Advance Care Planning: Making It Easier for Patients (and You)

Article Type
Changed
Wed, 09/13/2017 - 09:58
Display Headline
Advance Care Planning: Making It Easier for Patients (and You)

 

With the number of aging Americans projected to grow dramatically in the next several years, the need for primary palliative care and advance care planning (ACP) is more important than ever. Patients and their families want and expect palliative care when needed, but initial conversations about ACP can be difficult for them. Appropriate timing in raising this subject and clear communication can give patients the opportunity, while they are still independent, to set their goals for medical care.

For the past several decades, political decisions and judicial cases have shaped palliative care as we know it today. And its shape is still evolving. In support of ACP, advocacy groups at a national level are developing models that practitioners can use to engage patients in setting goals. And Medicare is now reimbursing primary care providers for this work that they have been doing for years (although many still may not be billing for the service).

Finally, the busy primary care office may have its own set of challenges in addressing ACP. Our aim in this review is to identify the barriers we face and the solutions we can implement to make a difference in our patients’ end-of-life care planning.

LANDMARK EVENTS HAVE DEFINED ACP TODAY

In 1969, Luis Kutner, an Illinois attorney, proposed the idea of a “living will,” envisioned as a document specifying the types of treatment a person would be willing to receive were he or she unable at a later time to participate in making a decision.1 In 1976, California became the first state to give living wills the power of the law through the Natural Death Act.2

Throughout the 1970s and ‘80s, several high-profile court cases brought this idea into the national spotlight. In 1975, the New Jersey Supreme Court granted the parents of 21-year-old Karen Ann Quinlan the right to discontinue the treatment sustaining her in a persistent vegetative state. Ms. Quinlan was removed from the ventilator and lived nine more months before dying in a nursing home.

In 1983, age 25, Nancy Cruzan was involved in a motor vehicle accident that left her in a persistent vegetative state. She remained so until 1988, when her parents asked that her feeding tube be removed. The hospital refused, indicating that it would lead to her death. The family sued, and the case eventually went to the US Supreme Court in 1989.

In a 5-to-4 decision, the Supreme Court ruled that a state was legally able to require “clear and convincing evidence” of a patient’s wish for removal of life-sustaining therapies. Cruzan’s family was able to provide such evidence, and her artificial nutrition was withheld. She died 12 days later.

The Cruzan case was instrumental in furthering ACP, leading to the passage of the Patient Self Determination Act by ­Congress in 1990. All federally funded health care facilities were now required to educate ­patients of their rights in determining their medical care and to ask about advance directives.3 The ACP movement gained ad­ditional momentum from the landmark SUPPORT study that documented short­comings in communication between phys­icians and patients/families about treat­ment preferences and end-of-life care in US hospitals.4

In the Terri Schiavo case, the patient’s husband disagreed with the life-sustaining decisions of his wife’s parents, given her persistent vegetative state and the fact that she had no chance of meaningful recovery. After a prolonged national debate, it was ultimately decided that the husband could elect to withhold artificial nutrition. (She died in 2005.) The Schiavo case, as well as the Institute of Medicine’s report on Dying in America, influenced Congress in 2016 to pass legislation funding ACP conversations.5

THE DEMONSTRATED BENEFITS OF ACP

When done comprehensively, ACP yields many benefits for patients and families and for the health care system. A systematic review demonstrated that, despite the few studies examining the economic cost of ACP, the process may lead to decreased health care costs in certain populations (nursing home residents, community-dwelling adults with dementia, and those living in high health care–spending regions) and at the very least does not increase health care costs.6 ACP has increased the number of do-not-resuscitate orders and has decreased hospitalizations, admissions to intensive care units, and rates of cardiopulmonary resuscitation, mechanical ventilation, and use of tube feeding.6-8

More noteworthy than the decrease in resource utilization and potential cost savings is the impact that ACP can have on a patient’s quality of life. Patients who receive aggressive care at the end of life tend to experience decreased quality of life compared with those receiving hospice care.7 Quality-of-life scores for patients in hospice improved with the length of enrollment in that care.7 When ACP discussions have taken place, the care patients receive at the end of life tends to conform more closely to their wishes and to increase family satis­faction.9-11

One reason that practitioners often give for not completing ACP is the fear of increasing patient or family anxiety or post-traumatic stress disorder (PTSD). However, studies have shown this concern to be unfounded.7,12 While ACP studies have not shown a decrease in rates of anxiety or PTSD, no study has shown an increase in these psychologic morbidities.8

Caveats to keep in mind. Not all studies have shown unambiguous benefits related to ACP. Among the systematic reviews previously noted, there was significant variability in quality of data. Additionally, some experts argue that the traditional view of ACP (ie, completion of a single advance directive/living will) is outdated and should be replaced with a method that prepares patients and families to anticipate “in-the-moment decision making.”13 While we still believe that completion of an advance directive is useful, the experts’ point is well taken, especially since many patients change their preferences over time (and typically toward more aggressive care).14,15 While the advance directive serves a role, it is more important to help patients recognize their goals and preferences and to facilitate ongoing discussions between the patient and his or her family/surrogate decision-maker and providers.

 

 

 

A SNAPSHOT OF PARTICIPATION IN ACP

Despite the ACP movement and the likely benefits associated with it, most individuals have not participated. Rates of completion do seem to be rising, but there is still room for improvement. Among all individuals older than 18, only 26.3% have an advance directive.16 In a cohort of older patients seen in an emergency department, only 40% had a living will, while nearly 54% had a designated health care power of attorney.17 Perhaps more alarming is the lack of ACP for those patients almost all providers would agree need it: the long-term care population. The National Center for Health Statistics has reported that only 28% of home health care patients, 65% of nursing home residents, and 88% of hospice patients have an advance directive on file.18

PROVIDER AND PATIENT BARRIERS TO ACP

If ACP can decrease resource utilization and improve caregiver compliance with a patient’s wishes for end of life, the obvious question is: Why isn’t it done more often? A longstanding barrier for providers has been that these types of discussions are time intensive and have not been billable. However, since January 1, 2016, we are now able to bill for these discussions. (More on this in a bit.) Providers do cite other barriers, though.

A recent systematic review showed that ACP is hindered by time constraints imposed by other clinical and administrative tasks that are heavily monitored.19 Barriers to engaging in ACP reported by patients include a reluctance to think about dying, a belief that family or providers will know what to do, difficulty understanding ACP forms, and the absence of a person who can serve as a surrogate decision-maker.20,21

NATIONAL MODELS TO HELP WITH IMPLEMENTATION

The percentage of individuals with an advance directive in the US has not increased significantly over the past decade.22 The lack of traction in completion and use of advance directives has led several authors to question the utility of this older model of ACP.22 Several experts in the field believe that more robust, ongoing goals-of-care conversations between patients, families, and providers are equally, or even more, important than the completion of actual advance directive documents.23,24

National models such as the POLST (Physician Orders for Life-Sustaining Treatment) paradigm have become popular in several states (www.polst.org). Intended for those with estimated life expectancy of less than one year, POLST is not an advance directive but a physician order for these seriously ill patients. Emergency medical service workers are legally able to follow a POLST document but not a living will or advance directive—a significant reason for those with end-stage illness to consider completing a POLST document with their health care provider. Programs such as “Respecting Choices” have incorporated POLST documentation as part of ongoing goals-of-care conversations between patients and health care providers (www.gundersenhealth.org/respecting-choices).

Many groups have developed products to encourage patients and their families to initiate conversations at home. An example is the Conversation Project, a free online resource available in multiple languages that can help break the ice for patients and get them talking about their wishes for end-of-life care (www.theconversationproject.org). It poses simple stimulating questions such as “What kind of role do you want to have in the decision-making process?” and “What are your concerns about treatment?”

HOW-TO TIPS FOR ACP IN OUTPATIENT SETTINGS

When approaching the topic of ACP with patients, it’s important to do so over time, starting as soon as possible with older patients and those with chronic illness that confers a high risk for significant morbidity or mortality. Assess each patient’s understanding of ACP and readiness to discuss the topic. Many patients think of ACP in the context of a document (eg, living will), so asking about the existence of a living will may help to start the conversation. Alternatively, consider inquiring about whether the patient has had experience with family or friends at the end of life or during a difficult medical situation, and whether the patient has thought about making personal plans for such a situation.25

When a patient is ready to have this conversation, your goal should be three-fold:26

  1. Help the patient articulate personal values, goals, and preferences.
  2. Ask the patient to formally assign health care power of attorney (POA) to a trusted individual or to name a surrogate decision-maker. Document this decision in the medical record.
  3. Help the patient translate expressed values into specific medical care plans, if applicable.

Because ACP conversations are often time consuming, it’s a good idea to schedule separate appointments to focus on this alone. If, however, a patient is unable to return for a dedicated ACP visit, a first step that can be completed in a reasonably short period would be choosing a surrogate decision-maker.

Helping a patient articulate personal values may be eased by asking such questions as, “Have you ever thought about what kind of care you would want if the time came when you could not make your own decisions?” or “What worries you the most about possibly not being able to make your own decisions?”27 If the patient is able to identify a surrogate decision-maker before the ACP appointment, ask that this person attend. A family member or close friend may remember instances in which the patient expressed health care preferences, and their presence can help to minimize gaps in communication.

Once the patient’s preferences are clear, document them in the medical record. Some preferences may be suitable for translation into POLST orders or an advance directive, but this is less important than the overall discussion. ACP should be an ongoing conversation, since a patient’s goals may change over time. And encourage the patient to share any desired change in plans with their surrogate decision-maker or update the POA document.

 

 

 

BE SURE TO BILL FOR ACP SERVICES

To encourage office-based providers to conduct ACP, the Centers for Medicare and Medicaid Services (CMS) implemented payment for CPT codes 99497 and 99498.

CPT code 99497 covers the first 30 minutes of face-to-face time with patients or their family members or medical decision-makers. This time can be used to discuss living wills or advance directives.

CPT code 99498 can be applied to each additional 30 minutes of ACP services. Typically, this billing code would be used as an add-on for a particular diagnosis, such as heart failure, chronic obstructive pulmonary disease, or pancreatic cancer.

CPT Code 99497 equates to 2.40 relative-value units (RVU) with an estimated payment of $85.99, while CPT code 99498 equates to 2.09 RVU with an estimated payment of $74.88.28

According to CMS, there is no annual limit to the number of times the ACP codes can be billed for a particular patient. And there are no restrictions regarding location of service, meaning a provider could perform this in an outpatient setting, an inpatient setting, or a long-term care facility. All health care providers are allowed to bill with this code. Also worth noting: You don’t need to complete any particular documentation for a visit to be billed as an ACP service. CMS provides a helpful Q & A at www.cms.gov/Medicare/Medicare-Fee-for-Service-Payment/PhysicianFeeSched/Downloads/FAQ-Advance-Care-Planning.pdf.

References

1. Kutner L. Due process of euthanasia: the living will, a proposal. Indiana Law J. 1969;44:539-554.
2. California Law Revision Commission. 2000 Health Care Decisions Law and Revised Power of Attorney Law. www.clrc.ca.gov/pub/Printed-Reports/Pub208.pdf. Accessed August 14, 2017.
3. H.R. 5067 - 101st Congress. Patient Self Determination Act of 1990. www.govtrack.us/congress/bills/101/hr5067. Accessed August 14, 2017.
4. The SUPPORT Principle Investigators. A controlled trial to improve care for seriously ill hospitalized patients. The Study to Understand Prognoses and Preferences for Outcomes and Risks of Treatments (SUPPORT). JAMA. 1995;274:1591-1598.
5. Institute of Medicine. Dying in America: Improving Quality and Honoring Individual Preferences Near the End of Life. Washington, DC: National Academies Press; 2015.
6. Dixon J, Matosevic T, Knapp M. The economic evidence for advance care planning: systematic review of evidence. Palliat Med. 2015;29:869-884.
7. Wright AA, Ray A, Mack JW, et al. Associations between end-of-life discussions, patient mental-health, medical care near death, and caregiver bereavement adjustment. JAMA. 2008;300:1665-1673.
8. Brinkman-Stoppelenburg A, Rietjens JAC, van der Heide A. The effects of advance care planning on end-of-life care: a systematic review. Palliat Med. 2014;28:1000-1025.
9. Detering KM, Hancock AD, Reade MC, et al. The impact of advance care planning on end of life care in elderly patients: randomised controlled trial. BMJ. 2010;340:c1345.
10. Morrison RS, Chichin E, Carter J, et al. The effect of a social work intervention to enhance advance care planning documentation in the nursing home. J Am Geriatr Soc. 2005;53:290-294.
11. Schamp R, Tenkku L. Managed death in a PACE: pathways in present and advance directives. J Am Med Dir Assoc. 2006;7:339-344.
12. Walczak A, Butow PN, Bu S, et al. A systematic review of evidence for end-of-life communication interventions: who do they target, how are they structured and do they work? Patient Educ Couns. 2016;99:3-16.
13. Sudore RL, Fried TR. Redefining the “planning” in advance care planning: preparing for end-of-life decision making. Ann Intern Med. 2010;153:256-261.
14. Straton JB, Wang NY, Meoni LA, et al. Physical functioning, depression, and preferences for treatment at the end of life: the Johns Hopkins Precursors study. J Am Geriatr Soc. 2004;52:577-582.
15. Fried TR, Byers AL, Gallo WT, et al. Prospective study of health status preferences and changes in preferences over time in older adults. Arch Intern Med. 2006;166:890-895.
16. Rao JK, Anderson LA, Lin F, et al. Completion of advance directives among U.S. consumers. Am J Prev Med. 2014;46:65-70.
17. Grudzen CR, Buonocore P, Steinberg J, et al; AAHPM Re­search Committee Writing Group. Concordance of advance care plans with inpatient directives in the electronic medical record for older patients admitted from the emergency department. J Pain Symptom Manage. 2016;51:647-651.
18. Jones AL, Moss AJ, Harris-Kojetin LD. Use of advance directives in long-term care populations. NCHS Data Brief. 2011;(54):1-8.
19. Lund S, Richardson A, May C. Barriers to advance care planning at the end of life: an explanatory systematic review of implementation studies. PLoS One. 2015;10:e0116629.
20. Fried TR, Bullock K, Iannone L, et al. Understanding advance care planning as a process of health behavior change. J Am Geriatr Soc. 2009;57:1547-1555.
21. Schickedanz AD, Schillinger D, Landefeld CS, et al. A clinical framework for improving the advance care planning process: start with patients’ self-identified barriers. J Am Geriatr Soc. 2009;57:31-39.
22. Winter L, Parks SM, Diamond JJ. Ask a different question, get a different answer: why living wills are poor guides to care preferences at the end of life. J Palliat Med. 2010;13:567-572.
23. Institute of Medicine. Dying in America: Improving Quality and Honoring Individual Preferences Near the End of Life. www.nap.edu/read/18748/chapter/1. Accessed August 14, 2017.
24. Sudore RL, Schickedanz AD, Landefeld CS, et al. Engagement in multiple steps of the advance care planning process: a descriptive study of diverse older adults. J Am Geriatr Soc. 2008;56:1006-1013.
25. McMahan RD, Knight SJ, Fried TR, et al. Advance care planning beyond advance directives: perspectives from patients and surrogates. J Pain Symptom Manage. 2013;46:355-365.
26. Lum HD, Sudore RL, Bekelman DB. Advance care planning in the elderly. Med Clin North Am. 2015;99:391-403.
27. Lum HD, Sudore RL. Advance care planning and goals of care communication in older adults with cardiovascular disease and multi-morbidity. Clin Geriatr Med. 2016;32:247-260.
28. American College of Physicians. Advanced Care Planning: Implementation for practices. www.acponline.org/system/files/documents/practice-resources/business-resources/payment/advance_care_planning_toolkit.pdf. Accessed August 14, 2017.

Article PDF
Author and Disclosure Information

John Liantonio, Michael Liquori, Lilia Lakhtman, and Susan Parks practice at Thomas Jefferson University Hospital in Philadelphia.

The authors reported no potential conflict of interestrelevant to this article, which was originally published in The Journal of Family Practice (2017;66[8]:487-491).

Issue
Clinician Reviews - 27(9)
Publications
Topics
Page Number
47-51
Sections
Author and Disclosure Information

John Liantonio, Michael Liquori, Lilia Lakhtman, and Susan Parks practice at Thomas Jefferson University Hospital in Philadelphia.

The authors reported no potential conflict of interestrelevant to this article, which was originally published in The Journal of Family Practice (2017;66[8]:487-491).

Author and Disclosure Information

John Liantonio, Michael Liquori, Lilia Lakhtman, and Susan Parks practice at Thomas Jefferson University Hospital in Philadelphia.

The authors reported no potential conflict of interestrelevant to this article, which was originally published in The Journal of Family Practice (2017;66[8]:487-491).

Article PDF
Article PDF
Related Articles

 

With the number of aging Americans projected to grow dramatically in the next several years, the need for primary palliative care and advance care planning (ACP) is more important than ever. Patients and their families want and expect palliative care when needed, but initial conversations about ACP can be difficult for them. Appropriate timing in raising this subject and clear communication can give patients the opportunity, while they are still independent, to set their goals for medical care.

For the past several decades, political decisions and judicial cases have shaped palliative care as we know it today. And its shape is still evolving. In support of ACP, advocacy groups at a national level are developing models that practitioners can use to engage patients in setting goals. And Medicare is now reimbursing primary care providers for this work that they have been doing for years (although many still may not be billing for the service).

Finally, the busy primary care office may have its own set of challenges in addressing ACP. Our aim in this review is to identify the barriers we face and the solutions we can implement to make a difference in our patients’ end-of-life care planning.

LANDMARK EVENTS HAVE DEFINED ACP TODAY

In 1969, Luis Kutner, an Illinois attorney, proposed the idea of a “living will,” envisioned as a document specifying the types of treatment a person would be willing to receive were he or she unable at a later time to participate in making a decision.1 In 1976, California became the first state to give living wills the power of the law through the Natural Death Act.2

Throughout the 1970s and ‘80s, several high-profile court cases brought this idea into the national spotlight. In 1975, the New Jersey Supreme Court granted the parents of 21-year-old Karen Ann Quinlan the right to discontinue the treatment sustaining her in a persistent vegetative state. Ms. Quinlan was removed from the ventilator and lived nine more months before dying in a nursing home.

In 1983, age 25, Nancy Cruzan was involved in a motor vehicle accident that left her in a persistent vegetative state. She remained so until 1988, when her parents asked that her feeding tube be removed. The hospital refused, indicating that it would lead to her death. The family sued, and the case eventually went to the US Supreme Court in 1989.

In a 5-to-4 decision, the Supreme Court ruled that a state was legally able to require “clear and convincing evidence” of a patient’s wish for removal of life-sustaining therapies. Cruzan’s family was able to provide such evidence, and her artificial nutrition was withheld. She died 12 days later.

The Cruzan case was instrumental in furthering ACP, leading to the passage of the Patient Self Determination Act by ­Congress in 1990. All federally funded health care facilities were now required to educate ­patients of their rights in determining their medical care and to ask about advance directives.3 The ACP movement gained ad­ditional momentum from the landmark SUPPORT study that documented short­comings in communication between phys­icians and patients/families about treat­ment preferences and end-of-life care in US hospitals.4

In the Terri Schiavo case, the patient’s husband disagreed with the life-sustaining decisions of his wife’s parents, given her persistent vegetative state and the fact that she had no chance of meaningful recovery. After a prolonged national debate, it was ultimately decided that the husband could elect to withhold artificial nutrition. (She died in 2005.) The Schiavo case, as well as the Institute of Medicine’s report on Dying in America, influenced Congress in 2016 to pass legislation funding ACP conversations.5

THE DEMONSTRATED BENEFITS OF ACP

When done comprehensively, ACP yields many benefits for patients and families and for the health care system. A systematic review demonstrated that, despite the few studies examining the economic cost of ACP, the process may lead to decreased health care costs in certain populations (nursing home residents, community-dwelling adults with dementia, and those living in high health care–spending regions) and at the very least does not increase health care costs.6 ACP has increased the number of do-not-resuscitate orders and has decreased hospitalizations, admissions to intensive care units, and rates of cardiopulmonary resuscitation, mechanical ventilation, and use of tube feeding.6-8

More noteworthy than the decrease in resource utilization and potential cost savings is the impact that ACP can have on a patient’s quality of life. Patients who receive aggressive care at the end of life tend to experience decreased quality of life compared with those receiving hospice care.7 Quality-of-life scores for patients in hospice improved with the length of enrollment in that care.7 When ACP discussions have taken place, the care patients receive at the end of life tends to conform more closely to their wishes and to increase family satis­faction.9-11

One reason that practitioners often give for not completing ACP is the fear of increasing patient or family anxiety or post-traumatic stress disorder (PTSD). However, studies have shown this concern to be unfounded.7,12 While ACP studies have not shown a decrease in rates of anxiety or PTSD, no study has shown an increase in these psychologic morbidities.8

Caveats to keep in mind. Not all studies have shown unambiguous benefits related to ACP. Among the systematic reviews previously noted, there was significant variability in quality of data. Additionally, some experts argue that the traditional view of ACP (ie, completion of a single advance directive/living will) is outdated and should be replaced with a method that prepares patients and families to anticipate “in-the-moment decision making.”13 While we still believe that completion of an advance directive is useful, the experts’ point is well taken, especially since many patients change their preferences over time (and typically toward more aggressive care).14,15 While the advance directive serves a role, it is more important to help patients recognize their goals and preferences and to facilitate ongoing discussions between the patient and his or her family/surrogate decision-maker and providers.

 

 

 

A SNAPSHOT OF PARTICIPATION IN ACP

Despite the ACP movement and the likely benefits associated with it, most individuals have not participated. Rates of completion do seem to be rising, but there is still room for improvement. Among all individuals older than 18, only 26.3% have an advance directive.16 In a cohort of older patients seen in an emergency department, only 40% had a living will, while nearly 54% had a designated health care power of attorney.17 Perhaps more alarming is the lack of ACP for those patients almost all providers would agree need it: the long-term care population. The National Center for Health Statistics has reported that only 28% of home health care patients, 65% of nursing home residents, and 88% of hospice patients have an advance directive on file.18

PROVIDER AND PATIENT BARRIERS TO ACP

If ACP can decrease resource utilization and improve caregiver compliance with a patient’s wishes for end of life, the obvious question is: Why isn’t it done more often? A longstanding barrier for providers has been that these types of discussions are time intensive and have not been billable. However, since January 1, 2016, we are now able to bill for these discussions. (More on this in a bit.) Providers do cite other barriers, though.

A recent systematic review showed that ACP is hindered by time constraints imposed by other clinical and administrative tasks that are heavily monitored.19 Barriers to engaging in ACP reported by patients include a reluctance to think about dying, a belief that family or providers will know what to do, difficulty understanding ACP forms, and the absence of a person who can serve as a surrogate decision-maker.20,21

NATIONAL MODELS TO HELP WITH IMPLEMENTATION

The percentage of individuals with an advance directive in the US has not increased significantly over the past decade.22 The lack of traction in completion and use of advance directives has led several authors to question the utility of this older model of ACP.22 Several experts in the field believe that more robust, ongoing goals-of-care conversations between patients, families, and providers are equally, or even more, important than the completion of actual advance directive documents.23,24

National models such as the POLST (Physician Orders for Life-Sustaining Treatment) paradigm have become popular in several states (www.polst.org). Intended for those with estimated life expectancy of less than one year, POLST is not an advance directive but a physician order for these seriously ill patients. Emergency medical service workers are legally able to follow a POLST document but not a living will or advance directive—a significant reason for those with end-stage illness to consider completing a POLST document with their health care provider. Programs such as “Respecting Choices” have incorporated POLST documentation as part of ongoing goals-of-care conversations between patients and health care providers (www.gundersenhealth.org/respecting-choices).

Many groups have developed products to encourage patients and their families to initiate conversations at home. An example is the Conversation Project, a free online resource available in multiple languages that can help break the ice for patients and get them talking about their wishes for end-of-life care (www.theconversationproject.org). It poses simple stimulating questions such as “What kind of role do you want to have in the decision-making process?” and “What are your concerns about treatment?”

HOW-TO TIPS FOR ACP IN OUTPATIENT SETTINGS

When approaching the topic of ACP with patients, it’s important to do so over time, starting as soon as possible with older patients and those with chronic illness that confers a high risk for significant morbidity or mortality. Assess each patient’s understanding of ACP and readiness to discuss the topic. Many patients think of ACP in the context of a document (eg, living will), so asking about the existence of a living will may help to start the conversation. Alternatively, consider inquiring about whether the patient has had experience with family or friends at the end of life or during a difficult medical situation, and whether the patient has thought about making personal plans for such a situation.25

When a patient is ready to have this conversation, your goal should be three-fold:26

  1. Help the patient articulate personal values, goals, and preferences.
  2. Ask the patient to formally assign health care power of attorney (POA) to a trusted individual or to name a surrogate decision-maker. Document this decision in the medical record.
  3. Help the patient translate expressed values into specific medical care plans, if applicable.

Because ACP conversations are often time consuming, it’s a good idea to schedule separate appointments to focus on this alone. If, however, a patient is unable to return for a dedicated ACP visit, a first step that can be completed in a reasonably short period would be choosing a surrogate decision-maker.

Helping a patient articulate personal values may be eased by asking such questions as, “Have you ever thought about what kind of care you would want if the time came when you could not make your own decisions?” or “What worries you the most about possibly not being able to make your own decisions?”27 If the patient is able to identify a surrogate decision-maker before the ACP appointment, ask that this person attend. A family member or close friend may remember instances in which the patient expressed health care preferences, and their presence can help to minimize gaps in communication.

Once the patient’s preferences are clear, document them in the medical record. Some preferences may be suitable for translation into POLST orders or an advance directive, but this is less important than the overall discussion. ACP should be an ongoing conversation, since a patient’s goals may change over time. And encourage the patient to share any desired change in plans with their surrogate decision-maker or update the POA document.

 

 

 

BE SURE TO BILL FOR ACP SERVICES

To encourage office-based providers to conduct ACP, the Centers for Medicare and Medicaid Services (CMS) implemented payment for CPT codes 99497 and 99498.

CPT code 99497 covers the first 30 minutes of face-to-face time with patients or their family members or medical decision-makers. This time can be used to discuss living wills or advance directives.

CPT code 99498 can be applied to each additional 30 minutes of ACP services. Typically, this billing code would be used as an add-on for a particular diagnosis, such as heart failure, chronic obstructive pulmonary disease, or pancreatic cancer.

CPT Code 99497 equates to 2.40 relative-value units (RVU) with an estimated payment of $85.99, while CPT code 99498 equates to 2.09 RVU with an estimated payment of $74.88.28

According to CMS, there is no annual limit to the number of times the ACP codes can be billed for a particular patient. And there are no restrictions regarding location of service, meaning a provider could perform this in an outpatient setting, an inpatient setting, or a long-term care facility. All health care providers are allowed to bill with this code. Also worth noting: You don’t need to complete any particular documentation for a visit to be billed as an ACP service. CMS provides a helpful Q & A at www.cms.gov/Medicare/Medicare-Fee-for-Service-Payment/PhysicianFeeSched/Downloads/FAQ-Advance-Care-Planning.pdf.

 

With the number of aging Americans projected to grow dramatically in the next several years, the need for primary palliative care and advance care planning (ACP) is more important than ever. Patients and their families want and expect palliative care when needed, but initial conversations about ACP can be difficult for them. Appropriate timing in raising this subject and clear communication can give patients the opportunity, while they are still independent, to set their goals for medical care.

For the past several decades, political decisions and judicial cases have shaped palliative care as we know it today. And its shape is still evolving. In support of ACP, advocacy groups at a national level are developing models that practitioners can use to engage patients in setting goals. And Medicare is now reimbursing primary care providers for this work that they have been doing for years (although many still may not be billing for the service).

Finally, the busy primary care office may have its own set of challenges in addressing ACP. Our aim in this review is to identify the barriers we face and the solutions we can implement to make a difference in our patients’ end-of-life care planning.

LANDMARK EVENTS HAVE DEFINED ACP TODAY

In 1969, Luis Kutner, an Illinois attorney, proposed the idea of a “living will,” envisioned as a document specifying the types of treatment a person would be willing to receive were he or she unable at a later time to participate in making a decision.1 In 1976, California became the first state to give living wills the power of the law through the Natural Death Act.2

Throughout the 1970s and ‘80s, several high-profile court cases brought this idea into the national spotlight. In 1975, the New Jersey Supreme Court granted the parents of 21-year-old Karen Ann Quinlan the right to discontinue the treatment sustaining her in a persistent vegetative state. Ms. Quinlan was removed from the ventilator and lived nine more months before dying in a nursing home.

In 1983, age 25, Nancy Cruzan was involved in a motor vehicle accident that left her in a persistent vegetative state. She remained so until 1988, when her parents asked that her feeding tube be removed. The hospital refused, indicating that it would lead to her death. The family sued, and the case eventually went to the US Supreme Court in 1989.

In a 5-to-4 decision, the Supreme Court ruled that a state was legally able to require “clear and convincing evidence” of a patient’s wish for removal of life-sustaining therapies. Cruzan’s family was able to provide such evidence, and her artificial nutrition was withheld. She died 12 days later.

The Cruzan case was instrumental in furthering ACP, leading to the passage of the Patient Self Determination Act by ­Congress in 1990. All federally funded health care facilities were now required to educate ­patients of their rights in determining their medical care and to ask about advance directives.3 The ACP movement gained ad­ditional momentum from the landmark SUPPORT study that documented short­comings in communication between phys­icians and patients/families about treat­ment preferences and end-of-life care in US hospitals.4

In the Terri Schiavo case, the patient’s husband disagreed with the life-sustaining decisions of his wife’s parents, given her persistent vegetative state and the fact that she had no chance of meaningful recovery. After a prolonged national debate, it was ultimately decided that the husband could elect to withhold artificial nutrition. (She died in 2005.) The Schiavo case, as well as the Institute of Medicine’s report on Dying in America, influenced Congress in 2016 to pass legislation funding ACP conversations.5

THE DEMONSTRATED BENEFITS OF ACP

When done comprehensively, ACP yields many benefits for patients and families and for the health care system. A systematic review demonstrated that, despite the few studies examining the economic cost of ACP, the process may lead to decreased health care costs in certain populations (nursing home residents, community-dwelling adults with dementia, and those living in high health care–spending regions) and at the very least does not increase health care costs.6 ACP has increased the number of do-not-resuscitate orders and has decreased hospitalizations, admissions to intensive care units, and rates of cardiopulmonary resuscitation, mechanical ventilation, and use of tube feeding.6-8

More noteworthy than the decrease in resource utilization and potential cost savings is the impact that ACP can have on a patient’s quality of life. Patients who receive aggressive care at the end of life tend to experience decreased quality of life compared with those receiving hospice care.7 Quality-of-life scores for patients in hospice improved with the length of enrollment in that care.7 When ACP discussions have taken place, the care patients receive at the end of life tends to conform more closely to their wishes and to increase family satis­faction.9-11

One reason that practitioners often give for not completing ACP is the fear of increasing patient or family anxiety or post-traumatic stress disorder (PTSD). However, studies have shown this concern to be unfounded.7,12 While ACP studies have not shown a decrease in rates of anxiety or PTSD, no study has shown an increase in these psychologic morbidities.8

Caveats to keep in mind. Not all studies have shown unambiguous benefits related to ACP. Among the systematic reviews previously noted, there was significant variability in quality of data. Additionally, some experts argue that the traditional view of ACP (ie, completion of a single advance directive/living will) is outdated and should be replaced with a method that prepares patients and families to anticipate “in-the-moment decision making.”13 While we still believe that completion of an advance directive is useful, the experts’ point is well taken, especially since many patients change their preferences over time (and typically toward more aggressive care).14,15 While the advance directive serves a role, it is more important to help patients recognize their goals and preferences and to facilitate ongoing discussions between the patient and his or her family/surrogate decision-maker and providers.

 

 

 

A SNAPSHOT OF PARTICIPATION IN ACP

Despite the ACP movement and the likely benefits associated with it, most individuals have not participated. Rates of completion do seem to be rising, but there is still room for improvement. Among all individuals older than 18, only 26.3% have an advance directive.16 In a cohort of older patients seen in an emergency department, only 40% had a living will, while nearly 54% had a designated health care power of attorney.17 Perhaps more alarming is the lack of ACP for those patients almost all providers would agree need it: the long-term care population. The National Center for Health Statistics has reported that only 28% of home health care patients, 65% of nursing home residents, and 88% of hospice patients have an advance directive on file.18

PROVIDER AND PATIENT BARRIERS TO ACP

If ACP can decrease resource utilization and improve caregiver compliance with a patient’s wishes for end of life, the obvious question is: Why isn’t it done more often? A longstanding barrier for providers has been that these types of discussions are time intensive and have not been billable. However, since January 1, 2016, we are now able to bill for these discussions. (More on this in a bit.) Providers do cite other barriers, though.

A recent systematic review showed that ACP is hindered by time constraints imposed by other clinical and administrative tasks that are heavily monitored.19 Barriers to engaging in ACP reported by patients include a reluctance to think about dying, a belief that family or providers will know what to do, difficulty understanding ACP forms, and the absence of a person who can serve as a surrogate decision-maker.20,21

NATIONAL MODELS TO HELP WITH IMPLEMENTATION

The percentage of individuals with an advance directive in the US has not increased significantly over the past decade.22 The lack of traction in completion and use of advance directives has led several authors to question the utility of this older model of ACP.22 Several experts in the field believe that more robust, ongoing goals-of-care conversations between patients, families, and providers are equally, or even more, important than the completion of actual advance directive documents.23,24

National models such as the POLST (Physician Orders for Life-Sustaining Treatment) paradigm have become popular in several states (www.polst.org). Intended for those with estimated life expectancy of less than one year, POLST is not an advance directive but a physician order for these seriously ill patients. Emergency medical service workers are legally able to follow a POLST document but not a living will or advance directive—a significant reason for those with end-stage illness to consider completing a POLST document with their health care provider. Programs such as “Respecting Choices” have incorporated POLST documentation as part of ongoing goals-of-care conversations between patients and health care providers (www.gundersenhealth.org/respecting-choices).

Many groups have developed products to encourage patients and their families to initiate conversations at home. An example is the Conversation Project, a free online resource available in multiple languages that can help break the ice for patients and get them talking about their wishes for end-of-life care (www.theconversationproject.org). It poses simple stimulating questions such as “What kind of role do you want to have in the decision-making process?” and “What are your concerns about treatment?”

HOW-TO TIPS FOR ACP IN OUTPATIENT SETTINGS

When approaching the topic of ACP with patients, it’s important to do so over time, starting as soon as possible with older patients and those with chronic illness that confers a high risk for significant morbidity or mortality. Assess each patient’s understanding of ACP and readiness to discuss the topic. Many patients think of ACP in the context of a document (eg, living will), so asking about the existence of a living will may help to start the conversation. Alternatively, consider inquiring about whether the patient has had experience with family or friends at the end of life or during a difficult medical situation, and whether the patient has thought about making personal plans for such a situation.25

When a patient is ready to have this conversation, your goal should be three-fold:26

  1. Help the patient articulate personal values, goals, and preferences.
  2. Ask the patient to formally assign health care power of attorney (POA) to a trusted individual or to name a surrogate decision-maker. Document this decision in the medical record.
  3. Help the patient translate expressed values into specific medical care plans, if applicable.

Because ACP conversations are often time consuming, it’s a good idea to schedule separate appointments to focus on this alone. If, however, a patient is unable to return for a dedicated ACP visit, a first step that can be completed in a reasonably short period would be choosing a surrogate decision-maker.

Helping a patient articulate personal values may be eased by asking such questions as, “Have you ever thought about what kind of care you would want if the time came when you could not make your own decisions?” or “What worries you the most about possibly not being able to make your own decisions?”27 If the patient is able to identify a surrogate decision-maker before the ACP appointment, ask that this person attend. A family member or close friend may remember instances in which the patient expressed health care preferences, and their presence can help to minimize gaps in communication.

Once the patient’s preferences are clear, document them in the medical record. Some preferences may be suitable for translation into POLST orders or an advance directive, but this is less important than the overall discussion. ACP should be an ongoing conversation, since a patient’s goals may change over time. And encourage the patient to share any desired change in plans with their surrogate decision-maker or update the POA document.

 

 

 

BE SURE TO BILL FOR ACP SERVICES

To encourage office-based providers to conduct ACP, the Centers for Medicare and Medicaid Services (CMS) implemented payment for CPT codes 99497 and 99498.

CPT code 99497 covers the first 30 minutes of face-to-face time with patients or their family members or medical decision-makers. This time can be used to discuss living wills or advance directives.

CPT code 99498 can be applied to each additional 30 minutes of ACP services. Typically, this billing code would be used as an add-on for a particular diagnosis, such as heart failure, chronic obstructive pulmonary disease, or pancreatic cancer.

CPT Code 99497 equates to 2.40 relative-value units (RVU) with an estimated payment of $85.99, while CPT code 99498 equates to 2.09 RVU with an estimated payment of $74.88.28

According to CMS, there is no annual limit to the number of times the ACP codes can be billed for a particular patient. And there are no restrictions regarding location of service, meaning a provider could perform this in an outpatient setting, an inpatient setting, or a long-term care facility. All health care providers are allowed to bill with this code. Also worth noting: You don’t need to complete any particular documentation for a visit to be billed as an ACP service. CMS provides a helpful Q & A at www.cms.gov/Medicare/Medicare-Fee-for-Service-Payment/PhysicianFeeSched/Downloads/FAQ-Advance-Care-Planning.pdf.

References

1. Kutner L. Due process of euthanasia: the living will, a proposal. Indiana Law J. 1969;44:539-554.
2. California Law Revision Commission. 2000 Health Care Decisions Law and Revised Power of Attorney Law. www.clrc.ca.gov/pub/Printed-Reports/Pub208.pdf. Accessed August 14, 2017.
3. H.R. 5067 - 101st Congress. Patient Self Determination Act of 1990. www.govtrack.us/congress/bills/101/hr5067. Accessed August 14, 2017.
4. The SUPPORT Principle Investigators. A controlled trial to improve care for seriously ill hospitalized patients. The Study to Understand Prognoses and Preferences for Outcomes and Risks of Treatments (SUPPORT). JAMA. 1995;274:1591-1598.
5. Institute of Medicine. Dying in America: Improving Quality and Honoring Individual Preferences Near the End of Life. Washington, DC: National Academies Press; 2015.
6. Dixon J, Matosevic T, Knapp M. The economic evidence for advance care planning: systematic review of evidence. Palliat Med. 2015;29:869-884.
7. Wright AA, Ray A, Mack JW, et al. Associations between end-of-life discussions, patient mental-health, medical care near death, and caregiver bereavement adjustment. JAMA. 2008;300:1665-1673.
8. Brinkman-Stoppelenburg A, Rietjens JAC, van der Heide A. The effects of advance care planning on end-of-life care: a systematic review. Palliat Med. 2014;28:1000-1025.
9. Detering KM, Hancock AD, Reade MC, et al. The impact of advance care planning on end of life care in elderly patients: randomised controlled trial. BMJ. 2010;340:c1345.
10. Morrison RS, Chichin E, Carter J, et al. The effect of a social work intervention to enhance advance care planning documentation in the nursing home. J Am Geriatr Soc. 2005;53:290-294.
11. Schamp R, Tenkku L. Managed death in a PACE: pathways in present and advance directives. J Am Med Dir Assoc. 2006;7:339-344.
12. Walczak A, Butow PN, Bu S, et al. A systematic review of evidence for end-of-life communication interventions: who do they target, how are they structured and do they work? Patient Educ Couns. 2016;99:3-16.
13. Sudore RL, Fried TR. Redefining the “planning” in advance care planning: preparing for end-of-life decision making. Ann Intern Med. 2010;153:256-261.
14. Straton JB, Wang NY, Meoni LA, et al. Physical functioning, depression, and preferences for treatment at the end of life: the Johns Hopkins Precursors study. J Am Geriatr Soc. 2004;52:577-582.
15. Fried TR, Byers AL, Gallo WT, et al. Prospective study of health status preferences and changes in preferences over time in older adults. Arch Intern Med. 2006;166:890-895.
16. Rao JK, Anderson LA, Lin F, et al. Completion of advance directives among U.S. consumers. Am J Prev Med. 2014;46:65-70.
17. Grudzen CR, Buonocore P, Steinberg J, et al; AAHPM Re­search Committee Writing Group. Concordance of advance care plans with inpatient directives in the electronic medical record for older patients admitted from the emergency department. J Pain Symptom Manage. 2016;51:647-651.
18. Jones AL, Moss AJ, Harris-Kojetin LD. Use of advance directives in long-term care populations. NCHS Data Brief. 2011;(54):1-8.
19. Lund S, Richardson A, May C. Barriers to advance care planning at the end of life: an explanatory systematic review of implementation studies. PLoS One. 2015;10:e0116629.
20. Fried TR, Bullock K, Iannone L, et al. Understanding advance care planning as a process of health behavior change. J Am Geriatr Soc. 2009;57:1547-1555.
21. Schickedanz AD, Schillinger D, Landefeld CS, et al. A clinical framework for improving the advance care planning process: start with patients’ self-identified barriers. J Am Geriatr Soc. 2009;57:31-39.
22. Winter L, Parks SM, Diamond JJ. Ask a different question, get a different answer: why living wills are poor guides to care preferences at the end of life. J Palliat Med. 2010;13:567-572.
23. Institute of Medicine. Dying in America: Improving Quality and Honoring Individual Preferences Near the End of Life. www.nap.edu/read/18748/chapter/1. Accessed August 14, 2017.
24. Sudore RL, Schickedanz AD, Landefeld CS, et al. Engagement in multiple steps of the advance care planning process: a descriptive study of diverse older adults. J Am Geriatr Soc. 2008;56:1006-1013.
25. McMahan RD, Knight SJ, Fried TR, et al. Advance care planning beyond advance directives: perspectives from patients and surrogates. J Pain Symptom Manage. 2013;46:355-365.
26. Lum HD, Sudore RL, Bekelman DB. Advance care planning in the elderly. Med Clin North Am. 2015;99:391-403.
27. Lum HD, Sudore RL. Advance care planning and goals of care communication in older adults with cardiovascular disease and multi-morbidity. Clin Geriatr Med. 2016;32:247-260.
28. American College of Physicians. Advanced Care Planning: Implementation for practices. www.acponline.org/system/files/documents/practice-resources/business-resources/payment/advance_care_planning_toolkit.pdf. Accessed August 14, 2017.

References

1. Kutner L. Due process of euthanasia: the living will, a proposal. Indiana Law J. 1969;44:539-554.
2. California Law Revision Commission. 2000 Health Care Decisions Law and Revised Power of Attorney Law. www.clrc.ca.gov/pub/Printed-Reports/Pub208.pdf. Accessed August 14, 2017.
3. H.R. 5067 - 101st Congress. Patient Self Determination Act of 1990. www.govtrack.us/congress/bills/101/hr5067. Accessed August 14, 2017.
4. The SUPPORT Principle Investigators. A controlled trial to improve care for seriously ill hospitalized patients. The Study to Understand Prognoses and Preferences for Outcomes and Risks of Treatments (SUPPORT). JAMA. 1995;274:1591-1598.
5. Institute of Medicine. Dying in America: Improving Quality and Honoring Individual Preferences Near the End of Life. Washington, DC: National Academies Press; 2015.
6. Dixon J, Matosevic T, Knapp M. The economic evidence for advance care planning: systematic review of evidence. Palliat Med. 2015;29:869-884.
7. Wright AA, Ray A, Mack JW, et al. Associations between end-of-life discussions, patient mental-health, medical care near death, and caregiver bereavement adjustment. JAMA. 2008;300:1665-1673.
8. Brinkman-Stoppelenburg A, Rietjens JAC, van der Heide A. The effects of advance care planning on end-of-life care: a systematic review. Palliat Med. 2014;28:1000-1025.
9. Detering KM, Hancock AD, Reade MC, et al. The impact of advance care planning on end of life care in elderly patients: randomised controlled trial. BMJ. 2010;340:c1345.
10. Morrison RS, Chichin E, Carter J, et al. The effect of a social work intervention to enhance advance care planning documentation in the nursing home. J Am Geriatr Soc. 2005;53:290-294.
11. Schamp R, Tenkku L. Managed death in a PACE: pathways in present and advance directives. J Am Med Dir Assoc. 2006;7:339-344.
12. Walczak A, Butow PN, Bu S, et al. A systematic review of evidence for end-of-life communication interventions: who do they target, how are they structured and do they work? Patient Educ Couns. 2016;99:3-16.
13. Sudore RL, Fried TR. Redefining the “planning” in advance care planning: preparing for end-of-life decision making. Ann Intern Med. 2010;153:256-261.
14. Straton JB, Wang NY, Meoni LA, et al. Physical functioning, depression, and preferences for treatment at the end of life: the Johns Hopkins Precursors study. J Am Geriatr Soc. 2004;52:577-582.
15. Fried TR, Byers AL, Gallo WT, et al. Prospective study of health status preferences and changes in preferences over time in older adults. Arch Intern Med. 2006;166:890-895.
16. Rao JK, Anderson LA, Lin F, et al. Completion of advance directives among U.S. consumers. Am J Prev Med. 2014;46:65-70.
17. Grudzen CR, Buonocore P, Steinberg J, et al; AAHPM Re­search Committee Writing Group. Concordance of advance care plans with inpatient directives in the electronic medical record for older patients admitted from the emergency department. J Pain Symptom Manage. 2016;51:647-651.
18. Jones AL, Moss AJ, Harris-Kojetin LD. Use of advance directives in long-term care populations. NCHS Data Brief. 2011;(54):1-8.
19. Lund S, Richardson A, May C. Barriers to advance care planning at the end of life: an explanatory systematic review of implementation studies. PLoS One. 2015;10:e0116629.
20. Fried TR, Bullock K, Iannone L, et al. Understanding advance care planning as a process of health behavior change. J Am Geriatr Soc. 2009;57:1547-1555.
21. Schickedanz AD, Schillinger D, Landefeld CS, et al. A clinical framework for improving the advance care planning process: start with patients’ self-identified barriers. J Am Geriatr Soc. 2009;57:31-39.
22. Winter L, Parks SM, Diamond JJ. Ask a different question, get a different answer: why living wills are poor guides to care preferences at the end of life. J Palliat Med. 2010;13:567-572.
23. Institute of Medicine. Dying in America: Improving Quality and Honoring Individual Preferences Near the End of Life. www.nap.edu/read/18748/chapter/1. Accessed August 14, 2017.
24. Sudore RL, Schickedanz AD, Landefeld CS, et al. Engagement in multiple steps of the advance care planning process: a descriptive study of diverse older adults. J Am Geriatr Soc. 2008;56:1006-1013.
25. McMahan RD, Knight SJ, Fried TR, et al. Advance care planning beyond advance directives: perspectives from patients and surrogates. J Pain Symptom Manage. 2013;46:355-365.
26. Lum HD, Sudore RL, Bekelman DB. Advance care planning in the elderly. Med Clin North Am. 2015;99:391-403.
27. Lum HD, Sudore RL. Advance care planning and goals of care communication in older adults with cardiovascular disease and multi-morbidity. Clin Geriatr Med. 2016;32:247-260.
28. American College of Physicians. Advanced Care Planning: Implementation for practices. www.acponline.org/system/files/documents/practice-resources/business-resources/payment/advance_care_planning_toolkit.pdf. Accessed August 14, 2017.

Issue
Clinician Reviews - 27(9)
Issue
Clinician Reviews - 27(9)
Page Number
47-51
Page Number
47-51
Publications
Publications
Topics
Article Type
Display Headline
Advance Care Planning: Making It Easier for Patients (and You)
Display Headline
Advance Care Planning: Making It Easier for Patients (and You)
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Article PDF Media

First EDition: ED Visits by Older Patients Increase in the Weeks After a Disaster

Article Type
Changed
Wed, 12/12/2018 - 20:58
Display Headline
First EDition: ED Visits by Older Patients Increase in the Weeks After a Disaster, more

ED Visits by Older Patients Increase in the Weeks After a Disaster

BY KELLIE DESANTIS

Visits to an ED by adults ages 65 years and older increase significantly in the weeks following a disaster, according to a study published in Disaster Medicine and Public Health Preparedness.1

Older adults are vulnerable to the effects of disasters because of their diminished ability to adequately prepare for and respond to the effects of a disaster. Older adults suffering from visual, auditory, proprioceptive, and cognitive impairments are especially vulnerable and have the most difficulty complying with evacuation and preparatory warnings. Individuals with multiple chronic diseases, living in long-term care facilities or suffering from cognitive impairments are among the most vulnerable.

To better understand the impact of natural disasters on this vulnerable population, researchers examined the effects of the 2012 disaster, Hurricane Sandy, on older adults living in New York City (NYC) during the disaster. Researchers turned to the New York State Department of Health (NYSDOH) for data. The NYSDOH compiles a comprehensive database of claims from all ED visits in the Statewide Planning and Research Cooperative System (SPARCS), which is the most complete source for ED utilization in New York state, and includes primary and secondary diagnosis codes and patient addresses.

Researchers evaluated ED utilization by adults 65 years and older in the weeks immediately before and after the Hurricane Sandy landfall. They excluded patients who lived in a nursing home, were incarcerated, or visited an ED associated with a specialty hospital (surgical subspecialty, oncological, or Veterans Administration). By using geographic distribution information available from SPARCS and the NYC Office of Emergency Management evacuation zones, researchers were able to compare the ED utilization for older adults living in the evacuation zones before the landfall of Hurricane Sandy and in the weeks shortly after the storm.

The analysis revealed a significant increase in ED utilization for older adults living in the evacuation zones in the 3 weeks after the storm compared to ED use before the storm. The number of weekly ED visits by older adults from all evacuation zones was 9,852 in the weeks before Hurricane Sandy and increased in the first week after the storm to 10,073. Among the most severely impacted were older adults in evacuation zone one, where ED utilization increased from 552 visits to 1,111 visits. The number of ED visits remained elevated for 3 weeks after the storm but returned to normal by the fourth week.

Researchers suggested several reasons for this increase in ED visits, including seeking refuge in the ED as a result of homelessness due to the disaster, the interruption of ongoing care for chronic illness, environmental exposure, and the lack of preparation for the lasting effect of the disaster.

To improve the response to such a disaster in the future, a NYC Hurricane Sandy Assessment report2 recommended developing a door-to-door service task force for older adults to improve preparedness for this vulnerable population. The task force would be responsible for implementing an action plan to ensure that healthcare services, communication, and provisions for this population continue without interruption in the weeks following a disaster. Legal and regulatory changes would allow for Medicare recipients to be eligible for "early medication refill" and pre-storm "early dialysis" programs to improve the continuity of care of the chronically ill.

1. Malik S, Lee DC, Doran KM, et al. Vulnerability of older adults in disasters: emergency department utilization by geriatric patients after hurricane sandy. Disaster Med Public Health Prep. 2017:1-10. doi:10.1017/dmp.2017.44

2. The City of New York, Office of the Mayor. Hurricane Sandy After Action Report. Published May 2013. http://www.nyc.gov/html/recovery/downlaods/pdf/sandy_aar_5.2.13.pdf. Accessed September 1, 2017.

Digital Rectal Examination of ED Patients with Acute GI Bleeding Cuts Rates of Admissions, Pharmacotherapy, and Endoscopy

BY JEFF BAUER

Patients presenting to the ED with acute gastrointestinal (GI) bleeding who receive a digital rectal examination have significantly lower rates of admissions, pharmacotherapy, and endoscopies, according to a retrospective study published in The American Journal of Medicine. Digital rectal examinations are an established part of the physical examination of a patient with GI bleeding, but physicians often are reluctant to conduct such examinations. Previous studies have found that 10% to 35% of patients with acute GI bleeding do not receive digital rectal examinations.

In the current study, researchers analyzed data from the electronic health records (EHRs) of patients ages 18 years and older who presented to the ED of a single institution with acute GI bleeding, as identified by International Classification of Diseases, Ninth Edition codes. They collected patients’ medical histories, demographic information, and clinical and laboratory data. ED clinician notes were used to determine which patients received a digital rectal examination. The outcomes researchers assessed were hospital admission, intensive care unit (ICU) admission, initiation of medical therapy (a proton pump inhibitor or octreotide), inpatient endoscopy (upper endoscopy or colonoscopy), and packed red blood cell (RBC) transfusion.

Overall, 1237 patients presented with acute GI bleeding. Most patients were Caucasian (49.2%) or Hispanic (38.4%), 44.9% were female, and the median age was 53 years.

Slightly more than one-half of patients (55.6%) received a digital rectal examination. In total, 736 patients were admitted—including 222 admissions to the ICU; 751 were started on a proton pump inhibitor or octreotide, 274 underwent endoscopy, and 321 received an RBC transfusion.

Patients were more likely to receive a digital rectal examination if they were older, Hispanic, or receiving an anticoagulant. Patients were less likely to undergo such examinations if they presented with altered mental status or hematemesis. Compared to patients who did not receive a digital rectal examination, those who did were significantly less likely to be admitted to the hospital (P = .004), to be starting on medical therapy (P = .04), or to undergo endoscopy (P = .02). There were no significant differences between these two groups in terms of ICU admissions, gastroenterology consultations, or transfusions.

Researchers suggested that the 44% rate of patients with acute GI bleeding who did not receive digital rectal examinations was higher than had been reported in previous studies. The difference had been the result of relying solely on ED clinician notes for this data, without including notes from admitting or consulting clinicians. The authors also were unable to determine the reasons these examinations were not conducted.

Shrestha MP, Borgstrom M, Trowers E. Digital rectal examination reduces hospital admissions, endoscopies, and medical therapy in patients with acute gastrointestinal bleeding. Am J Med. 2017;130(7):819-825. doi: 10.1016/j.amjmed.2017.01.036.

References

 

Article PDF
Issue
Emergency Medicine - 49(9)
Publications
Topics
Page Number
389-390, 390
Sections
Article PDF
Article PDF

ED Visits by Older Patients Increase in the Weeks After a Disaster

BY KELLIE DESANTIS

Visits to an ED by adults ages 65 years and older increase significantly in the weeks following a disaster, according to a study published in Disaster Medicine and Public Health Preparedness.1

Older adults are vulnerable to the effects of disasters because of their diminished ability to adequately prepare for and respond to the effects of a disaster. Older adults suffering from visual, auditory, proprioceptive, and cognitive impairments are especially vulnerable and have the most difficulty complying with evacuation and preparatory warnings. Individuals with multiple chronic diseases, living in long-term care facilities or suffering from cognitive impairments are among the most vulnerable.

To better understand the impact of natural disasters on this vulnerable population, researchers examined the effects of the 2012 disaster, Hurricane Sandy, on older adults living in New York City (NYC) during the disaster. Researchers turned to the New York State Department of Health (NYSDOH) for data. The NYSDOH compiles a comprehensive database of claims from all ED visits in the Statewide Planning and Research Cooperative System (SPARCS), which is the most complete source for ED utilization in New York state, and includes primary and secondary diagnosis codes and patient addresses.

Researchers evaluated ED utilization by adults 65 years and older in the weeks immediately before and after the Hurricane Sandy landfall. They excluded patients who lived in a nursing home, were incarcerated, or visited an ED associated with a specialty hospital (surgical subspecialty, oncological, or Veterans Administration). By using geographic distribution information available from SPARCS and the NYC Office of Emergency Management evacuation zones, researchers were able to compare the ED utilization for older adults living in the evacuation zones before the landfall of Hurricane Sandy and in the weeks shortly after the storm.

The analysis revealed a significant increase in ED utilization for older adults living in the evacuation zones in the 3 weeks after the storm compared to ED use before the storm. The number of weekly ED visits by older adults from all evacuation zones was 9,852 in the weeks before Hurricane Sandy and increased in the first week after the storm to 10,073. Among the most severely impacted were older adults in evacuation zone one, where ED utilization increased from 552 visits to 1,111 visits. The number of ED visits remained elevated for 3 weeks after the storm but returned to normal by the fourth week.

Researchers suggested several reasons for this increase in ED visits, including seeking refuge in the ED as a result of homelessness due to the disaster, the interruption of ongoing care for chronic illness, environmental exposure, and the lack of preparation for the lasting effect of the disaster.

To improve the response to such a disaster in the future, a NYC Hurricane Sandy Assessment report2 recommended developing a door-to-door service task force for older adults to improve preparedness for this vulnerable population. The task force would be responsible for implementing an action plan to ensure that healthcare services, communication, and provisions for this population continue without interruption in the weeks following a disaster. Legal and regulatory changes would allow for Medicare recipients to be eligible for "early medication refill" and pre-storm "early dialysis" programs to improve the continuity of care of the chronically ill.

1. Malik S, Lee DC, Doran KM, et al. Vulnerability of older adults in disasters: emergency department utilization by geriatric patients after hurricane sandy. Disaster Med Public Health Prep. 2017:1-10. doi:10.1017/dmp.2017.44

2. The City of New York, Office of the Mayor. Hurricane Sandy After Action Report. Published May 2013. http://www.nyc.gov/html/recovery/downlaods/pdf/sandy_aar_5.2.13.pdf. Accessed September 1, 2017.

Digital Rectal Examination of ED Patients with Acute GI Bleeding Cuts Rates of Admissions, Pharmacotherapy, and Endoscopy

BY JEFF BAUER

Patients presenting to the ED with acute gastrointestinal (GI) bleeding who receive a digital rectal examination have significantly lower rates of admissions, pharmacotherapy, and endoscopies, according to a retrospective study published in The American Journal of Medicine. Digital rectal examinations are an established part of the physical examination of a patient with GI bleeding, but physicians often are reluctant to conduct such examinations. Previous studies have found that 10% to 35% of patients with acute GI bleeding do not receive digital rectal examinations.

In the current study, researchers analyzed data from the electronic health records (EHRs) of patients ages 18 years and older who presented to the ED of a single institution with acute GI bleeding, as identified by International Classification of Diseases, Ninth Edition codes. They collected patients’ medical histories, demographic information, and clinical and laboratory data. ED clinician notes were used to determine which patients received a digital rectal examination. The outcomes researchers assessed were hospital admission, intensive care unit (ICU) admission, initiation of medical therapy (a proton pump inhibitor or octreotide), inpatient endoscopy (upper endoscopy or colonoscopy), and packed red blood cell (RBC) transfusion.

Overall, 1237 patients presented with acute GI bleeding. Most patients were Caucasian (49.2%) or Hispanic (38.4%), 44.9% were female, and the median age was 53 years.

Slightly more than one-half of patients (55.6%) received a digital rectal examination. In total, 736 patients were admitted—including 222 admissions to the ICU; 751 were started on a proton pump inhibitor or octreotide, 274 underwent endoscopy, and 321 received an RBC transfusion.

Patients were more likely to receive a digital rectal examination if they were older, Hispanic, or receiving an anticoagulant. Patients were less likely to undergo such examinations if they presented with altered mental status or hematemesis. Compared to patients who did not receive a digital rectal examination, those who did were significantly less likely to be admitted to the hospital (P = .004), to be starting on medical therapy (P = .04), or to undergo endoscopy (P = .02). There were no significant differences between these two groups in terms of ICU admissions, gastroenterology consultations, or transfusions.

Researchers suggested that the 44% rate of patients with acute GI bleeding who did not receive digital rectal examinations was higher than had been reported in previous studies. The difference had been the result of relying solely on ED clinician notes for this data, without including notes from admitting or consulting clinicians. The authors also were unable to determine the reasons these examinations were not conducted.

Shrestha MP, Borgstrom M, Trowers E. Digital rectal examination reduces hospital admissions, endoscopies, and medical therapy in patients with acute gastrointestinal bleeding. Am J Med. 2017;130(7):819-825. doi: 10.1016/j.amjmed.2017.01.036.

ED Visits by Older Patients Increase in the Weeks After a Disaster

BY KELLIE DESANTIS

Visits to an ED by adults ages 65 years and older increase significantly in the weeks following a disaster, according to a study published in Disaster Medicine and Public Health Preparedness.1

Older adults are vulnerable to the effects of disasters because of their diminished ability to adequately prepare for and respond to the effects of a disaster. Older adults suffering from visual, auditory, proprioceptive, and cognitive impairments are especially vulnerable and have the most difficulty complying with evacuation and preparatory warnings. Individuals with multiple chronic diseases, living in long-term care facilities or suffering from cognitive impairments are among the most vulnerable.

To better understand the impact of natural disasters on this vulnerable population, researchers examined the effects of the 2012 disaster, Hurricane Sandy, on older adults living in New York City (NYC) during the disaster. Researchers turned to the New York State Department of Health (NYSDOH) for data. The NYSDOH compiles a comprehensive database of claims from all ED visits in the Statewide Planning and Research Cooperative System (SPARCS), which is the most complete source for ED utilization in New York state, and includes primary and secondary diagnosis codes and patient addresses.

Researchers evaluated ED utilization by adults 65 years and older in the weeks immediately before and after the Hurricane Sandy landfall. They excluded patients who lived in a nursing home, were incarcerated, or visited an ED associated with a specialty hospital (surgical subspecialty, oncological, or Veterans Administration). By using geographic distribution information available from SPARCS and the NYC Office of Emergency Management evacuation zones, researchers were able to compare the ED utilization for older adults living in the evacuation zones before the landfall of Hurricane Sandy and in the weeks shortly after the storm.

The analysis revealed a significant increase in ED utilization for older adults living in the evacuation zones in the 3 weeks after the storm compared to ED use before the storm. The number of weekly ED visits by older adults from all evacuation zones was 9,852 in the weeks before Hurricane Sandy and increased in the first week after the storm to 10,073. Among the most severely impacted were older adults in evacuation zone one, where ED utilization increased from 552 visits to 1,111 visits. The number of ED visits remained elevated for 3 weeks after the storm but returned to normal by the fourth week.

Researchers suggested several reasons for this increase in ED visits, including seeking refuge in the ED as a result of homelessness due to the disaster, the interruption of ongoing care for chronic illness, environmental exposure, and the lack of preparation for the lasting effect of the disaster.

To improve the response to such a disaster in the future, a NYC Hurricane Sandy Assessment report2 recommended developing a door-to-door service task force for older adults to improve preparedness for this vulnerable population. The task force would be responsible for implementing an action plan to ensure that healthcare services, communication, and provisions for this population continue without interruption in the weeks following a disaster. Legal and regulatory changes would allow for Medicare recipients to be eligible for "early medication refill" and pre-storm "early dialysis" programs to improve the continuity of care of the chronically ill.

1. Malik S, Lee DC, Doran KM, et al. Vulnerability of older adults in disasters: emergency department utilization by geriatric patients after hurricane sandy. Disaster Med Public Health Prep. 2017:1-10. doi:10.1017/dmp.2017.44

2. The City of New York, Office of the Mayor. Hurricane Sandy After Action Report. Published May 2013. http://www.nyc.gov/html/recovery/downlaods/pdf/sandy_aar_5.2.13.pdf. Accessed September 1, 2017.

Digital Rectal Examination of ED Patients with Acute GI Bleeding Cuts Rates of Admissions, Pharmacotherapy, and Endoscopy

BY JEFF BAUER

Patients presenting to the ED with acute gastrointestinal (GI) bleeding who receive a digital rectal examination have significantly lower rates of admissions, pharmacotherapy, and endoscopies, according to a retrospective study published in The American Journal of Medicine. Digital rectal examinations are an established part of the physical examination of a patient with GI bleeding, but physicians often are reluctant to conduct such examinations. Previous studies have found that 10% to 35% of patients with acute GI bleeding do not receive digital rectal examinations.

In the current study, researchers analyzed data from the electronic health records (EHRs) of patients ages 18 years and older who presented to the ED of a single institution with acute GI bleeding, as identified by International Classification of Diseases, Ninth Edition codes. They collected patients’ medical histories, demographic information, and clinical and laboratory data. ED clinician notes were used to determine which patients received a digital rectal examination. The outcomes researchers assessed were hospital admission, intensive care unit (ICU) admission, initiation of medical therapy (a proton pump inhibitor or octreotide), inpatient endoscopy (upper endoscopy or colonoscopy), and packed red blood cell (RBC) transfusion.

Overall, 1237 patients presented with acute GI bleeding. Most patients were Caucasian (49.2%) or Hispanic (38.4%), 44.9% were female, and the median age was 53 years.

Slightly more than one-half of patients (55.6%) received a digital rectal examination. In total, 736 patients were admitted—including 222 admissions to the ICU; 751 were started on a proton pump inhibitor or octreotide, 274 underwent endoscopy, and 321 received an RBC transfusion.

Patients were more likely to receive a digital rectal examination if they were older, Hispanic, or receiving an anticoagulant. Patients were less likely to undergo such examinations if they presented with altered mental status or hematemesis. Compared to patients who did not receive a digital rectal examination, those who did were significantly less likely to be admitted to the hospital (P = .004), to be starting on medical therapy (P = .04), or to undergo endoscopy (P = .02). There were no significant differences between these two groups in terms of ICU admissions, gastroenterology consultations, or transfusions.

Researchers suggested that the 44% rate of patients with acute GI bleeding who did not receive digital rectal examinations was higher than had been reported in previous studies. The difference had been the result of relying solely on ED clinician notes for this data, without including notes from admitting or consulting clinicians. The authors also were unable to determine the reasons these examinations were not conducted.

Shrestha MP, Borgstrom M, Trowers E. Digital rectal examination reduces hospital admissions, endoscopies, and medical therapy in patients with acute gastrointestinal bleeding. Am J Med. 2017;130(7):819-825. doi: 10.1016/j.amjmed.2017.01.036.

References

 

References

 

Issue
Emergency Medicine - 49(9)
Issue
Emergency Medicine - 49(9)
Page Number
389-390, 390
Page Number
389-390, 390
Publications
Publications
Topics
Article Type
Display Headline
First EDition: ED Visits by Older Patients Increase in the Weeks After a Disaster, more
Display Headline
First EDition: ED Visits by Older Patients Increase in the Weeks After a Disaster, more
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Article PDF Media

Is Ketamine the New Wonder Drug for Treating Suicide?

Article Type
Changed
Thu, 04/26/2018 - 10:10
Although the initial findings involving the use of ketamine in suicidal patients are promising, further research is needed on the short- and long-term effects of this medication.

In 2014 the suicide rate in the U.S. was 13/100,000, the highest recorded in 28 years.1 Suicide is now considered the 10th leading cause of death for all ages, and the rate has increased every year from 2000 to 2014 among both women and men and in every age group except those aged ≥ 75 years.1-3 For those aged 15 to 44 years, suicide is among the top 3 causes of death worldwide.4-6

Background

In 2013, more than 490,000 hospital visits related to suicide attempts were reported in the U.S.4 Health care expenditures related to suicide are estimated at $56.9 billion in combined medical and work loss costs annually and an unmeasurable cost to the affected families.7 The mental health care community is desperate for ways to address this epidemic, and the National Academies of Medicine (NAM) has declared that research that directly addresses comparative effectiveness of treatment strategies following a suicide attempt should be a national priority.8

The most recent reports from 2014 indicate that the suicide rates are higher for male veterans than for male nonveterans (32.1 vs 20.9 per 100,000, respectively) and are much higher for female veterans than for female nonveterans (28.7 vs 5.2 per 100,000, respectively).3 Suicide rates also may be associated with veteran-specific comorbidities, such as higher rates of depression, anxiety, posttraumatic stress disorder (PTSD), and war-related trauma.3 According to the VHA, the suicide rate for veterans aged > 30 years also is rapidly increasing, and VHA has echoed the calls from NAM to make suicide prevention research a national priority.3

The VA has tried to stem the tide of suicides in veterans by implementing many advances in suicide prevention, including hiring suicide prevention coordinators at every VA hospital, enhanced monitoring, and the availability of 24-hour crisis hotline services. Yet the suicide rates for veterans continue to rise and remain higher than the rates in the general population.3

About 90% of deaths by suicide are by persons who have a treatable psychiatric disorder, most commonly a mood disorder, such as depression.4 However, most studies show that antidepressant therapy does not provide rapid or significant relief of suicidal ideation (SI).4 Therefore, the current standard of care for the treatment of acutely suicidal patients includes a combination of hospitalization, cognitive behavioral therapy or psychotherapy, case management, antidepressant medications, and electroconvulsive therapy (ECT).4 Even though these therapies have become more widely available over the past decade, rates of suicide continue to increase.1,4 These interventions have limited effectiveness in acute settings. Although both intensive outpatient follow-up and routine outpatient care have been studied in relation to the decrease of suicidal behavior, neither intervention has been shown to immediately reduce suicidal behavior significantly in patients.

Suicidality Interventions

Therapy and case management require patients to be well enough to make office visits and follow through with care for periods as long as 1 year, which is often not possible for individuals with severe depression.5 One-third of patients who attended 6 months of outpatient therapy consistently still met the criteria for major depressive disorder (MDD), a major risk for suicide attempt.9 Antidepressant medications take a minimum of 4 weeks to reach full efficacy, and many patients stop taking the medications before that point because of concern that the medication is not helping or because of adverse effects (AEs), such as sleep disturbance, sexual dysfunction, or weight gain.9

Electroconvulsive therapy has been shown to be an effective treatment for patients with depression and suicidal behavior, but adherence with 12 weeks of recommended therapy has been a barrier for this intervention. Additionally, ECT may not provide reduction in SI for 1 to 2 weeks.4,10 A review of research studies showed that nearly 50% of patients with high-expressed SI did not complete the prescribed amount of ECT due to the length of time to complete the recommended 12 sessions.10 Therefore, current treatment barriers for suicidal patients include: (1) long periods in treatment for therapy, medication, and ECT before any relief of symptoms is noted; (2) high recidivism rates for MDD symptoms and risk of suicide following treatment; and (3) high treatment dropout rates.

Pharmacologic treatments currently used in suicidal patients have not fared much better. Many have received FDA approval for treatment of associated mental health diagnoses such as bipolar disorder, schizophrenia, or MDD, but there are no approved treatments that specifically target suicidal behavior. Lithium is approved for reducing the long-term risk of SI primarily because it reduces the risk of mood disorders associated with SI, but lithium has not been shown to be effective in acute settings.11 Clozapine is approved for reducing the long-term risk of recurrent suicide in patients with schizophrenia or schizoaffective disorder.4 Clozapine has not been shown to be effective in patients with mood disorders, which make up the majority of patients who attempt suicide.4 Additionally, both medications are plagued by the same barriers listed earlier, such as long time to effect (it takes an average 4 weeks to reach efficacy), lack of efficacy in acute settings, and AEs (eg, sleep problems, weight gain, and sexual dysfunction).9 Thus finding better pharmacologic interventions for suicidal patients is a priority for current research.

 

 

Ketamine

Recently, researchers have identified ketamine as a potential therapeutic option for depression and SI. A single ketamine infusion treatment has a rapid response, minimal AEs, and potentially long-lasting efficacy with SI, which would make it ideal for the treatment of acutely suicidal patients.4 Ketamine is an N-methyl-D-aspartate receptor (NMDAR) inhibitor that also has been found to be a weak μ- and κ-opioid receptor agonist and an inhibitor of the reuptake of serotonin, dopamine, and norepinephrine. Inhibition of the NMDAR results in analgesia, and ketamine is approved for the induction of anesthesia, pain relief, and sedation.12

Although AEs such as hallucinations and sedation create the potential for dangerous recreational use, ketamine is safely used in health care settings for a variety of indications. Effects are noted within 5 minutes of administration if given by infusion, and the main effects can last between 20 and 40 minutes.

Ketamine has a complex pharmacology and plays a role in other cell signaling mechanisms, but the significance of these additional mechanisms in the therapeutic effects of ketamine have only recently been elucidated. Preclinical studies indicate a probable NMDAR inhibition-independent mechanism responsible for the antidepressant response to ketamine.13,14 The complex associations with rapamycin signaling, eukaryotic elongation factor 2 dephosphorylation, increased synthesis of brain-derived neurotrophic factor, and activation of glutamatergic AMPA (α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid) receptors have been linked to its rapid antidepressant effect and ketamine’s induction of synaptogenesis within the limbic system.13,14

 

Clinical Research

Ketamine was studied as an adjunctive treatment to psychotherapy for addictions as far back as the 1970s.15 The available reports indicate a universally positive result, with increased rates of remission and decreased rates of relapse attributed to ketamine’s ability to alter one’s thought processes by reinforcing limbic-cortex interactions that facilitate the growth of more positive cognitive schemas and improved emotional attitudes about the self in support of the recovery process.15

Neurobiologic studies have shown that treatment with ketamine has a direct and immediate effect on neuronal pathways of the limbic system. It is known to regulate the mind’s reaction to positive stimuli by reversing the depressed subject’s blunted reaction to positive faces.16 This rapid normalization of the positive faces test is unique to ketamine infusion and is not seen in tests with traditional antidepressants.

In 2000, the first placebo-controlled trial using ketamine for treatment resistant depression (TRD) demonstrated the rapid antidepressant effects of a single dose of ketamine, but this study only looked at these effects for 1 week.17 In multiple double blind, placebo-controlled trials since then, IV infusion of ketamine was shown to be an effective intervention for TRD.13,18,19 More recently, a published investigation involving the treatment of MDD showed that ketamine in conjunction with a selective serotonin reuptake inhibitor (SSRI) accelerated and enhanced the effectiveness of the SSRI in reducing depressive symptoms.20

Based on the rapid resolution of depressive symptoms using ketamine, researchers have looked at its effect on suicidality as a secondary measure. A case study of a patient with severe depressive episodes and multiple previous suicidal attempts reported that the patient responded to a single dose of ketamine, described the experience as “being reborn,” and maintained complete remission of SI for the 6-month study period.21 In a larger study, 133 TRD patients received a single IV dose of ketamine with significant reductions in SI independent of depressive and anxiety symptoms.22

Depression Treatment

These results have led to an excitement for ketamine therapy as a novel treatment of depression, and off-label use by treatment centers now exists in several countries to aid those with TRD.23 This off-label use continues to be controversial, as research has yet to determine the safest most effective route and duration of treatment and whether the ketamine treatment AEs will exceed any accrued therapeutic benefit.13

The American Psychological Association Council of Research Task Force on Novel Biomarkers and Treatment critically examined the clinical evidence of ketamine use and has raised important concerns about the use of ketamine in the outpatient setting, administered in the absence of consensus therapeutic monitoring guidelines, and ambitiously marketed as a panacea for TRD.13,24 A study showed permanent impairment of brain function for both groups compared with monkeys treated with saline infusions.25 In 2016, the FDA gave fast-track approval for an intranasal ketamine that would make the treatment more easily available in the outpatient setting, but this could lead to certain patients developing a dependency on ketamine or engaging in its diversion for recreational use. There are case reports and anecdotes in the literature of patients and research subjects developing drug-seeking behaviors and overuse of ketamine.24 Additionally, the comorbidities associated with TRD and SI have not been fully evaluated. For instance, there is evidence that depressed patients with obsessive compulsive disorder may have worse outcomes that include delayed onset SI.26

There also is concern for the use of ketamine for chronic opioid users. The combination of ketamine with opioids may increase the response to the opioid in an otherwise drug tolerant patient, leading to risk of death by overdose in patients who have not increased their usual dose.27 However, this effect was noted only when ketamine and opioids were administered together, and the effect does not seem to last postinfusion.27

The challenges in treatment of TRD include finding an effective formulation—IV infusion of ketamine requires cardiovascular monitoring and is administered by anesthesiologists. The short duration of action for depression requires repeated infusions, and the frequency and quantity of infusions have not been determined. Efforts to find other NMDAR inhibitors (eg, memantine, nitrous oxide, D-cycloserine, and others) that match ketamine’s antidepressant efficacy but with easier delivery methods and fewer risks have thus far been unsuccessful.13 It is now believed that ketamine’s unique ability to activate intracellular signaling pathways linked to synaptic plasticity gives it the antidepressant function. Recent studies have further narrowed ketamine’s antidepressant function to the R- enantiomer of the ketamine metabolite, hydroxynorketamine.14 The nasal spray for ketamine is the S- enantiomer, which has better bioavailability but may have less antidepressant efficacy compared with the racemic mixture used in ketamine infusions.

 

 

Suicide Ideation Treatment

The many challenges faced by researchers and clinicians trying to develop ketamine treatment for TRD may not apply to the treatment of SI. Whereas repeated doses of ketamine cannot reliably produce sustained remission of depression, the few studies that have looked at the long-term effects of ketamine treatment on SI indicate the potential for long-term efficacy after a single IV infusion.21,22 Although treatment with IV infusions have additional costs and logistics, if it is found beneficial, it could be given in the emergency department (ED) prior to hospitalization and potentially lead to better outcomes.

In 2011, a small preliminary observational study of patients with depression and SI presenting to the ED indicated that SI was rapidly reduced following an infusion of ketamine.28 This study showed that both depressive symptoms and suicidality rapidly and significantly diminished within 40 minutes with no evidence of the recurrence of symptoms 10 days postadministration.

 

A more recent study used ketamine in a military field hospital to treat SI and also concluded that it could be effective and safe when administered in an ED setting. This preliminary study suggests that ketamine could be a safe and potentially effective medication for rapid reduction of depression and suicidality in a busy ED setting.29 These limited studies involving the use of ketamine in patients with SI show promise with long-term effectiveness. However, more research is needed to clarify whether the efficacy with SI will be similar to the clinical experience seen in TRD; a duration of effect limited to 2 weeks with recurrence after treatment discontinued.24

Conclusion

There has been a compelling accumulation of scientific data since 2000 to support the use of ketamine for the treatment of depression and SI. Ketamine use in patients with these diagnoses showed a rapid decrease of symptoms and minimal AEs among a significant number of patients.22,30

Although the initial findings involving the use of ketamine in suicidal patients are promising, the clinical use of ketamine needs further research, using larger sample sizes and exploring both the short-term and long-term effects of this medication. Researchers need to further establish the safe and effective route, point of care, and patient type that would best respond to this novel treatment. The initial evidence would suggest that health care providers have every right to be hopeful that ketamine will become the first pharmacologic treatment of acute SI in a majority of patients presenting to EDs, mental health clinics, community hospitals, and VA medical centers.

References

1. Curtin SC, Warner MA, Hedegaard H. Increase in suicide in the United States 199-2014. NCHS data brief, no. 241. https://www.cdc.gov/nchs/data/data -briefs/db241.pdf. Published April 2016. Accessed August 3, 2017.

2. Nock MK, Borges G, Bromet EJ, Cha CB, Kessler RC, Lee S. Suicide and suicidal behavior. Epidemiol Rev. 2008;30(1):133-154.

3. U.S. Department of Veteran Affairs Office of Suicide Prevention. Suicide among veterans and other Americans 2001-2014. https://www.mentalhealth .va.gov/docs/2016suicidedatareport.pdf Published August 3, 2016. Accessed August 11, 2017.

4. Wilkinson ST, Sanacora G. Ketamine: a potential rapid-acting antisuicidal agent? Depress Anxiety. 2016;33(8):711-717.

5. Aleman A, Denys D. Mental health: a road map for suicide research and prevention. Nature. 2014;509(7501):421-423.

6. Griffiths JJ, Zarate CA, Jr, Rasimas JJ. Existing and novel biological therapeutics in suicide prevention. Am J Prev Med. 2014;47(3)(suppl 2):S195-S203.

7. Centers for Disease Control and Prevention. Leading causes of death reports, 1981-2015. https://www.cdc.gov/injury/wisqars/leading_causes_death.html. Updated February 19, 2017. Accessed August 14, 2017.

8. Institute of Medicine of the National Academies; Board on Health Care Services; Committee on Comparative Effectiveness Research Prioritization. Initial National Priorities for Comparative Effectiveness Research. Washington, DC: The National Academies Press; 2009.

9. Weinberger MI, Sirey JA, Bruce ML, Heo M, Papademetriou E, Meyers BS. Predictors of major depression six months after admission for outpatient treatment. Psychiatr Serv. 2008;59(10):1211-1215.

10. Kellner CH, Fink M, Knapp R, et al. Relief of expressed suicidal intent by ECT: a consortium for research in ECT study. Am J Psychiatry. 2005;162(5):977-982.

11. Lewitzka U, Jabs B, Fülle M, et al. Does lithium reduce acute suicidal ideation and behavior? A protocol for a randomized, placebo-controlled multicenter trial of lithium plus treatment as usual (TAU) in patients with suicidal major depressive episode. BMC Psychiatry. 2015;15:117.

12. Vadivelu N, Schermer E, Kodumudi V, Belani K, Urman RD, Kaye AD. Role of ketamine for analgesia in adults and children. J Anaesthesiol Clin Pharmacol. 2016;32(3):298-306.

13. Newport DJ, Carpenter LL, McDonald WM, et al; APA Council of Research Task Force on Novel Biomarkers and Treatments. Ketamine and other NMDA antagonists: early clinical trials and possible mechanisms in depression. Am J Psychiatry. 2015;172(10):950-966.

14. Zanos P, Moaddel R, Morris PJ, et al. NMDAR inhibition-independent antidepressant actions of ketamine metabolites. Nature. 2016;533(7604):481-486.

15. Krupitsky EM, Grinenko AY. Ketamine psychedelic therapy (KPT): a review of the results of ten years of research. J Psychoactive Drugs. 1997;29(2):165-183.

16. Murrough JW, Collins KA, Fields J, et al. Regulation of neural responses to emotion perception by ketamine in individuals with treatment-resistant major depressive disorder. Transl Psychiatry. 2015;5:e509.

17. Berman RM, Cappiello A, Anand A, et al. Antidepressant effects of ketamine in depressed patients. Biol Psychiatry. 2000;47(4):351-354.

18. Murrough JW, Iosifescu DV, Chang LC, et al. Antidepressant efficacy of ketamine in treatment-resistant major depression: a two-site randomized controlled trial. Am J Psychiatry. 2013;170(10):1134-1142.

19. Zarate CA Jr, Singh JB, Carlson PJ, et al. A randomized trial of an N-methyl-D-aspartate antagonist in treatment-resistant major depression. Arch Gen Psychiatry. 2006;63(8):856-864.

20. Hu YD, Xiang YT, Fang JX, et al. Single i.v. ketamine augmentation of newly initiated escitalopram for major depression: results from a randomized, placebo-controlled 4-week study. Psychol Med. 2016;46(3):623-635.

21. Aligeti S, Quinones M, Salazar R. Rapid resolution of suicidal behavior and depression with single low-dose ketamine intravenous push even after 6 months of follow-up. J Clin Psychopharmacol. 2014;34(4):533-535.

22. Ballard ED, Ionescu DF, Vande Voort JL, et al. Improvement in suicidal ideation after ketamine infusion: relationship to reductions in depression and anxiety. J Psychiatr Res. 2014;58:161-166.

23. Henderson TA. Practical application of the neuroregenerative properties of ketamine: real world treatment experience. Neural Regen Res. 2016;11(2):195-200.

24. Newport DJ, Schatzberg AF, Nemeroff CB. Whither ketamine as an antidepressant: panacea or toxin? Depress Anxiety. 2016;33(8):685-688.

25. Sun L, Li Q, Li Q, et al. Chronic ketamine exposure induces permanent impairment of brain functions in adolescent cynomolgus monkeys. Addict Biol. 2014;19(2):185-194.

26. Niciu MJ, Grunschel BD, Corlett PR, Pittenger C, Bloch MH. Two cases of delayed-onset suicidal ideation, dysphoria and anxiety after ketamine infusion in patients with obsessive-compulsive disorder and a history of major depressive disorder. J Psychopharmacol. 2013;27(7):651-654.

27. Huxtable CA, Roberts LJ, Somogyi AA, MacIntyre PE. Acute pain management in opioid-tolerant patients: a growing challenge. Anaesth Intensive Care. 2011;39(5):804-823.

28. Larkin GL, Beautrais AL. A preliminary naturalistic study of low-dose ketamine for depression and suicide ideation in the emergency department. Int J Neuropsychopharmacol. 2011;14(8):1127-1131.

29. Burger J, Capobianco M, Lovem R, et al. A double-blinded, randomized, placebo-controlled sub-dissociative dose ketamine pilot study in the treatment of acute depression and suicidality in a military emergency department setting. Mil Med. 2016;181(10):1195-1199.

30. Wan LB, Levitch CF, Perez AM, et al. Ketamine safety and tolerability in clinical trials for treatment-resistant depression. J Clin Psychiatry. 2015;76(3):247-252.

Article PDF
Author and Disclosure Information

Dr. Boilini is a staff psychiatrist, Ms. Baldwin is a clinical social worker, and Dr. Lamvu is a gynecologist, all at Orlando VAMC in Florida. Dr. Lamvu also is a professor at the University of Central Florida College of Medicine in Orlando.

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 - 34(9)
Publications
Topics
Page Number
12-16
Sections
Author and Disclosure Information

Dr. Boilini is a staff psychiatrist, Ms. Baldwin is a clinical social worker, and Dr. Lamvu is a gynecologist, all at Orlando VAMC in Florida. Dr. Lamvu also is a professor at the University of Central Florida College of Medicine in Orlando.

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. Boilini is a staff psychiatrist, Ms. Baldwin is a clinical social worker, and Dr. Lamvu is a gynecologist, all at Orlando VAMC in Florida. Dr. Lamvu also is a professor at the University of Central Florida College of Medicine in Orlando.

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
Although the initial findings involving the use of ketamine in suicidal patients are promising, further research is needed on the short- and long-term effects of this medication.
Although the initial findings involving the use of ketamine in suicidal patients are promising, further research is needed on the short- and long-term effects of this medication.

In 2014 the suicide rate in the U.S. was 13/100,000, the highest recorded in 28 years.1 Suicide is now considered the 10th leading cause of death for all ages, and the rate has increased every year from 2000 to 2014 among both women and men and in every age group except those aged ≥ 75 years.1-3 For those aged 15 to 44 years, suicide is among the top 3 causes of death worldwide.4-6

Background

In 2013, more than 490,000 hospital visits related to suicide attempts were reported in the U.S.4 Health care expenditures related to suicide are estimated at $56.9 billion in combined medical and work loss costs annually and an unmeasurable cost to the affected families.7 The mental health care community is desperate for ways to address this epidemic, and the National Academies of Medicine (NAM) has declared that research that directly addresses comparative effectiveness of treatment strategies following a suicide attempt should be a national priority.8

The most recent reports from 2014 indicate that the suicide rates are higher for male veterans than for male nonveterans (32.1 vs 20.9 per 100,000, respectively) and are much higher for female veterans than for female nonveterans (28.7 vs 5.2 per 100,000, respectively).3 Suicide rates also may be associated with veteran-specific comorbidities, such as higher rates of depression, anxiety, posttraumatic stress disorder (PTSD), and war-related trauma.3 According to the VHA, the suicide rate for veterans aged > 30 years also is rapidly increasing, and VHA has echoed the calls from NAM to make suicide prevention research a national priority.3

The VA has tried to stem the tide of suicides in veterans by implementing many advances in suicide prevention, including hiring suicide prevention coordinators at every VA hospital, enhanced monitoring, and the availability of 24-hour crisis hotline services. Yet the suicide rates for veterans continue to rise and remain higher than the rates in the general population.3

About 90% of deaths by suicide are by persons who have a treatable psychiatric disorder, most commonly a mood disorder, such as depression.4 However, most studies show that antidepressant therapy does not provide rapid or significant relief of suicidal ideation (SI).4 Therefore, the current standard of care for the treatment of acutely suicidal patients includes a combination of hospitalization, cognitive behavioral therapy or psychotherapy, case management, antidepressant medications, and electroconvulsive therapy (ECT).4 Even though these therapies have become more widely available over the past decade, rates of suicide continue to increase.1,4 These interventions have limited effectiveness in acute settings. Although both intensive outpatient follow-up and routine outpatient care have been studied in relation to the decrease of suicidal behavior, neither intervention has been shown to immediately reduce suicidal behavior significantly in patients.

Suicidality Interventions

Therapy and case management require patients to be well enough to make office visits and follow through with care for periods as long as 1 year, which is often not possible for individuals with severe depression.5 One-third of patients who attended 6 months of outpatient therapy consistently still met the criteria for major depressive disorder (MDD), a major risk for suicide attempt.9 Antidepressant medications take a minimum of 4 weeks to reach full efficacy, and many patients stop taking the medications before that point because of concern that the medication is not helping or because of adverse effects (AEs), such as sleep disturbance, sexual dysfunction, or weight gain.9

Electroconvulsive therapy has been shown to be an effective treatment for patients with depression and suicidal behavior, but adherence with 12 weeks of recommended therapy has been a barrier for this intervention. Additionally, ECT may not provide reduction in SI for 1 to 2 weeks.4,10 A review of research studies showed that nearly 50% of patients with high-expressed SI did not complete the prescribed amount of ECT due to the length of time to complete the recommended 12 sessions.10 Therefore, current treatment barriers for suicidal patients include: (1) long periods in treatment for therapy, medication, and ECT before any relief of symptoms is noted; (2) high recidivism rates for MDD symptoms and risk of suicide following treatment; and (3) high treatment dropout rates.

Pharmacologic treatments currently used in suicidal patients have not fared much better. Many have received FDA approval for treatment of associated mental health diagnoses such as bipolar disorder, schizophrenia, or MDD, but there are no approved treatments that specifically target suicidal behavior. Lithium is approved for reducing the long-term risk of SI primarily because it reduces the risk of mood disorders associated with SI, but lithium has not been shown to be effective in acute settings.11 Clozapine is approved for reducing the long-term risk of recurrent suicide in patients with schizophrenia or schizoaffective disorder.4 Clozapine has not been shown to be effective in patients with mood disorders, which make up the majority of patients who attempt suicide.4 Additionally, both medications are plagued by the same barriers listed earlier, such as long time to effect (it takes an average 4 weeks to reach efficacy), lack of efficacy in acute settings, and AEs (eg, sleep problems, weight gain, and sexual dysfunction).9 Thus finding better pharmacologic interventions for suicidal patients is a priority for current research.

 

 

Ketamine

Recently, researchers have identified ketamine as a potential therapeutic option for depression and SI. A single ketamine infusion treatment has a rapid response, minimal AEs, and potentially long-lasting efficacy with SI, which would make it ideal for the treatment of acutely suicidal patients.4 Ketamine is an N-methyl-D-aspartate receptor (NMDAR) inhibitor that also has been found to be a weak μ- and κ-opioid receptor agonist and an inhibitor of the reuptake of serotonin, dopamine, and norepinephrine. Inhibition of the NMDAR results in analgesia, and ketamine is approved for the induction of anesthesia, pain relief, and sedation.12

Although AEs such as hallucinations and sedation create the potential for dangerous recreational use, ketamine is safely used in health care settings for a variety of indications. Effects are noted within 5 minutes of administration if given by infusion, and the main effects can last between 20 and 40 minutes.

Ketamine has a complex pharmacology and plays a role in other cell signaling mechanisms, but the significance of these additional mechanisms in the therapeutic effects of ketamine have only recently been elucidated. Preclinical studies indicate a probable NMDAR inhibition-independent mechanism responsible for the antidepressant response to ketamine.13,14 The complex associations with rapamycin signaling, eukaryotic elongation factor 2 dephosphorylation, increased synthesis of brain-derived neurotrophic factor, and activation of glutamatergic AMPA (α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid) receptors have been linked to its rapid antidepressant effect and ketamine’s induction of synaptogenesis within the limbic system.13,14

 

Clinical Research

Ketamine was studied as an adjunctive treatment to psychotherapy for addictions as far back as the 1970s.15 The available reports indicate a universally positive result, with increased rates of remission and decreased rates of relapse attributed to ketamine’s ability to alter one’s thought processes by reinforcing limbic-cortex interactions that facilitate the growth of more positive cognitive schemas and improved emotional attitudes about the self in support of the recovery process.15

Neurobiologic studies have shown that treatment with ketamine has a direct and immediate effect on neuronal pathways of the limbic system. It is known to regulate the mind’s reaction to positive stimuli by reversing the depressed subject’s blunted reaction to positive faces.16 This rapid normalization of the positive faces test is unique to ketamine infusion and is not seen in tests with traditional antidepressants.

In 2000, the first placebo-controlled trial using ketamine for treatment resistant depression (TRD) demonstrated the rapid antidepressant effects of a single dose of ketamine, but this study only looked at these effects for 1 week.17 In multiple double blind, placebo-controlled trials since then, IV infusion of ketamine was shown to be an effective intervention for TRD.13,18,19 More recently, a published investigation involving the treatment of MDD showed that ketamine in conjunction with a selective serotonin reuptake inhibitor (SSRI) accelerated and enhanced the effectiveness of the SSRI in reducing depressive symptoms.20

Based on the rapid resolution of depressive symptoms using ketamine, researchers have looked at its effect on suicidality as a secondary measure. A case study of a patient with severe depressive episodes and multiple previous suicidal attempts reported that the patient responded to a single dose of ketamine, described the experience as “being reborn,” and maintained complete remission of SI for the 6-month study period.21 In a larger study, 133 TRD patients received a single IV dose of ketamine with significant reductions in SI independent of depressive and anxiety symptoms.22

Depression Treatment

These results have led to an excitement for ketamine therapy as a novel treatment of depression, and off-label use by treatment centers now exists in several countries to aid those with TRD.23 This off-label use continues to be controversial, as research has yet to determine the safest most effective route and duration of treatment and whether the ketamine treatment AEs will exceed any accrued therapeutic benefit.13

The American Psychological Association Council of Research Task Force on Novel Biomarkers and Treatment critically examined the clinical evidence of ketamine use and has raised important concerns about the use of ketamine in the outpatient setting, administered in the absence of consensus therapeutic monitoring guidelines, and ambitiously marketed as a panacea for TRD.13,24 A study showed permanent impairment of brain function for both groups compared with monkeys treated with saline infusions.25 In 2016, the FDA gave fast-track approval for an intranasal ketamine that would make the treatment more easily available in the outpatient setting, but this could lead to certain patients developing a dependency on ketamine or engaging in its diversion for recreational use. There are case reports and anecdotes in the literature of patients and research subjects developing drug-seeking behaviors and overuse of ketamine.24 Additionally, the comorbidities associated with TRD and SI have not been fully evaluated. For instance, there is evidence that depressed patients with obsessive compulsive disorder may have worse outcomes that include delayed onset SI.26

There also is concern for the use of ketamine for chronic opioid users. The combination of ketamine with opioids may increase the response to the opioid in an otherwise drug tolerant patient, leading to risk of death by overdose in patients who have not increased their usual dose.27 However, this effect was noted only when ketamine and opioids were administered together, and the effect does not seem to last postinfusion.27

The challenges in treatment of TRD include finding an effective formulation—IV infusion of ketamine requires cardiovascular monitoring and is administered by anesthesiologists. The short duration of action for depression requires repeated infusions, and the frequency and quantity of infusions have not been determined. Efforts to find other NMDAR inhibitors (eg, memantine, nitrous oxide, D-cycloserine, and others) that match ketamine’s antidepressant efficacy but with easier delivery methods and fewer risks have thus far been unsuccessful.13 It is now believed that ketamine’s unique ability to activate intracellular signaling pathways linked to synaptic plasticity gives it the antidepressant function. Recent studies have further narrowed ketamine’s antidepressant function to the R- enantiomer of the ketamine metabolite, hydroxynorketamine.14 The nasal spray for ketamine is the S- enantiomer, which has better bioavailability but may have less antidepressant efficacy compared with the racemic mixture used in ketamine infusions.

 

 

Suicide Ideation Treatment

The many challenges faced by researchers and clinicians trying to develop ketamine treatment for TRD may not apply to the treatment of SI. Whereas repeated doses of ketamine cannot reliably produce sustained remission of depression, the few studies that have looked at the long-term effects of ketamine treatment on SI indicate the potential for long-term efficacy after a single IV infusion.21,22 Although treatment with IV infusions have additional costs and logistics, if it is found beneficial, it could be given in the emergency department (ED) prior to hospitalization and potentially lead to better outcomes.

In 2011, a small preliminary observational study of patients with depression and SI presenting to the ED indicated that SI was rapidly reduced following an infusion of ketamine.28 This study showed that both depressive symptoms and suicidality rapidly and significantly diminished within 40 minutes with no evidence of the recurrence of symptoms 10 days postadministration.

 

A more recent study used ketamine in a military field hospital to treat SI and also concluded that it could be effective and safe when administered in an ED setting. This preliminary study suggests that ketamine could be a safe and potentially effective medication for rapid reduction of depression and suicidality in a busy ED setting.29 These limited studies involving the use of ketamine in patients with SI show promise with long-term effectiveness. However, more research is needed to clarify whether the efficacy with SI will be similar to the clinical experience seen in TRD; a duration of effect limited to 2 weeks with recurrence after treatment discontinued.24

Conclusion

There has been a compelling accumulation of scientific data since 2000 to support the use of ketamine for the treatment of depression and SI. Ketamine use in patients with these diagnoses showed a rapid decrease of symptoms and minimal AEs among a significant number of patients.22,30

Although the initial findings involving the use of ketamine in suicidal patients are promising, the clinical use of ketamine needs further research, using larger sample sizes and exploring both the short-term and long-term effects of this medication. Researchers need to further establish the safe and effective route, point of care, and patient type that would best respond to this novel treatment. The initial evidence would suggest that health care providers have every right to be hopeful that ketamine will become the first pharmacologic treatment of acute SI in a majority of patients presenting to EDs, mental health clinics, community hospitals, and VA medical centers.

In 2014 the suicide rate in the U.S. was 13/100,000, the highest recorded in 28 years.1 Suicide is now considered the 10th leading cause of death for all ages, and the rate has increased every year from 2000 to 2014 among both women and men and in every age group except those aged ≥ 75 years.1-3 For those aged 15 to 44 years, suicide is among the top 3 causes of death worldwide.4-6

Background

In 2013, more than 490,000 hospital visits related to suicide attempts were reported in the U.S.4 Health care expenditures related to suicide are estimated at $56.9 billion in combined medical and work loss costs annually and an unmeasurable cost to the affected families.7 The mental health care community is desperate for ways to address this epidemic, and the National Academies of Medicine (NAM) has declared that research that directly addresses comparative effectiveness of treatment strategies following a suicide attempt should be a national priority.8

The most recent reports from 2014 indicate that the suicide rates are higher for male veterans than for male nonveterans (32.1 vs 20.9 per 100,000, respectively) and are much higher for female veterans than for female nonveterans (28.7 vs 5.2 per 100,000, respectively).3 Suicide rates also may be associated with veteran-specific comorbidities, such as higher rates of depression, anxiety, posttraumatic stress disorder (PTSD), and war-related trauma.3 According to the VHA, the suicide rate for veterans aged > 30 years also is rapidly increasing, and VHA has echoed the calls from NAM to make suicide prevention research a national priority.3

The VA has tried to stem the tide of suicides in veterans by implementing many advances in suicide prevention, including hiring suicide prevention coordinators at every VA hospital, enhanced monitoring, and the availability of 24-hour crisis hotline services. Yet the suicide rates for veterans continue to rise and remain higher than the rates in the general population.3

About 90% of deaths by suicide are by persons who have a treatable psychiatric disorder, most commonly a mood disorder, such as depression.4 However, most studies show that antidepressant therapy does not provide rapid or significant relief of suicidal ideation (SI).4 Therefore, the current standard of care for the treatment of acutely suicidal patients includes a combination of hospitalization, cognitive behavioral therapy or psychotherapy, case management, antidepressant medications, and electroconvulsive therapy (ECT).4 Even though these therapies have become more widely available over the past decade, rates of suicide continue to increase.1,4 These interventions have limited effectiveness in acute settings. Although both intensive outpatient follow-up and routine outpatient care have been studied in relation to the decrease of suicidal behavior, neither intervention has been shown to immediately reduce suicidal behavior significantly in patients.

Suicidality Interventions

Therapy and case management require patients to be well enough to make office visits and follow through with care for periods as long as 1 year, which is often not possible for individuals with severe depression.5 One-third of patients who attended 6 months of outpatient therapy consistently still met the criteria for major depressive disorder (MDD), a major risk for suicide attempt.9 Antidepressant medications take a minimum of 4 weeks to reach full efficacy, and many patients stop taking the medications before that point because of concern that the medication is not helping or because of adverse effects (AEs), such as sleep disturbance, sexual dysfunction, or weight gain.9

Electroconvulsive therapy has been shown to be an effective treatment for patients with depression and suicidal behavior, but adherence with 12 weeks of recommended therapy has been a barrier for this intervention. Additionally, ECT may not provide reduction in SI for 1 to 2 weeks.4,10 A review of research studies showed that nearly 50% of patients with high-expressed SI did not complete the prescribed amount of ECT due to the length of time to complete the recommended 12 sessions.10 Therefore, current treatment barriers for suicidal patients include: (1) long periods in treatment for therapy, medication, and ECT before any relief of symptoms is noted; (2) high recidivism rates for MDD symptoms and risk of suicide following treatment; and (3) high treatment dropout rates.

Pharmacologic treatments currently used in suicidal patients have not fared much better. Many have received FDA approval for treatment of associated mental health diagnoses such as bipolar disorder, schizophrenia, or MDD, but there are no approved treatments that specifically target suicidal behavior. Lithium is approved for reducing the long-term risk of SI primarily because it reduces the risk of mood disorders associated with SI, but lithium has not been shown to be effective in acute settings.11 Clozapine is approved for reducing the long-term risk of recurrent suicide in patients with schizophrenia or schizoaffective disorder.4 Clozapine has not been shown to be effective in patients with mood disorders, which make up the majority of patients who attempt suicide.4 Additionally, both medications are plagued by the same barriers listed earlier, such as long time to effect (it takes an average 4 weeks to reach efficacy), lack of efficacy in acute settings, and AEs (eg, sleep problems, weight gain, and sexual dysfunction).9 Thus finding better pharmacologic interventions for suicidal patients is a priority for current research.

 

 

Ketamine

Recently, researchers have identified ketamine as a potential therapeutic option for depression and SI. A single ketamine infusion treatment has a rapid response, minimal AEs, and potentially long-lasting efficacy with SI, which would make it ideal for the treatment of acutely suicidal patients.4 Ketamine is an N-methyl-D-aspartate receptor (NMDAR) inhibitor that also has been found to be a weak μ- and κ-opioid receptor agonist and an inhibitor of the reuptake of serotonin, dopamine, and norepinephrine. Inhibition of the NMDAR results in analgesia, and ketamine is approved for the induction of anesthesia, pain relief, and sedation.12

Although AEs such as hallucinations and sedation create the potential for dangerous recreational use, ketamine is safely used in health care settings for a variety of indications. Effects are noted within 5 minutes of administration if given by infusion, and the main effects can last between 20 and 40 minutes.

Ketamine has a complex pharmacology and plays a role in other cell signaling mechanisms, but the significance of these additional mechanisms in the therapeutic effects of ketamine have only recently been elucidated. Preclinical studies indicate a probable NMDAR inhibition-independent mechanism responsible for the antidepressant response to ketamine.13,14 The complex associations with rapamycin signaling, eukaryotic elongation factor 2 dephosphorylation, increased synthesis of brain-derived neurotrophic factor, and activation of glutamatergic AMPA (α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid) receptors have been linked to its rapid antidepressant effect and ketamine’s induction of synaptogenesis within the limbic system.13,14

 

Clinical Research

Ketamine was studied as an adjunctive treatment to psychotherapy for addictions as far back as the 1970s.15 The available reports indicate a universally positive result, with increased rates of remission and decreased rates of relapse attributed to ketamine’s ability to alter one’s thought processes by reinforcing limbic-cortex interactions that facilitate the growth of more positive cognitive schemas and improved emotional attitudes about the self in support of the recovery process.15

Neurobiologic studies have shown that treatment with ketamine has a direct and immediate effect on neuronal pathways of the limbic system. It is known to regulate the mind’s reaction to positive stimuli by reversing the depressed subject’s blunted reaction to positive faces.16 This rapid normalization of the positive faces test is unique to ketamine infusion and is not seen in tests with traditional antidepressants.

In 2000, the first placebo-controlled trial using ketamine for treatment resistant depression (TRD) demonstrated the rapid antidepressant effects of a single dose of ketamine, but this study only looked at these effects for 1 week.17 In multiple double blind, placebo-controlled trials since then, IV infusion of ketamine was shown to be an effective intervention for TRD.13,18,19 More recently, a published investigation involving the treatment of MDD showed that ketamine in conjunction with a selective serotonin reuptake inhibitor (SSRI) accelerated and enhanced the effectiveness of the SSRI in reducing depressive symptoms.20

Based on the rapid resolution of depressive symptoms using ketamine, researchers have looked at its effect on suicidality as a secondary measure. A case study of a patient with severe depressive episodes and multiple previous suicidal attempts reported that the patient responded to a single dose of ketamine, described the experience as “being reborn,” and maintained complete remission of SI for the 6-month study period.21 In a larger study, 133 TRD patients received a single IV dose of ketamine with significant reductions in SI independent of depressive and anxiety symptoms.22

Depression Treatment

These results have led to an excitement for ketamine therapy as a novel treatment of depression, and off-label use by treatment centers now exists in several countries to aid those with TRD.23 This off-label use continues to be controversial, as research has yet to determine the safest most effective route and duration of treatment and whether the ketamine treatment AEs will exceed any accrued therapeutic benefit.13

The American Psychological Association Council of Research Task Force on Novel Biomarkers and Treatment critically examined the clinical evidence of ketamine use and has raised important concerns about the use of ketamine in the outpatient setting, administered in the absence of consensus therapeutic monitoring guidelines, and ambitiously marketed as a panacea for TRD.13,24 A study showed permanent impairment of brain function for both groups compared with monkeys treated with saline infusions.25 In 2016, the FDA gave fast-track approval for an intranasal ketamine that would make the treatment more easily available in the outpatient setting, but this could lead to certain patients developing a dependency on ketamine or engaging in its diversion for recreational use. There are case reports and anecdotes in the literature of patients and research subjects developing drug-seeking behaviors and overuse of ketamine.24 Additionally, the comorbidities associated with TRD and SI have not been fully evaluated. For instance, there is evidence that depressed patients with obsessive compulsive disorder may have worse outcomes that include delayed onset SI.26

There also is concern for the use of ketamine for chronic opioid users. The combination of ketamine with opioids may increase the response to the opioid in an otherwise drug tolerant patient, leading to risk of death by overdose in patients who have not increased their usual dose.27 However, this effect was noted only when ketamine and opioids were administered together, and the effect does not seem to last postinfusion.27

The challenges in treatment of TRD include finding an effective formulation—IV infusion of ketamine requires cardiovascular monitoring and is administered by anesthesiologists. The short duration of action for depression requires repeated infusions, and the frequency and quantity of infusions have not been determined. Efforts to find other NMDAR inhibitors (eg, memantine, nitrous oxide, D-cycloserine, and others) that match ketamine’s antidepressant efficacy but with easier delivery methods and fewer risks have thus far been unsuccessful.13 It is now believed that ketamine’s unique ability to activate intracellular signaling pathways linked to synaptic plasticity gives it the antidepressant function. Recent studies have further narrowed ketamine’s antidepressant function to the R- enantiomer of the ketamine metabolite, hydroxynorketamine.14 The nasal spray for ketamine is the S- enantiomer, which has better bioavailability but may have less antidepressant efficacy compared with the racemic mixture used in ketamine infusions.

 

 

Suicide Ideation Treatment

The many challenges faced by researchers and clinicians trying to develop ketamine treatment for TRD may not apply to the treatment of SI. Whereas repeated doses of ketamine cannot reliably produce sustained remission of depression, the few studies that have looked at the long-term effects of ketamine treatment on SI indicate the potential for long-term efficacy after a single IV infusion.21,22 Although treatment with IV infusions have additional costs and logistics, if it is found beneficial, it could be given in the emergency department (ED) prior to hospitalization and potentially lead to better outcomes.

In 2011, a small preliminary observational study of patients with depression and SI presenting to the ED indicated that SI was rapidly reduced following an infusion of ketamine.28 This study showed that both depressive symptoms and suicidality rapidly and significantly diminished within 40 minutes with no evidence of the recurrence of symptoms 10 days postadministration.

 

A more recent study used ketamine in a military field hospital to treat SI and also concluded that it could be effective and safe when administered in an ED setting. This preliminary study suggests that ketamine could be a safe and potentially effective medication for rapid reduction of depression and suicidality in a busy ED setting.29 These limited studies involving the use of ketamine in patients with SI show promise with long-term effectiveness. However, more research is needed to clarify whether the efficacy with SI will be similar to the clinical experience seen in TRD; a duration of effect limited to 2 weeks with recurrence after treatment discontinued.24

Conclusion

There has been a compelling accumulation of scientific data since 2000 to support the use of ketamine for the treatment of depression and SI. Ketamine use in patients with these diagnoses showed a rapid decrease of symptoms and minimal AEs among a significant number of patients.22,30

Although the initial findings involving the use of ketamine in suicidal patients are promising, the clinical use of ketamine needs further research, using larger sample sizes and exploring both the short-term and long-term effects of this medication. Researchers need to further establish the safe and effective route, point of care, and patient type that would best respond to this novel treatment. The initial evidence would suggest that health care providers have every right to be hopeful that ketamine will become the first pharmacologic treatment of acute SI in a majority of patients presenting to EDs, mental health clinics, community hospitals, and VA medical centers.

References

1. Curtin SC, Warner MA, Hedegaard H. Increase in suicide in the United States 199-2014. NCHS data brief, no. 241. https://www.cdc.gov/nchs/data/data -briefs/db241.pdf. Published April 2016. Accessed August 3, 2017.

2. Nock MK, Borges G, Bromet EJ, Cha CB, Kessler RC, Lee S. Suicide and suicidal behavior. Epidemiol Rev. 2008;30(1):133-154.

3. U.S. Department of Veteran Affairs Office of Suicide Prevention. Suicide among veterans and other Americans 2001-2014. https://www.mentalhealth .va.gov/docs/2016suicidedatareport.pdf Published August 3, 2016. Accessed August 11, 2017.

4. Wilkinson ST, Sanacora G. Ketamine: a potential rapid-acting antisuicidal agent? Depress Anxiety. 2016;33(8):711-717.

5. Aleman A, Denys D. Mental health: a road map for suicide research and prevention. Nature. 2014;509(7501):421-423.

6. Griffiths JJ, Zarate CA, Jr, Rasimas JJ. Existing and novel biological therapeutics in suicide prevention. Am J Prev Med. 2014;47(3)(suppl 2):S195-S203.

7. Centers for Disease Control and Prevention. Leading causes of death reports, 1981-2015. https://www.cdc.gov/injury/wisqars/leading_causes_death.html. Updated February 19, 2017. Accessed August 14, 2017.

8. Institute of Medicine of the National Academies; Board on Health Care Services; Committee on Comparative Effectiveness Research Prioritization. Initial National Priorities for Comparative Effectiveness Research. Washington, DC: The National Academies Press; 2009.

9. Weinberger MI, Sirey JA, Bruce ML, Heo M, Papademetriou E, Meyers BS. Predictors of major depression six months after admission for outpatient treatment. Psychiatr Serv. 2008;59(10):1211-1215.

10. Kellner CH, Fink M, Knapp R, et al. Relief of expressed suicidal intent by ECT: a consortium for research in ECT study. Am J Psychiatry. 2005;162(5):977-982.

11. Lewitzka U, Jabs B, Fülle M, et al. Does lithium reduce acute suicidal ideation and behavior? A protocol for a randomized, placebo-controlled multicenter trial of lithium plus treatment as usual (TAU) in patients with suicidal major depressive episode. BMC Psychiatry. 2015;15:117.

12. Vadivelu N, Schermer E, Kodumudi V, Belani K, Urman RD, Kaye AD. Role of ketamine for analgesia in adults and children. J Anaesthesiol Clin Pharmacol. 2016;32(3):298-306.

13. Newport DJ, Carpenter LL, McDonald WM, et al; APA Council of Research Task Force on Novel Biomarkers and Treatments. Ketamine and other NMDA antagonists: early clinical trials and possible mechanisms in depression. Am J Psychiatry. 2015;172(10):950-966.

14. Zanos P, Moaddel R, Morris PJ, et al. NMDAR inhibition-independent antidepressant actions of ketamine metabolites. Nature. 2016;533(7604):481-486.

15. Krupitsky EM, Grinenko AY. Ketamine psychedelic therapy (KPT): a review of the results of ten years of research. J Psychoactive Drugs. 1997;29(2):165-183.

16. Murrough JW, Collins KA, Fields J, et al. Regulation of neural responses to emotion perception by ketamine in individuals with treatment-resistant major depressive disorder. Transl Psychiatry. 2015;5:e509.

17. Berman RM, Cappiello A, Anand A, et al. Antidepressant effects of ketamine in depressed patients. Biol Psychiatry. 2000;47(4):351-354.

18. Murrough JW, Iosifescu DV, Chang LC, et al. Antidepressant efficacy of ketamine in treatment-resistant major depression: a two-site randomized controlled trial. Am J Psychiatry. 2013;170(10):1134-1142.

19. Zarate CA Jr, Singh JB, Carlson PJ, et al. A randomized trial of an N-methyl-D-aspartate antagonist in treatment-resistant major depression. Arch Gen Psychiatry. 2006;63(8):856-864.

20. Hu YD, Xiang YT, Fang JX, et al. Single i.v. ketamine augmentation of newly initiated escitalopram for major depression: results from a randomized, placebo-controlled 4-week study. Psychol Med. 2016;46(3):623-635.

21. Aligeti S, Quinones M, Salazar R. Rapid resolution of suicidal behavior and depression with single low-dose ketamine intravenous push even after 6 months of follow-up. J Clin Psychopharmacol. 2014;34(4):533-535.

22. Ballard ED, Ionescu DF, Vande Voort JL, et al. Improvement in suicidal ideation after ketamine infusion: relationship to reductions in depression and anxiety. J Psychiatr Res. 2014;58:161-166.

23. Henderson TA. Practical application of the neuroregenerative properties of ketamine: real world treatment experience. Neural Regen Res. 2016;11(2):195-200.

24. Newport DJ, Schatzberg AF, Nemeroff CB. Whither ketamine as an antidepressant: panacea or toxin? Depress Anxiety. 2016;33(8):685-688.

25. Sun L, Li Q, Li Q, et al. Chronic ketamine exposure induces permanent impairment of brain functions in adolescent cynomolgus monkeys. Addict Biol. 2014;19(2):185-194.

26. Niciu MJ, Grunschel BD, Corlett PR, Pittenger C, Bloch MH. Two cases of delayed-onset suicidal ideation, dysphoria and anxiety after ketamine infusion in patients with obsessive-compulsive disorder and a history of major depressive disorder. J Psychopharmacol. 2013;27(7):651-654.

27. Huxtable CA, Roberts LJ, Somogyi AA, MacIntyre PE. Acute pain management in opioid-tolerant patients: a growing challenge. Anaesth Intensive Care. 2011;39(5):804-823.

28. Larkin GL, Beautrais AL. A preliminary naturalistic study of low-dose ketamine for depression and suicide ideation in the emergency department. Int J Neuropsychopharmacol. 2011;14(8):1127-1131.

29. Burger J, Capobianco M, Lovem R, et al. A double-blinded, randomized, placebo-controlled sub-dissociative dose ketamine pilot study in the treatment of acute depression and suicidality in a military emergency department setting. Mil Med. 2016;181(10):1195-1199.

30. Wan LB, Levitch CF, Perez AM, et al. Ketamine safety and tolerability in clinical trials for treatment-resistant depression. J Clin Psychiatry. 2015;76(3):247-252.

References

1. Curtin SC, Warner MA, Hedegaard H. Increase in suicide in the United States 199-2014. NCHS data brief, no. 241. https://www.cdc.gov/nchs/data/data -briefs/db241.pdf. Published April 2016. Accessed August 3, 2017.

2. Nock MK, Borges G, Bromet EJ, Cha CB, Kessler RC, Lee S. Suicide and suicidal behavior. Epidemiol Rev. 2008;30(1):133-154.

3. U.S. Department of Veteran Affairs Office of Suicide Prevention. Suicide among veterans and other Americans 2001-2014. https://www.mentalhealth .va.gov/docs/2016suicidedatareport.pdf Published August 3, 2016. Accessed August 11, 2017.

4. Wilkinson ST, Sanacora G. Ketamine: a potential rapid-acting antisuicidal agent? Depress Anxiety. 2016;33(8):711-717.

5. Aleman A, Denys D. Mental health: a road map for suicide research and prevention. Nature. 2014;509(7501):421-423.

6. Griffiths JJ, Zarate CA, Jr, Rasimas JJ. Existing and novel biological therapeutics in suicide prevention. Am J Prev Med. 2014;47(3)(suppl 2):S195-S203.

7. Centers for Disease Control and Prevention. Leading causes of death reports, 1981-2015. https://www.cdc.gov/injury/wisqars/leading_causes_death.html. Updated February 19, 2017. Accessed August 14, 2017.

8. Institute of Medicine of the National Academies; Board on Health Care Services; Committee on Comparative Effectiveness Research Prioritization. Initial National Priorities for Comparative Effectiveness Research. Washington, DC: The National Academies Press; 2009.

9. Weinberger MI, Sirey JA, Bruce ML, Heo M, Papademetriou E, Meyers BS. Predictors of major depression six months after admission for outpatient treatment. Psychiatr Serv. 2008;59(10):1211-1215.

10. Kellner CH, Fink M, Knapp R, et al. Relief of expressed suicidal intent by ECT: a consortium for research in ECT study. Am J Psychiatry. 2005;162(5):977-982.

11. Lewitzka U, Jabs B, Fülle M, et al. Does lithium reduce acute suicidal ideation and behavior? A protocol for a randomized, placebo-controlled multicenter trial of lithium plus treatment as usual (TAU) in patients with suicidal major depressive episode. BMC Psychiatry. 2015;15:117.

12. Vadivelu N, Schermer E, Kodumudi V, Belani K, Urman RD, Kaye AD. Role of ketamine for analgesia in adults and children. J Anaesthesiol Clin Pharmacol. 2016;32(3):298-306.

13. Newport DJ, Carpenter LL, McDonald WM, et al; APA Council of Research Task Force on Novel Biomarkers and Treatments. Ketamine and other NMDA antagonists: early clinical trials and possible mechanisms in depression. Am J Psychiatry. 2015;172(10):950-966.

14. Zanos P, Moaddel R, Morris PJ, et al. NMDAR inhibition-independent antidepressant actions of ketamine metabolites. Nature. 2016;533(7604):481-486.

15. Krupitsky EM, Grinenko AY. Ketamine psychedelic therapy (KPT): a review of the results of ten years of research. J Psychoactive Drugs. 1997;29(2):165-183.

16. Murrough JW, Collins KA, Fields J, et al. Regulation of neural responses to emotion perception by ketamine in individuals with treatment-resistant major depressive disorder. Transl Psychiatry. 2015;5:e509.

17. Berman RM, Cappiello A, Anand A, et al. Antidepressant effects of ketamine in depressed patients. Biol Psychiatry. 2000;47(4):351-354.

18. Murrough JW, Iosifescu DV, Chang LC, et al. Antidepressant efficacy of ketamine in treatment-resistant major depression: a two-site randomized controlled trial. Am J Psychiatry. 2013;170(10):1134-1142.

19. Zarate CA Jr, Singh JB, Carlson PJ, et al. A randomized trial of an N-methyl-D-aspartate antagonist in treatment-resistant major depression. Arch Gen Psychiatry. 2006;63(8):856-864.

20. Hu YD, Xiang YT, Fang JX, et al. Single i.v. ketamine augmentation of newly initiated escitalopram for major depression: results from a randomized, placebo-controlled 4-week study. Psychol Med. 2016;46(3):623-635.

21. Aligeti S, Quinones M, Salazar R. Rapid resolution of suicidal behavior and depression with single low-dose ketamine intravenous push even after 6 months of follow-up. J Clin Psychopharmacol. 2014;34(4):533-535.

22. Ballard ED, Ionescu DF, Vande Voort JL, et al. Improvement in suicidal ideation after ketamine infusion: relationship to reductions in depression and anxiety. J Psychiatr Res. 2014;58:161-166.

23. Henderson TA. Practical application of the neuroregenerative properties of ketamine: real world treatment experience. Neural Regen Res. 2016;11(2):195-200.

24. Newport DJ, Schatzberg AF, Nemeroff CB. Whither ketamine as an antidepressant: panacea or toxin? Depress Anxiety. 2016;33(8):685-688.

25. Sun L, Li Q, Li Q, et al. Chronic ketamine exposure induces permanent impairment of brain functions in adolescent cynomolgus monkeys. Addict Biol. 2014;19(2):185-194.

26. Niciu MJ, Grunschel BD, Corlett PR, Pittenger C, Bloch MH. Two cases of delayed-onset suicidal ideation, dysphoria and anxiety after ketamine infusion in patients with obsessive-compulsive disorder and a history of major depressive disorder. J Psychopharmacol. 2013;27(7):651-654.

27. Huxtable CA, Roberts LJ, Somogyi AA, MacIntyre PE. Acute pain management in opioid-tolerant patients: a growing challenge. Anaesth Intensive Care. 2011;39(5):804-823.

28. Larkin GL, Beautrais AL. A preliminary naturalistic study of low-dose ketamine for depression and suicide ideation in the emergency department. Int J Neuropsychopharmacol. 2011;14(8):1127-1131.

29. Burger J, Capobianco M, Lovem R, et al. A double-blinded, randomized, placebo-controlled sub-dissociative dose ketamine pilot study in the treatment of acute depression and suicidality in a military emergency department setting. Mil Med. 2016;181(10):1195-1199.

30. Wan LB, Levitch CF, Perez AM, et al. Ketamine safety and tolerability in clinical trials for treatment-resistant depression. J Clin Psychiatry. 2015;76(3):247-252.

Issue
Federal Practitioner - 34(9)
Issue
Federal Practitioner - 34(9)
Page Number
12-16
Page Number
12-16
Publications
Publications
Topics
Article Type
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Article PDF Media

Behavioral Health: Using Rating Scales in a Clinical Setting

Article Type
Changed
Thu, 03/28/2019 - 14:47
Display Headline
Behavioral Health: Using Rating Scales in a Clinical Setting
 

In the current health care environment, there is an increasing demand for objective assessment of disease states.1 This is particularly apparent in the realm of behavioral health, where documentation of outcomes lags that of other areas of medicine.

In 2012, the additional health care costs incurred by persons with mental health diagnoses were estimated to be $293 billion among commercially insured, Medicaid, and Medicare beneficiaries in the United States—a figure that is 273% higher than the cost for those without psychiatric diagnoses.2 Psychiatric and medical illnesses can be so tightly linked that accurate diagnosis and treatment of psychiatric disorders becomes essential to control medical illnesses. It is not surprising that there is increased scrutiny to the ways in which behavioral health care can be objectively assessed and monitored, and payers such as the Centers for Medicare and Medicaid Services increasingly require objective documentation of disease state improvement for payment.3

Support for objective assessment of disease derives from the collaborative care model. This model is designed to better integrate mental health and primary care (among other practices) by establishing the Patient-Centered Medical Home and emphasizing screening and monitoring patient-reported outcomes over time to assess treatment response.4 This approach, which is endorsed by the American Psychiatric Association, is associated with significant improvements in outcomes compared with usual care.5 It tracks patient progress using validated clinical rating scales and other screening tools (eg, Patient Health Questionnaire [PHQ-9] for depression), an approach that is analogous to how patients with type 2 diabetes are monitored by A1C lab tests.6 An extensive body of research supports the impact of this approach on treatment. A 2012 Cochrane review associated collaborative care with significant improvements in depression and anxiety outcomes compared with usual treatment.7

Despite these findings, a recent Kennedy Forum brief asserts that behavioral health is characterized by a "lack of systematic measurement to determine whether patients are responding to treatment."8 That same brief points to the many validated, easy-to-administer rating scales and screening tools that can reliably measure the frequency and severity of psychiatric symptoms over time, and likens the lack of their use to "treating high blood pressure without using a blood pressure cuff to measure if a patient's blood pressure is improving."8 In fact, it is estimated that only 18% of psychiatrists and 11% of psychologists use rating scales routinely.9,10 This lack of use denies clinicians important information that can help detect deterioration or lack of improvement in their patients; implementing these scales in primary care can help early detection of behavioral health problems.

Behavioral health is replete with rating scales and screening tools, and the number of competing scales can make choosing a measure difficult.1 Nonetheless, not all scales are appropriate for clinical use; many are designed for research, for instance, and are lengthy and difficult to administer.

Let's review a number of rating scales that are brief, useful, and easy to administer. A framework for the screening tools addressed in this article is available on the federally funded Center for Integrated Health Solutions website (www.integration.samhsa.gov). This site promotes the use of tools designed to assist in screening and monitoring for depression, anxiety, bipolar disorder, substance use, and suicidality.11

QUALITY CRITERIA FOR RATING SCALES

The quality of a rating scale is determined by the following attributes.

Objectivity. The ability of a scale to obtain the same results, regardless of who administers, analyzes, or interprets it.

Reliability. The ability of a scale to convey consistent and reproducible information across time, patients, and raters.

Validity. The degree to which the scale measures what it is supposed to measure (eg, depressive symptoms). Sensitivity and specificity are measures of validity and provide additional information about the rating scale; namely, whether the scale can detect the presence of a disease (sensitivity) and whether it detects only that disease or condition and not another (specificity).

Establishment of norms. Whether a scale provides reference values for different clinical groups.

Practicability. The resources required to administer the assessment instrument in terms of time, staff, and material.12

In addition to meeting these quality criteria, selection of a scale can be based on whether it is self-rated or observer-rated. Advantages to self-rated scales, such as the PHQ-9, Mood Disorder Questionnaire (MDQ), and Generalized Anxiety Disorder 7-item (GAD-7) scale, are their practicability—they are easy to administer and don't require much time—and their use in evaluating and raising awareness of subjective states.

 

 

 

However, reliability may be a concern, as some patients may lack insight or exaggerate or mask symptoms when completing such scales.13 Both observer- and self-rated scales can be used together to minimize bias, identify symptoms that might have been missed/not addressed in the clinical interview, and drive clinical decision-making. Both can also help patients communicate with their providers and make them feel more involved in clinical decision-making.8

ENDORSED RATING SCALES

The following scales have met many of the quality criteria described here and are endorsed by the government payer system. They can easily be incorporated into clinical practice and will provide useful clinical information that can assist in diagnosis and monitoring patient outcomes.

Patient Health Questionnaire

PHQ-9 is a nine-item self-report questionnaire that can help to detect depression and supplement a thorough mental health interview. It scores the nine DSM-IV criteria for depression on a scale of 0 (not at all) to 3 (nearly every day). It is a public resource that is easy to find online, available without cost in several languages, and takes just a few minutes to complete.14

PHQ-9 has shown excellent test-retest reliability in screening for depression, and normative data on the instrument's use are available in various clinical populations.15 Research has shown that as PHQ-9 depression scores increase, functional status decreases, while depressive symptoms, sick days, and health care utilization increase.15 In one study, a PHQ-9 score of ≥ 10 had 88% sensitivity and specificity for detecting depression, with scores of 5, 10, 15, and 20 indicating mild, moderate, moderately severe, and severe depression, respectively.16 In addition to its use as a screening tool, PHQ-9 is a responsive and reliable measure of depression treatment outcomes.17

Mood Disorder Questionnaire

MDQ is another brief, self-report questionnaire that is available online. It is designed to identify and monitor patients who are likely to meet diagnostic criteria for bipolar disorder.18,19

The first question on the MDQ asks if the patient has experienced any of 13 common mood and behavior symptoms. The second question asks if these symptoms have ever occurred at the same time, and the third asks the degree to which the patient finds the symptoms to be problematic. The remaining two questions provide additional clinical information, addressing family history of manic-depressive illness or bipolar disorder and whether a diagnosis of either disorder has been made.

The MDQ has shown validity in assessing bipolar disorder symptoms in a general population, although recent research suggests that imprecise recall bias may limit its reliability in detecting hypomanic episodes earlier in life.20,21 Nonetheless, its specificity of > 97% means that it will effectively screen out just about all true negatives.18

Generalized Anxiety Disorder 7-item scale

The GAD-7 scale is a brief, self-administered questionnaire for screening and measuring severity of GAD.22 It asks patients to rate seven items that represent problems with general anxiety and scores each item on a scale of 0 (not at all) to 3 (nearly every day). Similar to the other measures, it is easily accessible online.

Research evidence supports the reliability and validity of GAD-7 as a measure of anxiety in the general population. Sensitivity and specificity are 89% and 82%, respectively. Normative data for age- and sex-specific subgroups support its use across age groups and in both males and females.23 The GAD-7 performs well for detecting and monitoring not only GAD but also panic disorder, social anxiety disorder, and posttraumatic stress disorder.24

CAGE questionnaire for detection of substance use

The CAGE questionnaire is a widely used screening tool that was originally developed to detect alcohol abuse but has been adapted to assess other substance abuse.25,26 The omission of substance abuse from diagnostic consideration can have a major effect on quality of care, because substance abuse can be the underlying cause of other diseases. Therefore, routine administration of this instrument in clinical practice can lead to better understanding and monitoring of patient health.27

Similar to other instruments, CAGE is free and available online.27 It contains four simple questions, with 1 point assigned to each positive answer (see Table); the simple mnemonic makes the questions easy to ­remember and to administer in a clinical setting.

CAGE has demonstrated validity, with one study determining that scores ≥ 2 had a specificity and sensitivity of 76% and 93%, respectively, for identifying excessive drinking, and a specificity and sensitivity of 77% and 91%, respectively, for identifying alcohol abuse.28

 

 

 

Columbia Suicide Severity Rating Scale (C-SSRS)

C-SSRS was developed by researchers at Columbia University to assess the severity of and track changes over time in suicidal ideation and behavior. C-SSRS is two pages and takes only a few minutes to administer; however, it also may be completed as a self-report measure. The questions are phrased in an interview format, and while clinicians are encouraged to receive training prior to its administration, specific training in mental health is not required.

The "Lifetime/Recent" version allows practitioners to gather lifetime history of suicidality as well as any recent suicidal ideation and/or behavior, whereas the "Since Last Visit" version of the scale assesses suicidality in patients who have completed at least one Lifetime/Recent C-SSRS assessment. A truncated, six-item "Screener" version is typically used in emergency situations. A risk assessment can be added to either the Full or Screener version to summarize the answers from C-SSRS and document risk and protective factors.29

Several studies have found C-SSRS to be reliable and valid for identifying suicide risk in children and adults.30,31USA Today reported that an individual exhibiting even a single behavior identified by the scale is eight to 10 times more likely to complete suicide.32 In addition, the C-SSRS has helped reduce the suicide rate by 65% in one of the largest providers of community-based behavioral health care in the United States.32

USING SCALES TO AUGMENT CARE

Each of the scales described in this article can easily be incorporated into clinical practice. The information the scales provide can be used to track progression of symptoms and effectiveness of treatment. Although rating scales should never be used alone to establish a diagnosis or clinical treatment plan, they can and should be used to augment information from the clinician's assessment and follow-up interviews.5

References

1. McDowell I. Measuring Health: A Guide to Rating Scales and Questionnaires. 3rd ed. New York, NY: Oxford University Press; 2006.
2. Kennedy Forum. Fixing behavioral health care in America: a national call for integrating and coordinating specialty behavioral health care with the medical system. http://thekennedyforum-dot-org.s3.amazonaws.com/documents/KennedyForum-BehavioralHealth_FINAL_3.pdf. Accessed August 14, 2017. 
3. The Office of the National Coordinator for Health Information Technology. Behavioral health (BH) Clinical Quality Measures (CQMs) Program initiatives. www.healthit.gov/sites/default/files/pdf/2012-09-27-behavioral-health-clinical-quality-measures-program-initiatives-public-forum.pdf. Accessed August 14, 2017.
4. Unutzer J, Harbin H, Schoenbaum M. The collaborative care model: an approach for integrating physical and mental health care in Medicaid health homes. www.medicaid.gov/State-Resource-Center/Medicaid-State-Technical-Assistance/Health-Homes-Technical-Assistance/Downloads/HH-IRC-Collaborative-5-13.pdf. Accessed August 14, 2017. 
5. World Group On Psychiatric Evaluation; American Psychiatric Association Steering Committee On Practice Guidelines. Practice guideline for the psychiatric evaluation of adults. 2nd ed. http://psychiatryonline.org/pb/assets/raw/sitewide/practice_guidelines/guidelines/psychevaladults.pdf. Accessed August 14, 2017. 
6. Melek S, Norris D, Paulus J. Economic Impact of Integrated Medical-Behavioral Healthcare: Implications for Psychiatry. Denver, CO: Milliman, Inc; 2014. 
7. Archer J, Bower P, Gilbody S, et al. Collaborative care for depression and anxiety problems. Cochrane Database Syst Rev. 2012;10:CD006525. 
8. Kennedy Forum. Fixing behavioral health care in America: a national call for measurement-based care.  www.thekennedyforum.org/a-national-call-for-measurement-based-care/. Accessed August 14, 2017.
9. Zimmerman M, McGlinchey JB. Why don't psychiatrists use scales to measure outcome when treating depressed patients? J Clin Psychiatry. 2008;69(12):1916-1919. 
10. Hatfield D, McCullough L, Frantz SH, et al. Do we know when our clients get worse? An investigation of therapists' ability to detect negative client change. Clin Psychol Psychother. 2010;17(1):25-32.
11. SAMHSA-HRSA Center for Integrated Solutions. Screening tools. www.integration.samhsa.gov/clinical-practice/screening-tools. Accessed August 14, 2017. 
12. Moller HJ. Standardised rating scales in psychiatry: methodological basis, their possibilities and limitations and descriptions of important rating scales. World J Biol Psychiatry. 2009;10(1):6-26.
13. Sajatovic M, Ramirez LF. Rating Scales in Mental Health. 2nd ed. Hudson, OH: Lexi-Comp; 2003.
14. Patient Health Questionnaire-9 (PHQ-9). www.agencymeddirectors.wa.gov/files/AssessmentTools/14-PHQ-9%20overview.pdf. Accessed August 14, 2017.
15. Patient Health Questionnaire-9 (PHQ-9). Rehab Measures Web site. www.rehabmeasures.org/Lists/RehabMeasures/DispForm.aspx?ID=954. Accessed August 14, 2017. 
16. Kroenke K, Spitzer RL, Williams JB. The PHQ-9: validity of a brief depression severity measure. J Gen Intern Med. 2001;16(9):606-613. 
17. Löwe B, Unützer J, Callahan CM, et al. Monitoring depression treatment outcomes with the Patient Health Questionnaire-9. Med Care. 2004;42(12):1194-1201.
18. Ketter TA. Strategies for monitoring outcomes in patients with bipolar disorder. Prim Care Companion J Clin Psychiatry. 2010;12(suppl 1):10-16.
19. The Mood Disorder Questionnaire. University of Texas Medical Branch. www.dbsalliance.org/pdfs/MDQ.pdf. Accessed August 14, 2017.
20. Hirschfeld RM, Holzer C, Calabrese JR, et al. Validity of the Mood Disorder Questionnaire: a general population study. Am J Psychiatry. 2003;160(1):178-180.
21. Boschloo L, Nolen WA, Spijker AT, et al. The Mood Disorder Questionnaire (MDQ) for detecting (hypo) manic episodes: its validity and impact of recall bias. J Affect Disord. 2013;151(1):203-208.
22. Spitzer RL, Kroenke K, Williams JB, et al. A brief measure for assessing generalized anxiety disorder: the GAD-7. Arch Intern Med. 2006;166(10):1092-1097.
23. Lowe B, Decker O, Müller S, et al. Validation and standardization of the Generalized Anxiety Disorder Screener (GAD-7) in the general population. Med Care. 2008;46(3):266-274. 
24. Kroenke K, Spitzer RL, Williams JB, et al. Anxiety disorders in primary care: prevalence, impairment, comorbidity, and detection. Ann Intern Med. 2007;146(5):317-325.
25. Ewing JA. Detecting alcoholism. The CAGE Questionnaire. JAMA. 1984;252(14):1905-1907.
26. CAGE substance abuse screening tool. Johns Hopkins Medicine. www.hopkinsmedicine.org/johns_hopkins_healthcare/downloads/cage%20substance%20screening%20tool.pdf. Accessed August 14, 2017.
27. O'Brien CP. The CAGE questionnaire for detection of alcoholism: a remarkably useful but simple tool. JAMA. 2008;300(17):2054-2056. 
28. Bernadt MW, Mumford J, Taylor C, et al. Comparison of questionnaire and laboratory tests in the detection of excessive drinking and alcoholism. Lancet. 1982;1(8267):325-328. 
29. Columbia Suicide-Severity Rating Scale (CS-SRS). http://cssrs.columbia.edu/the-columbia-scale-c-ssrs/cssrs-for-communities-and-healthcare/#filter=.general-use.english. Accessed August 14, 2017. 
30. Mundt JC, Greist JH, Jefferson JW, et al. Prediction of suicidal behavior in clinical research by lifetime suicidal ideation and behavior ascertained by the electronic Columbia-Suicide Severity Rating Scale. J Clin Psychiatry. 2013;74(9):887-893.
31. Posner K, Brown GK, Stanley B, et al. The Columbia-Suicide Severity Rating Scale: initial validity and internal consistency findings from three multisite studies with adolescents and adults. Am J Psychiatry. 2011;168(12):1266-1277. 
32. Esposito L. Suicide checklist spots people at highest risk. USA Today. http://usatoday30.usatoday.com/news/health/story/health/story/2011-11-09/Suicide-checklist-spots-peo ple-at-highest-risk/51135944/1. Accessed August 14, 2017.

Article PDF
Author and Disclosure Information

Julie M. Wood is a Consultant Medical Liaison of Neuroscience at Lilly USA, LLC, in Indianapolis. Sanjay Gupta is a Clinical Professor in the Department of Psychiatry at SUNY Upstate Medical University, Syracuse, and at SUNY Buffalo School of Medicine and Biomedical Sciences.

The authors report no financial relationships with any company whose products are mentioned in this article or with manufacturers of competing products.

This article was originally published in Current Psychiatry (2017;16[2]:21-25).

Issue
Clinician Reviews - 27(9)
Publications
Topics
Page Number
28-31,33
Sections
Author and Disclosure Information

Julie M. Wood is a Consultant Medical Liaison of Neuroscience at Lilly USA, LLC, in Indianapolis. Sanjay Gupta is a Clinical Professor in the Department of Psychiatry at SUNY Upstate Medical University, Syracuse, and at SUNY Buffalo School of Medicine and Biomedical Sciences.

The authors report no financial relationships with any company whose products are mentioned in this article or with manufacturers of competing products.

This article was originally published in Current Psychiatry (2017;16[2]:21-25).

Author and Disclosure Information

Julie M. Wood is a Consultant Medical Liaison of Neuroscience at Lilly USA, LLC, in Indianapolis. Sanjay Gupta is a Clinical Professor in the Department of Psychiatry at SUNY Upstate Medical University, Syracuse, and at SUNY Buffalo School of Medicine and Biomedical Sciences.

The authors report no financial relationships with any company whose products are mentioned in this article or with manufacturers of competing products.

This article was originally published in Current Psychiatry (2017;16[2]:21-25).

Article PDF
Article PDF
Related Articles
 

In the current health care environment, there is an increasing demand for objective assessment of disease states.1 This is particularly apparent in the realm of behavioral health, where documentation of outcomes lags that of other areas of medicine.

In 2012, the additional health care costs incurred by persons with mental health diagnoses were estimated to be $293 billion among commercially insured, Medicaid, and Medicare beneficiaries in the United States—a figure that is 273% higher than the cost for those without psychiatric diagnoses.2 Psychiatric and medical illnesses can be so tightly linked that accurate diagnosis and treatment of psychiatric disorders becomes essential to control medical illnesses. It is not surprising that there is increased scrutiny to the ways in which behavioral health care can be objectively assessed and monitored, and payers such as the Centers for Medicare and Medicaid Services increasingly require objective documentation of disease state improvement for payment.3

Support for objective assessment of disease derives from the collaborative care model. This model is designed to better integrate mental health and primary care (among other practices) by establishing the Patient-Centered Medical Home and emphasizing screening and monitoring patient-reported outcomes over time to assess treatment response.4 This approach, which is endorsed by the American Psychiatric Association, is associated with significant improvements in outcomes compared with usual care.5 It tracks patient progress using validated clinical rating scales and other screening tools (eg, Patient Health Questionnaire [PHQ-9] for depression), an approach that is analogous to how patients with type 2 diabetes are monitored by A1C lab tests.6 An extensive body of research supports the impact of this approach on treatment. A 2012 Cochrane review associated collaborative care with significant improvements in depression and anxiety outcomes compared with usual treatment.7

Despite these findings, a recent Kennedy Forum brief asserts that behavioral health is characterized by a "lack of systematic measurement to determine whether patients are responding to treatment."8 That same brief points to the many validated, easy-to-administer rating scales and screening tools that can reliably measure the frequency and severity of psychiatric symptoms over time, and likens the lack of their use to "treating high blood pressure without using a blood pressure cuff to measure if a patient's blood pressure is improving."8 In fact, it is estimated that only 18% of psychiatrists and 11% of psychologists use rating scales routinely.9,10 This lack of use denies clinicians important information that can help detect deterioration or lack of improvement in their patients; implementing these scales in primary care can help early detection of behavioral health problems.

Behavioral health is replete with rating scales and screening tools, and the number of competing scales can make choosing a measure difficult.1 Nonetheless, not all scales are appropriate for clinical use; many are designed for research, for instance, and are lengthy and difficult to administer.

Let's review a number of rating scales that are brief, useful, and easy to administer. A framework for the screening tools addressed in this article is available on the federally funded Center for Integrated Health Solutions website (www.integration.samhsa.gov). This site promotes the use of tools designed to assist in screening and monitoring for depression, anxiety, bipolar disorder, substance use, and suicidality.11

QUALITY CRITERIA FOR RATING SCALES

The quality of a rating scale is determined by the following attributes.

Objectivity. The ability of a scale to obtain the same results, regardless of who administers, analyzes, or interprets it.

Reliability. The ability of a scale to convey consistent and reproducible information across time, patients, and raters.

Validity. The degree to which the scale measures what it is supposed to measure (eg, depressive symptoms). Sensitivity and specificity are measures of validity and provide additional information about the rating scale; namely, whether the scale can detect the presence of a disease (sensitivity) and whether it detects only that disease or condition and not another (specificity).

Establishment of norms. Whether a scale provides reference values for different clinical groups.

Practicability. The resources required to administer the assessment instrument in terms of time, staff, and material.12

In addition to meeting these quality criteria, selection of a scale can be based on whether it is self-rated or observer-rated. Advantages to self-rated scales, such as the PHQ-9, Mood Disorder Questionnaire (MDQ), and Generalized Anxiety Disorder 7-item (GAD-7) scale, are their practicability—they are easy to administer and don't require much time—and their use in evaluating and raising awareness of subjective states.

 

 

 

However, reliability may be a concern, as some patients may lack insight or exaggerate or mask symptoms when completing such scales.13 Both observer- and self-rated scales can be used together to minimize bias, identify symptoms that might have been missed/not addressed in the clinical interview, and drive clinical decision-making. Both can also help patients communicate with their providers and make them feel more involved in clinical decision-making.8

ENDORSED RATING SCALES

The following scales have met many of the quality criteria described here and are endorsed by the government payer system. They can easily be incorporated into clinical practice and will provide useful clinical information that can assist in diagnosis and monitoring patient outcomes.

Patient Health Questionnaire

PHQ-9 is a nine-item self-report questionnaire that can help to detect depression and supplement a thorough mental health interview. It scores the nine DSM-IV criteria for depression on a scale of 0 (not at all) to 3 (nearly every day). It is a public resource that is easy to find online, available without cost in several languages, and takes just a few minutes to complete.14

PHQ-9 has shown excellent test-retest reliability in screening for depression, and normative data on the instrument's use are available in various clinical populations.15 Research has shown that as PHQ-9 depression scores increase, functional status decreases, while depressive symptoms, sick days, and health care utilization increase.15 In one study, a PHQ-9 score of ≥ 10 had 88% sensitivity and specificity for detecting depression, with scores of 5, 10, 15, and 20 indicating mild, moderate, moderately severe, and severe depression, respectively.16 In addition to its use as a screening tool, PHQ-9 is a responsive and reliable measure of depression treatment outcomes.17

Mood Disorder Questionnaire

MDQ is another brief, self-report questionnaire that is available online. It is designed to identify and monitor patients who are likely to meet diagnostic criteria for bipolar disorder.18,19

The first question on the MDQ asks if the patient has experienced any of 13 common mood and behavior symptoms. The second question asks if these symptoms have ever occurred at the same time, and the third asks the degree to which the patient finds the symptoms to be problematic. The remaining two questions provide additional clinical information, addressing family history of manic-depressive illness or bipolar disorder and whether a diagnosis of either disorder has been made.

The MDQ has shown validity in assessing bipolar disorder symptoms in a general population, although recent research suggests that imprecise recall bias may limit its reliability in detecting hypomanic episodes earlier in life.20,21 Nonetheless, its specificity of > 97% means that it will effectively screen out just about all true negatives.18

Generalized Anxiety Disorder 7-item scale

The GAD-7 scale is a brief, self-administered questionnaire for screening and measuring severity of GAD.22 It asks patients to rate seven items that represent problems with general anxiety and scores each item on a scale of 0 (not at all) to 3 (nearly every day). Similar to the other measures, it is easily accessible online.

Research evidence supports the reliability and validity of GAD-7 as a measure of anxiety in the general population. Sensitivity and specificity are 89% and 82%, respectively. Normative data for age- and sex-specific subgroups support its use across age groups and in both males and females.23 The GAD-7 performs well for detecting and monitoring not only GAD but also panic disorder, social anxiety disorder, and posttraumatic stress disorder.24

CAGE questionnaire for detection of substance use

The CAGE questionnaire is a widely used screening tool that was originally developed to detect alcohol abuse but has been adapted to assess other substance abuse.25,26 The omission of substance abuse from diagnostic consideration can have a major effect on quality of care, because substance abuse can be the underlying cause of other diseases. Therefore, routine administration of this instrument in clinical practice can lead to better understanding and monitoring of patient health.27

Similar to other instruments, CAGE is free and available online.27 It contains four simple questions, with 1 point assigned to each positive answer (see Table); the simple mnemonic makes the questions easy to ­remember and to administer in a clinical setting.

CAGE has demonstrated validity, with one study determining that scores ≥ 2 had a specificity and sensitivity of 76% and 93%, respectively, for identifying excessive drinking, and a specificity and sensitivity of 77% and 91%, respectively, for identifying alcohol abuse.28

 

 

 

Columbia Suicide Severity Rating Scale (C-SSRS)

C-SSRS was developed by researchers at Columbia University to assess the severity of and track changes over time in suicidal ideation and behavior. C-SSRS is two pages and takes only a few minutes to administer; however, it also may be completed as a self-report measure. The questions are phrased in an interview format, and while clinicians are encouraged to receive training prior to its administration, specific training in mental health is not required.

The "Lifetime/Recent" version allows practitioners to gather lifetime history of suicidality as well as any recent suicidal ideation and/or behavior, whereas the "Since Last Visit" version of the scale assesses suicidality in patients who have completed at least one Lifetime/Recent C-SSRS assessment. A truncated, six-item "Screener" version is typically used in emergency situations. A risk assessment can be added to either the Full or Screener version to summarize the answers from C-SSRS and document risk and protective factors.29

Several studies have found C-SSRS to be reliable and valid for identifying suicide risk in children and adults.30,31USA Today reported that an individual exhibiting even a single behavior identified by the scale is eight to 10 times more likely to complete suicide.32 In addition, the C-SSRS has helped reduce the suicide rate by 65% in one of the largest providers of community-based behavioral health care in the United States.32

USING SCALES TO AUGMENT CARE

Each of the scales described in this article can easily be incorporated into clinical practice. The information the scales provide can be used to track progression of symptoms and effectiveness of treatment. Although rating scales should never be used alone to establish a diagnosis or clinical treatment plan, they can and should be used to augment information from the clinician's assessment and follow-up interviews.5

 

In the current health care environment, there is an increasing demand for objective assessment of disease states.1 This is particularly apparent in the realm of behavioral health, where documentation of outcomes lags that of other areas of medicine.

In 2012, the additional health care costs incurred by persons with mental health diagnoses were estimated to be $293 billion among commercially insured, Medicaid, and Medicare beneficiaries in the United States—a figure that is 273% higher than the cost for those without psychiatric diagnoses.2 Psychiatric and medical illnesses can be so tightly linked that accurate diagnosis and treatment of psychiatric disorders becomes essential to control medical illnesses. It is not surprising that there is increased scrutiny to the ways in which behavioral health care can be objectively assessed and monitored, and payers such as the Centers for Medicare and Medicaid Services increasingly require objective documentation of disease state improvement for payment.3

Support for objective assessment of disease derives from the collaborative care model. This model is designed to better integrate mental health and primary care (among other practices) by establishing the Patient-Centered Medical Home and emphasizing screening and monitoring patient-reported outcomes over time to assess treatment response.4 This approach, which is endorsed by the American Psychiatric Association, is associated with significant improvements in outcomes compared with usual care.5 It tracks patient progress using validated clinical rating scales and other screening tools (eg, Patient Health Questionnaire [PHQ-9] for depression), an approach that is analogous to how patients with type 2 diabetes are monitored by A1C lab tests.6 An extensive body of research supports the impact of this approach on treatment. A 2012 Cochrane review associated collaborative care with significant improvements in depression and anxiety outcomes compared with usual treatment.7

Despite these findings, a recent Kennedy Forum brief asserts that behavioral health is characterized by a "lack of systematic measurement to determine whether patients are responding to treatment."8 That same brief points to the many validated, easy-to-administer rating scales and screening tools that can reliably measure the frequency and severity of psychiatric symptoms over time, and likens the lack of their use to "treating high blood pressure without using a blood pressure cuff to measure if a patient's blood pressure is improving."8 In fact, it is estimated that only 18% of psychiatrists and 11% of psychologists use rating scales routinely.9,10 This lack of use denies clinicians important information that can help detect deterioration or lack of improvement in their patients; implementing these scales in primary care can help early detection of behavioral health problems.

Behavioral health is replete with rating scales and screening tools, and the number of competing scales can make choosing a measure difficult.1 Nonetheless, not all scales are appropriate for clinical use; many are designed for research, for instance, and are lengthy and difficult to administer.

Let's review a number of rating scales that are brief, useful, and easy to administer. A framework for the screening tools addressed in this article is available on the federally funded Center for Integrated Health Solutions website (www.integration.samhsa.gov). This site promotes the use of tools designed to assist in screening and monitoring for depression, anxiety, bipolar disorder, substance use, and suicidality.11

QUALITY CRITERIA FOR RATING SCALES

The quality of a rating scale is determined by the following attributes.

Objectivity. The ability of a scale to obtain the same results, regardless of who administers, analyzes, or interprets it.

Reliability. The ability of a scale to convey consistent and reproducible information across time, patients, and raters.

Validity. The degree to which the scale measures what it is supposed to measure (eg, depressive symptoms). Sensitivity and specificity are measures of validity and provide additional information about the rating scale; namely, whether the scale can detect the presence of a disease (sensitivity) and whether it detects only that disease or condition and not another (specificity).

Establishment of norms. Whether a scale provides reference values for different clinical groups.

Practicability. The resources required to administer the assessment instrument in terms of time, staff, and material.12

In addition to meeting these quality criteria, selection of a scale can be based on whether it is self-rated or observer-rated. Advantages to self-rated scales, such as the PHQ-9, Mood Disorder Questionnaire (MDQ), and Generalized Anxiety Disorder 7-item (GAD-7) scale, are their practicability—they are easy to administer and don't require much time—and their use in evaluating and raising awareness of subjective states.

 

 

 

However, reliability may be a concern, as some patients may lack insight or exaggerate or mask symptoms when completing such scales.13 Both observer- and self-rated scales can be used together to minimize bias, identify symptoms that might have been missed/not addressed in the clinical interview, and drive clinical decision-making. Both can also help patients communicate with their providers and make them feel more involved in clinical decision-making.8

ENDORSED RATING SCALES

The following scales have met many of the quality criteria described here and are endorsed by the government payer system. They can easily be incorporated into clinical practice and will provide useful clinical information that can assist in diagnosis and monitoring patient outcomes.

Patient Health Questionnaire

PHQ-9 is a nine-item self-report questionnaire that can help to detect depression and supplement a thorough mental health interview. It scores the nine DSM-IV criteria for depression on a scale of 0 (not at all) to 3 (nearly every day). It is a public resource that is easy to find online, available without cost in several languages, and takes just a few minutes to complete.14

PHQ-9 has shown excellent test-retest reliability in screening for depression, and normative data on the instrument's use are available in various clinical populations.15 Research has shown that as PHQ-9 depression scores increase, functional status decreases, while depressive symptoms, sick days, and health care utilization increase.15 In one study, a PHQ-9 score of ≥ 10 had 88% sensitivity and specificity for detecting depression, with scores of 5, 10, 15, and 20 indicating mild, moderate, moderately severe, and severe depression, respectively.16 In addition to its use as a screening tool, PHQ-9 is a responsive and reliable measure of depression treatment outcomes.17

Mood Disorder Questionnaire

MDQ is another brief, self-report questionnaire that is available online. It is designed to identify and monitor patients who are likely to meet diagnostic criteria for bipolar disorder.18,19

The first question on the MDQ asks if the patient has experienced any of 13 common mood and behavior symptoms. The second question asks if these symptoms have ever occurred at the same time, and the third asks the degree to which the patient finds the symptoms to be problematic. The remaining two questions provide additional clinical information, addressing family history of manic-depressive illness or bipolar disorder and whether a diagnosis of either disorder has been made.

The MDQ has shown validity in assessing bipolar disorder symptoms in a general population, although recent research suggests that imprecise recall bias may limit its reliability in detecting hypomanic episodes earlier in life.20,21 Nonetheless, its specificity of > 97% means that it will effectively screen out just about all true negatives.18

Generalized Anxiety Disorder 7-item scale

The GAD-7 scale is a brief, self-administered questionnaire for screening and measuring severity of GAD.22 It asks patients to rate seven items that represent problems with general anxiety and scores each item on a scale of 0 (not at all) to 3 (nearly every day). Similar to the other measures, it is easily accessible online.

Research evidence supports the reliability and validity of GAD-7 as a measure of anxiety in the general population. Sensitivity and specificity are 89% and 82%, respectively. Normative data for age- and sex-specific subgroups support its use across age groups and in both males and females.23 The GAD-7 performs well for detecting and monitoring not only GAD but also panic disorder, social anxiety disorder, and posttraumatic stress disorder.24

CAGE questionnaire for detection of substance use

The CAGE questionnaire is a widely used screening tool that was originally developed to detect alcohol abuse but has been adapted to assess other substance abuse.25,26 The omission of substance abuse from diagnostic consideration can have a major effect on quality of care, because substance abuse can be the underlying cause of other diseases. Therefore, routine administration of this instrument in clinical practice can lead to better understanding and monitoring of patient health.27

Similar to other instruments, CAGE is free and available online.27 It contains four simple questions, with 1 point assigned to each positive answer (see Table); the simple mnemonic makes the questions easy to ­remember and to administer in a clinical setting.

CAGE has demonstrated validity, with one study determining that scores ≥ 2 had a specificity and sensitivity of 76% and 93%, respectively, for identifying excessive drinking, and a specificity and sensitivity of 77% and 91%, respectively, for identifying alcohol abuse.28

 

 

 

Columbia Suicide Severity Rating Scale (C-SSRS)

C-SSRS was developed by researchers at Columbia University to assess the severity of and track changes over time in suicidal ideation and behavior. C-SSRS is two pages and takes only a few minutes to administer; however, it also may be completed as a self-report measure. The questions are phrased in an interview format, and while clinicians are encouraged to receive training prior to its administration, specific training in mental health is not required.

The "Lifetime/Recent" version allows practitioners to gather lifetime history of suicidality as well as any recent suicidal ideation and/or behavior, whereas the "Since Last Visit" version of the scale assesses suicidality in patients who have completed at least one Lifetime/Recent C-SSRS assessment. A truncated, six-item "Screener" version is typically used in emergency situations. A risk assessment can be added to either the Full or Screener version to summarize the answers from C-SSRS and document risk and protective factors.29

Several studies have found C-SSRS to be reliable and valid for identifying suicide risk in children and adults.30,31USA Today reported that an individual exhibiting even a single behavior identified by the scale is eight to 10 times more likely to complete suicide.32 In addition, the C-SSRS has helped reduce the suicide rate by 65% in one of the largest providers of community-based behavioral health care in the United States.32

USING SCALES TO AUGMENT CARE

Each of the scales described in this article can easily be incorporated into clinical practice. The information the scales provide can be used to track progression of symptoms and effectiveness of treatment. Although rating scales should never be used alone to establish a diagnosis or clinical treatment plan, they can and should be used to augment information from the clinician's assessment and follow-up interviews.5

References

1. McDowell I. Measuring Health: A Guide to Rating Scales and Questionnaires. 3rd ed. New York, NY: Oxford University Press; 2006.
2. Kennedy Forum. Fixing behavioral health care in America: a national call for integrating and coordinating specialty behavioral health care with the medical system. http://thekennedyforum-dot-org.s3.amazonaws.com/documents/KennedyForum-BehavioralHealth_FINAL_3.pdf. Accessed August 14, 2017. 
3. The Office of the National Coordinator for Health Information Technology. Behavioral health (BH) Clinical Quality Measures (CQMs) Program initiatives. www.healthit.gov/sites/default/files/pdf/2012-09-27-behavioral-health-clinical-quality-measures-program-initiatives-public-forum.pdf. Accessed August 14, 2017.
4. Unutzer J, Harbin H, Schoenbaum M. The collaborative care model: an approach for integrating physical and mental health care in Medicaid health homes. www.medicaid.gov/State-Resource-Center/Medicaid-State-Technical-Assistance/Health-Homes-Technical-Assistance/Downloads/HH-IRC-Collaborative-5-13.pdf. Accessed August 14, 2017. 
5. World Group On Psychiatric Evaluation; American Psychiatric Association Steering Committee On Practice Guidelines. Practice guideline for the psychiatric evaluation of adults. 2nd ed. http://psychiatryonline.org/pb/assets/raw/sitewide/practice_guidelines/guidelines/psychevaladults.pdf. Accessed August 14, 2017. 
6. Melek S, Norris D, Paulus J. Economic Impact of Integrated Medical-Behavioral Healthcare: Implications for Psychiatry. Denver, CO: Milliman, Inc; 2014. 
7. Archer J, Bower P, Gilbody S, et al. Collaborative care for depression and anxiety problems. Cochrane Database Syst Rev. 2012;10:CD006525. 
8. Kennedy Forum. Fixing behavioral health care in America: a national call for measurement-based care.  www.thekennedyforum.org/a-national-call-for-measurement-based-care/. Accessed August 14, 2017.
9. Zimmerman M, McGlinchey JB. Why don't psychiatrists use scales to measure outcome when treating depressed patients? J Clin Psychiatry. 2008;69(12):1916-1919. 
10. Hatfield D, McCullough L, Frantz SH, et al. Do we know when our clients get worse? An investigation of therapists' ability to detect negative client change. Clin Psychol Psychother. 2010;17(1):25-32.
11. SAMHSA-HRSA Center for Integrated Solutions. Screening tools. www.integration.samhsa.gov/clinical-practice/screening-tools. Accessed August 14, 2017. 
12. Moller HJ. Standardised rating scales in psychiatry: methodological basis, their possibilities and limitations and descriptions of important rating scales. World J Biol Psychiatry. 2009;10(1):6-26.
13. Sajatovic M, Ramirez LF. Rating Scales in Mental Health. 2nd ed. Hudson, OH: Lexi-Comp; 2003.
14. Patient Health Questionnaire-9 (PHQ-9). www.agencymeddirectors.wa.gov/files/AssessmentTools/14-PHQ-9%20overview.pdf. Accessed August 14, 2017.
15. Patient Health Questionnaire-9 (PHQ-9). Rehab Measures Web site. www.rehabmeasures.org/Lists/RehabMeasures/DispForm.aspx?ID=954. Accessed August 14, 2017. 
16. Kroenke K, Spitzer RL, Williams JB. The PHQ-9: validity of a brief depression severity measure. J Gen Intern Med. 2001;16(9):606-613. 
17. Löwe B, Unützer J, Callahan CM, et al. Monitoring depression treatment outcomes with the Patient Health Questionnaire-9. Med Care. 2004;42(12):1194-1201.
18. Ketter TA. Strategies for monitoring outcomes in patients with bipolar disorder. Prim Care Companion J Clin Psychiatry. 2010;12(suppl 1):10-16.
19. The Mood Disorder Questionnaire. University of Texas Medical Branch. www.dbsalliance.org/pdfs/MDQ.pdf. Accessed August 14, 2017.
20. Hirschfeld RM, Holzer C, Calabrese JR, et al. Validity of the Mood Disorder Questionnaire: a general population study. Am J Psychiatry. 2003;160(1):178-180.
21. Boschloo L, Nolen WA, Spijker AT, et al. The Mood Disorder Questionnaire (MDQ) for detecting (hypo) manic episodes: its validity and impact of recall bias. J Affect Disord. 2013;151(1):203-208.
22. Spitzer RL, Kroenke K, Williams JB, et al. A brief measure for assessing generalized anxiety disorder: the GAD-7. Arch Intern Med. 2006;166(10):1092-1097.
23. Lowe B, Decker O, Müller S, et al. Validation and standardization of the Generalized Anxiety Disorder Screener (GAD-7) in the general population. Med Care. 2008;46(3):266-274. 
24. Kroenke K, Spitzer RL, Williams JB, et al. Anxiety disorders in primary care: prevalence, impairment, comorbidity, and detection. Ann Intern Med. 2007;146(5):317-325.
25. Ewing JA. Detecting alcoholism. The CAGE Questionnaire. JAMA. 1984;252(14):1905-1907.
26. CAGE substance abuse screening tool. Johns Hopkins Medicine. www.hopkinsmedicine.org/johns_hopkins_healthcare/downloads/cage%20substance%20screening%20tool.pdf. Accessed August 14, 2017.
27. O'Brien CP. The CAGE questionnaire for detection of alcoholism: a remarkably useful but simple tool. JAMA. 2008;300(17):2054-2056. 
28. Bernadt MW, Mumford J, Taylor C, et al. Comparison of questionnaire and laboratory tests in the detection of excessive drinking and alcoholism. Lancet. 1982;1(8267):325-328. 
29. Columbia Suicide-Severity Rating Scale (CS-SRS). http://cssrs.columbia.edu/the-columbia-scale-c-ssrs/cssrs-for-communities-and-healthcare/#filter=.general-use.english. Accessed August 14, 2017. 
30. Mundt JC, Greist JH, Jefferson JW, et al. Prediction of suicidal behavior in clinical research by lifetime suicidal ideation and behavior ascertained by the electronic Columbia-Suicide Severity Rating Scale. J Clin Psychiatry. 2013;74(9):887-893.
31. Posner K, Brown GK, Stanley B, et al. The Columbia-Suicide Severity Rating Scale: initial validity and internal consistency findings from three multisite studies with adolescents and adults. Am J Psychiatry. 2011;168(12):1266-1277. 
32. Esposito L. Suicide checklist spots people at highest risk. USA Today. http://usatoday30.usatoday.com/news/health/story/health/story/2011-11-09/Suicide-checklist-spots-peo ple-at-highest-risk/51135944/1. Accessed August 14, 2017.

References

1. McDowell I. Measuring Health: A Guide to Rating Scales and Questionnaires. 3rd ed. New York, NY: Oxford University Press; 2006.
2. Kennedy Forum. Fixing behavioral health care in America: a national call for integrating and coordinating specialty behavioral health care with the medical system. http://thekennedyforum-dot-org.s3.amazonaws.com/documents/KennedyForum-BehavioralHealth_FINAL_3.pdf. Accessed August 14, 2017. 
3. The Office of the National Coordinator for Health Information Technology. Behavioral health (BH) Clinical Quality Measures (CQMs) Program initiatives. www.healthit.gov/sites/default/files/pdf/2012-09-27-behavioral-health-clinical-quality-measures-program-initiatives-public-forum.pdf. Accessed August 14, 2017.
4. Unutzer J, Harbin H, Schoenbaum M. The collaborative care model: an approach for integrating physical and mental health care in Medicaid health homes. www.medicaid.gov/State-Resource-Center/Medicaid-State-Technical-Assistance/Health-Homes-Technical-Assistance/Downloads/HH-IRC-Collaborative-5-13.pdf. Accessed August 14, 2017. 
5. World Group On Psychiatric Evaluation; American Psychiatric Association Steering Committee On Practice Guidelines. Practice guideline for the psychiatric evaluation of adults. 2nd ed. http://psychiatryonline.org/pb/assets/raw/sitewide/practice_guidelines/guidelines/psychevaladults.pdf. Accessed August 14, 2017. 
6. Melek S, Norris D, Paulus J. Economic Impact of Integrated Medical-Behavioral Healthcare: Implications for Psychiatry. Denver, CO: Milliman, Inc; 2014. 
7. Archer J, Bower P, Gilbody S, et al. Collaborative care for depression and anxiety problems. Cochrane Database Syst Rev. 2012;10:CD006525. 
8. Kennedy Forum. Fixing behavioral health care in America: a national call for measurement-based care.  www.thekennedyforum.org/a-national-call-for-measurement-based-care/. Accessed August 14, 2017.
9. Zimmerman M, McGlinchey JB. Why don't psychiatrists use scales to measure outcome when treating depressed patients? J Clin Psychiatry. 2008;69(12):1916-1919. 
10. Hatfield D, McCullough L, Frantz SH, et al. Do we know when our clients get worse? An investigation of therapists' ability to detect negative client change. Clin Psychol Psychother. 2010;17(1):25-32.
11. SAMHSA-HRSA Center for Integrated Solutions. Screening tools. www.integration.samhsa.gov/clinical-practice/screening-tools. Accessed August 14, 2017. 
12. Moller HJ. Standardised rating scales in psychiatry: methodological basis, their possibilities and limitations and descriptions of important rating scales. World J Biol Psychiatry. 2009;10(1):6-26.
13. Sajatovic M, Ramirez LF. Rating Scales in Mental Health. 2nd ed. Hudson, OH: Lexi-Comp; 2003.
14. Patient Health Questionnaire-9 (PHQ-9). www.agencymeddirectors.wa.gov/files/AssessmentTools/14-PHQ-9%20overview.pdf. Accessed August 14, 2017.
15. Patient Health Questionnaire-9 (PHQ-9). Rehab Measures Web site. www.rehabmeasures.org/Lists/RehabMeasures/DispForm.aspx?ID=954. Accessed August 14, 2017. 
16. Kroenke K, Spitzer RL, Williams JB. The PHQ-9: validity of a brief depression severity measure. J Gen Intern Med. 2001;16(9):606-613. 
17. Löwe B, Unützer J, Callahan CM, et al. Monitoring depression treatment outcomes with the Patient Health Questionnaire-9. Med Care. 2004;42(12):1194-1201.
18. Ketter TA. Strategies for monitoring outcomes in patients with bipolar disorder. Prim Care Companion J Clin Psychiatry. 2010;12(suppl 1):10-16.
19. The Mood Disorder Questionnaire. University of Texas Medical Branch. www.dbsalliance.org/pdfs/MDQ.pdf. Accessed August 14, 2017.
20. Hirschfeld RM, Holzer C, Calabrese JR, et al. Validity of the Mood Disorder Questionnaire: a general population study. Am J Psychiatry. 2003;160(1):178-180.
21. Boschloo L, Nolen WA, Spijker AT, et al. The Mood Disorder Questionnaire (MDQ) for detecting (hypo) manic episodes: its validity and impact of recall bias. J Affect Disord. 2013;151(1):203-208.
22. Spitzer RL, Kroenke K, Williams JB, et al. A brief measure for assessing generalized anxiety disorder: the GAD-7. Arch Intern Med. 2006;166(10):1092-1097.
23. Lowe B, Decker O, Müller S, et al. Validation and standardization of the Generalized Anxiety Disorder Screener (GAD-7) in the general population. Med Care. 2008;46(3):266-274. 
24. Kroenke K, Spitzer RL, Williams JB, et al. Anxiety disorders in primary care: prevalence, impairment, comorbidity, and detection. Ann Intern Med. 2007;146(5):317-325.
25. Ewing JA. Detecting alcoholism. The CAGE Questionnaire. JAMA. 1984;252(14):1905-1907.
26. CAGE substance abuse screening tool. Johns Hopkins Medicine. www.hopkinsmedicine.org/johns_hopkins_healthcare/downloads/cage%20substance%20screening%20tool.pdf. Accessed August 14, 2017.
27. O'Brien CP. The CAGE questionnaire for detection of alcoholism: a remarkably useful but simple tool. JAMA. 2008;300(17):2054-2056. 
28. Bernadt MW, Mumford J, Taylor C, et al. Comparison of questionnaire and laboratory tests in the detection of excessive drinking and alcoholism. Lancet. 1982;1(8267):325-328. 
29. Columbia Suicide-Severity Rating Scale (CS-SRS). http://cssrs.columbia.edu/the-columbia-scale-c-ssrs/cssrs-for-communities-and-healthcare/#filter=.general-use.english. Accessed August 14, 2017. 
30. Mundt JC, Greist JH, Jefferson JW, et al. Prediction of suicidal behavior in clinical research by lifetime suicidal ideation and behavior ascertained by the electronic Columbia-Suicide Severity Rating Scale. J Clin Psychiatry. 2013;74(9):887-893.
31. Posner K, Brown GK, Stanley B, et al. The Columbia-Suicide Severity Rating Scale: initial validity and internal consistency findings from three multisite studies with adolescents and adults. Am J Psychiatry. 2011;168(12):1266-1277. 
32. Esposito L. Suicide checklist spots people at highest risk. USA Today. http://usatoday30.usatoday.com/news/health/story/health/story/2011-11-09/Suicide-checklist-spots-peo ple-at-highest-risk/51135944/1. Accessed August 14, 2017.

Issue
Clinician Reviews - 27(9)
Issue
Clinician Reviews - 27(9)
Page Number
28-31,33
Page Number
28-31,33
Publications
Publications
Topics
Article Type
Display Headline
Behavioral Health: Using Rating Scales in a Clinical Setting
Display Headline
Behavioral Health: Using Rating Scales in a Clinical Setting
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Article PDF Media

Atopic Dermatitis Prevention and Treatment

Article Type
Changed
Thu, 01/10/2019 - 13:45
Display Headline
Atopic Dermatitis Prevention and Treatment

Atopic dermatitis (AD) is a disease that finally is coming of age in dermatology research. New topical agents and systemic biologic agents offer patients with AD other options for medical management. This article provides a practical review of prevention strategies and treatment guidelines for AD.

PREVENTION

Prevention strategies for AD have been largely unsuccessful in the past, which may relate to factors such as prenatal triggers.1 However, some newer interventional studies have shown some promise in AD prevention in specific settings. For example, a randomized trial of infants in the United States and United Kingdom at high risk for AD (ie, family history of atopy) reported that the AD risk was reduced by 50% when patients were treated with at least once-daily application of full-body emollients for 6 months (beginning by 3 weeks of life).2 The strategy of daily application of emollients for avoidance of AD in infants with a family history of AD is reasonable but may not offer lifetime prevention, and the benefit in children not from AD families is unknown.

Other trials to prevent AD have included usage of dust avoidance and dust covers for mattresses. This strategy showed modest benefit in reducing the incidence of atopic diatheses in the first year3 but did not gain endorsement by the most recent guidelines of the American Academy of Dermatology (AAD).4

Prenatal and postnatal (maternal and child) supplementation of Lactobacillus rhamnosus has shown promise in prevention.5 The exact regimen likely makes an impact on efficacy. An early study showed the usage of probiotics (eg, Lactobacillus reuteri) prenatally in pregnant women and postnatally in infants resulted in no reduction in occurrence of AD and possible reduction in IgE-associated AD.6 Kalliomäki et al7 demonstrated that L rhamnosus GG alone reduced AD by half in at-risk infants in a double-blind, placebo-controlled trial. On the other hand, Taylor et al8 performed a study of probiotic supplementation in which patients at high risk for AD developed higher rates of allergen sensitization. The most successful recent trial involved the randomization of 415 pregnant women to receive interventions from 36 weeks’ gestation until 3 months postpartum.9 The intervention was a randomized comparison of milk without probiotics versus a blend of probiotic milk containing L rhamnosus GG, Lactobacillus acidophilus La-5, and Bifidobacterium animalis subsp lactis Bb-12. At 6 years of age, 81 babies who consumed probiotic milk and 82 babies who consumed milk without probiotics were available for testing. The strategy caused a statistically significant reduction in AD in the complete case analysis (odds ratio, 0.48; 95% confidence interval, 0.25-0.92; P=.027; number needed to treat, 6). Sadly, other allergic diseases were not prevented in this study.9

MANAGEMENT OF AD

There currently is no cure or perfected prevention technique for AD. As a result, therapy focuses on avoiding triggers and alleviating symptoms.10 Recent guidelines from the AAD state that“[t]he ultimate judgment regarding the propriety of any specific therapy must be made by the physician and the patient in light of all the circumstances presented by the individual patient, and the known variability and biologic behavior of the disease.”11 Skin-directed therapies are the first line of treatment including emollients, gentle skin care, and topical medicaments. In AD, therapies are needed to reduce disease activity and flare severity, clear flares, and provide relief.

Parental education and written eczema action plans are recommended to help patients and parents/guardians follow recommended regimens12; Tollefson and Bruckner13 for the American Academy of Pediatrics provide an action plan to guide the care of children with atopic dermatitis that is simple, but many others exist online. The eczema action plan usually provides information on how to bathe and what to do when the skin is actively inflamed.

In 2014, a 4-part series of guidelines of care for the management of AD was published by the AAD, replacing prior guidelines.4,11,14,15 The following sections review some of the important parameters of care highlighted in these management guidelines.

Psychological Support

Appropriate psychological support for AD patients can be sought through counselors, therapists, psychiatrists, and support groups such as the National Eczema Association (https://nationaleczema.org/).

Education

Education is the leading form of medical therapy in patients with AD. Eczema schools are popular in Europe and are just beginning to form in the United States (http://tuh.templehealth.org/content/eczema_school.htm), which can be helpful to educate caregivers and patients with AD. Patient resources online and through support groups with an online presence, in-person meetings, and patient/family conventions can be helpful to AD patients. Often, an initial office visit with a dermatologist involves a review of avoidance of triggers, usage of gentle skin care including bland emollients, and therapeutic regimens for disease activity. This form of verbal education is to be paired with an eczema action plan, a written document that allows individuals to reference recommendations and share information with other caregivers.12,13,16

Emollients and Gentle Skin Care

Gentle skin care regimens, which includes the usage of synthetic cleansers with a low pH to help maintain the acidity (acid mantle) of the skin, seek to reduce irritation and have been rated as level IA (highest level) in recent AAD guidelines.14 Although bathing frequency has been emphasized in the guidelines, AD severity as reflected by SCORAD (SCORing Atopic Dermatitis) was not different for daily bathing versus twice weekly.17 The American Academy of Pediatrics recommended a skin care regimen of bathing every 2 to 3 days in lukewarm water for 10 to 15 minutes, followed by application of emollients that are fragrance free and have few preservatives.13 Topical emollients with additives such as colloidal oatmeal, avenanthramides, or ceramides can be used to enhance the skin barrier and are well tolerated in all age groups.18,19 Despite enhanced emollients, the therapy of AD still requires usage of prescription or over-the-counter TCs and/or topical calcineurin inhibitors (TCIs) in many cases.20

 

 

Topical Medication

Children have a relatively higher body surface area–to-weight ratio, allowing for greater potential absorption of topical medicaments and potential side effects from absorption. Types of vehicle, cost, site of application, and availability may impact patient and physician preference in choice of therapeutic topical agent.14

Topical Corticosteroids
Topical corticosteroids (TCs) are the mainstay of treatment for AD and have been used for more than 60 years.14,20 Topical corticosteroids provide anti-inflammatory effects on T cells, monocytes, and macrophages, producing altered cytokine activity locally. Topical corticosteroids inhibit collagen synthesis, potentially causing skin atrophy. They also inhibit IL-1, IL-2, IL-6, IFN-α, and tumor necrosis factor α.21 Topical corticosteroids are classified as class I (ultra-high potency) to class VII (low potency). In children, low-potency TCs generally are applied to the face, intertriginous areas, groin, and genitalia, and mid-potency corticosteroids are applied to the body, arms, and legs. An even higher-strength agent can be prescribed as a rescue medication in severe cases. After clearance with once- or twice-daily therapy, twice-weekly usage can benefit disease activity.22 Topical corticosteroids reduce inflammation as well as Staphylococcus aureus load through inhibition of cytokines that inhibit antimicrobial peptides. Topical corticosteroids have been endorsed as level IA evidence therapy by the AAD guidelines.14

Topical corticosteroids, particularly prolonged usage of mid- to high-potency products, have been associated with side effects such as skin atrophy, striae, telangiectases, hypopigmentation, rosacea, acneiform eruptions, focal hypertrichosis, perioral dermatitis, and acne23; potential systemic side effects include hypothalamic-pituitary-adrenal axis suppression, cataracts, glaucoma (with periocular application), Cushing syndrome, hyperglycemia, hypertension,23 and growth retardation.14 Long-term corticosteroid therapy is associated with tachyphylaxis and potential rebound of disease with discontinuation.24 Based on the potential risk of side effects with TCs, the least potent product for the shortest time needed is recommended, with special care for thin skin. Discontinuation when clearance occurs is advised. Allergy to TCs and/or vehicle ingredients such as propylene glycol should be suspected in severe unremitting cases.14 A recent registry review of children screened for contact dermatitis demonstrated that children with AD had higher sensitization to the steroid tixocortol pivalate.25

Topical Calcineurin Inhibitors
Topical calcineurin inhibitors include pimecrolimus cream 1%, which is approved for mild to moderate AD in adults and children 2 years and older, and tacrolimus ointment 0.03% and 0.1%, which are approved for moderate to severe AD in adults and children aged 2 to 15 years (0.03% formulation only). Topical calcineurin inhibitors can be used as second-line agents in AD in patients who have inadequate response to TCs or who may not be able to use TCs due to the disease site.10,13,14 Guidelines from the AAD also have endorsed TCIs as level IA evidence for steroid-sparing agents.

Concerns about the reporting of cancers and lymphomas prompted the US Food and Drug Administration to issue a black box warning on TCIs more than 10 years ago. Pimecrolimus, which has little absorption and no notable immunosuppressive effects, has been used without detrimental effect on vaccination and delayed-type hypersensitivities, but many decades of data are lacking.10,13,14,17,26-29 Topical calcineurin inhibitors can be used as steroid-sparing agents in lieu of corticosteroids in specific locations such as the face and eyelids and for long-term suppressive therapy twice weekly.30 Intermittent usage and cycling with corticosteroids is advisable,28 but usage intermittently beyond 1 year has not been evaluated.

Topical calcineurin inhibitors are recommended as effective for acute and chronic AD. Their use as maintenance therapy in adults and children, for AD recalcitrant to steroids, for AD in sensitive areas, for steroid-induced atrophy, and for long-term uninterrupted topical steroid usage carries a level IA evidence recommendation. Furthermore, the AAD guidelines have recommended TCIs as steroid-sparing agents with level IA evidence and off-label use of TCIs in children younger than 2 years with level IA evidence. Pretreatment with TCs to reduce stinging has level IIB evidence. Usage for flare prevention is level IA evidence. Routine blood monitoring of TCI-treated patients was not recommended; in fact, the AAD guidelines provided this recommendation as level IA evidence against routine laboratory monitoring of TCI-treated patients.14

Topical Antibiotics
Topical antibiotics are indicated for the therapy of impetigo and can be used in the setting of impetiginized AD in conjunction with TCs. Recent AAD guidelines suggested against routine usage of topical antistaphylococcal agents as level IA evidence.14 There is one study supporting usage of topical mupirocin in addition to TCs to heal children with eczema area and severity index scores more than 7 more rapidly in the first week of AD therapy, but in the same study, additive benefit was not demonstrated in AD beyond the first week.31 There also are data supporting usage of intranasal mupirocin adjunctively with bleach baths in patients with moderate to severe AD, which was rated as level IIB evidence in the AAD guidelines.14,32 There are limited data on the long-term utility of topical anti-infectives in AD. The risks of long-term usage could include resistance formation to agents such as mupirocin, contact dermatitis, and lack of efficacy.

 

 

Additional Therapeutics

Wet Wraps
Penetration through the stratum corneum is needed for drug activity in AD. Penetration can be enhanced using wet wrap therapy or using ointments, which produce higher relative potency.13 Wet wraps overlying a dilute topical medicament have been described as effective in AD and are recommended in AAD guidelines as level IIB evidence.14 Different wet wrap techniques can be used, including wet pajamas covered by dry pajamas or saline-soaked gauze wrapped around the affected areas and then dry gauze applied over the wet gauze. The methodology used should be tailored to the patient as well as to whether the individual is an inpatient or outpatient.

Bleach Baths
Dilute sodium hypochlorite solution 0.005% (one-quarter cup bleach in 20 gallons of water) has been demonstrated to be beneficial in reduction of disease activity in AD patients with recurrent bacterial infections.32 This simple technique in addition to intranasal mupirocin can reduce AD severity and improve quality of life and is the only ongoing S aureus therapeutic management endorsed by the AAD guidelines for the management of AD.14,32

Topical and Oral Delivery

Antihistamines
Topical antihistamines are ineffective in AD. Oral antihistamines can be used to reduce pruritus and are most effective when given as sedating agents for sleep enhancement but may be given as nonsedating agents for patients with concomitant allergic disorders such as allergic rhinoconjunctivitis. Paradoxical hyperreactivity with sedating antihistamines is not uncommon in small children, and sedating antihistamine usage should be discontinued in these instances.13 Parents of children with AD have reported giving the child antihistamines to sleep was helpful, as well as putting on creams, using special clothes (eg, all cotton), and keeping the room cool.33 There is level IIIC evidence against use of systemic antihistamines and level IIA evidence for sedating and nonsedating, according to the AAD guidelines.14

Systemic Therapeutics

Oral therapeutics range from oral antihistamines to oral antibiotics and immunosuppressive medications. Oral antibiotics (level IIB evidence) are reserved for superinfected AD, which is not easily defined for the following reasons: there is no consensus definition of superinfected AD; the majority of active AD lesions when cultured will demonstrate S aureus growth; and most AD lesions ooze, thereby creating the appearance of superinfection. In real-world practice, superinfection can be diagnosed based on the presence of pustules; furuncles; or signs of infection such as tracking erythema, tenderness, severe erosions, or maceration. Clinical judgment is always required.

The immunosuppressive medications used in AD include leukotriene inhibitors, which rarely are effective for AD.34 More effective systemic agents for AD include cyclosporine (level I to IIB evidence), azathioprine (level IIB evidence), mycophenolate mofetil (level IIIC evidence), and methotrexate (level IIB evidence). These agents are indicated for pediatric or adult patients when topical agents and/or phototherapy have failed.15 Monitoring these agents for side effects includes ongoing evaluation for renal and liver toxicity. Short courses (ie, 6 months) are preferred to minimize side effects.35

Dupilumab, an injectable AD therapy, is approved in the United States. This agent is injected every 2 weeks and binds to the IL-4Rα shared by IL-4 and IL-13. In 4 weeks of monotherapy, 85% of adult patients treated had 50% or greater clearance.36 Recently published consensus opinion from the International Eczema Council recommends assessment of a variety of factors before initiating systemic therapy including comorbid illnesses such as contact allergy, trigger avoidance, superinfection, and impact on quality of life.37

Oral Corticosteroids
Systemic corticosteroids clear patients quickly but provide no sustained improvement; in fact, many patients rebound or have tachyphylaxis. Although short-term corticosteroid usage can break the itch-scratch cycle, long-term usage is associated with osteoporosis, Cushing syndrome, and aseptic necrosis of the femoral head. Decreased linear growth will occur during therapy in children; therefore, systemic steroids are not recommended in children with AD, except for additional or comorbid conditions (eg, asthma or contact dermatitis).4

Phototherapy
Phototherapy has been recommended in the AAD guidelines as a second-line treatment after failure of first-line agents (ie, TCIs and TCs) for clearance and or maintenance and should be tailored to the patient’s skin tone by an experienced physician. Narrowband UVB phototherapy may act through the suppression of T-cell activity in the skin and possibly via suppression of staphylococcal superantigens; however, many phototherapy types have been described for AD.38,39 Usage can be effective in school-aged children and teenagers but may be limited due to school attendance. Phototherapy was graded as level IIB evidence in the AAD guidelines.15 Side effects include aggravation of AD by exposure to heat and UV light, actinic damage, tenderness, erythema, pruritus, burning, and stinging. Lentigines; skin cancers (melanoma and nonmelanoma); folliculitis; and ocular toxicity, especially cataracts, can occur.15 Children younger than 6 years will find it difficult to stand in a phototherapy booth and may be poor candidates.15,38,39

 

 

Complementary and Alternative Medicine

Complementary and alternative medicine (CAM) also has been used for AD in the United States. In a review of the 2007 National Health Interview Survey of 9417 children aged 0 to 17 years, CAM was used for AD by 0.99% of children. Some CAM techniques were associated with worsening severity of AD, including herbal therapy, vitamins, homeopathic agents, diet, and movement techniques.40 Usage of Chinese herbal medications for AD can be associated with liver toxicity.41 Only one CAM therapy—massage therapy—has some mild supportive data.42

Allergen Avoidance and Diet
Bronsnick et al43 discussed the possible benefit of prenatal and postnatal probiotics for prevention of AD, which were not supported in the AAD guidelines for management of AD4; postnatal prebiotic supplementation; and exclusive breastfeeding and/or supplementation with hydrolyzed formula in at-risk children. Elimination diets for children and mothers were not recommended. The authors found no beneficial role of supplements including vitamin D, selenium, fish oil, borage oil, and zinc sulfate.43

A National Institute of Allergy and Infectious Diseases consensus group recommended avoidance of proven but not random elimination of food allergens in AD, asthma, and/or eosinophilic esophagitis.44 Restricted maternal diet was not recommended, and breastfeeding exclusively for the first 4 to 6 months was recommended. Hydrolyzed formulas were suggested as a possible preventive strategy in at-risk infants as a breastfeeding alternative, with cost of these formulas being a problem.44

In children younger than 5 years, food allergy screening for the most common allergens (eg, milk, eggs, peanuts, wheat, soy) should be considered in children with persistent unremitting dermatitis and/or known food challenge–induced reactions.4 Conservative measures to avoid house dust mite exposure in known sensitized individuals including dust covers for pillows and mattresses may be beneficial.4,45

Emerging Therapies

Recently approved therapies include better-targeted agents that appear to have a reasonable safety profile and may fulfill unmet needs in AD care. Of these agents, crisaborole, a topical boron-based phosphodiesterase 4 inhibitor, was approved in December 2016 for mild to moderate AD in patients 2 years and older.Topically, this agent seems to be efficacious in the absence of notable carcinogenicity.46

The systemic (injectable) biologic agent dupilumab was approved in March 2017 for moderate to severe AD. Phase 3 studies in adults with AD showed excellent success in adults with moderate to severe AD.37 This agent is a monoclonal antibody targeted at blockade of the crucial atopic inflammatory triggering pathway via blockade of the IL-4A receptor site, targeting IL-4 and IL-13 activity.36,47 There are many medications in the pipeline, which Renert-Yuval and Guttman-Yassky48 review. However, an overview of the landscape demonstrates that Janus kinase (JAK) inhibitors49 and biologic medications in addition to dupilumab affecting targeted inflammatory cascades in AD are in development. In particular, the JAK inhibitors appear promising due to availability both as oral and topical agents.49

Need for Ongoing Care and Monitoring

Atopic dermatitis is a chronic inflammatory skin disorder with a genetic basis. Once initiated, the process of AD may persist throughout the patient’s life and become a systemic disorder with comorbidities including sleep disturbance, reduced quality of life, and cardiovascular disease.50 Ongoing management of AD includes topical reduction in irritants and triggers, topical medicaments, and management of pruritus and infections. At this time, emollients and irritant avoidance paired with judicious topical medicaments including TCs and second-line or site-specific (eg, eyelids) usage of TCIs or phosphodiesterase 4 inhibitors remain the backbone of therapy. Ongoing review of therapeutics for associated morbidities is underway, which may guide future therapeutic interventions into AD. The future of prevention and therapy look bright, but time will tell.

References
  1. Kelleher M, Dunn-Galvin A, Hourihane JO, et al. Skin barrier dysfunction measured by transepidermal water loss at 2 days and 2 months predates and predicts atopic dermatitis at 1 year. J Allergy Clin Immunol. 2015;135:930-935.
  2. Simpson EL, Chalmers JR, Hanifin JM, et al. Emollient enhancement of the skin barrier from birth offers effective atopic dermatitis prevention. J Allergy Clin Immunol. 2014;134:818-823.
  3. Tsitoura S, Nestoridou K, Botis P, et al. Randomized trial to prevent sensitization to mite allergens in toddlers and preschoolers by allergen reduction and education: one-year results. Arch Pediatr Adolesc Med. 2002;156:1021-1027.
  4. Sidbury R, Tom WL, Bergman JN, et al. Guidelines of care for the management of atopic dermatitis: section 4. prevention of disease flares and use of adjunctive therapies and approaches. J Am Acad Dermatol. 2014;71:1218-1233.
  5. Foolad N, Brezinski EA, Chase EP, et al. Effect of nutrient supplementation on atopic dermatitis in children: a systematic review of probiotics, prebiotics, formula, and fatty acids. JAMA Dermatol. 2013;149:350-355.
  6. Abrahamsson TR, Jakobsson T, Böttcher MF, et al. Probiotics in prevention of IgE-associated eczema: a double-blind, randomized, placebo-controlled trial. J Allergy Clin Immunol. 2007;119:1174-1180.
  7. Kalliomäki M, Salminen S, Arvilommi H, et al. Probiotics in primary prevention of atopic disease: a randomised placebo-controlled trial. Lancet. 2001;357:1076-1079.
  8. Taylor AL, Dunstan JA, Prescott SL. Probiotic supplementation for the first 6 months of life fails to reduce the risk of atopic dermatitis and increases the risk of allergen sensitization in high-risk children: a randomized controlled trial. J Allergy Clin Immunol. 2007;119:184-191.
  9. Simpson MR, Dotterud CK, Storrø O, et al. Perinatal probiotic supplementation in the prevention of allergy related disease: 6 year follow up of a randomised controlled trial. BMC Dermatol. 2015;15:13. doi:10.1186/s12895-015-0030-1.
  10. Carr WW. Topical calcineurin inhibitors for atopic dermatitis: review and treatment recommendations. Paediatr Drugs. 2013;15:303-310.
  11. Eichenfield LF, Tom WL, Chamlin SL, et al. Guidelines of care for the management of atopic dermatitis: section 1. diagnosis and assessment of atopic dermatitis. J Am Acad Dermatol. 2014;70:338-351.
  12. Silverberg NB. Creating an action plan for eczema patients. Cutis. 2015;96:362-363.
  13. Tollefson MM, Bruckner AL; Section on Dermatology. Atopic dermatitis: skin-directed management. Pediatrics. 2014;134:E1735-E1744.
  14. Eichenfield LF, Tom WL, Berger TG, et al. Guidelines of care for the management of atopic dermatitis: section 2. management and treatment of atopic dermatitis with topical therapies. J Am Acad Dermatol. 2014;71:116-132.
  15. Sidbury R, Davis DM, Cohen DE, et al; American Academy of Dermatology. Guidelines of care for the management of atopic dermatitis: section 3. management and treatment with phototherapy and systemic agents. J Am Acad Dermatol. 2014;71:327-349.
  16. Shi VY, Nanda S, Lee K, et al. Improving patient education with an eczema action plan: a randomized controlled trial. JAMA Dermatol. 2013;149:481-483.
  17. Koutroulis I, Petrova K, Kratimenos P, et al. Frequency of bathing in the management of atopic dermatitis: to bathe or not to bathe? Clin Pediatr (Phila). 2014;53:677-681.
  18. Fowler JF, Nebus J, Wallo W, et al. Colloidal oatmeal formulations as adjunct treatments in atopic dermatitis. J Drugs Dermatol. 2012;11:804-807.
  19. Fowler J Jr, Silverberg N. Active naturals have a key role in atopic dermatitis. Semin Cutan Med Surg. 2008;27:8-10.
  20. Eichenfield LF. Consensus guidelines in diagnosis and treatment of atopic dermatitis. Allergy. 2004;59:86-92.
  21. Nghiem P, Pearson G, Langley RG. Tacrolimus and pimecrolimus: from clever prokaryotes to inhibiting calcineurin and treating atopic dermatitis. J Am Acad Dermatol. 2002;46:228-241.
  22. Schmitt J. Commentary: eczema and cancer risk. Br J Dermatol. 2011;165:463-464.
  23. Abramovits W, Hung P, Tong KB. Efficacy and economics of topical calcineurin inhibitors for the treatment of atopic dermatitis. Am J Clin Dermatol. 2006;7:213-222.
  24. Takahashi-Ando N, Jones MA, Fujisawa S, et al. Patient-reported outcomes after discontinuation of long-term topical corticosteroid treatment for atopic dermatitis: a targeted cross-sectional survey. Drug Healthc Patient Saf. 2015;7:57-62.
  25. Jacob SE, McGowan M, Silverberg NB, et al. Pediatric contact dermatitis registry data on contact allergy in children with atopic dermatitis. JAMA Dermatol. 2017;153:765-770.
  26. Werfel T. Topical use of pimecrolimus in atopic dermatitis: update on the safety and efficacy. J Dtsch Dermatol Ges. 2009;7:739-742.
  27. Wahn U, Bos JD, Goodfield M, et al. Efficacy and safety of pimecrolimus cream in the long-term management of atopic dermatitis in children. Pediatrics. 2002;110(1, pt 1):E2.
  28. Berger TG, Duvic M, Van Voorhees AS, et al; American Academy of Dermatology Association Task Force. The use of topical calcineurin inhibitors in dermatology: safety concerns. report of the American Academy of Dermatology Association Task Force. J Am Acad Dermatol. 2006;54:818-823.
  29. Paller AS. Latest approaches to treating atopic dermatitis. Chem Immunol Allergy. 2012;96:132-140.
  30. Thaçi D, Reitamo S, Gonzalez Ensenat MA, et al. Proactive disease management with 0.03% tacrolimus ointment for children with atopic dermatitis: results of a randomized, multicentre, comparative study. Br J Dermatol. 2008;159:1348-1356.
  31. Gong JQ, Lin L, Lin T, et al. Skin colonization by Staphylococcus aureus in patients with eczema and atopic dermatitis and relevant combined topical therapy: a double-blind multicentre randomized controlled trial. Br J Dermatol. 2006;155:680-687.
  32. Huang JT, Abrams M, Tlougan B, et al. Treatment of Staphylococcus aureus colonization in atopic dermatitis decreases disease severity. Pediatrics. 2009;123:E808-E814.
  33. Reid P, Lewis-Jones MS. Sleep difficulties and their management in preschoolers with atopic eczema. Clin Exp Dermatol. 1995;20:38-41.
  34. Silverberg NB, Paller AS. Leukotriene receptor antagonists are ineffective for severe atopic dermatitis. J Am Acad Dermatol. 2004;50:485-486.
  35. Wolverton SE. Comprehensive Dermatologic Drug Therapy. 3rd ed. New York, NY: Elsevier Saunders; 2013.
  36. Beck LA, Thaçi D, Hamilton JD, et al. Dupilumab treatment in adults with moderate-to-severe atopic dermatitis. N Engl J Med. 2014;371:130-139.
  37. Simpson EL, Bruin-Weller M, Flohr C, et al. When does atopic dermatitis warrant systemic therapy? recommendations from an expert panel of the International Eczema Council [published online August 10, 2017]. J Am Acad Dermatol. doi:10.1016/j.jaad.2017.06.042.
  38. Veith W, DeLeo V, Silverberg N. Medical phototherapy in childhood skin diseases. Minerva Pediatr. 2011;63:327-333.
  39. Song E, Reja D, Silverberg N, et al. Phototherapy: kids are not just little people. Clin Dermatol. 2015;33:672-680.
  40. Silverberg JI, Lee-Wong M, Silverberg NB. Complementary and alternative medicines and childhood eczema: a US population-based study. Dermatitis. 2014;25:246-254.
  41. Stickel F, Shouval D. Hepatotoxicity of herbal and dietary supplements: an update. Arch Toxicol. 2015;89:851-865.
  42. Schachner L, Field T, Hernandez-Reif M, et al. Atopic dermatitis symptoms decreased in children following massage therapy. Pediatr Dermatol. 1998;15:390-395.
  43. Bronsnick T, Murzaku EC, Rao BK. Diet in dermatology: part I. atopic dermatitis, acne, and nonmelanoma skin cancer. J Am Acad Dermatol. 2014;71:1039.e1-1039.e12.
  44. Boyce JA, Assa’ad A, Burks AW, et al. Guidelines for the diagnosis and management of food allergy in the United States: summary of the NIAID-sponsored expert panel report. Nutr Res. 2011;31:61-75.
  45. Silverberg NB, Lee-Wong M, Yosipovitch G. Diet and atopic dermatitis. Cutis. 2016;97:227-232.
  46. Hanifin JM, Chan SC, Cheng JB, et al. Type phosphodiesterase inhibitors have clinical and in vitro anti-inflammatory effects in atopic dermatitis. J Invest Dermatol. 1996;107:51-56.
  47. Boguniewicz M, Leung DY. Targeted therapy for allergic diseases: at the intersection of cutting-edge science and clinical practice. J Allergy Clin Immunol. 2015;135:354-356.
  48. Renert-Yuval Y, Guttman-Yassky E. Systemic therapies in atopic dermatitis: the pipeline. Clin Dermatol. 2017;35:387-397.
  49. Damsky W, King BA. JAK inhibitors in dermatology: the promise of a new drug class. J Am Acad Dermatol. 2017;76:736-744.
  50. Brunner PM, Silverberg JI, Guttman-Yassky E, et al. Increasing comorbidities suggest that atopic dermatitis is a systemic disorder. J Invest Dermatol. 2017;137:18-25.
Article PDF
Author and Disclosure Information

From the Department of Dermatology, Mt Sinai West of the Icahn School of Medicine, New York, New York.

Dr. Silverberg is on the advisory board for and has received honoraria from Pfizer Inc. She also is an investigator for Regeneron Pharmaceuticals, Inc.

Correspondence: Nanette B. Silverberg, MD, Mt Sinai West, 425 W 59th St, Ste 8B, New York, NY 10019 ([email protected]).

Issue
Cutis - 100(3)
Publications
Topics
Page Number
173-177, 192
Sections
Author and Disclosure Information

From the Department of Dermatology, Mt Sinai West of the Icahn School of Medicine, New York, New York.

Dr. Silverberg is on the advisory board for and has received honoraria from Pfizer Inc. She also is an investigator for Regeneron Pharmaceuticals, Inc.

Correspondence: Nanette B. Silverberg, MD, Mt Sinai West, 425 W 59th St, Ste 8B, New York, NY 10019 ([email protected]).

Author and Disclosure Information

From the Department of Dermatology, Mt Sinai West of the Icahn School of Medicine, New York, New York.

Dr. Silverberg is on the advisory board for and has received honoraria from Pfizer Inc. She also is an investigator for Regeneron Pharmaceuticals, Inc.

Correspondence: Nanette B. Silverberg, MD, Mt Sinai West, 425 W 59th St, Ste 8B, New York, NY 10019 ([email protected]).

Article PDF
Article PDF
Related Articles

Atopic dermatitis (AD) is a disease that finally is coming of age in dermatology research. New topical agents and systemic biologic agents offer patients with AD other options for medical management. This article provides a practical review of prevention strategies and treatment guidelines for AD.

PREVENTION

Prevention strategies for AD have been largely unsuccessful in the past, which may relate to factors such as prenatal triggers.1 However, some newer interventional studies have shown some promise in AD prevention in specific settings. For example, a randomized trial of infants in the United States and United Kingdom at high risk for AD (ie, family history of atopy) reported that the AD risk was reduced by 50% when patients were treated with at least once-daily application of full-body emollients for 6 months (beginning by 3 weeks of life).2 The strategy of daily application of emollients for avoidance of AD in infants with a family history of AD is reasonable but may not offer lifetime prevention, and the benefit in children not from AD families is unknown.

Other trials to prevent AD have included usage of dust avoidance and dust covers for mattresses. This strategy showed modest benefit in reducing the incidence of atopic diatheses in the first year3 but did not gain endorsement by the most recent guidelines of the American Academy of Dermatology (AAD).4

Prenatal and postnatal (maternal and child) supplementation of Lactobacillus rhamnosus has shown promise in prevention.5 The exact regimen likely makes an impact on efficacy. An early study showed the usage of probiotics (eg, Lactobacillus reuteri) prenatally in pregnant women and postnatally in infants resulted in no reduction in occurrence of AD and possible reduction in IgE-associated AD.6 Kalliomäki et al7 demonstrated that L rhamnosus GG alone reduced AD by half in at-risk infants in a double-blind, placebo-controlled trial. On the other hand, Taylor et al8 performed a study of probiotic supplementation in which patients at high risk for AD developed higher rates of allergen sensitization. The most successful recent trial involved the randomization of 415 pregnant women to receive interventions from 36 weeks’ gestation until 3 months postpartum.9 The intervention was a randomized comparison of milk without probiotics versus a blend of probiotic milk containing L rhamnosus GG, Lactobacillus acidophilus La-5, and Bifidobacterium animalis subsp lactis Bb-12. At 6 years of age, 81 babies who consumed probiotic milk and 82 babies who consumed milk without probiotics were available for testing. The strategy caused a statistically significant reduction in AD in the complete case analysis (odds ratio, 0.48; 95% confidence interval, 0.25-0.92; P=.027; number needed to treat, 6). Sadly, other allergic diseases were not prevented in this study.9

MANAGEMENT OF AD

There currently is no cure or perfected prevention technique for AD. As a result, therapy focuses on avoiding triggers and alleviating symptoms.10 Recent guidelines from the AAD state that“[t]he ultimate judgment regarding the propriety of any specific therapy must be made by the physician and the patient in light of all the circumstances presented by the individual patient, and the known variability and biologic behavior of the disease.”11 Skin-directed therapies are the first line of treatment including emollients, gentle skin care, and topical medicaments. In AD, therapies are needed to reduce disease activity and flare severity, clear flares, and provide relief.

Parental education and written eczema action plans are recommended to help patients and parents/guardians follow recommended regimens12; Tollefson and Bruckner13 for the American Academy of Pediatrics provide an action plan to guide the care of children with atopic dermatitis that is simple, but many others exist online. The eczema action plan usually provides information on how to bathe and what to do when the skin is actively inflamed.

In 2014, a 4-part series of guidelines of care for the management of AD was published by the AAD, replacing prior guidelines.4,11,14,15 The following sections review some of the important parameters of care highlighted in these management guidelines.

Psychological Support

Appropriate psychological support for AD patients can be sought through counselors, therapists, psychiatrists, and support groups such as the National Eczema Association (https://nationaleczema.org/).

Education

Education is the leading form of medical therapy in patients with AD. Eczema schools are popular in Europe and are just beginning to form in the United States (http://tuh.templehealth.org/content/eczema_school.htm), which can be helpful to educate caregivers and patients with AD. Patient resources online and through support groups with an online presence, in-person meetings, and patient/family conventions can be helpful to AD patients. Often, an initial office visit with a dermatologist involves a review of avoidance of triggers, usage of gentle skin care including bland emollients, and therapeutic regimens for disease activity. This form of verbal education is to be paired with an eczema action plan, a written document that allows individuals to reference recommendations and share information with other caregivers.12,13,16

Emollients and Gentle Skin Care

Gentle skin care regimens, which includes the usage of synthetic cleansers with a low pH to help maintain the acidity (acid mantle) of the skin, seek to reduce irritation and have been rated as level IA (highest level) in recent AAD guidelines.14 Although bathing frequency has been emphasized in the guidelines, AD severity as reflected by SCORAD (SCORing Atopic Dermatitis) was not different for daily bathing versus twice weekly.17 The American Academy of Pediatrics recommended a skin care regimen of bathing every 2 to 3 days in lukewarm water for 10 to 15 minutes, followed by application of emollients that are fragrance free and have few preservatives.13 Topical emollients with additives such as colloidal oatmeal, avenanthramides, or ceramides can be used to enhance the skin barrier and are well tolerated in all age groups.18,19 Despite enhanced emollients, the therapy of AD still requires usage of prescription or over-the-counter TCs and/or topical calcineurin inhibitors (TCIs) in many cases.20

 

 

Topical Medication

Children have a relatively higher body surface area–to-weight ratio, allowing for greater potential absorption of topical medicaments and potential side effects from absorption. Types of vehicle, cost, site of application, and availability may impact patient and physician preference in choice of therapeutic topical agent.14

Topical Corticosteroids
Topical corticosteroids (TCs) are the mainstay of treatment for AD and have been used for more than 60 years.14,20 Topical corticosteroids provide anti-inflammatory effects on T cells, monocytes, and macrophages, producing altered cytokine activity locally. Topical corticosteroids inhibit collagen synthesis, potentially causing skin atrophy. They also inhibit IL-1, IL-2, IL-6, IFN-α, and tumor necrosis factor α.21 Topical corticosteroids are classified as class I (ultra-high potency) to class VII (low potency). In children, low-potency TCs generally are applied to the face, intertriginous areas, groin, and genitalia, and mid-potency corticosteroids are applied to the body, arms, and legs. An even higher-strength agent can be prescribed as a rescue medication in severe cases. After clearance with once- or twice-daily therapy, twice-weekly usage can benefit disease activity.22 Topical corticosteroids reduce inflammation as well as Staphylococcus aureus load through inhibition of cytokines that inhibit antimicrobial peptides. Topical corticosteroids have been endorsed as level IA evidence therapy by the AAD guidelines.14

Topical corticosteroids, particularly prolonged usage of mid- to high-potency products, have been associated with side effects such as skin atrophy, striae, telangiectases, hypopigmentation, rosacea, acneiform eruptions, focal hypertrichosis, perioral dermatitis, and acne23; potential systemic side effects include hypothalamic-pituitary-adrenal axis suppression, cataracts, glaucoma (with periocular application), Cushing syndrome, hyperglycemia, hypertension,23 and growth retardation.14 Long-term corticosteroid therapy is associated with tachyphylaxis and potential rebound of disease with discontinuation.24 Based on the potential risk of side effects with TCs, the least potent product for the shortest time needed is recommended, with special care for thin skin. Discontinuation when clearance occurs is advised. Allergy to TCs and/or vehicle ingredients such as propylene glycol should be suspected in severe unremitting cases.14 A recent registry review of children screened for contact dermatitis demonstrated that children with AD had higher sensitization to the steroid tixocortol pivalate.25

Topical Calcineurin Inhibitors
Topical calcineurin inhibitors include pimecrolimus cream 1%, which is approved for mild to moderate AD in adults and children 2 years and older, and tacrolimus ointment 0.03% and 0.1%, which are approved for moderate to severe AD in adults and children aged 2 to 15 years (0.03% formulation only). Topical calcineurin inhibitors can be used as second-line agents in AD in patients who have inadequate response to TCs or who may not be able to use TCs due to the disease site.10,13,14 Guidelines from the AAD also have endorsed TCIs as level IA evidence for steroid-sparing agents.

Concerns about the reporting of cancers and lymphomas prompted the US Food and Drug Administration to issue a black box warning on TCIs more than 10 years ago. Pimecrolimus, which has little absorption and no notable immunosuppressive effects, has been used without detrimental effect on vaccination and delayed-type hypersensitivities, but many decades of data are lacking.10,13,14,17,26-29 Topical calcineurin inhibitors can be used as steroid-sparing agents in lieu of corticosteroids in specific locations such as the face and eyelids and for long-term suppressive therapy twice weekly.30 Intermittent usage and cycling with corticosteroids is advisable,28 but usage intermittently beyond 1 year has not been evaluated.

Topical calcineurin inhibitors are recommended as effective for acute and chronic AD. Their use as maintenance therapy in adults and children, for AD recalcitrant to steroids, for AD in sensitive areas, for steroid-induced atrophy, and for long-term uninterrupted topical steroid usage carries a level IA evidence recommendation. Furthermore, the AAD guidelines have recommended TCIs as steroid-sparing agents with level IA evidence and off-label use of TCIs in children younger than 2 years with level IA evidence. Pretreatment with TCs to reduce stinging has level IIB evidence. Usage for flare prevention is level IA evidence. Routine blood monitoring of TCI-treated patients was not recommended; in fact, the AAD guidelines provided this recommendation as level IA evidence against routine laboratory monitoring of TCI-treated patients.14

Topical Antibiotics
Topical antibiotics are indicated for the therapy of impetigo and can be used in the setting of impetiginized AD in conjunction with TCs. Recent AAD guidelines suggested against routine usage of topical antistaphylococcal agents as level IA evidence.14 There is one study supporting usage of topical mupirocin in addition to TCs to heal children with eczema area and severity index scores more than 7 more rapidly in the first week of AD therapy, but in the same study, additive benefit was not demonstrated in AD beyond the first week.31 There also are data supporting usage of intranasal mupirocin adjunctively with bleach baths in patients with moderate to severe AD, which was rated as level IIB evidence in the AAD guidelines.14,32 There are limited data on the long-term utility of topical anti-infectives in AD. The risks of long-term usage could include resistance formation to agents such as mupirocin, contact dermatitis, and lack of efficacy.

 

 

Additional Therapeutics

Wet Wraps
Penetration through the stratum corneum is needed for drug activity in AD. Penetration can be enhanced using wet wrap therapy or using ointments, which produce higher relative potency.13 Wet wraps overlying a dilute topical medicament have been described as effective in AD and are recommended in AAD guidelines as level IIB evidence.14 Different wet wrap techniques can be used, including wet pajamas covered by dry pajamas or saline-soaked gauze wrapped around the affected areas and then dry gauze applied over the wet gauze. The methodology used should be tailored to the patient as well as to whether the individual is an inpatient or outpatient.

Bleach Baths
Dilute sodium hypochlorite solution 0.005% (one-quarter cup bleach in 20 gallons of water) has been demonstrated to be beneficial in reduction of disease activity in AD patients with recurrent bacterial infections.32 This simple technique in addition to intranasal mupirocin can reduce AD severity and improve quality of life and is the only ongoing S aureus therapeutic management endorsed by the AAD guidelines for the management of AD.14,32

Topical and Oral Delivery

Antihistamines
Topical antihistamines are ineffective in AD. Oral antihistamines can be used to reduce pruritus and are most effective when given as sedating agents for sleep enhancement but may be given as nonsedating agents for patients with concomitant allergic disorders such as allergic rhinoconjunctivitis. Paradoxical hyperreactivity with sedating antihistamines is not uncommon in small children, and sedating antihistamine usage should be discontinued in these instances.13 Parents of children with AD have reported giving the child antihistamines to sleep was helpful, as well as putting on creams, using special clothes (eg, all cotton), and keeping the room cool.33 There is level IIIC evidence against use of systemic antihistamines and level IIA evidence for sedating and nonsedating, according to the AAD guidelines.14

Systemic Therapeutics

Oral therapeutics range from oral antihistamines to oral antibiotics and immunosuppressive medications. Oral antibiotics (level IIB evidence) are reserved for superinfected AD, which is not easily defined for the following reasons: there is no consensus definition of superinfected AD; the majority of active AD lesions when cultured will demonstrate S aureus growth; and most AD lesions ooze, thereby creating the appearance of superinfection. In real-world practice, superinfection can be diagnosed based on the presence of pustules; furuncles; or signs of infection such as tracking erythema, tenderness, severe erosions, or maceration. Clinical judgment is always required.

The immunosuppressive medications used in AD include leukotriene inhibitors, which rarely are effective for AD.34 More effective systemic agents for AD include cyclosporine (level I to IIB evidence), azathioprine (level IIB evidence), mycophenolate mofetil (level IIIC evidence), and methotrexate (level IIB evidence). These agents are indicated for pediatric or adult patients when topical agents and/or phototherapy have failed.15 Monitoring these agents for side effects includes ongoing evaluation for renal and liver toxicity. Short courses (ie, 6 months) are preferred to minimize side effects.35

Dupilumab, an injectable AD therapy, is approved in the United States. This agent is injected every 2 weeks and binds to the IL-4Rα shared by IL-4 and IL-13. In 4 weeks of monotherapy, 85% of adult patients treated had 50% or greater clearance.36 Recently published consensus opinion from the International Eczema Council recommends assessment of a variety of factors before initiating systemic therapy including comorbid illnesses such as contact allergy, trigger avoidance, superinfection, and impact on quality of life.37

Oral Corticosteroids
Systemic corticosteroids clear patients quickly but provide no sustained improvement; in fact, many patients rebound or have tachyphylaxis. Although short-term corticosteroid usage can break the itch-scratch cycle, long-term usage is associated with osteoporosis, Cushing syndrome, and aseptic necrosis of the femoral head. Decreased linear growth will occur during therapy in children; therefore, systemic steroids are not recommended in children with AD, except for additional or comorbid conditions (eg, asthma or contact dermatitis).4

Phototherapy
Phototherapy has been recommended in the AAD guidelines as a second-line treatment after failure of first-line agents (ie, TCIs and TCs) for clearance and or maintenance and should be tailored to the patient’s skin tone by an experienced physician. Narrowband UVB phototherapy may act through the suppression of T-cell activity in the skin and possibly via suppression of staphylococcal superantigens; however, many phototherapy types have been described for AD.38,39 Usage can be effective in school-aged children and teenagers but may be limited due to school attendance. Phototherapy was graded as level IIB evidence in the AAD guidelines.15 Side effects include aggravation of AD by exposure to heat and UV light, actinic damage, tenderness, erythema, pruritus, burning, and stinging. Lentigines; skin cancers (melanoma and nonmelanoma); folliculitis; and ocular toxicity, especially cataracts, can occur.15 Children younger than 6 years will find it difficult to stand in a phototherapy booth and may be poor candidates.15,38,39

 

 

Complementary and Alternative Medicine

Complementary and alternative medicine (CAM) also has been used for AD in the United States. In a review of the 2007 National Health Interview Survey of 9417 children aged 0 to 17 years, CAM was used for AD by 0.99% of children. Some CAM techniques were associated with worsening severity of AD, including herbal therapy, vitamins, homeopathic agents, diet, and movement techniques.40 Usage of Chinese herbal medications for AD can be associated with liver toxicity.41 Only one CAM therapy—massage therapy—has some mild supportive data.42

Allergen Avoidance and Diet
Bronsnick et al43 discussed the possible benefit of prenatal and postnatal probiotics for prevention of AD, which were not supported in the AAD guidelines for management of AD4; postnatal prebiotic supplementation; and exclusive breastfeeding and/or supplementation with hydrolyzed formula in at-risk children. Elimination diets for children and mothers were not recommended. The authors found no beneficial role of supplements including vitamin D, selenium, fish oil, borage oil, and zinc sulfate.43

A National Institute of Allergy and Infectious Diseases consensus group recommended avoidance of proven but not random elimination of food allergens in AD, asthma, and/or eosinophilic esophagitis.44 Restricted maternal diet was not recommended, and breastfeeding exclusively for the first 4 to 6 months was recommended. Hydrolyzed formulas were suggested as a possible preventive strategy in at-risk infants as a breastfeeding alternative, with cost of these formulas being a problem.44

In children younger than 5 years, food allergy screening for the most common allergens (eg, milk, eggs, peanuts, wheat, soy) should be considered in children with persistent unremitting dermatitis and/or known food challenge–induced reactions.4 Conservative measures to avoid house dust mite exposure in known sensitized individuals including dust covers for pillows and mattresses may be beneficial.4,45

Emerging Therapies

Recently approved therapies include better-targeted agents that appear to have a reasonable safety profile and may fulfill unmet needs in AD care. Of these agents, crisaborole, a topical boron-based phosphodiesterase 4 inhibitor, was approved in December 2016 for mild to moderate AD in patients 2 years and older.Topically, this agent seems to be efficacious in the absence of notable carcinogenicity.46

The systemic (injectable) biologic agent dupilumab was approved in March 2017 for moderate to severe AD. Phase 3 studies in adults with AD showed excellent success in adults with moderate to severe AD.37 This agent is a monoclonal antibody targeted at blockade of the crucial atopic inflammatory triggering pathway via blockade of the IL-4A receptor site, targeting IL-4 and IL-13 activity.36,47 There are many medications in the pipeline, which Renert-Yuval and Guttman-Yassky48 review. However, an overview of the landscape demonstrates that Janus kinase (JAK) inhibitors49 and biologic medications in addition to dupilumab affecting targeted inflammatory cascades in AD are in development. In particular, the JAK inhibitors appear promising due to availability both as oral and topical agents.49

Need for Ongoing Care and Monitoring

Atopic dermatitis is a chronic inflammatory skin disorder with a genetic basis. Once initiated, the process of AD may persist throughout the patient’s life and become a systemic disorder with comorbidities including sleep disturbance, reduced quality of life, and cardiovascular disease.50 Ongoing management of AD includes topical reduction in irritants and triggers, topical medicaments, and management of pruritus and infections. At this time, emollients and irritant avoidance paired with judicious topical medicaments including TCs and second-line or site-specific (eg, eyelids) usage of TCIs or phosphodiesterase 4 inhibitors remain the backbone of therapy. Ongoing review of therapeutics for associated morbidities is underway, which may guide future therapeutic interventions into AD. The future of prevention and therapy look bright, but time will tell.

Atopic dermatitis (AD) is a disease that finally is coming of age in dermatology research. New topical agents and systemic biologic agents offer patients with AD other options for medical management. This article provides a practical review of prevention strategies and treatment guidelines for AD.

PREVENTION

Prevention strategies for AD have been largely unsuccessful in the past, which may relate to factors such as prenatal triggers.1 However, some newer interventional studies have shown some promise in AD prevention in specific settings. For example, a randomized trial of infants in the United States and United Kingdom at high risk for AD (ie, family history of atopy) reported that the AD risk was reduced by 50% when patients were treated with at least once-daily application of full-body emollients for 6 months (beginning by 3 weeks of life).2 The strategy of daily application of emollients for avoidance of AD in infants with a family history of AD is reasonable but may not offer lifetime prevention, and the benefit in children not from AD families is unknown.

Other trials to prevent AD have included usage of dust avoidance and dust covers for mattresses. This strategy showed modest benefit in reducing the incidence of atopic diatheses in the first year3 but did not gain endorsement by the most recent guidelines of the American Academy of Dermatology (AAD).4

Prenatal and postnatal (maternal and child) supplementation of Lactobacillus rhamnosus has shown promise in prevention.5 The exact regimen likely makes an impact on efficacy. An early study showed the usage of probiotics (eg, Lactobacillus reuteri) prenatally in pregnant women and postnatally in infants resulted in no reduction in occurrence of AD and possible reduction in IgE-associated AD.6 Kalliomäki et al7 demonstrated that L rhamnosus GG alone reduced AD by half in at-risk infants in a double-blind, placebo-controlled trial. On the other hand, Taylor et al8 performed a study of probiotic supplementation in which patients at high risk for AD developed higher rates of allergen sensitization. The most successful recent trial involved the randomization of 415 pregnant women to receive interventions from 36 weeks’ gestation until 3 months postpartum.9 The intervention was a randomized comparison of milk without probiotics versus a blend of probiotic milk containing L rhamnosus GG, Lactobacillus acidophilus La-5, and Bifidobacterium animalis subsp lactis Bb-12. At 6 years of age, 81 babies who consumed probiotic milk and 82 babies who consumed milk without probiotics were available for testing. The strategy caused a statistically significant reduction in AD in the complete case analysis (odds ratio, 0.48; 95% confidence interval, 0.25-0.92; P=.027; number needed to treat, 6). Sadly, other allergic diseases were not prevented in this study.9

MANAGEMENT OF AD

There currently is no cure or perfected prevention technique for AD. As a result, therapy focuses on avoiding triggers and alleviating symptoms.10 Recent guidelines from the AAD state that“[t]he ultimate judgment regarding the propriety of any specific therapy must be made by the physician and the patient in light of all the circumstances presented by the individual patient, and the known variability and biologic behavior of the disease.”11 Skin-directed therapies are the first line of treatment including emollients, gentle skin care, and topical medicaments. In AD, therapies are needed to reduce disease activity and flare severity, clear flares, and provide relief.

Parental education and written eczema action plans are recommended to help patients and parents/guardians follow recommended regimens12; Tollefson and Bruckner13 for the American Academy of Pediatrics provide an action plan to guide the care of children with atopic dermatitis that is simple, but many others exist online. The eczema action plan usually provides information on how to bathe and what to do when the skin is actively inflamed.

In 2014, a 4-part series of guidelines of care for the management of AD was published by the AAD, replacing prior guidelines.4,11,14,15 The following sections review some of the important parameters of care highlighted in these management guidelines.

Psychological Support

Appropriate psychological support for AD patients can be sought through counselors, therapists, psychiatrists, and support groups such as the National Eczema Association (https://nationaleczema.org/).

Education

Education is the leading form of medical therapy in patients with AD. Eczema schools are popular in Europe and are just beginning to form in the United States (http://tuh.templehealth.org/content/eczema_school.htm), which can be helpful to educate caregivers and patients with AD. Patient resources online and through support groups with an online presence, in-person meetings, and patient/family conventions can be helpful to AD patients. Often, an initial office visit with a dermatologist involves a review of avoidance of triggers, usage of gentle skin care including bland emollients, and therapeutic regimens for disease activity. This form of verbal education is to be paired with an eczema action plan, a written document that allows individuals to reference recommendations and share information with other caregivers.12,13,16

Emollients and Gentle Skin Care

Gentle skin care regimens, which includes the usage of synthetic cleansers with a low pH to help maintain the acidity (acid mantle) of the skin, seek to reduce irritation and have been rated as level IA (highest level) in recent AAD guidelines.14 Although bathing frequency has been emphasized in the guidelines, AD severity as reflected by SCORAD (SCORing Atopic Dermatitis) was not different for daily bathing versus twice weekly.17 The American Academy of Pediatrics recommended a skin care regimen of bathing every 2 to 3 days in lukewarm water for 10 to 15 minutes, followed by application of emollients that are fragrance free and have few preservatives.13 Topical emollients with additives such as colloidal oatmeal, avenanthramides, or ceramides can be used to enhance the skin barrier and are well tolerated in all age groups.18,19 Despite enhanced emollients, the therapy of AD still requires usage of prescription or over-the-counter TCs and/or topical calcineurin inhibitors (TCIs) in many cases.20

 

 

Topical Medication

Children have a relatively higher body surface area–to-weight ratio, allowing for greater potential absorption of topical medicaments and potential side effects from absorption. Types of vehicle, cost, site of application, and availability may impact patient and physician preference in choice of therapeutic topical agent.14

Topical Corticosteroids
Topical corticosteroids (TCs) are the mainstay of treatment for AD and have been used for more than 60 years.14,20 Topical corticosteroids provide anti-inflammatory effects on T cells, monocytes, and macrophages, producing altered cytokine activity locally. Topical corticosteroids inhibit collagen synthesis, potentially causing skin atrophy. They also inhibit IL-1, IL-2, IL-6, IFN-α, and tumor necrosis factor α.21 Topical corticosteroids are classified as class I (ultra-high potency) to class VII (low potency). In children, low-potency TCs generally are applied to the face, intertriginous areas, groin, and genitalia, and mid-potency corticosteroids are applied to the body, arms, and legs. An even higher-strength agent can be prescribed as a rescue medication in severe cases. After clearance with once- or twice-daily therapy, twice-weekly usage can benefit disease activity.22 Topical corticosteroids reduce inflammation as well as Staphylococcus aureus load through inhibition of cytokines that inhibit antimicrobial peptides. Topical corticosteroids have been endorsed as level IA evidence therapy by the AAD guidelines.14

Topical corticosteroids, particularly prolonged usage of mid- to high-potency products, have been associated with side effects such as skin atrophy, striae, telangiectases, hypopigmentation, rosacea, acneiform eruptions, focal hypertrichosis, perioral dermatitis, and acne23; potential systemic side effects include hypothalamic-pituitary-adrenal axis suppression, cataracts, glaucoma (with periocular application), Cushing syndrome, hyperglycemia, hypertension,23 and growth retardation.14 Long-term corticosteroid therapy is associated with tachyphylaxis and potential rebound of disease with discontinuation.24 Based on the potential risk of side effects with TCs, the least potent product for the shortest time needed is recommended, with special care for thin skin. Discontinuation when clearance occurs is advised. Allergy to TCs and/or vehicle ingredients such as propylene glycol should be suspected in severe unremitting cases.14 A recent registry review of children screened for contact dermatitis demonstrated that children with AD had higher sensitization to the steroid tixocortol pivalate.25

Topical Calcineurin Inhibitors
Topical calcineurin inhibitors include pimecrolimus cream 1%, which is approved for mild to moderate AD in adults and children 2 years and older, and tacrolimus ointment 0.03% and 0.1%, which are approved for moderate to severe AD in adults and children aged 2 to 15 years (0.03% formulation only). Topical calcineurin inhibitors can be used as second-line agents in AD in patients who have inadequate response to TCs or who may not be able to use TCs due to the disease site.10,13,14 Guidelines from the AAD also have endorsed TCIs as level IA evidence for steroid-sparing agents.

Concerns about the reporting of cancers and lymphomas prompted the US Food and Drug Administration to issue a black box warning on TCIs more than 10 years ago. Pimecrolimus, which has little absorption and no notable immunosuppressive effects, has been used without detrimental effect on vaccination and delayed-type hypersensitivities, but many decades of data are lacking.10,13,14,17,26-29 Topical calcineurin inhibitors can be used as steroid-sparing agents in lieu of corticosteroids in specific locations such as the face and eyelids and for long-term suppressive therapy twice weekly.30 Intermittent usage and cycling with corticosteroids is advisable,28 but usage intermittently beyond 1 year has not been evaluated.

Topical calcineurin inhibitors are recommended as effective for acute and chronic AD. Their use as maintenance therapy in adults and children, for AD recalcitrant to steroids, for AD in sensitive areas, for steroid-induced atrophy, and for long-term uninterrupted topical steroid usage carries a level IA evidence recommendation. Furthermore, the AAD guidelines have recommended TCIs as steroid-sparing agents with level IA evidence and off-label use of TCIs in children younger than 2 years with level IA evidence. Pretreatment with TCs to reduce stinging has level IIB evidence. Usage for flare prevention is level IA evidence. Routine blood monitoring of TCI-treated patients was not recommended; in fact, the AAD guidelines provided this recommendation as level IA evidence against routine laboratory monitoring of TCI-treated patients.14

Topical Antibiotics
Topical antibiotics are indicated for the therapy of impetigo and can be used in the setting of impetiginized AD in conjunction with TCs. Recent AAD guidelines suggested against routine usage of topical antistaphylococcal agents as level IA evidence.14 There is one study supporting usage of topical mupirocin in addition to TCs to heal children with eczema area and severity index scores more than 7 more rapidly in the first week of AD therapy, but in the same study, additive benefit was not demonstrated in AD beyond the first week.31 There also are data supporting usage of intranasal mupirocin adjunctively with bleach baths in patients with moderate to severe AD, which was rated as level IIB evidence in the AAD guidelines.14,32 There are limited data on the long-term utility of topical anti-infectives in AD. The risks of long-term usage could include resistance formation to agents such as mupirocin, contact dermatitis, and lack of efficacy.

 

 

Additional Therapeutics

Wet Wraps
Penetration through the stratum corneum is needed for drug activity in AD. Penetration can be enhanced using wet wrap therapy or using ointments, which produce higher relative potency.13 Wet wraps overlying a dilute topical medicament have been described as effective in AD and are recommended in AAD guidelines as level IIB evidence.14 Different wet wrap techniques can be used, including wet pajamas covered by dry pajamas or saline-soaked gauze wrapped around the affected areas and then dry gauze applied over the wet gauze. The methodology used should be tailored to the patient as well as to whether the individual is an inpatient or outpatient.

Bleach Baths
Dilute sodium hypochlorite solution 0.005% (one-quarter cup bleach in 20 gallons of water) has been demonstrated to be beneficial in reduction of disease activity in AD patients with recurrent bacterial infections.32 This simple technique in addition to intranasal mupirocin can reduce AD severity and improve quality of life and is the only ongoing S aureus therapeutic management endorsed by the AAD guidelines for the management of AD.14,32

Topical and Oral Delivery

Antihistamines
Topical antihistamines are ineffective in AD. Oral antihistamines can be used to reduce pruritus and are most effective when given as sedating agents for sleep enhancement but may be given as nonsedating agents for patients with concomitant allergic disorders such as allergic rhinoconjunctivitis. Paradoxical hyperreactivity with sedating antihistamines is not uncommon in small children, and sedating antihistamine usage should be discontinued in these instances.13 Parents of children with AD have reported giving the child antihistamines to sleep was helpful, as well as putting on creams, using special clothes (eg, all cotton), and keeping the room cool.33 There is level IIIC evidence against use of systemic antihistamines and level IIA evidence for sedating and nonsedating, according to the AAD guidelines.14

Systemic Therapeutics

Oral therapeutics range from oral antihistamines to oral antibiotics and immunosuppressive medications. Oral antibiotics (level IIB evidence) are reserved for superinfected AD, which is not easily defined for the following reasons: there is no consensus definition of superinfected AD; the majority of active AD lesions when cultured will demonstrate S aureus growth; and most AD lesions ooze, thereby creating the appearance of superinfection. In real-world practice, superinfection can be diagnosed based on the presence of pustules; furuncles; or signs of infection such as tracking erythema, tenderness, severe erosions, or maceration. Clinical judgment is always required.

The immunosuppressive medications used in AD include leukotriene inhibitors, which rarely are effective for AD.34 More effective systemic agents for AD include cyclosporine (level I to IIB evidence), azathioprine (level IIB evidence), mycophenolate mofetil (level IIIC evidence), and methotrexate (level IIB evidence). These agents are indicated for pediatric or adult patients when topical agents and/or phototherapy have failed.15 Monitoring these agents for side effects includes ongoing evaluation for renal and liver toxicity. Short courses (ie, 6 months) are preferred to minimize side effects.35

Dupilumab, an injectable AD therapy, is approved in the United States. This agent is injected every 2 weeks and binds to the IL-4Rα shared by IL-4 and IL-13. In 4 weeks of monotherapy, 85% of adult patients treated had 50% or greater clearance.36 Recently published consensus opinion from the International Eczema Council recommends assessment of a variety of factors before initiating systemic therapy including comorbid illnesses such as contact allergy, trigger avoidance, superinfection, and impact on quality of life.37

Oral Corticosteroids
Systemic corticosteroids clear patients quickly but provide no sustained improvement; in fact, many patients rebound or have tachyphylaxis. Although short-term corticosteroid usage can break the itch-scratch cycle, long-term usage is associated with osteoporosis, Cushing syndrome, and aseptic necrosis of the femoral head. Decreased linear growth will occur during therapy in children; therefore, systemic steroids are not recommended in children with AD, except for additional or comorbid conditions (eg, asthma or contact dermatitis).4

Phototherapy
Phototherapy has been recommended in the AAD guidelines as a second-line treatment after failure of first-line agents (ie, TCIs and TCs) for clearance and or maintenance and should be tailored to the patient’s skin tone by an experienced physician. Narrowband UVB phototherapy may act through the suppression of T-cell activity in the skin and possibly via suppression of staphylococcal superantigens; however, many phototherapy types have been described for AD.38,39 Usage can be effective in school-aged children and teenagers but may be limited due to school attendance. Phototherapy was graded as level IIB evidence in the AAD guidelines.15 Side effects include aggravation of AD by exposure to heat and UV light, actinic damage, tenderness, erythema, pruritus, burning, and stinging. Lentigines; skin cancers (melanoma and nonmelanoma); folliculitis; and ocular toxicity, especially cataracts, can occur.15 Children younger than 6 years will find it difficult to stand in a phototherapy booth and may be poor candidates.15,38,39

 

 

Complementary and Alternative Medicine

Complementary and alternative medicine (CAM) also has been used for AD in the United States. In a review of the 2007 National Health Interview Survey of 9417 children aged 0 to 17 years, CAM was used for AD by 0.99% of children. Some CAM techniques were associated with worsening severity of AD, including herbal therapy, vitamins, homeopathic agents, diet, and movement techniques.40 Usage of Chinese herbal medications for AD can be associated with liver toxicity.41 Only one CAM therapy—massage therapy—has some mild supportive data.42

Allergen Avoidance and Diet
Bronsnick et al43 discussed the possible benefit of prenatal and postnatal probiotics for prevention of AD, which were not supported in the AAD guidelines for management of AD4; postnatal prebiotic supplementation; and exclusive breastfeeding and/or supplementation with hydrolyzed formula in at-risk children. Elimination diets for children and mothers were not recommended. The authors found no beneficial role of supplements including vitamin D, selenium, fish oil, borage oil, and zinc sulfate.43

A National Institute of Allergy and Infectious Diseases consensus group recommended avoidance of proven but not random elimination of food allergens in AD, asthma, and/or eosinophilic esophagitis.44 Restricted maternal diet was not recommended, and breastfeeding exclusively for the first 4 to 6 months was recommended. Hydrolyzed formulas were suggested as a possible preventive strategy in at-risk infants as a breastfeeding alternative, with cost of these formulas being a problem.44

In children younger than 5 years, food allergy screening for the most common allergens (eg, milk, eggs, peanuts, wheat, soy) should be considered in children with persistent unremitting dermatitis and/or known food challenge–induced reactions.4 Conservative measures to avoid house dust mite exposure in known sensitized individuals including dust covers for pillows and mattresses may be beneficial.4,45

Emerging Therapies

Recently approved therapies include better-targeted agents that appear to have a reasonable safety profile and may fulfill unmet needs in AD care. Of these agents, crisaborole, a topical boron-based phosphodiesterase 4 inhibitor, was approved in December 2016 for mild to moderate AD in patients 2 years and older.Topically, this agent seems to be efficacious in the absence of notable carcinogenicity.46

The systemic (injectable) biologic agent dupilumab was approved in March 2017 for moderate to severe AD. Phase 3 studies in adults with AD showed excellent success in adults with moderate to severe AD.37 This agent is a monoclonal antibody targeted at blockade of the crucial atopic inflammatory triggering pathway via blockade of the IL-4A receptor site, targeting IL-4 and IL-13 activity.36,47 There are many medications in the pipeline, which Renert-Yuval and Guttman-Yassky48 review. However, an overview of the landscape demonstrates that Janus kinase (JAK) inhibitors49 and biologic medications in addition to dupilumab affecting targeted inflammatory cascades in AD are in development. In particular, the JAK inhibitors appear promising due to availability both as oral and topical agents.49

Need for Ongoing Care and Monitoring

Atopic dermatitis is a chronic inflammatory skin disorder with a genetic basis. Once initiated, the process of AD may persist throughout the patient’s life and become a systemic disorder with comorbidities including sleep disturbance, reduced quality of life, and cardiovascular disease.50 Ongoing management of AD includes topical reduction in irritants and triggers, topical medicaments, and management of pruritus and infections. At this time, emollients and irritant avoidance paired with judicious topical medicaments including TCs and second-line or site-specific (eg, eyelids) usage of TCIs or phosphodiesterase 4 inhibitors remain the backbone of therapy. Ongoing review of therapeutics for associated morbidities is underway, which may guide future therapeutic interventions into AD. The future of prevention and therapy look bright, but time will tell.

References
  1. Kelleher M, Dunn-Galvin A, Hourihane JO, et al. Skin barrier dysfunction measured by transepidermal water loss at 2 days and 2 months predates and predicts atopic dermatitis at 1 year. J Allergy Clin Immunol. 2015;135:930-935.
  2. Simpson EL, Chalmers JR, Hanifin JM, et al. Emollient enhancement of the skin barrier from birth offers effective atopic dermatitis prevention. J Allergy Clin Immunol. 2014;134:818-823.
  3. Tsitoura S, Nestoridou K, Botis P, et al. Randomized trial to prevent sensitization to mite allergens in toddlers and preschoolers by allergen reduction and education: one-year results. Arch Pediatr Adolesc Med. 2002;156:1021-1027.
  4. Sidbury R, Tom WL, Bergman JN, et al. Guidelines of care for the management of atopic dermatitis: section 4. prevention of disease flares and use of adjunctive therapies and approaches. J Am Acad Dermatol. 2014;71:1218-1233.
  5. Foolad N, Brezinski EA, Chase EP, et al. Effect of nutrient supplementation on atopic dermatitis in children: a systematic review of probiotics, prebiotics, formula, and fatty acids. JAMA Dermatol. 2013;149:350-355.
  6. Abrahamsson TR, Jakobsson T, Böttcher MF, et al. Probiotics in prevention of IgE-associated eczema: a double-blind, randomized, placebo-controlled trial. J Allergy Clin Immunol. 2007;119:1174-1180.
  7. Kalliomäki M, Salminen S, Arvilommi H, et al. Probiotics in primary prevention of atopic disease: a randomised placebo-controlled trial. Lancet. 2001;357:1076-1079.
  8. Taylor AL, Dunstan JA, Prescott SL. Probiotic supplementation for the first 6 months of life fails to reduce the risk of atopic dermatitis and increases the risk of allergen sensitization in high-risk children: a randomized controlled trial. J Allergy Clin Immunol. 2007;119:184-191.
  9. Simpson MR, Dotterud CK, Storrø O, et al. Perinatal probiotic supplementation in the prevention of allergy related disease: 6 year follow up of a randomised controlled trial. BMC Dermatol. 2015;15:13. doi:10.1186/s12895-015-0030-1.
  10. Carr WW. Topical calcineurin inhibitors for atopic dermatitis: review and treatment recommendations. Paediatr Drugs. 2013;15:303-310.
  11. Eichenfield LF, Tom WL, Chamlin SL, et al. Guidelines of care for the management of atopic dermatitis: section 1. diagnosis and assessment of atopic dermatitis. J Am Acad Dermatol. 2014;70:338-351.
  12. Silverberg NB. Creating an action plan for eczema patients. Cutis. 2015;96:362-363.
  13. Tollefson MM, Bruckner AL; Section on Dermatology. Atopic dermatitis: skin-directed management. Pediatrics. 2014;134:E1735-E1744.
  14. Eichenfield LF, Tom WL, Berger TG, et al. Guidelines of care for the management of atopic dermatitis: section 2. management and treatment of atopic dermatitis with topical therapies. J Am Acad Dermatol. 2014;71:116-132.
  15. Sidbury R, Davis DM, Cohen DE, et al; American Academy of Dermatology. Guidelines of care for the management of atopic dermatitis: section 3. management and treatment with phototherapy and systemic agents. J Am Acad Dermatol. 2014;71:327-349.
  16. Shi VY, Nanda S, Lee K, et al. Improving patient education with an eczema action plan: a randomized controlled trial. JAMA Dermatol. 2013;149:481-483.
  17. Koutroulis I, Petrova K, Kratimenos P, et al. Frequency of bathing in the management of atopic dermatitis: to bathe or not to bathe? Clin Pediatr (Phila). 2014;53:677-681.
  18. Fowler JF, Nebus J, Wallo W, et al. Colloidal oatmeal formulations as adjunct treatments in atopic dermatitis. J Drugs Dermatol. 2012;11:804-807.
  19. Fowler J Jr, Silverberg N. Active naturals have a key role in atopic dermatitis. Semin Cutan Med Surg. 2008;27:8-10.
  20. Eichenfield LF. Consensus guidelines in diagnosis and treatment of atopic dermatitis. Allergy. 2004;59:86-92.
  21. Nghiem P, Pearson G, Langley RG. Tacrolimus and pimecrolimus: from clever prokaryotes to inhibiting calcineurin and treating atopic dermatitis. J Am Acad Dermatol. 2002;46:228-241.
  22. Schmitt J. Commentary: eczema and cancer risk. Br J Dermatol. 2011;165:463-464.
  23. Abramovits W, Hung P, Tong KB. Efficacy and economics of topical calcineurin inhibitors for the treatment of atopic dermatitis. Am J Clin Dermatol. 2006;7:213-222.
  24. Takahashi-Ando N, Jones MA, Fujisawa S, et al. Patient-reported outcomes after discontinuation of long-term topical corticosteroid treatment for atopic dermatitis: a targeted cross-sectional survey. Drug Healthc Patient Saf. 2015;7:57-62.
  25. Jacob SE, McGowan M, Silverberg NB, et al. Pediatric contact dermatitis registry data on contact allergy in children with atopic dermatitis. JAMA Dermatol. 2017;153:765-770.
  26. Werfel T. Topical use of pimecrolimus in atopic dermatitis: update on the safety and efficacy. J Dtsch Dermatol Ges. 2009;7:739-742.
  27. Wahn U, Bos JD, Goodfield M, et al. Efficacy and safety of pimecrolimus cream in the long-term management of atopic dermatitis in children. Pediatrics. 2002;110(1, pt 1):E2.
  28. Berger TG, Duvic M, Van Voorhees AS, et al; American Academy of Dermatology Association Task Force. The use of topical calcineurin inhibitors in dermatology: safety concerns. report of the American Academy of Dermatology Association Task Force. J Am Acad Dermatol. 2006;54:818-823.
  29. Paller AS. Latest approaches to treating atopic dermatitis. Chem Immunol Allergy. 2012;96:132-140.
  30. Thaçi D, Reitamo S, Gonzalez Ensenat MA, et al. Proactive disease management with 0.03% tacrolimus ointment for children with atopic dermatitis: results of a randomized, multicentre, comparative study. Br J Dermatol. 2008;159:1348-1356.
  31. Gong JQ, Lin L, Lin T, et al. Skin colonization by Staphylococcus aureus in patients with eczema and atopic dermatitis and relevant combined topical therapy: a double-blind multicentre randomized controlled trial. Br J Dermatol. 2006;155:680-687.
  32. Huang JT, Abrams M, Tlougan B, et al. Treatment of Staphylococcus aureus colonization in atopic dermatitis decreases disease severity. Pediatrics. 2009;123:E808-E814.
  33. Reid P, Lewis-Jones MS. Sleep difficulties and their management in preschoolers with atopic eczema. Clin Exp Dermatol. 1995;20:38-41.
  34. Silverberg NB, Paller AS. Leukotriene receptor antagonists are ineffective for severe atopic dermatitis. J Am Acad Dermatol. 2004;50:485-486.
  35. Wolverton SE. Comprehensive Dermatologic Drug Therapy. 3rd ed. New York, NY: Elsevier Saunders; 2013.
  36. Beck LA, Thaçi D, Hamilton JD, et al. Dupilumab treatment in adults with moderate-to-severe atopic dermatitis. N Engl J Med. 2014;371:130-139.
  37. Simpson EL, Bruin-Weller M, Flohr C, et al. When does atopic dermatitis warrant systemic therapy? recommendations from an expert panel of the International Eczema Council [published online August 10, 2017]. J Am Acad Dermatol. doi:10.1016/j.jaad.2017.06.042.
  38. Veith W, DeLeo V, Silverberg N. Medical phototherapy in childhood skin diseases. Minerva Pediatr. 2011;63:327-333.
  39. Song E, Reja D, Silverberg N, et al. Phototherapy: kids are not just little people. Clin Dermatol. 2015;33:672-680.
  40. Silverberg JI, Lee-Wong M, Silverberg NB. Complementary and alternative medicines and childhood eczema: a US population-based study. Dermatitis. 2014;25:246-254.
  41. Stickel F, Shouval D. Hepatotoxicity of herbal and dietary supplements: an update. Arch Toxicol. 2015;89:851-865.
  42. Schachner L, Field T, Hernandez-Reif M, et al. Atopic dermatitis symptoms decreased in children following massage therapy. Pediatr Dermatol. 1998;15:390-395.
  43. Bronsnick T, Murzaku EC, Rao BK. Diet in dermatology: part I. atopic dermatitis, acne, and nonmelanoma skin cancer. J Am Acad Dermatol. 2014;71:1039.e1-1039.e12.
  44. Boyce JA, Assa’ad A, Burks AW, et al. Guidelines for the diagnosis and management of food allergy in the United States: summary of the NIAID-sponsored expert panel report. Nutr Res. 2011;31:61-75.
  45. Silverberg NB, Lee-Wong M, Yosipovitch G. Diet and atopic dermatitis. Cutis. 2016;97:227-232.
  46. Hanifin JM, Chan SC, Cheng JB, et al. Type phosphodiesterase inhibitors have clinical and in vitro anti-inflammatory effects in atopic dermatitis. J Invest Dermatol. 1996;107:51-56.
  47. Boguniewicz M, Leung DY. Targeted therapy for allergic diseases: at the intersection of cutting-edge science and clinical practice. J Allergy Clin Immunol. 2015;135:354-356.
  48. Renert-Yuval Y, Guttman-Yassky E. Systemic therapies in atopic dermatitis: the pipeline. Clin Dermatol. 2017;35:387-397.
  49. Damsky W, King BA. JAK inhibitors in dermatology: the promise of a new drug class. J Am Acad Dermatol. 2017;76:736-744.
  50. Brunner PM, Silverberg JI, Guttman-Yassky E, et al. Increasing comorbidities suggest that atopic dermatitis is a systemic disorder. J Invest Dermatol. 2017;137:18-25.
References
  1. Kelleher M, Dunn-Galvin A, Hourihane JO, et al. Skin barrier dysfunction measured by transepidermal water loss at 2 days and 2 months predates and predicts atopic dermatitis at 1 year. J Allergy Clin Immunol. 2015;135:930-935.
  2. Simpson EL, Chalmers JR, Hanifin JM, et al. Emollient enhancement of the skin barrier from birth offers effective atopic dermatitis prevention. J Allergy Clin Immunol. 2014;134:818-823.
  3. Tsitoura S, Nestoridou K, Botis P, et al. Randomized trial to prevent sensitization to mite allergens in toddlers and preschoolers by allergen reduction and education: one-year results. Arch Pediatr Adolesc Med. 2002;156:1021-1027.
  4. Sidbury R, Tom WL, Bergman JN, et al. Guidelines of care for the management of atopic dermatitis: section 4. prevention of disease flares and use of adjunctive therapies and approaches. J Am Acad Dermatol. 2014;71:1218-1233.
  5. Foolad N, Brezinski EA, Chase EP, et al. Effect of nutrient supplementation on atopic dermatitis in children: a systematic review of probiotics, prebiotics, formula, and fatty acids. JAMA Dermatol. 2013;149:350-355.
  6. Abrahamsson TR, Jakobsson T, Böttcher MF, et al. Probiotics in prevention of IgE-associated eczema: a double-blind, randomized, placebo-controlled trial. J Allergy Clin Immunol. 2007;119:1174-1180.
  7. Kalliomäki M, Salminen S, Arvilommi H, et al. Probiotics in primary prevention of atopic disease: a randomised placebo-controlled trial. Lancet. 2001;357:1076-1079.
  8. Taylor AL, Dunstan JA, Prescott SL. Probiotic supplementation for the first 6 months of life fails to reduce the risk of atopic dermatitis and increases the risk of allergen sensitization in high-risk children: a randomized controlled trial. J Allergy Clin Immunol. 2007;119:184-191.
  9. Simpson MR, Dotterud CK, Storrø O, et al. Perinatal probiotic supplementation in the prevention of allergy related disease: 6 year follow up of a randomised controlled trial. BMC Dermatol. 2015;15:13. doi:10.1186/s12895-015-0030-1.
  10. Carr WW. Topical calcineurin inhibitors for atopic dermatitis: review and treatment recommendations. Paediatr Drugs. 2013;15:303-310.
  11. Eichenfield LF, Tom WL, Chamlin SL, et al. Guidelines of care for the management of atopic dermatitis: section 1. diagnosis and assessment of atopic dermatitis. J Am Acad Dermatol. 2014;70:338-351.
  12. Silverberg NB. Creating an action plan for eczema patients. Cutis. 2015;96:362-363.
  13. Tollefson MM, Bruckner AL; Section on Dermatology. Atopic dermatitis: skin-directed management. Pediatrics. 2014;134:E1735-E1744.
  14. Eichenfield LF, Tom WL, Berger TG, et al. Guidelines of care for the management of atopic dermatitis: section 2. management and treatment of atopic dermatitis with topical therapies. J Am Acad Dermatol. 2014;71:116-132.
  15. Sidbury R, Davis DM, Cohen DE, et al; American Academy of Dermatology. Guidelines of care for the management of atopic dermatitis: section 3. management and treatment with phototherapy and systemic agents. J Am Acad Dermatol. 2014;71:327-349.
  16. Shi VY, Nanda S, Lee K, et al. Improving patient education with an eczema action plan: a randomized controlled trial. JAMA Dermatol. 2013;149:481-483.
  17. Koutroulis I, Petrova K, Kratimenos P, et al. Frequency of bathing in the management of atopic dermatitis: to bathe or not to bathe? Clin Pediatr (Phila). 2014;53:677-681.
  18. Fowler JF, Nebus J, Wallo W, et al. Colloidal oatmeal formulations as adjunct treatments in atopic dermatitis. J Drugs Dermatol. 2012;11:804-807.
  19. Fowler J Jr, Silverberg N. Active naturals have a key role in atopic dermatitis. Semin Cutan Med Surg. 2008;27:8-10.
  20. Eichenfield LF. Consensus guidelines in diagnosis and treatment of atopic dermatitis. Allergy. 2004;59:86-92.
  21. Nghiem P, Pearson G, Langley RG. Tacrolimus and pimecrolimus: from clever prokaryotes to inhibiting calcineurin and treating atopic dermatitis. J Am Acad Dermatol. 2002;46:228-241.
  22. Schmitt J. Commentary: eczema and cancer risk. Br J Dermatol. 2011;165:463-464.
  23. Abramovits W, Hung P, Tong KB. Efficacy and economics of topical calcineurin inhibitors for the treatment of atopic dermatitis. Am J Clin Dermatol. 2006;7:213-222.
  24. Takahashi-Ando N, Jones MA, Fujisawa S, et al. Patient-reported outcomes after discontinuation of long-term topical corticosteroid treatment for atopic dermatitis: a targeted cross-sectional survey. Drug Healthc Patient Saf. 2015;7:57-62.
  25. Jacob SE, McGowan M, Silverberg NB, et al. Pediatric contact dermatitis registry data on contact allergy in children with atopic dermatitis. JAMA Dermatol. 2017;153:765-770.
  26. Werfel T. Topical use of pimecrolimus in atopic dermatitis: update on the safety and efficacy. J Dtsch Dermatol Ges. 2009;7:739-742.
  27. Wahn U, Bos JD, Goodfield M, et al. Efficacy and safety of pimecrolimus cream in the long-term management of atopic dermatitis in children. Pediatrics. 2002;110(1, pt 1):E2.
  28. Berger TG, Duvic M, Van Voorhees AS, et al; American Academy of Dermatology Association Task Force. The use of topical calcineurin inhibitors in dermatology: safety concerns. report of the American Academy of Dermatology Association Task Force. J Am Acad Dermatol. 2006;54:818-823.
  29. Paller AS. Latest approaches to treating atopic dermatitis. Chem Immunol Allergy. 2012;96:132-140.
  30. Thaçi D, Reitamo S, Gonzalez Ensenat MA, et al. Proactive disease management with 0.03% tacrolimus ointment for children with atopic dermatitis: results of a randomized, multicentre, comparative study. Br J Dermatol. 2008;159:1348-1356.
  31. Gong JQ, Lin L, Lin T, et al. Skin colonization by Staphylococcus aureus in patients with eczema and atopic dermatitis and relevant combined topical therapy: a double-blind multicentre randomized controlled trial. Br J Dermatol. 2006;155:680-687.
  32. Huang JT, Abrams M, Tlougan B, et al. Treatment of Staphylococcus aureus colonization in atopic dermatitis decreases disease severity. Pediatrics. 2009;123:E808-E814.
  33. Reid P, Lewis-Jones MS. Sleep difficulties and their management in preschoolers with atopic eczema. Clin Exp Dermatol. 1995;20:38-41.
  34. Silverberg NB, Paller AS. Leukotriene receptor antagonists are ineffective for severe atopic dermatitis. J Am Acad Dermatol. 2004;50:485-486.
  35. Wolverton SE. Comprehensive Dermatologic Drug Therapy. 3rd ed. New York, NY: Elsevier Saunders; 2013.
  36. Beck LA, Thaçi D, Hamilton JD, et al. Dupilumab treatment in adults with moderate-to-severe atopic dermatitis. N Engl J Med. 2014;371:130-139.
  37. Simpson EL, Bruin-Weller M, Flohr C, et al. When does atopic dermatitis warrant systemic therapy? recommendations from an expert panel of the International Eczema Council [published online August 10, 2017]. J Am Acad Dermatol. doi:10.1016/j.jaad.2017.06.042.
  38. Veith W, DeLeo V, Silverberg N. Medical phototherapy in childhood skin diseases. Minerva Pediatr. 2011;63:327-333.
  39. Song E, Reja D, Silverberg N, et al. Phototherapy: kids are not just little people. Clin Dermatol. 2015;33:672-680.
  40. Silverberg JI, Lee-Wong M, Silverberg NB. Complementary and alternative medicines and childhood eczema: a US population-based study. Dermatitis. 2014;25:246-254.
  41. Stickel F, Shouval D. Hepatotoxicity of herbal and dietary supplements: an update. Arch Toxicol. 2015;89:851-865.
  42. Schachner L, Field T, Hernandez-Reif M, et al. Atopic dermatitis symptoms decreased in children following massage therapy. Pediatr Dermatol. 1998;15:390-395.
  43. Bronsnick T, Murzaku EC, Rao BK. Diet in dermatology: part I. atopic dermatitis, acne, and nonmelanoma skin cancer. J Am Acad Dermatol. 2014;71:1039.e1-1039.e12.
  44. Boyce JA, Assa’ad A, Burks AW, et al. Guidelines for the diagnosis and management of food allergy in the United States: summary of the NIAID-sponsored expert panel report. Nutr Res. 2011;31:61-75.
  45. Silverberg NB, Lee-Wong M, Yosipovitch G. Diet and atopic dermatitis. Cutis. 2016;97:227-232.
  46. Hanifin JM, Chan SC, Cheng JB, et al. Type phosphodiesterase inhibitors have clinical and in vitro anti-inflammatory effects in atopic dermatitis. J Invest Dermatol. 1996;107:51-56.
  47. Boguniewicz M, Leung DY. Targeted therapy for allergic diseases: at the intersection of cutting-edge science and clinical practice. J Allergy Clin Immunol. 2015;135:354-356.
  48. Renert-Yuval Y, Guttman-Yassky E. Systemic therapies in atopic dermatitis: the pipeline. Clin Dermatol. 2017;35:387-397.
  49. Damsky W, King BA. JAK inhibitors in dermatology: the promise of a new drug class. J Am Acad Dermatol. 2017;76:736-744.
  50. Brunner PM, Silverberg JI, Guttman-Yassky E, et al. Increasing comorbidities suggest that atopic dermatitis is a systemic disorder. J Invest Dermatol. 2017;137:18-25.
Issue
Cutis - 100(3)
Issue
Cutis - 100(3)
Page Number
173-177, 192
Page Number
173-177, 192
Publications
Publications
Topics
Article Type
Display Headline
Atopic Dermatitis Prevention and Treatment
Display Headline
Atopic Dermatitis Prevention and Treatment
Sections
Inside the Article

Practice Points

  • Prevention of atopic dermatitis is desired in high-risk settings (ie, 1 or more relatives with atopy).
  • Emollient therapy from early infancy has been described as one method.
  • Other forms of disease prevention have not yet been adequately developed.
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Article PDF Media

2017 Update on female sexual dysfunction

Article Type
Changed
Thu, 12/15/2022 - 17:52
Display Headline
2017 Update on female sexual dysfunction

Illustration: Kimberly Martens for OBG Management
Neurologic functions, hormonal regulation, and psychological factors affect sexual desire and arousal to some extent. Menopause, and the genitourinary symptoms associated with it, also affect sexual function. Understanding the pathogenesis of sexual dysfunction is key to management decisions.

Sexual function is a complex, multifaceted process mediated by neurologic functions, hormonal regulation, and psychological factors. What could possibly go wrong?

As it turns out, quite a lot. Female sexual dysfunction is a common, vastly undertreated sexual health problem that can have wide-reaching effects on a woman’s life. These effects may include impaired body image, self-confidence, and self-worth. Sexual dysfunction also can contribute to relationship dissatisfaction and leave one feeling less connected with her partner.1,2 Studies have shown women with sexual dysfunction have higher health care expenditures3 and that depression and fatigue are common comorbidities, as is frequently seen in other chronic conditions such as diabetes and back pain.4

Understanding the pathogenesis of female sexual dysfunction helps to guide our approach to its management. Indeed, increased understanding of its pathology has helped to usher in new and emerging treatment options, as well as a personalized, biopsychosocial approach to its management.

 

Related article:
2016 Update on female sexual dysfunction

 

In this Update, I discuss the interplay of physiologic and psychological factors that affect female sexual function as well as the latest options for its management. I have also assembled a panel of experts to discuss 2 cases representative of sexual dysfunction that you may encounter in your clinical practice and how prescribing decisions are made for their management.

 

Read about factors that impact sexual function and agents to help manage dysfunction.

 

 

Multiple transmitters in the brain can increase or decrease sexual desire and function

Neurotransmitters involved in sexual excitation include brain dopamine, melanocortin, oxytocin, vasopressin, and norepinephrine, whereas brain opioids, serotonin, prolactin, and endocannabinoids function as sexual inhibitors. Inhibitory transmitters are activated normally during sexual refractoriness but also from primary aversion or secondary avoidance disorders.1 Drugs or conditions that reduce brain dopamine levels, increase the action of brain serotonin, or enhance brain opioid pathways have been shown to inhibit sexual desire, while those that increase hypothalamic and mesolimbic dopamine or decrease serotonin release have been shown to stimulate sexual desire.1 

Estradiol and progesterone can impact sexual function and desire

In addition to the neurotransmitters, hormones are important modulators of female sexual function. Decreasing levels of circulating estrogen after menopause lead to physiologic, biologic, and clinical changes in the urogenital tissues, such as decreased elastin, thinning of the epithelium, reduced vaginal blood flow, diminished lubrication, and decreased flexibility and elasticity. These changes result in the symptoms of genitourinary syndrome of menopause (GSM), which affects as many as half of all menopausal women.5,6 In clinical trials, dyspareunia and vaginal dryness are the most bothersome GSM symptoms reported.7

The role of hormonal regulation in sexual dysfunction among premenopausal women is not yet fully understood, but we do know that estradiol has been shown to improve sexual desire, progesterone tends to dampen sexual desire, and that testosterone at physiological levels has been shown in most studies to have a neutral effect on sexual desire in a well-estrogenized patient.8

 

Related article:
Focus on treating genital atrophy symptoms

 

Experience and behavior modulate or reinforce sexual dysfunction

The most common psychological factors that trigger or amplify female sexual dysfunction are depression, anxiety, distraction, negative body image, sexual abuse, and emotional neglect.9 Contextual or sociocultural factors, such as relationship discord, life-stage stressors (the empty nest syndrome or anxiety and sleep deprivation from a new baby), as well as cultural or religious values that suppress sexuality, also should be considered.9 Experience-based neuroplasticity (changes in brain pathways that become solidified by negative or positive experiences) may elucidate how a multimodal approach, utilizing medical and psychological treatment, can be beneficial for patients, particularly those with hypoactive sexual desire disorder (HSDD).1

New and emerging approaches to managing female sexual dysfunction

Three agents, one of which has been available for prescription for some time, one that is newly available, and one in the pipeline, are or may soon be in the gynecologist's armamentarium.

Flibanserin

Medications that target excitatory pathways or blunt inhibitory pathways are in development, and one, flibanserin (Addyi), has been US Food and Drug Administration (FDA)-approved for the treatment of acquired, generalized HSDD in premenopausal women.1,10 Flibanserin is a nonhormonal, centrally acting, postsynaptic serotonin 1A receptor agonist and a serotonin 2A receptor antagonist that is taken daily at bedtime (100 mg); several weeks are usually needed before any effects are noted.1 It is not approved for postmenopausal women and has a boxed warning about the risks of hypotension and syncope; its use is contraindicated in women who drink alcohol, in those who have hepatic impairment, and with the use of moderate or strong CYP3A4 inhibitors.11

Also keep in mind that flibanserin is only available through a Risk Evaluation and Mitigation Strategy program, so clinicians who wish to prescribe it must enroll in and complete training to become certified providers.9

 

Related article:
What you need to know (and do) to prescribe the new drug flibanserin

 

Prasterone

Prasterone (Intrarosa), a once-daily intravaginal dehydroepiandrosterone (DHEA) product, is a prohormone that increases local estrogen and testosterone and has the advantage of improved sexual function, desire, arousal, lubrication, orgasm, satisfaction, as well as pain at sexual activity.12 It was approved by the FDA in November 2016 to treat moderate to severe dyspareunia and has been available for prescribing since July 2017. Its cost is comparable to topical estrogen products, with a $25 copay program.

Because prasterone is not an estrogen, it does not have the boxed warning that all estrogen products are mandated by the FDA to have. This may make it more acceptable to patients, who often decline to use an estrogen product after seeing the boxed warning on the package. The Centers for Medicare and Medicaid Services (CMS) does not have prasterone on its list of potentially hazardous drugs for the elderly. However, keep in mind that because its label is for dyspareunia and not specifically for GSM, CMS considers it a drug of choice--in other words, like sildenafil (Viagra), a lifestyle choice and not for treatment of a medical condition. As such, at the present time, Medicare does not cover it.

Bremelanotide

Late-stage trials of bremelanotide, a melanocortin receptor agonist, are underway. Its mechanism of action is somewhat like that of flibanserin in that both drugs increase dopamine and norepinephrine levels. The advantage of bremelanotide is that it is used as needed. It is dosed subcutaneously (1.75 mg) and it can be used as often as a woman would like to use it. The FDA is expected to consider it for approval in about a year. Unpublished data from poster sessions at recent meetings show that, in a phase 3 study of 1,247 premenopausal women with HSDD (who had already been screened for depression and were found to have a physiologic condition), improvements in desire, arousal, lubrication, and orgasm were shown with bremelanotide. About 18% of women stopped using the drug because of adverse effects (nausea, vomiting, flushing, or headache) versus 2% for placebo. Like flibanserin, it is expected to be approved for premenopausal women only. 

 

Read how 3 experts would manage differing GSM symptoms.

 

 

What would you prescribe for these patients? 


CASE Genitourinary syndrome of menopause (GSM) in a 55-year-old woman

A 55-year-old widow is beginning a new relationship. She has not had partnered sexual activity for several years, but she recently has begun a relationship. She describes pain with attempted penetration with her new partner. Her last menstrual period was 3 years ago and she has experienced very minor menopausal symptoms, which are not bothersome. On examination, the vulva and vagina are pale, with thin epithelium and absent rugae. The tissue lacks elasticity. A virginal speculum is needed to visualize the cervix.

How would you go about deciding which of the many options for management of GSM you will recommend for this patient? What do you weigh as you consider DHEA versus estrogen and topical versus oral therapy?
 
JoAnn V. Pinkerton, MD: Vulvovaginal atrophy (VVA), part of GSM, is associated with postmenopausal estrogen deficiency and includes the signs and symptoms seen on this patient's physical exam: vaginal narrowing, pallor, loss of elasticity, as well as pain with intercourse.6 Estrogen therapy is the most effective treatment for vaginal atrophy.13 Since she does not have significant menopausal symptoms, low-dose vaginal estrogen preparations are effective and generally safe treatments for VVA; these include creams, tablets containing estradiol or conjugated equine estrogen (CEE), and a low-dose vaginal estradiol ring--all available at doses that result in minimal systemic absorption.

Choice is usually made based on patient desire and likely adherence. If the patient prefers nonestrogen therapies that improve VVA and have been approved for relief of dyspareunia in postmenopausal women, I would discuss with the patient the oral selective estrogen receptor modulator ospemifene,14 and the new intravaginal DHEA suppositories, prasterone.15 Ospemifene is taken daily as an oral tablet, has a small risk of blood clots, and is my choice for women who do not need systemic hormone therapy and prefer to avoid vaginal therapy.

Andrew M. Kaunitz, MD: GSM is prevalent in menopausal women and, if not treated, causes progressive vaginal dryness and sexual discomfort. When the main indication for hormonal management in a menopausal woman is GSM (as opposed to treatment of vasomotor symptoms or prevention of osteoporosis), the treatment of choice is low-dose local vaginal estrogen, ospemifene, or prasterone (DHEA). Prasterone is a vaginally administered nonestrogen steroid that was approved by the FDA to treat dyspareunia associated with GSM. DHEA is an endogenous inactive steroid that is converted locally into androgens and estrogens; one vaginal insert is placed nightly.16,17

This 55-year-old widow has not been sexually active for some time. The facts that attempted penetration was painful and only an ultrathin (virginal) speculum could be used for examination indicate that contraction of the pelvic floor muscles is likely present. Simply starting medical management may not lead to comfortable/successful penetrative sex for this woman. In addition to  medical management, she would likely benefit from referral for physical therapy. Using dilators and other strategies, along with the positive impact that medical management will have on the vaginal mucosa, a woman's physical therapist can work with this patient to help the pelvic floor muscles relax and facilitate comfortable penetrative sex.

James A. Simon, MD: With only minor vasomotor symptoms, I would assess the other potential benefits of a systemic therapy. These might include cardiovascular risk reduction (systemic estrogens or estrogens/progesterone in some), breast cancer risk reduction (some data suggesting ospemifene can accomplish this), osteoporosis prevention (systemic estrogens and estrogen/androgens), etc. If there is an option for a treatment to address more than one symptom, in this case GSM, assessing the risks/benefits of each of these therapies should be estimated for this specific patient.

If there are no systemic benefits to be had, then any of the local treatments should be helpful. As there are no head-to-head comparisons available, local estrogen cream, tablets, rings, local DHEA, or systemic ospemifene each should be considered possible treatments. I also feel this patient may benefit from supplementary self-dilation and/or physical therapy.

 

Related article:
2017 Update on menopause


 
CASE Dyspareunia and vasomotor symptoms in a 42-year-old breast cancer survivor

A 42-year-old woman with a BRCA1 mutation has undergone prophylactic mastectomies as well as hysterectomy with bilateral salpingo-oophorectomy. She reports mild to moderate hot flashes and bothersome vaginal dryness and dyspareunia. Examination confirms GSM.

Would you advise systemic hormone therapy for this patient? What would your recommendation be for management of her GSM symptoms?

Dr. Simon: While one's gut reaction would be to avoid systemic estrogen therapy in a patient with a BRCA1 mutation, the scientific information confirming this fear is lacking.18 Such patients may benefit significantly from systemic estrogen therapy (reduced risk of cardiovascular disease and cognitive decline, etc.), and with both breasts and both ovaries removed, estrogen's breast cancer risks, if any in this population, are largely avoided. The patient also may benefit from additional local therapy with either estrogens or DHEA.

Dr. Kaunitz: Due to her high lifetime risk of breast and ovarian cancer, this woman has proceeded with risk-reducing breast and gynecologic surgery. As more BRCA mutation carriers are being identified and undergo risk-reducing bilateral mastectomy (usually with reconstruction) and salpingo-oophorectomy, clinicians and mutation carriers more frequently face decisions regarding use of systemic hormone therapy.

Mutation carriers who have undergone bilateral risk-reducing mastectomy experience a very low baseline future risk for breast cancer; accordingly, concerns regarding this disease should not prevent use of systemic hormone therapy. Furthermore, without hormone replacement, induced menopause in women this age is associated with an elevated risk of osteoporosis, persistent vasomotor symptoms, cardiovascular disease, stroke, mood changes, dementia, Parkinson disease, and overall mortality. Recognizing the safety of estrogen therapy in this setting, this 42-year-old BRCA1 mutation carrier can initiate estrogen therapy. Standard dose estrogen therapy refers to oral estradiol 1.0 mg, conjugated equine estrogen 0.625 mg,or transdermal estradiol 0.05 mg. In younger women like this 42-year-old with surgically induced menopause, higher than standard replacement doses of estrogen are often appropriate.17

Due to concerns the hormone therapy might further increase future risk of breast cancer, some mutation carriers may delay or avoid risk-reducing bilateral salpingo-oophorectomy, a potentially lifesaving surgery which reduces not only future risk of ovarian cancer but also future risk for breast cancer.

Among mutation carriers with intact breasts, several studies address risk of breast cancer with use of systemic hormone therapy. Although limited in numbers of participants and years of follow-up, in aggregate, these studies provide reassurance that short-term use of systemic hormone therapy does not increase breast cancer risk in women with BRCA1 or BRCA2 mutations and intact breasts.19

Dr. Pinkerton: For this woman with early surgical menopause and hysterectomy, estrogen therapy could improve her vasomotor symptoms and decrease her risk of bone loss and GSM.17 In the Women's Health Initiative trial, there were 7 fewer breast cancers per 10,000 women-years in the estrogen-onlyarm.20 Observational studies suggest that hormone therapy, when given to the average age of menopause, decreases the risks of heart disease, Parkinson disease, and dementia.21 Limited observational evidence suggests that hormone therapy use does not further increase risk of breast cancer in women following oophorectomy for BRCA1 or BRCA2 gene mutation.22

The absolute risks observed with hormone therapy tended to be small, especially in younger, healthy women. Systemic hormone therapy could treat her hot flashes and her GSM symptoms and potentially decrease health risks associated with premature estrogen deficiency. Nonestrogen therapies for hot flashes include low-dose antidepressants, gabapentin, and mind-body options, such as cognitive behavioral therapy and hypnosis, but these would not decrease her health risks or treat her GSM.

If she only requests treatment of her GSM symptoms, she would be a candidate for low-dose vaginal estrogen therapy, given as a cream, tablet, or ring depending on her choice. I would not choose ospemifene as my first choice as she is having hot flashes, and there are no data yet on the drug's health benefits in early menopause. If she prefers nonestrogen vaginal therapy, the new intravaginal DHEA might be a good choice as both estrogen and testosterone are increased locally in the vagina while hormone levels remain in the postmenopausal range. There is no boxed warning on the patient insert, although safety in women with breast cancer or in those with elevated risk of breast cancer has not been tested.

 

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. Goldstein I, Kim NN, Clayton AH, et al. Hypoactive Sexual Desire Disorder: International Society for the Study of Women’s Sexual Health (ISSWSH) Expert Consensus Panel Review. Mayo Clin Proc. 2017;92(1):114–128.
  2. Kingsberg SA. Attitudinal survey of women living with low sexual desire. J Womens Health (Larchmt). 2014;23(10):817–823.
  3. Foley K, Foley D, Johnson BH. Healthcare resource utilization and expenditures of women diagnosed with hypoactive sexual desire disorder. J Med Econ. 2010;13(4):583–590.
  4. Biddle AK, West SL, D’Aloisio AA, Wheeler SB, Borisov NN, Thorp J. Hypoactive sexual desire disorder in postmenopausal women: quality of life and health burden. Value Health. 2009;12(5):763–772.
  5. Portman DJ, Gass ML; Vulvovaginal Atrophy Terminology Consensus Conference Panel. Genitourinary syndrome of menopause: new terminology for vulvovaginal atrophy from the International Society for the Study of Women’s Sexual Health and the North American Menopause Society. Menopause. 2014;21(10):1063–1068.
  6. Management of symptomatic vulvovaginal atrophy: 2013 position statement of The North American Menopause Society. Menopause. 2013;20(9):888–902.
  7. Ettinger B, Hait H, Reape KZ, Shu H. Measuring symptom relief in studies of vaginal and vulvar atrophy: the most bothersome symptom approach. Menopause. 2008;15(5):885–889.
  8. Dennerstein L, Randolph J, Taffe J, Dudley E, Burger H. Hormones, mood, sexuality, and the menopausal transition. Fertil Steril. 2002;77(suppl 4):S42–S48.
  9. Brotto LA, Bitzer J, Laan E, Leiblum S, Luria M. Women’s sexual desire and arousal disorders [published correction appears in J Sex Med. 2010;7(2 pt 1):856]. J Sex Med. 2010;7(1 pt 2):586–614.
  10. US Food and Drug Administration website. FDA approves first treatment for sexual desire disorder. https://www.fda.gov/NewsEvents/Newsroom/PressAnnouncements/ucm458734.htm. Accessed August 14, 2017.
  11. Addyi (flibanserin) [package insert]. Bridgewater, NJ: Valeant Pharmaceuticals North America, LLC; 2016.
  12. Labrie F, Derogatis L, Archer DF, et al; Members of the VVA Prasterone Research Group. Effect of intravaginal prasterone on sexual dysfunction in postmenopausal women with vulvovaginal atrophy. J Sex Med. 2015;12(12):2401–2412.
  13. Lethaby A, Ayeleke RO, Roberts H. Local oestrogen for vaginal atrophy in postmenopausal women. Cochrane Database Syst Rev. 2016;8:CD001500.
  14. Portman DJ, Bachmann GA, Simon JA; Ospemifene Study Group. Ospemifene, a novel selective estrogen receptor modulator for treating dyspareunia associated with postmenopausal vulvar and vaginal atrophy. Menopause. 2013;20(6):623–630.
  15. Labrie F, Archer DF, Koltun, W, et al; VVA Prasterone Research Group. Efficacy of intravaginal dehydroepiandrosterone (DHEA) on moderate to severe dyspareunia and vaginal dryness, symptoms of vulvovaginal atrophy, and of the genitourinary syndrome of menopause. Menopause. 2016;23(3):243–256.
  16. Kaunitz AM. Focus on treating genital atrophy symptoms. OBG Manag. 2017;29(1):14, 16–17.
  17. The 2017 hormone therapy position statement of The North American Menopause Society. Menopause. 2017;24(7):728–753.
  18. Crandall CJ, Hovey KM, Andrews CA, et al. Breast cancer, endometrial cancer, and cardiovascular events in participants who used vaginal estrogen in the Women’s Health Initiative Observational Study. Menopause. August 14, 2017. doi:10.1097/GME.0000000000000956.
  19. Domchek S, Kaunitz AM. Use of systemic hormone therapy in BRCA mutation carriers. Menopause. 2016;23(9):1026–1027.
  20. Anderson GL, Limacher M, Assaf AR, et al; Women’s Health Initiative Steering Committee. Effects of conjugated equine estrogen in postmenopausal women with hysterectomy: the Women’s Health Initiative randomized controlled trial. JAMA. 2004;291(14):1701–1712.
  21. Faubion SS, Kuhle CL, Shuster LT, Rocca WA. Long-term health consequences of premature or early menopause and considerations for management. Climacteric. 2015;18(4):483–491.
  22. Gabriel CA, Tigges-Cardwell J, Stopfer J, Erlichman J, Nathanson K, Domchek SM. Use of total abdominal hysterectomy and hormone replacement therapy in BRCA1 and BRCA2 mutation carriers undergoing risk-reducing salpingo-oophorectomy. Fam Cancer. 2009;8(1):23-28.
Article PDF
Author and Disclosure Information

Dr. Levy is Vice President for Health Policy at the American College of Obstetricians and Gynecologists, Washington, DC.

The author reports no financial relationships relevant to this article.

EXPERT PANEL


Andrew M. Kaunitz, MD, NCMP, is University of Florida Term Professor and Associate Chairman, Department of Obstetrics and Gynecology, University of Florida College of Medicine-Jacksonville; Medical Director and Director of Menopause and Gynecologic Ultrasound Services, UF Women's Health Specialists at Emerson, Jacksonville, Florida.



JoAnn V. Pinkerton, MD, NCMP, is Professor, Department of Obstetrics and Gynecology, and Director, Midlife Health, University of Virginia Health System, Charlottesville, Virginia; Executive Director, The North American Menopause Society, Pepper Pike, Ohio.



James A. Simon, MD, CCD, IF, NCMP, is Clinical Professor, Department of Obstetrics and Gynecology, George Washington University; Medical Director, Women's Health & Research Consultants, Washington, DC.



Dr. Kaunitz reports that he receives grant or research support from Bayer, Endoceutics, and TherapeuticsMD and is a consultant to Bayer Healthcare, AMAG Pharmaceuticals, Allergan, Pfizer, and Shionogi. Dr. Kaunitz is a member of the OBG Management Board of Editors.

Dr. Pinkerton reports that she receives grant or research support from Grants/Research at TherapeuticsMD (fees go to the University of Virginia). She is a member of the OBG Management Board of Editors.

Dr. Simon reports he has served (within the last year) or is currently serving as a consultant to or on the advisory boards of: AbbVie, Allergan, AMAG Pharmaceuticals, Amgen, Ascend Therapeutics, Azure Biotech, Bayer Healthcare Pharmaceuticals, CEEK Enterprises, Covance, Millendo Therapeutics, Mitsubishi Tanabe Pharma Development America, ObsEva, Radius Health, Sanofi, Sebela Pharmaceuticals, Sermonix Pharmaceuticals, Shionogi, Symbiotec Pharmalab, TherapeuticsMD, and Valeant Pharmaceuticals. He has also served (within the last year) or is currently serving on the speaker's bureaus of: Duchesnay USA, Novo Nordisk, Shionogi, and Valeant Pharmaceuticals. In the last year, he has received or is currently receiving grant/research support from: AbbVie, Allergan, Agile Therapeutics, Bayer Healthcare, New England Research Institute, ObsEva, Palatin Technologies, Symbio Research, and TherapeuticsMD. He is a stockholder (direct purchase) in Sermonix Pharmaceuticals. Dr. Simon is a member of the OBG Management Board of Editors.

Issue
OBG Management - 29(9)
Publications
Topics
Page Number
34-36, 38-41
Sections
Author and Disclosure Information

Dr. Levy is Vice President for Health Policy at the American College of Obstetricians and Gynecologists, Washington, DC.

The author reports no financial relationships relevant to this article.

EXPERT PANEL


Andrew M. Kaunitz, MD, NCMP, is University of Florida Term Professor and Associate Chairman, Department of Obstetrics and Gynecology, University of Florida College of Medicine-Jacksonville; Medical Director and Director of Menopause and Gynecologic Ultrasound Services, UF Women's Health Specialists at Emerson, Jacksonville, Florida.



JoAnn V. Pinkerton, MD, NCMP, is Professor, Department of Obstetrics and Gynecology, and Director, Midlife Health, University of Virginia Health System, Charlottesville, Virginia; Executive Director, The North American Menopause Society, Pepper Pike, Ohio.



James A. Simon, MD, CCD, IF, NCMP, is Clinical Professor, Department of Obstetrics and Gynecology, George Washington University; Medical Director, Women's Health & Research Consultants, Washington, DC.



Dr. Kaunitz reports that he receives grant or research support from Bayer, Endoceutics, and TherapeuticsMD and is a consultant to Bayer Healthcare, AMAG Pharmaceuticals, Allergan, Pfizer, and Shionogi. Dr. Kaunitz is a member of the OBG Management Board of Editors.

Dr. Pinkerton reports that she receives grant or research support from Grants/Research at TherapeuticsMD (fees go to the University of Virginia). She is a member of the OBG Management Board of Editors.

Dr. Simon reports he has served (within the last year) or is currently serving as a consultant to or on the advisory boards of: AbbVie, Allergan, AMAG Pharmaceuticals, Amgen, Ascend Therapeutics, Azure Biotech, Bayer Healthcare Pharmaceuticals, CEEK Enterprises, Covance, Millendo Therapeutics, Mitsubishi Tanabe Pharma Development America, ObsEva, Radius Health, Sanofi, Sebela Pharmaceuticals, Sermonix Pharmaceuticals, Shionogi, Symbiotec Pharmalab, TherapeuticsMD, and Valeant Pharmaceuticals. He has also served (within the last year) or is currently serving on the speaker's bureaus of: Duchesnay USA, Novo Nordisk, Shionogi, and Valeant Pharmaceuticals. In the last year, he has received or is currently receiving grant/research support from: AbbVie, Allergan, Agile Therapeutics, Bayer Healthcare, New England Research Institute, ObsEva, Palatin Technologies, Symbio Research, and TherapeuticsMD. He is a stockholder (direct purchase) in Sermonix Pharmaceuticals. Dr. Simon is a member of the OBG Management Board of Editors.

Author and Disclosure Information

Dr. Levy is Vice President for Health Policy at the American College of Obstetricians and Gynecologists, Washington, DC.

The author reports no financial relationships relevant to this article.

EXPERT PANEL


Andrew M. Kaunitz, MD, NCMP, is University of Florida Term Professor and Associate Chairman, Department of Obstetrics and Gynecology, University of Florida College of Medicine-Jacksonville; Medical Director and Director of Menopause and Gynecologic Ultrasound Services, UF Women's Health Specialists at Emerson, Jacksonville, Florida.



JoAnn V. Pinkerton, MD, NCMP, is Professor, Department of Obstetrics and Gynecology, and Director, Midlife Health, University of Virginia Health System, Charlottesville, Virginia; Executive Director, The North American Menopause Society, Pepper Pike, Ohio.



James A. Simon, MD, CCD, IF, NCMP, is Clinical Professor, Department of Obstetrics and Gynecology, George Washington University; Medical Director, Women's Health & Research Consultants, Washington, DC.



Dr. Kaunitz reports that he receives grant or research support from Bayer, Endoceutics, and TherapeuticsMD and is a consultant to Bayer Healthcare, AMAG Pharmaceuticals, Allergan, Pfizer, and Shionogi. Dr. Kaunitz is a member of the OBG Management Board of Editors.

Dr. Pinkerton reports that she receives grant or research support from Grants/Research at TherapeuticsMD (fees go to the University of Virginia). She is a member of the OBG Management Board of Editors.

Dr. Simon reports he has served (within the last year) or is currently serving as a consultant to or on the advisory boards of: AbbVie, Allergan, AMAG Pharmaceuticals, Amgen, Ascend Therapeutics, Azure Biotech, Bayer Healthcare Pharmaceuticals, CEEK Enterprises, Covance, Millendo Therapeutics, Mitsubishi Tanabe Pharma Development America, ObsEva, Radius Health, Sanofi, Sebela Pharmaceuticals, Sermonix Pharmaceuticals, Shionogi, Symbiotec Pharmalab, TherapeuticsMD, and Valeant Pharmaceuticals. He has also served (within the last year) or is currently serving on the speaker's bureaus of: Duchesnay USA, Novo Nordisk, Shionogi, and Valeant Pharmaceuticals. In the last year, he has received or is currently receiving grant/research support from: AbbVie, Allergan, Agile Therapeutics, Bayer Healthcare, New England Research Institute, ObsEva, Palatin Technologies, Symbio Research, and TherapeuticsMD. He is a stockholder (direct purchase) in Sermonix Pharmaceuticals. Dr. Simon is a member of the OBG Management Board of Editors.

Article PDF
Article PDF

Illustration: Kimberly Martens for OBG Management
Neurologic functions, hormonal regulation, and psychological factors affect sexual desire and arousal to some extent. Menopause, and the genitourinary symptoms associated with it, also affect sexual function. Understanding the pathogenesis of sexual dysfunction is key to management decisions.

Sexual function is a complex, multifaceted process mediated by neurologic functions, hormonal regulation, and psychological factors. What could possibly go wrong?

As it turns out, quite a lot. Female sexual dysfunction is a common, vastly undertreated sexual health problem that can have wide-reaching effects on a woman’s life. These effects may include impaired body image, self-confidence, and self-worth. Sexual dysfunction also can contribute to relationship dissatisfaction and leave one feeling less connected with her partner.1,2 Studies have shown women with sexual dysfunction have higher health care expenditures3 and that depression and fatigue are common comorbidities, as is frequently seen in other chronic conditions such as diabetes and back pain.4

Understanding the pathogenesis of female sexual dysfunction helps to guide our approach to its management. Indeed, increased understanding of its pathology has helped to usher in new and emerging treatment options, as well as a personalized, biopsychosocial approach to its management.

 

Related article:
2016 Update on female sexual dysfunction

 

In this Update, I discuss the interplay of physiologic and psychological factors that affect female sexual function as well as the latest options for its management. I have also assembled a panel of experts to discuss 2 cases representative of sexual dysfunction that you may encounter in your clinical practice and how prescribing decisions are made for their management.

 

Read about factors that impact sexual function and agents to help manage dysfunction.

 

 

Multiple transmitters in the brain can increase or decrease sexual desire and function

Neurotransmitters involved in sexual excitation include brain dopamine, melanocortin, oxytocin, vasopressin, and norepinephrine, whereas brain opioids, serotonin, prolactin, and endocannabinoids function as sexual inhibitors. Inhibitory transmitters are activated normally during sexual refractoriness but also from primary aversion or secondary avoidance disorders.1 Drugs or conditions that reduce brain dopamine levels, increase the action of brain serotonin, or enhance brain opioid pathways have been shown to inhibit sexual desire, while those that increase hypothalamic and mesolimbic dopamine or decrease serotonin release have been shown to stimulate sexual desire.1 

Estradiol and progesterone can impact sexual function and desire

In addition to the neurotransmitters, hormones are important modulators of female sexual function. Decreasing levels of circulating estrogen after menopause lead to physiologic, biologic, and clinical changes in the urogenital tissues, such as decreased elastin, thinning of the epithelium, reduced vaginal blood flow, diminished lubrication, and decreased flexibility and elasticity. These changes result in the symptoms of genitourinary syndrome of menopause (GSM), which affects as many as half of all menopausal women.5,6 In clinical trials, dyspareunia and vaginal dryness are the most bothersome GSM symptoms reported.7

The role of hormonal regulation in sexual dysfunction among premenopausal women is not yet fully understood, but we do know that estradiol has been shown to improve sexual desire, progesterone tends to dampen sexual desire, and that testosterone at physiological levels has been shown in most studies to have a neutral effect on sexual desire in a well-estrogenized patient.8

 

Related article:
Focus on treating genital atrophy symptoms

 

Experience and behavior modulate or reinforce sexual dysfunction

The most common psychological factors that trigger or amplify female sexual dysfunction are depression, anxiety, distraction, negative body image, sexual abuse, and emotional neglect.9 Contextual or sociocultural factors, such as relationship discord, life-stage stressors (the empty nest syndrome or anxiety and sleep deprivation from a new baby), as well as cultural or religious values that suppress sexuality, also should be considered.9 Experience-based neuroplasticity (changes in brain pathways that become solidified by negative or positive experiences) may elucidate how a multimodal approach, utilizing medical and psychological treatment, can be beneficial for patients, particularly those with hypoactive sexual desire disorder (HSDD).1

New and emerging approaches to managing female sexual dysfunction

Three agents, one of which has been available for prescription for some time, one that is newly available, and one in the pipeline, are or may soon be in the gynecologist's armamentarium.

Flibanserin

Medications that target excitatory pathways or blunt inhibitory pathways are in development, and one, flibanserin (Addyi), has been US Food and Drug Administration (FDA)-approved for the treatment of acquired, generalized HSDD in premenopausal women.1,10 Flibanserin is a nonhormonal, centrally acting, postsynaptic serotonin 1A receptor agonist and a serotonin 2A receptor antagonist that is taken daily at bedtime (100 mg); several weeks are usually needed before any effects are noted.1 It is not approved for postmenopausal women and has a boxed warning about the risks of hypotension and syncope; its use is contraindicated in women who drink alcohol, in those who have hepatic impairment, and with the use of moderate or strong CYP3A4 inhibitors.11

Also keep in mind that flibanserin is only available through a Risk Evaluation and Mitigation Strategy program, so clinicians who wish to prescribe it must enroll in and complete training to become certified providers.9

 

Related article:
What you need to know (and do) to prescribe the new drug flibanserin

 

Prasterone

Prasterone (Intrarosa), a once-daily intravaginal dehydroepiandrosterone (DHEA) product, is a prohormone that increases local estrogen and testosterone and has the advantage of improved sexual function, desire, arousal, lubrication, orgasm, satisfaction, as well as pain at sexual activity.12 It was approved by the FDA in November 2016 to treat moderate to severe dyspareunia and has been available for prescribing since July 2017. Its cost is comparable to topical estrogen products, with a $25 copay program.

Because prasterone is not an estrogen, it does not have the boxed warning that all estrogen products are mandated by the FDA to have. This may make it more acceptable to patients, who often decline to use an estrogen product after seeing the boxed warning on the package. The Centers for Medicare and Medicaid Services (CMS) does not have prasterone on its list of potentially hazardous drugs for the elderly. However, keep in mind that because its label is for dyspareunia and not specifically for GSM, CMS considers it a drug of choice--in other words, like sildenafil (Viagra), a lifestyle choice and not for treatment of a medical condition. As such, at the present time, Medicare does not cover it.

Bremelanotide

Late-stage trials of bremelanotide, a melanocortin receptor agonist, are underway. Its mechanism of action is somewhat like that of flibanserin in that both drugs increase dopamine and norepinephrine levels. The advantage of bremelanotide is that it is used as needed. It is dosed subcutaneously (1.75 mg) and it can be used as often as a woman would like to use it. The FDA is expected to consider it for approval in about a year. Unpublished data from poster sessions at recent meetings show that, in a phase 3 study of 1,247 premenopausal women with HSDD (who had already been screened for depression and were found to have a physiologic condition), improvements in desire, arousal, lubrication, and orgasm were shown with bremelanotide. About 18% of women stopped using the drug because of adverse effects (nausea, vomiting, flushing, or headache) versus 2% for placebo. Like flibanserin, it is expected to be approved for premenopausal women only. 

 

Read how 3 experts would manage differing GSM symptoms.

 

 

What would you prescribe for these patients? 


CASE Genitourinary syndrome of menopause (GSM) in a 55-year-old woman

A 55-year-old widow is beginning a new relationship. She has not had partnered sexual activity for several years, but she recently has begun a relationship. She describes pain with attempted penetration with her new partner. Her last menstrual period was 3 years ago and she has experienced very minor menopausal symptoms, which are not bothersome. On examination, the vulva and vagina are pale, with thin epithelium and absent rugae. The tissue lacks elasticity. A virginal speculum is needed to visualize the cervix.

How would you go about deciding which of the many options for management of GSM you will recommend for this patient? What do you weigh as you consider DHEA versus estrogen and topical versus oral therapy?
 
JoAnn V. Pinkerton, MD: Vulvovaginal atrophy (VVA), part of GSM, is associated with postmenopausal estrogen deficiency and includes the signs and symptoms seen on this patient's physical exam: vaginal narrowing, pallor, loss of elasticity, as well as pain with intercourse.6 Estrogen therapy is the most effective treatment for vaginal atrophy.13 Since she does not have significant menopausal symptoms, low-dose vaginal estrogen preparations are effective and generally safe treatments for VVA; these include creams, tablets containing estradiol or conjugated equine estrogen (CEE), and a low-dose vaginal estradiol ring--all available at doses that result in minimal systemic absorption.

Choice is usually made based on patient desire and likely adherence. If the patient prefers nonestrogen therapies that improve VVA and have been approved for relief of dyspareunia in postmenopausal women, I would discuss with the patient the oral selective estrogen receptor modulator ospemifene,14 and the new intravaginal DHEA suppositories, prasterone.15 Ospemifene is taken daily as an oral tablet, has a small risk of blood clots, and is my choice for women who do not need systemic hormone therapy and prefer to avoid vaginal therapy.

Andrew M. Kaunitz, MD: GSM is prevalent in menopausal women and, if not treated, causes progressive vaginal dryness and sexual discomfort. When the main indication for hormonal management in a menopausal woman is GSM (as opposed to treatment of vasomotor symptoms or prevention of osteoporosis), the treatment of choice is low-dose local vaginal estrogen, ospemifene, or prasterone (DHEA). Prasterone is a vaginally administered nonestrogen steroid that was approved by the FDA to treat dyspareunia associated with GSM. DHEA is an endogenous inactive steroid that is converted locally into androgens and estrogens; one vaginal insert is placed nightly.16,17

This 55-year-old widow has not been sexually active for some time. The facts that attempted penetration was painful and only an ultrathin (virginal) speculum could be used for examination indicate that contraction of the pelvic floor muscles is likely present. Simply starting medical management may not lead to comfortable/successful penetrative sex for this woman. In addition to  medical management, she would likely benefit from referral for physical therapy. Using dilators and other strategies, along with the positive impact that medical management will have on the vaginal mucosa, a woman's physical therapist can work with this patient to help the pelvic floor muscles relax and facilitate comfortable penetrative sex.

James A. Simon, MD: With only minor vasomotor symptoms, I would assess the other potential benefits of a systemic therapy. These might include cardiovascular risk reduction (systemic estrogens or estrogens/progesterone in some), breast cancer risk reduction (some data suggesting ospemifene can accomplish this), osteoporosis prevention (systemic estrogens and estrogen/androgens), etc. If there is an option for a treatment to address more than one symptom, in this case GSM, assessing the risks/benefits of each of these therapies should be estimated for this specific patient.

If there are no systemic benefits to be had, then any of the local treatments should be helpful. As there are no head-to-head comparisons available, local estrogen cream, tablets, rings, local DHEA, or systemic ospemifene each should be considered possible treatments. I also feel this patient may benefit from supplementary self-dilation and/or physical therapy.

 

Related article:
2017 Update on menopause


 
CASE Dyspareunia and vasomotor symptoms in a 42-year-old breast cancer survivor

A 42-year-old woman with a BRCA1 mutation has undergone prophylactic mastectomies as well as hysterectomy with bilateral salpingo-oophorectomy. She reports mild to moderate hot flashes and bothersome vaginal dryness and dyspareunia. Examination confirms GSM.

Would you advise systemic hormone therapy for this patient? What would your recommendation be for management of her GSM symptoms?

Dr. Simon: While one's gut reaction would be to avoid systemic estrogen therapy in a patient with a BRCA1 mutation, the scientific information confirming this fear is lacking.18 Such patients may benefit significantly from systemic estrogen therapy (reduced risk of cardiovascular disease and cognitive decline, etc.), and with both breasts and both ovaries removed, estrogen's breast cancer risks, if any in this population, are largely avoided. The patient also may benefit from additional local therapy with either estrogens or DHEA.

Dr. Kaunitz: Due to her high lifetime risk of breast and ovarian cancer, this woman has proceeded with risk-reducing breast and gynecologic surgery. As more BRCA mutation carriers are being identified and undergo risk-reducing bilateral mastectomy (usually with reconstruction) and salpingo-oophorectomy, clinicians and mutation carriers more frequently face decisions regarding use of systemic hormone therapy.

Mutation carriers who have undergone bilateral risk-reducing mastectomy experience a very low baseline future risk for breast cancer; accordingly, concerns regarding this disease should not prevent use of systemic hormone therapy. Furthermore, without hormone replacement, induced menopause in women this age is associated with an elevated risk of osteoporosis, persistent vasomotor symptoms, cardiovascular disease, stroke, mood changes, dementia, Parkinson disease, and overall mortality. Recognizing the safety of estrogen therapy in this setting, this 42-year-old BRCA1 mutation carrier can initiate estrogen therapy. Standard dose estrogen therapy refers to oral estradiol 1.0 mg, conjugated equine estrogen 0.625 mg,or transdermal estradiol 0.05 mg. In younger women like this 42-year-old with surgically induced menopause, higher than standard replacement doses of estrogen are often appropriate.17

Due to concerns the hormone therapy might further increase future risk of breast cancer, some mutation carriers may delay or avoid risk-reducing bilateral salpingo-oophorectomy, a potentially lifesaving surgery which reduces not only future risk of ovarian cancer but also future risk for breast cancer.

Among mutation carriers with intact breasts, several studies address risk of breast cancer with use of systemic hormone therapy. Although limited in numbers of participants and years of follow-up, in aggregate, these studies provide reassurance that short-term use of systemic hormone therapy does not increase breast cancer risk in women with BRCA1 or BRCA2 mutations and intact breasts.19

Dr. Pinkerton: For this woman with early surgical menopause and hysterectomy, estrogen therapy could improve her vasomotor symptoms and decrease her risk of bone loss and GSM.17 In the Women's Health Initiative trial, there were 7 fewer breast cancers per 10,000 women-years in the estrogen-onlyarm.20 Observational studies suggest that hormone therapy, when given to the average age of menopause, decreases the risks of heart disease, Parkinson disease, and dementia.21 Limited observational evidence suggests that hormone therapy use does not further increase risk of breast cancer in women following oophorectomy for BRCA1 or BRCA2 gene mutation.22

The absolute risks observed with hormone therapy tended to be small, especially in younger, healthy women. Systemic hormone therapy could treat her hot flashes and her GSM symptoms and potentially decrease health risks associated with premature estrogen deficiency. Nonestrogen therapies for hot flashes include low-dose antidepressants, gabapentin, and mind-body options, such as cognitive behavioral therapy and hypnosis, but these would not decrease her health risks or treat her GSM.

If she only requests treatment of her GSM symptoms, she would be a candidate for low-dose vaginal estrogen therapy, given as a cream, tablet, or ring depending on her choice. I would not choose ospemifene as my first choice as she is having hot flashes, and there are no data yet on the drug's health benefits in early menopause. If she prefers nonestrogen vaginal therapy, the new intravaginal DHEA might be a good choice as both estrogen and testosterone are increased locally in the vagina while hormone levels remain in the postmenopausal range. There is no boxed warning on the patient insert, although safety in women with breast cancer or in those with elevated risk of breast cancer has not been tested.

 

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.

Illustration: Kimberly Martens for OBG Management
Neurologic functions, hormonal regulation, and psychological factors affect sexual desire and arousal to some extent. Menopause, and the genitourinary symptoms associated with it, also affect sexual function. Understanding the pathogenesis of sexual dysfunction is key to management decisions.

Sexual function is a complex, multifaceted process mediated by neurologic functions, hormonal regulation, and psychological factors. What could possibly go wrong?

As it turns out, quite a lot. Female sexual dysfunction is a common, vastly undertreated sexual health problem that can have wide-reaching effects on a woman’s life. These effects may include impaired body image, self-confidence, and self-worth. Sexual dysfunction also can contribute to relationship dissatisfaction and leave one feeling less connected with her partner.1,2 Studies have shown women with sexual dysfunction have higher health care expenditures3 and that depression and fatigue are common comorbidities, as is frequently seen in other chronic conditions such as diabetes and back pain.4

Understanding the pathogenesis of female sexual dysfunction helps to guide our approach to its management. Indeed, increased understanding of its pathology has helped to usher in new and emerging treatment options, as well as a personalized, biopsychosocial approach to its management.

 

Related article:
2016 Update on female sexual dysfunction

 

In this Update, I discuss the interplay of physiologic and psychological factors that affect female sexual function as well as the latest options for its management. I have also assembled a panel of experts to discuss 2 cases representative of sexual dysfunction that you may encounter in your clinical practice and how prescribing decisions are made for their management.

 

Read about factors that impact sexual function and agents to help manage dysfunction.

 

 

Multiple transmitters in the brain can increase or decrease sexual desire and function

Neurotransmitters involved in sexual excitation include brain dopamine, melanocortin, oxytocin, vasopressin, and norepinephrine, whereas brain opioids, serotonin, prolactin, and endocannabinoids function as sexual inhibitors. Inhibitory transmitters are activated normally during sexual refractoriness but also from primary aversion or secondary avoidance disorders.1 Drugs or conditions that reduce brain dopamine levels, increase the action of brain serotonin, or enhance brain opioid pathways have been shown to inhibit sexual desire, while those that increase hypothalamic and mesolimbic dopamine or decrease serotonin release have been shown to stimulate sexual desire.1 

Estradiol and progesterone can impact sexual function and desire

In addition to the neurotransmitters, hormones are important modulators of female sexual function. Decreasing levels of circulating estrogen after menopause lead to physiologic, biologic, and clinical changes in the urogenital tissues, such as decreased elastin, thinning of the epithelium, reduced vaginal blood flow, diminished lubrication, and decreased flexibility and elasticity. These changes result in the symptoms of genitourinary syndrome of menopause (GSM), which affects as many as half of all menopausal women.5,6 In clinical trials, dyspareunia and vaginal dryness are the most bothersome GSM symptoms reported.7

The role of hormonal regulation in sexual dysfunction among premenopausal women is not yet fully understood, but we do know that estradiol has been shown to improve sexual desire, progesterone tends to dampen sexual desire, and that testosterone at physiological levels has been shown in most studies to have a neutral effect on sexual desire in a well-estrogenized patient.8

 

Related article:
Focus on treating genital atrophy symptoms

 

Experience and behavior modulate or reinforce sexual dysfunction

The most common psychological factors that trigger or amplify female sexual dysfunction are depression, anxiety, distraction, negative body image, sexual abuse, and emotional neglect.9 Contextual or sociocultural factors, such as relationship discord, life-stage stressors (the empty nest syndrome or anxiety and sleep deprivation from a new baby), as well as cultural or religious values that suppress sexuality, also should be considered.9 Experience-based neuroplasticity (changes in brain pathways that become solidified by negative or positive experiences) may elucidate how a multimodal approach, utilizing medical and psychological treatment, can be beneficial for patients, particularly those with hypoactive sexual desire disorder (HSDD).1

New and emerging approaches to managing female sexual dysfunction

Three agents, one of which has been available for prescription for some time, one that is newly available, and one in the pipeline, are or may soon be in the gynecologist's armamentarium.

Flibanserin

Medications that target excitatory pathways or blunt inhibitory pathways are in development, and one, flibanserin (Addyi), has been US Food and Drug Administration (FDA)-approved for the treatment of acquired, generalized HSDD in premenopausal women.1,10 Flibanserin is a nonhormonal, centrally acting, postsynaptic serotonin 1A receptor agonist and a serotonin 2A receptor antagonist that is taken daily at bedtime (100 mg); several weeks are usually needed before any effects are noted.1 It is not approved for postmenopausal women and has a boxed warning about the risks of hypotension and syncope; its use is contraindicated in women who drink alcohol, in those who have hepatic impairment, and with the use of moderate or strong CYP3A4 inhibitors.11

Also keep in mind that flibanserin is only available through a Risk Evaluation and Mitigation Strategy program, so clinicians who wish to prescribe it must enroll in and complete training to become certified providers.9

 

Related article:
What you need to know (and do) to prescribe the new drug flibanserin

 

Prasterone

Prasterone (Intrarosa), a once-daily intravaginal dehydroepiandrosterone (DHEA) product, is a prohormone that increases local estrogen and testosterone and has the advantage of improved sexual function, desire, arousal, lubrication, orgasm, satisfaction, as well as pain at sexual activity.12 It was approved by the FDA in November 2016 to treat moderate to severe dyspareunia and has been available for prescribing since July 2017. Its cost is comparable to topical estrogen products, with a $25 copay program.

Because prasterone is not an estrogen, it does not have the boxed warning that all estrogen products are mandated by the FDA to have. This may make it more acceptable to patients, who often decline to use an estrogen product after seeing the boxed warning on the package. The Centers for Medicare and Medicaid Services (CMS) does not have prasterone on its list of potentially hazardous drugs for the elderly. However, keep in mind that because its label is for dyspareunia and not specifically for GSM, CMS considers it a drug of choice--in other words, like sildenafil (Viagra), a lifestyle choice and not for treatment of a medical condition. As such, at the present time, Medicare does not cover it.

Bremelanotide

Late-stage trials of bremelanotide, a melanocortin receptor agonist, are underway. Its mechanism of action is somewhat like that of flibanserin in that both drugs increase dopamine and norepinephrine levels. The advantage of bremelanotide is that it is used as needed. It is dosed subcutaneously (1.75 mg) and it can be used as often as a woman would like to use it. The FDA is expected to consider it for approval in about a year. Unpublished data from poster sessions at recent meetings show that, in a phase 3 study of 1,247 premenopausal women with HSDD (who had already been screened for depression and were found to have a physiologic condition), improvements in desire, arousal, lubrication, and orgasm were shown with bremelanotide. About 18% of women stopped using the drug because of adverse effects (nausea, vomiting, flushing, or headache) versus 2% for placebo. Like flibanserin, it is expected to be approved for premenopausal women only. 

 

Read how 3 experts would manage differing GSM symptoms.

 

 

What would you prescribe for these patients? 


CASE Genitourinary syndrome of menopause (GSM) in a 55-year-old woman

A 55-year-old widow is beginning a new relationship. She has not had partnered sexual activity for several years, but she recently has begun a relationship. She describes pain with attempted penetration with her new partner. Her last menstrual period was 3 years ago and she has experienced very minor menopausal symptoms, which are not bothersome. On examination, the vulva and vagina are pale, with thin epithelium and absent rugae. The tissue lacks elasticity. A virginal speculum is needed to visualize the cervix.

How would you go about deciding which of the many options for management of GSM you will recommend for this patient? What do you weigh as you consider DHEA versus estrogen and topical versus oral therapy?
 
JoAnn V. Pinkerton, MD: Vulvovaginal atrophy (VVA), part of GSM, is associated with postmenopausal estrogen deficiency and includes the signs and symptoms seen on this patient's physical exam: vaginal narrowing, pallor, loss of elasticity, as well as pain with intercourse.6 Estrogen therapy is the most effective treatment for vaginal atrophy.13 Since she does not have significant menopausal symptoms, low-dose vaginal estrogen preparations are effective and generally safe treatments for VVA; these include creams, tablets containing estradiol or conjugated equine estrogen (CEE), and a low-dose vaginal estradiol ring--all available at doses that result in minimal systemic absorption.

Choice is usually made based on patient desire and likely adherence. If the patient prefers nonestrogen therapies that improve VVA and have been approved for relief of dyspareunia in postmenopausal women, I would discuss with the patient the oral selective estrogen receptor modulator ospemifene,14 and the new intravaginal DHEA suppositories, prasterone.15 Ospemifene is taken daily as an oral tablet, has a small risk of blood clots, and is my choice for women who do not need systemic hormone therapy and prefer to avoid vaginal therapy.

Andrew M. Kaunitz, MD: GSM is prevalent in menopausal women and, if not treated, causes progressive vaginal dryness and sexual discomfort. When the main indication for hormonal management in a menopausal woman is GSM (as opposed to treatment of vasomotor symptoms or prevention of osteoporosis), the treatment of choice is low-dose local vaginal estrogen, ospemifene, or prasterone (DHEA). Prasterone is a vaginally administered nonestrogen steroid that was approved by the FDA to treat dyspareunia associated with GSM. DHEA is an endogenous inactive steroid that is converted locally into androgens and estrogens; one vaginal insert is placed nightly.16,17

This 55-year-old widow has not been sexually active for some time. The facts that attempted penetration was painful and only an ultrathin (virginal) speculum could be used for examination indicate that contraction of the pelvic floor muscles is likely present. Simply starting medical management may not lead to comfortable/successful penetrative sex for this woman. In addition to  medical management, she would likely benefit from referral for physical therapy. Using dilators and other strategies, along with the positive impact that medical management will have on the vaginal mucosa, a woman's physical therapist can work with this patient to help the pelvic floor muscles relax and facilitate comfortable penetrative sex.

James A. Simon, MD: With only minor vasomotor symptoms, I would assess the other potential benefits of a systemic therapy. These might include cardiovascular risk reduction (systemic estrogens or estrogens/progesterone in some), breast cancer risk reduction (some data suggesting ospemifene can accomplish this), osteoporosis prevention (systemic estrogens and estrogen/androgens), etc. If there is an option for a treatment to address more than one symptom, in this case GSM, assessing the risks/benefits of each of these therapies should be estimated for this specific patient.

If there are no systemic benefits to be had, then any of the local treatments should be helpful. As there are no head-to-head comparisons available, local estrogen cream, tablets, rings, local DHEA, or systemic ospemifene each should be considered possible treatments. I also feel this patient may benefit from supplementary self-dilation and/or physical therapy.

 

Related article:
2017 Update on menopause


 
CASE Dyspareunia and vasomotor symptoms in a 42-year-old breast cancer survivor

A 42-year-old woman with a BRCA1 mutation has undergone prophylactic mastectomies as well as hysterectomy with bilateral salpingo-oophorectomy. She reports mild to moderate hot flashes and bothersome vaginal dryness and dyspareunia. Examination confirms GSM.

Would you advise systemic hormone therapy for this patient? What would your recommendation be for management of her GSM symptoms?

Dr. Simon: While one's gut reaction would be to avoid systemic estrogen therapy in a patient with a BRCA1 mutation, the scientific information confirming this fear is lacking.18 Such patients may benefit significantly from systemic estrogen therapy (reduced risk of cardiovascular disease and cognitive decline, etc.), and with both breasts and both ovaries removed, estrogen's breast cancer risks, if any in this population, are largely avoided. The patient also may benefit from additional local therapy with either estrogens or DHEA.

Dr. Kaunitz: Due to her high lifetime risk of breast and ovarian cancer, this woman has proceeded with risk-reducing breast and gynecologic surgery. As more BRCA mutation carriers are being identified and undergo risk-reducing bilateral mastectomy (usually with reconstruction) and salpingo-oophorectomy, clinicians and mutation carriers more frequently face decisions regarding use of systemic hormone therapy.

Mutation carriers who have undergone bilateral risk-reducing mastectomy experience a very low baseline future risk for breast cancer; accordingly, concerns regarding this disease should not prevent use of systemic hormone therapy. Furthermore, without hormone replacement, induced menopause in women this age is associated with an elevated risk of osteoporosis, persistent vasomotor symptoms, cardiovascular disease, stroke, mood changes, dementia, Parkinson disease, and overall mortality. Recognizing the safety of estrogen therapy in this setting, this 42-year-old BRCA1 mutation carrier can initiate estrogen therapy. Standard dose estrogen therapy refers to oral estradiol 1.0 mg, conjugated equine estrogen 0.625 mg,or transdermal estradiol 0.05 mg. In younger women like this 42-year-old with surgically induced menopause, higher than standard replacement doses of estrogen are often appropriate.17

Due to concerns the hormone therapy might further increase future risk of breast cancer, some mutation carriers may delay or avoid risk-reducing bilateral salpingo-oophorectomy, a potentially lifesaving surgery which reduces not only future risk of ovarian cancer but also future risk for breast cancer.

Among mutation carriers with intact breasts, several studies address risk of breast cancer with use of systemic hormone therapy. Although limited in numbers of participants and years of follow-up, in aggregate, these studies provide reassurance that short-term use of systemic hormone therapy does not increase breast cancer risk in women with BRCA1 or BRCA2 mutations and intact breasts.19

Dr. Pinkerton: For this woman with early surgical menopause and hysterectomy, estrogen therapy could improve her vasomotor symptoms and decrease her risk of bone loss and GSM.17 In the Women's Health Initiative trial, there were 7 fewer breast cancers per 10,000 women-years in the estrogen-onlyarm.20 Observational studies suggest that hormone therapy, when given to the average age of menopause, decreases the risks of heart disease, Parkinson disease, and dementia.21 Limited observational evidence suggests that hormone therapy use does not further increase risk of breast cancer in women following oophorectomy for BRCA1 or BRCA2 gene mutation.22

The absolute risks observed with hormone therapy tended to be small, especially in younger, healthy women. Systemic hormone therapy could treat her hot flashes and her GSM symptoms and potentially decrease health risks associated with premature estrogen deficiency. Nonestrogen therapies for hot flashes include low-dose antidepressants, gabapentin, and mind-body options, such as cognitive behavioral therapy and hypnosis, but these would not decrease her health risks or treat her GSM.

If she only requests treatment of her GSM symptoms, she would be a candidate for low-dose vaginal estrogen therapy, given as a cream, tablet, or ring depending on her choice. I would not choose ospemifene as my first choice as she is having hot flashes, and there are no data yet on the drug's health benefits in early menopause. If she prefers nonestrogen vaginal therapy, the new intravaginal DHEA might be a good choice as both estrogen and testosterone are increased locally in the vagina while hormone levels remain in the postmenopausal range. There is no boxed warning on the patient insert, although safety in women with breast cancer or in those with elevated risk of breast cancer has not been tested.

 

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. Goldstein I, Kim NN, Clayton AH, et al. Hypoactive Sexual Desire Disorder: International Society for the Study of Women’s Sexual Health (ISSWSH) Expert Consensus Panel Review. Mayo Clin Proc. 2017;92(1):114–128.
  2. Kingsberg SA. Attitudinal survey of women living with low sexual desire. J Womens Health (Larchmt). 2014;23(10):817–823.
  3. Foley K, Foley D, Johnson BH. Healthcare resource utilization and expenditures of women diagnosed with hypoactive sexual desire disorder. J Med Econ. 2010;13(4):583–590.
  4. Biddle AK, West SL, D’Aloisio AA, Wheeler SB, Borisov NN, Thorp J. Hypoactive sexual desire disorder in postmenopausal women: quality of life and health burden. Value Health. 2009;12(5):763–772.
  5. Portman DJ, Gass ML; Vulvovaginal Atrophy Terminology Consensus Conference Panel. Genitourinary syndrome of menopause: new terminology for vulvovaginal atrophy from the International Society for the Study of Women’s Sexual Health and the North American Menopause Society. Menopause. 2014;21(10):1063–1068.
  6. Management of symptomatic vulvovaginal atrophy: 2013 position statement of The North American Menopause Society. Menopause. 2013;20(9):888–902.
  7. Ettinger B, Hait H, Reape KZ, Shu H. Measuring symptom relief in studies of vaginal and vulvar atrophy: the most bothersome symptom approach. Menopause. 2008;15(5):885–889.
  8. Dennerstein L, Randolph J, Taffe J, Dudley E, Burger H. Hormones, mood, sexuality, and the menopausal transition. Fertil Steril. 2002;77(suppl 4):S42–S48.
  9. Brotto LA, Bitzer J, Laan E, Leiblum S, Luria M. Women’s sexual desire and arousal disorders [published correction appears in J Sex Med. 2010;7(2 pt 1):856]. J Sex Med. 2010;7(1 pt 2):586–614.
  10. US Food and Drug Administration website. FDA approves first treatment for sexual desire disorder. https://www.fda.gov/NewsEvents/Newsroom/PressAnnouncements/ucm458734.htm. Accessed August 14, 2017.
  11. Addyi (flibanserin) [package insert]. Bridgewater, NJ: Valeant Pharmaceuticals North America, LLC; 2016.
  12. Labrie F, Derogatis L, Archer DF, et al; Members of the VVA Prasterone Research Group. Effect of intravaginal prasterone on sexual dysfunction in postmenopausal women with vulvovaginal atrophy. J Sex Med. 2015;12(12):2401–2412.
  13. Lethaby A, Ayeleke RO, Roberts H. Local oestrogen for vaginal atrophy in postmenopausal women. Cochrane Database Syst Rev. 2016;8:CD001500.
  14. Portman DJ, Bachmann GA, Simon JA; Ospemifene Study Group. Ospemifene, a novel selective estrogen receptor modulator for treating dyspareunia associated with postmenopausal vulvar and vaginal atrophy. Menopause. 2013;20(6):623–630.
  15. Labrie F, Archer DF, Koltun, W, et al; VVA Prasterone Research Group. Efficacy of intravaginal dehydroepiandrosterone (DHEA) on moderate to severe dyspareunia and vaginal dryness, symptoms of vulvovaginal atrophy, and of the genitourinary syndrome of menopause. Menopause. 2016;23(3):243–256.
  16. Kaunitz AM. Focus on treating genital atrophy symptoms. OBG Manag. 2017;29(1):14, 16–17.
  17. The 2017 hormone therapy position statement of The North American Menopause Society. Menopause. 2017;24(7):728–753.
  18. Crandall CJ, Hovey KM, Andrews CA, et al. Breast cancer, endometrial cancer, and cardiovascular events in participants who used vaginal estrogen in the Women’s Health Initiative Observational Study. Menopause. August 14, 2017. doi:10.1097/GME.0000000000000956.
  19. Domchek S, Kaunitz AM. Use of systemic hormone therapy in BRCA mutation carriers. Menopause. 2016;23(9):1026–1027.
  20. Anderson GL, Limacher M, Assaf AR, et al; Women’s Health Initiative Steering Committee. Effects of conjugated equine estrogen in postmenopausal women with hysterectomy: the Women’s Health Initiative randomized controlled trial. JAMA. 2004;291(14):1701–1712.
  21. Faubion SS, Kuhle CL, Shuster LT, Rocca WA. Long-term health consequences of premature or early menopause and considerations for management. Climacteric. 2015;18(4):483–491.
  22. Gabriel CA, Tigges-Cardwell J, Stopfer J, Erlichman J, Nathanson K, Domchek SM. Use of total abdominal hysterectomy and hormone replacement therapy in BRCA1 and BRCA2 mutation carriers undergoing risk-reducing salpingo-oophorectomy. Fam Cancer. 2009;8(1):23-28.
References
  1. Goldstein I, Kim NN, Clayton AH, et al. Hypoactive Sexual Desire Disorder: International Society for the Study of Women’s Sexual Health (ISSWSH) Expert Consensus Panel Review. Mayo Clin Proc. 2017;92(1):114–128.
  2. Kingsberg SA. Attitudinal survey of women living with low sexual desire. J Womens Health (Larchmt). 2014;23(10):817–823.
  3. Foley K, Foley D, Johnson BH. Healthcare resource utilization and expenditures of women diagnosed with hypoactive sexual desire disorder. J Med Econ. 2010;13(4):583–590.
  4. Biddle AK, West SL, D’Aloisio AA, Wheeler SB, Borisov NN, Thorp J. Hypoactive sexual desire disorder in postmenopausal women: quality of life and health burden. Value Health. 2009;12(5):763–772.
  5. Portman DJ, Gass ML; Vulvovaginal Atrophy Terminology Consensus Conference Panel. Genitourinary syndrome of menopause: new terminology for vulvovaginal atrophy from the International Society for the Study of Women’s Sexual Health and the North American Menopause Society. Menopause. 2014;21(10):1063–1068.
  6. Management of symptomatic vulvovaginal atrophy: 2013 position statement of The North American Menopause Society. Menopause. 2013;20(9):888–902.
  7. Ettinger B, Hait H, Reape KZ, Shu H. Measuring symptom relief in studies of vaginal and vulvar atrophy: the most bothersome symptom approach. Menopause. 2008;15(5):885–889.
  8. Dennerstein L, Randolph J, Taffe J, Dudley E, Burger H. Hormones, mood, sexuality, and the menopausal transition. Fertil Steril. 2002;77(suppl 4):S42–S48.
  9. Brotto LA, Bitzer J, Laan E, Leiblum S, Luria M. Women’s sexual desire and arousal disorders [published correction appears in J Sex Med. 2010;7(2 pt 1):856]. J Sex Med. 2010;7(1 pt 2):586–614.
  10. US Food and Drug Administration website. FDA approves first treatment for sexual desire disorder. https://www.fda.gov/NewsEvents/Newsroom/PressAnnouncements/ucm458734.htm. Accessed August 14, 2017.
  11. Addyi (flibanserin) [package insert]. Bridgewater, NJ: Valeant Pharmaceuticals North America, LLC; 2016.
  12. Labrie F, Derogatis L, Archer DF, et al; Members of the VVA Prasterone Research Group. Effect of intravaginal prasterone on sexual dysfunction in postmenopausal women with vulvovaginal atrophy. J Sex Med. 2015;12(12):2401–2412.
  13. Lethaby A, Ayeleke RO, Roberts H. Local oestrogen for vaginal atrophy in postmenopausal women. Cochrane Database Syst Rev. 2016;8:CD001500.
  14. Portman DJ, Bachmann GA, Simon JA; Ospemifene Study Group. Ospemifene, a novel selective estrogen receptor modulator for treating dyspareunia associated with postmenopausal vulvar and vaginal atrophy. Menopause. 2013;20(6):623–630.
  15. Labrie F, Archer DF, Koltun, W, et al; VVA Prasterone Research Group. Efficacy of intravaginal dehydroepiandrosterone (DHEA) on moderate to severe dyspareunia and vaginal dryness, symptoms of vulvovaginal atrophy, and of the genitourinary syndrome of menopause. Menopause. 2016;23(3):243–256.
  16. Kaunitz AM. Focus on treating genital atrophy symptoms. OBG Manag. 2017;29(1):14, 16–17.
  17. The 2017 hormone therapy position statement of The North American Menopause Society. Menopause. 2017;24(7):728–753.
  18. Crandall CJ, Hovey KM, Andrews CA, et al. Breast cancer, endometrial cancer, and cardiovascular events in participants who used vaginal estrogen in the Women’s Health Initiative Observational Study. Menopause. August 14, 2017. doi:10.1097/GME.0000000000000956.
  19. Domchek S, Kaunitz AM. Use of systemic hormone therapy in BRCA mutation carriers. Menopause. 2016;23(9):1026–1027.
  20. Anderson GL, Limacher M, Assaf AR, et al; Women’s Health Initiative Steering Committee. Effects of conjugated equine estrogen in postmenopausal women with hysterectomy: the Women’s Health Initiative randomized controlled trial. JAMA. 2004;291(14):1701–1712.
  21. Faubion SS, Kuhle CL, Shuster LT, Rocca WA. Long-term health consequences of premature or early menopause and considerations for management. Climacteric. 2015;18(4):483–491.
  22. Gabriel CA, Tigges-Cardwell J, Stopfer J, Erlichman J, Nathanson K, Domchek SM. Use of total abdominal hysterectomy and hormone replacement therapy in BRCA1 and BRCA2 mutation carriers undergoing risk-reducing salpingo-oophorectomy. Fam Cancer. 2009;8(1):23-28.
Issue
OBG Management - 29(9)
Issue
OBG Management - 29(9)
Page Number
34-36, 38-41
Page Number
34-36, 38-41
Publications
Publications
Topics
Article Type
Display Headline
2017 Update on female sexual dysfunction
Display Headline
2017 Update on female sexual dysfunction
Sections
Inside the Article

Fast Tracks

  • Although not fully understood how, estradiol can improve sexual desire, progesterone tends to dampen sexual desire, and testosterone has a neutral effect in premenopausal women
  • Newly available since July 2017, prasterone is a once-daily intravaginal agent that treats moderate to severe dyspareunia and has costs similar to topical estrogens
  • Estrogen therapy may be considered in a breast cancer mutation carrier who has undergone prophylactic mastectomies and  bilateral salpingo-oophorectomy
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Article PDF Media

5 Points on Stiff Elbow

Article Type
Changed
Thu, 09/19/2019 - 13:20

Take-Home Points

  • Proper patient selection is critical as extensive postoperative rehabilitation is required to obtain an excellent outcome.
  • Open and arthroscopic approaches are effective treatment options for elbow contractures.
  • Elbow stability must be restored to obtain a successful outcome.
  • Knowledge of neurovascular anatomy is essential to prevent neurologic complications.
  • Prophylactic ulnar nerve release should be considered, especially in patients with limited flexion.

Elbow stiffness has several etiologies, posttraumatic being the most common. Elbow stiffness can have debilitating functional effects necessitating treatment. In a biomechanical study of normal elbow function, Morrey and colleagues1 determined that a flexion extension arc of 100° (30°-130°) and a forearm rotation arc of 100° (50° pronation-50° supination) are required in 90% of activities of daily living. Similarly, elbow flexion of <105° was poorly tolerated, whereas patients could easier adapt to flexion contractures up to 40°.2

The goal of initial evaluation should be to establish the cause of the contracture and the patient’s functional demands and ability to cooperate in the extensive postoperative rehabilitation that is essential in achieving an excellent functional outcome. In a thorough clinical examination, the clinician must note skin, range of motion (ROM), ligamentous stability, and neurovascular structures and give special attention to ulnar nerve function and symptoms. Mid-arc pain suggests additional intra-articular pathology, as stiffness typically causes pain only at the limits of motion as osteophytes impinge and soft tissue is under maximal tension. Routine elbow radiographs are required in all cases, and computed tomography (CT) can be useful in evaluating osseous sources of contracture. Suspected ligamentous instability and cartilaginous defects particularly in the setting of mid-arc pain are best evaluated with magnetic resonance imaging.3

In this 5-point review, we evaluate treatment options as well as rehabilitation protocols in the management of elbow stiffness.

1 Anatomy of Contracture: The Usual Suspects

The cause of elbow stiffness is incompletely understood. Several posited contributing factors include biology, complex intra-articular anatomy, capsular distention favoring a flexed position, and tenuous postoperative fixation necessitating prolonged immobilization. Identifying intrinsic and extrinsic anatomical sources of stiffness can help guide treatment.4 Intrinsic pathology includes intra-articular malunion, osteophytes, loose bodies, and adhesions; extrinsic pathology includes soft-tissue contracture, heterotopic ossification, and extra-articular malunion.

Compared with the normal elbow, the capsule becomes thickened and fibrotic and thereby prevents motion. Severe contractures, and extension contractures in particular, may require release of the posterior medial capsule and the posterior medial collateral ligament (MCL) to regain motion. In a series of 42 patients with flexion <100°, Park and colleagues5 noted that all patients required release of the posterior band of the MCL to regain flexion. Other muscular impediments to motion include contracture of the brachialis and scarring of the triceps to the posterior humerus. Scarring of the triceps to the humerus can limit flexion.

In the posttrauma setting, intra-articular and extra-articular malunion must be considered. Extension malunion of the distal humerus can reduce flexion,6 and shortening with compromise of the olecranon and coronoid fossae can limit both flexion and extension.

Last, heterotopic ossification and osteophytes should be assessed as potential causes of limited ROM. Both the coronoid process and the olecranon can develop osteophytes, and their respective fossae should be assessed with CT. Posterior impingement is rare at the tip of the olecranon; it occurs because of "widening" of the olecranon by "Mickey Mouse ear" osteophytes and bony encroachment along the medial and lateral columns. Thus, the olecranon must be narrowed and the fossa widened and deepened.

In case of concomitant ligament instability, we prefer to reconstruct the ligament first, and then perform contracture release as a staged procedure. We favor a staged approach because the rehabilitation regimens for instability and contracture release are diametrically opposed: Instability requires immobilization, and contracture release requires immediate motion. Last, incision placement and ulnar nerve management are crucial in minimizing the potential complications of the second procedure.

2 Nonoperative Treatment

In the absence of significant bony impediments to motion—such as heterotopic ossification or malunion—initial treatment should commence with nonoperative therapy. Therapy should be initiated as soon as concern for stiffness arises in order to prevent contracture. Initial nonoperative treatment can also serve as an important litmus test of postoperative adherence. Adequate patient relaxation is crucial in avoiding co-contracture resisting stretching forces. Passive ROM exercises use sustained force to allow time-dependent stress relaxation to increase tissue length as well as fatigue antagonist muscles. In addition, hold-and-relax techniques apply isometric resistance to induce relaxation of antagonist muscles.7 Active ROM should emphasize triceps isolation and elbow extension to prevent scarring of the triceps to the posterior humerus.

Corrective splinting can be an effective adjuvant to physiotherapy. Static progressive turnbuckle splints was described as an effective treatment for both elbow flexion and extension contractures, effecting an average 43° increase in elbow motion in a series of 15 patients.8 Similarly, Gelinas and colleagues9 noted improvement among 22 patients treated with turnbuckle splinting for an average of 4.5 months. In addition, serial extension splints may be used in the treatment of elbow flexion contractures.

3 Open Contacture Release and Surgical Approach

When nonoperative therapies fail to restore the functional arc of motion, patients with flexion contractures or extension contractures of >30° may be indicated for contracture release. Surgical approach should be determined by meticulous preoperative planning that notes prior incisions and CT findings. It can be helpful to organize common offending structures and their effects on flexion and extension (Table).

Table.
Careful attention to bony architecture and joint space can provide valuable information about motion impingement and arthrosis. In most cases, both the anterior compartment and the posterior compartment must be addressed, but this can often be achieved with a medial or lateral approach alone.
Figure 1.
Figure 1 shows a lateral radiograph of an active weightlifter with –10° of extension and 90° of flexion. CT showed osteophytes primarily over the anterior and posterior compartments of the ulnohumeral joint (Figure 2). Ulnar nerve paresthesia dictated our approach from the medial side.
Figure 2.

A medial over-the-top approach uses the medial supracondylar ridge as a landmark, subperiosteally reflecting the brachialis anteriorly.10 The ulnar nerve is neurolyzed and protected posteriorly. The flexor-pronator mass is split distally and elevated along with the brachialis as a single sleeve of muscle. The coronal plane of dissection should be the anterior half of the lateral epicondyle to avoid injury to the MCL. Large Bennett or Hohmann retractors can hinge on the lateral border of the humerus and provide clear visualization of the anterior capsule and the ulnohumeral joint. Exposure of the radiocapitellar joint is possible, but this joint is very deep in the operative field, and caution should be taken excising the anterolateral capsule because of the risk of radial nerve injury. The ulnar nerve can be temporarily transposed anteriorly to dissect posteriorly along the supracondylar ridge of the humerus. The triceps is reflected off the distal humerus. Occasionally, the posterior band of the MCL must be resected in severe extension contractures. If possible, the anterior bundle should be preserved. With this approach, the anterior capsule, distal humerus, coronoid process, posterior MCL, posterior capsule, and triceps can be addressed. The zone anterior to the radial head and the anterolateral and posterolateral capsule cannot be safely exposed with a medial approach. As described by Wada and colleagues,11 a primarily medial approach resulted in an average 64° increase in arc of motion.

Figure 3.
The lateral approach, commonly referred to as the column approach, uses the lateral supracondylar ridge to release distal fibers of the brachioradialis and the extensor carpi radialis longus (ECRL) (Figure 3). This exposure is limited by the radial nerve proximally and the posterior interosseous the ECRL and the extensor carpi radialis brevis. After the ECRL and the distal brachioradialis are released from the humerus, the superolateral capsule is visualized. After the brachialis and the radial nerve are elevated off the capsule, the capsule can be safely excised extending medially. Limited forearm rotation can also be addressed, as the proximal radioulnar joint (PRUJ) and the radiocapitellar joint can be exposed through this approach. Given the limits of lateral exposure, in some cases we release the lateral ulnar collateral ligament to better "hinge" open the elbow and obtain better visualization of the PRUJ and the medial joint. In isolation, the ligament can be repaired with suture anchors without causing instability or delaying rehabilitation. Husband and Hastings12 described a lateral approach with a mean 46° increased arc of motion—similar to the lateral column procedure with a mean 45° increased arc of motion, described by Mansat and Morrey.13 Last, a single posterior approach with wide medial and lateral flaps can also be used, but this has the distinct disadvantage of a risk of seroma or hematoma owing to the large dead space created. We typically prefer to initially approach contractures medially, as this allows ulnar nerve symptoms to be addressed. A lateral approach can be added to address forearm rotation and facilitate protection of the radial nerve/PIN during anterolateral capsular release in significant flexion contractures. Presence of heterotopic ossification or extra-articular malunion must also be considered, as it may dictate the surgical approach. If circumferential release of the elbow soft tissue and complete stripping of the distal humerus are performed for contracture release, simple collateral ligament repairs will be inadequate in providing stability immediately after surgery. In these extreme circumstances, we prefer to protect the ligament repair with
an internal joint stabilizer (Skeletal Dynamics) (Figure 4) and to initiate motion therapy immediately. External fixation (hinged or unhinged is rarely used in our practice.
Figure 4.

Arthroscopic Contracture Release and Technique

Recently, arthroscopic elbow contracture release, a technically demanding but effective treatment option, has gained popularity. Knowledge of neurovascular anatomy is a prerequisite to the prevention of devastating neurologic complications (ulnar, median, and radial nerve transections have been described14,15). Relative contraindications include extensive heterotopic ossification, ulnar nerve transposition, and limited arthroscopic experience. Functional improvements as well as average 26° to 42° increases in arc of motion have been described with arthroscopic release.16-18 In thin-framed patients with dense elbow capsular scarring (severe loss of elbow motion with hard block) and small joint space, arthroscopic release and particularly arthroscope insertion are notoriously difficult.

The patient may be placed in the prone, lateral decubitus, or supine position, depending on surgeon preference (Figure 5). Before surgery, portals and the ulnar nerve should be carefully outlined.19

Figure 5.
The median, ulnar, and medial antebrachial cutaneous nerves are at risk during placement of the proximal anteromedial portal, and the radial and lateral antebrachial cutaneous nerves are at risk during proximal anterolateral portal placement. Ulnar nerve decompression may also be performed through a limited incision before arthroscopic contracture release, and the nerve is protected during use of the shaver along the posteromedial joint capsule. Fluid management is required to ensure proper visualization and avoid excessive swelling.

We prefer to start by entering the posterior compartment and using the shaver to create a working space. All bone work and resectioning should be performed before capsular resection. After the joint and the olecranon fossa are identified, soft-tissue and bony débridement of the olecranon and the fossa can be performed. Care should be taken to protect the ulnar nerve when the posteromedial corner or medial gutter is approached.

Figure 6.
The anterior compartment is then visualized through the proximal anteromedial portal, and a working anterolateral portal is established (Figures 6A-6D). Often, bone forms in the coronoid fossa and the supracapitellar area, and this bone is resected. After completion of the bone work, the capsule is released or resected. In osteoarthritis cases, the capsule is usually relatively normal, and simple release proximally or distally is sufficient. In posttraumatic stiffness, however, marked capsular thickening and arthrofibrosis may occur, and in these cases more thorough capsular resection is advised. The capsulectomy is begun anteromedially, where the brachialis protects the median nerve and the brachial vessels. As the lateral border of the brachialis is approached, extreme caution must be taken to prevent radial or PIN injury. After the brachioradialis is visualized, more aggressive resection can be performed. Last, given the high risk of superficial infection classically associated with elbow arthroscopy, portal sites must be tightly closed and drains retained until scant drainage is noted.15

5 Additional Considerations

After surgery, the elbow is immobilized in maximal extension and supination with an anterior splint, and therapy is initiated either immediately or after temporary immobilization.16,19,20 Regional anesthesia is crucial in obtaining adequate pain control and establishing an immediate postoperative therapy program. The utility of continuous passive motion (CPM) in postoperative protocols is controversial. A retrospective case-control study of 32 patients matched on age, diagnosis, and contraction severity found no benefit of CPM use, and increased costs and hospital length of stay, leading the authors to recommend against CPM use.20

Neurovascular risks are associated with both open and arthroscopic elbow contracture release. Particularly concerning is the risk of traction ulnar neuropathy, described in upward of 20% of patients.21 Anatomical studies have found decreases in cubital tunnel and ulnar nerve area as elbow flexion increases with corresponding increased intraneural pressure,22 leading some authors to recommend prophylactic ulnar nerve release with limited preoperative flexion.15 Nevertheless, despite transposition, ulnar nerve symptoms were noted in 8 of 40 patients who underwent open contracture release for posttraumatic loss of elbow flexion.5 In a retrospective review of 164 open and arthroscopic elbow contracture releases, Williams and colleagues21 noted an 8.1% rate of postoperative new-onset ulnar nerve symptoms. The rate of ulnar neuropathy was nonsignificantly elevated among patients with preoperative flexion of <100° (15.2% vs 3.7%; P = .057). Recently, a retrospective review of 564 consecutive arthroscopic contracture releases found a significantly higher rate of delayed-onset ulnar neuritis among patients without prophylactic ulnar nerve decompression or transposition (11% vs 3%; P < .001).23 Further analysis revealed that, compared with decompression, ulnar nerve transposition did not offer additional benefit but was associated with a significantly higher rate of wound complications (19% vs 4%; P = .03). We favor prophylactic release, particularly in the setting of preoperative extension contracture. For open contracture release from the lateral approach, however, we do not routinely release the ulnar nerve unless there were preoperative symptoms.

Although open and arthroscopic contracture releases can provide durable outcomes in the setting of painless elbow stiffness, options are more limited in the treatment of the painful stiff elbow. Total elbow arthroplasty remains an option in low-demand elderly patients but is not without significant risk of complications.24 In addition, durability concerns and postoperative restrictions make total elbow arthroplasty less attractive to younger patients. Interposition arthroplasty may be indicated as a salvage procedure in the treatment of a young or high-demand patient with a stiff painful elbow.25 Elbow stability is crucial in obtaining a successful outcome, and data on optimal graft choices are limited.

Conclusion

Elbow stiffness, a common complication of trauma, significantly impairs activities of daily living. Early after trauma, therapy should be initiated to prevent contracture. In the absence of symptomatic arthritis, both open and arthroscopic contracture releases are effective surgical treatments in properly selected and motivated patients. Although more research is needed to establish the optimal surgical approach, severity and anatomical cause of contracture should guide decisions as to which approach to use. Having a thorough understanding of neurovascular anatomy and of prophylactic ulnar nerve decompression in the setting of limited preoperative flexion can mitigate complications.

References

1. Morrey BF, Askew LJ, Chao EY. A biomechanical study of normal functional elbow motion. J Bone Joint Surg Am. 1981;63(6):872-877.

2. Hotchkiss RN. Elbow contracture. In: Green DP, Rotchkiss RN, Pederson WC, Wolfe SW, eds. Green’s Operative Hand Surgery. 5th ed. New York, NY: Churchill-Livingstone; 2005:667-682.

3. Van Zeeland NL, Yamaguchi K. Arthroscopic capsular release of the elbow. J Shoulder Elbow Surg. 2010;19(2):13-19.

4. Morrey BF. Post-traumatic contracture of the elbow. Operative treatment, including distraction arthroplasty. J Bone Joint Surg Am. 1990;72(4):601-618.

5. Park MJ, Chang MJ, Lee YB, Kang HJ. Surgical release for posttraumatic loss of elbow flexion. J Bone Joint Surg Am. 2010;92(16):2692-2699.

6. Brouwer KM, Lindenhovius AL, Ring D. Loss of anterior translation of the distal humeral articular surface is associated with decreased elbow flexion. J Hand Surg Am. 2009;34(7):
1256-1260.

7. Taylor DC, Dalton JD, Seaber AV, Garrett WE. Viscoelastic properties of muscle-tendon units: the biomechanical effects of stretching. Am J Sports Med. 1990;18(3):300-309.

8. Green DP, McCoy H. Turnbuckle orthotic correction of elbow-flexion contractures after acute injuries. J Bone Joint Surg Am. 1979;61(7):1092-1095.

9. Gelinas JJ, Faber KJ, Patterson SD, King GJ. The effectiveness of turnbuckle splinting for elbow contractures. J Bone Joint Surg Br. 2000;82(1):74-78.

10. Hotchkiss RN, Kasparyan GN. The medial "over the top" approach to the elbow. Tech Orthop. 2000;15(2):105-112.

11. Wada T, Ishii S, Usui M, Miyano S. The medial approach for operative release of post-traumatic contracture of the elbow. J Bone Joint Surg Br. 2000;82(1):68-73.

12. Husband JB, Hastings H. The lateral approach for operative release of post-traumatic contracture of the elbow. J Bone Joint Surg Am. 1990;72(9):1353-1358.

13. Mansat P, Morrey BF. The column procedure: a limited lateral approach for extrinsic contracture of the elbow. J Bone Joint Surg Am. 1998;80(11):1603-1605.

14. Haapaniemi T, Berggren M, Adolfsson L. Complete transection of the median and radial nerves during arthroscopic release of post-traumatic elbow contracture. Arthroscopy. 1999;15(7):784-787.

15. Kelly EW, Morrey BF, O’Driscoll SW. Complications of elbow arthroscopy. J Bone Joint Surg Am. 2001;83(1):25-34.

16. Ball CM, Meunier M, Galatz LM, Calfee R, Yamaguchi K. Arthroscopic treatment of post-traumatic elbow contracture. J Shoulder Elbow Surg. 2002;11(6):624-629.

17. Ćefo I, Eygendaal D. Arthroscopic arthrolysis for posttraumatic elbow stiffness. J Shoulder Elbow Surg. 2011;20(3):434-439.

18. Nguyen D, Proper SI, MacDermid JC, King GJ, Faber KJ. Functional outcomes of arthroscopic capsular release of the elbow. Arthroscopy. 2006;22(8):842-849.

19. Sahajpal D, Choi T, Wright TW. Arthroscopic release of the stiff elbow. J Hand Surg. 2009;34(3):540-544.

20. Lindenhovius AL, Jupiter JB. The posttraumatic stiff elbow: a review of the literature. J Hand Surg. 2007;32(10):1605-1623.

21. Williams BG, Sotereanos DG, Baratz ME, Jarrett CD, Venouziou AI, Miller MC. The contracted elbow: is ulnar nerve release necessary? J Shoulder Elbow Surg. 2012;21(12):
1632-1636.

22. Gelberman RH, Yamaguchi K, Hollstien SB, et al. Changes in interstitial pressure and cross-sectional area of the cubital tunnel and of the ulnar nerve with flexion of the elbow. an experimental study in human cadavera. J Bone Joint Surg Am. 1998;80(4):492-501.

23. Blonna D, O’Driscoll SW. Delayed-onset ulnar neuritis after release of elbow contracture: preventive strategies derived from a study of 563 cases. Arthroscopy. 2014;30(8):947-956.

24. Mansat P, Morrey BF. Semiconstrained total elbow arthroplasty for ankylosed and stiff elbows. J Bone Joint Surg. 2000;82(9):1260-1268.

25. Hausman MR, Birnbaum PS. Interposition elbow arthroplasty. Tech Hand Up Extrem Surg. 2004;8(3):181-188.

Article PDF
Author and Disclosure Information

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

Issue
The American Journal of Orthopedics - 46(5)
Publications
Topics
Page Number
245-251
Sections
Author and Disclosure Information

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

Author and Disclosure Information

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

Article PDF
Article PDF

Take-Home Points

  • Proper patient selection is critical as extensive postoperative rehabilitation is required to obtain an excellent outcome.
  • Open and arthroscopic approaches are effective treatment options for elbow contractures.
  • Elbow stability must be restored to obtain a successful outcome.
  • Knowledge of neurovascular anatomy is essential to prevent neurologic complications.
  • Prophylactic ulnar nerve release should be considered, especially in patients with limited flexion.

Elbow stiffness has several etiologies, posttraumatic being the most common. Elbow stiffness can have debilitating functional effects necessitating treatment. In a biomechanical study of normal elbow function, Morrey and colleagues1 determined that a flexion extension arc of 100° (30°-130°) and a forearm rotation arc of 100° (50° pronation-50° supination) are required in 90% of activities of daily living. Similarly, elbow flexion of <105° was poorly tolerated, whereas patients could easier adapt to flexion contractures up to 40°.2

The goal of initial evaluation should be to establish the cause of the contracture and the patient’s functional demands and ability to cooperate in the extensive postoperative rehabilitation that is essential in achieving an excellent functional outcome. In a thorough clinical examination, the clinician must note skin, range of motion (ROM), ligamentous stability, and neurovascular structures and give special attention to ulnar nerve function and symptoms. Mid-arc pain suggests additional intra-articular pathology, as stiffness typically causes pain only at the limits of motion as osteophytes impinge and soft tissue is under maximal tension. Routine elbow radiographs are required in all cases, and computed tomography (CT) can be useful in evaluating osseous sources of contracture. Suspected ligamentous instability and cartilaginous defects particularly in the setting of mid-arc pain are best evaluated with magnetic resonance imaging.3

In this 5-point review, we evaluate treatment options as well as rehabilitation protocols in the management of elbow stiffness.

1 Anatomy of Contracture: The Usual Suspects

The cause of elbow stiffness is incompletely understood. Several posited contributing factors include biology, complex intra-articular anatomy, capsular distention favoring a flexed position, and tenuous postoperative fixation necessitating prolonged immobilization. Identifying intrinsic and extrinsic anatomical sources of stiffness can help guide treatment.4 Intrinsic pathology includes intra-articular malunion, osteophytes, loose bodies, and adhesions; extrinsic pathology includes soft-tissue contracture, heterotopic ossification, and extra-articular malunion.

Compared with the normal elbow, the capsule becomes thickened and fibrotic and thereby prevents motion. Severe contractures, and extension contractures in particular, may require release of the posterior medial capsule and the posterior medial collateral ligament (MCL) to regain motion. In a series of 42 patients with flexion <100°, Park and colleagues5 noted that all patients required release of the posterior band of the MCL to regain flexion. Other muscular impediments to motion include contracture of the brachialis and scarring of the triceps to the posterior humerus. Scarring of the triceps to the humerus can limit flexion.

In the posttrauma setting, intra-articular and extra-articular malunion must be considered. Extension malunion of the distal humerus can reduce flexion,6 and shortening with compromise of the olecranon and coronoid fossae can limit both flexion and extension.

Last, heterotopic ossification and osteophytes should be assessed as potential causes of limited ROM. Both the coronoid process and the olecranon can develop osteophytes, and their respective fossae should be assessed with CT. Posterior impingement is rare at the tip of the olecranon; it occurs because of "widening" of the olecranon by "Mickey Mouse ear" osteophytes and bony encroachment along the medial and lateral columns. Thus, the olecranon must be narrowed and the fossa widened and deepened.

In case of concomitant ligament instability, we prefer to reconstruct the ligament first, and then perform contracture release as a staged procedure. We favor a staged approach because the rehabilitation regimens for instability and contracture release are diametrically opposed: Instability requires immobilization, and contracture release requires immediate motion. Last, incision placement and ulnar nerve management are crucial in minimizing the potential complications of the second procedure.

2 Nonoperative Treatment

In the absence of significant bony impediments to motion—such as heterotopic ossification or malunion—initial treatment should commence with nonoperative therapy. Therapy should be initiated as soon as concern for stiffness arises in order to prevent contracture. Initial nonoperative treatment can also serve as an important litmus test of postoperative adherence. Adequate patient relaxation is crucial in avoiding co-contracture resisting stretching forces. Passive ROM exercises use sustained force to allow time-dependent stress relaxation to increase tissue length as well as fatigue antagonist muscles. In addition, hold-and-relax techniques apply isometric resistance to induce relaxation of antagonist muscles.7 Active ROM should emphasize triceps isolation and elbow extension to prevent scarring of the triceps to the posterior humerus.

Corrective splinting can be an effective adjuvant to physiotherapy. Static progressive turnbuckle splints was described as an effective treatment for both elbow flexion and extension contractures, effecting an average 43° increase in elbow motion in a series of 15 patients.8 Similarly, Gelinas and colleagues9 noted improvement among 22 patients treated with turnbuckle splinting for an average of 4.5 months. In addition, serial extension splints may be used in the treatment of elbow flexion contractures.

3 Open Contacture Release and Surgical Approach

When nonoperative therapies fail to restore the functional arc of motion, patients with flexion contractures or extension contractures of >30° may be indicated for contracture release. Surgical approach should be determined by meticulous preoperative planning that notes prior incisions and CT findings. It can be helpful to organize common offending structures and their effects on flexion and extension (Table).

Table.
Careful attention to bony architecture and joint space can provide valuable information about motion impingement and arthrosis. In most cases, both the anterior compartment and the posterior compartment must be addressed, but this can often be achieved with a medial or lateral approach alone.
Figure 1.
Figure 1 shows a lateral radiograph of an active weightlifter with –10° of extension and 90° of flexion. CT showed osteophytes primarily over the anterior and posterior compartments of the ulnohumeral joint (Figure 2). Ulnar nerve paresthesia dictated our approach from the medial side.
Figure 2.

A medial over-the-top approach uses the medial supracondylar ridge as a landmark, subperiosteally reflecting the brachialis anteriorly.10 The ulnar nerve is neurolyzed and protected posteriorly. The flexor-pronator mass is split distally and elevated along with the brachialis as a single sleeve of muscle. The coronal plane of dissection should be the anterior half of the lateral epicondyle to avoid injury to the MCL. Large Bennett or Hohmann retractors can hinge on the lateral border of the humerus and provide clear visualization of the anterior capsule and the ulnohumeral joint. Exposure of the radiocapitellar joint is possible, but this joint is very deep in the operative field, and caution should be taken excising the anterolateral capsule because of the risk of radial nerve injury. The ulnar nerve can be temporarily transposed anteriorly to dissect posteriorly along the supracondylar ridge of the humerus. The triceps is reflected off the distal humerus. Occasionally, the posterior band of the MCL must be resected in severe extension contractures. If possible, the anterior bundle should be preserved. With this approach, the anterior capsule, distal humerus, coronoid process, posterior MCL, posterior capsule, and triceps can be addressed. The zone anterior to the radial head and the anterolateral and posterolateral capsule cannot be safely exposed with a medial approach. As described by Wada and colleagues,11 a primarily medial approach resulted in an average 64° increase in arc of motion.

Figure 3.
The lateral approach, commonly referred to as the column approach, uses the lateral supracondylar ridge to release distal fibers of the brachioradialis and the extensor carpi radialis longus (ECRL) (Figure 3). This exposure is limited by the radial nerve proximally and the posterior interosseous the ECRL and the extensor carpi radialis brevis. After the ECRL and the distal brachioradialis are released from the humerus, the superolateral capsule is visualized. After the brachialis and the radial nerve are elevated off the capsule, the capsule can be safely excised extending medially. Limited forearm rotation can also be addressed, as the proximal radioulnar joint (PRUJ) and the radiocapitellar joint can be exposed through this approach. Given the limits of lateral exposure, in some cases we release the lateral ulnar collateral ligament to better "hinge" open the elbow and obtain better visualization of the PRUJ and the medial joint. In isolation, the ligament can be repaired with suture anchors without causing instability or delaying rehabilitation. Husband and Hastings12 described a lateral approach with a mean 46° increased arc of motion—similar to the lateral column procedure with a mean 45° increased arc of motion, described by Mansat and Morrey.13 Last, a single posterior approach with wide medial and lateral flaps can also be used, but this has the distinct disadvantage of a risk of seroma or hematoma owing to the large dead space created. We typically prefer to initially approach contractures medially, as this allows ulnar nerve symptoms to be addressed. A lateral approach can be added to address forearm rotation and facilitate protection of the radial nerve/PIN during anterolateral capsular release in significant flexion contractures. Presence of heterotopic ossification or extra-articular malunion must also be considered, as it may dictate the surgical approach. If circumferential release of the elbow soft tissue and complete stripping of the distal humerus are performed for contracture release, simple collateral ligament repairs will be inadequate in providing stability immediately after surgery. In these extreme circumstances, we prefer to protect the ligament repair with
an internal joint stabilizer (Skeletal Dynamics) (Figure 4) and to initiate motion therapy immediately. External fixation (hinged or unhinged is rarely used in our practice.
Figure 4.

Arthroscopic Contracture Release and Technique

Recently, arthroscopic elbow contracture release, a technically demanding but effective treatment option, has gained popularity. Knowledge of neurovascular anatomy is a prerequisite to the prevention of devastating neurologic complications (ulnar, median, and radial nerve transections have been described14,15). Relative contraindications include extensive heterotopic ossification, ulnar nerve transposition, and limited arthroscopic experience. Functional improvements as well as average 26° to 42° increases in arc of motion have been described with arthroscopic release.16-18 In thin-framed patients with dense elbow capsular scarring (severe loss of elbow motion with hard block) and small joint space, arthroscopic release and particularly arthroscope insertion are notoriously difficult.

The patient may be placed in the prone, lateral decubitus, or supine position, depending on surgeon preference (Figure 5). Before surgery, portals and the ulnar nerve should be carefully outlined.19

Figure 5.
The median, ulnar, and medial antebrachial cutaneous nerves are at risk during placement of the proximal anteromedial portal, and the radial and lateral antebrachial cutaneous nerves are at risk during proximal anterolateral portal placement. Ulnar nerve decompression may also be performed through a limited incision before arthroscopic contracture release, and the nerve is protected during use of the shaver along the posteromedial joint capsule. Fluid management is required to ensure proper visualization and avoid excessive swelling.

We prefer to start by entering the posterior compartment and using the shaver to create a working space. All bone work and resectioning should be performed before capsular resection. After the joint and the olecranon fossa are identified, soft-tissue and bony débridement of the olecranon and the fossa can be performed. Care should be taken to protect the ulnar nerve when the posteromedial corner or medial gutter is approached.

Figure 6.
The anterior compartment is then visualized through the proximal anteromedial portal, and a working anterolateral portal is established (Figures 6A-6D). Often, bone forms in the coronoid fossa and the supracapitellar area, and this bone is resected. After completion of the bone work, the capsule is released or resected. In osteoarthritis cases, the capsule is usually relatively normal, and simple release proximally or distally is sufficient. In posttraumatic stiffness, however, marked capsular thickening and arthrofibrosis may occur, and in these cases more thorough capsular resection is advised. The capsulectomy is begun anteromedially, where the brachialis protects the median nerve and the brachial vessels. As the lateral border of the brachialis is approached, extreme caution must be taken to prevent radial or PIN injury. After the brachioradialis is visualized, more aggressive resection can be performed. Last, given the high risk of superficial infection classically associated with elbow arthroscopy, portal sites must be tightly closed and drains retained until scant drainage is noted.15

5 Additional Considerations

After surgery, the elbow is immobilized in maximal extension and supination with an anterior splint, and therapy is initiated either immediately or after temporary immobilization.16,19,20 Regional anesthesia is crucial in obtaining adequate pain control and establishing an immediate postoperative therapy program. The utility of continuous passive motion (CPM) in postoperative protocols is controversial. A retrospective case-control study of 32 patients matched on age, diagnosis, and contraction severity found no benefit of CPM use, and increased costs and hospital length of stay, leading the authors to recommend against CPM use.20

Neurovascular risks are associated with both open and arthroscopic elbow contracture release. Particularly concerning is the risk of traction ulnar neuropathy, described in upward of 20% of patients.21 Anatomical studies have found decreases in cubital tunnel and ulnar nerve area as elbow flexion increases with corresponding increased intraneural pressure,22 leading some authors to recommend prophylactic ulnar nerve release with limited preoperative flexion.15 Nevertheless, despite transposition, ulnar nerve symptoms were noted in 8 of 40 patients who underwent open contracture release for posttraumatic loss of elbow flexion.5 In a retrospective review of 164 open and arthroscopic elbow contracture releases, Williams and colleagues21 noted an 8.1% rate of postoperative new-onset ulnar nerve symptoms. The rate of ulnar neuropathy was nonsignificantly elevated among patients with preoperative flexion of <100° (15.2% vs 3.7%; P = .057). Recently, a retrospective review of 564 consecutive arthroscopic contracture releases found a significantly higher rate of delayed-onset ulnar neuritis among patients without prophylactic ulnar nerve decompression or transposition (11% vs 3%; P < .001).23 Further analysis revealed that, compared with decompression, ulnar nerve transposition did not offer additional benefit but was associated with a significantly higher rate of wound complications (19% vs 4%; P = .03). We favor prophylactic release, particularly in the setting of preoperative extension contracture. For open contracture release from the lateral approach, however, we do not routinely release the ulnar nerve unless there were preoperative symptoms.

Although open and arthroscopic contracture releases can provide durable outcomes in the setting of painless elbow stiffness, options are more limited in the treatment of the painful stiff elbow. Total elbow arthroplasty remains an option in low-demand elderly patients but is not without significant risk of complications.24 In addition, durability concerns and postoperative restrictions make total elbow arthroplasty less attractive to younger patients. Interposition arthroplasty may be indicated as a salvage procedure in the treatment of a young or high-demand patient with a stiff painful elbow.25 Elbow stability is crucial in obtaining a successful outcome, and data on optimal graft choices are limited.

Conclusion

Elbow stiffness, a common complication of trauma, significantly impairs activities of daily living. Early after trauma, therapy should be initiated to prevent contracture. In the absence of symptomatic arthritis, both open and arthroscopic contracture releases are effective surgical treatments in properly selected and motivated patients. Although more research is needed to establish the optimal surgical approach, severity and anatomical cause of contracture should guide decisions as to which approach to use. Having a thorough understanding of neurovascular anatomy and of prophylactic ulnar nerve decompression in the setting of limited preoperative flexion can mitigate complications.

Take-Home Points

  • Proper patient selection is critical as extensive postoperative rehabilitation is required to obtain an excellent outcome.
  • Open and arthroscopic approaches are effective treatment options for elbow contractures.
  • Elbow stability must be restored to obtain a successful outcome.
  • Knowledge of neurovascular anatomy is essential to prevent neurologic complications.
  • Prophylactic ulnar nerve release should be considered, especially in patients with limited flexion.

Elbow stiffness has several etiologies, posttraumatic being the most common. Elbow stiffness can have debilitating functional effects necessitating treatment. In a biomechanical study of normal elbow function, Morrey and colleagues1 determined that a flexion extension arc of 100° (30°-130°) and a forearm rotation arc of 100° (50° pronation-50° supination) are required in 90% of activities of daily living. Similarly, elbow flexion of <105° was poorly tolerated, whereas patients could easier adapt to flexion contractures up to 40°.2

The goal of initial evaluation should be to establish the cause of the contracture and the patient’s functional demands and ability to cooperate in the extensive postoperative rehabilitation that is essential in achieving an excellent functional outcome. In a thorough clinical examination, the clinician must note skin, range of motion (ROM), ligamentous stability, and neurovascular structures and give special attention to ulnar nerve function and symptoms. Mid-arc pain suggests additional intra-articular pathology, as stiffness typically causes pain only at the limits of motion as osteophytes impinge and soft tissue is under maximal tension. Routine elbow radiographs are required in all cases, and computed tomography (CT) can be useful in evaluating osseous sources of contracture. Suspected ligamentous instability and cartilaginous defects particularly in the setting of mid-arc pain are best evaluated with magnetic resonance imaging.3

In this 5-point review, we evaluate treatment options as well as rehabilitation protocols in the management of elbow stiffness.

1 Anatomy of Contracture: The Usual Suspects

The cause of elbow stiffness is incompletely understood. Several posited contributing factors include biology, complex intra-articular anatomy, capsular distention favoring a flexed position, and tenuous postoperative fixation necessitating prolonged immobilization. Identifying intrinsic and extrinsic anatomical sources of stiffness can help guide treatment.4 Intrinsic pathology includes intra-articular malunion, osteophytes, loose bodies, and adhesions; extrinsic pathology includes soft-tissue contracture, heterotopic ossification, and extra-articular malunion.

Compared with the normal elbow, the capsule becomes thickened and fibrotic and thereby prevents motion. Severe contractures, and extension contractures in particular, may require release of the posterior medial capsule and the posterior medial collateral ligament (MCL) to regain motion. In a series of 42 patients with flexion <100°, Park and colleagues5 noted that all patients required release of the posterior band of the MCL to regain flexion. Other muscular impediments to motion include contracture of the brachialis and scarring of the triceps to the posterior humerus. Scarring of the triceps to the humerus can limit flexion.

In the posttrauma setting, intra-articular and extra-articular malunion must be considered. Extension malunion of the distal humerus can reduce flexion,6 and shortening with compromise of the olecranon and coronoid fossae can limit both flexion and extension.

Last, heterotopic ossification and osteophytes should be assessed as potential causes of limited ROM. Both the coronoid process and the olecranon can develop osteophytes, and their respective fossae should be assessed with CT. Posterior impingement is rare at the tip of the olecranon; it occurs because of "widening" of the olecranon by "Mickey Mouse ear" osteophytes and bony encroachment along the medial and lateral columns. Thus, the olecranon must be narrowed and the fossa widened and deepened.

In case of concomitant ligament instability, we prefer to reconstruct the ligament first, and then perform contracture release as a staged procedure. We favor a staged approach because the rehabilitation regimens for instability and contracture release are diametrically opposed: Instability requires immobilization, and contracture release requires immediate motion. Last, incision placement and ulnar nerve management are crucial in minimizing the potential complications of the second procedure.

2 Nonoperative Treatment

In the absence of significant bony impediments to motion—such as heterotopic ossification or malunion—initial treatment should commence with nonoperative therapy. Therapy should be initiated as soon as concern for stiffness arises in order to prevent contracture. Initial nonoperative treatment can also serve as an important litmus test of postoperative adherence. Adequate patient relaxation is crucial in avoiding co-contracture resisting stretching forces. Passive ROM exercises use sustained force to allow time-dependent stress relaxation to increase tissue length as well as fatigue antagonist muscles. In addition, hold-and-relax techniques apply isometric resistance to induce relaxation of antagonist muscles.7 Active ROM should emphasize triceps isolation and elbow extension to prevent scarring of the triceps to the posterior humerus.

Corrective splinting can be an effective adjuvant to physiotherapy. Static progressive turnbuckle splints was described as an effective treatment for both elbow flexion and extension contractures, effecting an average 43° increase in elbow motion in a series of 15 patients.8 Similarly, Gelinas and colleagues9 noted improvement among 22 patients treated with turnbuckle splinting for an average of 4.5 months. In addition, serial extension splints may be used in the treatment of elbow flexion contractures.

3 Open Contacture Release and Surgical Approach

When nonoperative therapies fail to restore the functional arc of motion, patients with flexion contractures or extension contractures of >30° may be indicated for contracture release. Surgical approach should be determined by meticulous preoperative planning that notes prior incisions and CT findings. It can be helpful to organize common offending structures and their effects on flexion and extension (Table).

Table.
Careful attention to bony architecture and joint space can provide valuable information about motion impingement and arthrosis. In most cases, both the anterior compartment and the posterior compartment must be addressed, but this can often be achieved with a medial or lateral approach alone.
Figure 1.
Figure 1 shows a lateral radiograph of an active weightlifter with –10° of extension and 90° of flexion. CT showed osteophytes primarily over the anterior and posterior compartments of the ulnohumeral joint (Figure 2). Ulnar nerve paresthesia dictated our approach from the medial side.
Figure 2.

A medial over-the-top approach uses the medial supracondylar ridge as a landmark, subperiosteally reflecting the brachialis anteriorly.10 The ulnar nerve is neurolyzed and protected posteriorly. The flexor-pronator mass is split distally and elevated along with the brachialis as a single sleeve of muscle. The coronal plane of dissection should be the anterior half of the lateral epicondyle to avoid injury to the MCL. Large Bennett or Hohmann retractors can hinge on the lateral border of the humerus and provide clear visualization of the anterior capsule and the ulnohumeral joint. Exposure of the radiocapitellar joint is possible, but this joint is very deep in the operative field, and caution should be taken excising the anterolateral capsule because of the risk of radial nerve injury. The ulnar nerve can be temporarily transposed anteriorly to dissect posteriorly along the supracondylar ridge of the humerus. The triceps is reflected off the distal humerus. Occasionally, the posterior band of the MCL must be resected in severe extension contractures. If possible, the anterior bundle should be preserved. With this approach, the anterior capsule, distal humerus, coronoid process, posterior MCL, posterior capsule, and triceps can be addressed. The zone anterior to the radial head and the anterolateral and posterolateral capsule cannot be safely exposed with a medial approach. As described by Wada and colleagues,11 a primarily medial approach resulted in an average 64° increase in arc of motion.

Figure 3.
The lateral approach, commonly referred to as the column approach, uses the lateral supracondylar ridge to release distal fibers of the brachioradialis and the extensor carpi radialis longus (ECRL) (Figure 3). This exposure is limited by the radial nerve proximally and the posterior interosseous the ECRL and the extensor carpi radialis brevis. After the ECRL and the distal brachioradialis are released from the humerus, the superolateral capsule is visualized. After the brachialis and the radial nerve are elevated off the capsule, the capsule can be safely excised extending medially. Limited forearm rotation can also be addressed, as the proximal radioulnar joint (PRUJ) and the radiocapitellar joint can be exposed through this approach. Given the limits of lateral exposure, in some cases we release the lateral ulnar collateral ligament to better "hinge" open the elbow and obtain better visualization of the PRUJ and the medial joint. In isolation, the ligament can be repaired with suture anchors without causing instability or delaying rehabilitation. Husband and Hastings12 described a lateral approach with a mean 46° increased arc of motion—similar to the lateral column procedure with a mean 45° increased arc of motion, described by Mansat and Morrey.13 Last, a single posterior approach with wide medial and lateral flaps can also be used, but this has the distinct disadvantage of a risk of seroma or hematoma owing to the large dead space created. We typically prefer to initially approach contractures medially, as this allows ulnar nerve symptoms to be addressed. A lateral approach can be added to address forearm rotation and facilitate protection of the radial nerve/PIN during anterolateral capsular release in significant flexion contractures. Presence of heterotopic ossification or extra-articular malunion must also be considered, as it may dictate the surgical approach. If circumferential release of the elbow soft tissue and complete stripping of the distal humerus are performed for contracture release, simple collateral ligament repairs will be inadequate in providing stability immediately after surgery. In these extreme circumstances, we prefer to protect the ligament repair with
an internal joint stabilizer (Skeletal Dynamics) (Figure 4) and to initiate motion therapy immediately. External fixation (hinged or unhinged is rarely used in our practice.
Figure 4.

Arthroscopic Contracture Release and Technique

Recently, arthroscopic elbow contracture release, a technically demanding but effective treatment option, has gained popularity. Knowledge of neurovascular anatomy is a prerequisite to the prevention of devastating neurologic complications (ulnar, median, and radial nerve transections have been described14,15). Relative contraindications include extensive heterotopic ossification, ulnar nerve transposition, and limited arthroscopic experience. Functional improvements as well as average 26° to 42° increases in arc of motion have been described with arthroscopic release.16-18 In thin-framed patients with dense elbow capsular scarring (severe loss of elbow motion with hard block) and small joint space, arthroscopic release and particularly arthroscope insertion are notoriously difficult.

The patient may be placed in the prone, lateral decubitus, or supine position, depending on surgeon preference (Figure 5). Before surgery, portals and the ulnar nerve should be carefully outlined.19

Figure 5.
The median, ulnar, and medial antebrachial cutaneous nerves are at risk during placement of the proximal anteromedial portal, and the radial and lateral antebrachial cutaneous nerves are at risk during proximal anterolateral portal placement. Ulnar nerve decompression may also be performed through a limited incision before arthroscopic contracture release, and the nerve is protected during use of the shaver along the posteromedial joint capsule. Fluid management is required to ensure proper visualization and avoid excessive swelling.

We prefer to start by entering the posterior compartment and using the shaver to create a working space. All bone work and resectioning should be performed before capsular resection. After the joint and the olecranon fossa are identified, soft-tissue and bony débridement of the olecranon and the fossa can be performed. Care should be taken to protect the ulnar nerve when the posteromedial corner or medial gutter is approached.

Figure 6.
The anterior compartment is then visualized through the proximal anteromedial portal, and a working anterolateral portal is established (Figures 6A-6D). Often, bone forms in the coronoid fossa and the supracapitellar area, and this bone is resected. After completion of the bone work, the capsule is released or resected. In osteoarthritis cases, the capsule is usually relatively normal, and simple release proximally or distally is sufficient. In posttraumatic stiffness, however, marked capsular thickening and arthrofibrosis may occur, and in these cases more thorough capsular resection is advised. The capsulectomy is begun anteromedially, where the brachialis protects the median nerve and the brachial vessels. As the lateral border of the brachialis is approached, extreme caution must be taken to prevent radial or PIN injury. After the brachioradialis is visualized, more aggressive resection can be performed. Last, given the high risk of superficial infection classically associated with elbow arthroscopy, portal sites must be tightly closed and drains retained until scant drainage is noted.15

5 Additional Considerations

After surgery, the elbow is immobilized in maximal extension and supination with an anterior splint, and therapy is initiated either immediately or after temporary immobilization.16,19,20 Regional anesthesia is crucial in obtaining adequate pain control and establishing an immediate postoperative therapy program. The utility of continuous passive motion (CPM) in postoperative protocols is controversial. A retrospective case-control study of 32 patients matched on age, diagnosis, and contraction severity found no benefit of CPM use, and increased costs and hospital length of stay, leading the authors to recommend against CPM use.20

Neurovascular risks are associated with both open and arthroscopic elbow contracture release. Particularly concerning is the risk of traction ulnar neuropathy, described in upward of 20% of patients.21 Anatomical studies have found decreases in cubital tunnel and ulnar nerve area as elbow flexion increases with corresponding increased intraneural pressure,22 leading some authors to recommend prophylactic ulnar nerve release with limited preoperative flexion.15 Nevertheless, despite transposition, ulnar nerve symptoms were noted in 8 of 40 patients who underwent open contracture release for posttraumatic loss of elbow flexion.5 In a retrospective review of 164 open and arthroscopic elbow contracture releases, Williams and colleagues21 noted an 8.1% rate of postoperative new-onset ulnar nerve symptoms. The rate of ulnar neuropathy was nonsignificantly elevated among patients with preoperative flexion of <100° (15.2% vs 3.7%; P = .057). Recently, a retrospective review of 564 consecutive arthroscopic contracture releases found a significantly higher rate of delayed-onset ulnar neuritis among patients without prophylactic ulnar nerve decompression or transposition (11% vs 3%; P < .001).23 Further analysis revealed that, compared with decompression, ulnar nerve transposition did not offer additional benefit but was associated with a significantly higher rate of wound complications (19% vs 4%; P = .03). We favor prophylactic release, particularly in the setting of preoperative extension contracture. For open contracture release from the lateral approach, however, we do not routinely release the ulnar nerve unless there were preoperative symptoms.

Although open and arthroscopic contracture releases can provide durable outcomes in the setting of painless elbow stiffness, options are more limited in the treatment of the painful stiff elbow. Total elbow arthroplasty remains an option in low-demand elderly patients but is not without significant risk of complications.24 In addition, durability concerns and postoperative restrictions make total elbow arthroplasty less attractive to younger patients. Interposition arthroplasty may be indicated as a salvage procedure in the treatment of a young or high-demand patient with a stiff painful elbow.25 Elbow stability is crucial in obtaining a successful outcome, and data on optimal graft choices are limited.

Conclusion

Elbow stiffness, a common complication of trauma, significantly impairs activities of daily living. Early after trauma, therapy should be initiated to prevent contracture. In the absence of symptomatic arthritis, both open and arthroscopic contracture releases are effective surgical treatments in properly selected and motivated patients. Although more research is needed to establish the optimal surgical approach, severity and anatomical cause of contracture should guide decisions as to which approach to use. Having a thorough understanding of neurovascular anatomy and of prophylactic ulnar nerve decompression in the setting of limited preoperative flexion can mitigate complications.

References

1. Morrey BF, Askew LJ, Chao EY. A biomechanical study of normal functional elbow motion. J Bone Joint Surg Am. 1981;63(6):872-877.

2. Hotchkiss RN. Elbow contracture. In: Green DP, Rotchkiss RN, Pederson WC, Wolfe SW, eds. Green’s Operative Hand Surgery. 5th ed. New York, NY: Churchill-Livingstone; 2005:667-682.

3. Van Zeeland NL, Yamaguchi K. Arthroscopic capsular release of the elbow. J Shoulder Elbow Surg. 2010;19(2):13-19.

4. Morrey BF. Post-traumatic contracture of the elbow. Operative treatment, including distraction arthroplasty. J Bone Joint Surg Am. 1990;72(4):601-618.

5. Park MJ, Chang MJ, Lee YB, Kang HJ. Surgical release for posttraumatic loss of elbow flexion. J Bone Joint Surg Am. 2010;92(16):2692-2699.

6. Brouwer KM, Lindenhovius AL, Ring D. Loss of anterior translation of the distal humeral articular surface is associated with decreased elbow flexion. J Hand Surg Am. 2009;34(7):
1256-1260.

7. Taylor DC, Dalton JD, Seaber AV, Garrett WE. Viscoelastic properties of muscle-tendon units: the biomechanical effects of stretching. Am J Sports Med. 1990;18(3):300-309.

8. Green DP, McCoy H. Turnbuckle orthotic correction of elbow-flexion contractures after acute injuries. J Bone Joint Surg Am. 1979;61(7):1092-1095.

9. Gelinas JJ, Faber KJ, Patterson SD, King GJ. The effectiveness of turnbuckle splinting for elbow contractures. J Bone Joint Surg Br. 2000;82(1):74-78.

10. Hotchkiss RN, Kasparyan GN. The medial "over the top" approach to the elbow. Tech Orthop. 2000;15(2):105-112.

11. Wada T, Ishii S, Usui M, Miyano S. The medial approach for operative release of post-traumatic contracture of the elbow. J Bone Joint Surg Br. 2000;82(1):68-73.

12. Husband JB, Hastings H. The lateral approach for operative release of post-traumatic contracture of the elbow. J Bone Joint Surg Am. 1990;72(9):1353-1358.

13. Mansat P, Morrey BF. The column procedure: a limited lateral approach for extrinsic contracture of the elbow. J Bone Joint Surg Am. 1998;80(11):1603-1605.

14. Haapaniemi T, Berggren M, Adolfsson L. Complete transection of the median and radial nerves during arthroscopic release of post-traumatic elbow contracture. Arthroscopy. 1999;15(7):784-787.

15. Kelly EW, Morrey BF, O’Driscoll SW. Complications of elbow arthroscopy. J Bone Joint Surg Am. 2001;83(1):25-34.

16. Ball CM, Meunier M, Galatz LM, Calfee R, Yamaguchi K. Arthroscopic treatment of post-traumatic elbow contracture. J Shoulder Elbow Surg. 2002;11(6):624-629.

17. Ćefo I, Eygendaal D. Arthroscopic arthrolysis for posttraumatic elbow stiffness. J Shoulder Elbow Surg. 2011;20(3):434-439.

18. Nguyen D, Proper SI, MacDermid JC, King GJ, Faber KJ. Functional outcomes of arthroscopic capsular release of the elbow. Arthroscopy. 2006;22(8):842-849.

19. Sahajpal D, Choi T, Wright TW. Arthroscopic release of the stiff elbow. J Hand Surg. 2009;34(3):540-544.

20. Lindenhovius AL, Jupiter JB. The posttraumatic stiff elbow: a review of the literature. J Hand Surg. 2007;32(10):1605-1623.

21. Williams BG, Sotereanos DG, Baratz ME, Jarrett CD, Venouziou AI, Miller MC. The contracted elbow: is ulnar nerve release necessary? J Shoulder Elbow Surg. 2012;21(12):
1632-1636.

22. Gelberman RH, Yamaguchi K, Hollstien SB, et al. Changes in interstitial pressure and cross-sectional area of the cubital tunnel and of the ulnar nerve with flexion of the elbow. an experimental study in human cadavera. J Bone Joint Surg Am. 1998;80(4):492-501.

23. Blonna D, O’Driscoll SW. Delayed-onset ulnar neuritis after release of elbow contracture: preventive strategies derived from a study of 563 cases. Arthroscopy. 2014;30(8):947-956.

24. Mansat P, Morrey BF. Semiconstrained total elbow arthroplasty for ankylosed and stiff elbows. J Bone Joint Surg. 2000;82(9):1260-1268.

25. Hausman MR, Birnbaum PS. Interposition elbow arthroplasty. Tech Hand Up Extrem Surg. 2004;8(3):181-188.

References

1. Morrey BF, Askew LJ, Chao EY. A biomechanical study of normal functional elbow motion. J Bone Joint Surg Am. 1981;63(6):872-877.

2. Hotchkiss RN. Elbow contracture. In: Green DP, Rotchkiss RN, Pederson WC, Wolfe SW, eds. Green’s Operative Hand Surgery. 5th ed. New York, NY: Churchill-Livingstone; 2005:667-682.

3. Van Zeeland NL, Yamaguchi K. Arthroscopic capsular release of the elbow. J Shoulder Elbow Surg. 2010;19(2):13-19.

4. Morrey BF. Post-traumatic contracture of the elbow. Operative treatment, including distraction arthroplasty. J Bone Joint Surg Am. 1990;72(4):601-618.

5. Park MJ, Chang MJ, Lee YB, Kang HJ. Surgical release for posttraumatic loss of elbow flexion. J Bone Joint Surg Am. 2010;92(16):2692-2699.

6. Brouwer KM, Lindenhovius AL, Ring D. Loss of anterior translation of the distal humeral articular surface is associated with decreased elbow flexion. J Hand Surg Am. 2009;34(7):
1256-1260.

7. Taylor DC, Dalton JD, Seaber AV, Garrett WE. Viscoelastic properties of muscle-tendon units: the biomechanical effects of stretching. Am J Sports Med. 1990;18(3):300-309.

8. Green DP, McCoy H. Turnbuckle orthotic correction of elbow-flexion contractures after acute injuries. J Bone Joint Surg Am. 1979;61(7):1092-1095.

9. Gelinas JJ, Faber KJ, Patterson SD, King GJ. The effectiveness of turnbuckle splinting for elbow contractures. J Bone Joint Surg Br. 2000;82(1):74-78.

10. Hotchkiss RN, Kasparyan GN. The medial "over the top" approach to the elbow. Tech Orthop. 2000;15(2):105-112.

11. Wada T, Ishii S, Usui M, Miyano S. The medial approach for operative release of post-traumatic contracture of the elbow. J Bone Joint Surg Br. 2000;82(1):68-73.

12. Husband JB, Hastings H. The lateral approach for operative release of post-traumatic contracture of the elbow. J Bone Joint Surg Am. 1990;72(9):1353-1358.

13. Mansat P, Morrey BF. The column procedure: a limited lateral approach for extrinsic contracture of the elbow. J Bone Joint Surg Am. 1998;80(11):1603-1605.

14. Haapaniemi T, Berggren M, Adolfsson L. Complete transection of the median and radial nerves during arthroscopic release of post-traumatic elbow contracture. Arthroscopy. 1999;15(7):784-787.

15. Kelly EW, Morrey BF, O’Driscoll SW. Complications of elbow arthroscopy. J Bone Joint Surg Am. 2001;83(1):25-34.

16. Ball CM, Meunier M, Galatz LM, Calfee R, Yamaguchi K. Arthroscopic treatment of post-traumatic elbow contracture. J Shoulder Elbow Surg. 2002;11(6):624-629.

17. Ćefo I, Eygendaal D. Arthroscopic arthrolysis for posttraumatic elbow stiffness. J Shoulder Elbow Surg. 2011;20(3):434-439.

18. Nguyen D, Proper SI, MacDermid JC, King GJ, Faber KJ. Functional outcomes of arthroscopic capsular release of the elbow. Arthroscopy. 2006;22(8):842-849.

19. Sahajpal D, Choi T, Wright TW. Arthroscopic release of the stiff elbow. J Hand Surg. 2009;34(3):540-544.

20. Lindenhovius AL, Jupiter JB. The posttraumatic stiff elbow: a review of the literature. J Hand Surg. 2007;32(10):1605-1623.

21. Williams BG, Sotereanos DG, Baratz ME, Jarrett CD, Venouziou AI, Miller MC. The contracted elbow: is ulnar nerve release necessary? J Shoulder Elbow Surg. 2012;21(12):
1632-1636.

22. Gelberman RH, Yamaguchi K, Hollstien SB, et al. Changes in interstitial pressure and cross-sectional area of the cubital tunnel and of the ulnar nerve with flexion of the elbow. an experimental study in human cadavera. J Bone Joint Surg Am. 1998;80(4):492-501.

23. Blonna D, O’Driscoll SW. Delayed-onset ulnar neuritis after release of elbow contracture: preventive strategies derived from a study of 563 cases. Arthroscopy. 2014;30(8):947-956.

24. Mansat P, Morrey BF. Semiconstrained total elbow arthroplasty for ankylosed and stiff elbows. J Bone Joint Surg. 2000;82(9):1260-1268.

25. Hausman MR, Birnbaum PS. Interposition elbow arthroplasty. Tech Hand Up Extrem Surg. 2004;8(3):181-188.

Issue
The American Journal of Orthopedics - 46(5)
Issue
The American Journal of Orthopedics - 46(5)
Page Number
245-251
Page Number
245-251
Publications
Publications
Topics
Article Type
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Article PDF Media

Distal Radius Fractures: Reconstruction Approaches, Planning, and Principles

Article Type
Changed
Thu, 09/19/2019 - 13:20

Take-Home Points

  • Restore proper anatomic parameters; compare to the other side.
  • Don't forget about the DRU joint.
  • CT can aide in identifying subtle articular depression and severe comminution to change operative management.
  • Remember, there still is a role for external fixators; an alternative remains an internal spanning plate.
  • Respect the soft tissues, which can aide in reduction, however don't leave the operating room without feeling confident about your fixation.

Distal radius fracture (DRF), a common fracture, accounts for almost one sixth of all emergency department visits.1 With the advent of emerging technologies and refined technique, treatment options for DRFs have evolved. Although controversy remains regarding nonoperative vs operative treatment of DRFs in the elderly,2,3 select situations (open injuries, complex high-energy injuries, young age) warrant definitive fixation. Previously, internal fixation options were limited. Current technologies include locked fixed-angle plating, fragment-specific fixation, and locked variable-angle plating. These modalities aid in achieving and maintaining more anatomical fixation. This article summarizes tips, tricks, and planning for definitive external and internal fixation of complex DRFs.

Anatomical Considerations and Classification

The wrist joint, part of the complex articular network that begins at the forearm and ends at the distal interphalangeal joint, is the foundation for fine- and gross-motor skills. Understanding the anatomy of this network can provide a valuable roadmap for operative reconstruction.

At the wrist level, the radius bears most of the weight-bearing, and in some studies exhibits up to 80% of the load.1,4 The triangular distal radius bears this weight through a biconcave articular surface with facets for the lunate and scaphoid separated by an anteroposterior ridge.5-7 The radius also articulates with the ulnar head at the sigmoid notch to form the distal radioulnar (DRU) joint. Restoring the relationships of the DRU joint, the triangular fibrocartilage complex, and the ulnar variance is of paramount importance.1,8,9

Classical teaching calls for restoration of radial inclination to about 23°, volar tilt to 11° to 12°, and radial length to about 11 mm. Especially regarding volar tilt and radial length, however, cadaveric and clinical studies have found more variance, leading to use of the contralateral extremity as an operative template, particularly when closed reduction thought to be adequate deviates significantly from these parameters.1,4,7

DRF classification based on these principles has led to abundant representation in the literature.10-13 Many authors have focused on fracture lines, comminution degree, articular surface violation, and other anatomical or radiographic characteristics of DRF classification and operative fixation approach.10-13 In 2001, Fernandez9 proposed a classification system focused on energy or mechanism of injury. In comparisons,14 the Fernandez system had the highest interobserver reliability—higher than that of AO (Arbeitsgemeinschaft für Osteosynthesefragen).

Considerations for Operative Treatment: Column Theory

In the restoration of anatomical alignment in complex DRFs, it is important to consider the 3 joints and the 3 columns—radial, intermediate, and ulnar (Figure 1). [[{"fid":"201864","view_mode":"medstat_image_flush_left","attributes":{"class":"media-element file-medstat-image-flush-left","data-delta":"1"},"fields":{"format":"medstat_image_flush_left","field_file_image_caption[und][0][value]":"Figure 1.","field_file_image_credit[und][0][value]":"","field_file_image_caption[und][0][format]":"plain_text","field_file_image_credit[und][0][format]":"plain_text"},"type":"media","field_deltas":{"1":{"format":"medstat_image_flush_left","field_file_image_caption[und][0][value]":"Figure 1.","field_file_image_credit[und][0][value]":""}}}]]In addition, parallels between the distal radius and the tibial plateau can be considered because of similarities in operative goals. Restoration of mechanical axis, length, alignment, rotation, and articular surfaces is paramount.15 Considering multiple surgical approaches to address "bicolumnar injuries" and reconstructing the "simpler" columnar injury first are common principles.16

The goals of fracture fixation at the wrist are the same as at any other joint: anatomical reduction, stable fixation, and early range of motion (ROM). Column restoration can result in consistent achievement of those goals. Intuitively, there is a close correlation between anatomical alignment and functional results.17 Rebuilding the structural foundation of the columns with respect to buttressing and restoring the 3 radial articulations with the ulna, scaphoid, and lunate can consistently yield restoration of length, inclination, and tilt (Figure 2). [[{"fid":"201865","view_mode":"medstat_image_flush_right","attributes":{"class":"media-element file-medstat-image-flush-right","data-delta":"2"},"fields":{"format":"medstat_image_flush_right","field_file_image_caption[und][0][value]":"Figure 2.","field_file_image_credit[und][0][value]":"","field_file_image_caption[und][0][format]":"plain_text","field_file_image_credit[und][0][format]":"plain_text"},"type":"media","field_deltas":{"2":{"format":"medstat_image_flush_right","field_file_image_caption[und][0][value]":"Figure 2.","field_file_image_credit[und][0][value]":""}}}]]Next, we discuss the options available and how to use each to an advantage, individually or in hybrid constructs.

External Fixation: Is There Still a Role?

In the setting of highly comminuted, complex fractures, external fixation with Kirschner wires (K-wires) is a reasonable choice, with restoration of motion and strength within 75% to 80% of the uninjured wrist.18 In a 2-year study of 113 patients with comminuted metaphyseal DRFs randomly assigned to either external fixation or casting, Kreder and colleagues19 found a trend toward better clinical, functional, and radiographic outcomes with external fixation with or without K-wire fixation. There was improved restoration of radial length and palmar tilt with external fixation. A study of unstable DRF in patients with osteoporosis found that redisplacement was more common after treatment with a cast than after treatment with an external fixator.20 Although closed reduction and casting continue to have a role in the treatment of DRF, Kreder and colleagues19 found that remanipulation was necessary in at least 9% of cases. According to a meta-analysis21 of the literature on DRF treatment, 4 articles directly address the question of the superiority of external fixation over closed reduction and casting, and 3 of the 4 found more favorable radiographic and functional outcomes with external fixation.

External fixation is useful in treating complex DRFs with metaphyseal comminution. It can also be effective in the presence of simple articular involvement without depression of the joint surface. External fixation devices can span areas of soft-
tissue injury and are useful as manipulation tools in achieving anatomical reduction. Although external fixation is effective, its complications include pin-tract infection, nerve injury, loss of reduction, and loss of digital ROM. In a meta-analysis, Li-hai and colleagues22 found that external fixators had a complication rate of 30.9%. With this technique, it is important to avoid midcarpal distraction, excessive ulnar deviation, and excessive palmar flexion. Papadonikolakis and colleagues23 found that dis
traction of as little as 2 mm to 5 mm significantly affected the function of the flexor digitorum superficialis at the metacarpophalangeal joint. Over-distraction in wrist flexion can lead to lengthening of the extensor tendons and loss of full digital ROM. Excessive flexion and ulnar deviation can lead to median nerve compression and associated symptoms, as well as poor extensor and radial tendon length. In addition, prolonged distraction in excessive flexion combined with swelling and inflammation during fracture healing causes digital stiffness and contracture.23 Biomechanical studies have found that proximal pin placement in the radius, along with distal pin fixation in 6 metacarpal cortices through the second and third metacarpals, helps provide the strongest fixation.24

As for technique, pins are placed in the second metacarpal and radial shaft. With respect to the radius, the incision is made just proximal to the edge of the abductor pollicis longus muscle in the "bare area." Ideal pin placement is between the extensor carpi radialis longus and the extensor carpi radialis brevis, with care taken to avoid the radial sensory nerve, which lies between the extensor carpi radialis longus and the brachialis and emerges 9 cm proximal to the radial styloid.25 Next, a 2.5-cm to 3-cm incision is made over the palpable edge of the index metacarpal near the base. During drilling, the guide is placed at intersecting 45° angles, and the distal pin is placed 2 cm to 3 cm from the proximal pin. The proximal metacarpal pin is placed at the base of the metacarpal. The second metacarpal pin can also be placed first, with the external fixator used to judge proximal placement of the radial pin within the bare area.

Various supplements to external fixation have positive outcomes. Wolfe and colleagues18 found that using K-wires with the external fixation construct added stability in flexion/extension, radial/ulnar deviation, and rotational motion. They noted that fixation stability may depend more on the augmentation to fixation than on the external fixator itself. In a prospective, randomized trial, Moroni and colleagues26,27 found that, compared with standard pins, hydroxyapatite-coated pins had higher extraction torque, which was associated with improved fixation. When combined with external fixation, calcium phosphate cement also provided additional stability, allowing the bone filler to help maintain articular reduction and cortical continuity.28,29

External fixation has its disadvantages and complications. It can be bulky, and theoretically it contributes to higher rates of stiffness in the wrist and fingers.30-32 Higher rates of pin-site infection have been reported, along with hardware failure and associated loss of reduction, in patients treated with external fixation (Figures 3A-3C).31-33[[{"fid":"201866","view_mode":"medstat_image_flush_left","attributes":{"class":"media-element file-medstat-image-flush-left","data-delta":"3"},"fields":{"format":"medstat_image_flush_left","field_file_image_caption[und][0][value]":"Figure 3.","field_file_image_credit[und][0][value]":"","field_file_image_caption[und][0][format]":"plain_text","field_file_image_credit[und][0][format]":"plain_text"},"type":"media","field_deltas":{"3":{"format":"medstat_image_flush_left","field_file_image_caption[und][0][value]":"Figure 3.","field_file_image_credit[und][0][value]":""}}}]]In addition, joint overdistraction can adversely affect the length-tension curve and contribute to potential reflex sympathetic dystrophy, which can be devastating (Figures 4A, 4B).1,21,31,33 Despite these complications, external fixation remains a powerful tool in the treatment of high-energy DRFs. [[{"fid":"201867","view_mode":"medstat_image_flush_right","attributes":{"class":"media-element file-medstat-image-flush-right","data-delta":"4"},"fields":{"format":"medstat_image_flush_right","field_file_image_caption[und][0][value]":"Figure 4.","field_file_image_credit[und][0][value]":"","field_file_image_caption[und][0][format]":"plain_text","field_file_image_credit[und][0][format]":"plain_text"},"type":"media","field_deltas":{"4":{"format":"medstat_image_flush_right","field_file_image_caption[und][0][value]":"Figure 4.","field_file_image_credit[und][0][value]":""}}}]]In many cases, authors who compared open reduction and internal fixation (ORIF) with external fixation found no significant differences in outcome scores or function.31-34 In a meta-analysis of 917 patients, Margaliot and colleagues33 found no differences in pain, grip strength, wrist ROM, or radiographic parameters. More recently, in prospective randomized trials, both Egol and colleagues31 and Grewal and colleagues34 compared hybrid external fixation with ORIF, and, though early outcomes favored ORIF, 1-year follow-up comparisons were even, and there were no significant differences. These consistently reproducible results reaffirm keeping external fixation in the orthopedic toolbox.

Definitive Reconstruction With ORIF

Early nonlocked dorsal plating options for DRF fixation had unacceptable rates of plate failure, poor cosmesis, and extensor tendon complications.17,35-37 Subsequent technologic advances—multiple approaches, lower profile plating, and rigid, fragment-specific fixation—have allowed even the most complex fracture patterns to be addressed (Table). In malunited fractures, bone graft may not be required if the fracture is extra-articular and treated with a volar locking plate. [[{"fid":"201868","view_mode":"medstat_image_flush_left","attributes":{"class":"media-element file-medstat-image-flush-left","data-delta":"5"},"fields":{"format":"medstat_image_flush_left","field_file_image_caption[und][0][value]":"Table.","field_file_image_credit[und][0][value]":"","field_file_image_caption[und][0][format]":"plain_text","field_file_image_credit[und][0][format]":"plain_text"},"type":"media","field_deltas":{"5":{"format":"medstat_image_flush_left","field_file_image_caption[und][0][value]":"Table.","field_file_image_credit[und][0][value]":""}}}]]Other options include corticocancellous autograft from the iliac crest, hydroxyapatite synthetic grafts, and osteoconductive bone graft substitutes, such as bone morphogenic proteins. In addition, healing times are similar in cases, regardless of whether a graft was used.38

Involvement of the radial and intermediate columns should be addressed first. Although some may prefer a single volar plate, others may use fragment-specific fixation to buttress a comminuted radial styloid (in orthogonal fashion) and/or a dorsal ulnar fragment to restore the intermediate column and thereby fully restore the radial articular surface.39,40 Typically, restoring the radial and intermediate columns for radial articular reduction subsequently and simultaneously restores the majority of radial height and length. After the radial and intermediate columns are reduced and stabilized, the need for ulna column fixation can be determined. Important factors in ulna column restoration are severe osteoporosis and ulna head and/or neck comminution. Significant comminution throughout the metaphysis of both the radius and the ulna may also warrant stabilizing the ulna with internal fixation. Finally, any DRU joint instability noted on examination should also favor fixing the ulnar side.

Assessment of the distal ulna in these complex fractures goes beyond the involvement of an ulnar styloid fracture. Typically, fractures at the base of the ulnar styloid have been reported to have little clinical relevance, including a low incidence of associated DRU joint problems.41-43 Decisions to address the ulnar column are largely swayed by any instability found on DRU joint testing, as laxity caused by severe comminution can dictate the need for distal ring fixation to provide support. Even in the presence of a high-energy fracture in severely osteoporotic bone, the argument can be made to prevent instability by supporting the ulnar column. Stabilization of the ulnar articular surface can also be made more facile by creating an easier "A" fracture pattern (per AO classification) from a complex "C" to further aid in achieving efficient anatomical reduction. After preoperative planning is completed, depending on which columns need to be addressed, several surgical approaches can be considered to achieve maximum exposure and soft-tissue mobilization in order to successfully complete the operative fixation goals.

Volar Approach

An approach is selected for ideal exposure of a facile environment for definitive fixation. Access to the radial column can be gained with the extended flexor carpi radialis (FCR) approach. This approach allows visualization and removal of the appropriate deforming forces on the radial column to allow for fracture reduction by "opening the book," similar to that of tibial plateau reconstruction.44,45 It may be prudent to perform a preincision Allen test as well as a preoperative DRU joint examination for comparison after ORIF is complete. Compared with the classic Henry approach near the distal radius, going through the volar sheath of the FCR avoids many of the perforating radial artery branches. Avoiding stripping the radial artery of its surrounding fat and lymphatics prevents postoperative "cold intolerance." Retracting the FCR ulnarly and then incising the dorsal FCR sheath provide ready access to the pronator quadratus after collective ulnar mobilization of both the FCR and the flexor pollicis longus.44 In addition, for work near the distal FCR sheath, care must be taken to avoid the branch of the palmar cutaneous nerve that emerges about 5 cm proximal to the wrist flexion crease.46

Once at the level of the pronator quadratus, an "L-shape" incision can be made to reflect the muscle off the radius. Care must be taken when working too distal to avoid transection of the inserting volar wrist ligaments.44 Leaving a cuff for repair of the pronator remains controversial. In a recent case-control series, however, Hershman and colleagues47 did not find significant differences in function or complication rate in patients with and without repair. After reflection, adequate exposure of the radial column should be achieved. Ready access to the radial styloid for orthogonal plating can be obtained by releasing the brachioradialis, which simultaneously releases one of the primary fracture deforming forces.44 With this incision and exposure, if needed, dorsal bone grafting can be achieved from the volar side; however, care must be taken to protect the first dorsal compartment.48 The cutaneous branch of the median nerve may be at risk with this exposure, but avoiding dissection ulnar to the FCR tendon can help to reduce this risk.49

Before surgery, if the fracture pattern dictates a more ulnar approach, we prefer the extended carpal tunnel approach. Using the plane between the palmaris longus and the flexor digitorum superficialis medially and the FCR laterally, the extended carpal tunnel approach provides an obvious release of the flexor retinaculum but, more important, allows for extensile access to the sigmoid notch, the DRU joint, and the ulnar column.

Dorsal Approach

The dorsal approach is necessary in a few select cases. With a focus on fragment-specific fixation, presence of a significant dorsal ulnar fragment should warrant a dorsal approach.50 In addition, in select, rare cases in which volar access is limited or unavailable, dorsal access is the only option.50 Finally, if direct articular visualization is required, the dorsal approach typically is favored as the stronger radiocarpal ligaments found on the volar side are maintained.

Access should begin with an incision centered over the dorsal distal radius; a safe access point is just ulnar to the Lister tubercle. On incision of the retinaculum through a full-thickness excision, the third dorsal compartment is opened and the extensor pollicis longus (EPL) mobilized, fully exposing the dorsal distal radius. Work can be performed on either side of the EPL between the second and fourth dorsal compartments. Exposure typically is not an issue because of the pliable soft tissue of the dorsum, with ready access from styloid to styloid.44 Here, low-profile plates and/or mini-fragment-specific plate options should be used to minimize potential tendon damage.51 Care must also be taken to avoid damaging the radiocarpal or scapholunate ligaments.49 On closure, the retinaculum is repaired primarily; however, though some proponents advocate relocating the EPL tendon into its groove, we prefer leaving the EPL free within the surrounding soft tissue to reduce tension and promote unhindered excursion. The dorsal approach, though controversial and used inconsistently, should remain an important tool in anatomical restoration, especially in cases of complex fracture patterns.

Conclusion

Controversy still marks the lack of consensus on deciding which DRF treatment is optimal. Some investigators question moving away from external fixation and cite the lack of significantly better data relative to ORIF.21,52 The same proponents note that the only advantage over external fixation is earlier return to function and cite reports of tendon rupture and complications with both dorsal and volar fixation options.34,53-58 Other investigators find that operative treatment generally does not provide a significant improvement over nonoperative treatment.59

With the advent of lower profile locked plating, fragment-specific fixation, and variable-angle devices, comparative clinical trials are finding it difficult to keep up.60-64 Results from ongoing prospective randomized trials like ORCHID (Open Reduction and Internal Fixation Versus Casting for Highly Comminuted Intra-Articular Fractures of the Distal Radius; 500 patients >65 years old, 15 centers) will provide more definitive answers about ideal treatment.65

Anatomical restoration involves a versatile array of fragment fixation and reconstruction. Careful preoperative planning and a consistent approach to restoring the radial, intermediate, and ulnar columns, along with a proper surgical approach, are ideal. Many advances in internal fixation have been exceedingly helpful. Use of external fixation, especially in a bridging fashion with or without supplementation, is still valuable in many situations.

References

1. Liporace FA, Adams MR, Capo JT, Koval KJ. Distal radius fractures. J Orthop Trauma. 2009;23(10):739-748.

2. Lee YS, Wei TY, Cheng YC, Hsu TL, Huang CR. A comparative study of Colles’ fractures in patients between fifty and seventy years of age: percutaneous K-wiring versus volar locking plating. Int Orthop. 2012;36(4):789-794.

3. Diaz-Garcia RJ, Oda T, Shauver MJ, Chung KC. A systematic review of outcomes and complications of treating unstable distal radius fractures in the elderly. J Hand Surg Am. 2011;36(5):824-835.e2.

4. Ring D. Treatment of the neglected distal radius fracture. Clin Orthop Relat Res. 2005;(431):85-92.

5. Berger RA. Arthroscopic anatomy of the wrist and distal radioulnar joint. Hand Clin. 1999;15(3):393-413, vii.

6. Berger RA. The anatomy of the ligaments of the wrist and distal radioulnar joints. Clin Orthop Relat Res. 2001;(383):32-40.

7. McCann PA, Clarke D, Amirfeyz R, Bhatia R. The cadaveric anatomy of the distal radius: implications for the use of volar plates. Ann R Coll Surg Engl. 2012;94(2):116-120.

8. Ekenstam F. Osseous anatomy and articular relationships about the distal ulna. Hand Clin. 1998;14(2):161-164.

9. Fernandez DL. Distal radius fracture: the rationale of a classification. Chir Main. 2001;20(6):411-425.

10. Raskin KB, Melone CP Jr. Unstable articular fractures of the distal radius. Comparative techniques of ligamentotaxis. Orthop Clin North Am. 1993;24(2):275-286.

11. Melone CP Jr. Distal radius fractures: patterns of articular fragmentation. Orthop Clin North Am. 1993;24(2):239-253.

12. Jenkins NH. The unstable Colles’ fracture. J Hand Surg Br. 1989;14(2):149-154.

13. Cooney WP, Dobyns JH, Linscheid RL. Arthroscopy of the wrist: anatomy and classification of carpal instability. Arthroscopy. 1990;6(2):133-140.

14. Kural C, Sungur I, Kaya I, Ugras A, Ertürk A, Cetinus E. Evaluation of the reliability of classification systems used for distal radius fractures. Orthopedics. 2010;33(11):801.

15. Lipton HA, Wollstein R. Operative treatment of intraarticular distal radial fractures. Clin Orthop Relat Res. 1996;(327):110-124.

16. Wolfe SW. Distal radius fractures. Green’s Operative Hand Surgery. 6th ed. Philadelphia, PA: Churchill Livingstone; 2011:561-638.

17. Rikli DA, Regazzoni P. Fractures of the distal end of the radius treated by internal fixation and early function. A preliminary report of 20 cases. J Bone Joint Surg Br. 1996;78(4):
588-592.

18. Wolfe SW, Austin G, Lorenze M, Swigart CR, Panjabi MM. A biomechanical comparison of different wrist external fixators with and without K-wire augmentation. J Hand Surg Am. 1999;24(3):516-524.

19. Kreder HJ, Agel J, McKee MD, Schemitsch EH, Stephen D, Hanel DP. A randomized, controlled trial of distal radius fractures with metaphyseal displacement but without joint incongruity: closed reduction and casting versus closed reduction, spanning external fixation, and optional percutaneous K-wires. J Orthop Trauma. 2006;20(2):115-121.

20. Moroni A, Vannini F, Faldini C, Pegreffi F, Giannini S. Cast vs external fixation: a comparative study in elderly osteoporotic distal radial fracture patients. Scand J Surg. 2004;93(1):64-67.

21. Paksima N, Panchal A, Posner MA, Green SM, Mehiman CT, Hiebert R. A meta-analysis of the literature on distal radius fractures: review of 615 articles. Bull Hosp Jt Dis. 2004;62(1-2):40-46.

22. Li-hai Z, Ya-nan W, Zhi M, et al. Volar locking plate versus external fixation for the treatment of unstable distal radial fractures: a meta-analysis of randomized controlled trials.
J Surg Res. 2015;193(1):324-333.

23. Papadonikolakis A, Shen J, Garrett JP, Davis SM, Ruch DS. The effect of increasing distraction on digital motion after external fixation of the wrist. J Hand Surg Am. 2005;30(4):
773-779.

24. Seitz WH Jr, Froimson AI, Brooks DB, et al. Biomechanical analysis of pin placement and pin size for external fixation of distal radius fractures. Clin Orthop Relat Res. 1990;(251):
207-212.

25. Beldner S, Zlotolow DA, Melone CP Jr, Agnes AM, Jones MH. Anatomy of the lateral antebrachial cutaneous and superficial radial nerves in the forearm: a cadaveric and clinical study. J Hand Surg Am. 2005;30(6):1226-1230.

26. Moroni A, Faldini C, Marchetti S, Manca M, Consoli V, Giannini S. Improvement of the bone-pin interface strength in osteoporotic bone with use of hydroxyapatite-coated tapered external-fixation pins. A prospective, randomized clinical study of wrist fractures. J Bone Joint Surg Am. 2001;83(5):717-721.

27. Moroni A, Heikkila J, Magyar G, Toksvig-Larsen S, Giannini S. Fixation strength and pin tract infection of hydroxyapatite-coated tapered pins. Clin Orthop Relat Res. 2001;(388):209-217.

28. Higgins TF, Dodds SD, Wolfe SW. A biomechanical analysis of fixation of intra-articular distal radial fractures with calcium-phosphate bone cement. J Bone Joint Surg Am. 2002;84(9):1579-1586.

29. Tobe M, Mizutani K, Tsubuku Y. Treatment of distal radius fracture with the use of calcium phosphate bone cement as a filler. Tech Hand Up Extrem Surg. 2004;8(2):95-101.

30. Capo JT, Rossy W, Henry P, Maurer RJ, Naidu S, Chen L.
External fixation of distal radius fractures: effect of distraction and duration. J Hand Surg Am. 2009;34(9):1605-1611.

31. Egol K, Walsh M, Tejwani N, McLaurin T, Wynn C, Paksima N. Bridging external fixation and supplementary Kirschner-wire fixation versus volar locked plating for unstable fractures of the distal radius: a randomised, prospective trial. J Bone Joint Surg Br. 2008;90(9):1214-1221.

32. Egol KA, Paksima N, Puopolo S, Klugman J, Hiebert R, Koval KJ. Treatment of external fixation pins about the wrist: a prospective, randomized trial. J Bone Joint Surg Am. 2006;88(2):349-354.

33. Margaliot Z, Haase SC, Kotsis SV, Kim HM, Chung KC. A meta-analysis of outcomes of external fixation versus plate osteosynthesis for unstable distal radius fractures. J Hand Surg Am. 2005;30(6):1185-1199.

34. Grewal R, MacDermid JC, King GJ, Faber KJ. Open reduction internal fixation versus percutaneous pinning with external fixation of distal radius fractures: a prospective, randomized clinical trial. J Hand Surg Am. 2011;36(12):
1899-1906.

35. Axelrod TS, McMurtry RY. Open reduction and internal fixation of comminuted, intraarticular fractures of the distal radius. J Hand Surg Am. 1990;15(1):1-11.

36. Hove LM, Nilsen PT, Furnes O, Oulie HE, Solheim E, Mölster AO. Open reduction and internal fixation of displaced intraarticular fractures of the distal radius. 31 patients followed for 3-7 years. Acta Orthop Scand. 1997;68(1):59-63.

37. Carter PR, Frederick HA, Laseter GF. Open reduction and internal fixation of unstable distal radius fractures with a low-profile plate: a multicenter study of 73 fractures. J Hand Surg Am. 1998;23(2):300-307.

38. Mugnai R, Tarallo L, Lancellotti E, et al. Corrective osteotomies of the radius: grafting or not? World J Orthop. 2016;7(2):128-135.

39. Tang P, Ding A, Uzumcugil A. Radial column and volar plating (RCVP) for distal radius fractures with a radial styloid component or severe comminution. Tech Hand Up Extrem Surg. 2010;14(3):143-149.

40. Helmerhorst GT, Kloen P. Orthogonal plating of intra-articular distal radius fractures with an associated radial column fracture via a single volar approach. Injury. 2012;43(8):1307-1312.

41. May MM, Lawton JN, Blazar PE. Ulnar styloid fractures associated with distal radius fractures: incidence and implications for distal radioulnar joint instability. J Hand Surg Am. 2002;27(6):965-971.

42. Souer JS, Ring D, Matschke S, Audige L, Marent-Huber M, Jupiter JB; AOCID Prospective ORIF Distal Radius Study Group. Effect of an unrepaired fracture of the ulnar styloid base on outcome after plate-and-screw fixation of a distal radial fracture. J Bone Joint Surg Am. 2009;91(4):830-838.

43. Noda K, Goto A, Murase T, Sugamoto K, Yoshikawa H, Moritomo H. Interosseous membrane of the forearm: an anatomical study of ligament attachment locations. J Hand Surg Am. 2009;34(3):415-422.

44. Catalano LW 3rd, Zlotolow DA, Hitchcock PB, Shah SN, Barron OA. Surgical exposures of the radius and ulna. J Am Acad Orthop Surg. 2011;19(7):430-438.

45. Orbay JL, Badia A, Indriago IR, et al. The extended flexor carpi radialis approach: a new perspective for the distal radius fracture. Tech Hand Up Extrem Surg. 2001;5(4):204-211.

46. Hobbs RA, Magnussen PA, Tonkin MA. Palmar cutaneous branch of the median nerve. J Hand Surg Am. 1990;15(1):38-43.

47. Hershman SH, Immerman I, Bechtel C, Lekic N, Paksima N, Egol KA. The effects of pronator quadratus repair on outcomes after volar plating of distal radius fractures. J Orthop Trauma. 2013;27(3):130-133.

48. Prommersberger KJ, Lanz UB. Corrective osteotomy of the distal radius through volar approach. Tech Hand Up Extrem Surg. 2004;8(2):70-77.

49. Ilyas AM. Surgical approaches to the distal radius. Hand (N Y). 2011;6(1):8-17.

50. Tavakolian JD, Jupiter JB. Dorsal plating for distal radius fractures. Hand Clin. 2005;21(3):341-346.

51. Yu YR, Makhni MC, Tabrizi S, Rozental TD, Mundanthanam G, Day CS. Complications of low-profile dorsal versus volar locking plates in the distal radius: a comparative study. J Hand Surg Am. 2011;36(7):1135-1141.

52. Mattila VM, Huttunen TT, Sillanpää P, Niemi S, Pihlajamäki H, Kannus P. Significant change in the surgical treatment of distal radius fractures: a nationwide study between 1998 and 2008 in Finland. J Trauma. 2011;71(4):939-942.

53. Wilcke MK, Abbaszadegan H, Adolphson PY. Wrist function recovers more rapidly after volar locked plating than after external fixation but the outcomes are similar after 1 year. Acta Orthop. 2011;82(1):76-81.

54. Ward CM, Kuhl TL, Adams BD. Early complications of volar plating of distal radius fractures and their relationship to surgeon experience. Hand (N Y). 2011;6(2):185-189.

55. Soong M, van Leerdam R, Guitton TG, Got C, Katarincic J, Ring D. Fracture of the distal radius: risk factors for complications after locked volar plate fixation. J Hand Surg Am. 2011;36(1):3-9.

56. Soong M, Earp BE, Bishop G, Leung A, Blazar P. Volar locking plate implant prominence and flexor tendon rupture. J Bone Joint Surg Am. 2011;93(4):328-335.

57. Jeudy J, Steiger V, Boyer P, Cronier P, Bizot P, Massin P. Treatment of complex fractures of the distal radius: a prospective randomised comparison of external fixation ‘versus’ locked volar plating. Injury. 2012;43(2):174-179.

58. Berglund LM, Messer TM. Complications of volar plate fixation for managing distal radius fractures. J Am Acad Orthop Surg. 2009;17(6):369-377.

59. Egol KA, Walsh M, Romo-Cardoso S, Dorsky S, Paksima N. Distal radial fractures in the elderly: operative compared with nonoperative treatment. J Bone Joint Surg Am. 2010;92(9):1851-1857.

60. Wall LB, Brodt MD, Silva MJ, Boyer MI, Calfee RP. The effects of screw length on stability of simulated osteoporotic distal radius fractures fixed with volar locking plates. J Hand Surg Am. 2012;37(3):446-453.

61. Dahl WJ, Nassab PF, Burgess KM, et al. Biomechanical properties of fixed-angle volar distal radius plates under dynamic loading. J Hand Surg Am. 2012;37(7):1381-1387.

62. Park JH, Hagopian J, Ilyas AM. Variable-angle locking screw volar plating of distal radius fractures. Hand Clin. 2010;26(3):373-380, vi.

63. Pensy RA, Brunton LM, Parks BG, Higgins JP, Chhabra AB. Single-incision extensile volar approach to the distal radius and concurrent carpal tunnel release: cadaveric study. J Hand Surg Am. 2010;35(2):217-222.

64. Klos K, Rausch S, Löffler M, et al. A biomechanical comparison of a biodegradable volar locked plate with two titanium volar locked plates in a distal radius fracture model. J Trauma. 2010;68(4):984-991.

65. Bartl C, Stengel D, Bruckner T, et al. Open reduction and internal fixation versus casting for highly comminuted and intra-articular fractures of the distal radius (ORCHID): protocol for a randomized clinical multi-center trial. Trials. 2011;12:84

Article PDF
Author and Disclosure Information

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

Issue
The American Journal of Orthopedics - 46(5)
Publications
Topics
Page Number
238-244
Sections
Author and Disclosure Information

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

Author and Disclosure Information

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

Article PDF
Article PDF

Take-Home Points

  • Restore proper anatomic parameters; compare to the other side.
  • Don't forget about the DRU joint.
  • CT can aide in identifying subtle articular depression and severe comminution to change operative management.
  • Remember, there still is a role for external fixators; an alternative remains an internal spanning plate.
  • Respect the soft tissues, which can aide in reduction, however don't leave the operating room without feeling confident about your fixation.

Distal radius fracture (DRF), a common fracture, accounts for almost one sixth of all emergency department visits.1 With the advent of emerging technologies and refined technique, treatment options for DRFs have evolved. Although controversy remains regarding nonoperative vs operative treatment of DRFs in the elderly,2,3 select situations (open injuries, complex high-energy injuries, young age) warrant definitive fixation. Previously, internal fixation options were limited. Current technologies include locked fixed-angle plating, fragment-specific fixation, and locked variable-angle plating. These modalities aid in achieving and maintaining more anatomical fixation. This article summarizes tips, tricks, and planning for definitive external and internal fixation of complex DRFs.

Anatomical Considerations and Classification

The wrist joint, part of the complex articular network that begins at the forearm and ends at the distal interphalangeal joint, is the foundation for fine- and gross-motor skills. Understanding the anatomy of this network can provide a valuable roadmap for operative reconstruction.

At the wrist level, the radius bears most of the weight-bearing, and in some studies exhibits up to 80% of the load.1,4 The triangular distal radius bears this weight through a biconcave articular surface with facets for the lunate and scaphoid separated by an anteroposterior ridge.5-7 The radius also articulates with the ulnar head at the sigmoid notch to form the distal radioulnar (DRU) joint. Restoring the relationships of the DRU joint, the triangular fibrocartilage complex, and the ulnar variance is of paramount importance.1,8,9

Classical teaching calls for restoration of radial inclination to about 23°, volar tilt to 11° to 12°, and radial length to about 11 mm. Especially regarding volar tilt and radial length, however, cadaveric and clinical studies have found more variance, leading to use of the contralateral extremity as an operative template, particularly when closed reduction thought to be adequate deviates significantly from these parameters.1,4,7

DRF classification based on these principles has led to abundant representation in the literature.10-13 Many authors have focused on fracture lines, comminution degree, articular surface violation, and other anatomical or radiographic characteristics of DRF classification and operative fixation approach.10-13 In 2001, Fernandez9 proposed a classification system focused on energy or mechanism of injury. In comparisons,14 the Fernandez system had the highest interobserver reliability—higher than that of AO (Arbeitsgemeinschaft für Osteosynthesefragen).

Considerations for Operative Treatment: Column Theory

In the restoration of anatomical alignment in complex DRFs, it is important to consider the 3 joints and the 3 columns—radial, intermediate, and ulnar (Figure 1). [[{"fid":"201864","view_mode":"medstat_image_flush_left","attributes":{"class":"media-element file-medstat-image-flush-left","data-delta":"1"},"fields":{"format":"medstat_image_flush_left","field_file_image_caption[und][0][value]":"Figure 1.","field_file_image_credit[und][0][value]":"","field_file_image_caption[und][0][format]":"plain_text","field_file_image_credit[und][0][format]":"plain_text"},"type":"media","field_deltas":{"1":{"format":"medstat_image_flush_left","field_file_image_caption[und][0][value]":"Figure 1.","field_file_image_credit[und][0][value]":""}}}]]In addition, parallels between the distal radius and the tibial plateau can be considered because of similarities in operative goals. Restoration of mechanical axis, length, alignment, rotation, and articular surfaces is paramount.15 Considering multiple surgical approaches to address "bicolumnar injuries" and reconstructing the "simpler" columnar injury first are common principles.16

The goals of fracture fixation at the wrist are the same as at any other joint: anatomical reduction, stable fixation, and early range of motion (ROM). Column restoration can result in consistent achievement of those goals. Intuitively, there is a close correlation between anatomical alignment and functional results.17 Rebuilding the structural foundation of the columns with respect to buttressing and restoring the 3 radial articulations with the ulna, scaphoid, and lunate can consistently yield restoration of length, inclination, and tilt (Figure 2). [[{"fid":"201865","view_mode":"medstat_image_flush_right","attributes":{"class":"media-element file-medstat-image-flush-right","data-delta":"2"},"fields":{"format":"medstat_image_flush_right","field_file_image_caption[und][0][value]":"Figure 2.","field_file_image_credit[und][0][value]":"","field_file_image_caption[und][0][format]":"plain_text","field_file_image_credit[und][0][format]":"plain_text"},"type":"media","field_deltas":{"2":{"format":"medstat_image_flush_right","field_file_image_caption[und][0][value]":"Figure 2.","field_file_image_credit[und][0][value]":""}}}]]Next, we discuss the options available and how to use each to an advantage, individually or in hybrid constructs.

External Fixation: Is There Still a Role?

In the setting of highly comminuted, complex fractures, external fixation with Kirschner wires (K-wires) is a reasonable choice, with restoration of motion and strength within 75% to 80% of the uninjured wrist.18 In a 2-year study of 113 patients with comminuted metaphyseal DRFs randomly assigned to either external fixation or casting, Kreder and colleagues19 found a trend toward better clinical, functional, and radiographic outcomes with external fixation with or without K-wire fixation. There was improved restoration of radial length and palmar tilt with external fixation. A study of unstable DRF in patients with osteoporosis found that redisplacement was more common after treatment with a cast than after treatment with an external fixator.20 Although closed reduction and casting continue to have a role in the treatment of DRF, Kreder and colleagues19 found that remanipulation was necessary in at least 9% of cases. According to a meta-analysis21 of the literature on DRF treatment, 4 articles directly address the question of the superiority of external fixation over closed reduction and casting, and 3 of the 4 found more favorable radiographic and functional outcomes with external fixation.

External fixation is useful in treating complex DRFs with metaphyseal comminution. It can also be effective in the presence of simple articular involvement without depression of the joint surface. External fixation devices can span areas of soft-
tissue injury and are useful as manipulation tools in achieving anatomical reduction. Although external fixation is effective, its complications include pin-tract infection, nerve injury, loss of reduction, and loss of digital ROM. In a meta-analysis, Li-hai and colleagues22 found that external fixators had a complication rate of 30.9%. With this technique, it is important to avoid midcarpal distraction, excessive ulnar deviation, and excessive palmar flexion. Papadonikolakis and colleagues23 found that dis
traction of as little as 2 mm to 5 mm significantly affected the function of the flexor digitorum superficialis at the metacarpophalangeal joint. Over-distraction in wrist flexion can lead to lengthening of the extensor tendons and loss of full digital ROM. Excessive flexion and ulnar deviation can lead to median nerve compression and associated symptoms, as well as poor extensor and radial tendon length. In addition, prolonged distraction in excessive flexion combined with swelling and inflammation during fracture healing causes digital stiffness and contracture.23 Biomechanical studies have found that proximal pin placement in the radius, along with distal pin fixation in 6 metacarpal cortices through the second and third metacarpals, helps provide the strongest fixation.24

As for technique, pins are placed in the second metacarpal and radial shaft. With respect to the radius, the incision is made just proximal to the edge of the abductor pollicis longus muscle in the "bare area." Ideal pin placement is between the extensor carpi radialis longus and the extensor carpi radialis brevis, with care taken to avoid the radial sensory nerve, which lies between the extensor carpi radialis longus and the brachialis and emerges 9 cm proximal to the radial styloid.25 Next, a 2.5-cm to 3-cm incision is made over the palpable edge of the index metacarpal near the base. During drilling, the guide is placed at intersecting 45° angles, and the distal pin is placed 2 cm to 3 cm from the proximal pin. The proximal metacarpal pin is placed at the base of the metacarpal. The second metacarpal pin can also be placed first, with the external fixator used to judge proximal placement of the radial pin within the bare area.

Various supplements to external fixation have positive outcomes. Wolfe and colleagues18 found that using K-wires with the external fixation construct added stability in flexion/extension, radial/ulnar deviation, and rotational motion. They noted that fixation stability may depend more on the augmentation to fixation than on the external fixator itself. In a prospective, randomized trial, Moroni and colleagues26,27 found that, compared with standard pins, hydroxyapatite-coated pins had higher extraction torque, which was associated with improved fixation. When combined with external fixation, calcium phosphate cement also provided additional stability, allowing the bone filler to help maintain articular reduction and cortical continuity.28,29

External fixation has its disadvantages and complications. It can be bulky, and theoretically it contributes to higher rates of stiffness in the wrist and fingers.30-32 Higher rates of pin-site infection have been reported, along with hardware failure and associated loss of reduction, in patients treated with external fixation (Figures 3A-3C).31-33[[{"fid":"201866","view_mode":"medstat_image_flush_left","attributes":{"class":"media-element file-medstat-image-flush-left","data-delta":"3"},"fields":{"format":"medstat_image_flush_left","field_file_image_caption[und][0][value]":"Figure 3.","field_file_image_credit[und][0][value]":"","field_file_image_caption[und][0][format]":"plain_text","field_file_image_credit[und][0][format]":"plain_text"},"type":"media","field_deltas":{"3":{"format":"medstat_image_flush_left","field_file_image_caption[und][0][value]":"Figure 3.","field_file_image_credit[und][0][value]":""}}}]]In addition, joint overdistraction can adversely affect the length-tension curve and contribute to potential reflex sympathetic dystrophy, which can be devastating (Figures 4A, 4B).1,21,31,33 Despite these complications, external fixation remains a powerful tool in the treatment of high-energy DRFs. [[{"fid":"201867","view_mode":"medstat_image_flush_right","attributes":{"class":"media-element file-medstat-image-flush-right","data-delta":"4"},"fields":{"format":"medstat_image_flush_right","field_file_image_caption[und][0][value]":"Figure 4.","field_file_image_credit[und][0][value]":"","field_file_image_caption[und][0][format]":"plain_text","field_file_image_credit[und][0][format]":"plain_text"},"type":"media","field_deltas":{"4":{"format":"medstat_image_flush_right","field_file_image_caption[und][0][value]":"Figure 4.","field_file_image_credit[und][0][value]":""}}}]]In many cases, authors who compared open reduction and internal fixation (ORIF) with external fixation found no significant differences in outcome scores or function.31-34 In a meta-analysis of 917 patients, Margaliot and colleagues33 found no differences in pain, grip strength, wrist ROM, or radiographic parameters. More recently, in prospective randomized trials, both Egol and colleagues31 and Grewal and colleagues34 compared hybrid external fixation with ORIF, and, though early outcomes favored ORIF, 1-year follow-up comparisons were even, and there were no significant differences. These consistently reproducible results reaffirm keeping external fixation in the orthopedic toolbox.

Definitive Reconstruction With ORIF

Early nonlocked dorsal plating options for DRF fixation had unacceptable rates of plate failure, poor cosmesis, and extensor tendon complications.17,35-37 Subsequent technologic advances—multiple approaches, lower profile plating, and rigid, fragment-specific fixation—have allowed even the most complex fracture patterns to be addressed (Table). In malunited fractures, bone graft may not be required if the fracture is extra-articular and treated with a volar locking plate. [[{"fid":"201868","view_mode":"medstat_image_flush_left","attributes":{"class":"media-element file-medstat-image-flush-left","data-delta":"5"},"fields":{"format":"medstat_image_flush_left","field_file_image_caption[und][0][value]":"Table.","field_file_image_credit[und][0][value]":"","field_file_image_caption[und][0][format]":"plain_text","field_file_image_credit[und][0][format]":"plain_text"},"type":"media","field_deltas":{"5":{"format":"medstat_image_flush_left","field_file_image_caption[und][0][value]":"Table.","field_file_image_credit[und][0][value]":""}}}]]Other options include corticocancellous autograft from the iliac crest, hydroxyapatite synthetic grafts, and osteoconductive bone graft substitutes, such as bone morphogenic proteins. In addition, healing times are similar in cases, regardless of whether a graft was used.38

Involvement of the radial and intermediate columns should be addressed first. Although some may prefer a single volar plate, others may use fragment-specific fixation to buttress a comminuted radial styloid (in orthogonal fashion) and/or a dorsal ulnar fragment to restore the intermediate column and thereby fully restore the radial articular surface.39,40 Typically, restoring the radial and intermediate columns for radial articular reduction subsequently and simultaneously restores the majority of radial height and length. After the radial and intermediate columns are reduced and stabilized, the need for ulna column fixation can be determined. Important factors in ulna column restoration are severe osteoporosis and ulna head and/or neck comminution. Significant comminution throughout the metaphysis of both the radius and the ulna may also warrant stabilizing the ulna with internal fixation. Finally, any DRU joint instability noted on examination should also favor fixing the ulnar side.

Assessment of the distal ulna in these complex fractures goes beyond the involvement of an ulnar styloid fracture. Typically, fractures at the base of the ulnar styloid have been reported to have little clinical relevance, including a low incidence of associated DRU joint problems.41-43 Decisions to address the ulnar column are largely swayed by any instability found on DRU joint testing, as laxity caused by severe comminution can dictate the need for distal ring fixation to provide support. Even in the presence of a high-energy fracture in severely osteoporotic bone, the argument can be made to prevent instability by supporting the ulnar column. Stabilization of the ulnar articular surface can also be made more facile by creating an easier "A" fracture pattern (per AO classification) from a complex "C" to further aid in achieving efficient anatomical reduction. After preoperative planning is completed, depending on which columns need to be addressed, several surgical approaches can be considered to achieve maximum exposure and soft-tissue mobilization in order to successfully complete the operative fixation goals.

Volar Approach

An approach is selected for ideal exposure of a facile environment for definitive fixation. Access to the radial column can be gained with the extended flexor carpi radialis (FCR) approach. This approach allows visualization and removal of the appropriate deforming forces on the radial column to allow for fracture reduction by "opening the book," similar to that of tibial plateau reconstruction.44,45 It may be prudent to perform a preincision Allen test as well as a preoperative DRU joint examination for comparison after ORIF is complete. Compared with the classic Henry approach near the distal radius, going through the volar sheath of the FCR avoids many of the perforating radial artery branches. Avoiding stripping the radial artery of its surrounding fat and lymphatics prevents postoperative "cold intolerance." Retracting the FCR ulnarly and then incising the dorsal FCR sheath provide ready access to the pronator quadratus after collective ulnar mobilization of both the FCR and the flexor pollicis longus.44 In addition, for work near the distal FCR sheath, care must be taken to avoid the branch of the palmar cutaneous nerve that emerges about 5 cm proximal to the wrist flexion crease.46

Once at the level of the pronator quadratus, an "L-shape" incision can be made to reflect the muscle off the radius. Care must be taken when working too distal to avoid transection of the inserting volar wrist ligaments.44 Leaving a cuff for repair of the pronator remains controversial. In a recent case-control series, however, Hershman and colleagues47 did not find significant differences in function or complication rate in patients with and without repair. After reflection, adequate exposure of the radial column should be achieved. Ready access to the radial styloid for orthogonal plating can be obtained by releasing the brachioradialis, which simultaneously releases one of the primary fracture deforming forces.44 With this incision and exposure, if needed, dorsal bone grafting can be achieved from the volar side; however, care must be taken to protect the first dorsal compartment.48 The cutaneous branch of the median nerve may be at risk with this exposure, but avoiding dissection ulnar to the FCR tendon can help to reduce this risk.49

Before surgery, if the fracture pattern dictates a more ulnar approach, we prefer the extended carpal tunnel approach. Using the plane between the palmaris longus and the flexor digitorum superficialis medially and the FCR laterally, the extended carpal tunnel approach provides an obvious release of the flexor retinaculum but, more important, allows for extensile access to the sigmoid notch, the DRU joint, and the ulnar column.

Dorsal Approach

The dorsal approach is necessary in a few select cases. With a focus on fragment-specific fixation, presence of a significant dorsal ulnar fragment should warrant a dorsal approach.50 In addition, in select, rare cases in which volar access is limited or unavailable, dorsal access is the only option.50 Finally, if direct articular visualization is required, the dorsal approach typically is favored as the stronger radiocarpal ligaments found on the volar side are maintained.

Access should begin with an incision centered over the dorsal distal radius; a safe access point is just ulnar to the Lister tubercle. On incision of the retinaculum through a full-thickness excision, the third dorsal compartment is opened and the extensor pollicis longus (EPL) mobilized, fully exposing the dorsal distal radius. Work can be performed on either side of the EPL between the second and fourth dorsal compartments. Exposure typically is not an issue because of the pliable soft tissue of the dorsum, with ready access from styloid to styloid.44 Here, low-profile plates and/or mini-fragment-specific plate options should be used to minimize potential tendon damage.51 Care must also be taken to avoid damaging the radiocarpal or scapholunate ligaments.49 On closure, the retinaculum is repaired primarily; however, though some proponents advocate relocating the EPL tendon into its groove, we prefer leaving the EPL free within the surrounding soft tissue to reduce tension and promote unhindered excursion. The dorsal approach, though controversial and used inconsistently, should remain an important tool in anatomical restoration, especially in cases of complex fracture patterns.

Conclusion

Controversy still marks the lack of consensus on deciding which DRF treatment is optimal. Some investigators question moving away from external fixation and cite the lack of significantly better data relative to ORIF.21,52 The same proponents note that the only advantage over external fixation is earlier return to function and cite reports of tendon rupture and complications with both dorsal and volar fixation options.34,53-58 Other investigators find that operative treatment generally does not provide a significant improvement over nonoperative treatment.59

With the advent of lower profile locked plating, fragment-specific fixation, and variable-angle devices, comparative clinical trials are finding it difficult to keep up.60-64 Results from ongoing prospective randomized trials like ORCHID (Open Reduction and Internal Fixation Versus Casting for Highly Comminuted Intra-Articular Fractures of the Distal Radius; 500 patients >65 years old, 15 centers) will provide more definitive answers about ideal treatment.65

Anatomical restoration involves a versatile array of fragment fixation and reconstruction. Careful preoperative planning and a consistent approach to restoring the radial, intermediate, and ulnar columns, along with a proper surgical approach, are ideal. Many advances in internal fixation have been exceedingly helpful. Use of external fixation, especially in a bridging fashion with or without supplementation, is still valuable in many situations.

Take-Home Points

  • Restore proper anatomic parameters; compare to the other side.
  • Don't forget about the DRU joint.
  • CT can aide in identifying subtle articular depression and severe comminution to change operative management.
  • Remember, there still is a role for external fixators; an alternative remains an internal spanning plate.
  • Respect the soft tissues, which can aide in reduction, however don't leave the operating room without feeling confident about your fixation.

Distal radius fracture (DRF), a common fracture, accounts for almost one sixth of all emergency department visits.1 With the advent of emerging technologies and refined technique, treatment options for DRFs have evolved. Although controversy remains regarding nonoperative vs operative treatment of DRFs in the elderly,2,3 select situations (open injuries, complex high-energy injuries, young age) warrant definitive fixation. Previously, internal fixation options were limited. Current technologies include locked fixed-angle plating, fragment-specific fixation, and locked variable-angle plating. These modalities aid in achieving and maintaining more anatomical fixation. This article summarizes tips, tricks, and planning for definitive external and internal fixation of complex DRFs.

Anatomical Considerations and Classification

The wrist joint, part of the complex articular network that begins at the forearm and ends at the distal interphalangeal joint, is the foundation for fine- and gross-motor skills. Understanding the anatomy of this network can provide a valuable roadmap for operative reconstruction.

At the wrist level, the radius bears most of the weight-bearing, and in some studies exhibits up to 80% of the load.1,4 The triangular distal radius bears this weight through a biconcave articular surface with facets for the lunate and scaphoid separated by an anteroposterior ridge.5-7 The radius also articulates with the ulnar head at the sigmoid notch to form the distal radioulnar (DRU) joint. Restoring the relationships of the DRU joint, the triangular fibrocartilage complex, and the ulnar variance is of paramount importance.1,8,9

Classical teaching calls for restoration of radial inclination to about 23°, volar tilt to 11° to 12°, and radial length to about 11 mm. Especially regarding volar tilt and radial length, however, cadaveric and clinical studies have found more variance, leading to use of the contralateral extremity as an operative template, particularly when closed reduction thought to be adequate deviates significantly from these parameters.1,4,7

DRF classification based on these principles has led to abundant representation in the literature.10-13 Many authors have focused on fracture lines, comminution degree, articular surface violation, and other anatomical or radiographic characteristics of DRF classification and operative fixation approach.10-13 In 2001, Fernandez9 proposed a classification system focused on energy or mechanism of injury. In comparisons,14 the Fernandez system had the highest interobserver reliability—higher than that of AO (Arbeitsgemeinschaft für Osteosynthesefragen).

Considerations for Operative Treatment: Column Theory

In the restoration of anatomical alignment in complex DRFs, it is important to consider the 3 joints and the 3 columns—radial, intermediate, and ulnar (Figure 1). [[{"fid":"201864","view_mode":"medstat_image_flush_left","attributes":{"class":"media-element file-medstat-image-flush-left","data-delta":"1"},"fields":{"format":"medstat_image_flush_left","field_file_image_caption[und][0][value]":"Figure 1.","field_file_image_credit[und][0][value]":"","field_file_image_caption[und][0][format]":"plain_text","field_file_image_credit[und][0][format]":"plain_text"},"type":"media","field_deltas":{"1":{"format":"medstat_image_flush_left","field_file_image_caption[und][0][value]":"Figure 1.","field_file_image_credit[und][0][value]":""}}}]]In addition, parallels between the distal radius and the tibial plateau can be considered because of similarities in operative goals. Restoration of mechanical axis, length, alignment, rotation, and articular surfaces is paramount.15 Considering multiple surgical approaches to address "bicolumnar injuries" and reconstructing the "simpler" columnar injury first are common principles.16

The goals of fracture fixation at the wrist are the same as at any other joint: anatomical reduction, stable fixation, and early range of motion (ROM). Column restoration can result in consistent achievement of those goals. Intuitively, there is a close correlation between anatomical alignment and functional results.17 Rebuilding the structural foundation of the columns with respect to buttressing and restoring the 3 radial articulations with the ulna, scaphoid, and lunate can consistently yield restoration of length, inclination, and tilt (Figure 2). [[{"fid":"201865","view_mode":"medstat_image_flush_right","attributes":{"class":"media-element file-medstat-image-flush-right","data-delta":"2"},"fields":{"format":"medstat_image_flush_right","field_file_image_caption[und][0][value]":"Figure 2.","field_file_image_credit[und][0][value]":"","field_file_image_caption[und][0][format]":"plain_text","field_file_image_credit[und][0][format]":"plain_text"},"type":"media","field_deltas":{"2":{"format":"medstat_image_flush_right","field_file_image_caption[und][0][value]":"Figure 2.","field_file_image_credit[und][0][value]":""}}}]]Next, we discuss the options available and how to use each to an advantage, individually or in hybrid constructs.

External Fixation: Is There Still a Role?

In the setting of highly comminuted, complex fractures, external fixation with Kirschner wires (K-wires) is a reasonable choice, with restoration of motion and strength within 75% to 80% of the uninjured wrist.18 In a 2-year study of 113 patients with comminuted metaphyseal DRFs randomly assigned to either external fixation or casting, Kreder and colleagues19 found a trend toward better clinical, functional, and radiographic outcomes with external fixation with or without K-wire fixation. There was improved restoration of radial length and palmar tilt with external fixation. A study of unstable DRF in patients with osteoporosis found that redisplacement was more common after treatment with a cast than after treatment with an external fixator.20 Although closed reduction and casting continue to have a role in the treatment of DRF, Kreder and colleagues19 found that remanipulation was necessary in at least 9% of cases. According to a meta-analysis21 of the literature on DRF treatment, 4 articles directly address the question of the superiority of external fixation over closed reduction and casting, and 3 of the 4 found more favorable radiographic and functional outcomes with external fixation.

External fixation is useful in treating complex DRFs with metaphyseal comminution. It can also be effective in the presence of simple articular involvement without depression of the joint surface. External fixation devices can span areas of soft-
tissue injury and are useful as manipulation tools in achieving anatomical reduction. Although external fixation is effective, its complications include pin-tract infection, nerve injury, loss of reduction, and loss of digital ROM. In a meta-analysis, Li-hai and colleagues22 found that external fixators had a complication rate of 30.9%. With this technique, it is important to avoid midcarpal distraction, excessive ulnar deviation, and excessive palmar flexion. Papadonikolakis and colleagues23 found that dis
traction of as little as 2 mm to 5 mm significantly affected the function of the flexor digitorum superficialis at the metacarpophalangeal joint. Over-distraction in wrist flexion can lead to lengthening of the extensor tendons and loss of full digital ROM. Excessive flexion and ulnar deviation can lead to median nerve compression and associated symptoms, as well as poor extensor and radial tendon length. In addition, prolonged distraction in excessive flexion combined with swelling and inflammation during fracture healing causes digital stiffness and contracture.23 Biomechanical studies have found that proximal pin placement in the radius, along with distal pin fixation in 6 metacarpal cortices through the second and third metacarpals, helps provide the strongest fixation.24

As for technique, pins are placed in the second metacarpal and radial shaft. With respect to the radius, the incision is made just proximal to the edge of the abductor pollicis longus muscle in the "bare area." Ideal pin placement is between the extensor carpi radialis longus and the extensor carpi radialis brevis, with care taken to avoid the radial sensory nerve, which lies between the extensor carpi radialis longus and the brachialis and emerges 9 cm proximal to the radial styloid.25 Next, a 2.5-cm to 3-cm incision is made over the palpable edge of the index metacarpal near the base. During drilling, the guide is placed at intersecting 45° angles, and the distal pin is placed 2 cm to 3 cm from the proximal pin. The proximal metacarpal pin is placed at the base of the metacarpal. The second metacarpal pin can also be placed first, with the external fixator used to judge proximal placement of the radial pin within the bare area.

Various supplements to external fixation have positive outcomes. Wolfe and colleagues18 found that using K-wires with the external fixation construct added stability in flexion/extension, radial/ulnar deviation, and rotational motion. They noted that fixation stability may depend more on the augmentation to fixation than on the external fixator itself. In a prospective, randomized trial, Moroni and colleagues26,27 found that, compared with standard pins, hydroxyapatite-coated pins had higher extraction torque, which was associated with improved fixation. When combined with external fixation, calcium phosphate cement also provided additional stability, allowing the bone filler to help maintain articular reduction and cortical continuity.28,29

External fixation has its disadvantages and complications. It can be bulky, and theoretically it contributes to higher rates of stiffness in the wrist and fingers.30-32 Higher rates of pin-site infection have been reported, along with hardware failure and associated loss of reduction, in patients treated with external fixation (Figures 3A-3C).31-33[[{"fid":"201866","view_mode":"medstat_image_flush_left","attributes":{"class":"media-element file-medstat-image-flush-left","data-delta":"3"},"fields":{"format":"medstat_image_flush_left","field_file_image_caption[und][0][value]":"Figure 3.","field_file_image_credit[und][0][value]":"","field_file_image_caption[und][0][format]":"plain_text","field_file_image_credit[und][0][format]":"plain_text"},"type":"media","field_deltas":{"3":{"format":"medstat_image_flush_left","field_file_image_caption[und][0][value]":"Figure 3.","field_file_image_credit[und][0][value]":""}}}]]In addition, joint overdistraction can adversely affect the length-tension curve and contribute to potential reflex sympathetic dystrophy, which can be devastating (Figures 4A, 4B).1,21,31,33 Despite these complications, external fixation remains a powerful tool in the treatment of high-energy DRFs. [[{"fid":"201867","view_mode":"medstat_image_flush_right","attributes":{"class":"media-element file-medstat-image-flush-right","data-delta":"4"},"fields":{"format":"medstat_image_flush_right","field_file_image_caption[und][0][value]":"Figure 4.","field_file_image_credit[und][0][value]":"","field_file_image_caption[und][0][format]":"plain_text","field_file_image_credit[und][0][format]":"plain_text"},"type":"media","field_deltas":{"4":{"format":"medstat_image_flush_right","field_file_image_caption[und][0][value]":"Figure 4.","field_file_image_credit[und][0][value]":""}}}]]In many cases, authors who compared open reduction and internal fixation (ORIF) with external fixation found no significant differences in outcome scores or function.31-34 In a meta-analysis of 917 patients, Margaliot and colleagues33 found no differences in pain, grip strength, wrist ROM, or radiographic parameters. More recently, in prospective randomized trials, both Egol and colleagues31 and Grewal and colleagues34 compared hybrid external fixation with ORIF, and, though early outcomes favored ORIF, 1-year follow-up comparisons were even, and there were no significant differences. These consistently reproducible results reaffirm keeping external fixation in the orthopedic toolbox.

Definitive Reconstruction With ORIF

Early nonlocked dorsal plating options for DRF fixation had unacceptable rates of plate failure, poor cosmesis, and extensor tendon complications.17,35-37 Subsequent technologic advances—multiple approaches, lower profile plating, and rigid, fragment-specific fixation—have allowed even the most complex fracture patterns to be addressed (Table). In malunited fractures, bone graft may not be required if the fracture is extra-articular and treated with a volar locking plate. [[{"fid":"201868","view_mode":"medstat_image_flush_left","attributes":{"class":"media-element file-medstat-image-flush-left","data-delta":"5"},"fields":{"format":"medstat_image_flush_left","field_file_image_caption[und][0][value]":"Table.","field_file_image_credit[und][0][value]":"","field_file_image_caption[und][0][format]":"plain_text","field_file_image_credit[und][0][format]":"plain_text"},"type":"media","field_deltas":{"5":{"format":"medstat_image_flush_left","field_file_image_caption[und][0][value]":"Table.","field_file_image_credit[und][0][value]":""}}}]]Other options include corticocancellous autograft from the iliac crest, hydroxyapatite synthetic grafts, and osteoconductive bone graft substitutes, such as bone morphogenic proteins. In addition, healing times are similar in cases, regardless of whether a graft was used.38

Involvement of the radial and intermediate columns should be addressed first. Although some may prefer a single volar plate, others may use fragment-specific fixation to buttress a comminuted radial styloid (in orthogonal fashion) and/or a dorsal ulnar fragment to restore the intermediate column and thereby fully restore the radial articular surface.39,40 Typically, restoring the radial and intermediate columns for radial articular reduction subsequently and simultaneously restores the majority of radial height and length. After the radial and intermediate columns are reduced and stabilized, the need for ulna column fixation can be determined. Important factors in ulna column restoration are severe osteoporosis and ulna head and/or neck comminution. Significant comminution throughout the metaphysis of both the radius and the ulna may also warrant stabilizing the ulna with internal fixation. Finally, any DRU joint instability noted on examination should also favor fixing the ulnar side.

Assessment of the distal ulna in these complex fractures goes beyond the involvement of an ulnar styloid fracture. Typically, fractures at the base of the ulnar styloid have been reported to have little clinical relevance, including a low incidence of associated DRU joint problems.41-43 Decisions to address the ulnar column are largely swayed by any instability found on DRU joint testing, as laxity caused by severe comminution can dictate the need for distal ring fixation to provide support. Even in the presence of a high-energy fracture in severely osteoporotic bone, the argument can be made to prevent instability by supporting the ulnar column. Stabilization of the ulnar articular surface can also be made more facile by creating an easier "A" fracture pattern (per AO classification) from a complex "C" to further aid in achieving efficient anatomical reduction. After preoperative planning is completed, depending on which columns need to be addressed, several surgical approaches can be considered to achieve maximum exposure and soft-tissue mobilization in order to successfully complete the operative fixation goals.

Volar Approach

An approach is selected for ideal exposure of a facile environment for definitive fixation. Access to the radial column can be gained with the extended flexor carpi radialis (FCR) approach. This approach allows visualization and removal of the appropriate deforming forces on the radial column to allow for fracture reduction by "opening the book," similar to that of tibial plateau reconstruction.44,45 It may be prudent to perform a preincision Allen test as well as a preoperative DRU joint examination for comparison after ORIF is complete. Compared with the classic Henry approach near the distal radius, going through the volar sheath of the FCR avoids many of the perforating radial artery branches. Avoiding stripping the radial artery of its surrounding fat and lymphatics prevents postoperative "cold intolerance." Retracting the FCR ulnarly and then incising the dorsal FCR sheath provide ready access to the pronator quadratus after collective ulnar mobilization of both the FCR and the flexor pollicis longus.44 In addition, for work near the distal FCR sheath, care must be taken to avoid the branch of the palmar cutaneous nerve that emerges about 5 cm proximal to the wrist flexion crease.46

Once at the level of the pronator quadratus, an "L-shape" incision can be made to reflect the muscle off the radius. Care must be taken when working too distal to avoid transection of the inserting volar wrist ligaments.44 Leaving a cuff for repair of the pronator remains controversial. In a recent case-control series, however, Hershman and colleagues47 did not find significant differences in function or complication rate in patients with and without repair. After reflection, adequate exposure of the radial column should be achieved. Ready access to the radial styloid for orthogonal plating can be obtained by releasing the brachioradialis, which simultaneously releases one of the primary fracture deforming forces.44 With this incision and exposure, if needed, dorsal bone grafting can be achieved from the volar side; however, care must be taken to protect the first dorsal compartment.48 The cutaneous branch of the median nerve may be at risk with this exposure, but avoiding dissection ulnar to the FCR tendon can help to reduce this risk.49

Before surgery, if the fracture pattern dictates a more ulnar approach, we prefer the extended carpal tunnel approach. Using the plane between the palmaris longus and the flexor digitorum superficialis medially and the FCR laterally, the extended carpal tunnel approach provides an obvious release of the flexor retinaculum but, more important, allows for extensile access to the sigmoid notch, the DRU joint, and the ulnar column.

Dorsal Approach

The dorsal approach is necessary in a few select cases. With a focus on fragment-specific fixation, presence of a significant dorsal ulnar fragment should warrant a dorsal approach.50 In addition, in select, rare cases in which volar access is limited or unavailable, dorsal access is the only option.50 Finally, if direct articular visualization is required, the dorsal approach typically is favored as the stronger radiocarpal ligaments found on the volar side are maintained.

Access should begin with an incision centered over the dorsal distal radius; a safe access point is just ulnar to the Lister tubercle. On incision of the retinaculum through a full-thickness excision, the third dorsal compartment is opened and the extensor pollicis longus (EPL) mobilized, fully exposing the dorsal distal radius. Work can be performed on either side of the EPL between the second and fourth dorsal compartments. Exposure typically is not an issue because of the pliable soft tissue of the dorsum, with ready access from styloid to styloid.44 Here, low-profile plates and/or mini-fragment-specific plate options should be used to minimize potential tendon damage.51 Care must also be taken to avoid damaging the radiocarpal or scapholunate ligaments.49 On closure, the retinaculum is repaired primarily; however, though some proponents advocate relocating the EPL tendon into its groove, we prefer leaving the EPL free within the surrounding soft tissue to reduce tension and promote unhindered excursion. The dorsal approach, though controversial and used inconsistently, should remain an important tool in anatomical restoration, especially in cases of complex fracture patterns.

Conclusion

Controversy still marks the lack of consensus on deciding which DRF treatment is optimal. Some investigators question moving away from external fixation and cite the lack of significantly better data relative to ORIF.21,52 The same proponents note that the only advantage over external fixation is earlier return to function and cite reports of tendon rupture and complications with both dorsal and volar fixation options.34,53-58 Other investigators find that operative treatment generally does not provide a significant improvement over nonoperative treatment.59

With the advent of lower profile locked plating, fragment-specific fixation, and variable-angle devices, comparative clinical trials are finding it difficult to keep up.60-64 Results from ongoing prospective randomized trials like ORCHID (Open Reduction and Internal Fixation Versus Casting for Highly Comminuted Intra-Articular Fractures of the Distal Radius; 500 patients >65 years old, 15 centers) will provide more definitive answers about ideal treatment.65

Anatomical restoration involves a versatile array of fragment fixation and reconstruction. Careful preoperative planning and a consistent approach to restoring the radial, intermediate, and ulnar columns, along with a proper surgical approach, are ideal. Many advances in internal fixation have been exceedingly helpful. Use of external fixation, especially in a bridging fashion with or without supplementation, is still valuable in many situations.

References

1. Liporace FA, Adams MR, Capo JT, Koval KJ. Distal radius fractures. J Orthop Trauma. 2009;23(10):739-748.

2. Lee YS, Wei TY, Cheng YC, Hsu TL, Huang CR. A comparative study of Colles’ fractures in patients between fifty and seventy years of age: percutaneous K-wiring versus volar locking plating. Int Orthop. 2012;36(4):789-794.

3. Diaz-Garcia RJ, Oda T, Shauver MJ, Chung KC. A systematic review of outcomes and complications of treating unstable distal radius fractures in the elderly. J Hand Surg Am. 2011;36(5):824-835.e2.

4. Ring D. Treatment of the neglected distal radius fracture. Clin Orthop Relat Res. 2005;(431):85-92.

5. Berger RA. Arthroscopic anatomy of the wrist and distal radioulnar joint. Hand Clin. 1999;15(3):393-413, vii.

6. Berger RA. The anatomy of the ligaments of the wrist and distal radioulnar joints. Clin Orthop Relat Res. 2001;(383):32-40.

7. McCann PA, Clarke D, Amirfeyz R, Bhatia R. The cadaveric anatomy of the distal radius: implications for the use of volar plates. Ann R Coll Surg Engl. 2012;94(2):116-120.

8. Ekenstam F. Osseous anatomy and articular relationships about the distal ulna. Hand Clin. 1998;14(2):161-164.

9. Fernandez DL. Distal radius fracture: the rationale of a classification. Chir Main. 2001;20(6):411-425.

10. Raskin KB, Melone CP Jr. Unstable articular fractures of the distal radius. Comparative techniques of ligamentotaxis. Orthop Clin North Am. 1993;24(2):275-286.

11. Melone CP Jr. Distal radius fractures: patterns of articular fragmentation. Orthop Clin North Am. 1993;24(2):239-253.

12. Jenkins NH. The unstable Colles’ fracture. J Hand Surg Br. 1989;14(2):149-154.

13. Cooney WP, Dobyns JH, Linscheid RL. Arthroscopy of the wrist: anatomy and classification of carpal instability. Arthroscopy. 1990;6(2):133-140.

14. Kural C, Sungur I, Kaya I, Ugras A, Ertürk A, Cetinus E. Evaluation of the reliability of classification systems used for distal radius fractures. Orthopedics. 2010;33(11):801.

15. Lipton HA, Wollstein R. Operative treatment of intraarticular distal radial fractures. Clin Orthop Relat Res. 1996;(327):110-124.

16. Wolfe SW. Distal radius fractures. Green’s Operative Hand Surgery. 6th ed. Philadelphia, PA: Churchill Livingstone; 2011:561-638.

17. Rikli DA, Regazzoni P. Fractures of the distal end of the radius treated by internal fixation and early function. A preliminary report of 20 cases. J Bone Joint Surg Br. 1996;78(4):
588-592.

18. Wolfe SW, Austin G, Lorenze M, Swigart CR, Panjabi MM. A biomechanical comparison of different wrist external fixators with and without K-wire augmentation. J Hand Surg Am. 1999;24(3):516-524.

19. Kreder HJ, Agel J, McKee MD, Schemitsch EH, Stephen D, Hanel DP. A randomized, controlled trial of distal radius fractures with metaphyseal displacement but without joint incongruity: closed reduction and casting versus closed reduction, spanning external fixation, and optional percutaneous K-wires. J Orthop Trauma. 2006;20(2):115-121.

20. Moroni A, Vannini F, Faldini C, Pegreffi F, Giannini S. Cast vs external fixation: a comparative study in elderly osteoporotic distal radial fracture patients. Scand J Surg. 2004;93(1):64-67.

21. Paksima N, Panchal A, Posner MA, Green SM, Mehiman CT, Hiebert R. A meta-analysis of the literature on distal radius fractures: review of 615 articles. Bull Hosp Jt Dis. 2004;62(1-2):40-46.

22. Li-hai Z, Ya-nan W, Zhi M, et al. Volar locking plate versus external fixation for the treatment of unstable distal radial fractures: a meta-analysis of randomized controlled trials.
J Surg Res. 2015;193(1):324-333.

23. Papadonikolakis A, Shen J, Garrett JP, Davis SM, Ruch DS. The effect of increasing distraction on digital motion after external fixation of the wrist. J Hand Surg Am. 2005;30(4):
773-779.

24. Seitz WH Jr, Froimson AI, Brooks DB, et al. Biomechanical analysis of pin placement and pin size for external fixation of distal radius fractures. Clin Orthop Relat Res. 1990;(251):
207-212.

25. Beldner S, Zlotolow DA, Melone CP Jr, Agnes AM, Jones MH. Anatomy of the lateral antebrachial cutaneous and superficial radial nerves in the forearm: a cadaveric and clinical study. J Hand Surg Am. 2005;30(6):1226-1230.

26. Moroni A, Faldini C, Marchetti S, Manca M, Consoli V, Giannini S. Improvement of the bone-pin interface strength in osteoporotic bone with use of hydroxyapatite-coated tapered external-fixation pins. A prospective, randomized clinical study of wrist fractures. J Bone Joint Surg Am. 2001;83(5):717-721.

27. Moroni A, Heikkila J, Magyar G, Toksvig-Larsen S, Giannini S. Fixation strength and pin tract infection of hydroxyapatite-coated tapered pins. Clin Orthop Relat Res. 2001;(388):209-217.

28. Higgins TF, Dodds SD, Wolfe SW. A biomechanical analysis of fixation of intra-articular distal radial fractures with calcium-phosphate bone cement. J Bone Joint Surg Am. 2002;84(9):1579-1586.

29. Tobe M, Mizutani K, Tsubuku Y. Treatment of distal radius fracture with the use of calcium phosphate bone cement as a filler. Tech Hand Up Extrem Surg. 2004;8(2):95-101.

30. Capo JT, Rossy W, Henry P, Maurer RJ, Naidu S, Chen L.
External fixation of distal radius fractures: effect of distraction and duration. J Hand Surg Am. 2009;34(9):1605-1611.

31. Egol K, Walsh M, Tejwani N, McLaurin T, Wynn C, Paksima N. Bridging external fixation and supplementary Kirschner-wire fixation versus volar locked plating for unstable fractures of the distal radius: a randomised, prospective trial. J Bone Joint Surg Br. 2008;90(9):1214-1221.

32. Egol KA, Paksima N, Puopolo S, Klugman J, Hiebert R, Koval KJ. Treatment of external fixation pins about the wrist: a prospective, randomized trial. J Bone Joint Surg Am. 2006;88(2):349-354.

33. Margaliot Z, Haase SC, Kotsis SV, Kim HM, Chung KC. A meta-analysis of outcomes of external fixation versus plate osteosynthesis for unstable distal radius fractures. J Hand Surg Am. 2005;30(6):1185-1199.

34. Grewal R, MacDermid JC, King GJ, Faber KJ. Open reduction internal fixation versus percutaneous pinning with external fixation of distal radius fractures: a prospective, randomized clinical trial. J Hand Surg Am. 2011;36(12):
1899-1906.

35. Axelrod TS, McMurtry RY. Open reduction and internal fixation of comminuted, intraarticular fractures of the distal radius. J Hand Surg Am. 1990;15(1):1-11.

36. Hove LM, Nilsen PT, Furnes O, Oulie HE, Solheim E, Mölster AO. Open reduction and internal fixation of displaced intraarticular fractures of the distal radius. 31 patients followed for 3-7 years. Acta Orthop Scand. 1997;68(1):59-63.

37. Carter PR, Frederick HA, Laseter GF. Open reduction and internal fixation of unstable distal radius fractures with a low-profile plate: a multicenter study of 73 fractures. J Hand Surg Am. 1998;23(2):300-307.

38. Mugnai R, Tarallo L, Lancellotti E, et al. Corrective osteotomies of the radius: grafting or not? World J Orthop. 2016;7(2):128-135.

39. Tang P, Ding A, Uzumcugil A. Radial column and volar plating (RCVP) for distal radius fractures with a radial styloid component or severe comminution. Tech Hand Up Extrem Surg. 2010;14(3):143-149.

40. Helmerhorst GT, Kloen P. Orthogonal plating of intra-articular distal radius fractures with an associated radial column fracture via a single volar approach. Injury. 2012;43(8):1307-1312.

41. May MM, Lawton JN, Blazar PE. Ulnar styloid fractures associated with distal radius fractures: incidence and implications for distal radioulnar joint instability. J Hand Surg Am. 2002;27(6):965-971.

42. Souer JS, Ring D, Matschke S, Audige L, Marent-Huber M, Jupiter JB; AOCID Prospective ORIF Distal Radius Study Group. Effect of an unrepaired fracture of the ulnar styloid base on outcome after plate-and-screw fixation of a distal radial fracture. J Bone Joint Surg Am. 2009;91(4):830-838.

43. Noda K, Goto A, Murase T, Sugamoto K, Yoshikawa H, Moritomo H. Interosseous membrane of the forearm: an anatomical study of ligament attachment locations. J Hand Surg Am. 2009;34(3):415-422.

44. Catalano LW 3rd, Zlotolow DA, Hitchcock PB, Shah SN, Barron OA. Surgical exposures of the radius and ulna. J Am Acad Orthop Surg. 2011;19(7):430-438.

45. Orbay JL, Badia A, Indriago IR, et al. The extended flexor carpi radialis approach: a new perspective for the distal radius fracture. Tech Hand Up Extrem Surg. 2001;5(4):204-211.

46. Hobbs RA, Magnussen PA, Tonkin MA. Palmar cutaneous branch of the median nerve. J Hand Surg Am. 1990;15(1):38-43.

47. Hershman SH, Immerman I, Bechtel C, Lekic N, Paksima N, Egol KA. The effects of pronator quadratus repair on outcomes after volar plating of distal radius fractures. J Orthop Trauma. 2013;27(3):130-133.

48. Prommersberger KJ, Lanz UB. Corrective osteotomy of the distal radius through volar approach. Tech Hand Up Extrem Surg. 2004;8(2):70-77.

49. Ilyas AM. Surgical approaches to the distal radius. Hand (N Y). 2011;6(1):8-17.

50. Tavakolian JD, Jupiter JB. Dorsal plating for distal radius fractures. Hand Clin. 2005;21(3):341-346.

51. Yu YR, Makhni MC, Tabrizi S, Rozental TD, Mundanthanam G, Day CS. Complications of low-profile dorsal versus volar locking plates in the distal radius: a comparative study. J Hand Surg Am. 2011;36(7):1135-1141.

52. Mattila VM, Huttunen TT, Sillanpää P, Niemi S, Pihlajamäki H, Kannus P. Significant change in the surgical treatment of distal radius fractures: a nationwide study between 1998 and 2008 in Finland. J Trauma. 2011;71(4):939-942.

53. Wilcke MK, Abbaszadegan H, Adolphson PY. Wrist function recovers more rapidly after volar locked plating than after external fixation but the outcomes are similar after 1 year. Acta Orthop. 2011;82(1):76-81.

54. Ward CM, Kuhl TL, Adams BD. Early complications of volar plating of distal radius fractures and their relationship to surgeon experience. Hand (N Y). 2011;6(2):185-189.

55. Soong M, van Leerdam R, Guitton TG, Got C, Katarincic J, Ring D. Fracture of the distal radius: risk factors for complications after locked volar plate fixation. J Hand Surg Am. 2011;36(1):3-9.

56. Soong M, Earp BE, Bishop G, Leung A, Blazar P. Volar locking plate implant prominence and flexor tendon rupture. J Bone Joint Surg Am. 2011;93(4):328-335.

57. Jeudy J, Steiger V, Boyer P, Cronier P, Bizot P, Massin P. Treatment of complex fractures of the distal radius: a prospective randomised comparison of external fixation ‘versus’ locked volar plating. Injury. 2012;43(2):174-179.

58. Berglund LM, Messer TM. Complications of volar plate fixation for managing distal radius fractures. J Am Acad Orthop Surg. 2009;17(6):369-377.

59. Egol KA, Walsh M, Romo-Cardoso S, Dorsky S, Paksima N. Distal radial fractures in the elderly: operative compared with nonoperative treatment. J Bone Joint Surg Am. 2010;92(9):1851-1857.

60. Wall LB, Brodt MD, Silva MJ, Boyer MI, Calfee RP. The effects of screw length on stability of simulated osteoporotic distal radius fractures fixed with volar locking plates. J Hand Surg Am. 2012;37(3):446-453.

61. Dahl WJ, Nassab PF, Burgess KM, et al. Biomechanical properties of fixed-angle volar distal radius plates under dynamic loading. J Hand Surg Am. 2012;37(7):1381-1387.

62. Park JH, Hagopian J, Ilyas AM. Variable-angle locking screw volar plating of distal radius fractures. Hand Clin. 2010;26(3):373-380, vi.

63. Pensy RA, Brunton LM, Parks BG, Higgins JP, Chhabra AB. Single-incision extensile volar approach to the distal radius and concurrent carpal tunnel release: cadaveric study. J Hand Surg Am. 2010;35(2):217-222.

64. Klos K, Rausch S, Löffler M, et al. A biomechanical comparison of a biodegradable volar locked plate with two titanium volar locked plates in a distal radius fracture model. J Trauma. 2010;68(4):984-991.

65. Bartl C, Stengel D, Bruckner T, et al. Open reduction and internal fixation versus casting for highly comminuted and intra-articular fractures of the distal radius (ORCHID): protocol for a randomized clinical multi-center trial. Trials. 2011;12:84

References

1. Liporace FA, Adams MR, Capo JT, Koval KJ. Distal radius fractures. J Orthop Trauma. 2009;23(10):739-748.

2. Lee YS, Wei TY, Cheng YC, Hsu TL, Huang CR. A comparative study of Colles’ fractures in patients between fifty and seventy years of age: percutaneous K-wiring versus volar locking plating. Int Orthop. 2012;36(4):789-794.

3. Diaz-Garcia RJ, Oda T, Shauver MJ, Chung KC. A systematic review of outcomes and complications of treating unstable distal radius fractures in the elderly. J Hand Surg Am. 2011;36(5):824-835.e2.

4. Ring D. Treatment of the neglected distal radius fracture. Clin Orthop Relat Res. 2005;(431):85-92.

5. Berger RA. Arthroscopic anatomy of the wrist and distal radioulnar joint. Hand Clin. 1999;15(3):393-413, vii.

6. Berger RA. The anatomy of the ligaments of the wrist and distal radioulnar joints. Clin Orthop Relat Res. 2001;(383):32-40.

7. McCann PA, Clarke D, Amirfeyz R, Bhatia R. The cadaveric anatomy of the distal radius: implications for the use of volar plates. Ann R Coll Surg Engl. 2012;94(2):116-120.

8. Ekenstam F. Osseous anatomy and articular relationships about the distal ulna. Hand Clin. 1998;14(2):161-164.

9. Fernandez DL. Distal radius fracture: the rationale of a classification. Chir Main. 2001;20(6):411-425.

10. Raskin KB, Melone CP Jr. Unstable articular fractures of the distal radius. Comparative techniques of ligamentotaxis. Orthop Clin North Am. 1993;24(2):275-286.

11. Melone CP Jr. Distal radius fractures: patterns of articular fragmentation. Orthop Clin North Am. 1993;24(2):239-253.

12. Jenkins NH. The unstable Colles’ fracture. J Hand Surg Br. 1989;14(2):149-154.

13. Cooney WP, Dobyns JH, Linscheid RL. Arthroscopy of the wrist: anatomy and classification of carpal instability. Arthroscopy. 1990;6(2):133-140.

14. Kural C, Sungur I, Kaya I, Ugras A, Ertürk A, Cetinus E. Evaluation of the reliability of classification systems used for distal radius fractures. Orthopedics. 2010;33(11):801.

15. Lipton HA, Wollstein R. Operative treatment of intraarticular distal radial fractures. Clin Orthop Relat Res. 1996;(327):110-124.

16. Wolfe SW. Distal radius fractures. Green’s Operative Hand Surgery. 6th ed. Philadelphia, PA: Churchill Livingstone; 2011:561-638.

17. Rikli DA, Regazzoni P. Fractures of the distal end of the radius treated by internal fixation and early function. A preliminary report of 20 cases. J Bone Joint Surg Br. 1996;78(4):
588-592.

18. Wolfe SW, Austin G, Lorenze M, Swigart CR, Panjabi MM. A biomechanical comparison of different wrist external fixators with and without K-wire augmentation. J Hand Surg Am. 1999;24(3):516-524.

19. Kreder HJ, Agel J, McKee MD, Schemitsch EH, Stephen D, Hanel DP. A randomized, controlled trial of distal radius fractures with metaphyseal displacement but without joint incongruity: closed reduction and casting versus closed reduction, spanning external fixation, and optional percutaneous K-wires. J Orthop Trauma. 2006;20(2):115-121.

20. Moroni A, Vannini F, Faldini C, Pegreffi F, Giannini S. Cast vs external fixation: a comparative study in elderly osteoporotic distal radial fracture patients. Scand J Surg. 2004;93(1):64-67.

21. Paksima N, Panchal A, Posner MA, Green SM, Mehiman CT, Hiebert R. A meta-analysis of the literature on distal radius fractures: review of 615 articles. Bull Hosp Jt Dis. 2004;62(1-2):40-46.

22. Li-hai Z, Ya-nan W, Zhi M, et al. Volar locking plate versus external fixation for the treatment of unstable distal radial fractures: a meta-analysis of randomized controlled trials.
J Surg Res. 2015;193(1):324-333.

23. Papadonikolakis A, Shen J, Garrett JP, Davis SM, Ruch DS. The effect of increasing distraction on digital motion after external fixation of the wrist. J Hand Surg Am. 2005;30(4):
773-779.

24. Seitz WH Jr, Froimson AI, Brooks DB, et al. Biomechanical analysis of pin placement and pin size for external fixation of distal radius fractures. Clin Orthop Relat Res. 1990;(251):
207-212.

25. Beldner S, Zlotolow DA, Melone CP Jr, Agnes AM, Jones MH. Anatomy of the lateral antebrachial cutaneous and superficial radial nerves in the forearm: a cadaveric and clinical study. J Hand Surg Am. 2005;30(6):1226-1230.

26. Moroni A, Faldini C, Marchetti S, Manca M, Consoli V, Giannini S. Improvement of the bone-pin interface strength in osteoporotic bone with use of hydroxyapatite-coated tapered external-fixation pins. A prospective, randomized clinical study of wrist fractures. J Bone Joint Surg Am. 2001;83(5):717-721.

27. Moroni A, Heikkila J, Magyar G, Toksvig-Larsen S, Giannini S. Fixation strength and pin tract infection of hydroxyapatite-coated tapered pins. Clin Orthop Relat Res. 2001;(388):209-217.

28. Higgins TF, Dodds SD, Wolfe SW. A biomechanical analysis of fixation of intra-articular distal radial fractures with calcium-phosphate bone cement. J Bone Joint Surg Am. 2002;84(9):1579-1586.

29. Tobe M, Mizutani K, Tsubuku Y. Treatment of distal radius fracture with the use of calcium phosphate bone cement as a filler. Tech Hand Up Extrem Surg. 2004;8(2):95-101.

30. Capo JT, Rossy W, Henry P, Maurer RJ, Naidu S, Chen L.
External fixation of distal radius fractures: effect of distraction and duration. J Hand Surg Am. 2009;34(9):1605-1611.

31. Egol K, Walsh M, Tejwani N, McLaurin T, Wynn C, Paksima N. Bridging external fixation and supplementary Kirschner-wire fixation versus volar locked plating for unstable fractures of the distal radius: a randomised, prospective trial. J Bone Joint Surg Br. 2008;90(9):1214-1221.

32. Egol KA, Paksima N, Puopolo S, Klugman J, Hiebert R, Koval KJ. Treatment of external fixation pins about the wrist: a prospective, randomized trial. J Bone Joint Surg Am. 2006;88(2):349-354.

33. Margaliot Z, Haase SC, Kotsis SV, Kim HM, Chung KC. A meta-analysis of outcomes of external fixation versus plate osteosynthesis for unstable distal radius fractures. J Hand Surg Am. 2005;30(6):1185-1199.

34. Grewal R, MacDermid JC, King GJ, Faber KJ. Open reduction internal fixation versus percutaneous pinning with external fixation of distal radius fractures: a prospective, randomized clinical trial. J Hand Surg Am. 2011;36(12):
1899-1906.

35. Axelrod TS, McMurtry RY. Open reduction and internal fixation of comminuted, intraarticular fractures of the distal radius. J Hand Surg Am. 1990;15(1):1-11.

36. Hove LM, Nilsen PT, Furnes O, Oulie HE, Solheim E, Mölster AO. Open reduction and internal fixation of displaced intraarticular fractures of the distal radius. 31 patients followed for 3-7 years. Acta Orthop Scand. 1997;68(1):59-63.

37. Carter PR, Frederick HA, Laseter GF. Open reduction and internal fixation of unstable distal radius fractures with a low-profile plate: a multicenter study of 73 fractures. J Hand Surg Am. 1998;23(2):300-307.

38. Mugnai R, Tarallo L, Lancellotti E, et al. Corrective osteotomies of the radius: grafting or not? World J Orthop. 2016;7(2):128-135.

39. Tang P, Ding A, Uzumcugil A. Radial column and volar plating (RCVP) for distal radius fractures with a radial styloid component or severe comminution. Tech Hand Up Extrem Surg. 2010;14(3):143-149.

40. Helmerhorst GT, Kloen P. Orthogonal plating of intra-articular distal radius fractures with an associated radial column fracture via a single volar approach. Injury. 2012;43(8):1307-1312.

41. May MM, Lawton JN, Blazar PE. Ulnar styloid fractures associated with distal radius fractures: incidence and implications for distal radioulnar joint instability. J Hand Surg Am. 2002;27(6):965-971.

42. Souer JS, Ring D, Matschke S, Audige L, Marent-Huber M, Jupiter JB; AOCID Prospective ORIF Distal Radius Study Group. Effect of an unrepaired fracture of the ulnar styloid base on outcome after plate-and-screw fixation of a distal radial fracture. J Bone Joint Surg Am. 2009;91(4):830-838.

43. Noda K, Goto A, Murase T, Sugamoto K, Yoshikawa H, Moritomo H. Interosseous membrane of the forearm: an anatomical study of ligament attachment locations. J Hand Surg Am. 2009;34(3):415-422.

44. Catalano LW 3rd, Zlotolow DA, Hitchcock PB, Shah SN, Barron OA. Surgical exposures of the radius and ulna. J Am Acad Orthop Surg. 2011;19(7):430-438.

45. Orbay JL, Badia A, Indriago IR, et al. The extended flexor carpi radialis approach: a new perspective for the distal radius fracture. Tech Hand Up Extrem Surg. 2001;5(4):204-211.

46. Hobbs RA, Magnussen PA, Tonkin MA. Palmar cutaneous branch of the median nerve. J Hand Surg Am. 1990;15(1):38-43.

47. Hershman SH, Immerman I, Bechtel C, Lekic N, Paksima N, Egol KA. The effects of pronator quadratus repair on outcomes after volar plating of distal radius fractures. J Orthop Trauma. 2013;27(3):130-133.

48. Prommersberger KJ, Lanz UB. Corrective osteotomy of the distal radius through volar approach. Tech Hand Up Extrem Surg. 2004;8(2):70-77.

49. Ilyas AM. Surgical approaches to the distal radius. Hand (N Y). 2011;6(1):8-17.

50. Tavakolian JD, Jupiter JB. Dorsal plating for distal radius fractures. Hand Clin. 2005;21(3):341-346.

51. Yu YR, Makhni MC, Tabrizi S, Rozental TD, Mundanthanam G, Day CS. Complications of low-profile dorsal versus volar locking plates in the distal radius: a comparative study. J Hand Surg Am. 2011;36(7):1135-1141.

52. Mattila VM, Huttunen TT, Sillanpää P, Niemi S, Pihlajamäki H, Kannus P. Significant change in the surgical treatment of distal radius fractures: a nationwide study between 1998 and 2008 in Finland. J Trauma. 2011;71(4):939-942.

53. Wilcke MK, Abbaszadegan H, Adolphson PY. Wrist function recovers more rapidly after volar locked plating than after external fixation but the outcomes are similar after 1 year. Acta Orthop. 2011;82(1):76-81.

54. Ward CM, Kuhl TL, Adams BD. Early complications of volar plating of distal radius fractures and their relationship to surgeon experience. Hand (N Y). 2011;6(2):185-189.

55. Soong M, van Leerdam R, Guitton TG, Got C, Katarincic J, Ring D. Fracture of the distal radius: risk factors for complications after locked volar plate fixation. J Hand Surg Am. 2011;36(1):3-9.

56. Soong M, Earp BE, Bishop G, Leung A, Blazar P. Volar locking plate implant prominence and flexor tendon rupture. J Bone Joint Surg Am. 2011;93(4):328-335.

57. Jeudy J, Steiger V, Boyer P, Cronier P, Bizot P, Massin P. Treatment of complex fractures of the distal radius: a prospective randomised comparison of external fixation ‘versus’ locked volar plating. Injury. 2012;43(2):174-179.

58. Berglund LM, Messer TM. Complications of volar plate fixation for managing distal radius fractures. J Am Acad Orthop Surg. 2009;17(6):369-377.

59. Egol KA, Walsh M, Romo-Cardoso S, Dorsky S, Paksima N. Distal radial fractures in the elderly: operative compared with nonoperative treatment. J Bone Joint Surg Am. 2010;92(9):1851-1857.

60. Wall LB, Brodt MD, Silva MJ, Boyer MI, Calfee RP. The effects of screw length on stability of simulated osteoporotic distal radius fractures fixed with volar locking plates. J Hand Surg Am. 2012;37(3):446-453.

61. Dahl WJ, Nassab PF, Burgess KM, et al. Biomechanical properties of fixed-angle volar distal radius plates under dynamic loading. J Hand Surg Am. 2012;37(7):1381-1387.

62. Park JH, Hagopian J, Ilyas AM. Variable-angle locking screw volar plating of distal radius fractures. Hand Clin. 2010;26(3):373-380, vi.

63. Pensy RA, Brunton LM, Parks BG, Higgins JP, Chhabra AB. Single-incision extensile volar approach to the distal radius and concurrent carpal tunnel release: cadaveric study. J Hand Surg Am. 2010;35(2):217-222.

64. Klos K, Rausch S, Löffler M, et al. A biomechanical comparison of a biodegradable volar locked plate with two titanium volar locked plates in a distal radius fracture model. J Trauma. 2010;68(4):984-991.

65. Bartl C, Stengel D, Bruckner T, et al. Open reduction and internal fixation versus casting for highly comminuted and intra-articular fractures of the distal radius (ORCHID): protocol for a randomized clinical multi-center trial. Trials. 2011;12:84

Issue
The American Journal of Orthopedics - 46(5)
Issue
The American Journal of Orthopedics - 46(5)
Page Number
238-244
Page Number
238-244
Publications
Publications
Topics
Article Type
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Article PDF Media

Comparison of Lateral Retinaculum Release and Lengthening in the Treatment of Patellofemoral Disorders

Article Type
Changed
Thu, 09/19/2019 - 13:21

Take-Home Points

  • Understanding the indications for treatment is essential.
  • Identifying the superficial (oblique fibers) and deep layers (transverse fibers) of the LR is very important and can lengthen the LR by as much as 20 mm.
  • Open procedures reduce the risk of hematomas and related pain.
  • The goal is to obtain 1 or 2 patellar quadrants of medial and lateral patellar glide in extensino and a neutral patella.
  • If the Z-plasty is combined with the MPFL reconstruction or tibial tubercle transfer, the LR is set to length after the tubercle transfer and before the MPFL reconstruction (to avoid overconstraint).

Anterior knee pain is a common clinical problem that can be challenging to correct, in large part because of multiple causative factors, including structural/anatomical, functional, alignment, and neuroperception/pain pathway factors. One difficult aspect of anatomical assessment is judging the soft-tissue balance between the medial restraints (medial patellofemoral ligament [MPFL]; medial quadriceps tendon to femoral ligament; medial patellotibial and patellomeniscal ligaments) and the lateral restraints (lateral retinaculum [LR] specifically). Both LR tightness and patellar instability can be interpreted as anterior knee pain. Differentiating these entities is one of the most difficult clinical challenges in orthopedics.

LR release (LRR) has been found to improve patellar mobility and tracking.1 In the absence of clearly defined guidelines, the procedure quickly gained in popularity because of its technical simplicity and the enticing "one tool fits all" treatment approach suggested in early reviews. Injudicious use of LRR, alone or in combination with other procedures, led to iatrogenic instability and chronic pain. LR lengthening (LRL) was introduced to address LR tightness while maintaining lateral soft-tissue integrity and avoiding some of the severe complications of LRR.2

Today, isolated use of LRR/LRL is recommended only for treatment of LR tightness and pain secondary to lateral patellar hypercompression.3 It can also be used as an adjunct treatment in the setting of patellofemoral instability. LRR/LRL should never be used as primary treatment for patellofemoral instability.

In this review of treatments for LR tightness and patellofemoral disorders, we compare the use of LRR and LRL.

Discussion

LR procedures are indicated for LR tightness, which is assessed by taking a history, performing a physical examination, and obtaining diagnostic imaging. Decisions should be based on all findings considered together and never on imaging findings alone.

Physical Examination

The physical examination should include assessment of limb alignment, patellar mobility, muscle balance, and dynamic patellar tracking.

Limb Alignment. Abnormal valgus, rotational deformities, and increased Q-angle are associated with LR tightness. Valgus alignment can be assessed on standing inspection; rotational deformities with increased hip anteversion by hip motion with the patient in the prone position (increased hip internal rotation, decreased hip external rotation); and Q-angle on weight-bearing standing examination and with the patient flexing and extending the knee while seated.

Patellar Mobility. The patellar glide and tilt tests provide the most direct evaluations of LR tightness. Medial displacement of <1 quadrant is consistent with tightness, and displacement of >3 quadrants is consistent with laxity. In full extension, the patellar glide test evaluates only the soft-tissue restraints; at 30° flexion, it also evaluates patellofemoral engagement. The patellar tilt test measures the lifting of the lateral edge of the patella. With normal elevation being 0° to 20°, lack of patellar tilt means the LR is tight, and tilt of >20° means it is loose. MPFL patency can be examined with the Lachman test; the examiner rapidly moves the patella laterally while feeling for the characteristic hard endpoint of lateral translation.

Muscle Balance. The tone, strength, and tightness of the core (abdomen, dorsal, and hip muscles) and lower extremities (quadriceps, hamstrings, gastrocnemius) should be evaluated.

Dynamic Patellar Tracking. The J-sign is the course (shaped like an inverted J) that the patella takes when it is medialized into the trochlea from its laterally displaced resting position as the knee goes from full extension to flexion. The J-sign can be associated with LR tightness, trochlear dysplasia, and patella alta.

Imaging

Although we cannot provide a comprehensive review of the imaging literature, the following radiologic examinations should be used to assess the patellofemoral joint.

30° Lateral Radiograph.  Increased tilt is seen when the lateral facet is not anterior to the patellar ridge. Also evaluated are trochlear anatomy, patellar height, and other factors involved in patellofemoral disorders.

30° Flexed Axial (Merchant) Radiograph. Patellar tilt, subluxation, and trochlear dysplasia are evaluated. Images obtained with progressive flexion can be very useful in verifying patellar tilt reduction. Lack of reduction during early flexion suggests LR tightness.4

Alignment Axial Radiographs (Scanogram). Valgus alignment is assessed with this full-length, standing, long-leg examination.

Computed Tomography/Magnetic Resonance Imaging. Many parameters of patellar alignment have been described. Basic assessment should include evaluation of patellar tilt, angle by the line across posterior condyles and a line through the greatest patellar width (>20° indicates abnormality and LR tightness) and tibial tubercle-trochlear groove distance (computed tomography or magnetic resonance imaging scan of the knee is used to measure this distance, and to confirm a significant amount in light of complex patellofemoral malalignment5).

Indications

Lateral compression syndrome with LR tightness is often successfully treated with isolated LRR, and results are reproducible and predictable.6 Surgical intervention for patellofemoral pain should be undertaken only after failed extensive nonoperative treatment with physical therapy and bracing/taping. Patients with LR tightness on preoperative examination, lateral patellar tilt on imaging, and normal Q-angle can obtain satisfactory results with this procedure. Patellar subluxation or dislocation history, high Q-angle (>20°), grade 3 or 4 chondral injury, and patellofemoral arthritis are associated with poorer outcomes when the procedure is performed in isolation.6International Patellofemoral Study Group members agreed that LRR/LRL is a valid treatment option when indicated, but it is rarely performed in isolation and constitutes only 1% to 2% of surgeries performed by this group of experts.7 When lateral compression syndrome progresses to arthritis, LRR/LRL can be performed with lateral patella facetectomy for maximal improvement.4 In the setting of patellofemoral instability, LRR/LRL can be combined with proximal and/or distal realignment surgery if the LR is tight. The LR is the last line of defense limiting lateral translation in the setting of an incompetent MPFL. Isolated LRR/LRL in the setting of instability further destabilizes the patella and worsens the instability. Therefore, LRR/LRL
is a poor surgical option as an isolated procedure for this condition and should be used only as an adjunct in cases of patellofemoral instability with LR tightness that does not allow the patella to be centralized into the trochlea.8 LRR/LRL can also be performed to improve patellar tracking in patello­femoral arthroplasty and total knee arthroplasty.

Lateral Retinaculum Release Versus Lengthening

LRR was first described for the treatment of patellar instability in 1891.9 It was also used for the treatment of lateral patellar hypercompression syndrome associated with LR tightness that led to lateral patellar tracking, joint overload, degeneration, and anterior knee pain.10 Metcalf10 further popularized the procedure by describing a minimally invasive arthroscopic version. However, the arthroscopic technique is as aggressive as the open technique and may be performed with less control, potentially making its results more variable. As proximal and distal releases are performed from the "inside out," more capsule and muscle disruption is needed to release the more superficial layers.

Z-plasty lengthening of the LR was described as an alternative for maintaining lateral patellar soft-tissue integrity while reducing the tension of the lateral tissue restraints.3 This is our preferred method.

Performing LRL instead of LRR avoids iatrogenic medial patellar instability, avoids overrelease and muscle injury, and improves soft-tissue balance.3 Open release or lengthening reduces inadvertent injury to the lateral superior/inferior geniculate arteries and allows direct hemostasis. Two prospective randomized studies found functional knee outcomes and return to athletic activities were improved more after LRL than LRR.11,12 These procedures had similar rates of postoperative knee stiffness, decreased muscle mass, and decreased strength. Each prospective study used an extensive LRR technique for LRR cases (various authors have recommended performing the release until the patella is perpendicular to the trochlea), which may have affected outcomes. In any case, with lengthening, the surgeon is less likely to excessively disrupt the lateral tissues.

Lateral Retinaculum Release.  LRR can be openly performed by lateral parapatellar incision,1 a mini-open percutaneous technique, or arthroscopy. For these open techniques, incisions of various sizes have been used to access the LR and incise it about 1 cm lateral to the patella starting at the distal end of the vastus lateralis and extending distally until patellar tilt reduction is sufficient. If tightness in deep flexion persists, the LRR can be extended distally to the tibial tubercle. Open techniques have the advantage of sparing the joint capsule. All-arthroscopic techniques involve using electrocautery to cut through the capsule and access the LR.

Lateral Retinaculum Lengthening. 

The LR is sharply divided into a superficial layer of superficial oblique fibers from the anterior iliotibial band and a deep layer of transverse fibers from the femur. For LRR, these 2 layers must be identified separate from the articular capsule.13

Figure 1.
Figure 1 illustrates the anatomy and the essential steps of the procedure. The fascia lata is carefully removed over the lateral patella, releasing some lateral tension and helping in the identification of the superficial and deep layers. The proximal lateral soft tissues of the patella are exposed, and the outer oblique layer of the LR is incised near the lateral border of the patella (Figure 2).
Figure 2.
The incision begins around the level of the proximal patellar pole and extends distally to the inferior patellar pole. The superficial oblique fibers of the LR are sharply dissected from the deep transverse fibers and are elevated as dissection is carried posteriorly to the posterior-most extent of the retinacular envelope, usually 1 cm to 2 cm (Figure 3). T
Figure 3.
he deep transverse fibers are then incised longitudinally (Figure 4). In many cases, a capsule that adheres to the deep layer can be separated from it. In cases with combined tibial tubercle transfers, the capsule is incised in order to mobilize the patella medially or distally.
Figure 4.
Then, the knee is moved through its full range of motion, positioned in 30° flexion, and engaged into the trochlear groove. The length of both the MPFL and the LR can then be adjusted. The cut edges of the superior oblique and deep transverse fibers are then sutured together with absorbable suture, and the appropriate amount of lengthening is performed to remove excess tension in lateral structures while maintaining lateral soft-tissue integrity (Figure 5).
Figure 5.
Neither the MPFL nor the LR is tensioned; rather, their lengths are adjusted, as they act as checkreins guiding the patella. If this procedure is performed with tibial tubercle transfer (medial and/or distal) or MPFL reconstruction, tubercle fixation is done first; next, MPFL length and then LR length are set.

Complications

Complications of performing LRR/LRL to change the lateral restraint include medial patellar instability, increased lateral pain, repair failure, recurrent lateral instability, quadriceps weakness and atrophy, postoperative hemarthrosis, knee stiffness, wound complications, and thermal skin injury.7 These complications often result from poor surgical technique and too aggressive release. Although recommended patellar tilt historically has varied from 45° to 90°, the current goal is to normalize the tight soft-tissue restraints without creating secondary instability.

The most significant complication of LRR is medial patellar instability caused by muscle atrophy and loss of soft-tissue restraint.14 Medial instability can be difficult to diagnose and should be considered in any patient with patellofemoral pain, popping, or patellar instability after LRR.15 A positive medial subluxation test or medial patellar apprehension test suggests medial instability.

Medial patellar instability usually requires surgical treatment. Direct LR repair, lateral soft-tissue reconstruction, and other procedures can be used to restore lateral restraint.15 However, these are salvage techniques, and patients often remain significantly limited by pain or instability. Therefore, the LR must be carefully addressed and preferably should undergo lengthening rather than release.

References

1. Merchant AC, Mercer RL. Lateral release of the patella. A preliminary report. Clin Orthop Relat Res. 1974;(103):40-45.

2. Ceder LC, Larson RL. Z-plasty lateral retinacular release for the treatment of patellar compression syndrome. Clin Orthop Relat Res. 1979;(144):110-113.

3. Biedert R. Lateral patellar hypercompression, tilt and mild lateral subluxation. In: Biedert R, ed. Patellofemoral Disorders. Chichester, England: Wiley; 2004:161-166.

4. Hinckel BB, Arendt EA. Lateral retinaculum lengthening or release. Oper Tech Sports Med. 2015;23(2):100-106.

5. Seitlinger G, Scheurecker G, Högler R, Labey L, Innocenti B, Hofmann S. Tibial tubercle–posterior cruciate ligament distance: a new measurement to define the position of the tibial tubercle in patients with patellar dislocation. Am J Sports Med. 2012;40(5):1119-1125.

6. Lattermann C, Toth J, Bach BR Jr. The role of lateral retinacular release in the treatment of patellar instability. Sports Med Arthrosc. 2007;15(2):57-60.

7. Fithian DC, Paxton EW, Post WR, Panni AS; International Patellofemoral Study Group. Lateral retinacular release: a survey of the International Patellofemoral Study Group. Arthroscopy. 2004;20(5):463-468.

8. Christoforakis J, Bull AM, Strachan RK, Shymkiw R, Senavongse W, Amis AA. Effects of lateral retinacular release on the lateral stability of the patella. Knee Surg Sports Traumatol Arthrosc. 2006;14(3):273-277.

9. Pollard B. Old dislocation of patella by intra-articular operation. Lancet. 1891;(988):17-22.

10. Metcalf RW. An arthroscopic method for lateral release of subluxating or dislocating patella. Clin Orthop Relat Res. 1982;167:9-18.

11. Pagenstert G, Wolf N, Bachmann M, et al. Open lateral patellar retinacular lengthening versus open retinacular release in lateral patellar hypercompression syndrome: a prospective double-blinded comparative study on complications and outcome. Arthroscopy. 2012;28(6):788-797.

12. O’Neill DB. Open lateral retinacular lengthening compared with arthroscopic release. A prospective, randomized outcome study. J Bone Joint Surg Am. 1997;79(12):1759-1769.

13. Merican AM, Amis AA. Anatomy of the lateral retinaculum of the knee. J Bone Joint Surg Br. 2008;90(4):527-534.

14. Hughston JC, Deese M. Medial subluxation of the patella as a complication of lateral retinacular release. Am J Sports Med. 1988;16(4):383-388.

15. McCarthy MA, Bollier MJ. Medial patella subluxation: diagnosis and treatment. Iowa Orthop J. 2015;35:26-33.

Article PDF
Author and Disclosure Information

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

Issue
The American Journal of Orthopedics - 46(5)
Publications
Topics
Page Number
224-228
Sections
Author and Disclosure Information

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

Author and Disclosure Information

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

Article PDF
Article PDF

Take-Home Points

  • Understanding the indications for treatment is essential.
  • Identifying the superficial (oblique fibers) and deep layers (transverse fibers) of the LR is very important and can lengthen the LR by as much as 20 mm.
  • Open procedures reduce the risk of hematomas and related pain.
  • The goal is to obtain 1 or 2 patellar quadrants of medial and lateral patellar glide in extensino and a neutral patella.
  • If the Z-plasty is combined with the MPFL reconstruction or tibial tubercle transfer, the LR is set to length after the tubercle transfer and before the MPFL reconstruction (to avoid overconstraint).

Anterior knee pain is a common clinical problem that can be challenging to correct, in large part because of multiple causative factors, including structural/anatomical, functional, alignment, and neuroperception/pain pathway factors. One difficult aspect of anatomical assessment is judging the soft-tissue balance between the medial restraints (medial patellofemoral ligament [MPFL]; medial quadriceps tendon to femoral ligament; medial patellotibial and patellomeniscal ligaments) and the lateral restraints (lateral retinaculum [LR] specifically). Both LR tightness and patellar instability can be interpreted as anterior knee pain. Differentiating these entities is one of the most difficult clinical challenges in orthopedics.

LR release (LRR) has been found to improve patellar mobility and tracking.1 In the absence of clearly defined guidelines, the procedure quickly gained in popularity because of its technical simplicity and the enticing "one tool fits all" treatment approach suggested in early reviews. Injudicious use of LRR, alone or in combination with other procedures, led to iatrogenic instability and chronic pain. LR lengthening (LRL) was introduced to address LR tightness while maintaining lateral soft-tissue integrity and avoiding some of the severe complications of LRR.2

Today, isolated use of LRR/LRL is recommended only for treatment of LR tightness and pain secondary to lateral patellar hypercompression.3 It can also be used as an adjunct treatment in the setting of patellofemoral instability. LRR/LRL should never be used as primary treatment for patellofemoral instability.

In this review of treatments for LR tightness and patellofemoral disorders, we compare the use of LRR and LRL.

Discussion

LR procedures are indicated for LR tightness, which is assessed by taking a history, performing a physical examination, and obtaining diagnostic imaging. Decisions should be based on all findings considered together and never on imaging findings alone.

Physical Examination

The physical examination should include assessment of limb alignment, patellar mobility, muscle balance, and dynamic patellar tracking.

Limb Alignment. Abnormal valgus, rotational deformities, and increased Q-angle are associated with LR tightness. Valgus alignment can be assessed on standing inspection; rotational deformities with increased hip anteversion by hip motion with the patient in the prone position (increased hip internal rotation, decreased hip external rotation); and Q-angle on weight-bearing standing examination and with the patient flexing and extending the knee while seated.

Patellar Mobility. The patellar glide and tilt tests provide the most direct evaluations of LR tightness. Medial displacement of <1 quadrant is consistent with tightness, and displacement of >3 quadrants is consistent with laxity. In full extension, the patellar glide test evaluates only the soft-tissue restraints; at 30° flexion, it also evaluates patellofemoral engagement. The patellar tilt test measures the lifting of the lateral edge of the patella. With normal elevation being 0° to 20°, lack of patellar tilt means the LR is tight, and tilt of >20° means it is loose. MPFL patency can be examined with the Lachman test; the examiner rapidly moves the patella laterally while feeling for the characteristic hard endpoint of lateral translation.

Muscle Balance. The tone, strength, and tightness of the core (abdomen, dorsal, and hip muscles) and lower extremities (quadriceps, hamstrings, gastrocnemius) should be evaluated.

Dynamic Patellar Tracking. The J-sign is the course (shaped like an inverted J) that the patella takes when it is medialized into the trochlea from its laterally displaced resting position as the knee goes from full extension to flexion. The J-sign can be associated with LR tightness, trochlear dysplasia, and patella alta.

Imaging

Although we cannot provide a comprehensive review of the imaging literature, the following radiologic examinations should be used to assess the patellofemoral joint.

30° Lateral Radiograph.  Increased tilt is seen when the lateral facet is not anterior to the patellar ridge. Also evaluated are trochlear anatomy, patellar height, and other factors involved in patellofemoral disorders.

30° Flexed Axial (Merchant) Radiograph. Patellar tilt, subluxation, and trochlear dysplasia are evaluated. Images obtained with progressive flexion can be very useful in verifying patellar tilt reduction. Lack of reduction during early flexion suggests LR tightness.4

Alignment Axial Radiographs (Scanogram). Valgus alignment is assessed with this full-length, standing, long-leg examination.

Computed Tomography/Magnetic Resonance Imaging. Many parameters of patellar alignment have been described. Basic assessment should include evaluation of patellar tilt, angle by the line across posterior condyles and a line through the greatest patellar width (>20° indicates abnormality and LR tightness) and tibial tubercle-trochlear groove distance (computed tomography or magnetic resonance imaging scan of the knee is used to measure this distance, and to confirm a significant amount in light of complex patellofemoral malalignment5).

Indications

Lateral compression syndrome with LR tightness is often successfully treated with isolated LRR, and results are reproducible and predictable.6 Surgical intervention for patellofemoral pain should be undertaken only after failed extensive nonoperative treatment with physical therapy and bracing/taping. Patients with LR tightness on preoperative examination, lateral patellar tilt on imaging, and normal Q-angle can obtain satisfactory results with this procedure. Patellar subluxation or dislocation history, high Q-angle (>20°), grade 3 or 4 chondral injury, and patellofemoral arthritis are associated with poorer outcomes when the procedure is performed in isolation.6International Patellofemoral Study Group members agreed that LRR/LRL is a valid treatment option when indicated, but it is rarely performed in isolation and constitutes only 1% to 2% of surgeries performed by this group of experts.7 When lateral compression syndrome progresses to arthritis, LRR/LRL can be performed with lateral patella facetectomy for maximal improvement.4 In the setting of patellofemoral instability, LRR/LRL can be combined with proximal and/or distal realignment surgery if the LR is tight. The LR is the last line of defense limiting lateral translation in the setting of an incompetent MPFL. Isolated LRR/LRL in the setting of instability further destabilizes the patella and worsens the instability. Therefore, LRR/LRL
is a poor surgical option as an isolated procedure for this condition and should be used only as an adjunct in cases of patellofemoral instability with LR tightness that does not allow the patella to be centralized into the trochlea.8 LRR/LRL can also be performed to improve patellar tracking in patello­femoral arthroplasty and total knee arthroplasty.

Lateral Retinaculum Release Versus Lengthening

LRR was first described for the treatment of patellar instability in 1891.9 It was also used for the treatment of lateral patellar hypercompression syndrome associated with LR tightness that led to lateral patellar tracking, joint overload, degeneration, and anterior knee pain.10 Metcalf10 further popularized the procedure by describing a minimally invasive arthroscopic version. However, the arthroscopic technique is as aggressive as the open technique and may be performed with less control, potentially making its results more variable. As proximal and distal releases are performed from the "inside out," more capsule and muscle disruption is needed to release the more superficial layers.

Z-plasty lengthening of the LR was described as an alternative for maintaining lateral patellar soft-tissue integrity while reducing the tension of the lateral tissue restraints.3 This is our preferred method.

Performing LRL instead of LRR avoids iatrogenic medial patellar instability, avoids overrelease and muscle injury, and improves soft-tissue balance.3 Open release or lengthening reduces inadvertent injury to the lateral superior/inferior geniculate arteries and allows direct hemostasis. Two prospective randomized studies found functional knee outcomes and return to athletic activities were improved more after LRL than LRR.11,12 These procedures had similar rates of postoperative knee stiffness, decreased muscle mass, and decreased strength. Each prospective study used an extensive LRR technique for LRR cases (various authors have recommended performing the release until the patella is perpendicular to the trochlea), which may have affected outcomes. In any case, with lengthening, the surgeon is less likely to excessively disrupt the lateral tissues.

Lateral Retinaculum Release.  LRR can be openly performed by lateral parapatellar incision,1 a mini-open percutaneous technique, or arthroscopy. For these open techniques, incisions of various sizes have been used to access the LR and incise it about 1 cm lateral to the patella starting at the distal end of the vastus lateralis and extending distally until patellar tilt reduction is sufficient. If tightness in deep flexion persists, the LRR can be extended distally to the tibial tubercle. Open techniques have the advantage of sparing the joint capsule. All-arthroscopic techniques involve using electrocautery to cut through the capsule and access the LR.

Lateral Retinaculum Lengthening. 

The LR is sharply divided into a superficial layer of superficial oblique fibers from the anterior iliotibial band and a deep layer of transverse fibers from the femur. For LRR, these 2 layers must be identified separate from the articular capsule.13

Figure 1.
Figure 1 illustrates the anatomy and the essential steps of the procedure. The fascia lata is carefully removed over the lateral patella, releasing some lateral tension and helping in the identification of the superficial and deep layers. The proximal lateral soft tissues of the patella are exposed, and the outer oblique layer of the LR is incised near the lateral border of the patella (Figure 2).
Figure 2.
The incision begins around the level of the proximal patellar pole and extends distally to the inferior patellar pole. The superficial oblique fibers of the LR are sharply dissected from the deep transverse fibers and are elevated as dissection is carried posteriorly to the posterior-most extent of the retinacular envelope, usually 1 cm to 2 cm (Figure 3). T
Figure 3.
he deep transverse fibers are then incised longitudinally (Figure 4). In many cases, a capsule that adheres to the deep layer can be separated from it. In cases with combined tibial tubercle transfers, the capsule is incised in order to mobilize the patella medially or distally.
Figure 4.
Then, the knee is moved through its full range of motion, positioned in 30° flexion, and engaged into the trochlear groove. The length of both the MPFL and the LR can then be adjusted. The cut edges of the superior oblique and deep transverse fibers are then sutured together with absorbable suture, and the appropriate amount of lengthening is performed to remove excess tension in lateral structures while maintaining lateral soft-tissue integrity (Figure 5).
Figure 5.
Neither the MPFL nor the LR is tensioned; rather, their lengths are adjusted, as they act as checkreins guiding the patella. If this procedure is performed with tibial tubercle transfer (medial and/or distal) or MPFL reconstruction, tubercle fixation is done first; next, MPFL length and then LR length are set.

Complications

Complications of performing LRR/LRL to change the lateral restraint include medial patellar instability, increased lateral pain, repair failure, recurrent lateral instability, quadriceps weakness and atrophy, postoperative hemarthrosis, knee stiffness, wound complications, and thermal skin injury.7 These complications often result from poor surgical technique and too aggressive release. Although recommended patellar tilt historically has varied from 45° to 90°, the current goal is to normalize the tight soft-tissue restraints without creating secondary instability.

The most significant complication of LRR is medial patellar instability caused by muscle atrophy and loss of soft-tissue restraint.14 Medial instability can be difficult to diagnose and should be considered in any patient with patellofemoral pain, popping, or patellar instability after LRR.15 A positive medial subluxation test or medial patellar apprehension test suggests medial instability.

Medial patellar instability usually requires surgical treatment. Direct LR repair, lateral soft-tissue reconstruction, and other procedures can be used to restore lateral restraint.15 However, these are salvage techniques, and patients often remain significantly limited by pain or instability. Therefore, the LR must be carefully addressed and preferably should undergo lengthening rather than release.

Take-Home Points

  • Understanding the indications for treatment is essential.
  • Identifying the superficial (oblique fibers) and deep layers (transverse fibers) of the LR is very important and can lengthen the LR by as much as 20 mm.
  • Open procedures reduce the risk of hematomas and related pain.
  • The goal is to obtain 1 or 2 patellar quadrants of medial and lateral patellar glide in extensino and a neutral patella.
  • If the Z-plasty is combined with the MPFL reconstruction or tibial tubercle transfer, the LR is set to length after the tubercle transfer and before the MPFL reconstruction (to avoid overconstraint).

Anterior knee pain is a common clinical problem that can be challenging to correct, in large part because of multiple causative factors, including structural/anatomical, functional, alignment, and neuroperception/pain pathway factors. One difficult aspect of anatomical assessment is judging the soft-tissue balance between the medial restraints (medial patellofemoral ligament [MPFL]; medial quadriceps tendon to femoral ligament; medial patellotibial and patellomeniscal ligaments) and the lateral restraints (lateral retinaculum [LR] specifically). Both LR tightness and patellar instability can be interpreted as anterior knee pain. Differentiating these entities is one of the most difficult clinical challenges in orthopedics.

LR release (LRR) has been found to improve patellar mobility and tracking.1 In the absence of clearly defined guidelines, the procedure quickly gained in popularity because of its technical simplicity and the enticing "one tool fits all" treatment approach suggested in early reviews. Injudicious use of LRR, alone or in combination with other procedures, led to iatrogenic instability and chronic pain. LR lengthening (LRL) was introduced to address LR tightness while maintaining lateral soft-tissue integrity and avoiding some of the severe complications of LRR.2

Today, isolated use of LRR/LRL is recommended only for treatment of LR tightness and pain secondary to lateral patellar hypercompression.3 It can also be used as an adjunct treatment in the setting of patellofemoral instability. LRR/LRL should never be used as primary treatment for patellofemoral instability.

In this review of treatments for LR tightness and patellofemoral disorders, we compare the use of LRR and LRL.

Discussion

LR procedures are indicated for LR tightness, which is assessed by taking a history, performing a physical examination, and obtaining diagnostic imaging. Decisions should be based on all findings considered together and never on imaging findings alone.

Physical Examination

The physical examination should include assessment of limb alignment, patellar mobility, muscle balance, and dynamic patellar tracking.

Limb Alignment. Abnormal valgus, rotational deformities, and increased Q-angle are associated with LR tightness. Valgus alignment can be assessed on standing inspection; rotational deformities with increased hip anteversion by hip motion with the patient in the prone position (increased hip internal rotation, decreased hip external rotation); and Q-angle on weight-bearing standing examination and with the patient flexing and extending the knee while seated.

Patellar Mobility. The patellar glide and tilt tests provide the most direct evaluations of LR tightness. Medial displacement of <1 quadrant is consistent with tightness, and displacement of >3 quadrants is consistent with laxity. In full extension, the patellar glide test evaluates only the soft-tissue restraints; at 30° flexion, it also evaluates patellofemoral engagement. The patellar tilt test measures the lifting of the lateral edge of the patella. With normal elevation being 0° to 20°, lack of patellar tilt means the LR is tight, and tilt of >20° means it is loose. MPFL patency can be examined with the Lachman test; the examiner rapidly moves the patella laterally while feeling for the characteristic hard endpoint of lateral translation.

Muscle Balance. The tone, strength, and tightness of the core (abdomen, dorsal, and hip muscles) and lower extremities (quadriceps, hamstrings, gastrocnemius) should be evaluated.

Dynamic Patellar Tracking. The J-sign is the course (shaped like an inverted J) that the patella takes when it is medialized into the trochlea from its laterally displaced resting position as the knee goes from full extension to flexion. The J-sign can be associated with LR tightness, trochlear dysplasia, and patella alta.

Imaging

Although we cannot provide a comprehensive review of the imaging literature, the following radiologic examinations should be used to assess the patellofemoral joint.

30° Lateral Radiograph.  Increased tilt is seen when the lateral facet is not anterior to the patellar ridge. Also evaluated are trochlear anatomy, patellar height, and other factors involved in patellofemoral disorders.

30° Flexed Axial (Merchant) Radiograph. Patellar tilt, subluxation, and trochlear dysplasia are evaluated. Images obtained with progressive flexion can be very useful in verifying patellar tilt reduction. Lack of reduction during early flexion suggests LR tightness.4

Alignment Axial Radiographs (Scanogram). Valgus alignment is assessed with this full-length, standing, long-leg examination.

Computed Tomography/Magnetic Resonance Imaging. Many parameters of patellar alignment have been described. Basic assessment should include evaluation of patellar tilt, angle by the line across posterior condyles and a line through the greatest patellar width (>20° indicates abnormality and LR tightness) and tibial tubercle-trochlear groove distance (computed tomography or magnetic resonance imaging scan of the knee is used to measure this distance, and to confirm a significant amount in light of complex patellofemoral malalignment5).

Indications

Lateral compression syndrome with LR tightness is often successfully treated with isolated LRR, and results are reproducible and predictable.6 Surgical intervention for patellofemoral pain should be undertaken only after failed extensive nonoperative treatment with physical therapy and bracing/taping. Patients with LR tightness on preoperative examination, lateral patellar tilt on imaging, and normal Q-angle can obtain satisfactory results with this procedure. Patellar subluxation or dislocation history, high Q-angle (>20°), grade 3 or 4 chondral injury, and patellofemoral arthritis are associated with poorer outcomes when the procedure is performed in isolation.6International Patellofemoral Study Group members agreed that LRR/LRL is a valid treatment option when indicated, but it is rarely performed in isolation and constitutes only 1% to 2% of surgeries performed by this group of experts.7 When lateral compression syndrome progresses to arthritis, LRR/LRL can be performed with lateral patella facetectomy for maximal improvement.4 In the setting of patellofemoral instability, LRR/LRL can be combined with proximal and/or distal realignment surgery if the LR is tight. The LR is the last line of defense limiting lateral translation in the setting of an incompetent MPFL. Isolated LRR/LRL in the setting of instability further destabilizes the patella and worsens the instability. Therefore, LRR/LRL
is a poor surgical option as an isolated procedure for this condition and should be used only as an adjunct in cases of patellofemoral instability with LR tightness that does not allow the patella to be centralized into the trochlea.8 LRR/LRL can also be performed to improve patellar tracking in patello­femoral arthroplasty and total knee arthroplasty.

Lateral Retinaculum Release Versus Lengthening

LRR was first described for the treatment of patellar instability in 1891.9 It was also used for the treatment of lateral patellar hypercompression syndrome associated with LR tightness that led to lateral patellar tracking, joint overload, degeneration, and anterior knee pain.10 Metcalf10 further popularized the procedure by describing a minimally invasive arthroscopic version. However, the arthroscopic technique is as aggressive as the open technique and may be performed with less control, potentially making its results more variable. As proximal and distal releases are performed from the "inside out," more capsule and muscle disruption is needed to release the more superficial layers.

Z-plasty lengthening of the LR was described as an alternative for maintaining lateral patellar soft-tissue integrity while reducing the tension of the lateral tissue restraints.3 This is our preferred method.

Performing LRL instead of LRR avoids iatrogenic medial patellar instability, avoids overrelease and muscle injury, and improves soft-tissue balance.3 Open release or lengthening reduces inadvertent injury to the lateral superior/inferior geniculate arteries and allows direct hemostasis. Two prospective randomized studies found functional knee outcomes and return to athletic activities were improved more after LRL than LRR.11,12 These procedures had similar rates of postoperative knee stiffness, decreased muscle mass, and decreased strength. Each prospective study used an extensive LRR technique for LRR cases (various authors have recommended performing the release until the patella is perpendicular to the trochlea), which may have affected outcomes. In any case, with lengthening, the surgeon is less likely to excessively disrupt the lateral tissues.

Lateral Retinaculum Release.  LRR can be openly performed by lateral parapatellar incision,1 a mini-open percutaneous technique, or arthroscopy. For these open techniques, incisions of various sizes have been used to access the LR and incise it about 1 cm lateral to the patella starting at the distal end of the vastus lateralis and extending distally until patellar tilt reduction is sufficient. If tightness in deep flexion persists, the LRR can be extended distally to the tibial tubercle. Open techniques have the advantage of sparing the joint capsule. All-arthroscopic techniques involve using electrocautery to cut through the capsule and access the LR.

Lateral Retinaculum Lengthening. 

The LR is sharply divided into a superficial layer of superficial oblique fibers from the anterior iliotibial band and a deep layer of transverse fibers from the femur. For LRR, these 2 layers must be identified separate from the articular capsule.13

Figure 1.
Figure 1 illustrates the anatomy and the essential steps of the procedure. The fascia lata is carefully removed over the lateral patella, releasing some lateral tension and helping in the identification of the superficial and deep layers. The proximal lateral soft tissues of the patella are exposed, and the outer oblique layer of the LR is incised near the lateral border of the patella (Figure 2).
Figure 2.
The incision begins around the level of the proximal patellar pole and extends distally to the inferior patellar pole. The superficial oblique fibers of the LR are sharply dissected from the deep transverse fibers and are elevated as dissection is carried posteriorly to the posterior-most extent of the retinacular envelope, usually 1 cm to 2 cm (Figure 3). T
Figure 3.
he deep transverse fibers are then incised longitudinally (Figure 4). In many cases, a capsule that adheres to the deep layer can be separated from it. In cases with combined tibial tubercle transfers, the capsule is incised in order to mobilize the patella medially or distally.
Figure 4.
Then, the knee is moved through its full range of motion, positioned in 30° flexion, and engaged into the trochlear groove. The length of both the MPFL and the LR can then be adjusted. The cut edges of the superior oblique and deep transverse fibers are then sutured together with absorbable suture, and the appropriate amount of lengthening is performed to remove excess tension in lateral structures while maintaining lateral soft-tissue integrity (Figure 5).
Figure 5.
Neither the MPFL nor the LR is tensioned; rather, their lengths are adjusted, as they act as checkreins guiding the patella. If this procedure is performed with tibial tubercle transfer (medial and/or distal) or MPFL reconstruction, tubercle fixation is done first; next, MPFL length and then LR length are set.

Complications

Complications of performing LRR/LRL to change the lateral restraint include medial patellar instability, increased lateral pain, repair failure, recurrent lateral instability, quadriceps weakness and atrophy, postoperative hemarthrosis, knee stiffness, wound complications, and thermal skin injury.7 These complications often result from poor surgical technique and too aggressive release. Although recommended patellar tilt historically has varied from 45° to 90°, the current goal is to normalize the tight soft-tissue restraints without creating secondary instability.

The most significant complication of LRR is medial patellar instability caused by muscle atrophy and loss of soft-tissue restraint.14 Medial instability can be difficult to diagnose and should be considered in any patient with patellofemoral pain, popping, or patellar instability after LRR.15 A positive medial subluxation test or medial patellar apprehension test suggests medial instability.

Medial patellar instability usually requires surgical treatment. Direct LR repair, lateral soft-tissue reconstruction, and other procedures can be used to restore lateral restraint.15 However, these are salvage techniques, and patients often remain significantly limited by pain or instability. Therefore, the LR must be carefully addressed and preferably should undergo lengthening rather than release.

References

1. Merchant AC, Mercer RL. Lateral release of the patella. A preliminary report. Clin Orthop Relat Res. 1974;(103):40-45.

2. Ceder LC, Larson RL. Z-plasty lateral retinacular release for the treatment of patellar compression syndrome. Clin Orthop Relat Res. 1979;(144):110-113.

3. Biedert R. Lateral patellar hypercompression, tilt and mild lateral subluxation. In: Biedert R, ed. Patellofemoral Disorders. Chichester, England: Wiley; 2004:161-166.

4. Hinckel BB, Arendt EA. Lateral retinaculum lengthening or release. Oper Tech Sports Med. 2015;23(2):100-106.

5. Seitlinger G, Scheurecker G, Högler R, Labey L, Innocenti B, Hofmann S. Tibial tubercle–posterior cruciate ligament distance: a new measurement to define the position of the tibial tubercle in patients with patellar dislocation. Am J Sports Med. 2012;40(5):1119-1125.

6. Lattermann C, Toth J, Bach BR Jr. The role of lateral retinacular release in the treatment of patellar instability. Sports Med Arthrosc. 2007;15(2):57-60.

7. Fithian DC, Paxton EW, Post WR, Panni AS; International Patellofemoral Study Group. Lateral retinacular release: a survey of the International Patellofemoral Study Group. Arthroscopy. 2004;20(5):463-468.

8. Christoforakis J, Bull AM, Strachan RK, Shymkiw R, Senavongse W, Amis AA. Effects of lateral retinacular release on the lateral stability of the patella. Knee Surg Sports Traumatol Arthrosc. 2006;14(3):273-277.

9. Pollard B. Old dislocation of patella by intra-articular operation. Lancet. 1891;(988):17-22.

10. Metcalf RW. An arthroscopic method for lateral release of subluxating or dislocating patella. Clin Orthop Relat Res. 1982;167:9-18.

11. Pagenstert G, Wolf N, Bachmann M, et al. Open lateral patellar retinacular lengthening versus open retinacular release in lateral patellar hypercompression syndrome: a prospective double-blinded comparative study on complications and outcome. Arthroscopy. 2012;28(6):788-797.

12. O’Neill DB. Open lateral retinacular lengthening compared with arthroscopic release. A prospective, randomized outcome study. J Bone Joint Surg Am. 1997;79(12):1759-1769.

13. Merican AM, Amis AA. Anatomy of the lateral retinaculum of the knee. J Bone Joint Surg Br. 2008;90(4):527-534.

14. Hughston JC, Deese M. Medial subluxation of the patella as a complication of lateral retinacular release. Am J Sports Med. 1988;16(4):383-388.

15. McCarthy MA, Bollier MJ. Medial patella subluxation: diagnosis and treatment. Iowa Orthop J. 2015;35:26-33.

References

1. Merchant AC, Mercer RL. Lateral release of the patella. A preliminary report. Clin Orthop Relat Res. 1974;(103):40-45.

2. Ceder LC, Larson RL. Z-plasty lateral retinacular release for the treatment of patellar compression syndrome. Clin Orthop Relat Res. 1979;(144):110-113.

3. Biedert R. Lateral patellar hypercompression, tilt and mild lateral subluxation. In: Biedert R, ed. Patellofemoral Disorders. Chichester, England: Wiley; 2004:161-166.

4. Hinckel BB, Arendt EA. Lateral retinaculum lengthening or release. Oper Tech Sports Med. 2015;23(2):100-106.

5. Seitlinger G, Scheurecker G, Högler R, Labey L, Innocenti B, Hofmann S. Tibial tubercle–posterior cruciate ligament distance: a new measurement to define the position of the tibial tubercle in patients with patellar dislocation. Am J Sports Med. 2012;40(5):1119-1125.

6. Lattermann C, Toth J, Bach BR Jr. The role of lateral retinacular release in the treatment of patellar instability. Sports Med Arthrosc. 2007;15(2):57-60.

7. Fithian DC, Paxton EW, Post WR, Panni AS; International Patellofemoral Study Group. Lateral retinacular release: a survey of the International Patellofemoral Study Group. Arthroscopy. 2004;20(5):463-468.

8. Christoforakis J, Bull AM, Strachan RK, Shymkiw R, Senavongse W, Amis AA. Effects of lateral retinacular release on the lateral stability of the patella. Knee Surg Sports Traumatol Arthrosc. 2006;14(3):273-277.

9. Pollard B. Old dislocation of patella by intra-articular operation. Lancet. 1891;(988):17-22.

10. Metcalf RW. An arthroscopic method for lateral release of subluxating or dislocating patella. Clin Orthop Relat Res. 1982;167:9-18.

11. Pagenstert G, Wolf N, Bachmann M, et al. Open lateral patellar retinacular lengthening versus open retinacular release in lateral patellar hypercompression syndrome: a prospective double-blinded comparative study on complications and outcome. Arthroscopy. 2012;28(6):788-797.

12. O’Neill DB. Open lateral retinacular lengthening compared with arthroscopic release. A prospective, randomized outcome study. J Bone Joint Surg Am. 1997;79(12):1759-1769.

13. Merican AM, Amis AA. Anatomy of the lateral retinaculum of the knee. J Bone Joint Surg Br. 2008;90(4):527-534.

14. Hughston JC, Deese M. Medial subluxation of the patella as a complication of lateral retinacular release. Am J Sports Med. 1988;16(4):383-388.

15. McCarthy MA, Bollier MJ. Medial patella subluxation: diagnosis and treatment. Iowa Orthop J. 2015;35:26-33.

Issue
The American Journal of Orthopedics - 46(5)
Issue
The American Journal of Orthopedics - 46(5)
Page Number
224-228
Page Number
224-228
Publications
Publications
Topics
Article Type
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Article PDF Media

Patella Alta Sees You, Do You See It?

Article Type
Changed
Thu, 09/19/2019 - 13:21

Take-Home Points

  • The decision to adda TTDO to an MPFL reconstruction is dependent on patellar height as assessed with the CDI, as well as multiple other patient and anatomical factors.

  • TTDOs that include a complete detachment of the tibial tubercle (as required for distalization) have increased risk of nonunion and hardware failure.

  • Poor surgical technique (failure to make a flat osteotomy cut, cortical only bone block, poor bony apposition of the detached bone block- particularly at the location of any transverse plane cut, and failure to minimize thermal damage through copious irrigation) can increase nonunion risk.

  • Postoperative rehabilitation should include a 6-week period of limited weight-bearing.

  • Reconstruction of the MPFL should be performed after any TTO is performed.

Patellar instability is the result of numerous anatomical factors, including trochlear dysplasia,1,2 patella alta,2-4 and increased tibial tubercle-trochlear groove (TT-TG) or tibial tubercle-posterior cruciate ligament distance.2,5 Of all the factors, TT-TG distance and the medial patellofemoral ligament (MPFL) have received the ost attention. Patellar height remains a crucial yet underappreciated contributor that is amenable to surgical correction with tibial tubercle distalization osteotomy (TTDO). The obvious question is how severe patella alta must be to require surgical correction. In other words, when is patella alta so severe that isolated MPFL reconstruction is insufficient to restore patellar stability?

The indications for TTDO are not completely clear and depend on multiple factors. Patient factors, physical examination findings, and radiographic measures must be considered. In general, adding TTDO to MPFL reconstruction should be considered when the degree of patella alta exceeds 1.4 on the Caton-Deschamps Index (CDI). Presence of trochlear dysplasia, patellar maltracking (J-sign), lateral patellar apprehension that persists at higher flexion angles, and decreased patellotrochlear articular cartilage contact on sagittal magnetic resonance imaging may drive the decision to proceed with TTDO when the CDI is lower.

Why You Need To Know About Patella Alta

Recurrent lateral patellar dislocation is a debilitating knee condition that often involves young, active patients and significantly affects their quality of life. The MPFL is a primary restraint to lateral patellar dislocation, and an MPFL injury is a key contributor to loss of patellar stability. MPFL reconstruction is increasingly being performed to treat recurrent lateral patellar instability.6 Patellar instability is the result of numerous anatomical factors, including trochlear dysplasia,1,2 patella alta,2-4 and increased TT-TG distance.2,5 This review focuses on patella alta.

The classic teaching of the Lyon School of Knee Surgery in France, the menu à la carte, is that patella alta exceeding 1.2 on the CDI is an indication for TTDO.2 Although this teaching is an excellent guide for normal anatomy, we must keep in mind that the classic surgical menu does not consider the influence of MPFL reconstruction, as development of the menu predated this surgical option. At that time, the proximal soft-tissue procedures included vastus medialis obliquus plasty and advancement, which are performed to balance soft tissues and treat patellar tilt. These procedures and MPFL reconstruction have different functions, and the difference may be important. Furthermore, performing TTDO alongside MPFL reconstruction significantly increases the risk of complications and alters the rehabilitation protocol. However, significant untreated patella alta has been implicated as a cause of failure of isolated MPFL reconstruction.7 Establishing when MPFL reconstruction alone is sufficient is therefore crucial in avoiding the increased morbidity associated with the addition of TTDO.

Discussion

Above a certain degree of patella alta, isolated MPFL reconstruction fails to restore patellar stability. What remains unknown is the appropriate CDI cutoff (1.4) and whether the same cutoff can be used for all patients. In 2013, Wagner and colleagues8 assessed the influence of patella alta on isolated MPFL reconstruction outcomes and found no significant difference, though their study did not include many patients with significant alta and was underpowered. In 2014, Feller and colleagues9 reported on a series of patients who were successfully treated with isolated MPFL reconstruction despite patella alta significantly exceeding the traditional CDI cutoff of 1.2. Their indication for performing the isolated procedure was normal patellar tracking—in particular, absence of the J-sign. Further analysis of these patients revealed a preponderance of relatively normal TT-TG distances and low-grade, if any, trochlear dysplasia in comparison with other patients treated with a combination of MPFL reconstruction and tibial tubercle osteotomy.

Together, the work of Wagner and colleagues8 and Feller and colleagues9 suggests the historical use of the CDI of 1.2 as a hard and fast indication for adding TTDO is aggressive. In fact, it is probably the case that there really is no single CDI cutoff that is an appropriate indication for adding TTDO in all patients with instability. This decision is, and should be, influenced by a multitude of other factors, including other anatomical factors, physical examination findings, patient factors, and, of course, patient preference.

An interesting idea to consider in treating patellar instability is the interplay of patella alta and trochlear dysplasia. Patella alta is theorized as contributing to patellar instability in part by delaying entry of the patella into the TG as the knee flexes, therefore requiring less force to laterally displace

the patella.10 Similarly, in the setting of trochlear dysplasia, a shallow TG leads to less bony constraint of the patella, particularly in the groove’s superior portions, which are more involved in lower grade dysplasia. Because trochlear dysplasia and patella alta decrease patellar stability by similar mechanisms, they clearly interact, and a patient with both is at higher risk for instability than a patient who exhibits either in isolation.11 Therefore, trochlear dysplasia, particularly higher grade, may be an indication for adding TTDO at lower CDI.

Other imaging and physical examination factors can provide additional insight into the process of patellar engagement into the trochlea in each patient. The patellotrochlear index (PTI) directly measures the relationship between the patella and the trochlea, rather than relative to the tibia, as with other measures of patellar height.12[[{"fid":"201853","view_mode":"medstat_image_flush_left","attributes":{"class":"media-element file-medstat-image-flush-left","data-delta":"1"},"fields":{"format":"medstat_image_flush_left","field_file_image_caption[und][0][value]":"Figure 1.","field_file_image_credit[und][0][value]":"","field_file_image_caption[und][0][format]":"plain_text","field_file_image_credit[und][0][format]":"plain_text"},"type":"media","field_deltas":{"1":{"format":"medstat_image_flush_left","field_file_image_caption[und][0][value]":"Figure 1.","field_file_image_credit[und][0][value]":""}}}]]The PTI is correlated with tibia-based measures of height, but the correlation is not perfect. Lower degrees of overlap between the patella and the trochlea (PTI <0.15) and significant functional patella alta may warrant adding TTDO in cases of borderline CDI (1.2-1.4). Figures 1A, 1B and 2A, 2B show the imaging of 2 patients with relatively similar patellar height (assessed with CDI) but quite different degrees of overlap between the patella and trochlea. [[{"fid":"201854","view_mode":"medstat_image_flush_right","attributes":{"class":"media-element file-medstat-image-flush-right","data-delta":"2"},"fields":{"format":"medstat_image_flush_right","field_file_image_caption[und][0][value]":"Figure 2.","field_file_image_credit[und][0][value]":"","field_file_image_caption[und][0][format]":"plain_text","field_file_image_credit[und][0][format]":"plain_text"},"type":"media","field_deltas":{"2":{"format":"medstat_image_flush_right","field_file_image_caption[und][0][value]":"Figure 2.","field_file_image_credit[und][0][value]":""}}}]]The patient with less overlap is more likely to have delayed patellar engagement and symptomatic patella alta and thus may be a poorer candidate for isolated MPFL reconstruction. For additional information, please refer to the work by Roland Biedert, MD, who has proposed trochlear lengthening in these situations.13

Physical examination (even in the era of advance imaging) continues to provide useful insight into whether to add TTDO. One physical examination test that can help in understanding patellar-
trochlear dynamics is the patellar apprehension and relief test. Patellar apprehension has been widely discussed, but equally important is the degree of knee flexion above which apprehension dissipates. As patella alta and trochlear dysplasia become more severe, more knee flexion is required to relieve apprehension. Apprehension that is relieved at 30° to 40° of flexion suggests that patellar stability stands a good chance of being restored with isolated MPFL reconstruction, whereas persistent instability >45° or especially 60° of knee flexion suggests that there is significant patella alta, trochlear dysplasia, or both and that TTDO should be added. A large J-sign during knee flexion and extension provides further evidence that entry of the patella into the TG is delayed, typically because of patella alta, trochlear dysplasia, or both, and possibly tight lateral structures or a lateralized tibial tubercle. This sign is another clue that isolated MPFL reconstruction may be insufficient to completely restore patellar stability.

References

1. Dejour H, Walch G, Neyret P, Adeleine P. Dysplasia of the femoral trochlea [in French]. Rev Chir Orthop Reparatrice Appar Mot. 1990;76(1):45-54.

2. Dejour H, Walch G, Nove-Josserand L, Guier C. Factors of patellar instability: an anatomic radiographic study. Knee Surg Sports Traumatol Arthrosc. 1994;2(1):19-26.

3. Geenen E, Molenaers G, Martens M. Patella alta in patellofemoral instability. Acta Orthop Belg. 1989;55(3):387-393.

4. Simmons E Jr, Cameron JC. Patella alta and recurrent dislocation of the patella. Clin Orthop Relat Res. 1992;(274):265-269.

5. Goutallier D, Bernageau J, Lecudonnec B. The measurement of the tibial tuberosity. Patella groove distanced technique and results (author’s transl) [in French]. Rev Chir Orthop Reparatrice Appar Mot. 1978;64(5):423-428.

6. Feller JA, Lind M, Nelson J, Diduch DR, Arendt E. Repair and reconstruction of the medial patellofemoral ligament for treatment of lateral patellar dislocations. In: Scott WN, ed. Insall & Scott—Surgery of the Knee. 5th ed. Philadelphia, PA: Churchill Livingstone; 2011:677-687.

7. Thaunat M, Erasmus PJ. Recurrent patellar dislocation after medial patellofemoral ligament reconstruction. Knee Surg Sports Traumatol Arthrosc. 2008;16(1):40-43.

8. Wagner D, Pfalzer F, Hingelbaum S, Huth J, Mauch F, Bauer G. The influence of risk factors on clinical outcomes following anatomical medial patellofemoral ligament (MPFL) reconstruction using the gracilis tendon. Knee Surg Sports Traumatol Arthrosc. 2013;21(2):318-324.

9. Feller JA, Richmond AK, Wasiak J. Medial patellofemoral ligament reconstruction as an isolated or combined procedure for recurrent patellar instability. Knee Surg Sports Traumatol Arthrosc. 2014;22(10):2470-2476.

10. Ward SR, Terk MR, Powers CM. Patella alta: association
with patellofemoral alignment and changes in contact area during weight-bearing. J Bone Joint Surg Am. 2007;89(8):
1749-1755.

11. Lewallen L, McIntosh A, Dahm D. First-time patellofemoral dislocation: risk factors for recurrent instability. J Knee Surg. 2015;28(4):303-309

12. Biedert RM, Albrecht S. The patellotrochlear index: a new index for assessing patellar height. Knee Surg Sports Traumatol Arthrosc. 2006;14(8):707-712.

13. Biedert RM. Trochlear lengthening osteotomy with or without elevation of the lateral trochlear facet. In: Zaffagnini S, Dejour D, Arendt EA, eds. Patellofemoral Pain, Instability, and Arthritis. Germany: Springer-Verlag Berlin Heidelberg; 2010:
209-215.

Article PDF
Author and Disclosure Information

Author's Disclosure Statement: The authors report no actual or potential conflict of interest in relation to this article.

Issue
The American Journal of Orthopedics - 46(5)
Publications
Topics
Page Number
229-231
Sections
Author and Disclosure Information

Author's Disclosure Statement: The authors report no actual or potential conflict of interest in relation to this article.

Author and Disclosure Information

Author's Disclosure Statement: The authors report no actual or potential conflict of interest in relation to this article.

Article PDF
Article PDF

Take-Home Points

  • The decision to adda TTDO to an MPFL reconstruction is dependent on patellar height as assessed with the CDI, as well as multiple other patient and anatomical factors.

  • TTDOs that include a complete detachment of the tibial tubercle (as required for distalization) have increased risk of nonunion and hardware failure.

  • Poor surgical technique (failure to make a flat osteotomy cut, cortical only bone block, poor bony apposition of the detached bone block- particularly at the location of any transverse plane cut, and failure to minimize thermal damage through copious irrigation) can increase nonunion risk.

  • Postoperative rehabilitation should include a 6-week period of limited weight-bearing.

  • Reconstruction of the MPFL should be performed after any TTO is performed.

Patellar instability is the result of numerous anatomical factors, including trochlear dysplasia,1,2 patella alta,2-4 and increased tibial tubercle-trochlear groove (TT-TG) or tibial tubercle-posterior cruciate ligament distance.2,5 Of all the factors, TT-TG distance and the medial patellofemoral ligament (MPFL) have received the ost attention. Patellar height remains a crucial yet underappreciated contributor that is amenable to surgical correction with tibial tubercle distalization osteotomy (TTDO). The obvious question is how severe patella alta must be to require surgical correction. In other words, when is patella alta so severe that isolated MPFL reconstruction is insufficient to restore patellar stability?

The indications for TTDO are not completely clear and depend on multiple factors. Patient factors, physical examination findings, and radiographic measures must be considered. In general, adding TTDO to MPFL reconstruction should be considered when the degree of patella alta exceeds 1.4 on the Caton-Deschamps Index (CDI). Presence of trochlear dysplasia, patellar maltracking (J-sign), lateral patellar apprehension that persists at higher flexion angles, and decreased patellotrochlear articular cartilage contact on sagittal magnetic resonance imaging may drive the decision to proceed with TTDO when the CDI is lower.

Why You Need To Know About Patella Alta

Recurrent lateral patellar dislocation is a debilitating knee condition that often involves young, active patients and significantly affects their quality of life. The MPFL is a primary restraint to lateral patellar dislocation, and an MPFL injury is a key contributor to loss of patellar stability. MPFL reconstruction is increasingly being performed to treat recurrent lateral patellar instability.6 Patellar instability is the result of numerous anatomical factors, including trochlear dysplasia,1,2 patella alta,2-4 and increased TT-TG distance.2,5 This review focuses on patella alta.

The classic teaching of the Lyon School of Knee Surgery in France, the menu à la carte, is that patella alta exceeding 1.2 on the CDI is an indication for TTDO.2 Although this teaching is an excellent guide for normal anatomy, we must keep in mind that the classic surgical menu does not consider the influence of MPFL reconstruction, as development of the menu predated this surgical option. At that time, the proximal soft-tissue procedures included vastus medialis obliquus plasty and advancement, which are performed to balance soft tissues and treat patellar tilt. These procedures and MPFL reconstruction have different functions, and the difference may be important. Furthermore, performing TTDO alongside MPFL reconstruction significantly increases the risk of complications and alters the rehabilitation protocol. However, significant untreated patella alta has been implicated as a cause of failure of isolated MPFL reconstruction.7 Establishing when MPFL reconstruction alone is sufficient is therefore crucial in avoiding the increased morbidity associated with the addition of TTDO.

Discussion

Above a certain degree of patella alta, isolated MPFL reconstruction fails to restore patellar stability. What remains unknown is the appropriate CDI cutoff (1.4) and whether the same cutoff can be used for all patients. In 2013, Wagner and colleagues8 assessed the influence of patella alta on isolated MPFL reconstruction outcomes and found no significant difference, though their study did not include many patients with significant alta and was underpowered. In 2014, Feller and colleagues9 reported on a series of patients who were successfully treated with isolated MPFL reconstruction despite patella alta significantly exceeding the traditional CDI cutoff of 1.2. Their indication for performing the isolated procedure was normal patellar tracking—in particular, absence of the J-sign. Further analysis of these patients revealed a preponderance of relatively normal TT-TG distances and low-grade, if any, trochlear dysplasia in comparison with other patients treated with a combination of MPFL reconstruction and tibial tubercle osteotomy.

Together, the work of Wagner and colleagues8 and Feller and colleagues9 suggests the historical use of the CDI of 1.2 as a hard and fast indication for adding TTDO is aggressive. In fact, it is probably the case that there really is no single CDI cutoff that is an appropriate indication for adding TTDO in all patients with instability. This decision is, and should be, influenced by a multitude of other factors, including other anatomical factors, physical examination findings, patient factors, and, of course, patient preference.

An interesting idea to consider in treating patellar instability is the interplay of patella alta and trochlear dysplasia. Patella alta is theorized as contributing to patellar instability in part by delaying entry of the patella into the TG as the knee flexes, therefore requiring less force to laterally displace

the patella.10 Similarly, in the setting of trochlear dysplasia, a shallow TG leads to less bony constraint of the patella, particularly in the groove’s superior portions, which are more involved in lower grade dysplasia. Because trochlear dysplasia and patella alta decrease patellar stability by similar mechanisms, they clearly interact, and a patient with both is at higher risk for instability than a patient who exhibits either in isolation.11 Therefore, trochlear dysplasia, particularly higher grade, may be an indication for adding TTDO at lower CDI.

Other imaging and physical examination factors can provide additional insight into the process of patellar engagement into the trochlea in each patient. The patellotrochlear index (PTI) directly measures the relationship between the patella and the trochlea, rather than relative to the tibia, as with other measures of patellar height.12[[{"fid":"201853","view_mode":"medstat_image_flush_left","attributes":{"class":"media-element file-medstat-image-flush-left","data-delta":"1"},"fields":{"format":"medstat_image_flush_left","field_file_image_caption[und][0][value]":"Figure 1.","field_file_image_credit[und][0][value]":"","field_file_image_caption[und][0][format]":"plain_text","field_file_image_credit[und][0][format]":"plain_text"},"type":"media","field_deltas":{"1":{"format":"medstat_image_flush_left","field_file_image_caption[und][0][value]":"Figure 1.","field_file_image_credit[und][0][value]":""}}}]]The PTI is correlated with tibia-based measures of height, but the correlation is not perfect. Lower degrees of overlap between the patella and the trochlea (PTI <0.15) and significant functional patella alta may warrant adding TTDO in cases of borderline CDI (1.2-1.4). Figures 1A, 1B and 2A, 2B show the imaging of 2 patients with relatively similar patellar height (assessed with CDI) but quite different degrees of overlap between the patella and trochlea. [[{"fid":"201854","view_mode":"medstat_image_flush_right","attributes":{"class":"media-element file-medstat-image-flush-right","data-delta":"2"},"fields":{"format":"medstat_image_flush_right","field_file_image_caption[und][0][value]":"Figure 2.","field_file_image_credit[und][0][value]":"","field_file_image_caption[und][0][format]":"plain_text","field_file_image_credit[und][0][format]":"plain_text"},"type":"media","field_deltas":{"2":{"format":"medstat_image_flush_right","field_file_image_caption[und][0][value]":"Figure 2.","field_file_image_credit[und][0][value]":""}}}]]The patient with less overlap is more likely to have delayed patellar engagement and symptomatic patella alta and thus may be a poorer candidate for isolated MPFL reconstruction. For additional information, please refer to the work by Roland Biedert, MD, who has proposed trochlear lengthening in these situations.13

Physical examination (even in the era of advance imaging) continues to provide useful insight into whether to add TTDO. One physical examination test that can help in understanding patellar-
trochlear dynamics is the patellar apprehension and relief test. Patellar apprehension has been widely discussed, but equally important is the degree of knee flexion above which apprehension dissipates. As patella alta and trochlear dysplasia become more severe, more knee flexion is required to relieve apprehension. Apprehension that is relieved at 30° to 40° of flexion suggests that patellar stability stands a good chance of being restored with isolated MPFL reconstruction, whereas persistent instability >45° or especially 60° of knee flexion suggests that there is significant patella alta, trochlear dysplasia, or both and that TTDO should be added. A large J-sign during knee flexion and extension provides further evidence that entry of the patella into the TG is delayed, typically because of patella alta, trochlear dysplasia, or both, and possibly tight lateral structures or a lateralized tibial tubercle. This sign is another clue that isolated MPFL reconstruction may be insufficient to completely restore patellar stability.

Take-Home Points

  • The decision to adda TTDO to an MPFL reconstruction is dependent on patellar height as assessed with the CDI, as well as multiple other patient and anatomical factors.

  • TTDOs that include a complete detachment of the tibial tubercle (as required for distalization) have increased risk of nonunion and hardware failure.

  • Poor surgical technique (failure to make a flat osteotomy cut, cortical only bone block, poor bony apposition of the detached bone block- particularly at the location of any transverse plane cut, and failure to minimize thermal damage through copious irrigation) can increase nonunion risk.

  • Postoperative rehabilitation should include a 6-week period of limited weight-bearing.

  • Reconstruction of the MPFL should be performed after any TTO is performed.

Patellar instability is the result of numerous anatomical factors, including trochlear dysplasia,1,2 patella alta,2-4 and increased tibial tubercle-trochlear groove (TT-TG) or tibial tubercle-posterior cruciate ligament distance.2,5 Of all the factors, TT-TG distance and the medial patellofemoral ligament (MPFL) have received the ost attention. Patellar height remains a crucial yet underappreciated contributor that is amenable to surgical correction with tibial tubercle distalization osteotomy (TTDO). The obvious question is how severe patella alta must be to require surgical correction. In other words, when is patella alta so severe that isolated MPFL reconstruction is insufficient to restore patellar stability?

The indications for TTDO are not completely clear and depend on multiple factors. Patient factors, physical examination findings, and radiographic measures must be considered. In general, adding TTDO to MPFL reconstruction should be considered when the degree of patella alta exceeds 1.4 on the Caton-Deschamps Index (CDI). Presence of trochlear dysplasia, patellar maltracking (J-sign), lateral patellar apprehension that persists at higher flexion angles, and decreased patellotrochlear articular cartilage contact on sagittal magnetic resonance imaging may drive the decision to proceed with TTDO when the CDI is lower.

Why You Need To Know About Patella Alta

Recurrent lateral patellar dislocation is a debilitating knee condition that often involves young, active patients and significantly affects their quality of life. The MPFL is a primary restraint to lateral patellar dislocation, and an MPFL injury is a key contributor to loss of patellar stability. MPFL reconstruction is increasingly being performed to treat recurrent lateral patellar instability.6 Patellar instability is the result of numerous anatomical factors, including trochlear dysplasia,1,2 patella alta,2-4 and increased TT-TG distance.2,5 This review focuses on patella alta.

The classic teaching of the Lyon School of Knee Surgery in France, the menu à la carte, is that patella alta exceeding 1.2 on the CDI is an indication for TTDO.2 Although this teaching is an excellent guide for normal anatomy, we must keep in mind that the classic surgical menu does not consider the influence of MPFL reconstruction, as development of the menu predated this surgical option. At that time, the proximal soft-tissue procedures included vastus medialis obliquus plasty and advancement, which are performed to balance soft tissues and treat patellar tilt. These procedures and MPFL reconstruction have different functions, and the difference may be important. Furthermore, performing TTDO alongside MPFL reconstruction significantly increases the risk of complications and alters the rehabilitation protocol. However, significant untreated patella alta has been implicated as a cause of failure of isolated MPFL reconstruction.7 Establishing when MPFL reconstruction alone is sufficient is therefore crucial in avoiding the increased morbidity associated with the addition of TTDO.

Discussion

Above a certain degree of patella alta, isolated MPFL reconstruction fails to restore patellar stability. What remains unknown is the appropriate CDI cutoff (1.4) and whether the same cutoff can be used for all patients. In 2013, Wagner and colleagues8 assessed the influence of patella alta on isolated MPFL reconstruction outcomes and found no significant difference, though their study did not include many patients with significant alta and was underpowered. In 2014, Feller and colleagues9 reported on a series of patients who were successfully treated with isolated MPFL reconstruction despite patella alta significantly exceeding the traditional CDI cutoff of 1.2. Their indication for performing the isolated procedure was normal patellar tracking—in particular, absence of the J-sign. Further analysis of these patients revealed a preponderance of relatively normal TT-TG distances and low-grade, if any, trochlear dysplasia in comparison with other patients treated with a combination of MPFL reconstruction and tibial tubercle osteotomy.

Together, the work of Wagner and colleagues8 and Feller and colleagues9 suggests the historical use of the CDI of 1.2 as a hard and fast indication for adding TTDO is aggressive. In fact, it is probably the case that there really is no single CDI cutoff that is an appropriate indication for adding TTDO in all patients with instability. This decision is, and should be, influenced by a multitude of other factors, including other anatomical factors, physical examination findings, patient factors, and, of course, patient preference.

An interesting idea to consider in treating patellar instability is the interplay of patella alta and trochlear dysplasia. Patella alta is theorized as contributing to patellar instability in part by delaying entry of the patella into the TG as the knee flexes, therefore requiring less force to laterally displace

the patella.10 Similarly, in the setting of trochlear dysplasia, a shallow TG leads to less bony constraint of the patella, particularly in the groove’s superior portions, which are more involved in lower grade dysplasia. Because trochlear dysplasia and patella alta decrease patellar stability by similar mechanisms, they clearly interact, and a patient with both is at higher risk for instability than a patient who exhibits either in isolation.11 Therefore, trochlear dysplasia, particularly higher grade, may be an indication for adding TTDO at lower CDI.

Other imaging and physical examination factors can provide additional insight into the process of patellar engagement into the trochlea in each patient. The patellotrochlear index (PTI) directly measures the relationship between the patella and the trochlea, rather than relative to the tibia, as with other measures of patellar height.12[[{"fid":"201853","view_mode":"medstat_image_flush_left","attributes":{"class":"media-element file-medstat-image-flush-left","data-delta":"1"},"fields":{"format":"medstat_image_flush_left","field_file_image_caption[und][0][value]":"Figure 1.","field_file_image_credit[und][0][value]":"","field_file_image_caption[und][0][format]":"plain_text","field_file_image_credit[und][0][format]":"plain_text"},"type":"media","field_deltas":{"1":{"format":"medstat_image_flush_left","field_file_image_caption[und][0][value]":"Figure 1.","field_file_image_credit[und][0][value]":""}}}]]The PTI is correlated with tibia-based measures of height, but the correlation is not perfect. Lower degrees of overlap between the patella and the trochlea (PTI <0.15) and significant functional patella alta may warrant adding TTDO in cases of borderline CDI (1.2-1.4). Figures 1A, 1B and 2A, 2B show the imaging of 2 patients with relatively similar patellar height (assessed with CDI) but quite different degrees of overlap between the patella and trochlea. [[{"fid":"201854","view_mode":"medstat_image_flush_right","attributes":{"class":"media-element file-medstat-image-flush-right","data-delta":"2"},"fields":{"format":"medstat_image_flush_right","field_file_image_caption[und][0][value]":"Figure 2.","field_file_image_credit[und][0][value]":"","field_file_image_caption[und][0][format]":"plain_text","field_file_image_credit[und][0][format]":"plain_text"},"type":"media","field_deltas":{"2":{"format":"medstat_image_flush_right","field_file_image_caption[und][0][value]":"Figure 2.","field_file_image_credit[und][0][value]":""}}}]]The patient with less overlap is more likely to have delayed patellar engagement and symptomatic patella alta and thus may be a poorer candidate for isolated MPFL reconstruction. For additional information, please refer to the work by Roland Biedert, MD, who has proposed trochlear lengthening in these situations.13

Physical examination (even in the era of advance imaging) continues to provide useful insight into whether to add TTDO. One physical examination test that can help in understanding patellar-
trochlear dynamics is the patellar apprehension and relief test. Patellar apprehension has been widely discussed, but equally important is the degree of knee flexion above which apprehension dissipates. As patella alta and trochlear dysplasia become more severe, more knee flexion is required to relieve apprehension. Apprehension that is relieved at 30° to 40° of flexion suggests that patellar stability stands a good chance of being restored with isolated MPFL reconstruction, whereas persistent instability >45° or especially 60° of knee flexion suggests that there is significant patella alta, trochlear dysplasia, or both and that TTDO should be added. A large J-sign during knee flexion and extension provides further evidence that entry of the patella into the TG is delayed, typically because of patella alta, trochlear dysplasia, or both, and possibly tight lateral structures or a lateralized tibial tubercle. This sign is another clue that isolated MPFL reconstruction may be insufficient to completely restore patellar stability.

References

1. Dejour H, Walch G, Neyret P, Adeleine P. Dysplasia of the femoral trochlea [in French]. Rev Chir Orthop Reparatrice Appar Mot. 1990;76(1):45-54.

2. Dejour H, Walch G, Nove-Josserand L, Guier C. Factors of patellar instability: an anatomic radiographic study. Knee Surg Sports Traumatol Arthrosc. 1994;2(1):19-26.

3. Geenen E, Molenaers G, Martens M. Patella alta in patellofemoral instability. Acta Orthop Belg. 1989;55(3):387-393.

4. Simmons E Jr, Cameron JC. Patella alta and recurrent dislocation of the patella. Clin Orthop Relat Res. 1992;(274):265-269.

5. Goutallier D, Bernageau J, Lecudonnec B. The measurement of the tibial tuberosity. Patella groove distanced technique and results (author’s transl) [in French]. Rev Chir Orthop Reparatrice Appar Mot. 1978;64(5):423-428.

6. Feller JA, Lind M, Nelson J, Diduch DR, Arendt E. Repair and reconstruction of the medial patellofemoral ligament for treatment of lateral patellar dislocations. In: Scott WN, ed. Insall & Scott—Surgery of the Knee. 5th ed. Philadelphia, PA: Churchill Livingstone; 2011:677-687.

7. Thaunat M, Erasmus PJ. Recurrent patellar dislocation after medial patellofemoral ligament reconstruction. Knee Surg Sports Traumatol Arthrosc. 2008;16(1):40-43.

8. Wagner D, Pfalzer F, Hingelbaum S, Huth J, Mauch F, Bauer G. The influence of risk factors on clinical outcomes following anatomical medial patellofemoral ligament (MPFL) reconstruction using the gracilis tendon. Knee Surg Sports Traumatol Arthrosc. 2013;21(2):318-324.

9. Feller JA, Richmond AK, Wasiak J. Medial patellofemoral ligament reconstruction as an isolated or combined procedure for recurrent patellar instability. Knee Surg Sports Traumatol Arthrosc. 2014;22(10):2470-2476.

10. Ward SR, Terk MR, Powers CM. Patella alta: association
with patellofemoral alignment and changes in contact area during weight-bearing. J Bone Joint Surg Am. 2007;89(8):
1749-1755.

11. Lewallen L, McIntosh A, Dahm D. First-time patellofemoral dislocation: risk factors for recurrent instability. J Knee Surg. 2015;28(4):303-309

12. Biedert RM, Albrecht S. The patellotrochlear index: a new index for assessing patellar height. Knee Surg Sports Traumatol Arthrosc. 2006;14(8):707-712.

13. Biedert RM. Trochlear lengthening osteotomy with or without elevation of the lateral trochlear facet. In: Zaffagnini S, Dejour D, Arendt EA, eds. Patellofemoral Pain, Instability, and Arthritis. Germany: Springer-Verlag Berlin Heidelberg; 2010:
209-215.

References

1. Dejour H, Walch G, Neyret P, Adeleine P. Dysplasia of the femoral trochlea [in French]. Rev Chir Orthop Reparatrice Appar Mot. 1990;76(1):45-54.

2. Dejour H, Walch G, Nove-Josserand L, Guier C. Factors of patellar instability: an anatomic radiographic study. Knee Surg Sports Traumatol Arthrosc. 1994;2(1):19-26.

3. Geenen E, Molenaers G, Martens M. Patella alta in patellofemoral instability. Acta Orthop Belg. 1989;55(3):387-393.

4. Simmons E Jr, Cameron JC. Patella alta and recurrent dislocation of the patella. Clin Orthop Relat Res. 1992;(274):265-269.

5. Goutallier D, Bernageau J, Lecudonnec B. The measurement of the tibial tuberosity. Patella groove distanced technique and results (author’s transl) [in French]. Rev Chir Orthop Reparatrice Appar Mot. 1978;64(5):423-428.

6. Feller JA, Lind M, Nelson J, Diduch DR, Arendt E. Repair and reconstruction of the medial patellofemoral ligament for treatment of lateral patellar dislocations. In: Scott WN, ed. Insall & Scott—Surgery of the Knee. 5th ed. Philadelphia, PA: Churchill Livingstone; 2011:677-687.

7. Thaunat M, Erasmus PJ. Recurrent patellar dislocation after medial patellofemoral ligament reconstruction. Knee Surg Sports Traumatol Arthrosc. 2008;16(1):40-43.

8. Wagner D, Pfalzer F, Hingelbaum S, Huth J, Mauch F, Bauer G. The influence of risk factors on clinical outcomes following anatomical medial patellofemoral ligament (MPFL) reconstruction using the gracilis tendon. Knee Surg Sports Traumatol Arthrosc. 2013;21(2):318-324.

9. Feller JA, Richmond AK, Wasiak J. Medial patellofemoral ligament reconstruction as an isolated or combined procedure for recurrent patellar instability. Knee Surg Sports Traumatol Arthrosc. 2014;22(10):2470-2476.

10. Ward SR, Terk MR, Powers CM. Patella alta: association
with patellofemoral alignment and changes in contact area during weight-bearing. J Bone Joint Surg Am. 2007;89(8):
1749-1755.

11. Lewallen L, McIntosh A, Dahm D. First-time patellofemoral dislocation: risk factors for recurrent instability. J Knee Surg. 2015;28(4):303-309

12. Biedert RM, Albrecht S. The patellotrochlear index: a new index for assessing patellar height. Knee Surg Sports Traumatol Arthrosc. 2006;14(8):707-712.

13. Biedert RM. Trochlear lengthening osteotomy with or without elevation of the lateral trochlear facet. In: Zaffagnini S, Dejour D, Arendt EA, eds. Patellofemoral Pain, Instability, and Arthritis. Germany: Springer-Verlag Berlin Heidelberg; 2010:
209-215.

Issue
The American Journal of Orthopedics - 46(5)
Issue
The American Journal of Orthopedics - 46(5)
Page Number
229-231
Page Number
229-231
Publications
Publications
Topics
Article Type
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Article PDF Media