Smoking Prevalence and Nicotine Patch Success Rate Within a VA Medical Center

Article Type
Changed
Tue, 12/13/2016 - 12:08
Display Headline
Smoking Prevalence and Nicotine Patch Success Rate Within a VA Medical Center

Article PDF
Author and Disclosure Information

Martin Hahn, PharmD; Jennifer Bean, PharmD, BCPS; M. Shawn McFarland, PharmD, BC-ADM; and Wes Carnahan, PharmD, BCPS

At the time of this study, Dr. Hahn was a PGY-1 pharmacy practice resident at the VA Tennessee Valley Healthcare System (VATVHS), Nashville. He is now a clinical/staff pharmacist at Skyline Medical Center, Nashville, Tennessee. Dr. Bean is a clinical pharmacy specialist in home-based primary care and mental health services, Dr. McFarland is a clinical pharmacy specialist in primary care, and Dr. Carnahan is a clinical pharmacy specialist in oncology and formulary management, all at the VATVHS. In addition, Drs. Bean and Carnahan are assistant professors and Dr. McFarland is an associate professor, all at the University of Tennessee College of Pharmacy.

Issue
Federal Practitioner - 27(10)
Publications
Topics
Page Number
14
Legacy Keywords
cigarette, smoke, nicotine, patch, cardiovascular, chronic obstructive pulmonary disease, lung cancer, myocardial infarction, Agency for Health Care Policy and Researchcigarette, smoke, nicotine, patch, cardiovascular, chronic obstructive pulmonary disease, lung cancer, myocardial infarction, Agency for Health Care Policy and Research
Sections
Author and Disclosure Information

Martin Hahn, PharmD; Jennifer Bean, PharmD, BCPS; M. Shawn McFarland, PharmD, BC-ADM; and Wes Carnahan, PharmD, BCPS

At the time of this study, Dr. Hahn was a PGY-1 pharmacy practice resident at the VA Tennessee Valley Healthcare System (VATVHS), Nashville. He is now a clinical/staff pharmacist at Skyline Medical Center, Nashville, Tennessee. Dr. Bean is a clinical pharmacy specialist in home-based primary care and mental health services, Dr. McFarland is a clinical pharmacy specialist in primary care, and Dr. Carnahan is a clinical pharmacy specialist in oncology and formulary management, all at the VATVHS. In addition, Drs. Bean and Carnahan are assistant professors and Dr. McFarland is an associate professor, all at the University of Tennessee College of Pharmacy.

Author and Disclosure Information

Martin Hahn, PharmD; Jennifer Bean, PharmD, BCPS; M. Shawn McFarland, PharmD, BC-ADM; and Wes Carnahan, PharmD, BCPS

At the time of this study, Dr. Hahn was a PGY-1 pharmacy practice resident at the VA Tennessee Valley Healthcare System (VATVHS), Nashville. He is now a clinical/staff pharmacist at Skyline Medical Center, Nashville, Tennessee. Dr. Bean is a clinical pharmacy specialist in home-based primary care and mental health services, Dr. McFarland is a clinical pharmacy specialist in primary care, and Dr. Carnahan is a clinical pharmacy specialist in oncology and formulary management, all at the VATVHS. In addition, Drs. Bean and Carnahan are assistant professors and Dr. McFarland is an associate professor, all at the University of Tennessee College of Pharmacy.

Article PDF
Article PDF

Issue
Federal Practitioner - 27(10)
Issue
Federal Practitioner - 27(10)
Page Number
14
Page Number
14
Publications
Publications
Topics
Article Type
Display Headline
Smoking Prevalence and Nicotine Patch Success Rate Within a VA Medical Center
Display Headline
Smoking Prevalence and Nicotine Patch Success Rate Within a VA Medical Center
Legacy Keywords
cigarette, smoke, nicotine, patch, cardiovascular, chronic obstructive pulmonary disease, lung cancer, myocardial infarction, Agency for Health Care Policy and Researchcigarette, smoke, nicotine, patch, cardiovascular, chronic obstructive pulmonary disease, lung cancer, myocardial infarction, Agency for Health Care Policy and Research
Legacy Keywords
cigarette, smoke, nicotine, patch, cardiovascular, chronic obstructive pulmonary disease, lung cancer, myocardial infarction, Agency for Health Care Policy and Researchcigarette, smoke, nicotine, patch, cardiovascular, chronic obstructive pulmonary disease, lung cancer, myocardial infarction, Agency for Health Care Policy and Research
Sections
Article Source

PURLs Copyright

Inside the Article

Article PDF Media

Investigating Infection in the Patient With RA

Article Type
Changed
Tue, 12/13/2016 - 12:08
Display Headline
Investigating Infection in the Patient With RA
Blood Cultures Needed for Those Taking Anti-TNF Therapy

In 2009, a question of proper practices was brought before the Arizona Regulatory Board of Physician Assistants. The patient in question was seen in an emergency department by a PA/physician health care team for complaints of cough and fever at home. In question was the team’s decision to discharge the patient, who had a history of immunosuppression with an anti–tumor necrosis factor (anti-TNF) medication for a chronic joint disorder, without ordering laboratory tests or blood cultures. Although there is textbook instruction for evaluation of patients with infection who use agents in this drug class,1 many clinicians may be unaware that blood cultures should be drawn as part of a proper workup.

In the United States, medications that have anti-TNF-alpha ­(anti-TNF-a) properties, including adalimumab, etanercept, and infliximab, are approved by the FDA to treat rheumatoid arthritis (RA).2 Many patients take a second medication; others, a combination of medications. Options ­include methotrexate, cyclo­sporine, prednisone, and hydroxychloroquine. Typically, anti-TNF medications represent second- or third-line therapy after unsatisfactory response to disease-modifying antirheumatic drugs (DMARDs).

Although anti-TNF medications are effective for treatment of autoimmune diseases like RA, it should be noted that TNF-a is an important component in the proper functioning of the immune system. Accordingly, it is recognized that patients who use anti-TNF medications are at increased risk for infection.3 Indeed, patients with RA are already at greater risk for contracting infectious illnesses, even before immunosuppressive medications are administered.4

TNF is one cytokine that is responsible for humoral activation and subsequent inflammation.5 In RA and certain other diseases (eg, Crohn’s disease, lupus), TNF-a’s interaction with other cytokines is at least partially responsible for the inflammation that leads to RA-associated joint damage.5 In­fliximab and adalimumab are monoclonal antibodies developed specifically to bind with TNF-a, thereby neutralizing a pathway for further release of inflammatory cytokines. 

In contrast to recombinant soluble receptors, such as etanercept, the site-specific monoclonal antibodies for TNF-a (ie, infliximab, adalimumab) may play a greater role in suppressing macrophages, which contribute significantly to cellular immunity. Macrophage inhibition may further increase the body’s vulnerability to opportunistic infection.6

Anti-TNF Agents: Clinical Trials

The relationship between treatment with immunosuppressive agents (including infliximab and other anti-TNF medications) and serious infection is evident in the literature. In a 2005 study, Listing et al4 followed patients for 12 months after they were started on etanercept, infliximab, anakinra (another cytokine inhibitor), or DMARDs (controls). Ninety-two (26.5%) of the 346 patients who were given infliximab, compared with 6% of 601 controls, developed infection; of these, 20 patients (21.7%) had a determinable serious infection (ie, that was considered life-threatening or resulted in inpatient treatment, significant disability, or death). Thus, serious infection affected 5.8% of the patient population studied who had received infliximab.

Five of the 512 patients who used etanercept were diagnosed with sepsis, but no episodes of bacteremia were recorded among the infliximab patients. The most prevalent serious infections observed were pneumonia, infective arthritis, and various skin and subcutaneous infections (see table4). Less prevalent respiratory tract ailments included bronchitis, lung abscess, and pleural infection. Overall, small numbers of cases of sepsis were reported, making it difficult for the study authors to comment on the disparity of sepsis rates among treatment groups.4

Another prospective study followed 28 patients (median age, 53) who were treated with infliximab between 1999 and 2002.7 Dosages administered ranged from the standard 3 mg/kg every 8 weeks to 10 mg/kg every 4 weeks, with variation in duration of treatment. A 13% higher rate of infection was ­reported among these patients, compared with patients who received conventional therapy (DMARDs) over a two-year ­period.

Six patients were determined to have serious, life-threatening infections (ie, requiring hospitalization or IV antibiotic therapy). Of these, two were given a diagnosis of bacteremia via blood cultures. The cultures grew Streptococcus pneumoniae in one patient, who was also diagnosed incidentally with Pneumocystis carinii through bronchial washings, and the other patient had blood cultures positive for Staphylococcus aureus and bilateral necrotizing pulmonary abscesses.

Case Studies

Individual case studies of infection in patients taking infliximab or other anti-TNF agents for RA reveal specific illnesses, their time lines, and relevant patient management. Crum et al2 reported an example of the common pathogen S pneumoniae in a 47-year-old patient who developed a left lower lobe pneumonia. Three of four blood cultures were positive, CBC with differential showed abnormal results, and the patient’s temperature on presentation was 39.5°C.

In a report of two cases of severe abdominal infection during administration of anti-TNF medication, one 40-year-old woman using infliximab for severe RA presented with a temperature of 39.2°C and a three-day history of abdominal pain. Escherichia coli was identified from blood cultures, although urine cultures were negative. CT led to a diagnosis of pyelonephritis. The patient had become septic but improved after 48 hours’ therapy with IV cefuroxime and gentamicin.8

 

 

The second patient was a man, age 60, with a two-week history of abdominal pain and a six-month history of effective etanercept use for persistent psoriatic arthritis. Contrast-­enhanced CT revealed a large splenic abscess, and blood cultures identified infection with S aureus. Management began with unsuccessful high-dose IV antibiotics, followed by laparotomy and splenectomy, postsurgical sepsis, and finally, high-dose inotropic therapy that proved effective.8

Infection With Listeria monocytogenes

Listeria monocytogenes, a pathogen sometimes identified in patients who receive anti-TNF-a therapy, has been the focus of several case reports. For example, Gluck et al9 describe a 60-year-old woman with a history of RA and previous treatment with multiple therapies who had recently completed a five-month regimen with infliximab (10 mg/kg every 4 weeks). Fourteen days after receiving her last dose, she presented to a hospital with anemia, fever (measurement undisclosed), and abdominal pain. She was given multiple diagnoses, including a gastric ulcer without hemorrhage, a pulmonary infiltrate, and acute cholecystitis. After she underwent cholecystectomy, her condition deteriorated, and head CT revealed several small subarachnoid hemorrhages and severe brain edema.

The patient experienced multi–organ system failure and died after a brain-stem herniation. What should be noted is that a blood culture taken one day after her surgery was positive for L monocytogenes, as was an intraoperative gallbladder swab-culture for the same organism—this, despite presumed antibiotic therapy.

A second patient described by Gluck et al9 was a 62-year-old woman with RA who developed cholecystitis after her second dose of infliximab, administered over a two-week period (200 mg IV/dose). As in the previous case, the patient manifested cerebral edema and had a fever after undergoing cholecystectomy. Blood cultures performed after the surgery were positive for L monocytogenes. The patient recovered slowly with proper antibiotic therapy, including ampicillin and gentamicin.

A third example of listeriosis involved a 52-year-old patient with RA who presented with symptoms including fever and abdominal pain. CT led to a diagnosis of terminal ileitis. One of two blood cultures taken was positive for L monocytogenes.6

Finally, a 79-year-old patient with a long-standing diagnosis of RA and prosthetic hips reported a fever of three weeks’ duration and left leg pain. As the patient had no fever on initial presentation, blood cultures were not drawn until five days after he was admitted. Three of four blood cultures were positive for L monocytogenes.6

Less Common Infections

Other less common bacterial etiologies have also been reported in the literature, including a fatal case of Salmonella enteritidis septicemia with a pleural empyema after treatment with infliximab.10 Another fatality was reported in a 54-year-old man who had necrotizing fasciitis, with a group A hemolytic streptococcus that grew in blood cultures. The patient, who had been taking in­fliximab, had no report of fever before or during his hospitalization.11

Discussion

Although infliximab is effective for controlling the symptoms of RA, clinicians must be attentive to fever in patients who use it—even a patient-­reported fever that may not be present on physical exam. Due to the potential blunting of the immune response associated with infliximab use, a serious underlying infection can occur without fever. General malaise may be the only symptom a patient reports.

Infliximab and other anti-TNF medications are included in a textbook list of immunosuppressive medications whose use warrants special attention for patients who are evaluated for infection in an emergency setting. According to Burns,1 “Rapid diagnosis and early initiation of therapy are essential in preventing serious morbidity and mortality in immunocompromised patients who often have subtle or unusual presentations and are difficult to ­diagnose.”1

Active tuberculosis (ATB) is another serious illness that has been reported with infliximab use.12,13 Patients should be screened for TB before taking infliximab, but clinicians must also be aware that ATB can develop as an unwanted result of infliximab therapy. Standard blood cultures may not be an effective means of identifying ATB.

Several types of infection may occur, with or without confirmation by positive blood cultures, including viral and fungal infections. Even bacterial infections, such as a urinary tract infection or bacterial pneumonia, may not yield positive blood cultures immediately, if ever. Each of these has been included among serious illnesses associated with use of anti-TNF medications.4 Nevertheless, blood cultures—Gram-stain, anaerobic, and aerobic blood cultures with sensitivities—should be considered an important component of any comprehensive infectious disease evaluation.

Although the purpose of this article has been to emphasize the importance of blood cultures in the timely diagnosis and treatment of patients taking ­anti-TNF medications who present with symptoms of infectious disease, it is important to remember that blood cultures are only part of a full infectious disease workup.

 

 

Conclusion

The clinical studies and case presentations discussed here serve to remind clinicians that timely acquisition and evaluation of blood cultures are essential to the identification of serious bacteriologic pathogens, and to the chances of recovery in the RA patient with a systemic infection who is immunosuppressed as a result of treatment with infliximab or other anti-TNF agent. Rapid identification of pathogenic organisms will facilitate the implementation of appropriate therapy, thus avoiding the often-deadly outcomes of severe infection and sepsis. Gram-stain, anaerobic, and aerobic blood cultures with sensitivities should be considered the standard of care in any infliximab-treated patient who presents with fever, a patient report of a fever, or any other signs, symptoms, or patient history that could suggest infection.

While this article focuses for the most part on infections associated with infliximab use, the same strategy in standard of practice should apply to all patients who use anti-TNF therapy for treatment of RA.

References

1. Burns MJ. Chapter 181. Other immunosuppressive medications. In: Marx J, Hockberger R, Walls R, et al, eds. Rosen’s Emergency Medicine: Concepts and Clinical Practice. 7th ed. Philadelphia, PA: Mosby Elsevier; 2009.

2. Crum NF, Lederman ER, Wallace MR. Infections associated with tumor necrosis factor-alpha antagonists. Medicine (Baltimore). 2005; 84(5)291-302.

3. Geraghty EM, Ristow B, Gordon SM, Aronowitz P. Overwhelming parasitemia with Plasmodium falciparum infection in a patient receiving infliximab therapy for rheumatoid arthritis. Clin Infect Dis. 2007;44(10):82-84.

4. Listing J, Strangfeld A, Kary S, et al. Infections in patients with rheumatoid arthritis treated with biologic agents. Arthritis Rheum. 2005;52(11):3403-3412.

5. Louie SG, Park B, Yoon H. Biological response modifiers in the management of rheumatoid arthritis. Am J Health Syst Pharm. 2003;60(4): 346-355.

6. Kesteman T, Yombi JC, Gigi J, Durez P. Listeria infections associated with infliximab: case reports. Clin Rheumatol. 2007;26(12):2173-2175.

7. Kroesen S, Widmer AF, Tyndall A, Hasler P. Serious bacterial infections in patients with rheumatoid arthritis under anti-TNF-alpha therapy. Rheumatology. 2003;42(5):617-621.

8. Goode S, Tierney G, Deighton C. Life threatening intra-abdominal sepsis in patients on anti-TNF-alpha therapy. Gut. 2005;54(4):590-591. 

9. Glück T, Linde HJ, Schölmerich J, et al. Anti-tumor necrosis factor therapy and Listeria monocytogenes infection: report of two cases. Arthritis Rheum. 2002;46(8):2255-2257.

10. Rijkeboer A, Voskuyl A, Van Agtmael M. Fatal Salmonella enteritidis septicaemia in a rheumatoid arthritis patient treated with a TNF-alpha antagonist. Scand J Infect Dis. 2007; 39(1):80-83.

11. Chan AT, Cleeve V, Daymond TJ. Necrotising fasciitis in a patient receiving infliximab for rheumatoid arthritis. Postgrad Med J. 2002; 78(915):47-48.

12. Favalli EG, Desiati F, Atzeni F, et al. Serious infections during anti-TNF-alpha treatment in rheumatoid arthritis patients. Autoimmun Rev. 2009;8(3):266-273.

13. Keane J, Gershon S, Wise RP, et al. Tuberculosis associated with infliximab, a tumor necrosis factor alpha-neutralizing agent. N Engl J Med. 2001;345(15):1098-1104.

Author and Disclosure Information

Geoffrey W. Hoffa, PA-C

Issue
Clinician Reviews - 20(10)
Publications
Topics
Page Number
38-43
Legacy Keywords
rheumatoid arthritis, infection, blood cultures, anti-tumor necrosis factor, infliximabrheumatoid arthritis, infection, blood cultures, anti-tumor necrosis factor, infliximab
Sections
Author and Disclosure Information

Geoffrey W. Hoffa, PA-C

Author and Disclosure Information

Geoffrey W. Hoffa, PA-C

Blood Cultures Needed for Those Taking Anti-TNF Therapy
Blood Cultures Needed for Those Taking Anti-TNF Therapy

In 2009, a question of proper practices was brought before the Arizona Regulatory Board of Physician Assistants. The patient in question was seen in an emergency department by a PA/physician health care team for complaints of cough and fever at home. In question was the team’s decision to discharge the patient, who had a history of immunosuppression with an anti–tumor necrosis factor (anti-TNF) medication for a chronic joint disorder, without ordering laboratory tests or blood cultures. Although there is textbook instruction for evaluation of patients with infection who use agents in this drug class,1 many clinicians may be unaware that blood cultures should be drawn as part of a proper workup.

In the United States, medications that have anti-TNF-alpha ­(anti-TNF-a) properties, including adalimumab, etanercept, and infliximab, are approved by the FDA to treat rheumatoid arthritis (RA).2 Many patients take a second medication; others, a combination of medications. Options ­include methotrexate, cyclo­sporine, prednisone, and hydroxychloroquine. Typically, anti-TNF medications represent second- or third-line therapy after unsatisfactory response to disease-modifying antirheumatic drugs (DMARDs).

Although anti-TNF medications are effective for treatment of autoimmune diseases like RA, it should be noted that TNF-a is an important component in the proper functioning of the immune system. Accordingly, it is recognized that patients who use anti-TNF medications are at increased risk for infection.3 Indeed, patients with RA are already at greater risk for contracting infectious illnesses, even before immunosuppressive medications are administered.4

TNF is one cytokine that is responsible for humoral activation and subsequent inflammation.5 In RA and certain other diseases (eg, Crohn’s disease, lupus), TNF-a’s interaction with other cytokines is at least partially responsible for the inflammation that leads to RA-associated joint damage.5 In­fliximab and adalimumab are monoclonal antibodies developed specifically to bind with TNF-a, thereby neutralizing a pathway for further release of inflammatory cytokines. 

In contrast to recombinant soluble receptors, such as etanercept, the site-specific monoclonal antibodies for TNF-a (ie, infliximab, adalimumab) may play a greater role in suppressing macrophages, which contribute significantly to cellular immunity. Macrophage inhibition may further increase the body’s vulnerability to opportunistic infection.6

Anti-TNF Agents: Clinical Trials

The relationship between treatment with immunosuppressive agents (including infliximab and other anti-TNF medications) and serious infection is evident in the literature. In a 2005 study, Listing et al4 followed patients for 12 months after they were started on etanercept, infliximab, anakinra (another cytokine inhibitor), or DMARDs (controls). Ninety-two (26.5%) of the 346 patients who were given infliximab, compared with 6% of 601 controls, developed infection; of these, 20 patients (21.7%) had a determinable serious infection (ie, that was considered life-threatening or resulted in inpatient treatment, significant disability, or death). Thus, serious infection affected 5.8% of the patient population studied who had received infliximab.

Five of the 512 patients who used etanercept were diagnosed with sepsis, but no episodes of bacteremia were recorded among the infliximab patients. The most prevalent serious infections observed were pneumonia, infective arthritis, and various skin and subcutaneous infections (see table4). Less prevalent respiratory tract ailments included bronchitis, lung abscess, and pleural infection. Overall, small numbers of cases of sepsis were reported, making it difficult for the study authors to comment on the disparity of sepsis rates among treatment groups.4

Another prospective study followed 28 patients (median age, 53) who were treated with infliximab between 1999 and 2002.7 Dosages administered ranged from the standard 3 mg/kg every 8 weeks to 10 mg/kg every 4 weeks, with variation in duration of treatment. A 13% higher rate of infection was ­reported among these patients, compared with patients who received conventional therapy (DMARDs) over a two-year ­period.

Six patients were determined to have serious, life-threatening infections (ie, requiring hospitalization or IV antibiotic therapy). Of these, two were given a diagnosis of bacteremia via blood cultures. The cultures grew Streptococcus pneumoniae in one patient, who was also diagnosed incidentally with Pneumocystis carinii through bronchial washings, and the other patient had blood cultures positive for Staphylococcus aureus and bilateral necrotizing pulmonary abscesses.

Case Studies

Individual case studies of infection in patients taking infliximab or other anti-TNF agents for RA reveal specific illnesses, their time lines, and relevant patient management. Crum et al2 reported an example of the common pathogen S pneumoniae in a 47-year-old patient who developed a left lower lobe pneumonia. Three of four blood cultures were positive, CBC with differential showed abnormal results, and the patient’s temperature on presentation was 39.5°C.

In a report of two cases of severe abdominal infection during administration of anti-TNF medication, one 40-year-old woman using infliximab for severe RA presented with a temperature of 39.2°C and a three-day history of abdominal pain. Escherichia coli was identified from blood cultures, although urine cultures were negative. CT led to a diagnosis of pyelonephritis. The patient had become septic but improved after 48 hours’ therapy with IV cefuroxime and gentamicin.8

 

 

The second patient was a man, age 60, with a two-week history of abdominal pain and a six-month history of effective etanercept use for persistent psoriatic arthritis. Contrast-­enhanced CT revealed a large splenic abscess, and blood cultures identified infection with S aureus. Management began with unsuccessful high-dose IV antibiotics, followed by laparotomy and splenectomy, postsurgical sepsis, and finally, high-dose inotropic therapy that proved effective.8

Infection With Listeria monocytogenes

Listeria monocytogenes, a pathogen sometimes identified in patients who receive anti-TNF-a therapy, has been the focus of several case reports. For example, Gluck et al9 describe a 60-year-old woman with a history of RA and previous treatment with multiple therapies who had recently completed a five-month regimen with infliximab (10 mg/kg every 4 weeks). Fourteen days after receiving her last dose, she presented to a hospital with anemia, fever (measurement undisclosed), and abdominal pain. She was given multiple diagnoses, including a gastric ulcer without hemorrhage, a pulmonary infiltrate, and acute cholecystitis. After she underwent cholecystectomy, her condition deteriorated, and head CT revealed several small subarachnoid hemorrhages and severe brain edema.

The patient experienced multi–organ system failure and died after a brain-stem herniation. What should be noted is that a blood culture taken one day after her surgery was positive for L monocytogenes, as was an intraoperative gallbladder swab-culture for the same organism—this, despite presumed antibiotic therapy.

A second patient described by Gluck et al9 was a 62-year-old woman with RA who developed cholecystitis after her second dose of infliximab, administered over a two-week period (200 mg IV/dose). As in the previous case, the patient manifested cerebral edema and had a fever after undergoing cholecystectomy. Blood cultures performed after the surgery were positive for L monocytogenes. The patient recovered slowly with proper antibiotic therapy, including ampicillin and gentamicin.

A third example of listeriosis involved a 52-year-old patient with RA who presented with symptoms including fever and abdominal pain. CT led to a diagnosis of terminal ileitis. One of two blood cultures taken was positive for L monocytogenes.6

Finally, a 79-year-old patient with a long-standing diagnosis of RA and prosthetic hips reported a fever of three weeks’ duration and left leg pain. As the patient had no fever on initial presentation, blood cultures were not drawn until five days after he was admitted. Three of four blood cultures were positive for L monocytogenes.6

Less Common Infections

Other less common bacterial etiologies have also been reported in the literature, including a fatal case of Salmonella enteritidis septicemia with a pleural empyema after treatment with infliximab.10 Another fatality was reported in a 54-year-old man who had necrotizing fasciitis, with a group A hemolytic streptococcus that grew in blood cultures. The patient, who had been taking in­fliximab, had no report of fever before or during his hospitalization.11

Discussion

Although infliximab is effective for controlling the symptoms of RA, clinicians must be attentive to fever in patients who use it—even a patient-­reported fever that may not be present on physical exam. Due to the potential blunting of the immune response associated with infliximab use, a serious underlying infection can occur without fever. General malaise may be the only symptom a patient reports.

Infliximab and other anti-TNF medications are included in a textbook list of immunosuppressive medications whose use warrants special attention for patients who are evaluated for infection in an emergency setting. According to Burns,1 “Rapid diagnosis and early initiation of therapy are essential in preventing serious morbidity and mortality in immunocompromised patients who often have subtle or unusual presentations and are difficult to ­diagnose.”1

Active tuberculosis (ATB) is another serious illness that has been reported with infliximab use.12,13 Patients should be screened for TB before taking infliximab, but clinicians must also be aware that ATB can develop as an unwanted result of infliximab therapy. Standard blood cultures may not be an effective means of identifying ATB.

Several types of infection may occur, with or without confirmation by positive blood cultures, including viral and fungal infections. Even bacterial infections, such as a urinary tract infection or bacterial pneumonia, may not yield positive blood cultures immediately, if ever. Each of these has been included among serious illnesses associated with use of anti-TNF medications.4 Nevertheless, blood cultures—Gram-stain, anaerobic, and aerobic blood cultures with sensitivities—should be considered an important component of any comprehensive infectious disease evaluation.

Although the purpose of this article has been to emphasize the importance of blood cultures in the timely diagnosis and treatment of patients taking ­anti-TNF medications who present with symptoms of infectious disease, it is important to remember that blood cultures are only part of a full infectious disease workup.

 

 

Conclusion

The clinical studies and case presentations discussed here serve to remind clinicians that timely acquisition and evaluation of blood cultures are essential to the identification of serious bacteriologic pathogens, and to the chances of recovery in the RA patient with a systemic infection who is immunosuppressed as a result of treatment with infliximab or other anti-TNF agent. Rapid identification of pathogenic organisms will facilitate the implementation of appropriate therapy, thus avoiding the often-deadly outcomes of severe infection and sepsis. Gram-stain, anaerobic, and aerobic blood cultures with sensitivities should be considered the standard of care in any infliximab-treated patient who presents with fever, a patient report of a fever, or any other signs, symptoms, or patient history that could suggest infection.

While this article focuses for the most part on infections associated with infliximab use, the same strategy in standard of practice should apply to all patients who use anti-TNF therapy for treatment of RA.

In 2009, a question of proper practices was brought before the Arizona Regulatory Board of Physician Assistants. The patient in question was seen in an emergency department by a PA/physician health care team for complaints of cough and fever at home. In question was the team’s decision to discharge the patient, who had a history of immunosuppression with an anti–tumor necrosis factor (anti-TNF) medication for a chronic joint disorder, without ordering laboratory tests or blood cultures. Although there is textbook instruction for evaluation of patients with infection who use agents in this drug class,1 many clinicians may be unaware that blood cultures should be drawn as part of a proper workup.

In the United States, medications that have anti-TNF-alpha ­(anti-TNF-a) properties, including adalimumab, etanercept, and infliximab, are approved by the FDA to treat rheumatoid arthritis (RA).2 Many patients take a second medication; others, a combination of medications. Options ­include methotrexate, cyclo­sporine, prednisone, and hydroxychloroquine. Typically, anti-TNF medications represent second- or third-line therapy after unsatisfactory response to disease-modifying antirheumatic drugs (DMARDs).

Although anti-TNF medications are effective for treatment of autoimmune diseases like RA, it should be noted that TNF-a is an important component in the proper functioning of the immune system. Accordingly, it is recognized that patients who use anti-TNF medications are at increased risk for infection.3 Indeed, patients with RA are already at greater risk for contracting infectious illnesses, even before immunosuppressive medications are administered.4

TNF is one cytokine that is responsible for humoral activation and subsequent inflammation.5 In RA and certain other diseases (eg, Crohn’s disease, lupus), TNF-a’s interaction with other cytokines is at least partially responsible for the inflammation that leads to RA-associated joint damage.5 In­fliximab and adalimumab are monoclonal antibodies developed specifically to bind with TNF-a, thereby neutralizing a pathway for further release of inflammatory cytokines. 

In contrast to recombinant soluble receptors, such as etanercept, the site-specific monoclonal antibodies for TNF-a (ie, infliximab, adalimumab) may play a greater role in suppressing macrophages, which contribute significantly to cellular immunity. Macrophage inhibition may further increase the body’s vulnerability to opportunistic infection.6

Anti-TNF Agents: Clinical Trials

The relationship between treatment with immunosuppressive agents (including infliximab and other anti-TNF medications) and serious infection is evident in the literature. In a 2005 study, Listing et al4 followed patients for 12 months after they were started on etanercept, infliximab, anakinra (another cytokine inhibitor), or DMARDs (controls). Ninety-two (26.5%) of the 346 patients who were given infliximab, compared with 6% of 601 controls, developed infection; of these, 20 patients (21.7%) had a determinable serious infection (ie, that was considered life-threatening or resulted in inpatient treatment, significant disability, or death). Thus, serious infection affected 5.8% of the patient population studied who had received infliximab.

Five of the 512 patients who used etanercept were diagnosed with sepsis, but no episodes of bacteremia were recorded among the infliximab patients. The most prevalent serious infections observed were pneumonia, infective arthritis, and various skin and subcutaneous infections (see table4). Less prevalent respiratory tract ailments included bronchitis, lung abscess, and pleural infection. Overall, small numbers of cases of sepsis were reported, making it difficult for the study authors to comment on the disparity of sepsis rates among treatment groups.4

Another prospective study followed 28 patients (median age, 53) who were treated with infliximab between 1999 and 2002.7 Dosages administered ranged from the standard 3 mg/kg every 8 weeks to 10 mg/kg every 4 weeks, with variation in duration of treatment. A 13% higher rate of infection was ­reported among these patients, compared with patients who received conventional therapy (DMARDs) over a two-year ­period.

Six patients were determined to have serious, life-threatening infections (ie, requiring hospitalization or IV antibiotic therapy). Of these, two were given a diagnosis of bacteremia via blood cultures. The cultures grew Streptococcus pneumoniae in one patient, who was also diagnosed incidentally with Pneumocystis carinii through bronchial washings, and the other patient had blood cultures positive for Staphylococcus aureus and bilateral necrotizing pulmonary abscesses.

Case Studies

Individual case studies of infection in patients taking infliximab or other anti-TNF agents for RA reveal specific illnesses, their time lines, and relevant patient management. Crum et al2 reported an example of the common pathogen S pneumoniae in a 47-year-old patient who developed a left lower lobe pneumonia. Three of four blood cultures were positive, CBC with differential showed abnormal results, and the patient’s temperature on presentation was 39.5°C.

In a report of two cases of severe abdominal infection during administration of anti-TNF medication, one 40-year-old woman using infliximab for severe RA presented with a temperature of 39.2°C and a three-day history of abdominal pain. Escherichia coli was identified from blood cultures, although urine cultures were negative. CT led to a diagnosis of pyelonephritis. The patient had become septic but improved after 48 hours’ therapy with IV cefuroxime and gentamicin.8

 

 

The second patient was a man, age 60, with a two-week history of abdominal pain and a six-month history of effective etanercept use for persistent psoriatic arthritis. Contrast-­enhanced CT revealed a large splenic abscess, and blood cultures identified infection with S aureus. Management began with unsuccessful high-dose IV antibiotics, followed by laparotomy and splenectomy, postsurgical sepsis, and finally, high-dose inotropic therapy that proved effective.8

Infection With Listeria monocytogenes

Listeria monocytogenes, a pathogen sometimes identified in patients who receive anti-TNF-a therapy, has been the focus of several case reports. For example, Gluck et al9 describe a 60-year-old woman with a history of RA and previous treatment with multiple therapies who had recently completed a five-month regimen with infliximab (10 mg/kg every 4 weeks). Fourteen days after receiving her last dose, she presented to a hospital with anemia, fever (measurement undisclosed), and abdominal pain. She was given multiple diagnoses, including a gastric ulcer without hemorrhage, a pulmonary infiltrate, and acute cholecystitis. After she underwent cholecystectomy, her condition deteriorated, and head CT revealed several small subarachnoid hemorrhages and severe brain edema.

The patient experienced multi–organ system failure and died after a brain-stem herniation. What should be noted is that a blood culture taken one day after her surgery was positive for L monocytogenes, as was an intraoperative gallbladder swab-culture for the same organism—this, despite presumed antibiotic therapy.

A second patient described by Gluck et al9 was a 62-year-old woman with RA who developed cholecystitis after her second dose of infliximab, administered over a two-week period (200 mg IV/dose). As in the previous case, the patient manifested cerebral edema and had a fever after undergoing cholecystectomy. Blood cultures performed after the surgery were positive for L monocytogenes. The patient recovered slowly with proper antibiotic therapy, including ampicillin and gentamicin.

A third example of listeriosis involved a 52-year-old patient with RA who presented with symptoms including fever and abdominal pain. CT led to a diagnosis of terminal ileitis. One of two blood cultures taken was positive for L monocytogenes.6

Finally, a 79-year-old patient with a long-standing diagnosis of RA and prosthetic hips reported a fever of three weeks’ duration and left leg pain. As the patient had no fever on initial presentation, blood cultures were not drawn until five days after he was admitted. Three of four blood cultures were positive for L monocytogenes.6

Less Common Infections

Other less common bacterial etiologies have also been reported in the literature, including a fatal case of Salmonella enteritidis septicemia with a pleural empyema after treatment with infliximab.10 Another fatality was reported in a 54-year-old man who had necrotizing fasciitis, with a group A hemolytic streptococcus that grew in blood cultures. The patient, who had been taking in­fliximab, had no report of fever before or during his hospitalization.11

Discussion

Although infliximab is effective for controlling the symptoms of RA, clinicians must be attentive to fever in patients who use it—even a patient-­reported fever that may not be present on physical exam. Due to the potential blunting of the immune response associated with infliximab use, a serious underlying infection can occur without fever. General malaise may be the only symptom a patient reports.

Infliximab and other anti-TNF medications are included in a textbook list of immunosuppressive medications whose use warrants special attention for patients who are evaluated for infection in an emergency setting. According to Burns,1 “Rapid diagnosis and early initiation of therapy are essential in preventing serious morbidity and mortality in immunocompromised patients who often have subtle or unusual presentations and are difficult to ­diagnose.”1

Active tuberculosis (ATB) is another serious illness that has been reported with infliximab use.12,13 Patients should be screened for TB before taking infliximab, but clinicians must also be aware that ATB can develop as an unwanted result of infliximab therapy. Standard blood cultures may not be an effective means of identifying ATB.

Several types of infection may occur, with or without confirmation by positive blood cultures, including viral and fungal infections. Even bacterial infections, such as a urinary tract infection or bacterial pneumonia, may not yield positive blood cultures immediately, if ever. Each of these has been included among serious illnesses associated with use of anti-TNF medications.4 Nevertheless, blood cultures—Gram-stain, anaerobic, and aerobic blood cultures with sensitivities—should be considered an important component of any comprehensive infectious disease evaluation.

Although the purpose of this article has been to emphasize the importance of blood cultures in the timely diagnosis and treatment of patients taking ­anti-TNF medications who present with symptoms of infectious disease, it is important to remember that blood cultures are only part of a full infectious disease workup.

 

 

Conclusion

The clinical studies and case presentations discussed here serve to remind clinicians that timely acquisition and evaluation of blood cultures are essential to the identification of serious bacteriologic pathogens, and to the chances of recovery in the RA patient with a systemic infection who is immunosuppressed as a result of treatment with infliximab or other anti-TNF agent. Rapid identification of pathogenic organisms will facilitate the implementation of appropriate therapy, thus avoiding the often-deadly outcomes of severe infection and sepsis. Gram-stain, anaerobic, and aerobic blood cultures with sensitivities should be considered the standard of care in any infliximab-treated patient who presents with fever, a patient report of a fever, or any other signs, symptoms, or patient history that could suggest infection.

While this article focuses for the most part on infections associated with infliximab use, the same strategy in standard of practice should apply to all patients who use anti-TNF therapy for treatment of RA.

References

1. Burns MJ. Chapter 181. Other immunosuppressive medications. In: Marx J, Hockberger R, Walls R, et al, eds. Rosen’s Emergency Medicine: Concepts and Clinical Practice. 7th ed. Philadelphia, PA: Mosby Elsevier; 2009.

2. Crum NF, Lederman ER, Wallace MR. Infections associated with tumor necrosis factor-alpha antagonists. Medicine (Baltimore). 2005; 84(5)291-302.

3. Geraghty EM, Ristow B, Gordon SM, Aronowitz P. Overwhelming parasitemia with Plasmodium falciparum infection in a patient receiving infliximab therapy for rheumatoid arthritis. Clin Infect Dis. 2007;44(10):82-84.

4. Listing J, Strangfeld A, Kary S, et al. Infections in patients with rheumatoid arthritis treated with biologic agents. Arthritis Rheum. 2005;52(11):3403-3412.

5. Louie SG, Park B, Yoon H. Biological response modifiers in the management of rheumatoid arthritis. Am J Health Syst Pharm. 2003;60(4): 346-355.

6. Kesteman T, Yombi JC, Gigi J, Durez P. Listeria infections associated with infliximab: case reports. Clin Rheumatol. 2007;26(12):2173-2175.

7. Kroesen S, Widmer AF, Tyndall A, Hasler P. Serious bacterial infections in patients with rheumatoid arthritis under anti-TNF-alpha therapy. Rheumatology. 2003;42(5):617-621.

8. Goode S, Tierney G, Deighton C. Life threatening intra-abdominal sepsis in patients on anti-TNF-alpha therapy. Gut. 2005;54(4):590-591. 

9. Glück T, Linde HJ, Schölmerich J, et al. Anti-tumor necrosis factor therapy and Listeria monocytogenes infection: report of two cases. Arthritis Rheum. 2002;46(8):2255-2257.

10. Rijkeboer A, Voskuyl A, Van Agtmael M. Fatal Salmonella enteritidis septicaemia in a rheumatoid arthritis patient treated with a TNF-alpha antagonist. Scand J Infect Dis. 2007; 39(1):80-83.

11. Chan AT, Cleeve V, Daymond TJ. Necrotising fasciitis in a patient receiving infliximab for rheumatoid arthritis. Postgrad Med J. 2002; 78(915):47-48.

12. Favalli EG, Desiati F, Atzeni F, et al. Serious infections during anti-TNF-alpha treatment in rheumatoid arthritis patients. Autoimmun Rev. 2009;8(3):266-273.

13. Keane J, Gershon S, Wise RP, et al. Tuberculosis associated with infliximab, a tumor necrosis factor alpha-neutralizing agent. N Engl J Med. 2001;345(15):1098-1104.

References

1. Burns MJ. Chapter 181. Other immunosuppressive medications. In: Marx J, Hockberger R, Walls R, et al, eds. Rosen’s Emergency Medicine: Concepts and Clinical Practice. 7th ed. Philadelphia, PA: Mosby Elsevier; 2009.

2. Crum NF, Lederman ER, Wallace MR. Infections associated with tumor necrosis factor-alpha antagonists. Medicine (Baltimore). 2005; 84(5)291-302.

3. Geraghty EM, Ristow B, Gordon SM, Aronowitz P. Overwhelming parasitemia with Plasmodium falciparum infection in a patient receiving infliximab therapy for rheumatoid arthritis. Clin Infect Dis. 2007;44(10):82-84.

4. Listing J, Strangfeld A, Kary S, et al. Infections in patients with rheumatoid arthritis treated with biologic agents. Arthritis Rheum. 2005;52(11):3403-3412.

5. Louie SG, Park B, Yoon H. Biological response modifiers in the management of rheumatoid arthritis. Am J Health Syst Pharm. 2003;60(4): 346-355.

6. Kesteman T, Yombi JC, Gigi J, Durez P. Listeria infections associated with infliximab: case reports. Clin Rheumatol. 2007;26(12):2173-2175.

7. Kroesen S, Widmer AF, Tyndall A, Hasler P. Serious bacterial infections in patients with rheumatoid arthritis under anti-TNF-alpha therapy. Rheumatology. 2003;42(5):617-621.

8. Goode S, Tierney G, Deighton C. Life threatening intra-abdominal sepsis in patients on anti-TNF-alpha therapy. Gut. 2005;54(4):590-591. 

9. Glück T, Linde HJ, Schölmerich J, et al. Anti-tumor necrosis factor therapy and Listeria monocytogenes infection: report of two cases. Arthritis Rheum. 2002;46(8):2255-2257.

10. Rijkeboer A, Voskuyl A, Van Agtmael M. Fatal Salmonella enteritidis septicaemia in a rheumatoid arthritis patient treated with a TNF-alpha antagonist. Scand J Infect Dis. 2007; 39(1):80-83.

11. Chan AT, Cleeve V, Daymond TJ. Necrotising fasciitis in a patient receiving infliximab for rheumatoid arthritis. Postgrad Med J. 2002; 78(915):47-48.

12. Favalli EG, Desiati F, Atzeni F, et al. Serious infections during anti-TNF-alpha treatment in rheumatoid arthritis patients. Autoimmun Rev. 2009;8(3):266-273.

13. Keane J, Gershon S, Wise RP, et al. Tuberculosis associated with infliximab, a tumor necrosis factor alpha-neutralizing agent. N Engl J Med. 2001;345(15):1098-1104.

Issue
Clinician Reviews - 20(10)
Issue
Clinician Reviews - 20(10)
Page Number
38-43
Page Number
38-43
Publications
Publications
Topics
Article Type
Display Headline
Investigating Infection in the Patient With RA
Display Headline
Investigating Infection in the Patient With RA
Legacy Keywords
rheumatoid arthritis, infection, blood cultures, anti-tumor necrosis factor, infliximabrheumatoid arthritis, infection, blood cultures, anti-tumor necrosis factor, infliximab
Legacy Keywords
rheumatoid arthritis, infection, blood cultures, anti-tumor necrosis factor, infliximabrheumatoid arthritis, infection, blood cultures, anti-tumor necrosis factor, infliximab
Sections
Article Source

PURLs Copyright

Inside the Article

Grand Rounds: Man, 72, With Peeling Penile Skin

Article Type
Changed
Mon, 07/09/2018 - 10:47
Display Headline
Man, 72, With Peeling Penile Skin

A 72-year-old man presented to his primary care provider’s office with complaints of peeling skin on his penis and frequent, burning urination. He said he had first noticed redness on his penis about four days earlier, adding that it was growing worse. He was unsure whether he was truly experiencing frequent urination or just more aware of urinating because of the burning pain. He reported no attempts to treat himself, stating that he was “just keeping an eye on it and hoping it would go away.”

The patient’s medical history was limited to hypertension, for which he was taking valsartan, and allergies, for which he took fexofenadine. His surgical history included a tonsillectomy and appendectomy during his early teens. He had no known allergies to any medications.

The patient was married and retired after an executive career. He and his wife split their residence between New York and Florida during seasonal changes and were living in Florida at the time. He reported social drinking (“on rare occasions, these days”) and smoking an occasional cigar. He reported that he showers only once or twice weekly because of dry skin.

The following vital signs were recorded: blood pressure, 110/72 mm Hg; heart rate, 68 beats/min; respirations, 15/min; temperature, 97.8°F; and O2 saturation, 99% on room air. He was 73” tall and weighed 197 lb, with a BMI of 26.

The patient was alert and oriented. His physical exam was overall unremarkable, with the exception of an uncircumcised penis with redness and inflammation on the glans penis and no discharge noted. The reddened area was bright and shiny with a moist appearance and well-defined borders. The man denied any risk for sexually transmitted disease (STD) and denied any penile discharge. He also denied fever, chills, or arthritis.

Urinalysis performed in the office was negative for a urinary tract infection or for elevated glucose. A laboratory report from six months earlier was reviewed; all findings were within normal range, including the blood glucose level, with special attention paid for possible underlying cause; and the prostate-specific antigen (PSA) level, obtained for possible prostatitis or prostate cancer.

The differential diagnosis included eczema or psoriasis, Zoon’s balanitis, penile cancer, balanitis xerotica obliterans (lichen sclerosus), candidiasis balanitis, and circinate balanitis (as occurs in patients with Reiter’s disease; see table1-5). The absence of circumcision and the patient’s report of infrequent bathing raised concern for a hygiene-related etiology; the final diagnosis, made empirically, was candidiasis balanitis. Regarding an underlying cause, the laboratory order included a urine culture, fasting complete blood count, chemistry panel, and PSA level.

The patient was given instructions to wash the affected area twice daily for one week with a lukewarm weak saline solution (1 tablespoon salt/L water),5,6 gently retracting the foreskin; he was also given a topical antifungal cream7 (ketoconazole 2%, although other choices are discussed below), to be applied two to three times daily until his symptoms resolved.6 He was advised to return in one week if the condition did not improve or grew worse5; referral to dermatology would then be considered. The patient was also advised that in the case of a recurrent episode, dermatology would be consulted. The possibility of circumcision was discussed,8 and the patient was given information about the procedure, with referral to a urologist in the area.

Discussion

Balanitis is an inflammation of the glans penis; balanoposthitis involves the foreskin and prepuce.9-11 Balanitis can occur in men of any age, with etiologies varying with a patient’s age. Typical signs and symptoms include redness and swelling of the glans penis or foreskin, itching and/or pain, urethral discharge, phimosis, swollen lymph nodes, ulceration or plaque appearance, and pain on urination.12

In addition to the differential diagnoses mentioned, several additional conditions can be considered in a man with penile lesions. In older men, it is particularly important to investigate such lesions thoroughly, following the patient until the underlying cause is determined and the best treatment choice is selected. Specialists in dermatology and urology can best identify persistent or chronic lesions and make appropriate treatment recommendations, including possible circumcision.

The condition is commonly associated with absence of circumcision, poor hygiene, and phimosis (the inability to retract the foreskin from the glans penis). Accumulation of glandular secretions (smegma) and sloughed epithelial cells under the foreskin can lead to irritation and subsequent infection.

Uncontrolled or poorly controlled diabetes can be implicated in candidiasis infections.1 Other causes and contributing factors include chemical irritants (eg, soaps, lubricating jelly), edematous conditions (including congestive heart failure, cirrhosis, and nephrosis), drug allergies, morbid obesity, and a number of viruses and other pathogens, including those associated with STDs.12

 

 

A more detailed laboratory work-up might include the following:

• Serum glucose test (as part of a diabetes screening; in older men, this inflammatory condition can be a presenting sign of diabetes mellitus6)

• Culture of discharge, if any is present

• Serology test for STDs

• Wet mount with potassium hydroxide (for Candida albicans infection)

• Ultrasound, in severe cases or when urinary obstruction is suspected.

Additionally, in chronic cases, the patient should be referred to dermatology or urology for biopsy.5,9 Testing for anaerobes should also be considered for the patient and his sexual partner; if results are positive, treatment with oral metronidazole (400 mg tid for 10 days) is advised.6

In this patient’s case, the test that would best support an in-office diagnosis of candidiasis balanitis is a wet mount with potassium hydroxide. This was not performed at the time of the case patient’s visit, however; the diagnosis was empirically determined.

Management, Including Patient Education

Treatment of candidiasis balanitis involves routinely cleaning the penis and foreskin, as the case patient was instructed; use of soap, an irritant, should be avoided until the condition is resolved.7,10 Appropriate topical antifungal creams include nystatin, ketoconazole, miconazole, clotrimazole, econazole, and terbinafine, applied two to three times daily for at least 10 days; a cream combining an imidazole with 1% hydrocortisone may be effective for patients with significant inflammation.5,6,8,10,13

The patient should be instructed to:

• Keep the area clean and dry

• Wash twice daily with weak saline solution after removing residual medication and before applying fresh medication

• Wear loose cotton underwear

• Avoid sharing towels or cleaning cloths

• Wash personal items and surfaces, if possible, with disinfectant

• Notify sexual partner(s) that they may need treatment

• Discontinue sexual intercourse until infection is resolved

• Continue treatment for 10 to 14 days, even though relief may occur early

• Follow up with the clinician if no improvement is seen within one week

• Consider circumcision, in case of chronic infection.1,2,8,12

Conclusion

It is important to diagnose balanitis correctly, as this condition can affect sexual and urinary function, and its effects should not be underestimated in older men. Differentiating between infectious, noninfectious, premalignant, and malignant lesions will lead to appropriate care and allow early diagnosis or prevention of curable malignancies.

References

1. Singh S, Bunker C. Male genital dermatoses in old age. Age Ageing. 2008;37(5):500-504.

2. Thompson IM, Teichman JM, Elston DM, Sea J. Noninfectious penile lesions. Am Fam Physician. 2010;81(2):167-174.

3. Lane JE, Johnson J. Persistent penile patch. Am Fam Physician. 2008;78(9):1081-1082.

4. Gupta S, Malhotra AK, Ajith C. Lichen sclerosus: role of occlusion of the genital skin in the pathogenesis. Indian J Dermatol Venereol Leprol. 2010;76(1):56-58.

5. British Association for Sexual Health and HIV, Clinical Effectiveness Group. 2008 UK National Guideline on the Management of Balanoposthitis. www.bashh.org/documents/2062. Accessed September 22, 2010.

6. Ashton R, Leppard B. Differential Diagnosis in Dermatology. 3rd ed. London: Radcliffe Publishing Ltd; 2004:321.

7. NHS Institute for Innovation and Improvement. Clinical Knowledge Summaries: Balanitis (June 2009). www.cks.nhs.uk/balanitis/management/scenario_balanitis_adults#-378526. Accessed September 22, 2010.

8. Parker J. Management of common fungal infections in primary care. Nurs Stand. 2009;23(43):42-46.

9. Green MB, Bailey PP. Infectious processes: urinary tract infections and sexually transmitted diseases. In: Buttaro TM, Trybulski J, Bailey PP, Sandberg-Cook J, eds. Primary Care: A Collaborative Practice. 3rd ed. St. Louis, MO: Mosby Elsevier; 2008:576-590.

10. Singh-Behl D, Tomecki KJ. Common skins infections 2009. www.clevelandclinicmeded .com/medicalpubs/diseasemanagement/dermatol ogy/common-skin-infections. Accessed September 22, 2010.

11. Ko WT, Adal KA, Tomecki KJ. Infectious diseases. Med Clin North Am. 1998;82:(5):1001-1031.

12. Morgan K, McCance, KL. Alterations of the reproductive systems. In: McCance KL, Huether SE, eds. Pathophysiology: The Biologic Basis for Disease in Adults and Children. 5th ed. St. Louis, MO: Elsevier Mosby; 2006:805-807.

13. Waugh MA, Evans EG, Nayyar KC, Fong R. Clotrimazole (Canestan) in the treatment of candidal balanitis in men: with incidental observations on diabetic candidal balanoposthitis. Br J Vener Dis. 1978;54(3):184-186.

Author and Disclosure Information

 

Jan Meires, EdD, FNP, BC, Bridgette Futch, RN, BSN, CEN, FNP-S

Issue
Clinician Reviews - 20(10)
Publications
Topics
Page Number
13-15
Legacy Keywords
balanitis, inflammation, glans penisbalanitis, inflammation, glans penis
Sections
Author and Disclosure Information

 

Jan Meires, EdD, FNP, BC, Bridgette Futch, RN, BSN, CEN, FNP-S

Author and Disclosure Information

 

Jan Meires, EdD, FNP, BC, Bridgette Futch, RN, BSN, CEN, FNP-S

A 72-year-old man presented to his primary care provider’s office with complaints of peeling skin on his penis and frequent, burning urination. He said he had first noticed redness on his penis about four days earlier, adding that it was growing worse. He was unsure whether he was truly experiencing frequent urination or just more aware of urinating because of the burning pain. He reported no attempts to treat himself, stating that he was “just keeping an eye on it and hoping it would go away.”

The patient’s medical history was limited to hypertension, for which he was taking valsartan, and allergies, for which he took fexofenadine. His surgical history included a tonsillectomy and appendectomy during his early teens. He had no known allergies to any medications.

The patient was married and retired after an executive career. He and his wife split their residence between New York and Florida during seasonal changes and were living in Florida at the time. He reported social drinking (“on rare occasions, these days”) and smoking an occasional cigar. He reported that he showers only once or twice weekly because of dry skin.

The following vital signs were recorded: blood pressure, 110/72 mm Hg; heart rate, 68 beats/min; respirations, 15/min; temperature, 97.8°F; and O2 saturation, 99% on room air. He was 73” tall and weighed 197 lb, with a BMI of 26.

The patient was alert and oriented. His physical exam was overall unremarkable, with the exception of an uncircumcised penis with redness and inflammation on the glans penis and no discharge noted. The reddened area was bright and shiny with a moist appearance and well-defined borders. The man denied any risk for sexually transmitted disease (STD) and denied any penile discharge. He also denied fever, chills, or arthritis.

Urinalysis performed in the office was negative for a urinary tract infection or for elevated glucose. A laboratory report from six months earlier was reviewed; all findings were within normal range, including the blood glucose level, with special attention paid for possible underlying cause; and the prostate-specific antigen (PSA) level, obtained for possible prostatitis or prostate cancer.

The differential diagnosis included eczema or psoriasis, Zoon’s balanitis, penile cancer, balanitis xerotica obliterans (lichen sclerosus), candidiasis balanitis, and circinate balanitis (as occurs in patients with Reiter’s disease; see table1-5). The absence of circumcision and the patient’s report of infrequent bathing raised concern for a hygiene-related etiology; the final diagnosis, made empirically, was candidiasis balanitis. Regarding an underlying cause, the laboratory order included a urine culture, fasting complete blood count, chemistry panel, and PSA level.

The patient was given instructions to wash the affected area twice daily for one week with a lukewarm weak saline solution (1 tablespoon salt/L water),5,6 gently retracting the foreskin; he was also given a topical antifungal cream7 (ketoconazole 2%, although other choices are discussed below), to be applied two to three times daily until his symptoms resolved.6 He was advised to return in one week if the condition did not improve or grew worse5; referral to dermatology would then be considered. The patient was also advised that in the case of a recurrent episode, dermatology would be consulted. The possibility of circumcision was discussed,8 and the patient was given information about the procedure, with referral to a urologist in the area.

Discussion

Balanitis is an inflammation of the glans penis; balanoposthitis involves the foreskin and prepuce.9-11 Balanitis can occur in men of any age, with etiologies varying with a patient’s age. Typical signs and symptoms include redness and swelling of the glans penis or foreskin, itching and/or pain, urethral discharge, phimosis, swollen lymph nodes, ulceration or plaque appearance, and pain on urination.12

In addition to the differential diagnoses mentioned, several additional conditions can be considered in a man with penile lesions. In older men, it is particularly important to investigate such lesions thoroughly, following the patient until the underlying cause is determined and the best treatment choice is selected. Specialists in dermatology and urology can best identify persistent or chronic lesions and make appropriate treatment recommendations, including possible circumcision.

The condition is commonly associated with absence of circumcision, poor hygiene, and phimosis (the inability to retract the foreskin from the glans penis). Accumulation of glandular secretions (smegma) and sloughed epithelial cells under the foreskin can lead to irritation and subsequent infection.

Uncontrolled or poorly controlled diabetes can be implicated in candidiasis infections.1 Other causes and contributing factors include chemical irritants (eg, soaps, lubricating jelly), edematous conditions (including congestive heart failure, cirrhosis, and nephrosis), drug allergies, morbid obesity, and a number of viruses and other pathogens, including those associated with STDs.12

 

 

A more detailed laboratory work-up might include the following:

• Serum glucose test (as part of a diabetes screening; in older men, this inflammatory condition can be a presenting sign of diabetes mellitus6)

• Culture of discharge, if any is present

• Serology test for STDs

• Wet mount with potassium hydroxide (for Candida albicans infection)

• Ultrasound, in severe cases or when urinary obstruction is suspected.

Additionally, in chronic cases, the patient should be referred to dermatology or urology for biopsy.5,9 Testing for anaerobes should also be considered for the patient and his sexual partner; if results are positive, treatment with oral metronidazole (400 mg tid for 10 days) is advised.6

In this patient’s case, the test that would best support an in-office diagnosis of candidiasis balanitis is a wet mount with potassium hydroxide. This was not performed at the time of the case patient’s visit, however; the diagnosis was empirically determined.

Management, Including Patient Education

Treatment of candidiasis balanitis involves routinely cleaning the penis and foreskin, as the case patient was instructed; use of soap, an irritant, should be avoided until the condition is resolved.7,10 Appropriate topical antifungal creams include nystatin, ketoconazole, miconazole, clotrimazole, econazole, and terbinafine, applied two to three times daily for at least 10 days; a cream combining an imidazole with 1% hydrocortisone may be effective for patients with significant inflammation.5,6,8,10,13

The patient should be instructed to:

• Keep the area clean and dry

• Wash twice daily with weak saline solution after removing residual medication and before applying fresh medication

• Wear loose cotton underwear

• Avoid sharing towels or cleaning cloths

• Wash personal items and surfaces, if possible, with disinfectant

• Notify sexual partner(s) that they may need treatment

• Discontinue sexual intercourse until infection is resolved

• Continue treatment for 10 to 14 days, even though relief may occur early

• Follow up with the clinician if no improvement is seen within one week

• Consider circumcision, in case of chronic infection.1,2,8,12

Conclusion

It is important to diagnose balanitis correctly, as this condition can affect sexual and urinary function, and its effects should not be underestimated in older men. Differentiating between infectious, noninfectious, premalignant, and malignant lesions will lead to appropriate care and allow early diagnosis or prevention of curable malignancies.

A 72-year-old man presented to his primary care provider’s office with complaints of peeling skin on his penis and frequent, burning urination. He said he had first noticed redness on his penis about four days earlier, adding that it was growing worse. He was unsure whether he was truly experiencing frequent urination or just more aware of urinating because of the burning pain. He reported no attempts to treat himself, stating that he was “just keeping an eye on it and hoping it would go away.”

The patient’s medical history was limited to hypertension, for which he was taking valsartan, and allergies, for which he took fexofenadine. His surgical history included a tonsillectomy and appendectomy during his early teens. He had no known allergies to any medications.

The patient was married and retired after an executive career. He and his wife split their residence between New York and Florida during seasonal changes and were living in Florida at the time. He reported social drinking (“on rare occasions, these days”) and smoking an occasional cigar. He reported that he showers only once or twice weekly because of dry skin.

The following vital signs were recorded: blood pressure, 110/72 mm Hg; heart rate, 68 beats/min; respirations, 15/min; temperature, 97.8°F; and O2 saturation, 99% on room air. He was 73” tall and weighed 197 lb, with a BMI of 26.

The patient was alert and oriented. His physical exam was overall unremarkable, with the exception of an uncircumcised penis with redness and inflammation on the glans penis and no discharge noted. The reddened area was bright and shiny with a moist appearance and well-defined borders. The man denied any risk for sexually transmitted disease (STD) and denied any penile discharge. He also denied fever, chills, or arthritis.

Urinalysis performed in the office was negative for a urinary tract infection or for elevated glucose. A laboratory report from six months earlier was reviewed; all findings were within normal range, including the blood glucose level, with special attention paid for possible underlying cause; and the prostate-specific antigen (PSA) level, obtained for possible prostatitis or prostate cancer.

The differential diagnosis included eczema or psoriasis, Zoon’s balanitis, penile cancer, balanitis xerotica obliterans (lichen sclerosus), candidiasis balanitis, and circinate balanitis (as occurs in patients with Reiter’s disease; see table1-5). The absence of circumcision and the patient’s report of infrequent bathing raised concern for a hygiene-related etiology; the final diagnosis, made empirically, was candidiasis balanitis. Regarding an underlying cause, the laboratory order included a urine culture, fasting complete blood count, chemistry panel, and PSA level.

The patient was given instructions to wash the affected area twice daily for one week with a lukewarm weak saline solution (1 tablespoon salt/L water),5,6 gently retracting the foreskin; he was also given a topical antifungal cream7 (ketoconazole 2%, although other choices are discussed below), to be applied two to three times daily until his symptoms resolved.6 He was advised to return in one week if the condition did not improve or grew worse5; referral to dermatology would then be considered. The patient was also advised that in the case of a recurrent episode, dermatology would be consulted. The possibility of circumcision was discussed,8 and the patient was given information about the procedure, with referral to a urologist in the area.

Discussion

Balanitis is an inflammation of the glans penis; balanoposthitis involves the foreskin and prepuce.9-11 Balanitis can occur in men of any age, with etiologies varying with a patient’s age. Typical signs and symptoms include redness and swelling of the glans penis or foreskin, itching and/or pain, urethral discharge, phimosis, swollen lymph nodes, ulceration or plaque appearance, and pain on urination.12

In addition to the differential diagnoses mentioned, several additional conditions can be considered in a man with penile lesions. In older men, it is particularly important to investigate such lesions thoroughly, following the patient until the underlying cause is determined and the best treatment choice is selected. Specialists in dermatology and urology can best identify persistent or chronic lesions and make appropriate treatment recommendations, including possible circumcision.

The condition is commonly associated with absence of circumcision, poor hygiene, and phimosis (the inability to retract the foreskin from the glans penis). Accumulation of glandular secretions (smegma) and sloughed epithelial cells under the foreskin can lead to irritation and subsequent infection.

Uncontrolled or poorly controlled diabetes can be implicated in candidiasis infections.1 Other causes and contributing factors include chemical irritants (eg, soaps, lubricating jelly), edematous conditions (including congestive heart failure, cirrhosis, and nephrosis), drug allergies, morbid obesity, and a number of viruses and other pathogens, including those associated with STDs.12

 

 

A more detailed laboratory work-up might include the following:

• Serum glucose test (as part of a diabetes screening; in older men, this inflammatory condition can be a presenting sign of diabetes mellitus6)

• Culture of discharge, if any is present

• Serology test for STDs

• Wet mount with potassium hydroxide (for Candida albicans infection)

• Ultrasound, in severe cases or when urinary obstruction is suspected.

Additionally, in chronic cases, the patient should be referred to dermatology or urology for biopsy.5,9 Testing for anaerobes should also be considered for the patient and his sexual partner; if results are positive, treatment with oral metronidazole (400 mg tid for 10 days) is advised.6

In this patient’s case, the test that would best support an in-office diagnosis of candidiasis balanitis is a wet mount with potassium hydroxide. This was not performed at the time of the case patient’s visit, however; the diagnosis was empirically determined.

Management, Including Patient Education

Treatment of candidiasis balanitis involves routinely cleaning the penis and foreskin, as the case patient was instructed; use of soap, an irritant, should be avoided until the condition is resolved.7,10 Appropriate topical antifungal creams include nystatin, ketoconazole, miconazole, clotrimazole, econazole, and terbinafine, applied two to three times daily for at least 10 days; a cream combining an imidazole with 1% hydrocortisone may be effective for patients with significant inflammation.5,6,8,10,13

The patient should be instructed to:

• Keep the area clean and dry

• Wash twice daily with weak saline solution after removing residual medication and before applying fresh medication

• Wear loose cotton underwear

• Avoid sharing towels or cleaning cloths

• Wash personal items and surfaces, if possible, with disinfectant

• Notify sexual partner(s) that they may need treatment

• Discontinue sexual intercourse until infection is resolved

• Continue treatment for 10 to 14 days, even though relief may occur early

• Follow up with the clinician if no improvement is seen within one week

• Consider circumcision, in case of chronic infection.1,2,8,12

Conclusion

It is important to diagnose balanitis correctly, as this condition can affect sexual and urinary function, and its effects should not be underestimated in older men. Differentiating between infectious, noninfectious, premalignant, and malignant lesions will lead to appropriate care and allow early diagnosis or prevention of curable malignancies.

References

1. Singh S, Bunker C. Male genital dermatoses in old age. Age Ageing. 2008;37(5):500-504.

2. Thompson IM, Teichman JM, Elston DM, Sea J. Noninfectious penile lesions. Am Fam Physician. 2010;81(2):167-174.

3. Lane JE, Johnson J. Persistent penile patch. Am Fam Physician. 2008;78(9):1081-1082.

4. Gupta S, Malhotra AK, Ajith C. Lichen sclerosus: role of occlusion of the genital skin in the pathogenesis. Indian J Dermatol Venereol Leprol. 2010;76(1):56-58.

5. British Association for Sexual Health and HIV, Clinical Effectiveness Group. 2008 UK National Guideline on the Management of Balanoposthitis. www.bashh.org/documents/2062. Accessed September 22, 2010.

6. Ashton R, Leppard B. Differential Diagnosis in Dermatology. 3rd ed. London: Radcliffe Publishing Ltd; 2004:321.

7. NHS Institute for Innovation and Improvement. Clinical Knowledge Summaries: Balanitis (June 2009). www.cks.nhs.uk/balanitis/management/scenario_balanitis_adults#-378526. Accessed September 22, 2010.

8. Parker J. Management of common fungal infections in primary care. Nurs Stand. 2009;23(43):42-46.

9. Green MB, Bailey PP. Infectious processes: urinary tract infections and sexually transmitted diseases. In: Buttaro TM, Trybulski J, Bailey PP, Sandberg-Cook J, eds. Primary Care: A Collaborative Practice. 3rd ed. St. Louis, MO: Mosby Elsevier; 2008:576-590.

10. Singh-Behl D, Tomecki KJ. Common skins infections 2009. www.clevelandclinicmeded .com/medicalpubs/diseasemanagement/dermatol ogy/common-skin-infections. Accessed September 22, 2010.

11. Ko WT, Adal KA, Tomecki KJ. Infectious diseases. Med Clin North Am. 1998;82:(5):1001-1031.

12. Morgan K, McCance, KL. Alterations of the reproductive systems. In: McCance KL, Huether SE, eds. Pathophysiology: The Biologic Basis for Disease in Adults and Children. 5th ed. St. Louis, MO: Elsevier Mosby; 2006:805-807.

13. Waugh MA, Evans EG, Nayyar KC, Fong R. Clotrimazole (Canestan) in the treatment of candidal balanitis in men: with incidental observations on diabetic candidal balanoposthitis. Br J Vener Dis. 1978;54(3):184-186.

References

1. Singh S, Bunker C. Male genital dermatoses in old age. Age Ageing. 2008;37(5):500-504.

2. Thompson IM, Teichman JM, Elston DM, Sea J. Noninfectious penile lesions. Am Fam Physician. 2010;81(2):167-174.

3. Lane JE, Johnson J. Persistent penile patch. Am Fam Physician. 2008;78(9):1081-1082.

4. Gupta S, Malhotra AK, Ajith C. Lichen sclerosus: role of occlusion of the genital skin in the pathogenesis. Indian J Dermatol Venereol Leprol. 2010;76(1):56-58.

5. British Association for Sexual Health and HIV, Clinical Effectiveness Group. 2008 UK National Guideline on the Management of Balanoposthitis. www.bashh.org/documents/2062. Accessed September 22, 2010.

6. Ashton R, Leppard B. Differential Diagnosis in Dermatology. 3rd ed. London: Radcliffe Publishing Ltd; 2004:321.

7. NHS Institute for Innovation and Improvement. Clinical Knowledge Summaries: Balanitis (June 2009). www.cks.nhs.uk/balanitis/management/scenario_balanitis_adults#-378526. Accessed September 22, 2010.

8. Parker J. Management of common fungal infections in primary care. Nurs Stand. 2009;23(43):42-46.

9. Green MB, Bailey PP. Infectious processes: urinary tract infections and sexually transmitted diseases. In: Buttaro TM, Trybulski J, Bailey PP, Sandberg-Cook J, eds. Primary Care: A Collaborative Practice. 3rd ed. St. Louis, MO: Mosby Elsevier; 2008:576-590.

10. Singh-Behl D, Tomecki KJ. Common skins infections 2009. www.clevelandclinicmeded .com/medicalpubs/diseasemanagement/dermatol ogy/common-skin-infections. Accessed September 22, 2010.

11. Ko WT, Adal KA, Tomecki KJ. Infectious diseases. Med Clin North Am. 1998;82:(5):1001-1031.

12. Morgan K, McCance, KL. Alterations of the reproductive systems. In: McCance KL, Huether SE, eds. Pathophysiology: The Biologic Basis for Disease in Adults and Children. 5th ed. St. Louis, MO: Elsevier Mosby; 2006:805-807.

13. Waugh MA, Evans EG, Nayyar KC, Fong R. Clotrimazole (Canestan) in the treatment of candidal balanitis in men: with incidental observations on diabetic candidal balanoposthitis. Br J Vener Dis. 1978;54(3):184-186.

Issue
Clinician Reviews - 20(10)
Issue
Clinician Reviews - 20(10)
Page Number
13-15
Page Number
13-15
Publications
Publications
Topics
Article Type
Display Headline
Man, 72, With Peeling Penile Skin
Display Headline
Man, 72, With Peeling Penile Skin
Legacy Keywords
balanitis, inflammation, glans penisbalanitis, inflammation, glans penis
Legacy Keywords
balanitis, inflammation, glans penisbalanitis, inflammation, glans penis
Sections
Disallow All Ads
Alternative CME

Hydroxocobalamin for Suspected Cyanide Poisoning

Article Type
Changed
Wed, 12/12/2018 - 19:30
Display Headline
Hydroxocobalamin for Suspected Cyanide Poisoning

Article PDF
Author and Disclosure Information

Lewis S. Nelson, MD (Series Editor), and Daniel M. Lugassy, MD

Issue
Emergency Medicine - 42(10)
Publications
Page Number
15-18
Legacy Keywords
cyanide poisoning, hydroxocobalamincyanide poisoning, hydroxocobalamin
Sections
Author and Disclosure Information

Lewis S. Nelson, MD (Series Editor), and Daniel M. Lugassy, MD

Author and Disclosure Information

Lewis S. Nelson, MD (Series Editor), and Daniel M. Lugassy, MD

Article PDF
Article PDF

Issue
Emergency Medicine - 42(10)
Issue
Emergency Medicine - 42(10)
Page Number
15-18
Page Number
15-18
Publications
Publications
Article Type
Display Headline
Hydroxocobalamin for Suspected Cyanide Poisoning
Display Headline
Hydroxocobalamin for Suspected Cyanide Poisoning
Legacy Keywords
cyanide poisoning, hydroxocobalamincyanide poisoning, hydroxocobalamin
Legacy Keywords
cyanide poisoning, hydroxocobalamincyanide poisoning, hydroxocobalamin
Sections
Article Source

PURLs Copyright

Inside the Article

Article PDF Media

Emergent Management of Pediatric Seizures

Article Type
Changed
Wed, 12/12/2018 - 19:30
Display Headline
Emergent Management of Pediatric Seizures

Article PDF
Author and Disclosure Information

Joesph King, MD, and Bruce Lo, MD, RDMS

Issue
Emergency Medicine - 42(10)
Publications
Topics
Page Number
6-13
Legacy Keywords
partial seizures, generalized seizures, febrile seizures, epilepsypartial seizures, generalized seizures, febrile seizures, epilepsy
Sections
Author and Disclosure Information

Joesph King, MD, and Bruce Lo, MD, RDMS

Author and Disclosure Information

Joesph King, MD, and Bruce Lo, MD, RDMS

Article PDF
Article PDF

Issue
Emergency Medicine - 42(10)
Issue
Emergency Medicine - 42(10)
Page Number
6-13
Page Number
6-13
Publications
Publications
Topics
Article Type
Display Headline
Emergent Management of Pediatric Seizures
Display Headline
Emergent Management of Pediatric Seizures
Legacy Keywords
partial seizures, generalized seizures, febrile seizures, epilepsypartial seizures, generalized seizures, febrile seizures, epilepsy
Legacy Keywords
partial seizures, generalized seizures, febrile seizures, epilepsypartial seizures, generalized seizures, febrile seizures, epilepsy
Sections
Article Source

PURLs Copyright

Inside the Article

Article PDF Media

Familial Tumoral Calcinosis

Article Type
Changed
Thu, 09/19/2019 - 13:59
Display Headline
Familial Tumoral Calcinosis

Article PDF
Author and Disclosure Information

Jean Jose, DO, Braden Fitcher, and Paul D. Clifford, MD

Dr. Jose is Assistant Professor of Clinical Radiology, Musculoskeletal Imaging Section, Department of Radiology, Mr. Fitcher is a Medical Student, and Dr. Clifford is Associate Professor of Clinical Radiology and Chief, Musculoskeletal Imaging Section, Department of Radiology, University of Miami Miller School of Medicine, Miami, Florida.

Issue
The American Journal of Orthopedics - 39(10)
Publications
Topics
Page Number
E111-E113
Legacy Keywords
Tumoral calcinosis; familial tumoral calcinosis; hyperphosphatemia; calcium hydroxyapatite; MRI; hyperostosis-hyperphosphataemia syndrome; CT; Familial Tumoral Calcinosis; Jose; Fitcher; Clifford; The American Journal of Orthopedics, ajo
Sections
Author and Disclosure Information

Jean Jose, DO, Braden Fitcher, and Paul D. Clifford, MD

Dr. Jose is Assistant Professor of Clinical Radiology, Musculoskeletal Imaging Section, Department of Radiology, Mr. Fitcher is a Medical Student, and Dr. Clifford is Associate Professor of Clinical Radiology and Chief, Musculoskeletal Imaging Section, Department of Radiology, University of Miami Miller School of Medicine, Miami, Florida.

Author and Disclosure Information

Jean Jose, DO, Braden Fitcher, and Paul D. Clifford, MD

Dr. Jose is Assistant Professor of Clinical Radiology, Musculoskeletal Imaging Section, Department of Radiology, Mr. Fitcher is a Medical Student, and Dr. Clifford is Associate Professor of Clinical Radiology and Chief, Musculoskeletal Imaging Section, Department of Radiology, University of Miami Miller School of Medicine, Miami, Florida.

Article PDF
Article PDF

Issue
The American Journal of Orthopedics - 39(10)
Issue
The American Journal of Orthopedics - 39(10)
Page Number
E111-E113
Page Number
E111-E113
Publications
Publications
Topics
Article Type
Display Headline
Familial Tumoral Calcinosis
Display Headline
Familial Tumoral Calcinosis
Legacy Keywords
Tumoral calcinosis; familial tumoral calcinosis; hyperphosphatemia; calcium hydroxyapatite; MRI; hyperostosis-hyperphosphataemia syndrome; CT; Familial Tumoral Calcinosis; Jose; Fitcher; Clifford; The American Journal of Orthopedics, ajo
Legacy Keywords
Tumoral calcinosis; familial tumoral calcinosis; hyperphosphatemia; calcium hydroxyapatite; MRI; hyperostosis-hyperphosphataemia syndrome; CT; Familial Tumoral Calcinosis; Jose; Fitcher; Clifford; The American Journal of Orthopedics, ajo
Sections
Article Source

PURLs Copyright

Inside the Article

Article PDF Media

UPDATE: PELVIC FLOOR DYSFUNCTION

Article Type
Changed
Tue, 08/28/2018 - 10:58
Display Headline
UPDATE: PELVIC FLOOR DYSFUNCTION

When a woman has advanced prolapse of the anterior vaginal wall, it is highly likely that she has apical prolapse as well. Consider a study by Rooney and associates that determined that clinically significant vault prolapse is present in most women who have anterior vaginal prolapse of stage II or higher.1 For that reason, suspension of the vaginal apex should be considered whenever surgical treatment of anterior wall defects is planned.

Sacrocolpopexy involves suspension of the vaginal vault from the anterior longitudinal ligament of the sacrum, using Y-shaped mesh to augment native tissue (FIGURE).2 It is an effective, durable treatment for vaginal apical prolapse. With a success rate approaching 93%, this procedure has become the gold standard for repair of vault prolapse. Among its advantages are maximization of vaginal depth and preservation of a normal vaginal axis.

Sacrocolpopexy preserves the vaginal axis



With the vaginal vault suspended from the anterior longitudinal
ligament of the sacrum, the normal vaginal axis is preserved
and vaginal depth is maximized.

Sacrocolpopexy can be performed via the abdominal, laparoscopic, or robotic-assisted approach (TABLE 1). Minimally invasive techniques are attractive because they involve faster recovery than abdominal sacrocolpopexy does. Minimally invasive techniques have also advanced to the point that they are both effective and durable. However, these advantages must be weighed against the effort required to learn the techniques, as well as their higher cost.

TABLE 1

How the 3 approaches to sacrocolpopexy compare

ApproachAdvantages and disadvantages
AbdominalShortest operative time
No significant Trendelenburg position required
Highest estimated blood loss
Longest length of stay
Low rate of complications
Longest postoperative recovery
Well-established long-term durability
LaparoscopicLonger operative time
Moderate Trendelenburg position required
Lower estimated blood loss
Shorter length of stay
Surgical technique least similar to abdominal procedure
Low rate of complications
Shorter postoperative recovery
Long-term durability less firmly established
Robotic-assistedLongest operative time
Steep Trendelenburg position required
Lower estimated blood loss
Shorter length of stay
Surgical technique resembles that of abdominal approach
Low rate of complications
Shorter postoperative recovery
Long-term durability appears to be good

In this article, we highlight:

  • a comparison of the laparoscopic and abdominal approaches to sacrocolpopexy
  • an investigation of the learning curve associated with robotic-assisted sacrocolpopexy
  • a study exploring the durability of robotic-assisted repair
  • an estimate of the costs associated with each route of operation.

Laparoscopic vs abdominal sacrocolpopexy—how do they compare?

Paraiso MF, Walters MD, Rackley RR, Melek S, Hugney C. Laparoscopic and abdominal sacral colpopexies: a comparative cohort study. Am J Obstet Gynecol. 2005;192(5):1752–1758.

When surgeons at the Cleveland Clinic performed a retrospective cohort study to compare laparoscopic and abdominal sacrocolpopexy, they found significantly longer operative time with the laparoscopic route, with an average difference of 51 minutes (P < .0001). However, the laparoscopic approach was associated with lower blood loss (although there was no difference between groups in hematocrit on postoperative day 1); shorter hospital stay (average of 1.8 days versus 4 days [P < .001]); and comparable rates of intraoperative and postoperative complications.

Details of the trial

Paraiso and colleagues reviewed the medical charts of 56 consecutive patients who had undergone laparoscopic sacrocolpopexy, comparing them with the charts of 61 consecutive patients who had undergone the procedure using the abdominal approach. The operations had been performed between 1998 and 2003 for treatment of posthysterectomy vaginal prolapse.

The groups underwent similar rates of concurrent procedures. The laparotomy group had a significantly higher number of Burch procedures (P = .007), and the laparoscopic group had a significantly higher rate of adhesiolysis (P = .002).

Among the complications noted— which occurred at comparable rates between groups—were cystotomy, enterotomy, need for transfusion, deep-vein thrombosis, ileus, small bowel obstruction, wound infection, ventral hernia, mesh erosion, and recurrent prolapse. One laparoscopic case was converted to laparotomy because of excessive bleeding during the rectopexy portion of the operation.

Laparoscopy may have taken longer than this trial suggests

This study is one of very few well-designed trials comparing laparoscopic sacrocolpopexy to the historical gold standard of abdominal sacrocolpopexy for vault prolapse.

Twenty-eight percent of laparoscopic procedures in this study used tacking devices in lieu of suturing. Had suturing been performed universally, an even greater difference in surgical time may have been observed.

 

 

There may also be differences between groups in the durability of the two types of repair, an outcome not included in this particular study.

WHAT THIS EVIDENCE MEANS FOR PRACTICE

The laparoscopic approach offers a shorter hospital stay with no increase in intraoperative or postoperative complications, compared with abdominal sacrocolpopexy. However, it entails a significantly longer operative time than the abdominal approach does.

How steep is the learning curve for robotic-assisted sacrocolpopexy?

Akl MN, Long JB, Giles DL, et al. Robotic-assisted sacrocolpopexy: technique and learning curve. Surg Endosc. 2009;23(10):2390–2394.

Akl and coworkers reviewed the medical records of all patients who had undergone robotic-assisted sacrocolpopexy at the Mayo Clinics in Arizona and Florida between 2004 and 2007. All operations were performed by the same four urogynecologists, with an average operative time of 197.9 minutes (standard deviation, ± 66.8 minutes). However, after the first 10 cases, the operative time decreased by 64.3 minutes—a decline of 25.4% (P < .01; 95% confidence interval [CI], 16.1–112.4 minutes).

Details of the trial

Researchers collected baseline information on participants’ age, stage of prolapse, and concomitant procedures. They also gathered data on average operative time, estimated blood loss, intraoperative and postoperative complications, conversion to laparotomy, and length of hospitalization.

Of 80 women who had advanced pelvic organ prolapse (stage III/IV) who underwent robotic-assisted sacrocolpopexy, 88% underwent concomitant robotic and vaginal procedures, including robotic supracervical hysterectomy, Burch procedure, paravaginal repair, lysis of adhesions, bilateral salpingooophorectomy, vaginal cystocele or rectocele repair, and placement of a midurethral sling.

Estimated blood loss for the robotic-assisted approach ranged from 25 mL to 300 mL, with a mean loss of 96.8 mL. Average length of hospitalization was 2.6 days. Four cases (5%) were converted to laparotomy because of limited exposure and one intraoperative bladder injury. Other intraoperative complications included small-bowel injury during trocar placement and one ureteral injury. Postoperative complications included one case of ileus and five (6%) vaginal mesh erosions. Three patients developed recurrent prolapse and underwent subsequent correction.

Learning curve could have been measured more precisely

The authors did not specifically measure the learning curve for robotic-assisted sacrocolpopexy, as they took into account the concomitant procedures. For this reason, the decrease in operative time observed after 10 cases may not accurately reflect an improvement in the performance of sacrocolpopexy.

Akl and colleagues consider this detail to be a strength of the study because most women who undergo prolapse surgery have concomitant procedures. However, recording the length of time it took to perform the sacrocolpopexy portion of the procedure would have been more accurate.

The average length of stay approached that of the abdominal route. Length of stay may decline as a surgeon gains experience with the robotic-assisted approach.

WHAT THIS EVIDENCE MEANS FOR PRACTICE

Robotic-assisted sacrocolpopexy has a steep learning curve with respect to technique and surgical time.

Does robotic-assisted sacrocolpopexy provide durable support?

Elliott DS, Krambeck AE, Chow GK. Long-term results of robotic assisted laparoscopic sacrocolpopexy for the treatment of high grade vaginal vault prolapse. J Urol. 2006;176(2):655–659.

Among the few recent series reporting long-term outcomes after robotic-assisted sacrocolpopexy is this observational study from the Mayo Clinic. It involved 30 women who underwent the operation for the treatment of Baden Walker grade 4/4 posthysterectomy vaginal vault prolapse. The authors concluded that advanced prolapse can be treated with robotic-assisted sacrocolpopexy with long-term success and minimal complications.

Details of the trial

Of 30 women in this trial, 52% underwent an anti-incontinence procedure at the time of sacrocolpopexy. Women who had multiple vaginal defects or a history of abdominal surgery were excluded from the study.

Average operative time was 3.1 hours (range, 2.15–4.75 hours) in the early phase of development of operative technique (described in the manuscript) but diminished over time to an average of 2.5 hours.

Twenty-nine patients were discharged from the hospital after an overnight stay. Very few immediate postoperative complications were observed. Two patients experienced mild port-site infections that required outpatient treatment, and one patient had persistent vaginal bleeding from the incision made during the anti-incontinence procedure.

Most patients were followed for at least 1 year

The mean follow-up in this study was 24 months (range, 16–39 months). During this period, 21 women were followed for a full year. Long-term observation revealed that the repair of vault prolapse remained successful in 19 of these women.

One patient experienced recurrent prolapse 7 months after surgery. Another developed a rectocele 9 months after sacrocolpopexy. Vaginal mesh erosions occurred in two patients within 6 months after the procedure; both patients were treated with outpatient resection of the exposed mesh, with no recurrence of the prolapse.

 

 

Although a larger sample size and longer follow-up would be ideal, this study demonstrates a low rate of recurrent prolapse 1 year after the procedure.

WHAT THIS EVIDENCE MEANS FOR PRACTICE

Robotic sacrocolpopexy appears to provide long-term durability for the treatment of advanced vaginal vault prolapse.

How to choose an approach when planning sacrocolpopexy

Depending on where you practice, you may have as many as three options: abdominal, laparoscopic, or robotic-assisted. Here are basic questions you should address when choosing one:

  • How familiar are you with the technique? if the answer is “not much,” you can anticipate that the cost and time required to perform it will be significantly higher.
  • Are the appropriate instruments and surgical team available?
  • Does the patient have comorbidities? Consider, for example, the fact that she may not be able to tolerate a steep Trendelenberg position—required for the robotic-assisted approach—if she has severe cardiac or pulmonary disease. However, if she has a risk of poor wound healing, a large abdominal incision may not be advisable and postoperative immobility can be risky. if she is obese, laparoscopic or robotic port placement is challenging, but visualization and retraction will be easier. The need for anticoagulation is another consideration, as it will affect estimated blood loss and the choice of an incision, among other things.
  • Let’s not forget the patient. Given the pros and cons, what approach does she prefer?

How much do laparoscopic, abdominal, and robotic-assisted sacrocolpopexy cost?

Judd JP, Siddiqui NY, Barnett JC, et al. Cost-minimization analysis of robotic-assisted, laparoscopic, and abdominal sacrocolpopexy. J Minim Invasive Gynecol. 2010;17: 493–499.

This cost-minimization analysis concluded that robotic-assisted sacrocolpopexy incurs the highest hospital charges but is reimbursed by Medicare at a rate similar to reimbursement for the abdominal and laparoscopic routes (TABLE 2).

TABLE 2

Cost of sacrocolpopexy is significant—especially using the robotic approach

ApproachCost of a procedureOperative time, min (range)
Robotic-assisted$8,508328 (130–383)
Laparoscopic$7,353269 (97–334)
Abdominal$5,792170 (110–286)
Source: Judd JP, Siddiqui NY, Barnett JC, Visco AG, Havrilesky LJ, Wu JM. Cost-minimization analysis of robotic-assisted, laparoscopic, and abdominal sacrocolpopexy. J Minim invasive Gynecol. 2010;17(4):493–499.

The analysis accounted for realistic practices, such as the inclusion of concurrent hysterectomy and other procedures.

Details of the trial

Surgeons from Duke University developed a decision-analysis model in which a hypothetical group of women with advanced vaginal prolapse could choose between one of the three routes of sacrocolpopexy: abdominal, laparoscopic, or robotic-assisted. Researchers postulated two different scenarios:

  • the hospital had ownership of a robotic system
  • the hospital invested in the initial purchase and maintenance of such a system.

Researchers reviewed the literature to formulate their estimates of operative time, rate of conversion to laparotomy, rate of transfusion, and length of hospital stay. In addition, the costs of initial anesthesia setup, professional fees, per-minute intraoperative fees, and postanesthesia care were applied to each approach. Operating room costs per minute and the cost of disposable items such as drapes, gowns, gloves, and single-use instruments were added. For the robotic approach, the costs of reusable instruments were distributed across 10 operations. Reusable instruments for laparoscopic and abdominal surgery were assumed to incur no additional investment. Last, postoperative care—including laboratory tests, pharmacy usage, and the need for a hospital room—were individualized for each route of surgery and applied to the cost.

Costs were estimated in 2008 US dollars, based on procedure costs incurred at Duke University Medical Center.

Physician reimbursement data were obtained from Medicare reimbursement rates for anesthesia and from surgeon Current Procedural Terminology (CPT) codes specific to each procedure.

Quality-of-life assessments were not measured. Nor was the cost to society of the postoperative loss of productivity and wages for each surgical route. Had these losses been recognized, the authors observed, the cost of robotic surgery may have been lower.

The cost of robotic surgery was equivalent to the cost of laparoscopy in only two instances:

  • when the operative time of robotic surgery was reduced to 149 minutes
  • when the cost of robotic disposable items was less than $2,132 (reduced from a baseline cost of $3,293).

WHAT THIS EVIDENCE MEANS FOR PRACTICE

Robotic sacrocolpopexy is costly. this is an important consideration when implementing new technology. cost-saving scenarios are useful to maximize patient benefit and minimize financial burden.

We want to hear from you! Tell us what you think.

References

1. Rooney K, Kenton K, Mueller ER, FitzGerald MP, Brubaker L. Advanced anterior vaginal wall prolapse is highly correlated with apical prolapse. Am J Obstet Gynecol. 2006;195(6):1837-1840.

2. Addison WA, Timmons MC. Abdominal approach to vaginal eversion. Clin Obstet Gynecol. 1993;36(4):995-1004.

Article PDF
Author and Disclosure Information

Cindy L. Amundsen, MD
Dr. Amundsen is Associate Professor of Obstetrics and Gynecology, Division of Urogynecology; Assistant Professor in the Division of Urology; and Fellowship Director of Female Pelvic Medicine and Reconstructive Surgery, Duke University Medical Center, Durham, NC.

Amie Kawasaki, MD
Dr. Kawasaki is a Fellow in Female Pelvic Medicine and Reconstructive Surgery, Division of Urogynecology, Department of Obstetrics and Gynecology, Duke University Medical Center, Durham, NC.

The authors report no financial relationships relevant to this article.

Issue
OBG Management - 22(10)
Publications
Topics
Page Number
20-29
Sections
Author and Disclosure Information

Cindy L. Amundsen, MD
Dr. Amundsen is Associate Professor of Obstetrics and Gynecology, Division of Urogynecology; Assistant Professor in the Division of Urology; and Fellowship Director of Female Pelvic Medicine and Reconstructive Surgery, Duke University Medical Center, Durham, NC.

Amie Kawasaki, MD
Dr. Kawasaki is a Fellow in Female Pelvic Medicine and Reconstructive Surgery, Division of Urogynecology, Department of Obstetrics and Gynecology, Duke University Medical Center, Durham, NC.

The authors report no financial relationships relevant to this article.

Author and Disclosure Information

Cindy L. Amundsen, MD
Dr. Amundsen is Associate Professor of Obstetrics and Gynecology, Division of Urogynecology; Assistant Professor in the Division of Urology; and Fellowship Director of Female Pelvic Medicine and Reconstructive Surgery, Duke University Medical Center, Durham, NC.

Amie Kawasaki, MD
Dr. Kawasaki is a Fellow in Female Pelvic Medicine and Reconstructive Surgery, Division of Urogynecology, Department of Obstetrics and Gynecology, Duke University Medical Center, Durham, NC.

The authors report no financial relationships relevant to this article.

Article PDF
Article PDF

When a woman has advanced prolapse of the anterior vaginal wall, it is highly likely that she has apical prolapse as well. Consider a study by Rooney and associates that determined that clinically significant vault prolapse is present in most women who have anterior vaginal prolapse of stage II or higher.1 For that reason, suspension of the vaginal apex should be considered whenever surgical treatment of anterior wall defects is planned.

Sacrocolpopexy involves suspension of the vaginal vault from the anterior longitudinal ligament of the sacrum, using Y-shaped mesh to augment native tissue (FIGURE).2 It is an effective, durable treatment for vaginal apical prolapse. With a success rate approaching 93%, this procedure has become the gold standard for repair of vault prolapse. Among its advantages are maximization of vaginal depth and preservation of a normal vaginal axis.

Sacrocolpopexy preserves the vaginal axis



With the vaginal vault suspended from the anterior longitudinal
ligament of the sacrum, the normal vaginal axis is preserved
and vaginal depth is maximized.

Sacrocolpopexy can be performed via the abdominal, laparoscopic, or robotic-assisted approach (TABLE 1). Minimally invasive techniques are attractive because they involve faster recovery than abdominal sacrocolpopexy does. Minimally invasive techniques have also advanced to the point that they are both effective and durable. However, these advantages must be weighed against the effort required to learn the techniques, as well as their higher cost.

TABLE 1

How the 3 approaches to sacrocolpopexy compare

ApproachAdvantages and disadvantages
AbdominalShortest operative time
No significant Trendelenburg position required
Highest estimated blood loss
Longest length of stay
Low rate of complications
Longest postoperative recovery
Well-established long-term durability
LaparoscopicLonger operative time
Moderate Trendelenburg position required
Lower estimated blood loss
Shorter length of stay
Surgical technique least similar to abdominal procedure
Low rate of complications
Shorter postoperative recovery
Long-term durability less firmly established
Robotic-assistedLongest operative time
Steep Trendelenburg position required
Lower estimated blood loss
Shorter length of stay
Surgical technique resembles that of abdominal approach
Low rate of complications
Shorter postoperative recovery
Long-term durability appears to be good

In this article, we highlight:

  • a comparison of the laparoscopic and abdominal approaches to sacrocolpopexy
  • an investigation of the learning curve associated with robotic-assisted sacrocolpopexy
  • a study exploring the durability of robotic-assisted repair
  • an estimate of the costs associated with each route of operation.

Laparoscopic vs abdominal sacrocolpopexy—how do they compare?

Paraiso MF, Walters MD, Rackley RR, Melek S, Hugney C. Laparoscopic and abdominal sacral colpopexies: a comparative cohort study. Am J Obstet Gynecol. 2005;192(5):1752–1758.

When surgeons at the Cleveland Clinic performed a retrospective cohort study to compare laparoscopic and abdominal sacrocolpopexy, they found significantly longer operative time with the laparoscopic route, with an average difference of 51 minutes (P < .0001). However, the laparoscopic approach was associated with lower blood loss (although there was no difference between groups in hematocrit on postoperative day 1); shorter hospital stay (average of 1.8 days versus 4 days [P < .001]); and comparable rates of intraoperative and postoperative complications.

Details of the trial

Paraiso and colleagues reviewed the medical charts of 56 consecutive patients who had undergone laparoscopic sacrocolpopexy, comparing them with the charts of 61 consecutive patients who had undergone the procedure using the abdominal approach. The operations had been performed between 1998 and 2003 for treatment of posthysterectomy vaginal prolapse.

The groups underwent similar rates of concurrent procedures. The laparotomy group had a significantly higher number of Burch procedures (P = .007), and the laparoscopic group had a significantly higher rate of adhesiolysis (P = .002).

Among the complications noted— which occurred at comparable rates between groups—were cystotomy, enterotomy, need for transfusion, deep-vein thrombosis, ileus, small bowel obstruction, wound infection, ventral hernia, mesh erosion, and recurrent prolapse. One laparoscopic case was converted to laparotomy because of excessive bleeding during the rectopexy portion of the operation.

Laparoscopy may have taken longer than this trial suggests

This study is one of very few well-designed trials comparing laparoscopic sacrocolpopexy to the historical gold standard of abdominal sacrocolpopexy for vault prolapse.

Twenty-eight percent of laparoscopic procedures in this study used tacking devices in lieu of suturing. Had suturing been performed universally, an even greater difference in surgical time may have been observed.

 

 

There may also be differences between groups in the durability of the two types of repair, an outcome not included in this particular study.

WHAT THIS EVIDENCE MEANS FOR PRACTICE

The laparoscopic approach offers a shorter hospital stay with no increase in intraoperative or postoperative complications, compared with abdominal sacrocolpopexy. However, it entails a significantly longer operative time than the abdominal approach does.

How steep is the learning curve for robotic-assisted sacrocolpopexy?

Akl MN, Long JB, Giles DL, et al. Robotic-assisted sacrocolpopexy: technique and learning curve. Surg Endosc. 2009;23(10):2390–2394.

Akl and coworkers reviewed the medical records of all patients who had undergone robotic-assisted sacrocolpopexy at the Mayo Clinics in Arizona and Florida between 2004 and 2007. All operations were performed by the same four urogynecologists, with an average operative time of 197.9 minutes (standard deviation, ± 66.8 minutes). However, after the first 10 cases, the operative time decreased by 64.3 minutes—a decline of 25.4% (P < .01; 95% confidence interval [CI], 16.1–112.4 minutes).

Details of the trial

Researchers collected baseline information on participants’ age, stage of prolapse, and concomitant procedures. They also gathered data on average operative time, estimated blood loss, intraoperative and postoperative complications, conversion to laparotomy, and length of hospitalization.

Of 80 women who had advanced pelvic organ prolapse (stage III/IV) who underwent robotic-assisted sacrocolpopexy, 88% underwent concomitant robotic and vaginal procedures, including robotic supracervical hysterectomy, Burch procedure, paravaginal repair, lysis of adhesions, bilateral salpingooophorectomy, vaginal cystocele or rectocele repair, and placement of a midurethral sling.

Estimated blood loss for the robotic-assisted approach ranged from 25 mL to 300 mL, with a mean loss of 96.8 mL. Average length of hospitalization was 2.6 days. Four cases (5%) were converted to laparotomy because of limited exposure and one intraoperative bladder injury. Other intraoperative complications included small-bowel injury during trocar placement and one ureteral injury. Postoperative complications included one case of ileus and five (6%) vaginal mesh erosions. Three patients developed recurrent prolapse and underwent subsequent correction.

Learning curve could have been measured more precisely

The authors did not specifically measure the learning curve for robotic-assisted sacrocolpopexy, as they took into account the concomitant procedures. For this reason, the decrease in operative time observed after 10 cases may not accurately reflect an improvement in the performance of sacrocolpopexy.

Akl and colleagues consider this detail to be a strength of the study because most women who undergo prolapse surgery have concomitant procedures. However, recording the length of time it took to perform the sacrocolpopexy portion of the procedure would have been more accurate.

The average length of stay approached that of the abdominal route. Length of stay may decline as a surgeon gains experience with the robotic-assisted approach.

WHAT THIS EVIDENCE MEANS FOR PRACTICE

Robotic-assisted sacrocolpopexy has a steep learning curve with respect to technique and surgical time.

Does robotic-assisted sacrocolpopexy provide durable support?

Elliott DS, Krambeck AE, Chow GK. Long-term results of robotic assisted laparoscopic sacrocolpopexy for the treatment of high grade vaginal vault prolapse. J Urol. 2006;176(2):655–659.

Among the few recent series reporting long-term outcomes after robotic-assisted sacrocolpopexy is this observational study from the Mayo Clinic. It involved 30 women who underwent the operation for the treatment of Baden Walker grade 4/4 posthysterectomy vaginal vault prolapse. The authors concluded that advanced prolapse can be treated with robotic-assisted sacrocolpopexy with long-term success and minimal complications.

Details of the trial

Of 30 women in this trial, 52% underwent an anti-incontinence procedure at the time of sacrocolpopexy. Women who had multiple vaginal defects or a history of abdominal surgery were excluded from the study.

Average operative time was 3.1 hours (range, 2.15–4.75 hours) in the early phase of development of operative technique (described in the manuscript) but diminished over time to an average of 2.5 hours.

Twenty-nine patients were discharged from the hospital after an overnight stay. Very few immediate postoperative complications were observed. Two patients experienced mild port-site infections that required outpatient treatment, and one patient had persistent vaginal bleeding from the incision made during the anti-incontinence procedure.

Most patients were followed for at least 1 year

The mean follow-up in this study was 24 months (range, 16–39 months). During this period, 21 women were followed for a full year. Long-term observation revealed that the repair of vault prolapse remained successful in 19 of these women.

One patient experienced recurrent prolapse 7 months after surgery. Another developed a rectocele 9 months after sacrocolpopexy. Vaginal mesh erosions occurred in two patients within 6 months after the procedure; both patients were treated with outpatient resection of the exposed mesh, with no recurrence of the prolapse.

 

 

Although a larger sample size and longer follow-up would be ideal, this study demonstrates a low rate of recurrent prolapse 1 year after the procedure.

WHAT THIS EVIDENCE MEANS FOR PRACTICE

Robotic sacrocolpopexy appears to provide long-term durability for the treatment of advanced vaginal vault prolapse.

How to choose an approach when planning sacrocolpopexy

Depending on where you practice, you may have as many as three options: abdominal, laparoscopic, or robotic-assisted. Here are basic questions you should address when choosing one:

  • How familiar are you with the technique? if the answer is “not much,” you can anticipate that the cost and time required to perform it will be significantly higher.
  • Are the appropriate instruments and surgical team available?
  • Does the patient have comorbidities? Consider, for example, the fact that she may not be able to tolerate a steep Trendelenberg position—required for the robotic-assisted approach—if she has severe cardiac or pulmonary disease. However, if she has a risk of poor wound healing, a large abdominal incision may not be advisable and postoperative immobility can be risky. if she is obese, laparoscopic or robotic port placement is challenging, but visualization and retraction will be easier. The need for anticoagulation is another consideration, as it will affect estimated blood loss and the choice of an incision, among other things.
  • Let’s not forget the patient. Given the pros and cons, what approach does she prefer?

How much do laparoscopic, abdominal, and robotic-assisted sacrocolpopexy cost?

Judd JP, Siddiqui NY, Barnett JC, et al. Cost-minimization analysis of robotic-assisted, laparoscopic, and abdominal sacrocolpopexy. J Minim Invasive Gynecol. 2010;17: 493–499.

This cost-minimization analysis concluded that robotic-assisted sacrocolpopexy incurs the highest hospital charges but is reimbursed by Medicare at a rate similar to reimbursement for the abdominal and laparoscopic routes (TABLE 2).

TABLE 2

Cost of sacrocolpopexy is significant—especially using the robotic approach

ApproachCost of a procedureOperative time, min (range)
Robotic-assisted$8,508328 (130–383)
Laparoscopic$7,353269 (97–334)
Abdominal$5,792170 (110–286)
Source: Judd JP, Siddiqui NY, Barnett JC, Visco AG, Havrilesky LJ, Wu JM. Cost-minimization analysis of robotic-assisted, laparoscopic, and abdominal sacrocolpopexy. J Minim invasive Gynecol. 2010;17(4):493–499.

The analysis accounted for realistic practices, such as the inclusion of concurrent hysterectomy and other procedures.

Details of the trial

Surgeons from Duke University developed a decision-analysis model in which a hypothetical group of women with advanced vaginal prolapse could choose between one of the three routes of sacrocolpopexy: abdominal, laparoscopic, or robotic-assisted. Researchers postulated two different scenarios:

  • the hospital had ownership of a robotic system
  • the hospital invested in the initial purchase and maintenance of such a system.

Researchers reviewed the literature to formulate their estimates of operative time, rate of conversion to laparotomy, rate of transfusion, and length of hospital stay. In addition, the costs of initial anesthesia setup, professional fees, per-minute intraoperative fees, and postanesthesia care were applied to each approach. Operating room costs per minute and the cost of disposable items such as drapes, gowns, gloves, and single-use instruments were added. For the robotic approach, the costs of reusable instruments were distributed across 10 operations. Reusable instruments for laparoscopic and abdominal surgery were assumed to incur no additional investment. Last, postoperative care—including laboratory tests, pharmacy usage, and the need for a hospital room—were individualized for each route of surgery and applied to the cost.

Costs were estimated in 2008 US dollars, based on procedure costs incurred at Duke University Medical Center.

Physician reimbursement data were obtained from Medicare reimbursement rates for anesthesia and from surgeon Current Procedural Terminology (CPT) codes specific to each procedure.

Quality-of-life assessments were not measured. Nor was the cost to society of the postoperative loss of productivity and wages for each surgical route. Had these losses been recognized, the authors observed, the cost of robotic surgery may have been lower.

The cost of robotic surgery was equivalent to the cost of laparoscopy in only two instances:

  • when the operative time of robotic surgery was reduced to 149 minutes
  • when the cost of robotic disposable items was less than $2,132 (reduced from a baseline cost of $3,293).

WHAT THIS EVIDENCE MEANS FOR PRACTICE

Robotic sacrocolpopexy is costly. this is an important consideration when implementing new technology. cost-saving scenarios are useful to maximize patient benefit and minimize financial burden.

We want to hear from you! Tell us what you think.

When a woman has advanced prolapse of the anterior vaginal wall, it is highly likely that she has apical prolapse as well. Consider a study by Rooney and associates that determined that clinically significant vault prolapse is present in most women who have anterior vaginal prolapse of stage II or higher.1 For that reason, suspension of the vaginal apex should be considered whenever surgical treatment of anterior wall defects is planned.

Sacrocolpopexy involves suspension of the vaginal vault from the anterior longitudinal ligament of the sacrum, using Y-shaped mesh to augment native tissue (FIGURE).2 It is an effective, durable treatment for vaginal apical prolapse. With a success rate approaching 93%, this procedure has become the gold standard for repair of vault prolapse. Among its advantages are maximization of vaginal depth and preservation of a normal vaginal axis.

Sacrocolpopexy preserves the vaginal axis



With the vaginal vault suspended from the anterior longitudinal
ligament of the sacrum, the normal vaginal axis is preserved
and vaginal depth is maximized.

Sacrocolpopexy can be performed via the abdominal, laparoscopic, or robotic-assisted approach (TABLE 1). Minimally invasive techniques are attractive because they involve faster recovery than abdominal sacrocolpopexy does. Minimally invasive techniques have also advanced to the point that they are both effective and durable. However, these advantages must be weighed against the effort required to learn the techniques, as well as their higher cost.

TABLE 1

How the 3 approaches to sacrocolpopexy compare

ApproachAdvantages and disadvantages
AbdominalShortest operative time
No significant Trendelenburg position required
Highest estimated blood loss
Longest length of stay
Low rate of complications
Longest postoperative recovery
Well-established long-term durability
LaparoscopicLonger operative time
Moderate Trendelenburg position required
Lower estimated blood loss
Shorter length of stay
Surgical technique least similar to abdominal procedure
Low rate of complications
Shorter postoperative recovery
Long-term durability less firmly established
Robotic-assistedLongest operative time
Steep Trendelenburg position required
Lower estimated blood loss
Shorter length of stay
Surgical technique resembles that of abdominal approach
Low rate of complications
Shorter postoperative recovery
Long-term durability appears to be good

In this article, we highlight:

  • a comparison of the laparoscopic and abdominal approaches to sacrocolpopexy
  • an investigation of the learning curve associated with robotic-assisted sacrocolpopexy
  • a study exploring the durability of robotic-assisted repair
  • an estimate of the costs associated with each route of operation.

Laparoscopic vs abdominal sacrocolpopexy—how do they compare?

Paraiso MF, Walters MD, Rackley RR, Melek S, Hugney C. Laparoscopic and abdominal sacral colpopexies: a comparative cohort study. Am J Obstet Gynecol. 2005;192(5):1752–1758.

When surgeons at the Cleveland Clinic performed a retrospective cohort study to compare laparoscopic and abdominal sacrocolpopexy, they found significantly longer operative time with the laparoscopic route, with an average difference of 51 minutes (P < .0001). However, the laparoscopic approach was associated with lower blood loss (although there was no difference between groups in hematocrit on postoperative day 1); shorter hospital stay (average of 1.8 days versus 4 days [P < .001]); and comparable rates of intraoperative and postoperative complications.

Details of the trial

Paraiso and colleagues reviewed the medical charts of 56 consecutive patients who had undergone laparoscopic sacrocolpopexy, comparing them with the charts of 61 consecutive patients who had undergone the procedure using the abdominal approach. The operations had been performed between 1998 and 2003 for treatment of posthysterectomy vaginal prolapse.

The groups underwent similar rates of concurrent procedures. The laparotomy group had a significantly higher number of Burch procedures (P = .007), and the laparoscopic group had a significantly higher rate of adhesiolysis (P = .002).

Among the complications noted— which occurred at comparable rates between groups—were cystotomy, enterotomy, need for transfusion, deep-vein thrombosis, ileus, small bowel obstruction, wound infection, ventral hernia, mesh erosion, and recurrent prolapse. One laparoscopic case was converted to laparotomy because of excessive bleeding during the rectopexy portion of the operation.

Laparoscopy may have taken longer than this trial suggests

This study is one of very few well-designed trials comparing laparoscopic sacrocolpopexy to the historical gold standard of abdominal sacrocolpopexy for vault prolapse.

Twenty-eight percent of laparoscopic procedures in this study used tacking devices in lieu of suturing. Had suturing been performed universally, an even greater difference in surgical time may have been observed.

 

 

There may also be differences between groups in the durability of the two types of repair, an outcome not included in this particular study.

WHAT THIS EVIDENCE MEANS FOR PRACTICE

The laparoscopic approach offers a shorter hospital stay with no increase in intraoperative or postoperative complications, compared with abdominal sacrocolpopexy. However, it entails a significantly longer operative time than the abdominal approach does.

How steep is the learning curve for robotic-assisted sacrocolpopexy?

Akl MN, Long JB, Giles DL, et al. Robotic-assisted sacrocolpopexy: technique and learning curve. Surg Endosc. 2009;23(10):2390–2394.

Akl and coworkers reviewed the medical records of all patients who had undergone robotic-assisted sacrocolpopexy at the Mayo Clinics in Arizona and Florida between 2004 and 2007. All operations were performed by the same four urogynecologists, with an average operative time of 197.9 minutes (standard deviation, ± 66.8 minutes). However, after the first 10 cases, the operative time decreased by 64.3 minutes—a decline of 25.4% (P < .01; 95% confidence interval [CI], 16.1–112.4 minutes).

Details of the trial

Researchers collected baseline information on participants’ age, stage of prolapse, and concomitant procedures. They also gathered data on average operative time, estimated blood loss, intraoperative and postoperative complications, conversion to laparotomy, and length of hospitalization.

Of 80 women who had advanced pelvic organ prolapse (stage III/IV) who underwent robotic-assisted sacrocolpopexy, 88% underwent concomitant robotic and vaginal procedures, including robotic supracervical hysterectomy, Burch procedure, paravaginal repair, lysis of adhesions, bilateral salpingooophorectomy, vaginal cystocele or rectocele repair, and placement of a midurethral sling.

Estimated blood loss for the robotic-assisted approach ranged from 25 mL to 300 mL, with a mean loss of 96.8 mL. Average length of hospitalization was 2.6 days. Four cases (5%) were converted to laparotomy because of limited exposure and one intraoperative bladder injury. Other intraoperative complications included small-bowel injury during trocar placement and one ureteral injury. Postoperative complications included one case of ileus and five (6%) vaginal mesh erosions. Three patients developed recurrent prolapse and underwent subsequent correction.

Learning curve could have been measured more precisely

The authors did not specifically measure the learning curve for robotic-assisted sacrocolpopexy, as they took into account the concomitant procedures. For this reason, the decrease in operative time observed after 10 cases may not accurately reflect an improvement in the performance of sacrocolpopexy.

Akl and colleagues consider this detail to be a strength of the study because most women who undergo prolapse surgery have concomitant procedures. However, recording the length of time it took to perform the sacrocolpopexy portion of the procedure would have been more accurate.

The average length of stay approached that of the abdominal route. Length of stay may decline as a surgeon gains experience with the robotic-assisted approach.

WHAT THIS EVIDENCE MEANS FOR PRACTICE

Robotic-assisted sacrocolpopexy has a steep learning curve with respect to technique and surgical time.

Does robotic-assisted sacrocolpopexy provide durable support?

Elliott DS, Krambeck AE, Chow GK. Long-term results of robotic assisted laparoscopic sacrocolpopexy for the treatment of high grade vaginal vault prolapse. J Urol. 2006;176(2):655–659.

Among the few recent series reporting long-term outcomes after robotic-assisted sacrocolpopexy is this observational study from the Mayo Clinic. It involved 30 women who underwent the operation for the treatment of Baden Walker grade 4/4 posthysterectomy vaginal vault prolapse. The authors concluded that advanced prolapse can be treated with robotic-assisted sacrocolpopexy with long-term success and minimal complications.

Details of the trial

Of 30 women in this trial, 52% underwent an anti-incontinence procedure at the time of sacrocolpopexy. Women who had multiple vaginal defects or a history of abdominal surgery were excluded from the study.

Average operative time was 3.1 hours (range, 2.15–4.75 hours) in the early phase of development of operative technique (described in the manuscript) but diminished over time to an average of 2.5 hours.

Twenty-nine patients were discharged from the hospital after an overnight stay. Very few immediate postoperative complications were observed. Two patients experienced mild port-site infections that required outpatient treatment, and one patient had persistent vaginal bleeding from the incision made during the anti-incontinence procedure.

Most patients were followed for at least 1 year

The mean follow-up in this study was 24 months (range, 16–39 months). During this period, 21 women were followed for a full year. Long-term observation revealed that the repair of vault prolapse remained successful in 19 of these women.

One patient experienced recurrent prolapse 7 months after surgery. Another developed a rectocele 9 months after sacrocolpopexy. Vaginal mesh erosions occurred in two patients within 6 months after the procedure; both patients were treated with outpatient resection of the exposed mesh, with no recurrence of the prolapse.

 

 

Although a larger sample size and longer follow-up would be ideal, this study demonstrates a low rate of recurrent prolapse 1 year after the procedure.

WHAT THIS EVIDENCE MEANS FOR PRACTICE

Robotic sacrocolpopexy appears to provide long-term durability for the treatment of advanced vaginal vault prolapse.

How to choose an approach when planning sacrocolpopexy

Depending on where you practice, you may have as many as three options: abdominal, laparoscopic, or robotic-assisted. Here are basic questions you should address when choosing one:

  • How familiar are you with the technique? if the answer is “not much,” you can anticipate that the cost and time required to perform it will be significantly higher.
  • Are the appropriate instruments and surgical team available?
  • Does the patient have comorbidities? Consider, for example, the fact that she may not be able to tolerate a steep Trendelenberg position—required for the robotic-assisted approach—if she has severe cardiac or pulmonary disease. However, if she has a risk of poor wound healing, a large abdominal incision may not be advisable and postoperative immobility can be risky. if she is obese, laparoscopic or robotic port placement is challenging, but visualization and retraction will be easier. The need for anticoagulation is another consideration, as it will affect estimated blood loss and the choice of an incision, among other things.
  • Let’s not forget the patient. Given the pros and cons, what approach does she prefer?

How much do laparoscopic, abdominal, and robotic-assisted sacrocolpopexy cost?

Judd JP, Siddiqui NY, Barnett JC, et al. Cost-minimization analysis of robotic-assisted, laparoscopic, and abdominal sacrocolpopexy. J Minim Invasive Gynecol. 2010;17: 493–499.

This cost-minimization analysis concluded that robotic-assisted sacrocolpopexy incurs the highest hospital charges but is reimbursed by Medicare at a rate similar to reimbursement for the abdominal and laparoscopic routes (TABLE 2).

TABLE 2

Cost of sacrocolpopexy is significant—especially using the robotic approach

ApproachCost of a procedureOperative time, min (range)
Robotic-assisted$8,508328 (130–383)
Laparoscopic$7,353269 (97–334)
Abdominal$5,792170 (110–286)
Source: Judd JP, Siddiqui NY, Barnett JC, Visco AG, Havrilesky LJ, Wu JM. Cost-minimization analysis of robotic-assisted, laparoscopic, and abdominal sacrocolpopexy. J Minim invasive Gynecol. 2010;17(4):493–499.

The analysis accounted for realistic practices, such as the inclusion of concurrent hysterectomy and other procedures.

Details of the trial

Surgeons from Duke University developed a decision-analysis model in which a hypothetical group of women with advanced vaginal prolapse could choose between one of the three routes of sacrocolpopexy: abdominal, laparoscopic, or robotic-assisted. Researchers postulated two different scenarios:

  • the hospital had ownership of a robotic system
  • the hospital invested in the initial purchase and maintenance of such a system.

Researchers reviewed the literature to formulate their estimates of operative time, rate of conversion to laparotomy, rate of transfusion, and length of hospital stay. In addition, the costs of initial anesthesia setup, professional fees, per-minute intraoperative fees, and postanesthesia care were applied to each approach. Operating room costs per minute and the cost of disposable items such as drapes, gowns, gloves, and single-use instruments were added. For the robotic approach, the costs of reusable instruments were distributed across 10 operations. Reusable instruments for laparoscopic and abdominal surgery were assumed to incur no additional investment. Last, postoperative care—including laboratory tests, pharmacy usage, and the need for a hospital room—were individualized for each route of surgery and applied to the cost.

Costs were estimated in 2008 US dollars, based on procedure costs incurred at Duke University Medical Center.

Physician reimbursement data were obtained from Medicare reimbursement rates for anesthesia and from surgeon Current Procedural Terminology (CPT) codes specific to each procedure.

Quality-of-life assessments were not measured. Nor was the cost to society of the postoperative loss of productivity and wages for each surgical route. Had these losses been recognized, the authors observed, the cost of robotic surgery may have been lower.

The cost of robotic surgery was equivalent to the cost of laparoscopy in only two instances:

  • when the operative time of robotic surgery was reduced to 149 minutes
  • when the cost of robotic disposable items was less than $2,132 (reduced from a baseline cost of $3,293).

WHAT THIS EVIDENCE MEANS FOR PRACTICE

Robotic sacrocolpopexy is costly. this is an important consideration when implementing new technology. cost-saving scenarios are useful to maximize patient benefit and minimize financial burden.

We want to hear from you! Tell us what you think.

References

1. Rooney K, Kenton K, Mueller ER, FitzGerald MP, Brubaker L. Advanced anterior vaginal wall prolapse is highly correlated with apical prolapse. Am J Obstet Gynecol. 2006;195(6):1837-1840.

2. Addison WA, Timmons MC. Abdominal approach to vaginal eversion. Clin Obstet Gynecol. 1993;36(4):995-1004.

References

1. Rooney K, Kenton K, Mueller ER, FitzGerald MP, Brubaker L. Advanced anterior vaginal wall prolapse is highly correlated with apical prolapse. Am J Obstet Gynecol. 2006;195(6):1837-1840.

2. Addison WA, Timmons MC. Abdominal approach to vaginal eversion. Clin Obstet Gynecol. 1993;36(4):995-1004.

Issue
OBG Management - 22(10)
Issue
OBG Management - 22(10)
Page Number
20-29
Page Number
20-29
Publications
Publications
Topics
Article Type
Display Headline
UPDATE: PELVIC FLOOR DYSFUNCTION
Display Headline
UPDATE: PELVIC FLOOR DYSFUNCTION
Sections
Article Source

PURLs Copyright

Inside the Article
Article PDF Media

Skilled US imaging of the adnexae: The non-neoplastic mass

Article Type
Changed
Tue, 08/28/2018 - 10:58
Display Headline
Skilled US imaging of the adnexae: The non-neoplastic mass

READ THE WHOLE SERIES

Part 1: A Starting Point (September 2010)

Part 3: Ovarian neoplasms (November 2010)

Part 4: The fallopian tubes (December 2010)

Scanning the ovaries is no simple task. As we mentioned in Part 1 of this four-part series, the practitioner must use the right equipment, take basic preparatory steps, be watchful for clues in the history, and reach a conclusion about what he or she sees. Not only that: The ultrasonographer must be extraordinarily vigilant, paying close attention to multiple characteristics of any mass, from thickness of the wall to the presence of papillations or a blood supply—signs of potential malignancy.

In this article, we detail the traits of various types of non-neoplastic ovarian masses, including:

  • functional cysts—follicles, the corpus luteum, and theca lutein cysts
  • nonfunctional cysts—serous masses and endometriomas
  • cystadenofibromas. Although these masses are usually categorized histo-logically as neoplasms, we include them here due to their almost daily appearance in a busy gynecologic ultrasonographic (US) facility.

In Part 3, we will cover ovarian neoplasms, and in Part 4, our focus will be tubal entities such as ectopic pregnancy and torsion.


FIGURE 1 What is a mass made of? 6 morphologic building blocks

Take an inventory of the mass

Any adnexal mass should be assessed in light of its essential characteristics (Figure 1).

Wall structure. Pay attention to thickness. We use an arbitrary cutoff of 4 mm, giving extra scrutiny to thicknesses exceeding that measurement. In our experience, the thicker the wall, the more likely the mass is to be malignant.

Septation and loculation. A mass is typically unilocular or multilocular. Multilocularity is more common in tumors of low malignant potential and malignant neoplasms.

Papillation. Any internal or external papillae or excrescences should draw your attention. Papillarity in an ovarian mass renders that mass suspicious for malignancy.

Measure (height and width) any papillae that are identified, and document them. Because papillae are associated with ovarian malignancy, further assessment is warranted immediately. The first step is determining whether the papillations contain blood vessels—a task for which color and power Doppler are helpful. We prefer power Doppler because it is more sensitive, detecting blood-flow velocity in the lowest detectable range of 2 cm/s, and because it is not directionally influenced.

Papillae that contain blood vessels with detectable flow are suspicious for malignancy.

Exacoustos and colleagues found that papillae as large as 15 mm in height and 10 mm in width (base) were present in 48% of borderline ovarian tumors but in only 4% of benign and 4% of malignant tumors. However, when the intracystic solid tissue exceeded those dimensions, the lesions were present in 48% of invasive ovarian tumors, 18% of borderline ovarian tumors, and 7% of benign masses.1

Internal echo-structure. A mass can be anechoic, a finding that usually indicates the presence of clear fluid. Mostly solid masses are echogenic. And masses that contain particulate matter, such as blood, cellular matter, or even mucous material, usually have echogenicity of a low level, often described as a “ground-glass” appearance. A mass can also have mixed echo-genicity, a finding usually found in cases involving teratoma or malignancy.

Shadowing. If it is present, it may signify the presence of an extremely dense, solid tissue, such as bone or calcification. The diagnosis of a benign teratoma (i.e., dermoid cyst) should be entertained if shadowing is present in a hyperechoic nodule or mass. Malignant masses very rarely, if ever, display frank shadowing.

Overall appearance. On rare occasions, a bizarre shape or “complex” appearance (as it is termed in most radiology reports) may indicate a malignant mass. More likely it indicates the presence of a teratoma, cystadenoma, or even an atypical corpus luteum. In some reports generated by US laboratories, the term “complex” is applied to all structures other than simple cysts.

Size. The size of a mass can be misleading, as small ovarian lesions with the appropriate sonographic characteristics may be malignant and some larger ones without those characteristics may not be. However, it is understood that the larger an ovarian lesion, the more likely it is a tumor. One important distinction: The amount of fluid in a cystic structure or the amount of old blood in an endometrioma is not the disease process…it is the byproduct of the process. So an 8-cm endometrioma may create fewer pain or fertility issues than a 2- or 3-cm endometrioma. Similarly, the amount of “chocolate” fluid is not automatically indicative of the amount of active endometriotic glands or their sequelae!2

Ascites. If it is present, it should be recorded and investigated further because it may be caused by a malignant intra-abdominal tumor.

 

 

Motion tenderness. If the to-and-fro movement of the vaginal probe elicits any motion tenderness, it, too, should be documented. It may be a sign of pelvic peritonitis. In such cases, an “ominous appearing” adnexal finding may represent an inflammatory, rather than malignant, mass.

When to use Doppler

One of the components of extensive evaluation of the adnexae in general and ovaries in particular is color or power Doppler interrogation—or both.

Tumors contain a relatively large number of pathologic blood vessels that lack the muscular layer found in normal blood vessels and, as a result, demonstrate lower resistance to flow. Diastolic flow is high in these vessels, and resistance and pulsatility indices are low.

We also pay attention when these blood vessels have a tortuous appearance, changes in caliber, anastomoses, and vascular lakes.3 The more tortuous the vessels, with multiple inter-vessel connections and dilatations with changing calibers, the greater the risk of malignancy.4 No less important is the presence of a vessel within a “complex” ovarian mass. A centrally located vessel (also called a “lead vessel”) is suspicious for malignancy.5

A gallery of non-neoplastic ovarian masses

Non-neoplastic cysts are, by far, the most common structures of the ovary. They may be functional, as in the case of the follicles, corpus luteum, and theca lutein cysts, or they may be nonfunctional, as in serous cysts and endometriomas. (As we noted in Part 1, do not call the follicles and corpus luteum “cysts” because this designation suggests pathology.)6


FIGURE 2 Simple cyst

This cyst is anechoic and unilocular with thin walls and no papillae.

Functional cysts

Functional cysts, also known as “simple” cysts, may grow as large as 4 to 5 cm in diameter (Figure 2). They are typically unilocular, anechoic, and thin-walled, with no papillae, and almost never malignant. They usually resolve and require no treatment unless rupture or torsion occurs. Except for the corpus luteum, they have no increased blood flow, and need be viewed only by transvaginal ultrasonography (TVS).

The corpus luteum also can be recognized by TVS. It can exhibit any of a variety of internal structures and echo patterns, due to the multitude of shapes of the blood or clot that can be seen within it (Figure 3).


FIGURE 3 Corpus luteum

A–C. Gray-scale, color Doppler, and power Doppler images, respectively, of a typical corpus luteum. B and C show the enveloping vessels, or “ring of fire.” D. A rather typical gray-scale appearance with a mesh-like, linear internal texture. E. A common feature of the corpus luteum is a linear interphase (arrow) between the clot (c) and the liquified serum (s).

The corpus luteum is typically enveloped by blood vessels, visible on color Doppler as what is called a “ring of fire.” It regresses without intervention. In hyperstimulated ovaries, however, more than one may be present; this poses a real diagnostic challenge when ectopic pregnancy is suspected because it is difficult to differentiate the two entities.

Because the corpus luteum can sometimes resemble some types of ovarian tumors on TVS, imaging during the secretory phase of the cycle in a woman of reproductive age is not ideal. Instead, she should be scanned (or rescanned) between days 5 and 9 of the cycle.


FIGURE 4 Hormonally stimulated ovaries

A, B. The right and left ovaries stimulated by follicle-stimulating hormone preparation (arrow points to hilus). C. An ovary stimulated by clomiphene.

Lutein cysts may reach 5 to 10 cm in diameter. They generally have a thick wall, are multilocular, and typically occur after hormonal induction of ovulation (Figure 4). They also can occur in diabetes, molar pregnancy, and hydrops fetalis. We have seen a unilateral theca lutein cyst in a normal pregnancy (Figure 5). No treatment is necessary unless rupture or torsion occurs.


FIGURE 5 Lutein cysts

A–C. The typical “stained glass” appearance of three lutein cysts of the right ovary in a pregnant patient. D. Color Doppler image of the ovary demonstrating high-velocity flow (peak systolic velocity of 20.4 cm/s).

Serous cysts

These cysts can reach 4 cm in diameter, have smooth walls with no papillae, are unilocular, and occur most often during menopause. No pathological blood flow is visible in their walls. Most gynecologists follow them (Figure 6).1,7


FIGURE 6 Serous cyst

A. Right ovary containing the cyst. B. Normal left ovary. C. Power Doppler interrogation showing no particular flow in the walls of a serous cyst.

Endometriomas

After the simple cyst, the endometrioma is the most prevalent ovarian or adnexal cyst (Figure 7). It usually has a thick wall and is filled with homogeneous fluid with low-level echo-genicity. It can reach 10 cm in size, and many are bilateral. It is sometimes called a “chocolate” cyst because of its dark blood content.


 

 

FIGURE 7 Endometriomas

Endometriomas have low echogenicity. A. Unilateral, unilocular cyst with thin walls. B. Bilateral endometriomas. C. Blood flow in a solid or papillary component of the endometrioma is an occasional finding. It should be investigated further because of the risk that it represents endometrioid cancer.

Endometriomas do not resolve; they usually require surgical excision, although very small ones wholly contained within an ovary are often managed medically or expectantly.

These masses rarely (<1%) give rise to endometrioid carcinoma. Should an endometrioma contain papillae with blood vessels, it is extremely suspicious for endometrioid cancer.


FIGURE 8 Cystic fibromas

A. Sonographic image shows a thin wall and hyperechoic, small mural nodules. B. Macroscopic appearance of an area of internal papillary excrescences. C. Measurement of the small, mural nodules. D. Lack of blood flow in the small papillae, a typical finding on color or power Doppler. E, F. Blood flow in the wall of the cyst and in the mural nodules.

Ovarian fibromas

A fibroma is a slow-growing, benign, solid ovarian tumor. It usually has a cystic component and then is called a cystadenofibroma.

The cystic variety is filled with anechoic fluid and has a thin wall. However, its pathognomonic feature is the small (2–3 mm), extremely hyperechoic mural nodules (papillae) it contains (Figure 8A–C). In the overwhelming majority of cases, no blood vessels are detectable, and the mass is unilocular (Figure 8D–E). It can be recognized in the ovary by the semilunar shape of the tissue surrounding it (crescent sign). The differential diagnosis includes the simple (serous) cyst.

The solid fibroma has a myometrium-like texture, with few or no detectable blood vessels in the stroma. The differential diagnosis includes the Brenner tumor and the Krukenberg tumor.

Federal health agency weighs in on adnexal mass assessment

According to a technology assessment from the Agency for Healthcare Research and Quality (AHRQ), “conventional gray-scale ultrasonography is the most common imaging modality used to differentiate benign from malignant adnexal masses. Especially with the advent of high-frequency transvaginal probes, the quality of the images allows description of the gross anatomic features of the lesion.”8 This descriptive ability is limited, however, “by the great variability of macroscopic characteristics of both benign and malignant masses. Furthermore, the technique is operator dependent.”8

To overcome these challenges, some experts have developed ultrasonographic (US) morphologic scoring systems, which assign a value to individual characteristics. Lerner and colleagues devised a 4-point system:

CharacteristicPoints
 0123
Wall structureSmooth or small irregularities (<3 mm) Solid or not applicablePapillarities larger than 3 mm
ShadowingYesNo  
SeptationNone or thin (<3 mm)Thick (≥3 mm)  
EchogenicitySonolucent or low-level echo or echogenic core Mixed or high 

The mean point value for benign masses was 1.8; for tumors of low malignant potential it was 3.9; and for malignant tumors it was 5.6 (P < .0005). Lerner and associates proposed a cutoff of 3. A score of 3 or higher, they felt, would be most predictive of malignancy, with sensitivity of 96.8% and specificity of 77%. Positive and negative predictive values were 29.4% and 99.6%, respectively.9

Almost all published scoring systems are based upon or derived from one reported by Sassone and coworkers.10 The most important and practical feature of all scoring systems is their ability to rule out malignancy.

Morphology and Doppler: A synergistic combination

As the same AHRQ report points out, “all of the diagnostic tests and scoring systems we evaluated exhibited a trade-off between sensitivity and specificity—studies of a given test that reported higher sensitivity had lower specificity, and vice versa.”8 Among evaluation methods, the combination of US morphology scores and Doppler imaging achieved the highest pooled sensitivity and specificity scores in distinguishing benign and malignant adnexal masses in postmenopausal women: 86% and 91%, respectively, according to the AHRQ report.8

Compare these figures with those of:

  • Bimanual pelvic examination (45% and 90%, respectively)
  • Doppler resistance index (72% and 90%)
  • Doppler pulsatility index (80% and 73%)
  • presence of blood vessels (88% and 78%).

The combination of US morphology scores and Doppler was comparable to the pooled sensitivity and specificity of magnetic resonance imaging (91% and 88%, respectively) and superior to computed tomography (90% and 75%, respectively).

Why the need to know?

Discrimination between benign and malignant masses serves a number of purposes, depending on the setting.

For example, if a symptomatic woman is found to have an adnexal mass, it is important to identify the type of mass causing the symptoms to determine the best course of treatment. And because surgery may be one of the treatment options, it is helpful to know whether a mass is likely to be malignant so that the patient can be referred to a specialist or center that has optimal surgical expertise.8

Some asymptomatic masses may be identified during the annual bimanual pelvic examination recommended by ACOG or during pregnancy-related US imaging. In this setting, it is important to ascertain whether the mass is likely to be malignant so that the patient can be referred to a specialist, if necessary. In addition, thorough assessment of the mass can help “avoid unnecessary diagnostic procedures, including surgery, and anxiety in women with asymptomatic, nonmalignant conditions. In some cases, there may be a rationale for removing certain asymptomatic benign lesions, including prevention of malignant transformation; prevention of ovarian torsion”; and prevention of rupture. Surgery may also be appropriate to avert the need for more complicated surgery in the future or to enhance fertility.8Janelle Yates, Senior Editor

 

 

Stay tuned!

Next issue, in Part 3 of this series, we will review the use of imaging in the investigation of ovarian neoplasms, both benign and malignant, with an abundance of US images to accompany our discussion.

We want to hear from you! Tell us what you think.

References

1. Exacoustos C, Romanini ME, Rinaldo D, et al. Preoperative sonographic features of borderline ovarian tumors. Ultrasound Obstet Gynecol. 2004;25(1):50-59.

2. Rulin MC, Preston AL. Adnexal masses in postmenopausal women. Obstet Gynecol. 1987;70(4):578-581.

3. Timor-Tritsch IE, Goldstein SR. The complexity of a complex mass and the simplicity of a simple cyst. J Ultraound Med. 2005;24(3):255-258.

4. Sladkevicius P, Jokubkiene L, Valentin L. Contribution of morphological assessment of the vessel tree by three-dimensional ultrasound to a correct diagnos is of malignancy in ovarian masses. Ultrasound Obstet Gynecol. 2007;30(6):874-882.

5. Testa AC, Mancari R, Di Legge A, et al. The “lead vessel”: a vascular ultrasound feature of metastasis in the ovaries. Ultrasound Obstet Gynecol. 2008;31:218-221.

6. Goldstein SR. Postmenopausal adnexal cysts: how clinical management has evolved. Am J Obstet Gynecol. 1996;175(6):1496-1501.

7. Levine D, Gosink BB, Wolf S, Feldesman MR, Pretorius D. Simple adnexal cysts: the natural history in postmenopausal women. Radiology. 1992;184(3):653-659.

8. Myers ER, Bastian LA, Havrilesky LJ, et al. Management of adnexal mass. Evidence Report Technol Assess. 2006;Feb;(130):1-145.

9. Lerner JP, Timor-Tritsch IE, Federman A, Abramovich G. Transvaginal ultrasonographic characterization of ovarian masses with an improved, weighted scoring system. Am J Obstet Gynecol. 1994;170(1 Pt 1):81-85.

10. Sassone AM, Timor-Tritsch IE, Artner A, et al. Transvaginal sonographic characterization of ovarian disease: evaluation of a new scoring system to predict ovarian malignancy. Obstet Gynecol. 2001;78:70-76.

Article PDF
Author and Disclosure Information

Ilan E. Timor-Tritsch, MD
Dr. Timor-Tritsch is Professor of ObGyn and Director of ObGyn Ultrasound at New York University Medical Center in New York City.

Steven R. Goldstein, MD
Dr. Goldstein is Professor of ObGyn, Director of Gynecologic Ultrasound, and Co-Director of Bone Densitometry at New York University Medical Center in New York City. He serves on the Board of Editors.

Dr. Timor-Tritsch reports no financial relationships relevant to this article. Dr. Goldstein reports that he is an advisor to Amgen, Boehringer Ingelheim, Eli Lilly, Novo Nordisk, Merck, and Pfizer. He also serves as a consultant to Cook ObGyn and Philips Ultrasound and as a speaker for Eli Lilly and Warner Chilcott. He is a director of Sonosite, Inc.

Issue
OBG Management - 22(10)
Publications
Page Number
44-56
Sections
Author and Disclosure Information

Ilan E. Timor-Tritsch, MD
Dr. Timor-Tritsch is Professor of ObGyn and Director of ObGyn Ultrasound at New York University Medical Center in New York City.

Steven R. Goldstein, MD
Dr. Goldstein is Professor of ObGyn, Director of Gynecologic Ultrasound, and Co-Director of Bone Densitometry at New York University Medical Center in New York City. He serves on the Board of Editors.

Dr. Timor-Tritsch reports no financial relationships relevant to this article. Dr. Goldstein reports that he is an advisor to Amgen, Boehringer Ingelheim, Eli Lilly, Novo Nordisk, Merck, and Pfizer. He also serves as a consultant to Cook ObGyn and Philips Ultrasound and as a speaker for Eli Lilly and Warner Chilcott. He is a director of Sonosite, Inc.

Author and Disclosure Information

Ilan E. Timor-Tritsch, MD
Dr. Timor-Tritsch is Professor of ObGyn and Director of ObGyn Ultrasound at New York University Medical Center in New York City.

Steven R. Goldstein, MD
Dr. Goldstein is Professor of ObGyn, Director of Gynecologic Ultrasound, and Co-Director of Bone Densitometry at New York University Medical Center in New York City. He serves on the Board of Editors.

Dr. Timor-Tritsch reports no financial relationships relevant to this article. Dr. Goldstein reports that he is an advisor to Amgen, Boehringer Ingelheim, Eli Lilly, Novo Nordisk, Merck, and Pfizer. He also serves as a consultant to Cook ObGyn and Philips Ultrasound and as a speaker for Eli Lilly and Warner Chilcott. He is a director of Sonosite, Inc.

Article PDF
Article PDF

READ THE WHOLE SERIES

Part 1: A Starting Point (September 2010)

Part 3: Ovarian neoplasms (November 2010)

Part 4: The fallopian tubes (December 2010)

Scanning the ovaries is no simple task. As we mentioned in Part 1 of this four-part series, the practitioner must use the right equipment, take basic preparatory steps, be watchful for clues in the history, and reach a conclusion about what he or she sees. Not only that: The ultrasonographer must be extraordinarily vigilant, paying close attention to multiple characteristics of any mass, from thickness of the wall to the presence of papillations or a blood supply—signs of potential malignancy.

In this article, we detail the traits of various types of non-neoplastic ovarian masses, including:

  • functional cysts—follicles, the corpus luteum, and theca lutein cysts
  • nonfunctional cysts—serous masses and endometriomas
  • cystadenofibromas. Although these masses are usually categorized histo-logically as neoplasms, we include them here due to their almost daily appearance in a busy gynecologic ultrasonographic (US) facility.

In Part 3, we will cover ovarian neoplasms, and in Part 4, our focus will be tubal entities such as ectopic pregnancy and torsion.


FIGURE 1 What is a mass made of? 6 morphologic building blocks

Take an inventory of the mass

Any adnexal mass should be assessed in light of its essential characteristics (Figure 1).

Wall structure. Pay attention to thickness. We use an arbitrary cutoff of 4 mm, giving extra scrutiny to thicknesses exceeding that measurement. In our experience, the thicker the wall, the more likely the mass is to be malignant.

Septation and loculation. A mass is typically unilocular or multilocular. Multilocularity is more common in tumors of low malignant potential and malignant neoplasms.

Papillation. Any internal or external papillae or excrescences should draw your attention. Papillarity in an ovarian mass renders that mass suspicious for malignancy.

Measure (height and width) any papillae that are identified, and document them. Because papillae are associated with ovarian malignancy, further assessment is warranted immediately. The first step is determining whether the papillations contain blood vessels—a task for which color and power Doppler are helpful. We prefer power Doppler because it is more sensitive, detecting blood-flow velocity in the lowest detectable range of 2 cm/s, and because it is not directionally influenced.

Papillae that contain blood vessels with detectable flow are suspicious for malignancy.

Exacoustos and colleagues found that papillae as large as 15 mm in height and 10 mm in width (base) were present in 48% of borderline ovarian tumors but in only 4% of benign and 4% of malignant tumors. However, when the intracystic solid tissue exceeded those dimensions, the lesions were present in 48% of invasive ovarian tumors, 18% of borderline ovarian tumors, and 7% of benign masses.1

Internal echo-structure. A mass can be anechoic, a finding that usually indicates the presence of clear fluid. Mostly solid masses are echogenic. And masses that contain particulate matter, such as blood, cellular matter, or even mucous material, usually have echogenicity of a low level, often described as a “ground-glass” appearance. A mass can also have mixed echo-genicity, a finding usually found in cases involving teratoma or malignancy.

Shadowing. If it is present, it may signify the presence of an extremely dense, solid tissue, such as bone or calcification. The diagnosis of a benign teratoma (i.e., dermoid cyst) should be entertained if shadowing is present in a hyperechoic nodule or mass. Malignant masses very rarely, if ever, display frank shadowing.

Overall appearance. On rare occasions, a bizarre shape or “complex” appearance (as it is termed in most radiology reports) may indicate a malignant mass. More likely it indicates the presence of a teratoma, cystadenoma, or even an atypical corpus luteum. In some reports generated by US laboratories, the term “complex” is applied to all structures other than simple cysts.

Size. The size of a mass can be misleading, as small ovarian lesions with the appropriate sonographic characteristics may be malignant and some larger ones without those characteristics may not be. However, it is understood that the larger an ovarian lesion, the more likely it is a tumor. One important distinction: The amount of fluid in a cystic structure or the amount of old blood in an endometrioma is not the disease process…it is the byproduct of the process. So an 8-cm endometrioma may create fewer pain or fertility issues than a 2- or 3-cm endometrioma. Similarly, the amount of “chocolate” fluid is not automatically indicative of the amount of active endometriotic glands or their sequelae!2

Ascites. If it is present, it should be recorded and investigated further because it may be caused by a malignant intra-abdominal tumor.

 

 

Motion tenderness. If the to-and-fro movement of the vaginal probe elicits any motion tenderness, it, too, should be documented. It may be a sign of pelvic peritonitis. In such cases, an “ominous appearing” adnexal finding may represent an inflammatory, rather than malignant, mass.

When to use Doppler

One of the components of extensive evaluation of the adnexae in general and ovaries in particular is color or power Doppler interrogation—or both.

Tumors contain a relatively large number of pathologic blood vessels that lack the muscular layer found in normal blood vessels and, as a result, demonstrate lower resistance to flow. Diastolic flow is high in these vessels, and resistance and pulsatility indices are low.

We also pay attention when these blood vessels have a tortuous appearance, changes in caliber, anastomoses, and vascular lakes.3 The more tortuous the vessels, with multiple inter-vessel connections and dilatations with changing calibers, the greater the risk of malignancy.4 No less important is the presence of a vessel within a “complex” ovarian mass. A centrally located vessel (also called a “lead vessel”) is suspicious for malignancy.5

A gallery of non-neoplastic ovarian masses

Non-neoplastic cysts are, by far, the most common structures of the ovary. They may be functional, as in the case of the follicles, corpus luteum, and theca lutein cysts, or they may be nonfunctional, as in serous cysts and endometriomas. (As we noted in Part 1, do not call the follicles and corpus luteum “cysts” because this designation suggests pathology.)6


FIGURE 2 Simple cyst

This cyst is anechoic and unilocular with thin walls and no papillae.

Functional cysts

Functional cysts, also known as “simple” cysts, may grow as large as 4 to 5 cm in diameter (Figure 2). They are typically unilocular, anechoic, and thin-walled, with no papillae, and almost never malignant. They usually resolve and require no treatment unless rupture or torsion occurs. Except for the corpus luteum, they have no increased blood flow, and need be viewed only by transvaginal ultrasonography (TVS).

The corpus luteum also can be recognized by TVS. It can exhibit any of a variety of internal structures and echo patterns, due to the multitude of shapes of the blood or clot that can be seen within it (Figure 3).


FIGURE 3 Corpus luteum

A–C. Gray-scale, color Doppler, and power Doppler images, respectively, of a typical corpus luteum. B and C show the enveloping vessels, or “ring of fire.” D. A rather typical gray-scale appearance with a mesh-like, linear internal texture. E. A common feature of the corpus luteum is a linear interphase (arrow) between the clot (c) and the liquified serum (s).

The corpus luteum is typically enveloped by blood vessels, visible on color Doppler as what is called a “ring of fire.” It regresses without intervention. In hyperstimulated ovaries, however, more than one may be present; this poses a real diagnostic challenge when ectopic pregnancy is suspected because it is difficult to differentiate the two entities.

Because the corpus luteum can sometimes resemble some types of ovarian tumors on TVS, imaging during the secretory phase of the cycle in a woman of reproductive age is not ideal. Instead, she should be scanned (or rescanned) between days 5 and 9 of the cycle.


FIGURE 4 Hormonally stimulated ovaries

A, B. The right and left ovaries stimulated by follicle-stimulating hormone preparation (arrow points to hilus). C. An ovary stimulated by clomiphene.

Lutein cysts may reach 5 to 10 cm in diameter. They generally have a thick wall, are multilocular, and typically occur after hormonal induction of ovulation (Figure 4). They also can occur in diabetes, molar pregnancy, and hydrops fetalis. We have seen a unilateral theca lutein cyst in a normal pregnancy (Figure 5). No treatment is necessary unless rupture or torsion occurs.


FIGURE 5 Lutein cysts

A–C. The typical “stained glass” appearance of three lutein cysts of the right ovary in a pregnant patient. D. Color Doppler image of the ovary demonstrating high-velocity flow (peak systolic velocity of 20.4 cm/s).

Serous cysts

These cysts can reach 4 cm in diameter, have smooth walls with no papillae, are unilocular, and occur most often during menopause. No pathological blood flow is visible in their walls. Most gynecologists follow them (Figure 6).1,7


FIGURE 6 Serous cyst

A. Right ovary containing the cyst. B. Normal left ovary. C. Power Doppler interrogation showing no particular flow in the walls of a serous cyst.

Endometriomas

After the simple cyst, the endometrioma is the most prevalent ovarian or adnexal cyst (Figure 7). It usually has a thick wall and is filled with homogeneous fluid with low-level echo-genicity. It can reach 10 cm in size, and many are bilateral. It is sometimes called a “chocolate” cyst because of its dark blood content.


 

 

FIGURE 7 Endometriomas

Endometriomas have low echogenicity. A. Unilateral, unilocular cyst with thin walls. B. Bilateral endometriomas. C. Blood flow in a solid or papillary component of the endometrioma is an occasional finding. It should be investigated further because of the risk that it represents endometrioid cancer.

Endometriomas do not resolve; they usually require surgical excision, although very small ones wholly contained within an ovary are often managed medically or expectantly.

These masses rarely (<1%) give rise to endometrioid carcinoma. Should an endometrioma contain papillae with blood vessels, it is extremely suspicious for endometrioid cancer.


FIGURE 8 Cystic fibromas

A. Sonographic image shows a thin wall and hyperechoic, small mural nodules. B. Macroscopic appearance of an area of internal papillary excrescences. C. Measurement of the small, mural nodules. D. Lack of blood flow in the small papillae, a typical finding on color or power Doppler. E, F. Blood flow in the wall of the cyst and in the mural nodules.

Ovarian fibromas

A fibroma is a slow-growing, benign, solid ovarian tumor. It usually has a cystic component and then is called a cystadenofibroma.

The cystic variety is filled with anechoic fluid and has a thin wall. However, its pathognomonic feature is the small (2–3 mm), extremely hyperechoic mural nodules (papillae) it contains (Figure 8A–C). In the overwhelming majority of cases, no blood vessels are detectable, and the mass is unilocular (Figure 8D–E). It can be recognized in the ovary by the semilunar shape of the tissue surrounding it (crescent sign). The differential diagnosis includes the simple (serous) cyst.

The solid fibroma has a myometrium-like texture, with few or no detectable blood vessels in the stroma. The differential diagnosis includes the Brenner tumor and the Krukenberg tumor.

Federal health agency weighs in on adnexal mass assessment

According to a technology assessment from the Agency for Healthcare Research and Quality (AHRQ), “conventional gray-scale ultrasonography is the most common imaging modality used to differentiate benign from malignant adnexal masses. Especially with the advent of high-frequency transvaginal probes, the quality of the images allows description of the gross anatomic features of the lesion.”8 This descriptive ability is limited, however, “by the great variability of macroscopic characteristics of both benign and malignant masses. Furthermore, the technique is operator dependent.”8

To overcome these challenges, some experts have developed ultrasonographic (US) morphologic scoring systems, which assign a value to individual characteristics. Lerner and colleagues devised a 4-point system:

CharacteristicPoints
 0123
Wall structureSmooth or small irregularities (<3 mm) Solid or not applicablePapillarities larger than 3 mm
ShadowingYesNo  
SeptationNone or thin (<3 mm)Thick (≥3 mm)  
EchogenicitySonolucent or low-level echo or echogenic core Mixed or high 

The mean point value for benign masses was 1.8; for tumors of low malignant potential it was 3.9; and for malignant tumors it was 5.6 (P < .0005). Lerner and associates proposed a cutoff of 3. A score of 3 or higher, they felt, would be most predictive of malignancy, with sensitivity of 96.8% and specificity of 77%. Positive and negative predictive values were 29.4% and 99.6%, respectively.9

Almost all published scoring systems are based upon or derived from one reported by Sassone and coworkers.10 The most important and practical feature of all scoring systems is their ability to rule out malignancy.

Morphology and Doppler: A synergistic combination

As the same AHRQ report points out, “all of the diagnostic tests and scoring systems we evaluated exhibited a trade-off between sensitivity and specificity—studies of a given test that reported higher sensitivity had lower specificity, and vice versa.”8 Among evaluation methods, the combination of US morphology scores and Doppler imaging achieved the highest pooled sensitivity and specificity scores in distinguishing benign and malignant adnexal masses in postmenopausal women: 86% and 91%, respectively, according to the AHRQ report.8

Compare these figures with those of:

  • Bimanual pelvic examination (45% and 90%, respectively)
  • Doppler resistance index (72% and 90%)
  • Doppler pulsatility index (80% and 73%)
  • presence of blood vessels (88% and 78%).

The combination of US morphology scores and Doppler was comparable to the pooled sensitivity and specificity of magnetic resonance imaging (91% and 88%, respectively) and superior to computed tomography (90% and 75%, respectively).

Why the need to know?

Discrimination between benign and malignant masses serves a number of purposes, depending on the setting.

For example, if a symptomatic woman is found to have an adnexal mass, it is important to identify the type of mass causing the symptoms to determine the best course of treatment. And because surgery may be one of the treatment options, it is helpful to know whether a mass is likely to be malignant so that the patient can be referred to a specialist or center that has optimal surgical expertise.8

Some asymptomatic masses may be identified during the annual bimanual pelvic examination recommended by ACOG or during pregnancy-related US imaging. In this setting, it is important to ascertain whether the mass is likely to be malignant so that the patient can be referred to a specialist, if necessary. In addition, thorough assessment of the mass can help “avoid unnecessary diagnostic procedures, including surgery, and anxiety in women with asymptomatic, nonmalignant conditions. In some cases, there may be a rationale for removing certain asymptomatic benign lesions, including prevention of malignant transformation; prevention of ovarian torsion”; and prevention of rupture. Surgery may also be appropriate to avert the need for more complicated surgery in the future or to enhance fertility.8Janelle Yates, Senior Editor

 

 

Stay tuned!

Next issue, in Part 3 of this series, we will review the use of imaging in the investigation of ovarian neoplasms, both benign and malignant, with an abundance of US images to accompany our discussion.

We want to hear from you! Tell us what you think.

READ THE WHOLE SERIES

Part 1: A Starting Point (September 2010)

Part 3: Ovarian neoplasms (November 2010)

Part 4: The fallopian tubes (December 2010)

Scanning the ovaries is no simple task. As we mentioned in Part 1 of this four-part series, the practitioner must use the right equipment, take basic preparatory steps, be watchful for clues in the history, and reach a conclusion about what he or she sees. Not only that: The ultrasonographer must be extraordinarily vigilant, paying close attention to multiple characteristics of any mass, from thickness of the wall to the presence of papillations or a blood supply—signs of potential malignancy.

In this article, we detail the traits of various types of non-neoplastic ovarian masses, including:

  • functional cysts—follicles, the corpus luteum, and theca lutein cysts
  • nonfunctional cysts—serous masses and endometriomas
  • cystadenofibromas. Although these masses are usually categorized histo-logically as neoplasms, we include them here due to their almost daily appearance in a busy gynecologic ultrasonographic (US) facility.

In Part 3, we will cover ovarian neoplasms, and in Part 4, our focus will be tubal entities such as ectopic pregnancy and torsion.


FIGURE 1 What is a mass made of? 6 morphologic building blocks

Take an inventory of the mass

Any adnexal mass should be assessed in light of its essential characteristics (Figure 1).

Wall structure. Pay attention to thickness. We use an arbitrary cutoff of 4 mm, giving extra scrutiny to thicknesses exceeding that measurement. In our experience, the thicker the wall, the more likely the mass is to be malignant.

Septation and loculation. A mass is typically unilocular or multilocular. Multilocularity is more common in tumors of low malignant potential and malignant neoplasms.

Papillation. Any internal or external papillae or excrescences should draw your attention. Papillarity in an ovarian mass renders that mass suspicious for malignancy.

Measure (height and width) any papillae that are identified, and document them. Because papillae are associated with ovarian malignancy, further assessment is warranted immediately. The first step is determining whether the papillations contain blood vessels—a task for which color and power Doppler are helpful. We prefer power Doppler because it is more sensitive, detecting blood-flow velocity in the lowest detectable range of 2 cm/s, and because it is not directionally influenced.

Papillae that contain blood vessels with detectable flow are suspicious for malignancy.

Exacoustos and colleagues found that papillae as large as 15 mm in height and 10 mm in width (base) were present in 48% of borderline ovarian tumors but in only 4% of benign and 4% of malignant tumors. However, when the intracystic solid tissue exceeded those dimensions, the lesions were present in 48% of invasive ovarian tumors, 18% of borderline ovarian tumors, and 7% of benign masses.1

Internal echo-structure. A mass can be anechoic, a finding that usually indicates the presence of clear fluid. Mostly solid masses are echogenic. And masses that contain particulate matter, such as blood, cellular matter, or even mucous material, usually have echogenicity of a low level, often described as a “ground-glass” appearance. A mass can also have mixed echo-genicity, a finding usually found in cases involving teratoma or malignancy.

Shadowing. If it is present, it may signify the presence of an extremely dense, solid tissue, such as bone or calcification. The diagnosis of a benign teratoma (i.e., dermoid cyst) should be entertained if shadowing is present in a hyperechoic nodule or mass. Malignant masses very rarely, if ever, display frank shadowing.

Overall appearance. On rare occasions, a bizarre shape or “complex” appearance (as it is termed in most radiology reports) may indicate a malignant mass. More likely it indicates the presence of a teratoma, cystadenoma, or even an atypical corpus luteum. In some reports generated by US laboratories, the term “complex” is applied to all structures other than simple cysts.

Size. The size of a mass can be misleading, as small ovarian lesions with the appropriate sonographic characteristics may be malignant and some larger ones without those characteristics may not be. However, it is understood that the larger an ovarian lesion, the more likely it is a tumor. One important distinction: The amount of fluid in a cystic structure or the amount of old blood in an endometrioma is not the disease process…it is the byproduct of the process. So an 8-cm endometrioma may create fewer pain or fertility issues than a 2- or 3-cm endometrioma. Similarly, the amount of “chocolate” fluid is not automatically indicative of the amount of active endometriotic glands or their sequelae!2

Ascites. If it is present, it should be recorded and investigated further because it may be caused by a malignant intra-abdominal tumor.

 

 

Motion tenderness. If the to-and-fro movement of the vaginal probe elicits any motion tenderness, it, too, should be documented. It may be a sign of pelvic peritonitis. In such cases, an “ominous appearing” adnexal finding may represent an inflammatory, rather than malignant, mass.

When to use Doppler

One of the components of extensive evaluation of the adnexae in general and ovaries in particular is color or power Doppler interrogation—or both.

Tumors contain a relatively large number of pathologic blood vessels that lack the muscular layer found in normal blood vessels and, as a result, demonstrate lower resistance to flow. Diastolic flow is high in these vessels, and resistance and pulsatility indices are low.

We also pay attention when these blood vessels have a tortuous appearance, changes in caliber, anastomoses, and vascular lakes.3 The more tortuous the vessels, with multiple inter-vessel connections and dilatations with changing calibers, the greater the risk of malignancy.4 No less important is the presence of a vessel within a “complex” ovarian mass. A centrally located vessel (also called a “lead vessel”) is suspicious for malignancy.5

A gallery of non-neoplastic ovarian masses

Non-neoplastic cysts are, by far, the most common structures of the ovary. They may be functional, as in the case of the follicles, corpus luteum, and theca lutein cysts, or they may be nonfunctional, as in serous cysts and endometriomas. (As we noted in Part 1, do not call the follicles and corpus luteum “cysts” because this designation suggests pathology.)6


FIGURE 2 Simple cyst

This cyst is anechoic and unilocular with thin walls and no papillae.

Functional cysts

Functional cysts, also known as “simple” cysts, may grow as large as 4 to 5 cm in diameter (Figure 2). They are typically unilocular, anechoic, and thin-walled, with no papillae, and almost never malignant. They usually resolve and require no treatment unless rupture or torsion occurs. Except for the corpus luteum, they have no increased blood flow, and need be viewed only by transvaginal ultrasonography (TVS).

The corpus luteum also can be recognized by TVS. It can exhibit any of a variety of internal structures and echo patterns, due to the multitude of shapes of the blood or clot that can be seen within it (Figure 3).


FIGURE 3 Corpus luteum

A–C. Gray-scale, color Doppler, and power Doppler images, respectively, of a typical corpus luteum. B and C show the enveloping vessels, or “ring of fire.” D. A rather typical gray-scale appearance with a mesh-like, linear internal texture. E. A common feature of the corpus luteum is a linear interphase (arrow) between the clot (c) and the liquified serum (s).

The corpus luteum is typically enveloped by blood vessels, visible on color Doppler as what is called a “ring of fire.” It regresses without intervention. In hyperstimulated ovaries, however, more than one may be present; this poses a real diagnostic challenge when ectopic pregnancy is suspected because it is difficult to differentiate the two entities.

Because the corpus luteum can sometimes resemble some types of ovarian tumors on TVS, imaging during the secretory phase of the cycle in a woman of reproductive age is not ideal. Instead, she should be scanned (or rescanned) between days 5 and 9 of the cycle.


FIGURE 4 Hormonally stimulated ovaries

A, B. The right and left ovaries stimulated by follicle-stimulating hormone preparation (arrow points to hilus). C. An ovary stimulated by clomiphene.

Lutein cysts may reach 5 to 10 cm in diameter. They generally have a thick wall, are multilocular, and typically occur after hormonal induction of ovulation (Figure 4). They also can occur in diabetes, molar pregnancy, and hydrops fetalis. We have seen a unilateral theca lutein cyst in a normal pregnancy (Figure 5). No treatment is necessary unless rupture or torsion occurs.


FIGURE 5 Lutein cysts

A–C. The typical “stained glass” appearance of three lutein cysts of the right ovary in a pregnant patient. D. Color Doppler image of the ovary demonstrating high-velocity flow (peak systolic velocity of 20.4 cm/s).

Serous cysts

These cysts can reach 4 cm in diameter, have smooth walls with no papillae, are unilocular, and occur most often during menopause. No pathological blood flow is visible in their walls. Most gynecologists follow them (Figure 6).1,7


FIGURE 6 Serous cyst

A. Right ovary containing the cyst. B. Normal left ovary. C. Power Doppler interrogation showing no particular flow in the walls of a serous cyst.

Endometriomas

After the simple cyst, the endometrioma is the most prevalent ovarian or adnexal cyst (Figure 7). It usually has a thick wall and is filled with homogeneous fluid with low-level echo-genicity. It can reach 10 cm in size, and many are bilateral. It is sometimes called a “chocolate” cyst because of its dark blood content.


 

 

FIGURE 7 Endometriomas

Endometriomas have low echogenicity. A. Unilateral, unilocular cyst with thin walls. B. Bilateral endometriomas. C. Blood flow in a solid or papillary component of the endometrioma is an occasional finding. It should be investigated further because of the risk that it represents endometrioid cancer.

Endometriomas do not resolve; they usually require surgical excision, although very small ones wholly contained within an ovary are often managed medically or expectantly.

These masses rarely (<1%) give rise to endometrioid carcinoma. Should an endometrioma contain papillae with blood vessels, it is extremely suspicious for endometrioid cancer.


FIGURE 8 Cystic fibromas

A. Sonographic image shows a thin wall and hyperechoic, small mural nodules. B. Macroscopic appearance of an area of internal papillary excrescences. C. Measurement of the small, mural nodules. D. Lack of blood flow in the small papillae, a typical finding on color or power Doppler. E, F. Blood flow in the wall of the cyst and in the mural nodules.

Ovarian fibromas

A fibroma is a slow-growing, benign, solid ovarian tumor. It usually has a cystic component and then is called a cystadenofibroma.

The cystic variety is filled with anechoic fluid and has a thin wall. However, its pathognomonic feature is the small (2–3 mm), extremely hyperechoic mural nodules (papillae) it contains (Figure 8A–C). In the overwhelming majority of cases, no blood vessels are detectable, and the mass is unilocular (Figure 8D–E). It can be recognized in the ovary by the semilunar shape of the tissue surrounding it (crescent sign). The differential diagnosis includes the simple (serous) cyst.

The solid fibroma has a myometrium-like texture, with few or no detectable blood vessels in the stroma. The differential diagnosis includes the Brenner tumor and the Krukenberg tumor.

Federal health agency weighs in on adnexal mass assessment

According to a technology assessment from the Agency for Healthcare Research and Quality (AHRQ), “conventional gray-scale ultrasonography is the most common imaging modality used to differentiate benign from malignant adnexal masses. Especially with the advent of high-frequency transvaginal probes, the quality of the images allows description of the gross anatomic features of the lesion.”8 This descriptive ability is limited, however, “by the great variability of macroscopic characteristics of both benign and malignant masses. Furthermore, the technique is operator dependent.”8

To overcome these challenges, some experts have developed ultrasonographic (US) morphologic scoring systems, which assign a value to individual characteristics. Lerner and colleagues devised a 4-point system:

CharacteristicPoints
 0123
Wall structureSmooth or small irregularities (<3 mm) Solid or not applicablePapillarities larger than 3 mm
ShadowingYesNo  
SeptationNone or thin (<3 mm)Thick (≥3 mm)  
EchogenicitySonolucent or low-level echo or echogenic core Mixed or high 

The mean point value for benign masses was 1.8; for tumors of low malignant potential it was 3.9; and for malignant tumors it was 5.6 (P < .0005). Lerner and associates proposed a cutoff of 3. A score of 3 or higher, they felt, would be most predictive of malignancy, with sensitivity of 96.8% and specificity of 77%. Positive and negative predictive values were 29.4% and 99.6%, respectively.9

Almost all published scoring systems are based upon or derived from one reported by Sassone and coworkers.10 The most important and practical feature of all scoring systems is their ability to rule out malignancy.

Morphology and Doppler: A synergistic combination

As the same AHRQ report points out, “all of the diagnostic tests and scoring systems we evaluated exhibited a trade-off between sensitivity and specificity—studies of a given test that reported higher sensitivity had lower specificity, and vice versa.”8 Among evaluation methods, the combination of US morphology scores and Doppler imaging achieved the highest pooled sensitivity and specificity scores in distinguishing benign and malignant adnexal masses in postmenopausal women: 86% and 91%, respectively, according to the AHRQ report.8

Compare these figures with those of:

  • Bimanual pelvic examination (45% and 90%, respectively)
  • Doppler resistance index (72% and 90%)
  • Doppler pulsatility index (80% and 73%)
  • presence of blood vessels (88% and 78%).

The combination of US morphology scores and Doppler was comparable to the pooled sensitivity and specificity of magnetic resonance imaging (91% and 88%, respectively) and superior to computed tomography (90% and 75%, respectively).

Why the need to know?

Discrimination between benign and malignant masses serves a number of purposes, depending on the setting.

For example, if a symptomatic woman is found to have an adnexal mass, it is important to identify the type of mass causing the symptoms to determine the best course of treatment. And because surgery may be one of the treatment options, it is helpful to know whether a mass is likely to be malignant so that the patient can be referred to a specialist or center that has optimal surgical expertise.8

Some asymptomatic masses may be identified during the annual bimanual pelvic examination recommended by ACOG or during pregnancy-related US imaging. In this setting, it is important to ascertain whether the mass is likely to be malignant so that the patient can be referred to a specialist, if necessary. In addition, thorough assessment of the mass can help “avoid unnecessary diagnostic procedures, including surgery, and anxiety in women with asymptomatic, nonmalignant conditions. In some cases, there may be a rationale for removing certain asymptomatic benign lesions, including prevention of malignant transformation; prevention of ovarian torsion”; and prevention of rupture. Surgery may also be appropriate to avert the need for more complicated surgery in the future or to enhance fertility.8Janelle Yates, Senior Editor

 

 

Stay tuned!

Next issue, in Part 3 of this series, we will review the use of imaging in the investigation of ovarian neoplasms, both benign and malignant, with an abundance of US images to accompany our discussion.

We want to hear from you! Tell us what you think.

References

1. Exacoustos C, Romanini ME, Rinaldo D, et al. Preoperative sonographic features of borderline ovarian tumors. Ultrasound Obstet Gynecol. 2004;25(1):50-59.

2. Rulin MC, Preston AL. Adnexal masses in postmenopausal women. Obstet Gynecol. 1987;70(4):578-581.

3. Timor-Tritsch IE, Goldstein SR. The complexity of a complex mass and the simplicity of a simple cyst. J Ultraound Med. 2005;24(3):255-258.

4. Sladkevicius P, Jokubkiene L, Valentin L. Contribution of morphological assessment of the vessel tree by three-dimensional ultrasound to a correct diagnos is of malignancy in ovarian masses. Ultrasound Obstet Gynecol. 2007;30(6):874-882.

5. Testa AC, Mancari R, Di Legge A, et al. The “lead vessel”: a vascular ultrasound feature of metastasis in the ovaries. Ultrasound Obstet Gynecol. 2008;31:218-221.

6. Goldstein SR. Postmenopausal adnexal cysts: how clinical management has evolved. Am J Obstet Gynecol. 1996;175(6):1496-1501.

7. Levine D, Gosink BB, Wolf S, Feldesman MR, Pretorius D. Simple adnexal cysts: the natural history in postmenopausal women. Radiology. 1992;184(3):653-659.

8. Myers ER, Bastian LA, Havrilesky LJ, et al. Management of adnexal mass. Evidence Report Technol Assess. 2006;Feb;(130):1-145.

9. Lerner JP, Timor-Tritsch IE, Federman A, Abramovich G. Transvaginal ultrasonographic characterization of ovarian masses with an improved, weighted scoring system. Am J Obstet Gynecol. 1994;170(1 Pt 1):81-85.

10. Sassone AM, Timor-Tritsch IE, Artner A, et al. Transvaginal sonographic characterization of ovarian disease: evaluation of a new scoring system to predict ovarian malignancy. Obstet Gynecol. 2001;78:70-76.

References

1. Exacoustos C, Romanini ME, Rinaldo D, et al. Preoperative sonographic features of borderline ovarian tumors. Ultrasound Obstet Gynecol. 2004;25(1):50-59.

2. Rulin MC, Preston AL. Adnexal masses in postmenopausal women. Obstet Gynecol. 1987;70(4):578-581.

3. Timor-Tritsch IE, Goldstein SR. The complexity of a complex mass and the simplicity of a simple cyst. J Ultraound Med. 2005;24(3):255-258.

4. Sladkevicius P, Jokubkiene L, Valentin L. Contribution of morphological assessment of the vessel tree by three-dimensional ultrasound to a correct diagnos is of malignancy in ovarian masses. Ultrasound Obstet Gynecol. 2007;30(6):874-882.

5. Testa AC, Mancari R, Di Legge A, et al. The “lead vessel”: a vascular ultrasound feature of metastasis in the ovaries. Ultrasound Obstet Gynecol. 2008;31:218-221.

6. Goldstein SR. Postmenopausal adnexal cysts: how clinical management has evolved. Am J Obstet Gynecol. 1996;175(6):1496-1501.

7. Levine D, Gosink BB, Wolf S, Feldesman MR, Pretorius D. Simple adnexal cysts: the natural history in postmenopausal women. Radiology. 1992;184(3):653-659.

8. Myers ER, Bastian LA, Havrilesky LJ, et al. Management of adnexal mass. Evidence Report Technol Assess. 2006;Feb;(130):1-145.

9. Lerner JP, Timor-Tritsch IE, Federman A, Abramovich G. Transvaginal ultrasonographic characterization of ovarian masses with an improved, weighted scoring system. Am J Obstet Gynecol. 1994;170(1 Pt 1):81-85.

10. Sassone AM, Timor-Tritsch IE, Artner A, et al. Transvaginal sonographic characterization of ovarian disease: evaluation of a new scoring system to predict ovarian malignancy. Obstet Gynecol. 2001;78:70-76.

Issue
OBG Management - 22(10)
Issue
OBG Management - 22(10)
Page Number
44-56
Page Number
44-56
Publications
Publications
Article Type
Display Headline
Skilled US imaging of the adnexae: The non-neoplastic mass
Display Headline
Skilled US imaging of the adnexae: The non-neoplastic mass
Sections
Article Source

PURLs Copyright

Inside the Article
Article PDF Media

An ObGyn’s guide to aromatase inhibitors as adjuvant therapy for breast CA

Article Type
Changed
Thu, 12/15/2022 - 18:30
Display Headline
An ObGyn’s guide to aromatase inhibitors as adjuvant therapy for breast CA

Think breast cancer survivors are unlikely to show up in your practice? You should think again.

At last official count, 2.5 million women in the United States had a history of breast cancer.1 Most of them are now free of malignancy, but others are still grappling with the disease in some form or fashion.2 All need continuing health care.

Roughly two thirds of women who have breast cancer have disease that is hormone-receptor–positive.2,3 Recently updated guidelines from the American Society of Clinical Oncology (ASCO) recommend that adjuvant therapy for postmenopausal women who have hormone-positive breast cancer include an aromatase inhibitor (AI) (see a summary of these guidelines on page 36). That makes it likely that a good number of breast cancer survivors who visit your practice are taking one of these medications: anastrozole (Arimidex), letrozole (Femara), or exemestane (Aromasin). These drugs are antiestrogens, given to postmenopausal women to reduce the likelihood of disease recurrence and progression.

The antiestrogenic properties of these drugs are what make them lifesavers. But the same qualities can create a range of health issues, from increased risk of osteoporosis and fracture to vasomotor and joint symptoms. And although ObGyns are not the physicians who prescribe these drugs, you may be the provider one of these women consults about their side effects and related issues.

To find out the latest on the management of women who are taking one of these agents, we inserted ourselves into the busy schedule of Andrew M. Kaunitz, MD, who agreed to address some fundamental—and some not so basic—questions about the drugs. In this extended Q&A, Dr. Kaunitz touches on mechanism of action, benefits versus risks, common side effects, compliance with therapy, and the ill effects of early discontinuation.


Aromatase inhibitors are better than tamoxifen at reducing the risk of breast cancer recurrence in postmenopausal women who have hormone-receptor–positive disease. But, they increase the risk of osteoporosis and fracture and often cause arthralgias and other complaints that ObGyn practitioners may be called upon to manage.

OBG Management: What is the overall aim of adjuvant endocrine therapy in the setting of breast cancer?

Dr. Kaunitz: Endocrine therapy—specifically, use of an AI—prevents the stimulation of breast cancer cells by endogenous estrogen. In other words, aromatase inhibitors suppress the growth of cancer cells that have estrogen receptors. These drugs also inhibit aromatase near any breast tumor and reduce estrogen levels in breast tissue.

OBG Management: What is the mechanism of action of adjuvant endocrine therapy?

Dr. Kaunitz: In postmenopausal women, androgens are converted to estrogens via the aromatase enzyme, which is present in adipose tissue and other sites. By blocking this enzyme, AIs reduce endogenous estrogen levels by as much as 95%.4

OBG Management: How does that differ from the mechanism of action of tamoxifen, another drug used in breast cancer patients?

Dr. Kaunitz: Tamoxifen is a selective estrogen receptor modulator (SERM). It blocks estrogen in breast tissue selectively, by competitively binding to estrogen receptors.5 However, tamoxifen has estrogenic effects in the uterus, bone, and liver, as well as other tissues.

The efficacy of AIs in preventing breast cancer recurrence in the first 2 years after breast cancer surgery is higher than that of tamoxifen. And unlike tamoxifen, the AIs do not increase the risk of venous thromboembolism or cause endometrial disease.

OBG Management: What effects do aromatase inhibitors have in premenopausal women?

Dr. Kaunitz: These agents are not recommended for use in premenopausal women because, in that population, the lion’s share of estrogen production takes place in the ovary rather than in adipose tissue and muscle. If you were to administer an AI to a premenopausal woman, the reduced hypothalamic and pituitary estrogen feedback could lead to ovarian stimulation—which could increase ovarian steroid production.

OBG Management: What about women who become amenorrheic as a result of chemotherapy or other cancer treatment? Do most oncologists assume that they are postmenopausal and prescribe an aromatase inhibitor?

Dr. Kaunitz: Clinicians should not assume that chemotherapy-induced amenorrhea signals permanent cessation of ovarian function. It is common for ovarian function to return in this setting. Accordingly, follicle-stimulating hormone (FSH) and estradiol levels should be assessed before an AI is considered as adjuvant therapy. Some investigators have suggested that the use of an AI in women who have chemotherapy-induced amenorrhea may actually increase the likelihood that ovarian function will return.6

 

 

OBG Management: Do all AIs produce the same effects?

Dr. Kaunitz: The AIs used in women with breast cancer are third-generation drugs. These AIs are classified as steroidal (type 1; exemestane) or nonsteroidal (type 2; anastrozole, letrozole). Exemestane, a steroid derived from androstenedione, inhibits the aromatase enzyme irreversibly. The nonsteroidal AIs are reversible.

Although all three AIs have numerous similarities, there are other distinctions between them in pharmacokinetics, mechanism of action, and toxicity—so they are not completely interchangeable.7 However, from our perspective as ObGyns caring for breast cancer survivors, we can assume that all three AIs will have similar effects on skeletal health and produce similar side effects in postmenopausal women.

Key points about AIs in breast Ca

  • The American Society of Clinical Oncology recommends that adjuvant therapy for postmenopausal women who have hormone-positive breast cancer include an aromatase inhibitor (AI).
  • AIs are not recommended for use in premenopausal women.
  • By blocking the aromatase enzyme, AIs reduce endogenous estrogen levels by as much as 95%.4
  • AIs are more effective than tamoxifen at preventing recurrence in the first 2 years after breast cancer surgery. Postmenopausal women taking an AI have a longer disease-free survival and time to recurrence than do women taking tamoxifen. They also have a lower incidence of contralateral breast cancer.
  • The most prominent side effects of AI therapy include arthralgias and hot flushes, while the most serious health impact appears to be a decrease in bone mineral density (BMD). However, endometrial cancer, vaginal bleeding and discharge, cerebrovascular events, venous thromboembolic events, and hot flushes all are less common among women taking an AI than among those taking tamoxifen.
  • The FDA strongly discourages the use of estrogen therapy—systemic or local—in women who are taking an AI. Accordingly, bisphospho-nate therapy is recommended as first-line treatment of low bone mineral density. Vaginal lubricants and moisturizers are the mainstay strategy for symptomatic genital atrophy. And gabapentin, selective serotonin reuptake inhibitors, and serotonin norepinephrine reuptake inhibitors are the mainstay of therapy for vasomotor flushes.
  • Roughly 50% of women who are prescribed adjuvant endocrine therapy with tamoxifen or an AI discontinue the drug early.32 Early discontinuation is associated with an increase in mortality.33

How much do we know about these drugs?

OBG Management: How long does a woman typically take an AI?

Dr. Kaunitz: At present, in women treated for early-stage, hormone-positive breast cancer, the optimal duration of treatment is unknown. Most oncologists prescribe an AI for 5 years, the length of treatment in a prominent trial of the drugs.8

OBG Management: Is that duration likely to increase as more data come in?

Dr. Kaunitz: The optimal duration of adjuvant AI therapy will be determined by the findings of long-term clinical trials. The National Surgical Adjuvant Breast and Bowel Project B-42 trial may provide new insights into optimal duration of AI treatment after initial tamoxifen therapy.9

OBG Management: How thoroughly have AIs been studied in regard to their use in breast cancer survivors and women who have early-stage disease? How would you characterize the quantity and quality of data that we have so far?

Dr. Kaunitz: AIs have been extensively studied. The most important clinical trials of AIs in this setting, including the Anastrozole, Tamoxifen Alone or in Combination (ATAC) trial (over 6,000 participants, median follow-up of 100 months) and the Breast International Group (BIG) trial (almost 5,000 participants, median follow-up of 76 months) have been detailed in the recent ASCO report.10 These two large landmark trials, in particular, formed the basis for ASCO’s recommendations to routinely incorporate AIs into the therapy of postmenopausal women who have hormone-receptor–positive breast cancer.

OBG Management: In treating breast cancer, what other applications are AIs used for?

Dr. Kaunitz: AIs appear to be slightly more effective than tamoxifen in treating postmenopausal women who have metastatic breast cancer.11

They are approved as first-line therapy for breast cancer in:

  • postmenopausal women who have hormone-receptor–positive disease
  • postmenopausal women who have locally advanced disease when the hormone receptor is unknown
  • postmenopausal women who have metastatic disease.

In addition, they are approved as second-line treatment of advanced breast cancer in postmenopausal women who have disease progression following tamoxifen therapy.12

How effective is AI therapy?

OBG Management: What do we know about the efficacy of these drugs?

Dr. Kaunitz: Most of the studies that have explored efficacy compare an AI with tamoxifen rather than with placebo. In the ATAC trial, after a median follow-up of 33 months, women who were taking anastrozole for early-stage breast cancer had longer disease-free survival and time to recurrence and a lower incidence of contralateral breast cancer than did women taking tamoxifen.8

 

 

After 4 years of follow-up in the ATAC trial, women taking anastrozole continued to have more favorable disease-free survival (86.9% vs 84.5% for anastrozole and tamoxifen, respectively; hazard ratio [HR], 0.86; 95% confidence interval [CI], 0.76–0.99; P =.03).13 They also had a more favorable time to recurrence than did women taking tamoxifen (HR, 0.83; 95% CI, 0.71–0.96; P =.015). And women taking anastrozole had a lower incidence of contralateral breast cancer, as well, although this different did not achieve statistical significance (HR, 0.62; 95% CI, 0.38–1.02; P =.062).13

In the BIG study, women taking letrozole had a 5-year disease-free survival estimate of 84.0%, compared with 81.4% for women taking tamoxifen.14 In addition, women taking letrozole were significantly less likely than those taking tamoxifen to experience an event that ended a period of disease-free survival (HR, 0.81; 95% CI, 0.70–0.93; P =.003), especially the event of distant recurrence (HR, 0.73; 95% CI, 0.60–0.88; P =.001).14

And a phase-3 study of exemestane versus tamoxifen in women who had metastatic breast cancer found that the AI produced a superior response rate (46% vs 31% for exemestane and tamoxifen, respectively; odds ratio [OR], 1.85; 95% CI, 1.21–2.82; P =.005). In addition, median progression-free survival was greater with exemestane (9.9 months; 95% CI, 8.7–11.8 months) than with tamoxifen (5.8 months; 95% CI, 5.3–8.1 months). However, there was no difference between arms in progression-free survival or overall survival.

ASCO guidelines emphasize the importance of aromatase inhibitors

Postmenopausal women who have hormone-receptor–positive breast cancer should consider taking an aromatase inhibitor (AI) to lengthen disease-free survival and lower the risk of recurrence. That’s one of the recommendations in updated guidelines issued earlier this year by the American Society of Clinical Oncology (ASCO). The guidelines suggest a duration of AI therapy of 5 years. In the event that a woman discontinues AI therapy before 5 years are up, she should consider using tamoxifen to bring the total duration of treatment to 5 years.

Other recommendations in the guidelines include:

  • Women who have taken tamoxifen for 5 years stand to benefit from switching to an AI for as long as 5 additional years.
  • When advising a woman about adjuvant therapy with an AI, clinicians should consider the potential adverse effects, which include osteoporosis, fracture, and arthralgias.
  • The third-generation AIs on the market today have not been found to have clinically important differences between them. A woman who cannot tolerate a particular AI should consider switching to a different AI.
  • Switching from an AI to tamoxifen (or vice versa) may be an appropriate option for patients who cannot tolerate a drug’s adverse effects. In the event of a switch to tamoxifen, the clinician should counsel the patient about its adverse effects, which include venous thromboembolism and endometrial polyps, hyperplasia, and cancer.

The full guidelines can be accessed at http://jco.ascopubs.org/content/early/2010/07/12/JCO.2009.26.3756.full.pdf.

Andrew M. Kaunitz, MD

How well tolerated are AIs?

OBG Management: What adverse effects are associated with AIs?

Dr. Kaunitz: Although AIs, overall, are safe medications, their use is associated with a number of adverse events. The most prominent side effects include arthralgias and hot flushes, while the most serious health impact appears to be a decrease in bone mineral density (BMD).

However, the drugs are generally perceived as being easier to tolerate than tamoxifen. That’s because endometrial cancer, vaginal bleeding and discharge, cerebrovascular events, venous thromboembolic events, and hot flushes all are less common among women taking an AI than among those taking tamoxifen.8,13

For overweight women, who face an elevated baseline risk of thromboembolism, the availability of AIs represents a major advantage over tamoxifen. Similarly, AIs offer advantages over tamoxifen for women who have an intact uterus. In addition, postmenopausal women who are taking a selective serotonin reuptake inhibitor (SSRI) such as paroxetine should take an AI rather than tamoxifen, because the concomitant use of SSRIs attenuates the efficacy of tamoxifen.15

What can be done about the most prominent risks?

OBG Management: Let’s focus on what’s probably the best-known adverse effect of AIs—the heightened risk of osteoporosis and fracture. How significant is this effect?

Dr. Kaunitz: Because use of an AI is associated with a profound reduction in endogenous estrogen levels, it also decreases BMD and can lead to osteoporotic fractures. All major phase-3 trials of adjuvant use of AIs in women who have early breast cancer found an increased risk of fracture, with no significant differences between AIs.16

Fortunately, bisphosphonate therapy (oral or intravenous) has been found to reduce bone loss associated with AI therapy.17,18

Assessing baseline BMD is important as women initiate AI therapy. Although no consensus exists regarding follow-up BMD assessment in the setting of AI use, an interval of 2 years is prudent, with the follow-up study preferably performed at the same imaging center and by the same technician as the first. If baseline osteoporosis is observed at the lumbar spine or hip, bisphosphonate therapy is appropriate. If a woman taking an AI has low bone mass (osteopenia) but not osteoporosis, bisphosphonate therapy should be considered if any of the following risk factors are present:

 

 

  • advanced age
  • history of fracture
  • glucocorticoid therapy
  • parental history of hip fracture
  • low body weight
  • current smoking status
  • excess alcohol consumption
  • rheumatoid arthritis
  • known risk factors for secondary osteoporosis.19

In breast cancer survivors initiating or continuing AI therapy, it is also appropriate to check a serum vitamin D level and ensure that intake of this nutrient is adequate.

Bisphosphonates may offer oncologic benefits, as well; preliminary evidence suggests that the drugs may prevent recurrence of the cancer and prolong survival.20

OBG Management: What can an ObGyn offer to a woman who complains of significant AI-related arthralgia?

Dr. Kaunitz: Bone and joint symptoms, including aches, pain, and stiffness that is bilateral and not associated with other evidence of rheumatologic disorders, are among the most common side effects of AI therapy. On the plus side of the equation, these symptoms are more likely to be mild to moderate than severe. On the negative side, no specific treatment has been found to be effective in relieving these symptoms, which usually resolve within 2 months or so after discontinuing AI therapy.10

OBG Management: Do AIs have a negative impact on cardiovascular health?

Dr. Kaunitz: Unlike tamoxifen, AIs do not increase the risk of thromboembolic disease. Although the use of an AI may modestly increase the risk of ischemic cardiovascular disease (and lipid changes), compared with tamoxifen, AIs do not appear to increase cardiovascular risk compared with placebo.21,22

OBG Management: Do the antiestrogenic effects of AIs have a significant impact on vaginal health and sexual desire?

Dr. Kaunitz: A review of published reports did not find that the use of AIs has a predictable impact on vaginal dryness or sexual desire.10 However, symptomatic genital atrophy is common in postmenopausal breast cancer survivors, whether or not they use adjuvant therapy.

Although the FDA considers the use of any estrogen (systemic or vaginal) following a diagnosis of breast cancer to be contraindicated, some breast cancer survivors who have symptomatic genital atrophy express an interest in the use of vaginal estrogen. Use of 25-μg estradiol tablets (Vagifem) is associated with a short-term increase in serum estradiol levels.23 This finding has reinforced caution among medical oncologists about the safety of vaginal estrogen in breast cancer survivors. (The 25-μg tablets are no longer marketed.) The lowest dosage of vaginal estrogen available for the treatment of genital atrophy is found in 10-μg estradiol tablets (Vagifem) and the estradiol (2-mg) 3-month vaginal ring (Estring). Nonetheless, in the absence of data, oncologists will likely continue to be concerned that even the lowest dosage of vaginal estrogen could attenuate the favorable impact of AIs on breast cancer. Accordingly, use of vaginal lubricants and moisturizers are the mainstay strategy for symptomatic genital atrophy.

OBG Management: What about the ubiquitous hot flush? Vasomotor symptoms may be more common in women who take tamoxifen, but women on AIs are also bothered by flushes. What are the alternatives to estrogen therapy?

Dr. Kaunitz: Both nonprescription and prescription alternatives are available. Nonprescription options include soy extract and red clover isoflavones, black cohosh, and Chinese herbs. However, none of these over-the-counter approaches has been found to be more effective than placebo in the treatment of menopausal hot flushes.24-26

As for prescription nonhormonal options, ObGyns should recognize that all such treatments are off-label and that none attain the efficacy of hormone therapy in the treatment of vasomotor symptoms. The best-studied and most effective medications include gabapentin, SSRIs (especially paroxetine), and serotonin-norepinephrine reuptake inhibitors (venlafaxine and desvenlafaxine).24,27

OBG Management: Is there any evidence that AIs impair cognitive function in postmenopausal women?

Dr. Kaunitz: Because estrogen is important for cognition, one might anticipate that the profound reduction in background estrogen associated with AI use would impair cognition. Fortunately, the evidence to date is reassuring. Substudies of the BIG trial and the Tamoxifen and Exemestane Adjuvant Multinational Trial indicate that, compared with tamoxifen (which is associated with declines in cognitive function in postmenopausal women), letrozole and exemestane do not diminish cognitive function.28,29

OBG Management: Overall, what is the typical impact of an AI on a woman’s quality of life?

Dr. Kaunitz: Most women do very well on an AI, finding it easier to tolerate than tamoxifen, as we have discussed. However, a significant minority of women is seriously bothered by the adverse effects, with arthralgias usually leading the pack of complaints.30,31

OBG Management: Do some women discontinue adjuvant endocrine therapy because of adverse effects?

Dr. Kaunitz: Regrettably, the answer is “Yes.” A recent study from Kaiser Permanente of northern California found that roughly 50% of women who are prescribed adjuvant endocrine therapy with tamoxifen or an AI discontinue the drug early.32

 

 

OBG Management: What can an ObGyn do to encourage compliance with and completion of AI therapy?

Dr. Kaunitz: First, it is critical that patients understand that AIs are lifesaving drugs. As a recent paper points out, early discontinuation or noncompliance with AI therapy is associated with higher mortality.33

Clinicians should also help breast cancer patients understand what common side effects to anticipate with these medications.

Finally, clinicians who understand the financial toll a breast cancer diagnosis and treatment can take are better positioned to help women overcome challenges that may interfere with long-term compliance with AI therapy.

OBG Management: Do you expect the use of AIs in breast cancer survivors to become more commonplace?

Dr. Kaunitz: Given how common breast cancer is, and given the new ASCO guidelines and the extensive literature upon which they are based, ObGyns will be seeing more women using AIs. Although we are not the physicians who prescribe AIs, we need to remain up to date on their benefits and side effects. This important class of drugs is positioned to improve outcomes for postmenopausal women with breast cancer.

We want to hear from you! Tell us what you think.

References

1. Horner MJ, Ries LAG, Krapcho M, et al. eds. SEER Cancer Statistics Review, 1975–2006. Bethesda, Md: National Cancer Institute; 2009.http://seer.cancer.gov/csr/1975_2006. Accessed August 18, 2010.

2. American Cancer Society. Breast cancer facts and figures, 2009–2010. Atlanta, Ga: American Cancer Society; 2010. http://www.cancer.org/Research/CancerFactsFigures/BreastCancerFactsFigures/breast-cancer-facts—figures-2009-2010. Accessed August 18, 2010.

3. Li CI, Daling JR, Malone KE. Incidence of invasive breast cancer by hormone receptor status from 1992 to 1998. J Clin Oncol. 2003;21(1):28-34.

4. Miller WR. Aromatase inhibitors: mechanism of action and role in the treatment of breast cancer. Semin Oncol. 2003;30(4 suppl 14):3-11.

5. Peng J, Sengupta S, Jordan VC. Potential of selective estrogen receptor modulators as treatments and preventives of breast cancer. Anticancer Agents Med Chem. 2009;9(5):481-499.

6. Smith IE, Dowsett M, Yap YS, et al. Adjuvant aromatase inhibitors for early breast cancer after chemotherapy-induced amenorrhea: caution and suggested guidelines. J Clin Oncol. 2006;24(16):2444-2447.

7. Nabholtz JM, Mouret-Reynier MA, Durando X, et al. Comparative review of anastrozole, letrozole and exemestane in the management of early breast cancer. Expert Opin Pharmacother. 2009;10(9):1435-1447.

8. Baum M, Budzar AU, Cuzick J, et al. ATAC Trialists’ Group. Anastrozole alone or in combination with tamoxifen versus tamoxifen alone for adjuvant treatment of postmenopausal women with early breast cancer: first results of the ATAC randomized trial. Lancet. 2002;359(9324):2131-2139.

9. Letrozole in treating postmenopausal women who have received hormone therapy for hormone receptor-positive breast cancer. National Cancer Institute Web site.http://www.cancer.gov/clinicaltrials/NSABP-B-42. Published August 16, 2010. Accessed August 18, 2010.

10. Burstein HJ, Prestrud AA, Seidenfeld J, et al. American Society of Clinical Oncology clinical practice guideline: update on adjuvant endocrine therapy for women with hormone-receptor–positive breast cancer. J Clin Oncol. 2010;28(23):3784-3796.

11. Mauri D, Pavlidis N, Polyzos NP, Ioanidis JP. Survival with aromatase inhibitors and inactivators versus standard hormonal therapy in advanced breast cancer: meta-analysis. J Natl Cancer Inst. 2006;98(18):1285-1291.

12. Arimidex [package insert]. AstraZeneca; 2009.

13. Baum M, Buzdar A, Cuzick J, et al. Anastrozole alone or in combination with tamoxifen versus tamoxifen alone for adjuvant treatment of postmenopausal women with early-stage breast cancer: results of the ATAC (Arimidex, Taxoxifen Alone or in Combination) trial efficacy and safety update analyses. Cancer. 2003;98(9):1802-1810.

14. Thurlimann B, Keshaviah A, Coates AS, et al. Breast International Group (BIG) 1-98 Collaborative Group. A comparison of letrozole and tamoxifen in postmenopausal women with early breast cancer. N Engl J Med. 2005;353(26):2747-2757.

15. Kelly CM, Juurlink DM, Gomes T, et al. Selective serotonin reuptake inhibitors and breast cancer mortality in women receiving tamoxifen: a population-based cohort study. 2010;340:c693.-doi: 10.1136/bmj.c693.

16. Geisler J, Lonning PE. Impact of aromatase inhibitors on bone health in breast cancer patients. J Steroid Biochem Mol Biol. 2010;118(4–5):294-299.

17. Van Poznak C, Hannon RA, Mackey JR, et al. Prevention of aromatase inhibitor-induced bone loss using risedronate: the SABRE trial. J Clin Oncol. 2010;28(6):967-975.

18. Brufsky AM, Bosserman LD, Caradonna RR, et al. Zoledronic acid effectively prevents aromatase-inhibitor associated bone loss in postmenopausal women with early breast cancer receiving adjuvant letrozole. Z-FAST study 36-month follow-up results. Clin Breast Cancer. 2009;9(2):77-85.

19. Kanis JA, Borgstrom F, De Laet C, et al. Assessment of fracture risk. Osteoporosis Int. 2005;16(6):581-589.

20. Reeder JG, Brufsky AM. The role of bisphosphonates in the adjuvant setting for breast cancer. Oncology. 2010;24(6):462-467,475.

21. Nabholtz JM. Long-term safety of aromatase inhibitors in the treatment of breast cancer. Ther Clin Risk Manag. 2008;4(1):189-204.

22. Cuppone F, Bria E, Verma S, et al. Do adjuvant aromatase inhibitors increase the cardiovascular risk in postmenopausal women with early breast cancer? Meta-analysis of randomized trials. Cancer. 2008;112(2):260-267.

23. Kendall A, Dowsett M, Folkerd E, Smith I. Caution: Vaginal estradiol appears to be contraindicated in postmenopausal women on adjuvant aromatase inhibitors. Ann Oncol. 2006;17(4):584-587.

24. Nelson HD. Menopause. Lancet. 2008;371(9614):760-770.

25. Geller SE, Shulman LP, van Breemen RB, et al. Safety and efficacy of black cohosh and red clover for the management of vasomotor symptoms: a randomized controlled trial. Menopause. 2009;16(6):1156-1166.

26. Kaunitz AM. Effective herbal treatment of vasomotor symptoms—are we any closer? Menopause. 2009;16(3):428-429.

27. Archer DF, Dupont CM, Constantine GD, Pickar JH, Olivier S. Study 319 Investigators. Desvenlafaxine for the treatment of vasomotor symptoms associated with menopause: a double-blind, randomized, placebo-controlled trial of efficacy and safety. Am J Obstet Gynecol. 2009;200(3):238.e1-e10.

28. Phillips KA, Ribi K, Sun Z, et al. Cognitive function in postmenopausal women receiving adjuvant letrozole or tamoxifen for breast cancer in the BIG 1-98 randomized trial [published online ahead of print April 10, 2010]. Breast. doi:10.1016/j.breast.2010.03.025.

29. Schilder CM, Seynaeve C, Beex LV, et al. Effects of tamoxifen and exemestane on cognitive functioning of postmenopausal patients with breast cancer: results from the neuropsychological side study of the Tamoxifen and Exemestane Adjuvant Multinational Trial. J Clin Oncol. 2010;28(8):1294-1300.

30. Din OS, Dodwell D, Wakefield RJ, Coleman RE. Aromatase inhibitor-induced arthralgia in early breast cancer: what do we know and how can we find out more? Breast Cancer Res Treat. 2010;120(3):525-538.

31. Burstein HJ. Aromatase inhibitor-associated arthralgia syndrome. Breast. 2007;16(3):223-234.

32. Hershman DL, Kushi LH, Shao T, et al. Early discontinuation and nonadherence to adjuvant hormonal therapy in a cohort of 8,769 early-stage breast cancer patients [published online ahead of print June 28, 2010]. J Clin Oncol. doi: 10.1200/JCO.2009.25.9655.

33. Hershman DL, Shao T, Kushi LH, et al. Early discontinuation and nonadherence to adjuvant hormonal therapy are associated with increased mortality in women with breast cancer [published online ahead of print August 28, 2010]. Breast Cancer Res Treat. doi: 10.1007/ s10549-010-1132-4.

Article PDF
Author and Disclosure Information

Hear Dr. Kaunitz describe bone-protective strategies for patients on AI therapy

Andrew M. Kaunitz, MD
Dr. Kaunitz is Professor and associate Chairman of the Department of Obstetrics and Gynecology at the University of Florida College of Medicine– Jacksonville in Jacksonville, Fla. He serves on the OBG Management Board of Editors.

Janelle Yates
Senior Editor, OBG Management

Dr. Kaunitz reports no financial relationships relevant to this article.

Issue
OBG Management - 22(10)
Publications
Topics
Page Number
30-40
Sections
Author and Disclosure Information

Hear Dr. Kaunitz describe bone-protective strategies for patients on AI therapy

Andrew M. Kaunitz, MD
Dr. Kaunitz is Professor and associate Chairman of the Department of Obstetrics and Gynecology at the University of Florida College of Medicine– Jacksonville in Jacksonville, Fla. He serves on the OBG Management Board of Editors.

Janelle Yates
Senior Editor, OBG Management

Dr. Kaunitz reports no financial relationships relevant to this article.

Author and Disclosure Information

Hear Dr. Kaunitz describe bone-protective strategies for patients on AI therapy

Andrew M. Kaunitz, MD
Dr. Kaunitz is Professor and associate Chairman of the Department of Obstetrics and Gynecology at the University of Florida College of Medicine– Jacksonville in Jacksonville, Fla. He serves on the OBG Management Board of Editors.

Janelle Yates
Senior Editor, OBG Management

Dr. Kaunitz reports no financial relationships relevant to this article.

Article PDF
Article PDF

Think breast cancer survivors are unlikely to show up in your practice? You should think again.

At last official count, 2.5 million women in the United States had a history of breast cancer.1 Most of them are now free of malignancy, but others are still grappling with the disease in some form or fashion.2 All need continuing health care.

Roughly two thirds of women who have breast cancer have disease that is hormone-receptor–positive.2,3 Recently updated guidelines from the American Society of Clinical Oncology (ASCO) recommend that adjuvant therapy for postmenopausal women who have hormone-positive breast cancer include an aromatase inhibitor (AI) (see a summary of these guidelines on page 36). That makes it likely that a good number of breast cancer survivors who visit your practice are taking one of these medications: anastrozole (Arimidex), letrozole (Femara), or exemestane (Aromasin). These drugs are antiestrogens, given to postmenopausal women to reduce the likelihood of disease recurrence and progression.

The antiestrogenic properties of these drugs are what make them lifesavers. But the same qualities can create a range of health issues, from increased risk of osteoporosis and fracture to vasomotor and joint symptoms. And although ObGyns are not the physicians who prescribe these drugs, you may be the provider one of these women consults about their side effects and related issues.

To find out the latest on the management of women who are taking one of these agents, we inserted ourselves into the busy schedule of Andrew M. Kaunitz, MD, who agreed to address some fundamental—and some not so basic—questions about the drugs. In this extended Q&A, Dr. Kaunitz touches on mechanism of action, benefits versus risks, common side effects, compliance with therapy, and the ill effects of early discontinuation.


Aromatase inhibitors are better than tamoxifen at reducing the risk of breast cancer recurrence in postmenopausal women who have hormone-receptor–positive disease. But, they increase the risk of osteoporosis and fracture and often cause arthralgias and other complaints that ObGyn practitioners may be called upon to manage.

OBG Management: What is the overall aim of adjuvant endocrine therapy in the setting of breast cancer?

Dr. Kaunitz: Endocrine therapy—specifically, use of an AI—prevents the stimulation of breast cancer cells by endogenous estrogen. In other words, aromatase inhibitors suppress the growth of cancer cells that have estrogen receptors. These drugs also inhibit aromatase near any breast tumor and reduce estrogen levels in breast tissue.

OBG Management: What is the mechanism of action of adjuvant endocrine therapy?

Dr. Kaunitz: In postmenopausal women, androgens are converted to estrogens via the aromatase enzyme, which is present in adipose tissue and other sites. By blocking this enzyme, AIs reduce endogenous estrogen levels by as much as 95%.4

OBG Management: How does that differ from the mechanism of action of tamoxifen, another drug used in breast cancer patients?

Dr. Kaunitz: Tamoxifen is a selective estrogen receptor modulator (SERM). It blocks estrogen in breast tissue selectively, by competitively binding to estrogen receptors.5 However, tamoxifen has estrogenic effects in the uterus, bone, and liver, as well as other tissues.

The efficacy of AIs in preventing breast cancer recurrence in the first 2 years after breast cancer surgery is higher than that of tamoxifen. And unlike tamoxifen, the AIs do not increase the risk of venous thromboembolism or cause endometrial disease.

OBG Management: What effects do aromatase inhibitors have in premenopausal women?

Dr. Kaunitz: These agents are not recommended for use in premenopausal women because, in that population, the lion’s share of estrogen production takes place in the ovary rather than in adipose tissue and muscle. If you were to administer an AI to a premenopausal woman, the reduced hypothalamic and pituitary estrogen feedback could lead to ovarian stimulation—which could increase ovarian steroid production.

OBG Management: What about women who become amenorrheic as a result of chemotherapy or other cancer treatment? Do most oncologists assume that they are postmenopausal and prescribe an aromatase inhibitor?

Dr. Kaunitz: Clinicians should not assume that chemotherapy-induced amenorrhea signals permanent cessation of ovarian function. It is common for ovarian function to return in this setting. Accordingly, follicle-stimulating hormone (FSH) and estradiol levels should be assessed before an AI is considered as adjuvant therapy. Some investigators have suggested that the use of an AI in women who have chemotherapy-induced amenorrhea may actually increase the likelihood that ovarian function will return.6

 

 

OBG Management: Do all AIs produce the same effects?

Dr. Kaunitz: The AIs used in women with breast cancer are third-generation drugs. These AIs are classified as steroidal (type 1; exemestane) or nonsteroidal (type 2; anastrozole, letrozole). Exemestane, a steroid derived from androstenedione, inhibits the aromatase enzyme irreversibly. The nonsteroidal AIs are reversible.

Although all three AIs have numerous similarities, there are other distinctions between them in pharmacokinetics, mechanism of action, and toxicity—so they are not completely interchangeable.7 However, from our perspective as ObGyns caring for breast cancer survivors, we can assume that all three AIs will have similar effects on skeletal health and produce similar side effects in postmenopausal women.

Key points about AIs in breast Ca

  • The American Society of Clinical Oncology recommends that adjuvant therapy for postmenopausal women who have hormone-positive breast cancer include an aromatase inhibitor (AI).
  • AIs are not recommended for use in premenopausal women.
  • By blocking the aromatase enzyme, AIs reduce endogenous estrogen levels by as much as 95%.4
  • AIs are more effective than tamoxifen at preventing recurrence in the first 2 years after breast cancer surgery. Postmenopausal women taking an AI have a longer disease-free survival and time to recurrence than do women taking tamoxifen. They also have a lower incidence of contralateral breast cancer.
  • The most prominent side effects of AI therapy include arthralgias and hot flushes, while the most serious health impact appears to be a decrease in bone mineral density (BMD). However, endometrial cancer, vaginal bleeding and discharge, cerebrovascular events, venous thromboembolic events, and hot flushes all are less common among women taking an AI than among those taking tamoxifen.
  • The FDA strongly discourages the use of estrogen therapy—systemic or local—in women who are taking an AI. Accordingly, bisphospho-nate therapy is recommended as first-line treatment of low bone mineral density. Vaginal lubricants and moisturizers are the mainstay strategy for symptomatic genital atrophy. And gabapentin, selective serotonin reuptake inhibitors, and serotonin norepinephrine reuptake inhibitors are the mainstay of therapy for vasomotor flushes.
  • Roughly 50% of women who are prescribed adjuvant endocrine therapy with tamoxifen or an AI discontinue the drug early.32 Early discontinuation is associated with an increase in mortality.33

How much do we know about these drugs?

OBG Management: How long does a woman typically take an AI?

Dr. Kaunitz: At present, in women treated for early-stage, hormone-positive breast cancer, the optimal duration of treatment is unknown. Most oncologists prescribe an AI for 5 years, the length of treatment in a prominent trial of the drugs.8

OBG Management: Is that duration likely to increase as more data come in?

Dr. Kaunitz: The optimal duration of adjuvant AI therapy will be determined by the findings of long-term clinical trials. The National Surgical Adjuvant Breast and Bowel Project B-42 trial may provide new insights into optimal duration of AI treatment after initial tamoxifen therapy.9

OBG Management: How thoroughly have AIs been studied in regard to their use in breast cancer survivors and women who have early-stage disease? How would you characterize the quantity and quality of data that we have so far?

Dr. Kaunitz: AIs have been extensively studied. The most important clinical trials of AIs in this setting, including the Anastrozole, Tamoxifen Alone or in Combination (ATAC) trial (over 6,000 participants, median follow-up of 100 months) and the Breast International Group (BIG) trial (almost 5,000 participants, median follow-up of 76 months) have been detailed in the recent ASCO report.10 These two large landmark trials, in particular, formed the basis for ASCO’s recommendations to routinely incorporate AIs into the therapy of postmenopausal women who have hormone-receptor–positive breast cancer.

OBG Management: In treating breast cancer, what other applications are AIs used for?

Dr. Kaunitz: AIs appear to be slightly more effective than tamoxifen in treating postmenopausal women who have metastatic breast cancer.11

They are approved as first-line therapy for breast cancer in:

  • postmenopausal women who have hormone-receptor–positive disease
  • postmenopausal women who have locally advanced disease when the hormone receptor is unknown
  • postmenopausal women who have metastatic disease.

In addition, they are approved as second-line treatment of advanced breast cancer in postmenopausal women who have disease progression following tamoxifen therapy.12

How effective is AI therapy?

OBG Management: What do we know about the efficacy of these drugs?

Dr. Kaunitz: Most of the studies that have explored efficacy compare an AI with tamoxifen rather than with placebo. In the ATAC trial, after a median follow-up of 33 months, women who were taking anastrozole for early-stage breast cancer had longer disease-free survival and time to recurrence and a lower incidence of contralateral breast cancer than did women taking tamoxifen.8

 

 

After 4 years of follow-up in the ATAC trial, women taking anastrozole continued to have more favorable disease-free survival (86.9% vs 84.5% for anastrozole and tamoxifen, respectively; hazard ratio [HR], 0.86; 95% confidence interval [CI], 0.76–0.99; P =.03).13 They also had a more favorable time to recurrence than did women taking tamoxifen (HR, 0.83; 95% CI, 0.71–0.96; P =.015). And women taking anastrozole had a lower incidence of contralateral breast cancer, as well, although this different did not achieve statistical significance (HR, 0.62; 95% CI, 0.38–1.02; P =.062).13

In the BIG study, women taking letrozole had a 5-year disease-free survival estimate of 84.0%, compared with 81.4% for women taking tamoxifen.14 In addition, women taking letrozole were significantly less likely than those taking tamoxifen to experience an event that ended a period of disease-free survival (HR, 0.81; 95% CI, 0.70–0.93; P =.003), especially the event of distant recurrence (HR, 0.73; 95% CI, 0.60–0.88; P =.001).14

And a phase-3 study of exemestane versus tamoxifen in women who had metastatic breast cancer found that the AI produced a superior response rate (46% vs 31% for exemestane and tamoxifen, respectively; odds ratio [OR], 1.85; 95% CI, 1.21–2.82; P =.005). In addition, median progression-free survival was greater with exemestane (9.9 months; 95% CI, 8.7–11.8 months) than with tamoxifen (5.8 months; 95% CI, 5.3–8.1 months). However, there was no difference between arms in progression-free survival or overall survival.

ASCO guidelines emphasize the importance of aromatase inhibitors

Postmenopausal women who have hormone-receptor–positive breast cancer should consider taking an aromatase inhibitor (AI) to lengthen disease-free survival and lower the risk of recurrence. That’s one of the recommendations in updated guidelines issued earlier this year by the American Society of Clinical Oncology (ASCO). The guidelines suggest a duration of AI therapy of 5 years. In the event that a woman discontinues AI therapy before 5 years are up, she should consider using tamoxifen to bring the total duration of treatment to 5 years.

Other recommendations in the guidelines include:

  • Women who have taken tamoxifen for 5 years stand to benefit from switching to an AI for as long as 5 additional years.
  • When advising a woman about adjuvant therapy with an AI, clinicians should consider the potential adverse effects, which include osteoporosis, fracture, and arthralgias.
  • The third-generation AIs on the market today have not been found to have clinically important differences between them. A woman who cannot tolerate a particular AI should consider switching to a different AI.
  • Switching from an AI to tamoxifen (or vice versa) may be an appropriate option for patients who cannot tolerate a drug’s adverse effects. In the event of a switch to tamoxifen, the clinician should counsel the patient about its adverse effects, which include venous thromboembolism and endometrial polyps, hyperplasia, and cancer.

The full guidelines can be accessed at http://jco.ascopubs.org/content/early/2010/07/12/JCO.2009.26.3756.full.pdf.

Andrew M. Kaunitz, MD

How well tolerated are AIs?

OBG Management: What adverse effects are associated with AIs?

Dr. Kaunitz: Although AIs, overall, are safe medications, their use is associated with a number of adverse events. The most prominent side effects include arthralgias and hot flushes, while the most serious health impact appears to be a decrease in bone mineral density (BMD).

However, the drugs are generally perceived as being easier to tolerate than tamoxifen. That’s because endometrial cancer, vaginal bleeding and discharge, cerebrovascular events, venous thromboembolic events, and hot flushes all are less common among women taking an AI than among those taking tamoxifen.8,13

For overweight women, who face an elevated baseline risk of thromboembolism, the availability of AIs represents a major advantage over tamoxifen. Similarly, AIs offer advantages over tamoxifen for women who have an intact uterus. In addition, postmenopausal women who are taking a selective serotonin reuptake inhibitor (SSRI) such as paroxetine should take an AI rather than tamoxifen, because the concomitant use of SSRIs attenuates the efficacy of tamoxifen.15

What can be done about the most prominent risks?

OBG Management: Let’s focus on what’s probably the best-known adverse effect of AIs—the heightened risk of osteoporosis and fracture. How significant is this effect?

Dr. Kaunitz: Because use of an AI is associated with a profound reduction in endogenous estrogen levels, it also decreases BMD and can lead to osteoporotic fractures. All major phase-3 trials of adjuvant use of AIs in women who have early breast cancer found an increased risk of fracture, with no significant differences between AIs.16

Fortunately, bisphosphonate therapy (oral or intravenous) has been found to reduce bone loss associated with AI therapy.17,18

Assessing baseline BMD is important as women initiate AI therapy. Although no consensus exists regarding follow-up BMD assessment in the setting of AI use, an interval of 2 years is prudent, with the follow-up study preferably performed at the same imaging center and by the same technician as the first. If baseline osteoporosis is observed at the lumbar spine or hip, bisphosphonate therapy is appropriate. If a woman taking an AI has low bone mass (osteopenia) but not osteoporosis, bisphosphonate therapy should be considered if any of the following risk factors are present:

 

 

  • advanced age
  • history of fracture
  • glucocorticoid therapy
  • parental history of hip fracture
  • low body weight
  • current smoking status
  • excess alcohol consumption
  • rheumatoid arthritis
  • known risk factors for secondary osteoporosis.19

In breast cancer survivors initiating or continuing AI therapy, it is also appropriate to check a serum vitamin D level and ensure that intake of this nutrient is adequate.

Bisphosphonates may offer oncologic benefits, as well; preliminary evidence suggests that the drugs may prevent recurrence of the cancer and prolong survival.20

OBG Management: What can an ObGyn offer to a woman who complains of significant AI-related arthralgia?

Dr. Kaunitz: Bone and joint symptoms, including aches, pain, and stiffness that is bilateral and not associated with other evidence of rheumatologic disorders, are among the most common side effects of AI therapy. On the plus side of the equation, these symptoms are more likely to be mild to moderate than severe. On the negative side, no specific treatment has been found to be effective in relieving these symptoms, which usually resolve within 2 months or so after discontinuing AI therapy.10

OBG Management: Do AIs have a negative impact on cardiovascular health?

Dr. Kaunitz: Unlike tamoxifen, AIs do not increase the risk of thromboembolic disease. Although the use of an AI may modestly increase the risk of ischemic cardiovascular disease (and lipid changes), compared with tamoxifen, AIs do not appear to increase cardiovascular risk compared with placebo.21,22

OBG Management: Do the antiestrogenic effects of AIs have a significant impact on vaginal health and sexual desire?

Dr. Kaunitz: A review of published reports did not find that the use of AIs has a predictable impact on vaginal dryness or sexual desire.10 However, symptomatic genital atrophy is common in postmenopausal breast cancer survivors, whether or not they use adjuvant therapy.

Although the FDA considers the use of any estrogen (systemic or vaginal) following a diagnosis of breast cancer to be contraindicated, some breast cancer survivors who have symptomatic genital atrophy express an interest in the use of vaginal estrogen. Use of 25-μg estradiol tablets (Vagifem) is associated with a short-term increase in serum estradiol levels.23 This finding has reinforced caution among medical oncologists about the safety of vaginal estrogen in breast cancer survivors. (The 25-μg tablets are no longer marketed.) The lowest dosage of vaginal estrogen available for the treatment of genital atrophy is found in 10-μg estradiol tablets (Vagifem) and the estradiol (2-mg) 3-month vaginal ring (Estring). Nonetheless, in the absence of data, oncologists will likely continue to be concerned that even the lowest dosage of vaginal estrogen could attenuate the favorable impact of AIs on breast cancer. Accordingly, use of vaginal lubricants and moisturizers are the mainstay strategy for symptomatic genital atrophy.

OBG Management: What about the ubiquitous hot flush? Vasomotor symptoms may be more common in women who take tamoxifen, but women on AIs are also bothered by flushes. What are the alternatives to estrogen therapy?

Dr. Kaunitz: Both nonprescription and prescription alternatives are available. Nonprescription options include soy extract and red clover isoflavones, black cohosh, and Chinese herbs. However, none of these over-the-counter approaches has been found to be more effective than placebo in the treatment of menopausal hot flushes.24-26

As for prescription nonhormonal options, ObGyns should recognize that all such treatments are off-label and that none attain the efficacy of hormone therapy in the treatment of vasomotor symptoms. The best-studied and most effective medications include gabapentin, SSRIs (especially paroxetine), and serotonin-norepinephrine reuptake inhibitors (venlafaxine and desvenlafaxine).24,27

OBG Management: Is there any evidence that AIs impair cognitive function in postmenopausal women?

Dr. Kaunitz: Because estrogen is important for cognition, one might anticipate that the profound reduction in background estrogen associated with AI use would impair cognition. Fortunately, the evidence to date is reassuring. Substudies of the BIG trial and the Tamoxifen and Exemestane Adjuvant Multinational Trial indicate that, compared with tamoxifen (which is associated with declines in cognitive function in postmenopausal women), letrozole and exemestane do not diminish cognitive function.28,29

OBG Management: Overall, what is the typical impact of an AI on a woman’s quality of life?

Dr. Kaunitz: Most women do very well on an AI, finding it easier to tolerate than tamoxifen, as we have discussed. However, a significant minority of women is seriously bothered by the adverse effects, with arthralgias usually leading the pack of complaints.30,31

OBG Management: Do some women discontinue adjuvant endocrine therapy because of adverse effects?

Dr. Kaunitz: Regrettably, the answer is “Yes.” A recent study from Kaiser Permanente of northern California found that roughly 50% of women who are prescribed adjuvant endocrine therapy with tamoxifen or an AI discontinue the drug early.32

 

 

OBG Management: What can an ObGyn do to encourage compliance with and completion of AI therapy?

Dr. Kaunitz: First, it is critical that patients understand that AIs are lifesaving drugs. As a recent paper points out, early discontinuation or noncompliance with AI therapy is associated with higher mortality.33

Clinicians should also help breast cancer patients understand what common side effects to anticipate with these medications.

Finally, clinicians who understand the financial toll a breast cancer diagnosis and treatment can take are better positioned to help women overcome challenges that may interfere with long-term compliance with AI therapy.

OBG Management: Do you expect the use of AIs in breast cancer survivors to become more commonplace?

Dr. Kaunitz: Given how common breast cancer is, and given the new ASCO guidelines and the extensive literature upon which they are based, ObGyns will be seeing more women using AIs. Although we are not the physicians who prescribe AIs, we need to remain up to date on their benefits and side effects. This important class of drugs is positioned to improve outcomes for postmenopausal women with breast cancer.

We want to hear from you! Tell us what you think.

Think breast cancer survivors are unlikely to show up in your practice? You should think again.

At last official count, 2.5 million women in the United States had a history of breast cancer.1 Most of them are now free of malignancy, but others are still grappling with the disease in some form or fashion.2 All need continuing health care.

Roughly two thirds of women who have breast cancer have disease that is hormone-receptor–positive.2,3 Recently updated guidelines from the American Society of Clinical Oncology (ASCO) recommend that adjuvant therapy for postmenopausal women who have hormone-positive breast cancer include an aromatase inhibitor (AI) (see a summary of these guidelines on page 36). That makes it likely that a good number of breast cancer survivors who visit your practice are taking one of these medications: anastrozole (Arimidex), letrozole (Femara), or exemestane (Aromasin). These drugs are antiestrogens, given to postmenopausal women to reduce the likelihood of disease recurrence and progression.

The antiestrogenic properties of these drugs are what make them lifesavers. But the same qualities can create a range of health issues, from increased risk of osteoporosis and fracture to vasomotor and joint symptoms. And although ObGyns are not the physicians who prescribe these drugs, you may be the provider one of these women consults about their side effects and related issues.

To find out the latest on the management of women who are taking one of these agents, we inserted ourselves into the busy schedule of Andrew M. Kaunitz, MD, who agreed to address some fundamental—and some not so basic—questions about the drugs. In this extended Q&A, Dr. Kaunitz touches on mechanism of action, benefits versus risks, common side effects, compliance with therapy, and the ill effects of early discontinuation.


Aromatase inhibitors are better than tamoxifen at reducing the risk of breast cancer recurrence in postmenopausal women who have hormone-receptor–positive disease. But, they increase the risk of osteoporosis and fracture and often cause arthralgias and other complaints that ObGyn practitioners may be called upon to manage.

OBG Management: What is the overall aim of adjuvant endocrine therapy in the setting of breast cancer?

Dr. Kaunitz: Endocrine therapy—specifically, use of an AI—prevents the stimulation of breast cancer cells by endogenous estrogen. In other words, aromatase inhibitors suppress the growth of cancer cells that have estrogen receptors. These drugs also inhibit aromatase near any breast tumor and reduce estrogen levels in breast tissue.

OBG Management: What is the mechanism of action of adjuvant endocrine therapy?

Dr. Kaunitz: In postmenopausal women, androgens are converted to estrogens via the aromatase enzyme, which is present in adipose tissue and other sites. By blocking this enzyme, AIs reduce endogenous estrogen levels by as much as 95%.4

OBG Management: How does that differ from the mechanism of action of tamoxifen, another drug used in breast cancer patients?

Dr. Kaunitz: Tamoxifen is a selective estrogen receptor modulator (SERM). It blocks estrogen in breast tissue selectively, by competitively binding to estrogen receptors.5 However, tamoxifen has estrogenic effects in the uterus, bone, and liver, as well as other tissues.

The efficacy of AIs in preventing breast cancer recurrence in the first 2 years after breast cancer surgery is higher than that of tamoxifen. And unlike tamoxifen, the AIs do not increase the risk of venous thromboembolism or cause endometrial disease.

OBG Management: What effects do aromatase inhibitors have in premenopausal women?

Dr. Kaunitz: These agents are not recommended for use in premenopausal women because, in that population, the lion’s share of estrogen production takes place in the ovary rather than in adipose tissue and muscle. If you were to administer an AI to a premenopausal woman, the reduced hypothalamic and pituitary estrogen feedback could lead to ovarian stimulation—which could increase ovarian steroid production.

OBG Management: What about women who become amenorrheic as a result of chemotherapy or other cancer treatment? Do most oncologists assume that they are postmenopausal and prescribe an aromatase inhibitor?

Dr. Kaunitz: Clinicians should not assume that chemotherapy-induced amenorrhea signals permanent cessation of ovarian function. It is common for ovarian function to return in this setting. Accordingly, follicle-stimulating hormone (FSH) and estradiol levels should be assessed before an AI is considered as adjuvant therapy. Some investigators have suggested that the use of an AI in women who have chemotherapy-induced amenorrhea may actually increase the likelihood that ovarian function will return.6

 

 

OBG Management: Do all AIs produce the same effects?

Dr. Kaunitz: The AIs used in women with breast cancer are third-generation drugs. These AIs are classified as steroidal (type 1; exemestane) or nonsteroidal (type 2; anastrozole, letrozole). Exemestane, a steroid derived from androstenedione, inhibits the aromatase enzyme irreversibly. The nonsteroidal AIs are reversible.

Although all three AIs have numerous similarities, there are other distinctions between them in pharmacokinetics, mechanism of action, and toxicity—so they are not completely interchangeable.7 However, from our perspective as ObGyns caring for breast cancer survivors, we can assume that all three AIs will have similar effects on skeletal health and produce similar side effects in postmenopausal women.

Key points about AIs in breast Ca

  • The American Society of Clinical Oncology recommends that adjuvant therapy for postmenopausal women who have hormone-positive breast cancer include an aromatase inhibitor (AI).
  • AIs are not recommended for use in premenopausal women.
  • By blocking the aromatase enzyme, AIs reduce endogenous estrogen levels by as much as 95%.4
  • AIs are more effective than tamoxifen at preventing recurrence in the first 2 years after breast cancer surgery. Postmenopausal women taking an AI have a longer disease-free survival and time to recurrence than do women taking tamoxifen. They also have a lower incidence of contralateral breast cancer.
  • The most prominent side effects of AI therapy include arthralgias and hot flushes, while the most serious health impact appears to be a decrease in bone mineral density (BMD). However, endometrial cancer, vaginal bleeding and discharge, cerebrovascular events, venous thromboembolic events, and hot flushes all are less common among women taking an AI than among those taking tamoxifen.
  • The FDA strongly discourages the use of estrogen therapy—systemic or local—in women who are taking an AI. Accordingly, bisphospho-nate therapy is recommended as first-line treatment of low bone mineral density. Vaginal lubricants and moisturizers are the mainstay strategy for symptomatic genital atrophy. And gabapentin, selective serotonin reuptake inhibitors, and serotonin norepinephrine reuptake inhibitors are the mainstay of therapy for vasomotor flushes.
  • Roughly 50% of women who are prescribed adjuvant endocrine therapy with tamoxifen or an AI discontinue the drug early.32 Early discontinuation is associated with an increase in mortality.33

How much do we know about these drugs?

OBG Management: How long does a woman typically take an AI?

Dr. Kaunitz: At present, in women treated for early-stage, hormone-positive breast cancer, the optimal duration of treatment is unknown. Most oncologists prescribe an AI for 5 years, the length of treatment in a prominent trial of the drugs.8

OBG Management: Is that duration likely to increase as more data come in?

Dr. Kaunitz: The optimal duration of adjuvant AI therapy will be determined by the findings of long-term clinical trials. The National Surgical Adjuvant Breast and Bowel Project B-42 trial may provide new insights into optimal duration of AI treatment after initial tamoxifen therapy.9

OBG Management: How thoroughly have AIs been studied in regard to their use in breast cancer survivors and women who have early-stage disease? How would you characterize the quantity and quality of data that we have so far?

Dr. Kaunitz: AIs have been extensively studied. The most important clinical trials of AIs in this setting, including the Anastrozole, Tamoxifen Alone or in Combination (ATAC) trial (over 6,000 participants, median follow-up of 100 months) and the Breast International Group (BIG) trial (almost 5,000 participants, median follow-up of 76 months) have been detailed in the recent ASCO report.10 These two large landmark trials, in particular, formed the basis for ASCO’s recommendations to routinely incorporate AIs into the therapy of postmenopausal women who have hormone-receptor–positive breast cancer.

OBG Management: In treating breast cancer, what other applications are AIs used for?

Dr. Kaunitz: AIs appear to be slightly more effective than tamoxifen in treating postmenopausal women who have metastatic breast cancer.11

They are approved as first-line therapy for breast cancer in:

  • postmenopausal women who have hormone-receptor–positive disease
  • postmenopausal women who have locally advanced disease when the hormone receptor is unknown
  • postmenopausal women who have metastatic disease.

In addition, they are approved as second-line treatment of advanced breast cancer in postmenopausal women who have disease progression following tamoxifen therapy.12

How effective is AI therapy?

OBG Management: What do we know about the efficacy of these drugs?

Dr. Kaunitz: Most of the studies that have explored efficacy compare an AI with tamoxifen rather than with placebo. In the ATAC trial, after a median follow-up of 33 months, women who were taking anastrozole for early-stage breast cancer had longer disease-free survival and time to recurrence and a lower incidence of contralateral breast cancer than did women taking tamoxifen.8

 

 

After 4 years of follow-up in the ATAC trial, women taking anastrozole continued to have more favorable disease-free survival (86.9% vs 84.5% for anastrozole and tamoxifen, respectively; hazard ratio [HR], 0.86; 95% confidence interval [CI], 0.76–0.99; P =.03).13 They also had a more favorable time to recurrence than did women taking tamoxifen (HR, 0.83; 95% CI, 0.71–0.96; P =.015). And women taking anastrozole had a lower incidence of contralateral breast cancer, as well, although this different did not achieve statistical significance (HR, 0.62; 95% CI, 0.38–1.02; P =.062).13

In the BIG study, women taking letrozole had a 5-year disease-free survival estimate of 84.0%, compared with 81.4% for women taking tamoxifen.14 In addition, women taking letrozole were significantly less likely than those taking tamoxifen to experience an event that ended a period of disease-free survival (HR, 0.81; 95% CI, 0.70–0.93; P =.003), especially the event of distant recurrence (HR, 0.73; 95% CI, 0.60–0.88; P =.001).14

And a phase-3 study of exemestane versus tamoxifen in women who had metastatic breast cancer found that the AI produced a superior response rate (46% vs 31% for exemestane and tamoxifen, respectively; odds ratio [OR], 1.85; 95% CI, 1.21–2.82; P =.005). In addition, median progression-free survival was greater with exemestane (9.9 months; 95% CI, 8.7–11.8 months) than with tamoxifen (5.8 months; 95% CI, 5.3–8.1 months). However, there was no difference between arms in progression-free survival or overall survival.

ASCO guidelines emphasize the importance of aromatase inhibitors

Postmenopausal women who have hormone-receptor–positive breast cancer should consider taking an aromatase inhibitor (AI) to lengthen disease-free survival and lower the risk of recurrence. That’s one of the recommendations in updated guidelines issued earlier this year by the American Society of Clinical Oncology (ASCO). The guidelines suggest a duration of AI therapy of 5 years. In the event that a woman discontinues AI therapy before 5 years are up, she should consider using tamoxifen to bring the total duration of treatment to 5 years.

Other recommendations in the guidelines include:

  • Women who have taken tamoxifen for 5 years stand to benefit from switching to an AI for as long as 5 additional years.
  • When advising a woman about adjuvant therapy with an AI, clinicians should consider the potential adverse effects, which include osteoporosis, fracture, and arthralgias.
  • The third-generation AIs on the market today have not been found to have clinically important differences between them. A woman who cannot tolerate a particular AI should consider switching to a different AI.
  • Switching from an AI to tamoxifen (or vice versa) may be an appropriate option for patients who cannot tolerate a drug’s adverse effects. In the event of a switch to tamoxifen, the clinician should counsel the patient about its adverse effects, which include venous thromboembolism and endometrial polyps, hyperplasia, and cancer.

The full guidelines can be accessed at http://jco.ascopubs.org/content/early/2010/07/12/JCO.2009.26.3756.full.pdf.

Andrew M. Kaunitz, MD

How well tolerated are AIs?

OBG Management: What adverse effects are associated with AIs?

Dr. Kaunitz: Although AIs, overall, are safe medications, their use is associated with a number of adverse events. The most prominent side effects include arthralgias and hot flushes, while the most serious health impact appears to be a decrease in bone mineral density (BMD).

However, the drugs are generally perceived as being easier to tolerate than tamoxifen. That’s because endometrial cancer, vaginal bleeding and discharge, cerebrovascular events, venous thromboembolic events, and hot flushes all are less common among women taking an AI than among those taking tamoxifen.8,13

For overweight women, who face an elevated baseline risk of thromboembolism, the availability of AIs represents a major advantage over tamoxifen. Similarly, AIs offer advantages over tamoxifen for women who have an intact uterus. In addition, postmenopausal women who are taking a selective serotonin reuptake inhibitor (SSRI) such as paroxetine should take an AI rather than tamoxifen, because the concomitant use of SSRIs attenuates the efficacy of tamoxifen.15

What can be done about the most prominent risks?

OBG Management: Let’s focus on what’s probably the best-known adverse effect of AIs—the heightened risk of osteoporosis and fracture. How significant is this effect?

Dr. Kaunitz: Because use of an AI is associated with a profound reduction in endogenous estrogen levels, it also decreases BMD and can lead to osteoporotic fractures. All major phase-3 trials of adjuvant use of AIs in women who have early breast cancer found an increased risk of fracture, with no significant differences between AIs.16

Fortunately, bisphosphonate therapy (oral or intravenous) has been found to reduce bone loss associated with AI therapy.17,18

Assessing baseline BMD is important as women initiate AI therapy. Although no consensus exists regarding follow-up BMD assessment in the setting of AI use, an interval of 2 years is prudent, with the follow-up study preferably performed at the same imaging center and by the same technician as the first. If baseline osteoporosis is observed at the lumbar spine or hip, bisphosphonate therapy is appropriate. If a woman taking an AI has low bone mass (osteopenia) but not osteoporosis, bisphosphonate therapy should be considered if any of the following risk factors are present:

 

 

  • advanced age
  • history of fracture
  • glucocorticoid therapy
  • parental history of hip fracture
  • low body weight
  • current smoking status
  • excess alcohol consumption
  • rheumatoid arthritis
  • known risk factors for secondary osteoporosis.19

In breast cancer survivors initiating or continuing AI therapy, it is also appropriate to check a serum vitamin D level and ensure that intake of this nutrient is adequate.

Bisphosphonates may offer oncologic benefits, as well; preliminary evidence suggests that the drugs may prevent recurrence of the cancer and prolong survival.20

OBG Management: What can an ObGyn offer to a woman who complains of significant AI-related arthralgia?

Dr. Kaunitz: Bone and joint symptoms, including aches, pain, and stiffness that is bilateral and not associated with other evidence of rheumatologic disorders, are among the most common side effects of AI therapy. On the plus side of the equation, these symptoms are more likely to be mild to moderate than severe. On the negative side, no specific treatment has been found to be effective in relieving these symptoms, which usually resolve within 2 months or so after discontinuing AI therapy.10

OBG Management: Do AIs have a negative impact on cardiovascular health?

Dr. Kaunitz: Unlike tamoxifen, AIs do not increase the risk of thromboembolic disease. Although the use of an AI may modestly increase the risk of ischemic cardiovascular disease (and lipid changes), compared with tamoxifen, AIs do not appear to increase cardiovascular risk compared with placebo.21,22

OBG Management: Do the antiestrogenic effects of AIs have a significant impact on vaginal health and sexual desire?

Dr. Kaunitz: A review of published reports did not find that the use of AIs has a predictable impact on vaginal dryness or sexual desire.10 However, symptomatic genital atrophy is common in postmenopausal breast cancer survivors, whether or not they use adjuvant therapy.

Although the FDA considers the use of any estrogen (systemic or vaginal) following a diagnosis of breast cancer to be contraindicated, some breast cancer survivors who have symptomatic genital atrophy express an interest in the use of vaginal estrogen. Use of 25-μg estradiol tablets (Vagifem) is associated with a short-term increase in serum estradiol levels.23 This finding has reinforced caution among medical oncologists about the safety of vaginal estrogen in breast cancer survivors. (The 25-μg tablets are no longer marketed.) The lowest dosage of vaginal estrogen available for the treatment of genital atrophy is found in 10-μg estradiol tablets (Vagifem) and the estradiol (2-mg) 3-month vaginal ring (Estring). Nonetheless, in the absence of data, oncologists will likely continue to be concerned that even the lowest dosage of vaginal estrogen could attenuate the favorable impact of AIs on breast cancer. Accordingly, use of vaginal lubricants and moisturizers are the mainstay strategy for symptomatic genital atrophy.

OBG Management: What about the ubiquitous hot flush? Vasomotor symptoms may be more common in women who take tamoxifen, but women on AIs are also bothered by flushes. What are the alternatives to estrogen therapy?

Dr. Kaunitz: Both nonprescription and prescription alternatives are available. Nonprescription options include soy extract and red clover isoflavones, black cohosh, and Chinese herbs. However, none of these over-the-counter approaches has been found to be more effective than placebo in the treatment of menopausal hot flushes.24-26

As for prescription nonhormonal options, ObGyns should recognize that all such treatments are off-label and that none attain the efficacy of hormone therapy in the treatment of vasomotor symptoms. The best-studied and most effective medications include gabapentin, SSRIs (especially paroxetine), and serotonin-norepinephrine reuptake inhibitors (venlafaxine and desvenlafaxine).24,27

OBG Management: Is there any evidence that AIs impair cognitive function in postmenopausal women?

Dr. Kaunitz: Because estrogen is important for cognition, one might anticipate that the profound reduction in background estrogen associated with AI use would impair cognition. Fortunately, the evidence to date is reassuring. Substudies of the BIG trial and the Tamoxifen and Exemestane Adjuvant Multinational Trial indicate that, compared with tamoxifen (which is associated with declines in cognitive function in postmenopausal women), letrozole and exemestane do not diminish cognitive function.28,29

OBG Management: Overall, what is the typical impact of an AI on a woman’s quality of life?

Dr. Kaunitz: Most women do very well on an AI, finding it easier to tolerate than tamoxifen, as we have discussed. However, a significant minority of women is seriously bothered by the adverse effects, with arthralgias usually leading the pack of complaints.30,31

OBG Management: Do some women discontinue adjuvant endocrine therapy because of adverse effects?

Dr. Kaunitz: Regrettably, the answer is “Yes.” A recent study from Kaiser Permanente of northern California found that roughly 50% of women who are prescribed adjuvant endocrine therapy with tamoxifen or an AI discontinue the drug early.32

 

 

OBG Management: What can an ObGyn do to encourage compliance with and completion of AI therapy?

Dr. Kaunitz: First, it is critical that patients understand that AIs are lifesaving drugs. As a recent paper points out, early discontinuation or noncompliance with AI therapy is associated with higher mortality.33

Clinicians should also help breast cancer patients understand what common side effects to anticipate with these medications.

Finally, clinicians who understand the financial toll a breast cancer diagnosis and treatment can take are better positioned to help women overcome challenges that may interfere with long-term compliance with AI therapy.

OBG Management: Do you expect the use of AIs in breast cancer survivors to become more commonplace?

Dr. Kaunitz: Given how common breast cancer is, and given the new ASCO guidelines and the extensive literature upon which they are based, ObGyns will be seeing more women using AIs. Although we are not the physicians who prescribe AIs, we need to remain up to date on their benefits and side effects. This important class of drugs is positioned to improve outcomes for postmenopausal women with breast cancer.

We want to hear from you! Tell us what you think.

References

1. Horner MJ, Ries LAG, Krapcho M, et al. eds. SEER Cancer Statistics Review, 1975–2006. Bethesda, Md: National Cancer Institute; 2009.http://seer.cancer.gov/csr/1975_2006. Accessed August 18, 2010.

2. American Cancer Society. Breast cancer facts and figures, 2009–2010. Atlanta, Ga: American Cancer Society; 2010. http://www.cancer.org/Research/CancerFactsFigures/BreastCancerFactsFigures/breast-cancer-facts—figures-2009-2010. Accessed August 18, 2010.

3. Li CI, Daling JR, Malone KE. Incidence of invasive breast cancer by hormone receptor status from 1992 to 1998. J Clin Oncol. 2003;21(1):28-34.

4. Miller WR. Aromatase inhibitors: mechanism of action and role in the treatment of breast cancer. Semin Oncol. 2003;30(4 suppl 14):3-11.

5. Peng J, Sengupta S, Jordan VC. Potential of selective estrogen receptor modulators as treatments and preventives of breast cancer. Anticancer Agents Med Chem. 2009;9(5):481-499.

6. Smith IE, Dowsett M, Yap YS, et al. Adjuvant aromatase inhibitors for early breast cancer after chemotherapy-induced amenorrhea: caution and suggested guidelines. J Clin Oncol. 2006;24(16):2444-2447.

7. Nabholtz JM, Mouret-Reynier MA, Durando X, et al. Comparative review of anastrozole, letrozole and exemestane in the management of early breast cancer. Expert Opin Pharmacother. 2009;10(9):1435-1447.

8. Baum M, Budzar AU, Cuzick J, et al. ATAC Trialists’ Group. Anastrozole alone or in combination with tamoxifen versus tamoxifen alone for adjuvant treatment of postmenopausal women with early breast cancer: first results of the ATAC randomized trial. Lancet. 2002;359(9324):2131-2139.

9. Letrozole in treating postmenopausal women who have received hormone therapy for hormone receptor-positive breast cancer. National Cancer Institute Web site.http://www.cancer.gov/clinicaltrials/NSABP-B-42. Published August 16, 2010. Accessed August 18, 2010.

10. Burstein HJ, Prestrud AA, Seidenfeld J, et al. American Society of Clinical Oncology clinical practice guideline: update on adjuvant endocrine therapy for women with hormone-receptor–positive breast cancer. J Clin Oncol. 2010;28(23):3784-3796.

11. Mauri D, Pavlidis N, Polyzos NP, Ioanidis JP. Survival with aromatase inhibitors and inactivators versus standard hormonal therapy in advanced breast cancer: meta-analysis. J Natl Cancer Inst. 2006;98(18):1285-1291.

12. Arimidex [package insert]. AstraZeneca; 2009.

13. Baum M, Buzdar A, Cuzick J, et al. Anastrozole alone or in combination with tamoxifen versus tamoxifen alone for adjuvant treatment of postmenopausal women with early-stage breast cancer: results of the ATAC (Arimidex, Taxoxifen Alone or in Combination) trial efficacy and safety update analyses. Cancer. 2003;98(9):1802-1810.

14. Thurlimann B, Keshaviah A, Coates AS, et al. Breast International Group (BIG) 1-98 Collaborative Group. A comparison of letrozole and tamoxifen in postmenopausal women with early breast cancer. N Engl J Med. 2005;353(26):2747-2757.

15. Kelly CM, Juurlink DM, Gomes T, et al. Selective serotonin reuptake inhibitors and breast cancer mortality in women receiving tamoxifen: a population-based cohort study. 2010;340:c693.-doi: 10.1136/bmj.c693.

16. Geisler J, Lonning PE. Impact of aromatase inhibitors on bone health in breast cancer patients. J Steroid Biochem Mol Biol. 2010;118(4–5):294-299.

17. Van Poznak C, Hannon RA, Mackey JR, et al. Prevention of aromatase inhibitor-induced bone loss using risedronate: the SABRE trial. J Clin Oncol. 2010;28(6):967-975.

18. Brufsky AM, Bosserman LD, Caradonna RR, et al. Zoledronic acid effectively prevents aromatase-inhibitor associated bone loss in postmenopausal women with early breast cancer receiving adjuvant letrozole. Z-FAST study 36-month follow-up results. Clin Breast Cancer. 2009;9(2):77-85.

19. Kanis JA, Borgstrom F, De Laet C, et al. Assessment of fracture risk. Osteoporosis Int. 2005;16(6):581-589.

20. Reeder JG, Brufsky AM. The role of bisphosphonates in the adjuvant setting for breast cancer. Oncology. 2010;24(6):462-467,475.

21. Nabholtz JM. Long-term safety of aromatase inhibitors in the treatment of breast cancer. Ther Clin Risk Manag. 2008;4(1):189-204.

22. Cuppone F, Bria E, Verma S, et al. Do adjuvant aromatase inhibitors increase the cardiovascular risk in postmenopausal women with early breast cancer? Meta-analysis of randomized trials. Cancer. 2008;112(2):260-267.

23. Kendall A, Dowsett M, Folkerd E, Smith I. Caution: Vaginal estradiol appears to be contraindicated in postmenopausal women on adjuvant aromatase inhibitors. Ann Oncol. 2006;17(4):584-587.

24. Nelson HD. Menopause. Lancet. 2008;371(9614):760-770.

25. Geller SE, Shulman LP, van Breemen RB, et al. Safety and efficacy of black cohosh and red clover for the management of vasomotor symptoms: a randomized controlled trial. Menopause. 2009;16(6):1156-1166.

26. Kaunitz AM. Effective herbal treatment of vasomotor symptoms—are we any closer? Menopause. 2009;16(3):428-429.

27. Archer DF, Dupont CM, Constantine GD, Pickar JH, Olivier S. Study 319 Investigators. Desvenlafaxine for the treatment of vasomotor symptoms associated with menopause: a double-blind, randomized, placebo-controlled trial of efficacy and safety. Am J Obstet Gynecol. 2009;200(3):238.e1-e10.

28. Phillips KA, Ribi K, Sun Z, et al. Cognitive function in postmenopausal women receiving adjuvant letrozole or tamoxifen for breast cancer in the BIG 1-98 randomized trial [published online ahead of print April 10, 2010]. Breast. doi:10.1016/j.breast.2010.03.025.

29. Schilder CM, Seynaeve C, Beex LV, et al. Effects of tamoxifen and exemestane on cognitive functioning of postmenopausal patients with breast cancer: results from the neuropsychological side study of the Tamoxifen and Exemestane Adjuvant Multinational Trial. J Clin Oncol. 2010;28(8):1294-1300.

30. Din OS, Dodwell D, Wakefield RJ, Coleman RE. Aromatase inhibitor-induced arthralgia in early breast cancer: what do we know and how can we find out more? Breast Cancer Res Treat. 2010;120(3):525-538.

31. Burstein HJ. Aromatase inhibitor-associated arthralgia syndrome. Breast. 2007;16(3):223-234.

32. Hershman DL, Kushi LH, Shao T, et al. Early discontinuation and nonadherence to adjuvant hormonal therapy in a cohort of 8,769 early-stage breast cancer patients [published online ahead of print June 28, 2010]. J Clin Oncol. doi: 10.1200/JCO.2009.25.9655.

33. Hershman DL, Shao T, Kushi LH, et al. Early discontinuation and nonadherence to adjuvant hormonal therapy are associated with increased mortality in women with breast cancer [published online ahead of print August 28, 2010]. Breast Cancer Res Treat. doi: 10.1007/ s10549-010-1132-4.

References

1. Horner MJ, Ries LAG, Krapcho M, et al. eds. SEER Cancer Statistics Review, 1975–2006. Bethesda, Md: National Cancer Institute; 2009.http://seer.cancer.gov/csr/1975_2006. Accessed August 18, 2010.

2. American Cancer Society. Breast cancer facts and figures, 2009–2010. Atlanta, Ga: American Cancer Society; 2010. http://www.cancer.org/Research/CancerFactsFigures/BreastCancerFactsFigures/breast-cancer-facts—figures-2009-2010. Accessed August 18, 2010.

3. Li CI, Daling JR, Malone KE. Incidence of invasive breast cancer by hormone receptor status from 1992 to 1998. J Clin Oncol. 2003;21(1):28-34.

4. Miller WR. Aromatase inhibitors: mechanism of action and role in the treatment of breast cancer. Semin Oncol. 2003;30(4 suppl 14):3-11.

5. Peng J, Sengupta S, Jordan VC. Potential of selective estrogen receptor modulators as treatments and preventives of breast cancer. Anticancer Agents Med Chem. 2009;9(5):481-499.

6. Smith IE, Dowsett M, Yap YS, et al. Adjuvant aromatase inhibitors for early breast cancer after chemotherapy-induced amenorrhea: caution and suggested guidelines. J Clin Oncol. 2006;24(16):2444-2447.

7. Nabholtz JM, Mouret-Reynier MA, Durando X, et al. Comparative review of anastrozole, letrozole and exemestane in the management of early breast cancer. Expert Opin Pharmacother. 2009;10(9):1435-1447.

8. Baum M, Budzar AU, Cuzick J, et al. ATAC Trialists’ Group. Anastrozole alone or in combination with tamoxifen versus tamoxifen alone for adjuvant treatment of postmenopausal women with early breast cancer: first results of the ATAC randomized trial. Lancet. 2002;359(9324):2131-2139.

9. Letrozole in treating postmenopausal women who have received hormone therapy for hormone receptor-positive breast cancer. National Cancer Institute Web site.http://www.cancer.gov/clinicaltrials/NSABP-B-42. Published August 16, 2010. Accessed August 18, 2010.

10. Burstein HJ, Prestrud AA, Seidenfeld J, et al. American Society of Clinical Oncology clinical practice guideline: update on adjuvant endocrine therapy for women with hormone-receptor–positive breast cancer. J Clin Oncol. 2010;28(23):3784-3796.

11. Mauri D, Pavlidis N, Polyzos NP, Ioanidis JP. Survival with aromatase inhibitors and inactivators versus standard hormonal therapy in advanced breast cancer: meta-analysis. J Natl Cancer Inst. 2006;98(18):1285-1291.

12. Arimidex [package insert]. AstraZeneca; 2009.

13. Baum M, Buzdar A, Cuzick J, et al. Anastrozole alone or in combination with tamoxifen versus tamoxifen alone for adjuvant treatment of postmenopausal women with early-stage breast cancer: results of the ATAC (Arimidex, Taxoxifen Alone or in Combination) trial efficacy and safety update analyses. Cancer. 2003;98(9):1802-1810.

14. Thurlimann B, Keshaviah A, Coates AS, et al. Breast International Group (BIG) 1-98 Collaborative Group. A comparison of letrozole and tamoxifen in postmenopausal women with early breast cancer. N Engl J Med. 2005;353(26):2747-2757.

15. Kelly CM, Juurlink DM, Gomes T, et al. Selective serotonin reuptake inhibitors and breast cancer mortality in women receiving tamoxifen: a population-based cohort study. 2010;340:c693.-doi: 10.1136/bmj.c693.

16. Geisler J, Lonning PE. Impact of aromatase inhibitors on bone health in breast cancer patients. J Steroid Biochem Mol Biol. 2010;118(4–5):294-299.

17. Van Poznak C, Hannon RA, Mackey JR, et al. Prevention of aromatase inhibitor-induced bone loss using risedronate: the SABRE trial. J Clin Oncol. 2010;28(6):967-975.

18. Brufsky AM, Bosserman LD, Caradonna RR, et al. Zoledronic acid effectively prevents aromatase-inhibitor associated bone loss in postmenopausal women with early breast cancer receiving adjuvant letrozole. Z-FAST study 36-month follow-up results. Clin Breast Cancer. 2009;9(2):77-85.

19. Kanis JA, Borgstrom F, De Laet C, et al. Assessment of fracture risk. Osteoporosis Int. 2005;16(6):581-589.

20. Reeder JG, Brufsky AM. The role of bisphosphonates in the adjuvant setting for breast cancer. Oncology. 2010;24(6):462-467,475.

21. Nabholtz JM. Long-term safety of aromatase inhibitors in the treatment of breast cancer. Ther Clin Risk Manag. 2008;4(1):189-204.

22. Cuppone F, Bria E, Verma S, et al. Do adjuvant aromatase inhibitors increase the cardiovascular risk in postmenopausal women with early breast cancer? Meta-analysis of randomized trials. Cancer. 2008;112(2):260-267.

23. Kendall A, Dowsett M, Folkerd E, Smith I. Caution: Vaginal estradiol appears to be contraindicated in postmenopausal women on adjuvant aromatase inhibitors. Ann Oncol. 2006;17(4):584-587.

24. Nelson HD. Menopause. Lancet. 2008;371(9614):760-770.

25. Geller SE, Shulman LP, van Breemen RB, et al. Safety and efficacy of black cohosh and red clover for the management of vasomotor symptoms: a randomized controlled trial. Menopause. 2009;16(6):1156-1166.

26. Kaunitz AM. Effective herbal treatment of vasomotor symptoms—are we any closer? Menopause. 2009;16(3):428-429.

27. Archer DF, Dupont CM, Constantine GD, Pickar JH, Olivier S. Study 319 Investigators. Desvenlafaxine for the treatment of vasomotor symptoms associated with menopause: a double-blind, randomized, placebo-controlled trial of efficacy and safety. Am J Obstet Gynecol. 2009;200(3):238.e1-e10.

28. Phillips KA, Ribi K, Sun Z, et al. Cognitive function in postmenopausal women receiving adjuvant letrozole or tamoxifen for breast cancer in the BIG 1-98 randomized trial [published online ahead of print April 10, 2010]. Breast. doi:10.1016/j.breast.2010.03.025.

29. Schilder CM, Seynaeve C, Beex LV, et al. Effects of tamoxifen and exemestane on cognitive functioning of postmenopausal patients with breast cancer: results from the neuropsychological side study of the Tamoxifen and Exemestane Adjuvant Multinational Trial. J Clin Oncol. 2010;28(8):1294-1300.

30. Din OS, Dodwell D, Wakefield RJ, Coleman RE. Aromatase inhibitor-induced arthralgia in early breast cancer: what do we know and how can we find out more? Breast Cancer Res Treat. 2010;120(3):525-538.

31. Burstein HJ. Aromatase inhibitor-associated arthralgia syndrome. Breast. 2007;16(3):223-234.

32. Hershman DL, Kushi LH, Shao T, et al. Early discontinuation and nonadherence to adjuvant hormonal therapy in a cohort of 8,769 early-stage breast cancer patients [published online ahead of print June 28, 2010]. J Clin Oncol. doi: 10.1200/JCO.2009.25.9655.

33. Hershman DL, Shao T, Kushi LH, et al. Early discontinuation and nonadherence to adjuvant hormonal therapy are associated with increased mortality in women with breast cancer [published online ahead of print August 28, 2010]. Breast Cancer Res Treat. doi: 10.1007/ s10549-010-1132-4.

Issue
OBG Management - 22(10)
Issue
OBG Management - 22(10)
Page Number
30-40
Page Number
30-40
Publications
Publications
Topics
Article Type
Display Headline
An ObGyn’s guide to aromatase inhibitors as adjuvant therapy for breast CA
Display Headline
An ObGyn’s guide to aromatase inhibitors as adjuvant therapy for breast CA
Sections
Article Source

PURLs Copyright

Inside the Article
Article PDF Media

Breast cancer prevention and Tx: An evidence-based guide

Article Type
Changed
Thu, 12/15/2022 - 18:30
Display Headline
Breast cancer prevention and Tx: An evidence-based guide

PRACTICE RECOMMENDATIONS

Offer screening magnetic resonance imaging (MRI) to patients with a known BRCA 1 or 2 mutation, a strong family history of breast cancer, or a lifetime risk of breast cancer >20% to 25%. B

For early-stage breast cancer, lumpectomy and sentinel node mapping with excision is the preferred method for staging. A

Monitor patients receiving tamoxifen for signs and symptoms of venous thromboembolism, cataracts, and uterine malignancy, and patients on aromatase inhibitors for the development of osteoporosis. A

Strength of recommendation (SOR)

A Good-quality patient-oriented evidence
B
Inconsistent or limited-quality patient-oriented evidence
C
Consensus, usual practice, opinion, disease-oriented evidence, case series

Late last year, the US Preventive Services Task Force (USPSTF) sparked a nationwide controversy when it announced that it was recommending against routine screening mammography for women younger than age 50.1 Indeed, that’s a recommendation that many other organizations, including the American Cancer Society (ACS),2 the American College of Obstetricians and Gynecologists (ACOG),3 and the National Comprehensive Cancer Network (NCCN),4 disagree with. But the age at which women should begin routine mammography isn’t the only controversial question. Experts disagree on the benefits of breast self-examination, the optimal frequency of clinical breast exams, and the use of digital mammography—among other issues. This evidence-based review can help you cut through the confusion.

CASE Carrie, a 39-year-old woman who has never been pregnant, comes in for an annual Pap smear and gynecologic exam. She has a negative past medical history, but a positive family history for breast cancer—both her mother and 1 of her sisters had the disease. How would you assess Carrie’s risk of breast cancer, and what preventive measures would you recommend?

Use this predictive model to pinpoint your patient’s risk

When making decisions regarding primary prevention of and screening for breast cancer, an accurate assessment of risk is critical. Many predictive models have been developed with that in mind. The most widely studied, the Gail model, incorporates a number of important risk factors (TABLE 1), including age; race; family history; reproductive factors such as age of menarche, menopause, and first childbirth; and previous history of breast biopsy and atypical findings, to calculate a woman’s 5-year risk.5

A risk calculator (the Breast Cancer Risk Assessment Tool) based on the Gail model is available on the National Cancer Institute’s Web site, at http://www.cancer.gov/bcrisktool. Generally, a score ≥1.66%,5 which indicates that a patient has at least a 1.66% chance of developing breast cancer over the next 5 years, is considered high risk.6,7

CASE Carrie’s 2 first-degree relatives affected by breast cancer and her nulliparous status place her at increased risk. Further questioning reveals a particularly strong family history, as both relatives were diagnosed before the age of 50 (her mom at 45 years of age and her sister, at 39). Carrie’s 5-year risk is 1.8%.

TABLE 1
Risk factors for breast cancer5,29

  • Age (>50 years)
  • Sex
  • Ethnicity*
  • Family history (≥1 first-degree relative diagnosed with breast cancer, particularly if diagnosed at <50 years of age)
  • Early menarche (<12 years)
  • Late menopause (>55 years)
  • Late age at first full-term pregnancy (>30 years)
  • Nulliparity
  • Breast density
  • History of breast biopsies
  • Atypical hyperplasia or LCIS on prior biopsy
  • History of radiation to chest wall
  • Lack of breastfeeding
  • Physical inactivity
  • Obesity
  • Alcohol use
  • Exogenous hormones (HRT)
*African American and Caucasian women are at higher risk compared with Asian, Hispanic, and Native American women.
1 drink/day results in minimal increase in risk; 2-5 drinks/day result in 1.5 increased risk compared with nondrinkers.
HRT, hormone replacement therapy; LCIS, lobular carcinoma in situ.

All women can benefit from these preventive measures

As primary care physicians, we have a responsibility to stress lifestyle modification as the mainstay of breast cancer prevention. Whether or not a woman is at high risk, advise her that maintaining a normal weight, exercising vigorously, limiting alcohol consumption, and breastfeeding are evidence-based methods of primary prevention. Diets low in fat and high in fiber may be associated with a lower risk of invasive breast cancer, but there is no conclusive evidence to support specific dietary interventions to reduce the risk.8-11 Nor has a link between active or passive smoking, antioxidants, or fruit and vegetable intake been firmly established.12

There is a clear association between prolonged estrogen exposure and breast cancer, however. Many reproductive factors, such as early menarche, late menopause, later age at time of first full-term pregnancy, and nulliparity, increase a woman’s exposure to endogenous estrogen—and her risk of developing breast cancer.12,13

Exposure to exogenous estrogen is also linked to the development of breast cancer. In 2002, the Women’s Health Initiative (WHI) was stopped early after a report was released stating that the risks of hormone replacement therapy (HRT)—a higher incidence of cardiovascular events, stroke, and venous thromboembolism, as well as breast cancer—outweighed the benefits.14 Subsequent analyses have found a relationship between the declining incidence of breast cancer and the marked decrease in HRT use prompted by the WHI report. While causality has not been firmly established, multiple studies strongly suggest it.15,16

 

 

The association between oral contraceptives (OCs) and breast cancer is more controversial. Some studies have found an increased breast cancer risk among OC users, but both the relative risk and absolute risk were found to be very small and to dissipate 10 years after stopping OC use. More recent studies with newer formulations containing lower doses of estrogen have failed to show an increased risk.8

Breast cancer screening: The parameters have changed

Various organizations have published guidelines for breast cancer screening (TABLE 2), and all are somewhat different. Here’s what you need to know.

Breast self-examination (BSE), which women were previously advised to perform monthly, has not been shown to improve mortality in any age group, and is no longer routinely recommended.17 While both the USPSTF and the Canadian Task Force on Preventive Health Care recommend against teaching women BSE, the ACS, ACOG, and NCCN encourage self-examination—particularly among women older than 40 years.1-4,17,18

Clinical breast examination has an average sensitivity of 50% and detects approximately 5% of mammographically occult cancers.19 It is still not clear whether clinical breast exams save lives, however—a finding that is reflected in the USPSTF’s “I” (insufficient evidence to assess the benefits and harms) recommendation.1 Other consensus guidelines still recommend clinical breast examination, albeit at varying frequencies.

Screening mammography decreases mortality rates by anywhere from 28% to 65%, depending on the statistical model used.20 The benefit is greatest in women between the ages of 50 and 69 years, however, and most groups agree that mammography every 1 to 2 years is advisable for this age group. (There is limited data on the value of mammography for women 70 years of age and older, and no consensus on the age at which to stop screening.1,21) But because the mortality benefit from screening mammography is lower for women aged 40 to 49, guidelines for this age group are more controversial.

Mammography’s sensitivity is affected by a variety of factors, including age and menopausal status, prior breast surgery or radiation, breast density, and the experience of the radiologist. Women in their 40s have denser breast tissue than older women, making mammography less sensitive for this age group. Because of that, and because the overall incidence of breast cancer is lower for women younger than 50, some argue that screening mammography for women between the ages of 40 and 49 years leads to unacceptably high false-positive rates (9.8% annually22) and that the harm associated with mammography may outweigh the benefit. Others counter that tumors in younger women tend to be more aggressive and faster growing, making early detection even more critical than for older women.

What should you advise women in this age group? You might point out that the USPSTF recommends against routine screening, but indicates that the decision to begin (or defer) routine mammography before age 50 should be individualized, based on the needs and values of each patient.1

Digital mammography. A recent study of more than 43,000 women demonstrated that digital mammography is more accurate than film—but only for certain groups: These include women <50 years of age, women with dense breasts, and pre- and perimenopausal women. 23 Because it is still not clear whether the increased accuracy will translate into a mortality benefit, more research is needed before digital mammography is widely adopted. The USPSTF maintains that there is insufficient evidence to assess the benefits and harms of using either digital mammography or magnetic resonance imaging (MRI) rather than film mammography to screen for breast cancer.1

MRI. In 2007, the ACS published guidelines on the use of MRI as an adjunct to mammography for breast cancer screening in high-risk women.24 According to ACS guidelines, screening MRI should be offered to patients with a known BRCA 1 or 2 mutation (5%-10% of all breast cancers are associated with a mutation in the BRCA 1 or BRCA 2 gene, which is transmitted in an autosomal dominant pattern6). It also should be offered to those with a strong family history, or a lifetime risk of developing breast cancer that is >20% to 25%. And finally, MRI should be offered to women who had chest wall radiation when they were between the ages of 10 and 30 years—another significant risk factor for breast cancer—and those with other genetic syndromes that increase their lifetime risk of breast cancer.24

 

 

Evidence is insufficient for or against MRI screening for women with a personal history of breast cancer, atypical hyperplasia, or lobular carcinoma in situ, however, and neither breast ultrasound (which is generally used diagnostically, not for screening purposes) nor MRI has been shown to be helpful as a screening tool in women with <15% lifetime risk of developing breast cancer.24,25

TABLE 2
Guidelines for breast cancer screening for women with average risk

OrganizationAge (years)Breast self-examClinical breast examMammography
American Cancer Society220-40OptionalEvery 3 yNA
>40EncouragesAnnuallyAnnually
American College of Obstetricians and Gynecologists340-49EncouragesAnnually1-2 y
50-69EncouragesAnnuallyAnnually
Canadian Task force on Preventive Health Care1840-49Recommends against teachingInsufficient evidenceInsufficient evidence
50-69Recommends against teaching1-2 y1-2 y
National Comprehensive Cancer Network420-40Encourages1-3 yNA
>40EncouragesAnnuallyAnnually
US Preventive Services Task Force140-49Recommends against teachingInsufficient evidenceNot routinely recommended
50-74Recommends against teachingInsufficient evidenceEvery 2 y
NA, not addressed.

When to consider chemoprevention

For women like Carrie, who are at high risk of developing breast cancer, selective estrogen receptor modulator (SERM) therapy and surgical interventions may be options to consider. The Breast Cancer Prevention Trial demonstrated the efficacy of tamoxifen as a preventive agent. This landmark trial showed that for high-risk women older than 35, 5 years of tamoxifen therapy can reduce the incidence of invasive breast cancer by nearly 50%.26

Women with the BRCA 1 or 2 mutation—all of whom should be offered genetic counseling—were included in the study. Tamoxifen reduced the incidence of breast cancer in BRCA 2 carriers by 62%, the researchers found, but did not reduce risk in carriers of the BRCA 1 gene. This is likely due to the high prevalence of estrogen receptor-negative breast cancers among BRCA 1 carriers.26

More recently, the Study of Tamoxifen and Raloxifene (STAR) trial compared the efficacy of tamoxifen and raloxifene, a second-generation SERM, in high-risk postmenopausal women ages 35 and older. The drugs were found to be equally effective in reducing the risk of invasive breast cancer, but raloxifene had a better side effect profile, with a lower incidence of thromboembolism and cataracts. 27

What the guidelines call for. In 2003, the USPSTF recommended that clinicians discuss chemoprevention with women at high risk for breast cancer and low risk for adverse effects of SERMs.28

The most recent update to the NCCN breast cancer risk reduction guidelines recommends that clinicians offer tamoxifen to premenopausal women with a 5-year projected breast cancer risk ≥1.7% and offer tamoxifen or raloxifene to high-risk postmenopausal women.29 It is worth noting, however, that SERMs can have significant adverse effects, including venous thromboembolism, stroke, cataracts, uterine malignancy, and hot flashes, while lifestyle modifications and the avoidance of HRT have few, if any, negative effects.

CASE After consultation with a genetic counselor, Carrie underwent testing for both the BRCA 1 and BRCA 2 mutations. She tested negative for both. She declined chemoprevention and prophylactic surgery, opting for enhanced screening with yearly mammography and MRI and lifestyle modification instead.

When a mass is found

For women ages 30 or older with palpable masses or solid masses ≥2 cm found on imaging, core needle biopsy is recommended.30,31 Biopsy is indicated for women younger than 30 as well, if the mass is >2 cm or imaging is suspicious. In general, a needle biopsy read as benign is considered adequate for diagnostic purposes only if the lesion appeared benign on imaging.

For lesions shown to be cystic on imaging, recommendations for follow-up or additional testing are based on the characteristics of the cyst. For simple cysts, 2- to 4-month follow up for stability, followed by routine screening, is adequate.21 Additional evaluation of complex cysts is indicated, including aspiration for complicated cysts and biopsy for complex cysts. After aspiration, surgical excision of bloody aspirates or persistent masses is recommended.30,31

Staging using the TNM system
The TNM (tumor, node, metastases) classification system is used for the staging of breast cancer:

  • T refers to the tumor type, size, and extent of local involvement
  • N describes regional lymph node involvement
  • M refers to distant metastases.

The TNM classifications are also grouped by stage (I through IV).,

Lumpectomy and sentinel node mapping with excision is the preferred method for staging of early-stage breast cancer without palpable lymphadenopathy—provided that the surgical team has documented experience with sentinel node biopsy.32 Sentinel node biopsy is preferred because of its safety, low (<10%) false negative rate, and decreased morbidity compared with full axillary dissection, although dissection is recommended for patients with more advanced cancer or a positive sentinel node.32 The comparative effects of sentinel node biopsy vs axillary node dissection on tumor recurrence and patient survival are not known.33

 

 

Testing for tumor markers such as estrogen and progesterone receptors and human epidermal growth factor receptor 2 (HER2) expression status in biopsy-proven breast cancer is now the standard of care. Seventy percent of breast cancers are estrogen receptor-positive, with increasing frequency associated with older age.34 Estrogen/progesterone receptor positivity is associated with a more favorable outcome, and multiple hormonal therapies can be aimed at these receptors.34 While HER2 overexpression—which occurs in 15% to 30% of newly diagnosed breast cancers35—is associated with more aggressive tumors, women with this type of tumor cell can benefit from trastuzumab, an anti-HER2 drug.36

Key factors that affect prognosis

Important factors affecting prognosis and treatment of localized breast cancer are tumor size, age and menopausal status, tumor expression of hormone receptors and/ or the HER2 protein, as well as the status of the draining axillary nodes. Factors that predict a greater chance of recurrence include the spread of disease to axillary nodes, larger tumor size, invasive histology, inflammatory pathology, lack of estrogen/progesterone receptors, and age <50 years or premenopausal status.

Treatment options include surgical resection, radiation, and systemic adjuvant therapy in the form of chemotherapy, endocrine therapy, or anti-HER2 monoclonal antibodies.37 (For more on treatment, see “Surgery, radiation, and systemic therapy: Making the most of what’s in our arsenal” at jfponline.com.)

Don’t overlook quality-of-life issues
Follow-up of breast cancer patients should go beyond treatment and work-up for recurrence and metastatic disease to focus on health and lifestyle issues, such as stress reduction, mood, smoking cessation, diet and exercise, treatment of hot flashes, sexual dysfunction, and bone health. A recent study found both reduced recurrence and increased survival in women receiving psychological interventions to improve quality-of-life measures after an 11-year follow-up.38

Refer women to targeted Web sites such as the National Breast Cancer Awareness Month organization (http://www.nbcam.org/), the National Breast Cancer Foundation (http://community.nationalbreastcancer.org/), and the Susan G. Komen Breast Cancer Foundation (http://ww5.komen.org/). Offer treatment for bothersome symptoms. Hot flashes and depression, for example, often related to endocrine therapy, can be treated with selective serotonin reuptake inhibitors (SSRIs). That said, some SSRIs decrease the active metabolite of tamoxifen by inhibiting CYP2D6 enzyme and must, therefore, be used with caution. However, venlafaxine and citalopram are less likely to alter tamoxifen metabolism than other SSRIs.39

CASE When Carrie was 47, she had an abnormal MRI of the left breast. Core needle biopsy and pathology of the lesion revealed an estrogen and progesterone receptor-positive tumor that was negative for HER2 overexpression. She underwent lumpectomy, which revealed a 1.5 cm tumor, followed by a negative sentinel node biopsy, and was diagnosed with stage I (T1N0M0) breast cancer. Carrie had radiation after surgery; she did not require chemotherapy, but was told to take tamoxifen for 5 years. This adjuvant endocrine therapy led to hot flashes and depression, both of which were successfully treated with venlafaxine. Carrie is currently cancer-free and participates in a breast cancer survivor program that includes regular visits with her primary physician and her oncologist.

CORRESPONDENCE Denise Sur, MD, 1920 Colorado Avenue, Santa Monica, CA 90404; [email protected] 

Online exclusive

Surgery, radiation, and systemic therapy: Making the most of what’s in our arsenal

Breast cancer surgery has changed dramatically over the years. Multiple studies have shown that breast-conserving therapy (lumpectomy followed by radiation) for carefully selected women is comparable to mastectomy for local recurrence and survival. While there has been much interest in determining whether a subset of patients could forego radiation after lumpectomy, a meta-analysis by the Early Breast Cancer Trialists Collaborative Group demonstrated that radiation after lumpectomy provides an absolute local recurrence risk reduction of 19%, and a 5.4% absolute reduction in 15-year breast cancer mortality rates compared with lumpectomy without radiation.1 Thus, radiation after lumpectomy remains the standard of care for all women undergoing breast-conserving therapy, regardless of tumor characteristics.

In certain women with a high risk of recurrence (≥4 positive nodes), radiation is also recommended after mastectomy. Women undergoing mastectomy have numerous options for immediate or delayed breast reconstruction. Consultation with a multidisciplinary team, including a plastic surgeon, prior to any surgical intervention is advised.2

Multiple systemic chemotherapy regimens have been shown to be beneficial in carefully selected patients with breast cancer. Systematic reviews have demonstrated that an anthracycline-based regimen can decrease annual breast cancer mortality by 38% in women <50 years old and by 20% in women ages 50 to 69 years.1 in more recent randomized controlled trials, the addition of taxanes to anthracycline-based regimens has produced promising results.3

Numerous hormonal therapies benefit women with estrogen or progesterone receptor-positive breast cancer. Tamoxifen blocks the activity of estrogen on receptors located in breast cancer tissue, for example; aromatase inhibitors block the conversion of androgens to estrogen; and gonadotropin-releasing hormone (GnRH) analogs such as leuprolide and goserelin suppress ovarian production of estrogen.

For postmenopausal women, options include an aromatase inhibitor alone or tamoxifen followed by an aromatase inhibitor.

In premenopausal women, aromatase inhibitors are not very effective, as decreasing peripheral estrogen stimulates the ovaries to produce more estrogen. Thus, for these patients, adjuvant endocrine therapy consists of tamoxifen, with ovarian ablation (via surgery or radiation) or ovarian suppression with a GnRH analog. If the patient goes through menopause as a result of this therapy, she may benefit from aromatase inhibitors at that time.4,5

Women with breast cancer that overexpresses the HER2 gene benefit from adjuvant treatment with trastuzumab, an anti-HER2 antibody.6 While current guidelines advise treatment for 1 year, multiple studies are evaluating dosing schedules and optimal duration of treatment. for now, patients should be monitored for signs of cardiotoxicity at baseline and every 3 months thereafter until completion of therapy.4

References

1. Early Breast Caner Trialists’ Collaborative Group (EBCTCG). Effects of chemotherapy and hormonal therapy for early breast cancer on recurrence and 15-year survival: an overview of the randomised trials. Lancet. 2005;265:1687-1717.

2. Codeiro P. Breast reconstruction after surgery for breast cancer. N Engl J Med. 2008;359:1590-1601.

3. DeLaurentiis M, Cancello G, D’Agostino D, et al. Taxane-based combinations as adjuvant chemotherapy of early breast cancer: a meta-analysis of randomized trials. J Clin Oncol. 2008;26:44-53.

4. National Comprehensive Cancer Network. Breast cancer risk reduction clinical practice guidelines in oncology. J Natl Compr Canc Netw. 2003;1:280-296.

5. Baum M, Budzar AU, Cuzick J, et al. Anastrozole alone or in combination with tamoxifen versus tamoxifen alone for adjuvant treatment of postmenopausal women with early breast cancer: first results of the ATAC randomised trial. Lancet. 2002;359:2131-2139.

6. Piccart-Gebhart MJ, Procter M, Leyland-Jones B, et al. Herceptin adjuvant (HERA) Trial Study Team. Trastuzumab after adjuvant chemotherapy in HER2-positive breast cancer. N Engl J Med. 2005;353:1659-1672.

References

1. US Preventive Services Task Force Screening for breast cancer. Available at: http://www.uspreventiveservicestaskforce.org/uspstf/uspsbrca.htm. Accessed September 2, 2010.

2. American Cancer Society. American Cancer Society guidelines for the early detection of cancer. Available at: http://www.cancer.org/Healthy/FindCancerEarly/CancerScreeningGuidelines/american-cancer-society-guidelines-for-the-early-detection-of-cancer. Accessed September 2, 2010.

3. American College of Obstetricians and Gynecologists. Response of the American College of Obstetricians and Gynecologists to the new breast cancer screening recommendations from the US Preventive Services Task Force. Available at: http://www.acog.org/from_home/misc/uspstfresponse.cfm. Accessed September 2, 2010.

4. National Comprehensive Cancer Network. Breast cancer screening and diagnosis. Clinical Practice Guidelines in Oncology-V.1.2010. Fort Washington, Pa: National Comprehensive Cancer Network: November 3, 2009. Available at: http://www.nccn.org/professionals/physician_gls/f_guidelines.asp. Accessed September 17 , 2010.

5. National Cancer Institute. Breast cancer risk assessment tool. Available at: http://www.cancer.gov/bcrisktool/. Accessed September 2, 2010

6. Robson M, Offit K. Clinical practice. Management of an inherited predisposition to breast cancer. N Engl J Med. 2007;357:154-162.

7. Fisher B, Costantino JP, Wickerham DL, et al. Tamoxifen for prevention of breast cancer: report of the National Surgical Adjuvant Breast and Bowel Project P-1 Study. J Natl Cancer Inst. 1998;90:1371-1388.

8. Cummings SR, Tice JA, Bauer S, et al. Prevention of breast cancer in postmenopausal women: approaches to estimating and reducing risk. J Natl Cancer Inst. 2009;18:101,-384-398.

9. Prentice RL, Caan B, Chlebowski RT, et al. Low-fat dietary pattern and risk of invasive breast cancer: the Women’s Health Initiative Randomized Controlled Dietary Modification Trial. JAMA. 2006;295:629-642.

10. Pierce JP, Natarajan L, Caan BJ, et al. Influence of a diet very high in vegetables, fruit, and fiber and low in fat on prognosis following treatment for breast cancer: the Women’s Healthy Eating and Living (WHEL) randomized trial. JAMA. 2007;298:289-298.

11. Chlebowski RT, Blackburn GL, Thomson CA, et al. Dietary fat reduction and breast cancer outcome: interim efficacy results from the Women’s Intervention Nutrition Study (WINS). J Natl Cancer Inst. 2006;98:1767-1776.

12. PDQ Cancer Information Summary. Breast Cancer Prevention (PRQ) – Health Professional. Date last modified April 30, 2009. Available at: http://www.cancer.gov/cancertopics/pdq/prevention/breast/healthprofessional. Accessed May 12, 2009.

13. Hulka BS, Moorman PG. Breast cancer: hormones and other risk factors. Maturitas. 2001;38:103-113.

14. Rossouw JE, Anderson GL, Prentice RL, et al. Risks and benefits of estrogen plus progestin in healthy postmenopausal women: principal results From the Women’s Health Initiative randomized controlled trial. JAMA. 2002;288:321-333.

15. Ravdin PM, Cronin KA, Howlader N, et al. The decrease in breast-cancer incidence in 2003 in the United States. N Engl J Med. 2007;356:1670-1674.

16. Chlebowski RT, Kuller LH, Prentice RL, et al. WHI Investigators. Breast cancer after use of estrogen plus progestin in postmenopausal women. N Engl J Med. 2009;360:573-587.

17. Kosters JP, Gotzsche PC. Regular self-examination or clinical examination for early detection of breast cancer. Cochrane Database Syst Rev. 2003;(2):CD003373.-

18. Canadian Task Force on the Periodic Health Examination Ottawa, Canada: Health Canada; 1994:788-795 (reaffirmed by the Canadian Task Force on the Periodic Health Examination 1999, 2001).Available at: http://www.ctfphc.org/index/html. Accessed August 12, 2009.

19. Green BB, Taplin SH. Breast cancer screening controversies. J Am Board Fam Pract. 2003;16:233-241.

20. Berry DA, Cronin KA, Plevritis SK, et al. Effect of screening and adjuvant therapy on mortality from breast cancer. N Engl J Med. 2005;353:1784-1792.

21. Tice JA, Kerlikowske K. Screening and prevention of breast cancer in primary care. Prim Care. 2009;36:533-558.

22. US Preventive Services Task Force. Screening for breast cancer. Systematic evidence review update for the US Preventive Services Task Force. Available at: http://www.ncbi.nlm.nih.gov/bookshelf/br.fcgi?book=es74. Accessed September 2, 2010.

23. Pisano ED, Gatsonis C, Hendrick E, et al. Digital Mammographic Imaging Screening Trial (DMIST) Investigators Group. Diagnostic performance of digital versus film mammography for breast-cancer screening. N Engl J Med. 2005;353:1773-1783.

24. Saslow D, Boetes C, Burke W, et al. American Cancer Society Breast Cancer Advisory Group. American Cancer Society guidelines for breast screening with MRI as an adjunct to mammography. CA Cancer J Clin. 2007;57:75-89.

25. Berg W, Blume J, Cormack J, et al. Combined screening with ultrasound and mammography vs mammography alone in women at elevated risk of breast cancer. JAMA. 2008;299:2151-2163.

26. King M, Wieand S, Hale K, et al. Tamoxifen and breast cancer incidence among women with inherited mutations in BRCA1 and BRCA2: National Surgical Adjuvant Breast and Bowel Project (NSABP-P1) Breast Cancer Prevention Trial. JAMA. 2001;286:2251-2256.

27. Vogel VG, Costantino JP, Wickerham DL, et al. Effects of tamoxifen vs raloxifene on the risk of developing invasive breast cancer and other disease outcomes: the NSABP Study of Tamoxifen and Raloxifene (STAR) P-2 trial. JAMA. 2006;295:2727-2741.

28. Berg AO. US Preventive Services Task Force. Chemoprevention of breast cancer: recommendations and rationale. Am J Nurs. 2003;103:107-113.

29. National Comprehensive Cancer Network. Breast cancer risk reduction. Clinical Practice Guidelines in Oncology-V.2.2010. Fort Washington, Pa: National Comprehensive Cancer Network; August 7, 2010. Available at: http://www.nccn.org/professionals/physician_gls/f_guidelines.asp. Accessed September 17, 2010.

30. Sickles EA, Filly RA, Cllen PW. Benign breast lesions: ultrasound detection and diagnosis. Radiology. 1984;151:467.-

31. Beavers TB, Anderson BO, Bonaccio E, et al. NCCN clinical practice guidelines in oncology: breast cancer screening and diagnosis. J Natl Compr Canc Netw. 2009;7:1060-1096.

32. Mansel RE, Fallowfield L, Kissin M, et al. Randomized multicenter trial of sentinel node biopsy versus standard axillary treatment in operable breast cancer: the ALMANAC trial. J Natl Canc Inst. 2006;98:599-609

33. Lyman GH, Guiliano AE, Somerfield MR, et al. American Society of Clinical Oncology guideline recommendations for sentinel lymph node biopsy in early- stage breast cancer. J Clin Oncol. 2005;23:7703-7720.

34. Turner N, Jones A. Management of breast cancer-Part II. BMJ. 2008;337:a540.-

35. Harris L, Fritsche H, Mennel R, et al:. American Society of Clinical Oncology 2007 update of recommendations for the use of tumor markers in breast cancer J Clin Oncol. 2007;25:5287-5312.

36. Piccart-Gebhart MJ, Procter M, Leyland-Jones B, et al. Herceptin adjuvant (HERA) Trial Study Team. Trastuzumab after adjuvant chemotherapy in HER2-positive breast cancer. N Engl J Med. 2005;353:1659-1672.

37. PDQ Cancer Information Summary Breast cancer treatment– health professional. Available at: http://www.cancer.gov/cancertopics/pdq/treatment/breast/HealthProfessional. Accessed May 12, 2009.

38. Anderson BL, Yang HC, Farrar WB, et al. Psychologic intervention improves survival for breast cancer patients: a randomized clinical trial. Cancer. 2008;113:3450-3458.

39. Jin Y, Desta Z, Stearns V, et al. CYP2D6 genotype, antidepressant use, and tamoxifen metabolism during adjuvant breast cancer treatment. J Natl Cancer Inst. 2005;97:30-39.

Article PDF
Author and Disclosure Information

Denise Sur, MD
Holly Milne, MD
Family Medicine, University of California Los Angeles
[email protected]

The authors reported no potential conflict of interest relevant to this article.

Issue
The Journal of Family Practice - 59(10)
Publications
Topics
Page Number
575-581a
Legacy Keywords
Denise Sur; breast cancer prevention; screening mammography; controversy; breast health
Sections
Author and Disclosure Information

Denise Sur, MD
Holly Milne, MD
Family Medicine, University of California Los Angeles
[email protected]

The authors reported no potential conflict of interest relevant to this article.

Author and Disclosure Information

Denise Sur, MD
Holly Milne, MD
Family Medicine, University of California Los Angeles
[email protected]

The authors reported no potential conflict of interest relevant to this article.

Article PDF
Article PDF

PRACTICE RECOMMENDATIONS

Offer screening magnetic resonance imaging (MRI) to patients with a known BRCA 1 or 2 mutation, a strong family history of breast cancer, or a lifetime risk of breast cancer >20% to 25%. B

For early-stage breast cancer, lumpectomy and sentinel node mapping with excision is the preferred method for staging. A

Monitor patients receiving tamoxifen for signs and symptoms of venous thromboembolism, cataracts, and uterine malignancy, and patients on aromatase inhibitors for the development of osteoporosis. A

Strength of recommendation (SOR)

A Good-quality patient-oriented evidence
B
Inconsistent or limited-quality patient-oriented evidence
C
Consensus, usual practice, opinion, disease-oriented evidence, case series

Late last year, the US Preventive Services Task Force (USPSTF) sparked a nationwide controversy when it announced that it was recommending against routine screening mammography for women younger than age 50.1 Indeed, that’s a recommendation that many other organizations, including the American Cancer Society (ACS),2 the American College of Obstetricians and Gynecologists (ACOG),3 and the National Comprehensive Cancer Network (NCCN),4 disagree with. But the age at which women should begin routine mammography isn’t the only controversial question. Experts disagree on the benefits of breast self-examination, the optimal frequency of clinical breast exams, and the use of digital mammography—among other issues. This evidence-based review can help you cut through the confusion.

CASE Carrie, a 39-year-old woman who has never been pregnant, comes in for an annual Pap smear and gynecologic exam. She has a negative past medical history, but a positive family history for breast cancer—both her mother and 1 of her sisters had the disease. How would you assess Carrie’s risk of breast cancer, and what preventive measures would you recommend?

Use this predictive model to pinpoint your patient’s risk

When making decisions regarding primary prevention of and screening for breast cancer, an accurate assessment of risk is critical. Many predictive models have been developed with that in mind. The most widely studied, the Gail model, incorporates a number of important risk factors (TABLE 1), including age; race; family history; reproductive factors such as age of menarche, menopause, and first childbirth; and previous history of breast biopsy and atypical findings, to calculate a woman’s 5-year risk.5

A risk calculator (the Breast Cancer Risk Assessment Tool) based on the Gail model is available on the National Cancer Institute’s Web site, at http://www.cancer.gov/bcrisktool. Generally, a score ≥1.66%,5 which indicates that a patient has at least a 1.66% chance of developing breast cancer over the next 5 years, is considered high risk.6,7

CASE Carrie’s 2 first-degree relatives affected by breast cancer and her nulliparous status place her at increased risk. Further questioning reveals a particularly strong family history, as both relatives were diagnosed before the age of 50 (her mom at 45 years of age and her sister, at 39). Carrie’s 5-year risk is 1.8%.

TABLE 1
Risk factors for breast cancer5,29

  • Age (>50 years)
  • Sex
  • Ethnicity*
  • Family history (≥1 first-degree relative diagnosed with breast cancer, particularly if diagnosed at <50 years of age)
  • Early menarche (<12 years)
  • Late menopause (>55 years)
  • Late age at first full-term pregnancy (>30 years)
  • Nulliparity
  • Breast density
  • History of breast biopsies
  • Atypical hyperplasia or LCIS on prior biopsy
  • History of radiation to chest wall
  • Lack of breastfeeding
  • Physical inactivity
  • Obesity
  • Alcohol use
  • Exogenous hormones (HRT)
*African American and Caucasian women are at higher risk compared with Asian, Hispanic, and Native American women.
1 drink/day results in minimal increase in risk; 2-5 drinks/day result in 1.5 increased risk compared with nondrinkers.
HRT, hormone replacement therapy; LCIS, lobular carcinoma in situ.

All women can benefit from these preventive measures

As primary care physicians, we have a responsibility to stress lifestyle modification as the mainstay of breast cancer prevention. Whether or not a woman is at high risk, advise her that maintaining a normal weight, exercising vigorously, limiting alcohol consumption, and breastfeeding are evidence-based methods of primary prevention. Diets low in fat and high in fiber may be associated with a lower risk of invasive breast cancer, but there is no conclusive evidence to support specific dietary interventions to reduce the risk.8-11 Nor has a link between active or passive smoking, antioxidants, or fruit and vegetable intake been firmly established.12

There is a clear association between prolonged estrogen exposure and breast cancer, however. Many reproductive factors, such as early menarche, late menopause, later age at time of first full-term pregnancy, and nulliparity, increase a woman’s exposure to endogenous estrogen—and her risk of developing breast cancer.12,13

Exposure to exogenous estrogen is also linked to the development of breast cancer. In 2002, the Women’s Health Initiative (WHI) was stopped early after a report was released stating that the risks of hormone replacement therapy (HRT)—a higher incidence of cardiovascular events, stroke, and venous thromboembolism, as well as breast cancer—outweighed the benefits.14 Subsequent analyses have found a relationship between the declining incidence of breast cancer and the marked decrease in HRT use prompted by the WHI report. While causality has not been firmly established, multiple studies strongly suggest it.15,16

 

 

The association between oral contraceptives (OCs) and breast cancer is more controversial. Some studies have found an increased breast cancer risk among OC users, but both the relative risk and absolute risk were found to be very small and to dissipate 10 years after stopping OC use. More recent studies with newer formulations containing lower doses of estrogen have failed to show an increased risk.8

Breast cancer screening: The parameters have changed

Various organizations have published guidelines for breast cancer screening (TABLE 2), and all are somewhat different. Here’s what you need to know.

Breast self-examination (BSE), which women were previously advised to perform monthly, has not been shown to improve mortality in any age group, and is no longer routinely recommended.17 While both the USPSTF and the Canadian Task Force on Preventive Health Care recommend against teaching women BSE, the ACS, ACOG, and NCCN encourage self-examination—particularly among women older than 40 years.1-4,17,18

Clinical breast examination has an average sensitivity of 50% and detects approximately 5% of mammographically occult cancers.19 It is still not clear whether clinical breast exams save lives, however—a finding that is reflected in the USPSTF’s “I” (insufficient evidence to assess the benefits and harms) recommendation.1 Other consensus guidelines still recommend clinical breast examination, albeit at varying frequencies.

Screening mammography decreases mortality rates by anywhere from 28% to 65%, depending on the statistical model used.20 The benefit is greatest in women between the ages of 50 and 69 years, however, and most groups agree that mammography every 1 to 2 years is advisable for this age group. (There is limited data on the value of mammography for women 70 years of age and older, and no consensus on the age at which to stop screening.1,21) But because the mortality benefit from screening mammography is lower for women aged 40 to 49, guidelines for this age group are more controversial.

Mammography’s sensitivity is affected by a variety of factors, including age and menopausal status, prior breast surgery or radiation, breast density, and the experience of the radiologist. Women in their 40s have denser breast tissue than older women, making mammography less sensitive for this age group. Because of that, and because the overall incidence of breast cancer is lower for women younger than 50, some argue that screening mammography for women between the ages of 40 and 49 years leads to unacceptably high false-positive rates (9.8% annually22) and that the harm associated with mammography may outweigh the benefit. Others counter that tumors in younger women tend to be more aggressive and faster growing, making early detection even more critical than for older women.

What should you advise women in this age group? You might point out that the USPSTF recommends against routine screening, but indicates that the decision to begin (or defer) routine mammography before age 50 should be individualized, based on the needs and values of each patient.1

Digital mammography. A recent study of more than 43,000 women demonstrated that digital mammography is more accurate than film—but only for certain groups: These include women <50 years of age, women with dense breasts, and pre- and perimenopausal women. 23 Because it is still not clear whether the increased accuracy will translate into a mortality benefit, more research is needed before digital mammography is widely adopted. The USPSTF maintains that there is insufficient evidence to assess the benefits and harms of using either digital mammography or magnetic resonance imaging (MRI) rather than film mammography to screen for breast cancer.1

MRI. In 2007, the ACS published guidelines on the use of MRI as an adjunct to mammography for breast cancer screening in high-risk women.24 According to ACS guidelines, screening MRI should be offered to patients with a known BRCA 1 or 2 mutation (5%-10% of all breast cancers are associated with a mutation in the BRCA 1 or BRCA 2 gene, which is transmitted in an autosomal dominant pattern6). It also should be offered to those with a strong family history, or a lifetime risk of developing breast cancer that is >20% to 25%. And finally, MRI should be offered to women who had chest wall radiation when they were between the ages of 10 and 30 years—another significant risk factor for breast cancer—and those with other genetic syndromes that increase their lifetime risk of breast cancer.24

 

 

Evidence is insufficient for or against MRI screening for women with a personal history of breast cancer, atypical hyperplasia, or lobular carcinoma in situ, however, and neither breast ultrasound (which is generally used diagnostically, not for screening purposes) nor MRI has been shown to be helpful as a screening tool in women with <15% lifetime risk of developing breast cancer.24,25

TABLE 2
Guidelines for breast cancer screening for women with average risk

OrganizationAge (years)Breast self-examClinical breast examMammography
American Cancer Society220-40OptionalEvery 3 yNA
>40EncouragesAnnuallyAnnually
American College of Obstetricians and Gynecologists340-49EncouragesAnnually1-2 y
50-69EncouragesAnnuallyAnnually
Canadian Task force on Preventive Health Care1840-49Recommends against teachingInsufficient evidenceInsufficient evidence
50-69Recommends against teaching1-2 y1-2 y
National Comprehensive Cancer Network420-40Encourages1-3 yNA
>40EncouragesAnnuallyAnnually
US Preventive Services Task Force140-49Recommends against teachingInsufficient evidenceNot routinely recommended
50-74Recommends against teachingInsufficient evidenceEvery 2 y
NA, not addressed.

When to consider chemoprevention

For women like Carrie, who are at high risk of developing breast cancer, selective estrogen receptor modulator (SERM) therapy and surgical interventions may be options to consider. The Breast Cancer Prevention Trial demonstrated the efficacy of tamoxifen as a preventive agent. This landmark trial showed that for high-risk women older than 35, 5 years of tamoxifen therapy can reduce the incidence of invasive breast cancer by nearly 50%.26

Women with the BRCA 1 or 2 mutation—all of whom should be offered genetic counseling—were included in the study. Tamoxifen reduced the incidence of breast cancer in BRCA 2 carriers by 62%, the researchers found, but did not reduce risk in carriers of the BRCA 1 gene. This is likely due to the high prevalence of estrogen receptor-negative breast cancers among BRCA 1 carriers.26

More recently, the Study of Tamoxifen and Raloxifene (STAR) trial compared the efficacy of tamoxifen and raloxifene, a second-generation SERM, in high-risk postmenopausal women ages 35 and older. The drugs were found to be equally effective in reducing the risk of invasive breast cancer, but raloxifene had a better side effect profile, with a lower incidence of thromboembolism and cataracts. 27

What the guidelines call for. In 2003, the USPSTF recommended that clinicians discuss chemoprevention with women at high risk for breast cancer and low risk for adverse effects of SERMs.28

The most recent update to the NCCN breast cancer risk reduction guidelines recommends that clinicians offer tamoxifen to premenopausal women with a 5-year projected breast cancer risk ≥1.7% and offer tamoxifen or raloxifene to high-risk postmenopausal women.29 It is worth noting, however, that SERMs can have significant adverse effects, including venous thromboembolism, stroke, cataracts, uterine malignancy, and hot flashes, while lifestyle modifications and the avoidance of HRT have few, if any, negative effects.

CASE After consultation with a genetic counselor, Carrie underwent testing for both the BRCA 1 and BRCA 2 mutations. She tested negative for both. She declined chemoprevention and prophylactic surgery, opting for enhanced screening with yearly mammography and MRI and lifestyle modification instead.

When a mass is found

For women ages 30 or older with palpable masses or solid masses ≥2 cm found on imaging, core needle biopsy is recommended.30,31 Biopsy is indicated for women younger than 30 as well, if the mass is >2 cm or imaging is suspicious. In general, a needle biopsy read as benign is considered adequate for diagnostic purposes only if the lesion appeared benign on imaging.

For lesions shown to be cystic on imaging, recommendations for follow-up or additional testing are based on the characteristics of the cyst. For simple cysts, 2- to 4-month follow up for stability, followed by routine screening, is adequate.21 Additional evaluation of complex cysts is indicated, including aspiration for complicated cysts and biopsy for complex cysts. After aspiration, surgical excision of bloody aspirates or persistent masses is recommended.30,31

Staging using the TNM system
The TNM (tumor, node, metastases) classification system is used for the staging of breast cancer:

  • T refers to the tumor type, size, and extent of local involvement
  • N describes regional lymph node involvement
  • M refers to distant metastases.

The TNM classifications are also grouped by stage (I through IV).,

Lumpectomy and sentinel node mapping with excision is the preferred method for staging of early-stage breast cancer without palpable lymphadenopathy—provided that the surgical team has documented experience with sentinel node biopsy.32 Sentinel node biopsy is preferred because of its safety, low (<10%) false negative rate, and decreased morbidity compared with full axillary dissection, although dissection is recommended for patients with more advanced cancer or a positive sentinel node.32 The comparative effects of sentinel node biopsy vs axillary node dissection on tumor recurrence and patient survival are not known.33

 

 

Testing for tumor markers such as estrogen and progesterone receptors and human epidermal growth factor receptor 2 (HER2) expression status in biopsy-proven breast cancer is now the standard of care. Seventy percent of breast cancers are estrogen receptor-positive, with increasing frequency associated with older age.34 Estrogen/progesterone receptor positivity is associated with a more favorable outcome, and multiple hormonal therapies can be aimed at these receptors.34 While HER2 overexpression—which occurs in 15% to 30% of newly diagnosed breast cancers35—is associated with more aggressive tumors, women with this type of tumor cell can benefit from trastuzumab, an anti-HER2 drug.36

Key factors that affect prognosis

Important factors affecting prognosis and treatment of localized breast cancer are tumor size, age and menopausal status, tumor expression of hormone receptors and/ or the HER2 protein, as well as the status of the draining axillary nodes. Factors that predict a greater chance of recurrence include the spread of disease to axillary nodes, larger tumor size, invasive histology, inflammatory pathology, lack of estrogen/progesterone receptors, and age <50 years or premenopausal status.

Treatment options include surgical resection, radiation, and systemic adjuvant therapy in the form of chemotherapy, endocrine therapy, or anti-HER2 monoclonal antibodies.37 (For more on treatment, see “Surgery, radiation, and systemic therapy: Making the most of what’s in our arsenal” at jfponline.com.)

Don’t overlook quality-of-life issues
Follow-up of breast cancer patients should go beyond treatment and work-up for recurrence and metastatic disease to focus on health and lifestyle issues, such as stress reduction, mood, smoking cessation, diet and exercise, treatment of hot flashes, sexual dysfunction, and bone health. A recent study found both reduced recurrence and increased survival in women receiving psychological interventions to improve quality-of-life measures after an 11-year follow-up.38

Refer women to targeted Web sites such as the National Breast Cancer Awareness Month organization (http://www.nbcam.org/), the National Breast Cancer Foundation (http://community.nationalbreastcancer.org/), and the Susan G. Komen Breast Cancer Foundation (http://ww5.komen.org/). Offer treatment for bothersome symptoms. Hot flashes and depression, for example, often related to endocrine therapy, can be treated with selective serotonin reuptake inhibitors (SSRIs). That said, some SSRIs decrease the active metabolite of tamoxifen by inhibiting CYP2D6 enzyme and must, therefore, be used with caution. However, venlafaxine and citalopram are less likely to alter tamoxifen metabolism than other SSRIs.39

CASE When Carrie was 47, she had an abnormal MRI of the left breast. Core needle biopsy and pathology of the lesion revealed an estrogen and progesterone receptor-positive tumor that was negative for HER2 overexpression. She underwent lumpectomy, which revealed a 1.5 cm tumor, followed by a negative sentinel node biopsy, and was diagnosed with stage I (T1N0M0) breast cancer. Carrie had radiation after surgery; she did not require chemotherapy, but was told to take tamoxifen for 5 years. This adjuvant endocrine therapy led to hot flashes and depression, both of which were successfully treated with venlafaxine. Carrie is currently cancer-free and participates in a breast cancer survivor program that includes regular visits with her primary physician and her oncologist.

CORRESPONDENCE Denise Sur, MD, 1920 Colorado Avenue, Santa Monica, CA 90404; [email protected] 

Online exclusive

Surgery, radiation, and systemic therapy: Making the most of what’s in our arsenal

Breast cancer surgery has changed dramatically over the years. Multiple studies have shown that breast-conserving therapy (lumpectomy followed by radiation) for carefully selected women is comparable to mastectomy for local recurrence and survival. While there has been much interest in determining whether a subset of patients could forego radiation after lumpectomy, a meta-analysis by the Early Breast Cancer Trialists Collaborative Group demonstrated that radiation after lumpectomy provides an absolute local recurrence risk reduction of 19%, and a 5.4% absolute reduction in 15-year breast cancer mortality rates compared with lumpectomy without radiation.1 Thus, radiation after lumpectomy remains the standard of care for all women undergoing breast-conserving therapy, regardless of tumor characteristics.

In certain women with a high risk of recurrence (≥4 positive nodes), radiation is also recommended after mastectomy. Women undergoing mastectomy have numerous options for immediate or delayed breast reconstruction. Consultation with a multidisciplinary team, including a plastic surgeon, prior to any surgical intervention is advised.2

Multiple systemic chemotherapy regimens have been shown to be beneficial in carefully selected patients with breast cancer. Systematic reviews have demonstrated that an anthracycline-based regimen can decrease annual breast cancer mortality by 38% in women <50 years old and by 20% in women ages 50 to 69 years.1 in more recent randomized controlled trials, the addition of taxanes to anthracycline-based regimens has produced promising results.3

Numerous hormonal therapies benefit women with estrogen or progesterone receptor-positive breast cancer. Tamoxifen blocks the activity of estrogen on receptors located in breast cancer tissue, for example; aromatase inhibitors block the conversion of androgens to estrogen; and gonadotropin-releasing hormone (GnRH) analogs such as leuprolide and goserelin suppress ovarian production of estrogen.

For postmenopausal women, options include an aromatase inhibitor alone or tamoxifen followed by an aromatase inhibitor.

In premenopausal women, aromatase inhibitors are not very effective, as decreasing peripheral estrogen stimulates the ovaries to produce more estrogen. Thus, for these patients, adjuvant endocrine therapy consists of tamoxifen, with ovarian ablation (via surgery or radiation) or ovarian suppression with a GnRH analog. If the patient goes through menopause as a result of this therapy, she may benefit from aromatase inhibitors at that time.4,5

Women with breast cancer that overexpresses the HER2 gene benefit from adjuvant treatment with trastuzumab, an anti-HER2 antibody.6 While current guidelines advise treatment for 1 year, multiple studies are evaluating dosing schedules and optimal duration of treatment. for now, patients should be monitored for signs of cardiotoxicity at baseline and every 3 months thereafter until completion of therapy.4

References

1. Early Breast Caner Trialists’ Collaborative Group (EBCTCG). Effects of chemotherapy and hormonal therapy for early breast cancer on recurrence and 15-year survival: an overview of the randomised trials. Lancet. 2005;265:1687-1717.

2. Codeiro P. Breast reconstruction after surgery for breast cancer. N Engl J Med. 2008;359:1590-1601.

3. DeLaurentiis M, Cancello G, D’Agostino D, et al. Taxane-based combinations as adjuvant chemotherapy of early breast cancer: a meta-analysis of randomized trials. J Clin Oncol. 2008;26:44-53.

4. National Comprehensive Cancer Network. Breast cancer risk reduction clinical practice guidelines in oncology. J Natl Compr Canc Netw. 2003;1:280-296.

5. Baum M, Budzar AU, Cuzick J, et al. Anastrozole alone or in combination with tamoxifen versus tamoxifen alone for adjuvant treatment of postmenopausal women with early breast cancer: first results of the ATAC randomised trial. Lancet. 2002;359:2131-2139.

6. Piccart-Gebhart MJ, Procter M, Leyland-Jones B, et al. Herceptin adjuvant (HERA) Trial Study Team. Trastuzumab after adjuvant chemotherapy in HER2-positive breast cancer. N Engl J Med. 2005;353:1659-1672.

PRACTICE RECOMMENDATIONS

Offer screening magnetic resonance imaging (MRI) to patients with a known BRCA 1 or 2 mutation, a strong family history of breast cancer, or a lifetime risk of breast cancer >20% to 25%. B

For early-stage breast cancer, lumpectomy and sentinel node mapping with excision is the preferred method for staging. A

Monitor patients receiving tamoxifen for signs and symptoms of venous thromboembolism, cataracts, and uterine malignancy, and patients on aromatase inhibitors for the development of osteoporosis. A

Strength of recommendation (SOR)

A Good-quality patient-oriented evidence
B
Inconsistent or limited-quality patient-oriented evidence
C
Consensus, usual practice, opinion, disease-oriented evidence, case series

Late last year, the US Preventive Services Task Force (USPSTF) sparked a nationwide controversy when it announced that it was recommending against routine screening mammography for women younger than age 50.1 Indeed, that’s a recommendation that many other organizations, including the American Cancer Society (ACS),2 the American College of Obstetricians and Gynecologists (ACOG),3 and the National Comprehensive Cancer Network (NCCN),4 disagree with. But the age at which women should begin routine mammography isn’t the only controversial question. Experts disagree on the benefits of breast self-examination, the optimal frequency of clinical breast exams, and the use of digital mammography—among other issues. This evidence-based review can help you cut through the confusion.

CASE Carrie, a 39-year-old woman who has never been pregnant, comes in for an annual Pap smear and gynecologic exam. She has a negative past medical history, but a positive family history for breast cancer—both her mother and 1 of her sisters had the disease. How would you assess Carrie’s risk of breast cancer, and what preventive measures would you recommend?

Use this predictive model to pinpoint your patient’s risk

When making decisions regarding primary prevention of and screening for breast cancer, an accurate assessment of risk is critical. Many predictive models have been developed with that in mind. The most widely studied, the Gail model, incorporates a number of important risk factors (TABLE 1), including age; race; family history; reproductive factors such as age of menarche, menopause, and first childbirth; and previous history of breast biopsy and atypical findings, to calculate a woman’s 5-year risk.5

A risk calculator (the Breast Cancer Risk Assessment Tool) based on the Gail model is available on the National Cancer Institute’s Web site, at http://www.cancer.gov/bcrisktool. Generally, a score ≥1.66%,5 which indicates that a patient has at least a 1.66% chance of developing breast cancer over the next 5 years, is considered high risk.6,7

CASE Carrie’s 2 first-degree relatives affected by breast cancer and her nulliparous status place her at increased risk. Further questioning reveals a particularly strong family history, as both relatives were diagnosed before the age of 50 (her mom at 45 years of age and her sister, at 39). Carrie’s 5-year risk is 1.8%.

TABLE 1
Risk factors for breast cancer5,29

  • Age (>50 years)
  • Sex
  • Ethnicity*
  • Family history (≥1 first-degree relative diagnosed with breast cancer, particularly if diagnosed at <50 years of age)
  • Early menarche (<12 years)
  • Late menopause (>55 years)
  • Late age at first full-term pregnancy (>30 years)
  • Nulliparity
  • Breast density
  • History of breast biopsies
  • Atypical hyperplasia or LCIS on prior biopsy
  • History of radiation to chest wall
  • Lack of breastfeeding
  • Physical inactivity
  • Obesity
  • Alcohol use
  • Exogenous hormones (HRT)
*African American and Caucasian women are at higher risk compared with Asian, Hispanic, and Native American women.
1 drink/day results in minimal increase in risk; 2-5 drinks/day result in 1.5 increased risk compared with nondrinkers.
HRT, hormone replacement therapy; LCIS, lobular carcinoma in situ.

All women can benefit from these preventive measures

As primary care physicians, we have a responsibility to stress lifestyle modification as the mainstay of breast cancer prevention. Whether or not a woman is at high risk, advise her that maintaining a normal weight, exercising vigorously, limiting alcohol consumption, and breastfeeding are evidence-based methods of primary prevention. Diets low in fat and high in fiber may be associated with a lower risk of invasive breast cancer, but there is no conclusive evidence to support specific dietary interventions to reduce the risk.8-11 Nor has a link between active or passive smoking, antioxidants, or fruit and vegetable intake been firmly established.12

There is a clear association between prolonged estrogen exposure and breast cancer, however. Many reproductive factors, such as early menarche, late menopause, later age at time of first full-term pregnancy, and nulliparity, increase a woman’s exposure to endogenous estrogen—and her risk of developing breast cancer.12,13

Exposure to exogenous estrogen is also linked to the development of breast cancer. In 2002, the Women’s Health Initiative (WHI) was stopped early after a report was released stating that the risks of hormone replacement therapy (HRT)—a higher incidence of cardiovascular events, stroke, and venous thromboembolism, as well as breast cancer—outweighed the benefits.14 Subsequent analyses have found a relationship between the declining incidence of breast cancer and the marked decrease in HRT use prompted by the WHI report. While causality has not been firmly established, multiple studies strongly suggest it.15,16

 

 

The association between oral contraceptives (OCs) and breast cancer is more controversial. Some studies have found an increased breast cancer risk among OC users, but both the relative risk and absolute risk were found to be very small and to dissipate 10 years after stopping OC use. More recent studies with newer formulations containing lower doses of estrogen have failed to show an increased risk.8

Breast cancer screening: The parameters have changed

Various organizations have published guidelines for breast cancer screening (TABLE 2), and all are somewhat different. Here’s what you need to know.

Breast self-examination (BSE), which women were previously advised to perform monthly, has not been shown to improve mortality in any age group, and is no longer routinely recommended.17 While both the USPSTF and the Canadian Task Force on Preventive Health Care recommend against teaching women BSE, the ACS, ACOG, and NCCN encourage self-examination—particularly among women older than 40 years.1-4,17,18

Clinical breast examination has an average sensitivity of 50% and detects approximately 5% of mammographically occult cancers.19 It is still not clear whether clinical breast exams save lives, however—a finding that is reflected in the USPSTF’s “I” (insufficient evidence to assess the benefits and harms) recommendation.1 Other consensus guidelines still recommend clinical breast examination, albeit at varying frequencies.

Screening mammography decreases mortality rates by anywhere from 28% to 65%, depending on the statistical model used.20 The benefit is greatest in women between the ages of 50 and 69 years, however, and most groups agree that mammography every 1 to 2 years is advisable for this age group. (There is limited data on the value of mammography for women 70 years of age and older, and no consensus on the age at which to stop screening.1,21) But because the mortality benefit from screening mammography is lower for women aged 40 to 49, guidelines for this age group are more controversial.

Mammography’s sensitivity is affected by a variety of factors, including age and menopausal status, prior breast surgery or radiation, breast density, and the experience of the radiologist. Women in their 40s have denser breast tissue than older women, making mammography less sensitive for this age group. Because of that, and because the overall incidence of breast cancer is lower for women younger than 50, some argue that screening mammography for women between the ages of 40 and 49 years leads to unacceptably high false-positive rates (9.8% annually22) and that the harm associated with mammography may outweigh the benefit. Others counter that tumors in younger women tend to be more aggressive and faster growing, making early detection even more critical than for older women.

What should you advise women in this age group? You might point out that the USPSTF recommends against routine screening, but indicates that the decision to begin (or defer) routine mammography before age 50 should be individualized, based on the needs and values of each patient.1

Digital mammography. A recent study of more than 43,000 women demonstrated that digital mammography is more accurate than film—but only for certain groups: These include women <50 years of age, women with dense breasts, and pre- and perimenopausal women. 23 Because it is still not clear whether the increased accuracy will translate into a mortality benefit, more research is needed before digital mammography is widely adopted. The USPSTF maintains that there is insufficient evidence to assess the benefits and harms of using either digital mammography or magnetic resonance imaging (MRI) rather than film mammography to screen for breast cancer.1

MRI. In 2007, the ACS published guidelines on the use of MRI as an adjunct to mammography for breast cancer screening in high-risk women.24 According to ACS guidelines, screening MRI should be offered to patients with a known BRCA 1 or 2 mutation (5%-10% of all breast cancers are associated with a mutation in the BRCA 1 or BRCA 2 gene, which is transmitted in an autosomal dominant pattern6). It also should be offered to those with a strong family history, or a lifetime risk of developing breast cancer that is >20% to 25%. And finally, MRI should be offered to women who had chest wall radiation when they were between the ages of 10 and 30 years—another significant risk factor for breast cancer—and those with other genetic syndromes that increase their lifetime risk of breast cancer.24

 

 

Evidence is insufficient for or against MRI screening for women with a personal history of breast cancer, atypical hyperplasia, or lobular carcinoma in situ, however, and neither breast ultrasound (which is generally used diagnostically, not for screening purposes) nor MRI has been shown to be helpful as a screening tool in women with <15% lifetime risk of developing breast cancer.24,25

TABLE 2
Guidelines for breast cancer screening for women with average risk

OrganizationAge (years)Breast self-examClinical breast examMammography
American Cancer Society220-40OptionalEvery 3 yNA
>40EncouragesAnnuallyAnnually
American College of Obstetricians and Gynecologists340-49EncouragesAnnually1-2 y
50-69EncouragesAnnuallyAnnually
Canadian Task force on Preventive Health Care1840-49Recommends against teachingInsufficient evidenceInsufficient evidence
50-69Recommends against teaching1-2 y1-2 y
National Comprehensive Cancer Network420-40Encourages1-3 yNA
>40EncouragesAnnuallyAnnually
US Preventive Services Task Force140-49Recommends against teachingInsufficient evidenceNot routinely recommended
50-74Recommends against teachingInsufficient evidenceEvery 2 y
NA, not addressed.

When to consider chemoprevention

For women like Carrie, who are at high risk of developing breast cancer, selective estrogen receptor modulator (SERM) therapy and surgical interventions may be options to consider. The Breast Cancer Prevention Trial demonstrated the efficacy of tamoxifen as a preventive agent. This landmark trial showed that for high-risk women older than 35, 5 years of tamoxifen therapy can reduce the incidence of invasive breast cancer by nearly 50%.26

Women with the BRCA 1 or 2 mutation—all of whom should be offered genetic counseling—were included in the study. Tamoxifen reduced the incidence of breast cancer in BRCA 2 carriers by 62%, the researchers found, but did not reduce risk in carriers of the BRCA 1 gene. This is likely due to the high prevalence of estrogen receptor-negative breast cancers among BRCA 1 carriers.26

More recently, the Study of Tamoxifen and Raloxifene (STAR) trial compared the efficacy of tamoxifen and raloxifene, a second-generation SERM, in high-risk postmenopausal women ages 35 and older. The drugs were found to be equally effective in reducing the risk of invasive breast cancer, but raloxifene had a better side effect profile, with a lower incidence of thromboembolism and cataracts. 27

What the guidelines call for. In 2003, the USPSTF recommended that clinicians discuss chemoprevention with women at high risk for breast cancer and low risk for adverse effects of SERMs.28

The most recent update to the NCCN breast cancer risk reduction guidelines recommends that clinicians offer tamoxifen to premenopausal women with a 5-year projected breast cancer risk ≥1.7% and offer tamoxifen or raloxifene to high-risk postmenopausal women.29 It is worth noting, however, that SERMs can have significant adverse effects, including venous thromboembolism, stroke, cataracts, uterine malignancy, and hot flashes, while lifestyle modifications and the avoidance of HRT have few, if any, negative effects.

CASE After consultation with a genetic counselor, Carrie underwent testing for both the BRCA 1 and BRCA 2 mutations. She tested negative for both. She declined chemoprevention and prophylactic surgery, opting for enhanced screening with yearly mammography and MRI and lifestyle modification instead.

When a mass is found

For women ages 30 or older with palpable masses or solid masses ≥2 cm found on imaging, core needle biopsy is recommended.30,31 Biopsy is indicated for women younger than 30 as well, if the mass is >2 cm or imaging is suspicious. In general, a needle biopsy read as benign is considered adequate for diagnostic purposes only if the lesion appeared benign on imaging.

For lesions shown to be cystic on imaging, recommendations for follow-up or additional testing are based on the characteristics of the cyst. For simple cysts, 2- to 4-month follow up for stability, followed by routine screening, is adequate.21 Additional evaluation of complex cysts is indicated, including aspiration for complicated cysts and biopsy for complex cysts. After aspiration, surgical excision of bloody aspirates or persistent masses is recommended.30,31

Staging using the TNM system
The TNM (tumor, node, metastases) classification system is used for the staging of breast cancer:

  • T refers to the tumor type, size, and extent of local involvement
  • N describes regional lymph node involvement
  • M refers to distant metastases.

The TNM classifications are also grouped by stage (I through IV).,

Lumpectomy and sentinel node mapping with excision is the preferred method for staging of early-stage breast cancer without palpable lymphadenopathy—provided that the surgical team has documented experience with sentinel node biopsy.32 Sentinel node biopsy is preferred because of its safety, low (<10%) false negative rate, and decreased morbidity compared with full axillary dissection, although dissection is recommended for patients with more advanced cancer or a positive sentinel node.32 The comparative effects of sentinel node biopsy vs axillary node dissection on tumor recurrence and patient survival are not known.33

 

 

Testing for tumor markers such as estrogen and progesterone receptors and human epidermal growth factor receptor 2 (HER2) expression status in biopsy-proven breast cancer is now the standard of care. Seventy percent of breast cancers are estrogen receptor-positive, with increasing frequency associated with older age.34 Estrogen/progesterone receptor positivity is associated with a more favorable outcome, and multiple hormonal therapies can be aimed at these receptors.34 While HER2 overexpression—which occurs in 15% to 30% of newly diagnosed breast cancers35—is associated with more aggressive tumors, women with this type of tumor cell can benefit from trastuzumab, an anti-HER2 drug.36

Key factors that affect prognosis

Important factors affecting prognosis and treatment of localized breast cancer are tumor size, age and menopausal status, tumor expression of hormone receptors and/ or the HER2 protein, as well as the status of the draining axillary nodes. Factors that predict a greater chance of recurrence include the spread of disease to axillary nodes, larger tumor size, invasive histology, inflammatory pathology, lack of estrogen/progesterone receptors, and age <50 years or premenopausal status.

Treatment options include surgical resection, radiation, and systemic adjuvant therapy in the form of chemotherapy, endocrine therapy, or anti-HER2 monoclonal antibodies.37 (For more on treatment, see “Surgery, radiation, and systemic therapy: Making the most of what’s in our arsenal” at jfponline.com.)

Don’t overlook quality-of-life issues
Follow-up of breast cancer patients should go beyond treatment and work-up for recurrence and metastatic disease to focus on health and lifestyle issues, such as stress reduction, mood, smoking cessation, diet and exercise, treatment of hot flashes, sexual dysfunction, and bone health. A recent study found both reduced recurrence and increased survival in women receiving psychological interventions to improve quality-of-life measures after an 11-year follow-up.38

Refer women to targeted Web sites such as the National Breast Cancer Awareness Month organization (http://www.nbcam.org/), the National Breast Cancer Foundation (http://community.nationalbreastcancer.org/), and the Susan G. Komen Breast Cancer Foundation (http://ww5.komen.org/). Offer treatment for bothersome symptoms. Hot flashes and depression, for example, often related to endocrine therapy, can be treated with selective serotonin reuptake inhibitors (SSRIs). That said, some SSRIs decrease the active metabolite of tamoxifen by inhibiting CYP2D6 enzyme and must, therefore, be used with caution. However, venlafaxine and citalopram are less likely to alter tamoxifen metabolism than other SSRIs.39

CASE When Carrie was 47, she had an abnormal MRI of the left breast. Core needle biopsy and pathology of the lesion revealed an estrogen and progesterone receptor-positive tumor that was negative for HER2 overexpression. She underwent lumpectomy, which revealed a 1.5 cm tumor, followed by a negative sentinel node biopsy, and was diagnosed with stage I (T1N0M0) breast cancer. Carrie had radiation after surgery; she did not require chemotherapy, but was told to take tamoxifen for 5 years. This adjuvant endocrine therapy led to hot flashes and depression, both of which were successfully treated with venlafaxine. Carrie is currently cancer-free and participates in a breast cancer survivor program that includes regular visits with her primary physician and her oncologist.

CORRESPONDENCE Denise Sur, MD, 1920 Colorado Avenue, Santa Monica, CA 90404; [email protected] 

Online exclusive

Surgery, radiation, and systemic therapy: Making the most of what’s in our arsenal

Breast cancer surgery has changed dramatically over the years. Multiple studies have shown that breast-conserving therapy (lumpectomy followed by radiation) for carefully selected women is comparable to mastectomy for local recurrence and survival. While there has been much interest in determining whether a subset of patients could forego radiation after lumpectomy, a meta-analysis by the Early Breast Cancer Trialists Collaborative Group demonstrated that radiation after lumpectomy provides an absolute local recurrence risk reduction of 19%, and a 5.4% absolute reduction in 15-year breast cancer mortality rates compared with lumpectomy without radiation.1 Thus, radiation after lumpectomy remains the standard of care for all women undergoing breast-conserving therapy, regardless of tumor characteristics.

In certain women with a high risk of recurrence (≥4 positive nodes), radiation is also recommended after mastectomy. Women undergoing mastectomy have numerous options for immediate or delayed breast reconstruction. Consultation with a multidisciplinary team, including a plastic surgeon, prior to any surgical intervention is advised.2

Multiple systemic chemotherapy regimens have been shown to be beneficial in carefully selected patients with breast cancer. Systematic reviews have demonstrated that an anthracycline-based regimen can decrease annual breast cancer mortality by 38% in women <50 years old and by 20% in women ages 50 to 69 years.1 in more recent randomized controlled trials, the addition of taxanes to anthracycline-based regimens has produced promising results.3

Numerous hormonal therapies benefit women with estrogen or progesterone receptor-positive breast cancer. Tamoxifen blocks the activity of estrogen on receptors located in breast cancer tissue, for example; aromatase inhibitors block the conversion of androgens to estrogen; and gonadotropin-releasing hormone (GnRH) analogs such as leuprolide and goserelin suppress ovarian production of estrogen.

For postmenopausal women, options include an aromatase inhibitor alone or tamoxifen followed by an aromatase inhibitor.

In premenopausal women, aromatase inhibitors are not very effective, as decreasing peripheral estrogen stimulates the ovaries to produce more estrogen. Thus, for these patients, adjuvant endocrine therapy consists of tamoxifen, with ovarian ablation (via surgery or radiation) or ovarian suppression with a GnRH analog. If the patient goes through menopause as a result of this therapy, she may benefit from aromatase inhibitors at that time.4,5

Women with breast cancer that overexpresses the HER2 gene benefit from adjuvant treatment with trastuzumab, an anti-HER2 antibody.6 While current guidelines advise treatment for 1 year, multiple studies are evaluating dosing schedules and optimal duration of treatment. for now, patients should be monitored for signs of cardiotoxicity at baseline and every 3 months thereafter until completion of therapy.4

References

1. Early Breast Caner Trialists’ Collaborative Group (EBCTCG). Effects of chemotherapy and hormonal therapy for early breast cancer on recurrence and 15-year survival: an overview of the randomised trials. Lancet. 2005;265:1687-1717.

2. Codeiro P. Breast reconstruction after surgery for breast cancer. N Engl J Med. 2008;359:1590-1601.

3. DeLaurentiis M, Cancello G, D’Agostino D, et al. Taxane-based combinations as adjuvant chemotherapy of early breast cancer: a meta-analysis of randomized trials. J Clin Oncol. 2008;26:44-53.

4. National Comprehensive Cancer Network. Breast cancer risk reduction clinical practice guidelines in oncology. J Natl Compr Canc Netw. 2003;1:280-296.

5. Baum M, Budzar AU, Cuzick J, et al. Anastrozole alone or in combination with tamoxifen versus tamoxifen alone for adjuvant treatment of postmenopausal women with early breast cancer: first results of the ATAC randomised trial. Lancet. 2002;359:2131-2139.

6. Piccart-Gebhart MJ, Procter M, Leyland-Jones B, et al. Herceptin adjuvant (HERA) Trial Study Team. Trastuzumab after adjuvant chemotherapy in HER2-positive breast cancer. N Engl J Med. 2005;353:1659-1672.

References

1. US Preventive Services Task Force Screening for breast cancer. Available at: http://www.uspreventiveservicestaskforce.org/uspstf/uspsbrca.htm. Accessed September 2, 2010.

2. American Cancer Society. American Cancer Society guidelines for the early detection of cancer. Available at: http://www.cancer.org/Healthy/FindCancerEarly/CancerScreeningGuidelines/american-cancer-society-guidelines-for-the-early-detection-of-cancer. Accessed September 2, 2010.

3. American College of Obstetricians and Gynecologists. Response of the American College of Obstetricians and Gynecologists to the new breast cancer screening recommendations from the US Preventive Services Task Force. Available at: http://www.acog.org/from_home/misc/uspstfresponse.cfm. Accessed September 2, 2010.

4. National Comprehensive Cancer Network. Breast cancer screening and diagnosis. Clinical Practice Guidelines in Oncology-V.1.2010. Fort Washington, Pa: National Comprehensive Cancer Network: November 3, 2009. Available at: http://www.nccn.org/professionals/physician_gls/f_guidelines.asp. Accessed September 17 , 2010.

5. National Cancer Institute. Breast cancer risk assessment tool. Available at: http://www.cancer.gov/bcrisktool/. Accessed September 2, 2010

6. Robson M, Offit K. Clinical practice. Management of an inherited predisposition to breast cancer. N Engl J Med. 2007;357:154-162.

7. Fisher B, Costantino JP, Wickerham DL, et al. Tamoxifen for prevention of breast cancer: report of the National Surgical Adjuvant Breast and Bowel Project P-1 Study. J Natl Cancer Inst. 1998;90:1371-1388.

8. Cummings SR, Tice JA, Bauer S, et al. Prevention of breast cancer in postmenopausal women: approaches to estimating and reducing risk. J Natl Cancer Inst. 2009;18:101,-384-398.

9. Prentice RL, Caan B, Chlebowski RT, et al. Low-fat dietary pattern and risk of invasive breast cancer: the Women’s Health Initiative Randomized Controlled Dietary Modification Trial. JAMA. 2006;295:629-642.

10. Pierce JP, Natarajan L, Caan BJ, et al. Influence of a diet very high in vegetables, fruit, and fiber and low in fat on prognosis following treatment for breast cancer: the Women’s Healthy Eating and Living (WHEL) randomized trial. JAMA. 2007;298:289-298.

11. Chlebowski RT, Blackburn GL, Thomson CA, et al. Dietary fat reduction and breast cancer outcome: interim efficacy results from the Women’s Intervention Nutrition Study (WINS). J Natl Cancer Inst. 2006;98:1767-1776.

12. PDQ Cancer Information Summary. Breast Cancer Prevention (PRQ) – Health Professional. Date last modified April 30, 2009. Available at: http://www.cancer.gov/cancertopics/pdq/prevention/breast/healthprofessional. Accessed May 12, 2009.

13. Hulka BS, Moorman PG. Breast cancer: hormones and other risk factors. Maturitas. 2001;38:103-113.

14. Rossouw JE, Anderson GL, Prentice RL, et al. Risks and benefits of estrogen plus progestin in healthy postmenopausal women: principal results From the Women’s Health Initiative randomized controlled trial. JAMA. 2002;288:321-333.

15. Ravdin PM, Cronin KA, Howlader N, et al. The decrease in breast-cancer incidence in 2003 in the United States. N Engl J Med. 2007;356:1670-1674.

16. Chlebowski RT, Kuller LH, Prentice RL, et al. WHI Investigators. Breast cancer after use of estrogen plus progestin in postmenopausal women. N Engl J Med. 2009;360:573-587.

17. Kosters JP, Gotzsche PC. Regular self-examination or clinical examination for early detection of breast cancer. Cochrane Database Syst Rev. 2003;(2):CD003373.-

18. Canadian Task Force on the Periodic Health Examination Ottawa, Canada: Health Canada; 1994:788-795 (reaffirmed by the Canadian Task Force on the Periodic Health Examination 1999, 2001).Available at: http://www.ctfphc.org/index/html. Accessed August 12, 2009.

19. Green BB, Taplin SH. Breast cancer screening controversies. J Am Board Fam Pract. 2003;16:233-241.

20. Berry DA, Cronin KA, Plevritis SK, et al. Effect of screening and adjuvant therapy on mortality from breast cancer. N Engl J Med. 2005;353:1784-1792.

21. Tice JA, Kerlikowske K. Screening and prevention of breast cancer in primary care. Prim Care. 2009;36:533-558.

22. US Preventive Services Task Force. Screening for breast cancer. Systematic evidence review update for the US Preventive Services Task Force. Available at: http://www.ncbi.nlm.nih.gov/bookshelf/br.fcgi?book=es74. Accessed September 2, 2010.

23. Pisano ED, Gatsonis C, Hendrick E, et al. Digital Mammographic Imaging Screening Trial (DMIST) Investigators Group. Diagnostic performance of digital versus film mammography for breast-cancer screening. N Engl J Med. 2005;353:1773-1783.

24. Saslow D, Boetes C, Burke W, et al. American Cancer Society Breast Cancer Advisory Group. American Cancer Society guidelines for breast screening with MRI as an adjunct to mammography. CA Cancer J Clin. 2007;57:75-89.

25. Berg W, Blume J, Cormack J, et al. Combined screening with ultrasound and mammography vs mammography alone in women at elevated risk of breast cancer. JAMA. 2008;299:2151-2163.

26. King M, Wieand S, Hale K, et al. Tamoxifen and breast cancer incidence among women with inherited mutations in BRCA1 and BRCA2: National Surgical Adjuvant Breast and Bowel Project (NSABP-P1) Breast Cancer Prevention Trial. JAMA. 2001;286:2251-2256.

27. Vogel VG, Costantino JP, Wickerham DL, et al. Effects of tamoxifen vs raloxifene on the risk of developing invasive breast cancer and other disease outcomes: the NSABP Study of Tamoxifen and Raloxifene (STAR) P-2 trial. JAMA. 2006;295:2727-2741.

28. Berg AO. US Preventive Services Task Force. Chemoprevention of breast cancer: recommendations and rationale. Am J Nurs. 2003;103:107-113.

29. National Comprehensive Cancer Network. Breast cancer risk reduction. Clinical Practice Guidelines in Oncology-V.2.2010. Fort Washington, Pa: National Comprehensive Cancer Network; August 7, 2010. Available at: http://www.nccn.org/professionals/physician_gls/f_guidelines.asp. Accessed September 17, 2010.

30. Sickles EA, Filly RA, Cllen PW. Benign breast lesions: ultrasound detection and diagnosis. Radiology. 1984;151:467.-

31. Beavers TB, Anderson BO, Bonaccio E, et al. NCCN clinical practice guidelines in oncology: breast cancer screening and diagnosis. J Natl Compr Canc Netw. 2009;7:1060-1096.

32. Mansel RE, Fallowfield L, Kissin M, et al. Randomized multicenter trial of sentinel node biopsy versus standard axillary treatment in operable breast cancer: the ALMANAC trial. J Natl Canc Inst. 2006;98:599-609

33. Lyman GH, Guiliano AE, Somerfield MR, et al. American Society of Clinical Oncology guideline recommendations for sentinel lymph node biopsy in early- stage breast cancer. J Clin Oncol. 2005;23:7703-7720.

34. Turner N, Jones A. Management of breast cancer-Part II. BMJ. 2008;337:a540.-

35. Harris L, Fritsche H, Mennel R, et al:. American Society of Clinical Oncology 2007 update of recommendations for the use of tumor markers in breast cancer J Clin Oncol. 2007;25:5287-5312.

36. Piccart-Gebhart MJ, Procter M, Leyland-Jones B, et al. Herceptin adjuvant (HERA) Trial Study Team. Trastuzumab after adjuvant chemotherapy in HER2-positive breast cancer. N Engl J Med. 2005;353:1659-1672.

37. PDQ Cancer Information Summary Breast cancer treatment– health professional. Available at: http://www.cancer.gov/cancertopics/pdq/treatment/breast/HealthProfessional. Accessed May 12, 2009.

38. Anderson BL, Yang HC, Farrar WB, et al. Psychologic intervention improves survival for breast cancer patients: a randomized clinical trial. Cancer. 2008;113:3450-3458.

39. Jin Y, Desta Z, Stearns V, et al. CYP2D6 genotype, antidepressant use, and tamoxifen metabolism during adjuvant breast cancer treatment. J Natl Cancer Inst. 2005;97:30-39.

References

1. US Preventive Services Task Force Screening for breast cancer. Available at: http://www.uspreventiveservicestaskforce.org/uspstf/uspsbrca.htm. Accessed September 2, 2010.

2. American Cancer Society. American Cancer Society guidelines for the early detection of cancer. Available at: http://www.cancer.org/Healthy/FindCancerEarly/CancerScreeningGuidelines/american-cancer-society-guidelines-for-the-early-detection-of-cancer. Accessed September 2, 2010.

3. American College of Obstetricians and Gynecologists. Response of the American College of Obstetricians and Gynecologists to the new breast cancer screening recommendations from the US Preventive Services Task Force. Available at: http://www.acog.org/from_home/misc/uspstfresponse.cfm. Accessed September 2, 2010.

4. National Comprehensive Cancer Network. Breast cancer screening and diagnosis. Clinical Practice Guidelines in Oncology-V.1.2010. Fort Washington, Pa: National Comprehensive Cancer Network: November 3, 2009. Available at: http://www.nccn.org/professionals/physician_gls/f_guidelines.asp. Accessed September 17 , 2010.

5. National Cancer Institute. Breast cancer risk assessment tool. Available at: http://www.cancer.gov/bcrisktool/. Accessed September 2, 2010

6. Robson M, Offit K. Clinical practice. Management of an inherited predisposition to breast cancer. N Engl J Med. 2007;357:154-162.

7. Fisher B, Costantino JP, Wickerham DL, et al. Tamoxifen for prevention of breast cancer: report of the National Surgical Adjuvant Breast and Bowel Project P-1 Study. J Natl Cancer Inst. 1998;90:1371-1388.

8. Cummings SR, Tice JA, Bauer S, et al. Prevention of breast cancer in postmenopausal women: approaches to estimating and reducing risk. J Natl Cancer Inst. 2009;18:101,-384-398.

9. Prentice RL, Caan B, Chlebowski RT, et al. Low-fat dietary pattern and risk of invasive breast cancer: the Women’s Health Initiative Randomized Controlled Dietary Modification Trial. JAMA. 2006;295:629-642.

10. Pierce JP, Natarajan L, Caan BJ, et al. Influence of a diet very high in vegetables, fruit, and fiber and low in fat on prognosis following treatment for breast cancer: the Women’s Healthy Eating and Living (WHEL) randomized trial. JAMA. 2007;298:289-298.

11. Chlebowski RT, Blackburn GL, Thomson CA, et al. Dietary fat reduction and breast cancer outcome: interim efficacy results from the Women’s Intervention Nutrition Study (WINS). J Natl Cancer Inst. 2006;98:1767-1776.

12. PDQ Cancer Information Summary. Breast Cancer Prevention (PRQ) – Health Professional. Date last modified April 30, 2009. Available at: http://www.cancer.gov/cancertopics/pdq/prevention/breast/healthprofessional. Accessed May 12, 2009.

13. Hulka BS, Moorman PG. Breast cancer: hormones and other risk factors. Maturitas. 2001;38:103-113.

14. Rossouw JE, Anderson GL, Prentice RL, et al. Risks and benefits of estrogen plus progestin in healthy postmenopausal women: principal results From the Women’s Health Initiative randomized controlled trial. JAMA. 2002;288:321-333.

15. Ravdin PM, Cronin KA, Howlader N, et al. The decrease in breast-cancer incidence in 2003 in the United States. N Engl J Med. 2007;356:1670-1674.

16. Chlebowski RT, Kuller LH, Prentice RL, et al. WHI Investigators. Breast cancer after use of estrogen plus progestin in postmenopausal women. N Engl J Med. 2009;360:573-587.

17. Kosters JP, Gotzsche PC. Regular self-examination or clinical examination for early detection of breast cancer. Cochrane Database Syst Rev. 2003;(2):CD003373.-

18. Canadian Task Force on the Periodic Health Examination Ottawa, Canada: Health Canada; 1994:788-795 (reaffirmed by the Canadian Task Force on the Periodic Health Examination 1999, 2001).Available at: http://www.ctfphc.org/index/html. Accessed August 12, 2009.

19. Green BB, Taplin SH. Breast cancer screening controversies. J Am Board Fam Pract. 2003;16:233-241.

20. Berry DA, Cronin KA, Plevritis SK, et al. Effect of screening and adjuvant therapy on mortality from breast cancer. N Engl J Med. 2005;353:1784-1792.

21. Tice JA, Kerlikowske K. Screening and prevention of breast cancer in primary care. Prim Care. 2009;36:533-558.

22. US Preventive Services Task Force. Screening for breast cancer. Systematic evidence review update for the US Preventive Services Task Force. Available at: http://www.ncbi.nlm.nih.gov/bookshelf/br.fcgi?book=es74. Accessed September 2, 2010.

23. Pisano ED, Gatsonis C, Hendrick E, et al. Digital Mammographic Imaging Screening Trial (DMIST) Investigators Group. Diagnostic performance of digital versus film mammography for breast-cancer screening. N Engl J Med. 2005;353:1773-1783.

24. Saslow D, Boetes C, Burke W, et al. American Cancer Society Breast Cancer Advisory Group. American Cancer Society guidelines for breast screening with MRI as an adjunct to mammography. CA Cancer J Clin. 2007;57:75-89.

25. Berg W, Blume J, Cormack J, et al. Combined screening with ultrasound and mammography vs mammography alone in women at elevated risk of breast cancer. JAMA. 2008;299:2151-2163.

26. King M, Wieand S, Hale K, et al. Tamoxifen and breast cancer incidence among women with inherited mutations in BRCA1 and BRCA2: National Surgical Adjuvant Breast and Bowel Project (NSABP-P1) Breast Cancer Prevention Trial. JAMA. 2001;286:2251-2256.

27. Vogel VG, Costantino JP, Wickerham DL, et al. Effects of tamoxifen vs raloxifene on the risk of developing invasive breast cancer and other disease outcomes: the NSABP Study of Tamoxifen and Raloxifene (STAR) P-2 trial. JAMA. 2006;295:2727-2741.

28. Berg AO. US Preventive Services Task Force. Chemoprevention of breast cancer: recommendations and rationale. Am J Nurs. 2003;103:107-113.

29. National Comprehensive Cancer Network. Breast cancer risk reduction. Clinical Practice Guidelines in Oncology-V.2.2010. Fort Washington, Pa: National Comprehensive Cancer Network; August 7, 2010. Available at: http://www.nccn.org/professionals/physician_gls/f_guidelines.asp. Accessed September 17, 2010.

30. Sickles EA, Filly RA, Cllen PW. Benign breast lesions: ultrasound detection and diagnosis. Radiology. 1984;151:467.-

31. Beavers TB, Anderson BO, Bonaccio E, et al. NCCN clinical practice guidelines in oncology: breast cancer screening and diagnosis. J Natl Compr Canc Netw. 2009;7:1060-1096.

32. Mansel RE, Fallowfield L, Kissin M, et al. Randomized multicenter trial of sentinel node biopsy versus standard axillary treatment in operable breast cancer: the ALMANAC trial. J Natl Canc Inst. 2006;98:599-609

33. Lyman GH, Guiliano AE, Somerfield MR, et al. American Society of Clinical Oncology guideline recommendations for sentinel lymph node biopsy in early- stage breast cancer. J Clin Oncol. 2005;23:7703-7720.

34. Turner N, Jones A. Management of breast cancer-Part II. BMJ. 2008;337:a540.-

35. Harris L, Fritsche H, Mennel R, et al:. American Society of Clinical Oncology 2007 update of recommendations for the use of tumor markers in breast cancer J Clin Oncol. 2007;25:5287-5312.

36. Piccart-Gebhart MJ, Procter M, Leyland-Jones B, et al. Herceptin adjuvant (HERA) Trial Study Team. Trastuzumab after adjuvant chemotherapy in HER2-positive breast cancer. N Engl J Med. 2005;353:1659-1672.

37. PDQ Cancer Information Summary Breast cancer treatment– health professional. Available at: http://www.cancer.gov/cancertopics/pdq/treatment/breast/HealthProfessional. Accessed May 12, 2009.

38. Anderson BL, Yang HC, Farrar WB, et al. Psychologic intervention improves survival for breast cancer patients: a randomized clinical trial. Cancer. 2008;113:3450-3458.

39. Jin Y, Desta Z, Stearns V, et al. CYP2D6 genotype, antidepressant use, and tamoxifen metabolism during adjuvant breast cancer treatment. J Natl Cancer Inst. 2005;97:30-39.

Issue
The Journal of Family Practice - 59(10)
Issue
The Journal of Family Practice - 59(10)
Page Number
575-581a
Page Number
575-581a
Publications
Publications
Topics
Article Type
Display Headline
Breast cancer prevention and Tx: An evidence-based guide
Display Headline
Breast cancer prevention and Tx: An evidence-based guide
Legacy Keywords
Denise Sur; breast cancer prevention; screening mammography; controversy; breast health
Legacy Keywords
Denise Sur; breast cancer prevention; screening mammography; controversy; breast health
Sections
Article Source

PURLs Copyright

Inside the Article

Article PDF Media