2016 Update on menopause

Article Type
Changed
Tue, 08/28/2018 - 11:07
Display Headline
2016 Update on menopause

In this Update, I discuss important new study results regarding the cardiovascular safety of hormone therapy (HT) in early menopausal women. In addition, I review survey data that reveal a huge number of US women are using compounded HT preparations, which have unproven efficacy and safety.

Earlier initiation is better: ELITE trial provides strong support for the estrogen timing hypothesis
Hodis HN, Mack WJ, Henderson VW, et al; for the ELITE Research Group. Vascular effects of early versus late postmenopausal treatment with estradiol. N Engl J Med. 2016;374(13):1221-1231.

Keaney JF, Solomon G. Postmenopausal hormone therapy and atherosclerosis--time is of the essence [editorial]. N Engl J Med. 2016;374(13):1279-1280.

A substantial amount of published data, including from the Women's Health Initiative (WHI), supports the timing hypothesis, which proposes that HT slows the progression of atherosclerosis among recently menopausal women but has a neutral or adverse effect among women who are a decade or more past menopause onset.1 To directly test this hypothesis, Hodis and colleagues randomly assigned healthy postmenopausal women (<6 years or ≥10 years past menopause) without cardiovascular disease (CVD) to oral estradiol 1 mg or placebo. Women with a uterus also were randomly assigned to receive either vaginal progesterone gel or placebo gel. The primary outcome was the rate of change in carotid artery intima-media thickness (CIMT), which was assessed at baseline and each 6 months of the study. (An earlier report had noted that baseline CIMT correlated well with CVD risk factors.2) Coronary artery atherosclerosis, a secondary outcome, was assessed at study completion using computed tomography (CT).

Details of the study
Among the 643 participants in the Early versus Late Intervention Trial with Estradiol (ELITE), the median years since menopause and the median age at enrollment were 3.5 and 55.4, respectively, in the early postmenopause group and 14.3 and 63.6, respectively, in the late postmenopause group.  

Among the younger women, after a median of 5 years of study medications, the estradiol group had less progression of CIMT than the placebo group (P = .008). By contrast, in the older group, rates of CIMT progression were similar in the HT and placebo groups (P = .29). The relationship between estrogen and CIMT progression differed significantly between the younger and older groups (P = .007). Use of progesterone did not change these trends. Coronary artery CT parameters did not differ significantly between the placebo and HT groups in the age group or in the time-since-menopause group.

What this evidence means for practice
In an editorial accompanying the published results of the ELITE trial, Keaney and Solomon concluded that, although estrogen had a favorable effect on atherosclerosis in early menopause, it would be premature to recommend HT for prevention of cardiovascular events. I agree with them, but I also would like to note that the use of HT for the treatment of menopausal symptoms has plummeted since the initial WHI findings in 2002, with infrequent HT use even among symptomatic women in early menopause.3 (And I refer you to the special inset featuring JoAnn E. Manson, MD, DrPH) The takeaway message is that this important new clinical trial provides additional reassurance regarding the cardiovascular safety of HT when initiated by recently menopausal women to treat bothersome vasomotor symptoms. This message represents welcome news for women with bothersome menopausal symptoms considering use of HT.

A word about the vaginal progesterone gel used in the ELITE trial in relation to clinical practice: Given the need for vaginal placement of progesterone gel, potential messiness, and high cost, few clinicians may prescribe this formulation, and few women probably would choose to use it. As an alternative, micronized progesterone 100-mg capsules are less expensive and well accepted by most patients. These capsules are formulated with peanut oil. Because they may cause women to feel drowsy, the capsules should be taken at bedtime. In women with an intact uterus who are taking oral estradiol 1-mg tablets, one appropriate progestogen regimen for endometrial suppression is a 100-mg micronized progesterone capsule each night, continuously.

 

WHI, ELITE and the timing hypothesis:
New evidence on HT in early menopause is reassuring

 

Q&A with JoAnn E. Manson, MD, DrPH
In this interview, Dr. JoAnn Manson discusses the reassuring results of recent hormone therapy (HT) trials in early versus later postmenopausal women, examines these outcomes in the context of the Women's Health Initiative (WHI) trial and ELITE trial, and debunks an enduring common misconception about the WHI.

Q You have said for several years that there has been a misconception about the WHI trial. What is that misconception, and what has been its impact on clinicians, women, and the use of HT?
A The WHI HT trial has been largely misunderstood. It was designed to address the balance of benefits and risks of long-term HT for the prevention of chronic disease in postmenopausal women across a broad range of ages (average age 63).1,2 It was not intended to evaluate the clinical role of HT for managing menopausal symptoms in young and early menopausal women.3 Overall, the WHI study findings have been inappropriately extrapolated to women in their 40s and early 50s who report distressing hot flashes, night sweats, and other menopausal symptoms, and they are often used as a reason to deny therapy when in fact many of these women would be appropriate candidates for HT.

There is increasing evidence that younger women in early menopause who are taking HT have a lower risk of adverse outcomes and lower absolute risks of disease than older women.2,3 In younger, early menopausal women with bothersome hot flashes, night sweats, or other menopausal symptoms and who have no contraindications to HT, the benefits of treatment are likely to outweigh the risks, and these patients derive quality-of-life benefits from treatment.

Q How do the results of the recent ELITE (Early versus Late Intervention Trial with Estradiol) trial build on cardiovascular safety, in particular, of HT and when HT is optimally initiated?
A The ELITE trial directly tested the "timing hypothesis" and the role of HT in slowing the progression of atherosclerosis in early menopause (defined as within 6 years of menopause onset) compared with the effect in women in later menopause (defined as at least 10 yearspast menopause).4 The investigators used carotid artery intima-media thickness (CIMT) as a surrogate end point. In this trial, 643 women were randomly assigned according to whether they were in early or later menopause to receive either placebo or estradiol 1 mg daily; women with a uterus also received progesterone 45 mg as a 4% vaginal gel or matching placebo gel. The median duration of intervention was 5 years.
 
The ELITE study results provide support for the "critical window hypothesis" in that the estradiol-treated younger women closer to onset of menopause had slowing of atherosclerosis compared with the placebo group, while the older women more distant from menopause did not have slowing of atherosclerosis with estradiol.
 
The ELITE trial was not large enough, however, to assess clinical end points--rates of heart attack, stroke, or other cardiovascular events. So it remains unclear whether the findings for the surrogate end point of CIMT would translate into a reduced risk of clinical events in the younger women. Nevertheless, ELITE does provide more reassurance about the use of HT in early menopause and supports the possibility that the overall results of the WHI among women enrolled at an average age of 63 years may not apply directly to younger women in early menopause.

Q What impact on clinical practice do you anticipate as a result of the ELITE trial results?
A The findings provide further support for the timing hypothesis and offer additional reassurance regarding the safety  of HT in early menopause for management of menopausal symptoms. However, the trial does not provide conclusive evidence to support recommendations to use HT for the express purpose of preventing cardiovascular disease (CVD), even if HT is started in early menopause. Using a surrogate end point for atherosclerosis (CIMT) is not the same as looking at clinical events. There are many biologic pathways for heart attacks, strokes, and other cardiovascular events. In addition to atherosclerosis, for example, there is thrombosis, clotting, thrombo-occlusion within a blood vessel, and plaque rupture. Again, we do not know whether the CIMT-based results would translate directly into a reduction in clinical heart attacks and stroke.

The main takeaway point from the ELITE trial results is further reassurance for use of HT for management of menopausal symptoms in early menopause, but not for long-term chronic disease prevention at any age.

Q Another recent study, published in the Journal of Clinical Endocrinology and Metabolism, addresses HT and the timing hypothesis but in this instance relating to glucose tolerance.5 What did these study authors find? 
A This study by Pereira and colleagues is very interesting and suggests that the window of opportunity for initiating estrogen therapy may apply not only to coronary events but also to glucose tolerance, insulin sensitivity, and diabetes risk.5

The authors investigated the effects of short-term high-dose transdermal estradiol on the insulin-mediated glucose disposal rate (GDR), which is a measure of insulin-stimulated glucose uptake. Participants in this randomized, crossover, placebo-controlled study included 22 women who were in early menopause (6 years or less since final menses) and 24 women who were in later menopause (10 years or longer since final menses). All of the women were naïve to hormone therapy, and baseline GDR did not differ between groups. After 1 week of treatment with transdermal estradiol (a high dose of 150 μg) or placebo, the participants' GDR was measured via a hyperinsulinemic-euglycemic clamp.
 
The investigators found that in the younger women, estradiol had a favorable effect on insulin sensitivity and GDR, whereas in the older women, there was no evidence of a favorable effect and, in fact, there was a signal for risk and more adverse findings in this group.

Several studies in the WHI also looked at glucose tolerance and at the risk of being diagnosed with diabetes. While the results of the WHI estrogen-alone trial revealed a reduction in diabetes and favorable effects across age groups, in the WHI estrogen-plus-progestin trial we did see a signal that the results for diabetes may have been more favorable in the younger than in the older women, somewhat consistent with the findings of Pereira and colleagues.2,5
Overall this issue requires more research, but the Pereira study provides further support for the possibility that estrogen's metabolic effects may vary by age and time since menopause, and there is evidence that the estrogen receptors may be more functional and more sensitive in early rather than later menopause. These findings are very interesting and consistent with the overall hypothesis about the importance of age and time since menopause in relation to estrogen action. Again, they offer further support for use of HT for managing bothersome menopausal symptoms in early menopause, but they should not be interpreted as endorsing the use of HT to prevent either diabetes or CVD, due to the potential for other risks.

Q Where would you like to see future research conducted regarding the timing hypothesis?
A I would like to see more research on the role of oral versus transdermal estrogen in relation to insulin sensitivity, diabetes risk, and CVD risk, and more research on the role of estrogen dose, different types of progestogens, and the benefits and risks of novel formulations, including selective estrogen receptor modulators and tissue selective estrogen complexes.
 

 


Dr. Manson is Professor of Medicine and the Michael and Lee Bell Professor of Women's Health at Harvard Medical School and Chief of the Division of Preventive Medicine at Brigham and Women's Hospital, Boston, Massachusetts. She is a past President of the North American Menopause Society (NAMS) and a NAMS Certified Menopause Practitioner.

The author reports no financial relationships relevant to this article.

 


References

 

  1. Rossouw JE, Anderson GL, Prentice RL, et al; Writing Group for the Women's Health Initiative Investigators. Risks and benefits of estrogen plus progestin in healthy postmenopausal women: principal results from the Women's Health Initiative randomized controlled trial. JAMA. 2002;288(3):321-333.
  2. Manson JE, Chlebowski RT, Stefanick ML, et al. Menopausal hormone therapy and health outcomes during the intervention and extended poststopping phases of the Women's Health Initiative randomized trials. JAMA. 2013(13);310:1353-1368.
  3. Manson JE, Kaunitz AM. Menopause management-- getting clinical care back on track. N Engl J Med. 2016;374(19):803-806.
  4. Hodis HN, Mack WJ, Henderson VW, et al; ELITE Research Group. Vascular effects of early versus late postmenopausal treatment with estradiol. N Engl J Med. 2016;374(13):1221-1231.
  5. Pereira RI, Casey BA, Swibas TA, Erickson CB, Wolfe P, Van Pelt RE. Timing of estradiol treatment after menopause may determine benefit or harm to insulin action. J Clin Endocrinol Metab. 2015;100(12):4456-4462.
 

 

FDA-approved HT is preferable to compounded HT formulations
Pinkerton JV, Santoro N. Compounded bioidentical hormone therapy: identifying use trends and knowledge gaps among US women. Menopause. 2015;22(9):926-936.

Pinkerton JV, Constantine GD. Compounded non-FDA-approved menopausal hormone therapy prescriptions have increased: results of a pharmacy survey. Menopause. 2016;23(4):359-367.

Gass ML, Stuenkel CA, Utian WH, LaCroix A, Liu JH, Shifren JL.; North American Menopause Society (NAMS) Advisory Panel consisting of representatives of NAMS Board of Trustees and other experts in women's health. Use of compounded hormone therapy in the United States: report of The North American Menopause Society Survey. Menopause. 2015;22(12):1276-1284.

Consider how you would manage this clinical scenario: During a well-woman visit, your 54-year-old patient mentions that, after seeing an advertisement on television, she visited a clinic that sells compounded hormones. There, she underwent some testing and received an estrogen-testosterone implant and a progesterone cream that she applies to her skin each night to treat her menopausal symptoms. Now what?

The use of HT for menopausal symptoms declined considerably following the 2002 publication of the initial findings from the WHI, and its use remains low.4 Symptomatic menopausal women often find that their physicians are reluctant to consider prescribing treatment for menopausal symptoms because of safety concerns regarding HT use. Further, confusion about HT safety has opened the door to the increasing use of compounded bioidentical HT formulations, which are not approved by the US Food and Drug Administration (FDA).3 Since the publication of my 2015 Update on menopause (OBG Manag. 2015;27(6):37−40,42−43), several reports have addressed the use of "custom compounded" bioidentical menopausal HT in US women.

Millions use compounded HT for menopausal symptoms
A recent study by Pinkerton and Santoro that analyzed data from 2 national surveys suggested that as many as 2.5 million US women currently use non−FDA-approved custom-compounded HT. The authors also found that more than three-quarters of women using compounded HT are unaware that these medications, which include oral, topical, injectable, and implantable (pellet) formulations, are not FDA approved. In a study by Pinkerton and Constantine, total annual sales of compounded HT were estimated at approximately $1.5 billion. The dramatic growth in the use of compounded HT appears to have stemmed from celebrity endorsements, aggressive and unregulated marketing, and beliefs about the safety of "natural" hormones.5

Spurious laboratory testing. Women seeking care from physicians and clinics that provide compounded HT are often advised to undergo saliva and serum testing to determine hormone levels. Many women are unaware, however, that saliva testing does not correlate with serum levels of hormones. Further, in contrast with conditions such as thyroid disease and diabetes, routine laboratory testing is neither indicated nor helpful in the management of menopausal symptoms.6 Of note, insurance companies often do not reimburse for the cost of saliva hormone testing or for non-FDA-approved hormones.5

Inadequate endometrial protection. Topical progesterone cream, which is not absorbed in sufficient quantities to generate therapeutic effects, is often prescribed by practitioners who sell bioidentical compounded hormones to their patients.7 According to a report by the North American Menopause Society, several cases of endometrial cancer have been reported among women using compounded HT. These cases may reflect use of systemic estrogen without adequate progesterone protection, as could occur when topical progesterone cream is prescribed to women with an intact uterus using systemic estrogen therapy. 

What this evidence means for practice
Clinicians should be alert to the growing prevalence of use of compounded HT and should educate themselves and their patients about the differences between non−FDA-approved HT and FDA-approved HT. Further, women interested in using "natural," "bioidentical," or "custom compounded" HT should be aware that FDA-approved estradiol (oral, transdermal, and vaginal) and progesterone (oral and vaginal) formulations are available.

Because the FDA does not test custom compounded hormones for efficacy or safety and the standardization and purity of these products are uncertain, the American College of Obstetricians and Gynecologists has stated that FDA-approved HT is preferred for management of menopausal symptoms.8 Similarly, the North American Menopause Society does not recommend the use of compounded HT for treatment of menopausal symptoms unless a patient is allergic to ingredients contained in FDA-approved HT formulations.9

Share your thoughts! Send your Letter to the Editor to [email protected]. Please include your name and the city and state in which you practice.

References

 

 

  1. Manson JE, Chlebowski RT, Stefanick ML, et al. Menopausal hormone therapy and health outcomes during the intervention and extended poststopping phases of the Women’s Health Initiative randomized trials. JAMA. 2013;310(13):1353–1368.
  2. Hodis HN, Mack WJ, Shoupe D, et al. Methods and baseline cardiovascular data from the Early versus Late Intervention Trial with Estradiol testing the menopausal hormone timing hypothesis. Menopause. 2015;22(4):391–401.
  3. Manson JE, Kaunitz AM. Menopause management—getting clinical care back on track. N Engl J Med. 2016;374(9):803–806.
  4. Rossouw JE, Anderson GL, Prentice RL, et al; Writing Group for the Women’s Health Initiative Investigators. Risks and benefits of estrogen plus progestin in healthy postmenopausal women: principal results From the Women’s Health Initiative randomized controlled trial. JAMA. 2002;288(3):321–333.
  5. Kaunitz AM, Kaunitz JD. Compounded bioidentical hormone therapy: time for a reality check? Menopause. 2015;22(9):919–920.
  6. Kaunitz AM, Manson JE. Management of menopausal symptoms. Obstet Gynecol. 2015;126(4):859–876.
  7. Benster B, Carey A, Wadsworth F, Vashisht A, Domoney C, Studd J. A double-blind placebo-controlled study to evaluate the effect of progestelle progesterone cream on postmenopausal women. Menopause Int. 2009;15(2):63–69.
  8. American College of Obstetricians Gynecologists. ACOG Practice Bulletin No. 141: management of menopausal symptoms. Obstet Gynecol. 2014;123(1):202–216.
  9. North American Menopause Society. The 2012 hormone therapy position statement of The North American Menopause Society. Menopause. 2012;19(3):257–271.
Article PDF
Author and Disclosure Information

 

Andrew M. Kaunitz, MD

 

Dr. Kaunitz is University of Florida Research Foundation Professor and Associate Chairman, Department of Obstetrics and Gynecology, University of Florida College of Medicine—Jacksonville. Dr. Kaunitz serves as Medical Director and directs Menopause and Gynecologic Ultrasound Services at UF Women’s Health Specialists—Emerson. Dr. Kaunitz is a NAMS Certified Menopause Practitioner. He serves on the OBG Management Board of Editors.

 

Dr. Kaunitz reports receiving grant or research support from Bayer, Pfizer, Radius, Shionogi, and TherapeuticsMD.

Issue
OBG Management - 28(7)
Publications
Topics
Page Number
38-43
Legacy Keywords
Andrew M. Kaunitz MD, JoAnn Manson MD DrPh, menopause, vardiovascular safety, hormone therapy, HT, early menopausal women, compounded HT preparations, FDA, Women's Health Initiative, WHI, timing hypothesis, atherosclerosis, estrogen, progesterone
Sections
Author and Disclosure Information

 

Andrew M. Kaunitz, MD

 

Dr. Kaunitz is University of Florida Research Foundation Professor and Associate Chairman, Department of Obstetrics and Gynecology, University of Florida College of Medicine—Jacksonville. Dr. Kaunitz serves as Medical Director and directs Menopause and Gynecologic Ultrasound Services at UF Women’s Health Specialists—Emerson. Dr. Kaunitz is a NAMS Certified Menopause Practitioner. He serves on the OBG Management Board of Editors.

 

Dr. Kaunitz reports receiving grant or research support from Bayer, Pfizer, Radius, Shionogi, and TherapeuticsMD.

Author and Disclosure Information

 

Andrew M. Kaunitz, MD

 

Dr. Kaunitz is University of Florida Research Foundation Professor and Associate Chairman, Department of Obstetrics and Gynecology, University of Florida College of Medicine—Jacksonville. Dr. Kaunitz serves as Medical Director and directs Menopause and Gynecologic Ultrasound Services at UF Women’s Health Specialists—Emerson. Dr. Kaunitz is a NAMS Certified Menopause Practitioner. He serves on the OBG Management Board of Editors.

 

Dr. Kaunitz reports receiving grant or research support from Bayer, Pfizer, Radius, Shionogi, and TherapeuticsMD.

Article PDF
Article PDF
Related Articles

In this Update, I discuss important new study results regarding the cardiovascular safety of hormone therapy (HT) in early menopausal women. In addition, I review survey data that reveal a huge number of US women are using compounded HT preparations, which have unproven efficacy and safety.

Earlier initiation is better: ELITE trial provides strong support for the estrogen timing hypothesis
Hodis HN, Mack WJ, Henderson VW, et al; for the ELITE Research Group. Vascular effects of early versus late postmenopausal treatment with estradiol. N Engl J Med. 2016;374(13):1221-1231.

Keaney JF, Solomon G. Postmenopausal hormone therapy and atherosclerosis--time is of the essence [editorial]. N Engl J Med. 2016;374(13):1279-1280.

A substantial amount of published data, including from the Women's Health Initiative (WHI), supports the timing hypothesis, which proposes that HT slows the progression of atherosclerosis among recently menopausal women but has a neutral or adverse effect among women who are a decade or more past menopause onset.1 To directly test this hypothesis, Hodis and colleagues randomly assigned healthy postmenopausal women (<6 years or ≥10 years past menopause) without cardiovascular disease (CVD) to oral estradiol 1 mg or placebo. Women with a uterus also were randomly assigned to receive either vaginal progesterone gel or placebo gel. The primary outcome was the rate of change in carotid artery intima-media thickness (CIMT), which was assessed at baseline and each 6 months of the study. (An earlier report had noted that baseline CIMT correlated well with CVD risk factors.2) Coronary artery atherosclerosis, a secondary outcome, was assessed at study completion using computed tomography (CT).

Details of the study
Among the 643 participants in the Early versus Late Intervention Trial with Estradiol (ELITE), the median years since menopause and the median age at enrollment were 3.5 and 55.4, respectively, in the early postmenopause group and 14.3 and 63.6, respectively, in the late postmenopause group.  

Among the younger women, after a median of 5 years of study medications, the estradiol group had less progression of CIMT than the placebo group (P = .008). By contrast, in the older group, rates of CIMT progression were similar in the HT and placebo groups (P = .29). The relationship between estrogen and CIMT progression differed significantly between the younger and older groups (P = .007). Use of progesterone did not change these trends. Coronary artery CT parameters did not differ significantly between the placebo and HT groups in the age group or in the time-since-menopause group.

What this evidence means for practice
In an editorial accompanying the published results of the ELITE trial, Keaney and Solomon concluded that, although estrogen had a favorable effect on atherosclerosis in early menopause, it would be premature to recommend HT for prevention of cardiovascular events. I agree with them, but I also would like to note that the use of HT for the treatment of menopausal symptoms has plummeted since the initial WHI findings in 2002, with infrequent HT use even among symptomatic women in early menopause.3 (And I refer you to the special inset featuring JoAnn E. Manson, MD, DrPH) The takeaway message is that this important new clinical trial provides additional reassurance regarding the cardiovascular safety of HT when initiated by recently menopausal women to treat bothersome vasomotor symptoms. This message represents welcome news for women with bothersome menopausal symptoms considering use of HT.

A word about the vaginal progesterone gel used in the ELITE trial in relation to clinical practice: Given the need for vaginal placement of progesterone gel, potential messiness, and high cost, few clinicians may prescribe this formulation, and few women probably would choose to use it. As an alternative, micronized progesterone 100-mg capsules are less expensive and well accepted by most patients. These capsules are formulated with peanut oil. Because they may cause women to feel drowsy, the capsules should be taken at bedtime. In women with an intact uterus who are taking oral estradiol 1-mg tablets, one appropriate progestogen regimen for endometrial suppression is a 100-mg micronized progesterone capsule each night, continuously.

 

WHI, ELITE and the timing hypothesis:
New evidence on HT in early menopause is reassuring

 

Q&A with JoAnn E. Manson, MD, DrPH
In this interview, Dr. JoAnn Manson discusses the reassuring results of recent hormone therapy (HT) trials in early versus later postmenopausal women, examines these outcomes in the context of the Women's Health Initiative (WHI) trial and ELITE trial, and debunks an enduring common misconception about the WHI.

Q You have said for several years that there has been a misconception about the WHI trial. What is that misconception, and what has been its impact on clinicians, women, and the use of HT?
A The WHI HT trial has been largely misunderstood. It was designed to address the balance of benefits and risks of long-term HT for the prevention of chronic disease in postmenopausal women across a broad range of ages (average age 63).1,2 It was not intended to evaluate the clinical role of HT for managing menopausal symptoms in young and early menopausal women.3 Overall, the WHI study findings have been inappropriately extrapolated to women in their 40s and early 50s who report distressing hot flashes, night sweats, and other menopausal symptoms, and they are often used as a reason to deny therapy when in fact many of these women would be appropriate candidates for HT.

There is increasing evidence that younger women in early menopause who are taking HT have a lower risk of adverse outcomes and lower absolute risks of disease than older women.2,3 In younger, early menopausal women with bothersome hot flashes, night sweats, or other menopausal symptoms and who have no contraindications to HT, the benefits of treatment are likely to outweigh the risks, and these patients derive quality-of-life benefits from treatment.

Q How do the results of the recent ELITE (Early versus Late Intervention Trial with Estradiol) trial build on cardiovascular safety, in particular, of HT and when HT is optimally initiated?
A The ELITE trial directly tested the "timing hypothesis" and the role of HT in slowing the progression of atherosclerosis in early menopause (defined as within 6 years of menopause onset) compared with the effect in women in later menopause (defined as at least 10 yearspast menopause).4 The investigators used carotid artery intima-media thickness (CIMT) as a surrogate end point. In this trial, 643 women were randomly assigned according to whether they were in early or later menopause to receive either placebo or estradiol 1 mg daily; women with a uterus also received progesterone 45 mg as a 4% vaginal gel or matching placebo gel. The median duration of intervention was 5 years.
 
The ELITE study results provide support for the "critical window hypothesis" in that the estradiol-treated younger women closer to onset of menopause had slowing of atherosclerosis compared with the placebo group, while the older women more distant from menopause did not have slowing of atherosclerosis with estradiol.
 
The ELITE trial was not large enough, however, to assess clinical end points--rates of heart attack, stroke, or other cardiovascular events. So it remains unclear whether the findings for the surrogate end point of CIMT would translate into a reduced risk of clinical events in the younger women. Nevertheless, ELITE does provide more reassurance about the use of HT in early menopause and supports the possibility that the overall results of the WHI among women enrolled at an average age of 63 years may not apply directly to younger women in early menopause.

Q What impact on clinical practice do you anticipate as a result of the ELITE trial results?
A The findings provide further support for the timing hypothesis and offer additional reassurance regarding the safety  of HT in early menopause for management of menopausal symptoms. However, the trial does not provide conclusive evidence to support recommendations to use HT for the express purpose of preventing cardiovascular disease (CVD), even if HT is started in early menopause. Using a surrogate end point for atherosclerosis (CIMT) is not the same as looking at clinical events. There are many biologic pathways for heart attacks, strokes, and other cardiovascular events. In addition to atherosclerosis, for example, there is thrombosis, clotting, thrombo-occlusion within a blood vessel, and plaque rupture. Again, we do not know whether the CIMT-based results would translate directly into a reduction in clinical heart attacks and stroke.

The main takeaway point from the ELITE trial results is further reassurance for use of HT for management of menopausal symptoms in early menopause, but not for long-term chronic disease prevention at any age.

Q Another recent study, published in the Journal of Clinical Endocrinology and Metabolism, addresses HT and the timing hypothesis but in this instance relating to glucose tolerance.5 What did these study authors find? 
A This study by Pereira and colleagues is very interesting and suggests that the window of opportunity for initiating estrogen therapy may apply not only to coronary events but also to glucose tolerance, insulin sensitivity, and diabetes risk.5

The authors investigated the effects of short-term high-dose transdermal estradiol on the insulin-mediated glucose disposal rate (GDR), which is a measure of insulin-stimulated glucose uptake. Participants in this randomized, crossover, placebo-controlled study included 22 women who were in early menopause (6 years or less since final menses) and 24 women who were in later menopause (10 years or longer since final menses). All of the women were naïve to hormone therapy, and baseline GDR did not differ between groups. After 1 week of treatment with transdermal estradiol (a high dose of 150 μg) or placebo, the participants' GDR was measured via a hyperinsulinemic-euglycemic clamp.
 
The investigators found that in the younger women, estradiol had a favorable effect on insulin sensitivity and GDR, whereas in the older women, there was no evidence of a favorable effect and, in fact, there was a signal for risk and more adverse findings in this group.

Several studies in the WHI also looked at glucose tolerance and at the risk of being diagnosed with diabetes. While the results of the WHI estrogen-alone trial revealed a reduction in diabetes and favorable effects across age groups, in the WHI estrogen-plus-progestin trial we did see a signal that the results for diabetes may have been more favorable in the younger than in the older women, somewhat consistent with the findings of Pereira and colleagues.2,5
Overall this issue requires more research, but the Pereira study provides further support for the possibility that estrogen's metabolic effects may vary by age and time since menopause, and there is evidence that the estrogen receptors may be more functional and more sensitive in early rather than later menopause. These findings are very interesting and consistent with the overall hypothesis about the importance of age and time since menopause in relation to estrogen action. Again, they offer further support for use of HT for managing bothersome menopausal symptoms in early menopause, but they should not be interpreted as endorsing the use of HT to prevent either diabetes or CVD, due to the potential for other risks.

Q Where would you like to see future research conducted regarding the timing hypothesis?
A I would like to see more research on the role of oral versus transdermal estrogen in relation to insulin sensitivity, diabetes risk, and CVD risk, and more research on the role of estrogen dose, different types of progestogens, and the benefits and risks of novel formulations, including selective estrogen receptor modulators and tissue selective estrogen complexes.
 

 


Dr. Manson is Professor of Medicine and the Michael and Lee Bell Professor of Women's Health at Harvard Medical School and Chief of the Division of Preventive Medicine at Brigham and Women's Hospital, Boston, Massachusetts. She is a past President of the North American Menopause Society (NAMS) and a NAMS Certified Menopause Practitioner.

The author reports no financial relationships relevant to this article.

 


References

 

  1. Rossouw JE, Anderson GL, Prentice RL, et al; Writing Group for the Women's Health Initiative Investigators. Risks and benefits of estrogen plus progestin in healthy postmenopausal women: principal results from the Women's Health Initiative randomized controlled trial. JAMA. 2002;288(3):321-333.
  2. Manson JE, Chlebowski RT, Stefanick ML, et al. Menopausal hormone therapy and health outcomes during the intervention and extended poststopping phases of the Women's Health Initiative randomized trials. JAMA. 2013(13);310:1353-1368.
  3. Manson JE, Kaunitz AM. Menopause management-- getting clinical care back on track. N Engl J Med. 2016;374(19):803-806.
  4. Hodis HN, Mack WJ, Henderson VW, et al; ELITE Research Group. Vascular effects of early versus late postmenopausal treatment with estradiol. N Engl J Med. 2016;374(13):1221-1231.
  5. Pereira RI, Casey BA, Swibas TA, Erickson CB, Wolfe P, Van Pelt RE. Timing of estradiol treatment after menopause may determine benefit or harm to insulin action. J Clin Endocrinol Metab. 2015;100(12):4456-4462.
 

 

FDA-approved HT is preferable to compounded HT formulations
Pinkerton JV, Santoro N. Compounded bioidentical hormone therapy: identifying use trends and knowledge gaps among US women. Menopause. 2015;22(9):926-936.

Pinkerton JV, Constantine GD. Compounded non-FDA-approved menopausal hormone therapy prescriptions have increased: results of a pharmacy survey. Menopause. 2016;23(4):359-367.

Gass ML, Stuenkel CA, Utian WH, LaCroix A, Liu JH, Shifren JL.; North American Menopause Society (NAMS) Advisory Panel consisting of representatives of NAMS Board of Trustees and other experts in women's health. Use of compounded hormone therapy in the United States: report of The North American Menopause Society Survey. Menopause. 2015;22(12):1276-1284.

Consider how you would manage this clinical scenario: During a well-woman visit, your 54-year-old patient mentions that, after seeing an advertisement on television, she visited a clinic that sells compounded hormones. There, she underwent some testing and received an estrogen-testosterone implant and a progesterone cream that she applies to her skin each night to treat her menopausal symptoms. Now what?

The use of HT for menopausal symptoms declined considerably following the 2002 publication of the initial findings from the WHI, and its use remains low.4 Symptomatic menopausal women often find that their physicians are reluctant to consider prescribing treatment for menopausal symptoms because of safety concerns regarding HT use. Further, confusion about HT safety has opened the door to the increasing use of compounded bioidentical HT formulations, which are not approved by the US Food and Drug Administration (FDA).3 Since the publication of my 2015 Update on menopause (OBG Manag. 2015;27(6):37−40,42−43), several reports have addressed the use of "custom compounded" bioidentical menopausal HT in US women.

Millions use compounded HT for menopausal symptoms
A recent study by Pinkerton and Santoro that analyzed data from 2 national surveys suggested that as many as 2.5 million US women currently use non−FDA-approved custom-compounded HT. The authors also found that more than three-quarters of women using compounded HT are unaware that these medications, which include oral, topical, injectable, and implantable (pellet) formulations, are not FDA approved. In a study by Pinkerton and Constantine, total annual sales of compounded HT were estimated at approximately $1.5 billion. The dramatic growth in the use of compounded HT appears to have stemmed from celebrity endorsements, aggressive and unregulated marketing, and beliefs about the safety of "natural" hormones.5

Spurious laboratory testing. Women seeking care from physicians and clinics that provide compounded HT are often advised to undergo saliva and serum testing to determine hormone levels. Many women are unaware, however, that saliva testing does not correlate with serum levels of hormones. Further, in contrast with conditions such as thyroid disease and diabetes, routine laboratory testing is neither indicated nor helpful in the management of menopausal symptoms.6 Of note, insurance companies often do not reimburse for the cost of saliva hormone testing or for non-FDA-approved hormones.5

Inadequate endometrial protection. Topical progesterone cream, which is not absorbed in sufficient quantities to generate therapeutic effects, is often prescribed by practitioners who sell bioidentical compounded hormones to their patients.7 According to a report by the North American Menopause Society, several cases of endometrial cancer have been reported among women using compounded HT. These cases may reflect use of systemic estrogen without adequate progesterone protection, as could occur when topical progesterone cream is prescribed to women with an intact uterus using systemic estrogen therapy. 

What this evidence means for practice
Clinicians should be alert to the growing prevalence of use of compounded HT and should educate themselves and their patients about the differences between non−FDA-approved HT and FDA-approved HT. Further, women interested in using "natural," "bioidentical," or "custom compounded" HT should be aware that FDA-approved estradiol (oral, transdermal, and vaginal) and progesterone (oral and vaginal) formulations are available.

Because the FDA does not test custom compounded hormones for efficacy or safety and the standardization and purity of these products are uncertain, the American College of Obstetricians and Gynecologists has stated that FDA-approved HT is preferred for management of menopausal symptoms.8 Similarly, the North American Menopause Society does not recommend the use of compounded HT for treatment of menopausal symptoms unless a patient is allergic to ingredients contained in FDA-approved HT formulations.9

Share your thoughts! Send your Letter to the Editor to [email protected]. Please include your name and the city and state in which you practice.

In this Update, I discuss important new study results regarding the cardiovascular safety of hormone therapy (HT) in early menopausal women. In addition, I review survey data that reveal a huge number of US women are using compounded HT preparations, which have unproven efficacy and safety.

Earlier initiation is better: ELITE trial provides strong support for the estrogen timing hypothesis
Hodis HN, Mack WJ, Henderson VW, et al; for the ELITE Research Group. Vascular effects of early versus late postmenopausal treatment with estradiol. N Engl J Med. 2016;374(13):1221-1231.

Keaney JF, Solomon G. Postmenopausal hormone therapy and atherosclerosis--time is of the essence [editorial]. N Engl J Med. 2016;374(13):1279-1280.

A substantial amount of published data, including from the Women's Health Initiative (WHI), supports the timing hypothesis, which proposes that HT slows the progression of atherosclerosis among recently menopausal women but has a neutral or adverse effect among women who are a decade or more past menopause onset.1 To directly test this hypothesis, Hodis and colleagues randomly assigned healthy postmenopausal women (<6 years or ≥10 years past menopause) without cardiovascular disease (CVD) to oral estradiol 1 mg or placebo. Women with a uterus also were randomly assigned to receive either vaginal progesterone gel or placebo gel. The primary outcome was the rate of change in carotid artery intima-media thickness (CIMT), which was assessed at baseline and each 6 months of the study. (An earlier report had noted that baseline CIMT correlated well with CVD risk factors.2) Coronary artery atherosclerosis, a secondary outcome, was assessed at study completion using computed tomography (CT).

Details of the study
Among the 643 participants in the Early versus Late Intervention Trial with Estradiol (ELITE), the median years since menopause and the median age at enrollment were 3.5 and 55.4, respectively, in the early postmenopause group and 14.3 and 63.6, respectively, in the late postmenopause group.  

Among the younger women, after a median of 5 years of study medications, the estradiol group had less progression of CIMT than the placebo group (P = .008). By contrast, in the older group, rates of CIMT progression were similar in the HT and placebo groups (P = .29). The relationship between estrogen and CIMT progression differed significantly between the younger and older groups (P = .007). Use of progesterone did not change these trends. Coronary artery CT parameters did not differ significantly between the placebo and HT groups in the age group or in the time-since-menopause group.

What this evidence means for practice
In an editorial accompanying the published results of the ELITE trial, Keaney and Solomon concluded that, although estrogen had a favorable effect on atherosclerosis in early menopause, it would be premature to recommend HT for prevention of cardiovascular events. I agree with them, but I also would like to note that the use of HT for the treatment of menopausal symptoms has plummeted since the initial WHI findings in 2002, with infrequent HT use even among symptomatic women in early menopause.3 (And I refer you to the special inset featuring JoAnn E. Manson, MD, DrPH) The takeaway message is that this important new clinical trial provides additional reassurance regarding the cardiovascular safety of HT when initiated by recently menopausal women to treat bothersome vasomotor symptoms. This message represents welcome news for women with bothersome menopausal symptoms considering use of HT.

A word about the vaginal progesterone gel used in the ELITE trial in relation to clinical practice: Given the need for vaginal placement of progesterone gel, potential messiness, and high cost, few clinicians may prescribe this formulation, and few women probably would choose to use it. As an alternative, micronized progesterone 100-mg capsules are less expensive and well accepted by most patients. These capsules are formulated with peanut oil. Because they may cause women to feel drowsy, the capsules should be taken at bedtime. In women with an intact uterus who are taking oral estradiol 1-mg tablets, one appropriate progestogen regimen for endometrial suppression is a 100-mg micronized progesterone capsule each night, continuously.

 

WHI, ELITE and the timing hypothesis:
New evidence on HT in early menopause is reassuring

 

Q&A with JoAnn E. Manson, MD, DrPH
In this interview, Dr. JoAnn Manson discusses the reassuring results of recent hormone therapy (HT) trials in early versus later postmenopausal women, examines these outcomes in the context of the Women's Health Initiative (WHI) trial and ELITE trial, and debunks an enduring common misconception about the WHI.

Q You have said for several years that there has been a misconception about the WHI trial. What is that misconception, and what has been its impact on clinicians, women, and the use of HT?
A The WHI HT trial has been largely misunderstood. It was designed to address the balance of benefits and risks of long-term HT for the prevention of chronic disease in postmenopausal women across a broad range of ages (average age 63).1,2 It was not intended to evaluate the clinical role of HT for managing menopausal symptoms in young and early menopausal women.3 Overall, the WHI study findings have been inappropriately extrapolated to women in their 40s and early 50s who report distressing hot flashes, night sweats, and other menopausal symptoms, and they are often used as a reason to deny therapy when in fact many of these women would be appropriate candidates for HT.

There is increasing evidence that younger women in early menopause who are taking HT have a lower risk of adverse outcomes and lower absolute risks of disease than older women.2,3 In younger, early menopausal women with bothersome hot flashes, night sweats, or other menopausal symptoms and who have no contraindications to HT, the benefits of treatment are likely to outweigh the risks, and these patients derive quality-of-life benefits from treatment.

Q How do the results of the recent ELITE (Early versus Late Intervention Trial with Estradiol) trial build on cardiovascular safety, in particular, of HT and when HT is optimally initiated?
A The ELITE trial directly tested the "timing hypothesis" and the role of HT in slowing the progression of atherosclerosis in early menopause (defined as within 6 years of menopause onset) compared with the effect in women in later menopause (defined as at least 10 yearspast menopause).4 The investigators used carotid artery intima-media thickness (CIMT) as a surrogate end point. In this trial, 643 women were randomly assigned according to whether they were in early or later menopause to receive either placebo or estradiol 1 mg daily; women with a uterus also received progesterone 45 mg as a 4% vaginal gel or matching placebo gel. The median duration of intervention was 5 years.
 
The ELITE study results provide support for the "critical window hypothesis" in that the estradiol-treated younger women closer to onset of menopause had slowing of atherosclerosis compared with the placebo group, while the older women more distant from menopause did not have slowing of atherosclerosis with estradiol.
 
The ELITE trial was not large enough, however, to assess clinical end points--rates of heart attack, stroke, or other cardiovascular events. So it remains unclear whether the findings for the surrogate end point of CIMT would translate into a reduced risk of clinical events in the younger women. Nevertheless, ELITE does provide more reassurance about the use of HT in early menopause and supports the possibility that the overall results of the WHI among women enrolled at an average age of 63 years may not apply directly to younger women in early menopause.

Q What impact on clinical practice do you anticipate as a result of the ELITE trial results?
A The findings provide further support for the timing hypothesis and offer additional reassurance regarding the safety  of HT in early menopause for management of menopausal symptoms. However, the trial does not provide conclusive evidence to support recommendations to use HT for the express purpose of preventing cardiovascular disease (CVD), even if HT is started in early menopause. Using a surrogate end point for atherosclerosis (CIMT) is not the same as looking at clinical events. There are many biologic pathways for heart attacks, strokes, and other cardiovascular events. In addition to atherosclerosis, for example, there is thrombosis, clotting, thrombo-occlusion within a blood vessel, and plaque rupture. Again, we do not know whether the CIMT-based results would translate directly into a reduction in clinical heart attacks and stroke.

The main takeaway point from the ELITE trial results is further reassurance for use of HT for management of menopausal symptoms in early menopause, but not for long-term chronic disease prevention at any age.

Q Another recent study, published in the Journal of Clinical Endocrinology and Metabolism, addresses HT and the timing hypothesis but in this instance relating to glucose tolerance.5 What did these study authors find? 
A This study by Pereira and colleagues is very interesting and suggests that the window of opportunity for initiating estrogen therapy may apply not only to coronary events but also to glucose tolerance, insulin sensitivity, and diabetes risk.5

The authors investigated the effects of short-term high-dose transdermal estradiol on the insulin-mediated glucose disposal rate (GDR), which is a measure of insulin-stimulated glucose uptake. Participants in this randomized, crossover, placebo-controlled study included 22 women who were in early menopause (6 years or less since final menses) and 24 women who were in later menopause (10 years or longer since final menses). All of the women were naïve to hormone therapy, and baseline GDR did not differ between groups. After 1 week of treatment with transdermal estradiol (a high dose of 150 μg) or placebo, the participants' GDR was measured via a hyperinsulinemic-euglycemic clamp.
 
The investigators found that in the younger women, estradiol had a favorable effect on insulin sensitivity and GDR, whereas in the older women, there was no evidence of a favorable effect and, in fact, there was a signal for risk and more adverse findings in this group.

Several studies in the WHI also looked at glucose tolerance and at the risk of being diagnosed with diabetes. While the results of the WHI estrogen-alone trial revealed a reduction in diabetes and favorable effects across age groups, in the WHI estrogen-plus-progestin trial we did see a signal that the results for diabetes may have been more favorable in the younger than in the older women, somewhat consistent with the findings of Pereira and colleagues.2,5
Overall this issue requires more research, but the Pereira study provides further support for the possibility that estrogen's metabolic effects may vary by age and time since menopause, and there is evidence that the estrogen receptors may be more functional and more sensitive in early rather than later menopause. These findings are very interesting and consistent with the overall hypothesis about the importance of age and time since menopause in relation to estrogen action. Again, they offer further support for use of HT for managing bothersome menopausal symptoms in early menopause, but they should not be interpreted as endorsing the use of HT to prevent either diabetes or CVD, due to the potential for other risks.

Q Where would you like to see future research conducted regarding the timing hypothesis?
A I would like to see more research on the role of oral versus transdermal estrogen in relation to insulin sensitivity, diabetes risk, and CVD risk, and more research on the role of estrogen dose, different types of progestogens, and the benefits and risks of novel formulations, including selective estrogen receptor modulators and tissue selective estrogen complexes.
 

 


Dr. Manson is Professor of Medicine and the Michael and Lee Bell Professor of Women's Health at Harvard Medical School and Chief of the Division of Preventive Medicine at Brigham and Women's Hospital, Boston, Massachusetts. She is a past President of the North American Menopause Society (NAMS) and a NAMS Certified Menopause Practitioner.

The author reports no financial relationships relevant to this article.

 


References

 

  1. Rossouw JE, Anderson GL, Prentice RL, et al; Writing Group for the Women's Health Initiative Investigators. Risks and benefits of estrogen plus progestin in healthy postmenopausal women: principal results from the Women's Health Initiative randomized controlled trial. JAMA. 2002;288(3):321-333.
  2. Manson JE, Chlebowski RT, Stefanick ML, et al. Menopausal hormone therapy and health outcomes during the intervention and extended poststopping phases of the Women's Health Initiative randomized trials. JAMA. 2013(13);310:1353-1368.
  3. Manson JE, Kaunitz AM. Menopause management-- getting clinical care back on track. N Engl J Med. 2016;374(19):803-806.
  4. Hodis HN, Mack WJ, Henderson VW, et al; ELITE Research Group. Vascular effects of early versus late postmenopausal treatment with estradiol. N Engl J Med. 2016;374(13):1221-1231.
  5. Pereira RI, Casey BA, Swibas TA, Erickson CB, Wolfe P, Van Pelt RE. Timing of estradiol treatment after menopause may determine benefit or harm to insulin action. J Clin Endocrinol Metab. 2015;100(12):4456-4462.
 

 

FDA-approved HT is preferable to compounded HT formulations
Pinkerton JV, Santoro N. Compounded bioidentical hormone therapy: identifying use trends and knowledge gaps among US women. Menopause. 2015;22(9):926-936.

Pinkerton JV, Constantine GD. Compounded non-FDA-approved menopausal hormone therapy prescriptions have increased: results of a pharmacy survey. Menopause. 2016;23(4):359-367.

Gass ML, Stuenkel CA, Utian WH, LaCroix A, Liu JH, Shifren JL.; North American Menopause Society (NAMS) Advisory Panel consisting of representatives of NAMS Board of Trustees and other experts in women's health. Use of compounded hormone therapy in the United States: report of The North American Menopause Society Survey. Menopause. 2015;22(12):1276-1284.

Consider how you would manage this clinical scenario: During a well-woman visit, your 54-year-old patient mentions that, after seeing an advertisement on television, she visited a clinic that sells compounded hormones. There, she underwent some testing and received an estrogen-testosterone implant and a progesterone cream that she applies to her skin each night to treat her menopausal symptoms. Now what?

The use of HT for menopausal symptoms declined considerably following the 2002 publication of the initial findings from the WHI, and its use remains low.4 Symptomatic menopausal women often find that their physicians are reluctant to consider prescribing treatment for menopausal symptoms because of safety concerns regarding HT use. Further, confusion about HT safety has opened the door to the increasing use of compounded bioidentical HT formulations, which are not approved by the US Food and Drug Administration (FDA).3 Since the publication of my 2015 Update on menopause (OBG Manag. 2015;27(6):37−40,42−43), several reports have addressed the use of "custom compounded" bioidentical menopausal HT in US women.

Millions use compounded HT for menopausal symptoms
A recent study by Pinkerton and Santoro that analyzed data from 2 national surveys suggested that as many as 2.5 million US women currently use non−FDA-approved custom-compounded HT. The authors also found that more than three-quarters of women using compounded HT are unaware that these medications, which include oral, topical, injectable, and implantable (pellet) formulations, are not FDA approved. In a study by Pinkerton and Constantine, total annual sales of compounded HT were estimated at approximately $1.5 billion. The dramatic growth in the use of compounded HT appears to have stemmed from celebrity endorsements, aggressive and unregulated marketing, and beliefs about the safety of "natural" hormones.5

Spurious laboratory testing. Women seeking care from physicians and clinics that provide compounded HT are often advised to undergo saliva and serum testing to determine hormone levels. Many women are unaware, however, that saliva testing does not correlate with serum levels of hormones. Further, in contrast with conditions such as thyroid disease and diabetes, routine laboratory testing is neither indicated nor helpful in the management of menopausal symptoms.6 Of note, insurance companies often do not reimburse for the cost of saliva hormone testing or for non-FDA-approved hormones.5

Inadequate endometrial protection. Topical progesterone cream, which is not absorbed in sufficient quantities to generate therapeutic effects, is often prescribed by practitioners who sell bioidentical compounded hormones to their patients.7 According to a report by the North American Menopause Society, several cases of endometrial cancer have been reported among women using compounded HT. These cases may reflect use of systemic estrogen without adequate progesterone protection, as could occur when topical progesterone cream is prescribed to women with an intact uterus using systemic estrogen therapy. 

What this evidence means for practice
Clinicians should be alert to the growing prevalence of use of compounded HT and should educate themselves and their patients about the differences between non−FDA-approved HT and FDA-approved HT. Further, women interested in using "natural," "bioidentical," or "custom compounded" HT should be aware that FDA-approved estradiol (oral, transdermal, and vaginal) and progesterone (oral and vaginal) formulations are available.

Because the FDA does not test custom compounded hormones for efficacy or safety and the standardization and purity of these products are uncertain, the American College of Obstetricians and Gynecologists has stated that FDA-approved HT is preferred for management of menopausal symptoms.8 Similarly, the North American Menopause Society does not recommend the use of compounded HT for treatment of menopausal symptoms unless a patient is allergic to ingredients contained in FDA-approved HT formulations.9

Share your thoughts! Send your Letter to the Editor to [email protected]. Please include your name and the city and state in which you practice.

References

 

 

  1. Manson JE, Chlebowski RT, Stefanick ML, et al. Menopausal hormone therapy and health outcomes during the intervention and extended poststopping phases of the Women’s Health Initiative randomized trials. JAMA. 2013;310(13):1353–1368.
  2. Hodis HN, Mack WJ, Shoupe D, et al. Methods and baseline cardiovascular data from the Early versus Late Intervention Trial with Estradiol testing the menopausal hormone timing hypothesis. Menopause. 2015;22(4):391–401.
  3. Manson JE, Kaunitz AM. Menopause management—getting clinical care back on track. N Engl J Med. 2016;374(9):803–806.
  4. Rossouw JE, Anderson GL, Prentice RL, et al; Writing Group for the Women’s Health Initiative Investigators. Risks and benefits of estrogen plus progestin in healthy postmenopausal women: principal results From the Women’s Health Initiative randomized controlled trial. JAMA. 2002;288(3):321–333.
  5. Kaunitz AM, Kaunitz JD. Compounded bioidentical hormone therapy: time for a reality check? Menopause. 2015;22(9):919–920.
  6. Kaunitz AM, Manson JE. Management of menopausal symptoms. Obstet Gynecol. 2015;126(4):859–876.
  7. Benster B, Carey A, Wadsworth F, Vashisht A, Domoney C, Studd J. A double-blind placebo-controlled study to evaluate the effect of progestelle progesterone cream on postmenopausal women. Menopause Int. 2009;15(2):63–69.
  8. American College of Obstetricians Gynecologists. ACOG Practice Bulletin No. 141: management of menopausal symptoms. Obstet Gynecol. 2014;123(1):202–216.
  9. North American Menopause Society. The 2012 hormone therapy position statement of The North American Menopause Society. Menopause. 2012;19(3):257–271.
References

 

 

  1. Manson JE, Chlebowski RT, Stefanick ML, et al. Menopausal hormone therapy and health outcomes during the intervention and extended poststopping phases of the Women’s Health Initiative randomized trials. JAMA. 2013;310(13):1353–1368.
  2. Hodis HN, Mack WJ, Shoupe D, et al. Methods and baseline cardiovascular data from the Early versus Late Intervention Trial with Estradiol testing the menopausal hormone timing hypothesis. Menopause. 2015;22(4):391–401.
  3. Manson JE, Kaunitz AM. Menopause management—getting clinical care back on track. N Engl J Med. 2016;374(9):803–806.
  4. Rossouw JE, Anderson GL, Prentice RL, et al; Writing Group for the Women’s Health Initiative Investigators. Risks and benefits of estrogen plus progestin in healthy postmenopausal women: principal results From the Women’s Health Initiative randomized controlled trial. JAMA. 2002;288(3):321–333.
  5. Kaunitz AM, Kaunitz JD. Compounded bioidentical hormone therapy: time for a reality check? Menopause. 2015;22(9):919–920.
  6. Kaunitz AM, Manson JE. Management of menopausal symptoms. Obstet Gynecol. 2015;126(4):859–876.
  7. Benster B, Carey A, Wadsworth F, Vashisht A, Domoney C, Studd J. A double-blind placebo-controlled study to evaluate the effect of progestelle progesterone cream on postmenopausal women. Menopause Int. 2009;15(2):63–69.
  8. American College of Obstetricians Gynecologists. ACOG Practice Bulletin No. 141: management of menopausal symptoms. Obstet Gynecol. 2014;123(1):202–216.
  9. North American Menopause Society. The 2012 hormone therapy position statement of The North American Menopause Society. Menopause. 2012;19(3):257–271.
Issue
OBG Management - 28(7)
Issue
OBG Management - 28(7)
Page Number
38-43
Page Number
38-43
Publications
Publications
Topics
Article Type
Display Headline
2016 Update on menopause
Display Headline
2016 Update on menopause
Legacy Keywords
Andrew M. Kaunitz MD, JoAnn Manson MD DrPh, menopause, vardiovascular safety, hormone therapy, HT, early menopausal women, compounded HT preparations, FDA, Women's Health Initiative, WHI, timing hypothesis, atherosclerosis, estrogen, progesterone
Legacy Keywords
Andrew M. Kaunitz MD, JoAnn Manson MD DrPh, menopause, vardiovascular safety, hormone therapy, HT, early menopausal women, compounded HT preparations, FDA, Women's Health Initiative, WHI, timing hypothesis, atherosclerosis, estrogen, progesterone
Sections
Inside the Article

    In this article

 

• Cardiovascular safety of HT
• JoAnn E. Manson discusses new data on HT benefits vs risks
• Use of compounded hormones growing
Article PDF Media

How do you break the ice with patients to ask about their sexual health?

Article Type
Changed
Wed, 08/29/2018 - 11:00
Display Headline
How do you break the ice with patients to ask about their sexual health?

CASE Patient may benefit from treatment for dyspareuniaA 54-year-old woman has been in your care for more than 15 years. Three years ago, at her well-woman examination, she was not yet having symptoms of menopause. Now, during her current examination, she reports hot flashes, which she says are not bothersome. In passing, she also says, “I don’t want to take hormone therapy,” but then is not overly conversational or responsive to your questions. She does mention having had 3 urinary tract infections over the past 8 months. On physical examination, you note mildly atrophied vaginal tissue.

Your patient does not bring up any sexual concerns, and so far you have not directly asked about sexual health. However, the time remaining in this visit is limited, and your patient, whose daughter is sitting in the waiting area, seems anxious to finish and leave. Still, you want to broach the subject of your patient’s sexual health. What are your best options?

We learned a lot about women’s perceptions regarding their sexual health in the 2008 Prevalence of Female Sexual Problems Associated with Distress and Determinants of Treatment Seeking study (PRESIDE). Approximately 43% of 31,581 questionnaire respondents reported dysfunction in sexual desire, arousal, or orgasm.1 Results also showed that 11.5% of the respondents with any of these types of female sexual dysfunction (FSD) were distressed about it. For clinicians, knowing who these women are is key in recognizing and treating FSD.

Important to the opening case, in PRESIDE, Shifren and colleagues found that women in their midlife years (aged 45 to 64) had the highest rate of any distressing sexual problem: 14.8%. Younger women (aged 18 to 44 years) had a rate of 10.8%; older women (aged 65 years or older) had a rate of 8.9%.1

The most prevalent FSD was hypoactive sexual desire disorder,1 which in 2013 was renamed sexual interest and arousal disorder in the Diagnostic and Statistical Manual of Mental Disorders, Fifth Edition.2 As with any distressing FSD, reports of being distressed about low sexual desire were highest for midlife women (12.3%) relative to younger (8.9%) and older (7.4%) women.1

Unfortunately, decreased desire can have a ripple effect that goes well beyond a patient’s sexual health. A less-than-satisfying sex life can have a significant negative impact on self-image, possibly leading to depression or overall mood instability, which in turn can put undue strain on personal relationships.1,3 A patient’s entire quality of life can be affected negatively.

With so much at stake, it is important for physicians to take a more active role in addressing the sexual health of their patients. Emphasizing wellness can help reduce the stigma of sexual dysfunction, break the silence, and open up patient–physician communication.4 There is also much to be gained by helping patients realize that having positive and respectful relationships is protective for health, including sexual health.4 Likewise, patients benefit from acknowledging that sexual health is an element of overall health and contributes to it.4

Toward these ends, more discussion with patients is needed. According to a 2008 national study, although 63% of US ObGyns surveyed indicated that they routinely asked their patients about sexual activity, only 40% asked about sexual problems, and only 29% asked patients if their sex lives were satisfying.5

Without communication, information is missed, and clinicians easily can overlook their patients’ sexual dysfunction and need for intervention. For midlife women, who are disproportionately affected by dysfunction relative to younger and older women, and for whom the rate of menopausal symptoms increases over the transition years, the results of going undiagnosed and untreated can be especially troubling. As reported in one study, for example, the rate of bothersome vulvovaginal atrophy, which can be a source of sexual dysfunction, increased from less than 5% at premenopause to almost 50% at 3 years postmenopause.6 What is standing in our way, however, and how can we overcome the hurdles to an open-door approach and meaningful conversation?

Obstacles to taking a sexual historyInitiating a sexual history can be like opening Pandora’s box. How do clinicians deal with the problems that come out? Some clinicians worry about embarrassing a patient with the first few questions about sexual health. Male gynecologists may feel awkward asking a patient about sex—particularly an older, midlife patient. The problem with not starting the conversation is that the midlife patient is often the one in the most distress, and the one most in need of treatment. Only by having the sexual health discussion can clinicians identify any issues and begin to address them.

 

 

 

Icebreakers to jump-start the conversation
Asking open-ended questions works best. Here are some options for starting a conversation with a midlife patient:

 

  1. say, “Many women around menopause develop sexual problems. Have you noticed any changes?”
  2. say, “It is part of my routine to ask about sexual health. Tell me if you have any concerns.”
  3. add a brief sexual symptom checklist (FIGURE 1) to the patient history or intake form. The checklist shown here starts by asking if the patient is satisfied, yes or no, with her sexual function. If yes, the satisfied patient (and the clinician) can proceed to the next section on the form. If no, the dissatisfied patient can answer additional questions about problems related to sexual desire, arousal, orgasm, and dyspareunia.

Such tools as checklists are often needed to bridge the wide communication gap between patients and physicians. Of the 255 women who reported experiencing dyspareunia in the Revealing Vaginal Effects at Midlife (REVEAL) study, almost half (44%) indicated that they had not spoken with their health care clinician about it.7 Another 44% had spoken about the problem but on their own initiative. In only 10% of cases had a physician started the conversation.

Clinicians can and should do better. Many of us have known our patients for years—given them their annual examinations, delivered their babies, performed their surgeries, become familiar with their bodies and intimate medical histories. We are uniquely qualified to start conversations on sexual health. A clinician who examines tissues and sees a decrease in vaginal caliber and pallor must say something. In some cases, the vagina is dry, but the patient has not been having lubrication problems. In other cases, a more serious condition might be involved. The important thing is to open up a conversation and talk about treatments.

CASE Continued
As today’s office visit wraps up and your patient begins moving for the door, you say, “Your hot flashes aren’t bothering you, but some women start experiencing certain sexual problems around this time in life. Have you noticed any issues?”

“Well, I have been having more burning during intercourse,” your patient responds.

On hearing this, you say, “That’s very important, Mrs. X, and I am glad you told me about it. I would like to discuss your concern a bit more, so let’s make another appointment to do just that.”

At the next visit, as part of the discussion, you give your patient a 15-minute sexual status examination.

Sexual status examination
Performing this examination helps clinicians see patterns in both sexual behavior and sexual health, which in turn can make it easier to recognize any dysfunction that might subsequently develop. The key to this process is establishing trust with the patient and having her feel comfortable with the discussion.

The patient remains fully clothed during this 15-minute session, which takes place with guarantees of nonjudgmental listening, confidentiality, privacy, and no interruptions. With the topic of sex being so personal, it should be emphasized that she is simply giving the clinician information, as she does on other health-related matters.

Establish her sexual status. Begin by asking the patient to describe her most recent or typical sexual encounter, including details such as day, time, location, type of activity, thoughts and feelings, and responses.

Potential issues can become apparent immediately. A patient may not have had a sexual encounter recently, or ever. Another may want sex, or more sex, but sees obstacles or lack of opportunity. Each of these is an issue to be explored, if the patient allows.

A patient can be sexually active in a number of ways, as the definition varies among population groups (race and age) and individuals. Sex is not only intercourse or oral sex—it is also kissing, touching, and hugging. Some people have an expansive view of what it is to be sexually active. When the patient mentions an encounter, ask what day, what time, where (at home, in a hotel room, at the office), and what type of activity (foreplay, oral sex, manual stimulation, intercourse, and position). Following up, ask what the patient was thinking or feeling about the encounter. For example, were there distracting thoughts or feelings of guilt? How did the patient and her partner respond during the encounter?

Assess for sexual dysfunction. After assessing the patient’s sexual status, turn to dysfunction. Arousal, pain, orgasm, and satisfaction are 4 areas of interest. Did the patient have difficulty becoming aroused? Was there a problem with lubrication? Did she have an orgasm? Was sex painful? How did she feel in terms of overall satisfaction?

 

 

In general, patients are comfortable speaking about sexual function and health. Having this talk can help identify a pattern, which can be discussed further during another visit. Such a follow-up would not take long—a level 3 visit should suffice.

Differential diagnosis. Consider the effects of current medications.8,9 The psychiatric illnesses and general health factors that may affect sexual function should be considered as well (FIGURE 2).10–22

 

When is it important to refer?
There are many reasons to refer a patient to another physician, including:

 

  • a recommended treatment is not working
  • abuse is suspected
  • the patient shows symptoms of depression, anxiety, or another psychiatric condition
  • a chronic, generalized (vs situational) disorder may be involved
  • physical pain issues must be addressed
  • you simply do not feel comfortable with a particular problem or patient.

Given the range of potential issues associated with sexual function, it is important to be able to provide the patient with expert assistance from a multidisciplinary team of specialists. This team can include psychologists, psychiatrists, counselors, sex educators, and, for pain issues, pelvic floor specialists and pelvic floor physical therapists. These colleagues are thoroughly familiar with the kinds of issues that can arise, and can offer alternative and adjunctive therapies.

Referrals also can be made for the latest nonpharmacologic and FDA-approved pharmacologic treatment options. Specialists tend to be familiar with these options, some of which are available only recently.

It is important to ask patients about sexual function and, if necessary, give them access to the best treatment options.

CASE Resolved
During the sexual status examination, your patient describes her most recent sexual encounter with her husband. She is frustrated with her lack of sexual response and describes a dry, tearing sensation during intercourse. You recommend first-line treatment with vaginal lubricants, preferably iso-osmolar aqueous− or silicone/dimethicone−based lubricants during intercourse. You also can discuss topical estrogen therapy via estradiol cream, conjugated equine estrogen cream, estradiol tablets in the vagina, or the estrogen ring. She is reassured that topical estrogen use will not pose significant risk for cancer, stroke, heart disease, or blood clot and that progesterone treatment is not necessary.

For patients who are particularly concerned about vaginal estrogen use, 2 or 3 times weekly use of a vaginal moisturizer could be an alternative for genitourinary symptoms and dyspareunia.


Share your thoughts on this article! Send your Letter to the Editor to [email protected]. Please include your name and the city and state in which you practice.

References
  1. Shifren JL, Monz BU, Russo PA, Segreti A, Johannes CB. Sexual problems and distress in United States women: prevalence and correlates. Obstet Gynecol. 2008;112(5):970−978.
  2. American Psychiatric Association. Diagnostic and Statistical Manual of Mental Disorders, Fifth Edition (DSM-5). Washington, DC: American Psychiatric Association; 2013.
  3. Leiblum SR, Koochaki PE, Rodenberg CA, Barton IP, Rosen RC. Hypoactive sexual desire disorder in postmenopausal women: US results from the Women’s International Study of Health and Sexuality (WISHeS). Menopause. 2006;13(1):46−56.
  4. Satcher D, Hook EW 3rd, Coleman E. Sexual health in America: improving patient care and public health. JAMA. 2015;314(8):765−766.
  5. Sobecki JN, Curlin FA, Rasinski KA, Lindau ST. What we don’t talk about when we don’t talk about sex: results of a national survey of U.S. obstetrician/gynecologists. J Sex Med. 2012;9(5):1285−1294.
  6. Dennerstein L, Dudley EC, Hopper JL, Guthrie JR, Burger HG. A prospective population-based study of menopausal symptoms. Obstet Gynecol. 2000;96(3):351−358.
  7. Shifren JL, Johannes CB, Monz BU, Russo PA, Bennett L, Rosen R. Help-seeking behavior of women with self-reported distressing sexual problems. J Womens Health. 2009;18(4):461−468.
  8. Basson R, Schultz WW. Sexual sequelae of general medical disorders. Lancet. 2007;369(9559):409−424.
  9. Kingsberg SA, Janata JW. Female sexual disorders: assessment, diagnosis, and treatment. Urol Clin North Am. 2007;34(4):497−506, v−vi.
  10. Casper RC, Redmond DE Jr, Katz MM, Schaffer CB, Davis JM, Koslow SH. Somatic symptoms in primary affective disorder. Presence and relationship to the classification of depression. Arch Gen Psychiatry. 1985;42(11):1098−1104.
  11. van Lankveld JJ, Grotjohann Y. Psychiatric comorbidity in heterosexual couples with sexual dysfunction assessed with the Composite International Diagnostic Interview. Arch Sex Behav. 2000;29(5):479−498.
  12. Shifren JL, Monz BU, Russo PA, Segreti A, Johannes CB. Sexual problems and distress in United States women: prevalence and correlates. Obstet Gynecol. 2008;112(5):970−978.
  13. Friedman S, Harrison G. Sexual histories, attitudes, and behavior of schizophrenic and “normal” women. Arch Sex Behav. 1984;13(6):555−567.
  14. Okeahialam BN, Obeka NC. Sexual dysfunction in female hypertensives. J Natl Med Assoc. 2006;98(4):638−640.
  15. Rees PM, Fowler CJ, Maas CP. Sexual function in men and women with neurological disorders. Lancet. 2007;369(9560):512−525.
  16. Bhasin S, Enzlin P, Coviello A, Basson R. Sexual dysfunction in men and women with endocrine disorders. Lancet. 2007;369(9561):597−611.
  17. Aslan G, KöseoTimesğlu H, Sadik O, Gimen S, Cihan A, Esen A. Sexual function in women with urinary incontinence. Int J Impot Res. 2005;17(3):248−251.
  18. Smith EM, Ritchie JM, Galask R, Pugh EE, Jia J, Ricks-McGillan J. Case–control study of vulvar vestibulitis risk associated with genital infections. Infect Dis Obstet Gynecol. 2002;10(4):193−202.
  19. Baksu B, Davas I, Agar E, Akyol A, Varolan A. The effect of mode of delivery on postpartum sexual functioning in primiparous women. Int Urogynecol J Pelvic Floor Dysfunct. 2007;18(4):401−406.
  20. Abdel-Nasser AM, Ali EI. Determinants of sexual disability and dissatisfaction in female patients with rheumatoid arthritis. Clin Rheumatol. 2006;25(6):822−830.
  21. Sampogna F, Gisondi P, Tabolli S, Abeni D; IDI Multipurpose Psoriasis Research on Vital Experiences investigators. Impairment of sexual life in patients with psoriasis. Dermatology. 2007;214(2):144−150.
  22. Mathias C, Cardeal Mendes CM, Pondé de Sena E, et al. An open-label, fixed-dose study of bupropion effect on sexual function scores in women treated for breast cancer. Ann Oncol. 2006;17(12):1792−1796.
Article PDF
Author and Disclosure Information

Dr. Iglesia is Director, Section of Female Pelvic Medicine and Reconstructive Surgery, MedStar Washington Hospital Center, Washington, DC, Professor, Departments of Obstetrics/Gynecology and Urology, Georgetown University School of Medicine, Washington, DC. Dr. Iglesia serves on the OBG Management Board of Editors.

The author reports no financial relationships relevant to this article.

Issue
OBG Management - 28(7)
Publications
Topics
Page Number
19, 20, 22-24
Legacy Keywords
Cheryl B. Iglesia MD, sexual health, sexual dysfunction, dyspareunia, well woman examination, menopausal symptoms, hormone therapy, female sexual dysfunction, FSD, premenopause, conversation, checklists, dry vagina, sexual arousal, orgasm, sexual desire, abuse, pelvic pain
Sections
Author and Disclosure Information

Dr. Iglesia is Director, Section of Female Pelvic Medicine and Reconstructive Surgery, MedStar Washington Hospital Center, Washington, DC, Professor, Departments of Obstetrics/Gynecology and Urology, Georgetown University School of Medicine, Washington, DC. Dr. Iglesia serves on the OBG Management Board of Editors.

The author reports no financial relationships relevant to this article.

Author and Disclosure Information

Dr. Iglesia is Director, Section of Female Pelvic Medicine and Reconstructive Surgery, MedStar Washington Hospital Center, Washington, DC, Professor, Departments of Obstetrics/Gynecology and Urology, Georgetown University School of Medicine, Washington, DC. Dr. Iglesia serves on the OBG Management Board of Editors.

The author reports no financial relationships relevant to this article.

Article PDF
Article PDF
Related Articles

CASE Patient may benefit from treatment for dyspareuniaA 54-year-old woman has been in your care for more than 15 years. Three years ago, at her well-woman examination, she was not yet having symptoms of menopause. Now, during her current examination, she reports hot flashes, which she says are not bothersome. In passing, she also says, “I don’t want to take hormone therapy,” but then is not overly conversational or responsive to your questions. She does mention having had 3 urinary tract infections over the past 8 months. On physical examination, you note mildly atrophied vaginal tissue.

Your patient does not bring up any sexual concerns, and so far you have not directly asked about sexual health. However, the time remaining in this visit is limited, and your patient, whose daughter is sitting in the waiting area, seems anxious to finish and leave. Still, you want to broach the subject of your patient’s sexual health. What are your best options?

We learned a lot about women’s perceptions regarding their sexual health in the 2008 Prevalence of Female Sexual Problems Associated with Distress and Determinants of Treatment Seeking study (PRESIDE). Approximately 43% of 31,581 questionnaire respondents reported dysfunction in sexual desire, arousal, or orgasm.1 Results also showed that 11.5% of the respondents with any of these types of female sexual dysfunction (FSD) were distressed about it. For clinicians, knowing who these women are is key in recognizing and treating FSD.

Important to the opening case, in PRESIDE, Shifren and colleagues found that women in their midlife years (aged 45 to 64) had the highest rate of any distressing sexual problem: 14.8%. Younger women (aged 18 to 44 years) had a rate of 10.8%; older women (aged 65 years or older) had a rate of 8.9%.1

The most prevalent FSD was hypoactive sexual desire disorder,1 which in 2013 was renamed sexual interest and arousal disorder in the Diagnostic and Statistical Manual of Mental Disorders, Fifth Edition.2 As with any distressing FSD, reports of being distressed about low sexual desire were highest for midlife women (12.3%) relative to younger (8.9%) and older (7.4%) women.1

Unfortunately, decreased desire can have a ripple effect that goes well beyond a patient’s sexual health. A less-than-satisfying sex life can have a significant negative impact on self-image, possibly leading to depression or overall mood instability, which in turn can put undue strain on personal relationships.1,3 A patient’s entire quality of life can be affected negatively.

With so much at stake, it is important for physicians to take a more active role in addressing the sexual health of their patients. Emphasizing wellness can help reduce the stigma of sexual dysfunction, break the silence, and open up patient–physician communication.4 There is also much to be gained by helping patients realize that having positive and respectful relationships is protective for health, including sexual health.4 Likewise, patients benefit from acknowledging that sexual health is an element of overall health and contributes to it.4

Toward these ends, more discussion with patients is needed. According to a 2008 national study, although 63% of US ObGyns surveyed indicated that they routinely asked their patients about sexual activity, only 40% asked about sexual problems, and only 29% asked patients if their sex lives were satisfying.5

Without communication, information is missed, and clinicians easily can overlook their patients’ sexual dysfunction and need for intervention. For midlife women, who are disproportionately affected by dysfunction relative to younger and older women, and for whom the rate of menopausal symptoms increases over the transition years, the results of going undiagnosed and untreated can be especially troubling. As reported in one study, for example, the rate of bothersome vulvovaginal atrophy, which can be a source of sexual dysfunction, increased from less than 5% at premenopause to almost 50% at 3 years postmenopause.6 What is standing in our way, however, and how can we overcome the hurdles to an open-door approach and meaningful conversation?

Obstacles to taking a sexual historyInitiating a sexual history can be like opening Pandora’s box. How do clinicians deal with the problems that come out? Some clinicians worry about embarrassing a patient with the first few questions about sexual health. Male gynecologists may feel awkward asking a patient about sex—particularly an older, midlife patient. The problem with not starting the conversation is that the midlife patient is often the one in the most distress, and the one most in need of treatment. Only by having the sexual health discussion can clinicians identify any issues and begin to address them.

 

 

 

Icebreakers to jump-start the conversation
Asking open-ended questions works best. Here are some options for starting a conversation with a midlife patient:

 

  1. say, “Many women around menopause develop sexual problems. Have you noticed any changes?”
  2. say, “It is part of my routine to ask about sexual health. Tell me if you have any concerns.”
  3. add a brief sexual symptom checklist (FIGURE 1) to the patient history or intake form. The checklist shown here starts by asking if the patient is satisfied, yes or no, with her sexual function. If yes, the satisfied patient (and the clinician) can proceed to the next section on the form. If no, the dissatisfied patient can answer additional questions about problems related to sexual desire, arousal, orgasm, and dyspareunia.

Such tools as checklists are often needed to bridge the wide communication gap between patients and physicians. Of the 255 women who reported experiencing dyspareunia in the Revealing Vaginal Effects at Midlife (REVEAL) study, almost half (44%) indicated that they had not spoken with their health care clinician about it.7 Another 44% had spoken about the problem but on their own initiative. In only 10% of cases had a physician started the conversation.

Clinicians can and should do better. Many of us have known our patients for years—given them their annual examinations, delivered their babies, performed their surgeries, become familiar with their bodies and intimate medical histories. We are uniquely qualified to start conversations on sexual health. A clinician who examines tissues and sees a decrease in vaginal caliber and pallor must say something. In some cases, the vagina is dry, but the patient has not been having lubrication problems. In other cases, a more serious condition might be involved. The important thing is to open up a conversation and talk about treatments.

CASE Continued
As today’s office visit wraps up and your patient begins moving for the door, you say, “Your hot flashes aren’t bothering you, but some women start experiencing certain sexual problems around this time in life. Have you noticed any issues?”

“Well, I have been having more burning during intercourse,” your patient responds.

On hearing this, you say, “That’s very important, Mrs. X, and I am glad you told me about it. I would like to discuss your concern a bit more, so let’s make another appointment to do just that.”

At the next visit, as part of the discussion, you give your patient a 15-minute sexual status examination.

Sexual status examination
Performing this examination helps clinicians see patterns in both sexual behavior and sexual health, which in turn can make it easier to recognize any dysfunction that might subsequently develop. The key to this process is establishing trust with the patient and having her feel comfortable with the discussion.

The patient remains fully clothed during this 15-minute session, which takes place with guarantees of nonjudgmental listening, confidentiality, privacy, and no interruptions. With the topic of sex being so personal, it should be emphasized that she is simply giving the clinician information, as she does on other health-related matters.

Establish her sexual status. Begin by asking the patient to describe her most recent or typical sexual encounter, including details such as day, time, location, type of activity, thoughts and feelings, and responses.

Potential issues can become apparent immediately. A patient may not have had a sexual encounter recently, or ever. Another may want sex, or more sex, but sees obstacles or lack of opportunity. Each of these is an issue to be explored, if the patient allows.

A patient can be sexually active in a number of ways, as the definition varies among population groups (race and age) and individuals. Sex is not only intercourse or oral sex—it is also kissing, touching, and hugging. Some people have an expansive view of what it is to be sexually active. When the patient mentions an encounter, ask what day, what time, where (at home, in a hotel room, at the office), and what type of activity (foreplay, oral sex, manual stimulation, intercourse, and position). Following up, ask what the patient was thinking or feeling about the encounter. For example, were there distracting thoughts or feelings of guilt? How did the patient and her partner respond during the encounter?

Assess for sexual dysfunction. After assessing the patient’s sexual status, turn to dysfunction. Arousal, pain, orgasm, and satisfaction are 4 areas of interest. Did the patient have difficulty becoming aroused? Was there a problem with lubrication? Did she have an orgasm? Was sex painful? How did she feel in terms of overall satisfaction?

 

 

In general, patients are comfortable speaking about sexual function and health. Having this talk can help identify a pattern, which can be discussed further during another visit. Such a follow-up would not take long—a level 3 visit should suffice.

Differential diagnosis. Consider the effects of current medications.8,9 The psychiatric illnesses and general health factors that may affect sexual function should be considered as well (FIGURE 2).10–22

 

When is it important to refer?
There are many reasons to refer a patient to another physician, including:

 

  • a recommended treatment is not working
  • abuse is suspected
  • the patient shows symptoms of depression, anxiety, or another psychiatric condition
  • a chronic, generalized (vs situational) disorder may be involved
  • physical pain issues must be addressed
  • you simply do not feel comfortable with a particular problem or patient.

Given the range of potential issues associated with sexual function, it is important to be able to provide the patient with expert assistance from a multidisciplinary team of specialists. This team can include psychologists, psychiatrists, counselors, sex educators, and, for pain issues, pelvic floor specialists and pelvic floor physical therapists. These colleagues are thoroughly familiar with the kinds of issues that can arise, and can offer alternative and adjunctive therapies.

Referrals also can be made for the latest nonpharmacologic and FDA-approved pharmacologic treatment options. Specialists tend to be familiar with these options, some of which are available only recently.

It is important to ask patients about sexual function and, if necessary, give them access to the best treatment options.

CASE Resolved
During the sexual status examination, your patient describes her most recent sexual encounter with her husband. She is frustrated with her lack of sexual response and describes a dry, tearing sensation during intercourse. You recommend first-line treatment with vaginal lubricants, preferably iso-osmolar aqueous− or silicone/dimethicone−based lubricants during intercourse. You also can discuss topical estrogen therapy via estradiol cream, conjugated equine estrogen cream, estradiol tablets in the vagina, or the estrogen ring. She is reassured that topical estrogen use will not pose significant risk for cancer, stroke, heart disease, or blood clot and that progesterone treatment is not necessary.

For patients who are particularly concerned about vaginal estrogen use, 2 or 3 times weekly use of a vaginal moisturizer could be an alternative for genitourinary symptoms and dyspareunia.


Share your thoughts on this article! Send your Letter to the Editor to [email protected]. Please include your name and the city and state in which you practice.

CASE Patient may benefit from treatment for dyspareuniaA 54-year-old woman has been in your care for more than 15 years. Three years ago, at her well-woman examination, she was not yet having symptoms of menopause. Now, during her current examination, she reports hot flashes, which she says are not bothersome. In passing, she also says, “I don’t want to take hormone therapy,” but then is not overly conversational or responsive to your questions. She does mention having had 3 urinary tract infections over the past 8 months. On physical examination, you note mildly atrophied vaginal tissue.

Your patient does not bring up any sexual concerns, and so far you have not directly asked about sexual health. However, the time remaining in this visit is limited, and your patient, whose daughter is sitting in the waiting area, seems anxious to finish and leave. Still, you want to broach the subject of your patient’s sexual health. What are your best options?

We learned a lot about women’s perceptions regarding their sexual health in the 2008 Prevalence of Female Sexual Problems Associated with Distress and Determinants of Treatment Seeking study (PRESIDE). Approximately 43% of 31,581 questionnaire respondents reported dysfunction in sexual desire, arousal, or orgasm.1 Results also showed that 11.5% of the respondents with any of these types of female sexual dysfunction (FSD) were distressed about it. For clinicians, knowing who these women are is key in recognizing and treating FSD.

Important to the opening case, in PRESIDE, Shifren and colleagues found that women in their midlife years (aged 45 to 64) had the highest rate of any distressing sexual problem: 14.8%. Younger women (aged 18 to 44 years) had a rate of 10.8%; older women (aged 65 years or older) had a rate of 8.9%.1

The most prevalent FSD was hypoactive sexual desire disorder,1 which in 2013 was renamed sexual interest and arousal disorder in the Diagnostic and Statistical Manual of Mental Disorders, Fifth Edition.2 As with any distressing FSD, reports of being distressed about low sexual desire were highest for midlife women (12.3%) relative to younger (8.9%) and older (7.4%) women.1

Unfortunately, decreased desire can have a ripple effect that goes well beyond a patient’s sexual health. A less-than-satisfying sex life can have a significant negative impact on self-image, possibly leading to depression or overall mood instability, which in turn can put undue strain on personal relationships.1,3 A patient’s entire quality of life can be affected negatively.

With so much at stake, it is important for physicians to take a more active role in addressing the sexual health of their patients. Emphasizing wellness can help reduce the stigma of sexual dysfunction, break the silence, and open up patient–physician communication.4 There is also much to be gained by helping patients realize that having positive and respectful relationships is protective for health, including sexual health.4 Likewise, patients benefit from acknowledging that sexual health is an element of overall health and contributes to it.4

Toward these ends, more discussion with patients is needed. According to a 2008 national study, although 63% of US ObGyns surveyed indicated that they routinely asked their patients about sexual activity, only 40% asked about sexual problems, and only 29% asked patients if their sex lives were satisfying.5

Without communication, information is missed, and clinicians easily can overlook their patients’ sexual dysfunction and need for intervention. For midlife women, who are disproportionately affected by dysfunction relative to younger and older women, and for whom the rate of menopausal symptoms increases over the transition years, the results of going undiagnosed and untreated can be especially troubling. As reported in one study, for example, the rate of bothersome vulvovaginal atrophy, which can be a source of sexual dysfunction, increased from less than 5% at premenopause to almost 50% at 3 years postmenopause.6 What is standing in our way, however, and how can we overcome the hurdles to an open-door approach and meaningful conversation?

Obstacles to taking a sexual historyInitiating a sexual history can be like opening Pandora’s box. How do clinicians deal with the problems that come out? Some clinicians worry about embarrassing a patient with the first few questions about sexual health. Male gynecologists may feel awkward asking a patient about sex—particularly an older, midlife patient. The problem with not starting the conversation is that the midlife patient is often the one in the most distress, and the one most in need of treatment. Only by having the sexual health discussion can clinicians identify any issues and begin to address them.

 

 

 

Icebreakers to jump-start the conversation
Asking open-ended questions works best. Here are some options for starting a conversation with a midlife patient:

 

  1. say, “Many women around menopause develop sexual problems. Have you noticed any changes?”
  2. say, “It is part of my routine to ask about sexual health. Tell me if you have any concerns.”
  3. add a brief sexual symptom checklist (FIGURE 1) to the patient history or intake form. The checklist shown here starts by asking if the patient is satisfied, yes or no, with her sexual function. If yes, the satisfied patient (and the clinician) can proceed to the next section on the form. If no, the dissatisfied patient can answer additional questions about problems related to sexual desire, arousal, orgasm, and dyspareunia.

Such tools as checklists are often needed to bridge the wide communication gap between patients and physicians. Of the 255 women who reported experiencing dyspareunia in the Revealing Vaginal Effects at Midlife (REVEAL) study, almost half (44%) indicated that they had not spoken with their health care clinician about it.7 Another 44% had spoken about the problem but on their own initiative. In only 10% of cases had a physician started the conversation.

Clinicians can and should do better. Many of us have known our patients for years—given them their annual examinations, delivered their babies, performed their surgeries, become familiar with their bodies and intimate medical histories. We are uniquely qualified to start conversations on sexual health. A clinician who examines tissues and sees a decrease in vaginal caliber and pallor must say something. In some cases, the vagina is dry, but the patient has not been having lubrication problems. In other cases, a more serious condition might be involved. The important thing is to open up a conversation and talk about treatments.

CASE Continued
As today’s office visit wraps up and your patient begins moving for the door, you say, “Your hot flashes aren’t bothering you, but some women start experiencing certain sexual problems around this time in life. Have you noticed any issues?”

“Well, I have been having more burning during intercourse,” your patient responds.

On hearing this, you say, “That’s very important, Mrs. X, and I am glad you told me about it. I would like to discuss your concern a bit more, so let’s make another appointment to do just that.”

At the next visit, as part of the discussion, you give your patient a 15-minute sexual status examination.

Sexual status examination
Performing this examination helps clinicians see patterns in both sexual behavior and sexual health, which in turn can make it easier to recognize any dysfunction that might subsequently develop. The key to this process is establishing trust with the patient and having her feel comfortable with the discussion.

The patient remains fully clothed during this 15-minute session, which takes place with guarantees of nonjudgmental listening, confidentiality, privacy, and no interruptions. With the topic of sex being so personal, it should be emphasized that she is simply giving the clinician information, as she does on other health-related matters.

Establish her sexual status. Begin by asking the patient to describe her most recent or typical sexual encounter, including details such as day, time, location, type of activity, thoughts and feelings, and responses.

Potential issues can become apparent immediately. A patient may not have had a sexual encounter recently, or ever. Another may want sex, or more sex, but sees obstacles or lack of opportunity. Each of these is an issue to be explored, if the patient allows.

A patient can be sexually active in a number of ways, as the definition varies among population groups (race and age) and individuals. Sex is not only intercourse or oral sex—it is also kissing, touching, and hugging. Some people have an expansive view of what it is to be sexually active. When the patient mentions an encounter, ask what day, what time, where (at home, in a hotel room, at the office), and what type of activity (foreplay, oral sex, manual stimulation, intercourse, and position). Following up, ask what the patient was thinking or feeling about the encounter. For example, were there distracting thoughts or feelings of guilt? How did the patient and her partner respond during the encounter?

Assess for sexual dysfunction. After assessing the patient’s sexual status, turn to dysfunction. Arousal, pain, orgasm, and satisfaction are 4 areas of interest. Did the patient have difficulty becoming aroused? Was there a problem with lubrication? Did she have an orgasm? Was sex painful? How did she feel in terms of overall satisfaction?

 

 

In general, patients are comfortable speaking about sexual function and health. Having this talk can help identify a pattern, which can be discussed further during another visit. Such a follow-up would not take long—a level 3 visit should suffice.

Differential diagnosis. Consider the effects of current medications.8,9 The psychiatric illnesses and general health factors that may affect sexual function should be considered as well (FIGURE 2).10–22

 

When is it important to refer?
There are many reasons to refer a patient to another physician, including:

 

  • a recommended treatment is not working
  • abuse is suspected
  • the patient shows symptoms of depression, anxiety, or another psychiatric condition
  • a chronic, generalized (vs situational) disorder may be involved
  • physical pain issues must be addressed
  • you simply do not feel comfortable with a particular problem or patient.

Given the range of potential issues associated with sexual function, it is important to be able to provide the patient with expert assistance from a multidisciplinary team of specialists. This team can include psychologists, psychiatrists, counselors, sex educators, and, for pain issues, pelvic floor specialists and pelvic floor physical therapists. These colleagues are thoroughly familiar with the kinds of issues that can arise, and can offer alternative and adjunctive therapies.

Referrals also can be made for the latest nonpharmacologic and FDA-approved pharmacologic treatment options. Specialists tend to be familiar with these options, some of which are available only recently.

It is important to ask patients about sexual function and, if necessary, give them access to the best treatment options.

CASE Resolved
During the sexual status examination, your patient describes her most recent sexual encounter with her husband. She is frustrated with her lack of sexual response and describes a dry, tearing sensation during intercourse. You recommend first-line treatment with vaginal lubricants, preferably iso-osmolar aqueous− or silicone/dimethicone−based lubricants during intercourse. You also can discuss topical estrogen therapy via estradiol cream, conjugated equine estrogen cream, estradiol tablets in the vagina, or the estrogen ring. She is reassured that topical estrogen use will not pose significant risk for cancer, stroke, heart disease, or blood clot and that progesterone treatment is not necessary.

For patients who are particularly concerned about vaginal estrogen use, 2 or 3 times weekly use of a vaginal moisturizer could be an alternative for genitourinary symptoms and dyspareunia.


Share your thoughts on this article! Send your Letter to the Editor to [email protected]. Please include your name and the city and state in which you practice.

References
  1. Shifren JL, Monz BU, Russo PA, Segreti A, Johannes CB. Sexual problems and distress in United States women: prevalence and correlates. Obstet Gynecol. 2008;112(5):970−978.
  2. American Psychiatric Association. Diagnostic and Statistical Manual of Mental Disorders, Fifth Edition (DSM-5). Washington, DC: American Psychiatric Association; 2013.
  3. Leiblum SR, Koochaki PE, Rodenberg CA, Barton IP, Rosen RC. Hypoactive sexual desire disorder in postmenopausal women: US results from the Women’s International Study of Health and Sexuality (WISHeS). Menopause. 2006;13(1):46−56.
  4. Satcher D, Hook EW 3rd, Coleman E. Sexual health in America: improving patient care and public health. JAMA. 2015;314(8):765−766.
  5. Sobecki JN, Curlin FA, Rasinski KA, Lindau ST. What we don’t talk about when we don’t talk about sex: results of a national survey of U.S. obstetrician/gynecologists. J Sex Med. 2012;9(5):1285−1294.
  6. Dennerstein L, Dudley EC, Hopper JL, Guthrie JR, Burger HG. A prospective population-based study of menopausal symptoms. Obstet Gynecol. 2000;96(3):351−358.
  7. Shifren JL, Johannes CB, Monz BU, Russo PA, Bennett L, Rosen R. Help-seeking behavior of women with self-reported distressing sexual problems. J Womens Health. 2009;18(4):461−468.
  8. Basson R, Schultz WW. Sexual sequelae of general medical disorders. Lancet. 2007;369(9559):409−424.
  9. Kingsberg SA, Janata JW. Female sexual disorders: assessment, diagnosis, and treatment. Urol Clin North Am. 2007;34(4):497−506, v−vi.
  10. Casper RC, Redmond DE Jr, Katz MM, Schaffer CB, Davis JM, Koslow SH. Somatic symptoms in primary affective disorder. Presence and relationship to the classification of depression. Arch Gen Psychiatry. 1985;42(11):1098−1104.
  11. van Lankveld JJ, Grotjohann Y. Psychiatric comorbidity in heterosexual couples with sexual dysfunction assessed with the Composite International Diagnostic Interview. Arch Sex Behav. 2000;29(5):479−498.
  12. Shifren JL, Monz BU, Russo PA, Segreti A, Johannes CB. Sexual problems and distress in United States women: prevalence and correlates. Obstet Gynecol. 2008;112(5):970−978.
  13. Friedman S, Harrison G. Sexual histories, attitudes, and behavior of schizophrenic and “normal” women. Arch Sex Behav. 1984;13(6):555−567.
  14. Okeahialam BN, Obeka NC. Sexual dysfunction in female hypertensives. J Natl Med Assoc. 2006;98(4):638−640.
  15. Rees PM, Fowler CJ, Maas CP. Sexual function in men and women with neurological disorders. Lancet. 2007;369(9560):512−525.
  16. Bhasin S, Enzlin P, Coviello A, Basson R. Sexual dysfunction in men and women with endocrine disorders. Lancet. 2007;369(9561):597−611.
  17. Aslan G, KöseoTimesğlu H, Sadik O, Gimen S, Cihan A, Esen A. Sexual function in women with urinary incontinence. Int J Impot Res. 2005;17(3):248−251.
  18. Smith EM, Ritchie JM, Galask R, Pugh EE, Jia J, Ricks-McGillan J. Case–control study of vulvar vestibulitis risk associated with genital infections. Infect Dis Obstet Gynecol. 2002;10(4):193−202.
  19. Baksu B, Davas I, Agar E, Akyol A, Varolan A. The effect of mode of delivery on postpartum sexual functioning in primiparous women. Int Urogynecol J Pelvic Floor Dysfunct. 2007;18(4):401−406.
  20. Abdel-Nasser AM, Ali EI. Determinants of sexual disability and dissatisfaction in female patients with rheumatoid arthritis. Clin Rheumatol. 2006;25(6):822−830.
  21. Sampogna F, Gisondi P, Tabolli S, Abeni D; IDI Multipurpose Psoriasis Research on Vital Experiences investigators. Impairment of sexual life in patients with psoriasis. Dermatology. 2007;214(2):144−150.
  22. Mathias C, Cardeal Mendes CM, Pondé de Sena E, et al. An open-label, fixed-dose study of bupropion effect on sexual function scores in women treated for breast cancer. Ann Oncol. 2006;17(12):1792−1796.
References
  1. Shifren JL, Monz BU, Russo PA, Segreti A, Johannes CB. Sexual problems and distress in United States women: prevalence and correlates. Obstet Gynecol. 2008;112(5):970−978.
  2. American Psychiatric Association. Diagnostic and Statistical Manual of Mental Disorders, Fifth Edition (DSM-5). Washington, DC: American Psychiatric Association; 2013.
  3. Leiblum SR, Koochaki PE, Rodenberg CA, Barton IP, Rosen RC. Hypoactive sexual desire disorder in postmenopausal women: US results from the Women’s International Study of Health and Sexuality (WISHeS). Menopause. 2006;13(1):46−56.
  4. Satcher D, Hook EW 3rd, Coleman E. Sexual health in America: improving patient care and public health. JAMA. 2015;314(8):765−766.
  5. Sobecki JN, Curlin FA, Rasinski KA, Lindau ST. What we don’t talk about when we don’t talk about sex: results of a national survey of U.S. obstetrician/gynecologists. J Sex Med. 2012;9(5):1285−1294.
  6. Dennerstein L, Dudley EC, Hopper JL, Guthrie JR, Burger HG. A prospective population-based study of menopausal symptoms. Obstet Gynecol. 2000;96(3):351−358.
  7. Shifren JL, Johannes CB, Monz BU, Russo PA, Bennett L, Rosen R. Help-seeking behavior of women with self-reported distressing sexual problems. J Womens Health. 2009;18(4):461−468.
  8. Basson R, Schultz WW. Sexual sequelae of general medical disorders. Lancet. 2007;369(9559):409−424.
  9. Kingsberg SA, Janata JW. Female sexual disorders: assessment, diagnosis, and treatment. Urol Clin North Am. 2007;34(4):497−506, v−vi.
  10. Casper RC, Redmond DE Jr, Katz MM, Schaffer CB, Davis JM, Koslow SH. Somatic symptoms in primary affective disorder. Presence and relationship to the classification of depression. Arch Gen Psychiatry. 1985;42(11):1098−1104.
  11. van Lankveld JJ, Grotjohann Y. Psychiatric comorbidity in heterosexual couples with sexual dysfunction assessed with the Composite International Diagnostic Interview. Arch Sex Behav. 2000;29(5):479−498.
  12. Shifren JL, Monz BU, Russo PA, Segreti A, Johannes CB. Sexual problems and distress in United States women: prevalence and correlates. Obstet Gynecol. 2008;112(5):970−978.
  13. Friedman S, Harrison G. Sexual histories, attitudes, and behavior of schizophrenic and “normal” women. Arch Sex Behav. 1984;13(6):555−567.
  14. Okeahialam BN, Obeka NC. Sexual dysfunction in female hypertensives. J Natl Med Assoc. 2006;98(4):638−640.
  15. Rees PM, Fowler CJ, Maas CP. Sexual function in men and women with neurological disorders. Lancet. 2007;369(9560):512−525.
  16. Bhasin S, Enzlin P, Coviello A, Basson R. Sexual dysfunction in men and women with endocrine disorders. Lancet. 2007;369(9561):597−611.
  17. Aslan G, KöseoTimesğlu H, Sadik O, Gimen S, Cihan A, Esen A. Sexual function in women with urinary incontinence. Int J Impot Res. 2005;17(3):248−251.
  18. Smith EM, Ritchie JM, Galask R, Pugh EE, Jia J, Ricks-McGillan J. Case–control study of vulvar vestibulitis risk associated with genital infections. Infect Dis Obstet Gynecol. 2002;10(4):193−202.
  19. Baksu B, Davas I, Agar E, Akyol A, Varolan A. The effect of mode of delivery on postpartum sexual functioning in primiparous women. Int Urogynecol J Pelvic Floor Dysfunct. 2007;18(4):401−406.
  20. Abdel-Nasser AM, Ali EI. Determinants of sexual disability and dissatisfaction in female patients with rheumatoid arthritis. Clin Rheumatol. 2006;25(6):822−830.
  21. Sampogna F, Gisondi P, Tabolli S, Abeni D; IDI Multipurpose Psoriasis Research on Vital Experiences investigators. Impairment of sexual life in patients with psoriasis. Dermatology. 2007;214(2):144−150.
  22. Mathias C, Cardeal Mendes CM, Pondé de Sena E, et al. An open-label, fixed-dose study of bupropion effect on sexual function scores in women treated for breast cancer. Ann Oncol. 2006;17(12):1792−1796.
Issue
OBG Management - 28(7)
Issue
OBG Management - 28(7)
Page Number
19, 20, 22-24
Page Number
19, 20, 22-24
Publications
Publications
Topics
Article Type
Display Headline
How do you break the ice with patients to ask about their sexual health?
Display Headline
How do you break the ice with patients to ask about their sexual health?
Legacy Keywords
Cheryl B. Iglesia MD, sexual health, sexual dysfunction, dyspareunia, well woman examination, menopausal symptoms, hormone therapy, female sexual dysfunction, FSD, premenopause, conversation, checklists, dry vagina, sexual arousal, orgasm, sexual desire, abuse, pelvic pain
Legacy Keywords
Cheryl B. Iglesia MD, sexual health, sexual dysfunction, dyspareunia, well woman examination, menopausal symptoms, hormone therapy, female sexual dysfunction, FSD, premenopause, conversation, checklists, dry vagina, sexual arousal, orgasm, sexual desire, abuse, pelvic pain
Sections
Inside the Article

    In this Article

 

  • Conversation icebreakers
  • The sexual status examination
  • When to refer
Disallow All Ads
Alternative CME
Use ProPublica
Article PDF Media

Acellular Dermal Matrix in Rotator Cuff Surgery

Article Type
Changed
Thu, 09/19/2019 - 13:25
Display Headline
Acellular Dermal Matrix in Rotator Cuff Surgery

Rotator cuff repairs (RCRs) can be challenging due to poor tendon quality and the inability of tendon to heal to bone. Smoking, age over 63 years, fatty infiltration, and massive cuff tears are all factors implicated in increased failure rates.1-3 Tears >3 cm have a structural failure rate ranging from 11% to 95% in the literature.1-5 Massive tears (tears >5 cm or involving 2 or more tendons) are even more complex and have failure rates of 20% to 90%.5,6 The weakest link in the RCR construct is the suture-tendon interface, and suture pullout through the tendon is thought to be the most common method of failure.6 The purpose of this review is to examine whether literature supports the use of acellular dermal matrices (ADMs) in rotator cuff surgery.

The high rate of structural failures after RCR has led surgeons to seek means to augment repairs and new means of reconstruction for irreparable tears. Freeze dried allograft tendons have been used historically with mixed results, including reports of complete graft failures and foreign body reaction.7-10 Porcine intestinal submucosal membrane “patches” gained popularity due to off-the- shelf availability of the graft. However, these were found to have poor outcomes with early graft rejection and intense inflammatory reaction.11,12 Recently, ADMs have gained significant interest due to favorable biomechanical properties and clinical outcomes.13-19

An ADM is an allograft composed of mostly type I collagen that is processed to remove donor cells while preserving the extracellular matrix. There are several commercially available ADMs with different methods of processing and sterilization, as well as handling characteristics.20,21 In vivo studies have demonstrated that removing the cellular components allows infiltration of native cellular agents, such as fibroblasts, vascular tissue, and tenocytes, while causing minimal host inflammatory reaction.21-23 In addition, superior suture pullout strength has been demonstrated by multiple benchtop and preclinical studies.23,24 Therefore, ADMs play a dual role of strengthening the repair while allowing infiltration of host cells and growth factors to potentially promote healing at the repair site.

Emerging Evidence

Multiple biomechanical studies have evaluated ADMs in RC models.24-28 Barber and colleagues24 demonstrated that ADM had significantly higher loads to failure (229 N) than porcine skin (128 N), bovine skin (76 N), and porcine small intestine submucosa (32 N) (P < .001). In another study, Barber and colleagues25 subsequently demonstrated, in a cadaver RC tear model, an increase in mean failure strength in augmented repairs with ADM (325 N) compared to cadaveric controls (273 N) (P = .047).

A subsequent study by Barber and Aziz-Jacobo26 compared ADMs to a control model of allograft RC. The ADMs had significantly higher tensile modulus (P < .001) and higher suture retention measure by a single-pull destructive test of a simple vertical stitch (P < .05) than the RC allograft. The ultimate load to failure of the ADM model was higher than the RC allograft control (523±154 N vs 208±115 N); however, this difference did not reach statistical significance.26 Beitzel and colleagues27 evaluated ADM augmentation in a cadaver RC model and found a statistically significant increase in load to failure in ADM augmented repairs vs nonaugmented controls, (575.8 N vs 348.9 N, P = .025). Ely and colleagues28 also demonstrated that repairs augmented with ADM had a higher load to failure (643 N vs 551 N) and less gap formation (2.2 mm vs 2.8 mm) compared to controls, although this difference was not statistically significant.

 

 

These biomechanical studies have been translated to clinical findings. A level II, prospective, randomized controlled study by Barber and colleagues29 evaluated 42 patients with >3 cm, 2-tendon RCTs repaired arthroscopically.Twenty-two patients were randomized to single-row arthroscopic repair, and 20 patients to single-row arthroscopic repair augmented by ADM by an onlay technique (Figure 1) as described by Labbé.30 At average follow-up of 24 months, 85% of the augmented repairs were intact on magnetic resonance imaging (MRI) at follow-up, compared to 40% in the control group (P < .05). Agrawal31 retrospectively reviewed 14 patients with either RCTs >3 cm or recurrent RCT (may be <3 cm) that were arthroscopically repaired with a double-row technique with ADM augmentation. Postoperative MRI obtained at average of 16.8 months revealed 85.7% of repairs to be intact, with 14.3% having recurrent tears of <1 cm. Rotini and colleagues32 evaluated a smaller subset of 5 patients with large/massive primary cuff tears, arthroscopically repaired with double-row technique and ADM augmentation. Follow-up MRI at an average of 1 year demonstrated 3 intact repairs, 1 partial recurrence, and 1 complete recurrence. These clinical studies demonstrate that RCRs augmented with ADM have a much higher rate of structural integrity on postoperative imaging compared to what has been previously reported in the literature.1-6

Although an “off-label” indication, the use of ADM in massive RC tears has been described with good clinical results.14,17,19,33 The ADM is used to bridge the gap by suturing it to the edge of the retracted tendon and anchoring it to the tuberosity (Figures 2A-2E). Improvement in pain, function, and active range of motion can be achieved. Burkhead and colleagues14 obtained postoperative MRIs at average follow-up of 1.2 years and found only 3 of 11 repairs with evidence of re-tear, all noted to be smaller than preoperative tears. Gupta and colleagues17 obtained postoperative ultrasounds in 24 patients at average 3 years and showed 76% of tears to be fully intact, with the remaining 24% having only a partial tear, and 0% with full re-tears. Venouziou and colleagues19 evaluated 14 patients with minimum 18-month follow-up and Kokkalis and colleagues33 evaluated 21 patients with a 29-month follow-up; both described successful clinical outcomes but did not provide postoperative imaging evaluation. Multiple studies have adapted this technique to a fully arthroscopic method and have had similarly positive results clinically and with MRI.13,16,18,34,35 Bond and colleagues13 reported 16 cases with massive irreparable tears repaired arthroscopically with ADM to span the tendon gap. At an average follow-up of 26.8 months, 75% had good or excellent clinical results, and at an average of 1 year postoperatively 13 of 16 cases had an intact repair on gadolinium enhanced MRI.13 These studies suggest that ADM can be used for bridging massive irreparable RC tears with good clinical and radiographic outcomes.

Superior capsule reconstruction is a biomechanically proven concept that has been described in previous studies.36,37 In the original technique, autologous tensor fascia lata (TFL) is anchored from the glenoid margin to the greater tuberosity footprint to restore the superior stability of the glenohumeral joint, without altering the native glenohumeral contact forces.38 This concept has gained popularity in the United States, but with the use of an ADM instead of harvesting TFL (Figures 3A, 3B). However, there are no published biomechanical or clinical studies with the use of ADM in superior capsular reconstruction.

Conclusion

The use of ADM is an emerging solution for augmenting primary RCRs and the treatment of irreparable RC tears. The biomechanical and clinical studies summarized support the use of ADM in RC surgery. Further randomized studies are needed to add to the growing evidence on the use of ADMs.

References

1.    Green A. Chronic massive rotator cuff tears: evaluation and management. J Am Acad Orthop Surg. 2003;11(5):321-331.

2.    Boileau P, Brassart N, Watkinson DJ, Carles M, Hatzidakis AM, Krishnan SG. Arthroscopic repair of full-thickness tears of the supraspinatus: does the tendon really heal? J Bone Joint Surg Am. 2005;87(6):1229-1240.

3.    Iannotti JP, Deutsch A, Green A, et al. Time to failure after rotator cuff repair: a prospective imaging study. J Bone Joint Surg Am. 2013;95(11):965-971.

4.    Karas EH, Iannotti JP. Failed repair of the rotator cuff: evaluation and treatment of complications. Instr Course Lect. 1998;47:87-95.

5.    Burkhart SS. Biomechanics of rotator cuff repair: converting the ritual to a science. Instr Course Lect. 1998;47:43-50.

6.    Derwin KA, Badylak SF, Steinmann SP, Iannotti JP. Extracellular matrix scaffold devices for rotator cuff repair. J Shoulder Elbow Surg. 2010;19:467-476.

7.    Neviaser JS, Neviaser RJ, Neviaser TJ. The repair of chronic massive ruptures of the rotator cuff of the shoulder by use of a freeze-dried rotator cuff. J Bone Joint Surg Am. 1978;60(5):681-684.

8.    Ito J, Morioka T. Surgical treatment for large and massive tears of the rotator cuff. Int Orthop. 2003;27(4):228-231.

9.    Nasca RJ. The use of freeze-dried allografts in the management of global rotator cuff tears. Clin Orthop Related Res. 1988;228:218-226.

10.  Moore DR, Cain EL, Schwartz ML, Clancy WG Jr. Allograft reconstruction for massive, irreparable rotator cuff tears. Am J Sports Med. 2006;34(3):392-396.

11.  Walton JR, Bowman NK, Khatib Y, Linklater J, Murrell GA. Restore orthobiologic implant: not recommended for augmentation of rotator cuff repairs. J Bone Joint Surg Am. 2007;89(4):786-791.

12.  Iannotti JP, Codsi MJ, Kwon YW, Derwin K, Ciccone J, Brems JJ. Porcine small intestine submucosa augmentation of surgical repair of chronic two-tendon rotator cuff tears. A randomized, controlled trial. J Bone Joint Surg Am. 2006;88(6):1238-1244.

13.  Bond JL, Dopirak RM, Higgins J, Burns J, Snyder SJ. Arthroscopic replacement of massive, irreparable rotator cuff tears using a GraftJacket allograft: technique and preliminary results. Arthroscopy. 2008;24(4):403-409.

14.  Burkhead WZ Jr, Schiffern SC, Krishnan SG. Use of Graft Jacket as an augmentation for massive rotator cuff tears. Semin Arthoplasty. 2007;18(1):11-18.

15.  Dehler T, Pennings AL, ElMaraghy AW. Dermal allograft reconstruction of a chronic pectoralis major tear. J Shoulder Elbow Surg. 2013;22(10):e18-e22.

16.  Dopirak R, Bond JL, Snyder SJ. Arthroscopic total rotator cuff replacement with an acellular dermal allograft matrix. Int J Shoulder Surg. 2007;1(1):7-15.

17.  Gupta AK, Hug K, Berkoff DJ, et al. Dermal tissue allograft for the repair of massive irreparable rotator cuff tears. Am J Sports Med. 2012;40(1):141-147.

18.  Modi A, Singh HP, Pandey R, Armstrong A. Management of irreparable rotator cuff tears with the GraftJacket allograft as an interpositional graft. Shoulder Elbow. 2013;5(3):188-194.

19.  Venouziou AI, Kokkalis ZT, Sotereanos DG. Human dermal allograft interposition for the reconstruction of massive irreparable rotator cuff tears. Am J Orthop. 2013;42(2):63-70.

20.  Acevedo DC, Shore B, Mirzayan R. Orthopedic applications of acellular human dermal allograft for shoulder and elbow surgery. Orthop Clin North Am. 2015;46(3):377-388.

21.  Beniker D, McQuillan D, Livesey S, et al. The use of acellular dermal matrix as a scaffold for periosteum replacement. Orthopedics. 2003;26(5 Suppl):s591-s596.

22.  Smith RD, Carr A, Dakin SG, Snelling SJ, Yapp C, Hakimi O. The response of tenocytes to commercial scaffolds used for rotator cuff repair. Eur Cell Mater. 2016;31:107-118.

23.   Adams JE, Zobitz ME, Reach JS Jr, An KN, Steinmann SP. Rotator cuff repair using an acellular dermal matrix graft: an in vivo study in a canine model. Arthroscopy. 2006;22(7):700-709.

24.  Barber FA, Herbert MA, Coons DA. Tendon augmentation grafts: biomechanical failure loads and failure patterns. Arthroscopy. 2006;22(5):534-538.

25.  Barber FA, Herbert MA, Boothby MH. Ultimate tensile failure loads of a human dermal allograft rotator cuff augmentation. Arthroscopy. 2008;24(1):20-24.

26.  Barber AF, Aziz-Jacobo J. Biomechanical testing of commercially available soft-tissue augmentation materials. Arthroscopy. 2009;25(11):1233-1239.

27.  Beitzel K, Chowaniec DM, McCarthy MB, et al. Stability of double-row rotator cuff repair is not adversely affected by scaffold interposition between tendon and bone. Am J Sports Med. 2012;40(5):1148-1154.

28.  Ely EE, Figueroa NM, Gilot GJ. Biomechanical analysis of rotator cuff repairs with extraccellular matrix graft augmentation. Orthopedics. 2014;37(9):608-614.

29.  Barber AF, Burns JP, Deutsch A, Labbé MR, Litchfield RB. A prospective, randomized evaluation of acellular human dermal matrix augmentation for arthroscopic rotator cuff repair. Arthroscopy. 2012;28(1):8-15.

30.  Labbé MR. Arthroscopic technique for patch augmentation of rotator cuff repairs. Arthroscopy. 2006;22(1):1136.e1-e6.

31.  Agrawal V. Healing rates for challenging rotator cuff tears utilizing an acellular human dermal reinforcement graft. Int J Shoulder Surg. 2012;6(2):36-44.

32.  Rotini R, Marinelli A, Guerra E, et al. Human dermal matrix scaffold augmentation for large and massive rotator cuff repairs: preliminary clinical and MRI results at 1-year follow-up. Musculoskelet Surg. 2011;95 Suppl 1:S13-S23.

33.  Kokkalis ZT, Mavrogenis AF, Scarlat M, et al. Human dermal allograft for massive rotator cuff tears. Orthopedics. 2014;37(12):e1108-e1116.

34.  Wong I, Burns J, Snyder S. Arthroscopic GraftJacket repair of rotator cuff tears. J Shoulder Elbow Surg. 2010;19(2 Suppl):104-109.

35.  Snyder SJ, Bond JL. Technique for arthroscopic replacement of severely damaged rotator cuff using “GraftJacket” allograft. Oper Tech Sports Med. 2007;15(2):86-94.

36.  Mihata T, McGarry MH, Pirolo JM, Kinoshita M, Lee TQ. Superior capsule reconstruction to restore superior stability in irreparable rotator cuff tears: a biomechanical cadaveric study. Am J Sports Med. 2012;40(10):2248-2255.

37.  Mihata T, McGarry MH, Kahn T, Goldberg I, Neo M, Lee TQ. Biomechanical role of capsular continuity in superior capsule reconstruction for irreparable tears of the supraspinatus tendon. Am J Sports Med. 2016;44(6):1423-1430.

38.  Mihata T, Lee TQ, Watanabe C, et al. Clinical results of arthroscopic superior capsule reconstruction for irreparable rotator cuff tears. Arthroscopy. 2013;29(3):459-470.

Article PDF
Author and Disclosure Information

Joseph Cooper, MD, and Raffy Mirzayan, MD

Authors’ Disclosure Statement: Dr. Mirzayan reports that he receives royalties from Thieme and Lippincott; receives research grants from Arthrex, Joint Restoration Foundation, and BioD, LLC and educational grants from Arthrex; and has stock in Alignmed, Cayenne Medical, and USIS/ITS Implants. Dr. Cooper reports no actual or potential conflict of interest in relation to this article.

Issue
The American Journal of Orthopedics - 45(5)
Publications
Topics
Page Number
301-305
Legacy Keywords
acellular dermal matrix, ADM, rotator cuff surgery, rotator cuff, RCR, repair, tendon, review, shoulder, cooper, mirzayan
Sections
Author and Disclosure Information

Joseph Cooper, MD, and Raffy Mirzayan, MD

Authors’ Disclosure Statement: Dr. Mirzayan reports that he receives royalties from Thieme and Lippincott; receives research grants from Arthrex, Joint Restoration Foundation, and BioD, LLC and educational grants from Arthrex; and has stock in Alignmed, Cayenne Medical, and USIS/ITS Implants. Dr. Cooper reports no actual or potential conflict of interest in relation to this article.

Author and Disclosure Information

Joseph Cooper, MD, and Raffy Mirzayan, MD

Authors’ Disclosure Statement: Dr. Mirzayan reports that he receives royalties from Thieme and Lippincott; receives research grants from Arthrex, Joint Restoration Foundation, and BioD, LLC and educational grants from Arthrex; and has stock in Alignmed, Cayenne Medical, and USIS/ITS Implants. Dr. Cooper reports no actual or potential conflict of interest in relation to this article.

Article PDF
Article PDF

Rotator cuff repairs (RCRs) can be challenging due to poor tendon quality and the inability of tendon to heal to bone. Smoking, age over 63 years, fatty infiltration, and massive cuff tears are all factors implicated in increased failure rates.1-3 Tears >3 cm have a structural failure rate ranging from 11% to 95% in the literature.1-5 Massive tears (tears >5 cm or involving 2 or more tendons) are even more complex and have failure rates of 20% to 90%.5,6 The weakest link in the RCR construct is the suture-tendon interface, and suture pullout through the tendon is thought to be the most common method of failure.6 The purpose of this review is to examine whether literature supports the use of acellular dermal matrices (ADMs) in rotator cuff surgery.

The high rate of structural failures after RCR has led surgeons to seek means to augment repairs and new means of reconstruction for irreparable tears. Freeze dried allograft tendons have been used historically with mixed results, including reports of complete graft failures and foreign body reaction.7-10 Porcine intestinal submucosal membrane “patches” gained popularity due to off-the- shelf availability of the graft. However, these were found to have poor outcomes with early graft rejection and intense inflammatory reaction.11,12 Recently, ADMs have gained significant interest due to favorable biomechanical properties and clinical outcomes.13-19

An ADM is an allograft composed of mostly type I collagen that is processed to remove donor cells while preserving the extracellular matrix. There are several commercially available ADMs with different methods of processing and sterilization, as well as handling characteristics.20,21 In vivo studies have demonstrated that removing the cellular components allows infiltration of native cellular agents, such as fibroblasts, vascular tissue, and tenocytes, while causing minimal host inflammatory reaction.21-23 In addition, superior suture pullout strength has been demonstrated by multiple benchtop and preclinical studies.23,24 Therefore, ADMs play a dual role of strengthening the repair while allowing infiltration of host cells and growth factors to potentially promote healing at the repair site.

Emerging Evidence

Multiple biomechanical studies have evaluated ADMs in RC models.24-28 Barber and colleagues24 demonstrated that ADM had significantly higher loads to failure (229 N) than porcine skin (128 N), bovine skin (76 N), and porcine small intestine submucosa (32 N) (P < .001). In another study, Barber and colleagues25 subsequently demonstrated, in a cadaver RC tear model, an increase in mean failure strength in augmented repairs with ADM (325 N) compared to cadaveric controls (273 N) (P = .047).

A subsequent study by Barber and Aziz-Jacobo26 compared ADMs to a control model of allograft RC. The ADMs had significantly higher tensile modulus (P < .001) and higher suture retention measure by a single-pull destructive test of a simple vertical stitch (P < .05) than the RC allograft. The ultimate load to failure of the ADM model was higher than the RC allograft control (523±154 N vs 208±115 N); however, this difference did not reach statistical significance.26 Beitzel and colleagues27 evaluated ADM augmentation in a cadaver RC model and found a statistically significant increase in load to failure in ADM augmented repairs vs nonaugmented controls, (575.8 N vs 348.9 N, P = .025). Ely and colleagues28 also demonstrated that repairs augmented with ADM had a higher load to failure (643 N vs 551 N) and less gap formation (2.2 mm vs 2.8 mm) compared to controls, although this difference was not statistically significant.

 

 

These biomechanical studies have been translated to clinical findings. A level II, prospective, randomized controlled study by Barber and colleagues29 evaluated 42 patients with >3 cm, 2-tendon RCTs repaired arthroscopically.Twenty-two patients were randomized to single-row arthroscopic repair, and 20 patients to single-row arthroscopic repair augmented by ADM by an onlay technique (Figure 1) as described by Labbé.30 At average follow-up of 24 months, 85% of the augmented repairs were intact on magnetic resonance imaging (MRI) at follow-up, compared to 40% in the control group (P < .05). Agrawal31 retrospectively reviewed 14 patients with either RCTs >3 cm or recurrent RCT (may be <3 cm) that were arthroscopically repaired with a double-row technique with ADM augmentation. Postoperative MRI obtained at average of 16.8 months revealed 85.7% of repairs to be intact, with 14.3% having recurrent tears of <1 cm. Rotini and colleagues32 evaluated a smaller subset of 5 patients with large/massive primary cuff tears, arthroscopically repaired with double-row technique and ADM augmentation. Follow-up MRI at an average of 1 year demonstrated 3 intact repairs, 1 partial recurrence, and 1 complete recurrence. These clinical studies demonstrate that RCRs augmented with ADM have a much higher rate of structural integrity on postoperative imaging compared to what has been previously reported in the literature.1-6

Although an “off-label” indication, the use of ADM in massive RC tears has been described with good clinical results.14,17,19,33 The ADM is used to bridge the gap by suturing it to the edge of the retracted tendon and anchoring it to the tuberosity (Figures 2A-2E). Improvement in pain, function, and active range of motion can be achieved. Burkhead and colleagues14 obtained postoperative MRIs at average follow-up of 1.2 years and found only 3 of 11 repairs with evidence of re-tear, all noted to be smaller than preoperative tears. Gupta and colleagues17 obtained postoperative ultrasounds in 24 patients at average 3 years and showed 76% of tears to be fully intact, with the remaining 24% having only a partial tear, and 0% with full re-tears. Venouziou and colleagues19 evaluated 14 patients with minimum 18-month follow-up and Kokkalis and colleagues33 evaluated 21 patients with a 29-month follow-up; both described successful clinical outcomes but did not provide postoperative imaging evaluation. Multiple studies have adapted this technique to a fully arthroscopic method and have had similarly positive results clinically and with MRI.13,16,18,34,35 Bond and colleagues13 reported 16 cases with massive irreparable tears repaired arthroscopically with ADM to span the tendon gap. At an average follow-up of 26.8 months, 75% had good or excellent clinical results, and at an average of 1 year postoperatively 13 of 16 cases had an intact repair on gadolinium enhanced MRI.13 These studies suggest that ADM can be used for bridging massive irreparable RC tears with good clinical and radiographic outcomes.

Superior capsule reconstruction is a biomechanically proven concept that has been described in previous studies.36,37 In the original technique, autologous tensor fascia lata (TFL) is anchored from the glenoid margin to the greater tuberosity footprint to restore the superior stability of the glenohumeral joint, without altering the native glenohumeral contact forces.38 This concept has gained popularity in the United States, but with the use of an ADM instead of harvesting TFL (Figures 3A, 3B). However, there are no published biomechanical or clinical studies with the use of ADM in superior capsular reconstruction.

Conclusion

The use of ADM is an emerging solution for augmenting primary RCRs and the treatment of irreparable RC tears. The biomechanical and clinical studies summarized support the use of ADM in RC surgery. Further randomized studies are needed to add to the growing evidence on the use of ADMs.

Rotator cuff repairs (RCRs) can be challenging due to poor tendon quality and the inability of tendon to heal to bone. Smoking, age over 63 years, fatty infiltration, and massive cuff tears are all factors implicated in increased failure rates.1-3 Tears >3 cm have a structural failure rate ranging from 11% to 95% in the literature.1-5 Massive tears (tears >5 cm or involving 2 or more tendons) are even more complex and have failure rates of 20% to 90%.5,6 The weakest link in the RCR construct is the suture-tendon interface, and suture pullout through the tendon is thought to be the most common method of failure.6 The purpose of this review is to examine whether literature supports the use of acellular dermal matrices (ADMs) in rotator cuff surgery.

The high rate of structural failures after RCR has led surgeons to seek means to augment repairs and new means of reconstruction for irreparable tears. Freeze dried allograft tendons have been used historically with mixed results, including reports of complete graft failures and foreign body reaction.7-10 Porcine intestinal submucosal membrane “patches” gained popularity due to off-the- shelf availability of the graft. However, these were found to have poor outcomes with early graft rejection and intense inflammatory reaction.11,12 Recently, ADMs have gained significant interest due to favorable biomechanical properties and clinical outcomes.13-19

An ADM is an allograft composed of mostly type I collagen that is processed to remove donor cells while preserving the extracellular matrix. There are several commercially available ADMs with different methods of processing and sterilization, as well as handling characteristics.20,21 In vivo studies have demonstrated that removing the cellular components allows infiltration of native cellular agents, such as fibroblasts, vascular tissue, and tenocytes, while causing minimal host inflammatory reaction.21-23 In addition, superior suture pullout strength has been demonstrated by multiple benchtop and preclinical studies.23,24 Therefore, ADMs play a dual role of strengthening the repair while allowing infiltration of host cells and growth factors to potentially promote healing at the repair site.

Emerging Evidence

Multiple biomechanical studies have evaluated ADMs in RC models.24-28 Barber and colleagues24 demonstrated that ADM had significantly higher loads to failure (229 N) than porcine skin (128 N), bovine skin (76 N), and porcine small intestine submucosa (32 N) (P < .001). In another study, Barber and colleagues25 subsequently demonstrated, in a cadaver RC tear model, an increase in mean failure strength in augmented repairs with ADM (325 N) compared to cadaveric controls (273 N) (P = .047).

A subsequent study by Barber and Aziz-Jacobo26 compared ADMs to a control model of allograft RC. The ADMs had significantly higher tensile modulus (P < .001) and higher suture retention measure by a single-pull destructive test of a simple vertical stitch (P < .05) than the RC allograft. The ultimate load to failure of the ADM model was higher than the RC allograft control (523±154 N vs 208±115 N); however, this difference did not reach statistical significance.26 Beitzel and colleagues27 evaluated ADM augmentation in a cadaver RC model and found a statistically significant increase in load to failure in ADM augmented repairs vs nonaugmented controls, (575.8 N vs 348.9 N, P = .025). Ely and colleagues28 also demonstrated that repairs augmented with ADM had a higher load to failure (643 N vs 551 N) and less gap formation (2.2 mm vs 2.8 mm) compared to controls, although this difference was not statistically significant.

 

 

These biomechanical studies have been translated to clinical findings. A level II, prospective, randomized controlled study by Barber and colleagues29 evaluated 42 patients with >3 cm, 2-tendon RCTs repaired arthroscopically.Twenty-two patients were randomized to single-row arthroscopic repair, and 20 patients to single-row arthroscopic repair augmented by ADM by an onlay technique (Figure 1) as described by Labbé.30 At average follow-up of 24 months, 85% of the augmented repairs were intact on magnetic resonance imaging (MRI) at follow-up, compared to 40% in the control group (P < .05). Agrawal31 retrospectively reviewed 14 patients with either RCTs >3 cm or recurrent RCT (may be <3 cm) that were arthroscopically repaired with a double-row technique with ADM augmentation. Postoperative MRI obtained at average of 16.8 months revealed 85.7% of repairs to be intact, with 14.3% having recurrent tears of <1 cm. Rotini and colleagues32 evaluated a smaller subset of 5 patients with large/massive primary cuff tears, arthroscopically repaired with double-row technique and ADM augmentation. Follow-up MRI at an average of 1 year demonstrated 3 intact repairs, 1 partial recurrence, and 1 complete recurrence. These clinical studies demonstrate that RCRs augmented with ADM have a much higher rate of structural integrity on postoperative imaging compared to what has been previously reported in the literature.1-6

Although an “off-label” indication, the use of ADM in massive RC tears has been described with good clinical results.14,17,19,33 The ADM is used to bridge the gap by suturing it to the edge of the retracted tendon and anchoring it to the tuberosity (Figures 2A-2E). Improvement in pain, function, and active range of motion can be achieved. Burkhead and colleagues14 obtained postoperative MRIs at average follow-up of 1.2 years and found only 3 of 11 repairs with evidence of re-tear, all noted to be smaller than preoperative tears. Gupta and colleagues17 obtained postoperative ultrasounds in 24 patients at average 3 years and showed 76% of tears to be fully intact, with the remaining 24% having only a partial tear, and 0% with full re-tears. Venouziou and colleagues19 evaluated 14 patients with minimum 18-month follow-up and Kokkalis and colleagues33 evaluated 21 patients with a 29-month follow-up; both described successful clinical outcomes but did not provide postoperative imaging evaluation. Multiple studies have adapted this technique to a fully arthroscopic method and have had similarly positive results clinically and with MRI.13,16,18,34,35 Bond and colleagues13 reported 16 cases with massive irreparable tears repaired arthroscopically with ADM to span the tendon gap. At an average follow-up of 26.8 months, 75% had good or excellent clinical results, and at an average of 1 year postoperatively 13 of 16 cases had an intact repair on gadolinium enhanced MRI.13 These studies suggest that ADM can be used for bridging massive irreparable RC tears with good clinical and radiographic outcomes.

Superior capsule reconstruction is a biomechanically proven concept that has been described in previous studies.36,37 In the original technique, autologous tensor fascia lata (TFL) is anchored from the glenoid margin to the greater tuberosity footprint to restore the superior stability of the glenohumeral joint, without altering the native glenohumeral contact forces.38 This concept has gained popularity in the United States, but with the use of an ADM instead of harvesting TFL (Figures 3A, 3B). However, there are no published biomechanical or clinical studies with the use of ADM in superior capsular reconstruction.

Conclusion

The use of ADM is an emerging solution for augmenting primary RCRs and the treatment of irreparable RC tears. The biomechanical and clinical studies summarized support the use of ADM in RC surgery. Further randomized studies are needed to add to the growing evidence on the use of ADMs.

References

1.    Green A. Chronic massive rotator cuff tears: evaluation and management. J Am Acad Orthop Surg. 2003;11(5):321-331.

2.    Boileau P, Brassart N, Watkinson DJ, Carles M, Hatzidakis AM, Krishnan SG. Arthroscopic repair of full-thickness tears of the supraspinatus: does the tendon really heal? J Bone Joint Surg Am. 2005;87(6):1229-1240.

3.    Iannotti JP, Deutsch A, Green A, et al. Time to failure after rotator cuff repair: a prospective imaging study. J Bone Joint Surg Am. 2013;95(11):965-971.

4.    Karas EH, Iannotti JP. Failed repair of the rotator cuff: evaluation and treatment of complications. Instr Course Lect. 1998;47:87-95.

5.    Burkhart SS. Biomechanics of rotator cuff repair: converting the ritual to a science. Instr Course Lect. 1998;47:43-50.

6.    Derwin KA, Badylak SF, Steinmann SP, Iannotti JP. Extracellular matrix scaffold devices for rotator cuff repair. J Shoulder Elbow Surg. 2010;19:467-476.

7.    Neviaser JS, Neviaser RJ, Neviaser TJ. The repair of chronic massive ruptures of the rotator cuff of the shoulder by use of a freeze-dried rotator cuff. J Bone Joint Surg Am. 1978;60(5):681-684.

8.    Ito J, Morioka T. Surgical treatment for large and massive tears of the rotator cuff. Int Orthop. 2003;27(4):228-231.

9.    Nasca RJ. The use of freeze-dried allografts in the management of global rotator cuff tears. Clin Orthop Related Res. 1988;228:218-226.

10.  Moore DR, Cain EL, Schwartz ML, Clancy WG Jr. Allograft reconstruction for massive, irreparable rotator cuff tears. Am J Sports Med. 2006;34(3):392-396.

11.  Walton JR, Bowman NK, Khatib Y, Linklater J, Murrell GA. Restore orthobiologic implant: not recommended for augmentation of rotator cuff repairs. J Bone Joint Surg Am. 2007;89(4):786-791.

12.  Iannotti JP, Codsi MJ, Kwon YW, Derwin K, Ciccone J, Brems JJ. Porcine small intestine submucosa augmentation of surgical repair of chronic two-tendon rotator cuff tears. A randomized, controlled trial. J Bone Joint Surg Am. 2006;88(6):1238-1244.

13.  Bond JL, Dopirak RM, Higgins J, Burns J, Snyder SJ. Arthroscopic replacement of massive, irreparable rotator cuff tears using a GraftJacket allograft: technique and preliminary results. Arthroscopy. 2008;24(4):403-409.

14.  Burkhead WZ Jr, Schiffern SC, Krishnan SG. Use of Graft Jacket as an augmentation for massive rotator cuff tears. Semin Arthoplasty. 2007;18(1):11-18.

15.  Dehler T, Pennings AL, ElMaraghy AW. Dermal allograft reconstruction of a chronic pectoralis major tear. J Shoulder Elbow Surg. 2013;22(10):e18-e22.

16.  Dopirak R, Bond JL, Snyder SJ. Arthroscopic total rotator cuff replacement with an acellular dermal allograft matrix. Int J Shoulder Surg. 2007;1(1):7-15.

17.  Gupta AK, Hug K, Berkoff DJ, et al. Dermal tissue allograft for the repair of massive irreparable rotator cuff tears. Am J Sports Med. 2012;40(1):141-147.

18.  Modi A, Singh HP, Pandey R, Armstrong A. Management of irreparable rotator cuff tears with the GraftJacket allograft as an interpositional graft. Shoulder Elbow. 2013;5(3):188-194.

19.  Venouziou AI, Kokkalis ZT, Sotereanos DG. Human dermal allograft interposition for the reconstruction of massive irreparable rotator cuff tears. Am J Orthop. 2013;42(2):63-70.

20.  Acevedo DC, Shore B, Mirzayan R. Orthopedic applications of acellular human dermal allograft for shoulder and elbow surgery. Orthop Clin North Am. 2015;46(3):377-388.

21.  Beniker D, McQuillan D, Livesey S, et al. The use of acellular dermal matrix as a scaffold for periosteum replacement. Orthopedics. 2003;26(5 Suppl):s591-s596.

22.  Smith RD, Carr A, Dakin SG, Snelling SJ, Yapp C, Hakimi O. The response of tenocytes to commercial scaffolds used for rotator cuff repair. Eur Cell Mater. 2016;31:107-118.

23.   Adams JE, Zobitz ME, Reach JS Jr, An KN, Steinmann SP. Rotator cuff repair using an acellular dermal matrix graft: an in vivo study in a canine model. Arthroscopy. 2006;22(7):700-709.

24.  Barber FA, Herbert MA, Coons DA. Tendon augmentation grafts: biomechanical failure loads and failure patterns. Arthroscopy. 2006;22(5):534-538.

25.  Barber FA, Herbert MA, Boothby MH. Ultimate tensile failure loads of a human dermal allograft rotator cuff augmentation. Arthroscopy. 2008;24(1):20-24.

26.  Barber AF, Aziz-Jacobo J. Biomechanical testing of commercially available soft-tissue augmentation materials. Arthroscopy. 2009;25(11):1233-1239.

27.  Beitzel K, Chowaniec DM, McCarthy MB, et al. Stability of double-row rotator cuff repair is not adversely affected by scaffold interposition between tendon and bone. Am J Sports Med. 2012;40(5):1148-1154.

28.  Ely EE, Figueroa NM, Gilot GJ. Biomechanical analysis of rotator cuff repairs with extraccellular matrix graft augmentation. Orthopedics. 2014;37(9):608-614.

29.  Barber AF, Burns JP, Deutsch A, Labbé MR, Litchfield RB. A prospective, randomized evaluation of acellular human dermal matrix augmentation for arthroscopic rotator cuff repair. Arthroscopy. 2012;28(1):8-15.

30.  Labbé MR. Arthroscopic technique for patch augmentation of rotator cuff repairs. Arthroscopy. 2006;22(1):1136.e1-e6.

31.  Agrawal V. Healing rates for challenging rotator cuff tears utilizing an acellular human dermal reinforcement graft. Int J Shoulder Surg. 2012;6(2):36-44.

32.  Rotini R, Marinelli A, Guerra E, et al. Human dermal matrix scaffold augmentation for large and massive rotator cuff repairs: preliminary clinical and MRI results at 1-year follow-up. Musculoskelet Surg. 2011;95 Suppl 1:S13-S23.

33.  Kokkalis ZT, Mavrogenis AF, Scarlat M, et al. Human dermal allograft for massive rotator cuff tears. Orthopedics. 2014;37(12):e1108-e1116.

34.  Wong I, Burns J, Snyder S. Arthroscopic GraftJacket repair of rotator cuff tears. J Shoulder Elbow Surg. 2010;19(2 Suppl):104-109.

35.  Snyder SJ, Bond JL. Technique for arthroscopic replacement of severely damaged rotator cuff using “GraftJacket” allograft. Oper Tech Sports Med. 2007;15(2):86-94.

36.  Mihata T, McGarry MH, Pirolo JM, Kinoshita M, Lee TQ. Superior capsule reconstruction to restore superior stability in irreparable rotator cuff tears: a biomechanical cadaveric study. Am J Sports Med. 2012;40(10):2248-2255.

37.  Mihata T, McGarry MH, Kahn T, Goldberg I, Neo M, Lee TQ. Biomechanical role of capsular continuity in superior capsule reconstruction for irreparable tears of the supraspinatus tendon. Am J Sports Med. 2016;44(6):1423-1430.

38.  Mihata T, Lee TQ, Watanabe C, et al. Clinical results of arthroscopic superior capsule reconstruction for irreparable rotator cuff tears. Arthroscopy. 2013;29(3):459-470.

References

1.    Green A. Chronic massive rotator cuff tears: evaluation and management. J Am Acad Orthop Surg. 2003;11(5):321-331.

2.    Boileau P, Brassart N, Watkinson DJ, Carles M, Hatzidakis AM, Krishnan SG. Arthroscopic repair of full-thickness tears of the supraspinatus: does the tendon really heal? J Bone Joint Surg Am. 2005;87(6):1229-1240.

3.    Iannotti JP, Deutsch A, Green A, et al. Time to failure after rotator cuff repair: a prospective imaging study. J Bone Joint Surg Am. 2013;95(11):965-971.

4.    Karas EH, Iannotti JP. Failed repair of the rotator cuff: evaluation and treatment of complications. Instr Course Lect. 1998;47:87-95.

5.    Burkhart SS. Biomechanics of rotator cuff repair: converting the ritual to a science. Instr Course Lect. 1998;47:43-50.

6.    Derwin KA, Badylak SF, Steinmann SP, Iannotti JP. Extracellular matrix scaffold devices for rotator cuff repair. J Shoulder Elbow Surg. 2010;19:467-476.

7.    Neviaser JS, Neviaser RJ, Neviaser TJ. The repair of chronic massive ruptures of the rotator cuff of the shoulder by use of a freeze-dried rotator cuff. J Bone Joint Surg Am. 1978;60(5):681-684.

8.    Ito J, Morioka T. Surgical treatment for large and massive tears of the rotator cuff. Int Orthop. 2003;27(4):228-231.

9.    Nasca RJ. The use of freeze-dried allografts in the management of global rotator cuff tears. Clin Orthop Related Res. 1988;228:218-226.

10.  Moore DR, Cain EL, Schwartz ML, Clancy WG Jr. Allograft reconstruction for massive, irreparable rotator cuff tears. Am J Sports Med. 2006;34(3):392-396.

11.  Walton JR, Bowman NK, Khatib Y, Linklater J, Murrell GA. Restore orthobiologic implant: not recommended for augmentation of rotator cuff repairs. J Bone Joint Surg Am. 2007;89(4):786-791.

12.  Iannotti JP, Codsi MJ, Kwon YW, Derwin K, Ciccone J, Brems JJ. Porcine small intestine submucosa augmentation of surgical repair of chronic two-tendon rotator cuff tears. A randomized, controlled trial. J Bone Joint Surg Am. 2006;88(6):1238-1244.

13.  Bond JL, Dopirak RM, Higgins J, Burns J, Snyder SJ. Arthroscopic replacement of massive, irreparable rotator cuff tears using a GraftJacket allograft: technique and preliminary results. Arthroscopy. 2008;24(4):403-409.

14.  Burkhead WZ Jr, Schiffern SC, Krishnan SG. Use of Graft Jacket as an augmentation for massive rotator cuff tears. Semin Arthoplasty. 2007;18(1):11-18.

15.  Dehler T, Pennings AL, ElMaraghy AW. Dermal allograft reconstruction of a chronic pectoralis major tear. J Shoulder Elbow Surg. 2013;22(10):e18-e22.

16.  Dopirak R, Bond JL, Snyder SJ. Arthroscopic total rotator cuff replacement with an acellular dermal allograft matrix. Int J Shoulder Surg. 2007;1(1):7-15.

17.  Gupta AK, Hug K, Berkoff DJ, et al. Dermal tissue allograft for the repair of massive irreparable rotator cuff tears. Am J Sports Med. 2012;40(1):141-147.

18.  Modi A, Singh HP, Pandey R, Armstrong A. Management of irreparable rotator cuff tears with the GraftJacket allograft as an interpositional graft. Shoulder Elbow. 2013;5(3):188-194.

19.  Venouziou AI, Kokkalis ZT, Sotereanos DG. Human dermal allograft interposition for the reconstruction of massive irreparable rotator cuff tears. Am J Orthop. 2013;42(2):63-70.

20.  Acevedo DC, Shore B, Mirzayan R. Orthopedic applications of acellular human dermal allograft for shoulder and elbow surgery. Orthop Clin North Am. 2015;46(3):377-388.

21.  Beniker D, McQuillan D, Livesey S, et al. The use of acellular dermal matrix as a scaffold for periosteum replacement. Orthopedics. 2003;26(5 Suppl):s591-s596.

22.  Smith RD, Carr A, Dakin SG, Snelling SJ, Yapp C, Hakimi O. The response of tenocytes to commercial scaffolds used for rotator cuff repair. Eur Cell Mater. 2016;31:107-118.

23.   Adams JE, Zobitz ME, Reach JS Jr, An KN, Steinmann SP. Rotator cuff repair using an acellular dermal matrix graft: an in vivo study in a canine model. Arthroscopy. 2006;22(7):700-709.

24.  Barber FA, Herbert MA, Coons DA. Tendon augmentation grafts: biomechanical failure loads and failure patterns. Arthroscopy. 2006;22(5):534-538.

25.  Barber FA, Herbert MA, Boothby MH. Ultimate tensile failure loads of a human dermal allograft rotator cuff augmentation. Arthroscopy. 2008;24(1):20-24.

26.  Barber AF, Aziz-Jacobo J. Biomechanical testing of commercially available soft-tissue augmentation materials. Arthroscopy. 2009;25(11):1233-1239.

27.  Beitzel K, Chowaniec DM, McCarthy MB, et al. Stability of double-row rotator cuff repair is not adversely affected by scaffold interposition between tendon and bone. Am J Sports Med. 2012;40(5):1148-1154.

28.  Ely EE, Figueroa NM, Gilot GJ. Biomechanical analysis of rotator cuff repairs with extraccellular matrix graft augmentation. Orthopedics. 2014;37(9):608-614.

29.  Barber AF, Burns JP, Deutsch A, Labbé MR, Litchfield RB. A prospective, randomized evaluation of acellular human dermal matrix augmentation for arthroscopic rotator cuff repair. Arthroscopy. 2012;28(1):8-15.

30.  Labbé MR. Arthroscopic technique for patch augmentation of rotator cuff repairs. Arthroscopy. 2006;22(1):1136.e1-e6.

31.  Agrawal V. Healing rates for challenging rotator cuff tears utilizing an acellular human dermal reinforcement graft. Int J Shoulder Surg. 2012;6(2):36-44.

32.  Rotini R, Marinelli A, Guerra E, et al. Human dermal matrix scaffold augmentation for large and massive rotator cuff repairs: preliminary clinical and MRI results at 1-year follow-up. Musculoskelet Surg. 2011;95 Suppl 1:S13-S23.

33.  Kokkalis ZT, Mavrogenis AF, Scarlat M, et al. Human dermal allograft for massive rotator cuff tears. Orthopedics. 2014;37(12):e1108-e1116.

34.  Wong I, Burns J, Snyder S. Arthroscopic GraftJacket repair of rotator cuff tears. J Shoulder Elbow Surg. 2010;19(2 Suppl):104-109.

35.  Snyder SJ, Bond JL. Technique for arthroscopic replacement of severely damaged rotator cuff using “GraftJacket” allograft. Oper Tech Sports Med. 2007;15(2):86-94.

36.  Mihata T, McGarry MH, Pirolo JM, Kinoshita M, Lee TQ. Superior capsule reconstruction to restore superior stability in irreparable rotator cuff tears: a biomechanical cadaveric study. Am J Sports Med. 2012;40(10):2248-2255.

37.  Mihata T, McGarry MH, Kahn T, Goldberg I, Neo M, Lee TQ. Biomechanical role of capsular continuity in superior capsule reconstruction for irreparable tears of the supraspinatus tendon. Am J Sports Med. 2016;44(6):1423-1430.

38.  Mihata T, Lee TQ, Watanabe C, et al. Clinical results of arthroscopic superior capsule reconstruction for irreparable rotator cuff tears. Arthroscopy. 2013;29(3):459-470.

Issue
The American Journal of Orthopedics - 45(5)
Issue
The American Journal of Orthopedics - 45(5)
Page Number
301-305
Page Number
301-305
Publications
Publications
Topics
Article Type
Display Headline
Acellular Dermal Matrix in Rotator Cuff Surgery
Display Headline
Acellular Dermal Matrix in Rotator Cuff Surgery
Legacy Keywords
acellular dermal matrix, ADM, rotator cuff surgery, rotator cuff, RCR, repair, tendon, review, shoulder, cooper, mirzayan
Legacy Keywords
acellular dermal matrix, ADM, rotator cuff surgery, rotator cuff, RCR, repair, tendon, review, shoulder, cooper, mirzayan
Sections
Article Source

PURLs Copyright

Inside the Article

Article PDF Media

Platelet-Rich Plasma (PRP) in Orthopedic Sports Medicine

Article Type
Changed
Thu, 09/19/2019 - 13:25
Display Headline
Platelet-Rich Plasma (PRP) in Orthopedic Sports Medicine

Platelet-rich plasma (PRP) is a refined product of autologous blood with a platelet concentration greater than that of whole blood. It is prepared via plasmapheresis utilizing a 2-stage centrifugation process and more than 40 commercially available systems are marketed to concentrate whole blood to PRP.1 It is rich in biologic factors (growth factors, cytokines, proteins, cellular components) essential to the body’s response to injury. For this reason, it was first used in oromaxillofacial surgery in the 1950s, but its effects on the musculoskeletal system have yet to be clearly elucidated.2 However, this lack of clarity has not deterred its widespread use among orthopedic surgeons. In this review, we aim to delineate the current understanding of PRP and its proven effectiveness in the treatment of rotator cuff tears, knee osteoarthritis, ulnar collateral ligament (UCL) tears, lateral epicondylitis, hamstring injuries, and Achilles tendinopathy.

Rotator Cuff Tears

Rotator cuff tears are one of the most common etiologies for shoulder pain and disability. The incidence continues to increase with the active aging population.3 Rotator cuff tears treated with arthroscopic repair have exhibited satisfactory pain relief and functional outcomes.4-7 Despite advances in fixation techniques, the quality and speed of tendon-to-bone healing remains unpredictable, with repaired tendons exhibiting inferior mechanical properties that are susceptible to re-tear.8-10

Numerous studies have investigated PRP application during arthroscopic rotator cuff repair (RCR) in an attempt to enhance and accelerate the repair process.11-15 However, wide variability exists among protocols of how and when PRP is utilized to augment the repair. Warth and colleagues16 performed a meta-analysis of 11 Level I/II studies evaluating RCR with PRP augmentation. With regards to clinical outcome scores, they found no significant difference in pre- and postoperative American Shoulder and Elbow Surgeons (ASES), Constant, Disability of the Arm, Shoulder and Hand (DASH), or visual analog scale (VAS) pain scores between those patients with or without PRP augmentation. However, they did note a significant increase in Constant scores when PRP was delivered to the tendon-bone interface rather than over the surface of the repair site. There was no significant difference in structural outcomes (evaluated by magnetic resonance imaging [MRI] re-tear rates) between those RCRs with and without PRP augmentation, except in those tears >3 cm in anterior-posterior length using double-row technique, with the PRP group exhibiting a significantly decreased re-tear rate (25.9% vs 57.1%).16 Zhao and colleagues17 reported similar results in a meta-analysis of 8 randomized controlled trials, exhibiting no significant differences in clinical outcome scores or re-tear rates after RCR with and without PRP augmentation. Overall, most studies have failed to demonstrate a significant benefit with regards to re-tear rates or shoulder-specific outcomes with the addition of PRP during arthroscopic RCR.

Knee Osteoarthritis

Osteoarthritis is the most common musculoskeletal disorder, with an estimated prevalence of 10% of the world’s population age 60 years and older.18 The knee is commonly symptomatic, resulting in pain, disability, and significant healthcare costs. Novel biologic, nonoperative therapies, including intra-articular viscosupplementation and PRP injections, have been proposed to treat the early stages of osteoarthritis to provide symptomatic relief and delay surgical intervention.

A multitude of studies have been performed investigating the effects of PRP on knee osteoarthritis, revealing mixed results.19-22 Campbell and colleagues23 published a 2015 systematic review of 3 overlapping meta-analyses comparing the outcomes of intra-articular injection of PRP vs control (hyaluronic acid [HA] or placebo) in 3278 knees. They reported a significant improvement in patient outcome scores for the PRP group when compared to control from 2 to 12 months after injection, but due to significant differences within the included studies, the ideal number of injections or time intervals between injections remains unclear. Meheux and colleagues24 reported a 2016 systematic review including 6 studies (817 knees) comparing PRP and HA injections. They demonstrated significantly better improvements in Western Ontario and McMaster Universities Arthritis Index (WOMAC) outcome scores with PRP vs HA injections at 3 and 12 months postinjection. Similarly, Smith25 conducted a Food and Drug Administration-sanctioned, randomized, double-blind, placebo-controlled clinical trial investigating the effects of intra-articular leukocyte-poor autologous conditioned plasma (ACP) in 30 patients. He reported an improvement in the ACP treatment group WOMAC scores by 78% compared to 7% improvement in the placebo group after 12 months. Despite the heterogeneity amongst studies, the majority of published data suggests better symptomatic relief in patients with early knee degenerative changes, and use of PRP may be considered in this population.

Ulnar Collateral Ligament Injuries

The anterior band of the UCL of the elbow provides stability to valgus stress. Overhead, high-velocity throwing athletes may cause repetitive injury to the UCL, resulting in partial or complete tears of the ligament. This may result in medial elbow pain, as well as decreased throwing velocity and accuracy. Athletes with complete UCL tears have few nonoperative treatment options and generally, operative treatment with UCL reconstruction is recommended for those athletes desiring to return to sport. However, it remains unclear how to definitively treat athletes with partial UCL tears. Recently, there has been an interest in treating these injuries with PRP in conjunction with physical therapy to facilitate a more predictable outcome.

Podesta and colleagues26 published a case series of 34 athletes with MRI-diagnosed partial UCL tears who underwent ultrasound-guided UCL injections and physical therapy. At an average follow-up of 70 weeks, they reported an average return to play (RTP) of 12 weeks, with significant improvements in Kerlan-Jobe Orthopaedic Clinic (KJOC) and DASH outcome scores, and decreased dynamic ulnohumeral joint widening to valgus stress on ultrasound. Most athletes (30/34) returned to their previous level of play, and 1 patient underwent subsequent UCL reconstruction. This study demonstrates that PRP may be used in conjunction with physical therapy and an interval throwing program for the treatment of partial UCL tears, but without a comparison control group, more studies are necessary to delineate the role of PRP in this population.

Lateral Elbow Epicondylitis

Lateral elbow epicondylitis, also known as “tennis elbow,” is thought to be caused by repetitive wrist extension and is more likely to present in patients with various comorbidities such as rotator cuff pathology or a history of smoking.27-29 The condition typically presents as radiating pain centered about the lateral epicondyle. Annual incidence ranges from 0.34% to 3%, with the most recent large-scale, population-based study estimating that nearly 1 million individuals in the United States develop lateral elbow epicondylitis each year.30 For the majority of patients, symptoms resolve after 6 to 12 months of various nonoperative or minimally invasive treatments.31-33 Those who develop chronic symptoms (>12 months) may benefit from surgical intervention.34 The use of PRP has become a contentious topic of debate in treating lateral epicondylitis. Its use and efficacy have been empirically examined and compared among more traditional treatments.35-37

In a small case-series of 6 patients, contrast-enhanced ultrasound imaging was utilized to demonstrate that PRP injection therapy may induce vascularization of the myotendinous junction of the common extensor tendon up to 6 months following injection.38 These physiologic changes may precede observable clinical improvements. Brklijac and colleagues39 prospectively followed 34 patients who had refractory symptoms despite conservative treatment and elected to undergo injection with PRP. At a mean follow-up of 26 weeks, 88.2% of the patients demonstrated improvements on their Oxford Elbow Score (OES). While potentially promising, case series lack large sample sizes, longitudinal analysis, and adequate control groups for comparative analyses of treatments, thereby increasing the likelihood of unintended selection bias.

Randomized controlled trials have demonstrated no difference between PRP and corticosteroid (CS) injection treatments in the short term for symptomatic lateral elbow epicondylitis. At 15 days, 1 month, and 6 months postinjection, no significant difference was found between PRP and CS injections in dynamometer strength measurements nor patient outcome scores (VAS, DASH, OES, and Mayo Clinic Performance Index for Elbow [MMCPIE]).40,41 In fact, multiple randomized controlled trials have demonstrated PRP to be less effective at 1 and 3 months compared to CS injections, as assessed by the Patient Rated Tennis-Elbow Evaluation (PRTEE) questionnaire, VAS, MMCPIE, and Nirschl scores.42,43 One mid-term, multi-center randomized controlled trial published by Mishra and colleagues44 compared PRP injections to an active control group, demonstrating a significant improvement in VAS pain scores at 24 weeks, but no difference in the PRTEE outcome. The available evidence indicates PRP injection therapy remains limited in utility for treatment of lateral epicondylitis, particularly in the short term when compared to CS injections. In the midterm to long term, PRP therapy may provide some benefit, but ultimately, well-designed prospective randomized controlled trials are needed to delineate the effects of PRP versus the natural course of tendon healing and symptom resolution.

 

 

Hamstring Injuries

Acute hamstring injuries are common across all levels and types of sport, particularly those in which sprinting or running is involved. While there is no consensus within the literature on how RTP after hamstring injury should be managed or defined, most injuries seem to resolve around 3 to 6 weeks.45 The proximal myotendinous junction of the long head of the biceps femoris and semitendinosus are commonly associated with significant pain and edema after acute hamstring injury.46 The amount of edema resulting from grade 1 and 2 hamstring injuries has been found to correlate (minimally) with time to RTP in elite athletes.47 PRP injection near the proximal myotendinous hamstring origin has been theorized to help speed the recovery process after acute hamstring injury. To date, the literature demonstrates mixed and limited benefit of PRP injection therapy for acute hamstring injury.

Few studies have shown improvements of PRP therapy over typical nonoperative management (rest, physical therapy, nonsteroidal anti-inflammatory drugs) in acute hamstring injury, but the results must be interpreted carefully.48,49 Wetzel and colleagues48 retrospectively reviewed 17 patients with acute hamstring injury, 12 of whom failed typical management and received PRP injection at the hamstring origin. This group demonstrated significant improvements in their VAS and Nirschl scores at follow-up, whereas the 5 patients who did not receive the injection did not. However, this study exhibited significant limitations inherent to a retrospective review with a small sample size. Hamid and colleagues49 conducted a randomized controlled trial of 24 athletes with diagnosed grade 2a acute hamstring injuries, comparing autologous PRP therapy combined with a rehabilitation program versus rehabilitation program alone. RTP, changes in pain severity (Brief Pain Injury-Short Form [BPI-SF] questions 2-6), and pain interference (BPI-SF questions 9A-9G) scores over time were examined. Athletes in the PRP group exhibited no difference in outcomes scores, but returned to play sooner than controls (26.7 vs 42.5 days).

Mejia and Bradley50 have reported their experience in treating 12 National Football League (NFL) players with acute MRI grade 1 or 2 hamstring injuries with a series of PRP injections at the site of injury. They found a 1-game difference in earlier RTP when compared to the predicted RTP based on MRI grading. Similarly, Hamid and colleagues49 performed a randomized control trial published in 2014, reporting an earlier RTP (26.7 vs 42.5 days) when comparing single PRP injection vs rehabilitation alone in 28 patients diagnosed with acute ultrasound grade 2 hamstring injuries. On the contrary, a small case-control study of NFL players and a retrospective cohort study of athletes with severe hamstring injuries demonstrated no difference in RTP when PRP injected patients were compared with controls.51,52 Larger randomized controlled trials have demonstrated comparable results, including a study of 90 professional athletes in whom a single PRP injection did not decrease RTP or lessen the risk of re-injury at 2 and 6 months.53 In another large multicenter randomized controlled trial examining 80 competitive and recreational athletes, PRP did not accelerate RTP, lessen the risk of 2-month or 1-year re-injury rate, or improve secondary measures of MRI parameters, subjective patient satisfaction, or the hamstring outcome score.54 Although further study is warranted, available evidence suggests limited utility of PRP injection in the treatment of acute hamstring injuries.

Achilles Tendinopathy

Noninsertional Achilles tendinopathy is a common source of pain for both recreational and competitive athletes. Typically thought of as an overuse syndrome, Achilles tendinopathy may result in significant pain and swelling, often at the site of its tenuous blood supply, approximately 2 to 7 cm proximal to its insertion.55 Conservative management frequently begins with rest, activity/shoe modification, physical therapy, and eccentric loading exercises.56 For those whom conservative management has failed to reduce symptoms after 6 months, more invasive treatment options may be considered. Peritendinous PRP injection has become an alternative approach in treating Achilles tendinopathy refractory to conservative treatment.

In the few randomized controlled trials published, the data demonstrates no significant improvements in clinical outcomes from PRP injection for Achilles tendinopathy. Kearney and colleagues57 conducted a pilot study of 20 patients randomized into PRP injection or eccentric loading program for mid-substance Achilles tendinopathy, in which Victorian Institute of Sports Assessment (VISA-A), EuroQol 5 dimensions questionnaire (EQ-5D), and complications associated with the injection were recorded at 6 weeks, 3 months, and 6 months. Although this was a pilot study with a small sample size, no significant difference was found between groups across these time periods. Similarly, de Vos and colleagues58,59 conducted a double-blind randomized controlled trial of 54 patients with chronic mid-substance Achilles tendinopathy and randomized them into eccentric exercise therapy with either a PRP injection or a saline injected placebo groups. VISA-A scores were recorded and imaging parameters assessing tendon structure by ultrasonographic tissue characterization and color Doppler ultrasonography were taken with follow-up at 6, 12, and 24 weeks. VISA-A scores improved significantly in both groups after 24 weeks, but the difference was not statistically significant between groups. In addition, tendon structure and neovascularization (exhibited by color Doppler ultrasonography) improved in both groups, with no significant difference between groups. The current literature does not support the use of PRP in treatment of Achilles tendinopathy, as it has failed to reveal additional benefits over conventional treatment alone. Future prospective, well-designed randomized controlled trials with large sample sizes will need to be conducted to ultimately conclude whether or not PRP deserves a role in the treatment of Achilles tendinopathy.

Summary

In theory, the use of PRP within orthopedic surgery makes a great deal of sense to accelerate and augment the healing process of the aforementioned musculoskeletal injuries. However, the vast majority of published literature is Level III and IV evidence. Future research may provide the missing critical information of optimal growth factor, platelet, and leukocyte concentrations necessary for the desired effect, as well as the appropriate delivery method and timing of PRP application in different target tissues. Evidence-based guidelines to direct the use of PRP will benefit from more homogenous, repeatable, and randomized controlled trials.

References

1.    Hsu WK, Mishra A, Rodeo SR, et al. Platelet-rich plasma in orthopaedic applications: evidence-based recommendations for treatment. J Am Acad Orthop Surg. 2013;21(12):739-748.

2.    Marx RE. Platelet-rich plasma: evidence to support its use. J Oral Maxillofac Surg. 2004;62(4):489-496.

3.    Jo CH, Kim JE, Yoon KS, et al. Does platelet-rich plasma accelerate recovery after rotator cuff repair? A prospective cohort study. Am J Sports Med. 2011;39(10):2082-2090.

4.    Burkhart SS, Danaceau SM, Pearce CE Jr. Arthroscopic rotator cuff repair: Analysis of results by tear size and by repair technique-margin convergence versus direct tendon-to-bone repair. Arthroscopy. 2001;17(9):905-912.

5.    Severud EL, Ruotolo C, Abbott DD, Nottage WM. All-arthroscopic versus mini-open rotator cuff repair: A long-term retrospective outcome comparison. Arthroscopy. 2003;19(3):234-238.

6.    Huang R, Wang S, Wang Y, Qin X, Sun Y. Systematic review of all-arthroscopic versus mini-open repair of rotator cuff tears: a meta-analysis. Sci Rep. 2016;6:22857.

7.    Watson EM, Sonnabend DH. Outcome of rotator cuff repair. J Shoulder Elbow Surg. 2002;11(3):201-211.

8.    Butler DL, Juncosa N, Dressler MR. Functional efficacy of tendon repair processes. Annu Rev Biomed Eng. 2004;6:303-329.

9.    Galatz LM, Ball CM, Teefey SA, Middleton WD, Yamaguchi K. The outcome and repair integrity of completely arthroscopically repaired large and massive rotator cuff tears. J Bone Joint Surg Am. 2004;86-A(2):219-224.

10.  Lafosse L, Brozska R, Toussaint B, Gobezie R. The outcome and structural integrity of arthroscopic rotator cuff repair with use of the double-row suture anchor technique. J Bone Joint Surg Am. 2007;89(7):1533-1541.

11.  Castricini R, Longo UG, De Benedetto M, et al. Platelet-rich plasma augmentation for arthroscopic rotator cuff repair: a randomized controlled trial. Am J Sports Med. 2011;39(2):258-265.

12.  Randelli P, Arrigoni P, Ragone V, Aliprandi A, Cabitza P. Platelet rich plasma in arthroscopic rotator cuff repair: a prospective RCT study, 2-year follow-up. J Shoulder Elbow Surg. 2011;20(4):518-528.

13.  Weber SC, Kauffman JI, Parise C, Weber SJ, Katz SD. Platelet-rich fibrin matrix in the management of arthroscopic repair of the rotator cuff: a prospective, randomized, double-blinded study. Am J Sports Med. 2013;41(2):263-270.

14.  Gumina S, Campagna V, Ferrazza G, et al. Use of platelet-leukocyte membrane in arthroscopic repair of large rotator cuff tears: a prospective randomized study. J Bone Joint Surg Am. 2012;94(15):1345-1352.

15.  Rodeo SA, Delos D, Williams RJ, Adler RS, Pearle A, Warren RF.  The effect of platelet-rich fibrin matrix on rotator cuff tendon healing: a prospective, randomized clinical study. Am J Sports Med. 2012;40(6):1234-1241.

16.  Warth RJ, Dornan GJ, James EW, Horan MP, Millett PJ. Clinical and structural outcomes after arthroscopic repair of full-thickness rotator cuff tears with and without platelet-rich product supplementation: a meta-analysis and meta-regression. Arthroscopy. 2015;31(2):306-320.

17.  Zhao JG, Zhao L, Jiang YX, Wang ZL, Wang J, Zhang P. Platelet-rich plasma in arthroscopic rotator cuff repair: a meta-analysis of randomized controlled trials. Arthroscopy. 2015;31(1):125-135.

18.  Glyn-Jones S, Palmer AJ, Agricola R, et al. Osteoarthritis. Lancet. 2015;386(9991):376-387.

19.  Cerza F, Carni S, Carcangiu A, et al. Comparison between hyaluronic acid and platelet-rich plasma, intra-articular infiltration in the treatment of gonarthrosis. Am J Sports Med. 2012;40(12):2822-2827.

20.  Filardo G, Kon E, Di Martino A, et al. Platelet-rich plasma vs hyaluronic acid to treat knee degenerative pathology: study design and preliminary results of a randomized controlled trial. BMC Musculoskelet Disord. 2012;13:229.

21.  Patel S, Dhillon MS, Aggarwal S, Marwaha N, Jain A. Treatment with platelet-rich plasma is more effective than placebo for knee osteoarthritis: a prospective, double-blind, randomized trial. Am J Sports Med. 2013;41(2):356-364.

22.  Sanchez M, Fiz N, Azofra J, et al. A randomized clinical trial evaluating plasma rich in growth factors (PRGF-Endoret) versus hyaluronic acid in the short-term treatment of symptomatic knee osteoarthritis. Arthroscopy. 2012;28(8):1070-1078.

23.  Campbell KA, Saltzman BM, Mascarenhas R, et al. Does intra-articular platelet-rich plasma injection provide clinically superior outcomes compared with other therapies in the treatment of knee osteoarthritis? A systematic review of overlapping meta-analyses. Arthroscopy. 2015;31(11):2213-2221.

24.  Meheux CJ, McCulloch PC, Lintner DM, Varner KE, Harris JD. Efficacy of intra-articular platelet-rich plasma injections in knee osteoarthritis: A systematic review. Arthroscopy. 2016;32(3):495-505.

25.  Smith PA. Intra-articular autologous conditioned plasma injections provide safe and efficacious treatment for knee osteoarthritis: An FDA-sanctioned, randomized, double-blind, placebo-controlled clinical trial. Am J Sports Med. 2016;44(4):884-891.

26.   Podesta L, Crow SA, Volkmer D, Bert T, Yocum LA. Treatment of partial ulnar collateral ligament tears in the elbow with platelet-rich plasma. Am J Sports Med. 2013;41(7):1689-1694.

27.  Herquelot E, Gueguen A, Roquelaure Y, et al. Work-related risk factors for incidence of lateral epicondylitis in a large working population. Scand J Work Environ Health. 2013;39(6):578-588.

28.  Titchener AG, Fakis A, Tambe AA, Smith C, Hubbard RB, Clark DI. Risk factors in lateral epicondylitis (tennis elbow): a case-control study. J Hand Surg Eur Vol. 2013;38(2):159-164.

29.  Gruchow HW, Pelletier D. An epidemiologic study of tennis elbow. Incidence, recurrence, and effectiveness of prevention strategies. Am J Sports Med. 1979;7(4):234-238.

30.  Sanders TL Jr, Maradit Kremers H, Bryan AJ, Ransom JE, Smith J, Morrey BF.  The epidemiology and health care burden of tennis elbow: a population-based study. Am J Sports Med. 2015;43(5):1066-1071.

31.  Coonrad RW, Hooper WR. Tennis elbow: its course, natural history, conservative and surgical management. J Bone Joint Surg Am. 1973;55(6):1177-1182.

32.  Taylor SA, Hannafin JA. Evaluation and management of elbow tendinopathy. Sports Health. 2012;4(5):384-393.

33.  Sims SE, Miller K, Elfar JC, Hammert WC. Non-surgical treatment of lateral epicondylitis: a systematic review of randomized controlled trials. Hand (NY). 2014;9(4):419-446.

34.  Brummel J, Baker CL 3rd, Hopkins R, Baker CL Jr. Epicondylitis: lateral. Sports Med Arthrosc. 2014;22(3):e1-e6.

35.  de Vos RJ, Windt J, Weir A. Strong evidence against platelet-rich plasma injections for chronic lateral epicondylar tendinopathy: a systematic review. Br J Sports Med. 2014;48(12):952-956.

36.  Ahmad Z, Brooks R, Kang SN, et al. The effect of platelet-rich plasma on clinical outcomes in lateral epicondylitis. Arthroscopy. 2013;29(11):1851-1862.

37.  Arirachakaran A, Sukthuayat A, Sisayanarane T, Laoratanavoraphong S, Kanchanatawan W, Kongtharvonskul J. Platelet-rich plasma versus autologous blood versus steroid injection in lateral epicondylitis: systematic review and network meta-analysis. J Orthop Traumatol. 2016;17(2):101-112.

38.  Chaudhury S, de La Lama M, Adler RS, et al. Platelet-rich plasma for the treatment of lateral epicondylitis: sonographic assessment of tendon morphology and vascularity (pilot study). Skeletal Radiol. 2013;42(1):91-97.

39.  Brkljac M, Kumar S, Kalloo D, Hirehal K. The effect of platelet-rich plasma injection on lateral epicondylitis following failed conservative management. J Orthop. 2015;12(Suppl 2):S166-S170.

40.  Yadav R, Kothari SY, Borah D. Comparison of local injection of platelet rich plasma and corticosteroids in the treatment of lateral epicondylitis of humerus. J Clin Diagn Res. 2015;9(7):RC05-RC07.

41.  Gautam VK, Verma S, Batra S, Bhatnagar N, Arora S. Platelet-rich plasma versus corticosteroid injection for recalcitrant lateral epicondylitis: clinical and ultrasonographic evaluation. J Orthop Surg (Hong Kong). 2015;23(1):1-5.

42.  Krogh TP, Fredberg U, Stengaard-Pedersen K, Christensen R, Jensen P, Ellingsen T. Treatment of lateral epicondylitis with platelet-rich plasma, glucocorticoid, or saline: a randomized, double-blind, placebo-controlled trial. Am J Sports Med. 2013;41(3):625-635.

43.  Behera P, Dhillon M, Aggarwal S, Marwaha N, Prakash M. Leukocyte-poor platelet-rich plasma versus bupivacaine for recalcitrant lateral epicondylar tendinopathy. J Orthop Surg (Hong Kong). 2015;23(1):6-10.

44.  Mishra AK, Skrepnik NV, Edwards SG, et al. Efficacy of platelet-rich plasma for chronic tennis elbow: a double-blind, prospective, multicenter, randomized controlled trial of 230 patients. Am J Sports Med. 2014;42(2):463-471.

45.  van der Horst N, van de Hoef S, Reurink G, Huisstede B, Backx F. Return to play after hamstring injuries: a qualitative systematic review of definitions and criteria. Sports Med. 2016;46(6):899-912.

46.  Crema MD, Guermazi A, Tol JL, Niu J, Hamilton B, Roemer FW. Acute hamstring injury in football players: Association between anatomical location and extent of injury-A large single-center MRI report. J Sci Med Sport. 2016;19(4):317-322.

47.  Ekstrand J, Lee JC, Healy JC. MRI findings and return to play in football: a prospective analysis of 255 hamstring injuries in the UEFA Elite Club Injury Study. Br J Sports Med. 2016;50(12):738-743.

48.  Wetzel RJ, Patel RM, Terry MA. Platelet-rich plasma as an effective treatment for proximal hamstring injuries. Orthopedics. 2013;36(1):e64-e70.

49.  Hamid A, Mohamed Ali MR, Yusof A, George J, Lee LP. Platelet-rich plasma injections for the treatment of hamstring injuries: a randomized controlled trial. Am J Sports Med. 2014;42(10):2410-2418.

50.  Mejia HA, Bradley JP.  The effects of platelet-rich plasma on muscle: basic science and clinical application. Operative Techniques in Sports Medicine. 2011;19(3):149-153.

51.  Guillodo Y, Madouas G, Simon T, Le Dauphin H, Saraux A. Platelet-rich plasma (PRP) treatment of sports-related severe acute hamstring injuries. Muscles Ligaments Tendons J. 2015;5(4):284-288.

52.  Rettig AC, Meyer S, Bhadra AK. Platelet-rich plasma in addition to rehabilitation for acute hamstring injuries in NFL players: Clinical effects and time to return to play. Orthop J Sports Med. 2013;1(1):2325967113494354.

53.   Hamilton B, Tol JL, Almusa E, et al. Platelet-rich plasma does not enhance return to play in hamstring injuries: a randomised controlled trial. Br J Sports Med. 2015;49(14):943-950.

54.  Reurink G, Goudswaard GJ, Moen MH, et al. Rationale, secondary outcome scores and 1-year follow-up of a randomised trial of platelet-rich plasma injections in acute hamstring muscle injury: the Dutch Hamstring Injection Therapy study. Br J Sports Med. 2015;49(18):1206-1212.

55.  Kujala UM, Sarna S, Kaprio J. Cumulative incidence of achilles tendon rupture and tendinopathy in male former elite athletes. Clin J Sport Med. 2005;15(3):133-135.

56.  Alfredson H. Clinical commentary of the evolution of the treatment for chronic painful mid-portion Achilles tendinopathy. Braz J Phys Ther. 2015;19(5):429-432.

57.  Kearney RS, Parsons N, Costa ML. Achilles tendinopathy management: A pilot randomised controlled trial comparing platelet-rich plasma injection with an eccentric loading programme. Bone Joint Res. 2013;2(10):227-232.

58.  de Vos RJ, Weir A, Tol JL, Verhaar JA, Weinans H, van Schie HT. No effects of PRP on ultrasonographic tendon structure and neovascularisation in chronic midportion Achilles tendinopathy. Br J Sports Med. 2011;45(5):387-392.

59.  de Vos RJ, Weir A, van Schie HT, et al. Platelet-rich plasma injection for chronic Achilles tendinopathy: a randomized controlled trial. JAMA. 2010;303(2):144-149.

Article PDF
Author and Disclosure Information

Ryan A. Mlynarek, MD, Andrew W. Kuhn, BA, and Asheesh Bedi, MD

Authors’ Disclosure Statement: The authors report no actual or potential conflict of interest in relation to this article.

Issue
The American Journal of Orthopedics - 45(5)
Publications
Topics
Page Number
290-294, 326
Legacy Keywords
platelet-rich plasma, PRP, biologics, sports medicine, review, injections, mlynarek, kuhn, bedi
Sections
Author and Disclosure Information

Ryan A. Mlynarek, MD, Andrew W. Kuhn, BA, and Asheesh Bedi, MD

Authors’ Disclosure Statement: The authors report no actual or potential conflict of interest in relation to this article.

Author and Disclosure Information

Ryan A. Mlynarek, MD, Andrew W. Kuhn, BA, and Asheesh Bedi, MD

Authors’ Disclosure Statement: The authors report no actual or potential conflict of interest in relation to this article.

Article PDF
Article PDF

Platelet-rich plasma (PRP) is a refined product of autologous blood with a platelet concentration greater than that of whole blood. It is prepared via plasmapheresis utilizing a 2-stage centrifugation process and more than 40 commercially available systems are marketed to concentrate whole blood to PRP.1 It is rich in biologic factors (growth factors, cytokines, proteins, cellular components) essential to the body’s response to injury. For this reason, it was first used in oromaxillofacial surgery in the 1950s, but its effects on the musculoskeletal system have yet to be clearly elucidated.2 However, this lack of clarity has not deterred its widespread use among orthopedic surgeons. In this review, we aim to delineate the current understanding of PRP and its proven effectiveness in the treatment of rotator cuff tears, knee osteoarthritis, ulnar collateral ligament (UCL) tears, lateral epicondylitis, hamstring injuries, and Achilles tendinopathy.

Rotator Cuff Tears

Rotator cuff tears are one of the most common etiologies for shoulder pain and disability. The incidence continues to increase with the active aging population.3 Rotator cuff tears treated with arthroscopic repair have exhibited satisfactory pain relief and functional outcomes.4-7 Despite advances in fixation techniques, the quality and speed of tendon-to-bone healing remains unpredictable, with repaired tendons exhibiting inferior mechanical properties that are susceptible to re-tear.8-10

Numerous studies have investigated PRP application during arthroscopic rotator cuff repair (RCR) in an attempt to enhance and accelerate the repair process.11-15 However, wide variability exists among protocols of how and when PRP is utilized to augment the repair. Warth and colleagues16 performed a meta-analysis of 11 Level I/II studies evaluating RCR with PRP augmentation. With regards to clinical outcome scores, they found no significant difference in pre- and postoperative American Shoulder and Elbow Surgeons (ASES), Constant, Disability of the Arm, Shoulder and Hand (DASH), or visual analog scale (VAS) pain scores between those patients with or without PRP augmentation. However, they did note a significant increase in Constant scores when PRP was delivered to the tendon-bone interface rather than over the surface of the repair site. There was no significant difference in structural outcomes (evaluated by magnetic resonance imaging [MRI] re-tear rates) between those RCRs with and without PRP augmentation, except in those tears >3 cm in anterior-posterior length using double-row technique, with the PRP group exhibiting a significantly decreased re-tear rate (25.9% vs 57.1%).16 Zhao and colleagues17 reported similar results in a meta-analysis of 8 randomized controlled trials, exhibiting no significant differences in clinical outcome scores or re-tear rates after RCR with and without PRP augmentation. Overall, most studies have failed to demonstrate a significant benefit with regards to re-tear rates or shoulder-specific outcomes with the addition of PRP during arthroscopic RCR.

Knee Osteoarthritis

Osteoarthritis is the most common musculoskeletal disorder, with an estimated prevalence of 10% of the world’s population age 60 years and older.18 The knee is commonly symptomatic, resulting in pain, disability, and significant healthcare costs. Novel biologic, nonoperative therapies, including intra-articular viscosupplementation and PRP injections, have been proposed to treat the early stages of osteoarthritis to provide symptomatic relief and delay surgical intervention.

A multitude of studies have been performed investigating the effects of PRP on knee osteoarthritis, revealing mixed results.19-22 Campbell and colleagues23 published a 2015 systematic review of 3 overlapping meta-analyses comparing the outcomes of intra-articular injection of PRP vs control (hyaluronic acid [HA] or placebo) in 3278 knees. They reported a significant improvement in patient outcome scores for the PRP group when compared to control from 2 to 12 months after injection, but due to significant differences within the included studies, the ideal number of injections or time intervals between injections remains unclear. Meheux and colleagues24 reported a 2016 systematic review including 6 studies (817 knees) comparing PRP and HA injections. They demonstrated significantly better improvements in Western Ontario and McMaster Universities Arthritis Index (WOMAC) outcome scores with PRP vs HA injections at 3 and 12 months postinjection. Similarly, Smith25 conducted a Food and Drug Administration-sanctioned, randomized, double-blind, placebo-controlled clinical trial investigating the effects of intra-articular leukocyte-poor autologous conditioned plasma (ACP) in 30 patients. He reported an improvement in the ACP treatment group WOMAC scores by 78% compared to 7% improvement in the placebo group after 12 months. Despite the heterogeneity amongst studies, the majority of published data suggests better symptomatic relief in patients with early knee degenerative changes, and use of PRP may be considered in this population.

Ulnar Collateral Ligament Injuries

The anterior band of the UCL of the elbow provides stability to valgus stress. Overhead, high-velocity throwing athletes may cause repetitive injury to the UCL, resulting in partial or complete tears of the ligament. This may result in medial elbow pain, as well as decreased throwing velocity and accuracy. Athletes with complete UCL tears have few nonoperative treatment options and generally, operative treatment with UCL reconstruction is recommended for those athletes desiring to return to sport. However, it remains unclear how to definitively treat athletes with partial UCL tears. Recently, there has been an interest in treating these injuries with PRP in conjunction with physical therapy to facilitate a more predictable outcome.

Podesta and colleagues26 published a case series of 34 athletes with MRI-diagnosed partial UCL tears who underwent ultrasound-guided UCL injections and physical therapy. At an average follow-up of 70 weeks, they reported an average return to play (RTP) of 12 weeks, with significant improvements in Kerlan-Jobe Orthopaedic Clinic (KJOC) and DASH outcome scores, and decreased dynamic ulnohumeral joint widening to valgus stress on ultrasound. Most athletes (30/34) returned to their previous level of play, and 1 patient underwent subsequent UCL reconstruction. This study demonstrates that PRP may be used in conjunction with physical therapy and an interval throwing program for the treatment of partial UCL tears, but without a comparison control group, more studies are necessary to delineate the role of PRP in this population.

Lateral Elbow Epicondylitis

Lateral elbow epicondylitis, also known as “tennis elbow,” is thought to be caused by repetitive wrist extension and is more likely to present in patients with various comorbidities such as rotator cuff pathology or a history of smoking.27-29 The condition typically presents as radiating pain centered about the lateral epicondyle. Annual incidence ranges from 0.34% to 3%, with the most recent large-scale, population-based study estimating that nearly 1 million individuals in the United States develop lateral elbow epicondylitis each year.30 For the majority of patients, symptoms resolve after 6 to 12 months of various nonoperative or minimally invasive treatments.31-33 Those who develop chronic symptoms (>12 months) may benefit from surgical intervention.34 The use of PRP has become a contentious topic of debate in treating lateral epicondylitis. Its use and efficacy have been empirically examined and compared among more traditional treatments.35-37

In a small case-series of 6 patients, contrast-enhanced ultrasound imaging was utilized to demonstrate that PRP injection therapy may induce vascularization of the myotendinous junction of the common extensor tendon up to 6 months following injection.38 These physiologic changes may precede observable clinical improvements. Brklijac and colleagues39 prospectively followed 34 patients who had refractory symptoms despite conservative treatment and elected to undergo injection with PRP. At a mean follow-up of 26 weeks, 88.2% of the patients demonstrated improvements on their Oxford Elbow Score (OES). While potentially promising, case series lack large sample sizes, longitudinal analysis, and adequate control groups for comparative analyses of treatments, thereby increasing the likelihood of unintended selection bias.

Randomized controlled trials have demonstrated no difference between PRP and corticosteroid (CS) injection treatments in the short term for symptomatic lateral elbow epicondylitis. At 15 days, 1 month, and 6 months postinjection, no significant difference was found between PRP and CS injections in dynamometer strength measurements nor patient outcome scores (VAS, DASH, OES, and Mayo Clinic Performance Index for Elbow [MMCPIE]).40,41 In fact, multiple randomized controlled trials have demonstrated PRP to be less effective at 1 and 3 months compared to CS injections, as assessed by the Patient Rated Tennis-Elbow Evaluation (PRTEE) questionnaire, VAS, MMCPIE, and Nirschl scores.42,43 One mid-term, multi-center randomized controlled trial published by Mishra and colleagues44 compared PRP injections to an active control group, demonstrating a significant improvement in VAS pain scores at 24 weeks, but no difference in the PRTEE outcome. The available evidence indicates PRP injection therapy remains limited in utility for treatment of lateral epicondylitis, particularly in the short term when compared to CS injections. In the midterm to long term, PRP therapy may provide some benefit, but ultimately, well-designed prospective randomized controlled trials are needed to delineate the effects of PRP versus the natural course of tendon healing and symptom resolution.

 

 

Hamstring Injuries

Acute hamstring injuries are common across all levels and types of sport, particularly those in which sprinting or running is involved. While there is no consensus within the literature on how RTP after hamstring injury should be managed or defined, most injuries seem to resolve around 3 to 6 weeks.45 The proximal myotendinous junction of the long head of the biceps femoris and semitendinosus are commonly associated with significant pain and edema after acute hamstring injury.46 The amount of edema resulting from grade 1 and 2 hamstring injuries has been found to correlate (minimally) with time to RTP in elite athletes.47 PRP injection near the proximal myotendinous hamstring origin has been theorized to help speed the recovery process after acute hamstring injury. To date, the literature demonstrates mixed and limited benefit of PRP injection therapy for acute hamstring injury.

Few studies have shown improvements of PRP therapy over typical nonoperative management (rest, physical therapy, nonsteroidal anti-inflammatory drugs) in acute hamstring injury, but the results must be interpreted carefully.48,49 Wetzel and colleagues48 retrospectively reviewed 17 patients with acute hamstring injury, 12 of whom failed typical management and received PRP injection at the hamstring origin. This group demonstrated significant improvements in their VAS and Nirschl scores at follow-up, whereas the 5 patients who did not receive the injection did not. However, this study exhibited significant limitations inherent to a retrospective review with a small sample size. Hamid and colleagues49 conducted a randomized controlled trial of 24 athletes with diagnosed grade 2a acute hamstring injuries, comparing autologous PRP therapy combined with a rehabilitation program versus rehabilitation program alone. RTP, changes in pain severity (Brief Pain Injury-Short Form [BPI-SF] questions 2-6), and pain interference (BPI-SF questions 9A-9G) scores over time were examined. Athletes in the PRP group exhibited no difference in outcomes scores, but returned to play sooner than controls (26.7 vs 42.5 days).

Mejia and Bradley50 have reported their experience in treating 12 National Football League (NFL) players with acute MRI grade 1 or 2 hamstring injuries with a series of PRP injections at the site of injury. They found a 1-game difference in earlier RTP when compared to the predicted RTP based on MRI grading. Similarly, Hamid and colleagues49 performed a randomized control trial published in 2014, reporting an earlier RTP (26.7 vs 42.5 days) when comparing single PRP injection vs rehabilitation alone in 28 patients diagnosed with acute ultrasound grade 2 hamstring injuries. On the contrary, a small case-control study of NFL players and a retrospective cohort study of athletes with severe hamstring injuries demonstrated no difference in RTP when PRP injected patients were compared with controls.51,52 Larger randomized controlled trials have demonstrated comparable results, including a study of 90 professional athletes in whom a single PRP injection did not decrease RTP or lessen the risk of re-injury at 2 and 6 months.53 In another large multicenter randomized controlled trial examining 80 competitive and recreational athletes, PRP did not accelerate RTP, lessen the risk of 2-month or 1-year re-injury rate, or improve secondary measures of MRI parameters, subjective patient satisfaction, or the hamstring outcome score.54 Although further study is warranted, available evidence suggests limited utility of PRP injection in the treatment of acute hamstring injuries.

Achilles Tendinopathy

Noninsertional Achilles tendinopathy is a common source of pain for both recreational and competitive athletes. Typically thought of as an overuse syndrome, Achilles tendinopathy may result in significant pain and swelling, often at the site of its tenuous blood supply, approximately 2 to 7 cm proximal to its insertion.55 Conservative management frequently begins with rest, activity/shoe modification, physical therapy, and eccentric loading exercises.56 For those whom conservative management has failed to reduce symptoms after 6 months, more invasive treatment options may be considered. Peritendinous PRP injection has become an alternative approach in treating Achilles tendinopathy refractory to conservative treatment.

In the few randomized controlled trials published, the data demonstrates no significant improvements in clinical outcomes from PRP injection for Achilles tendinopathy. Kearney and colleagues57 conducted a pilot study of 20 patients randomized into PRP injection or eccentric loading program for mid-substance Achilles tendinopathy, in which Victorian Institute of Sports Assessment (VISA-A), EuroQol 5 dimensions questionnaire (EQ-5D), and complications associated with the injection were recorded at 6 weeks, 3 months, and 6 months. Although this was a pilot study with a small sample size, no significant difference was found between groups across these time periods. Similarly, de Vos and colleagues58,59 conducted a double-blind randomized controlled trial of 54 patients with chronic mid-substance Achilles tendinopathy and randomized them into eccentric exercise therapy with either a PRP injection or a saline injected placebo groups. VISA-A scores were recorded and imaging parameters assessing tendon structure by ultrasonographic tissue characterization and color Doppler ultrasonography were taken with follow-up at 6, 12, and 24 weeks. VISA-A scores improved significantly in both groups after 24 weeks, but the difference was not statistically significant between groups. In addition, tendon structure and neovascularization (exhibited by color Doppler ultrasonography) improved in both groups, with no significant difference between groups. The current literature does not support the use of PRP in treatment of Achilles tendinopathy, as it has failed to reveal additional benefits over conventional treatment alone. Future prospective, well-designed randomized controlled trials with large sample sizes will need to be conducted to ultimately conclude whether or not PRP deserves a role in the treatment of Achilles tendinopathy.

Summary

In theory, the use of PRP within orthopedic surgery makes a great deal of sense to accelerate and augment the healing process of the aforementioned musculoskeletal injuries. However, the vast majority of published literature is Level III and IV evidence. Future research may provide the missing critical information of optimal growth factor, platelet, and leukocyte concentrations necessary for the desired effect, as well as the appropriate delivery method and timing of PRP application in different target tissues. Evidence-based guidelines to direct the use of PRP will benefit from more homogenous, repeatable, and randomized controlled trials.

Platelet-rich plasma (PRP) is a refined product of autologous blood with a platelet concentration greater than that of whole blood. It is prepared via plasmapheresis utilizing a 2-stage centrifugation process and more than 40 commercially available systems are marketed to concentrate whole blood to PRP.1 It is rich in biologic factors (growth factors, cytokines, proteins, cellular components) essential to the body’s response to injury. For this reason, it was first used in oromaxillofacial surgery in the 1950s, but its effects on the musculoskeletal system have yet to be clearly elucidated.2 However, this lack of clarity has not deterred its widespread use among orthopedic surgeons. In this review, we aim to delineate the current understanding of PRP and its proven effectiveness in the treatment of rotator cuff tears, knee osteoarthritis, ulnar collateral ligament (UCL) tears, lateral epicondylitis, hamstring injuries, and Achilles tendinopathy.

Rotator Cuff Tears

Rotator cuff tears are one of the most common etiologies for shoulder pain and disability. The incidence continues to increase with the active aging population.3 Rotator cuff tears treated with arthroscopic repair have exhibited satisfactory pain relief and functional outcomes.4-7 Despite advances in fixation techniques, the quality and speed of tendon-to-bone healing remains unpredictable, with repaired tendons exhibiting inferior mechanical properties that are susceptible to re-tear.8-10

Numerous studies have investigated PRP application during arthroscopic rotator cuff repair (RCR) in an attempt to enhance and accelerate the repair process.11-15 However, wide variability exists among protocols of how and when PRP is utilized to augment the repair. Warth and colleagues16 performed a meta-analysis of 11 Level I/II studies evaluating RCR with PRP augmentation. With regards to clinical outcome scores, they found no significant difference in pre- and postoperative American Shoulder and Elbow Surgeons (ASES), Constant, Disability of the Arm, Shoulder and Hand (DASH), or visual analog scale (VAS) pain scores between those patients with or without PRP augmentation. However, they did note a significant increase in Constant scores when PRP was delivered to the tendon-bone interface rather than over the surface of the repair site. There was no significant difference in structural outcomes (evaluated by magnetic resonance imaging [MRI] re-tear rates) between those RCRs with and without PRP augmentation, except in those tears >3 cm in anterior-posterior length using double-row technique, with the PRP group exhibiting a significantly decreased re-tear rate (25.9% vs 57.1%).16 Zhao and colleagues17 reported similar results in a meta-analysis of 8 randomized controlled trials, exhibiting no significant differences in clinical outcome scores or re-tear rates after RCR with and without PRP augmentation. Overall, most studies have failed to demonstrate a significant benefit with regards to re-tear rates or shoulder-specific outcomes with the addition of PRP during arthroscopic RCR.

Knee Osteoarthritis

Osteoarthritis is the most common musculoskeletal disorder, with an estimated prevalence of 10% of the world’s population age 60 years and older.18 The knee is commonly symptomatic, resulting in pain, disability, and significant healthcare costs. Novel biologic, nonoperative therapies, including intra-articular viscosupplementation and PRP injections, have been proposed to treat the early stages of osteoarthritis to provide symptomatic relief and delay surgical intervention.

A multitude of studies have been performed investigating the effects of PRP on knee osteoarthritis, revealing mixed results.19-22 Campbell and colleagues23 published a 2015 systematic review of 3 overlapping meta-analyses comparing the outcomes of intra-articular injection of PRP vs control (hyaluronic acid [HA] or placebo) in 3278 knees. They reported a significant improvement in patient outcome scores for the PRP group when compared to control from 2 to 12 months after injection, but due to significant differences within the included studies, the ideal number of injections or time intervals between injections remains unclear. Meheux and colleagues24 reported a 2016 systematic review including 6 studies (817 knees) comparing PRP and HA injections. They demonstrated significantly better improvements in Western Ontario and McMaster Universities Arthritis Index (WOMAC) outcome scores with PRP vs HA injections at 3 and 12 months postinjection. Similarly, Smith25 conducted a Food and Drug Administration-sanctioned, randomized, double-blind, placebo-controlled clinical trial investigating the effects of intra-articular leukocyte-poor autologous conditioned plasma (ACP) in 30 patients. He reported an improvement in the ACP treatment group WOMAC scores by 78% compared to 7% improvement in the placebo group after 12 months. Despite the heterogeneity amongst studies, the majority of published data suggests better symptomatic relief in patients with early knee degenerative changes, and use of PRP may be considered in this population.

Ulnar Collateral Ligament Injuries

The anterior band of the UCL of the elbow provides stability to valgus stress. Overhead, high-velocity throwing athletes may cause repetitive injury to the UCL, resulting in partial or complete tears of the ligament. This may result in medial elbow pain, as well as decreased throwing velocity and accuracy. Athletes with complete UCL tears have few nonoperative treatment options and generally, operative treatment with UCL reconstruction is recommended for those athletes desiring to return to sport. However, it remains unclear how to definitively treat athletes with partial UCL tears. Recently, there has been an interest in treating these injuries with PRP in conjunction with physical therapy to facilitate a more predictable outcome.

Podesta and colleagues26 published a case series of 34 athletes with MRI-diagnosed partial UCL tears who underwent ultrasound-guided UCL injections and physical therapy. At an average follow-up of 70 weeks, they reported an average return to play (RTP) of 12 weeks, with significant improvements in Kerlan-Jobe Orthopaedic Clinic (KJOC) and DASH outcome scores, and decreased dynamic ulnohumeral joint widening to valgus stress on ultrasound. Most athletes (30/34) returned to their previous level of play, and 1 patient underwent subsequent UCL reconstruction. This study demonstrates that PRP may be used in conjunction with physical therapy and an interval throwing program for the treatment of partial UCL tears, but without a comparison control group, more studies are necessary to delineate the role of PRP in this population.

Lateral Elbow Epicondylitis

Lateral elbow epicondylitis, also known as “tennis elbow,” is thought to be caused by repetitive wrist extension and is more likely to present in patients with various comorbidities such as rotator cuff pathology or a history of smoking.27-29 The condition typically presents as radiating pain centered about the lateral epicondyle. Annual incidence ranges from 0.34% to 3%, with the most recent large-scale, population-based study estimating that nearly 1 million individuals in the United States develop lateral elbow epicondylitis each year.30 For the majority of patients, symptoms resolve after 6 to 12 months of various nonoperative or minimally invasive treatments.31-33 Those who develop chronic symptoms (>12 months) may benefit from surgical intervention.34 The use of PRP has become a contentious topic of debate in treating lateral epicondylitis. Its use and efficacy have been empirically examined and compared among more traditional treatments.35-37

In a small case-series of 6 patients, contrast-enhanced ultrasound imaging was utilized to demonstrate that PRP injection therapy may induce vascularization of the myotendinous junction of the common extensor tendon up to 6 months following injection.38 These physiologic changes may precede observable clinical improvements. Brklijac and colleagues39 prospectively followed 34 patients who had refractory symptoms despite conservative treatment and elected to undergo injection with PRP. At a mean follow-up of 26 weeks, 88.2% of the patients demonstrated improvements on their Oxford Elbow Score (OES). While potentially promising, case series lack large sample sizes, longitudinal analysis, and adequate control groups for comparative analyses of treatments, thereby increasing the likelihood of unintended selection bias.

Randomized controlled trials have demonstrated no difference between PRP and corticosteroid (CS) injection treatments in the short term for symptomatic lateral elbow epicondylitis. At 15 days, 1 month, and 6 months postinjection, no significant difference was found between PRP and CS injections in dynamometer strength measurements nor patient outcome scores (VAS, DASH, OES, and Mayo Clinic Performance Index for Elbow [MMCPIE]).40,41 In fact, multiple randomized controlled trials have demonstrated PRP to be less effective at 1 and 3 months compared to CS injections, as assessed by the Patient Rated Tennis-Elbow Evaluation (PRTEE) questionnaire, VAS, MMCPIE, and Nirschl scores.42,43 One mid-term, multi-center randomized controlled trial published by Mishra and colleagues44 compared PRP injections to an active control group, demonstrating a significant improvement in VAS pain scores at 24 weeks, but no difference in the PRTEE outcome. The available evidence indicates PRP injection therapy remains limited in utility for treatment of lateral epicondylitis, particularly in the short term when compared to CS injections. In the midterm to long term, PRP therapy may provide some benefit, but ultimately, well-designed prospective randomized controlled trials are needed to delineate the effects of PRP versus the natural course of tendon healing and symptom resolution.

 

 

Hamstring Injuries

Acute hamstring injuries are common across all levels and types of sport, particularly those in which sprinting or running is involved. While there is no consensus within the literature on how RTP after hamstring injury should be managed or defined, most injuries seem to resolve around 3 to 6 weeks.45 The proximal myotendinous junction of the long head of the biceps femoris and semitendinosus are commonly associated with significant pain and edema after acute hamstring injury.46 The amount of edema resulting from grade 1 and 2 hamstring injuries has been found to correlate (minimally) with time to RTP in elite athletes.47 PRP injection near the proximal myotendinous hamstring origin has been theorized to help speed the recovery process after acute hamstring injury. To date, the literature demonstrates mixed and limited benefit of PRP injection therapy for acute hamstring injury.

Few studies have shown improvements of PRP therapy over typical nonoperative management (rest, physical therapy, nonsteroidal anti-inflammatory drugs) in acute hamstring injury, but the results must be interpreted carefully.48,49 Wetzel and colleagues48 retrospectively reviewed 17 patients with acute hamstring injury, 12 of whom failed typical management and received PRP injection at the hamstring origin. This group demonstrated significant improvements in their VAS and Nirschl scores at follow-up, whereas the 5 patients who did not receive the injection did not. However, this study exhibited significant limitations inherent to a retrospective review with a small sample size. Hamid and colleagues49 conducted a randomized controlled trial of 24 athletes with diagnosed grade 2a acute hamstring injuries, comparing autologous PRP therapy combined with a rehabilitation program versus rehabilitation program alone. RTP, changes in pain severity (Brief Pain Injury-Short Form [BPI-SF] questions 2-6), and pain interference (BPI-SF questions 9A-9G) scores over time were examined. Athletes in the PRP group exhibited no difference in outcomes scores, but returned to play sooner than controls (26.7 vs 42.5 days).

Mejia and Bradley50 have reported their experience in treating 12 National Football League (NFL) players with acute MRI grade 1 or 2 hamstring injuries with a series of PRP injections at the site of injury. They found a 1-game difference in earlier RTP when compared to the predicted RTP based on MRI grading. Similarly, Hamid and colleagues49 performed a randomized control trial published in 2014, reporting an earlier RTP (26.7 vs 42.5 days) when comparing single PRP injection vs rehabilitation alone in 28 patients diagnosed with acute ultrasound grade 2 hamstring injuries. On the contrary, a small case-control study of NFL players and a retrospective cohort study of athletes with severe hamstring injuries demonstrated no difference in RTP when PRP injected patients were compared with controls.51,52 Larger randomized controlled trials have demonstrated comparable results, including a study of 90 professional athletes in whom a single PRP injection did not decrease RTP or lessen the risk of re-injury at 2 and 6 months.53 In another large multicenter randomized controlled trial examining 80 competitive and recreational athletes, PRP did not accelerate RTP, lessen the risk of 2-month or 1-year re-injury rate, or improve secondary measures of MRI parameters, subjective patient satisfaction, or the hamstring outcome score.54 Although further study is warranted, available evidence suggests limited utility of PRP injection in the treatment of acute hamstring injuries.

Achilles Tendinopathy

Noninsertional Achilles tendinopathy is a common source of pain for both recreational and competitive athletes. Typically thought of as an overuse syndrome, Achilles tendinopathy may result in significant pain and swelling, often at the site of its tenuous blood supply, approximately 2 to 7 cm proximal to its insertion.55 Conservative management frequently begins with rest, activity/shoe modification, physical therapy, and eccentric loading exercises.56 For those whom conservative management has failed to reduce symptoms after 6 months, more invasive treatment options may be considered. Peritendinous PRP injection has become an alternative approach in treating Achilles tendinopathy refractory to conservative treatment.

In the few randomized controlled trials published, the data demonstrates no significant improvements in clinical outcomes from PRP injection for Achilles tendinopathy. Kearney and colleagues57 conducted a pilot study of 20 patients randomized into PRP injection or eccentric loading program for mid-substance Achilles tendinopathy, in which Victorian Institute of Sports Assessment (VISA-A), EuroQol 5 dimensions questionnaire (EQ-5D), and complications associated with the injection were recorded at 6 weeks, 3 months, and 6 months. Although this was a pilot study with a small sample size, no significant difference was found between groups across these time periods. Similarly, de Vos and colleagues58,59 conducted a double-blind randomized controlled trial of 54 patients with chronic mid-substance Achilles tendinopathy and randomized them into eccentric exercise therapy with either a PRP injection or a saline injected placebo groups. VISA-A scores were recorded and imaging parameters assessing tendon structure by ultrasonographic tissue characterization and color Doppler ultrasonography were taken with follow-up at 6, 12, and 24 weeks. VISA-A scores improved significantly in both groups after 24 weeks, but the difference was not statistically significant between groups. In addition, tendon structure and neovascularization (exhibited by color Doppler ultrasonography) improved in both groups, with no significant difference between groups. The current literature does not support the use of PRP in treatment of Achilles tendinopathy, as it has failed to reveal additional benefits over conventional treatment alone. Future prospective, well-designed randomized controlled trials with large sample sizes will need to be conducted to ultimately conclude whether or not PRP deserves a role in the treatment of Achilles tendinopathy.

Summary

In theory, the use of PRP within orthopedic surgery makes a great deal of sense to accelerate and augment the healing process of the aforementioned musculoskeletal injuries. However, the vast majority of published literature is Level III and IV evidence. Future research may provide the missing critical information of optimal growth factor, platelet, and leukocyte concentrations necessary for the desired effect, as well as the appropriate delivery method and timing of PRP application in different target tissues. Evidence-based guidelines to direct the use of PRP will benefit from more homogenous, repeatable, and randomized controlled trials.

References

1.    Hsu WK, Mishra A, Rodeo SR, et al. Platelet-rich plasma in orthopaedic applications: evidence-based recommendations for treatment. J Am Acad Orthop Surg. 2013;21(12):739-748.

2.    Marx RE. Platelet-rich plasma: evidence to support its use. J Oral Maxillofac Surg. 2004;62(4):489-496.

3.    Jo CH, Kim JE, Yoon KS, et al. Does platelet-rich plasma accelerate recovery after rotator cuff repair? A prospective cohort study. Am J Sports Med. 2011;39(10):2082-2090.

4.    Burkhart SS, Danaceau SM, Pearce CE Jr. Arthroscopic rotator cuff repair: Analysis of results by tear size and by repair technique-margin convergence versus direct tendon-to-bone repair. Arthroscopy. 2001;17(9):905-912.

5.    Severud EL, Ruotolo C, Abbott DD, Nottage WM. All-arthroscopic versus mini-open rotator cuff repair: A long-term retrospective outcome comparison. Arthroscopy. 2003;19(3):234-238.

6.    Huang R, Wang S, Wang Y, Qin X, Sun Y. Systematic review of all-arthroscopic versus mini-open repair of rotator cuff tears: a meta-analysis. Sci Rep. 2016;6:22857.

7.    Watson EM, Sonnabend DH. Outcome of rotator cuff repair. J Shoulder Elbow Surg. 2002;11(3):201-211.

8.    Butler DL, Juncosa N, Dressler MR. Functional efficacy of tendon repair processes. Annu Rev Biomed Eng. 2004;6:303-329.

9.    Galatz LM, Ball CM, Teefey SA, Middleton WD, Yamaguchi K. The outcome and repair integrity of completely arthroscopically repaired large and massive rotator cuff tears. J Bone Joint Surg Am. 2004;86-A(2):219-224.

10.  Lafosse L, Brozska R, Toussaint B, Gobezie R. The outcome and structural integrity of arthroscopic rotator cuff repair with use of the double-row suture anchor technique. J Bone Joint Surg Am. 2007;89(7):1533-1541.

11.  Castricini R, Longo UG, De Benedetto M, et al. Platelet-rich plasma augmentation for arthroscopic rotator cuff repair: a randomized controlled trial. Am J Sports Med. 2011;39(2):258-265.

12.  Randelli P, Arrigoni P, Ragone V, Aliprandi A, Cabitza P. Platelet rich plasma in arthroscopic rotator cuff repair: a prospective RCT study, 2-year follow-up. J Shoulder Elbow Surg. 2011;20(4):518-528.

13.  Weber SC, Kauffman JI, Parise C, Weber SJ, Katz SD. Platelet-rich fibrin matrix in the management of arthroscopic repair of the rotator cuff: a prospective, randomized, double-blinded study. Am J Sports Med. 2013;41(2):263-270.

14.  Gumina S, Campagna V, Ferrazza G, et al. Use of platelet-leukocyte membrane in arthroscopic repair of large rotator cuff tears: a prospective randomized study. J Bone Joint Surg Am. 2012;94(15):1345-1352.

15.  Rodeo SA, Delos D, Williams RJ, Adler RS, Pearle A, Warren RF.  The effect of platelet-rich fibrin matrix on rotator cuff tendon healing: a prospective, randomized clinical study. Am J Sports Med. 2012;40(6):1234-1241.

16.  Warth RJ, Dornan GJ, James EW, Horan MP, Millett PJ. Clinical and structural outcomes after arthroscopic repair of full-thickness rotator cuff tears with and without platelet-rich product supplementation: a meta-analysis and meta-regression. Arthroscopy. 2015;31(2):306-320.

17.  Zhao JG, Zhao L, Jiang YX, Wang ZL, Wang J, Zhang P. Platelet-rich plasma in arthroscopic rotator cuff repair: a meta-analysis of randomized controlled trials. Arthroscopy. 2015;31(1):125-135.

18.  Glyn-Jones S, Palmer AJ, Agricola R, et al. Osteoarthritis. Lancet. 2015;386(9991):376-387.

19.  Cerza F, Carni S, Carcangiu A, et al. Comparison between hyaluronic acid and platelet-rich plasma, intra-articular infiltration in the treatment of gonarthrosis. Am J Sports Med. 2012;40(12):2822-2827.

20.  Filardo G, Kon E, Di Martino A, et al. Platelet-rich plasma vs hyaluronic acid to treat knee degenerative pathology: study design and preliminary results of a randomized controlled trial. BMC Musculoskelet Disord. 2012;13:229.

21.  Patel S, Dhillon MS, Aggarwal S, Marwaha N, Jain A. Treatment with platelet-rich plasma is more effective than placebo for knee osteoarthritis: a prospective, double-blind, randomized trial. Am J Sports Med. 2013;41(2):356-364.

22.  Sanchez M, Fiz N, Azofra J, et al. A randomized clinical trial evaluating plasma rich in growth factors (PRGF-Endoret) versus hyaluronic acid in the short-term treatment of symptomatic knee osteoarthritis. Arthroscopy. 2012;28(8):1070-1078.

23.  Campbell KA, Saltzman BM, Mascarenhas R, et al. Does intra-articular platelet-rich plasma injection provide clinically superior outcomes compared with other therapies in the treatment of knee osteoarthritis? A systematic review of overlapping meta-analyses. Arthroscopy. 2015;31(11):2213-2221.

24.  Meheux CJ, McCulloch PC, Lintner DM, Varner KE, Harris JD. Efficacy of intra-articular platelet-rich plasma injections in knee osteoarthritis: A systematic review. Arthroscopy. 2016;32(3):495-505.

25.  Smith PA. Intra-articular autologous conditioned plasma injections provide safe and efficacious treatment for knee osteoarthritis: An FDA-sanctioned, randomized, double-blind, placebo-controlled clinical trial. Am J Sports Med. 2016;44(4):884-891.

26.   Podesta L, Crow SA, Volkmer D, Bert T, Yocum LA. Treatment of partial ulnar collateral ligament tears in the elbow with platelet-rich plasma. Am J Sports Med. 2013;41(7):1689-1694.

27.  Herquelot E, Gueguen A, Roquelaure Y, et al. Work-related risk factors for incidence of lateral epicondylitis in a large working population. Scand J Work Environ Health. 2013;39(6):578-588.

28.  Titchener AG, Fakis A, Tambe AA, Smith C, Hubbard RB, Clark DI. Risk factors in lateral epicondylitis (tennis elbow): a case-control study. J Hand Surg Eur Vol. 2013;38(2):159-164.

29.  Gruchow HW, Pelletier D. An epidemiologic study of tennis elbow. Incidence, recurrence, and effectiveness of prevention strategies. Am J Sports Med. 1979;7(4):234-238.

30.  Sanders TL Jr, Maradit Kremers H, Bryan AJ, Ransom JE, Smith J, Morrey BF.  The epidemiology and health care burden of tennis elbow: a population-based study. Am J Sports Med. 2015;43(5):1066-1071.

31.  Coonrad RW, Hooper WR. Tennis elbow: its course, natural history, conservative and surgical management. J Bone Joint Surg Am. 1973;55(6):1177-1182.

32.  Taylor SA, Hannafin JA. Evaluation and management of elbow tendinopathy. Sports Health. 2012;4(5):384-393.

33.  Sims SE, Miller K, Elfar JC, Hammert WC. Non-surgical treatment of lateral epicondylitis: a systematic review of randomized controlled trials. Hand (NY). 2014;9(4):419-446.

34.  Brummel J, Baker CL 3rd, Hopkins R, Baker CL Jr. Epicondylitis: lateral. Sports Med Arthrosc. 2014;22(3):e1-e6.

35.  de Vos RJ, Windt J, Weir A. Strong evidence against platelet-rich plasma injections for chronic lateral epicondylar tendinopathy: a systematic review. Br J Sports Med. 2014;48(12):952-956.

36.  Ahmad Z, Brooks R, Kang SN, et al. The effect of platelet-rich plasma on clinical outcomes in lateral epicondylitis. Arthroscopy. 2013;29(11):1851-1862.

37.  Arirachakaran A, Sukthuayat A, Sisayanarane T, Laoratanavoraphong S, Kanchanatawan W, Kongtharvonskul J. Platelet-rich plasma versus autologous blood versus steroid injection in lateral epicondylitis: systematic review and network meta-analysis. J Orthop Traumatol. 2016;17(2):101-112.

38.  Chaudhury S, de La Lama M, Adler RS, et al. Platelet-rich plasma for the treatment of lateral epicondylitis: sonographic assessment of tendon morphology and vascularity (pilot study). Skeletal Radiol. 2013;42(1):91-97.

39.  Brkljac M, Kumar S, Kalloo D, Hirehal K. The effect of platelet-rich plasma injection on lateral epicondylitis following failed conservative management. J Orthop. 2015;12(Suppl 2):S166-S170.

40.  Yadav R, Kothari SY, Borah D. Comparison of local injection of platelet rich plasma and corticosteroids in the treatment of lateral epicondylitis of humerus. J Clin Diagn Res. 2015;9(7):RC05-RC07.

41.  Gautam VK, Verma S, Batra S, Bhatnagar N, Arora S. Platelet-rich plasma versus corticosteroid injection for recalcitrant lateral epicondylitis: clinical and ultrasonographic evaluation. J Orthop Surg (Hong Kong). 2015;23(1):1-5.

42.  Krogh TP, Fredberg U, Stengaard-Pedersen K, Christensen R, Jensen P, Ellingsen T. Treatment of lateral epicondylitis with platelet-rich plasma, glucocorticoid, or saline: a randomized, double-blind, placebo-controlled trial. Am J Sports Med. 2013;41(3):625-635.

43.  Behera P, Dhillon M, Aggarwal S, Marwaha N, Prakash M. Leukocyte-poor platelet-rich plasma versus bupivacaine for recalcitrant lateral epicondylar tendinopathy. J Orthop Surg (Hong Kong). 2015;23(1):6-10.

44.  Mishra AK, Skrepnik NV, Edwards SG, et al. Efficacy of platelet-rich plasma for chronic tennis elbow: a double-blind, prospective, multicenter, randomized controlled trial of 230 patients. Am J Sports Med. 2014;42(2):463-471.

45.  van der Horst N, van de Hoef S, Reurink G, Huisstede B, Backx F. Return to play after hamstring injuries: a qualitative systematic review of definitions and criteria. Sports Med. 2016;46(6):899-912.

46.  Crema MD, Guermazi A, Tol JL, Niu J, Hamilton B, Roemer FW. Acute hamstring injury in football players: Association between anatomical location and extent of injury-A large single-center MRI report. J Sci Med Sport. 2016;19(4):317-322.

47.  Ekstrand J, Lee JC, Healy JC. MRI findings and return to play in football: a prospective analysis of 255 hamstring injuries in the UEFA Elite Club Injury Study. Br J Sports Med. 2016;50(12):738-743.

48.  Wetzel RJ, Patel RM, Terry MA. Platelet-rich plasma as an effective treatment for proximal hamstring injuries. Orthopedics. 2013;36(1):e64-e70.

49.  Hamid A, Mohamed Ali MR, Yusof A, George J, Lee LP. Platelet-rich plasma injections for the treatment of hamstring injuries: a randomized controlled trial. Am J Sports Med. 2014;42(10):2410-2418.

50.  Mejia HA, Bradley JP.  The effects of platelet-rich plasma on muscle: basic science and clinical application. Operative Techniques in Sports Medicine. 2011;19(3):149-153.

51.  Guillodo Y, Madouas G, Simon T, Le Dauphin H, Saraux A. Platelet-rich plasma (PRP) treatment of sports-related severe acute hamstring injuries. Muscles Ligaments Tendons J. 2015;5(4):284-288.

52.  Rettig AC, Meyer S, Bhadra AK. Platelet-rich plasma in addition to rehabilitation for acute hamstring injuries in NFL players: Clinical effects and time to return to play. Orthop J Sports Med. 2013;1(1):2325967113494354.

53.   Hamilton B, Tol JL, Almusa E, et al. Platelet-rich plasma does not enhance return to play in hamstring injuries: a randomised controlled trial. Br J Sports Med. 2015;49(14):943-950.

54.  Reurink G, Goudswaard GJ, Moen MH, et al. Rationale, secondary outcome scores and 1-year follow-up of a randomised trial of platelet-rich plasma injections in acute hamstring muscle injury: the Dutch Hamstring Injection Therapy study. Br J Sports Med. 2015;49(18):1206-1212.

55.  Kujala UM, Sarna S, Kaprio J. Cumulative incidence of achilles tendon rupture and tendinopathy in male former elite athletes. Clin J Sport Med. 2005;15(3):133-135.

56.  Alfredson H. Clinical commentary of the evolution of the treatment for chronic painful mid-portion Achilles tendinopathy. Braz J Phys Ther. 2015;19(5):429-432.

57.  Kearney RS, Parsons N, Costa ML. Achilles tendinopathy management: A pilot randomised controlled trial comparing platelet-rich plasma injection with an eccentric loading programme. Bone Joint Res. 2013;2(10):227-232.

58.  de Vos RJ, Weir A, Tol JL, Verhaar JA, Weinans H, van Schie HT. No effects of PRP on ultrasonographic tendon structure and neovascularisation in chronic midportion Achilles tendinopathy. Br J Sports Med. 2011;45(5):387-392.

59.  de Vos RJ, Weir A, van Schie HT, et al. Platelet-rich plasma injection for chronic Achilles tendinopathy: a randomized controlled trial. JAMA. 2010;303(2):144-149.

References

1.    Hsu WK, Mishra A, Rodeo SR, et al. Platelet-rich plasma in orthopaedic applications: evidence-based recommendations for treatment. J Am Acad Orthop Surg. 2013;21(12):739-748.

2.    Marx RE. Platelet-rich plasma: evidence to support its use. J Oral Maxillofac Surg. 2004;62(4):489-496.

3.    Jo CH, Kim JE, Yoon KS, et al. Does platelet-rich plasma accelerate recovery after rotator cuff repair? A prospective cohort study. Am J Sports Med. 2011;39(10):2082-2090.

4.    Burkhart SS, Danaceau SM, Pearce CE Jr. Arthroscopic rotator cuff repair: Analysis of results by tear size and by repair technique-margin convergence versus direct tendon-to-bone repair. Arthroscopy. 2001;17(9):905-912.

5.    Severud EL, Ruotolo C, Abbott DD, Nottage WM. All-arthroscopic versus mini-open rotator cuff repair: A long-term retrospective outcome comparison. Arthroscopy. 2003;19(3):234-238.

6.    Huang R, Wang S, Wang Y, Qin X, Sun Y. Systematic review of all-arthroscopic versus mini-open repair of rotator cuff tears: a meta-analysis. Sci Rep. 2016;6:22857.

7.    Watson EM, Sonnabend DH. Outcome of rotator cuff repair. J Shoulder Elbow Surg. 2002;11(3):201-211.

8.    Butler DL, Juncosa N, Dressler MR. Functional efficacy of tendon repair processes. Annu Rev Biomed Eng. 2004;6:303-329.

9.    Galatz LM, Ball CM, Teefey SA, Middleton WD, Yamaguchi K. The outcome and repair integrity of completely arthroscopically repaired large and massive rotator cuff tears. J Bone Joint Surg Am. 2004;86-A(2):219-224.

10.  Lafosse L, Brozska R, Toussaint B, Gobezie R. The outcome and structural integrity of arthroscopic rotator cuff repair with use of the double-row suture anchor technique. J Bone Joint Surg Am. 2007;89(7):1533-1541.

11.  Castricini R, Longo UG, De Benedetto M, et al. Platelet-rich plasma augmentation for arthroscopic rotator cuff repair: a randomized controlled trial. Am J Sports Med. 2011;39(2):258-265.

12.  Randelli P, Arrigoni P, Ragone V, Aliprandi A, Cabitza P. Platelet rich plasma in arthroscopic rotator cuff repair: a prospective RCT study, 2-year follow-up. J Shoulder Elbow Surg. 2011;20(4):518-528.

13.  Weber SC, Kauffman JI, Parise C, Weber SJ, Katz SD. Platelet-rich fibrin matrix in the management of arthroscopic repair of the rotator cuff: a prospective, randomized, double-blinded study. Am J Sports Med. 2013;41(2):263-270.

14.  Gumina S, Campagna V, Ferrazza G, et al. Use of platelet-leukocyte membrane in arthroscopic repair of large rotator cuff tears: a prospective randomized study. J Bone Joint Surg Am. 2012;94(15):1345-1352.

15.  Rodeo SA, Delos D, Williams RJ, Adler RS, Pearle A, Warren RF.  The effect of platelet-rich fibrin matrix on rotator cuff tendon healing: a prospective, randomized clinical study. Am J Sports Med. 2012;40(6):1234-1241.

16.  Warth RJ, Dornan GJ, James EW, Horan MP, Millett PJ. Clinical and structural outcomes after arthroscopic repair of full-thickness rotator cuff tears with and without platelet-rich product supplementation: a meta-analysis and meta-regression. Arthroscopy. 2015;31(2):306-320.

17.  Zhao JG, Zhao L, Jiang YX, Wang ZL, Wang J, Zhang P. Platelet-rich plasma in arthroscopic rotator cuff repair: a meta-analysis of randomized controlled trials. Arthroscopy. 2015;31(1):125-135.

18.  Glyn-Jones S, Palmer AJ, Agricola R, et al. Osteoarthritis. Lancet. 2015;386(9991):376-387.

19.  Cerza F, Carni S, Carcangiu A, et al. Comparison between hyaluronic acid and platelet-rich plasma, intra-articular infiltration in the treatment of gonarthrosis. Am J Sports Med. 2012;40(12):2822-2827.

20.  Filardo G, Kon E, Di Martino A, et al. Platelet-rich plasma vs hyaluronic acid to treat knee degenerative pathology: study design and preliminary results of a randomized controlled trial. BMC Musculoskelet Disord. 2012;13:229.

21.  Patel S, Dhillon MS, Aggarwal S, Marwaha N, Jain A. Treatment with platelet-rich plasma is more effective than placebo for knee osteoarthritis: a prospective, double-blind, randomized trial. Am J Sports Med. 2013;41(2):356-364.

22.  Sanchez M, Fiz N, Azofra J, et al. A randomized clinical trial evaluating plasma rich in growth factors (PRGF-Endoret) versus hyaluronic acid in the short-term treatment of symptomatic knee osteoarthritis. Arthroscopy. 2012;28(8):1070-1078.

23.  Campbell KA, Saltzman BM, Mascarenhas R, et al. Does intra-articular platelet-rich plasma injection provide clinically superior outcomes compared with other therapies in the treatment of knee osteoarthritis? A systematic review of overlapping meta-analyses. Arthroscopy. 2015;31(11):2213-2221.

24.  Meheux CJ, McCulloch PC, Lintner DM, Varner KE, Harris JD. Efficacy of intra-articular platelet-rich plasma injections in knee osteoarthritis: A systematic review. Arthroscopy. 2016;32(3):495-505.

25.  Smith PA. Intra-articular autologous conditioned plasma injections provide safe and efficacious treatment for knee osteoarthritis: An FDA-sanctioned, randomized, double-blind, placebo-controlled clinical trial. Am J Sports Med. 2016;44(4):884-891.

26.   Podesta L, Crow SA, Volkmer D, Bert T, Yocum LA. Treatment of partial ulnar collateral ligament tears in the elbow with platelet-rich plasma. Am J Sports Med. 2013;41(7):1689-1694.

27.  Herquelot E, Gueguen A, Roquelaure Y, et al. Work-related risk factors for incidence of lateral epicondylitis in a large working population. Scand J Work Environ Health. 2013;39(6):578-588.

28.  Titchener AG, Fakis A, Tambe AA, Smith C, Hubbard RB, Clark DI. Risk factors in lateral epicondylitis (tennis elbow): a case-control study. J Hand Surg Eur Vol. 2013;38(2):159-164.

29.  Gruchow HW, Pelletier D. An epidemiologic study of tennis elbow. Incidence, recurrence, and effectiveness of prevention strategies. Am J Sports Med. 1979;7(4):234-238.

30.  Sanders TL Jr, Maradit Kremers H, Bryan AJ, Ransom JE, Smith J, Morrey BF.  The epidemiology and health care burden of tennis elbow: a population-based study. Am J Sports Med. 2015;43(5):1066-1071.

31.  Coonrad RW, Hooper WR. Tennis elbow: its course, natural history, conservative and surgical management. J Bone Joint Surg Am. 1973;55(6):1177-1182.

32.  Taylor SA, Hannafin JA. Evaluation and management of elbow tendinopathy. Sports Health. 2012;4(5):384-393.

33.  Sims SE, Miller K, Elfar JC, Hammert WC. Non-surgical treatment of lateral epicondylitis: a systematic review of randomized controlled trials. Hand (NY). 2014;9(4):419-446.

34.  Brummel J, Baker CL 3rd, Hopkins R, Baker CL Jr. Epicondylitis: lateral. Sports Med Arthrosc. 2014;22(3):e1-e6.

35.  de Vos RJ, Windt J, Weir A. Strong evidence against platelet-rich plasma injections for chronic lateral epicondylar tendinopathy: a systematic review. Br J Sports Med. 2014;48(12):952-956.

36.  Ahmad Z, Brooks R, Kang SN, et al. The effect of platelet-rich plasma on clinical outcomes in lateral epicondylitis. Arthroscopy. 2013;29(11):1851-1862.

37.  Arirachakaran A, Sukthuayat A, Sisayanarane T, Laoratanavoraphong S, Kanchanatawan W, Kongtharvonskul J. Platelet-rich plasma versus autologous blood versus steroid injection in lateral epicondylitis: systematic review and network meta-analysis. J Orthop Traumatol. 2016;17(2):101-112.

38.  Chaudhury S, de La Lama M, Adler RS, et al. Platelet-rich plasma for the treatment of lateral epicondylitis: sonographic assessment of tendon morphology and vascularity (pilot study). Skeletal Radiol. 2013;42(1):91-97.

39.  Brkljac M, Kumar S, Kalloo D, Hirehal K. The effect of platelet-rich plasma injection on lateral epicondylitis following failed conservative management. J Orthop. 2015;12(Suppl 2):S166-S170.

40.  Yadav R, Kothari SY, Borah D. Comparison of local injection of platelet rich plasma and corticosteroids in the treatment of lateral epicondylitis of humerus. J Clin Diagn Res. 2015;9(7):RC05-RC07.

41.  Gautam VK, Verma S, Batra S, Bhatnagar N, Arora S. Platelet-rich plasma versus corticosteroid injection for recalcitrant lateral epicondylitis: clinical and ultrasonographic evaluation. J Orthop Surg (Hong Kong). 2015;23(1):1-5.

42.  Krogh TP, Fredberg U, Stengaard-Pedersen K, Christensen R, Jensen P, Ellingsen T. Treatment of lateral epicondylitis with platelet-rich plasma, glucocorticoid, or saline: a randomized, double-blind, placebo-controlled trial. Am J Sports Med. 2013;41(3):625-635.

43.  Behera P, Dhillon M, Aggarwal S, Marwaha N, Prakash M. Leukocyte-poor platelet-rich plasma versus bupivacaine for recalcitrant lateral epicondylar tendinopathy. J Orthop Surg (Hong Kong). 2015;23(1):6-10.

44.  Mishra AK, Skrepnik NV, Edwards SG, et al. Efficacy of platelet-rich plasma for chronic tennis elbow: a double-blind, prospective, multicenter, randomized controlled trial of 230 patients. Am J Sports Med. 2014;42(2):463-471.

45.  van der Horst N, van de Hoef S, Reurink G, Huisstede B, Backx F. Return to play after hamstring injuries: a qualitative systematic review of definitions and criteria. Sports Med. 2016;46(6):899-912.

46.  Crema MD, Guermazi A, Tol JL, Niu J, Hamilton B, Roemer FW. Acute hamstring injury in football players: Association between anatomical location and extent of injury-A large single-center MRI report. J Sci Med Sport. 2016;19(4):317-322.

47.  Ekstrand J, Lee JC, Healy JC. MRI findings and return to play in football: a prospective analysis of 255 hamstring injuries in the UEFA Elite Club Injury Study. Br J Sports Med. 2016;50(12):738-743.

48.  Wetzel RJ, Patel RM, Terry MA. Platelet-rich plasma as an effective treatment for proximal hamstring injuries. Orthopedics. 2013;36(1):e64-e70.

49.  Hamid A, Mohamed Ali MR, Yusof A, George J, Lee LP. Platelet-rich plasma injections for the treatment of hamstring injuries: a randomized controlled trial. Am J Sports Med. 2014;42(10):2410-2418.

50.  Mejia HA, Bradley JP.  The effects of platelet-rich plasma on muscle: basic science and clinical application. Operative Techniques in Sports Medicine. 2011;19(3):149-153.

51.  Guillodo Y, Madouas G, Simon T, Le Dauphin H, Saraux A. Platelet-rich plasma (PRP) treatment of sports-related severe acute hamstring injuries. Muscles Ligaments Tendons J. 2015;5(4):284-288.

52.  Rettig AC, Meyer S, Bhadra AK. Platelet-rich plasma in addition to rehabilitation for acute hamstring injuries in NFL players: Clinical effects and time to return to play. Orthop J Sports Med. 2013;1(1):2325967113494354.

53.   Hamilton B, Tol JL, Almusa E, et al. Platelet-rich plasma does not enhance return to play in hamstring injuries: a randomised controlled trial. Br J Sports Med. 2015;49(14):943-950.

54.  Reurink G, Goudswaard GJ, Moen MH, et al. Rationale, secondary outcome scores and 1-year follow-up of a randomised trial of platelet-rich plasma injections in acute hamstring muscle injury: the Dutch Hamstring Injection Therapy study. Br J Sports Med. 2015;49(18):1206-1212.

55.  Kujala UM, Sarna S, Kaprio J. Cumulative incidence of achilles tendon rupture and tendinopathy in male former elite athletes. Clin J Sport Med. 2005;15(3):133-135.

56.  Alfredson H. Clinical commentary of the evolution of the treatment for chronic painful mid-portion Achilles tendinopathy. Braz J Phys Ther. 2015;19(5):429-432.

57.  Kearney RS, Parsons N, Costa ML. Achilles tendinopathy management: A pilot randomised controlled trial comparing platelet-rich plasma injection with an eccentric loading programme. Bone Joint Res. 2013;2(10):227-232.

58.  de Vos RJ, Weir A, Tol JL, Verhaar JA, Weinans H, van Schie HT. No effects of PRP on ultrasonographic tendon structure and neovascularisation in chronic midportion Achilles tendinopathy. Br J Sports Med. 2011;45(5):387-392.

59.  de Vos RJ, Weir A, van Schie HT, et al. Platelet-rich plasma injection for chronic Achilles tendinopathy: a randomized controlled trial. JAMA. 2010;303(2):144-149.

Issue
The American Journal of Orthopedics - 45(5)
Issue
The American Journal of Orthopedics - 45(5)
Page Number
290-294, 326
Page Number
290-294, 326
Publications
Publications
Topics
Article Type
Display Headline
Platelet-Rich Plasma (PRP) in Orthopedic Sports Medicine
Display Headline
Platelet-Rich Plasma (PRP) in Orthopedic Sports Medicine
Legacy Keywords
platelet-rich plasma, PRP, biologics, sports medicine, review, injections, mlynarek, kuhn, bedi
Legacy Keywords
platelet-rich plasma, PRP, biologics, sports medicine, review, injections, mlynarek, kuhn, bedi
Sections
Article Source

PURLs Copyright

Inside the Article

Article PDF Media

Stem Cells in Orthopedics: A Comprehensive Guide for the General Orthopedist

Article Type
Changed
Thu, 09/19/2019 - 13:25
Display Headline
Stem Cells in Orthopedics: A Comprehensive Guide for the General Orthopedist

Biologic use in orthopedics is a continuously evolving field that complements technical, anatomic, and biomechanical advancements in orthopedics. Biologic agents are receiving increasing attention for their use in augmenting healing of muscles, tendons, ligaments, and osseous structures. As biologic augmentation strategies become increasingly utilized in bony and soft-tissue injuries, research on stem cell use in orthopedics continues to increase. Stem cell-based therapies for the repair or regeneration of muscle and tendon represent a promising technology going forward for numerous diseases.1

Stem cells by definition are undifferentiated cells that have 4 main characteristics: (1) mobilization during angiogenesis, (2) differentiation into specialized cell types, (3) proliferation and regeneration, and (4) release of immune regulators and growth factors.2 Mesenchymal stem cells (MSCs) have garnered the most attention in the field of surgery due to their ability to differentiate into the tissues of interest for the surgeon.3 This includes both bone marrow-derived mesenchymal stem cells (bm-MSCs) and adipose-derived mesenchymal stem cells (a-MSCs). These multipotent stem cells in adults originate from mesenchymal tissues, including bone marrow, tendon, adipose, and muscle tissue.4 They are attractive for clinical use because of their multipotent potential and relative ease of growth in culture.5 They also exert a paracrine effect to modulate and control inflammation, stimulate endogenous cell repair and proliferation, inhibit apoptosis, and improve blood flow through secretion of chemokines, cytokines, and growth factors.6,7

Questions exist regarding the best way to administer stem cells, whether systematic administration is possible for these cells to localize to the tissue in need, or more likely if direct application to the pathologic area is necessary.8,9 A number of sources, purification process, and modes of delivery are available, but the most effective means of preparation and administration are still under investigation. The goal of this review is to illustrate the current state of knowledge surrounding stem cell therapy in orthopedics with a focus on osteoarthritis, tendinopathy, articular cartilage, and enhancement of surgical procedures.

Important Considerations

Common stem cell isolates include embryonic, induced pluripotent, and mesenchymal formulations (Table 1). MSCs can be obtained from multiple sites, including but not limited to the adult bone marrow, adipose, muscular, or tendinous tissues, and their use has been highlighted in the study of numerous orthopedic and nonorthopedic pathologies over the course of the last decade. Research on the use of embryonic stem cells in medical therapy with human implications has received substantial attention, with many ethical concerns by those opposed, and the existence of a potential risk of malignant alterations.8,10 Amniotic-derived stem cells can be isolated from amniotic fluid, umbilical cord blood, or the placenta and thus do not harbor the same social constraints as the aforementioned embryonic cells; however, they do not harbor the same magnitude of multi-differentiation potential, either.4

Adult MSCs are more locally available and easy to obtain for treatment when compared with embryonic and fetal stem cells, and the former has a lower immunogenicity, which allows allogeneic use.11 Safety has been preliminarily demonstrated in use thus far; Centeno and colleagues12 found no neoplastic tissue generation at the site of stem cell injection after 3 years postinjection for a cohort of patients who were treated with autologous bm-MSCs for various pathologies. Self-limited pain and swelling are the most commonly reported adverse events after use.13 However, long-term data are lacking in many instances to definitively suggest the absence of possible complications.

Basic Science

Stem cell research encompasses a wide range of rapidly developing treatment strategies that are applicable to virtually every field of medicine. In general, stem cells can be classified as embryonic stem cells (ESCs), induced pluripotent stem (iPS) cells, or adult-derived MSCs. ESCs are embryonic cells derived typically from fetal tissue, whereas iPS cells are dedifferentiated from adult tissue, thus avoiding many of the ethical and legal challenges imposed by research with ESCs. However, oncogenic and lingering politico-legal concerns with introducing dedifferentiated ESCs or iPS cells into healthy tissue necessitate the development, isolation, and expansion of multi- but not pluripotent stem cell lines.14 To date, the most advantageous and widely utilized from any perspective are MSCs, which can further differentiate into cartilage, tendon, muscle, and bony tissue.7,15,16

MSCs are defined by their ability to demonstrate in vitro differentiation into osteoblasts, adipocytes, or chondroblasts, adhere to plastic, express CD105, CD73, and CD90, and not express CD43, CD23, CD14 or CD11b, CD79 or CD19, or HLA-DR.17 Porada and Almeida-Porada18 have outlined 6 reasons highlighting the advantages of MSCs: 1) ease of isolation, 2) high differentiation capabilities, 3) strong colony expansion without differentiation loss, 4) immunosuppression following transplantation, 5) powerful anti-inflammatory properties, and 6) their ability to localize to damaged tissue. The anti-inflammatory properties of MSCs are particularly important as they promote allo- and xenotransplantation from donor tissues.19,20 MSCs can be isolated from numerous sources, including but not limited to bone marrow, periosteum, adipocyte, and muscle.21-23 Interestingly, the source tissue used to isolate MSCs can affect differentiation capabilities, colony size, and growth rate (Table 2).24 Advantages of a-MSCs include high prevalence and ease of harvest; however, several animal studies have shown inferior results when compared to bm-MSCs.25-27 More research is needed to determine the ideal source material for MSCs, which will likely depend in part on the procedure for which they are employed.27

 

 

Following harvesting, isolation, and expansion, MSC delivery methods for treatments typically consist of either cell-based or tissue engineering approaches. Cell-based techniques involve the injection of MSCs into damaged tissues. Purely cell-based therapy has shown success in limited clinical trials involving knee osteoarthritis, cartilage repair, and meniscal repair.28-30 However, additional studies with longer follow-up are required to validate these preliminary findings. Tissue engineering approaches involve the construction of a 3-dimensional scaffold seeded with MSCs that is later surgically implanted. While promising in theory, limited and often conflicting data exist regarding the efficacy of tissue-engineered MSC implantation.31-32 Suboptimal scaffold vascularity is a major limitation to scaffold design, which may be alleviated in part with the advent of 3-dimensional printing and the ability to more precisely alter scaffold architecture.14,33 Additional limitations include ensuring MSC purity and differentiation potential following harvesting and expansion. At present, the use of tissue engineering with MSCs is promising but it remains a nascent technology with additional preclinical studies required to confirm implant efficacy and safety.

Clinical Entities

Osteoarthritis

MSC therapies have emerged as promising treatment strategies in the setting of early osteoarthritis (OA). In addition to their regenerative potential, MSCs demonstrate potent anti-inflammatory properties, increasing their attractiveness as biologic agents in the setting of OA.34 Over the past decade, multiple human trials have been published demonstrating the efficacy of MSC injections into patients with OA.35,36 In a study evaluating a-MSC injection into elderly patients (age >65 years) with knee OA, Koh and colleagues29 found that 88% demonstrated improved cartilage status at 2-year follow-up, while no patient underwent a total knee arthroplasty during this time period. In another study investigating patients with unicompartmental knee OA with varus alignment undergoing high tibial osteotomy and microfracture, Wong and colleagues37 reported improved clinical, patient-reported, and magnetic resonance imaging (MRI)-based outcomes in a group receiving a preoperative MSC injection compared to a control group. Further, in a recent randomized control trial of patients with knee osteoarthritis, Vega and colleagues38 reported improved cartilage and quality of life outcomes at 1 year following MSC injection compared to a control group receiving a hyaluronic acid injection. In addition to knee OA, studies have also reported improvement in ankle OA following MSC injection.39 While promising, many of the preliminary clinical studies evaluating the efficacy of MSC therapies in the treatment of OA are hindered by small patient populations and short-term follow-up. Additional large-scale, randomized studies are required and many are ongoing presently in hopes of validating these preliminary findings.36

Tendinopathy

The quality of repaired tissue in primary tendon-to-tendon and tendon-to-bone healing has long been a topic of great interest.40 The healing potential of tendons is inferior to that of other bony and connective tissues,41 with tendon healing typically resulting in a biomechanically and histologically inferior structure to the native tissue.42 As such, this has been a particularly salient opportunity for stem cell use with hopes of recapitulating a more normal tendon or tendon enthesis following injury. In addition to the acute injury, there is great interest in the application of stem cells to chronic states of injury such as tendinopathy.

In equine models, the effect of autologous bm-MSCs treatment on tendinopathy of the superficial digital flexor tendon has been studied. Godwin and colleagues43 evaluated 141 race horses with spontaneous superficial digital flexor tendinopathy treated in this manner, and reported a reinjury percentage in these treated horses of just 27.4%, which compared favorably to historical controls and alternative therapeutics. Machova Urdzikova and colleagues44 injected MSCs at Achilles tendinopathy locations to augment nonoperative healing in 40 rats, and identified more native histological organization and improved vascularization in comparison to control rat specimens. Oshita and colleagues45 reported histologic improvement of tendinopathy findings in 8 rats receiving a-MSCs at the location of induced Achilles tendinopathy that was significantly superior to a control cohort. Bm-MSCs were used by Yuksel and colleagues46 in comparison with platelet-rich plasma (PRP) for treatment of Achilles tendon ruptures created surgically in rat models. They demonstrated successful effects with its use in terms of recovery for the tendon’s histopathologic, immunohistochemical, and biomechanical properties, related to significantly greater levels of anti-inflammatory cytokines. However, these aforementioned findings have not been uniform across the literature—other authors have reported findings that MSC transplantation alone did not repair Achilles tendon injury with such high levels of success.47

Human treatment of tendinopathies with stem cells has been scarcely studied to date. Pascual-Garrido and colleagues48 evaluated 8 patients with refractory patellar tendinopathy treated with injection of autologous bm-MSCs and reported successful results at 2- to 5-year follow-up, with significant improvements in patient-reported outcome measures for 100% of patients. Seven of 8 (87.5%) noted that they would undergo the procedure again.

 

 

Articular Cartilage Injury

Chondral injury is a particularly important subject given the limited potential of chondrocytes to replicate or migrate to the site of pathology.49 Stem cell use in this setting assists with programmed growth factor release and alteration of the anatomic microenvironment to facilitate regeneration and repair of the chondral surface. Autologous stem cell use through microfracture provides a perforation into the bone marrow and a subsequent fibrin clot formation containing platelets, growth factors, vascular elements, and MSCs.50 A similar concept to PRP is currently being explored with bm-MSCs. Isolated bm-MSCs are commonly referred to as bone marrow aspirate or bone marrow aspirate concentrate (BMAC). Commercially available systems are now available to aid in the harvesting and implementation of BMAC. One of the more promising avenues for BMAC implementation is in articular cartilage repair or regeneration due to chondrogenic potential of BMAC when used in isolation or when combined with microfracture, chondrocyte transfer, or collagen scaffolds.19,51 Synovial-derived stem cells as an additional source for stem cell use has demonstrated excellent chondrogenic potential in animal studies with full-thickness lesion healing and native-appearing cartilage histologically.52 Incorporation of a-MSCs into scaffolds for surgical implantation has demonstrated success in repairing full-thickness chondral defects with continuous joint surface and extracellular proteins, surface markers, and gene products similar to the native cartilage in animal models.53,54 In light of the promising basic science and animal studies, clinical studies have begun to emerge.55-57

Fortier and colleagues58 found MRI and histologic evidence of full-thickness chondral repair and increased integration with neighboring cartilage when BMAC was concurrently used at the time of microfracture in an equine model. Fortier and colleagues58 also demonstrated greater healing in equine models with acute full-thickness cartilage defects treated by microfracture with MSCs than without delivery of MSCs. Kim and colleagues59,60 similarly reported superiority in clinical outcomes for patients with osteochondral lesions of the talus treated with marrow stimulation and MSC injection than by the former in isolation.

In humans, stem cell use for chondral repair has additionally proven promising. A systematic review of the literature suggested good to excellent overall outcomes for the treatment of moderate focal chondral defects with BMAC with or without scaffolds and microfracture with inclusion of 8 total publications.61 This review included Gobbi and colleagues,62 who prospectively treated 15 patients with a mean focal chondral defect size of 9.2 cm2 about the knee. Use of BMAC covered with a collagen I/III matrix produced significant improvements in patient-reported outcome scores and MRI demonstrated complete hyaline-like cartilage coverage in 80%, with second-look arthroscopy demonstrating normal to nearly normal tissue. Gobbi and colleagues55 also found evidence for superiority of chondral defects treated with BMAC compared to matrix-induced autologous chondrocyte implantation (MACI) for patellofemoral lesions in 37 patients (MRI showed complete filling of defects in 81% of BMAC-treated patients vs 76% of MACI-treated patients).

Meniscal Repair

Clinical application of MSCs in the treatment of meniscal pathology is evolving as well. ASCs have been added to modify the biomechanical environment of avascular zone meniscal tears at the time of suture repair in a rabbit, and have demonstrated increased healing rates in small and larger lesions, although the effect lessens with delay in repair.63 Angele and colleagues64 treated meniscal defects in a rabbit model with scaffolds with bm-MSCs compared with empty scaffolds or control cohorts and found a higher proportion of menisci with healed meniscus-like fibrocartilage when MSCs were utilized.

In humans, Vangsness and colleagues30 treated knees with partial medial meniscectomy with allogeneic stem cells and reported an increase in meniscal volume and decrease in pain in those patients when compared to a cohort of knees treated with hyaluronic acid. Despite promising early results, additional clinical studies are necessary to determine the external validity and broad applicability of stem cell use in meniscal repair.

Rotator Cuff Repair

The number of local resident stem cells at the site of rotator cuff tear has been shown to decrease with tear size, chronicity, and degree of fatty infiltration, suggesting that those with the greatest need for a good reparative environment are those least equipped to heal.65 The need for improvement in this domain is related to the still relatively high re-tear rate after rotator cuff repair despite improvements in instrumentation and surgical technique.66 The native fibrocartilaginous transition zone between the humerus and the rotator cuff becomes a fibrovascular scar tissue after rupture and repair with poorer material properties than the native tissue.67 Thus, a-MSCs have been evaluated in this setting to determine if the biomechanical and histological properties of the repair may improve.68

 

 

In rat models, Valencia Mora and colleagues68 reported on the application of a-MSCs in a rat rotator cuff repair model compared to an untreated group. They found no differences between those treated rats and those without a-MSCs use in terms of biomechanical properties of the tendon-to-bone healing, but those with stem cell use had less inflammation shown histologically (diminished presence of edema and neutrophils) at 2- and 4-week time points, which the authors suggested may lead to a more elastic repair and less scar at the bone-tendon healing site. Oh and colleagues1 evaluated the use of a-MSCs in a rabbit subscapularis tear model, and reported significantly reduced fatty infiltration at the site of chronic rotator cuff tear after repair with its application at the repair site; while the load-to-failure was higher in those rabbits with ASCs administration, it was short of reaching statistical significance. Yokoya and colleagues69 demonstrated regeneration of rotator cuff tendon-to-bone insertional site anatomy and in the belly of the cuff tendon in a rabbit model with MSCs applied at the operative site. However, Gulotta and colleagues70 did not see the same improvement in their similar study in the rat model; these authors failed to see improvement in structure, strength, or composition of the tendinous attachment site despite addition of MSCs.

Clinical studies on augmented rotator cuff repair have also found mixed results. MSCs for this purpose have been cultivated from arthroscopic bone marrow aspiration of the proximal humerus71 and subacromial bursa72 with successful and reproducibly high concentrations of stem cells. Hernigou and colleagues73 found a significant improvement in rate of healing (87% intact cuffs vs 44% in the control group) and repair surface tendon integrity (via ultrasound and MRI) for patients at a minimum of 10 years after rotator cuff repair with MSC injection at the time of surgery. The authors found a direct correlation in these outcomes with the number of MSCs injected at the time of repair. Ellera Gomes and colleagues74 injected bm-MSCs obtained from the iliac crest into the tendinous repair site in 14 consecutive patients with full-thickness rotator cuff tears treated by transosseous sutures via a mini-open approach. MRI demonstrated integrity of the repair site in all patients at more than 1-year follow-up.

Achilles Tendon Repair

The goal with stem cell use in Achilles repair is to accelerate the healing and rehabilitation. Several animal studies have demonstrated improved mechanical properties and collagen composition of tendon repairs augmented with stem cells, including Achilles tendon repair in a rat model. Adams and colleagues75 compared suture alone (36 tendons) to suture plus stem cell concentrate injection (36 tendons) and stem cell loaded suture (36 tendons) in Achilles tendon repair with rat models. The suture-alone cohort had lower ultimate failure loads at 14 days after surgery, indicating biomechanical superiority with stem cell augmentation means. Transplantation of hypoxic MSCs at the time of Achilles tendon repair may be a promising option for superior biomechanical failure loads and histologic findings as per recent rat model findings by Huang and colleagues.76 Yao and colleagues77 demonstrated increased strength of suture repair for Achilles repair in rat models at early time points when using MSC-coated suture in comparison to standard suture, and suggested that the addition of stem cells may improve early mechanical properties during the tendon repair process. A-MSC addition to PRP has provided significantly increased tensile strength to rabbit models with Achilles tendon repair as well.78

In evaluation of stem cell use for this purpose with humans, Stein and colleagues79 reviewed 28 sports-related Achilles tendon ruptures in 27 patients treated with open repair and BMAC injection. At a mean follow-up of 29.7 months, the authors reported no re-ruptures, with 92% return to sport at 5.9 months, and excellent clinical outcomes. This small cohort study found no adverse outcomes related to the BMAC addition, and thus proposed further study of the efficacy of stem cell treatment for Achilles tendon repair.

Anterior Cruciate Ligament Reconstruction

Bm-MSCs genetically modified with bone morphogenetic protein 2 (BMP2) and basic fibroblast growth factor (bFGF) have shown great promise in improvement of the formation of mechanically sound tendon-bone interface in anterior cruciate ligament (ACL) reconstruction.80 Similar to the other surgical procedures mentioned in this review, animal studies have successfully evaluated the augmentation of osteointegration of tendon to bone in the setting of ACL reconstruction. Jang and colleagues3 investigated the use of nonautologous transplantation of human umbilical cord blood-derived MSCs in a rabbit ACL reconstruction model. The authors demonstrated a lack of immune rejection, and enhanced tendon-bone healing with broad fibrocartilage formation at the transition zone (similar to the native ACL) and decreased femoral and tibial tunnel widening as compared to a control cohort at 12-weeks after surgery. In a rat model, Kanaya and colleagues81 reported improved histological scores and slight improvements in biomechanical integrity of partially transected rat ACLs treated with intra-articular MSC injection. Stem cell use in the form of suture-supporting scaffolds seeded with MSCs has been evaluated in a total ACL transection rabbit model; the authors of this report demonstrated total ACL regeneration in one-third of samples treated with this augmentation option, in comparison to complete failure in all suture and scaffold alone groups.82

 

 

The use of autologous MSCs in ACL healing remains limited to preclinical research and small case series of patients. One human trial by Silva and colleagues83 evaluated the graft-to-bone site of healing in ACL reconstruction for 20 patients who received an intraoperative infiltration of their graft with adult bm-MSCs. MRI and histologic analysis showed no difference in comparison to control groups, but the authors’ conclusion proposed that the number of stem cells injected might have been too minimal to show a clinical effect.

Other Applications

Although outside the scope of this article, stem cells have demonstrated efficacy in the treatment of a number of osseous clinical entities. This includes the treatment of fracture nonunion, augmentation of spinal fusion, and assistance in the treatment of osteonecrosis.84

Summary

As a scientific community, our understanding of the use of stem cells, their nuances, and their indications has expanded dramatically over the last several years. Stem cell treatment has particularly infiltrated the world of operative and nonoperative sports medicine, given in part the active patient population seeking greater levels of improvement.85 Stem cell therapy offers a potentially effective therapy for a multitude of pathologies because of these cells’ anti-inflammatory, immunoregulatory, angiogenic, and paracrine effects.86 It thus remains a very dynamic option in the study of musculoskeletal tissue regeneration. While the potential exists for stem cell use in daily surgery practices, it is still premature to predict whether this can be expected.

The ideal stem cell sources (including allogeneic or autologous), preparation, cell number, timing, and means of application continue to be evaluated, as well as those advantageous pathologies that can benefit from the technology. In order to better answer these pertinent questions, we need to make sure we have a safe, economic, and ethically acceptable means for stem cell translational research efforts. More high-level studies with standardized protocols need to be performed. It is necessary to improve national and international collaboration in research, as well as collaboration with governing bodies, to attempt to further scientific advancement in this field of research.49 Further study on embryonic stem cell use may be valuable as well, pending governmental approval. Finally, more dedicated research efforts must be placed on the utility of adjuncts with stem cell use, including PRP and scaffolds, which may increase protection, nutritional support, and mechanical stimulation of the administered stem cells.

References

1.    Oh JH, Chung SW, Kim SH, Chung JY, Kim JY. 2013 Neer Award: Effect of the adipose-derived stem cell for the improvement of fatty degeneration and rotator cuff healing in rabbit model. J Shoulder Elb Surg. 2014;23(4):445-455.

2.    Caplan AI, Correa D. PDGF in bone formation and regeneration: new insights into a novel mechanism involving MSCs. J Orthop Res. 2011;29(12):1795-1803.

3.    Jang KM, Lim HC, Jung WY, Moon SW, Wang JH. Efficacy and safety of human umbilical cord blood-derived mesenchymal stem cells in anterior cruciate ligament reconstruction of a rabbit model: new strategy to enhance tendon graft healing. Arthroscopy. 2015;31(8):1530-1539.

4.    Muttini A, Salini V, Valbonetti L, Abate M. Stem cell therapy of tendinopathies: suggestions from veterinary medicine. Muscles Ligaments Tendons J. 2012;2(3):187-192.

5.    Xia P, Wang X, Lin Q, Li X. Efficacy of mesenchymal stem cells injection for the management of knee osteoarthritis: a systematic review and meta-analysis. Int Orthop. 2015;39(12):2363-2372.

6.    Veronesi F, Giavaresi G, Tschon M, Borsari V, Nicoli Aldini N, Fini M. Clinical use of bone marrow, bone marrow concentrate, and expanded bone marrow mesenchymal stem cells in cartilage disease. Stem Cells Dev. 2013;22(2):181-192.

7.    Caplan AI. Review: mesenchymal stem cells: cell-based reconstructive therapy in orthopedics. Tissue Eng. 2005;11(7-8):1198-1211.

8.    Hirzinger C, Tauber M, Korntner S, et al. ACL injuries and stem cell therapy. Arch Orthop Trauma Surg. 2014;134(11):1573-1578.

9.    Becerra P, Valdés Vázquez MA, Dudhia J, et al. Distribution of injected technetium(99m)-labeled mesenchymal stem cells in horses with naturally occurring tendinopathy. J Orthop Res. 2013;31(7):1096-1102.

10.  Lodi D, Iannitti T, Palmieri B. Stem cells in clinical practice: applications and warnings. J Exp Clin Cancer Res. 2011;30:9.

11.  García-Gómez I, Elvira G, Zapata AG, et al. Mesenchymal stem cells: biological properties and clinical applications. Expert Opin Biol Ther. 2010;10(10):1453-1468.

12.  Centeno CJ, Schultz JR, Cheever M, et al. Safety and complications reporting update on the re-implantation of culture-expanded mesenchymal stem cells using autologous platelet lysate technique. Curr Stem Cell Res Ther. 2011;6(4):368-378.

13.  Centeno CJ, Al-Sayegh H, Freeman MD, Smith J, Murrell WD, Bubnov R. A multi-center analysis of adverse events among two thousand, three hundred and seventy two adult patients undergoing adult autologous stem cell therapy for orthopaedic conditions. Int Orthop. 2016 Mar 30. [Epub ahead of print]

14.  Schmitt A, van Griensven M, Imhoff AB, Buchmann S. Application of stem cells in orthopedics. Stem Cells Int. 2012;2012:394962.

15.  Tuan RS, Boland G, Tuli R. Adult mesenchymal stem cells and cell-based tissue engineering. Arthritis Res Ther. 2003;5(1):32-45.

16.  Anz AW, Hackel JG, Nilssen EC, Andrews JR. Application of biologics in the treatment of the rotator cuff, meniscus, cartilage, and osteoarthritis. J Am Acad Orthop Surg. 2014;22(2):68-79.

17.  Dominici M, Le Blanc K, Mueller I, et al. Minimal criteria for defining multipotent mesenchymal stromal cells. The International Society for Cellular Therapy position statement. Cytotherapy. 2006;8(4):315-317.

18.  Porada CD, Almeida-Porada G. Mesenchymal stem cells as therapeutics and vehicles for gene and drug delivery. Adv Drug Deliv Rev. 2010;62(12):1156-1566.

19.  Filardo G, Madry H, Jelic M, Roffi A, Cucchiarini M, Kon E. Mesenchymal stem cells for the treatment of cartilage lesions: from preclinical findings to clinical application in orthopaedics. Knee Surg Sports Traumatol Arthrosc. 2013;21(8):1717-1729.

20.  Liechty KW, MacKenzie TC, Shaaban AF, et al. Human mesenchymal stem cells engraft and demonstrate site-specific differentiation after in utero transplantation in sheep. Nat Med. 2000;6(11):1282-1286.

21.  Hung SC, Chen NJ, Hsieh SL, Li H, Ma HL, Lo WH. Isolation and characterization of size-sieved stem cells from human bone marrow. Stem Cells. 2002;20(3):249-258.

22.  De Bari C, Dell’Accio F, Vanlauwe J, et al. Mesenchymal multipotency of adult human periosteal cells demonstrated by single-cell lineage analysis. Arthritis Rheum. 2006;54(4):1209-1221.

23.  Zuk PA, Zhu M, Ashjian P, et al. Human adipose tissue is a source of multipotent stem cells. Mol Biol Cell. 2002;13(12):4279-4295.

24.  Mafi R, Hindocha S, Mafi P, Griffin M, Khan WS. Sources of adult mesenchymal stem cells applicable for musculoskeletal applications - a systematic review of the literature. Open Orthop J. 2011;5 Suppl 2:242-248.

25.  Frisbie DD, Kisiday JD, Kawcak CE, Werpy NM, McIlwraith CW. Evaluation of adipose-derived stromal vascular fraction or bone marrow-derived mesenchymal stem cells for treatment of osteoarthritis. J Orthop Res. 2009;27(12):1675-1680.

26.  Vidal MA, Robinson SO, Lopez MJ, et al. Comparison of chondrogenic potential in equine mesenchymal stromal cells derived from adipose tissue and bone marrow. Vet Surg. 2008;37(8):713-724.

27.  Yoshimura H, Muneta T, Nimura A, Yokoyama A, Koga H, Sekiya I. Comparison of rat mesenchymal stem cells derived from bone marrow, synovium, periosteum, adipose tissue, and muscle. Cell Tissue Res. 2007;327(3):449-462.

28.  Hogan MV, Walker GN, Cui LR, Fu FH, Huard J. The role of stem cells and tissue engineering in orthopaedic sports medicine: current evidence and future directions. Arthroscopy. 2015;31(5):1017-1021.

29.  Koh YG, Choi YJ, Kwon SK, Kim YS, Yeo JE. Clinical results and second-look arthroscopic findings after treatment with adipose-derived stem cells for knee osteoarthritis. Knee Surg Sports Traumatol Arthrosc. 2015;23(5):1308-1316.

30.  Vangsness CT Jr, Farr J 2nd, Boyd J, Dellaero DT, Mills CR, LeRoux-Williams M. Adult human mesenchymal stem cells delivered via intra-articular injection to the knee following partial medial meniscectomy: a randomized, double-blind, controlled study. J Bone Joint Surg Am. 2014;96(2):90-98.

31.  Goodrich LR, Chen AC, Werpy NM, et al. Addition of mesenchymal stem cells to autologous platelet-enhanced fibrin scaffolds in chondral defects: does it enhance repair? J Bone Joint Surg Am. 2016;98(1):23-34.

32.  Kim YS, Choi YJ, Suh DS, et al. Mesenchymal stem cell implantation in osteoarthritic knees: is fibrin glue effective as a scaffold? Am J Sports Med. 2015;43(1):176-185.

33.  Steinert AF, Rackwitz L, Gilbert F, Nöth U, Tuan RS. Concise review: the clinical application of mesenchymal stem cells for musculoskeletal regeneration: current status and perspectives. Stem Cells Transl Med. 2012;1(3):237-247.

34.  Pers YM, Ruiz M, Noël D, Jorgensen C. Mesenchymal stem cells for the management of inflammation in osteoarthritis: state of the art and perspectives. Osteoarthritis Cartilage. 2015;23(11):2027-2035.

35.  Mamidi MK, Das AK, Zakaria Z, Bhonde R. Mesenchymal stromal cells for cartilage repair in osteoarthritis. Osteoarthritis Cartilage. 2016 Mar 10. [Epub ahead of print]

36.  Wyles CC, Houdek MT, Behfar A, Sierra RJ. Mesenchymal stem cell therapy for osteoarthritis: current perspectives. Stem Cells Cloning. 2015;8:117-124.

37.  Wong KL, Lee KB, Tai BC, Law P, Lee EH, Hui JH. Injectable cultured bone marrow-derived mesenchymal stem cells in varus knees with cartilage defects undergoing high tibial osteotomy: a prospective, randomized controlled clinical trial with 2 years’ follow-up. Arthroscopy. 2013;29(12):2020-2028.

38.  Vega A, Martín-Ferrero MA, Del Canto F, et al. Treatment of knee osteoarthritis with allogeneic bone marrow mesenchymal stem cells: a randomized controlled trial. Transplantation. 2015;99(8):1681-1690.

39.  Kim YS, Lee M, Koh YG. Additional mesenchymal stem cell injection improves the outcomes of marrow stimulation combined with supramalleolar osteotomy in varus ankle osteoarthritis: short-term clinical results with second-look arthroscopic evaluation. J Exp Orthop. 2016;3(1):12.

40.  Kraus TM, Imhoff FB, Reinert J, et al. Stem cells and bFGF in tendon healing: Effects of lentiviral gene transfer and long-term follow-up in a rat Achilles tendon defect model. BMC Musculoskelet Disord. 2016;17(1):148.

41.  Thomopoulos S, Parks WC, Rifkin DB, Derwin KA. Mechanisms of tendon injury and repair. J Orthop Res. 2015;33(6):832-839.

42.  Müller SA, Todorov A, Heisterbach PE, Martin I, Majewski M. Tendon healing: an overview of physiology, biology, and pathology of tendon healing and systematic review of state of the art in tendon bioengineering. Knee Surg Sports Traumatol Arthrosc. 2015;23(7):2097-3105.

43.  Godwin EE, Young NJ, Dudhia J, Beamish IC, Smith RK. Implantation of bone marrow-derived mesenchymal stem cells demonstrates improved outcome in horses with overstrain injury of the superficial digital flexor tendon. Equine Vet J. 2012;44(1):25-32.

44.  Machova Urdzikova L, Sedlacek R, Suchy T, et al. Human multipotent mesenchymal stem cells improve healing after collagenase tendon injury in the rat. Biomed Eng Online. 2014;13:42.

45.  Oshita T, Tobita M, Tajima S, Mizuno H. Adipose-derived stem cells improve collagenase-induced tendinopathy in a rat model. Am J Sports Med. 2016 Apr 11. [Epub ahead of print]

46.  Yuksel S, Guleç MA, Gultekin MZ, et al. Comparison of the early-period effects of bone marrow-derived mesenchymal stem cells and platelet-rich plasma on achilles tendon ruptures in rats. Connect Tissue Res. 2016 May 18. [Epub ahead of print]

47.  Chen L, Liu JP, Tang KL, et al. Tendon derived stem cells promote platelet-rich plasma healing in collagenase-induced rat achilles tendinopathy. Cell Physiol Biochem. 2014;34(6):2153-2168.

48.  Pascual-Garrido C, Rolón A, Makino A. Treatment of chronic patellar tendinopathy with autologous bone marrow stem cells: a 5-year-followup. Stem Cells Int. 2012;2012:953510.

49.  Zlotnicki JP, Geeslin AG, Murray IR, et al. Biologic treatments for sports injuries ii think tank-current concepts, future research, and barriers to advancement, part 3: articular cartilage. Orthop J Sports Med. 2016;4(4):2325967116642433.

50.  McCormack RA, Shreve M, Strauss EJ. Biologic augmentation in rotator cuff repair--should we do it, who should get it, and has it worked? Bull Hosp Jt Dis (2013). 2014;72(1):89-96.

51.  Mosna F, Sensebé L, Krampera M. Human bone marrow and adipose tissue mesenchymal stem cells: a user’s guide. Stem Cells Dev. 2010;19(10):1449-1470.

52.  Nakamura T, Sekiya I, Muneta T, et al. Arthroscopic, histological and MRI analyses of cartilage repair after a minimally invasive method of transplantation of allogeneic synovial mesenchymal stromal cells into cartilage defects in pigs. Cytotherapy. 2012;14(3):327-338.

53.  Dragoo JL, Carlson G, McCormick F, et al. Healing full-thickness cartilage defects using adipose-derived stem cells. Tissue Eng. 2007;13(7):1615-1621.

54.  Masuoka K, Asazuma T, Hattori H, et al. Tissue engineering of articular cartilage with autologous cultured adipose tissue-derived stromal cells using atelocollagen honeycomb-shaped scaffold with a membrane sealing in rabbits. J Biomed Mater Res B Appl Biomater. 2006 79(1):25-34.

55.  Gobbi A, Karnatzikos G, Sankineani SR. One-step surgery with multipotent stem cells for the treatment of large full-thickness chondral defects of the knee. Am J Sports Med. 2014;42(3):648-657.

56.  Kim JD, Lee GW, Jung GH, et al. Clinical outcome of autologous bone marrow aspirates concentrate (BMAC) injection in degenerative arthritis of the knee. Eur J Orthop Surg Traumatol. 2014;24(8):1505-1511.

57.  Krych AJ, Nawabi DH, Farshad-Amacker NA, et al. Bone marrow concentrate improves early cartilage phase maturation of a scaffold plug in the knee: a comparative magnetic resonance imaging analysis to platelet-rich plasma and control. Am J Sports Med. 2016;44(1):91-98.

58.  Fortier LA, Potter HG, Rickey EJ, et al. Concentrated bone marrow aspirate improves full-thickness cartilage repair compared with microfracture in the equine model. J Bone Joint Surg Am. 2010;92(10):1927-1937.

59.  Kim YS, Park EH, Kim YC, Koh YG. Clinical outcomes of mesenchymal stem cell injection with arthroscopic treatment in older patients with osteochondral lesions of the talus. Am J Sports Med. 2013;41(5):1090-1099.

60.  Kim YS, Lee HJ, Choi YJ, Kim YI, Koh YG. Does an injection of a stromal vascular fraction containing adipose-derived mesenchymal stem cells influence the outcomes of marrow stimulation in osteochondral lesions of the talus? A clinical and magnetic resonance imaging study. Am J Sports Med. 2014;42(10):2424-2434.

61.  Chahla J, Dean CS, Moatshe G, Pascual-Garrido C, Serra Cruz R, LaPrade RF. Concentrated bone marrow aspirate for the treatment of chondral injuries and osteoarthritis of the knee: a systematic review of outcomes. Orthop J Sports Med. 2016;4(1):2325967115625481.

62.  Gobbi A, Karnatzikos G, Scotti C, Mahajan V, Mazzucco L, Grigolo B. One-step cartilage repair with bone marrow aspirate concentrated cells and collagen matrix in full-thickness knee cartilage lesions: results at 2-year follow-up. Cartilage. 2011;2(3):286-299.

63.  Ruiz-Ibán MÁ, Díaz-Heredia J, García-Gómez I, Gonzalez-Lizán F, Elías-Martín E, Abraira V. The effect of the addition of adipose-derived mesenchymal stem cells to a meniscal repair in the avascular zone: an experimental study in rabbits. Arthroscopy. 2011;27(12):1688-1696.

64.  Angele P, Johnstone B, Kujat R, et al. Stem cell based tissue engineering for meniscus repair. J Biomed Mater Res A. 2008;85(2):445-455.

65.  Hernigou P, Merouse G, Duffiet P, Chevalier N, Rouard H. Reduced levels of mesenchymal stem cells at the tendon-bone interface tuberosity in patients with symptomatic rotator cuff tear. Int Orthop. 2015;39(6):1219-1225.

66.  Goutallier D, Postel JM, Gleyze P, Leguilloux P, Van Driessche S. Influence of cuff muscle fatty degeneration on anatomic and functional outcomes after simple suture of full-thickness tears. J Shoulder Elbow Surg. 2003;12(6):550-554.

67.   Kovacevic D, Rodeo SA. Biological augmentation of rotator cuff tendon repair. Clin Orthop Relat Res. 2008;466(3):622-633.

68.   Valencia Mora M, Antuña Antuña S, García Arranz M, Carrascal MT, Barco R. Application of adipose tissue-derived stem cells in a rat rotator cuff repair model. Injury. 2014;45 Suppl 4:S22-S27.

69.  Yokoya S, Mochizuki Y, Natsu K, Omae H, Nagata Y, Ochi M. Rotator cuff regeneration using a bioabsorbable material with bone marrow-derived mesenchymal stem cells in a rabbit model. Am J Sports Med. 2012;40(6):1259-1268.

70.  Gulotta LV, Kovacevic D, Ehteshami JR, Dagher E, Packer JD, Rodeo SA. Application of bone marrow-derived mesenchymal stem cells in a rotator cuff repair model. Am J Sports Med. 2009;37(11):2126-2133.

71.  Beitzel K, McCarthy MB, Cote MP, et al. Comparison of mesenchymal stem cells (osteoprogenitors) harvested from proximal humerus and distal femur during arthroscopic surgery. Arthroscopy. 2013;29(2):301-308.

72.  Utsunomiya H, Uchida S, Sekiya I, Sakai A, Moridera K, Nakamura T. Isolation and characterization of human mesenchymal stem cells derived from shoulder tissues involved in rotator cuff tears. Am J Sports Med. 2013;41(3):657-668.

73.  Hernigou P, Flouzat Lachaniette CH, Delambre J, et al. Biologic augmentation of rotator cuff repair with mesenchymal stem cells during arthroscopy improves healing and prevents further tears: a case-controlled study. Int Orthop. 2014;38(9):1811-1818.

74.  Ellera Gomes JL, da Silva RC, Silla LM, Abreu MR, Pellanda R. Conventional rotator cuff repair complemented by the aid of mononuclear autologous stem cells. Knee Surg Sports Traumatol Arthrosc. 2012;20(2):373-377.

75.  Adams SB Jr, Thorpe MA, Parks BG, Aghazarian G, Allen E, Schon LC. Stem cell-bearing suture improves Achilles tendon healing in a rat model. Foot Ankle Int. 2014;35(3):293-299.

76.  Huang TF, Yew TL, Chiang ER, et al. Mesenchymal stem cells from a hypoxic culture improve and engraft Achilles tendon repair. Am J Sports Med. 2013;41(5):1117-1125.

77.  Yao J, Woon CY, Behn A, et al. The effect of suture coated with mesenchymal stem cells and bioactive substrate on tendon repair strength in a rat model. J Hand Surg Am. 2012;37(8):1639-1645.

78.  Uysal CA, Tobita M, Hyakusoku H, Mizuno H. Adipose-derived stem cells enhance primary tendon repair: biomechanical and immunohistochemical evaluation. J Plast Reconstr Aesthet Surg. 2012;65(12):1712-1719.

79.  Stein BE, Stroh DA, Schon LC. Outcomes of acute Achilles tendon rupture repair with bone marrow aspirate concentrate augmentation. Int Orthop. 2015;39(5):901-905.

80.  Chen B, Li B, Qi YJ, et al. Enhancement of tendon-to-bone healing after anterior cruciate ligament reconstruction using bone marrow-derived mesenchymal stem cells genetically modified with bFGF/BMP2. Sci Rep. 2016;6:25940.

81.  Kanaya A, Deie M, Adachi N, Nishimori M, Yanada S, Ochi M. Intra-articular injection of mesenchymal stromal cells in partially torn anterior cruciate ligaments in a rat model. Arthroscopy. 2007;23(6):610-617.

82.  Figueroa D, Espinosa M, Calvo R, et al. Anterior cruciate ligament regeneration using mesenchymal stem cells and collagen type I scaffold in a rabbit model. Knee Surg Sports Traumatol Arthrosc. 2014;22(5):1196-1202.

83.  Silva A, Sampaio R, Fernandes R, Pinto E. Is there a role for adult non-cultivated bone marrow stem cells in ACL reconstruction? Knee Surg Sports Traumatol Arthrosc. 2014;22(1):66-71.

84.  Pepke W, Kasten P, Beckmann NA, Janicki P, Egermann M. Core decompression and autologous bone marrow concentrate for treatment of femoral head osteonecrosis: a randomized prospective study. Orthop Rev (Pavia). 2016;8(1):6162.

85.   Kopka M, Bradley JP.  The use of biologic agents in athletes with knee injuries. J Knee Surg. 2016 May 20. [Epub ahead of print]

86.  Valencia Mora M, Ruiz Ibán MA, Díaz Heredia J, Barco Laakso R, Cuéllar R, García Arranz M. Stem cell therapy in the management of shoulder rotator cuff disorders. World J Stem Cells. 2015;7(4):691-699.

87.  Johnstone B, Hering TM, Caplan AI, Goldberg VM, Yoo JU. In vitro chondrogenesis of bone marrow-derived mesenchymal progenitor cells. Exp Cell Res. 1998;238(1):265-272.

88.  Ferrari G, Cusella-De Angelis G, Coletta M, et al. Muscle regeneration by bone marrow-derived myogenic progenitors. Science. 1998;279(5356):1528-1530.

89.  Pittenger MF, Mackay AM, Beck SC, et al. Multilineage potential of adult human mesenchymal stem cells. Science. 1999;284(5411):143-147.

90.  Fukuda K. Molecular characterization of regenerated cardiomyocytes derived from adult mesenchymal stem cells. Congenit Anom (Kyoto). 2002;42(1):1-9.

91.  Ito T, Suzuki A, Okabe M, Imai E, Hori M. Application of bone marrow-derived stem cells in experimental nephrology. Exp Nephrol. 2001;9(6):444-450.

92.  Qu-Petersen Z, Deasy B, Jankowski R, et al. Identification of a novel population of muscle stem cells in mice: potential for muscle regeneration. J Cell Biol. 2002;157(5):851-864.

93.  Shi S, Gronthos S, Chen S, et al. Bone formation by human postnatal bone marrow stromal stem cells is enhanced by telomerase expression. Nat Biotechnol. 2002;20(6):587-591.

94.  Deans TL, Elisseeff JH. Stem cells in musculoskeletal engineered tissue. Curr Opin Biotechnol. 2009;20(5):537-544.

95.  Funk JF, Matziolis G, Krocker D, Perka C. [Promotion of bone healing through clinical application of autologous periosteum derived stem cells in a case of atrophic non-union]. Z Orthop Unfall. 2007;145(6):790-794.

Article PDF
Author and Disclosure Information

Bryan M. Saltzman, MD, Benjamin D. Kuhns, MD, MS, Alexander E. Weber, MD, Adam Yanke, MD, and Shane J. Nho, MD, MS 

Authors’ Disclosure Statement: Dr. Saltzman reports that he receives royalties from Nova Science Publishers and Postgraduate Institute for Medicine. Dr. Yanke reports that he receives research support from Arthrex and NuTech. Dr. Nho reports that he receives research support from Allosource, Arthrex, Athletico, DJ Orthopaedics, Linvatec, Miomed, Smith & Nephew, and Stryker; is on the editorial/governing board of The American Journal of Orthopedics; is on the board of the American Orthopaedic Society for Sports Medicine and the Arthroscopy Association of North America; is a paid consultant for Ossur and Stryker; and receives publishing royalties from Springer. The other authors report no actual or potential conflict of interest in relation to this article.

Issue
The American Journal of Orthopedics - 45(5)
Publications
Topics
Page Number
280-288, 326
Legacy Keywords
review, stem cells, biologics, guide, orthopedist, biologic adjuvants, treatment, biomechanical, cells, platelet-rich plasma, PRP, repair, saltzman, kuhns, weber, yanke, nho
Sections
Author and Disclosure Information

Bryan M. Saltzman, MD, Benjamin D. Kuhns, MD, MS, Alexander E. Weber, MD, Adam Yanke, MD, and Shane J. Nho, MD, MS 

Authors’ Disclosure Statement: Dr. Saltzman reports that he receives royalties from Nova Science Publishers and Postgraduate Institute for Medicine. Dr. Yanke reports that he receives research support from Arthrex and NuTech. Dr. Nho reports that he receives research support from Allosource, Arthrex, Athletico, DJ Orthopaedics, Linvatec, Miomed, Smith & Nephew, and Stryker; is on the editorial/governing board of The American Journal of Orthopedics; is on the board of the American Orthopaedic Society for Sports Medicine and the Arthroscopy Association of North America; is a paid consultant for Ossur and Stryker; and receives publishing royalties from Springer. The other authors report no actual or potential conflict of interest in relation to this article.

Author and Disclosure Information

Bryan M. Saltzman, MD, Benjamin D. Kuhns, MD, MS, Alexander E. Weber, MD, Adam Yanke, MD, and Shane J. Nho, MD, MS 

Authors’ Disclosure Statement: Dr. Saltzman reports that he receives royalties from Nova Science Publishers and Postgraduate Institute for Medicine. Dr. Yanke reports that he receives research support from Arthrex and NuTech. Dr. Nho reports that he receives research support from Allosource, Arthrex, Athletico, DJ Orthopaedics, Linvatec, Miomed, Smith & Nephew, and Stryker; is on the editorial/governing board of The American Journal of Orthopedics; is on the board of the American Orthopaedic Society for Sports Medicine and the Arthroscopy Association of North America; is a paid consultant for Ossur and Stryker; and receives publishing royalties from Springer. The other authors report no actual or potential conflict of interest in relation to this article.

Article PDF
Article PDF

Biologic use in orthopedics is a continuously evolving field that complements technical, anatomic, and biomechanical advancements in orthopedics. Biologic agents are receiving increasing attention for their use in augmenting healing of muscles, tendons, ligaments, and osseous structures. As biologic augmentation strategies become increasingly utilized in bony and soft-tissue injuries, research on stem cell use in orthopedics continues to increase. Stem cell-based therapies for the repair or regeneration of muscle and tendon represent a promising technology going forward for numerous diseases.1

Stem cells by definition are undifferentiated cells that have 4 main characteristics: (1) mobilization during angiogenesis, (2) differentiation into specialized cell types, (3) proliferation and regeneration, and (4) release of immune regulators and growth factors.2 Mesenchymal stem cells (MSCs) have garnered the most attention in the field of surgery due to their ability to differentiate into the tissues of interest for the surgeon.3 This includes both bone marrow-derived mesenchymal stem cells (bm-MSCs) and adipose-derived mesenchymal stem cells (a-MSCs). These multipotent stem cells in adults originate from mesenchymal tissues, including bone marrow, tendon, adipose, and muscle tissue.4 They are attractive for clinical use because of their multipotent potential and relative ease of growth in culture.5 They also exert a paracrine effect to modulate and control inflammation, stimulate endogenous cell repair and proliferation, inhibit apoptosis, and improve blood flow through secretion of chemokines, cytokines, and growth factors.6,7

Questions exist regarding the best way to administer stem cells, whether systematic administration is possible for these cells to localize to the tissue in need, or more likely if direct application to the pathologic area is necessary.8,9 A number of sources, purification process, and modes of delivery are available, but the most effective means of preparation and administration are still under investigation. The goal of this review is to illustrate the current state of knowledge surrounding stem cell therapy in orthopedics with a focus on osteoarthritis, tendinopathy, articular cartilage, and enhancement of surgical procedures.

Important Considerations

Common stem cell isolates include embryonic, induced pluripotent, and mesenchymal formulations (Table 1). MSCs can be obtained from multiple sites, including but not limited to the adult bone marrow, adipose, muscular, or tendinous tissues, and their use has been highlighted in the study of numerous orthopedic and nonorthopedic pathologies over the course of the last decade. Research on the use of embryonic stem cells in medical therapy with human implications has received substantial attention, with many ethical concerns by those opposed, and the existence of a potential risk of malignant alterations.8,10 Amniotic-derived stem cells can be isolated from amniotic fluid, umbilical cord blood, or the placenta and thus do not harbor the same social constraints as the aforementioned embryonic cells; however, they do not harbor the same magnitude of multi-differentiation potential, either.4

Adult MSCs are more locally available and easy to obtain for treatment when compared with embryonic and fetal stem cells, and the former has a lower immunogenicity, which allows allogeneic use.11 Safety has been preliminarily demonstrated in use thus far; Centeno and colleagues12 found no neoplastic tissue generation at the site of stem cell injection after 3 years postinjection for a cohort of patients who were treated with autologous bm-MSCs for various pathologies. Self-limited pain and swelling are the most commonly reported adverse events after use.13 However, long-term data are lacking in many instances to definitively suggest the absence of possible complications.

Basic Science

Stem cell research encompasses a wide range of rapidly developing treatment strategies that are applicable to virtually every field of medicine. In general, stem cells can be classified as embryonic stem cells (ESCs), induced pluripotent stem (iPS) cells, or adult-derived MSCs. ESCs are embryonic cells derived typically from fetal tissue, whereas iPS cells are dedifferentiated from adult tissue, thus avoiding many of the ethical and legal challenges imposed by research with ESCs. However, oncogenic and lingering politico-legal concerns with introducing dedifferentiated ESCs or iPS cells into healthy tissue necessitate the development, isolation, and expansion of multi- but not pluripotent stem cell lines.14 To date, the most advantageous and widely utilized from any perspective are MSCs, which can further differentiate into cartilage, tendon, muscle, and bony tissue.7,15,16

MSCs are defined by their ability to demonstrate in vitro differentiation into osteoblasts, adipocytes, or chondroblasts, adhere to plastic, express CD105, CD73, and CD90, and not express CD43, CD23, CD14 or CD11b, CD79 or CD19, or HLA-DR.17 Porada and Almeida-Porada18 have outlined 6 reasons highlighting the advantages of MSCs: 1) ease of isolation, 2) high differentiation capabilities, 3) strong colony expansion without differentiation loss, 4) immunosuppression following transplantation, 5) powerful anti-inflammatory properties, and 6) their ability to localize to damaged tissue. The anti-inflammatory properties of MSCs are particularly important as they promote allo- and xenotransplantation from donor tissues.19,20 MSCs can be isolated from numerous sources, including but not limited to bone marrow, periosteum, adipocyte, and muscle.21-23 Interestingly, the source tissue used to isolate MSCs can affect differentiation capabilities, colony size, and growth rate (Table 2).24 Advantages of a-MSCs include high prevalence and ease of harvest; however, several animal studies have shown inferior results when compared to bm-MSCs.25-27 More research is needed to determine the ideal source material for MSCs, which will likely depend in part on the procedure for which they are employed.27

 

 

Following harvesting, isolation, and expansion, MSC delivery methods for treatments typically consist of either cell-based or tissue engineering approaches. Cell-based techniques involve the injection of MSCs into damaged tissues. Purely cell-based therapy has shown success in limited clinical trials involving knee osteoarthritis, cartilage repair, and meniscal repair.28-30 However, additional studies with longer follow-up are required to validate these preliminary findings. Tissue engineering approaches involve the construction of a 3-dimensional scaffold seeded with MSCs that is later surgically implanted. While promising in theory, limited and often conflicting data exist regarding the efficacy of tissue-engineered MSC implantation.31-32 Suboptimal scaffold vascularity is a major limitation to scaffold design, which may be alleviated in part with the advent of 3-dimensional printing and the ability to more precisely alter scaffold architecture.14,33 Additional limitations include ensuring MSC purity and differentiation potential following harvesting and expansion. At present, the use of tissue engineering with MSCs is promising but it remains a nascent technology with additional preclinical studies required to confirm implant efficacy and safety.

Clinical Entities

Osteoarthritis

MSC therapies have emerged as promising treatment strategies in the setting of early osteoarthritis (OA). In addition to their regenerative potential, MSCs demonstrate potent anti-inflammatory properties, increasing their attractiveness as biologic agents in the setting of OA.34 Over the past decade, multiple human trials have been published demonstrating the efficacy of MSC injections into patients with OA.35,36 In a study evaluating a-MSC injection into elderly patients (age >65 years) with knee OA, Koh and colleagues29 found that 88% demonstrated improved cartilage status at 2-year follow-up, while no patient underwent a total knee arthroplasty during this time period. In another study investigating patients with unicompartmental knee OA with varus alignment undergoing high tibial osteotomy and microfracture, Wong and colleagues37 reported improved clinical, patient-reported, and magnetic resonance imaging (MRI)-based outcomes in a group receiving a preoperative MSC injection compared to a control group. Further, in a recent randomized control trial of patients with knee osteoarthritis, Vega and colleagues38 reported improved cartilage and quality of life outcomes at 1 year following MSC injection compared to a control group receiving a hyaluronic acid injection. In addition to knee OA, studies have also reported improvement in ankle OA following MSC injection.39 While promising, many of the preliminary clinical studies evaluating the efficacy of MSC therapies in the treatment of OA are hindered by small patient populations and short-term follow-up. Additional large-scale, randomized studies are required and many are ongoing presently in hopes of validating these preliminary findings.36

Tendinopathy

The quality of repaired tissue in primary tendon-to-tendon and tendon-to-bone healing has long been a topic of great interest.40 The healing potential of tendons is inferior to that of other bony and connective tissues,41 with tendon healing typically resulting in a biomechanically and histologically inferior structure to the native tissue.42 As such, this has been a particularly salient opportunity for stem cell use with hopes of recapitulating a more normal tendon or tendon enthesis following injury. In addition to the acute injury, there is great interest in the application of stem cells to chronic states of injury such as tendinopathy.

In equine models, the effect of autologous bm-MSCs treatment on tendinopathy of the superficial digital flexor tendon has been studied. Godwin and colleagues43 evaluated 141 race horses with spontaneous superficial digital flexor tendinopathy treated in this manner, and reported a reinjury percentage in these treated horses of just 27.4%, which compared favorably to historical controls and alternative therapeutics. Machova Urdzikova and colleagues44 injected MSCs at Achilles tendinopathy locations to augment nonoperative healing in 40 rats, and identified more native histological organization and improved vascularization in comparison to control rat specimens. Oshita and colleagues45 reported histologic improvement of tendinopathy findings in 8 rats receiving a-MSCs at the location of induced Achilles tendinopathy that was significantly superior to a control cohort. Bm-MSCs were used by Yuksel and colleagues46 in comparison with platelet-rich plasma (PRP) for treatment of Achilles tendon ruptures created surgically in rat models. They demonstrated successful effects with its use in terms of recovery for the tendon’s histopathologic, immunohistochemical, and biomechanical properties, related to significantly greater levels of anti-inflammatory cytokines. However, these aforementioned findings have not been uniform across the literature—other authors have reported findings that MSC transplantation alone did not repair Achilles tendon injury with such high levels of success.47

Human treatment of tendinopathies with stem cells has been scarcely studied to date. Pascual-Garrido and colleagues48 evaluated 8 patients with refractory patellar tendinopathy treated with injection of autologous bm-MSCs and reported successful results at 2- to 5-year follow-up, with significant improvements in patient-reported outcome measures for 100% of patients. Seven of 8 (87.5%) noted that they would undergo the procedure again.

 

 

Articular Cartilage Injury

Chondral injury is a particularly important subject given the limited potential of chondrocytes to replicate or migrate to the site of pathology.49 Stem cell use in this setting assists with programmed growth factor release and alteration of the anatomic microenvironment to facilitate regeneration and repair of the chondral surface. Autologous stem cell use through microfracture provides a perforation into the bone marrow and a subsequent fibrin clot formation containing platelets, growth factors, vascular elements, and MSCs.50 A similar concept to PRP is currently being explored with bm-MSCs. Isolated bm-MSCs are commonly referred to as bone marrow aspirate or bone marrow aspirate concentrate (BMAC). Commercially available systems are now available to aid in the harvesting and implementation of BMAC. One of the more promising avenues for BMAC implementation is in articular cartilage repair or regeneration due to chondrogenic potential of BMAC when used in isolation or when combined with microfracture, chondrocyte transfer, or collagen scaffolds.19,51 Synovial-derived stem cells as an additional source for stem cell use has demonstrated excellent chondrogenic potential in animal studies with full-thickness lesion healing and native-appearing cartilage histologically.52 Incorporation of a-MSCs into scaffolds for surgical implantation has demonstrated success in repairing full-thickness chondral defects with continuous joint surface and extracellular proteins, surface markers, and gene products similar to the native cartilage in animal models.53,54 In light of the promising basic science and animal studies, clinical studies have begun to emerge.55-57

Fortier and colleagues58 found MRI and histologic evidence of full-thickness chondral repair and increased integration with neighboring cartilage when BMAC was concurrently used at the time of microfracture in an equine model. Fortier and colleagues58 also demonstrated greater healing in equine models with acute full-thickness cartilage defects treated by microfracture with MSCs than without delivery of MSCs. Kim and colleagues59,60 similarly reported superiority in clinical outcomes for patients with osteochondral lesions of the talus treated with marrow stimulation and MSC injection than by the former in isolation.

In humans, stem cell use for chondral repair has additionally proven promising. A systematic review of the literature suggested good to excellent overall outcomes for the treatment of moderate focal chondral defects with BMAC with or without scaffolds and microfracture with inclusion of 8 total publications.61 This review included Gobbi and colleagues,62 who prospectively treated 15 patients with a mean focal chondral defect size of 9.2 cm2 about the knee. Use of BMAC covered with a collagen I/III matrix produced significant improvements in patient-reported outcome scores and MRI demonstrated complete hyaline-like cartilage coverage in 80%, with second-look arthroscopy demonstrating normal to nearly normal tissue. Gobbi and colleagues55 also found evidence for superiority of chondral defects treated with BMAC compared to matrix-induced autologous chondrocyte implantation (MACI) for patellofemoral lesions in 37 patients (MRI showed complete filling of defects in 81% of BMAC-treated patients vs 76% of MACI-treated patients).

Meniscal Repair

Clinical application of MSCs in the treatment of meniscal pathology is evolving as well. ASCs have been added to modify the biomechanical environment of avascular zone meniscal tears at the time of suture repair in a rabbit, and have demonstrated increased healing rates in small and larger lesions, although the effect lessens with delay in repair.63 Angele and colleagues64 treated meniscal defects in a rabbit model with scaffolds with bm-MSCs compared with empty scaffolds or control cohorts and found a higher proportion of menisci with healed meniscus-like fibrocartilage when MSCs were utilized.

In humans, Vangsness and colleagues30 treated knees with partial medial meniscectomy with allogeneic stem cells and reported an increase in meniscal volume and decrease in pain in those patients when compared to a cohort of knees treated with hyaluronic acid. Despite promising early results, additional clinical studies are necessary to determine the external validity and broad applicability of stem cell use in meniscal repair.

Rotator Cuff Repair

The number of local resident stem cells at the site of rotator cuff tear has been shown to decrease with tear size, chronicity, and degree of fatty infiltration, suggesting that those with the greatest need for a good reparative environment are those least equipped to heal.65 The need for improvement in this domain is related to the still relatively high re-tear rate after rotator cuff repair despite improvements in instrumentation and surgical technique.66 The native fibrocartilaginous transition zone between the humerus and the rotator cuff becomes a fibrovascular scar tissue after rupture and repair with poorer material properties than the native tissue.67 Thus, a-MSCs have been evaluated in this setting to determine if the biomechanical and histological properties of the repair may improve.68

 

 

In rat models, Valencia Mora and colleagues68 reported on the application of a-MSCs in a rat rotator cuff repair model compared to an untreated group. They found no differences between those treated rats and those without a-MSCs use in terms of biomechanical properties of the tendon-to-bone healing, but those with stem cell use had less inflammation shown histologically (diminished presence of edema and neutrophils) at 2- and 4-week time points, which the authors suggested may lead to a more elastic repair and less scar at the bone-tendon healing site. Oh and colleagues1 evaluated the use of a-MSCs in a rabbit subscapularis tear model, and reported significantly reduced fatty infiltration at the site of chronic rotator cuff tear after repair with its application at the repair site; while the load-to-failure was higher in those rabbits with ASCs administration, it was short of reaching statistical significance. Yokoya and colleagues69 demonstrated regeneration of rotator cuff tendon-to-bone insertional site anatomy and in the belly of the cuff tendon in a rabbit model with MSCs applied at the operative site. However, Gulotta and colleagues70 did not see the same improvement in their similar study in the rat model; these authors failed to see improvement in structure, strength, or composition of the tendinous attachment site despite addition of MSCs.

Clinical studies on augmented rotator cuff repair have also found mixed results. MSCs for this purpose have been cultivated from arthroscopic bone marrow aspiration of the proximal humerus71 and subacromial bursa72 with successful and reproducibly high concentrations of stem cells. Hernigou and colleagues73 found a significant improvement in rate of healing (87% intact cuffs vs 44% in the control group) and repair surface tendon integrity (via ultrasound and MRI) for patients at a minimum of 10 years after rotator cuff repair with MSC injection at the time of surgery. The authors found a direct correlation in these outcomes with the number of MSCs injected at the time of repair. Ellera Gomes and colleagues74 injected bm-MSCs obtained from the iliac crest into the tendinous repair site in 14 consecutive patients with full-thickness rotator cuff tears treated by transosseous sutures via a mini-open approach. MRI demonstrated integrity of the repair site in all patients at more than 1-year follow-up.

Achilles Tendon Repair

The goal with stem cell use in Achilles repair is to accelerate the healing and rehabilitation. Several animal studies have demonstrated improved mechanical properties and collagen composition of tendon repairs augmented with stem cells, including Achilles tendon repair in a rat model. Adams and colleagues75 compared suture alone (36 tendons) to suture plus stem cell concentrate injection (36 tendons) and stem cell loaded suture (36 tendons) in Achilles tendon repair with rat models. The suture-alone cohort had lower ultimate failure loads at 14 days after surgery, indicating biomechanical superiority with stem cell augmentation means. Transplantation of hypoxic MSCs at the time of Achilles tendon repair may be a promising option for superior biomechanical failure loads and histologic findings as per recent rat model findings by Huang and colleagues.76 Yao and colleagues77 demonstrated increased strength of suture repair for Achilles repair in rat models at early time points when using MSC-coated suture in comparison to standard suture, and suggested that the addition of stem cells may improve early mechanical properties during the tendon repair process. A-MSC addition to PRP has provided significantly increased tensile strength to rabbit models with Achilles tendon repair as well.78

In evaluation of stem cell use for this purpose with humans, Stein and colleagues79 reviewed 28 sports-related Achilles tendon ruptures in 27 patients treated with open repair and BMAC injection. At a mean follow-up of 29.7 months, the authors reported no re-ruptures, with 92% return to sport at 5.9 months, and excellent clinical outcomes. This small cohort study found no adverse outcomes related to the BMAC addition, and thus proposed further study of the efficacy of stem cell treatment for Achilles tendon repair.

Anterior Cruciate Ligament Reconstruction

Bm-MSCs genetically modified with bone morphogenetic protein 2 (BMP2) and basic fibroblast growth factor (bFGF) have shown great promise in improvement of the formation of mechanically sound tendon-bone interface in anterior cruciate ligament (ACL) reconstruction.80 Similar to the other surgical procedures mentioned in this review, animal studies have successfully evaluated the augmentation of osteointegration of tendon to bone in the setting of ACL reconstruction. Jang and colleagues3 investigated the use of nonautologous transplantation of human umbilical cord blood-derived MSCs in a rabbit ACL reconstruction model. The authors demonstrated a lack of immune rejection, and enhanced tendon-bone healing with broad fibrocartilage formation at the transition zone (similar to the native ACL) and decreased femoral and tibial tunnel widening as compared to a control cohort at 12-weeks after surgery. In a rat model, Kanaya and colleagues81 reported improved histological scores and slight improvements in biomechanical integrity of partially transected rat ACLs treated with intra-articular MSC injection. Stem cell use in the form of suture-supporting scaffolds seeded with MSCs has been evaluated in a total ACL transection rabbit model; the authors of this report demonstrated total ACL regeneration in one-third of samples treated with this augmentation option, in comparison to complete failure in all suture and scaffold alone groups.82

 

 

The use of autologous MSCs in ACL healing remains limited to preclinical research and small case series of patients. One human trial by Silva and colleagues83 evaluated the graft-to-bone site of healing in ACL reconstruction for 20 patients who received an intraoperative infiltration of their graft with adult bm-MSCs. MRI and histologic analysis showed no difference in comparison to control groups, but the authors’ conclusion proposed that the number of stem cells injected might have been too minimal to show a clinical effect.

Other Applications

Although outside the scope of this article, stem cells have demonstrated efficacy in the treatment of a number of osseous clinical entities. This includes the treatment of fracture nonunion, augmentation of spinal fusion, and assistance in the treatment of osteonecrosis.84

Summary

As a scientific community, our understanding of the use of stem cells, their nuances, and their indications has expanded dramatically over the last several years. Stem cell treatment has particularly infiltrated the world of operative and nonoperative sports medicine, given in part the active patient population seeking greater levels of improvement.85 Stem cell therapy offers a potentially effective therapy for a multitude of pathologies because of these cells’ anti-inflammatory, immunoregulatory, angiogenic, and paracrine effects.86 It thus remains a very dynamic option in the study of musculoskeletal tissue regeneration. While the potential exists for stem cell use in daily surgery practices, it is still premature to predict whether this can be expected.

The ideal stem cell sources (including allogeneic or autologous), preparation, cell number, timing, and means of application continue to be evaluated, as well as those advantageous pathologies that can benefit from the technology. In order to better answer these pertinent questions, we need to make sure we have a safe, economic, and ethically acceptable means for stem cell translational research efforts. More high-level studies with standardized protocols need to be performed. It is necessary to improve national and international collaboration in research, as well as collaboration with governing bodies, to attempt to further scientific advancement in this field of research.49 Further study on embryonic stem cell use may be valuable as well, pending governmental approval. Finally, more dedicated research efforts must be placed on the utility of adjuncts with stem cell use, including PRP and scaffolds, which may increase protection, nutritional support, and mechanical stimulation of the administered stem cells.

Biologic use in orthopedics is a continuously evolving field that complements technical, anatomic, and biomechanical advancements in orthopedics. Biologic agents are receiving increasing attention for their use in augmenting healing of muscles, tendons, ligaments, and osseous structures. As biologic augmentation strategies become increasingly utilized in bony and soft-tissue injuries, research on stem cell use in orthopedics continues to increase. Stem cell-based therapies for the repair or regeneration of muscle and tendon represent a promising technology going forward for numerous diseases.1

Stem cells by definition are undifferentiated cells that have 4 main characteristics: (1) mobilization during angiogenesis, (2) differentiation into specialized cell types, (3) proliferation and regeneration, and (4) release of immune regulators and growth factors.2 Mesenchymal stem cells (MSCs) have garnered the most attention in the field of surgery due to their ability to differentiate into the tissues of interest for the surgeon.3 This includes both bone marrow-derived mesenchymal stem cells (bm-MSCs) and adipose-derived mesenchymal stem cells (a-MSCs). These multipotent stem cells in adults originate from mesenchymal tissues, including bone marrow, tendon, adipose, and muscle tissue.4 They are attractive for clinical use because of their multipotent potential and relative ease of growth in culture.5 They also exert a paracrine effect to modulate and control inflammation, stimulate endogenous cell repair and proliferation, inhibit apoptosis, and improve blood flow through secretion of chemokines, cytokines, and growth factors.6,7

Questions exist regarding the best way to administer stem cells, whether systematic administration is possible for these cells to localize to the tissue in need, or more likely if direct application to the pathologic area is necessary.8,9 A number of sources, purification process, and modes of delivery are available, but the most effective means of preparation and administration are still under investigation. The goal of this review is to illustrate the current state of knowledge surrounding stem cell therapy in orthopedics with a focus on osteoarthritis, tendinopathy, articular cartilage, and enhancement of surgical procedures.

Important Considerations

Common stem cell isolates include embryonic, induced pluripotent, and mesenchymal formulations (Table 1). MSCs can be obtained from multiple sites, including but not limited to the adult bone marrow, adipose, muscular, or tendinous tissues, and their use has been highlighted in the study of numerous orthopedic and nonorthopedic pathologies over the course of the last decade. Research on the use of embryonic stem cells in medical therapy with human implications has received substantial attention, with many ethical concerns by those opposed, and the existence of a potential risk of malignant alterations.8,10 Amniotic-derived stem cells can be isolated from amniotic fluid, umbilical cord blood, or the placenta and thus do not harbor the same social constraints as the aforementioned embryonic cells; however, they do not harbor the same magnitude of multi-differentiation potential, either.4

Adult MSCs are more locally available and easy to obtain for treatment when compared with embryonic and fetal stem cells, and the former has a lower immunogenicity, which allows allogeneic use.11 Safety has been preliminarily demonstrated in use thus far; Centeno and colleagues12 found no neoplastic tissue generation at the site of stem cell injection after 3 years postinjection for a cohort of patients who were treated with autologous bm-MSCs for various pathologies. Self-limited pain and swelling are the most commonly reported adverse events after use.13 However, long-term data are lacking in many instances to definitively suggest the absence of possible complications.

Basic Science

Stem cell research encompasses a wide range of rapidly developing treatment strategies that are applicable to virtually every field of medicine. In general, stem cells can be classified as embryonic stem cells (ESCs), induced pluripotent stem (iPS) cells, or adult-derived MSCs. ESCs are embryonic cells derived typically from fetal tissue, whereas iPS cells are dedifferentiated from adult tissue, thus avoiding many of the ethical and legal challenges imposed by research with ESCs. However, oncogenic and lingering politico-legal concerns with introducing dedifferentiated ESCs or iPS cells into healthy tissue necessitate the development, isolation, and expansion of multi- but not pluripotent stem cell lines.14 To date, the most advantageous and widely utilized from any perspective are MSCs, which can further differentiate into cartilage, tendon, muscle, and bony tissue.7,15,16

MSCs are defined by their ability to demonstrate in vitro differentiation into osteoblasts, adipocytes, or chondroblasts, adhere to plastic, express CD105, CD73, and CD90, and not express CD43, CD23, CD14 or CD11b, CD79 or CD19, or HLA-DR.17 Porada and Almeida-Porada18 have outlined 6 reasons highlighting the advantages of MSCs: 1) ease of isolation, 2) high differentiation capabilities, 3) strong colony expansion without differentiation loss, 4) immunosuppression following transplantation, 5) powerful anti-inflammatory properties, and 6) their ability to localize to damaged tissue. The anti-inflammatory properties of MSCs are particularly important as they promote allo- and xenotransplantation from donor tissues.19,20 MSCs can be isolated from numerous sources, including but not limited to bone marrow, periosteum, adipocyte, and muscle.21-23 Interestingly, the source tissue used to isolate MSCs can affect differentiation capabilities, colony size, and growth rate (Table 2).24 Advantages of a-MSCs include high prevalence and ease of harvest; however, several animal studies have shown inferior results when compared to bm-MSCs.25-27 More research is needed to determine the ideal source material for MSCs, which will likely depend in part on the procedure for which they are employed.27

 

 

Following harvesting, isolation, and expansion, MSC delivery methods for treatments typically consist of either cell-based or tissue engineering approaches. Cell-based techniques involve the injection of MSCs into damaged tissues. Purely cell-based therapy has shown success in limited clinical trials involving knee osteoarthritis, cartilage repair, and meniscal repair.28-30 However, additional studies with longer follow-up are required to validate these preliminary findings. Tissue engineering approaches involve the construction of a 3-dimensional scaffold seeded with MSCs that is later surgically implanted. While promising in theory, limited and often conflicting data exist regarding the efficacy of tissue-engineered MSC implantation.31-32 Suboptimal scaffold vascularity is a major limitation to scaffold design, which may be alleviated in part with the advent of 3-dimensional printing and the ability to more precisely alter scaffold architecture.14,33 Additional limitations include ensuring MSC purity and differentiation potential following harvesting and expansion. At present, the use of tissue engineering with MSCs is promising but it remains a nascent technology with additional preclinical studies required to confirm implant efficacy and safety.

Clinical Entities

Osteoarthritis

MSC therapies have emerged as promising treatment strategies in the setting of early osteoarthritis (OA). In addition to their regenerative potential, MSCs demonstrate potent anti-inflammatory properties, increasing their attractiveness as biologic agents in the setting of OA.34 Over the past decade, multiple human trials have been published demonstrating the efficacy of MSC injections into patients with OA.35,36 In a study evaluating a-MSC injection into elderly patients (age >65 years) with knee OA, Koh and colleagues29 found that 88% demonstrated improved cartilage status at 2-year follow-up, while no patient underwent a total knee arthroplasty during this time period. In another study investigating patients with unicompartmental knee OA with varus alignment undergoing high tibial osteotomy and microfracture, Wong and colleagues37 reported improved clinical, patient-reported, and magnetic resonance imaging (MRI)-based outcomes in a group receiving a preoperative MSC injection compared to a control group. Further, in a recent randomized control trial of patients with knee osteoarthritis, Vega and colleagues38 reported improved cartilage and quality of life outcomes at 1 year following MSC injection compared to a control group receiving a hyaluronic acid injection. In addition to knee OA, studies have also reported improvement in ankle OA following MSC injection.39 While promising, many of the preliminary clinical studies evaluating the efficacy of MSC therapies in the treatment of OA are hindered by small patient populations and short-term follow-up. Additional large-scale, randomized studies are required and many are ongoing presently in hopes of validating these preliminary findings.36

Tendinopathy

The quality of repaired tissue in primary tendon-to-tendon and tendon-to-bone healing has long been a topic of great interest.40 The healing potential of tendons is inferior to that of other bony and connective tissues,41 with tendon healing typically resulting in a biomechanically and histologically inferior structure to the native tissue.42 As such, this has been a particularly salient opportunity for stem cell use with hopes of recapitulating a more normal tendon or tendon enthesis following injury. In addition to the acute injury, there is great interest in the application of stem cells to chronic states of injury such as tendinopathy.

In equine models, the effect of autologous bm-MSCs treatment on tendinopathy of the superficial digital flexor tendon has been studied. Godwin and colleagues43 evaluated 141 race horses with spontaneous superficial digital flexor tendinopathy treated in this manner, and reported a reinjury percentage in these treated horses of just 27.4%, which compared favorably to historical controls and alternative therapeutics. Machova Urdzikova and colleagues44 injected MSCs at Achilles tendinopathy locations to augment nonoperative healing in 40 rats, and identified more native histological organization and improved vascularization in comparison to control rat specimens. Oshita and colleagues45 reported histologic improvement of tendinopathy findings in 8 rats receiving a-MSCs at the location of induced Achilles tendinopathy that was significantly superior to a control cohort. Bm-MSCs were used by Yuksel and colleagues46 in comparison with platelet-rich plasma (PRP) for treatment of Achilles tendon ruptures created surgically in rat models. They demonstrated successful effects with its use in terms of recovery for the tendon’s histopathologic, immunohistochemical, and biomechanical properties, related to significantly greater levels of anti-inflammatory cytokines. However, these aforementioned findings have not been uniform across the literature—other authors have reported findings that MSC transplantation alone did not repair Achilles tendon injury with such high levels of success.47

Human treatment of tendinopathies with stem cells has been scarcely studied to date. Pascual-Garrido and colleagues48 evaluated 8 patients with refractory patellar tendinopathy treated with injection of autologous bm-MSCs and reported successful results at 2- to 5-year follow-up, with significant improvements in patient-reported outcome measures for 100% of patients. Seven of 8 (87.5%) noted that they would undergo the procedure again.

 

 

Articular Cartilage Injury

Chondral injury is a particularly important subject given the limited potential of chondrocytes to replicate or migrate to the site of pathology.49 Stem cell use in this setting assists with programmed growth factor release and alteration of the anatomic microenvironment to facilitate regeneration and repair of the chondral surface. Autologous stem cell use through microfracture provides a perforation into the bone marrow and a subsequent fibrin clot formation containing platelets, growth factors, vascular elements, and MSCs.50 A similar concept to PRP is currently being explored with bm-MSCs. Isolated bm-MSCs are commonly referred to as bone marrow aspirate or bone marrow aspirate concentrate (BMAC). Commercially available systems are now available to aid in the harvesting and implementation of BMAC. One of the more promising avenues for BMAC implementation is in articular cartilage repair or regeneration due to chondrogenic potential of BMAC when used in isolation or when combined with microfracture, chondrocyte transfer, or collagen scaffolds.19,51 Synovial-derived stem cells as an additional source for stem cell use has demonstrated excellent chondrogenic potential in animal studies with full-thickness lesion healing and native-appearing cartilage histologically.52 Incorporation of a-MSCs into scaffolds for surgical implantation has demonstrated success in repairing full-thickness chondral defects with continuous joint surface and extracellular proteins, surface markers, and gene products similar to the native cartilage in animal models.53,54 In light of the promising basic science and animal studies, clinical studies have begun to emerge.55-57

Fortier and colleagues58 found MRI and histologic evidence of full-thickness chondral repair and increased integration with neighboring cartilage when BMAC was concurrently used at the time of microfracture in an equine model. Fortier and colleagues58 also demonstrated greater healing in equine models with acute full-thickness cartilage defects treated by microfracture with MSCs than without delivery of MSCs. Kim and colleagues59,60 similarly reported superiority in clinical outcomes for patients with osteochondral lesions of the talus treated with marrow stimulation and MSC injection than by the former in isolation.

In humans, stem cell use for chondral repair has additionally proven promising. A systematic review of the literature suggested good to excellent overall outcomes for the treatment of moderate focal chondral defects with BMAC with or without scaffolds and microfracture with inclusion of 8 total publications.61 This review included Gobbi and colleagues,62 who prospectively treated 15 patients with a mean focal chondral defect size of 9.2 cm2 about the knee. Use of BMAC covered with a collagen I/III matrix produced significant improvements in patient-reported outcome scores and MRI demonstrated complete hyaline-like cartilage coverage in 80%, with second-look arthroscopy demonstrating normal to nearly normal tissue. Gobbi and colleagues55 also found evidence for superiority of chondral defects treated with BMAC compared to matrix-induced autologous chondrocyte implantation (MACI) for patellofemoral lesions in 37 patients (MRI showed complete filling of defects in 81% of BMAC-treated patients vs 76% of MACI-treated patients).

Meniscal Repair

Clinical application of MSCs in the treatment of meniscal pathology is evolving as well. ASCs have been added to modify the biomechanical environment of avascular zone meniscal tears at the time of suture repair in a rabbit, and have demonstrated increased healing rates in small and larger lesions, although the effect lessens with delay in repair.63 Angele and colleagues64 treated meniscal defects in a rabbit model with scaffolds with bm-MSCs compared with empty scaffolds or control cohorts and found a higher proportion of menisci with healed meniscus-like fibrocartilage when MSCs were utilized.

In humans, Vangsness and colleagues30 treated knees with partial medial meniscectomy with allogeneic stem cells and reported an increase in meniscal volume and decrease in pain in those patients when compared to a cohort of knees treated with hyaluronic acid. Despite promising early results, additional clinical studies are necessary to determine the external validity and broad applicability of stem cell use in meniscal repair.

Rotator Cuff Repair

The number of local resident stem cells at the site of rotator cuff tear has been shown to decrease with tear size, chronicity, and degree of fatty infiltration, suggesting that those with the greatest need for a good reparative environment are those least equipped to heal.65 The need for improvement in this domain is related to the still relatively high re-tear rate after rotator cuff repair despite improvements in instrumentation and surgical technique.66 The native fibrocartilaginous transition zone between the humerus and the rotator cuff becomes a fibrovascular scar tissue after rupture and repair with poorer material properties than the native tissue.67 Thus, a-MSCs have been evaluated in this setting to determine if the biomechanical and histological properties of the repair may improve.68

 

 

In rat models, Valencia Mora and colleagues68 reported on the application of a-MSCs in a rat rotator cuff repair model compared to an untreated group. They found no differences between those treated rats and those without a-MSCs use in terms of biomechanical properties of the tendon-to-bone healing, but those with stem cell use had less inflammation shown histologically (diminished presence of edema and neutrophils) at 2- and 4-week time points, which the authors suggested may lead to a more elastic repair and less scar at the bone-tendon healing site. Oh and colleagues1 evaluated the use of a-MSCs in a rabbit subscapularis tear model, and reported significantly reduced fatty infiltration at the site of chronic rotator cuff tear after repair with its application at the repair site; while the load-to-failure was higher in those rabbits with ASCs administration, it was short of reaching statistical significance. Yokoya and colleagues69 demonstrated regeneration of rotator cuff tendon-to-bone insertional site anatomy and in the belly of the cuff tendon in a rabbit model with MSCs applied at the operative site. However, Gulotta and colleagues70 did not see the same improvement in their similar study in the rat model; these authors failed to see improvement in structure, strength, or composition of the tendinous attachment site despite addition of MSCs.

Clinical studies on augmented rotator cuff repair have also found mixed results. MSCs for this purpose have been cultivated from arthroscopic bone marrow aspiration of the proximal humerus71 and subacromial bursa72 with successful and reproducibly high concentrations of stem cells. Hernigou and colleagues73 found a significant improvement in rate of healing (87% intact cuffs vs 44% in the control group) and repair surface tendon integrity (via ultrasound and MRI) for patients at a minimum of 10 years after rotator cuff repair with MSC injection at the time of surgery. The authors found a direct correlation in these outcomes with the number of MSCs injected at the time of repair. Ellera Gomes and colleagues74 injected bm-MSCs obtained from the iliac crest into the tendinous repair site in 14 consecutive patients with full-thickness rotator cuff tears treated by transosseous sutures via a mini-open approach. MRI demonstrated integrity of the repair site in all patients at more than 1-year follow-up.

Achilles Tendon Repair

The goal with stem cell use in Achilles repair is to accelerate the healing and rehabilitation. Several animal studies have demonstrated improved mechanical properties and collagen composition of tendon repairs augmented with stem cells, including Achilles tendon repair in a rat model. Adams and colleagues75 compared suture alone (36 tendons) to suture plus stem cell concentrate injection (36 tendons) and stem cell loaded suture (36 tendons) in Achilles tendon repair with rat models. The suture-alone cohort had lower ultimate failure loads at 14 days after surgery, indicating biomechanical superiority with stem cell augmentation means. Transplantation of hypoxic MSCs at the time of Achilles tendon repair may be a promising option for superior biomechanical failure loads and histologic findings as per recent rat model findings by Huang and colleagues.76 Yao and colleagues77 demonstrated increased strength of suture repair for Achilles repair in rat models at early time points when using MSC-coated suture in comparison to standard suture, and suggested that the addition of stem cells may improve early mechanical properties during the tendon repair process. A-MSC addition to PRP has provided significantly increased tensile strength to rabbit models with Achilles tendon repair as well.78

In evaluation of stem cell use for this purpose with humans, Stein and colleagues79 reviewed 28 sports-related Achilles tendon ruptures in 27 patients treated with open repair and BMAC injection. At a mean follow-up of 29.7 months, the authors reported no re-ruptures, with 92% return to sport at 5.9 months, and excellent clinical outcomes. This small cohort study found no adverse outcomes related to the BMAC addition, and thus proposed further study of the efficacy of stem cell treatment for Achilles tendon repair.

Anterior Cruciate Ligament Reconstruction

Bm-MSCs genetically modified with bone morphogenetic protein 2 (BMP2) and basic fibroblast growth factor (bFGF) have shown great promise in improvement of the formation of mechanically sound tendon-bone interface in anterior cruciate ligament (ACL) reconstruction.80 Similar to the other surgical procedures mentioned in this review, animal studies have successfully evaluated the augmentation of osteointegration of tendon to bone in the setting of ACL reconstruction. Jang and colleagues3 investigated the use of nonautologous transplantation of human umbilical cord blood-derived MSCs in a rabbit ACL reconstruction model. The authors demonstrated a lack of immune rejection, and enhanced tendon-bone healing with broad fibrocartilage formation at the transition zone (similar to the native ACL) and decreased femoral and tibial tunnel widening as compared to a control cohort at 12-weeks after surgery. In a rat model, Kanaya and colleagues81 reported improved histological scores and slight improvements in biomechanical integrity of partially transected rat ACLs treated with intra-articular MSC injection. Stem cell use in the form of suture-supporting scaffolds seeded with MSCs has been evaluated in a total ACL transection rabbit model; the authors of this report demonstrated total ACL regeneration in one-third of samples treated with this augmentation option, in comparison to complete failure in all suture and scaffold alone groups.82

 

 

The use of autologous MSCs in ACL healing remains limited to preclinical research and small case series of patients. One human trial by Silva and colleagues83 evaluated the graft-to-bone site of healing in ACL reconstruction for 20 patients who received an intraoperative infiltration of their graft with adult bm-MSCs. MRI and histologic analysis showed no difference in comparison to control groups, but the authors’ conclusion proposed that the number of stem cells injected might have been too minimal to show a clinical effect.

Other Applications

Although outside the scope of this article, stem cells have demonstrated efficacy in the treatment of a number of osseous clinical entities. This includes the treatment of fracture nonunion, augmentation of spinal fusion, and assistance in the treatment of osteonecrosis.84

Summary

As a scientific community, our understanding of the use of stem cells, their nuances, and their indications has expanded dramatically over the last several years. Stem cell treatment has particularly infiltrated the world of operative and nonoperative sports medicine, given in part the active patient population seeking greater levels of improvement.85 Stem cell therapy offers a potentially effective therapy for a multitude of pathologies because of these cells’ anti-inflammatory, immunoregulatory, angiogenic, and paracrine effects.86 It thus remains a very dynamic option in the study of musculoskeletal tissue regeneration. While the potential exists for stem cell use in daily surgery practices, it is still premature to predict whether this can be expected.

The ideal stem cell sources (including allogeneic or autologous), preparation, cell number, timing, and means of application continue to be evaluated, as well as those advantageous pathologies that can benefit from the technology. In order to better answer these pertinent questions, we need to make sure we have a safe, economic, and ethically acceptable means for stem cell translational research efforts. More high-level studies with standardized protocols need to be performed. It is necessary to improve national and international collaboration in research, as well as collaboration with governing bodies, to attempt to further scientific advancement in this field of research.49 Further study on embryonic stem cell use may be valuable as well, pending governmental approval. Finally, more dedicated research efforts must be placed on the utility of adjuncts with stem cell use, including PRP and scaffolds, which may increase protection, nutritional support, and mechanical stimulation of the administered stem cells.

References

1.    Oh JH, Chung SW, Kim SH, Chung JY, Kim JY. 2013 Neer Award: Effect of the adipose-derived stem cell for the improvement of fatty degeneration and rotator cuff healing in rabbit model. J Shoulder Elb Surg. 2014;23(4):445-455.

2.    Caplan AI, Correa D. PDGF in bone formation and regeneration: new insights into a novel mechanism involving MSCs. J Orthop Res. 2011;29(12):1795-1803.

3.    Jang KM, Lim HC, Jung WY, Moon SW, Wang JH. Efficacy and safety of human umbilical cord blood-derived mesenchymal stem cells in anterior cruciate ligament reconstruction of a rabbit model: new strategy to enhance tendon graft healing. Arthroscopy. 2015;31(8):1530-1539.

4.    Muttini A, Salini V, Valbonetti L, Abate M. Stem cell therapy of tendinopathies: suggestions from veterinary medicine. Muscles Ligaments Tendons J. 2012;2(3):187-192.

5.    Xia P, Wang X, Lin Q, Li X. Efficacy of mesenchymal stem cells injection for the management of knee osteoarthritis: a systematic review and meta-analysis. Int Orthop. 2015;39(12):2363-2372.

6.    Veronesi F, Giavaresi G, Tschon M, Borsari V, Nicoli Aldini N, Fini M. Clinical use of bone marrow, bone marrow concentrate, and expanded bone marrow mesenchymal stem cells in cartilage disease. Stem Cells Dev. 2013;22(2):181-192.

7.    Caplan AI. Review: mesenchymal stem cells: cell-based reconstructive therapy in orthopedics. Tissue Eng. 2005;11(7-8):1198-1211.

8.    Hirzinger C, Tauber M, Korntner S, et al. ACL injuries and stem cell therapy. Arch Orthop Trauma Surg. 2014;134(11):1573-1578.

9.    Becerra P, Valdés Vázquez MA, Dudhia J, et al. Distribution of injected technetium(99m)-labeled mesenchymal stem cells in horses with naturally occurring tendinopathy. J Orthop Res. 2013;31(7):1096-1102.

10.  Lodi D, Iannitti T, Palmieri B. Stem cells in clinical practice: applications and warnings. J Exp Clin Cancer Res. 2011;30:9.

11.  García-Gómez I, Elvira G, Zapata AG, et al. Mesenchymal stem cells: biological properties and clinical applications. Expert Opin Biol Ther. 2010;10(10):1453-1468.

12.  Centeno CJ, Schultz JR, Cheever M, et al. Safety and complications reporting update on the re-implantation of culture-expanded mesenchymal stem cells using autologous platelet lysate technique. Curr Stem Cell Res Ther. 2011;6(4):368-378.

13.  Centeno CJ, Al-Sayegh H, Freeman MD, Smith J, Murrell WD, Bubnov R. A multi-center analysis of adverse events among two thousand, three hundred and seventy two adult patients undergoing adult autologous stem cell therapy for orthopaedic conditions. Int Orthop. 2016 Mar 30. [Epub ahead of print]

14.  Schmitt A, van Griensven M, Imhoff AB, Buchmann S. Application of stem cells in orthopedics. Stem Cells Int. 2012;2012:394962.

15.  Tuan RS, Boland G, Tuli R. Adult mesenchymal stem cells and cell-based tissue engineering. Arthritis Res Ther. 2003;5(1):32-45.

16.  Anz AW, Hackel JG, Nilssen EC, Andrews JR. Application of biologics in the treatment of the rotator cuff, meniscus, cartilage, and osteoarthritis. J Am Acad Orthop Surg. 2014;22(2):68-79.

17.  Dominici M, Le Blanc K, Mueller I, et al. Minimal criteria for defining multipotent mesenchymal stromal cells. The International Society for Cellular Therapy position statement. Cytotherapy. 2006;8(4):315-317.

18.  Porada CD, Almeida-Porada G. Mesenchymal stem cells as therapeutics and vehicles for gene and drug delivery. Adv Drug Deliv Rev. 2010;62(12):1156-1566.

19.  Filardo G, Madry H, Jelic M, Roffi A, Cucchiarini M, Kon E. Mesenchymal stem cells for the treatment of cartilage lesions: from preclinical findings to clinical application in orthopaedics. Knee Surg Sports Traumatol Arthrosc. 2013;21(8):1717-1729.

20.  Liechty KW, MacKenzie TC, Shaaban AF, et al. Human mesenchymal stem cells engraft and demonstrate site-specific differentiation after in utero transplantation in sheep. Nat Med. 2000;6(11):1282-1286.

21.  Hung SC, Chen NJ, Hsieh SL, Li H, Ma HL, Lo WH. Isolation and characterization of size-sieved stem cells from human bone marrow. Stem Cells. 2002;20(3):249-258.

22.  De Bari C, Dell’Accio F, Vanlauwe J, et al. Mesenchymal multipotency of adult human periosteal cells demonstrated by single-cell lineage analysis. Arthritis Rheum. 2006;54(4):1209-1221.

23.  Zuk PA, Zhu M, Ashjian P, et al. Human adipose tissue is a source of multipotent stem cells. Mol Biol Cell. 2002;13(12):4279-4295.

24.  Mafi R, Hindocha S, Mafi P, Griffin M, Khan WS. Sources of adult mesenchymal stem cells applicable for musculoskeletal applications - a systematic review of the literature. Open Orthop J. 2011;5 Suppl 2:242-248.

25.  Frisbie DD, Kisiday JD, Kawcak CE, Werpy NM, McIlwraith CW. Evaluation of adipose-derived stromal vascular fraction or bone marrow-derived mesenchymal stem cells for treatment of osteoarthritis. J Orthop Res. 2009;27(12):1675-1680.

26.  Vidal MA, Robinson SO, Lopez MJ, et al. Comparison of chondrogenic potential in equine mesenchymal stromal cells derived from adipose tissue and bone marrow. Vet Surg. 2008;37(8):713-724.

27.  Yoshimura H, Muneta T, Nimura A, Yokoyama A, Koga H, Sekiya I. Comparison of rat mesenchymal stem cells derived from bone marrow, synovium, periosteum, adipose tissue, and muscle. Cell Tissue Res. 2007;327(3):449-462.

28.  Hogan MV, Walker GN, Cui LR, Fu FH, Huard J. The role of stem cells and tissue engineering in orthopaedic sports medicine: current evidence and future directions. Arthroscopy. 2015;31(5):1017-1021.

29.  Koh YG, Choi YJ, Kwon SK, Kim YS, Yeo JE. Clinical results and second-look arthroscopic findings after treatment with adipose-derived stem cells for knee osteoarthritis. Knee Surg Sports Traumatol Arthrosc. 2015;23(5):1308-1316.

30.  Vangsness CT Jr, Farr J 2nd, Boyd J, Dellaero DT, Mills CR, LeRoux-Williams M. Adult human mesenchymal stem cells delivered via intra-articular injection to the knee following partial medial meniscectomy: a randomized, double-blind, controlled study. J Bone Joint Surg Am. 2014;96(2):90-98.

31.  Goodrich LR, Chen AC, Werpy NM, et al. Addition of mesenchymal stem cells to autologous platelet-enhanced fibrin scaffolds in chondral defects: does it enhance repair? J Bone Joint Surg Am. 2016;98(1):23-34.

32.  Kim YS, Choi YJ, Suh DS, et al. Mesenchymal stem cell implantation in osteoarthritic knees: is fibrin glue effective as a scaffold? Am J Sports Med. 2015;43(1):176-185.

33.  Steinert AF, Rackwitz L, Gilbert F, Nöth U, Tuan RS. Concise review: the clinical application of mesenchymal stem cells for musculoskeletal regeneration: current status and perspectives. Stem Cells Transl Med. 2012;1(3):237-247.

34.  Pers YM, Ruiz M, Noël D, Jorgensen C. Mesenchymal stem cells for the management of inflammation in osteoarthritis: state of the art and perspectives. Osteoarthritis Cartilage. 2015;23(11):2027-2035.

35.  Mamidi MK, Das AK, Zakaria Z, Bhonde R. Mesenchymal stromal cells for cartilage repair in osteoarthritis. Osteoarthritis Cartilage. 2016 Mar 10. [Epub ahead of print]

36.  Wyles CC, Houdek MT, Behfar A, Sierra RJ. Mesenchymal stem cell therapy for osteoarthritis: current perspectives. Stem Cells Cloning. 2015;8:117-124.

37.  Wong KL, Lee KB, Tai BC, Law P, Lee EH, Hui JH. Injectable cultured bone marrow-derived mesenchymal stem cells in varus knees with cartilage defects undergoing high tibial osteotomy: a prospective, randomized controlled clinical trial with 2 years’ follow-up. Arthroscopy. 2013;29(12):2020-2028.

38.  Vega A, Martín-Ferrero MA, Del Canto F, et al. Treatment of knee osteoarthritis with allogeneic bone marrow mesenchymal stem cells: a randomized controlled trial. Transplantation. 2015;99(8):1681-1690.

39.  Kim YS, Lee M, Koh YG. Additional mesenchymal stem cell injection improves the outcomes of marrow stimulation combined with supramalleolar osteotomy in varus ankle osteoarthritis: short-term clinical results with second-look arthroscopic evaluation. J Exp Orthop. 2016;3(1):12.

40.  Kraus TM, Imhoff FB, Reinert J, et al. Stem cells and bFGF in tendon healing: Effects of lentiviral gene transfer and long-term follow-up in a rat Achilles tendon defect model. BMC Musculoskelet Disord. 2016;17(1):148.

41.  Thomopoulos S, Parks WC, Rifkin DB, Derwin KA. Mechanisms of tendon injury and repair. J Orthop Res. 2015;33(6):832-839.

42.  Müller SA, Todorov A, Heisterbach PE, Martin I, Majewski M. Tendon healing: an overview of physiology, biology, and pathology of tendon healing and systematic review of state of the art in tendon bioengineering. Knee Surg Sports Traumatol Arthrosc. 2015;23(7):2097-3105.

43.  Godwin EE, Young NJ, Dudhia J, Beamish IC, Smith RK. Implantation of bone marrow-derived mesenchymal stem cells demonstrates improved outcome in horses with overstrain injury of the superficial digital flexor tendon. Equine Vet J. 2012;44(1):25-32.

44.  Machova Urdzikova L, Sedlacek R, Suchy T, et al. Human multipotent mesenchymal stem cells improve healing after collagenase tendon injury in the rat. Biomed Eng Online. 2014;13:42.

45.  Oshita T, Tobita M, Tajima S, Mizuno H. Adipose-derived stem cells improve collagenase-induced tendinopathy in a rat model. Am J Sports Med. 2016 Apr 11. [Epub ahead of print]

46.  Yuksel S, Guleç MA, Gultekin MZ, et al. Comparison of the early-period effects of bone marrow-derived mesenchymal stem cells and platelet-rich plasma on achilles tendon ruptures in rats. Connect Tissue Res. 2016 May 18. [Epub ahead of print]

47.  Chen L, Liu JP, Tang KL, et al. Tendon derived stem cells promote platelet-rich plasma healing in collagenase-induced rat achilles tendinopathy. Cell Physiol Biochem. 2014;34(6):2153-2168.

48.  Pascual-Garrido C, Rolón A, Makino A. Treatment of chronic patellar tendinopathy with autologous bone marrow stem cells: a 5-year-followup. Stem Cells Int. 2012;2012:953510.

49.  Zlotnicki JP, Geeslin AG, Murray IR, et al. Biologic treatments for sports injuries ii think tank-current concepts, future research, and barriers to advancement, part 3: articular cartilage. Orthop J Sports Med. 2016;4(4):2325967116642433.

50.  McCormack RA, Shreve M, Strauss EJ. Biologic augmentation in rotator cuff repair--should we do it, who should get it, and has it worked? Bull Hosp Jt Dis (2013). 2014;72(1):89-96.

51.  Mosna F, Sensebé L, Krampera M. Human bone marrow and adipose tissue mesenchymal stem cells: a user’s guide. Stem Cells Dev. 2010;19(10):1449-1470.

52.  Nakamura T, Sekiya I, Muneta T, et al. Arthroscopic, histological and MRI analyses of cartilage repair after a minimally invasive method of transplantation of allogeneic synovial mesenchymal stromal cells into cartilage defects in pigs. Cytotherapy. 2012;14(3):327-338.

53.  Dragoo JL, Carlson G, McCormick F, et al. Healing full-thickness cartilage defects using adipose-derived stem cells. Tissue Eng. 2007;13(7):1615-1621.

54.  Masuoka K, Asazuma T, Hattori H, et al. Tissue engineering of articular cartilage with autologous cultured adipose tissue-derived stromal cells using atelocollagen honeycomb-shaped scaffold with a membrane sealing in rabbits. J Biomed Mater Res B Appl Biomater. 2006 79(1):25-34.

55.  Gobbi A, Karnatzikos G, Sankineani SR. One-step surgery with multipotent stem cells for the treatment of large full-thickness chondral defects of the knee. Am J Sports Med. 2014;42(3):648-657.

56.  Kim JD, Lee GW, Jung GH, et al. Clinical outcome of autologous bone marrow aspirates concentrate (BMAC) injection in degenerative arthritis of the knee. Eur J Orthop Surg Traumatol. 2014;24(8):1505-1511.

57.  Krych AJ, Nawabi DH, Farshad-Amacker NA, et al. Bone marrow concentrate improves early cartilage phase maturation of a scaffold plug in the knee: a comparative magnetic resonance imaging analysis to platelet-rich plasma and control. Am J Sports Med. 2016;44(1):91-98.

58.  Fortier LA, Potter HG, Rickey EJ, et al. Concentrated bone marrow aspirate improves full-thickness cartilage repair compared with microfracture in the equine model. J Bone Joint Surg Am. 2010;92(10):1927-1937.

59.  Kim YS, Park EH, Kim YC, Koh YG. Clinical outcomes of mesenchymal stem cell injection with arthroscopic treatment in older patients with osteochondral lesions of the talus. Am J Sports Med. 2013;41(5):1090-1099.

60.  Kim YS, Lee HJ, Choi YJ, Kim YI, Koh YG. Does an injection of a stromal vascular fraction containing adipose-derived mesenchymal stem cells influence the outcomes of marrow stimulation in osteochondral lesions of the talus? A clinical and magnetic resonance imaging study. Am J Sports Med. 2014;42(10):2424-2434.

61.  Chahla J, Dean CS, Moatshe G, Pascual-Garrido C, Serra Cruz R, LaPrade RF. Concentrated bone marrow aspirate for the treatment of chondral injuries and osteoarthritis of the knee: a systematic review of outcomes. Orthop J Sports Med. 2016;4(1):2325967115625481.

62.  Gobbi A, Karnatzikos G, Scotti C, Mahajan V, Mazzucco L, Grigolo B. One-step cartilage repair with bone marrow aspirate concentrated cells and collagen matrix in full-thickness knee cartilage lesions: results at 2-year follow-up. Cartilage. 2011;2(3):286-299.

63.  Ruiz-Ibán MÁ, Díaz-Heredia J, García-Gómez I, Gonzalez-Lizán F, Elías-Martín E, Abraira V. The effect of the addition of adipose-derived mesenchymal stem cells to a meniscal repair in the avascular zone: an experimental study in rabbits. Arthroscopy. 2011;27(12):1688-1696.

64.  Angele P, Johnstone B, Kujat R, et al. Stem cell based tissue engineering for meniscus repair. J Biomed Mater Res A. 2008;85(2):445-455.

65.  Hernigou P, Merouse G, Duffiet P, Chevalier N, Rouard H. Reduced levels of mesenchymal stem cells at the tendon-bone interface tuberosity in patients with symptomatic rotator cuff tear. Int Orthop. 2015;39(6):1219-1225.

66.  Goutallier D, Postel JM, Gleyze P, Leguilloux P, Van Driessche S. Influence of cuff muscle fatty degeneration on anatomic and functional outcomes after simple suture of full-thickness tears. J Shoulder Elbow Surg. 2003;12(6):550-554.

67.   Kovacevic D, Rodeo SA. Biological augmentation of rotator cuff tendon repair. Clin Orthop Relat Res. 2008;466(3):622-633.

68.   Valencia Mora M, Antuña Antuña S, García Arranz M, Carrascal MT, Barco R. Application of adipose tissue-derived stem cells in a rat rotator cuff repair model. Injury. 2014;45 Suppl 4:S22-S27.

69.  Yokoya S, Mochizuki Y, Natsu K, Omae H, Nagata Y, Ochi M. Rotator cuff regeneration using a bioabsorbable material with bone marrow-derived mesenchymal stem cells in a rabbit model. Am J Sports Med. 2012;40(6):1259-1268.

70.  Gulotta LV, Kovacevic D, Ehteshami JR, Dagher E, Packer JD, Rodeo SA. Application of bone marrow-derived mesenchymal stem cells in a rotator cuff repair model. Am J Sports Med. 2009;37(11):2126-2133.

71.  Beitzel K, McCarthy MB, Cote MP, et al. Comparison of mesenchymal stem cells (osteoprogenitors) harvested from proximal humerus and distal femur during arthroscopic surgery. Arthroscopy. 2013;29(2):301-308.

72.  Utsunomiya H, Uchida S, Sekiya I, Sakai A, Moridera K, Nakamura T. Isolation and characterization of human mesenchymal stem cells derived from shoulder tissues involved in rotator cuff tears. Am J Sports Med. 2013;41(3):657-668.

73.  Hernigou P, Flouzat Lachaniette CH, Delambre J, et al. Biologic augmentation of rotator cuff repair with mesenchymal stem cells during arthroscopy improves healing and prevents further tears: a case-controlled study. Int Orthop. 2014;38(9):1811-1818.

74.  Ellera Gomes JL, da Silva RC, Silla LM, Abreu MR, Pellanda R. Conventional rotator cuff repair complemented by the aid of mononuclear autologous stem cells. Knee Surg Sports Traumatol Arthrosc. 2012;20(2):373-377.

75.  Adams SB Jr, Thorpe MA, Parks BG, Aghazarian G, Allen E, Schon LC. Stem cell-bearing suture improves Achilles tendon healing in a rat model. Foot Ankle Int. 2014;35(3):293-299.

76.  Huang TF, Yew TL, Chiang ER, et al. Mesenchymal stem cells from a hypoxic culture improve and engraft Achilles tendon repair. Am J Sports Med. 2013;41(5):1117-1125.

77.  Yao J, Woon CY, Behn A, et al. The effect of suture coated with mesenchymal stem cells and bioactive substrate on tendon repair strength in a rat model. J Hand Surg Am. 2012;37(8):1639-1645.

78.  Uysal CA, Tobita M, Hyakusoku H, Mizuno H. Adipose-derived stem cells enhance primary tendon repair: biomechanical and immunohistochemical evaluation. J Plast Reconstr Aesthet Surg. 2012;65(12):1712-1719.

79.  Stein BE, Stroh DA, Schon LC. Outcomes of acute Achilles tendon rupture repair with bone marrow aspirate concentrate augmentation. Int Orthop. 2015;39(5):901-905.

80.  Chen B, Li B, Qi YJ, et al. Enhancement of tendon-to-bone healing after anterior cruciate ligament reconstruction using bone marrow-derived mesenchymal stem cells genetically modified with bFGF/BMP2. Sci Rep. 2016;6:25940.

81.  Kanaya A, Deie M, Adachi N, Nishimori M, Yanada S, Ochi M. Intra-articular injection of mesenchymal stromal cells in partially torn anterior cruciate ligaments in a rat model. Arthroscopy. 2007;23(6):610-617.

82.  Figueroa D, Espinosa M, Calvo R, et al. Anterior cruciate ligament regeneration using mesenchymal stem cells and collagen type I scaffold in a rabbit model. Knee Surg Sports Traumatol Arthrosc. 2014;22(5):1196-1202.

83.  Silva A, Sampaio R, Fernandes R, Pinto E. Is there a role for adult non-cultivated bone marrow stem cells in ACL reconstruction? Knee Surg Sports Traumatol Arthrosc. 2014;22(1):66-71.

84.  Pepke W, Kasten P, Beckmann NA, Janicki P, Egermann M. Core decompression and autologous bone marrow concentrate for treatment of femoral head osteonecrosis: a randomized prospective study. Orthop Rev (Pavia). 2016;8(1):6162.

85.   Kopka M, Bradley JP.  The use of biologic agents in athletes with knee injuries. J Knee Surg. 2016 May 20. [Epub ahead of print]

86.  Valencia Mora M, Ruiz Ibán MA, Díaz Heredia J, Barco Laakso R, Cuéllar R, García Arranz M. Stem cell therapy in the management of shoulder rotator cuff disorders. World J Stem Cells. 2015;7(4):691-699.

87.  Johnstone B, Hering TM, Caplan AI, Goldberg VM, Yoo JU. In vitro chondrogenesis of bone marrow-derived mesenchymal progenitor cells. Exp Cell Res. 1998;238(1):265-272.

88.  Ferrari G, Cusella-De Angelis G, Coletta M, et al. Muscle regeneration by bone marrow-derived myogenic progenitors. Science. 1998;279(5356):1528-1530.

89.  Pittenger MF, Mackay AM, Beck SC, et al. Multilineage potential of adult human mesenchymal stem cells. Science. 1999;284(5411):143-147.

90.  Fukuda K. Molecular characterization of regenerated cardiomyocytes derived from adult mesenchymal stem cells. Congenit Anom (Kyoto). 2002;42(1):1-9.

91.  Ito T, Suzuki A, Okabe M, Imai E, Hori M. Application of bone marrow-derived stem cells in experimental nephrology. Exp Nephrol. 2001;9(6):444-450.

92.  Qu-Petersen Z, Deasy B, Jankowski R, et al. Identification of a novel population of muscle stem cells in mice: potential for muscle regeneration. J Cell Biol. 2002;157(5):851-864.

93.  Shi S, Gronthos S, Chen S, et al. Bone formation by human postnatal bone marrow stromal stem cells is enhanced by telomerase expression. Nat Biotechnol. 2002;20(6):587-591.

94.  Deans TL, Elisseeff JH. Stem cells in musculoskeletal engineered tissue. Curr Opin Biotechnol. 2009;20(5):537-544.

95.  Funk JF, Matziolis G, Krocker D, Perka C. [Promotion of bone healing through clinical application of autologous periosteum derived stem cells in a case of atrophic non-union]. Z Orthop Unfall. 2007;145(6):790-794.

References

1.    Oh JH, Chung SW, Kim SH, Chung JY, Kim JY. 2013 Neer Award: Effect of the adipose-derived stem cell for the improvement of fatty degeneration and rotator cuff healing in rabbit model. J Shoulder Elb Surg. 2014;23(4):445-455.

2.    Caplan AI, Correa D. PDGF in bone formation and regeneration: new insights into a novel mechanism involving MSCs. J Orthop Res. 2011;29(12):1795-1803.

3.    Jang KM, Lim HC, Jung WY, Moon SW, Wang JH. Efficacy and safety of human umbilical cord blood-derived mesenchymal stem cells in anterior cruciate ligament reconstruction of a rabbit model: new strategy to enhance tendon graft healing. Arthroscopy. 2015;31(8):1530-1539.

4.    Muttini A, Salini V, Valbonetti L, Abate M. Stem cell therapy of tendinopathies: suggestions from veterinary medicine. Muscles Ligaments Tendons J. 2012;2(3):187-192.

5.    Xia P, Wang X, Lin Q, Li X. Efficacy of mesenchymal stem cells injection for the management of knee osteoarthritis: a systematic review and meta-analysis. Int Orthop. 2015;39(12):2363-2372.

6.    Veronesi F, Giavaresi G, Tschon M, Borsari V, Nicoli Aldini N, Fini M. Clinical use of bone marrow, bone marrow concentrate, and expanded bone marrow mesenchymal stem cells in cartilage disease. Stem Cells Dev. 2013;22(2):181-192.

7.    Caplan AI. Review: mesenchymal stem cells: cell-based reconstructive therapy in orthopedics. Tissue Eng. 2005;11(7-8):1198-1211.

8.    Hirzinger C, Tauber M, Korntner S, et al. ACL injuries and stem cell therapy. Arch Orthop Trauma Surg. 2014;134(11):1573-1578.

9.    Becerra P, Valdés Vázquez MA, Dudhia J, et al. Distribution of injected technetium(99m)-labeled mesenchymal stem cells in horses with naturally occurring tendinopathy. J Orthop Res. 2013;31(7):1096-1102.

10.  Lodi D, Iannitti T, Palmieri B. Stem cells in clinical practice: applications and warnings. J Exp Clin Cancer Res. 2011;30:9.

11.  García-Gómez I, Elvira G, Zapata AG, et al. Mesenchymal stem cells: biological properties and clinical applications. Expert Opin Biol Ther. 2010;10(10):1453-1468.

12.  Centeno CJ, Schultz JR, Cheever M, et al. Safety and complications reporting update on the re-implantation of culture-expanded mesenchymal stem cells using autologous platelet lysate technique. Curr Stem Cell Res Ther. 2011;6(4):368-378.

13.  Centeno CJ, Al-Sayegh H, Freeman MD, Smith J, Murrell WD, Bubnov R. A multi-center analysis of adverse events among two thousand, three hundred and seventy two adult patients undergoing adult autologous stem cell therapy for orthopaedic conditions. Int Orthop. 2016 Mar 30. [Epub ahead of print]

14.  Schmitt A, van Griensven M, Imhoff AB, Buchmann S. Application of stem cells in orthopedics. Stem Cells Int. 2012;2012:394962.

15.  Tuan RS, Boland G, Tuli R. Adult mesenchymal stem cells and cell-based tissue engineering. Arthritis Res Ther. 2003;5(1):32-45.

16.  Anz AW, Hackel JG, Nilssen EC, Andrews JR. Application of biologics in the treatment of the rotator cuff, meniscus, cartilage, and osteoarthritis. J Am Acad Orthop Surg. 2014;22(2):68-79.

17.  Dominici M, Le Blanc K, Mueller I, et al. Minimal criteria for defining multipotent mesenchymal stromal cells. The International Society for Cellular Therapy position statement. Cytotherapy. 2006;8(4):315-317.

18.  Porada CD, Almeida-Porada G. Mesenchymal stem cells as therapeutics and vehicles for gene and drug delivery. Adv Drug Deliv Rev. 2010;62(12):1156-1566.

19.  Filardo G, Madry H, Jelic M, Roffi A, Cucchiarini M, Kon E. Mesenchymal stem cells for the treatment of cartilage lesions: from preclinical findings to clinical application in orthopaedics. Knee Surg Sports Traumatol Arthrosc. 2013;21(8):1717-1729.

20.  Liechty KW, MacKenzie TC, Shaaban AF, et al. Human mesenchymal stem cells engraft and demonstrate site-specific differentiation after in utero transplantation in sheep. Nat Med. 2000;6(11):1282-1286.

21.  Hung SC, Chen NJ, Hsieh SL, Li H, Ma HL, Lo WH. Isolation and characterization of size-sieved stem cells from human bone marrow. Stem Cells. 2002;20(3):249-258.

22.  De Bari C, Dell’Accio F, Vanlauwe J, et al. Mesenchymal multipotency of adult human periosteal cells demonstrated by single-cell lineage analysis. Arthritis Rheum. 2006;54(4):1209-1221.

23.  Zuk PA, Zhu M, Ashjian P, et al. Human adipose tissue is a source of multipotent stem cells. Mol Biol Cell. 2002;13(12):4279-4295.

24.  Mafi R, Hindocha S, Mafi P, Griffin M, Khan WS. Sources of adult mesenchymal stem cells applicable for musculoskeletal applications - a systematic review of the literature. Open Orthop J. 2011;5 Suppl 2:242-248.

25.  Frisbie DD, Kisiday JD, Kawcak CE, Werpy NM, McIlwraith CW. Evaluation of adipose-derived stromal vascular fraction or bone marrow-derived mesenchymal stem cells for treatment of osteoarthritis. J Orthop Res. 2009;27(12):1675-1680.

26.  Vidal MA, Robinson SO, Lopez MJ, et al. Comparison of chondrogenic potential in equine mesenchymal stromal cells derived from adipose tissue and bone marrow. Vet Surg. 2008;37(8):713-724.

27.  Yoshimura H, Muneta T, Nimura A, Yokoyama A, Koga H, Sekiya I. Comparison of rat mesenchymal stem cells derived from bone marrow, synovium, periosteum, adipose tissue, and muscle. Cell Tissue Res. 2007;327(3):449-462.

28.  Hogan MV, Walker GN, Cui LR, Fu FH, Huard J. The role of stem cells and tissue engineering in orthopaedic sports medicine: current evidence and future directions. Arthroscopy. 2015;31(5):1017-1021.

29.  Koh YG, Choi YJ, Kwon SK, Kim YS, Yeo JE. Clinical results and second-look arthroscopic findings after treatment with adipose-derived stem cells for knee osteoarthritis. Knee Surg Sports Traumatol Arthrosc. 2015;23(5):1308-1316.

30.  Vangsness CT Jr, Farr J 2nd, Boyd J, Dellaero DT, Mills CR, LeRoux-Williams M. Adult human mesenchymal stem cells delivered via intra-articular injection to the knee following partial medial meniscectomy: a randomized, double-blind, controlled study. J Bone Joint Surg Am. 2014;96(2):90-98.

31.  Goodrich LR, Chen AC, Werpy NM, et al. Addition of mesenchymal stem cells to autologous platelet-enhanced fibrin scaffolds in chondral defects: does it enhance repair? J Bone Joint Surg Am. 2016;98(1):23-34.

32.  Kim YS, Choi YJ, Suh DS, et al. Mesenchymal stem cell implantation in osteoarthritic knees: is fibrin glue effective as a scaffold? Am J Sports Med. 2015;43(1):176-185.

33.  Steinert AF, Rackwitz L, Gilbert F, Nöth U, Tuan RS. Concise review: the clinical application of mesenchymal stem cells for musculoskeletal regeneration: current status and perspectives. Stem Cells Transl Med. 2012;1(3):237-247.

34.  Pers YM, Ruiz M, Noël D, Jorgensen C. Mesenchymal stem cells for the management of inflammation in osteoarthritis: state of the art and perspectives. Osteoarthritis Cartilage. 2015;23(11):2027-2035.

35.  Mamidi MK, Das AK, Zakaria Z, Bhonde R. Mesenchymal stromal cells for cartilage repair in osteoarthritis. Osteoarthritis Cartilage. 2016 Mar 10. [Epub ahead of print]

36.  Wyles CC, Houdek MT, Behfar A, Sierra RJ. Mesenchymal stem cell therapy for osteoarthritis: current perspectives. Stem Cells Cloning. 2015;8:117-124.

37.  Wong KL, Lee KB, Tai BC, Law P, Lee EH, Hui JH. Injectable cultured bone marrow-derived mesenchymal stem cells in varus knees with cartilage defects undergoing high tibial osteotomy: a prospective, randomized controlled clinical trial with 2 years’ follow-up. Arthroscopy. 2013;29(12):2020-2028.

38.  Vega A, Martín-Ferrero MA, Del Canto F, et al. Treatment of knee osteoarthritis with allogeneic bone marrow mesenchymal stem cells: a randomized controlled trial. Transplantation. 2015;99(8):1681-1690.

39.  Kim YS, Lee M, Koh YG. Additional mesenchymal stem cell injection improves the outcomes of marrow stimulation combined with supramalleolar osteotomy in varus ankle osteoarthritis: short-term clinical results with second-look arthroscopic evaluation. J Exp Orthop. 2016;3(1):12.

40.  Kraus TM, Imhoff FB, Reinert J, et al. Stem cells and bFGF in tendon healing: Effects of lentiviral gene transfer and long-term follow-up in a rat Achilles tendon defect model. BMC Musculoskelet Disord. 2016;17(1):148.

41.  Thomopoulos S, Parks WC, Rifkin DB, Derwin KA. Mechanisms of tendon injury and repair. J Orthop Res. 2015;33(6):832-839.

42.  Müller SA, Todorov A, Heisterbach PE, Martin I, Majewski M. Tendon healing: an overview of physiology, biology, and pathology of tendon healing and systematic review of state of the art in tendon bioengineering. Knee Surg Sports Traumatol Arthrosc. 2015;23(7):2097-3105.

43.  Godwin EE, Young NJ, Dudhia J, Beamish IC, Smith RK. Implantation of bone marrow-derived mesenchymal stem cells demonstrates improved outcome in horses with overstrain injury of the superficial digital flexor tendon. Equine Vet J. 2012;44(1):25-32.

44.  Machova Urdzikova L, Sedlacek R, Suchy T, et al. Human multipotent mesenchymal stem cells improve healing after collagenase tendon injury in the rat. Biomed Eng Online. 2014;13:42.

45.  Oshita T, Tobita M, Tajima S, Mizuno H. Adipose-derived stem cells improve collagenase-induced tendinopathy in a rat model. Am J Sports Med. 2016 Apr 11. [Epub ahead of print]

46.  Yuksel S, Guleç MA, Gultekin MZ, et al. Comparison of the early-period effects of bone marrow-derived mesenchymal stem cells and platelet-rich plasma on achilles tendon ruptures in rats. Connect Tissue Res. 2016 May 18. [Epub ahead of print]

47.  Chen L, Liu JP, Tang KL, et al. Tendon derived stem cells promote platelet-rich plasma healing in collagenase-induced rat achilles tendinopathy. Cell Physiol Biochem. 2014;34(6):2153-2168.

48.  Pascual-Garrido C, Rolón A, Makino A. Treatment of chronic patellar tendinopathy with autologous bone marrow stem cells: a 5-year-followup. Stem Cells Int. 2012;2012:953510.

49.  Zlotnicki JP, Geeslin AG, Murray IR, et al. Biologic treatments for sports injuries ii think tank-current concepts, future research, and barriers to advancement, part 3: articular cartilage. Orthop J Sports Med. 2016;4(4):2325967116642433.

50.  McCormack RA, Shreve M, Strauss EJ. Biologic augmentation in rotator cuff repair--should we do it, who should get it, and has it worked? Bull Hosp Jt Dis (2013). 2014;72(1):89-96.

51.  Mosna F, Sensebé L, Krampera M. Human bone marrow and adipose tissue mesenchymal stem cells: a user’s guide. Stem Cells Dev. 2010;19(10):1449-1470.

52.  Nakamura T, Sekiya I, Muneta T, et al. Arthroscopic, histological and MRI analyses of cartilage repair after a minimally invasive method of transplantation of allogeneic synovial mesenchymal stromal cells into cartilage defects in pigs. Cytotherapy. 2012;14(3):327-338.

53.  Dragoo JL, Carlson G, McCormick F, et al. Healing full-thickness cartilage defects using adipose-derived stem cells. Tissue Eng. 2007;13(7):1615-1621.

54.  Masuoka K, Asazuma T, Hattori H, et al. Tissue engineering of articular cartilage with autologous cultured adipose tissue-derived stromal cells using atelocollagen honeycomb-shaped scaffold with a membrane sealing in rabbits. J Biomed Mater Res B Appl Biomater. 2006 79(1):25-34.

55.  Gobbi A, Karnatzikos G, Sankineani SR. One-step surgery with multipotent stem cells for the treatment of large full-thickness chondral defects of the knee. Am J Sports Med. 2014;42(3):648-657.

56.  Kim JD, Lee GW, Jung GH, et al. Clinical outcome of autologous bone marrow aspirates concentrate (BMAC) injection in degenerative arthritis of the knee. Eur J Orthop Surg Traumatol. 2014;24(8):1505-1511.

57.  Krych AJ, Nawabi DH, Farshad-Amacker NA, et al. Bone marrow concentrate improves early cartilage phase maturation of a scaffold plug in the knee: a comparative magnetic resonance imaging analysis to platelet-rich plasma and control. Am J Sports Med. 2016;44(1):91-98.

58.  Fortier LA, Potter HG, Rickey EJ, et al. Concentrated bone marrow aspirate improves full-thickness cartilage repair compared with microfracture in the equine model. J Bone Joint Surg Am. 2010;92(10):1927-1937.

59.  Kim YS, Park EH, Kim YC, Koh YG. Clinical outcomes of mesenchymal stem cell injection with arthroscopic treatment in older patients with osteochondral lesions of the talus. Am J Sports Med. 2013;41(5):1090-1099.

60.  Kim YS, Lee HJ, Choi YJ, Kim YI, Koh YG. Does an injection of a stromal vascular fraction containing adipose-derived mesenchymal stem cells influence the outcomes of marrow stimulation in osteochondral lesions of the talus? A clinical and magnetic resonance imaging study. Am J Sports Med. 2014;42(10):2424-2434.

61.  Chahla J, Dean CS, Moatshe G, Pascual-Garrido C, Serra Cruz R, LaPrade RF. Concentrated bone marrow aspirate for the treatment of chondral injuries and osteoarthritis of the knee: a systematic review of outcomes. Orthop J Sports Med. 2016;4(1):2325967115625481.

62.  Gobbi A, Karnatzikos G, Scotti C, Mahajan V, Mazzucco L, Grigolo B. One-step cartilage repair with bone marrow aspirate concentrated cells and collagen matrix in full-thickness knee cartilage lesions: results at 2-year follow-up. Cartilage. 2011;2(3):286-299.

63.  Ruiz-Ibán MÁ, Díaz-Heredia J, García-Gómez I, Gonzalez-Lizán F, Elías-Martín E, Abraira V. The effect of the addition of adipose-derived mesenchymal stem cells to a meniscal repair in the avascular zone: an experimental study in rabbits. Arthroscopy. 2011;27(12):1688-1696.

64.  Angele P, Johnstone B, Kujat R, et al. Stem cell based tissue engineering for meniscus repair. J Biomed Mater Res A. 2008;85(2):445-455.

65.  Hernigou P, Merouse G, Duffiet P, Chevalier N, Rouard H. Reduced levels of mesenchymal stem cells at the tendon-bone interface tuberosity in patients with symptomatic rotator cuff tear. Int Orthop. 2015;39(6):1219-1225.

66.  Goutallier D, Postel JM, Gleyze P, Leguilloux P, Van Driessche S. Influence of cuff muscle fatty degeneration on anatomic and functional outcomes after simple suture of full-thickness tears. J Shoulder Elbow Surg. 2003;12(6):550-554.

67.   Kovacevic D, Rodeo SA. Biological augmentation of rotator cuff tendon repair. Clin Orthop Relat Res. 2008;466(3):622-633.

68.   Valencia Mora M, Antuña Antuña S, García Arranz M, Carrascal MT, Barco R. Application of adipose tissue-derived stem cells in a rat rotator cuff repair model. Injury. 2014;45 Suppl 4:S22-S27.

69.  Yokoya S, Mochizuki Y, Natsu K, Omae H, Nagata Y, Ochi M. Rotator cuff regeneration using a bioabsorbable material with bone marrow-derived mesenchymal stem cells in a rabbit model. Am J Sports Med. 2012;40(6):1259-1268.

70.  Gulotta LV, Kovacevic D, Ehteshami JR, Dagher E, Packer JD, Rodeo SA. Application of bone marrow-derived mesenchymal stem cells in a rotator cuff repair model. Am J Sports Med. 2009;37(11):2126-2133.

71.  Beitzel K, McCarthy MB, Cote MP, et al. Comparison of mesenchymal stem cells (osteoprogenitors) harvested from proximal humerus and distal femur during arthroscopic surgery. Arthroscopy. 2013;29(2):301-308.

72.  Utsunomiya H, Uchida S, Sekiya I, Sakai A, Moridera K, Nakamura T. Isolation and characterization of human mesenchymal stem cells derived from shoulder tissues involved in rotator cuff tears. Am J Sports Med. 2013;41(3):657-668.

73.  Hernigou P, Flouzat Lachaniette CH, Delambre J, et al. Biologic augmentation of rotator cuff repair with mesenchymal stem cells during arthroscopy improves healing and prevents further tears: a case-controlled study. Int Orthop. 2014;38(9):1811-1818.

74.  Ellera Gomes JL, da Silva RC, Silla LM, Abreu MR, Pellanda R. Conventional rotator cuff repair complemented by the aid of mononuclear autologous stem cells. Knee Surg Sports Traumatol Arthrosc. 2012;20(2):373-377.

75.  Adams SB Jr, Thorpe MA, Parks BG, Aghazarian G, Allen E, Schon LC. Stem cell-bearing suture improves Achilles tendon healing in a rat model. Foot Ankle Int. 2014;35(3):293-299.

76.  Huang TF, Yew TL, Chiang ER, et al. Mesenchymal stem cells from a hypoxic culture improve and engraft Achilles tendon repair. Am J Sports Med. 2013;41(5):1117-1125.

77.  Yao J, Woon CY, Behn A, et al. The effect of suture coated with mesenchymal stem cells and bioactive substrate on tendon repair strength in a rat model. J Hand Surg Am. 2012;37(8):1639-1645.

78.  Uysal CA, Tobita M, Hyakusoku H, Mizuno H. Adipose-derived stem cells enhance primary tendon repair: biomechanical and immunohistochemical evaluation. J Plast Reconstr Aesthet Surg. 2012;65(12):1712-1719.

79.  Stein BE, Stroh DA, Schon LC. Outcomes of acute Achilles tendon rupture repair with bone marrow aspirate concentrate augmentation. Int Orthop. 2015;39(5):901-905.

80.  Chen B, Li B, Qi YJ, et al. Enhancement of tendon-to-bone healing after anterior cruciate ligament reconstruction using bone marrow-derived mesenchymal stem cells genetically modified with bFGF/BMP2. Sci Rep. 2016;6:25940.

81.  Kanaya A, Deie M, Adachi N, Nishimori M, Yanada S, Ochi M. Intra-articular injection of mesenchymal stromal cells in partially torn anterior cruciate ligaments in a rat model. Arthroscopy. 2007;23(6):610-617.

82.  Figueroa D, Espinosa M, Calvo R, et al. Anterior cruciate ligament regeneration using mesenchymal stem cells and collagen type I scaffold in a rabbit model. Knee Surg Sports Traumatol Arthrosc. 2014;22(5):1196-1202.

83.  Silva A, Sampaio R, Fernandes R, Pinto E. Is there a role for adult non-cultivated bone marrow stem cells in ACL reconstruction? Knee Surg Sports Traumatol Arthrosc. 2014;22(1):66-71.

84.  Pepke W, Kasten P, Beckmann NA, Janicki P, Egermann M. Core decompression and autologous bone marrow concentrate for treatment of femoral head osteonecrosis: a randomized prospective study. Orthop Rev (Pavia). 2016;8(1):6162.

85.   Kopka M, Bradley JP.  The use of biologic agents in athletes with knee injuries. J Knee Surg. 2016 May 20. [Epub ahead of print]

86.  Valencia Mora M, Ruiz Ibán MA, Díaz Heredia J, Barco Laakso R, Cuéllar R, García Arranz M. Stem cell therapy in the management of shoulder rotator cuff disorders. World J Stem Cells. 2015;7(4):691-699.

87.  Johnstone B, Hering TM, Caplan AI, Goldberg VM, Yoo JU. In vitro chondrogenesis of bone marrow-derived mesenchymal progenitor cells. Exp Cell Res. 1998;238(1):265-272.

88.  Ferrari G, Cusella-De Angelis G, Coletta M, et al. Muscle regeneration by bone marrow-derived myogenic progenitors. Science. 1998;279(5356):1528-1530.

89.  Pittenger MF, Mackay AM, Beck SC, et al. Multilineage potential of adult human mesenchymal stem cells. Science. 1999;284(5411):143-147.

90.  Fukuda K. Molecular characterization of regenerated cardiomyocytes derived from adult mesenchymal stem cells. Congenit Anom (Kyoto). 2002;42(1):1-9.

91.  Ito T, Suzuki A, Okabe M, Imai E, Hori M. Application of bone marrow-derived stem cells in experimental nephrology. Exp Nephrol. 2001;9(6):444-450.

92.  Qu-Petersen Z, Deasy B, Jankowski R, et al. Identification of a novel population of muscle stem cells in mice: potential for muscle regeneration. J Cell Biol. 2002;157(5):851-864.

93.  Shi S, Gronthos S, Chen S, et al. Bone formation by human postnatal bone marrow stromal stem cells is enhanced by telomerase expression. Nat Biotechnol. 2002;20(6):587-591.

94.  Deans TL, Elisseeff JH. Stem cells in musculoskeletal engineered tissue. Curr Opin Biotechnol. 2009;20(5):537-544.

95.  Funk JF, Matziolis G, Krocker D, Perka C. [Promotion of bone healing through clinical application of autologous periosteum derived stem cells in a case of atrophic non-union]. Z Orthop Unfall. 2007;145(6):790-794.

Issue
The American Journal of Orthopedics - 45(5)
Issue
The American Journal of Orthopedics - 45(5)
Page Number
280-288, 326
Page Number
280-288, 326
Publications
Publications
Topics
Article Type
Display Headline
Stem Cells in Orthopedics: A Comprehensive Guide for the General Orthopedist
Display Headline
Stem Cells in Orthopedics: A Comprehensive Guide for the General Orthopedist
Legacy Keywords
review, stem cells, biologics, guide, orthopedist, biologic adjuvants, treatment, biomechanical, cells, platelet-rich plasma, PRP, repair, saltzman, kuhns, weber, yanke, nho
Legacy Keywords
review, stem cells, biologics, guide, orthopedist, biologic adjuvants, treatment, biomechanical, cells, platelet-rich plasma, PRP, repair, saltzman, kuhns, weber, yanke, nho
Sections
Article Source

PURLs Copyright

Inside the Article

Article PDF Media

Current and Future Stem Cell Regulation: A Call to Action

Article Type
Changed
Thu, 09/19/2019 - 13:25
Display Headline
Current and Future Stem Cell Regulation: A Call to Action

The 2 cardinal properties of stem cells are the ability to self-renew and the ability to differentiate into distinctive end-stage cell types. The work of Caplan1 captured our early attention, with cells cultured from bone marrow differentiating into a number of different cell types of orthopedic interest. Our latest attention has been captured by the additional abilities of these cells to mobilize, monitor, and interact with their surrounding environment.2-4 In response to their environment, stem cells are able to release a broad spectrum of macromolecules with trophic, chemotactic, and immunomodulatory potential, which allows them to participate in injury response, tissue healing, and tissue regeneration.4 These cells are innate to the body’s monitoring, maintenance, repair, and stress response systems.2,4-11 Basic science and animal studies have illustrated the potential of cells with stem potential regardless of their environment/source of harvest.

Where Can We Get Stem Cells?

Cells with stem properties are present in many environmental niches, including the bone marrow, peripheral circulatory system, adipose tissue, synovial tissue, muscle tissue, and tendon tissue.12-15 A number of cell types with stem properties populate the bone marrow niche, including hematopoietic stem/progenitor cells (HSPC), perivascular stromal cells (PSC), endothelial stem cells (ESC), and immature cells with qualities like embryonal stem cells termed very small embryonal-like stem cells (VESL).12,15-19 All of these cells have stem properties and have been shown to differentiate to tissues of orthopedic interest.The interplay, interaction, and potential of these cell types is complex and incompletely understood.12,15-19 When bone marrow is aspirated for culturing purposes, it is unclear which cell line produces the plastic-adherent multipotent cells grown in culture, which are often referred to as mesenchymal stem cells (MSCs). Researches propose that HSPC and/or VESL circulate peripherally in small numbers but leave the bone marrow in certain mobilization instances and are important for the monitoring and maintenance of the majority of tissues in our bodies.5,16 Current clinical utilization of these cell types by the orthopedic community primarily utilizes point-of-care bone marrow aspiration and concentration, while the hematology oncology community mobilizes cells from the bone marrow to the blood stream with pharmaceutical agents and harvests cells via apheresis. Bone marrow aspiration produces variable numbers of stem cells, with studies ranging from 1 stem cell per mL of tissue collected to 300,000 stem cells per mL of tissue collected.20Mobilization and apheresis can produce large volumes of peripheral blood-derived cells with 600,000 HSPC per mL and 2.32 million PSC per mL of tissue collected.21

In adipose tissue, cells adherent to the abluminal side of blood vessels known as pericytes also carry stem qualities. Aspiration and processing of adipose tissue can access these stem cells, producing a product often referred to as stromal vascular fraction (SVF). Processing of lipoaspirate to create stromal vascular fraction requires mechanical or enzymatic processing. This also produces variable numbers of stem cells, with quantitative studies ranging from 5000 to 1.5 million stem cells per mL of tissue collected.20 Similar to adipose-derived stem cells, synovial-derived and muscle-derived stem cells also require mechanical or enzymatic processing. For applications where it is believed that a large number of cells is necessary, investigators often utilize culturing techniques for all sources with the exception of mobilization and apheresis harvest. As clinicians, 3 challenges have proven more important than which cell type to utilize: 1) patient-care logistics regarding collection and application; 2) the undefined dose-response curve regarding stem cell treatments; and 3) evolving government/community regulation.

Regulation of Stem Cell Therapies

The regulation of stem cell technologies is a double-edged sword for development. While loose regulation encourages clinical application and experimentation, patient safety and efficacy concerns are raised, and a technology’s worth is not proven before clinical application. Tight regulation temporarily hampers progress, yet ensures the proof of safety and efficacy prior to widespread implementation. Within the United States, the Food and Drug Administration (FDA) has tightened regulation, established precedent, and intervened in the ability of clinicians to utilize stem cell therapies in humans, through “warning letters,” “untitled letters,” and industry guidance documents.22-30

The FDA categorizes stem cell therapies as human cells, tissues, and cellular- and tissue-based products (HCT/Ps). Section 361 of the Public Health Safety (PHS) Act established and outlined the authority of the FDA to regulate low-risk HCT/Ps in order to prevent the introduction, transmission, and spread of communicable disease. Section 361 provided standards for safety without requiring preclinical development. The FDA established 4 principles to determine the risk of HCT/Ps: the extent of manipulation involved in manufacture, the metabolic activity/autologous nature of the product, whether the product represents a tissue combined with another product, and whether the product is utilized in a fashion homologous with its original function (Figure 1). If a product/therapy meets requirements around all 4 of these principles, then it is deemed a low-risk product and regulated under Section 361 alone. If a product/therapy does not meet requirements around all 4 of these principles, then the FDA regulates the product/therapy under additional codes including Section 351 of the PHS Act. Section 351 outlines a developmental process including preclinical animal trials, phased clinical study, and premarket review by the FDA prior to offering the product/treatment in clinical practice. The developmental process requires investigators and/or industry developers to initiate an Investigational New Drug (IND) program whose end goal is to present data from all developmental study and obtain a Biologic License Application (BLA) approval to market the product.22-23 To establish safety and efficacy, the traditional IND program involves a preclinical animal study, a small pilot human study (Phase I), a small initial randomized controlled trial (Phase II), followed by a large multicenter randomized controlled trial (Phase III) (Figure 2). The FDA has recognized little to no stem cell treatments as products regulated by Section 361 alone. Additionally, the FDA has established precedent regarding allograft stem cells, cells obtained from fat harvest, amniotic/placental products, and cultured cells, suggesting that these products are not low risk and require an IND pathway outlined in Section 351.24-30

 

 

Bone Marrow Aspiration

Surprisingly, the FDA has not moved to regulate the point-of-care use of bone marrow aspirate or platelet-rich plasma and has labeled these as “not HCTPs.” The stem cell concentration of bone marrow aspirate is technique-dependent, declines with age, and has been found to be an important factor for clinical benefit.31 While it is possible to aspirate from multiple sites, posterior iliac crest harvest produces the highest stem cell yield.32-34 Hernigou and colleagues35-36 have outlined safe zones for trocar placement and illustrated that strong aspiration with small-volume syringes, 10-mL syringes, optimizes stem cell harvest. Additionally, studies by Hernigou and colleagues31,37-38 involving tibial nonunion, avascular necrosis of the femur, and augmentation of rotator cuff repair are guideposts to clinicians utilizing bone marrow aspirate.

Amniotic Stem Cell Technologies and Adipose-Derived Stem Cells

While some argue that there is regulatory confusion around amniotic/placental-derived tissues and adipose-derived products, the FDA has clearly established precedent establishing these as products requiring Section 351 development.26-29 Companies are marketing products derived from perinatal byproducts, yet there are multiple FDA letters suggesting that these are not products regulated solely under PHS Act 361 because they do not meet the criteria of homologous use and are not autologous.28-29 Use of these products places risk upon the clinician and the patient. Some argue that adipose-derived stem cell products are 361 products. While the FDA has approved devices for the mechanical processing of lipoaspirate, they have established precedent suggesting that they consider orthopedic applications nonhomologous and any processing that “alters the original relevant characteristics of adipose tissue relating to the tissue’s utility for reconstruction, repair, or replacement” as more than minimal manipulation.26,27 The FDA originally planned an open forum for discussion with clinicians and industry for April 2016. This open forum was delayed due to the volume of interest, and a workshop has been planned for Fall 2016.

Future Regulation of Stem Cell Technologies

While many countries have mirrored the FDA with tight regulatory mechanisms, a few countries have established modern regulatory mechanisms aimed at the promotion of conscientious development, including South Korea, Japan, and England. For example, in 2014 Japan labeled stem cell technologies as “regenerative medicine products,” setting them apart from pharmaceuticals, and implemented a new approval system allowing early observed commercialization with reimbursement after less stringent safety and efficacy milestones.22The observed commercialization lowers time and financial hurdles for development while still requiring the proof of the technology’s worth. Countries that have effected change have positioned themselves to be pioneers in this emerging field.

In March 2016, the Reliable and Effective Growth for Regenerative Health Options that Improve Wellness (REGROW) Act of 2016 (S. 2689 / H.R. 4762) was introduced into the United States Congress. This bipartisan, bicameral legislation was introduced, read twice, and referred to subcommittee. Its goal is to reduce barriers and accelerate development of biologic therapies while keeping the frame work set forth under Sections 351 and 361 of the PHS Act.39 Similar to the pathway in Japan, the REGROW Act would establish a conditional approval pathway that would ensure products are safe and effective while also evolving the regulatory pathway towards progress (Figure 3). Development would still require an IND application after preclinical animal study. However, after safety was established with human Phase I data and preliminary evidence of efficacy with Phase II data, patients could be treated with the investigational therapies and reimbursement collected for a limited period of time (5 years) prior to a large Phase III human clinical trial. Patients treated with the new therapy would be monitored closely. All results would be reported to the FDA in a BLA. This change in legislation would lower but not remove regulatory hurdles necessary for development.

Conclusion

The future of stem cell treatments hinges upon the creation of new favorable regulatory mechanisms that will promote clinical application while ensuring that safety and efficacy milestones are reached. Clinical researchers require freedom to develop these technologies while protecting patients and ensuring the validity of treatments. The coordination of research and regulatory affairs on a global level is necessary focusing on the harmonization of guidelines, regulations, and mechanisms for simultaneous adoption in different countries. The global orthopedic community has made strides regarding the science of stem cell technologies; it is time for us to initiate progressive change regarding regulation so that we can determine what is effective clinically.

References

1.    Caplan AI. Mesenchymal stem cells. J Orthop Res. 1991;9(5):641-650.

2.    Wright DE, Wagers AJ, Gulati AP, Johnson FL, Weissman IL. Physiological migration of hematopoietic stem and progenitor cells. Science. 2001;294(5548):1933-1936.

3.    Caplan AI. Adult mesenchymal stem cells for tissue engineering versus regenerative medicine. J Cell Physiol. 2007;213(2):341-347.

4.    Murphy MB, Moncivais K, Caplan AI. Mesenchymal stem cells: environmentally responsive therapeutics for regenerative medicine. Exp Mol Med. 2013;45:e54.

5.    Ogawa M, LaRue AC, Mehrotra M. Hematopoietic stem cells are pluripotent and not just “hematopoietic.” Blood Cells Mol Dis. 2013;51(1):3-8.

6.    Cesselli D, Beltrami AP, Rigo S, et al. Multipotent progenitor cells are present in human peripheral blood. Circ Res. 2009;104(10):1225-1234.

7.    Massberg S, Schaerli P, Knezevic-Maramica I, et al. Immunosurveillance by hematopoietic progenitor cells trafficking through blood, lymph, and peripheral tissues. Cell. 2007;131(5):994-1008.

8.    Wang Y, Johnsen HE, Mortensen S, et al. Changes in circulating mesenchymal stem cells, stem cell homing factor, and vascular growth factors in patients with acute ST elevation myocardial infarction treated with primary percutaneous coronary intervention. Heart. 2006;92(6):768-774.

9.    Mansilla E, Marín GH, Drago H, et al. Bloodstream cells phenotypically identical to human mesenchymal bone marrow stem cells circulate in large amounts under the influence of acute large skin damage: new evidence for their use in regenerative medicine. Transplant Proc. 2006;38(3):967-969.

10.  Rankin SM. Impact of bone marrow on respiratory disease. Curr Opin Pharmacol. 2008;8(3):236-241.

11.  Rochefort GY, Delorme B, Lopez A, et al. Multipotential mesenchymal stem cells are mobilized into peripheral blood by hypoxia. Stem Cells. 2006;24(10):2202-2208.

12.   Ugarte F, Forsberg EC. Haematopoietic stem cell niches: new insights inspire new questions. EMBO J. 2013;32(19):2535-2547.

13.  Harvanová D, Tóthová T, Sarišský M, Amrichová J, Rosocha J. Isolation and characterization of synovial mesenchymal stem cells. Folia Biol (Praha). 2011;57(3):119-124.

14.  Crisan M, Yap S, Casteilla L, et al. A perivascular origin for mesenchymal stem cells in multiple human organs. Cell Stem Cell. 2008;3(3):301-313.

15.  Frenette PS, Pinho S, Lucas D, Scheiermann C. Mesenchymal stem cell: keystone of the hematopoietic stem cell niche and a stepping-stone for regenerative medicine. Annu Rev Immunol. 2013;31:285-316.

16.  Bonig H, Papayannopoulou T. Hematopoietic stem cell mobilization: updated conceptual renditions. Leukemia. 2013;27(1):24-31.

17.  Ratajczak MZ, Marycz K, Poniewierska-Baran A, Fiedorowicz K, Zbucka-Kretowska M, Moniuszko M. Very small embryonic-like stem cells as a novel developmental concept and the hierarchy of the stem cell compartment. Adv Med Sci. 2014;59(2):273-280.

18.  Smith JN, Calvi LM. Concise review: current concepts in bone marrow microenvironmental regulation of hematopoietic stem and progenitor cells. Stem Cells. 2013;31(6):1044-1050.

19.  Morrison SJ, Scadden DT. The bone marrow niche for haematopoietic stem cells. Nature. 2014;505(7483):327-334.

20.   Vangsness CT Jr, Sternberg H, Harris L. Umbilical cord tissue offers the greatest number of harvestable mesenchymal stem cells for research and clinical application: a literature review of different harvest sites. Arthroscopy. 2015;31(9):1836-1843.

21.  Saw KY, Anz A, Merican S, et al. Articular cartilage regeneration with autologous peripheral blood progenitor cells and hyaluronic acid after arthroscopic subchondral drilling: a report of 5 cases with histology. Arthroscopy. 2011;27(4):493-506.

22.  Board on Health Sciences Policy; Board on Life Sciences; Division on Earth and Life Studies; Institute of Medicine; National Academy of Sciences. Stem Cell Therapies: Opportunities for Ensuring the Quality and Safety of Clinical Offerings: Summary of a Joint Workshop. Washington, DC: National Academies Press (US); 2014.

23.  US Food and Drug Administration. Minimal manipulation of human cells, tissues, and cellular and tissue-based products: draft guidance for industry and food and drug administration staff. http://www.fda.gov/BiologicsBloodVaccines/GuidanceComplianceRegulatoryInformation/Guidances/CellularandGeneTherapy/ucm427692.htm. Updated February 3, 2015. Accessed June 10, 2016.

24.  US Food and Drug Administration. PureGen™ osteoprogenitor cell allograft, parcell laboratories, LLC - untitled letter. http://www.fda.gov/BiologicsBloodVaccines/GuidanceComplianceRegulatoryInformation/ComplianceActivities/Enforcement/UntitledLetters/ucm264011.htm. Published June 23, 2011. Accessed June 10, 2016.

25.  US Food and Drug Administration. Map3 chips allograft-untitled letter. http://www.fda.gov/BiologicsBloodVaccines/GuidanceComplianceRegulatoryInformation/ComplianceActivities/Enforcement/UntitledLetters/ucm418126.htm. Updated December 30, 2014. Accessed June 10, 2016.

26.  US Food and Drug Administration. Irvine stem cell treatment center 12/30/15: warning letter. http://www.fda.gov/ICECI/EnforcementActions/WarningLetters/2015/ucm479837.htm. Published December 30, 2015. Accessed June 10, 2016.

27.  US Food and Drug Administration. IntelliCell Biosciences, Inc. 3/13/12: warning letter. http://www.fda.gov/ICECI/EnforcementActions/WarningLetters/2012/ucm297245.htm. Published March 13, 2012. Accessed June 10, 2016.

28.  US Food and Drug Administration. Osiris Therapeutics, Inc. - untitled letter. http://www.fda.gov/BiologicsBloodVaccines/GuidanceComplianceRegulatoryInformation/ComplianceActivities/Enforcement/UntitledLetters/ucm371540.htm. Updated October 21, 2013. Accessed June 10, 2016.

29.  US Food and Drug Administration. BioD- untitled letter. http://www.fda.gov/downloads/BiologicsBloodVaccines/GuidanceComplianceRegulatoryInformation/ComplianceActivities/Enforcement/UntitledLetters/UCM452862.pdf. Published June 22, 2015. Accessed June 10, 2016.

30.  US Food and Drug Administration. Regenerative Sciences, Inc. http://www.fda.gov/BiologicsBloodVaccines/GuidanceComplianceRegulatoryInformation/ComplianceActivities/Enforcement/UntitledLetters/ucm091991.htm. Published July 25, 2008. Accessed June 10, 2016.

31.  Hernigou P, Poignard A, Beaujean F, Rouard H. Percutaneous autologous bone-marrow grafting for nonunions. Influence of the number and concentration of progenitor cells. J Bone Joint Surg Am. 2005;87(7):1430-1437.

32.  Narbona-Carceles J, Vaquero J, Suárez-Sancho S, Forriol F, Fernández-Santos ME. Bone marrow mesenchymal stem cell aspirates from alternative sources: is the knee as good as the iliac crest? Injury. 2014;45 Suppl 4:S42-S47.

33.  Hyer CF, Berlet GC, Bussewitz BW, Hankins T, Ziegler HL, Philbin TM. Quantitative assessment of the yield of osteoblastic connective tissue progenitors in bone marrow aspirate from the iliac crest, tibia, and calcaneus. J Bone Joint Surg Am. 2013;95(14):1312-1316.

34.  Pierini M, Di Bella C, Dozza B, et al. The posterior iliac crest outperforms the anterior iliac crest when obtaining mesenchymal stem cells from bone marrow. J Bone Joint Surg Am. 2013;95(12):1101-1107.

35.  Hernigou J, Picard L, Alves A, Silvera J, Homma Y, Hernigou P. Understanding bone safety zones during bone marrow aspiration from the iliac crest: the sector rule. Int Orthop. 2014;38(11):2377-2384.

36.  Hernigou P, Homma Y, Flouzat Lachaniette CH, et al. Benefits of small volume and small syringe for bone marrow aspirations of mesenchymal stem cells. Int Orthop. 2013;37(11):2279-2287.

37.  Hernigou P, Flouzat Lachaniette CH, Delambre J, et al. Biologic augmentation of rotator cuff repair with mesenchymal stem cells during arthroscopy improves healing and prevents further tears: a case-controlled study. Int Orthop. 2014;38(9):1811-1818.

38.  Hernigou P, Poignard A, Zilber S, Rouard H. Cell therapy of hip osteonecrosis with autologous bone marrow grafting. Indian J Orthop. 2009;43(1):40-45.

39.  114th Congress (2015-2016). S.2689 - REGROW Act. https://www.congress.gov/bill/114th-congress/senate-bill/2689/text. Accessed June 10, 2016.

Article PDF
Author and Disclosure Information

Adam Anz, MD

Author’s Disclosure Statement: Dr. Anz reports that he is a paid consultant to Arthrex and Smith & Nephew.

Issue
The American Journal of Orthopedics - 45(5)
Publications
Topics
Page Number
274-278, 318
Legacy Keywords
stem cell regulation, stem cell, biologics, review, Anz, sports medicine, arthroscope
Sections
Author and Disclosure Information

Adam Anz, MD

Author’s Disclosure Statement: Dr. Anz reports that he is a paid consultant to Arthrex and Smith & Nephew.

Author and Disclosure Information

Adam Anz, MD

Author’s Disclosure Statement: Dr. Anz reports that he is a paid consultant to Arthrex and Smith & Nephew.

Article PDF
Article PDF

The 2 cardinal properties of stem cells are the ability to self-renew and the ability to differentiate into distinctive end-stage cell types. The work of Caplan1 captured our early attention, with cells cultured from bone marrow differentiating into a number of different cell types of orthopedic interest. Our latest attention has been captured by the additional abilities of these cells to mobilize, monitor, and interact with their surrounding environment.2-4 In response to their environment, stem cells are able to release a broad spectrum of macromolecules with trophic, chemotactic, and immunomodulatory potential, which allows them to participate in injury response, tissue healing, and tissue regeneration.4 These cells are innate to the body’s monitoring, maintenance, repair, and stress response systems.2,4-11 Basic science and animal studies have illustrated the potential of cells with stem potential regardless of their environment/source of harvest.

Where Can We Get Stem Cells?

Cells with stem properties are present in many environmental niches, including the bone marrow, peripheral circulatory system, adipose tissue, synovial tissue, muscle tissue, and tendon tissue.12-15 A number of cell types with stem properties populate the bone marrow niche, including hematopoietic stem/progenitor cells (HSPC), perivascular stromal cells (PSC), endothelial stem cells (ESC), and immature cells with qualities like embryonal stem cells termed very small embryonal-like stem cells (VESL).12,15-19 All of these cells have stem properties and have been shown to differentiate to tissues of orthopedic interest.The interplay, interaction, and potential of these cell types is complex and incompletely understood.12,15-19 When bone marrow is aspirated for culturing purposes, it is unclear which cell line produces the plastic-adherent multipotent cells grown in culture, which are often referred to as mesenchymal stem cells (MSCs). Researches propose that HSPC and/or VESL circulate peripherally in small numbers but leave the bone marrow in certain mobilization instances and are important for the monitoring and maintenance of the majority of tissues in our bodies.5,16 Current clinical utilization of these cell types by the orthopedic community primarily utilizes point-of-care bone marrow aspiration and concentration, while the hematology oncology community mobilizes cells from the bone marrow to the blood stream with pharmaceutical agents and harvests cells via apheresis. Bone marrow aspiration produces variable numbers of stem cells, with studies ranging from 1 stem cell per mL of tissue collected to 300,000 stem cells per mL of tissue collected.20Mobilization and apheresis can produce large volumes of peripheral blood-derived cells with 600,000 HSPC per mL and 2.32 million PSC per mL of tissue collected.21

In adipose tissue, cells adherent to the abluminal side of blood vessels known as pericytes also carry stem qualities. Aspiration and processing of adipose tissue can access these stem cells, producing a product often referred to as stromal vascular fraction (SVF). Processing of lipoaspirate to create stromal vascular fraction requires mechanical or enzymatic processing. This also produces variable numbers of stem cells, with quantitative studies ranging from 5000 to 1.5 million stem cells per mL of tissue collected.20 Similar to adipose-derived stem cells, synovial-derived and muscle-derived stem cells also require mechanical or enzymatic processing. For applications where it is believed that a large number of cells is necessary, investigators often utilize culturing techniques for all sources with the exception of mobilization and apheresis harvest. As clinicians, 3 challenges have proven more important than which cell type to utilize: 1) patient-care logistics regarding collection and application; 2) the undefined dose-response curve regarding stem cell treatments; and 3) evolving government/community regulation.

Regulation of Stem Cell Therapies

The regulation of stem cell technologies is a double-edged sword for development. While loose regulation encourages clinical application and experimentation, patient safety and efficacy concerns are raised, and a technology’s worth is not proven before clinical application. Tight regulation temporarily hampers progress, yet ensures the proof of safety and efficacy prior to widespread implementation. Within the United States, the Food and Drug Administration (FDA) has tightened regulation, established precedent, and intervened in the ability of clinicians to utilize stem cell therapies in humans, through “warning letters,” “untitled letters,” and industry guidance documents.22-30

The FDA categorizes stem cell therapies as human cells, tissues, and cellular- and tissue-based products (HCT/Ps). Section 361 of the Public Health Safety (PHS) Act established and outlined the authority of the FDA to regulate low-risk HCT/Ps in order to prevent the introduction, transmission, and spread of communicable disease. Section 361 provided standards for safety without requiring preclinical development. The FDA established 4 principles to determine the risk of HCT/Ps: the extent of manipulation involved in manufacture, the metabolic activity/autologous nature of the product, whether the product represents a tissue combined with another product, and whether the product is utilized in a fashion homologous with its original function (Figure 1). If a product/therapy meets requirements around all 4 of these principles, then it is deemed a low-risk product and regulated under Section 361 alone. If a product/therapy does not meet requirements around all 4 of these principles, then the FDA regulates the product/therapy under additional codes including Section 351 of the PHS Act. Section 351 outlines a developmental process including preclinical animal trials, phased clinical study, and premarket review by the FDA prior to offering the product/treatment in clinical practice. The developmental process requires investigators and/or industry developers to initiate an Investigational New Drug (IND) program whose end goal is to present data from all developmental study and obtain a Biologic License Application (BLA) approval to market the product.22-23 To establish safety and efficacy, the traditional IND program involves a preclinical animal study, a small pilot human study (Phase I), a small initial randomized controlled trial (Phase II), followed by a large multicenter randomized controlled trial (Phase III) (Figure 2). The FDA has recognized little to no stem cell treatments as products regulated by Section 361 alone. Additionally, the FDA has established precedent regarding allograft stem cells, cells obtained from fat harvest, amniotic/placental products, and cultured cells, suggesting that these products are not low risk and require an IND pathway outlined in Section 351.24-30

 

 

Bone Marrow Aspiration

Surprisingly, the FDA has not moved to regulate the point-of-care use of bone marrow aspirate or platelet-rich plasma and has labeled these as “not HCTPs.” The stem cell concentration of bone marrow aspirate is technique-dependent, declines with age, and has been found to be an important factor for clinical benefit.31 While it is possible to aspirate from multiple sites, posterior iliac crest harvest produces the highest stem cell yield.32-34 Hernigou and colleagues35-36 have outlined safe zones for trocar placement and illustrated that strong aspiration with small-volume syringes, 10-mL syringes, optimizes stem cell harvest. Additionally, studies by Hernigou and colleagues31,37-38 involving tibial nonunion, avascular necrosis of the femur, and augmentation of rotator cuff repair are guideposts to clinicians utilizing bone marrow aspirate.

Amniotic Stem Cell Technologies and Adipose-Derived Stem Cells

While some argue that there is regulatory confusion around amniotic/placental-derived tissues and adipose-derived products, the FDA has clearly established precedent establishing these as products requiring Section 351 development.26-29 Companies are marketing products derived from perinatal byproducts, yet there are multiple FDA letters suggesting that these are not products regulated solely under PHS Act 361 because they do not meet the criteria of homologous use and are not autologous.28-29 Use of these products places risk upon the clinician and the patient. Some argue that adipose-derived stem cell products are 361 products. While the FDA has approved devices for the mechanical processing of lipoaspirate, they have established precedent suggesting that they consider orthopedic applications nonhomologous and any processing that “alters the original relevant characteristics of adipose tissue relating to the tissue’s utility for reconstruction, repair, or replacement” as more than minimal manipulation.26,27 The FDA originally planned an open forum for discussion with clinicians and industry for April 2016. This open forum was delayed due to the volume of interest, and a workshop has been planned for Fall 2016.

Future Regulation of Stem Cell Technologies

While many countries have mirrored the FDA with tight regulatory mechanisms, a few countries have established modern regulatory mechanisms aimed at the promotion of conscientious development, including South Korea, Japan, and England. For example, in 2014 Japan labeled stem cell technologies as “regenerative medicine products,” setting them apart from pharmaceuticals, and implemented a new approval system allowing early observed commercialization with reimbursement after less stringent safety and efficacy milestones.22The observed commercialization lowers time and financial hurdles for development while still requiring the proof of the technology’s worth. Countries that have effected change have positioned themselves to be pioneers in this emerging field.

In March 2016, the Reliable and Effective Growth for Regenerative Health Options that Improve Wellness (REGROW) Act of 2016 (S. 2689 / H.R. 4762) was introduced into the United States Congress. This bipartisan, bicameral legislation was introduced, read twice, and referred to subcommittee. Its goal is to reduce barriers and accelerate development of biologic therapies while keeping the frame work set forth under Sections 351 and 361 of the PHS Act.39 Similar to the pathway in Japan, the REGROW Act would establish a conditional approval pathway that would ensure products are safe and effective while also evolving the regulatory pathway towards progress (Figure 3). Development would still require an IND application after preclinical animal study. However, after safety was established with human Phase I data and preliminary evidence of efficacy with Phase II data, patients could be treated with the investigational therapies and reimbursement collected for a limited period of time (5 years) prior to a large Phase III human clinical trial. Patients treated with the new therapy would be monitored closely. All results would be reported to the FDA in a BLA. This change in legislation would lower but not remove regulatory hurdles necessary for development.

Conclusion

The future of stem cell treatments hinges upon the creation of new favorable regulatory mechanisms that will promote clinical application while ensuring that safety and efficacy milestones are reached. Clinical researchers require freedom to develop these technologies while protecting patients and ensuring the validity of treatments. The coordination of research and regulatory affairs on a global level is necessary focusing on the harmonization of guidelines, regulations, and mechanisms for simultaneous adoption in different countries. The global orthopedic community has made strides regarding the science of stem cell technologies; it is time for us to initiate progressive change regarding regulation so that we can determine what is effective clinically.

The 2 cardinal properties of stem cells are the ability to self-renew and the ability to differentiate into distinctive end-stage cell types. The work of Caplan1 captured our early attention, with cells cultured from bone marrow differentiating into a number of different cell types of orthopedic interest. Our latest attention has been captured by the additional abilities of these cells to mobilize, monitor, and interact with their surrounding environment.2-4 In response to their environment, stem cells are able to release a broad spectrum of macromolecules with trophic, chemotactic, and immunomodulatory potential, which allows them to participate in injury response, tissue healing, and tissue regeneration.4 These cells are innate to the body’s monitoring, maintenance, repair, and stress response systems.2,4-11 Basic science and animal studies have illustrated the potential of cells with stem potential regardless of their environment/source of harvest.

Where Can We Get Stem Cells?

Cells with stem properties are present in many environmental niches, including the bone marrow, peripheral circulatory system, adipose tissue, synovial tissue, muscle tissue, and tendon tissue.12-15 A number of cell types with stem properties populate the bone marrow niche, including hematopoietic stem/progenitor cells (HSPC), perivascular stromal cells (PSC), endothelial stem cells (ESC), and immature cells with qualities like embryonal stem cells termed very small embryonal-like stem cells (VESL).12,15-19 All of these cells have stem properties and have been shown to differentiate to tissues of orthopedic interest.The interplay, interaction, and potential of these cell types is complex and incompletely understood.12,15-19 When bone marrow is aspirated for culturing purposes, it is unclear which cell line produces the plastic-adherent multipotent cells grown in culture, which are often referred to as mesenchymal stem cells (MSCs). Researches propose that HSPC and/or VESL circulate peripherally in small numbers but leave the bone marrow in certain mobilization instances and are important for the monitoring and maintenance of the majority of tissues in our bodies.5,16 Current clinical utilization of these cell types by the orthopedic community primarily utilizes point-of-care bone marrow aspiration and concentration, while the hematology oncology community mobilizes cells from the bone marrow to the blood stream with pharmaceutical agents and harvests cells via apheresis. Bone marrow aspiration produces variable numbers of stem cells, with studies ranging from 1 stem cell per mL of tissue collected to 300,000 stem cells per mL of tissue collected.20Mobilization and apheresis can produce large volumes of peripheral blood-derived cells with 600,000 HSPC per mL and 2.32 million PSC per mL of tissue collected.21

In adipose tissue, cells adherent to the abluminal side of blood vessels known as pericytes also carry stem qualities. Aspiration and processing of adipose tissue can access these stem cells, producing a product often referred to as stromal vascular fraction (SVF). Processing of lipoaspirate to create stromal vascular fraction requires mechanical or enzymatic processing. This also produces variable numbers of stem cells, with quantitative studies ranging from 5000 to 1.5 million stem cells per mL of tissue collected.20 Similar to adipose-derived stem cells, synovial-derived and muscle-derived stem cells also require mechanical or enzymatic processing. For applications where it is believed that a large number of cells is necessary, investigators often utilize culturing techniques for all sources with the exception of mobilization and apheresis harvest. As clinicians, 3 challenges have proven more important than which cell type to utilize: 1) patient-care logistics regarding collection and application; 2) the undefined dose-response curve regarding stem cell treatments; and 3) evolving government/community regulation.

Regulation of Stem Cell Therapies

The regulation of stem cell technologies is a double-edged sword for development. While loose regulation encourages clinical application and experimentation, patient safety and efficacy concerns are raised, and a technology’s worth is not proven before clinical application. Tight regulation temporarily hampers progress, yet ensures the proof of safety and efficacy prior to widespread implementation. Within the United States, the Food and Drug Administration (FDA) has tightened regulation, established precedent, and intervened in the ability of clinicians to utilize stem cell therapies in humans, through “warning letters,” “untitled letters,” and industry guidance documents.22-30

The FDA categorizes stem cell therapies as human cells, tissues, and cellular- and tissue-based products (HCT/Ps). Section 361 of the Public Health Safety (PHS) Act established and outlined the authority of the FDA to regulate low-risk HCT/Ps in order to prevent the introduction, transmission, and spread of communicable disease. Section 361 provided standards for safety without requiring preclinical development. The FDA established 4 principles to determine the risk of HCT/Ps: the extent of manipulation involved in manufacture, the metabolic activity/autologous nature of the product, whether the product represents a tissue combined with another product, and whether the product is utilized in a fashion homologous with its original function (Figure 1). If a product/therapy meets requirements around all 4 of these principles, then it is deemed a low-risk product and regulated under Section 361 alone. If a product/therapy does not meet requirements around all 4 of these principles, then the FDA regulates the product/therapy under additional codes including Section 351 of the PHS Act. Section 351 outlines a developmental process including preclinical animal trials, phased clinical study, and premarket review by the FDA prior to offering the product/treatment in clinical practice. The developmental process requires investigators and/or industry developers to initiate an Investigational New Drug (IND) program whose end goal is to present data from all developmental study and obtain a Biologic License Application (BLA) approval to market the product.22-23 To establish safety and efficacy, the traditional IND program involves a preclinical animal study, a small pilot human study (Phase I), a small initial randomized controlled trial (Phase II), followed by a large multicenter randomized controlled trial (Phase III) (Figure 2). The FDA has recognized little to no stem cell treatments as products regulated by Section 361 alone. Additionally, the FDA has established precedent regarding allograft stem cells, cells obtained from fat harvest, amniotic/placental products, and cultured cells, suggesting that these products are not low risk and require an IND pathway outlined in Section 351.24-30

 

 

Bone Marrow Aspiration

Surprisingly, the FDA has not moved to regulate the point-of-care use of bone marrow aspirate or platelet-rich plasma and has labeled these as “not HCTPs.” The stem cell concentration of bone marrow aspirate is technique-dependent, declines with age, and has been found to be an important factor for clinical benefit.31 While it is possible to aspirate from multiple sites, posterior iliac crest harvest produces the highest stem cell yield.32-34 Hernigou and colleagues35-36 have outlined safe zones for trocar placement and illustrated that strong aspiration with small-volume syringes, 10-mL syringes, optimizes stem cell harvest. Additionally, studies by Hernigou and colleagues31,37-38 involving tibial nonunion, avascular necrosis of the femur, and augmentation of rotator cuff repair are guideposts to clinicians utilizing bone marrow aspirate.

Amniotic Stem Cell Technologies and Adipose-Derived Stem Cells

While some argue that there is regulatory confusion around amniotic/placental-derived tissues and adipose-derived products, the FDA has clearly established precedent establishing these as products requiring Section 351 development.26-29 Companies are marketing products derived from perinatal byproducts, yet there are multiple FDA letters suggesting that these are not products regulated solely under PHS Act 361 because they do not meet the criteria of homologous use and are not autologous.28-29 Use of these products places risk upon the clinician and the patient. Some argue that adipose-derived stem cell products are 361 products. While the FDA has approved devices for the mechanical processing of lipoaspirate, they have established precedent suggesting that they consider orthopedic applications nonhomologous and any processing that “alters the original relevant characteristics of adipose tissue relating to the tissue’s utility for reconstruction, repair, or replacement” as more than minimal manipulation.26,27 The FDA originally planned an open forum for discussion with clinicians and industry for April 2016. This open forum was delayed due to the volume of interest, and a workshop has been planned for Fall 2016.

Future Regulation of Stem Cell Technologies

While many countries have mirrored the FDA with tight regulatory mechanisms, a few countries have established modern regulatory mechanisms aimed at the promotion of conscientious development, including South Korea, Japan, and England. For example, in 2014 Japan labeled stem cell technologies as “regenerative medicine products,” setting them apart from pharmaceuticals, and implemented a new approval system allowing early observed commercialization with reimbursement after less stringent safety and efficacy milestones.22The observed commercialization lowers time and financial hurdles for development while still requiring the proof of the technology’s worth. Countries that have effected change have positioned themselves to be pioneers in this emerging field.

In March 2016, the Reliable and Effective Growth for Regenerative Health Options that Improve Wellness (REGROW) Act of 2016 (S. 2689 / H.R. 4762) was introduced into the United States Congress. This bipartisan, bicameral legislation was introduced, read twice, and referred to subcommittee. Its goal is to reduce barriers and accelerate development of biologic therapies while keeping the frame work set forth under Sections 351 and 361 of the PHS Act.39 Similar to the pathway in Japan, the REGROW Act would establish a conditional approval pathway that would ensure products are safe and effective while also evolving the regulatory pathway towards progress (Figure 3). Development would still require an IND application after preclinical animal study. However, after safety was established with human Phase I data and preliminary evidence of efficacy with Phase II data, patients could be treated with the investigational therapies and reimbursement collected for a limited period of time (5 years) prior to a large Phase III human clinical trial. Patients treated with the new therapy would be monitored closely. All results would be reported to the FDA in a BLA. This change in legislation would lower but not remove regulatory hurdles necessary for development.

Conclusion

The future of stem cell treatments hinges upon the creation of new favorable regulatory mechanisms that will promote clinical application while ensuring that safety and efficacy milestones are reached. Clinical researchers require freedom to develop these technologies while protecting patients and ensuring the validity of treatments. The coordination of research and regulatory affairs on a global level is necessary focusing on the harmonization of guidelines, regulations, and mechanisms for simultaneous adoption in different countries. The global orthopedic community has made strides regarding the science of stem cell technologies; it is time for us to initiate progressive change regarding regulation so that we can determine what is effective clinically.

References

1.    Caplan AI. Mesenchymal stem cells. J Orthop Res. 1991;9(5):641-650.

2.    Wright DE, Wagers AJ, Gulati AP, Johnson FL, Weissman IL. Physiological migration of hematopoietic stem and progenitor cells. Science. 2001;294(5548):1933-1936.

3.    Caplan AI. Adult mesenchymal stem cells for tissue engineering versus regenerative medicine. J Cell Physiol. 2007;213(2):341-347.

4.    Murphy MB, Moncivais K, Caplan AI. Mesenchymal stem cells: environmentally responsive therapeutics for regenerative medicine. Exp Mol Med. 2013;45:e54.

5.    Ogawa M, LaRue AC, Mehrotra M. Hematopoietic stem cells are pluripotent and not just “hematopoietic.” Blood Cells Mol Dis. 2013;51(1):3-8.

6.    Cesselli D, Beltrami AP, Rigo S, et al. Multipotent progenitor cells are present in human peripheral blood. Circ Res. 2009;104(10):1225-1234.

7.    Massberg S, Schaerli P, Knezevic-Maramica I, et al. Immunosurveillance by hematopoietic progenitor cells trafficking through blood, lymph, and peripheral tissues. Cell. 2007;131(5):994-1008.

8.    Wang Y, Johnsen HE, Mortensen S, et al. Changes in circulating mesenchymal stem cells, stem cell homing factor, and vascular growth factors in patients with acute ST elevation myocardial infarction treated with primary percutaneous coronary intervention. Heart. 2006;92(6):768-774.

9.    Mansilla E, Marín GH, Drago H, et al. Bloodstream cells phenotypically identical to human mesenchymal bone marrow stem cells circulate in large amounts under the influence of acute large skin damage: new evidence for their use in regenerative medicine. Transplant Proc. 2006;38(3):967-969.

10.  Rankin SM. Impact of bone marrow on respiratory disease. Curr Opin Pharmacol. 2008;8(3):236-241.

11.  Rochefort GY, Delorme B, Lopez A, et al. Multipotential mesenchymal stem cells are mobilized into peripheral blood by hypoxia. Stem Cells. 2006;24(10):2202-2208.

12.   Ugarte F, Forsberg EC. Haematopoietic stem cell niches: new insights inspire new questions. EMBO J. 2013;32(19):2535-2547.

13.  Harvanová D, Tóthová T, Sarišský M, Amrichová J, Rosocha J. Isolation and characterization of synovial mesenchymal stem cells. Folia Biol (Praha). 2011;57(3):119-124.

14.  Crisan M, Yap S, Casteilla L, et al. A perivascular origin for mesenchymal stem cells in multiple human organs. Cell Stem Cell. 2008;3(3):301-313.

15.  Frenette PS, Pinho S, Lucas D, Scheiermann C. Mesenchymal stem cell: keystone of the hematopoietic stem cell niche and a stepping-stone for regenerative medicine. Annu Rev Immunol. 2013;31:285-316.

16.  Bonig H, Papayannopoulou T. Hematopoietic stem cell mobilization: updated conceptual renditions. Leukemia. 2013;27(1):24-31.

17.  Ratajczak MZ, Marycz K, Poniewierska-Baran A, Fiedorowicz K, Zbucka-Kretowska M, Moniuszko M. Very small embryonic-like stem cells as a novel developmental concept and the hierarchy of the stem cell compartment. Adv Med Sci. 2014;59(2):273-280.

18.  Smith JN, Calvi LM. Concise review: current concepts in bone marrow microenvironmental regulation of hematopoietic stem and progenitor cells. Stem Cells. 2013;31(6):1044-1050.

19.  Morrison SJ, Scadden DT. The bone marrow niche for haematopoietic stem cells. Nature. 2014;505(7483):327-334.

20.   Vangsness CT Jr, Sternberg H, Harris L. Umbilical cord tissue offers the greatest number of harvestable mesenchymal stem cells for research and clinical application: a literature review of different harvest sites. Arthroscopy. 2015;31(9):1836-1843.

21.  Saw KY, Anz A, Merican S, et al. Articular cartilage regeneration with autologous peripheral blood progenitor cells and hyaluronic acid after arthroscopic subchondral drilling: a report of 5 cases with histology. Arthroscopy. 2011;27(4):493-506.

22.  Board on Health Sciences Policy; Board on Life Sciences; Division on Earth and Life Studies; Institute of Medicine; National Academy of Sciences. Stem Cell Therapies: Opportunities for Ensuring the Quality and Safety of Clinical Offerings: Summary of a Joint Workshop. Washington, DC: National Academies Press (US); 2014.

23.  US Food and Drug Administration. Minimal manipulation of human cells, tissues, and cellular and tissue-based products: draft guidance for industry and food and drug administration staff. http://www.fda.gov/BiologicsBloodVaccines/GuidanceComplianceRegulatoryInformation/Guidances/CellularandGeneTherapy/ucm427692.htm. Updated February 3, 2015. Accessed June 10, 2016.

24.  US Food and Drug Administration. PureGen™ osteoprogenitor cell allograft, parcell laboratories, LLC - untitled letter. http://www.fda.gov/BiologicsBloodVaccines/GuidanceComplianceRegulatoryInformation/ComplianceActivities/Enforcement/UntitledLetters/ucm264011.htm. Published June 23, 2011. Accessed June 10, 2016.

25.  US Food and Drug Administration. Map3 chips allograft-untitled letter. http://www.fda.gov/BiologicsBloodVaccines/GuidanceComplianceRegulatoryInformation/ComplianceActivities/Enforcement/UntitledLetters/ucm418126.htm. Updated December 30, 2014. Accessed June 10, 2016.

26.  US Food and Drug Administration. Irvine stem cell treatment center 12/30/15: warning letter. http://www.fda.gov/ICECI/EnforcementActions/WarningLetters/2015/ucm479837.htm. Published December 30, 2015. Accessed June 10, 2016.

27.  US Food and Drug Administration. IntelliCell Biosciences, Inc. 3/13/12: warning letter. http://www.fda.gov/ICECI/EnforcementActions/WarningLetters/2012/ucm297245.htm. Published March 13, 2012. Accessed June 10, 2016.

28.  US Food and Drug Administration. Osiris Therapeutics, Inc. - untitled letter. http://www.fda.gov/BiologicsBloodVaccines/GuidanceComplianceRegulatoryInformation/ComplianceActivities/Enforcement/UntitledLetters/ucm371540.htm. Updated October 21, 2013. Accessed June 10, 2016.

29.  US Food and Drug Administration. BioD- untitled letter. http://www.fda.gov/downloads/BiologicsBloodVaccines/GuidanceComplianceRegulatoryInformation/ComplianceActivities/Enforcement/UntitledLetters/UCM452862.pdf. Published June 22, 2015. Accessed June 10, 2016.

30.  US Food and Drug Administration. Regenerative Sciences, Inc. http://www.fda.gov/BiologicsBloodVaccines/GuidanceComplianceRegulatoryInformation/ComplianceActivities/Enforcement/UntitledLetters/ucm091991.htm. Published July 25, 2008. Accessed June 10, 2016.

31.  Hernigou P, Poignard A, Beaujean F, Rouard H. Percutaneous autologous bone-marrow grafting for nonunions. Influence of the number and concentration of progenitor cells. J Bone Joint Surg Am. 2005;87(7):1430-1437.

32.  Narbona-Carceles J, Vaquero J, Suárez-Sancho S, Forriol F, Fernández-Santos ME. Bone marrow mesenchymal stem cell aspirates from alternative sources: is the knee as good as the iliac crest? Injury. 2014;45 Suppl 4:S42-S47.

33.  Hyer CF, Berlet GC, Bussewitz BW, Hankins T, Ziegler HL, Philbin TM. Quantitative assessment of the yield of osteoblastic connective tissue progenitors in bone marrow aspirate from the iliac crest, tibia, and calcaneus. J Bone Joint Surg Am. 2013;95(14):1312-1316.

34.  Pierini M, Di Bella C, Dozza B, et al. The posterior iliac crest outperforms the anterior iliac crest when obtaining mesenchymal stem cells from bone marrow. J Bone Joint Surg Am. 2013;95(12):1101-1107.

35.  Hernigou J, Picard L, Alves A, Silvera J, Homma Y, Hernigou P. Understanding bone safety zones during bone marrow aspiration from the iliac crest: the sector rule. Int Orthop. 2014;38(11):2377-2384.

36.  Hernigou P, Homma Y, Flouzat Lachaniette CH, et al. Benefits of small volume and small syringe for bone marrow aspirations of mesenchymal stem cells. Int Orthop. 2013;37(11):2279-2287.

37.  Hernigou P, Flouzat Lachaniette CH, Delambre J, et al. Biologic augmentation of rotator cuff repair with mesenchymal stem cells during arthroscopy improves healing and prevents further tears: a case-controlled study. Int Orthop. 2014;38(9):1811-1818.

38.  Hernigou P, Poignard A, Zilber S, Rouard H. Cell therapy of hip osteonecrosis with autologous bone marrow grafting. Indian J Orthop. 2009;43(1):40-45.

39.  114th Congress (2015-2016). S.2689 - REGROW Act. https://www.congress.gov/bill/114th-congress/senate-bill/2689/text. Accessed June 10, 2016.

References

1.    Caplan AI. Mesenchymal stem cells. J Orthop Res. 1991;9(5):641-650.

2.    Wright DE, Wagers AJ, Gulati AP, Johnson FL, Weissman IL. Physiological migration of hematopoietic stem and progenitor cells. Science. 2001;294(5548):1933-1936.

3.    Caplan AI. Adult mesenchymal stem cells for tissue engineering versus regenerative medicine. J Cell Physiol. 2007;213(2):341-347.

4.    Murphy MB, Moncivais K, Caplan AI. Mesenchymal stem cells: environmentally responsive therapeutics for regenerative medicine. Exp Mol Med. 2013;45:e54.

5.    Ogawa M, LaRue AC, Mehrotra M. Hematopoietic stem cells are pluripotent and not just “hematopoietic.” Blood Cells Mol Dis. 2013;51(1):3-8.

6.    Cesselli D, Beltrami AP, Rigo S, et al. Multipotent progenitor cells are present in human peripheral blood. Circ Res. 2009;104(10):1225-1234.

7.    Massberg S, Schaerli P, Knezevic-Maramica I, et al. Immunosurveillance by hematopoietic progenitor cells trafficking through blood, lymph, and peripheral tissues. Cell. 2007;131(5):994-1008.

8.    Wang Y, Johnsen HE, Mortensen S, et al. Changes in circulating mesenchymal stem cells, stem cell homing factor, and vascular growth factors in patients with acute ST elevation myocardial infarction treated with primary percutaneous coronary intervention. Heart. 2006;92(6):768-774.

9.    Mansilla E, Marín GH, Drago H, et al. Bloodstream cells phenotypically identical to human mesenchymal bone marrow stem cells circulate in large amounts under the influence of acute large skin damage: new evidence for their use in regenerative medicine. Transplant Proc. 2006;38(3):967-969.

10.  Rankin SM. Impact of bone marrow on respiratory disease. Curr Opin Pharmacol. 2008;8(3):236-241.

11.  Rochefort GY, Delorme B, Lopez A, et al. Multipotential mesenchymal stem cells are mobilized into peripheral blood by hypoxia. Stem Cells. 2006;24(10):2202-2208.

12.   Ugarte F, Forsberg EC. Haematopoietic stem cell niches: new insights inspire new questions. EMBO J. 2013;32(19):2535-2547.

13.  Harvanová D, Tóthová T, Sarišský M, Amrichová J, Rosocha J. Isolation and characterization of synovial mesenchymal stem cells. Folia Biol (Praha). 2011;57(3):119-124.

14.  Crisan M, Yap S, Casteilla L, et al. A perivascular origin for mesenchymal stem cells in multiple human organs. Cell Stem Cell. 2008;3(3):301-313.

15.  Frenette PS, Pinho S, Lucas D, Scheiermann C. Mesenchymal stem cell: keystone of the hematopoietic stem cell niche and a stepping-stone for regenerative medicine. Annu Rev Immunol. 2013;31:285-316.

16.  Bonig H, Papayannopoulou T. Hematopoietic stem cell mobilization: updated conceptual renditions. Leukemia. 2013;27(1):24-31.

17.  Ratajczak MZ, Marycz K, Poniewierska-Baran A, Fiedorowicz K, Zbucka-Kretowska M, Moniuszko M. Very small embryonic-like stem cells as a novel developmental concept and the hierarchy of the stem cell compartment. Adv Med Sci. 2014;59(2):273-280.

18.  Smith JN, Calvi LM. Concise review: current concepts in bone marrow microenvironmental regulation of hematopoietic stem and progenitor cells. Stem Cells. 2013;31(6):1044-1050.

19.  Morrison SJ, Scadden DT. The bone marrow niche for haematopoietic stem cells. Nature. 2014;505(7483):327-334.

20.   Vangsness CT Jr, Sternberg H, Harris L. Umbilical cord tissue offers the greatest number of harvestable mesenchymal stem cells for research and clinical application: a literature review of different harvest sites. Arthroscopy. 2015;31(9):1836-1843.

21.  Saw KY, Anz A, Merican S, et al. Articular cartilage regeneration with autologous peripheral blood progenitor cells and hyaluronic acid after arthroscopic subchondral drilling: a report of 5 cases with histology. Arthroscopy. 2011;27(4):493-506.

22.  Board on Health Sciences Policy; Board on Life Sciences; Division on Earth and Life Studies; Institute of Medicine; National Academy of Sciences. Stem Cell Therapies: Opportunities for Ensuring the Quality and Safety of Clinical Offerings: Summary of a Joint Workshop. Washington, DC: National Academies Press (US); 2014.

23.  US Food and Drug Administration. Minimal manipulation of human cells, tissues, and cellular and tissue-based products: draft guidance for industry and food and drug administration staff. http://www.fda.gov/BiologicsBloodVaccines/GuidanceComplianceRegulatoryInformation/Guidances/CellularandGeneTherapy/ucm427692.htm. Updated February 3, 2015. Accessed June 10, 2016.

24.  US Food and Drug Administration. PureGen™ osteoprogenitor cell allograft, parcell laboratories, LLC - untitled letter. http://www.fda.gov/BiologicsBloodVaccines/GuidanceComplianceRegulatoryInformation/ComplianceActivities/Enforcement/UntitledLetters/ucm264011.htm. Published June 23, 2011. Accessed June 10, 2016.

25.  US Food and Drug Administration. Map3 chips allograft-untitled letter. http://www.fda.gov/BiologicsBloodVaccines/GuidanceComplianceRegulatoryInformation/ComplianceActivities/Enforcement/UntitledLetters/ucm418126.htm. Updated December 30, 2014. Accessed June 10, 2016.

26.  US Food and Drug Administration. Irvine stem cell treatment center 12/30/15: warning letter. http://www.fda.gov/ICECI/EnforcementActions/WarningLetters/2015/ucm479837.htm. Published December 30, 2015. Accessed June 10, 2016.

27.  US Food and Drug Administration. IntelliCell Biosciences, Inc. 3/13/12: warning letter. http://www.fda.gov/ICECI/EnforcementActions/WarningLetters/2012/ucm297245.htm. Published March 13, 2012. Accessed June 10, 2016.

28.  US Food and Drug Administration. Osiris Therapeutics, Inc. - untitled letter. http://www.fda.gov/BiologicsBloodVaccines/GuidanceComplianceRegulatoryInformation/ComplianceActivities/Enforcement/UntitledLetters/ucm371540.htm. Updated October 21, 2013. Accessed June 10, 2016.

29.  US Food and Drug Administration. BioD- untitled letter. http://www.fda.gov/downloads/BiologicsBloodVaccines/GuidanceComplianceRegulatoryInformation/ComplianceActivities/Enforcement/UntitledLetters/UCM452862.pdf. Published June 22, 2015. Accessed June 10, 2016.

30.  US Food and Drug Administration. Regenerative Sciences, Inc. http://www.fda.gov/BiologicsBloodVaccines/GuidanceComplianceRegulatoryInformation/ComplianceActivities/Enforcement/UntitledLetters/ucm091991.htm. Published July 25, 2008. Accessed June 10, 2016.

31.  Hernigou P, Poignard A, Beaujean F, Rouard H. Percutaneous autologous bone-marrow grafting for nonunions. Influence of the number and concentration of progenitor cells. J Bone Joint Surg Am. 2005;87(7):1430-1437.

32.  Narbona-Carceles J, Vaquero J, Suárez-Sancho S, Forriol F, Fernández-Santos ME. Bone marrow mesenchymal stem cell aspirates from alternative sources: is the knee as good as the iliac crest? Injury. 2014;45 Suppl 4:S42-S47.

33.  Hyer CF, Berlet GC, Bussewitz BW, Hankins T, Ziegler HL, Philbin TM. Quantitative assessment of the yield of osteoblastic connective tissue progenitors in bone marrow aspirate from the iliac crest, tibia, and calcaneus. J Bone Joint Surg Am. 2013;95(14):1312-1316.

34.  Pierini M, Di Bella C, Dozza B, et al. The posterior iliac crest outperforms the anterior iliac crest when obtaining mesenchymal stem cells from bone marrow. J Bone Joint Surg Am. 2013;95(12):1101-1107.

35.  Hernigou J, Picard L, Alves A, Silvera J, Homma Y, Hernigou P. Understanding bone safety zones during bone marrow aspiration from the iliac crest: the sector rule. Int Orthop. 2014;38(11):2377-2384.

36.  Hernigou P, Homma Y, Flouzat Lachaniette CH, et al. Benefits of small volume and small syringe for bone marrow aspirations of mesenchymal stem cells. Int Orthop. 2013;37(11):2279-2287.

37.  Hernigou P, Flouzat Lachaniette CH, Delambre J, et al. Biologic augmentation of rotator cuff repair with mesenchymal stem cells during arthroscopy improves healing and prevents further tears: a case-controlled study. Int Orthop. 2014;38(9):1811-1818.

38.  Hernigou P, Poignard A, Zilber S, Rouard H. Cell therapy of hip osteonecrosis with autologous bone marrow grafting. Indian J Orthop. 2009;43(1):40-45.

39.  114th Congress (2015-2016). S.2689 - REGROW Act. https://www.congress.gov/bill/114th-congress/senate-bill/2689/text. Accessed June 10, 2016.

Issue
The American Journal of Orthopedics - 45(5)
Issue
The American Journal of Orthopedics - 45(5)
Page Number
274-278, 318
Page Number
274-278, 318
Publications
Publications
Topics
Article Type
Display Headline
Current and Future Stem Cell Regulation: A Call to Action
Display Headline
Current and Future Stem Cell Regulation: A Call to Action
Legacy Keywords
stem cell regulation, stem cell, biologics, review, Anz, sports medicine, arthroscope
Legacy Keywords
stem cell regulation, stem cell, biologics, review, Anz, sports medicine, arthroscope
Sections
Article Source

PURLs Copyright

Inside the Article

Article PDF Media

Febrile, Immunocompromised Man With Rash

Article Type
Changed
Mon, 07/09/2018 - 10:47
Display Headline
Febrile, Immunocompromised Man With Rash

IN THIS ARTICLE

 

  • Conditions associated with increased risk for case disease
  • Outcome for the case patient
  • Differential diagnosis

A 78-year-old white man with chronic lymphocytic leukemia is admitted to the hospital with worsening cough, shortness of breath, and fever. His medical history is significant for pneumonia caused by Pneumocystis jirovecii in the past year. In the weeks preceding hospital admission, the patient developed an erythematous rash over his trunk (see photographs).

During the man’s hospital stay, this eruption becomes increasingly pruritic and spreads to his proximal extremities. His pulmonary symptoms improve slightly following the initiation of broad-spectrum antibiotic therapy (piperacillin/tazobactam and vancomycin), but CT performed one week after admission reveals worsening pulmonary disease (see image). The radiologist’s differential diagnosis includes neoplasm, fungal infection, Kaposi sarcoma, and autoimmune disease.

 

A. The patient's back shows a distribution of
lesions, with areas of excoriation caused by
scratching.
B. A close-up reveals erythematous
papules and keratotic papules.

Suspecting that the progressive rash is related to the systemic process, the provider orders a punch biopsy in an effort to reach a diagnosis with minimally invasive studies. When the patient’s clinical status further declines, he undergoes video-assisted thoracoscopic surgery to obtain an excisional biopsy of one of the pulmonary nodules. Subsequent analysis reveals fungal organisms consistent with histoplasmosis. Interestingly, in the histologic review of the skin biopsy, focal acantholytic dyskeratosis—suggestive of Grover disease—is identified.

 

CT of the chest demonstrates numerous pulmonarynodules, air bronchograms, and reticulonodularinfiltrates.

Continue for discussion >>

 

 

DISCUSSION
Grover disease (GD), also known as transient acantholytic dermatosis, is a skin condition of uncertain pathophysiology. Its clinical presentation can be difficult to distinguish from other dermopathies.1,2

Incidence
GD most commonly appears in fair-skinned persons of late middle age, with men affected at two to three times the rate seen in women.1,2 Although GD has been documented in patients ranging in age from 4 to 100, this dermopathy is rare in younger patients.1-3 Persons with a prior history of atopic dermatitis, contact dermatitis, or xerosis cutis are at increased risk for GD—likely due to an increased dermatologic sensitivity to irritants resulting from the aforementioned disorders.1,4 Risk for GD is also elevated in patients with chronic medical conditions, immunodeficiency, febrile illnesses, or malignancies (see Table 1).2-5

The true incidence of GD is not known; biopsy-proven GD is uncommon, and specific data on the incidence and prevalence of the condition are lacking. Swiss researchers who reviewed more than 30,000 skin biopsies in the late 1990s noted only 24 diagnosed cases of GD, and similar findings have been reported in the United States.1,6 However, the variable presentation and often mild nature of GD may result in cases of misdiagnosis, lack of diagnosis, or empiric treatment in the absence of a formal diagnosis.7

Causative factors
Although the pathophysiology of GD is uncertain, the most likely cause is an occlusion of the eccrine glands.3 This is followed by acantholysis, or separation of keratinocytes within the epidermis, which in turn leads to the development of vesicular lesions.

Though diagnosed most often in the winter, GD has also been associated with exposure to sunlight, heat, xerosis, and diaphoresis.1,3 Hospitalized or bedridden patients are at risk for occlusion of the eccrine glands and thus for GD. Use of certain therapies, including sulfadoxine/pyrimethamine (an antimalarial treatment), ionizing radiation, and interleukin-4, may also be precursors for the condition.2

Other exacerbating factors have been suggested, but reports are largely limited to case studies and other anecdotal publications.2 Concrete data regarding the etiology and pathophysiology of GD are still relatively scarce.

Clinical presentation
Patients with GD present with pruritic dermatitis on the trunk and proximal extremities, most classically on the anterior chest and mid back.2,3 The severity of the rash does not necessarily correlate to the degree of pruritus. Some patients report only mild pruritus, while others experience debilitating discomfort and pain. In most cases, erythematous and violaceous papules and vesicles appear first, followed by keratotic erosions.3

GD is a self-limited disorder that often resolves within a few weeks, although some cases will persist for several months.3,5 Severity and duration of symptoms appear to be correlated with increasing age; elderly patients experience worse pruritus for longer periods than do younger patients.2

Although the condition is sometimes referred to as transient acantholytic dermatosis, there are three typical presentations of GD: transient eruptive, persistent pruritic, and chronic asymptomatic.4 Transient eruptive GD presents suddenly, with intense pruritus, and tends to subside over several weeks. Persistent pruritic disease generally causes a milder pruritus, with symptoms that last for several months and are not well controlled by medication. Chronic asymptomatic GD can be difficult to treat medically, yet this form of the disease typically causes little to no irritation and requires minimal therapeutic intervention.4

Systemic symptoms of GD have not been observed. Pruritus and rash are the main features in most affected patients. However, pruritic papulovesicular eruptions are commonly seen in other conditions with similar characteristics (see Table 2,3,4), and GD is comparatively rare. While clinical appearance alone may suggest a diagnosis of GD, further testing may be needed to eliminate other conditions from the differential.

Treatment and prognosis
In the absence of randomized therapeutic trials for GD, there are no strict guidelines for treatment. When irritation, inflammation, and pruritus become bothersome, several interventions may be considered. The first step may consist of efforts to modify aggravating factors, such as dry skin, occlusion, excess heat, and rapid temperature changes. Indeed, for mild cases of GD, this may be all that is required.

The firstline pharmacotherapy for GD is medium- to high-potency topical corticosteroids, which reduce inflammation and pruritus in approximately half of affected patients.3,6,8 Topical emollients and oral antihistamines can also provide symptom relief. Vitamin D analogues are considered secondline therapy, and retinoids (both topical and systemic) have also been shown to reduce GD severity.3,4,8

Severe, refractory cases may require more aggressive systemic therapy with corticosteroids or retinoids. For pruritic relief, several weeks of oral corticosteroids may be necessary—and GD may rebound after treatment ceases.3,4 Therefore, oral corticosteroids should only be considered for severe or persistent cases, since the systemic adverse effects (eg, immunosuppression, weight gain, dysglycemia) of these drugs may outweigh the benefits in patients with GD. Other interventions, including phototherapy and immunosuppressive drugs (eg, etanercept) have also demonstrated benefit in select patients.4,9,10

The self-limited nature of GD, along with its lack of systemic symptoms, is associated with a generally benign course of disease and no long-term sequelae.3,5

Continue to outcome for the case patient >>

 

 

OUTCOME FOR THE CASE PATIENT
This case involved an immunocompromised patient with systemic symptoms, vasculitic cutaneous lesions, and significant pulmonary disease. The differential diagnosis was extensive, and diagnosis based on clinical grounds alone was extremely challenging. In these circumstances, diagnostic testing was essential to reach a final diagnosis.

In this case, the skin biopsy yielded a diagnosis of GD, and the rash was found to be unrelated to the patient’s systemic and pulmonary symptoms. The providers were then able to focus on the diagnosis of histoplasmosis, with only minimal intervention for the patient’s GD (ie, oral diphenhydramine prn for pruritus).

CONCLUSION
In many cases of GD, skin biopsy can guide providers when the history and physical examination do not yield a clear diagnosis. The histopathology of affected tissue can provide invaluable information about an underlying disease process, particularly in complex cases such as this patient’s. Skin biopsy provides a minimally invasive opportunity to obtain a diagnosis in patients with a condition that affects multiple organ systems, and its use should be considered in disease processes with cutaneous manifestations.

REFERENCES
1. Scheinfeld N, Mones J. Seasonal variation of transient acantholytic dyskeratosis (Grover’s disease). J Am Acad Dermatol. 2006;55(2): 263-268.
2. Parsons JM. Transient acantholytic dermatosis (Grover’s disease): a global perspective. J Am Acad Dermatol. 1996;35(5 part 1):653-666.
3. Weaver J, Bergfeld WF. Grover disease (transient acantholytic dermatosis). Arch Pathol Lab Med. 2009;133(9):1490-1494.
4. Quirk CJ, Heenan PJ. Grover’s disease: 34 years on. Australas J Dermatol. 2004;45(2):83-86.
5. Ippoliti G, Paulli M, Lucioni M, et al. Grover’s disease after heart transplantation: a case report. Case Rep Transplant. 2012;2012:126592.
6. Streit M, Paredes BE, Braathen LR, Brand CU. Transitory acantholytic dermatosis (Grover’s disease): an analysis of the clinical spectrum based on 21 histologically assessed cases [in German]. Hautarzt. 2000;51:244-249.
7. Joshi R, Taneja A. Grover’s disease with acrosyringeal acantholysis: a rare histological presentation of an uncommon disease. Indian J Dermatol. 2014;59(6):621-623.
8. Riemann H, High WA. Grover’s disease (transient and persistent acantholytic dermatosis). UpToDate. 2015. www.uptodate.com/contents/grovers-disease-transient-and-persistent-acantholytic-dermatosis. Accessed June 4, 2016.
9. Breuckmann F, Appelhans C, Altmeyer P, Kreuter A. Medium-dose ultraviolet A1 phototherapy in transient acantholytic dermatosis (Grover’s disease). J Am Acad Dermatol. 2005;52(1):169-170.
10. Norman R, Chau V. Use of etanercept in treating pruritus and preventing new lesions in Grover disease. J Am Acad Dermatol. 2011;64(4):796-798.

Article PDF
Author and Disclosure Information

 

Christopher Roman, MMS, PA-C, Lana Moulding, MS, PA-C

Christopher Roman is an Assistant Professor in the Physician Assistant Program at Butler University in Indianapolis. Lana Moulding practices urgent care near Nashville.

The authors have no financial relationships to disclose.

Issue
Clinician Reviews - 26(7)
Publications
Topics
Page Number
34-37
Legacy Keywords
focal acantholytic dyskeratosis, Grover disease
Sections
Author and Disclosure Information

 

Christopher Roman, MMS, PA-C, Lana Moulding, MS, PA-C

Christopher Roman is an Assistant Professor in the Physician Assistant Program at Butler University in Indianapolis. Lana Moulding practices urgent care near Nashville.

The authors have no financial relationships to disclose.

Author and Disclosure Information

 

Christopher Roman, MMS, PA-C, Lana Moulding, MS, PA-C

Christopher Roman is an Assistant Professor in the Physician Assistant Program at Butler University in Indianapolis. Lana Moulding practices urgent care near Nashville.

The authors have no financial relationships to disclose.

Article PDF
Article PDF
Related Articles

IN THIS ARTICLE

 

  • Conditions associated with increased risk for case disease
  • Outcome for the case patient
  • Differential diagnosis

A 78-year-old white man with chronic lymphocytic leukemia is admitted to the hospital with worsening cough, shortness of breath, and fever. His medical history is significant for pneumonia caused by Pneumocystis jirovecii in the past year. In the weeks preceding hospital admission, the patient developed an erythematous rash over his trunk (see photographs).

During the man’s hospital stay, this eruption becomes increasingly pruritic and spreads to his proximal extremities. His pulmonary symptoms improve slightly following the initiation of broad-spectrum antibiotic therapy (piperacillin/tazobactam and vancomycin), but CT performed one week after admission reveals worsening pulmonary disease (see image). The radiologist’s differential diagnosis includes neoplasm, fungal infection, Kaposi sarcoma, and autoimmune disease.

 

A. The patient's back shows a distribution of
lesions, with areas of excoriation caused by
scratching.
B. A close-up reveals erythematous
papules and keratotic papules.

Suspecting that the progressive rash is related to the systemic process, the provider orders a punch biopsy in an effort to reach a diagnosis with minimally invasive studies. When the patient’s clinical status further declines, he undergoes video-assisted thoracoscopic surgery to obtain an excisional biopsy of one of the pulmonary nodules. Subsequent analysis reveals fungal organisms consistent with histoplasmosis. Interestingly, in the histologic review of the skin biopsy, focal acantholytic dyskeratosis—suggestive of Grover disease—is identified.

 

CT of the chest demonstrates numerous pulmonarynodules, air bronchograms, and reticulonodularinfiltrates.

Continue for discussion >>

 

 

DISCUSSION
Grover disease (GD), also known as transient acantholytic dermatosis, is a skin condition of uncertain pathophysiology. Its clinical presentation can be difficult to distinguish from other dermopathies.1,2

Incidence
GD most commonly appears in fair-skinned persons of late middle age, with men affected at two to three times the rate seen in women.1,2 Although GD has been documented in patients ranging in age from 4 to 100, this dermopathy is rare in younger patients.1-3 Persons with a prior history of atopic dermatitis, contact dermatitis, or xerosis cutis are at increased risk for GD—likely due to an increased dermatologic sensitivity to irritants resulting from the aforementioned disorders.1,4 Risk for GD is also elevated in patients with chronic medical conditions, immunodeficiency, febrile illnesses, or malignancies (see Table 1).2-5

The true incidence of GD is not known; biopsy-proven GD is uncommon, and specific data on the incidence and prevalence of the condition are lacking. Swiss researchers who reviewed more than 30,000 skin biopsies in the late 1990s noted only 24 diagnosed cases of GD, and similar findings have been reported in the United States.1,6 However, the variable presentation and often mild nature of GD may result in cases of misdiagnosis, lack of diagnosis, or empiric treatment in the absence of a formal diagnosis.7

Causative factors
Although the pathophysiology of GD is uncertain, the most likely cause is an occlusion of the eccrine glands.3 This is followed by acantholysis, or separation of keratinocytes within the epidermis, which in turn leads to the development of vesicular lesions.

Though diagnosed most often in the winter, GD has also been associated with exposure to sunlight, heat, xerosis, and diaphoresis.1,3 Hospitalized or bedridden patients are at risk for occlusion of the eccrine glands and thus for GD. Use of certain therapies, including sulfadoxine/pyrimethamine (an antimalarial treatment), ionizing radiation, and interleukin-4, may also be precursors for the condition.2

Other exacerbating factors have been suggested, but reports are largely limited to case studies and other anecdotal publications.2 Concrete data regarding the etiology and pathophysiology of GD are still relatively scarce.

Clinical presentation
Patients with GD present with pruritic dermatitis on the trunk and proximal extremities, most classically on the anterior chest and mid back.2,3 The severity of the rash does not necessarily correlate to the degree of pruritus. Some patients report only mild pruritus, while others experience debilitating discomfort and pain. In most cases, erythematous and violaceous papules and vesicles appear first, followed by keratotic erosions.3

GD is a self-limited disorder that often resolves within a few weeks, although some cases will persist for several months.3,5 Severity and duration of symptoms appear to be correlated with increasing age; elderly patients experience worse pruritus for longer periods than do younger patients.2

Although the condition is sometimes referred to as transient acantholytic dermatosis, there are three typical presentations of GD: transient eruptive, persistent pruritic, and chronic asymptomatic.4 Transient eruptive GD presents suddenly, with intense pruritus, and tends to subside over several weeks. Persistent pruritic disease generally causes a milder pruritus, with symptoms that last for several months and are not well controlled by medication. Chronic asymptomatic GD can be difficult to treat medically, yet this form of the disease typically causes little to no irritation and requires minimal therapeutic intervention.4

Systemic symptoms of GD have not been observed. Pruritus and rash are the main features in most affected patients. However, pruritic papulovesicular eruptions are commonly seen in other conditions with similar characteristics (see Table 2,3,4), and GD is comparatively rare. While clinical appearance alone may suggest a diagnosis of GD, further testing may be needed to eliminate other conditions from the differential.

Treatment and prognosis
In the absence of randomized therapeutic trials for GD, there are no strict guidelines for treatment. When irritation, inflammation, and pruritus become bothersome, several interventions may be considered. The first step may consist of efforts to modify aggravating factors, such as dry skin, occlusion, excess heat, and rapid temperature changes. Indeed, for mild cases of GD, this may be all that is required.

The firstline pharmacotherapy for GD is medium- to high-potency topical corticosteroids, which reduce inflammation and pruritus in approximately half of affected patients.3,6,8 Topical emollients and oral antihistamines can also provide symptom relief. Vitamin D analogues are considered secondline therapy, and retinoids (both topical and systemic) have also been shown to reduce GD severity.3,4,8

Severe, refractory cases may require more aggressive systemic therapy with corticosteroids or retinoids. For pruritic relief, several weeks of oral corticosteroids may be necessary—and GD may rebound after treatment ceases.3,4 Therefore, oral corticosteroids should only be considered for severe or persistent cases, since the systemic adverse effects (eg, immunosuppression, weight gain, dysglycemia) of these drugs may outweigh the benefits in patients with GD. Other interventions, including phototherapy and immunosuppressive drugs (eg, etanercept) have also demonstrated benefit in select patients.4,9,10

The self-limited nature of GD, along with its lack of systemic symptoms, is associated with a generally benign course of disease and no long-term sequelae.3,5

Continue to outcome for the case patient >>

 

 

OUTCOME FOR THE CASE PATIENT
This case involved an immunocompromised patient with systemic symptoms, vasculitic cutaneous lesions, and significant pulmonary disease. The differential diagnosis was extensive, and diagnosis based on clinical grounds alone was extremely challenging. In these circumstances, diagnostic testing was essential to reach a final diagnosis.

In this case, the skin biopsy yielded a diagnosis of GD, and the rash was found to be unrelated to the patient’s systemic and pulmonary symptoms. The providers were then able to focus on the diagnosis of histoplasmosis, with only minimal intervention for the patient’s GD (ie, oral diphenhydramine prn for pruritus).

CONCLUSION
In many cases of GD, skin biopsy can guide providers when the history and physical examination do not yield a clear diagnosis. The histopathology of affected tissue can provide invaluable information about an underlying disease process, particularly in complex cases such as this patient’s. Skin biopsy provides a minimally invasive opportunity to obtain a diagnosis in patients with a condition that affects multiple organ systems, and its use should be considered in disease processes with cutaneous manifestations.

REFERENCES
1. Scheinfeld N, Mones J. Seasonal variation of transient acantholytic dyskeratosis (Grover’s disease). J Am Acad Dermatol. 2006;55(2): 263-268.
2. Parsons JM. Transient acantholytic dermatosis (Grover’s disease): a global perspective. J Am Acad Dermatol. 1996;35(5 part 1):653-666.
3. Weaver J, Bergfeld WF. Grover disease (transient acantholytic dermatosis). Arch Pathol Lab Med. 2009;133(9):1490-1494.
4. Quirk CJ, Heenan PJ. Grover’s disease: 34 years on. Australas J Dermatol. 2004;45(2):83-86.
5. Ippoliti G, Paulli M, Lucioni M, et al. Grover’s disease after heart transplantation: a case report. Case Rep Transplant. 2012;2012:126592.
6. Streit M, Paredes BE, Braathen LR, Brand CU. Transitory acantholytic dermatosis (Grover’s disease): an analysis of the clinical spectrum based on 21 histologically assessed cases [in German]. Hautarzt. 2000;51:244-249.
7. Joshi R, Taneja A. Grover’s disease with acrosyringeal acantholysis: a rare histological presentation of an uncommon disease. Indian J Dermatol. 2014;59(6):621-623.
8. Riemann H, High WA. Grover’s disease (transient and persistent acantholytic dermatosis). UpToDate. 2015. www.uptodate.com/contents/grovers-disease-transient-and-persistent-acantholytic-dermatosis. Accessed June 4, 2016.
9. Breuckmann F, Appelhans C, Altmeyer P, Kreuter A. Medium-dose ultraviolet A1 phototherapy in transient acantholytic dermatosis (Grover’s disease). J Am Acad Dermatol. 2005;52(1):169-170.
10. Norman R, Chau V. Use of etanercept in treating pruritus and preventing new lesions in Grover disease. J Am Acad Dermatol. 2011;64(4):796-798.

IN THIS ARTICLE

 

  • Conditions associated with increased risk for case disease
  • Outcome for the case patient
  • Differential diagnosis

A 78-year-old white man with chronic lymphocytic leukemia is admitted to the hospital with worsening cough, shortness of breath, and fever. His medical history is significant for pneumonia caused by Pneumocystis jirovecii in the past year. In the weeks preceding hospital admission, the patient developed an erythematous rash over his trunk (see photographs).

During the man’s hospital stay, this eruption becomes increasingly pruritic and spreads to his proximal extremities. His pulmonary symptoms improve slightly following the initiation of broad-spectrum antibiotic therapy (piperacillin/tazobactam and vancomycin), but CT performed one week after admission reveals worsening pulmonary disease (see image). The radiologist’s differential diagnosis includes neoplasm, fungal infection, Kaposi sarcoma, and autoimmune disease.

 

A. The patient's back shows a distribution of
lesions, with areas of excoriation caused by
scratching.
B. A close-up reveals erythematous
papules and keratotic papules.

Suspecting that the progressive rash is related to the systemic process, the provider orders a punch biopsy in an effort to reach a diagnosis with minimally invasive studies. When the patient’s clinical status further declines, he undergoes video-assisted thoracoscopic surgery to obtain an excisional biopsy of one of the pulmonary nodules. Subsequent analysis reveals fungal organisms consistent with histoplasmosis. Interestingly, in the histologic review of the skin biopsy, focal acantholytic dyskeratosis—suggestive of Grover disease—is identified.

 

CT of the chest demonstrates numerous pulmonarynodules, air bronchograms, and reticulonodularinfiltrates.

Continue for discussion >>

 

 

DISCUSSION
Grover disease (GD), also known as transient acantholytic dermatosis, is a skin condition of uncertain pathophysiology. Its clinical presentation can be difficult to distinguish from other dermopathies.1,2

Incidence
GD most commonly appears in fair-skinned persons of late middle age, with men affected at two to three times the rate seen in women.1,2 Although GD has been documented in patients ranging in age from 4 to 100, this dermopathy is rare in younger patients.1-3 Persons with a prior history of atopic dermatitis, contact dermatitis, or xerosis cutis are at increased risk for GD—likely due to an increased dermatologic sensitivity to irritants resulting from the aforementioned disorders.1,4 Risk for GD is also elevated in patients with chronic medical conditions, immunodeficiency, febrile illnesses, or malignancies (see Table 1).2-5

The true incidence of GD is not known; biopsy-proven GD is uncommon, and specific data on the incidence and prevalence of the condition are lacking. Swiss researchers who reviewed more than 30,000 skin biopsies in the late 1990s noted only 24 diagnosed cases of GD, and similar findings have been reported in the United States.1,6 However, the variable presentation and often mild nature of GD may result in cases of misdiagnosis, lack of diagnosis, or empiric treatment in the absence of a formal diagnosis.7

Causative factors
Although the pathophysiology of GD is uncertain, the most likely cause is an occlusion of the eccrine glands.3 This is followed by acantholysis, or separation of keratinocytes within the epidermis, which in turn leads to the development of vesicular lesions.

Though diagnosed most often in the winter, GD has also been associated with exposure to sunlight, heat, xerosis, and diaphoresis.1,3 Hospitalized or bedridden patients are at risk for occlusion of the eccrine glands and thus for GD. Use of certain therapies, including sulfadoxine/pyrimethamine (an antimalarial treatment), ionizing radiation, and interleukin-4, may also be precursors for the condition.2

Other exacerbating factors have been suggested, but reports are largely limited to case studies and other anecdotal publications.2 Concrete data regarding the etiology and pathophysiology of GD are still relatively scarce.

Clinical presentation
Patients with GD present with pruritic dermatitis on the trunk and proximal extremities, most classically on the anterior chest and mid back.2,3 The severity of the rash does not necessarily correlate to the degree of pruritus. Some patients report only mild pruritus, while others experience debilitating discomfort and pain. In most cases, erythematous and violaceous papules and vesicles appear first, followed by keratotic erosions.3

GD is a self-limited disorder that often resolves within a few weeks, although some cases will persist for several months.3,5 Severity and duration of symptoms appear to be correlated with increasing age; elderly patients experience worse pruritus for longer periods than do younger patients.2

Although the condition is sometimes referred to as transient acantholytic dermatosis, there are three typical presentations of GD: transient eruptive, persistent pruritic, and chronic asymptomatic.4 Transient eruptive GD presents suddenly, with intense pruritus, and tends to subside over several weeks. Persistent pruritic disease generally causes a milder pruritus, with symptoms that last for several months and are not well controlled by medication. Chronic asymptomatic GD can be difficult to treat medically, yet this form of the disease typically causes little to no irritation and requires minimal therapeutic intervention.4

Systemic symptoms of GD have not been observed. Pruritus and rash are the main features in most affected patients. However, pruritic papulovesicular eruptions are commonly seen in other conditions with similar characteristics (see Table 2,3,4), and GD is comparatively rare. While clinical appearance alone may suggest a diagnosis of GD, further testing may be needed to eliminate other conditions from the differential.

Treatment and prognosis
In the absence of randomized therapeutic trials for GD, there are no strict guidelines for treatment. When irritation, inflammation, and pruritus become bothersome, several interventions may be considered. The first step may consist of efforts to modify aggravating factors, such as dry skin, occlusion, excess heat, and rapid temperature changes. Indeed, for mild cases of GD, this may be all that is required.

The firstline pharmacotherapy for GD is medium- to high-potency topical corticosteroids, which reduce inflammation and pruritus in approximately half of affected patients.3,6,8 Topical emollients and oral antihistamines can also provide symptom relief. Vitamin D analogues are considered secondline therapy, and retinoids (both topical and systemic) have also been shown to reduce GD severity.3,4,8

Severe, refractory cases may require more aggressive systemic therapy with corticosteroids or retinoids. For pruritic relief, several weeks of oral corticosteroids may be necessary—and GD may rebound after treatment ceases.3,4 Therefore, oral corticosteroids should only be considered for severe or persistent cases, since the systemic adverse effects (eg, immunosuppression, weight gain, dysglycemia) of these drugs may outweigh the benefits in patients with GD. Other interventions, including phototherapy and immunosuppressive drugs (eg, etanercept) have also demonstrated benefit in select patients.4,9,10

The self-limited nature of GD, along with its lack of systemic symptoms, is associated with a generally benign course of disease and no long-term sequelae.3,5

Continue to outcome for the case patient >>

 

 

OUTCOME FOR THE CASE PATIENT
This case involved an immunocompromised patient with systemic symptoms, vasculitic cutaneous lesions, and significant pulmonary disease. The differential diagnosis was extensive, and diagnosis based on clinical grounds alone was extremely challenging. In these circumstances, diagnostic testing was essential to reach a final diagnosis.

In this case, the skin biopsy yielded a diagnosis of GD, and the rash was found to be unrelated to the patient’s systemic and pulmonary symptoms. The providers were then able to focus on the diagnosis of histoplasmosis, with only minimal intervention for the patient’s GD (ie, oral diphenhydramine prn for pruritus).

CONCLUSION
In many cases of GD, skin biopsy can guide providers when the history and physical examination do not yield a clear diagnosis. The histopathology of affected tissue can provide invaluable information about an underlying disease process, particularly in complex cases such as this patient’s. Skin biopsy provides a minimally invasive opportunity to obtain a diagnosis in patients with a condition that affects multiple organ systems, and its use should be considered in disease processes with cutaneous manifestations.

REFERENCES
1. Scheinfeld N, Mones J. Seasonal variation of transient acantholytic dyskeratosis (Grover’s disease). J Am Acad Dermatol. 2006;55(2): 263-268.
2. Parsons JM. Transient acantholytic dermatosis (Grover’s disease): a global perspective. J Am Acad Dermatol. 1996;35(5 part 1):653-666.
3. Weaver J, Bergfeld WF. Grover disease (transient acantholytic dermatosis). Arch Pathol Lab Med. 2009;133(9):1490-1494.
4. Quirk CJ, Heenan PJ. Grover’s disease: 34 years on. Australas J Dermatol. 2004;45(2):83-86.
5. Ippoliti G, Paulli M, Lucioni M, et al. Grover’s disease after heart transplantation: a case report. Case Rep Transplant. 2012;2012:126592.
6. Streit M, Paredes BE, Braathen LR, Brand CU. Transitory acantholytic dermatosis (Grover’s disease): an analysis of the clinical spectrum based on 21 histologically assessed cases [in German]. Hautarzt. 2000;51:244-249.
7. Joshi R, Taneja A. Grover’s disease with acrosyringeal acantholysis: a rare histological presentation of an uncommon disease. Indian J Dermatol. 2014;59(6):621-623.
8. Riemann H, High WA. Grover’s disease (transient and persistent acantholytic dermatosis). UpToDate. 2015. www.uptodate.com/contents/grovers-disease-transient-and-persistent-acantholytic-dermatosis. Accessed June 4, 2016.
9. Breuckmann F, Appelhans C, Altmeyer P, Kreuter A. Medium-dose ultraviolet A1 phototherapy in transient acantholytic dermatosis (Grover’s disease). J Am Acad Dermatol. 2005;52(1):169-170.
10. Norman R, Chau V. Use of etanercept in treating pruritus and preventing new lesions in Grover disease. J Am Acad Dermatol. 2011;64(4):796-798.

Issue
Clinician Reviews - 26(7)
Issue
Clinician Reviews - 26(7)
Page Number
34-37
Page Number
34-37
Publications
Publications
Topics
Article Type
Display Headline
Febrile, Immunocompromised Man With Rash
Display Headline
Febrile, Immunocompromised Man With Rash
Legacy Keywords
focal acantholytic dyskeratosis, Grover disease
Legacy Keywords
focal acantholytic dyskeratosis, Grover disease
Sections
Disallow All Ads
Alternative CME
Article PDF Media

Survival After Long-Term Residence in an Intensive Care Unit

Article Type
Changed
Wed, 03/27/2019 - 11:52
Display Headline
Survival After Long-Term Residence in an Intensive Care Unit
A higher mortality trend correlated with increased age and length of stay for medical and surgical patients in the intensive care unit.

Admission to an intensive care unit (ICU) is lifesaving for some patients, but for many, the admission carries high expectations and financial costs and fails to provide desirable outcomes. Patients who receive intensive care have a mortality rate of about 20%, and the costs of this care comprise about 4% of the U.S. health care budget.1,2 In a study of Medicare recipients, treatment intensity and expenses increased between the mid-1980s and 1999 but without any increase in survivorship; per capita ICU expenses were higher for patients who did not survive the ICU.3 Use of the ICU in patients’ final stages of life has increased in proportion since then, and the demand for critical care is likely to continue as the relative proportion of elderly patients in the population rises.2,4,5

Physicians and nurses who responded to a European survey on the inappropriateness of intensive care overwhelmingly endorsed the problems of “too much care” (89%) and “other patients would benefit more” (38%).6 Receiving terminal care in the ICU runs counter to the preferences of most patients.7 Therefore, the challenges are to define the true beneficiaries of critical care and to minimize the discomfort and unrealistic expectations of patients who will not benefit from intensive care.

For ICU patients, morbidity and mortality depend on the interaction of an acute insult (or a surgery), major comorbidities, and physiologic reserve. Aside from those with objective criteria of extreme illness, many patients have an indeterminate prognosis that is difficult to reliably predict.8,9 Several prognostic scores, including the APACHE (Acute Physiologic Assessment and Chronic Health Evaluation) and SOFA (Sequential Organ Failure Assessment) scores, have proved useful in understanding the illness burden of a population when comparing outcomes in different ICUs. Yet their use in assessing the survival of individual patients has not been advocated.10-15 The utility of such models is further challenged by the significant differences in survival between patients with similar illness scores; by the sometimes poor applicability of a model’s derivation cohort to other ICU populations (surgical in particular); by cases of huge disparities between actual and predicted mortality; and by the periodic need to recalibrate models according to advances in care.16-20

 

Physician intuition regarding prognosis is highly variable. In a series of medical (floor and ICU) admissions, resident physician estimates of illness severity and postdischarge status were associated with stepwise differences in mortality and APACHE scores.21,22 However, in a pure ICU population, in most cases seasoned providers could not accurately predict a patient’s chance of survival.23 Physicians are likewise poor in predicting family preferences regarding aggressive care vs alternatives, and often, survival is couched in terms of ICU survival, which for family members may not be as meaningful as long-term survival or functional recovery. Further, quality of life and patient preferences are not discussed in most cases, even those associated with poor outcomes.24 There also is a large amount of heterogeneity in the end of-life care of ICU patients. For example, cardiopulmonary resuscitation was attempted in up to 70% of dying patients in some ICUs and in as little as 4% in other ICUs.25 Thus, the limitations of predictive models, combined with misperceptions of patient preference, poor communication, and local traditions, lead to aggressive care being given to patients who might not benefit from or desire such care.

It has been stated that the trajectory of most critical illness is unclear enough so that patients should be admitted to the ICU for a trial of therapy, and that in outcome predictions, the response to intensive treatment may be more useful than laboratory and other data comprising illness severity scores.15,26 However, there is no consensus as to what constitutes a trial of intensive care therapy—vs a round of chemotherapy, a course of antibiotics, or a palliative ileostomy—yet this is the basis of many ICU admissions. Slight corrections in laboratory or physiologic findings often lead to continuation of aggressive care, often without any discussion of expected outcomes and the process of identifying and caring for patients who do not respond to therapy. Intensive care also may be prolonged because of several medical, personal, and social factors (Table 1).

At best, deciding how long to provide intensive care involves a synthesis of information about the trajectory, physiologic reserve, beliefs, values, and preferences of the patient. Any or all of these elements may not be known to the care decision-makers.

The authors conducted a study to determine whether a particular duration of care exists that represents a reasonable trial of therapy. As the VA Palo Alto Health Care System (VAPAHCS) ICU treats both medical and surgical patients, the authors were able to compare these subpopulations’ outcomes while providing the same standard of care. They analyzed the aggregate of patients as well as the medical and surgical subpopulations.

 

 

Methods

The VA Research and Development Committee and the Stanford Panel on Human Subjects approved the authors’ data collection and reporting.The study was conducted at the 15-bed mixed-medical/surgical VAPAHCS ICU. Analyzed data were drawn from all patients admitted during a 19-month period (July 14, 2008, to January 28, 2010). A serial log was used to prospectively capture basic data regarding each admission. Medical patients received care from the ICU service, and surgical patients were comanaged by the surgical and ICU teams.

A mortality database was constructed with data from the Decedent Affairs Office and from the national VistA database. The data included all deaths recorded either inside or outside the hospital or systemwide nursing facility. Mortality reported in the Computerized Patient Record System (CPRS) was queried further for patients with a length of stay (LOS) of more than 14 days.

Statistical Analysis

Calculations were based on denominators of individual patients or on number of admissions. All mortality calculations were based on a denominator of individual patients. For mortality analysis, only the last admission was included, unless a patient survived a full year between admissions. The Kruskal-Wallis test for nonnormally distributed data and the Dunn posttest for multiple comparisons were used for continuous variables (eg, age, LOS, risk scores); the Fisher exact test was used for categorical data; and the log-rank test was used to compare survival curves. For all analyses, P < .05 was considered statistically significant.

 

Mortality and Functional Status

Mortality risk scores on ICU admission were calculated with the Mortality Prediction Model–Admission III (MPM-III), using data from the CPRS. Specifics on this calculation are described in the eAppendix.

Current survival status of patients who were in the ICU more than 14 days was determined from the CPRS and telephone discussions with the patient or with relatives. Functional status was evaluated with the 36-Item Short Form Health Survey (SF-36), which has been used in comparable studies.27,28 Disposition at 6 months and 1 year was established by inspecting the CPRS for dates corresponding to these exact periods. For example, a patient in the hospital about 1 year after ICU discharge would be considered to be at home if discharged 1 day before the 365-day anniversary. In a few cases, progress notes indicated that the patient was receiving around-the-clock nursing care at home; in the analysis, these cases were included with those of patients known to be in traditional nursing facilities. In cases in which the CPRS lacked mortality information, the patient was presumed to be alive even if there were no records of clinic visits or other medical attention. Serial admission data from a mixed-medical/surgical ICU were collected over a 19-month period (July 14, 2008, to January 28, 2010) and analyzed.

Results

The final data set consisted of 1,113 admissions and 976 patients (one-third medical, two-thirds surgical). In this cohort, 12% of all patients studied were readmitted to the ICU at least once, and 12% of all ICU admissions were repeat admissions. The medical/surgical proportion was similar for readmitted patients. Demographics and other data are available in eTable 1.

Length of Stay

The distribution of all patients by LOS in the study period is shown in eFigure 1A. Data are skewed rightward toward longer LOS. The median LOS of 3 days for the entire population differed according to specialty, with a median of 3 days for medical patients (interquartile range, 2-7 days) and a median of 2 days for surgical patients (interquartile range, 1-5 days; P < .01 for medical vs surgical patients).

The LOS differed between ICU patients admitted for the first time and those readmitted within the 19-month study period. For both admission categories, LOS was longer for medical patients than for surgical patients. However, there were no significant differences between percentages of medical and surgical patients who were readmitted (Table 2). Despite comprising about 12% of the population, patients with more than 1 admission accounted for 23% of admissions and 25% of all bed occupancies during the study period.

Figure e1B shows ICU bed occupancy for different LOS intervals (calculated as bed days) and indicates that despite accounting for a small percentage of admissions, patients with long LOS accounted for a significant portion of total occupancy (32% for more than 1 month, 45% for more than 14 days). The medical and surgical contributions of these long-LOS patients were about equal. The figures indicate that more than half of medical ICU patient occupancy involved LOS of more than 14 days, while surgical patients tended to have shorter LOS.

 

 

Mortality

Of all the patients in this study, 5.1% died in the ICU; the mortality rate was 11% for medical patients and 2.1% for surgical patients. Thirty days after discharge, overall mortality was 10.4%, or 23.5% for medical patients and 3.9% for surgical patients. Finally, 1 year after discharge, mortality rates were 21.5% (overall), 39.4% (medical patients), and 12.5% (surgical patients) (Table 3). Survival curves demonstrated the difference between medical and surgical patients at 30 days and 1 year (Figures 1A & 1B).

 

Impact of LOS on Mortality

One-year mortality was 17% for patients who were in the ICU less than 14 days and 40% for those in the ICU more than 14 days (relative risk [RR] = 2.35; P < .01) (Table 4).

In the under-14-days group, mortality was significantly higher (RR = 3.3; P < .0001) for medical patients (33%) than for surgical patients (10%). A significant association between LOS and mortality was found for admissions of 0 to 7 days (r2 = 0.63; P < .05) and up to 6 weeks (r2 = 0.88; P < .01) (Figures 2A & 2B). At each LOS, mortality was significantly higher for medical patients than for surgical patients.
Survival curves of both medical and surgical patients with LOS of 0 to 7 days, 8 to 14 days, and more than 14 days showed a similar significantly higher mortality rate associated with longer ICU duration (Figures 3A & 3B).

Mortality also was higher in patients with more than 1 ICU admission. For the aggregate of ICU patients, readmission status was significantly associated with a 10% increase in mortality. For both single- and multiple-admission status, the mortality rate was 2.5-fold higher for medical patients than for surgical patients. The increased mortality associated with readmission status was not significantly different for either medical or surgical patients analyzed as subgroups (eAppendix Table.)

Impact of Age on Mortality

Figures 4A and 4B shows 30-day and 1-year mortality associated with age; regression analysis indicated that age is an independent predictor of ICU mortality. For 30-day mortality, increased age was positively associated with mortality in medical patients but not in surgical patients (r2 = 0.91; P < .0001). Age had a significant impact on 1-year mortality for both medical and surgical patients but less so in the latter (r2 = 0.95 and 0.65, respectively; P < .001 for both). Although increased mortality was associated with both LOS and age, there was no clear association between the latter 2 variables.

Survival of Chronic Critical Illness

As eTable 2 shows, 21.5% of all patients died either in the ICU or within the first year after ICU discharge. To evaluate the survival of chronic ICU residence, the authors performed a detailed analysis of functional status and mortality of patients with LOS of more than 14 days. Seventy-one patients fit that profile (their mean LOS was 41 days; median, 28 days). Of these patients, 11 died in the ICU, and another 17 died within 6 months (including 2 in a stepdown unit and 7 in hospice). Overall, 28 (39%) of the 71 patients died either in the ICU or within 6 months (35% aggregate, 53% of medical patients, and 27% of surgical patients in ICU > 2 weeks). Another 8 patients (11%) died between 6 and 12 months after discharge. One-year mortality among patients in the ICU more than 14 days was 40% overall, 50% for medical patients, and 29% for surgical patients—or twice that predicted by the MPM-III model, which figured mortality rates of 25% and 12% for medical and surgical patients, respectively. In this cohort, the mean MPM-III score was 18.6% for 1-year survivors and 29.3% for nonsurvivors (P = .016, Mann-Whitney U test). Mortality was associated with a trend toward higher MPM-III scores in both medical and surgical patients but did not reach statistical significance.

Of the cohort patients who lived at least 6 months after ICU discharge, 45% were still in a hospital or were in a nursing facility at 6 months. Of the patients who lived at least 1 year, 33% were still in a hospital or were in a nursing facility (Figure 5). At 1 year, mean age was 63 years for survivors and 69 years for the deceased (P < .01 by Student t test).

There were no significant associations among age, LOS, and nursing facility residence at 1 year. Compared with surgical patients, a larger percentage of medical patients required skilled nursing at 1 year (RR = 1.95; P = .042 by Fisher exact test).

 

 

Quality-of-Life Survey

The authors successfully contacted 32 of the 39 patients who lived at least 1 year after discharge after an ICU stay of more than 14 days. The subgroups’ median SF-36 scores were similar: 57 for medical patients and 51 for surgical patients. These average scores over 8 domains are similar to those reported by Graf and colleagues for 9 months after ICU discharge (53.7) and are lower than the normative data reported by those authors for the German population (mean, 66.5).29

Discussion

The goals of the present study were 2-fold—to gain a better understanding of the survival and functioning of patients after ICU residence and to define what may constitute a trial of therapy in ICU, or specifically to determine whether there is a particular ICU interval or point at which further care fails to improve survival. The study also compared medical and surgical subpopulations.

The main finding of this study was a 4-fold difference between ICU mortality and 1-year mortality. This mortality increase occurred in both medical and surgical patients, but there were large differences in magnitude between these groups. The survival rates generally were better than those of other general intensive care populations, though such a comparison should be made with caution, as survival differs by country, population, admitting practices, and a variety of other hospital characteristics.30,31 Although some findings of the present study may relate to its largely male U.S. veteran population, the authors believe they have provided a data-collection-and-analysis model that can be used by any hospital trying to understand the course and outcome of its ICU patients and recognizing the value of this knowledge in discussions on goals of care.

 

Mortality and LOS

As each interval of ICU residence was associated with a stepwise increase in mortality, there was no clear cutoff for diminishing return. To create a reference point, the authors analyzed the data of patients who were in an ICU more than 14 days—thinking that this duration may represent an outer limit of a reasonable trial of therapy and a measure that probably distinguishes acute from chronic critical illness.32 Use of this interval represented a conservative approach, as only 6.5% of the patients in this cohort had a LOS of more than 14 days. This small percentage of patients accounted for 45% of total bed occupancy in this study and 54% of all medical bed occupancy. In the more-than-14-days group, mortality was 37.5% for surgical patients and 46.3% for medical patients. Thus, LOS may be a dynamic measure of physiologic reserve and disease severity—reflecting variables such as response to therapy, severity of comorbidities, resistance to new problems, and rebound from chronic stress, inflammation, and catabolism. This view is supported by the nearly 2-fold higher mortality in medical patients and nearly 3-fold higher mortality in surgical patients in comparison with MPM-III predictions.

Twelve percent of all patients were admitted to ICU multiple times, and these admissions accounted for 25% of all bed occupancies. Multiple admissions indicate a high disease burden or a low physiologic reserve that prevents full recovery from critical illness. As mortality was higher in patients with multiple admissions, ICU readmission should be regarded as a marker for poor overall recovery and should prompt consideration of both initial discharge criteria and trajectory as well as goals of care.

Medical vs Surgical Patients

In this cohort, medical and surgical patients were distinguished on several grounds. Despite the similar mean age of these subpopulations, medical patients had longer LOS and higher short- and long-term mortality. These findings are not surprising, as medical patients in the ICU have high rates of end-stage disease, malignancy, and high comorbidity burden and are often admitted to have potentially life-ending conditions stabilized. Surgical patients generally are selected on their ability to withstand major systemic perturbations—palliative and emergency operations excepted—and generally have medical conditions optimized before surgery. As the expectation of postoperative survival likely biases clinician behavior toward aggressive care, some short-term survival may reflect this behavior.

In contrast, such biased behavior is not an issue in 1-year survival, which instead accurately reflects underlying health. The different slopes of medical and surgical patients on age-vs-mortality in Figures 4A & 4B indicate the different physiologic makeups of these ICU patients. With short and long LOS compared, the difference between surgical and medical patients in the ICU is striking: Sixty-one percent of all surgical bed days vs 45% of all medical bed days are for LOS less than 14 days. Nevertheless, chronic critical illness has a significant impact on both medical and surgical patients and tends to equalize some of the survival differences between these groups. These populations had similar ICU readmission rates as well as similar higher mortality rates for LOS of more than 14 days and especially for LOS of more than 1 month. With longer LOS, the survival curve of surgical patients begins to resemble that of medical patients—suggesting that the phenotype of chronic critical illness becomes the dominant force influencing survival and function (Figures 3A & 3B). Indeed, for surgical patients, the highest mortality categories were ICU readmission and LOS of more than 30 days.

 

 

The mortality rate was significantly lower for surgical patients than it was for medical patients at all intervals studied, with the largest separation in the short-term categories of ICU and 30-day mortality. The post-ICU mortality rates for medical and surgical patients are similar to those reported in several other studies, including a study of veterans.14-16,33,34 Among the present patients with LOS of more than 14 days, surviving surgical patients were significantly younger than nonsurviving surgical patients and both surviving and nonsurviving medical patients.

  

The few SF-36 responses collected revealed no differences between medical and surgical patients.

A Trial of Therapy

The present data are useful in describing the landscape of post-ICU survival to patients and their families. The data demonstrated a higher mortality trend that correlated with increases in age and increases in ICU duration and readmission. Within this continuum, there was no break point at which survivors and nonsurvivors clearly separated. The data therefore lack a boundary that can be used to define a trial of therapy. However, the added risks of age and recovery longer than 1 week are clear and should be included in care decisions. The generally better survival of surgical patients (nearly all of whom had elective surgery) in comparison with medical patients suggests these populations should be considered separately.

In the absence of a point distinguishing survivors from nonsurvivors, the authors performed a more detailed analysis of patients in the ICU for more than 14 days to provide some perspective on health care dependence in the subsequent year. That ICU survival does not necessarily equate to overall survival and independence long after ICU residence is an important matter for patients and families to consider when making decisions about critical care residence. The 14-day LOS data, though using a fairly arbitrary time point, suggest that patients who cannot recover from critical illness in less than 14 days should be advised of the range of short- and long-term mortality and the likelihood of high dependence on medical care within the subsequent year.

The concepts of hospital-dependent patient and persistent inflammation, immunosuppression, and catabolism syndrome have been introduced to describe the condition of progressive deterioration and inability to regain full independence after illness.32,35 These illness patterns deserve attention in prognosis discussions. The present study focused not on ICU survival but on 1-year mortality and functional independence, and it is these longer term outcomes that critical care professionals should consider. Intensive care units are successful in improving short-term survival, but a long line of successful ICU discharges may lead an intensivist to think that longer term survival is important as well and convey this impression to patients and their families.

Study Strengths

This study is one of a few to investigate the short- and long-term survival of an unselected cohort of critically ill patients and is unique in its inclusion of both medical and surgical patients receiving care in the same environment. Medical and surgical patients have different survival profiles that may necessitate separate studies of these subpopulations. However, the finding of different survival profiles under the same care highlights the intrinsic differences between these groups. Use of a 1.5-year study period allowed the authors to capture ICU patients with long LOS and to include multiple episodes of care provided by more than 10 different attending physicians. Therefore, these data likely were not influenced by any rare events or idiosyncrasies in practice styles. Further, the same teams of physicians and nurses cared for all the medical and surgical patients, and all unit-based protocols and quality improvement activities were applied to all patients.

Study Limitations

The intensive care patients come from a large catchment area; however, conditions seen in tertiary referral centers, such as bone marrow transplants, cerebrovascular, transplant surgery, and ventricular-assist devices are not represented in this population.

In this study, bed days were used as a crude measure of care burden. From a nursing perspective, however, the workload may be higher with quick-turnover beds than with long-term residents. On the other hand, long-term ICU residents are visited by multiple consultants and receive a much larger set of interventions, including weeks of ventilation and hemodialysis, line changes, and family meetings. A comparison of the costs involved for different ICU subpopulations would add valuable information to this discussion.

The authors took a conservative approach in establishing the mortality and residence of patients 1 year after their ICU stays. At 6 months, 1-year patients without evidence of hospital or nursing facility residence were assumed to be home. In reality, nearly all these patients had multiple admissions or emergency department stays, or there was other evidence of intensive care. Some patients who were assumed to be home may have left the area and become untraceable. All estimates of care dependence and mortality should therefore be considered minimums. The authors cannot envision how any of their estimates could overstate the morbidity and mortality.

 

 

The concept of hospital dependence is applicable to the majority of the ICU survivors, though the authors did not attempt to create a quantitative measure of this status.36 Another study limitation is that absence of hospitalization does not equal functional independence. A better definition of this status, and its application to a broad spectrum of LOS, would be a valuable adjunct to ICU decision making.

The convention by which the authors considered the first day of their study period a “fresh slate” did not adjust for the situation that some first admissions actually were readmissions. Assuming the validity of the finding that readmitted patients had a higher burden of morbidity and mortality, misclassification of admission status would tend to inflate the mortality of single-admission patients and minimize the magnitude of the differences found in this study. Similarly, an admission near the end of the study period may have been analyzed as a single admission, even if the patient was readmitted and died the next year. The latter situation also would tend to inflate the mortality of the single-admission category. None of these possible mathematical errors negates the fact that a second ICU admission should be regarded as a marker for poor recovery.

A more accurate estimate of short- and long-term prognosis likely can be obtained by examining laboratory studies and interventions such as vasopressors, dialysis, and ventilation at defined time points. Although the authors did not attempt it, development of such a model would be a valuable undertaking. They focused on describing the expected course of ICU patients and determining what patterns emerged from care duration. As this study found that the prognosis for long-term ICU residents remained guarded a long time after discharge, survival models of patients with 1- to 2-week ICU residences likely would be valuable in clinical decision making.

A quality-of-life survey was administered only to patients in the ICU longer than 2 weeks. This limited study was conducted to explore the feasibility of assessing outcomes other than survival and to determine the staffing requirements needed to research this further. A more meaningful analysis would come from a broader analysis of scores from 3 or 4 different ICU lengths of stay.

Clinician and family behavior can influence some of the outcomes measured in this study—particularly in cases in which an illness is poorly characterized and an evidence basis for decision making is lacking. In these situations, values and individual clinician judgment likely predominate, possibly introducing variability to care duration. Nevertheless, cumulative mortality 1 month or more after ICU residence would eliminate biased clinician behavior. The heterogeneity of care providers’ and families’ decision making, captured in this analysis, likely is a normal phenomenon that should help inform physicians’ understanding of prolonged ICU residence. 

References

1.  Halpern NA. Can the costs of critical care be controlled? Curr Opin Crit Care. 2009;15(6):591-596.

2.  Angus DC, Barnato AE, Linde-Zwirble WT, et al; Robert Wood Johnson Foundation ICU End-of-Life Peer Group. Use of intensive care at the end of life in the United States: an epidemiologic study. Crit Care Med. 2004;32(3):638-643.

3.  Barnato AE, McClellan MB, Kagay CR, Garber AM. Trends in inpatient treatment intensity among Medicare beneficiaries at the end of life. Health Serv Res. 2004;39(2):363-375.

4.  Teno JM, Gozalo PL, Bynum JP, et al. Change in end-of-life care for Medicare beneficiaries: site of death, place of care, and health care transitions in 2000, 2005, and 2009. JAMA. 2013;309(5):470-477.

5.  Zilberberg MD, Shorr AF. Economics at the end of life: hospital and ICU perspectives. Semin Respir Crit Care Med. 2012;33(4):362-369.

6.  Piers RD, Azoulay E, Ricou B, et al; APPROPRICUS Study Group of the Ethics Section of the ESICM. Perceptions of appropriateness of care among European and Israeli intensive care unit nurses and physicians. JAMA. 2011;306(24):2694-2703.

7.  Higginson IJ, Sen-Gupta GJ. Place of care in advanced cancer: a qualitative systematic literature review of patient p. J Palliat Med. 2000;3(3):287-300.

8.  McClish DK, Powell SH. How well can physicians estimate mortality in a medical intensive care unit? Med Decis Making. 1989;9(2):125-132.

9.  Barrera R, Nygard S, Sogoloff H, Groeger J, Wilson R. Accuracy of predictions of survival at admission to the intensive care unit. J Crit Care. 2001;16(1):32-35.

10.  Johnson AE, Kramer AA, Clifford GD. A new severity of illness scale using a subset of Acute Physiology and Chronic Health Evaluation data elements shows comparable predictive accuracy. Crit Care Med. 2013;41(7):1711-1718.

11.  D'Hoore W, Bouckaert A, Tilquin C. Practical considerations on the use of the Charlson comorbidity index with administrative data bases. J Clin Epidemiol. 1996;49(12):1429-1433.

12.  Knaus WA, Wagner DP, Zimmerman JE, Draper EA. Variations in mortality and length of stay in intensive care units. Ann Intern Med. 1993;118(10):753-761.

13.  Lemeshow S, Teres D, Klar J, Avrunin JS, Gehlbach SH, Rapoport J. Mortality Probability Models (MPM II) based on an international cohort of intensive care unit patients. JAMA. 1993;270(20):2478-2486.

14.  Render ML, Kim HM, Welsh DE, et al; VA ICU Project (VIP) Investigators. Automated intensive care unit risk adjustment: results from a national Veterans Affairs study. Crit Care Med. 2003;31(6):1638-1646.

15.  Viviand X, Gouvernet J, Granthil C, François G. Simplification of the SAPS by selecting independent variables. Intensive Care Med. 1991;17(3):164-168.

16.  Higgins TL, Teres D, Copes WS, Nathanson BH, Stark M, Kramer AA. Assessing contemporary intensive care unit outcome: an updated Mortality Probability Admission Model (MPM0-III). Crit Care Med. 2007;35(3):827-835.

17.  Knaus WA, Draper EA, Wagner DP, Zimmerman JE. APACHE II: a severity of disease classification system. Crit Care Med. 1985;13(10):818-829.

18.  Poses RM, McClish DK, Smith WR, et al. Results of report cards for patients with congestive heart failure depend on the method used to adjust for severity. Ann Intern Med. 2000;133(1):10-20.

19.  Schuster DP. Predicting outcome after ICU admission. The art and science of assessing risk. Chest. 1992;102(6):1861-1870.

20.  Teno JM, Fisher E, Hamel MB, et al. Decision-making and outcomes of prolonged ICU stays in seriously ill patients. J Am Geriatr Soc. 2000;48(suppl 5):S70-S74.

21.  Charlson M, Szatrowski TP, Peterson J, Gold J. Validation of a combined comorbidity index. J Clin Epidemiol. 1994;47(11):1245-1251.

22.  Pompei P, Charlson ME, Ales K, MacKenzie CR, Norton M. Relating patient characteristics at the time of admission to outcomes of hospitalization. J Clin Epidemiol. 1991;44(10):1063-1069.

23.  Meadow W, Pohlman A, Frain L, et al. Power and limitations of daily prognostications of death in the medical intensive care unit. Crit Care Med. 2011;39(3):474-479.

24.  Douglas SL, Daly BJ, Lipson AR. Neglect of quality-of-life considerations in intensive care unit family meetings for long-stay intensive care unit patients. Crit Care Med. 2012;40(2):461-467.

25.  Prendergast TJ, Claessens MT, Luce JM. A national survey of end-of-life care for critically ill patients. Am J Respir Crit Care Med. 1998;158(4):1163-1167.

26.  Luce JM. A history of resolving conflicts over end-of-life care in intensive care units in the United States. Crit Care Med. 2010;38(8):1623-1629.

27. Bashour CA, Yared JP, Ryan TA, et al. Long-term survival and functional capacity in cardiac surgery patients after prolonged intensive care. Crit Care Med. 2000;28(12):3847-3853.

28. Eddleston JM, White P, Guthrie E. Survival, morbidity, and quality of life after discharge from intensive care. Crit Care Med. 2000;28(7):2293-2299. 

29. Graf J, Koch M, Dujardin R, Kersten A, Janssens U. Health-related quality of life before, 1 month after, and 9 months after intensive care in medical cardiovascular and pulmonary patients. Crit Care Med. 2003;31(8):2163-2169. 

30.  Montuclard L, Garrouste-Orgeas M, Timsit JF, Misset B, De Jonghe B, Carlet J. Outcome, functional autonomy, and quality of life of elderly patients with a long-term intensive care unit stay. Crit Care Med. 2000;28(10):3389-3395.

31.  Teno JM, Harrell FE Jr, Knaus W, et al. Prediction of survival for older hospitalized patients: the HELP survival model. Hospitalized Elderly Longitudinal Project. J Am Geriatr Soc. 2000;48(suppl 5):S16-S24.

32.  Lamas D. Chronic critical illness. N Engl J Med. 2014;370(2):175-177.

33.  Konopad E, Noseworthy TW, Johnston R, Shustack A, Grace M. Quality of life measures before and one year after admission to an intensive care unit. Crit Care Med. 1995;23(10):1653-1659.

34.  Render ML, Kim HM, Deddens J, et al. Variation in outcomes in Veterans Affairs intensive care units with a computerized severity measure. Crit Care Med. 2005;33(5):930-939.

35.  Gentile LF, Cuenca AG, Efron PA, et al. Persistent inflammation and immunosuppression: a common syndrome and new horizon for surgical intensive care. J Trauma Acute Care Surg. 2012;72(6):1491-1501.

36.  Reuben DB, Tinetti ME. The hospital-dependent patient. N Engl J Med. 2014;370(8):694-697.

Article PDF
Author and Disclosure Information

Dr. Lighthall is a staff physician in the Department of Anesthesia at the VA Palo Alto Health Care System and an associate professor of anesthesiology and perioperative and pain medicine at the Stanford School of Medicine, both in California. Dr. Verduzco is an anesthesiologist at Santa Clara Valley Medical Center in San Jose, California.

Author disclosures
The authors report no actual or potential conflicts of interest with regard to this article.

Disclaimer
The opinions expressed herein are those of the authors and do not necessarily reflect those of Federal Practitioner, Frontline Medical Communications Inc., the U.S. Government, or any of its agencies. This article may discuss unlabeled or investigational use of certain drugs. Please review the complete prescribing information for specific drugs or drug combinations—including indications, contraindications, warnings, and adverse effects—before administering pharmacologic therapy to patients.

Issue
Federal Practitioner - 33(6)
Publications
Topics
Page Number
18-27
Legacy Keywords
Geoffrey Lighthall, Luis Verduzco, intensive care unit survival, ICU, ICU long-term residence
Sections
Author and Disclosure Information

Dr. Lighthall is a staff physician in the Department of Anesthesia at the VA Palo Alto Health Care System and an associate professor of anesthesiology and perioperative and pain medicine at the Stanford School of Medicine, both in California. Dr. Verduzco is an anesthesiologist at Santa Clara Valley Medical Center in San Jose, California.

Author disclosures
The authors report no actual or potential conflicts of interest with regard to this article.

Disclaimer
The opinions expressed herein are those of the authors and do not necessarily reflect those of Federal Practitioner, Frontline Medical Communications Inc., the U.S. Government, or any of its agencies. This article may discuss unlabeled or investigational use of certain drugs. Please review the complete prescribing information for specific drugs or drug combinations—including indications, contraindications, warnings, and adverse effects—before administering pharmacologic therapy to patients.

Author and Disclosure Information

Dr. Lighthall is a staff physician in the Department of Anesthesia at the VA Palo Alto Health Care System and an associate professor of anesthesiology and perioperative and pain medicine at the Stanford School of Medicine, both in California. Dr. Verduzco is an anesthesiologist at Santa Clara Valley Medical Center in San Jose, California.

Author disclosures
The authors report no actual or potential conflicts of interest with regard to this article.

Disclaimer
The opinions expressed herein are those of the authors and do not necessarily reflect those of Federal Practitioner, Frontline Medical Communications Inc., the U.S. Government, or any of its agencies. This article may discuss unlabeled or investigational use of certain drugs. Please review the complete prescribing information for specific drugs or drug combinations—including indications, contraindications, warnings, and adverse effects—before administering pharmacologic therapy to patients.

Article PDF
Article PDF
Related Articles
A higher mortality trend correlated with increased age and length of stay for medical and surgical patients in the intensive care unit.
A higher mortality trend correlated with increased age and length of stay for medical and surgical patients in the intensive care unit.

Admission to an intensive care unit (ICU) is lifesaving for some patients, but for many, the admission carries high expectations and financial costs and fails to provide desirable outcomes. Patients who receive intensive care have a mortality rate of about 20%, and the costs of this care comprise about 4% of the U.S. health care budget.1,2 In a study of Medicare recipients, treatment intensity and expenses increased between the mid-1980s and 1999 but without any increase in survivorship; per capita ICU expenses were higher for patients who did not survive the ICU.3 Use of the ICU in patients’ final stages of life has increased in proportion since then, and the demand for critical care is likely to continue as the relative proportion of elderly patients in the population rises.2,4,5

Physicians and nurses who responded to a European survey on the inappropriateness of intensive care overwhelmingly endorsed the problems of “too much care” (89%) and “other patients would benefit more” (38%).6 Receiving terminal care in the ICU runs counter to the preferences of most patients.7 Therefore, the challenges are to define the true beneficiaries of critical care and to minimize the discomfort and unrealistic expectations of patients who will not benefit from intensive care.

For ICU patients, morbidity and mortality depend on the interaction of an acute insult (or a surgery), major comorbidities, and physiologic reserve. Aside from those with objective criteria of extreme illness, many patients have an indeterminate prognosis that is difficult to reliably predict.8,9 Several prognostic scores, including the APACHE (Acute Physiologic Assessment and Chronic Health Evaluation) and SOFA (Sequential Organ Failure Assessment) scores, have proved useful in understanding the illness burden of a population when comparing outcomes in different ICUs. Yet their use in assessing the survival of individual patients has not been advocated.10-15 The utility of such models is further challenged by the significant differences in survival between patients with similar illness scores; by the sometimes poor applicability of a model’s derivation cohort to other ICU populations (surgical in particular); by cases of huge disparities between actual and predicted mortality; and by the periodic need to recalibrate models according to advances in care.16-20

 

Physician intuition regarding prognosis is highly variable. In a series of medical (floor and ICU) admissions, resident physician estimates of illness severity and postdischarge status were associated with stepwise differences in mortality and APACHE scores.21,22 However, in a pure ICU population, in most cases seasoned providers could not accurately predict a patient’s chance of survival.23 Physicians are likewise poor in predicting family preferences regarding aggressive care vs alternatives, and often, survival is couched in terms of ICU survival, which for family members may not be as meaningful as long-term survival or functional recovery. Further, quality of life and patient preferences are not discussed in most cases, even those associated with poor outcomes.24 There also is a large amount of heterogeneity in the end of-life care of ICU patients. For example, cardiopulmonary resuscitation was attempted in up to 70% of dying patients in some ICUs and in as little as 4% in other ICUs.25 Thus, the limitations of predictive models, combined with misperceptions of patient preference, poor communication, and local traditions, lead to aggressive care being given to patients who might not benefit from or desire such care.

It has been stated that the trajectory of most critical illness is unclear enough so that patients should be admitted to the ICU for a trial of therapy, and that in outcome predictions, the response to intensive treatment may be more useful than laboratory and other data comprising illness severity scores.15,26 However, there is no consensus as to what constitutes a trial of intensive care therapy—vs a round of chemotherapy, a course of antibiotics, or a palliative ileostomy—yet this is the basis of many ICU admissions. Slight corrections in laboratory or physiologic findings often lead to continuation of aggressive care, often without any discussion of expected outcomes and the process of identifying and caring for patients who do not respond to therapy. Intensive care also may be prolonged because of several medical, personal, and social factors (Table 1).

At best, deciding how long to provide intensive care involves a synthesis of information about the trajectory, physiologic reserve, beliefs, values, and preferences of the patient. Any or all of these elements may not be known to the care decision-makers.

The authors conducted a study to determine whether a particular duration of care exists that represents a reasonable trial of therapy. As the VA Palo Alto Health Care System (VAPAHCS) ICU treats both medical and surgical patients, the authors were able to compare these subpopulations’ outcomes while providing the same standard of care. They analyzed the aggregate of patients as well as the medical and surgical subpopulations.

 

 

Methods

The VA Research and Development Committee and the Stanford Panel on Human Subjects approved the authors’ data collection and reporting.The study was conducted at the 15-bed mixed-medical/surgical VAPAHCS ICU. Analyzed data were drawn from all patients admitted during a 19-month period (July 14, 2008, to January 28, 2010). A serial log was used to prospectively capture basic data regarding each admission. Medical patients received care from the ICU service, and surgical patients were comanaged by the surgical and ICU teams.

A mortality database was constructed with data from the Decedent Affairs Office and from the national VistA database. The data included all deaths recorded either inside or outside the hospital or systemwide nursing facility. Mortality reported in the Computerized Patient Record System (CPRS) was queried further for patients with a length of stay (LOS) of more than 14 days.

Statistical Analysis

Calculations were based on denominators of individual patients or on number of admissions. All mortality calculations were based on a denominator of individual patients. For mortality analysis, only the last admission was included, unless a patient survived a full year between admissions. The Kruskal-Wallis test for nonnormally distributed data and the Dunn posttest for multiple comparisons were used for continuous variables (eg, age, LOS, risk scores); the Fisher exact test was used for categorical data; and the log-rank test was used to compare survival curves. For all analyses, P < .05 was considered statistically significant.

 

Mortality and Functional Status

Mortality risk scores on ICU admission were calculated with the Mortality Prediction Model–Admission III (MPM-III), using data from the CPRS. Specifics on this calculation are described in the eAppendix.

Current survival status of patients who were in the ICU more than 14 days was determined from the CPRS and telephone discussions with the patient or with relatives. Functional status was evaluated with the 36-Item Short Form Health Survey (SF-36), which has been used in comparable studies.27,28 Disposition at 6 months and 1 year was established by inspecting the CPRS for dates corresponding to these exact periods. For example, a patient in the hospital about 1 year after ICU discharge would be considered to be at home if discharged 1 day before the 365-day anniversary. In a few cases, progress notes indicated that the patient was receiving around-the-clock nursing care at home; in the analysis, these cases were included with those of patients known to be in traditional nursing facilities. In cases in which the CPRS lacked mortality information, the patient was presumed to be alive even if there were no records of clinic visits or other medical attention. Serial admission data from a mixed-medical/surgical ICU were collected over a 19-month period (July 14, 2008, to January 28, 2010) and analyzed.

Results

The final data set consisted of 1,113 admissions and 976 patients (one-third medical, two-thirds surgical). In this cohort, 12% of all patients studied were readmitted to the ICU at least once, and 12% of all ICU admissions were repeat admissions. The medical/surgical proportion was similar for readmitted patients. Demographics and other data are available in eTable 1.

Length of Stay

The distribution of all patients by LOS in the study period is shown in eFigure 1A. Data are skewed rightward toward longer LOS. The median LOS of 3 days for the entire population differed according to specialty, with a median of 3 days for medical patients (interquartile range, 2-7 days) and a median of 2 days for surgical patients (interquartile range, 1-5 days; P < .01 for medical vs surgical patients).

The LOS differed between ICU patients admitted for the first time and those readmitted within the 19-month study period. For both admission categories, LOS was longer for medical patients than for surgical patients. However, there were no significant differences between percentages of medical and surgical patients who were readmitted (Table 2). Despite comprising about 12% of the population, patients with more than 1 admission accounted for 23% of admissions and 25% of all bed occupancies during the study period.

Figure e1B shows ICU bed occupancy for different LOS intervals (calculated as bed days) and indicates that despite accounting for a small percentage of admissions, patients with long LOS accounted for a significant portion of total occupancy (32% for more than 1 month, 45% for more than 14 days). The medical and surgical contributions of these long-LOS patients were about equal. The figures indicate that more than half of medical ICU patient occupancy involved LOS of more than 14 days, while surgical patients tended to have shorter LOS.

 

 

Mortality

Of all the patients in this study, 5.1% died in the ICU; the mortality rate was 11% for medical patients and 2.1% for surgical patients. Thirty days after discharge, overall mortality was 10.4%, or 23.5% for medical patients and 3.9% for surgical patients. Finally, 1 year after discharge, mortality rates were 21.5% (overall), 39.4% (medical patients), and 12.5% (surgical patients) (Table 3). Survival curves demonstrated the difference between medical and surgical patients at 30 days and 1 year (Figures 1A & 1B).

 

Impact of LOS on Mortality

One-year mortality was 17% for patients who were in the ICU less than 14 days and 40% for those in the ICU more than 14 days (relative risk [RR] = 2.35; P < .01) (Table 4).

In the under-14-days group, mortality was significantly higher (RR = 3.3; P < .0001) for medical patients (33%) than for surgical patients (10%). A significant association between LOS and mortality was found for admissions of 0 to 7 days (r2 = 0.63; P < .05) and up to 6 weeks (r2 = 0.88; P < .01) (Figures 2A & 2B). At each LOS, mortality was significantly higher for medical patients than for surgical patients.
Survival curves of both medical and surgical patients with LOS of 0 to 7 days, 8 to 14 days, and more than 14 days showed a similar significantly higher mortality rate associated with longer ICU duration (Figures 3A & 3B).

Mortality also was higher in patients with more than 1 ICU admission. For the aggregate of ICU patients, readmission status was significantly associated with a 10% increase in mortality. For both single- and multiple-admission status, the mortality rate was 2.5-fold higher for medical patients than for surgical patients. The increased mortality associated with readmission status was not significantly different for either medical or surgical patients analyzed as subgroups (eAppendix Table.)

Impact of Age on Mortality

Figures 4A and 4B shows 30-day and 1-year mortality associated with age; regression analysis indicated that age is an independent predictor of ICU mortality. For 30-day mortality, increased age was positively associated with mortality in medical patients but not in surgical patients (r2 = 0.91; P < .0001). Age had a significant impact on 1-year mortality for both medical and surgical patients but less so in the latter (r2 = 0.95 and 0.65, respectively; P < .001 for both). Although increased mortality was associated with both LOS and age, there was no clear association between the latter 2 variables.

Survival of Chronic Critical Illness

As eTable 2 shows, 21.5% of all patients died either in the ICU or within the first year after ICU discharge. To evaluate the survival of chronic ICU residence, the authors performed a detailed analysis of functional status and mortality of patients with LOS of more than 14 days. Seventy-one patients fit that profile (their mean LOS was 41 days; median, 28 days). Of these patients, 11 died in the ICU, and another 17 died within 6 months (including 2 in a stepdown unit and 7 in hospice). Overall, 28 (39%) of the 71 patients died either in the ICU or within 6 months (35% aggregate, 53% of medical patients, and 27% of surgical patients in ICU > 2 weeks). Another 8 patients (11%) died between 6 and 12 months after discharge. One-year mortality among patients in the ICU more than 14 days was 40% overall, 50% for medical patients, and 29% for surgical patients—or twice that predicted by the MPM-III model, which figured mortality rates of 25% and 12% for medical and surgical patients, respectively. In this cohort, the mean MPM-III score was 18.6% for 1-year survivors and 29.3% for nonsurvivors (P = .016, Mann-Whitney U test). Mortality was associated with a trend toward higher MPM-III scores in both medical and surgical patients but did not reach statistical significance.

Of the cohort patients who lived at least 6 months after ICU discharge, 45% were still in a hospital or were in a nursing facility at 6 months. Of the patients who lived at least 1 year, 33% were still in a hospital or were in a nursing facility (Figure 5). At 1 year, mean age was 63 years for survivors and 69 years for the deceased (P < .01 by Student t test).

There were no significant associations among age, LOS, and nursing facility residence at 1 year. Compared with surgical patients, a larger percentage of medical patients required skilled nursing at 1 year (RR = 1.95; P = .042 by Fisher exact test).

 

 

Quality-of-Life Survey

The authors successfully contacted 32 of the 39 patients who lived at least 1 year after discharge after an ICU stay of more than 14 days. The subgroups’ median SF-36 scores were similar: 57 for medical patients and 51 for surgical patients. These average scores over 8 domains are similar to those reported by Graf and colleagues for 9 months after ICU discharge (53.7) and are lower than the normative data reported by those authors for the German population (mean, 66.5).29

Discussion

The goals of the present study were 2-fold—to gain a better understanding of the survival and functioning of patients after ICU residence and to define what may constitute a trial of therapy in ICU, or specifically to determine whether there is a particular ICU interval or point at which further care fails to improve survival. The study also compared medical and surgical subpopulations.

The main finding of this study was a 4-fold difference between ICU mortality and 1-year mortality. This mortality increase occurred in both medical and surgical patients, but there were large differences in magnitude between these groups. The survival rates generally were better than those of other general intensive care populations, though such a comparison should be made with caution, as survival differs by country, population, admitting practices, and a variety of other hospital characteristics.30,31 Although some findings of the present study may relate to its largely male U.S. veteran population, the authors believe they have provided a data-collection-and-analysis model that can be used by any hospital trying to understand the course and outcome of its ICU patients and recognizing the value of this knowledge in discussions on goals of care.

 

Mortality and LOS

As each interval of ICU residence was associated with a stepwise increase in mortality, there was no clear cutoff for diminishing return. To create a reference point, the authors analyzed the data of patients who were in an ICU more than 14 days—thinking that this duration may represent an outer limit of a reasonable trial of therapy and a measure that probably distinguishes acute from chronic critical illness.32 Use of this interval represented a conservative approach, as only 6.5% of the patients in this cohort had a LOS of more than 14 days. This small percentage of patients accounted for 45% of total bed occupancy in this study and 54% of all medical bed occupancy. In the more-than-14-days group, mortality was 37.5% for surgical patients and 46.3% for medical patients. Thus, LOS may be a dynamic measure of physiologic reserve and disease severity—reflecting variables such as response to therapy, severity of comorbidities, resistance to new problems, and rebound from chronic stress, inflammation, and catabolism. This view is supported by the nearly 2-fold higher mortality in medical patients and nearly 3-fold higher mortality in surgical patients in comparison with MPM-III predictions.

Twelve percent of all patients were admitted to ICU multiple times, and these admissions accounted for 25% of all bed occupancies. Multiple admissions indicate a high disease burden or a low physiologic reserve that prevents full recovery from critical illness. As mortality was higher in patients with multiple admissions, ICU readmission should be regarded as a marker for poor overall recovery and should prompt consideration of both initial discharge criteria and trajectory as well as goals of care.

Medical vs Surgical Patients

In this cohort, medical and surgical patients were distinguished on several grounds. Despite the similar mean age of these subpopulations, medical patients had longer LOS and higher short- and long-term mortality. These findings are not surprising, as medical patients in the ICU have high rates of end-stage disease, malignancy, and high comorbidity burden and are often admitted to have potentially life-ending conditions stabilized. Surgical patients generally are selected on their ability to withstand major systemic perturbations—palliative and emergency operations excepted—and generally have medical conditions optimized before surgery. As the expectation of postoperative survival likely biases clinician behavior toward aggressive care, some short-term survival may reflect this behavior.

In contrast, such biased behavior is not an issue in 1-year survival, which instead accurately reflects underlying health. The different slopes of medical and surgical patients on age-vs-mortality in Figures 4A & 4B indicate the different physiologic makeups of these ICU patients. With short and long LOS compared, the difference between surgical and medical patients in the ICU is striking: Sixty-one percent of all surgical bed days vs 45% of all medical bed days are for LOS less than 14 days. Nevertheless, chronic critical illness has a significant impact on both medical and surgical patients and tends to equalize some of the survival differences between these groups. These populations had similar ICU readmission rates as well as similar higher mortality rates for LOS of more than 14 days and especially for LOS of more than 1 month. With longer LOS, the survival curve of surgical patients begins to resemble that of medical patients—suggesting that the phenotype of chronic critical illness becomes the dominant force influencing survival and function (Figures 3A & 3B). Indeed, for surgical patients, the highest mortality categories were ICU readmission and LOS of more than 30 days.

 

 

The mortality rate was significantly lower for surgical patients than it was for medical patients at all intervals studied, with the largest separation in the short-term categories of ICU and 30-day mortality. The post-ICU mortality rates for medical and surgical patients are similar to those reported in several other studies, including a study of veterans.14-16,33,34 Among the present patients with LOS of more than 14 days, surviving surgical patients were significantly younger than nonsurviving surgical patients and both surviving and nonsurviving medical patients.

  

The few SF-36 responses collected revealed no differences between medical and surgical patients.

A Trial of Therapy

The present data are useful in describing the landscape of post-ICU survival to patients and their families. The data demonstrated a higher mortality trend that correlated with increases in age and increases in ICU duration and readmission. Within this continuum, there was no break point at which survivors and nonsurvivors clearly separated. The data therefore lack a boundary that can be used to define a trial of therapy. However, the added risks of age and recovery longer than 1 week are clear and should be included in care decisions. The generally better survival of surgical patients (nearly all of whom had elective surgery) in comparison with medical patients suggests these populations should be considered separately.

In the absence of a point distinguishing survivors from nonsurvivors, the authors performed a more detailed analysis of patients in the ICU for more than 14 days to provide some perspective on health care dependence in the subsequent year. That ICU survival does not necessarily equate to overall survival and independence long after ICU residence is an important matter for patients and families to consider when making decisions about critical care residence. The 14-day LOS data, though using a fairly arbitrary time point, suggest that patients who cannot recover from critical illness in less than 14 days should be advised of the range of short- and long-term mortality and the likelihood of high dependence on medical care within the subsequent year.

The concepts of hospital-dependent patient and persistent inflammation, immunosuppression, and catabolism syndrome have been introduced to describe the condition of progressive deterioration and inability to regain full independence after illness.32,35 These illness patterns deserve attention in prognosis discussions. The present study focused not on ICU survival but on 1-year mortality and functional independence, and it is these longer term outcomes that critical care professionals should consider. Intensive care units are successful in improving short-term survival, but a long line of successful ICU discharges may lead an intensivist to think that longer term survival is important as well and convey this impression to patients and their families.

Study Strengths

This study is one of a few to investigate the short- and long-term survival of an unselected cohort of critically ill patients and is unique in its inclusion of both medical and surgical patients receiving care in the same environment. Medical and surgical patients have different survival profiles that may necessitate separate studies of these subpopulations. However, the finding of different survival profiles under the same care highlights the intrinsic differences between these groups. Use of a 1.5-year study period allowed the authors to capture ICU patients with long LOS and to include multiple episodes of care provided by more than 10 different attending physicians. Therefore, these data likely were not influenced by any rare events or idiosyncrasies in practice styles. Further, the same teams of physicians and nurses cared for all the medical and surgical patients, and all unit-based protocols and quality improvement activities were applied to all patients.

Study Limitations

The intensive care patients come from a large catchment area; however, conditions seen in tertiary referral centers, such as bone marrow transplants, cerebrovascular, transplant surgery, and ventricular-assist devices are not represented in this population.

In this study, bed days were used as a crude measure of care burden. From a nursing perspective, however, the workload may be higher with quick-turnover beds than with long-term residents. On the other hand, long-term ICU residents are visited by multiple consultants and receive a much larger set of interventions, including weeks of ventilation and hemodialysis, line changes, and family meetings. A comparison of the costs involved for different ICU subpopulations would add valuable information to this discussion.

The authors took a conservative approach in establishing the mortality and residence of patients 1 year after their ICU stays. At 6 months, 1-year patients without evidence of hospital or nursing facility residence were assumed to be home. In reality, nearly all these patients had multiple admissions or emergency department stays, or there was other evidence of intensive care. Some patients who were assumed to be home may have left the area and become untraceable. All estimates of care dependence and mortality should therefore be considered minimums. The authors cannot envision how any of their estimates could overstate the morbidity and mortality.

 

 

The concept of hospital dependence is applicable to the majority of the ICU survivors, though the authors did not attempt to create a quantitative measure of this status.36 Another study limitation is that absence of hospitalization does not equal functional independence. A better definition of this status, and its application to a broad spectrum of LOS, would be a valuable adjunct to ICU decision making.

The convention by which the authors considered the first day of their study period a “fresh slate” did not adjust for the situation that some first admissions actually were readmissions. Assuming the validity of the finding that readmitted patients had a higher burden of morbidity and mortality, misclassification of admission status would tend to inflate the mortality of single-admission patients and minimize the magnitude of the differences found in this study. Similarly, an admission near the end of the study period may have been analyzed as a single admission, even if the patient was readmitted and died the next year. The latter situation also would tend to inflate the mortality of the single-admission category. None of these possible mathematical errors negates the fact that a second ICU admission should be regarded as a marker for poor recovery.

A more accurate estimate of short- and long-term prognosis likely can be obtained by examining laboratory studies and interventions such as vasopressors, dialysis, and ventilation at defined time points. Although the authors did not attempt it, development of such a model would be a valuable undertaking. They focused on describing the expected course of ICU patients and determining what patterns emerged from care duration. As this study found that the prognosis for long-term ICU residents remained guarded a long time after discharge, survival models of patients with 1- to 2-week ICU residences likely would be valuable in clinical decision making.

A quality-of-life survey was administered only to patients in the ICU longer than 2 weeks. This limited study was conducted to explore the feasibility of assessing outcomes other than survival and to determine the staffing requirements needed to research this further. A more meaningful analysis would come from a broader analysis of scores from 3 or 4 different ICU lengths of stay.

Clinician and family behavior can influence some of the outcomes measured in this study—particularly in cases in which an illness is poorly characterized and an evidence basis for decision making is lacking. In these situations, values and individual clinician judgment likely predominate, possibly introducing variability to care duration. Nevertheless, cumulative mortality 1 month or more after ICU residence would eliminate biased clinician behavior. The heterogeneity of care providers’ and families’ decision making, captured in this analysis, likely is a normal phenomenon that should help inform physicians’ understanding of prolonged ICU residence. 

Admission to an intensive care unit (ICU) is lifesaving for some patients, but for many, the admission carries high expectations and financial costs and fails to provide desirable outcomes. Patients who receive intensive care have a mortality rate of about 20%, and the costs of this care comprise about 4% of the U.S. health care budget.1,2 In a study of Medicare recipients, treatment intensity and expenses increased between the mid-1980s and 1999 but without any increase in survivorship; per capita ICU expenses were higher for patients who did not survive the ICU.3 Use of the ICU in patients’ final stages of life has increased in proportion since then, and the demand for critical care is likely to continue as the relative proportion of elderly patients in the population rises.2,4,5

Physicians and nurses who responded to a European survey on the inappropriateness of intensive care overwhelmingly endorsed the problems of “too much care” (89%) and “other patients would benefit more” (38%).6 Receiving terminal care in the ICU runs counter to the preferences of most patients.7 Therefore, the challenges are to define the true beneficiaries of critical care and to minimize the discomfort and unrealistic expectations of patients who will not benefit from intensive care.

For ICU patients, morbidity and mortality depend on the interaction of an acute insult (or a surgery), major comorbidities, and physiologic reserve. Aside from those with objective criteria of extreme illness, many patients have an indeterminate prognosis that is difficult to reliably predict.8,9 Several prognostic scores, including the APACHE (Acute Physiologic Assessment and Chronic Health Evaluation) and SOFA (Sequential Organ Failure Assessment) scores, have proved useful in understanding the illness burden of a population when comparing outcomes in different ICUs. Yet their use in assessing the survival of individual patients has not been advocated.10-15 The utility of such models is further challenged by the significant differences in survival between patients with similar illness scores; by the sometimes poor applicability of a model’s derivation cohort to other ICU populations (surgical in particular); by cases of huge disparities between actual and predicted mortality; and by the periodic need to recalibrate models according to advances in care.16-20

 

Physician intuition regarding prognosis is highly variable. In a series of medical (floor and ICU) admissions, resident physician estimates of illness severity and postdischarge status were associated with stepwise differences in mortality and APACHE scores.21,22 However, in a pure ICU population, in most cases seasoned providers could not accurately predict a patient’s chance of survival.23 Physicians are likewise poor in predicting family preferences regarding aggressive care vs alternatives, and often, survival is couched in terms of ICU survival, which for family members may not be as meaningful as long-term survival or functional recovery. Further, quality of life and patient preferences are not discussed in most cases, even those associated with poor outcomes.24 There also is a large amount of heterogeneity in the end of-life care of ICU patients. For example, cardiopulmonary resuscitation was attempted in up to 70% of dying patients in some ICUs and in as little as 4% in other ICUs.25 Thus, the limitations of predictive models, combined with misperceptions of patient preference, poor communication, and local traditions, lead to aggressive care being given to patients who might not benefit from or desire such care.

It has been stated that the trajectory of most critical illness is unclear enough so that patients should be admitted to the ICU for a trial of therapy, and that in outcome predictions, the response to intensive treatment may be more useful than laboratory and other data comprising illness severity scores.15,26 However, there is no consensus as to what constitutes a trial of intensive care therapy—vs a round of chemotherapy, a course of antibiotics, or a palliative ileostomy—yet this is the basis of many ICU admissions. Slight corrections in laboratory or physiologic findings often lead to continuation of aggressive care, often without any discussion of expected outcomes and the process of identifying and caring for patients who do not respond to therapy. Intensive care also may be prolonged because of several medical, personal, and social factors (Table 1).

At best, deciding how long to provide intensive care involves a synthesis of information about the trajectory, physiologic reserve, beliefs, values, and preferences of the patient. Any or all of these elements may not be known to the care decision-makers.

The authors conducted a study to determine whether a particular duration of care exists that represents a reasonable trial of therapy. As the VA Palo Alto Health Care System (VAPAHCS) ICU treats both medical and surgical patients, the authors were able to compare these subpopulations’ outcomes while providing the same standard of care. They analyzed the aggregate of patients as well as the medical and surgical subpopulations.

 

 

Methods

The VA Research and Development Committee and the Stanford Panel on Human Subjects approved the authors’ data collection and reporting.The study was conducted at the 15-bed mixed-medical/surgical VAPAHCS ICU. Analyzed data were drawn from all patients admitted during a 19-month period (July 14, 2008, to January 28, 2010). A serial log was used to prospectively capture basic data regarding each admission. Medical patients received care from the ICU service, and surgical patients were comanaged by the surgical and ICU teams.

A mortality database was constructed with data from the Decedent Affairs Office and from the national VistA database. The data included all deaths recorded either inside or outside the hospital or systemwide nursing facility. Mortality reported in the Computerized Patient Record System (CPRS) was queried further for patients with a length of stay (LOS) of more than 14 days.

Statistical Analysis

Calculations were based on denominators of individual patients or on number of admissions. All mortality calculations were based on a denominator of individual patients. For mortality analysis, only the last admission was included, unless a patient survived a full year between admissions. The Kruskal-Wallis test for nonnormally distributed data and the Dunn posttest for multiple comparisons were used for continuous variables (eg, age, LOS, risk scores); the Fisher exact test was used for categorical data; and the log-rank test was used to compare survival curves. For all analyses, P < .05 was considered statistically significant.

 

Mortality and Functional Status

Mortality risk scores on ICU admission were calculated with the Mortality Prediction Model–Admission III (MPM-III), using data from the CPRS. Specifics on this calculation are described in the eAppendix.

Current survival status of patients who were in the ICU more than 14 days was determined from the CPRS and telephone discussions with the patient or with relatives. Functional status was evaluated with the 36-Item Short Form Health Survey (SF-36), which has been used in comparable studies.27,28 Disposition at 6 months and 1 year was established by inspecting the CPRS for dates corresponding to these exact periods. For example, a patient in the hospital about 1 year after ICU discharge would be considered to be at home if discharged 1 day before the 365-day anniversary. In a few cases, progress notes indicated that the patient was receiving around-the-clock nursing care at home; in the analysis, these cases were included with those of patients known to be in traditional nursing facilities. In cases in which the CPRS lacked mortality information, the patient was presumed to be alive even if there were no records of clinic visits or other medical attention. Serial admission data from a mixed-medical/surgical ICU were collected over a 19-month period (July 14, 2008, to January 28, 2010) and analyzed.

Results

The final data set consisted of 1,113 admissions and 976 patients (one-third medical, two-thirds surgical). In this cohort, 12% of all patients studied were readmitted to the ICU at least once, and 12% of all ICU admissions were repeat admissions. The medical/surgical proportion was similar for readmitted patients. Demographics and other data are available in eTable 1.

Length of Stay

The distribution of all patients by LOS in the study period is shown in eFigure 1A. Data are skewed rightward toward longer LOS. The median LOS of 3 days for the entire population differed according to specialty, with a median of 3 days for medical patients (interquartile range, 2-7 days) and a median of 2 days for surgical patients (interquartile range, 1-5 days; P < .01 for medical vs surgical patients).

The LOS differed between ICU patients admitted for the first time and those readmitted within the 19-month study period. For both admission categories, LOS was longer for medical patients than for surgical patients. However, there were no significant differences between percentages of medical and surgical patients who were readmitted (Table 2). Despite comprising about 12% of the population, patients with more than 1 admission accounted for 23% of admissions and 25% of all bed occupancies during the study period.

Figure e1B shows ICU bed occupancy for different LOS intervals (calculated as bed days) and indicates that despite accounting for a small percentage of admissions, patients with long LOS accounted for a significant portion of total occupancy (32% for more than 1 month, 45% for more than 14 days). The medical and surgical contributions of these long-LOS patients were about equal. The figures indicate that more than half of medical ICU patient occupancy involved LOS of more than 14 days, while surgical patients tended to have shorter LOS.

 

 

Mortality

Of all the patients in this study, 5.1% died in the ICU; the mortality rate was 11% for medical patients and 2.1% for surgical patients. Thirty days after discharge, overall mortality was 10.4%, or 23.5% for medical patients and 3.9% for surgical patients. Finally, 1 year after discharge, mortality rates were 21.5% (overall), 39.4% (medical patients), and 12.5% (surgical patients) (Table 3). Survival curves demonstrated the difference between medical and surgical patients at 30 days and 1 year (Figures 1A & 1B).

 

Impact of LOS on Mortality

One-year mortality was 17% for patients who were in the ICU less than 14 days and 40% for those in the ICU more than 14 days (relative risk [RR] = 2.35; P < .01) (Table 4).

In the under-14-days group, mortality was significantly higher (RR = 3.3; P < .0001) for medical patients (33%) than for surgical patients (10%). A significant association between LOS and mortality was found for admissions of 0 to 7 days (r2 = 0.63; P < .05) and up to 6 weeks (r2 = 0.88; P < .01) (Figures 2A & 2B). At each LOS, mortality was significantly higher for medical patients than for surgical patients.
Survival curves of both medical and surgical patients with LOS of 0 to 7 days, 8 to 14 days, and more than 14 days showed a similar significantly higher mortality rate associated with longer ICU duration (Figures 3A & 3B).

Mortality also was higher in patients with more than 1 ICU admission. For the aggregate of ICU patients, readmission status was significantly associated with a 10% increase in mortality. For both single- and multiple-admission status, the mortality rate was 2.5-fold higher for medical patients than for surgical patients. The increased mortality associated with readmission status was not significantly different for either medical or surgical patients analyzed as subgroups (eAppendix Table.)

Impact of Age on Mortality

Figures 4A and 4B shows 30-day and 1-year mortality associated with age; regression analysis indicated that age is an independent predictor of ICU mortality. For 30-day mortality, increased age was positively associated with mortality in medical patients but not in surgical patients (r2 = 0.91; P < .0001). Age had a significant impact on 1-year mortality for both medical and surgical patients but less so in the latter (r2 = 0.95 and 0.65, respectively; P < .001 for both). Although increased mortality was associated with both LOS and age, there was no clear association between the latter 2 variables.

Survival of Chronic Critical Illness

As eTable 2 shows, 21.5% of all patients died either in the ICU or within the first year after ICU discharge. To evaluate the survival of chronic ICU residence, the authors performed a detailed analysis of functional status and mortality of patients with LOS of more than 14 days. Seventy-one patients fit that profile (their mean LOS was 41 days; median, 28 days). Of these patients, 11 died in the ICU, and another 17 died within 6 months (including 2 in a stepdown unit and 7 in hospice). Overall, 28 (39%) of the 71 patients died either in the ICU or within 6 months (35% aggregate, 53% of medical patients, and 27% of surgical patients in ICU > 2 weeks). Another 8 patients (11%) died between 6 and 12 months after discharge. One-year mortality among patients in the ICU more than 14 days was 40% overall, 50% for medical patients, and 29% for surgical patients—or twice that predicted by the MPM-III model, which figured mortality rates of 25% and 12% for medical and surgical patients, respectively. In this cohort, the mean MPM-III score was 18.6% for 1-year survivors and 29.3% for nonsurvivors (P = .016, Mann-Whitney U test). Mortality was associated with a trend toward higher MPM-III scores in both medical and surgical patients but did not reach statistical significance.

Of the cohort patients who lived at least 6 months after ICU discharge, 45% were still in a hospital or were in a nursing facility at 6 months. Of the patients who lived at least 1 year, 33% were still in a hospital or were in a nursing facility (Figure 5). At 1 year, mean age was 63 years for survivors and 69 years for the deceased (P < .01 by Student t test).

There were no significant associations among age, LOS, and nursing facility residence at 1 year. Compared with surgical patients, a larger percentage of medical patients required skilled nursing at 1 year (RR = 1.95; P = .042 by Fisher exact test).

 

 

Quality-of-Life Survey

The authors successfully contacted 32 of the 39 patients who lived at least 1 year after discharge after an ICU stay of more than 14 days. The subgroups’ median SF-36 scores were similar: 57 for medical patients and 51 for surgical patients. These average scores over 8 domains are similar to those reported by Graf and colleagues for 9 months after ICU discharge (53.7) and are lower than the normative data reported by those authors for the German population (mean, 66.5).29

Discussion

The goals of the present study were 2-fold—to gain a better understanding of the survival and functioning of patients after ICU residence and to define what may constitute a trial of therapy in ICU, or specifically to determine whether there is a particular ICU interval or point at which further care fails to improve survival. The study also compared medical and surgical subpopulations.

The main finding of this study was a 4-fold difference between ICU mortality and 1-year mortality. This mortality increase occurred in both medical and surgical patients, but there were large differences in magnitude between these groups. The survival rates generally were better than those of other general intensive care populations, though such a comparison should be made with caution, as survival differs by country, population, admitting practices, and a variety of other hospital characteristics.30,31 Although some findings of the present study may relate to its largely male U.S. veteran population, the authors believe they have provided a data-collection-and-analysis model that can be used by any hospital trying to understand the course and outcome of its ICU patients and recognizing the value of this knowledge in discussions on goals of care.

 

Mortality and LOS

As each interval of ICU residence was associated with a stepwise increase in mortality, there was no clear cutoff for diminishing return. To create a reference point, the authors analyzed the data of patients who were in an ICU more than 14 days—thinking that this duration may represent an outer limit of a reasonable trial of therapy and a measure that probably distinguishes acute from chronic critical illness.32 Use of this interval represented a conservative approach, as only 6.5% of the patients in this cohort had a LOS of more than 14 days. This small percentage of patients accounted for 45% of total bed occupancy in this study and 54% of all medical bed occupancy. In the more-than-14-days group, mortality was 37.5% for surgical patients and 46.3% for medical patients. Thus, LOS may be a dynamic measure of physiologic reserve and disease severity—reflecting variables such as response to therapy, severity of comorbidities, resistance to new problems, and rebound from chronic stress, inflammation, and catabolism. This view is supported by the nearly 2-fold higher mortality in medical patients and nearly 3-fold higher mortality in surgical patients in comparison with MPM-III predictions.

Twelve percent of all patients were admitted to ICU multiple times, and these admissions accounted for 25% of all bed occupancies. Multiple admissions indicate a high disease burden or a low physiologic reserve that prevents full recovery from critical illness. As mortality was higher in patients with multiple admissions, ICU readmission should be regarded as a marker for poor overall recovery and should prompt consideration of both initial discharge criteria and trajectory as well as goals of care.

Medical vs Surgical Patients

In this cohort, medical and surgical patients were distinguished on several grounds. Despite the similar mean age of these subpopulations, medical patients had longer LOS and higher short- and long-term mortality. These findings are not surprising, as medical patients in the ICU have high rates of end-stage disease, malignancy, and high comorbidity burden and are often admitted to have potentially life-ending conditions stabilized. Surgical patients generally are selected on their ability to withstand major systemic perturbations—palliative and emergency operations excepted—and generally have medical conditions optimized before surgery. As the expectation of postoperative survival likely biases clinician behavior toward aggressive care, some short-term survival may reflect this behavior.

In contrast, such biased behavior is not an issue in 1-year survival, which instead accurately reflects underlying health. The different slopes of medical and surgical patients on age-vs-mortality in Figures 4A & 4B indicate the different physiologic makeups of these ICU patients. With short and long LOS compared, the difference between surgical and medical patients in the ICU is striking: Sixty-one percent of all surgical bed days vs 45% of all medical bed days are for LOS less than 14 days. Nevertheless, chronic critical illness has a significant impact on both medical and surgical patients and tends to equalize some of the survival differences between these groups. These populations had similar ICU readmission rates as well as similar higher mortality rates for LOS of more than 14 days and especially for LOS of more than 1 month. With longer LOS, the survival curve of surgical patients begins to resemble that of medical patients—suggesting that the phenotype of chronic critical illness becomes the dominant force influencing survival and function (Figures 3A & 3B). Indeed, for surgical patients, the highest mortality categories were ICU readmission and LOS of more than 30 days.

 

 

The mortality rate was significantly lower for surgical patients than it was for medical patients at all intervals studied, with the largest separation in the short-term categories of ICU and 30-day mortality. The post-ICU mortality rates for medical and surgical patients are similar to those reported in several other studies, including a study of veterans.14-16,33,34 Among the present patients with LOS of more than 14 days, surviving surgical patients were significantly younger than nonsurviving surgical patients and both surviving and nonsurviving medical patients.

  

The few SF-36 responses collected revealed no differences between medical and surgical patients.

A Trial of Therapy

The present data are useful in describing the landscape of post-ICU survival to patients and their families. The data demonstrated a higher mortality trend that correlated with increases in age and increases in ICU duration and readmission. Within this continuum, there was no break point at which survivors and nonsurvivors clearly separated. The data therefore lack a boundary that can be used to define a trial of therapy. However, the added risks of age and recovery longer than 1 week are clear and should be included in care decisions. The generally better survival of surgical patients (nearly all of whom had elective surgery) in comparison with medical patients suggests these populations should be considered separately.

In the absence of a point distinguishing survivors from nonsurvivors, the authors performed a more detailed analysis of patients in the ICU for more than 14 days to provide some perspective on health care dependence in the subsequent year. That ICU survival does not necessarily equate to overall survival and independence long after ICU residence is an important matter for patients and families to consider when making decisions about critical care residence. The 14-day LOS data, though using a fairly arbitrary time point, suggest that patients who cannot recover from critical illness in less than 14 days should be advised of the range of short- and long-term mortality and the likelihood of high dependence on medical care within the subsequent year.

The concepts of hospital-dependent patient and persistent inflammation, immunosuppression, and catabolism syndrome have been introduced to describe the condition of progressive deterioration and inability to regain full independence after illness.32,35 These illness patterns deserve attention in prognosis discussions. The present study focused not on ICU survival but on 1-year mortality and functional independence, and it is these longer term outcomes that critical care professionals should consider. Intensive care units are successful in improving short-term survival, but a long line of successful ICU discharges may lead an intensivist to think that longer term survival is important as well and convey this impression to patients and their families.

Study Strengths

This study is one of a few to investigate the short- and long-term survival of an unselected cohort of critically ill patients and is unique in its inclusion of both medical and surgical patients receiving care in the same environment. Medical and surgical patients have different survival profiles that may necessitate separate studies of these subpopulations. However, the finding of different survival profiles under the same care highlights the intrinsic differences between these groups. Use of a 1.5-year study period allowed the authors to capture ICU patients with long LOS and to include multiple episodes of care provided by more than 10 different attending physicians. Therefore, these data likely were not influenced by any rare events or idiosyncrasies in practice styles. Further, the same teams of physicians and nurses cared for all the medical and surgical patients, and all unit-based protocols and quality improvement activities were applied to all patients.

Study Limitations

The intensive care patients come from a large catchment area; however, conditions seen in tertiary referral centers, such as bone marrow transplants, cerebrovascular, transplant surgery, and ventricular-assist devices are not represented in this population.

In this study, bed days were used as a crude measure of care burden. From a nursing perspective, however, the workload may be higher with quick-turnover beds than with long-term residents. On the other hand, long-term ICU residents are visited by multiple consultants and receive a much larger set of interventions, including weeks of ventilation and hemodialysis, line changes, and family meetings. A comparison of the costs involved for different ICU subpopulations would add valuable information to this discussion.

The authors took a conservative approach in establishing the mortality and residence of patients 1 year after their ICU stays. At 6 months, 1-year patients without evidence of hospital or nursing facility residence were assumed to be home. In reality, nearly all these patients had multiple admissions or emergency department stays, or there was other evidence of intensive care. Some patients who were assumed to be home may have left the area and become untraceable. All estimates of care dependence and mortality should therefore be considered minimums. The authors cannot envision how any of their estimates could overstate the morbidity and mortality.

 

 

The concept of hospital dependence is applicable to the majority of the ICU survivors, though the authors did not attempt to create a quantitative measure of this status.36 Another study limitation is that absence of hospitalization does not equal functional independence. A better definition of this status, and its application to a broad spectrum of LOS, would be a valuable adjunct to ICU decision making.

The convention by which the authors considered the first day of their study period a “fresh slate” did not adjust for the situation that some first admissions actually were readmissions. Assuming the validity of the finding that readmitted patients had a higher burden of morbidity and mortality, misclassification of admission status would tend to inflate the mortality of single-admission patients and minimize the magnitude of the differences found in this study. Similarly, an admission near the end of the study period may have been analyzed as a single admission, even if the patient was readmitted and died the next year. The latter situation also would tend to inflate the mortality of the single-admission category. None of these possible mathematical errors negates the fact that a second ICU admission should be regarded as a marker for poor recovery.

A more accurate estimate of short- and long-term prognosis likely can be obtained by examining laboratory studies and interventions such as vasopressors, dialysis, and ventilation at defined time points. Although the authors did not attempt it, development of such a model would be a valuable undertaking. They focused on describing the expected course of ICU patients and determining what patterns emerged from care duration. As this study found that the prognosis for long-term ICU residents remained guarded a long time after discharge, survival models of patients with 1- to 2-week ICU residences likely would be valuable in clinical decision making.

A quality-of-life survey was administered only to patients in the ICU longer than 2 weeks. This limited study was conducted to explore the feasibility of assessing outcomes other than survival and to determine the staffing requirements needed to research this further. A more meaningful analysis would come from a broader analysis of scores from 3 or 4 different ICU lengths of stay.

Clinician and family behavior can influence some of the outcomes measured in this study—particularly in cases in which an illness is poorly characterized and an evidence basis for decision making is lacking. In these situations, values and individual clinician judgment likely predominate, possibly introducing variability to care duration. Nevertheless, cumulative mortality 1 month or more after ICU residence would eliminate biased clinician behavior. The heterogeneity of care providers’ and families’ decision making, captured in this analysis, likely is a normal phenomenon that should help inform physicians’ understanding of prolonged ICU residence. 

References

1.  Halpern NA. Can the costs of critical care be controlled? Curr Opin Crit Care. 2009;15(6):591-596.

2.  Angus DC, Barnato AE, Linde-Zwirble WT, et al; Robert Wood Johnson Foundation ICU End-of-Life Peer Group. Use of intensive care at the end of life in the United States: an epidemiologic study. Crit Care Med. 2004;32(3):638-643.

3.  Barnato AE, McClellan MB, Kagay CR, Garber AM. Trends in inpatient treatment intensity among Medicare beneficiaries at the end of life. Health Serv Res. 2004;39(2):363-375.

4.  Teno JM, Gozalo PL, Bynum JP, et al. Change in end-of-life care for Medicare beneficiaries: site of death, place of care, and health care transitions in 2000, 2005, and 2009. JAMA. 2013;309(5):470-477.

5.  Zilberberg MD, Shorr AF. Economics at the end of life: hospital and ICU perspectives. Semin Respir Crit Care Med. 2012;33(4):362-369.

6.  Piers RD, Azoulay E, Ricou B, et al; APPROPRICUS Study Group of the Ethics Section of the ESICM. Perceptions of appropriateness of care among European and Israeli intensive care unit nurses and physicians. JAMA. 2011;306(24):2694-2703.

7.  Higginson IJ, Sen-Gupta GJ. Place of care in advanced cancer: a qualitative systematic literature review of patient p. J Palliat Med. 2000;3(3):287-300.

8.  McClish DK, Powell SH. How well can physicians estimate mortality in a medical intensive care unit? Med Decis Making. 1989;9(2):125-132.

9.  Barrera R, Nygard S, Sogoloff H, Groeger J, Wilson R. Accuracy of predictions of survival at admission to the intensive care unit. J Crit Care. 2001;16(1):32-35.

10.  Johnson AE, Kramer AA, Clifford GD. A new severity of illness scale using a subset of Acute Physiology and Chronic Health Evaluation data elements shows comparable predictive accuracy. Crit Care Med. 2013;41(7):1711-1718.

11.  D'Hoore W, Bouckaert A, Tilquin C. Practical considerations on the use of the Charlson comorbidity index with administrative data bases. J Clin Epidemiol. 1996;49(12):1429-1433.

12.  Knaus WA, Wagner DP, Zimmerman JE, Draper EA. Variations in mortality and length of stay in intensive care units. Ann Intern Med. 1993;118(10):753-761.

13.  Lemeshow S, Teres D, Klar J, Avrunin JS, Gehlbach SH, Rapoport J. Mortality Probability Models (MPM II) based on an international cohort of intensive care unit patients. JAMA. 1993;270(20):2478-2486.

14.  Render ML, Kim HM, Welsh DE, et al; VA ICU Project (VIP) Investigators. Automated intensive care unit risk adjustment: results from a national Veterans Affairs study. Crit Care Med. 2003;31(6):1638-1646.

15.  Viviand X, Gouvernet J, Granthil C, François G. Simplification of the SAPS by selecting independent variables. Intensive Care Med. 1991;17(3):164-168.

16.  Higgins TL, Teres D, Copes WS, Nathanson BH, Stark M, Kramer AA. Assessing contemporary intensive care unit outcome: an updated Mortality Probability Admission Model (MPM0-III). Crit Care Med. 2007;35(3):827-835.

17.  Knaus WA, Draper EA, Wagner DP, Zimmerman JE. APACHE II: a severity of disease classification system. Crit Care Med. 1985;13(10):818-829.

18.  Poses RM, McClish DK, Smith WR, et al. Results of report cards for patients with congestive heart failure depend on the method used to adjust for severity. Ann Intern Med. 2000;133(1):10-20.

19.  Schuster DP. Predicting outcome after ICU admission. The art and science of assessing risk. Chest. 1992;102(6):1861-1870.

20.  Teno JM, Fisher E, Hamel MB, et al. Decision-making and outcomes of prolonged ICU stays in seriously ill patients. J Am Geriatr Soc. 2000;48(suppl 5):S70-S74.

21.  Charlson M, Szatrowski TP, Peterson J, Gold J. Validation of a combined comorbidity index. J Clin Epidemiol. 1994;47(11):1245-1251.

22.  Pompei P, Charlson ME, Ales K, MacKenzie CR, Norton M. Relating patient characteristics at the time of admission to outcomes of hospitalization. J Clin Epidemiol. 1991;44(10):1063-1069.

23.  Meadow W, Pohlman A, Frain L, et al. Power and limitations of daily prognostications of death in the medical intensive care unit. Crit Care Med. 2011;39(3):474-479.

24.  Douglas SL, Daly BJ, Lipson AR. Neglect of quality-of-life considerations in intensive care unit family meetings for long-stay intensive care unit patients. Crit Care Med. 2012;40(2):461-467.

25.  Prendergast TJ, Claessens MT, Luce JM. A national survey of end-of-life care for critically ill patients. Am J Respir Crit Care Med. 1998;158(4):1163-1167.

26.  Luce JM. A history of resolving conflicts over end-of-life care in intensive care units in the United States. Crit Care Med. 2010;38(8):1623-1629.

27. Bashour CA, Yared JP, Ryan TA, et al. Long-term survival and functional capacity in cardiac surgery patients after prolonged intensive care. Crit Care Med. 2000;28(12):3847-3853.

28. Eddleston JM, White P, Guthrie E. Survival, morbidity, and quality of life after discharge from intensive care. Crit Care Med. 2000;28(7):2293-2299. 

29. Graf J, Koch M, Dujardin R, Kersten A, Janssens U. Health-related quality of life before, 1 month after, and 9 months after intensive care in medical cardiovascular and pulmonary patients. Crit Care Med. 2003;31(8):2163-2169. 

30.  Montuclard L, Garrouste-Orgeas M, Timsit JF, Misset B, De Jonghe B, Carlet J. Outcome, functional autonomy, and quality of life of elderly patients with a long-term intensive care unit stay. Crit Care Med. 2000;28(10):3389-3395.

31.  Teno JM, Harrell FE Jr, Knaus W, et al. Prediction of survival for older hospitalized patients: the HELP survival model. Hospitalized Elderly Longitudinal Project. J Am Geriatr Soc. 2000;48(suppl 5):S16-S24.

32.  Lamas D. Chronic critical illness. N Engl J Med. 2014;370(2):175-177.

33.  Konopad E, Noseworthy TW, Johnston R, Shustack A, Grace M. Quality of life measures before and one year after admission to an intensive care unit. Crit Care Med. 1995;23(10):1653-1659.

34.  Render ML, Kim HM, Deddens J, et al. Variation in outcomes in Veterans Affairs intensive care units with a computerized severity measure. Crit Care Med. 2005;33(5):930-939.

35.  Gentile LF, Cuenca AG, Efron PA, et al. Persistent inflammation and immunosuppression: a common syndrome and new horizon for surgical intensive care. J Trauma Acute Care Surg. 2012;72(6):1491-1501.

36.  Reuben DB, Tinetti ME. The hospital-dependent patient. N Engl J Med. 2014;370(8):694-697.

References

1.  Halpern NA. Can the costs of critical care be controlled? Curr Opin Crit Care. 2009;15(6):591-596.

2.  Angus DC, Barnato AE, Linde-Zwirble WT, et al; Robert Wood Johnson Foundation ICU End-of-Life Peer Group. Use of intensive care at the end of life in the United States: an epidemiologic study. Crit Care Med. 2004;32(3):638-643.

3.  Barnato AE, McClellan MB, Kagay CR, Garber AM. Trends in inpatient treatment intensity among Medicare beneficiaries at the end of life. Health Serv Res. 2004;39(2):363-375.

4.  Teno JM, Gozalo PL, Bynum JP, et al. Change in end-of-life care for Medicare beneficiaries: site of death, place of care, and health care transitions in 2000, 2005, and 2009. JAMA. 2013;309(5):470-477.

5.  Zilberberg MD, Shorr AF. Economics at the end of life: hospital and ICU perspectives. Semin Respir Crit Care Med. 2012;33(4):362-369.

6.  Piers RD, Azoulay E, Ricou B, et al; APPROPRICUS Study Group of the Ethics Section of the ESICM. Perceptions of appropriateness of care among European and Israeli intensive care unit nurses and physicians. JAMA. 2011;306(24):2694-2703.

7.  Higginson IJ, Sen-Gupta GJ. Place of care in advanced cancer: a qualitative systematic literature review of patient p. J Palliat Med. 2000;3(3):287-300.

8.  McClish DK, Powell SH. How well can physicians estimate mortality in a medical intensive care unit? Med Decis Making. 1989;9(2):125-132.

9.  Barrera R, Nygard S, Sogoloff H, Groeger J, Wilson R. Accuracy of predictions of survival at admission to the intensive care unit. J Crit Care. 2001;16(1):32-35.

10.  Johnson AE, Kramer AA, Clifford GD. A new severity of illness scale using a subset of Acute Physiology and Chronic Health Evaluation data elements shows comparable predictive accuracy. Crit Care Med. 2013;41(7):1711-1718.

11.  D'Hoore W, Bouckaert A, Tilquin C. Practical considerations on the use of the Charlson comorbidity index with administrative data bases. J Clin Epidemiol. 1996;49(12):1429-1433.

12.  Knaus WA, Wagner DP, Zimmerman JE, Draper EA. Variations in mortality and length of stay in intensive care units. Ann Intern Med. 1993;118(10):753-761.

13.  Lemeshow S, Teres D, Klar J, Avrunin JS, Gehlbach SH, Rapoport J. Mortality Probability Models (MPM II) based on an international cohort of intensive care unit patients. JAMA. 1993;270(20):2478-2486.

14.  Render ML, Kim HM, Welsh DE, et al; VA ICU Project (VIP) Investigators. Automated intensive care unit risk adjustment: results from a national Veterans Affairs study. Crit Care Med. 2003;31(6):1638-1646.

15.  Viviand X, Gouvernet J, Granthil C, François G. Simplification of the SAPS by selecting independent variables. Intensive Care Med. 1991;17(3):164-168.

16.  Higgins TL, Teres D, Copes WS, Nathanson BH, Stark M, Kramer AA. Assessing contemporary intensive care unit outcome: an updated Mortality Probability Admission Model (MPM0-III). Crit Care Med. 2007;35(3):827-835.

17.  Knaus WA, Draper EA, Wagner DP, Zimmerman JE. APACHE II: a severity of disease classification system. Crit Care Med. 1985;13(10):818-829.

18.  Poses RM, McClish DK, Smith WR, et al. Results of report cards for patients with congestive heart failure depend on the method used to adjust for severity. Ann Intern Med. 2000;133(1):10-20.

19.  Schuster DP. Predicting outcome after ICU admission. The art and science of assessing risk. Chest. 1992;102(6):1861-1870.

20.  Teno JM, Fisher E, Hamel MB, et al. Decision-making and outcomes of prolonged ICU stays in seriously ill patients. J Am Geriatr Soc. 2000;48(suppl 5):S70-S74.

21.  Charlson M, Szatrowski TP, Peterson J, Gold J. Validation of a combined comorbidity index. J Clin Epidemiol. 1994;47(11):1245-1251.

22.  Pompei P, Charlson ME, Ales K, MacKenzie CR, Norton M. Relating patient characteristics at the time of admission to outcomes of hospitalization. J Clin Epidemiol. 1991;44(10):1063-1069.

23.  Meadow W, Pohlman A, Frain L, et al. Power and limitations of daily prognostications of death in the medical intensive care unit. Crit Care Med. 2011;39(3):474-479.

24.  Douglas SL, Daly BJ, Lipson AR. Neglect of quality-of-life considerations in intensive care unit family meetings for long-stay intensive care unit patients. Crit Care Med. 2012;40(2):461-467.

25.  Prendergast TJ, Claessens MT, Luce JM. A national survey of end-of-life care for critically ill patients. Am J Respir Crit Care Med. 1998;158(4):1163-1167.

26.  Luce JM. A history of resolving conflicts over end-of-life care in intensive care units in the United States. Crit Care Med. 2010;38(8):1623-1629.

27. Bashour CA, Yared JP, Ryan TA, et al. Long-term survival and functional capacity in cardiac surgery patients after prolonged intensive care. Crit Care Med. 2000;28(12):3847-3853.

28. Eddleston JM, White P, Guthrie E. Survival, morbidity, and quality of life after discharge from intensive care. Crit Care Med. 2000;28(7):2293-2299. 

29. Graf J, Koch M, Dujardin R, Kersten A, Janssens U. Health-related quality of life before, 1 month after, and 9 months after intensive care in medical cardiovascular and pulmonary patients. Crit Care Med. 2003;31(8):2163-2169. 

30.  Montuclard L, Garrouste-Orgeas M, Timsit JF, Misset B, De Jonghe B, Carlet J. Outcome, functional autonomy, and quality of life of elderly patients with a long-term intensive care unit stay. Crit Care Med. 2000;28(10):3389-3395.

31.  Teno JM, Harrell FE Jr, Knaus W, et al. Prediction of survival for older hospitalized patients: the HELP survival model. Hospitalized Elderly Longitudinal Project. J Am Geriatr Soc. 2000;48(suppl 5):S16-S24.

32.  Lamas D. Chronic critical illness. N Engl J Med. 2014;370(2):175-177.

33.  Konopad E, Noseworthy TW, Johnston R, Shustack A, Grace M. Quality of life measures before and one year after admission to an intensive care unit. Crit Care Med. 1995;23(10):1653-1659.

34.  Render ML, Kim HM, Deddens J, et al. Variation in outcomes in Veterans Affairs intensive care units with a computerized severity measure. Crit Care Med. 2005;33(5):930-939.

35.  Gentile LF, Cuenca AG, Efron PA, et al. Persistent inflammation and immunosuppression: a common syndrome and new horizon for surgical intensive care. J Trauma Acute Care Surg. 2012;72(6):1491-1501.

36.  Reuben DB, Tinetti ME. The hospital-dependent patient. N Engl J Med. 2014;370(8):694-697.

Issue
Federal Practitioner - 33(6)
Issue
Federal Practitioner - 33(6)
Page Number
18-27
Page Number
18-27
Publications
Publications
Topics
Article Type
Display Headline
Survival After Long-Term Residence in an Intensive Care Unit
Display Headline
Survival After Long-Term Residence in an Intensive Care Unit
Legacy Keywords
Geoffrey Lighthall, Luis Verduzco, intensive care unit survival, ICU, ICU long-term residence
Legacy Keywords
Geoffrey Lighthall, Luis Verduzco, intensive care unit survival, ICU, ICU long-term residence
Sections
Disallow All Ads
Alternative CME
Use ProPublica
Article PDF Media

Disease-Modifying Therapies in Multiple Sclerosis: Overview and Treatment Considerations

Article Type
Changed
Tue, 04/24/2018 - 16:18
Display Headline
Disease-Modifying Therapies in Multiple Sclerosis: Overview and Treatment Considerations
Controlling symptoms can slow the physical and emotional disabilities associated with multiple sclerosis and help patients attain the highest quality of life possible for as long as possible.

Multiple sclerosis (MS) is a disorder characterized by inflammation, demyelination, and degeneration of the central nervous system (CNS). The hallmark of the disorder is relapses and remissions of neurologic symptoms occurring early in the disease course, which are often associated with areas of CNS inflammation and myelin loss.1-3 The inciting cause for this inflammation is unknown but is believed to be multifactorial, with environmental and genetic influences creating an adaptive, T cell-mediated autoimmune response against the CNS.4 Separate from the acute attacks, progressive neurodegeneration can occur more chronically and is characterized by axonal loss and grey matter atrophy thought to be due to direct cytotoxic activity of the innate immune system as well as toxic intermediates, such as nitric oxide.4,5 Despite the multiple insults early on, neurologic disability typically becomes more apparent over time.6 The disability threshold theory argues that neurologic function compensates for brain tissue loss until a threshold of accumulated damage is exceeded.7

Background

The incidence of MS follows a geographic gradient; rates rise as the distance from the equator increases.8,9 This is thought to be due to the gradient of relative sun exposure and its role in the production of vitamin D, which plays an important role in immune regulation when converted to its active hormonal form. Multiple sclerosis is more prevalent in non-Hispanic white patients than it is in other racial groups, and women are affected nearly 2 to 3 times more often than are men.10 About 450,000 individuals in the U.S. and more than 2 million worldwide have MS.11-14

Multiple sclerosis is the most common cause of nontraumatic neurologic disability in young adults. It is typically diagnosed in the third and fourth decades of life, and those who are diagnosed after age 50 years often can recount neurologic symptoms that began years before. However, pediatric-onset and new-onset cases in the elderly have been reported. It has been estimated that up to 10% of patients with MS have onset before 18 years of age.15-17 Compared with adult-onset MS, pediatric-onset is associated with a longer period between initial attack and physical disability, although the average age of disability onset is about 10 years younger.17,18

 

Disease Courses

Relapsing-remitting MS (RRMS) is the most common disease course overall, and this pattern affects 97% of individuals with disease onset before age 18 years.15-17 The clinically isolated syndrome disease course leads to clinically definite MS in one-third of patients within 1 year and in one-half of patients within 2 years.19 In the majority of cases, the RRMS course transitions over time to secondary-progressive MS (SPMS), which is a disease pattern of progressively worsening disability with few neurologic relapses. Although inflammation is present at all stages, the difference is in the predominance of cell types involved.5 Why the shift from active to chronic inflammation occurs and how to prevent it remain central questions in MS research.4 Regardless, tentative evidence suggests that prevention of relapses may reduce disability accumulation and risk of conversion to progressive MS.20

A minority of patients with MS are diagnosed with primary-progressive MS (PPMS) at onset, which is characterized by a disease pattern that follows a relatively steady progression of neurologic symptoms over time, without clear relapses or remissions of these symptoms, though phases of stability or fluctuations in disability may still occur.21 It is typically diagnosed at an older age than is RRMS, and it is rare in children; suspicion of PPMS in this age group should prompt detailed assessment of alternative diagnoses.17,22 Primary-progressive MS is more equally distributed in men and women than is RRMS.

Regardless of onset type, disability progression seems to occur at the same rate among all patients with MS after a certain threshold is reached. The established assessment scale for disability progression in MS is the Kurtzke Expanded Disability Status scale (EDSS), which has a scoring range from 0 to 10. Data from several patient registries have shown that once EDSS step 4 is reached, progression thereafter occurs at a predictable rate that is similar across MS phenotypes.23 The time it takes patients to subsequently reach higher EDSS steps may be independent of preceding factors.23

MS Symptom Burden

The neurologic symptoms that patients experience are fluctuating and disabling throughout the disease course, irrespective of onset type. Typical MS symptoms include mobility impairment, changes in cognition and mood, pain and other sensation disturbances, bowel and bladder dysfunction, fatigue, and visual disturbances. The burden of these symptoms can significantly impact quality of life (QOL) for patients and their families. The symptom burden can pose a direct threat to a patient’s autonomy, necessitating adaptation to an unpredictable disease that requires frequent health care visits to many different health care providers (eg, neurologists; primary care providers; physiatrists; urologists; ophthalmologists; and speech, physical, and occupational therapists), periodic testing, and often costly medications.24

 

 

Compared with patients who have other chronic conditions, patients with MS experience diminished societal roles, along with decreased assessments in health, energy, and physical functions.25 These often lead to early exit from the workforce and limitations in household responsibilities, which further impact QOL.26 Including both direct and indirect costs of the disease, a patient with MS can expect a lifetime financial burden of nearly $1.2 million.27

Large population cohort studies in MS, along with MS registry studies of patients untreated with disease-modifying therapies, have shown reduced survival rates by an average of 7 to 14 years.23,28 Multiple sclerosis is the main cause of death in about 50% of cases (EDSS step 10), which is defined as “acute death due to brain stem involvement or to respiratory failure, or death consequent to the chronic bedridden state with terminal pneumonia, sepsis, uremia, or cardiorespiratory failure [and excluding] intercurrent causes of death.”23 For the remaining patients with MS, cause of death is similar to those of the general population, such as cardiovascular disease and cancer.23 However, the incidence of suicide is higher among patients with MS.23

All these factors underscore the importance of early diagnosis as well as early initiation of effective disease-modifying therapy.

The diagnosis of MS is difficult largely due to the lack of definitive diagnostic testing and specific biomarkers for disease activity and because of the wide range of differential diagnoses that can mimic MS.19,21,29 Diagnosis of MS requires that more likely diagnoses have been excluded as well as that lesions (scleroses) are disseminated in space within the CNS and disseminated in time. The 2010 Revised McDonald Diagnostic Criteria for MS are outlined in Table 1.

Disease-Modifying Therapies

The goal of MS disease-modifying therapy is to reduce the early clinical and subclinical disease activity that eventually contributes to long-term disability.31,32 There are currently 13 FDA-approved disease-modifying therapies for MS. These include 7 self-injecting therapies, 3 oral therapies, and 3 infusion therapies. These 13 medications have 8 different mechanisms of action (MOA) that target distinct areas of the immune-mediated disease process. They also differ in their frequencies and routes of administration in addition to their adverse effect (AE) profiles (Tables 2, 3, and 4).

 
 

Treatment Considerations

In 1993, interferon beta-1b became the first FDA-approved MS medication. In the following 2 decades, there became 12 additional FDA-approved medications for MS, beginning with other injectables. The first infusion therapy was introduced in 2004, followed by various oral medications. The treatment landscape continues to change rapidly. This therapeutic revolution has occurred largely due to the improved understanding of the pathophysiology of MS and unquestionably has improved the prognosis and overall QOL for patients. The question is no longer how to treat MS but rather how to personalize and optimize treatment for each patient.20

Despite all available treatment options, none are curative, and none have been proven to offer neuroprotection or contribute to neural repair. To date, no studies have led to FDA-approved therapies for PPMS. Further, the efficacy of any of these medications varies from patient to patient. Due largely to the lack of biomarkers for disease activity and treatment response, drug efficacy continues to be measured according to the current gold standard, which is identification of gadolinium-enhancing lesions in the white matter on magnetic resonance imaging (MRI), combined with other markers of disease, including clinical relapse rate and confirmed disability progression.19 In general, the injectable therapies are expected to protect against about 20% to 35% of relapses; the oral agents, 50% to 55%; and the infusion therapies, > 60%.2

In conjunction with a medication’s efficacy rate and safety profile, the frequency and route of administration also must be considered. In general, MS medications are exceedingly expensive, some costing up to tens-of-thousands of dollars per year.48 All these factors have the real potential to negatively impact patient adherence. Nonadherence and gaps in treatment have been correlated with increased rates of relapses and progression of disability as well as negative MRI outcomes.49-53

When to Initiate Treatment

Once a patient is diagnosed, a common question is, when is the right time to initiate treatment? The primary target of the current MS medications is to decrease CNS inflammation (relapses). The ideal time to start treatment is as promptly as possible after confirmation of the diagnosis to combat the early inflammatory relapsing phase of the disease. There seems to be an early window in the disease course when achieving disease control can have a profound impact on long-term disability. Disease control is typically defined as decreasing relapses, slowing the accumulation of lesions visualized on MRI, and preventing the disability that occurs from both incomplete recovery after relapses and overall disease progression.54,55

 

 

Certain clinical indicators, such as higher relapse rates early in the disease course and MRI characteristics, including total lesion burden and the location of lesions within the CNS, seem to be associated with a higher risk of disease progression.56 These are potential prognostic indicators that can help tailor the choice of disease-modifying therapy for patients.57 Those with highly inflammatory and potentially aggressive disease at onset, for example, may benefit from early initiation of higher efficacy therapies, whereas those with more benign forms of MS at onset may fare well on lower efficacy therapies. In general, when it comes to currently available MS treatments, higher efficacy is often tied to riskier AE profiles, so the best medication may be the “least efficacious” one that can still control the disease.20

Hauser and colleagues suggested a treatment decision-making model that identifies the interferons, glatiramer acetate, dimethyl fumarate, and teriflunomide as acceptable first-line therapies; fingolimod and natalizumab as acceptable second-line options; and mitoxantrone and alemtuzumab as acceptable third-line therapeutic options.20 The authors generally agree with Hauser and colleagues’ model, and it is important to consider individual patient factors (eg, comorbidities, concurrent medications, life circumstances) and disease severity when deciding on a treatment plan.

Perhaps an even more difficult question is, when is the right time to switch therapies? There remains a dearth of either guidelines or comparative studies for treatment management decisions. Further, without reliable biomarkers, the clinical and pathologic heterogeneity of MS makes treatment difficult.4,19 In practice, there is general consensus that 1 year of treatment monitoring for effects on clinical and radiologic outcomes is an acceptable time frame to evaluate effectiveness of a disease-modifying treatment. If adherence is maintained and there is still evidence of clinical or MRI activity (suggesting a suboptimal response), an alternative therapy, particularly one with a different MOA, should be strongly considered. This highlights the importance of broad access to all available MS therapies to allow for early selection of a correct therapy that patients will remain adherent to and that controls their disease.

 

Conclusion

Multiple sclerosis remains a highly unpredictable disease, and relapses have the ability to produce a measurable and sustained impact on the level of disability.58 Still, the influence of reduced relapses on preventing disability in an individual patient remains unclear. Large, long-term, prospective cohort studies may clarify whether early treatment affects disease progression and disability.20 However, it is quite evident that effective relapse reduction decreases discomfort, reduces days lost from work and other important activities of daily life, and improves QOL.58,59

There is still much to learn about this unique disease, but emerging evidence in the medical literature highlights the importance of setting treatment goals that include targeting disease activity to achieve early and effective control. Attaining control with a MS medication seems to be a key component of slowing the physical and emotional disability that can accumulate, helping patients remain active and maintain the highest QOL possible for as long as possible.

References

1.  Lublin FD, Reingold SC. Defining the clinical course of multiple sclerosis: results of an international survey. National Multiple Sclerosis Society (USA) Advisory Committee on Clinical Trials of New Agents in Multiple Sclerosis. Neurology. 1996;46(4):907-911. 

2.  Frischer JM, Bramow S, Dal-Bianco A, et al. The relation between inflammation and neurodegeneration in multiple sclerosis brains. Brain. 2009;132(pt 5):1175-1189. 

3.  Charil A, Filippi M. Inflammatory demyelination and neurodegeneration in early multiple sclerosis. J Neurol Sci. 2007;259(1-2):7-15.

4.  Weiner HL. The challenge of multiple sclerosis: how do we cure a chronic heterogeneous disease? Ann Neurol. 2009;65(3):239-248.

5.  Grigoriadis N, van Pesch V; ParadigMS Group. A basic overview of multiple sclerosis immunopathology. Eur J Neurol. 2015;22(suppl 2):3-13. 

6.  Lassmann H, van Horssen J, Mahad D. Progressive multiple sclerosis: pathology and pathogenesis. Nat Rev Neurol. 2012;8(11):647-656. 

7.  Rudick RA, Lee JC, Simon J, Fisher E. Significance of T2 lesions in multiple sclerosis: a 13-year longitudinal study. Ann Neurol. 2006;60(2):236-242.

8.  Alla S, Mason DF. Multiple sclerosis in New Zealand. J Clin Neurosci. 2014;21(8):1288-1291. 

9.  Simpson S Jr, Blizzard L, Otahal P, Van der Mei I, Taylor B. Latitude is significantly associated with the prevalence of multiple sclerosis: a meta-analysis. J Neurol Neurosurg Psychiatry. 2011;82(10):1132-1141. 

10.  Evans C, Beland SG, Kulaga S, et al. Incidence and prevalence of multiple sclerosis in the Americas: a systematic review. Neuroepidemiology. 2013;40(3):195-210. 

11.  Giesser BS. Diagnosis of multiple sclerosis. Neurol Clin. 2011;29(2):381-388. 

12.  Noseworthy JH, Lucchinetti C, Rodriguez M, Weinshenker BG. Multiple sclerosis. N Engl J Med. 2000;343(13):938-952. 

13.  Weinshenker BG. The natural history of multiple sclerosis. Neurol Clin. 1995;13(1):119-146. 

14. Weinshenker BG. The natural history of multiple sclerosis: update 1998. Semin Neurol. 1998;18(3):301-307. 

15. Simone IL, Carrara D, Tortorella C, Ceccarelli A, Livrea P. Early onset multiple sclerosis. Neurol Sci. 2000;21(4)(suppl 2):S861-S863.

16. Reinhardt K, Weiss S, Rosenbauer J, Gärtner J, von Kries R. Multiple sclerosis in children and adolescents: incidence and clinical picture--new insights from the nationwide German surveillance (2009-2011). Eur J Neurol. 2014;21(4):654-659.

17. Waldman A, Ghezzi A, Bar-Or A, Mikaeloff Y, Tardieu M, Banwell B. Multiple sclerosis in children: an update on clinical diagnosis, therapeutic strategies, and research. Lancet Neurol. 2014;13(9):936-948. 

18.  Renoux C, Vukusic S, Mikaeloff Y, et al; Adult Neurology Departments KIDMUS Study Group. Natural history of multiple sclerosis with childhood onset. N Engl J Med. 2007;356(25):2603-2613.

19.  D'Ambrosio A, Pontecorvo S, Colastanti T, Zamboni S, Francia A, Margutti P. Peripheral blood biomarkers in multiple sclerosis. Autoimmun Rev. 2015;14(12):1097-1110.

20.  Hauser SL, Chan JR, Oksenberg JR. Multiple sclerosis: prospects and promise. Ann Neurol. 2013;74(3):317-327.

21.  Lublin FD, Reingold SC, Cohen JA, et al. Defining the clinical course of multiple sclerosis: the 2013 revisions. Neurology. 2014;83(3):278-286. 

22.  Polman CH, Reingold SC, Banwell B, et al. Diagnostic criteria for multiple sclerosis: 2010 revisions to the McDonald criteria. Ann Neurol. 2011;69(2):292-302.

23. Hurwitz BJ. Analysis of current multiple sclerosis registries. Neurology. 2011;76(1)(suppl 1):S7-S13.

24. Boeije HR, Duijnstee MS, Grypdonck MH, Pool A. Encountering the downward phase: biographical work in people with multiple sclerosis living at home. Soc Sci Med. 2002;55(6):881-893. 

25. Sprangers MA, de Regt EB, Andries F, et al. Which chronic conditions are associated with better or poorer quality of life? J Clin Epidemiol. 2000;53(9):895-907. 

26. Julian LJ, Vella L, Vollmer T, Hadjimichael O, Mohr DC. Employment in multiple sclerosis. Exiting and re-entering the work force. J Neurol. 2008;255(9):1354-1360. 

27. Trisolini M, Honeycutt A, Wiener J, Lesesne S. Global economic impact of multiple sclerosis. Multiple Sclerosis International Federation website. http://www.msif.org/wp-content/uploads/2014/09/Global_economic_impact_of_MS.pdf. Published May 2010. Accessed May 6, 2016

28. Scalfari A, Knappertz V, Cutter G, Goodin DS, Ashton R, Ebers GC. Mortality in patients with multiple sclerosis. Neurology. 2013;81(2):184-192.

29. Gurevich M, Miron G, Achiron A. Optimizing multiple sclerosis diagnosis: gene expression and genomic association. Ann Clin Transl Neurol. 2015;2(3):271-277.

30. National Multiple Sclerosis Society, European Committee for Treatment and Research in Multiple Sclerosis. Tip sheet: 2010 revised McDonald diagnostic criteria for MS. National Multiple Sclerosis Society website. http://www.nationalmssociety.org/NationalMSSociety/media/MSNationalFiles/Brochures/Paper-TipSheet_-2010-Revisions-to-the-McDonald-Criteria-for-the-Diagnosis-of-MS.pdf. Accessed April 22, 2016. 

31. Freedman MS, Selchen D, Arnold DL, et al; Canadian Multiple Sclerosis Working Group. Treatment optimization in MS: Canadian MS Working Group updated recommendations. Can J Neurol Sci. 2013;40(3):307-323. 

32. Gold R, Wolinsky JS, Amato MP, Comi G. Evolving expectations around early management of multiple sclerosis. Ther Adv Neurol Disord. 2010;3(6):351-367. 

33. Copaxone [package insert]. Overland Park, KS: Teva Neuroscience, Inc; 2014. 

34. Avonex [package insert]. Cambridge, MA: Biogen Idec Inc; 1996. 

35. Rebif [package insert]. Rockland, MA: EMD Serono, Inc; New York, NY: Pfizer, Inc; 2012. 

36. Betaseron [package insert]. Montville, NJ: Bayer HealthCare Pharmaceuticals Inc; 2012. 

37. Extavia [package insert]. East Hanover, NJ: Novartis Pharmaceuticals Corp; 2014. 

38. Plegridy [package insert]. Cambridge, MA: Biogen Idec Inc; 2013. 

39. Calabresi PA, Kieseier BC, Arnold DL, et al. Pegylated interferon ß-1a for relapsing-remitting multiple sclerosis (ADVANCE): a randomised, phase 3, double-blind study. Lancet Neurol. 2014;13(7):657-665. 

40. Tecfidera [package insert]. Cambridge, MA: Biogen Idec Inc; 2015. 

41. Gilenya [package insert]. East Hanover, NJ: Novartis Pharmaceuticals Corp; 2016. 

42. Aubagio [package insert]. Cambridge, MA: Genzyme Corp; 2012.

43. Lemtrada [package insert]. Cambridge, MA: Genzyme Corp; 2014. 

44. Cohen JA, Coles AJ, Arnold DL, et al; CARE-MS I investigators. Alemtuzumab versus interferon beta 1a as first-line treatment for patients with relapsing-remitting multiple sclerosis: a randomised controlled phase 3 trial. Lancet. 2012;380(9856):1819-1828. 

45. Coles AJ, Twyman CL, Arnold DL, et al; CARE-MS II investigators. Alemtuzumab for patients with relapsing multiple sclerosis after disease-modifying therapy: a randomised controlled phase 3 trial. Lancet. 2012;380(9856):1829-1839. 

46. Novantrone [package insert]. Rockland, MA: EMD Serono, Inc; 2008. 

47. Tysabri [medication guide]. Cambridge, MA: Biogen Idec Inc; 2015.

48. Hartung DM, Bourdette DN, Ahmed SM, Whitham RH. The cost of multiple sclerosis drugs in the US and the pharmaceutical industry: too big to fail. Neurology. 2015;84(21):2185-2192. 

49. Cohen BA, Coyle PK, Leist T, Oleen-Burkey MA, Schwartz M, Zwibel H. Therapy Optimization in Multiple Sclerosis: a cohort study of therapy adherence and risk of relapse. Mult Scler Relat Disord. 2015;4(1):75-82.

50. Cohen B, Leist T, Coyle P, Zwibel H, Markowitz C, Tullman M. MS therapy adherence and relapse risk. Neurology. 2013;80(7) (suppl):P01.193. 

51. Richert ND, Zierak MC, Bash CN, Lewis BK, McFarland HF, Frank JA. MRI and clinical activity in MS patients after terminating treatment with interferon beta-1b. Mult Scler. 2000;6(2):86-90. 

52. Siger M, Durko A, Nicpan A, Konarska M, Grudziecka M, Selmaj K. Discontinuation of interferon beta therapy in multiple sclerosis patients with high pre-treatment disease activity leads to prompt return to previous disease activity. J Neurol Sci. 2011;303(1-2):50-52. 

53. Wu X, Dastidar P, Kuusisto H, Ukkonen M, Huhtala H, Elovaara I. Increased disability and MRI lesions after discontinuation of IFN-beta-1a in secondary progressive MS. Acta Neurol Scand. 2005;112(4):242-247. 

54. Scalfari A, Neuhaus A, Degenhardt A, et al. The natural history of multiple sclerosis: a geographically based study 10: relapses and long-term disability. Brain. 2010;133(pt 7):1914-1929. 

55. Bates D. Treatment effects of immunomodulatory therapies at different stages of multiple sclerosis in short-term trials. Neurology. 2011;76(1)(suppl 1):S14-S25. 

56. Fisniku LK, Brex PA, Altmann DR, et al. Disability and T2 MRI lesions: a 20-year follow-up of patients with relapse onset of multiple sclerosis. Brain. 2008;131(pt 3):808-817. 

57. Cross AH, Naismith RT. Established and novel disease-modifying treatments in multiple sclerosis. J Intern Med. 2014;275(4):350-363. 

58. Lublin FD, Baier M, Cutter G. Effect of relapses on development of residual deficit in multiple sclerosis. Neurology. 2003;61(11):1528-1532. 

59. Kalb R. The emotional and psychological impact of multiple sclerosis relapses. J Neurol Sci. 2007;256(suppl 1):S29-S33.

Article PDF
Author and Disclosure Information

Dr. Robertson is director of the Multiple Sclerosis Center of Excellence at James A. Haley Veterans' Hospital in Tampa, Florida. Dr. Moreo is a PGY-1 multiple sclerosis fellow, and Dr. Robertson is an assistant professor of neurology and director of the multiple sclerosis division, both at the University of South Florida in Tampa.

Author disclosures
Dr. Robertson has served as a consultant for Biogen, Genzyme, Teva Neuroscience, and Pfizer; is on the speakers’ bureaus of Biogen, Pfizer, EMD Serono, Genzyme, Novartis, Teva Neuroscience, Mallinckrodt, and Acorda; and has received grant support from Biogen, Genzyme, Novartis, Sun Pharma, MedImmune, Actelion, Mallinckrodt, EMD Serono, and Genetech.

Disclaimer
The opinions expressed herein are those of the authors and do not necessarily reflect those of Federal Practitioner, Frontline Medical Communications Inc., the U.S. Government, or any of its agencies. This article may discuss unlabeled or investigational use of certain drugs. Please review complete prescribing information for specific drugs or drug combinations—including indications, contraindications, warnings, and adverse effects—before administering pharmacologic therapy to patients.

Issue
Federal Practitioner - 33(6)
Publications
Topics
Page Number
28-33
Legacy Keywords
Derrick Robertson MD, Natalie Moreo MD, multiple sclerosis, MS
Sections
Author and Disclosure Information

Dr. Robertson is director of the Multiple Sclerosis Center of Excellence at James A. Haley Veterans' Hospital in Tampa, Florida. Dr. Moreo is a PGY-1 multiple sclerosis fellow, and Dr. Robertson is an assistant professor of neurology and director of the multiple sclerosis division, both at the University of South Florida in Tampa.

Author disclosures
Dr. Robertson has served as a consultant for Biogen, Genzyme, Teva Neuroscience, and Pfizer; is on the speakers’ bureaus of Biogen, Pfizer, EMD Serono, Genzyme, Novartis, Teva Neuroscience, Mallinckrodt, and Acorda; and has received grant support from Biogen, Genzyme, Novartis, Sun Pharma, MedImmune, Actelion, Mallinckrodt, EMD Serono, and Genetech.

Disclaimer
The opinions expressed herein are those of the authors and do not necessarily reflect those of Federal Practitioner, Frontline Medical Communications Inc., the U.S. Government, or any of its agencies. This article may discuss unlabeled or investigational use of certain drugs. Please review complete prescribing information for specific drugs or drug combinations—including indications, contraindications, warnings, and adverse effects—before administering pharmacologic therapy to patients.

Author and Disclosure Information

Dr. Robertson is director of the Multiple Sclerosis Center of Excellence at James A. Haley Veterans' Hospital in Tampa, Florida. Dr. Moreo is a PGY-1 multiple sclerosis fellow, and Dr. Robertson is an assistant professor of neurology and director of the multiple sclerosis division, both at the University of South Florida in Tampa.

Author disclosures
Dr. Robertson has served as a consultant for Biogen, Genzyme, Teva Neuroscience, and Pfizer; is on the speakers’ bureaus of Biogen, Pfizer, EMD Serono, Genzyme, Novartis, Teva Neuroscience, Mallinckrodt, and Acorda; and has received grant support from Biogen, Genzyme, Novartis, Sun Pharma, MedImmune, Actelion, Mallinckrodt, EMD Serono, and Genetech.

Disclaimer
The opinions expressed herein are those of the authors and do not necessarily reflect those of Federal Practitioner, Frontline Medical Communications Inc., the U.S. Government, or any of its agencies. This article may discuss unlabeled or investigational use of certain drugs. Please review complete prescribing information for specific drugs or drug combinations—including indications, contraindications, warnings, and adverse effects—before administering pharmacologic therapy to patients.

Article PDF
Article PDF
Related Articles
Controlling symptoms can slow the physical and emotional disabilities associated with multiple sclerosis and help patients attain the highest quality of life possible for as long as possible.
Controlling symptoms can slow the physical and emotional disabilities associated with multiple sclerosis and help patients attain the highest quality of life possible for as long as possible.

Multiple sclerosis (MS) is a disorder characterized by inflammation, demyelination, and degeneration of the central nervous system (CNS). The hallmark of the disorder is relapses and remissions of neurologic symptoms occurring early in the disease course, which are often associated with areas of CNS inflammation and myelin loss.1-3 The inciting cause for this inflammation is unknown but is believed to be multifactorial, with environmental and genetic influences creating an adaptive, T cell-mediated autoimmune response against the CNS.4 Separate from the acute attacks, progressive neurodegeneration can occur more chronically and is characterized by axonal loss and grey matter atrophy thought to be due to direct cytotoxic activity of the innate immune system as well as toxic intermediates, such as nitric oxide.4,5 Despite the multiple insults early on, neurologic disability typically becomes more apparent over time.6 The disability threshold theory argues that neurologic function compensates for brain tissue loss until a threshold of accumulated damage is exceeded.7

Background

The incidence of MS follows a geographic gradient; rates rise as the distance from the equator increases.8,9 This is thought to be due to the gradient of relative sun exposure and its role in the production of vitamin D, which plays an important role in immune regulation when converted to its active hormonal form. Multiple sclerosis is more prevalent in non-Hispanic white patients than it is in other racial groups, and women are affected nearly 2 to 3 times more often than are men.10 About 450,000 individuals in the U.S. and more than 2 million worldwide have MS.11-14

Multiple sclerosis is the most common cause of nontraumatic neurologic disability in young adults. It is typically diagnosed in the third and fourth decades of life, and those who are diagnosed after age 50 years often can recount neurologic symptoms that began years before. However, pediatric-onset and new-onset cases in the elderly have been reported. It has been estimated that up to 10% of patients with MS have onset before 18 years of age.15-17 Compared with adult-onset MS, pediatric-onset is associated with a longer period between initial attack and physical disability, although the average age of disability onset is about 10 years younger.17,18

 

Disease Courses

Relapsing-remitting MS (RRMS) is the most common disease course overall, and this pattern affects 97% of individuals with disease onset before age 18 years.15-17 The clinically isolated syndrome disease course leads to clinically definite MS in one-third of patients within 1 year and in one-half of patients within 2 years.19 In the majority of cases, the RRMS course transitions over time to secondary-progressive MS (SPMS), which is a disease pattern of progressively worsening disability with few neurologic relapses. Although inflammation is present at all stages, the difference is in the predominance of cell types involved.5 Why the shift from active to chronic inflammation occurs and how to prevent it remain central questions in MS research.4 Regardless, tentative evidence suggests that prevention of relapses may reduce disability accumulation and risk of conversion to progressive MS.20

A minority of patients with MS are diagnosed with primary-progressive MS (PPMS) at onset, which is characterized by a disease pattern that follows a relatively steady progression of neurologic symptoms over time, without clear relapses or remissions of these symptoms, though phases of stability or fluctuations in disability may still occur.21 It is typically diagnosed at an older age than is RRMS, and it is rare in children; suspicion of PPMS in this age group should prompt detailed assessment of alternative diagnoses.17,22 Primary-progressive MS is more equally distributed in men and women than is RRMS.

Regardless of onset type, disability progression seems to occur at the same rate among all patients with MS after a certain threshold is reached. The established assessment scale for disability progression in MS is the Kurtzke Expanded Disability Status scale (EDSS), which has a scoring range from 0 to 10. Data from several patient registries have shown that once EDSS step 4 is reached, progression thereafter occurs at a predictable rate that is similar across MS phenotypes.23 The time it takes patients to subsequently reach higher EDSS steps may be independent of preceding factors.23

MS Symptom Burden

The neurologic symptoms that patients experience are fluctuating and disabling throughout the disease course, irrespective of onset type. Typical MS symptoms include mobility impairment, changes in cognition and mood, pain and other sensation disturbances, bowel and bladder dysfunction, fatigue, and visual disturbances. The burden of these symptoms can significantly impact quality of life (QOL) for patients and their families. The symptom burden can pose a direct threat to a patient’s autonomy, necessitating adaptation to an unpredictable disease that requires frequent health care visits to many different health care providers (eg, neurologists; primary care providers; physiatrists; urologists; ophthalmologists; and speech, physical, and occupational therapists), periodic testing, and often costly medications.24

 

 

Compared with patients who have other chronic conditions, patients with MS experience diminished societal roles, along with decreased assessments in health, energy, and physical functions.25 These often lead to early exit from the workforce and limitations in household responsibilities, which further impact QOL.26 Including both direct and indirect costs of the disease, a patient with MS can expect a lifetime financial burden of nearly $1.2 million.27

Large population cohort studies in MS, along with MS registry studies of patients untreated with disease-modifying therapies, have shown reduced survival rates by an average of 7 to 14 years.23,28 Multiple sclerosis is the main cause of death in about 50% of cases (EDSS step 10), which is defined as “acute death due to brain stem involvement or to respiratory failure, or death consequent to the chronic bedridden state with terminal pneumonia, sepsis, uremia, or cardiorespiratory failure [and excluding] intercurrent causes of death.”23 For the remaining patients with MS, cause of death is similar to those of the general population, such as cardiovascular disease and cancer.23 However, the incidence of suicide is higher among patients with MS.23

All these factors underscore the importance of early diagnosis as well as early initiation of effective disease-modifying therapy.

The diagnosis of MS is difficult largely due to the lack of definitive diagnostic testing and specific biomarkers for disease activity and because of the wide range of differential diagnoses that can mimic MS.19,21,29 Diagnosis of MS requires that more likely diagnoses have been excluded as well as that lesions (scleroses) are disseminated in space within the CNS and disseminated in time. The 2010 Revised McDonald Diagnostic Criteria for MS are outlined in Table 1.

Disease-Modifying Therapies

The goal of MS disease-modifying therapy is to reduce the early clinical and subclinical disease activity that eventually contributes to long-term disability.31,32 There are currently 13 FDA-approved disease-modifying therapies for MS. These include 7 self-injecting therapies, 3 oral therapies, and 3 infusion therapies. These 13 medications have 8 different mechanisms of action (MOA) that target distinct areas of the immune-mediated disease process. They also differ in their frequencies and routes of administration in addition to their adverse effect (AE) profiles (Tables 2, 3, and 4).

 
 

Treatment Considerations

In 1993, interferon beta-1b became the first FDA-approved MS medication. In the following 2 decades, there became 12 additional FDA-approved medications for MS, beginning with other injectables. The first infusion therapy was introduced in 2004, followed by various oral medications. The treatment landscape continues to change rapidly. This therapeutic revolution has occurred largely due to the improved understanding of the pathophysiology of MS and unquestionably has improved the prognosis and overall QOL for patients. The question is no longer how to treat MS but rather how to personalize and optimize treatment for each patient.20

Despite all available treatment options, none are curative, and none have been proven to offer neuroprotection or contribute to neural repair. To date, no studies have led to FDA-approved therapies for PPMS. Further, the efficacy of any of these medications varies from patient to patient. Due largely to the lack of biomarkers for disease activity and treatment response, drug efficacy continues to be measured according to the current gold standard, which is identification of gadolinium-enhancing lesions in the white matter on magnetic resonance imaging (MRI), combined with other markers of disease, including clinical relapse rate and confirmed disability progression.19 In general, the injectable therapies are expected to protect against about 20% to 35% of relapses; the oral agents, 50% to 55%; and the infusion therapies, > 60%.2

In conjunction with a medication’s efficacy rate and safety profile, the frequency and route of administration also must be considered. In general, MS medications are exceedingly expensive, some costing up to tens-of-thousands of dollars per year.48 All these factors have the real potential to negatively impact patient adherence. Nonadherence and gaps in treatment have been correlated with increased rates of relapses and progression of disability as well as negative MRI outcomes.49-53

When to Initiate Treatment

Once a patient is diagnosed, a common question is, when is the right time to initiate treatment? The primary target of the current MS medications is to decrease CNS inflammation (relapses). The ideal time to start treatment is as promptly as possible after confirmation of the diagnosis to combat the early inflammatory relapsing phase of the disease. There seems to be an early window in the disease course when achieving disease control can have a profound impact on long-term disability. Disease control is typically defined as decreasing relapses, slowing the accumulation of lesions visualized on MRI, and preventing the disability that occurs from both incomplete recovery after relapses and overall disease progression.54,55

 

 

Certain clinical indicators, such as higher relapse rates early in the disease course and MRI characteristics, including total lesion burden and the location of lesions within the CNS, seem to be associated with a higher risk of disease progression.56 These are potential prognostic indicators that can help tailor the choice of disease-modifying therapy for patients.57 Those with highly inflammatory and potentially aggressive disease at onset, for example, may benefit from early initiation of higher efficacy therapies, whereas those with more benign forms of MS at onset may fare well on lower efficacy therapies. In general, when it comes to currently available MS treatments, higher efficacy is often tied to riskier AE profiles, so the best medication may be the “least efficacious” one that can still control the disease.20

Hauser and colleagues suggested a treatment decision-making model that identifies the interferons, glatiramer acetate, dimethyl fumarate, and teriflunomide as acceptable first-line therapies; fingolimod and natalizumab as acceptable second-line options; and mitoxantrone and alemtuzumab as acceptable third-line therapeutic options.20 The authors generally agree with Hauser and colleagues’ model, and it is important to consider individual patient factors (eg, comorbidities, concurrent medications, life circumstances) and disease severity when deciding on a treatment plan.

Perhaps an even more difficult question is, when is the right time to switch therapies? There remains a dearth of either guidelines or comparative studies for treatment management decisions. Further, without reliable biomarkers, the clinical and pathologic heterogeneity of MS makes treatment difficult.4,19 In practice, there is general consensus that 1 year of treatment monitoring for effects on clinical and radiologic outcomes is an acceptable time frame to evaluate effectiveness of a disease-modifying treatment. If adherence is maintained and there is still evidence of clinical or MRI activity (suggesting a suboptimal response), an alternative therapy, particularly one with a different MOA, should be strongly considered. This highlights the importance of broad access to all available MS therapies to allow for early selection of a correct therapy that patients will remain adherent to and that controls their disease.

 

Conclusion

Multiple sclerosis remains a highly unpredictable disease, and relapses have the ability to produce a measurable and sustained impact on the level of disability.58 Still, the influence of reduced relapses on preventing disability in an individual patient remains unclear. Large, long-term, prospective cohort studies may clarify whether early treatment affects disease progression and disability.20 However, it is quite evident that effective relapse reduction decreases discomfort, reduces days lost from work and other important activities of daily life, and improves QOL.58,59

There is still much to learn about this unique disease, but emerging evidence in the medical literature highlights the importance of setting treatment goals that include targeting disease activity to achieve early and effective control. Attaining control with a MS medication seems to be a key component of slowing the physical and emotional disability that can accumulate, helping patients remain active and maintain the highest QOL possible for as long as possible.

Multiple sclerosis (MS) is a disorder characterized by inflammation, demyelination, and degeneration of the central nervous system (CNS). The hallmark of the disorder is relapses and remissions of neurologic symptoms occurring early in the disease course, which are often associated with areas of CNS inflammation and myelin loss.1-3 The inciting cause for this inflammation is unknown but is believed to be multifactorial, with environmental and genetic influences creating an adaptive, T cell-mediated autoimmune response against the CNS.4 Separate from the acute attacks, progressive neurodegeneration can occur more chronically and is characterized by axonal loss and grey matter atrophy thought to be due to direct cytotoxic activity of the innate immune system as well as toxic intermediates, such as nitric oxide.4,5 Despite the multiple insults early on, neurologic disability typically becomes more apparent over time.6 The disability threshold theory argues that neurologic function compensates for brain tissue loss until a threshold of accumulated damage is exceeded.7

Background

The incidence of MS follows a geographic gradient; rates rise as the distance from the equator increases.8,9 This is thought to be due to the gradient of relative sun exposure and its role in the production of vitamin D, which plays an important role in immune regulation when converted to its active hormonal form. Multiple sclerosis is more prevalent in non-Hispanic white patients than it is in other racial groups, and women are affected nearly 2 to 3 times more often than are men.10 About 450,000 individuals in the U.S. and more than 2 million worldwide have MS.11-14

Multiple sclerosis is the most common cause of nontraumatic neurologic disability in young adults. It is typically diagnosed in the third and fourth decades of life, and those who are diagnosed after age 50 years often can recount neurologic symptoms that began years before. However, pediatric-onset and new-onset cases in the elderly have been reported. It has been estimated that up to 10% of patients with MS have onset before 18 years of age.15-17 Compared with adult-onset MS, pediatric-onset is associated with a longer period between initial attack and physical disability, although the average age of disability onset is about 10 years younger.17,18

 

Disease Courses

Relapsing-remitting MS (RRMS) is the most common disease course overall, and this pattern affects 97% of individuals with disease onset before age 18 years.15-17 The clinically isolated syndrome disease course leads to clinically definite MS in one-third of patients within 1 year and in one-half of patients within 2 years.19 In the majority of cases, the RRMS course transitions over time to secondary-progressive MS (SPMS), which is a disease pattern of progressively worsening disability with few neurologic relapses. Although inflammation is present at all stages, the difference is in the predominance of cell types involved.5 Why the shift from active to chronic inflammation occurs and how to prevent it remain central questions in MS research.4 Regardless, tentative evidence suggests that prevention of relapses may reduce disability accumulation and risk of conversion to progressive MS.20

A minority of patients with MS are diagnosed with primary-progressive MS (PPMS) at onset, which is characterized by a disease pattern that follows a relatively steady progression of neurologic symptoms over time, without clear relapses or remissions of these symptoms, though phases of stability or fluctuations in disability may still occur.21 It is typically diagnosed at an older age than is RRMS, and it is rare in children; suspicion of PPMS in this age group should prompt detailed assessment of alternative diagnoses.17,22 Primary-progressive MS is more equally distributed in men and women than is RRMS.

Regardless of onset type, disability progression seems to occur at the same rate among all patients with MS after a certain threshold is reached. The established assessment scale for disability progression in MS is the Kurtzke Expanded Disability Status scale (EDSS), which has a scoring range from 0 to 10. Data from several patient registries have shown that once EDSS step 4 is reached, progression thereafter occurs at a predictable rate that is similar across MS phenotypes.23 The time it takes patients to subsequently reach higher EDSS steps may be independent of preceding factors.23

MS Symptom Burden

The neurologic symptoms that patients experience are fluctuating and disabling throughout the disease course, irrespective of onset type. Typical MS symptoms include mobility impairment, changes in cognition and mood, pain and other sensation disturbances, bowel and bladder dysfunction, fatigue, and visual disturbances. The burden of these symptoms can significantly impact quality of life (QOL) for patients and their families. The symptom burden can pose a direct threat to a patient’s autonomy, necessitating adaptation to an unpredictable disease that requires frequent health care visits to many different health care providers (eg, neurologists; primary care providers; physiatrists; urologists; ophthalmologists; and speech, physical, and occupational therapists), periodic testing, and often costly medications.24

 

 

Compared with patients who have other chronic conditions, patients with MS experience diminished societal roles, along with decreased assessments in health, energy, and physical functions.25 These often lead to early exit from the workforce and limitations in household responsibilities, which further impact QOL.26 Including both direct and indirect costs of the disease, a patient with MS can expect a lifetime financial burden of nearly $1.2 million.27

Large population cohort studies in MS, along with MS registry studies of patients untreated with disease-modifying therapies, have shown reduced survival rates by an average of 7 to 14 years.23,28 Multiple sclerosis is the main cause of death in about 50% of cases (EDSS step 10), which is defined as “acute death due to brain stem involvement or to respiratory failure, or death consequent to the chronic bedridden state with terminal pneumonia, sepsis, uremia, or cardiorespiratory failure [and excluding] intercurrent causes of death.”23 For the remaining patients with MS, cause of death is similar to those of the general population, such as cardiovascular disease and cancer.23 However, the incidence of suicide is higher among patients with MS.23

All these factors underscore the importance of early diagnosis as well as early initiation of effective disease-modifying therapy.

The diagnosis of MS is difficult largely due to the lack of definitive diagnostic testing and specific biomarkers for disease activity and because of the wide range of differential diagnoses that can mimic MS.19,21,29 Diagnosis of MS requires that more likely diagnoses have been excluded as well as that lesions (scleroses) are disseminated in space within the CNS and disseminated in time. The 2010 Revised McDonald Diagnostic Criteria for MS are outlined in Table 1.

Disease-Modifying Therapies

The goal of MS disease-modifying therapy is to reduce the early clinical and subclinical disease activity that eventually contributes to long-term disability.31,32 There are currently 13 FDA-approved disease-modifying therapies for MS. These include 7 self-injecting therapies, 3 oral therapies, and 3 infusion therapies. These 13 medications have 8 different mechanisms of action (MOA) that target distinct areas of the immune-mediated disease process. They also differ in their frequencies and routes of administration in addition to their adverse effect (AE) profiles (Tables 2, 3, and 4).

 
 

Treatment Considerations

In 1993, interferon beta-1b became the first FDA-approved MS medication. In the following 2 decades, there became 12 additional FDA-approved medications for MS, beginning with other injectables. The first infusion therapy was introduced in 2004, followed by various oral medications. The treatment landscape continues to change rapidly. This therapeutic revolution has occurred largely due to the improved understanding of the pathophysiology of MS and unquestionably has improved the prognosis and overall QOL for patients. The question is no longer how to treat MS but rather how to personalize and optimize treatment for each patient.20

Despite all available treatment options, none are curative, and none have been proven to offer neuroprotection or contribute to neural repair. To date, no studies have led to FDA-approved therapies for PPMS. Further, the efficacy of any of these medications varies from patient to patient. Due largely to the lack of biomarkers for disease activity and treatment response, drug efficacy continues to be measured according to the current gold standard, which is identification of gadolinium-enhancing lesions in the white matter on magnetic resonance imaging (MRI), combined with other markers of disease, including clinical relapse rate and confirmed disability progression.19 In general, the injectable therapies are expected to protect against about 20% to 35% of relapses; the oral agents, 50% to 55%; and the infusion therapies, > 60%.2

In conjunction with a medication’s efficacy rate and safety profile, the frequency and route of administration also must be considered. In general, MS medications are exceedingly expensive, some costing up to tens-of-thousands of dollars per year.48 All these factors have the real potential to negatively impact patient adherence. Nonadherence and gaps in treatment have been correlated with increased rates of relapses and progression of disability as well as negative MRI outcomes.49-53

When to Initiate Treatment

Once a patient is diagnosed, a common question is, when is the right time to initiate treatment? The primary target of the current MS medications is to decrease CNS inflammation (relapses). The ideal time to start treatment is as promptly as possible after confirmation of the diagnosis to combat the early inflammatory relapsing phase of the disease. There seems to be an early window in the disease course when achieving disease control can have a profound impact on long-term disability. Disease control is typically defined as decreasing relapses, slowing the accumulation of lesions visualized on MRI, and preventing the disability that occurs from both incomplete recovery after relapses and overall disease progression.54,55

 

 

Certain clinical indicators, such as higher relapse rates early in the disease course and MRI characteristics, including total lesion burden and the location of lesions within the CNS, seem to be associated with a higher risk of disease progression.56 These are potential prognostic indicators that can help tailor the choice of disease-modifying therapy for patients.57 Those with highly inflammatory and potentially aggressive disease at onset, for example, may benefit from early initiation of higher efficacy therapies, whereas those with more benign forms of MS at onset may fare well on lower efficacy therapies. In general, when it comes to currently available MS treatments, higher efficacy is often tied to riskier AE profiles, so the best medication may be the “least efficacious” one that can still control the disease.20

Hauser and colleagues suggested a treatment decision-making model that identifies the interferons, glatiramer acetate, dimethyl fumarate, and teriflunomide as acceptable first-line therapies; fingolimod and natalizumab as acceptable second-line options; and mitoxantrone and alemtuzumab as acceptable third-line therapeutic options.20 The authors generally agree with Hauser and colleagues’ model, and it is important to consider individual patient factors (eg, comorbidities, concurrent medications, life circumstances) and disease severity when deciding on a treatment plan.

Perhaps an even more difficult question is, when is the right time to switch therapies? There remains a dearth of either guidelines or comparative studies for treatment management decisions. Further, without reliable biomarkers, the clinical and pathologic heterogeneity of MS makes treatment difficult.4,19 In practice, there is general consensus that 1 year of treatment monitoring for effects on clinical and radiologic outcomes is an acceptable time frame to evaluate effectiveness of a disease-modifying treatment. If adherence is maintained and there is still evidence of clinical or MRI activity (suggesting a suboptimal response), an alternative therapy, particularly one with a different MOA, should be strongly considered. This highlights the importance of broad access to all available MS therapies to allow for early selection of a correct therapy that patients will remain adherent to and that controls their disease.

 

Conclusion

Multiple sclerosis remains a highly unpredictable disease, and relapses have the ability to produce a measurable and sustained impact on the level of disability.58 Still, the influence of reduced relapses on preventing disability in an individual patient remains unclear. Large, long-term, prospective cohort studies may clarify whether early treatment affects disease progression and disability.20 However, it is quite evident that effective relapse reduction decreases discomfort, reduces days lost from work and other important activities of daily life, and improves QOL.58,59

There is still much to learn about this unique disease, but emerging evidence in the medical literature highlights the importance of setting treatment goals that include targeting disease activity to achieve early and effective control. Attaining control with a MS medication seems to be a key component of slowing the physical and emotional disability that can accumulate, helping patients remain active and maintain the highest QOL possible for as long as possible.

References

1.  Lublin FD, Reingold SC. Defining the clinical course of multiple sclerosis: results of an international survey. National Multiple Sclerosis Society (USA) Advisory Committee on Clinical Trials of New Agents in Multiple Sclerosis. Neurology. 1996;46(4):907-911. 

2.  Frischer JM, Bramow S, Dal-Bianco A, et al. The relation between inflammation and neurodegeneration in multiple sclerosis brains. Brain. 2009;132(pt 5):1175-1189. 

3.  Charil A, Filippi M. Inflammatory demyelination and neurodegeneration in early multiple sclerosis. J Neurol Sci. 2007;259(1-2):7-15.

4.  Weiner HL. The challenge of multiple sclerosis: how do we cure a chronic heterogeneous disease? Ann Neurol. 2009;65(3):239-248.

5.  Grigoriadis N, van Pesch V; ParadigMS Group. A basic overview of multiple sclerosis immunopathology. Eur J Neurol. 2015;22(suppl 2):3-13. 

6.  Lassmann H, van Horssen J, Mahad D. Progressive multiple sclerosis: pathology and pathogenesis. Nat Rev Neurol. 2012;8(11):647-656. 

7.  Rudick RA, Lee JC, Simon J, Fisher E. Significance of T2 lesions in multiple sclerosis: a 13-year longitudinal study. Ann Neurol. 2006;60(2):236-242.

8.  Alla S, Mason DF. Multiple sclerosis in New Zealand. J Clin Neurosci. 2014;21(8):1288-1291. 

9.  Simpson S Jr, Blizzard L, Otahal P, Van der Mei I, Taylor B. Latitude is significantly associated with the prevalence of multiple sclerosis: a meta-analysis. J Neurol Neurosurg Psychiatry. 2011;82(10):1132-1141. 

10.  Evans C, Beland SG, Kulaga S, et al. Incidence and prevalence of multiple sclerosis in the Americas: a systematic review. Neuroepidemiology. 2013;40(3):195-210. 

11.  Giesser BS. Diagnosis of multiple sclerosis. Neurol Clin. 2011;29(2):381-388. 

12.  Noseworthy JH, Lucchinetti C, Rodriguez M, Weinshenker BG. Multiple sclerosis. N Engl J Med. 2000;343(13):938-952. 

13.  Weinshenker BG. The natural history of multiple sclerosis. Neurol Clin. 1995;13(1):119-146. 

14. Weinshenker BG. The natural history of multiple sclerosis: update 1998. Semin Neurol. 1998;18(3):301-307. 

15. Simone IL, Carrara D, Tortorella C, Ceccarelli A, Livrea P. Early onset multiple sclerosis. Neurol Sci. 2000;21(4)(suppl 2):S861-S863.

16. Reinhardt K, Weiss S, Rosenbauer J, Gärtner J, von Kries R. Multiple sclerosis in children and adolescents: incidence and clinical picture--new insights from the nationwide German surveillance (2009-2011). Eur J Neurol. 2014;21(4):654-659.

17. Waldman A, Ghezzi A, Bar-Or A, Mikaeloff Y, Tardieu M, Banwell B. Multiple sclerosis in children: an update on clinical diagnosis, therapeutic strategies, and research. Lancet Neurol. 2014;13(9):936-948. 

18.  Renoux C, Vukusic S, Mikaeloff Y, et al; Adult Neurology Departments KIDMUS Study Group. Natural history of multiple sclerosis with childhood onset. N Engl J Med. 2007;356(25):2603-2613.

19.  D'Ambrosio A, Pontecorvo S, Colastanti T, Zamboni S, Francia A, Margutti P. Peripheral blood biomarkers in multiple sclerosis. Autoimmun Rev. 2015;14(12):1097-1110.

20.  Hauser SL, Chan JR, Oksenberg JR. Multiple sclerosis: prospects and promise. Ann Neurol. 2013;74(3):317-327.

21.  Lublin FD, Reingold SC, Cohen JA, et al. Defining the clinical course of multiple sclerosis: the 2013 revisions. Neurology. 2014;83(3):278-286. 

22.  Polman CH, Reingold SC, Banwell B, et al. Diagnostic criteria for multiple sclerosis: 2010 revisions to the McDonald criteria. Ann Neurol. 2011;69(2):292-302.

23. Hurwitz BJ. Analysis of current multiple sclerosis registries. Neurology. 2011;76(1)(suppl 1):S7-S13.

24. Boeije HR, Duijnstee MS, Grypdonck MH, Pool A. Encountering the downward phase: biographical work in people with multiple sclerosis living at home. Soc Sci Med. 2002;55(6):881-893. 

25. Sprangers MA, de Regt EB, Andries F, et al. Which chronic conditions are associated with better or poorer quality of life? J Clin Epidemiol. 2000;53(9):895-907. 

26. Julian LJ, Vella L, Vollmer T, Hadjimichael O, Mohr DC. Employment in multiple sclerosis. Exiting and re-entering the work force. J Neurol. 2008;255(9):1354-1360. 

27. Trisolini M, Honeycutt A, Wiener J, Lesesne S. Global economic impact of multiple sclerosis. Multiple Sclerosis International Federation website. http://www.msif.org/wp-content/uploads/2014/09/Global_economic_impact_of_MS.pdf. Published May 2010. Accessed May 6, 2016

28. Scalfari A, Knappertz V, Cutter G, Goodin DS, Ashton R, Ebers GC. Mortality in patients with multiple sclerosis. Neurology. 2013;81(2):184-192.

29. Gurevich M, Miron G, Achiron A. Optimizing multiple sclerosis diagnosis: gene expression and genomic association. Ann Clin Transl Neurol. 2015;2(3):271-277.

30. National Multiple Sclerosis Society, European Committee for Treatment and Research in Multiple Sclerosis. Tip sheet: 2010 revised McDonald diagnostic criteria for MS. National Multiple Sclerosis Society website. http://www.nationalmssociety.org/NationalMSSociety/media/MSNationalFiles/Brochures/Paper-TipSheet_-2010-Revisions-to-the-McDonald-Criteria-for-the-Diagnosis-of-MS.pdf. Accessed April 22, 2016. 

31. Freedman MS, Selchen D, Arnold DL, et al; Canadian Multiple Sclerosis Working Group. Treatment optimization in MS: Canadian MS Working Group updated recommendations. Can J Neurol Sci. 2013;40(3):307-323. 

32. Gold R, Wolinsky JS, Amato MP, Comi G. Evolving expectations around early management of multiple sclerosis. Ther Adv Neurol Disord. 2010;3(6):351-367. 

33. Copaxone [package insert]. Overland Park, KS: Teva Neuroscience, Inc; 2014. 

34. Avonex [package insert]. Cambridge, MA: Biogen Idec Inc; 1996. 

35. Rebif [package insert]. Rockland, MA: EMD Serono, Inc; New York, NY: Pfizer, Inc; 2012. 

36. Betaseron [package insert]. Montville, NJ: Bayer HealthCare Pharmaceuticals Inc; 2012. 

37. Extavia [package insert]. East Hanover, NJ: Novartis Pharmaceuticals Corp; 2014. 

38. Plegridy [package insert]. Cambridge, MA: Biogen Idec Inc; 2013. 

39. Calabresi PA, Kieseier BC, Arnold DL, et al. Pegylated interferon ß-1a for relapsing-remitting multiple sclerosis (ADVANCE): a randomised, phase 3, double-blind study. Lancet Neurol. 2014;13(7):657-665. 

40. Tecfidera [package insert]. Cambridge, MA: Biogen Idec Inc; 2015. 

41. Gilenya [package insert]. East Hanover, NJ: Novartis Pharmaceuticals Corp; 2016. 

42. Aubagio [package insert]. Cambridge, MA: Genzyme Corp; 2012.

43. Lemtrada [package insert]. Cambridge, MA: Genzyme Corp; 2014. 

44. Cohen JA, Coles AJ, Arnold DL, et al; CARE-MS I investigators. Alemtuzumab versus interferon beta 1a as first-line treatment for patients with relapsing-remitting multiple sclerosis: a randomised controlled phase 3 trial. Lancet. 2012;380(9856):1819-1828. 

45. Coles AJ, Twyman CL, Arnold DL, et al; CARE-MS II investigators. Alemtuzumab for patients with relapsing multiple sclerosis after disease-modifying therapy: a randomised controlled phase 3 trial. Lancet. 2012;380(9856):1829-1839. 

46. Novantrone [package insert]. Rockland, MA: EMD Serono, Inc; 2008. 

47. Tysabri [medication guide]. Cambridge, MA: Biogen Idec Inc; 2015.

48. Hartung DM, Bourdette DN, Ahmed SM, Whitham RH. The cost of multiple sclerosis drugs in the US and the pharmaceutical industry: too big to fail. Neurology. 2015;84(21):2185-2192. 

49. Cohen BA, Coyle PK, Leist T, Oleen-Burkey MA, Schwartz M, Zwibel H. Therapy Optimization in Multiple Sclerosis: a cohort study of therapy adherence and risk of relapse. Mult Scler Relat Disord. 2015;4(1):75-82.

50. Cohen B, Leist T, Coyle P, Zwibel H, Markowitz C, Tullman M. MS therapy adherence and relapse risk. Neurology. 2013;80(7) (suppl):P01.193. 

51. Richert ND, Zierak MC, Bash CN, Lewis BK, McFarland HF, Frank JA. MRI and clinical activity in MS patients after terminating treatment with interferon beta-1b. Mult Scler. 2000;6(2):86-90. 

52. Siger M, Durko A, Nicpan A, Konarska M, Grudziecka M, Selmaj K. Discontinuation of interferon beta therapy in multiple sclerosis patients with high pre-treatment disease activity leads to prompt return to previous disease activity. J Neurol Sci. 2011;303(1-2):50-52. 

53. Wu X, Dastidar P, Kuusisto H, Ukkonen M, Huhtala H, Elovaara I. Increased disability and MRI lesions after discontinuation of IFN-beta-1a in secondary progressive MS. Acta Neurol Scand. 2005;112(4):242-247. 

54. Scalfari A, Neuhaus A, Degenhardt A, et al. The natural history of multiple sclerosis: a geographically based study 10: relapses and long-term disability. Brain. 2010;133(pt 7):1914-1929. 

55. Bates D. Treatment effects of immunomodulatory therapies at different stages of multiple sclerosis in short-term trials. Neurology. 2011;76(1)(suppl 1):S14-S25. 

56. Fisniku LK, Brex PA, Altmann DR, et al. Disability and T2 MRI lesions: a 20-year follow-up of patients with relapse onset of multiple sclerosis. Brain. 2008;131(pt 3):808-817. 

57. Cross AH, Naismith RT. Established and novel disease-modifying treatments in multiple sclerosis. J Intern Med. 2014;275(4):350-363. 

58. Lublin FD, Baier M, Cutter G. Effect of relapses on development of residual deficit in multiple sclerosis. Neurology. 2003;61(11):1528-1532. 

59. Kalb R. The emotional and psychological impact of multiple sclerosis relapses. J Neurol Sci. 2007;256(suppl 1):S29-S33.

References

1.  Lublin FD, Reingold SC. Defining the clinical course of multiple sclerosis: results of an international survey. National Multiple Sclerosis Society (USA) Advisory Committee on Clinical Trials of New Agents in Multiple Sclerosis. Neurology. 1996;46(4):907-911. 

2.  Frischer JM, Bramow S, Dal-Bianco A, et al. The relation between inflammation and neurodegeneration in multiple sclerosis brains. Brain. 2009;132(pt 5):1175-1189. 

3.  Charil A, Filippi M. Inflammatory demyelination and neurodegeneration in early multiple sclerosis. J Neurol Sci. 2007;259(1-2):7-15.

4.  Weiner HL. The challenge of multiple sclerosis: how do we cure a chronic heterogeneous disease? Ann Neurol. 2009;65(3):239-248.

5.  Grigoriadis N, van Pesch V; ParadigMS Group. A basic overview of multiple sclerosis immunopathology. Eur J Neurol. 2015;22(suppl 2):3-13. 

6.  Lassmann H, van Horssen J, Mahad D. Progressive multiple sclerosis: pathology and pathogenesis. Nat Rev Neurol. 2012;8(11):647-656. 

7.  Rudick RA, Lee JC, Simon J, Fisher E. Significance of T2 lesions in multiple sclerosis: a 13-year longitudinal study. Ann Neurol. 2006;60(2):236-242.

8.  Alla S, Mason DF. Multiple sclerosis in New Zealand. J Clin Neurosci. 2014;21(8):1288-1291. 

9.  Simpson S Jr, Blizzard L, Otahal P, Van der Mei I, Taylor B. Latitude is significantly associated with the prevalence of multiple sclerosis: a meta-analysis. J Neurol Neurosurg Psychiatry. 2011;82(10):1132-1141. 

10.  Evans C, Beland SG, Kulaga S, et al. Incidence and prevalence of multiple sclerosis in the Americas: a systematic review. Neuroepidemiology. 2013;40(3):195-210. 

11.  Giesser BS. Diagnosis of multiple sclerosis. Neurol Clin. 2011;29(2):381-388. 

12.  Noseworthy JH, Lucchinetti C, Rodriguez M, Weinshenker BG. Multiple sclerosis. N Engl J Med. 2000;343(13):938-952. 

13.  Weinshenker BG. The natural history of multiple sclerosis. Neurol Clin. 1995;13(1):119-146. 

14. Weinshenker BG. The natural history of multiple sclerosis: update 1998. Semin Neurol. 1998;18(3):301-307. 

15. Simone IL, Carrara D, Tortorella C, Ceccarelli A, Livrea P. Early onset multiple sclerosis. Neurol Sci. 2000;21(4)(suppl 2):S861-S863.

16. Reinhardt K, Weiss S, Rosenbauer J, Gärtner J, von Kries R. Multiple sclerosis in children and adolescents: incidence and clinical picture--new insights from the nationwide German surveillance (2009-2011). Eur J Neurol. 2014;21(4):654-659.

17. Waldman A, Ghezzi A, Bar-Or A, Mikaeloff Y, Tardieu M, Banwell B. Multiple sclerosis in children: an update on clinical diagnosis, therapeutic strategies, and research. Lancet Neurol. 2014;13(9):936-948. 

18.  Renoux C, Vukusic S, Mikaeloff Y, et al; Adult Neurology Departments KIDMUS Study Group. Natural history of multiple sclerosis with childhood onset. N Engl J Med. 2007;356(25):2603-2613.

19.  D'Ambrosio A, Pontecorvo S, Colastanti T, Zamboni S, Francia A, Margutti P. Peripheral blood biomarkers in multiple sclerosis. Autoimmun Rev. 2015;14(12):1097-1110.

20.  Hauser SL, Chan JR, Oksenberg JR. Multiple sclerosis: prospects and promise. Ann Neurol. 2013;74(3):317-327.

21.  Lublin FD, Reingold SC, Cohen JA, et al. Defining the clinical course of multiple sclerosis: the 2013 revisions. Neurology. 2014;83(3):278-286. 

22.  Polman CH, Reingold SC, Banwell B, et al. Diagnostic criteria for multiple sclerosis: 2010 revisions to the McDonald criteria. Ann Neurol. 2011;69(2):292-302.

23. Hurwitz BJ. Analysis of current multiple sclerosis registries. Neurology. 2011;76(1)(suppl 1):S7-S13.

24. Boeije HR, Duijnstee MS, Grypdonck MH, Pool A. Encountering the downward phase: biographical work in people with multiple sclerosis living at home. Soc Sci Med. 2002;55(6):881-893. 

25. Sprangers MA, de Regt EB, Andries F, et al. Which chronic conditions are associated with better or poorer quality of life? J Clin Epidemiol. 2000;53(9):895-907. 

26. Julian LJ, Vella L, Vollmer T, Hadjimichael O, Mohr DC. Employment in multiple sclerosis. Exiting and re-entering the work force. J Neurol. 2008;255(9):1354-1360. 

27. Trisolini M, Honeycutt A, Wiener J, Lesesne S. Global economic impact of multiple sclerosis. Multiple Sclerosis International Federation website. http://www.msif.org/wp-content/uploads/2014/09/Global_economic_impact_of_MS.pdf. Published May 2010. Accessed May 6, 2016

28. Scalfari A, Knappertz V, Cutter G, Goodin DS, Ashton R, Ebers GC. Mortality in patients with multiple sclerosis. Neurology. 2013;81(2):184-192.

29. Gurevich M, Miron G, Achiron A. Optimizing multiple sclerosis diagnosis: gene expression and genomic association. Ann Clin Transl Neurol. 2015;2(3):271-277.

30. National Multiple Sclerosis Society, European Committee for Treatment and Research in Multiple Sclerosis. Tip sheet: 2010 revised McDonald diagnostic criteria for MS. National Multiple Sclerosis Society website. http://www.nationalmssociety.org/NationalMSSociety/media/MSNationalFiles/Brochures/Paper-TipSheet_-2010-Revisions-to-the-McDonald-Criteria-for-the-Diagnosis-of-MS.pdf. Accessed April 22, 2016. 

31. Freedman MS, Selchen D, Arnold DL, et al; Canadian Multiple Sclerosis Working Group. Treatment optimization in MS: Canadian MS Working Group updated recommendations. Can J Neurol Sci. 2013;40(3):307-323. 

32. Gold R, Wolinsky JS, Amato MP, Comi G. Evolving expectations around early management of multiple sclerosis. Ther Adv Neurol Disord. 2010;3(6):351-367. 

33. Copaxone [package insert]. Overland Park, KS: Teva Neuroscience, Inc; 2014. 

34. Avonex [package insert]. Cambridge, MA: Biogen Idec Inc; 1996. 

35. Rebif [package insert]. Rockland, MA: EMD Serono, Inc; New York, NY: Pfizer, Inc; 2012. 

36. Betaseron [package insert]. Montville, NJ: Bayer HealthCare Pharmaceuticals Inc; 2012. 

37. Extavia [package insert]. East Hanover, NJ: Novartis Pharmaceuticals Corp; 2014. 

38. Plegridy [package insert]. Cambridge, MA: Biogen Idec Inc; 2013. 

39. Calabresi PA, Kieseier BC, Arnold DL, et al. Pegylated interferon ß-1a for relapsing-remitting multiple sclerosis (ADVANCE): a randomised, phase 3, double-blind study. Lancet Neurol. 2014;13(7):657-665. 

40. Tecfidera [package insert]. Cambridge, MA: Biogen Idec Inc; 2015. 

41. Gilenya [package insert]. East Hanover, NJ: Novartis Pharmaceuticals Corp; 2016. 

42. Aubagio [package insert]. Cambridge, MA: Genzyme Corp; 2012.

43. Lemtrada [package insert]. Cambridge, MA: Genzyme Corp; 2014. 

44. Cohen JA, Coles AJ, Arnold DL, et al; CARE-MS I investigators. Alemtuzumab versus interferon beta 1a as first-line treatment for patients with relapsing-remitting multiple sclerosis: a randomised controlled phase 3 trial. Lancet. 2012;380(9856):1819-1828. 

45. Coles AJ, Twyman CL, Arnold DL, et al; CARE-MS II investigators. Alemtuzumab for patients with relapsing multiple sclerosis after disease-modifying therapy: a randomised controlled phase 3 trial. Lancet. 2012;380(9856):1829-1839. 

46. Novantrone [package insert]. Rockland, MA: EMD Serono, Inc; 2008. 

47. Tysabri [medication guide]. Cambridge, MA: Biogen Idec Inc; 2015.

48. Hartung DM, Bourdette DN, Ahmed SM, Whitham RH. The cost of multiple sclerosis drugs in the US and the pharmaceutical industry: too big to fail. Neurology. 2015;84(21):2185-2192. 

49. Cohen BA, Coyle PK, Leist T, Oleen-Burkey MA, Schwartz M, Zwibel H. Therapy Optimization in Multiple Sclerosis: a cohort study of therapy adherence and risk of relapse. Mult Scler Relat Disord. 2015;4(1):75-82.

50. Cohen B, Leist T, Coyle P, Zwibel H, Markowitz C, Tullman M. MS therapy adherence and relapse risk. Neurology. 2013;80(7) (suppl):P01.193. 

51. Richert ND, Zierak MC, Bash CN, Lewis BK, McFarland HF, Frank JA. MRI and clinical activity in MS patients after terminating treatment with interferon beta-1b. Mult Scler. 2000;6(2):86-90. 

52. Siger M, Durko A, Nicpan A, Konarska M, Grudziecka M, Selmaj K. Discontinuation of interferon beta therapy in multiple sclerosis patients with high pre-treatment disease activity leads to prompt return to previous disease activity. J Neurol Sci. 2011;303(1-2):50-52. 

53. Wu X, Dastidar P, Kuusisto H, Ukkonen M, Huhtala H, Elovaara I. Increased disability and MRI lesions after discontinuation of IFN-beta-1a in secondary progressive MS. Acta Neurol Scand. 2005;112(4):242-247. 

54. Scalfari A, Neuhaus A, Degenhardt A, et al. The natural history of multiple sclerosis: a geographically based study 10: relapses and long-term disability. Brain. 2010;133(pt 7):1914-1929. 

55. Bates D. Treatment effects of immunomodulatory therapies at different stages of multiple sclerosis in short-term trials. Neurology. 2011;76(1)(suppl 1):S14-S25. 

56. Fisniku LK, Brex PA, Altmann DR, et al. Disability and T2 MRI lesions: a 20-year follow-up of patients with relapse onset of multiple sclerosis. Brain. 2008;131(pt 3):808-817. 

57. Cross AH, Naismith RT. Established and novel disease-modifying treatments in multiple sclerosis. J Intern Med. 2014;275(4):350-363. 

58. Lublin FD, Baier M, Cutter G. Effect of relapses on development of residual deficit in multiple sclerosis. Neurology. 2003;61(11):1528-1532. 

59. Kalb R. The emotional and psychological impact of multiple sclerosis relapses. J Neurol Sci. 2007;256(suppl 1):S29-S33.

Issue
Federal Practitioner - 33(6)
Issue
Federal Practitioner - 33(6)
Page Number
28-33
Page Number
28-33
Publications
Publications
Topics
Article Type
Display Headline
Disease-Modifying Therapies in Multiple Sclerosis: Overview and Treatment Considerations
Display Headline
Disease-Modifying Therapies in Multiple Sclerosis: Overview and Treatment Considerations
Legacy Keywords
Derrick Robertson MD, Natalie Moreo MD, multiple sclerosis, MS
Legacy Keywords
Derrick Robertson MD, Natalie Moreo MD, multiple sclerosis, MS
Sections
Disallow All Ads
Alternative CME
Use ProPublica
Article PDF Media

The Relationship Between Male Patients’ Antihypertensive Medication Beliefs and Erectile Function

Article Type
Changed
Tue, 04/24/2018 - 14:51
Display Headline
The Relationship Between Male Patients’ Antihypertensive Medication Beliefs and Erectile Function
There was no association between the demographic variables of age, race, marital status, and education and respondents’ medication beliefs or erectile function in this pilot study.

Erectile dysfunction (ED) is a multifactorial disease that can lead to treatment nonadherence in men taking medications for chronic health problems. Despite the importance of adhering to medication protocols for disease management, several studies demonstrated that a lack of adherence to medication protocols may be a direct response to the development of ED.

The Health Belief Model (HBM) is a conceptual framework used to better understand the relationship between health behavior and adherence in men diagnosed with hypertension. The HBM consists of 3 components that help explain health care behaviors: (1) individual perceptions; (2) modifying factors; and (3) likelihood of action. These components align with the variables in this study, which include beliefs about medicines, illness perception, and erectile function.1 Thus, the research challenge was to determine the relationship between knowledge of hypertension and sexual satisfaction among men with hypertension by using instruments in the form of 2 questionnaires: (1) the Beliefs about Medicines Questionnaire (BMQ), and (2) the International Index of Erectile Functioning (IIEF).2

Methods

A descriptive correlational design was used to explore the relationship between male patients’ antihypertensive medication beliefs and erectile function.

A convenience sample of 47 eligible men (age range 18-75 years) was recruited for this pilot study. Inclusion criteria were hypertension diagnosed by a medical professional, current use of antihypertensive medication, ability to read and write in English, and a signed informed consent. Study participants were recruited from a nurse-managed center in Detroit, Michigan, and from 6 facilities identified through Project Healthy Living 2012. The mission of the nurse-managed center is to assist low-income and underinsured populations in Wayne County, Michigan. Project Healthy Living 2012 performs free and low-cost health screenings and provides general health information to the public at various locations in the Detroit metropolitan area. Written permission to collect data was obtained from the director of Project Healthy Living 2012. Recruitment began after institutional review board approval was obtained. Participants signed an informed consent form that fully explained the nature of the study and the benefits and potential risks of participation.

Instruments

This study used the BMQ and IIEF health-related questionnaires. In addition, demographic information (eg, age, race, marital status, education, length of time with hypertension diagnosis, and current antihypertensive medications) was collected.

The BMQ was originally developed to understand patients’ commonly held beliefs about medications and the factors influencing their adherence to prescribed medication regimens.3 This questionnaire has 2 scales, BMQ Specific and BMQ General (only BMQ Specific was relevant to this study).

The BMQ Specific scale consists of two 5-item subscales, specific-necessity and specific-concern, that are designed to assess respondents’ beliefs about prescribed medications they are taking for specific conditions, such as hypertension. According to Horne and colleagues, the specific-necessity construct represents the perceived role of medication in protecting against deterioration of present and future health status.4 Statement examples include “My current health depends on my medicines,” and “My medicines protect me from becoming worse.” The specific-concern construct has an emotional component (eg, “Having to take my medicines worries me”) and a cognitive component (eg, “My medicines are a mystery to me”).

Items are scored on a 5-point Likert scale ranging from 1 (strongly disagree) to 5 (strongly agree). Higher BMQ scores signify stronger beliefs. Studies using the BMQ subscales have shown that respondents with stronger beliefs about the necessity of their medications and fewer concerns about their medications are more likely to be adherent.

The self-administered IIEF is the instrument most widely used to assess sexual functioning and has been deployed in numerous clinical trials as the primary endpoint in evaluating the efficacy of phosphodiesterase 5-inhibitor therapy in the treatment of ED. The IIEF consists of 15 statements grouped into 5 domains assessing erectile function (6 items), orgasmic function (2 items), sexual desire (2 items), intercourse satisfaction (3 items), and overall satisfaction (2 items). Each domain is scored separately (Table 1).

Data Collection and Analysis

The researcher administered paper-and-pencil versions of the BMQ and the IIEF. Participants were allowed to complete these surveys in a semiprivate setting. No identifying information was collected—only general demographic information. IBM SPSS Version 16 statistical software was used to analyze the data.

Univariate descriptive statistics were run on all study variables, and Spearman correlation coefficients were computed to answer the research question regarding the relationship between specific beliefs about antihypertensive medications and self-reported erectile functioning in men with hypertension.

Results

Forty-five men who were taking antihypertensive medication prescribed by a primary care provider agreed to participate. Their mean age was 55.8 years. The majority were African American (82%), married (58%) with some post-high school education (76%) (Table 2). Mean length of time with a hypertension diagnosis was 9.2 years. All respondents reported taking at least 1 antihypertensive medication. The medication most often reported being used was amlodipine, followed by lisinopril and metoprolol.

 

 

All of the respondents self-reported sexual dysfunction as indicated by the mean scores on the 5 domains of the IIEF (Table 3). Moreover, the vast majority of the total scores on the 5 domains fell within the moderate-to-severe dysfunction levels (Table 4).

 

BMQ Scores

The mean BMQ specific-necessity score was 16.7 (Table 5). More than half the respondents agreed or strongly agreed with 4 of the 5 statements regarding the necessity of their medications; the exception was “My life would be impossible without my medicines” (29.8%). The statement that generated the highest percentage of agreement was “My medicines protect me from becoming worse” (72.3%) (Table 6).

 

The mean BMQ specific-concern score was 13.7. Less than one-fourth of the respondents agreed or strongly agreed with 3 statements (of 4) regarding medication concerns: “My medicine is a mystery to me” (21.3%), “I sometimes worry about becoming too dependent on my medicine” (24.1%), and “My medicines disrupt my life” (25.6%). Regarding the fourth statement, “I sometimes worry about the long-term effects of my medicines,” 58.7% agreed or strongly agreed.

Spearman correlation analyses were performed to examine the relationships between BMQ specific-necessity, BMQ specific-concern, and the 5 IIEF sexual functioning domains. No statistically significant relationships were detected.

Time With Hypertension Diagnosis

The study also evaluated the results based on length of time the men had a hypertension diagnosis (≤ 10 years vs ≥ 11 years). An independent t test was used to compare mean BMQ specific-necessity and specific-concern scores. Crosstabs was used to compare length of time with diagnosis and the 5 IIEF domains. Mean specific-necessity score was 18.1 for the 20 respondents with a diagnosis of ≥ 11 years and 15.0 for the 22 respondents with a diagnosis of ≤ 10 years; this difference was statistically significant (P = .048). In addition, mean specific-concern score was 14.7 for respondents with a diagnosis of ≥ 11 years and 12.4 for those with a diagnosis of ≤ 10 years; this difference was not statistically significant but was trending in that direction (P = .076).

Length of time with a hypertension diagnosis did not necessarily relate to sexual function or satisfaction. Moderate-to-severe dysfunction in sexual desire was reported by 42% of the respondents with a diagnosis of ≥ 11 years compared with 57% of those with a diagnosis of ≤ 10 years. The same was found for overall satisfaction.

Discussion

Men with chronic health conditions, such as hypertension, are often prescribed medications that interfere with their sexual functioning.5 Patients who do not understand the importance of medication adherence in treating long-term chronic health conditions may discontinue taking these medications to improve their sexual functioning.5 Some men overestimate the control they have over chronic health conditions; others underestimate their control.

With a better understanding of men’s perceived ability to control chronic hypertension, practitioners can begin to provide appropriate educational interventions. These interventions must inform patients about the role of medication adherence in maintaining and enhancing sexual functioning and must impart a greater sense of control and empowerment over their condition, thereby increasing the likelihood of medication adherence.

To the authors’ knowledge, this is the first study to examine the potential associations of medication beliefs (using the BMQ) and erectile function in men with hypertension. There was no association between any of the demographic variables of age, race, marital status, and education, with respondents’ medication beliefs or erectile function. The IIEF domain that had the highest level of severe and moderate dysfunction combined was orgasmic function (n = 36; 80%), which suggests that antihypertensive medication may interfere with orgasmic function because achieving an orgasm is a physiologic process. This is consistent with the findings of Hellstrom and colleagues—that the vast majority of ED is related to orgasmic factors.6 Although 80% of respondents reported either moderate or severe dysfunction in this area, only 53% (n = 24) reported moderate or severe dysfunction in overall satisfaction. This might suggest that men taking antihypertensive medication, and their partners, have learned other methods or strategies for achieving sexual satisfaction.

The BMQ specific-necessity and specific-concern results offer insight into how men with hypertension view the need for antihypertensive medication. Mean (SD) specific-necessity score was 16.7 (5); the maximum was 25. Although no other studies have investigated men’s beliefs about antihypertensive medications, studies of other chronic illnesses have measured beliefs about the medications used in treatment. For example, Neame and Hammond reported a mean specific-necessity score of 19.9 and a mean specific-concern score of 15.9 in a sample of 108 men with arthritis.7 Of the respondents, 74.3% agreed or strongly agreed that their arthritis medication was necessary for health. However, 47.4% were concerned about potential adverse effects. Lennerling and Forsberg conducted a cross-sectional study of 250 renal transplant recipients.8 Respondents reported a mean specific-necessity score of 23 and a median specific-concern score of 11. In addition, length of time with hypertension diagnosis affected specific-necessity and specific-concern scores. For example, men with a hypertension diagnosis for 10 or fewer years had higher scores on both of these BMQ subscales.

 

 

The inability to detect a statistically significant correlation between BMQ specific-necessity and the 5 IIEF domains suggests that the sexual functioning of men with hypertension is not necessarily related to their antihypertensive medication beliefs. More than 50% agreed or strongly agreed that antihypertensive medications were necessary, but not that life would be impossible without them. Moreover, there was no statistically significant relationship between any of the 5 IIEF domains and the BMQ specific-concern subscale, which suggests that the sexual functioning of men with hypertension may not be related to their concern about their medications.

The findings of this study support HBM and provide a possible explanation for medication nonadherence in men diagnosed with hypertension. The HBM provides a conceptual framework that positions the study variables (eg, beliefs about medicines, illness perception, and erectile function) in terms of health care behaviors that can lead to increased medication adherence among men with hypertension. The HBM also allows the researcher to gain a better understanding about participants’ health behaviors, how these health behaviors are determined by personal beliefs or perceptions about a disease, and strategies available to decrease nonadherence.1

The HBM postulates that patients’ appraisals of disease risk (susceptibility) and severity influence their behavior.9 One study assessed respondents’ perceptions of the seriousness of their hypertension and its relationship to ED and of the consequences of failing to take prescribed blood pressure medications.9 When using the HBM, it is important to consider patients’ perceived barriers to and incentives for engaging in specific behaviors. The same study used the BMQ to address specific questions about respondents’ beliefs and feelings about their prescribed medications and medication adherence. The HBM suggests considering action cues that encourage patients to act by reminding them of the need to change their behavior.

 

Limitations

This study had several limitations. First, the sample size was small (47), which makes it difficult to generalize findings to a broader population of men with a hypertension diagnosis. In addition, because the study was underpowered, its ability to detect significant differences was compromised. Second, the study used a convenience sample of predominantly African American men. As always, there are concerns of self-selection and failure to represent the overall population. Third, the setting for completing the surveys was only semiprivate, and some respondents may have been uncomfortable, perhaps, working too quickly and not really thinking about the questions or their answers. Fourth, the ED survey was self-administered, so there is a concern about the truthfulness of responses. Fifth, failure to ask respondents whether they were taking a phosphodiesterase 5 inhibitor for ED could have significantly impacted study findings. Sixth, respondents were not asked about other medications, such as antidepressants and nonsteroidal anti-inflammatory drugs, which could have affected erectile function.

Clinical Implications

Despite the study limitations, several findings have important clinical implications. First, the vast majority of participants in this pilot study self-reported moderate or severe sexual dysfunction on all 5 IIEF domains. This finding is important because this was a convenience sample, and many of the IIEF statements are personal. The high rate of reported sexual dysfunction suggests that the incidence of ED may be underreported in the larger population. Second, mean BMQ scores were similar to those reported in other studies involving chronic illness: higher necessity and lower concern. Third, there was no statistically significant relationship between BMQ necessity and concern and IIEF sexual functioning. More research is needed to determine how to interpret these findings. Fourth, there was a significant relationship between length of time with hypertension diagnosis and BMQ specific-necessity score: The longer the diagnosis, the higher the score. However, this relationship did not hold for BMQ specific-concern, though it trended toward significance. Moreover, length of time with hypertension diagnosis did not necessarily predict or influence erectile function as measured with the IIEF. In fact, men with a hypertension diagnosis of ≥ 11 years reported less moderate-to-severe sexual dysfunction in overall satisfaction and sexual desire. Although there are several methodologic concerns about this study, its results offer direction for both clinical practice and future research.

Studies of erectile function and its relationship to hypertension have generated both cause for concern and reason for further research. The present study focused on gaining a better understanding of the relationship between antihypertensive medication beliefs and erectile function. Future clinical studies should explore the effects of antihypertensive medication on erectile function and men’s lack of knowledge and education about the importance of taking medication to prevent complications of hypertension. It is essential that this research be applied to improve the understanding of erectile function in men with hypertension. This will ultimately allow for better patient management and contribute to the overall sexual health and well-being of patients with hypertension.

Although it is important to identify men’s antihypertensive medication beliefs and the relationship of these beliefs to sexual satisfaction, most longitudinal studies suggest that the ED rate is high and that it increases with age.10 Therefore, it is crucial that men differentiate between how antihypertensive medications affect erectile function and changes associated with aging. The present study found no statistically significant relationships between the BMQ specific-necessity and specific-concern subscales and overall satisfaction with sexual functioning (IIEF). In addition, the study found no significant correlations between the BMQ specific and general scales and the 5 IIEF domains.

Conclusion

It was evident from this research that men with chronic health problems, such as hypertension, are often prescribed medications that affect sexual functioning. Unfortunately, the effect on sexual functioning often plays a significant role in the discontinuation of long-term therapy. Many of this study’s participants self-reported moderate or severe sexual dysfunction. Results showed no statistically significant relationships between either BMQ subscale or any of the 5 IIEF domains. Research is needed to further explore the association between ED and antihypertensive medication and men’s lack of knowledge and education about the importance of treatment adherence.

References

1. Hochbaum GM; U.S. Public Health Service, Division of Special Health Services. Public Participation in Medical Screening Programs: A Socio-Psychological Study. Public Health Service publication, no. 572 Washington, DC: U.S. Dept. of Health, Education, and Welfare, Public Health Service, Bureau of State Services, Division of Special Health Services, Tuberculosis Program; 1958.

2. International Index of Erectile Function (IIEF). Weill Cornell Medical College James Buchanan Brady Foundation Department of Urology website. https://www.cornellurology.com/images/uploads/IBPH-IIEF-Assessments.pdf. Accessed May 14, 2016.

3. Porteous T, Francis J, Bond C, Hannaford P. Temporal stability of beliefs about medicines: implications for optimizing adherence. Patient Educ Couns. 2010; 79(2):225-230.

4. Horne R, Weinman J, Hankins M. The beliefs about medicines questionnaire: the development and evaluation of a new method for assessing the cognitive representation of medication. Psychol Health. 1999;14(1):1-24.

5. Fogari R, Zoppi A, Corradi L, Mugellini A, Poletti L, Lusardi P. Sexual function in hypertensive males treated with lisinopril or atenolol: a cross-over study. Am J Hypertens. 1998;11(10):1244-1247.

6. Hellstrom WJ, Gittelman M, Karlin G, et al. Vardenafil for treatment of men with erectile dysfunction: efficacy and safety in a randomized, double-blind, placebo-controlled trial. J Androl. 2002;23(6):763-771.

7. Neame R, Hammond A. Beliefs about medications: a questionnaire survey of people with rheumatoid arthritis. Rheumatology (Oxford). 2005;44(6):762-767.

8. Lennerling A, Forsberg A. Self-reported non-adherence and beliefs about medication in a Swedish kidney transplant population. Open Nurs J. 2012;6:41-46.

9. Kressin NR, Wang F, Long J, et al. Hypertensive patients' race, health beliefs, process of care, and medication adherence. J Gen Intern Med. 2007; 22(6):768-774.

10. Inman BA, Sauver JL, Jacobson DJ, et al. A population-based, longitudinal study of erectile dysfunction and future coronary artery disease. Mayo Clin Proc. 2009;84(2):108-113.

Article PDF
Author and Disclosure Information

Dr. Gray is a board-certified family nurse practitioner in primary care at the John D. Dingell VAMC in Detroit, Michigan, and a PhD candidate in educational studies at Eastern Michigan University in Ypsilanti. Dr. Groh is a board-certified psychiatric nurse practitioner at the McAuley Health Center in Detroit, and professor at the University of Detroit Mercy College of Health Professions. 

Author disclosures
The authors report no actual or potential conflicts of interest with regard to this article.

Disclaimer
The opinions expressed herein are those of the authors and do not necessarily reflect those of Federal Practitioner, Frontline Medical Communications Inc., the U.S. Government, or any of its agencies. This article may discuss unlabeled or investigational use of certain drugs. Please review the complete prescribing information for specific drugs or drug combinations—including indications, contraindications, warnings, and adverse effects—before administering pharmacologic therapy to patients.

 

Issue
Federal Practitioner - 33(6)
Publications
Topics
Page Number
36-41
Legacy Keywords
adherence, hypertension
Sections
Author and Disclosure Information

Dr. Gray is a board-certified family nurse practitioner in primary care at the John D. Dingell VAMC in Detroit, Michigan, and a PhD candidate in educational studies at Eastern Michigan University in Ypsilanti. Dr. Groh is a board-certified psychiatric nurse practitioner at the McAuley Health Center in Detroit, and professor at the University of Detroit Mercy College of Health Professions. 

Author disclosures
The authors report no actual or potential conflicts of interest with regard to this article.

Disclaimer
The opinions expressed herein are those of the authors and do not necessarily reflect those of Federal Practitioner, Frontline Medical Communications Inc., the U.S. Government, or any of its agencies. This article may discuss unlabeled or investigational use of certain drugs. Please review the complete prescribing information for specific drugs or drug combinations—including indications, contraindications, warnings, and adverse effects—before administering pharmacologic therapy to patients.

 

Author and Disclosure Information

Dr. Gray is a board-certified family nurse practitioner in primary care at the John D. Dingell VAMC in Detroit, Michigan, and a PhD candidate in educational studies at Eastern Michigan University in Ypsilanti. Dr. Groh is a board-certified psychiatric nurse practitioner at the McAuley Health Center in Detroit, and professor at the University of Detroit Mercy College of Health Professions. 

Author disclosures
The authors report no actual or potential conflicts of interest with regard to this article.

Disclaimer
The opinions expressed herein are those of the authors and do not necessarily reflect those of Federal Practitioner, Frontline Medical Communications Inc., the U.S. Government, or any of its agencies. This article may discuss unlabeled or investigational use of certain drugs. Please review the complete prescribing information for specific drugs or drug combinations—including indications, contraindications, warnings, and adverse effects—before administering pharmacologic therapy to patients.

 

Article PDF
Article PDF
Related Articles
There was no association between the demographic variables of age, race, marital status, and education and respondents’ medication beliefs or erectile function in this pilot study.
There was no association between the demographic variables of age, race, marital status, and education and respondents’ medication beliefs or erectile function in this pilot study.

Erectile dysfunction (ED) is a multifactorial disease that can lead to treatment nonadherence in men taking medications for chronic health problems. Despite the importance of adhering to medication protocols for disease management, several studies demonstrated that a lack of adherence to medication protocols may be a direct response to the development of ED.

The Health Belief Model (HBM) is a conceptual framework used to better understand the relationship between health behavior and adherence in men diagnosed with hypertension. The HBM consists of 3 components that help explain health care behaviors: (1) individual perceptions; (2) modifying factors; and (3) likelihood of action. These components align with the variables in this study, which include beliefs about medicines, illness perception, and erectile function.1 Thus, the research challenge was to determine the relationship between knowledge of hypertension and sexual satisfaction among men with hypertension by using instruments in the form of 2 questionnaires: (1) the Beliefs about Medicines Questionnaire (BMQ), and (2) the International Index of Erectile Functioning (IIEF).2

Methods

A descriptive correlational design was used to explore the relationship between male patients’ antihypertensive medication beliefs and erectile function.

A convenience sample of 47 eligible men (age range 18-75 years) was recruited for this pilot study. Inclusion criteria were hypertension diagnosed by a medical professional, current use of antihypertensive medication, ability to read and write in English, and a signed informed consent. Study participants were recruited from a nurse-managed center in Detroit, Michigan, and from 6 facilities identified through Project Healthy Living 2012. The mission of the nurse-managed center is to assist low-income and underinsured populations in Wayne County, Michigan. Project Healthy Living 2012 performs free and low-cost health screenings and provides general health information to the public at various locations in the Detroit metropolitan area. Written permission to collect data was obtained from the director of Project Healthy Living 2012. Recruitment began after institutional review board approval was obtained. Participants signed an informed consent form that fully explained the nature of the study and the benefits and potential risks of participation.

Instruments

This study used the BMQ and IIEF health-related questionnaires. In addition, demographic information (eg, age, race, marital status, education, length of time with hypertension diagnosis, and current antihypertensive medications) was collected.

The BMQ was originally developed to understand patients’ commonly held beliefs about medications and the factors influencing their adherence to prescribed medication regimens.3 This questionnaire has 2 scales, BMQ Specific and BMQ General (only BMQ Specific was relevant to this study).

The BMQ Specific scale consists of two 5-item subscales, specific-necessity and specific-concern, that are designed to assess respondents’ beliefs about prescribed medications they are taking for specific conditions, such as hypertension. According to Horne and colleagues, the specific-necessity construct represents the perceived role of medication in protecting against deterioration of present and future health status.4 Statement examples include “My current health depends on my medicines,” and “My medicines protect me from becoming worse.” The specific-concern construct has an emotional component (eg, “Having to take my medicines worries me”) and a cognitive component (eg, “My medicines are a mystery to me”).

Items are scored on a 5-point Likert scale ranging from 1 (strongly disagree) to 5 (strongly agree). Higher BMQ scores signify stronger beliefs. Studies using the BMQ subscales have shown that respondents with stronger beliefs about the necessity of their medications and fewer concerns about their medications are more likely to be adherent.

The self-administered IIEF is the instrument most widely used to assess sexual functioning and has been deployed in numerous clinical trials as the primary endpoint in evaluating the efficacy of phosphodiesterase 5-inhibitor therapy in the treatment of ED. The IIEF consists of 15 statements grouped into 5 domains assessing erectile function (6 items), orgasmic function (2 items), sexual desire (2 items), intercourse satisfaction (3 items), and overall satisfaction (2 items). Each domain is scored separately (Table 1).

Data Collection and Analysis

The researcher administered paper-and-pencil versions of the BMQ and the IIEF. Participants were allowed to complete these surveys in a semiprivate setting. No identifying information was collected—only general demographic information. IBM SPSS Version 16 statistical software was used to analyze the data.

Univariate descriptive statistics were run on all study variables, and Spearman correlation coefficients were computed to answer the research question regarding the relationship between specific beliefs about antihypertensive medications and self-reported erectile functioning in men with hypertension.

Results

Forty-five men who were taking antihypertensive medication prescribed by a primary care provider agreed to participate. Their mean age was 55.8 years. The majority were African American (82%), married (58%) with some post-high school education (76%) (Table 2). Mean length of time with a hypertension diagnosis was 9.2 years. All respondents reported taking at least 1 antihypertensive medication. The medication most often reported being used was amlodipine, followed by lisinopril and metoprolol.

 

 

All of the respondents self-reported sexual dysfunction as indicated by the mean scores on the 5 domains of the IIEF (Table 3). Moreover, the vast majority of the total scores on the 5 domains fell within the moderate-to-severe dysfunction levels (Table 4).

 

BMQ Scores

The mean BMQ specific-necessity score was 16.7 (Table 5). More than half the respondents agreed or strongly agreed with 4 of the 5 statements regarding the necessity of their medications; the exception was “My life would be impossible without my medicines” (29.8%). The statement that generated the highest percentage of agreement was “My medicines protect me from becoming worse” (72.3%) (Table 6).

 

The mean BMQ specific-concern score was 13.7. Less than one-fourth of the respondents agreed or strongly agreed with 3 statements (of 4) regarding medication concerns: “My medicine is a mystery to me” (21.3%), “I sometimes worry about becoming too dependent on my medicine” (24.1%), and “My medicines disrupt my life” (25.6%). Regarding the fourth statement, “I sometimes worry about the long-term effects of my medicines,” 58.7% agreed or strongly agreed.

Spearman correlation analyses were performed to examine the relationships between BMQ specific-necessity, BMQ specific-concern, and the 5 IIEF sexual functioning domains. No statistically significant relationships were detected.

Time With Hypertension Diagnosis

The study also evaluated the results based on length of time the men had a hypertension diagnosis (≤ 10 years vs ≥ 11 years). An independent t test was used to compare mean BMQ specific-necessity and specific-concern scores. Crosstabs was used to compare length of time with diagnosis and the 5 IIEF domains. Mean specific-necessity score was 18.1 for the 20 respondents with a diagnosis of ≥ 11 years and 15.0 for the 22 respondents with a diagnosis of ≤ 10 years; this difference was statistically significant (P = .048). In addition, mean specific-concern score was 14.7 for respondents with a diagnosis of ≥ 11 years and 12.4 for those with a diagnosis of ≤ 10 years; this difference was not statistically significant but was trending in that direction (P = .076).

Length of time with a hypertension diagnosis did not necessarily relate to sexual function or satisfaction. Moderate-to-severe dysfunction in sexual desire was reported by 42% of the respondents with a diagnosis of ≥ 11 years compared with 57% of those with a diagnosis of ≤ 10 years. The same was found for overall satisfaction.

Discussion

Men with chronic health conditions, such as hypertension, are often prescribed medications that interfere with their sexual functioning.5 Patients who do not understand the importance of medication adherence in treating long-term chronic health conditions may discontinue taking these medications to improve their sexual functioning.5 Some men overestimate the control they have over chronic health conditions; others underestimate their control.

With a better understanding of men’s perceived ability to control chronic hypertension, practitioners can begin to provide appropriate educational interventions. These interventions must inform patients about the role of medication adherence in maintaining and enhancing sexual functioning and must impart a greater sense of control and empowerment over their condition, thereby increasing the likelihood of medication adherence.

To the authors’ knowledge, this is the first study to examine the potential associations of medication beliefs (using the BMQ) and erectile function in men with hypertension. There was no association between any of the demographic variables of age, race, marital status, and education, with respondents’ medication beliefs or erectile function. The IIEF domain that had the highest level of severe and moderate dysfunction combined was orgasmic function (n = 36; 80%), which suggests that antihypertensive medication may interfere with orgasmic function because achieving an orgasm is a physiologic process. This is consistent with the findings of Hellstrom and colleagues—that the vast majority of ED is related to orgasmic factors.6 Although 80% of respondents reported either moderate or severe dysfunction in this area, only 53% (n = 24) reported moderate or severe dysfunction in overall satisfaction. This might suggest that men taking antihypertensive medication, and their partners, have learned other methods or strategies for achieving sexual satisfaction.

The BMQ specific-necessity and specific-concern results offer insight into how men with hypertension view the need for antihypertensive medication. Mean (SD) specific-necessity score was 16.7 (5); the maximum was 25. Although no other studies have investigated men’s beliefs about antihypertensive medications, studies of other chronic illnesses have measured beliefs about the medications used in treatment. For example, Neame and Hammond reported a mean specific-necessity score of 19.9 and a mean specific-concern score of 15.9 in a sample of 108 men with arthritis.7 Of the respondents, 74.3% agreed or strongly agreed that their arthritis medication was necessary for health. However, 47.4% were concerned about potential adverse effects. Lennerling and Forsberg conducted a cross-sectional study of 250 renal transplant recipients.8 Respondents reported a mean specific-necessity score of 23 and a median specific-concern score of 11. In addition, length of time with hypertension diagnosis affected specific-necessity and specific-concern scores. For example, men with a hypertension diagnosis for 10 or fewer years had higher scores on both of these BMQ subscales.

 

 

The inability to detect a statistically significant correlation between BMQ specific-necessity and the 5 IIEF domains suggests that the sexual functioning of men with hypertension is not necessarily related to their antihypertensive medication beliefs. More than 50% agreed or strongly agreed that antihypertensive medications were necessary, but not that life would be impossible without them. Moreover, there was no statistically significant relationship between any of the 5 IIEF domains and the BMQ specific-concern subscale, which suggests that the sexual functioning of men with hypertension may not be related to their concern about their medications.

The findings of this study support HBM and provide a possible explanation for medication nonadherence in men diagnosed with hypertension. The HBM provides a conceptual framework that positions the study variables (eg, beliefs about medicines, illness perception, and erectile function) in terms of health care behaviors that can lead to increased medication adherence among men with hypertension. The HBM also allows the researcher to gain a better understanding about participants’ health behaviors, how these health behaviors are determined by personal beliefs or perceptions about a disease, and strategies available to decrease nonadherence.1

The HBM postulates that patients’ appraisals of disease risk (susceptibility) and severity influence their behavior.9 One study assessed respondents’ perceptions of the seriousness of their hypertension and its relationship to ED and of the consequences of failing to take prescribed blood pressure medications.9 When using the HBM, it is important to consider patients’ perceived barriers to and incentives for engaging in specific behaviors. The same study used the BMQ to address specific questions about respondents’ beliefs and feelings about their prescribed medications and medication adherence. The HBM suggests considering action cues that encourage patients to act by reminding them of the need to change their behavior.

 

Limitations

This study had several limitations. First, the sample size was small (47), which makes it difficult to generalize findings to a broader population of men with a hypertension diagnosis. In addition, because the study was underpowered, its ability to detect significant differences was compromised. Second, the study used a convenience sample of predominantly African American men. As always, there are concerns of self-selection and failure to represent the overall population. Third, the setting for completing the surveys was only semiprivate, and some respondents may have been uncomfortable, perhaps, working too quickly and not really thinking about the questions or their answers. Fourth, the ED survey was self-administered, so there is a concern about the truthfulness of responses. Fifth, failure to ask respondents whether they were taking a phosphodiesterase 5 inhibitor for ED could have significantly impacted study findings. Sixth, respondents were not asked about other medications, such as antidepressants and nonsteroidal anti-inflammatory drugs, which could have affected erectile function.

Clinical Implications

Despite the study limitations, several findings have important clinical implications. First, the vast majority of participants in this pilot study self-reported moderate or severe sexual dysfunction on all 5 IIEF domains. This finding is important because this was a convenience sample, and many of the IIEF statements are personal. The high rate of reported sexual dysfunction suggests that the incidence of ED may be underreported in the larger population. Second, mean BMQ scores were similar to those reported in other studies involving chronic illness: higher necessity and lower concern. Third, there was no statistically significant relationship between BMQ necessity and concern and IIEF sexual functioning. More research is needed to determine how to interpret these findings. Fourth, there was a significant relationship between length of time with hypertension diagnosis and BMQ specific-necessity score: The longer the diagnosis, the higher the score. However, this relationship did not hold for BMQ specific-concern, though it trended toward significance. Moreover, length of time with hypertension diagnosis did not necessarily predict or influence erectile function as measured with the IIEF. In fact, men with a hypertension diagnosis of ≥ 11 years reported less moderate-to-severe sexual dysfunction in overall satisfaction and sexual desire. Although there are several methodologic concerns about this study, its results offer direction for both clinical practice and future research.

Studies of erectile function and its relationship to hypertension have generated both cause for concern and reason for further research. The present study focused on gaining a better understanding of the relationship between antihypertensive medication beliefs and erectile function. Future clinical studies should explore the effects of antihypertensive medication on erectile function and men’s lack of knowledge and education about the importance of taking medication to prevent complications of hypertension. It is essential that this research be applied to improve the understanding of erectile function in men with hypertension. This will ultimately allow for better patient management and contribute to the overall sexual health and well-being of patients with hypertension.

Although it is important to identify men’s antihypertensive medication beliefs and the relationship of these beliefs to sexual satisfaction, most longitudinal studies suggest that the ED rate is high and that it increases with age.10 Therefore, it is crucial that men differentiate between how antihypertensive medications affect erectile function and changes associated with aging. The present study found no statistically significant relationships between the BMQ specific-necessity and specific-concern subscales and overall satisfaction with sexual functioning (IIEF). In addition, the study found no significant correlations between the BMQ specific and general scales and the 5 IIEF domains.

Conclusion

It was evident from this research that men with chronic health problems, such as hypertension, are often prescribed medications that affect sexual functioning. Unfortunately, the effect on sexual functioning often plays a significant role in the discontinuation of long-term therapy. Many of this study’s participants self-reported moderate or severe sexual dysfunction. Results showed no statistically significant relationships between either BMQ subscale or any of the 5 IIEF domains. Research is needed to further explore the association between ED and antihypertensive medication and men’s lack of knowledge and education about the importance of treatment adherence.

Erectile dysfunction (ED) is a multifactorial disease that can lead to treatment nonadherence in men taking medications for chronic health problems. Despite the importance of adhering to medication protocols for disease management, several studies demonstrated that a lack of adherence to medication protocols may be a direct response to the development of ED.

The Health Belief Model (HBM) is a conceptual framework used to better understand the relationship between health behavior and adherence in men diagnosed with hypertension. The HBM consists of 3 components that help explain health care behaviors: (1) individual perceptions; (2) modifying factors; and (3) likelihood of action. These components align with the variables in this study, which include beliefs about medicines, illness perception, and erectile function.1 Thus, the research challenge was to determine the relationship between knowledge of hypertension and sexual satisfaction among men with hypertension by using instruments in the form of 2 questionnaires: (1) the Beliefs about Medicines Questionnaire (BMQ), and (2) the International Index of Erectile Functioning (IIEF).2

Methods

A descriptive correlational design was used to explore the relationship between male patients’ antihypertensive medication beliefs and erectile function.

A convenience sample of 47 eligible men (age range 18-75 years) was recruited for this pilot study. Inclusion criteria were hypertension diagnosed by a medical professional, current use of antihypertensive medication, ability to read and write in English, and a signed informed consent. Study participants were recruited from a nurse-managed center in Detroit, Michigan, and from 6 facilities identified through Project Healthy Living 2012. The mission of the nurse-managed center is to assist low-income and underinsured populations in Wayne County, Michigan. Project Healthy Living 2012 performs free and low-cost health screenings and provides general health information to the public at various locations in the Detroit metropolitan area. Written permission to collect data was obtained from the director of Project Healthy Living 2012. Recruitment began after institutional review board approval was obtained. Participants signed an informed consent form that fully explained the nature of the study and the benefits and potential risks of participation.

Instruments

This study used the BMQ and IIEF health-related questionnaires. In addition, demographic information (eg, age, race, marital status, education, length of time with hypertension diagnosis, and current antihypertensive medications) was collected.

The BMQ was originally developed to understand patients’ commonly held beliefs about medications and the factors influencing their adherence to prescribed medication regimens.3 This questionnaire has 2 scales, BMQ Specific and BMQ General (only BMQ Specific was relevant to this study).

The BMQ Specific scale consists of two 5-item subscales, specific-necessity and specific-concern, that are designed to assess respondents’ beliefs about prescribed medications they are taking for specific conditions, such as hypertension. According to Horne and colleagues, the specific-necessity construct represents the perceived role of medication in protecting against deterioration of present and future health status.4 Statement examples include “My current health depends on my medicines,” and “My medicines protect me from becoming worse.” The specific-concern construct has an emotional component (eg, “Having to take my medicines worries me”) and a cognitive component (eg, “My medicines are a mystery to me”).

Items are scored on a 5-point Likert scale ranging from 1 (strongly disagree) to 5 (strongly agree). Higher BMQ scores signify stronger beliefs. Studies using the BMQ subscales have shown that respondents with stronger beliefs about the necessity of their medications and fewer concerns about their medications are more likely to be adherent.

The self-administered IIEF is the instrument most widely used to assess sexual functioning and has been deployed in numerous clinical trials as the primary endpoint in evaluating the efficacy of phosphodiesterase 5-inhibitor therapy in the treatment of ED. The IIEF consists of 15 statements grouped into 5 domains assessing erectile function (6 items), orgasmic function (2 items), sexual desire (2 items), intercourse satisfaction (3 items), and overall satisfaction (2 items). Each domain is scored separately (Table 1).

Data Collection and Analysis

The researcher administered paper-and-pencil versions of the BMQ and the IIEF. Participants were allowed to complete these surveys in a semiprivate setting. No identifying information was collected—only general demographic information. IBM SPSS Version 16 statistical software was used to analyze the data.

Univariate descriptive statistics were run on all study variables, and Spearman correlation coefficients were computed to answer the research question regarding the relationship between specific beliefs about antihypertensive medications and self-reported erectile functioning in men with hypertension.

Results

Forty-five men who were taking antihypertensive medication prescribed by a primary care provider agreed to participate. Their mean age was 55.8 years. The majority were African American (82%), married (58%) with some post-high school education (76%) (Table 2). Mean length of time with a hypertension diagnosis was 9.2 years. All respondents reported taking at least 1 antihypertensive medication. The medication most often reported being used was amlodipine, followed by lisinopril and metoprolol.

 

 

All of the respondents self-reported sexual dysfunction as indicated by the mean scores on the 5 domains of the IIEF (Table 3). Moreover, the vast majority of the total scores on the 5 domains fell within the moderate-to-severe dysfunction levels (Table 4).

 

BMQ Scores

The mean BMQ specific-necessity score was 16.7 (Table 5). More than half the respondents agreed or strongly agreed with 4 of the 5 statements regarding the necessity of their medications; the exception was “My life would be impossible without my medicines” (29.8%). The statement that generated the highest percentage of agreement was “My medicines protect me from becoming worse” (72.3%) (Table 6).

 

The mean BMQ specific-concern score was 13.7. Less than one-fourth of the respondents agreed or strongly agreed with 3 statements (of 4) regarding medication concerns: “My medicine is a mystery to me” (21.3%), “I sometimes worry about becoming too dependent on my medicine” (24.1%), and “My medicines disrupt my life” (25.6%). Regarding the fourth statement, “I sometimes worry about the long-term effects of my medicines,” 58.7% agreed or strongly agreed.

Spearman correlation analyses were performed to examine the relationships between BMQ specific-necessity, BMQ specific-concern, and the 5 IIEF sexual functioning domains. No statistically significant relationships were detected.

Time With Hypertension Diagnosis

The study also evaluated the results based on length of time the men had a hypertension diagnosis (≤ 10 years vs ≥ 11 years). An independent t test was used to compare mean BMQ specific-necessity and specific-concern scores. Crosstabs was used to compare length of time with diagnosis and the 5 IIEF domains. Mean specific-necessity score was 18.1 for the 20 respondents with a diagnosis of ≥ 11 years and 15.0 for the 22 respondents with a diagnosis of ≤ 10 years; this difference was statistically significant (P = .048). In addition, mean specific-concern score was 14.7 for respondents with a diagnosis of ≥ 11 years and 12.4 for those with a diagnosis of ≤ 10 years; this difference was not statistically significant but was trending in that direction (P = .076).

Length of time with a hypertension diagnosis did not necessarily relate to sexual function or satisfaction. Moderate-to-severe dysfunction in sexual desire was reported by 42% of the respondents with a diagnosis of ≥ 11 years compared with 57% of those with a diagnosis of ≤ 10 years. The same was found for overall satisfaction.

Discussion

Men with chronic health conditions, such as hypertension, are often prescribed medications that interfere with their sexual functioning.5 Patients who do not understand the importance of medication adherence in treating long-term chronic health conditions may discontinue taking these medications to improve their sexual functioning.5 Some men overestimate the control they have over chronic health conditions; others underestimate their control.

With a better understanding of men’s perceived ability to control chronic hypertension, practitioners can begin to provide appropriate educational interventions. These interventions must inform patients about the role of medication adherence in maintaining and enhancing sexual functioning and must impart a greater sense of control and empowerment over their condition, thereby increasing the likelihood of medication adherence.

To the authors’ knowledge, this is the first study to examine the potential associations of medication beliefs (using the BMQ) and erectile function in men with hypertension. There was no association between any of the demographic variables of age, race, marital status, and education, with respondents’ medication beliefs or erectile function. The IIEF domain that had the highest level of severe and moderate dysfunction combined was orgasmic function (n = 36; 80%), which suggests that antihypertensive medication may interfere with orgasmic function because achieving an orgasm is a physiologic process. This is consistent with the findings of Hellstrom and colleagues—that the vast majority of ED is related to orgasmic factors.6 Although 80% of respondents reported either moderate or severe dysfunction in this area, only 53% (n = 24) reported moderate or severe dysfunction in overall satisfaction. This might suggest that men taking antihypertensive medication, and their partners, have learned other methods or strategies for achieving sexual satisfaction.

The BMQ specific-necessity and specific-concern results offer insight into how men with hypertension view the need for antihypertensive medication. Mean (SD) specific-necessity score was 16.7 (5); the maximum was 25. Although no other studies have investigated men’s beliefs about antihypertensive medications, studies of other chronic illnesses have measured beliefs about the medications used in treatment. For example, Neame and Hammond reported a mean specific-necessity score of 19.9 and a mean specific-concern score of 15.9 in a sample of 108 men with arthritis.7 Of the respondents, 74.3% agreed or strongly agreed that their arthritis medication was necessary for health. However, 47.4% were concerned about potential adverse effects. Lennerling and Forsberg conducted a cross-sectional study of 250 renal transplant recipients.8 Respondents reported a mean specific-necessity score of 23 and a median specific-concern score of 11. In addition, length of time with hypertension diagnosis affected specific-necessity and specific-concern scores. For example, men with a hypertension diagnosis for 10 or fewer years had higher scores on both of these BMQ subscales.

 

 

The inability to detect a statistically significant correlation between BMQ specific-necessity and the 5 IIEF domains suggests that the sexual functioning of men with hypertension is not necessarily related to their antihypertensive medication beliefs. More than 50% agreed or strongly agreed that antihypertensive medications were necessary, but not that life would be impossible without them. Moreover, there was no statistically significant relationship between any of the 5 IIEF domains and the BMQ specific-concern subscale, which suggests that the sexual functioning of men with hypertension may not be related to their concern about their medications.

The findings of this study support HBM and provide a possible explanation for medication nonadherence in men diagnosed with hypertension. The HBM provides a conceptual framework that positions the study variables (eg, beliefs about medicines, illness perception, and erectile function) in terms of health care behaviors that can lead to increased medication adherence among men with hypertension. The HBM also allows the researcher to gain a better understanding about participants’ health behaviors, how these health behaviors are determined by personal beliefs or perceptions about a disease, and strategies available to decrease nonadherence.1

The HBM postulates that patients’ appraisals of disease risk (susceptibility) and severity influence their behavior.9 One study assessed respondents’ perceptions of the seriousness of their hypertension and its relationship to ED and of the consequences of failing to take prescribed blood pressure medications.9 When using the HBM, it is important to consider patients’ perceived barriers to and incentives for engaging in specific behaviors. The same study used the BMQ to address specific questions about respondents’ beliefs and feelings about their prescribed medications and medication adherence. The HBM suggests considering action cues that encourage patients to act by reminding them of the need to change their behavior.

 

Limitations

This study had several limitations. First, the sample size was small (47), which makes it difficult to generalize findings to a broader population of men with a hypertension diagnosis. In addition, because the study was underpowered, its ability to detect significant differences was compromised. Second, the study used a convenience sample of predominantly African American men. As always, there are concerns of self-selection and failure to represent the overall population. Third, the setting for completing the surveys was only semiprivate, and some respondents may have been uncomfortable, perhaps, working too quickly and not really thinking about the questions or their answers. Fourth, the ED survey was self-administered, so there is a concern about the truthfulness of responses. Fifth, failure to ask respondents whether they were taking a phosphodiesterase 5 inhibitor for ED could have significantly impacted study findings. Sixth, respondents were not asked about other medications, such as antidepressants and nonsteroidal anti-inflammatory drugs, which could have affected erectile function.

Clinical Implications

Despite the study limitations, several findings have important clinical implications. First, the vast majority of participants in this pilot study self-reported moderate or severe sexual dysfunction on all 5 IIEF domains. This finding is important because this was a convenience sample, and many of the IIEF statements are personal. The high rate of reported sexual dysfunction suggests that the incidence of ED may be underreported in the larger population. Second, mean BMQ scores were similar to those reported in other studies involving chronic illness: higher necessity and lower concern. Third, there was no statistically significant relationship between BMQ necessity and concern and IIEF sexual functioning. More research is needed to determine how to interpret these findings. Fourth, there was a significant relationship between length of time with hypertension diagnosis and BMQ specific-necessity score: The longer the diagnosis, the higher the score. However, this relationship did not hold for BMQ specific-concern, though it trended toward significance. Moreover, length of time with hypertension diagnosis did not necessarily predict or influence erectile function as measured with the IIEF. In fact, men with a hypertension diagnosis of ≥ 11 years reported less moderate-to-severe sexual dysfunction in overall satisfaction and sexual desire. Although there are several methodologic concerns about this study, its results offer direction for both clinical practice and future research.

Studies of erectile function and its relationship to hypertension have generated both cause for concern and reason for further research. The present study focused on gaining a better understanding of the relationship between antihypertensive medication beliefs and erectile function. Future clinical studies should explore the effects of antihypertensive medication on erectile function and men’s lack of knowledge and education about the importance of taking medication to prevent complications of hypertension. It is essential that this research be applied to improve the understanding of erectile function in men with hypertension. This will ultimately allow for better patient management and contribute to the overall sexual health and well-being of patients with hypertension.

Although it is important to identify men’s antihypertensive medication beliefs and the relationship of these beliefs to sexual satisfaction, most longitudinal studies suggest that the ED rate is high and that it increases with age.10 Therefore, it is crucial that men differentiate between how antihypertensive medications affect erectile function and changes associated with aging. The present study found no statistically significant relationships between the BMQ specific-necessity and specific-concern subscales and overall satisfaction with sexual functioning (IIEF). In addition, the study found no significant correlations between the BMQ specific and general scales and the 5 IIEF domains.

Conclusion

It was evident from this research that men with chronic health problems, such as hypertension, are often prescribed medications that affect sexual functioning. Unfortunately, the effect on sexual functioning often plays a significant role in the discontinuation of long-term therapy. Many of this study’s participants self-reported moderate or severe sexual dysfunction. Results showed no statistically significant relationships between either BMQ subscale or any of the 5 IIEF domains. Research is needed to further explore the association between ED and antihypertensive medication and men’s lack of knowledge and education about the importance of treatment adherence.

References

1. Hochbaum GM; U.S. Public Health Service, Division of Special Health Services. Public Participation in Medical Screening Programs: A Socio-Psychological Study. Public Health Service publication, no. 572 Washington, DC: U.S. Dept. of Health, Education, and Welfare, Public Health Service, Bureau of State Services, Division of Special Health Services, Tuberculosis Program; 1958.

2. International Index of Erectile Function (IIEF). Weill Cornell Medical College James Buchanan Brady Foundation Department of Urology website. https://www.cornellurology.com/images/uploads/IBPH-IIEF-Assessments.pdf. Accessed May 14, 2016.

3. Porteous T, Francis J, Bond C, Hannaford P. Temporal stability of beliefs about medicines: implications for optimizing adherence. Patient Educ Couns. 2010; 79(2):225-230.

4. Horne R, Weinman J, Hankins M. The beliefs about medicines questionnaire: the development and evaluation of a new method for assessing the cognitive representation of medication. Psychol Health. 1999;14(1):1-24.

5. Fogari R, Zoppi A, Corradi L, Mugellini A, Poletti L, Lusardi P. Sexual function in hypertensive males treated with lisinopril or atenolol: a cross-over study. Am J Hypertens. 1998;11(10):1244-1247.

6. Hellstrom WJ, Gittelman M, Karlin G, et al. Vardenafil for treatment of men with erectile dysfunction: efficacy and safety in a randomized, double-blind, placebo-controlled trial. J Androl. 2002;23(6):763-771.

7. Neame R, Hammond A. Beliefs about medications: a questionnaire survey of people with rheumatoid arthritis. Rheumatology (Oxford). 2005;44(6):762-767.

8. Lennerling A, Forsberg A. Self-reported non-adherence and beliefs about medication in a Swedish kidney transplant population. Open Nurs J. 2012;6:41-46.

9. Kressin NR, Wang F, Long J, et al. Hypertensive patients' race, health beliefs, process of care, and medication adherence. J Gen Intern Med. 2007; 22(6):768-774.

10. Inman BA, Sauver JL, Jacobson DJ, et al. A population-based, longitudinal study of erectile dysfunction and future coronary artery disease. Mayo Clin Proc. 2009;84(2):108-113.

References

1. Hochbaum GM; U.S. Public Health Service, Division of Special Health Services. Public Participation in Medical Screening Programs: A Socio-Psychological Study. Public Health Service publication, no. 572 Washington, DC: U.S. Dept. of Health, Education, and Welfare, Public Health Service, Bureau of State Services, Division of Special Health Services, Tuberculosis Program; 1958.

2. International Index of Erectile Function (IIEF). Weill Cornell Medical College James Buchanan Brady Foundation Department of Urology website. https://www.cornellurology.com/images/uploads/IBPH-IIEF-Assessments.pdf. Accessed May 14, 2016.

3. Porteous T, Francis J, Bond C, Hannaford P. Temporal stability of beliefs about medicines: implications for optimizing adherence. Patient Educ Couns. 2010; 79(2):225-230.

4. Horne R, Weinman J, Hankins M. The beliefs about medicines questionnaire: the development and evaluation of a new method for assessing the cognitive representation of medication. Psychol Health. 1999;14(1):1-24.

5. Fogari R, Zoppi A, Corradi L, Mugellini A, Poletti L, Lusardi P. Sexual function in hypertensive males treated with lisinopril or atenolol: a cross-over study. Am J Hypertens. 1998;11(10):1244-1247.

6. Hellstrom WJ, Gittelman M, Karlin G, et al. Vardenafil for treatment of men with erectile dysfunction: efficacy and safety in a randomized, double-blind, placebo-controlled trial. J Androl. 2002;23(6):763-771.

7. Neame R, Hammond A. Beliefs about medications: a questionnaire survey of people with rheumatoid arthritis. Rheumatology (Oxford). 2005;44(6):762-767.

8. Lennerling A, Forsberg A. Self-reported non-adherence and beliefs about medication in a Swedish kidney transplant population. Open Nurs J. 2012;6:41-46.

9. Kressin NR, Wang F, Long J, et al. Hypertensive patients' race, health beliefs, process of care, and medication adherence. J Gen Intern Med. 2007; 22(6):768-774.

10. Inman BA, Sauver JL, Jacobson DJ, et al. A population-based, longitudinal study of erectile dysfunction and future coronary artery disease. Mayo Clin Proc. 2009;84(2):108-113.

Issue
Federal Practitioner - 33(6)
Issue
Federal Practitioner - 33(6)
Page Number
36-41
Page Number
36-41
Publications
Publications
Topics
Article Type
Display Headline
The Relationship Between Male Patients’ Antihypertensive Medication Beliefs and Erectile Function
Display Headline
The Relationship Between Male Patients’ Antihypertensive Medication Beliefs and Erectile Function
Legacy Keywords
adherence, hypertension
Legacy Keywords
adherence, hypertension
Sections
Disallow All Ads
Alternative CME
Use ProPublica
Article PDF Media