Infectious Diseases Board Review: Menopause in Women Living With HIV

Article Type
Changed
Wed, 09/02/2020 - 10:12
Display Headline
Infectious Diseases Board Review: Menopause in Women Living With HIV

More than half of the 37.9 million persons living with HIV (PLWH) worldwide are women.1 Between 2010 and 2016, 58% of women living with HIV (WLWH) in the United States were older than 45 years.2 As such, an increasing number of WLWH are entering menopause and living well beyond menopause. Despite this, health care providers expressed a lack of confidence in managing menopause in WLWH, and menopausal symptoms often are not recognized by providers.3 Enhancing our knowledge about menopause in WLWH is important, since the physiologic changes associated with menopause impact short- and long-term quality of life and mortality. 

Test your knowledge of this topic HERE.

Amenorrhea

Menstrual irregularities, including amenorrhea and anovulation, are more frequently found in women of low socioeconomic status, presumably due to associated physical and emotional stress.4 In addition, women with low body mass index (BMI) have decreased serum estradiol levels, which lead to amenorrhea.4,5 Furthermore, low parity and many legal and illegal drugs are associated with amenorrhea, including hormonal contraceptives, opiates, stimulants, antipsychotics, and chemotherapeutic agents.6-8

Because these factors associated with amenorrhea are common in WLWH, it is not surprising that amenorrhea and anovulation are frequently found in this population. However, HIV infection itself also appears to be an independent risk factor for amenorrhea. A recent meta-analysis of 8925 women showed a significant association between HIV status and amenorrhea, even when women with and without HIV had similar rates of substance abuse and smoking and similar socioeconomic status.9 The impact of HIV on an increased frequency of amenorrhea was strongest in women with low BMI. Some, but not all, of the studies included in the meta-analysis found a negative association between CD4 cell count and amenorrhea. In addition, a study comparing amenorrhea frequency within subgroups of WLWH also found a higher rate of amenorrhea in women with lower CD4 cell counts.10

“Prolonged” amenorrhea, defined as amenorrhea lasting 1 year or more, also occurs at a high frequency in WLWH.6 This has made determination of age of menopause extremely challenging, since it is likely that many studies defining menopause are misidentifying “prolonged” amenorrhea as menopause. The Women’s Interagency HIV Study (WIHS), a multicenter observational study of women of similar socioeconomic status living with and without HIV, found that more than 50% of WLWH with “prolonged” amenorrhea had serum follicle-stimulating hormone (FSH) levels in the premenopausal range.8 In a later study from the same cohort, 37% of 660 WLWH with “prolonged” amenorrhea had documented resumption of menses.6

Test your knowledge of this topic HERE.

Age at Menopause

In the United States, the median age of menopause is between 50 and 52 years in middle-class white women.11,12 Earlier menopause has been observed in women who are African American, are nulliparous, have a lower BMI, smoke tobacco, and have more stress, less education, and higher unemployment rates.11,13,14 Because 57% of women diagnosed with HIV in 2018 were African American and many WLWH have other risk factors associated with earlier menopause, studies examining the age of menopause in WLWH need to use a comparator group of women without HIV with similar characteristics and control for these factors to determine the influence of HIV on the age of menopause.

It is also necessary to accurately define menopause. The World Health Organization defines natural menopause as the permanent cessation of menstruation for 12 consecutive months without any obvious pathologic or physiologic causes.15 Most studies have used this definition, and many have found that the age of menopause is earlier in WLWH and is associated with immunosuppression.14,16,17 The Ms Study found that women with CD4 cell counts < 200 cells/μL had an increased risk of amenorrhea lasting at least 12 months, when compared to women with CD4 cell counts ≥ 200 cells/μL. The median age of menopause was 42.5 years in women with CD4 cell counts < 200 cells/μL, 46.0 years in women with CD4 cell counts between 200 cells/μL and 500 cells/μL, and 46.5 years in women with CD4 cell counts > 500 cells/μL.14 Similarly, in a cohort of 667 Brazilian WLWH, among whom 160 were postmenopausal, Calvet and colleagues found that 33% of women with CD4 cell counts < 50 cells/μL had premature menopause, as compared to 8% of women with CD4 cell counts ≥ 350 cells/μL.17 In De Pommerol and colleague’s study of 404 WLWH, among whom 69 were found to be postmenopausal, women with CD4 cell counts < 200 cells/μL were more likely to have premature menopause, as compared to women with CD4 cell counts ≥ 350 cells/μL.16

Despite these findings, given the data from WIHS showing that many women with amenorrhea for at least 12 consecutive months had FSH levels in the premenopausal range8 and that 37% of WLWH have resumption of menses after 12 consecutive months of amenorrhea,6 it is probable that the conclusions about the age of menopause in WLWH are invalid, since many of the participants likely had prolonged amenorrhea, not menopause. WIHS found no significant difference in the median age of menopause when WLWH were compared to women without HIV. The median age of menopause was 47.7 years in WLWH and 48.0 years in women without HIV.18

Menopause-Associated Symptoms

The perimenopausal period, which begins, on average, 4 years prior to the final menstrual period, is characterized by hormonal fluctuations leading to irregular menstrual cycles.19,20 Symptoms associated with these physiologic changes during the perimenopausal period include vasomotor symptoms (hot flashes), genitourinary symptoms (vaginal dryness and dyspareunia), anxiety, depression, sleep disturbances, and joint aches.21,22 Such menopausal symptoms can be distressing and negatively impact quality of life.23 In WLWH, severe menopausal symptoms have been associated with suboptimal adherence to antiretroviral therapy (ART).24 

It can be difficult to determine which symptoms are caused by the physiologic changes of menopause in WLWH, as these women have multiple potential reasons for these symptoms, such as ART, comorbidities, and HIV infection itself.25 However, several studies show that there are symptoms that occur more commonly in the perimenopausal period and that WLWH experience these symptoms earlier and with greater intensity.26-30 In addition, the burden of commonly reported HIV symptoms, such as fatigue and muscle aches/joint pains, is higher in women after menopause, suggesting this burden may be exacerbated by menopause.31

Vasomotor

In the United States, the most common symptom during perimenopause is hot flashes, which occur in 38% to 80% of women.32,33 Vasomotor symptoms are most common in women who smoke, use illicit substances, have a high BMI, are of lower socioeconomic status, and are African American.11 As expected, prior studies focusing on hot flash prevalence among premenopausal, perimenopausal, and postmenopausal WLWH found that postmenopausal women experience more hot flashes than premenopausal or perimenopausal women.27,28 In addition, a comparison of women with and without HIV demonstrated a higher prevalence of hot flashes among WLWH.26,29 Vasomotor symptoms can be severely distressing, with hot flashes contributing to increased risk of depression.25,34 In a cross-sectional analysis of 835 WLWH and 335 women without HIV from the WIHS cohort, persistent vasomotor symptoms predicted elevated depressive symptoms in both WLWH and women without HIV.34 In a similar cross-sectional analysis of 536 women, among whom 54% were WLWH and 37% were perimenopausal, psychological symptoms were prevalent in 61% of the women with vasomotor symptoms.29

Genitourinary

Estrogen deficiency, which accompanies the perimenopausal period, leads to vulvovaginal atrophy (VVA), manifesting with symptoms of vaginal dryness, itching, burning, urinary urgency, and dyspareunia (painful intercourse).33,35,36 Unlike vasomotor symptoms, which diminish with time, genitourinary symptoms generally worsen if left untreated.37 Furthermore, these symptoms are often underreported and underdiagnosed.38,39 VVA was found in 43% to 84% of postmenopausal women.36,40,41 In the AGATA study, the prevalence of VVA was associated with years since menopause. 36 Vaginal dryness and dyspareunia were common.

Genitourinary symptoms are most common among women who are African American, have an increased BMI, are of lower socioeconomic status, use tobacco, have a prior history of pelvic inflammatory disease, and have anxiety and depression.11,42,43 Similar to hot flashes, many of these predisposing factors are more common in WLWH. Fantry and colleagues found that 49.6% of WLWH had vaginal dryness.27 Although 56% of postmenopausal women and 36% of perimenopausal women complained of vaginal dryness, in a multivariate analysis only cocaine use, which can decrease estradiol levels,44 was associated with a higher frequency of vaginal dryness.27

Dyspareunia is also common among WLWH. In a cross-sectional study of 178 women without HIV and 128 WLWH between 40 and 60 years of age, Valadares et al found a high prevalence of dyspareunia in WLWH: 41.8%.45 However, this was not significantly higher than the prevalence of dyspareunia in women without HIV: 34.8%.45 HIV infection itself was not associated with the presence of dyspareunia.

Psychiatric

Anxiety and depression are also common symptoms in perimenopausal women.46-48 Studies have shown that depression is diagnosed 2.5 times more frequently among perimenopausal women than premenopausal women.48 In a study by Miller et al that focused on 536 WLWH, among whom 37% were perimenopausal, 89% reported psychological symptoms.29 Ferreira et al found that perimenopausal WLWH had an increased incidence of psychological symptoms, such as depression and anxiety, compared to women without HIV infection.26 Whether this increased prevalence of psychological symptoms seen in WLWH can be attributed to menopause is unclear, since one third to one half of men and women living with HIV experience symptoms of depression.49 However, in the WIHS, which compared findings from 835 WLWH to findings from 335 women without HIV from all menopausal stages, elevated depressive symptoms were seen in the early perimenopausal period.34 There was no increased incidence of such symptoms during the premenopausal or postmenopausal stage, suggesting that factors related to menopause contribute to depressive symptoms during the perimenopausal stage.34

Persistent menopausal symptoms, especially hot flashes, also predicted elevated depressive symptoms in several studies, suggesting the importance of appropriately identifying and treating menopausal symptoms.29,34 In addition, cognitive decline associated with menopause contributes to depression.50,51

Other Symptoms

Sleep disturbances are common among perimenopausal women, with an estimated prevalence between 38% and 46%.52-54 Hot flashes, anxiety, and depression appear to be factors that contribute to sleep difficulty.52-54 In a cross-sectional study of 273 WLWH and 264 women without HIV between 40 and 60 years of age, insomnia was found in 51% of perimenopausal and 53% of postmenopausal WLWH. The prevalence of insomnia in WLWH and women without HIV was the same.55 Joint aches are also commonly reported in the perimenopausal period, with a prevalence as high as 50% to 60% among perimenopausal women in the United States.22,29 Miller and colleagues found that 63% of menopausal WLWH reported arthralgia.29

Test your knowledge of this topic HERE.

Treatment

Despite the increased severity of menopausal symptoms experienced among WLWH, menopausal replacement therapy (MRT) is used less frequently in WLWH than in  women living without HIV.55 Topical treatment is recommended for women who are experiencing vaginal dryness. First-line treatment is topical nonhormonal therapy, such as moisturizers and lubricants.56 If symptoms are not relieved, then topical vaginal estrogen therapy is recommended.56 Randomized placebo-controlled studies have verified the safety and efficacy of topical estrogen in the general population, and there is no reason to expect different outcomes in WLWH.57,58 

For women experiencing severe hot flashes and vaginal dryness, short-term oral MRT is indicated.56 MRT should be limited to the shortest period of time at the lowest effective dose needed to address these symptoms, as MRT is associated with increased risks of breast cancer, cardiovascular disease, and thromboembolism and increased morbidity.56 Drug interactions between MRT and ART are of concern for non-nucleoside reverse transcriptase inhibitors (NNRTIs), protease inhibitors (PIs), and cobicistat, as these ARTs and MRT are metabolized by the CYP3A4 system.59 With any PI, there is potential for an increase or decrease in estradiol or conjugated estrogen levels; an increase in medroxyprogesterone and micronized progesterone levels; and an increase in drospirenone levels. With doravirine and rilpivirine, there is no change in expected hormonal concentrations, but with other NNRTIs (efavirenz, etravirine, and nevirapine) there is a possibility of a decrease in estradiol, conjugated estrogen, medroxyprogesterone, micronized progesterone, and drospirenone levels. None of the integrase strand transfer inhibitors alone leads to changes in hormone level, but elvitegravir is only used when co-formulated with cobicistat, which may lead to increased or decreased estrogen, progesterone, and drospirenone levels.60 Since all of these drug interactions are uncertain, and even act in varying directions, clinicians should monitor menopausal symptoms and titrate MRT to the dose that achieves relief of menopausal symptoms. 

Cardiovascular Risk

Estrogen deficiency that occurs during menopause leads to an increased risk of cardiovascular disease, particularly with changes in lipid profiles, insulin resistance, and body composition (eg, increased fat mass and waist circumference).61 HIV infection also is associated with a higher risk of cardiovascular disease, with studies consistently reporting a 1.5- to 2-fold increase in the rate of cardiovascular events in PLWH compared to persons without HIV.62 The inflammatory effects of HIV as well as ART exposure, specifically to PIs and abacavir, increase the risk for cardiovascular disease.62 In addition, traditional risk factors, including dyslipidemia, contribute to cardiovascular disease risk in this population.63,64 

The increased risk for cardiovascular disease seen in HIV infection is likely compounded with the increased risk associated with menopause. Postmenopausal WLWH appear to be at higher risk of cardiovascular disease compared to postmenopausal women without HIV. Modifiable risk factors for cardiovascular disease, such as decreased fitness and smoking, are more commonly seen in WLWH.65 Even prior to menopause, WLWH experience lipodystrophy syndrome, with increased truncal visceral adiposity and decreased subcutaneous fat and muscle mass.65,66 Microbial translocation due to HIV-related damage of the intestinal mucosa can lead to elevated levels of lipopolysaccharides, a component of the cell wall of gram-negative bacteria; this subsequently activates monocytes, macrophages, and
T cells. In a study that compared postmenopausal WLWH to age-matched women without HIV, this HIV-related immune activation was correlated with an increase in biomarkers of cardiovascular disease, suggesting WLWH are at higher risk of developing cardiovascular disease.67 Similarly, when comparing sex hormone concentrations in premenopausal WLWH and women without HIV, WLWH had lower estrogen and androgen levels, both of which are linked to carotid artery stiffness.68

In addition, postmenopausal WLWH are at higher risk of cardiovascular disease compared to premenopausal WLWH. WLWH with reduced ovarian reserve had increased subclinical coronary atherosclerotic plaque compared to premenopausal WLWH, even when controlling for cardiovascular disease risk factors.69

In summary, cardiovascular disease risk is increased in postmenopausal WLWH.69 Appropriate measures, such as lipid control, antiplatelet therapy, smoking cessation, aerobic exercise, and other lifestyle changes, should be initiated in WLWH as in any other population. 

 

Test your knowledge of this topic HERE.

Osteoporosis

Menopause, with its associated estrogen deficiency, is the most important risk factor linked to increased bone turnover and bone loss.70 In addition, HIV is associated with bone loss, with low bone mineral density (BMD) described even among men and premenopausal women with HIV infection.71 Although decreased BMD associated with HIV stabilizes or even improves after initiation of ART in the younger population,72-74 chronic inflammation caused by HIV stimulates osteoclast differentiation and resorption.71 Other factors that appear to contribute to decreased BMD among PLWH include ART; vitamin D deficiency; low BMI; poor nutrition; inactivity; use of tobacco, alcohol, and illicit drugs; hepatitis B and C coinfection; and frailty, defined as increased vulnerability to stresses related to aging.72-80 Among ARTs, tenofovir disoproxil fumarate is associated with an increased risk of osteoporosis, and switching from this agent to tenofovir alafenamide improves bone density.81 Prolonged amenorrhea is also an added risk factor for osteoporosis in WLWH.82

Once WLWH enter menopause, they have higher rates of osteoporosis and bone loss compared to women without HIV.83 Among postmenopausal WLWH, those taking ritonavir were found to have increased differentiation of osteoclast cells and increased bone loss.84 Similarly, methadone use in postmenopausal women has been associated with increased declines in BMD.85 African-American postmenopausal WLWH appear to be at the greatest risk for bone loss.86

Given the evidence of low BMD and increased fracture risk that occurs during menopause among women living without HIV, and the additional bone loss observed in PLWH, current guidelines recommend screening postmenopausal women ≥ 50 years of age with dual-energy X-ray absorptiometry (DEXA) scan.87 Preventive therapy, such as smoking cessation, adequate nutrition, alcohol reduction, and weight-bearing exercises, should be discussed and recommended to all menopausal WLWH.88 Adequate calcium and vitamin D intake should be discussed as well, with current evidence indicating that low-dose vitamin D supplementation at 1000 IU is as effective as high-dose vitamin D supplementation at 3000 IU in increasing BMD.89 If the DEXA scan shows a T-score < –2.5 at the femoral neck or spine, or between –1 and –2.5 with a 10-year probability of hip fracture ≥ 3% or a 10-year probability of any osteoporosis-related fracture ≥ 20%, bisphosphonates or other medical therapy should be considered. Although the data are limited in WLWH, bisphosphonates have been shown to be effective in improving BMD.90

Cognition

Both men and women living with HIV are at higher risk for cognitive impairment, ranging from minor cognitive-motor disorder to HIV-associated dementia.91 In addition, the menopause transition is characterized by cognitive changes, such as memory loss and difficulty concentrating.92,93 Studies focusing on the effects of both HIV infection and menopause on cognition have been limited thus far. A cross-sectional study demonstrated that HIV infection, but not menopausal stage, was associated with worse performance on cognitive measures.94 While menopausal stage was not associated with cognitive decline, menopausal symptoms like depression, anxiety, and vasomotor symptoms were associated with lower cognitive performance, highlighting the importance of recognition and treatment of menopausal symptoms.94

Test your knowledge of this topic HERE.

Cervical Dysplasia

WLWH are at increased risk for low- and high-grade squamous intraepithelial lesions (SILs) and more rapid progression to cervical carcinoma, as compared to women without HIV.95 This increased risk of cervical disease is associated with age, human papillomavirus genotype, and degree of immunosuppression.96 In addition, menopause appears to affect the risk of cervical disease. Postmenopausal WLWH had a higher risk of progression of SILs and persistence of lower-grade SILs compared to premenopausal women.97,98 Although studies on progression to cervical cancer in postmenopausal WLWH remain limited, current data suggest that postmenopausal WLWH should continue to be monitored and screened similarly to premenopausal women. 

HIV Acquisition and Transmission

Women aged 50 years and older are primarily exposed to HIV through heterosexual contact.99 While the lack of awareness of HIV risk and less frequent use of barrier protection can contribute to new HIV infection in older women, physiologic changes associated with menopause also may be playing a role.100 Vaginal wall thinning and immunologic changes of the cervix that occur during menopause may serve as a risk factor for HIV acquisition. The cervicovaginal mucosa of postmenopausal women had higher levels of p24 antigen after ex vivo HIV-1 infection, suggesting higher susceptibility to acquire HIV infection.101 Postmenopausal women have been shown to have increased cervical CCR5 expression, which serves as an entry point of HIV into target cells.102 Finally, anti-HIV-1 activity was significantly decreased in postmenopausal women compared to premenopausal women.103 In addition, ex vivo studies demonstrated reduced tenofovir disoproxil fumarate and emtricitabine triphosphate concentrations in cervical tissue of postmenopausal women, suggesting that postmenopausal women may need higher doses of pre-exposure prophylaxis to achieve protective efficacy.104 

In contrast, although data are limited, postmenopausal WLWH do not appear to be at increased risk of vaginally transmitting HIV. The intensity of HIV shedding did not differ between premenopausal or postmenopausal women.105 There was a high prevalence of low-level HIV RNA in genital secretions among perimenopausal WLWH, suggesting WLWH in menopause do not present a major public health risk for HIV transmission.106

HIV Progression

With prior data suggesting that younger persons experience better immunologic and virologic responses to ART,107-109 it had previously been hypothesized that virologic and immunologic responses to ART will decline once WLWH reach menopause. However, current studies suggest that menopause does not affect the progression of HIV and that ART-naive women should respond to ART, regardless of their menopausal status. Treatment responses to ART, determined by the median changes in CD4 cell counts and percentages and viral load, in ART-naive individuals did not differ between premenopausal and postmenopausal women.110 In addition, there appear to be no significant changes in CD4 cell counts as WLWH progress through menopause.111

Conclusion

As individuals with HIV infection live longer, an increasing number of women will enter menopause and live many years beyond menopause. WLWH experience earlier and more severe menopausal symptoms, but evidence on the appropriate management of these symptoms is still lacking. These conditions require proper surveillance, and can be prevented with an improved understanding of the effects of menopause on WLWH. However, there remain significant gaps in our understanding of menopause in WLWH. As practitioners encounter an increasing number of perimenopausal and postmenopausal WLWH, studies of the effects of HIV on comorbidities and symptoms of menopause and their appropriate management are necessary to improve care of WLWH.

Test your knowledge of this topic HERE.

References

1. UNAIDS. AIDSInfo. 2019 estimates https://www.unaids.org/en/resources/infographics/girls-and-women-living-with-HIV. Accessed April 30, 2020.

2. Centers for Disease Control and Prevention. Estimated HIV incidence and prevalence in the United States, 2010–2016. HIV Surveillance Supplemental Report 2019;24(No. 1). www.cdc.gov/hiv/library/reports/hiv-surveillance.html. Accessed April 30, 2020.

3. Chirwa M, Ma R, Guallar C, et al. Managing menopause in women living with HIV: A survey of primary care practitioners. Post Reprod Health. 2017;23:111-115.

4. Munster K, Helm P, Schmidt L. Secondary amenorrhea: Prevalence and medical contract–A cross sectional study from a Danish county. Br J Obstet Gynecol. 1992;99:430-433.

5. Vyver E, Steinegger C, Katzman DK, et al. Eating disorders and menstrual dysfunction in adolescents. Ann N Y Acad Sci. 2008;1135: 253-264.

6. Cejtin HE, Evans CT, Greenblatt R, et al. Prolonged amenorrhea and resumption of menses in women with HIV. J Womens Health (Larchmt). 2018;27:1441‐1448.

7. Bai J, Greenwald E, Caterini H, et al. Drug-related menstrual aberrations. Obstet Gynecol. 1974;44:713-719.

8. Cejtin HE, Kalinowski A, Bacchetti P. Effects of human immunodeficiency virus on protracted amenorrhea and ovarian dysfunction. Obstet Gynecol. 2006;108:1423-1431.

9. King EM, Albert AY, Murray MCM. HIV and amenorrhea: a meta-analysis. AIDS. 2019;33:483‐491.

10. Watts DH, Spino C, Zaborski L. Comparison of gynecologic history and laboratory results in HIV-positive women with CDR+ lymphocyte counts between 200 and 500 cells/μl and below 100 cells/ μl. J Acquir Immune Defic Syndr Hum Retrovirol. 1999;20:455-462.

11. Gold EB, Crawford SL, Avis NE, et al. Factors related to age at natural menopause: longitudinal analyses from SWAN. Am J Epidemiol. 2013;178:70-83.

12. Thomas F, Renaud F, Benefice E, et al. International variability of ages at menarche and menopause: patterns and main determinants. Hum Biol. 2001;73:271-290.

13. Bromberger JT, Matthews KA, Kuller LH, et al. Prospective study of the determinants of age at menopause. Am J Epidemiol. 1997; 145:24-33.

14. Schoenbaum E, Hartel D, Lo Y, et al. HIV infection, drug use, and onset of natural menopause. Clinical Infect Dis. 2005;41: 1517-1524.

15. Research on the menopause in the 1990s. Report of a WHO scientific group. World Health Organ Tech Rep Ser. 1996;866:1-107.

16. de Pommerol M, Hessamfar M, Lawson-Ayayi S, et al. Menopause and HIV infection: age at onset and associated factors, ANRS CO3 Aquitaine cohort. Int J STD AIDS. 2011;22:67-72.

17. Calvet G, Grinsztejn G. Predictors of early menopause in HIV infected women: a prospective cohort study. Am J Obstet Gynecol. 2015;212:765.

18. Cejtin SH, Taylor R, Watts DH. Assessment of menopausal status among women in the Women’s Interagency HIV study (WIHS). Proceedings of the XV International AIDS Conference; July 11-16, 2004; Bangkok, Thailand.

19. Taffe JR, Dennerstein L. Menstrual patterns leading to the final menstrual period. Menopause. 2002;9:32-40.

20. Miro F, Parker SW, Aspinall LJ, et al. Origins and consequences of the elongation of the human menstrual cycle during the menopausal transition: the FREEDOM Study. J Clin Endocrinol Metab. 2004;89:4910-4915.

21. McKinlay SM, Brambilla DJ, Posner JG. The normal menopause transition. Maturitas. 1992;14:103-115.

22. Blümel JE, Chedraui P, Baron G, et al. Menopause could be involved in the pathogenesis of muscle and joint aches in mid-aged women. Maturitas. 2013;75:94-100.

23. Woods NF, Mitchell ES. Symptoms interference with work and relationships during the menopausal transition and early postmenopause: observations from the Seattle Midlife Women’s Health Study. Menopause. 2011;18:654-661.

24. Duff PK, Money DM, Ogilvie GS, et al. Severe menopausal symptoms associated with reduced adherence to antiretroviral therapy among perimenopausal and menopausal women living with HIV in Metro Vancouver. Menopause. 2018;25:531-537.

25.  Johnson TM, Cohen HW, Howard AA, et al. Attribution of menopause symptoms in human immunodeficiency virus–infected or at-risk drug-using women. Menopause. 2008;15:551-557.

26. Ferreira CE, Pinto-Neto AM, Conde DM, et al. Menopausal symptoms in women infected with HIV: prevalence and associated factors. Gynecol Endocrinol. 2007;23:198-205.

27. Fantry L, Zhan M, Taylor G, et al. Age at menopause and menopausal symptoms in HIV-infected women. AIDS Patient Care STD. 2005;19:703-711.

28. Boonyanurak P, Bunupuradah T, Wilawan K, et al. Age at menopause and menopause-related symptoms in human immunodeficiency virus-infected Thai women. Menopause. 2012;19: 820-824.

29. Miller SA, Santoro N, Lo Y. Menopausal symptoms in HIV-infected and drug-using women. Menopause. 2005;12:348-356.

30. Looby S, Shifren J, Corless I. Increased hot flash severity and related interference in perimenopausal HIV-infected women. Menopause. 2014;21:403-409.

31. Schnall R, Jia H, Olender S, et al. In people living with HIV (PLWH), menopause (natural or surgical) contributes to the greater symptom burden in women: results from an online US survey. Menopause. 2018;25:744-752.

32. Thurston RC, Joffe H. Vasomotor symptoms and menopause: findings from the Study of Women’s Health across the Nation. Obstet Gynecol Clin North Am. 2011;38:489-501.

33. Woods NF, Mitchell ES. Symptoms during the perimenopause: prevalence, severity, trajectory, and significance in women’s lives. Am J Med. 2005;118 Suppl 12B:14-24.

34. Maki PM, Rubin LH, Cohen M, et al. Depressive symptoms are increased in the early perimenopausal stage in ethnically diverse human immunodeficiency virus-infected and human immunodeficiency virus-uninfected women. Menopause. 2012;19: 1215-1233.

35. Dennerstein L, Dudley EC, Hopper JL, et al. A prospective population-based study of menopausal symptoms. Obstet Gynecol. 2000;96:351-358.

36. Palma F, Volpe A, Villa P, et al. Vaginal atrophy of women in postmenopause. Results from a multicentric observational study: The AGATA study. Maturitas. 2016;83:40-44.

37. Cutler WB, Garcia CR, McCoy N. Perimenopausal sexuality. Arch Sex Behav. 1987;16:225-234.

38. Moreira ED, Glasser DB, Nicolosi A, et al. GSSAB Investigators’ Group. Sexual problems and help-seeking behavior in adults in the United Kingdom and continental Europe. BJU Int. 2008;101:1005-1111.

39. MacBride MB, Rhodes DJ, Shuster LT. Vulvovaginal atrophy. Mayo Clin Proc. 2010;85:87-94.

40. Nappi RE, Kokot-Kierepa M. Women’s voices in the menopause: results from an international survey on vaginal atrophy. Maturitas. 2010;67:233-238.

41. Santoro N, Komi J. Prevalence and impact of vaginal symptoms among postmenopausal women. J Sex Med. 2009;6:2133-2142.

42. Valadares AL, Pinto-Neto AM, Conde DM, et al. A population-based study of dyspareunia in a cohort of middle-aged Brazilian women. Menopause. 2008;15:1184-1190.

43. Latthe P, Migini L, Gray R, et al. Factors predisposing women to chronic pelvic pain: a systemic review. BMJ. 2006;332:749-755.

44. Potter DA, Moreno A, Luther MF, et al. Effects of follicular-phase cocaine administration on menstrual and ovarian cyclicity in rhesus monkeys. Am J Obstet Gynecol. 1998;178:118-125.

45. Valadares AL, Pinto-Neto AM, Gomes D, et al. Dyspareunia in HIV-positive and HIV-negative middle-aged women: a cross-sectional study. BMJ Open. 2014;4:e004974.

46. Bromberger JT, Meyer PM, Kravitz HM, et al. Psychologic distress and natural menopause: a multiethnic community study. Am J Public Health. 2001;91:1435-1442.

47. Avis NE, Brambilla D, McKinlay SM, Vass K. A longitudinal analysis of the association between menopause and depression. Results from the Massachusetts Women’s Health Study. Ann Epidemiol. 1994;4:214-220.

48. Freeman EW, Sammel MD, Lin H, Nelson DB. Associations of hormones and menopausal status with depressed mood in women with no history of depression. Arch Gen Psychiatry. 2006; 63:375-382.

49. Eller LS, Corless I, Bunch EH, et al. Self-care strategies for depressive symptoms in people with HIV disease. J Adv Nurs. 2005;51:119-130.

50. Fuh JL, Wang SJ, Lee SJ, et al. A longitudinal study of cognition change during early menopausal transition in a rural community. Maturitas. 2006;53:447-453.

51. Hinkin CH, Castellon SA, Atkinson JH, et al. Neuropsychiatric aspects of HIV infection among older adults. J Clin Epidemiol. 2001;54:S44-S52

52. Kravitz HM, Ganz PA, Bromberger J, et al. Sleep difficulty in women at midlife: a community survey of sleep and the menopausal transition. Menopause. 2003;10:19-28.

53. Freedman RR, Roehrs TA. Effects of REM sleep and ambient temperature on hot flash-induced sleep disturbance. Menopause. 2006;13:576-583.

54. Erlik Y, Tataryn IV, Meldrum DR, et al. Association of waking episodes aspects of HIV infection among older adults. J Clin Epidemiol. 2001;54:S44–52.

55. Lui-Filho JF, Valadares AR, Gomes D, et al. Menopausal symptoms and associated factors in HIV-positive women. Maturitas. 2013;76:172-178.

56. Management of symptomatic vulvovaginal atrophy: 2013 position statement of The North American Menopause Society. Menopause. 2013;20:888‐904.

57. Fernandes T, Pedro AO, Baccaro LF, et al. Hormonal, metabolic, and endometrial safety of testosterone vaginal cream versus estrogens for the treatment of vulvovaginal atrophy in postmenopausal women: a randomized, placebo-controlled study. Menopause. 2018; 25:641‐647.

58.  Kroll R, Archer DF, Lin Y, et al. A randomized, multicenter, double-blind study to evaluate the safety and efficacy of estradiol vaginal cream 0.003% in postmenopausal women with dyspareunia as the most bothersome symptom. Menopause. 2018;25:133‐138.

59. Panel on Antiretroviral Guidelines for Adults and Adolescents. Guidelines for the use of antiretroviral agents in adults and adolescents with HIV. Department of Health and Human Services. Tables 21a-d.www.aidsinfo.nih.gov/ContentFiles/ AdultandAdolescentGL.pdf. Accessed May 4, 2020.

60. Tittle, V, Bull, L, Boffito, M. Pharmacokinetic and pharmacodynamics drug interactions between antiretrovirals and oral contraceptives. Clin Pharmacokinet. 2015;54:23-34.

61. Sower M, Zheng H, Tomey K, et al. Changes in body composition in women over six years at midlife: ovarian and chronological aging. J Clin Endocrin Metab. 2007;92:895- 901.

62. Eyawo O, Brockman G, Goldsmith CH, et al. Risk of myocardial infarction among people living with HIV: an updated systematic review and meta-analysis. BMJ Open. 2019;9:e025874.

63. Flooris-Moore M, Howard AA, Lo Y, et al. Increased serum lipids are associated with higher CD4 lymphocyte count in HIV-infected women. HIV Med. 2006;7:421-430.

64. Hadigan C, Meigs JB, Corcoran C, et al. Metabolic abnormalities and cardiovascular disease risk factors in adults with human immunodeficiency virus infection and lipodystrophy. Clin Infect Dis. 2001;32:130-139.

65. Grinspoon S, Carr A. Cardiovascular risk and body fat abnormalities in HIV-infected adults. N Engl J Med. 2005; 352:48–62.

66. Study of Fat Redistribution and Metabolic Change in HIV Infection (FRAM). Fat distribution in women with HIV infection. J Acquir Immune Defic Syndr. 2006;42:562-571.

67. Alcaide ML, Parmigiani A, Pallikkuth S, et al. Immune activation in HIV-infected aging women on antiretrovirals--implications for age-associated comorbidities: a cross-sectional pilot study. PLoS One. 2013;8:e63804.

68. Karim R, Mack WJ, Kono N, et al. Gonadotropin and sex steroid levels in HIV-infected premenopausal women and their association with subclinical atherosclerosis in HIV-infected and -uninfected women in the women’s interagency HIV study (WIHS). J Clin Endocrinol Metab. 2013;98:E610‐E618.

69. Looby SE, Fitch KV, Srinivasa S, et al. Reduced ovarian reserve relates to monocyte activation and subclinical coronary atherosclerotic plaque in women with HIV. AIDS. 2016;30:383‐393.

70. Akhter MP, Lappe JM, Davies KM, et al. Transmenopausal changes in the trabecular bone structure. Bone. 2007;41:111-116.

71. Gibellini D, De Crignis E, Ponti C. HIV-1 triggers apoptosis in primary osteoblasts and HOBIT cells through TNF-alpha activation. J Med Virol. 2008;80:1507-1514.

72. Cassetti I, Madruga JV, Suleiman JM, et al. The safety and efficacy of tenofovir DF in combination with lamivudine and efavirenz through 6 years in antiretroviral-naive HIV- 1-infected patients. HIV Clin Trials. 2007;8:164-172.

73. McComsey GA, Kitch D, Daar ES, et al. Bone mineral density and fractures in antiretroviral-naive persons randomized to receive abacavir-lamivudine or tenofovir disoproxil fumarate-emtricitabine along with efavirenz or atazanavir-ritonavir: AIDS Clinical Trials Group A5224s, a substudy of ACTG A5202. J Infect Dis. 2011;203: 1791-1801.

74. Hansen AB, Obel N, Nielsen H, et al. Bone mineral density changes in protease inhibitor-sparing vs. nucleoside reverse transcriptase inhibitor-sparing highly active antiretroviral therapy: Data from a randomized trial. HIV Med. 2011;12:157-165.

75. FDao CN, Patel P, Overton ET, et al. Study to understand the natural history of HIV and AIDS in the era of effective therapy (SUN) investigators. Low vitamin D among HIV-infected adults: prevalence of and risk factors for low vitamin D levels in cohort of HIV-infected adults and comparison to prevalence among adults in the US general population. Clin Infect Dis. 2011;52:396-405.

76.  Jacobson DL, Spiegelman D, Know TK, Wilson IB. Evolution and predictors of change in total bone mineral density over time in HIV-infected men and women in the nutrition for healthy living study. J Acquir Immune Defic Syndr Hum Retrovirol. 2008;49:298-308.

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

78. Pedrazzoni M, Vescovi L, Maninetti M, et al. Effects of chronic heroine abuse on bone and mineral metabolism. Acta Endocrinol. 1993;129:42-45.

79. Lo Re V 3rd, Guaraldi G, Leonard MB, et al. Viral hepatitis is associated with reduced bone mineral density in HIV-infected women but not men. AIDS. 1990;23:2191-2198.

80. Bregigeon S, Galinier A, Zaegel-Faucher O, et al. Frailty in HIV infected people: a new risk factor for bone mineral density loss [published correction appears in AIDS. AIDS. 2017;31: 1573‐1577.

81. Mills A, Arribas JR, Andrade-Villanueva J, et al. Switching from tenofovir disoproxil fumarate to tenofovir alafenamide in antiretroviral regimens for virologically suppressed adults with HIV-1 infection: a randomised, active-controlled, multicentre, open-label, phase 3, non-inferiority study. Lancet Infect Dis. 2015;16:43-45.

82. King EM, Nesbitt A, Albert AYK, et al. Prolonged amenorrhea and low hip bone mineral density in women living with HIV-a controlled cross-sectional study. J Acquir Immune Defic Syndr. 2020;83:
486‐495.

83. Yin MT, Mcmahon DJ, Ferris DC, et al. Low bone mass and high bone turnover in postmenopausal human immunodeficiency virus-infected women. J Clin Endocrinol Metab. 2010;95:620-629.

84. Yin MT, Modarresi R, Shane E, et al. Effects of HIV infection and antiretroviral therapy with ritonavir on induction of osteoclast-like cells in postmenopausal women. Osteoporos Int. 2011;22:1459-1466.

85. Sharma A, Cohen HW, Freeman R, et al. Prospective evaluation of bone mineral density among middle-aged HIV-infected and uninfected women: association between methadone use and bone loss. Maturitas. 2011;70:295-301.

86. Sharma A, Flom PL, Rosen CJ, et al. Racial differences in bone loss and relation to menopause among HIV-infected and uninfected women. Bone. 2015;77:24-30.

87. Aberg JA, Gallant JE, Ghanem KG, et al, Infectious Diseases Society of America. Primary care guidelines for the management of persons infected with HIV: 2013 update by the HIV medicine association of the Infectious Diseases Society of America. Clin Infect Dis. 2014;58:1‐10.

88. National Osteoporosis Foundation. Clinician’s guide to prevention and treatment of osteoporosis 2014. Washington, DC: National Osteoporosis Foundation; 2014.

89. Yin MT, Choudhury A, Bucovsky M, et al. A randomized placebo-controlled trial of low- versus moderate-dose vitamin d3 supplementation on bone mineral density in postmenopausal women with HIV. J Acquir Immune Defic Syndr. 2019;80:342-349.

90. McComsey GA, Tebas P, Shane E, et al. Bone disease in HIV infection: a practical review and recommendations for HIV care providers. Clin Infect Dis. 2010;51:937-946.

91. Price RW. Neurological complications of HIV infection. Lancet. 1996;348:445-452.

92. Soares CN, Maki PM. Menopausal transition, mood, and cognition: an integrated view to close the gaps. Menopause. 2010;17:812-814.

93. Greendale GA, Wight RG, Huang MH, et al. Menopause-associated symptoms and cognitive performance: results from the study of women’s health across the nation. Am J Epidemiol. 2010;171:1214-1224.

94. Rubin LH, Sundermann EE, Cook JA, et al. An investigation of menopausal stage and symptoms on cognition in HIV-infected women. Menopause. 2014;21:997-1006.

95. Ellerbrock TV, Chiasson MA, Bush TJ, et al. Incidence of cervical squamous intraepithelial lesions in HIV-infected women. JAMA. 2000;283:1031-1037.

96. Mandelblatt JS, Kanetsky P, Eggert L, et al. Is HIV infection a cofactor for cervical squamous cell neoplasia? Cancer Epidemiol Biomarkers Prev. 1999;8:97-106.

97. Kim SC, Messing S, Shah K, et al. Effects of highly active antiretroviral therapy (HAART) and menopause on risk of progression of cervical dysplasia in human immune deficiency virus (HIV) infected women. Infect Dis Obstet Gynecol. 2013;2013:784718.

98. Ceccaldi PF, Ferreira C, Coussy F, et al. Cervical disease in postmenopausal HIV-1 infected women. J Gynecol Obstet Biol Reprod. 2010;39:466-470.

99. Centers for Disease Control and Prevention. HIV and older Americans. www.cdc.gov/hiv/group/age/olderamericans/index.html. Accessed May 11, 2020.

100. Levy JA, Ory MG, Crystal S. HIV/AIDS interventions for midlife and older adults: current status and challenges. J Acquir Immune Defic Syndr. 2003;33 Suppl 2:S59-S67.

101. Thurman AR, Yousefieh N, Chandra N, et al. Comparison of mucosal markers of human immunodeficiency virus susceptibility in healthy premenopausal versus postmenopausal women. AIDS Res Hum Retroviruses. 2017;33:807-819.

102. Meditz AL, Moreau KL, MaWhinney S, et al. CCR5 expression is elevated on endocervical CD4+ T cells in healthy postmenopausal women. J Acquir Immune Defic Syndr. 2012;59:221-228.

103. Chappell CA, Isaacs CE, Xu W, et al. The effect of menopause on the innate antiviral activity of cervicovaginal lavage. Am J Obstet Gynecol. 2015;213:204.

104. Nicol MR, Brewers LM, Kashuba ADM, et al. The role of menopause in tenofovir diphosphate and emtricitabine triphosphate concentrations in cervical tissue. AIDS. 2018;32:11-15.

105. Melo KC, Melo MR, Ricci BV, Segurado AC. Correlates of human immunodeficiency virus cervicovaginal shedding among postmenopausal and fertile-aged women. Menopause. 2012;19:150-156.

106. Landolt NK, Do T, Kasipong N, et al. Low-level genital HIV shedding in Thai HIV-infected women with suppressed plasma viral load after menopause: a longitudinal study. J Virus Erad. 2017;3:204-207.

107. Viard JP, Mocroft A, Chiesi A, et al. Influence of age of CD4 cell recovery in human immunodeficiency virus-infected patients receiving highly active antiretroviral therapy: evidence from the Euro SIDA study. J Infect Dis. 2001;193:1290-1294.

108. Grabar S, Kousignian I, Sobel A, et al. Immunological and clinical responses to highly active antiretroviral therapy over 50 years of age. Results from the French Hospital Database on HIV. AIDS. 2004;18:2029-2038.

109. Cuzin L, Delpierre C, Gerard S, et al. Immunologic and clinical responses to highly active antiretroviral therapy in patients with HIV infection aged >50 years. Clin Infect Dis. 2007;45:654-657.

110. Patterson KB, Cohn SE, Uynik J, et al. Treatment responses in antiretroviral treatment-naïve premenopausal and postmenopausal HIV-1 infected women: an analysis from AIDS clinical trials group studies. Clin Infect Dis. 2009;49:473476.

111. van Benthem BH, Vernazza P, Coutinho RA, et al. The impact of pregnancy and menopause on CD4 lymphocyte count in HIV-infected women. AIDS. 2002;16:919-922.

Author and Disclosure Information

Minji Kang, MD
Infectious Diseases Fellow, Division of Infectious Diseases, University of California San Diego, San Diego, CA

Lori E. Fantry, MD, MPH
Professor of Medicine, University of Arizona/Banner University Medical Center, Tucson, AZ

The authors have reported no conflicts of interest relevant to this article.

Publications
Topics
Sections
Author and Disclosure Information

Minji Kang, MD
Infectious Diseases Fellow, Division of Infectious Diseases, University of California San Diego, San Diego, CA

Lori E. Fantry, MD, MPH
Professor of Medicine, University of Arizona/Banner University Medical Center, Tucson, AZ

The authors have reported no conflicts of interest relevant to this article.

Author and Disclosure Information

Minji Kang, MD
Infectious Diseases Fellow, Division of Infectious Diseases, University of California San Diego, San Diego, CA

Lori E. Fantry, MD, MPH
Professor of Medicine, University of Arizona/Banner University Medical Center, Tucson, AZ

The authors have reported no conflicts of interest relevant to this article.

More than half of the 37.9 million persons living with HIV (PLWH) worldwide are women.1 Between 2010 and 2016, 58% of women living with HIV (WLWH) in the United States were older than 45 years.2 As such, an increasing number of WLWH are entering menopause and living well beyond menopause. Despite this, health care providers expressed a lack of confidence in managing menopause in WLWH, and menopausal symptoms often are not recognized by providers.3 Enhancing our knowledge about menopause in WLWH is important, since the physiologic changes associated with menopause impact short- and long-term quality of life and mortality. 

Test your knowledge of this topic HERE.

Amenorrhea

Menstrual irregularities, including amenorrhea and anovulation, are more frequently found in women of low socioeconomic status, presumably due to associated physical and emotional stress.4 In addition, women with low body mass index (BMI) have decreased serum estradiol levels, which lead to amenorrhea.4,5 Furthermore, low parity and many legal and illegal drugs are associated with amenorrhea, including hormonal contraceptives, opiates, stimulants, antipsychotics, and chemotherapeutic agents.6-8

Because these factors associated with amenorrhea are common in WLWH, it is not surprising that amenorrhea and anovulation are frequently found in this population. However, HIV infection itself also appears to be an independent risk factor for amenorrhea. A recent meta-analysis of 8925 women showed a significant association between HIV status and amenorrhea, even when women with and without HIV had similar rates of substance abuse and smoking and similar socioeconomic status.9 The impact of HIV on an increased frequency of amenorrhea was strongest in women with low BMI. Some, but not all, of the studies included in the meta-analysis found a negative association between CD4 cell count and amenorrhea. In addition, a study comparing amenorrhea frequency within subgroups of WLWH also found a higher rate of amenorrhea in women with lower CD4 cell counts.10

“Prolonged” amenorrhea, defined as amenorrhea lasting 1 year or more, also occurs at a high frequency in WLWH.6 This has made determination of age of menopause extremely challenging, since it is likely that many studies defining menopause are misidentifying “prolonged” amenorrhea as menopause. The Women’s Interagency HIV Study (WIHS), a multicenter observational study of women of similar socioeconomic status living with and without HIV, found that more than 50% of WLWH with “prolonged” amenorrhea had serum follicle-stimulating hormone (FSH) levels in the premenopausal range.8 In a later study from the same cohort, 37% of 660 WLWH with “prolonged” amenorrhea had documented resumption of menses.6

Test your knowledge of this topic HERE.

Age at Menopause

In the United States, the median age of menopause is between 50 and 52 years in middle-class white women.11,12 Earlier menopause has been observed in women who are African American, are nulliparous, have a lower BMI, smoke tobacco, and have more stress, less education, and higher unemployment rates.11,13,14 Because 57% of women diagnosed with HIV in 2018 were African American and many WLWH have other risk factors associated with earlier menopause, studies examining the age of menopause in WLWH need to use a comparator group of women without HIV with similar characteristics and control for these factors to determine the influence of HIV on the age of menopause.

It is also necessary to accurately define menopause. The World Health Organization defines natural menopause as the permanent cessation of menstruation for 12 consecutive months without any obvious pathologic or physiologic causes.15 Most studies have used this definition, and many have found that the age of menopause is earlier in WLWH and is associated with immunosuppression.14,16,17 The Ms Study found that women with CD4 cell counts < 200 cells/μL had an increased risk of amenorrhea lasting at least 12 months, when compared to women with CD4 cell counts ≥ 200 cells/μL. The median age of menopause was 42.5 years in women with CD4 cell counts < 200 cells/μL, 46.0 years in women with CD4 cell counts between 200 cells/μL and 500 cells/μL, and 46.5 years in women with CD4 cell counts > 500 cells/μL.14 Similarly, in a cohort of 667 Brazilian WLWH, among whom 160 were postmenopausal, Calvet and colleagues found that 33% of women with CD4 cell counts < 50 cells/μL had premature menopause, as compared to 8% of women with CD4 cell counts ≥ 350 cells/μL.17 In De Pommerol and colleague’s study of 404 WLWH, among whom 69 were found to be postmenopausal, women with CD4 cell counts < 200 cells/μL were more likely to have premature menopause, as compared to women with CD4 cell counts ≥ 350 cells/μL.16

Despite these findings, given the data from WIHS showing that many women with amenorrhea for at least 12 consecutive months had FSH levels in the premenopausal range8 and that 37% of WLWH have resumption of menses after 12 consecutive months of amenorrhea,6 it is probable that the conclusions about the age of menopause in WLWH are invalid, since many of the participants likely had prolonged amenorrhea, not menopause. WIHS found no significant difference in the median age of menopause when WLWH were compared to women without HIV. The median age of menopause was 47.7 years in WLWH and 48.0 years in women without HIV.18

Menopause-Associated Symptoms

The perimenopausal period, which begins, on average, 4 years prior to the final menstrual period, is characterized by hormonal fluctuations leading to irregular menstrual cycles.19,20 Symptoms associated with these physiologic changes during the perimenopausal period include vasomotor symptoms (hot flashes), genitourinary symptoms (vaginal dryness and dyspareunia), anxiety, depression, sleep disturbances, and joint aches.21,22 Such menopausal symptoms can be distressing and negatively impact quality of life.23 In WLWH, severe menopausal symptoms have been associated with suboptimal adherence to antiretroviral therapy (ART).24 

It can be difficult to determine which symptoms are caused by the physiologic changes of menopause in WLWH, as these women have multiple potential reasons for these symptoms, such as ART, comorbidities, and HIV infection itself.25 However, several studies show that there are symptoms that occur more commonly in the perimenopausal period and that WLWH experience these symptoms earlier and with greater intensity.26-30 In addition, the burden of commonly reported HIV symptoms, such as fatigue and muscle aches/joint pains, is higher in women after menopause, suggesting this burden may be exacerbated by menopause.31

Vasomotor

In the United States, the most common symptom during perimenopause is hot flashes, which occur in 38% to 80% of women.32,33 Vasomotor symptoms are most common in women who smoke, use illicit substances, have a high BMI, are of lower socioeconomic status, and are African American.11 As expected, prior studies focusing on hot flash prevalence among premenopausal, perimenopausal, and postmenopausal WLWH found that postmenopausal women experience more hot flashes than premenopausal or perimenopausal women.27,28 In addition, a comparison of women with and without HIV demonstrated a higher prevalence of hot flashes among WLWH.26,29 Vasomotor symptoms can be severely distressing, with hot flashes contributing to increased risk of depression.25,34 In a cross-sectional analysis of 835 WLWH and 335 women without HIV from the WIHS cohort, persistent vasomotor symptoms predicted elevated depressive symptoms in both WLWH and women without HIV.34 In a similar cross-sectional analysis of 536 women, among whom 54% were WLWH and 37% were perimenopausal, psychological symptoms were prevalent in 61% of the women with vasomotor symptoms.29

Genitourinary

Estrogen deficiency, which accompanies the perimenopausal period, leads to vulvovaginal atrophy (VVA), manifesting with symptoms of vaginal dryness, itching, burning, urinary urgency, and dyspareunia (painful intercourse).33,35,36 Unlike vasomotor symptoms, which diminish with time, genitourinary symptoms generally worsen if left untreated.37 Furthermore, these symptoms are often underreported and underdiagnosed.38,39 VVA was found in 43% to 84% of postmenopausal women.36,40,41 In the AGATA study, the prevalence of VVA was associated with years since menopause. 36 Vaginal dryness and dyspareunia were common.

Genitourinary symptoms are most common among women who are African American, have an increased BMI, are of lower socioeconomic status, use tobacco, have a prior history of pelvic inflammatory disease, and have anxiety and depression.11,42,43 Similar to hot flashes, many of these predisposing factors are more common in WLWH. Fantry and colleagues found that 49.6% of WLWH had vaginal dryness.27 Although 56% of postmenopausal women and 36% of perimenopausal women complained of vaginal dryness, in a multivariate analysis only cocaine use, which can decrease estradiol levels,44 was associated with a higher frequency of vaginal dryness.27

Dyspareunia is also common among WLWH. In a cross-sectional study of 178 women without HIV and 128 WLWH between 40 and 60 years of age, Valadares et al found a high prevalence of dyspareunia in WLWH: 41.8%.45 However, this was not significantly higher than the prevalence of dyspareunia in women without HIV: 34.8%.45 HIV infection itself was not associated with the presence of dyspareunia.

Psychiatric

Anxiety and depression are also common symptoms in perimenopausal women.46-48 Studies have shown that depression is diagnosed 2.5 times more frequently among perimenopausal women than premenopausal women.48 In a study by Miller et al that focused on 536 WLWH, among whom 37% were perimenopausal, 89% reported psychological symptoms.29 Ferreira et al found that perimenopausal WLWH had an increased incidence of psychological symptoms, such as depression and anxiety, compared to women without HIV infection.26 Whether this increased prevalence of psychological symptoms seen in WLWH can be attributed to menopause is unclear, since one third to one half of men and women living with HIV experience symptoms of depression.49 However, in the WIHS, which compared findings from 835 WLWH to findings from 335 women without HIV from all menopausal stages, elevated depressive symptoms were seen in the early perimenopausal period.34 There was no increased incidence of such symptoms during the premenopausal or postmenopausal stage, suggesting that factors related to menopause contribute to depressive symptoms during the perimenopausal stage.34

Persistent menopausal symptoms, especially hot flashes, also predicted elevated depressive symptoms in several studies, suggesting the importance of appropriately identifying and treating menopausal symptoms.29,34 In addition, cognitive decline associated with menopause contributes to depression.50,51

Other Symptoms

Sleep disturbances are common among perimenopausal women, with an estimated prevalence between 38% and 46%.52-54 Hot flashes, anxiety, and depression appear to be factors that contribute to sleep difficulty.52-54 In a cross-sectional study of 273 WLWH and 264 women without HIV between 40 and 60 years of age, insomnia was found in 51% of perimenopausal and 53% of postmenopausal WLWH. The prevalence of insomnia in WLWH and women without HIV was the same.55 Joint aches are also commonly reported in the perimenopausal period, with a prevalence as high as 50% to 60% among perimenopausal women in the United States.22,29 Miller and colleagues found that 63% of menopausal WLWH reported arthralgia.29

Test your knowledge of this topic HERE.

Treatment

Despite the increased severity of menopausal symptoms experienced among WLWH, menopausal replacement therapy (MRT) is used less frequently in WLWH than in  women living without HIV.55 Topical treatment is recommended for women who are experiencing vaginal dryness. First-line treatment is topical nonhormonal therapy, such as moisturizers and lubricants.56 If symptoms are not relieved, then topical vaginal estrogen therapy is recommended.56 Randomized placebo-controlled studies have verified the safety and efficacy of topical estrogen in the general population, and there is no reason to expect different outcomes in WLWH.57,58 

For women experiencing severe hot flashes and vaginal dryness, short-term oral MRT is indicated.56 MRT should be limited to the shortest period of time at the lowest effective dose needed to address these symptoms, as MRT is associated with increased risks of breast cancer, cardiovascular disease, and thromboembolism and increased morbidity.56 Drug interactions between MRT and ART are of concern for non-nucleoside reverse transcriptase inhibitors (NNRTIs), protease inhibitors (PIs), and cobicistat, as these ARTs and MRT are metabolized by the CYP3A4 system.59 With any PI, there is potential for an increase or decrease in estradiol or conjugated estrogen levels; an increase in medroxyprogesterone and micronized progesterone levels; and an increase in drospirenone levels. With doravirine and rilpivirine, there is no change in expected hormonal concentrations, but with other NNRTIs (efavirenz, etravirine, and nevirapine) there is a possibility of a decrease in estradiol, conjugated estrogen, medroxyprogesterone, micronized progesterone, and drospirenone levels. None of the integrase strand transfer inhibitors alone leads to changes in hormone level, but elvitegravir is only used when co-formulated with cobicistat, which may lead to increased or decreased estrogen, progesterone, and drospirenone levels.60 Since all of these drug interactions are uncertain, and even act in varying directions, clinicians should monitor menopausal symptoms and titrate MRT to the dose that achieves relief of menopausal symptoms. 

Cardiovascular Risk

Estrogen deficiency that occurs during menopause leads to an increased risk of cardiovascular disease, particularly with changes in lipid profiles, insulin resistance, and body composition (eg, increased fat mass and waist circumference).61 HIV infection also is associated with a higher risk of cardiovascular disease, with studies consistently reporting a 1.5- to 2-fold increase in the rate of cardiovascular events in PLWH compared to persons without HIV.62 The inflammatory effects of HIV as well as ART exposure, specifically to PIs and abacavir, increase the risk for cardiovascular disease.62 In addition, traditional risk factors, including dyslipidemia, contribute to cardiovascular disease risk in this population.63,64 

The increased risk for cardiovascular disease seen in HIV infection is likely compounded with the increased risk associated with menopause. Postmenopausal WLWH appear to be at higher risk of cardiovascular disease compared to postmenopausal women without HIV. Modifiable risk factors for cardiovascular disease, such as decreased fitness and smoking, are more commonly seen in WLWH.65 Even prior to menopause, WLWH experience lipodystrophy syndrome, with increased truncal visceral adiposity and decreased subcutaneous fat and muscle mass.65,66 Microbial translocation due to HIV-related damage of the intestinal mucosa can lead to elevated levels of lipopolysaccharides, a component of the cell wall of gram-negative bacteria; this subsequently activates monocytes, macrophages, and
T cells. In a study that compared postmenopausal WLWH to age-matched women without HIV, this HIV-related immune activation was correlated with an increase in biomarkers of cardiovascular disease, suggesting WLWH are at higher risk of developing cardiovascular disease.67 Similarly, when comparing sex hormone concentrations in premenopausal WLWH and women without HIV, WLWH had lower estrogen and androgen levels, both of which are linked to carotid artery stiffness.68

In addition, postmenopausal WLWH are at higher risk of cardiovascular disease compared to premenopausal WLWH. WLWH with reduced ovarian reserve had increased subclinical coronary atherosclerotic plaque compared to premenopausal WLWH, even when controlling for cardiovascular disease risk factors.69

In summary, cardiovascular disease risk is increased in postmenopausal WLWH.69 Appropriate measures, such as lipid control, antiplatelet therapy, smoking cessation, aerobic exercise, and other lifestyle changes, should be initiated in WLWH as in any other population. 

 

Test your knowledge of this topic HERE.

Osteoporosis

Menopause, with its associated estrogen deficiency, is the most important risk factor linked to increased bone turnover and bone loss.70 In addition, HIV is associated with bone loss, with low bone mineral density (BMD) described even among men and premenopausal women with HIV infection.71 Although decreased BMD associated with HIV stabilizes or even improves after initiation of ART in the younger population,72-74 chronic inflammation caused by HIV stimulates osteoclast differentiation and resorption.71 Other factors that appear to contribute to decreased BMD among PLWH include ART; vitamin D deficiency; low BMI; poor nutrition; inactivity; use of tobacco, alcohol, and illicit drugs; hepatitis B and C coinfection; and frailty, defined as increased vulnerability to stresses related to aging.72-80 Among ARTs, tenofovir disoproxil fumarate is associated with an increased risk of osteoporosis, and switching from this agent to tenofovir alafenamide improves bone density.81 Prolonged amenorrhea is also an added risk factor for osteoporosis in WLWH.82

Once WLWH enter menopause, they have higher rates of osteoporosis and bone loss compared to women without HIV.83 Among postmenopausal WLWH, those taking ritonavir were found to have increased differentiation of osteoclast cells and increased bone loss.84 Similarly, methadone use in postmenopausal women has been associated with increased declines in BMD.85 African-American postmenopausal WLWH appear to be at the greatest risk for bone loss.86

Given the evidence of low BMD and increased fracture risk that occurs during menopause among women living without HIV, and the additional bone loss observed in PLWH, current guidelines recommend screening postmenopausal women ≥ 50 years of age with dual-energy X-ray absorptiometry (DEXA) scan.87 Preventive therapy, such as smoking cessation, adequate nutrition, alcohol reduction, and weight-bearing exercises, should be discussed and recommended to all menopausal WLWH.88 Adequate calcium and vitamin D intake should be discussed as well, with current evidence indicating that low-dose vitamin D supplementation at 1000 IU is as effective as high-dose vitamin D supplementation at 3000 IU in increasing BMD.89 If the DEXA scan shows a T-score < –2.5 at the femoral neck or spine, or between –1 and –2.5 with a 10-year probability of hip fracture ≥ 3% or a 10-year probability of any osteoporosis-related fracture ≥ 20%, bisphosphonates or other medical therapy should be considered. Although the data are limited in WLWH, bisphosphonates have been shown to be effective in improving BMD.90

Cognition

Both men and women living with HIV are at higher risk for cognitive impairment, ranging from minor cognitive-motor disorder to HIV-associated dementia.91 In addition, the menopause transition is characterized by cognitive changes, such as memory loss and difficulty concentrating.92,93 Studies focusing on the effects of both HIV infection and menopause on cognition have been limited thus far. A cross-sectional study demonstrated that HIV infection, but not menopausal stage, was associated with worse performance on cognitive measures.94 While menopausal stage was not associated with cognitive decline, menopausal symptoms like depression, anxiety, and vasomotor symptoms were associated with lower cognitive performance, highlighting the importance of recognition and treatment of menopausal symptoms.94

Test your knowledge of this topic HERE.

Cervical Dysplasia

WLWH are at increased risk for low- and high-grade squamous intraepithelial lesions (SILs) and more rapid progression to cervical carcinoma, as compared to women without HIV.95 This increased risk of cervical disease is associated with age, human papillomavirus genotype, and degree of immunosuppression.96 In addition, menopause appears to affect the risk of cervical disease. Postmenopausal WLWH had a higher risk of progression of SILs and persistence of lower-grade SILs compared to premenopausal women.97,98 Although studies on progression to cervical cancer in postmenopausal WLWH remain limited, current data suggest that postmenopausal WLWH should continue to be monitored and screened similarly to premenopausal women. 

HIV Acquisition and Transmission

Women aged 50 years and older are primarily exposed to HIV through heterosexual contact.99 While the lack of awareness of HIV risk and less frequent use of barrier protection can contribute to new HIV infection in older women, physiologic changes associated with menopause also may be playing a role.100 Vaginal wall thinning and immunologic changes of the cervix that occur during menopause may serve as a risk factor for HIV acquisition. The cervicovaginal mucosa of postmenopausal women had higher levels of p24 antigen after ex vivo HIV-1 infection, suggesting higher susceptibility to acquire HIV infection.101 Postmenopausal women have been shown to have increased cervical CCR5 expression, which serves as an entry point of HIV into target cells.102 Finally, anti-HIV-1 activity was significantly decreased in postmenopausal women compared to premenopausal women.103 In addition, ex vivo studies demonstrated reduced tenofovir disoproxil fumarate and emtricitabine triphosphate concentrations in cervical tissue of postmenopausal women, suggesting that postmenopausal women may need higher doses of pre-exposure prophylaxis to achieve protective efficacy.104 

In contrast, although data are limited, postmenopausal WLWH do not appear to be at increased risk of vaginally transmitting HIV. The intensity of HIV shedding did not differ between premenopausal or postmenopausal women.105 There was a high prevalence of low-level HIV RNA in genital secretions among perimenopausal WLWH, suggesting WLWH in menopause do not present a major public health risk for HIV transmission.106

HIV Progression

With prior data suggesting that younger persons experience better immunologic and virologic responses to ART,107-109 it had previously been hypothesized that virologic and immunologic responses to ART will decline once WLWH reach menopause. However, current studies suggest that menopause does not affect the progression of HIV and that ART-naive women should respond to ART, regardless of their menopausal status. Treatment responses to ART, determined by the median changes in CD4 cell counts and percentages and viral load, in ART-naive individuals did not differ between premenopausal and postmenopausal women.110 In addition, there appear to be no significant changes in CD4 cell counts as WLWH progress through menopause.111

Conclusion

As individuals with HIV infection live longer, an increasing number of women will enter menopause and live many years beyond menopause. WLWH experience earlier and more severe menopausal symptoms, but evidence on the appropriate management of these symptoms is still lacking. These conditions require proper surveillance, and can be prevented with an improved understanding of the effects of menopause on WLWH. However, there remain significant gaps in our understanding of menopause in WLWH. As practitioners encounter an increasing number of perimenopausal and postmenopausal WLWH, studies of the effects of HIV on comorbidities and symptoms of menopause and their appropriate management are necessary to improve care of WLWH.

Test your knowledge of this topic HERE.

More than half of the 37.9 million persons living with HIV (PLWH) worldwide are women.1 Between 2010 and 2016, 58% of women living with HIV (WLWH) in the United States were older than 45 years.2 As such, an increasing number of WLWH are entering menopause and living well beyond menopause. Despite this, health care providers expressed a lack of confidence in managing menopause in WLWH, and menopausal symptoms often are not recognized by providers.3 Enhancing our knowledge about menopause in WLWH is important, since the physiologic changes associated with menopause impact short- and long-term quality of life and mortality. 

Test your knowledge of this topic HERE.

Amenorrhea

Menstrual irregularities, including amenorrhea and anovulation, are more frequently found in women of low socioeconomic status, presumably due to associated physical and emotional stress.4 In addition, women with low body mass index (BMI) have decreased serum estradiol levels, which lead to amenorrhea.4,5 Furthermore, low parity and many legal and illegal drugs are associated with amenorrhea, including hormonal contraceptives, opiates, stimulants, antipsychotics, and chemotherapeutic agents.6-8

Because these factors associated with amenorrhea are common in WLWH, it is not surprising that amenorrhea and anovulation are frequently found in this population. However, HIV infection itself also appears to be an independent risk factor for amenorrhea. A recent meta-analysis of 8925 women showed a significant association between HIV status and amenorrhea, even when women with and without HIV had similar rates of substance abuse and smoking and similar socioeconomic status.9 The impact of HIV on an increased frequency of amenorrhea was strongest in women with low BMI. Some, but not all, of the studies included in the meta-analysis found a negative association between CD4 cell count and amenorrhea. In addition, a study comparing amenorrhea frequency within subgroups of WLWH also found a higher rate of amenorrhea in women with lower CD4 cell counts.10

“Prolonged” amenorrhea, defined as amenorrhea lasting 1 year or more, also occurs at a high frequency in WLWH.6 This has made determination of age of menopause extremely challenging, since it is likely that many studies defining menopause are misidentifying “prolonged” amenorrhea as menopause. The Women’s Interagency HIV Study (WIHS), a multicenter observational study of women of similar socioeconomic status living with and without HIV, found that more than 50% of WLWH with “prolonged” amenorrhea had serum follicle-stimulating hormone (FSH) levels in the premenopausal range.8 In a later study from the same cohort, 37% of 660 WLWH with “prolonged” amenorrhea had documented resumption of menses.6

Test your knowledge of this topic HERE.

Age at Menopause

In the United States, the median age of menopause is between 50 and 52 years in middle-class white women.11,12 Earlier menopause has been observed in women who are African American, are nulliparous, have a lower BMI, smoke tobacco, and have more stress, less education, and higher unemployment rates.11,13,14 Because 57% of women diagnosed with HIV in 2018 were African American and many WLWH have other risk factors associated with earlier menopause, studies examining the age of menopause in WLWH need to use a comparator group of women without HIV with similar characteristics and control for these factors to determine the influence of HIV on the age of menopause.

It is also necessary to accurately define menopause. The World Health Organization defines natural menopause as the permanent cessation of menstruation for 12 consecutive months without any obvious pathologic or physiologic causes.15 Most studies have used this definition, and many have found that the age of menopause is earlier in WLWH and is associated with immunosuppression.14,16,17 The Ms Study found that women with CD4 cell counts < 200 cells/μL had an increased risk of amenorrhea lasting at least 12 months, when compared to women with CD4 cell counts ≥ 200 cells/μL. The median age of menopause was 42.5 years in women with CD4 cell counts < 200 cells/μL, 46.0 years in women with CD4 cell counts between 200 cells/μL and 500 cells/μL, and 46.5 years in women with CD4 cell counts > 500 cells/μL.14 Similarly, in a cohort of 667 Brazilian WLWH, among whom 160 were postmenopausal, Calvet and colleagues found that 33% of women with CD4 cell counts < 50 cells/μL had premature menopause, as compared to 8% of women with CD4 cell counts ≥ 350 cells/μL.17 In De Pommerol and colleague’s study of 404 WLWH, among whom 69 were found to be postmenopausal, women with CD4 cell counts < 200 cells/μL were more likely to have premature menopause, as compared to women with CD4 cell counts ≥ 350 cells/μL.16

Despite these findings, given the data from WIHS showing that many women with amenorrhea for at least 12 consecutive months had FSH levels in the premenopausal range8 and that 37% of WLWH have resumption of menses after 12 consecutive months of amenorrhea,6 it is probable that the conclusions about the age of menopause in WLWH are invalid, since many of the participants likely had prolonged amenorrhea, not menopause. WIHS found no significant difference in the median age of menopause when WLWH were compared to women without HIV. The median age of menopause was 47.7 years in WLWH and 48.0 years in women without HIV.18

Menopause-Associated Symptoms

The perimenopausal period, which begins, on average, 4 years prior to the final menstrual period, is characterized by hormonal fluctuations leading to irregular menstrual cycles.19,20 Symptoms associated with these physiologic changes during the perimenopausal period include vasomotor symptoms (hot flashes), genitourinary symptoms (vaginal dryness and dyspareunia), anxiety, depression, sleep disturbances, and joint aches.21,22 Such menopausal symptoms can be distressing and negatively impact quality of life.23 In WLWH, severe menopausal symptoms have been associated with suboptimal adherence to antiretroviral therapy (ART).24 

It can be difficult to determine which symptoms are caused by the physiologic changes of menopause in WLWH, as these women have multiple potential reasons for these symptoms, such as ART, comorbidities, and HIV infection itself.25 However, several studies show that there are symptoms that occur more commonly in the perimenopausal period and that WLWH experience these symptoms earlier and with greater intensity.26-30 In addition, the burden of commonly reported HIV symptoms, such as fatigue and muscle aches/joint pains, is higher in women after menopause, suggesting this burden may be exacerbated by menopause.31

Vasomotor

In the United States, the most common symptom during perimenopause is hot flashes, which occur in 38% to 80% of women.32,33 Vasomotor symptoms are most common in women who smoke, use illicit substances, have a high BMI, are of lower socioeconomic status, and are African American.11 As expected, prior studies focusing on hot flash prevalence among premenopausal, perimenopausal, and postmenopausal WLWH found that postmenopausal women experience more hot flashes than premenopausal or perimenopausal women.27,28 In addition, a comparison of women with and without HIV demonstrated a higher prevalence of hot flashes among WLWH.26,29 Vasomotor symptoms can be severely distressing, with hot flashes contributing to increased risk of depression.25,34 In a cross-sectional analysis of 835 WLWH and 335 women without HIV from the WIHS cohort, persistent vasomotor symptoms predicted elevated depressive symptoms in both WLWH and women without HIV.34 In a similar cross-sectional analysis of 536 women, among whom 54% were WLWH and 37% were perimenopausal, psychological symptoms were prevalent in 61% of the women with vasomotor symptoms.29

Genitourinary

Estrogen deficiency, which accompanies the perimenopausal period, leads to vulvovaginal atrophy (VVA), manifesting with symptoms of vaginal dryness, itching, burning, urinary urgency, and dyspareunia (painful intercourse).33,35,36 Unlike vasomotor symptoms, which diminish with time, genitourinary symptoms generally worsen if left untreated.37 Furthermore, these symptoms are often underreported and underdiagnosed.38,39 VVA was found in 43% to 84% of postmenopausal women.36,40,41 In the AGATA study, the prevalence of VVA was associated with years since menopause. 36 Vaginal dryness and dyspareunia were common.

Genitourinary symptoms are most common among women who are African American, have an increased BMI, are of lower socioeconomic status, use tobacco, have a prior history of pelvic inflammatory disease, and have anxiety and depression.11,42,43 Similar to hot flashes, many of these predisposing factors are more common in WLWH. Fantry and colleagues found that 49.6% of WLWH had vaginal dryness.27 Although 56% of postmenopausal women and 36% of perimenopausal women complained of vaginal dryness, in a multivariate analysis only cocaine use, which can decrease estradiol levels,44 was associated with a higher frequency of vaginal dryness.27

Dyspareunia is also common among WLWH. In a cross-sectional study of 178 women without HIV and 128 WLWH between 40 and 60 years of age, Valadares et al found a high prevalence of dyspareunia in WLWH: 41.8%.45 However, this was not significantly higher than the prevalence of dyspareunia in women without HIV: 34.8%.45 HIV infection itself was not associated with the presence of dyspareunia.

Psychiatric

Anxiety and depression are also common symptoms in perimenopausal women.46-48 Studies have shown that depression is diagnosed 2.5 times more frequently among perimenopausal women than premenopausal women.48 In a study by Miller et al that focused on 536 WLWH, among whom 37% were perimenopausal, 89% reported psychological symptoms.29 Ferreira et al found that perimenopausal WLWH had an increased incidence of psychological symptoms, such as depression and anxiety, compared to women without HIV infection.26 Whether this increased prevalence of psychological symptoms seen in WLWH can be attributed to menopause is unclear, since one third to one half of men and women living with HIV experience symptoms of depression.49 However, in the WIHS, which compared findings from 835 WLWH to findings from 335 women without HIV from all menopausal stages, elevated depressive symptoms were seen in the early perimenopausal period.34 There was no increased incidence of such symptoms during the premenopausal or postmenopausal stage, suggesting that factors related to menopause contribute to depressive symptoms during the perimenopausal stage.34

Persistent menopausal symptoms, especially hot flashes, also predicted elevated depressive symptoms in several studies, suggesting the importance of appropriately identifying and treating menopausal symptoms.29,34 In addition, cognitive decline associated with menopause contributes to depression.50,51

Other Symptoms

Sleep disturbances are common among perimenopausal women, with an estimated prevalence between 38% and 46%.52-54 Hot flashes, anxiety, and depression appear to be factors that contribute to sleep difficulty.52-54 In a cross-sectional study of 273 WLWH and 264 women without HIV between 40 and 60 years of age, insomnia was found in 51% of perimenopausal and 53% of postmenopausal WLWH. The prevalence of insomnia in WLWH and women without HIV was the same.55 Joint aches are also commonly reported in the perimenopausal period, with a prevalence as high as 50% to 60% among perimenopausal women in the United States.22,29 Miller and colleagues found that 63% of menopausal WLWH reported arthralgia.29

Test your knowledge of this topic HERE.

Treatment

Despite the increased severity of menopausal symptoms experienced among WLWH, menopausal replacement therapy (MRT) is used less frequently in WLWH than in  women living without HIV.55 Topical treatment is recommended for women who are experiencing vaginal dryness. First-line treatment is topical nonhormonal therapy, such as moisturizers and lubricants.56 If symptoms are not relieved, then topical vaginal estrogen therapy is recommended.56 Randomized placebo-controlled studies have verified the safety and efficacy of topical estrogen in the general population, and there is no reason to expect different outcomes in WLWH.57,58 

For women experiencing severe hot flashes and vaginal dryness, short-term oral MRT is indicated.56 MRT should be limited to the shortest period of time at the lowest effective dose needed to address these symptoms, as MRT is associated with increased risks of breast cancer, cardiovascular disease, and thromboembolism and increased morbidity.56 Drug interactions between MRT and ART are of concern for non-nucleoside reverse transcriptase inhibitors (NNRTIs), protease inhibitors (PIs), and cobicistat, as these ARTs and MRT are metabolized by the CYP3A4 system.59 With any PI, there is potential for an increase or decrease in estradiol or conjugated estrogen levels; an increase in medroxyprogesterone and micronized progesterone levels; and an increase in drospirenone levels. With doravirine and rilpivirine, there is no change in expected hormonal concentrations, but with other NNRTIs (efavirenz, etravirine, and nevirapine) there is a possibility of a decrease in estradiol, conjugated estrogen, medroxyprogesterone, micronized progesterone, and drospirenone levels. None of the integrase strand transfer inhibitors alone leads to changes in hormone level, but elvitegravir is only used when co-formulated with cobicistat, which may lead to increased or decreased estrogen, progesterone, and drospirenone levels.60 Since all of these drug interactions are uncertain, and even act in varying directions, clinicians should monitor menopausal symptoms and titrate MRT to the dose that achieves relief of menopausal symptoms. 

Cardiovascular Risk

Estrogen deficiency that occurs during menopause leads to an increased risk of cardiovascular disease, particularly with changes in lipid profiles, insulin resistance, and body composition (eg, increased fat mass and waist circumference).61 HIV infection also is associated with a higher risk of cardiovascular disease, with studies consistently reporting a 1.5- to 2-fold increase in the rate of cardiovascular events in PLWH compared to persons without HIV.62 The inflammatory effects of HIV as well as ART exposure, specifically to PIs and abacavir, increase the risk for cardiovascular disease.62 In addition, traditional risk factors, including dyslipidemia, contribute to cardiovascular disease risk in this population.63,64 

The increased risk for cardiovascular disease seen in HIV infection is likely compounded with the increased risk associated with menopause. Postmenopausal WLWH appear to be at higher risk of cardiovascular disease compared to postmenopausal women without HIV. Modifiable risk factors for cardiovascular disease, such as decreased fitness and smoking, are more commonly seen in WLWH.65 Even prior to menopause, WLWH experience lipodystrophy syndrome, with increased truncal visceral adiposity and decreased subcutaneous fat and muscle mass.65,66 Microbial translocation due to HIV-related damage of the intestinal mucosa can lead to elevated levels of lipopolysaccharides, a component of the cell wall of gram-negative bacteria; this subsequently activates monocytes, macrophages, and
T cells. In a study that compared postmenopausal WLWH to age-matched women without HIV, this HIV-related immune activation was correlated with an increase in biomarkers of cardiovascular disease, suggesting WLWH are at higher risk of developing cardiovascular disease.67 Similarly, when comparing sex hormone concentrations in premenopausal WLWH and women without HIV, WLWH had lower estrogen and androgen levels, both of which are linked to carotid artery stiffness.68

In addition, postmenopausal WLWH are at higher risk of cardiovascular disease compared to premenopausal WLWH. WLWH with reduced ovarian reserve had increased subclinical coronary atherosclerotic plaque compared to premenopausal WLWH, even when controlling for cardiovascular disease risk factors.69

In summary, cardiovascular disease risk is increased in postmenopausal WLWH.69 Appropriate measures, such as lipid control, antiplatelet therapy, smoking cessation, aerobic exercise, and other lifestyle changes, should be initiated in WLWH as in any other population. 

 

Test your knowledge of this topic HERE.

Osteoporosis

Menopause, with its associated estrogen deficiency, is the most important risk factor linked to increased bone turnover and bone loss.70 In addition, HIV is associated with bone loss, with low bone mineral density (BMD) described even among men and premenopausal women with HIV infection.71 Although decreased BMD associated with HIV stabilizes or even improves after initiation of ART in the younger population,72-74 chronic inflammation caused by HIV stimulates osteoclast differentiation and resorption.71 Other factors that appear to contribute to decreased BMD among PLWH include ART; vitamin D deficiency; low BMI; poor nutrition; inactivity; use of tobacco, alcohol, and illicit drugs; hepatitis B and C coinfection; and frailty, defined as increased vulnerability to stresses related to aging.72-80 Among ARTs, tenofovir disoproxil fumarate is associated with an increased risk of osteoporosis, and switching from this agent to tenofovir alafenamide improves bone density.81 Prolonged amenorrhea is also an added risk factor for osteoporosis in WLWH.82

Once WLWH enter menopause, they have higher rates of osteoporosis and bone loss compared to women without HIV.83 Among postmenopausal WLWH, those taking ritonavir were found to have increased differentiation of osteoclast cells and increased bone loss.84 Similarly, methadone use in postmenopausal women has been associated with increased declines in BMD.85 African-American postmenopausal WLWH appear to be at the greatest risk for bone loss.86

Given the evidence of low BMD and increased fracture risk that occurs during menopause among women living without HIV, and the additional bone loss observed in PLWH, current guidelines recommend screening postmenopausal women ≥ 50 years of age with dual-energy X-ray absorptiometry (DEXA) scan.87 Preventive therapy, such as smoking cessation, adequate nutrition, alcohol reduction, and weight-bearing exercises, should be discussed and recommended to all menopausal WLWH.88 Adequate calcium and vitamin D intake should be discussed as well, with current evidence indicating that low-dose vitamin D supplementation at 1000 IU is as effective as high-dose vitamin D supplementation at 3000 IU in increasing BMD.89 If the DEXA scan shows a T-score < –2.5 at the femoral neck or spine, or between –1 and –2.5 with a 10-year probability of hip fracture ≥ 3% or a 10-year probability of any osteoporosis-related fracture ≥ 20%, bisphosphonates or other medical therapy should be considered. Although the data are limited in WLWH, bisphosphonates have been shown to be effective in improving BMD.90

Cognition

Both men and women living with HIV are at higher risk for cognitive impairment, ranging from minor cognitive-motor disorder to HIV-associated dementia.91 In addition, the menopause transition is characterized by cognitive changes, such as memory loss and difficulty concentrating.92,93 Studies focusing on the effects of both HIV infection and menopause on cognition have been limited thus far. A cross-sectional study demonstrated that HIV infection, but not menopausal stage, was associated with worse performance on cognitive measures.94 While menopausal stage was not associated with cognitive decline, menopausal symptoms like depression, anxiety, and vasomotor symptoms were associated with lower cognitive performance, highlighting the importance of recognition and treatment of menopausal symptoms.94

Test your knowledge of this topic HERE.

Cervical Dysplasia

WLWH are at increased risk for low- and high-grade squamous intraepithelial lesions (SILs) and more rapid progression to cervical carcinoma, as compared to women without HIV.95 This increased risk of cervical disease is associated with age, human papillomavirus genotype, and degree of immunosuppression.96 In addition, menopause appears to affect the risk of cervical disease. Postmenopausal WLWH had a higher risk of progression of SILs and persistence of lower-grade SILs compared to premenopausal women.97,98 Although studies on progression to cervical cancer in postmenopausal WLWH remain limited, current data suggest that postmenopausal WLWH should continue to be monitored and screened similarly to premenopausal women. 

HIV Acquisition and Transmission

Women aged 50 years and older are primarily exposed to HIV through heterosexual contact.99 While the lack of awareness of HIV risk and less frequent use of barrier protection can contribute to new HIV infection in older women, physiologic changes associated with menopause also may be playing a role.100 Vaginal wall thinning and immunologic changes of the cervix that occur during menopause may serve as a risk factor for HIV acquisition. The cervicovaginal mucosa of postmenopausal women had higher levels of p24 antigen after ex vivo HIV-1 infection, suggesting higher susceptibility to acquire HIV infection.101 Postmenopausal women have been shown to have increased cervical CCR5 expression, which serves as an entry point of HIV into target cells.102 Finally, anti-HIV-1 activity was significantly decreased in postmenopausal women compared to premenopausal women.103 In addition, ex vivo studies demonstrated reduced tenofovir disoproxil fumarate and emtricitabine triphosphate concentrations in cervical tissue of postmenopausal women, suggesting that postmenopausal women may need higher doses of pre-exposure prophylaxis to achieve protective efficacy.104 

In contrast, although data are limited, postmenopausal WLWH do not appear to be at increased risk of vaginally transmitting HIV. The intensity of HIV shedding did not differ between premenopausal or postmenopausal women.105 There was a high prevalence of low-level HIV RNA in genital secretions among perimenopausal WLWH, suggesting WLWH in menopause do not present a major public health risk for HIV transmission.106

HIV Progression

With prior data suggesting that younger persons experience better immunologic and virologic responses to ART,107-109 it had previously been hypothesized that virologic and immunologic responses to ART will decline once WLWH reach menopause. However, current studies suggest that menopause does not affect the progression of HIV and that ART-naive women should respond to ART, regardless of their menopausal status. Treatment responses to ART, determined by the median changes in CD4 cell counts and percentages and viral load, in ART-naive individuals did not differ between premenopausal and postmenopausal women.110 In addition, there appear to be no significant changes in CD4 cell counts as WLWH progress through menopause.111

Conclusion

As individuals with HIV infection live longer, an increasing number of women will enter menopause and live many years beyond menopause. WLWH experience earlier and more severe menopausal symptoms, but evidence on the appropriate management of these symptoms is still lacking. These conditions require proper surveillance, and can be prevented with an improved understanding of the effects of menopause on WLWH. However, there remain significant gaps in our understanding of menopause in WLWH. As practitioners encounter an increasing number of perimenopausal and postmenopausal WLWH, studies of the effects of HIV on comorbidities and symptoms of menopause and their appropriate management are necessary to improve care of WLWH.

Test your knowledge of this topic HERE.

References

1. UNAIDS. AIDSInfo. 2019 estimates https://www.unaids.org/en/resources/infographics/girls-and-women-living-with-HIV. Accessed April 30, 2020.

2. Centers for Disease Control and Prevention. Estimated HIV incidence and prevalence in the United States, 2010–2016. HIV Surveillance Supplemental Report 2019;24(No. 1). www.cdc.gov/hiv/library/reports/hiv-surveillance.html. Accessed April 30, 2020.

3. Chirwa M, Ma R, Guallar C, et al. Managing menopause in women living with HIV: A survey of primary care practitioners. Post Reprod Health. 2017;23:111-115.

4. Munster K, Helm P, Schmidt L. Secondary amenorrhea: Prevalence and medical contract–A cross sectional study from a Danish county. Br J Obstet Gynecol. 1992;99:430-433.

5. Vyver E, Steinegger C, Katzman DK, et al. Eating disorders and menstrual dysfunction in adolescents. Ann N Y Acad Sci. 2008;1135: 253-264.

6. Cejtin HE, Evans CT, Greenblatt R, et al. Prolonged amenorrhea and resumption of menses in women with HIV. J Womens Health (Larchmt). 2018;27:1441‐1448.

7. Bai J, Greenwald E, Caterini H, et al. Drug-related menstrual aberrations. Obstet Gynecol. 1974;44:713-719.

8. Cejtin HE, Kalinowski A, Bacchetti P. Effects of human immunodeficiency virus on protracted amenorrhea and ovarian dysfunction. Obstet Gynecol. 2006;108:1423-1431.

9. King EM, Albert AY, Murray MCM. HIV and amenorrhea: a meta-analysis. AIDS. 2019;33:483‐491.

10. Watts DH, Spino C, Zaborski L. Comparison of gynecologic history and laboratory results in HIV-positive women with CDR+ lymphocyte counts between 200 and 500 cells/μl and below 100 cells/ μl. J Acquir Immune Defic Syndr Hum Retrovirol. 1999;20:455-462.

11. Gold EB, Crawford SL, Avis NE, et al. Factors related to age at natural menopause: longitudinal analyses from SWAN. Am J Epidemiol. 2013;178:70-83.

12. Thomas F, Renaud F, Benefice E, et al. International variability of ages at menarche and menopause: patterns and main determinants. Hum Biol. 2001;73:271-290.

13. Bromberger JT, Matthews KA, Kuller LH, et al. Prospective study of the determinants of age at menopause. Am J Epidemiol. 1997; 145:24-33.

14. Schoenbaum E, Hartel D, Lo Y, et al. HIV infection, drug use, and onset of natural menopause. Clinical Infect Dis. 2005;41: 1517-1524.

15. Research on the menopause in the 1990s. Report of a WHO scientific group. World Health Organ Tech Rep Ser. 1996;866:1-107.

16. de Pommerol M, Hessamfar M, Lawson-Ayayi S, et al. Menopause and HIV infection: age at onset and associated factors, ANRS CO3 Aquitaine cohort. Int J STD AIDS. 2011;22:67-72.

17. Calvet G, Grinsztejn G. Predictors of early menopause in HIV infected women: a prospective cohort study. Am J Obstet Gynecol. 2015;212:765.

18. Cejtin SH, Taylor R, Watts DH. Assessment of menopausal status among women in the Women’s Interagency HIV study (WIHS). Proceedings of the XV International AIDS Conference; July 11-16, 2004; Bangkok, Thailand.

19. Taffe JR, Dennerstein L. Menstrual patterns leading to the final menstrual period. Menopause. 2002;9:32-40.

20. Miro F, Parker SW, Aspinall LJ, et al. Origins and consequences of the elongation of the human menstrual cycle during the menopausal transition: the FREEDOM Study. J Clin Endocrinol Metab. 2004;89:4910-4915.

21. McKinlay SM, Brambilla DJ, Posner JG. The normal menopause transition. Maturitas. 1992;14:103-115.

22. Blümel JE, Chedraui P, Baron G, et al. Menopause could be involved in the pathogenesis of muscle and joint aches in mid-aged women. Maturitas. 2013;75:94-100.

23. Woods NF, Mitchell ES. Symptoms interference with work and relationships during the menopausal transition and early postmenopause: observations from the Seattle Midlife Women’s Health Study. Menopause. 2011;18:654-661.

24. Duff PK, Money DM, Ogilvie GS, et al. Severe menopausal symptoms associated with reduced adherence to antiretroviral therapy among perimenopausal and menopausal women living with HIV in Metro Vancouver. Menopause. 2018;25:531-537.

25.  Johnson TM, Cohen HW, Howard AA, et al. Attribution of menopause symptoms in human immunodeficiency virus–infected or at-risk drug-using women. Menopause. 2008;15:551-557.

26. Ferreira CE, Pinto-Neto AM, Conde DM, et al. Menopausal symptoms in women infected with HIV: prevalence and associated factors. Gynecol Endocrinol. 2007;23:198-205.

27. Fantry L, Zhan M, Taylor G, et al. Age at menopause and menopausal symptoms in HIV-infected women. AIDS Patient Care STD. 2005;19:703-711.

28. Boonyanurak P, Bunupuradah T, Wilawan K, et al. Age at menopause and menopause-related symptoms in human immunodeficiency virus-infected Thai women. Menopause. 2012;19: 820-824.

29. Miller SA, Santoro N, Lo Y. Menopausal symptoms in HIV-infected and drug-using women. Menopause. 2005;12:348-356.

30. Looby S, Shifren J, Corless I. Increased hot flash severity and related interference in perimenopausal HIV-infected women. Menopause. 2014;21:403-409.

31. Schnall R, Jia H, Olender S, et al. In people living with HIV (PLWH), menopause (natural or surgical) contributes to the greater symptom burden in women: results from an online US survey. Menopause. 2018;25:744-752.

32. Thurston RC, Joffe H. Vasomotor symptoms and menopause: findings from the Study of Women’s Health across the Nation. Obstet Gynecol Clin North Am. 2011;38:489-501.

33. Woods NF, Mitchell ES. Symptoms during the perimenopause: prevalence, severity, trajectory, and significance in women’s lives. Am J Med. 2005;118 Suppl 12B:14-24.

34. Maki PM, Rubin LH, Cohen M, et al. Depressive symptoms are increased in the early perimenopausal stage in ethnically diverse human immunodeficiency virus-infected and human immunodeficiency virus-uninfected women. Menopause. 2012;19: 1215-1233.

35. Dennerstein L, Dudley EC, Hopper JL, et al. A prospective population-based study of menopausal symptoms. Obstet Gynecol. 2000;96:351-358.

36. Palma F, Volpe A, Villa P, et al. Vaginal atrophy of women in postmenopause. Results from a multicentric observational study: The AGATA study. Maturitas. 2016;83:40-44.

37. Cutler WB, Garcia CR, McCoy N. Perimenopausal sexuality. Arch Sex Behav. 1987;16:225-234.

38. Moreira ED, Glasser DB, Nicolosi A, et al. GSSAB Investigators’ Group. Sexual problems and help-seeking behavior in adults in the United Kingdom and continental Europe. BJU Int. 2008;101:1005-1111.

39. MacBride MB, Rhodes DJ, Shuster LT. Vulvovaginal atrophy. Mayo Clin Proc. 2010;85:87-94.

40. Nappi RE, Kokot-Kierepa M. Women’s voices in the menopause: results from an international survey on vaginal atrophy. Maturitas. 2010;67:233-238.

41. Santoro N, Komi J. Prevalence and impact of vaginal symptoms among postmenopausal women. J Sex Med. 2009;6:2133-2142.

42. Valadares AL, Pinto-Neto AM, Conde DM, et al. A population-based study of dyspareunia in a cohort of middle-aged Brazilian women. Menopause. 2008;15:1184-1190.

43. Latthe P, Migini L, Gray R, et al. Factors predisposing women to chronic pelvic pain: a systemic review. BMJ. 2006;332:749-755.

44. Potter DA, Moreno A, Luther MF, et al. Effects of follicular-phase cocaine administration on menstrual and ovarian cyclicity in rhesus monkeys. Am J Obstet Gynecol. 1998;178:118-125.

45. Valadares AL, Pinto-Neto AM, Gomes D, et al. Dyspareunia in HIV-positive and HIV-negative middle-aged women: a cross-sectional study. BMJ Open. 2014;4:e004974.

46. Bromberger JT, Meyer PM, Kravitz HM, et al. Psychologic distress and natural menopause: a multiethnic community study. Am J Public Health. 2001;91:1435-1442.

47. Avis NE, Brambilla D, McKinlay SM, Vass K. A longitudinal analysis of the association between menopause and depression. Results from the Massachusetts Women’s Health Study. Ann Epidemiol. 1994;4:214-220.

48. Freeman EW, Sammel MD, Lin H, Nelson DB. Associations of hormones and menopausal status with depressed mood in women with no history of depression. Arch Gen Psychiatry. 2006; 63:375-382.

49. Eller LS, Corless I, Bunch EH, et al. Self-care strategies for depressive symptoms in people with HIV disease. J Adv Nurs. 2005;51:119-130.

50. Fuh JL, Wang SJ, Lee SJ, et al. A longitudinal study of cognition change during early menopausal transition in a rural community. Maturitas. 2006;53:447-453.

51. Hinkin CH, Castellon SA, Atkinson JH, et al. Neuropsychiatric aspects of HIV infection among older adults. J Clin Epidemiol. 2001;54:S44-S52

52. Kravitz HM, Ganz PA, Bromberger J, et al. Sleep difficulty in women at midlife: a community survey of sleep and the menopausal transition. Menopause. 2003;10:19-28.

53. Freedman RR, Roehrs TA. Effects of REM sleep and ambient temperature on hot flash-induced sleep disturbance. Menopause. 2006;13:576-583.

54. Erlik Y, Tataryn IV, Meldrum DR, et al. Association of waking episodes aspects of HIV infection among older adults. J Clin Epidemiol. 2001;54:S44–52.

55. Lui-Filho JF, Valadares AR, Gomes D, et al. Menopausal symptoms and associated factors in HIV-positive women. Maturitas. 2013;76:172-178.

56. Management of symptomatic vulvovaginal atrophy: 2013 position statement of The North American Menopause Society. Menopause. 2013;20:888‐904.

57. Fernandes T, Pedro AO, Baccaro LF, et al. Hormonal, metabolic, and endometrial safety of testosterone vaginal cream versus estrogens for the treatment of vulvovaginal atrophy in postmenopausal women: a randomized, placebo-controlled study. Menopause. 2018; 25:641‐647.

58.  Kroll R, Archer DF, Lin Y, et al. A randomized, multicenter, double-blind study to evaluate the safety and efficacy of estradiol vaginal cream 0.003% in postmenopausal women with dyspareunia as the most bothersome symptom. Menopause. 2018;25:133‐138.

59. Panel on Antiretroviral Guidelines for Adults and Adolescents. Guidelines for the use of antiretroviral agents in adults and adolescents with HIV. Department of Health and Human Services. Tables 21a-d.www.aidsinfo.nih.gov/ContentFiles/ AdultandAdolescentGL.pdf. Accessed May 4, 2020.

60. Tittle, V, Bull, L, Boffito, M. Pharmacokinetic and pharmacodynamics drug interactions between antiretrovirals and oral contraceptives. Clin Pharmacokinet. 2015;54:23-34.

61. Sower M, Zheng H, Tomey K, et al. Changes in body composition in women over six years at midlife: ovarian and chronological aging. J Clin Endocrin Metab. 2007;92:895- 901.

62. Eyawo O, Brockman G, Goldsmith CH, et al. Risk of myocardial infarction among people living with HIV: an updated systematic review and meta-analysis. BMJ Open. 2019;9:e025874.

63. Flooris-Moore M, Howard AA, Lo Y, et al. Increased serum lipids are associated with higher CD4 lymphocyte count in HIV-infected women. HIV Med. 2006;7:421-430.

64. Hadigan C, Meigs JB, Corcoran C, et al. Metabolic abnormalities and cardiovascular disease risk factors in adults with human immunodeficiency virus infection and lipodystrophy. Clin Infect Dis. 2001;32:130-139.

65. Grinspoon S, Carr A. Cardiovascular risk and body fat abnormalities in HIV-infected adults. N Engl J Med. 2005; 352:48–62.

66. Study of Fat Redistribution and Metabolic Change in HIV Infection (FRAM). Fat distribution in women with HIV infection. J Acquir Immune Defic Syndr. 2006;42:562-571.

67. Alcaide ML, Parmigiani A, Pallikkuth S, et al. Immune activation in HIV-infected aging women on antiretrovirals--implications for age-associated comorbidities: a cross-sectional pilot study. PLoS One. 2013;8:e63804.

68. Karim R, Mack WJ, Kono N, et al. Gonadotropin and sex steroid levels in HIV-infected premenopausal women and their association with subclinical atherosclerosis in HIV-infected and -uninfected women in the women’s interagency HIV study (WIHS). J Clin Endocrinol Metab. 2013;98:E610‐E618.

69. Looby SE, Fitch KV, Srinivasa S, et al. Reduced ovarian reserve relates to monocyte activation and subclinical coronary atherosclerotic plaque in women with HIV. AIDS. 2016;30:383‐393.

70. Akhter MP, Lappe JM, Davies KM, et al. Transmenopausal changes in the trabecular bone structure. Bone. 2007;41:111-116.

71. Gibellini D, De Crignis E, Ponti C. HIV-1 triggers apoptosis in primary osteoblasts and HOBIT cells through TNF-alpha activation. J Med Virol. 2008;80:1507-1514.

72. Cassetti I, Madruga JV, Suleiman JM, et al. The safety and efficacy of tenofovir DF in combination with lamivudine and efavirenz through 6 years in antiretroviral-naive HIV- 1-infected patients. HIV Clin Trials. 2007;8:164-172.

73. McComsey GA, Kitch D, Daar ES, et al. Bone mineral density and fractures in antiretroviral-naive persons randomized to receive abacavir-lamivudine or tenofovir disoproxil fumarate-emtricitabine along with efavirenz or atazanavir-ritonavir: AIDS Clinical Trials Group A5224s, a substudy of ACTG A5202. J Infect Dis. 2011;203: 1791-1801.

74. Hansen AB, Obel N, Nielsen H, et al. Bone mineral density changes in protease inhibitor-sparing vs. nucleoside reverse transcriptase inhibitor-sparing highly active antiretroviral therapy: Data from a randomized trial. HIV Med. 2011;12:157-165.

75. FDao CN, Patel P, Overton ET, et al. Study to understand the natural history of HIV and AIDS in the era of effective therapy (SUN) investigators. Low vitamin D among HIV-infected adults: prevalence of and risk factors for low vitamin D levels in cohort of HIV-infected adults and comparison to prevalence among adults in the US general population. Clin Infect Dis. 2011;52:396-405.

76.  Jacobson DL, Spiegelman D, Know TK, Wilson IB. Evolution and predictors of change in total bone mineral density over time in HIV-infected men and women in the nutrition for healthy living study. J Acquir Immune Defic Syndr Hum Retrovirol. 2008;49:298-308.

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

78. Pedrazzoni M, Vescovi L, Maninetti M, et al. Effects of chronic heroine abuse on bone and mineral metabolism. Acta Endocrinol. 1993;129:42-45.

79. Lo Re V 3rd, Guaraldi G, Leonard MB, et al. Viral hepatitis is associated with reduced bone mineral density in HIV-infected women but not men. AIDS. 1990;23:2191-2198.

80. Bregigeon S, Galinier A, Zaegel-Faucher O, et al. Frailty in HIV infected people: a new risk factor for bone mineral density loss [published correction appears in AIDS. AIDS. 2017;31: 1573‐1577.

81. Mills A, Arribas JR, Andrade-Villanueva J, et al. Switching from tenofovir disoproxil fumarate to tenofovir alafenamide in antiretroviral regimens for virologically suppressed adults with HIV-1 infection: a randomised, active-controlled, multicentre, open-label, phase 3, non-inferiority study. Lancet Infect Dis. 2015;16:43-45.

82. King EM, Nesbitt A, Albert AYK, et al. Prolonged amenorrhea and low hip bone mineral density in women living with HIV-a controlled cross-sectional study. J Acquir Immune Defic Syndr. 2020;83:
486‐495.

83. Yin MT, Mcmahon DJ, Ferris DC, et al. Low bone mass and high bone turnover in postmenopausal human immunodeficiency virus-infected women. J Clin Endocrinol Metab. 2010;95:620-629.

84. Yin MT, Modarresi R, Shane E, et al. Effects of HIV infection and antiretroviral therapy with ritonavir on induction of osteoclast-like cells in postmenopausal women. Osteoporos Int. 2011;22:1459-1466.

85. Sharma A, Cohen HW, Freeman R, et al. Prospective evaluation of bone mineral density among middle-aged HIV-infected and uninfected women: association between methadone use and bone loss. Maturitas. 2011;70:295-301.

86. Sharma A, Flom PL, Rosen CJ, et al. Racial differences in bone loss and relation to menopause among HIV-infected and uninfected women. Bone. 2015;77:24-30.

87. Aberg JA, Gallant JE, Ghanem KG, et al, Infectious Diseases Society of America. Primary care guidelines for the management of persons infected with HIV: 2013 update by the HIV medicine association of the Infectious Diseases Society of America. Clin Infect Dis. 2014;58:1‐10.

88. National Osteoporosis Foundation. Clinician’s guide to prevention and treatment of osteoporosis 2014. Washington, DC: National Osteoporosis Foundation; 2014.

89. Yin MT, Choudhury A, Bucovsky M, et al. A randomized placebo-controlled trial of low- versus moderate-dose vitamin d3 supplementation on bone mineral density in postmenopausal women with HIV. J Acquir Immune Defic Syndr. 2019;80:342-349.

90. McComsey GA, Tebas P, Shane E, et al. Bone disease in HIV infection: a practical review and recommendations for HIV care providers. Clin Infect Dis. 2010;51:937-946.

91. Price RW. Neurological complications of HIV infection. Lancet. 1996;348:445-452.

92. Soares CN, Maki PM. Menopausal transition, mood, and cognition: an integrated view to close the gaps. Menopause. 2010;17:812-814.

93. Greendale GA, Wight RG, Huang MH, et al. Menopause-associated symptoms and cognitive performance: results from the study of women’s health across the nation. Am J Epidemiol. 2010;171:1214-1224.

94. Rubin LH, Sundermann EE, Cook JA, et al. An investigation of menopausal stage and symptoms on cognition in HIV-infected women. Menopause. 2014;21:997-1006.

95. Ellerbrock TV, Chiasson MA, Bush TJ, et al. Incidence of cervical squamous intraepithelial lesions in HIV-infected women. JAMA. 2000;283:1031-1037.

96. Mandelblatt JS, Kanetsky P, Eggert L, et al. Is HIV infection a cofactor for cervical squamous cell neoplasia? Cancer Epidemiol Biomarkers Prev. 1999;8:97-106.

97. Kim SC, Messing S, Shah K, et al. Effects of highly active antiretroviral therapy (HAART) and menopause on risk of progression of cervical dysplasia in human immune deficiency virus (HIV) infected women. Infect Dis Obstet Gynecol. 2013;2013:784718.

98. Ceccaldi PF, Ferreira C, Coussy F, et al. Cervical disease in postmenopausal HIV-1 infected women. J Gynecol Obstet Biol Reprod. 2010;39:466-470.

99. Centers for Disease Control and Prevention. HIV and older Americans. www.cdc.gov/hiv/group/age/olderamericans/index.html. Accessed May 11, 2020.

100. Levy JA, Ory MG, Crystal S. HIV/AIDS interventions for midlife and older adults: current status and challenges. J Acquir Immune Defic Syndr. 2003;33 Suppl 2:S59-S67.

101. Thurman AR, Yousefieh N, Chandra N, et al. Comparison of mucosal markers of human immunodeficiency virus susceptibility in healthy premenopausal versus postmenopausal women. AIDS Res Hum Retroviruses. 2017;33:807-819.

102. Meditz AL, Moreau KL, MaWhinney S, et al. CCR5 expression is elevated on endocervical CD4+ T cells in healthy postmenopausal women. J Acquir Immune Defic Syndr. 2012;59:221-228.

103. Chappell CA, Isaacs CE, Xu W, et al. The effect of menopause on the innate antiviral activity of cervicovaginal lavage. Am J Obstet Gynecol. 2015;213:204.

104. Nicol MR, Brewers LM, Kashuba ADM, et al. The role of menopause in tenofovir diphosphate and emtricitabine triphosphate concentrations in cervical tissue. AIDS. 2018;32:11-15.

105. Melo KC, Melo MR, Ricci BV, Segurado AC. Correlates of human immunodeficiency virus cervicovaginal shedding among postmenopausal and fertile-aged women. Menopause. 2012;19:150-156.

106. Landolt NK, Do T, Kasipong N, et al. Low-level genital HIV shedding in Thai HIV-infected women with suppressed plasma viral load after menopause: a longitudinal study. J Virus Erad. 2017;3:204-207.

107. Viard JP, Mocroft A, Chiesi A, et al. Influence of age of CD4 cell recovery in human immunodeficiency virus-infected patients receiving highly active antiretroviral therapy: evidence from the Euro SIDA study. J Infect Dis. 2001;193:1290-1294.

108. Grabar S, Kousignian I, Sobel A, et al. Immunological and clinical responses to highly active antiretroviral therapy over 50 years of age. Results from the French Hospital Database on HIV. AIDS. 2004;18:2029-2038.

109. Cuzin L, Delpierre C, Gerard S, et al. Immunologic and clinical responses to highly active antiretroviral therapy in patients with HIV infection aged >50 years. Clin Infect Dis. 2007;45:654-657.

110. Patterson KB, Cohn SE, Uynik J, et al. Treatment responses in antiretroviral treatment-naïve premenopausal and postmenopausal HIV-1 infected women: an analysis from AIDS clinical trials group studies. Clin Infect Dis. 2009;49:473476.

111. van Benthem BH, Vernazza P, Coutinho RA, et al. The impact of pregnancy and menopause on CD4 lymphocyte count in HIV-infected women. AIDS. 2002;16:919-922.

References

1. UNAIDS. AIDSInfo. 2019 estimates https://www.unaids.org/en/resources/infographics/girls-and-women-living-with-HIV. Accessed April 30, 2020.

2. Centers for Disease Control and Prevention. Estimated HIV incidence and prevalence in the United States, 2010–2016. HIV Surveillance Supplemental Report 2019;24(No. 1). www.cdc.gov/hiv/library/reports/hiv-surveillance.html. Accessed April 30, 2020.

3. Chirwa M, Ma R, Guallar C, et al. Managing menopause in women living with HIV: A survey of primary care practitioners. Post Reprod Health. 2017;23:111-115.

4. Munster K, Helm P, Schmidt L. Secondary amenorrhea: Prevalence and medical contract–A cross sectional study from a Danish county. Br J Obstet Gynecol. 1992;99:430-433.

5. Vyver E, Steinegger C, Katzman DK, et al. Eating disorders and menstrual dysfunction in adolescents. Ann N Y Acad Sci. 2008;1135: 253-264.

6. Cejtin HE, Evans CT, Greenblatt R, et al. Prolonged amenorrhea and resumption of menses in women with HIV. J Womens Health (Larchmt). 2018;27:1441‐1448.

7. Bai J, Greenwald E, Caterini H, et al. Drug-related menstrual aberrations. Obstet Gynecol. 1974;44:713-719.

8. Cejtin HE, Kalinowski A, Bacchetti P. Effects of human immunodeficiency virus on protracted amenorrhea and ovarian dysfunction. Obstet Gynecol. 2006;108:1423-1431.

9. King EM, Albert AY, Murray MCM. HIV and amenorrhea: a meta-analysis. AIDS. 2019;33:483‐491.

10. Watts DH, Spino C, Zaborski L. Comparison of gynecologic history and laboratory results in HIV-positive women with CDR+ lymphocyte counts between 200 and 500 cells/μl and below 100 cells/ μl. J Acquir Immune Defic Syndr Hum Retrovirol. 1999;20:455-462.

11. Gold EB, Crawford SL, Avis NE, et al. Factors related to age at natural menopause: longitudinal analyses from SWAN. Am J Epidemiol. 2013;178:70-83.

12. Thomas F, Renaud F, Benefice E, et al. International variability of ages at menarche and menopause: patterns and main determinants. Hum Biol. 2001;73:271-290.

13. Bromberger JT, Matthews KA, Kuller LH, et al. Prospective study of the determinants of age at menopause. Am J Epidemiol. 1997; 145:24-33.

14. Schoenbaum E, Hartel D, Lo Y, et al. HIV infection, drug use, and onset of natural menopause. Clinical Infect Dis. 2005;41: 1517-1524.

15. Research on the menopause in the 1990s. Report of a WHO scientific group. World Health Organ Tech Rep Ser. 1996;866:1-107.

16. de Pommerol M, Hessamfar M, Lawson-Ayayi S, et al. Menopause and HIV infection: age at onset and associated factors, ANRS CO3 Aquitaine cohort. Int J STD AIDS. 2011;22:67-72.

17. Calvet G, Grinsztejn G. Predictors of early menopause in HIV infected women: a prospective cohort study. Am J Obstet Gynecol. 2015;212:765.

18. Cejtin SH, Taylor R, Watts DH. Assessment of menopausal status among women in the Women’s Interagency HIV study (WIHS). Proceedings of the XV International AIDS Conference; July 11-16, 2004; Bangkok, Thailand.

19. Taffe JR, Dennerstein L. Menstrual patterns leading to the final menstrual period. Menopause. 2002;9:32-40.

20. Miro F, Parker SW, Aspinall LJ, et al. Origins and consequences of the elongation of the human menstrual cycle during the menopausal transition: the FREEDOM Study. J Clin Endocrinol Metab. 2004;89:4910-4915.

21. McKinlay SM, Brambilla DJ, Posner JG. The normal menopause transition. Maturitas. 1992;14:103-115.

22. Blümel JE, Chedraui P, Baron G, et al. Menopause could be involved in the pathogenesis of muscle and joint aches in mid-aged women. Maturitas. 2013;75:94-100.

23. Woods NF, Mitchell ES. Symptoms interference with work and relationships during the menopausal transition and early postmenopause: observations from the Seattle Midlife Women’s Health Study. Menopause. 2011;18:654-661.

24. Duff PK, Money DM, Ogilvie GS, et al. Severe menopausal symptoms associated with reduced adherence to antiretroviral therapy among perimenopausal and menopausal women living with HIV in Metro Vancouver. Menopause. 2018;25:531-537.

25.  Johnson TM, Cohen HW, Howard AA, et al. Attribution of menopause symptoms in human immunodeficiency virus–infected or at-risk drug-using women. Menopause. 2008;15:551-557.

26. Ferreira CE, Pinto-Neto AM, Conde DM, et al. Menopausal symptoms in women infected with HIV: prevalence and associated factors. Gynecol Endocrinol. 2007;23:198-205.

27. Fantry L, Zhan M, Taylor G, et al. Age at menopause and menopausal symptoms in HIV-infected women. AIDS Patient Care STD. 2005;19:703-711.

28. Boonyanurak P, Bunupuradah T, Wilawan K, et al. Age at menopause and menopause-related symptoms in human immunodeficiency virus-infected Thai women. Menopause. 2012;19: 820-824.

29. Miller SA, Santoro N, Lo Y. Menopausal symptoms in HIV-infected and drug-using women. Menopause. 2005;12:348-356.

30. Looby S, Shifren J, Corless I. Increased hot flash severity and related interference in perimenopausal HIV-infected women. Menopause. 2014;21:403-409.

31. Schnall R, Jia H, Olender S, et al. In people living with HIV (PLWH), menopause (natural or surgical) contributes to the greater symptom burden in women: results from an online US survey. Menopause. 2018;25:744-752.

32. Thurston RC, Joffe H. Vasomotor symptoms and menopause: findings from the Study of Women’s Health across the Nation. Obstet Gynecol Clin North Am. 2011;38:489-501.

33. Woods NF, Mitchell ES. Symptoms during the perimenopause: prevalence, severity, trajectory, and significance in women’s lives. Am J Med. 2005;118 Suppl 12B:14-24.

34. Maki PM, Rubin LH, Cohen M, et al. Depressive symptoms are increased in the early perimenopausal stage in ethnically diverse human immunodeficiency virus-infected and human immunodeficiency virus-uninfected women. Menopause. 2012;19: 1215-1233.

35. Dennerstein L, Dudley EC, Hopper JL, et al. A prospective population-based study of menopausal symptoms. Obstet Gynecol. 2000;96:351-358.

36. Palma F, Volpe A, Villa P, et al. Vaginal atrophy of women in postmenopause. Results from a multicentric observational study: The AGATA study. Maturitas. 2016;83:40-44.

37. Cutler WB, Garcia CR, McCoy N. Perimenopausal sexuality. Arch Sex Behav. 1987;16:225-234.

38. Moreira ED, Glasser DB, Nicolosi A, et al. GSSAB Investigators’ Group. Sexual problems and help-seeking behavior in adults in the United Kingdom and continental Europe. BJU Int. 2008;101:1005-1111.

39. MacBride MB, Rhodes DJ, Shuster LT. Vulvovaginal atrophy. Mayo Clin Proc. 2010;85:87-94.

40. Nappi RE, Kokot-Kierepa M. Women’s voices in the menopause: results from an international survey on vaginal atrophy. Maturitas. 2010;67:233-238.

41. Santoro N, Komi J. Prevalence and impact of vaginal symptoms among postmenopausal women. J Sex Med. 2009;6:2133-2142.

42. Valadares AL, Pinto-Neto AM, Conde DM, et al. A population-based study of dyspareunia in a cohort of middle-aged Brazilian women. Menopause. 2008;15:1184-1190.

43. Latthe P, Migini L, Gray R, et al. Factors predisposing women to chronic pelvic pain: a systemic review. BMJ. 2006;332:749-755.

44. Potter DA, Moreno A, Luther MF, et al. Effects of follicular-phase cocaine administration on menstrual and ovarian cyclicity in rhesus monkeys. Am J Obstet Gynecol. 1998;178:118-125.

45. Valadares AL, Pinto-Neto AM, Gomes D, et al. Dyspareunia in HIV-positive and HIV-negative middle-aged women: a cross-sectional study. BMJ Open. 2014;4:e004974.

46. Bromberger JT, Meyer PM, Kravitz HM, et al. Psychologic distress and natural menopause: a multiethnic community study. Am J Public Health. 2001;91:1435-1442.

47. Avis NE, Brambilla D, McKinlay SM, Vass K. A longitudinal analysis of the association between menopause and depression. Results from the Massachusetts Women’s Health Study. Ann Epidemiol. 1994;4:214-220.

48. Freeman EW, Sammel MD, Lin H, Nelson DB. Associations of hormones and menopausal status with depressed mood in women with no history of depression. Arch Gen Psychiatry. 2006; 63:375-382.

49. Eller LS, Corless I, Bunch EH, et al. Self-care strategies for depressive symptoms in people with HIV disease. J Adv Nurs. 2005;51:119-130.

50. Fuh JL, Wang SJ, Lee SJ, et al. A longitudinal study of cognition change during early menopausal transition in a rural community. Maturitas. 2006;53:447-453.

51. Hinkin CH, Castellon SA, Atkinson JH, et al. Neuropsychiatric aspects of HIV infection among older adults. J Clin Epidemiol. 2001;54:S44-S52

52. Kravitz HM, Ganz PA, Bromberger J, et al. Sleep difficulty in women at midlife: a community survey of sleep and the menopausal transition. Menopause. 2003;10:19-28.

53. Freedman RR, Roehrs TA. Effects of REM sleep and ambient temperature on hot flash-induced sleep disturbance. Menopause. 2006;13:576-583.

54. Erlik Y, Tataryn IV, Meldrum DR, et al. Association of waking episodes aspects of HIV infection among older adults. J Clin Epidemiol. 2001;54:S44–52.

55. Lui-Filho JF, Valadares AR, Gomes D, et al. Menopausal symptoms and associated factors in HIV-positive women. Maturitas. 2013;76:172-178.

56. Management of symptomatic vulvovaginal atrophy: 2013 position statement of The North American Menopause Society. Menopause. 2013;20:888‐904.

57. Fernandes T, Pedro AO, Baccaro LF, et al. Hormonal, metabolic, and endometrial safety of testosterone vaginal cream versus estrogens for the treatment of vulvovaginal atrophy in postmenopausal women: a randomized, placebo-controlled study. Menopause. 2018; 25:641‐647.

58.  Kroll R, Archer DF, Lin Y, et al. A randomized, multicenter, double-blind study to evaluate the safety and efficacy of estradiol vaginal cream 0.003% in postmenopausal women with dyspareunia as the most bothersome symptom. Menopause. 2018;25:133‐138.

59. Panel on Antiretroviral Guidelines for Adults and Adolescents. Guidelines for the use of antiretroviral agents in adults and adolescents with HIV. Department of Health and Human Services. Tables 21a-d.www.aidsinfo.nih.gov/ContentFiles/ AdultandAdolescentGL.pdf. Accessed May 4, 2020.

60. Tittle, V, Bull, L, Boffito, M. Pharmacokinetic and pharmacodynamics drug interactions between antiretrovirals and oral contraceptives. Clin Pharmacokinet. 2015;54:23-34.

61. Sower M, Zheng H, Tomey K, et al. Changes in body composition in women over six years at midlife: ovarian and chronological aging. J Clin Endocrin Metab. 2007;92:895- 901.

62. Eyawo O, Brockman G, Goldsmith CH, et al. Risk of myocardial infarction among people living with HIV: an updated systematic review and meta-analysis. BMJ Open. 2019;9:e025874.

63. Flooris-Moore M, Howard AA, Lo Y, et al. Increased serum lipids are associated with higher CD4 lymphocyte count in HIV-infected women. HIV Med. 2006;7:421-430.

64. Hadigan C, Meigs JB, Corcoran C, et al. Metabolic abnormalities and cardiovascular disease risk factors in adults with human immunodeficiency virus infection and lipodystrophy. Clin Infect Dis. 2001;32:130-139.

65. Grinspoon S, Carr A. Cardiovascular risk and body fat abnormalities in HIV-infected adults. N Engl J Med. 2005; 352:48–62.

66. Study of Fat Redistribution and Metabolic Change in HIV Infection (FRAM). Fat distribution in women with HIV infection. J Acquir Immune Defic Syndr. 2006;42:562-571.

67. Alcaide ML, Parmigiani A, Pallikkuth S, et al. Immune activation in HIV-infected aging women on antiretrovirals--implications for age-associated comorbidities: a cross-sectional pilot study. PLoS One. 2013;8:e63804.

68. Karim R, Mack WJ, Kono N, et al. Gonadotropin and sex steroid levels in HIV-infected premenopausal women and their association with subclinical atherosclerosis in HIV-infected and -uninfected women in the women’s interagency HIV study (WIHS). J Clin Endocrinol Metab. 2013;98:E610‐E618.

69. Looby SE, Fitch KV, Srinivasa S, et al. Reduced ovarian reserve relates to monocyte activation and subclinical coronary atherosclerotic plaque in women with HIV. AIDS. 2016;30:383‐393.

70. Akhter MP, Lappe JM, Davies KM, et al. Transmenopausal changes in the trabecular bone structure. Bone. 2007;41:111-116.

71. Gibellini D, De Crignis E, Ponti C. HIV-1 triggers apoptosis in primary osteoblasts and HOBIT cells through TNF-alpha activation. J Med Virol. 2008;80:1507-1514.

72. Cassetti I, Madruga JV, Suleiman JM, et al. The safety and efficacy of tenofovir DF in combination with lamivudine and efavirenz through 6 years in antiretroviral-naive HIV- 1-infected patients. HIV Clin Trials. 2007;8:164-172.

73. McComsey GA, Kitch D, Daar ES, et al. Bone mineral density and fractures in antiretroviral-naive persons randomized to receive abacavir-lamivudine or tenofovir disoproxil fumarate-emtricitabine along with efavirenz or atazanavir-ritonavir: AIDS Clinical Trials Group A5224s, a substudy of ACTG A5202. J Infect Dis. 2011;203: 1791-1801.

74. Hansen AB, Obel N, Nielsen H, et al. Bone mineral density changes in protease inhibitor-sparing vs. nucleoside reverse transcriptase inhibitor-sparing highly active antiretroviral therapy: Data from a randomized trial. HIV Med. 2011;12:157-165.

75. FDao CN, Patel P, Overton ET, et al. Study to understand the natural history of HIV and AIDS in the era of effective therapy (SUN) investigators. Low vitamin D among HIV-infected adults: prevalence of and risk factors for low vitamin D levels in cohort of HIV-infected adults and comparison to prevalence among adults in the US general population. Clin Infect Dis. 2011;52:396-405.

76.  Jacobson DL, Spiegelman D, Know TK, Wilson IB. Evolution and predictors of change in total bone mineral density over time in HIV-infected men and women in the nutrition for healthy living study. J Acquir Immune Defic Syndr Hum Retrovirol. 2008;49:298-308.

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

78. Pedrazzoni M, Vescovi L, Maninetti M, et al. Effects of chronic heroine abuse on bone and mineral metabolism. Acta Endocrinol. 1993;129:42-45.

79. Lo Re V 3rd, Guaraldi G, Leonard MB, et al. Viral hepatitis is associated with reduced bone mineral density in HIV-infected women but not men. AIDS. 1990;23:2191-2198.

80. Bregigeon S, Galinier A, Zaegel-Faucher O, et al. Frailty in HIV infected people: a new risk factor for bone mineral density loss [published correction appears in AIDS. AIDS. 2017;31: 1573‐1577.

81. Mills A, Arribas JR, Andrade-Villanueva J, et al. Switching from tenofovir disoproxil fumarate to tenofovir alafenamide in antiretroviral regimens for virologically suppressed adults with HIV-1 infection: a randomised, active-controlled, multicentre, open-label, phase 3, non-inferiority study. Lancet Infect Dis. 2015;16:43-45.

82. King EM, Nesbitt A, Albert AYK, et al. Prolonged amenorrhea and low hip bone mineral density in women living with HIV-a controlled cross-sectional study. J Acquir Immune Defic Syndr. 2020;83:
486‐495.

83. Yin MT, Mcmahon DJ, Ferris DC, et al. Low bone mass and high bone turnover in postmenopausal human immunodeficiency virus-infected women. J Clin Endocrinol Metab. 2010;95:620-629.

84. Yin MT, Modarresi R, Shane E, et al. Effects of HIV infection and antiretroviral therapy with ritonavir on induction of osteoclast-like cells in postmenopausal women. Osteoporos Int. 2011;22:1459-1466.

85. Sharma A, Cohen HW, Freeman R, et al. Prospective evaluation of bone mineral density among middle-aged HIV-infected and uninfected women: association between methadone use and bone loss. Maturitas. 2011;70:295-301.

86. Sharma A, Flom PL, Rosen CJ, et al. Racial differences in bone loss and relation to menopause among HIV-infected and uninfected women. Bone. 2015;77:24-30.

87. Aberg JA, Gallant JE, Ghanem KG, et al, Infectious Diseases Society of America. Primary care guidelines for the management of persons infected with HIV: 2013 update by the HIV medicine association of the Infectious Diseases Society of America. Clin Infect Dis. 2014;58:1‐10.

88. National Osteoporosis Foundation. Clinician’s guide to prevention and treatment of osteoporosis 2014. Washington, DC: National Osteoporosis Foundation; 2014.

89. Yin MT, Choudhury A, Bucovsky M, et al. A randomized placebo-controlled trial of low- versus moderate-dose vitamin d3 supplementation on bone mineral density in postmenopausal women with HIV. J Acquir Immune Defic Syndr. 2019;80:342-349.

90. McComsey GA, Tebas P, Shane E, et al. Bone disease in HIV infection: a practical review and recommendations for HIV care providers. Clin Infect Dis. 2010;51:937-946.

91. Price RW. Neurological complications of HIV infection. Lancet. 1996;348:445-452.

92. Soares CN, Maki PM. Menopausal transition, mood, and cognition: an integrated view to close the gaps. Menopause. 2010;17:812-814.

93. Greendale GA, Wight RG, Huang MH, et al. Menopause-associated symptoms and cognitive performance: results from the study of women’s health across the nation. Am J Epidemiol. 2010;171:1214-1224.

94. Rubin LH, Sundermann EE, Cook JA, et al. An investigation of menopausal stage and symptoms on cognition in HIV-infected women. Menopause. 2014;21:997-1006.

95. Ellerbrock TV, Chiasson MA, Bush TJ, et al. Incidence of cervical squamous intraepithelial lesions in HIV-infected women. JAMA. 2000;283:1031-1037.

96. Mandelblatt JS, Kanetsky P, Eggert L, et al. Is HIV infection a cofactor for cervical squamous cell neoplasia? Cancer Epidemiol Biomarkers Prev. 1999;8:97-106.

97. Kim SC, Messing S, Shah K, et al. Effects of highly active antiretroviral therapy (HAART) and menopause on risk of progression of cervical dysplasia in human immune deficiency virus (HIV) infected women. Infect Dis Obstet Gynecol. 2013;2013:784718.

98. Ceccaldi PF, Ferreira C, Coussy F, et al. Cervical disease in postmenopausal HIV-1 infected women. J Gynecol Obstet Biol Reprod. 2010;39:466-470.

99. Centers for Disease Control and Prevention. HIV and older Americans. www.cdc.gov/hiv/group/age/olderamericans/index.html. Accessed May 11, 2020.

100. Levy JA, Ory MG, Crystal S. HIV/AIDS interventions for midlife and older adults: current status and challenges. J Acquir Immune Defic Syndr. 2003;33 Suppl 2:S59-S67.

101. Thurman AR, Yousefieh N, Chandra N, et al. Comparison of mucosal markers of human immunodeficiency virus susceptibility in healthy premenopausal versus postmenopausal women. AIDS Res Hum Retroviruses. 2017;33:807-819.

102. Meditz AL, Moreau KL, MaWhinney S, et al. CCR5 expression is elevated on endocervical CD4+ T cells in healthy postmenopausal women. J Acquir Immune Defic Syndr. 2012;59:221-228.

103. Chappell CA, Isaacs CE, Xu W, et al. The effect of menopause on the innate antiviral activity of cervicovaginal lavage. Am J Obstet Gynecol. 2015;213:204.

104. Nicol MR, Brewers LM, Kashuba ADM, et al. The role of menopause in tenofovir diphosphate and emtricitabine triphosphate concentrations in cervical tissue. AIDS. 2018;32:11-15.

105. Melo KC, Melo MR, Ricci BV, Segurado AC. Correlates of human immunodeficiency virus cervicovaginal shedding among postmenopausal and fertile-aged women. Menopause. 2012;19:150-156.

106. Landolt NK, Do T, Kasipong N, et al. Low-level genital HIV shedding in Thai HIV-infected women with suppressed plasma viral load after menopause: a longitudinal study. J Virus Erad. 2017;3:204-207.

107. Viard JP, Mocroft A, Chiesi A, et al. Influence of age of CD4 cell recovery in human immunodeficiency virus-infected patients receiving highly active antiretroviral therapy: evidence from the Euro SIDA study. J Infect Dis. 2001;193:1290-1294.

108. Grabar S, Kousignian I, Sobel A, et al. Immunological and clinical responses to highly active antiretroviral therapy over 50 years of age. Results from the French Hospital Database on HIV. AIDS. 2004;18:2029-2038.

109. Cuzin L, Delpierre C, Gerard S, et al. Immunologic and clinical responses to highly active antiretroviral therapy in patients with HIV infection aged >50 years. Clin Infect Dis. 2007;45:654-657.

110. Patterson KB, Cohn SE, Uynik J, et al. Treatment responses in antiretroviral treatment-naïve premenopausal and postmenopausal HIV-1 infected women: an analysis from AIDS clinical trials group studies. Clin Infect Dis. 2009;49:473476.

111. van Benthem BH, Vernazza P, Coutinho RA, et al. The impact of pregnancy and menopause on CD4 lymphocyte count in HIV-infected women. AIDS. 2002;16:919-922.

Publications
Publications
Topics
Article Type
Display Headline
Infectious Diseases Board Review: Menopause in Women Living With HIV
Display Headline
Infectious Diseases Board Review: Menopause in Women Living With HIV
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Gate On Date
Tue, 02/25/2020 - 14:15
Un-Gate On Date
Tue, 02/25/2020 - 14:15
Use ProPublica
CFC Schedule Remove Status
Tue, 02/25/2020 - 14:15
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article

Menopause in HIV-Infected Women

Article Type
Changed
Wed, 02/28/2018 - 15:38
Display Headline
Menopause in HIV-Infected Women

From the University of Maryland School of Medicine, Baltimore, MD.

 

Abstract

  • Objective: To review the current literature on menopause in HIV-infected women.
  • Methods: We searched PubMed for articles published in English using the search terms HIV and menopause, HIV and amenorrhea, HIV and menopause symptoms, HIV and vasomotor symptoms, HIV and vaginal dryness, HIV and dyspareunia, HIV and menopause and cardiovascular disease, HIV and menopause and osteoporosis, HIV and menopause and cognition, HIV and menopause and cervical dysplasia, menopause and HIV transmission, and menopause and HIV progression. Major studies on menopause in other populations were also reviewed to provide background data.
  • Results: While studies on the age of menopause in HIV-infected women give conflicting results, immuno-suppression associated with HIV appears to contribute to an earlier onset of menopause. HIV-infected women experience menopausal symptoms, especially vasomotor symptoms, earlier and in greater intensity. In addition, menopause and HIV infection have additive effects on one another, further increasing the disease risks of cardiovascular disease, osteoporosis, and progression of cervical dysplasia. The effects of menopause on HIV infection itself seems limited. While some data suggest an increased risk of acquisition in non–HIV-infected menopausal women, menopause has no effect on the transmission or progression of HIV in menopausal HIV-infected women.
  • Conclusion: As HIV-infected individuals live longer, practitioners will encounter an increasing number of women entering menopause and living into their postmenopausal years. Future studies on the age of menopause, symptoms of menopause, and the effects of menopause on long term comorbidities such as cognitive decline, cardiovascular disease, and bone density loss are necessary to improve care of this expanding population of women living with HIV.

 

Since the introduction of highly active antiretroviral therapy (HAART) in 1996, there has been a significant decrease in morbidity and mortality worldwide among individuals living with human immunodeficiency virus (HIV) [1]. It is projected that by the year 2020, half of persons living with HIV infection in the United States will be over the age of 50 years [2]. For HIV-infected women, this longer survival translates into an increased number of women entering into menopause and living well beyond menopause. Enhancing our knowledge about menopause in HIV-infected women is important since the physiologic changes associated with menopause impact short- and long-term quality of life and mortality. Symptoms associated with menopause can be mistaken for symptoms suggestive of infections, cancers, and drug toxicity. Furthermore, changes in cognition, body composition, lipids, glucose metabolism, and bone mass are influential factors determining morbidity and mortality in later years.

 

 

 

Effect of HIV on the Menstrual Cycle

Menstrual irregularities, including amenorrhea and anovulation, are more frequently found in women of low socioeconomic class who experience more social and physical stress like poverty and physical illnesses [3]. In addition, women with low body mass index (BMI) have decreased serum estradiol levels which lead to amenorrhea [3,4]. Furthermore, several studies have demonstrated that methadone, heroin, and morphine use are associated with amenorrhea. Opiate use inhibits the central neural reproductive drive leading to amenorrhea even in the absence of menopause [5–7].

As these demographics, body habitus, and lifestyle characteristics are frequently found among HIV-infected women, it is not surprising that amenorrhea and anovulation are common in this population [8–14]. In fact, studies show that there is an increased prevalence of amenorrhea and anovulation among HIV-infected women when compared to non–HIV-infected women [8]. Some studies suggest that women with lower CD4 cell counts and higher viral loads have increased frequency of amenorrhea and irregular menstruation compared to those with higher CD4 cell counts and lower viral loads [9,10]. However, it remains unclear if HIV infection itself, instead of the associated social and medical factors, is responsible for the higher frequency of amenorrhea [11–13]. For example, in a prospective study comparing 802 HIV-infected women with 273 non–HIV-infected women, there was no difference in the prevalence of amenorrhea when controlling for BMI, substance use, and age [13].

The World Health Organization (WHO) currently defines natural menopause as the permanent cessation of menstruation for 12 consecutive months without any obvious pathological or physiologic causes [15]. However, given the increased prevalence of amenorrhea in HIV-infected women, amenorrhea seen with HIV infection can be mistaken for menopause. The Women’s Interagency HIV Study (WIHS), a multicenter, observational study of HIV-infected women and non–HIV-infected women of similar socioeconomic status, found that more than half of HIV-infected women with prolonged amenorrhea of at least 1 year had serum follicle-stimulating hormone (FSH) levels in the premenopausal range of less than 25 mIU/mL [16]. Hence, this implies that some of these women may have had prolonged amenorrhea rather than menopause [17]. The traditional definition of menopause may need to be altered in this population.

Age at Menopause

Natural menopause, retrospectively determined by the cessation of menstrual cycles for 12 consecutive months, is a reflection of complete, or near complete, ovarian follicular depletion with subsequent low estrogen levels and high FSH concentrations [18]. In the United States, studies have found the mean age of menopause to be between 50 to 52 years old [19,20].  These studies, however, focused predominantly on menopause in middle class, white women. Early menopause, defined as the permanent cessation of menstruation between 40 to 45 years of age, affects 5% of the women in the United States, while premature menopause or primary ovarian insufficiency, which occurs at younger than 40 years of age, affects 1% of the women [21].

As earlier menopause is associated with increased risks of diabetes [22], cardiovascular disease [23]stroke [24], and osteoporosis [25], identifying the mean age of menopause is important in the management of HIV-infected women. Among women in the United States, early menopause has been observed in women who are African American, nulliparous, have lower BMI, smoke tobacco, and have more stress, less education, and more unemployment [26–29]. Unhealthy lifestyles can also contribute to an earlier age of menopause. Smoking is one of the most consistent and modifiable risk factors associated with an earlier onset of natural menopause, accelerating menopause by up to 2 years [26,30]. Substances present in cigarettes are associated with irreversible damage of ovarian follicles and impaired liver estrogen metabolism [30]. Cocaine use has also been associated with lower estradiol levels, suggesting possible ovary-toxic effects [7,31].

Many of these characteristics and unhealthy lifestyles are prevalent among HIV-infected women. Prevalence of current smoking among HIV-infected persons is found to be approximately 42% [32] in comparison with the 19% seen in the general population in the United States [33]. Specifically, among women participating in WIHS, 56% of the women were found to be current smokers with an additional 16% of the women found to be prior smokers [34]. In addition, African Americans account for the highest proportion of new HIV infections in the United States with an estimated 64% of all new HIV infections in women found to be in African Americans [35]. Furthermore, HIV-infected women are of lower socioeconomic status, with increased prevalence of substance use than that typically found in women enrolled in studies on the age of menopause [36]. Hence, when examining the influence of HIV on the age of menopause, one needs to have a comparator of non–HIV-infected group with similar characteristics. Studies without comparison groups have reported the median age of menopause in HIV-infected women to be between 47 and 50 years old [37–42].

There are only few studies that have focused on the age of menopause in HIV-infected women with a similar comparative non–HIV-infected group.Cejtin et al studied the age of menopause in women enrolled in the WIHS [43]. HIV-infected women partaking in the WIHS were primarily African American and of lower socioeconomic status with heterosexual transmission rather than injection drug use as the major HIV risk factor [44]. They found no significant difference in the median age of menopause when HIV-infected women were compared to non–HIV-infected women. Median age of menopause was 47.7 years in HIV-infected women and 48.0 years in non–HIV-infected women [43].

In contrast, in the Ms Study, a prospective cohort comparing 302 HIV-infected with 259 non-HIV-infected women, HIV-infected women were 73% more likely to experience early menopause than non-HIV-infected women [45]. Similar to the WIHS, there was a high prevalence of African Americans but unlike the WIHS the majority of participants had used heroin or cocaine within the past 5 years. The high prevalence of drug use and current or former cigarette use in the Ms Study likely contributed to the relatively early onset of menopause. Furthermore, the WIHS and Ms Study used different definition of menopause. The WIHS defined menopause as 6 consecutive months of amenorrhea with an FSH level greater than 25 mIU/mL while the Ms Study defined menopause as the cessation of menstrual period for 12 consecutive months [43,45]. Given the fact that 52% of the women in the Ms Study had high-risk behaviors associated with amenorrhea and that menopause was defined as 12 months of amenorrhea without corresponding FSH levels, it is possible that the Ms Study included many women with amenorrhea who had not yet reached menopause. On the other hand, although the 6 months’ duration of amenorrhea used in the WIHS to define menopause had the potential to include women who only had amenorrhea without menopause, the use of FSH levels to define menopause most likely eliminated women who only had amenorrhea.

HIV-infected women have several factors associated with early menopause which are similar to that in the general population, including African American race, injection drug use, cigarette smoking, and menarche before age of 11 [37,41]. In addition, multiple studies have shown that a key factor associated with early age of menopause among HIV-infected women is the degree of immunosuppression [37,41,45]. The Ms Study found that women with CD4 cell counts < 200 cells/mmhad an increased risk ofamenorrhea lasting at least 12 months when compared to women with CD4 cell counts ≥ 200 cells/mm3. The median age of menopause was 42.5 years in women with CD4 cell counts < 200 cells/mm3, 46.0 years in women with CD4 cell counts between 200 cells/mmand 500 cells/mm3, and 46.5 years in women with CD4 cell counts > 500 cells/mm[45]. Similarly, in a cohort of 667 Brazilian HIV-infected women, among whom 160 women were postmenopausal, Calvet et al found 33% of women with CD4 cell counts < 50 cells/mmto have premature menopause, compared to 8% of women with CD4 cell counts ≥ 350 cells/mm[41]. De Pommerol et al  studied 404 HIV-infected women among whom 69 were found to be postmenopausal. They found that women with CD4 cell counts < 200 cells/mmwere more likely to have premature menopause compared to women with CD4 cell counts ≥ 350 cells/mm[37].

Besides the degree of immunosuppression, another factor contributing to early menopause unique to HIV-infected women is chronic hepatitis C infection [41].

 

 

 

Menopause-Associated Symptoms

The perimenopausal period, which begins on average 4 years prior to the final menstrual period, is characterized by hormonal fluctuations leading to irregular menstrual cycles. Symptoms associated with these physiologic changes during the perimenopausal period include vasomotor symptoms (hot flashes), genitourinary symptoms (vaginal dryness and dyspareunia), anxiety, depression, sleep disturbances, and joint aches [46–53]. Such menopausal symptoms can be distressing, negatively impacting quality of life [54].

It can be difficult to determine which symptoms are caused by the physiologic changes of menopause in HIV-infected women as they have multiple potential reasons for these symptoms, such as antiretroviral therapy, comorbidities, and HIV infection itself [55]. However, several studies clearly show that there are symptoms that occur more commonly in the perimenopausal period and that HIV-infected women experience these symptoms earlier and with greater intensity [38–40,42,56,57]. In a cross-sectional study of 536 women among whom 54% were HIV-infected, Miller et al found that menopausal symptoms were reported significantly more frequently in HIV-infected women compared with non–HIV-infected women [56]. As symptoms can occur in greater intensity and impair quality of life, it is important that providers be able to recognize, understand, and appropriately treat menopausal symptoms in HIV-infected women.

Vasomotor Symptoms

In the United States the most common symptom during perimenopause is hot flashes, which occur in 38% to 80% of women [58,59]. Vasomotor symptoms are most common in women who smoke, use illicit substances, have a high BMI, are of lower socioeconomic status, and are African American [19]. As expected, prior studies focusing on hot flash prevalence among premenopausal, perimenopausal, and postmenopausal HIV-infected women found that postmenopausal women experience more hot flashes than premenopausal or perimenopausal women [40,42]. In addition, a comparison of HIV-infected and non–HIV-infected women demonstrated a higher prevalence of hot flashes among HIV-infected women [38,56]. Ferreira et al found that 78% of Brazilian HIV-infected women reported vasomotor symptoms compared to 60% of non–HIV-infected women [38]. Similarly, Miller et al reported that 64% of HIV-infected women reported vasomotor symptoms compared to 58% of non–HIV-infected women [56].

Vasomotor symptoms can be severely distressing with hot flashes contributing to increased risk of depression [56,60]. In a cross-sectional analysis of 835 HIV-infected and 335 non–HIV-infected women from the WIHS, persistent vasomotor symptoms predicted elevated depressive symptoms in both HIV-infected and non-HIV-infected women [60]. In a similar cross-sectional analysis of 536 women, among whom 54% were HIV positive and 37% were perimenopausal, psychological symptoms were prevalent in 61% of the women with vasomotor symptoms [56].

Oddly enough, higher CD4 cell counts appear to be associated with increased prevalence of vasomotor symptoms [39,56]. Clark et al demonstrated that menopausal HIV-infected women with CD4 cell counts > 500 cells/mmwere more likely to report hot flashes [39]. Similarly, Miller et al observed a reduction in the prevalence of menopausal symptoms as CD4 cell counts declined among HIV-infected non-HAART users [56]. The rationale behind this is unclear but some experts postulated that it may be due to the effects of HAART.

Genitourinary Symptoms

With estrogen deficiency, which accompanies the perimenopausal period, vulvovaginal atrophy (VVA) occurs leading to symptoms of vaginal dryness, itching, burning, urgency, and dyspareunia (painful intercourse) [59,61,62]. Unlike vasomotor symptoms, which diminish with time, genitourinary symptoms generally worsen if left untreated [63]. Furthermore, these symptoms are often underreported and underdiagnosed [64,65]. Several studies using telephone and online surveys have found that the prevalence of symptoms of VVA is between 43% and 63% in postmenopausal women [66–69]. Even higher rates were found in the Agata Study in which pelvic exams in 913 Italian women were performed to obtain objective signs of VVA [62]. The prevalence of VVA was 64% 1 year after menopause and 84% 6 years after menopause. Vaginal dryness was found in 100% of participants with VVA or 82% of total study participants. In addition, 77% of women with VVA, or 40% of total study participants, reported dyspareunia.

Genitourinary symptoms are most common among women who are African American, have an increased BMI, are from lower socioeconomic class, use tobacco [19], have prior history of pelvic inflammatory disease, and have anxiety and depression [70,71]. Similarly to hot flashes, many of these predisposing factors are more common in HIV-infected women. Fantry et al found that 49.6% of HIV-infected women had vaginal dryness. Although 56% of postmenopausal women and 36% of perimenopausal women complained of vaginal dryness, in a multivariate analysis only cocaine use, which can decrease estradiol levels [7,31] was associated with a higher frequency of vaginal dryness [40].

Similarly, dyspareunia is also common among HIV-infected women. In a cross-sectional study of 178 non–HIV-infected and 128 HIV-infected women between 40 and 60 years of age, Valadares et al found that the frequency of dyspareunia in HIV-infected women was high at 41.8% [72]. However, this was not significantly higher compared to the prevalence of 34.8% in non–HIV-infected women. HIV infection itself was not associated with the presence of dyspareunia

Psychiatric Symptoms

Anxiety and depression are also common symptoms in perimenopausal women [73–76]. Studies have shown that depression is diagnosed 2.5 times more frequently among perimenopausal than premenopausal women [76].

In a study by Miller et al that focused on 536 HIV-infected women, among whom 37% were perimenopausal, 89% reported psychological symptoms [56]. Ferreira et al found that HIV-infected perimenopausal women had an increased incidence of psychological symptoms compared to non–HIV-infected women [38]. Whether this increased prevalence of psychological symptoms seen in HIV-infected women can be attributed to menopause is unclear since one third to one half of men and women living with HIV experience symptoms of depression [77]. However, in the WIHS, which compared 835 HIV-infected with 335 non-HIV-infected women from all menopausal stages, elevated depressive symptoms were seen in the early perimenopausal period [60]. There was no increased incidence of such symptoms during the premenopausal or postmenopausal period, suggesting the contribution of menopause to depressive symptoms during the perimenopausal period [60].

Persistent menopausal symptoms, especially hot flashes, also predicted elevated depressive symptoms in several studies [56,60] suggesting the importance of appropriately identifying and treating menopausal symptoms. In addition, cognitive decline associated with menopause contributes to depression [78–80].

Other Symptoms

Sleep disturbances are also common among perimenopausal women, with prevalence estimated to be between 38% and 46% [81–84]. Hot flashes, anxiety, and depression appear to be contributing factors [81–84]. In a cross-sectional study of 273 HIV-infected and 264 non-HIV-infected women between 40 and 60 years of age, insomnia was found in 51% of perimenopausal and 53% of postmenopausal HIV-infected women. HIV-infected women had the same prevalence of insomnia compared to non–HIV-infected women [85]. Joint aches are also commonly reported in the perimenopausal period, with prevalence as high as 50% to 60% among perimenopausal women in the United States [52,53]. In HIV-infected women, Miller et al found that 63% of menopausal women reported arthralgia [56].

Treatment

For women experiencing severe hot flashes and vaginal dryness, short-term menopausal hormone therapy (MHT) is indicated to relieve symptoms. MHT should be limited to the shortest period of time at the lowest effective dose as MHT is associated with increased risks of breast cancer, cardiovascular disease, thromboembolism, and increased morbidity [86]. Despite the increased severity of menopausal symptoms experienced among HIV-infected women, the prevalence of the use of MHT in this population is lower compared to non–HIV-infected women [85].

Topical treatment is recommended for women who are experiencing solely vaginal atrophy. First-line treatment is topical nonhormonal therapy such as moisturizers and lubricants [87]. If symptoms are not relieved, then topical vaginal estrogen therapy is recommended [87]. Although topical therapy can result in estrogen absorption into the circulation, it is to a much lesser extent than systemic estrogen therapy [88].

Overall, there is lack of data on the potential interactions between MHT and HAART. Much of the potential interactions are inferred from pharmacokinetic and pharmacodynamics studies between HAART and oral contraceptives. Hormone therapy, protease inhibitors (PIs), colbicistat, and non-nucleoside reverse transcriptase inhibitors (NNRTIs) are all metabolized by the CYP3A4 enzyme [89–91]. Current evidence suggests that concomitant use of hormone therapy with NNRTIs and PIs does not significantly alter the pharmacokinetics of HAART or the clinical outcomes of HIV [91]. However, there is evidence that concomitant use of nevirapine and PIs boosted with ritonavir leads to decrease in estrogen levels so higher doses of MHT may have to be used to achieve symptomatic relief [91]. There is no data on the interaction between PIs boosted with colbicistat and estrogen [92]. Integrase inhibitors, nucleoside and nucleotide reverse transcriptase inhibitors (NRTIs), and the CCR5 antagonist maraviroc have no significant interactions with estrogen containing compounds [89,90,92].

Cardiovascular Risk

Estrogen deficiency resulting from menopause leads to several long-term effects, including cardiovascular disease and osteoporosis. The loss of protective effects of estrogen leads to an increased risk of cardiovascular disease particularly with changes in lipid profiles [93]. Perimenopausal women experience changes in body composition with increased fat mass and waist circumference, as well as dyslipidemia and insulin resistance, all of which are associated with higher risk of cardiovascular disease [94].

HIV infection also incurs a higher risk of cardiovascular disease [95–99]. The inflammatory effects of HIV, HAART, and traditional risk factors including dyslipidemia all contribute to cardiovascular disease but the degree to which each factor contributes to elevated risk is unknown [95,98]. In addition, modifiable risk factors for cardiovascular disease such as decreased fitness and smoking are more commonly seen in HIV-infected women [100]. Even prior to menopause, HIV-infected women experience lipodystrophy syndrome with increase in truncal visceral adiposity and decrease in subcutaneous fat and muscle mass [101,102]. Whether such changes in body composition are exacerbated during the perimenopausal period remain unclear. In the SWEET study, which focused on 702 South African women among whom 21% were HIV-infected, there was lower lean mass but minimal difference in the fat mass of postmenopausal women compared to premenopausal women [103]. As the study was based in South Africa with only 21% HIV-infected, the results of this study should be viewed with caution. While changes in body composition were not observed in postmenopausal women in the SWEET study, increased truncal adiposity seen in premenopausal HIV-infected women is likely to pose an additional risk for cardiovascular disease during the menopause transition.

Several studies have been conducted to demonstrate an increased risk of cardiovascular disease, especially among young HIV-infected men [95–99]. However, no study has focused specifically on the risk of cardiovascular disease in postmenopausal HIV-infected women to date. Despite the lack of studies, it is plausible that the increased risk of cardiovascular disease seen in HIV infection is likely to be compounded with the increased risk seen during menopause. Postmenopausal HIV-infected women may be at significantly higher risk of cardiovascular disease. Appropriate measures such as lipid control, antiplatelet therapy, smoking cessation, and other lifestyle changes should be initiated as in any other population. Further studies are necessary focusing on the effects of menopause on cardiovascular disease risk in HIV-infected women.

 

 

 

Osteoporosis

Menopause, with its associated estrogen deficiency, is the most important risk factor associated with increased bone turnover and bone loss and can worsen HIV associated bone loss [104]. Among HIV-infected individuals, low bone mineral density (BMD) has been described even among premenopausal women and younger men [105–107]. Evidence suggests that the decreased BMD associated with HIV stabilizes or even improves after initiation of HAART in the younger population [105–107]. However, once HIV-infected women enter menopause, they have higher rates of bone loss compared to non–HIV-infected women with significantly increased prevalence of osteoporosis compared to non–HIV-infected women [108–112].

Chronic inflammation by HIV stimulates osteoclast differentiation and resorption [113]. In addition, HAART [114–116], vitamin D deficiency [117], low BMI, poor nutrition [118], inactivity, use of tobacco, alcohol, and illicit drugs [119,120], and coinfection with hepatitis B and C [121] all appear to contribute to decreased BMD among HIV-infected men and women [118]. Among HIV-infected postmenopausal women, those taking ritonavir were found to have increased differentiation of osteoclast cells and increased bone loss [122]. Similarly, methadone use in postmenopausal women has been associated with increased BMD decline [123]. African-American, HIV-infected postmenopausal women appear to be at the greatest risk for bone loss [109].

Multiple studies focusing on HIV-infected men have demonstrated an increased prevalence of fractures compared to non–HIV-infected men [124–126]. However, current studies on postmenopausal HIV-infected women demonstrate that fracture incidence is similar between HIV-infected and non–HIV-infected postmenopausal women [108,112]. Nevertheless, given the evidence of low BMD and increased fracture risk seen during menopause among non–HIV-infected women compounded with the additional bone loss seen in HIV-infected individuals, enhanced screening in postmenopausal HIV-infected women is prudent. Although the U.S. Preventive Services Task Force (USPSTF) makes no mention of HIV as a risk factor for enhanced screening [127] and the Infectious Diseases Society of America (IDSA) only recommends screening beginning at the age of 50 years old if there are additional risk factors other than HIV [128], the more recently published Primary care guidelines for the management of persons infected with HIV recommends screening postmenopausal women ≥ 50 years of age with dual-energy X-ray absorptiometry (DEXA) scan [86]. Preventative therapy such as smoking cessation, adequate nutrition, alcohol reduction, weight bearing exercises, and adequate daily vitamin D and calcium should be discussed and recommended in all menopausal HIV-infected women [129]. If the DEXA scan shows osteoporosis, bisphosphonates or other medical therapy should be considered. Although the data are limited, bisphosphonates have been shown to be effective in improving BMD [130–132].

Cognition

The menopause transition is characterized by cognitive changes such as memory loss and difficulty concentrating [133–136]. Both HIV-infected men and women are at higher risk of cognitive impairment [137–139]. Cognitive impairment can range from minor cognitive-motor disorder to HIV-associated dementia due to the immunologic, hormonal, and inflammatory effects of HIV on cognition [137–139]. In addition, those with HIV infection appear to have increased risk factors for cognitive impairment including low education level, psychiatric illnesses, increased social stress, and chemical dependence [137].

Studies focusing on the effects of both HIV infection and menopause on cognition have been limited thus far. In a cross-sectional study of 708 HIV-infected and 278 non–HIV-infected premenopausal, perimenopausal, and postmenopausal women, Rubin et al demonstrated that HIV infection, but not menopausal stage, was associated with worse performance on cognitive measures [140]. While menopausal stage was not associated with cognitive decline, menopausal symptoms like depression, anxiety, and vasomotor symptoms were associated with lower cognitive performance [140].

Though limited, current data appear to indicate that HIV infection, not menopause, contributes to cognitive dysfunction [140]. Symptoms of menopause, however, do appear to exacerbate cognitive decline indicating the importance of recognition and treatment of menopausal symptoms. This is especially important in HIV-infected women since decrease in cognition and depression can interfere with day to day function including medication adherence [141,142].

Cervical Dysplasia

As more HIV-infected women reach older age, the effects of prolonged survival and especially menopause on squamous intraepithelial lesions (SILs) are being investigated to determine if general guidelines of cervical cancer screening should be applied to postmenopausal women.

In a retrospective analysis of Papanicolaou smear results of 245 HIV-infected women, Kim et al noted that menopausal women had a 70% higher risk of progression of SILs than premenopausal women [143]. Similar results were found in a smaller retrospective study of 18 postmenopausal HIV-infected women in which postmenopausal women had a higher prevalence of SILs and persistence of low-grade SILs [144].

Although studies on progression to cervical cancer in postmenopausal HIV-infected women remain limited, current data suggest that postmenopausal HIV-infected women should continue to be monitored and screened similarly to the screening recommendations for premenopausal women. Nevertheless, further studies examining the natural course of cervical lesions are needed to establish the best practice guidelines for screening postmenopausal women.

HIV Acquisition and Transmission

The incidence of new HIV infections in older American women has increased. HIV acquisition from heterosexual contact appears to be higher in older women compared to younger women, with a study suggesting that women over age 45 years had almost a fourfold higher risk of HIV acquisition compared to those under the age of 45 years [145]. While the lack of awareness of HIV risk and less frequent use of protection may contribute to increases in new HIV infection in older women, hormonal changes associated with older age, specifically menopause, may be playing a role. Vaginal wall thinning that occurs during menopause may serve as a risk factor for HIV acquisition.

In a study by Meditz et al, the percentage of endocervical or blood CD4 T cells did not differ between premenopausal and postmenopausal women, but postmenopausal women had greater percentage of CCR5 expression. As CCR5 serves as an entry point of HIV into target cells, this suggests the possibility that postmenopausal women may be at increased risk for HIV acquisition [146]. More recently, Chappell et al also revealed that anti-HIV-1 activity was significantly decreased in postmenopausal compared to premenopausal women, suggesting that there may be an increased susceptibility to HIV-1 infection in postmenopausal women [147]. Hence there appears to be menopause-related immunologic changes of the cervix that may contribute to an increased risk of HIV acquisition in postmenopausal women.

In contrast, although data is limited, postmenopausal HIV-infected women do not appear to be at increased risk of transmitting HIV to non–HIV-infected individuals. Melo et al compared the intensity of HIV shedding between premenopausal and postmenopausal women and found that HIV shedding did not differ between premenopausal or postmenopausal women [148].

HIV Progression

Several studies have focused on the effects of HIV infection on menopause, but minimal data are available on the effects of menopause on the progression of HIV infection. With prior data suggesting that younger persons experience better immunological and virological responses to HAART [149–151], it has previously been hypothesized that virologic and immunologic responses to HAART can decline once HIV-infected women reach menopause. However, current evidence suggests that treatment responses to HAART, determined by the median changes in CD4 cell counts and percentages and viral load, in HAART-naive patients did not differ between premenopausal and postmenopausal women [152]. In addition, there appears to be no significant changes in CD4 cell counts as HIV-infected women progress through menopause [153]. These studies suggest that menopause does not affect the progression of HIV and that HAART-naive women should respond to HAART regardless of their menopausal status.

Conclusion

As HIV-infected individuals live longer, increasing number of women will enter into menopause and live many years beyond menopause. HIV-infected women experience earlier and more severe menopausal symptoms, but knowledge is still lacking on the appropriate management of these symptoms. In addition, current evidence suggests that immunosuppression associated with HIV contributes to an early onset of menopause which leads to increased risks of cardiovascular disease, osteoporosis, and progression of cervical dysplasia. These conditions require proper surveillance and can be prevented with improved understanding of influences of menopause on HIV-infected women. Furthermore, although there is some evidence suggesting that menopause has no effect on HIV transmission and progression, further studies on the immunologic and virologic effects of menopause are necessary.

There still remain significant gaps in our understanding of menopause in HIV-infected women.  As practitioners encounter an increasing number of perimenopausal and postmenopausal HIV-infected women, future studies on the effects of HIV on co-morbidities and symptoms of menopause and their appropriate management are necessary to improve care of women living with HIV.

 

Corresponding author: Lori E. Fantry, MD, MPH, 29 S. Greene St., Suite 300, Baltimore, MD 21201, lfantry@medicine.umaryland.edu.

Financial disclosures: None.

References

1. CASCADE Collaboration. Survival after introduction of HAART in people with known duration of HIV-1 infection. Lancet 2000;355:1158–9.

2. Brooks JT, Buchaz K, Gebo KA, Mermin J. HIV infection and older Americans: the public health perspective. Am J Pub Health 2012;102:1516–26.

3. Munster K, Helm P, Schmidt L. Secondary amenorrhea: Prevalence and medical contract–A cross sectional study from a Danish county. Br J Obstet Gynecol 1992;99:430–3.

4. Vyver E, Steinegger C, Katzman DK, et al. Eating disorders and menstrual dysfunction in adolescents. Ann N Y Acad Sci 2008;1135:253–64.

5. Abs R, Verhelst J, Maeyaert J, et al. Endocrine consequences of long-term intrathecal administration of opioids. J Clin Endocrinol Metab 2000;85:2215–22.

6. Pelosi MA, Sama JC, Caterini H, et al. Galactorrhea-amenorrhea syndrome associated with heroin addiction. Am J Obstet Gynecol 1974;118:966–70.

7. Bai J, Greenwald E, Caterini H, et al. Drug-related menstrual aberrations. Obstet Gynecol 1974;44:713–9.

8. Chirgwin KD, Feldman J, Muneyyirci-Delale O, et al. Menstrual function in HIV-infected women without AIDS. J Acquir Immune Defic Syndr Hum Retrovirol 1996;12:489–94.

9. Clark RA, Mulligan K, Stamenovic E, et al. Frequency of anovulation and early menopause among women enrolled in selected adult AIDS clinical trials group studies. J Infect Dis 2001;184:1325–7.

10. Watts DH, Spino C, Zaborski L. Comparison of gynecologic history and laboratory results in HIV-positive women with CDR+ lymphocyte counts between 200 and 500 cells/µl and below 100 cells/ µl. J Acquir Immune Defic Syndr Hum Retrovirol 1999;20:455–62.

11. Ellerbrock TV, Wrig TC, Bush TJ, et al. Characteristics of menstruation in women infected with HIV. Obstet Gynecol 1996;87:1030–4.

12. Shah PN, Smith JR, Wells C, et al. Menstrual symptoms in women infected by the HIV. Obstet Gynecol 1994;83:397–400.

13. Harlow SC, Schuman P, Cohen M, et al. Effect of HIV infection on menstrual cycle length. J Acquir Immune Defic Syndr Hum Retrovirol 2000;24:68–75.

14. Grinspoon S, Corocran C, Miller K, et al. Bone composition and endocrine function in women with AIDS wasting. J Clin Edocrinol Metab 1997;82:1332–7.

15. Research on the menopause in the 1990s. Report of a WHO scientific group. World Health Organ Tech Rep Ser 1996;866:1–107.

16. Cejtin HE, Kalinowski A, Bacchetti P. Effects of human immunodeficiency virus on protracted amenorrhea and ovarian dysfunction. Obstet Gynecol 2006;108:1423–31.

17. Freeman EW, Sammel MD, Garcia CR, et al.  Follicular phase hormone levels and menstrual bleeding status in the approach to menopause. Fertil Steril 2005;83:383–92.

18. Soules MR, Sherman S, Parrott E, et al. Executive summary: Stages of Reproductive Aging Workshop (STRAW). Fertil Steril 2001;76:874–8.

19. Gold EB, Crawford SL, Avis NE, et al. Factors related to age at natural menopause: longitudinal analyses from SWAN. Am J Epidemiol 2013;178:70–83.

20. Thomas F, Renaud F, Benefice E, et al. International variability of ages at menarche and menopause: patterns and main determinants. Hum Biol 2001;73:271–90.

21. Shuster LT, Rhodes DJ, Gostout BS, et al. Premature menopause or early menopause: long-term health consequences. Maturitas 2010;65:161–6.

22. Carr MC. The emergence of the metabolic syndrome with menopause. J Clin Endocrinol Metab 2003;88:2404–11.

23. Wellons M, Ouyang P, Schreiner PJ, et al. Early menopause predicts future coronary heart disease and stroke: the multi-ethnic study of atherosclerosis. Menopause 2012;19:1081–7.

24. Rocca WA, Grossardt BR, Miller VM, et al. Premature menopause or early menopause and risk of ischemic stroke. Menopause 2012;19:272–7.

25. Svejme O, Ahlborg HG, Nilsson JA, et al. Early menopause and risk of osteoporosis, fracture and mortality: a 34-year prospective observational study in 390 women. BJOG 2012;119:810–6.

26. Cooper GS, Sandler DP, Bohlig M. Active and passive smoking and the occurrence of natural menopause. Epidemiology 1999;10:771–3.

27. Luoto R, Kaprio J, Uutela A. Age at natural menopause and socioeconomic status in Finland. Am J Epidemiol 1994;139:64–76.

28. Bromberger JT, Matthews KA, Kuller LH, et al. Prospective study of the determinants of age at menopause. Am J Epidemiol 1997;145:24–33.

29. Gold EB, Crawford SL, Avis NE, et al. Factors related to age at natural menopause: longitudinal analyses from SWAN. Am J Epidemiol 2013;178:70–83.

30. Tziomalos K, Charsoulis F. Endocrine effects of tobacco smoking. Clin Endocrinol 2004;61:664–74.

31. Potter DA, Moreno A, Luther MF, et al. Effects of follicular-phase cocaine administration on menstrual and ovarian cyclicity in rhesus monkeys. Am J Obstet Gynecol 1998;178:118–25.

32. Mdodo R, Frazier EL, Dube SR, et al. Cigarette smoking prevalence among adults with HIV compared with the general adult population in the United States: Cross-sectional survey. Ann Intern Med 2015;162:335–44.

33. Centers for Disease Control and Prevention (CDC). Vital signs: current cigarette smoking among adults aged ≥ 18 years–United States, 2005-2010. MMWR Morb Mortal Wkly Rep 2011;60:1207–12.

34. Feldman J, Mikoff H, Schneider M, et al. Association of cigarette smoking with HIV prognosis among women in the HAART era: A report from the Women’s Interagency HIV study. Am J Public Health 2006:96:1060–5.

35. Centers for Disease Control and Prevention. Estimated HIV incidence among adults and adolescents in the United States, 2007–2010. HIV Surveillance Supplemental Report 2012;17(4).

36. Galea S, Ahren J, Vlahov D. Contextual determinants of drug use risk behavior: a theoretical framework. J Urban Health 2003;80:50–8.

37. de Pommerol M, Hessamfar M, Lawson-Ayayi S, et al. Menopause and HIV infection: age at onset and associated factors, ANRS CO3 Aquitaine cohort. Int J STD AIDS 2011;22:67–72.

38. Ferreira CE, Pinto-Neto AM, Conde DM, et al. Menopausal symptoms in women infected with HIV: prevalence and associated factors. Gynecol Endocrinol 2007;23:198–205.

39. Clark RA, Cohn SE, Jarck C, et al. Perimenopausal symptomatology among HIV infected women at least 40 years of age. J Acquir Immune Defic Syndr Hum Retrovirol 2000;23:99–100.

40. Fantry L, Zhan M, Taylor G, et al. Age at menopause and menopausal symptoms in HIV-infected women. AIDS Patient Care STD 2005;19:703–11.

41. Calvet G, Grinsztejn G. Predictors of early menopause in HIV infected women: a prospective cohort study. Am J Obstet Gynecol 2015;212:765.

42. Boonyanurak P, Bunupuradah T, Wilawan K, et al. Age at menopause and menopause-related symptoms in human immunodeficiency virus-infected Thai women. Menopause 2012;19:820–4.

43. Cejtin SH, Taylor R, Watts DH. Assessment of menopausal status among women in the Women’s Interagency HIV study (WIHS). Proceedings of the 57th International AIDS Conference 2004; Bangkok, Thailand.

44. WIHS Data Management and Analysis Center (WDMAC). Women’s Interagency HIV Study (WIHS) Dossier. October 2014. Available at https://statepiaps.jhsph.edu/wihs/invest-info/dossier.pdf.

45. Schoenbaum E, Hartel D, Lo Y, et al. HIV infection, drug use, and onset of natural menopause. Clinical Infect Dis 2005;41:1517–24.

46. Taffe JR, Dennerstein L. Menstrual patterns leading to the final menstrual period. Menopause 2002;9:32–40.

47. Miro F, Parker SW, Aspinall LJ, et al. Origins and consequences of the elongation of the human menstrual cycle during the menopausal transition: the FREEDOM Study. J Clin Endocrinol Metab 2004;89:4910–5.

48. Harlow SD, Gass M, Hall JE, et al. Executive summary of the Stages of Reproductive Aging Workshop + 10: addressing the unfinished agenda of staging reproductive aging. J Clin Endocrinol Metab 2012;97:1159–68.

49. Freeman EW, Sammel MD, Gracia CR, et al. Follicular phase hormone levels and menstrual bleeding status in the approach to menopause. Fertil Steril 2005;83:383–92.

50. Burger HG, Hale GE, Dennerstein L, Robertson DM. Cycle and hormone changes during perimenopause: the key role of ovarian function. Menopause 2008;15:603–12.

51. McKinlay SM, Brambilla DJ, Posner JG. The normal menopause transition. Maturitas 1992;14:103–15.

52. Szoeke CE, Cicuttini F, Guthrie J, Dennerstein L. Self-reported arthritis and the menopause. Climacteric 2005;8:49–55.

53. Blümel JE, Chedraui P, Baron G, et al. Menopause could be involved in the pathogenesis of muscle and joint aches in mid-aged women. Maturitas 2013;75:94–100.

54. Woods NF, Mitchell ES. Symptoms interference with work and relationships during the menopausal transition and early postmenopause: observations from the Seattle Midlife Women’s Health Study. Menopause 2011;18:654–61.

55. Johnson TM, Cohen HW, Howard AA, et al. Attribution of menopause symptoms in human immunodeficiency virus–infected or at-risk drug-using women Menopause 2008;15:551–7.

56. Miller SA, Santoro N, Lo Y. Menopausal symptoms in HIV-infected and drug-using women. Menopause 2005;12:348–56.

57. Looby S, Shifren J, Corless I. Increased hot flash severity and related interference in perimenopausal HIV-infected women. Menopause 2014;21:403–9.

58. Thurston RC, Joffe H. Vasomotor symptoms and menopause: findings from the Study of Women’s Health across the Nation. Obstet Gynecol Clin North Am 2011;38:489–501.

59. Woods NF, Mitchell ES. Symptoms during the perimenopause: prevalence, severity, trajectory, and significance in women’s lives. Am J Med 2005;118 Suppl 12B:14.

60. Maki PM, Rubin LH, Cohen M, et al. Depressive symptoms are increased in the early perimenopausal stage in ethnically diverse human immunodeficiency virus-infected and human immunodeficiency virus-uninfected women. Menopause 2012;19:1215–33.

61. Dennerstein L, Dudley EC, Hopper JL, et al. A prospective population-based study of menopausal symptoms. Obstet Gynecol 2000;96:351–8.

62. Palma F, Volpe A, Villa P, et al. Vaginal atrophy of women in postmenopause. Results from a multicentric observational study: The AGATA study. Maturitas 2015 Sep 14.

63. Cutler WB, Garcia CR, McCoy N. Perimenopausal sexuality.  Arch Sex Behav 1987;16:225–34.

64. Moreira ED, Glasser DB, Nicolosi A, et al. GSSAB Investigators’ Group.  Sexual problems and help-seeking behavior in adults in the United Kingdom and continental Europe. BJU Int 2008;101:1005–11.

65. MacBride MB, Rhodes DJ, Shuster LT. Vulvovaginal atrophy.  Mayo Clin Proc 2010;85:87–94.

66. Nappi RE, Kokot-Kierepa M. Women’s voices in the menopause: results from an international survey on vaginal atrophy. Maturitas 2010;67:233–8.

67. Santoro N, Komi J. Prevalence and impact of vaginal symptoms among postmenopausal women. J Sex Med 2009;6:2133–42.

68. Levine KB, Williams RE, Hartmann KE. Vulvovaginal atrophy is strongly associated with female sexual dysfunction among sexually active post-menopausal women. Menopause 2008;15(4 Pt 1):661–6.

69. Cumming GP, Currie HD, Moncur R, Lee AJ. Web-based survey on the effect of menopause on women’s libido in a computer-literate population. Menopause Int 2009;15:8–12.

70. Valadares AL, Pinto-Neto AM, Conde DM, et al. A population-based study of dyspareunia in a cohort of middle-aged Brazilian women. Menopause 2008;15:1184–90.

71. Latthe P, Migini L, Gray R, et al. Factors predisposing women to chronic pelvic pain: a systemic review. BMJ 2006;332:749–55.

72. Valadares AL, Pinto-Neto AM, Gomes D, et al. Dyspareunia in HIV-positive and HIV-negative middle-aged women: a cross-sectional study. BMJ Open 2014;4:e004974.

73. Bromberger JT, Meyer PM, Kravitz HM, et al. Psychologic distress and natural menopause: a multiethnic community study. Am J Public Health 2001;91:1435–42.

74. Avis NE, Brambilla D, McKinlay SM, Vass K. A longitudinal analysis of the association between menopause and depression. Results from the Massachusetts Women’s Health Study. Ann Epidemiol 1994;4:214–20.

75. Cohen LS, Soares CN, Joffe H. Diagnosis and management of mood disorders during the menopausal transition. Am J Med 2005;118 Suppl 12B:93–7.

76. Freeman EW, Sammel MD, Lin H, Nelson DB. Associations of hormones and menopausal status with depressed mood in women with no history of depression. Arch Gen Psychiatry 2006;63:375–82.

77. Eller LS, Corless I, Bunch EH, et al. Self-care strategies for depressive symptoms in people with HIV disease. J Adv Nurs 2005;51:119–30.

78. Fuh JL, Wang SJ, Lee SJ, et al. A longitudinal study of cognition change during early menopausal transition in a rural community. Maturitas 2006;53:447–53.

79. Greendale GA, Huang MH, Wight RG, et al. Effects of the menopause transition and hormone use on cognitive performance in midlife women. Neurology 2009;72:1850–7.

80. Hinkin CH, Castellon SA, Atkinson JH, et al. Neuropsychiatric aspects of HIV infection among older adults. J Clin Epidemiol 2001;54:S44–52.

81. Kravitz HM, Ganz PA, Bromberger J, et al. Sleep difficulty in women at midlife: a community survey of sleep and the menopausal transition. Menopause 2003;10:19–28.

82. Freedman RR, Roehrs TA. Effects of REM sleep and ambient temperature on hot flash-induced sleep disturbance. Menopause 2006;13:576–83.

83. Erlik Y, Tataryn IV, Meldrum DR, et al. Association of waking episodes with menopausal hot flushes. JAMA 1981; 245:1741–4.

84. Freedman RR, Roehrs TA. Sleep disturbance in menopause. Menopause 2007;14:826–9.

85. Lui-Filho JF, Valadares AR, Gomes D, et al. Menopausal symptoms and associated factors in HIV-positive women. Maturitas 2013;76:172–8.

86. Aberg JA, Gallant JE, Ghanem KG, et al, Infectious Diseases Society of America. Primary care guidelines for the management of persons infected with HIV: 2013 update by the HIV medicine association of the Infectious Diseases Society of America. Clin Infect Dis 2014;58:e1–34.

87. The role of local vaginal estrogen for treatment of vaginal atrophy in postmenopausal women: 2007 position statement of The North American Menopause Society. Menopause 2007;14:357–69.

88. Dorr MB, Nelson AL, Mayer PR, et al. Plasma estrogen concentrations after oral and vaginal estrogen administration in women with atrophic vaginitis. Fertil Steril 2010;94:2365–8.

89. El-Ibiary SY, Cocohoba JM. Effects of antiretrovirals on the pharmacokinetics of hormonal contraception. Eur J Contracept Reprod Health Care 2008;13:123–32.

90. Tittle V, Bull L, Boffito M, Nwokolo N. Pharmacokinetic and pharmacodynamics drug interactions between antiretrovirals and oral contraceptives. Clin Pharmacokinet 2015;54:23–34.

91. Thurman AR, Anderson S, Doncel G. Effects of hormonal contraception on anti-retroviral drug metabolism, pharmacokinetics and pharmacodynamics. Am J Reprod Immunol 2014:71:523–30.

92. Panel on Antiretroviral Guidelines for Adults and Adolescents. Guidelines for the use of antiretroviral agents in HIV-1-infected adults and adolescents. US. Department of Health and Human Services. Availabe at www.aidsinfo.nih.gov/ContentFiles/AdultandAdolescentGL.pdf.

93. Berg G, Mesch V, Boero L, et al. Lipid and lipoprotein profile in menopausal transition: effects of hormones, age and fat distribution. Hormone Metab Res 2004;36:215–20.

94. Sower M, Zheng H, Tomey K, et al. Changes in body composition in women over six years at midlife: ovarian and chronological aging. J Clin Endocrin Metab 2007;92:895–901.

95. Flooris-Moore M, Howard AA, Lo Y, et al. Increased serum lipids are associated with higher CD4 lymphocyte count in HIV-infected women. HIV Med 2006;7:421–30.

96. Grunfeld C, Delaney JA, Wanke C, et al. Preclinical atherosclerosis due to HIV infection: carotid intima-medial thickness measurements from the FRAM study. AIDS 2009;23:1841–9.

97. Palacios R, Alonso I, Hidalgo A, et al. Peripheral arterial disease in HIV patients older than 50 years of age. AIDS Res Hum Retroviruses 2008;24:1043–6.

98. Hadigan C, Meigs JB, Corcoran C, et al. Metabolic abnormalities and cardiovascular disease risk factors in adults with human immunodeficiency virus infection and lipodystrophy. Clin Infect Dis 2001;32:130–9.

99. Triant VA, Lee H, Hadigan C, Grinspoon SK. Increased acute myocardial infarction rates and cardiovascular risk factors among patients with human immunodeficiency virus disease. J Clin Endocrin Metab 2007;92:2506–12.

100. Dolan SE, Frontera W, Librizzi J et al. The effects of a supervised home based aerobic and progressive resistance training regimen in HIV-infected women: randomized trial. Arch Intern Med 2006;166:1225–31.

101. Grinspoon S, Carr A. Cardiovascular risk and body fat abnormalities in HIV-infected adults. N Engl J Med 2005;352:48–62

102. Study of Fat Redistribution and Metabolic Change in HIV Infection (FRAM). Fat distribution in women with HIV infection. J Acquir Immune Defic Syndr 2006;42:562–71.

103. Jaff NG, Norris SA, Snyman T, et al. Body composition in the study of women entering and in Endocrine Transition (SWEET): A perspective of African women who have a high prevalence of obesity and HIV infection. Metabolism 2015;64:1031–41.

104. Akhter MP, Lappe JM, Davies KM, et al. Transmenopausal changes in the trabecular bone structure. Bone 2007;41:111–6.

105. Cassetti I, Madruga JV, Suleiman JM, et al. The safety and efficacy of tenofovir DF in combination with lamivudine and efavirenz through 6 years in antiretroviral-naive HIV-1-infected patients. HIV Clin Trials 2007;8:164–72.

106. McComsey GA, Kitch D, Daar ES, et al. Bone mineral density and fractures in antiretroviral-naive persons randomized to receive abacavir-lamivudine or tenofovir disoproxil fumarate-emtricitabine along with efavirenz or atazanavir-ritonavir: AIDS Clinical Trials Group A5224s, a substudy of ACTG A5202. J Infect Dis 2011;203:1791–801.

107. Hansen AB, Obel N, Nielsen H, et al. Bone mineral density changes in protease inhibitor-sparing vs. nucleoside reverse transcriptase inhibitor-sparing highly active antiretroviral therapy: Data from a randomized trial. HIV Med 2011;12:157–65.

108. Yin MT, Zhang CA, McMahon DJ, et al. Higher rates of bone loss in postmenopausal HIV-infected women: a longitudinal study. J Clin Endocrinol Metab 2012;97:554–62.

109. Sharma A, Flom PL, Rosen CJ, et al. Racial differences in bone loss and relation to menopause among HIV-infected and uninfected women. Bone 2015;77:24–30.

110. Caputo BV, Traversa-Caputo GC, Costa C, et al. Evaluation of bone alterations in the jaws of HIV-infected menopausal women. Braz Oral Res 2013;27:231–7.

111. Bone mass and mineral metabolism in HIV+ postmenopausal women. Osteoporos Int 2005;26:1345–52.

112. Yin MT, Mcmahon DJ, Ferris DC, et al. Low bone mass and high bone turnover in postmenopausal human immunodeficiency virus-infected women. J Clin Endocrinol Metab 2010;95:620–9.

113. Gibellini D, De Crignis E, Ponti C. HIV-1 triggers apoptosis in primary osteoblasts and HOBIT cells through TNF-alpha activation. J Med Virol 2008;80:1507–14.

114. Tebas P, Powderly WG, Claxton S, et al. Accelerated bone mineral loss in HIV-infected patients receiving potent antiretroviral therapy. AIDS 2000;14:F63–7.

115. Van Rompay KK, Brignolo LL, Meyer DJ, et al. Biological effects of short-term or prolonged administration of 9-[2(phosphonomethoxy)propyl] adenine (tenofovir) to newborn and infant rhesus macaques. Antimicrob Agents Chemother 2004;48:1469–87.

116. Brown TT, Qaqish RB. Antiretroviral therapy and the prevalence of osteopenia and osteoporosis: a meta-analytic review. AIDS 2006;20:2165–74.

117. Dao CN, Patel P, Overton ET, Rhame F, et al. Study to understand the natural history of HIV and AIDS in the era of effective therapy (SUN) investigators. Low vitamin D among HIV-infected adults: prevalence of and risk factors for low vitamin D levels in cohort of HIV-infected adults and comparison to prevalence among adults in the US general population. Clin Infect Dis 2011;52:396–405.

118. Jacobson DL, Spiegelman D, Know TK, Wilson IB. Evolution and predictors of change in total bone mineral density over time in HIV-infected men and women in the nutrition for healthy living study. J Acquir Immune Defic Syndr Hum Retrovirol 2008;49:298–308.

119. Kanis JA, Borgstrom F, De Laet C, et al. Assessment of fracture risk. Osteoporosis Int  2005;16:581–9

120. Pedrazzoni M, Vescovi L, Maninetti M, et al. Effects of chronic heroine abuse on bone and mineral metabolism. Acta Endocrinol 1993;129:42–5.

121. Lo Re V 3rd, Guaraldi G, Leonard MB, et al. Viral hepatitis is associated with reduced bone mineral density in HIV-infected women but not men. AIDS 1990;23:2191–8.

122. Yin MT, Modarresi R, Shane E, et al. Effects of HIV infection and antiretroviral therapy with ritonavir on induction of osteoclast-like cells in postmenopausal women. Osteoporos Int 2011;22:1459–66.

123. Sharma A, Cohen HW, Freeman R, et al. Prospective evaluation of bone mineral density among middle-aged HIV-infected and uninfected women: association between methadone use and bone loss. Maturitas 2011;70:295–301.

124. Triant VA, Brown TT, Lee H, Grinspoon SK. Fracture prevalence among human immunodeficiency virus (HIV)-infected versus non-HIV-infected patients in a large U.S. healthcare system. J Clin Endocrinol Metab 2008;93:3499–504.

125. Womack JA, Goulet JL, Gibert C, et al. Veterans Aging Cohort Study Project Team. Increased risk of fragility fractures among HIV infected compared to uninfected male veterans. PLoS One Feb 16 2011;6:e17217.

126. Young B, Dao CN, Buchacz K, et al, HIV Outpatient Study (HOPS) Investigators. Increased rates of bone fracture among HIV-infected persons in the HIV Outpatient Study (HOPS) compared with the US general population, 2000–2006. Clin Infect Dis 2011;52:1061–8.

127. U.S. Preventive Services Task Force. Screening for osteoporosis: U.S. Preventive Services Task Force recommendation statement. Ann Intern Med 2011; 154:356–64.

128. Aberg JA, Kaplan JE, Libman H, et al; HIV Medicine Association of the Infectious Diseases Society of America. Primary care guidelines for the management of persons infected with human immunodeficiency virus: 2009 update by the HIV medicine Association of the Infectious Diseases Society of America. Clin Infect Dis 2009;49:651–81.

129. National Osteoporosis Foundation. Clinician’s guide to prevention and treatment of osteoporosis 2014. Washington, DC: National Osteoporosis Foundation; 2014.

130. McComsey GA, Tebas P, Shane E, et al. Bone disease in HIV infection: a practical review and recommendations for HIV care providers. Clin Infect Dis 2010;51:937–46.

131. McComsey GA, Kendall MA, Tebas P, et al. Alendronate with calcium and vitamin D supplementation is safe and effective for the treatment of decreased bone mineral density in HIV. AIDS 2007;21:2473–82.

132. Lin D, Rieder MJ. Interventions for the treatment of decreased bone mineral density associated with HIV infection. Cochrane Database Syst Rev 2007:CD005645.

133. Haring B, Leng X, Robinson J. Cardiovascular disease and cognitive decline in postmenopausal women: results from the Women’s Health Initiative Memory Study. J Am Heart Assoc 2013;2:e000369.

134. Soares CN, Maki PM. Menopausal transition, mood, and cognition: an integrated view to close the gaps. Menopause 2010;17:812–4.

135. Greendale GA, Derby CA, Maki PM. Perimenopause and cognition. Obstet Gynecol Clin North Am 2011;38:519–35.

136. Greendale GA, Wight RG, Huang MH, et al. Menopause-associated symptoms and cognitive performance: results from the study of women’s health across the nation. Am J Epidemiol 2010;171:1214–24.

137. Price RW. Neurological complications of HIV infection. Lancet 1996;348:445–52.

138. Antinori A, Arendt G, Becker JT, et al. Updated research nosology for HIV-associated neurocognitive disorders. Neurology 2007;69:1789–99.

139. Gisslén M, Price RW, Nilsson S. The definition of HIV-associated neurocognitive disorders: are we overestimating the real prevalence? BMC Infect Dis 2011;11:356.

140. Rubin LH, Sundermann EE, Cook JA, et al. An investigation of menopausal stage and symptoms on cognition in HIV-infected women. Menopause 2014;21:997–1006.

141. Cook JA, Cohen MH, Burke J, et al. Effects of depressive symptoms and mental health quality of life on use of highly active antiretroviral therapy among HIV-seropositive women. J Acquir Immune Defic Syndr 2002;30:401–9.

142. Cook JA, Grey D, Burke J, et al. Depressive symptoms and AIDS-related mortality among a multisite cohort of HIV-positive women. Am J Pub Health 2004;94:1133–40.

143. Kim SC, Messing S, Shah K, et al. Effects of highly active antiretroviral therapy (HAART) and menopause on risk of progression of cervical dysplasia in human immune deficiency virus (HIV) infected women. Infect Dis Obstet Gynecol 2013;2013:784718.

144. Ceccaldi PF, Ferreira C, Coussy F, et al. Cervical disease in postmenopausal HIV-1 infected women. J Gynecol Obstet Biol Reprod 2010;39:466–70.

145. European Study Group on Heterosexual Transmission of HIV. Comparison of female to male and male to female transmission of HIV in 563 stable couples. BMJ 1992;304:809–13.

146. Meditz AL, Moreau KL, MaWhinney S, et al. CCR5 expression is elevated on endocervical CD4+ T cells in healthy postmenopausal women. J Acquir Immune Defic Syndr 2012;59:221–8.

147. Chappell CA, Isaacs CE, Xu W, et al. The effect of menopause on the innate antiviral activity of cervicovaginal lavage. Am J Obstet Gynecol 2015;213:204.

148. Melo KC, Melo MR, Ricci BV, Segurado AC. Correlates of human immunodeficiency virus cervicovaginal shedding among postmenopausal and fertile-aged women. Menopause 2012;19:150–6.

149. Viard JP, Mocroft A, Chiesi A, et al. Influence of age of CD4 cell recovery in human immunodeficiency virus-infected patients receiving highly active antiretroviral therapy: evidence from the Euro SIDA study. J Infect Dis 2001;193:1290–4.

150. Grabar S, Kousignian I, Sobel A, et al. Immunological and clinical responses to highly active antiretroviral therapy over 50 years of age. Results from the French Hospital Database on HIV. AIDS 2004;18:2029–38.

151. Cuzin L, Delpierre C, Gerard S, et al. Immunologic and clinical responses to highly active antiretroviral therapy in patients with HIV infection aged >50 years. Clin Infect Dis 2007;45:654–7.

152. Patterson KB, Cohn SE, Uynik J, et al. Treatment responses in antiretroviral treatment-naïve premenopausal and postmenopausal HIV-1 infected women: an analysis from AIDS clinical trials group studies. Clin Infect Dis 2009;49:473–6.

153. van Benthem BH, Vernazza P, Coutinho RA, et al. The impact of pregnancy and menopause on CD4 lymphocyte count in HIV-infected women. AIDS 2002;16:919–24.

Issue
Journal of Clinical Outcomes Management - January 2016, VOL. 23, NO. 1
Publications
Topics
Sections

From the University of Maryland School of Medicine, Baltimore, MD.

 

Abstract

  • Objective: To review the current literature on menopause in HIV-infected women.
  • Methods: We searched PubMed for articles published in English using the search terms HIV and menopause, HIV and amenorrhea, HIV and menopause symptoms, HIV and vasomotor symptoms, HIV and vaginal dryness, HIV and dyspareunia, HIV and menopause and cardiovascular disease, HIV and menopause and osteoporosis, HIV and menopause and cognition, HIV and menopause and cervical dysplasia, menopause and HIV transmission, and menopause and HIV progression. Major studies on menopause in other populations were also reviewed to provide background data.
  • Results: While studies on the age of menopause in HIV-infected women give conflicting results, immuno-suppression associated with HIV appears to contribute to an earlier onset of menopause. HIV-infected women experience menopausal symptoms, especially vasomotor symptoms, earlier and in greater intensity. In addition, menopause and HIV infection have additive effects on one another, further increasing the disease risks of cardiovascular disease, osteoporosis, and progression of cervical dysplasia. The effects of menopause on HIV infection itself seems limited. While some data suggest an increased risk of acquisition in non–HIV-infected menopausal women, menopause has no effect on the transmission or progression of HIV in menopausal HIV-infected women.
  • Conclusion: As HIV-infected individuals live longer, practitioners will encounter an increasing number of women entering menopause and living into their postmenopausal years. Future studies on the age of menopause, symptoms of menopause, and the effects of menopause on long term comorbidities such as cognitive decline, cardiovascular disease, and bone density loss are necessary to improve care of this expanding population of women living with HIV.

 

Since the introduction of highly active antiretroviral therapy (HAART) in 1996, there has been a significant decrease in morbidity and mortality worldwide among individuals living with human immunodeficiency virus (HIV) [1]. It is projected that by the year 2020, half of persons living with HIV infection in the United States will be over the age of 50 years [2]. For HIV-infected women, this longer survival translates into an increased number of women entering into menopause and living well beyond menopause. Enhancing our knowledge about menopause in HIV-infected women is important since the physiologic changes associated with menopause impact short- and long-term quality of life and mortality. Symptoms associated with menopause can be mistaken for symptoms suggestive of infections, cancers, and drug toxicity. Furthermore, changes in cognition, body composition, lipids, glucose metabolism, and bone mass are influential factors determining morbidity and mortality in later years.

 

 

 

Effect of HIV on the Menstrual Cycle

Menstrual irregularities, including amenorrhea and anovulation, are more frequently found in women of low socioeconomic class who experience more social and physical stress like poverty and physical illnesses [3]. In addition, women with low body mass index (BMI) have decreased serum estradiol levels which lead to amenorrhea [3,4]. Furthermore, several studies have demonstrated that methadone, heroin, and morphine use are associated with amenorrhea. Opiate use inhibits the central neural reproductive drive leading to amenorrhea even in the absence of menopause [5–7].

As these demographics, body habitus, and lifestyle characteristics are frequently found among HIV-infected women, it is not surprising that amenorrhea and anovulation are common in this population [8–14]. In fact, studies show that there is an increased prevalence of amenorrhea and anovulation among HIV-infected women when compared to non–HIV-infected women [8]. Some studies suggest that women with lower CD4 cell counts and higher viral loads have increased frequency of amenorrhea and irregular menstruation compared to those with higher CD4 cell counts and lower viral loads [9,10]. However, it remains unclear if HIV infection itself, instead of the associated social and medical factors, is responsible for the higher frequency of amenorrhea [11–13]. For example, in a prospective study comparing 802 HIV-infected women with 273 non–HIV-infected women, there was no difference in the prevalence of amenorrhea when controlling for BMI, substance use, and age [13].

The World Health Organization (WHO) currently defines natural menopause as the permanent cessation of menstruation for 12 consecutive months without any obvious pathological or physiologic causes [15]. However, given the increased prevalence of amenorrhea in HIV-infected women, amenorrhea seen with HIV infection can be mistaken for menopause. The Women’s Interagency HIV Study (WIHS), a multicenter, observational study of HIV-infected women and non–HIV-infected women of similar socioeconomic status, found that more than half of HIV-infected women with prolonged amenorrhea of at least 1 year had serum follicle-stimulating hormone (FSH) levels in the premenopausal range of less than 25 mIU/mL [16]. Hence, this implies that some of these women may have had prolonged amenorrhea rather than menopause [17]. The traditional definition of menopause may need to be altered in this population.

Age at Menopause

Natural menopause, retrospectively determined by the cessation of menstrual cycles for 12 consecutive months, is a reflection of complete, or near complete, ovarian follicular depletion with subsequent low estrogen levels and high FSH concentrations [18]. In the United States, studies have found the mean age of menopause to be between 50 to 52 years old [19,20].  These studies, however, focused predominantly on menopause in middle class, white women. Early menopause, defined as the permanent cessation of menstruation between 40 to 45 years of age, affects 5% of the women in the United States, while premature menopause or primary ovarian insufficiency, which occurs at younger than 40 years of age, affects 1% of the women [21].

As earlier menopause is associated with increased risks of diabetes [22], cardiovascular disease [23]stroke [24], and osteoporosis [25], identifying the mean age of menopause is important in the management of HIV-infected women. Among women in the United States, early menopause has been observed in women who are African American, nulliparous, have lower BMI, smoke tobacco, and have more stress, less education, and more unemployment [26–29]. Unhealthy lifestyles can also contribute to an earlier age of menopause. Smoking is one of the most consistent and modifiable risk factors associated with an earlier onset of natural menopause, accelerating menopause by up to 2 years [26,30]. Substances present in cigarettes are associated with irreversible damage of ovarian follicles and impaired liver estrogen metabolism [30]. Cocaine use has also been associated with lower estradiol levels, suggesting possible ovary-toxic effects [7,31].

Many of these characteristics and unhealthy lifestyles are prevalent among HIV-infected women. Prevalence of current smoking among HIV-infected persons is found to be approximately 42% [32] in comparison with the 19% seen in the general population in the United States [33]. Specifically, among women participating in WIHS, 56% of the women were found to be current smokers with an additional 16% of the women found to be prior smokers [34]. In addition, African Americans account for the highest proportion of new HIV infections in the United States with an estimated 64% of all new HIV infections in women found to be in African Americans [35]. Furthermore, HIV-infected women are of lower socioeconomic status, with increased prevalence of substance use than that typically found in women enrolled in studies on the age of menopause [36]. Hence, when examining the influence of HIV on the age of menopause, one needs to have a comparator of non–HIV-infected group with similar characteristics. Studies without comparison groups have reported the median age of menopause in HIV-infected women to be between 47 and 50 years old [37–42].

There are only few studies that have focused on the age of menopause in HIV-infected women with a similar comparative non–HIV-infected group.Cejtin et al studied the age of menopause in women enrolled in the WIHS [43]. HIV-infected women partaking in the WIHS were primarily African American and of lower socioeconomic status with heterosexual transmission rather than injection drug use as the major HIV risk factor [44]. They found no significant difference in the median age of menopause when HIV-infected women were compared to non–HIV-infected women. Median age of menopause was 47.7 years in HIV-infected women and 48.0 years in non–HIV-infected women [43].

In contrast, in the Ms Study, a prospective cohort comparing 302 HIV-infected with 259 non-HIV-infected women, HIV-infected women were 73% more likely to experience early menopause than non-HIV-infected women [45]. Similar to the WIHS, there was a high prevalence of African Americans but unlike the WIHS the majority of participants had used heroin or cocaine within the past 5 years. The high prevalence of drug use and current or former cigarette use in the Ms Study likely contributed to the relatively early onset of menopause. Furthermore, the WIHS and Ms Study used different definition of menopause. The WIHS defined menopause as 6 consecutive months of amenorrhea with an FSH level greater than 25 mIU/mL while the Ms Study defined menopause as the cessation of menstrual period for 12 consecutive months [43,45]. Given the fact that 52% of the women in the Ms Study had high-risk behaviors associated with amenorrhea and that menopause was defined as 12 months of amenorrhea without corresponding FSH levels, it is possible that the Ms Study included many women with amenorrhea who had not yet reached menopause. On the other hand, although the 6 months’ duration of amenorrhea used in the WIHS to define menopause had the potential to include women who only had amenorrhea without menopause, the use of FSH levels to define menopause most likely eliminated women who only had amenorrhea.

HIV-infected women have several factors associated with early menopause which are similar to that in the general population, including African American race, injection drug use, cigarette smoking, and menarche before age of 11 [37,41]. In addition, multiple studies have shown that a key factor associated with early age of menopause among HIV-infected women is the degree of immunosuppression [37,41,45]. The Ms Study found that women with CD4 cell counts < 200 cells/mmhad an increased risk ofamenorrhea lasting at least 12 months when compared to women with CD4 cell counts ≥ 200 cells/mm3. The median age of menopause was 42.5 years in women with CD4 cell counts < 200 cells/mm3, 46.0 years in women with CD4 cell counts between 200 cells/mmand 500 cells/mm3, and 46.5 years in women with CD4 cell counts > 500 cells/mm[45]. Similarly, in a cohort of 667 Brazilian HIV-infected women, among whom 160 women were postmenopausal, Calvet et al found 33% of women with CD4 cell counts < 50 cells/mmto have premature menopause, compared to 8% of women with CD4 cell counts ≥ 350 cells/mm[41]. De Pommerol et al  studied 404 HIV-infected women among whom 69 were found to be postmenopausal. They found that women with CD4 cell counts < 200 cells/mmwere more likely to have premature menopause compared to women with CD4 cell counts ≥ 350 cells/mm[37].

Besides the degree of immunosuppression, another factor contributing to early menopause unique to HIV-infected women is chronic hepatitis C infection [41].

 

 

 

Menopause-Associated Symptoms

The perimenopausal period, which begins on average 4 years prior to the final menstrual period, is characterized by hormonal fluctuations leading to irregular menstrual cycles. Symptoms associated with these physiologic changes during the perimenopausal period include vasomotor symptoms (hot flashes), genitourinary symptoms (vaginal dryness and dyspareunia), anxiety, depression, sleep disturbances, and joint aches [46–53]. Such menopausal symptoms can be distressing, negatively impacting quality of life [54].

It can be difficult to determine which symptoms are caused by the physiologic changes of menopause in HIV-infected women as they have multiple potential reasons for these symptoms, such as antiretroviral therapy, comorbidities, and HIV infection itself [55]. However, several studies clearly show that there are symptoms that occur more commonly in the perimenopausal period and that HIV-infected women experience these symptoms earlier and with greater intensity [38–40,42,56,57]. In a cross-sectional study of 536 women among whom 54% were HIV-infected, Miller et al found that menopausal symptoms were reported significantly more frequently in HIV-infected women compared with non–HIV-infected women [56]. As symptoms can occur in greater intensity and impair quality of life, it is important that providers be able to recognize, understand, and appropriately treat menopausal symptoms in HIV-infected women.

Vasomotor Symptoms

In the United States the most common symptom during perimenopause is hot flashes, which occur in 38% to 80% of women [58,59]. Vasomotor symptoms are most common in women who smoke, use illicit substances, have a high BMI, are of lower socioeconomic status, and are African American [19]. As expected, prior studies focusing on hot flash prevalence among premenopausal, perimenopausal, and postmenopausal HIV-infected women found that postmenopausal women experience more hot flashes than premenopausal or perimenopausal women [40,42]. In addition, a comparison of HIV-infected and non–HIV-infected women demonstrated a higher prevalence of hot flashes among HIV-infected women [38,56]. Ferreira et al found that 78% of Brazilian HIV-infected women reported vasomotor symptoms compared to 60% of non–HIV-infected women [38]. Similarly, Miller et al reported that 64% of HIV-infected women reported vasomotor symptoms compared to 58% of non–HIV-infected women [56].

Vasomotor symptoms can be severely distressing with hot flashes contributing to increased risk of depression [56,60]. In a cross-sectional analysis of 835 HIV-infected and 335 non–HIV-infected women from the WIHS, persistent vasomotor symptoms predicted elevated depressive symptoms in both HIV-infected and non-HIV-infected women [60]. In a similar cross-sectional analysis of 536 women, among whom 54% were HIV positive and 37% were perimenopausal, psychological symptoms were prevalent in 61% of the women with vasomotor symptoms [56].

Oddly enough, higher CD4 cell counts appear to be associated with increased prevalence of vasomotor symptoms [39,56]. Clark et al demonstrated that menopausal HIV-infected women with CD4 cell counts > 500 cells/mmwere more likely to report hot flashes [39]. Similarly, Miller et al observed a reduction in the prevalence of menopausal symptoms as CD4 cell counts declined among HIV-infected non-HAART users [56]. The rationale behind this is unclear but some experts postulated that it may be due to the effects of HAART.

Genitourinary Symptoms

With estrogen deficiency, which accompanies the perimenopausal period, vulvovaginal atrophy (VVA) occurs leading to symptoms of vaginal dryness, itching, burning, urgency, and dyspareunia (painful intercourse) [59,61,62]. Unlike vasomotor symptoms, which diminish with time, genitourinary symptoms generally worsen if left untreated [63]. Furthermore, these symptoms are often underreported and underdiagnosed [64,65]. Several studies using telephone and online surveys have found that the prevalence of symptoms of VVA is between 43% and 63% in postmenopausal women [66–69]. Even higher rates were found in the Agata Study in which pelvic exams in 913 Italian women were performed to obtain objective signs of VVA [62]. The prevalence of VVA was 64% 1 year after menopause and 84% 6 years after menopause. Vaginal dryness was found in 100% of participants with VVA or 82% of total study participants. In addition, 77% of women with VVA, or 40% of total study participants, reported dyspareunia.

Genitourinary symptoms are most common among women who are African American, have an increased BMI, are from lower socioeconomic class, use tobacco [19], have prior history of pelvic inflammatory disease, and have anxiety and depression [70,71]. Similarly to hot flashes, many of these predisposing factors are more common in HIV-infected women. Fantry et al found that 49.6% of HIV-infected women had vaginal dryness. Although 56% of postmenopausal women and 36% of perimenopausal women complained of vaginal dryness, in a multivariate analysis only cocaine use, which can decrease estradiol levels [7,31] was associated with a higher frequency of vaginal dryness [40].

Similarly, dyspareunia is also common among HIV-infected women. In a cross-sectional study of 178 non–HIV-infected and 128 HIV-infected women between 40 and 60 years of age, Valadares et al found that the frequency of dyspareunia in HIV-infected women was high at 41.8% [72]. However, this was not significantly higher compared to the prevalence of 34.8% in non–HIV-infected women. HIV infection itself was not associated with the presence of dyspareunia

Psychiatric Symptoms

Anxiety and depression are also common symptoms in perimenopausal women [73–76]. Studies have shown that depression is diagnosed 2.5 times more frequently among perimenopausal than premenopausal women [76].

In a study by Miller et al that focused on 536 HIV-infected women, among whom 37% were perimenopausal, 89% reported psychological symptoms [56]. Ferreira et al found that HIV-infected perimenopausal women had an increased incidence of psychological symptoms compared to non–HIV-infected women [38]. Whether this increased prevalence of psychological symptoms seen in HIV-infected women can be attributed to menopause is unclear since one third to one half of men and women living with HIV experience symptoms of depression [77]. However, in the WIHS, which compared 835 HIV-infected with 335 non-HIV-infected women from all menopausal stages, elevated depressive symptoms were seen in the early perimenopausal period [60]. There was no increased incidence of such symptoms during the premenopausal or postmenopausal period, suggesting the contribution of menopause to depressive symptoms during the perimenopausal period [60].

Persistent menopausal symptoms, especially hot flashes, also predicted elevated depressive symptoms in several studies [56,60] suggesting the importance of appropriately identifying and treating menopausal symptoms. In addition, cognitive decline associated with menopause contributes to depression [78–80].

Other Symptoms

Sleep disturbances are also common among perimenopausal women, with prevalence estimated to be between 38% and 46% [81–84]. Hot flashes, anxiety, and depression appear to be contributing factors [81–84]. In a cross-sectional study of 273 HIV-infected and 264 non-HIV-infected women between 40 and 60 years of age, insomnia was found in 51% of perimenopausal and 53% of postmenopausal HIV-infected women. HIV-infected women had the same prevalence of insomnia compared to non–HIV-infected women [85]. Joint aches are also commonly reported in the perimenopausal period, with prevalence as high as 50% to 60% among perimenopausal women in the United States [52,53]. In HIV-infected women, Miller et al found that 63% of menopausal women reported arthralgia [56].

Treatment

For women experiencing severe hot flashes and vaginal dryness, short-term menopausal hormone therapy (MHT) is indicated to relieve symptoms. MHT should be limited to the shortest period of time at the lowest effective dose as MHT is associated with increased risks of breast cancer, cardiovascular disease, thromboembolism, and increased morbidity [86]. Despite the increased severity of menopausal symptoms experienced among HIV-infected women, the prevalence of the use of MHT in this population is lower compared to non–HIV-infected women [85].

Topical treatment is recommended for women who are experiencing solely vaginal atrophy. First-line treatment is topical nonhormonal therapy such as moisturizers and lubricants [87]. If symptoms are not relieved, then topical vaginal estrogen therapy is recommended [87]. Although topical therapy can result in estrogen absorption into the circulation, it is to a much lesser extent than systemic estrogen therapy [88].

Overall, there is lack of data on the potential interactions between MHT and HAART. Much of the potential interactions are inferred from pharmacokinetic and pharmacodynamics studies between HAART and oral contraceptives. Hormone therapy, protease inhibitors (PIs), colbicistat, and non-nucleoside reverse transcriptase inhibitors (NNRTIs) are all metabolized by the CYP3A4 enzyme [89–91]. Current evidence suggests that concomitant use of hormone therapy with NNRTIs and PIs does not significantly alter the pharmacokinetics of HAART or the clinical outcomes of HIV [91]. However, there is evidence that concomitant use of nevirapine and PIs boosted with ritonavir leads to decrease in estrogen levels so higher doses of MHT may have to be used to achieve symptomatic relief [91]. There is no data on the interaction between PIs boosted with colbicistat and estrogen [92]. Integrase inhibitors, nucleoside and nucleotide reverse transcriptase inhibitors (NRTIs), and the CCR5 antagonist maraviroc have no significant interactions with estrogen containing compounds [89,90,92].

Cardiovascular Risk

Estrogen deficiency resulting from menopause leads to several long-term effects, including cardiovascular disease and osteoporosis. The loss of protective effects of estrogen leads to an increased risk of cardiovascular disease particularly with changes in lipid profiles [93]. Perimenopausal women experience changes in body composition with increased fat mass and waist circumference, as well as dyslipidemia and insulin resistance, all of which are associated with higher risk of cardiovascular disease [94].

HIV infection also incurs a higher risk of cardiovascular disease [95–99]. The inflammatory effects of HIV, HAART, and traditional risk factors including dyslipidemia all contribute to cardiovascular disease but the degree to which each factor contributes to elevated risk is unknown [95,98]. In addition, modifiable risk factors for cardiovascular disease such as decreased fitness and smoking are more commonly seen in HIV-infected women [100]. Even prior to menopause, HIV-infected women experience lipodystrophy syndrome with increase in truncal visceral adiposity and decrease in subcutaneous fat and muscle mass [101,102]. Whether such changes in body composition are exacerbated during the perimenopausal period remain unclear. In the SWEET study, which focused on 702 South African women among whom 21% were HIV-infected, there was lower lean mass but minimal difference in the fat mass of postmenopausal women compared to premenopausal women [103]. As the study was based in South Africa with only 21% HIV-infected, the results of this study should be viewed with caution. While changes in body composition were not observed in postmenopausal women in the SWEET study, increased truncal adiposity seen in premenopausal HIV-infected women is likely to pose an additional risk for cardiovascular disease during the menopause transition.

Several studies have been conducted to demonstrate an increased risk of cardiovascular disease, especially among young HIV-infected men [95–99]. However, no study has focused specifically on the risk of cardiovascular disease in postmenopausal HIV-infected women to date. Despite the lack of studies, it is plausible that the increased risk of cardiovascular disease seen in HIV infection is likely to be compounded with the increased risk seen during menopause. Postmenopausal HIV-infected women may be at significantly higher risk of cardiovascular disease. Appropriate measures such as lipid control, antiplatelet therapy, smoking cessation, and other lifestyle changes should be initiated as in any other population. Further studies are necessary focusing on the effects of menopause on cardiovascular disease risk in HIV-infected women.

 

 

 

Osteoporosis

Menopause, with its associated estrogen deficiency, is the most important risk factor associated with increased bone turnover and bone loss and can worsen HIV associated bone loss [104]. Among HIV-infected individuals, low bone mineral density (BMD) has been described even among premenopausal women and younger men [105–107]. Evidence suggests that the decreased BMD associated with HIV stabilizes or even improves after initiation of HAART in the younger population [105–107]. However, once HIV-infected women enter menopause, they have higher rates of bone loss compared to non–HIV-infected women with significantly increased prevalence of osteoporosis compared to non–HIV-infected women [108–112].

Chronic inflammation by HIV stimulates osteoclast differentiation and resorption [113]. In addition, HAART [114–116], vitamin D deficiency [117], low BMI, poor nutrition [118], inactivity, use of tobacco, alcohol, and illicit drugs [119,120], and coinfection with hepatitis B and C [121] all appear to contribute to decreased BMD among HIV-infected men and women [118]. Among HIV-infected postmenopausal women, those taking ritonavir were found to have increased differentiation of osteoclast cells and increased bone loss [122]. Similarly, methadone use in postmenopausal women has been associated with increased BMD decline [123]. African-American, HIV-infected postmenopausal women appear to be at the greatest risk for bone loss [109].

Multiple studies focusing on HIV-infected men have demonstrated an increased prevalence of fractures compared to non–HIV-infected men [124–126]. However, current studies on postmenopausal HIV-infected women demonstrate that fracture incidence is similar between HIV-infected and non–HIV-infected postmenopausal women [108,112]. Nevertheless, given the evidence of low BMD and increased fracture risk seen during menopause among non–HIV-infected women compounded with the additional bone loss seen in HIV-infected individuals, enhanced screening in postmenopausal HIV-infected women is prudent. Although the U.S. Preventive Services Task Force (USPSTF) makes no mention of HIV as a risk factor for enhanced screening [127] and the Infectious Diseases Society of America (IDSA) only recommends screening beginning at the age of 50 years old if there are additional risk factors other than HIV [128], the more recently published Primary care guidelines for the management of persons infected with HIV recommends screening postmenopausal women ≥ 50 years of age with dual-energy X-ray absorptiometry (DEXA) scan [86]. Preventative therapy such as smoking cessation, adequate nutrition, alcohol reduction, weight bearing exercises, and adequate daily vitamin D and calcium should be discussed and recommended in all menopausal HIV-infected women [129]. If the DEXA scan shows osteoporosis, bisphosphonates or other medical therapy should be considered. Although the data are limited, bisphosphonates have been shown to be effective in improving BMD [130–132].

Cognition

The menopause transition is characterized by cognitive changes such as memory loss and difficulty concentrating [133–136]. Both HIV-infected men and women are at higher risk of cognitive impairment [137–139]. Cognitive impairment can range from minor cognitive-motor disorder to HIV-associated dementia due to the immunologic, hormonal, and inflammatory effects of HIV on cognition [137–139]. In addition, those with HIV infection appear to have increased risk factors for cognitive impairment including low education level, psychiatric illnesses, increased social stress, and chemical dependence [137].

Studies focusing on the effects of both HIV infection and menopause on cognition have been limited thus far. In a cross-sectional study of 708 HIV-infected and 278 non–HIV-infected premenopausal, perimenopausal, and postmenopausal women, Rubin et al demonstrated that HIV infection, but not menopausal stage, was associated with worse performance on cognitive measures [140]. While menopausal stage was not associated with cognitive decline, menopausal symptoms like depression, anxiety, and vasomotor symptoms were associated with lower cognitive performance [140].

Though limited, current data appear to indicate that HIV infection, not menopause, contributes to cognitive dysfunction [140]. Symptoms of menopause, however, do appear to exacerbate cognitive decline indicating the importance of recognition and treatment of menopausal symptoms. This is especially important in HIV-infected women since decrease in cognition and depression can interfere with day to day function including medication adherence [141,142].

Cervical Dysplasia

As more HIV-infected women reach older age, the effects of prolonged survival and especially menopause on squamous intraepithelial lesions (SILs) are being investigated to determine if general guidelines of cervical cancer screening should be applied to postmenopausal women.

In a retrospective analysis of Papanicolaou smear results of 245 HIV-infected women, Kim et al noted that menopausal women had a 70% higher risk of progression of SILs than premenopausal women [143]. Similar results were found in a smaller retrospective study of 18 postmenopausal HIV-infected women in which postmenopausal women had a higher prevalence of SILs and persistence of low-grade SILs [144].

Although studies on progression to cervical cancer in postmenopausal HIV-infected women remain limited, current data suggest that postmenopausal HIV-infected women should continue to be monitored and screened similarly to the screening recommendations for premenopausal women. Nevertheless, further studies examining the natural course of cervical lesions are needed to establish the best practice guidelines for screening postmenopausal women.

HIV Acquisition and Transmission

The incidence of new HIV infections in older American women has increased. HIV acquisition from heterosexual contact appears to be higher in older women compared to younger women, with a study suggesting that women over age 45 years had almost a fourfold higher risk of HIV acquisition compared to those under the age of 45 years [145]. While the lack of awareness of HIV risk and less frequent use of protection may contribute to increases in new HIV infection in older women, hormonal changes associated with older age, specifically menopause, may be playing a role. Vaginal wall thinning that occurs during menopause may serve as a risk factor for HIV acquisition.

In a study by Meditz et al, the percentage of endocervical or blood CD4 T cells did not differ between premenopausal and postmenopausal women, but postmenopausal women had greater percentage of CCR5 expression. As CCR5 serves as an entry point of HIV into target cells, this suggests the possibility that postmenopausal women may be at increased risk for HIV acquisition [146]. More recently, Chappell et al also revealed that anti-HIV-1 activity was significantly decreased in postmenopausal compared to premenopausal women, suggesting that there may be an increased susceptibility to HIV-1 infection in postmenopausal women [147]. Hence there appears to be menopause-related immunologic changes of the cervix that may contribute to an increased risk of HIV acquisition in postmenopausal women.

In contrast, although data is limited, postmenopausal HIV-infected women do not appear to be at increased risk of transmitting HIV to non–HIV-infected individuals. Melo et al compared the intensity of HIV shedding between premenopausal and postmenopausal women and found that HIV shedding did not differ between premenopausal or postmenopausal women [148].

HIV Progression

Several studies have focused on the effects of HIV infection on menopause, but minimal data are available on the effects of menopause on the progression of HIV infection. With prior data suggesting that younger persons experience better immunological and virological responses to HAART [149–151], it has previously been hypothesized that virologic and immunologic responses to HAART can decline once HIV-infected women reach menopause. However, current evidence suggests that treatment responses to HAART, determined by the median changes in CD4 cell counts and percentages and viral load, in HAART-naive patients did not differ between premenopausal and postmenopausal women [152]. In addition, there appears to be no significant changes in CD4 cell counts as HIV-infected women progress through menopause [153]. These studies suggest that menopause does not affect the progression of HIV and that HAART-naive women should respond to HAART regardless of their menopausal status.

Conclusion

As HIV-infected individuals live longer, increasing number of women will enter into menopause and live many years beyond menopause. HIV-infected women experience earlier and more severe menopausal symptoms, but knowledge is still lacking on the appropriate management of these symptoms. In addition, current evidence suggests that immunosuppression associated with HIV contributes to an early onset of menopause which leads to increased risks of cardiovascular disease, osteoporosis, and progression of cervical dysplasia. These conditions require proper surveillance and can be prevented with improved understanding of influences of menopause on HIV-infected women. Furthermore, although there is some evidence suggesting that menopause has no effect on HIV transmission and progression, further studies on the immunologic and virologic effects of menopause are necessary.

There still remain significant gaps in our understanding of menopause in HIV-infected women.  As practitioners encounter an increasing number of perimenopausal and postmenopausal HIV-infected women, future studies on the effects of HIV on co-morbidities and symptoms of menopause and their appropriate management are necessary to improve care of women living with HIV.

 

Corresponding author: Lori E. Fantry, MD, MPH, 29 S. Greene St., Suite 300, Baltimore, MD 21201, lfantry@medicine.umaryland.edu.

Financial disclosures: None.

From the University of Maryland School of Medicine, Baltimore, MD.

 

Abstract

  • Objective: To review the current literature on menopause in HIV-infected women.
  • Methods: We searched PubMed for articles published in English using the search terms HIV and menopause, HIV and amenorrhea, HIV and menopause symptoms, HIV and vasomotor symptoms, HIV and vaginal dryness, HIV and dyspareunia, HIV and menopause and cardiovascular disease, HIV and menopause and osteoporosis, HIV and menopause and cognition, HIV and menopause and cervical dysplasia, menopause and HIV transmission, and menopause and HIV progression. Major studies on menopause in other populations were also reviewed to provide background data.
  • Results: While studies on the age of menopause in HIV-infected women give conflicting results, immuno-suppression associated with HIV appears to contribute to an earlier onset of menopause. HIV-infected women experience menopausal symptoms, especially vasomotor symptoms, earlier and in greater intensity. In addition, menopause and HIV infection have additive effects on one another, further increasing the disease risks of cardiovascular disease, osteoporosis, and progression of cervical dysplasia. The effects of menopause on HIV infection itself seems limited. While some data suggest an increased risk of acquisition in non–HIV-infected menopausal women, menopause has no effect on the transmission or progression of HIV in menopausal HIV-infected women.
  • Conclusion: As HIV-infected individuals live longer, practitioners will encounter an increasing number of women entering menopause and living into their postmenopausal years. Future studies on the age of menopause, symptoms of menopause, and the effects of menopause on long term comorbidities such as cognitive decline, cardiovascular disease, and bone density loss are necessary to improve care of this expanding population of women living with HIV.

 

Since the introduction of highly active antiretroviral therapy (HAART) in 1996, there has been a significant decrease in morbidity and mortality worldwide among individuals living with human immunodeficiency virus (HIV) [1]. It is projected that by the year 2020, half of persons living with HIV infection in the United States will be over the age of 50 years [2]. For HIV-infected women, this longer survival translates into an increased number of women entering into menopause and living well beyond menopause. Enhancing our knowledge about menopause in HIV-infected women is important since the physiologic changes associated with menopause impact short- and long-term quality of life and mortality. Symptoms associated with menopause can be mistaken for symptoms suggestive of infections, cancers, and drug toxicity. Furthermore, changes in cognition, body composition, lipids, glucose metabolism, and bone mass are influential factors determining morbidity and mortality in later years.

 

 

 

Effect of HIV on the Menstrual Cycle

Menstrual irregularities, including amenorrhea and anovulation, are more frequently found in women of low socioeconomic class who experience more social and physical stress like poverty and physical illnesses [3]. In addition, women with low body mass index (BMI) have decreased serum estradiol levels which lead to amenorrhea [3,4]. Furthermore, several studies have demonstrated that methadone, heroin, and morphine use are associated with amenorrhea. Opiate use inhibits the central neural reproductive drive leading to amenorrhea even in the absence of menopause [5–7].

As these demographics, body habitus, and lifestyle characteristics are frequently found among HIV-infected women, it is not surprising that amenorrhea and anovulation are common in this population [8–14]. In fact, studies show that there is an increased prevalence of amenorrhea and anovulation among HIV-infected women when compared to non–HIV-infected women [8]. Some studies suggest that women with lower CD4 cell counts and higher viral loads have increased frequency of amenorrhea and irregular menstruation compared to those with higher CD4 cell counts and lower viral loads [9,10]. However, it remains unclear if HIV infection itself, instead of the associated social and medical factors, is responsible for the higher frequency of amenorrhea [11–13]. For example, in a prospective study comparing 802 HIV-infected women with 273 non–HIV-infected women, there was no difference in the prevalence of amenorrhea when controlling for BMI, substance use, and age [13].

The World Health Organization (WHO) currently defines natural menopause as the permanent cessation of menstruation for 12 consecutive months without any obvious pathological or physiologic causes [15]. However, given the increased prevalence of amenorrhea in HIV-infected women, amenorrhea seen with HIV infection can be mistaken for menopause. The Women’s Interagency HIV Study (WIHS), a multicenter, observational study of HIV-infected women and non–HIV-infected women of similar socioeconomic status, found that more than half of HIV-infected women with prolonged amenorrhea of at least 1 year had serum follicle-stimulating hormone (FSH) levels in the premenopausal range of less than 25 mIU/mL [16]. Hence, this implies that some of these women may have had prolonged amenorrhea rather than menopause [17]. The traditional definition of menopause may need to be altered in this population.

Age at Menopause

Natural menopause, retrospectively determined by the cessation of menstrual cycles for 12 consecutive months, is a reflection of complete, or near complete, ovarian follicular depletion with subsequent low estrogen levels and high FSH concentrations [18]. In the United States, studies have found the mean age of menopause to be between 50 to 52 years old [19,20].  These studies, however, focused predominantly on menopause in middle class, white women. Early menopause, defined as the permanent cessation of menstruation between 40 to 45 years of age, affects 5% of the women in the United States, while premature menopause or primary ovarian insufficiency, which occurs at younger than 40 years of age, affects 1% of the women [21].

As earlier menopause is associated with increased risks of diabetes [22], cardiovascular disease [23]stroke [24], and osteoporosis [25], identifying the mean age of menopause is important in the management of HIV-infected women. Among women in the United States, early menopause has been observed in women who are African American, nulliparous, have lower BMI, smoke tobacco, and have more stress, less education, and more unemployment [26–29]. Unhealthy lifestyles can also contribute to an earlier age of menopause. Smoking is one of the most consistent and modifiable risk factors associated with an earlier onset of natural menopause, accelerating menopause by up to 2 years [26,30]. Substances present in cigarettes are associated with irreversible damage of ovarian follicles and impaired liver estrogen metabolism [30]. Cocaine use has also been associated with lower estradiol levels, suggesting possible ovary-toxic effects [7,31].

Many of these characteristics and unhealthy lifestyles are prevalent among HIV-infected women. Prevalence of current smoking among HIV-infected persons is found to be approximately 42% [32] in comparison with the 19% seen in the general population in the United States [33]. Specifically, among women participating in WIHS, 56% of the women were found to be current smokers with an additional 16% of the women found to be prior smokers [34]. In addition, African Americans account for the highest proportion of new HIV infections in the United States with an estimated 64% of all new HIV infections in women found to be in African Americans [35]. Furthermore, HIV-infected women are of lower socioeconomic status, with increased prevalence of substance use than that typically found in women enrolled in studies on the age of menopause [36]. Hence, when examining the influence of HIV on the age of menopause, one needs to have a comparator of non–HIV-infected group with similar characteristics. Studies without comparison groups have reported the median age of menopause in HIV-infected women to be between 47 and 50 years old [37–42].

There are only few studies that have focused on the age of menopause in HIV-infected women with a similar comparative non–HIV-infected group.Cejtin et al studied the age of menopause in women enrolled in the WIHS [43]. HIV-infected women partaking in the WIHS were primarily African American and of lower socioeconomic status with heterosexual transmission rather than injection drug use as the major HIV risk factor [44]. They found no significant difference in the median age of menopause when HIV-infected women were compared to non–HIV-infected women. Median age of menopause was 47.7 years in HIV-infected women and 48.0 years in non–HIV-infected women [43].

In contrast, in the Ms Study, a prospective cohort comparing 302 HIV-infected with 259 non-HIV-infected women, HIV-infected women were 73% more likely to experience early menopause than non-HIV-infected women [45]. Similar to the WIHS, there was a high prevalence of African Americans but unlike the WIHS the majority of participants had used heroin or cocaine within the past 5 years. The high prevalence of drug use and current or former cigarette use in the Ms Study likely contributed to the relatively early onset of menopause. Furthermore, the WIHS and Ms Study used different definition of menopause. The WIHS defined menopause as 6 consecutive months of amenorrhea with an FSH level greater than 25 mIU/mL while the Ms Study defined menopause as the cessation of menstrual period for 12 consecutive months [43,45]. Given the fact that 52% of the women in the Ms Study had high-risk behaviors associated with amenorrhea and that menopause was defined as 12 months of amenorrhea without corresponding FSH levels, it is possible that the Ms Study included many women with amenorrhea who had not yet reached menopause. On the other hand, although the 6 months’ duration of amenorrhea used in the WIHS to define menopause had the potential to include women who only had amenorrhea without menopause, the use of FSH levels to define menopause most likely eliminated women who only had amenorrhea.

HIV-infected women have several factors associated with early menopause which are similar to that in the general population, including African American race, injection drug use, cigarette smoking, and menarche before age of 11 [37,41]. In addition, multiple studies have shown that a key factor associated with early age of menopause among HIV-infected women is the degree of immunosuppression [37,41,45]. The Ms Study found that women with CD4 cell counts < 200 cells/mmhad an increased risk ofamenorrhea lasting at least 12 months when compared to women with CD4 cell counts ≥ 200 cells/mm3. The median age of menopause was 42.5 years in women with CD4 cell counts < 200 cells/mm3, 46.0 years in women with CD4 cell counts between 200 cells/mmand 500 cells/mm3, and 46.5 years in women with CD4 cell counts > 500 cells/mm[45]. Similarly, in a cohort of 667 Brazilian HIV-infected women, among whom 160 women were postmenopausal, Calvet et al found 33% of women with CD4 cell counts < 50 cells/mmto have premature menopause, compared to 8% of women with CD4 cell counts ≥ 350 cells/mm[41]. De Pommerol et al  studied 404 HIV-infected women among whom 69 were found to be postmenopausal. They found that women with CD4 cell counts < 200 cells/mmwere more likely to have premature menopause compared to women with CD4 cell counts ≥ 350 cells/mm[37].

Besides the degree of immunosuppression, another factor contributing to early menopause unique to HIV-infected women is chronic hepatitis C infection [41].

 

 

 

Menopause-Associated Symptoms

The perimenopausal period, which begins on average 4 years prior to the final menstrual period, is characterized by hormonal fluctuations leading to irregular menstrual cycles. Symptoms associated with these physiologic changes during the perimenopausal period include vasomotor symptoms (hot flashes), genitourinary symptoms (vaginal dryness and dyspareunia), anxiety, depression, sleep disturbances, and joint aches [46–53]. Such menopausal symptoms can be distressing, negatively impacting quality of life [54].

It can be difficult to determine which symptoms are caused by the physiologic changes of menopause in HIV-infected women as they have multiple potential reasons for these symptoms, such as antiretroviral therapy, comorbidities, and HIV infection itself [55]. However, several studies clearly show that there are symptoms that occur more commonly in the perimenopausal period and that HIV-infected women experience these symptoms earlier and with greater intensity [38–40,42,56,57]. In a cross-sectional study of 536 women among whom 54% were HIV-infected, Miller et al found that menopausal symptoms were reported significantly more frequently in HIV-infected women compared with non–HIV-infected women [56]. As symptoms can occur in greater intensity and impair quality of life, it is important that providers be able to recognize, understand, and appropriately treat menopausal symptoms in HIV-infected women.

Vasomotor Symptoms

In the United States the most common symptom during perimenopause is hot flashes, which occur in 38% to 80% of women [58,59]. Vasomotor symptoms are most common in women who smoke, use illicit substances, have a high BMI, are of lower socioeconomic status, and are African American [19]. As expected, prior studies focusing on hot flash prevalence among premenopausal, perimenopausal, and postmenopausal HIV-infected women found that postmenopausal women experience more hot flashes than premenopausal or perimenopausal women [40,42]. In addition, a comparison of HIV-infected and non–HIV-infected women demonstrated a higher prevalence of hot flashes among HIV-infected women [38,56]. Ferreira et al found that 78% of Brazilian HIV-infected women reported vasomotor symptoms compared to 60% of non–HIV-infected women [38]. Similarly, Miller et al reported that 64% of HIV-infected women reported vasomotor symptoms compared to 58% of non–HIV-infected women [56].

Vasomotor symptoms can be severely distressing with hot flashes contributing to increased risk of depression [56,60]. In a cross-sectional analysis of 835 HIV-infected and 335 non–HIV-infected women from the WIHS, persistent vasomotor symptoms predicted elevated depressive symptoms in both HIV-infected and non-HIV-infected women [60]. In a similar cross-sectional analysis of 536 women, among whom 54% were HIV positive and 37% were perimenopausal, psychological symptoms were prevalent in 61% of the women with vasomotor symptoms [56].

Oddly enough, higher CD4 cell counts appear to be associated with increased prevalence of vasomotor symptoms [39,56]. Clark et al demonstrated that menopausal HIV-infected women with CD4 cell counts > 500 cells/mmwere more likely to report hot flashes [39]. Similarly, Miller et al observed a reduction in the prevalence of menopausal symptoms as CD4 cell counts declined among HIV-infected non-HAART users [56]. The rationale behind this is unclear but some experts postulated that it may be due to the effects of HAART.

Genitourinary Symptoms

With estrogen deficiency, which accompanies the perimenopausal period, vulvovaginal atrophy (VVA) occurs leading to symptoms of vaginal dryness, itching, burning, urgency, and dyspareunia (painful intercourse) [59,61,62]. Unlike vasomotor symptoms, which diminish with time, genitourinary symptoms generally worsen if left untreated [63]. Furthermore, these symptoms are often underreported and underdiagnosed [64,65]. Several studies using telephone and online surveys have found that the prevalence of symptoms of VVA is between 43% and 63% in postmenopausal women [66–69]. Even higher rates were found in the Agata Study in which pelvic exams in 913 Italian women were performed to obtain objective signs of VVA [62]. The prevalence of VVA was 64% 1 year after menopause and 84% 6 years after menopause. Vaginal dryness was found in 100% of participants with VVA or 82% of total study participants. In addition, 77% of women with VVA, or 40% of total study participants, reported dyspareunia.

Genitourinary symptoms are most common among women who are African American, have an increased BMI, are from lower socioeconomic class, use tobacco [19], have prior history of pelvic inflammatory disease, and have anxiety and depression [70,71]. Similarly to hot flashes, many of these predisposing factors are more common in HIV-infected women. Fantry et al found that 49.6% of HIV-infected women had vaginal dryness. Although 56% of postmenopausal women and 36% of perimenopausal women complained of vaginal dryness, in a multivariate analysis only cocaine use, which can decrease estradiol levels [7,31] was associated with a higher frequency of vaginal dryness [40].

Similarly, dyspareunia is also common among HIV-infected women. In a cross-sectional study of 178 non–HIV-infected and 128 HIV-infected women between 40 and 60 years of age, Valadares et al found that the frequency of dyspareunia in HIV-infected women was high at 41.8% [72]. However, this was not significantly higher compared to the prevalence of 34.8% in non–HIV-infected women. HIV infection itself was not associated with the presence of dyspareunia

Psychiatric Symptoms

Anxiety and depression are also common symptoms in perimenopausal women [73–76]. Studies have shown that depression is diagnosed 2.5 times more frequently among perimenopausal than premenopausal women [76].

In a study by Miller et al that focused on 536 HIV-infected women, among whom 37% were perimenopausal, 89% reported psychological symptoms [56]. Ferreira et al found that HIV-infected perimenopausal women had an increased incidence of psychological symptoms compared to non–HIV-infected women [38]. Whether this increased prevalence of psychological symptoms seen in HIV-infected women can be attributed to menopause is unclear since one third to one half of men and women living with HIV experience symptoms of depression [77]. However, in the WIHS, which compared 835 HIV-infected with 335 non-HIV-infected women from all menopausal stages, elevated depressive symptoms were seen in the early perimenopausal period [60]. There was no increased incidence of such symptoms during the premenopausal or postmenopausal period, suggesting the contribution of menopause to depressive symptoms during the perimenopausal period [60].

Persistent menopausal symptoms, especially hot flashes, also predicted elevated depressive symptoms in several studies [56,60] suggesting the importance of appropriately identifying and treating menopausal symptoms. In addition, cognitive decline associated with menopause contributes to depression [78–80].

Other Symptoms

Sleep disturbances are also common among perimenopausal women, with prevalence estimated to be between 38% and 46% [81–84]. Hot flashes, anxiety, and depression appear to be contributing factors [81–84]. In a cross-sectional study of 273 HIV-infected and 264 non-HIV-infected women between 40 and 60 years of age, insomnia was found in 51% of perimenopausal and 53% of postmenopausal HIV-infected women. HIV-infected women had the same prevalence of insomnia compared to non–HIV-infected women [85]. Joint aches are also commonly reported in the perimenopausal period, with prevalence as high as 50% to 60% among perimenopausal women in the United States [52,53]. In HIV-infected women, Miller et al found that 63% of menopausal women reported arthralgia [56].

Treatment

For women experiencing severe hot flashes and vaginal dryness, short-term menopausal hormone therapy (MHT) is indicated to relieve symptoms. MHT should be limited to the shortest period of time at the lowest effective dose as MHT is associated with increased risks of breast cancer, cardiovascular disease, thromboembolism, and increased morbidity [86]. Despite the increased severity of menopausal symptoms experienced among HIV-infected women, the prevalence of the use of MHT in this population is lower compared to non–HIV-infected women [85].

Topical treatment is recommended for women who are experiencing solely vaginal atrophy. First-line treatment is topical nonhormonal therapy such as moisturizers and lubricants [87]. If symptoms are not relieved, then topical vaginal estrogen therapy is recommended [87]. Although topical therapy can result in estrogen absorption into the circulation, it is to a much lesser extent than systemic estrogen therapy [88].

Overall, there is lack of data on the potential interactions between MHT and HAART. Much of the potential interactions are inferred from pharmacokinetic and pharmacodynamics studies between HAART and oral contraceptives. Hormone therapy, protease inhibitors (PIs), colbicistat, and non-nucleoside reverse transcriptase inhibitors (NNRTIs) are all metabolized by the CYP3A4 enzyme [89–91]. Current evidence suggests that concomitant use of hormone therapy with NNRTIs and PIs does not significantly alter the pharmacokinetics of HAART or the clinical outcomes of HIV [91]. However, there is evidence that concomitant use of nevirapine and PIs boosted with ritonavir leads to decrease in estrogen levels so higher doses of MHT may have to be used to achieve symptomatic relief [91]. There is no data on the interaction between PIs boosted with colbicistat and estrogen [92]. Integrase inhibitors, nucleoside and nucleotide reverse transcriptase inhibitors (NRTIs), and the CCR5 antagonist maraviroc have no significant interactions with estrogen containing compounds [89,90,92].

Cardiovascular Risk

Estrogen deficiency resulting from menopause leads to several long-term effects, including cardiovascular disease and osteoporosis. The loss of protective effects of estrogen leads to an increased risk of cardiovascular disease particularly with changes in lipid profiles [93]. Perimenopausal women experience changes in body composition with increased fat mass and waist circumference, as well as dyslipidemia and insulin resistance, all of which are associated with higher risk of cardiovascular disease [94].

HIV infection also incurs a higher risk of cardiovascular disease [95–99]. The inflammatory effects of HIV, HAART, and traditional risk factors including dyslipidemia all contribute to cardiovascular disease but the degree to which each factor contributes to elevated risk is unknown [95,98]. In addition, modifiable risk factors for cardiovascular disease such as decreased fitness and smoking are more commonly seen in HIV-infected women [100]. Even prior to menopause, HIV-infected women experience lipodystrophy syndrome with increase in truncal visceral adiposity and decrease in subcutaneous fat and muscle mass [101,102]. Whether such changes in body composition are exacerbated during the perimenopausal period remain unclear. In the SWEET study, which focused on 702 South African women among whom 21% were HIV-infected, there was lower lean mass but minimal difference in the fat mass of postmenopausal women compared to premenopausal women [103]. As the study was based in South Africa with only 21% HIV-infected, the results of this study should be viewed with caution. While changes in body composition were not observed in postmenopausal women in the SWEET study, increased truncal adiposity seen in premenopausal HIV-infected women is likely to pose an additional risk for cardiovascular disease during the menopause transition.

Several studies have been conducted to demonstrate an increased risk of cardiovascular disease, especially among young HIV-infected men [95–99]. However, no study has focused specifically on the risk of cardiovascular disease in postmenopausal HIV-infected women to date. Despite the lack of studies, it is plausible that the increased risk of cardiovascular disease seen in HIV infection is likely to be compounded with the increased risk seen during menopause. Postmenopausal HIV-infected women may be at significantly higher risk of cardiovascular disease. Appropriate measures such as lipid control, antiplatelet therapy, smoking cessation, and other lifestyle changes should be initiated as in any other population. Further studies are necessary focusing on the effects of menopause on cardiovascular disease risk in HIV-infected women.

 

 

 

Osteoporosis

Menopause, with its associated estrogen deficiency, is the most important risk factor associated with increased bone turnover and bone loss and can worsen HIV associated bone loss [104]. Among HIV-infected individuals, low bone mineral density (BMD) has been described even among premenopausal women and younger men [105–107]. Evidence suggests that the decreased BMD associated with HIV stabilizes or even improves after initiation of HAART in the younger population [105–107]. However, once HIV-infected women enter menopause, they have higher rates of bone loss compared to non–HIV-infected women with significantly increased prevalence of osteoporosis compared to non–HIV-infected women [108–112].

Chronic inflammation by HIV stimulates osteoclast differentiation and resorption [113]. In addition, HAART [114–116], vitamin D deficiency [117], low BMI, poor nutrition [118], inactivity, use of tobacco, alcohol, and illicit drugs [119,120], and coinfection with hepatitis B and C [121] all appear to contribute to decreased BMD among HIV-infected men and women [118]. Among HIV-infected postmenopausal women, those taking ritonavir were found to have increased differentiation of osteoclast cells and increased bone loss [122]. Similarly, methadone use in postmenopausal women has been associated with increased BMD decline [123]. African-American, HIV-infected postmenopausal women appear to be at the greatest risk for bone loss [109].

Multiple studies focusing on HIV-infected men have demonstrated an increased prevalence of fractures compared to non–HIV-infected men [124–126]. However, current studies on postmenopausal HIV-infected women demonstrate that fracture incidence is similar between HIV-infected and non–HIV-infected postmenopausal women [108,112]. Nevertheless, given the evidence of low BMD and increased fracture risk seen during menopause among non–HIV-infected women compounded with the additional bone loss seen in HIV-infected individuals, enhanced screening in postmenopausal HIV-infected women is prudent. Although the U.S. Preventive Services Task Force (USPSTF) makes no mention of HIV as a risk factor for enhanced screening [127] and the Infectious Diseases Society of America (IDSA) only recommends screening beginning at the age of 50 years old if there are additional risk factors other than HIV [128], the more recently published Primary care guidelines for the management of persons infected with HIV recommends screening postmenopausal women ≥ 50 years of age with dual-energy X-ray absorptiometry (DEXA) scan [86]. Preventative therapy such as smoking cessation, adequate nutrition, alcohol reduction, weight bearing exercises, and adequate daily vitamin D and calcium should be discussed and recommended in all menopausal HIV-infected women [129]. If the DEXA scan shows osteoporosis, bisphosphonates or other medical therapy should be considered. Although the data are limited, bisphosphonates have been shown to be effective in improving BMD [130–132].

Cognition

The menopause transition is characterized by cognitive changes such as memory loss and difficulty concentrating [133–136]. Both HIV-infected men and women are at higher risk of cognitive impairment [137–139]. Cognitive impairment can range from minor cognitive-motor disorder to HIV-associated dementia due to the immunologic, hormonal, and inflammatory effects of HIV on cognition [137–139]. In addition, those with HIV infection appear to have increased risk factors for cognitive impairment including low education level, psychiatric illnesses, increased social stress, and chemical dependence [137].

Studies focusing on the effects of both HIV infection and menopause on cognition have been limited thus far. In a cross-sectional study of 708 HIV-infected and 278 non–HIV-infected premenopausal, perimenopausal, and postmenopausal women, Rubin et al demonstrated that HIV infection, but not menopausal stage, was associated with worse performance on cognitive measures [140]. While menopausal stage was not associated with cognitive decline, menopausal symptoms like depression, anxiety, and vasomotor symptoms were associated with lower cognitive performance [140].

Though limited, current data appear to indicate that HIV infection, not menopause, contributes to cognitive dysfunction [140]. Symptoms of menopause, however, do appear to exacerbate cognitive decline indicating the importance of recognition and treatment of menopausal symptoms. This is especially important in HIV-infected women since decrease in cognition and depression can interfere with day to day function including medication adherence [141,142].

Cervical Dysplasia

As more HIV-infected women reach older age, the effects of prolonged survival and especially menopause on squamous intraepithelial lesions (SILs) are being investigated to determine if general guidelines of cervical cancer screening should be applied to postmenopausal women.

In a retrospective analysis of Papanicolaou smear results of 245 HIV-infected women, Kim et al noted that menopausal women had a 70% higher risk of progression of SILs than premenopausal women [143]. Similar results were found in a smaller retrospective study of 18 postmenopausal HIV-infected women in which postmenopausal women had a higher prevalence of SILs and persistence of low-grade SILs [144].

Although studies on progression to cervical cancer in postmenopausal HIV-infected women remain limited, current data suggest that postmenopausal HIV-infected women should continue to be monitored and screened similarly to the screening recommendations for premenopausal women. Nevertheless, further studies examining the natural course of cervical lesions are needed to establish the best practice guidelines for screening postmenopausal women.

HIV Acquisition and Transmission

The incidence of new HIV infections in older American women has increased. HIV acquisition from heterosexual contact appears to be higher in older women compared to younger women, with a study suggesting that women over age 45 years had almost a fourfold higher risk of HIV acquisition compared to those under the age of 45 years [145]. While the lack of awareness of HIV risk and less frequent use of protection may contribute to increases in new HIV infection in older women, hormonal changes associated with older age, specifically menopause, may be playing a role. Vaginal wall thinning that occurs during menopause may serve as a risk factor for HIV acquisition.

In a study by Meditz et al, the percentage of endocervical or blood CD4 T cells did not differ between premenopausal and postmenopausal women, but postmenopausal women had greater percentage of CCR5 expression. As CCR5 serves as an entry point of HIV into target cells, this suggests the possibility that postmenopausal women may be at increased risk for HIV acquisition [146]. More recently, Chappell et al also revealed that anti-HIV-1 activity was significantly decreased in postmenopausal compared to premenopausal women, suggesting that there may be an increased susceptibility to HIV-1 infection in postmenopausal women [147]. Hence there appears to be menopause-related immunologic changes of the cervix that may contribute to an increased risk of HIV acquisition in postmenopausal women.

In contrast, although data is limited, postmenopausal HIV-infected women do not appear to be at increased risk of transmitting HIV to non–HIV-infected individuals. Melo et al compared the intensity of HIV shedding between premenopausal and postmenopausal women and found that HIV shedding did not differ between premenopausal or postmenopausal women [148].

HIV Progression

Several studies have focused on the effects of HIV infection on menopause, but minimal data are available on the effects of menopause on the progression of HIV infection. With prior data suggesting that younger persons experience better immunological and virological responses to HAART [149–151], it has previously been hypothesized that virologic and immunologic responses to HAART can decline once HIV-infected women reach menopause. However, current evidence suggests that treatment responses to HAART, determined by the median changes in CD4 cell counts and percentages and viral load, in HAART-naive patients did not differ between premenopausal and postmenopausal women [152]. In addition, there appears to be no significant changes in CD4 cell counts as HIV-infected women progress through menopause [153]. These studies suggest that menopause does not affect the progression of HIV and that HAART-naive women should respond to HAART regardless of their menopausal status.

Conclusion

As HIV-infected individuals live longer, increasing number of women will enter into menopause and live many years beyond menopause. HIV-infected women experience earlier and more severe menopausal symptoms, but knowledge is still lacking on the appropriate management of these symptoms. In addition, current evidence suggests that immunosuppression associated with HIV contributes to an early onset of menopause which leads to increased risks of cardiovascular disease, osteoporosis, and progression of cervical dysplasia. These conditions require proper surveillance and can be prevented with improved understanding of influences of menopause on HIV-infected women. Furthermore, although there is some evidence suggesting that menopause has no effect on HIV transmission and progression, further studies on the immunologic and virologic effects of menopause are necessary.

There still remain significant gaps in our understanding of menopause in HIV-infected women.  As practitioners encounter an increasing number of perimenopausal and postmenopausal HIV-infected women, future studies on the effects of HIV on co-morbidities and symptoms of menopause and their appropriate management are necessary to improve care of women living with HIV.

 

Corresponding author: Lori E. Fantry, MD, MPH, 29 S. Greene St., Suite 300, Baltimore, MD 21201, lfantry@medicine.umaryland.edu.

Financial disclosures: None.

References

1. CASCADE Collaboration. Survival after introduction of HAART in people with known duration of HIV-1 infection. Lancet 2000;355:1158–9.

2. Brooks JT, Buchaz K, Gebo KA, Mermin J. HIV infection and older Americans: the public health perspective. Am J Pub Health 2012;102:1516–26.

3. Munster K, Helm P, Schmidt L. Secondary amenorrhea: Prevalence and medical contract–A cross sectional study from a Danish county. Br J Obstet Gynecol 1992;99:430–3.

4. Vyver E, Steinegger C, Katzman DK, et al. Eating disorders and menstrual dysfunction in adolescents. Ann N Y Acad Sci 2008;1135:253–64.

5. Abs R, Verhelst J, Maeyaert J, et al. Endocrine consequences of long-term intrathecal administration of opioids. J Clin Endocrinol Metab 2000;85:2215–22.

6. Pelosi MA, Sama JC, Caterini H, et al. Galactorrhea-amenorrhea syndrome associated with heroin addiction. Am J Obstet Gynecol 1974;118:966–70.

7. Bai J, Greenwald E, Caterini H, et al. Drug-related menstrual aberrations. Obstet Gynecol 1974;44:713–9.

8. Chirgwin KD, Feldman J, Muneyyirci-Delale O, et al. Menstrual function in HIV-infected women without AIDS. J Acquir Immune Defic Syndr Hum Retrovirol 1996;12:489–94.

9. Clark RA, Mulligan K, Stamenovic E, et al. Frequency of anovulation and early menopause among women enrolled in selected adult AIDS clinical trials group studies. J Infect Dis 2001;184:1325–7.

10. Watts DH, Spino C, Zaborski L. Comparison of gynecologic history and laboratory results in HIV-positive women with CDR+ lymphocyte counts between 200 and 500 cells/µl and below 100 cells/ µl. J Acquir Immune Defic Syndr Hum Retrovirol 1999;20:455–62.

11. Ellerbrock TV, Wrig TC, Bush TJ, et al. Characteristics of menstruation in women infected with HIV. Obstet Gynecol 1996;87:1030–4.

12. Shah PN, Smith JR, Wells C, et al. Menstrual symptoms in women infected by the HIV. Obstet Gynecol 1994;83:397–400.

13. Harlow SC, Schuman P, Cohen M, et al. Effect of HIV infection on menstrual cycle length. J Acquir Immune Defic Syndr Hum Retrovirol 2000;24:68–75.

14. Grinspoon S, Corocran C, Miller K, et al. Bone composition and endocrine function in women with AIDS wasting. J Clin Edocrinol Metab 1997;82:1332–7.

15. Research on the menopause in the 1990s. Report of a WHO scientific group. World Health Organ Tech Rep Ser 1996;866:1–107.

16. Cejtin HE, Kalinowski A, Bacchetti P. Effects of human immunodeficiency virus on protracted amenorrhea and ovarian dysfunction. Obstet Gynecol 2006;108:1423–31.

17. Freeman EW, Sammel MD, Garcia CR, et al.  Follicular phase hormone levels and menstrual bleeding status in the approach to menopause. Fertil Steril 2005;83:383–92.

18. Soules MR, Sherman S, Parrott E, et al. Executive summary: Stages of Reproductive Aging Workshop (STRAW). Fertil Steril 2001;76:874–8.

19. Gold EB, Crawford SL, Avis NE, et al. Factors related to age at natural menopause: longitudinal analyses from SWAN. Am J Epidemiol 2013;178:70–83.

20. Thomas F, Renaud F, Benefice E, et al. International variability of ages at menarche and menopause: patterns and main determinants. Hum Biol 2001;73:271–90.

21. Shuster LT, Rhodes DJ, Gostout BS, et al. Premature menopause or early menopause: long-term health consequences. Maturitas 2010;65:161–6.

22. Carr MC. The emergence of the metabolic syndrome with menopause. J Clin Endocrinol Metab 2003;88:2404–11.

23. Wellons M, Ouyang P, Schreiner PJ, et al. Early menopause predicts future coronary heart disease and stroke: the multi-ethnic study of atherosclerosis. Menopause 2012;19:1081–7.

24. Rocca WA, Grossardt BR, Miller VM, et al. Premature menopause or early menopause and risk of ischemic stroke. Menopause 2012;19:272–7.

25. Svejme O, Ahlborg HG, Nilsson JA, et al. Early menopause and risk of osteoporosis, fracture and mortality: a 34-year prospective observational study in 390 women. BJOG 2012;119:810–6.

26. Cooper GS, Sandler DP, Bohlig M. Active and passive smoking and the occurrence of natural menopause. Epidemiology 1999;10:771–3.

27. Luoto R, Kaprio J, Uutela A. Age at natural menopause and socioeconomic status in Finland. Am J Epidemiol 1994;139:64–76.

28. Bromberger JT, Matthews KA, Kuller LH, et al. Prospective study of the determinants of age at menopause. Am J Epidemiol 1997;145:24–33.

29. Gold EB, Crawford SL, Avis NE, et al. Factors related to age at natural menopause: longitudinal analyses from SWAN. Am J Epidemiol 2013;178:70–83.

30. Tziomalos K, Charsoulis F. Endocrine effects of tobacco smoking. Clin Endocrinol 2004;61:664–74.

31. Potter DA, Moreno A, Luther MF, et al. Effects of follicular-phase cocaine administration on menstrual and ovarian cyclicity in rhesus monkeys. Am J Obstet Gynecol 1998;178:118–25.

32. Mdodo R, Frazier EL, Dube SR, et al. Cigarette smoking prevalence among adults with HIV compared with the general adult population in the United States: Cross-sectional survey. Ann Intern Med 2015;162:335–44.

33. Centers for Disease Control and Prevention (CDC). Vital signs: current cigarette smoking among adults aged ≥ 18 years–United States, 2005-2010. MMWR Morb Mortal Wkly Rep 2011;60:1207–12.

34. Feldman J, Mikoff H, Schneider M, et al. Association of cigarette smoking with HIV prognosis among women in the HAART era: A report from the Women’s Interagency HIV study. Am J Public Health 2006:96:1060–5.

35. Centers for Disease Control and Prevention. Estimated HIV incidence among adults and adolescents in the United States, 2007–2010. HIV Surveillance Supplemental Report 2012;17(4).

36. Galea S, Ahren J, Vlahov D. Contextual determinants of drug use risk behavior: a theoretical framework. J Urban Health 2003;80:50–8.

37. de Pommerol M, Hessamfar M, Lawson-Ayayi S, et al. Menopause and HIV infection: age at onset and associated factors, ANRS CO3 Aquitaine cohort. Int J STD AIDS 2011;22:67–72.

38. Ferreira CE, Pinto-Neto AM, Conde DM, et al. Menopausal symptoms in women infected with HIV: prevalence and associated factors. Gynecol Endocrinol 2007;23:198–205.

39. Clark RA, Cohn SE, Jarck C, et al. Perimenopausal symptomatology among HIV infected women at least 40 years of age. J Acquir Immune Defic Syndr Hum Retrovirol 2000;23:99–100.

40. Fantry L, Zhan M, Taylor G, et al. Age at menopause and menopausal symptoms in HIV-infected women. AIDS Patient Care STD 2005;19:703–11.

41. Calvet G, Grinsztejn G. Predictors of early menopause in HIV infected women: a prospective cohort study. Am J Obstet Gynecol 2015;212:765.

42. Boonyanurak P, Bunupuradah T, Wilawan K, et al. Age at menopause and menopause-related symptoms in human immunodeficiency virus-infected Thai women. Menopause 2012;19:820–4.

43. Cejtin SH, Taylor R, Watts DH. Assessment of menopausal status among women in the Women’s Interagency HIV study (WIHS). Proceedings of the 57th International AIDS Conference 2004; Bangkok, Thailand.

44. WIHS Data Management and Analysis Center (WDMAC). Women’s Interagency HIV Study (WIHS) Dossier. October 2014. Available at https://statepiaps.jhsph.edu/wihs/invest-info/dossier.pdf.

45. Schoenbaum E, Hartel D, Lo Y, et al. HIV infection, drug use, and onset of natural menopause. Clinical Infect Dis 2005;41:1517–24.

46. Taffe JR, Dennerstein L. Menstrual patterns leading to the final menstrual period. Menopause 2002;9:32–40.

47. Miro F, Parker SW, Aspinall LJ, et al. Origins and consequences of the elongation of the human menstrual cycle during the menopausal transition: the FREEDOM Study. J Clin Endocrinol Metab 2004;89:4910–5.

48. Harlow SD, Gass M, Hall JE, et al. Executive summary of the Stages of Reproductive Aging Workshop + 10: addressing the unfinished agenda of staging reproductive aging. J Clin Endocrinol Metab 2012;97:1159–68.

49. Freeman EW, Sammel MD, Gracia CR, et al. Follicular phase hormone levels and menstrual bleeding status in the approach to menopause. Fertil Steril 2005;83:383–92.

50. Burger HG, Hale GE, Dennerstein L, Robertson DM. Cycle and hormone changes during perimenopause: the key role of ovarian function. Menopause 2008;15:603–12.

51. McKinlay SM, Brambilla DJ, Posner JG. The normal menopause transition. Maturitas 1992;14:103–15.

52. Szoeke CE, Cicuttini F, Guthrie J, Dennerstein L. Self-reported arthritis and the menopause. Climacteric 2005;8:49–55.

53. Blümel JE, Chedraui P, Baron G, et al. Menopause could be involved in the pathogenesis of muscle and joint aches in mid-aged women. Maturitas 2013;75:94–100.

54. Woods NF, Mitchell ES. Symptoms interference with work and relationships during the menopausal transition and early postmenopause: observations from the Seattle Midlife Women’s Health Study. Menopause 2011;18:654–61.

55. Johnson TM, Cohen HW, Howard AA, et al. Attribution of menopause symptoms in human immunodeficiency virus–infected or at-risk drug-using women Menopause 2008;15:551–7.

56. Miller SA, Santoro N, Lo Y. Menopausal symptoms in HIV-infected and drug-using women. Menopause 2005;12:348–56.

57. Looby S, Shifren J, Corless I. Increased hot flash severity and related interference in perimenopausal HIV-infected women. Menopause 2014;21:403–9.

58. Thurston RC, Joffe H. Vasomotor symptoms and menopause: findings from the Study of Women’s Health across the Nation. Obstet Gynecol Clin North Am 2011;38:489–501.

59. Woods NF, Mitchell ES. Symptoms during the perimenopause: prevalence, severity, trajectory, and significance in women’s lives. Am J Med 2005;118 Suppl 12B:14.

60. Maki PM, Rubin LH, Cohen M, et al. Depressive symptoms are increased in the early perimenopausal stage in ethnically diverse human immunodeficiency virus-infected and human immunodeficiency virus-uninfected women. Menopause 2012;19:1215–33.

61. Dennerstein L, Dudley EC, Hopper JL, et al. A prospective population-based study of menopausal symptoms. Obstet Gynecol 2000;96:351–8.

62. Palma F, Volpe A, Villa P, et al. Vaginal atrophy of women in postmenopause. Results from a multicentric observational study: The AGATA study. Maturitas 2015 Sep 14.

63. Cutler WB, Garcia CR, McCoy N. Perimenopausal sexuality.  Arch Sex Behav 1987;16:225–34.

64. Moreira ED, Glasser DB, Nicolosi A, et al. GSSAB Investigators’ Group.  Sexual problems and help-seeking behavior in adults in the United Kingdom and continental Europe. BJU Int 2008;101:1005–11.

65. MacBride MB, Rhodes DJ, Shuster LT. Vulvovaginal atrophy.  Mayo Clin Proc 2010;85:87–94.

66. Nappi RE, Kokot-Kierepa M. Women’s voices in the menopause: results from an international survey on vaginal atrophy. Maturitas 2010;67:233–8.

67. Santoro N, Komi J. Prevalence and impact of vaginal symptoms among postmenopausal women. J Sex Med 2009;6:2133–42.

68. Levine KB, Williams RE, Hartmann KE. Vulvovaginal atrophy is strongly associated with female sexual dysfunction among sexually active post-menopausal women. Menopause 2008;15(4 Pt 1):661–6.

69. Cumming GP, Currie HD, Moncur R, Lee AJ. Web-based survey on the effect of menopause on women’s libido in a computer-literate population. Menopause Int 2009;15:8–12.

70. Valadares AL, Pinto-Neto AM, Conde DM, et al. A population-based study of dyspareunia in a cohort of middle-aged Brazilian women. Menopause 2008;15:1184–90.

71. Latthe P, Migini L, Gray R, et al. Factors predisposing women to chronic pelvic pain: a systemic review. BMJ 2006;332:749–55.

72. Valadares AL, Pinto-Neto AM, Gomes D, et al. Dyspareunia in HIV-positive and HIV-negative middle-aged women: a cross-sectional study. BMJ Open 2014;4:e004974.

73. Bromberger JT, Meyer PM, Kravitz HM, et al. Psychologic distress and natural menopause: a multiethnic community study. Am J Public Health 2001;91:1435–42.

74. Avis NE, Brambilla D, McKinlay SM, Vass K. A longitudinal analysis of the association between menopause and depression. Results from the Massachusetts Women’s Health Study. Ann Epidemiol 1994;4:214–20.

75. Cohen LS, Soares CN, Joffe H. Diagnosis and management of mood disorders during the menopausal transition. Am J Med 2005;118 Suppl 12B:93–7.

76. Freeman EW, Sammel MD, Lin H, Nelson DB. Associations of hormones and menopausal status with depressed mood in women with no history of depression. Arch Gen Psychiatry 2006;63:375–82.

77. Eller LS, Corless I, Bunch EH, et al. Self-care strategies for depressive symptoms in people with HIV disease. J Adv Nurs 2005;51:119–30.

78. Fuh JL, Wang SJ, Lee SJ, et al. A longitudinal study of cognition change during early menopausal transition in a rural community. Maturitas 2006;53:447–53.

79. Greendale GA, Huang MH, Wight RG, et al. Effects of the menopause transition and hormone use on cognitive performance in midlife women. Neurology 2009;72:1850–7.

80. Hinkin CH, Castellon SA, Atkinson JH, et al. Neuropsychiatric aspects of HIV infection among older adults. J Clin Epidemiol 2001;54:S44–52.

81. Kravitz HM, Ganz PA, Bromberger J, et al. Sleep difficulty in women at midlife: a community survey of sleep and the menopausal transition. Menopause 2003;10:19–28.

82. Freedman RR, Roehrs TA. Effects of REM sleep and ambient temperature on hot flash-induced sleep disturbance. Menopause 2006;13:576–83.

83. Erlik Y, Tataryn IV, Meldrum DR, et al. Association of waking episodes with menopausal hot flushes. JAMA 1981; 245:1741–4.

84. Freedman RR, Roehrs TA. Sleep disturbance in menopause. Menopause 2007;14:826–9.

85. Lui-Filho JF, Valadares AR, Gomes D, et al. Menopausal symptoms and associated factors in HIV-positive women. Maturitas 2013;76:172–8.

86. Aberg JA, Gallant JE, Ghanem KG, et al, Infectious Diseases Society of America. Primary care guidelines for the management of persons infected with HIV: 2013 update by the HIV medicine association of the Infectious Diseases Society of America. Clin Infect Dis 2014;58:e1–34.

87. The role of local vaginal estrogen for treatment of vaginal atrophy in postmenopausal women: 2007 position statement of The North American Menopause Society. Menopause 2007;14:357–69.

88. Dorr MB, Nelson AL, Mayer PR, et al. Plasma estrogen concentrations after oral and vaginal estrogen administration in women with atrophic vaginitis. Fertil Steril 2010;94:2365–8.

89. El-Ibiary SY, Cocohoba JM. Effects of antiretrovirals on the pharmacokinetics of hormonal contraception. Eur J Contracept Reprod Health Care 2008;13:123–32.

90. Tittle V, Bull L, Boffito M, Nwokolo N. Pharmacokinetic and pharmacodynamics drug interactions between antiretrovirals and oral contraceptives. Clin Pharmacokinet 2015;54:23–34.

91. Thurman AR, Anderson S, Doncel G. Effects of hormonal contraception on anti-retroviral drug metabolism, pharmacokinetics and pharmacodynamics. Am J Reprod Immunol 2014:71:523–30.

92. Panel on Antiretroviral Guidelines for Adults and Adolescents. Guidelines for the use of antiretroviral agents in HIV-1-infected adults and adolescents. US. Department of Health and Human Services. Availabe at www.aidsinfo.nih.gov/ContentFiles/AdultandAdolescentGL.pdf.

93. Berg G, Mesch V, Boero L, et al. Lipid and lipoprotein profile in menopausal transition: effects of hormones, age and fat distribution. Hormone Metab Res 2004;36:215–20.

94. Sower M, Zheng H, Tomey K, et al. Changes in body composition in women over six years at midlife: ovarian and chronological aging. J Clin Endocrin Metab 2007;92:895–901.

95. Flooris-Moore M, Howard AA, Lo Y, et al. Increased serum lipids are associated with higher CD4 lymphocyte count in HIV-infected women. HIV Med 2006;7:421–30.

96. Grunfeld C, Delaney JA, Wanke C, et al. Preclinical atherosclerosis due to HIV infection: carotid intima-medial thickness measurements from the FRAM study. AIDS 2009;23:1841–9.

97. Palacios R, Alonso I, Hidalgo A, et al. Peripheral arterial disease in HIV patients older than 50 years of age. AIDS Res Hum Retroviruses 2008;24:1043–6.

98. Hadigan C, Meigs JB, Corcoran C, et al. Metabolic abnormalities and cardiovascular disease risk factors in adults with human immunodeficiency virus infection and lipodystrophy. Clin Infect Dis 2001;32:130–9.

99. Triant VA, Lee H, Hadigan C, Grinspoon SK. Increased acute myocardial infarction rates and cardiovascular risk factors among patients with human immunodeficiency virus disease. J Clin Endocrin Metab 2007;92:2506–12.

100. Dolan SE, Frontera W, Librizzi J et al. The effects of a supervised home based aerobic and progressive resistance training regimen in HIV-infected women: randomized trial. Arch Intern Med 2006;166:1225–31.

101. Grinspoon S, Carr A. Cardiovascular risk and body fat abnormalities in HIV-infected adults. N Engl J Med 2005;352:48–62

102. Study of Fat Redistribution and Metabolic Change in HIV Infection (FRAM). Fat distribution in women with HIV infection. J Acquir Immune Defic Syndr 2006;42:562–71.

103. Jaff NG, Norris SA, Snyman T, et al. Body composition in the study of women entering and in Endocrine Transition (SWEET): A perspective of African women who have a high prevalence of obesity and HIV infection. Metabolism 2015;64:1031–41.

104. Akhter MP, Lappe JM, Davies KM, et al. Transmenopausal changes in the trabecular bone structure. Bone 2007;41:111–6.

105. Cassetti I, Madruga JV, Suleiman JM, et al. The safety and efficacy of tenofovir DF in combination with lamivudine and efavirenz through 6 years in antiretroviral-naive HIV-1-infected patients. HIV Clin Trials 2007;8:164–72.

106. McComsey GA, Kitch D, Daar ES, et al. Bone mineral density and fractures in antiretroviral-naive persons randomized to receive abacavir-lamivudine or tenofovir disoproxil fumarate-emtricitabine along with efavirenz or atazanavir-ritonavir: AIDS Clinical Trials Group A5224s, a substudy of ACTG A5202. J Infect Dis 2011;203:1791–801.

107. Hansen AB, Obel N, Nielsen H, et al. Bone mineral density changes in protease inhibitor-sparing vs. nucleoside reverse transcriptase inhibitor-sparing highly active antiretroviral therapy: Data from a randomized trial. HIV Med 2011;12:157–65.

108. Yin MT, Zhang CA, McMahon DJ, et al. Higher rates of bone loss in postmenopausal HIV-infected women: a longitudinal study. J Clin Endocrinol Metab 2012;97:554–62.

109. Sharma A, Flom PL, Rosen CJ, et al. Racial differences in bone loss and relation to menopause among HIV-infected and uninfected women. Bone 2015;77:24–30.

110. Caputo BV, Traversa-Caputo GC, Costa C, et al. Evaluation of bone alterations in the jaws of HIV-infected menopausal women. Braz Oral Res 2013;27:231–7.

111. Bone mass and mineral metabolism in HIV+ postmenopausal women. Osteoporos Int 2005;26:1345–52.

112. Yin MT, Mcmahon DJ, Ferris DC, et al. Low bone mass and high bone turnover in postmenopausal human immunodeficiency virus-infected women. J Clin Endocrinol Metab 2010;95:620–9.

113. Gibellini D, De Crignis E, Ponti C. HIV-1 triggers apoptosis in primary osteoblasts and HOBIT cells through TNF-alpha activation. J Med Virol 2008;80:1507–14.

114. Tebas P, Powderly WG, Claxton S, et al. Accelerated bone mineral loss in HIV-infected patients receiving potent antiretroviral therapy. AIDS 2000;14:F63–7.

115. Van Rompay KK, Brignolo LL, Meyer DJ, et al. Biological effects of short-term or prolonged administration of 9-[2(phosphonomethoxy)propyl] adenine (tenofovir) to newborn and infant rhesus macaques. Antimicrob Agents Chemother 2004;48:1469–87.

116. Brown TT, Qaqish RB. Antiretroviral therapy and the prevalence of osteopenia and osteoporosis: a meta-analytic review. AIDS 2006;20:2165–74.

117. Dao CN, Patel P, Overton ET, Rhame F, et al. Study to understand the natural history of HIV and AIDS in the era of effective therapy (SUN) investigators. Low vitamin D among HIV-infected adults: prevalence of and risk factors for low vitamin D levels in cohort of HIV-infected adults and comparison to prevalence among adults in the US general population. Clin Infect Dis 2011;52:396–405.

118. Jacobson DL, Spiegelman D, Know TK, Wilson IB. Evolution and predictors of change in total bone mineral density over time in HIV-infected men and women in the nutrition for healthy living study. J Acquir Immune Defic Syndr Hum Retrovirol 2008;49:298–308.

119. Kanis JA, Borgstrom F, De Laet C, et al. Assessment of fracture risk. Osteoporosis Int  2005;16:581–9

120. Pedrazzoni M, Vescovi L, Maninetti M, et al. Effects of chronic heroine abuse on bone and mineral metabolism. Acta Endocrinol 1993;129:42–5.

121. Lo Re V 3rd, Guaraldi G, Leonard MB, et al. Viral hepatitis is associated with reduced bone mineral density in HIV-infected women but not men. AIDS 1990;23:2191–8.

122. Yin MT, Modarresi R, Shane E, et al. Effects of HIV infection and antiretroviral therapy with ritonavir on induction of osteoclast-like cells in postmenopausal women. Osteoporos Int 2011;22:1459–66.

123. Sharma A, Cohen HW, Freeman R, et al. Prospective evaluation of bone mineral density among middle-aged HIV-infected and uninfected women: association between methadone use and bone loss. Maturitas 2011;70:295–301.

124. Triant VA, Brown TT, Lee H, Grinspoon SK. Fracture prevalence among human immunodeficiency virus (HIV)-infected versus non-HIV-infected patients in a large U.S. healthcare system. J Clin Endocrinol Metab 2008;93:3499–504.

125. Womack JA, Goulet JL, Gibert C, et al. Veterans Aging Cohort Study Project Team. Increased risk of fragility fractures among HIV infected compared to uninfected male veterans. PLoS One Feb 16 2011;6:e17217.

126. Young B, Dao CN, Buchacz K, et al, HIV Outpatient Study (HOPS) Investigators. Increased rates of bone fracture among HIV-infected persons in the HIV Outpatient Study (HOPS) compared with the US general population, 2000–2006. Clin Infect Dis 2011;52:1061–8.

127. U.S. Preventive Services Task Force. Screening for osteoporosis: U.S. Preventive Services Task Force recommendation statement. Ann Intern Med 2011; 154:356–64.

128. Aberg JA, Kaplan JE, Libman H, et al; HIV Medicine Association of the Infectious Diseases Society of America. Primary care guidelines for the management of persons infected with human immunodeficiency virus: 2009 update by the HIV medicine Association of the Infectious Diseases Society of America. Clin Infect Dis 2009;49:651–81.

129. National Osteoporosis Foundation. Clinician’s guide to prevention and treatment of osteoporosis 2014. Washington, DC: National Osteoporosis Foundation; 2014.

130. McComsey GA, Tebas P, Shane E, et al. Bone disease in HIV infection: a practical review and recommendations for HIV care providers. Clin Infect Dis 2010;51:937–46.

131. McComsey GA, Kendall MA, Tebas P, et al. Alendronate with calcium and vitamin D supplementation is safe and effective for the treatment of decreased bone mineral density in HIV. AIDS 2007;21:2473–82.

132. Lin D, Rieder MJ. Interventions for the treatment of decreased bone mineral density associated with HIV infection. Cochrane Database Syst Rev 2007:CD005645.

133. Haring B, Leng X, Robinson J. Cardiovascular disease and cognitive decline in postmenopausal women: results from the Women’s Health Initiative Memory Study. J Am Heart Assoc 2013;2:e000369.

134. Soares CN, Maki PM. Menopausal transition, mood, and cognition: an integrated view to close the gaps. Menopause 2010;17:812–4.

135. Greendale GA, Derby CA, Maki PM. Perimenopause and cognition. Obstet Gynecol Clin North Am 2011;38:519–35.

136. Greendale GA, Wight RG, Huang MH, et al. Menopause-associated symptoms and cognitive performance: results from the study of women’s health across the nation. Am J Epidemiol 2010;171:1214–24.

137. Price RW. Neurological complications of HIV infection. Lancet 1996;348:445–52.

138. Antinori A, Arendt G, Becker JT, et al. Updated research nosology for HIV-associated neurocognitive disorders. Neurology 2007;69:1789–99.

139. Gisslén M, Price RW, Nilsson S. The definition of HIV-associated neurocognitive disorders: are we overestimating the real prevalence? BMC Infect Dis 2011;11:356.

140. Rubin LH, Sundermann EE, Cook JA, et al. An investigation of menopausal stage and symptoms on cognition in HIV-infected women. Menopause 2014;21:997–1006.

141. Cook JA, Cohen MH, Burke J, et al. Effects of depressive symptoms and mental health quality of life on use of highly active antiretroviral therapy among HIV-seropositive women. J Acquir Immune Defic Syndr 2002;30:401–9.

142. Cook JA, Grey D, Burke J, et al. Depressive symptoms and AIDS-related mortality among a multisite cohort of HIV-positive women. Am J Pub Health 2004;94:1133–40.

143. Kim SC, Messing S, Shah K, et al. Effects of highly active antiretroviral therapy (HAART) and menopause on risk of progression of cervical dysplasia in human immune deficiency virus (HIV) infected women. Infect Dis Obstet Gynecol 2013;2013:784718.

144. Ceccaldi PF, Ferreira C, Coussy F, et al. Cervical disease in postmenopausal HIV-1 infected women. J Gynecol Obstet Biol Reprod 2010;39:466–70.

145. European Study Group on Heterosexual Transmission of HIV. Comparison of female to male and male to female transmission of HIV in 563 stable couples. BMJ 1992;304:809–13.

146. Meditz AL, Moreau KL, MaWhinney S, et al. CCR5 expression is elevated on endocervical CD4+ T cells in healthy postmenopausal women. J Acquir Immune Defic Syndr 2012;59:221–8.

147. Chappell CA, Isaacs CE, Xu W, et al. The effect of menopause on the innate antiviral activity of cervicovaginal lavage. Am J Obstet Gynecol 2015;213:204.

148. Melo KC, Melo MR, Ricci BV, Segurado AC. Correlates of human immunodeficiency virus cervicovaginal shedding among postmenopausal and fertile-aged women. Menopause 2012;19:150–6.

149. Viard JP, Mocroft A, Chiesi A, et al. Influence of age of CD4 cell recovery in human immunodeficiency virus-infected patients receiving highly active antiretroviral therapy: evidence from the Euro SIDA study. J Infect Dis 2001;193:1290–4.

150. Grabar S, Kousignian I, Sobel A, et al. Immunological and clinical responses to highly active antiretroviral therapy over 50 years of age. Results from the French Hospital Database on HIV. AIDS 2004;18:2029–38.

151. Cuzin L, Delpierre C, Gerard S, et al. Immunologic and clinical responses to highly active antiretroviral therapy in patients with HIV infection aged >50 years. Clin Infect Dis 2007;45:654–7.

152. Patterson KB, Cohn SE, Uynik J, et al. Treatment responses in antiretroviral treatment-naïve premenopausal and postmenopausal HIV-1 infected women: an analysis from AIDS clinical trials group studies. Clin Infect Dis 2009;49:473–6.

153. van Benthem BH, Vernazza P, Coutinho RA, et al. The impact of pregnancy and menopause on CD4 lymphocyte count in HIV-infected women. AIDS 2002;16:919–24.

References

1. CASCADE Collaboration. Survival after introduction of HAART in people with known duration of HIV-1 infection. Lancet 2000;355:1158–9.

2. Brooks JT, Buchaz K, Gebo KA, Mermin J. HIV infection and older Americans: the public health perspective. Am J Pub Health 2012;102:1516–26.

3. Munster K, Helm P, Schmidt L. Secondary amenorrhea: Prevalence and medical contract–A cross sectional study from a Danish county. Br J Obstet Gynecol 1992;99:430–3.

4. Vyver E, Steinegger C, Katzman DK, et al. Eating disorders and menstrual dysfunction in adolescents. Ann N Y Acad Sci 2008;1135:253–64.

5. Abs R, Verhelst J, Maeyaert J, et al. Endocrine consequences of long-term intrathecal administration of opioids. J Clin Endocrinol Metab 2000;85:2215–22.

6. Pelosi MA, Sama JC, Caterini H, et al. Galactorrhea-amenorrhea syndrome associated with heroin addiction. Am J Obstet Gynecol 1974;118:966–70.

7. Bai J, Greenwald E, Caterini H, et al. Drug-related menstrual aberrations. Obstet Gynecol 1974;44:713–9.

8. Chirgwin KD, Feldman J, Muneyyirci-Delale O, et al. Menstrual function in HIV-infected women without AIDS. J Acquir Immune Defic Syndr Hum Retrovirol 1996;12:489–94.

9. Clark RA, Mulligan K, Stamenovic E, et al. Frequency of anovulation and early menopause among women enrolled in selected adult AIDS clinical trials group studies. J Infect Dis 2001;184:1325–7.

10. Watts DH, Spino C, Zaborski L. Comparison of gynecologic history and laboratory results in HIV-positive women with CDR+ lymphocyte counts between 200 and 500 cells/µl and below 100 cells/ µl. J Acquir Immune Defic Syndr Hum Retrovirol 1999;20:455–62.

11. Ellerbrock TV, Wrig TC, Bush TJ, et al. Characteristics of menstruation in women infected with HIV. Obstet Gynecol 1996;87:1030–4.

12. Shah PN, Smith JR, Wells C, et al. Menstrual symptoms in women infected by the HIV. Obstet Gynecol 1994;83:397–400.

13. Harlow SC, Schuman P, Cohen M, et al. Effect of HIV infection on menstrual cycle length. J Acquir Immune Defic Syndr Hum Retrovirol 2000;24:68–75.

14. Grinspoon S, Corocran C, Miller K, et al. Bone composition and endocrine function in women with AIDS wasting. J Clin Edocrinol Metab 1997;82:1332–7.

15. Research on the menopause in the 1990s. Report of a WHO scientific group. World Health Organ Tech Rep Ser 1996;866:1–107.

16. Cejtin HE, Kalinowski A, Bacchetti P. Effects of human immunodeficiency virus on protracted amenorrhea and ovarian dysfunction. Obstet Gynecol 2006;108:1423–31.

17. Freeman EW, Sammel MD, Garcia CR, et al.  Follicular phase hormone levels and menstrual bleeding status in the approach to menopause. Fertil Steril 2005;83:383–92.

18. Soules MR, Sherman S, Parrott E, et al. Executive summary: Stages of Reproductive Aging Workshop (STRAW). Fertil Steril 2001;76:874–8.

19. Gold EB, Crawford SL, Avis NE, et al. Factors related to age at natural menopause: longitudinal analyses from SWAN. Am J Epidemiol 2013;178:70–83.

20. Thomas F, Renaud F, Benefice E, et al. International variability of ages at menarche and menopause: patterns and main determinants. Hum Biol 2001;73:271–90.

21. Shuster LT, Rhodes DJ, Gostout BS, et al. Premature menopause or early menopause: long-term health consequences. Maturitas 2010;65:161–6.

22. Carr MC. The emergence of the metabolic syndrome with menopause. J Clin Endocrinol Metab 2003;88:2404–11.

23. Wellons M, Ouyang P, Schreiner PJ, et al. Early menopause predicts future coronary heart disease and stroke: the multi-ethnic study of atherosclerosis. Menopause 2012;19:1081–7.

24. Rocca WA, Grossardt BR, Miller VM, et al. Premature menopause or early menopause and risk of ischemic stroke. Menopause 2012;19:272–7.

25. Svejme O, Ahlborg HG, Nilsson JA, et al. Early menopause and risk of osteoporosis, fracture and mortality: a 34-year prospective observational study in 390 women. BJOG 2012;119:810–6.

26. Cooper GS, Sandler DP, Bohlig M. Active and passive smoking and the occurrence of natural menopause. Epidemiology 1999;10:771–3.

27. Luoto R, Kaprio J, Uutela A. Age at natural menopause and socioeconomic status in Finland. Am J Epidemiol 1994;139:64–76.

28. Bromberger JT, Matthews KA, Kuller LH, et al. Prospective study of the determinants of age at menopause. Am J Epidemiol 1997;145:24–33.

29. Gold EB, Crawford SL, Avis NE, et al. Factors related to age at natural menopause: longitudinal analyses from SWAN. Am J Epidemiol 2013;178:70–83.

30. Tziomalos K, Charsoulis F. Endocrine effects of tobacco smoking. Clin Endocrinol 2004;61:664–74.

31. Potter DA, Moreno A, Luther MF, et al. Effects of follicular-phase cocaine administration on menstrual and ovarian cyclicity in rhesus monkeys. Am J Obstet Gynecol 1998;178:118–25.

32. Mdodo R, Frazier EL, Dube SR, et al. Cigarette smoking prevalence among adults with HIV compared with the general adult population in the United States: Cross-sectional survey. Ann Intern Med 2015;162:335–44.

33. Centers for Disease Control and Prevention (CDC). Vital signs: current cigarette smoking among adults aged ≥ 18 years–United States, 2005-2010. MMWR Morb Mortal Wkly Rep 2011;60:1207–12.

34. Feldman J, Mikoff H, Schneider M, et al. Association of cigarette smoking with HIV prognosis among women in the HAART era: A report from the Women’s Interagency HIV study. Am J Public Health 2006:96:1060–5.

35. Centers for Disease Control and Prevention. Estimated HIV incidence among adults and adolescents in the United States, 2007–2010. HIV Surveillance Supplemental Report 2012;17(4).

36. Galea S, Ahren J, Vlahov D. Contextual determinants of drug use risk behavior: a theoretical framework. J Urban Health 2003;80:50–8.

37. de Pommerol M, Hessamfar M, Lawson-Ayayi S, et al. Menopause and HIV infection: age at onset and associated factors, ANRS CO3 Aquitaine cohort. Int J STD AIDS 2011;22:67–72.

38. Ferreira CE, Pinto-Neto AM, Conde DM, et al. Menopausal symptoms in women infected with HIV: prevalence and associated factors. Gynecol Endocrinol 2007;23:198–205.

39. Clark RA, Cohn SE, Jarck C, et al. Perimenopausal symptomatology among HIV infected women at least 40 years of age. J Acquir Immune Defic Syndr Hum Retrovirol 2000;23:99–100.

40. Fantry L, Zhan M, Taylor G, et al. Age at menopause and menopausal symptoms in HIV-infected women. AIDS Patient Care STD 2005;19:703–11.

41. Calvet G, Grinsztejn G. Predictors of early menopause in HIV infected women: a prospective cohort study. Am J Obstet Gynecol 2015;212:765.

42. Boonyanurak P, Bunupuradah T, Wilawan K, et al. Age at menopause and menopause-related symptoms in human immunodeficiency virus-infected Thai women. Menopause 2012;19:820–4.

43. Cejtin SH, Taylor R, Watts DH. Assessment of menopausal status among women in the Women’s Interagency HIV study (WIHS). Proceedings of the 57th International AIDS Conference 2004; Bangkok, Thailand.

44. WIHS Data Management and Analysis Center (WDMAC). Women’s Interagency HIV Study (WIHS) Dossier. October 2014. Available at https://statepiaps.jhsph.edu/wihs/invest-info/dossier.pdf.

45. Schoenbaum E, Hartel D, Lo Y, et al. HIV infection, drug use, and onset of natural menopause. Clinical Infect Dis 2005;41:1517–24.

46. Taffe JR, Dennerstein L. Menstrual patterns leading to the final menstrual period. Menopause 2002;9:32–40.

47. Miro F, Parker SW, Aspinall LJ, et al. Origins and consequences of the elongation of the human menstrual cycle during the menopausal transition: the FREEDOM Study. J Clin Endocrinol Metab 2004;89:4910–5.

48. Harlow SD, Gass M, Hall JE, et al. Executive summary of the Stages of Reproductive Aging Workshop + 10: addressing the unfinished agenda of staging reproductive aging. J Clin Endocrinol Metab 2012;97:1159–68.

49. Freeman EW, Sammel MD, Gracia CR, et al. Follicular phase hormone levels and menstrual bleeding status in the approach to menopause. Fertil Steril 2005;83:383–92.

50. Burger HG, Hale GE, Dennerstein L, Robertson DM. Cycle and hormone changes during perimenopause: the key role of ovarian function. Menopause 2008;15:603–12.

51. McKinlay SM, Brambilla DJ, Posner JG. The normal menopause transition. Maturitas 1992;14:103–15.

52. Szoeke CE, Cicuttini F, Guthrie J, Dennerstein L. Self-reported arthritis and the menopause. Climacteric 2005;8:49–55.

53. Blümel JE, Chedraui P, Baron G, et al. Menopause could be involved in the pathogenesis of muscle and joint aches in mid-aged women. Maturitas 2013;75:94–100.

54. Woods NF, Mitchell ES. Symptoms interference with work and relationships during the menopausal transition and early postmenopause: observations from the Seattle Midlife Women’s Health Study. Menopause 2011;18:654–61.

55. Johnson TM, Cohen HW, Howard AA, et al. Attribution of menopause symptoms in human immunodeficiency virus–infected or at-risk drug-using women Menopause 2008;15:551–7.

56. Miller SA, Santoro N, Lo Y. Menopausal symptoms in HIV-infected and drug-using women. Menopause 2005;12:348–56.

57. Looby S, Shifren J, Corless I. Increased hot flash severity and related interference in perimenopausal HIV-infected women. Menopause 2014;21:403–9.

58. Thurston RC, Joffe H. Vasomotor symptoms and menopause: findings from the Study of Women’s Health across the Nation. Obstet Gynecol Clin North Am 2011;38:489–501.

59. Woods NF, Mitchell ES. Symptoms during the perimenopause: prevalence, severity, trajectory, and significance in women’s lives. Am J Med 2005;118 Suppl 12B:14.

60. Maki PM, Rubin LH, Cohen M, et al. Depressive symptoms are increased in the early perimenopausal stage in ethnically diverse human immunodeficiency virus-infected and human immunodeficiency virus-uninfected women. Menopause 2012;19:1215–33.

61. Dennerstein L, Dudley EC, Hopper JL, et al. A prospective population-based study of menopausal symptoms. Obstet Gynecol 2000;96:351–8.

62. Palma F, Volpe A, Villa P, et al. Vaginal atrophy of women in postmenopause. Results from a multicentric observational study: The AGATA study. Maturitas 2015 Sep 14.

63. Cutler WB, Garcia CR, McCoy N. Perimenopausal sexuality.  Arch Sex Behav 1987;16:225–34.

64. Moreira ED, Glasser DB, Nicolosi A, et al. GSSAB Investigators’ Group.  Sexual problems and help-seeking behavior in adults in the United Kingdom and continental Europe. BJU Int 2008;101:1005–11.

65. MacBride MB, Rhodes DJ, Shuster LT. Vulvovaginal atrophy.  Mayo Clin Proc 2010;85:87–94.

66. Nappi RE, Kokot-Kierepa M. Women’s voices in the menopause: results from an international survey on vaginal atrophy. Maturitas 2010;67:233–8.

67. Santoro N, Komi J. Prevalence and impact of vaginal symptoms among postmenopausal women. J Sex Med 2009;6:2133–42.

68. Levine KB, Williams RE, Hartmann KE. Vulvovaginal atrophy is strongly associated with female sexual dysfunction among sexually active post-menopausal women. Menopause 2008;15(4 Pt 1):661–6.

69. Cumming GP, Currie HD, Moncur R, Lee AJ. Web-based survey on the effect of menopause on women’s libido in a computer-literate population. Menopause Int 2009;15:8–12.

70. Valadares AL, Pinto-Neto AM, Conde DM, et al. A population-based study of dyspareunia in a cohort of middle-aged Brazilian women. Menopause 2008;15:1184–90.

71. Latthe P, Migini L, Gray R, et al. Factors predisposing women to chronic pelvic pain: a systemic review. BMJ 2006;332:749–55.

72. Valadares AL, Pinto-Neto AM, Gomes D, et al. Dyspareunia in HIV-positive and HIV-negative middle-aged women: a cross-sectional study. BMJ Open 2014;4:e004974.

73. Bromberger JT, Meyer PM, Kravitz HM, et al. Psychologic distress and natural menopause: a multiethnic community study. Am J Public Health 2001;91:1435–42.

74. Avis NE, Brambilla D, McKinlay SM, Vass K. A longitudinal analysis of the association between menopause and depression. Results from the Massachusetts Women’s Health Study. Ann Epidemiol 1994;4:214–20.

75. Cohen LS, Soares CN, Joffe H. Diagnosis and management of mood disorders during the menopausal transition. Am J Med 2005;118 Suppl 12B:93–7.

76. Freeman EW, Sammel MD, Lin H, Nelson DB. Associations of hormones and menopausal status with depressed mood in women with no history of depression. Arch Gen Psychiatry 2006;63:375–82.

77. Eller LS, Corless I, Bunch EH, et al. Self-care strategies for depressive symptoms in people with HIV disease. J Adv Nurs 2005;51:119–30.

78. Fuh JL, Wang SJ, Lee SJ, et al. A longitudinal study of cognition change during early menopausal transition in a rural community. Maturitas 2006;53:447–53.

79. Greendale GA, Huang MH, Wight RG, et al. Effects of the menopause transition and hormone use on cognitive performance in midlife women. Neurology 2009;72:1850–7.

80. Hinkin CH, Castellon SA, Atkinson JH, et al. Neuropsychiatric aspects of HIV infection among older adults. J Clin Epidemiol 2001;54:S44–52.

81. Kravitz HM, Ganz PA, Bromberger J, et al. Sleep difficulty in women at midlife: a community survey of sleep and the menopausal transition. Menopause 2003;10:19–28.

82. Freedman RR, Roehrs TA. Effects of REM sleep and ambient temperature on hot flash-induced sleep disturbance. Menopause 2006;13:576–83.

83. Erlik Y, Tataryn IV, Meldrum DR, et al. Association of waking episodes with menopausal hot flushes. JAMA 1981; 245:1741–4.

84. Freedman RR, Roehrs TA. Sleep disturbance in menopause. Menopause 2007;14:826–9.

85. Lui-Filho JF, Valadares AR, Gomes D, et al. Menopausal symptoms and associated factors in HIV-positive women. Maturitas 2013;76:172–8.

86. Aberg JA, Gallant JE, Ghanem KG, et al, Infectious Diseases Society of America. Primary care guidelines for the management of persons infected with HIV: 2013 update by the HIV medicine association of the Infectious Diseases Society of America. Clin Infect Dis 2014;58:e1–34.

87. The role of local vaginal estrogen for treatment of vaginal atrophy in postmenopausal women: 2007 position statement of The North American Menopause Society. Menopause 2007;14:357–69.

88. Dorr MB, Nelson AL, Mayer PR, et al. Plasma estrogen concentrations after oral and vaginal estrogen administration in women with atrophic vaginitis. Fertil Steril 2010;94:2365–8.

89. El-Ibiary SY, Cocohoba JM. Effects of antiretrovirals on the pharmacokinetics of hormonal contraception. Eur J Contracept Reprod Health Care 2008;13:123–32.

90. Tittle V, Bull L, Boffito M, Nwokolo N. Pharmacokinetic and pharmacodynamics drug interactions between antiretrovirals and oral contraceptives. Clin Pharmacokinet 2015;54:23–34.

91. Thurman AR, Anderson S, Doncel G. Effects of hormonal contraception on anti-retroviral drug metabolism, pharmacokinetics and pharmacodynamics. Am J Reprod Immunol 2014:71:523–30.

92. Panel on Antiretroviral Guidelines for Adults and Adolescents. Guidelines for the use of antiretroviral agents in HIV-1-infected adults and adolescents. US. Department of Health and Human Services. Availabe at www.aidsinfo.nih.gov/ContentFiles/AdultandAdolescentGL.pdf.

93. Berg G, Mesch V, Boero L, et al. Lipid and lipoprotein profile in menopausal transition: effects of hormones, age and fat distribution. Hormone Metab Res 2004;36:215–20.

94. Sower M, Zheng H, Tomey K, et al. Changes in body composition in women over six years at midlife: ovarian and chronological aging. J Clin Endocrin Metab 2007;92:895–901.

95. Flooris-Moore M, Howard AA, Lo Y, et al. Increased serum lipids are associated with higher CD4 lymphocyte count in HIV-infected women. HIV Med 2006;7:421–30.

96. Grunfeld C, Delaney JA, Wanke C, et al. Preclinical atherosclerosis due to HIV infection: carotid intima-medial thickness measurements from the FRAM study. AIDS 2009;23:1841–9.

97. Palacios R, Alonso I, Hidalgo A, et al. Peripheral arterial disease in HIV patients older than 50 years of age. AIDS Res Hum Retroviruses 2008;24:1043–6.

98. Hadigan C, Meigs JB, Corcoran C, et al. Metabolic abnormalities and cardiovascular disease risk factors in adults with human immunodeficiency virus infection and lipodystrophy. Clin Infect Dis 2001;32:130–9.

99. Triant VA, Lee H, Hadigan C, Grinspoon SK. Increased acute myocardial infarction rates and cardiovascular risk factors among patients with human immunodeficiency virus disease. J Clin Endocrin Metab 2007;92:2506–12.

100. Dolan SE, Frontera W, Librizzi J et al. The effects of a supervised home based aerobic and progressive resistance training regimen in HIV-infected women: randomized trial. Arch Intern Med 2006;166:1225–31.

101. Grinspoon S, Carr A. Cardiovascular risk and body fat abnormalities in HIV-infected adults. N Engl J Med 2005;352:48–62

102. Study of Fat Redistribution and Metabolic Change in HIV Infection (FRAM). Fat distribution in women with HIV infection. J Acquir Immune Defic Syndr 2006;42:562–71.

103. Jaff NG, Norris SA, Snyman T, et al. Body composition in the study of women entering and in Endocrine Transition (SWEET): A perspective of African women who have a high prevalence of obesity and HIV infection. Metabolism 2015;64:1031–41.

104. Akhter MP, Lappe JM, Davies KM, et al. Transmenopausal changes in the trabecular bone structure. Bone 2007;41:111–6.

105. Cassetti I, Madruga JV, Suleiman JM, et al. The safety and efficacy of tenofovir DF in combination with lamivudine and efavirenz through 6 years in antiretroviral-naive HIV-1-infected patients. HIV Clin Trials 2007;8:164–72.

106. McComsey GA, Kitch D, Daar ES, et al. Bone mineral density and fractures in antiretroviral-naive persons randomized to receive abacavir-lamivudine or tenofovir disoproxil fumarate-emtricitabine along with efavirenz or atazanavir-ritonavir: AIDS Clinical Trials Group A5224s, a substudy of ACTG A5202. J Infect Dis 2011;203:1791–801.

107. Hansen AB, Obel N, Nielsen H, et al. Bone mineral density changes in protease inhibitor-sparing vs. nucleoside reverse transcriptase inhibitor-sparing highly active antiretroviral therapy: Data from a randomized trial. HIV Med 2011;12:157–65.

108. Yin MT, Zhang CA, McMahon DJ, et al. Higher rates of bone loss in postmenopausal HIV-infected women: a longitudinal study. J Clin Endocrinol Metab 2012;97:554–62.

109. Sharma A, Flom PL, Rosen CJ, et al. Racial differences in bone loss and relation to menopause among HIV-infected and uninfected women. Bone 2015;77:24–30.

110. Caputo BV, Traversa-Caputo GC, Costa C, et al. Evaluation of bone alterations in the jaws of HIV-infected menopausal women. Braz Oral Res 2013;27:231–7.

111. Bone mass and mineral metabolism in HIV+ postmenopausal women. Osteoporos Int 2005;26:1345–52.

112. Yin MT, Mcmahon DJ, Ferris DC, et al. Low bone mass and high bone turnover in postmenopausal human immunodeficiency virus-infected women. J Clin Endocrinol Metab 2010;95:620–9.

113. Gibellini D, De Crignis E, Ponti C. HIV-1 triggers apoptosis in primary osteoblasts and HOBIT cells through TNF-alpha activation. J Med Virol 2008;80:1507–14.

114. Tebas P, Powderly WG, Claxton S, et al. Accelerated bone mineral loss in HIV-infected patients receiving potent antiretroviral therapy. AIDS 2000;14:F63–7.

115. Van Rompay KK, Brignolo LL, Meyer DJ, et al. Biological effects of short-term or prolonged administration of 9-[2(phosphonomethoxy)propyl] adenine (tenofovir) to newborn and infant rhesus macaques. Antimicrob Agents Chemother 2004;48:1469–87.

116. Brown TT, Qaqish RB. Antiretroviral therapy and the prevalence of osteopenia and osteoporosis: a meta-analytic review. AIDS 2006;20:2165–74.

117. Dao CN, Patel P, Overton ET, Rhame F, et al. Study to understand the natural history of HIV and AIDS in the era of effective therapy (SUN) investigators. Low vitamin D among HIV-infected adults: prevalence of and risk factors for low vitamin D levels in cohort of HIV-infected adults and comparison to prevalence among adults in the US general population. Clin Infect Dis 2011;52:396–405.

118. Jacobson DL, Spiegelman D, Know TK, Wilson IB. Evolution and predictors of change in total bone mineral density over time in HIV-infected men and women in the nutrition for healthy living study. J Acquir Immune Defic Syndr Hum Retrovirol 2008;49:298–308.

119. Kanis JA, Borgstrom F, De Laet C, et al. Assessment of fracture risk. Osteoporosis Int  2005;16:581–9

120. Pedrazzoni M, Vescovi L, Maninetti M, et al. Effects of chronic heroine abuse on bone and mineral metabolism. Acta Endocrinol 1993;129:42–5.

121. Lo Re V 3rd, Guaraldi G, Leonard MB, et al. Viral hepatitis is associated with reduced bone mineral density in HIV-infected women but not men. AIDS 1990;23:2191–8.

122. Yin MT, Modarresi R, Shane E, et al. Effects of HIV infection and antiretroviral therapy with ritonavir on induction of osteoclast-like cells in postmenopausal women. Osteoporos Int 2011;22:1459–66.

123. Sharma A, Cohen HW, Freeman R, et al. Prospective evaluation of bone mineral density among middle-aged HIV-infected and uninfected women: association between methadone use and bone loss. Maturitas 2011;70:295–301.

124. Triant VA, Brown TT, Lee H, Grinspoon SK. Fracture prevalence among human immunodeficiency virus (HIV)-infected versus non-HIV-infected patients in a large U.S. healthcare system. J Clin Endocrinol Metab 2008;93:3499–504.

125. Womack JA, Goulet JL, Gibert C, et al. Veterans Aging Cohort Study Project Team. Increased risk of fragility fractures among HIV infected compared to uninfected male veterans. PLoS One Feb 16 2011;6:e17217.

126. Young B, Dao CN, Buchacz K, et al, HIV Outpatient Study (HOPS) Investigators. Increased rates of bone fracture among HIV-infected persons in the HIV Outpatient Study (HOPS) compared with the US general population, 2000–2006. Clin Infect Dis 2011;52:1061–8.

127. U.S. Preventive Services Task Force. Screening for osteoporosis: U.S. Preventive Services Task Force recommendation statement. Ann Intern Med 2011; 154:356–64.

128. Aberg JA, Kaplan JE, Libman H, et al; HIV Medicine Association of the Infectious Diseases Society of America. Primary care guidelines for the management of persons infected with human immunodeficiency virus: 2009 update by the HIV medicine Association of the Infectious Diseases Society of America. Clin Infect Dis 2009;49:651–81.

129. National Osteoporosis Foundation. Clinician’s guide to prevention and treatment of osteoporosis 2014. Washington, DC: National Osteoporosis Foundation; 2014.

130. McComsey GA, Tebas P, Shane E, et al. Bone disease in HIV infection: a practical review and recommendations for HIV care providers. Clin Infect Dis 2010;51:937–46.

131. McComsey GA, Kendall MA, Tebas P, et al. Alendronate with calcium and vitamin D supplementation is safe and effective for the treatment of decreased bone mineral density in HIV. AIDS 2007;21:2473–82.

132. Lin D, Rieder MJ. Interventions for the treatment of decreased bone mineral density associated with HIV infection. Cochrane Database Syst Rev 2007:CD005645.

133. Haring B, Leng X, Robinson J. Cardiovascular disease and cognitive decline in postmenopausal women: results from the Women’s Health Initiative Memory Study. J Am Heart Assoc 2013;2:e000369.

134. Soares CN, Maki PM. Menopausal transition, mood, and cognition: an integrated view to close the gaps. Menopause 2010;17:812–4.

135. Greendale GA, Derby CA, Maki PM. Perimenopause and cognition. Obstet Gynecol Clin North Am 2011;38:519–35.

136. Greendale GA, Wight RG, Huang MH, et al. Menopause-associated symptoms and cognitive performance: results from the study of women’s health across the nation. Am J Epidemiol 2010;171:1214–24.

137. Price RW. Neurological complications of HIV infection. Lancet 1996;348:445–52.

138. Antinori A, Arendt G, Becker JT, et al. Updated research nosology for HIV-associated neurocognitive disorders. Neurology 2007;69:1789–99.

139. Gisslén M, Price RW, Nilsson S. The definition of HIV-associated neurocognitive disorders: are we overestimating the real prevalence? BMC Infect Dis 2011;11:356.

140. Rubin LH, Sundermann EE, Cook JA, et al. An investigation of menopausal stage and symptoms on cognition in HIV-infected women. Menopause 2014;21:997–1006.

141. Cook JA, Cohen MH, Burke J, et al. Effects of depressive symptoms and mental health quality of life on use of highly active antiretroviral therapy among HIV-seropositive women. J Acquir Immune Defic Syndr 2002;30:401–9.

142. Cook JA, Grey D, Burke J, et al. Depressive symptoms and AIDS-related mortality among a multisite cohort of HIV-positive women. Am J Pub Health 2004;94:1133–40.

143. Kim SC, Messing S, Shah K, et al. Effects of highly active antiretroviral therapy (HAART) and menopause on risk of progression of cervical dysplasia in human immune deficiency virus (HIV) infected women. Infect Dis Obstet Gynecol 2013;2013:784718.

144. Ceccaldi PF, Ferreira C, Coussy F, et al. Cervical disease in postmenopausal HIV-1 infected women. J Gynecol Obstet Biol Reprod 2010;39:466–70.

145. European Study Group on Heterosexual Transmission of HIV. Comparison of female to male and male to female transmission of HIV in 563 stable couples. BMJ 1992;304:809–13.

146. Meditz AL, Moreau KL, MaWhinney S, et al. CCR5 expression is elevated on endocervical CD4+ T cells in healthy postmenopausal women. J Acquir Immune Defic Syndr 2012;59:221–8.

147. Chappell CA, Isaacs CE, Xu W, et al. The effect of menopause on the innate antiviral activity of cervicovaginal lavage. Am J Obstet Gynecol 2015;213:204.

148. Melo KC, Melo MR, Ricci BV, Segurado AC. Correlates of human immunodeficiency virus cervicovaginal shedding among postmenopausal and fertile-aged women. Menopause 2012;19:150–6.

149. Viard JP, Mocroft A, Chiesi A, et al. Influence of age of CD4 cell recovery in human immunodeficiency virus-infected patients receiving highly active antiretroviral therapy: evidence from the Euro SIDA study. J Infect Dis 2001;193:1290–4.

150. Grabar S, Kousignian I, Sobel A, et al. Immunological and clinical responses to highly active antiretroviral therapy over 50 years of age. Results from the French Hospital Database on HIV. AIDS 2004;18:2029–38.

151. Cuzin L, Delpierre C, Gerard S, et al. Immunologic and clinical responses to highly active antiretroviral therapy in patients with HIV infection aged >50 years. Clin Infect Dis 2007;45:654–7.

152. Patterson KB, Cohn SE, Uynik J, et al. Treatment responses in antiretroviral treatment-naïve premenopausal and postmenopausal HIV-1 infected women: an analysis from AIDS clinical trials group studies. Clin Infect Dis 2009;49:473–6.

153. van Benthem BH, Vernazza P, Coutinho RA, et al. The impact of pregnancy and menopause on CD4 lymphocyte count in HIV-infected women. AIDS 2002;16:919–24.

Issue
Journal of Clinical Outcomes Management - January 2016, VOL. 23, NO. 1
Issue
Journal of Clinical Outcomes Management - January 2016, VOL. 23, NO. 1
Publications
Publications
Topics
Article Type
Display Headline
Menopause in HIV-Infected Women
Display Headline
Menopause in HIV-Infected Women
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default