Skip to main content
Advertisement
Browse Subject Areas
?

Click through the PLOS taxonomy to find articles in your field.

For more information about PLOS Subject Areas, click here.

  • Loading metrics

Low bone mass in people living with HIV on long-term anti-retroviral therapy: A single center study in Uganda

  • Erisa Sabakaki Mwaka ,

    Roles Conceptualization, Data curation, Formal analysis, Funding acquisition, Investigation, Methodology, Validation, Writing – original draft, Writing – review & editing

    erisamwaka@gmail.com

    Affiliations School of Biomedical Sciences, College of Health Sciences, Makerere University, Kampala, Uganda, Infectious Diseases Institute, College of Health Sciences, Makerere University, Kampala, Uganda

  • Ian Guyton Munabi,

    Roles Conceptualization, Data curation, Formal analysis, Methodology, Writing – review & editing

    Affiliation School of Biomedical Sciences, College of Health Sciences, Makerere University, Kampala, Uganda

  • Barbara Castelnuovo,

    Roles Data curation, Formal analysis, Methodology, Supervision, Writing – review & editing

    Affiliation Infectious Diseases Institute, College of Health Sciences, Makerere University, Kampala, Uganda

  • Arvind Kaimal,

    Roles Data curation, Investigation, Methodology, Writing – review & editing

    Affiliation Infectious Diseases Institute, College of Health Sciences, Makerere University, Kampala, Uganda

  • William Kasozi,

    Roles Data curation, Investigation, Methodology, Writing – review & editing

    Affiliation Infectious Diseases Institute, College of Health Sciences, Makerere University, Kampala, Uganda

  • Andrew Kambugu,

    Roles Conceptualization, Project administration, Resources, Supervision, Writing – review & editing

    Affiliation School of Biomedical Sciences, College of Health Sciences, Makerere University, Kampala, Uganda

  • Philippa Musoke,

    Roles Formal analysis, Investigation, Methodology, Supervision, Writing – review & editing

    Affiliation Department of Paediatrics and Child Health, School of Medicine, College of Health Sciences, Makerere University, Kampala, Uganda

  • Elly Katabira

    Roles Conceptualization, Formal analysis, Supervision, Writing – review & editing

    Affiliation Department of Medicine, School of Medicine, College of Health Sciences, Makerere University, Kampala, Uganda

Abstract

Background

This study set out to determine the prevalence of low bone mass following long-term exposure to antiretroviral therapy in Ugandan people living with HIV.

Methods

A cross-sectional study was conducted among 199 people living with HIV that had been on anti-retroviral therapy for at least 10 years. All participants had dual X-ray absorptiometry to determine their bone mineral density. The data collected included antiretroviral drug history and behavioral risk data Descriptive statistics were used to summarize the data. Inferential statistics were analyzed using multilevel binomial longitudinal Markov chain Monte Carlo mixed multivariate regression modelling using the rstanarm package.

Results

One hundred ninety nine adults were enrolled with equal representation of males and females. The mean age was 39.5 (SD 8.5) years. Mean durations on anti-retroviral treatment was 12.1 (SD 1.44) years, CD4 cell count was 563.9 cells/mm3. 178 (89.5%) had viral suppression with <50 viral copies/ml. There were 4 (2.0%) and 36 (18%) participants with low bone mass of the hip and lumbar spine respectively. Each unit increase in body mass index was associated with a significant reduction in the odds for low bone mineral density of the hip and lumbar spine. The duration on and exposure to the various antiretroviral medications had no significant effect on the participant’s odds for developing low bone mass. All the coefficients of the variables in a multivariable model for either hip or lumbar spine bone mass were not significant.

Conclusion

These results provide additional evidence that patients on long term ART achieve bone mass stabilization. Maintaining adequate body weight is important in maintaining good bone health in people on antiretroviral therapy.

Introduction

Globally, nearly 70% of the people living with the human immunodeficiency virus (HIV), which is an estimated 37.9 million people, are in Sub-Saharan Africa (SSA) [1]. As of June 2019, 24.5 million people were accessing antiretroviral therapy and currently the World Health Organization (WHO) recommends that all HIV infected individuals should be initiated on ART upon diagnosis [2]. The wide spread use of antiretroviral therapy (ART) to treat HIV infection has led to increased lifespans of HIV-infected persons and a marked reduction in AIDS associated complications but created additional health challenges related to aging with HIV [3,4].

The burden of non-communicable diseases (NCDs) in sub-Saharan Africa (SSA) is higher than the global average; and should therefore be prioritized on the health and development agenda [5]. Non-communicable diseases are becoming a leading cause of morbidity and mortality also in the HIV-infected populations especially those on long-term anti-retroviral therapy (ART) [6]. Long-term use of ART has been associated with several adverse effects on bone mineral density (BMD) that call for continual monitoring, especially now that a sizable proportion of Ugandan people living with HIV (PLWH) have been on ART for long durations. It is important to note that 67% of adults living with HIV are accessing ART in eastern and southern Africa [1] with a sizable proportion of these being on ART for over a decade [6], yet relatively little is known about the impact of ART on bone health.

The prevalence of low BMD in PLWH and ART-treated individuals is reported to be twice that in health individuals [7]. The prevalence of osteopenia and osteoporosis in PLWH ranges from 48% to 55% and 10% to 34% respectively [8,9] with HIV-infected individuals having a 6.3 times greater chance of developing low BMD when compared to the HIV-uninfected, and HIV infected individuals on ART-treated having a 2.5 times greater risk of developing low BMD compared to the ART-naïve individuals [3]. Results form a meta-analysis [7] showed that HIV infected individuals had twice the risk of developing hip and lumbar spine low BMD (odds ratio 2.4 and 2.6 respectively), compared to the HIV-uninfected. In the same meta-analysis, the odds of developing low BMD at the hip and lumbar spine was 2.8 times (p = 0.004) and 3.4 times (p = 0.0002) respectively when compared to ART-naïve individuals.

Generally, BMD is higher among PLWH in low and middle-income countries (LMIC) compared to resource rich settings with prevalence rates as high as 85% [1014]. Data from developed countries consistently show that HIV infection is associated with low BMD and an increased fracture risk, however, there is limited published data from resource limited settings (RLS) on longitudinal changes in BMD, the effect of ART regimens on BMD and almost no data on fragility fractures among HIV infected individuals. This could be attributed to bone health not being a priority in these settings, limited availability of dual energy X-ray absorptiometry (DXA) machines and the prohibitive cost of DXA scanning in most SSA countries [14].

The risk factors for low BMD in the developed world remain similar to those in RLS [8,15,16]. However, some of these risk factors such as low body mass index (BMI), malnutrition, advanced disease, longer duration of disease and a higher viral load are more prevalent in RLS [10,11,17,18]. The prevalence of low BMD in the Ugandan population is not known. Furthermore, there are no local guidelines for the diagnosis and management of osteoporosis in the country. This study therefore set out to determine the prevalence and predictors of low BMD following long-term exposure to ART in Ugandan PLWH.

Much as many studies with relatively young populations [8,9,17,19] use the World Health Organization criteria for categorizing bone loss, in this study we used the International Society of Clinical Densitometry (ISCD) official position that recommends the use of “Bone mass” instead of “Bone mineral density” for pre-menopausal females, males younger than 50 years and children [20].

Materials and methods

Study design and setting

This was a cross-sectional study conducted over an 18 month period in a cohort of PLWH receiving treatment from the Makerere University Infectious Diseases Institutes (IDI) HIV care clinics in Kampala, the capital city of Uganda. The IDI is a Center of Excellence for HIVAIDS research and service delivery in Uganda [21]. The clinic opens 10 hours a day, 5 days a week and currently supports the care for over 8,000 HIV/AIDS clients.

The study population comprised of adult PLWH, who are part of the IDI Antiretroviral Treatment Long-term (ALT) cohort, which has been enrolling patients receiving ART consecutively for at least 10 years and plan to follow them up for another 10 years. The ALT cohort currently comprises 1000 individuals who have been undergoing annual evaluations for more than 10 years. Details of the ALT cohort have been published and described elsewhere [22]. Using randomly generated computer numbers we randomly enrolled adults above 18 years who had been receiving ART for at least 10 years and were willing and able to comply with all study procedures. We excluded clients who were hospitalized or bedridden, enrolled in clinical trials, those with pathological fractures, history of trauma or surgery at the measurement site and those that were unwilling or unable to comply with any part of the study protocol. One hundred ninety nine PLWH were enrolled in the study (Fig 1).

The sample size was calculated using the www.openepi.com online proportions sample size calculator for: an 80.4% prevalence of low BMD among HIV positive persons on ART [11], population size of 1000 HIV positive clients on ART, confidence limits of 5% and design effect of 1.3 arising from the multiple medications for each client at any one time to give a calculated sample size of 190 respondents for a power of 90. To this was included a 5% allowance for loss and omissions, binging the final sample size to 200 respondents.

Participants were consecutively recruited over the 18-month study period. Clients were invited to participate and enrolled during their routine annual clinic visit.

Study procedures

All participants were subjected to lumbar spine, left total hip and left femoral neck body scans, to assess body mineral content and bone mineral density (BMD), using dual energy X-ray absorptiometry (DXA) (Hologic Discovery Wi Apex 3.1, Hologic Bedford Inc., Bedford, MA, USA) using standard protocols. Bone mineral density was expressed in grams of mineral per square centimetre. The reference population for this scanning was age, sex, race and BMI matched from the National Health and Nutrition Examination Survey (NHANES) cohort [23]. Bone mass was categorized using the official position of the International Society of Clinical Densitometry (ISCD) since most participants were below 50 years of age [20]. The ISCD official position recommends the use of the Z-score for pre-menopausal females, males younger than 50 years and children. A Z-score of -2.0 or lower was defined as having low bone mass for age and a Z-score above -2.0 was defined as bone mass within the expected range for age [20]. The World Health Organization (WHO) was used for post-menopausal females and males above 50 years of age [24,25]. A T-score that was within one standard deviation (SD) of the reference BMD was classified as normal, 1 to 2.5 SD below the reference BMD as osteopenia, and greater than 2.5 SD below the reference BMD as osteoporosis. For this study all participants with low bone mass or osteopenia/osteoporosis were categorized as having “low bone mass” while the rest were categorized as having normal bone mass (BM). This was done for ease of data analysis. The following data was also collected at enrolment: age, sex, height and weight for use in calculating Body Mass Index (BMI), the type of ART (categorized as Nucleoside reverse transcriptase inhibitors, Non-nucleoside reverse transcriptase inhibitors, protease inhibitors, and Integrase inhibitors), duration on specific antiretroviral agents, behavioural risk information including alcohol and tobacco use, and history of previous fractures of the wrist, hip or spine, using a study questionnaire and data abstraction tool.

Data collection and statistical analysis

Data were collected initially collected using case report forms and then entered into DataFax forms that were specially designed for this study. This data management system is designated to manage paper data forms. The forms are faxed to the DataFax server where they are read using intelligent characters-recognition and then added to the study database. The latest viral load results and nadir CD4 count were retrieved from the Integrated Clinic Enterprise Application (ICEA) database, an in-house software that provides good quality data collection, with minimal missing or incorrect information [26]. These validated records were then exported as excel files for transformation so that each row represented an individual drug exposure prior to further analysis in the R-statistical computing environment [27]. The final long dataset comprised of 1384 anti-retroviral drug exposures. The analysis was informed by the study hypothesis that: exposure (time on treatment and type of drugs) to ART was associated with differences in BM among respondents after controlling for age, alcohol consumption, sex, smoking, most recent viral load, CD4 cell count, line of treatment (switch) and body mass index. During analysis descriptive statistics were generated and summarised as frequencies and correlations in tables. Inferential statistics were generated with multilevel binomial longitudinal Markov chain Monte Carlo (MCMC) mixed multivariate regression modelling using the rstanarm package [2830]. The diagnostics for each model, one for the spine and other for hip BM, were used to identify any poorly performing values and assess the sampling quality. The absence of a value of “zero” in the 95% confidence interval for the coefficients was used to identify statistically significant output values. The mean coefficients were summarised in tables but exponentiated to obtain odds ratios. All records with missing data were removed from the database prior to the inferential MCMC part of the analysis. Ethics approval was obtained from the Makerere University School of Biomedical Sciences Higher Degrees and Research Ethics Committee (SBSHDREC 410) and the Uganda National Council of Science and Technology (HS 2179). All participants provided informed consent to participate in the study.

Results

One hundred ninety nine PLWH were recruited between July 2017 and July 2018. Fewer participants (4/199, 2%) had left hip low bone mass compared to the lumbar spine (36/199, 18%) as shown in Table 1. There was equal representation of both males (50%) and females (50%). The mean age was 39.5 (SD 8.5) years and female participants were on average younger than male participants by 1.38 years (standard error = 1.21, t-value = -1.14, P-value = 0.25). At the time of recruitment, participants had on average been living with HIV for 12.6 (SD 1.55) years since diagnosis and a mean of 12.1 (SD 1.44) years on ART. One hundred seventy eight (89.5%) of the participants had viral suppression with <50 viral copies/ml. The mean CD4 cell count was 564 cells/mm3 (SD 20.4, CI 524–604).

thumbnail
Table 1. Descriptive statistics for the study population.

https://doi.org/10.1371/journal.pone.0246389.t001

The correlations between the study variables as summarized in Table 2, were moderate to weak. There was moderate correlation for observed low BM between the hip and the lumbar spine (R = 0.30, P-value = <0.01).

thumbnail
Table 2. Showing the correlations between the study variables.

https://doi.org/10.1371/journal.pone.0246389.t002

Fig 2 provides a summary of the different ART drug classes in the various prescriptions for each participant, from the time of initiation on ART till recruitment into the study. From this table non-reverse transcriptase inhibitors (NRTI) were the most commonly prescribed medications.

thumbnail
Fig 2. ART treatment at each treatment switch.

Nucleoside reverse transcriptase inhibitors (NRTI), Non-nucleoside reverse transcriptase inhibitors (NNRTI), protease inhibitors (PI), and Integrase inhibitors (II).

https://doi.org/10.1371/journal.pone.0246389.g002

There were three incomplete participants records 4/199 (2%), which were excluded, leaving 196 complete respondents’ records for further analysis.

Table 3 provides a summary of the best-fitted univariable and multivariable MCMC models for BMI determined using hip BM and lumbar spine BM. Overall on the univariable analysis, each unit change in the classification of BMI was associated with a large reduction in the odds for low BM. This was significant for both the hip (OR = 0.06, 95% CI 0.01 to 0.47) and lumbar spine (OR = 0.004, 95% CI 0.0002 to 0.07). Also, in this table note that, for both hip (OR = 70.11, 95% CI 1.75 to 3,071) and lumbar spine (OR = 14,185, 95% CI 20.70 to 18,255,921), participants who were underweight were several times more likely to have a low BM compared to the normal weight participants. On the other hand, participants who were overweight were less likely to have low BM. This was significant for the lumbar spine observations (OR = 0.002, 95% CI <0.01 to 0.29). Also, either being female or each unit increase in age was associated with significantly lower odds of low BM on univariable modeling. All the coefficients for the variables of the best multivariable model for both hip and lumbar spine BM were not significant. As shown in Table 3, for both the univariable and multivariable modeling, the duration on and exposure to the various ART medications had no significant effect on the participant’s odds for developing low BM. For the multivariable model interactions in Table 3, it is important to note that the interaction between being female and having ever smoked was associated with significantly lower odds for observed low BM affecting the lumbar spine (OR<0.01, 95% CI <0.01 to 0.04).

Discussion

In this cross-sectional study we observed that the duration of exposure to ART medication and ART classes have no significant effect on participants’ risk for developing low BM and that high BMI is associated with reduced risk of developing low BMD in PLWH on long-term ART. People living with HIV are at increased risk of developing low BM when compared to age-matched HIV uninfected controls [34,35]. In this study, there were more cases of low BM as defined by the lumbar spine than the hip by a factor of 9. The prevalence of low BM was 18.1%. The prevalence of low BM in our study is consistent with what is reported in literature [8,9,17,19]. It is important to note that most longitudinal studies reporting on bone health in PLWH on long term ART use the WHO criteria for categorizing BMD instead of the ISCD official position that recommend the use of BM instead of BMD; despite having relatively young populations.

The ISCD also asserts that osteoporosis cannot be diagnosed in men under 50 years of age using BMD alone [20]. Therefore in the absence of local national guidance on the diagnosis and management of osteoporosis, we considered the official position of the International Society of Clinical Densitometry (ISCD) to categorize participants as having low BM or normal bone, since most participants were below 50 years of age [20]. However, the WHO criteria were also used to categorize post-menopausal females and males above 50 years. For ease of analysis, those categorized as osteopenia or osteoporosis were grouped together and considered as having low BM. The official position of International osteoporosis foundation (IOF) recommends BMD measurements at both the lumbar spine and the hip, and to use the lowest BMD recorded [3639]. The IOF official position further recommends that lumbar spine BMD be used for monitoring response to treatment while hip BMD is considered the best predictor for hip fractures [40].

The prevalence of low BM in our study is consistent with what was previously reported in a study conducted at our institution to investigate low BMD among patients failing first-line ART, though the mean duration on antiretroviral medication at the time was only 3.7 years [17]. With a mean ART experience of more than 12 years, we believe that our participants had stable BM. Antiretroviral therapy has been shown to significantly contribute to low BM irrespective of classes [9,10,14,4143]; although this accelerated bone loss has been mainly associated with tenofovir disoproxil fumarate (TDF) containing regimens compared to non-TDF-containing regimens [14,4345]. This initial bone loss is transient, possibly due to increased bone catabolism after viral load suppression and immune reconstitution [4649], and has been shown to be most marked in the first two years of ART initiation with about 2–6% loss in BMD [50] followed by a long period of stable BMD or increase [51,52]. These changes in BMD over time have been shown to be comparable to those in uninfected health controls [51]. The pathogenesis of increased bone turn over in HIV infected individuals is rather complex and multifactorial [7]. HIV infection per se increases inflammatory cytokines that are thought to increase bone turn-over through the stimulation of osteoclast and bone resorption [7,53]. The mechanism of action of anti-retroviral drugs on low BM remains controversial [3,12]. Evidence from longitudinal data shows that among the different antiretroviral drugs, tenofovir is associated with bone loss [45,5458] and this has been attributed to increased bone turnover. The exact mechanism by which TDF reduces bone mass is yet to be elucidated but, some studies have linked it to proximal renal tubular dysfunction [5962] with possible resultant impairment in Vitamin D metabolism [59,61,63,64]. Protease inhibitors have been reported to suppress osteoclast function [65]. Several cross-sectional studies from LMIC have shown an association between non-nucleoside reverse transcriptase inhibitors (NNRTIs) particularly Efavirenz, which is widely used to treat HIV in LMIC, and low 25-hydroxyvitamin D [6669]. Low levels of Vitamin D have been associated with a higher risk of HIV disease progression, death and virologic failure after ART [7072]. Sub-optimal Vitamin D levels and cumulative exposure of tenofovir and protease inhibitors like lopinavir/ritonavir (LVP/RTV) have also been reported to independently increase the risk of low BMD [72,73] and fragility fractures [74]. TDF has been associated with a bigger decline in BMD than stavudine [55] or abacavir [45].

In the setting of long term viral suppression, understanding the complications of HIV infection and ART is paramount to successful management of HIV and its associated complications like low Bone mineral density [9]. Our findings further suggest that the duration of exposure to the various ART classes, is not associated with low BM; which is consistent with other studies which have reported stable BMD in PLWH on long-term ART [44,51,52,7578], suggesting a long-term benefit of antiretroviral therapy. Bolland et. al [52] reported stable BMD in a cohort of 44 HIV infected middle-aged men on ART who were followed up for up to12 years. They further reported a BMD increase of 6.9% at the lumbar spine; and actually argued that it was not necessary to follow up men established on ART who do not have risk factors for BMD loss. Important to note, is the 5% mean increase in weight over the 12 years in their cohort. Increase in weight reduces the odds of developing low BMD, which is consistent with what was observed in our study. Bone loss is influenced by several factors including body weight, age, physical activity level, diet to mention a few [79]. Low BMI is a well-documented risk factor for osteoporosis and hip fractures in both men and women [10,80]. Weight loss is associated with 1% to 2% bone loss at the hip [8183] and has been associated with bone mobilization and decrease in bone mineral content and BMD [8486]. Therefore it is important for on ART to maintain adequate weight if they are to maintain good bone health.

In the setting of long term viral suppression, understanding the complications of HIV infection and ART is key to successful management of HIV and its associated complications [9]. We hope our results will raise awareness on bone health among clinicians and contribute to appropriate planning and management of low BMD among PLWH with the ultimate goal of making significant contribution towards reducing the NCD burden among this population.

Strength of the study

The study was conducted among a well-characterized and organized cohort that had been consistently followed up every six months since 2004 and there was equal representation of both males and females. Secondly, all BMD scans were performed using the same machine and by the same technicians. Lastly, we utilized both Z-score and T-scores to diagnose low BMD in participants < 50 years and those above 50 years respectively as recommended by WHO [24,25].

Limitations

This was a cross-sectional study conducted at one site; therefore, the study findings might not be generalizable to PLWH in the general population. Secondly, our results are based on only one DXA scan, participants had uniform exposure to ART and there was no control group; thus making it difficult to make any inferences on the effect of individual anti-retroviral drugs and regimens on bone loss.

Conclusion

These results provide additional evidence that patients on long term ART achieve BM stabilization. It is important for PLWH on ART to maintain adequate weight if they are to maintain good bone health. There is need to develop local guidelines and protocols for the prevention and treatment of low bone mass not only in HIV, but also in the general public.

Acknowledgments

We are very grateful to our research participants for their voluntary participation in this study.

References

  1. 1. HIV/AIDS JUNPo. Fact Sheet—Latest Global and Regional Statistics on the Status of the HIV Epidemic. 2019.
  2. 2. Consolidated guidelines on the use of antiretroviral drugs for treating and preventing HIV infection: Recommendations for a public health approach.: World Health Organization; 2016 [Second:[https://www.who.int/hiv/pub/arv/arv-2016/en/.
  3. 3. Brown TT, Qaqish RB. Antiretroviral therapy and the prevalence of osteopenia and osteoporosis: a meta-analytic review. Aids. 2006;20(17):2165–74. pmid:17086056
  4. 4. Battalora LA, Young B, Overton ET. Bones, fractures, antiretroviral therapy and HIV. Current infectious disease reports. 2014;16(2):393. pmid:24535243
  5. 5. Gouda HN, Charlson F, Sorsdahl K, Ahmadzada S, Ferrari AJ, Erskine H, et al. Burden of non-communicable diseases in sub-Saharan Africa, 1990–2017: results from the Global Burden of Disease Study 2017. The Lancet Global Health. 2019;7(10):e1375–e87. pmid:31537368
  6. 6. Kiragga AN, Mubiru F, Kambugu AD, Kamya MR, Castelnuovo B. A decade of antiretroviral therapy in Uganda: what are the emerging causes of death? BMC infectious diseases. 2019;19(1):77. pmid:30665434
  7. 7. Goh S, Lai PSM, Tan ATB, Ponnampalavanar S. Reduced bone mineral density in human immunodeficiency virus-infected individuals: a meta-analysis of its prevalence and risk factors. Osteoporosis International. 2018;29(3):595–613. pmid:29159533
  8. 8. Bonjoch A, Figueras M, Estany C, Perez-Alvarez N, Rosales J, del Rio L, et al. High prevalence of and progression to low bone mineral density in HIV-infected patients: a longitudinal cohort study. Aids. 2010;24(18):2827–33. pmid:21045635
  9. 9. Escota GV, Mondy K, Bush T, Conley L, Brooks JT, Önen N, et al. High Prevalence of Low Bone Mineral Density and Substantial Bone Loss over 4 Years Among HIV-Infected Persons in the Era of Modern Antiretroviral Therapy. AIDS research and human retroviruses. 2016;32(1):59–67. pmid:26366785
  10. 10. Alonge T, Okoje-Adesomoju V, Atalabi O, Obamuyide H, Olaleye D, Adewole I. Prevalence of abnormal bone mineral density in HIV-positive patients in Ibadan, Nigeria. Journal of the West African College of Surgeons. 2013;3(4):1. pmid:26046022
  11. 11. Dravid A, Kulkarni M, Borkar A, Dhande S. Prevalence of low bone mineral density among HIV patients on long-term suppressive antiretroviral therapy in resource limited setting of western India. Journal of the International AIDS Society. 2014;17(4). pmid:25394074
  12. 12. Aydın OA, Karaosmanoglu HK, Karahasanoglu R, Tahmaz M, Nazlıcan O. Prevalence and risk factors of osteopenia/osteoporosis in Turkish HIV/AIDS patients. The Brazilian Journal of Infectious Diseases. 2013;17(6):707–11. pmid:24076108
  13. 13. Neto LFP, Ragi-Eis S, Vieira NF, Soprani M, Neves MB, Ribeiro-Rodrigues R, et al. Low bone mass prevalence, therapy type, and clinical risk factors in an HIV-infected Brazilian population. Journal of Clinical Densitometry. 2011;14(4):434–9. pmid:22051092
  14. 14. Matovu FK, Wattanachanya L, Beksinska M, Pettifor JM, Ruxrungtham K. Bone health and HIV in resource-limited settings: a scoping review. Current Opinion in HIV and AIDS. 2016;11(3):306–25. pmid:27023284
  15. 15. Yin MT, Overton ET. Increasing clarity on bone loss associated with antiretroviral initiation. Journal of Infectious Diseases. 2011;203(12):1705–7.
  16. 16. Kim H-S, Chin BS, Shin H-S. Prevalence and risk factors of low bone mineral density in Korean HIV-infected patients: impact of abacavir and zidovudine. Journal of Korean medical science. 2013;28(6):827–32. pmid:23772145
  17. 17. Wandera B, Kiragga A, Semitala F, Ellis J, Colebunders R, Paton N, et al. Low Bone Mineral Density among Ugandan HIV infected patients on failing first line Antiretroviral Therapy; a sub-study of the EARNEST trial. Proceedings of the 21st Conference on Retroviruses and Opportunistic Infections Boston, MA March 3–6, 2014 Boston, MA: National AIDS Treatment Advocavy project; 2014 [http://www.natap.org/2014/CROI/croi_65.htm.
  18. 18. Masyeni S, Utama S, Somia A, Widiana R, Merati TP. Factors influencing bone mineral density in ARV-naive patients at Sanglah Hospital, Bali. Acta Med Indones. 2013;45(3):175–9. pmid:24045386
  19. 19. Chisati EM, Constantinou D, Lampiao F. Reduced bone mineral density among HIV infected patients on anti-retroviral therapy in Blantyre, Malawi: Prevalence and associated factors. PloS one. 2020;15(1):e0227893. pmid:31935270
  20. 20. 2019 ISCD Official Positions: Adult: International Society for Clinical Densitometry; 2019 [https://www.iscd.org/official-positions/2019-iscd-official-positions-adult/.
  21. 21. Nwaka S, Ochem A, Besson D, Ramirez B, Fakorede F, Botros S, et al. Analysis of pan-African Centres of excellence in health innovation highlights opportunities and challenges for local innovation and financing in the continent. BMC international health and human rights. 2012;12(1):11. pmid:22838941
  22. 22. Castelnuovo B, Kiragga A, Musaazi J, Sempa J, Mubiru F, Wanyama J, et al. Outcomes in a cohort of patients started on antiretroviral treatment and followed up for a decade in an urban clinic in Uganda. PloS one. 2015;10(12):e0142722. pmid:26642214
  23. 23. National Health and Nutrition Examination Survey (NHANES) [https://www.cdc.gov/nchs/nhanes/index.htm.
  24. 24. Czerwiński E, Badurski J, Marcinowska-Suchowierska E, Osieleniec J. Current understanding of osteoporosis according to the position of the World Health Organization (WHO) and International Osteoporosis Foundation. Ortopedia, traumatologia, rehabilitacja. 2006;9(4):337–56.
  25. 25. WHO, editor World Health Organisation (WHO) scientific group on the assessment of osteoporosis at primary health care level. Summary meeting report; 2004.
  26. 26. Castelnuovo B, Kiragga A, Afayo V, Ncube M, Orama R, Magero S, et al. Implementation of provider-based electronic medical records and improvement of the quality of data in a large HIV program in Sub-Saharan Africa. PloS one. 2012;7(12):e51631. pmid:23284728
  27. 27. Fox J, Andersen R. Using the R statistical computing environment to teach social statistics courses. Department of Sociology, McMaster University. 2005:2–4.
  28. 28. Goodrich B, Gabry J, Ali I, Brilleman S. rstanarm: Bayesian applied regression modeling via Stan. 2018.
  29. 29. Bürkner P-C. Advanced Bayesian Multilevel Modeling with the R Package brms. The R Journal. 2018;10(1):395–411.
  30. 30. Bürkner P-C. brms: An R Package for Bayesian Multilevel Models Using Stan. Journal of Statistical Software. 2017;80(1):1–28.
  31. 31. Zhou B-F, China CM-AGotWGoOi. Predictive values of body mass index and waist circumference for risk factors of certain related diseases in Chinese adults—study on optimal cut-off points of body mass index and waist circumference in Chinese adults. Biomedical and environmental sciences: BES. 2002;15(1):83. pmid:12046553
  32. 32. Ogunlade O, Adalumo O, Asafa M. Challenges of body mass index classification: New criteria for young adult Nigerians. Nigerian Journal of Health Sciences. 2015;15(2):71.
  33. 33. Rahman M, Berenson AB. Accuracy of current body mass index obesity classification for white, black and Hispanic reproductive-age women. Obstetrics and gynecology. 2010;115(5):982. pmid:20410772
  34. 34. Fausto A, Bongiovanni M, Cicconi P, Menicagli L, Ligabò EV, Melzi S, et al. Potential predictive factors of osteoporosis in HIV-positive subjects. Bone. 2006;38(6):893–7. pmid:16377270
  35. 35. Grund B, Peng G, Gibert CL, Hoy JF, Isaksson RL, Shlay JC, et al. Continuous antiretroviral therapy decreases bone mineral density. AIDS (London, England). 2009;23(12):1519. pmid:19531929
  36. 36. International OSteoporosis Foundation: Official positions Adults 2019 [https://iscd.app.box.com/s/5r713cfzvf4gr28q7zdccg2i7169fv86.
  37. 37. Leib ES, Lewiecki EM, Binkley N, Hamdy RC. Official positions of the international society for clinical densitometry. Journal of Clinical Densitometry. 2004;7(1):1–5. pmid:14742881
  38. 38. Binkley N, Bilezikian JP, Kendler DL, Leib ES, Lewiecki EM, Petak SM. Official positions of the International Society for Clinical Densitometry and executive summary of the 2005 Position Development Conference. Journal of Clinical Densitometry. 2006;9(1):4–14. pmid:16731426
  39. 39. Kanis J, Cooper C, Rizzoli R, Reginster J-Y. European guidance for the diagnosis and management of osteoporosis in postmenopausal women. Osteoporosis International. 2019;30(1):3–44. pmid:30324412
  40. 40. Sheu A, Diamond T. Diagnostic tests: bone mineral density: testing for osteoporosis. Australian prescriber. 2016;39(2):35. pmid:27340320
  41. 41. Mirembe BG, Kelly CW, Mgodi N, Greenspan S, Dai JY, Mayo A, et al. Bone mineral density changes among young, healthy African women receiving oral tenofovir for HIV preexposure prophylaxis. Journal of acquired immune deficiency syndromes (1999). 2016;71(3):287. pmid:26866954
  42. 42. Collins BN, Levin KP, Bryant-Stephens T. Pediatricians’ practices and attitudes about environmental tobacco smoke and parental smoking. The Journal of pediatrics. 2007;150(5):547–52. pmid:17452234
  43. 43. Grant PM, Cotter AG. Tenofovir and bone health. Current Opinion in HIV and AIDS. 2016;11(3):326. pmid:26859637
  44. 44. Brown TT, Moser C, Currier JS, Ribaudo HJ, Rothenberg J, Kelesidis T, et al. Changes in bone mineral density after initiation of antiretroviral treatment with tenofovir disoproxil fumarate/emtricitabine plus atazanavir/ritonavir, darunavir/ritonavir, or raltegravir. The Journal of infectious diseases. 2015;212(8):1241–9. pmid:25948863
  45. 45. McComsey GA, Kitch D, Daar ES, Tierney C, Jahed NC, Tebas P, 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. Journal of Infectious Diseases. 2011;203(12):1791–801. pmid:21606537
  46. 46. Bolland MJ, Grey A, Reid IR. Skeletal health in adults with HIV infection. The lancet Diabetes & endocrinology. 2015;3(1):63–74. pmid:24731663
  47. 47. Hoy JF, Grund B, Roediger M, Schwartz AV, Shepherd J, Avihingsanon A, et al. Immediate initiation of antiretroviral therapy for HIV infection accelerates bone loss relative to deferring therapy: findings from the START bone mineral density substudy, a randomized trial. Journal of bone and mineral research. 2017;32(9):1945–55. pmid:28650589
  48. 48. Grant PM, Kitch D, McComsey GA, Dube MP, Haubrich R, Huang J, et al. Low baseline CD4+ count is associated with greater bone mineral density loss after antiretroviral therapy initiation. Clinical infectious diseases. 2013;57(10):1483–8. pmid:23943825
  49. 49. Ofotokun I, Titanji K, Vikulina T, Roser-Page S, Yamaguchi M, Zayzafoon M, et al. Role of T-cell reconstitution in HIV-1 antiretroviral therapy-induced bone loss. Nature communications. 2015;6(1):1–15. pmid:26392000
  50. 50. Duvivier C, Kolta S, Assoumou L, Ghosn J, Rozenberg S, Murphy RL, et al. Greater decrease in bone mineral density with protease inhibitor regimens compared with nonnucleoside reverse transcriptase inhibitor regimens in HIV-1 infected naive patients. Aids. 2009;23(7):817–24. pmid:19363330
  51. 51. Bolland MJ, Grey A, Horne AM, Briggs SE, Thomas MG, Ellis-Pegler RB, et al. Stable bone mineral density over 6 years in HIV-infected men treated with highly active antiretroviral therapy (HAART). Clinical endocrinology. 2012;76(5):643–8. pmid:22040002
  52. 52. Bolland MJ, Horne AM, Briggs SE, Thomas MG, Reid IR, Gamble GD, et al. Long-Term Stable Bone Mineral Density in HIV-Infected Men Without Risk Factors for Osteoporosis Treated with Antiretroviral Therapy. Calcified tissue international. 2019;105(4):423–9. pmid:31250043
  53. 53. Fakruddin JM, Laurence J. HIV envelope gp120-mediated regulation of osteoclastogenesis via receptor activator of nuclear factor κB ligand (RANKL) secretion and its modulation by certain HIV protease inhibitors through interferon-γ/RANKL cross-talk. Journal of Biological Chemistry. 2003;278(48):48251–8. pmid:12975380
  54. 54. Bernardino JI, Mocroft A, Mallon PW, Wallet C, Gerstoft J, Russell C, et al. Bone mineral density and inflammatory and bone biomarkers after darunavir–ritonavir combined with either raltegravir or tenofovir–emtricitabine in antiretroviral-naive adults with HIV-1: a substudy of the NEAT001/ANRS143 randomised trial. The Lancet HIV. 2015;2(11):e464–e73. pmid:26520926
  55. 55. Gallant JE, Staszewski S, Pozniak AL, DeJesus E, Suleiman JM, Miller MD, et al. Efficacy and safety of tenofovir DF vs stavudine in combination therapy in antiretroviral-naive patients: a 3-year randomized trial. JAMA: the journal of the American Medical Association. 2004;292(2):191–201. pmid:15249568
  56. 56. Stellbrink HJ, Orkin C, Arribas JR, Compston J, Gerstoft J, Van Wijngaerden E, et al. Comparison of changes in bone density and turnover with abacavir-lamivudine versus tenofovir-emtricitabine in HIV-infected adults: 48-week results from the ASSERT study. Clin Infect Dis. 2010;51(8):963–72. pmid:20828304
  57. 57. Assoumou L, Katlama C, Viard J-P, Bentata M, Simon A, Roux C, et al. Changes in bone mineral density over a 2-year period in HIV-1-infected men under combined antiretroviral therapy with osteopenia. AIDS. 2013;27(15):2425–30. pmid:24029735
  58. 58. Taiwo BO, Chan ES, Fichtenbaum CJ, Ribaudo H, Tsibris A, Klingman KL, et al. Less bone loss with maraviroc-compared to tenofovir-containing antiretroviral therapy in the ACTG A5303 Study. Clinical Infectious Diseases. 2015:civ455.
  59. 59. Fux CA, Rauch A, Simcock M, Bucher HC, Hirschel B, Opravil M, et al. Short communication Tenofovir use is associated with an increase in serum alkaline phosphatase in the Swiss HIV Cohort Study. Antiviral therapy. 2008;13:1077–82. pmid:19195333
  60. 60. Kohler JJ, Hosseini SH, Hoying-Brandt A, Green E, Johnson DM, Russ R, et al. Tenofovir renal toxicity targets mitochondria of renal proximal tubules. Laboratory investigation. 2009;89(5):513–9. pmid:19274046
  61. 61. Nelson MR, Katlama C, Montaner JS, Cooper DA, Gazzard B, Clotet B, et al. The safety of tenofovir disoproxil fumarate for the treatment of HIV infection in adults: the first 4 years. Aids. 2007;21(10):1273–81. pmid:17545703
  62. 62. Labarga P, Barreiro P, Martin-Carbonero L, Rodriguez-Novoa S, Solera C, Medrano J, et al. Kidney tubular abnormalities in the absence of impaired glomerular function in HIV patients treated with tenofovir. Aids. 2009;23(6):689–96. pmid:19262355
  63. 63. Brown TT, McComsey GA, King MS, Qaqish RB, Bernstein BM, da Silva BA. Loss of bone mineral density after antiretroviral therapy initiation, independent of antiretroviral regimen. JAIDS Journal of Acquired Immune Deficiency Syndromes. 2009;51(5):554–61. pmid:19512937
  64. 64. Zimmermann AE, Pizzoferrato T, Bedford J, Morris A, Hoffman R, Braden G. Tenofovir-associated acute and chronic kidney disease: a case of multiple drug interactions. Clinical Infectious Diseases. 2006;42(2):283–90. pmid:16355343
  65. 65. Wang MW-H, Wei S, Faccio R, Takeshita S, Tebas P, Powderly WG, et al. The HIV protease inhibitor ritonavir blocks osteoclastogenesis and function by impairing RANKL-induced signaling. The Journal of clinical investigation. 2004;114(2):206–13. pmid:15254587
  66. 66. Steenhoff AP, Schall JI, Samuel J, Seme B, Marape M, Ratshaa B, et al. Vitamin D3 supplementation in Batswana children and adults with HIV: a pilot double blind randomized controlled trial. PloS one. 2015;10(2):e0117123. pmid:25706751
  67. 67. Hidron AI, Hill B, Guest JL, Rimland D. Risk Factors for Vitamin D Deficiency among Veterans with and without HIV Infection. PloS one. 2015;10(4):e0124168. pmid:25898185
  68. 68. Wohl D, Doroana M, Orkin C, Pilotto J, Sungkanuparph S, Yeni P, et al., editors. Change in vitamin D levels smaller and risk of development of severe vitamin D deficiency lower among HIV-1-infected, treatment-naive adults receiving TMC278 compared with EFV: 48-week results from the phase III ECHO trial. 18th Conference on Retroviruses and Opportunistic Infections; 2011.
  69. 69. Wiboonchutikul S, Sungkanuparph S, Kiertiburanakul S, Chailurkit L-O, Charoenyingwattana A, Wangsomboonsiri W, et al. Vitamin D insufficiency and deficiency among HIV-1-infected patients in a tropical setting. Journal of the International Association of Physicians in AIDS Care (JIAPAC). 2012:1545109711432142.
  70. 70. Mansueto P, Seidita A, Vitale G, Gangemi S, Iaria C, Cascio A. Vitamin D Deficiency in HIV Infection: Not Only a Bone Disorder. BioMed research international. 2015;2015.
  71. 71. Havers F, Smeaton L, Gupte N, Detrick B, Bollinger RC, Hakim J, et al. 25-Hydroxyvitamin D insufficiency and deficiency is associated with HIV disease progression and virological failure post-antiretroviral therapy initiation in diverse multinational settings. Journal of Infectious Diseases. 2014;210(2):244–53. pmid:24799602
  72. 72. Dave JA, Cohen K, Micklesfield LK, Maartens G, Levitt NS. Antiretroviral Therapy, Especially Efavirenz, Is Associated with Low Bone Mineral Density in HIV-Infected South Africans. PloS one. 2015;10(12):e0144286. pmid:26633015
  73. 73. Paul T, Asha H, Thomas N, Seshadri M, Rupali P, Abraham O, et al. Hypovitaminosis D and bone mineral density in human immunodeficiency virus-infected men from India, with or without antiretroviral therapy. Endocrine Practice. 2010.
  74. 74. Bedimo R, Maalouf NM, Zhang S, Drechsler H, Tebas P. Osteoporotic fracture risk associated with cumulative exposure to tenofovir and other antiretroviral agents. AIDS. 2012;26(7):825–31. pmid:22301411
  75. 75. Cassetti I, Madruga JVR, Suleiman JMA, Etzel A, Zhong L, Cheng AK, et al. The safety and efficacy of tenofovir df in combination with lamivudine and efavirenz through 6 years in antiretroviral-naïve HIV-1—infected patients. HIV clinical trials. 2007;8(3):164–72. pmid:17621463
  76. 76. Madeddu G, Spanu A, Solinas P, Babudieri S, Calia G, Lovigu C, et al. Different impact of NNRTI and PI-including HAART on bone mineral density loss in HIV-infected patients. Eur Rev Med Pharmacol Sci. 2015;19(23):4576–89. pmid:26698255
  77. 77. Nolan D, Upton R, McKinnon E, John M, James I, Adler B, et al. Stable or increasing bone mineral density in HIV-infected patients treated with nelfinavir or indinavir. Aids. 2001;15(10):1275–80. pmid:11426072
  78. 78. Dolan SE, Kanter JR, Grinspoon S. Longitudinal analysis of bone density in human immunodeficiency virus-infected women. The Journal of Clinical Endocrinology & Metabolism. 2006;91(8):2938–45. pmid:16735489
  79. 79. Palermo A, Tuccinardi D, Defeudis G, Watanabe M, D’Onofrio L, Lauria Pantano A, et al. BMI and BMD: the potential interplay between obesity and bone fragility. International journal of environmental research and public health. 2016;13(6):544. pmid:27240395
  80. 80. De Laet C, Kanis J, Odén A, Johanson H, Johnell O, Delmas P, et al. Body mass index as a predictor of fracture risk: a meta-analysis. Osteoporosis international. 2005;16(11):1330–8. pmid:15928804
  81. 81. Langlois J, Mussolino M, Visser M, Looker A, Harris T, Madans J. Weight loss from maximum body weight among middle-aged and older white women and the risk of hip fracture: the NHANES I epidemiologic follow-up study. Osteoporosis international. 2001;12(9):763–8. pmid:11605743
  82. 82. Ensrud KE, Fullman RL, Barrett-Connor E, Cauley JA, Stefanick ML, Fink HA, et al. Voluntary weight reduction in older men increases hip bone loss: the osteoporotic fractures in men study. The Journal of Clinical Endocrinology & Metabolism. 2005;90(4):1998–2004. pmid:15671096
  83. 83. Bleicher K, Cumming RG, Naganathan V, Travison TG, Sambrook PN, Blyth FM, et al. The role of fat and lean mass in bone loss in older men: findings from the CHAMP study. Bone. 2011;49(6):1299–305. pmid:21925297
  84. 84. Compston J, Laskey M, Croucher P, Coxon A, Kreitzman S. Effect of diet-induced weight loss on total body bone mass. Clinical Science. 1992;82(4):429–32. pmid:1315653
  85. 85. Center JR, White CP. Obesity: bariatric surgery, weight loss and bone. Nature Reviews Endocrinology. 2013;9(11):630. pmid:24080734
  86. 86. Meyer HE, Søgaard AJ, Falch JA, Jørgensen L, Emaus N. Weight change over three decades and the risk of osteoporosis in men: the Norwegian Epidemiological Osteoporosis Studies (NOREPOS). American journal of epidemiology. 2008;168(4):454–60. pmid:18599490