User login
Take these steps to improve your flu season preparedness
Last year’s influenza season was severe enough that hospitals around the United States set up special evaluation areas beyond their emergency departments, at times spilling over to tents or other temporary structures in what otherwise would be parking lots. The scale and potential severity of the annual epidemic can be difficult to convey to our patients, who sometimes say “just the flu” to refer to an illness responsible for more than 170 pediatric deaths in the United States this past year.1 The Centers for Disease Control and Prevention (CDC) recently updated its 5-year estimates of influenza-related deaths in the United States; influenza mortality ranges from about 12,000 deaths in a mild season (such as 2011-2012) to 56,000 in a more severe season (eg, 2012-2013).2
Although influenza cannot be completely prevented, the following strategies can help reduce the risk for the illness and limit its severity if contracted.
Prevention
Strategy 1: Vaccinate against influenza
While the efficacy of vaccines varies from year to year, vaccination remains the core of influenza prevention efforts. In this decade, vaccine effectiveness has ranged from 19% to 60%.3 However, models suggest that even when the vaccine is only 20% effective, vaccinating 140 million people (the average number of doses delivered annually in the United States over the past 5 seasons) prevents 21 million infections, 130,000 hospitalizations, and more than 61,000 deaths.4 In a case-control study, Flannery et al found that vaccination was 65% effective in preventing laboratory-confirmed influenza-associated death in children over 4 seasons (July 2010 through June 2014).5
Deciding who should be vaccinated is simpler than in prior years: Rather than targeting people who are at higher risk (those ages 65 and older, or those with comorbidities), the current CDC recommendation is to vaccinate nearly everyone ages 6 months or older, with limited exceptions.6,7 (See Table 18).
Formulations. Many types of influenza vaccine are approved for use in the United States; these differ in the number of strains included (3 or 4), the amount of antigen present for each strain, the presence of an adjuvant, the growth medium used for the virus, and the route of administration (see Table 29). The relative merits of each type are a matter of some debate. There is ongoing research into the comparative efficacy of vaccines comprised of egg- vs cell-based cultures, as well as studies comparing high-dose or adjuvant vaccines to standard-dose inactivated vaccines.
Previously, the CDC has recommended preferential use (or avoidance) of some vaccine types, based on their efficacy. For the 2018-2019 flu season, however, the CDC has rescinded its recommendation against vaccine containing live attenuated virus (LAIV; FluMist brand) and expresses no preference for any vaccine formulation for patients of appropriate age and health status.10 The American Academy of Pediatrics (AAP), however, is recommends that LAIV be used only if patients and their families decline injectable vaccines.11
Timing. Influenza vaccines are now distributed as early as July to some locations, raising concerns about waning immunity from early vaccination (July/August) if the influenza season does not peak until February or March.8,12,13 Currently, the CDC recommends balancing the possible benefit of delayed vaccination against the risks of missed opportunities to vaccinate, a possible early season, and logistical problems related to vaccinating the same number of people in a smaller time interval. Offering vaccination by the end of October, if possible, is recommended in order for immunity to develop by mid-November.8 Note: Children ages 6 months to 8 years will need to receive their initial vaccination in 2 half-doses administered at least 28 days apart; completing their vaccination by the end of October would require starting the process weeks earlier.
[polldaddy:10124269]
Continue to: Strategy 2
Strategy 2: Make use of chemoprophylaxis
Preventive use of antiviral medication (chemoprophylaxis) may be a useful adjunct or alternative to vaccination in certain circumstances: if the patient is at high risk for complications, has been exposed to someone with influenza, has contraindications to vaccination, or received the vaccine within the past 2 weeks. The CDC also suggests that chemoprophylaxis be considered for those with immune deficiencies or who are otherwise immunosuppressed after exposure.14 Antivirals can also be used to control outbreaks in long-term care facilities; in these cases, the recommendedregimen is daily administration for at least 2 weeks, continuing until at least 7 days after the identification of the last case.14 Oseltamivir (Tamiflu) and zanamivir (Relenza) are the recommended prophylactic agents; a related intravenous medication, peramivir (Rapivab), is recommend for treatment only (see Table 314).
Strategy 3: Prevent comorbidities and opportunistic infections
Morbidity associated with influenza often comes from secondary infection. Pneumonia is among the most common complications, so influenza season is a good time to ensure that patients are appropriately vaccinated against pneumococcus, as well. Pneumococcal conjugate vaccine (Prevnar or PCV13) is recommended for children younger than 2 years of age, to be administered in a series of 4 doses: at 2, 4, 6, and 12-15 months. Vaccination with PCV13 is also recommended for those ages 65 or older, to be followed at least one year later with pneumococcal polysaccharide vaccine (Pneumovax or PPSV23).15 Additional doses of PCV13, PPSV23, or both may be indicated, depending on health status.
Strategy 4: Encourage good hygiene
The availability of immunizations and antivirals does not replace good hygiene. Frequent handwashing reduces the transmission of respiratory viruses, including influenza.16 Few studies have evaluated the use of alcohol-based hand sanitizers, but available evidence suggests they are effective in lowering viral transmission.16
Barriers, such as masks, gloves, and gowns, are helpful for health care workers.16 Surgical masks are often considered more comfortable to wear than N95 respirators. It may therefore be welcome news that when a 2009 randomized study assessed their use by hospital-based nurses, masks were non-inferior in protecting these health care workers against influenza.17
Presenteeism, the practice of going to work while sick, should be discouraged. People at risk for influenza may wish to avoid crowds during flu season; those with symptoms should be encouraged to stay home and limit contact with others.
Continue to: Treatment
Treatment
Strategy 1: Make prompt use of antivirals
Despite available preventive measures, tens of millions of people in the United States develop influenza every year. Use of antiviral medication, begun early in the course of illness, can reduce the duration of symptoms and may reduce the risk for complications.
The neuraminidase inhibitor (NI) group of antivirals—oseltamivir, zanamivir, and peramivir—is effective against influenza types A and B and current resistance rates are low.
The adamantine family of antivirals, amantadine and rimantadine, treat type A only. Since the circulating influenza strains in the past several seasons have demonstrated resistance >99%, these medications are not currently recommended.14
NIs reduce the duration of influenza symptoms by 10% to 20%, shortening the illness by 6 to 24 hours.18,19 In otherwise healthy patients, this benefit must be balanced against the increased risk for nausea and vomiting (oseltamivir), bronchospasm and sinusitis (zanamivir), and diarrhea (peramivir). In adults, NIs reduce the risk for lower respiratory tract complications and hospitalization. A 2015 meta-analysis by Dobson et al found a relative risk for hospitalization among those prescribed oseltamivir vs placebo of 37%.18
In the past, antivirals were used only in high-risk patients, such as children younger than 2 years, adults older than 65 years, and those with chronic health conditions.14 Now, antivirals are recommended for those who are at higher risk for complications (see Table 4), those with “severe, complicated, or progressive illness,” and hospitalized patients.14
Continue to: Antiviral treatment may have some value...
Antiviral treatment may have some value for hospitalized patients when started even 5 days after symptom onset. Treatment may be extended beyond the usual recommendations (5 days for oseltamivir or zanamivir) in immunosuppressed patients or the critically ill. Additionally, recent guidelines include consideration of antiviral treatment in outpatients who are at normal risk if treatment can be started within 48 hours of symptom onset.14
The CDC currently recommends use of oseltamivir rather than other antivirals for most hospitalized patients, based on the availability of data on its use in this setting.14 Intravenous peramivir is recommended for patients who cannot tolerate or absorb oral medication; inhaled zanamivir or IV peramivir are preferred for patients with end-stage renal disease who are not undergoing dialysis (see Table 3).14
Strategy 2: Exercise caution when it comes to supportive care
There are other medications that may offer symptom relief or prevent complications, especially when antivirals are contraindicated or unavailable.
Corticosteroids are recommended as part of the treatment of community-acquired pneumonia,20 but their role in influenza is controversial. A 2016 Cochrane review21 found no randomized controlled trials on the topic. Although the balance of available data from observational studies indicated that use of corticosteroids was associated with increased mortality, the authors also noted that all the studies included in their meta-analysis were of “very low quality.” They concluded that “the use of steroids in influenza remains a clinical judgement call.”
Statins may be associated with improved outcomes in influenza and pneumonia. Studies thus far have given contradictory results,22,23 and a planned Cochrane review of the question has been withdrawn.24
Continue to: Over-the-counter medications...
Over-the-counter medications, such as aspirin, acetaminophen, and ibuprofen are often used to manage the fever and myalgia associated with influenza. Patients should be cautioned against using the same ingredient in multiple different branded medications. Acetaminophen, for example, is not limited to Tylenol-branded products. To avoid Reye’s syndrome, children and teens with febrile illness, such as influenza, should not use aspirin.
CORRESPONDENCE
Jennifer L. Hamilton, MD, PhD, Drexel Family Medicine, 10 Shurs Lane, Suite 301, Philadelphia, PA 19127; [email protected].
1. CDC. Weekly US influenza surveillance report. https://www.cdc.gov/flu/weekly/index.htm. Published June 8, 2018. Accessed August 22, 2018.
2. CDC. Estimated influenza illnesses, medical visits, hospitalizations, and deaths averted by vaccination in the United States. Published April 19, 2017. https://www.cdc.gov/flu/about/disease/2015-16.htm. Accessed Setptember 18, 2018.
3. CDC. Seasonal influenza vaccine effectiveness, 2005-2018. https://www.cdc.gov/flu/professionals/vaccination/effectiveness-studies.htm. Published February 15, 2018. Accessed August 22, 2018.
4. Sah P, Medlock J, Fitzpatrick MC, et al. Optimizing the impact of low-efficacy influenza vaccines. Proc Natl Acad Sci. 2018:201802479.
5. Flannery B, Reynolds SB, Blanton L, et al. Influenza vaccine effectiveness against pediatric deaths: 2010-2014. Pediatrics. 2017;139: e20164244.
6. Kim DK, Riley LE, Hunter P. Advisory Committee on Immunization Practices recommended immunization schedule for adults aged 19 years or older—United States, 2018. MMWR Morb Mortal Wkly Rep. 2018;67:158–160.
7. Robinson CL, Romero JR, Kempe A, et al. Advisory Committee on Immunization Practices recommended immunization schedule for children and adolescents aged 18 years or younger—United States, 2018. MMWR Morb Mortal Wkly Rep. 2018;67:156–157.
8. Grohskopf LA, Sokolow LZ, Broder KR, et al. Prevention and control of seasonal influenza with vaccines: recommendations of the Advisory Committee on Immunization Practices—United States, 2017-18 influenza season. MMWR Recomm Rep. 2017;66:1-20.
9. CDC. Influenza vaccines—United States, 2017–18 influenza season. https://www.cdc.gov/flu/protect/vaccine/vaccines.htm. Published May 16, 2018. Accessed August 22, 2018.
10. Grohskopf LA, Sokolow LZ, Fry AM, et al. Update: ACIP recommendations for the use of quadrivalent live attenuated influenza vaccine (LAIV4)—United States, 2018-19 influenza season. MMWR Morb Mortal Wkly Rep. 2018;67:643–645.
11. Jenco M. AAP: Give children IIV flu shot; use LAIV as last resort. AAP News. May 21, 2018. http://www.aappublications.org/news/2018/05/21/fluvaccine051818. Accessed August 22, 2018.
12. Glinka ER, Smith DM, Johns ST. Timing matters—influenza vaccination to HIV-infected patients. HIV Med. 2016;17:601-604.
13. Castilla J, Martínez-Baz I, Martínez-Artola V, et al. Decline in influenza vaccine effectiveness with time after vaccination, Navarre, Spain, season 2011/12. Euro Surveill. 2013;18:20388.
14. CDC. Influenza antiviral medications: summary for clinicians. https://www.cdc.gov/flu/professionals/antivirals/summary-clinicians.htm. Published May 11, 2018. Accessed August 22, 2018.
15. CDC. Pneumococcal vaccination summary: who and when to vaccinate. https://www.cdc.gov/vaccines/vpd/pneumo/hcp/who-when-to-vaccinate.html. Published February 28, 2018. Accessed August 22, 2018.
16. Jefferson T, Del Mar CB, Dooley L, et al. Physical interventions to interrupt or reduce the spread of respiratory viruses. Cochrane Database Syst Rev. 2011;(7):CD006207.
17. Loeb M, Dafoe N, Mahony J, et al. Surgical mask vs N95 respirator for preventing influenza among health care workers: a randomized trial. JAMA. 2009;302:1865-1871.
18. Dobson J, Whitley RJ, Pocock S, Monto AS. Oseltamivir treatment for influenza in adults: a meta-analysis of randomised controlled trials. Lancet. 2015;385:1729-1737.
19. Ghebrehewet S, MacPherson P, Ho A. Influenza. BMJ. 2016;355:i6258.
20. Kaysin A, Viera AJ. Community-acquired pneumonia in adults: diagnosis and management. Am Fam Physician. 2016;94:698-706.
21. Rodrigo C, Leonardi‐Bee J, Nguyen‐Van‐Tam J, et al. Corticosteroids as adjunctive therapy in the treatment of influenza. Cochrane Database Syst Rev. 2016;3:CD010406.
22. Brassard P, Wu JW, Ernst P, et al. The effect of statins on influenza-like illness morbidity and mortality. Pharmacoepidemiol Drug Saf. 2017;26:63-70.
23. Fedson DS. Treating influenza with statins and other immunomodulatory agents. Antiviral Res. 2013;99:417-435.
24. Khandaker G, Rashid H, Chow MY, et al. Statins for influenza and pneumonia. Cochrane Database Syst Rev. January 9, 2017 [withdrawn].
Last year’s influenza season was severe enough that hospitals around the United States set up special evaluation areas beyond their emergency departments, at times spilling over to tents or other temporary structures in what otherwise would be parking lots. The scale and potential severity of the annual epidemic can be difficult to convey to our patients, who sometimes say “just the flu” to refer to an illness responsible for more than 170 pediatric deaths in the United States this past year.1 The Centers for Disease Control and Prevention (CDC) recently updated its 5-year estimates of influenza-related deaths in the United States; influenza mortality ranges from about 12,000 deaths in a mild season (such as 2011-2012) to 56,000 in a more severe season (eg, 2012-2013).2
Although influenza cannot be completely prevented, the following strategies can help reduce the risk for the illness and limit its severity if contracted.
Prevention
Strategy 1: Vaccinate against influenza
While the efficacy of vaccines varies from year to year, vaccination remains the core of influenza prevention efforts. In this decade, vaccine effectiveness has ranged from 19% to 60%.3 However, models suggest that even when the vaccine is only 20% effective, vaccinating 140 million people (the average number of doses delivered annually in the United States over the past 5 seasons) prevents 21 million infections, 130,000 hospitalizations, and more than 61,000 deaths.4 In a case-control study, Flannery et al found that vaccination was 65% effective in preventing laboratory-confirmed influenza-associated death in children over 4 seasons (July 2010 through June 2014).5
Deciding who should be vaccinated is simpler than in prior years: Rather than targeting people who are at higher risk (those ages 65 and older, or those with comorbidities), the current CDC recommendation is to vaccinate nearly everyone ages 6 months or older, with limited exceptions.6,7 (See Table 18).
Formulations. Many types of influenza vaccine are approved for use in the United States; these differ in the number of strains included (3 or 4), the amount of antigen present for each strain, the presence of an adjuvant, the growth medium used for the virus, and the route of administration (see Table 29). The relative merits of each type are a matter of some debate. There is ongoing research into the comparative efficacy of vaccines comprised of egg- vs cell-based cultures, as well as studies comparing high-dose or adjuvant vaccines to standard-dose inactivated vaccines.
Previously, the CDC has recommended preferential use (or avoidance) of some vaccine types, based on their efficacy. For the 2018-2019 flu season, however, the CDC has rescinded its recommendation against vaccine containing live attenuated virus (LAIV; FluMist brand) and expresses no preference for any vaccine formulation for patients of appropriate age and health status.10 The American Academy of Pediatrics (AAP), however, is recommends that LAIV be used only if patients and their families decline injectable vaccines.11
Timing. Influenza vaccines are now distributed as early as July to some locations, raising concerns about waning immunity from early vaccination (July/August) if the influenza season does not peak until February or March.8,12,13 Currently, the CDC recommends balancing the possible benefit of delayed vaccination against the risks of missed opportunities to vaccinate, a possible early season, and logistical problems related to vaccinating the same number of people in a smaller time interval. Offering vaccination by the end of October, if possible, is recommended in order for immunity to develop by mid-November.8 Note: Children ages 6 months to 8 years will need to receive their initial vaccination in 2 half-doses administered at least 28 days apart; completing their vaccination by the end of October would require starting the process weeks earlier.
[polldaddy:10124269]
Continue to: Strategy 2
Strategy 2: Make use of chemoprophylaxis
Preventive use of antiviral medication (chemoprophylaxis) may be a useful adjunct or alternative to vaccination in certain circumstances: if the patient is at high risk for complications, has been exposed to someone with influenza, has contraindications to vaccination, or received the vaccine within the past 2 weeks. The CDC also suggests that chemoprophylaxis be considered for those with immune deficiencies or who are otherwise immunosuppressed after exposure.14 Antivirals can also be used to control outbreaks in long-term care facilities; in these cases, the recommendedregimen is daily administration for at least 2 weeks, continuing until at least 7 days after the identification of the last case.14 Oseltamivir (Tamiflu) and zanamivir (Relenza) are the recommended prophylactic agents; a related intravenous medication, peramivir (Rapivab), is recommend for treatment only (see Table 314).
Strategy 3: Prevent comorbidities and opportunistic infections
Morbidity associated with influenza often comes from secondary infection. Pneumonia is among the most common complications, so influenza season is a good time to ensure that patients are appropriately vaccinated against pneumococcus, as well. Pneumococcal conjugate vaccine (Prevnar or PCV13) is recommended for children younger than 2 years of age, to be administered in a series of 4 doses: at 2, 4, 6, and 12-15 months. Vaccination with PCV13 is also recommended for those ages 65 or older, to be followed at least one year later with pneumococcal polysaccharide vaccine (Pneumovax or PPSV23).15 Additional doses of PCV13, PPSV23, or both may be indicated, depending on health status.
Strategy 4: Encourage good hygiene
The availability of immunizations and antivirals does not replace good hygiene. Frequent handwashing reduces the transmission of respiratory viruses, including influenza.16 Few studies have evaluated the use of alcohol-based hand sanitizers, but available evidence suggests they are effective in lowering viral transmission.16
Barriers, such as masks, gloves, and gowns, are helpful for health care workers.16 Surgical masks are often considered more comfortable to wear than N95 respirators. It may therefore be welcome news that when a 2009 randomized study assessed their use by hospital-based nurses, masks were non-inferior in protecting these health care workers against influenza.17
Presenteeism, the practice of going to work while sick, should be discouraged. People at risk for influenza may wish to avoid crowds during flu season; those with symptoms should be encouraged to stay home and limit contact with others.
Continue to: Treatment
Treatment
Strategy 1: Make prompt use of antivirals
Despite available preventive measures, tens of millions of people in the United States develop influenza every year. Use of antiviral medication, begun early in the course of illness, can reduce the duration of symptoms and may reduce the risk for complications.
The neuraminidase inhibitor (NI) group of antivirals—oseltamivir, zanamivir, and peramivir—is effective against influenza types A and B and current resistance rates are low.
The adamantine family of antivirals, amantadine and rimantadine, treat type A only. Since the circulating influenza strains in the past several seasons have demonstrated resistance >99%, these medications are not currently recommended.14
NIs reduce the duration of influenza symptoms by 10% to 20%, shortening the illness by 6 to 24 hours.18,19 In otherwise healthy patients, this benefit must be balanced against the increased risk for nausea and vomiting (oseltamivir), bronchospasm and sinusitis (zanamivir), and diarrhea (peramivir). In adults, NIs reduce the risk for lower respiratory tract complications and hospitalization. A 2015 meta-analysis by Dobson et al found a relative risk for hospitalization among those prescribed oseltamivir vs placebo of 37%.18
In the past, antivirals were used only in high-risk patients, such as children younger than 2 years, adults older than 65 years, and those with chronic health conditions.14 Now, antivirals are recommended for those who are at higher risk for complications (see Table 4), those with “severe, complicated, or progressive illness,” and hospitalized patients.14
Continue to: Antiviral treatment may have some value...
Antiviral treatment may have some value for hospitalized patients when started even 5 days after symptom onset. Treatment may be extended beyond the usual recommendations (5 days for oseltamivir or zanamivir) in immunosuppressed patients or the critically ill. Additionally, recent guidelines include consideration of antiviral treatment in outpatients who are at normal risk if treatment can be started within 48 hours of symptom onset.14
The CDC currently recommends use of oseltamivir rather than other antivirals for most hospitalized patients, based on the availability of data on its use in this setting.14 Intravenous peramivir is recommended for patients who cannot tolerate or absorb oral medication; inhaled zanamivir or IV peramivir are preferred for patients with end-stage renal disease who are not undergoing dialysis (see Table 3).14
Strategy 2: Exercise caution when it comes to supportive care
There are other medications that may offer symptom relief or prevent complications, especially when antivirals are contraindicated or unavailable.
Corticosteroids are recommended as part of the treatment of community-acquired pneumonia,20 but their role in influenza is controversial. A 2016 Cochrane review21 found no randomized controlled trials on the topic. Although the balance of available data from observational studies indicated that use of corticosteroids was associated with increased mortality, the authors also noted that all the studies included in their meta-analysis were of “very low quality.” They concluded that “the use of steroids in influenza remains a clinical judgement call.”
Statins may be associated with improved outcomes in influenza and pneumonia. Studies thus far have given contradictory results,22,23 and a planned Cochrane review of the question has been withdrawn.24
Continue to: Over-the-counter medications...
Over-the-counter medications, such as aspirin, acetaminophen, and ibuprofen are often used to manage the fever and myalgia associated with influenza. Patients should be cautioned against using the same ingredient in multiple different branded medications. Acetaminophen, for example, is not limited to Tylenol-branded products. To avoid Reye’s syndrome, children and teens with febrile illness, such as influenza, should not use aspirin.
CORRESPONDENCE
Jennifer L. Hamilton, MD, PhD, Drexel Family Medicine, 10 Shurs Lane, Suite 301, Philadelphia, PA 19127; [email protected].
Last year’s influenza season was severe enough that hospitals around the United States set up special evaluation areas beyond their emergency departments, at times spilling over to tents or other temporary structures in what otherwise would be parking lots. The scale and potential severity of the annual epidemic can be difficult to convey to our patients, who sometimes say “just the flu” to refer to an illness responsible for more than 170 pediatric deaths in the United States this past year.1 The Centers for Disease Control and Prevention (CDC) recently updated its 5-year estimates of influenza-related deaths in the United States; influenza mortality ranges from about 12,000 deaths in a mild season (such as 2011-2012) to 56,000 in a more severe season (eg, 2012-2013).2
Although influenza cannot be completely prevented, the following strategies can help reduce the risk for the illness and limit its severity if contracted.
Prevention
Strategy 1: Vaccinate against influenza
While the efficacy of vaccines varies from year to year, vaccination remains the core of influenza prevention efforts. In this decade, vaccine effectiveness has ranged from 19% to 60%.3 However, models suggest that even when the vaccine is only 20% effective, vaccinating 140 million people (the average number of doses delivered annually in the United States over the past 5 seasons) prevents 21 million infections, 130,000 hospitalizations, and more than 61,000 deaths.4 In a case-control study, Flannery et al found that vaccination was 65% effective in preventing laboratory-confirmed influenza-associated death in children over 4 seasons (July 2010 through June 2014).5
Deciding who should be vaccinated is simpler than in prior years: Rather than targeting people who are at higher risk (those ages 65 and older, or those with comorbidities), the current CDC recommendation is to vaccinate nearly everyone ages 6 months or older, with limited exceptions.6,7 (See Table 18).
Formulations. Many types of influenza vaccine are approved for use in the United States; these differ in the number of strains included (3 or 4), the amount of antigen present for each strain, the presence of an adjuvant, the growth medium used for the virus, and the route of administration (see Table 29). The relative merits of each type are a matter of some debate. There is ongoing research into the comparative efficacy of vaccines comprised of egg- vs cell-based cultures, as well as studies comparing high-dose or adjuvant vaccines to standard-dose inactivated vaccines.
Previously, the CDC has recommended preferential use (or avoidance) of some vaccine types, based on their efficacy. For the 2018-2019 flu season, however, the CDC has rescinded its recommendation against vaccine containing live attenuated virus (LAIV; FluMist brand) and expresses no preference for any vaccine formulation for patients of appropriate age and health status.10 The American Academy of Pediatrics (AAP), however, is recommends that LAIV be used only if patients and their families decline injectable vaccines.11
Timing. Influenza vaccines are now distributed as early as July to some locations, raising concerns about waning immunity from early vaccination (July/August) if the influenza season does not peak until February or March.8,12,13 Currently, the CDC recommends balancing the possible benefit of delayed vaccination against the risks of missed opportunities to vaccinate, a possible early season, and logistical problems related to vaccinating the same number of people in a smaller time interval. Offering vaccination by the end of October, if possible, is recommended in order for immunity to develop by mid-November.8 Note: Children ages 6 months to 8 years will need to receive their initial vaccination in 2 half-doses administered at least 28 days apart; completing their vaccination by the end of October would require starting the process weeks earlier.
[polldaddy:10124269]
Continue to: Strategy 2
Strategy 2: Make use of chemoprophylaxis
Preventive use of antiviral medication (chemoprophylaxis) may be a useful adjunct or alternative to vaccination in certain circumstances: if the patient is at high risk for complications, has been exposed to someone with influenza, has contraindications to vaccination, or received the vaccine within the past 2 weeks. The CDC also suggests that chemoprophylaxis be considered for those with immune deficiencies or who are otherwise immunosuppressed after exposure.14 Antivirals can also be used to control outbreaks in long-term care facilities; in these cases, the recommendedregimen is daily administration for at least 2 weeks, continuing until at least 7 days after the identification of the last case.14 Oseltamivir (Tamiflu) and zanamivir (Relenza) are the recommended prophylactic agents; a related intravenous medication, peramivir (Rapivab), is recommend for treatment only (see Table 314).
Strategy 3: Prevent comorbidities and opportunistic infections
Morbidity associated with influenza often comes from secondary infection. Pneumonia is among the most common complications, so influenza season is a good time to ensure that patients are appropriately vaccinated against pneumococcus, as well. Pneumococcal conjugate vaccine (Prevnar or PCV13) is recommended for children younger than 2 years of age, to be administered in a series of 4 doses: at 2, 4, 6, and 12-15 months. Vaccination with PCV13 is also recommended for those ages 65 or older, to be followed at least one year later with pneumococcal polysaccharide vaccine (Pneumovax or PPSV23).15 Additional doses of PCV13, PPSV23, or both may be indicated, depending on health status.
Strategy 4: Encourage good hygiene
The availability of immunizations and antivirals does not replace good hygiene. Frequent handwashing reduces the transmission of respiratory viruses, including influenza.16 Few studies have evaluated the use of alcohol-based hand sanitizers, but available evidence suggests they are effective in lowering viral transmission.16
Barriers, such as masks, gloves, and gowns, are helpful for health care workers.16 Surgical masks are often considered more comfortable to wear than N95 respirators. It may therefore be welcome news that when a 2009 randomized study assessed their use by hospital-based nurses, masks were non-inferior in protecting these health care workers against influenza.17
Presenteeism, the practice of going to work while sick, should be discouraged. People at risk for influenza may wish to avoid crowds during flu season; those with symptoms should be encouraged to stay home and limit contact with others.
Continue to: Treatment
Treatment
Strategy 1: Make prompt use of antivirals
Despite available preventive measures, tens of millions of people in the United States develop influenza every year. Use of antiviral medication, begun early in the course of illness, can reduce the duration of symptoms and may reduce the risk for complications.
The neuraminidase inhibitor (NI) group of antivirals—oseltamivir, zanamivir, and peramivir—is effective against influenza types A and B and current resistance rates are low.
The adamantine family of antivirals, amantadine and rimantadine, treat type A only. Since the circulating influenza strains in the past several seasons have demonstrated resistance >99%, these medications are not currently recommended.14
NIs reduce the duration of influenza symptoms by 10% to 20%, shortening the illness by 6 to 24 hours.18,19 In otherwise healthy patients, this benefit must be balanced against the increased risk for nausea and vomiting (oseltamivir), bronchospasm and sinusitis (zanamivir), and diarrhea (peramivir). In adults, NIs reduce the risk for lower respiratory tract complications and hospitalization. A 2015 meta-analysis by Dobson et al found a relative risk for hospitalization among those prescribed oseltamivir vs placebo of 37%.18
In the past, antivirals were used only in high-risk patients, such as children younger than 2 years, adults older than 65 years, and those with chronic health conditions.14 Now, antivirals are recommended for those who are at higher risk for complications (see Table 4), those with “severe, complicated, or progressive illness,” and hospitalized patients.14
Continue to: Antiviral treatment may have some value...
Antiviral treatment may have some value for hospitalized patients when started even 5 days after symptom onset. Treatment may be extended beyond the usual recommendations (5 days for oseltamivir or zanamivir) in immunosuppressed patients or the critically ill. Additionally, recent guidelines include consideration of antiviral treatment in outpatients who are at normal risk if treatment can be started within 48 hours of symptom onset.14
The CDC currently recommends use of oseltamivir rather than other antivirals for most hospitalized patients, based on the availability of data on its use in this setting.14 Intravenous peramivir is recommended for patients who cannot tolerate or absorb oral medication; inhaled zanamivir or IV peramivir are preferred for patients with end-stage renal disease who are not undergoing dialysis (see Table 3).14
Strategy 2: Exercise caution when it comes to supportive care
There are other medications that may offer symptom relief or prevent complications, especially when antivirals are contraindicated or unavailable.
Corticosteroids are recommended as part of the treatment of community-acquired pneumonia,20 but their role in influenza is controversial. A 2016 Cochrane review21 found no randomized controlled trials on the topic. Although the balance of available data from observational studies indicated that use of corticosteroids was associated with increased mortality, the authors also noted that all the studies included in their meta-analysis were of “very low quality.” They concluded that “the use of steroids in influenza remains a clinical judgement call.”
Statins may be associated with improved outcomes in influenza and pneumonia. Studies thus far have given contradictory results,22,23 and a planned Cochrane review of the question has been withdrawn.24
Continue to: Over-the-counter medications...
Over-the-counter medications, such as aspirin, acetaminophen, and ibuprofen are often used to manage the fever and myalgia associated with influenza. Patients should be cautioned against using the same ingredient in multiple different branded medications. Acetaminophen, for example, is not limited to Tylenol-branded products. To avoid Reye’s syndrome, children and teens with febrile illness, such as influenza, should not use aspirin.
CORRESPONDENCE
Jennifer L. Hamilton, MD, PhD, Drexel Family Medicine, 10 Shurs Lane, Suite 301, Philadelphia, PA 19127; [email protected].
1. CDC. Weekly US influenza surveillance report. https://www.cdc.gov/flu/weekly/index.htm. Published June 8, 2018. Accessed August 22, 2018.
2. CDC. Estimated influenza illnesses, medical visits, hospitalizations, and deaths averted by vaccination in the United States. Published April 19, 2017. https://www.cdc.gov/flu/about/disease/2015-16.htm. Accessed Setptember 18, 2018.
3. CDC. Seasonal influenza vaccine effectiveness, 2005-2018. https://www.cdc.gov/flu/professionals/vaccination/effectiveness-studies.htm. Published February 15, 2018. Accessed August 22, 2018.
4. Sah P, Medlock J, Fitzpatrick MC, et al. Optimizing the impact of low-efficacy influenza vaccines. Proc Natl Acad Sci. 2018:201802479.
5. Flannery B, Reynolds SB, Blanton L, et al. Influenza vaccine effectiveness against pediatric deaths: 2010-2014. Pediatrics. 2017;139: e20164244.
6. Kim DK, Riley LE, Hunter P. Advisory Committee on Immunization Practices recommended immunization schedule for adults aged 19 years or older—United States, 2018. MMWR Morb Mortal Wkly Rep. 2018;67:158–160.
7. Robinson CL, Romero JR, Kempe A, et al. Advisory Committee on Immunization Practices recommended immunization schedule for children and adolescents aged 18 years or younger—United States, 2018. MMWR Morb Mortal Wkly Rep. 2018;67:156–157.
8. Grohskopf LA, Sokolow LZ, Broder KR, et al. Prevention and control of seasonal influenza with vaccines: recommendations of the Advisory Committee on Immunization Practices—United States, 2017-18 influenza season. MMWR Recomm Rep. 2017;66:1-20.
9. CDC. Influenza vaccines—United States, 2017–18 influenza season. https://www.cdc.gov/flu/protect/vaccine/vaccines.htm. Published May 16, 2018. Accessed August 22, 2018.
10. Grohskopf LA, Sokolow LZ, Fry AM, et al. Update: ACIP recommendations for the use of quadrivalent live attenuated influenza vaccine (LAIV4)—United States, 2018-19 influenza season. MMWR Morb Mortal Wkly Rep. 2018;67:643–645.
11. Jenco M. AAP: Give children IIV flu shot; use LAIV as last resort. AAP News. May 21, 2018. http://www.aappublications.org/news/2018/05/21/fluvaccine051818. Accessed August 22, 2018.
12. Glinka ER, Smith DM, Johns ST. Timing matters—influenza vaccination to HIV-infected patients. HIV Med. 2016;17:601-604.
13. Castilla J, Martínez-Baz I, Martínez-Artola V, et al. Decline in influenza vaccine effectiveness with time after vaccination, Navarre, Spain, season 2011/12. Euro Surveill. 2013;18:20388.
14. CDC. Influenza antiviral medications: summary for clinicians. https://www.cdc.gov/flu/professionals/antivirals/summary-clinicians.htm. Published May 11, 2018. Accessed August 22, 2018.
15. CDC. Pneumococcal vaccination summary: who and when to vaccinate. https://www.cdc.gov/vaccines/vpd/pneumo/hcp/who-when-to-vaccinate.html. Published February 28, 2018. Accessed August 22, 2018.
16. Jefferson T, Del Mar CB, Dooley L, et al. Physical interventions to interrupt or reduce the spread of respiratory viruses. Cochrane Database Syst Rev. 2011;(7):CD006207.
17. Loeb M, Dafoe N, Mahony J, et al. Surgical mask vs N95 respirator for preventing influenza among health care workers: a randomized trial. JAMA. 2009;302:1865-1871.
18. Dobson J, Whitley RJ, Pocock S, Monto AS. Oseltamivir treatment for influenza in adults: a meta-analysis of randomised controlled trials. Lancet. 2015;385:1729-1737.
19. Ghebrehewet S, MacPherson P, Ho A. Influenza. BMJ. 2016;355:i6258.
20. Kaysin A, Viera AJ. Community-acquired pneumonia in adults: diagnosis and management. Am Fam Physician. 2016;94:698-706.
21. Rodrigo C, Leonardi‐Bee J, Nguyen‐Van‐Tam J, et al. Corticosteroids as adjunctive therapy in the treatment of influenza. Cochrane Database Syst Rev. 2016;3:CD010406.
22. Brassard P, Wu JW, Ernst P, et al. The effect of statins on influenza-like illness morbidity and mortality. Pharmacoepidemiol Drug Saf. 2017;26:63-70.
23. Fedson DS. Treating influenza with statins and other immunomodulatory agents. Antiviral Res. 2013;99:417-435.
24. Khandaker G, Rashid H, Chow MY, et al. Statins for influenza and pneumonia. Cochrane Database Syst Rev. January 9, 2017 [withdrawn].
1. CDC. Weekly US influenza surveillance report. https://www.cdc.gov/flu/weekly/index.htm. Published June 8, 2018. Accessed August 22, 2018.
2. CDC. Estimated influenza illnesses, medical visits, hospitalizations, and deaths averted by vaccination in the United States. Published April 19, 2017. https://www.cdc.gov/flu/about/disease/2015-16.htm. Accessed Setptember 18, 2018.
3. CDC. Seasonal influenza vaccine effectiveness, 2005-2018. https://www.cdc.gov/flu/professionals/vaccination/effectiveness-studies.htm. Published February 15, 2018. Accessed August 22, 2018.
4. Sah P, Medlock J, Fitzpatrick MC, et al. Optimizing the impact of low-efficacy influenza vaccines. Proc Natl Acad Sci. 2018:201802479.
5. Flannery B, Reynolds SB, Blanton L, et al. Influenza vaccine effectiveness against pediatric deaths: 2010-2014. Pediatrics. 2017;139: e20164244.
6. Kim DK, Riley LE, Hunter P. Advisory Committee on Immunization Practices recommended immunization schedule for adults aged 19 years or older—United States, 2018. MMWR Morb Mortal Wkly Rep. 2018;67:158–160.
7. Robinson CL, Romero JR, Kempe A, et al. Advisory Committee on Immunization Practices recommended immunization schedule for children and adolescents aged 18 years or younger—United States, 2018. MMWR Morb Mortal Wkly Rep. 2018;67:156–157.
8. Grohskopf LA, Sokolow LZ, Broder KR, et al. Prevention and control of seasonal influenza with vaccines: recommendations of the Advisory Committee on Immunization Practices—United States, 2017-18 influenza season. MMWR Recomm Rep. 2017;66:1-20.
9. CDC. Influenza vaccines—United States, 2017–18 influenza season. https://www.cdc.gov/flu/protect/vaccine/vaccines.htm. Published May 16, 2018. Accessed August 22, 2018.
10. Grohskopf LA, Sokolow LZ, Fry AM, et al. Update: ACIP recommendations for the use of quadrivalent live attenuated influenza vaccine (LAIV4)—United States, 2018-19 influenza season. MMWR Morb Mortal Wkly Rep. 2018;67:643–645.
11. Jenco M. AAP: Give children IIV flu shot; use LAIV as last resort. AAP News. May 21, 2018. http://www.aappublications.org/news/2018/05/21/fluvaccine051818. Accessed August 22, 2018.
12. Glinka ER, Smith DM, Johns ST. Timing matters—influenza vaccination to HIV-infected patients. HIV Med. 2016;17:601-604.
13. Castilla J, Martínez-Baz I, Martínez-Artola V, et al. Decline in influenza vaccine effectiveness with time after vaccination, Navarre, Spain, season 2011/12. Euro Surveill. 2013;18:20388.
14. CDC. Influenza antiviral medications: summary for clinicians. https://www.cdc.gov/flu/professionals/antivirals/summary-clinicians.htm. Published May 11, 2018. Accessed August 22, 2018.
15. CDC. Pneumococcal vaccination summary: who and when to vaccinate. https://www.cdc.gov/vaccines/vpd/pneumo/hcp/who-when-to-vaccinate.html. Published February 28, 2018. Accessed August 22, 2018.
16. Jefferson T, Del Mar CB, Dooley L, et al. Physical interventions to interrupt or reduce the spread of respiratory viruses. Cochrane Database Syst Rev. 2011;(7):CD006207.
17. Loeb M, Dafoe N, Mahony J, et al. Surgical mask vs N95 respirator for preventing influenza among health care workers: a randomized trial. JAMA. 2009;302:1865-1871.
18. Dobson J, Whitley RJ, Pocock S, Monto AS. Oseltamivir treatment for influenza in adults: a meta-analysis of randomised controlled trials. Lancet. 2015;385:1729-1737.
19. Ghebrehewet S, MacPherson P, Ho A. Influenza. BMJ. 2016;355:i6258.
20. Kaysin A, Viera AJ. Community-acquired pneumonia in adults: diagnosis and management. Am Fam Physician. 2016;94:698-706.
21. Rodrigo C, Leonardi‐Bee J, Nguyen‐Van‐Tam J, et al. Corticosteroids as adjunctive therapy in the treatment of influenza. Cochrane Database Syst Rev. 2016;3:CD010406.
22. Brassard P, Wu JW, Ernst P, et al. The effect of statins on influenza-like illness morbidity and mortality. Pharmacoepidemiol Drug Saf. 2017;26:63-70.
23. Fedson DS. Treating influenza with statins and other immunomodulatory agents. Antiviral Res. 2013;99:417-435.
24. Khandaker G, Rashid H, Chow MY, et al. Statins for influenza and pneumonia. Cochrane Database Syst Rev. January 9, 2017 [withdrawn].
PRACTICE RECOMMENDATIONS
› Recommend influenza vaccination for all patients at least 6 months old unless a specific contraindication exists. A
› Recommend pneumococcal vaccination to appropriate patients to reduce the risk for a common complication of influenza. A
› Encourage hygiene-based measures to limit infection, including frequent handwashing or use of a hand sanitizer. B
› Prescribe oseltamivir to hospitalized influenza patients to limit the duration and severity of infection. B
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
Multiple payment systems impede universal HPV screening access
Multiple payment systems impede universal HPV screening access
Unless the issue of multiple systems of payment and access (that is, multiple insurance companies and providers) can be resolved in the United States, I do not believe there will be an advancement across the board for human papillomavirus (HPV) screening. In my opinion, we need to work toward access to health care for all and a single-payer system.
C.L. Conrad-Forrest, MD
Davis, California
Dr. Barbieri responds
I agree with Dr. Conrad-Forrest that improving cervical cancer screening would be advanced by interoperable electronic medical records and health systems that are designed to manage population health. I predict that a large integrated health system will be the first to adopt the use of high-risk HPV testing to screen for cervical cancer in the United States.
Share your thoughts! Send your Letter to the Editor to [email protected]. Please include your name and the city and state in which you practice.
Multiple payment systems impede universal HPV screening access
Unless the issue of multiple systems of payment and access (that is, multiple insurance companies and providers) can be resolved in the United States, I do not believe there will be an advancement across the board for human papillomavirus (HPV) screening. In my opinion, we need to work toward access to health care for all and a single-payer system.
C.L. Conrad-Forrest, MD
Davis, California
Dr. Barbieri responds
I agree with Dr. Conrad-Forrest that improving cervical cancer screening would be advanced by interoperable electronic medical records and health systems that are designed to manage population health. I predict that a large integrated health system will be the first to adopt the use of high-risk HPV testing to screen for cervical cancer in the United States.
Share your thoughts! Send your Letter to the Editor to [email protected]. Please include your name and the city and state in which you practice.
Multiple payment systems impede universal HPV screening access
Unless the issue of multiple systems of payment and access (that is, multiple insurance companies and providers) can be resolved in the United States, I do not believe there will be an advancement across the board for human papillomavirus (HPV) screening. In my opinion, we need to work toward access to health care for all and a single-payer system.
C.L. Conrad-Forrest, MD
Davis, California
Dr. Barbieri responds
I agree with Dr. Conrad-Forrest that improving cervical cancer screening would be advanced by interoperable electronic medical records and health systems that are designed to manage population health. I predict that a large integrated health system will be the first to adopt the use of high-risk HPV testing to screen for cervical cancer in the United States.
Share your thoughts! Send your Letter to the Editor to [email protected]. Please include your name and the city and state in which you practice.
Obesity: When to consider surgery
Patients with overweight and obesity are at increased risk of multiple morbidities, including cardiovascular disease, stroke, type 2 diabetes (T2D), osteoarthritis, obstructive sleep apnea (OSA), and all-cause mortality.1 Even modest weight loss—5% to 10%—can lead to a clinically relevant reduction in this risk of disease.2,3 The American Academy of Family Physicians recognizes obesity as a disease, and recommends screening of all adults for obesity and referral for those with body mass index (BMI)* ≥30 to intensive, multicomponent behavioral interventions.4,5
For some patients, diet, exercise, and behavioral modifications are sufficient; for the great majority, however, weight loss achieved by lifestyle modification is counteracted by metabolic adaptations that promote weight regain.6 For patients with obesity who are unable to achieve or maintain sufficient weight loss to improve health outcomes with lifestyle modification alone, options include pharmacotherapy, devices, endoscopic bariatric therapies, and bariatric surgery.
Bariatric surgery is the most effective of these treatments, due to its association with significant and sustained weight loss, reduction in obesity-related comorbidities, and improved quality of life.1,7 Furthermore, compared with usual care, bariatric surgery is associated with a reduced number of cardiovascular deaths, a lower incidence of cardiovascular events in adults with obesity, and a long-term reduction in overall mortality.8-10
What are the options? Who is a candidate?
The 3 most common bariatric procedures in the United States are sleeve gastrectomy (SG), Roux-en-Y gastric bypass (RYGB), and laparoscopic adjustable gastric band (LAGB).11 SG and RYGB are performed more often than the LAGB, consequent to greater efficacy and fewer complications.12 Weight loss is maximal at 1 to 2 years, and is estimated to be 15% of total body weight for LAGB; 25% for SG; and 35% for RYGB.13,14
Not all patients are candidates for bariatric surgery. Contraindications include chronic obstructive pulmonary disease or respiratory dysfunction, poor cardiac reserve, nonadherence to medical treatment, and severe psychological disorders.15 Because some patients have difficulty maintaining weight loss following bariatric surgery and, on average, patients regain at least some weight, patients must understand that long-term lifestyle changes and follow-up are critical to the success of bariatric surgery.16
When should bariatric surgery be considered?
American Heart Association/American College of Cardiology/The Obesity Society guidelines16 conceptualize 2 indications for bariatric surgery:
- adults with BMI ≥40
- adults with BMI ≥35 who have obesity-related comorbid conditions and are motivated to lose weight but have not responded to behavioral treatment, with or without pharmacotherapy, to achieve sufficient weight loss for target health goals.
American Association of Clinical Endocrinologists guidelines17 conceptualize 3 indications for bariatric surgery:
- adults with BMI ≥40
- adults with BMI ≥35 with 1 or more severe obesity-related complications
- adults with BMI 30-34.9 with diabetes or metabolic syndrome (evidence for this recommendation is limited).
Continue to: The 3 illustrative vignettes presented...
The 3 illustrative vignettes presented in this article offer examples of patients with obesity who could benefit from bariatric surgery. Each has been unable to achieve or maintain sufficient weight loss to improve health outcomes with nonsurgical interventions alone.
CASE 1
Sleep apnea persists despite weight loss
Robin W, a 50-year-old woman with class-II obesity (5’8”; 250 lb; BMI, 38 ), OSA requiring continuous positive airway pressure (CPAP), hyperlipidemia, hypertension, and iron-deficiency anemia secondary to menorrhagia, and taking an iron supplement, presents for weight management. She has lost 50 lb, reducing her BMI from 45.6 with behavioral modifications and pharmacotherapy, but she has been unsuccessful at achieving further weight loss despite a reduced-calorie diet and at least 30 minutes of physical activity most days.
Ms. W is frustrated that she has reached a weight plateau; she is motivated to lose more weight. Her goal is to improve her weight-related comorbid conditions and reduce her medication requirement. Despite the initial weight loss, she continues to require CPAP therapy for OSA and remains on 3 medications for hypertension. She does not have cardiac or respiratory disease, psychiatric diagnoses, or a history of gastroesophageal reflux disease (GERD).
Is bariatric surgery a reasonable option for Ms. W? If so, which procedure would you recommend?
Good option for Ms. W: Sleeve gastrectomy
It is reasonable to consider bariatric surgery—in particular, SG—for this patient with class-II obesity and multiple weight-related comorbid conditions because she has been unable to achieve further weight loss with more conservative measures.
Continue to: How does the procedure work?
How does the procedure work? SG removes a large portion of the stomach along the greater curvature, reducing the organ to approximately 15% to 25% of its original size.18 The procedure leaves the pyloric valve intact and does not involve removal or bypass of the intestines.
How appealing and successful is it? The majority of patients who undergo SG experience significant weight loss; studies report approximately 25% total body weight loss after 1 to 2 years.14 Furthermore, most patients with T2D experience resolution of, or improvement in, disease markers.19 Because SG leaves the pylorus intact, there are fewer restrictions on what a patient can eat after surgery, compared with RYGB. With further weight loss, Ms. W may experience improvement in, or resolution of, hypertension, hyperlipidemia, and OSA.
The SG procedure itself is simpler than some other bariatric procedures and presents less risk of malabsorption because the intestines are left intact. Patients who undergo SG report feeling less hungry because the fundus of the stomach, which secretes ghrelin (the so-called hunger hormone), is removed.18,20
What are special considerations, including candidacy? Patients with GERD are not ideal candidates for this procedure because exacerbation of the disease is a potential associated adverse event. SG is a reasonable surgical option for Ms. W because the procedure is less likely to exacerbate her nutritional deficiency (iron-deficiency anemia), compared to RYGB, and she does not have a history of GERD.
What are the complications? Complications of SG occur at a lower rate than they do with RYGB, which is associated with a greater risk of nutritional deficiency.18 Common early complications of SG include leaking, bleeding, stenosis, GERD, and vomiting due to excessive eating. Late complications include stomach expansion by 12 months, leading to decreased restriction.15 Unlike RYGB and LAGB, SG is not reversible.
Continue to: CASE 2
CASE 2
Severe obesity, polypharmacy for type 2 diabetes
Anne P, a 42-year-old woman with class-III obesity (5’6”; 290 lb; BMI, 46.8 kg/m2), presents to discuss bariatric surgery. Comorbidities include T2D, for which she takes metformin, a glucagon-like peptide-1 (GLP-1) receptor agonist, and a sodium–glucose cotransporter-2 (SGLT-2) inhibitor; GERD; hypertension, for which she takes an angiotensin-converting enzyme inhibitor and a calcium-channel blocker; hyperlipidemia, for which she takes a statin; and osteoarthritis.
Ms. P lost 30 pounds—reducing her BMI from 51.6—when the sulfonylurea and thiazolidinedione she was taking were switched to the GLP-1 receptor agonist and the SGLT2 inhibitor. She also made behavioral modifications, including 30 minutes a day of physical activity and a reduced-calorie meal plan under the guidance of a dietitian.
However, Ms. P has been unable to lose more weight or reduce her hemoglobin A1c (HbA1c) level below 8%. Her goal is to avoid the need to take insulin (which several members of her family take), lower her HbA1c level, and decrease her medication requirement.
Ms. P does not have cardiac or respiratory disease or psychiatric diagnoses. Which surgical intervention would you recommend for her?
Good option for Ms. P: Roux-en-Y gastric bypass
RYGB is a reasonable option for a patient with class-III obesity and multiple comorbidities, including poorly controlled T2D and GERD, who has failed conservative measures but wants to lose more weight, reduce her HbA1c, reduce her medication requirement, and avoid the need for insulin.
Continue to: How does the procedure work?
How does the procedure work? RYGB constructs a small pouch from the proximal portion of the stomach and attaches it directly to the jejunum, thus bypassing part of the stomach and duodenum. The procedure is effective for weight loss because it is both restrictive and malabsorptive: patients not only eat smaller portions, but cannot absorb all they eat. Other mechanisms attributed to RYGB that are hypothesized to promote weight loss include21:
- alteration of endogenous gut hormones, which promotes postprandial satiety
- increased levels of bile acids, which promotes alteration of the gut microbiome
- intestinal hypertrophy.
How successful is it? RYGB is associated with significant total body weight loss of approximately 35% at 2 years.9 The procedure has been shown to produce superior outcomes in reducing comorbid disease compared to other bariatric procedures or medical therapy alone. Of the procedures discussed in this article, RYGB is associated with the greatest reduction in triglycerides, HbA1c, and use of diabetes medications, including insulin.22
What are special considerations, including candidacy? For patients with mild or moderate T2D (calculated using the Individualized Metabolic Surgery Score [http://riskcalc.org/Metabolic_Surgery_Score/], which categorizes patients by number of diabetes medications, insulin use, duration of diabetes before surgery, and HbA1c), RYGB is recommended over SG because it leads to greater long-term remission of T2D.
RYGB is associated with a lower rate of GERD than SG and can even alleviate GERD in patients who have the disease. Furthermore, for patients with limited pancreatic beta cell reserve, RYBG and SG have similarly low efficacy for T2D remission; SG is therefore recommended over RYGB in this specific circumstance, given its slightly lower risk profile.23
What are the complications? Patients who undergo any bariatric surgical procedure require long-term follow-up and vitamin supplementation, but those who undergo RYGB require stricter dietary adherence after the procedure; lifelong vitamin (D, B12, folic acid, and thiamine), iron, and calcium supplementation; and long-term follow-up to reduce the risk and severity of complications and to monitor for nutritional deficiencies.7 As such, patients who have shown poor adherence to medical treatment are not good candidates for the procedure.
Continue to: Early complications include...
Early complications include leak, stricture, obstruction, and failure of the staple partition of the upper stomach. Late complications include nutritional deficiencies, as noted, and ulceration of the anastomosis. Dumping syndrome (overly rapid transit of food from the stomach into the small intestine) can develop early or late; early dumping leads to osmotic diarrhea and abdominal cramping, and late dumping leads to reactive hypoglycemia.15
Technically, RYGB is a reversible procedure, although generally it is reversed only in extreme circumstances.
CASE 3
Fatty liver disease, hesitation to undergo surgery
Walt Z, a 35 year-old-man with class-II obesity (5’10”; 265 lb; BMI, 38 kg/m2), T2D, and hepatic steatosis, presents for weight management. He has been able to lose modest weight over the years with behavioral modifications, but has been unsuccessful in maintaining that loss. He requests referral to a bariatric surgeon but is concerned about the permanence and invasiveness of most bariatric procedures.
Which surgical intervention would you recommend for this patient?
Good option for Mr. Z: Laparoscopic adjustable gastric band
Given that Mr. Z is a candidate for a surgical intervention but does not want a permanent or invasive procedure, LAGB is a reasonable option.
Continue to: How does the procedure work?
How does the procedure work? LAGB is a reversible procedure in which an inflatable band is placed around the fundus of the stomach to create a small pouch. The band can be adjusted to regulate food intake by adding or removing saline through a subcutaneous access port.
How appealing and successful is it? LAGB results in approximately 15% total body weight loss at 2 years.13 Because the procedure is purely restrictive, it carries a reduced risk of nutritional deficiency associated more commonly with malabsorptive procedures.
What are special considerations, including candidacy? As noted, Mr. Z expressed concern about the permanence and invasiveness of most bariatric procedures, and therefore wants to undergo a reversible procedure; LAGB can be a reasonable option for such a patient. Patients who want a reversible or minimally invasive procedure should also be made aware that endoscopic bariatric therapies and other devices are being developed to fill the treatment gap in the management of obesity.
What are the complications? Although LAGB is the least invasive procedure discussed here, it is associated with the highest rate of complications—most commonly, complications associated with the band itself (eg, nausea, vomiting, obstruction, band erosion or migration, esophageal dysmotility leading to acid reflux) and failure to lose weight.7 LAGB also requires more postoperative visits than other procedures, to optimize band tightness. A high number of bands are removed eventually because of complications or inadequate weight loss, or both.13,24
Shared decision-making and dialogue are essential to overcome obstacles
Despite the known benefits of bariatric surgery, including greater reduction in the risk and severity of obesity-related comorbid conditions than seen with other interventions and a long-term reduction in overall mortality when compared with usual care, fewer than 1% of eligible patients undergo a weight-loss procedure.25 Likely, this is due to:
- limited patient knowledge of the health benefits of surgery
- limited provider comfort recommending surgery
- inadequate insurance coverage, which might, in part, be due to a lack of prospective studies comparing various bariatric procedures.18
Continue to: Ultimately, the decision whether to undergo a bariatric procedure...
Ultimately, the decision whether to undergo a bariatric procedure, and which one(s) to consider, should be the product of a thorough conversation between patient and provider.
CORRESPONDENCE
Sarah R. Barenbaum, MD, Department of Internal Medicine, New York–Presbyterian Hospital/Weill Cornell Medical College, 530 East 70th Street, M-507, New York, NY 10021; [email protected]
1. Must A, Spadano J, Coakley EH, et al. The disease burden associated with overweight and obesity. JAMA. 1999;282:1523-1529.
2. Wing RR, Lang W, Wadden TA, et al. Benefits of modest weight loss in improving cardiovascular risk factors in overweight and obese individuals with type 2 diabetes. Diabetes Care. 2011;34:1481-1486.
3. Magkos F, Fraterrigo G, Yoshino J, et al. Effects of moderate and subsequent progressive weight loss on metabolic function and adipose tissue biology in humans with obesity. Cell Metab. 2016;23:591-601.
4. American Academy of Family Physicians. Clinical preventive service recommendation: Obesity. www.aafp.org/patient-care/clinical-recommendations/all/obesity.html. Accessed August 22, 2018.
5. American Academy of Family Physicians: USPSTF draft recommendation: Intensive behavioral interventions recommended for obesity. www.aafp.org/news/health-of-the-public/20180221uspstfobesity.html. Published February 21, 2018. Accessed August 22, 2018.
6. Saunders KH, Shukla AP, Igel LI, Aronne LJ. Obesity: When to consider medication. J Fam Pract. 2017;66:608-616.
7. Roux CW, Heneghan HM. Bariatric surgery for obesity. Med Clin North Am. 2018;102:165-182.
8. Sjöström L, Peltonen M, Jacobson P, et al. Bariatric surgery and long-term cardiovascular events. JAMA. 2012;307:56-65.
9. Sjöström L. Review of the key results from the Swedish Obese Subjects (SOS) trial - a prospective controlled intervention study of bariatric surgery. J Intern Med. 2013;273:219-234.
10. Reges O, Greenland P, Dicker D, et al. Association of bariatric surgery using laparoscopic banding, Roux-en-Y, gastric bypass, or laparoscopic sleeve gastrectomy vs usual care obesity management with all-cause mortality. JAMA. 2018;319:279-290.
11. Lee JH, Nguyen QN, Le QA. Comparative effectiveness of 3 bariatric surgery procedures: Roux-en-Y gastric bypass, laparoscopic adjustable gastric band, and sleeve gastrectomy. Surg Obes Relat Dis. 2016;12:997-1002.
12. American Society for Metabolic and Bariatric Surgery. Estimate of bariatric surgery numbers, 2011-2017. https://asmbs.org/resources/estimate-of-bariatric-surgery-numbers. Published June 2018. Accessed August 22, 2018.
13. Courcoulas AP, King WC, Belle SH, et al. Seven-year weight trajectories and health outcomes in the Longitudinal Assessment of Bariatric Surgery (LABS) Study. JAMA Surg. 2018;153:427-434.
14. Heymsfield SB, Wadden TA. Mechanisms, pathophysiology, and management of obesity. N Engl J Med. 2017;376:254-266.
15. Colquitt JL, Pickett K, Loveman E, Frampton GK. Surgery for weight loss in adults. Cochrane Database Syst Rev. 2014;(8):CD003641.
16. Jensen MD, Ryan DH, Apovian CM, et al; American College of Cardiology/American Heart Association Task Force on Practice Guidelines; Obesity Society. 2013 AHA/ACC/TOS guideline for the management of overweight and obesity in adults: a report of the American College of Cardiology/American Heart Association Task Force on Practice Guidelines and The Obesity Society. Circulation. 2014;129:S102-S138.
17. Garvey WT, Mechanick JI, Brett EM, et al; Reviewers of the AACE/ACE Obesity Clinical Practice Guidelines. American Association of Clinical Endocrinologists and American College of Endocrinology clinical practice guidelines for comprehensive medical care of patients with obesity. Endocr Pract. 2016;22 Suppl 3:1-203.
18. Carlin Am, Zeni Tm, English WJ, et al; Michigan Bariatric Surgery Collaborative. The comparative effectiveness of sleeve gastrectomy, gastric bypass, and adjustable gastric banding procedures for the treatment of morbid obesity. Ann Surg. 2013;257:791-797.
19. Gill RS, Birch DW, Shi X, et al. Sleeve gastrectomy and type 2 diabetes mellitus: a systematic review. Surg Obes Relat Dis. 2010;6:707-713.
20. Karamanakos SN, Vagenas K, Kalfarentzos F, et al. Weight loss, appetite suppression, and changes in fasting and postprandial ghrelin and peptide-YY levels after Roux-en-Y gastric bypass and sleeve gastrectomy. Ann Surg. 2008;247:401-407.
21. Abdeen G, le Roux CW. Mechanism underlying the weight loss and complications of Roux-en-Y gastric bypass. Obes Surg. 2016;26:410-421.
22. Schauer PR, Bhatt DL, Kirwan JP et al; STAMPEDE Investigators. Bariatric surgery versus intensive medical therapy for diabetes - 5-year outcomes. N Engl J Med. 2017;376:641-651.
23. Aminian A, Brethauer SA, Andalib A, et al. Individualized metabolic surgery score: procedure selection based on diabetes severity. Ann Surg. 2017;266:4:650-657.
24. Smetana GW, Jones DB, Wee CC. Beyond the guidelines: Should this patient have weight loss surgery? Grand rounds discussion from Beth Israel Deaconess Medical Center. Ann Intern Med. 2017;166:808-817.
25. Wolfe BM, Morton JM. Weighing in on bariatric surgery: procedure use, readmission rates, and mortality [editorial]. JAMA. 2005;294:1960-1963.
Patients with overweight and obesity are at increased risk of multiple morbidities, including cardiovascular disease, stroke, type 2 diabetes (T2D), osteoarthritis, obstructive sleep apnea (OSA), and all-cause mortality.1 Even modest weight loss—5% to 10%—can lead to a clinically relevant reduction in this risk of disease.2,3 The American Academy of Family Physicians recognizes obesity as a disease, and recommends screening of all adults for obesity and referral for those with body mass index (BMI)* ≥30 to intensive, multicomponent behavioral interventions.4,5
For some patients, diet, exercise, and behavioral modifications are sufficient; for the great majority, however, weight loss achieved by lifestyle modification is counteracted by metabolic adaptations that promote weight regain.6 For patients with obesity who are unable to achieve or maintain sufficient weight loss to improve health outcomes with lifestyle modification alone, options include pharmacotherapy, devices, endoscopic bariatric therapies, and bariatric surgery.
Bariatric surgery is the most effective of these treatments, due to its association with significant and sustained weight loss, reduction in obesity-related comorbidities, and improved quality of life.1,7 Furthermore, compared with usual care, bariatric surgery is associated with a reduced number of cardiovascular deaths, a lower incidence of cardiovascular events in adults with obesity, and a long-term reduction in overall mortality.8-10
What are the options? Who is a candidate?
The 3 most common bariatric procedures in the United States are sleeve gastrectomy (SG), Roux-en-Y gastric bypass (RYGB), and laparoscopic adjustable gastric band (LAGB).11 SG and RYGB are performed more often than the LAGB, consequent to greater efficacy and fewer complications.12 Weight loss is maximal at 1 to 2 years, and is estimated to be 15% of total body weight for LAGB; 25% for SG; and 35% for RYGB.13,14
Not all patients are candidates for bariatric surgery. Contraindications include chronic obstructive pulmonary disease or respiratory dysfunction, poor cardiac reserve, nonadherence to medical treatment, and severe psychological disorders.15 Because some patients have difficulty maintaining weight loss following bariatric surgery and, on average, patients regain at least some weight, patients must understand that long-term lifestyle changes and follow-up are critical to the success of bariatric surgery.16
When should bariatric surgery be considered?
American Heart Association/American College of Cardiology/The Obesity Society guidelines16 conceptualize 2 indications for bariatric surgery:
- adults with BMI ≥40
- adults with BMI ≥35 who have obesity-related comorbid conditions and are motivated to lose weight but have not responded to behavioral treatment, with or without pharmacotherapy, to achieve sufficient weight loss for target health goals.
American Association of Clinical Endocrinologists guidelines17 conceptualize 3 indications for bariatric surgery:
- adults with BMI ≥40
- adults with BMI ≥35 with 1 or more severe obesity-related complications
- adults with BMI 30-34.9 with diabetes or metabolic syndrome (evidence for this recommendation is limited).
Continue to: The 3 illustrative vignettes presented...
The 3 illustrative vignettes presented in this article offer examples of patients with obesity who could benefit from bariatric surgery. Each has been unable to achieve or maintain sufficient weight loss to improve health outcomes with nonsurgical interventions alone.
CASE 1
Sleep apnea persists despite weight loss
Robin W, a 50-year-old woman with class-II obesity (5’8”; 250 lb; BMI, 38 ), OSA requiring continuous positive airway pressure (CPAP), hyperlipidemia, hypertension, and iron-deficiency anemia secondary to menorrhagia, and taking an iron supplement, presents for weight management. She has lost 50 lb, reducing her BMI from 45.6 with behavioral modifications and pharmacotherapy, but she has been unsuccessful at achieving further weight loss despite a reduced-calorie diet and at least 30 minutes of physical activity most days.
Ms. W is frustrated that she has reached a weight plateau; she is motivated to lose more weight. Her goal is to improve her weight-related comorbid conditions and reduce her medication requirement. Despite the initial weight loss, she continues to require CPAP therapy for OSA and remains on 3 medications for hypertension. She does not have cardiac or respiratory disease, psychiatric diagnoses, or a history of gastroesophageal reflux disease (GERD).
Is bariatric surgery a reasonable option for Ms. W? If so, which procedure would you recommend?
Good option for Ms. W: Sleeve gastrectomy
It is reasonable to consider bariatric surgery—in particular, SG—for this patient with class-II obesity and multiple weight-related comorbid conditions because she has been unable to achieve further weight loss with more conservative measures.
Continue to: How does the procedure work?
How does the procedure work? SG removes a large portion of the stomach along the greater curvature, reducing the organ to approximately 15% to 25% of its original size.18 The procedure leaves the pyloric valve intact and does not involve removal or bypass of the intestines.
How appealing and successful is it? The majority of patients who undergo SG experience significant weight loss; studies report approximately 25% total body weight loss after 1 to 2 years.14 Furthermore, most patients with T2D experience resolution of, or improvement in, disease markers.19 Because SG leaves the pylorus intact, there are fewer restrictions on what a patient can eat after surgery, compared with RYGB. With further weight loss, Ms. W may experience improvement in, or resolution of, hypertension, hyperlipidemia, and OSA.
The SG procedure itself is simpler than some other bariatric procedures and presents less risk of malabsorption because the intestines are left intact. Patients who undergo SG report feeling less hungry because the fundus of the stomach, which secretes ghrelin (the so-called hunger hormone), is removed.18,20
What are special considerations, including candidacy? Patients with GERD are not ideal candidates for this procedure because exacerbation of the disease is a potential associated adverse event. SG is a reasonable surgical option for Ms. W because the procedure is less likely to exacerbate her nutritional deficiency (iron-deficiency anemia), compared to RYGB, and she does not have a history of GERD.
What are the complications? Complications of SG occur at a lower rate than they do with RYGB, which is associated with a greater risk of nutritional deficiency.18 Common early complications of SG include leaking, bleeding, stenosis, GERD, and vomiting due to excessive eating. Late complications include stomach expansion by 12 months, leading to decreased restriction.15 Unlike RYGB and LAGB, SG is not reversible.
Continue to: CASE 2
CASE 2
Severe obesity, polypharmacy for type 2 diabetes
Anne P, a 42-year-old woman with class-III obesity (5’6”; 290 lb; BMI, 46.8 kg/m2), presents to discuss bariatric surgery. Comorbidities include T2D, for which she takes metformin, a glucagon-like peptide-1 (GLP-1) receptor agonist, and a sodium–glucose cotransporter-2 (SGLT-2) inhibitor; GERD; hypertension, for which she takes an angiotensin-converting enzyme inhibitor and a calcium-channel blocker; hyperlipidemia, for which she takes a statin; and osteoarthritis.
Ms. P lost 30 pounds—reducing her BMI from 51.6—when the sulfonylurea and thiazolidinedione she was taking were switched to the GLP-1 receptor agonist and the SGLT2 inhibitor. She also made behavioral modifications, including 30 minutes a day of physical activity and a reduced-calorie meal plan under the guidance of a dietitian.
However, Ms. P has been unable to lose more weight or reduce her hemoglobin A1c (HbA1c) level below 8%. Her goal is to avoid the need to take insulin (which several members of her family take), lower her HbA1c level, and decrease her medication requirement.
Ms. P does not have cardiac or respiratory disease or psychiatric diagnoses. Which surgical intervention would you recommend for her?
Good option for Ms. P: Roux-en-Y gastric bypass
RYGB is a reasonable option for a patient with class-III obesity and multiple comorbidities, including poorly controlled T2D and GERD, who has failed conservative measures but wants to lose more weight, reduce her HbA1c, reduce her medication requirement, and avoid the need for insulin.
Continue to: How does the procedure work?
How does the procedure work? RYGB constructs a small pouch from the proximal portion of the stomach and attaches it directly to the jejunum, thus bypassing part of the stomach and duodenum. The procedure is effective for weight loss because it is both restrictive and malabsorptive: patients not only eat smaller portions, but cannot absorb all they eat. Other mechanisms attributed to RYGB that are hypothesized to promote weight loss include21:
- alteration of endogenous gut hormones, which promotes postprandial satiety
- increased levels of bile acids, which promotes alteration of the gut microbiome
- intestinal hypertrophy.
How successful is it? RYGB is associated with significant total body weight loss of approximately 35% at 2 years.9 The procedure has been shown to produce superior outcomes in reducing comorbid disease compared to other bariatric procedures or medical therapy alone. Of the procedures discussed in this article, RYGB is associated with the greatest reduction in triglycerides, HbA1c, and use of diabetes medications, including insulin.22
What are special considerations, including candidacy? For patients with mild or moderate T2D (calculated using the Individualized Metabolic Surgery Score [http://riskcalc.org/Metabolic_Surgery_Score/], which categorizes patients by number of diabetes medications, insulin use, duration of diabetes before surgery, and HbA1c), RYGB is recommended over SG because it leads to greater long-term remission of T2D.
RYGB is associated with a lower rate of GERD than SG and can even alleviate GERD in patients who have the disease. Furthermore, for patients with limited pancreatic beta cell reserve, RYBG and SG have similarly low efficacy for T2D remission; SG is therefore recommended over RYGB in this specific circumstance, given its slightly lower risk profile.23
What are the complications? Patients who undergo any bariatric surgical procedure require long-term follow-up and vitamin supplementation, but those who undergo RYGB require stricter dietary adherence after the procedure; lifelong vitamin (D, B12, folic acid, and thiamine), iron, and calcium supplementation; and long-term follow-up to reduce the risk and severity of complications and to monitor for nutritional deficiencies.7 As such, patients who have shown poor adherence to medical treatment are not good candidates for the procedure.
Continue to: Early complications include...
Early complications include leak, stricture, obstruction, and failure of the staple partition of the upper stomach. Late complications include nutritional deficiencies, as noted, and ulceration of the anastomosis. Dumping syndrome (overly rapid transit of food from the stomach into the small intestine) can develop early or late; early dumping leads to osmotic diarrhea and abdominal cramping, and late dumping leads to reactive hypoglycemia.15
Technically, RYGB is a reversible procedure, although generally it is reversed only in extreme circumstances.
CASE 3
Fatty liver disease, hesitation to undergo surgery
Walt Z, a 35 year-old-man with class-II obesity (5’10”; 265 lb; BMI, 38 kg/m2), T2D, and hepatic steatosis, presents for weight management. He has been able to lose modest weight over the years with behavioral modifications, but has been unsuccessful in maintaining that loss. He requests referral to a bariatric surgeon but is concerned about the permanence and invasiveness of most bariatric procedures.
Which surgical intervention would you recommend for this patient?
Good option for Mr. Z: Laparoscopic adjustable gastric band
Given that Mr. Z is a candidate for a surgical intervention but does not want a permanent or invasive procedure, LAGB is a reasonable option.
Continue to: How does the procedure work?
How does the procedure work? LAGB is a reversible procedure in which an inflatable band is placed around the fundus of the stomach to create a small pouch. The band can be adjusted to regulate food intake by adding or removing saline through a subcutaneous access port.
How appealing and successful is it? LAGB results in approximately 15% total body weight loss at 2 years.13 Because the procedure is purely restrictive, it carries a reduced risk of nutritional deficiency associated more commonly with malabsorptive procedures.
What are special considerations, including candidacy? As noted, Mr. Z expressed concern about the permanence and invasiveness of most bariatric procedures, and therefore wants to undergo a reversible procedure; LAGB can be a reasonable option for such a patient. Patients who want a reversible or minimally invasive procedure should also be made aware that endoscopic bariatric therapies and other devices are being developed to fill the treatment gap in the management of obesity.
What are the complications? Although LAGB is the least invasive procedure discussed here, it is associated with the highest rate of complications—most commonly, complications associated with the band itself (eg, nausea, vomiting, obstruction, band erosion or migration, esophageal dysmotility leading to acid reflux) and failure to lose weight.7 LAGB also requires more postoperative visits than other procedures, to optimize band tightness. A high number of bands are removed eventually because of complications or inadequate weight loss, or both.13,24
Shared decision-making and dialogue are essential to overcome obstacles
Despite the known benefits of bariatric surgery, including greater reduction in the risk and severity of obesity-related comorbid conditions than seen with other interventions and a long-term reduction in overall mortality when compared with usual care, fewer than 1% of eligible patients undergo a weight-loss procedure.25 Likely, this is due to:
- limited patient knowledge of the health benefits of surgery
- limited provider comfort recommending surgery
- inadequate insurance coverage, which might, in part, be due to a lack of prospective studies comparing various bariatric procedures.18
Continue to: Ultimately, the decision whether to undergo a bariatric procedure...
Ultimately, the decision whether to undergo a bariatric procedure, and which one(s) to consider, should be the product of a thorough conversation between patient and provider.
CORRESPONDENCE
Sarah R. Barenbaum, MD, Department of Internal Medicine, New York–Presbyterian Hospital/Weill Cornell Medical College, 530 East 70th Street, M-507, New York, NY 10021; [email protected]
Patients with overweight and obesity are at increased risk of multiple morbidities, including cardiovascular disease, stroke, type 2 diabetes (T2D), osteoarthritis, obstructive sleep apnea (OSA), and all-cause mortality.1 Even modest weight loss—5% to 10%—can lead to a clinically relevant reduction in this risk of disease.2,3 The American Academy of Family Physicians recognizes obesity as a disease, and recommends screening of all adults for obesity and referral for those with body mass index (BMI)* ≥30 to intensive, multicomponent behavioral interventions.4,5
For some patients, diet, exercise, and behavioral modifications are sufficient; for the great majority, however, weight loss achieved by lifestyle modification is counteracted by metabolic adaptations that promote weight regain.6 For patients with obesity who are unable to achieve or maintain sufficient weight loss to improve health outcomes with lifestyle modification alone, options include pharmacotherapy, devices, endoscopic bariatric therapies, and bariatric surgery.
Bariatric surgery is the most effective of these treatments, due to its association with significant and sustained weight loss, reduction in obesity-related comorbidities, and improved quality of life.1,7 Furthermore, compared with usual care, bariatric surgery is associated with a reduced number of cardiovascular deaths, a lower incidence of cardiovascular events in adults with obesity, and a long-term reduction in overall mortality.8-10
What are the options? Who is a candidate?
The 3 most common bariatric procedures in the United States are sleeve gastrectomy (SG), Roux-en-Y gastric bypass (RYGB), and laparoscopic adjustable gastric band (LAGB).11 SG and RYGB are performed more often than the LAGB, consequent to greater efficacy and fewer complications.12 Weight loss is maximal at 1 to 2 years, and is estimated to be 15% of total body weight for LAGB; 25% for SG; and 35% for RYGB.13,14
Not all patients are candidates for bariatric surgery. Contraindications include chronic obstructive pulmonary disease or respiratory dysfunction, poor cardiac reserve, nonadherence to medical treatment, and severe psychological disorders.15 Because some patients have difficulty maintaining weight loss following bariatric surgery and, on average, patients regain at least some weight, patients must understand that long-term lifestyle changes and follow-up are critical to the success of bariatric surgery.16
When should bariatric surgery be considered?
American Heart Association/American College of Cardiology/The Obesity Society guidelines16 conceptualize 2 indications for bariatric surgery:
- adults with BMI ≥40
- adults with BMI ≥35 who have obesity-related comorbid conditions and are motivated to lose weight but have not responded to behavioral treatment, with or without pharmacotherapy, to achieve sufficient weight loss for target health goals.
American Association of Clinical Endocrinologists guidelines17 conceptualize 3 indications for bariatric surgery:
- adults with BMI ≥40
- adults with BMI ≥35 with 1 or more severe obesity-related complications
- adults with BMI 30-34.9 with diabetes or metabolic syndrome (evidence for this recommendation is limited).
Continue to: The 3 illustrative vignettes presented...
The 3 illustrative vignettes presented in this article offer examples of patients with obesity who could benefit from bariatric surgery. Each has been unable to achieve or maintain sufficient weight loss to improve health outcomes with nonsurgical interventions alone.
CASE 1
Sleep apnea persists despite weight loss
Robin W, a 50-year-old woman with class-II obesity (5’8”; 250 lb; BMI, 38 ), OSA requiring continuous positive airway pressure (CPAP), hyperlipidemia, hypertension, and iron-deficiency anemia secondary to menorrhagia, and taking an iron supplement, presents for weight management. She has lost 50 lb, reducing her BMI from 45.6 with behavioral modifications and pharmacotherapy, but she has been unsuccessful at achieving further weight loss despite a reduced-calorie diet and at least 30 minutes of physical activity most days.
Ms. W is frustrated that she has reached a weight plateau; she is motivated to lose more weight. Her goal is to improve her weight-related comorbid conditions and reduce her medication requirement. Despite the initial weight loss, she continues to require CPAP therapy for OSA and remains on 3 medications for hypertension. She does not have cardiac or respiratory disease, psychiatric diagnoses, or a history of gastroesophageal reflux disease (GERD).
Is bariatric surgery a reasonable option for Ms. W? If so, which procedure would you recommend?
Good option for Ms. W: Sleeve gastrectomy
It is reasonable to consider bariatric surgery—in particular, SG—for this patient with class-II obesity and multiple weight-related comorbid conditions because she has been unable to achieve further weight loss with more conservative measures.
Continue to: How does the procedure work?
How does the procedure work? SG removes a large portion of the stomach along the greater curvature, reducing the organ to approximately 15% to 25% of its original size.18 The procedure leaves the pyloric valve intact and does not involve removal or bypass of the intestines.
How appealing and successful is it? The majority of patients who undergo SG experience significant weight loss; studies report approximately 25% total body weight loss after 1 to 2 years.14 Furthermore, most patients with T2D experience resolution of, or improvement in, disease markers.19 Because SG leaves the pylorus intact, there are fewer restrictions on what a patient can eat after surgery, compared with RYGB. With further weight loss, Ms. W may experience improvement in, or resolution of, hypertension, hyperlipidemia, and OSA.
The SG procedure itself is simpler than some other bariatric procedures and presents less risk of malabsorption because the intestines are left intact. Patients who undergo SG report feeling less hungry because the fundus of the stomach, which secretes ghrelin (the so-called hunger hormone), is removed.18,20
What are special considerations, including candidacy? Patients with GERD are not ideal candidates for this procedure because exacerbation of the disease is a potential associated adverse event. SG is a reasonable surgical option for Ms. W because the procedure is less likely to exacerbate her nutritional deficiency (iron-deficiency anemia), compared to RYGB, and she does not have a history of GERD.
What are the complications? Complications of SG occur at a lower rate than they do with RYGB, which is associated with a greater risk of nutritional deficiency.18 Common early complications of SG include leaking, bleeding, stenosis, GERD, and vomiting due to excessive eating. Late complications include stomach expansion by 12 months, leading to decreased restriction.15 Unlike RYGB and LAGB, SG is not reversible.
Continue to: CASE 2
CASE 2
Severe obesity, polypharmacy for type 2 diabetes
Anne P, a 42-year-old woman with class-III obesity (5’6”; 290 lb; BMI, 46.8 kg/m2), presents to discuss bariatric surgery. Comorbidities include T2D, for which she takes metformin, a glucagon-like peptide-1 (GLP-1) receptor agonist, and a sodium–glucose cotransporter-2 (SGLT-2) inhibitor; GERD; hypertension, for which she takes an angiotensin-converting enzyme inhibitor and a calcium-channel blocker; hyperlipidemia, for which she takes a statin; and osteoarthritis.
Ms. P lost 30 pounds—reducing her BMI from 51.6—when the sulfonylurea and thiazolidinedione she was taking were switched to the GLP-1 receptor agonist and the SGLT2 inhibitor. She also made behavioral modifications, including 30 minutes a day of physical activity and a reduced-calorie meal plan under the guidance of a dietitian.
However, Ms. P has been unable to lose more weight or reduce her hemoglobin A1c (HbA1c) level below 8%. Her goal is to avoid the need to take insulin (which several members of her family take), lower her HbA1c level, and decrease her medication requirement.
Ms. P does not have cardiac or respiratory disease or psychiatric diagnoses. Which surgical intervention would you recommend for her?
Good option for Ms. P: Roux-en-Y gastric bypass
RYGB is a reasonable option for a patient with class-III obesity and multiple comorbidities, including poorly controlled T2D and GERD, who has failed conservative measures but wants to lose more weight, reduce her HbA1c, reduce her medication requirement, and avoid the need for insulin.
Continue to: How does the procedure work?
How does the procedure work? RYGB constructs a small pouch from the proximal portion of the stomach and attaches it directly to the jejunum, thus bypassing part of the stomach and duodenum. The procedure is effective for weight loss because it is both restrictive and malabsorptive: patients not only eat smaller portions, but cannot absorb all they eat. Other mechanisms attributed to RYGB that are hypothesized to promote weight loss include21:
- alteration of endogenous gut hormones, which promotes postprandial satiety
- increased levels of bile acids, which promotes alteration of the gut microbiome
- intestinal hypertrophy.
How successful is it? RYGB is associated with significant total body weight loss of approximately 35% at 2 years.9 The procedure has been shown to produce superior outcomes in reducing comorbid disease compared to other bariatric procedures or medical therapy alone. Of the procedures discussed in this article, RYGB is associated with the greatest reduction in triglycerides, HbA1c, and use of diabetes medications, including insulin.22
What are special considerations, including candidacy? For patients with mild or moderate T2D (calculated using the Individualized Metabolic Surgery Score [http://riskcalc.org/Metabolic_Surgery_Score/], which categorizes patients by number of diabetes medications, insulin use, duration of diabetes before surgery, and HbA1c), RYGB is recommended over SG because it leads to greater long-term remission of T2D.
RYGB is associated with a lower rate of GERD than SG and can even alleviate GERD in patients who have the disease. Furthermore, for patients with limited pancreatic beta cell reserve, RYBG and SG have similarly low efficacy for T2D remission; SG is therefore recommended over RYGB in this specific circumstance, given its slightly lower risk profile.23
What are the complications? Patients who undergo any bariatric surgical procedure require long-term follow-up and vitamin supplementation, but those who undergo RYGB require stricter dietary adherence after the procedure; lifelong vitamin (D, B12, folic acid, and thiamine), iron, and calcium supplementation; and long-term follow-up to reduce the risk and severity of complications and to monitor for nutritional deficiencies.7 As such, patients who have shown poor adherence to medical treatment are not good candidates for the procedure.
Continue to: Early complications include...
Early complications include leak, stricture, obstruction, and failure of the staple partition of the upper stomach. Late complications include nutritional deficiencies, as noted, and ulceration of the anastomosis. Dumping syndrome (overly rapid transit of food from the stomach into the small intestine) can develop early or late; early dumping leads to osmotic diarrhea and abdominal cramping, and late dumping leads to reactive hypoglycemia.15
Technically, RYGB is a reversible procedure, although generally it is reversed only in extreme circumstances.
CASE 3
Fatty liver disease, hesitation to undergo surgery
Walt Z, a 35 year-old-man with class-II obesity (5’10”; 265 lb; BMI, 38 kg/m2), T2D, and hepatic steatosis, presents for weight management. He has been able to lose modest weight over the years with behavioral modifications, but has been unsuccessful in maintaining that loss. He requests referral to a bariatric surgeon but is concerned about the permanence and invasiveness of most bariatric procedures.
Which surgical intervention would you recommend for this patient?
Good option for Mr. Z: Laparoscopic adjustable gastric band
Given that Mr. Z is a candidate for a surgical intervention but does not want a permanent or invasive procedure, LAGB is a reasonable option.
Continue to: How does the procedure work?
How does the procedure work? LAGB is a reversible procedure in which an inflatable band is placed around the fundus of the stomach to create a small pouch. The band can be adjusted to regulate food intake by adding or removing saline through a subcutaneous access port.
How appealing and successful is it? LAGB results in approximately 15% total body weight loss at 2 years.13 Because the procedure is purely restrictive, it carries a reduced risk of nutritional deficiency associated more commonly with malabsorptive procedures.
What are special considerations, including candidacy? As noted, Mr. Z expressed concern about the permanence and invasiveness of most bariatric procedures, and therefore wants to undergo a reversible procedure; LAGB can be a reasonable option for such a patient. Patients who want a reversible or minimally invasive procedure should also be made aware that endoscopic bariatric therapies and other devices are being developed to fill the treatment gap in the management of obesity.
What are the complications? Although LAGB is the least invasive procedure discussed here, it is associated with the highest rate of complications—most commonly, complications associated with the band itself (eg, nausea, vomiting, obstruction, band erosion or migration, esophageal dysmotility leading to acid reflux) and failure to lose weight.7 LAGB also requires more postoperative visits than other procedures, to optimize band tightness. A high number of bands are removed eventually because of complications or inadequate weight loss, or both.13,24
Shared decision-making and dialogue are essential to overcome obstacles
Despite the known benefits of bariatric surgery, including greater reduction in the risk and severity of obesity-related comorbid conditions than seen with other interventions and a long-term reduction in overall mortality when compared with usual care, fewer than 1% of eligible patients undergo a weight-loss procedure.25 Likely, this is due to:
- limited patient knowledge of the health benefits of surgery
- limited provider comfort recommending surgery
- inadequate insurance coverage, which might, in part, be due to a lack of prospective studies comparing various bariatric procedures.18
Continue to: Ultimately, the decision whether to undergo a bariatric procedure...
Ultimately, the decision whether to undergo a bariatric procedure, and which one(s) to consider, should be the product of a thorough conversation between patient and provider.
CORRESPONDENCE
Sarah R. Barenbaum, MD, Department of Internal Medicine, New York–Presbyterian Hospital/Weill Cornell Medical College, 530 East 70th Street, M-507, New York, NY 10021; [email protected]
1. Must A, Spadano J, Coakley EH, et al. The disease burden associated with overweight and obesity. JAMA. 1999;282:1523-1529.
2. Wing RR, Lang W, Wadden TA, et al. Benefits of modest weight loss in improving cardiovascular risk factors in overweight and obese individuals with type 2 diabetes. Diabetes Care. 2011;34:1481-1486.
3. Magkos F, Fraterrigo G, Yoshino J, et al. Effects of moderate and subsequent progressive weight loss on metabolic function and adipose tissue biology in humans with obesity. Cell Metab. 2016;23:591-601.
4. American Academy of Family Physicians. Clinical preventive service recommendation: Obesity. www.aafp.org/patient-care/clinical-recommendations/all/obesity.html. Accessed August 22, 2018.
5. American Academy of Family Physicians: USPSTF draft recommendation: Intensive behavioral interventions recommended for obesity. www.aafp.org/news/health-of-the-public/20180221uspstfobesity.html. Published February 21, 2018. Accessed August 22, 2018.
6. Saunders KH, Shukla AP, Igel LI, Aronne LJ. Obesity: When to consider medication. J Fam Pract. 2017;66:608-616.
7. Roux CW, Heneghan HM. Bariatric surgery for obesity. Med Clin North Am. 2018;102:165-182.
8. Sjöström L, Peltonen M, Jacobson P, et al. Bariatric surgery and long-term cardiovascular events. JAMA. 2012;307:56-65.
9. Sjöström L. Review of the key results from the Swedish Obese Subjects (SOS) trial - a prospective controlled intervention study of bariatric surgery. J Intern Med. 2013;273:219-234.
10. Reges O, Greenland P, Dicker D, et al. Association of bariatric surgery using laparoscopic banding, Roux-en-Y, gastric bypass, or laparoscopic sleeve gastrectomy vs usual care obesity management with all-cause mortality. JAMA. 2018;319:279-290.
11. Lee JH, Nguyen QN, Le QA. Comparative effectiveness of 3 bariatric surgery procedures: Roux-en-Y gastric bypass, laparoscopic adjustable gastric band, and sleeve gastrectomy. Surg Obes Relat Dis. 2016;12:997-1002.
12. American Society for Metabolic and Bariatric Surgery. Estimate of bariatric surgery numbers, 2011-2017. https://asmbs.org/resources/estimate-of-bariatric-surgery-numbers. Published June 2018. Accessed August 22, 2018.
13. Courcoulas AP, King WC, Belle SH, et al. Seven-year weight trajectories and health outcomes in the Longitudinal Assessment of Bariatric Surgery (LABS) Study. JAMA Surg. 2018;153:427-434.
14. Heymsfield SB, Wadden TA. Mechanisms, pathophysiology, and management of obesity. N Engl J Med. 2017;376:254-266.
15. Colquitt JL, Pickett K, Loveman E, Frampton GK. Surgery for weight loss in adults. Cochrane Database Syst Rev. 2014;(8):CD003641.
16. Jensen MD, Ryan DH, Apovian CM, et al; American College of Cardiology/American Heart Association Task Force on Practice Guidelines; Obesity Society. 2013 AHA/ACC/TOS guideline for the management of overweight and obesity in adults: a report of the American College of Cardiology/American Heart Association Task Force on Practice Guidelines and The Obesity Society. Circulation. 2014;129:S102-S138.
17. Garvey WT, Mechanick JI, Brett EM, et al; Reviewers of the AACE/ACE Obesity Clinical Practice Guidelines. American Association of Clinical Endocrinologists and American College of Endocrinology clinical practice guidelines for comprehensive medical care of patients with obesity. Endocr Pract. 2016;22 Suppl 3:1-203.
18. Carlin Am, Zeni Tm, English WJ, et al; Michigan Bariatric Surgery Collaborative. The comparative effectiveness of sleeve gastrectomy, gastric bypass, and adjustable gastric banding procedures for the treatment of morbid obesity. Ann Surg. 2013;257:791-797.
19. Gill RS, Birch DW, Shi X, et al. Sleeve gastrectomy and type 2 diabetes mellitus: a systematic review. Surg Obes Relat Dis. 2010;6:707-713.
20. Karamanakos SN, Vagenas K, Kalfarentzos F, et al. Weight loss, appetite suppression, and changes in fasting and postprandial ghrelin and peptide-YY levels after Roux-en-Y gastric bypass and sleeve gastrectomy. Ann Surg. 2008;247:401-407.
21. Abdeen G, le Roux CW. Mechanism underlying the weight loss and complications of Roux-en-Y gastric bypass. Obes Surg. 2016;26:410-421.
22. Schauer PR, Bhatt DL, Kirwan JP et al; STAMPEDE Investigators. Bariatric surgery versus intensive medical therapy for diabetes - 5-year outcomes. N Engl J Med. 2017;376:641-651.
23. Aminian A, Brethauer SA, Andalib A, et al. Individualized metabolic surgery score: procedure selection based on diabetes severity. Ann Surg. 2017;266:4:650-657.
24. Smetana GW, Jones DB, Wee CC. Beyond the guidelines: Should this patient have weight loss surgery? Grand rounds discussion from Beth Israel Deaconess Medical Center. Ann Intern Med. 2017;166:808-817.
25. Wolfe BM, Morton JM. Weighing in on bariatric surgery: procedure use, readmission rates, and mortality [editorial]. JAMA. 2005;294:1960-1963.
1. Must A, Spadano J, Coakley EH, et al. The disease burden associated with overweight and obesity. JAMA. 1999;282:1523-1529.
2. Wing RR, Lang W, Wadden TA, et al. Benefits of modest weight loss in improving cardiovascular risk factors in overweight and obese individuals with type 2 diabetes. Diabetes Care. 2011;34:1481-1486.
3. Magkos F, Fraterrigo G, Yoshino J, et al. Effects of moderate and subsequent progressive weight loss on metabolic function and adipose tissue biology in humans with obesity. Cell Metab. 2016;23:591-601.
4. American Academy of Family Physicians. Clinical preventive service recommendation: Obesity. www.aafp.org/patient-care/clinical-recommendations/all/obesity.html. Accessed August 22, 2018.
5. American Academy of Family Physicians: USPSTF draft recommendation: Intensive behavioral interventions recommended for obesity. www.aafp.org/news/health-of-the-public/20180221uspstfobesity.html. Published February 21, 2018. Accessed August 22, 2018.
6. Saunders KH, Shukla AP, Igel LI, Aronne LJ. Obesity: When to consider medication. J Fam Pract. 2017;66:608-616.
7. Roux CW, Heneghan HM. Bariatric surgery for obesity. Med Clin North Am. 2018;102:165-182.
8. Sjöström L, Peltonen M, Jacobson P, et al. Bariatric surgery and long-term cardiovascular events. JAMA. 2012;307:56-65.
9. Sjöström L. Review of the key results from the Swedish Obese Subjects (SOS) trial - a prospective controlled intervention study of bariatric surgery. J Intern Med. 2013;273:219-234.
10. Reges O, Greenland P, Dicker D, et al. Association of bariatric surgery using laparoscopic banding, Roux-en-Y, gastric bypass, or laparoscopic sleeve gastrectomy vs usual care obesity management with all-cause mortality. JAMA. 2018;319:279-290.
11. Lee JH, Nguyen QN, Le QA. Comparative effectiveness of 3 bariatric surgery procedures: Roux-en-Y gastric bypass, laparoscopic adjustable gastric band, and sleeve gastrectomy. Surg Obes Relat Dis. 2016;12:997-1002.
12. American Society for Metabolic and Bariatric Surgery. Estimate of bariatric surgery numbers, 2011-2017. https://asmbs.org/resources/estimate-of-bariatric-surgery-numbers. Published June 2018. Accessed August 22, 2018.
13. Courcoulas AP, King WC, Belle SH, et al. Seven-year weight trajectories and health outcomes in the Longitudinal Assessment of Bariatric Surgery (LABS) Study. JAMA Surg. 2018;153:427-434.
14. Heymsfield SB, Wadden TA. Mechanisms, pathophysiology, and management of obesity. N Engl J Med. 2017;376:254-266.
15. Colquitt JL, Pickett K, Loveman E, Frampton GK. Surgery for weight loss in adults. Cochrane Database Syst Rev. 2014;(8):CD003641.
16. Jensen MD, Ryan DH, Apovian CM, et al; American College of Cardiology/American Heart Association Task Force on Practice Guidelines; Obesity Society. 2013 AHA/ACC/TOS guideline for the management of overweight and obesity in adults: a report of the American College of Cardiology/American Heart Association Task Force on Practice Guidelines and The Obesity Society. Circulation. 2014;129:S102-S138.
17. Garvey WT, Mechanick JI, Brett EM, et al; Reviewers of the AACE/ACE Obesity Clinical Practice Guidelines. American Association of Clinical Endocrinologists and American College of Endocrinology clinical practice guidelines for comprehensive medical care of patients with obesity. Endocr Pract. 2016;22 Suppl 3:1-203.
18. Carlin Am, Zeni Tm, English WJ, et al; Michigan Bariatric Surgery Collaborative. The comparative effectiveness of sleeve gastrectomy, gastric bypass, and adjustable gastric banding procedures for the treatment of morbid obesity. Ann Surg. 2013;257:791-797.
19. Gill RS, Birch DW, Shi X, et al. Sleeve gastrectomy and type 2 diabetes mellitus: a systematic review. Surg Obes Relat Dis. 2010;6:707-713.
20. Karamanakos SN, Vagenas K, Kalfarentzos F, et al. Weight loss, appetite suppression, and changes in fasting and postprandial ghrelin and peptide-YY levels after Roux-en-Y gastric bypass and sleeve gastrectomy. Ann Surg. 2008;247:401-407.
21. Abdeen G, le Roux CW. Mechanism underlying the weight loss and complications of Roux-en-Y gastric bypass. Obes Surg. 2016;26:410-421.
22. Schauer PR, Bhatt DL, Kirwan JP et al; STAMPEDE Investigators. Bariatric surgery versus intensive medical therapy for diabetes - 5-year outcomes. N Engl J Med. 2017;376:641-651.
23. Aminian A, Brethauer SA, Andalib A, et al. Individualized metabolic surgery score: procedure selection based on diabetes severity. Ann Surg. 2017;266:4:650-657.
24. Smetana GW, Jones DB, Wee CC. Beyond the guidelines: Should this patient have weight loss surgery? Grand rounds discussion from Beth Israel Deaconess Medical Center. Ann Intern Med. 2017;166:808-817.
25. Wolfe BM, Morton JM. Weighing in on bariatric surgery: procedure use, readmission rates, and mortality [editorial]. JAMA. 2005;294:1960-1963.
PRACTICE RECOMMENDATIONS
Among adult patients with body mass index* ≥40, or ≥35 with obesity-related comorbid conditions:
› Consider bariatric surgery in those who are motivated to lose weight but who have not responded to lifestyle modification with or without pharmacotherapy in order to achieve sufficient and sustained weight loss. A
› Consider bariatric surgery to help patients achieve target health goals and reduce/improve obesity-related comorbidities. 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
*Calculated as weight in kilograms divided by height in meters squared.
Researchers share Nobel Prize for cancer immunotherapy discoveries
Two researchers who made groundbreaking discoveries leading to the development of immune checkpoint therapy have jointly received the 2018 Nobel Prize in Physiology or Medicine.
James P. Allison, PhD, and Tasuku Honjo, MD, PhD, share the prize for their work in showing how strategies for “inhibiting the brakes” on T cells can be used in the treatment of cancer, the Nobel Assembly at Karolinska Institutet, Stockholm, said in a news release.
The discoveries of Dr. Allison and Dr. Honjo represent a landmark in the fight against cancer, according to the Nobel Assembly.
“This new form of immunotherapy unleashes a vigorous, and often durable, immune response directed against essentially any tumor already recognized by the immune system,” members of the Nobel Committee and Assembly said in a simultaneously published manuscript explaining the scientific background behind the discovery.
The “unprecedented” research activity in the immune checkpoint field demonstrates how influential the discoveries of Dr. Allison and Dr. Honjo have been, molecular genetics professor C. I. Edvard Smith, MD, PhD, and coauthors said in the manuscript.
Their findings have “conferred great benefit on mankind” and add a “new pillar” to existing cancer treatments, Dr. Smith and coauthors said in their report.
Dr. Allison was one of several scientists to observe that the cytotoxic T-lymphocyte antigen 4 (CTLA4) protein functions as a brake on T cells, the Nobel Assembly said in the press release. He developed an antibody that could bind to CTLA4 and block its function, and demonstrated its activity in experiments taking place near the end of 1994.
The “spectacular” results showed that, in mice, the antibodies could inhibit the brake and unlock antitumor T-cell activity, according to the release.
Studies of CTLA-4 blockade in humans from several research groups yielded promising results, and in 2010, a major breakthrough came, according to the Nobel Assembly, when it was reported that the CTLA4 blocker ipilimumab improved survival in patients with metastatic melanoma.
Dr. Honjo discovered the programmed cell death protein (PD-1) in 1992, a few years before Dr. Allison’s discovery, and explored its function in experiments carried out over many years at his laboratory at Kyoto University.
Dr. Honjo’s experiments showed that PD-1 functioned as a T-cell brake, but by a different mechanism than CTLA4, paving the way for studying PD-1 as a target of cancer treatment.
That led to a considerable amount of clinical development, including a key 2012 study showing that patients treated with the anti-PD-1 antibody nivolumab led to long-term remissions and possible cures in some patients, the assembly said in their press release.
Scientists have attempted to harness the immune system in the fight against cancer for more than 100 years, according to the assembly. “Until the seminal discoveries by the two laureates, progress into clinical development was modest. Checkpoint therapy has now revolutionized cancer treatment and has fundamentally changed the way we view how cancer can be managed.”
Two researchers who made groundbreaking discoveries leading to the development of immune checkpoint therapy have jointly received the 2018 Nobel Prize in Physiology or Medicine.
James P. Allison, PhD, and Tasuku Honjo, MD, PhD, share the prize for their work in showing how strategies for “inhibiting the brakes” on T cells can be used in the treatment of cancer, the Nobel Assembly at Karolinska Institutet, Stockholm, said in a news release.
The discoveries of Dr. Allison and Dr. Honjo represent a landmark in the fight against cancer, according to the Nobel Assembly.
“This new form of immunotherapy unleashes a vigorous, and often durable, immune response directed against essentially any tumor already recognized by the immune system,” members of the Nobel Committee and Assembly said in a simultaneously published manuscript explaining the scientific background behind the discovery.
The “unprecedented” research activity in the immune checkpoint field demonstrates how influential the discoveries of Dr. Allison and Dr. Honjo have been, molecular genetics professor C. I. Edvard Smith, MD, PhD, and coauthors said in the manuscript.
Their findings have “conferred great benefit on mankind” and add a “new pillar” to existing cancer treatments, Dr. Smith and coauthors said in their report.
Dr. Allison was one of several scientists to observe that the cytotoxic T-lymphocyte antigen 4 (CTLA4) protein functions as a brake on T cells, the Nobel Assembly said in the press release. He developed an antibody that could bind to CTLA4 and block its function, and demonstrated its activity in experiments taking place near the end of 1994.
The “spectacular” results showed that, in mice, the antibodies could inhibit the brake and unlock antitumor T-cell activity, according to the release.
Studies of CTLA-4 blockade in humans from several research groups yielded promising results, and in 2010, a major breakthrough came, according to the Nobel Assembly, when it was reported that the CTLA4 blocker ipilimumab improved survival in patients with metastatic melanoma.
Dr. Honjo discovered the programmed cell death protein (PD-1) in 1992, a few years before Dr. Allison’s discovery, and explored its function in experiments carried out over many years at his laboratory at Kyoto University.
Dr. Honjo’s experiments showed that PD-1 functioned as a T-cell brake, but by a different mechanism than CTLA4, paving the way for studying PD-1 as a target of cancer treatment.
That led to a considerable amount of clinical development, including a key 2012 study showing that patients treated with the anti-PD-1 antibody nivolumab led to long-term remissions and possible cures in some patients, the assembly said in their press release.
Scientists have attempted to harness the immune system in the fight against cancer for more than 100 years, according to the assembly. “Until the seminal discoveries by the two laureates, progress into clinical development was modest. Checkpoint therapy has now revolutionized cancer treatment and has fundamentally changed the way we view how cancer can be managed.”
Two researchers who made groundbreaking discoveries leading to the development of immune checkpoint therapy have jointly received the 2018 Nobel Prize in Physiology or Medicine.
James P. Allison, PhD, and Tasuku Honjo, MD, PhD, share the prize for their work in showing how strategies for “inhibiting the brakes” on T cells can be used in the treatment of cancer, the Nobel Assembly at Karolinska Institutet, Stockholm, said in a news release.
The discoveries of Dr. Allison and Dr. Honjo represent a landmark in the fight against cancer, according to the Nobel Assembly.
“This new form of immunotherapy unleashes a vigorous, and often durable, immune response directed against essentially any tumor already recognized by the immune system,” members of the Nobel Committee and Assembly said in a simultaneously published manuscript explaining the scientific background behind the discovery.
The “unprecedented” research activity in the immune checkpoint field demonstrates how influential the discoveries of Dr. Allison and Dr. Honjo have been, molecular genetics professor C. I. Edvard Smith, MD, PhD, and coauthors said in the manuscript.
Their findings have “conferred great benefit on mankind” and add a “new pillar” to existing cancer treatments, Dr. Smith and coauthors said in their report.
Dr. Allison was one of several scientists to observe that the cytotoxic T-lymphocyte antigen 4 (CTLA4) protein functions as a brake on T cells, the Nobel Assembly said in the press release. He developed an antibody that could bind to CTLA4 and block its function, and demonstrated its activity in experiments taking place near the end of 1994.
The “spectacular” results showed that, in mice, the antibodies could inhibit the brake and unlock antitumor T-cell activity, according to the release.
Studies of CTLA-4 blockade in humans from several research groups yielded promising results, and in 2010, a major breakthrough came, according to the Nobel Assembly, when it was reported that the CTLA4 blocker ipilimumab improved survival in patients with metastatic melanoma.
Dr. Honjo discovered the programmed cell death protein (PD-1) in 1992, a few years before Dr. Allison’s discovery, and explored its function in experiments carried out over many years at his laboratory at Kyoto University.
Dr. Honjo’s experiments showed that PD-1 functioned as a T-cell brake, but by a different mechanism than CTLA4, paving the way for studying PD-1 as a target of cancer treatment.
That led to a considerable amount of clinical development, including a key 2012 study showing that patients treated with the anti-PD-1 antibody nivolumab led to long-term remissions and possible cures in some patients, the assembly said in their press release.
Scientists have attempted to harness the immune system in the fight against cancer for more than 100 years, according to the assembly. “Until the seminal discoveries by the two laureates, progress into clinical development was modest. Checkpoint therapy has now revolutionized cancer treatment and has fundamentally changed the way we view how cancer can be managed.”
The oncologist’s dilemma
The textbook answer would have been straightforward: No additional chemotherapy. Focus on palliative care.
But Max (not his real name) was not a textbook case. He was a person, and he was terrified of dying.
When I met him in clinic, four lines of chemotherapy had not slowed the spread of his rare cancer that was already metastatic at the time of diagnosis. His skin was yellow. His hair had fallen out in clumps. His legs were swollen to the point that he couldn’t walk. He had trouble transferring from his wheelchair on his own.
All this was offset by the college hoodie he wore, a disarming display of his youth. I had to look at the chart to remind myself. He was only 19 years old.
“Will Dr. D give Max chemotherapy?” his mother asked me, referring to the attending oncologist who had been caring for her son since the beginning. “I think he needs it right away,” she said. She was holding back tears.
Protected by the fact that I was a visiting fellow in her clinic for the day, and I was just meeting Max, I deferred the decision to Dr. D.
Dr. D and I reviewed Max’s case outside the room, scrolling through his PET scans showing spread of cancer in his liver, lungs, and bones in only 2 short months. We recounted the multiple lines of chemotherapy and immunotherapy he had tried and that had failed him.
Palliative care? I offered. She agreed.
But as we went back in together, it was harder. Max’s mother began to cry as Dr. D tried to broach the option. To her, palliative care meant death. That was not something she could swallow. Her questions turned back only to the next chemotherapy we would be giving.
I learned that Max’s father worked in a hospital. He knew how serious it was. “He calls me every day, crying,” my attending later told me solemnly. Begging her to do something. Pleading for more chemotherapy.
After some painful back and forth in the room, we didn’t come to a resolution. Dr. D would call them later, she said.
It was a busy clinic day, and we saw the rest of her patients.
“What are we going to do about Max?” I asked at the end of the day. I was writing the note in his chart. We still didn’t have a plan.
More chemotherapy, from any technical and data-based standpoint, was not the right choice. We had no evidence it would improve survival. We did have evidence that it could worsen the quality of life when time was limited. If we gave him more chemotherapy, it would be beyond guidelines, beyond evidence. We would be off the grid.
I thought about the conversations I’d been privy to about giving toxic therapies near the end of life. While a handful felt productive, others were uncomfortably strained. The latter involved a tension that manifests when the goals of the patient and the oncologist are misaligned. The words may be slightly different each time, but the theme is the same.
The oncologist says: “More chemotherapy is not going to work.”
The patient says: “But we don’t know. It’s better than doing nothing. Can’t we try?”
This can be excruciatingly challenging because both sides are correct. Both sides are logical, and yet they are talking past each other.
From the point of view of the person desperate to survive, anything is worth a try. Without trying some form of treatment, there’s a zero percent chance of surviving. Low odds of something working are still better than zero percent odds.
But it’s not an issue of logic. For people like Max’s parents, the questions come from a place of helplessness. The cognitive dissonance sets in because our job is to help and we struggle when we feel we cannot. We want to say yes. I wish we had a treatment to slow Max’s cancer, too. I wish more chemotherapy would help him.
What I’ve learned from these conversations is the importance of defining terms. Specifically, what do we mean when we say a treatment will or will not “work”? What are we trying to achieve? To prolong life by weeks? By months or years? To make your pain go away? To help you feel stronger? To allow you to spend time doing what you love?
The relevant question is not: Will this treatment work? It is: What are your goals, and will this treatment help you achieve them?
Defining terms in that way can mean the difference of a conversation where two sides are talking past one another to one that comes to a mutual understanding – even if the ultimate conclusion is painful for everyone.
Yet sometimes, even for the most skilled communicators, there may be compromise. There may be an agreement to trial Nth-line chemotherapy, even without evidence that it will achieve a person’s goals. It will require nuanced informed consent from both sides. And it will come from a place of compassion.
This is what I thought about as I signed chemotherapy orders for Max that evening. I felt for him, for his parents, and for his oncologist.
Because even though I suspected the treatment wouldn’t work the way they all hoped it would, I also wasn’t the one fielding daily phone calls from a father, begging his oncologist to do something, anything at all, to save his son’s life.
Dr. Yurkiewicz is a fellow in hematology and oncology at Stanford (Calif.) University. Follow her on Twitter @ilanayurkiewicz.
The textbook answer would have been straightforward: No additional chemotherapy. Focus on palliative care.
But Max (not his real name) was not a textbook case. He was a person, and he was terrified of dying.
When I met him in clinic, four lines of chemotherapy had not slowed the spread of his rare cancer that was already metastatic at the time of diagnosis. His skin was yellow. His hair had fallen out in clumps. His legs were swollen to the point that he couldn’t walk. He had trouble transferring from his wheelchair on his own.
All this was offset by the college hoodie he wore, a disarming display of his youth. I had to look at the chart to remind myself. He was only 19 years old.
“Will Dr. D give Max chemotherapy?” his mother asked me, referring to the attending oncologist who had been caring for her son since the beginning. “I think he needs it right away,” she said. She was holding back tears.
Protected by the fact that I was a visiting fellow in her clinic for the day, and I was just meeting Max, I deferred the decision to Dr. D.
Dr. D and I reviewed Max’s case outside the room, scrolling through his PET scans showing spread of cancer in his liver, lungs, and bones in only 2 short months. We recounted the multiple lines of chemotherapy and immunotherapy he had tried and that had failed him.
Palliative care? I offered. She agreed.
But as we went back in together, it was harder. Max’s mother began to cry as Dr. D tried to broach the option. To her, palliative care meant death. That was not something she could swallow. Her questions turned back only to the next chemotherapy we would be giving.
I learned that Max’s father worked in a hospital. He knew how serious it was. “He calls me every day, crying,” my attending later told me solemnly. Begging her to do something. Pleading for more chemotherapy.
After some painful back and forth in the room, we didn’t come to a resolution. Dr. D would call them later, she said.
It was a busy clinic day, and we saw the rest of her patients.
“What are we going to do about Max?” I asked at the end of the day. I was writing the note in his chart. We still didn’t have a plan.
More chemotherapy, from any technical and data-based standpoint, was not the right choice. We had no evidence it would improve survival. We did have evidence that it could worsen the quality of life when time was limited. If we gave him more chemotherapy, it would be beyond guidelines, beyond evidence. We would be off the grid.
I thought about the conversations I’d been privy to about giving toxic therapies near the end of life. While a handful felt productive, others were uncomfortably strained. The latter involved a tension that manifests when the goals of the patient and the oncologist are misaligned. The words may be slightly different each time, but the theme is the same.
The oncologist says: “More chemotherapy is not going to work.”
The patient says: “But we don’t know. It’s better than doing nothing. Can’t we try?”
This can be excruciatingly challenging because both sides are correct. Both sides are logical, and yet they are talking past each other.
From the point of view of the person desperate to survive, anything is worth a try. Without trying some form of treatment, there’s a zero percent chance of surviving. Low odds of something working are still better than zero percent odds.
But it’s not an issue of logic. For people like Max’s parents, the questions come from a place of helplessness. The cognitive dissonance sets in because our job is to help and we struggle when we feel we cannot. We want to say yes. I wish we had a treatment to slow Max’s cancer, too. I wish more chemotherapy would help him.
What I’ve learned from these conversations is the importance of defining terms. Specifically, what do we mean when we say a treatment will or will not “work”? What are we trying to achieve? To prolong life by weeks? By months or years? To make your pain go away? To help you feel stronger? To allow you to spend time doing what you love?
The relevant question is not: Will this treatment work? It is: What are your goals, and will this treatment help you achieve them?
Defining terms in that way can mean the difference of a conversation where two sides are talking past one another to one that comes to a mutual understanding – even if the ultimate conclusion is painful for everyone.
Yet sometimes, even for the most skilled communicators, there may be compromise. There may be an agreement to trial Nth-line chemotherapy, even without evidence that it will achieve a person’s goals. It will require nuanced informed consent from both sides. And it will come from a place of compassion.
This is what I thought about as I signed chemotherapy orders for Max that evening. I felt for him, for his parents, and for his oncologist.
Because even though I suspected the treatment wouldn’t work the way they all hoped it would, I also wasn’t the one fielding daily phone calls from a father, begging his oncologist to do something, anything at all, to save his son’s life.
Dr. Yurkiewicz is a fellow in hematology and oncology at Stanford (Calif.) University. Follow her on Twitter @ilanayurkiewicz.
The textbook answer would have been straightforward: No additional chemotherapy. Focus on palliative care.
But Max (not his real name) was not a textbook case. He was a person, and he was terrified of dying.
When I met him in clinic, four lines of chemotherapy had not slowed the spread of his rare cancer that was already metastatic at the time of diagnosis. His skin was yellow. His hair had fallen out in clumps. His legs were swollen to the point that he couldn’t walk. He had trouble transferring from his wheelchair on his own.
All this was offset by the college hoodie he wore, a disarming display of his youth. I had to look at the chart to remind myself. He was only 19 years old.
“Will Dr. D give Max chemotherapy?” his mother asked me, referring to the attending oncologist who had been caring for her son since the beginning. “I think he needs it right away,” she said. She was holding back tears.
Protected by the fact that I was a visiting fellow in her clinic for the day, and I was just meeting Max, I deferred the decision to Dr. D.
Dr. D and I reviewed Max’s case outside the room, scrolling through his PET scans showing spread of cancer in his liver, lungs, and bones in only 2 short months. We recounted the multiple lines of chemotherapy and immunotherapy he had tried and that had failed him.
Palliative care? I offered. She agreed.
But as we went back in together, it was harder. Max’s mother began to cry as Dr. D tried to broach the option. To her, palliative care meant death. That was not something she could swallow. Her questions turned back only to the next chemotherapy we would be giving.
I learned that Max’s father worked in a hospital. He knew how serious it was. “He calls me every day, crying,” my attending later told me solemnly. Begging her to do something. Pleading for more chemotherapy.
After some painful back and forth in the room, we didn’t come to a resolution. Dr. D would call them later, she said.
It was a busy clinic day, and we saw the rest of her patients.
“What are we going to do about Max?” I asked at the end of the day. I was writing the note in his chart. We still didn’t have a plan.
More chemotherapy, from any technical and data-based standpoint, was not the right choice. We had no evidence it would improve survival. We did have evidence that it could worsen the quality of life when time was limited. If we gave him more chemotherapy, it would be beyond guidelines, beyond evidence. We would be off the grid.
I thought about the conversations I’d been privy to about giving toxic therapies near the end of life. While a handful felt productive, others were uncomfortably strained. The latter involved a tension that manifests when the goals of the patient and the oncologist are misaligned. The words may be slightly different each time, but the theme is the same.
The oncologist says: “More chemotherapy is not going to work.”
The patient says: “But we don’t know. It’s better than doing nothing. Can’t we try?”
This can be excruciatingly challenging because both sides are correct. Both sides are logical, and yet they are talking past each other.
From the point of view of the person desperate to survive, anything is worth a try. Without trying some form of treatment, there’s a zero percent chance of surviving. Low odds of something working are still better than zero percent odds.
But it’s not an issue of logic. For people like Max’s parents, the questions come from a place of helplessness. The cognitive dissonance sets in because our job is to help and we struggle when we feel we cannot. We want to say yes. I wish we had a treatment to slow Max’s cancer, too. I wish more chemotherapy would help him.
What I’ve learned from these conversations is the importance of defining terms. Specifically, what do we mean when we say a treatment will or will not “work”? What are we trying to achieve? To prolong life by weeks? By months or years? To make your pain go away? To help you feel stronger? To allow you to spend time doing what you love?
The relevant question is not: Will this treatment work? It is: What are your goals, and will this treatment help you achieve them?
Defining terms in that way can mean the difference of a conversation where two sides are talking past one another to one that comes to a mutual understanding – even if the ultimate conclusion is painful for everyone.
Yet sometimes, even for the most skilled communicators, there may be compromise. There may be an agreement to trial Nth-line chemotherapy, even without evidence that it will achieve a person’s goals. It will require nuanced informed consent from both sides. And it will come from a place of compassion.
This is what I thought about as I signed chemotherapy orders for Max that evening. I felt for him, for his parents, and for his oncologist.
Because even though I suspected the treatment wouldn’t work the way they all hoped it would, I also wasn’t the one fielding daily phone calls from a father, begging his oncologist to do something, anything at all, to save his son’s life.
Dr. Yurkiewicz is a fellow in hematology and oncology at Stanford (Calif.) University. Follow her on Twitter @ilanayurkiewicz.
Get on top of home BP monitoring
opioid use in osteoarthritis may mean more activity-limiting pain, sulfasalazine is pinpointed as the reason for high triple-therapy discontinuation, and point-of-care test for respiratory viruses lowers antibiotic use. Also today,
Amazon Alexa
Apple Podcasts
Spotify
opioid use in osteoarthritis may mean more activity-limiting pain, sulfasalazine is pinpointed as the reason for high triple-therapy discontinuation, and point-of-care test for respiratory viruses lowers antibiotic use. Also today,
Amazon Alexa
Apple Podcasts
Spotify
opioid use in osteoarthritis may mean more activity-limiting pain, sulfasalazine is pinpointed as the reason for high triple-therapy discontinuation, and point-of-care test for respiratory viruses lowers antibiotic use. Also today,
Amazon Alexa
Apple Podcasts
Spotify
Office approach to small fiber neuropathy
Peripheral neuropathy is the most common reason for an outpatient neurology visit in the United States and accounts for over $10 billion in healthcare spending each year.1,2 When the disorder affects only small, thinly myelinated or unmyelinated nerve fibers, it is referred to as small fiber neuropathy, which commonly presents as numbness and burning pain in the feet.
This article details the manifestations and evaluation of small fiber neuropathy, with an eye toward diagnosing an underlying cause amenable to treatment.
OLDER PATIENTS MOST AFFECTED
The epidemiology of small fiber neuropathy is not well established. It occurs more commonly in older patients, but data are mixed on prevalence by sex.3–6 In a Dutch study,3 the overall prevalence was at least 53 cases per 100,000, with the highest rate in men over age 65.
CHARACTERISTIC SENSORY DISTURBANCES
Sensations vary in quality and time
Patients with small fiber neuropathy typically present with a symmetric length-dependent (“stocking-glove”) distribution of sensory changes, starting in the feet and gradually ascending up the legs and then to the hands.
Commonly reported neuropathic symptoms include various combinations of burning, numbness, tingling, itching, sunburn-like, and frostbite-like sensations. Nonneuropathic symptoms may include tightness, a vise-like squeezing of the feet, and the sensation of a sock rolled up at the end of the shoe. Cramps or spasms may also be reported but rarely occur in isolation.7
Symptoms are typically worse at the end of the day and while sitting or lying down at night. They can arise spontaneously but may also be triggered by something as minor as the touch of clothing or cool air against the skin. Bedsheet sensitivity of the feet is reported so often that it is used as an outcome measure in clinical trials. Symptoms can also be exacerbated by extremes in ambient temperature and are especially worse in cold weather.
Random patterns suggest an immune cause
Symptoms may also have a non–length-dependent distribution that is asymmetric, patchy, intermittent, and migratory, and can involve the face, proximal limbs, and trunk. Symptoms may vary throughout the day, eg, starting with electric-shock sensations on one side of the face, followed by perineal numbness and then tingling in the arms lasting for a few minutes to several hours. While such patterns may be seen with diabetes and other common etiologies, they often suggest an underlying immune-mediated disorder such as Sjögren syndrome or sarcoidosis.8–10 Although large fiber polyneuropathy may also be non–length-dependent, the deficits are usually fixed, with no migratory component.
Autonomic features may be prominent
Autonomic symptoms occur in nearly half of patients and can be as troublesome as neuropathic pain.3 Small nerve fibers mediate somatic and autonomic functions, an evolutionary link that may reflect visceral defense mechanisms responding to pain as a signal of danger.11 This may help explain the multisystemic nature of symptoms, which can include sweating abnormalities, bowel and bladder disturbances, dry eyes, dry mouth, gastrointestinal dysmotility, skin changes (eg, discoloration, loss of hair, shiny skin), sexual dysfunction, orthostatic hypotension, and palpitations. In some cases, isolated dysautonomia may be seen.
TARGETED EXAMINATION
History: Medications, alcohol, infections
When a patient presents with neuropathic pain in the feet, a detailed history should be obtained, including alcohol use, family history of neuropathy, and use of neurotoxic medications such as metronidazole, colchicine, and chemotherapeutic agents.
Human immunodeficiency virus (HIV) and hepatitis C infection are well known to be associated with small fiber neuropathy, so relevant risk factors (eg, blood transfusions, sexual history, intravenous drug use) should be asked about. Recent illnesses and vaccinations are another important line of questioning, as a small-fiber variant of Guillain-Barré syndrome has been described.12
Assess reflexes, strength, sensation
On physical examination, particular attention should be focused on searching for abnormalities indicating large nerve fiber involvement (eg, absent deep tendon reflexes, weakness of the toes). However, absent ankle deep tendon reflexes and reduced vibratory sense may also occur in healthy elderly people.
Similarly, proprioception, motor strength, balance, and vibratory sensation are functions of large myelinated nerve fibers, and thus remain unaffected in patients with only small fiber neuropathy.
Evidence of a systemic disorder should also be sought, as it may indicate an underlying etiology.
DIAGNOSTIC TESTING
Although patients with either large or small fiber neuropathy may have subjective hyperesthesia or numbness of the distal lower extremities, the absence of significant abnormalities on neurologic examination should prompt consideration of small fiber neuropathy.
Electromyography worthwhile
Nerve conduction studies and needle electrode examination evaluate only large nerve fiber conditions. While electromyographic results are normal in patients with isolated small fiber neuropathy, the test can help evaluate subclinical large nerve fiber involvement and alternative diagnoses such as bilateral S1 radiculopathy. Nerve conduction studies may be less useful in patients over age 75, as they may lack sural sensory responses because of aging changes.13
Skin biopsy easy to do
Skin biopsy for evaluating intraepidermal nerve fiber density is one of the most widely used tests for small fiber neuropathy. This minimally invasive procedure can now be performed in a primary care office using readily available tools or prepackaged kits and analyzed by several commercial laboratories.
Reduced intraepidermal nerve fiber density on skin biopsy has been described in various other conditions such as fibromyalgia and chronic pain syndromes.16,17 The clinical significance of these findings remains uncertain.
Quantitative sudomotor axon reflex testing
Quantitative sudomotor axon reflex testing (QSART) is a noninvasive autonomic study that assesses the volume of sweat produced by the limbs in response to acetylcholine. A measure of postganglionic sympathetic sudomotor nerve function, QSART has a sensitivity of up to 80% and can be used to diagnose small fiber neuropathy.18 In a series of 115 patients with sarcoidosis small fiber neuropathy,9 the QSART and skin biopsy findings were concordant in 17 cases and complementary in 29, allowing for confirmation of small fiber neuropathy in patients whose condition would have remained undiagnosed had only one test been performed. QSART can also be considered in cases where skin biopsy may be contraindicated (eg, patient use of anticoagulation). Of note, the study may be affected by a number of external factors, including caffeine, tobacco, antihistamines, and tricyclic antidepressants; these should be held before testing.
Other diagnostic studies
Other tests may be helpful, as follows:
Tilt-table and cardiovagal testing may be useful for patients with orthostasis and palpitations.
Thermoregulatory sweat testing can be used to evaluate patients with abnormal patterns of sweating, eg, hyperhidrosis of the face and head.
INITIAL TESTING FOR AN UNDERLYING CAUSE
Glucose tolerance test for diabetes
Diabetes is the most common identifiable cause of small fiber neuropathy and accounts for about a third of all cases.5 Impaired glucose tolerance is also thought to be a risk factor and has been found in up to 50% of idiopathic cases, but the association is still being debated.21
While testing for hemoglobin A1c is more convenient for the patient, especially because it does not require fasting, a 2-hour oral glucose tolerance test is more sensitive for detecting glucose dysmetabolism.22
Lipid panel for metabolic syndrome
Small fiber neuropathy is associated with individual components of the metabolic syndrome, which include obesity, hyperglycemia, and dyslipidemia. Of these, dyslipidemia has emerged as the primary factor involved in the development of small fiber neuropathy, via an inflammatory pathway or oxidative stress mechanism.23,24
Vitamin B12 deficiency testing
Vitamin B12 deficiency, a potentially correctable cause of small fiber neuropathy, may be underdiagnosed, especially as values obtained by blood testing may not reflect tissue uptake. Causes of vitamin B12 deficiency include reduced intake, pernicious anemia, and medications that can affect absorption of vitamin B12 (eg, proton pump inhibitors, histamine 2 receptor antagonists, metformin).
Testing should include:
- Complete blood cell count to evaluate for vitamin B12-related macrocytic anemia and other hematologic abnormalities
- Serum vitamin B12 level
- Methylmalonic acid or homocysteine level in patients with subclinical or mild vitamin B12 deficiency, manifested as low to normal vitamin B12 levels (< 400 pg/mL); methylmalonic acid and homocysteine require vitamin B12 as a cofactor for enzymatic conversion, and either or both may be elevated in early vitamin B12 deficiency.
Celiac antibody panel
Celiac disease, a T-cell mediated enteropathy characterized by gluten intolerance and a herpetiform-like rash, can be associated with small fiber neuropathy.25 In some cases, neuropathy symptoms are preceded by the onset of gastrointestinal symptoms, or they may occur in isolation.25
Inflammatory disease testing
Sjögren syndrome accounts for nearly 10% of cases of small fiber neuropathy. Associated neuropathic symptoms are often non–length-dependent, can precede sicca symptoms for up to 6 years, and in some cases are the sole manifestation of the disease.10 Small fiber neuropathy may also be associated with vasculitis, systemic lupus erythematosus, and other connective tissue disorders.
Testing should include:
- Erythrocyte sedimentation rate, C-reactive protein, and antinuclear antibodies: though these are nonspecific markers of inflammation, they may support an immune-mediated etiology if positive
- Extractable nuclear antigen panel: Sjögren syndrome A and B autoantibodies are the most important components in this setting5,11
- The Schirmer test or salivary gland biopsy should be considered for seronegative patients with sicca or a suspected immune-mediated etiology, as the sensitivity of antibody testing ranges from only 10% to 55%.10
Thyroid function testing
Hypothyroidism, and less commonly hyperthyroidism, are associated with small fiber neuropathy.
Metabolic tests for liver and kidney disease
Renal insufficiency and liver impairment are well-known causes of small nerve fiber dysfunction. Testing should include:
- Comprehensive metabolic panel
- Gamma-glutamyltransferase if alcohol abuse is suspected, since heavy alcohol use is one of the most common causes of both large and small fiber neuropathy.
HIV and hepatitis C testing
For patients with relevant risk factors, HIV and hepatitis C testing should be part of the initial workup (and as second-tier testing for others). Patients who test positive for hepatitis C should undergo further testing for cryoglobulinemia, which can present with painful small fiber neuropathy.26
Serum and urine immunoelectrophoresis
Paraproteinemia, with causes ranging from monoclonal gammopathy of uncertain significance to multiple myeloma, has been associated with small fiber neuropathy. An abnormal serum or urine immunoelectrophoresis test warrants further investigation and possibly referral to a hematology-oncology specialist.
SECOND-TIER TESTING
Less common treatable causes of small fiber neuropathy may also be evaluated.
Copper, vitamin B1 (thiamine), or vitamin B6 (pyridoxine) deficiency testing. Although vitamin B6 toxicity may also result in neuropathy due to its toxic effect on the dorsal root ganglia, the mildly elevated vitamin B6 levels often found in patients being evaluated for neuropathy are unlikely to be the primary cause of symptoms. Many laboratories require fasting samples for accurate vitamin B6 levels.
Angiotensin-converting enzyme levels for sarcoidosis. Small fiber neuropathy is common in sarcoidosis, occurring in more than 30% of patients with systemic disease.27 However, screening for sarcoidosis by measuring serum levels is often falsely positive and is not cost-effective. In a study of 195 patients with idiopathic small fiber neuropathy,11 44% had an elevated serum level, but no evidence of sarcoidosis was seen on further testing, which included computed tomography of the chest in 29 patients.12 Thus, this test is best used for patients with evidence of systemic disease.
Amyloid testing for amyloidosis. Fat pad or bone marrow biopsy should be considered in the appropriate clinical setting.
Paraneoplastic autoantibody panel for occult cancer. Such testing may also be considered if clinically warranted. However, if a patient is found to have low positive titers of paraneoplastic antibodies and suspicion is low for an occult cancer (eg, no weight loss or early satiety), repeat confirmatory testing at another laboratory should be done before embarking on an extensive search for malignancy.
Ganglionic acetylcholine receptor antibody testing for autoimmune autonomic ganglionopathy. This should be ordered for patients with prominent autonomic dysfunction. The antibody test can be ordered separately or as part of an autoantibody panel. The antibody may indicate a primary immune-mediated process or a paraneoplastic disease.28
Genetic mutation testing. Recent discoveries of gene mutations leading to peripheral nerve hyperexcitability of voltage-gated sodium channels have elucidated a hereditary cause of small fiber neuropathy in nearly 30% of cases that were once thought to be idiopathic.29,30 Genetic testing for mutations in SCN9A and SCN10 (which code for the Nav1.7 and Nav1.8 sodium channels, respectively) is commercially available and may be considered for those with a family history of neuropathic pain in the feet or for young, otherwise healthy patients.
Fabry disease is an X-linked lysosomal disorder characterized by angiokeratomas, cardiac and renal impairment, and small fiber neuropathy. Treatment is now available, but screening is not cost-efficient and should only be pursued in patients with other symptoms of the disease.31,32
OTHER POSSIBLE CAUSES
Guillain-Barré syndrome
A Guillain-Barré syndrome variant has been reported that is characterized by ascending limb paresthesias and cerebrospinal fluid albuminocytologic dissociation in the setting of preserved deep tendon reflexes and normal findings on EMG.12 The clinical course is similar to that of typical Guillain-Barré syndrome, in that symptoms follow an upper respiratory or gastrointestinal tract infection, reach their nadir at 4 weeks, and then gradually improve. Some patients respond to intravenous immune globulin.
Vaccine-associated
Postvaccination small fiber neuropathy has also been reported. The nature of the association is unclear.33
Parkinson disease
Small fiber neuropathy is associated with Parkinson disease. It is attributed to a number of proposed factors, including neurodegeneration that occurs parallel to central nervous system decline, as well as intestinal malabsorption with resultant vitamin deficiency.34,35
Rapid glycemic lowering
Aggressive treatment of diabetes, defined as at least a 2-point reduction of serum hemoglobin A1c level over 3 months, may result in acute small fiber neuropathy. It manifests as severe distal extremity pain and dysautonomia.
In a retrospective study,36 104 (10.9%) of 954 patients presenting to a tertiary diabetic clinic developed treatment-induced diabetic neuropathy with symptoms occurring within 8 weeks of rapid glycemic control. The severity of neuropathy correlated with the degree and rate of glycemic lowering. The condition was reversible in some cases.
TREATING SPECIFIC DISORDERS
For patients with an identified cause of neuropathy, targeted treatment offers the best chance of halting progression and possibly improving symptoms. Below are recommendations for addressing neuropathy associated with the common diagnoses.
Diabetes, impaired glucose tolerance, and metabolic syndrome. In addition to glycemic- and lipid-lowering therapies, lifestyle modifications with a specific focus on exercise and nutrition are integral to treating diabetes and related disorders.
In the Look AHEAD (Action for Health in Diabetes) study,37 which evaluated the effects of intensive lifestyle intervention on neuropathy in 5,145 overweight patients with type 2 diabetes, patients in the intervention group had lower pain scores and better touch sensation in the toes compared with controls at 1 year. Differences correlated with the degree of weight loss and reduction of hemoglobin A1c and lipid levels.
As running and walking may not be feasible for many patients owing to pain, stationary cycling, aqua therapy, and swimming are other options. A stationary recumbent bike may be useful for older patients with balance issues.
Vitamin B12 deficiency. As reduced absorption rather than low dietary intake is the primary cause of vitamin B12 deficiency for many patients, parenteral rather than oral supplementation may be best. A suggested regimen is subcutaneous or intramuscular methylcobalamin injection of 1,000 µg given daily for 1 week, then once weekly for 1 month, followed by a maintenance dose once a month for at least 6 to 12 months. Alternatively, a daily dose of vitamin B12 1,000 µg can be taken sublingually.
Sjögren syndrome. According to anecdotal case reports, intravenous immune globulin, corticosteroids, and other immunosuppressants help painful small fiber neuropathy and dysautonomia associated with Sjögren syndrome.10
Sarcoidosis. Sarcoidosis-associated small fiber neuropathy may also respond to intravenous immune globulin, as well as infliximab and combination therapy.9 Culver et al38 found that cibinetide, an experimental erythropoetin agonist, resulted in improved corneal nerve fiber measures in patients with small fiber neuropathy associated with sarcoidosis.
Celiac disease. A gluten-free diet is the treatment for celiac disease and can help some patients.
GENERAL MANAGEMENT
For all patients, regardless of whether the cause of small fiber neuropathy has been identified, managing symptoms remains key, as pain and autonomic dysfunction can markedly impair quality of life. A multidisciplinary approach that incorporates pain medications, physical therapy, and lifestyle modifications is ideal. Integrative holistic treatments such as natural supplements, yoga, and other mind-body therapies may also help.
Pain control
Mexiletine, a voltage-gated sodium channel blocker used as an antiarrhythmic, may help refractory pain or hereditary small fiber neuropathy related to sodium channel dysfunction. However, it is not recommended for diabetic neuropathy.39
Combination regimens that use drugs with different mechanisms of action can be effective. In one study, combined gabapentin and nortriptyline were more effective than either drug alone for neuropathic pain.40
Inhaled cannabis reduced pain in patients with HIV and diabetic neuropathy in a number of studies. Side effects included euphoria, somnolence, and cognitive impairment.41,42 The use of medical marijuana is not yet legal nationwide and may affect employability even in states in which it has been legalized.
Owing to the opioid epidemic and high addiction potential, opioids are no longer a preferred recommendation for chronic treatment of noncancer-related neuropathy. A population-based study of 2,892 patients with neuropathy found that those on chronic opioid therapy (≥ 90 days) had worse functional outcomes and higher rates of addiction and overdose than those on short-term therapy.43 However, the opioid agonist tramadol was found to be effective in reducing neuropathic pain and may be a safer option for patients with chronic small fiber neuropathy.44
Integrative, holistic therapies
PROGNOSIS
For many patients, small fiber neuropathy is a slowly progressive disorder that reaches a clinical plateau lasting for years, with progression to large fiber involvement reported in 13% to 36% of cases; over half of patients in one series either improved or remained stable over a period of 2 years.5,57 Long-term studies are needed to fully understand the natural disease course. In the meantime, treating underlying disease and managing symptoms are imperative to patient care.
- Burke JF, Skolarus LE, Callaghan BC, Kerber KA. Choosing Wisely: highest-cost tests in outpatient neurology. Ann Neurol 2013; 73(5):679–683. doi:10.1002/ana.23865
- Gordois A, Scuffham P, Shearer A, Oglesby A, Tobian JA. The health care costs of diabetic peripheral neuropathy in the US. Diabetes Care 2003; 26(6):1790–1795. pmid:12766111
- Peters MJ, Bakkers M, Merkies IS, Hoeijmakers JG, van Raak EP, Faber CG. Incidence and prevalence of small-fiber neuropathy: a survey in the Netherlands. Neurology 2013; 81(15):1356–1360. doi:10.1212/WNL.0b013e3182a8236e
- Periquet MI, Novak V, Collins MP, et al. Painful sensory neuropathy: prospective evaluation using skin biopsy. Neurology 1999; 53(8):1641–1647. pmid:10563606
- Devigili G, Tugnoli V, Penza P, et al. The diagnostic criteria for small fibre neuropathy: from symptoms to neuropathology. Brain 2008; 131(pt 7):1912–1925. doi:10.1093/brain/awn093
- Lacomis D. Small-fiber neuropathy. Muscle Nerve 2002; 26(2):173–188. doi:10.1002/mus.10181
- Lopate G, Streif E, Harms M, Weihl C, Pestronk A. Cramps and small-fiber neuropathy. Muscle Nerve 2013; 48(2):252–255. doi:10.1002/mus.23757
- Khan S, Zhou L. Characterization of non-length-dependent small-fiber sensory neuropathy. Muscle Nerve 2012; 45(1):86–91. doi:10.1002/mus.22255
- Tavee JO, Karwa K, Ahmed Z, Thompson N, Parambil J, Culver DA. Sarcoidosis-associated small fiber neuropathy in a large cohort: clinical aspects and response to IVIG and anti-TNF alpha treatment. Respir Med 2017; 126:135–138. doi:10.1016/j.rmed.2017.03.011
- Berkowitz AL, Samuels MA. The neurology of Sjogren’s syndrome and the rheumatology of peripheral neuropathy and myelitis. Pract Neurol 2014; 14(1):14–22. doi:10.1136/practneurol-2013-000651
- Lang M, Treister R, Oaklander AL. Diagnostic value of blood tests for occult causes of initially idiopathic small-fiber polyneuropathy. J Neurol 2016; 263(12):2515–2527. doi:10.1007/s00415-016-8270-5
- Seneviratne U, Gunasekera S. Acute small fibre sensory neuropathy: another variant of Guillain-Barré syndrome? J Neurol Neurosurg Psychiatry 2002; 72(4):540–542. pmid:11909922
- Tavee JO, Polston D, Zhou L, Shields RW, Butler RS, Levin KH. Sural sensory nerve action potential, epidermal nerve fiber density, and quantitative sudomotor axon reflex in the healthy elderly. Muscle Nerve 2014; 49(4):564–569. doi:10.1002/mus.23971
- Tavee J, Zhou L. Small fiber neuropathy: a burning problem. Cleve Clin J Med 2009; 76(5):297–305. doi:10.3949/ccjm.76a.08070
- Herrmann DN, Griffin JW, Hauer P, Cornblath DR, McArthur JC. Epidermal nerve fiber density and sural nerve morphometry in peripheral neuropathies. Neurology 1999; 53(8):1634–1640. pmid:10563605
- Oaklander AL, Herzog ZD, Downs HM, Klein MM. Objective evidence that small-fiber polyneuropathy underlies some illnesses currently labeled as fibromyalgia. Pain 2013; 154(11):2310–2316. doi:10.1016/j.pain.2013.06.001
- Üçeyler N, Zeller D, Kahn AK, et al. Small fibre pathology in patients with fibromyalgia syndrome. Brain 2013; 136(pt 6):1857–1867. doi:10.1093/brain/awt053
- Stewart JD, Low PA, Fealey RD. Distal small fiber neuropathy: results of tests of sweating and autonomic cardiovascular reflexes. Muscle Nerve 1992; 15(6):661–665. doi:10.1002/mus.880150605
- Malik RA, Kallinikos P, Abbott CA, et al. Corneal confocal microscopy: a non-invasive surrogate of nerve fibre damage and repair in diabetic patients. Diabetologia 2003; 46(5):683–688. doi:10.1007/s00125-003-1086-8
- de Greef BTA, Hoeijmakers JGJ, Gorissen-Brouwers CML, Geerts M, Faber CG, Merkies ISJ. Associated conditions in small fiber neuropathy—a large cohort study and review of the literature. Eur J Neurol 2018; 25(2):348–355. doi:10.1111/ene.13508
- Smith AG. Impaired glucose tolerance and metabolic syndrome in idiopathic neuropathy. J Peripher Nerv Syst 2012; 17(suppl 2):15–21. doi:10.1111/j.1529-8027.2012.00390.x
- Hoffman-Snyder C, Smith BE, Ross MA, Hernandez J, Bosch EP. Value of the oral glucose tolerance test in the evaluation of chronic idiopathic axonal polyneuropathy. Arch Neurol 2006; 63(8):1075–1079. doi:10.1001/archneur.63.8.noc50336
- Vincent AM, Hinder LM, Pop-Busui R, Feldman EL. Hyperlipidemia: a new therapeutic target for diabetic neuropathy. J Peripher Nerv Syst 2009; 14(4):257–267. doi:10.1111/j.1529-8027.2009.00237.x
- Wiggin TD, Sullivan KA, Pop-Busui R, Amato A, Sima AA, Feldman EL. Elevated triglycerides correlate with progression of diabetic neuropathy. Diabetes 2009; 58(7):1634–1640. doi:10.2337/db08-1771
- Chin RL, Sander HW, Brannagan TH, et al. Celiac neuropathy. Neurology 2003; 60(10):1581–1585. pmid:12771245
- Gemignani F, Brindani F, Alfieri S, et al. Clinical spectrum of cryoglobulinaemic neuropathy. J Neurol Neurosurg Psychiatry 2005; 76(10):1410–1414. doi:10.1136/jnnp.2004.057620
- Bakkers M, Merkies IS, Lauria G, et al. Intraepidermal nerve fiber density and its application in sarcoidosis. Neurology 2009; 73(14):1142–1148. doi:10.1212/WNL.0b013e3181bacf05
- Vernino S, Low PA, Fealey RD, Stewart JD, Farrugia G, Lennon VA. Autoantibodies to ganglionic acetylcholine receptors in autoimmune autonomic neuropathies. N Engl J Med 2000; 343(12):847–855. doi:10.1056/NEJM200009213431204
- Faber CG, Hoeijmakers JG, Ahn HS, et al. Gain of function Nav1.7 mutations in idiopathic small fiber neuropathy. Ann Neurol 2012; 71(1):26–39. doi:10.1002/ana.22485
- Brouwer BA, Merkies IS, Gerrits MM, Waxman SG, Hoeijmakers JG, Faber CG. Painful neuropathies: the emerging role of sodium channelopathies. J Peripher Nerv Syst 2014; 19(2):53–65. doi:10.1111/jns5.12071
- Samuelsson K, Kostulas K, Vrethem M, Rolfs A, Press R. Idiopathic small fiber neuropathy: phenotype, etiologies, and the search for Fabry disease. J Clin Neurol 2014; 10(2):108–118. doi:10.3988/jcn.2014.10.2.108
- de Greef BT, Hoeijmakers JG, Wolters EE, et al. No Fabry disease in patients presenting with isolated small fiber neuropathy. PLoS One 2016; 11(2):e0148316. doi:10.1371/journal.pone.0148316
- Souayah N, Ajroud-Driss S, Sander HW, Brannagan TH, Hays AP, Chin RL. Small fiber neuropathy following vaccination for rabies, varicella or Lyme disease. Vaccine 2009; 27(52):7322–7325. doi:10.1016/j.vaccine.2009.09.077
- Nolano M, Provitera V, Manganelli F, et al. Loss of cutaneous large and small fibers in naive and l-dopa–treated PD patients. Neurology 2017; 89(8):776–784. doi:10.1212/WNL.0000000000004274
- Zis P, Grünewald RA, Chaudhuri RK, Hadjivassiliou M. Peripheral neuropathy in idiopathic Parkinson’s disease: a systematic review. J Neurol Sci 2017; 378:204–209. doi:10.1016/j.jns.2017.05.023
- Gibbons CH, Freeman R. Treatment-induced neuropathy of diabetes: an acute, iatrogenic complication of diabetes. Brain 2015; 138(pt 1):43–52. doi:10.1093/brain/awu307
- Look AHEAD Research Group. Effects of a long-term lifestyle modification programme on peripheral neuropathy in overweight or obese adults with type 2 diabetes: the Look AHEAD study. Diabetologia 2017; 60(6):980–988. doi:10.1007/s00125-017-4253-z
- Culver DA, Dahan A, Bajorunas D, et al. Cibinetide improves corneal nerve fiber abundance in patients with sarcoidosis-associated small nerve fiber loss and neuropathic pain. Invest Ophthalmol Vis Sci 2017; 58(6):BIO52–BIO60. doi:10.1167/iovs.16-21291
- Bril V, England J, Franklin GM, et al; American Academy of Neurology; American Association of Neuromuscular and Electrodiagnostic Medicine; American Academy of Physical Medicine and Rehabilitation. Evidence-based guideline: treatment of painful diabetic neuropathy: report of the American Academy of Neurology, the American Association of Neuromuscular and Electrodiagnostic Medicine, and the American Academy of Physical Medicine and Rehabilitation. PM R 2011; 3(4):345–352.e21. doi:10.1016/j.pmrj.2011.03.008
- Gilron I, Bailey JM, Tu D, Holden RR, Jackson AC, Houlden RL. Nortriptyline and gabapentin, alone and in combination for neuropathic pain: a double-blind, randomised controlled crossover trial. Lancet 2009; 374(9697):1252–1261. doi:10.1016/S0140-6736(09)61081-3
- Ellis RJ, Toperoff W, Vaida F, et al. Smoked medicinal cannabis for neuropathic pain in HIV: a randomized, crossover clinical trial. Neuropsychopharmacology 2009; 34(3):672–680. doi:10.1038/npp.2008.120
- Wallace MS, Marcotte TD, Umlauf A, Gouaux B, Atkinson JH. Efficacy of inhaled cannabis on painful diabetic neuropathy. J Pain 2015; 16(7):616–627. doi:10.1016/j.jpain.2015.03.008
- Hoffman EM, Watson JC, St Sauver J, Staff NP, Klein CJ. Association of long-term opioid therapy with functional status, adverse outcomes, and mortality among patients with polyneuropathy. JAMA Neurol 2017; 74(7):773–779. doi:10.1001/jamaneurol.2017.0486
- Harati Y, Gooch C, Swenson M, et al. Double-blind randomized trial of tramadol for the treatment of the pain of diabetic neuropathy. Neurology 1998; 50(6):1842–1846. pmid:9633738
- Sima AA, Calvani M, Mehra M, Amato A; Acetyl-L-Carnitine Study Group. Acetyl-L-carnitine improves pain, nerve regeneration, and vibratory perception in patients with chronic diabetic neuropathy: an analysis of two randomized placebo-controlled trials. Diabetes Care 2005; 28(1):89–94. pmid:15616239
- Ziegler D, Hanefeld M, Ruhnau KJ, et al. Treatment of symptomatic diabetic peripheral neuropathy with the anti-oxidant alpha-lipoic acid. A 3-week multicentre randomized controlled trial (ALADIN Study). Diabetologia 1995; 38(12):1425–1433. pmid:8786016
- Scarpini E, Sacilotto G, Baron P, Cusini M, Scarlato G. Effect of acetyl-L-carnitine in the treatment of painful peripheral neuropathies in HIV+ patients. J Peripher Nerv Syst 1997; 2(3):250-252. pmid: 10975731
- Hershman DL, Unger JM, Crew KD, et al. Randomized double-blind placebo-controlled trial of acetyl-L-carnitine for the prevention of taxane-induced neuropathy in women undergoing adjuvant breast cancer therapy. J Clin Oncol 2013; 31(20):2627-2633. doi:10.1200/JCO.2012.44.8738
- Amara S. Oral glutamine for the prevention of chemotherapy-induced peripheral neuropathy. Ann Pharmacother 2008; 42(10):1481-1485. doi:10.1345/aph.1L179
- Huang JS, Wu CL, Fan CW, Chen WH, Yeh KY, Chang PH. Intravenous glutamine appears to reduce the severity of symptomatic platinum-induced neuropathy: a prospective randomized study. J Chemother 2015; 27(4):235-240. doi:10.1179/1973947815Y.0000000011
- Banafshe HR, Hamidi GA, Noureddini M, Mirhashemi SM, Mokhtari R, Shoferpour M. Effect of curcumin on diabetic peripheral neuropathic pain: possible involvement of opioid system. Eur J Pharmacol 2014; 723:202-206. doi:10.1016/j.ejphar.2013.11.033
- Mendonça LM, da Silva Machado C, Teixeira CC, de Freitas LA, Bianchi MD, Antunes LM. Curcumin reduces cisplatin-induced neurotoxicity in NGF-differentiated PC12 cells. Neurotoxicology 2013; 34:205-211. doi:10.1016/j.neuro.2012.09.011
- Wagner K, Lee KS, Yang J, Hammock BD. Epoxy fatty acids mediate analgesia in murine diabetic neuropathy. Eur J Pain 2017; 21(3):456-465. doi:10.1002/ejp.939
- Lewis EJ, Perkins BA, Lovblom LE, Bazinet RP, Wolever TMS, Bril V. Effect of omega-3 supplementation on neuropathy in type 1 diabetes: a 12-month pilot trial. Neurology 2017; 88(24):2294–2301. doi:10.1212/WNL.0000000000004033
- Hu D, Wang C, Li F, et al. A combined water extract of frankincense and myrrh alleviates neuropathic pain in mice via modulation of TRPV1. Neural Plast 2017; 2017:3710821. doi:10.1155/2017/3710821
- Tavee J, Rensel M, Planchon SM, Butler RS, Stone L. Effects of meditation on pain and quality of life in multiple sclerosis and peripheral neuropathy: a pilot study. Int J MS Care 2011; 13(4):163–168. doi:10.7224/1537-2073-13.4.163
- Khoshnoodi MA, Truelove S, Burakgazi A, Hoke A, Mammen AL, Polydefkis M. Longitudinal assessment of small fiber neuropathy: evidence of a non-length-dependent distal axonopathy. JAMA Neurol 2016; 73(6):684–690. doi:10.1001/jamaneurol.2016.0057
Peripheral neuropathy is the most common reason for an outpatient neurology visit in the United States and accounts for over $10 billion in healthcare spending each year.1,2 When the disorder affects only small, thinly myelinated or unmyelinated nerve fibers, it is referred to as small fiber neuropathy, which commonly presents as numbness and burning pain in the feet.
This article details the manifestations and evaluation of small fiber neuropathy, with an eye toward diagnosing an underlying cause amenable to treatment.
OLDER PATIENTS MOST AFFECTED
The epidemiology of small fiber neuropathy is not well established. It occurs more commonly in older patients, but data are mixed on prevalence by sex.3–6 In a Dutch study,3 the overall prevalence was at least 53 cases per 100,000, with the highest rate in men over age 65.
CHARACTERISTIC SENSORY DISTURBANCES
Sensations vary in quality and time
Patients with small fiber neuropathy typically present with a symmetric length-dependent (“stocking-glove”) distribution of sensory changes, starting in the feet and gradually ascending up the legs and then to the hands.
Commonly reported neuropathic symptoms include various combinations of burning, numbness, tingling, itching, sunburn-like, and frostbite-like sensations. Nonneuropathic symptoms may include tightness, a vise-like squeezing of the feet, and the sensation of a sock rolled up at the end of the shoe. Cramps or spasms may also be reported but rarely occur in isolation.7
Symptoms are typically worse at the end of the day and while sitting or lying down at night. They can arise spontaneously but may also be triggered by something as minor as the touch of clothing or cool air against the skin. Bedsheet sensitivity of the feet is reported so often that it is used as an outcome measure in clinical trials. Symptoms can also be exacerbated by extremes in ambient temperature and are especially worse in cold weather.
Random patterns suggest an immune cause
Symptoms may also have a non–length-dependent distribution that is asymmetric, patchy, intermittent, and migratory, and can involve the face, proximal limbs, and trunk. Symptoms may vary throughout the day, eg, starting with electric-shock sensations on one side of the face, followed by perineal numbness and then tingling in the arms lasting for a few minutes to several hours. While such patterns may be seen with diabetes and other common etiologies, they often suggest an underlying immune-mediated disorder such as Sjögren syndrome or sarcoidosis.8–10 Although large fiber polyneuropathy may also be non–length-dependent, the deficits are usually fixed, with no migratory component.
Autonomic features may be prominent
Autonomic symptoms occur in nearly half of patients and can be as troublesome as neuropathic pain.3 Small nerve fibers mediate somatic and autonomic functions, an evolutionary link that may reflect visceral defense mechanisms responding to pain as a signal of danger.11 This may help explain the multisystemic nature of symptoms, which can include sweating abnormalities, bowel and bladder disturbances, dry eyes, dry mouth, gastrointestinal dysmotility, skin changes (eg, discoloration, loss of hair, shiny skin), sexual dysfunction, orthostatic hypotension, and palpitations. In some cases, isolated dysautonomia may be seen.
TARGETED EXAMINATION
History: Medications, alcohol, infections
When a patient presents with neuropathic pain in the feet, a detailed history should be obtained, including alcohol use, family history of neuropathy, and use of neurotoxic medications such as metronidazole, colchicine, and chemotherapeutic agents.
Human immunodeficiency virus (HIV) and hepatitis C infection are well known to be associated with small fiber neuropathy, so relevant risk factors (eg, blood transfusions, sexual history, intravenous drug use) should be asked about. Recent illnesses and vaccinations are another important line of questioning, as a small-fiber variant of Guillain-Barré syndrome has been described.12
Assess reflexes, strength, sensation
On physical examination, particular attention should be focused on searching for abnormalities indicating large nerve fiber involvement (eg, absent deep tendon reflexes, weakness of the toes). However, absent ankle deep tendon reflexes and reduced vibratory sense may also occur in healthy elderly people.
Similarly, proprioception, motor strength, balance, and vibratory sensation are functions of large myelinated nerve fibers, and thus remain unaffected in patients with only small fiber neuropathy.
Evidence of a systemic disorder should also be sought, as it may indicate an underlying etiology.
DIAGNOSTIC TESTING
Although patients with either large or small fiber neuropathy may have subjective hyperesthesia or numbness of the distal lower extremities, the absence of significant abnormalities on neurologic examination should prompt consideration of small fiber neuropathy.
Electromyography worthwhile
Nerve conduction studies and needle electrode examination evaluate only large nerve fiber conditions. While electromyographic results are normal in patients with isolated small fiber neuropathy, the test can help evaluate subclinical large nerve fiber involvement and alternative diagnoses such as bilateral S1 radiculopathy. Nerve conduction studies may be less useful in patients over age 75, as they may lack sural sensory responses because of aging changes.13
Skin biopsy easy to do
Skin biopsy for evaluating intraepidermal nerve fiber density is one of the most widely used tests for small fiber neuropathy. This minimally invasive procedure can now be performed in a primary care office using readily available tools or prepackaged kits and analyzed by several commercial laboratories.
Reduced intraepidermal nerve fiber density on skin biopsy has been described in various other conditions such as fibromyalgia and chronic pain syndromes.16,17 The clinical significance of these findings remains uncertain.
Quantitative sudomotor axon reflex testing
Quantitative sudomotor axon reflex testing (QSART) is a noninvasive autonomic study that assesses the volume of sweat produced by the limbs in response to acetylcholine. A measure of postganglionic sympathetic sudomotor nerve function, QSART has a sensitivity of up to 80% and can be used to diagnose small fiber neuropathy.18 In a series of 115 patients with sarcoidosis small fiber neuropathy,9 the QSART and skin biopsy findings were concordant in 17 cases and complementary in 29, allowing for confirmation of small fiber neuropathy in patients whose condition would have remained undiagnosed had only one test been performed. QSART can also be considered in cases where skin biopsy may be contraindicated (eg, patient use of anticoagulation). Of note, the study may be affected by a number of external factors, including caffeine, tobacco, antihistamines, and tricyclic antidepressants; these should be held before testing.
Other diagnostic studies
Other tests may be helpful, as follows:
Tilt-table and cardiovagal testing may be useful for patients with orthostasis and palpitations.
Thermoregulatory sweat testing can be used to evaluate patients with abnormal patterns of sweating, eg, hyperhidrosis of the face and head.
INITIAL TESTING FOR AN UNDERLYING CAUSE
Glucose tolerance test for diabetes
Diabetes is the most common identifiable cause of small fiber neuropathy and accounts for about a third of all cases.5 Impaired glucose tolerance is also thought to be a risk factor and has been found in up to 50% of idiopathic cases, but the association is still being debated.21
While testing for hemoglobin A1c is more convenient for the patient, especially because it does not require fasting, a 2-hour oral glucose tolerance test is more sensitive for detecting glucose dysmetabolism.22
Lipid panel for metabolic syndrome
Small fiber neuropathy is associated with individual components of the metabolic syndrome, which include obesity, hyperglycemia, and dyslipidemia. Of these, dyslipidemia has emerged as the primary factor involved in the development of small fiber neuropathy, via an inflammatory pathway or oxidative stress mechanism.23,24
Vitamin B12 deficiency testing
Vitamin B12 deficiency, a potentially correctable cause of small fiber neuropathy, may be underdiagnosed, especially as values obtained by blood testing may not reflect tissue uptake. Causes of vitamin B12 deficiency include reduced intake, pernicious anemia, and medications that can affect absorption of vitamin B12 (eg, proton pump inhibitors, histamine 2 receptor antagonists, metformin).
Testing should include:
- Complete blood cell count to evaluate for vitamin B12-related macrocytic anemia and other hematologic abnormalities
- Serum vitamin B12 level
- Methylmalonic acid or homocysteine level in patients with subclinical or mild vitamin B12 deficiency, manifested as low to normal vitamin B12 levels (< 400 pg/mL); methylmalonic acid and homocysteine require vitamin B12 as a cofactor for enzymatic conversion, and either or both may be elevated in early vitamin B12 deficiency.
Celiac antibody panel
Celiac disease, a T-cell mediated enteropathy characterized by gluten intolerance and a herpetiform-like rash, can be associated with small fiber neuropathy.25 In some cases, neuropathy symptoms are preceded by the onset of gastrointestinal symptoms, or they may occur in isolation.25
Inflammatory disease testing
Sjögren syndrome accounts for nearly 10% of cases of small fiber neuropathy. Associated neuropathic symptoms are often non–length-dependent, can precede sicca symptoms for up to 6 years, and in some cases are the sole manifestation of the disease.10 Small fiber neuropathy may also be associated with vasculitis, systemic lupus erythematosus, and other connective tissue disorders.
Testing should include:
- Erythrocyte sedimentation rate, C-reactive protein, and antinuclear antibodies: though these are nonspecific markers of inflammation, they may support an immune-mediated etiology if positive
- Extractable nuclear antigen panel: Sjögren syndrome A and B autoantibodies are the most important components in this setting5,11
- The Schirmer test or salivary gland biopsy should be considered for seronegative patients with sicca or a suspected immune-mediated etiology, as the sensitivity of antibody testing ranges from only 10% to 55%.10
Thyroid function testing
Hypothyroidism, and less commonly hyperthyroidism, are associated with small fiber neuropathy.
Metabolic tests for liver and kidney disease
Renal insufficiency and liver impairment are well-known causes of small nerve fiber dysfunction. Testing should include:
- Comprehensive metabolic panel
- Gamma-glutamyltransferase if alcohol abuse is suspected, since heavy alcohol use is one of the most common causes of both large and small fiber neuropathy.
HIV and hepatitis C testing
For patients with relevant risk factors, HIV and hepatitis C testing should be part of the initial workup (and as second-tier testing for others). Patients who test positive for hepatitis C should undergo further testing for cryoglobulinemia, which can present with painful small fiber neuropathy.26
Serum and urine immunoelectrophoresis
Paraproteinemia, with causes ranging from monoclonal gammopathy of uncertain significance to multiple myeloma, has been associated with small fiber neuropathy. An abnormal serum or urine immunoelectrophoresis test warrants further investigation and possibly referral to a hematology-oncology specialist.
SECOND-TIER TESTING
Less common treatable causes of small fiber neuropathy may also be evaluated.
Copper, vitamin B1 (thiamine), or vitamin B6 (pyridoxine) deficiency testing. Although vitamin B6 toxicity may also result in neuropathy due to its toxic effect on the dorsal root ganglia, the mildly elevated vitamin B6 levels often found in patients being evaluated for neuropathy are unlikely to be the primary cause of symptoms. Many laboratories require fasting samples for accurate vitamin B6 levels.
Angiotensin-converting enzyme levels for sarcoidosis. Small fiber neuropathy is common in sarcoidosis, occurring in more than 30% of patients with systemic disease.27 However, screening for sarcoidosis by measuring serum levels is often falsely positive and is not cost-effective. In a study of 195 patients with idiopathic small fiber neuropathy,11 44% had an elevated serum level, but no evidence of sarcoidosis was seen on further testing, which included computed tomography of the chest in 29 patients.12 Thus, this test is best used for patients with evidence of systemic disease.
Amyloid testing for amyloidosis. Fat pad or bone marrow biopsy should be considered in the appropriate clinical setting.
Paraneoplastic autoantibody panel for occult cancer. Such testing may also be considered if clinically warranted. However, if a patient is found to have low positive titers of paraneoplastic antibodies and suspicion is low for an occult cancer (eg, no weight loss or early satiety), repeat confirmatory testing at another laboratory should be done before embarking on an extensive search for malignancy.
Ganglionic acetylcholine receptor antibody testing for autoimmune autonomic ganglionopathy. This should be ordered for patients with prominent autonomic dysfunction. The antibody test can be ordered separately or as part of an autoantibody panel. The antibody may indicate a primary immune-mediated process or a paraneoplastic disease.28
Genetic mutation testing. Recent discoveries of gene mutations leading to peripheral nerve hyperexcitability of voltage-gated sodium channels have elucidated a hereditary cause of small fiber neuropathy in nearly 30% of cases that were once thought to be idiopathic.29,30 Genetic testing for mutations in SCN9A and SCN10 (which code for the Nav1.7 and Nav1.8 sodium channels, respectively) is commercially available and may be considered for those with a family history of neuropathic pain in the feet or for young, otherwise healthy patients.
Fabry disease is an X-linked lysosomal disorder characterized by angiokeratomas, cardiac and renal impairment, and small fiber neuropathy. Treatment is now available, but screening is not cost-efficient and should only be pursued in patients with other symptoms of the disease.31,32
OTHER POSSIBLE CAUSES
Guillain-Barré syndrome
A Guillain-Barré syndrome variant has been reported that is characterized by ascending limb paresthesias and cerebrospinal fluid albuminocytologic dissociation in the setting of preserved deep tendon reflexes and normal findings on EMG.12 The clinical course is similar to that of typical Guillain-Barré syndrome, in that symptoms follow an upper respiratory or gastrointestinal tract infection, reach their nadir at 4 weeks, and then gradually improve. Some patients respond to intravenous immune globulin.
Vaccine-associated
Postvaccination small fiber neuropathy has also been reported. The nature of the association is unclear.33
Parkinson disease
Small fiber neuropathy is associated with Parkinson disease. It is attributed to a number of proposed factors, including neurodegeneration that occurs parallel to central nervous system decline, as well as intestinal malabsorption with resultant vitamin deficiency.34,35
Rapid glycemic lowering
Aggressive treatment of diabetes, defined as at least a 2-point reduction of serum hemoglobin A1c level over 3 months, may result in acute small fiber neuropathy. It manifests as severe distal extremity pain and dysautonomia.
In a retrospective study,36 104 (10.9%) of 954 patients presenting to a tertiary diabetic clinic developed treatment-induced diabetic neuropathy with symptoms occurring within 8 weeks of rapid glycemic control. The severity of neuropathy correlated with the degree and rate of glycemic lowering. The condition was reversible in some cases.
TREATING SPECIFIC DISORDERS
For patients with an identified cause of neuropathy, targeted treatment offers the best chance of halting progression and possibly improving symptoms. Below are recommendations for addressing neuropathy associated with the common diagnoses.
Diabetes, impaired glucose tolerance, and metabolic syndrome. In addition to glycemic- and lipid-lowering therapies, lifestyle modifications with a specific focus on exercise and nutrition are integral to treating diabetes and related disorders.
In the Look AHEAD (Action for Health in Diabetes) study,37 which evaluated the effects of intensive lifestyle intervention on neuropathy in 5,145 overweight patients with type 2 diabetes, patients in the intervention group had lower pain scores and better touch sensation in the toes compared with controls at 1 year. Differences correlated with the degree of weight loss and reduction of hemoglobin A1c and lipid levels.
As running and walking may not be feasible for many patients owing to pain, stationary cycling, aqua therapy, and swimming are other options. A stationary recumbent bike may be useful for older patients with balance issues.
Vitamin B12 deficiency. As reduced absorption rather than low dietary intake is the primary cause of vitamin B12 deficiency for many patients, parenteral rather than oral supplementation may be best. A suggested regimen is subcutaneous or intramuscular methylcobalamin injection of 1,000 µg given daily for 1 week, then once weekly for 1 month, followed by a maintenance dose once a month for at least 6 to 12 months. Alternatively, a daily dose of vitamin B12 1,000 µg can be taken sublingually.
Sjögren syndrome. According to anecdotal case reports, intravenous immune globulin, corticosteroids, and other immunosuppressants help painful small fiber neuropathy and dysautonomia associated with Sjögren syndrome.10
Sarcoidosis. Sarcoidosis-associated small fiber neuropathy may also respond to intravenous immune globulin, as well as infliximab and combination therapy.9 Culver et al38 found that cibinetide, an experimental erythropoetin agonist, resulted in improved corneal nerve fiber measures in patients with small fiber neuropathy associated with sarcoidosis.
Celiac disease. A gluten-free diet is the treatment for celiac disease and can help some patients.
GENERAL MANAGEMENT
For all patients, regardless of whether the cause of small fiber neuropathy has been identified, managing symptoms remains key, as pain and autonomic dysfunction can markedly impair quality of life. A multidisciplinary approach that incorporates pain medications, physical therapy, and lifestyle modifications is ideal. Integrative holistic treatments such as natural supplements, yoga, and other mind-body therapies may also help.
Pain control
Mexiletine, a voltage-gated sodium channel blocker used as an antiarrhythmic, may help refractory pain or hereditary small fiber neuropathy related to sodium channel dysfunction. However, it is not recommended for diabetic neuropathy.39
Combination regimens that use drugs with different mechanisms of action can be effective. In one study, combined gabapentin and nortriptyline were more effective than either drug alone for neuropathic pain.40
Inhaled cannabis reduced pain in patients with HIV and diabetic neuropathy in a number of studies. Side effects included euphoria, somnolence, and cognitive impairment.41,42 The use of medical marijuana is not yet legal nationwide and may affect employability even in states in which it has been legalized.
Owing to the opioid epidemic and high addiction potential, opioids are no longer a preferred recommendation for chronic treatment of noncancer-related neuropathy. A population-based study of 2,892 patients with neuropathy found that those on chronic opioid therapy (≥ 90 days) had worse functional outcomes and higher rates of addiction and overdose than those on short-term therapy.43 However, the opioid agonist tramadol was found to be effective in reducing neuropathic pain and may be a safer option for patients with chronic small fiber neuropathy.44
Integrative, holistic therapies
PROGNOSIS
For many patients, small fiber neuropathy is a slowly progressive disorder that reaches a clinical plateau lasting for years, with progression to large fiber involvement reported in 13% to 36% of cases; over half of patients in one series either improved or remained stable over a period of 2 years.5,57 Long-term studies are needed to fully understand the natural disease course. In the meantime, treating underlying disease and managing symptoms are imperative to patient care.
Peripheral neuropathy is the most common reason for an outpatient neurology visit in the United States and accounts for over $10 billion in healthcare spending each year.1,2 When the disorder affects only small, thinly myelinated or unmyelinated nerve fibers, it is referred to as small fiber neuropathy, which commonly presents as numbness and burning pain in the feet.
This article details the manifestations and evaluation of small fiber neuropathy, with an eye toward diagnosing an underlying cause amenable to treatment.
OLDER PATIENTS MOST AFFECTED
The epidemiology of small fiber neuropathy is not well established. It occurs more commonly in older patients, but data are mixed on prevalence by sex.3–6 In a Dutch study,3 the overall prevalence was at least 53 cases per 100,000, with the highest rate in men over age 65.
CHARACTERISTIC SENSORY DISTURBANCES
Sensations vary in quality and time
Patients with small fiber neuropathy typically present with a symmetric length-dependent (“stocking-glove”) distribution of sensory changes, starting in the feet and gradually ascending up the legs and then to the hands.
Commonly reported neuropathic symptoms include various combinations of burning, numbness, tingling, itching, sunburn-like, and frostbite-like sensations. Nonneuropathic symptoms may include tightness, a vise-like squeezing of the feet, and the sensation of a sock rolled up at the end of the shoe. Cramps or spasms may also be reported but rarely occur in isolation.7
Symptoms are typically worse at the end of the day and while sitting or lying down at night. They can arise spontaneously but may also be triggered by something as minor as the touch of clothing or cool air against the skin. Bedsheet sensitivity of the feet is reported so often that it is used as an outcome measure in clinical trials. Symptoms can also be exacerbated by extremes in ambient temperature and are especially worse in cold weather.
Random patterns suggest an immune cause
Symptoms may also have a non–length-dependent distribution that is asymmetric, patchy, intermittent, and migratory, and can involve the face, proximal limbs, and trunk. Symptoms may vary throughout the day, eg, starting with electric-shock sensations on one side of the face, followed by perineal numbness and then tingling in the arms lasting for a few minutes to several hours. While such patterns may be seen with diabetes and other common etiologies, they often suggest an underlying immune-mediated disorder such as Sjögren syndrome or sarcoidosis.8–10 Although large fiber polyneuropathy may also be non–length-dependent, the deficits are usually fixed, with no migratory component.
Autonomic features may be prominent
Autonomic symptoms occur in nearly half of patients and can be as troublesome as neuropathic pain.3 Small nerve fibers mediate somatic and autonomic functions, an evolutionary link that may reflect visceral defense mechanisms responding to pain as a signal of danger.11 This may help explain the multisystemic nature of symptoms, which can include sweating abnormalities, bowel and bladder disturbances, dry eyes, dry mouth, gastrointestinal dysmotility, skin changes (eg, discoloration, loss of hair, shiny skin), sexual dysfunction, orthostatic hypotension, and palpitations. In some cases, isolated dysautonomia may be seen.
TARGETED EXAMINATION
History: Medications, alcohol, infections
When a patient presents with neuropathic pain in the feet, a detailed history should be obtained, including alcohol use, family history of neuropathy, and use of neurotoxic medications such as metronidazole, colchicine, and chemotherapeutic agents.
Human immunodeficiency virus (HIV) and hepatitis C infection are well known to be associated with small fiber neuropathy, so relevant risk factors (eg, blood transfusions, sexual history, intravenous drug use) should be asked about. Recent illnesses and vaccinations are another important line of questioning, as a small-fiber variant of Guillain-Barré syndrome has been described.12
Assess reflexes, strength, sensation
On physical examination, particular attention should be focused on searching for abnormalities indicating large nerve fiber involvement (eg, absent deep tendon reflexes, weakness of the toes). However, absent ankle deep tendon reflexes and reduced vibratory sense may also occur in healthy elderly people.
Similarly, proprioception, motor strength, balance, and vibratory sensation are functions of large myelinated nerve fibers, and thus remain unaffected in patients with only small fiber neuropathy.
Evidence of a systemic disorder should also be sought, as it may indicate an underlying etiology.
DIAGNOSTIC TESTING
Although patients with either large or small fiber neuropathy may have subjective hyperesthesia or numbness of the distal lower extremities, the absence of significant abnormalities on neurologic examination should prompt consideration of small fiber neuropathy.
Electromyography worthwhile
Nerve conduction studies and needle electrode examination evaluate only large nerve fiber conditions. While electromyographic results are normal in patients with isolated small fiber neuropathy, the test can help evaluate subclinical large nerve fiber involvement and alternative diagnoses such as bilateral S1 radiculopathy. Nerve conduction studies may be less useful in patients over age 75, as they may lack sural sensory responses because of aging changes.13
Skin biopsy easy to do
Skin biopsy for evaluating intraepidermal nerve fiber density is one of the most widely used tests for small fiber neuropathy. This minimally invasive procedure can now be performed in a primary care office using readily available tools or prepackaged kits and analyzed by several commercial laboratories.
Reduced intraepidermal nerve fiber density on skin biopsy has been described in various other conditions such as fibromyalgia and chronic pain syndromes.16,17 The clinical significance of these findings remains uncertain.
Quantitative sudomotor axon reflex testing
Quantitative sudomotor axon reflex testing (QSART) is a noninvasive autonomic study that assesses the volume of sweat produced by the limbs in response to acetylcholine. A measure of postganglionic sympathetic sudomotor nerve function, QSART has a sensitivity of up to 80% and can be used to diagnose small fiber neuropathy.18 In a series of 115 patients with sarcoidosis small fiber neuropathy,9 the QSART and skin biopsy findings were concordant in 17 cases and complementary in 29, allowing for confirmation of small fiber neuropathy in patients whose condition would have remained undiagnosed had only one test been performed. QSART can also be considered in cases where skin biopsy may be contraindicated (eg, patient use of anticoagulation). Of note, the study may be affected by a number of external factors, including caffeine, tobacco, antihistamines, and tricyclic antidepressants; these should be held before testing.
Other diagnostic studies
Other tests may be helpful, as follows:
Tilt-table and cardiovagal testing may be useful for patients with orthostasis and palpitations.
Thermoregulatory sweat testing can be used to evaluate patients with abnormal patterns of sweating, eg, hyperhidrosis of the face and head.
INITIAL TESTING FOR AN UNDERLYING CAUSE
Glucose tolerance test for diabetes
Diabetes is the most common identifiable cause of small fiber neuropathy and accounts for about a third of all cases.5 Impaired glucose tolerance is also thought to be a risk factor and has been found in up to 50% of idiopathic cases, but the association is still being debated.21
While testing for hemoglobin A1c is more convenient for the patient, especially because it does not require fasting, a 2-hour oral glucose tolerance test is more sensitive for detecting glucose dysmetabolism.22
Lipid panel for metabolic syndrome
Small fiber neuropathy is associated with individual components of the metabolic syndrome, which include obesity, hyperglycemia, and dyslipidemia. Of these, dyslipidemia has emerged as the primary factor involved in the development of small fiber neuropathy, via an inflammatory pathway or oxidative stress mechanism.23,24
Vitamin B12 deficiency testing
Vitamin B12 deficiency, a potentially correctable cause of small fiber neuropathy, may be underdiagnosed, especially as values obtained by blood testing may not reflect tissue uptake. Causes of vitamin B12 deficiency include reduced intake, pernicious anemia, and medications that can affect absorption of vitamin B12 (eg, proton pump inhibitors, histamine 2 receptor antagonists, metformin).
Testing should include:
- Complete blood cell count to evaluate for vitamin B12-related macrocytic anemia and other hematologic abnormalities
- Serum vitamin B12 level
- Methylmalonic acid or homocysteine level in patients with subclinical or mild vitamin B12 deficiency, manifested as low to normal vitamin B12 levels (< 400 pg/mL); methylmalonic acid and homocysteine require vitamin B12 as a cofactor for enzymatic conversion, and either or both may be elevated in early vitamin B12 deficiency.
Celiac antibody panel
Celiac disease, a T-cell mediated enteropathy characterized by gluten intolerance and a herpetiform-like rash, can be associated with small fiber neuropathy.25 In some cases, neuropathy symptoms are preceded by the onset of gastrointestinal symptoms, or they may occur in isolation.25
Inflammatory disease testing
Sjögren syndrome accounts for nearly 10% of cases of small fiber neuropathy. Associated neuropathic symptoms are often non–length-dependent, can precede sicca symptoms for up to 6 years, and in some cases are the sole manifestation of the disease.10 Small fiber neuropathy may also be associated with vasculitis, systemic lupus erythematosus, and other connective tissue disorders.
Testing should include:
- Erythrocyte sedimentation rate, C-reactive protein, and antinuclear antibodies: though these are nonspecific markers of inflammation, they may support an immune-mediated etiology if positive
- Extractable nuclear antigen panel: Sjögren syndrome A and B autoantibodies are the most important components in this setting5,11
- The Schirmer test or salivary gland biopsy should be considered for seronegative patients with sicca or a suspected immune-mediated etiology, as the sensitivity of antibody testing ranges from only 10% to 55%.10
Thyroid function testing
Hypothyroidism, and less commonly hyperthyroidism, are associated with small fiber neuropathy.
Metabolic tests for liver and kidney disease
Renal insufficiency and liver impairment are well-known causes of small nerve fiber dysfunction. Testing should include:
- Comprehensive metabolic panel
- Gamma-glutamyltransferase if alcohol abuse is suspected, since heavy alcohol use is one of the most common causes of both large and small fiber neuropathy.
HIV and hepatitis C testing
For patients with relevant risk factors, HIV and hepatitis C testing should be part of the initial workup (and as second-tier testing for others). Patients who test positive for hepatitis C should undergo further testing for cryoglobulinemia, which can present with painful small fiber neuropathy.26
Serum and urine immunoelectrophoresis
Paraproteinemia, with causes ranging from monoclonal gammopathy of uncertain significance to multiple myeloma, has been associated with small fiber neuropathy. An abnormal serum or urine immunoelectrophoresis test warrants further investigation and possibly referral to a hematology-oncology specialist.
SECOND-TIER TESTING
Less common treatable causes of small fiber neuropathy may also be evaluated.
Copper, vitamin B1 (thiamine), or vitamin B6 (pyridoxine) deficiency testing. Although vitamin B6 toxicity may also result in neuropathy due to its toxic effect on the dorsal root ganglia, the mildly elevated vitamin B6 levels often found in patients being evaluated for neuropathy are unlikely to be the primary cause of symptoms. Many laboratories require fasting samples for accurate vitamin B6 levels.
Angiotensin-converting enzyme levels for sarcoidosis. Small fiber neuropathy is common in sarcoidosis, occurring in more than 30% of patients with systemic disease.27 However, screening for sarcoidosis by measuring serum levels is often falsely positive and is not cost-effective. In a study of 195 patients with idiopathic small fiber neuropathy,11 44% had an elevated serum level, but no evidence of sarcoidosis was seen on further testing, which included computed tomography of the chest in 29 patients.12 Thus, this test is best used for patients with evidence of systemic disease.
Amyloid testing for amyloidosis. Fat pad or bone marrow biopsy should be considered in the appropriate clinical setting.
Paraneoplastic autoantibody panel for occult cancer. Such testing may also be considered if clinically warranted. However, if a patient is found to have low positive titers of paraneoplastic antibodies and suspicion is low for an occult cancer (eg, no weight loss or early satiety), repeat confirmatory testing at another laboratory should be done before embarking on an extensive search for malignancy.
Ganglionic acetylcholine receptor antibody testing for autoimmune autonomic ganglionopathy. This should be ordered for patients with prominent autonomic dysfunction. The antibody test can be ordered separately or as part of an autoantibody panel. The antibody may indicate a primary immune-mediated process or a paraneoplastic disease.28
Genetic mutation testing. Recent discoveries of gene mutations leading to peripheral nerve hyperexcitability of voltage-gated sodium channels have elucidated a hereditary cause of small fiber neuropathy in nearly 30% of cases that were once thought to be idiopathic.29,30 Genetic testing for mutations in SCN9A and SCN10 (which code for the Nav1.7 and Nav1.8 sodium channels, respectively) is commercially available and may be considered for those with a family history of neuropathic pain in the feet or for young, otherwise healthy patients.
Fabry disease is an X-linked lysosomal disorder characterized by angiokeratomas, cardiac and renal impairment, and small fiber neuropathy. Treatment is now available, but screening is not cost-efficient and should only be pursued in patients with other symptoms of the disease.31,32
OTHER POSSIBLE CAUSES
Guillain-Barré syndrome
A Guillain-Barré syndrome variant has been reported that is characterized by ascending limb paresthesias and cerebrospinal fluid albuminocytologic dissociation in the setting of preserved deep tendon reflexes and normal findings on EMG.12 The clinical course is similar to that of typical Guillain-Barré syndrome, in that symptoms follow an upper respiratory or gastrointestinal tract infection, reach their nadir at 4 weeks, and then gradually improve. Some patients respond to intravenous immune globulin.
Vaccine-associated
Postvaccination small fiber neuropathy has also been reported. The nature of the association is unclear.33
Parkinson disease
Small fiber neuropathy is associated with Parkinson disease. It is attributed to a number of proposed factors, including neurodegeneration that occurs parallel to central nervous system decline, as well as intestinal malabsorption with resultant vitamin deficiency.34,35
Rapid glycemic lowering
Aggressive treatment of diabetes, defined as at least a 2-point reduction of serum hemoglobin A1c level over 3 months, may result in acute small fiber neuropathy. It manifests as severe distal extremity pain and dysautonomia.
In a retrospective study,36 104 (10.9%) of 954 patients presenting to a tertiary diabetic clinic developed treatment-induced diabetic neuropathy with symptoms occurring within 8 weeks of rapid glycemic control. The severity of neuropathy correlated with the degree and rate of glycemic lowering. The condition was reversible in some cases.
TREATING SPECIFIC DISORDERS
For patients with an identified cause of neuropathy, targeted treatment offers the best chance of halting progression and possibly improving symptoms. Below are recommendations for addressing neuropathy associated with the common diagnoses.
Diabetes, impaired glucose tolerance, and metabolic syndrome. In addition to glycemic- and lipid-lowering therapies, lifestyle modifications with a specific focus on exercise and nutrition are integral to treating diabetes and related disorders.
In the Look AHEAD (Action for Health in Diabetes) study,37 which evaluated the effects of intensive lifestyle intervention on neuropathy in 5,145 overweight patients with type 2 diabetes, patients in the intervention group had lower pain scores and better touch sensation in the toes compared with controls at 1 year. Differences correlated with the degree of weight loss and reduction of hemoglobin A1c and lipid levels.
As running and walking may not be feasible for many patients owing to pain, stationary cycling, aqua therapy, and swimming are other options. A stationary recumbent bike may be useful for older patients with balance issues.
Vitamin B12 deficiency. As reduced absorption rather than low dietary intake is the primary cause of vitamin B12 deficiency for many patients, parenteral rather than oral supplementation may be best. A suggested regimen is subcutaneous or intramuscular methylcobalamin injection of 1,000 µg given daily for 1 week, then once weekly for 1 month, followed by a maintenance dose once a month for at least 6 to 12 months. Alternatively, a daily dose of vitamin B12 1,000 µg can be taken sublingually.
Sjögren syndrome. According to anecdotal case reports, intravenous immune globulin, corticosteroids, and other immunosuppressants help painful small fiber neuropathy and dysautonomia associated with Sjögren syndrome.10
Sarcoidosis. Sarcoidosis-associated small fiber neuropathy may also respond to intravenous immune globulin, as well as infliximab and combination therapy.9 Culver et al38 found that cibinetide, an experimental erythropoetin agonist, resulted in improved corneal nerve fiber measures in patients with small fiber neuropathy associated with sarcoidosis.
Celiac disease. A gluten-free diet is the treatment for celiac disease and can help some patients.
GENERAL MANAGEMENT
For all patients, regardless of whether the cause of small fiber neuropathy has been identified, managing symptoms remains key, as pain and autonomic dysfunction can markedly impair quality of life. A multidisciplinary approach that incorporates pain medications, physical therapy, and lifestyle modifications is ideal. Integrative holistic treatments such as natural supplements, yoga, and other mind-body therapies may also help.
Pain control
Mexiletine, a voltage-gated sodium channel blocker used as an antiarrhythmic, may help refractory pain or hereditary small fiber neuropathy related to sodium channel dysfunction. However, it is not recommended for diabetic neuropathy.39
Combination regimens that use drugs with different mechanisms of action can be effective. In one study, combined gabapentin and nortriptyline were more effective than either drug alone for neuropathic pain.40
Inhaled cannabis reduced pain in patients with HIV and diabetic neuropathy in a number of studies. Side effects included euphoria, somnolence, and cognitive impairment.41,42 The use of medical marijuana is not yet legal nationwide and may affect employability even in states in which it has been legalized.
Owing to the opioid epidemic and high addiction potential, opioids are no longer a preferred recommendation for chronic treatment of noncancer-related neuropathy. A population-based study of 2,892 patients with neuropathy found that those on chronic opioid therapy (≥ 90 days) had worse functional outcomes and higher rates of addiction and overdose than those on short-term therapy.43 However, the opioid agonist tramadol was found to be effective in reducing neuropathic pain and may be a safer option for patients with chronic small fiber neuropathy.44
Integrative, holistic therapies
PROGNOSIS
For many patients, small fiber neuropathy is a slowly progressive disorder that reaches a clinical plateau lasting for years, with progression to large fiber involvement reported in 13% to 36% of cases; over half of patients in one series either improved or remained stable over a period of 2 years.5,57 Long-term studies are needed to fully understand the natural disease course. In the meantime, treating underlying disease and managing symptoms are imperative to patient care.
- Burke JF, Skolarus LE, Callaghan BC, Kerber KA. Choosing Wisely: highest-cost tests in outpatient neurology. Ann Neurol 2013; 73(5):679–683. doi:10.1002/ana.23865
- Gordois A, Scuffham P, Shearer A, Oglesby A, Tobian JA. The health care costs of diabetic peripheral neuropathy in the US. Diabetes Care 2003; 26(6):1790–1795. pmid:12766111
- Peters MJ, Bakkers M, Merkies IS, Hoeijmakers JG, van Raak EP, Faber CG. Incidence and prevalence of small-fiber neuropathy: a survey in the Netherlands. Neurology 2013; 81(15):1356–1360. doi:10.1212/WNL.0b013e3182a8236e
- Periquet MI, Novak V, Collins MP, et al. Painful sensory neuropathy: prospective evaluation using skin biopsy. Neurology 1999; 53(8):1641–1647. pmid:10563606
- Devigili G, Tugnoli V, Penza P, et al. The diagnostic criteria for small fibre neuropathy: from symptoms to neuropathology. Brain 2008; 131(pt 7):1912–1925. doi:10.1093/brain/awn093
- Lacomis D. Small-fiber neuropathy. Muscle Nerve 2002; 26(2):173–188. doi:10.1002/mus.10181
- Lopate G, Streif E, Harms M, Weihl C, Pestronk A. Cramps and small-fiber neuropathy. Muscle Nerve 2013; 48(2):252–255. doi:10.1002/mus.23757
- Khan S, Zhou L. Characterization of non-length-dependent small-fiber sensory neuropathy. Muscle Nerve 2012; 45(1):86–91. doi:10.1002/mus.22255
- Tavee JO, Karwa K, Ahmed Z, Thompson N, Parambil J, Culver DA. Sarcoidosis-associated small fiber neuropathy in a large cohort: clinical aspects and response to IVIG and anti-TNF alpha treatment. Respir Med 2017; 126:135–138. doi:10.1016/j.rmed.2017.03.011
- Berkowitz AL, Samuels MA. The neurology of Sjogren’s syndrome and the rheumatology of peripheral neuropathy and myelitis. Pract Neurol 2014; 14(1):14–22. doi:10.1136/practneurol-2013-000651
- Lang M, Treister R, Oaklander AL. Diagnostic value of blood tests for occult causes of initially idiopathic small-fiber polyneuropathy. J Neurol 2016; 263(12):2515–2527. doi:10.1007/s00415-016-8270-5
- Seneviratne U, Gunasekera S. Acute small fibre sensory neuropathy: another variant of Guillain-Barré syndrome? J Neurol Neurosurg Psychiatry 2002; 72(4):540–542. pmid:11909922
- Tavee JO, Polston D, Zhou L, Shields RW, Butler RS, Levin KH. Sural sensory nerve action potential, epidermal nerve fiber density, and quantitative sudomotor axon reflex in the healthy elderly. Muscle Nerve 2014; 49(4):564–569. doi:10.1002/mus.23971
- Tavee J, Zhou L. Small fiber neuropathy: a burning problem. Cleve Clin J Med 2009; 76(5):297–305. doi:10.3949/ccjm.76a.08070
- Herrmann DN, Griffin JW, Hauer P, Cornblath DR, McArthur JC. Epidermal nerve fiber density and sural nerve morphometry in peripheral neuropathies. Neurology 1999; 53(8):1634–1640. pmid:10563605
- Oaklander AL, Herzog ZD, Downs HM, Klein MM. Objective evidence that small-fiber polyneuropathy underlies some illnesses currently labeled as fibromyalgia. Pain 2013; 154(11):2310–2316. doi:10.1016/j.pain.2013.06.001
- Üçeyler N, Zeller D, Kahn AK, et al. Small fibre pathology in patients with fibromyalgia syndrome. Brain 2013; 136(pt 6):1857–1867. doi:10.1093/brain/awt053
- Stewart JD, Low PA, Fealey RD. Distal small fiber neuropathy: results of tests of sweating and autonomic cardiovascular reflexes. Muscle Nerve 1992; 15(6):661–665. doi:10.1002/mus.880150605
- Malik RA, Kallinikos P, Abbott CA, et al. Corneal confocal microscopy: a non-invasive surrogate of nerve fibre damage and repair in diabetic patients. Diabetologia 2003; 46(5):683–688. doi:10.1007/s00125-003-1086-8
- de Greef BTA, Hoeijmakers JGJ, Gorissen-Brouwers CML, Geerts M, Faber CG, Merkies ISJ. Associated conditions in small fiber neuropathy—a large cohort study and review of the literature. Eur J Neurol 2018; 25(2):348–355. doi:10.1111/ene.13508
- Smith AG. Impaired glucose tolerance and metabolic syndrome in idiopathic neuropathy. J Peripher Nerv Syst 2012; 17(suppl 2):15–21. doi:10.1111/j.1529-8027.2012.00390.x
- Hoffman-Snyder C, Smith BE, Ross MA, Hernandez J, Bosch EP. Value of the oral glucose tolerance test in the evaluation of chronic idiopathic axonal polyneuropathy. Arch Neurol 2006; 63(8):1075–1079. doi:10.1001/archneur.63.8.noc50336
- Vincent AM, Hinder LM, Pop-Busui R, Feldman EL. Hyperlipidemia: a new therapeutic target for diabetic neuropathy. J Peripher Nerv Syst 2009; 14(4):257–267. doi:10.1111/j.1529-8027.2009.00237.x
- Wiggin TD, Sullivan KA, Pop-Busui R, Amato A, Sima AA, Feldman EL. Elevated triglycerides correlate with progression of diabetic neuropathy. Diabetes 2009; 58(7):1634–1640. doi:10.2337/db08-1771
- Chin RL, Sander HW, Brannagan TH, et al. Celiac neuropathy. Neurology 2003; 60(10):1581–1585. pmid:12771245
- Gemignani F, Brindani F, Alfieri S, et al. Clinical spectrum of cryoglobulinaemic neuropathy. J Neurol Neurosurg Psychiatry 2005; 76(10):1410–1414. doi:10.1136/jnnp.2004.057620
- Bakkers M, Merkies IS, Lauria G, et al. Intraepidermal nerve fiber density and its application in sarcoidosis. Neurology 2009; 73(14):1142–1148. doi:10.1212/WNL.0b013e3181bacf05
- Vernino S, Low PA, Fealey RD, Stewart JD, Farrugia G, Lennon VA. Autoantibodies to ganglionic acetylcholine receptors in autoimmune autonomic neuropathies. N Engl J Med 2000; 343(12):847–855. doi:10.1056/NEJM200009213431204
- Faber CG, Hoeijmakers JG, Ahn HS, et al. Gain of function Nav1.7 mutations in idiopathic small fiber neuropathy. Ann Neurol 2012; 71(1):26–39. doi:10.1002/ana.22485
- Brouwer BA, Merkies IS, Gerrits MM, Waxman SG, Hoeijmakers JG, Faber CG. Painful neuropathies: the emerging role of sodium channelopathies. J Peripher Nerv Syst 2014; 19(2):53–65. doi:10.1111/jns5.12071
- Samuelsson K, Kostulas K, Vrethem M, Rolfs A, Press R. Idiopathic small fiber neuropathy: phenotype, etiologies, and the search for Fabry disease. J Clin Neurol 2014; 10(2):108–118. doi:10.3988/jcn.2014.10.2.108
- de Greef BT, Hoeijmakers JG, Wolters EE, et al. No Fabry disease in patients presenting with isolated small fiber neuropathy. PLoS One 2016; 11(2):e0148316. doi:10.1371/journal.pone.0148316
- Souayah N, Ajroud-Driss S, Sander HW, Brannagan TH, Hays AP, Chin RL. Small fiber neuropathy following vaccination for rabies, varicella or Lyme disease. Vaccine 2009; 27(52):7322–7325. doi:10.1016/j.vaccine.2009.09.077
- Nolano M, Provitera V, Manganelli F, et al. Loss of cutaneous large and small fibers in naive and l-dopa–treated PD patients. Neurology 2017; 89(8):776–784. doi:10.1212/WNL.0000000000004274
- Zis P, Grünewald RA, Chaudhuri RK, Hadjivassiliou M. Peripheral neuropathy in idiopathic Parkinson’s disease: a systematic review. J Neurol Sci 2017; 378:204–209. doi:10.1016/j.jns.2017.05.023
- Gibbons CH, Freeman R. Treatment-induced neuropathy of diabetes: an acute, iatrogenic complication of diabetes. Brain 2015; 138(pt 1):43–52. doi:10.1093/brain/awu307
- Look AHEAD Research Group. Effects of a long-term lifestyle modification programme on peripheral neuropathy in overweight or obese adults with type 2 diabetes: the Look AHEAD study. Diabetologia 2017; 60(6):980–988. doi:10.1007/s00125-017-4253-z
- Culver DA, Dahan A, Bajorunas D, et al. Cibinetide improves corneal nerve fiber abundance in patients with sarcoidosis-associated small nerve fiber loss and neuropathic pain. Invest Ophthalmol Vis Sci 2017; 58(6):BIO52–BIO60. doi:10.1167/iovs.16-21291
- Bril V, England J, Franklin GM, et al; American Academy of Neurology; American Association of Neuromuscular and Electrodiagnostic Medicine; American Academy of Physical Medicine and Rehabilitation. Evidence-based guideline: treatment of painful diabetic neuropathy: report of the American Academy of Neurology, the American Association of Neuromuscular and Electrodiagnostic Medicine, and the American Academy of Physical Medicine and Rehabilitation. PM R 2011; 3(4):345–352.e21. doi:10.1016/j.pmrj.2011.03.008
- Gilron I, Bailey JM, Tu D, Holden RR, Jackson AC, Houlden RL. Nortriptyline and gabapentin, alone and in combination for neuropathic pain: a double-blind, randomised controlled crossover trial. Lancet 2009; 374(9697):1252–1261. doi:10.1016/S0140-6736(09)61081-3
- Ellis RJ, Toperoff W, Vaida F, et al. Smoked medicinal cannabis for neuropathic pain in HIV: a randomized, crossover clinical trial. Neuropsychopharmacology 2009; 34(3):672–680. doi:10.1038/npp.2008.120
- Wallace MS, Marcotte TD, Umlauf A, Gouaux B, Atkinson JH. Efficacy of inhaled cannabis on painful diabetic neuropathy. J Pain 2015; 16(7):616–627. doi:10.1016/j.jpain.2015.03.008
- Hoffman EM, Watson JC, St Sauver J, Staff NP, Klein CJ. Association of long-term opioid therapy with functional status, adverse outcomes, and mortality among patients with polyneuropathy. JAMA Neurol 2017; 74(7):773–779. doi:10.1001/jamaneurol.2017.0486
- Harati Y, Gooch C, Swenson M, et al. Double-blind randomized trial of tramadol for the treatment of the pain of diabetic neuropathy. Neurology 1998; 50(6):1842–1846. pmid:9633738
- Sima AA, Calvani M, Mehra M, Amato A; Acetyl-L-Carnitine Study Group. Acetyl-L-carnitine improves pain, nerve regeneration, and vibratory perception in patients with chronic diabetic neuropathy: an analysis of two randomized placebo-controlled trials. Diabetes Care 2005; 28(1):89–94. pmid:15616239
- Ziegler D, Hanefeld M, Ruhnau KJ, et al. Treatment of symptomatic diabetic peripheral neuropathy with the anti-oxidant alpha-lipoic acid. A 3-week multicentre randomized controlled trial (ALADIN Study). Diabetologia 1995; 38(12):1425–1433. pmid:8786016
- Scarpini E, Sacilotto G, Baron P, Cusini M, Scarlato G. Effect of acetyl-L-carnitine in the treatment of painful peripheral neuropathies in HIV+ patients. J Peripher Nerv Syst 1997; 2(3):250-252. pmid: 10975731
- Hershman DL, Unger JM, Crew KD, et al. Randomized double-blind placebo-controlled trial of acetyl-L-carnitine for the prevention of taxane-induced neuropathy in women undergoing adjuvant breast cancer therapy. J Clin Oncol 2013; 31(20):2627-2633. doi:10.1200/JCO.2012.44.8738
- Amara S. Oral glutamine for the prevention of chemotherapy-induced peripheral neuropathy. Ann Pharmacother 2008; 42(10):1481-1485. doi:10.1345/aph.1L179
- Huang JS, Wu CL, Fan CW, Chen WH, Yeh KY, Chang PH. Intravenous glutamine appears to reduce the severity of symptomatic platinum-induced neuropathy: a prospective randomized study. J Chemother 2015; 27(4):235-240. doi:10.1179/1973947815Y.0000000011
- Banafshe HR, Hamidi GA, Noureddini M, Mirhashemi SM, Mokhtari R, Shoferpour M. Effect of curcumin on diabetic peripheral neuropathic pain: possible involvement of opioid system. Eur J Pharmacol 2014; 723:202-206. doi:10.1016/j.ejphar.2013.11.033
- Mendonça LM, da Silva Machado C, Teixeira CC, de Freitas LA, Bianchi MD, Antunes LM. Curcumin reduces cisplatin-induced neurotoxicity in NGF-differentiated PC12 cells. Neurotoxicology 2013; 34:205-211. doi:10.1016/j.neuro.2012.09.011
- Wagner K, Lee KS, Yang J, Hammock BD. Epoxy fatty acids mediate analgesia in murine diabetic neuropathy. Eur J Pain 2017; 21(3):456-465. doi:10.1002/ejp.939
- Lewis EJ, Perkins BA, Lovblom LE, Bazinet RP, Wolever TMS, Bril V. Effect of omega-3 supplementation on neuropathy in type 1 diabetes: a 12-month pilot trial. Neurology 2017; 88(24):2294–2301. doi:10.1212/WNL.0000000000004033
- Hu D, Wang C, Li F, et al. A combined water extract of frankincense and myrrh alleviates neuropathic pain in mice via modulation of TRPV1. Neural Plast 2017; 2017:3710821. doi:10.1155/2017/3710821
- Tavee J, Rensel M, Planchon SM, Butler RS, Stone L. Effects of meditation on pain and quality of life in multiple sclerosis and peripheral neuropathy: a pilot study. Int J MS Care 2011; 13(4):163–168. doi:10.7224/1537-2073-13.4.163
- Khoshnoodi MA, Truelove S, Burakgazi A, Hoke A, Mammen AL, Polydefkis M. Longitudinal assessment of small fiber neuropathy: evidence of a non-length-dependent distal axonopathy. JAMA Neurol 2016; 73(6):684–690. doi:10.1001/jamaneurol.2016.0057
- Burke JF, Skolarus LE, Callaghan BC, Kerber KA. Choosing Wisely: highest-cost tests in outpatient neurology. Ann Neurol 2013; 73(5):679–683. doi:10.1002/ana.23865
- Gordois A, Scuffham P, Shearer A, Oglesby A, Tobian JA. The health care costs of diabetic peripheral neuropathy in the US. Diabetes Care 2003; 26(6):1790–1795. pmid:12766111
- Peters MJ, Bakkers M, Merkies IS, Hoeijmakers JG, van Raak EP, Faber CG. Incidence and prevalence of small-fiber neuropathy: a survey in the Netherlands. Neurology 2013; 81(15):1356–1360. doi:10.1212/WNL.0b013e3182a8236e
- Periquet MI, Novak V, Collins MP, et al. Painful sensory neuropathy: prospective evaluation using skin biopsy. Neurology 1999; 53(8):1641–1647. pmid:10563606
- Devigili G, Tugnoli V, Penza P, et al. The diagnostic criteria for small fibre neuropathy: from symptoms to neuropathology. Brain 2008; 131(pt 7):1912–1925. doi:10.1093/brain/awn093
- Lacomis D. Small-fiber neuropathy. Muscle Nerve 2002; 26(2):173–188. doi:10.1002/mus.10181
- Lopate G, Streif E, Harms M, Weihl C, Pestronk A. Cramps and small-fiber neuropathy. Muscle Nerve 2013; 48(2):252–255. doi:10.1002/mus.23757
- Khan S, Zhou L. Characterization of non-length-dependent small-fiber sensory neuropathy. Muscle Nerve 2012; 45(1):86–91. doi:10.1002/mus.22255
- Tavee JO, Karwa K, Ahmed Z, Thompson N, Parambil J, Culver DA. Sarcoidosis-associated small fiber neuropathy in a large cohort: clinical aspects and response to IVIG and anti-TNF alpha treatment. Respir Med 2017; 126:135–138. doi:10.1016/j.rmed.2017.03.011
- Berkowitz AL, Samuels MA. The neurology of Sjogren’s syndrome and the rheumatology of peripheral neuropathy and myelitis. Pract Neurol 2014; 14(1):14–22. doi:10.1136/practneurol-2013-000651
- Lang M, Treister R, Oaklander AL. Diagnostic value of blood tests for occult causes of initially idiopathic small-fiber polyneuropathy. J Neurol 2016; 263(12):2515–2527. doi:10.1007/s00415-016-8270-5
- Seneviratne U, Gunasekera S. Acute small fibre sensory neuropathy: another variant of Guillain-Barré syndrome? J Neurol Neurosurg Psychiatry 2002; 72(4):540–542. pmid:11909922
- Tavee JO, Polston D, Zhou L, Shields RW, Butler RS, Levin KH. Sural sensory nerve action potential, epidermal nerve fiber density, and quantitative sudomotor axon reflex in the healthy elderly. Muscle Nerve 2014; 49(4):564–569. doi:10.1002/mus.23971
- Tavee J, Zhou L. Small fiber neuropathy: a burning problem. Cleve Clin J Med 2009; 76(5):297–305. doi:10.3949/ccjm.76a.08070
- Herrmann DN, Griffin JW, Hauer P, Cornblath DR, McArthur JC. Epidermal nerve fiber density and sural nerve morphometry in peripheral neuropathies. Neurology 1999; 53(8):1634–1640. pmid:10563605
- Oaklander AL, Herzog ZD, Downs HM, Klein MM. Objective evidence that small-fiber polyneuropathy underlies some illnesses currently labeled as fibromyalgia. Pain 2013; 154(11):2310–2316. doi:10.1016/j.pain.2013.06.001
- Üçeyler N, Zeller D, Kahn AK, et al. Small fibre pathology in patients with fibromyalgia syndrome. Brain 2013; 136(pt 6):1857–1867. doi:10.1093/brain/awt053
- Stewart JD, Low PA, Fealey RD. Distal small fiber neuropathy: results of tests of sweating and autonomic cardiovascular reflexes. Muscle Nerve 1992; 15(6):661–665. doi:10.1002/mus.880150605
- Malik RA, Kallinikos P, Abbott CA, et al. Corneal confocal microscopy: a non-invasive surrogate of nerve fibre damage and repair in diabetic patients. Diabetologia 2003; 46(5):683–688. doi:10.1007/s00125-003-1086-8
- de Greef BTA, Hoeijmakers JGJ, Gorissen-Brouwers CML, Geerts M, Faber CG, Merkies ISJ. Associated conditions in small fiber neuropathy—a large cohort study and review of the literature. Eur J Neurol 2018; 25(2):348–355. doi:10.1111/ene.13508
- Smith AG. Impaired glucose tolerance and metabolic syndrome in idiopathic neuropathy. J Peripher Nerv Syst 2012; 17(suppl 2):15–21. doi:10.1111/j.1529-8027.2012.00390.x
- Hoffman-Snyder C, Smith BE, Ross MA, Hernandez J, Bosch EP. Value of the oral glucose tolerance test in the evaluation of chronic idiopathic axonal polyneuropathy. Arch Neurol 2006; 63(8):1075–1079. doi:10.1001/archneur.63.8.noc50336
- Vincent AM, Hinder LM, Pop-Busui R, Feldman EL. Hyperlipidemia: a new therapeutic target for diabetic neuropathy. J Peripher Nerv Syst 2009; 14(4):257–267. doi:10.1111/j.1529-8027.2009.00237.x
- Wiggin TD, Sullivan KA, Pop-Busui R, Amato A, Sima AA, Feldman EL. Elevated triglycerides correlate with progression of diabetic neuropathy. Diabetes 2009; 58(7):1634–1640. doi:10.2337/db08-1771
- Chin RL, Sander HW, Brannagan TH, et al. Celiac neuropathy. Neurology 2003; 60(10):1581–1585. pmid:12771245
- Gemignani F, Brindani F, Alfieri S, et al. Clinical spectrum of cryoglobulinaemic neuropathy. J Neurol Neurosurg Psychiatry 2005; 76(10):1410–1414. doi:10.1136/jnnp.2004.057620
- Bakkers M, Merkies IS, Lauria G, et al. Intraepidermal nerve fiber density and its application in sarcoidosis. Neurology 2009; 73(14):1142–1148. doi:10.1212/WNL.0b013e3181bacf05
- Vernino S, Low PA, Fealey RD, Stewart JD, Farrugia G, Lennon VA. Autoantibodies to ganglionic acetylcholine receptors in autoimmune autonomic neuropathies. N Engl J Med 2000; 343(12):847–855. doi:10.1056/NEJM200009213431204
- Faber CG, Hoeijmakers JG, Ahn HS, et al. Gain of function Nav1.7 mutations in idiopathic small fiber neuropathy. Ann Neurol 2012; 71(1):26–39. doi:10.1002/ana.22485
- Brouwer BA, Merkies IS, Gerrits MM, Waxman SG, Hoeijmakers JG, Faber CG. Painful neuropathies: the emerging role of sodium channelopathies. J Peripher Nerv Syst 2014; 19(2):53–65. doi:10.1111/jns5.12071
- Samuelsson K, Kostulas K, Vrethem M, Rolfs A, Press R. Idiopathic small fiber neuropathy: phenotype, etiologies, and the search for Fabry disease. J Clin Neurol 2014; 10(2):108–118. doi:10.3988/jcn.2014.10.2.108
- de Greef BT, Hoeijmakers JG, Wolters EE, et al. No Fabry disease in patients presenting with isolated small fiber neuropathy. PLoS One 2016; 11(2):e0148316. doi:10.1371/journal.pone.0148316
- Souayah N, Ajroud-Driss S, Sander HW, Brannagan TH, Hays AP, Chin RL. Small fiber neuropathy following vaccination for rabies, varicella or Lyme disease. Vaccine 2009; 27(52):7322–7325. doi:10.1016/j.vaccine.2009.09.077
- Nolano M, Provitera V, Manganelli F, et al. Loss of cutaneous large and small fibers in naive and l-dopa–treated PD patients. Neurology 2017; 89(8):776–784. doi:10.1212/WNL.0000000000004274
- Zis P, Grünewald RA, Chaudhuri RK, Hadjivassiliou M. Peripheral neuropathy in idiopathic Parkinson’s disease: a systematic review. J Neurol Sci 2017; 378:204–209. doi:10.1016/j.jns.2017.05.023
- Gibbons CH, Freeman R. Treatment-induced neuropathy of diabetes: an acute, iatrogenic complication of diabetes. Brain 2015; 138(pt 1):43–52. doi:10.1093/brain/awu307
- Look AHEAD Research Group. Effects of a long-term lifestyle modification programme on peripheral neuropathy in overweight or obese adults with type 2 diabetes: the Look AHEAD study. Diabetologia 2017; 60(6):980–988. doi:10.1007/s00125-017-4253-z
- Culver DA, Dahan A, Bajorunas D, et al. Cibinetide improves corneal nerve fiber abundance in patients with sarcoidosis-associated small nerve fiber loss and neuropathic pain. Invest Ophthalmol Vis Sci 2017; 58(6):BIO52–BIO60. doi:10.1167/iovs.16-21291
- Bril V, England J, Franklin GM, et al; American Academy of Neurology; American Association of Neuromuscular and Electrodiagnostic Medicine; American Academy of Physical Medicine and Rehabilitation. Evidence-based guideline: treatment of painful diabetic neuropathy: report of the American Academy of Neurology, the American Association of Neuromuscular and Electrodiagnostic Medicine, and the American Academy of Physical Medicine and Rehabilitation. PM R 2011; 3(4):345–352.e21. doi:10.1016/j.pmrj.2011.03.008
- Gilron I, Bailey JM, Tu D, Holden RR, Jackson AC, Houlden RL. Nortriptyline and gabapentin, alone and in combination for neuropathic pain: a double-blind, randomised controlled crossover trial. Lancet 2009; 374(9697):1252–1261. doi:10.1016/S0140-6736(09)61081-3
- Ellis RJ, Toperoff W, Vaida F, et al. Smoked medicinal cannabis for neuropathic pain in HIV: a randomized, crossover clinical trial. Neuropsychopharmacology 2009; 34(3):672–680. doi:10.1038/npp.2008.120
- Wallace MS, Marcotte TD, Umlauf A, Gouaux B, Atkinson JH. Efficacy of inhaled cannabis on painful diabetic neuropathy. J Pain 2015; 16(7):616–627. doi:10.1016/j.jpain.2015.03.008
- Hoffman EM, Watson JC, St Sauver J, Staff NP, Klein CJ. Association of long-term opioid therapy with functional status, adverse outcomes, and mortality among patients with polyneuropathy. JAMA Neurol 2017; 74(7):773–779. doi:10.1001/jamaneurol.2017.0486
- Harati Y, Gooch C, Swenson M, et al. Double-blind randomized trial of tramadol for the treatment of the pain of diabetic neuropathy. Neurology 1998; 50(6):1842–1846. pmid:9633738
- Sima AA, Calvani M, Mehra M, Amato A; Acetyl-L-Carnitine Study Group. Acetyl-L-carnitine improves pain, nerve regeneration, and vibratory perception in patients with chronic diabetic neuropathy: an analysis of two randomized placebo-controlled trials. Diabetes Care 2005; 28(1):89–94. pmid:15616239
- Ziegler D, Hanefeld M, Ruhnau KJ, et al. Treatment of symptomatic diabetic peripheral neuropathy with the anti-oxidant alpha-lipoic acid. A 3-week multicentre randomized controlled trial (ALADIN Study). Diabetologia 1995; 38(12):1425–1433. pmid:8786016
- Scarpini E, Sacilotto G, Baron P, Cusini M, Scarlato G. Effect of acetyl-L-carnitine in the treatment of painful peripheral neuropathies in HIV+ patients. J Peripher Nerv Syst 1997; 2(3):250-252. pmid: 10975731
- Hershman DL, Unger JM, Crew KD, et al. Randomized double-blind placebo-controlled trial of acetyl-L-carnitine for the prevention of taxane-induced neuropathy in women undergoing adjuvant breast cancer therapy. J Clin Oncol 2013; 31(20):2627-2633. doi:10.1200/JCO.2012.44.8738
- Amara S. Oral glutamine for the prevention of chemotherapy-induced peripheral neuropathy. Ann Pharmacother 2008; 42(10):1481-1485. doi:10.1345/aph.1L179
- Huang JS, Wu CL, Fan CW, Chen WH, Yeh KY, Chang PH. Intravenous glutamine appears to reduce the severity of symptomatic platinum-induced neuropathy: a prospective randomized study. J Chemother 2015; 27(4):235-240. doi:10.1179/1973947815Y.0000000011
- Banafshe HR, Hamidi GA, Noureddini M, Mirhashemi SM, Mokhtari R, Shoferpour M. Effect of curcumin on diabetic peripheral neuropathic pain: possible involvement of opioid system. Eur J Pharmacol 2014; 723:202-206. doi:10.1016/j.ejphar.2013.11.033
- Mendonça LM, da Silva Machado C, Teixeira CC, de Freitas LA, Bianchi MD, Antunes LM. Curcumin reduces cisplatin-induced neurotoxicity in NGF-differentiated PC12 cells. Neurotoxicology 2013; 34:205-211. doi:10.1016/j.neuro.2012.09.011
- Wagner K, Lee KS, Yang J, Hammock BD. Epoxy fatty acids mediate analgesia in murine diabetic neuropathy. Eur J Pain 2017; 21(3):456-465. doi:10.1002/ejp.939
- Lewis EJ, Perkins BA, Lovblom LE, Bazinet RP, Wolever TMS, Bril V. Effect of omega-3 supplementation on neuropathy in type 1 diabetes: a 12-month pilot trial. Neurology 2017; 88(24):2294–2301. doi:10.1212/WNL.0000000000004033
- Hu D, Wang C, Li F, et al. A combined water extract of frankincense and myrrh alleviates neuropathic pain in mice via modulation of TRPV1. Neural Plast 2017; 2017:3710821. doi:10.1155/2017/3710821
- Tavee J, Rensel M, Planchon SM, Butler RS, Stone L. Effects of meditation on pain and quality of life in multiple sclerosis and peripheral neuropathy: a pilot study. Int J MS Care 2011; 13(4):163–168. doi:10.7224/1537-2073-13.4.163
- Khoshnoodi MA, Truelove S, Burakgazi A, Hoke A, Mammen AL, Polydefkis M. Longitudinal assessment of small fiber neuropathy: evidence of a non-length-dependent distal axonopathy. JAMA Neurol 2016; 73(6):684–690. doi:10.1001/jamaneurol.2016.0057
KEY POINTS
- Patients typically develop a symmetric “stocking-glove” pattern of sensory loss in the feet and hands.
- The diagnosis may be confirmed with skin biopsy for nerve fiber density, which can easily be done in a clinic setting with commercially available kits.
- Diabetes is the most common identifiable cause of small fiber neuropathy.
- Serologic testing can help uncover a vitamin deficiency or other potentially treatable condition.
- Antiepileptics, antidepressants, and topical agents are first-line drugs for managing pain.
Genitourinary syndrome of menopause in breast cancer survivors: Treatments are available
Many breast cancer survivors and women at high risk of breast cancer suffer from genitourinary syndrome of menopause (GSM), a term that encompasses any urinary, genital, or sexual dysfunction related to a hypoestrogenic state. Although GSM is usually caused by postmenopausal estrogen loss, it can also be caused by cancer treatments such as chemotherapy, radiation, and systemic endocrine therapy (eg, tamoxifen, aromatase inhibitors). These treatments can substantially decrease systemic estrogen levels, causing GSM symptoms that can profoundly worsen quality of life.
Managing GSM in these women poses a dilemma because systemic estrogen-containing therapies can increase the risk of breast cancer, and nonhormonal vaginal lubricants and moisturizers provide only minimal benefit. Fortunately, there are hormonal options, including locally applied estrogen, intravaginal dehydroepiandrosterone (DHEA), and estrogen receptor agonists/antagonists (ospemifene and bazedoxifene).
Here, we review the clinical management of GSM in breast cancer survivors and women at high risk of breast cancer and the efficacy and safety of available treatments, including their impact on breast cancer risk.
DRYNESS, IRRITATION, ATROPHY
The term GSM describes vulvovaginal and genitourinary symptoms associated with estrogen loss after menopause. Common symptoms are vaginal dryness, dyspareunia, irritation of genital skin, and pruritus.
LOCAL ESTROGEN THERAPY
Systemic estrogen therapy is widely used and effective for GSM, but there are concerns that it could increase the risk of breast cancer. After the Women’s Health Initiative in 2002 showed higher rates of cardiovascular disease and breast cancer with systemic estrogen-progestin use,5 the use of this hormone therapy declined by approximately 80%.6 Since then, healthcare providers have turned to local (ie, vaginal) estrogen therapies to manage GSM. These therapies have several advantages over systemic hormone therapy:
- Lower risk of adverse effects on the breast and cardiovascular system
- Greater efficacy in treating GSM
- In general, no need for progesterone when low-dose local estrogen is given to a woman with a uterus.7
Is locally applied estrogen systemically absorbed?
Despite these advantages, concerns remain as to whether vaginal estrogen therapy has adverse consequences associated with systemic absorption, particularly from atrophic vaginal tissues.
Santen,8 in a 2015 review of 33 studies, concluded that systemic absorption from low-dose vaginal estrogen is minimal, which provides some rationale for using it to treat vulvovaginal atrophy in postmenopausal women. This finding also suggests that the US Food and Drug Administration (FDA) “black box” warning of possible toxicities with vaginal estrogen is likely overstated, given that serum estrogen levels remained within normal postmenopausal levels.
Nevertheless, many providers are apprehensive about prescribing vaginal estrogen in women with a history of breast cancer because the threshold for systemic estrogen levels associated with breast cancer recurrence has not been established.
ACOG statement. In 2016, a committee of the American College of Obstetricians and Gynecologists cited data showing that low-dose vaginal estrogens do not result in sustained serum estrogen levels exceeding the normal postmenopausal range, and that the use of vaginal estrogens does not increase the risk of cancer recurrence.9 However, they recommend caution with vaginal estrogen use, especially in women with a history of estrogen-dependent breast cancer, reserving it for patients with GSM symptoms nonresponsive to nonhormonal treatment and specifying that it be used in low doses.
Vaginal estrogen formulations
Several types of locally applied estrogens are available, each with different properties and affinity for estrogen receptors. These include conjugated estrogens, 17-beta estradiol, estradiol acetate, and estradiol hemihydrate. Three delivery systems are FDA-approved: creams, rings, and tablets (Table 2).
Vaginal creams. Two vaginal creams are available, one (Estrace) containing 17-beta estradiol and the other (Premarin) containing conjugated estrogens.
The FDA-approved dosage for 17-beta estradiol is 2 to 4 g/day for 1 or 2 weeks, then gradually reduced to half the dose for a similar time. Maintenance dosing is 1 g 1 to 3 times per week. However, the ACOG statement notes that the FDA-approved dosages are higher than those proven to be effective and currently used in clinical practice, eg, 0.5 g twice a week.9
The FDA-approved dosage of conjugated estrogen cream for moderate to severe dyspareunia is 0.5 g daily for 21 days, then off for 7 days, or 0.5 g twice a week.
Vaginal tablets. The vaginal tablet Vagifem and its generic equivalent Yuvafem contain 10 µg of estradiol hemihydrate. The FDA-approved dosage is 10 µg daily for 2 weeks, followed by 10 µg twice a week, inserted into the lower third of the vagina. This dosage is significantly lower than that of estrogen creams.
Vaginal insert. A newly approved vaginal insert (Imvexxy) contains estradiol 4 µg (the lowest dose of vaginal estradiol available) or 10 µg, in a coconut oil vehicle. Its indications are for moderate to severe dyspareunia due to menopause and atrophic vaginitis due to menopause. A study cited in its package insert (www.accessdata.fda.gov/drugsatfda_docs/label/2018/208564s000lbl.pdf) showed that, in patients who used this product, systemic absorption of estradiol remained within the postmenopausal range. Its effects on breast cancer have not yet been studied.
Vaginal rings. Two vaginal rings are marketed. One (Estring) contains 17-beta estradiol, and the other (Femring) contains estradiol acetate. Only the 17-beta estradiol ring delivers a low dose to vaginal tissues, releasing 7.5 µg/day for 90 days. The estradiol acetate ring releases 0.05 mg/day or 0.10 mg/day and is a systemic treatment meant to be used with a progestin, not for local therapy.
VAGINAL ANDROGEN THERAPY: DHEA
After menopause, as the ovaries stop making estrogen from androstenedione, some production continues in other tissues, with DHEA as the primary precursor of androgens that are ultimately converted to estrogen. This has led to the theory that the cause of GSM is not estrogen deficiency but androgen deficiency. Evidence reviewed by Labrie et al11 shows that vulvovaginal atrophy is caused by decreased DHEA availability, which in turn causes sex steroid deficiency-related menopausal symptoms.11 Thus, it is reasonable to conclude that menopausal symptoms can be relieved by giving DHEA.
This theory has been borne out in clinical trials, in which DHEA in a vaginal tablet formulation increased the maturation of vaginal cells and lowered vaginal pH, leading to relief of GSM symptoms.12
The only DHEA product FDA-approved for treating GSM-related symptoms is prasterone (Intrarosa), indicated for moderate to severe dyspareunia due to vulvovaginal atrophy. The recommended dosing is a single 6.5-mg intravaginal tablet (0.5% prasterone) inserted nightly at bedtime. Its efficacy for treating hypoactive sexual desire disorder in postmenopausal women is being investigated.
Breast cancer implications
Because DHEA is converted to estrogen by aromatization, healthcare providers might hesitate to use it in women who have a history of hormone-sensitive cancer. Data on the safety of intravaginal DHEA use in breast cancer survivors are limited. However, studies have found that prasterone has highly beneficial effects on dyspareunia, vaginal dryness, and objective signs of vulvovaginal atrophy without significant drug-related adverse effects.12,13 In these studies, serum estrogen levels in women treated with DHEA were within the values observed in normal postmenopausal women. In addition, there are no aromatase enzymes in the endometrium, so even high doses of vaginal DHEA (in contrast to high doses of vaginal estrogen) will not stimulate the endometrium.
Clinically, this evidence indicates that DHEA exerts both estrogenic and androgenic activity in the vagina without increasing serum estrogen levels, making it a good alternative to topical estrogen therapy.
OSPEMIFENE: AN ESTROGEN RECEPTOR AGONIST/ANTAGONIST
Ospemifene (Osphena) is an estrogen receptor agonist/antagonist, a class of drugs previously called selective estrogen receptor modulators (SERMs). It is FDA-approved to treat moderate to severe dyspareunia secondary to vulvar and vaginal atrophy.
Ospemifene has unique estrogenic effects on the vaginal mucosa, having been shown to increase the number of epithelial cells, lower the vaginal pH, and decrease the percentage of parabasal cells seen on Papanicolaou smears after 12 weeks of use.14
Unlike tamoxifen, another drug of this class, ospemifene does not change the endometrial lining.14 Similarly, ospemifene acts as an estrogenic agonist in bone and, thus, has the potential for use in preventing and managing osteoporosis or for use in women at risk of fractures.
Breast cancer impact
In preclinical trials, ospemifene was found to have antiestrogenic effects on breast tissue, similar to those seen with tamoxifen.
In a model using human tumor grafts, ospemifene decreased tumor growth in mice implanted with estrogen receptor-positive breast cancer cells.15
In a mouse model using breast cancer cells that were biologically and histologically similar to those of humans, ospemifene had strong antiestrogenic effects on the breast tissue.16 The evidence suggests that ospemifene has a favorable effect on vulvar and vaginal atrophy.17
Ospemifene is FDA-approved to treat moderate to severe dyspareunia secondary to menopause. Recommended dosing is 60 mg/day orally with food.
Its antiestrogenic effects on breast tissue make it a promising option for women with a history of estrogen-receptor positive breast cancer. However, further study is needed to fully understand its effects on human breast tissue. According to the manufacturer’s package insert (www.osphena.com/files/pdf/osphena_prescribing_information.pdf), because the drug has not been adequately studied in women with breast cancer, it should not be used in women with known or suspected breast cancer or a history of breast cancer.
CONJUGATED ESTROGENS PLUS BAZEDOXIFENE
The combination of conjugated estrogens and bazedoxifene (Duavee) is a progesterone-free alternative for treating various menopausal symptoms. Bazedoxifene is another estrogen receptor agonist/antagonist, and it was added to counteract estrogen’s effects on the endometrium, thus replacing progesterone. This protective effect has been validated in clinical trials, which also found a favorable safety profile in breast tissue.18,19
SMART trials. The efficacy of this combination was studied in a series of large phase 3 multicenter trials called the SMART (Selective Estrogens, Menopause, and Response to Therapy) trials.20–23 Treated patients had markedly fewer vasomotor symptoms at 1 year, along with an increase in superficial cells and intermediate cells of the vaginal epithelium and a decrease in parabasal cells. They also had a substantial decrease in the incidence of dyspareunia.
Its effects on breast tissue were evaluated in the SMART-5 trial. Therapy had no net impact on breast density, suggesting that it has an estrogen-neutral effect on the breast.23
These results suggest that combined conjugated estrogens and bazedoxifene could be a noteworthy treatment option for GSM in women with a history of estrogen receptor-positive breast cancer, particularly in those with vasomotor symptoms and bone loss. However, the combination has not been studied specifically in breast cancer survivors.
Dosage. The FDA-approved dosing is 20 mg/0.45 mg per day orally to treat vasomotor symptoms, GSM, and osteoporosis in postmenopausal women with a uterus.
LASER THERAPY AND RADIOFREQUENCY HEAT: AN OFF-LABEL OPTION
Low-dose radiofrequency thermal therapy, delivered by carbon dioxide laser or by radiofrequency heat, has been used with some success to treat urinary stress incontinence and vaginal laxity in postpartum women. It may be an option for GSM, although it is not FDA-approved for this indication, and the FDA has recently issued a warning about it.24
Marketing literature promotes laser therapy as an effective option that stimulates vaginal connective tissue to produce new collagen, which then promotes improved blood flow and tissue regeneration for vaginal lubrication and elasticity.
A study comparing fractional carbon dioxide vaginal laser treatment and local estrogen therapy in postmenopausal women with vulvovaginal atrophy found that laser therapy was an effective treatment for vulvovaginal atrophy (dyspareunia, dryness, and burning), both alone and with local estrogen.25
Despite the promising effects of laser therapy for treating vulvovaginal atrophy in GSM, studies have not determined its short-term or long-term safety profile. Furthermore, laser therapy does not improve impaired sexual function, ie, decreased libido, arousal, and sexual satisfaction. Another important consideration is that the cost of laser therapy in 2017 was estimated to be $2,000 to $3,000 per treatment, not covered by healthcare insurance.
CLINICAL APPROACH
Symptoms of GSM are common in breast cancer survivors, both pre- and postmenopausal, especially those treated with tamoxifen or an aromatase inhibitor. Estimates are that 60% of postmenopausal breast cancer survivors and 40% of premenopausal breast cancer survivors suffer from GSM.26 Unfortunately, many women do not seek medical attention for their symptoms.
A variety of hormonal and nonhormonal options are available for these patients. We recommend an interdisciplinary approach to treatment, with the decision to use hormonal options made in collaboration with the patient’s oncologist and the patient herself, in an informed, shared decision-making process that takes into consideration the risks and possible benefits depending on the symptoms.
The first step in selecting a management plan for GSM symptoms in women with breast cancer is to conduct a thorough assessment to provide data for individualizing the care plan. The decision to use a hormonal option should be made in collaboration with a woman’s oncologist and should include an informed decision-making process during which the potential risks and benefits, including the breast cancer impact, are fully disclosed.
Alternatives to systemic estrogen
Vaginal estrogen is an effective and safe option to treat GSM in women with either estrogen receptor-negative or estrogen receptor-positive breast cancer. It often completely cures the symptoms without any noticeable increase in serum estrogen levels.
Vaginal DHEA therapy is a nonestrogen option shown to effectively treat GSM without increasing systemic levels of estrogen or testosterone. This profile makes vaginal DHEA therapy a particularly attractive treatment for symptoms of GSM in women at risk for breast cancer.
Use of an estrogen receptor agonist/antagonist in breast cancer survivors needs careful consideration. Ospemifene has antiestrogenic effects that make it a good option for women with bone loss and those at high risk for breast cancer, but it should not be used concurrently with tamoxifen or raloxifene. Additionally, ospemifene does not cause uterine hyperplasia, so it can be used in women with a uterus.
Although more study is needed, we do have options to improve the overall quality of life in breast cancer survivors with GSM.
- Lester J, Pahouja G, Andersen B, Lustberg M. Atrophic vaginitis in breast cancer survivors: a difficult survivorship issue. J Pers Med 2015; 5(2):50–66. doi:10.3390/jpm5020050
- Chin SN, Trinkaus M, Simmons C, et al. Prevalence and severity of urogenital symptoms in postmenopausal women receiving endocrine therapy for breast cancer. Clin Breast Cancer 2009; 9(2):108–117. doi:10.3816/CBC.2009.n.020
- Fallowfield L, Cella D, Cuzick J, Francis S, Locker G, Howell A. Quality of life of postmenopausal women in the Arimidex, Tamoxifen, Alone or in Combination (ATAC) adjuvant breast cancer trial. J Clin Oncol 2004; 22(21):4261–4271. doi:10.1200/JCO.2004.08.029
- Cella D, Fallowfield LJ. Recognition and management of treatment-related side effects for breast cancer patients receiving adjuvant endocrine therapy. Breast Cancer Res Treat 2008; 107(2):167–180. doi:10.1007/s10549-007-9548-1
- 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(3):321–333. pmid:12117397
- Tsai SA, Stefanick ML, Stafford RS. Trends in menopausal hormone therapy use of US office-based physicians, 2000–2009. Menopause 2011; 18(4):385–392. doi:10.1097/gme.0b013e3181f43404
- North American Menopause Society. Management of symptomatic vulvovaginal atrophy: 2013 position statement of The North American Menopause Society. Menopause 2013; 20(9):888–902. doi:10.1097/GME.0b013e3182a122c2
- Santen RJ. Vaginal administration of estradiol: effects of dose, preparation and timing on plasma estradiol levels. Climacteric 2015; 18(2):121–134. doi:10.3109/13697137.2014.947254
- American College of Obstetricians and Gynecologists Committee on Gynecologic Practice, Farrell R. ACOG Committee Opinion No. 659: the use of vaginal estrogen in women with a history of estrogen-dependent breast cancer. Obstet Gynecol 2016; 127(3):e93–e96. doi:10.1097/AOG.0000000000001351
- Santoro N, Epperson CN, Mathews SB. Menopausal symptoms and their management. Endocrinol Metab Clin North Am 2015; 44(3):497–515. doi:10.1016/j.ecl.2015.05.001
- Labrie F, Archer DF, Martel C, Vaillancourt M, Montesino M. Combined data of intravaginal prasterone against vulvovaginal atrophy of menopause. Menopause 2017; 24(11):1246–1256. doi:10.1097/GME.0000000000000910
- Labrie F, Archer D, Bouchard C, et al. Serum steroid levels during 12-week intravaginal dehydroepiandrosterone administration. Menopause 2009; 16(5):897–906. doi:10.1097/gme.0b013e31819e8930
- Labrie F, Cusan L, Gomez JL, et al. Effect of intravaginal DHEA on serum DHEA and eleven of its metabolites in postmenopausal women. J Steroid Biochem Mol Biol 2008; 111(3-5):178–194. doi:10.1016/j.jsbmb.2008.06.003
- Soe LH, Wurz GT, Kao CJ, Degregorio MW. Ospemifene for the treatment of dyspareunia associated with vulvar and vaginal atrophy: potential benefits in bone and breast. Int J Womens Health 2013; 5:605–611. doi:10.2147/IJWH.S39146
- Taras TL, Wurz GT, DeGregorio MW. In vitro and in vivo biologic effects of ospemifene (FC-1271a) in breast cancer. J Steroid Biochem Mol Biol 2001; 77(4–5):271–279. pmid:11457665
- Wurz GT, Read KC, Marchisano-Karpman C, et al. Ospemifene inhibits the growth of dimethylbenzanthracene-induced mammary tumors in Sencar mice. J Steroid Biochem Mol Biol 2005; 97(3):230–240. doi:10.1016/j.jsbmb.2005.06.027
- Archer DF, Carr BR, Pinkerton JV, Taylor HS, Constantine GD. Effects of ospemifene on the female reproductive and urinary tracts: translation from preclinical models into clinical evidence. Menopause 2015; 22(7):786–796. doi:10.1097/GME.0000000000000365
- Mirkin S, Pickar JH. Management of osteoporosis and menopausal symptoms: focus on bazedoxifene/conjugated estrogen combination. Int J Womens Health 2013; 5:465–475. doi:10.2147/IJWH.S39455
- Kagan R, Goldstein SR, Pickar JH, Komm BS. Patient considerations in the management of menopausal symptoms: role of conjugated estrogens with bazedoxifene. Ther Clin Risk Manag 2016; 12:549–562. doi:10.2147/TCRM.S63833
- Lobo RA, Pinkerton JV, Gass ML, et al. Evaluation of bazedoxifene/conjugated estrogens for the treatment of menopausal symptoms and effects on metabolic parameters and overall safety profile. Fertil Steril 2009; 92(3):1025–1038. doi:10.1016/j.fertnstert.2009.03.113
- Pinkerton JV, Utian WH, Constantine GD, Olivier S, Pickar JH. Relief of vasomotor symptoms with the tissue-selective estrogen complex containing bazedoxifene/conjugated estrogens: a randomized, controlled trial. Menopause 2009; 16(6):1116–1124. doi:10.1097/gme.0b013e3181a7df0d
- Kagan R, Williams RS, Pan K, Mirkin S, Pickar JH. A randomized, placebo- and active-controlled trial of bazedoxifene/conjugated estrogens for treatment of moderate to severe vulvar/vaginal atrophy in postmenopausal women. Menopause 2010; 17(2):281–289. doi:10.1097/GME.0b013e3181b7c65f
- Pinkerton JV, Harvey JA, Pan K, et al. Breast effects of bazedoxifene-conjugated estrogens: a randomized controlled trial. Obstet Gynecol 2013; 121(5):959–968. doi:10.1097/AOG.0b013e31828c5974
- FDA. U.S. Food & Drug Administration. FDA Statement. Statement from FDA Commissioner Scott Gottlieb, M.D., on efforts to safeguard women’s health from deceptive health claims and significant risks related to devices marketed for use in medical procedures for “vaginal rejuvenation.” www.fda.gov/NewsEvents/Newsroom/PressAnnouncements/ucm615130.htm. Accessed August 20, 2018.
- Cruz VL, Steiner ML, Pompei LM, et al. Randomized, double-blind, placebo-controlled clinical trial for evaluating the efficacy of fractional CO2 laser compared with topical estriol in the treatment of vaginal atrophy in postmenopausal women. Menopause 2018; 25(1):21–28. doi:10.1097/GME.0000000000000955
- Biglia N, Bounous VE, D’Alonzo M, et al. Vaginal atrophy in breast cancer survivors: attitude and approaches among oncologists. Clin Breast Cancer 2017; 17(8):611–617. doi:10.1016/j.clbc.2017.05.008
Many breast cancer survivors and women at high risk of breast cancer suffer from genitourinary syndrome of menopause (GSM), a term that encompasses any urinary, genital, or sexual dysfunction related to a hypoestrogenic state. Although GSM is usually caused by postmenopausal estrogen loss, it can also be caused by cancer treatments such as chemotherapy, radiation, and systemic endocrine therapy (eg, tamoxifen, aromatase inhibitors). These treatments can substantially decrease systemic estrogen levels, causing GSM symptoms that can profoundly worsen quality of life.
Managing GSM in these women poses a dilemma because systemic estrogen-containing therapies can increase the risk of breast cancer, and nonhormonal vaginal lubricants and moisturizers provide only minimal benefit. Fortunately, there are hormonal options, including locally applied estrogen, intravaginal dehydroepiandrosterone (DHEA), and estrogen receptor agonists/antagonists (ospemifene and bazedoxifene).
Here, we review the clinical management of GSM in breast cancer survivors and women at high risk of breast cancer and the efficacy and safety of available treatments, including their impact on breast cancer risk.
DRYNESS, IRRITATION, ATROPHY
The term GSM describes vulvovaginal and genitourinary symptoms associated with estrogen loss after menopause. Common symptoms are vaginal dryness, dyspareunia, irritation of genital skin, and pruritus.
LOCAL ESTROGEN THERAPY
Systemic estrogen therapy is widely used and effective for GSM, but there are concerns that it could increase the risk of breast cancer. After the Women’s Health Initiative in 2002 showed higher rates of cardiovascular disease and breast cancer with systemic estrogen-progestin use,5 the use of this hormone therapy declined by approximately 80%.6 Since then, healthcare providers have turned to local (ie, vaginal) estrogen therapies to manage GSM. These therapies have several advantages over systemic hormone therapy:
- Lower risk of adverse effects on the breast and cardiovascular system
- Greater efficacy in treating GSM
- In general, no need for progesterone when low-dose local estrogen is given to a woman with a uterus.7
Is locally applied estrogen systemically absorbed?
Despite these advantages, concerns remain as to whether vaginal estrogen therapy has adverse consequences associated with systemic absorption, particularly from atrophic vaginal tissues.
Santen,8 in a 2015 review of 33 studies, concluded that systemic absorption from low-dose vaginal estrogen is minimal, which provides some rationale for using it to treat vulvovaginal atrophy in postmenopausal women. This finding also suggests that the US Food and Drug Administration (FDA) “black box” warning of possible toxicities with vaginal estrogen is likely overstated, given that serum estrogen levels remained within normal postmenopausal levels.
Nevertheless, many providers are apprehensive about prescribing vaginal estrogen in women with a history of breast cancer because the threshold for systemic estrogen levels associated with breast cancer recurrence has not been established.
ACOG statement. In 2016, a committee of the American College of Obstetricians and Gynecologists cited data showing that low-dose vaginal estrogens do not result in sustained serum estrogen levels exceeding the normal postmenopausal range, and that the use of vaginal estrogens does not increase the risk of cancer recurrence.9 However, they recommend caution with vaginal estrogen use, especially in women with a history of estrogen-dependent breast cancer, reserving it for patients with GSM symptoms nonresponsive to nonhormonal treatment and specifying that it be used in low doses.
Vaginal estrogen formulations
Several types of locally applied estrogens are available, each with different properties and affinity for estrogen receptors. These include conjugated estrogens, 17-beta estradiol, estradiol acetate, and estradiol hemihydrate. Three delivery systems are FDA-approved: creams, rings, and tablets (Table 2).
Vaginal creams. Two vaginal creams are available, one (Estrace) containing 17-beta estradiol and the other (Premarin) containing conjugated estrogens.
The FDA-approved dosage for 17-beta estradiol is 2 to 4 g/day for 1 or 2 weeks, then gradually reduced to half the dose for a similar time. Maintenance dosing is 1 g 1 to 3 times per week. However, the ACOG statement notes that the FDA-approved dosages are higher than those proven to be effective and currently used in clinical practice, eg, 0.5 g twice a week.9
The FDA-approved dosage of conjugated estrogen cream for moderate to severe dyspareunia is 0.5 g daily for 21 days, then off for 7 days, or 0.5 g twice a week.
Vaginal tablets. The vaginal tablet Vagifem and its generic equivalent Yuvafem contain 10 µg of estradiol hemihydrate. The FDA-approved dosage is 10 µg daily for 2 weeks, followed by 10 µg twice a week, inserted into the lower third of the vagina. This dosage is significantly lower than that of estrogen creams.
Vaginal insert. A newly approved vaginal insert (Imvexxy) contains estradiol 4 µg (the lowest dose of vaginal estradiol available) or 10 µg, in a coconut oil vehicle. Its indications are for moderate to severe dyspareunia due to menopause and atrophic vaginitis due to menopause. A study cited in its package insert (www.accessdata.fda.gov/drugsatfda_docs/label/2018/208564s000lbl.pdf) showed that, in patients who used this product, systemic absorption of estradiol remained within the postmenopausal range. Its effects on breast cancer have not yet been studied.
Vaginal rings. Two vaginal rings are marketed. One (Estring) contains 17-beta estradiol, and the other (Femring) contains estradiol acetate. Only the 17-beta estradiol ring delivers a low dose to vaginal tissues, releasing 7.5 µg/day for 90 days. The estradiol acetate ring releases 0.05 mg/day or 0.10 mg/day and is a systemic treatment meant to be used with a progestin, not for local therapy.
VAGINAL ANDROGEN THERAPY: DHEA
After menopause, as the ovaries stop making estrogen from androstenedione, some production continues in other tissues, with DHEA as the primary precursor of androgens that are ultimately converted to estrogen. This has led to the theory that the cause of GSM is not estrogen deficiency but androgen deficiency. Evidence reviewed by Labrie et al11 shows that vulvovaginal atrophy is caused by decreased DHEA availability, which in turn causes sex steroid deficiency-related menopausal symptoms.11 Thus, it is reasonable to conclude that menopausal symptoms can be relieved by giving DHEA.
This theory has been borne out in clinical trials, in which DHEA in a vaginal tablet formulation increased the maturation of vaginal cells and lowered vaginal pH, leading to relief of GSM symptoms.12
The only DHEA product FDA-approved for treating GSM-related symptoms is prasterone (Intrarosa), indicated for moderate to severe dyspareunia due to vulvovaginal atrophy. The recommended dosing is a single 6.5-mg intravaginal tablet (0.5% prasterone) inserted nightly at bedtime. Its efficacy for treating hypoactive sexual desire disorder in postmenopausal women is being investigated.
Breast cancer implications
Because DHEA is converted to estrogen by aromatization, healthcare providers might hesitate to use it in women who have a history of hormone-sensitive cancer. Data on the safety of intravaginal DHEA use in breast cancer survivors are limited. However, studies have found that prasterone has highly beneficial effects on dyspareunia, vaginal dryness, and objective signs of vulvovaginal atrophy without significant drug-related adverse effects.12,13 In these studies, serum estrogen levels in women treated with DHEA were within the values observed in normal postmenopausal women. In addition, there are no aromatase enzymes in the endometrium, so even high doses of vaginal DHEA (in contrast to high doses of vaginal estrogen) will not stimulate the endometrium.
Clinically, this evidence indicates that DHEA exerts both estrogenic and androgenic activity in the vagina without increasing serum estrogen levels, making it a good alternative to topical estrogen therapy.
OSPEMIFENE: AN ESTROGEN RECEPTOR AGONIST/ANTAGONIST
Ospemifene (Osphena) is an estrogen receptor agonist/antagonist, a class of drugs previously called selective estrogen receptor modulators (SERMs). It is FDA-approved to treat moderate to severe dyspareunia secondary to vulvar and vaginal atrophy.
Ospemifene has unique estrogenic effects on the vaginal mucosa, having been shown to increase the number of epithelial cells, lower the vaginal pH, and decrease the percentage of parabasal cells seen on Papanicolaou smears after 12 weeks of use.14
Unlike tamoxifen, another drug of this class, ospemifene does not change the endometrial lining.14 Similarly, ospemifene acts as an estrogenic agonist in bone and, thus, has the potential for use in preventing and managing osteoporosis or for use in women at risk of fractures.
Breast cancer impact
In preclinical trials, ospemifene was found to have antiestrogenic effects on breast tissue, similar to those seen with tamoxifen.
In a model using human tumor grafts, ospemifene decreased tumor growth in mice implanted with estrogen receptor-positive breast cancer cells.15
In a mouse model using breast cancer cells that were biologically and histologically similar to those of humans, ospemifene had strong antiestrogenic effects on the breast tissue.16 The evidence suggests that ospemifene has a favorable effect on vulvar and vaginal atrophy.17
Ospemifene is FDA-approved to treat moderate to severe dyspareunia secondary to menopause. Recommended dosing is 60 mg/day orally with food.
Its antiestrogenic effects on breast tissue make it a promising option for women with a history of estrogen-receptor positive breast cancer. However, further study is needed to fully understand its effects on human breast tissue. According to the manufacturer’s package insert (www.osphena.com/files/pdf/osphena_prescribing_information.pdf), because the drug has not been adequately studied in women with breast cancer, it should not be used in women with known or suspected breast cancer or a history of breast cancer.
CONJUGATED ESTROGENS PLUS BAZEDOXIFENE
The combination of conjugated estrogens and bazedoxifene (Duavee) is a progesterone-free alternative for treating various menopausal symptoms. Bazedoxifene is another estrogen receptor agonist/antagonist, and it was added to counteract estrogen’s effects on the endometrium, thus replacing progesterone. This protective effect has been validated in clinical trials, which also found a favorable safety profile in breast tissue.18,19
SMART trials. The efficacy of this combination was studied in a series of large phase 3 multicenter trials called the SMART (Selective Estrogens, Menopause, and Response to Therapy) trials.20–23 Treated patients had markedly fewer vasomotor symptoms at 1 year, along with an increase in superficial cells and intermediate cells of the vaginal epithelium and a decrease in parabasal cells. They also had a substantial decrease in the incidence of dyspareunia.
Its effects on breast tissue were evaluated in the SMART-5 trial. Therapy had no net impact on breast density, suggesting that it has an estrogen-neutral effect on the breast.23
These results suggest that combined conjugated estrogens and bazedoxifene could be a noteworthy treatment option for GSM in women with a history of estrogen receptor-positive breast cancer, particularly in those with vasomotor symptoms and bone loss. However, the combination has not been studied specifically in breast cancer survivors.
Dosage. The FDA-approved dosing is 20 mg/0.45 mg per day orally to treat vasomotor symptoms, GSM, and osteoporosis in postmenopausal women with a uterus.
LASER THERAPY AND RADIOFREQUENCY HEAT: AN OFF-LABEL OPTION
Low-dose radiofrequency thermal therapy, delivered by carbon dioxide laser or by radiofrequency heat, has been used with some success to treat urinary stress incontinence and vaginal laxity in postpartum women. It may be an option for GSM, although it is not FDA-approved for this indication, and the FDA has recently issued a warning about it.24
Marketing literature promotes laser therapy as an effective option that stimulates vaginal connective tissue to produce new collagen, which then promotes improved blood flow and tissue regeneration for vaginal lubrication and elasticity.
A study comparing fractional carbon dioxide vaginal laser treatment and local estrogen therapy in postmenopausal women with vulvovaginal atrophy found that laser therapy was an effective treatment for vulvovaginal atrophy (dyspareunia, dryness, and burning), both alone and with local estrogen.25
Despite the promising effects of laser therapy for treating vulvovaginal atrophy in GSM, studies have not determined its short-term or long-term safety profile. Furthermore, laser therapy does not improve impaired sexual function, ie, decreased libido, arousal, and sexual satisfaction. Another important consideration is that the cost of laser therapy in 2017 was estimated to be $2,000 to $3,000 per treatment, not covered by healthcare insurance.
CLINICAL APPROACH
Symptoms of GSM are common in breast cancer survivors, both pre- and postmenopausal, especially those treated with tamoxifen or an aromatase inhibitor. Estimates are that 60% of postmenopausal breast cancer survivors and 40% of premenopausal breast cancer survivors suffer from GSM.26 Unfortunately, many women do not seek medical attention for their symptoms.
A variety of hormonal and nonhormonal options are available for these patients. We recommend an interdisciplinary approach to treatment, with the decision to use hormonal options made in collaboration with the patient’s oncologist and the patient herself, in an informed, shared decision-making process that takes into consideration the risks and possible benefits depending on the symptoms.
The first step in selecting a management plan for GSM symptoms in women with breast cancer is to conduct a thorough assessment to provide data for individualizing the care plan. The decision to use a hormonal option should be made in collaboration with a woman’s oncologist and should include an informed decision-making process during which the potential risks and benefits, including the breast cancer impact, are fully disclosed.
Alternatives to systemic estrogen
Vaginal estrogen is an effective and safe option to treat GSM in women with either estrogen receptor-negative or estrogen receptor-positive breast cancer. It often completely cures the symptoms without any noticeable increase in serum estrogen levels.
Vaginal DHEA therapy is a nonestrogen option shown to effectively treat GSM without increasing systemic levels of estrogen or testosterone. This profile makes vaginal DHEA therapy a particularly attractive treatment for symptoms of GSM in women at risk for breast cancer.
Use of an estrogen receptor agonist/antagonist in breast cancer survivors needs careful consideration. Ospemifene has antiestrogenic effects that make it a good option for women with bone loss and those at high risk for breast cancer, but it should not be used concurrently with tamoxifen or raloxifene. Additionally, ospemifene does not cause uterine hyperplasia, so it can be used in women with a uterus.
Although more study is needed, we do have options to improve the overall quality of life in breast cancer survivors with GSM.
Many breast cancer survivors and women at high risk of breast cancer suffer from genitourinary syndrome of menopause (GSM), a term that encompasses any urinary, genital, or sexual dysfunction related to a hypoestrogenic state. Although GSM is usually caused by postmenopausal estrogen loss, it can also be caused by cancer treatments such as chemotherapy, radiation, and systemic endocrine therapy (eg, tamoxifen, aromatase inhibitors). These treatments can substantially decrease systemic estrogen levels, causing GSM symptoms that can profoundly worsen quality of life.
Managing GSM in these women poses a dilemma because systemic estrogen-containing therapies can increase the risk of breast cancer, and nonhormonal vaginal lubricants and moisturizers provide only minimal benefit. Fortunately, there are hormonal options, including locally applied estrogen, intravaginal dehydroepiandrosterone (DHEA), and estrogen receptor agonists/antagonists (ospemifene and bazedoxifene).
Here, we review the clinical management of GSM in breast cancer survivors and women at high risk of breast cancer and the efficacy and safety of available treatments, including their impact on breast cancer risk.
DRYNESS, IRRITATION, ATROPHY
The term GSM describes vulvovaginal and genitourinary symptoms associated with estrogen loss after menopause. Common symptoms are vaginal dryness, dyspareunia, irritation of genital skin, and pruritus.
LOCAL ESTROGEN THERAPY
Systemic estrogen therapy is widely used and effective for GSM, but there are concerns that it could increase the risk of breast cancer. After the Women’s Health Initiative in 2002 showed higher rates of cardiovascular disease and breast cancer with systemic estrogen-progestin use,5 the use of this hormone therapy declined by approximately 80%.6 Since then, healthcare providers have turned to local (ie, vaginal) estrogen therapies to manage GSM. These therapies have several advantages over systemic hormone therapy:
- Lower risk of adverse effects on the breast and cardiovascular system
- Greater efficacy in treating GSM
- In general, no need for progesterone when low-dose local estrogen is given to a woman with a uterus.7
Is locally applied estrogen systemically absorbed?
Despite these advantages, concerns remain as to whether vaginal estrogen therapy has adverse consequences associated with systemic absorption, particularly from atrophic vaginal tissues.
Santen,8 in a 2015 review of 33 studies, concluded that systemic absorption from low-dose vaginal estrogen is minimal, which provides some rationale for using it to treat vulvovaginal atrophy in postmenopausal women. This finding also suggests that the US Food and Drug Administration (FDA) “black box” warning of possible toxicities with vaginal estrogen is likely overstated, given that serum estrogen levels remained within normal postmenopausal levels.
Nevertheless, many providers are apprehensive about prescribing vaginal estrogen in women with a history of breast cancer because the threshold for systemic estrogen levels associated with breast cancer recurrence has not been established.
ACOG statement. In 2016, a committee of the American College of Obstetricians and Gynecologists cited data showing that low-dose vaginal estrogens do not result in sustained serum estrogen levels exceeding the normal postmenopausal range, and that the use of vaginal estrogens does not increase the risk of cancer recurrence.9 However, they recommend caution with vaginal estrogen use, especially in women with a history of estrogen-dependent breast cancer, reserving it for patients with GSM symptoms nonresponsive to nonhormonal treatment and specifying that it be used in low doses.
Vaginal estrogen formulations
Several types of locally applied estrogens are available, each with different properties and affinity for estrogen receptors. These include conjugated estrogens, 17-beta estradiol, estradiol acetate, and estradiol hemihydrate. Three delivery systems are FDA-approved: creams, rings, and tablets (Table 2).
Vaginal creams. Two vaginal creams are available, one (Estrace) containing 17-beta estradiol and the other (Premarin) containing conjugated estrogens.
The FDA-approved dosage for 17-beta estradiol is 2 to 4 g/day for 1 or 2 weeks, then gradually reduced to half the dose for a similar time. Maintenance dosing is 1 g 1 to 3 times per week. However, the ACOG statement notes that the FDA-approved dosages are higher than those proven to be effective and currently used in clinical practice, eg, 0.5 g twice a week.9
The FDA-approved dosage of conjugated estrogen cream for moderate to severe dyspareunia is 0.5 g daily for 21 days, then off for 7 days, or 0.5 g twice a week.
Vaginal tablets. The vaginal tablet Vagifem and its generic equivalent Yuvafem contain 10 µg of estradiol hemihydrate. The FDA-approved dosage is 10 µg daily for 2 weeks, followed by 10 µg twice a week, inserted into the lower third of the vagina. This dosage is significantly lower than that of estrogen creams.
Vaginal insert. A newly approved vaginal insert (Imvexxy) contains estradiol 4 µg (the lowest dose of vaginal estradiol available) or 10 µg, in a coconut oil vehicle. Its indications are for moderate to severe dyspareunia due to menopause and atrophic vaginitis due to menopause. A study cited in its package insert (www.accessdata.fda.gov/drugsatfda_docs/label/2018/208564s000lbl.pdf) showed that, in patients who used this product, systemic absorption of estradiol remained within the postmenopausal range. Its effects on breast cancer have not yet been studied.
Vaginal rings. Two vaginal rings are marketed. One (Estring) contains 17-beta estradiol, and the other (Femring) contains estradiol acetate. Only the 17-beta estradiol ring delivers a low dose to vaginal tissues, releasing 7.5 µg/day for 90 days. The estradiol acetate ring releases 0.05 mg/day or 0.10 mg/day and is a systemic treatment meant to be used with a progestin, not for local therapy.
VAGINAL ANDROGEN THERAPY: DHEA
After menopause, as the ovaries stop making estrogen from androstenedione, some production continues in other tissues, with DHEA as the primary precursor of androgens that are ultimately converted to estrogen. This has led to the theory that the cause of GSM is not estrogen deficiency but androgen deficiency. Evidence reviewed by Labrie et al11 shows that vulvovaginal atrophy is caused by decreased DHEA availability, which in turn causes sex steroid deficiency-related menopausal symptoms.11 Thus, it is reasonable to conclude that menopausal symptoms can be relieved by giving DHEA.
This theory has been borne out in clinical trials, in which DHEA in a vaginal tablet formulation increased the maturation of vaginal cells and lowered vaginal pH, leading to relief of GSM symptoms.12
The only DHEA product FDA-approved for treating GSM-related symptoms is prasterone (Intrarosa), indicated for moderate to severe dyspareunia due to vulvovaginal atrophy. The recommended dosing is a single 6.5-mg intravaginal tablet (0.5% prasterone) inserted nightly at bedtime. Its efficacy for treating hypoactive sexual desire disorder in postmenopausal women is being investigated.
Breast cancer implications
Because DHEA is converted to estrogen by aromatization, healthcare providers might hesitate to use it in women who have a history of hormone-sensitive cancer. Data on the safety of intravaginal DHEA use in breast cancer survivors are limited. However, studies have found that prasterone has highly beneficial effects on dyspareunia, vaginal dryness, and objective signs of vulvovaginal atrophy without significant drug-related adverse effects.12,13 In these studies, serum estrogen levels in women treated with DHEA were within the values observed in normal postmenopausal women. In addition, there are no aromatase enzymes in the endometrium, so even high doses of vaginal DHEA (in contrast to high doses of vaginal estrogen) will not stimulate the endometrium.
Clinically, this evidence indicates that DHEA exerts both estrogenic and androgenic activity in the vagina without increasing serum estrogen levels, making it a good alternative to topical estrogen therapy.
OSPEMIFENE: AN ESTROGEN RECEPTOR AGONIST/ANTAGONIST
Ospemifene (Osphena) is an estrogen receptor agonist/antagonist, a class of drugs previously called selective estrogen receptor modulators (SERMs). It is FDA-approved to treat moderate to severe dyspareunia secondary to vulvar and vaginal atrophy.
Ospemifene has unique estrogenic effects on the vaginal mucosa, having been shown to increase the number of epithelial cells, lower the vaginal pH, and decrease the percentage of parabasal cells seen on Papanicolaou smears after 12 weeks of use.14
Unlike tamoxifen, another drug of this class, ospemifene does not change the endometrial lining.14 Similarly, ospemifene acts as an estrogenic agonist in bone and, thus, has the potential for use in preventing and managing osteoporosis or for use in women at risk of fractures.
Breast cancer impact
In preclinical trials, ospemifene was found to have antiestrogenic effects on breast tissue, similar to those seen with tamoxifen.
In a model using human tumor grafts, ospemifene decreased tumor growth in mice implanted with estrogen receptor-positive breast cancer cells.15
In a mouse model using breast cancer cells that were biologically and histologically similar to those of humans, ospemifene had strong antiestrogenic effects on the breast tissue.16 The evidence suggests that ospemifene has a favorable effect on vulvar and vaginal atrophy.17
Ospemifene is FDA-approved to treat moderate to severe dyspareunia secondary to menopause. Recommended dosing is 60 mg/day orally with food.
Its antiestrogenic effects on breast tissue make it a promising option for women with a history of estrogen-receptor positive breast cancer. However, further study is needed to fully understand its effects on human breast tissue. According to the manufacturer’s package insert (www.osphena.com/files/pdf/osphena_prescribing_information.pdf), because the drug has not been adequately studied in women with breast cancer, it should not be used in women with known or suspected breast cancer or a history of breast cancer.
CONJUGATED ESTROGENS PLUS BAZEDOXIFENE
The combination of conjugated estrogens and bazedoxifene (Duavee) is a progesterone-free alternative for treating various menopausal symptoms. Bazedoxifene is another estrogen receptor agonist/antagonist, and it was added to counteract estrogen’s effects on the endometrium, thus replacing progesterone. This protective effect has been validated in clinical trials, which also found a favorable safety profile in breast tissue.18,19
SMART trials. The efficacy of this combination was studied in a series of large phase 3 multicenter trials called the SMART (Selective Estrogens, Menopause, and Response to Therapy) trials.20–23 Treated patients had markedly fewer vasomotor symptoms at 1 year, along with an increase in superficial cells and intermediate cells of the vaginal epithelium and a decrease in parabasal cells. They also had a substantial decrease in the incidence of dyspareunia.
Its effects on breast tissue were evaluated in the SMART-5 trial. Therapy had no net impact on breast density, suggesting that it has an estrogen-neutral effect on the breast.23
These results suggest that combined conjugated estrogens and bazedoxifene could be a noteworthy treatment option for GSM in women with a history of estrogen receptor-positive breast cancer, particularly in those with vasomotor symptoms and bone loss. However, the combination has not been studied specifically in breast cancer survivors.
Dosage. The FDA-approved dosing is 20 mg/0.45 mg per day orally to treat vasomotor symptoms, GSM, and osteoporosis in postmenopausal women with a uterus.
LASER THERAPY AND RADIOFREQUENCY HEAT: AN OFF-LABEL OPTION
Low-dose radiofrequency thermal therapy, delivered by carbon dioxide laser or by radiofrequency heat, has been used with some success to treat urinary stress incontinence and vaginal laxity in postpartum women. It may be an option for GSM, although it is not FDA-approved for this indication, and the FDA has recently issued a warning about it.24
Marketing literature promotes laser therapy as an effective option that stimulates vaginal connective tissue to produce new collagen, which then promotes improved blood flow and tissue regeneration for vaginal lubrication and elasticity.
A study comparing fractional carbon dioxide vaginal laser treatment and local estrogen therapy in postmenopausal women with vulvovaginal atrophy found that laser therapy was an effective treatment for vulvovaginal atrophy (dyspareunia, dryness, and burning), both alone and with local estrogen.25
Despite the promising effects of laser therapy for treating vulvovaginal atrophy in GSM, studies have not determined its short-term or long-term safety profile. Furthermore, laser therapy does not improve impaired sexual function, ie, decreased libido, arousal, and sexual satisfaction. Another important consideration is that the cost of laser therapy in 2017 was estimated to be $2,000 to $3,000 per treatment, not covered by healthcare insurance.
CLINICAL APPROACH
Symptoms of GSM are common in breast cancer survivors, both pre- and postmenopausal, especially those treated with tamoxifen or an aromatase inhibitor. Estimates are that 60% of postmenopausal breast cancer survivors and 40% of premenopausal breast cancer survivors suffer from GSM.26 Unfortunately, many women do not seek medical attention for their symptoms.
A variety of hormonal and nonhormonal options are available for these patients. We recommend an interdisciplinary approach to treatment, with the decision to use hormonal options made in collaboration with the patient’s oncologist and the patient herself, in an informed, shared decision-making process that takes into consideration the risks and possible benefits depending on the symptoms.
The first step in selecting a management plan for GSM symptoms in women with breast cancer is to conduct a thorough assessment to provide data for individualizing the care plan. The decision to use a hormonal option should be made in collaboration with a woman’s oncologist and should include an informed decision-making process during which the potential risks and benefits, including the breast cancer impact, are fully disclosed.
Alternatives to systemic estrogen
Vaginal estrogen is an effective and safe option to treat GSM in women with either estrogen receptor-negative or estrogen receptor-positive breast cancer. It often completely cures the symptoms without any noticeable increase in serum estrogen levels.
Vaginal DHEA therapy is a nonestrogen option shown to effectively treat GSM without increasing systemic levels of estrogen or testosterone. This profile makes vaginal DHEA therapy a particularly attractive treatment for symptoms of GSM in women at risk for breast cancer.
Use of an estrogen receptor agonist/antagonist in breast cancer survivors needs careful consideration. Ospemifene has antiestrogenic effects that make it a good option for women with bone loss and those at high risk for breast cancer, but it should not be used concurrently with tamoxifen or raloxifene. Additionally, ospemifene does not cause uterine hyperplasia, so it can be used in women with a uterus.
Although more study is needed, we do have options to improve the overall quality of life in breast cancer survivors with GSM.
- Lester J, Pahouja G, Andersen B, Lustberg M. Atrophic vaginitis in breast cancer survivors: a difficult survivorship issue. J Pers Med 2015; 5(2):50–66. doi:10.3390/jpm5020050
- Chin SN, Trinkaus M, Simmons C, et al. Prevalence and severity of urogenital symptoms in postmenopausal women receiving endocrine therapy for breast cancer. Clin Breast Cancer 2009; 9(2):108–117. doi:10.3816/CBC.2009.n.020
- Fallowfield L, Cella D, Cuzick J, Francis S, Locker G, Howell A. Quality of life of postmenopausal women in the Arimidex, Tamoxifen, Alone or in Combination (ATAC) adjuvant breast cancer trial. J Clin Oncol 2004; 22(21):4261–4271. doi:10.1200/JCO.2004.08.029
- Cella D, Fallowfield LJ. Recognition and management of treatment-related side effects for breast cancer patients receiving adjuvant endocrine therapy. Breast Cancer Res Treat 2008; 107(2):167–180. doi:10.1007/s10549-007-9548-1
- 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(3):321–333. pmid:12117397
- Tsai SA, Stefanick ML, Stafford RS. Trends in menopausal hormone therapy use of US office-based physicians, 2000–2009. Menopause 2011; 18(4):385–392. doi:10.1097/gme.0b013e3181f43404
- North American Menopause Society. Management of symptomatic vulvovaginal atrophy: 2013 position statement of The North American Menopause Society. Menopause 2013; 20(9):888–902. doi:10.1097/GME.0b013e3182a122c2
- Santen RJ. Vaginal administration of estradiol: effects of dose, preparation and timing on plasma estradiol levels. Climacteric 2015; 18(2):121–134. doi:10.3109/13697137.2014.947254
- American College of Obstetricians and Gynecologists Committee on Gynecologic Practice, Farrell R. ACOG Committee Opinion No. 659: the use of vaginal estrogen in women with a history of estrogen-dependent breast cancer. Obstet Gynecol 2016; 127(3):e93–e96. doi:10.1097/AOG.0000000000001351
- Santoro N, Epperson CN, Mathews SB. Menopausal symptoms and their management. Endocrinol Metab Clin North Am 2015; 44(3):497–515. doi:10.1016/j.ecl.2015.05.001
- Labrie F, Archer DF, Martel C, Vaillancourt M, Montesino M. Combined data of intravaginal prasterone against vulvovaginal atrophy of menopause. Menopause 2017; 24(11):1246–1256. doi:10.1097/GME.0000000000000910
- Labrie F, Archer D, Bouchard C, et al. Serum steroid levels during 12-week intravaginal dehydroepiandrosterone administration. Menopause 2009; 16(5):897–906. doi:10.1097/gme.0b013e31819e8930
- Labrie F, Cusan L, Gomez JL, et al. Effect of intravaginal DHEA on serum DHEA and eleven of its metabolites in postmenopausal women. J Steroid Biochem Mol Biol 2008; 111(3-5):178–194. doi:10.1016/j.jsbmb.2008.06.003
- Soe LH, Wurz GT, Kao CJ, Degregorio MW. Ospemifene for the treatment of dyspareunia associated with vulvar and vaginal atrophy: potential benefits in bone and breast. Int J Womens Health 2013; 5:605–611. doi:10.2147/IJWH.S39146
- Taras TL, Wurz GT, DeGregorio MW. In vitro and in vivo biologic effects of ospemifene (FC-1271a) in breast cancer. J Steroid Biochem Mol Biol 2001; 77(4–5):271–279. pmid:11457665
- Wurz GT, Read KC, Marchisano-Karpman C, et al. Ospemifene inhibits the growth of dimethylbenzanthracene-induced mammary tumors in Sencar mice. J Steroid Biochem Mol Biol 2005; 97(3):230–240. doi:10.1016/j.jsbmb.2005.06.027
- Archer DF, Carr BR, Pinkerton JV, Taylor HS, Constantine GD. Effects of ospemifene on the female reproductive and urinary tracts: translation from preclinical models into clinical evidence. Menopause 2015; 22(7):786–796. doi:10.1097/GME.0000000000000365
- Mirkin S, Pickar JH. Management of osteoporosis and menopausal symptoms: focus on bazedoxifene/conjugated estrogen combination. Int J Womens Health 2013; 5:465–475. doi:10.2147/IJWH.S39455
- Kagan R, Goldstein SR, Pickar JH, Komm BS. Patient considerations in the management of menopausal symptoms: role of conjugated estrogens with bazedoxifene. Ther Clin Risk Manag 2016; 12:549–562. doi:10.2147/TCRM.S63833
- Lobo RA, Pinkerton JV, Gass ML, et al. Evaluation of bazedoxifene/conjugated estrogens for the treatment of menopausal symptoms and effects on metabolic parameters and overall safety profile. Fertil Steril 2009; 92(3):1025–1038. doi:10.1016/j.fertnstert.2009.03.113
- Pinkerton JV, Utian WH, Constantine GD, Olivier S, Pickar JH. Relief of vasomotor symptoms with the tissue-selective estrogen complex containing bazedoxifene/conjugated estrogens: a randomized, controlled trial. Menopause 2009; 16(6):1116–1124. doi:10.1097/gme.0b013e3181a7df0d
- Kagan R, Williams RS, Pan K, Mirkin S, Pickar JH. A randomized, placebo- and active-controlled trial of bazedoxifene/conjugated estrogens for treatment of moderate to severe vulvar/vaginal atrophy in postmenopausal women. Menopause 2010; 17(2):281–289. doi:10.1097/GME.0b013e3181b7c65f
- Pinkerton JV, Harvey JA, Pan K, et al. Breast effects of bazedoxifene-conjugated estrogens: a randomized controlled trial. Obstet Gynecol 2013; 121(5):959–968. doi:10.1097/AOG.0b013e31828c5974
- FDA. U.S. Food & Drug Administration. FDA Statement. Statement from FDA Commissioner Scott Gottlieb, M.D., on efforts to safeguard women’s health from deceptive health claims and significant risks related to devices marketed for use in medical procedures for “vaginal rejuvenation.” www.fda.gov/NewsEvents/Newsroom/PressAnnouncements/ucm615130.htm. Accessed August 20, 2018.
- Cruz VL, Steiner ML, Pompei LM, et al. Randomized, double-blind, placebo-controlled clinical trial for evaluating the efficacy of fractional CO2 laser compared with topical estriol in the treatment of vaginal atrophy in postmenopausal women. Menopause 2018; 25(1):21–28. doi:10.1097/GME.0000000000000955
- Biglia N, Bounous VE, D’Alonzo M, et al. Vaginal atrophy in breast cancer survivors: attitude and approaches among oncologists. Clin Breast Cancer 2017; 17(8):611–617. doi:10.1016/j.clbc.2017.05.008
- Lester J, Pahouja G, Andersen B, Lustberg M. Atrophic vaginitis in breast cancer survivors: a difficult survivorship issue. J Pers Med 2015; 5(2):50–66. doi:10.3390/jpm5020050
- Chin SN, Trinkaus M, Simmons C, et al. Prevalence and severity of urogenital symptoms in postmenopausal women receiving endocrine therapy for breast cancer. Clin Breast Cancer 2009; 9(2):108–117. doi:10.3816/CBC.2009.n.020
- Fallowfield L, Cella D, Cuzick J, Francis S, Locker G, Howell A. Quality of life of postmenopausal women in the Arimidex, Tamoxifen, Alone or in Combination (ATAC) adjuvant breast cancer trial. J Clin Oncol 2004; 22(21):4261–4271. doi:10.1200/JCO.2004.08.029
- Cella D, Fallowfield LJ. Recognition and management of treatment-related side effects for breast cancer patients receiving adjuvant endocrine therapy. Breast Cancer Res Treat 2008; 107(2):167–180. doi:10.1007/s10549-007-9548-1
- 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(3):321–333. pmid:12117397
- Tsai SA, Stefanick ML, Stafford RS. Trends in menopausal hormone therapy use of US office-based physicians, 2000–2009. Menopause 2011; 18(4):385–392. doi:10.1097/gme.0b013e3181f43404
- North American Menopause Society. Management of symptomatic vulvovaginal atrophy: 2013 position statement of The North American Menopause Society. Menopause 2013; 20(9):888–902. doi:10.1097/GME.0b013e3182a122c2
- Santen RJ. Vaginal administration of estradiol: effects of dose, preparation and timing on plasma estradiol levels. Climacteric 2015; 18(2):121–134. doi:10.3109/13697137.2014.947254
- American College of Obstetricians and Gynecologists Committee on Gynecologic Practice, Farrell R. ACOG Committee Opinion No. 659: the use of vaginal estrogen in women with a history of estrogen-dependent breast cancer. Obstet Gynecol 2016; 127(3):e93–e96. doi:10.1097/AOG.0000000000001351
- Santoro N, Epperson CN, Mathews SB. Menopausal symptoms and their management. Endocrinol Metab Clin North Am 2015; 44(3):497–515. doi:10.1016/j.ecl.2015.05.001
- Labrie F, Archer DF, Martel C, Vaillancourt M, Montesino M. Combined data of intravaginal prasterone against vulvovaginal atrophy of menopause. Menopause 2017; 24(11):1246–1256. doi:10.1097/GME.0000000000000910
- Labrie F, Archer D, Bouchard C, et al. Serum steroid levels during 12-week intravaginal dehydroepiandrosterone administration. Menopause 2009; 16(5):897–906. doi:10.1097/gme.0b013e31819e8930
- Labrie F, Cusan L, Gomez JL, et al. Effect of intravaginal DHEA on serum DHEA and eleven of its metabolites in postmenopausal women. J Steroid Biochem Mol Biol 2008; 111(3-5):178–194. doi:10.1016/j.jsbmb.2008.06.003
- Soe LH, Wurz GT, Kao CJ, Degregorio MW. Ospemifene for the treatment of dyspareunia associated with vulvar and vaginal atrophy: potential benefits in bone and breast. Int J Womens Health 2013; 5:605–611. doi:10.2147/IJWH.S39146
- Taras TL, Wurz GT, DeGregorio MW. In vitro and in vivo biologic effects of ospemifene (FC-1271a) in breast cancer. J Steroid Biochem Mol Biol 2001; 77(4–5):271–279. pmid:11457665
- Wurz GT, Read KC, Marchisano-Karpman C, et al. Ospemifene inhibits the growth of dimethylbenzanthracene-induced mammary tumors in Sencar mice. J Steroid Biochem Mol Biol 2005; 97(3):230–240. doi:10.1016/j.jsbmb.2005.06.027
- Archer DF, Carr BR, Pinkerton JV, Taylor HS, Constantine GD. Effects of ospemifene on the female reproductive and urinary tracts: translation from preclinical models into clinical evidence. Menopause 2015; 22(7):786–796. doi:10.1097/GME.0000000000000365
- Mirkin S, Pickar JH. Management of osteoporosis and menopausal symptoms: focus on bazedoxifene/conjugated estrogen combination. Int J Womens Health 2013; 5:465–475. doi:10.2147/IJWH.S39455
- Kagan R, Goldstein SR, Pickar JH, Komm BS. Patient considerations in the management of menopausal symptoms: role of conjugated estrogens with bazedoxifene. Ther Clin Risk Manag 2016; 12:549–562. doi:10.2147/TCRM.S63833
- Lobo RA, Pinkerton JV, Gass ML, et al. Evaluation of bazedoxifene/conjugated estrogens for the treatment of menopausal symptoms and effects on metabolic parameters and overall safety profile. Fertil Steril 2009; 92(3):1025–1038. doi:10.1016/j.fertnstert.2009.03.113
- Pinkerton JV, Utian WH, Constantine GD, Olivier S, Pickar JH. Relief of vasomotor symptoms with the tissue-selective estrogen complex containing bazedoxifene/conjugated estrogens: a randomized, controlled trial. Menopause 2009; 16(6):1116–1124. doi:10.1097/gme.0b013e3181a7df0d
- Kagan R, Williams RS, Pan K, Mirkin S, Pickar JH. A randomized, placebo- and active-controlled trial of bazedoxifene/conjugated estrogens for treatment of moderate to severe vulvar/vaginal atrophy in postmenopausal women. Menopause 2010; 17(2):281–289. doi:10.1097/GME.0b013e3181b7c65f
- Pinkerton JV, Harvey JA, Pan K, et al. Breast effects of bazedoxifene-conjugated estrogens: a randomized controlled trial. Obstet Gynecol 2013; 121(5):959–968. doi:10.1097/AOG.0b013e31828c5974
- FDA. U.S. Food & Drug Administration. FDA Statement. Statement from FDA Commissioner Scott Gottlieb, M.D., on efforts to safeguard women’s health from deceptive health claims and significant risks related to devices marketed for use in medical procedures for “vaginal rejuvenation.” www.fda.gov/NewsEvents/Newsroom/PressAnnouncements/ucm615130.htm. Accessed August 20, 2018.
- Cruz VL, Steiner ML, Pompei LM, et al. Randomized, double-blind, placebo-controlled clinical trial for evaluating the efficacy of fractional CO2 laser compared with topical estriol in the treatment of vaginal atrophy in postmenopausal women. Menopause 2018; 25(1):21–28. doi:10.1097/GME.0000000000000955
- Biglia N, Bounous VE, D’Alonzo M, et al. Vaginal atrophy in breast cancer survivors: attitude and approaches among oncologists. Clin Breast Cancer 2017; 17(8):611–617. doi:10.1016/j.clbc.2017.05.008
KEY POINTS
- In general, locally applied hormonal therapies relieve GSM symptoms without increasing breast cancer risk.
- DHEA relieves vaginal symptoms without increasing serum estrogen levels.
- Ospemifene has antiestrogenic effects on breast tissue that make it an attractive option for women with breast cancer.
- The combination of conjugated estrogens and bazedoxifene offers a progesterone-free treatment for GSM symptoms in women desiring systemic hormone therapy.
Bicuspid aortic valve: Basics and beyond
Bicuspid aortic valve may initially be asymptomatic, but it is associated with progressive valvular and aortic abnormalities that can lead to chronic heart failure and sudden death. Regular monitoring is required with an eye toward surgery when indicated.
This article reviews inheritance patterns and conditions associated with bicuspid aortic valve. We discuss diagnosis, management, and monitoring, and offer surgical recommendations. Special guidance for dental procedures, pregnancy, and athletes is also provided.
A YOUNG MAN WITH PALPITATIONS AND A MURMUR
A 34-year-old man presented to an outpatient clinic with occasional palpitations over the past several months. He reported that he had been diagnosed with a murmur as a child but had received no further testing.
Physical examination at this time revealed a faint systolic crescendo-decrescendo murmur along the right sternal border without radiation to the carotid arteries or to the apex. Transthoracic echocardiography (TTE) showed a bicuspid aortic valve with fusion of the right and left coronary cusps, with no aortic valve stenosis or insufficiency. There was mild dilation of the aortic root, but the mid-ascending aorta could not be evaluated because of limited acoustic windows.
Is further diagnostic testing needed, and if so, what? May he participate in exertional physical activity? Does his newborn son need evaluation?
ABNORMALITIES OCCUR DURING EMBRYOGENESIS
Bicuspid aortic valve develops because of abnormal valvulogenesis. Adjacent cusps fail to separate from each other, resulting in only 2 cusps, with 1 usually larger than the other. Morphology varies according to which commissures are fused.1
Bicuspid aortic valve is associated with abnormalities in the coronary artery anatomy in about 2% of patients, including anomalous origins of the coronary arteries and upwardly displaced coronary ostia.2 Such features need to be considered before surgical intervention.
Bicuspid aortic valve can be found in 1% to 2% of the general population, with a male-to-female predominance of 3:1.1,3,4 It is one of the most common congenital cardiac malformations and is the leading congenital cause of aortic valve stenosis.1,3 However, routine screening of newborns for the condition is not recommended, and most cases are diagnosed incidentally.
GENETIC FACTORS PROMINENT
Bicuspid aortic valve is thought to be primarily inherited in an autosomal-dominant pattern, but there is evidence of genetic heterogeneity, and the pattern may be variable.5,6
No single gene responsible for bicuspid aortic valve has been identified. The condition may occur as a component of different pleiotropic genetic syndromes such as Loeys-Dietz, DiGeorge, and Marfan syndromes,7,8 as well as in patients with Turner syndrome and Williams syndrome.8–11 It also commonly coexists with other congenital heart diseases, including ventricular septal defect, isolated aortic arch obstruction, and patent ductus arteriosus.9
Studies have found a 15% rate of familial clustering.6,12 In a study of 142 patients with bicuspid aortic valve, 20% of first-degree relatives had some cardiac abnormality found by screening, of whom 68% had bicuspid aortic valve. Of these, 71% were newly detected abnormalities.13
CHARACTERISTIC CLICK AND MURMUR
Physical examination findings of a functionally normal bicuspid aortic valve include a systolic ejection click followed by an early peaking systolic murmur at the apex or left lower sternal border. With progression of aortic stenosis, the ejection murmur has a harsher sound, with later peaking, and the S2 sound diminishes or becomes inaudible.14 If aortic regurgitation is present, a diastolic decrescendo murmur is heard best at the left lower sternal border.
DISEASE PROGRESSION
Although bicuspid aortic valve is typically asymptomatic at first, it is commonly associated with progressive valvulopathy and thoracic aortic disease.1,3,4,15 It can lead to chronic heart failure and increase the risk of acute aortic syndromes and sudden cardiac death.15
Michelena et al16 studied 212 cases of asymptomatic bicuspid aortic valve. Although the survival rate 20 years after diagnosis was the same as for an age-matched cohort in the general population, the frequency of adverse cardiovascular events and surgical interventions was higher.
Aortic stenosis progresses rapidly
Aortic stenosis associated with a bicuspid aortic valve tends to affect younger patients and progress more rapidly than when associated with a tricuspid valve.17
In a study of 542 patients with congenital bicuspid aortic valve undergoing aortic valve replacement,3 75% had isolated aortic stenosis, 10% had aortic stenosis with some degree of aortic insufficiency, and 13% had isolated aortic insufficiency. Given the tendency of aortic stenosis to progress rapidly, early surgery is often pursued.17,18
Aneurysmal disease is common
The thoracic aorta is at increased risk of aneurysmal disease, coarctation, and dissection in patients with a bicuspid aortic valve.1,6,15
Michelena et al16 reported that in patients without an aneurysm at the time of bicuspid aortic valve diagnosis, the 25-year risk of aneurysm formation was approximately 26%. In patients with an aneurysm at the time of diagnosis, the 15-year risk of aortic surgery after the diagnosis of aneurysm was about 46% and the risk of aortic dissection after aneurysm diagnosis was 7%.15 Compared with the general population, the age-adjusted relative risk of aortic aneurysm in patients with bicuspid aortic valve was 86.2, and that of aortic dissection was 8.4. Although the absolute incidence of dissection is low in these patients, it is markedly higher than in the general population, particularly in older patients (age > 50) and those with an aneurysm at the time of diagnosis.15
The risk of infective endocarditis
Patients with bicuspid aortic valve are highly prone to infective endocarditis for reasons that remain poorly understood. The pathogens in most cases are staphylococci or viridans streptococci.19 Patients with infective endocarditis typically require emergency surgery. Complications including valvular abscess, myocardial abscess, and overt heart failure are common.19
Lamas and Eykyn20 studied 408 cases of native valve endocarditis; in 12.3%, the patient had a bicuspid aortic valve. In this subset, all were male, the mean age was 39 at diagnosis, 82% needed surgery, and the death rate was 14%.
Patients with bicuspid aortic valve do not routinely need antibiotics before dental and surgical procedures, but if they have had endocarditis in the past, they need antibiotics to prevent a recurrence.21
REGULAR MONITORING NEEDED
Because complications may be life-threatening, early detection of progressive disease by regular screening is critical. Echocardiographic evaluation of valvular function, ventricular dimensions and function, and diameter of the aortic root and ascending aorta should be performed in every patient with bicuspid aortic valve. If initial imaging is normal and there is no aortic dilation, imaging should be repeated every 5 to 10 years. If any abnormality is found, repeat imaging is needed every year.22
Magnetic resonance imaging (MRI) or computed tomographic (CT) angiography may be required to better assess the aorta for patients requiring a surgical intervention, or when aortic dimensions are not clearly visualized on TTE. MRI has 2 advantages over CT angiography: it poses no radiation risk, and it provides more information on left ventricular function and dimensions, in addition to valve assessment.23,24
No published study has compared MRI or CT angiography and transesophageal echocardiography (TEE), but in a study of 174 patients with dilated aortic root, combined TTE and TEE detected aortic valve morphology accurately in 98% of cases. As TEE is more invasive, it is not recommended for regular surveillance (Figures 1 and 2).25
FAMILY SCREENING RECOMMENDED
Close relatives should be evaluated for aortic valve and thoracic aortic disease.12,13,23,26
The American College of Cardiology (ACC) and the American Heart Association (AHA), backed by radiologic and cardiovascular associations, concur in recommending echocardiographic screening and routine screening of the thoracic aorta for aortic root dilation in first-degree relatives (ie, siblings, parents, and children) of patients with bicuspid aortic valve (class I recommendation).22,27,28
A comprehensive physical examination is recommended for family members in addition to TTE, with careful assessment of the aortic valve in short and long axes, and of the aortic root.14 If the aorta cannot be adequately evaluated with TTE, further assessment should be pursued with CT angiography or MRI.
EXERCISE RESTRICTIONS
The 2015 ACC/AHA guidelines for competitive athletes with cardiovascular abnormalities recommend annual screening with TTE or MRI angiography for athletes with bicuspid aortic valve and coexisting dilation of the ascending aorta (aortic diameter 40–42 mm in men and 36–39 mm in women) (class I recommendation, level of evidence C).29
Athletes with a bicuspid aortic valve and a normal aortic root and ascending aorta may participate in all competitive activities.29 However, those with a dilated aorta should avoid strenuous activities because of the increased risk of rupture.30 The ACC/AHA recommendations29 depend on the diameter of the ascending aorta and the nature of the sport:
- For an aortic diameter 40 to 42 mm in men or 36 to 39 mm in women, and no features of connective tissue disease or familial thoracic ascending aortic syndrome, low- and moderate-intensity sports with a low likelihood of significant body contact may be considered; consider avoiding intense weight training (class IIb, level of evidence C)
- For an aortic diameter 43 to 45 mm, low-intensity sports with a low likelihood of body contact may be considered (class IIb, level of evidence C)
- For an aortic diameter greater than 43 mm in men or greater than 40 mm in women, sports involving body collision should be avoided (class III, level of evidence C)
- For an aortic diameter greater than 45 mm, sports activities should be avoided (class III, level of evidence C).
PREGNANCY CONSIDERATIONS
Bicuspid aortic valve is associated with aortic dissection, mainly in the third trimester.31 Patients should ideally undergo echocardiographic screening before conception. The 2010 ACC/AHA guidelines for managing thoracic aortic disease recommend monthly or bimonthly echocardiography until delivery in pregnant women with a dilated thoracic aorta.22
Patients with bicuspid aortic valve and aortic root enlargement of more than 40 mm should have preconception counseling about surgery for aortic root replacement before becoming pregnant. If the diagnosis of enlarged aortic root is made during pregnancy, echocardiographic surveillance at 4- to 6-week intervals is indicated.32
SURGICAL MANAGEMENT
In the past, beta-blockers and angiotensin-converting enzyme inhibitors were recommended to minimize shear stress, with the goal of slowing progression of aortic dilation. However, evidence to support their use is inadequate.33,34
The only definitive treatment is surgery, with various procedures that lower the risk of death or dissection.24,35
The dimensions of the aortic root or ascending aorta should be examined vigilantly, according to the 2014 ACC/AHA guidelines27:
- Repairing the aortic sinuses or replacing the ascending aorta is indicated if the diameter of the aortic sinuses or ascending aorta is greater than 5.5 cm (class I, level of evidence B)
- Repairing the aortic sinuses or replacing the ascending aorta is reasonable if the diameter of the aortic sinuses or ascending aorta is greater than 5.0 cm and the patient has a risk factor for dissection such as a family history of aortic dissection or an increase in diameter of 0.5 cm or greater per year (class IIa, level of evidence C)
- Replacement of the ascending aorta is reasonable if the diameter of the ascending aorta is greater than 4.5 cm and the patient is undergoing aortic valve surgery for severe aortic stenosis or regurgitation.
Valve repair or replacement
Aortic valve repair or replacement is sometimes done separately from aortic root repair.
The value of aortic valve repair is debatable, but a series of 728 patients at Cleveland Clinic showed a very low mortality rate (0.41%) and an annual reoperation rate of 2.6% during up to 15 years of follow-up.36
Aortic valve replacement is usually considered for patients with severe valve dysfunction, abnormal left ventricular dimensions, or symptoms. It is important to determine if the patient is a good surgical candidate and to refer early for surgical evaluation to avoid the higher risk of death associated with emergency surgery.36
Transcatheter aortic valve replacement has been studied in patients deemed to be at too high a risk for surgical replacement. Short- and intermediate-term outcomes have been good in these patients, but long-term data are lacking.37
Surveillance after surgery
The type of operation determines postoperative surveillance.
After isolated aortic valve repair or replacement, patients should continue with surveillance at least annually to monitor for progressive aortopathy, as they remain at increased risk of dissection or rupture after isolated valve surgery, especially if they had aortic insufficiency preoperatively.38
After definitive surgery with replacement or repair of the ascending aorta, no clear recommendations have been established for continued surveillance. However, it is reasonable to image these patients with either MRI or CT angiography 3 to 5 years after their surgery to monitor for anastomotic complications.
CASE QUESTIONS ANSWERED
Our patient should undergo repeat TTE in 1 year. He should also undergo CT angiography of the ascending aorta if it is not seen by TTE. He can participate in low-intensity sports but should avoid intense weight training. His parents, siblings, and children should be screened for bicuspid aortic valve or associated aortopathies.
- Roberts WC. The congenitally bicuspid aortic valve. A study of 85 autopsy cases. Am J Cardiol 1970; 26(1):72–83. pmid:5427836
- Michalowska IM, Hryniewiecki T, Kwiatek P, Stoklosa P, Swoboda-Rydz U, Szymanski P. Coronary artery variants and anomalies in patients with bicuspid aortic valve. J Thorac Imaging 2016; 31(3):156–162. doi:10.1097/RTI.0000000000000205
- Sabet HY, Edwards WD, Tazelaar HD, Daly RC. Congenitally bicuspid aortic valves: a surgical pathology study of 542 cases (1991 through 1996) and a literature review of 2,715 additional cases. Mayo Clin Proc 1999; 74(1):14–26. doi:10.4065/74.1.14
- Tutar E, Ekici F, Atalay S, Nacar N. The prevalence of bicuspid aortic valve in newborns by echocardiographic screening. Am Heart J 2005; 150(3):513–515. doi:10.1016/j.ahj.2004.10.036
- Benson DW. The genetics of congenital heart disease: a point in the revolution. Cardiol Clin 2002; 20(3):385–394. pmid:12371007
- Emanuel R, Withers R, O’Brien K, Ross P, Feizi O. Congenitally bicuspid aortic valves. Clinicogenetic study of 41 families. Br Heart J 1978; 40(12):1402–1407. pmid:737099
- Giusti B, Sticchi E, De Cario R, Magi A, Nistri S, Pepe G. Genetic bases of bicuspid aortic valve: the contribution of traditional and high-throughput sequencing approaches on research and diagnosis. Front Physiol 2017; 8:612. doi:10.3389/fphys.2017.00612
- Sachdev V, Matura LA, Sidenko S, et al. Aortic valve disease in Turner syndrome. J Am Coll Cardiol 2008; 51(19):1904–1909. doi:10.1016/j.jacc.2008.02.035
- Duran AC, Frescura C, Sans-Coma V, Angelini A, Basso C, Thiene G. Bicuspid aortic valves in hearts with other congenital heart disease. J Heart Valve Dis 1995; 4(6):581–590. pmid:8611973
- De Rubens Figueroa J, Rodríguez LM, Hach JL, Del Castillo Ruíz V, Martínez HO. Cardiovascular spectrum in Williams-Beuren syndrome: the Mexican experience in 40 patients. Tex Heart Inst J 2008; 35(3):279–285. pmid:18941598
- Yuan SM, Jing H. The bicuspid aortic valve and related disorders. Sao Paulo Med J 2010; 128(5):296–301. pmid:21181071
- Cripe L, Andelfinger G, Martin LJ, Shooner K, Benson DW. Bicuspid aortic valve is heritable. J Am Coll Cardiol 2004; 44(1):138–143. doi:10.1016/j.jacc.2004.03.050
- Kerstjens-Frederikse WS, Sarvaas GJ, Ruiter JS, et al. Left ventricular outflow tract obstruction: should cardiac screening be offered to first-degree relatives? Heart 2011; 97(15):1228–1232. doi:10.1136/hrt.2010.211433
- Siu SC, Silversides CK. Bicuspid aortic valve disease. J Am Coll Cardiol 2010; 55(25):2789–2800. doi:10.1016/j.jacc.2009.12.068
- Michelena HI, Khanna AD, Mahoney D, et al. Incidence of aortic complications in patients with bicuspid aortic valves. JAMA 2011; 306(10):1104–1112.
- Michelena HI, Desjardins VA, Avierinos JF, et al. Natural history of asymptomatic patients with normally functioning or minimally dysfunctional bicuspid aortic valve in the community. Circulation 2008; 117(21):2776–2784. doi:10.1161/CIRCULATIONAHA.107.740878
- Beppu S, Suzuki S, Matsuda H, Ohmori F, Nagata S, Miyatake K. Rapidity of progression of aortic stenosis in patients with congenital bicuspid aortic valves. Am J Cardiol 1993; 71(4):322–327. pmid:8427176
- Roberts WC, Ko JM. Frequency by decades of unicuspid, bicuspid, and tricuspid aortic valves in adults having isolated aortic valve replacement for aortic stenosis, with or without associated aortic regurgitation. Circulation 2005; 111(7):920–925. doi:10.1161/01.CIR.0000155623.48408.C5
- Yener N, Oktar GL, Erer D, Yardimci MM, Yener A. Bicuspid aortic valve. Ann Thorac Cardiovasc Surg 2002; 8(5):264–267. pmid:12472407
- Lamas CC, Eykyn SJ. Bicuspid aortic valve—a silent danger: analysis of 50 cases of infective endocarditis. Clin Infect Dis 2000; 30(2):336–341. doi:10.1086/313646
- Wilson W, Taubert KA, Gewitz M, et al. Prevention of infective endocarditis: guidelines from the American Heart Association. Circulation 2007; 116(15):1736–1754. doi:10.1161/CIRCULATIONAHA.106.183095
- Hiratzka L, Bakris G, Beckman JA, et al. 2010 ACCF/AHA/AATS/ACR/ASA/SCA/SCAI/SIR/STS/SVM guidelines for the diagnosis and management of patients with thoracic aortic disease. Circulation 2010; 121(13):e266–e369. doi:10.1161/CIR.0b013e3181d4739e
- Chun EJ, Choi SI, Lim C, et al. Aortic stenosis: evaluation with multidetector CT angiography and MR imaging. Korean J Radiol 2008; 9(5):439–448. doi:10.3348/kjr.2008.9.5.439
- Kiefer TL, Wang A, Hughes GC, Bashore TM. Management of patients with bicuspid aortic valve disease. Curr Treat Options Cardiovasc Med 2011; 13(6):489–505. doi:10.1007/s11936-011-0152-7
- Alegret JM, Palazon O, Duran I, Vernis JM. Aortic valve morphology definition with transthoracic combined with transesophageal echocardiography in a population with high prevalence of bicuspid aortic valve. Int J Cardiovasc Imaging 2005; 21(2-3):213–217. doi:10.1007/s10554-004-3901-9
- Biner S, Rafique AM, Ray I, Cuk O, Siegel RJ, Tolstrup K. Aortopathy is prevalent in relatives of bicuspid aortic valve patients. J Am Coll Cardiol 2009; 53(24):2288–2295. doi:10.1016/j.jacc.2009.03.027
- Nishimura RA, Otto CM, Bonow RO, et al. 2014 AHA/ACC guideline for the management of patients with valvular heart disease. J Thorac Cardiovasc Surg 2014; 148(1):e1-e132. doi:10.1016/j.jtcvs.2014.05.014
- Warnes CA, Williams RG, Bashore TM, et al. ACC/AHA 2008 guidelines for the management of adults with congenital heart disease. J Am Coll Cardiol 2008; 52(23):e143–e263. doi:10.1016/j.jacc.2008.10.001
- Braverman AC, Harris KM, Kovacs RJ, Maron BJ. Eligibility and disqualification recommendations for competitive athletes with cardiovascular abnormalities: Task Force 7: aortic diseases, including Marfan syndrome. Circulation 2015; 132(22):e303–e309. doi:10.1161/CIR.0000000000000243
- De Mozzi P, Longo UG, Galanti G, Maffulli N. Bicuspid aortic valve: a literature review and its impact on sport activity. Br Med Bull 2008; 85:63–85. doi:10.1093/bmb/ldn002
- Thorne SA. Pregnancy in heart disease. Heart 2004; 90(4):450–456. pmid:15020530
- Immer FF, Bansi AG, Immer-Bansi AS, et al. Aortic dissection in pregnancy: analysis of risk factors and outcome. Ann Thorac Surg 2003; 76(1):309–314. pmid:12842575
- Allen BD, Markl M, Barker AJ, et al. Influence of beta-blocker therapy on aortic blood flow in patients with bicuspid aortic valve. Int J Cardiovasc Imaging 2016; 32(4):621–628. doi:10.1007/s10554-015-0819-3
- Ohnemus D, Oster ME, Gatlin S, Jokhadar M, Mahle WT. The effect of angiotensin-converting enzyme inhibitors on the rate of ascending aorta dilation in patients with bicuspid aortic valve. Congenit Heart Dis 2015; 10(1):E1–E5. doi:10.1111/chd.12184
- Masri A, Kalahasti V, Alkharabsheh S, et al. Characteristics and long-term outcomes of contemporary patients with bicuspid aortic valves. J Thorac Cardiovasc Surg 2016; 151(6):1650–1659.e1. doi:10.1016/j.jtcvs.2015.12.019
- Svensson LG, Al Kindi AH, Vivacqua A, et al. Long-term durability of bicuspid aortic valve repair. Ann Thorac Surg 2014; 97(5):1539–1548. doi:10.1016/j.athoracsur.2013.11.036
- Mylotte D, Lefevre T, Sondergaard L, et al. Transcatheter aortic valve replacement in bicuspid aortic valve disease. J Am Coll Cardiol 2014; 64(22):2330–2339. doi:10.1016/j.jacc.2014.09.039
- Girdauskas E, Disha K, Raisin HH, Secknus MA, Borger MA, Kuntze T. Risk of late aortic events after an isolated aortic valve replacement for bicuspid aortic valve stenosis with concomitant ascending aortic dilation. Eur J Cardiothorac Surg 2012; 42(5):832–838. doi:10.1093/ejcts/ezs137
Bicuspid aortic valve may initially be asymptomatic, but it is associated with progressive valvular and aortic abnormalities that can lead to chronic heart failure and sudden death. Regular monitoring is required with an eye toward surgery when indicated.
This article reviews inheritance patterns and conditions associated with bicuspid aortic valve. We discuss diagnosis, management, and monitoring, and offer surgical recommendations. Special guidance for dental procedures, pregnancy, and athletes is also provided.
A YOUNG MAN WITH PALPITATIONS AND A MURMUR
A 34-year-old man presented to an outpatient clinic with occasional palpitations over the past several months. He reported that he had been diagnosed with a murmur as a child but had received no further testing.
Physical examination at this time revealed a faint systolic crescendo-decrescendo murmur along the right sternal border without radiation to the carotid arteries or to the apex. Transthoracic echocardiography (TTE) showed a bicuspid aortic valve with fusion of the right and left coronary cusps, with no aortic valve stenosis or insufficiency. There was mild dilation of the aortic root, but the mid-ascending aorta could not be evaluated because of limited acoustic windows.
Is further diagnostic testing needed, and if so, what? May he participate in exertional physical activity? Does his newborn son need evaluation?
ABNORMALITIES OCCUR DURING EMBRYOGENESIS
Bicuspid aortic valve develops because of abnormal valvulogenesis. Adjacent cusps fail to separate from each other, resulting in only 2 cusps, with 1 usually larger than the other. Morphology varies according to which commissures are fused.1
Bicuspid aortic valve is associated with abnormalities in the coronary artery anatomy in about 2% of patients, including anomalous origins of the coronary arteries and upwardly displaced coronary ostia.2 Such features need to be considered before surgical intervention.
Bicuspid aortic valve can be found in 1% to 2% of the general population, with a male-to-female predominance of 3:1.1,3,4 It is one of the most common congenital cardiac malformations and is the leading congenital cause of aortic valve stenosis.1,3 However, routine screening of newborns for the condition is not recommended, and most cases are diagnosed incidentally.
GENETIC FACTORS PROMINENT
Bicuspid aortic valve is thought to be primarily inherited in an autosomal-dominant pattern, but there is evidence of genetic heterogeneity, and the pattern may be variable.5,6
No single gene responsible for bicuspid aortic valve has been identified. The condition may occur as a component of different pleiotropic genetic syndromes such as Loeys-Dietz, DiGeorge, and Marfan syndromes,7,8 as well as in patients with Turner syndrome and Williams syndrome.8–11 It also commonly coexists with other congenital heart diseases, including ventricular septal defect, isolated aortic arch obstruction, and patent ductus arteriosus.9
Studies have found a 15% rate of familial clustering.6,12 In a study of 142 patients with bicuspid aortic valve, 20% of first-degree relatives had some cardiac abnormality found by screening, of whom 68% had bicuspid aortic valve. Of these, 71% were newly detected abnormalities.13
CHARACTERISTIC CLICK AND MURMUR
Physical examination findings of a functionally normal bicuspid aortic valve include a systolic ejection click followed by an early peaking systolic murmur at the apex or left lower sternal border. With progression of aortic stenosis, the ejection murmur has a harsher sound, with later peaking, and the S2 sound diminishes or becomes inaudible.14 If aortic regurgitation is present, a diastolic decrescendo murmur is heard best at the left lower sternal border.
DISEASE PROGRESSION
Although bicuspid aortic valve is typically asymptomatic at first, it is commonly associated with progressive valvulopathy and thoracic aortic disease.1,3,4,15 It can lead to chronic heart failure and increase the risk of acute aortic syndromes and sudden cardiac death.15
Michelena et al16 studied 212 cases of asymptomatic bicuspid aortic valve. Although the survival rate 20 years after diagnosis was the same as for an age-matched cohort in the general population, the frequency of adverse cardiovascular events and surgical interventions was higher.
Aortic stenosis progresses rapidly
Aortic stenosis associated with a bicuspid aortic valve tends to affect younger patients and progress more rapidly than when associated with a tricuspid valve.17
In a study of 542 patients with congenital bicuspid aortic valve undergoing aortic valve replacement,3 75% had isolated aortic stenosis, 10% had aortic stenosis with some degree of aortic insufficiency, and 13% had isolated aortic insufficiency. Given the tendency of aortic stenosis to progress rapidly, early surgery is often pursued.17,18
Aneurysmal disease is common
The thoracic aorta is at increased risk of aneurysmal disease, coarctation, and dissection in patients with a bicuspid aortic valve.1,6,15
Michelena et al16 reported that in patients without an aneurysm at the time of bicuspid aortic valve diagnosis, the 25-year risk of aneurysm formation was approximately 26%. In patients with an aneurysm at the time of diagnosis, the 15-year risk of aortic surgery after the diagnosis of aneurysm was about 46% and the risk of aortic dissection after aneurysm diagnosis was 7%.15 Compared with the general population, the age-adjusted relative risk of aortic aneurysm in patients with bicuspid aortic valve was 86.2, and that of aortic dissection was 8.4. Although the absolute incidence of dissection is low in these patients, it is markedly higher than in the general population, particularly in older patients (age > 50) and those with an aneurysm at the time of diagnosis.15
The risk of infective endocarditis
Patients with bicuspid aortic valve are highly prone to infective endocarditis for reasons that remain poorly understood. The pathogens in most cases are staphylococci or viridans streptococci.19 Patients with infective endocarditis typically require emergency surgery. Complications including valvular abscess, myocardial abscess, and overt heart failure are common.19
Lamas and Eykyn20 studied 408 cases of native valve endocarditis; in 12.3%, the patient had a bicuspid aortic valve. In this subset, all were male, the mean age was 39 at diagnosis, 82% needed surgery, and the death rate was 14%.
Patients with bicuspid aortic valve do not routinely need antibiotics before dental and surgical procedures, but if they have had endocarditis in the past, they need antibiotics to prevent a recurrence.21
REGULAR MONITORING NEEDED
Because complications may be life-threatening, early detection of progressive disease by regular screening is critical. Echocardiographic evaluation of valvular function, ventricular dimensions and function, and diameter of the aortic root and ascending aorta should be performed in every patient with bicuspid aortic valve. If initial imaging is normal and there is no aortic dilation, imaging should be repeated every 5 to 10 years. If any abnormality is found, repeat imaging is needed every year.22
Magnetic resonance imaging (MRI) or computed tomographic (CT) angiography may be required to better assess the aorta for patients requiring a surgical intervention, or when aortic dimensions are not clearly visualized on TTE. MRI has 2 advantages over CT angiography: it poses no radiation risk, and it provides more information on left ventricular function and dimensions, in addition to valve assessment.23,24
No published study has compared MRI or CT angiography and transesophageal echocardiography (TEE), but in a study of 174 patients with dilated aortic root, combined TTE and TEE detected aortic valve morphology accurately in 98% of cases. As TEE is more invasive, it is not recommended for regular surveillance (Figures 1 and 2).25
FAMILY SCREENING RECOMMENDED
Close relatives should be evaluated for aortic valve and thoracic aortic disease.12,13,23,26
The American College of Cardiology (ACC) and the American Heart Association (AHA), backed by radiologic and cardiovascular associations, concur in recommending echocardiographic screening and routine screening of the thoracic aorta for aortic root dilation in first-degree relatives (ie, siblings, parents, and children) of patients with bicuspid aortic valve (class I recommendation).22,27,28
A comprehensive physical examination is recommended for family members in addition to TTE, with careful assessment of the aortic valve in short and long axes, and of the aortic root.14 If the aorta cannot be adequately evaluated with TTE, further assessment should be pursued with CT angiography or MRI.
EXERCISE RESTRICTIONS
The 2015 ACC/AHA guidelines for competitive athletes with cardiovascular abnormalities recommend annual screening with TTE or MRI angiography for athletes with bicuspid aortic valve and coexisting dilation of the ascending aorta (aortic diameter 40–42 mm in men and 36–39 mm in women) (class I recommendation, level of evidence C).29
Athletes with a bicuspid aortic valve and a normal aortic root and ascending aorta may participate in all competitive activities.29 However, those with a dilated aorta should avoid strenuous activities because of the increased risk of rupture.30 The ACC/AHA recommendations29 depend on the diameter of the ascending aorta and the nature of the sport:
- For an aortic diameter 40 to 42 mm in men or 36 to 39 mm in women, and no features of connective tissue disease or familial thoracic ascending aortic syndrome, low- and moderate-intensity sports with a low likelihood of significant body contact may be considered; consider avoiding intense weight training (class IIb, level of evidence C)
- For an aortic diameter 43 to 45 mm, low-intensity sports with a low likelihood of body contact may be considered (class IIb, level of evidence C)
- For an aortic diameter greater than 43 mm in men or greater than 40 mm in women, sports involving body collision should be avoided (class III, level of evidence C)
- For an aortic diameter greater than 45 mm, sports activities should be avoided (class III, level of evidence C).
PREGNANCY CONSIDERATIONS
Bicuspid aortic valve is associated with aortic dissection, mainly in the third trimester.31 Patients should ideally undergo echocardiographic screening before conception. The 2010 ACC/AHA guidelines for managing thoracic aortic disease recommend monthly or bimonthly echocardiography until delivery in pregnant women with a dilated thoracic aorta.22
Patients with bicuspid aortic valve and aortic root enlargement of more than 40 mm should have preconception counseling about surgery for aortic root replacement before becoming pregnant. If the diagnosis of enlarged aortic root is made during pregnancy, echocardiographic surveillance at 4- to 6-week intervals is indicated.32
SURGICAL MANAGEMENT
In the past, beta-blockers and angiotensin-converting enzyme inhibitors were recommended to minimize shear stress, with the goal of slowing progression of aortic dilation. However, evidence to support their use is inadequate.33,34
The only definitive treatment is surgery, with various procedures that lower the risk of death or dissection.24,35
The dimensions of the aortic root or ascending aorta should be examined vigilantly, according to the 2014 ACC/AHA guidelines27:
- Repairing the aortic sinuses or replacing the ascending aorta is indicated if the diameter of the aortic sinuses or ascending aorta is greater than 5.5 cm (class I, level of evidence B)
- Repairing the aortic sinuses or replacing the ascending aorta is reasonable if the diameter of the aortic sinuses or ascending aorta is greater than 5.0 cm and the patient has a risk factor for dissection such as a family history of aortic dissection or an increase in diameter of 0.5 cm or greater per year (class IIa, level of evidence C)
- Replacement of the ascending aorta is reasonable if the diameter of the ascending aorta is greater than 4.5 cm and the patient is undergoing aortic valve surgery for severe aortic stenosis or regurgitation.
Valve repair or replacement
Aortic valve repair or replacement is sometimes done separately from aortic root repair.
The value of aortic valve repair is debatable, but a series of 728 patients at Cleveland Clinic showed a very low mortality rate (0.41%) and an annual reoperation rate of 2.6% during up to 15 years of follow-up.36
Aortic valve replacement is usually considered for patients with severe valve dysfunction, abnormal left ventricular dimensions, or symptoms. It is important to determine if the patient is a good surgical candidate and to refer early for surgical evaluation to avoid the higher risk of death associated with emergency surgery.36
Transcatheter aortic valve replacement has been studied in patients deemed to be at too high a risk for surgical replacement. Short- and intermediate-term outcomes have been good in these patients, but long-term data are lacking.37
Surveillance after surgery
The type of operation determines postoperative surveillance.
After isolated aortic valve repair or replacement, patients should continue with surveillance at least annually to monitor for progressive aortopathy, as they remain at increased risk of dissection or rupture after isolated valve surgery, especially if they had aortic insufficiency preoperatively.38
After definitive surgery with replacement or repair of the ascending aorta, no clear recommendations have been established for continued surveillance. However, it is reasonable to image these patients with either MRI or CT angiography 3 to 5 years after their surgery to monitor for anastomotic complications.
CASE QUESTIONS ANSWERED
Our patient should undergo repeat TTE in 1 year. He should also undergo CT angiography of the ascending aorta if it is not seen by TTE. He can participate in low-intensity sports but should avoid intense weight training. His parents, siblings, and children should be screened for bicuspid aortic valve or associated aortopathies.
Bicuspid aortic valve may initially be asymptomatic, but it is associated with progressive valvular and aortic abnormalities that can lead to chronic heart failure and sudden death. Regular monitoring is required with an eye toward surgery when indicated.
This article reviews inheritance patterns and conditions associated with bicuspid aortic valve. We discuss diagnosis, management, and monitoring, and offer surgical recommendations. Special guidance for dental procedures, pregnancy, and athletes is also provided.
A YOUNG MAN WITH PALPITATIONS AND A MURMUR
A 34-year-old man presented to an outpatient clinic with occasional palpitations over the past several months. He reported that he had been diagnosed with a murmur as a child but had received no further testing.
Physical examination at this time revealed a faint systolic crescendo-decrescendo murmur along the right sternal border without radiation to the carotid arteries or to the apex. Transthoracic echocardiography (TTE) showed a bicuspid aortic valve with fusion of the right and left coronary cusps, with no aortic valve stenosis or insufficiency. There was mild dilation of the aortic root, but the mid-ascending aorta could not be evaluated because of limited acoustic windows.
Is further diagnostic testing needed, and if so, what? May he participate in exertional physical activity? Does his newborn son need evaluation?
ABNORMALITIES OCCUR DURING EMBRYOGENESIS
Bicuspid aortic valve develops because of abnormal valvulogenesis. Adjacent cusps fail to separate from each other, resulting in only 2 cusps, with 1 usually larger than the other. Morphology varies according to which commissures are fused.1
Bicuspid aortic valve is associated with abnormalities in the coronary artery anatomy in about 2% of patients, including anomalous origins of the coronary arteries and upwardly displaced coronary ostia.2 Such features need to be considered before surgical intervention.
Bicuspid aortic valve can be found in 1% to 2% of the general population, with a male-to-female predominance of 3:1.1,3,4 It is one of the most common congenital cardiac malformations and is the leading congenital cause of aortic valve stenosis.1,3 However, routine screening of newborns for the condition is not recommended, and most cases are diagnosed incidentally.
GENETIC FACTORS PROMINENT
Bicuspid aortic valve is thought to be primarily inherited in an autosomal-dominant pattern, but there is evidence of genetic heterogeneity, and the pattern may be variable.5,6
No single gene responsible for bicuspid aortic valve has been identified. The condition may occur as a component of different pleiotropic genetic syndromes such as Loeys-Dietz, DiGeorge, and Marfan syndromes,7,8 as well as in patients with Turner syndrome and Williams syndrome.8–11 It also commonly coexists with other congenital heart diseases, including ventricular septal defect, isolated aortic arch obstruction, and patent ductus arteriosus.9
Studies have found a 15% rate of familial clustering.6,12 In a study of 142 patients with bicuspid aortic valve, 20% of first-degree relatives had some cardiac abnormality found by screening, of whom 68% had bicuspid aortic valve. Of these, 71% were newly detected abnormalities.13
CHARACTERISTIC CLICK AND MURMUR
Physical examination findings of a functionally normal bicuspid aortic valve include a systolic ejection click followed by an early peaking systolic murmur at the apex or left lower sternal border. With progression of aortic stenosis, the ejection murmur has a harsher sound, with later peaking, and the S2 sound diminishes or becomes inaudible.14 If aortic regurgitation is present, a diastolic decrescendo murmur is heard best at the left lower sternal border.
DISEASE PROGRESSION
Although bicuspid aortic valve is typically asymptomatic at first, it is commonly associated with progressive valvulopathy and thoracic aortic disease.1,3,4,15 It can lead to chronic heart failure and increase the risk of acute aortic syndromes and sudden cardiac death.15
Michelena et al16 studied 212 cases of asymptomatic bicuspid aortic valve. Although the survival rate 20 years after diagnosis was the same as for an age-matched cohort in the general population, the frequency of adverse cardiovascular events and surgical interventions was higher.
Aortic stenosis progresses rapidly
Aortic stenosis associated with a bicuspid aortic valve tends to affect younger patients and progress more rapidly than when associated with a tricuspid valve.17
In a study of 542 patients with congenital bicuspid aortic valve undergoing aortic valve replacement,3 75% had isolated aortic stenosis, 10% had aortic stenosis with some degree of aortic insufficiency, and 13% had isolated aortic insufficiency. Given the tendency of aortic stenosis to progress rapidly, early surgery is often pursued.17,18
Aneurysmal disease is common
The thoracic aorta is at increased risk of aneurysmal disease, coarctation, and dissection in patients with a bicuspid aortic valve.1,6,15
Michelena et al16 reported that in patients without an aneurysm at the time of bicuspid aortic valve diagnosis, the 25-year risk of aneurysm formation was approximately 26%. In patients with an aneurysm at the time of diagnosis, the 15-year risk of aortic surgery after the diagnosis of aneurysm was about 46% and the risk of aortic dissection after aneurysm diagnosis was 7%.15 Compared with the general population, the age-adjusted relative risk of aortic aneurysm in patients with bicuspid aortic valve was 86.2, and that of aortic dissection was 8.4. Although the absolute incidence of dissection is low in these patients, it is markedly higher than in the general population, particularly in older patients (age > 50) and those with an aneurysm at the time of diagnosis.15
The risk of infective endocarditis
Patients with bicuspid aortic valve are highly prone to infective endocarditis for reasons that remain poorly understood. The pathogens in most cases are staphylococci or viridans streptococci.19 Patients with infective endocarditis typically require emergency surgery. Complications including valvular abscess, myocardial abscess, and overt heart failure are common.19
Lamas and Eykyn20 studied 408 cases of native valve endocarditis; in 12.3%, the patient had a bicuspid aortic valve. In this subset, all were male, the mean age was 39 at diagnosis, 82% needed surgery, and the death rate was 14%.
Patients with bicuspid aortic valve do not routinely need antibiotics before dental and surgical procedures, but if they have had endocarditis in the past, they need antibiotics to prevent a recurrence.21
REGULAR MONITORING NEEDED
Because complications may be life-threatening, early detection of progressive disease by regular screening is critical. Echocardiographic evaluation of valvular function, ventricular dimensions and function, and diameter of the aortic root and ascending aorta should be performed in every patient with bicuspid aortic valve. If initial imaging is normal and there is no aortic dilation, imaging should be repeated every 5 to 10 years. If any abnormality is found, repeat imaging is needed every year.22
Magnetic resonance imaging (MRI) or computed tomographic (CT) angiography may be required to better assess the aorta for patients requiring a surgical intervention, or when aortic dimensions are not clearly visualized on TTE. MRI has 2 advantages over CT angiography: it poses no radiation risk, and it provides more information on left ventricular function and dimensions, in addition to valve assessment.23,24
No published study has compared MRI or CT angiography and transesophageal echocardiography (TEE), but in a study of 174 patients with dilated aortic root, combined TTE and TEE detected aortic valve morphology accurately in 98% of cases. As TEE is more invasive, it is not recommended for regular surveillance (Figures 1 and 2).25
FAMILY SCREENING RECOMMENDED
Close relatives should be evaluated for aortic valve and thoracic aortic disease.12,13,23,26
The American College of Cardiology (ACC) and the American Heart Association (AHA), backed by radiologic and cardiovascular associations, concur in recommending echocardiographic screening and routine screening of the thoracic aorta for aortic root dilation in first-degree relatives (ie, siblings, parents, and children) of patients with bicuspid aortic valve (class I recommendation).22,27,28
A comprehensive physical examination is recommended for family members in addition to TTE, with careful assessment of the aortic valve in short and long axes, and of the aortic root.14 If the aorta cannot be adequately evaluated with TTE, further assessment should be pursued with CT angiography or MRI.
EXERCISE RESTRICTIONS
The 2015 ACC/AHA guidelines for competitive athletes with cardiovascular abnormalities recommend annual screening with TTE or MRI angiography for athletes with bicuspid aortic valve and coexisting dilation of the ascending aorta (aortic diameter 40–42 mm in men and 36–39 mm in women) (class I recommendation, level of evidence C).29
Athletes with a bicuspid aortic valve and a normal aortic root and ascending aorta may participate in all competitive activities.29 However, those with a dilated aorta should avoid strenuous activities because of the increased risk of rupture.30 The ACC/AHA recommendations29 depend on the diameter of the ascending aorta and the nature of the sport:
- For an aortic diameter 40 to 42 mm in men or 36 to 39 mm in women, and no features of connective tissue disease or familial thoracic ascending aortic syndrome, low- and moderate-intensity sports with a low likelihood of significant body contact may be considered; consider avoiding intense weight training (class IIb, level of evidence C)
- For an aortic diameter 43 to 45 mm, low-intensity sports with a low likelihood of body contact may be considered (class IIb, level of evidence C)
- For an aortic diameter greater than 43 mm in men or greater than 40 mm in women, sports involving body collision should be avoided (class III, level of evidence C)
- For an aortic diameter greater than 45 mm, sports activities should be avoided (class III, level of evidence C).
PREGNANCY CONSIDERATIONS
Bicuspid aortic valve is associated with aortic dissection, mainly in the third trimester.31 Patients should ideally undergo echocardiographic screening before conception. The 2010 ACC/AHA guidelines for managing thoracic aortic disease recommend monthly or bimonthly echocardiography until delivery in pregnant women with a dilated thoracic aorta.22
Patients with bicuspid aortic valve and aortic root enlargement of more than 40 mm should have preconception counseling about surgery for aortic root replacement before becoming pregnant. If the diagnosis of enlarged aortic root is made during pregnancy, echocardiographic surveillance at 4- to 6-week intervals is indicated.32
SURGICAL MANAGEMENT
In the past, beta-blockers and angiotensin-converting enzyme inhibitors were recommended to minimize shear stress, with the goal of slowing progression of aortic dilation. However, evidence to support their use is inadequate.33,34
The only definitive treatment is surgery, with various procedures that lower the risk of death or dissection.24,35
The dimensions of the aortic root or ascending aorta should be examined vigilantly, according to the 2014 ACC/AHA guidelines27:
- Repairing the aortic sinuses or replacing the ascending aorta is indicated if the diameter of the aortic sinuses or ascending aorta is greater than 5.5 cm (class I, level of evidence B)
- Repairing the aortic sinuses or replacing the ascending aorta is reasonable if the diameter of the aortic sinuses or ascending aorta is greater than 5.0 cm and the patient has a risk factor for dissection such as a family history of aortic dissection or an increase in diameter of 0.5 cm or greater per year (class IIa, level of evidence C)
- Replacement of the ascending aorta is reasonable if the diameter of the ascending aorta is greater than 4.5 cm and the patient is undergoing aortic valve surgery for severe aortic stenosis or regurgitation.
Valve repair or replacement
Aortic valve repair or replacement is sometimes done separately from aortic root repair.
The value of aortic valve repair is debatable, but a series of 728 patients at Cleveland Clinic showed a very low mortality rate (0.41%) and an annual reoperation rate of 2.6% during up to 15 years of follow-up.36
Aortic valve replacement is usually considered for patients with severe valve dysfunction, abnormal left ventricular dimensions, or symptoms. It is important to determine if the patient is a good surgical candidate and to refer early for surgical evaluation to avoid the higher risk of death associated with emergency surgery.36
Transcatheter aortic valve replacement has been studied in patients deemed to be at too high a risk for surgical replacement. Short- and intermediate-term outcomes have been good in these patients, but long-term data are lacking.37
Surveillance after surgery
The type of operation determines postoperative surveillance.
After isolated aortic valve repair or replacement, patients should continue with surveillance at least annually to monitor for progressive aortopathy, as they remain at increased risk of dissection or rupture after isolated valve surgery, especially if they had aortic insufficiency preoperatively.38
After definitive surgery with replacement or repair of the ascending aorta, no clear recommendations have been established for continued surveillance. However, it is reasonable to image these patients with either MRI or CT angiography 3 to 5 years after their surgery to monitor for anastomotic complications.
CASE QUESTIONS ANSWERED
Our patient should undergo repeat TTE in 1 year. He should also undergo CT angiography of the ascending aorta if it is not seen by TTE. He can participate in low-intensity sports but should avoid intense weight training. His parents, siblings, and children should be screened for bicuspid aortic valve or associated aortopathies.
- Roberts WC. The congenitally bicuspid aortic valve. A study of 85 autopsy cases. Am J Cardiol 1970; 26(1):72–83. pmid:5427836
- Michalowska IM, Hryniewiecki T, Kwiatek P, Stoklosa P, Swoboda-Rydz U, Szymanski P. Coronary artery variants and anomalies in patients with bicuspid aortic valve. J Thorac Imaging 2016; 31(3):156–162. doi:10.1097/RTI.0000000000000205
- Sabet HY, Edwards WD, Tazelaar HD, Daly RC. Congenitally bicuspid aortic valves: a surgical pathology study of 542 cases (1991 through 1996) and a literature review of 2,715 additional cases. Mayo Clin Proc 1999; 74(1):14–26. doi:10.4065/74.1.14
- Tutar E, Ekici F, Atalay S, Nacar N. The prevalence of bicuspid aortic valve in newborns by echocardiographic screening. Am Heart J 2005; 150(3):513–515. doi:10.1016/j.ahj.2004.10.036
- Benson DW. The genetics of congenital heart disease: a point in the revolution. Cardiol Clin 2002; 20(3):385–394. pmid:12371007
- Emanuel R, Withers R, O’Brien K, Ross P, Feizi O. Congenitally bicuspid aortic valves. Clinicogenetic study of 41 families. Br Heart J 1978; 40(12):1402–1407. pmid:737099
- Giusti B, Sticchi E, De Cario R, Magi A, Nistri S, Pepe G. Genetic bases of bicuspid aortic valve: the contribution of traditional and high-throughput sequencing approaches on research and diagnosis. Front Physiol 2017; 8:612. doi:10.3389/fphys.2017.00612
- Sachdev V, Matura LA, Sidenko S, et al. Aortic valve disease in Turner syndrome. J Am Coll Cardiol 2008; 51(19):1904–1909. doi:10.1016/j.jacc.2008.02.035
- Duran AC, Frescura C, Sans-Coma V, Angelini A, Basso C, Thiene G. Bicuspid aortic valves in hearts with other congenital heart disease. J Heart Valve Dis 1995; 4(6):581–590. pmid:8611973
- De Rubens Figueroa J, Rodríguez LM, Hach JL, Del Castillo Ruíz V, Martínez HO. Cardiovascular spectrum in Williams-Beuren syndrome: the Mexican experience in 40 patients. Tex Heart Inst J 2008; 35(3):279–285. pmid:18941598
- Yuan SM, Jing H. The bicuspid aortic valve and related disorders. Sao Paulo Med J 2010; 128(5):296–301. pmid:21181071
- Cripe L, Andelfinger G, Martin LJ, Shooner K, Benson DW. Bicuspid aortic valve is heritable. J Am Coll Cardiol 2004; 44(1):138–143. doi:10.1016/j.jacc.2004.03.050
- Kerstjens-Frederikse WS, Sarvaas GJ, Ruiter JS, et al. Left ventricular outflow tract obstruction: should cardiac screening be offered to first-degree relatives? Heart 2011; 97(15):1228–1232. doi:10.1136/hrt.2010.211433
- Siu SC, Silversides CK. Bicuspid aortic valve disease. J Am Coll Cardiol 2010; 55(25):2789–2800. doi:10.1016/j.jacc.2009.12.068
- Michelena HI, Khanna AD, Mahoney D, et al. Incidence of aortic complications in patients with bicuspid aortic valves. JAMA 2011; 306(10):1104–1112.
- Michelena HI, Desjardins VA, Avierinos JF, et al. Natural history of asymptomatic patients with normally functioning or minimally dysfunctional bicuspid aortic valve in the community. Circulation 2008; 117(21):2776–2784. doi:10.1161/CIRCULATIONAHA.107.740878
- Beppu S, Suzuki S, Matsuda H, Ohmori F, Nagata S, Miyatake K. Rapidity of progression of aortic stenosis in patients with congenital bicuspid aortic valves. Am J Cardiol 1993; 71(4):322–327. pmid:8427176
- Roberts WC, Ko JM. Frequency by decades of unicuspid, bicuspid, and tricuspid aortic valves in adults having isolated aortic valve replacement for aortic stenosis, with or without associated aortic regurgitation. Circulation 2005; 111(7):920–925. doi:10.1161/01.CIR.0000155623.48408.C5
- Yener N, Oktar GL, Erer D, Yardimci MM, Yener A. Bicuspid aortic valve. Ann Thorac Cardiovasc Surg 2002; 8(5):264–267. pmid:12472407
- Lamas CC, Eykyn SJ. Bicuspid aortic valve—a silent danger: analysis of 50 cases of infective endocarditis. Clin Infect Dis 2000; 30(2):336–341. doi:10.1086/313646
- Wilson W, Taubert KA, Gewitz M, et al. Prevention of infective endocarditis: guidelines from the American Heart Association. Circulation 2007; 116(15):1736–1754. doi:10.1161/CIRCULATIONAHA.106.183095
- Hiratzka L, Bakris G, Beckman JA, et al. 2010 ACCF/AHA/AATS/ACR/ASA/SCA/SCAI/SIR/STS/SVM guidelines for the diagnosis and management of patients with thoracic aortic disease. Circulation 2010; 121(13):e266–e369. doi:10.1161/CIR.0b013e3181d4739e
- Chun EJ, Choi SI, Lim C, et al. Aortic stenosis: evaluation with multidetector CT angiography and MR imaging. Korean J Radiol 2008; 9(5):439–448. doi:10.3348/kjr.2008.9.5.439
- Kiefer TL, Wang A, Hughes GC, Bashore TM. Management of patients with bicuspid aortic valve disease. Curr Treat Options Cardiovasc Med 2011; 13(6):489–505. doi:10.1007/s11936-011-0152-7
- Alegret JM, Palazon O, Duran I, Vernis JM. Aortic valve morphology definition with transthoracic combined with transesophageal echocardiography in a population with high prevalence of bicuspid aortic valve. Int J Cardiovasc Imaging 2005; 21(2-3):213–217. doi:10.1007/s10554-004-3901-9
- Biner S, Rafique AM, Ray I, Cuk O, Siegel RJ, Tolstrup K. Aortopathy is prevalent in relatives of bicuspid aortic valve patients. J Am Coll Cardiol 2009; 53(24):2288–2295. doi:10.1016/j.jacc.2009.03.027
- Nishimura RA, Otto CM, Bonow RO, et al. 2014 AHA/ACC guideline for the management of patients with valvular heart disease. J Thorac Cardiovasc Surg 2014; 148(1):e1-e132. doi:10.1016/j.jtcvs.2014.05.014
- Warnes CA, Williams RG, Bashore TM, et al. ACC/AHA 2008 guidelines for the management of adults with congenital heart disease. J Am Coll Cardiol 2008; 52(23):e143–e263. doi:10.1016/j.jacc.2008.10.001
- Braverman AC, Harris KM, Kovacs RJ, Maron BJ. Eligibility and disqualification recommendations for competitive athletes with cardiovascular abnormalities: Task Force 7: aortic diseases, including Marfan syndrome. Circulation 2015; 132(22):e303–e309. doi:10.1161/CIR.0000000000000243
- De Mozzi P, Longo UG, Galanti G, Maffulli N. Bicuspid aortic valve: a literature review and its impact on sport activity. Br Med Bull 2008; 85:63–85. doi:10.1093/bmb/ldn002
- Thorne SA. Pregnancy in heart disease. Heart 2004; 90(4):450–456. pmid:15020530
- Immer FF, Bansi AG, Immer-Bansi AS, et al. Aortic dissection in pregnancy: analysis of risk factors and outcome. Ann Thorac Surg 2003; 76(1):309–314. pmid:12842575
- Allen BD, Markl M, Barker AJ, et al. Influence of beta-blocker therapy on aortic blood flow in patients with bicuspid aortic valve. Int J Cardiovasc Imaging 2016; 32(4):621–628. doi:10.1007/s10554-015-0819-3
- Ohnemus D, Oster ME, Gatlin S, Jokhadar M, Mahle WT. The effect of angiotensin-converting enzyme inhibitors on the rate of ascending aorta dilation in patients with bicuspid aortic valve. Congenit Heart Dis 2015; 10(1):E1–E5. doi:10.1111/chd.12184
- Masri A, Kalahasti V, Alkharabsheh S, et al. Characteristics and long-term outcomes of contemporary patients with bicuspid aortic valves. J Thorac Cardiovasc Surg 2016; 151(6):1650–1659.e1. doi:10.1016/j.jtcvs.2015.12.019
- Svensson LG, Al Kindi AH, Vivacqua A, et al. Long-term durability of bicuspid aortic valve repair. Ann Thorac Surg 2014; 97(5):1539–1548. doi:10.1016/j.athoracsur.2013.11.036
- Mylotte D, Lefevre T, Sondergaard L, et al. Transcatheter aortic valve replacement in bicuspid aortic valve disease. J Am Coll Cardiol 2014; 64(22):2330–2339. doi:10.1016/j.jacc.2014.09.039
- Girdauskas E, Disha K, Raisin HH, Secknus MA, Borger MA, Kuntze T. Risk of late aortic events after an isolated aortic valve replacement for bicuspid aortic valve stenosis with concomitant ascending aortic dilation. Eur J Cardiothorac Surg 2012; 42(5):832–838. doi:10.1093/ejcts/ezs137
- Roberts WC. The congenitally bicuspid aortic valve. A study of 85 autopsy cases. Am J Cardiol 1970; 26(1):72–83. pmid:5427836
- Michalowska IM, Hryniewiecki T, Kwiatek P, Stoklosa P, Swoboda-Rydz U, Szymanski P. Coronary artery variants and anomalies in patients with bicuspid aortic valve. J Thorac Imaging 2016; 31(3):156–162. doi:10.1097/RTI.0000000000000205
- Sabet HY, Edwards WD, Tazelaar HD, Daly RC. Congenitally bicuspid aortic valves: a surgical pathology study of 542 cases (1991 through 1996) and a literature review of 2,715 additional cases. Mayo Clin Proc 1999; 74(1):14–26. doi:10.4065/74.1.14
- Tutar E, Ekici F, Atalay S, Nacar N. The prevalence of bicuspid aortic valve in newborns by echocardiographic screening. Am Heart J 2005; 150(3):513–515. doi:10.1016/j.ahj.2004.10.036
- Benson DW. The genetics of congenital heart disease: a point in the revolution. Cardiol Clin 2002; 20(3):385–394. pmid:12371007
- Emanuel R, Withers R, O’Brien K, Ross P, Feizi O. Congenitally bicuspid aortic valves. Clinicogenetic study of 41 families. Br Heart J 1978; 40(12):1402–1407. pmid:737099
- Giusti B, Sticchi E, De Cario R, Magi A, Nistri S, Pepe G. Genetic bases of bicuspid aortic valve: the contribution of traditional and high-throughput sequencing approaches on research and diagnosis. Front Physiol 2017; 8:612. doi:10.3389/fphys.2017.00612
- Sachdev V, Matura LA, Sidenko S, et al. Aortic valve disease in Turner syndrome. J Am Coll Cardiol 2008; 51(19):1904–1909. doi:10.1016/j.jacc.2008.02.035
- Duran AC, Frescura C, Sans-Coma V, Angelini A, Basso C, Thiene G. Bicuspid aortic valves in hearts with other congenital heart disease. J Heart Valve Dis 1995; 4(6):581–590. pmid:8611973
- De Rubens Figueroa J, Rodríguez LM, Hach JL, Del Castillo Ruíz V, Martínez HO. Cardiovascular spectrum in Williams-Beuren syndrome: the Mexican experience in 40 patients. Tex Heart Inst J 2008; 35(3):279–285. pmid:18941598
- Yuan SM, Jing H. The bicuspid aortic valve and related disorders. Sao Paulo Med J 2010; 128(5):296–301. pmid:21181071
- Cripe L, Andelfinger G, Martin LJ, Shooner K, Benson DW. Bicuspid aortic valve is heritable. J Am Coll Cardiol 2004; 44(1):138–143. doi:10.1016/j.jacc.2004.03.050
- Kerstjens-Frederikse WS, Sarvaas GJ, Ruiter JS, et al. Left ventricular outflow tract obstruction: should cardiac screening be offered to first-degree relatives? Heart 2011; 97(15):1228–1232. doi:10.1136/hrt.2010.211433
- Siu SC, Silversides CK. Bicuspid aortic valve disease. J Am Coll Cardiol 2010; 55(25):2789–2800. doi:10.1016/j.jacc.2009.12.068
- Michelena HI, Khanna AD, Mahoney D, et al. Incidence of aortic complications in patients with bicuspid aortic valves. JAMA 2011; 306(10):1104–1112.
- Michelena HI, Desjardins VA, Avierinos JF, et al. Natural history of asymptomatic patients with normally functioning or minimally dysfunctional bicuspid aortic valve in the community. Circulation 2008; 117(21):2776–2784. doi:10.1161/CIRCULATIONAHA.107.740878
- Beppu S, Suzuki S, Matsuda H, Ohmori F, Nagata S, Miyatake K. Rapidity of progression of aortic stenosis in patients with congenital bicuspid aortic valves. Am J Cardiol 1993; 71(4):322–327. pmid:8427176
- Roberts WC, Ko JM. Frequency by decades of unicuspid, bicuspid, and tricuspid aortic valves in adults having isolated aortic valve replacement for aortic stenosis, with or without associated aortic regurgitation. Circulation 2005; 111(7):920–925. doi:10.1161/01.CIR.0000155623.48408.C5
- Yener N, Oktar GL, Erer D, Yardimci MM, Yener A. Bicuspid aortic valve. Ann Thorac Cardiovasc Surg 2002; 8(5):264–267. pmid:12472407
- Lamas CC, Eykyn SJ. Bicuspid aortic valve—a silent danger: analysis of 50 cases of infective endocarditis. Clin Infect Dis 2000; 30(2):336–341. doi:10.1086/313646
- Wilson W, Taubert KA, Gewitz M, et al. Prevention of infective endocarditis: guidelines from the American Heart Association. Circulation 2007; 116(15):1736–1754. doi:10.1161/CIRCULATIONAHA.106.183095
- Hiratzka L, Bakris G, Beckman JA, et al. 2010 ACCF/AHA/AATS/ACR/ASA/SCA/SCAI/SIR/STS/SVM guidelines for the diagnosis and management of patients with thoracic aortic disease. Circulation 2010; 121(13):e266–e369. doi:10.1161/CIR.0b013e3181d4739e
- Chun EJ, Choi SI, Lim C, et al. Aortic stenosis: evaluation with multidetector CT angiography and MR imaging. Korean J Radiol 2008; 9(5):439–448. doi:10.3348/kjr.2008.9.5.439
- Kiefer TL, Wang A, Hughes GC, Bashore TM. Management of patients with bicuspid aortic valve disease. Curr Treat Options Cardiovasc Med 2011; 13(6):489–505. doi:10.1007/s11936-011-0152-7
- Alegret JM, Palazon O, Duran I, Vernis JM. Aortic valve morphology definition with transthoracic combined with transesophageal echocardiography in a population with high prevalence of bicuspid aortic valve. Int J Cardiovasc Imaging 2005; 21(2-3):213–217. doi:10.1007/s10554-004-3901-9
- Biner S, Rafique AM, Ray I, Cuk O, Siegel RJ, Tolstrup K. Aortopathy is prevalent in relatives of bicuspid aortic valve patients. J Am Coll Cardiol 2009; 53(24):2288–2295. doi:10.1016/j.jacc.2009.03.027
- Nishimura RA, Otto CM, Bonow RO, et al. 2014 AHA/ACC guideline for the management of patients with valvular heart disease. J Thorac Cardiovasc Surg 2014; 148(1):e1-e132. doi:10.1016/j.jtcvs.2014.05.014
- Warnes CA, Williams RG, Bashore TM, et al. ACC/AHA 2008 guidelines for the management of adults with congenital heart disease. J Am Coll Cardiol 2008; 52(23):e143–e263. doi:10.1016/j.jacc.2008.10.001
- Braverman AC, Harris KM, Kovacs RJ, Maron BJ. Eligibility and disqualification recommendations for competitive athletes with cardiovascular abnormalities: Task Force 7: aortic diseases, including Marfan syndrome. Circulation 2015; 132(22):e303–e309. doi:10.1161/CIR.0000000000000243
- De Mozzi P, Longo UG, Galanti G, Maffulli N. Bicuspid aortic valve: a literature review and its impact on sport activity. Br Med Bull 2008; 85:63–85. doi:10.1093/bmb/ldn002
- Thorne SA. Pregnancy in heart disease. Heart 2004; 90(4):450–456. pmid:15020530
- Immer FF, Bansi AG, Immer-Bansi AS, et al. Aortic dissection in pregnancy: analysis of risk factors and outcome. Ann Thorac Surg 2003; 76(1):309–314. pmid:12842575
- Allen BD, Markl M, Barker AJ, et al. Influence of beta-blocker therapy on aortic blood flow in patients with bicuspid aortic valve. Int J Cardiovasc Imaging 2016; 32(4):621–628. doi:10.1007/s10554-015-0819-3
- Ohnemus D, Oster ME, Gatlin S, Jokhadar M, Mahle WT. The effect of angiotensin-converting enzyme inhibitors on the rate of ascending aorta dilation in patients with bicuspid aortic valve. Congenit Heart Dis 2015; 10(1):E1–E5. doi:10.1111/chd.12184
- Masri A, Kalahasti V, Alkharabsheh S, et al. Characteristics and long-term outcomes of contemporary patients with bicuspid aortic valves. J Thorac Cardiovasc Surg 2016; 151(6):1650–1659.e1. doi:10.1016/j.jtcvs.2015.12.019
- Svensson LG, Al Kindi AH, Vivacqua A, et al. Long-term durability of bicuspid aortic valve repair. Ann Thorac Surg 2014; 97(5):1539–1548. doi:10.1016/j.athoracsur.2013.11.036
- Mylotte D, Lefevre T, Sondergaard L, et al. Transcatheter aortic valve replacement in bicuspid aortic valve disease. J Am Coll Cardiol 2014; 64(22):2330–2339. doi:10.1016/j.jacc.2014.09.039
- Girdauskas E, Disha K, Raisin HH, Secknus MA, Borger MA, Kuntze T. Risk of late aortic events after an isolated aortic valve replacement for bicuspid aortic valve stenosis with concomitant ascending aortic dilation. Eur J Cardiothorac Surg 2012; 42(5):832–838. doi:10.1093/ejcts/ezs137
KEY POINTS
- Associated aortopathies such as aortic root dilation, aneurysm, dissection, and coarctation may initially be asymptomatic.
- Regular surveillance with transthoracic echocardiography (TTE) is required.
- Transesophageal echocardiography should be performed if TTE does not clearly show the aorta and aortic root. Magnetic resonance imaging or computed tomographic angiography may also be needed to measure the aortic root and ascending thoracic aorta.
- If initial imaging is normal and there is no aortic dilation, repeat imaging should be done every 5 to 10 years. If any abnormality is found, annual surveillance is needed.
- Women with a bicuspid aortic valve who are contemplating pregnancy should undergo echocardiography first, and some may need to undergo surgery.
Ablation of atrial fibrillation: Facts for the referring physician
A 64-year-old man with hypertension but without known structural heart disease presents for a second opinion on management of his atrial fibrillation. The condition was first diagnosed at age 38, when he experienced palpitations and shortness of breath on exertion; at times he also experienced decreased endurance and fatigue without overt palpitations. At first, these episodes occurred about twice a year, and the patient was managed with a beta-blocker for rate control and an oral anticoagulant.
Over the past 10 years, the episodes have become more frequent and longer-lasting and have required frequent cardioversions. He was given flecainide for rhythm control but continued to have frequent episodes, and so about 1 year ago he was switched to amiodarone, which controlled his rhythm better. However, after reading about side effects of amiodarone, he decided to seek a second opinion.
He was evaluated by our team and eventually underwent radiofrequency ablation. During the procedure, he was noted to have diffuse scarring and fibrosis of his left atrium, and afterward he continued to require antiarrhythmic drugs to maintain sinus rhythm.
Should he have been referred sooner? What factors should primary care physicians consider when referring a patient with atrial fibrillation for ablation?
THE EPIDEMIC OF ATRIAL FIBRILLATION
Atrial fibrillation is a large and growing public health problem. In 2010, it was estimated to affect 2.7 to 6.1 million people in the United States, and with the rapid aging of our population, its prevalence is expected to rise to between 5.6 and 12 million by 2050.1–3 It is associated with significant morbidity, poor quality of life, and increased risk of death, heart failure, stroke, and cognitive impairment.
The number of new cases per year has increased over the years despite research and preventive measures, which may reflect aging of the population and increased survival rates in patients with cardiovascular or comorbid conditions.1,4
Thus, atrial fibrillation is one of the most common cardiovascular conditions encountered by primary care physicians and cardiologists, putting them at the forefront of its management. Proper treatment in its early stages and referral to a specialist for advanced management may alter its natural history and improve clinical outcomes.
HOW DOES ATRIAL FIBRILLATION ARISE AND PERSIST?
Much is still unknown about the pathogenesis of atrial fibrillation, but considerable progress has been made in the past few decades, opening the door for clinical ablative strategies.
Multiple wavelet hypothesis
Until the late 1980s, the most widely accepted conceptual mechanism of atrial fibrillation was the multiple wavelet hypothesis developed by Moe et al.5 According to this hypothesis, atrial fibrillation begins with multiple independent wavelets occurring simultaneously and spreading randomly throughout both atria, and it persists if there are a minimum number of coexisting wavelets, increased atrial mass, and heterogeneous conduction delays across the atrial tissue.
The surgical maze procedure, in which a series of incisions arranged in a maze-like pattern is created in the left atrium, was predicated on this model. The theory was that these surgical lesions would compartmentalize the atria into discrete electrical segments and thereby reduce the number of circulating random wavelets.6,7
However, experimental and clinical studies suggest that although randomly propagating wavelets can contribute to maintaining atrial fibrillation, focal triggers are noted in most cases.
Focal triggers
In 1997, Jaïs et al8 observed that atrial fibrillation is often triggered by a rapidly firing ectopic focus and that ablation of that focus can eliminate it. These ectopic foci are often found at or near the ostia of the pulmonary veins or near the superior vena cava.8,9 It is now well established that ectopic foci in the pulmonary veins are crucial triggers that initiate atrial fibrillation.
Trigger-and-substrate theory
The substrate for maintaining atrial fibrillation consists of an abnormal left atrium with heterogeneous fibrosis (scarring) and conduction delays. Any heart disease that increases left atrial pressure could lead to atrial dilation and remodeling, which could be substrates for atrial fibrillation. Extensive atrial remodeling and scarring are associated with progression and persistence of atrial fibrillation and make rhythm control more challenging.
Atrial fibrillation begets atrial fibrillation
As shown in the case above, over time, paroxysmal atrial fibrillation often progresses to persistent and long-standing atrial fibrillation if not aggressively managed initially.
In 1972, Davies and Pomerance10 performed 100 autopsies and found that the people who had had atrial fibrillation for longer than 1 month had lost muscle mass in the sinus node and internodal tract, and their atria were dilated. The study introduced the concept that atrial fibrillation itself causes pathologic changes in the atrium.
Wijffels et al,11 in an experiment in goats, showed that atrial fibrillation produced by rapid bursts of atrial pacing was initially paroxysmal. However, as they continued to induce atrial fibrillation over and over again, it lasted progressively longer until it would persist for more than 24 hours. Thus, in a relatively short time, the atria went from supporting paroxysmal fibrillation to supporting persistent fibrillation.
Atrial fibrillation leads to electrophysiologic and anatomic remodeling in the atrium, which leads to a shorter action potential duration and a shorter refractory period. This in turn makes it easier for atrial fibrillation to persist.12
Because atrial fibrillation tends to progress, intervening early may improve its outcomes. Early ablation has been shown to improve the chances of staying in sinus rhythm in both paroxysmal and persistent atrial fibrillation.13–15
CATHETER ABLATION OF ATRIAL FIBRILLATION
The goal of ablation is to prevent atrial fibrillation by eliminating the trigger that initiates it, altering the arrhythmogenic substrate, or both.
Pulmonary vein isolation
The most common ablation strategy is to electrically isolate the pulmonary veins by creating circumferential lesions around their antra. This creates a nonconducting rim of scar tissue, electrically disconnecting the pulmonary veins from the atrium.
Ablation outside of the pulmonary veins
Because recurrence rates are high in patients with persistent atrial fibrillation who undergo pulmonary vein ablation alone, the search continues for adjunctive strategies to improve outcomes. Although these strategies have a sound rationale based on experimental data and anecdotal evidence in humans, they have not yet been convincingly shown to be helpful in large clinical studies. Nonetheless, it is possible that more extensive substrate ablation—atrial “debulking”—could improve outcomes by reducing the amount of tissue that can fibrillate.
Linear ablation. Creating lines of ablation (as in the maze procedure) isolates different segments of the left atrium. Often, these lines are created along the roof of the left atrium between the right and left upper pulmonary veins and from the mitral valve to the left inferior pulmonary vein. The benefit of linear ablation has not been proven, and gaps in such lines may introduce atrial flutter.
Triggers not in the pulmonary veins. Common sites of nonpulmonary vein triggers include the posterior wall of the left atrium, the superior vena cava, the coronary sinus, and along the ligament of Marshall. Provocative maneuvers such as isoproterenol infusion can help find those triggers, which can then be ablated. A limitation is that there is no protocol proven to reproducibly elicit triggers.
Complex fractionated atrial electrograms are areas in the atrium with highly fractionated, low voltage potentials. They may be critical sites of substrate for atrial fibrillation, and many electrophysiologists target them in patients with persistent atrial fibrillation. But despite initial enthusiasm, doing so has not resulted in better outcomes in persistent atrial fibrillation.
Rotors. Animal studies have shown that atrial fibrillation can be triggered or maintained by localized sources of organized reentrant circuits (rotors) or focal impulses. Recent studies have shown that these electrical rotors and focal sources could potentially be mapped and ablated in humans. But positive results in initial reports have not been reproduced, and this remains an area of controversy.
Our practice. We isolate the pulmonary veins with antral ablations, ablate the posterior wall, and extend the ablation toward the septum and inferior to the right pulmonary veins, with good long-term outcomes.14 The rationale behind ablating the posterior wall is that it shares embryologic origins with the pulmonary veins and may be a common source of triggers in atrial fibrillation.
We do not routinely create empiric ablation lines in the left or right atrium unless the patient has atrial flutter. Empiric ablation lines have not been convincingly shown to provide additional benefit compared with our extensive ablation approach, which involves the posterior wall. Empiric ablation of the appendage or coronary sinus is typically reserved for repeat ablation in patients with recurrent persistent atrial fibrillation.
RATIONALE FOR TREATING ATRIAL FIBRILLATION WITH ABLATION
To control symptoms
At this time, the primary aim of atrial fibrillation ablation is to reduce symptoms and improve quality of life. In theory, ablation could also decrease the risk of stroke, heart failure, and death. However, these outcomes have not been systematically evaluated in any large randomized controlled trial.
To control rhythm and improve survival
Randomized controlled trials of rhythm vs rate control of atrial fibrillation16–18 have failed to demonstrate that restoring sinus rhythm is associated with better survival. All of these trials used antiarrhythmic drugs for rhythm control. However, nonrandomized studies19,20 showed that maintaining sinus rhythm is associated with a significant reduction in mortality rates, whereas the use of antiarrhythmic drugs increased mortality risk.
This suggests that the beneficial effect of restoring sinus rhythm may be offset by adverse effects of antiarrhythmic drugs, and if rhythm control could be achieved by a method other than antiarrhythmic drug therapy, it may be superior to rate control. On the other hand, these data may be affected by residual confounding. This topic deserves further research, but maintaining sinus rhythm is typically preferred whenever possible.
Discontinuing anticoagulation is not a goal at this time
Retrospective studies have reported a low risk of stroke in patients who discontinue anticoagulation several months after undergoing atrial fibrillation ablation.21–23 However, atrial fibrillation can recur, and risk of stroke increases with age.
Therefore, guidelines24 still recommend continuing anticoagulation after ablation. Generally, we do not offer ablation with a goal of discontinuing anticoagulation. That said, stopping anticoagulation may be considered after long-term suppression of paroxysmal atrial fibrillation on a case-by-case basis in patients deemed to be at low risk. Left atrial appendage closure devices may eventually allow concomitant atrial fibrillation ablation and closure of the appendage, so that anticoagulation could then be stopped. This remains a topic of investigation.
Who should be considered for ablation?
There are no absolute age or comorbidity contraindications to ablation. Everyone who has atrial fibrillation deserves, in our opinion, a referral to the electrophysiology clinic.
PROCEDURAL CONSIDERATIONS
Atrial fibrillation ablation is most often performed by electrophysiologists using a minimally invasive endovascular approach. The patient can be under either moderate sedation or general anesthesia; we prefer general anesthesia for patient comfort, safety, and efficacy.
We use an electrogram-based technique to target and eliminate electrical potentials and ensure continuity of ablation sets, with additional guidance by 3-dimensional cardiac mapping systems and intracardiac echocardiography. We also use contact force-sensing catheters to ensure catheter-tissue contact during ablation and to avoid excessive contact, which may enhance the safety of the procedure.
Energy: Hot or cold
Two types of energy can be used for ablation:
Radiofrequency energy (low voltage, high frequency—30 kHz to 1.5 mHz) is delivered to the endocardial surface via a point-source catheter. The radiofrequency energy produces controlled, focal thermal ablation.
In a randomized trial,25 these ablation technologies were shown to be equivalent for preventing recurrences of atrial fibrillation. We use both in our practice. The choice depends primarily on the planned ablation set, given that balloon cryoablation can achieve antral isolation of the pulmonary veins but allows little or no substrate modification.
Improved ablation technology
Contact force-sensing catheters. Radiofrequency ablation catheters are now equipped with a pressure sensor at the tip that measures how hard the catheter is pressing on the heart wall.26,27 In our experience, this has improved the outcomes of ablation procedures, primarily in persistent atrial fibrillation.28
Complications of ablation
Although catheter ablation for atrial fibrillation is safe, it is still one of the most complex electrophysiologic procedures. Improvements in technology and techniques and accumulated experience over the past 15 years have made ablation safer, especially in tertiary care centers. But adverse outcomes are more frequent in low-volume centers.29
Minor procedural complications include pericarditis, complications at the site of vascular access, and anesthesia-related complications. While they do not affect the long-term outcome for the patient, they may increase hospital length of stay and cause temporary inconvenience.
Major complications include cardiac perforation and tamponade, periprocedural stroke, pulmonary vein stenosis, atrioesophageal fistula, phrenic nerve paralysis, major bleeding, myocardial infarction, and death. In a worldwide survey published in 2005, when atrial fibrillation ablation was still novel, the rate of major complications was 6%.30 By 2010, this had declined to 4.5%,31 and the rates of major complications may be significantly lower in more experienced centers.29 In our practice, in 2015, the rate of major complications was 1.3% (unpublished data).
Outcomes of catheter ablation
Clinical outcomes depend on many factors including the type of atrial fibrillation (paroxysmal vs nonparoxysmal), overall health of the atria (atrial size and scarring), patient age and comorbidities, and most importantly, the center’s and operator’s experience.
In randomized controlled trials comparing ablation and antiarrhythmic drug therapy, the efficacy of ablation in maintaining sinus rhythm has been in the range of 66% to 86% vs 16% to 22% for drug therapy,32,33 but these trials have been predominantly in middle-aged white men with paroxysmal atrial fibrillation. These trials also showed that catheter ablation reduced symptoms and improved quality of life. Ablation is less effective in persistent than in paroxysmal atrial fibrillation.34
In a long-term study from our group,14 660 (79.4%) of 831 patients who underwent ablation in 2005 were arrhythmia-free and not on antiarrhythmic drug therapy after a total of 1,019 ablations (an average of 1.2 ablations per patient) at a median of 55 months; 125 patients (15%, 41 with more than 1 ablation) continued to have atrial arrhythmia, controlled with drugs in 87 patients (69.6%). Only 38 patients (4.6%) continued to have drug-resistant atrial fibrillation and were treated with rate control with negative dromotropic agents.
Recent evidence
The largest randomized controlled trial of catheter ablation vs drug therapy for atrial fibrillation (Catheter Ablation Versus Antiarrhythmic Drug Therapy for Atrial Fibrillation [CABANA]) was completed recently, and the results were presented at a national meeting, although they have not yet been published in a peer-reviewed journal.35
A total of 2,204 patients with atrial fibrillation (42.4% paroxysmal, 47.3% persistent, and 10.3% long-standing persistent) were randomized to either ablation or drug therapy. Median follow-up was 4 years. The crossover rate was high—9.2% of those randomized to ablation did not undergo it, and 27.5% of those randomized to drug therapy underwent ablation.
The incidence of the primary end point (a composite of death, disabling stroke, serious bleeding, and cardiac arrest) was not significantly different between the 2 groups in the intention-to-treat analysis; however, given the high crossover rates, the as-treated and per-protocol analyses become important, and as-treated and per-protocol analyses revealed a significant benefit of ablation compared with drug therapy. The hazard ratio (HR) for the primary composite outcome was 0.67 (P = .006) on as-treated analysis and 0.73 (P = .05) on per-protocol analysis. The HR for all-cause mortality was 0.60 (P = .005) on as-treated analysis.
PERIPROCEDURAL CONSIDERATIONS
Periprocedural anticoagulation
The risk of thromboembolism is increased during, immediately following, and for several weeks to months after atrial fibrillation ablation.36,37
During the procedure, the risk is related to transseptal sheath placement, electrode catheters in the left atrium, and char formation on ablation catheters. These risks are mitigated with proper and careful sheath and catheter manipulation, maintenance of bubble-free irrigation through lines and sheaths, use of irrigated catheters, and initiation of heparin before transseptal access. Heparin is also infused during the procedure, with close monitoring of activated clotting time.
Postprocedurally, the transiently increased clotting risk could be due to damaged endothelium from the ablation itself and stunning of atrial tissue, which results in impaired contraction. Damaged endothelium improves as the tissue heals, and the stunning resolves by electrical reverse remodeling with sinus rhythm maintenance.
In view of these risks, the referring physician and electrophysiologist must pay careful attention to anticoagulation before and after ablation.
Before the procedure. It is safe to continue anticoagulation uninterrupted through the procedure.38,39 If the patient is on warfarin, we want the international normalized ratio to be in the therapeutic range when we perform atrial fibrillation ablation, and the patient takes his or her usual dose on the day of the procedure. If taking a direct oral anticoagulant, patients typically skip a dose the day before ablation and again on the morning of the procedure, and resume taking it immediately afterward while in the anesthesia recovery room.
During the procedure, we start heparin before transseptal puncture, adjust it to achieve an activated clotting time of 300 to 400 seconds, and keep it in this range as long as there are sheaths or catheters in the left atrium.
After the procedure. The current guidelines24 recommend that oral anticoagulation be continued without interruption for at least 2 months after the procedure, and in most cases indefinitely, depending on age and comorbidities. The decision to stop anticoagulation after 2 months is typically based on the stroke risk as assessed by the CHA2DS2-VASc score (www.chadsvasc.org) and not on the success of the ablation procedure.
ANTIARRHYTHMIC DRUGS AFTER THE PROCEDURE
Some patients actually experience more atrial fibrillation in the first weeks to months after the procedure. The mechanism in this setting may be different from that causing the arrhythmia in the first place. The causes of early recurrence of atrial arrhythmias include postablation inflammation, temporary autonomic imbalance, and delay of atrial radiofrequency lesion formation.40,41 These arrhythmias may completely resolve as the ablation lesions heal and scars mature.
It has been hypothesized that short-term use of antiarrhythmic drugs after atrial fibrillation ablation is effective in preventing arrhythmias because it alters atrial electrophysiologic characteristics induced by the above transient factors. A recent systematic review of 6 clinical trials showed that short-term use of antiarrhythmic drugs reduces the risk of early arrhythmia recurrence but does not reduce recurrence in the long term.42
In terms of outcomes, any arrhythmias that occur in the first 3 months do not necessarily affect long-term success. This is referred to as the “blanking period.” However, generally speaking, it is preferable to maintain sinus rhythm during that time to avoid further anatomic or electrical left atrial adverse remodeling. In many situations, patients continue taking the same antiarrhythmic agent or start on antiarrhythmic therapy in the first few months after ablation.43,44
The mechanisms of late recurrence of atrial arrhythmias after ablation are thought to be different from those in early recurrence. Late recurrence has been ascribed to incomplete pulmonary vein isolation, recovery of pulmonary vein-left atrium connections, or recovery of any other lines of ablation created in the procedure.45,46 For late recurrence of atrial arrhythmia, studies and guidelines suggest that repeat ablation may be an option.24,47
PRACTICAL CONSIDERATIONS FOR PROCEDURAL PLANNING
Before the procedure, some electrophysiologists use cardiac computed tomography or magnetic resonance imaging to evaluate the pulmonary vein anatomy. This helps in planning and in selecting the appropriate tools for the procedure.
The patient is asked to fast on the day of the procedure. The procedure can take 3 to 6 hours, depending on the patient’s anatomy and the operator’s technique and experience. It can be performed with the patient under general anesthesia or conscious sedation. Currently, we use general anesthesia most of the time to maximize patient comfort.
After the procedure, our patients must stay in bed for 4 hours and stay overnight for observation. If no complications arise, they are discharged the next day.
- Go AS. The epidemiology of atrial fibrillation in elderly persons: the tip of the iceberg. Am J Geriatr Cardiol 2005; 14(2):56–61. pmid:15785146
- Go AS, Hylek EM, Phillips KA, et al. Prevalence of diagnosed atrial fibrillation in adults: national implications for rhythm management and stroke prevention: the AnTicoagulation and Risk Factors in Atrial Fibrillation (ATRIA) Study. JAMA 2001; 285(18):2370–2375. pmid:11343485
- Miyasaka Y, Barnes ME, Gersh BJ, et al. Secular trends in incidence of atrial fibrillation in Olmsted County, Minnesota, 1980 to 2000, and implications on the projections for future prevalence. Circulation 2006; 114(2):119–125. doi:10.1161/CIRCULATIONAHA.105.595140
- Piccini JP, Hammill BG, Sinner MF, et al. Incidence and prevalence of atrial fibrillation and associated mortality among Medicare beneficiaries, 1993–2007. Circ Cardiovasc Qual Outcomes 2012; 5(1):85–93. doi:10.1161/CIRCOUTCOMES.111.962688
- Moe GK, Rheinboldt WC, Abildskov JA. A computer model of atrial fibrillation. Am Heart J 1964; 67:200–220. pmid:14118488
- Cox JL, Schuessler RB, Boineau JP. The surgical treatment of atrial fibrillation. I. Summary of the current concepts of the mechanisms of atrial flutter and atrial fibrillation. J Thorac Cardiovasc Surg 1991; 101(3):402–405. pmid:1999933
- Cox JL, Schuessler RB, D’Agostino HJ Jr, et al. The surgical treatment of atrial fibrillation. III. Development of a definitive surgical procedure. J Thorac Cardiovasc Surg 1991; 101(4):569–583. pmid:2008095
- Jaïs P, Haïssaguerre M, Shah DC, et al. A focal source of atrial fibrillation treated by discrete radiofrequency ablation. Circulation 1997; 95(3):572–576. pmid:9024141
- Haïssaguerre M, Jaïs P, Shah DC, et al. Spontaneous initiation of atrial fibrillation by ectopic beats originating in the pulmonary veins. N Engl J Med 1998; 339(10):659–666. doi:10.1056/NEJM199809033391003
- Davies MJ, Pomerance A. Pathology of atrial fibrillation in man. Br Heart J 1972; 34(5):520–525. pmid:5031645
- Wijffels MC, Kirchhof CJ, Dorland R, Allessie MA. Atrial fibrillation begets atrial fibrillation. A study in awake chronically instrumented goats. Circulation 1995; 92(7):1954–1968. pmid:7671380
- Nattel S. New ideas about atrial fibrillation 50 years on. Nature 2002; 415(6868):219–226. doi:10.1038/415219a
- Medi C, Sparks PB, Morton JB, et al. Pulmonary vein antral isolation for paroxysmal atrial fibrillation: results from long-term follow-up. J Cardiovasc Electrophysiol 2011; 22(2):137–141. doi:10.1111/j.1540-8167.2010.01885.x
- Hussein AA, Saliba WI, Martin DO, et al. Natural history and long-term outcomes of ablated atrial fibrillation. Circ Arrhythm Electrophysiol 2011; 4(3):271–278. doi:10.1161/CIRCEP.111.962100
- Hussein AA, Saliba WI, Barakat A, et al. Radiofrequency ablation of persistent atrial fibrillation: diagnosis-to-ablation time, markers of pathways of atrial remodeling, and outcomes. Circ Arrhythm Electrophysiol 2016; 9(1):e003669. doi:10.1161/CIRCEP.115.003669
- Carlsson J, Miketic S, Windeler J, et al. Randomized trial of rate-control versus rhythm-control in persistent atrial fibrillation: the Strategies of Treatment of Atrial Fibrillation (STAF) study. J Am Coll Cardiol 2003; 41(10):1690–1696. pmid:12767648
- Van Gelder IC, Hagens VE, Bosker HA, et al; Rate Control versus Electrical Cardioversion for Persistent Atrial Fibrillation Study Group. A comparison of rate control and rhythm control in patients with recurrent persistent atrial fibrillation. N Engl J Med 2002; 347(23):1834–1840. doi:10.1056/NEJMoa021375
- Wyse DG, Waldo AL, DiMarco JP, et al; Atrial Fibrillation Follow-up Investigation of Rhythm Management (AFFIRM) Investigators. A comparison of rate control and rhythm control in patients with atrial fibrillation. N Engl J Med 2002; 347(23):1825–1833. doi:10.1056/NEJMoa021328
- Hagens VE, Crijns HJ, Van Veldhuisen DJ, et al; RAte Control versus Electrical cardioversion for persistent atrial fibrillation study group. Rate control versus rhythm control for patients with persistent atrial fibrillation with mild to moderate heart failure: results from the RAte Control versus Electrical cardioversion (RACE) study. Am Heart J 2005; 149(6):1106–111. doi:10.1016/j.ahj.2004.11.030
- Pedersen OD, Bagger H, Keller N, Marchant B, Køber L, Torp-Pedersen C. Efficacy of dofetilide in the treatment of atrial fibrillation-flutter in patients with reduced left ventricular function: a Danish investigations of arrhythmia and mortality on dofetilide (diamond) substudy. Circulation 2001; 104(3):292–296. pmid:11457747
- Guiot A, Jongnarangsin K, Chugh A, et al. Anticoagulant therapy and risk of cerebrovascular events after catheter ablation of atrial fibrillation in the elderly. J Cardiovasc Electrophysiol 2012; 23(1):36–43. doi:10.1111/j.1540-8167.2011.02141.x
- Oral H, Chugh A, Ozaydin M, et al. Risk of thromboembolic events after percutaneous left atrial radiofrequency ablation of atrial fibrillation. Circulation 2006; 114(8):759–765. doi:10.1161/CIRCULATIONAHA.106.641225
- Themistoclakis S, Corrado A, Marchlinski FE, et al. The risk of thromboembolism and need for oral anticoagulation after successful atrial fibrillation ablation. J Am Coll Cardiol 2010; 55(8):735–743. doi:10.1016/j.jacc.2009.11.039
- Calkins H, Hindricks G, Cappato R, et al. 2017 HRS/EHRA/ECAS/APHRS/SOLAECE expert consensus statement on catheter and surgical ablation of atrial fibrillation: executive summary. J Arrhythm 2017; 33(5):369–409. doi:10.1016/j.joa.2017.08.001
- Kuck KH, Brugada J, Fürnkranz A, et al; FIRE AND ICE Investigators. Cryoballoon or radiofrequency ablation for paroxysmal atrial fibrillation. N Engl J Med 2016; 374(23):2235–2245. doi:10.1056/NEJMoa1602014
- Reddy VY, Dukkipati SR, Neuzil P, et al. Randomized, controlled trial of the safety and effectiveness of a contact force-sensing irrigated catheter for ablation of paroxysmal atrial fibrillation: results of the TactiCath Contact Force Ablation Catheter Study for Atrial Fibrillation (TOCCASTAR) Study. Circulation 2015; 132(10):907–915. doi:10.1161/CIRCULATIONAHA.114.014092
- Natale A, Reddy VY, Monir G, et al. Paroxysmal AF catheter ablation with a contact force sensing catheter: results of the prospective, multicenter SMART-AF trial. J Am Coll Cardiol 2014; 64(7):647–656. doi:10.1016/j.jacc.2014.04.072
- Hussein AA, Barakat AF, Saliba WI, et al. Persistent atrial fibrillation ablation with or without contact force sensing. J Cardiovasc Electrophysiol 2017; 28(5):483–488. doi:10.1111/jce.13179
- Deshmukh A, Patel NJ, Pant I, et al. In-hospital complications associated with catheter ablation of atrial fibrillation in the United States between 2000 and 2010: analysis of 93,801 procedures. Circulation 2013; 128(19):2104–2112. doi:10.1161/CIRCULATIONAHA.113.003862
- Cappato R, Calkins H, Chen SA, et al. Worldwide survey on the methods, efficacy, and safety of catheter ablation for human atrial fibrillation. Circulation 2005; 111(9):1100–1105. doi:10.1161/01.CIR.0000157153.30978.67
- Cappato R, Calkins H, Chen SA, et al. Updated worldwide survey on the methods, efficacy, and safety of catheter ablation for human atrial fibrillation. Circ Arrhythm Electrophysiol 2010; 3(1):32–38. doi:10.1161/CIRCEP.109.859116
- Wazni OM, Marrouche NF, Martin DO, et al. Radiofrequency ablation vs antiarrhythmic drugs as first-line treatment of symptomatic atrial fibrillation: a randomized trial. JAMA 2005; 293(21):2634–2640. doi:10.1001/jama.293.21.2634
- Jaïs P, Cauchemez B, Macle L, et al. Catheter ablation versus antiarrhythmic drugs for atrial fibrillation: the A4 study. Circulation 2008; 118(24):2498–2505. doi:10.1161/CIRCULATIONAHA.108.772582
- Brooks AG, Stiles MK, Laborderie J, et al. Outcomes of long-standing persistent atrial fibrillation ablation: a systematic review. Heart Rhythm 2010; 7(6):835–846. doi:10.1016/j.hrthm.2010.01.017
- Packer DL, Lee KL, Mark DB, Robb RA. Catheter Ablation versus Antiarrhythmic Drug Therapy for Atrial Fibrillation Trial, CABANA. http://cabanatrial.org/. Accessed September 10, 2018.
- Scherr D, Sharma K, Dalal D, et al. Incidence and predictors of periprocedural cerebrovascular accident in patients undergoing catheter ablation of atrial fibrillation. J Cardiovasc Electrophysiol 2009; 20(12):1357–1363. doi:10.1111/j.1540-8167.2009.01540.x
- Wazni OM, Rossillo A, Marrouche NF, et al. Embolic events and char formation during pulmonary vein isolation in patients with atrial fibrillation: impact of different anticoagulation regimens and importance of intracardiac echo imaging. J Cardiovasc Electrophysiol 2005; 16(6):576–581. doi:10.1111/j.1540-8167.2005.40480.x
- Hussein AA, Martin DO, Saliba W, et al. Radiofrequency ablation of atrial fibrillation under therapeutic international normalized ratio: a safe and efficacious periprocedural anticoagulation strategy. Heart Rhythm 2009; 6(10):1425–1429. doi:10.1016/j.hrthm.2009.07.007
- Bassiouny M, Saliba W, Rickard J, et al. Use of dabigatran for periprocedural anticoagulation in patients undergoing catheter ablation for atrial fibrillation. Circ Arrhythm Electrophysiol 2013; 6(3):460–466. doi:10.1161/CIRCEP.113.000320
- Koyama T, Tada H, Sekiguchi Y, et al. Prevention of atrial fibrillation recurrence with corticosteroids after radiofrequency catheter ablation: a randomized controlled trial. J Am Coll Cardiol 2010; 56(18):1463–1472. doi:10.1016/j.jacc.2010.04.057
- Oral H, Knight BP, Ozaydin M, et al. Clinical significance of early recurrences of atrial fibrillation after pulmonary vein isolation. J Am Coll Cardiol 2002; 40(1):100–104. pmid:12103262
- Chen W, Liu H, Ling Z, et al. Efficacy of short-term antiarrhythmic drugs use after catheter ablation of atrial fibrillation—a systematic review with meta-analyses and trial sequential analyses of randomized controlled trials. PLoS One 2016; 11(5):e0156121. doi:10.1371/journal.pone.0156121
- Leong-Sit P, Roux JF, Zado E, et al. Antiarrhythmics after ablation of atrial fibrillation (5A Study): six-month follow-up study. Circ Arrhythm Electrophysiol 2011; 4(1):11–14. doi:10.1161/CIRCEP.110.955393
- Roux JF, Zado E, Callans DJ, et al. Antiarrhythmics after ablation of atrial fibrillation (5A Study). Circulation 2009; 120(12):1036–1040. doi:10.1161/CIRCULATIONAHA.108.839639
- Sotomi Y, Inoue K, Ito N, et al. Cause of very late recurrence of atrial fibrillation or flutter after catheter ablation for atrial fibrillation. Am J Cardiol 2013; 111(4):552–556. doi:10.1016/j.amjcard.2012.10.040
- Lee SH, Tai CT, Hsieh MH, et al. Predictors of early and late recurrence of atrial fibrillation after catheter ablation of paroxysmal atrial fibrillation. J Interv Card Electrophysiol. 2004 Jun;10(3):221-6. doi:10.1023/B:JICE.0000026915.02503.92
- Zhang XD, Gu J, Jiang WF, et al. Optimal rhythm-control strategy for recurrent atrial tachycardia after catheter ablation of persistent atrial fibrillation: a randomized clinical trial. Eur Heart J 2014; 35(20):1327–1334. doi:10.1093/eurheartj/ehu017
A 64-year-old man with hypertension but without known structural heart disease presents for a second opinion on management of his atrial fibrillation. The condition was first diagnosed at age 38, when he experienced palpitations and shortness of breath on exertion; at times he also experienced decreased endurance and fatigue without overt palpitations. At first, these episodes occurred about twice a year, and the patient was managed with a beta-blocker for rate control and an oral anticoagulant.
Over the past 10 years, the episodes have become more frequent and longer-lasting and have required frequent cardioversions. He was given flecainide for rhythm control but continued to have frequent episodes, and so about 1 year ago he was switched to amiodarone, which controlled his rhythm better. However, after reading about side effects of amiodarone, he decided to seek a second opinion.
He was evaluated by our team and eventually underwent radiofrequency ablation. During the procedure, he was noted to have diffuse scarring and fibrosis of his left atrium, and afterward he continued to require antiarrhythmic drugs to maintain sinus rhythm.
Should he have been referred sooner? What factors should primary care physicians consider when referring a patient with atrial fibrillation for ablation?
THE EPIDEMIC OF ATRIAL FIBRILLATION
Atrial fibrillation is a large and growing public health problem. In 2010, it was estimated to affect 2.7 to 6.1 million people in the United States, and with the rapid aging of our population, its prevalence is expected to rise to between 5.6 and 12 million by 2050.1–3 It is associated with significant morbidity, poor quality of life, and increased risk of death, heart failure, stroke, and cognitive impairment.
The number of new cases per year has increased over the years despite research and preventive measures, which may reflect aging of the population and increased survival rates in patients with cardiovascular or comorbid conditions.1,4
Thus, atrial fibrillation is one of the most common cardiovascular conditions encountered by primary care physicians and cardiologists, putting them at the forefront of its management. Proper treatment in its early stages and referral to a specialist for advanced management may alter its natural history and improve clinical outcomes.
HOW DOES ATRIAL FIBRILLATION ARISE AND PERSIST?
Much is still unknown about the pathogenesis of atrial fibrillation, but considerable progress has been made in the past few decades, opening the door for clinical ablative strategies.
Multiple wavelet hypothesis
Until the late 1980s, the most widely accepted conceptual mechanism of atrial fibrillation was the multiple wavelet hypothesis developed by Moe et al.5 According to this hypothesis, atrial fibrillation begins with multiple independent wavelets occurring simultaneously and spreading randomly throughout both atria, and it persists if there are a minimum number of coexisting wavelets, increased atrial mass, and heterogeneous conduction delays across the atrial tissue.
The surgical maze procedure, in which a series of incisions arranged in a maze-like pattern is created in the left atrium, was predicated on this model. The theory was that these surgical lesions would compartmentalize the atria into discrete electrical segments and thereby reduce the number of circulating random wavelets.6,7
However, experimental and clinical studies suggest that although randomly propagating wavelets can contribute to maintaining atrial fibrillation, focal triggers are noted in most cases.
Focal triggers
In 1997, Jaïs et al8 observed that atrial fibrillation is often triggered by a rapidly firing ectopic focus and that ablation of that focus can eliminate it. These ectopic foci are often found at or near the ostia of the pulmonary veins or near the superior vena cava.8,9 It is now well established that ectopic foci in the pulmonary veins are crucial triggers that initiate atrial fibrillation.
Trigger-and-substrate theory
The substrate for maintaining atrial fibrillation consists of an abnormal left atrium with heterogeneous fibrosis (scarring) and conduction delays. Any heart disease that increases left atrial pressure could lead to atrial dilation and remodeling, which could be substrates for atrial fibrillation. Extensive atrial remodeling and scarring are associated with progression and persistence of atrial fibrillation and make rhythm control more challenging.
Atrial fibrillation begets atrial fibrillation
As shown in the case above, over time, paroxysmal atrial fibrillation often progresses to persistent and long-standing atrial fibrillation if not aggressively managed initially.
In 1972, Davies and Pomerance10 performed 100 autopsies and found that the people who had had atrial fibrillation for longer than 1 month had lost muscle mass in the sinus node and internodal tract, and their atria were dilated. The study introduced the concept that atrial fibrillation itself causes pathologic changes in the atrium.
Wijffels et al,11 in an experiment in goats, showed that atrial fibrillation produced by rapid bursts of atrial pacing was initially paroxysmal. However, as they continued to induce atrial fibrillation over and over again, it lasted progressively longer until it would persist for more than 24 hours. Thus, in a relatively short time, the atria went from supporting paroxysmal fibrillation to supporting persistent fibrillation.
Atrial fibrillation leads to electrophysiologic and anatomic remodeling in the atrium, which leads to a shorter action potential duration and a shorter refractory period. This in turn makes it easier for atrial fibrillation to persist.12
Because atrial fibrillation tends to progress, intervening early may improve its outcomes. Early ablation has been shown to improve the chances of staying in sinus rhythm in both paroxysmal and persistent atrial fibrillation.13–15
CATHETER ABLATION OF ATRIAL FIBRILLATION
The goal of ablation is to prevent atrial fibrillation by eliminating the trigger that initiates it, altering the arrhythmogenic substrate, or both.
Pulmonary vein isolation
The most common ablation strategy is to electrically isolate the pulmonary veins by creating circumferential lesions around their antra. This creates a nonconducting rim of scar tissue, electrically disconnecting the pulmonary veins from the atrium.
Ablation outside of the pulmonary veins
Because recurrence rates are high in patients with persistent atrial fibrillation who undergo pulmonary vein ablation alone, the search continues for adjunctive strategies to improve outcomes. Although these strategies have a sound rationale based on experimental data and anecdotal evidence in humans, they have not yet been convincingly shown to be helpful in large clinical studies. Nonetheless, it is possible that more extensive substrate ablation—atrial “debulking”—could improve outcomes by reducing the amount of tissue that can fibrillate.
Linear ablation. Creating lines of ablation (as in the maze procedure) isolates different segments of the left atrium. Often, these lines are created along the roof of the left atrium between the right and left upper pulmonary veins and from the mitral valve to the left inferior pulmonary vein. The benefit of linear ablation has not been proven, and gaps in such lines may introduce atrial flutter.
Triggers not in the pulmonary veins. Common sites of nonpulmonary vein triggers include the posterior wall of the left atrium, the superior vena cava, the coronary sinus, and along the ligament of Marshall. Provocative maneuvers such as isoproterenol infusion can help find those triggers, which can then be ablated. A limitation is that there is no protocol proven to reproducibly elicit triggers.
Complex fractionated atrial electrograms are areas in the atrium with highly fractionated, low voltage potentials. They may be critical sites of substrate for atrial fibrillation, and many electrophysiologists target them in patients with persistent atrial fibrillation. But despite initial enthusiasm, doing so has not resulted in better outcomes in persistent atrial fibrillation.
Rotors. Animal studies have shown that atrial fibrillation can be triggered or maintained by localized sources of organized reentrant circuits (rotors) or focal impulses. Recent studies have shown that these electrical rotors and focal sources could potentially be mapped and ablated in humans. But positive results in initial reports have not been reproduced, and this remains an area of controversy.
Our practice. We isolate the pulmonary veins with antral ablations, ablate the posterior wall, and extend the ablation toward the septum and inferior to the right pulmonary veins, with good long-term outcomes.14 The rationale behind ablating the posterior wall is that it shares embryologic origins with the pulmonary veins and may be a common source of triggers in atrial fibrillation.
We do not routinely create empiric ablation lines in the left or right atrium unless the patient has atrial flutter. Empiric ablation lines have not been convincingly shown to provide additional benefit compared with our extensive ablation approach, which involves the posterior wall. Empiric ablation of the appendage or coronary sinus is typically reserved for repeat ablation in patients with recurrent persistent atrial fibrillation.
RATIONALE FOR TREATING ATRIAL FIBRILLATION WITH ABLATION
To control symptoms
At this time, the primary aim of atrial fibrillation ablation is to reduce symptoms and improve quality of life. In theory, ablation could also decrease the risk of stroke, heart failure, and death. However, these outcomes have not been systematically evaluated in any large randomized controlled trial.
To control rhythm and improve survival
Randomized controlled trials of rhythm vs rate control of atrial fibrillation16–18 have failed to demonstrate that restoring sinus rhythm is associated with better survival. All of these trials used antiarrhythmic drugs for rhythm control. However, nonrandomized studies19,20 showed that maintaining sinus rhythm is associated with a significant reduction in mortality rates, whereas the use of antiarrhythmic drugs increased mortality risk.
This suggests that the beneficial effect of restoring sinus rhythm may be offset by adverse effects of antiarrhythmic drugs, and if rhythm control could be achieved by a method other than antiarrhythmic drug therapy, it may be superior to rate control. On the other hand, these data may be affected by residual confounding. This topic deserves further research, but maintaining sinus rhythm is typically preferred whenever possible.
Discontinuing anticoagulation is not a goal at this time
Retrospective studies have reported a low risk of stroke in patients who discontinue anticoagulation several months after undergoing atrial fibrillation ablation.21–23 However, atrial fibrillation can recur, and risk of stroke increases with age.
Therefore, guidelines24 still recommend continuing anticoagulation after ablation. Generally, we do not offer ablation with a goal of discontinuing anticoagulation. That said, stopping anticoagulation may be considered after long-term suppression of paroxysmal atrial fibrillation on a case-by-case basis in patients deemed to be at low risk. Left atrial appendage closure devices may eventually allow concomitant atrial fibrillation ablation and closure of the appendage, so that anticoagulation could then be stopped. This remains a topic of investigation.
Who should be considered for ablation?
There are no absolute age or comorbidity contraindications to ablation. Everyone who has atrial fibrillation deserves, in our opinion, a referral to the electrophysiology clinic.
PROCEDURAL CONSIDERATIONS
Atrial fibrillation ablation is most often performed by electrophysiologists using a minimally invasive endovascular approach. The patient can be under either moderate sedation or general anesthesia; we prefer general anesthesia for patient comfort, safety, and efficacy.
We use an electrogram-based technique to target and eliminate electrical potentials and ensure continuity of ablation sets, with additional guidance by 3-dimensional cardiac mapping systems and intracardiac echocardiography. We also use contact force-sensing catheters to ensure catheter-tissue contact during ablation and to avoid excessive contact, which may enhance the safety of the procedure.
Energy: Hot or cold
Two types of energy can be used for ablation:
Radiofrequency energy (low voltage, high frequency—30 kHz to 1.5 mHz) is delivered to the endocardial surface via a point-source catheter. The radiofrequency energy produces controlled, focal thermal ablation.
In a randomized trial,25 these ablation technologies were shown to be equivalent for preventing recurrences of atrial fibrillation. We use both in our practice. The choice depends primarily on the planned ablation set, given that balloon cryoablation can achieve antral isolation of the pulmonary veins but allows little or no substrate modification.
Improved ablation technology
Contact force-sensing catheters. Radiofrequency ablation catheters are now equipped with a pressure sensor at the tip that measures how hard the catheter is pressing on the heart wall.26,27 In our experience, this has improved the outcomes of ablation procedures, primarily in persistent atrial fibrillation.28
Complications of ablation
Although catheter ablation for atrial fibrillation is safe, it is still one of the most complex electrophysiologic procedures. Improvements in technology and techniques and accumulated experience over the past 15 years have made ablation safer, especially in tertiary care centers. But adverse outcomes are more frequent in low-volume centers.29
Minor procedural complications include pericarditis, complications at the site of vascular access, and anesthesia-related complications. While they do not affect the long-term outcome for the patient, they may increase hospital length of stay and cause temporary inconvenience.
Major complications include cardiac perforation and tamponade, periprocedural stroke, pulmonary vein stenosis, atrioesophageal fistula, phrenic nerve paralysis, major bleeding, myocardial infarction, and death. In a worldwide survey published in 2005, when atrial fibrillation ablation was still novel, the rate of major complications was 6%.30 By 2010, this had declined to 4.5%,31 and the rates of major complications may be significantly lower in more experienced centers.29 In our practice, in 2015, the rate of major complications was 1.3% (unpublished data).
Outcomes of catheter ablation
Clinical outcomes depend on many factors including the type of atrial fibrillation (paroxysmal vs nonparoxysmal), overall health of the atria (atrial size and scarring), patient age and comorbidities, and most importantly, the center’s and operator’s experience.
In randomized controlled trials comparing ablation and antiarrhythmic drug therapy, the efficacy of ablation in maintaining sinus rhythm has been in the range of 66% to 86% vs 16% to 22% for drug therapy,32,33 but these trials have been predominantly in middle-aged white men with paroxysmal atrial fibrillation. These trials also showed that catheter ablation reduced symptoms and improved quality of life. Ablation is less effective in persistent than in paroxysmal atrial fibrillation.34
In a long-term study from our group,14 660 (79.4%) of 831 patients who underwent ablation in 2005 were arrhythmia-free and not on antiarrhythmic drug therapy after a total of 1,019 ablations (an average of 1.2 ablations per patient) at a median of 55 months; 125 patients (15%, 41 with more than 1 ablation) continued to have atrial arrhythmia, controlled with drugs in 87 patients (69.6%). Only 38 patients (4.6%) continued to have drug-resistant atrial fibrillation and were treated with rate control with negative dromotropic agents.
Recent evidence
The largest randomized controlled trial of catheter ablation vs drug therapy for atrial fibrillation (Catheter Ablation Versus Antiarrhythmic Drug Therapy for Atrial Fibrillation [CABANA]) was completed recently, and the results were presented at a national meeting, although they have not yet been published in a peer-reviewed journal.35
A total of 2,204 patients with atrial fibrillation (42.4% paroxysmal, 47.3% persistent, and 10.3% long-standing persistent) were randomized to either ablation or drug therapy. Median follow-up was 4 years. The crossover rate was high—9.2% of those randomized to ablation did not undergo it, and 27.5% of those randomized to drug therapy underwent ablation.
The incidence of the primary end point (a composite of death, disabling stroke, serious bleeding, and cardiac arrest) was not significantly different between the 2 groups in the intention-to-treat analysis; however, given the high crossover rates, the as-treated and per-protocol analyses become important, and as-treated and per-protocol analyses revealed a significant benefit of ablation compared with drug therapy. The hazard ratio (HR) for the primary composite outcome was 0.67 (P = .006) on as-treated analysis and 0.73 (P = .05) on per-protocol analysis. The HR for all-cause mortality was 0.60 (P = .005) on as-treated analysis.
PERIPROCEDURAL CONSIDERATIONS
Periprocedural anticoagulation
The risk of thromboembolism is increased during, immediately following, and for several weeks to months after atrial fibrillation ablation.36,37
During the procedure, the risk is related to transseptal sheath placement, electrode catheters in the left atrium, and char formation on ablation catheters. These risks are mitigated with proper and careful sheath and catheter manipulation, maintenance of bubble-free irrigation through lines and sheaths, use of irrigated catheters, and initiation of heparin before transseptal access. Heparin is also infused during the procedure, with close monitoring of activated clotting time.
Postprocedurally, the transiently increased clotting risk could be due to damaged endothelium from the ablation itself and stunning of atrial tissue, which results in impaired contraction. Damaged endothelium improves as the tissue heals, and the stunning resolves by electrical reverse remodeling with sinus rhythm maintenance.
In view of these risks, the referring physician and electrophysiologist must pay careful attention to anticoagulation before and after ablation.
Before the procedure. It is safe to continue anticoagulation uninterrupted through the procedure.38,39 If the patient is on warfarin, we want the international normalized ratio to be in the therapeutic range when we perform atrial fibrillation ablation, and the patient takes his or her usual dose on the day of the procedure. If taking a direct oral anticoagulant, patients typically skip a dose the day before ablation and again on the morning of the procedure, and resume taking it immediately afterward while in the anesthesia recovery room.
During the procedure, we start heparin before transseptal puncture, adjust it to achieve an activated clotting time of 300 to 400 seconds, and keep it in this range as long as there are sheaths or catheters in the left atrium.
After the procedure. The current guidelines24 recommend that oral anticoagulation be continued without interruption for at least 2 months after the procedure, and in most cases indefinitely, depending on age and comorbidities. The decision to stop anticoagulation after 2 months is typically based on the stroke risk as assessed by the CHA2DS2-VASc score (www.chadsvasc.org) and not on the success of the ablation procedure.
ANTIARRHYTHMIC DRUGS AFTER THE PROCEDURE
Some patients actually experience more atrial fibrillation in the first weeks to months after the procedure. The mechanism in this setting may be different from that causing the arrhythmia in the first place. The causes of early recurrence of atrial arrhythmias include postablation inflammation, temporary autonomic imbalance, and delay of atrial radiofrequency lesion formation.40,41 These arrhythmias may completely resolve as the ablation lesions heal and scars mature.
It has been hypothesized that short-term use of antiarrhythmic drugs after atrial fibrillation ablation is effective in preventing arrhythmias because it alters atrial electrophysiologic characteristics induced by the above transient factors. A recent systematic review of 6 clinical trials showed that short-term use of antiarrhythmic drugs reduces the risk of early arrhythmia recurrence but does not reduce recurrence in the long term.42
In terms of outcomes, any arrhythmias that occur in the first 3 months do not necessarily affect long-term success. This is referred to as the “blanking period.” However, generally speaking, it is preferable to maintain sinus rhythm during that time to avoid further anatomic or electrical left atrial adverse remodeling. In many situations, patients continue taking the same antiarrhythmic agent or start on antiarrhythmic therapy in the first few months after ablation.43,44
The mechanisms of late recurrence of atrial arrhythmias after ablation are thought to be different from those in early recurrence. Late recurrence has been ascribed to incomplete pulmonary vein isolation, recovery of pulmonary vein-left atrium connections, or recovery of any other lines of ablation created in the procedure.45,46 For late recurrence of atrial arrhythmia, studies and guidelines suggest that repeat ablation may be an option.24,47
PRACTICAL CONSIDERATIONS FOR PROCEDURAL PLANNING
Before the procedure, some electrophysiologists use cardiac computed tomography or magnetic resonance imaging to evaluate the pulmonary vein anatomy. This helps in planning and in selecting the appropriate tools for the procedure.
The patient is asked to fast on the day of the procedure. The procedure can take 3 to 6 hours, depending on the patient’s anatomy and the operator’s technique and experience. It can be performed with the patient under general anesthesia or conscious sedation. Currently, we use general anesthesia most of the time to maximize patient comfort.
After the procedure, our patients must stay in bed for 4 hours and stay overnight for observation. If no complications arise, they are discharged the next day.
A 64-year-old man with hypertension but without known structural heart disease presents for a second opinion on management of his atrial fibrillation. The condition was first diagnosed at age 38, when he experienced palpitations and shortness of breath on exertion; at times he also experienced decreased endurance and fatigue without overt palpitations. At first, these episodes occurred about twice a year, and the patient was managed with a beta-blocker for rate control and an oral anticoagulant.
Over the past 10 years, the episodes have become more frequent and longer-lasting and have required frequent cardioversions. He was given flecainide for rhythm control but continued to have frequent episodes, and so about 1 year ago he was switched to amiodarone, which controlled his rhythm better. However, after reading about side effects of amiodarone, he decided to seek a second opinion.
He was evaluated by our team and eventually underwent radiofrequency ablation. During the procedure, he was noted to have diffuse scarring and fibrosis of his left atrium, and afterward he continued to require antiarrhythmic drugs to maintain sinus rhythm.
Should he have been referred sooner? What factors should primary care physicians consider when referring a patient with atrial fibrillation for ablation?
THE EPIDEMIC OF ATRIAL FIBRILLATION
Atrial fibrillation is a large and growing public health problem. In 2010, it was estimated to affect 2.7 to 6.1 million people in the United States, and with the rapid aging of our population, its prevalence is expected to rise to between 5.6 and 12 million by 2050.1–3 It is associated with significant morbidity, poor quality of life, and increased risk of death, heart failure, stroke, and cognitive impairment.
The number of new cases per year has increased over the years despite research and preventive measures, which may reflect aging of the population and increased survival rates in patients with cardiovascular or comorbid conditions.1,4
Thus, atrial fibrillation is one of the most common cardiovascular conditions encountered by primary care physicians and cardiologists, putting them at the forefront of its management. Proper treatment in its early stages and referral to a specialist for advanced management may alter its natural history and improve clinical outcomes.
HOW DOES ATRIAL FIBRILLATION ARISE AND PERSIST?
Much is still unknown about the pathogenesis of atrial fibrillation, but considerable progress has been made in the past few decades, opening the door for clinical ablative strategies.
Multiple wavelet hypothesis
Until the late 1980s, the most widely accepted conceptual mechanism of atrial fibrillation was the multiple wavelet hypothesis developed by Moe et al.5 According to this hypothesis, atrial fibrillation begins with multiple independent wavelets occurring simultaneously and spreading randomly throughout both atria, and it persists if there are a minimum number of coexisting wavelets, increased atrial mass, and heterogeneous conduction delays across the atrial tissue.
The surgical maze procedure, in which a series of incisions arranged in a maze-like pattern is created in the left atrium, was predicated on this model. The theory was that these surgical lesions would compartmentalize the atria into discrete electrical segments and thereby reduce the number of circulating random wavelets.6,7
However, experimental and clinical studies suggest that although randomly propagating wavelets can contribute to maintaining atrial fibrillation, focal triggers are noted in most cases.
Focal triggers
In 1997, Jaïs et al8 observed that atrial fibrillation is often triggered by a rapidly firing ectopic focus and that ablation of that focus can eliminate it. These ectopic foci are often found at or near the ostia of the pulmonary veins or near the superior vena cava.8,9 It is now well established that ectopic foci in the pulmonary veins are crucial triggers that initiate atrial fibrillation.
Trigger-and-substrate theory
The substrate for maintaining atrial fibrillation consists of an abnormal left atrium with heterogeneous fibrosis (scarring) and conduction delays. Any heart disease that increases left atrial pressure could lead to atrial dilation and remodeling, which could be substrates for atrial fibrillation. Extensive atrial remodeling and scarring are associated with progression and persistence of atrial fibrillation and make rhythm control more challenging.
Atrial fibrillation begets atrial fibrillation
As shown in the case above, over time, paroxysmal atrial fibrillation often progresses to persistent and long-standing atrial fibrillation if not aggressively managed initially.
In 1972, Davies and Pomerance10 performed 100 autopsies and found that the people who had had atrial fibrillation for longer than 1 month had lost muscle mass in the sinus node and internodal tract, and their atria were dilated. The study introduced the concept that atrial fibrillation itself causes pathologic changes in the atrium.
Wijffels et al,11 in an experiment in goats, showed that atrial fibrillation produced by rapid bursts of atrial pacing was initially paroxysmal. However, as they continued to induce atrial fibrillation over and over again, it lasted progressively longer until it would persist for more than 24 hours. Thus, in a relatively short time, the atria went from supporting paroxysmal fibrillation to supporting persistent fibrillation.
Atrial fibrillation leads to electrophysiologic and anatomic remodeling in the atrium, which leads to a shorter action potential duration and a shorter refractory period. This in turn makes it easier for atrial fibrillation to persist.12
Because atrial fibrillation tends to progress, intervening early may improve its outcomes. Early ablation has been shown to improve the chances of staying in sinus rhythm in both paroxysmal and persistent atrial fibrillation.13–15
CATHETER ABLATION OF ATRIAL FIBRILLATION
The goal of ablation is to prevent atrial fibrillation by eliminating the trigger that initiates it, altering the arrhythmogenic substrate, or both.
Pulmonary vein isolation
The most common ablation strategy is to electrically isolate the pulmonary veins by creating circumferential lesions around their antra. This creates a nonconducting rim of scar tissue, electrically disconnecting the pulmonary veins from the atrium.
Ablation outside of the pulmonary veins
Because recurrence rates are high in patients with persistent atrial fibrillation who undergo pulmonary vein ablation alone, the search continues for adjunctive strategies to improve outcomes. Although these strategies have a sound rationale based on experimental data and anecdotal evidence in humans, they have not yet been convincingly shown to be helpful in large clinical studies. Nonetheless, it is possible that more extensive substrate ablation—atrial “debulking”—could improve outcomes by reducing the amount of tissue that can fibrillate.
Linear ablation. Creating lines of ablation (as in the maze procedure) isolates different segments of the left atrium. Often, these lines are created along the roof of the left atrium between the right and left upper pulmonary veins and from the mitral valve to the left inferior pulmonary vein. The benefit of linear ablation has not been proven, and gaps in such lines may introduce atrial flutter.
Triggers not in the pulmonary veins. Common sites of nonpulmonary vein triggers include the posterior wall of the left atrium, the superior vena cava, the coronary sinus, and along the ligament of Marshall. Provocative maneuvers such as isoproterenol infusion can help find those triggers, which can then be ablated. A limitation is that there is no protocol proven to reproducibly elicit triggers.
Complex fractionated atrial electrograms are areas in the atrium with highly fractionated, low voltage potentials. They may be critical sites of substrate for atrial fibrillation, and many electrophysiologists target them in patients with persistent atrial fibrillation. But despite initial enthusiasm, doing so has not resulted in better outcomes in persistent atrial fibrillation.
Rotors. Animal studies have shown that atrial fibrillation can be triggered or maintained by localized sources of organized reentrant circuits (rotors) or focal impulses. Recent studies have shown that these electrical rotors and focal sources could potentially be mapped and ablated in humans. But positive results in initial reports have not been reproduced, and this remains an area of controversy.
Our practice. We isolate the pulmonary veins with antral ablations, ablate the posterior wall, and extend the ablation toward the septum and inferior to the right pulmonary veins, with good long-term outcomes.14 The rationale behind ablating the posterior wall is that it shares embryologic origins with the pulmonary veins and may be a common source of triggers in atrial fibrillation.
We do not routinely create empiric ablation lines in the left or right atrium unless the patient has atrial flutter. Empiric ablation lines have not been convincingly shown to provide additional benefit compared with our extensive ablation approach, which involves the posterior wall. Empiric ablation of the appendage or coronary sinus is typically reserved for repeat ablation in patients with recurrent persistent atrial fibrillation.
RATIONALE FOR TREATING ATRIAL FIBRILLATION WITH ABLATION
To control symptoms
At this time, the primary aim of atrial fibrillation ablation is to reduce symptoms and improve quality of life. In theory, ablation could also decrease the risk of stroke, heart failure, and death. However, these outcomes have not been systematically evaluated in any large randomized controlled trial.
To control rhythm and improve survival
Randomized controlled trials of rhythm vs rate control of atrial fibrillation16–18 have failed to demonstrate that restoring sinus rhythm is associated with better survival. All of these trials used antiarrhythmic drugs for rhythm control. However, nonrandomized studies19,20 showed that maintaining sinus rhythm is associated with a significant reduction in mortality rates, whereas the use of antiarrhythmic drugs increased mortality risk.
This suggests that the beneficial effect of restoring sinus rhythm may be offset by adverse effects of antiarrhythmic drugs, and if rhythm control could be achieved by a method other than antiarrhythmic drug therapy, it may be superior to rate control. On the other hand, these data may be affected by residual confounding. This topic deserves further research, but maintaining sinus rhythm is typically preferred whenever possible.
Discontinuing anticoagulation is not a goal at this time
Retrospective studies have reported a low risk of stroke in patients who discontinue anticoagulation several months after undergoing atrial fibrillation ablation.21–23 However, atrial fibrillation can recur, and risk of stroke increases with age.
Therefore, guidelines24 still recommend continuing anticoagulation after ablation. Generally, we do not offer ablation with a goal of discontinuing anticoagulation. That said, stopping anticoagulation may be considered after long-term suppression of paroxysmal atrial fibrillation on a case-by-case basis in patients deemed to be at low risk. Left atrial appendage closure devices may eventually allow concomitant atrial fibrillation ablation and closure of the appendage, so that anticoagulation could then be stopped. This remains a topic of investigation.
Who should be considered for ablation?
There are no absolute age or comorbidity contraindications to ablation. Everyone who has atrial fibrillation deserves, in our opinion, a referral to the electrophysiology clinic.
PROCEDURAL CONSIDERATIONS
Atrial fibrillation ablation is most often performed by electrophysiologists using a minimally invasive endovascular approach. The patient can be under either moderate sedation or general anesthesia; we prefer general anesthesia for patient comfort, safety, and efficacy.
We use an electrogram-based technique to target and eliminate electrical potentials and ensure continuity of ablation sets, with additional guidance by 3-dimensional cardiac mapping systems and intracardiac echocardiography. We also use contact force-sensing catheters to ensure catheter-tissue contact during ablation and to avoid excessive contact, which may enhance the safety of the procedure.
Energy: Hot or cold
Two types of energy can be used for ablation:
Radiofrequency energy (low voltage, high frequency—30 kHz to 1.5 mHz) is delivered to the endocardial surface via a point-source catheter. The radiofrequency energy produces controlled, focal thermal ablation.
In a randomized trial,25 these ablation technologies were shown to be equivalent for preventing recurrences of atrial fibrillation. We use both in our practice. The choice depends primarily on the planned ablation set, given that balloon cryoablation can achieve antral isolation of the pulmonary veins but allows little or no substrate modification.
Improved ablation technology
Contact force-sensing catheters. Radiofrequency ablation catheters are now equipped with a pressure sensor at the tip that measures how hard the catheter is pressing on the heart wall.26,27 In our experience, this has improved the outcomes of ablation procedures, primarily in persistent atrial fibrillation.28
Complications of ablation
Although catheter ablation for atrial fibrillation is safe, it is still one of the most complex electrophysiologic procedures. Improvements in technology and techniques and accumulated experience over the past 15 years have made ablation safer, especially in tertiary care centers. But adverse outcomes are more frequent in low-volume centers.29
Minor procedural complications include pericarditis, complications at the site of vascular access, and anesthesia-related complications. While they do not affect the long-term outcome for the patient, they may increase hospital length of stay and cause temporary inconvenience.
Major complications include cardiac perforation and tamponade, periprocedural stroke, pulmonary vein stenosis, atrioesophageal fistula, phrenic nerve paralysis, major bleeding, myocardial infarction, and death. In a worldwide survey published in 2005, when atrial fibrillation ablation was still novel, the rate of major complications was 6%.30 By 2010, this had declined to 4.5%,31 and the rates of major complications may be significantly lower in more experienced centers.29 In our practice, in 2015, the rate of major complications was 1.3% (unpublished data).
Outcomes of catheter ablation
Clinical outcomes depend on many factors including the type of atrial fibrillation (paroxysmal vs nonparoxysmal), overall health of the atria (atrial size and scarring), patient age and comorbidities, and most importantly, the center’s and operator’s experience.
In randomized controlled trials comparing ablation and antiarrhythmic drug therapy, the efficacy of ablation in maintaining sinus rhythm has been in the range of 66% to 86% vs 16% to 22% for drug therapy,32,33 but these trials have been predominantly in middle-aged white men with paroxysmal atrial fibrillation. These trials also showed that catheter ablation reduced symptoms and improved quality of life. Ablation is less effective in persistent than in paroxysmal atrial fibrillation.34
In a long-term study from our group,14 660 (79.4%) of 831 patients who underwent ablation in 2005 were arrhythmia-free and not on antiarrhythmic drug therapy after a total of 1,019 ablations (an average of 1.2 ablations per patient) at a median of 55 months; 125 patients (15%, 41 with more than 1 ablation) continued to have atrial arrhythmia, controlled with drugs in 87 patients (69.6%). Only 38 patients (4.6%) continued to have drug-resistant atrial fibrillation and were treated with rate control with negative dromotropic agents.
Recent evidence
The largest randomized controlled trial of catheter ablation vs drug therapy for atrial fibrillation (Catheter Ablation Versus Antiarrhythmic Drug Therapy for Atrial Fibrillation [CABANA]) was completed recently, and the results were presented at a national meeting, although they have not yet been published in a peer-reviewed journal.35
A total of 2,204 patients with atrial fibrillation (42.4% paroxysmal, 47.3% persistent, and 10.3% long-standing persistent) were randomized to either ablation or drug therapy. Median follow-up was 4 years. The crossover rate was high—9.2% of those randomized to ablation did not undergo it, and 27.5% of those randomized to drug therapy underwent ablation.
The incidence of the primary end point (a composite of death, disabling stroke, serious bleeding, and cardiac arrest) was not significantly different between the 2 groups in the intention-to-treat analysis; however, given the high crossover rates, the as-treated and per-protocol analyses become important, and as-treated and per-protocol analyses revealed a significant benefit of ablation compared with drug therapy. The hazard ratio (HR) for the primary composite outcome was 0.67 (P = .006) on as-treated analysis and 0.73 (P = .05) on per-protocol analysis. The HR for all-cause mortality was 0.60 (P = .005) on as-treated analysis.
PERIPROCEDURAL CONSIDERATIONS
Periprocedural anticoagulation
The risk of thromboembolism is increased during, immediately following, and for several weeks to months after atrial fibrillation ablation.36,37
During the procedure, the risk is related to transseptal sheath placement, electrode catheters in the left atrium, and char formation on ablation catheters. These risks are mitigated with proper and careful sheath and catheter manipulation, maintenance of bubble-free irrigation through lines and sheaths, use of irrigated catheters, and initiation of heparin before transseptal access. Heparin is also infused during the procedure, with close monitoring of activated clotting time.
Postprocedurally, the transiently increased clotting risk could be due to damaged endothelium from the ablation itself and stunning of atrial tissue, which results in impaired contraction. Damaged endothelium improves as the tissue heals, and the stunning resolves by electrical reverse remodeling with sinus rhythm maintenance.
In view of these risks, the referring physician and electrophysiologist must pay careful attention to anticoagulation before and after ablation.
Before the procedure. It is safe to continue anticoagulation uninterrupted through the procedure.38,39 If the patient is on warfarin, we want the international normalized ratio to be in the therapeutic range when we perform atrial fibrillation ablation, and the patient takes his or her usual dose on the day of the procedure. If taking a direct oral anticoagulant, patients typically skip a dose the day before ablation and again on the morning of the procedure, and resume taking it immediately afterward while in the anesthesia recovery room.
During the procedure, we start heparin before transseptal puncture, adjust it to achieve an activated clotting time of 300 to 400 seconds, and keep it in this range as long as there are sheaths or catheters in the left atrium.
After the procedure. The current guidelines24 recommend that oral anticoagulation be continued without interruption for at least 2 months after the procedure, and in most cases indefinitely, depending on age and comorbidities. The decision to stop anticoagulation after 2 months is typically based on the stroke risk as assessed by the CHA2DS2-VASc score (www.chadsvasc.org) and not on the success of the ablation procedure.
ANTIARRHYTHMIC DRUGS AFTER THE PROCEDURE
Some patients actually experience more atrial fibrillation in the first weeks to months after the procedure. The mechanism in this setting may be different from that causing the arrhythmia in the first place. The causes of early recurrence of atrial arrhythmias include postablation inflammation, temporary autonomic imbalance, and delay of atrial radiofrequency lesion formation.40,41 These arrhythmias may completely resolve as the ablation lesions heal and scars mature.
It has been hypothesized that short-term use of antiarrhythmic drugs after atrial fibrillation ablation is effective in preventing arrhythmias because it alters atrial electrophysiologic characteristics induced by the above transient factors. A recent systematic review of 6 clinical trials showed that short-term use of antiarrhythmic drugs reduces the risk of early arrhythmia recurrence but does not reduce recurrence in the long term.42
In terms of outcomes, any arrhythmias that occur in the first 3 months do not necessarily affect long-term success. This is referred to as the “blanking period.” However, generally speaking, it is preferable to maintain sinus rhythm during that time to avoid further anatomic or electrical left atrial adverse remodeling. In many situations, patients continue taking the same antiarrhythmic agent or start on antiarrhythmic therapy in the first few months after ablation.43,44
The mechanisms of late recurrence of atrial arrhythmias after ablation are thought to be different from those in early recurrence. Late recurrence has been ascribed to incomplete pulmonary vein isolation, recovery of pulmonary vein-left atrium connections, or recovery of any other lines of ablation created in the procedure.45,46 For late recurrence of atrial arrhythmia, studies and guidelines suggest that repeat ablation may be an option.24,47
PRACTICAL CONSIDERATIONS FOR PROCEDURAL PLANNING
Before the procedure, some electrophysiologists use cardiac computed tomography or magnetic resonance imaging to evaluate the pulmonary vein anatomy. This helps in planning and in selecting the appropriate tools for the procedure.
The patient is asked to fast on the day of the procedure. The procedure can take 3 to 6 hours, depending on the patient’s anatomy and the operator’s technique and experience. It can be performed with the patient under general anesthesia or conscious sedation. Currently, we use general anesthesia most of the time to maximize patient comfort.
After the procedure, our patients must stay in bed for 4 hours and stay overnight for observation. If no complications arise, they are discharged the next day.
- Go AS. The epidemiology of atrial fibrillation in elderly persons: the tip of the iceberg. Am J Geriatr Cardiol 2005; 14(2):56–61. pmid:15785146
- Go AS, Hylek EM, Phillips KA, et al. Prevalence of diagnosed atrial fibrillation in adults: national implications for rhythm management and stroke prevention: the AnTicoagulation and Risk Factors in Atrial Fibrillation (ATRIA) Study. JAMA 2001; 285(18):2370–2375. pmid:11343485
- Miyasaka Y, Barnes ME, Gersh BJ, et al. Secular trends in incidence of atrial fibrillation in Olmsted County, Minnesota, 1980 to 2000, and implications on the projections for future prevalence. Circulation 2006; 114(2):119–125. doi:10.1161/CIRCULATIONAHA.105.595140
- Piccini JP, Hammill BG, Sinner MF, et al. Incidence and prevalence of atrial fibrillation and associated mortality among Medicare beneficiaries, 1993–2007. Circ Cardiovasc Qual Outcomes 2012; 5(1):85–93. doi:10.1161/CIRCOUTCOMES.111.962688
- Moe GK, Rheinboldt WC, Abildskov JA. A computer model of atrial fibrillation. Am Heart J 1964; 67:200–220. pmid:14118488
- Cox JL, Schuessler RB, Boineau JP. The surgical treatment of atrial fibrillation. I. Summary of the current concepts of the mechanisms of atrial flutter and atrial fibrillation. J Thorac Cardiovasc Surg 1991; 101(3):402–405. pmid:1999933
- Cox JL, Schuessler RB, D’Agostino HJ Jr, et al. The surgical treatment of atrial fibrillation. III. Development of a definitive surgical procedure. J Thorac Cardiovasc Surg 1991; 101(4):569–583. pmid:2008095
- Jaïs P, Haïssaguerre M, Shah DC, et al. A focal source of atrial fibrillation treated by discrete radiofrequency ablation. Circulation 1997; 95(3):572–576. pmid:9024141
- Haïssaguerre M, Jaïs P, Shah DC, et al. Spontaneous initiation of atrial fibrillation by ectopic beats originating in the pulmonary veins. N Engl J Med 1998; 339(10):659–666. doi:10.1056/NEJM199809033391003
- Davies MJ, Pomerance A. Pathology of atrial fibrillation in man. Br Heart J 1972; 34(5):520–525. pmid:5031645
- Wijffels MC, Kirchhof CJ, Dorland R, Allessie MA. Atrial fibrillation begets atrial fibrillation. A study in awake chronically instrumented goats. Circulation 1995; 92(7):1954–1968. pmid:7671380
- Nattel S. New ideas about atrial fibrillation 50 years on. Nature 2002; 415(6868):219–226. doi:10.1038/415219a
- Medi C, Sparks PB, Morton JB, et al. Pulmonary vein antral isolation for paroxysmal atrial fibrillation: results from long-term follow-up. J Cardiovasc Electrophysiol 2011; 22(2):137–141. doi:10.1111/j.1540-8167.2010.01885.x
- Hussein AA, Saliba WI, Martin DO, et al. Natural history and long-term outcomes of ablated atrial fibrillation. Circ Arrhythm Electrophysiol 2011; 4(3):271–278. doi:10.1161/CIRCEP.111.962100
- Hussein AA, Saliba WI, Barakat A, et al. Radiofrequency ablation of persistent atrial fibrillation: diagnosis-to-ablation time, markers of pathways of atrial remodeling, and outcomes. Circ Arrhythm Electrophysiol 2016; 9(1):e003669. doi:10.1161/CIRCEP.115.003669
- Carlsson J, Miketic S, Windeler J, et al. Randomized trial of rate-control versus rhythm-control in persistent atrial fibrillation: the Strategies of Treatment of Atrial Fibrillation (STAF) study. J Am Coll Cardiol 2003; 41(10):1690–1696. pmid:12767648
- Van Gelder IC, Hagens VE, Bosker HA, et al; Rate Control versus Electrical Cardioversion for Persistent Atrial Fibrillation Study Group. A comparison of rate control and rhythm control in patients with recurrent persistent atrial fibrillation. N Engl J Med 2002; 347(23):1834–1840. doi:10.1056/NEJMoa021375
- Wyse DG, Waldo AL, DiMarco JP, et al; Atrial Fibrillation Follow-up Investigation of Rhythm Management (AFFIRM) Investigators. A comparison of rate control and rhythm control in patients with atrial fibrillation. N Engl J Med 2002; 347(23):1825–1833. doi:10.1056/NEJMoa021328
- Hagens VE, Crijns HJ, Van Veldhuisen DJ, et al; RAte Control versus Electrical cardioversion for persistent atrial fibrillation study group. Rate control versus rhythm control for patients with persistent atrial fibrillation with mild to moderate heart failure: results from the RAte Control versus Electrical cardioversion (RACE) study. Am Heart J 2005; 149(6):1106–111. doi:10.1016/j.ahj.2004.11.030
- Pedersen OD, Bagger H, Keller N, Marchant B, Køber L, Torp-Pedersen C. Efficacy of dofetilide in the treatment of atrial fibrillation-flutter in patients with reduced left ventricular function: a Danish investigations of arrhythmia and mortality on dofetilide (diamond) substudy. Circulation 2001; 104(3):292–296. pmid:11457747
- Guiot A, Jongnarangsin K, Chugh A, et al. Anticoagulant therapy and risk of cerebrovascular events after catheter ablation of atrial fibrillation in the elderly. J Cardiovasc Electrophysiol 2012; 23(1):36–43. doi:10.1111/j.1540-8167.2011.02141.x
- Oral H, Chugh A, Ozaydin M, et al. Risk of thromboembolic events after percutaneous left atrial radiofrequency ablation of atrial fibrillation. Circulation 2006; 114(8):759–765. doi:10.1161/CIRCULATIONAHA.106.641225
- Themistoclakis S, Corrado A, Marchlinski FE, et al. The risk of thromboembolism and need for oral anticoagulation after successful atrial fibrillation ablation. J Am Coll Cardiol 2010; 55(8):735–743. doi:10.1016/j.jacc.2009.11.039
- Calkins H, Hindricks G, Cappato R, et al. 2017 HRS/EHRA/ECAS/APHRS/SOLAECE expert consensus statement on catheter and surgical ablation of atrial fibrillation: executive summary. J Arrhythm 2017; 33(5):369–409. doi:10.1016/j.joa.2017.08.001
- Kuck KH, Brugada J, Fürnkranz A, et al; FIRE AND ICE Investigators. Cryoballoon or radiofrequency ablation for paroxysmal atrial fibrillation. N Engl J Med 2016; 374(23):2235–2245. doi:10.1056/NEJMoa1602014
- Reddy VY, Dukkipati SR, Neuzil P, et al. Randomized, controlled trial of the safety and effectiveness of a contact force-sensing irrigated catheter for ablation of paroxysmal atrial fibrillation: results of the TactiCath Contact Force Ablation Catheter Study for Atrial Fibrillation (TOCCASTAR) Study. Circulation 2015; 132(10):907–915. doi:10.1161/CIRCULATIONAHA.114.014092
- Natale A, Reddy VY, Monir G, et al. Paroxysmal AF catheter ablation with a contact force sensing catheter: results of the prospective, multicenter SMART-AF trial. J Am Coll Cardiol 2014; 64(7):647–656. doi:10.1016/j.jacc.2014.04.072
- Hussein AA, Barakat AF, Saliba WI, et al. Persistent atrial fibrillation ablation with or without contact force sensing. J Cardiovasc Electrophysiol 2017; 28(5):483–488. doi:10.1111/jce.13179
- Deshmukh A, Patel NJ, Pant I, et al. In-hospital complications associated with catheter ablation of atrial fibrillation in the United States between 2000 and 2010: analysis of 93,801 procedures. Circulation 2013; 128(19):2104–2112. doi:10.1161/CIRCULATIONAHA.113.003862
- Cappato R, Calkins H, Chen SA, et al. Worldwide survey on the methods, efficacy, and safety of catheter ablation for human atrial fibrillation. Circulation 2005; 111(9):1100–1105. doi:10.1161/01.CIR.0000157153.30978.67
- Cappato R, Calkins H, Chen SA, et al. Updated worldwide survey on the methods, efficacy, and safety of catheter ablation for human atrial fibrillation. Circ Arrhythm Electrophysiol 2010; 3(1):32–38. doi:10.1161/CIRCEP.109.859116
- Wazni OM, Marrouche NF, Martin DO, et al. Radiofrequency ablation vs antiarrhythmic drugs as first-line treatment of symptomatic atrial fibrillation: a randomized trial. JAMA 2005; 293(21):2634–2640. doi:10.1001/jama.293.21.2634
- Jaïs P, Cauchemez B, Macle L, et al. Catheter ablation versus antiarrhythmic drugs for atrial fibrillation: the A4 study. Circulation 2008; 118(24):2498–2505. doi:10.1161/CIRCULATIONAHA.108.772582
- Brooks AG, Stiles MK, Laborderie J, et al. Outcomes of long-standing persistent atrial fibrillation ablation: a systematic review. Heart Rhythm 2010; 7(6):835–846. doi:10.1016/j.hrthm.2010.01.017
- Packer DL, Lee KL, Mark DB, Robb RA. Catheter Ablation versus Antiarrhythmic Drug Therapy for Atrial Fibrillation Trial, CABANA. http://cabanatrial.org/. Accessed September 10, 2018.
- Scherr D, Sharma K, Dalal D, et al. Incidence and predictors of periprocedural cerebrovascular accident in patients undergoing catheter ablation of atrial fibrillation. J Cardiovasc Electrophysiol 2009; 20(12):1357–1363. doi:10.1111/j.1540-8167.2009.01540.x
- Wazni OM, Rossillo A, Marrouche NF, et al. Embolic events and char formation during pulmonary vein isolation in patients with atrial fibrillation: impact of different anticoagulation regimens and importance of intracardiac echo imaging. J Cardiovasc Electrophysiol 2005; 16(6):576–581. doi:10.1111/j.1540-8167.2005.40480.x
- Hussein AA, Martin DO, Saliba W, et al. Radiofrequency ablation of atrial fibrillation under therapeutic international normalized ratio: a safe and efficacious periprocedural anticoagulation strategy. Heart Rhythm 2009; 6(10):1425–1429. doi:10.1016/j.hrthm.2009.07.007
- Bassiouny M, Saliba W, Rickard J, et al. Use of dabigatran for periprocedural anticoagulation in patients undergoing catheter ablation for atrial fibrillation. Circ Arrhythm Electrophysiol 2013; 6(3):460–466. doi:10.1161/CIRCEP.113.000320
- Koyama T, Tada H, Sekiguchi Y, et al. Prevention of atrial fibrillation recurrence with corticosteroids after radiofrequency catheter ablation: a randomized controlled trial. J Am Coll Cardiol 2010; 56(18):1463–1472. doi:10.1016/j.jacc.2010.04.057
- Oral H, Knight BP, Ozaydin M, et al. Clinical significance of early recurrences of atrial fibrillation after pulmonary vein isolation. J Am Coll Cardiol 2002; 40(1):100–104. pmid:12103262
- Chen W, Liu H, Ling Z, et al. Efficacy of short-term antiarrhythmic drugs use after catheter ablation of atrial fibrillation—a systematic review with meta-analyses and trial sequential analyses of randomized controlled trials. PLoS One 2016; 11(5):e0156121. doi:10.1371/journal.pone.0156121
- Leong-Sit P, Roux JF, Zado E, et al. Antiarrhythmics after ablation of atrial fibrillation (5A Study): six-month follow-up study. Circ Arrhythm Electrophysiol 2011; 4(1):11–14. doi:10.1161/CIRCEP.110.955393
- Roux JF, Zado E, Callans DJ, et al. Antiarrhythmics after ablation of atrial fibrillation (5A Study). Circulation 2009; 120(12):1036–1040. doi:10.1161/CIRCULATIONAHA.108.839639
- Sotomi Y, Inoue K, Ito N, et al. Cause of very late recurrence of atrial fibrillation or flutter after catheter ablation for atrial fibrillation. Am J Cardiol 2013; 111(4):552–556. doi:10.1016/j.amjcard.2012.10.040
- Lee SH, Tai CT, Hsieh MH, et al. Predictors of early and late recurrence of atrial fibrillation after catheter ablation of paroxysmal atrial fibrillation. J Interv Card Electrophysiol. 2004 Jun;10(3):221-6. doi:10.1023/B:JICE.0000026915.02503.92
- Zhang XD, Gu J, Jiang WF, et al. Optimal rhythm-control strategy for recurrent atrial tachycardia after catheter ablation of persistent atrial fibrillation: a randomized clinical trial. Eur Heart J 2014; 35(20):1327–1334. doi:10.1093/eurheartj/ehu017
- Go AS. The epidemiology of atrial fibrillation in elderly persons: the tip of the iceberg. Am J Geriatr Cardiol 2005; 14(2):56–61. pmid:15785146
- Go AS, Hylek EM, Phillips KA, et al. Prevalence of diagnosed atrial fibrillation in adults: national implications for rhythm management and stroke prevention: the AnTicoagulation and Risk Factors in Atrial Fibrillation (ATRIA) Study. JAMA 2001; 285(18):2370–2375. pmid:11343485
- Miyasaka Y, Barnes ME, Gersh BJ, et al. Secular trends in incidence of atrial fibrillation in Olmsted County, Minnesota, 1980 to 2000, and implications on the projections for future prevalence. Circulation 2006; 114(2):119–125. doi:10.1161/CIRCULATIONAHA.105.595140
- Piccini JP, Hammill BG, Sinner MF, et al. Incidence and prevalence of atrial fibrillation and associated mortality among Medicare beneficiaries, 1993–2007. Circ Cardiovasc Qual Outcomes 2012; 5(1):85–93. doi:10.1161/CIRCOUTCOMES.111.962688
- Moe GK, Rheinboldt WC, Abildskov JA. A computer model of atrial fibrillation. Am Heart J 1964; 67:200–220. pmid:14118488
- Cox JL, Schuessler RB, Boineau JP. The surgical treatment of atrial fibrillation. I. Summary of the current concepts of the mechanisms of atrial flutter and atrial fibrillation. J Thorac Cardiovasc Surg 1991; 101(3):402–405. pmid:1999933
- Cox JL, Schuessler RB, D’Agostino HJ Jr, et al. The surgical treatment of atrial fibrillation. III. Development of a definitive surgical procedure. J Thorac Cardiovasc Surg 1991; 101(4):569–583. pmid:2008095
- Jaïs P, Haïssaguerre M, Shah DC, et al. A focal source of atrial fibrillation treated by discrete radiofrequency ablation. Circulation 1997; 95(3):572–576. pmid:9024141
- Haïssaguerre M, Jaïs P, Shah DC, et al. Spontaneous initiation of atrial fibrillation by ectopic beats originating in the pulmonary veins. N Engl J Med 1998; 339(10):659–666. doi:10.1056/NEJM199809033391003
- Davies MJ, Pomerance A. Pathology of atrial fibrillation in man. Br Heart J 1972; 34(5):520–525. pmid:5031645
- Wijffels MC, Kirchhof CJ, Dorland R, Allessie MA. Atrial fibrillation begets atrial fibrillation. A study in awake chronically instrumented goats. Circulation 1995; 92(7):1954–1968. pmid:7671380
- Nattel S. New ideas about atrial fibrillation 50 years on. Nature 2002; 415(6868):219–226. doi:10.1038/415219a
- Medi C, Sparks PB, Morton JB, et al. Pulmonary vein antral isolation for paroxysmal atrial fibrillation: results from long-term follow-up. J Cardiovasc Electrophysiol 2011; 22(2):137–141. doi:10.1111/j.1540-8167.2010.01885.x
- Hussein AA, Saliba WI, Martin DO, et al. Natural history and long-term outcomes of ablated atrial fibrillation. Circ Arrhythm Electrophysiol 2011; 4(3):271–278. doi:10.1161/CIRCEP.111.962100
- Hussein AA, Saliba WI, Barakat A, et al. Radiofrequency ablation of persistent atrial fibrillation: diagnosis-to-ablation time, markers of pathways of atrial remodeling, and outcomes. Circ Arrhythm Electrophysiol 2016; 9(1):e003669. doi:10.1161/CIRCEP.115.003669
- Carlsson J, Miketic S, Windeler J, et al. Randomized trial of rate-control versus rhythm-control in persistent atrial fibrillation: the Strategies of Treatment of Atrial Fibrillation (STAF) study. J Am Coll Cardiol 2003; 41(10):1690–1696. pmid:12767648
- Van Gelder IC, Hagens VE, Bosker HA, et al; Rate Control versus Electrical Cardioversion for Persistent Atrial Fibrillation Study Group. A comparison of rate control and rhythm control in patients with recurrent persistent atrial fibrillation. N Engl J Med 2002; 347(23):1834–1840. doi:10.1056/NEJMoa021375
- Wyse DG, Waldo AL, DiMarco JP, et al; Atrial Fibrillation Follow-up Investigation of Rhythm Management (AFFIRM) Investigators. A comparison of rate control and rhythm control in patients with atrial fibrillation. N Engl J Med 2002; 347(23):1825–1833. doi:10.1056/NEJMoa021328
- Hagens VE, Crijns HJ, Van Veldhuisen DJ, et al; RAte Control versus Electrical cardioversion for persistent atrial fibrillation study group. Rate control versus rhythm control for patients with persistent atrial fibrillation with mild to moderate heart failure: results from the RAte Control versus Electrical cardioversion (RACE) study. Am Heart J 2005; 149(6):1106–111. doi:10.1016/j.ahj.2004.11.030
- Pedersen OD, Bagger H, Keller N, Marchant B, Køber L, Torp-Pedersen C. Efficacy of dofetilide in the treatment of atrial fibrillation-flutter in patients with reduced left ventricular function: a Danish investigations of arrhythmia and mortality on dofetilide (diamond) substudy. Circulation 2001; 104(3):292–296. pmid:11457747
- Guiot A, Jongnarangsin K, Chugh A, et al. Anticoagulant therapy and risk of cerebrovascular events after catheter ablation of atrial fibrillation in the elderly. J Cardiovasc Electrophysiol 2012; 23(1):36–43. doi:10.1111/j.1540-8167.2011.02141.x
- Oral H, Chugh A, Ozaydin M, et al. Risk of thromboembolic events after percutaneous left atrial radiofrequency ablation of atrial fibrillation. Circulation 2006; 114(8):759–765. doi:10.1161/CIRCULATIONAHA.106.641225
- Themistoclakis S, Corrado A, Marchlinski FE, et al. The risk of thromboembolism and need for oral anticoagulation after successful atrial fibrillation ablation. J Am Coll Cardiol 2010; 55(8):735–743. doi:10.1016/j.jacc.2009.11.039
- Calkins H, Hindricks G, Cappato R, et al. 2017 HRS/EHRA/ECAS/APHRS/SOLAECE expert consensus statement on catheter and surgical ablation of atrial fibrillation: executive summary. J Arrhythm 2017; 33(5):369–409. doi:10.1016/j.joa.2017.08.001
- Kuck KH, Brugada J, Fürnkranz A, et al; FIRE AND ICE Investigators. Cryoballoon or radiofrequency ablation for paroxysmal atrial fibrillation. N Engl J Med 2016; 374(23):2235–2245. doi:10.1056/NEJMoa1602014
- Reddy VY, Dukkipati SR, Neuzil P, et al. Randomized, controlled trial of the safety and effectiveness of a contact force-sensing irrigated catheter for ablation of paroxysmal atrial fibrillation: results of the TactiCath Contact Force Ablation Catheter Study for Atrial Fibrillation (TOCCASTAR) Study. Circulation 2015; 132(10):907–915. doi:10.1161/CIRCULATIONAHA.114.014092
- Natale A, Reddy VY, Monir G, et al. Paroxysmal AF catheter ablation with a contact force sensing catheter: results of the prospective, multicenter SMART-AF trial. J Am Coll Cardiol 2014; 64(7):647–656. doi:10.1016/j.jacc.2014.04.072
- Hussein AA, Barakat AF, Saliba WI, et al. Persistent atrial fibrillation ablation with or without contact force sensing. J Cardiovasc Electrophysiol 2017; 28(5):483–488. doi:10.1111/jce.13179
- Deshmukh A, Patel NJ, Pant I, et al. In-hospital complications associated with catheter ablation of atrial fibrillation in the United States between 2000 and 2010: analysis of 93,801 procedures. Circulation 2013; 128(19):2104–2112. doi:10.1161/CIRCULATIONAHA.113.003862
- Cappato R, Calkins H, Chen SA, et al. Worldwide survey on the methods, efficacy, and safety of catheter ablation for human atrial fibrillation. Circulation 2005; 111(9):1100–1105. doi:10.1161/01.CIR.0000157153.30978.67
- Cappato R, Calkins H, Chen SA, et al. Updated worldwide survey on the methods, efficacy, and safety of catheter ablation for human atrial fibrillation. Circ Arrhythm Electrophysiol 2010; 3(1):32–38. doi:10.1161/CIRCEP.109.859116
- Wazni OM, Marrouche NF, Martin DO, et al. Radiofrequency ablation vs antiarrhythmic drugs as first-line treatment of symptomatic atrial fibrillation: a randomized trial. JAMA 2005; 293(21):2634–2640. doi:10.1001/jama.293.21.2634
- Jaïs P, Cauchemez B, Macle L, et al. Catheter ablation versus antiarrhythmic drugs for atrial fibrillation: the A4 study. Circulation 2008; 118(24):2498–2505. doi:10.1161/CIRCULATIONAHA.108.772582
- Brooks AG, Stiles MK, Laborderie J, et al. Outcomes of long-standing persistent atrial fibrillation ablation: a systematic review. Heart Rhythm 2010; 7(6):835–846. doi:10.1016/j.hrthm.2010.01.017
- Packer DL, Lee KL, Mark DB, Robb RA. Catheter Ablation versus Antiarrhythmic Drug Therapy for Atrial Fibrillation Trial, CABANA. http://cabanatrial.org/. Accessed September 10, 2018.
- Scherr D, Sharma K, Dalal D, et al. Incidence and predictors of periprocedural cerebrovascular accident in patients undergoing catheter ablation of atrial fibrillation. J Cardiovasc Electrophysiol 2009; 20(12):1357–1363. doi:10.1111/j.1540-8167.2009.01540.x
- Wazni OM, Rossillo A, Marrouche NF, et al. Embolic events and char formation during pulmonary vein isolation in patients with atrial fibrillation: impact of different anticoagulation regimens and importance of intracardiac echo imaging. J Cardiovasc Electrophysiol 2005; 16(6):576–581. doi:10.1111/j.1540-8167.2005.40480.x
- Hussein AA, Martin DO, Saliba W, et al. Radiofrequency ablation of atrial fibrillation under therapeutic international normalized ratio: a safe and efficacious periprocedural anticoagulation strategy. Heart Rhythm 2009; 6(10):1425–1429. doi:10.1016/j.hrthm.2009.07.007
- Bassiouny M, Saliba W, Rickard J, et al. Use of dabigatran for periprocedural anticoagulation in patients undergoing catheter ablation for atrial fibrillation. Circ Arrhythm Electrophysiol 2013; 6(3):460–466. doi:10.1161/CIRCEP.113.000320
- Koyama T, Tada H, Sekiguchi Y, et al. Prevention of atrial fibrillation recurrence with corticosteroids after radiofrequency catheter ablation: a randomized controlled trial. J Am Coll Cardiol 2010; 56(18):1463–1472. doi:10.1016/j.jacc.2010.04.057
- Oral H, Knight BP, Ozaydin M, et al. Clinical significance of early recurrences of atrial fibrillation after pulmonary vein isolation. J Am Coll Cardiol 2002; 40(1):100–104. pmid:12103262
- Chen W, Liu H, Ling Z, et al. Efficacy of short-term antiarrhythmic drugs use after catheter ablation of atrial fibrillation—a systematic review with meta-analyses and trial sequential analyses of randomized controlled trials. PLoS One 2016; 11(5):e0156121. doi:10.1371/journal.pone.0156121
- Leong-Sit P, Roux JF, Zado E, et al. Antiarrhythmics after ablation of atrial fibrillation (5A Study): six-month follow-up study. Circ Arrhythm Electrophysiol 2011; 4(1):11–14. doi:10.1161/CIRCEP.110.955393
- Roux JF, Zado E, Callans DJ, et al. Antiarrhythmics after ablation of atrial fibrillation (5A Study). Circulation 2009; 120(12):1036–1040. doi:10.1161/CIRCULATIONAHA.108.839639
- Sotomi Y, Inoue K, Ito N, et al. Cause of very late recurrence of atrial fibrillation or flutter after catheter ablation for atrial fibrillation. Am J Cardiol 2013; 111(4):552–556. doi:10.1016/j.amjcard.2012.10.040
- Lee SH, Tai CT, Hsieh MH, et al. Predictors of early and late recurrence of atrial fibrillation after catheter ablation of paroxysmal atrial fibrillation. J Interv Card Electrophysiol. 2004 Jun;10(3):221-6. doi:10.1023/B:JICE.0000026915.02503.92
- Zhang XD, Gu J, Jiang WF, et al. Optimal rhythm-control strategy for recurrent atrial tachycardia after catheter ablation of persistent atrial fibrillation: a randomized clinical trial. Eur Heart J 2014; 35(20):1327–1334. doi:10.1093/eurheartj/ehu017
KEY POINTS
- Atrial fibrillation is increasing in prevalence with the aging of the US population and is associated with worsening quality of life and increased risk of stroke, heart failure, and death.
- Atrial fibrillation results in adverse atrial remodeling and fibrosis, eventually leading to persistence of the arrhythmia and making rhythm control difficult.
- Catheter ablation has evolved to be a safe procedure with technologic advancements, especially in experienced tertiary care centers.
- The primary aim of atrial fibrillation ablation is to reduce symptoms and improve quality of life. In theory, it could also decrease the risk of stroke, heart failure, and death, but these outcomes have not been systematically evaluated in a large randomized controlled trial.