User login
Reproductive planning for women after solid-organ transplant
Increasing numbers of women of childbearing age are receiving solid-organ transplants. All need counseling on how to prevent pregnancy while they are taking immunosuppressive agents. Some want to become pregnant after their transplant and thus require counseling and follow-up to maintain good health during pregnancy (Table 1).1
Primary care physicians can assist with basic contraception counseling and pregnancy planning for their patients who have had solid-organ transplants. In this review, we describe contraceptive options and pregnancy planning for these women.
TRANSPLANTS IN WOMEN ARE INCREASING
Over the past 20 years, the number of solid-organ transplants in US women has increased steadily. Since 1988, 38% of the 634,000 transplants performed were in women, and 47% of these women were of childbearing age (ages 18 to 49).2 Kidneys accounted for 60% of solid-organ transplants,2 and kidney transplant is now commonly performed in women of childbearing age. In 2012, of 176,000 patients with a functioning renal graft, 40.5% were women, and recipients between ages 20 and 44 composed the second-largest age group.3
FERTILITY IN WOMEN WITH END-STAGE RENAL DISEASE
Women in their reproductive years who have end-stage renal disease have lower fertility rates than women in the general population. In women undergoing peritoneal dialysis or hemodialysis, conception rates decrease to around 0.5% per year.4 This lower rate is most likely related to hypothalamic-pituitary-gonadal dysfunction, leading to reduced or total impairment of ovulation, menstrual irregularities, and infertility.5
Fertility often returns within a few months after transplant,1,6 and reported posttransplant pregnancy rates range from 3.3% to 18%,7–9 with up to one-third of pregnancies being unintended.6,10 These numbers are likely an underestimate because they do not reflect all pregnancies that are terminated, as many women do not voluntarily report having had an abortion.
Fertility is also severely diminished in women with end-stage liver disease. After liver transplant, sex hormone levels return to normal for many women, and menses soon resume.11
In 2005, the National Transplantation Pregnancy Registry reported 1,418 pregnancies in 919 female recipients of solid-organ transplants. In 2010, this number had increased to 1,940 pregnancies in 1,185 recipients, of whom 75% were kidney transplant recipients.12
A successful pregnancy outcome is most likely when a minimum of 1 year intervenes between transplant and conception.12,13
TERATOGENICITY OF IMMUNOSUPPRESSANTS
Immunosuppressant drugs commonly used for maintenance therapy after solid-organ transplant include the following:
- Calcineurin inhibitors (eg, cyclosporine, tacrolimus)
- Antiproliferative and antimetabolite agents (eg, mycophenolate mofetil, azathioprine)
- Corticosteroids
- Mammalian target of rapamycin inhibitors (eg, sirolimus, everolimus)
- T-cell costimulation blockers (eg, belatacept).14
The US Food and Drug Administration (FDA) previously classified mycophenolate mofetil and azathioprine in pregnancy risk category D (positive evidence of human fetal risk). The teratogenic risk of mycophenolate mofetil is well established in studies documenting specific congenital malformations and fetal loss in the first trimester.13,15 The teratogenic risk of azathioprine, on the other hand, is estimated to be minimal to small.16 Many of the associated fetal abnormalities may be related to the complexity of the underlying medical condition of the mother rather than to the medication.16
In June 2015, the FDA’s new Pregnancy and Lactation Labeling Rule went into effect, which removes the pregnancy letter categories A, B, C, D, and X from labeling.17 This rule was designed to help providers counsel their patients regarding the specific risks and benefits of a drug when used by pregnant or nursing women. However, the ABCDX categories are still commonly used. Table 2 shows information about the risks during pregnancy and lactation posed by the immunosuppressive drugs commonly used by posttransplant patients.18
CRITERIA FOR A SUCCESSFUL PREGNANCY
To ensure a safe and successful pregnancy with the fewest fetal and maternal complications, women are generally advised to avoid pregnancy for at least 1 year after transplant.19,20
In addition, women should meet certain clinical prerequisites after transplant before they conceive, as outlined by the American Society of Transplantation.19,20 These include:
- No rejection within the previous year
- Adequate and stable graft function (eg, serum creatinine < 1.5 mg/dL and urinary protein excretion < 500 mg/24 hours)
- No acute infection that might affect the fetus
- Maintenance immunosuppression at stable dosages.
Other circumstances to consider include episodes of rejection in the first year after transplant (as evidenced by biopsy results or glomerular filtration rate), the woman’s age (advanced maternal age is unfavorable), or any history of noncompliance.
Every pregnancy in a transplant recipient must be carefully planned. Primary care providers should encourage patients to meet with their transplant team and obstetricians early and often to allow time for the care team to adjust the type and dosing of immunosuppressant drugs, to ensure stable graft function, and to optimize any current chronic medical conditions such as diabetes mellitus or hypertension before conception.
CONTRACEPTIVE COUNSELING AFTER TRANSPLANT
Pregnancy should be avoided while transplant patients are taking FDA category D immunosuppressant drugs and, as already mentioned, during the first year after transplant. Unintended pregnancy can have serious health consequences for the mother and the fetus, as well as poor pregnancy outcomes. The US Centers for Disease Control and Prevention (CDC) lists solid-organ transplant within the past 2 years as a condition that can lead to adverse events as a result of pregnancy.21 After a transplant, a woman’s risks from an unintended pregnancy are always greater than the risks from any contraceptive, and this is important to reinforce in counseling.
Two forms of reliable contraception should be used at all times, and consistent condom use should be encouraged as one of the methods. Condoms are not reliable when used as the sole contraceptive method because they have an 18% typical-use failure rate. However, they are an excellent adjunct to other contraceptive methods because they have the additional benefit of protecting against sexually transmitted disease.
Choosing the appropriate contraceptive method for recipients of solid-organ transplants can be challenging because of several factors, including the recipient’s preexisting medical problems and drug interactions of immunosuppressant medications.
CDC criteria and categories for contraceptive use
In 2010, the CDC released the US version of the Medical Eligibility Criteria (US MEC) for contraceptive use, which was based on the 2009 World Health Organization Medical Eligibility Criteria (WHO MEC); these criteria were revised in August 2016.21
- Category 1: A condition for which there is no restriction for the use of the contraceptive method
- Category 2: A condition for which the advantages of using the method generally outweigh the theoretical or proven risks
- Category 3: A condition for which the theoretical or proven risks usually outweigh the advantages of using the method
- Category 4: A condition that represents an unacceptable health risk if the contraceptive method is used.
These recommendations aimed to improve family planning options by clarifying the possible safe and effective contraceptive options available while considering the patient’s medical condition. The CDC added solid-organ transplant recipients to this document because of the prevalence of this group in the United States.
The CDC categorizes a patient’s medical condition after transplant as either complicated or uncomplicated. Complicated conditions include acute or chronic graft failure, graft rejection, and cardiac allograft vasculopathy.21
Effectiveness of contraceptive methods
Contraceptive methods can be divided into 4 categories based on estimated effectiveness, ie, the pregnancy rate with “typical use” of that particular method in 1 year21–23:
- Very effective (0%–0.9%)
- Effective (1%–9%)
- Moderately effective (10%–25%)
- Less effective (26%–32%).
Typical use refers to failure rates for women and men whose use is not consistent nor always correct. Correct use, also described in the sections that follow, refers to failure rates for those whose use is consistent and always correct.
Women should be counseled regarding all available contraceptive options that are medically suitable for them, so they can choose the method that best fits their needs and lifestyle. They should receive counseling on emergency contraception, barrier protection against sexually transmitted disease, and the correct use of the contraceptive method they choose. They should be advised that if their chosen contraceptive method is unsatisfactory for any reason, they can switch to another method. Most importantly, providers need to impress on their patients that the risks associated with unintended pregnancy are far greater than the risks from any of the contraceptive methods.
VERY EFFECTIVE CONTRACEPTIVES (UNINTENDED PREGNANCY RATE 0%–0.9%)
This tier of contraception is the most effective regardless of the patient’s adherence; it includes long-acting, reversible contraceptives and permanent sterilization (both male and female) (Table 3).21–23
Long-acting reversible contraceptives include intrauterine devices (IUDs) and the subdermal etonogestrel implant. Given their efficacy and favorable safety profile, long-acting reversible contraceptives are being promoted for use in women who have chronic medical conditions, such as transplants.24
Intrauterine devices
IUDs are long-acting and reversible. They can be used by women who are nulliparous and those of all ages, including adolescents.22
Two types of IUDs are available in the United States: nonhormonal (copper) and hormonal (levonorgestrel). The copper IUD is effective for at least 10 years, whereas the levonorgestrel IUDs last for 3 to 5 years.22
Four levonorgestrel IUDs are currently available in the United States. Their sizes and doses vary: Mirena (52 µg), Skyla (13.5 µg), Liletta (52 µg), and Kyleena (19.5 µg).
Fewer than 1% of women become pregnant in the first year of IUD use.22,23 IUDs are an ideal option for women with solid-organ transplants because they are so effective and because the patient does not have to do anything once the IUD is in.22–24 The levonorgestrel IUD Mirena has the additional advantage of reducing heavy menstrual bleeding and is currently the only hormonal IUD with FDA approval for the management of menorrhagia.
About 12% of women in the general population use IUDs as their contraceptive method of choice,25 whereas after solid-organ transplantation about 15% to 20% of women do.26
Two historic concerns regarding IUDs may explain their low rate of use in transplant recipients.
First, IUDs were believed to be less effective in women on immunosuppressive drugs because IUDs act by inducing a local inflammatory reaction. However, IUDs involve macrophage activation, which is independent of the immune processes modified by immunosuppressants (primarily T-cell function).27 A recent pilot study showed a strong inflammatory reaction in the endometrium of transplant recipients after levonorgestrel IUD insertion.28
Second, there was concern about the increased risk of pelvic inflammatory disease with IUDs, but studies have shown levonorgestrel IUDs to be safe in transplant patients.29,30
The CDC21 lists copper and levonorgestrel IUDs in MEC category 3 (the risks generally outweigh the advantages) for initiation in patients with complicated transplants and in category 2 (advantages generally outweigh the risks) in patients with uncomplicated organ transplants. The devices are in category 2 for both complicated and uncomplicated cases if the IUD is already in place.
Subdermal implant
A subdermal implant consisting of a single rod containing 68 mg of etonogestrel is commercially available in the United States. It is one of the most effective contraceptive methods, with the lowest rates of pregnancy—less than 1% per year, with protection lasting at least 3 years.22,23 This low risk makes the subdermal implant a suitable method of contraception after transplant. Daily compliance is not required, and there are no hepatic first-pass effects, which results in higher bioavailability and less chance of drug interactions.
The main disadvantage of the subdermal implant and IUDs is unscheduled bleeding. An important benefit is prolonged amenorrhea, not only for patient convenience, but for reduction of endometrial cancer risk. Insertion and removal of the implant are considered minor office procedures. The implants are classified as US MEC category 2 in uncomplicated cases; initiation in complicated cases is considered category 3 but continuation is considered category 2.21
Permanent sterilization
Permanent sterilization is another option for women and men. In women, the fallopian tubes can be occluded with a coil system implanted vaginally through a hysteroscope, or they can be severed, tied, or clamped in a laparoscopic procedure or during cesarean delivery. Pregnancy rates after tubal ligation are less than 1%,23,31 although concern exists for high failure rates with the hysteroscopic method.
Because younger patients are more likely than older patients to subsequently regret having the procedure done, all available contraceptive options should be discussed with them.31
For men, permanent sterilization is done by vasectomy, which has less associated risk and cost compared with sterilization for women.
EFFECTIVE CONTRACEPTIVE METHODS (UNINTENDED PREGNANCY RATE 1%–9%)
Effective contraceptive methods, the next tier down from very effective methods, include injectable contraceptives, combined hormonal contraceptives, and progestin-only contraceptives (Table 4).
Injectable contraceptives
Depot medroxyprogesterone acetate is an injectable progestin-only contraceptive that carries a pregnancy risk of 6% with typical use and less than 1% with correct use.23 Thus, some failures are due to patients not returning for follow-up, but in some patients this method is not effective. Injections are given intramuscularly once every 3 months, avoiding the need for daily use.
A valid concern for transplant patients is that medroxyprogesterone acetate reduces bone mineral density. Although the bone effects are reversible in healthy adult women, caution is needed when prescribing this option to transplant patients who are already at increased risk of bone disease attributable to renal osteodystrophy and chronic corticosteroid use. 32,33
Recently, a subcutaneous formulation of depot medroxyprogesterone acetate (104 mg)was added to the WHO MEC for contraceptive use.34,35 The recommendations for the subcutaneous form are similar to those for the intramuscular form. In healthy women, the subcutaneous formulation is as safe and effective as the intramuscular form,36 but its efficacy after solid-organ transplant has not been determined. Both forms of depot medroxyprogesterone acetate are category 2 in the US MEC for both complicated and uncomplicated transplant cases.21
Combined hormonal contraceptives
Combined hormonal contraceptives contain both estrogen and progesterone and are available as pills, patches, or rings. Each product has an unintended pregnancy risk of 9% with typical use and less than 1% with correct use.23 They require strict patient adherence to regular daily use, which likely explains their high failure rate with typical use.
Combined hormonal contraceptives reduce mortality risk in women in the general population,37 but their effect on mortality risk after transplant is unknown and needs further study. In women who received liver transplants, low-dose combined hormonal contraceptives have been found to be effective and well tolerated, but initiation should be delayed at least 6 months until postoperative organ stability is demonstrated.11
Combined oral contraceptives are the most widely prescribed because they are convenient and familiar and have an acceptable safety profile in transplant patients,11,33,37 despite their high failure rate with typical use. They regulate the menstrual cycle and reduce anemia associated with menstruation.
The transdermal contraceptive patch has a mechanism of action similar to that of the combined oral contraceptives, but it delivers estrogen and progesterone transdermally through the abdominal wall, thus avoiding first-pass metabolism in the liver and enzymatic degradation in the gut. It delivers 35 µg of ethinyl estradiol and 150 µg of norelgestromin (an active metabolite of norgestimate) daily.38 It may cause higher circulating levels of estrogen than a combined oral contraceptive and may be associated with a higher risk of venous thromboembolism, but the evidence is conflicting.39–42
The vaginal ring, made of Silastic, delivers ethinyl estradiol in a low dose (15 µg/day) and etonorgestrel 0.12 mg/day. Like the patch, it has the advantage of bypassing first-pass metabolism in the liver, making it a good option for transplant patients who are taking antirejection drugs, thus avoiding drug interactions.41
Both the transdermal patch and vaginal ring were studied in transplant patients and had favorable results.24,43 The combined hormonal oral contraceptive pills, patch, and ring are in category 4 (unacceptable health risk) in the US MEC in patients with complicated cases, but they are in category 2 in uncomplicated cases.21
Combined hormonal contraceptives should not be considered first-line options by themselves for transplant patients because of their high failure rate with typical use.24
Progestin-only pills
Although progestin-only pills have not been studied specifically in transplant patients, they can be considered for women who have contraindications to estrogen use. Estrogen use is contraindicated in women with a history of venous thromboembolism, thrombogenic mutations, estrogen-dependent neoplasia, hepatocellular adenoma, severe hypertension, vascular disease, and Budd-Chiari syndrome.
Progestin-only pills inhibit ovulation in only about half of a woman’s cycles, but they prevent conception by other mechanisms as well, such as causing thickening of the cervical mucus. They also alter the endometrium to make it unfavorable for implantation and reduce the ciliary activity of the fallopian tube.
Strict adherence is important for effectiveness because progestin-only pills have a shorter half-life than combined hormonal contraceptives and also suppress ovulation less effectively.22 Failure rates are similar or somewhat higher than with combined hormonal contraceptives; with typical use, about 9 in 100 women can become pregnant in the first year.23 According to the US MEC,21 progestin-only pills are classified as category 2 for patients after both complicated and uncomplicated transplants.
MODERATELY EFFECTIVE METHODS (PREGNANCY RATE 10%–25%)
This tier of contraceptives includes all barrier methods, ie, male and female condoms, vaginal diaphragms, cervical caps, and sponges (Table 5).
Condoms (male and female)
When male condoms are used as the only birth control method, pregnancy occurs less often (18% with typical use and 2% with correct use) than with female condoms (21% with typical use and 5% with correct use).23 Male and female condoms are the only contraceptive methods that also prevent transmission of sexually transmitted disease.24
Caps, sponges, diaphragms
Cervical caps, vaginal sponges, and vaginal diaphragms are other forms of barrier contraceptives. All barrier methods should be combined with another contraceptive method to provide reliable protection against pregnancy. These methods are considered category 1 according to the US MEC.
LESS-EFFECTIVE METHODS
Fertility awareness-based methods such as the rhythm method have an associated pregnancy rate of about 25% with typical use and 3% to 5% with correct use23 and cannot be relied on for use by transplant recipients.24
Withdrawal and spermicides are considered least effective and unreliable for pregnancy prevention.
KNOW YOUR OPTIONS
With the growing number of women in their reproductive years receiving solid-organ transplants in the United States, it is increasingly important for healthcare providers to be aware of contraceptive options and reproductive life planning for this high-risk population.
Safe and effective forms of contraception are available, and additional information to guide the choice can be found in the Summary Chart of US MEC for Contraceptive Use, which is also available in a free smart phone app through the CDC.44
Pregnancy after transplant carries high risks, requiring these patients to have special counseling and monitoring. Fortunately, planned pregnancy at least 1 year after transplant can lead to successful outcomes in these women.
- McKay DB, Josephson MA. Pregnancy in recipients of solid organs: effects on mother and child. N Engl J Med 2006; 354:1281–1293.
- US Department of Health and Human Services. Organ procurement and transplantation network. https://optn.transplant.hrsa.gov/. Accessed July 17, 2017.
- United States Renal Data System. 2014 annual data report. https://www.usrds.org/2014/view/Default.aspx. Accessed July 17, 2017.
- Hou S. Pregnancy in chronic renal insufficiency and end-stage renal disease. Am J Kidney Dis 1999; 33:235–252.
- Josephson MA, McKay DB. Women and transplantation: fertility, sexuality, pregnancy, contraception. Adv Chronic Kidney Dis 2013; 20:433–440.
- Gill JS, Zalunardo N, Rose C, Tonelli M. The pregnancy rate and live birth rate in kidney transplant recipients. Am J Transplant 2009; 9:1541–1549.
- Mohapatra A, Basu G. Pregnancy in kidney disease. Health Sciences 2012; 1(2). http://healthsciences.ac.in/july-sep-12/downloads/pregnancy_in_kidney_disease.pdf. Accessed July 25, 2017.
- Potluri K, Moldenhauer J, Karlman R, Hou S. Beta HCG levels in a pregnant dialysis patient: a cautionary tale. NDT Plus 2011; 4:42–43.
- Kennedy C, Hussein W, Spencer S, et al. Reproductive health in Irish female renal transplant recipients. Ir J Med Sci 2012; 181:59–63.
- Ghazizadeh S, Lessan-Pezeshki M, Khatami M, et al. Unwanted pregnancy among kidney transplant recipients in Iran. Transplant Proc 2005; 37:3085–3086.
- Jabiry-Zieniewicz Z, Bobrowska K, Kaminski P, Wielgos M, Zieniewicz K, Krawczyk M. Low-dose hormonal contraception after liver transplantation. Transplant Proc 2007; 39:1530–1532.
- Coscia LA, Constantinescu S, Moritz MJ, et al. Report from the National Transplantation Pregnancy Registry (NTPR): outcomes of pregnancy after transplantation. Clin Transpl 2010: 24:65–85.
- Mohamed-Ahmed O, Nelson-Piercy C, Bramham K, et al. Pregnancy outcomes in liver and cardiothoracic transplant recipients: a UK national cohort study. PLoS One 2014; 9:e89151.
- Enderby C, Keller CA. An overview of immunosuppression in solid organ transplantation. Am J Manag Care 2015; 21(suppl 1):s12–s23.
- Hoeltzenbein M, Elefant E, Vial T, et al. Teratogenicity of mycophenolate confirmed in a prospective study of the European Network of Teratology Information Services. Am J Med Genet A 2012; 158A:588–596.
- Polifka JE, Friedman JM. Teratogen update: azathioprine and 6-mercaptopurine. Teratology 2002; 65:240–261.
- Dinatale M. The pregnancy and lactation labeling rule (PLLR). US Food and Drug Administration, 2016. https://www.fda.gov/downloads/AdvisoryCommittees/CommitteesMeetingMaterials/PediatricAdvisoryCommittee/UCM520454.pdf. Accessed July 25, 2017.
- Lexicomp. http://online.lexi.com/lco/action/api/find/globalid/6612?utd=1. Accessed July 27, 2017.
- Kidney Disease: Improving Global Outcomes (KDIGO) Transplant Work Group. KDIGO clinical practice guideline for the care of kidney transplant recipients. Am J Transplant 2009; 9(suppl 3):S1–S155.
- Deshpande NA, Coscia LA, Gomez-Lobo V, Moritz MJ, Armenti VT. Pregnancy after solid organ transplantation: a guide for obstetric management. Rev Obstet Gynecol 2013; 6:116–125.
- Curtis KM, Tepper NK, Jatlaoui TC, et al. US medical eligibility criteria for contraceptive use, 2016. MMWR Recomm Rep 2016; 65:1–103.
- American College of Obstetricians and Gynecologists. ACOG Practice Bulletin No. 121: Long-acting reversible contraception: implants and intrauterine devices. Obstet Gynecol 2011; 118:184–196.
- Trussell J. Contraceptive failure in the United States. Contraception 2011; 83:397–404.
- Krajewski CM, Geetha D, Gomez-Lobo V. Contraceptive options for women with a history of solid-organ transplantation. Transplantation 2013; 95:1183–1186.
- Stern LF, Simons HR, Kohn JE, Debevec EJ, Morfesis JM, Patel AA. Differences in contraceptive use between family planning providers and the U.S. population: results of a nationwide survey. Contraception 2015; 91:464–469.
- Rafie S, Lai S, Garcia JE, Mody SK. Contraceptive use in female recipients of a solid-organ transplant. Prog Transplant 2014; 24:344–348.
- Labied S, Galant C, Nisolle M, et al. Differential elevation of matrix metalloproteinase expression in women exposed to levonorgestrel-releasing intrauterine system for a short or prolonged period of time. Hum Reprod 2009; 24:113–121.
- Kim CR, Martinez-Maza O, Magpantay L, et al. Immunologic evaluation of the endometrium with a levonorgestrel intrauterine device in solid organ transplant women and healthy controls. Contraception 2016; 94:534–540.
- Ramhendar T, Byrne P. Use of the levonorgestrel-releasing intrauterine system in renal transplant recipients: a retrospective case review. Contraception 2012; 86:288–289.
- Huguelet PS, Sheehan C, Spitzer RF, Scott S. Use of the levonorgestrel 52-mg intrauterine system in adolescent and young adult solid organ transplant recipients: a case series. Contraception 2017; 95:378–381.
- Peterson HB, Xia Z, Hughes JM, Wilcox LS, Tylor LR, Trussell J. The risk of pregnancy after tubal sterilization: findings from the US Collaborative Review of Sterilization. Am J Obstet Gynecol 1996; 174:1161–1168.
- Canalis E, Mazziotti G, Giustina A, Bilezikian JP. Glucocorticoid-induced osteoporosis: pathophysiology and therapy. Osteoporos Int 2007; 18:1319–1328.
- Krajewski C, Sucato G. Reproductive health care after transplantation. Best Pract Res Clin Obstet Gynaecol 2014; 28:1222–1234.
- World Health Organization. Medical eligibility criteria for contraceptive use. Fifth edition 2015. http://apps.who.int/iris/bitstream/10665/172915/1/WHO_RHR_15.07_eng.pdf. Accessed July 27, 2017.
- Pietrzak B, Bobrowska K, Jabiry-Zieniewicz Z, et al. Oral and transdermal hormonal contraception in women after kidney transplantation. Transplant Proc 2007; 39:2759–2762.
- Jain J, Jakimiuk AJ, Bode FR, Ross D, Kaunitz AM. Contraceptive efficacy and safety of DMPA-SC. Contraception 2004; 70:269–275.
- Vessey M, Painter R, Yeates D. Mortality in relation to oral contraceptive use and cigarette smoking. Lancet 2003; 362:185–191.
- van den Heuvel MW, van Bragt AJ, Alnabawy AK, Kaptein MC. Comparison of ethinylestradiol pharmacokinetics in three hormonal contraceptive formulations: the vaginal ring, the transdermal patch and an oral contraceptive. Contraception 2005; 72:168–174.
- Jick SS, Kaye JA, Russmann S, Jick H. Risk of nonfatal venous thromboembolism in women using a contraceptive transdermal patch and oral contraceptives containing norgestimate and 35 microg of ethinyl estradiol. Contraception 2006; 73:223–228.
- Jick S, Kaye JA, Li L, Jick H. Further results on the risk of nonfatal venous thromboembolism in users of the contraceptive transdermal patch compared to users of oral contraceptives containing norgestimate and 35 microg of ethinyl estradiol. Contraception 2007; 76:4–7.
- Estes CM, Westhoff C. Contraception for the transplant patient. Semin Perinatol 2007; 31:372–377.
- Cole JA, Norman H, Doherty M, Walker AM. Venous thromboembolism, myocardial infarction, and stroke among transdermal contraceptive system users. Obstet Gynecol 2007; 109:339–346.
- Paternoster DM, Riboni F, Bertolino M, et al. The contraceptive vaginal ring in women with renal and liver transplantation: analysis of preliminary results. Transplant Proc 2010; 42:1162–1165.
- Centers for Disease Control and Prevention (CDC). Summary chart of US medical eligibility criteria for contraceptive use. https://www.cdc.gov/reproductivehealth/unintendedpregnancy/pdf/legal_summary-chart_english_final_tag508.pdf. Accessed July 17, 2017.
Increasing numbers of women of childbearing age are receiving solid-organ transplants. All need counseling on how to prevent pregnancy while they are taking immunosuppressive agents. Some want to become pregnant after their transplant and thus require counseling and follow-up to maintain good health during pregnancy (Table 1).1
Primary care physicians can assist with basic contraception counseling and pregnancy planning for their patients who have had solid-organ transplants. In this review, we describe contraceptive options and pregnancy planning for these women.
TRANSPLANTS IN WOMEN ARE INCREASING
Over the past 20 years, the number of solid-organ transplants in US women has increased steadily. Since 1988, 38% of the 634,000 transplants performed were in women, and 47% of these women were of childbearing age (ages 18 to 49).2 Kidneys accounted for 60% of solid-organ transplants,2 and kidney transplant is now commonly performed in women of childbearing age. In 2012, of 176,000 patients with a functioning renal graft, 40.5% were women, and recipients between ages 20 and 44 composed the second-largest age group.3
FERTILITY IN WOMEN WITH END-STAGE RENAL DISEASE
Women in their reproductive years who have end-stage renal disease have lower fertility rates than women in the general population. In women undergoing peritoneal dialysis or hemodialysis, conception rates decrease to around 0.5% per year.4 This lower rate is most likely related to hypothalamic-pituitary-gonadal dysfunction, leading to reduced or total impairment of ovulation, menstrual irregularities, and infertility.5
Fertility often returns within a few months after transplant,1,6 and reported posttransplant pregnancy rates range from 3.3% to 18%,7–9 with up to one-third of pregnancies being unintended.6,10 These numbers are likely an underestimate because they do not reflect all pregnancies that are terminated, as many women do not voluntarily report having had an abortion.
Fertility is also severely diminished in women with end-stage liver disease. After liver transplant, sex hormone levels return to normal for many women, and menses soon resume.11
In 2005, the National Transplantation Pregnancy Registry reported 1,418 pregnancies in 919 female recipients of solid-organ transplants. In 2010, this number had increased to 1,940 pregnancies in 1,185 recipients, of whom 75% were kidney transplant recipients.12
A successful pregnancy outcome is most likely when a minimum of 1 year intervenes between transplant and conception.12,13
TERATOGENICITY OF IMMUNOSUPPRESSANTS
Immunosuppressant drugs commonly used for maintenance therapy after solid-organ transplant include the following:
- Calcineurin inhibitors (eg, cyclosporine, tacrolimus)
- Antiproliferative and antimetabolite agents (eg, mycophenolate mofetil, azathioprine)
- Corticosteroids
- Mammalian target of rapamycin inhibitors (eg, sirolimus, everolimus)
- T-cell costimulation blockers (eg, belatacept).14
The US Food and Drug Administration (FDA) previously classified mycophenolate mofetil and azathioprine in pregnancy risk category D (positive evidence of human fetal risk). The teratogenic risk of mycophenolate mofetil is well established in studies documenting specific congenital malformations and fetal loss in the first trimester.13,15 The teratogenic risk of azathioprine, on the other hand, is estimated to be minimal to small.16 Many of the associated fetal abnormalities may be related to the complexity of the underlying medical condition of the mother rather than to the medication.16
In June 2015, the FDA’s new Pregnancy and Lactation Labeling Rule went into effect, which removes the pregnancy letter categories A, B, C, D, and X from labeling.17 This rule was designed to help providers counsel their patients regarding the specific risks and benefits of a drug when used by pregnant or nursing women. However, the ABCDX categories are still commonly used. Table 2 shows information about the risks during pregnancy and lactation posed by the immunosuppressive drugs commonly used by posttransplant patients.18
CRITERIA FOR A SUCCESSFUL PREGNANCY
To ensure a safe and successful pregnancy with the fewest fetal and maternal complications, women are generally advised to avoid pregnancy for at least 1 year after transplant.19,20
In addition, women should meet certain clinical prerequisites after transplant before they conceive, as outlined by the American Society of Transplantation.19,20 These include:
- No rejection within the previous year
- Adequate and stable graft function (eg, serum creatinine < 1.5 mg/dL and urinary protein excretion < 500 mg/24 hours)
- No acute infection that might affect the fetus
- Maintenance immunosuppression at stable dosages.
Other circumstances to consider include episodes of rejection in the first year after transplant (as evidenced by biopsy results or glomerular filtration rate), the woman’s age (advanced maternal age is unfavorable), or any history of noncompliance.
Every pregnancy in a transplant recipient must be carefully planned. Primary care providers should encourage patients to meet with their transplant team and obstetricians early and often to allow time for the care team to adjust the type and dosing of immunosuppressant drugs, to ensure stable graft function, and to optimize any current chronic medical conditions such as diabetes mellitus or hypertension before conception.
CONTRACEPTIVE COUNSELING AFTER TRANSPLANT
Pregnancy should be avoided while transplant patients are taking FDA category D immunosuppressant drugs and, as already mentioned, during the first year after transplant. Unintended pregnancy can have serious health consequences for the mother and the fetus, as well as poor pregnancy outcomes. The US Centers for Disease Control and Prevention (CDC) lists solid-organ transplant within the past 2 years as a condition that can lead to adverse events as a result of pregnancy.21 After a transplant, a woman’s risks from an unintended pregnancy are always greater than the risks from any contraceptive, and this is important to reinforce in counseling.
Two forms of reliable contraception should be used at all times, and consistent condom use should be encouraged as one of the methods. Condoms are not reliable when used as the sole contraceptive method because they have an 18% typical-use failure rate. However, they are an excellent adjunct to other contraceptive methods because they have the additional benefit of protecting against sexually transmitted disease.
Choosing the appropriate contraceptive method for recipients of solid-organ transplants can be challenging because of several factors, including the recipient’s preexisting medical problems and drug interactions of immunosuppressant medications.
CDC criteria and categories for contraceptive use
In 2010, the CDC released the US version of the Medical Eligibility Criteria (US MEC) for contraceptive use, which was based on the 2009 World Health Organization Medical Eligibility Criteria (WHO MEC); these criteria were revised in August 2016.21
- Category 1: A condition for which there is no restriction for the use of the contraceptive method
- Category 2: A condition for which the advantages of using the method generally outweigh the theoretical or proven risks
- Category 3: A condition for which the theoretical or proven risks usually outweigh the advantages of using the method
- Category 4: A condition that represents an unacceptable health risk if the contraceptive method is used.
These recommendations aimed to improve family planning options by clarifying the possible safe and effective contraceptive options available while considering the patient’s medical condition. The CDC added solid-organ transplant recipients to this document because of the prevalence of this group in the United States.
The CDC categorizes a patient’s medical condition after transplant as either complicated or uncomplicated. Complicated conditions include acute or chronic graft failure, graft rejection, and cardiac allograft vasculopathy.21
Effectiveness of contraceptive methods
Contraceptive methods can be divided into 4 categories based on estimated effectiveness, ie, the pregnancy rate with “typical use” of that particular method in 1 year21–23:
- Very effective (0%–0.9%)
- Effective (1%–9%)
- Moderately effective (10%–25%)
- Less effective (26%–32%).
Typical use refers to failure rates for women and men whose use is not consistent nor always correct. Correct use, also described in the sections that follow, refers to failure rates for those whose use is consistent and always correct.
Women should be counseled regarding all available contraceptive options that are medically suitable for them, so they can choose the method that best fits their needs and lifestyle. They should receive counseling on emergency contraception, barrier protection against sexually transmitted disease, and the correct use of the contraceptive method they choose. They should be advised that if their chosen contraceptive method is unsatisfactory for any reason, they can switch to another method. Most importantly, providers need to impress on their patients that the risks associated with unintended pregnancy are far greater than the risks from any of the contraceptive methods.
VERY EFFECTIVE CONTRACEPTIVES (UNINTENDED PREGNANCY RATE 0%–0.9%)
This tier of contraception is the most effective regardless of the patient’s adherence; it includes long-acting, reversible contraceptives and permanent sterilization (both male and female) (Table 3).21–23
Long-acting reversible contraceptives include intrauterine devices (IUDs) and the subdermal etonogestrel implant. Given their efficacy and favorable safety profile, long-acting reversible contraceptives are being promoted for use in women who have chronic medical conditions, such as transplants.24
Intrauterine devices
IUDs are long-acting and reversible. They can be used by women who are nulliparous and those of all ages, including adolescents.22
Two types of IUDs are available in the United States: nonhormonal (copper) and hormonal (levonorgestrel). The copper IUD is effective for at least 10 years, whereas the levonorgestrel IUDs last for 3 to 5 years.22
Four levonorgestrel IUDs are currently available in the United States. Their sizes and doses vary: Mirena (52 µg), Skyla (13.5 µg), Liletta (52 µg), and Kyleena (19.5 µg).
Fewer than 1% of women become pregnant in the first year of IUD use.22,23 IUDs are an ideal option for women with solid-organ transplants because they are so effective and because the patient does not have to do anything once the IUD is in.22–24 The levonorgestrel IUD Mirena has the additional advantage of reducing heavy menstrual bleeding and is currently the only hormonal IUD with FDA approval for the management of menorrhagia.
About 12% of women in the general population use IUDs as their contraceptive method of choice,25 whereas after solid-organ transplantation about 15% to 20% of women do.26
Two historic concerns regarding IUDs may explain their low rate of use in transplant recipients.
First, IUDs were believed to be less effective in women on immunosuppressive drugs because IUDs act by inducing a local inflammatory reaction. However, IUDs involve macrophage activation, which is independent of the immune processes modified by immunosuppressants (primarily T-cell function).27 A recent pilot study showed a strong inflammatory reaction in the endometrium of transplant recipients after levonorgestrel IUD insertion.28
Second, there was concern about the increased risk of pelvic inflammatory disease with IUDs, but studies have shown levonorgestrel IUDs to be safe in transplant patients.29,30
The CDC21 lists copper and levonorgestrel IUDs in MEC category 3 (the risks generally outweigh the advantages) for initiation in patients with complicated transplants and in category 2 (advantages generally outweigh the risks) in patients with uncomplicated organ transplants. The devices are in category 2 for both complicated and uncomplicated cases if the IUD is already in place.
Subdermal implant
A subdermal implant consisting of a single rod containing 68 mg of etonogestrel is commercially available in the United States. It is one of the most effective contraceptive methods, with the lowest rates of pregnancy—less than 1% per year, with protection lasting at least 3 years.22,23 This low risk makes the subdermal implant a suitable method of contraception after transplant. Daily compliance is not required, and there are no hepatic first-pass effects, which results in higher bioavailability and less chance of drug interactions.
The main disadvantage of the subdermal implant and IUDs is unscheduled bleeding. An important benefit is prolonged amenorrhea, not only for patient convenience, but for reduction of endometrial cancer risk. Insertion and removal of the implant are considered minor office procedures. The implants are classified as US MEC category 2 in uncomplicated cases; initiation in complicated cases is considered category 3 but continuation is considered category 2.21
Permanent sterilization
Permanent sterilization is another option for women and men. In women, the fallopian tubes can be occluded with a coil system implanted vaginally through a hysteroscope, or they can be severed, tied, or clamped in a laparoscopic procedure or during cesarean delivery. Pregnancy rates after tubal ligation are less than 1%,23,31 although concern exists for high failure rates with the hysteroscopic method.
Because younger patients are more likely than older patients to subsequently regret having the procedure done, all available contraceptive options should be discussed with them.31
For men, permanent sterilization is done by vasectomy, which has less associated risk and cost compared with sterilization for women.
EFFECTIVE CONTRACEPTIVE METHODS (UNINTENDED PREGNANCY RATE 1%–9%)
Effective contraceptive methods, the next tier down from very effective methods, include injectable contraceptives, combined hormonal contraceptives, and progestin-only contraceptives (Table 4).
Injectable contraceptives
Depot medroxyprogesterone acetate is an injectable progestin-only contraceptive that carries a pregnancy risk of 6% with typical use and less than 1% with correct use.23 Thus, some failures are due to patients not returning for follow-up, but in some patients this method is not effective. Injections are given intramuscularly once every 3 months, avoiding the need for daily use.
A valid concern for transplant patients is that medroxyprogesterone acetate reduces bone mineral density. Although the bone effects are reversible in healthy adult women, caution is needed when prescribing this option to transplant patients who are already at increased risk of bone disease attributable to renal osteodystrophy and chronic corticosteroid use. 32,33
Recently, a subcutaneous formulation of depot medroxyprogesterone acetate (104 mg)was added to the WHO MEC for contraceptive use.34,35 The recommendations for the subcutaneous form are similar to those for the intramuscular form. In healthy women, the subcutaneous formulation is as safe and effective as the intramuscular form,36 but its efficacy after solid-organ transplant has not been determined. Both forms of depot medroxyprogesterone acetate are category 2 in the US MEC for both complicated and uncomplicated transplant cases.21
Combined hormonal contraceptives
Combined hormonal contraceptives contain both estrogen and progesterone and are available as pills, patches, or rings. Each product has an unintended pregnancy risk of 9% with typical use and less than 1% with correct use.23 They require strict patient adherence to regular daily use, which likely explains their high failure rate with typical use.
Combined hormonal contraceptives reduce mortality risk in women in the general population,37 but their effect on mortality risk after transplant is unknown and needs further study. In women who received liver transplants, low-dose combined hormonal contraceptives have been found to be effective and well tolerated, but initiation should be delayed at least 6 months until postoperative organ stability is demonstrated.11
Combined oral contraceptives are the most widely prescribed because they are convenient and familiar and have an acceptable safety profile in transplant patients,11,33,37 despite their high failure rate with typical use. They regulate the menstrual cycle and reduce anemia associated with menstruation.
The transdermal contraceptive patch has a mechanism of action similar to that of the combined oral contraceptives, but it delivers estrogen and progesterone transdermally through the abdominal wall, thus avoiding first-pass metabolism in the liver and enzymatic degradation in the gut. It delivers 35 µg of ethinyl estradiol and 150 µg of norelgestromin (an active metabolite of norgestimate) daily.38 It may cause higher circulating levels of estrogen than a combined oral contraceptive and may be associated with a higher risk of venous thromboembolism, but the evidence is conflicting.39–42
The vaginal ring, made of Silastic, delivers ethinyl estradiol in a low dose (15 µg/day) and etonorgestrel 0.12 mg/day. Like the patch, it has the advantage of bypassing first-pass metabolism in the liver, making it a good option for transplant patients who are taking antirejection drugs, thus avoiding drug interactions.41
Both the transdermal patch and vaginal ring were studied in transplant patients and had favorable results.24,43 The combined hormonal oral contraceptive pills, patch, and ring are in category 4 (unacceptable health risk) in the US MEC in patients with complicated cases, but they are in category 2 in uncomplicated cases.21
Combined hormonal contraceptives should not be considered first-line options by themselves for transplant patients because of their high failure rate with typical use.24
Progestin-only pills
Although progestin-only pills have not been studied specifically in transplant patients, they can be considered for women who have contraindications to estrogen use. Estrogen use is contraindicated in women with a history of venous thromboembolism, thrombogenic mutations, estrogen-dependent neoplasia, hepatocellular adenoma, severe hypertension, vascular disease, and Budd-Chiari syndrome.
Progestin-only pills inhibit ovulation in only about half of a woman’s cycles, but they prevent conception by other mechanisms as well, such as causing thickening of the cervical mucus. They also alter the endometrium to make it unfavorable for implantation and reduce the ciliary activity of the fallopian tube.
Strict adherence is important for effectiveness because progestin-only pills have a shorter half-life than combined hormonal contraceptives and also suppress ovulation less effectively.22 Failure rates are similar or somewhat higher than with combined hormonal contraceptives; with typical use, about 9 in 100 women can become pregnant in the first year.23 According to the US MEC,21 progestin-only pills are classified as category 2 for patients after both complicated and uncomplicated transplants.
MODERATELY EFFECTIVE METHODS (PREGNANCY RATE 10%–25%)
This tier of contraceptives includes all barrier methods, ie, male and female condoms, vaginal diaphragms, cervical caps, and sponges (Table 5).
Condoms (male and female)
When male condoms are used as the only birth control method, pregnancy occurs less often (18% with typical use and 2% with correct use) than with female condoms (21% with typical use and 5% with correct use).23 Male and female condoms are the only contraceptive methods that also prevent transmission of sexually transmitted disease.24
Caps, sponges, diaphragms
Cervical caps, vaginal sponges, and vaginal diaphragms are other forms of barrier contraceptives. All barrier methods should be combined with another contraceptive method to provide reliable protection against pregnancy. These methods are considered category 1 according to the US MEC.
LESS-EFFECTIVE METHODS
Fertility awareness-based methods such as the rhythm method have an associated pregnancy rate of about 25% with typical use and 3% to 5% with correct use23 and cannot be relied on for use by transplant recipients.24
Withdrawal and spermicides are considered least effective and unreliable for pregnancy prevention.
KNOW YOUR OPTIONS
With the growing number of women in their reproductive years receiving solid-organ transplants in the United States, it is increasingly important for healthcare providers to be aware of contraceptive options and reproductive life planning for this high-risk population.
Safe and effective forms of contraception are available, and additional information to guide the choice can be found in the Summary Chart of US MEC for Contraceptive Use, which is also available in a free smart phone app through the CDC.44
Pregnancy after transplant carries high risks, requiring these patients to have special counseling and monitoring. Fortunately, planned pregnancy at least 1 year after transplant can lead to successful outcomes in these women.
Increasing numbers of women of childbearing age are receiving solid-organ transplants. All need counseling on how to prevent pregnancy while they are taking immunosuppressive agents. Some want to become pregnant after their transplant and thus require counseling and follow-up to maintain good health during pregnancy (Table 1).1
Primary care physicians can assist with basic contraception counseling and pregnancy planning for their patients who have had solid-organ transplants. In this review, we describe contraceptive options and pregnancy planning for these women.
TRANSPLANTS IN WOMEN ARE INCREASING
Over the past 20 years, the number of solid-organ transplants in US women has increased steadily. Since 1988, 38% of the 634,000 transplants performed were in women, and 47% of these women were of childbearing age (ages 18 to 49).2 Kidneys accounted for 60% of solid-organ transplants,2 and kidney transplant is now commonly performed in women of childbearing age. In 2012, of 176,000 patients with a functioning renal graft, 40.5% were women, and recipients between ages 20 and 44 composed the second-largest age group.3
FERTILITY IN WOMEN WITH END-STAGE RENAL DISEASE
Women in their reproductive years who have end-stage renal disease have lower fertility rates than women in the general population. In women undergoing peritoneal dialysis or hemodialysis, conception rates decrease to around 0.5% per year.4 This lower rate is most likely related to hypothalamic-pituitary-gonadal dysfunction, leading to reduced or total impairment of ovulation, menstrual irregularities, and infertility.5
Fertility often returns within a few months after transplant,1,6 and reported posttransplant pregnancy rates range from 3.3% to 18%,7–9 with up to one-third of pregnancies being unintended.6,10 These numbers are likely an underestimate because they do not reflect all pregnancies that are terminated, as many women do not voluntarily report having had an abortion.
Fertility is also severely diminished in women with end-stage liver disease. After liver transplant, sex hormone levels return to normal for many women, and menses soon resume.11
In 2005, the National Transplantation Pregnancy Registry reported 1,418 pregnancies in 919 female recipients of solid-organ transplants. In 2010, this number had increased to 1,940 pregnancies in 1,185 recipients, of whom 75% were kidney transplant recipients.12
A successful pregnancy outcome is most likely when a minimum of 1 year intervenes between transplant and conception.12,13
TERATOGENICITY OF IMMUNOSUPPRESSANTS
Immunosuppressant drugs commonly used for maintenance therapy after solid-organ transplant include the following:
- Calcineurin inhibitors (eg, cyclosporine, tacrolimus)
- Antiproliferative and antimetabolite agents (eg, mycophenolate mofetil, azathioprine)
- Corticosteroids
- Mammalian target of rapamycin inhibitors (eg, sirolimus, everolimus)
- T-cell costimulation blockers (eg, belatacept).14
The US Food and Drug Administration (FDA) previously classified mycophenolate mofetil and azathioprine in pregnancy risk category D (positive evidence of human fetal risk). The teratogenic risk of mycophenolate mofetil is well established in studies documenting specific congenital malformations and fetal loss in the first trimester.13,15 The teratogenic risk of azathioprine, on the other hand, is estimated to be minimal to small.16 Many of the associated fetal abnormalities may be related to the complexity of the underlying medical condition of the mother rather than to the medication.16
In June 2015, the FDA’s new Pregnancy and Lactation Labeling Rule went into effect, which removes the pregnancy letter categories A, B, C, D, and X from labeling.17 This rule was designed to help providers counsel their patients regarding the specific risks and benefits of a drug when used by pregnant or nursing women. However, the ABCDX categories are still commonly used. Table 2 shows information about the risks during pregnancy and lactation posed by the immunosuppressive drugs commonly used by posttransplant patients.18
CRITERIA FOR A SUCCESSFUL PREGNANCY
To ensure a safe and successful pregnancy with the fewest fetal and maternal complications, women are generally advised to avoid pregnancy for at least 1 year after transplant.19,20
In addition, women should meet certain clinical prerequisites after transplant before they conceive, as outlined by the American Society of Transplantation.19,20 These include:
- No rejection within the previous year
- Adequate and stable graft function (eg, serum creatinine < 1.5 mg/dL and urinary protein excretion < 500 mg/24 hours)
- No acute infection that might affect the fetus
- Maintenance immunosuppression at stable dosages.
Other circumstances to consider include episodes of rejection in the first year after transplant (as evidenced by biopsy results or glomerular filtration rate), the woman’s age (advanced maternal age is unfavorable), or any history of noncompliance.
Every pregnancy in a transplant recipient must be carefully planned. Primary care providers should encourage patients to meet with their transplant team and obstetricians early and often to allow time for the care team to adjust the type and dosing of immunosuppressant drugs, to ensure stable graft function, and to optimize any current chronic medical conditions such as diabetes mellitus or hypertension before conception.
CONTRACEPTIVE COUNSELING AFTER TRANSPLANT
Pregnancy should be avoided while transplant patients are taking FDA category D immunosuppressant drugs and, as already mentioned, during the first year after transplant. Unintended pregnancy can have serious health consequences for the mother and the fetus, as well as poor pregnancy outcomes. The US Centers for Disease Control and Prevention (CDC) lists solid-organ transplant within the past 2 years as a condition that can lead to adverse events as a result of pregnancy.21 After a transplant, a woman’s risks from an unintended pregnancy are always greater than the risks from any contraceptive, and this is important to reinforce in counseling.
Two forms of reliable contraception should be used at all times, and consistent condom use should be encouraged as one of the methods. Condoms are not reliable when used as the sole contraceptive method because they have an 18% typical-use failure rate. However, they are an excellent adjunct to other contraceptive methods because they have the additional benefit of protecting against sexually transmitted disease.
Choosing the appropriate contraceptive method for recipients of solid-organ transplants can be challenging because of several factors, including the recipient’s preexisting medical problems and drug interactions of immunosuppressant medications.
CDC criteria and categories for contraceptive use
In 2010, the CDC released the US version of the Medical Eligibility Criteria (US MEC) for contraceptive use, which was based on the 2009 World Health Organization Medical Eligibility Criteria (WHO MEC); these criteria were revised in August 2016.21
- Category 1: A condition for which there is no restriction for the use of the contraceptive method
- Category 2: A condition for which the advantages of using the method generally outweigh the theoretical or proven risks
- Category 3: A condition for which the theoretical or proven risks usually outweigh the advantages of using the method
- Category 4: A condition that represents an unacceptable health risk if the contraceptive method is used.
These recommendations aimed to improve family planning options by clarifying the possible safe and effective contraceptive options available while considering the patient’s medical condition. The CDC added solid-organ transplant recipients to this document because of the prevalence of this group in the United States.
The CDC categorizes a patient’s medical condition after transplant as either complicated or uncomplicated. Complicated conditions include acute or chronic graft failure, graft rejection, and cardiac allograft vasculopathy.21
Effectiveness of contraceptive methods
Contraceptive methods can be divided into 4 categories based on estimated effectiveness, ie, the pregnancy rate with “typical use” of that particular method in 1 year21–23:
- Very effective (0%–0.9%)
- Effective (1%–9%)
- Moderately effective (10%–25%)
- Less effective (26%–32%).
Typical use refers to failure rates for women and men whose use is not consistent nor always correct. Correct use, also described in the sections that follow, refers to failure rates for those whose use is consistent and always correct.
Women should be counseled regarding all available contraceptive options that are medically suitable for them, so they can choose the method that best fits their needs and lifestyle. They should receive counseling on emergency contraception, barrier protection against sexually transmitted disease, and the correct use of the contraceptive method they choose. They should be advised that if their chosen contraceptive method is unsatisfactory for any reason, they can switch to another method. Most importantly, providers need to impress on their patients that the risks associated with unintended pregnancy are far greater than the risks from any of the contraceptive methods.
VERY EFFECTIVE CONTRACEPTIVES (UNINTENDED PREGNANCY RATE 0%–0.9%)
This tier of contraception is the most effective regardless of the patient’s adherence; it includes long-acting, reversible contraceptives and permanent sterilization (both male and female) (Table 3).21–23
Long-acting reversible contraceptives include intrauterine devices (IUDs) and the subdermal etonogestrel implant. Given their efficacy and favorable safety profile, long-acting reversible contraceptives are being promoted for use in women who have chronic medical conditions, such as transplants.24
Intrauterine devices
IUDs are long-acting and reversible. They can be used by women who are nulliparous and those of all ages, including adolescents.22
Two types of IUDs are available in the United States: nonhormonal (copper) and hormonal (levonorgestrel). The copper IUD is effective for at least 10 years, whereas the levonorgestrel IUDs last for 3 to 5 years.22
Four levonorgestrel IUDs are currently available in the United States. Their sizes and doses vary: Mirena (52 µg), Skyla (13.5 µg), Liletta (52 µg), and Kyleena (19.5 µg).
Fewer than 1% of women become pregnant in the first year of IUD use.22,23 IUDs are an ideal option for women with solid-organ transplants because they are so effective and because the patient does not have to do anything once the IUD is in.22–24 The levonorgestrel IUD Mirena has the additional advantage of reducing heavy menstrual bleeding and is currently the only hormonal IUD with FDA approval for the management of menorrhagia.
About 12% of women in the general population use IUDs as their contraceptive method of choice,25 whereas after solid-organ transplantation about 15% to 20% of women do.26
Two historic concerns regarding IUDs may explain their low rate of use in transplant recipients.
First, IUDs were believed to be less effective in women on immunosuppressive drugs because IUDs act by inducing a local inflammatory reaction. However, IUDs involve macrophage activation, which is independent of the immune processes modified by immunosuppressants (primarily T-cell function).27 A recent pilot study showed a strong inflammatory reaction in the endometrium of transplant recipients after levonorgestrel IUD insertion.28
Second, there was concern about the increased risk of pelvic inflammatory disease with IUDs, but studies have shown levonorgestrel IUDs to be safe in transplant patients.29,30
The CDC21 lists copper and levonorgestrel IUDs in MEC category 3 (the risks generally outweigh the advantages) for initiation in patients with complicated transplants and in category 2 (advantages generally outweigh the risks) in patients with uncomplicated organ transplants. The devices are in category 2 for both complicated and uncomplicated cases if the IUD is already in place.
Subdermal implant
A subdermal implant consisting of a single rod containing 68 mg of etonogestrel is commercially available in the United States. It is one of the most effective contraceptive methods, with the lowest rates of pregnancy—less than 1% per year, with protection lasting at least 3 years.22,23 This low risk makes the subdermal implant a suitable method of contraception after transplant. Daily compliance is not required, and there are no hepatic first-pass effects, which results in higher bioavailability and less chance of drug interactions.
The main disadvantage of the subdermal implant and IUDs is unscheduled bleeding. An important benefit is prolonged amenorrhea, not only for patient convenience, but for reduction of endometrial cancer risk. Insertion and removal of the implant are considered minor office procedures. The implants are classified as US MEC category 2 in uncomplicated cases; initiation in complicated cases is considered category 3 but continuation is considered category 2.21
Permanent sterilization
Permanent sterilization is another option for women and men. In women, the fallopian tubes can be occluded with a coil system implanted vaginally through a hysteroscope, or they can be severed, tied, or clamped in a laparoscopic procedure or during cesarean delivery. Pregnancy rates after tubal ligation are less than 1%,23,31 although concern exists for high failure rates with the hysteroscopic method.
Because younger patients are more likely than older patients to subsequently regret having the procedure done, all available contraceptive options should be discussed with them.31
For men, permanent sterilization is done by vasectomy, which has less associated risk and cost compared with sterilization for women.
EFFECTIVE CONTRACEPTIVE METHODS (UNINTENDED PREGNANCY RATE 1%–9%)
Effective contraceptive methods, the next tier down from very effective methods, include injectable contraceptives, combined hormonal contraceptives, and progestin-only contraceptives (Table 4).
Injectable contraceptives
Depot medroxyprogesterone acetate is an injectable progestin-only contraceptive that carries a pregnancy risk of 6% with typical use and less than 1% with correct use.23 Thus, some failures are due to patients not returning for follow-up, but in some patients this method is not effective. Injections are given intramuscularly once every 3 months, avoiding the need for daily use.
A valid concern for transplant patients is that medroxyprogesterone acetate reduces bone mineral density. Although the bone effects are reversible in healthy adult women, caution is needed when prescribing this option to transplant patients who are already at increased risk of bone disease attributable to renal osteodystrophy and chronic corticosteroid use. 32,33
Recently, a subcutaneous formulation of depot medroxyprogesterone acetate (104 mg)was added to the WHO MEC for contraceptive use.34,35 The recommendations for the subcutaneous form are similar to those for the intramuscular form. In healthy women, the subcutaneous formulation is as safe and effective as the intramuscular form,36 but its efficacy after solid-organ transplant has not been determined. Both forms of depot medroxyprogesterone acetate are category 2 in the US MEC for both complicated and uncomplicated transplant cases.21
Combined hormonal contraceptives
Combined hormonal contraceptives contain both estrogen and progesterone and are available as pills, patches, or rings. Each product has an unintended pregnancy risk of 9% with typical use and less than 1% with correct use.23 They require strict patient adherence to regular daily use, which likely explains their high failure rate with typical use.
Combined hormonal contraceptives reduce mortality risk in women in the general population,37 but their effect on mortality risk after transplant is unknown and needs further study. In women who received liver transplants, low-dose combined hormonal contraceptives have been found to be effective and well tolerated, but initiation should be delayed at least 6 months until postoperative organ stability is demonstrated.11
Combined oral contraceptives are the most widely prescribed because they are convenient and familiar and have an acceptable safety profile in transplant patients,11,33,37 despite their high failure rate with typical use. They regulate the menstrual cycle and reduce anemia associated with menstruation.
The transdermal contraceptive patch has a mechanism of action similar to that of the combined oral contraceptives, but it delivers estrogen and progesterone transdermally through the abdominal wall, thus avoiding first-pass metabolism in the liver and enzymatic degradation in the gut. It delivers 35 µg of ethinyl estradiol and 150 µg of norelgestromin (an active metabolite of norgestimate) daily.38 It may cause higher circulating levels of estrogen than a combined oral contraceptive and may be associated with a higher risk of venous thromboembolism, but the evidence is conflicting.39–42
The vaginal ring, made of Silastic, delivers ethinyl estradiol in a low dose (15 µg/day) and etonorgestrel 0.12 mg/day. Like the patch, it has the advantage of bypassing first-pass metabolism in the liver, making it a good option for transplant patients who are taking antirejection drugs, thus avoiding drug interactions.41
Both the transdermal patch and vaginal ring were studied in transplant patients and had favorable results.24,43 The combined hormonal oral contraceptive pills, patch, and ring are in category 4 (unacceptable health risk) in the US MEC in patients with complicated cases, but they are in category 2 in uncomplicated cases.21
Combined hormonal contraceptives should not be considered first-line options by themselves for transplant patients because of their high failure rate with typical use.24
Progestin-only pills
Although progestin-only pills have not been studied specifically in transplant patients, they can be considered for women who have contraindications to estrogen use. Estrogen use is contraindicated in women with a history of venous thromboembolism, thrombogenic mutations, estrogen-dependent neoplasia, hepatocellular adenoma, severe hypertension, vascular disease, and Budd-Chiari syndrome.
Progestin-only pills inhibit ovulation in only about half of a woman’s cycles, but they prevent conception by other mechanisms as well, such as causing thickening of the cervical mucus. They also alter the endometrium to make it unfavorable for implantation and reduce the ciliary activity of the fallopian tube.
Strict adherence is important for effectiveness because progestin-only pills have a shorter half-life than combined hormonal contraceptives and also suppress ovulation less effectively.22 Failure rates are similar or somewhat higher than with combined hormonal contraceptives; with typical use, about 9 in 100 women can become pregnant in the first year.23 According to the US MEC,21 progestin-only pills are classified as category 2 for patients after both complicated and uncomplicated transplants.
MODERATELY EFFECTIVE METHODS (PREGNANCY RATE 10%–25%)
This tier of contraceptives includes all barrier methods, ie, male and female condoms, vaginal diaphragms, cervical caps, and sponges (Table 5).
Condoms (male and female)
When male condoms are used as the only birth control method, pregnancy occurs less often (18% with typical use and 2% with correct use) than with female condoms (21% with typical use and 5% with correct use).23 Male and female condoms are the only contraceptive methods that also prevent transmission of sexually transmitted disease.24
Caps, sponges, diaphragms
Cervical caps, vaginal sponges, and vaginal diaphragms are other forms of barrier contraceptives. All barrier methods should be combined with another contraceptive method to provide reliable protection against pregnancy. These methods are considered category 1 according to the US MEC.
LESS-EFFECTIVE METHODS
Fertility awareness-based methods such as the rhythm method have an associated pregnancy rate of about 25% with typical use and 3% to 5% with correct use23 and cannot be relied on for use by transplant recipients.24
Withdrawal and spermicides are considered least effective and unreliable for pregnancy prevention.
KNOW YOUR OPTIONS
With the growing number of women in their reproductive years receiving solid-organ transplants in the United States, it is increasingly important for healthcare providers to be aware of contraceptive options and reproductive life planning for this high-risk population.
Safe and effective forms of contraception are available, and additional information to guide the choice can be found in the Summary Chart of US MEC for Contraceptive Use, which is also available in a free smart phone app through the CDC.44
Pregnancy after transplant carries high risks, requiring these patients to have special counseling and monitoring. Fortunately, planned pregnancy at least 1 year after transplant can lead to successful outcomes in these women.
- McKay DB, Josephson MA. Pregnancy in recipients of solid organs: effects on mother and child. N Engl J Med 2006; 354:1281–1293.
- US Department of Health and Human Services. Organ procurement and transplantation network. https://optn.transplant.hrsa.gov/. Accessed July 17, 2017.
- United States Renal Data System. 2014 annual data report. https://www.usrds.org/2014/view/Default.aspx. Accessed July 17, 2017.
- Hou S. Pregnancy in chronic renal insufficiency and end-stage renal disease. Am J Kidney Dis 1999; 33:235–252.
- Josephson MA, McKay DB. Women and transplantation: fertility, sexuality, pregnancy, contraception. Adv Chronic Kidney Dis 2013; 20:433–440.
- Gill JS, Zalunardo N, Rose C, Tonelli M. The pregnancy rate and live birth rate in kidney transplant recipients. Am J Transplant 2009; 9:1541–1549.
- Mohapatra A, Basu G. Pregnancy in kidney disease. Health Sciences 2012; 1(2). http://healthsciences.ac.in/july-sep-12/downloads/pregnancy_in_kidney_disease.pdf. Accessed July 25, 2017.
- Potluri K, Moldenhauer J, Karlman R, Hou S. Beta HCG levels in a pregnant dialysis patient: a cautionary tale. NDT Plus 2011; 4:42–43.
- Kennedy C, Hussein W, Spencer S, et al. Reproductive health in Irish female renal transplant recipients. Ir J Med Sci 2012; 181:59–63.
- Ghazizadeh S, Lessan-Pezeshki M, Khatami M, et al. Unwanted pregnancy among kidney transplant recipients in Iran. Transplant Proc 2005; 37:3085–3086.
- Jabiry-Zieniewicz Z, Bobrowska K, Kaminski P, Wielgos M, Zieniewicz K, Krawczyk M. Low-dose hormonal contraception after liver transplantation. Transplant Proc 2007; 39:1530–1532.
- Coscia LA, Constantinescu S, Moritz MJ, et al. Report from the National Transplantation Pregnancy Registry (NTPR): outcomes of pregnancy after transplantation. Clin Transpl 2010: 24:65–85.
- Mohamed-Ahmed O, Nelson-Piercy C, Bramham K, et al. Pregnancy outcomes in liver and cardiothoracic transplant recipients: a UK national cohort study. PLoS One 2014; 9:e89151.
- Enderby C, Keller CA. An overview of immunosuppression in solid organ transplantation. Am J Manag Care 2015; 21(suppl 1):s12–s23.
- Hoeltzenbein M, Elefant E, Vial T, et al. Teratogenicity of mycophenolate confirmed in a prospective study of the European Network of Teratology Information Services. Am J Med Genet A 2012; 158A:588–596.
- Polifka JE, Friedman JM. Teratogen update: azathioprine and 6-mercaptopurine. Teratology 2002; 65:240–261.
- Dinatale M. The pregnancy and lactation labeling rule (PLLR). US Food and Drug Administration, 2016. https://www.fda.gov/downloads/AdvisoryCommittees/CommitteesMeetingMaterials/PediatricAdvisoryCommittee/UCM520454.pdf. Accessed July 25, 2017.
- Lexicomp. http://online.lexi.com/lco/action/api/find/globalid/6612?utd=1. Accessed July 27, 2017.
- Kidney Disease: Improving Global Outcomes (KDIGO) Transplant Work Group. KDIGO clinical practice guideline for the care of kidney transplant recipients. Am J Transplant 2009; 9(suppl 3):S1–S155.
- Deshpande NA, Coscia LA, Gomez-Lobo V, Moritz MJ, Armenti VT. Pregnancy after solid organ transplantation: a guide for obstetric management. Rev Obstet Gynecol 2013; 6:116–125.
- Curtis KM, Tepper NK, Jatlaoui TC, et al. US medical eligibility criteria for contraceptive use, 2016. MMWR Recomm Rep 2016; 65:1–103.
- American College of Obstetricians and Gynecologists. ACOG Practice Bulletin No. 121: Long-acting reversible contraception: implants and intrauterine devices. Obstet Gynecol 2011; 118:184–196.
- Trussell J. Contraceptive failure in the United States. Contraception 2011; 83:397–404.
- Krajewski CM, Geetha D, Gomez-Lobo V. Contraceptive options for women with a history of solid-organ transplantation. Transplantation 2013; 95:1183–1186.
- Stern LF, Simons HR, Kohn JE, Debevec EJ, Morfesis JM, Patel AA. Differences in contraceptive use between family planning providers and the U.S. population: results of a nationwide survey. Contraception 2015; 91:464–469.
- Rafie S, Lai S, Garcia JE, Mody SK. Contraceptive use in female recipients of a solid-organ transplant. Prog Transplant 2014; 24:344–348.
- Labied S, Galant C, Nisolle M, et al. Differential elevation of matrix metalloproteinase expression in women exposed to levonorgestrel-releasing intrauterine system for a short or prolonged period of time. Hum Reprod 2009; 24:113–121.
- Kim CR, Martinez-Maza O, Magpantay L, et al. Immunologic evaluation of the endometrium with a levonorgestrel intrauterine device in solid organ transplant women and healthy controls. Contraception 2016; 94:534–540.
- Ramhendar T, Byrne P. Use of the levonorgestrel-releasing intrauterine system in renal transplant recipients: a retrospective case review. Contraception 2012; 86:288–289.
- Huguelet PS, Sheehan C, Spitzer RF, Scott S. Use of the levonorgestrel 52-mg intrauterine system in adolescent and young adult solid organ transplant recipients: a case series. Contraception 2017; 95:378–381.
- Peterson HB, Xia Z, Hughes JM, Wilcox LS, Tylor LR, Trussell J. The risk of pregnancy after tubal sterilization: findings from the US Collaborative Review of Sterilization. Am J Obstet Gynecol 1996; 174:1161–1168.
- Canalis E, Mazziotti G, Giustina A, Bilezikian JP. Glucocorticoid-induced osteoporosis: pathophysiology and therapy. Osteoporos Int 2007; 18:1319–1328.
- Krajewski C, Sucato G. Reproductive health care after transplantation. Best Pract Res Clin Obstet Gynaecol 2014; 28:1222–1234.
- World Health Organization. Medical eligibility criteria for contraceptive use. Fifth edition 2015. http://apps.who.int/iris/bitstream/10665/172915/1/WHO_RHR_15.07_eng.pdf. Accessed July 27, 2017.
- Pietrzak B, Bobrowska K, Jabiry-Zieniewicz Z, et al. Oral and transdermal hormonal contraception in women after kidney transplantation. Transplant Proc 2007; 39:2759–2762.
- Jain J, Jakimiuk AJ, Bode FR, Ross D, Kaunitz AM. Contraceptive efficacy and safety of DMPA-SC. Contraception 2004; 70:269–275.
- Vessey M, Painter R, Yeates D. Mortality in relation to oral contraceptive use and cigarette smoking. Lancet 2003; 362:185–191.
- van den Heuvel MW, van Bragt AJ, Alnabawy AK, Kaptein MC. Comparison of ethinylestradiol pharmacokinetics in three hormonal contraceptive formulations: the vaginal ring, the transdermal patch and an oral contraceptive. Contraception 2005; 72:168–174.
- Jick SS, Kaye JA, Russmann S, Jick H. Risk of nonfatal venous thromboembolism in women using a contraceptive transdermal patch and oral contraceptives containing norgestimate and 35 microg of ethinyl estradiol. Contraception 2006; 73:223–228.
- Jick S, Kaye JA, Li L, Jick H. Further results on the risk of nonfatal venous thromboembolism in users of the contraceptive transdermal patch compared to users of oral contraceptives containing norgestimate and 35 microg of ethinyl estradiol. Contraception 2007; 76:4–7.
- Estes CM, Westhoff C. Contraception for the transplant patient. Semin Perinatol 2007; 31:372–377.
- Cole JA, Norman H, Doherty M, Walker AM. Venous thromboembolism, myocardial infarction, and stroke among transdermal contraceptive system users. Obstet Gynecol 2007; 109:339–346.
- Paternoster DM, Riboni F, Bertolino M, et al. The contraceptive vaginal ring in women with renal and liver transplantation: analysis of preliminary results. Transplant Proc 2010; 42:1162–1165.
- Centers for Disease Control and Prevention (CDC). Summary chart of US medical eligibility criteria for contraceptive use. https://www.cdc.gov/reproductivehealth/unintendedpregnancy/pdf/legal_summary-chart_english_final_tag508.pdf. Accessed July 17, 2017.
- McKay DB, Josephson MA. Pregnancy in recipients of solid organs: effects on mother and child. N Engl J Med 2006; 354:1281–1293.
- US Department of Health and Human Services. Organ procurement and transplantation network. https://optn.transplant.hrsa.gov/. Accessed July 17, 2017.
- United States Renal Data System. 2014 annual data report. https://www.usrds.org/2014/view/Default.aspx. Accessed July 17, 2017.
- Hou S. Pregnancy in chronic renal insufficiency and end-stage renal disease. Am J Kidney Dis 1999; 33:235–252.
- Josephson MA, McKay DB. Women and transplantation: fertility, sexuality, pregnancy, contraception. Adv Chronic Kidney Dis 2013; 20:433–440.
- Gill JS, Zalunardo N, Rose C, Tonelli M. The pregnancy rate and live birth rate in kidney transplant recipients. Am J Transplant 2009; 9:1541–1549.
- Mohapatra A, Basu G. Pregnancy in kidney disease. Health Sciences 2012; 1(2). http://healthsciences.ac.in/july-sep-12/downloads/pregnancy_in_kidney_disease.pdf. Accessed July 25, 2017.
- Potluri K, Moldenhauer J, Karlman R, Hou S. Beta HCG levels in a pregnant dialysis patient: a cautionary tale. NDT Plus 2011; 4:42–43.
- Kennedy C, Hussein W, Spencer S, et al. Reproductive health in Irish female renal transplant recipients. Ir J Med Sci 2012; 181:59–63.
- Ghazizadeh S, Lessan-Pezeshki M, Khatami M, et al. Unwanted pregnancy among kidney transplant recipients in Iran. Transplant Proc 2005; 37:3085–3086.
- Jabiry-Zieniewicz Z, Bobrowska K, Kaminski P, Wielgos M, Zieniewicz K, Krawczyk M. Low-dose hormonal contraception after liver transplantation. Transplant Proc 2007; 39:1530–1532.
- Coscia LA, Constantinescu S, Moritz MJ, et al. Report from the National Transplantation Pregnancy Registry (NTPR): outcomes of pregnancy after transplantation. Clin Transpl 2010: 24:65–85.
- Mohamed-Ahmed O, Nelson-Piercy C, Bramham K, et al. Pregnancy outcomes in liver and cardiothoracic transplant recipients: a UK national cohort study. PLoS One 2014; 9:e89151.
- Enderby C, Keller CA. An overview of immunosuppression in solid organ transplantation. Am J Manag Care 2015; 21(suppl 1):s12–s23.
- Hoeltzenbein M, Elefant E, Vial T, et al. Teratogenicity of mycophenolate confirmed in a prospective study of the European Network of Teratology Information Services. Am J Med Genet A 2012; 158A:588–596.
- Polifka JE, Friedman JM. Teratogen update: azathioprine and 6-mercaptopurine. Teratology 2002; 65:240–261.
- Dinatale M. The pregnancy and lactation labeling rule (PLLR). US Food and Drug Administration, 2016. https://www.fda.gov/downloads/AdvisoryCommittees/CommitteesMeetingMaterials/PediatricAdvisoryCommittee/UCM520454.pdf. Accessed July 25, 2017.
- Lexicomp. http://online.lexi.com/lco/action/api/find/globalid/6612?utd=1. Accessed July 27, 2017.
- Kidney Disease: Improving Global Outcomes (KDIGO) Transplant Work Group. KDIGO clinical practice guideline for the care of kidney transplant recipients. Am J Transplant 2009; 9(suppl 3):S1–S155.
- Deshpande NA, Coscia LA, Gomez-Lobo V, Moritz MJ, Armenti VT. Pregnancy after solid organ transplantation: a guide for obstetric management. Rev Obstet Gynecol 2013; 6:116–125.
- Curtis KM, Tepper NK, Jatlaoui TC, et al. US medical eligibility criteria for contraceptive use, 2016. MMWR Recomm Rep 2016; 65:1–103.
- American College of Obstetricians and Gynecologists. ACOG Practice Bulletin No. 121: Long-acting reversible contraception: implants and intrauterine devices. Obstet Gynecol 2011; 118:184–196.
- Trussell J. Contraceptive failure in the United States. Contraception 2011; 83:397–404.
- Krajewski CM, Geetha D, Gomez-Lobo V. Contraceptive options for women with a history of solid-organ transplantation. Transplantation 2013; 95:1183–1186.
- Stern LF, Simons HR, Kohn JE, Debevec EJ, Morfesis JM, Patel AA. Differences in contraceptive use between family planning providers and the U.S. population: results of a nationwide survey. Contraception 2015; 91:464–469.
- Rafie S, Lai S, Garcia JE, Mody SK. Contraceptive use in female recipients of a solid-organ transplant. Prog Transplant 2014; 24:344–348.
- Labied S, Galant C, Nisolle M, et al. Differential elevation of matrix metalloproteinase expression in women exposed to levonorgestrel-releasing intrauterine system for a short or prolonged period of time. Hum Reprod 2009; 24:113–121.
- Kim CR, Martinez-Maza O, Magpantay L, et al. Immunologic evaluation of the endometrium with a levonorgestrel intrauterine device in solid organ transplant women and healthy controls. Contraception 2016; 94:534–540.
- Ramhendar T, Byrne P. Use of the levonorgestrel-releasing intrauterine system in renal transplant recipients: a retrospective case review. Contraception 2012; 86:288–289.
- Huguelet PS, Sheehan C, Spitzer RF, Scott S. Use of the levonorgestrel 52-mg intrauterine system in adolescent and young adult solid organ transplant recipients: a case series. Contraception 2017; 95:378–381.
- Peterson HB, Xia Z, Hughes JM, Wilcox LS, Tylor LR, Trussell J. The risk of pregnancy after tubal sterilization: findings from the US Collaborative Review of Sterilization. Am J Obstet Gynecol 1996; 174:1161–1168.
- Canalis E, Mazziotti G, Giustina A, Bilezikian JP. Glucocorticoid-induced osteoporosis: pathophysiology and therapy. Osteoporos Int 2007; 18:1319–1328.
- Krajewski C, Sucato G. Reproductive health care after transplantation. Best Pract Res Clin Obstet Gynaecol 2014; 28:1222–1234.
- World Health Organization. Medical eligibility criteria for contraceptive use. Fifth edition 2015. http://apps.who.int/iris/bitstream/10665/172915/1/WHO_RHR_15.07_eng.pdf. Accessed July 27, 2017.
- Pietrzak B, Bobrowska K, Jabiry-Zieniewicz Z, et al. Oral and transdermal hormonal contraception in women after kidney transplantation. Transplant Proc 2007; 39:2759–2762.
- Jain J, Jakimiuk AJ, Bode FR, Ross D, Kaunitz AM. Contraceptive efficacy and safety of DMPA-SC. Contraception 2004; 70:269–275.
- Vessey M, Painter R, Yeates D. Mortality in relation to oral contraceptive use and cigarette smoking. Lancet 2003; 362:185–191.
- van den Heuvel MW, van Bragt AJ, Alnabawy AK, Kaptein MC. Comparison of ethinylestradiol pharmacokinetics in three hormonal contraceptive formulations: the vaginal ring, the transdermal patch and an oral contraceptive. Contraception 2005; 72:168–174.
- Jick SS, Kaye JA, Russmann S, Jick H. Risk of nonfatal venous thromboembolism in women using a contraceptive transdermal patch and oral contraceptives containing norgestimate and 35 microg of ethinyl estradiol. Contraception 2006; 73:223–228.
- Jick S, Kaye JA, Li L, Jick H. Further results on the risk of nonfatal venous thromboembolism in users of the contraceptive transdermal patch compared to users of oral contraceptives containing norgestimate and 35 microg of ethinyl estradiol. Contraception 2007; 76:4–7.
- Estes CM, Westhoff C. Contraception for the transplant patient. Semin Perinatol 2007; 31:372–377.
- Cole JA, Norman H, Doherty M, Walker AM. Venous thromboembolism, myocardial infarction, and stroke among transdermal contraceptive system users. Obstet Gynecol 2007; 109:339–346.
- Paternoster DM, Riboni F, Bertolino M, et al. The contraceptive vaginal ring in women with renal and liver transplantation: analysis of preliminary results. Transplant Proc 2010; 42:1162–1165.
- Centers for Disease Control and Prevention (CDC). Summary chart of US medical eligibility criteria for contraceptive use. https://www.cdc.gov/reproductivehealth/unintendedpregnancy/pdf/legal_summary-chart_english_final_tag508.pdf. Accessed July 17, 2017.
KEY POINTS
- The number of solid-organ transplants in US women of childbearing age has increased over the past 20 years.
- Women should wait at least 1 year after receiving a solid-organ transplant before attempting to become pregnant, and then should do so only when cleared by the transplant team and obstetrician, with close monitoring.
- The various types of contraception can be grouped by their effectiveness and by the medical eligibility criteria set by the US Centers for Disease Control and Prevention.
- Transplant recipients of childbearing age should use 2 contraceptive methods concurrently, one of which should be condoms.
Combined hormonal contraceptives and migraine: An update on the evidence
Combined hormonal contraceptives are contraindicated in women who have migraine with aura because they pose a risk of stroke. But how great is the risk, and how strong is the evidence, particularly with today’s low-dose contraceptives? Can we view migraine with aura as a relative contraindication rather than an absolute one?
This article reviews migraine diagnosis, the effects of estrogen and the menstrual cycle on migraine, the evidence of stroke risk with combined hormonal contraceptive use, and how the frequency of aura may affect risk. It offers practical advice on choosing contraceptive formulations and counseling patients on risks and benefits.
WHAT THE GUIDELINES SAY
Current guidelines restrict the use of combined hormonal contraceptives in the setting of migraine with aura, but not in migraine without aura.
A practice bulletin from the American College of Obstetrics and Gynecology in 2010 noted that extended-cycle or continuous hormonal contraceptives, including oral and parenteral products, might provide relief of migraines by eliminating the drops in estrogen levels that precipitate them.1 However, the bulletin also cautioned that though cerebrovascular accidents in women are rare, the impact of a stroke is so devastating that clinicians should consider intrauterine devices, progestin-only options, and other nonestrogen methods in women who have migraine with focal neurologic signs, women who smoke, and women age 35 or older.1
In 2016, the US Centers for Disease Control and Prevention published updates to its medical eligibility criteria for contraceptive use in various medical conditions. In the case of migraine without aura, the guidelines note no limitation to the use of combined hormonal contraceptives, regardless of the patient’s age. In the case of migraine with aura, the consensus was that the risk associated with combined hormonal contraception typically outweighs its benefits, noting “an unacceptable health risk if the contraceptive method is used.”2
We believe a fresh look at the data is warranted.
EARLY ORAL CONTRACEPTIVES WERE ALL HIGH-DOSE
This issue first surfaced in the decade and a half after the initial launch of oral contraceptives in 1960. The products then were all high-dose pills, containing up to 150 µg of mestranol. In subsequent decades, the dose of estrogen was successively reduced, so that now some pills contain only 10 µg of ethinyl estradiol. High-dose pills—which today contain 50 µg of ethinyl estradiol—account for less than 1% of pills currently sold in the United States and have been eliminated in many countries.
DIAGNOSTIC CRITERIA FOR MIGRAINE
According to the International Classification of Headache Disorders (ICHD),3 the diagnosis of migraine requires 2 of the 4 following criteria:
- Unilateral location
- Pulsating or throbbing pain
- Pain of at least moderate intensity
- Pain aggravated by activity, or causing a preference to avoid activity.
An additional criterion is either nausea or a combination of photophobia and phonophobia with the episode. This criterion can be met if the patient prefers to avoid bright lights and loud noises during an attack.
Headache experts have suggested that patients with a stable pattern of episodic, disabling headache and normal findings on physical examination should be considered to have migraine if there is no contradictory evidence.4,5
Migraine with aura requires at least 2 of the following 4 characteristics3:
- 1 aura symptom, spreading gradually over 5 minutes, or 2 or more aura symptoms occurring in succession, or both
- Each aura symptom lasting 5 to 60 minutes (not “a few seconds,” not “hours”)
- The aura followed by the onset of headache within 60 minutes
- At least 1 aura symptom is unilateral.
Visual blurring, floaters, or split-second flashes before or during a migraine headache do not meet the criteria for aura.
MIGRAINE IS COMMON AND UNDERRECOGNIZED
In a study of 1,203 patients seeking care from a primary care provider for headache,6 94% of the 377 who turned in a diary with enough data to make a diagnosis were diagnosed with a migraine or probable migraine by an expert panel. A quarter of patients who likely had migraine based on an expert review of symptoms did not receive a migraine diagnosis at the time of their office visit.
Similarly, in a large epidemiologic study,7 30,758 adults were asked if they had headaches and, if so, how they named them. Headaches were reported by 23,564 of the participants and were subsequently diagnosed by formal ICHD criteria. Of the 3,074 individuals who met the criteria for migraine, only 53.4% correctly recognized their headaches as migraine. The most common erroneous labels were “sinus headache” and “stress headache.”7
HOW ESTROGEN AFFECTS MIGRAINE
Of note, migraine can be exacerbated during times of cycle irregularity, such as adolescence and perimenopause, the 2 times during a woman’s life associated with the highest risk of unintended pregnancy.10,11
STROKE RISK: ESTROGEN DOSE MATTERS
Shortly after the first combined oral contraceptives were released, reports of adverse events began to appear, although serious events were relatively rare. In response, prescribing guidelines advised against giving oral contraceptives to women with a history of deep vein thrombosis, myocardial infarction, stroke, or hypertension. Also, over the years, the hormonal content of the formulations was successively reduced, and with each reduction in estrogen, a decrease was observed in venous thrombosis and pulmonary embolism.12,13 Current low-dose formulations are considerably safer than high-dose options but are not entirely without risk.14
Stroke risk with combined oral contraceptives was first highlighted in a landmark article in 1975.15 However, the authors were unable to correlate the risk with the estrogen concentration of the pill, since 23 of the 25 women who suffered thrombotic stroke while taking the mestranol-containing formulation took 100-μg pills, and all 20 women who had strokes while taking the ethinyl estradiol formulation took 50-μg pills. Thus, by today’s standards, they were all taking high-dose pills. The risk of thrombotic stroke was 4 to 5 times higher in users than in nonusers.
In 1996, a study from the World Health Organization16 reported an increased risk of stroke with high-dose combined oral contraceptives (odds ratio [OR] 5.30, 95% confidence interval [CI] 2.56–11.0). With preparations containing less than 50 μg of ethinyl estradiol, the risk was not statistically significant (OR 1.53, 95% CI 0.71–3.31). These numbers were for Europe only; in developing countries, the risk was elevated regardless of dose, presumably due to additional risk factors in combined oral contraceptive users. The majority of strokes were in smokers taking 50-μg pills, with an average age greater than 35.
In 2002, a 5-year case-control study in Denmark found that the risk of stroke with combined oral contraceptives correlated directly with the estrogen content, from no increased risk with the newest and lowest-dose formulation (containing ethinyl estradiol 20 µg) to an OR of 4.5 with the older high-dose (50 µg) formulations.17
Reassuringly, a 2012 retrospective review of the Danish national registry13 revealed a low absolute risk of arterial events in users of combined oral contraceptives: 21.4 per 100,000 person-years for thrombotic stroke, and 10.1 per 100,000 person-years for myocardial infarction. Further, these risks were substantially lower with 20-μg ethinyl estradiol products than with those containing 30 to 40 μg.13 An important limitation of this large database review is that it did not control for important stroke risk factors such as obesity and smoking.
Although international studies14,16 continue to show a small but increased risk, more than 30 years have passed since a US study found an increased risk of stroke with combined oral contraceptives.
The discrepancy between US and international studies is possibly explained by the strong relative contraindication in the United States to the use of combined oral contraceptives in smokers over the age of 35 and the more prevalent use of high-dose pills in international studies. High-dose pills had been used in most of the stroke cases in the 1996 World Health Organization study16 but were used by only 0.7% of the women in the case and control groups in 2 pooled US studies from the same time period.18 Similarly, in these US studies, only 17% of the women were smokers on combined oral contraceptives, whereas in the international study, 51% of the women who had strokes and 38% of those in the control groups were smokers.
A large US study19 reviewing 3.6 million woman-years of use found no increased stroke risk (OR 0.96) in current users of low-dose combined oral contraceptives, results similar to those of a pooled analysis of US studies.18 Though this pooled analysis showed an adjusted increased risk of ischemic stroke in women reporting a history of migraine (OR 2.08, 95% CI 1.19–3.65), these conclusions were based on only 4 cases. The prevalence of migraine was identical in women who did or did not have strokes, 7.8% vs 7.7%, respectively, but the risk was judged to be increased after adjusting for other factors. But one important factor was not adjusted for: only 11 of the 1,017 women in the case and control groups were using 50-μg ethinyl estradiol pills, and 4 of the strokes were in this group of 11 women.
STROKE RISK INCREASES WITH FREQUENCY OF MIGRAINE AURA
Use of combined hormonal contraceptives in women who have migraine with aura remains controversial, based on good evidence that aura increases stroke risk20 and good evidence that high-dose oral contraceptives increase stroke risk.15
A cohort study encompassing more than 470,000 person-years with a median follow-up of 26 years found that while migraine without aura conferred no increase in risk of all-cause mortality, migraine with aura did.21
The longitudinal Women’s Health Study analyzed data from 27,798 women over age 45 and found that migraine with aura conferred an increased risk of cardiovascular disease (including stroke) that varied directly with aura frequency.22 Aura frequency less than once a month conferred a risk 2 times higher than in women without migraine, and the risk was more than 4 times higher when aura frequency exceeded once a week.
Similarly, an analysis of the World Health Organization study of stroke in young women found that the adjusted risk of ischemic stroke was significantly and directly associated with aura frequency.20
Potential explanations for this increased risk with greater aura frequency include changes induced during spreading cortical depression, shared genetic predispositions, and common underlying comorbidities such as patent foramen ovale.23–26
Though studies have shown that combined oral contraceptives in continuous regimens27 or in regimens that minimize drops in estrogen levels28 can help improve general headache and menstrual-related migraine, these studies have excluded patients who have migraine with aura.
In a pilot study,29 28 women referred to a tertiary headache clinic who had migraine with aura and intractable menstrual-related migraine were offered combined hormonal contraception in the form of a vaginal ring that releases only 15 μg ethinyl estradiol per 24 hours, thereby reducing peak estrogen exposure to a level lower than those encountered with the native menstrual cycle (with the suppression of ovulation). The women used this continuous ultra-low-dose hormonal contraception without placebo days. After a mean follow-up of 8 months, this regimen reduced aura frequency from a baseline average of 3.2 per month to only 0.2 per month. No woman had an increase in aura frequency, and menstrual-related migraine was eliminated in 21 (91.3%) of the 23 evaluable patients.
CHOOSING THE OPTIMAL CONTRACEPTIVE FORMULATION
Today, ultra-low-dose combined oral contraceptives (containing 10–15 µg of ethinyl estradiol) inhibit ovulation with doses of estrogen that are in a midphysiologic range. Consequently, they expose women to lower peak concentrations of estrogen than they would experience in their natural menstrual cycle (Figure 1). If a combined oral contraceptive is used in women with migraine with aura, lower estrogen doses (≤ 20 µg ethinyl estradiol) are preferred to decrease aura frequency and minimize the risk of stroke associated with high-dose ethinyl estradiol formulations.
Does the progestin matter?
Though there has been debate about whether different types of progestins alter the risk of venous thromboembolism,30,31 the chosen progestin does not seem to affect arterial risks such as stroke and myocardial infarction.14
All current guidelines note that progestin-only pills can be safely offered to women with migraine with aura. However, progestin-only pills have a shorter half-life than combined hormonal contraceptives and must be taken consistently and on time to ensure contraceptive efficacy and minimize abnormal bleeding. Patients who cannot adhere to a strict daily pill regimen may increase their risk of unintended pregnancy. In addition, progestin-only pills do not help with reducing episodes of migraine because they prevent ovulation only about half of the time.2 In contrast, a progestin-only arm implant is not only considered safe to use in women with migraine with aura, it may also prevent ovulation more reliably. Though progestin arm implants have the potential to reduce menstrual migraine and aura, this requires further study to confirm.
For menstrual-related migraine
In clinical practice, providers may offer certain combined hormonal contraceptives to women with debilitating menstrual-related migraine to prevent attacks. Although menstrual-related migraine rarely if ever is accompanied by aura, these patients may still have migraine with aura at other times of the month.
In women with menstrual-related migraine, any decrease in estrogen level greater than 10 µg of ethinyl estradiol may trigger an estrogen-withdrawal migraine. All currently available regimens of combined hormonal contraceptives that follow a 21-days-on, 7-days-off plan entail a drop in ethinyl estradiol of more than 10 µg (Figure 1).
Continuous regimens: Who needs a menstrual cycle anyway?
Of note: ultra-low-estrogen combined hormonal contraceptives that have placebo intervals may not inhibit ovulation consistently in all women.32 Contraceptive efficacy is still maintained, as contraception does not require inhibition of ovulation. Other mechanisms such as thickening of cervical mucus help with pregnancy prevention.
However, if ovulation is not inhibited, the consequent postovulatory decline in estrogen will continue to contribute to estrogen-withdrawal migraine.33,34 Reducing the number of placebo days may help inhibit ovulation. Adding back adequate estrogen during the placebo break (eg, either 0.9 mg conjugated equine estrogen with a 20-µg ethinyl estradiol combined oral contraceptive, or 0.075 mg transdermal 17B estradiol with a 15-µg combined hormonal contraceptive) can prevent these migraines.33,34
Some extended-cycle regimens, which give 4 withdrawal bleeds per year, will likewise prevent estrogen-withdrawal migraine if the decline in estrogen is limited to 10 µg (Table 1). Unfortunately, most extended regimens (Seasonale, Seasonique, and their generics) entail a 20- or 30-µg drop.
Continuous or extended-cycle regimens can be prescribed using any generic 20-µg combined hormonal contraceptive that the patient tolerates, along with specific instructions on the prescription to take the pills in a continuous fashion, eg, “Do not take the placebo pills; start the next pill pack immediately after 21 days.”
Postmenopausal hormone therapy
Neither smoking nor migraine is a contraindication to the use of postmenopausal hormone therapy, which is substantially lower in dosage than combined hormonal contraceptives.
ADVISING PATIENTS ON RISKS VS BENEFITS
It is important to remember that the risks of unintended pregnancy are always greater than the risks of any contraceptive, especially in women with chronic medical conditions, including those who have migraine with aura. Other benefits include the following:
Lower mortality risk. A 2010 analysis demonstrated that in nearly 46,000 women followed since 1968, those taking combined oral contraceptives had statistically significantly lower death rates from any cause and a lower risk of death from cancer and cardiovascular diseases than women who had never taken combined oral contraceptives.36
Stroke. Though the absolute risk of stroke to an individual woman taking a low-dose or ultra-low-dose combined hormonal contraceptive has been shown to be similar to that in women who are not taking combined hormonal contraception, its impact on an otherwise healthy woman could be devastating. Clinicians must remember that current guidelines still caution against prescribing combined hormonal contraceptives in women with migraine with aura and thus should counsel their patients accordingly and document the discussion in the medical record.
Noncontraceptive benefits. Women may be prescribed a combined hormonal contraceptive for benefits beyond contraception. The obvious reasons include beneficial effects on endometriosis, anemia, acne, hirsutism, dysmenorrhea, and prevention of ovarian cysts. But other important major benefits2 include substantial reductions in the risk of ovarian cancer (> 50% decrease after 10 years)37 and endometrial cancer (additional 24% reduction for each 5 years of use),38 and a modest decrease in the risk of colon cancer (37% less risk in ever-users).39 Further, combined oral contraceptive use has been associated with a decrease in mortality rates,40,41 with no increased risk of nonreproductive cancers.41
Ultra-low-dose, continuous formulations may benefit women by decreasing the frequency of migraine with aura and menstrual-related migraine. There is no evidence that reducing aura frequency also reduces stroke risk, but this represents an important area for future research.
WHAT WOULD WE DO?
For a patient who has a history of migraine with aura, if the goal is only to prevent pregnancy, we would recommend another contraceptive option that does not involve estrogen. However, we would consider prescribing a combined hormonal contraceptive in a low-dose regimen if the patient prefers this regimen for other health benefits (eg, acne control), if she has no other risk factors for stroke, and if she gives her informed consent after a discussion of the risks and benefits. Women who have menstrual-related migraine refractory to or who cannot tolerate other migraine therapies are often willing to try a low-dose estrogen-containing contraceptive for control of their migraine, especially if they have tried it in the past and believe that it helped prevent migraine. Patients should have follow-up within 3 months to discuss whether they have benefited from the regimen in terms of headache frequency or severity.
- ACOG Practice Bulletin No. 110: noncontraceptive uses of hormonal contraceptives. Obstet Gynecol 2010; 115:206–218.
- Centers for Disease Control and Prevention. US Medical Eligibility Criteria for Contraceptive Use, 2016. MMWR Recommendations and reports: Morbidity and mortality weekly report Recommendations and reports/Centers for Disease Control 2016; 65:1–104.
- Headache Classification Committee of the International Headache Society (IHS). The international classification of headache disorders, 3rd edition (beta version). Cephalalgia 2013; 33:629–808.
- Lipton RB, Cady RK, Stewart WF, Wilks K, Hall C. Diagnostic lessons from the Spectrum study. Neurology 2002; 58(suppl 6):S27–S31.
- Lipton RB, Stewart WF, Cady R, et al. 2000 Wolfe Award. Sumatriptan for the range of headaches in migraine sufferers: results of the Spectrum Study. Headache 2000; 40:783–791.
- Tepper SJ, Dahlof CG, Dowson A, et al. Prevalence and diagnosis of migraine in patients consulting their physician with a complaint of headache: data from the Landmark Study. Headache 2004; 44:856–864.
- Lipton RB, Stewart WF, Liberman JN. Self-awareness of migraine: interpreting the labels that headache sufferers apply to their headaches. Neurology 2002; 58(suppl 6):S21–S26.
- Chai NC, Peterlin BL, Calhoun AH. Migraine and estrogen. Curr Opin Neurol 2014; 27:315–324.
- Calhoun AH. Menstrual migraine: update on pathophysiology and approach to therapy and management. Curr Treat Options Neurol 2012; 14:1–14.
- McNamara M, Batur P, DeSapri KT. In the clinic. Perimenopause. Ann Intern Med 2015; 162:ITC1–ITC15.
- O’Brien HL, Cohen JM. Young adults with headaches: the transition from adolescents to adults. Headache 2015; 55:1404–1409.
- Vessey M, Mant D, Smith A, Yeates D. Oral contraceptives and venous thromboembolism: findings in a large prospective study. Br Med J (Clin Res Ed) 1986; 292:526.
- Lidegaard O, Lokkegaard E, Jensen A, Skovlund CW, Keiding N. Thrombotic stroke and myocardial infarction with hormonal contraception. N Engl J Med 2012; 366:2257–2266.
- MacGregor EA. Contraception and headache. Headache 2013; 53:247–276.
- Oral contraceptives and stroke in young women. Associated risk factors. JAMA 1975; 231:718–722.
- Ischaemic stroke and combined oral contraceptives: results of an international, multicentre, case-control study. WHO Collaborative Study of Cardiovascular Disease and Steroid Hormone Contraception. Lancet 1996; 348:498–505.
- Lidegaard O, Kreiner S. Contraceptives and cerebral thrombosis: a five-year national case-control study. Contraception 2002; 65:197–205.
- Schwartz SM, Petitti DB, Siscovick DS, et al. Stroke and use of low-dose oral contraceptives in young women: a pooled analysis of two US studies. Stroke 1998; 29:2277–2284.
- Petitti DB, Sidney S, Bernstein A, Wolf S, Quesenberry C, Ziel HK. Stroke in users of low-dose oral contraceptives. N Engl J Med 1996; 335:8–15.
- Donaghy M, Chang CL, Poulter N; European Collaborators of the World Health Organisation Collaborative Study of Cardiovascular Disease and Steroid Hormone Contraception. Duration, frequency, recency, and type of migraine and the risk of ischaemic stroke in women of childbearing age. J Neurol Neurosurg Psychiatry 2002; 73:747–750.
- Gudmundsson LS, Scher AI, Aspelund T, et al. Migraine with aura and risk of cardiovascular and all cause mortality in men and women: prospective cohort study. BMJ 2010; 341:c3966.
- Kurth T, Slomke MA, Kase CS, et al. Migraine, headache, and the risk of stroke in women: a prospective study. Neurology 2005; 64:1020–1026.
- Lee ST, Chu K, Jung KH, et al. Decreased number and function of endothelial progenitor cells in patients with migraine. Neurology 2008; 70:1510–1517.
- Kunz GA, Liang G, Cuculi F, et al. Circulating endothelial progenitor cells predict coronary artery disease severity. Am Heart J 2006; 152:190–195.
- Kurth T, Gaziano JM, Cook NR, Logroscino G, Diener HC, Buring JE. Migraine and risk of cardiovascular disease in women. JAMA 2006; 296:283–291.
- Pezzini A, Del Zotto E, Giossi A, Volonghi I, Grassi M, Padovani A. The migraine-ischemic stroke connection: potential pathogenic mechanisms. Curr Mol Med 2009; 9:215–226.
- Sulak P, Willis S, Kuehl T, Coffee A, Clark J. Headaches and oral contraceptives: impact of eliminating the standard 7-day placebo interval. Headache 2007; 47:27–37.
- Nappi RE, Terreno E, Sances G, et al. Effect of a contraceptive pill containing estradiol valerate and dienogest (E2V/DNG) in women with menstrually-related migraine (MRM). Contraception 2013; 88:369–375.
- Calhoun A, Ford S, Pruitt A. The impact of extended-cycle vaginal ring contraception on migraine aura: a retrospective case series. Headache 2012; 52:1246–1253.
- Wu CQ, Grandi SM, Filion KB, Abenhaim HA, Joseph L, Eisenberg MJ. Drospirenone-containing oral contraceptive pills and the risk of venous and arterial thrombosis: a systematic review. BJOG 2013; 120:801–810.
- Dinger J, Bardenheuer K, Heinemann K. Cardiovascular and general safety of a 24-day regimen of drospirenone-containing combined oral contraceptives: final results from the International Active Surveillance Study of Women Taking Oral Contraceptives. Contraception 2014; 89:253–263.
- Benson LS, Micks EA. Why stop now? Extended and continuous regimens of combined hormonal contraceptive methods. Obstet Gynecol Clin North Am 2015; 42:669–681.
- Mannix LK, Calhoun AH. Menstrual migraine. Curr Treat Options Neurol 2004; 6:489–498.
- Calhoun AH. A novel specific prophylaxis for menstrual-associated migraine. South Med J 2004; 97:819–822.
- Calhoun AH. Current topics and controversies in menstrual migraine. Headache 2012; 52(suppl 1):8–11.
- Hannaford PC, Iversen L, Macfarlane TV, Elliott AM, Angus V, Lee AJ. Mortality among contraceptive pill users: cohort evidence from Royal College of General Practitioners’ Oral Contraception Study. BMJ 2010; 340:c927.
- Havrilesky LJ, Moorman PG, Lowery WJ, et al. Oral contraceptive pills as primary prevention for ovarian cancer: a systematic review and meta-analysis. Obstet Gynecol 2013; 122:139 -147.
- Collaborative Group on Epidemiological Studies on Endometrial Cancer. Endometrial cancer and oral contraceptives: an individual participant meta-analysis of 27,276 women with endometrial cancer from 36 epidemiological studies. Lancet Oncol 2015; 16:1061–1070.
- Fernandez E, La Vecchia C, Franceschi S, et al. Oral contraceptive use and risk of colorectal cancer. Epidemiology 1998; 9:295–300.
- Merritt MA, Riboli E, Murphy N, et al. Reproductive factors and risk of mortality in the European Prospective Investigation into Cancer and Nutrition; a cohort study. BMC Med 2015; 13:252.
- Vessey M, Yeates D. Oral contraceptive use and cancer: final report from the Oxford-Family Planning Association Contraceptive Study. Contraception 2013; 88:678–683.
Combined hormonal contraceptives are contraindicated in women who have migraine with aura because they pose a risk of stroke. But how great is the risk, and how strong is the evidence, particularly with today’s low-dose contraceptives? Can we view migraine with aura as a relative contraindication rather than an absolute one?
This article reviews migraine diagnosis, the effects of estrogen and the menstrual cycle on migraine, the evidence of stroke risk with combined hormonal contraceptive use, and how the frequency of aura may affect risk. It offers practical advice on choosing contraceptive formulations and counseling patients on risks and benefits.
WHAT THE GUIDELINES SAY
Current guidelines restrict the use of combined hormonal contraceptives in the setting of migraine with aura, but not in migraine without aura.
A practice bulletin from the American College of Obstetrics and Gynecology in 2010 noted that extended-cycle or continuous hormonal contraceptives, including oral and parenteral products, might provide relief of migraines by eliminating the drops in estrogen levels that precipitate them.1 However, the bulletin also cautioned that though cerebrovascular accidents in women are rare, the impact of a stroke is so devastating that clinicians should consider intrauterine devices, progestin-only options, and other nonestrogen methods in women who have migraine with focal neurologic signs, women who smoke, and women age 35 or older.1
In 2016, the US Centers for Disease Control and Prevention published updates to its medical eligibility criteria for contraceptive use in various medical conditions. In the case of migraine without aura, the guidelines note no limitation to the use of combined hormonal contraceptives, regardless of the patient’s age. In the case of migraine with aura, the consensus was that the risk associated with combined hormonal contraception typically outweighs its benefits, noting “an unacceptable health risk if the contraceptive method is used.”2
We believe a fresh look at the data is warranted.
EARLY ORAL CONTRACEPTIVES WERE ALL HIGH-DOSE
This issue first surfaced in the decade and a half after the initial launch of oral contraceptives in 1960. The products then were all high-dose pills, containing up to 150 µg of mestranol. In subsequent decades, the dose of estrogen was successively reduced, so that now some pills contain only 10 µg of ethinyl estradiol. High-dose pills—which today contain 50 µg of ethinyl estradiol—account for less than 1% of pills currently sold in the United States and have been eliminated in many countries.
DIAGNOSTIC CRITERIA FOR MIGRAINE
According to the International Classification of Headache Disorders (ICHD),3 the diagnosis of migraine requires 2 of the 4 following criteria:
- Unilateral location
- Pulsating or throbbing pain
- Pain of at least moderate intensity
- Pain aggravated by activity, or causing a preference to avoid activity.
An additional criterion is either nausea or a combination of photophobia and phonophobia with the episode. This criterion can be met if the patient prefers to avoid bright lights and loud noises during an attack.
Headache experts have suggested that patients with a stable pattern of episodic, disabling headache and normal findings on physical examination should be considered to have migraine if there is no contradictory evidence.4,5
Migraine with aura requires at least 2 of the following 4 characteristics3:
- 1 aura symptom, spreading gradually over 5 minutes, or 2 or more aura symptoms occurring in succession, or both
- Each aura symptom lasting 5 to 60 minutes (not “a few seconds,” not “hours”)
- The aura followed by the onset of headache within 60 minutes
- At least 1 aura symptom is unilateral.
Visual blurring, floaters, or split-second flashes before or during a migraine headache do not meet the criteria for aura.
MIGRAINE IS COMMON AND UNDERRECOGNIZED
In a study of 1,203 patients seeking care from a primary care provider for headache,6 94% of the 377 who turned in a diary with enough data to make a diagnosis were diagnosed with a migraine or probable migraine by an expert panel. A quarter of patients who likely had migraine based on an expert review of symptoms did not receive a migraine diagnosis at the time of their office visit.
Similarly, in a large epidemiologic study,7 30,758 adults were asked if they had headaches and, if so, how they named them. Headaches were reported by 23,564 of the participants and were subsequently diagnosed by formal ICHD criteria. Of the 3,074 individuals who met the criteria for migraine, only 53.4% correctly recognized their headaches as migraine. The most common erroneous labels were “sinus headache” and “stress headache.”7
HOW ESTROGEN AFFECTS MIGRAINE
Of note, migraine can be exacerbated during times of cycle irregularity, such as adolescence and perimenopause, the 2 times during a woman’s life associated with the highest risk of unintended pregnancy.10,11
STROKE RISK: ESTROGEN DOSE MATTERS
Shortly after the first combined oral contraceptives were released, reports of adverse events began to appear, although serious events were relatively rare. In response, prescribing guidelines advised against giving oral contraceptives to women with a history of deep vein thrombosis, myocardial infarction, stroke, or hypertension. Also, over the years, the hormonal content of the formulations was successively reduced, and with each reduction in estrogen, a decrease was observed in venous thrombosis and pulmonary embolism.12,13 Current low-dose formulations are considerably safer than high-dose options but are not entirely without risk.14
Stroke risk with combined oral contraceptives was first highlighted in a landmark article in 1975.15 However, the authors were unable to correlate the risk with the estrogen concentration of the pill, since 23 of the 25 women who suffered thrombotic stroke while taking the mestranol-containing formulation took 100-μg pills, and all 20 women who had strokes while taking the ethinyl estradiol formulation took 50-μg pills. Thus, by today’s standards, they were all taking high-dose pills. The risk of thrombotic stroke was 4 to 5 times higher in users than in nonusers.
In 1996, a study from the World Health Organization16 reported an increased risk of stroke with high-dose combined oral contraceptives (odds ratio [OR] 5.30, 95% confidence interval [CI] 2.56–11.0). With preparations containing less than 50 μg of ethinyl estradiol, the risk was not statistically significant (OR 1.53, 95% CI 0.71–3.31). These numbers were for Europe only; in developing countries, the risk was elevated regardless of dose, presumably due to additional risk factors in combined oral contraceptive users. The majority of strokes were in smokers taking 50-μg pills, with an average age greater than 35.
In 2002, a 5-year case-control study in Denmark found that the risk of stroke with combined oral contraceptives correlated directly with the estrogen content, from no increased risk with the newest and lowest-dose formulation (containing ethinyl estradiol 20 µg) to an OR of 4.5 with the older high-dose (50 µg) formulations.17
Reassuringly, a 2012 retrospective review of the Danish national registry13 revealed a low absolute risk of arterial events in users of combined oral contraceptives: 21.4 per 100,000 person-years for thrombotic stroke, and 10.1 per 100,000 person-years for myocardial infarction. Further, these risks were substantially lower with 20-μg ethinyl estradiol products than with those containing 30 to 40 μg.13 An important limitation of this large database review is that it did not control for important stroke risk factors such as obesity and smoking.
Although international studies14,16 continue to show a small but increased risk, more than 30 years have passed since a US study found an increased risk of stroke with combined oral contraceptives.
The discrepancy between US and international studies is possibly explained by the strong relative contraindication in the United States to the use of combined oral contraceptives in smokers over the age of 35 and the more prevalent use of high-dose pills in international studies. High-dose pills had been used in most of the stroke cases in the 1996 World Health Organization study16 but were used by only 0.7% of the women in the case and control groups in 2 pooled US studies from the same time period.18 Similarly, in these US studies, only 17% of the women were smokers on combined oral contraceptives, whereas in the international study, 51% of the women who had strokes and 38% of those in the control groups were smokers.
A large US study19 reviewing 3.6 million woman-years of use found no increased stroke risk (OR 0.96) in current users of low-dose combined oral contraceptives, results similar to those of a pooled analysis of US studies.18 Though this pooled analysis showed an adjusted increased risk of ischemic stroke in women reporting a history of migraine (OR 2.08, 95% CI 1.19–3.65), these conclusions were based on only 4 cases. The prevalence of migraine was identical in women who did or did not have strokes, 7.8% vs 7.7%, respectively, but the risk was judged to be increased after adjusting for other factors. But one important factor was not adjusted for: only 11 of the 1,017 women in the case and control groups were using 50-μg ethinyl estradiol pills, and 4 of the strokes were in this group of 11 women.
STROKE RISK INCREASES WITH FREQUENCY OF MIGRAINE AURA
Use of combined hormonal contraceptives in women who have migraine with aura remains controversial, based on good evidence that aura increases stroke risk20 and good evidence that high-dose oral contraceptives increase stroke risk.15
A cohort study encompassing more than 470,000 person-years with a median follow-up of 26 years found that while migraine without aura conferred no increase in risk of all-cause mortality, migraine with aura did.21
The longitudinal Women’s Health Study analyzed data from 27,798 women over age 45 and found that migraine with aura conferred an increased risk of cardiovascular disease (including stroke) that varied directly with aura frequency.22 Aura frequency less than once a month conferred a risk 2 times higher than in women without migraine, and the risk was more than 4 times higher when aura frequency exceeded once a week.
Similarly, an analysis of the World Health Organization study of stroke in young women found that the adjusted risk of ischemic stroke was significantly and directly associated with aura frequency.20
Potential explanations for this increased risk with greater aura frequency include changes induced during spreading cortical depression, shared genetic predispositions, and common underlying comorbidities such as patent foramen ovale.23–26
Though studies have shown that combined oral contraceptives in continuous regimens27 or in regimens that minimize drops in estrogen levels28 can help improve general headache and menstrual-related migraine, these studies have excluded patients who have migraine with aura.
In a pilot study,29 28 women referred to a tertiary headache clinic who had migraine with aura and intractable menstrual-related migraine were offered combined hormonal contraception in the form of a vaginal ring that releases only 15 μg ethinyl estradiol per 24 hours, thereby reducing peak estrogen exposure to a level lower than those encountered with the native menstrual cycle (with the suppression of ovulation). The women used this continuous ultra-low-dose hormonal contraception without placebo days. After a mean follow-up of 8 months, this regimen reduced aura frequency from a baseline average of 3.2 per month to only 0.2 per month. No woman had an increase in aura frequency, and menstrual-related migraine was eliminated in 21 (91.3%) of the 23 evaluable patients.
CHOOSING THE OPTIMAL CONTRACEPTIVE FORMULATION
Today, ultra-low-dose combined oral contraceptives (containing 10–15 µg of ethinyl estradiol) inhibit ovulation with doses of estrogen that are in a midphysiologic range. Consequently, they expose women to lower peak concentrations of estrogen than they would experience in their natural menstrual cycle (Figure 1). If a combined oral contraceptive is used in women with migraine with aura, lower estrogen doses (≤ 20 µg ethinyl estradiol) are preferred to decrease aura frequency and minimize the risk of stroke associated with high-dose ethinyl estradiol formulations.
Does the progestin matter?
Though there has been debate about whether different types of progestins alter the risk of venous thromboembolism,30,31 the chosen progestin does not seem to affect arterial risks such as stroke and myocardial infarction.14
All current guidelines note that progestin-only pills can be safely offered to women with migraine with aura. However, progestin-only pills have a shorter half-life than combined hormonal contraceptives and must be taken consistently and on time to ensure contraceptive efficacy and minimize abnormal bleeding. Patients who cannot adhere to a strict daily pill regimen may increase their risk of unintended pregnancy. In addition, progestin-only pills do not help with reducing episodes of migraine because they prevent ovulation only about half of the time.2 In contrast, a progestin-only arm implant is not only considered safe to use in women with migraine with aura, it may also prevent ovulation more reliably. Though progestin arm implants have the potential to reduce menstrual migraine and aura, this requires further study to confirm.
For menstrual-related migraine
In clinical practice, providers may offer certain combined hormonal contraceptives to women with debilitating menstrual-related migraine to prevent attacks. Although menstrual-related migraine rarely if ever is accompanied by aura, these patients may still have migraine with aura at other times of the month.
In women with menstrual-related migraine, any decrease in estrogen level greater than 10 µg of ethinyl estradiol may trigger an estrogen-withdrawal migraine. All currently available regimens of combined hormonal contraceptives that follow a 21-days-on, 7-days-off plan entail a drop in ethinyl estradiol of more than 10 µg (Figure 1).
Continuous regimens: Who needs a menstrual cycle anyway?
Of note: ultra-low-estrogen combined hormonal contraceptives that have placebo intervals may not inhibit ovulation consistently in all women.32 Contraceptive efficacy is still maintained, as contraception does not require inhibition of ovulation. Other mechanisms such as thickening of cervical mucus help with pregnancy prevention.
However, if ovulation is not inhibited, the consequent postovulatory decline in estrogen will continue to contribute to estrogen-withdrawal migraine.33,34 Reducing the number of placebo days may help inhibit ovulation. Adding back adequate estrogen during the placebo break (eg, either 0.9 mg conjugated equine estrogen with a 20-µg ethinyl estradiol combined oral contraceptive, or 0.075 mg transdermal 17B estradiol with a 15-µg combined hormonal contraceptive) can prevent these migraines.33,34
Some extended-cycle regimens, which give 4 withdrawal bleeds per year, will likewise prevent estrogen-withdrawal migraine if the decline in estrogen is limited to 10 µg (Table 1). Unfortunately, most extended regimens (Seasonale, Seasonique, and their generics) entail a 20- or 30-µg drop.
Continuous or extended-cycle regimens can be prescribed using any generic 20-µg combined hormonal contraceptive that the patient tolerates, along with specific instructions on the prescription to take the pills in a continuous fashion, eg, “Do not take the placebo pills; start the next pill pack immediately after 21 days.”
Postmenopausal hormone therapy
Neither smoking nor migraine is a contraindication to the use of postmenopausal hormone therapy, which is substantially lower in dosage than combined hormonal contraceptives.
ADVISING PATIENTS ON RISKS VS BENEFITS
It is important to remember that the risks of unintended pregnancy are always greater than the risks of any contraceptive, especially in women with chronic medical conditions, including those who have migraine with aura. Other benefits include the following:
Lower mortality risk. A 2010 analysis demonstrated that in nearly 46,000 women followed since 1968, those taking combined oral contraceptives had statistically significantly lower death rates from any cause and a lower risk of death from cancer and cardiovascular diseases than women who had never taken combined oral contraceptives.36
Stroke. Though the absolute risk of stroke to an individual woman taking a low-dose or ultra-low-dose combined hormonal contraceptive has been shown to be similar to that in women who are not taking combined hormonal contraception, its impact on an otherwise healthy woman could be devastating. Clinicians must remember that current guidelines still caution against prescribing combined hormonal contraceptives in women with migraine with aura and thus should counsel their patients accordingly and document the discussion in the medical record.
Noncontraceptive benefits. Women may be prescribed a combined hormonal contraceptive for benefits beyond contraception. The obvious reasons include beneficial effects on endometriosis, anemia, acne, hirsutism, dysmenorrhea, and prevention of ovarian cysts. But other important major benefits2 include substantial reductions in the risk of ovarian cancer (> 50% decrease after 10 years)37 and endometrial cancer (additional 24% reduction for each 5 years of use),38 and a modest decrease in the risk of colon cancer (37% less risk in ever-users).39 Further, combined oral contraceptive use has been associated with a decrease in mortality rates,40,41 with no increased risk of nonreproductive cancers.41
Ultra-low-dose, continuous formulations may benefit women by decreasing the frequency of migraine with aura and menstrual-related migraine. There is no evidence that reducing aura frequency also reduces stroke risk, but this represents an important area for future research.
WHAT WOULD WE DO?
For a patient who has a history of migraine with aura, if the goal is only to prevent pregnancy, we would recommend another contraceptive option that does not involve estrogen. However, we would consider prescribing a combined hormonal contraceptive in a low-dose regimen if the patient prefers this regimen for other health benefits (eg, acne control), if she has no other risk factors for stroke, and if she gives her informed consent after a discussion of the risks and benefits. Women who have menstrual-related migraine refractory to or who cannot tolerate other migraine therapies are often willing to try a low-dose estrogen-containing contraceptive for control of their migraine, especially if they have tried it in the past and believe that it helped prevent migraine. Patients should have follow-up within 3 months to discuss whether they have benefited from the regimen in terms of headache frequency or severity.
Combined hormonal contraceptives are contraindicated in women who have migraine with aura because they pose a risk of stroke. But how great is the risk, and how strong is the evidence, particularly with today’s low-dose contraceptives? Can we view migraine with aura as a relative contraindication rather than an absolute one?
This article reviews migraine diagnosis, the effects of estrogen and the menstrual cycle on migraine, the evidence of stroke risk with combined hormonal contraceptive use, and how the frequency of aura may affect risk. It offers practical advice on choosing contraceptive formulations and counseling patients on risks and benefits.
WHAT THE GUIDELINES SAY
Current guidelines restrict the use of combined hormonal contraceptives in the setting of migraine with aura, but not in migraine without aura.
A practice bulletin from the American College of Obstetrics and Gynecology in 2010 noted that extended-cycle or continuous hormonal contraceptives, including oral and parenteral products, might provide relief of migraines by eliminating the drops in estrogen levels that precipitate them.1 However, the bulletin also cautioned that though cerebrovascular accidents in women are rare, the impact of a stroke is so devastating that clinicians should consider intrauterine devices, progestin-only options, and other nonestrogen methods in women who have migraine with focal neurologic signs, women who smoke, and women age 35 or older.1
In 2016, the US Centers for Disease Control and Prevention published updates to its medical eligibility criteria for contraceptive use in various medical conditions. In the case of migraine without aura, the guidelines note no limitation to the use of combined hormonal contraceptives, regardless of the patient’s age. In the case of migraine with aura, the consensus was that the risk associated with combined hormonal contraception typically outweighs its benefits, noting “an unacceptable health risk if the contraceptive method is used.”2
We believe a fresh look at the data is warranted.
EARLY ORAL CONTRACEPTIVES WERE ALL HIGH-DOSE
This issue first surfaced in the decade and a half after the initial launch of oral contraceptives in 1960. The products then were all high-dose pills, containing up to 150 µg of mestranol. In subsequent decades, the dose of estrogen was successively reduced, so that now some pills contain only 10 µg of ethinyl estradiol. High-dose pills—which today contain 50 µg of ethinyl estradiol—account for less than 1% of pills currently sold in the United States and have been eliminated in many countries.
DIAGNOSTIC CRITERIA FOR MIGRAINE
According to the International Classification of Headache Disorders (ICHD),3 the diagnosis of migraine requires 2 of the 4 following criteria:
- Unilateral location
- Pulsating or throbbing pain
- Pain of at least moderate intensity
- Pain aggravated by activity, or causing a preference to avoid activity.
An additional criterion is either nausea or a combination of photophobia and phonophobia with the episode. This criterion can be met if the patient prefers to avoid bright lights and loud noises during an attack.
Headache experts have suggested that patients with a stable pattern of episodic, disabling headache and normal findings on physical examination should be considered to have migraine if there is no contradictory evidence.4,5
Migraine with aura requires at least 2 of the following 4 characteristics3:
- 1 aura symptom, spreading gradually over 5 minutes, or 2 or more aura symptoms occurring in succession, or both
- Each aura symptom lasting 5 to 60 minutes (not “a few seconds,” not “hours”)
- The aura followed by the onset of headache within 60 minutes
- At least 1 aura symptom is unilateral.
Visual blurring, floaters, or split-second flashes before or during a migraine headache do not meet the criteria for aura.
MIGRAINE IS COMMON AND UNDERRECOGNIZED
In a study of 1,203 patients seeking care from a primary care provider for headache,6 94% of the 377 who turned in a diary with enough data to make a diagnosis were diagnosed with a migraine or probable migraine by an expert panel. A quarter of patients who likely had migraine based on an expert review of symptoms did not receive a migraine diagnosis at the time of their office visit.
Similarly, in a large epidemiologic study,7 30,758 adults were asked if they had headaches and, if so, how they named them. Headaches were reported by 23,564 of the participants and were subsequently diagnosed by formal ICHD criteria. Of the 3,074 individuals who met the criteria for migraine, only 53.4% correctly recognized their headaches as migraine. The most common erroneous labels were “sinus headache” and “stress headache.”7
HOW ESTROGEN AFFECTS MIGRAINE
Of note, migraine can be exacerbated during times of cycle irregularity, such as adolescence and perimenopause, the 2 times during a woman’s life associated with the highest risk of unintended pregnancy.10,11
STROKE RISK: ESTROGEN DOSE MATTERS
Shortly after the first combined oral contraceptives were released, reports of adverse events began to appear, although serious events were relatively rare. In response, prescribing guidelines advised against giving oral contraceptives to women with a history of deep vein thrombosis, myocardial infarction, stroke, or hypertension. Also, over the years, the hormonal content of the formulations was successively reduced, and with each reduction in estrogen, a decrease was observed in venous thrombosis and pulmonary embolism.12,13 Current low-dose formulations are considerably safer than high-dose options but are not entirely without risk.14
Stroke risk with combined oral contraceptives was first highlighted in a landmark article in 1975.15 However, the authors were unable to correlate the risk with the estrogen concentration of the pill, since 23 of the 25 women who suffered thrombotic stroke while taking the mestranol-containing formulation took 100-μg pills, and all 20 women who had strokes while taking the ethinyl estradiol formulation took 50-μg pills. Thus, by today’s standards, they were all taking high-dose pills. The risk of thrombotic stroke was 4 to 5 times higher in users than in nonusers.
In 1996, a study from the World Health Organization16 reported an increased risk of stroke with high-dose combined oral contraceptives (odds ratio [OR] 5.30, 95% confidence interval [CI] 2.56–11.0). With preparations containing less than 50 μg of ethinyl estradiol, the risk was not statistically significant (OR 1.53, 95% CI 0.71–3.31). These numbers were for Europe only; in developing countries, the risk was elevated regardless of dose, presumably due to additional risk factors in combined oral contraceptive users. The majority of strokes were in smokers taking 50-μg pills, with an average age greater than 35.
In 2002, a 5-year case-control study in Denmark found that the risk of stroke with combined oral contraceptives correlated directly with the estrogen content, from no increased risk with the newest and lowest-dose formulation (containing ethinyl estradiol 20 µg) to an OR of 4.5 with the older high-dose (50 µg) formulations.17
Reassuringly, a 2012 retrospective review of the Danish national registry13 revealed a low absolute risk of arterial events in users of combined oral contraceptives: 21.4 per 100,000 person-years for thrombotic stroke, and 10.1 per 100,000 person-years for myocardial infarction. Further, these risks were substantially lower with 20-μg ethinyl estradiol products than with those containing 30 to 40 μg.13 An important limitation of this large database review is that it did not control for important stroke risk factors such as obesity and smoking.
Although international studies14,16 continue to show a small but increased risk, more than 30 years have passed since a US study found an increased risk of stroke with combined oral contraceptives.
The discrepancy between US and international studies is possibly explained by the strong relative contraindication in the United States to the use of combined oral contraceptives in smokers over the age of 35 and the more prevalent use of high-dose pills in international studies. High-dose pills had been used in most of the stroke cases in the 1996 World Health Organization study16 but were used by only 0.7% of the women in the case and control groups in 2 pooled US studies from the same time period.18 Similarly, in these US studies, only 17% of the women were smokers on combined oral contraceptives, whereas in the international study, 51% of the women who had strokes and 38% of those in the control groups were smokers.
A large US study19 reviewing 3.6 million woman-years of use found no increased stroke risk (OR 0.96) in current users of low-dose combined oral contraceptives, results similar to those of a pooled analysis of US studies.18 Though this pooled analysis showed an adjusted increased risk of ischemic stroke in women reporting a history of migraine (OR 2.08, 95% CI 1.19–3.65), these conclusions were based on only 4 cases. The prevalence of migraine was identical in women who did or did not have strokes, 7.8% vs 7.7%, respectively, but the risk was judged to be increased after adjusting for other factors. But one important factor was not adjusted for: only 11 of the 1,017 women in the case and control groups were using 50-μg ethinyl estradiol pills, and 4 of the strokes were in this group of 11 women.
STROKE RISK INCREASES WITH FREQUENCY OF MIGRAINE AURA
Use of combined hormonal contraceptives in women who have migraine with aura remains controversial, based on good evidence that aura increases stroke risk20 and good evidence that high-dose oral contraceptives increase stroke risk.15
A cohort study encompassing more than 470,000 person-years with a median follow-up of 26 years found that while migraine without aura conferred no increase in risk of all-cause mortality, migraine with aura did.21
The longitudinal Women’s Health Study analyzed data from 27,798 women over age 45 and found that migraine with aura conferred an increased risk of cardiovascular disease (including stroke) that varied directly with aura frequency.22 Aura frequency less than once a month conferred a risk 2 times higher than in women without migraine, and the risk was more than 4 times higher when aura frequency exceeded once a week.
Similarly, an analysis of the World Health Organization study of stroke in young women found that the adjusted risk of ischemic stroke was significantly and directly associated with aura frequency.20
Potential explanations for this increased risk with greater aura frequency include changes induced during spreading cortical depression, shared genetic predispositions, and common underlying comorbidities such as patent foramen ovale.23–26
Though studies have shown that combined oral contraceptives in continuous regimens27 or in regimens that minimize drops in estrogen levels28 can help improve general headache and menstrual-related migraine, these studies have excluded patients who have migraine with aura.
In a pilot study,29 28 women referred to a tertiary headache clinic who had migraine with aura and intractable menstrual-related migraine were offered combined hormonal contraception in the form of a vaginal ring that releases only 15 μg ethinyl estradiol per 24 hours, thereby reducing peak estrogen exposure to a level lower than those encountered with the native menstrual cycle (with the suppression of ovulation). The women used this continuous ultra-low-dose hormonal contraception without placebo days. After a mean follow-up of 8 months, this regimen reduced aura frequency from a baseline average of 3.2 per month to only 0.2 per month. No woman had an increase in aura frequency, and menstrual-related migraine was eliminated in 21 (91.3%) of the 23 evaluable patients.
CHOOSING THE OPTIMAL CONTRACEPTIVE FORMULATION
Today, ultra-low-dose combined oral contraceptives (containing 10–15 µg of ethinyl estradiol) inhibit ovulation with doses of estrogen that are in a midphysiologic range. Consequently, they expose women to lower peak concentrations of estrogen than they would experience in their natural menstrual cycle (Figure 1). If a combined oral contraceptive is used in women with migraine with aura, lower estrogen doses (≤ 20 µg ethinyl estradiol) are preferred to decrease aura frequency and minimize the risk of stroke associated with high-dose ethinyl estradiol formulations.
Does the progestin matter?
Though there has been debate about whether different types of progestins alter the risk of venous thromboembolism,30,31 the chosen progestin does not seem to affect arterial risks such as stroke and myocardial infarction.14
All current guidelines note that progestin-only pills can be safely offered to women with migraine with aura. However, progestin-only pills have a shorter half-life than combined hormonal contraceptives and must be taken consistently and on time to ensure contraceptive efficacy and minimize abnormal bleeding. Patients who cannot adhere to a strict daily pill regimen may increase their risk of unintended pregnancy. In addition, progestin-only pills do not help with reducing episodes of migraine because they prevent ovulation only about half of the time.2 In contrast, a progestin-only arm implant is not only considered safe to use in women with migraine with aura, it may also prevent ovulation more reliably. Though progestin arm implants have the potential to reduce menstrual migraine and aura, this requires further study to confirm.
For menstrual-related migraine
In clinical practice, providers may offer certain combined hormonal contraceptives to women with debilitating menstrual-related migraine to prevent attacks. Although menstrual-related migraine rarely if ever is accompanied by aura, these patients may still have migraine with aura at other times of the month.
In women with menstrual-related migraine, any decrease in estrogen level greater than 10 µg of ethinyl estradiol may trigger an estrogen-withdrawal migraine. All currently available regimens of combined hormonal contraceptives that follow a 21-days-on, 7-days-off plan entail a drop in ethinyl estradiol of more than 10 µg (Figure 1).
Continuous regimens: Who needs a menstrual cycle anyway?
Of note: ultra-low-estrogen combined hormonal contraceptives that have placebo intervals may not inhibit ovulation consistently in all women.32 Contraceptive efficacy is still maintained, as contraception does not require inhibition of ovulation. Other mechanisms such as thickening of cervical mucus help with pregnancy prevention.
However, if ovulation is not inhibited, the consequent postovulatory decline in estrogen will continue to contribute to estrogen-withdrawal migraine.33,34 Reducing the number of placebo days may help inhibit ovulation. Adding back adequate estrogen during the placebo break (eg, either 0.9 mg conjugated equine estrogen with a 20-µg ethinyl estradiol combined oral contraceptive, or 0.075 mg transdermal 17B estradiol with a 15-µg combined hormonal contraceptive) can prevent these migraines.33,34
Some extended-cycle regimens, which give 4 withdrawal bleeds per year, will likewise prevent estrogen-withdrawal migraine if the decline in estrogen is limited to 10 µg (Table 1). Unfortunately, most extended regimens (Seasonale, Seasonique, and their generics) entail a 20- or 30-µg drop.
Continuous or extended-cycle regimens can be prescribed using any generic 20-µg combined hormonal contraceptive that the patient tolerates, along with specific instructions on the prescription to take the pills in a continuous fashion, eg, “Do not take the placebo pills; start the next pill pack immediately after 21 days.”
Postmenopausal hormone therapy
Neither smoking nor migraine is a contraindication to the use of postmenopausal hormone therapy, which is substantially lower in dosage than combined hormonal contraceptives.
ADVISING PATIENTS ON RISKS VS BENEFITS
It is important to remember that the risks of unintended pregnancy are always greater than the risks of any contraceptive, especially in women with chronic medical conditions, including those who have migraine with aura. Other benefits include the following:
Lower mortality risk. A 2010 analysis demonstrated that in nearly 46,000 women followed since 1968, those taking combined oral contraceptives had statistically significantly lower death rates from any cause and a lower risk of death from cancer and cardiovascular diseases than women who had never taken combined oral contraceptives.36
Stroke. Though the absolute risk of stroke to an individual woman taking a low-dose or ultra-low-dose combined hormonal contraceptive has been shown to be similar to that in women who are not taking combined hormonal contraception, its impact on an otherwise healthy woman could be devastating. Clinicians must remember that current guidelines still caution against prescribing combined hormonal contraceptives in women with migraine with aura and thus should counsel their patients accordingly and document the discussion in the medical record.
Noncontraceptive benefits. Women may be prescribed a combined hormonal contraceptive for benefits beyond contraception. The obvious reasons include beneficial effects on endometriosis, anemia, acne, hirsutism, dysmenorrhea, and prevention of ovarian cysts. But other important major benefits2 include substantial reductions in the risk of ovarian cancer (> 50% decrease after 10 years)37 and endometrial cancer (additional 24% reduction for each 5 years of use),38 and a modest decrease in the risk of colon cancer (37% less risk in ever-users).39 Further, combined oral contraceptive use has been associated with a decrease in mortality rates,40,41 with no increased risk of nonreproductive cancers.41
Ultra-low-dose, continuous formulations may benefit women by decreasing the frequency of migraine with aura and menstrual-related migraine. There is no evidence that reducing aura frequency also reduces stroke risk, but this represents an important area for future research.
WHAT WOULD WE DO?
For a patient who has a history of migraine with aura, if the goal is only to prevent pregnancy, we would recommend another contraceptive option that does not involve estrogen. However, we would consider prescribing a combined hormonal contraceptive in a low-dose regimen if the patient prefers this regimen for other health benefits (eg, acne control), if she has no other risk factors for stroke, and if she gives her informed consent after a discussion of the risks and benefits. Women who have menstrual-related migraine refractory to or who cannot tolerate other migraine therapies are often willing to try a low-dose estrogen-containing contraceptive for control of their migraine, especially if they have tried it in the past and believe that it helped prevent migraine. Patients should have follow-up within 3 months to discuss whether they have benefited from the regimen in terms of headache frequency or severity.
- ACOG Practice Bulletin No. 110: noncontraceptive uses of hormonal contraceptives. Obstet Gynecol 2010; 115:206–218.
- Centers for Disease Control and Prevention. US Medical Eligibility Criteria for Contraceptive Use, 2016. MMWR Recommendations and reports: Morbidity and mortality weekly report Recommendations and reports/Centers for Disease Control 2016; 65:1–104.
- Headache Classification Committee of the International Headache Society (IHS). The international classification of headache disorders, 3rd edition (beta version). Cephalalgia 2013; 33:629–808.
- Lipton RB, Cady RK, Stewart WF, Wilks K, Hall C. Diagnostic lessons from the Spectrum study. Neurology 2002; 58(suppl 6):S27–S31.
- Lipton RB, Stewart WF, Cady R, et al. 2000 Wolfe Award. Sumatriptan for the range of headaches in migraine sufferers: results of the Spectrum Study. Headache 2000; 40:783–791.
- Tepper SJ, Dahlof CG, Dowson A, et al. Prevalence and diagnosis of migraine in patients consulting their physician with a complaint of headache: data from the Landmark Study. Headache 2004; 44:856–864.
- Lipton RB, Stewart WF, Liberman JN. Self-awareness of migraine: interpreting the labels that headache sufferers apply to their headaches. Neurology 2002; 58(suppl 6):S21–S26.
- Chai NC, Peterlin BL, Calhoun AH. Migraine and estrogen. Curr Opin Neurol 2014; 27:315–324.
- Calhoun AH. Menstrual migraine: update on pathophysiology and approach to therapy and management. Curr Treat Options Neurol 2012; 14:1–14.
- McNamara M, Batur P, DeSapri KT. In the clinic. Perimenopause. Ann Intern Med 2015; 162:ITC1–ITC15.
- O’Brien HL, Cohen JM. Young adults with headaches: the transition from adolescents to adults. Headache 2015; 55:1404–1409.
- Vessey M, Mant D, Smith A, Yeates D. Oral contraceptives and venous thromboembolism: findings in a large prospective study. Br Med J (Clin Res Ed) 1986; 292:526.
- Lidegaard O, Lokkegaard E, Jensen A, Skovlund CW, Keiding N. Thrombotic stroke and myocardial infarction with hormonal contraception. N Engl J Med 2012; 366:2257–2266.
- MacGregor EA. Contraception and headache. Headache 2013; 53:247–276.
- Oral contraceptives and stroke in young women. Associated risk factors. JAMA 1975; 231:718–722.
- Ischaemic stroke and combined oral contraceptives: results of an international, multicentre, case-control study. WHO Collaborative Study of Cardiovascular Disease and Steroid Hormone Contraception. Lancet 1996; 348:498–505.
- Lidegaard O, Kreiner S. Contraceptives and cerebral thrombosis: a five-year national case-control study. Contraception 2002; 65:197–205.
- Schwartz SM, Petitti DB, Siscovick DS, et al. Stroke and use of low-dose oral contraceptives in young women: a pooled analysis of two US studies. Stroke 1998; 29:2277–2284.
- Petitti DB, Sidney S, Bernstein A, Wolf S, Quesenberry C, Ziel HK. Stroke in users of low-dose oral contraceptives. N Engl J Med 1996; 335:8–15.
- Donaghy M, Chang CL, Poulter N; European Collaborators of the World Health Organisation Collaborative Study of Cardiovascular Disease and Steroid Hormone Contraception. Duration, frequency, recency, and type of migraine and the risk of ischaemic stroke in women of childbearing age. J Neurol Neurosurg Psychiatry 2002; 73:747–750.
- Gudmundsson LS, Scher AI, Aspelund T, et al. Migraine with aura and risk of cardiovascular and all cause mortality in men and women: prospective cohort study. BMJ 2010; 341:c3966.
- Kurth T, Slomke MA, Kase CS, et al. Migraine, headache, and the risk of stroke in women: a prospective study. Neurology 2005; 64:1020–1026.
- Lee ST, Chu K, Jung KH, et al. Decreased number and function of endothelial progenitor cells in patients with migraine. Neurology 2008; 70:1510–1517.
- Kunz GA, Liang G, Cuculi F, et al. Circulating endothelial progenitor cells predict coronary artery disease severity. Am Heart J 2006; 152:190–195.
- Kurth T, Gaziano JM, Cook NR, Logroscino G, Diener HC, Buring JE. Migraine and risk of cardiovascular disease in women. JAMA 2006; 296:283–291.
- Pezzini A, Del Zotto E, Giossi A, Volonghi I, Grassi M, Padovani A. The migraine-ischemic stroke connection: potential pathogenic mechanisms. Curr Mol Med 2009; 9:215–226.
- Sulak P, Willis S, Kuehl T, Coffee A, Clark J. Headaches and oral contraceptives: impact of eliminating the standard 7-day placebo interval. Headache 2007; 47:27–37.
- Nappi RE, Terreno E, Sances G, et al. Effect of a contraceptive pill containing estradiol valerate and dienogest (E2V/DNG) in women with menstrually-related migraine (MRM). Contraception 2013; 88:369–375.
- Calhoun A, Ford S, Pruitt A. The impact of extended-cycle vaginal ring contraception on migraine aura: a retrospective case series. Headache 2012; 52:1246–1253.
- Wu CQ, Grandi SM, Filion KB, Abenhaim HA, Joseph L, Eisenberg MJ. Drospirenone-containing oral contraceptive pills and the risk of venous and arterial thrombosis: a systematic review. BJOG 2013; 120:801–810.
- Dinger J, Bardenheuer K, Heinemann K. Cardiovascular and general safety of a 24-day regimen of drospirenone-containing combined oral contraceptives: final results from the International Active Surveillance Study of Women Taking Oral Contraceptives. Contraception 2014; 89:253–263.
- Benson LS, Micks EA. Why stop now? Extended and continuous regimens of combined hormonal contraceptive methods. Obstet Gynecol Clin North Am 2015; 42:669–681.
- Mannix LK, Calhoun AH. Menstrual migraine. Curr Treat Options Neurol 2004; 6:489–498.
- Calhoun AH. A novel specific prophylaxis for menstrual-associated migraine. South Med J 2004; 97:819–822.
- Calhoun AH. Current topics and controversies in menstrual migraine. Headache 2012; 52(suppl 1):8–11.
- Hannaford PC, Iversen L, Macfarlane TV, Elliott AM, Angus V, Lee AJ. Mortality among contraceptive pill users: cohort evidence from Royal College of General Practitioners’ Oral Contraception Study. BMJ 2010; 340:c927.
- Havrilesky LJ, Moorman PG, Lowery WJ, et al. Oral contraceptive pills as primary prevention for ovarian cancer: a systematic review and meta-analysis. Obstet Gynecol 2013; 122:139 -147.
- Collaborative Group on Epidemiological Studies on Endometrial Cancer. Endometrial cancer and oral contraceptives: an individual participant meta-analysis of 27,276 women with endometrial cancer from 36 epidemiological studies. Lancet Oncol 2015; 16:1061–1070.
- Fernandez E, La Vecchia C, Franceschi S, et al. Oral contraceptive use and risk of colorectal cancer. Epidemiology 1998; 9:295–300.
- Merritt MA, Riboli E, Murphy N, et al. Reproductive factors and risk of mortality in the European Prospective Investigation into Cancer and Nutrition; a cohort study. BMC Med 2015; 13:252.
- Vessey M, Yeates D. Oral contraceptive use and cancer: final report from the Oxford-Family Planning Association Contraceptive Study. Contraception 2013; 88:678–683.
- ACOG Practice Bulletin No. 110: noncontraceptive uses of hormonal contraceptives. Obstet Gynecol 2010; 115:206–218.
- Centers for Disease Control and Prevention. US Medical Eligibility Criteria for Contraceptive Use, 2016. MMWR Recommendations and reports: Morbidity and mortality weekly report Recommendations and reports/Centers for Disease Control 2016; 65:1–104.
- Headache Classification Committee of the International Headache Society (IHS). The international classification of headache disorders, 3rd edition (beta version). Cephalalgia 2013; 33:629–808.
- Lipton RB, Cady RK, Stewart WF, Wilks K, Hall C. Diagnostic lessons from the Spectrum study. Neurology 2002; 58(suppl 6):S27–S31.
- Lipton RB, Stewart WF, Cady R, et al. 2000 Wolfe Award. Sumatriptan for the range of headaches in migraine sufferers: results of the Spectrum Study. Headache 2000; 40:783–791.
- Tepper SJ, Dahlof CG, Dowson A, et al. Prevalence and diagnosis of migraine in patients consulting their physician with a complaint of headache: data from the Landmark Study. Headache 2004; 44:856–864.
- Lipton RB, Stewart WF, Liberman JN. Self-awareness of migraine: interpreting the labels that headache sufferers apply to their headaches. Neurology 2002; 58(suppl 6):S21–S26.
- Chai NC, Peterlin BL, Calhoun AH. Migraine and estrogen. Curr Opin Neurol 2014; 27:315–324.
- Calhoun AH. Menstrual migraine: update on pathophysiology and approach to therapy and management. Curr Treat Options Neurol 2012; 14:1–14.
- McNamara M, Batur P, DeSapri KT. In the clinic. Perimenopause. Ann Intern Med 2015; 162:ITC1–ITC15.
- O’Brien HL, Cohen JM. Young adults with headaches: the transition from adolescents to adults. Headache 2015; 55:1404–1409.
- Vessey M, Mant D, Smith A, Yeates D. Oral contraceptives and venous thromboembolism: findings in a large prospective study. Br Med J (Clin Res Ed) 1986; 292:526.
- Lidegaard O, Lokkegaard E, Jensen A, Skovlund CW, Keiding N. Thrombotic stroke and myocardial infarction with hormonal contraception. N Engl J Med 2012; 366:2257–2266.
- MacGregor EA. Contraception and headache. Headache 2013; 53:247–276.
- Oral contraceptives and stroke in young women. Associated risk factors. JAMA 1975; 231:718–722.
- Ischaemic stroke and combined oral contraceptives: results of an international, multicentre, case-control study. WHO Collaborative Study of Cardiovascular Disease and Steroid Hormone Contraception. Lancet 1996; 348:498–505.
- Lidegaard O, Kreiner S. Contraceptives and cerebral thrombosis: a five-year national case-control study. Contraception 2002; 65:197–205.
- Schwartz SM, Petitti DB, Siscovick DS, et al. Stroke and use of low-dose oral contraceptives in young women: a pooled analysis of two US studies. Stroke 1998; 29:2277–2284.
- Petitti DB, Sidney S, Bernstein A, Wolf S, Quesenberry C, Ziel HK. Stroke in users of low-dose oral contraceptives. N Engl J Med 1996; 335:8–15.
- Donaghy M, Chang CL, Poulter N; European Collaborators of the World Health Organisation Collaborative Study of Cardiovascular Disease and Steroid Hormone Contraception. Duration, frequency, recency, and type of migraine and the risk of ischaemic stroke in women of childbearing age. J Neurol Neurosurg Psychiatry 2002; 73:747–750.
- Gudmundsson LS, Scher AI, Aspelund T, et al. Migraine with aura and risk of cardiovascular and all cause mortality in men and women: prospective cohort study. BMJ 2010; 341:c3966.
- Kurth T, Slomke MA, Kase CS, et al. Migraine, headache, and the risk of stroke in women: a prospective study. Neurology 2005; 64:1020–1026.
- Lee ST, Chu K, Jung KH, et al. Decreased number and function of endothelial progenitor cells in patients with migraine. Neurology 2008; 70:1510–1517.
- Kunz GA, Liang G, Cuculi F, et al. Circulating endothelial progenitor cells predict coronary artery disease severity. Am Heart J 2006; 152:190–195.
- Kurth T, Gaziano JM, Cook NR, Logroscino G, Diener HC, Buring JE. Migraine and risk of cardiovascular disease in women. JAMA 2006; 296:283–291.
- Pezzini A, Del Zotto E, Giossi A, Volonghi I, Grassi M, Padovani A. The migraine-ischemic stroke connection: potential pathogenic mechanisms. Curr Mol Med 2009; 9:215–226.
- Sulak P, Willis S, Kuehl T, Coffee A, Clark J. Headaches and oral contraceptives: impact of eliminating the standard 7-day placebo interval. Headache 2007; 47:27–37.
- Nappi RE, Terreno E, Sances G, et al. Effect of a contraceptive pill containing estradiol valerate and dienogest (E2V/DNG) in women with menstrually-related migraine (MRM). Contraception 2013; 88:369–375.
- Calhoun A, Ford S, Pruitt A. The impact of extended-cycle vaginal ring contraception on migraine aura: a retrospective case series. Headache 2012; 52:1246–1253.
- Wu CQ, Grandi SM, Filion KB, Abenhaim HA, Joseph L, Eisenberg MJ. Drospirenone-containing oral contraceptive pills and the risk of venous and arterial thrombosis: a systematic review. BJOG 2013; 120:801–810.
- Dinger J, Bardenheuer K, Heinemann K. Cardiovascular and general safety of a 24-day regimen of drospirenone-containing combined oral contraceptives: final results from the International Active Surveillance Study of Women Taking Oral Contraceptives. Contraception 2014; 89:253–263.
- Benson LS, Micks EA. Why stop now? Extended and continuous regimens of combined hormonal contraceptive methods. Obstet Gynecol Clin North Am 2015; 42:669–681.
- Mannix LK, Calhoun AH. Menstrual migraine. Curr Treat Options Neurol 2004; 6:489–498.
- Calhoun AH. A novel specific prophylaxis for menstrual-associated migraine. South Med J 2004; 97:819–822.
- Calhoun AH. Current topics and controversies in menstrual migraine. Headache 2012; 52(suppl 1):8–11.
- Hannaford PC, Iversen L, Macfarlane TV, Elliott AM, Angus V, Lee AJ. Mortality among contraceptive pill users: cohort evidence from Royal College of General Practitioners’ Oral Contraception Study. BMJ 2010; 340:c927.
- Havrilesky LJ, Moorman PG, Lowery WJ, et al. Oral contraceptive pills as primary prevention for ovarian cancer: a systematic review and meta-analysis. Obstet Gynecol 2013; 122:139 -147.
- Collaborative Group on Epidemiological Studies on Endometrial Cancer. Endometrial cancer and oral contraceptives: an individual participant meta-analysis of 27,276 women with endometrial cancer from 36 epidemiological studies. Lancet Oncol 2015; 16:1061–1070.
- Fernandez E, La Vecchia C, Franceschi S, et al. Oral contraceptive use and risk of colorectal cancer. Epidemiology 1998; 9:295–300.
- Merritt MA, Riboli E, Murphy N, et al. Reproductive factors and risk of mortality in the European Prospective Investigation into Cancer and Nutrition; a cohort study. BMC Med 2015; 13:252.
- Vessey M, Yeates D. Oral contraceptive use and cancer: final report from the Oxford-Family Planning Association Contraceptive Study. Contraception 2013; 88:678–683.
KEY POINTS
- There is no restriction on the use of combined hormonal contraceptives by women with migraine without aura, and the risk vs benefit for women with aura is debatable.
- Migraine with aura—but not migraine without aura—is associated with a twofold increased risk of ischemic stroke, although the absolute risk is small in healthy women who do not smoke.
- Combined hormonal contraceptives are associated with ischemic stroke, but the risk is dose-dependent. Ultra-low-dose formulations (containing ≤ 20 μg of ethinyl estradiol) do not pose an increased risk of stroke in healthy nonsmokers.
Delirium in hospitalized patients: Risks and benefits of antipsychotics
Delirium is common in hospitalized patients and contributes to healthcare costs and poor patient outcomes, including death. Its diagnosis and management remain clinically challenging. Although consensus panel guidelines recommend antipsychotic medications to treat delirium when conservative measures fail, few head-to-head trials have been done to tell us which antipsychotic drug to select, and antipsychotic use poses risks in the elderly.
Here, we review the risks and benefits of using antipsychotic drugs to manage delirium and describe an approach to selecting and using 5 commonly used antipsychotics.
SCOPE OF THE PROBLEM
Delirium is common and serious, affecting 11% to 42% of patients hospitalized on general medical wards.1 The burden to the public and individual patient is extremely high. Delirium has been found to result in an additional $16,303 to $64,421 per delirious patient per year, with a subsequent total 1-year health-attributable cost between $38 billion and $152 billion in the United States.2 Furthermore, many patients who become delirious in the hospital lose their independence and are placed in long-term care facilities.3
Although delirium was originally thought to be a time-limited neurocognitive disorder, recent evidence shows that it persists much longer4 and that some patients never return to their previous level of function, suggesting that a single episode of delirium can significantly alter the course of an underlying dementia with the dramatic initiation of cognitive decline.3 Most alarmingly, delirium is associated with an increased rate of death.1
DSM-5 DEFINITION
According to the Diagnostic and Statistical Manual of Mental Disorders, 5th edition (DSM-5),5 delirium is a neurocognitive disorder characterized by the acute onset of disturbance in attention, awareness, and cognition that fluctuates in severity throughout the day and is the direct physiologic consequence of another medical condition. The cognitive impairment seen in delirium is typically global and can affect memory, orientation, language, visuospatial ability, and perception. Other prominent features include psychomotor disturbance, sleep-cycle derangement, and emotional lability.
The pathogenesis of delirium is not clearly delineated but may relate to cholinergic deficiency and dopaminergic excess.
THE FIRST STEPS: NONPHARMACOLOGIC MANAGEMENT
Inouye3 outlined a general 3-part approach to managing delirium:
Identify and address predisposing factors. All patients found to have an acute change in mental status should be evaluated for the underlying cause, with special attention to the most common causes, ie, infection, metabolic derangement, and substance intoxication and withdrawal. A thorough medication reconciliation should also be done to identify medications with psychoactive or anticholinergic effects.
Provide supportive care, eg, addressing volume and nutritional status, mobilizing the patient early, and giving prophylaxis against deep venous thrombosis.
Manage symptoms. Behavioral strategies should be instituted in every delirious patient and should include frequent reorientation, use of observers, encouragement of family involvement, avoidance of physical restraints and Foley catheters, use of vision and hearing aids, and normalizing the sleep-wake cycle.
ANTIPSYCHOTICS: ARE THEY SAFE AND EFFECTIVE?
The US Food and Drug Administration (FDA) has not approved any medications for delirium. However, multiple consensus statements, including those by the American Psychiatric Association,6 the Canadian Coalition for Seniors’ Mental Health,7 and the UK National Institute for Health and Care Excellence,8 advocate for psychopharmacologic management of delirium symptoms in the following situations:
- The patient is in significant distress from his or her symptoms
- The patient poses a safety risk to self or others
- The patient is impeding essential aspects of his or her medical care.
Guidelines from these organizations recommend antipsychotic medications as the first-line drugs for managing delirium symptoms not caused by substance withdrawal. Nevertheless, the use of antipsychotics in the management of delirium remains controversial. While a number of studies suggest these drugs are beneficial,9–11 others do not.12 These consensus panels advocate for the judicious use of antipsychotics, limited to the specific situations outlined above.
The use of antipsychotics in elderly and medically complex patients poses risks. One of the most significant safety concerns is increased risk of death due to adverse cardiac events caused by prolongation of the QT interval.
Antipsychotics, QT prolongation, and torsades de pointes
Most antipsychotics have the potential to prolong the time of ventricular depolarization and repolarization and the QT interval to some extent, which can lead to torsades de pointes.13 Other risk factors for prolonged QT interval and torsades de pointes include:
- Long QT syndrome (a genetic arrhythmia)
- Female sex
- Old age
- Electrolyte abnormalities (hypokalemia, hypocalcemia, hypomagnesemia)
- Preexisting heart conditions such as bradycardia, left ventricular dysfunction, heart failure, mitral valve prolapse, and previous myocardial infarction
- Medical conditions that cause electrolyte derangements
- Medications, including antiarrhythmics, antibiotics (macrolides, quinolones), antifungals, antimalarials, antiemetics, some opioids (methadone), and most antipsychotics.
Haloperidol. Postmarketing analysis in 2007 found 73 cases of haloperidol-related torsades de pointes. However, many of these were confounded by other QT-prolonging medications and medical conditions.14
The QT-prolonging effect of haloperidol administered orally or intramuscularly is actually quite small. The equivalent oral dose of 15 mg of haloperidol (assuming 50% bioavailability) given orally or intramuscularly increases the corrected QT interval (QTc) by only 7 to 8 milliseconds. But intravenous haloperidol can cause much more significant QT prolongation: 8 of the 11 reported cases of fatal torsades de pointes occurred when haloperidol was given intravenously.14 Therefore, the FDA recommends cardiac monitoring for all patients receiving intravenous haloperidol.
Oral olanzapine, risperidone, and quetiapine prolong the QT interval approximately as much as oral haloperidol.
Aripiprazole has not been associated with significant QT prolongation.13
Atypical antipsychotics and stroke
The FDA has issued multiple warnings for prescribing antipsychotic medications in the elderly. In 2003, it warned prescribers of increased cerebrovascular adverse events, including stroke, in elderly patients with dementia who were treated with an atypical antipsychotic (risperidone, olanzapine, or aripiprazole) vs placebo.15
Atypical antipsychotics and risk of death
In 2005, the FDA issued a black-box warning about increased all-cause mortality risk in patients with dementia treated with atypical antipsychotics for behavioral disturbance (relative risk 1.6–1.7).16
This warning was likely based on a meta-analysis by Schneider et al17 of trials in which patients with dementia were randomized to receive either an atypical antipsychotic or placebo. The death rate was 3.5% in patients treated with an atypical antipsychotic vs 2.3% in patients treated with placebo, indicating a number needed to harm of 100. The most common causes of death were cardiovascular disease and pneumonia. However, the trials in this meta-analysis included only patients who were prescribed atypical antipsychotics for ongoing management of behavioral disturbances due to dementia in either the outpatient or nursing home setting. None of the trials looked at patients who were prescribed atypical antipsychotics for a limited time in a closely monitored inpatient setting.
Effectiveness of antipsychotics
While several studies since the FDA black-box warning have shown that antipsychotics are safe, the efficacy of these drugs in delirium management remains controversial.
In a 2016 meta-analysis, Kishi et al18 found that antipsychotics were superior to placebo in terms of response rate (defined as improvement of delirium severity rating scores), with a number needed to treat of 2.
In contrast, a meta-analysis by Neufeld et al12 found that antipsychotic use was not associated with a change in delirium duration, severity, or length of stay in the hospital or intensive care unit. However, the studies in this meta-analysis varied widely in age range, study design, drug comparison, and treatment strategy (with drugs given as both prophylaxis and treatment). Thus, the results are difficult to interpret.
Kishi et al18 found no difference in the incidence of death, extrapyramidal symptoms, akathisia, or QT prolongation between patients treated with antipsychotic drugs vs placebo.
In a prospective observational study, Hatta et al19 followed 2,453 inpatients who became delirious. Only 22 (0.9%) experienced adverse events attributable to antipsychotic use, the most common being aspiration pneumonia (0.7%), followed by cardiovascular events (0.2%). Notably, no patient died of antipsychotic-related events. In this study, the antipsychotic was stopped as soon as the delirium symptoms resolved, in most cases in 3 to 7 days.
Taken together, these studies indicate that despite the risk of QT prolongation with antipsychotic use and increased rates of morbidity with antipsychotic use in dementia, time-limited management of delirium with antipsychotics is effective9–11 and safe.
SELECTING AND USING ANTIPSYCHOTICS TO TREAT DELIRIUM
Identifying a single preferred agent is difficult, since we lack enough evidence from randomized controlled trials that directly compared the various antipsychotics used in delirium management.
Both typical and atypical antipsychotics are used in clinical practice to manage delirium. The typical antipsychotic most often used is haloperidol, while the most commonly used atypical antipsychotics for delirium include olanzapine, quetiapine, risperidone, and (more recently) aripiprazole.
The American Psychiatric Association guidelines6 suggest using haloperidol because it is the antipsychotic that has been most studied for delirium,20 and we have decades of experience with its use. Despite this, recent prospective studies have suggested that the atypical antipsychotics may be better because they have a faster onset of action and lower incidence of extrapyramidal symptoms.18,21
Because we lack enough head-to-head trials comparing the efficacy of the 5 most commonly used antipsychotics for the management of delirium, and because the prospective trials that do exist show equal efficacy across the antipsychotics studied,22 we suggest considering the unique pharmacologic properties of each drug within the patient’s clinical context when selecting which antipsychotic to use.
Table 123–25 summarizes some key characteristics of the 5 most commonly used antipsychotics.
Haloperidol
Haloperidol, a typical antipsychotic, is a potent antagonist of the dopamine D2 receptor.
Haloperidol has the advantage of having the strongest evidence base for use in delirium. In addition, it is available in oral, intravenous, and intramuscular dosage forms, and it has minimal effects on vital signs, negligible anticholinergic activity, and minimal interactions with other medications.21
Intravenous haloperidol poses a significant risk of QT prolongation and so should be used judiciously in patients with preexisting cardiac conditions or other risk factors for QT prolongation as outlined above, and with careful cardiac monitoring. Parenteral haloperidol is approximately twice as potent as oral haloperidol.
Some evidence suggests a higher risk of acute dystonia and other extrapyramidal symptoms with haloperidol than with the atypical antipsychotics.21,26 In contrast, a 2013 prospective study showed that low doses of haloperidol (< 3.5 mg/day) did not result in a greater frequency of extrapyramidal symptoms.22 Nevertheless, if a patient has a history of extrapyramidal symptoms, haloperidol should likely be avoided in favor of an atypical antipsychotic.
Atypical antipsychotics
Olanzapine, quetiapine, and risperidone are atypical antipsychotics that, like haloperidol, antagonize the dopamine D2 receptor, but also have antagonist action at serotonin, histamine, and alpha-2 receptors. This multireceptor antagonism reduces the risk of extrapyramidal symptoms but increases the risk of orthostatic hypotension.
Quetiapine, in particular, imposes an unacceptably high risk of orthostatic hypotension and so is not recommended for use in delirium in the emergency department.27 Additionally, quetiapine is anticholinergic, raising concerns about constipation and urinary retention.
Although the association between fall risk and antipsychotic use remains controversial,28,29 a study found that olanzapine conferred a lower fall risk than quetiapine and risperidone.30
Of these drugs, only olanzapine is available in an intramuscular dosage form. Both risperidone and olanzapine are available in dissolvable tablets; however, they are not sublingually absorbed.
Randomized controlled trials have shown that olanzapine is effective in managing cancer-related nausea, and therefore it may be useful in managing delirium in oncology patients.31,32
Patients with Parkinson disease are exquisitely sensitive to the antidopaminergic effects of antipsychotics but are also vulnerable to delirium, so they present a unique treatment challenge. The agent of choice in patients with Parkinson disease is quetiapine, as multiple trials have shown it has no effect on the motor symptoms of Parkinson disease (reviewed by Desmarais et al in a systematic meta-analysis33).
Aripiprazole is increasingly used to manage delirium. Its mechanism of action differs from that of the other atypical antipsychotics, as it is a partial dopamine agonist. It is available in oral, orally dissolvable, and intramuscular forms. It appears to be slightly less effective than the other atypical antipsychotics,34 but it may be useful for hypoactive delirium as it is less sedating than the other agents.35 Because its effect on the QT interval is negligible, it may also be favored in patients who have a high baseline QTc or other predisposing factors for torsades de pointes.
BALANCING THE RISKS
Antipsychotic drugs have been shown to be effective and generally safe. Antipsychotics do prolong the QT interval. However, other than with intravenous administration of haloperidol, the absolute effect is minimal. Although large meta-analyses have shown a higher rate of all-cause mortality in elderly outpatients with dementia who are prescribed atypical antipsychotics, an increase in death rates has not been borne out by prospective studies focusing on hospitalized patients who receive low doses of antipsychotics for a limited time.
There are no head-to-head randomized controlled trials comparing the efficacy of all of the 5 most commonly used antipsychotics. Therefore, we suggest considering the unique psychopharmacologic properties of each agent within the patient’s clinical setting, specifically taking into account the risk of cardiac arrhythmia, risk of orthostasis and falls, history of extrapyramidal symptoms, other comorbidities such as Parkinson disease and cancer, and the desired route of administration.
At the time the patient is discharged, we recommend a careful medication reconciliation and discontinuation of the antipsychotic drug once delirium has resolved. Studies show that at least 26% of antipsychotics initiated in the hospital are continued after discharge.36,37
Current delirium consensus statements recommend limiting the use of antipsychotics to target patient distress, impediment of care, or safety, because of the putative risks of antipsychotic use in the elderly. However, a growing body of evidence shows that low-dose, time-limited antipsychotic use is safe and effective in the treatment of delirium. In fact, González et al found that delirium is an independent risk factor for death, and each 48-hour increase in delirium is associated with an increased mortality risk of 11%, suggesting that delay in treating delirium may actually increase the risk of death.38
Therefore, we must balance the risks of prescribing antipsychotics in medically vulnerable patients against the increasing burden of evidence supporting the serious risks of morbidity and mortality of delirium, as well as the costs. Much remains to be studied to optimize antipsychotic use in delirium.
- Siddiqi N, House AO, Holmes JD. Occurrence and outcome of delirium in medical in-patients: a systematic literature review. Age Ageing 2006; 35:350–364.
- Leslie DL, Marcantonio ER, Zhang Y, Leo-Summers L, Inouye SK. One-year health care costs associated with delirium in the elderly population. Arch Intern Med 2008; 168:27–32.
- Inouye SK. Delirium in older persons. N Engl J Med 2006; 354:1157–1165.
- Levkoff SE, Evans DA, Liptzin B, et al. Delirium: the occurrence and persistence of symptoms among elderly hospitalized patients. Arch Intern Med 1992; 152:334–340.
- American Psychiatric Association. Diagnostic and statistical manual of mental disorders. 5th ed. Arlington, VA: American Psychiatric Publishing; 2013.
- Trzepacz P, Breitbart W, Franklin J, Levenson J, Martini DR, Wang P; American Psychiatric Association (APA). Practice guideline for the treatment of patients with delirium. http://psychiatryonline.org/pb/assets/raw/sitewide/practice_guidelines/guidelines/delirium.pdf. Accessed July 13, 2017.
- Canadian Coalition for Seniors’ Mental Health. National guidelines for seniors’ mental health: the assessment and treatment of delirium. http://ccsmh.ca/wp-content/uploads/2016/03/NatlGuideline_Delirium.pdf. Accessed July 13, 2017.
- National Institute for Health and Care Excellence (NICE). Delirium: prevention, diagnosis and management. www.nice.org.uk/guidance/cg103. Accessed July 13, 2017.
- Bourne RS, Tahir TA, Borthwick M, Sampson EL. Drug treatment of delirium: past, present and future. J Psychosom Res 2008; 65:273–282.
- Campbell N, Boustani MA, Ayub A, et al. Pharmacological management of delirium in hospitalized adults—a systematic evidence review. J Gen Intern Med 2009; 24:848–853.
- Devlin JW, Skrobik Y. Antipsychotics for the prevention and treatment of delirium in the intensive care unit: what is their role? Harv Rev Psychiatry 2011; 19:59–67.
- Neufeld KJ, Yue J, Robinson TN, Inouye SK, Needham DM. Antipsychotic medication for prevention and treatment of delirium in hospitalized adults: a systematic review and meta-analysis. J Am Geriatr Soc 2016; 64:705–714.
- Beach SR, Celano MC, Noseworthy PA, Januzzi JL, Huffman JC. QTc prolongation, torsades de pointes, and psychotropic medications. Psychosomatics 2013; 54:1–13.
- US Food and Drug Administration (FDA). Information for healthcare professionals: haloperidol (marketed as Haldol, Haldol decanoate and Haldol lactate). www.fda.gov/Drugs/DrugSafety/ucm085203.htm. Accessed July 13, 2017.
- US Food and Drug Administration Center for Drug Evaluation and Research. Approval package for: Application Number: NDA 20-272/S-033, 20-588/S-021 & 21-444/S-004. www.accessdata.fda.gov/drugsatfda_docs/nda/2003/020588_S021_RISPERDAL_TABLETS.pdf. Accessed July 13, 2017.
- US Food and Drug Administration. Public health advisory: deaths with antipsychotics in elderly patients with behavioral disturbances. www.fda.gov/drugs/drugsafety/postmarketdrugsafetyinformationforpatientsandproviders/ucm053171. Accessed July 13, 2017.
- Schneider LS, Dagerman KS, Insel P. Risk of death with atypical antipsychotic drug treatment for dementia: meta-analysis of randomized placebo-controlled trials. JAMA 2005; 294:1934–1943.
- Kishi T, Hirota T, Matsunaga S, Iwata N. Antipsychotic medications for the treatment of delirium: a systematic review and meta-analysis of randomised controlled trials. J Neurol Neurosurg Psychiatry 2016; 87:767–774.
- Hatta K, Kishi Y, Wada K, et al. Antipsychotics for delirium in the general hospital setting in consecutive 2453 inpatients: a prospective observational study. Int J Geriatr Psychiatry 2014; 29;253–262.
- Breitbart W, Marotta R, Platt MM, et al. A double-blind trial of haloperidol, chlorpromazine, and lorazepam in the treatment of delirium in hospitalized AIDS patients. Am J Psychiatry 1996; 153:231–237.
- Wilson MP, Pepper D, Currier GW, Holloman GH Jr, Feifel D. The psychopharmacology of agitation: consensus statement of the American Association For Emergency Psychiatry Project Beta Psychopharmacology Workgroup. West J Emerg Med 2012; 13:26–34.
- Yoon HJ, Park KM, Choi WJ, et al. Efficacy and safety of haloperidol versus atypical antipsychotic medications in the treatment of delirium. BMC Psychiatry 2013; 13:240.
- American Psychiatric Association. Manual of Clinical Psychopharmacology. 8th ed. Arlington, VA: American Psychiatric Publishing; 2015.
- Conley RR, Kelly DL. Pharmacologic Treatment of Schizophrenia. 3rd ed. West Islip, NY: Professional Communications; 2007.
- American Psychiatric Association (APA). The American Psychiatric Publishing Textbook of Psychosomatic Medicine. Psychiatric Care of the Medically Ill, 2nd ed. Arlington, VA: American Psychiatric Publishing; 2011.
- Boettger S, Jenewein J, Breitbart W. Haloperidol, risperidone, olanzapine and aripiprazole in the management of delirium: a comparison of efficacy, safety, and side effects. Palliat Support Care 2015; 13:1079–1085.
- Currier GW, Trenton AJ, Walsh PG, van Wijngaarden E. A pilot, open-label study of quetiapine for treatment of moderate psychotic agitation in the emergency setting. J Psychiatr Pract 2006; 12:223–228.
- Chatterjee S, Chen H, Johnson ML, Aparasu RR. Risk of falls and fractures in older adults using atypical antipsychotic agents: a propensity score-adjusted, retrospective cohort study. Am J Geriatr Pharmacother 2012; 10:84–94.
- Rigler SK, Shireman TI, Cook-Wiens GJ, et al. Fracture risk in nursing home residents initiating antipsychotic medications. J Am Geriatr Soc 2013; 61: 715–722.
- Bozat-Emre S, Doupe M, Kozyrskyj AL, Grymonpre R, Mahmud SM. Atypical antipsychotic drug use and falls among nursing home residents in Winnipeg, Canada. Int J Geriatr Psychiatry 2015; 30:842–850.
- Navari RM, Gray SE, Kerr AC. Olanzapine versus aprepitant for the prevention of chemotherapy-induced nausea and vomiting: a randomized phase III trial. J Support Oncol 2011; 9:188–195.
- Navari RM. Olanzapine for the prevention and treatment of chronic nausea and chemotherapy-induced nausea and vomiting. Eur J Pharmacol 2014; 722:180–186.
- Desmarais P, Massoud F, Filion J, Nguyen QD, Bajsarowicz P. Quetiapine for psychosis in Parkinson disease and neurodegenerative Parkinsonian disorders: a systematic review. J Geriatr Psychiatry Neurol 2016; 29:227–236.
- Citrome L. Comparison of intramuscular ziprasidone, olanzapine, or aripiprazole for agitation: a quantitative review of efficacy and safety. J Clin Psychiatry 2007; 68:1876–1885.
- Marder SR, McQuade RD, Stock E, et al. Aripiprazole in the treatment of schizophrenia: safety and tolerability in short-term, placebo-controlled trials. Schizophr Res 2003; 61:123–136.
- Loh KP, Ramdass S, Garb JL, et al. Long-term outcomes of elders discharged on antipsychotics. J Hosp Med 2016; 11:550–555.
- Herzig SJ, Rothberg MB, Guess JR, et al. Antipsychotic use in hospitalized adults: rates, indications, and predictors. J Am Geriatr Soc 2016; 64:299–305.
- González M, Martínez G, Calderón J, et al. Impact of delirium on short-term mortality in elderly inpatients: a prospective cohort study. Psychosomatics 2009; 50:234–238.
Delirium is common in hospitalized patients and contributes to healthcare costs and poor patient outcomes, including death. Its diagnosis and management remain clinically challenging. Although consensus panel guidelines recommend antipsychotic medications to treat delirium when conservative measures fail, few head-to-head trials have been done to tell us which antipsychotic drug to select, and antipsychotic use poses risks in the elderly.
Here, we review the risks and benefits of using antipsychotic drugs to manage delirium and describe an approach to selecting and using 5 commonly used antipsychotics.
SCOPE OF THE PROBLEM
Delirium is common and serious, affecting 11% to 42% of patients hospitalized on general medical wards.1 The burden to the public and individual patient is extremely high. Delirium has been found to result in an additional $16,303 to $64,421 per delirious patient per year, with a subsequent total 1-year health-attributable cost between $38 billion and $152 billion in the United States.2 Furthermore, many patients who become delirious in the hospital lose their independence and are placed in long-term care facilities.3
Although delirium was originally thought to be a time-limited neurocognitive disorder, recent evidence shows that it persists much longer4 and that some patients never return to their previous level of function, suggesting that a single episode of delirium can significantly alter the course of an underlying dementia with the dramatic initiation of cognitive decline.3 Most alarmingly, delirium is associated with an increased rate of death.1
DSM-5 DEFINITION
According to the Diagnostic and Statistical Manual of Mental Disorders, 5th edition (DSM-5),5 delirium is a neurocognitive disorder characterized by the acute onset of disturbance in attention, awareness, and cognition that fluctuates in severity throughout the day and is the direct physiologic consequence of another medical condition. The cognitive impairment seen in delirium is typically global and can affect memory, orientation, language, visuospatial ability, and perception. Other prominent features include psychomotor disturbance, sleep-cycle derangement, and emotional lability.
The pathogenesis of delirium is not clearly delineated but may relate to cholinergic deficiency and dopaminergic excess.
THE FIRST STEPS: NONPHARMACOLOGIC MANAGEMENT
Inouye3 outlined a general 3-part approach to managing delirium:
Identify and address predisposing factors. All patients found to have an acute change in mental status should be evaluated for the underlying cause, with special attention to the most common causes, ie, infection, metabolic derangement, and substance intoxication and withdrawal. A thorough medication reconciliation should also be done to identify medications with psychoactive or anticholinergic effects.
Provide supportive care, eg, addressing volume and nutritional status, mobilizing the patient early, and giving prophylaxis against deep venous thrombosis.
Manage symptoms. Behavioral strategies should be instituted in every delirious patient and should include frequent reorientation, use of observers, encouragement of family involvement, avoidance of physical restraints and Foley catheters, use of vision and hearing aids, and normalizing the sleep-wake cycle.
ANTIPSYCHOTICS: ARE THEY SAFE AND EFFECTIVE?
The US Food and Drug Administration (FDA) has not approved any medications for delirium. However, multiple consensus statements, including those by the American Psychiatric Association,6 the Canadian Coalition for Seniors’ Mental Health,7 and the UK National Institute for Health and Care Excellence,8 advocate for psychopharmacologic management of delirium symptoms in the following situations:
- The patient is in significant distress from his or her symptoms
- The patient poses a safety risk to self or others
- The patient is impeding essential aspects of his or her medical care.
Guidelines from these organizations recommend antipsychotic medications as the first-line drugs for managing delirium symptoms not caused by substance withdrawal. Nevertheless, the use of antipsychotics in the management of delirium remains controversial. While a number of studies suggest these drugs are beneficial,9–11 others do not.12 These consensus panels advocate for the judicious use of antipsychotics, limited to the specific situations outlined above.
The use of antipsychotics in elderly and medically complex patients poses risks. One of the most significant safety concerns is increased risk of death due to adverse cardiac events caused by prolongation of the QT interval.
Antipsychotics, QT prolongation, and torsades de pointes
Most antipsychotics have the potential to prolong the time of ventricular depolarization and repolarization and the QT interval to some extent, which can lead to torsades de pointes.13 Other risk factors for prolonged QT interval and torsades de pointes include:
- Long QT syndrome (a genetic arrhythmia)
- Female sex
- Old age
- Electrolyte abnormalities (hypokalemia, hypocalcemia, hypomagnesemia)
- Preexisting heart conditions such as bradycardia, left ventricular dysfunction, heart failure, mitral valve prolapse, and previous myocardial infarction
- Medical conditions that cause electrolyte derangements
- Medications, including antiarrhythmics, antibiotics (macrolides, quinolones), antifungals, antimalarials, antiemetics, some opioids (methadone), and most antipsychotics.
Haloperidol. Postmarketing analysis in 2007 found 73 cases of haloperidol-related torsades de pointes. However, many of these were confounded by other QT-prolonging medications and medical conditions.14
The QT-prolonging effect of haloperidol administered orally or intramuscularly is actually quite small. The equivalent oral dose of 15 mg of haloperidol (assuming 50% bioavailability) given orally or intramuscularly increases the corrected QT interval (QTc) by only 7 to 8 milliseconds. But intravenous haloperidol can cause much more significant QT prolongation: 8 of the 11 reported cases of fatal torsades de pointes occurred when haloperidol was given intravenously.14 Therefore, the FDA recommends cardiac monitoring for all patients receiving intravenous haloperidol.
Oral olanzapine, risperidone, and quetiapine prolong the QT interval approximately as much as oral haloperidol.
Aripiprazole has not been associated with significant QT prolongation.13
Atypical antipsychotics and stroke
The FDA has issued multiple warnings for prescribing antipsychotic medications in the elderly. In 2003, it warned prescribers of increased cerebrovascular adverse events, including stroke, in elderly patients with dementia who were treated with an atypical antipsychotic (risperidone, olanzapine, or aripiprazole) vs placebo.15
Atypical antipsychotics and risk of death
In 2005, the FDA issued a black-box warning about increased all-cause mortality risk in patients with dementia treated with atypical antipsychotics for behavioral disturbance (relative risk 1.6–1.7).16
This warning was likely based on a meta-analysis by Schneider et al17 of trials in which patients with dementia were randomized to receive either an atypical antipsychotic or placebo. The death rate was 3.5% in patients treated with an atypical antipsychotic vs 2.3% in patients treated with placebo, indicating a number needed to harm of 100. The most common causes of death were cardiovascular disease and pneumonia. However, the trials in this meta-analysis included only patients who were prescribed atypical antipsychotics for ongoing management of behavioral disturbances due to dementia in either the outpatient or nursing home setting. None of the trials looked at patients who were prescribed atypical antipsychotics for a limited time in a closely monitored inpatient setting.
Effectiveness of antipsychotics
While several studies since the FDA black-box warning have shown that antipsychotics are safe, the efficacy of these drugs in delirium management remains controversial.
In a 2016 meta-analysis, Kishi et al18 found that antipsychotics were superior to placebo in terms of response rate (defined as improvement of delirium severity rating scores), with a number needed to treat of 2.
In contrast, a meta-analysis by Neufeld et al12 found that antipsychotic use was not associated with a change in delirium duration, severity, or length of stay in the hospital or intensive care unit. However, the studies in this meta-analysis varied widely in age range, study design, drug comparison, and treatment strategy (with drugs given as both prophylaxis and treatment). Thus, the results are difficult to interpret.
Kishi et al18 found no difference in the incidence of death, extrapyramidal symptoms, akathisia, or QT prolongation between patients treated with antipsychotic drugs vs placebo.
In a prospective observational study, Hatta et al19 followed 2,453 inpatients who became delirious. Only 22 (0.9%) experienced adverse events attributable to antipsychotic use, the most common being aspiration pneumonia (0.7%), followed by cardiovascular events (0.2%). Notably, no patient died of antipsychotic-related events. In this study, the antipsychotic was stopped as soon as the delirium symptoms resolved, in most cases in 3 to 7 days.
Taken together, these studies indicate that despite the risk of QT prolongation with antipsychotic use and increased rates of morbidity with antipsychotic use in dementia, time-limited management of delirium with antipsychotics is effective9–11 and safe.
SELECTING AND USING ANTIPSYCHOTICS TO TREAT DELIRIUM
Identifying a single preferred agent is difficult, since we lack enough evidence from randomized controlled trials that directly compared the various antipsychotics used in delirium management.
Both typical and atypical antipsychotics are used in clinical practice to manage delirium. The typical antipsychotic most often used is haloperidol, while the most commonly used atypical antipsychotics for delirium include olanzapine, quetiapine, risperidone, and (more recently) aripiprazole.
The American Psychiatric Association guidelines6 suggest using haloperidol because it is the antipsychotic that has been most studied for delirium,20 and we have decades of experience with its use. Despite this, recent prospective studies have suggested that the atypical antipsychotics may be better because they have a faster onset of action and lower incidence of extrapyramidal symptoms.18,21
Because we lack enough head-to-head trials comparing the efficacy of the 5 most commonly used antipsychotics for the management of delirium, and because the prospective trials that do exist show equal efficacy across the antipsychotics studied,22 we suggest considering the unique pharmacologic properties of each drug within the patient’s clinical context when selecting which antipsychotic to use.
Table 123–25 summarizes some key characteristics of the 5 most commonly used antipsychotics.
Haloperidol
Haloperidol, a typical antipsychotic, is a potent antagonist of the dopamine D2 receptor.
Haloperidol has the advantage of having the strongest evidence base for use in delirium. In addition, it is available in oral, intravenous, and intramuscular dosage forms, and it has minimal effects on vital signs, negligible anticholinergic activity, and minimal interactions with other medications.21
Intravenous haloperidol poses a significant risk of QT prolongation and so should be used judiciously in patients with preexisting cardiac conditions or other risk factors for QT prolongation as outlined above, and with careful cardiac monitoring. Parenteral haloperidol is approximately twice as potent as oral haloperidol.
Some evidence suggests a higher risk of acute dystonia and other extrapyramidal symptoms with haloperidol than with the atypical antipsychotics.21,26 In contrast, a 2013 prospective study showed that low doses of haloperidol (< 3.5 mg/day) did not result in a greater frequency of extrapyramidal symptoms.22 Nevertheless, if a patient has a history of extrapyramidal symptoms, haloperidol should likely be avoided in favor of an atypical antipsychotic.
Atypical antipsychotics
Olanzapine, quetiapine, and risperidone are atypical antipsychotics that, like haloperidol, antagonize the dopamine D2 receptor, but also have antagonist action at serotonin, histamine, and alpha-2 receptors. This multireceptor antagonism reduces the risk of extrapyramidal symptoms but increases the risk of orthostatic hypotension.
Quetiapine, in particular, imposes an unacceptably high risk of orthostatic hypotension and so is not recommended for use in delirium in the emergency department.27 Additionally, quetiapine is anticholinergic, raising concerns about constipation and urinary retention.
Although the association between fall risk and antipsychotic use remains controversial,28,29 a study found that olanzapine conferred a lower fall risk than quetiapine and risperidone.30
Of these drugs, only olanzapine is available in an intramuscular dosage form. Both risperidone and olanzapine are available in dissolvable tablets; however, they are not sublingually absorbed.
Randomized controlled trials have shown that olanzapine is effective in managing cancer-related nausea, and therefore it may be useful in managing delirium in oncology patients.31,32
Patients with Parkinson disease are exquisitely sensitive to the antidopaminergic effects of antipsychotics but are also vulnerable to delirium, so they present a unique treatment challenge. The agent of choice in patients with Parkinson disease is quetiapine, as multiple trials have shown it has no effect on the motor symptoms of Parkinson disease (reviewed by Desmarais et al in a systematic meta-analysis33).
Aripiprazole is increasingly used to manage delirium. Its mechanism of action differs from that of the other atypical antipsychotics, as it is a partial dopamine agonist. It is available in oral, orally dissolvable, and intramuscular forms. It appears to be slightly less effective than the other atypical antipsychotics,34 but it may be useful for hypoactive delirium as it is less sedating than the other agents.35 Because its effect on the QT interval is negligible, it may also be favored in patients who have a high baseline QTc or other predisposing factors for torsades de pointes.
BALANCING THE RISKS
Antipsychotic drugs have been shown to be effective and generally safe. Antipsychotics do prolong the QT interval. However, other than with intravenous administration of haloperidol, the absolute effect is minimal. Although large meta-analyses have shown a higher rate of all-cause mortality in elderly outpatients with dementia who are prescribed atypical antipsychotics, an increase in death rates has not been borne out by prospective studies focusing on hospitalized patients who receive low doses of antipsychotics for a limited time.
There are no head-to-head randomized controlled trials comparing the efficacy of all of the 5 most commonly used antipsychotics. Therefore, we suggest considering the unique psychopharmacologic properties of each agent within the patient’s clinical setting, specifically taking into account the risk of cardiac arrhythmia, risk of orthostasis and falls, history of extrapyramidal symptoms, other comorbidities such as Parkinson disease and cancer, and the desired route of administration.
At the time the patient is discharged, we recommend a careful medication reconciliation and discontinuation of the antipsychotic drug once delirium has resolved. Studies show that at least 26% of antipsychotics initiated in the hospital are continued after discharge.36,37
Current delirium consensus statements recommend limiting the use of antipsychotics to target patient distress, impediment of care, or safety, because of the putative risks of antipsychotic use in the elderly. However, a growing body of evidence shows that low-dose, time-limited antipsychotic use is safe and effective in the treatment of delirium. In fact, González et al found that delirium is an independent risk factor for death, and each 48-hour increase in delirium is associated with an increased mortality risk of 11%, suggesting that delay in treating delirium may actually increase the risk of death.38
Therefore, we must balance the risks of prescribing antipsychotics in medically vulnerable patients against the increasing burden of evidence supporting the serious risks of morbidity and mortality of delirium, as well as the costs. Much remains to be studied to optimize antipsychotic use in delirium.
Delirium is common in hospitalized patients and contributes to healthcare costs and poor patient outcomes, including death. Its diagnosis and management remain clinically challenging. Although consensus panel guidelines recommend antipsychotic medications to treat delirium when conservative measures fail, few head-to-head trials have been done to tell us which antipsychotic drug to select, and antipsychotic use poses risks in the elderly.
Here, we review the risks and benefits of using antipsychotic drugs to manage delirium and describe an approach to selecting and using 5 commonly used antipsychotics.
SCOPE OF THE PROBLEM
Delirium is common and serious, affecting 11% to 42% of patients hospitalized on general medical wards.1 The burden to the public and individual patient is extremely high. Delirium has been found to result in an additional $16,303 to $64,421 per delirious patient per year, with a subsequent total 1-year health-attributable cost between $38 billion and $152 billion in the United States.2 Furthermore, many patients who become delirious in the hospital lose their independence and are placed in long-term care facilities.3
Although delirium was originally thought to be a time-limited neurocognitive disorder, recent evidence shows that it persists much longer4 and that some patients never return to their previous level of function, suggesting that a single episode of delirium can significantly alter the course of an underlying dementia with the dramatic initiation of cognitive decline.3 Most alarmingly, delirium is associated with an increased rate of death.1
DSM-5 DEFINITION
According to the Diagnostic and Statistical Manual of Mental Disorders, 5th edition (DSM-5),5 delirium is a neurocognitive disorder characterized by the acute onset of disturbance in attention, awareness, and cognition that fluctuates in severity throughout the day and is the direct physiologic consequence of another medical condition. The cognitive impairment seen in delirium is typically global and can affect memory, orientation, language, visuospatial ability, and perception. Other prominent features include psychomotor disturbance, sleep-cycle derangement, and emotional lability.
The pathogenesis of delirium is not clearly delineated but may relate to cholinergic deficiency and dopaminergic excess.
THE FIRST STEPS: NONPHARMACOLOGIC MANAGEMENT
Inouye3 outlined a general 3-part approach to managing delirium:
Identify and address predisposing factors. All patients found to have an acute change in mental status should be evaluated for the underlying cause, with special attention to the most common causes, ie, infection, metabolic derangement, and substance intoxication and withdrawal. A thorough medication reconciliation should also be done to identify medications with psychoactive or anticholinergic effects.
Provide supportive care, eg, addressing volume and nutritional status, mobilizing the patient early, and giving prophylaxis against deep venous thrombosis.
Manage symptoms. Behavioral strategies should be instituted in every delirious patient and should include frequent reorientation, use of observers, encouragement of family involvement, avoidance of physical restraints and Foley catheters, use of vision and hearing aids, and normalizing the sleep-wake cycle.
ANTIPSYCHOTICS: ARE THEY SAFE AND EFFECTIVE?
The US Food and Drug Administration (FDA) has not approved any medications for delirium. However, multiple consensus statements, including those by the American Psychiatric Association,6 the Canadian Coalition for Seniors’ Mental Health,7 and the UK National Institute for Health and Care Excellence,8 advocate for psychopharmacologic management of delirium symptoms in the following situations:
- The patient is in significant distress from his or her symptoms
- The patient poses a safety risk to self or others
- The patient is impeding essential aspects of his or her medical care.
Guidelines from these organizations recommend antipsychotic medications as the first-line drugs for managing delirium symptoms not caused by substance withdrawal. Nevertheless, the use of antipsychotics in the management of delirium remains controversial. While a number of studies suggest these drugs are beneficial,9–11 others do not.12 These consensus panels advocate for the judicious use of antipsychotics, limited to the specific situations outlined above.
The use of antipsychotics in elderly and medically complex patients poses risks. One of the most significant safety concerns is increased risk of death due to adverse cardiac events caused by prolongation of the QT interval.
Antipsychotics, QT prolongation, and torsades de pointes
Most antipsychotics have the potential to prolong the time of ventricular depolarization and repolarization and the QT interval to some extent, which can lead to torsades de pointes.13 Other risk factors for prolonged QT interval and torsades de pointes include:
- Long QT syndrome (a genetic arrhythmia)
- Female sex
- Old age
- Electrolyte abnormalities (hypokalemia, hypocalcemia, hypomagnesemia)
- Preexisting heart conditions such as bradycardia, left ventricular dysfunction, heart failure, mitral valve prolapse, and previous myocardial infarction
- Medical conditions that cause electrolyte derangements
- Medications, including antiarrhythmics, antibiotics (macrolides, quinolones), antifungals, antimalarials, antiemetics, some opioids (methadone), and most antipsychotics.
Haloperidol. Postmarketing analysis in 2007 found 73 cases of haloperidol-related torsades de pointes. However, many of these were confounded by other QT-prolonging medications and medical conditions.14
The QT-prolonging effect of haloperidol administered orally or intramuscularly is actually quite small. The equivalent oral dose of 15 mg of haloperidol (assuming 50% bioavailability) given orally or intramuscularly increases the corrected QT interval (QTc) by only 7 to 8 milliseconds. But intravenous haloperidol can cause much more significant QT prolongation: 8 of the 11 reported cases of fatal torsades de pointes occurred when haloperidol was given intravenously.14 Therefore, the FDA recommends cardiac monitoring for all patients receiving intravenous haloperidol.
Oral olanzapine, risperidone, and quetiapine prolong the QT interval approximately as much as oral haloperidol.
Aripiprazole has not been associated with significant QT prolongation.13
Atypical antipsychotics and stroke
The FDA has issued multiple warnings for prescribing antipsychotic medications in the elderly. In 2003, it warned prescribers of increased cerebrovascular adverse events, including stroke, in elderly patients with dementia who were treated with an atypical antipsychotic (risperidone, olanzapine, or aripiprazole) vs placebo.15
Atypical antipsychotics and risk of death
In 2005, the FDA issued a black-box warning about increased all-cause mortality risk in patients with dementia treated with atypical antipsychotics for behavioral disturbance (relative risk 1.6–1.7).16
This warning was likely based on a meta-analysis by Schneider et al17 of trials in which patients with dementia were randomized to receive either an atypical antipsychotic or placebo. The death rate was 3.5% in patients treated with an atypical antipsychotic vs 2.3% in patients treated with placebo, indicating a number needed to harm of 100. The most common causes of death were cardiovascular disease and pneumonia. However, the trials in this meta-analysis included only patients who were prescribed atypical antipsychotics for ongoing management of behavioral disturbances due to dementia in either the outpatient or nursing home setting. None of the trials looked at patients who were prescribed atypical antipsychotics for a limited time in a closely monitored inpatient setting.
Effectiveness of antipsychotics
While several studies since the FDA black-box warning have shown that antipsychotics are safe, the efficacy of these drugs in delirium management remains controversial.
In a 2016 meta-analysis, Kishi et al18 found that antipsychotics were superior to placebo in terms of response rate (defined as improvement of delirium severity rating scores), with a number needed to treat of 2.
In contrast, a meta-analysis by Neufeld et al12 found that antipsychotic use was not associated with a change in delirium duration, severity, or length of stay in the hospital or intensive care unit. However, the studies in this meta-analysis varied widely in age range, study design, drug comparison, and treatment strategy (with drugs given as both prophylaxis and treatment). Thus, the results are difficult to interpret.
Kishi et al18 found no difference in the incidence of death, extrapyramidal symptoms, akathisia, or QT prolongation between patients treated with antipsychotic drugs vs placebo.
In a prospective observational study, Hatta et al19 followed 2,453 inpatients who became delirious. Only 22 (0.9%) experienced adverse events attributable to antipsychotic use, the most common being aspiration pneumonia (0.7%), followed by cardiovascular events (0.2%). Notably, no patient died of antipsychotic-related events. In this study, the antipsychotic was stopped as soon as the delirium symptoms resolved, in most cases in 3 to 7 days.
Taken together, these studies indicate that despite the risk of QT prolongation with antipsychotic use and increased rates of morbidity with antipsychotic use in dementia, time-limited management of delirium with antipsychotics is effective9–11 and safe.
SELECTING AND USING ANTIPSYCHOTICS TO TREAT DELIRIUM
Identifying a single preferred agent is difficult, since we lack enough evidence from randomized controlled trials that directly compared the various antipsychotics used in delirium management.
Both typical and atypical antipsychotics are used in clinical practice to manage delirium. The typical antipsychotic most often used is haloperidol, while the most commonly used atypical antipsychotics for delirium include olanzapine, quetiapine, risperidone, and (more recently) aripiprazole.
The American Psychiatric Association guidelines6 suggest using haloperidol because it is the antipsychotic that has been most studied for delirium,20 and we have decades of experience with its use. Despite this, recent prospective studies have suggested that the atypical antipsychotics may be better because they have a faster onset of action and lower incidence of extrapyramidal symptoms.18,21
Because we lack enough head-to-head trials comparing the efficacy of the 5 most commonly used antipsychotics for the management of delirium, and because the prospective trials that do exist show equal efficacy across the antipsychotics studied,22 we suggest considering the unique pharmacologic properties of each drug within the patient’s clinical context when selecting which antipsychotic to use.
Table 123–25 summarizes some key characteristics of the 5 most commonly used antipsychotics.
Haloperidol
Haloperidol, a typical antipsychotic, is a potent antagonist of the dopamine D2 receptor.
Haloperidol has the advantage of having the strongest evidence base for use in delirium. In addition, it is available in oral, intravenous, and intramuscular dosage forms, and it has minimal effects on vital signs, negligible anticholinergic activity, and minimal interactions with other medications.21
Intravenous haloperidol poses a significant risk of QT prolongation and so should be used judiciously in patients with preexisting cardiac conditions or other risk factors for QT prolongation as outlined above, and with careful cardiac monitoring. Parenteral haloperidol is approximately twice as potent as oral haloperidol.
Some evidence suggests a higher risk of acute dystonia and other extrapyramidal symptoms with haloperidol than with the atypical antipsychotics.21,26 In contrast, a 2013 prospective study showed that low doses of haloperidol (< 3.5 mg/day) did not result in a greater frequency of extrapyramidal symptoms.22 Nevertheless, if a patient has a history of extrapyramidal symptoms, haloperidol should likely be avoided in favor of an atypical antipsychotic.
Atypical antipsychotics
Olanzapine, quetiapine, and risperidone are atypical antipsychotics that, like haloperidol, antagonize the dopamine D2 receptor, but also have antagonist action at serotonin, histamine, and alpha-2 receptors. This multireceptor antagonism reduces the risk of extrapyramidal symptoms but increases the risk of orthostatic hypotension.
Quetiapine, in particular, imposes an unacceptably high risk of orthostatic hypotension and so is not recommended for use in delirium in the emergency department.27 Additionally, quetiapine is anticholinergic, raising concerns about constipation and urinary retention.
Although the association between fall risk and antipsychotic use remains controversial,28,29 a study found that olanzapine conferred a lower fall risk than quetiapine and risperidone.30
Of these drugs, only olanzapine is available in an intramuscular dosage form. Both risperidone and olanzapine are available in dissolvable tablets; however, they are not sublingually absorbed.
Randomized controlled trials have shown that olanzapine is effective in managing cancer-related nausea, and therefore it may be useful in managing delirium in oncology patients.31,32
Patients with Parkinson disease are exquisitely sensitive to the antidopaminergic effects of antipsychotics but are also vulnerable to delirium, so they present a unique treatment challenge. The agent of choice in patients with Parkinson disease is quetiapine, as multiple trials have shown it has no effect on the motor symptoms of Parkinson disease (reviewed by Desmarais et al in a systematic meta-analysis33).
Aripiprazole is increasingly used to manage delirium. Its mechanism of action differs from that of the other atypical antipsychotics, as it is a partial dopamine agonist. It is available in oral, orally dissolvable, and intramuscular forms. It appears to be slightly less effective than the other atypical antipsychotics,34 but it may be useful for hypoactive delirium as it is less sedating than the other agents.35 Because its effect on the QT interval is negligible, it may also be favored in patients who have a high baseline QTc or other predisposing factors for torsades de pointes.
BALANCING THE RISKS
Antipsychotic drugs have been shown to be effective and generally safe. Antipsychotics do prolong the QT interval. However, other than with intravenous administration of haloperidol, the absolute effect is minimal. Although large meta-analyses have shown a higher rate of all-cause mortality in elderly outpatients with dementia who are prescribed atypical antipsychotics, an increase in death rates has not been borne out by prospective studies focusing on hospitalized patients who receive low doses of antipsychotics for a limited time.
There are no head-to-head randomized controlled trials comparing the efficacy of all of the 5 most commonly used antipsychotics. Therefore, we suggest considering the unique psychopharmacologic properties of each agent within the patient’s clinical setting, specifically taking into account the risk of cardiac arrhythmia, risk of orthostasis and falls, history of extrapyramidal symptoms, other comorbidities such as Parkinson disease and cancer, and the desired route of administration.
At the time the patient is discharged, we recommend a careful medication reconciliation and discontinuation of the antipsychotic drug once delirium has resolved. Studies show that at least 26% of antipsychotics initiated in the hospital are continued after discharge.36,37
Current delirium consensus statements recommend limiting the use of antipsychotics to target patient distress, impediment of care, or safety, because of the putative risks of antipsychotic use in the elderly. However, a growing body of evidence shows that low-dose, time-limited antipsychotic use is safe and effective in the treatment of delirium. In fact, González et al found that delirium is an independent risk factor for death, and each 48-hour increase in delirium is associated with an increased mortality risk of 11%, suggesting that delay in treating delirium may actually increase the risk of death.38
Therefore, we must balance the risks of prescribing antipsychotics in medically vulnerable patients against the increasing burden of evidence supporting the serious risks of morbidity and mortality of delirium, as well as the costs. Much remains to be studied to optimize antipsychotic use in delirium.
- Siddiqi N, House AO, Holmes JD. Occurrence and outcome of delirium in medical in-patients: a systematic literature review. Age Ageing 2006; 35:350–364.
- Leslie DL, Marcantonio ER, Zhang Y, Leo-Summers L, Inouye SK. One-year health care costs associated with delirium in the elderly population. Arch Intern Med 2008; 168:27–32.
- Inouye SK. Delirium in older persons. N Engl J Med 2006; 354:1157–1165.
- Levkoff SE, Evans DA, Liptzin B, et al. Delirium: the occurrence and persistence of symptoms among elderly hospitalized patients. Arch Intern Med 1992; 152:334–340.
- American Psychiatric Association. Diagnostic and statistical manual of mental disorders. 5th ed. Arlington, VA: American Psychiatric Publishing; 2013.
- Trzepacz P, Breitbart W, Franklin J, Levenson J, Martini DR, Wang P; American Psychiatric Association (APA). Practice guideline for the treatment of patients with delirium. http://psychiatryonline.org/pb/assets/raw/sitewide/practice_guidelines/guidelines/delirium.pdf. Accessed July 13, 2017.
- Canadian Coalition for Seniors’ Mental Health. National guidelines for seniors’ mental health: the assessment and treatment of delirium. http://ccsmh.ca/wp-content/uploads/2016/03/NatlGuideline_Delirium.pdf. Accessed July 13, 2017.
- National Institute for Health and Care Excellence (NICE). Delirium: prevention, diagnosis and management. www.nice.org.uk/guidance/cg103. Accessed July 13, 2017.
- Bourne RS, Tahir TA, Borthwick M, Sampson EL. Drug treatment of delirium: past, present and future. J Psychosom Res 2008; 65:273–282.
- Campbell N, Boustani MA, Ayub A, et al. Pharmacological management of delirium in hospitalized adults—a systematic evidence review. J Gen Intern Med 2009; 24:848–853.
- Devlin JW, Skrobik Y. Antipsychotics for the prevention and treatment of delirium in the intensive care unit: what is their role? Harv Rev Psychiatry 2011; 19:59–67.
- Neufeld KJ, Yue J, Robinson TN, Inouye SK, Needham DM. Antipsychotic medication for prevention and treatment of delirium in hospitalized adults: a systematic review and meta-analysis. J Am Geriatr Soc 2016; 64:705–714.
- Beach SR, Celano MC, Noseworthy PA, Januzzi JL, Huffman JC. QTc prolongation, torsades de pointes, and psychotropic medications. Psychosomatics 2013; 54:1–13.
- US Food and Drug Administration (FDA). Information for healthcare professionals: haloperidol (marketed as Haldol, Haldol decanoate and Haldol lactate). www.fda.gov/Drugs/DrugSafety/ucm085203.htm. Accessed July 13, 2017.
- US Food and Drug Administration Center for Drug Evaluation and Research. Approval package for: Application Number: NDA 20-272/S-033, 20-588/S-021 & 21-444/S-004. www.accessdata.fda.gov/drugsatfda_docs/nda/2003/020588_S021_RISPERDAL_TABLETS.pdf. Accessed July 13, 2017.
- US Food and Drug Administration. Public health advisory: deaths with antipsychotics in elderly patients with behavioral disturbances. www.fda.gov/drugs/drugsafety/postmarketdrugsafetyinformationforpatientsandproviders/ucm053171. Accessed July 13, 2017.
- Schneider LS, Dagerman KS, Insel P. Risk of death with atypical antipsychotic drug treatment for dementia: meta-analysis of randomized placebo-controlled trials. JAMA 2005; 294:1934–1943.
- Kishi T, Hirota T, Matsunaga S, Iwata N. Antipsychotic medications for the treatment of delirium: a systematic review and meta-analysis of randomised controlled trials. J Neurol Neurosurg Psychiatry 2016; 87:767–774.
- Hatta K, Kishi Y, Wada K, et al. Antipsychotics for delirium in the general hospital setting in consecutive 2453 inpatients: a prospective observational study. Int J Geriatr Psychiatry 2014; 29;253–262.
- Breitbart W, Marotta R, Platt MM, et al. A double-blind trial of haloperidol, chlorpromazine, and lorazepam in the treatment of delirium in hospitalized AIDS patients. Am J Psychiatry 1996; 153:231–237.
- Wilson MP, Pepper D, Currier GW, Holloman GH Jr, Feifel D. The psychopharmacology of agitation: consensus statement of the American Association For Emergency Psychiatry Project Beta Psychopharmacology Workgroup. West J Emerg Med 2012; 13:26–34.
- Yoon HJ, Park KM, Choi WJ, et al. Efficacy and safety of haloperidol versus atypical antipsychotic medications in the treatment of delirium. BMC Psychiatry 2013; 13:240.
- American Psychiatric Association. Manual of Clinical Psychopharmacology. 8th ed. Arlington, VA: American Psychiatric Publishing; 2015.
- Conley RR, Kelly DL. Pharmacologic Treatment of Schizophrenia. 3rd ed. West Islip, NY: Professional Communications; 2007.
- American Psychiatric Association (APA). The American Psychiatric Publishing Textbook of Psychosomatic Medicine. Psychiatric Care of the Medically Ill, 2nd ed. Arlington, VA: American Psychiatric Publishing; 2011.
- Boettger S, Jenewein J, Breitbart W. Haloperidol, risperidone, olanzapine and aripiprazole in the management of delirium: a comparison of efficacy, safety, and side effects. Palliat Support Care 2015; 13:1079–1085.
- Currier GW, Trenton AJ, Walsh PG, van Wijngaarden E. A pilot, open-label study of quetiapine for treatment of moderate psychotic agitation in the emergency setting. J Psychiatr Pract 2006; 12:223–228.
- Chatterjee S, Chen H, Johnson ML, Aparasu RR. Risk of falls and fractures in older adults using atypical antipsychotic agents: a propensity score-adjusted, retrospective cohort study. Am J Geriatr Pharmacother 2012; 10:84–94.
- Rigler SK, Shireman TI, Cook-Wiens GJ, et al. Fracture risk in nursing home residents initiating antipsychotic medications. J Am Geriatr Soc 2013; 61: 715–722.
- Bozat-Emre S, Doupe M, Kozyrskyj AL, Grymonpre R, Mahmud SM. Atypical antipsychotic drug use and falls among nursing home residents in Winnipeg, Canada. Int J Geriatr Psychiatry 2015; 30:842–850.
- Navari RM, Gray SE, Kerr AC. Olanzapine versus aprepitant for the prevention of chemotherapy-induced nausea and vomiting: a randomized phase III trial. J Support Oncol 2011; 9:188–195.
- Navari RM. Olanzapine for the prevention and treatment of chronic nausea and chemotherapy-induced nausea and vomiting. Eur J Pharmacol 2014; 722:180–186.
- Desmarais P, Massoud F, Filion J, Nguyen QD, Bajsarowicz P. Quetiapine for psychosis in Parkinson disease and neurodegenerative Parkinsonian disorders: a systematic review. J Geriatr Psychiatry Neurol 2016; 29:227–236.
- Citrome L. Comparison of intramuscular ziprasidone, olanzapine, or aripiprazole for agitation: a quantitative review of efficacy and safety. J Clin Psychiatry 2007; 68:1876–1885.
- Marder SR, McQuade RD, Stock E, et al. Aripiprazole in the treatment of schizophrenia: safety and tolerability in short-term, placebo-controlled trials. Schizophr Res 2003; 61:123–136.
- Loh KP, Ramdass S, Garb JL, et al. Long-term outcomes of elders discharged on antipsychotics. J Hosp Med 2016; 11:550–555.
- Herzig SJ, Rothberg MB, Guess JR, et al. Antipsychotic use in hospitalized adults: rates, indications, and predictors. J Am Geriatr Soc 2016; 64:299–305.
- González M, Martínez G, Calderón J, et al. Impact of delirium on short-term mortality in elderly inpatients: a prospective cohort study. Psychosomatics 2009; 50:234–238.
- Siddiqi N, House AO, Holmes JD. Occurrence and outcome of delirium in medical in-patients: a systematic literature review. Age Ageing 2006; 35:350–364.
- Leslie DL, Marcantonio ER, Zhang Y, Leo-Summers L, Inouye SK. One-year health care costs associated with delirium in the elderly population. Arch Intern Med 2008; 168:27–32.
- Inouye SK. Delirium in older persons. N Engl J Med 2006; 354:1157–1165.
- Levkoff SE, Evans DA, Liptzin B, et al. Delirium: the occurrence and persistence of symptoms among elderly hospitalized patients. Arch Intern Med 1992; 152:334–340.
- American Psychiatric Association. Diagnostic and statistical manual of mental disorders. 5th ed. Arlington, VA: American Psychiatric Publishing; 2013.
- Trzepacz P, Breitbart W, Franklin J, Levenson J, Martini DR, Wang P; American Psychiatric Association (APA). Practice guideline for the treatment of patients with delirium. http://psychiatryonline.org/pb/assets/raw/sitewide/practice_guidelines/guidelines/delirium.pdf. Accessed July 13, 2017.
- Canadian Coalition for Seniors’ Mental Health. National guidelines for seniors’ mental health: the assessment and treatment of delirium. http://ccsmh.ca/wp-content/uploads/2016/03/NatlGuideline_Delirium.pdf. Accessed July 13, 2017.
- National Institute for Health and Care Excellence (NICE). Delirium: prevention, diagnosis and management. www.nice.org.uk/guidance/cg103. Accessed July 13, 2017.
- Bourne RS, Tahir TA, Borthwick M, Sampson EL. Drug treatment of delirium: past, present and future. J Psychosom Res 2008; 65:273–282.
- Campbell N, Boustani MA, Ayub A, et al. Pharmacological management of delirium in hospitalized adults—a systematic evidence review. J Gen Intern Med 2009; 24:848–853.
- Devlin JW, Skrobik Y. Antipsychotics for the prevention and treatment of delirium in the intensive care unit: what is their role? Harv Rev Psychiatry 2011; 19:59–67.
- Neufeld KJ, Yue J, Robinson TN, Inouye SK, Needham DM. Antipsychotic medication for prevention and treatment of delirium in hospitalized adults: a systematic review and meta-analysis. J Am Geriatr Soc 2016; 64:705–714.
- Beach SR, Celano MC, Noseworthy PA, Januzzi JL, Huffman JC. QTc prolongation, torsades de pointes, and psychotropic medications. Psychosomatics 2013; 54:1–13.
- US Food and Drug Administration (FDA). Information for healthcare professionals: haloperidol (marketed as Haldol, Haldol decanoate and Haldol lactate). www.fda.gov/Drugs/DrugSafety/ucm085203.htm. Accessed July 13, 2017.
- US Food and Drug Administration Center for Drug Evaluation and Research. Approval package for: Application Number: NDA 20-272/S-033, 20-588/S-021 & 21-444/S-004. www.accessdata.fda.gov/drugsatfda_docs/nda/2003/020588_S021_RISPERDAL_TABLETS.pdf. Accessed July 13, 2017.
- US Food and Drug Administration. Public health advisory: deaths with antipsychotics in elderly patients with behavioral disturbances. www.fda.gov/drugs/drugsafety/postmarketdrugsafetyinformationforpatientsandproviders/ucm053171. Accessed July 13, 2017.
- Schneider LS, Dagerman KS, Insel P. Risk of death with atypical antipsychotic drug treatment for dementia: meta-analysis of randomized placebo-controlled trials. JAMA 2005; 294:1934–1943.
- Kishi T, Hirota T, Matsunaga S, Iwata N. Antipsychotic medications for the treatment of delirium: a systematic review and meta-analysis of randomised controlled trials. J Neurol Neurosurg Psychiatry 2016; 87:767–774.
- Hatta K, Kishi Y, Wada K, et al. Antipsychotics for delirium in the general hospital setting in consecutive 2453 inpatients: a prospective observational study. Int J Geriatr Psychiatry 2014; 29;253–262.
- Breitbart W, Marotta R, Platt MM, et al. A double-blind trial of haloperidol, chlorpromazine, and lorazepam in the treatment of delirium in hospitalized AIDS patients. Am J Psychiatry 1996; 153:231–237.
- Wilson MP, Pepper D, Currier GW, Holloman GH Jr, Feifel D. The psychopharmacology of agitation: consensus statement of the American Association For Emergency Psychiatry Project Beta Psychopharmacology Workgroup. West J Emerg Med 2012; 13:26–34.
- Yoon HJ, Park KM, Choi WJ, et al. Efficacy and safety of haloperidol versus atypical antipsychotic medications in the treatment of delirium. BMC Psychiatry 2013; 13:240.
- American Psychiatric Association. Manual of Clinical Psychopharmacology. 8th ed. Arlington, VA: American Psychiatric Publishing; 2015.
- Conley RR, Kelly DL. Pharmacologic Treatment of Schizophrenia. 3rd ed. West Islip, NY: Professional Communications; 2007.
- American Psychiatric Association (APA). The American Psychiatric Publishing Textbook of Psychosomatic Medicine. Psychiatric Care of the Medically Ill, 2nd ed. Arlington, VA: American Psychiatric Publishing; 2011.
- Boettger S, Jenewein J, Breitbart W. Haloperidol, risperidone, olanzapine and aripiprazole in the management of delirium: a comparison of efficacy, safety, and side effects. Palliat Support Care 2015; 13:1079–1085.
- Currier GW, Trenton AJ, Walsh PG, van Wijngaarden E. A pilot, open-label study of quetiapine for treatment of moderate psychotic agitation in the emergency setting. J Psychiatr Pract 2006; 12:223–228.
- Chatterjee S, Chen H, Johnson ML, Aparasu RR. Risk of falls and fractures in older adults using atypical antipsychotic agents: a propensity score-adjusted, retrospective cohort study. Am J Geriatr Pharmacother 2012; 10:84–94.
- Rigler SK, Shireman TI, Cook-Wiens GJ, et al. Fracture risk in nursing home residents initiating antipsychotic medications. J Am Geriatr Soc 2013; 61: 715–722.
- Bozat-Emre S, Doupe M, Kozyrskyj AL, Grymonpre R, Mahmud SM. Atypical antipsychotic drug use and falls among nursing home residents in Winnipeg, Canada. Int J Geriatr Psychiatry 2015; 30:842–850.
- Navari RM, Gray SE, Kerr AC. Olanzapine versus aprepitant for the prevention of chemotherapy-induced nausea and vomiting: a randomized phase III trial. J Support Oncol 2011; 9:188–195.
- Navari RM. Olanzapine for the prevention and treatment of chronic nausea and chemotherapy-induced nausea and vomiting. Eur J Pharmacol 2014; 722:180–186.
- Desmarais P, Massoud F, Filion J, Nguyen QD, Bajsarowicz P. Quetiapine for psychosis in Parkinson disease and neurodegenerative Parkinsonian disorders: a systematic review. J Geriatr Psychiatry Neurol 2016; 29:227–236.
- Citrome L. Comparison of intramuscular ziprasidone, olanzapine, or aripiprazole for agitation: a quantitative review of efficacy and safety. J Clin Psychiatry 2007; 68:1876–1885.
- Marder SR, McQuade RD, Stock E, et al. Aripiprazole in the treatment of schizophrenia: safety and tolerability in short-term, placebo-controlled trials. Schizophr Res 2003; 61:123–136.
- Loh KP, Ramdass S, Garb JL, et al. Long-term outcomes of elders discharged on antipsychotics. J Hosp Med 2016; 11:550–555.
- Herzig SJ, Rothberg MB, Guess JR, et al. Antipsychotic use in hospitalized adults: rates, indications, and predictors. J Am Geriatr Soc 2016; 64:299–305.
- González M, Martínez G, Calderón J, et al. Impact of delirium on short-term mortality in elderly inpatients: a prospective cohort study. Psychosomatics 2009; 50:234–238.
KEY POINTS
- Delirium is common in hospitalized patients and often leads to loss of independence and nursing-home placement.
- The first-line treatment is to identify and address predisposing factors, provide supportive care, and manage symptoms through behavioral strategies.
- Most antipsychotic medications can prolong the QT interval and thus pose a risk for torsades de pointes. The effect is greatest with intravenous haloperidol and least with aripiprazole.
- Lacking head-to-head trials of antipsychotics, we suggest selecting the drug based on its pharmacologic properties and the patient’s clinical context.
Fixed-Dose Combination Pills Enhance Adherence and Persistence to Antihypertensive Medications
Study Overview
Objective. To evaluate long-term adherence to antihypertensive therapy among patients on fixed-dose combination medication as well as antihypertensive monotherapy; and to identify demographic and clinical risk factors associated with selection of and adherence and persistence to antihypertensive medication therapy.
Design. Retrospective cohort study using claims data from a large nationwide insurer.
Setting and participants. The study population included patients older than age 18 who initiated antihypertensive medication between 1 January 2009 and 31 December 2012 and who were continually enrolled at least 180 days before and 365 days after the index date, defined as the date of initiation of antihypertensive therapy. Patients were excluded from the study if they had previously filled any antihypertensive medication at any time prior to the index date. Patients were categorized based on the number and type of antihypertensive medications (fixed-dose combination, defined as a single pill containing multiple medications; multi-pill combination, defined as 2 or more distinct antihypertensive tablets or capsules; or single therapy, defined as only 1 medication) using National Drug Codes (NDC). Study authors also measured patient baseline characteristics, such as age, region, gender, diagnoses as defined by ICD-9 codes, patient utilization characteristics (both outpatient visits and hospitalizations) and characteristics of the initiated medication, including patient copayment and number of days of medication supplied.
Main outcome measures. The primary outcome of inte-rest was persistence, defined as having supply for any antihypertensive medication that overlapped with the 365th day after initiation (index date), whether the initiated medication or other antihypertensive. Additional outcomes included adherence to at least 1 antihypertensive in the 12 months after initiation and refilling at least 1 antihypertensive medication. To determine adherence, the study authors calculated the proportion of days the patient had any antihypertensive available to them (proportion of days covered; PDC). PDC > 80% to at least 1 antihypertensive in the 12 months after initiation was defined as “fully adherent.”
Statistical analysis utilized modified multivariable Poisson regression models and sensitivity analyses were performed. The main study comparisons focused on patients initiating fixed-dose combination therapy and monotherapy because these groups were more comparable in terms of baseline characteristics and medications initiated than the multi-pill combination group.
Main results. The study sample consisted of 484,493 patients who initiated an oral antihypertensive, including 78,958 patient initiating fixed-dose combinations, 380,269 filled a single therapy, and 22,266 who initiated multi-pill combinations. The most frequently initiated fixed-dose combination was lisinopril-hydrochlorothiazide. Lisinopril, hydrochlorothiazide, and amlodipine with the most frequently initiated monotherapy. The mean age of the study population was 47.2 years and 51.8% were women. Patients initiating multiple pill combinations were older (mean age 52.5) and tended to be sicker with more comorbidities than fixed-dose combinations or monotherapy. Patients initiating fixed-dose combination had higher prescription copayments than patients using single medication (prescription copay $14.4 versus $9.6). Patients initiating fixed-dose combinations were 9% more likely to be persistent (relative risk [RR] 1.09, 95% CI 1.08–1.10) and 13% more likely to be adherent (RR 1.13, 95% CI 1.11–1.14) than those who started on a monotherapy. Refill rates were also slightly higher among fixed-dose combination initiators (RR 1.06, 95% CI 1.05-1.07).
Conclusion. Compared with monotherapy, fixed-dose combination therapy appears to improve adherence and persistence to antihypertensive medications.
Commentary
Approximately half of US of individuals with diagnosed hypertension obtain control of their condition based on currently defined targets [1]. The most effective approach to blood pressure management has been controversial. The JNC8 [2] guidelines liberalized blood pressure targets, while recent results from the SPRINT (systolic blood pressure intervention trial) [3] indicates that lower blood pressure targets are able to prevent hypertension-related complications without significant additional risk. Given these conflicts, there is clearly ambiguity in the most effective approach to initiating antihypertensive treatment. Prior studies have shown that fewer than 50% of patients continue to take their medications just 12 months after initiation [4,5].
Fixed-dose combination therapy for blood pressure management has been cited as better for adherence and is now making its way into clinical guidelines [6–8]. However, it should be noted that fixed-dose combination therapy for blood pressure management limits dosing flexibility. Dose titration may be needed, potentially leading to additional prescriptions, thus potentially complicating the drug regimen and adding additional cost. Complicating matters further, quality metrics and reporting requirements for hypertension require primary care providers to achieve blood pressure control while also ensuring patient adherence and concomitantly avoiding side effects related to medication therapy.
This study was conducted using claims data for commercially insured patients or those with Medicare Advan-tage and is unlikely to be representative of the entire population. Additionally, the study authors did not have detailed clinical information about patients, limiting the ability to understand the true clinical implications. Further, patients may have initiated medications for indications other than hypertension. In addition, causality cannot be established given the retrospective observational cohort nature of this study.
Applications for Clinical Practice
Primary care physicians face substantial challenges in the treatment of hypertension, including with respect to selection of initial medication therapy. Results from this study add to the evidence base that fixed-dose combination therapy is more effective in obtaining blood pressure control than monotherapy or multiple-pill therapy. Medication adherence in primary care practice is challenging. Strategies such as fixed-dose combination therapy are reasonable to employ to improve medication adherence; however, costs must be considered.
—Ajay Dharod, MD, Wake Forest School of Medicine, Winston-Salem, NC
1. Gu Q, Burt VL, Dillon CF, Yoon S. Trends in antihypertensive medication use and blood pressure control among United States adults with hypertension. Circulation 2012;126:2105–14.
2. James PA, Oparil S, Carter BL, et al. 2014 Evidence-based guideline for the management of high blood pressure in adults: report from the panel members appointed to the Eighth Joint National Committee (JNC 8). JAMA 2014;311:507–20.
3. Group TSR. A randomized trial of intensive versus standard blood-pressure control. N Engl J Med 2015;373:2103–16.
4. Yeaw J, Benner JS, Walt JG, et al. Comparing adherence and persistence across 6 chronic medication classes. J Manag Care Pharm 2009;15:728–40.
5. Baroletti S, Dell’Orfano H. Medication adherence in cardiovascular disease. Circulation 2010;121:1455–8.
6. Bangalore S, Kamalakkannan G, Parkar S, Messerli FH. Fixed-dose combinations improve medication compliance: a meta-analysis. Am J Med 2007;120:713–9.
7. Gupta AK, Arshad S, Poulter NR. Compliance, safety, and effectiveness of fixed-dose combinations of antihypertensive agents. Hypertension 2010;55:399–407.
8. Pan F, Chernew ME, Fendrick AM. Impact of fixed-dose combination drugs on adherence to prescription medications. J Gen Intern Med 2008;23:611–4.
Study Overview
Objective. To evaluate long-term adherence to antihypertensive therapy among patients on fixed-dose combination medication as well as antihypertensive monotherapy; and to identify demographic and clinical risk factors associated with selection of and adherence and persistence to antihypertensive medication therapy.
Design. Retrospective cohort study using claims data from a large nationwide insurer.
Setting and participants. The study population included patients older than age 18 who initiated antihypertensive medication between 1 January 2009 and 31 December 2012 and who were continually enrolled at least 180 days before and 365 days after the index date, defined as the date of initiation of antihypertensive therapy. Patients were excluded from the study if they had previously filled any antihypertensive medication at any time prior to the index date. Patients were categorized based on the number and type of antihypertensive medications (fixed-dose combination, defined as a single pill containing multiple medications; multi-pill combination, defined as 2 or more distinct antihypertensive tablets or capsules; or single therapy, defined as only 1 medication) using National Drug Codes (NDC). Study authors also measured patient baseline characteristics, such as age, region, gender, diagnoses as defined by ICD-9 codes, patient utilization characteristics (both outpatient visits and hospitalizations) and characteristics of the initiated medication, including patient copayment and number of days of medication supplied.
Main outcome measures. The primary outcome of inte-rest was persistence, defined as having supply for any antihypertensive medication that overlapped with the 365th day after initiation (index date), whether the initiated medication or other antihypertensive. Additional outcomes included adherence to at least 1 antihypertensive in the 12 months after initiation and refilling at least 1 antihypertensive medication. To determine adherence, the study authors calculated the proportion of days the patient had any antihypertensive available to them (proportion of days covered; PDC). PDC > 80% to at least 1 antihypertensive in the 12 months after initiation was defined as “fully adherent.”
Statistical analysis utilized modified multivariable Poisson regression models and sensitivity analyses were performed. The main study comparisons focused on patients initiating fixed-dose combination therapy and monotherapy because these groups were more comparable in terms of baseline characteristics and medications initiated than the multi-pill combination group.
Main results. The study sample consisted of 484,493 patients who initiated an oral antihypertensive, including 78,958 patient initiating fixed-dose combinations, 380,269 filled a single therapy, and 22,266 who initiated multi-pill combinations. The most frequently initiated fixed-dose combination was lisinopril-hydrochlorothiazide. Lisinopril, hydrochlorothiazide, and amlodipine with the most frequently initiated monotherapy. The mean age of the study population was 47.2 years and 51.8% were women. Patients initiating multiple pill combinations were older (mean age 52.5) and tended to be sicker with more comorbidities than fixed-dose combinations or monotherapy. Patients initiating fixed-dose combination had higher prescription copayments than patients using single medication (prescription copay $14.4 versus $9.6). Patients initiating fixed-dose combinations were 9% more likely to be persistent (relative risk [RR] 1.09, 95% CI 1.08–1.10) and 13% more likely to be adherent (RR 1.13, 95% CI 1.11–1.14) than those who started on a monotherapy. Refill rates were also slightly higher among fixed-dose combination initiators (RR 1.06, 95% CI 1.05-1.07).
Conclusion. Compared with monotherapy, fixed-dose combination therapy appears to improve adherence and persistence to antihypertensive medications.
Commentary
Approximately half of US of individuals with diagnosed hypertension obtain control of their condition based on currently defined targets [1]. The most effective approach to blood pressure management has been controversial. The JNC8 [2] guidelines liberalized blood pressure targets, while recent results from the SPRINT (systolic blood pressure intervention trial) [3] indicates that lower blood pressure targets are able to prevent hypertension-related complications without significant additional risk. Given these conflicts, there is clearly ambiguity in the most effective approach to initiating antihypertensive treatment. Prior studies have shown that fewer than 50% of patients continue to take their medications just 12 months after initiation [4,5].
Fixed-dose combination therapy for blood pressure management has been cited as better for adherence and is now making its way into clinical guidelines [6–8]. However, it should be noted that fixed-dose combination therapy for blood pressure management limits dosing flexibility. Dose titration may be needed, potentially leading to additional prescriptions, thus potentially complicating the drug regimen and adding additional cost. Complicating matters further, quality metrics and reporting requirements for hypertension require primary care providers to achieve blood pressure control while also ensuring patient adherence and concomitantly avoiding side effects related to medication therapy.
This study was conducted using claims data for commercially insured patients or those with Medicare Advan-tage and is unlikely to be representative of the entire population. Additionally, the study authors did not have detailed clinical information about patients, limiting the ability to understand the true clinical implications. Further, patients may have initiated medications for indications other than hypertension. In addition, causality cannot be established given the retrospective observational cohort nature of this study.
Applications for Clinical Practice
Primary care physicians face substantial challenges in the treatment of hypertension, including with respect to selection of initial medication therapy. Results from this study add to the evidence base that fixed-dose combination therapy is more effective in obtaining blood pressure control than monotherapy or multiple-pill therapy. Medication adherence in primary care practice is challenging. Strategies such as fixed-dose combination therapy are reasonable to employ to improve medication adherence; however, costs must be considered.
—Ajay Dharod, MD, Wake Forest School of Medicine, Winston-Salem, NC
Study Overview
Objective. To evaluate long-term adherence to antihypertensive therapy among patients on fixed-dose combination medication as well as antihypertensive monotherapy; and to identify demographic and clinical risk factors associated with selection of and adherence and persistence to antihypertensive medication therapy.
Design. Retrospective cohort study using claims data from a large nationwide insurer.
Setting and participants. The study population included patients older than age 18 who initiated antihypertensive medication between 1 January 2009 and 31 December 2012 and who were continually enrolled at least 180 days before and 365 days after the index date, defined as the date of initiation of antihypertensive therapy. Patients were excluded from the study if they had previously filled any antihypertensive medication at any time prior to the index date. Patients were categorized based on the number and type of antihypertensive medications (fixed-dose combination, defined as a single pill containing multiple medications; multi-pill combination, defined as 2 or more distinct antihypertensive tablets or capsules; or single therapy, defined as only 1 medication) using National Drug Codes (NDC). Study authors also measured patient baseline characteristics, such as age, region, gender, diagnoses as defined by ICD-9 codes, patient utilization characteristics (both outpatient visits and hospitalizations) and characteristics of the initiated medication, including patient copayment and number of days of medication supplied.
Main outcome measures. The primary outcome of inte-rest was persistence, defined as having supply for any antihypertensive medication that overlapped with the 365th day after initiation (index date), whether the initiated medication or other antihypertensive. Additional outcomes included adherence to at least 1 antihypertensive in the 12 months after initiation and refilling at least 1 antihypertensive medication. To determine adherence, the study authors calculated the proportion of days the patient had any antihypertensive available to them (proportion of days covered; PDC). PDC > 80% to at least 1 antihypertensive in the 12 months after initiation was defined as “fully adherent.”
Statistical analysis utilized modified multivariable Poisson regression models and sensitivity analyses were performed. The main study comparisons focused on patients initiating fixed-dose combination therapy and monotherapy because these groups were more comparable in terms of baseline characteristics and medications initiated than the multi-pill combination group.
Main results. The study sample consisted of 484,493 patients who initiated an oral antihypertensive, including 78,958 patient initiating fixed-dose combinations, 380,269 filled a single therapy, and 22,266 who initiated multi-pill combinations. The most frequently initiated fixed-dose combination was lisinopril-hydrochlorothiazide. Lisinopril, hydrochlorothiazide, and amlodipine with the most frequently initiated monotherapy. The mean age of the study population was 47.2 years and 51.8% were women. Patients initiating multiple pill combinations were older (mean age 52.5) and tended to be sicker with more comorbidities than fixed-dose combinations or monotherapy. Patients initiating fixed-dose combination had higher prescription copayments than patients using single medication (prescription copay $14.4 versus $9.6). Patients initiating fixed-dose combinations were 9% more likely to be persistent (relative risk [RR] 1.09, 95% CI 1.08–1.10) and 13% more likely to be adherent (RR 1.13, 95% CI 1.11–1.14) than those who started on a monotherapy. Refill rates were also slightly higher among fixed-dose combination initiators (RR 1.06, 95% CI 1.05-1.07).
Conclusion. Compared with monotherapy, fixed-dose combination therapy appears to improve adherence and persistence to antihypertensive medications.
Commentary
Approximately half of US of individuals with diagnosed hypertension obtain control of their condition based on currently defined targets [1]. The most effective approach to blood pressure management has been controversial. The JNC8 [2] guidelines liberalized blood pressure targets, while recent results from the SPRINT (systolic blood pressure intervention trial) [3] indicates that lower blood pressure targets are able to prevent hypertension-related complications without significant additional risk. Given these conflicts, there is clearly ambiguity in the most effective approach to initiating antihypertensive treatment. Prior studies have shown that fewer than 50% of patients continue to take their medications just 12 months after initiation [4,5].
Fixed-dose combination therapy for blood pressure management has been cited as better for adherence and is now making its way into clinical guidelines [6–8]. However, it should be noted that fixed-dose combination therapy for blood pressure management limits dosing flexibility. Dose titration may be needed, potentially leading to additional prescriptions, thus potentially complicating the drug regimen and adding additional cost. Complicating matters further, quality metrics and reporting requirements for hypertension require primary care providers to achieve blood pressure control while also ensuring patient adherence and concomitantly avoiding side effects related to medication therapy.
This study was conducted using claims data for commercially insured patients or those with Medicare Advan-tage and is unlikely to be representative of the entire population. Additionally, the study authors did not have detailed clinical information about patients, limiting the ability to understand the true clinical implications. Further, patients may have initiated medications for indications other than hypertension. In addition, causality cannot be established given the retrospective observational cohort nature of this study.
Applications for Clinical Practice
Primary care physicians face substantial challenges in the treatment of hypertension, including with respect to selection of initial medication therapy. Results from this study add to the evidence base that fixed-dose combination therapy is more effective in obtaining blood pressure control than monotherapy or multiple-pill therapy. Medication adherence in primary care practice is challenging. Strategies such as fixed-dose combination therapy are reasonable to employ to improve medication adherence; however, costs must be considered.
—Ajay Dharod, MD, Wake Forest School of Medicine, Winston-Salem, NC
1. Gu Q, Burt VL, Dillon CF, Yoon S. Trends in antihypertensive medication use and blood pressure control among United States adults with hypertension. Circulation 2012;126:2105–14.
2. James PA, Oparil S, Carter BL, et al. 2014 Evidence-based guideline for the management of high blood pressure in adults: report from the panel members appointed to the Eighth Joint National Committee (JNC 8). JAMA 2014;311:507–20.
3. Group TSR. A randomized trial of intensive versus standard blood-pressure control. N Engl J Med 2015;373:2103–16.
4. Yeaw J, Benner JS, Walt JG, et al. Comparing adherence and persistence across 6 chronic medication classes. J Manag Care Pharm 2009;15:728–40.
5. Baroletti S, Dell’Orfano H. Medication adherence in cardiovascular disease. Circulation 2010;121:1455–8.
6. Bangalore S, Kamalakkannan G, Parkar S, Messerli FH. Fixed-dose combinations improve medication compliance: a meta-analysis. Am J Med 2007;120:713–9.
7. Gupta AK, Arshad S, Poulter NR. Compliance, safety, and effectiveness of fixed-dose combinations of antihypertensive agents. Hypertension 2010;55:399–407.
8. Pan F, Chernew ME, Fendrick AM. Impact of fixed-dose combination drugs on adherence to prescription medications. J Gen Intern Med 2008;23:611–4.
1. Gu Q, Burt VL, Dillon CF, Yoon S. Trends in antihypertensive medication use and blood pressure control among United States adults with hypertension. Circulation 2012;126:2105–14.
2. James PA, Oparil S, Carter BL, et al. 2014 Evidence-based guideline for the management of high blood pressure in adults: report from the panel members appointed to the Eighth Joint National Committee (JNC 8). JAMA 2014;311:507–20.
3. Group TSR. A randomized trial of intensive versus standard blood-pressure control. N Engl J Med 2015;373:2103–16.
4. Yeaw J, Benner JS, Walt JG, et al. Comparing adherence and persistence across 6 chronic medication classes. J Manag Care Pharm 2009;15:728–40.
5. Baroletti S, Dell’Orfano H. Medication adherence in cardiovascular disease. Circulation 2010;121:1455–8.
6. Bangalore S, Kamalakkannan G, Parkar S, Messerli FH. Fixed-dose combinations improve medication compliance: a meta-analysis. Am J Med 2007;120:713–9.
7. Gupta AK, Arshad S, Poulter NR. Compliance, safety, and effectiveness of fixed-dose combinations of antihypertensive agents. Hypertension 2010;55:399–407.
8. Pan F, Chernew ME, Fendrick AM. Impact of fixed-dose combination drugs on adherence to prescription medications. J Gen Intern Med 2008;23:611–4.
Antimicrobial Stewardship Programs: Effects on Clinical and Economic Outcomes and Future Directions
From the Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ.
Abstract
- Objective: To review the evidence evaluating inpatient antimicrobial stewardship programs (ASPs) with a focus on clinical and economic outcomes.
- Methods: Pubmed/MEDLINE and the Cochrane Database of Systematic Reviews were used to identify systematic reviews, meta-analyses, randomized controlled trials, and other relevant literature evaluating the clinical and economic impact of ASP interventions.
- Results: A total of 5 meta-analyses, 3 systematic reviews, and 10 clinical studies (2 randomized controlled, 5 observational, and 3 quasi-experimental studies) were identified for analysis. ASPs were associated with a reduction in antimicrobial consumption and use. However, due to the heterogeneity of outcomes measured among studies, the effectiveness of ASPs varied with the measures used. There are data supporting the cost savings associated with ASPs, but these studies are more sparse. Most of the available evidence supporting ASPs is of low quality, and intervention strategies vary widely among available studies.
- Conclusion: Much of the evidence reviewed supports the assertion that ASPs result in a more judicious use of antimicrobials and lead to better patient care in the inpatient setting. While clinical outcomes vary between programs, there are ubiquitous positive benefits associated with ASPs in terms of antimicrobial consumption, C. difficile infection rates, and resistance, with few adverse effects. To date, economic outcomes have been difficult to uniformly quantify, but there are data supporting the economic benefits of ASPs. As the number of ASPs continues to grow, it is imperative that standardized metrics are considered in order to accurately measure the benefits of these essential programs.
Key words: Antimicrobial stewardship; antimicrobial consumption; resistance.
Antimicrobial resistance is a public health concern that has been escalating over the years and is now identified as a global crisis [1–3]. This is partly due to the widespread use of the same antibiotics that have existed for decades, combined with a lack of sufficient novel antibiotic discovery and development [4]. Bacteria that are resistant to our last-line-of-defense medications have recently emerged, and these resistant organisms may spread to treatment-naive patients [5]. Multidrug-resistant organisms are often found, treated, and likely originate within the hospital practice setting, where antimicrobials can be prescribed by any licensed provider [6]. Upwards of 50% of antibiotics administered are unnecessary and contribute to the problem of increasing resistance [7]. The seriousness of this situation is increasingly apparent; in 2014 the World Health Organization (WHO), President Obama, and Prime Minister Cameron issued statements urging solutions to the resistance crisis [8].
While the urgency of the situation is recognized today, efforts aimed at a more judicious use of antibiotics to curb resistance began as early as the 1960s and led to the first antimicrobial stewardship programs (ASPs) [9–11]. ASPs have since been defined as “coordinated interventions designed to improve and measure the appropriate use of antimicrobial agents by promoting the selection of the optimal antimicrobial drug regimen including dosing, duration of therapy, and route of administration” [1]. The primary objectives of these types of programs are to avoid or reduce adverse events (eg, Clostridium difficile infection) and resistance driven by a shift in minimum inhibitory concentrations (MICs) and to reverse the unnecessary economic burden caused by the inappropriate prescribing of these agents [1].
This article examines the evidence evaluating the reported effectiveness of inpatient ASPs, examining both clinical and economic outcomes. In addition, we touch on ASP history, current status, and future directions in light of current trends. While ASPs are expanding into the outpatient and nursing home settings, we will limit our review here to the inpatient setting.
Historical Background
Modern antibiotics date back to the late 1930s when penicillin and sulfonamides were introduced to the medical market, and resistance to these drug classes was reported just a few years after their introduction. The same bacterial resistance mechanisms that neutralized their efficacy then exist today, and these mechanisms continue to confer resistance among those classes [5].
While “stewardship” was not described as such until the late 1990s [12], institutions have historically been proactive in creating standards around antimicrobial utilization to encourage judicious use of these agents. The earliest form of tracking antibiotic use was in the form of paper charts as “antibiotic logs” [9] and “punch cards” [10] in the 1960s. The idea of a team approach to stewardship dates back to the 1970s, with the example of Hartford Hospital in Hartford, Connecticut, which employed an antimicrobial standards model run by an infectious disease (ID) physician and clinical pharmacists [11]. In 1977, the Infectious Diseases Society of America (IDSA) released a statement that clinical pharmacists may have a substantial impact on patient care, including in ID, contributing to the idea that a team of physicians collaborating with pharmacists presents the best way to combat inappropriate medication use. Pharmacist involvement has since been shown to restrict broad overutilized antimicrobial agents and reduce the rate of C. difficile infection by a significant amount [13].
In 1997 the IDSA and the Society for Healthcare Epidemiology of America (SHEA) published guidelines to assist in the prevention of the growing issue of resistance, mentioning the importance of antimicrobial stewardship [14]. A decade later they released joint guidelines for ASP implementation [15], and the Pediatric Infectious Disease Society (PIDS) joined them in 2012 to publish a joint statement acknowledging and endorsing stewardship [16]. In 2014, the Centers of Disease Control and Prevention (CDC) recommended that every hospital should have an ASP. As of 1 January 2017, the Joint Commission requires an ASP as a standard for accreditation at hospitals, critical access hospitals, and nursing care [17]. Guidelines for implementation of an ASP are currently available through the IDSA and SHEA [1,16].
ASP Interventions
There are 2 main strategies that ASPs have to combat inappropriate antimicrobial use, and each has its own set of systematic interventions. These strategies are referred to as “prospective audit with intervention and feedback” and “prior authorization” [6]. Although most ASPs will incorporate these main strategies, each institution typically creates its own strategies and regulations independently.
Prospective audit with intervention and feedback describes the process of providing recommendations after reviewing utilization and trends of antimicrobial use. This is sometimes referred to as the “back-end” intervention, in which decisions are made after antibiotics have been administered. Interventions that are commonly used under this strategy include discontinuation of antibiotics due to culture data, de-escalation to drugs with narrower spectra, IV to oral conversions, and cessation of surgical prophylaxis [6].
Prior authorization, also referred to as a “front-end” intervention, is the process of approving medications before they are used. Interventions include a restricted formulary for antimicrobials that can be managed through a paging system or a built-in computer restriction program, as well as other guidelines and protocols for dosing and duration of therapy. Restrictions typically focus on broad spectrum antibiotics as well as the more costly drugs on formularies. These solutions reduce the need for manual intervention as technology makes it possible to create automated restriction-based services that prevent inappropriate prescribing [6].
Aside from these main techniques, other strategies are taken to achieve the goal of attaining optimal clinical outcomes while limiting further antimicrobial resistance and adverse effects. Different clinical settings have different needs, and ASPs are customized to each setting’s resources, prescribing habits, and other local specificities [1]. These differences present difficulty with interpreting diverse datasets, but certain themes arise in the literature: commonly assessed clinical outcomes of inpatient ASPs include hospital length of stay (LOS) and readmission, reinfection, mortality, and resistance rates. These outcomes are putatively driven by the more prudent use of antimicrobials, particularly by decreased rates of antimicrobial consumption.
ASP Team Members
While ASPs may differ between institutions, the staff members involved are typically the same, and leadership is always an important aspect of a program. The CDC recommends that ASP leadership consist of a program leader (an ID physician) and a pharmacy leader, who co-lead the team [18]. In addition, the Joint Commission recommends that the multidisciplinary team should include an infection preventionist (ie, infection control and hospital epidemiologist) and practitioner [17]; these specialists have a role in prevention, awareness, and policy [19]. The integration of infection control with stewardship yields the best results [15], as infection control aims to prevent antibiotic use altogether, while stewardship increases the quality of antibiotic regimens that are being prescribed [20].
It is also beneficial to incorporate a microbiologist as an integral part of the team, responsible for performing and interpreting laboratory data (ie, cultures). Nurses should be integrated into ASPs due to the overlap of their routine activities with ASP interventions [21]; other clinicians (regardless of their infectious disease clinical background), quality control, information technology, and environmental services should all collaborate in the hospital-wide systems related to the program where appropriate [18].
Evidence Review
Results
Antimicrobial Usage
The most widely studied aspect of ASPs in the current review was the effect of ASP interventions on antimicrobial consumption and use. Three systematic reviews [22–24] showed improved antibiotic prescribing practices and reduced consumption rates overall, as did several studies inside and outside the intensive care unit (ICU) [25–31].One study found an insignificant declining usage trend [32]. An important underlying facet of this observation is that even as total antibiotic consumption decreases, certain antibiotic and antibiotic class consumption may increase. This is evident in several studies, which showed that as aminoglycoside, carbapenem, and β-lactam-β-lactamase inhibitor use increased, clindamycin (1 case), glycopeptide, fluoroquinolone, and macrolide use decreased [27,28,30]. A potential confounding factor relating to decreased glycopeptide use in Bevilacqua et al [30] was that there was an epidemic of glycopeptide-resistant enterococci during the study period, potentially causing prescribers to naturally avoid it. In any case, since the aim of ASPs is to encourage a more judicious usage of antimicrobials, the observed decreases in consumption of those restricted medications is intuitive. These observations about antimicrobial consumption related to ASPs are relevant because they putatively drive improvements in clinical outcomes, especially those related to reduced adverse events associated with these agents, such as the risk of C. difficile infection with certain drugs (eg, fluoroquinolones, clindamycin, and broad-spectrum antibiotics) and prolonged antibiotic usage [33–35]. There is evidence that these benefits are not limited to antibiotics but extend to antifungal agents and possibly antivirals [22,27,36].
Utilization, Mortality, and Infection Rates
ASPs typically intend to improve patient-focused clinical parameters such as hospital LOS, hospital readmissions, mortality, and incidence of infections acquired secondary to antibiotic usage during a hospital stay, especially C. difficile infection. Most of the reviewed evidence indicates that there has been no significant LOS benefit due to stewardship interventions [24–26,32,37], and one meta-analysis noted that when overall hospital LOS was significantly reduced, ICU-specific LOS was not [22]. Generally, there was also not a significant change in hospital readmission rates [24,26,32]. However, 2 retrospective observational studies found mixed results for both LOS and readmission rates relative to ASP interventions; while both noted a significantly reduced LOS, one study [38] showed an all-cause readmission benefit in a fairly healthy patient population (but no benefit for readmissions due to the specific infections of interest), and the another [29] showed a benefit for readmissions due to infections but an increased rate of readmissions in the intervention group overall. In this latter study, hospitalizations within the previous 3 months were significantly higher at baseline for the intervention group (55% vs. 46%, P = 0.042), suggesting sicker patients and possibly providing an explanation for this unique observation. Even so, a meta-analysis of 5 studies found a significantly elevated risk of readmission associated with ASP interventions (RR 1.26, 95% CI 1.02–1.57; P = 0.03); the authors noted that non–infection-related readmissions accounted for 61% of readmissions, but this was not significantly different between intervention and non-intervention arms [37].
With regard to mortality, most studies found no significant reductions related to stewardship interventions [22,24,26,29,32]. In a prospective randomized controlled trial, all reported deaths (7/160, 4.4%) were in the ASP intervention arm, but these were attributed to the severities of infection or an underlying, chronic disease [25]. One meta-analysis, however, found that there were significant mortality reductions related to stewardship guidelines for empirical antibiotic treatment (OR 0.65, 95% CI 0.54–0.80, P < 0.001; I2 = 65%) and to de-escalation of therapy based on culture results (RR 0.44, 95% CI 0.30–0.66, P < 0.001; I2 = 59%), based on 40 and 25 studies, respectively [39]; but both results exhibited substantial heterogeneity (defined as I2 = 50%–90% [40]) among the relevant studies. Another meta-analysis found that there was no significant change in mortality related to stewardship interventions intending to improve antibiotic appropriateness (RR 0.92, 95% CI 0.69–1.2, P = 0.56; I2 = 72%) or intending to reduce excessive prescribing (RR 0.92, 95% CI 0.81–1.06, P = 0.25; I2 = 0%), but that there was a significant mortality benefit associated with interventions aimed at increasing guideline compliance for pneumonia diagnoses (RR 0.89, 95% CI 0.82–0.97, P = 0.005; I2 = 0%) [37]. In the case of Schuts et al [39], search criteria specifically sought studies that assessed clinical outcomes (eg, mortality), whereas the search of Davey et al [37] focused on studies whose aim was to improve antibiotic prescribing, with a main comparison being between restrictive and persuasive interventions; while the difference may seem subtle, the body of data compiled from these searches may characterize the ASP effect of mortality differently. No significant evidence was found to suggest that reduced antimicrobial consumption increases mortality.
Improving the use of antimicrobial agents should limit collateral damage associated with their use (eg, damage to normal flora and increased resistance), and ideally infections should be better managed. As previously mentioned, one of the concerns with antibiotic usage (particularly fluoroquinolones, macrolides, and broad-spectrum agents) is that collateral damage could lead to increased rates of C. difficile infection. One meta-analysis showed no significant reduction in the rate of C. difficile infection (as well as overall infection rate) relative to ASPs [22]; however, this finding was based on only 3 of the 26 studies analyzed, and only 1 of those 3 studies utilized restrictions for flouroquinolones and cephalosporins. An interrupted time series (ITS) study similarly found no significant reduction in C. difficile infection rate [32]; however, this study was conducted in a hospital with low baseline antibiotic prescribing (it was ranked second-to-last in terms of antibiotic usage among its peer institutions), inherently limiting the risk of C. difficile infection among patients in the pre-ASP setting. In contrast to these findings, a meta-analysis specifically designed to assess the incidence of C. difficile infection relative to stewardship programs found a significantly reduced risk of infection based on 16 studies (RR 0.48, 95% CI 0.38–0.62, P < 0.001; I2 = 76%) [41], and the systematic review conducted by Filice et al [24] found a significant benefit with regard to the C. difficile infection rate in 4 of 6 studies. These results are consistent with those presented as evidence for the impact of stewardship on C. difficile infection by the CDC [42]. Aside from C. difficile infection, one retrospective observational study found that the 14-day reinfection rate (ie, reinfection with the same infection at the same anatomical location) was significantly reduced following stewardship intervention (0% vs. 10%, P = 0.009) [29]. This finding, combined with the C. difficile infection examples, provide evidence for better infection management of ASPs.
While the general trend seems to suggest mixed or no significant benefit for several clinical outcomes, it is important to note that variation in outcomes could be due to differences in the types of ASP interventions and intervention study periods across differing programs. Davey et al [37] found variation in prescribing outcomes based on whether restrictive (ie, restrict prescriber freedom with antimicrobials) or persuasive (ie, suggest changes to prescriber) interventions were used, and on the timeframe in which they were used. At one month into an ASP, restrictive interventions resulted in better prescribing practices relative to persuasive interventions based on 27 studies (effect size 32.0%, 95% CI 2.5%–61.4%), but by 6 months the 2 were not statistically different (effect size 10.1%, 95% CI –47.5% to 66.0%). At 12 and 24 months, persuasive interventions demonstrated greater effects on prescribing outcomes, but these were not significant. These findings provide evidence that different study timeframes can impact ASP practices differently (and these already vary widely in the literature). Considering the variety of ASP interventions employed across the different studies, these factors almost certainly impact the reported antimicrobial consumption rates and outcomes to different degrees as a consequence. A high degree of heterogeneity among an analyzed dataset could itself be the reason for net non-significance within single systematic reviews and meta-analyses.
Resistance
Another goal of ASPs is the prevention of antimicrobial resistance, an area where the evidence generally suggests benefit associated with ASP interventions. Resistance rates to common troublesome organisms, such as methicillin-resistant S. aureus (MRSA), imipenem-resistant P. aeruginosa, and extended-spectrum β-lactamase (ESBL)–producing Klebsiella spp were significantly reduced in a meta-analysis; ESBL-producing E. coli infections were not, however [22]. An ITS study found significantly reduced MRSA resistance, as well as reduced Pseudomonal resistance to imipenem-cilastin and levofloxacin (all P < 0.001), but no significant changes with respect to piperacillin/tazobactam, cefepime, or amikacin resistance [32]. This study also noted increased E. coli resistance to levofloxacin and ceftriaxone (both P < 0.001). No significant changes in resistance were noted for vancomycin-resistant enterococci. It may be a reasonable expectation that decreasing inappropriate antimicrobial use may decrease long-term antimicrobial resistance; but as most studies only span a few years, only the minute changes in resistance are understood [23]. Longer duration studies are needed to better understand resistance outcomes.
Of note is a phenomenon known as the “squeezing the balloon” effect. This can be associated with ASPs, potentially resulting in paradoxically increased resistance [43]. That is, when usage restrictions are placed on certain antibiotics, the use of other non-restricted antibiotics may increase, possibly leading to increased resistance of those non-restricted antibiotics [22] (“constraining one end [of a balloon] causes the other end to bulge … limiting the use of one class of compounds may be counteracted by corresponding changes in prescribing and drug resistance that are even more ominous” [43]). Karanika et al [22] took this phenomonen into consideration, and assessed restricted and non-restricted antimicrobial consumption separately. They found a reduction in consumption for both restricted and non-restricted antibiotics, which included “high potential resistance” antibiotics, specifically carbapenems and glycopeptides. In the study conducted by Cairns et al [28], a similar effect was observed; while the use of other classes of antibiotics decreased (eg, cephalosporins and aminoglycosides), the use of β–lactam–β–lactamase inhibitor combinations actually increased by 48% (change in use: +48.2% [95% CI 21.8%–47.9%]). Hohn et al [26] noted an increased usage rate of carbapenems, even though several other classes of antibiotics had reduced usage. Unfortunately, neither study reported resistance rates, so the impact of these findings is unknown. Finally, Jenkins et al [32] assessed trends in antimicrobial use as changes in rates of consumption. Among the various antibiotics assessed in this study, the rate of flouroquinolone use decreased both before and after the intervention period, although the rate of decreased usage slowed post-ASP (the change in rate post-ASP was +2.2% [95% CI 1.4%–3.1%], P < 0.001). They observed a small (but significant) increase in resistance of E. coli to levofloxacin pre- vs. post-intervention (11.0% vs. 13.9%, P < 0.001); in contrast, a significant decrease in resistance of P. aeruginosa was observed (30.5% vs. 21.4%, P < 0.001). While these examples help illustrate the concept of changes in antibiotic usage patterns associated with an ASP, at best they approximate the “squeezing the balloon” effect since these studies present data for antibiotics that were either restricted or for which restriction was not clearly specified. The “squeezing the balloon” effect is most relevant for the unintended, potentially increased usage of non-restricted drugs secondary to ASP restrictions. Higher resistance rates among certain drug classes observed in the context of this effect would constitute a drawback to an ASP program.
Adverse Effects
Reduced toxicities and adverse effects are expected with reduced usage of antimicrobials. The systematic review conducted by Filice et al [24] examined the incidence of adverse effects related to antibiotic usage, and their findings suggest, at the least, that stewardship programs generally do not cause harm, as only 2 of the studies they examined reported adverse events. Following stewardship interventions, 5.5% of the patients deteriorated; and of those, the large majority (75%) deteriorated due to progression of oncological malignancies. To further illustrate the effect of stewardship interventions on toxicities and side effects of antimicrobials, Schuts et al demonstrated that the risk of nephrotoxicity while on antimicrobial therapy was reduced based on 14 studies of moderate heterogeneity as a result of an ASP (OR 0.46, 95% CI 0.28–0.77, P = 0.003; I2 = 34%) [39,44]. It is intuitive that reduced drug exposure results in reduced adverse effects, as such these results are expected.
Economic Outcomes
Although the focus of ASPs is often to improve clinical outcomes, economic outcomes are an important component of ASPs; these programs bring associated economic value that should be highlighted and further detailed [22,45,46]. Since clinical outcomes are often the main objective of ASPs, most available studies have been clinical effect studies (rather than economic analyses), in which economic assessments are often a secondary consideration, if included.
As a result, cost evaluations are conducted on direct cost reductions whereas indirect cost reductions are often not critically evaluated. ASPs reduce hospital expenditures by limiting hospital-acquired infections and the associated medical costs where they are effective at decreasing consumption of antimicrobials [22,45], and by reducing antibiotic misuse, iatrogenic infections, and the rates of antibiotic-resistant organisms [47]. In one retrospective observational study, annual costs of antibiotics dropped by 33% with re-implementation of an ASP, mirrored by an overall decrease in antibiotic consumption of about 10%, over the course of the intervention study period [30]. Of note is that at 1 year post-ASP re-implementation, antibiotic consumption actually increased (by 5.4%); however, because antibiotic usage had changed to more appropriate and cost-effective therapies, cost expenditures associated with antibiotics were still reduced by 13% for that year relative to pre-ASP re-implementation. Aside from economic evaluations centered on consumption rates, there is the potential to further evaluate economic benefits associated with stewardship when looking at other outcomes, including hospital LOS [22], as well as indirect costs such as morbidity and mortality, societal, and operational costs [46]. Currently, these detailed analyses are lacking. In conjunction with more standardized clinical metrics, these assessments are needed to better delineate the full cost effectiveness of ASPs.
Evidence Summary
The evidence for inpatient ASP effectiveness is promising but mixed. Much of the evidence is low-level, based on observational studies that are retrospective in nature, and systematic reviews and meta-analyses are based on these types of studies. Studies have been conducted over a range of years, and the duration of intervention periods often vary widely between studies; it is difficult to capture and account for all of the infection, prescribing, and drug availability patterns (as well as the intervention differences or new drug approvals) throughout these time periods. To complicate the matter, both the quality of data as well as the quality of the ASPs are highly variable.
As such, the findings across pooled studies for ASPs are hard to amalgamate and draw concrete conclusions from. This difficulty is due to the inherent heterogeneity when comparing smaller individual studies in systematic reviews and meta-analyses. Currently, there are numerous ways to implement an ASP, but there is not a standardized system of specific interventions or metrics. Until we can directly compare similar ASPs and interventions among various institutions, it will be challenging to generalize positive benefits from systematic reviews and meta-analyses. Currently, the CDC is involved in a new initiative in which data from various hospitals are compiled to create a surveillance database [48]. Although this is a step in the right direction for standardized metrics for stewardship, for the current review the lack of standard metrics leads to conflicting results of heterogenic studies, making it difficult to show clear benefits in clinical outcomes.
Despite the vast array of ASPs, their differences, and a range of clinical measures—many with conflicting evidence—there is a noticeable trend toward a more prudent use of antimicrobials. Based on the review of available evidence, inpatient ASPs improve patient care and preserve an important health care resource—antibiotics. As has been presented, this is demonstrated by the alterations in consumption of these agents, has ramifications for secondary outcomes such as reduced instances of C. difficile infections, resistance, and adverse effects, and overall translates into better patient care and reduced costs. But while we can conclude that the direct interventions of stewardship in reducing and restricting antibiotic use have been effective, we cannot clearly state the overall magnitude of benefit, the effectiveness of various ASP structures and components on clinical outcomes (such as LOS, mortality, etc.), and the cost savings due to the heterogeneity of the available evidence.
Future Directions
Moving forward, the future of ASPs encompasses several potential developments. First and foremost, as technological advancements continue to develop, there is a need to integrate and utilize developments in information technology (IT). Baysari et al conducted a review on the value of utilizing IT interventions, focusing mainly on decision support (stand-alone or as a component of other hospital procedures), approval, and surveillance systems [49]. There was benefit associated with these IT interventions in terms of the improvement in the appropriate use of antimicrobials (RR 1.49, 95% CI, 1.07–2.08, P < 0.05; I2 = 93%), but there was no demonstrated benefit in terms of patient mortality or hospital LOS. Aside from this study, broad evidence is still lacking to support the use of IT systems in ASPs because meaningful comparisons amongst the interventions have not been made due to widespread variability in study design and outcome measures. However, it is generally agreed that ASPs must integrate with IT systems as the widespread use of technology within the healthcare field continues to grow. Evidence needs to be provided in the form of higher quality studies centered on similar outcomes to show appropriate approaches for ASPs to leverage IT systems. At a minimum, the integration of IT into ASPs should not hinder clinical outcomes. An important consideration is the variation in practice settings where antibiotic stewardship is to be implemented; eg, a small community hospital will be less equipped to incorporate and support technological tools compared to a large tertiary teaching hospital. Therefore, any antibiotic stewardship IT intervention must be customized to meet local needs, prescriber behaviors, minimize barriers to implementation, and utilize available resources.
Another area of focus for future ASPs is the use of rapid diagnostics. Currently, when patients present with signs and symptoms of an infection, an empiric antimicrobial regimen is started that is then de-escalated as necessary; rapid testing will help to initiate appropriate therapy more quickly and increase antimicrobial effectiveness. Rapid tests range from rapid polymerase chain reaction (PCR)-based screening [50], to Verigene gram-positive blood culture (BC-GP) tests [51], next-generation sequencing methods, and matrix assisted laser desorption/ionization time-of-flight mass spectrometry (MALDI-TOF MS) [52]. Rapid diagnostic tools should be viewed as aides to assist ASPs in decreasing antibiotic consumption and improving patient outcomes; these various tools have been shown to improve clinical outcomes when integrated into ASPs, but offer little value addressing the goals of ASPs when used outside of stewardship programs and their sensitive timeframes [53].
In terms of future ASP expansion, stewardship implementation can become more unified and broad in scope. ASPs should expand to include antifungal interventions, an area which is showing progress [36]. ASPs can also be implemented in new areas throughout the hospital (eg, pediatrics and emergency room), as well as areas outside of the hospital setting, including long-term care facilities, dialysis centers, and other institutions [54–56]. A prospective randomized control study was conducted in 30 nursing homes to evaluate the use of a novel resident antimicrobial management plan (RAMP) for improved use of antimicrobials [57]. This study found that the RAMP had no associated adverse effects and suggests that ASP is an important tool in nursing homes. In addition, the general outpatient and pediatric settings show promise for ASPs [56,58,59], but more research is needed to support expansion and to identify how ASP interventions should be applied in these various practice settings. The antimicrobial stewardship interventions that will be utilized will need to be carefully delineated to consider the scale, underlying need, and potential challenges in those settings.
While the future of antibiotic stewardship is unclear, there is certainty that it will continue to develop in both scope and depth to encompass new areas of focus, new settings to improve outcomes, and employ new tools to refine approaches. An important first step for the continued development of ASPs is alignment and standardization, since without alignment it will continue to be difficult to compare outcomes. This issue is currently being addressed by a number of different organizations. With current support from the Joint Commission, the CDC, as well as the President’s Council of Advisors on Science and Technology (PCAST) [8], regulatory requirements for ASPs are well underway, and these drivers will appropriately position ASPs for further advancements. By reducing variability amongst ASPs and delineating implementation of ASPs, there can be a clear identification of both economic and clinical benefits associated with specific interventions.
Corresponding author: Luigi Brunetti, PharmD, MPH, Rutgers, The State University of New Jersey, 160 Frelinghuysen Rd., Piscataway, NJ 08854, [email protected].
Financial disclosures: None.
1. Barlam TF, Cosgrove SE, Abbo AM, et al. Implementing an antimicrobial stewardship program: guidelines by the Infectious Diseases Society of America and the Society of Healthcare Epidemiology of America. Clin Infect Dis 2016;62:e51–77.
2. Hughes D. Selection and evolution of resistance to antimicrobial drugs. IUBMB Life 2014;66:521–9.
3. World Health Organzation. The evolving threat of antimicrobial resistance – options for action. Geneva: WHO Press; 2012.
4. Gould IM, Bal AM. New antibiotic agents in the pipeline and how they can help overcome microbial resistance. Virulence 2013;4:185–91.
5. Davies J, Davies D. Origins and evolution of antibiotic resistance. Microbiol Mol Biol Rev 2010;74:417–33.
6. Owens RC Jr. Antimicrobial stewardship: concepts and strategies in the 21st century. Diagn Microbiol Infect Dis 2008;61:110–28.
7. Antibiotic resistance threats in the United States, 2013 [Internet]. Centers for Disease Control and Prevention. Available at www.cdc.gov/drugresistance/pdf/ar-threats-2013-508.pdf.
8. Nathan C, Cars O. Antibiotic resistance – problems, progress, prospects. N Engl J Med 2014;371:1761–3.
9. McGoldrick, M. Antimicrobial stewardship. Home Healthc Nurse 2014;32:559–60.
10. Ruedy J. A method of determining patterns of use of antibacterial drugs. Can Med Assoc J 1966;95:807–12.
11. Briceland LL, Nightingdale CH, Quintiliani R, et al. Antibiotic streamlining from combination therapy to monotherapy utilizing an interdisciplinary approach. Arch Inter Med 1988;148:2019–22.
12. McGowan JE Jr, Gerding DN. Does antibiotic restriction prevent resistance? New Horiz 1996;4: 370–6.
13. Cappelletty D, Jacobs D. Evaluating the impact of a pharmacist’s absence from an antimicrobial stewardship team. Am J Health Syst Pharm 2013;70:1065–69.
14. Shales DM, Gerding DN, John JF Jr, et al. Society for Healthcare Epidemiology of America and Infectious Diseases Society of America Joint Committee on the prevention of antimicrobial resistance: guidelines for the prevention of antimicrobial resistance in hospitals. Infect Control Hosp Epidemiol 1997;18:275–91.
15. Dellit TH, Owens RC, McGowan JE, et al. Infectious Diseases Society of America and the Society for Healthcare Epidemiology of America guidelines for developing an institutional program to enhance antimicrobial stewardship. Clin Infect Dis 2007;44:159–77.
16. Policy statement on antimicrobial stewardship by the Society for Healthcare Epidemiology of America (SHEA), the Infectious Diseases Society of America (IDSA), and the Pediatric Infectious Diseases Society (PIDS). Infect Ctrl Hosp Epidemiol 2012;33:322–7.
17. The Joint Commission. Approved: New antimicrobial stewardship standard. Joint Commission Perspectives 2016;36:1–8.
18. Pollack LA, Srinivasan A. Core elements of hospital antibiotic stewardship programs from the Centers for Disease Control and Prevention. Clin Infect Dis 2014;59(Suppl 3):S97–100.
19. Moody J. Infection preventionists have a role in accelerating progress toward preventing the emergence and cross-transmission of MDROs. Prevention Strategist 2012 Summer:52–6.
20. Spellberg B, Bartlett JG, Gilbert DN. The future of antibiotics and resistance. N Engl J Med 2013;368:299–302.
21. Olans RN, Olans RD, Demaria A. The critical role of the staff nurse in antimicrobial stewardship--unrecognized, but already there. Clin Infect Dis 2016;62:84–9.
22. Karanika S, Paudel S, Grigoras C, et al. Systematic review and meta-analysis of clinical and economic outcomes from the implementation of hospital-based antimicrobial stewardship programs. Antimicrob Agents Chemother 2016;60:4840–52.
23. Wagner B, Filice GA, Drekonja D, et al. Antimicrobial stewardship programs in inpatient hospital settings: a systematic review. Infect Control Hosp Epidemiol 2014;35:1209–28.
24. Filice G, Drekonja D, Greer N, et al. Antimicrobial stewardship programs in inpatient settings: a systematic review. VA-ESP Project #09-009; 2013.
25. Cairns KA, Doyle JS, Trevillyan JM, et al. The impact of a multidisciplinary antimicrobial stewardship team on the timeliness of antimicrobial therapy in patients with positive blood cultures: a randomized controlled trial. J Antimicrob Chemother 2016;71:3276–83.
26. Hohn A, Heising B, Hertel S, et al. Antibiotic consumption after implementation of a procalcitonin-guided antimicrobial stewardship programme in surgical patients admitted to an intensive care unit: a retrospective before-and-after analysis. Infection 2015;43:405–12.
27. Singh S, Zhang YZ, Chalkley S, et al. A three-point time series study of antibiotic usage on an intensive care unit, following an antibiotic stewardship programme, after an outbreak of multi-resistant Acinetobacter baumannii. Eur J Clin Microbiol Infect Dis 2015;34:1893–900.
28. Cairns KA, Jenney AW, Abbott IJ, et al. Prescribing trends before and after implementation of an antimicrobial stewardship program. Med J Aust 2013;198:262–6.
29. Liew YX, Lee W, Loh JC, et al. Impact of an antimicrobial stewardship programme on patient safety in Singapore General Hospital. Int J Antimicrob Agents 2012;40:55–60.
30. Bevilacqua S, Demoré B, Boschetti E, et al. 15 years of antibiotic stewardship policy in the Nancy Teaching Hospital. Med Mal Infect 2011;41:532–9.
31. Danaher PJ, Milazzo NA, Kerr KJ, et al. The antibiotic support team--a successful educational approach to antibiotic stewardship. Mil Med 2009;174:201–5.
32. Jenkins TC, Knepper BC, Shihadeh K, et al. Long-term outcomes of an antimicrobial stewardship program implemented in a hospital with low baseline antibiotic use. Infect Control Hosp Epidemiol 2015;36:664–72.
33. Brown KA, Khanafer N, Daneman N, Fisman DN. Meta-analysis of antibiotics and the risk of community-associated Clostridium difficile infection. Antimicrob Agents Chemother 2013;57:2326–32.
34. Deshpande A, Pasupuleti V, Thota P, et al. Community-associated Clostridium difficile infection and antibiotics: a meta-analysis. J Antimicrob Chemother 2013;68:1951–61.
35. Slimings C, Riley TV. Antibiotics and hospital-acquired Clostridium difficile infection: update of systematic review and meta-analysis. J Antimicrob Chemother 2014;69:881–91.
36. Antworth A, Collins CD, Kunapuli A, et al. Impact of an antimicrobial stewardship program comprehensive care bundle on management of candidemia. Pharmacotherapy 2013;33:137–43.
37. Davey P, Brown E, Charani E, et al. Interventions to improve antibiotic prescribing practices for hospital inpatients. Cochrane Database Syst Rev 2013;4:CD003543.
38. Pasquale TR, Trienski TL, Olexia DE, et al. Impact of an antimicrobial stewardship program on patients with acute bacterial skin and skin structure infections. Am J Health Syst Pharm 2014;71:1136–9.
39. Schuts EC, Hulscher ME, Mouton JW, et al. Current evidence on hospital antimicrobial stewardship objectives: a systematic review and meta-analysis. Lancet Infect Dis 2016;16:847–56.
40. Higgins JPT, Green S, editors. Identifying and measuring heterogeneity. Cochrane Handbook for Systematic Reviews of Interventions, version 5.1.0. [Internet]. The Cochrane Collaboration, March 2011. Available at http://handbook.cochrane.org/chapter_9/9_5_2_identifying_and_measuring_heterogeneity.htm.
41. Feazel LM, Malhotra A, Perencevich EN, et al. Effect of antibiotic stewardship programmes on Clostridium difficile incidence: a systematic review and meta-analysis. J Antimicrob Chemother 2014;69:1748–54.
42. Impact of antibiotic stewardship programs on Clostridium difficile (C. diff) infections [Internet]. Centers for Disease Control and Prevention. [Updated 2016 May 13; cited 2016 Oct 11]. Available at www.cdc.gov/getsmart/healthcare/evidence/asp-int-cdiff.html.
43. Burke JP. Antibiotic resistance – squeezing the balloon? JAMA 1998;280:1270–1.
44. This nephrotoxicity result is corrected from the originally published result; communicated by Jan M Prins on behalf of the authors for reference [39]. Prins, JM (Department of Internal Medicine, Division of Infectious Diseases, Academic Medical Centre, Amsterdam, Netherlands). Email communication with Joseph Eckart (Pharmacy Practice & Administration, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ). 2016 Oct 9.
45. Coulter S, Merollini K, Roberts JA, et al. The need for cost-effectiveness analyses of antimicrobial stewardship programmes: a structured review. Int J Antimicrob Agents 2015;46:140–9.
46. Dik J, Vemer P, Friedrich A, et al. Financial evaluations of antibiotic stewardship programs—a systematic review. Frontiers Microbiol 2015;6:317.
47. Campbell KA, Stein S, Looze C, Bosco JA. Antibiotic stewardship in orthopaedic surgery: principles and practice. J Am Acad Orthop Surg 2014;22:772–81.
48. Surveillance for antimicrobial use and antimicrobial resistance options, 2015 [Internet]. Centers for Disease Control and Prevention. [Updated 2016 May 3; cited 2016 Nov 22]. Available at www.cdc.gov/nhsn/acute-care-hospital/aur/index.html.
49. Baysari MT, Lehnbom EC, Li L, Hargreaves A, et al. The effectiveness of information technology to improve antimicrobial prescribing in hospitals: a systematic review and meta-analysis. Int J Med Inform. 2016;92:15-34.
50. Bauer KA, West JE, Balada-llasat JM, et al. An antimicrobial stewardship program’s impact with rapid polymerase chain reaction methicillin-resistant Staphylococcus aureus/S. aureus blood culture test in patients with S. aureus bacteremia. Clin Infect Dis 2010;51:1074–80.
51. Sango A, Mccarter YS, Johnson D, et al. Stewardship approach for optimizing antimicrobial therapy through use of a rapid microarray assay on blood cultures positive for Enterococcus species. J Clin Microbiol 2013;51:4008–11.
52. Perez KK, Olsen RJ, Musick WL, et al. Integrating rapid diagnostics and antimicrobial stewardship improves outcomes in patients with antibiotic-resistant Gram-negative bacteremia. J Infect 2014;69:216–25.
53. Bauer KA, Perez KK, Forrest GN, Goff DA. Review of rapid diagnostic tests used by antimicrobial stewardship programs. Clin Infect Dis 2014;59 Suppl 3:S134–145.
54. Dyar OJ, Pagani L, Pulcini C. Strategies and challenges of antimicrobial stewardship in long-term care facilities. Clin Microbiol Infect 2015;21:10–9.
55. D’Agata EM. Antimicrobial use and stewardship programs among dialysis centers. Semin Dial 2013;26:457–64.
56. Smith MJ, Gerber JS, Hersh AL. Inpatient antimicrobial stewardship in pediatrics: a systematic review. J Pediatric Infect Dis Soc 2015;4:e127–135.
57. Fleet E, Gopal Rao G, Patel B, et al. Impact of implementation of a novel antimicrobial stewardship tool on antibiotic use in nursing homes: a prospective cluster randomized control pilot study. J Antimicrob Chemother 2014;69:2265–73.
58. Drekonja DM, Filice GA, Greer N, et al. Antimicrobial stewardship in outpatient settings: a systematic review. Infect Control Hosp Epidemiol 2015;36:142–52.
59. Drekonja D, Filice G, Greer N, et al. Antimicrobial stewardship programs in outpatient settings: a systematic review. VA-ESP Project #09-009; 2014.
60. Zhang YZ, Singh S. Antibiotic stewardship programmes in intensive care units: why, how, and where are they leading us. World J Crit Care Med 2015;4:13–28. (referenced in online Table)
From the Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ.
Abstract
- Objective: To review the evidence evaluating inpatient antimicrobial stewardship programs (ASPs) with a focus on clinical and economic outcomes.
- Methods: Pubmed/MEDLINE and the Cochrane Database of Systematic Reviews were used to identify systematic reviews, meta-analyses, randomized controlled trials, and other relevant literature evaluating the clinical and economic impact of ASP interventions.
- Results: A total of 5 meta-analyses, 3 systematic reviews, and 10 clinical studies (2 randomized controlled, 5 observational, and 3 quasi-experimental studies) were identified for analysis. ASPs were associated with a reduction in antimicrobial consumption and use. However, due to the heterogeneity of outcomes measured among studies, the effectiveness of ASPs varied with the measures used. There are data supporting the cost savings associated with ASPs, but these studies are more sparse. Most of the available evidence supporting ASPs is of low quality, and intervention strategies vary widely among available studies.
- Conclusion: Much of the evidence reviewed supports the assertion that ASPs result in a more judicious use of antimicrobials and lead to better patient care in the inpatient setting. While clinical outcomes vary between programs, there are ubiquitous positive benefits associated with ASPs in terms of antimicrobial consumption, C. difficile infection rates, and resistance, with few adverse effects. To date, economic outcomes have been difficult to uniformly quantify, but there are data supporting the economic benefits of ASPs. As the number of ASPs continues to grow, it is imperative that standardized metrics are considered in order to accurately measure the benefits of these essential programs.
Key words: Antimicrobial stewardship; antimicrobial consumption; resistance.
Antimicrobial resistance is a public health concern that has been escalating over the years and is now identified as a global crisis [1–3]. This is partly due to the widespread use of the same antibiotics that have existed for decades, combined with a lack of sufficient novel antibiotic discovery and development [4]. Bacteria that are resistant to our last-line-of-defense medications have recently emerged, and these resistant organisms may spread to treatment-naive patients [5]. Multidrug-resistant organisms are often found, treated, and likely originate within the hospital practice setting, where antimicrobials can be prescribed by any licensed provider [6]. Upwards of 50% of antibiotics administered are unnecessary and contribute to the problem of increasing resistance [7]. The seriousness of this situation is increasingly apparent; in 2014 the World Health Organization (WHO), President Obama, and Prime Minister Cameron issued statements urging solutions to the resistance crisis [8].
While the urgency of the situation is recognized today, efforts aimed at a more judicious use of antibiotics to curb resistance began as early as the 1960s and led to the first antimicrobial stewardship programs (ASPs) [9–11]. ASPs have since been defined as “coordinated interventions designed to improve and measure the appropriate use of antimicrobial agents by promoting the selection of the optimal antimicrobial drug regimen including dosing, duration of therapy, and route of administration” [1]. The primary objectives of these types of programs are to avoid or reduce adverse events (eg, Clostridium difficile infection) and resistance driven by a shift in minimum inhibitory concentrations (MICs) and to reverse the unnecessary economic burden caused by the inappropriate prescribing of these agents [1].
This article examines the evidence evaluating the reported effectiveness of inpatient ASPs, examining both clinical and economic outcomes. In addition, we touch on ASP history, current status, and future directions in light of current trends. While ASPs are expanding into the outpatient and nursing home settings, we will limit our review here to the inpatient setting.
Historical Background
Modern antibiotics date back to the late 1930s when penicillin and sulfonamides were introduced to the medical market, and resistance to these drug classes was reported just a few years after their introduction. The same bacterial resistance mechanisms that neutralized their efficacy then exist today, and these mechanisms continue to confer resistance among those classes [5].
While “stewardship” was not described as such until the late 1990s [12], institutions have historically been proactive in creating standards around antimicrobial utilization to encourage judicious use of these agents. The earliest form of tracking antibiotic use was in the form of paper charts as “antibiotic logs” [9] and “punch cards” [10] in the 1960s. The idea of a team approach to stewardship dates back to the 1970s, with the example of Hartford Hospital in Hartford, Connecticut, which employed an antimicrobial standards model run by an infectious disease (ID) physician and clinical pharmacists [11]. In 1977, the Infectious Diseases Society of America (IDSA) released a statement that clinical pharmacists may have a substantial impact on patient care, including in ID, contributing to the idea that a team of physicians collaborating with pharmacists presents the best way to combat inappropriate medication use. Pharmacist involvement has since been shown to restrict broad overutilized antimicrobial agents and reduce the rate of C. difficile infection by a significant amount [13].
In 1997 the IDSA and the Society for Healthcare Epidemiology of America (SHEA) published guidelines to assist in the prevention of the growing issue of resistance, mentioning the importance of antimicrobial stewardship [14]. A decade later they released joint guidelines for ASP implementation [15], and the Pediatric Infectious Disease Society (PIDS) joined them in 2012 to publish a joint statement acknowledging and endorsing stewardship [16]. In 2014, the Centers of Disease Control and Prevention (CDC) recommended that every hospital should have an ASP. As of 1 January 2017, the Joint Commission requires an ASP as a standard for accreditation at hospitals, critical access hospitals, and nursing care [17]. Guidelines for implementation of an ASP are currently available through the IDSA and SHEA [1,16].
ASP Interventions
There are 2 main strategies that ASPs have to combat inappropriate antimicrobial use, and each has its own set of systematic interventions. These strategies are referred to as “prospective audit with intervention and feedback” and “prior authorization” [6]. Although most ASPs will incorporate these main strategies, each institution typically creates its own strategies and regulations independently.
Prospective audit with intervention and feedback describes the process of providing recommendations after reviewing utilization and trends of antimicrobial use. This is sometimes referred to as the “back-end” intervention, in which decisions are made after antibiotics have been administered. Interventions that are commonly used under this strategy include discontinuation of antibiotics due to culture data, de-escalation to drugs with narrower spectra, IV to oral conversions, and cessation of surgical prophylaxis [6].
Prior authorization, also referred to as a “front-end” intervention, is the process of approving medications before they are used. Interventions include a restricted formulary for antimicrobials that can be managed through a paging system or a built-in computer restriction program, as well as other guidelines and protocols for dosing and duration of therapy. Restrictions typically focus on broad spectrum antibiotics as well as the more costly drugs on formularies. These solutions reduce the need for manual intervention as technology makes it possible to create automated restriction-based services that prevent inappropriate prescribing [6].
Aside from these main techniques, other strategies are taken to achieve the goal of attaining optimal clinical outcomes while limiting further antimicrobial resistance and adverse effects. Different clinical settings have different needs, and ASPs are customized to each setting’s resources, prescribing habits, and other local specificities [1]. These differences present difficulty with interpreting diverse datasets, but certain themes arise in the literature: commonly assessed clinical outcomes of inpatient ASPs include hospital length of stay (LOS) and readmission, reinfection, mortality, and resistance rates. These outcomes are putatively driven by the more prudent use of antimicrobials, particularly by decreased rates of antimicrobial consumption.
ASP Team Members
While ASPs may differ between institutions, the staff members involved are typically the same, and leadership is always an important aspect of a program. The CDC recommends that ASP leadership consist of a program leader (an ID physician) and a pharmacy leader, who co-lead the team [18]. In addition, the Joint Commission recommends that the multidisciplinary team should include an infection preventionist (ie, infection control and hospital epidemiologist) and practitioner [17]; these specialists have a role in prevention, awareness, and policy [19]. The integration of infection control with stewardship yields the best results [15], as infection control aims to prevent antibiotic use altogether, while stewardship increases the quality of antibiotic regimens that are being prescribed [20].
It is also beneficial to incorporate a microbiologist as an integral part of the team, responsible for performing and interpreting laboratory data (ie, cultures). Nurses should be integrated into ASPs due to the overlap of their routine activities with ASP interventions [21]; other clinicians (regardless of their infectious disease clinical background), quality control, information technology, and environmental services should all collaborate in the hospital-wide systems related to the program where appropriate [18].
Evidence Review
Results
Antimicrobial Usage
The most widely studied aspect of ASPs in the current review was the effect of ASP interventions on antimicrobial consumption and use. Three systematic reviews [22–24] showed improved antibiotic prescribing practices and reduced consumption rates overall, as did several studies inside and outside the intensive care unit (ICU) [25–31].One study found an insignificant declining usage trend [32]. An important underlying facet of this observation is that even as total antibiotic consumption decreases, certain antibiotic and antibiotic class consumption may increase. This is evident in several studies, which showed that as aminoglycoside, carbapenem, and β-lactam-β-lactamase inhibitor use increased, clindamycin (1 case), glycopeptide, fluoroquinolone, and macrolide use decreased [27,28,30]. A potential confounding factor relating to decreased glycopeptide use in Bevilacqua et al [30] was that there was an epidemic of glycopeptide-resistant enterococci during the study period, potentially causing prescribers to naturally avoid it. In any case, since the aim of ASPs is to encourage a more judicious usage of antimicrobials, the observed decreases in consumption of those restricted medications is intuitive. These observations about antimicrobial consumption related to ASPs are relevant because they putatively drive improvements in clinical outcomes, especially those related to reduced adverse events associated with these agents, such as the risk of C. difficile infection with certain drugs (eg, fluoroquinolones, clindamycin, and broad-spectrum antibiotics) and prolonged antibiotic usage [33–35]. There is evidence that these benefits are not limited to antibiotics but extend to antifungal agents and possibly antivirals [22,27,36].
Utilization, Mortality, and Infection Rates
ASPs typically intend to improve patient-focused clinical parameters such as hospital LOS, hospital readmissions, mortality, and incidence of infections acquired secondary to antibiotic usage during a hospital stay, especially C. difficile infection. Most of the reviewed evidence indicates that there has been no significant LOS benefit due to stewardship interventions [24–26,32,37], and one meta-analysis noted that when overall hospital LOS was significantly reduced, ICU-specific LOS was not [22]. Generally, there was also not a significant change in hospital readmission rates [24,26,32]. However, 2 retrospective observational studies found mixed results for both LOS and readmission rates relative to ASP interventions; while both noted a significantly reduced LOS, one study [38] showed an all-cause readmission benefit in a fairly healthy patient population (but no benefit for readmissions due to the specific infections of interest), and the another [29] showed a benefit for readmissions due to infections but an increased rate of readmissions in the intervention group overall. In this latter study, hospitalizations within the previous 3 months were significantly higher at baseline for the intervention group (55% vs. 46%, P = 0.042), suggesting sicker patients and possibly providing an explanation for this unique observation. Even so, a meta-analysis of 5 studies found a significantly elevated risk of readmission associated with ASP interventions (RR 1.26, 95% CI 1.02–1.57; P = 0.03); the authors noted that non–infection-related readmissions accounted for 61% of readmissions, but this was not significantly different between intervention and non-intervention arms [37].
With regard to mortality, most studies found no significant reductions related to stewardship interventions [22,24,26,29,32]. In a prospective randomized controlled trial, all reported deaths (7/160, 4.4%) were in the ASP intervention arm, but these were attributed to the severities of infection or an underlying, chronic disease [25]. One meta-analysis, however, found that there were significant mortality reductions related to stewardship guidelines for empirical antibiotic treatment (OR 0.65, 95% CI 0.54–0.80, P < 0.001; I2 = 65%) and to de-escalation of therapy based on culture results (RR 0.44, 95% CI 0.30–0.66, P < 0.001; I2 = 59%), based on 40 and 25 studies, respectively [39]; but both results exhibited substantial heterogeneity (defined as I2 = 50%–90% [40]) among the relevant studies. Another meta-analysis found that there was no significant change in mortality related to stewardship interventions intending to improve antibiotic appropriateness (RR 0.92, 95% CI 0.69–1.2, P = 0.56; I2 = 72%) or intending to reduce excessive prescribing (RR 0.92, 95% CI 0.81–1.06, P = 0.25; I2 = 0%), but that there was a significant mortality benefit associated with interventions aimed at increasing guideline compliance for pneumonia diagnoses (RR 0.89, 95% CI 0.82–0.97, P = 0.005; I2 = 0%) [37]. In the case of Schuts et al [39], search criteria specifically sought studies that assessed clinical outcomes (eg, mortality), whereas the search of Davey et al [37] focused on studies whose aim was to improve antibiotic prescribing, with a main comparison being between restrictive and persuasive interventions; while the difference may seem subtle, the body of data compiled from these searches may characterize the ASP effect of mortality differently. No significant evidence was found to suggest that reduced antimicrobial consumption increases mortality.
Improving the use of antimicrobial agents should limit collateral damage associated with their use (eg, damage to normal flora and increased resistance), and ideally infections should be better managed. As previously mentioned, one of the concerns with antibiotic usage (particularly fluoroquinolones, macrolides, and broad-spectrum agents) is that collateral damage could lead to increased rates of C. difficile infection. One meta-analysis showed no significant reduction in the rate of C. difficile infection (as well as overall infection rate) relative to ASPs [22]; however, this finding was based on only 3 of the 26 studies analyzed, and only 1 of those 3 studies utilized restrictions for flouroquinolones and cephalosporins. An interrupted time series (ITS) study similarly found no significant reduction in C. difficile infection rate [32]; however, this study was conducted in a hospital with low baseline antibiotic prescribing (it was ranked second-to-last in terms of antibiotic usage among its peer institutions), inherently limiting the risk of C. difficile infection among patients in the pre-ASP setting. In contrast to these findings, a meta-analysis specifically designed to assess the incidence of C. difficile infection relative to stewardship programs found a significantly reduced risk of infection based on 16 studies (RR 0.48, 95% CI 0.38–0.62, P < 0.001; I2 = 76%) [41], and the systematic review conducted by Filice et al [24] found a significant benefit with regard to the C. difficile infection rate in 4 of 6 studies. These results are consistent with those presented as evidence for the impact of stewardship on C. difficile infection by the CDC [42]. Aside from C. difficile infection, one retrospective observational study found that the 14-day reinfection rate (ie, reinfection with the same infection at the same anatomical location) was significantly reduced following stewardship intervention (0% vs. 10%, P = 0.009) [29]. This finding, combined with the C. difficile infection examples, provide evidence for better infection management of ASPs.
While the general trend seems to suggest mixed or no significant benefit for several clinical outcomes, it is important to note that variation in outcomes could be due to differences in the types of ASP interventions and intervention study periods across differing programs. Davey et al [37] found variation in prescribing outcomes based on whether restrictive (ie, restrict prescriber freedom with antimicrobials) or persuasive (ie, suggest changes to prescriber) interventions were used, and on the timeframe in which they were used. At one month into an ASP, restrictive interventions resulted in better prescribing practices relative to persuasive interventions based on 27 studies (effect size 32.0%, 95% CI 2.5%–61.4%), but by 6 months the 2 were not statistically different (effect size 10.1%, 95% CI –47.5% to 66.0%). At 12 and 24 months, persuasive interventions demonstrated greater effects on prescribing outcomes, but these were not significant. These findings provide evidence that different study timeframes can impact ASP practices differently (and these already vary widely in the literature). Considering the variety of ASP interventions employed across the different studies, these factors almost certainly impact the reported antimicrobial consumption rates and outcomes to different degrees as a consequence. A high degree of heterogeneity among an analyzed dataset could itself be the reason for net non-significance within single systematic reviews and meta-analyses.
Resistance
Another goal of ASPs is the prevention of antimicrobial resistance, an area where the evidence generally suggests benefit associated with ASP interventions. Resistance rates to common troublesome organisms, such as methicillin-resistant S. aureus (MRSA), imipenem-resistant P. aeruginosa, and extended-spectrum β-lactamase (ESBL)–producing Klebsiella spp were significantly reduced in a meta-analysis; ESBL-producing E. coli infections were not, however [22]. An ITS study found significantly reduced MRSA resistance, as well as reduced Pseudomonal resistance to imipenem-cilastin and levofloxacin (all P < 0.001), but no significant changes with respect to piperacillin/tazobactam, cefepime, or amikacin resistance [32]. This study also noted increased E. coli resistance to levofloxacin and ceftriaxone (both P < 0.001). No significant changes in resistance were noted for vancomycin-resistant enterococci. It may be a reasonable expectation that decreasing inappropriate antimicrobial use may decrease long-term antimicrobial resistance; but as most studies only span a few years, only the minute changes in resistance are understood [23]. Longer duration studies are needed to better understand resistance outcomes.
Of note is a phenomenon known as the “squeezing the balloon” effect. This can be associated with ASPs, potentially resulting in paradoxically increased resistance [43]. That is, when usage restrictions are placed on certain antibiotics, the use of other non-restricted antibiotics may increase, possibly leading to increased resistance of those non-restricted antibiotics [22] (“constraining one end [of a balloon] causes the other end to bulge … limiting the use of one class of compounds may be counteracted by corresponding changes in prescribing and drug resistance that are even more ominous” [43]). Karanika et al [22] took this phenomonen into consideration, and assessed restricted and non-restricted antimicrobial consumption separately. They found a reduction in consumption for both restricted and non-restricted antibiotics, which included “high potential resistance” antibiotics, specifically carbapenems and glycopeptides. In the study conducted by Cairns et al [28], a similar effect was observed; while the use of other classes of antibiotics decreased (eg, cephalosporins and aminoglycosides), the use of β–lactam–β–lactamase inhibitor combinations actually increased by 48% (change in use: +48.2% [95% CI 21.8%–47.9%]). Hohn et al [26] noted an increased usage rate of carbapenems, even though several other classes of antibiotics had reduced usage. Unfortunately, neither study reported resistance rates, so the impact of these findings is unknown. Finally, Jenkins et al [32] assessed trends in antimicrobial use as changes in rates of consumption. Among the various antibiotics assessed in this study, the rate of flouroquinolone use decreased both before and after the intervention period, although the rate of decreased usage slowed post-ASP (the change in rate post-ASP was +2.2% [95% CI 1.4%–3.1%], P < 0.001). They observed a small (but significant) increase in resistance of E. coli to levofloxacin pre- vs. post-intervention (11.0% vs. 13.9%, P < 0.001); in contrast, a significant decrease in resistance of P. aeruginosa was observed (30.5% vs. 21.4%, P < 0.001). While these examples help illustrate the concept of changes in antibiotic usage patterns associated with an ASP, at best they approximate the “squeezing the balloon” effect since these studies present data for antibiotics that were either restricted or for which restriction was not clearly specified. The “squeezing the balloon” effect is most relevant for the unintended, potentially increased usage of non-restricted drugs secondary to ASP restrictions. Higher resistance rates among certain drug classes observed in the context of this effect would constitute a drawback to an ASP program.
Adverse Effects
Reduced toxicities and adverse effects are expected with reduced usage of antimicrobials. The systematic review conducted by Filice et al [24] examined the incidence of adverse effects related to antibiotic usage, and their findings suggest, at the least, that stewardship programs generally do not cause harm, as only 2 of the studies they examined reported adverse events. Following stewardship interventions, 5.5% of the patients deteriorated; and of those, the large majority (75%) deteriorated due to progression of oncological malignancies. To further illustrate the effect of stewardship interventions on toxicities and side effects of antimicrobials, Schuts et al demonstrated that the risk of nephrotoxicity while on antimicrobial therapy was reduced based on 14 studies of moderate heterogeneity as a result of an ASP (OR 0.46, 95% CI 0.28–0.77, P = 0.003; I2 = 34%) [39,44]. It is intuitive that reduced drug exposure results in reduced adverse effects, as such these results are expected.
Economic Outcomes
Although the focus of ASPs is often to improve clinical outcomes, economic outcomes are an important component of ASPs; these programs bring associated economic value that should be highlighted and further detailed [22,45,46]. Since clinical outcomes are often the main objective of ASPs, most available studies have been clinical effect studies (rather than economic analyses), in which economic assessments are often a secondary consideration, if included.
As a result, cost evaluations are conducted on direct cost reductions whereas indirect cost reductions are often not critically evaluated. ASPs reduce hospital expenditures by limiting hospital-acquired infections and the associated medical costs where they are effective at decreasing consumption of antimicrobials [22,45], and by reducing antibiotic misuse, iatrogenic infections, and the rates of antibiotic-resistant organisms [47]. In one retrospective observational study, annual costs of antibiotics dropped by 33% with re-implementation of an ASP, mirrored by an overall decrease in antibiotic consumption of about 10%, over the course of the intervention study period [30]. Of note is that at 1 year post-ASP re-implementation, antibiotic consumption actually increased (by 5.4%); however, because antibiotic usage had changed to more appropriate and cost-effective therapies, cost expenditures associated with antibiotics were still reduced by 13% for that year relative to pre-ASP re-implementation. Aside from economic evaluations centered on consumption rates, there is the potential to further evaluate economic benefits associated with stewardship when looking at other outcomes, including hospital LOS [22], as well as indirect costs such as morbidity and mortality, societal, and operational costs [46]. Currently, these detailed analyses are lacking. In conjunction with more standardized clinical metrics, these assessments are needed to better delineate the full cost effectiveness of ASPs.
Evidence Summary
The evidence for inpatient ASP effectiveness is promising but mixed. Much of the evidence is low-level, based on observational studies that are retrospective in nature, and systematic reviews and meta-analyses are based on these types of studies. Studies have been conducted over a range of years, and the duration of intervention periods often vary widely between studies; it is difficult to capture and account for all of the infection, prescribing, and drug availability patterns (as well as the intervention differences or new drug approvals) throughout these time periods. To complicate the matter, both the quality of data as well as the quality of the ASPs are highly variable.
As such, the findings across pooled studies for ASPs are hard to amalgamate and draw concrete conclusions from. This difficulty is due to the inherent heterogeneity when comparing smaller individual studies in systematic reviews and meta-analyses. Currently, there are numerous ways to implement an ASP, but there is not a standardized system of specific interventions or metrics. Until we can directly compare similar ASPs and interventions among various institutions, it will be challenging to generalize positive benefits from systematic reviews and meta-analyses. Currently, the CDC is involved in a new initiative in which data from various hospitals are compiled to create a surveillance database [48]. Although this is a step in the right direction for standardized metrics for stewardship, for the current review the lack of standard metrics leads to conflicting results of heterogenic studies, making it difficult to show clear benefits in clinical outcomes.
Despite the vast array of ASPs, their differences, and a range of clinical measures—many with conflicting evidence—there is a noticeable trend toward a more prudent use of antimicrobials. Based on the review of available evidence, inpatient ASPs improve patient care and preserve an important health care resource—antibiotics. As has been presented, this is demonstrated by the alterations in consumption of these agents, has ramifications for secondary outcomes such as reduced instances of C. difficile infections, resistance, and adverse effects, and overall translates into better patient care and reduced costs. But while we can conclude that the direct interventions of stewardship in reducing and restricting antibiotic use have been effective, we cannot clearly state the overall magnitude of benefit, the effectiveness of various ASP structures and components on clinical outcomes (such as LOS, mortality, etc.), and the cost savings due to the heterogeneity of the available evidence.
Future Directions
Moving forward, the future of ASPs encompasses several potential developments. First and foremost, as technological advancements continue to develop, there is a need to integrate and utilize developments in information technology (IT). Baysari et al conducted a review on the value of utilizing IT interventions, focusing mainly on decision support (stand-alone or as a component of other hospital procedures), approval, and surveillance systems [49]. There was benefit associated with these IT interventions in terms of the improvement in the appropriate use of antimicrobials (RR 1.49, 95% CI, 1.07–2.08, P < 0.05; I2 = 93%), but there was no demonstrated benefit in terms of patient mortality or hospital LOS. Aside from this study, broad evidence is still lacking to support the use of IT systems in ASPs because meaningful comparisons amongst the interventions have not been made due to widespread variability in study design and outcome measures. However, it is generally agreed that ASPs must integrate with IT systems as the widespread use of technology within the healthcare field continues to grow. Evidence needs to be provided in the form of higher quality studies centered on similar outcomes to show appropriate approaches for ASPs to leverage IT systems. At a minimum, the integration of IT into ASPs should not hinder clinical outcomes. An important consideration is the variation in practice settings where antibiotic stewardship is to be implemented; eg, a small community hospital will be less equipped to incorporate and support technological tools compared to a large tertiary teaching hospital. Therefore, any antibiotic stewardship IT intervention must be customized to meet local needs, prescriber behaviors, minimize barriers to implementation, and utilize available resources.
Another area of focus for future ASPs is the use of rapid diagnostics. Currently, when patients present with signs and symptoms of an infection, an empiric antimicrobial regimen is started that is then de-escalated as necessary; rapid testing will help to initiate appropriate therapy more quickly and increase antimicrobial effectiveness. Rapid tests range from rapid polymerase chain reaction (PCR)-based screening [50], to Verigene gram-positive blood culture (BC-GP) tests [51], next-generation sequencing methods, and matrix assisted laser desorption/ionization time-of-flight mass spectrometry (MALDI-TOF MS) [52]. Rapid diagnostic tools should be viewed as aides to assist ASPs in decreasing antibiotic consumption and improving patient outcomes; these various tools have been shown to improve clinical outcomes when integrated into ASPs, but offer little value addressing the goals of ASPs when used outside of stewardship programs and their sensitive timeframes [53].
In terms of future ASP expansion, stewardship implementation can become more unified and broad in scope. ASPs should expand to include antifungal interventions, an area which is showing progress [36]. ASPs can also be implemented in new areas throughout the hospital (eg, pediatrics and emergency room), as well as areas outside of the hospital setting, including long-term care facilities, dialysis centers, and other institutions [54–56]. A prospective randomized control study was conducted in 30 nursing homes to evaluate the use of a novel resident antimicrobial management plan (RAMP) for improved use of antimicrobials [57]. This study found that the RAMP had no associated adverse effects and suggests that ASP is an important tool in nursing homes. In addition, the general outpatient and pediatric settings show promise for ASPs [56,58,59], but more research is needed to support expansion and to identify how ASP interventions should be applied in these various practice settings. The antimicrobial stewardship interventions that will be utilized will need to be carefully delineated to consider the scale, underlying need, and potential challenges in those settings.
While the future of antibiotic stewardship is unclear, there is certainty that it will continue to develop in both scope and depth to encompass new areas of focus, new settings to improve outcomes, and employ new tools to refine approaches. An important first step for the continued development of ASPs is alignment and standardization, since without alignment it will continue to be difficult to compare outcomes. This issue is currently being addressed by a number of different organizations. With current support from the Joint Commission, the CDC, as well as the President’s Council of Advisors on Science and Technology (PCAST) [8], regulatory requirements for ASPs are well underway, and these drivers will appropriately position ASPs for further advancements. By reducing variability amongst ASPs and delineating implementation of ASPs, there can be a clear identification of both economic and clinical benefits associated with specific interventions.
Corresponding author: Luigi Brunetti, PharmD, MPH, Rutgers, The State University of New Jersey, 160 Frelinghuysen Rd., Piscataway, NJ 08854, [email protected].
Financial disclosures: None.
From the Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ.
Abstract
- Objective: To review the evidence evaluating inpatient antimicrobial stewardship programs (ASPs) with a focus on clinical and economic outcomes.
- Methods: Pubmed/MEDLINE and the Cochrane Database of Systematic Reviews were used to identify systematic reviews, meta-analyses, randomized controlled trials, and other relevant literature evaluating the clinical and economic impact of ASP interventions.
- Results: A total of 5 meta-analyses, 3 systematic reviews, and 10 clinical studies (2 randomized controlled, 5 observational, and 3 quasi-experimental studies) were identified for analysis. ASPs were associated with a reduction in antimicrobial consumption and use. However, due to the heterogeneity of outcomes measured among studies, the effectiveness of ASPs varied with the measures used. There are data supporting the cost savings associated with ASPs, but these studies are more sparse. Most of the available evidence supporting ASPs is of low quality, and intervention strategies vary widely among available studies.
- Conclusion: Much of the evidence reviewed supports the assertion that ASPs result in a more judicious use of antimicrobials and lead to better patient care in the inpatient setting. While clinical outcomes vary between programs, there are ubiquitous positive benefits associated with ASPs in terms of antimicrobial consumption, C. difficile infection rates, and resistance, with few adverse effects. To date, economic outcomes have been difficult to uniformly quantify, but there are data supporting the economic benefits of ASPs. As the number of ASPs continues to grow, it is imperative that standardized metrics are considered in order to accurately measure the benefits of these essential programs.
Key words: Antimicrobial stewardship; antimicrobial consumption; resistance.
Antimicrobial resistance is a public health concern that has been escalating over the years and is now identified as a global crisis [1–3]. This is partly due to the widespread use of the same antibiotics that have existed for decades, combined with a lack of sufficient novel antibiotic discovery and development [4]. Bacteria that are resistant to our last-line-of-defense medications have recently emerged, and these resistant organisms may spread to treatment-naive patients [5]. Multidrug-resistant organisms are often found, treated, and likely originate within the hospital practice setting, where antimicrobials can be prescribed by any licensed provider [6]. Upwards of 50% of antibiotics administered are unnecessary and contribute to the problem of increasing resistance [7]. The seriousness of this situation is increasingly apparent; in 2014 the World Health Organization (WHO), President Obama, and Prime Minister Cameron issued statements urging solutions to the resistance crisis [8].
While the urgency of the situation is recognized today, efforts aimed at a more judicious use of antibiotics to curb resistance began as early as the 1960s and led to the first antimicrobial stewardship programs (ASPs) [9–11]. ASPs have since been defined as “coordinated interventions designed to improve and measure the appropriate use of antimicrobial agents by promoting the selection of the optimal antimicrobial drug regimen including dosing, duration of therapy, and route of administration” [1]. The primary objectives of these types of programs are to avoid or reduce adverse events (eg, Clostridium difficile infection) and resistance driven by a shift in minimum inhibitory concentrations (MICs) and to reverse the unnecessary economic burden caused by the inappropriate prescribing of these agents [1].
This article examines the evidence evaluating the reported effectiveness of inpatient ASPs, examining both clinical and economic outcomes. In addition, we touch on ASP history, current status, and future directions in light of current trends. While ASPs are expanding into the outpatient and nursing home settings, we will limit our review here to the inpatient setting.
Historical Background
Modern antibiotics date back to the late 1930s when penicillin and sulfonamides were introduced to the medical market, and resistance to these drug classes was reported just a few years after their introduction. The same bacterial resistance mechanisms that neutralized their efficacy then exist today, and these mechanisms continue to confer resistance among those classes [5].
While “stewardship” was not described as such until the late 1990s [12], institutions have historically been proactive in creating standards around antimicrobial utilization to encourage judicious use of these agents. The earliest form of tracking antibiotic use was in the form of paper charts as “antibiotic logs” [9] and “punch cards” [10] in the 1960s. The idea of a team approach to stewardship dates back to the 1970s, with the example of Hartford Hospital in Hartford, Connecticut, which employed an antimicrobial standards model run by an infectious disease (ID) physician and clinical pharmacists [11]. In 1977, the Infectious Diseases Society of America (IDSA) released a statement that clinical pharmacists may have a substantial impact on patient care, including in ID, contributing to the idea that a team of physicians collaborating with pharmacists presents the best way to combat inappropriate medication use. Pharmacist involvement has since been shown to restrict broad overutilized antimicrobial agents and reduce the rate of C. difficile infection by a significant amount [13].
In 1997 the IDSA and the Society for Healthcare Epidemiology of America (SHEA) published guidelines to assist in the prevention of the growing issue of resistance, mentioning the importance of antimicrobial stewardship [14]. A decade later they released joint guidelines for ASP implementation [15], and the Pediatric Infectious Disease Society (PIDS) joined them in 2012 to publish a joint statement acknowledging and endorsing stewardship [16]. In 2014, the Centers of Disease Control and Prevention (CDC) recommended that every hospital should have an ASP. As of 1 January 2017, the Joint Commission requires an ASP as a standard for accreditation at hospitals, critical access hospitals, and nursing care [17]. Guidelines for implementation of an ASP are currently available through the IDSA and SHEA [1,16].
ASP Interventions
There are 2 main strategies that ASPs have to combat inappropriate antimicrobial use, and each has its own set of systematic interventions. These strategies are referred to as “prospective audit with intervention and feedback” and “prior authorization” [6]. Although most ASPs will incorporate these main strategies, each institution typically creates its own strategies and regulations independently.
Prospective audit with intervention and feedback describes the process of providing recommendations after reviewing utilization and trends of antimicrobial use. This is sometimes referred to as the “back-end” intervention, in which decisions are made after antibiotics have been administered. Interventions that are commonly used under this strategy include discontinuation of antibiotics due to culture data, de-escalation to drugs with narrower spectra, IV to oral conversions, and cessation of surgical prophylaxis [6].
Prior authorization, also referred to as a “front-end” intervention, is the process of approving medications before they are used. Interventions include a restricted formulary for antimicrobials that can be managed through a paging system or a built-in computer restriction program, as well as other guidelines and protocols for dosing and duration of therapy. Restrictions typically focus on broad spectrum antibiotics as well as the more costly drugs on formularies. These solutions reduce the need for manual intervention as technology makes it possible to create automated restriction-based services that prevent inappropriate prescribing [6].
Aside from these main techniques, other strategies are taken to achieve the goal of attaining optimal clinical outcomes while limiting further antimicrobial resistance and adverse effects. Different clinical settings have different needs, and ASPs are customized to each setting’s resources, prescribing habits, and other local specificities [1]. These differences present difficulty with interpreting diverse datasets, but certain themes arise in the literature: commonly assessed clinical outcomes of inpatient ASPs include hospital length of stay (LOS) and readmission, reinfection, mortality, and resistance rates. These outcomes are putatively driven by the more prudent use of antimicrobials, particularly by decreased rates of antimicrobial consumption.
ASP Team Members
While ASPs may differ between institutions, the staff members involved are typically the same, and leadership is always an important aspect of a program. The CDC recommends that ASP leadership consist of a program leader (an ID physician) and a pharmacy leader, who co-lead the team [18]. In addition, the Joint Commission recommends that the multidisciplinary team should include an infection preventionist (ie, infection control and hospital epidemiologist) and practitioner [17]; these specialists have a role in prevention, awareness, and policy [19]. The integration of infection control with stewardship yields the best results [15], as infection control aims to prevent antibiotic use altogether, while stewardship increases the quality of antibiotic regimens that are being prescribed [20].
It is also beneficial to incorporate a microbiologist as an integral part of the team, responsible for performing and interpreting laboratory data (ie, cultures). Nurses should be integrated into ASPs due to the overlap of their routine activities with ASP interventions [21]; other clinicians (regardless of their infectious disease clinical background), quality control, information technology, and environmental services should all collaborate in the hospital-wide systems related to the program where appropriate [18].
Evidence Review
Results
Antimicrobial Usage
The most widely studied aspect of ASPs in the current review was the effect of ASP interventions on antimicrobial consumption and use. Three systematic reviews [22–24] showed improved antibiotic prescribing practices and reduced consumption rates overall, as did several studies inside and outside the intensive care unit (ICU) [25–31].One study found an insignificant declining usage trend [32]. An important underlying facet of this observation is that even as total antibiotic consumption decreases, certain antibiotic and antibiotic class consumption may increase. This is evident in several studies, which showed that as aminoglycoside, carbapenem, and β-lactam-β-lactamase inhibitor use increased, clindamycin (1 case), glycopeptide, fluoroquinolone, and macrolide use decreased [27,28,30]. A potential confounding factor relating to decreased glycopeptide use in Bevilacqua et al [30] was that there was an epidemic of glycopeptide-resistant enterococci during the study period, potentially causing prescribers to naturally avoid it. In any case, since the aim of ASPs is to encourage a more judicious usage of antimicrobials, the observed decreases in consumption of those restricted medications is intuitive. These observations about antimicrobial consumption related to ASPs are relevant because they putatively drive improvements in clinical outcomes, especially those related to reduced adverse events associated with these agents, such as the risk of C. difficile infection with certain drugs (eg, fluoroquinolones, clindamycin, and broad-spectrum antibiotics) and prolonged antibiotic usage [33–35]. There is evidence that these benefits are not limited to antibiotics but extend to antifungal agents and possibly antivirals [22,27,36].
Utilization, Mortality, and Infection Rates
ASPs typically intend to improve patient-focused clinical parameters such as hospital LOS, hospital readmissions, mortality, and incidence of infections acquired secondary to antibiotic usage during a hospital stay, especially C. difficile infection. Most of the reviewed evidence indicates that there has been no significant LOS benefit due to stewardship interventions [24–26,32,37], and one meta-analysis noted that when overall hospital LOS was significantly reduced, ICU-specific LOS was not [22]. Generally, there was also not a significant change in hospital readmission rates [24,26,32]. However, 2 retrospective observational studies found mixed results for both LOS and readmission rates relative to ASP interventions; while both noted a significantly reduced LOS, one study [38] showed an all-cause readmission benefit in a fairly healthy patient population (but no benefit for readmissions due to the specific infections of interest), and the another [29] showed a benefit for readmissions due to infections but an increased rate of readmissions in the intervention group overall. In this latter study, hospitalizations within the previous 3 months were significantly higher at baseline for the intervention group (55% vs. 46%, P = 0.042), suggesting sicker patients and possibly providing an explanation for this unique observation. Even so, a meta-analysis of 5 studies found a significantly elevated risk of readmission associated with ASP interventions (RR 1.26, 95% CI 1.02–1.57; P = 0.03); the authors noted that non–infection-related readmissions accounted for 61% of readmissions, but this was not significantly different between intervention and non-intervention arms [37].
With regard to mortality, most studies found no significant reductions related to stewardship interventions [22,24,26,29,32]. In a prospective randomized controlled trial, all reported deaths (7/160, 4.4%) were in the ASP intervention arm, but these were attributed to the severities of infection or an underlying, chronic disease [25]. One meta-analysis, however, found that there were significant mortality reductions related to stewardship guidelines for empirical antibiotic treatment (OR 0.65, 95% CI 0.54–0.80, P < 0.001; I2 = 65%) and to de-escalation of therapy based on culture results (RR 0.44, 95% CI 0.30–0.66, P < 0.001; I2 = 59%), based on 40 and 25 studies, respectively [39]; but both results exhibited substantial heterogeneity (defined as I2 = 50%–90% [40]) among the relevant studies. Another meta-analysis found that there was no significant change in mortality related to stewardship interventions intending to improve antibiotic appropriateness (RR 0.92, 95% CI 0.69–1.2, P = 0.56; I2 = 72%) or intending to reduce excessive prescribing (RR 0.92, 95% CI 0.81–1.06, P = 0.25; I2 = 0%), but that there was a significant mortality benefit associated with interventions aimed at increasing guideline compliance for pneumonia diagnoses (RR 0.89, 95% CI 0.82–0.97, P = 0.005; I2 = 0%) [37]. In the case of Schuts et al [39], search criteria specifically sought studies that assessed clinical outcomes (eg, mortality), whereas the search of Davey et al [37] focused on studies whose aim was to improve antibiotic prescribing, with a main comparison being between restrictive and persuasive interventions; while the difference may seem subtle, the body of data compiled from these searches may characterize the ASP effect of mortality differently. No significant evidence was found to suggest that reduced antimicrobial consumption increases mortality.
Improving the use of antimicrobial agents should limit collateral damage associated with their use (eg, damage to normal flora and increased resistance), and ideally infections should be better managed. As previously mentioned, one of the concerns with antibiotic usage (particularly fluoroquinolones, macrolides, and broad-spectrum agents) is that collateral damage could lead to increased rates of C. difficile infection. One meta-analysis showed no significant reduction in the rate of C. difficile infection (as well as overall infection rate) relative to ASPs [22]; however, this finding was based on only 3 of the 26 studies analyzed, and only 1 of those 3 studies utilized restrictions for flouroquinolones and cephalosporins. An interrupted time series (ITS) study similarly found no significant reduction in C. difficile infection rate [32]; however, this study was conducted in a hospital with low baseline antibiotic prescribing (it was ranked second-to-last in terms of antibiotic usage among its peer institutions), inherently limiting the risk of C. difficile infection among patients in the pre-ASP setting. In contrast to these findings, a meta-analysis specifically designed to assess the incidence of C. difficile infection relative to stewardship programs found a significantly reduced risk of infection based on 16 studies (RR 0.48, 95% CI 0.38–0.62, P < 0.001; I2 = 76%) [41], and the systematic review conducted by Filice et al [24] found a significant benefit with regard to the C. difficile infection rate in 4 of 6 studies. These results are consistent with those presented as evidence for the impact of stewardship on C. difficile infection by the CDC [42]. Aside from C. difficile infection, one retrospective observational study found that the 14-day reinfection rate (ie, reinfection with the same infection at the same anatomical location) was significantly reduced following stewardship intervention (0% vs. 10%, P = 0.009) [29]. This finding, combined with the C. difficile infection examples, provide evidence for better infection management of ASPs.
While the general trend seems to suggest mixed or no significant benefit for several clinical outcomes, it is important to note that variation in outcomes could be due to differences in the types of ASP interventions and intervention study periods across differing programs. Davey et al [37] found variation in prescribing outcomes based on whether restrictive (ie, restrict prescriber freedom with antimicrobials) or persuasive (ie, suggest changes to prescriber) interventions were used, and on the timeframe in which they were used. At one month into an ASP, restrictive interventions resulted in better prescribing practices relative to persuasive interventions based on 27 studies (effect size 32.0%, 95% CI 2.5%–61.4%), but by 6 months the 2 were not statistically different (effect size 10.1%, 95% CI –47.5% to 66.0%). At 12 and 24 months, persuasive interventions demonstrated greater effects on prescribing outcomes, but these were not significant. These findings provide evidence that different study timeframes can impact ASP practices differently (and these already vary widely in the literature). Considering the variety of ASP interventions employed across the different studies, these factors almost certainly impact the reported antimicrobial consumption rates and outcomes to different degrees as a consequence. A high degree of heterogeneity among an analyzed dataset could itself be the reason for net non-significance within single systematic reviews and meta-analyses.
Resistance
Another goal of ASPs is the prevention of antimicrobial resistance, an area where the evidence generally suggests benefit associated with ASP interventions. Resistance rates to common troublesome organisms, such as methicillin-resistant S. aureus (MRSA), imipenem-resistant P. aeruginosa, and extended-spectrum β-lactamase (ESBL)–producing Klebsiella spp were significantly reduced in a meta-analysis; ESBL-producing E. coli infections were not, however [22]. An ITS study found significantly reduced MRSA resistance, as well as reduced Pseudomonal resistance to imipenem-cilastin and levofloxacin (all P < 0.001), but no significant changes with respect to piperacillin/tazobactam, cefepime, or amikacin resistance [32]. This study also noted increased E. coli resistance to levofloxacin and ceftriaxone (both P < 0.001). No significant changes in resistance were noted for vancomycin-resistant enterococci. It may be a reasonable expectation that decreasing inappropriate antimicrobial use may decrease long-term antimicrobial resistance; but as most studies only span a few years, only the minute changes in resistance are understood [23]. Longer duration studies are needed to better understand resistance outcomes.
Of note is a phenomenon known as the “squeezing the balloon” effect. This can be associated with ASPs, potentially resulting in paradoxically increased resistance [43]. That is, when usage restrictions are placed on certain antibiotics, the use of other non-restricted antibiotics may increase, possibly leading to increased resistance of those non-restricted antibiotics [22] (“constraining one end [of a balloon] causes the other end to bulge … limiting the use of one class of compounds may be counteracted by corresponding changes in prescribing and drug resistance that are even more ominous” [43]). Karanika et al [22] took this phenomonen into consideration, and assessed restricted and non-restricted antimicrobial consumption separately. They found a reduction in consumption for both restricted and non-restricted antibiotics, which included “high potential resistance” antibiotics, specifically carbapenems and glycopeptides. In the study conducted by Cairns et al [28], a similar effect was observed; while the use of other classes of antibiotics decreased (eg, cephalosporins and aminoglycosides), the use of β–lactam–β–lactamase inhibitor combinations actually increased by 48% (change in use: +48.2% [95% CI 21.8%–47.9%]). Hohn et al [26] noted an increased usage rate of carbapenems, even though several other classes of antibiotics had reduced usage. Unfortunately, neither study reported resistance rates, so the impact of these findings is unknown. Finally, Jenkins et al [32] assessed trends in antimicrobial use as changes in rates of consumption. Among the various antibiotics assessed in this study, the rate of flouroquinolone use decreased both before and after the intervention period, although the rate of decreased usage slowed post-ASP (the change in rate post-ASP was +2.2% [95% CI 1.4%–3.1%], P < 0.001). They observed a small (but significant) increase in resistance of E. coli to levofloxacin pre- vs. post-intervention (11.0% vs. 13.9%, P < 0.001); in contrast, a significant decrease in resistance of P. aeruginosa was observed (30.5% vs. 21.4%, P < 0.001). While these examples help illustrate the concept of changes in antibiotic usage patterns associated with an ASP, at best they approximate the “squeezing the balloon” effect since these studies present data for antibiotics that were either restricted or for which restriction was not clearly specified. The “squeezing the balloon” effect is most relevant for the unintended, potentially increased usage of non-restricted drugs secondary to ASP restrictions. Higher resistance rates among certain drug classes observed in the context of this effect would constitute a drawback to an ASP program.
Adverse Effects
Reduced toxicities and adverse effects are expected with reduced usage of antimicrobials. The systematic review conducted by Filice et al [24] examined the incidence of adverse effects related to antibiotic usage, and their findings suggest, at the least, that stewardship programs generally do not cause harm, as only 2 of the studies they examined reported adverse events. Following stewardship interventions, 5.5% of the patients deteriorated; and of those, the large majority (75%) deteriorated due to progression of oncological malignancies. To further illustrate the effect of stewardship interventions on toxicities and side effects of antimicrobials, Schuts et al demonstrated that the risk of nephrotoxicity while on antimicrobial therapy was reduced based on 14 studies of moderate heterogeneity as a result of an ASP (OR 0.46, 95% CI 0.28–0.77, P = 0.003; I2 = 34%) [39,44]. It is intuitive that reduced drug exposure results in reduced adverse effects, as such these results are expected.
Economic Outcomes
Although the focus of ASPs is often to improve clinical outcomes, economic outcomes are an important component of ASPs; these programs bring associated economic value that should be highlighted and further detailed [22,45,46]. Since clinical outcomes are often the main objective of ASPs, most available studies have been clinical effect studies (rather than economic analyses), in which economic assessments are often a secondary consideration, if included.
As a result, cost evaluations are conducted on direct cost reductions whereas indirect cost reductions are often not critically evaluated. ASPs reduce hospital expenditures by limiting hospital-acquired infections and the associated medical costs where they are effective at decreasing consumption of antimicrobials [22,45], and by reducing antibiotic misuse, iatrogenic infections, and the rates of antibiotic-resistant organisms [47]. In one retrospective observational study, annual costs of antibiotics dropped by 33% with re-implementation of an ASP, mirrored by an overall decrease in antibiotic consumption of about 10%, over the course of the intervention study period [30]. Of note is that at 1 year post-ASP re-implementation, antibiotic consumption actually increased (by 5.4%); however, because antibiotic usage had changed to more appropriate and cost-effective therapies, cost expenditures associated with antibiotics were still reduced by 13% for that year relative to pre-ASP re-implementation. Aside from economic evaluations centered on consumption rates, there is the potential to further evaluate economic benefits associated with stewardship when looking at other outcomes, including hospital LOS [22], as well as indirect costs such as morbidity and mortality, societal, and operational costs [46]. Currently, these detailed analyses are lacking. In conjunction with more standardized clinical metrics, these assessments are needed to better delineate the full cost effectiveness of ASPs.
Evidence Summary
The evidence for inpatient ASP effectiveness is promising but mixed. Much of the evidence is low-level, based on observational studies that are retrospective in nature, and systematic reviews and meta-analyses are based on these types of studies. Studies have been conducted over a range of years, and the duration of intervention periods often vary widely between studies; it is difficult to capture and account for all of the infection, prescribing, and drug availability patterns (as well as the intervention differences or new drug approvals) throughout these time periods. To complicate the matter, both the quality of data as well as the quality of the ASPs are highly variable.
As such, the findings across pooled studies for ASPs are hard to amalgamate and draw concrete conclusions from. This difficulty is due to the inherent heterogeneity when comparing smaller individual studies in systematic reviews and meta-analyses. Currently, there are numerous ways to implement an ASP, but there is not a standardized system of specific interventions or metrics. Until we can directly compare similar ASPs and interventions among various institutions, it will be challenging to generalize positive benefits from systematic reviews and meta-analyses. Currently, the CDC is involved in a new initiative in which data from various hospitals are compiled to create a surveillance database [48]. Although this is a step in the right direction for standardized metrics for stewardship, for the current review the lack of standard metrics leads to conflicting results of heterogenic studies, making it difficult to show clear benefits in clinical outcomes.
Despite the vast array of ASPs, their differences, and a range of clinical measures—many with conflicting evidence—there is a noticeable trend toward a more prudent use of antimicrobials. Based on the review of available evidence, inpatient ASPs improve patient care and preserve an important health care resource—antibiotics. As has been presented, this is demonstrated by the alterations in consumption of these agents, has ramifications for secondary outcomes such as reduced instances of C. difficile infections, resistance, and adverse effects, and overall translates into better patient care and reduced costs. But while we can conclude that the direct interventions of stewardship in reducing and restricting antibiotic use have been effective, we cannot clearly state the overall magnitude of benefit, the effectiveness of various ASP structures and components on clinical outcomes (such as LOS, mortality, etc.), and the cost savings due to the heterogeneity of the available evidence.
Future Directions
Moving forward, the future of ASPs encompasses several potential developments. First and foremost, as technological advancements continue to develop, there is a need to integrate and utilize developments in information technology (IT). Baysari et al conducted a review on the value of utilizing IT interventions, focusing mainly on decision support (stand-alone or as a component of other hospital procedures), approval, and surveillance systems [49]. There was benefit associated with these IT interventions in terms of the improvement in the appropriate use of antimicrobials (RR 1.49, 95% CI, 1.07–2.08, P < 0.05; I2 = 93%), but there was no demonstrated benefit in terms of patient mortality or hospital LOS. Aside from this study, broad evidence is still lacking to support the use of IT systems in ASPs because meaningful comparisons amongst the interventions have not been made due to widespread variability in study design and outcome measures. However, it is generally agreed that ASPs must integrate with IT systems as the widespread use of technology within the healthcare field continues to grow. Evidence needs to be provided in the form of higher quality studies centered on similar outcomes to show appropriate approaches for ASPs to leverage IT systems. At a minimum, the integration of IT into ASPs should not hinder clinical outcomes. An important consideration is the variation in practice settings where antibiotic stewardship is to be implemented; eg, a small community hospital will be less equipped to incorporate and support technological tools compared to a large tertiary teaching hospital. Therefore, any antibiotic stewardship IT intervention must be customized to meet local needs, prescriber behaviors, minimize barriers to implementation, and utilize available resources.
Another area of focus for future ASPs is the use of rapid diagnostics. Currently, when patients present with signs and symptoms of an infection, an empiric antimicrobial regimen is started that is then de-escalated as necessary; rapid testing will help to initiate appropriate therapy more quickly and increase antimicrobial effectiveness. Rapid tests range from rapid polymerase chain reaction (PCR)-based screening [50], to Verigene gram-positive blood culture (BC-GP) tests [51], next-generation sequencing methods, and matrix assisted laser desorption/ionization time-of-flight mass spectrometry (MALDI-TOF MS) [52]. Rapid diagnostic tools should be viewed as aides to assist ASPs in decreasing antibiotic consumption and improving patient outcomes; these various tools have been shown to improve clinical outcomes when integrated into ASPs, but offer little value addressing the goals of ASPs when used outside of stewardship programs and their sensitive timeframes [53].
In terms of future ASP expansion, stewardship implementation can become more unified and broad in scope. ASPs should expand to include antifungal interventions, an area which is showing progress [36]. ASPs can also be implemented in new areas throughout the hospital (eg, pediatrics and emergency room), as well as areas outside of the hospital setting, including long-term care facilities, dialysis centers, and other institutions [54–56]. A prospective randomized control study was conducted in 30 nursing homes to evaluate the use of a novel resident antimicrobial management plan (RAMP) for improved use of antimicrobials [57]. This study found that the RAMP had no associated adverse effects and suggests that ASP is an important tool in nursing homes. In addition, the general outpatient and pediatric settings show promise for ASPs [56,58,59], but more research is needed to support expansion and to identify how ASP interventions should be applied in these various practice settings. The antimicrobial stewardship interventions that will be utilized will need to be carefully delineated to consider the scale, underlying need, and potential challenges in those settings.
While the future of antibiotic stewardship is unclear, there is certainty that it will continue to develop in both scope and depth to encompass new areas of focus, new settings to improve outcomes, and employ new tools to refine approaches. An important first step for the continued development of ASPs is alignment and standardization, since without alignment it will continue to be difficult to compare outcomes. This issue is currently being addressed by a number of different organizations. With current support from the Joint Commission, the CDC, as well as the President’s Council of Advisors on Science and Technology (PCAST) [8], regulatory requirements for ASPs are well underway, and these drivers will appropriately position ASPs for further advancements. By reducing variability amongst ASPs and delineating implementation of ASPs, there can be a clear identification of both economic and clinical benefits associated with specific interventions.
Corresponding author: Luigi Brunetti, PharmD, MPH, Rutgers, The State University of New Jersey, 160 Frelinghuysen Rd., Piscataway, NJ 08854, [email protected].
Financial disclosures: None.
1. Barlam TF, Cosgrove SE, Abbo AM, et al. Implementing an antimicrobial stewardship program: guidelines by the Infectious Diseases Society of America and the Society of Healthcare Epidemiology of America. Clin Infect Dis 2016;62:e51–77.
2. Hughes D. Selection and evolution of resistance to antimicrobial drugs. IUBMB Life 2014;66:521–9.
3. World Health Organzation. The evolving threat of antimicrobial resistance – options for action. Geneva: WHO Press; 2012.
4. Gould IM, Bal AM. New antibiotic agents in the pipeline and how they can help overcome microbial resistance. Virulence 2013;4:185–91.
5. Davies J, Davies D. Origins and evolution of antibiotic resistance. Microbiol Mol Biol Rev 2010;74:417–33.
6. Owens RC Jr. Antimicrobial stewardship: concepts and strategies in the 21st century. Diagn Microbiol Infect Dis 2008;61:110–28.
7. Antibiotic resistance threats in the United States, 2013 [Internet]. Centers for Disease Control and Prevention. Available at www.cdc.gov/drugresistance/pdf/ar-threats-2013-508.pdf.
8. Nathan C, Cars O. Antibiotic resistance – problems, progress, prospects. N Engl J Med 2014;371:1761–3.
9. McGoldrick, M. Antimicrobial stewardship. Home Healthc Nurse 2014;32:559–60.
10. Ruedy J. A method of determining patterns of use of antibacterial drugs. Can Med Assoc J 1966;95:807–12.
11. Briceland LL, Nightingdale CH, Quintiliani R, et al. Antibiotic streamlining from combination therapy to monotherapy utilizing an interdisciplinary approach. Arch Inter Med 1988;148:2019–22.
12. McGowan JE Jr, Gerding DN. Does antibiotic restriction prevent resistance? New Horiz 1996;4: 370–6.
13. Cappelletty D, Jacobs D. Evaluating the impact of a pharmacist’s absence from an antimicrobial stewardship team. Am J Health Syst Pharm 2013;70:1065–69.
14. Shales DM, Gerding DN, John JF Jr, et al. Society for Healthcare Epidemiology of America and Infectious Diseases Society of America Joint Committee on the prevention of antimicrobial resistance: guidelines for the prevention of antimicrobial resistance in hospitals. Infect Control Hosp Epidemiol 1997;18:275–91.
15. Dellit TH, Owens RC, McGowan JE, et al. Infectious Diseases Society of America and the Society for Healthcare Epidemiology of America guidelines for developing an institutional program to enhance antimicrobial stewardship. Clin Infect Dis 2007;44:159–77.
16. Policy statement on antimicrobial stewardship by the Society for Healthcare Epidemiology of America (SHEA), the Infectious Diseases Society of America (IDSA), and the Pediatric Infectious Diseases Society (PIDS). Infect Ctrl Hosp Epidemiol 2012;33:322–7.
17. The Joint Commission. Approved: New antimicrobial stewardship standard. Joint Commission Perspectives 2016;36:1–8.
18. Pollack LA, Srinivasan A. Core elements of hospital antibiotic stewardship programs from the Centers for Disease Control and Prevention. Clin Infect Dis 2014;59(Suppl 3):S97–100.
19. Moody J. Infection preventionists have a role in accelerating progress toward preventing the emergence and cross-transmission of MDROs. Prevention Strategist 2012 Summer:52–6.
20. Spellberg B, Bartlett JG, Gilbert DN. The future of antibiotics and resistance. N Engl J Med 2013;368:299–302.
21. Olans RN, Olans RD, Demaria A. The critical role of the staff nurse in antimicrobial stewardship--unrecognized, but already there. Clin Infect Dis 2016;62:84–9.
22. Karanika S, Paudel S, Grigoras C, et al. Systematic review and meta-analysis of clinical and economic outcomes from the implementation of hospital-based antimicrobial stewardship programs. Antimicrob Agents Chemother 2016;60:4840–52.
23. Wagner B, Filice GA, Drekonja D, et al. Antimicrobial stewardship programs in inpatient hospital settings: a systematic review. Infect Control Hosp Epidemiol 2014;35:1209–28.
24. Filice G, Drekonja D, Greer N, et al. Antimicrobial stewardship programs in inpatient settings: a systematic review. VA-ESP Project #09-009; 2013.
25. Cairns KA, Doyle JS, Trevillyan JM, et al. The impact of a multidisciplinary antimicrobial stewardship team on the timeliness of antimicrobial therapy in patients with positive blood cultures: a randomized controlled trial. J Antimicrob Chemother 2016;71:3276–83.
26. Hohn A, Heising B, Hertel S, et al. Antibiotic consumption after implementation of a procalcitonin-guided antimicrobial stewardship programme in surgical patients admitted to an intensive care unit: a retrospective before-and-after analysis. Infection 2015;43:405–12.
27. Singh S, Zhang YZ, Chalkley S, et al. A three-point time series study of antibiotic usage on an intensive care unit, following an antibiotic stewardship programme, after an outbreak of multi-resistant Acinetobacter baumannii. Eur J Clin Microbiol Infect Dis 2015;34:1893–900.
28. Cairns KA, Jenney AW, Abbott IJ, et al. Prescribing trends before and after implementation of an antimicrobial stewardship program. Med J Aust 2013;198:262–6.
29. Liew YX, Lee W, Loh JC, et al. Impact of an antimicrobial stewardship programme on patient safety in Singapore General Hospital. Int J Antimicrob Agents 2012;40:55–60.
30. Bevilacqua S, Demoré B, Boschetti E, et al. 15 years of antibiotic stewardship policy in the Nancy Teaching Hospital. Med Mal Infect 2011;41:532–9.
31. Danaher PJ, Milazzo NA, Kerr KJ, et al. The antibiotic support team--a successful educational approach to antibiotic stewardship. Mil Med 2009;174:201–5.
32. Jenkins TC, Knepper BC, Shihadeh K, et al. Long-term outcomes of an antimicrobial stewardship program implemented in a hospital with low baseline antibiotic use. Infect Control Hosp Epidemiol 2015;36:664–72.
33. Brown KA, Khanafer N, Daneman N, Fisman DN. Meta-analysis of antibiotics and the risk of community-associated Clostridium difficile infection. Antimicrob Agents Chemother 2013;57:2326–32.
34. Deshpande A, Pasupuleti V, Thota P, et al. Community-associated Clostridium difficile infection and antibiotics: a meta-analysis. J Antimicrob Chemother 2013;68:1951–61.
35. Slimings C, Riley TV. Antibiotics and hospital-acquired Clostridium difficile infection: update of systematic review and meta-analysis. J Antimicrob Chemother 2014;69:881–91.
36. Antworth A, Collins CD, Kunapuli A, et al. Impact of an antimicrobial stewardship program comprehensive care bundle on management of candidemia. Pharmacotherapy 2013;33:137–43.
37. Davey P, Brown E, Charani E, et al. Interventions to improve antibiotic prescribing practices for hospital inpatients. Cochrane Database Syst Rev 2013;4:CD003543.
38. Pasquale TR, Trienski TL, Olexia DE, et al. Impact of an antimicrobial stewardship program on patients with acute bacterial skin and skin structure infections. Am J Health Syst Pharm 2014;71:1136–9.
39. Schuts EC, Hulscher ME, Mouton JW, et al. Current evidence on hospital antimicrobial stewardship objectives: a systematic review and meta-analysis. Lancet Infect Dis 2016;16:847–56.
40. Higgins JPT, Green S, editors. Identifying and measuring heterogeneity. Cochrane Handbook for Systematic Reviews of Interventions, version 5.1.0. [Internet]. The Cochrane Collaboration, March 2011. Available at http://handbook.cochrane.org/chapter_9/9_5_2_identifying_and_measuring_heterogeneity.htm.
41. Feazel LM, Malhotra A, Perencevich EN, et al. Effect of antibiotic stewardship programmes on Clostridium difficile incidence: a systematic review and meta-analysis. J Antimicrob Chemother 2014;69:1748–54.
42. Impact of antibiotic stewardship programs on Clostridium difficile (C. diff) infections [Internet]. Centers for Disease Control and Prevention. [Updated 2016 May 13; cited 2016 Oct 11]. Available at www.cdc.gov/getsmart/healthcare/evidence/asp-int-cdiff.html.
43. Burke JP. Antibiotic resistance – squeezing the balloon? JAMA 1998;280:1270–1.
44. This nephrotoxicity result is corrected from the originally published result; communicated by Jan M Prins on behalf of the authors for reference [39]. Prins, JM (Department of Internal Medicine, Division of Infectious Diseases, Academic Medical Centre, Amsterdam, Netherlands). Email communication with Joseph Eckart (Pharmacy Practice & Administration, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ). 2016 Oct 9.
45. Coulter S, Merollini K, Roberts JA, et al. The need for cost-effectiveness analyses of antimicrobial stewardship programmes: a structured review. Int J Antimicrob Agents 2015;46:140–9.
46. Dik J, Vemer P, Friedrich A, et al. Financial evaluations of antibiotic stewardship programs—a systematic review. Frontiers Microbiol 2015;6:317.
47. Campbell KA, Stein S, Looze C, Bosco JA. Antibiotic stewardship in orthopaedic surgery: principles and practice. J Am Acad Orthop Surg 2014;22:772–81.
48. Surveillance for antimicrobial use and antimicrobial resistance options, 2015 [Internet]. Centers for Disease Control and Prevention. [Updated 2016 May 3; cited 2016 Nov 22]. Available at www.cdc.gov/nhsn/acute-care-hospital/aur/index.html.
49. Baysari MT, Lehnbom EC, Li L, Hargreaves A, et al. The effectiveness of information technology to improve antimicrobial prescribing in hospitals: a systematic review and meta-analysis. Int J Med Inform. 2016;92:15-34.
50. Bauer KA, West JE, Balada-llasat JM, et al. An antimicrobial stewardship program’s impact with rapid polymerase chain reaction methicillin-resistant Staphylococcus aureus/S. aureus blood culture test in patients with S. aureus bacteremia. Clin Infect Dis 2010;51:1074–80.
51. Sango A, Mccarter YS, Johnson D, et al. Stewardship approach for optimizing antimicrobial therapy through use of a rapid microarray assay on blood cultures positive for Enterococcus species. J Clin Microbiol 2013;51:4008–11.
52. Perez KK, Olsen RJ, Musick WL, et al. Integrating rapid diagnostics and antimicrobial stewardship improves outcomes in patients with antibiotic-resistant Gram-negative bacteremia. J Infect 2014;69:216–25.
53. Bauer KA, Perez KK, Forrest GN, Goff DA. Review of rapid diagnostic tests used by antimicrobial stewardship programs. Clin Infect Dis 2014;59 Suppl 3:S134–145.
54. Dyar OJ, Pagani L, Pulcini C. Strategies and challenges of antimicrobial stewardship in long-term care facilities. Clin Microbiol Infect 2015;21:10–9.
55. D’Agata EM. Antimicrobial use and stewardship programs among dialysis centers. Semin Dial 2013;26:457–64.
56. Smith MJ, Gerber JS, Hersh AL. Inpatient antimicrobial stewardship in pediatrics: a systematic review. J Pediatric Infect Dis Soc 2015;4:e127–135.
57. Fleet E, Gopal Rao G, Patel B, et al. Impact of implementation of a novel antimicrobial stewardship tool on antibiotic use in nursing homes: a prospective cluster randomized control pilot study. J Antimicrob Chemother 2014;69:2265–73.
58. Drekonja DM, Filice GA, Greer N, et al. Antimicrobial stewardship in outpatient settings: a systematic review. Infect Control Hosp Epidemiol 2015;36:142–52.
59. Drekonja D, Filice G, Greer N, et al. Antimicrobial stewardship programs in outpatient settings: a systematic review. VA-ESP Project #09-009; 2014.
60. Zhang YZ, Singh S. Antibiotic stewardship programmes in intensive care units: why, how, and where are they leading us. World J Crit Care Med 2015;4:13–28. (referenced in online Table)
1. Barlam TF, Cosgrove SE, Abbo AM, et al. Implementing an antimicrobial stewardship program: guidelines by the Infectious Diseases Society of America and the Society of Healthcare Epidemiology of America. Clin Infect Dis 2016;62:e51–77.
2. Hughes D. Selection and evolution of resistance to antimicrobial drugs. IUBMB Life 2014;66:521–9.
3. World Health Organzation. The evolving threat of antimicrobial resistance – options for action. Geneva: WHO Press; 2012.
4. Gould IM, Bal AM. New antibiotic agents in the pipeline and how they can help overcome microbial resistance. Virulence 2013;4:185–91.
5. Davies J, Davies D. Origins and evolution of antibiotic resistance. Microbiol Mol Biol Rev 2010;74:417–33.
6. Owens RC Jr. Antimicrobial stewardship: concepts and strategies in the 21st century. Diagn Microbiol Infect Dis 2008;61:110–28.
7. Antibiotic resistance threats in the United States, 2013 [Internet]. Centers for Disease Control and Prevention. Available at www.cdc.gov/drugresistance/pdf/ar-threats-2013-508.pdf.
8. Nathan C, Cars O. Antibiotic resistance – problems, progress, prospects. N Engl J Med 2014;371:1761–3.
9. McGoldrick, M. Antimicrobial stewardship. Home Healthc Nurse 2014;32:559–60.
10. Ruedy J. A method of determining patterns of use of antibacterial drugs. Can Med Assoc J 1966;95:807–12.
11. Briceland LL, Nightingdale CH, Quintiliani R, et al. Antibiotic streamlining from combination therapy to monotherapy utilizing an interdisciplinary approach. Arch Inter Med 1988;148:2019–22.
12. McGowan JE Jr, Gerding DN. Does antibiotic restriction prevent resistance? New Horiz 1996;4: 370–6.
13. Cappelletty D, Jacobs D. Evaluating the impact of a pharmacist’s absence from an antimicrobial stewardship team. Am J Health Syst Pharm 2013;70:1065–69.
14. Shales DM, Gerding DN, John JF Jr, et al. Society for Healthcare Epidemiology of America and Infectious Diseases Society of America Joint Committee on the prevention of antimicrobial resistance: guidelines for the prevention of antimicrobial resistance in hospitals. Infect Control Hosp Epidemiol 1997;18:275–91.
15. Dellit TH, Owens RC, McGowan JE, et al. Infectious Diseases Society of America and the Society for Healthcare Epidemiology of America guidelines for developing an institutional program to enhance antimicrobial stewardship. Clin Infect Dis 2007;44:159–77.
16. Policy statement on antimicrobial stewardship by the Society for Healthcare Epidemiology of America (SHEA), the Infectious Diseases Society of America (IDSA), and the Pediatric Infectious Diseases Society (PIDS). Infect Ctrl Hosp Epidemiol 2012;33:322–7.
17. The Joint Commission. Approved: New antimicrobial stewardship standard. Joint Commission Perspectives 2016;36:1–8.
18. Pollack LA, Srinivasan A. Core elements of hospital antibiotic stewardship programs from the Centers for Disease Control and Prevention. Clin Infect Dis 2014;59(Suppl 3):S97–100.
19. Moody J. Infection preventionists have a role in accelerating progress toward preventing the emergence and cross-transmission of MDROs. Prevention Strategist 2012 Summer:52–6.
20. Spellberg B, Bartlett JG, Gilbert DN. The future of antibiotics and resistance. N Engl J Med 2013;368:299–302.
21. Olans RN, Olans RD, Demaria A. The critical role of the staff nurse in antimicrobial stewardship--unrecognized, but already there. Clin Infect Dis 2016;62:84–9.
22. Karanika S, Paudel S, Grigoras C, et al. Systematic review and meta-analysis of clinical and economic outcomes from the implementation of hospital-based antimicrobial stewardship programs. Antimicrob Agents Chemother 2016;60:4840–52.
23. Wagner B, Filice GA, Drekonja D, et al. Antimicrobial stewardship programs in inpatient hospital settings: a systematic review. Infect Control Hosp Epidemiol 2014;35:1209–28.
24. Filice G, Drekonja D, Greer N, et al. Antimicrobial stewardship programs in inpatient settings: a systematic review. VA-ESP Project #09-009; 2013.
25. Cairns KA, Doyle JS, Trevillyan JM, et al. The impact of a multidisciplinary antimicrobial stewardship team on the timeliness of antimicrobial therapy in patients with positive blood cultures: a randomized controlled trial. J Antimicrob Chemother 2016;71:3276–83.
26. Hohn A, Heising B, Hertel S, et al. Antibiotic consumption after implementation of a procalcitonin-guided antimicrobial stewardship programme in surgical patients admitted to an intensive care unit: a retrospective before-and-after analysis. Infection 2015;43:405–12.
27. Singh S, Zhang YZ, Chalkley S, et al. A three-point time series study of antibiotic usage on an intensive care unit, following an antibiotic stewardship programme, after an outbreak of multi-resistant Acinetobacter baumannii. Eur J Clin Microbiol Infect Dis 2015;34:1893–900.
28. Cairns KA, Jenney AW, Abbott IJ, et al. Prescribing trends before and after implementation of an antimicrobial stewardship program. Med J Aust 2013;198:262–6.
29. Liew YX, Lee W, Loh JC, et al. Impact of an antimicrobial stewardship programme on patient safety in Singapore General Hospital. Int J Antimicrob Agents 2012;40:55–60.
30. Bevilacqua S, Demoré B, Boschetti E, et al. 15 years of antibiotic stewardship policy in the Nancy Teaching Hospital. Med Mal Infect 2011;41:532–9.
31. Danaher PJ, Milazzo NA, Kerr KJ, et al. The antibiotic support team--a successful educational approach to antibiotic stewardship. Mil Med 2009;174:201–5.
32. Jenkins TC, Knepper BC, Shihadeh K, et al. Long-term outcomes of an antimicrobial stewardship program implemented in a hospital with low baseline antibiotic use. Infect Control Hosp Epidemiol 2015;36:664–72.
33. Brown KA, Khanafer N, Daneman N, Fisman DN. Meta-analysis of antibiotics and the risk of community-associated Clostridium difficile infection. Antimicrob Agents Chemother 2013;57:2326–32.
34. Deshpande A, Pasupuleti V, Thota P, et al. Community-associated Clostridium difficile infection and antibiotics: a meta-analysis. J Antimicrob Chemother 2013;68:1951–61.
35. Slimings C, Riley TV. Antibiotics and hospital-acquired Clostridium difficile infection: update of systematic review and meta-analysis. J Antimicrob Chemother 2014;69:881–91.
36. Antworth A, Collins CD, Kunapuli A, et al. Impact of an antimicrobial stewardship program comprehensive care bundle on management of candidemia. Pharmacotherapy 2013;33:137–43.
37. Davey P, Brown E, Charani E, et al. Interventions to improve antibiotic prescribing practices for hospital inpatients. Cochrane Database Syst Rev 2013;4:CD003543.
38. Pasquale TR, Trienski TL, Olexia DE, et al. Impact of an antimicrobial stewardship program on patients with acute bacterial skin and skin structure infections. Am J Health Syst Pharm 2014;71:1136–9.
39. Schuts EC, Hulscher ME, Mouton JW, et al. Current evidence on hospital antimicrobial stewardship objectives: a systematic review and meta-analysis. Lancet Infect Dis 2016;16:847–56.
40. Higgins JPT, Green S, editors. Identifying and measuring heterogeneity. Cochrane Handbook for Systematic Reviews of Interventions, version 5.1.0. [Internet]. The Cochrane Collaboration, March 2011. Available at http://handbook.cochrane.org/chapter_9/9_5_2_identifying_and_measuring_heterogeneity.htm.
41. Feazel LM, Malhotra A, Perencevich EN, et al. Effect of antibiotic stewardship programmes on Clostridium difficile incidence: a systematic review and meta-analysis. J Antimicrob Chemother 2014;69:1748–54.
42. Impact of antibiotic stewardship programs on Clostridium difficile (C. diff) infections [Internet]. Centers for Disease Control and Prevention. [Updated 2016 May 13; cited 2016 Oct 11]. Available at www.cdc.gov/getsmart/healthcare/evidence/asp-int-cdiff.html.
43. Burke JP. Antibiotic resistance – squeezing the balloon? JAMA 1998;280:1270–1.
44. This nephrotoxicity result is corrected from the originally published result; communicated by Jan M Prins on behalf of the authors for reference [39]. Prins, JM (Department of Internal Medicine, Division of Infectious Diseases, Academic Medical Centre, Amsterdam, Netherlands). Email communication with Joseph Eckart (Pharmacy Practice & Administration, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ). 2016 Oct 9.
45. Coulter S, Merollini K, Roberts JA, et al. The need for cost-effectiveness analyses of antimicrobial stewardship programmes: a structured review. Int J Antimicrob Agents 2015;46:140–9.
46. Dik J, Vemer P, Friedrich A, et al. Financial evaluations of antibiotic stewardship programs—a systematic review. Frontiers Microbiol 2015;6:317.
47. Campbell KA, Stein S, Looze C, Bosco JA. Antibiotic stewardship in orthopaedic surgery: principles and practice. J Am Acad Orthop Surg 2014;22:772–81.
48. Surveillance for antimicrobial use and antimicrobial resistance options, 2015 [Internet]. Centers for Disease Control and Prevention. [Updated 2016 May 3; cited 2016 Nov 22]. Available at www.cdc.gov/nhsn/acute-care-hospital/aur/index.html.
49. Baysari MT, Lehnbom EC, Li L, Hargreaves A, et al. The effectiveness of information technology to improve antimicrobial prescribing in hospitals: a systematic review and meta-analysis. Int J Med Inform. 2016;92:15-34.
50. Bauer KA, West JE, Balada-llasat JM, et al. An antimicrobial stewardship program’s impact with rapid polymerase chain reaction methicillin-resistant Staphylococcus aureus/S. aureus blood culture test in patients with S. aureus bacteremia. Clin Infect Dis 2010;51:1074–80.
51. Sango A, Mccarter YS, Johnson D, et al. Stewardship approach for optimizing antimicrobial therapy through use of a rapid microarray assay on blood cultures positive for Enterococcus species. J Clin Microbiol 2013;51:4008–11.
52. Perez KK, Olsen RJ, Musick WL, et al. Integrating rapid diagnostics and antimicrobial stewardship improves outcomes in patients with antibiotic-resistant Gram-negative bacteremia. J Infect 2014;69:216–25.
53. Bauer KA, Perez KK, Forrest GN, Goff DA. Review of rapid diagnostic tests used by antimicrobial stewardship programs. Clin Infect Dis 2014;59 Suppl 3:S134–145.
54. Dyar OJ, Pagani L, Pulcini C. Strategies and challenges of antimicrobial stewardship in long-term care facilities. Clin Microbiol Infect 2015;21:10–9.
55. D’Agata EM. Antimicrobial use and stewardship programs among dialysis centers. Semin Dial 2013;26:457–64.
56. Smith MJ, Gerber JS, Hersh AL. Inpatient antimicrobial stewardship in pediatrics: a systematic review. J Pediatric Infect Dis Soc 2015;4:e127–135.
57. Fleet E, Gopal Rao G, Patel B, et al. Impact of implementation of a novel antimicrobial stewardship tool on antibiotic use in nursing homes: a prospective cluster randomized control pilot study. J Antimicrob Chemother 2014;69:2265–73.
58. Drekonja DM, Filice GA, Greer N, et al. Antimicrobial stewardship in outpatient settings: a systematic review. Infect Control Hosp Epidemiol 2015;36:142–52.
59. Drekonja D, Filice G, Greer N, et al. Antimicrobial stewardship programs in outpatient settings: a systematic review. VA-ESP Project #09-009; 2014.
60. Zhang YZ, Singh S. Antibiotic stewardship programmes in intensive care units: why, how, and where are they leading us. World J Crit Care Med 2015;4:13–28. (referenced in online Table)
ERAAs for menopause treatment: Welcome the ‘designer estrogens’
Estrogen receptor agonist-antagonists (ERAAs), previously called selective estrogen receptor modulators (SERMs), have extended the options for treating the various conditions that menopausal women suffer from. These drugs act differently on estrogen receptors in different tissues, stimulating receptors in some tissues but inhibiting them in others. This allows selective inhibition or stimulation of estrogen-like action in various target tissues.1
This article highlights the use of ERAAs to treat menopausal vasomotor symptoms (eg, hot flashes, night sweats), genitourinary syndrome of menopause, osteoporosis, breast cancer (and the risk of breast cancer), and other health concerns unique to women at midlife.
SYMPTOMS OF MENOPAUSE: COMMON AND TROUBLESOME
Vasomotor symptoms such as hot flashes and night sweats are common during perimenopause—most women experience them. They are most frequent during the menopause transition but can persist for 10 years or more afterward.2
Genitourinary syndrome of menopause is also common and often worsens with years after menopause.3 It can lead to dyspareunia and vaginal dryness, which may in turn result in lower libido, vaginismus, and hypoactive sexual desire disorder, problems that often arise at the same time as vaginal dryness and atrophy.4
Osteopenia and osteoporosis. A drop in systemic estrogen leads to a decline in bone mineral density, increasing the risk of fractures.5
ESTROGEN-PROGESTIN TREATMENT: THE GOLD STANDARD, BUT NOT IDEAL
The current gold standard for treating moderate to severe hot flashes is estrogen, available in oral, transdermal, and vaginal formulations.6 Estrogen also has antiresorptive effects on bone and is approved for preventing osteoporosis. Systemic estrogen may also be prescribed for genitourinary syndrome of menopause if local vaginal treatment alone is insufficient.
If women who have an intact uterus receive estrogen, they should also receive a progestin to protect against endometrial hyperplasia and reduce the risk of endometrial cancer.
Despite its status as the gold standard, estrogen-progestin therapy presents challenges. In some women, progestins cause side effects such as breast tenderness, bloating, fatigue, and depression.7 Estrogen-progestin therapy often causes vaginal bleeding, which for some women is troublesome or distressing; bleeding may be the reason for repeated evaluations, can increase anxiety, and can lead to poor adherence with hormonal treatment. Women who carry a higher-than-normal risk of developing breast cancer or fear that taking hormones will lead to breast cancer may show decreased adherence to therapy. Women who have estrogen receptor-positive breast cancer cannot take estrogen.
Individualized options are needed for women who have progestin-related side effects, unwanted vaginal bleeding, or a higher risk of breast cancer.
WELCOME THE ERAAs
An ideal treatment for menopause would relieve vasomotor symptoms and genitourinary syndrome of menopause and increase bone mineral density without causing breast tenderness, vaginal bleeding, or endometrial proliferation.
The “designer estrogens,” or ERAAs, have specific positive effects on the bone, heart, and brain with neutral or antagonist effects on estrogen receptors in other tissues such as the breasts and endometrium.8 While not entirely free of adverse effects, these agents have been developed with the aim of minimizing the most common ones related to estrogen and progestin.
Several ERAAs are currently approved by the US Food and Drug Administration (FDA)for various indications, each having a unique profile. Clomifene was the first agent of this class, and it is still used clinically to induce ovulation. This article highlights subsequently approved agents, ie, tamoxifen, raloxifene, ospemifene, and the combination of conjugated estrogens and bazedoxifene (Table 1).
All ERAAs increase the risk of venous thromboembolism, and therefore none of them should be used in women with known venous thromboembolism or at high risk of it.
TAMOXIFEN: CANCER TREATMENT AND PREVENTION
After clomiphene, tamoxifen was the second ERAA on the market. Although researchers were looking for a new contraceptive drug, they found tamoxifen to be useful as a chemotherapeutic agent for breast cancer. First used in 1971, tamoxifen continues to be one of the most commonly prescribed chemotherapeutic medications today.
The FDA has approved tamoxifen to treat breast cancer as well as to prevent breast cancer in pre- and postmenopausal women at risk. It may also have beneficial effects on bone and on cardiovascular risk factors, but these are not approved uses for it.
Trials of tamoxifen for cancer treatment
The Early Breast Cancer Trialists’ Collaborative Group9 performed a meta-analysis and found that 5 years of adjuvant treatment with tamoxifen is associated with a 26% reduction in mortality and a 47% reduction in breast cancer recurrence at 10 years. In absolute terms, we estimate that 21 women would need to be treated to prevent 1 death and 8 would need to be treated to prevent 1 recurrence.
The ATLAS Trial (Adjuvant Tamoxifen Longer Against Shorter)10 and later the UK ATTOM (Adjuvant Tamoxifen Treatment to Offer More)11 trial confirmed an even greater reduction in recurrence and mortality after a total of 10 years of treatment.
Trials of tamoxifen for cancer prevention
Cuzik et al12 performed a meta-analysis of 4 trials of tamoxifen’s effectiveness in preventing breast cancer for women at elevated risk. The incidence of estrogen receptor-positive breast cancer was 48% lower with tamoxifen use, but there was no effect on estrogen-negative breast cancer. From their data, we estimate that 77 women would need to be treated to prevent 1 case of breast cancer.
The IBIS-I trial (International Breast Cancer Intervention Study I)13 found that, in healthy women at high risk of breast cancer, the benefit of taking tamoxifen for 5 years as preventive treatment persisted long afterward. The investigators estimated that at 20 years of follow-up the risk of breast cancer would be 12.3% in placebo recipients and 7.8% in tamoxifen recipients, a 4.5% absolute risk reduction; number needed to treat (NNT) 22.
Data on tamoxifen and osteoporosis
The Breast Cancer Prevention Trial revealed a 19% reduction in the incidence of osteoporotic fractures with tamoxifen, but the difference was not statistically significant.14 The 1-year rates of fracture in women age 50 and older were 0.727% with placebo and 0.567% with tamoxifen, an absolute difference of 0.151%; therefore, if the effect is real, 662 women age 50 or older would need to be treated for 1 year to prevent 1 fracture. Tamoxifen is not FDA-approved to treat osteoporosis.
Data on tamoxifen and cardiovascular risk reduction
Chang et al,15 in a study in women at risk of breast cancer, incidentally found that tamoxifen was associated with a 13% reduction in total cholesterol compared with placebo.
Herrington and Klein,16 in a systematic review, noted similar findings in multiple studies of tamoxifen, with decreases in total cholesterol ranging from 7% to 17% and decreases in low-density lipoprotein cholesterol ranging from 10% to 28%. However, they found no change in high-density lipoprotein cholesterol concentrations or in the cardiovascular mortality rate.
The ATLAS trial10 revealed a relative risk reduction of 0.76 (95% confidence interval [CI] 0.60–0.95, P = .02) in ischemic heart disease for women who took tamoxifen for 10 years compared with 5 years. We calculate that ischemic heart disease occurred in 163 (2.5%) of 6,440 women who took tamoxifen for 5 years compared with 127 (1.9%) of 6,454 women who took it for 10 years, a 0.6% absolute risk reduction, NNT = 167.
Adverse effects of tamoxifen
Uterine neoplasia. Women taking tamoxifen have a 2.5-fold increased risk of endometrial cancer.14 Tamoxifen also increases the risk of benign uterine disease such as endometrial hyperplasia and polyps. As many as 39% of women taking tamoxifen will have evidence of benign uterine changes on pathology.17 Other adverse effects:
Venous thromboembolism (the risk of pulmonary embolism is increased approximately threefold14)
Cataracts (there is a slight increase in cataract diagnosis in tamoxifen users)
Vasomotor symptoms, which limit the use of tamoxifen in many women.
Ideal candidate for tamoxifen
The ideal candidate for tamoxifen is a woman with breast cancer that is estrogen receptor-positive and who has a history of osteopenia or osteoporosis and no risk factors for venous thromboembolism.
RALOXIFENE: FOR OSTEOPOROSIS AND FOR CANCER PREVENTION
Raloxifene, a second-generation ERAA, was first approved for preventing and treating osteoporosis and later for reducing the risk of invasive estrogen receptor-positive breast cancer in postmenopausal women.
Trials of raloxifene for osteoporosis
The MORE trial (Multiple Outcomes of Raloxifene)18 was a large multicenter randomized double-blind study. Raloxifene recipients showed a significant increase in bone mineral density in the lumbar spine and femoral neck at year 3 (P < .001) compared with those receiving placebo. Even after only 1 year of treatment, raloxifene significantly reduced the risk of new fractures, despite only modest gains in bone mineral density. After 3 years of treatment, new clinical vertebral fractures had occurred in 3.5% of the placebo group compared with 2.1% of the group receiving raloxifene 60 mg.19 Relative risk reductions were similar in women who had already had a clinical vertebral fracture at baseline, whose absolute risk is higher. However, no significant effect was seen on the incidence of hip or nonvertebral fractures.
The CORE trial (Continuing Outcomes Relevant to Raloxifene)20 extended the treatment of the women enrolled in the MORE trial another 4 years and found that the benefit of raloxifene with regard to bone mineral density persisted with continued use.
Trials of raloxifene for breast cancer prevention
The MORE trial,21 in postmenopausal women with osteoporosis included breast cancer as a secondary end point, and raloxifene was shown to decrease the incidence of invasive breast cancer. At a median of 40 months, invasive breast cancer had arisen in 13 (0.25%) of the 5,129 women assigned to raloxifene and 27 (1.0%) of the 2,576 women assigned to placebo. The authors calculated that 126 women would need to be treated to prevent 1 case of breast cancer.
The CORE trial,22 as noted, extended the treatment of the women enrolled in the MORE trial another 4 years. The risk of any invasive breast cancer in postmenopausal women with osteoporosis was significantly reduced by 59% after 8 years, and the risk of estrogen receptor-positive invasive breast cancer was reduced by 66%.
There is evidence that raloxifene’s effect on breast cancer risk persists after discontinuation of use.23
Does raloxifene reduce mortality?
Grady et al24 studied the effect of raloxifene on all-cause mortality in a pooled analysis of mortality data from the MORE, CORE, and Raloxifene Use for the Heart (RUTH)25 trials. In older postmenopausal women, the rate of all-cause mortality was 8.65% in those taking placebo compared with 7.88% in those taking raloxifene 60 mg daily—10% lower. The mechanism behind the lower mortality rate is unclear, and Grady et al recommend that the finding be interpreted with caution.
Trials of raloxifene for heart protection
The RUTH trial25 was a 5.6-year study undertaken to study the effects of raloxifene on coronary outcomes and invasive breast cancer in postmenopausal women. Results were mixed. Active treatment:
- Did not significantly affect the risk of coronary artery disease compared with placebo
- Significantly decreased the risk of invasive breast cancer
- Significantly decreased the risk of clinical vertebral fractures
- Increased the risk of fatal stroke (59 vs 39 events, hazard ratio 1.49, 95% CI 1.00–2.24) and venous thromboembolism (103 vs 71 events, hazard ratio 1.44, 95% CI 1.06–1.95).
The STAR trial (Study of Tamoxifen and Raloxifene)26,27 compared raloxifene and tamoxifen in postmenopausal women at increased risk of breast cancer. Women were randomized to receive either tamoxifen 20 mg or raloxifene 60 mg for 5 years. Results:
- No difference in the number of new cases of invasive breast cancer between the groups
- Fewer cases of noninvasive breast cancer in the tamoxifen group, but the difference was not statistically significant
- Fewer cases of uterine cancer in the raloxifene group, annual incidence rates 0.125% vs 0.199%, absolute risk reduction 0.74%, NNT 1,351, relative risk with raloxifene 0.62, 95% CI 0.30–0.50
- Fewer thromboembolic events with raloxifene
- Fewer cataracts with raloxifene.
Adverse effects of raloxifene
Raloxifene increases the risk of venous thromboembolism and stroke in women at high risk of coronary artery disease.19
Ideal candidates for raloxifene
Postmenopausal women with osteopenia or osteoporosis and a higher risk of breast cancer who have minimal to no vasomotor symptoms or genitourinary syndrome of menopause are good candidates for raloxifene. Raloxifene is also a good choice for women who have genitourinary syndrome of menopause treated with local vaginal estrogen. Raloxifene has no effect on vasomotor symptoms or genitourinary syndrome of menopause.
OSPEMIFENE: FOR GENITOURINARY SYNDROME OF MENOPAUSE
Although ospemifene does not have the steroid structure of estrogen, it acts as an estrogen agonist specifically in the vaginal mucosa and an antagonist in other tissues.28 It has been shown on Papanicolaou smears to reduce the number of parabasal cells and increase the number of intermediate and superficial cells after 3 months of treatment.29
Ospemifene 60 mg taken orally with food is approved by the FDA to treat genitourinary syndrome of menopause.
Why ospemifene is needed
First-line treatment options for genitourinary syndrome of menopause include over-the-counter lubricants. However, there is no evidence that these products reverse vaginal atrophy,30 and many women report no relief of symptoms with them.
While various local estrogen preparations positively affect genitourinary syndrome of menopause, some of them can be messy, which can limit-long term adherence.
In one of the largest surveys on genitourinary syndrome of menopause (the REVIVE survey—the Real Women’s View of Treatment Options for Menopausal Vaginal Changes29), 59% of women reported that their vaginal symptoms negatively affected sexual activity. The problem affects not only the patient but also her sexual partner.31 Another large study showed that 38% of women and 39% of male partners reported that it had a worse-than-expected impact on their intimate relationships.31
Genitourinary syndrome of menopause also makes pelvic examinations difficult, may worsen or exacerbate cystitis, and may increase urinary tract infections.
Trials of ospemifene for genitourinary syndrome of menopause
To date, 3 randomized, double-blind clinical trials have demonstrated ospemifene 60 mg to be superior to placebo in treating genitourinary syndrome of menopause. Two were short-term (12-week) and showed significant positive changes in the percent of superficial cells, vaginal pH (lower is better), and number of parabasal cells, along with improvements in the Likert rating of both vaginal dryness and dyspareunia.32,33
A long-term (52-week) randomized placebo-controlled trial compared ospemifene and placebo and showed significant improvement in vaginal maturation index and pH at weeks 12 and 52.34 Other outcome measures included petechiae, pallor, friability, erythema, and dryness, all of which improved from baseline (P < .001). At the end of the trial, 80% of the patients who received ospemifene had no vaginal atrophy.
No serious adverse events were noted in any of the clinical trials to date, and a systemic review and meta-analysis demonstrated ospemifene to be safe and efficacious.35 The most frequently reported reasons for discontinuation were hot flashes, vaginal discharge, muscle spasms, and hyperhidrosis, but the rates of these effects were similar to those with placebo.
Trial of ospemifene’s effect on bone turnover
As an estrogen receptor agonist in bone, ospemifene decreases the levels of bone turnover markers in postmenopausal women.36 A study found ospemifene to be about as effective as raloxifene in suppressing bone turnover,37 but ospemifene does not carry FDA approval for preventing or treating osteoporosis.
Other effects
In experiments in rats, the incidence of breast cancer appears to be lower with ospemifene, and the higher the dose, the lower the incidence.38
Ospemifene also has antagonistic effects on uterine tissue, and no cases of endometrial hyperplasia or carcinoma have been reported in short-term or long-term studies.35
Ospemifene has no effect however on vasomotor symptoms and may in fact worsen vasomotor symptoms in women suffering with hot flashes and night sweats. Further investigation into its long-term safety and effects on breast tissue and bone would provide more insight.
Ideal candidates for ospemifene
Ospemifene could help postmenopausal women with genitourinary syndrome of menopause for whom over-the-counter lubricants fail, who dislike local vaginal estrogen, or who decline systemic hormone therapy, and who do not meet the criteria for treatment with systemic hormone therapy.
CONJUGATED ESTROGENS AND BAZEDOXIFENE COMBINATION
A combination agent consisting of conjugated estrogens 0.45 mg plus bazedoxifene 20 mg has been approved by the FDA for treating moderate to severe vasomotor symptoms associated with menopause and also for preventing postmenopausal osteoporosis in women who have an intact uterus.
Trials of estrogen-bazedoxifene for vasomotor symptoms
The Selective Estrogen Menopause and Response to Therapy (SMART) trials39,40 were a series of randomized, double-blind, placebo-controlled phase 3 studies evaluating the efficacy and safety of the estrogen-bazedoxifene combination in postmenopausal women.
The SMART-2 trial39 evaluated the combination of conjugated estrogens (either 0.45 mg or 0.625) plus bazedoxifene 20 mg and found both dosages significantly reduced the number and severity of hot flashes at weeks 4 and 12 (P < .001). At week 12, the combination with 0.45 mg of estrogen reduced vasomotor symptoms from baseline by 74% (10.3 hot flashes per week at baseline vs 2.8 at week 12); the combination with 0.625 mg of estrogen reduced vasomotor symptoms by 80% (10.4 vs 2.4 flashes); and placebo reduced them by 51% (10.5 vs 5.4 flashes).
For bone density. The SMART-1 trial40 showed that the estrogen-bazedoxifene combination in both estrogen dosages significantly increased mean lumbar spine bone mineral density (P < .001) and total hip bone mineral density (P < .05) from baseline at 12 and 24 months compared with placebo. Increases in density tended to be higher with the higher estrogen dose (0.625 mg), but less with higher doses of bazedoxifene.41 At 24 months, the increase in bone mineral density was even greater than in women treated with raloxifene.42 However, the effect of estrogen-bazedoxifene on the incidence of fractures remains to be studied.
For genitourinary syndrome of menopause. The SMART-3 trial showed that treatment with conjugated estrogens plus bazedoxifene (0.45/20 mg or 0.625/20 mg) was more effective than placebo in increasing the percent of superficial and intermediate cells and decreased the number of parabasal cells at 12 weeks compared with placebo (P < .01).43 Both doses also significantly decreased the mean vaginal pH and improved vaginal dryness.
Patients treated with estrogen-bazedoxifene for a minimum of 12 weeks in a double-blind placebo-controlled study also showed a significant improvement in sexual function and quality-of-life measurements based on 3 well-defined scales, which included ease of lubrication, satisfaction with treatment, control of hot flashes, and sleep parameters.43
Low rates of side effects
To evaluate this regimen’s antagonistic effects on uterine tissue, endometrial hyperplasia was diagnosed by blinded pathologists using endometrial biopsies taken at 6, 12, and 24 months or more if cancer was a suspected diagnosis. At 12 and 24 months of treatment, the incidence of hyperplasia with bazedoxifene 20 or 40 mg at doses of either 0.45 or 0.625 mg of conjugated estrogens was less than 1%, which was similar to placebo rates over the 24 months.44 The lowest dose studied, bazedoxifene 10 mg, did not prevent hyperplasia with conjugated estrogens 0.45 or 0.625 mg, and its use was discontinued.
Rates of amenorrhea with bazedoxifene 20 or 40 mg and conjugated estrogens 0.45 or 0.625 mg were very favorable (83%–93%) and similar to those with placebo.45 For women with continued bleeding on hormone therapy requiring multiple evaluations, or for women who won’t accept the risk of bleeding on hormone therapy, conjugated estrogens and bazedoxifene may be a sustainable option. However, any woman with abnormal bleeding should undergo prompt immediate evaluation.
A typical side effect of estrogen replacement therapy is breast tenderness. For women seeking vasomotor symptom treatment but who experience breast tenderness, this may be a deterrent from continuing hormone therapy. As shown in the SMART-1 and SMART-2 trials,46 conjugated estrogens and bazedoxifene did not cause an increase in breast tenderness, which may enhance medication adherence.
Ideal candidates for conjugated estrogens plus bazedoxifene
This product could help postmenopausal women who have an intact uterus and are suffering with moderate to severe vasomotor symptoms and genitourinary syndrome of menopause who cannot tolerate the side effects of hormone therapy such as bleeding, bloating, or breast tenderness, or who prefer to take an estrogen but without a progestin. It is also ideal for women at higher risk of osteoporosis.
WHO SHOULD GET WHAT?
Not all postmenopausal women have vasomotor symptoms, genitourinary syndrome of menopause, or bone loss. For those who do, standard hormone therapy is an option.
For those who have symptoms and a lower threshold of side effects such as breast tenderness and vaginal bleeding, a combination of an estrogen plus an ERAA (eg, bazedoxifene) is an option.
For women who have no vasomotor symptoms but do have genitourinary syndrome of menopause and don’t want local vaginal treatment, ospemifene is an option.
For women with no vasomotor symptoms but who have bone loss and increased risk of estrogen receptor-positive breast cancer, raloxifene is a good option.
Both premenopausal and postmenopausal women who are at increased risk for breast cancer should be considered for tamoxifen chemoprevention. Postmenopausal women with a uterus at increased risk for breast cancer should be considered for raloxifene, as it has no uterine effect. Raloxifene is not indicated in premenopausal women.
No woman at increased risk of venous thromboembolism is a candidate for ERAA treatment or for oral estrogen. However, the clinician has multiple options to improve quality of life and work productivity and reduce office visits of women at midlife, especially when they are individually assessed and treated.
- Giannini A, Russo E, Mannella P, Simoncini T. Selective steroid receptor modulators in reproductive medicine. Minerva Ginecol 2015; 67:431–455.
- Feldman BM, Voda A, Gronseth E. The prevalence of hot flash and associated variables among perimenopausal women. Res Nurs Health 1985; 8:261–268.
- Versi E, Harvey MA, Cardozo L, Brincat M, Studd JW. Urogenital prolapse and atrophy at menopause: a prevalence study. Int Urogynecol J Pelvic Floor Dysfunct 2001; 12:107–110.
- Hess R, Chang CC, Conigliaro J, McNeil M. Understanding physicians’ attitudes towards hormone therapy. Womens Health Issues 2005; 15:31–38.
- Melton LJ 3rd, Khosla S, Atkinson EJ, O’Fallon WM, Riggs BL. Relationship of bone turnover to bone density and fractures. J Bone Miner Res 1997; 12:1083–1091.
- Sikon A, Thacker HL. Treatment options for menopausal hot flashes. Cleve Clin J Med 2004; 71:578–582.
- Levine JP. Treating menopausal symptoms with a tissue-selective estrogen complex. Gend Med 2011; 8:57–68.
- Pinkerton JV, Thomas S. Use of SERMs for treatment in postmenopausal women. J Steroid Biochem Mol Biol 2014; 142:142–154.
- Tamoxifen for early breast cancer: an overview of the randomised trials. Early Breast Cancer Trialists’ Collaborative Group. Lancet 1998; 351:1451–1467.
- Davies C, Pan H, Godwin J, et al; Adjuvant Tamoxifen: Longer Against Shorter (ATLAS) Collaborative Group. Long-term effects of continuing adjuvant tamoxifen to 10 years versus stopping at 5 years after diagnosis of oestrogen receptor-positive breast cancer: ATLAS, a randomised trial. Lancet 2013; 381:805–816.
- Gray RG, Rea D, Handley K, et al. aTTom: Long-term effects of continuing adjuvant tamoxifen to 10 years versus stopping at 5 years in 6,953 women with early breast cancer. J Clin Oncol 2013; (suppl): abstract 5.
- Cuzick J, Powles T, Veronesi U, et al. Overview of the main outcomes in breast-cancer prevention trials. Lancet 2003; 361:296–300.
- Cuzick J, Sestak I, Cawthorn S, et al. Tamoxifen for prevention of breast cancer: extended long-term follow-up of the IBIS-I breast cancer prevention trial. Lancet Oncol 2015; 16:67–75.
- Fisher B, Costantino JP, Wickerham DL, et al. Tamoxifen for prevention of breast cancer: report of the National Surgical Adjuvant Breast and Bowel Project P-1 Study. J Natl Cancer Inst 1998; 90:1371–1388.
- Chang J, Powles TJ, Ashley SE, et al. The effect of tamoxifen and hormone replacement therapy on serum cholesterol, bone mineral density and coagulation factors in healthy postmenopausal women participating in a randomised, controlled tamoxifen prevention study. Ann Oncol 1996; 7:671–675.
- Herrington DM, Klein KP. Effects of SERMs on important indicators of cardiovascular health: lipoproteins, hemostatic factors and endothelial function. Womens Health Issues 2001; 11:95–102.
- Kedar RP, Bourne TH, Powles TJ, et al. Effects of tamoxifen on uterus and ovaries of postmenopausal women in a randomized breast cancer prevention trial. Lancet 1994; 343:1318–1321.
- Ettinger B, Black DM, Mitlak BH, et al. Reduction of vertebral fracture risk in postmenopausal women with osteoporosis treated with raloxifene: results from a 3-year randomized clinical trial. Multiple Outcomes of Raloxifene Evaluation (MORE) Investigators. JAMA 1999; 282:637–645.
- Maricic M, Adachi JD, Sarkar S, Wu W, Wong M, Harper KD. Early effects of raloxifene on clinical vertebral fractures at 12 months in postmenopausal women with osteoporosis. Arch Intern Med 2002; 162:1140–1143.
- Recker RR, Mitlak BH, Ni X, Krege JH. Long-term raloxifene for postmenopausal osteoporosis. Curr Med Res Opin 2011; 27:1755–1761.
- Cummings SR, Eckert S, Krueger KA, et al. The effect of raloxifene on risk of breast cancer in postmenopausal women: results from the MORE randomized trial. Multiple Outcomes of Raloxifene Evaluation. JAMA 1999; 281:2189–2197.
- Martino S, Cauley JA, Barrett-Connor E, et al; CORE Investigators. Continuing outcomes relevant to Evista: breast cancer incidence in postmenopausal osteoporotic women in a randomized trial of raloxifene. J Natl Cancer Inst 2004; 96:1751–1761.
- Vogel VG, Qu Y, Wong M, Mitchell B, Mershon JL. Incidence of invasive breast cancer in postmenopausal women after discontinuation of long-term raloxifene administration. Clin Breast Cancer 2009; 9:45–50.
- Grady D, Cauley JA, Stock JL, et al. Effect of raloxifene on all-cause mortality. Am J Med 2010; 123:469.e1–e7.
- Barrett-Connor E, Mosca L, Collins P, et al; Raloxifene Use for The Heart (RUTH) Trial Investigators. Effects of raloxifene on cardiovascular events and breast cancer in postmenopausal women. N Engl J Med 2006; 355:125–137.
- Vogel VG. The NSABP Study of Tamoxifen and Raloxifene (STAR) trial. Expert Rev Anticancer Ther 2009; 9:51–60.
- Vogel VG, Costantino JP, Wickerham DL, et al; National Surgical Adjuvant Breast and Bowel Project (NSABP). Effects of tamoxifen vs raloxifene on the risk of developing invasive breast cancer and other disease outcomes: the NSABP Study of Tamoxifen and Raloxifene (STAR) P-2 trial. JAMA 2006; 295:2727–2741.
- Barnes KN, Pearce EF, Yancey AM, Forinash AB. Ospemifene in the treatment of vulvovaginal atrophy. Ann Pharmacother 2014; 48:752–757.
- Rutanen EM, Heikkinen J, Halonen K, Komi J, Lammintausta R, Ylikorkala O. Effects of ospemifene, a novel SERM, on hormones, genital tract, climacteric symptoms, and quality of life in postmenopausal women: a double-blind, randomized trial. Menopause 2003; 10:433–439.
- Constantine G, Graham S, Koltun WD, Kingsberg SA. Assessment of ospemifene or lubricants on clinical signs of VVA. J Sex Med 2014; 11:1033–1041.
- Kingsberg SA, Wysocki S, Magnus L, Krychman ML. Vulvar and vaginal atrophy in postmenopausal women: findings from the REVIVE survey. J Sex Med 2013; 10:1790–1799.
- Portman DJ, Bachmann GA, Simon JA; Ospemifene Study Group. Ospemifene, a novel selective estrogen receptor modulator for treating dyspareunia associated with postmenopausal vulvar and vaginal atrophy. Menopause 2013; 20:623–630.
- Bachmann GA, Komi JO; Ospemifene Study Group. Ospemifene effectively treats vulvovaginal atrophy in postmenopausal women: results from a pivotal phase 3 study. Menopause 2010; 17:480–486.
- Goldstein SR, Bachmann GA, Koninckx PR, Lin VH, Portman DJ, Ylikorkala O; Ospemifene Study Group. Ospemifene 12-month safety and efficacy in postmenopausal women with vulvar and vaginal atrophy. Climacteric 2014; 17:173–182.
- Cui Y, Zong H, Yan H, Li N, Zhang Y. The efficacy and safety of ospemifene in treating dyspareunia associated with postmenopausal vulvar and vaginal atrophy: a systematic review and meta-analysis. J Sex Med 2014; 11:487–497.
- Komi J, Heikkinen J, Rutanen EM, Halonen K, Lammintausta R, Ylikorkala O. Effects of ospemifene, a novel SERM, on biochemical markers of bone turnover in healthy postmenopausal women. Gynecal Endocrinol 2004; 18:152–158.
- Komi J, Lankinen KS, DeGregorio M, et al. Effects of ospemifene and raloxifene on biochemical markers of bone turnover in postmenopausal women. J Bone Miner Metab 2006; 24:314–318.
- 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; 97:230–240.
- 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; 16:1116–1124.
- Pickar JH, Mirkin S. Tissue-selective agents: selective estrogen receptor modulators and the tissue-selective estrogen complex. Menopause Int 2010; 16:121–128.
- Levine JP. Treating menopausal symptoms with a tissue-selective estrogen complex. Gend Med 2011; 8:57–68.
- Lindsay R, Gallagher JC, Kagan R, Pickar JH, Constantine G. Efficacy of tissue-selective estrange complex of bazedoxifene/conjugated estrogens for osteoporosis prevention in at-risk postmenopausal women. Fertil Steril 2009; 92:1045–1052.
- Bachmann G, Bobula J, Mirkin S. Effects of bazedoxifene/conjugated estrogens on quality of life in postmenopausal women with symptoms of vulvar/vaginal atrophy. Climacteric 2010; 13:132–140.
- Pickar JH, Yeh IT, Bachmann G, Speroff L. Endometrial effects of a tissue selective estrogen complex containing bazedoxifene/conjugated estrogens as a menopausal therapy. Fertil Steril 2009; 92:1018–1024.
- Archer DF, Lewis V, Carr BR, Olivier S, Pickar JH. Bazedoxifene/conjugated estrogens (BZA/CE): incidence of uterine bleeding in postmenopausal women. Fertil Steril 2009: 92:1039–1044.
- Pinkerton JV, Abraham L, Bushmakin AG, et al. Evaluation of the efficacy and safety of bazedoxifene/conjugated estrogens for secondary outcomes including vasomotor symptoms in postmenopausal women by years since menopause in the Selective estrogens, Menopause and Response to Therapy (SMART) trials. J Womens Health (Larchmt) 2014; 23:18–28.
Estrogen receptor agonist-antagonists (ERAAs), previously called selective estrogen receptor modulators (SERMs), have extended the options for treating the various conditions that menopausal women suffer from. These drugs act differently on estrogen receptors in different tissues, stimulating receptors in some tissues but inhibiting them in others. This allows selective inhibition or stimulation of estrogen-like action in various target tissues.1
This article highlights the use of ERAAs to treat menopausal vasomotor symptoms (eg, hot flashes, night sweats), genitourinary syndrome of menopause, osteoporosis, breast cancer (and the risk of breast cancer), and other health concerns unique to women at midlife.
SYMPTOMS OF MENOPAUSE: COMMON AND TROUBLESOME
Vasomotor symptoms such as hot flashes and night sweats are common during perimenopause—most women experience them. They are most frequent during the menopause transition but can persist for 10 years or more afterward.2
Genitourinary syndrome of menopause is also common and often worsens with years after menopause.3 It can lead to dyspareunia and vaginal dryness, which may in turn result in lower libido, vaginismus, and hypoactive sexual desire disorder, problems that often arise at the same time as vaginal dryness and atrophy.4
Osteopenia and osteoporosis. A drop in systemic estrogen leads to a decline in bone mineral density, increasing the risk of fractures.5
ESTROGEN-PROGESTIN TREATMENT: THE GOLD STANDARD, BUT NOT IDEAL
The current gold standard for treating moderate to severe hot flashes is estrogen, available in oral, transdermal, and vaginal formulations.6 Estrogen also has antiresorptive effects on bone and is approved for preventing osteoporosis. Systemic estrogen may also be prescribed for genitourinary syndrome of menopause if local vaginal treatment alone is insufficient.
If women who have an intact uterus receive estrogen, they should also receive a progestin to protect against endometrial hyperplasia and reduce the risk of endometrial cancer.
Despite its status as the gold standard, estrogen-progestin therapy presents challenges. In some women, progestins cause side effects such as breast tenderness, bloating, fatigue, and depression.7 Estrogen-progestin therapy often causes vaginal bleeding, which for some women is troublesome or distressing; bleeding may be the reason for repeated evaluations, can increase anxiety, and can lead to poor adherence with hormonal treatment. Women who carry a higher-than-normal risk of developing breast cancer or fear that taking hormones will lead to breast cancer may show decreased adherence to therapy. Women who have estrogen receptor-positive breast cancer cannot take estrogen.
Individualized options are needed for women who have progestin-related side effects, unwanted vaginal bleeding, or a higher risk of breast cancer.
WELCOME THE ERAAs
An ideal treatment for menopause would relieve vasomotor symptoms and genitourinary syndrome of menopause and increase bone mineral density without causing breast tenderness, vaginal bleeding, or endometrial proliferation.
The “designer estrogens,” or ERAAs, have specific positive effects on the bone, heart, and brain with neutral or antagonist effects on estrogen receptors in other tissues such as the breasts and endometrium.8 While not entirely free of adverse effects, these agents have been developed with the aim of minimizing the most common ones related to estrogen and progestin.
Several ERAAs are currently approved by the US Food and Drug Administration (FDA)for various indications, each having a unique profile. Clomifene was the first agent of this class, and it is still used clinically to induce ovulation. This article highlights subsequently approved agents, ie, tamoxifen, raloxifene, ospemifene, and the combination of conjugated estrogens and bazedoxifene (Table 1).
All ERAAs increase the risk of venous thromboembolism, and therefore none of them should be used in women with known venous thromboembolism or at high risk of it.
TAMOXIFEN: CANCER TREATMENT AND PREVENTION
After clomiphene, tamoxifen was the second ERAA on the market. Although researchers were looking for a new contraceptive drug, they found tamoxifen to be useful as a chemotherapeutic agent for breast cancer. First used in 1971, tamoxifen continues to be one of the most commonly prescribed chemotherapeutic medications today.
The FDA has approved tamoxifen to treat breast cancer as well as to prevent breast cancer in pre- and postmenopausal women at risk. It may also have beneficial effects on bone and on cardiovascular risk factors, but these are not approved uses for it.
Trials of tamoxifen for cancer treatment
The Early Breast Cancer Trialists’ Collaborative Group9 performed a meta-analysis and found that 5 years of adjuvant treatment with tamoxifen is associated with a 26% reduction in mortality and a 47% reduction in breast cancer recurrence at 10 years. In absolute terms, we estimate that 21 women would need to be treated to prevent 1 death and 8 would need to be treated to prevent 1 recurrence.
The ATLAS Trial (Adjuvant Tamoxifen Longer Against Shorter)10 and later the UK ATTOM (Adjuvant Tamoxifen Treatment to Offer More)11 trial confirmed an even greater reduction in recurrence and mortality after a total of 10 years of treatment.
Trials of tamoxifen for cancer prevention
Cuzik et al12 performed a meta-analysis of 4 trials of tamoxifen’s effectiveness in preventing breast cancer for women at elevated risk. The incidence of estrogen receptor-positive breast cancer was 48% lower with tamoxifen use, but there was no effect on estrogen-negative breast cancer. From their data, we estimate that 77 women would need to be treated to prevent 1 case of breast cancer.
The IBIS-I trial (International Breast Cancer Intervention Study I)13 found that, in healthy women at high risk of breast cancer, the benefit of taking tamoxifen for 5 years as preventive treatment persisted long afterward. The investigators estimated that at 20 years of follow-up the risk of breast cancer would be 12.3% in placebo recipients and 7.8% in tamoxifen recipients, a 4.5% absolute risk reduction; number needed to treat (NNT) 22.
Data on tamoxifen and osteoporosis
The Breast Cancer Prevention Trial revealed a 19% reduction in the incidence of osteoporotic fractures with tamoxifen, but the difference was not statistically significant.14 The 1-year rates of fracture in women age 50 and older were 0.727% with placebo and 0.567% with tamoxifen, an absolute difference of 0.151%; therefore, if the effect is real, 662 women age 50 or older would need to be treated for 1 year to prevent 1 fracture. Tamoxifen is not FDA-approved to treat osteoporosis.
Data on tamoxifen and cardiovascular risk reduction
Chang et al,15 in a study in women at risk of breast cancer, incidentally found that tamoxifen was associated with a 13% reduction in total cholesterol compared with placebo.
Herrington and Klein,16 in a systematic review, noted similar findings in multiple studies of tamoxifen, with decreases in total cholesterol ranging from 7% to 17% and decreases in low-density lipoprotein cholesterol ranging from 10% to 28%. However, they found no change in high-density lipoprotein cholesterol concentrations or in the cardiovascular mortality rate.
The ATLAS trial10 revealed a relative risk reduction of 0.76 (95% confidence interval [CI] 0.60–0.95, P = .02) in ischemic heart disease for women who took tamoxifen for 10 years compared with 5 years. We calculate that ischemic heart disease occurred in 163 (2.5%) of 6,440 women who took tamoxifen for 5 years compared with 127 (1.9%) of 6,454 women who took it for 10 years, a 0.6% absolute risk reduction, NNT = 167.
Adverse effects of tamoxifen
Uterine neoplasia. Women taking tamoxifen have a 2.5-fold increased risk of endometrial cancer.14 Tamoxifen also increases the risk of benign uterine disease such as endometrial hyperplasia and polyps. As many as 39% of women taking tamoxifen will have evidence of benign uterine changes on pathology.17 Other adverse effects:
Venous thromboembolism (the risk of pulmonary embolism is increased approximately threefold14)
Cataracts (there is a slight increase in cataract diagnosis in tamoxifen users)
Vasomotor symptoms, which limit the use of tamoxifen in many women.
Ideal candidate for tamoxifen
The ideal candidate for tamoxifen is a woman with breast cancer that is estrogen receptor-positive and who has a history of osteopenia or osteoporosis and no risk factors for venous thromboembolism.
RALOXIFENE: FOR OSTEOPOROSIS AND FOR CANCER PREVENTION
Raloxifene, a second-generation ERAA, was first approved for preventing and treating osteoporosis and later for reducing the risk of invasive estrogen receptor-positive breast cancer in postmenopausal women.
Trials of raloxifene for osteoporosis
The MORE trial (Multiple Outcomes of Raloxifene)18 was a large multicenter randomized double-blind study. Raloxifene recipients showed a significant increase in bone mineral density in the lumbar spine and femoral neck at year 3 (P < .001) compared with those receiving placebo. Even after only 1 year of treatment, raloxifene significantly reduced the risk of new fractures, despite only modest gains in bone mineral density. After 3 years of treatment, new clinical vertebral fractures had occurred in 3.5% of the placebo group compared with 2.1% of the group receiving raloxifene 60 mg.19 Relative risk reductions were similar in women who had already had a clinical vertebral fracture at baseline, whose absolute risk is higher. However, no significant effect was seen on the incidence of hip or nonvertebral fractures.
The CORE trial (Continuing Outcomes Relevant to Raloxifene)20 extended the treatment of the women enrolled in the MORE trial another 4 years and found that the benefit of raloxifene with regard to bone mineral density persisted with continued use.
Trials of raloxifene for breast cancer prevention
The MORE trial,21 in postmenopausal women with osteoporosis included breast cancer as a secondary end point, and raloxifene was shown to decrease the incidence of invasive breast cancer. At a median of 40 months, invasive breast cancer had arisen in 13 (0.25%) of the 5,129 women assigned to raloxifene and 27 (1.0%) of the 2,576 women assigned to placebo. The authors calculated that 126 women would need to be treated to prevent 1 case of breast cancer.
The CORE trial,22 as noted, extended the treatment of the women enrolled in the MORE trial another 4 years. The risk of any invasive breast cancer in postmenopausal women with osteoporosis was significantly reduced by 59% after 8 years, and the risk of estrogen receptor-positive invasive breast cancer was reduced by 66%.
There is evidence that raloxifene’s effect on breast cancer risk persists after discontinuation of use.23
Does raloxifene reduce mortality?
Grady et al24 studied the effect of raloxifene on all-cause mortality in a pooled analysis of mortality data from the MORE, CORE, and Raloxifene Use for the Heart (RUTH)25 trials. In older postmenopausal women, the rate of all-cause mortality was 8.65% in those taking placebo compared with 7.88% in those taking raloxifene 60 mg daily—10% lower. The mechanism behind the lower mortality rate is unclear, and Grady et al recommend that the finding be interpreted with caution.
Trials of raloxifene for heart protection
The RUTH trial25 was a 5.6-year study undertaken to study the effects of raloxifene on coronary outcomes and invasive breast cancer in postmenopausal women. Results were mixed. Active treatment:
- Did not significantly affect the risk of coronary artery disease compared with placebo
- Significantly decreased the risk of invasive breast cancer
- Significantly decreased the risk of clinical vertebral fractures
- Increased the risk of fatal stroke (59 vs 39 events, hazard ratio 1.49, 95% CI 1.00–2.24) and venous thromboembolism (103 vs 71 events, hazard ratio 1.44, 95% CI 1.06–1.95).
The STAR trial (Study of Tamoxifen and Raloxifene)26,27 compared raloxifene and tamoxifen in postmenopausal women at increased risk of breast cancer. Women were randomized to receive either tamoxifen 20 mg or raloxifene 60 mg for 5 years. Results:
- No difference in the number of new cases of invasive breast cancer between the groups
- Fewer cases of noninvasive breast cancer in the tamoxifen group, but the difference was not statistically significant
- Fewer cases of uterine cancer in the raloxifene group, annual incidence rates 0.125% vs 0.199%, absolute risk reduction 0.74%, NNT 1,351, relative risk with raloxifene 0.62, 95% CI 0.30–0.50
- Fewer thromboembolic events with raloxifene
- Fewer cataracts with raloxifene.
Adverse effects of raloxifene
Raloxifene increases the risk of venous thromboembolism and stroke in women at high risk of coronary artery disease.19
Ideal candidates for raloxifene
Postmenopausal women with osteopenia or osteoporosis and a higher risk of breast cancer who have minimal to no vasomotor symptoms or genitourinary syndrome of menopause are good candidates for raloxifene. Raloxifene is also a good choice for women who have genitourinary syndrome of menopause treated with local vaginal estrogen. Raloxifene has no effect on vasomotor symptoms or genitourinary syndrome of menopause.
OSPEMIFENE: FOR GENITOURINARY SYNDROME OF MENOPAUSE
Although ospemifene does not have the steroid structure of estrogen, it acts as an estrogen agonist specifically in the vaginal mucosa and an antagonist in other tissues.28 It has been shown on Papanicolaou smears to reduce the number of parabasal cells and increase the number of intermediate and superficial cells after 3 months of treatment.29
Ospemifene 60 mg taken orally with food is approved by the FDA to treat genitourinary syndrome of menopause.
Why ospemifene is needed
First-line treatment options for genitourinary syndrome of menopause include over-the-counter lubricants. However, there is no evidence that these products reverse vaginal atrophy,30 and many women report no relief of symptoms with them.
While various local estrogen preparations positively affect genitourinary syndrome of menopause, some of them can be messy, which can limit-long term adherence.
In one of the largest surveys on genitourinary syndrome of menopause (the REVIVE survey—the Real Women’s View of Treatment Options for Menopausal Vaginal Changes29), 59% of women reported that their vaginal symptoms negatively affected sexual activity. The problem affects not only the patient but also her sexual partner.31 Another large study showed that 38% of women and 39% of male partners reported that it had a worse-than-expected impact on their intimate relationships.31
Genitourinary syndrome of menopause also makes pelvic examinations difficult, may worsen or exacerbate cystitis, and may increase urinary tract infections.
Trials of ospemifene for genitourinary syndrome of menopause
To date, 3 randomized, double-blind clinical trials have demonstrated ospemifene 60 mg to be superior to placebo in treating genitourinary syndrome of menopause. Two were short-term (12-week) and showed significant positive changes in the percent of superficial cells, vaginal pH (lower is better), and number of parabasal cells, along with improvements in the Likert rating of both vaginal dryness and dyspareunia.32,33
A long-term (52-week) randomized placebo-controlled trial compared ospemifene and placebo and showed significant improvement in vaginal maturation index and pH at weeks 12 and 52.34 Other outcome measures included petechiae, pallor, friability, erythema, and dryness, all of which improved from baseline (P < .001). At the end of the trial, 80% of the patients who received ospemifene had no vaginal atrophy.
No serious adverse events were noted in any of the clinical trials to date, and a systemic review and meta-analysis demonstrated ospemifene to be safe and efficacious.35 The most frequently reported reasons for discontinuation were hot flashes, vaginal discharge, muscle spasms, and hyperhidrosis, but the rates of these effects were similar to those with placebo.
Trial of ospemifene’s effect on bone turnover
As an estrogen receptor agonist in bone, ospemifene decreases the levels of bone turnover markers in postmenopausal women.36 A study found ospemifene to be about as effective as raloxifene in suppressing bone turnover,37 but ospemifene does not carry FDA approval for preventing or treating osteoporosis.
Other effects
In experiments in rats, the incidence of breast cancer appears to be lower with ospemifene, and the higher the dose, the lower the incidence.38
Ospemifene also has antagonistic effects on uterine tissue, and no cases of endometrial hyperplasia or carcinoma have been reported in short-term or long-term studies.35
Ospemifene has no effect however on vasomotor symptoms and may in fact worsen vasomotor symptoms in women suffering with hot flashes and night sweats. Further investigation into its long-term safety and effects on breast tissue and bone would provide more insight.
Ideal candidates for ospemifene
Ospemifene could help postmenopausal women with genitourinary syndrome of menopause for whom over-the-counter lubricants fail, who dislike local vaginal estrogen, or who decline systemic hormone therapy, and who do not meet the criteria for treatment with systemic hormone therapy.
CONJUGATED ESTROGENS AND BAZEDOXIFENE COMBINATION
A combination agent consisting of conjugated estrogens 0.45 mg plus bazedoxifene 20 mg has been approved by the FDA for treating moderate to severe vasomotor symptoms associated with menopause and also for preventing postmenopausal osteoporosis in women who have an intact uterus.
Trials of estrogen-bazedoxifene for vasomotor symptoms
The Selective Estrogen Menopause and Response to Therapy (SMART) trials39,40 were a series of randomized, double-blind, placebo-controlled phase 3 studies evaluating the efficacy and safety of the estrogen-bazedoxifene combination in postmenopausal women.
The SMART-2 trial39 evaluated the combination of conjugated estrogens (either 0.45 mg or 0.625) plus bazedoxifene 20 mg and found both dosages significantly reduced the number and severity of hot flashes at weeks 4 and 12 (P < .001). At week 12, the combination with 0.45 mg of estrogen reduced vasomotor symptoms from baseline by 74% (10.3 hot flashes per week at baseline vs 2.8 at week 12); the combination with 0.625 mg of estrogen reduced vasomotor symptoms by 80% (10.4 vs 2.4 flashes); and placebo reduced them by 51% (10.5 vs 5.4 flashes).
For bone density. The SMART-1 trial40 showed that the estrogen-bazedoxifene combination in both estrogen dosages significantly increased mean lumbar spine bone mineral density (P < .001) and total hip bone mineral density (P < .05) from baseline at 12 and 24 months compared with placebo. Increases in density tended to be higher with the higher estrogen dose (0.625 mg), but less with higher doses of bazedoxifene.41 At 24 months, the increase in bone mineral density was even greater than in women treated with raloxifene.42 However, the effect of estrogen-bazedoxifene on the incidence of fractures remains to be studied.
For genitourinary syndrome of menopause. The SMART-3 trial showed that treatment with conjugated estrogens plus bazedoxifene (0.45/20 mg or 0.625/20 mg) was more effective than placebo in increasing the percent of superficial and intermediate cells and decreased the number of parabasal cells at 12 weeks compared with placebo (P < .01).43 Both doses also significantly decreased the mean vaginal pH and improved vaginal dryness.
Patients treated with estrogen-bazedoxifene for a minimum of 12 weeks in a double-blind placebo-controlled study also showed a significant improvement in sexual function and quality-of-life measurements based on 3 well-defined scales, which included ease of lubrication, satisfaction with treatment, control of hot flashes, and sleep parameters.43
Low rates of side effects
To evaluate this regimen’s antagonistic effects on uterine tissue, endometrial hyperplasia was diagnosed by blinded pathologists using endometrial biopsies taken at 6, 12, and 24 months or more if cancer was a suspected diagnosis. At 12 and 24 months of treatment, the incidence of hyperplasia with bazedoxifene 20 or 40 mg at doses of either 0.45 or 0.625 mg of conjugated estrogens was less than 1%, which was similar to placebo rates over the 24 months.44 The lowest dose studied, bazedoxifene 10 mg, did not prevent hyperplasia with conjugated estrogens 0.45 or 0.625 mg, and its use was discontinued.
Rates of amenorrhea with bazedoxifene 20 or 40 mg and conjugated estrogens 0.45 or 0.625 mg were very favorable (83%–93%) and similar to those with placebo.45 For women with continued bleeding on hormone therapy requiring multiple evaluations, or for women who won’t accept the risk of bleeding on hormone therapy, conjugated estrogens and bazedoxifene may be a sustainable option. However, any woman with abnormal bleeding should undergo prompt immediate evaluation.
A typical side effect of estrogen replacement therapy is breast tenderness. For women seeking vasomotor symptom treatment but who experience breast tenderness, this may be a deterrent from continuing hormone therapy. As shown in the SMART-1 and SMART-2 trials,46 conjugated estrogens and bazedoxifene did not cause an increase in breast tenderness, which may enhance medication adherence.
Ideal candidates for conjugated estrogens plus bazedoxifene
This product could help postmenopausal women who have an intact uterus and are suffering with moderate to severe vasomotor symptoms and genitourinary syndrome of menopause who cannot tolerate the side effects of hormone therapy such as bleeding, bloating, or breast tenderness, or who prefer to take an estrogen but without a progestin. It is also ideal for women at higher risk of osteoporosis.
WHO SHOULD GET WHAT?
Not all postmenopausal women have vasomotor symptoms, genitourinary syndrome of menopause, or bone loss. For those who do, standard hormone therapy is an option.
For those who have symptoms and a lower threshold of side effects such as breast tenderness and vaginal bleeding, a combination of an estrogen plus an ERAA (eg, bazedoxifene) is an option.
For women who have no vasomotor symptoms but do have genitourinary syndrome of menopause and don’t want local vaginal treatment, ospemifene is an option.
For women with no vasomotor symptoms but who have bone loss and increased risk of estrogen receptor-positive breast cancer, raloxifene is a good option.
Both premenopausal and postmenopausal women who are at increased risk for breast cancer should be considered for tamoxifen chemoprevention. Postmenopausal women with a uterus at increased risk for breast cancer should be considered for raloxifene, as it has no uterine effect. Raloxifene is not indicated in premenopausal women.
No woman at increased risk of venous thromboembolism is a candidate for ERAA treatment or for oral estrogen. However, the clinician has multiple options to improve quality of life and work productivity and reduce office visits of women at midlife, especially when they are individually assessed and treated.
Estrogen receptor agonist-antagonists (ERAAs), previously called selective estrogen receptor modulators (SERMs), have extended the options for treating the various conditions that menopausal women suffer from. These drugs act differently on estrogen receptors in different tissues, stimulating receptors in some tissues but inhibiting them in others. This allows selective inhibition or stimulation of estrogen-like action in various target tissues.1
This article highlights the use of ERAAs to treat menopausal vasomotor symptoms (eg, hot flashes, night sweats), genitourinary syndrome of menopause, osteoporosis, breast cancer (and the risk of breast cancer), and other health concerns unique to women at midlife.
SYMPTOMS OF MENOPAUSE: COMMON AND TROUBLESOME
Vasomotor symptoms such as hot flashes and night sweats are common during perimenopause—most women experience them. They are most frequent during the menopause transition but can persist for 10 years or more afterward.2
Genitourinary syndrome of menopause is also common and often worsens with years after menopause.3 It can lead to dyspareunia and vaginal dryness, which may in turn result in lower libido, vaginismus, and hypoactive sexual desire disorder, problems that often arise at the same time as vaginal dryness and atrophy.4
Osteopenia and osteoporosis. A drop in systemic estrogen leads to a decline in bone mineral density, increasing the risk of fractures.5
ESTROGEN-PROGESTIN TREATMENT: THE GOLD STANDARD, BUT NOT IDEAL
The current gold standard for treating moderate to severe hot flashes is estrogen, available in oral, transdermal, and vaginal formulations.6 Estrogen also has antiresorptive effects on bone and is approved for preventing osteoporosis. Systemic estrogen may also be prescribed for genitourinary syndrome of menopause if local vaginal treatment alone is insufficient.
If women who have an intact uterus receive estrogen, they should also receive a progestin to protect against endometrial hyperplasia and reduce the risk of endometrial cancer.
Despite its status as the gold standard, estrogen-progestin therapy presents challenges. In some women, progestins cause side effects such as breast tenderness, bloating, fatigue, and depression.7 Estrogen-progestin therapy often causes vaginal bleeding, which for some women is troublesome or distressing; bleeding may be the reason for repeated evaluations, can increase anxiety, and can lead to poor adherence with hormonal treatment. Women who carry a higher-than-normal risk of developing breast cancer or fear that taking hormones will lead to breast cancer may show decreased adherence to therapy. Women who have estrogen receptor-positive breast cancer cannot take estrogen.
Individualized options are needed for women who have progestin-related side effects, unwanted vaginal bleeding, or a higher risk of breast cancer.
WELCOME THE ERAAs
An ideal treatment for menopause would relieve vasomotor symptoms and genitourinary syndrome of menopause and increase bone mineral density without causing breast tenderness, vaginal bleeding, or endometrial proliferation.
The “designer estrogens,” or ERAAs, have specific positive effects on the bone, heart, and brain with neutral or antagonist effects on estrogen receptors in other tissues such as the breasts and endometrium.8 While not entirely free of adverse effects, these agents have been developed with the aim of minimizing the most common ones related to estrogen and progestin.
Several ERAAs are currently approved by the US Food and Drug Administration (FDA)for various indications, each having a unique profile. Clomifene was the first agent of this class, and it is still used clinically to induce ovulation. This article highlights subsequently approved agents, ie, tamoxifen, raloxifene, ospemifene, and the combination of conjugated estrogens and bazedoxifene (Table 1).
All ERAAs increase the risk of venous thromboembolism, and therefore none of them should be used in women with known venous thromboembolism or at high risk of it.
TAMOXIFEN: CANCER TREATMENT AND PREVENTION
After clomiphene, tamoxifen was the second ERAA on the market. Although researchers were looking for a new contraceptive drug, they found tamoxifen to be useful as a chemotherapeutic agent for breast cancer. First used in 1971, tamoxifen continues to be one of the most commonly prescribed chemotherapeutic medications today.
The FDA has approved tamoxifen to treat breast cancer as well as to prevent breast cancer in pre- and postmenopausal women at risk. It may also have beneficial effects on bone and on cardiovascular risk factors, but these are not approved uses for it.
Trials of tamoxifen for cancer treatment
The Early Breast Cancer Trialists’ Collaborative Group9 performed a meta-analysis and found that 5 years of adjuvant treatment with tamoxifen is associated with a 26% reduction in mortality and a 47% reduction in breast cancer recurrence at 10 years. In absolute terms, we estimate that 21 women would need to be treated to prevent 1 death and 8 would need to be treated to prevent 1 recurrence.
The ATLAS Trial (Adjuvant Tamoxifen Longer Against Shorter)10 and later the UK ATTOM (Adjuvant Tamoxifen Treatment to Offer More)11 trial confirmed an even greater reduction in recurrence and mortality after a total of 10 years of treatment.
Trials of tamoxifen for cancer prevention
Cuzik et al12 performed a meta-analysis of 4 trials of tamoxifen’s effectiveness in preventing breast cancer for women at elevated risk. The incidence of estrogen receptor-positive breast cancer was 48% lower with tamoxifen use, but there was no effect on estrogen-negative breast cancer. From their data, we estimate that 77 women would need to be treated to prevent 1 case of breast cancer.
The IBIS-I trial (International Breast Cancer Intervention Study I)13 found that, in healthy women at high risk of breast cancer, the benefit of taking tamoxifen for 5 years as preventive treatment persisted long afterward. The investigators estimated that at 20 years of follow-up the risk of breast cancer would be 12.3% in placebo recipients and 7.8% in tamoxifen recipients, a 4.5% absolute risk reduction; number needed to treat (NNT) 22.
Data on tamoxifen and osteoporosis
The Breast Cancer Prevention Trial revealed a 19% reduction in the incidence of osteoporotic fractures with tamoxifen, but the difference was not statistically significant.14 The 1-year rates of fracture in women age 50 and older were 0.727% with placebo and 0.567% with tamoxifen, an absolute difference of 0.151%; therefore, if the effect is real, 662 women age 50 or older would need to be treated for 1 year to prevent 1 fracture. Tamoxifen is not FDA-approved to treat osteoporosis.
Data on tamoxifen and cardiovascular risk reduction
Chang et al,15 in a study in women at risk of breast cancer, incidentally found that tamoxifen was associated with a 13% reduction in total cholesterol compared with placebo.
Herrington and Klein,16 in a systematic review, noted similar findings in multiple studies of tamoxifen, with decreases in total cholesterol ranging from 7% to 17% and decreases in low-density lipoprotein cholesterol ranging from 10% to 28%. However, they found no change in high-density lipoprotein cholesterol concentrations or in the cardiovascular mortality rate.
The ATLAS trial10 revealed a relative risk reduction of 0.76 (95% confidence interval [CI] 0.60–0.95, P = .02) in ischemic heart disease for women who took tamoxifen for 10 years compared with 5 years. We calculate that ischemic heart disease occurred in 163 (2.5%) of 6,440 women who took tamoxifen for 5 years compared with 127 (1.9%) of 6,454 women who took it for 10 years, a 0.6% absolute risk reduction, NNT = 167.
Adverse effects of tamoxifen
Uterine neoplasia. Women taking tamoxifen have a 2.5-fold increased risk of endometrial cancer.14 Tamoxifen also increases the risk of benign uterine disease such as endometrial hyperplasia and polyps. As many as 39% of women taking tamoxifen will have evidence of benign uterine changes on pathology.17 Other adverse effects:
Venous thromboembolism (the risk of pulmonary embolism is increased approximately threefold14)
Cataracts (there is a slight increase in cataract diagnosis in tamoxifen users)
Vasomotor symptoms, which limit the use of tamoxifen in many women.
Ideal candidate for tamoxifen
The ideal candidate for tamoxifen is a woman with breast cancer that is estrogen receptor-positive and who has a history of osteopenia or osteoporosis and no risk factors for venous thromboembolism.
RALOXIFENE: FOR OSTEOPOROSIS AND FOR CANCER PREVENTION
Raloxifene, a second-generation ERAA, was first approved for preventing and treating osteoporosis and later for reducing the risk of invasive estrogen receptor-positive breast cancer in postmenopausal women.
Trials of raloxifene for osteoporosis
The MORE trial (Multiple Outcomes of Raloxifene)18 was a large multicenter randomized double-blind study. Raloxifene recipients showed a significant increase in bone mineral density in the lumbar spine and femoral neck at year 3 (P < .001) compared with those receiving placebo. Even after only 1 year of treatment, raloxifene significantly reduced the risk of new fractures, despite only modest gains in bone mineral density. After 3 years of treatment, new clinical vertebral fractures had occurred in 3.5% of the placebo group compared with 2.1% of the group receiving raloxifene 60 mg.19 Relative risk reductions were similar in women who had already had a clinical vertebral fracture at baseline, whose absolute risk is higher. However, no significant effect was seen on the incidence of hip or nonvertebral fractures.
The CORE trial (Continuing Outcomes Relevant to Raloxifene)20 extended the treatment of the women enrolled in the MORE trial another 4 years and found that the benefit of raloxifene with regard to bone mineral density persisted with continued use.
Trials of raloxifene for breast cancer prevention
The MORE trial,21 in postmenopausal women with osteoporosis included breast cancer as a secondary end point, and raloxifene was shown to decrease the incidence of invasive breast cancer. At a median of 40 months, invasive breast cancer had arisen in 13 (0.25%) of the 5,129 women assigned to raloxifene and 27 (1.0%) of the 2,576 women assigned to placebo. The authors calculated that 126 women would need to be treated to prevent 1 case of breast cancer.
The CORE trial,22 as noted, extended the treatment of the women enrolled in the MORE trial another 4 years. The risk of any invasive breast cancer in postmenopausal women with osteoporosis was significantly reduced by 59% after 8 years, and the risk of estrogen receptor-positive invasive breast cancer was reduced by 66%.
There is evidence that raloxifene’s effect on breast cancer risk persists after discontinuation of use.23
Does raloxifene reduce mortality?
Grady et al24 studied the effect of raloxifene on all-cause mortality in a pooled analysis of mortality data from the MORE, CORE, and Raloxifene Use for the Heart (RUTH)25 trials. In older postmenopausal women, the rate of all-cause mortality was 8.65% in those taking placebo compared with 7.88% in those taking raloxifene 60 mg daily—10% lower. The mechanism behind the lower mortality rate is unclear, and Grady et al recommend that the finding be interpreted with caution.
Trials of raloxifene for heart protection
The RUTH trial25 was a 5.6-year study undertaken to study the effects of raloxifene on coronary outcomes and invasive breast cancer in postmenopausal women. Results were mixed. Active treatment:
- Did not significantly affect the risk of coronary artery disease compared with placebo
- Significantly decreased the risk of invasive breast cancer
- Significantly decreased the risk of clinical vertebral fractures
- Increased the risk of fatal stroke (59 vs 39 events, hazard ratio 1.49, 95% CI 1.00–2.24) and venous thromboembolism (103 vs 71 events, hazard ratio 1.44, 95% CI 1.06–1.95).
The STAR trial (Study of Tamoxifen and Raloxifene)26,27 compared raloxifene and tamoxifen in postmenopausal women at increased risk of breast cancer. Women were randomized to receive either tamoxifen 20 mg or raloxifene 60 mg for 5 years. Results:
- No difference in the number of new cases of invasive breast cancer between the groups
- Fewer cases of noninvasive breast cancer in the tamoxifen group, but the difference was not statistically significant
- Fewer cases of uterine cancer in the raloxifene group, annual incidence rates 0.125% vs 0.199%, absolute risk reduction 0.74%, NNT 1,351, relative risk with raloxifene 0.62, 95% CI 0.30–0.50
- Fewer thromboembolic events with raloxifene
- Fewer cataracts with raloxifene.
Adverse effects of raloxifene
Raloxifene increases the risk of venous thromboembolism and stroke in women at high risk of coronary artery disease.19
Ideal candidates for raloxifene
Postmenopausal women with osteopenia or osteoporosis and a higher risk of breast cancer who have minimal to no vasomotor symptoms or genitourinary syndrome of menopause are good candidates for raloxifene. Raloxifene is also a good choice for women who have genitourinary syndrome of menopause treated with local vaginal estrogen. Raloxifene has no effect on vasomotor symptoms or genitourinary syndrome of menopause.
OSPEMIFENE: FOR GENITOURINARY SYNDROME OF MENOPAUSE
Although ospemifene does not have the steroid structure of estrogen, it acts as an estrogen agonist specifically in the vaginal mucosa and an antagonist in other tissues.28 It has been shown on Papanicolaou smears to reduce the number of parabasal cells and increase the number of intermediate and superficial cells after 3 months of treatment.29
Ospemifene 60 mg taken orally with food is approved by the FDA to treat genitourinary syndrome of menopause.
Why ospemifene is needed
First-line treatment options for genitourinary syndrome of menopause include over-the-counter lubricants. However, there is no evidence that these products reverse vaginal atrophy,30 and many women report no relief of symptoms with them.
While various local estrogen preparations positively affect genitourinary syndrome of menopause, some of them can be messy, which can limit-long term adherence.
In one of the largest surveys on genitourinary syndrome of menopause (the REVIVE survey—the Real Women’s View of Treatment Options for Menopausal Vaginal Changes29), 59% of women reported that their vaginal symptoms negatively affected sexual activity. The problem affects not only the patient but also her sexual partner.31 Another large study showed that 38% of women and 39% of male partners reported that it had a worse-than-expected impact on their intimate relationships.31
Genitourinary syndrome of menopause also makes pelvic examinations difficult, may worsen or exacerbate cystitis, and may increase urinary tract infections.
Trials of ospemifene for genitourinary syndrome of menopause
To date, 3 randomized, double-blind clinical trials have demonstrated ospemifene 60 mg to be superior to placebo in treating genitourinary syndrome of menopause. Two were short-term (12-week) and showed significant positive changes in the percent of superficial cells, vaginal pH (lower is better), and number of parabasal cells, along with improvements in the Likert rating of both vaginal dryness and dyspareunia.32,33
A long-term (52-week) randomized placebo-controlled trial compared ospemifene and placebo and showed significant improvement in vaginal maturation index and pH at weeks 12 and 52.34 Other outcome measures included petechiae, pallor, friability, erythema, and dryness, all of which improved from baseline (P < .001). At the end of the trial, 80% of the patients who received ospemifene had no vaginal atrophy.
No serious adverse events were noted in any of the clinical trials to date, and a systemic review and meta-analysis demonstrated ospemifene to be safe and efficacious.35 The most frequently reported reasons for discontinuation were hot flashes, vaginal discharge, muscle spasms, and hyperhidrosis, but the rates of these effects were similar to those with placebo.
Trial of ospemifene’s effect on bone turnover
As an estrogen receptor agonist in bone, ospemifene decreases the levels of bone turnover markers in postmenopausal women.36 A study found ospemifene to be about as effective as raloxifene in suppressing bone turnover,37 but ospemifene does not carry FDA approval for preventing or treating osteoporosis.
Other effects
In experiments in rats, the incidence of breast cancer appears to be lower with ospemifene, and the higher the dose, the lower the incidence.38
Ospemifene also has antagonistic effects on uterine tissue, and no cases of endometrial hyperplasia or carcinoma have been reported in short-term or long-term studies.35
Ospemifene has no effect however on vasomotor symptoms and may in fact worsen vasomotor symptoms in women suffering with hot flashes and night sweats. Further investigation into its long-term safety and effects on breast tissue and bone would provide more insight.
Ideal candidates for ospemifene
Ospemifene could help postmenopausal women with genitourinary syndrome of menopause for whom over-the-counter lubricants fail, who dislike local vaginal estrogen, or who decline systemic hormone therapy, and who do not meet the criteria for treatment with systemic hormone therapy.
CONJUGATED ESTROGENS AND BAZEDOXIFENE COMBINATION
A combination agent consisting of conjugated estrogens 0.45 mg plus bazedoxifene 20 mg has been approved by the FDA for treating moderate to severe vasomotor symptoms associated with menopause and also for preventing postmenopausal osteoporosis in women who have an intact uterus.
Trials of estrogen-bazedoxifene for vasomotor symptoms
The Selective Estrogen Menopause and Response to Therapy (SMART) trials39,40 were a series of randomized, double-blind, placebo-controlled phase 3 studies evaluating the efficacy and safety of the estrogen-bazedoxifene combination in postmenopausal women.
The SMART-2 trial39 evaluated the combination of conjugated estrogens (either 0.45 mg or 0.625) plus bazedoxifene 20 mg and found both dosages significantly reduced the number and severity of hot flashes at weeks 4 and 12 (P < .001). At week 12, the combination with 0.45 mg of estrogen reduced vasomotor symptoms from baseline by 74% (10.3 hot flashes per week at baseline vs 2.8 at week 12); the combination with 0.625 mg of estrogen reduced vasomotor symptoms by 80% (10.4 vs 2.4 flashes); and placebo reduced them by 51% (10.5 vs 5.4 flashes).
For bone density. The SMART-1 trial40 showed that the estrogen-bazedoxifene combination in both estrogen dosages significantly increased mean lumbar spine bone mineral density (P < .001) and total hip bone mineral density (P < .05) from baseline at 12 and 24 months compared with placebo. Increases in density tended to be higher with the higher estrogen dose (0.625 mg), but less with higher doses of bazedoxifene.41 At 24 months, the increase in bone mineral density was even greater than in women treated with raloxifene.42 However, the effect of estrogen-bazedoxifene on the incidence of fractures remains to be studied.
For genitourinary syndrome of menopause. The SMART-3 trial showed that treatment with conjugated estrogens plus bazedoxifene (0.45/20 mg or 0.625/20 mg) was more effective than placebo in increasing the percent of superficial and intermediate cells and decreased the number of parabasal cells at 12 weeks compared with placebo (P < .01).43 Both doses also significantly decreased the mean vaginal pH and improved vaginal dryness.
Patients treated with estrogen-bazedoxifene for a minimum of 12 weeks in a double-blind placebo-controlled study also showed a significant improvement in sexual function and quality-of-life measurements based on 3 well-defined scales, which included ease of lubrication, satisfaction with treatment, control of hot flashes, and sleep parameters.43
Low rates of side effects
To evaluate this regimen’s antagonistic effects on uterine tissue, endometrial hyperplasia was diagnosed by blinded pathologists using endometrial biopsies taken at 6, 12, and 24 months or more if cancer was a suspected diagnosis. At 12 and 24 months of treatment, the incidence of hyperplasia with bazedoxifene 20 or 40 mg at doses of either 0.45 or 0.625 mg of conjugated estrogens was less than 1%, which was similar to placebo rates over the 24 months.44 The lowest dose studied, bazedoxifene 10 mg, did not prevent hyperplasia with conjugated estrogens 0.45 or 0.625 mg, and its use was discontinued.
Rates of amenorrhea with bazedoxifene 20 or 40 mg and conjugated estrogens 0.45 or 0.625 mg were very favorable (83%–93%) and similar to those with placebo.45 For women with continued bleeding on hormone therapy requiring multiple evaluations, or for women who won’t accept the risk of bleeding on hormone therapy, conjugated estrogens and bazedoxifene may be a sustainable option. However, any woman with abnormal bleeding should undergo prompt immediate evaluation.
A typical side effect of estrogen replacement therapy is breast tenderness. For women seeking vasomotor symptom treatment but who experience breast tenderness, this may be a deterrent from continuing hormone therapy. As shown in the SMART-1 and SMART-2 trials,46 conjugated estrogens and bazedoxifene did not cause an increase in breast tenderness, which may enhance medication adherence.
Ideal candidates for conjugated estrogens plus bazedoxifene
This product could help postmenopausal women who have an intact uterus and are suffering with moderate to severe vasomotor symptoms and genitourinary syndrome of menopause who cannot tolerate the side effects of hormone therapy such as bleeding, bloating, or breast tenderness, or who prefer to take an estrogen but without a progestin. It is also ideal for women at higher risk of osteoporosis.
WHO SHOULD GET WHAT?
Not all postmenopausal women have vasomotor symptoms, genitourinary syndrome of menopause, or bone loss. For those who do, standard hormone therapy is an option.
For those who have symptoms and a lower threshold of side effects such as breast tenderness and vaginal bleeding, a combination of an estrogen plus an ERAA (eg, bazedoxifene) is an option.
For women who have no vasomotor symptoms but do have genitourinary syndrome of menopause and don’t want local vaginal treatment, ospemifene is an option.
For women with no vasomotor symptoms but who have bone loss and increased risk of estrogen receptor-positive breast cancer, raloxifene is a good option.
Both premenopausal and postmenopausal women who are at increased risk for breast cancer should be considered for tamoxifen chemoprevention. Postmenopausal women with a uterus at increased risk for breast cancer should be considered for raloxifene, as it has no uterine effect. Raloxifene is not indicated in premenopausal women.
No woman at increased risk of venous thromboembolism is a candidate for ERAA treatment or for oral estrogen. However, the clinician has multiple options to improve quality of life and work productivity and reduce office visits of women at midlife, especially when they are individually assessed and treated.
- Giannini A, Russo E, Mannella P, Simoncini T. Selective steroid receptor modulators in reproductive medicine. Minerva Ginecol 2015; 67:431–455.
- Feldman BM, Voda A, Gronseth E. The prevalence of hot flash and associated variables among perimenopausal women. Res Nurs Health 1985; 8:261–268.
- Versi E, Harvey MA, Cardozo L, Brincat M, Studd JW. Urogenital prolapse and atrophy at menopause: a prevalence study. Int Urogynecol J Pelvic Floor Dysfunct 2001; 12:107–110.
- Hess R, Chang CC, Conigliaro J, McNeil M. Understanding physicians’ attitudes towards hormone therapy. Womens Health Issues 2005; 15:31–38.
- Melton LJ 3rd, Khosla S, Atkinson EJ, O’Fallon WM, Riggs BL. Relationship of bone turnover to bone density and fractures. J Bone Miner Res 1997; 12:1083–1091.
- Sikon A, Thacker HL. Treatment options for menopausal hot flashes. Cleve Clin J Med 2004; 71:578–582.
- Levine JP. Treating menopausal symptoms with a tissue-selective estrogen complex. Gend Med 2011; 8:57–68.
- Pinkerton JV, Thomas S. Use of SERMs for treatment in postmenopausal women. J Steroid Biochem Mol Biol 2014; 142:142–154.
- Tamoxifen for early breast cancer: an overview of the randomised trials. Early Breast Cancer Trialists’ Collaborative Group. Lancet 1998; 351:1451–1467.
- Davies C, Pan H, Godwin J, et al; Adjuvant Tamoxifen: Longer Against Shorter (ATLAS) Collaborative Group. Long-term effects of continuing adjuvant tamoxifen to 10 years versus stopping at 5 years after diagnosis of oestrogen receptor-positive breast cancer: ATLAS, a randomised trial. Lancet 2013; 381:805–816.
- Gray RG, Rea D, Handley K, et al. aTTom: Long-term effects of continuing adjuvant tamoxifen to 10 years versus stopping at 5 years in 6,953 women with early breast cancer. J Clin Oncol 2013; (suppl): abstract 5.
- Cuzick J, Powles T, Veronesi U, et al. Overview of the main outcomes in breast-cancer prevention trials. Lancet 2003; 361:296–300.
- Cuzick J, Sestak I, Cawthorn S, et al. Tamoxifen for prevention of breast cancer: extended long-term follow-up of the IBIS-I breast cancer prevention trial. Lancet Oncol 2015; 16:67–75.
- Fisher B, Costantino JP, Wickerham DL, et al. Tamoxifen for prevention of breast cancer: report of the National Surgical Adjuvant Breast and Bowel Project P-1 Study. J Natl Cancer Inst 1998; 90:1371–1388.
- Chang J, Powles TJ, Ashley SE, et al. The effect of tamoxifen and hormone replacement therapy on serum cholesterol, bone mineral density and coagulation factors in healthy postmenopausal women participating in a randomised, controlled tamoxifen prevention study. Ann Oncol 1996; 7:671–675.
- Herrington DM, Klein KP. Effects of SERMs on important indicators of cardiovascular health: lipoproteins, hemostatic factors and endothelial function. Womens Health Issues 2001; 11:95–102.
- Kedar RP, Bourne TH, Powles TJ, et al. Effects of tamoxifen on uterus and ovaries of postmenopausal women in a randomized breast cancer prevention trial. Lancet 1994; 343:1318–1321.
- Ettinger B, Black DM, Mitlak BH, et al. Reduction of vertebral fracture risk in postmenopausal women with osteoporosis treated with raloxifene: results from a 3-year randomized clinical trial. Multiple Outcomes of Raloxifene Evaluation (MORE) Investigators. JAMA 1999; 282:637–645.
- Maricic M, Adachi JD, Sarkar S, Wu W, Wong M, Harper KD. Early effects of raloxifene on clinical vertebral fractures at 12 months in postmenopausal women with osteoporosis. Arch Intern Med 2002; 162:1140–1143.
- Recker RR, Mitlak BH, Ni X, Krege JH. Long-term raloxifene for postmenopausal osteoporosis. Curr Med Res Opin 2011; 27:1755–1761.
- Cummings SR, Eckert S, Krueger KA, et al. The effect of raloxifene on risk of breast cancer in postmenopausal women: results from the MORE randomized trial. Multiple Outcomes of Raloxifene Evaluation. JAMA 1999; 281:2189–2197.
- Martino S, Cauley JA, Barrett-Connor E, et al; CORE Investigators. Continuing outcomes relevant to Evista: breast cancer incidence in postmenopausal osteoporotic women in a randomized trial of raloxifene. J Natl Cancer Inst 2004; 96:1751–1761.
- Vogel VG, Qu Y, Wong M, Mitchell B, Mershon JL. Incidence of invasive breast cancer in postmenopausal women after discontinuation of long-term raloxifene administration. Clin Breast Cancer 2009; 9:45–50.
- Grady D, Cauley JA, Stock JL, et al. Effect of raloxifene on all-cause mortality. Am J Med 2010; 123:469.e1–e7.
- Barrett-Connor E, Mosca L, Collins P, et al; Raloxifene Use for The Heart (RUTH) Trial Investigators. Effects of raloxifene on cardiovascular events and breast cancer in postmenopausal women. N Engl J Med 2006; 355:125–137.
- Vogel VG. The NSABP Study of Tamoxifen and Raloxifene (STAR) trial. Expert Rev Anticancer Ther 2009; 9:51–60.
- Vogel VG, Costantino JP, Wickerham DL, et al; National Surgical Adjuvant Breast and Bowel Project (NSABP). Effects of tamoxifen vs raloxifene on the risk of developing invasive breast cancer and other disease outcomes: the NSABP Study of Tamoxifen and Raloxifene (STAR) P-2 trial. JAMA 2006; 295:2727–2741.
- Barnes KN, Pearce EF, Yancey AM, Forinash AB. Ospemifene in the treatment of vulvovaginal atrophy. Ann Pharmacother 2014; 48:752–757.
- Rutanen EM, Heikkinen J, Halonen K, Komi J, Lammintausta R, Ylikorkala O. Effects of ospemifene, a novel SERM, on hormones, genital tract, climacteric symptoms, and quality of life in postmenopausal women: a double-blind, randomized trial. Menopause 2003; 10:433–439.
- Constantine G, Graham S, Koltun WD, Kingsberg SA. Assessment of ospemifene or lubricants on clinical signs of VVA. J Sex Med 2014; 11:1033–1041.
- Kingsberg SA, Wysocki S, Magnus L, Krychman ML. Vulvar and vaginal atrophy in postmenopausal women: findings from the REVIVE survey. J Sex Med 2013; 10:1790–1799.
- Portman DJ, Bachmann GA, Simon JA; Ospemifene Study Group. Ospemifene, a novel selective estrogen receptor modulator for treating dyspareunia associated with postmenopausal vulvar and vaginal atrophy. Menopause 2013; 20:623–630.
- Bachmann GA, Komi JO; Ospemifene Study Group. Ospemifene effectively treats vulvovaginal atrophy in postmenopausal women: results from a pivotal phase 3 study. Menopause 2010; 17:480–486.
- Goldstein SR, Bachmann GA, Koninckx PR, Lin VH, Portman DJ, Ylikorkala O; Ospemifene Study Group. Ospemifene 12-month safety and efficacy in postmenopausal women with vulvar and vaginal atrophy. Climacteric 2014; 17:173–182.
- Cui Y, Zong H, Yan H, Li N, Zhang Y. The efficacy and safety of ospemifene in treating dyspareunia associated with postmenopausal vulvar and vaginal atrophy: a systematic review and meta-analysis. J Sex Med 2014; 11:487–497.
- Komi J, Heikkinen J, Rutanen EM, Halonen K, Lammintausta R, Ylikorkala O. Effects of ospemifene, a novel SERM, on biochemical markers of bone turnover in healthy postmenopausal women. Gynecal Endocrinol 2004; 18:152–158.
- Komi J, Lankinen KS, DeGregorio M, et al. Effects of ospemifene and raloxifene on biochemical markers of bone turnover in postmenopausal women. J Bone Miner Metab 2006; 24:314–318.
- 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; 97:230–240.
- 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; 16:1116–1124.
- Pickar JH, Mirkin S. Tissue-selective agents: selective estrogen receptor modulators and the tissue-selective estrogen complex. Menopause Int 2010; 16:121–128.
- Levine JP. Treating menopausal symptoms with a tissue-selective estrogen complex. Gend Med 2011; 8:57–68.
- Lindsay R, Gallagher JC, Kagan R, Pickar JH, Constantine G. Efficacy of tissue-selective estrange complex of bazedoxifene/conjugated estrogens for osteoporosis prevention in at-risk postmenopausal women. Fertil Steril 2009; 92:1045–1052.
- Bachmann G, Bobula J, Mirkin S. Effects of bazedoxifene/conjugated estrogens on quality of life in postmenopausal women with symptoms of vulvar/vaginal atrophy. Climacteric 2010; 13:132–140.
- Pickar JH, Yeh IT, Bachmann G, Speroff L. Endometrial effects of a tissue selective estrogen complex containing bazedoxifene/conjugated estrogens as a menopausal therapy. Fertil Steril 2009; 92:1018–1024.
- Archer DF, Lewis V, Carr BR, Olivier S, Pickar JH. Bazedoxifene/conjugated estrogens (BZA/CE): incidence of uterine bleeding in postmenopausal women. Fertil Steril 2009: 92:1039–1044.
- Pinkerton JV, Abraham L, Bushmakin AG, et al. Evaluation of the efficacy and safety of bazedoxifene/conjugated estrogens for secondary outcomes including vasomotor symptoms in postmenopausal women by years since menopause in the Selective estrogens, Menopause and Response to Therapy (SMART) trials. J Womens Health (Larchmt) 2014; 23:18–28.
- Giannini A, Russo E, Mannella P, Simoncini T. Selective steroid receptor modulators in reproductive medicine. Minerva Ginecol 2015; 67:431–455.
- Feldman BM, Voda A, Gronseth E. The prevalence of hot flash and associated variables among perimenopausal women. Res Nurs Health 1985; 8:261–268.
- Versi E, Harvey MA, Cardozo L, Brincat M, Studd JW. Urogenital prolapse and atrophy at menopause: a prevalence study. Int Urogynecol J Pelvic Floor Dysfunct 2001; 12:107–110.
- Hess R, Chang CC, Conigliaro J, McNeil M. Understanding physicians’ attitudes towards hormone therapy. Womens Health Issues 2005; 15:31–38.
- Melton LJ 3rd, Khosla S, Atkinson EJ, O’Fallon WM, Riggs BL. Relationship of bone turnover to bone density and fractures. J Bone Miner Res 1997; 12:1083–1091.
- Sikon A, Thacker HL. Treatment options for menopausal hot flashes. Cleve Clin J Med 2004; 71:578–582.
- Levine JP. Treating menopausal symptoms with a tissue-selective estrogen complex. Gend Med 2011; 8:57–68.
- Pinkerton JV, Thomas S. Use of SERMs for treatment in postmenopausal women. J Steroid Biochem Mol Biol 2014; 142:142–154.
- Tamoxifen for early breast cancer: an overview of the randomised trials. Early Breast Cancer Trialists’ Collaborative Group. Lancet 1998; 351:1451–1467.
- Davies C, Pan H, Godwin J, et al; Adjuvant Tamoxifen: Longer Against Shorter (ATLAS) Collaborative Group. Long-term effects of continuing adjuvant tamoxifen to 10 years versus stopping at 5 years after diagnosis of oestrogen receptor-positive breast cancer: ATLAS, a randomised trial. Lancet 2013; 381:805–816.
- Gray RG, Rea D, Handley K, et al. aTTom: Long-term effects of continuing adjuvant tamoxifen to 10 years versus stopping at 5 years in 6,953 women with early breast cancer. J Clin Oncol 2013; (suppl): abstract 5.
- Cuzick J, Powles T, Veronesi U, et al. Overview of the main outcomes in breast-cancer prevention trials. Lancet 2003; 361:296–300.
- Cuzick J, Sestak I, Cawthorn S, et al. Tamoxifen for prevention of breast cancer: extended long-term follow-up of the IBIS-I breast cancer prevention trial. Lancet Oncol 2015; 16:67–75.
- Fisher B, Costantino JP, Wickerham DL, et al. Tamoxifen for prevention of breast cancer: report of the National Surgical Adjuvant Breast and Bowel Project P-1 Study. J Natl Cancer Inst 1998; 90:1371–1388.
- Chang J, Powles TJ, Ashley SE, et al. The effect of tamoxifen and hormone replacement therapy on serum cholesterol, bone mineral density and coagulation factors in healthy postmenopausal women participating in a randomised, controlled tamoxifen prevention study. Ann Oncol 1996; 7:671–675.
- Herrington DM, Klein KP. Effects of SERMs on important indicators of cardiovascular health: lipoproteins, hemostatic factors and endothelial function. Womens Health Issues 2001; 11:95–102.
- Kedar RP, Bourne TH, Powles TJ, et al. Effects of tamoxifen on uterus and ovaries of postmenopausal women in a randomized breast cancer prevention trial. Lancet 1994; 343:1318–1321.
- Ettinger B, Black DM, Mitlak BH, et al. Reduction of vertebral fracture risk in postmenopausal women with osteoporosis treated with raloxifene: results from a 3-year randomized clinical trial. Multiple Outcomes of Raloxifene Evaluation (MORE) Investigators. JAMA 1999; 282:637–645.
- Maricic M, Adachi JD, Sarkar S, Wu W, Wong M, Harper KD. Early effects of raloxifene on clinical vertebral fractures at 12 months in postmenopausal women with osteoporosis. Arch Intern Med 2002; 162:1140–1143.
- Recker RR, Mitlak BH, Ni X, Krege JH. Long-term raloxifene for postmenopausal osteoporosis. Curr Med Res Opin 2011; 27:1755–1761.
- Cummings SR, Eckert S, Krueger KA, et al. The effect of raloxifene on risk of breast cancer in postmenopausal women: results from the MORE randomized trial. Multiple Outcomes of Raloxifene Evaluation. JAMA 1999; 281:2189–2197.
- Martino S, Cauley JA, Barrett-Connor E, et al; CORE Investigators. Continuing outcomes relevant to Evista: breast cancer incidence in postmenopausal osteoporotic women in a randomized trial of raloxifene. J Natl Cancer Inst 2004; 96:1751–1761.
- Vogel VG, Qu Y, Wong M, Mitchell B, Mershon JL. Incidence of invasive breast cancer in postmenopausal women after discontinuation of long-term raloxifene administration. Clin Breast Cancer 2009; 9:45–50.
- Grady D, Cauley JA, Stock JL, et al. Effect of raloxifene on all-cause mortality. Am J Med 2010; 123:469.e1–e7.
- Barrett-Connor E, Mosca L, Collins P, et al; Raloxifene Use for The Heart (RUTH) Trial Investigators. Effects of raloxifene on cardiovascular events and breast cancer in postmenopausal women. N Engl J Med 2006; 355:125–137.
- Vogel VG. The NSABP Study of Tamoxifen and Raloxifene (STAR) trial. Expert Rev Anticancer Ther 2009; 9:51–60.
- Vogel VG, Costantino JP, Wickerham DL, et al; National Surgical Adjuvant Breast and Bowel Project (NSABP). Effects of tamoxifen vs raloxifene on the risk of developing invasive breast cancer and other disease outcomes: the NSABP Study of Tamoxifen and Raloxifene (STAR) P-2 trial. JAMA 2006; 295:2727–2741.
- Barnes KN, Pearce EF, Yancey AM, Forinash AB. Ospemifene in the treatment of vulvovaginal atrophy. Ann Pharmacother 2014; 48:752–757.
- Rutanen EM, Heikkinen J, Halonen K, Komi J, Lammintausta R, Ylikorkala O. Effects of ospemifene, a novel SERM, on hormones, genital tract, climacteric symptoms, and quality of life in postmenopausal women: a double-blind, randomized trial. Menopause 2003; 10:433–439.
- Constantine G, Graham S, Koltun WD, Kingsberg SA. Assessment of ospemifene or lubricants on clinical signs of VVA. J Sex Med 2014; 11:1033–1041.
- Kingsberg SA, Wysocki S, Magnus L, Krychman ML. Vulvar and vaginal atrophy in postmenopausal women: findings from the REVIVE survey. J Sex Med 2013; 10:1790–1799.
- Portman DJ, Bachmann GA, Simon JA; Ospemifene Study Group. Ospemifene, a novel selective estrogen receptor modulator for treating dyspareunia associated with postmenopausal vulvar and vaginal atrophy. Menopause 2013; 20:623–630.
- Bachmann GA, Komi JO; Ospemifene Study Group. Ospemifene effectively treats vulvovaginal atrophy in postmenopausal women: results from a pivotal phase 3 study. Menopause 2010; 17:480–486.
- Goldstein SR, Bachmann GA, Koninckx PR, Lin VH, Portman DJ, Ylikorkala O; Ospemifene Study Group. Ospemifene 12-month safety and efficacy in postmenopausal women with vulvar and vaginal atrophy. Climacteric 2014; 17:173–182.
- Cui Y, Zong H, Yan H, Li N, Zhang Y. The efficacy and safety of ospemifene in treating dyspareunia associated with postmenopausal vulvar and vaginal atrophy: a systematic review and meta-analysis. J Sex Med 2014; 11:487–497.
- Komi J, Heikkinen J, Rutanen EM, Halonen K, Lammintausta R, Ylikorkala O. Effects of ospemifene, a novel SERM, on biochemical markers of bone turnover in healthy postmenopausal women. Gynecal Endocrinol 2004; 18:152–158.
- Komi J, Lankinen KS, DeGregorio M, et al. Effects of ospemifene and raloxifene on biochemical markers of bone turnover in postmenopausal women. J Bone Miner Metab 2006; 24:314–318.
- 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; 97:230–240.
- 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; 16:1116–1124.
- Pickar JH, Mirkin S. Tissue-selective agents: selective estrogen receptor modulators and the tissue-selective estrogen complex. Menopause Int 2010; 16:121–128.
- Levine JP. Treating menopausal symptoms with a tissue-selective estrogen complex. Gend Med 2011; 8:57–68.
- Lindsay R, Gallagher JC, Kagan R, Pickar JH, Constantine G. Efficacy of tissue-selective estrange complex of bazedoxifene/conjugated estrogens for osteoporosis prevention in at-risk postmenopausal women. Fertil Steril 2009; 92:1045–1052.
- Bachmann G, Bobula J, Mirkin S. Effects of bazedoxifene/conjugated estrogens on quality of life in postmenopausal women with symptoms of vulvar/vaginal atrophy. Climacteric 2010; 13:132–140.
- Pickar JH, Yeh IT, Bachmann G, Speroff L. Endometrial effects of a tissue selective estrogen complex containing bazedoxifene/conjugated estrogens as a menopausal therapy. Fertil Steril 2009; 92:1018–1024.
- Archer DF, Lewis V, Carr BR, Olivier S, Pickar JH. Bazedoxifene/conjugated estrogens (BZA/CE): incidence of uterine bleeding in postmenopausal women. Fertil Steril 2009: 92:1039–1044.
- Pinkerton JV, Abraham L, Bushmakin AG, et al. Evaluation of the efficacy and safety of bazedoxifene/conjugated estrogens for secondary outcomes including vasomotor symptoms in postmenopausal women by years since menopause in the Selective estrogens, Menopause and Response to Therapy (SMART) trials. J Womens Health (Larchmt) 2014; 23:18–28.
KEY POINTS
- Tamoxifen is approved to prevent and treat breast cancer. It may also have beneficial effects on bone and on cardiovascular risk factors, but these are not approved uses.
- Raloxifene, a second-generation ERAA, was initially approved for preventing and treating osteoporosis and later received approval to reduce the risk of invasive estrogen receptor-positive breast cancer in postmenopausal women.
- Ospemifene is approved for treatment of genitourinary syndrome of menopause.
- The combination of conjugated estrogen and bazedoxifene is approved for treating moderate to severe vasomotor symptoms associated with menopause and also for preventing postmenopausal osteoporosis in women with an intact uterus.
Use and misuse of opioid agonists in opioid addiction
For a patient struggling with opioid addiction, opioid agonist therapy with methadone or buprenorphine can reduce craving and opioid use and may even save his or her life. But many clinicians are unfamiliar with this evidence-based treatment,1,2 which is best started early in the course of addiction.3
This article outlines the pharmacology of these drugs, their clinical uses, and the challenges of using them to treat opioid addiction.
DIAGNOSTIC CRITERIA
Opioid addiction, formally known as opioid use disorder, is a pattern of opioid misuse leading to clinically significant impairment in multiple areas of life. The Diagnostic and Statistical Manual of Mental Disorders, Fifth Edition, lists 11 diagnostic criteria, but only 2 need to be present within the past year to make the diagnosis4:
- Taking opioids longer or in higher doses than was intended
- A persistent desire or unsuccessful efforts to cut down or control opioid use
- Spending a great deal of time obtaining, using, or recovering from using opioids
- Craving opioids
- Repeatedly failing to fulfill obligations at work, school, or home due to opioid use
- Continuing to use opioids even though it causes or exacerbates social or interpersonal problems
- Giving up or curtailing important social, occupational, or recreational activities because of opioid use
- Repeatedly using opioids in situations in which it is physically hazardous
- Continuing to use opioids despite knowledge of having a persistent or recurrent physical or psychological problem that is likely to have been caused or exacerbated by the substance
- Tolerance
- Withdrawal.
Recent estimates indicate that 2.23 million people in the United States have opioid use disorder (426,000 with heroin and 1.8 million with prescription opioids).5
Progression from prescription opioids to heroin
We have observed that many patients with opioid use disorder start by misusing prescription opioids. Over time, tolerance can develop, which drives patients to use higher and higher doses.6
As the addiction progresses, a subset of prescription opioid users advances to using heroin, which is typically less expensive and easier to obtain.7 Most patients start with the intranasal route but eventually inject it intravenously.6,7
For many addicts, heroin use has medical consequences such as hepatitis C virus (HCV) and human immunodeficiency virus (HIV) infection, psychiatric problems such as depression and anxiety, and illegal activities such as theft and sex work.8 People who use heroin appear to have more severe addiction and a lower socioeconomic status than prescription opioid users.9–11 But recently, a growing number of middle class individuals are becoming addicted to heroin.12
METHADONE
Methadone is a long-acting synthetic opioid that functions as a full agonist on the mu-opioid receptor. The drug binds, occupies, and stimulates the receptor, preventing withdrawal symptoms and reducing opioid cravings for at least 24 hours.13
Adverse effects of methadone
The most common adverse effects include lightheadedness, dizziness, sedation, nausea, vomiting, and sweating.14 Other adverse effects:
Unintentional overdose. The risk is serious, as a single 30-mg dose can be fatal in people who are opioid-naïve.13
QTc prolongation, which can lead to torsade de pointes. This risk, which is dose-related, must be taken into consideration in patients who have any cardiac symptoms (eg, syncope, arrhythmia), pathology (familial QT prolongation), or other risk factors for QTc prolongation (eg, hypokalemia, QTc-prolonging medications).15
Respiratory depression, which can be fatal. This dose-related risk is heightened during the first 4 weeks of treatment if titration is too rapid or if methadone is used in combination with other drugs that cause central nervous system or respiratory depression.13,14
Starting methadone
To prevent respiratory depression and death related to rapid induction, the general rule is to start methadone at a low daily dose (20–30 mg) depending on the patient’s withdrawal symptoms.14 During this period, patients need to be closely monitored and educated on the perils of concomitant use of central nervous system depressants.14
In most patients, the dose is titrated up until their withdrawal symptoms and cravings are eliminated, which generally requires 60 to 120 mg daily.14 Hepatic and renal impairment, pregnancy, and advanced age can alter methadone pharmacokinetics and may therefore necessitate dose adjustment.
BUPRENORPHINE
Buprenorphine is an alkaloid thebaine opioid derivative that acts as a partial mu-opioid agonist and a kappa antagonist.16 Like methadone, buprenorphine is used to manage cravings and withdrawal symptoms.16 Dosages of 4 to 16 mg (up to 32 mg) per day of buprenorphine are usually required to adequately control opioid cravings.16
Sublingual and subdermal products
Buprenorphine is currently available in the United States in sublingual and subdermal formulations.16,17
Sublingual buprenorphine is usually combined with naloxone in a 4:1 ratio to deter intravenous use. Intravenous injection of the combination product can precipitate withdrawal due to the antagonist action of naloxone. (Taken orally or sublingually, naloxone is poorly absorbed and has little or no clinical effect.) Buprenorphine-naloxone is available in tablets, a sublingual film strip, and a buccal film strip. Buprenorphine is also available by itself in a sublingual formulation.
The US Food and Drug Administration has approved a buprenorphine subdermal implant, Probuphine. Four rods, about 1 inch long, are placed under the skin in the inner aspect of the upper arm and provide the equivalent of 8 mg of buprenorphine daily for 6 months.17 However, this method is formulated only for maintenance treatment and cannot be used for induction. Additionally, it is recommended that the implants be surgically removed at the end of 6 months, after which another set of implants can be inserted in the other arm or the patient can switch to sublingual therapy, depending on the clinical situation and patient preference.17
Generally safer than methadone
Buprenorphine works on the same receptor as methadone and therefore has a similar side effect profile. However, buprenorphine has a ceiling effect, which greatly reduces the risk of fatal respiratory depression.18 It also does not cause clinically significant QTc prolongation and is preferable in patients who have cardiac risk factors.18
Another advantage is that buprenorphine has fewer identified medication interactions than methadone.18 Further, induction of buprenorphine in patients with opioid use disorder has been shown to be safer than methadone.19
Although buprenorphine has been found to be 6 times safer than methadone with regard to overdose among the general population,20 it can still cause fatal intoxication if used in combination with central nervous system depressants.21
Buprenorphine has been also associated with hepatotoxicity, though the risk of new-onset liver disease appears to be low.22
NALTREXONE IS LESS EFFECTIVE THAN METHADONE, BUPRENORPHINE
Besides methadone and buprenorphine, the only other approved option for treating opioid use disorder is the opioid antagonist naltrexone.
Naltrexone has significantly less abuse potential, as it provides no euphoria, but patients do not like it. Even with the long-acting formulation (Vivitrol), naltrexone treatment is significantly less effective than methadone or buprenorphine.23–25 Further, although naltrexone is not a controlled substance and so does not face the same scrutiny as the agonist therapies, there are other significant barriers. Additional information on naltrexone is presented in reviews by Modesto-Lowe and Van Kirk24 and Woody.25
OBSTACLES TO TREATMENT
People hold conflicting views about opioid agonist therapy. Some believe that “trading one drug for another” is not a legitimate therapeutic strategy, and they may feel ashamed of being on maintenance therapy.26 Similarly, some argue that the answer to establishing stable abstinence does not lie simply in prescribing medications.
The contrary argument is that these medications, if used appropriately, confer many benefits such as reducing the medical and psychosocial sequelae of opioid addiction.18 In fact, properly treated patients no longer meet the diagnostic criteria of opioid use disorder, and both methadone and buprenorphine are on the World Health Organization’s (WHO) list of essential medicines.27
Despite endorsement by the WHO, the stigma attached to the opioid agonists has been difficult to overcome. Patients with opioid use disorder may be viewed with distrust by healthcare providers and often do not feel welcome in healthcare settings or in self-help recovery groups.28
Barriers to methadone therapy
Federal regulations on methadone prescribing and use were established to promote patient safety and decrease diversion, but they may also complicate access to care.29 They stipulate that to qualify for methadone maintenance, patients need to demonstrate opioid addiction for 1 year, except for pregnant women and those who have been incarcerated in the past 6 months. Patients under the age of 18 must have 2 documented failed treatment episodes as well as approval by a guardian to receive treatment.
Inconvenience. Methadone can be prescribed for opioid dependence only by an accredited treatment program. Patients must therefore travel to the clinic and wait to be evaluated on a daily basis for a minimum of 90 days. Only after they demonstrate consistent responsible behavior and negative results on urine testing do they become eligible to take methadone home.29 If a patient is to travel out of the area during the initial 90 days of treatment, he or she must make arrangements in advance to find a clinic that will provide a “guest dose.”
The inconvenience arising from the regulations may deter some patients from seeking methadone therapy. In spite of this, once patients are started on methadone, more of them continue treatment than with buprenorphine.18 A proposed reason is that methadone is a potent full opioid agonist and therefore relieves withdrawal symptoms and craving more effectively than buprenorphine, which is a partial agonist.30 Another possible reason is the higher level of supervision afforded by methadone clinics, which require daily contact for at least 90 days.
Safety concerns arise from methadone diversion, as illicit use may have lethal consequences. In the past decade, deaths from methadone overdose have risen significantly, most of them due to respiratory depression or torsade de pointes.13 However, most cases of diversion and overdose involve methadone that is prescribed for pain by individual practitioners and not from maintenance programs.13
Advantages of buprenorphine
Together, methadone’s lethality, stigma, and inconvenience may contribute to patients preferring buprenorphine.31
The regulations governing buprenorphine’s use are less restrictive than those with methadone. For example, patients must have a diagnosis of opioid addiction to be prescribed buprenorphine, but they are not required to carry the diagnosis for a year before treatment.31 Additionally, they do not need to travel to a federally approved opioid treatment center daily and can receive buprenorphine directly from a physician in an outpatient setting.
Under the Drug Abuse Treatment Act (DATA) of 2000, any physician can apply for a waiver to prescribe and dispense buprenorphine in his or her office. To qualify for an initial waiver, physicians must either obtain certification in the fields of addiction medicine or addiction psychiatry or complete an approved 8-hour training session.32 Each physician starts with a maximum of 30 patients, but can apply to treat up to 100 patients after 1 year and eventually up to 275 patients. Physicians must document every buprenorphine prescription they write and be able to refer patients for counseling.31
As of February 2017, nurse practitioners and physician assistants can also apply for a DATA 2000 waiver. All waivered providers are subject to unannounced visits from the Drug Enforcement Administration once every 5 years.32
While there are no federal restrictions on the amount of buprenorphine that can be dispensed, some states and some insurance companies have placed restrictions on dose or length of treatment.33 Buprenorphine patients can fill their prescriptions at any pharmacy and are permitted to bring their medication home, which improves access to care. However, office-based outpatient treatment is not without risk, and preventing buprenorphine diversion remains a challenge.34
‘Lending’ buprenorphine is a felony
Addicts have illegally used buprenorphine to self-treat opioid withdrawal, craving, and dependence.35 Its misuse has also been coupled with self-treatment of conditions that include depression and pain.36
A survey found that 83.7% of patients deem buprenorphine diversion to be appropriate; further, most patients said they consider it unethical to withhold prescribed buprenorphine from individuals showing symptoms of withdrawal.34 Physicians who prescribe buprenorphine must inform their patients that even “lending” or giving away their medication is a felony.
Prescribing physicians must also be diligent about monitoring for signs of diversion such as inconsistent urine toxicology screens, “lost” medication, and requests for early refills or escalating doses.37
EVALUATING PATIENTS FOR OPIOID REPLACEMENT THERAPY
In addition to federal regulations, we propose a 4-step approach to evaluate eligibility for opioid replacement therapy based on existing guidelines.37–39
Step 1: History and physical examination
The history should give particular attention to the patient’s cardiac, pulmonary, and hepatic status, with consideration of the risks of any medical comorbidities (eg, bacterial endocarditis, HIV and HCV infection) that might influence treatment.37
It is also essential to evaluate for any contraindications or drug interactions before prescribing methadone or buprenorphine.38
Contraindications to methadone maintenance include40:
- Cor pulmonale
- Methadone hypersensitivity
- Pseudomembranous colitis
- Selegiline use (due to risk of serotonin syndrome)
- Ileum paralyticus.
Contraindications to buprenorphine use include:
- Hypersensitivity to naloxone or buprenorphine
- Impaired liver function (due to the risk of inadvertent overdose associated with slowed metabolism).
Concurrent use of alcohol or illicit benzodiazepines is a relative contraindication to both methadone and buprenorphine due to the risk of respiratory depression and overdose.37 Likewise, avoid coprescribing opioid agonists and benzodiazepines whenever possible. Obtain a complete list of current medications and query a prescription-monitoring database to determine whether any controlled substances are currently prescribed.37
During the physical examination, look for stigmata of intravenous drug use such as track marks or abscesses37 and document any physical findings consistent with intoxication or withdrawal. Patients must be completely detoxed or in withdrawal before beginning buprenorphine induction; premature induction can precipitate withdrawal.38
A discussion of pregnant patients with opioid use disorder is beyond the scope of this paper. However, it is incumbent on the prescriber to inquire whether the client is pregnant or intends to become pregnant and what birth control methods are in place.
Step 2: Assess psychiatric status
Assessment of the patient’s psychiatric status, including an assessment of alcohol and other drug use, will help determine his or her eligibility for opioid agonists.37 To prepare for the need to manage patients with psychiatrically complex issues, it is helpful to develop relationships with addiction specialists and psychiatrists who are familiar with opioid replacement therapy in your area. This will make it easier to collaborate on patients’ care.
Ask all patients directly about suicidal or homicidal ideation. Any patient with active suicidal or homicidal ideation should be assessed for need of immediate hospitalization by a psychiatrist or another qualified mental health professional. Patients with a history of suicidal ideation should be monitored closely by a mental health professional throughout treatment.37
Many if not most patients with opioid use disorder have concurrent psychiatric disorders, and the interplay between these disorders is complex.40,41 Depression, for example, can precede and even precipitate drug use (an observation supporting the “self-medication theory”).42 If the underlying depressive disorder is not addressed, relapse is nearly inevitable.
It has also been shown that both chronic opioid use and withdrawal can exacerbate aversive emotional states. This escalation of symptoms may result from the pharmacologic effects of opioids or from psychosocial sequelae that can arise from chronic opioid use.41 In this situation, maintaining abstinence can lead to resolution of depressive symptoms. As depression and opioid use can occur together, successful treatment requires equal attention to both illnesses.
Other common comorbidities in patients with opioid use disorder include posttraumatic stress disorder, attention deficit hyperactivity disorder, antisocial personality disorder, and concurrent substance abuse disorders.43 The confluence of antisocial personality disorder is particularly important, as patients with antisocial personality disorder display disruptive and maladaptive behaviors.
Identify any psychotropic medication that is prescribed and check carefully for drug interactions. This applies especially to methadone, as many psychiatric medications also prolong the QT interval. Moreover, patients may not be forthcoming about the use of psychiatric medication.
Find out whether the patient is using any other addictive substances, particularly those that affect the central nervous system, as those who use fentanyl, benzodiazepines, or alcohol are at the highest risk of overdose.31 Often the best option for those with concurrent substance use disorders is inpatient detoxification followed by residential rehabilitation care. Either buprenorphine or methadone can then be initiated upon return to an outpatient setting.
Step 3: Assess psychosocial status
To what extent do the patient’s home environment and support systems promote a drug-free lifestyle? Unfortunately, the psychosocial status of many of these patients is fragile, and they may live in areas where illicit drugs are readily available (which can be urban, suburban, or rural), making it difficult to stay substance-free.38
Generally, lifestyle modifications are needed to transform maladaptive behaviors and promote an environment conducive to long-term recovery. Referrals to social services to address housing, vocational needs, and entitlements may be helpful.39
Step 4: Assess readiness to change
According to one model, people go through 5 stages when changing a behavior: precontemplation, contemplation, preparation for action, action, and maintenance.43 In general, the further along the stages a patient is, the more appropriate he or she is for office-based treatment with buprenorphine.39
The level of change can be assessed with tools such as Stages of Change Readiness and Treatment Eagerness Scale (SOCRATES). Use of stage-specific strategies may enhance a patient’s readiness to cease opioid use.43
Precontemplation. Those in the precontemplation stage are not ready to think about changing their behavior.43 They may be unaware of or unwilling to consider the risks associated with their opioid use and resistant to the idea of quitting. Engagement with opioid agonists for individuals in this stage is low and dropout rates are likely high.
Thus, the proper approach for “precontemplators” is to help them develop some ambivalence about their opioid use. One tactic is to involve the patient in a discussion of the personal benefits and risks of opioid use.
Contemplation. Individuals in the contemplation stage have begun to weigh the costs and benefits of opioid use and express ambivalence about it.44 Because the patient is willing to explore the risks of ongoing use and consider the benefits of treatment, the goal in this stage is to elicit a commitment from the individual to seek treatment.
Preparation. The person in this stage moves from thinking about treatment to planning what action to take.45 As the individual prepares to enter treatment, indecision tends to resurface, as well as self-doubt about his or her ability to change. During this stage, it is important for the provider to spell out goals (abstinence) and strategies (eg, counseling, medication) and enhance a sense of self-efficacy.
Action and maintenance. Patients in these stages engage in treatment and employ new strategies to abstain from opioid use. Maintaining these behaviors can be a daily struggle. Expressing confidence in the patient’s ability to abstain from use will support his or her progress. Behavioral interventions such as strategic avoidance of triggers and engagement in alternative activities (eg, support groups, exercise, faith-based practices) will help to maintain abstinence.
A CHRONIC CONDITION
Opioid use disorder, like many chronic illnesses, requires long-term attention to attain successful patient outcomes. The opioid agonists methadone and buprenorphine are the mainstay of treatment for it, conferring benefits such as reducing opioid use and preventing relapse.
Candidates for opioid agonist therapy should undergo a multidisciplinary assessment, including an evaluation on the patient’s readiness to change his or her opioid use.
Patient education should include a discussion of the risks of methadone (eg, respiratory depression, fatal overdose, and QTc prolongation) and buprenorphine (eg hepatotoxicity) and their benefits (eg, controlling craving, decreasing the risk of relapse). Patients should also be educated about overdose and diversion.
Despite the difficulties inherent in treating patients with opioid use disorder, when used appropriately, opioid agonist therapy can be lifesaving for patients struggling with long-term opioid addiction.
Acknowledgment: We thank Katelyn Colosi, BS, and Drs. Susan Wolfe, Dennis Bouffard, and Sinha Shirshendu for their helpful comments.
- Wakeman SE, Pham-Kanter G, Donelan K. Attitudes, practices, and preparedness to care for patients with substance use disorder: results from a survey of general internists. Subst Abus 2016; 37:635–641.
- Samuels EA, Dwyer K, Mello MJ, Baird J, Kellogg AR, Bernstein E. Emergency department-based opioid harm reduction: moving physicians from willing to doing. Acad Emerg Med 2016; 23:455–465.
- Mohlman MK, Tanzman B, Finison K, Pinette M, Jones C. Impact of medication-assisted treatment for opioid addiction on Medicaid expenditures and health services utilization rates in Vermont. J Subst Abuse Treat 2016; 67:9–14.
- American Psychiatric Association. Diagnostic and Statistical Manual of Mental Disorders, Fifth Edition. Arlington, VA, American Psychiatric Association, 2013.
- Center for Behavioral Health Statistics and Quality. Behavioral health trends in the United States: results from the 2014 National Survey on Drug Use and Health. www.samhsa.gov/data. Accessed April 6, 2017.
- Compton WM, Jones CM, Baldwin GT. Relationship between nonmedical prescription-opioid use and heroin use. N Engl J Med 2016; 374:154–163.
- Ruan X, Wyche MQ, Kaye AD. Analyzing the relationship between nonmedical prescription-opioid use and heroin use. J Opioid Manage 2016; 12:11–14.
- Hser YI, Evans E, Grella C, Ling W, Anglin D. Long-term course of opioid addiction. Harv Rev Psychiatry 2015; 23:76–89.
- Nielsen S, Hillhouse M, Mooney L, Ang A, Ling W. Buprenorphine pharmacotherapy and behavioral treatment: comparison of outcomes among prescription opioid users, heroin users and combination users. J Subst Abuse Treat 2015; 48:70–76.
- Moore BA, Fiellin DA, Barry DT, et al. Primary care office-based buprenorphine treatment: comparison of heroin and prescription opioid dependent patients. J Gen Intern Med 2007; 22:527–530.
- Fischer B, Patra J, Cruz MF, Gittins J, Rehm J. Comparing heroin users and prescription opioid users in a Canadian multi-site population of illicit opioid users. Drug Alcohol Rev 2008; 27:625–632.
- Compton WM, Jones CM, Baldwin GT. Relationship between nonmedical prescription-opioid use and heroin use. N Engl J Med 2016; 374:154–163.
- Jones CM, Baldwin GT, Manocchio T, White JO, Mack KA. Trends in methadone distribution for pain treatment, methadone diversion, and overdose deaths—United States, 2002–2014. MMWR Morb Mortal Wkly Rep 2016; 65:667–671.
- Baxter LE Sr, Campbell A, Deshields M, et al. Safe methadone induction and stabilization: report of an expert panel. J Addict Med 2013; 7:377–386.
- Alinejad S, Kazemi T, Zamani N, Hoffman RS, Mehrpour O. A systematic review of the cardiotoxicity of methadone. EXCLI J 2015; 14:577–600.
- Johnson RE, Strain EC, Amass L. Buprenorphine: how to use it right. Drug Alcohol Depend 2003; 70(suppl 2):S59–S77.
- Ling W. Buprenorphine implant for opioid addiction. Pain Manage 2012; 2:345–350.
- Saxon AJ, Hser YI, Woody G, Ling W. Medication-assisted treatment for opioid addiction: methadone and buprenorphine. J Food Drug Anal 2013; 21:S69–S72.
- Kimber J, Larney S, Hickman M, Randall D, Degenhardt L. Mortality risk of opioid substitution therapy with methadone versus buprenorphine: a retrospective cohort study. Lancet Psychiatry 2015; 2:901–908.
- Marteau D, McDonald R, Patel K. The relative risk of fatal poisoning by methadone or buprenorphine within the wider population of England and Wales. BMJ Open 2015; 5: e007629.
- Kintz P. Deaths involving buprenorphine: a compendium of French cases. Forensic Sci Int 2001; 121:65–69.
- Zuin M, Giorgini A, Selmi C, et al. Acute liver and renal failure during treatment with buprenorphine at therapeutic dose. Dig Liver Dis 2009; 41:e8–e10.
- Klein JW. Pharmacotherapy for substance use disorders. Med Clin North Am 2016; 100:891–910.
- Modesto-Lowe V, Van Kirk J. Clinical uses of naltrexone: a review of the evidence. Exp Clin Psychopharmocol 2002; 10:213–227.
- Woody GE. Agonist models for treating persons with substance use disorders. Curr Psychiatry Rep 2014; 16:489.
- Sanders JJ, Roose RJ, Lubrano MC, Lucan SC. Meaning and methadone: patient perceptions of methadone dose and a model to promote adherence to maintenance treatment. J Addict Med 2013; 7:307–313.
- Herget G. Methadone and buprenorphine added to the WHO list of essential medicines. HIV/AIDS Policy Law Rev 2005; 10:23–24.
- Suzuki J, Dodds T. Clinical recommendation of 12-step meeting attendance and discussion regarding disclosure of buprenorphine use among patients in office-based opioid treatment. Subst Abus 2016; 37:31–34.
- Rettig RA, Yarmolinsky A. Federal Regulation of Methadone Treatment. Washington, DC: National Academies Press; 1995.
- Srivastava A, Kahan M, Nader M. Primary care management of opioid use disorders: abstinence, methadone, or buprenorphine-naloxone? Can Fam Physician 2017; 63:200–205.
- Substance Abuse and Mental Health Services Administration. Federal Guidelines for Opioid Treatment Programs. Rockville, MD: Substance Abuse and Mental Health Services Administration; 2015.
- Substance Abuse and Mental Health Services Administration SAMSHA. Buprenorphine waiver management. www.samhsa.gov/medication-assisted-treatment/buprenorphine-waiver-management. Accessed April 6, 2017.
- Mark TL, Lubran R, McCance-Kats EF, Chalk M, Richardson J. Medicaid coverage of medications to treat alcohol and opioid dependence. J Subst Abuse Treat 2015; 55:1–5.
- Johnson B, Richert T. Diversion of methadone and buprenorphine from opioid substitution treatment: the importance of patients’ attitudes and norms. J Subst Abuse Treat 2015; 54:50–55.
- Yokell MA, Zaller ND, Green TC, Rich JD. Buprenorphine and buprenorphine/naloxone diversion, misuse, and illicit use: an international review. Curr Drug Abuse Rev 2011; 4:28–41.
- Schuman-Olivier Z, Albanese M, Nelson SE, et al. Self-treatment: illicit buprenorphine use by opioid-dependent treatment seekers. J Subst Abuse Treat 2010; 39:41–50.
- American Society of Addiction Medicine. National practice guidelines for the use of medications in the treatment of addiction involving opioid use. www.asam.org/docs/default-source/practice-support/guidelines-and-consensus-docs/asam-national-practice-guideline-supplement.pdf. Accessed April 6, 2017.
- McNicholas L. Clinical guidelines for the use of buprenorphine in the treatment of opioid addiction. Rockville, MD: US Department of Health and Human Services, Substance Abuse and Mental Health Service Administration; 2004.
- Center for Substance Abuse Treatment. Clinical guidelines for the use of buprenorphine in the treatment of opioid addiction. Rockville (MD): Substance Abuse and Mental Health Services Administration (US); 2004. (Treatment Improvement Protocol (TIP) Series, No. 40.) www.ncbi.nlm.nih.gov/books/NBK64245. Accessed April 6, 2017.
- Zippel-Schultz B, Specka M, Cimander K, et al. Outcomes of patients in long-term opioid maintenance treatment. Subst Use Misuse 2016; 51:1493–1503.
- Martins SS, Keyes KM, Storr CL, Zhu H, Chilcoat HD. Pathways between nonmedical opioid use/dependence and psychiatric disorders: results from the National Epidemiologic Survey on Alcohol and Related Conditions. Drug Alcohol Depend 2009; 103:16–24.
- Khantzian EJ. The self-medication hypothesis of addictive disorders: focus on heroin and cocaine dependence. Am J Psychiatry 1985; 142:1259–1264.
- Belding MA, Iguchi MY, Lamb RJ, Lakin M, Terry R. Stages and processes of change among polydrug users in methadone maintenance treatment. Drug Alcohol Depend 1995; 39:45–53.
- Peteet JR, Brenner S, Curtiss D, Ferrigno M, Kauffman J. A stage of change approach to addiction in the medical setting. Gen Hosp Psychiatry 1998; 20:267–273.
- Vijay A, Bazazi AR, Yee I, Kamarulzaman A, Altice FL. Treatment readiness, attitudes toward, and experiences with methadone and buprenorphine maintenance therapy among people who inject drugs in Malaysia. J Subst Abuse Treat 2015; 54:29–36.
For a patient struggling with opioid addiction, opioid agonist therapy with methadone or buprenorphine can reduce craving and opioid use and may even save his or her life. But many clinicians are unfamiliar with this evidence-based treatment,1,2 which is best started early in the course of addiction.3
This article outlines the pharmacology of these drugs, their clinical uses, and the challenges of using them to treat opioid addiction.
DIAGNOSTIC CRITERIA
Opioid addiction, formally known as opioid use disorder, is a pattern of opioid misuse leading to clinically significant impairment in multiple areas of life. The Diagnostic and Statistical Manual of Mental Disorders, Fifth Edition, lists 11 diagnostic criteria, but only 2 need to be present within the past year to make the diagnosis4:
- Taking opioids longer or in higher doses than was intended
- A persistent desire or unsuccessful efforts to cut down or control opioid use
- Spending a great deal of time obtaining, using, or recovering from using opioids
- Craving opioids
- Repeatedly failing to fulfill obligations at work, school, or home due to opioid use
- Continuing to use opioids even though it causes or exacerbates social or interpersonal problems
- Giving up or curtailing important social, occupational, or recreational activities because of opioid use
- Repeatedly using opioids in situations in which it is physically hazardous
- Continuing to use opioids despite knowledge of having a persistent or recurrent physical or psychological problem that is likely to have been caused or exacerbated by the substance
- Tolerance
- Withdrawal.
Recent estimates indicate that 2.23 million people in the United States have opioid use disorder (426,000 with heroin and 1.8 million with prescription opioids).5
Progression from prescription opioids to heroin
We have observed that many patients with opioid use disorder start by misusing prescription opioids. Over time, tolerance can develop, which drives patients to use higher and higher doses.6
As the addiction progresses, a subset of prescription opioid users advances to using heroin, which is typically less expensive and easier to obtain.7 Most patients start with the intranasal route but eventually inject it intravenously.6,7
For many addicts, heroin use has medical consequences such as hepatitis C virus (HCV) and human immunodeficiency virus (HIV) infection, psychiatric problems such as depression and anxiety, and illegal activities such as theft and sex work.8 People who use heroin appear to have more severe addiction and a lower socioeconomic status than prescription opioid users.9–11 But recently, a growing number of middle class individuals are becoming addicted to heroin.12
METHADONE
Methadone is a long-acting synthetic opioid that functions as a full agonist on the mu-opioid receptor. The drug binds, occupies, and stimulates the receptor, preventing withdrawal symptoms and reducing opioid cravings for at least 24 hours.13
Adverse effects of methadone
The most common adverse effects include lightheadedness, dizziness, sedation, nausea, vomiting, and sweating.14 Other adverse effects:
Unintentional overdose. The risk is serious, as a single 30-mg dose can be fatal in people who are opioid-naïve.13
QTc prolongation, which can lead to torsade de pointes. This risk, which is dose-related, must be taken into consideration in patients who have any cardiac symptoms (eg, syncope, arrhythmia), pathology (familial QT prolongation), or other risk factors for QTc prolongation (eg, hypokalemia, QTc-prolonging medications).15
Respiratory depression, which can be fatal. This dose-related risk is heightened during the first 4 weeks of treatment if titration is too rapid or if methadone is used in combination with other drugs that cause central nervous system or respiratory depression.13,14
Starting methadone
To prevent respiratory depression and death related to rapid induction, the general rule is to start methadone at a low daily dose (20–30 mg) depending on the patient’s withdrawal symptoms.14 During this period, patients need to be closely monitored and educated on the perils of concomitant use of central nervous system depressants.14
In most patients, the dose is titrated up until their withdrawal symptoms and cravings are eliminated, which generally requires 60 to 120 mg daily.14 Hepatic and renal impairment, pregnancy, and advanced age can alter methadone pharmacokinetics and may therefore necessitate dose adjustment.
BUPRENORPHINE
Buprenorphine is an alkaloid thebaine opioid derivative that acts as a partial mu-opioid agonist and a kappa antagonist.16 Like methadone, buprenorphine is used to manage cravings and withdrawal symptoms.16 Dosages of 4 to 16 mg (up to 32 mg) per day of buprenorphine are usually required to adequately control opioid cravings.16
Sublingual and subdermal products
Buprenorphine is currently available in the United States in sublingual and subdermal formulations.16,17
Sublingual buprenorphine is usually combined with naloxone in a 4:1 ratio to deter intravenous use. Intravenous injection of the combination product can precipitate withdrawal due to the antagonist action of naloxone. (Taken orally or sublingually, naloxone is poorly absorbed and has little or no clinical effect.) Buprenorphine-naloxone is available in tablets, a sublingual film strip, and a buccal film strip. Buprenorphine is also available by itself in a sublingual formulation.
The US Food and Drug Administration has approved a buprenorphine subdermal implant, Probuphine. Four rods, about 1 inch long, are placed under the skin in the inner aspect of the upper arm and provide the equivalent of 8 mg of buprenorphine daily for 6 months.17 However, this method is formulated only for maintenance treatment and cannot be used for induction. Additionally, it is recommended that the implants be surgically removed at the end of 6 months, after which another set of implants can be inserted in the other arm or the patient can switch to sublingual therapy, depending on the clinical situation and patient preference.17
Generally safer than methadone
Buprenorphine works on the same receptor as methadone and therefore has a similar side effect profile. However, buprenorphine has a ceiling effect, which greatly reduces the risk of fatal respiratory depression.18 It also does not cause clinically significant QTc prolongation and is preferable in patients who have cardiac risk factors.18
Another advantage is that buprenorphine has fewer identified medication interactions than methadone.18 Further, induction of buprenorphine in patients with opioid use disorder has been shown to be safer than methadone.19
Although buprenorphine has been found to be 6 times safer than methadone with regard to overdose among the general population,20 it can still cause fatal intoxication if used in combination with central nervous system depressants.21
Buprenorphine has been also associated with hepatotoxicity, though the risk of new-onset liver disease appears to be low.22
NALTREXONE IS LESS EFFECTIVE THAN METHADONE, BUPRENORPHINE
Besides methadone and buprenorphine, the only other approved option for treating opioid use disorder is the opioid antagonist naltrexone.
Naltrexone has significantly less abuse potential, as it provides no euphoria, but patients do not like it. Even with the long-acting formulation (Vivitrol), naltrexone treatment is significantly less effective than methadone or buprenorphine.23–25 Further, although naltrexone is not a controlled substance and so does not face the same scrutiny as the agonist therapies, there are other significant barriers. Additional information on naltrexone is presented in reviews by Modesto-Lowe and Van Kirk24 and Woody.25
OBSTACLES TO TREATMENT
People hold conflicting views about opioid agonist therapy. Some believe that “trading one drug for another” is not a legitimate therapeutic strategy, and they may feel ashamed of being on maintenance therapy.26 Similarly, some argue that the answer to establishing stable abstinence does not lie simply in prescribing medications.
The contrary argument is that these medications, if used appropriately, confer many benefits such as reducing the medical and psychosocial sequelae of opioid addiction.18 In fact, properly treated patients no longer meet the diagnostic criteria of opioid use disorder, and both methadone and buprenorphine are on the World Health Organization’s (WHO) list of essential medicines.27
Despite endorsement by the WHO, the stigma attached to the opioid agonists has been difficult to overcome. Patients with opioid use disorder may be viewed with distrust by healthcare providers and often do not feel welcome in healthcare settings or in self-help recovery groups.28
Barriers to methadone therapy
Federal regulations on methadone prescribing and use were established to promote patient safety and decrease diversion, but they may also complicate access to care.29 They stipulate that to qualify for methadone maintenance, patients need to demonstrate opioid addiction for 1 year, except for pregnant women and those who have been incarcerated in the past 6 months. Patients under the age of 18 must have 2 documented failed treatment episodes as well as approval by a guardian to receive treatment.
Inconvenience. Methadone can be prescribed for opioid dependence only by an accredited treatment program. Patients must therefore travel to the clinic and wait to be evaluated on a daily basis for a minimum of 90 days. Only after they demonstrate consistent responsible behavior and negative results on urine testing do they become eligible to take methadone home.29 If a patient is to travel out of the area during the initial 90 days of treatment, he or she must make arrangements in advance to find a clinic that will provide a “guest dose.”
The inconvenience arising from the regulations may deter some patients from seeking methadone therapy. In spite of this, once patients are started on methadone, more of them continue treatment than with buprenorphine.18 A proposed reason is that methadone is a potent full opioid agonist and therefore relieves withdrawal symptoms and craving more effectively than buprenorphine, which is a partial agonist.30 Another possible reason is the higher level of supervision afforded by methadone clinics, which require daily contact for at least 90 days.
Safety concerns arise from methadone diversion, as illicit use may have lethal consequences. In the past decade, deaths from methadone overdose have risen significantly, most of them due to respiratory depression or torsade de pointes.13 However, most cases of diversion and overdose involve methadone that is prescribed for pain by individual practitioners and not from maintenance programs.13
Advantages of buprenorphine
Together, methadone’s lethality, stigma, and inconvenience may contribute to patients preferring buprenorphine.31
The regulations governing buprenorphine’s use are less restrictive than those with methadone. For example, patients must have a diagnosis of opioid addiction to be prescribed buprenorphine, but they are not required to carry the diagnosis for a year before treatment.31 Additionally, they do not need to travel to a federally approved opioid treatment center daily and can receive buprenorphine directly from a physician in an outpatient setting.
Under the Drug Abuse Treatment Act (DATA) of 2000, any physician can apply for a waiver to prescribe and dispense buprenorphine in his or her office. To qualify for an initial waiver, physicians must either obtain certification in the fields of addiction medicine or addiction psychiatry or complete an approved 8-hour training session.32 Each physician starts with a maximum of 30 patients, but can apply to treat up to 100 patients after 1 year and eventually up to 275 patients. Physicians must document every buprenorphine prescription they write and be able to refer patients for counseling.31
As of February 2017, nurse practitioners and physician assistants can also apply for a DATA 2000 waiver. All waivered providers are subject to unannounced visits from the Drug Enforcement Administration once every 5 years.32
While there are no federal restrictions on the amount of buprenorphine that can be dispensed, some states and some insurance companies have placed restrictions on dose or length of treatment.33 Buprenorphine patients can fill their prescriptions at any pharmacy and are permitted to bring their medication home, which improves access to care. However, office-based outpatient treatment is not without risk, and preventing buprenorphine diversion remains a challenge.34
‘Lending’ buprenorphine is a felony
Addicts have illegally used buprenorphine to self-treat opioid withdrawal, craving, and dependence.35 Its misuse has also been coupled with self-treatment of conditions that include depression and pain.36
A survey found that 83.7% of patients deem buprenorphine diversion to be appropriate; further, most patients said they consider it unethical to withhold prescribed buprenorphine from individuals showing symptoms of withdrawal.34 Physicians who prescribe buprenorphine must inform their patients that even “lending” or giving away their medication is a felony.
Prescribing physicians must also be diligent about monitoring for signs of diversion such as inconsistent urine toxicology screens, “lost” medication, and requests for early refills or escalating doses.37
EVALUATING PATIENTS FOR OPIOID REPLACEMENT THERAPY
In addition to federal regulations, we propose a 4-step approach to evaluate eligibility for opioid replacement therapy based on existing guidelines.37–39
Step 1: History and physical examination
The history should give particular attention to the patient’s cardiac, pulmonary, and hepatic status, with consideration of the risks of any medical comorbidities (eg, bacterial endocarditis, HIV and HCV infection) that might influence treatment.37
It is also essential to evaluate for any contraindications or drug interactions before prescribing methadone or buprenorphine.38
Contraindications to methadone maintenance include40:
- Cor pulmonale
- Methadone hypersensitivity
- Pseudomembranous colitis
- Selegiline use (due to risk of serotonin syndrome)
- Ileum paralyticus.
Contraindications to buprenorphine use include:
- Hypersensitivity to naloxone or buprenorphine
- Impaired liver function (due to the risk of inadvertent overdose associated with slowed metabolism).
Concurrent use of alcohol or illicit benzodiazepines is a relative contraindication to both methadone and buprenorphine due to the risk of respiratory depression and overdose.37 Likewise, avoid coprescribing opioid agonists and benzodiazepines whenever possible. Obtain a complete list of current medications and query a prescription-monitoring database to determine whether any controlled substances are currently prescribed.37
During the physical examination, look for stigmata of intravenous drug use such as track marks or abscesses37 and document any physical findings consistent with intoxication or withdrawal. Patients must be completely detoxed or in withdrawal before beginning buprenorphine induction; premature induction can precipitate withdrawal.38
A discussion of pregnant patients with opioid use disorder is beyond the scope of this paper. However, it is incumbent on the prescriber to inquire whether the client is pregnant or intends to become pregnant and what birth control methods are in place.
Step 2: Assess psychiatric status
Assessment of the patient’s psychiatric status, including an assessment of alcohol and other drug use, will help determine his or her eligibility for opioid agonists.37 To prepare for the need to manage patients with psychiatrically complex issues, it is helpful to develop relationships with addiction specialists and psychiatrists who are familiar with opioid replacement therapy in your area. This will make it easier to collaborate on patients’ care.
Ask all patients directly about suicidal or homicidal ideation. Any patient with active suicidal or homicidal ideation should be assessed for need of immediate hospitalization by a psychiatrist or another qualified mental health professional. Patients with a history of suicidal ideation should be monitored closely by a mental health professional throughout treatment.37
Many if not most patients with opioid use disorder have concurrent psychiatric disorders, and the interplay between these disorders is complex.40,41 Depression, for example, can precede and even precipitate drug use (an observation supporting the “self-medication theory”).42 If the underlying depressive disorder is not addressed, relapse is nearly inevitable.
It has also been shown that both chronic opioid use and withdrawal can exacerbate aversive emotional states. This escalation of symptoms may result from the pharmacologic effects of opioids or from psychosocial sequelae that can arise from chronic opioid use.41 In this situation, maintaining abstinence can lead to resolution of depressive symptoms. As depression and opioid use can occur together, successful treatment requires equal attention to both illnesses.
Other common comorbidities in patients with opioid use disorder include posttraumatic stress disorder, attention deficit hyperactivity disorder, antisocial personality disorder, and concurrent substance abuse disorders.43 The confluence of antisocial personality disorder is particularly important, as patients with antisocial personality disorder display disruptive and maladaptive behaviors.
Identify any psychotropic medication that is prescribed and check carefully for drug interactions. This applies especially to methadone, as many psychiatric medications also prolong the QT interval. Moreover, patients may not be forthcoming about the use of psychiatric medication.
Find out whether the patient is using any other addictive substances, particularly those that affect the central nervous system, as those who use fentanyl, benzodiazepines, or alcohol are at the highest risk of overdose.31 Often the best option for those with concurrent substance use disorders is inpatient detoxification followed by residential rehabilitation care. Either buprenorphine or methadone can then be initiated upon return to an outpatient setting.
Step 3: Assess psychosocial status
To what extent do the patient’s home environment and support systems promote a drug-free lifestyle? Unfortunately, the psychosocial status of many of these patients is fragile, and they may live in areas where illicit drugs are readily available (which can be urban, suburban, or rural), making it difficult to stay substance-free.38
Generally, lifestyle modifications are needed to transform maladaptive behaviors and promote an environment conducive to long-term recovery. Referrals to social services to address housing, vocational needs, and entitlements may be helpful.39
Step 4: Assess readiness to change
According to one model, people go through 5 stages when changing a behavior: precontemplation, contemplation, preparation for action, action, and maintenance.43 In general, the further along the stages a patient is, the more appropriate he or she is for office-based treatment with buprenorphine.39
The level of change can be assessed with tools such as Stages of Change Readiness and Treatment Eagerness Scale (SOCRATES). Use of stage-specific strategies may enhance a patient’s readiness to cease opioid use.43
Precontemplation. Those in the precontemplation stage are not ready to think about changing their behavior.43 They may be unaware of or unwilling to consider the risks associated with their opioid use and resistant to the idea of quitting. Engagement with opioid agonists for individuals in this stage is low and dropout rates are likely high.
Thus, the proper approach for “precontemplators” is to help them develop some ambivalence about their opioid use. One tactic is to involve the patient in a discussion of the personal benefits and risks of opioid use.
Contemplation. Individuals in the contemplation stage have begun to weigh the costs and benefits of opioid use and express ambivalence about it.44 Because the patient is willing to explore the risks of ongoing use and consider the benefits of treatment, the goal in this stage is to elicit a commitment from the individual to seek treatment.
Preparation. The person in this stage moves from thinking about treatment to planning what action to take.45 As the individual prepares to enter treatment, indecision tends to resurface, as well as self-doubt about his or her ability to change. During this stage, it is important for the provider to spell out goals (abstinence) and strategies (eg, counseling, medication) and enhance a sense of self-efficacy.
Action and maintenance. Patients in these stages engage in treatment and employ new strategies to abstain from opioid use. Maintaining these behaviors can be a daily struggle. Expressing confidence in the patient’s ability to abstain from use will support his or her progress. Behavioral interventions such as strategic avoidance of triggers and engagement in alternative activities (eg, support groups, exercise, faith-based practices) will help to maintain abstinence.
A CHRONIC CONDITION
Opioid use disorder, like many chronic illnesses, requires long-term attention to attain successful patient outcomes. The opioid agonists methadone and buprenorphine are the mainstay of treatment for it, conferring benefits such as reducing opioid use and preventing relapse.
Candidates for opioid agonist therapy should undergo a multidisciplinary assessment, including an evaluation on the patient’s readiness to change his or her opioid use.
Patient education should include a discussion of the risks of methadone (eg, respiratory depression, fatal overdose, and QTc prolongation) and buprenorphine (eg hepatotoxicity) and their benefits (eg, controlling craving, decreasing the risk of relapse). Patients should also be educated about overdose and diversion.
Despite the difficulties inherent in treating patients with opioid use disorder, when used appropriately, opioid agonist therapy can be lifesaving for patients struggling with long-term opioid addiction.
Acknowledgment: We thank Katelyn Colosi, BS, and Drs. Susan Wolfe, Dennis Bouffard, and Sinha Shirshendu for their helpful comments.
For a patient struggling with opioid addiction, opioid agonist therapy with methadone or buprenorphine can reduce craving and opioid use and may even save his or her life. But many clinicians are unfamiliar with this evidence-based treatment,1,2 which is best started early in the course of addiction.3
This article outlines the pharmacology of these drugs, their clinical uses, and the challenges of using them to treat opioid addiction.
DIAGNOSTIC CRITERIA
Opioid addiction, formally known as opioid use disorder, is a pattern of opioid misuse leading to clinically significant impairment in multiple areas of life. The Diagnostic and Statistical Manual of Mental Disorders, Fifth Edition, lists 11 diagnostic criteria, but only 2 need to be present within the past year to make the diagnosis4:
- Taking opioids longer or in higher doses than was intended
- A persistent desire or unsuccessful efforts to cut down or control opioid use
- Spending a great deal of time obtaining, using, or recovering from using opioids
- Craving opioids
- Repeatedly failing to fulfill obligations at work, school, or home due to opioid use
- Continuing to use opioids even though it causes or exacerbates social or interpersonal problems
- Giving up or curtailing important social, occupational, or recreational activities because of opioid use
- Repeatedly using opioids in situations in which it is physically hazardous
- Continuing to use opioids despite knowledge of having a persistent or recurrent physical or psychological problem that is likely to have been caused or exacerbated by the substance
- Tolerance
- Withdrawal.
Recent estimates indicate that 2.23 million people in the United States have opioid use disorder (426,000 with heroin and 1.8 million with prescription opioids).5
Progression from prescription opioids to heroin
We have observed that many patients with opioid use disorder start by misusing prescription opioids. Over time, tolerance can develop, which drives patients to use higher and higher doses.6
As the addiction progresses, a subset of prescription opioid users advances to using heroin, which is typically less expensive and easier to obtain.7 Most patients start with the intranasal route but eventually inject it intravenously.6,7
For many addicts, heroin use has medical consequences such as hepatitis C virus (HCV) and human immunodeficiency virus (HIV) infection, psychiatric problems such as depression and anxiety, and illegal activities such as theft and sex work.8 People who use heroin appear to have more severe addiction and a lower socioeconomic status than prescription opioid users.9–11 But recently, a growing number of middle class individuals are becoming addicted to heroin.12
METHADONE
Methadone is a long-acting synthetic opioid that functions as a full agonist on the mu-opioid receptor. The drug binds, occupies, and stimulates the receptor, preventing withdrawal symptoms and reducing opioid cravings for at least 24 hours.13
Adverse effects of methadone
The most common adverse effects include lightheadedness, dizziness, sedation, nausea, vomiting, and sweating.14 Other adverse effects:
Unintentional overdose. The risk is serious, as a single 30-mg dose can be fatal in people who are opioid-naïve.13
QTc prolongation, which can lead to torsade de pointes. This risk, which is dose-related, must be taken into consideration in patients who have any cardiac symptoms (eg, syncope, arrhythmia), pathology (familial QT prolongation), or other risk factors for QTc prolongation (eg, hypokalemia, QTc-prolonging medications).15
Respiratory depression, which can be fatal. This dose-related risk is heightened during the first 4 weeks of treatment if titration is too rapid or if methadone is used in combination with other drugs that cause central nervous system or respiratory depression.13,14
Starting methadone
To prevent respiratory depression and death related to rapid induction, the general rule is to start methadone at a low daily dose (20–30 mg) depending on the patient’s withdrawal symptoms.14 During this period, patients need to be closely monitored and educated on the perils of concomitant use of central nervous system depressants.14
In most patients, the dose is titrated up until their withdrawal symptoms and cravings are eliminated, which generally requires 60 to 120 mg daily.14 Hepatic and renal impairment, pregnancy, and advanced age can alter methadone pharmacokinetics and may therefore necessitate dose adjustment.
BUPRENORPHINE
Buprenorphine is an alkaloid thebaine opioid derivative that acts as a partial mu-opioid agonist and a kappa antagonist.16 Like methadone, buprenorphine is used to manage cravings and withdrawal symptoms.16 Dosages of 4 to 16 mg (up to 32 mg) per day of buprenorphine are usually required to adequately control opioid cravings.16
Sublingual and subdermal products
Buprenorphine is currently available in the United States in sublingual and subdermal formulations.16,17
Sublingual buprenorphine is usually combined with naloxone in a 4:1 ratio to deter intravenous use. Intravenous injection of the combination product can precipitate withdrawal due to the antagonist action of naloxone. (Taken orally or sublingually, naloxone is poorly absorbed and has little or no clinical effect.) Buprenorphine-naloxone is available in tablets, a sublingual film strip, and a buccal film strip. Buprenorphine is also available by itself in a sublingual formulation.
The US Food and Drug Administration has approved a buprenorphine subdermal implant, Probuphine. Four rods, about 1 inch long, are placed under the skin in the inner aspect of the upper arm and provide the equivalent of 8 mg of buprenorphine daily for 6 months.17 However, this method is formulated only for maintenance treatment and cannot be used for induction. Additionally, it is recommended that the implants be surgically removed at the end of 6 months, after which another set of implants can be inserted in the other arm or the patient can switch to sublingual therapy, depending on the clinical situation and patient preference.17
Generally safer than methadone
Buprenorphine works on the same receptor as methadone and therefore has a similar side effect profile. However, buprenorphine has a ceiling effect, which greatly reduces the risk of fatal respiratory depression.18 It also does not cause clinically significant QTc prolongation and is preferable in patients who have cardiac risk factors.18
Another advantage is that buprenorphine has fewer identified medication interactions than methadone.18 Further, induction of buprenorphine in patients with opioid use disorder has been shown to be safer than methadone.19
Although buprenorphine has been found to be 6 times safer than methadone with regard to overdose among the general population,20 it can still cause fatal intoxication if used in combination with central nervous system depressants.21
Buprenorphine has been also associated with hepatotoxicity, though the risk of new-onset liver disease appears to be low.22
NALTREXONE IS LESS EFFECTIVE THAN METHADONE, BUPRENORPHINE
Besides methadone and buprenorphine, the only other approved option for treating opioid use disorder is the opioid antagonist naltrexone.
Naltrexone has significantly less abuse potential, as it provides no euphoria, but patients do not like it. Even with the long-acting formulation (Vivitrol), naltrexone treatment is significantly less effective than methadone or buprenorphine.23–25 Further, although naltrexone is not a controlled substance and so does not face the same scrutiny as the agonist therapies, there are other significant barriers. Additional information on naltrexone is presented in reviews by Modesto-Lowe and Van Kirk24 and Woody.25
OBSTACLES TO TREATMENT
People hold conflicting views about opioid agonist therapy. Some believe that “trading one drug for another” is not a legitimate therapeutic strategy, and they may feel ashamed of being on maintenance therapy.26 Similarly, some argue that the answer to establishing stable abstinence does not lie simply in prescribing medications.
The contrary argument is that these medications, if used appropriately, confer many benefits such as reducing the medical and psychosocial sequelae of opioid addiction.18 In fact, properly treated patients no longer meet the diagnostic criteria of opioid use disorder, and both methadone and buprenorphine are on the World Health Organization’s (WHO) list of essential medicines.27
Despite endorsement by the WHO, the stigma attached to the opioid agonists has been difficult to overcome. Patients with opioid use disorder may be viewed with distrust by healthcare providers and often do not feel welcome in healthcare settings or in self-help recovery groups.28
Barriers to methadone therapy
Federal regulations on methadone prescribing and use were established to promote patient safety and decrease diversion, but they may also complicate access to care.29 They stipulate that to qualify for methadone maintenance, patients need to demonstrate opioid addiction for 1 year, except for pregnant women and those who have been incarcerated in the past 6 months. Patients under the age of 18 must have 2 documented failed treatment episodes as well as approval by a guardian to receive treatment.
Inconvenience. Methadone can be prescribed for opioid dependence only by an accredited treatment program. Patients must therefore travel to the clinic and wait to be evaluated on a daily basis for a minimum of 90 days. Only after they demonstrate consistent responsible behavior and negative results on urine testing do they become eligible to take methadone home.29 If a patient is to travel out of the area during the initial 90 days of treatment, he or she must make arrangements in advance to find a clinic that will provide a “guest dose.”
The inconvenience arising from the regulations may deter some patients from seeking methadone therapy. In spite of this, once patients are started on methadone, more of them continue treatment than with buprenorphine.18 A proposed reason is that methadone is a potent full opioid agonist and therefore relieves withdrawal symptoms and craving more effectively than buprenorphine, which is a partial agonist.30 Another possible reason is the higher level of supervision afforded by methadone clinics, which require daily contact for at least 90 days.
Safety concerns arise from methadone diversion, as illicit use may have lethal consequences. In the past decade, deaths from methadone overdose have risen significantly, most of them due to respiratory depression or torsade de pointes.13 However, most cases of diversion and overdose involve methadone that is prescribed for pain by individual practitioners and not from maintenance programs.13
Advantages of buprenorphine
Together, methadone’s lethality, stigma, and inconvenience may contribute to patients preferring buprenorphine.31
The regulations governing buprenorphine’s use are less restrictive than those with methadone. For example, patients must have a diagnosis of opioid addiction to be prescribed buprenorphine, but they are not required to carry the diagnosis for a year before treatment.31 Additionally, they do not need to travel to a federally approved opioid treatment center daily and can receive buprenorphine directly from a physician in an outpatient setting.
Under the Drug Abuse Treatment Act (DATA) of 2000, any physician can apply for a waiver to prescribe and dispense buprenorphine in his or her office. To qualify for an initial waiver, physicians must either obtain certification in the fields of addiction medicine or addiction psychiatry or complete an approved 8-hour training session.32 Each physician starts with a maximum of 30 patients, but can apply to treat up to 100 patients after 1 year and eventually up to 275 patients. Physicians must document every buprenorphine prescription they write and be able to refer patients for counseling.31
As of February 2017, nurse practitioners and physician assistants can also apply for a DATA 2000 waiver. All waivered providers are subject to unannounced visits from the Drug Enforcement Administration once every 5 years.32
While there are no federal restrictions on the amount of buprenorphine that can be dispensed, some states and some insurance companies have placed restrictions on dose or length of treatment.33 Buprenorphine patients can fill their prescriptions at any pharmacy and are permitted to bring their medication home, which improves access to care. However, office-based outpatient treatment is not without risk, and preventing buprenorphine diversion remains a challenge.34
‘Lending’ buprenorphine is a felony
Addicts have illegally used buprenorphine to self-treat opioid withdrawal, craving, and dependence.35 Its misuse has also been coupled with self-treatment of conditions that include depression and pain.36
A survey found that 83.7% of patients deem buprenorphine diversion to be appropriate; further, most patients said they consider it unethical to withhold prescribed buprenorphine from individuals showing symptoms of withdrawal.34 Physicians who prescribe buprenorphine must inform their patients that even “lending” or giving away their medication is a felony.
Prescribing physicians must also be diligent about monitoring for signs of diversion such as inconsistent urine toxicology screens, “lost” medication, and requests for early refills or escalating doses.37
EVALUATING PATIENTS FOR OPIOID REPLACEMENT THERAPY
In addition to federal regulations, we propose a 4-step approach to evaluate eligibility for opioid replacement therapy based on existing guidelines.37–39
Step 1: History and physical examination
The history should give particular attention to the patient’s cardiac, pulmonary, and hepatic status, with consideration of the risks of any medical comorbidities (eg, bacterial endocarditis, HIV and HCV infection) that might influence treatment.37
It is also essential to evaluate for any contraindications or drug interactions before prescribing methadone or buprenorphine.38
Contraindications to methadone maintenance include40:
- Cor pulmonale
- Methadone hypersensitivity
- Pseudomembranous colitis
- Selegiline use (due to risk of serotonin syndrome)
- Ileum paralyticus.
Contraindications to buprenorphine use include:
- Hypersensitivity to naloxone or buprenorphine
- Impaired liver function (due to the risk of inadvertent overdose associated with slowed metabolism).
Concurrent use of alcohol or illicit benzodiazepines is a relative contraindication to both methadone and buprenorphine due to the risk of respiratory depression and overdose.37 Likewise, avoid coprescribing opioid agonists and benzodiazepines whenever possible. Obtain a complete list of current medications and query a prescription-monitoring database to determine whether any controlled substances are currently prescribed.37
During the physical examination, look for stigmata of intravenous drug use such as track marks or abscesses37 and document any physical findings consistent with intoxication or withdrawal. Patients must be completely detoxed or in withdrawal before beginning buprenorphine induction; premature induction can precipitate withdrawal.38
A discussion of pregnant patients with opioid use disorder is beyond the scope of this paper. However, it is incumbent on the prescriber to inquire whether the client is pregnant or intends to become pregnant and what birth control methods are in place.
Step 2: Assess psychiatric status
Assessment of the patient’s psychiatric status, including an assessment of alcohol and other drug use, will help determine his or her eligibility for opioid agonists.37 To prepare for the need to manage patients with psychiatrically complex issues, it is helpful to develop relationships with addiction specialists and psychiatrists who are familiar with opioid replacement therapy in your area. This will make it easier to collaborate on patients’ care.
Ask all patients directly about suicidal or homicidal ideation. Any patient with active suicidal or homicidal ideation should be assessed for need of immediate hospitalization by a psychiatrist or another qualified mental health professional. Patients with a history of suicidal ideation should be monitored closely by a mental health professional throughout treatment.37
Many if not most patients with opioid use disorder have concurrent psychiatric disorders, and the interplay between these disorders is complex.40,41 Depression, for example, can precede and even precipitate drug use (an observation supporting the “self-medication theory”).42 If the underlying depressive disorder is not addressed, relapse is nearly inevitable.
It has also been shown that both chronic opioid use and withdrawal can exacerbate aversive emotional states. This escalation of symptoms may result from the pharmacologic effects of opioids or from psychosocial sequelae that can arise from chronic opioid use.41 In this situation, maintaining abstinence can lead to resolution of depressive symptoms. As depression and opioid use can occur together, successful treatment requires equal attention to both illnesses.
Other common comorbidities in patients with opioid use disorder include posttraumatic stress disorder, attention deficit hyperactivity disorder, antisocial personality disorder, and concurrent substance abuse disorders.43 The confluence of antisocial personality disorder is particularly important, as patients with antisocial personality disorder display disruptive and maladaptive behaviors.
Identify any psychotropic medication that is prescribed and check carefully for drug interactions. This applies especially to methadone, as many psychiatric medications also prolong the QT interval. Moreover, patients may not be forthcoming about the use of psychiatric medication.
Find out whether the patient is using any other addictive substances, particularly those that affect the central nervous system, as those who use fentanyl, benzodiazepines, or alcohol are at the highest risk of overdose.31 Often the best option for those with concurrent substance use disorders is inpatient detoxification followed by residential rehabilitation care. Either buprenorphine or methadone can then be initiated upon return to an outpatient setting.
Step 3: Assess psychosocial status
To what extent do the patient’s home environment and support systems promote a drug-free lifestyle? Unfortunately, the psychosocial status of many of these patients is fragile, and they may live in areas where illicit drugs are readily available (which can be urban, suburban, or rural), making it difficult to stay substance-free.38
Generally, lifestyle modifications are needed to transform maladaptive behaviors and promote an environment conducive to long-term recovery. Referrals to social services to address housing, vocational needs, and entitlements may be helpful.39
Step 4: Assess readiness to change
According to one model, people go through 5 stages when changing a behavior: precontemplation, contemplation, preparation for action, action, and maintenance.43 In general, the further along the stages a patient is, the more appropriate he or she is for office-based treatment with buprenorphine.39
The level of change can be assessed with tools such as Stages of Change Readiness and Treatment Eagerness Scale (SOCRATES). Use of stage-specific strategies may enhance a patient’s readiness to cease opioid use.43
Precontemplation. Those in the precontemplation stage are not ready to think about changing their behavior.43 They may be unaware of or unwilling to consider the risks associated with their opioid use and resistant to the idea of quitting. Engagement with opioid agonists for individuals in this stage is low and dropout rates are likely high.
Thus, the proper approach for “precontemplators” is to help them develop some ambivalence about their opioid use. One tactic is to involve the patient in a discussion of the personal benefits and risks of opioid use.
Contemplation. Individuals in the contemplation stage have begun to weigh the costs and benefits of opioid use and express ambivalence about it.44 Because the patient is willing to explore the risks of ongoing use and consider the benefits of treatment, the goal in this stage is to elicit a commitment from the individual to seek treatment.
Preparation. The person in this stage moves from thinking about treatment to planning what action to take.45 As the individual prepares to enter treatment, indecision tends to resurface, as well as self-doubt about his or her ability to change. During this stage, it is important for the provider to spell out goals (abstinence) and strategies (eg, counseling, medication) and enhance a sense of self-efficacy.
Action and maintenance. Patients in these stages engage in treatment and employ new strategies to abstain from opioid use. Maintaining these behaviors can be a daily struggle. Expressing confidence in the patient’s ability to abstain from use will support his or her progress. Behavioral interventions such as strategic avoidance of triggers and engagement in alternative activities (eg, support groups, exercise, faith-based practices) will help to maintain abstinence.
A CHRONIC CONDITION
Opioid use disorder, like many chronic illnesses, requires long-term attention to attain successful patient outcomes. The opioid agonists methadone and buprenorphine are the mainstay of treatment for it, conferring benefits such as reducing opioid use and preventing relapse.
Candidates for opioid agonist therapy should undergo a multidisciplinary assessment, including an evaluation on the patient’s readiness to change his or her opioid use.
Patient education should include a discussion of the risks of methadone (eg, respiratory depression, fatal overdose, and QTc prolongation) and buprenorphine (eg hepatotoxicity) and their benefits (eg, controlling craving, decreasing the risk of relapse). Patients should also be educated about overdose and diversion.
Despite the difficulties inherent in treating patients with opioid use disorder, when used appropriately, opioid agonist therapy can be lifesaving for patients struggling with long-term opioid addiction.
Acknowledgment: We thank Katelyn Colosi, BS, and Drs. Susan Wolfe, Dennis Bouffard, and Sinha Shirshendu for their helpful comments.
- Wakeman SE, Pham-Kanter G, Donelan K. Attitudes, practices, and preparedness to care for patients with substance use disorder: results from a survey of general internists. Subst Abus 2016; 37:635–641.
- Samuels EA, Dwyer K, Mello MJ, Baird J, Kellogg AR, Bernstein E. Emergency department-based opioid harm reduction: moving physicians from willing to doing. Acad Emerg Med 2016; 23:455–465.
- Mohlman MK, Tanzman B, Finison K, Pinette M, Jones C. Impact of medication-assisted treatment for opioid addiction on Medicaid expenditures and health services utilization rates in Vermont. J Subst Abuse Treat 2016; 67:9–14.
- American Psychiatric Association. Diagnostic and Statistical Manual of Mental Disorders, Fifth Edition. Arlington, VA, American Psychiatric Association, 2013.
- Center for Behavioral Health Statistics and Quality. Behavioral health trends in the United States: results from the 2014 National Survey on Drug Use and Health. www.samhsa.gov/data. Accessed April 6, 2017.
- Compton WM, Jones CM, Baldwin GT. Relationship between nonmedical prescription-opioid use and heroin use. N Engl J Med 2016; 374:154–163.
- Ruan X, Wyche MQ, Kaye AD. Analyzing the relationship between nonmedical prescription-opioid use and heroin use. J Opioid Manage 2016; 12:11–14.
- Hser YI, Evans E, Grella C, Ling W, Anglin D. Long-term course of opioid addiction. Harv Rev Psychiatry 2015; 23:76–89.
- Nielsen S, Hillhouse M, Mooney L, Ang A, Ling W. Buprenorphine pharmacotherapy and behavioral treatment: comparison of outcomes among prescription opioid users, heroin users and combination users. J Subst Abuse Treat 2015; 48:70–76.
- Moore BA, Fiellin DA, Barry DT, et al. Primary care office-based buprenorphine treatment: comparison of heroin and prescription opioid dependent patients. J Gen Intern Med 2007; 22:527–530.
- Fischer B, Patra J, Cruz MF, Gittins J, Rehm J. Comparing heroin users and prescription opioid users in a Canadian multi-site population of illicit opioid users. Drug Alcohol Rev 2008; 27:625–632.
- Compton WM, Jones CM, Baldwin GT. Relationship between nonmedical prescription-opioid use and heroin use. N Engl J Med 2016; 374:154–163.
- Jones CM, Baldwin GT, Manocchio T, White JO, Mack KA. Trends in methadone distribution for pain treatment, methadone diversion, and overdose deaths—United States, 2002–2014. MMWR Morb Mortal Wkly Rep 2016; 65:667–671.
- Baxter LE Sr, Campbell A, Deshields M, et al. Safe methadone induction and stabilization: report of an expert panel. J Addict Med 2013; 7:377–386.
- Alinejad S, Kazemi T, Zamani N, Hoffman RS, Mehrpour O. A systematic review of the cardiotoxicity of methadone. EXCLI J 2015; 14:577–600.
- Johnson RE, Strain EC, Amass L. Buprenorphine: how to use it right. Drug Alcohol Depend 2003; 70(suppl 2):S59–S77.
- Ling W. Buprenorphine implant for opioid addiction. Pain Manage 2012; 2:345–350.
- Saxon AJ, Hser YI, Woody G, Ling W. Medication-assisted treatment for opioid addiction: methadone and buprenorphine. J Food Drug Anal 2013; 21:S69–S72.
- Kimber J, Larney S, Hickman M, Randall D, Degenhardt L. Mortality risk of opioid substitution therapy with methadone versus buprenorphine: a retrospective cohort study. Lancet Psychiatry 2015; 2:901–908.
- Marteau D, McDonald R, Patel K. The relative risk of fatal poisoning by methadone or buprenorphine within the wider population of England and Wales. BMJ Open 2015; 5: e007629.
- Kintz P. Deaths involving buprenorphine: a compendium of French cases. Forensic Sci Int 2001; 121:65–69.
- Zuin M, Giorgini A, Selmi C, et al. Acute liver and renal failure during treatment with buprenorphine at therapeutic dose. Dig Liver Dis 2009; 41:e8–e10.
- Klein JW. Pharmacotherapy for substance use disorders. Med Clin North Am 2016; 100:891–910.
- Modesto-Lowe V, Van Kirk J. Clinical uses of naltrexone: a review of the evidence. Exp Clin Psychopharmocol 2002; 10:213–227.
- Woody GE. Agonist models for treating persons with substance use disorders. Curr Psychiatry Rep 2014; 16:489.
- Sanders JJ, Roose RJ, Lubrano MC, Lucan SC. Meaning and methadone: patient perceptions of methadone dose and a model to promote adherence to maintenance treatment. J Addict Med 2013; 7:307–313.
- Herget G. Methadone and buprenorphine added to the WHO list of essential medicines. HIV/AIDS Policy Law Rev 2005; 10:23–24.
- Suzuki J, Dodds T. Clinical recommendation of 12-step meeting attendance and discussion regarding disclosure of buprenorphine use among patients in office-based opioid treatment. Subst Abus 2016; 37:31–34.
- Rettig RA, Yarmolinsky A. Federal Regulation of Methadone Treatment. Washington, DC: National Academies Press; 1995.
- Srivastava A, Kahan M, Nader M. Primary care management of opioid use disorders: abstinence, methadone, or buprenorphine-naloxone? Can Fam Physician 2017; 63:200–205.
- Substance Abuse and Mental Health Services Administration. Federal Guidelines for Opioid Treatment Programs. Rockville, MD: Substance Abuse and Mental Health Services Administration; 2015.
- Substance Abuse and Mental Health Services Administration SAMSHA. Buprenorphine waiver management. www.samhsa.gov/medication-assisted-treatment/buprenorphine-waiver-management. Accessed April 6, 2017.
- Mark TL, Lubran R, McCance-Kats EF, Chalk M, Richardson J. Medicaid coverage of medications to treat alcohol and opioid dependence. J Subst Abuse Treat 2015; 55:1–5.
- Johnson B, Richert T. Diversion of methadone and buprenorphine from opioid substitution treatment: the importance of patients’ attitudes and norms. J Subst Abuse Treat 2015; 54:50–55.
- Yokell MA, Zaller ND, Green TC, Rich JD. Buprenorphine and buprenorphine/naloxone diversion, misuse, and illicit use: an international review. Curr Drug Abuse Rev 2011; 4:28–41.
- Schuman-Olivier Z, Albanese M, Nelson SE, et al. Self-treatment: illicit buprenorphine use by opioid-dependent treatment seekers. J Subst Abuse Treat 2010; 39:41–50.
- American Society of Addiction Medicine. National practice guidelines for the use of medications in the treatment of addiction involving opioid use. www.asam.org/docs/default-source/practice-support/guidelines-and-consensus-docs/asam-national-practice-guideline-supplement.pdf. Accessed April 6, 2017.
- McNicholas L. Clinical guidelines for the use of buprenorphine in the treatment of opioid addiction. Rockville, MD: US Department of Health and Human Services, Substance Abuse and Mental Health Service Administration; 2004.
- Center for Substance Abuse Treatment. Clinical guidelines for the use of buprenorphine in the treatment of opioid addiction. Rockville (MD): Substance Abuse and Mental Health Services Administration (US); 2004. (Treatment Improvement Protocol (TIP) Series, No. 40.) www.ncbi.nlm.nih.gov/books/NBK64245. Accessed April 6, 2017.
- Zippel-Schultz B, Specka M, Cimander K, et al. Outcomes of patients in long-term opioid maintenance treatment. Subst Use Misuse 2016; 51:1493–1503.
- Martins SS, Keyes KM, Storr CL, Zhu H, Chilcoat HD. Pathways between nonmedical opioid use/dependence and psychiatric disorders: results from the National Epidemiologic Survey on Alcohol and Related Conditions. Drug Alcohol Depend 2009; 103:16–24.
- Khantzian EJ. The self-medication hypothesis of addictive disorders: focus on heroin and cocaine dependence. Am J Psychiatry 1985; 142:1259–1264.
- Belding MA, Iguchi MY, Lamb RJ, Lakin M, Terry R. Stages and processes of change among polydrug users in methadone maintenance treatment. Drug Alcohol Depend 1995; 39:45–53.
- Peteet JR, Brenner S, Curtiss D, Ferrigno M, Kauffman J. A stage of change approach to addiction in the medical setting. Gen Hosp Psychiatry 1998; 20:267–273.
- Vijay A, Bazazi AR, Yee I, Kamarulzaman A, Altice FL. Treatment readiness, attitudes toward, and experiences with methadone and buprenorphine maintenance therapy among people who inject drugs in Malaysia. J Subst Abuse Treat 2015; 54:29–36.
- Wakeman SE, Pham-Kanter G, Donelan K. Attitudes, practices, and preparedness to care for patients with substance use disorder: results from a survey of general internists. Subst Abus 2016; 37:635–641.
- Samuels EA, Dwyer K, Mello MJ, Baird J, Kellogg AR, Bernstein E. Emergency department-based opioid harm reduction: moving physicians from willing to doing. Acad Emerg Med 2016; 23:455–465.
- Mohlman MK, Tanzman B, Finison K, Pinette M, Jones C. Impact of medication-assisted treatment for opioid addiction on Medicaid expenditures and health services utilization rates in Vermont. J Subst Abuse Treat 2016; 67:9–14.
- American Psychiatric Association. Diagnostic and Statistical Manual of Mental Disorders, Fifth Edition. Arlington, VA, American Psychiatric Association, 2013.
- Center for Behavioral Health Statistics and Quality. Behavioral health trends in the United States: results from the 2014 National Survey on Drug Use and Health. www.samhsa.gov/data. Accessed April 6, 2017.
- Compton WM, Jones CM, Baldwin GT. Relationship between nonmedical prescription-opioid use and heroin use. N Engl J Med 2016; 374:154–163.
- Ruan X, Wyche MQ, Kaye AD. Analyzing the relationship between nonmedical prescription-opioid use and heroin use. J Opioid Manage 2016; 12:11–14.
- Hser YI, Evans E, Grella C, Ling W, Anglin D. Long-term course of opioid addiction. Harv Rev Psychiatry 2015; 23:76–89.
- Nielsen S, Hillhouse M, Mooney L, Ang A, Ling W. Buprenorphine pharmacotherapy and behavioral treatment: comparison of outcomes among prescription opioid users, heroin users and combination users. J Subst Abuse Treat 2015; 48:70–76.
- Moore BA, Fiellin DA, Barry DT, et al. Primary care office-based buprenorphine treatment: comparison of heroin and prescription opioid dependent patients. J Gen Intern Med 2007; 22:527–530.
- Fischer B, Patra J, Cruz MF, Gittins J, Rehm J. Comparing heroin users and prescription opioid users in a Canadian multi-site population of illicit opioid users. Drug Alcohol Rev 2008; 27:625–632.
- Compton WM, Jones CM, Baldwin GT. Relationship between nonmedical prescription-opioid use and heroin use. N Engl J Med 2016; 374:154–163.
- Jones CM, Baldwin GT, Manocchio T, White JO, Mack KA. Trends in methadone distribution for pain treatment, methadone diversion, and overdose deaths—United States, 2002–2014. MMWR Morb Mortal Wkly Rep 2016; 65:667–671.
- Baxter LE Sr, Campbell A, Deshields M, et al. Safe methadone induction and stabilization: report of an expert panel. J Addict Med 2013; 7:377–386.
- Alinejad S, Kazemi T, Zamani N, Hoffman RS, Mehrpour O. A systematic review of the cardiotoxicity of methadone. EXCLI J 2015; 14:577–600.
- Johnson RE, Strain EC, Amass L. Buprenorphine: how to use it right. Drug Alcohol Depend 2003; 70(suppl 2):S59–S77.
- Ling W. Buprenorphine implant for opioid addiction. Pain Manage 2012; 2:345–350.
- Saxon AJ, Hser YI, Woody G, Ling W. Medication-assisted treatment for opioid addiction: methadone and buprenorphine. J Food Drug Anal 2013; 21:S69–S72.
- Kimber J, Larney S, Hickman M, Randall D, Degenhardt L. Mortality risk of opioid substitution therapy with methadone versus buprenorphine: a retrospective cohort study. Lancet Psychiatry 2015; 2:901–908.
- Marteau D, McDonald R, Patel K. The relative risk of fatal poisoning by methadone or buprenorphine within the wider population of England and Wales. BMJ Open 2015; 5: e007629.
- Kintz P. Deaths involving buprenorphine: a compendium of French cases. Forensic Sci Int 2001; 121:65–69.
- Zuin M, Giorgini A, Selmi C, et al. Acute liver and renal failure during treatment with buprenorphine at therapeutic dose. Dig Liver Dis 2009; 41:e8–e10.
- Klein JW. Pharmacotherapy for substance use disorders. Med Clin North Am 2016; 100:891–910.
- Modesto-Lowe V, Van Kirk J. Clinical uses of naltrexone: a review of the evidence. Exp Clin Psychopharmocol 2002; 10:213–227.
- Woody GE. Agonist models for treating persons with substance use disorders. Curr Psychiatry Rep 2014; 16:489.
- Sanders JJ, Roose RJ, Lubrano MC, Lucan SC. Meaning and methadone: patient perceptions of methadone dose and a model to promote adherence to maintenance treatment. J Addict Med 2013; 7:307–313.
- Herget G. Methadone and buprenorphine added to the WHO list of essential medicines. HIV/AIDS Policy Law Rev 2005; 10:23–24.
- Suzuki J, Dodds T. Clinical recommendation of 12-step meeting attendance and discussion regarding disclosure of buprenorphine use among patients in office-based opioid treatment. Subst Abus 2016; 37:31–34.
- Rettig RA, Yarmolinsky A. Federal Regulation of Methadone Treatment. Washington, DC: National Academies Press; 1995.
- Srivastava A, Kahan M, Nader M. Primary care management of opioid use disorders: abstinence, methadone, or buprenorphine-naloxone? Can Fam Physician 2017; 63:200–205.
- Substance Abuse and Mental Health Services Administration. Federal Guidelines for Opioid Treatment Programs. Rockville, MD: Substance Abuse and Mental Health Services Administration; 2015.
- Substance Abuse and Mental Health Services Administration SAMSHA. Buprenorphine waiver management. www.samhsa.gov/medication-assisted-treatment/buprenorphine-waiver-management. Accessed April 6, 2017.
- Mark TL, Lubran R, McCance-Kats EF, Chalk M, Richardson J. Medicaid coverage of medications to treat alcohol and opioid dependence. J Subst Abuse Treat 2015; 55:1–5.
- Johnson B, Richert T. Diversion of methadone and buprenorphine from opioid substitution treatment: the importance of patients’ attitudes and norms. J Subst Abuse Treat 2015; 54:50–55.
- Yokell MA, Zaller ND, Green TC, Rich JD. Buprenorphine and buprenorphine/naloxone diversion, misuse, and illicit use: an international review. Curr Drug Abuse Rev 2011; 4:28–41.
- Schuman-Olivier Z, Albanese M, Nelson SE, et al. Self-treatment: illicit buprenorphine use by opioid-dependent treatment seekers. J Subst Abuse Treat 2010; 39:41–50.
- American Society of Addiction Medicine. National practice guidelines for the use of medications in the treatment of addiction involving opioid use. www.asam.org/docs/default-source/practice-support/guidelines-and-consensus-docs/asam-national-practice-guideline-supplement.pdf. Accessed April 6, 2017.
- McNicholas L. Clinical guidelines for the use of buprenorphine in the treatment of opioid addiction. Rockville, MD: US Department of Health and Human Services, Substance Abuse and Mental Health Service Administration; 2004.
- Center for Substance Abuse Treatment. Clinical guidelines for the use of buprenorphine in the treatment of opioid addiction. Rockville (MD): Substance Abuse and Mental Health Services Administration (US); 2004. (Treatment Improvement Protocol (TIP) Series, No. 40.) www.ncbi.nlm.nih.gov/books/NBK64245. Accessed April 6, 2017.
- Zippel-Schultz B, Specka M, Cimander K, et al. Outcomes of patients in long-term opioid maintenance treatment. Subst Use Misuse 2016; 51:1493–1503.
- Martins SS, Keyes KM, Storr CL, Zhu H, Chilcoat HD. Pathways between nonmedical opioid use/dependence and psychiatric disorders: results from the National Epidemiologic Survey on Alcohol and Related Conditions. Drug Alcohol Depend 2009; 103:16–24.
- Khantzian EJ. The self-medication hypothesis of addictive disorders: focus on heroin and cocaine dependence. Am J Psychiatry 1985; 142:1259–1264.
- Belding MA, Iguchi MY, Lamb RJ, Lakin M, Terry R. Stages and processes of change among polydrug users in methadone maintenance treatment. Drug Alcohol Depend 1995; 39:45–53.
- Peteet JR, Brenner S, Curtiss D, Ferrigno M, Kauffman J. A stage of change approach to addiction in the medical setting. Gen Hosp Psychiatry 1998; 20:267–273.
- Vijay A, Bazazi AR, Yee I, Kamarulzaman A, Altice FL. Treatment readiness, attitudes toward, and experiences with methadone and buprenorphine maintenance therapy among people who inject drugs in Malaysia. J Subst Abuse Treat 2015; 54:29–36.
KEY POINTS
- Opioid use disorder is potentially lethal and has become more prevalent in the United States over the past few decades.
- The opioid agonist methadone and the partial agonist buprenorphine are the currently recommended treatments for patients who need opioid maintenance therapy. However, they carry the risk of adverse effects (eg, respiratory depression, QTc interval prolongation, hepatotoxicity), diversion, and overdose.
- Patients being considered for opioid agonist therapy need a comprehensive assessment including a thorough medical history and physical examination, psychiatric evaluation, psychosocial appraisal, and determination of readiness to change.
- When methadone and buprenorphine are properly prescribed they confer significant benefits, including reduction or elimination of opioid use, reductions in overdose risk, and positive changes in behavior and lifestyle.
A rational approach to opioid use disorder in primary care
As a medical student, I understood that dealing with death was part of the practice of medicine. I was prepared to help my patients face the end of life from disease and old age and had steeled myself against the inevitable losses I would see from trauma and infection. However, I had no sense of the incredible burden that opioid addiction and death from unintentional overdose would one day cause.
MORE DEATHS FROM OVERDOSE THAN FROM MOTOR VEHICLE ACCIDENTS
To highlight the point, unintentional overdose deaths in 2008 exceeded motor vehicle accidents as the leading cause of accidental death in the United States.1 Since then, the problem has only worsened; by 2014 the US Centers for Disease Control and Prevention reported that 78 Americans were dying each day from unintentional opioid overdose.2
Yet the scourge of deaths from opioid overdose is only the most obvious way that opioid use disorder destroys the lives of patients suffering from addiction, as well as their friends and family. Among many other heartaches, opioid use disorder is associated with severely impaired social function, increased rates of hepatitis C and human immunodeficiency virus (HIV) infection, and serious legal consequences and incarceration.3 Sadly, opioid use disorder has torn apart countless families. Addiction may be a brain disease, but its scope of morbidity extends far beyond the individual with the affliction.
PLENTY OF BLAME TO GO AROUND
To some extent, physicians are culpable in propagating this epidemic, and not just in their obvious role as opioid suppliers. To be certain, opioid overprescribing is a tremendous problem; in 2014, more than 240 million prescriptions for opioids were issued, enough for every American adult to have his or her own bottle of pills.4
However, there is plenty of blame to go around in the medical system for the problems of overprescribing and inappropriate opioid use. Among other factors, medical schools have historically failed to teach young physicians how to treat pain or prescribe opioids safely,5 and pain specialists are often inaccessible to primary care providers.6 Additionally, pharmaceutical companies have been found guilty of marketing opioids to prescribers in misleading ways,7 and well-intentioned but misguided campaigns such as the “pain as a fifth vital sign” movement may have inadvertently contributed to opioid overprescribing as well.8
TACKLING THE CHALLENGE
Prescribers need to tackle these challenges by educating themselves about when and how to prescribe opioids for chronic pain. Breaking the cycle of overprescribing can be achieved by learning to prescribe opioids rationally, cautiously, and as part of a comprehensive multimodal pain management plan with a commitment to risk assessment and harm reduction. It also means having an exit strategy at the start of opioid therapy. This must include recognizing problematic opioid use when it occurs and having options to offer patients when opioid use disorder becomes the primary problem.
Recognizing the problem
Physicians are notoriously poor at predicting and detecting the presence of aberrant drug use behaviors and opioid use disorder. For example, in a study of patients clinicians thought were not at risk for misuse of medications, 60% had urine drug tests showing either the presence of illicit drugs or no evidence of the prescribed drug.9
The prevalence of problematic opioid use in patients on chronic opioid therapy for pain has been variably reported in the literature, but one systematic review found that misuse rates ranged from 21% to 29% (95% confidence interval 13%–38%) and addiction rates averaged 8% to 12% (3%–17%).10 These numbers are alarming, and prescribers need to know how to screen for and diagnose opioid addiction when they see it.
Importantly, there is a wide spectrum of opioid misuse behaviors, and the wise prescriber will thoughtfully consider each circumstance before assuming a patient has a substance use disorder. For example, one patient may skip doses and “hoard” unused pills for fear that he or she will run out of medication during a pain flare, while another may use opioids for nonmedical reasons such as to get high. Both examples represent aberrant drug use, but in the first case patient education may sufficiently address the problem, while the second may herald a more dangerous and less correctable problem.
Responding with empathy
Simply recognizing that a problem exists is not enough. Once we identify problematic opioid use, we also need to know how to address it.
Managing opioid misuse behaviors requires empathy, and prescribers should consider a patient’s motivation and emotive response to counsel. For instance, the patient who skips doses and hoards pills may fear that their well-controlled pain will suddenly worsen if their doctor’s opioid prescribing becomes more restrictive as new guidelines are released.
The lesson is that safe opioid prescribing may require a more restrictive approach than was understood in prior years, but rational prescribing also means careful consideration before arbitrarily tapering or discontinuing opioids in a patient who has demonstrated benefit without evidence of harm, even if new guidelines now recommend against starting opioid therapy for similar pain syndromes. For example, the American College of Physicians released a guideline earlier this year that recommended against opioids to treat low back pain, but it did not recommend stopping opioids if patients were already taking them and benefiting from their use.11
Sometimes the best course of action is to discontinue opioid therapy. This decision may trigger a grief-like reaction in some patients and there can be distinct communication challenges during each coping phase.12 The prescriber should frame opioid prescribing discussions on the changing balance of perceived benefits, risks, and harms; in some cases, the treatment may have “failed” or no longer be appropriate, but the patient may still be suffering from pain. Further, the patient may now need help with a newly recognized substance use disorder and may be particularly vulnerable during this time.
The wrong approach, in my opinion, is to discharge the patient from care because of addiction. This approach may seem justified to the provider who feels betrayed by a patient who has used a prescription differently than intended and has thus placed everyone at risk. However, providers should not take it personally; by definition, a patient with addiction has lost control over use of a drug and may have a stronger relationship with the drug than with you. Instead, we should attempt to intervene to protect a patient’s health and chances of survival. It is critical that physicians learn to leverage treatment resources to provide the support patients need to start the long process of recovery. This may involve detoxification and rehabilitation programs, but in many cases opioid agonist therapy also has a role.
Medication-assisted therapy
Medication-assisted therapy with methadone or buprenorphine can be an extremely important part of this process and is a strategy that Modesto-Lowe et al explore in this issue of the Journal.13 As they point out, patients and providers often misunderstand the use of opioid agonists to treat opioid use disorder; many perceive this as merely substituting one form of addiction for another. However, compelling data support this approach. Studies have shown that opioid agonist therapy is associated with decreased illicit opioid use, better retention in substance use treatment programs, reduced hepatitis C and HIV seroconversion, reduced rates of criminal activity and incarceration, decreased overdose risk, and improved survival.14
Opioid agonists are not a cure-all and come with their own challenges, but for many patients they can “create the space” needed to do the real work of recovery—healing their damaged relationships with themselves, their family, and their society.
Providers need to educate themselves regarding the options available and when and how to use them. They should familiarize themselves with methadone and buprenorphine treatment programs in their community. Better yet, with only 8 hours of additional training, primary care physicians can become waivered to prescribe buprenorphine to treat opioid addiction right in the office. Treating addiction is quickly becoming part of primary care, and clinicians in practice can no longer turn a blind eye toward this problem.
- Miniño AM, Murphy SL, Xu J, Kochanek KD. Deaths: final data for 2008. Natl Vital Stat Rep 2011; 59:1–126.
- Rudd RA, Aleshire N, Zibbell JE, Gladden RM. Increases in drug and opioid overdose deaths—United States, 2000–2014. MMWR Morb Mortal Wkly Rep 2016; 64(50–51):1378–1382.
- Hser YI, Evans E, Grella C, Ling W, Anglin D. Long-term course of opioid addiction. Harv Rev Psychiatry 2015; 23:76–89.
- The opioid epidemic: by the numbers. Department of Health and Human Services; 2016 [updated June 2016.] www.hhs.gov/sites/default/files/Factsheet-opioids-061516.pdf. Accessed April 18, 2017.
- Roehr B. US needs new strategy to help 116 million patients in chronic pain. BMJ 2011; 343:d4206.
- Breuer B, Pappagallo M, Tai JY, Portenoy RK. U.S. board-certified pain physician practices: uniformity and census data of their locations. J Pain 2007; 8:244–250.
- Morreale M. Why is the pendulum swinging? The opiate epidemic in the USA. Acad Psychiatry 2016; 40:839–840.
- Hirsch R. The opioid epidemic: It’s time to place blame where it belongs. KevinMD.com. April 6, 2016. http://www.kevinmd.com/blog/2016/04/the-opioid-epidemic-its-time-to-place-blame-where-it-belongs.html. Accessed April 8, 2017.
- Bronstein K, Passik S, Munitz L, Leider H. Can clinicians accurately predict which patients are misusing their medications? J Pain 2011; 12(suppl):P3. Abstract 111.
- Vowles KE, McEntee ML, Julnes PS, Frohe T, Ney JP, van der Goes DN. Rates of opioid misuse, abuse, and addiction in chronic pain: a systematic review and data synthesis. Pain 2015; 156:569–576.
- Qaseem A, Wilt T, McClean R, Forciea MA. Noninvasive treatments for acute, subacute, and chronic low back pain: a clinical practice guideline from the American College of Physicians. Ann Intern Med 2017; 166:514–530.
- Tobin D, Andrews R, Becker W. Prescribing opioids in primary care: safely starting, monitoring, and stopping. Cleve Clin J Med 2016; 83:207–215.
- Modesto-Lowe V, Sweizbin B, Cheplin M, Hoefer G. Use and misuse of opioid agonists in opioid addiction. Cleve Clin J Med 2017; 84:377–384.
- Nielsen S, Larance B, Degenhardt L, Gowing L, Kehler C, Lintzeris N. Opioid agonist treatment for pharmaceutical opioid dependent people. Cochrane Database Syst Rev 2016(5):CD011117.
As a medical student, I understood that dealing with death was part of the practice of medicine. I was prepared to help my patients face the end of life from disease and old age and had steeled myself against the inevitable losses I would see from trauma and infection. However, I had no sense of the incredible burden that opioid addiction and death from unintentional overdose would one day cause.
MORE DEATHS FROM OVERDOSE THAN FROM MOTOR VEHICLE ACCIDENTS
To highlight the point, unintentional overdose deaths in 2008 exceeded motor vehicle accidents as the leading cause of accidental death in the United States.1 Since then, the problem has only worsened; by 2014 the US Centers for Disease Control and Prevention reported that 78 Americans were dying each day from unintentional opioid overdose.2
Yet the scourge of deaths from opioid overdose is only the most obvious way that opioid use disorder destroys the lives of patients suffering from addiction, as well as their friends and family. Among many other heartaches, opioid use disorder is associated with severely impaired social function, increased rates of hepatitis C and human immunodeficiency virus (HIV) infection, and serious legal consequences and incarceration.3 Sadly, opioid use disorder has torn apart countless families. Addiction may be a brain disease, but its scope of morbidity extends far beyond the individual with the affliction.
PLENTY OF BLAME TO GO AROUND
To some extent, physicians are culpable in propagating this epidemic, and not just in their obvious role as opioid suppliers. To be certain, opioid overprescribing is a tremendous problem; in 2014, more than 240 million prescriptions for opioids were issued, enough for every American adult to have his or her own bottle of pills.4
However, there is plenty of blame to go around in the medical system for the problems of overprescribing and inappropriate opioid use. Among other factors, medical schools have historically failed to teach young physicians how to treat pain or prescribe opioids safely,5 and pain specialists are often inaccessible to primary care providers.6 Additionally, pharmaceutical companies have been found guilty of marketing opioids to prescribers in misleading ways,7 and well-intentioned but misguided campaigns such as the “pain as a fifth vital sign” movement may have inadvertently contributed to opioid overprescribing as well.8
TACKLING THE CHALLENGE
Prescribers need to tackle these challenges by educating themselves about when and how to prescribe opioids for chronic pain. Breaking the cycle of overprescribing can be achieved by learning to prescribe opioids rationally, cautiously, and as part of a comprehensive multimodal pain management plan with a commitment to risk assessment and harm reduction. It also means having an exit strategy at the start of opioid therapy. This must include recognizing problematic opioid use when it occurs and having options to offer patients when opioid use disorder becomes the primary problem.
Recognizing the problem
Physicians are notoriously poor at predicting and detecting the presence of aberrant drug use behaviors and opioid use disorder. For example, in a study of patients clinicians thought were not at risk for misuse of medications, 60% had urine drug tests showing either the presence of illicit drugs or no evidence of the prescribed drug.9
The prevalence of problematic opioid use in patients on chronic opioid therapy for pain has been variably reported in the literature, but one systematic review found that misuse rates ranged from 21% to 29% (95% confidence interval 13%–38%) and addiction rates averaged 8% to 12% (3%–17%).10 These numbers are alarming, and prescribers need to know how to screen for and diagnose opioid addiction when they see it.
Importantly, there is a wide spectrum of opioid misuse behaviors, and the wise prescriber will thoughtfully consider each circumstance before assuming a patient has a substance use disorder. For example, one patient may skip doses and “hoard” unused pills for fear that he or she will run out of medication during a pain flare, while another may use opioids for nonmedical reasons such as to get high. Both examples represent aberrant drug use, but in the first case patient education may sufficiently address the problem, while the second may herald a more dangerous and less correctable problem.
Responding with empathy
Simply recognizing that a problem exists is not enough. Once we identify problematic opioid use, we also need to know how to address it.
Managing opioid misuse behaviors requires empathy, and prescribers should consider a patient’s motivation and emotive response to counsel. For instance, the patient who skips doses and hoards pills may fear that their well-controlled pain will suddenly worsen if their doctor’s opioid prescribing becomes more restrictive as new guidelines are released.
The lesson is that safe opioid prescribing may require a more restrictive approach than was understood in prior years, but rational prescribing also means careful consideration before arbitrarily tapering or discontinuing opioids in a patient who has demonstrated benefit without evidence of harm, even if new guidelines now recommend against starting opioid therapy for similar pain syndromes. For example, the American College of Physicians released a guideline earlier this year that recommended against opioids to treat low back pain, but it did not recommend stopping opioids if patients were already taking them and benefiting from their use.11
Sometimes the best course of action is to discontinue opioid therapy. This decision may trigger a grief-like reaction in some patients and there can be distinct communication challenges during each coping phase.12 The prescriber should frame opioid prescribing discussions on the changing balance of perceived benefits, risks, and harms; in some cases, the treatment may have “failed” or no longer be appropriate, but the patient may still be suffering from pain. Further, the patient may now need help with a newly recognized substance use disorder and may be particularly vulnerable during this time.
The wrong approach, in my opinion, is to discharge the patient from care because of addiction. This approach may seem justified to the provider who feels betrayed by a patient who has used a prescription differently than intended and has thus placed everyone at risk. However, providers should not take it personally; by definition, a patient with addiction has lost control over use of a drug and may have a stronger relationship with the drug than with you. Instead, we should attempt to intervene to protect a patient’s health and chances of survival. It is critical that physicians learn to leverage treatment resources to provide the support patients need to start the long process of recovery. This may involve detoxification and rehabilitation programs, but in many cases opioid agonist therapy also has a role.
Medication-assisted therapy
Medication-assisted therapy with methadone or buprenorphine can be an extremely important part of this process and is a strategy that Modesto-Lowe et al explore in this issue of the Journal.13 As they point out, patients and providers often misunderstand the use of opioid agonists to treat opioid use disorder; many perceive this as merely substituting one form of addiction for another. However, compelling data support this approach. Studies have shown that opioid agonist therapy is associated with decreased illicit opioid use, better retention in substance use treatment programs, reduced hepatitis C and HIV seroconversion, reduced rates of criminal activity and incarceration, decreased overdose risk, and improved survival.14
Opioid agonists are not a cure-all and come with their own challenges, but for many patients they can “create the space” needed to do the real work of recovery—healing their damaged relationships with themselves, their family, and their society.
Providers need to educate themselves regarding the options available and when and how to use them. They should familiarize themselves with methadone and buprenorphine treatment programs in their community. Better yet, with only 8 hours of additional training, primary care physicians can become waivered to prescribe buprenorphine to treat opioid addiction right in the office. Treating addiction is quickly becoming part of primary care, and clinicians in practice can no longer turn a blind eye toward this problem.
As a medical student, I understood that dealing with death was part of the practice of medicine. I was prepared to help my patients face the end of life from disease and old age and had steeled myself against the inevitable losses I would see from trauma and infection. However, I had no sense of the incredible burden that opioid addiction and death from unintentional overdose would one day cause.
MORE DEATHS FROM OVERDOSE THAN FROM MOTOR VEHICLE ACCIDENTS
To highlight the point, unintentional overdose deaths in 2008 exceeded motor vehicle accidents as the leading cause of accidental death in the United States.1 Since then, the problem has only worsened; by 2014 the US Centers for Disease Control and Prevention reported that 78 Americans were dying each day from unintentional opioid overdose.2
Yet the scourge of deaths from opioid overdose is only the most obvious way that opioid use disorder destroys the lives of patients suffering from addiction, as well as their friends and family. Among many other heartaches, opioid use disorder is associated with severely impaired social function, increased rates of hepatitis C and human immunodeficiency virus (HIV) infection, and serious legal consequences and incarceration.3 Sadly, opioid use disorder has torn apart countless families. Addiction may be a brain disease, but its scope of morbidity extends far beyond the individual with the affliction.
PLENTY OF BLAME TO GO AROUND
To some extent, physicians are culpable in propagating this epidemic, and not just in their obvious role as opioid suppliers. To be certain, opioid overprescribing is a tremendous problem; in 2014, more than 240 million prescriptions for opioids were issued, enough for every American adult to have his or her own bottle of pills.4
However, there is plenty of blame to go around in the medical system for the problems of overprescribing and inappropriate opioid use. Among other factors, medical schools have historically failed to teach young physicians how to treat pain or prescribe opioids safely,5 and pain specialists are often inaccessible to primary care providers.6 Additionally, pharmaceutical companies have been found guilty of marketing opioids to prescribers in misleading ways,7 and well-intentioned but misguided campaigns such as the “pain as a fifth vital sign” movement may have inadvertently contributed to opioid overprescribing as well.8
TACKLING THE CHALLENGE
Prescribers need to tackle these challenges by educating themselves about when and how to prescribe opioids for chronic pain. Breaking the cycle of overprescribing can be achieved by learning to prescribe opioids rationally, cautiously, and as part of a comprehensive multimodal pain management plan with a commitment to risk assessment and harm reduction. It also means having an exit strategy at the start of opioid therapy. This must include recognizing problematic opioid use when it occurs and having options to offer patients when opioid use disorder becomes the primary problem.
Recognizing the problem
Physicians are notoriously poor at predicting and detecting the presence of aberrant drug use behaviors and opioid use disorder. For example, in a study of patients clinicians thought were not at risk for misuse of medications, 60% had urine drug tests showing either the presence of illicit drugs or no evidence of the prescribed drug.9
The prevalence of problematic opioid use in patients on chronic opioid therapy for pain has been variably reported in the literature, but one systematic review found that misuse rates ranged from 21% to 29% (95% confidence interval 13%–38%) and addiction rates averaged 8% to 12% (3%–17%).10 These numbers are alarming, and prescribers need to know how to screen for and diagnose opioid addiction when they see it.
Importantly, there is a wide spectrum of opioid misuse behaviors, and the wise prescriber will thoughtfully consider each circumstance before assuming a patient has a substance use disorder. For example, one patient may skip doses and “hoard” unused pills for fear that he or she will run out of medication during a pain flare, while another may use opioids for nonmedical reasons such as to get high. Both examples represent aberrant drug use, but in the first case patient education may sufficiently address the problem, while the second may herald a more dangerous and less correctable problem.
Responding with empathy
Simply recognizing that a problem exists is not enough. Once we identify problematic opioid use, we also need to know how to address it.
Managing opioid misuse behaviors requires empathy, and prescribers should consider a patient’s motivation and emotive response to counsel. For instance, the patient who skips doses and hoards pills may fear that their well-controlled pain will suddenly worsen if their doctor’s opioid prescribing becomes more restrictive as new guidelines are released.
The lesson is that safe opioid prescribing may require a more restrictive approach than was understood in prior years, but rational prescribing also means careful consideration before arbitrarily tapering or discontinuing opioids in a patient who has demonstrated benefit without evidence of harm, even if new guidelines now recommend against starting opioid therapy for similar pain syndromes. For example, the American College of Physicians released a guideline earlier this year that recommended against opioids to treat low back pain, but it did not recommend stopping opioids if patients were already taking them and benefiting from their use.11
Sometimes the best course of action is to discontinue opioid therapy. This decision may trigger a grief-like reaction in some patients and there can be distinct communication challenges during each coping phase.12 The prescriber should frame opioid prescribing discussions on the changing balance of perceived benefits, risks, and harms; in some cases, the treatment may have “failed” or no longer be appropriate, but the patient may still be suffering from pain. Further, the patient may now need help with a newly recognized substance use disorder and may be particularly vulnerable during this time.
The wrong approach, in my opinion, is to discharge the patient from care because of addiction. This approach may seem justified to the provider who feels betrayed by a patient who has used a prescription differently than intended and has thus placed everyone at risk. However, providers should not take it personally; by definition, a patient with addiction has lost control over use of a drug and may have a stronger relationship with the drug than with you. Instead, we should attempt to intervene to protect a patient’s health and chances of survival. It is critical that physicians learn to leverage treatment resources to provide the support patients need to start the long process of recovery. This may involve detoxification and rehabilitation programs, but in many cases opioid agonist therapy also has a role.
Medication-assisted therapy
Medication-assisted therapy with methadone or buprenorphine can be an extremely important part of this process and is a strategy that Modesto-Lowe et al explore in this issue of the Journal.13 As they point out, patients and providers often misunderstand the use of opioid agonists to treat opioid use disorder; many perceive this as merely substituting one form of addiction for another. However, compelling data support this approach. Studies have shown that opioid agonist therapy is associated with decreased illicit opioid use, better retention in substance use treatment programs, reduced hepatitis C and HIV seroconversion, reduced rates of criminal activity and incarceration, decreased overdose risk, and improved survival.14
Opioid agonists are not a cure-all and come with their own challenges, but for many patients they can “create the space” needed to do the real work of recovery—healing their damaged relationships with themselves, their family, and their society.
Providers need to educate themselves regarding the options available and when and how to use them. They should familiarize themselves with methadone and buprenorphine treatment programs in their community. Better yet, with only 8 hours of additional training, primary care physicians can become waivered to prescribe buprenorphine to treat opioid addiction right in the office. Treating addiction is quickly becoming part of primary care, and clinicians in practice can no longer turn a blind eye toward this problem.
- Miniño AM, Murphy SL, Xu J, Kochanek KD. Deaths: final data for 2008. Natl Vital Stat Rep 2011; 59:1–126.
- Rudd RA, Aleshire N, Zibbell JE, Gladden RM. Increases in drug and opioid overdose deaths—United States, 2000–2014. MMWR Morb Mortal Wkly Rep 2016; 64(50–51):1378–1382.
- Hser YI, Evans E, Grella C, Ling W, Anglin D. Long-term course of opioid addiction. Harv Rev Psychiatry 2015; 23:76–89.
- The opioid epidemic: by the numbers. Department of Health and Human Services; 2016 [updated June 2016.] www.hhs.gov/sites/default/files/Factsheet-opioids-061516.pdf. Accessed April 18, 2017.
- Roehr B. US needs new strategy to help 116 million patients in chronic pain. BMJ 2011; 343:d4206.
- Breuer B, Pappagallo M, Tai JY, Portenoy RK. U.S. board-certified pain physician practices: uniformity and census data of their locations. J Pain 2007; 8:244–250.
- Morreale M. Why is the pendulum swinging? The opiate epidemic in the USA. Acad Psychiatry 2016; 40:839–840.
- Hirsch R. The opioid epidemic: It’s time to place blame where it belongs. KevinMD.com. April 6, 2016. http://www.kevinmd.com/blog/2016/04/the-opioid-epidemic-its-time-to-place-blame-where-it-belongs.html. Accessed April 8, 2017.
- Bronstein K, Passik S, Munitz L, Leider H. Can clinicians accurately predict which patients are misusing their medications? J Pain 2011; 12(suppl):P3. Abstract 111.
- Vowles KE, McEntee ML, Julnes PS, Frohe T, Ney JP, van der Goes DN. Rates of opioid misuse, abuse, and addiction in chronic pain: a systematic review and data synthesis. Pain 2015; 156:569–576.
- Qaseem A, Wilt T, McClean R, Forciea MA. Noninvasive treatments for acute, subacute, and chronic low back pain: a clinical practice guideline from the American College of Physicians. Ann Intern Med 2017; 166:514–530.
- Tobin D, Andrews R, Becker W. Prescribing opioids in primary care: safely starting, monitoring, and stopping. Cleve Clin J Med 2016; 83:207–215.
- Modesto-Lowe V, Sweizbin B, Cheplin M, Hoefer G. Use and misuse of opioid agonists in opioid addiction. Cleve Clin J Med 2017; 84:377–384.
- Nielsen S, Larance B, Degenhardt L, Gowing L, Kehler C, Lintzeris N. Opioid agonist treatment for pharmaceutical opioid dependent people. Cochrane Database Syst Rev 2016(5):CD011117.
- Miniño AM, Murphy SL, Xu J, Kochanek KD. Deaths: final data for 2008. Natl Vital Stat Rep 2011; 59:1–126.
- Rudd RA, Aleshire N, Zibbell JE, Gladden RM. Increases in drug and opioid overdose deaths—United States, 2000–2014. MMWR Morb Mortal Wkly Rep 2016; 64(50–51):1378–1382.
- Hser YI, Evans E, Grella C, Ling W, Anglin D. Long-term course of opioid addiction. Harv Rev Psychiatry 2015; 23:76–89.
- The opioid epidemic: by the numbers. Department of Health and Human Services; 2016 [updated June 2016.] www.hhs.gov/sites/default/files/Factsheet-opioids-061516.pdf. Accessed April 18, 2017.
- Roehr B. US needs new strategy to help 116 million patients in chronic pain. BMJ 2011; 343:d4206.
- Breuer B, Pappagallo M, Tai JY, Portenoy RK. U.S. board-certified pain physician practices: uniformity and census data of their locations. J Pain 2007; 8:244–250.
- Morreale M. Why is the pendulum swinging? The opiate epidemic in the USA. Acad Psychiatry 2016; 40:839–840.
- Hirsch R. The opioid epidemic: It’s time to place blame where it belongs. KevinMD.com. April 6, 2016. http://www.kevinmd.com/blog/2016/04/the-opioid-epidemic-its-time-to-place-blame-where-it-belongs.html. Accessed April 8, 2017.
- Bronstein K, Passik S, Munitz L, Leider H. Can clinicians accurately predict which patients are misusing their medications? J Pain 2011; 12(suppl):P3. Abstract 111.
- Vowles KE, McEntee ML, Julnes PS, Frohe T, Ney JP, van der Goes DN. Rates of opioid misuse, abuse, and addiction in chronic pain: a systematic review and data synthesis. Pain 2015; 156:569–576.
- Qaseem A, Wilt T, McClean R, Forciea MA. Noninvasive treatments for acute, subacute, and chronic low back pain: a clinical practice guideline from the American College of Physicians. Ann Intern Med 2017; 166:514–530.
- Tobin D, Andrews R, Becker W. Prescribing opioids in primary care: safely starting, monitoring, and stopping. Cleve Clin J Med 2016; 83:207–215.
- Modesto-Lowe V, Sweizbin B, Cheplin M, Hoefer G. Use and misuse of opioid agonists in opioid addiction. Cleve Clin J Med 2017; 84:377–384.
- Nielsen S, Larance B, Degenhardt L, Gowing L, Kehler C, Lintzeris N. Opioid agonist treatment for pharmaceutical opioid dependent people. Cochrane Database Syst Rev 2016(5):CD011117.
Diabetes control during Ramadan fasting
An estimated 50 million patients with diabetes worldwide practice daily fasting during Ramadan, the ninth month of the Islamic calendar, which lasts 29 or 30 days. In the United States, Ramadan begins this year at sundown on Friday, May 26, and ends at sundown on Sunday, June 25.
According to the Multi-Country Retrospective Observational Study of the Management and Outcomes of Patients With Diabetes During Ramadan, conducted in 13 countries, 94.2% of Muslim diabetic patients fasted at least 15 days, and 67.6% of these fasted every day.1
The daily fasting period, which may extend from 14 to 18 hours, starts before sunrise and ends after sunset. The meal taken before sunrise is called Suhur, and the meal after sunset is called Iftar. The fast requires abstaining from eating, drinking, sexual activity, medications, and smoking. For diabetic patients, this poses medical challenges, increasing the risk of acute metabolic complications.
The goal of caring for diabetic patients during Ramadan fasting is to help them to fast without major complications and to empower them to modify their lifestyle in order to achieve this goal.
POSSIBLE METABOLIC COMPLICATIONS
Metabolic complications during Ramadan fasting include hypoglycemia, hyperglycemia, diabetic ketoacidosis, dehydration, and thrombosis.
Hypoglycemia
For patients with type 1 diabetes, fasting increases the risk of hypoglycemia 4.7 times, and the risk is 7.5 times higher for patients with type 2 diabetes.2 However, this is often underreported, as mild to moderate hypoglycemia does not usually require medical assistance.
Precipitating factors include long fasting hours, missing the Suhur meal, and failure to modify drug dosage and timing.
Hyperglycemia
The risk of severe hyperglycemia during fasting is 3.2 times higher in patients with type 1 diabetes and 5 times higher in those with type 2 diabetes.2 Precipitating factors include lack of diet control during the Iftar meal and excessive reduction in the dosage of diabetes medications due to fear of hypoglycemia.
Diabetic ketoacidosis
Ketoacidosis can be precipitated by a lack of diet control during the Iftar meal, excessive reduction in the dosage of insulin due to fear of hypoglycemia, acute stress, and illness or infection.
Dehydration and thrombosis
Patients can become dehydrated during long fasting hours in especially hot weather, by sweating during physical activity, and by osmotic diuresis in poorly controlled diabetes.
Diabetes is a procoagulant condition, and dehydration increases the risk of thrombosis.
OVERALL MANAGEMENT GOALS DURING RAMADAN FASTING
Important aspects of managing diabetes during Ramadan fasting are:
- The pre-Ramadan evaluation and risk stratification
- Promoting patient awareness with Ramadan-focused diabetes education
- Providing instruction on dietary modification
- Modification of the dosage and timing of diabetes medication
- Encouraging frequent monitoring of blood glucose levels
- Advising the patient when to break the fast
- Managing complications.
PRE-RAMADAN MEDICAL EVALUATION AND RISK STRATIFICATION
All diabetic patients who fast during Ramadan should undergo an evaluation 1 or 2 months before the start of Ramadan to determine their level of diabetes control and the presence of acute and chronic complications of diabetes and other comorbid conditions. Also important is to determine the patient’s social circumstances, ie, knowledge about diabetes, socioeconomic factors, religious beliefs, educational status, diabetes self-management skills, and family support in case of hypoglycemia or complications.
The evaluation helps to determine the patient’s risk of diabetes-related complications from Ramadan fasting, which is categorized as very high, high, or moderate/low according to the criteria of the International Diabetes Federation (Table 1).3 Patients should be advised as to the feasibility of fasting based on this risk categorization.
Even though the recommendation is to avoid fasting if the risk is very high or high, many patients fast. But patients should be advised about Islamic regulations exempting people from fasting (Table 2).4
RAMADAN-FOCUSED DIABETES EDUCATION
Improving the patient’s awareness of the risks of Ramadan fasting reduces the chance of complications. Education should include information on diet and exercise, changes in the timing and dosing of medications, signs and symptoms of hypoglycemia and hyperglycemia, the importance of monitoring blood glucose levels on fasting days, and the importance of breaking the fast in case of complications.5
DIET AND EXERCISE
All diabetic patients should be encouraged to remember to eat the predawn meal on fasting days. They should maintain a balanced diet, with complex carbohydrates with slow energy release for the predawn meal and simple carbohydrates for the sunset meal. Foods with a low glycemic index and high fiber content are recommended, and patients should be advised to avoid saturated fats and to drink plenty of fluids between sunset and sunrise to avoid dehydration.6
Diabetic patients can perform their usual physical activity, including moderate exercise, but should avoid excessive physical activity especially toward evening hours to prevent hypoglycemia.
Some patients may decide not to monitor their blood glucose as they believe that pricking the finger for blood sugar testing breaks the fast.7 Patients should be advised that this is a misconception.
ADJUSTING DIABETES MEDICATIONS
Oral diabetes drugs
Drugs such as metformin, alpha glucosidase inhibitors, thiazolidinediones, the short-acting insulin secretagogue nateglinide, dipeptidyl peptidase 4 inhibitors (eg, sitagliptin), and glucagon-like peptide 1 receptor agonists are associated with a lower risk of hypoglycemia and can be used during Ramadan fasting without significant changes in the daily dose (Table 3).8
Sulfonylureas carry a higher risk of hypoglycemia and should be used cautiously during fasting, with appropriate modification in dose and timing.9,10
Sodium-glucose cotransporter 2 inhibitors, when not combined with insulin or sulfonylureas, carry a lower risk of hypoglycemia, but during Ramadan fasting there is an increased risk of dehydration, urinary tract infection, and postural hypotension since fluids cannot be taken during fasting hours.
Dipeptidyl peptidase 4 inhibitors carry a low risk of hypoglycemia and can be used during Ramadan without dosing modification. Glucagon-like peptide 1 agonists also can be used without adjusting the dosage.11
Insulins
Insulin treatment is associated with a higher risk of hypoglycemia during Ramadan fasting.12 During fasting, the risk of hypoglycemia from premixed insulin can be minimized by changing to a multiple-dose regimen involving a basal insulin and short-acting insulin before meals, with adjustment of the short- acting insulin dose based on the anticipated carbohydrate intake for each meal.13
Patients taking premixed insulin preparations consisting of 70% intermediate-acting or long-acting insulin and 30% short-acting insulin should change to a 50/50 preparation during Ramadan fasting to reduce hypoglycemic risk and improve glycemic control; taking more of the fast-acting component controls postprandial hyperglycemia, and taking less of the intermediate or long-acting component minimizes the risk of hypoglycemia during fasting hours.14,15
Insulin analogues carry a lower risk of hypoglycemia than human insulin. Compared with a human insulin 70/30 preparation, an analogue premix containing 75% neutral protamine lispro and 25% insulin lispro resulted in better glycemic control during Ramadan fasting.16 This could be related to the pharmacodynamics of low-ratio premix analogues, as well as to the mealtime flexibility of analogue insulin, as the injections of the 75/25 mix were given immediately before the morning and evening meals. Insulin analogues are also less likely to cause postprandial hypoglycemia.16
A multiple-dose insulin regimen involving a long-acting basal insulin (eg, glargine, detemir, degludec) and a short-acting insulin (eg, glulisine, aspart, lispro) before meals is preferred in view of better glycemic control and lower risk of hypoglycemia.17
Use of an insulin pump during Ramadan is associated with a reduced risk of hypoglycemia.18 In patients with an insulin pump, the rate of basal insulin must be reduced during daytime, and the postprandial bolus of insulin must be increased after breaking the fast.
FREQUENT MONITORING OF BLOOD GLUCOSE DURING FASTING
Frequent monitoring reduces the risk of both hypoglycemia and hyperglycemia and helps control blood sugar levels during Ramadan fasting. As mentioned above, pricking the finger for blood sugar testing during fasting hours does not break the fast, and this should be emphasized during Ramadan-focused diabetes education.
The exact frequency of blood sugar testing is not defined. In patients with well-controlled diabetes without complications, testing once or twice a day is enough. Patients with poorly controlled diabetes and those with complications should test more often.
ADVICE REGARDING WHEN TO BREAK THE FAST
If signs or symptoms of hypoglycemia develop, the patient should break the fast in order to avoid serious complications. This is acceptable under Islamic law.3,19–21
MANAGEMENT OF COMPLICATIONS
Management of diabetic complications in patients during Ramadan fasting is similar to that for other diabetic patients and includes management of hypo- and hyperglycemia, diabetic ketoacidosis, and dehydration.
- Babineaux SM, Toaima D, Boye KS, et al. Multi-country retrospective observational study of the management and outcomes of patients with type 2 diabetes during Ramadan in 2010 (CREED). Diabet Med 2015; 32:819–828.
- Salti I, Benard E, Detournay B, et al; EPIDIAR Study Group. A population-based study of diabetes and its characteristics during the fasting month of Ramadan in 13 countries: results of the Epidemiology of Diabetes and Ramadan 1422/2001 (EPIDIAR) study. Diabetes Care 2004; 27:2306–2311.
- International Diabetes Federation and the DAR International Alliance. Diabetes and Ramadan: Practical Guidelines. Brussels, Belgium: International Diabetes Federation, 2016. www.idf.org/guidelines/diabetes-in-ramadan and www.daralliance.org. Accessed March 8, 2017.
- Al-Arouj M, Bouguerra R, Buse J, et al. Recommendations for management of diabetes during Ramadan. Diabetes Care 2005; 28:2305–2311.
- Masood SN, Masood Y, Hakim R, Alvi SFD, Shera AS. Ramadan fasting related awareness, practices and experiences of a representative group of urban Pakistani Diabetics. Pak J Med Sci 2012; 28:432–436.
- Bravis V, Hui E, Salih S, Mehar S, Hassanein M, Devendra D. Ramadan education and awareness in diabetes (READ) programme for Muslims with type 2 diabetes who fast during Ramadan. Diabet Med 2010; 27:327–331.
- Masood SN, Sheikh MA, Masood Y, Hakeem R, Shera AS. Beliefs of people with diabetes about skin prick during Ramadan fasting. Diabetes Care 2014; 37:e68–e69.
- Aravind S, Ismail SB, Balamurugan R, et al. Hypoglycemia in patients with type 2 diabetes from India and Malaysia treated with sitagliptin or a sulfonylurea during Ramadan: a randomized, pragmatic study. Curr Med Res Opin 2012; 28:1289–1296.
- Glimepiride in Ramadan (GLIRA) Study Group. The efficacy and safety of glimepiride in the management of type 2 diabetes in Muslim patients during Ramadan. Diabetes Care 2005; 28:421–422.
- Hassanein M, Abdallah K, Schweizer A. A double-blind, randomized trial, including frequent patient-physician contacts and Ramadan-focused advice, assessing vildagliptin and gliclazide in patients with type 2 diabetes fasting during Ramadan: the STEADFAST study. Vasc Health Risk Manag 2014; 10:319–326.
- Brady EM, Davies MJ, Gray LJ, et al. A randomized controlled trial comparing the GLP-1 receptor agonist liraglutide to a sulphonylurea as add on to metformin in patients with established type 2 diabetes during Ramadan: the Treat 4 Ramadan trial. Diabetes Obes Metab 2014; 16:527–536.
- Ibrahim M, Abu Al Magd M, Annabi FA, et al. Recommendations for management of diabetes during Ramadan: update 2015. BMJ Open Diabetes Res Care 2015; 3:e000108.
- Kassem HS, Zantout MS, Azar ST. Insulin therapy during Ramadan fast for type 1 diabetes patients. J Endocrinol Invest 2005; 28:802–805.
- Hui E, Bravis V, Salih S, Hassanein M, Devendra D. Comparison of humalog mix 50 with human insulin mix 30 in type 2 diabetes patients during Ramadan. Int J Clin Pract 2010; 64:1095–1099.
- Hassanein M, Belhadj M, Abdallah K, et al. Management of type 2 diabetes in Ramadan: low ratio premix insulin working group practical advice. Indian J Endocrinol Metab 2014; 18:794–799.
- Mattoo V, Milicevic Z, Malone JK, et al; Ramadan Study Group. A comparison of insulin lispro Mix25 and human insulin 30/70 in the treatment of type 2 diabetes during Ramadan. Diabetes Res Clin Pract 2003; 59:137–143.
- Pathan MF, Sahay RK, Zargar AH, et al. South Asian Consensus Guideline: use of insulin in diabetes during Ramadan. Indian J Endocrinol Metab 2012; 16:499–502.
- Khalil AB, Beshyah SA, Abu Awad SM, et al. Ramadan fasting in diabetes patients on insulin pump therapy augmented by continuous glucose monitoring: an observational real-life study. Diabetes Technol Ther 2012; 14:813–818.
- Holy Qur’an 2:195.
- Holy Qur’an 4:29.
- Bashir MI, Pathan MF, Raza SA. Role of oral hypoglycemic agents in the management of type 2 diabetes mellitus during Ramadan. Indian J Endocrinol Metab 2012; 16:503–507.
An estimated 50 million patients with diabetes worldwide practice daily fasting during Ramadan, the ninth month of the Islamic calendar, which lasts 29 or 30 days. In the United States, Ramadan begins this year at sundown on Friday, May 26, and ends at sundown on Sunday, June 25.
According to the Multi-Country Retrospective Observational Study of the Management and Outcomes of Patients With Diabetes During Ramadan, conducted in 13 countries, 94.2% of Muslim diabetic patients fasted at least 15 days, and 67.6% of these fasted every day.1
The daily fasting period, which may extend from 14 to 18 hours, starts before sunrise and ends after sunset. The meal taken before sunrise is called Suhur, and the meal after sunset is called Iftar. The fast requires abstaining from eating, drinking, sexual activity, medications, and smoking. For diabetic patients, this poses medical challenges, increasing the risk of acute metabolic complications.
The goal of caring for diabetic patients during Ramadan fasting is to help them to fast without major complications and to empower them to modify their lifestyle in order to achieve this goal.
POSSIBLE METABOLIC COMPLICATIONS
Metabolic complications during Ramadan fasting include hypoglycemia, hyperglycemia, diabetic ketoacidosis, dehydration, and thrombosis.
Hypoglycemia
For patients with type 1 diabetes, fasting increases the risk of hypoglycemia 4.7 times, and the risk is 7.5 times higher for patients with type 2 diabetes.2 However, this is often underreported, as mild to moderate hypoglycemia does not usually require medical assistance.
Precipitating factors include long fasting hours, missing the Suhur meal, and failure to modify drug dosage and timing.
Hyperglycemia
The risk of severe hyperglycemia during fasting is 3.2 times higher in patients with type 1 diabetes and 5 times higher in those with type 2 diabetes.2 Precipitating factors include lack of diet control during the Iftar meal and excessive reduction in the dosage of diabetes medications due to fear of hypoglycemia.
Diabetic ketoacidosis
Ketoacidosis can be precipitated by a lack of diet control during the Iftar meal, excessive reduction in the dosage of insulin due to fear of hypoglycemia, acute stress, and illness or infection.
Dehydration and thrombosis
Patients can become dehydrated during long fasting hours in especially hot weather, by sweating during physical activity, and by osmotic diuresis in poorly controlled diabetes.
Diabetes is a procoagulant condition, and dehydration increases the risk of thrombosis.
OVERALL MANAGEMENT GOALS DURING RAMADAN FASTING
Important aspects of managing diabetes during Ramadan fasting are:
- The pre-Ramadan evaluation and risk stratification
- Promoting patient awareness with Ramadan-focused diabetes education
- Providing instruction on dietary modification
- Modification of the dosage and timing of diabetes medication
- Encouraging frequent monitoring of blood glucose levels
- Advising the patient when to break the fast
- Managing complications.
PRE-RAMADAN MEDICAL EVALUATION AND RISK STRATIFICATION
All diabetic patients who fast during Ramadan should undergo an evaluation 1 or 2 months before the start of Ramadan to determine their level of diabetes control and the presence of acute and chronic complications of diabetes and other comorbid conditions. Also important is to determine the patient’s social circumstances, ie, knowledge about diabetes, socioeconomic factors, religious beliefs, educational status, diabetes self-management skills, and family support in case of hypoglycemia or complications.
The evaluation helps to determine the patient’s risk of diabetes-related complications from Ramadan fasting, which is categorized as very high, high, or moderate/low according to the criteria of the International Diabetes Federation (Table 1).3 Patients should be advised as to the feasibility of fasting based on this risk categorization.
Even though the recommendation is to avoid fasting if the risk is very high or high, many patients fast. But patients should be advised about Islamic regulations exempting people from fasting (Table 2).4
RAMADAN-FOCUSED DIABETES EDUCATION
Improving the patient’s awareness of the risks of Ramadan fasting reduces the chance of complications. Education should include information on diet and exercise, changes in the timing and dosing of medications, signs and symptoms of hypoglycemia and hyperglycemia, the importance of monitoring blood glucose levels on fasting days, and the importance of breaking the fast in case of complications.5
DIET AND EXERCISE
All diabetic patients should be encouraged to remember to eat the predawn meal on fasting days. They should maintain a balanced diet, with complex carbohydrates with slow energy release for the predawn meal and simple carbohydrates for the sunset meal. Foods with a low glycemic index and high fiber content are recommended, and patients should be advised to avoid saturated fats and to drink plenty of fluids between sunset and sunrise to avoid dehydration.6
Diabetic patients can perform their usual physical activity, including moderate exercise, but should avoid excessive physical activity especially toward evening hours to prevent hypoglycemia.
Some patients may decide not to monitor their blood glucose as they believe that pricking the finger for blood sugar testing breaks the fast.7 Patients should be advised that this is a misconception.
ADJUSTING DIABETES MEDICATIONS
Oral diabetes drugs
Drugs such as metformin, alpha glucosidase inhibitors, thiazolidinediones, the short-acting insulin secretagogue nateglinide, dipeptidyl peptidase 4 inhibitors (eg, sitagliptin), and glucagon-like peptide 1 receptor agonists are associated with a lower risk of hypoglycemia and can be used during Ramadan fasting without significant changes in the daily dose (Table 3).8
Sulfonylureas carry a higher risk of hypoglycemia and should be used cautiously during fasting, with appropriate modification in dose and timing.9,10
Sodium-glucose cotransporter 2 inhibitors, when not combined with insulin or sulfonylureas, carry a lower risk of hypoglycemia, but during Ramadan fasting there is an increased risk of dehydration, urinary tract infection, and postural hypotension since fluids cannot be taken during fasting hours.
Dipeptidyl peptidase 4 inhibitors carry a low risk of hypoglycemia and can be used during Ramadan without dosing modification. Glucagon-like peptide 1 agonists also can be used without adjusting the dosage.11
Insulins
Insulin treatment is associated with a higher risk of hypoglycemia during Ramadan fasting.12 During fasting, the risk of hypoglycemia from premixed insulin can be minimized by changing to a multiple-dose regimen involving a basal insulin and short-acting insulin before meals, with adjustment of the short- acting insulin dose based on the anticipated carbohydrate intake for each meal.13
Patients taking premixed insulin preparations consisting of 70% intermediate-acting or long-acting insulin and 30% short-acting insulin should change to a 50/50 preparation during Ramadan fasting to reduce hypoglycemic risk and improve glycemic control; taking more of the fast-acting component controls postprandial hyperglycemia, and taking less of the intermediate or long-acting component minimizes the risk of hypoglycemia during fasting hours.14,15
Insulin analogues carry a lower risk of hypoglycemia than human insulin. Compared with a human insulin 70/30 preparation, an analogue premix containing 75% neutral protamine lispro and 25% insulin lispro resulted in better glycemic control during Ramadan fasting.16 This could be related to the pharmacodynamics of low-ratio premix analogues, as well as to the mealtime flexibility of analogue insulin, as the injections of the 75/25 mix were given immediately before the morning and evening meals. Insulin analogues are also less likely to cause postprandial hypoglycemia.16
A multiple-dose insulin regimen involving a long-acting basal insulin (eg, glargine, detemir, degludec) and a short-acting insulin (eg, glulisine, aspart, lispro) before meals is preferred in view of better glycemic control and lower risk of hypoglycemia.17
Use of an insulin pump during Ramadan is associated with a reduced risk of hypoglycemia.18 In patients with an insulin pump, the rate of basal insulin must be reduced during daytime, and the postprandial bolus of insulin must be increased after breaking the fast.
FREQUENT MONITORING OF BLOOD GLUCOSE DURING FASTING
Frequent monitoring reduces the risk of both hypoglycemia and hyperglycemia and helps control blood sugar levels during Ramadan fasting. As mentioned above, pricking the finger for blood sugar testing during fasting hours does not break the fast, and this should be emphasized during Ramadan-focused diabetes education.
The exact frequency of blood sugar testing is not defined. In patients with well-controlled diabetes without complications, testing once or twice a day is enough. Patients with poorly controlled diabetes and those with complications should test more often.
ADVICE REGARDING WHEN TO BREAK THE FAST
If signs or symptoms of hypoglycemia develop, the patient should break the fast in order to avoid serious complications. This is acceptable under Islamic law.3,19–21
MANAGEMENT OF COMPLICATIONS
Management of diabetic complications in patients during Ramadan fasting is similar to that for other diabetic patients and includes management of hypo- and hyperglycemia, diabetic ketoacidosis, and dehydration.
An estimated 50 million patients with diabetes worldwide practice daily fasting during Ramadan, the ninth month of the Islamic calendar, which lasts 29 or 30 days. In the United States, Ramadan begins this year at sundown on Friday, May 26, and ends at sundown on Sunday, June 25.
According to the Multi-Country Retrospective Observational Study of the Management and Outcomes of Patients With Diabetes During Ramadan, conducted in 13 countries, 94.2% of Muslim diabetic patients fasted at least 15 days, and 67.6% of these fasted every day.1
The daily fasting period, which may extend from 14 to 18 hours, starts before sunrise and ends after sunset. The meal taken before sunrise is called Suhur, and the meal after sunset is called Iftar. The fast requires abstaining from eating, drinking, sexual activity, medications, and smoking. For diabetic patients, this poses medical challenges, increasing the risk of acute metabolic complications.
The goal of caring for diabetic patients during Ramadan fasting is to help them to fast without major complications and to empower them to modify their lifestyle in order to achieve this goal.
POSSIBLE METABOLIC COMPLICATIONS
Metabolic complications during Ramadan fasting include hypoglycemia, hyperglycemia, diabetic ketoacidosis, dehydration, and thrombosis.
Hypoglycemia
For patients with type 1 diabetes, fasting increases the risk of hypoglycemia 4.7 times, and the risk is 7.5 times higher for patients with type 2 diabetes.2 However, this is often underreported, as mild to moderate hypoglycemia does not usually require medical assistance.
Precipitating factors include long fasting hours, missing the Suhur meal, and failure to modify drug dosage and timing.
Hyperglycemia
The risk of severe hyperglycemia during fasting is 3.2 times higher in patients with type 1 diabetes and 5 times higher in those with type 2 diabetes.2 Precipitating factors include lack of diet control during the Iftar meal and excessive reduction in the dosage of diabetes medications due to fear of hypoglycemia.
Diabetic ketoacidosis
Ketoacidosis can be precipitated by a lack of diet control during the Iftar meal, excessive reduction in the dosage of insulin due to fear of hypoglycemia, acute stress, and illness or infection.
Dehydration and thrombosis
Patients can become dehydrated during long fasting hours in especially hot weather, by sweating during physical activity, and by osmotic diuresis in poorly controlled diabetes.
Diabetes is a procoagulant condition, and dehydration increases the risk of thrombosis.
OVERALL MANAGEMENT GOALS DURING RAMADAN FASTING
Important aspects of managing diabetes during Ramadan fasting are:
- The pre-Ramadan evaluation and risk stratification
- Promoting patient awareness with Ramadan-focused diabetes education
- Providing instruction on dietary modification
- Modification of the dosage and timing of diabetes medication
- Encouraging frequent monitoring of blood glucose levels
- Advising the patient when to break the fast
- Managing complications.
PRE-RAMADAN MEDICAL EVALUATION AND RISK STRATIFICATION
All diabetic patients who fast during Ramadan should undergo an evaluation 1 or 2 months before the start of Ramadan to determine their level of diabetes control and the presence of acute and chronic complications of diabetes and other comorbid conditions. Also important is to determine the patient’s social circumstances, ie, knowledge about diabetes, socioeconomic factors, religious beliefs, educational status, diabetes self-management skills, and family support in case of hypoglycemia or complications.
The evaluation helps to determine the patient’s risk of diabetes-related complications from Ramadan fasting, which is categorized as very high, high, or moderate/low according to the criteria of the International Diabetes Federation (Table 1).3 Patients should be advised as to the feasibility of fasting based on this risk categorization.
Even though the recommendation is to avoid fasting if the risk is very high or high, many patients fast. But patients should be advised about Islamic regulations exempting people from fasting (Table 2).4
RAMADAN-FOCUSED DIABETES EDUCATION
Improving the patient’s awareness of the risks of Ramadan fasting reduces the chance of complications. Education should include information on diet and exercise, changes in the timing and dosing of medications, signs and symptoms of hypoglycemia and hyperglycemia, the importance of monitoring blood glucose levels on fasting days, and the importance of breaking the fast in case of complications.5
DIET AND EXERCISE
All diabetic patients should be encouraged to remember to eat the predawn meal on fasting days. They should maintain a balanced diet, with complex carbohydrates with slow energy release for the predawn meal and simple carbohydrates for the sunset meal. Foods with a low glycemic index and high fiber content are recommended, and patients should be advised to avoid saturated fats and to drink plenty of fluids between sunset and sunrise to avoid dehydration.6
Diabetic patients can perform their usual physical activity, including moderate exercise, but should avoid excessive physical activity especially toward evening hours to prevent hypoglycemia.
Some patients may decide not to monitor their blood glucose as they believe that pricking the finger for blood sugar testing breaks the fast.7 Patients should be advised that this is a misconception.
ADJUSTING DIABETES MEDICATIONS
Oral diabetes drugs
Drugs such as metformin, alpha glucosidase inhibitors, thiazolidinediones, the short-acting insulin secretagogue nateglinide, dipeptidyl peptidase 4 inhibitors (eg, sitagliptin), and glucagon-like peptide 1 receptor agonists are associated with a lower risk of hypoglycemia and can be used during Ramadan fasting without significant changes in the daily dose (Table 3).8
Sulfonylureas carry a higher risk of hypoglycemia and should be used cautiously during fasting, with appropriate modification in dose and timing.9,10
Sodium-glucose cotransporter 2 inhibitors, when not combined with insulin or sulfonylureas, carry a lower risk of hypoglycemia, but during Ramadan fasting there is an increased risk of dehydration, urinary tract infection, and postural hypotension since fluids cannot be taken during fasting hours.
Dipeptidyl peptidase 4 inhibitors carry a low risk of hypoglycemia and can be used during Ramadan without dosing modification. Glucagon-like peptide 1 agonists also can be used without adjusting the dosage.11
Insulins
Insulin treatment is associated with a higher risk of hypoglycemia during Ramadan fasting.12 During fasting, the risk of hypoglycemia from premixed insulin can be minimized by changing to a multiple-dose regimen involving a basal insulin and short-acting insulin before meals, with adjustment of the short- acting insulin dose based on the anticipated carbohydrate intake for each meal.13
Patients taking premixed insulin preparations consisting of 70% intermediate-acting or long-acting insulin and 30% short-acting insulin should change to a 50/50 preparation during Ramadan fasting to reduce hypoglycemic risk and improve glycemic control; taking more of the fast-acting component controls postprandial hyperglycemia, and taking less of the intermediate or long-acting component minimizes the risk of hypoglycemia during fasting hours.14,15
Insulin analogues carry a lower risk of hypoglycemia than human insulin. Compared with a human insulin 70/30 preparation, an analogue premix containing 75% neutral protamine lispro and 25% insulin lispro resulted in better glycemic control during Ramadan fasting.16 This could be related to the pharmacodynamics of low-ratio premix analogues, as well as to the mealtime flexibility of analogue insulin, as the injections of the 75/25 mix were given immediately before the morning and evening meals. Insulin analogues are also less likely to cause postprandial hypoglycemia.16
A multiple-dose insulin regimen involving a long-acting basal insulin (eg, glargine, detemir, degludec) and a short-acting insulin (eg, glulisine, aspart, lispro) before meals is preferred in view of better glycemic control and lower risk of hypoglycemia.17
Use of an insulin pump during Ramadan is associated with a reduced risk of hypoglycemia.18 In patients with an insulin pump, the rate of basal insulin must be reduced during daytime, and the postprandial bolus of insulin must be increased after breaking the fast.
FREQUENT MONITORING OF BLOOD GLUCOSE DURING FASTING
Frequent monitoring reduces the risk of both hypoglycemia and hyperglycemia and helps control blood sugar levels during Ramadan fasting. As mentioned above, pricking the finger for blood sugar testing during fasting hours does not break the fast, and this should be emphasized during Ramadan-focused diabetes education.
The exact frequency of blood sugar testing is not defined. In patients with well-controlled diabetes without complications, testing once or twice a day is enough. Patients with poorly controlled diabetes and those with complications should test more often.
ADVICE REGARDING WHEN TO BREAK THE FAST
If signs or symptoms of hypoglycemia develop, the patient should break the fast in order to avoid serious complications. This is acceptable under Islamic law.3,19–21
MANAGEMENT OF COMPLICATIONS
Management of diabetic complications in patients during Ramadan fasting is similar to that for other diabetic patients and includes management of hypo- and hyperglycemia, diabetic ketoacidosis, and dehydration.
- Babineaux SM, Toaima D, Boye KS, et al. Multi-country retrospective observational study of the management and outcomes of patients with type 2 diabetes during Ramadan in 2010 (CREED). Diabet Med 2015; 32:819–828.
- Salti I, Benard E, Detournay B, et al; EPIDIAR Study Group. A population-based study of diabetes and its characteristics during the fasting month of Ramadan in 13 countries: results of the Epidemiology of Diabetes and Ramadan 1422/2001 (EPIDIAR) study. Diabetes Care 2004; 27:2306–2311.
- International Diabetes Federation and the DAR International Alliance. Diabetes and Ramadan: Practical Guidelines. Brussels, Belgium: International Diabetes Federation, 2016. www.idf.org/guidelines/diabetes-in-ramadan and www.daralliance.org. Accessed March 8, 2017.
- Al-Arouj M, Bouguerra R, Buse J, et al. Recommendations for management of diabetes during Ramadan. Diabetes Care 2005; 28:2305–2311.
- Masood SN, Masood Y, Hakim R, Alvi SFD, Shera AS. Ramadan fasting related awareness, practices and experiences of a representative group of urban Pakistani Diabetics. Pak J Med Sci 2012; 28:432–436.
- Bravis V, Hui E, Salih S, Mehar S, Hassanein M, Devendra D. Ramadan education and awareness in diabetes (READ) programme for Muslims with type 2 diabetes who fast during Ramadan. Diabet Med 2010; 27:327–331.
- Masood SN, Sheikh MA, Masood Y, Hakeem R, Shera AS. Beliefs of people with diabetes about skin prick during Ramadan fasting. Diabetes Care 2014; 37:e68–e69.
- Aravind S, Ismail SB, Balamurugan R, et al. Hypoglycemia in patients with type 2 diabetes from India and Malaysia treated with sitagliptin or a sulfonylurea during Ramadan: a randomized, pragmatic study. Curr Med Res Opin 2012; 28:1289–1296.
- Glimepiride in Ramadan (GLIRA) Study Group. The efficacy and safety of glimepiride in the management of type 2 diabetes in Muslim patients during Ramadan. Diabetes Care 2005; 28:421–422.
- Hassanein M, Abdallah K, Schweizer A. A double-blind, randomized trial, including frequent patient-physician contacts and Ramadan-focused advice, assessing vildagliptin and gliclazide in patients with type 2 diabetes fasting during Ramadan: the STEADFAST study. Vasc Health Risk Manag 2014; 10:319–326.
- Brady EM, Davies MJ, Gray LJ, et al. A randomized controlled trial comparing the GLP-1 receptor agonist liraglutide to a sulphonylurea as add on to metformin in patients with established type 2 diabetes during Ramadan: the Treat 4 Ramadan trial. Diabetes Obes Metab 2014; 16:527–536.
- Ibrahim M, Abu Al Magd M, Annabi FA, et al. Recommendations for management of diabetes during Ramadan: update 2015. BMJ Open Diabetes Res Care 2015; 3:e000108.
- Kassem HS, Zantout MS, Azar ST. Insulin therapy during Ramadan fast for type 1 diabetes patients. J Endocrinol Invest 2005; 28:802–805.
- Hui E, Bravis V, Salih S, Hassanein M, Devendra D. Comparison of humalog mix 50 with human insulin mix 30 in type 2 diabetes patients during Ramadan. Int J Clin Pract 2010; 64:1095–1099.
- Hassanein M, Belhadj M, Abdallah K, et al. Management of type 2 diabetes in Ramadan: low ratio premix insulin working group practical advice. Indian J Endocrinol Metab 2014; 18:794–799.
- Mattoo V, Milicevic Z, Malone JK, et al; Ramadan Study Group. A comparison of insulin lispro Mix25 and human insulin 30/70 in the treatment of type 2 diabetes during Ramadan. Diabetes Res Clin Pract 2003; 59:137–143.
- Pathan MF, Sahay RK, Zargar AH, et al. South Asian Consensus Guideline: use of insulin in diabetes during Ramadan. Indian J Endocrinol Metab 2012; 16:499–502.
- Khalil AB, Beshyah SA, Abu Awad SM, et al. Ramadan fasting in diabetes patients on insulin pump therapy augmented by continuous glucose monitoring: an observational real-life study. Diabetes Technol Ther 2012; 14:813–818.
- Holy Qur’an 2:195.
- Holy Qur’an 4:29.
- Bashir MI, Pathan MF, Raza SA. Role of oral hypoglycemic agents in the management of type 2 diabetes mellitus during Ramadan. Indian J Endocrinol Metab 2012; 16:503–507.
- Babineaux SM, Toaima D, Boye KS, et al. Multi-country retrospective observational study of the management and outcomes of patients with type 2 diabetes during Ramadan in 2010 (CREED). Diabet Med 2015; 32:819–828.
- Salti I, Benard E, Detournay B, et al; EPIDIAR Study Group. A population-based study of diabetes and its characteristics during the fasting month of Ramadan in 13 countries: results of the Epidemiology of Diabetes and Ramadan 1422/2001 (EPIDIAR) study. Diabetes Care 2004; 27:2306–2311.
- International Diabetes Federation and the DAR International Alliance. Diabetes and Ramadan: Practical Guidelines. Brussels, Belgium: International Diabetes Federation, 2016. www.idf.org/guidelines/diabetes-in-ramadan and www.daralliance.org. Accessed March 8, 2017.
- Al-Arouj M, Bouguerra R, Buse J, et al. Recommendations for management of diabetes during Ramadan. Diabetes Care 2005; 28:2305–2311.
- Masood SN, Masood Y, Hakim R, Alvi SFD, Shera AS. Ramadan fasting related awareness, practices and experiences of a representative group of urban Pakistani Diabetics. Pak J Med Sci 2012; 28:432–436.
- Bravis V, Hui E, Salih S, Mehar S, Hassanein M, Devendra D. Ramadan education and awareness in diabetes (READ) programme for Muslims with type 2 diabetes who fast during Ramadan. Diabet Med 2010; 27:327–331.
- Masood SN, Sheikh MA, Masood Y, Hakeem R, Shera AS. Beliefs of people with diabetes about skin prick during Ramadan fasting. Diabetes Care 2014; 37:e68–e69.
- Aravind S, Ismail SB, Balamurugan R, et al. Hypoglycemia in patients with type 2 diabetes from India and Malaysia treated with sitagliptin or a sulfonylurea during Ramadan: a randomized, pragmatic study. Curr Med Res Opin 2012; 28:1289–1296.
- Glimepiride in Ramadan (GLIRA) Study Group. The efficacy and safety of glimepiride in the management of type 2 diabetes in Muslim patients during Ramadan. Diabetes Care 2005; 28:421–422.
- Hassanein M, Abdallah K, Schweizer A. A double-blind, randomized trial, including frequent patient-physician contacts and Ramadan-focused advice, assessing vildagliptin and gliclazide in patients with type 2 diabetes fasting during Ramadan: the STEADFAST study. Vasc Health Risk Manag 2014; 10:319–326.
- Brady EM, Davies MJ, Gray LJ, et al. A randomized controlled trial comparing the GLP-1 receptor agonist liraglutide to a sulphonylurea as add on to metformin in patients with established type 2 diabetes during Ramadan: the Treat 4 Ramadan trial. Diabetes Obes Metab 2014; 16:527–536.
- Ibrahim M, Abu Al Magd M, Annabi FA, et al. Recommendations for management of diabetes during Ramadan: update 2015. BMJ Open Diabetes Res Care 2015; 3:e000108.
- Kassem HS, Zantout MS, Azar ST. Insulin therapy during Ramadan fast for type 1 diabetes patients. J Endocrinol Invest 2005; 28:802–805.
- Hui E, Bravis V, Salih S, Hassanein M, Devendra D. Comparison of humalog mix 50 with human insulin mix 30 in type 2 diabetes patients during Ramadan. Int J Clin Pract 2010; 64:1095–1099.
- Hassanein M, Belhadj M, Abdallah K, et al. Management of type 2 diabetes in Ramadan: low ratio premix insulin working group practical advice. Indian J Endocrinol Metab 2014; 18:794–799.
- Mattoo V, Milicevic Z, Malone JK, et al; Ramadan Study Group. A comparison of insulin lispro Mix25 and human insulin 30/70 in the treatment of type 2 diabetes during Ramadan. Diabetes Res Clin Pract 2003; 59:137–143.
- Pathan MF, Sahay RK, Zargar AH, et al. South Asian Consensus Guideline: use of insulin in diabetes during Ramadan. Indian J Endocrinol Metab 2012; 16:499–502.
- Khalil AB, Beshyah SA, Abu Awad SM, et al. Ramadan fasting in diabetes patients on insulin pump therapy augmented by continuous glucose monitoring: an observational real-life study. Diabetes Technol Ther 2012; 14:813–818.
- Holy Qur’an 2:195.
- Holy Qur’an 4:29.
- Bashir MI, Pathan MF, Raza SA. Role of oral hypoglycemic agents in the management of type 2 diabetes mellitus during Ramadan. Indian J Endocrinol Metab 2012; 16:503–507.
KEY POINTS
- A diabetic patient who develops signs or symptoms of hypoglycemia during Ramadan fasting should break the fast to avoid serious complications.
- Management of complications in diabetic patients during Ramadan is similar to that for nonfasting diabetic patients. Complications include hypo- and hyperglycemia, diabetic ketoacidosis, and dehydration.
- A common misconception among patients is that pricking the finger for blood sugar testing during fasting hours breaks the fast; this should be addressed during Ramadan-focused diabetes education.
Strategies for management of intermittent fasting in patients with diabetes
Islam is the second most common religion in the world, and there are 1.6 billion Muslims, many in areas where diabetes is prevalent. Each year observant Muslims fast during the daylight hours for the holy month of Ramadan. It is estimated that 50 million diabetic people fast between dawn and sundown during Ramadan, and Muslims are not the only group of patients who fast for religious or other reasons. It is important for healthcare providers to guide patients with diabetes in avoiding problems related to prolonged fasting.
In this issue of the Cleveland Clinic Journal of Medicine, Drs. A.V. and Zagar address management of diabetes specifically relating to Ramadan fasting, with considerations that also apply to other diabetic patients who fast for religious or for medical reasons.
Fortunately, we now have antihyperglycemic agents that are unlikely to cause hypoglycemia if used alone or in combination, as long as the regimen does not include insulin or a sulfonylurea. These include:
- Metformin and thiazolidinediones (pioglitazone and rosiglitazone), which improve insulin sensitivity
- Glucagon-like peptide 1 (GLP-1) agonists (exenatide, liraglutide, dulaglutide, and albaglutide), which facilitate insulin release in a glucose-dependent fashion
- Dipeptidyl peptidase 4 inhibitors (sitagliptin, saxagliptin, alogliptin, and linagliptin), which augment endogenous incretin hormones, primarily GLP-1, and also facilitate insulin production in a glucose-dependent fashion
- Alpha glucosidase inhibitors (acarbose and miglitol), which slow carbohydrate absorption.
Introduced in recent years, the sodium-glucose cotransporter 2 (SGLT-2) inhibitors canagliflozin, dapagliflozin, and empagliflozin lower blood glucose by reducing the renal threshold for reabsorption of glucose, coupled with reabsorption of sodium leading to daily urinary excretion of about 200 calories. These agents alone or taken with any of the agents above should not cause hypoglycemia. However, they can lead to dehydration if fasting precludes the intake of water as well as food.
The primary concern during fasting is hypoglycemia when diabetes regimens involve insulin or insulin secretagogues, most commonly sulfonylureas. Long-acting basal insulin should not require adjustment during fasting if the dose is not excessive. The amount and timing of short-acting analogues administered before meals should be adjusted to the timing of meals, and doses should be adjusted proportionally to the anticipated carbohydrate intake. Premixed insulins such as intermediate-acting (protamine suspension) insulin and a short-acting insulin in 70/30, 75/25, or 50/50 ratios should be avoided. They do not lend themselves to changes in timing, and the short-acting component is fixed and cannot be changed for varied intake without changing the intermediate-acting portion, which functions as the basal insulin.
Sulfonylurea doses can be reduced or the larger dose moved to before the evening meal, but these agents still pose a risk of hypoglycemia during fasting hours. And as Drs. A.V. and Zagar state, glimepiride, glipizide, and gliclazide are the only agents in the class that should be considered; glyburide (ie, glibenclamide) poses too great a risk of hypoglycemia. On the other hand, the short-acting secretagogue nateglinide can be used safely before meals without much risk of hypoglycemia.
We have focused primarily on hypoglycemia risk. But if antihyperglycemic agents are halted completely or if the reduction is too severe, patients are at risk for hyperglycemia and even diabetic ketoacidosis. Careful monitoring of blood glucose levels during the fasting period is most important for patients taking agents that can cause hypoglycemia, and patients should be advised to break the fast if dangerously low glycemic levels occur. Similarly, if severe hyperglycemia or ketoacidosis develops, patients should be advised to seek medical advice promptly.
Islam is the second most common religion in the world, and there are 1.6 billion Muslims, many in areas where diabetes is prevalent. Each year observant Muslims fast during the daylight hours for the holy month of Ramadan. It is estimated that 50 million diabetic people fast between dawn and sundown during Ramadan, and Muslims are not the only group of patients who fast for religious or other reasons. It is important for healthcare providers to guide patients with diabetes in avoiding problems related to prolonged fasting.
In this issue of the Cleveland Clinic Journal of Medicine, Drs. A.V. and Zagar address management of diabetes specifically relating to Ramadan fasting, with considerations that also apply to other diabetic patients who fast for religious or for medical reasons.
Fortunately, we now have antihyperglycemic agents that are unlikely to cause hypoglycemia if used alone or in combination, as long as the regimen does not include insulin or a sulfonylurea. These include:
- Metformin and thiazolidinediones (pioglitazone and rosiglitazone), which improve insulin sensitivity
- Glucagon-like peptide 1 (GLP-1) agonists (exenatide, liraglutide, dulaglutide, and albaglutide), which facilitate insulin release in a glucose-dependent fashion
- Dipeptidyl peptidase 4 inhibitors (sitagliptin, saxagliptin, alogliptin, and linagliptin), which augment endogenous incretin hormones, primarily GLP-1, and also facilitate insulin production in a glucose-dependent fashion
- Alpha glucosidase inhibitors (acarbose and miglitol), which slow carbohydrate absorption.
Introduced in recent years, the sodium-glucose cotransporter 2 (SGLT-2) inhibitors canagliflozin, dapagliflozin, and empagliflozin lower blood glucose by reducing the renal threshold for reabsorption of glucose, coupled with reabsorption of sodium leading to daily urinary excretion of about 200 calories. These agents alone or taken with any of the agents above should not cause hypoglycemia. However, they can lead to dehydration if fasting precludes the intake of water as well as food.
The primary concern during fasting is hypoglycemia when diabetes regimens involve insulin or insulin secretagogues, most commonly sulfonylureas. Long-acting basal insulin should not require adjustment during fasting if the dose is not excessive. The amount and timing of short-acting analogues administered before meals should be adjusted to the timing of meals, and doses should be adjusted proportionally to the anticipated carbohydrate intake. Premixed insulins such as intermediate-acting (protamine suspension) insulin and a short-acting insulin in 70/30, 75/25, or 50/50 ratios should be avoided. They do not lend themselves to changes in timing, and the short-acting component is fixed and cannot be changed for varied intake without changing the intermediate-acting portion, which functions as the basal insulin.
Sulfonylurea doses can be reduced or the larger dose moved to before the evening meal, but these agents still pose a risk of hypoglycemia during fasting hours. And as Drs. A.V. and Zagar state, glimepiride, glipizide, and gliclazide are the only agents in the class that should be considered; glyburide (ie, glibenclamide) poses too great a risk of hypoglycemia. On the other hand, the short-acting secretagogue nateglinide can be used safely before meals without much risk of hypoglycemia.
We have focused primarily on hypoglycemia risk. But if antihyperglycemic agents are halted completely or if the reduction is too severe, patients are at risk for hyperglycemia and even diabetic ketoacidosis. Careful monitoring of blood glucose levels during the fasting period is most important for patients taking agents that can cause hypoglycemia, and patients should be advised to break the fast if dangerously low glycemic levels occur. Similarly, if severe hyperglycemia or ketoacidosis develops, patients should be advised to seek medical advice promptly.
Islam is the second most common religion in the world, and there are 1.6 billion Muslims, many in areas where diabetes is prevalent. Each year observant Muslims fast during the daylight hours for the holy month of Ramadan. It is estimated that 50 million diabetic people fast between dawn and sundown during Ramadan, and Muslims are not the only group of patients who fast for religious or other reasons. It is important for healthcare providers to guide patients with diabetes in avoiding problems related to prolonged fasting.
In this issue of the Cleveland Clinic Journal of Medicine, Drs. A.V. and Zagar address management of diabetes specifically relating to Ramadan fasting, with considerations that also apply to other diabetic patients who fast for religious or for medical reasons.
Fortunately, we now have antihyperglycemic agents that are unlikely to cause hypoglycemia if used alone or in combination, as long as the regimen does not include insulin or a sulfonylurea. These include:
- Metformin and thiazolidinediones (pioglitazone and rosiglitazone), which improve insulin sensitivity
- Glucagon-like peptide 1 (GLP-1) agonists (exenatide, liraglutide, dulaglutide, and albaglutide), which facilitate insulin release in a glucose-dependent fashion
- Dipeptidyl peptidase 4 inhibitors (sitagliptin, saxagliptin, alogliptin, and linagliptin), which augment endogenous incretin hormones, primarily GLP-1, and also facilitate insulin production in a glucose-dependent fashion
- Alpha glucosidase inhibitors (acarbose and miglitol), which slow carbohydrate absorption.
Introduced in recent years, the sodium-glucose cotransporter 2 (SGLT-2) inhibitors canagliflozin, dapagliflozin, and empagliflozin lower blood glucose by reducing the renal threshold for reabsorption of glucose, coupled with reabsorption of sodium leading to daily urinary excretion of about 200 calories. These agents alone or taken with any of the agents above should not cause hypoglycemia. However, they can lead to dehydration if fasting precludes the intake of water as well as food.
The primary concern during fasting is hypoglycemia when diabetes regimens involve insulin or insulin secretagogues, most commonly sulfonylureas. Long-acting basal insulin should not require adjustment during fasting if the dose is not excessive. The amount and timing of short-acting analogues administered before meals should be adjusted to the timing of meals, and doses should be adjusted proportionally to the anticipated carbohydrate intake. Premixed insulins such as intermediate-acting (protamine suspension) insulin and a short-acting insulin in 70/30, 75/25, or 50/50 ratios should be avoided. They do not lend themselves to changes in timing, and the short-acting component is fixed and cannot be changed for varied intake without changing the intermediate-acting portion, which functions as the basal insulin.
Sulfonylurea doses can be reduced or the larger dose moved to before the evening meal, but these agents still pose a risk of hypoglycemia during fasting hours. And as Drs. A.V. and Zagar state, glimepiride, glipizide, and gliclazide are the only agents in the class that should be considered; glyburide (ie, glibenclamide) poses too great a risk of hypoglycemia. On the other hand, the short-acting secretagogue nateglinide can be used safely before meals without much risk of hypoglycemia.
We have focused primarily on hypoglycemia risk. But if antihyperglycemic agents are halted completely or if the reduction is too severe, patients are at risk for hyperglycemia and even diabetic ketoacidosis. Careful monitoring of blood glucose levels during the fasting period is most important for patients taking agents that can cause hypoglycemia, and patients should be advised to break the fast if dangerously low glycemic levels occur. Similarly, if severe hyperglycemia or ketoacidosis develops, patients should be advised to seek medical advice promptly.