692
Views
0
CrossRef citations to date
0
Altmetric
Review

HIV/AIDS and bone: an unrecognised threat?

, , &
Pages 326-329 | Received 03 Oct 2013, Accepted 13 Jan 2014, Published online: 10 Feb 2015

Abstract

Since the first isolated cases of acquired immunodeficiency syndrome (AIDS) was described in homosexual men in the United States in 1981, the Human Immunodeficiency Virus (HIV) infection has spread globally affecting 33 million people. With the advent of highly active antiretroviral therapy (HAART) there has been a paradigm shift in the clinical profile of patients with HIV. Prior to effective HAART, patients usually had a progressive fatal course characterised by recurrent opportunistic infections. Since the introduction of HAART, which has dramatically improved these patients survival, conditions associated with accelerated aging, have come to the fore, especially cardiovascular disease, malignancy, diabetes mellitus and metabolic bone disease. The aim of this review is to increase the knowledge of HIV associated metabolic bone disease, alert health care providers of the possible bone complications associated with HIV and HAART and stimulate further research in the field.

Introduction

The human immunodeficiency virus (HIV) infection has spread globally, affecting 33 million people since the first isolated cases of acquired immunodeficiency syndrome (AIDS) were described in homosexual men in the USA in 1981.Citation1 It is estimated to have lead to 35 million deaths. The brunt of the pandemic is concentrated in sub-Saharan Africa, with 22.5 million people being infected and a seroprevalence of 5%.Citation1

With the advent of highly active antiretroviral therapy (HAART), there has been a shift in the clinical profile of patients with HIV. Prior to effective HAART, patients usually had a progressive course, characterised by recurrent opportunistic infections, eventually culminating in death. Since the introduction of HAART, which has dramatically improved patient survival, non-infective complications have come to the fore, especially cardiovascular disease, malignancy, diabetes mellitus and metabolic bone disease.Citation2–5 The latter has been neglected in terms of research and management in South Africa. This is evident from the lack of sufficient South African data on HIV/AIDS and metabolic bone disease. Additionally, the South African guidelines for the management of HIV do not incorporate the possibility of bone disease.Citation6 As a result, metabolic bone disease and fractures are greatly overlooked and untreated in HIV patients.

Metabolic bone disease associated with HIV/AIDS results from a complex interplay of a host of different factors, including HIV infection itself, HAART and vitamin D deficiency.Citation7–9 The bone disease that is observed can usually be divided into three main categories; osteoporosis, osteomalacia, or a combination of the two. Osteoporosis is characterised by decreased bone mineral density (BMD), changes in bone microarchitecture, decreased bone strength and an increased risk of fragility fractures.Citation10 Fractures of the hip, spine and distal radius are the most common in osteoporosis.Citation10 Osteomalacia is a disorder characterised by defective mineralisation, the accumulation of unmineralised bone matrix and an increase in osteoid thickness. Vitamin D deficiency, primary or secondary, is the most common cause of osteomalacia. Primary vitamin D deficiency occurs mainly because of decreased sun exposure, and to a lesser extent, decreased dietary vitamin D. Secondary causes of vitamin D deficiency include partial gastrectomy, chronic renal failure and anticonvulsants, for example, phenobarbitone.

HIV/AIDS and metabolic bone disease

Definite increases in the prevalence of osteopaenia and osteoporosis have been described in patients suffering from HIV/AIDS, especially in the Northern Hemisphere.Citation5 There is a paucity of sub-Saharan data, and specifically South African data.

A recent meta-analysis by Brown et al using BMD as a marker of metabolic bone disease in HIV sufferers, demonstrated a 6.7-fold increase in the odds ratio of developing osteopaenia.Citation5 The odds ratio for osteoporosis was 3.7. A substudy of the AIDS Clinical Trials Group, a North American group investigating the effect of antiretroviral therapy on BMD, demonstrated a similar prevalence of osteopenia and osteoporosis. The baseline prevalence of osteopaenia and osteoporosis before the initiation of HAART was 28% and 7%, respectively.Citation9 More than 60% of patients included in the trials were males, which differs from the HIV gender distribution in South Africa and the rest of the world.Citation1,11 A South African study by Hamill et al in which BMD in black premenopausal South African females was evaluated, could not illustrate a difference between the HIV-negative and HIV-positive patients, irrespective of the cluster of differentiation (CD4) count.Citation12 This illustrates the indefiniteness of current South African data.

HAART has a considerable impact on bone health, independent of the regimen used.Citation9,13 The bone loss seen in patients with HIV on HAART seems to stabilise between 24 and 48 weeks.Citation9 This effect is similar to that observed with glucocorticoid therapy.Citation14 A sub-Saharan study performed in Dakar, Senegal, utilised quantitative heel ultrasound (QUS) for the evaluation of BMD.Citation15 Patients on HAART had a significantly lower BMD (p 0.003) when compared to HIV-negative HAART-naïve controls. Although reduced body mass index (BMI) explained one third of the reduced QUS BMD observed, HAART was responsible for the majority of BMD loss.

The Southern African HIV Clinicians Society guidelines for antiretroviral therapy, published in 2012, only mention the bone toxic effects of tenofovir, although it has been shown that other drugs are also bone toxic.Citation6,9,13 The three most common drugs used in South Africa are a combination of tenofovir, efavirenz (EFV) and lamivudine (3TC). It was shown in a South American study that this combination caused a statistically significant reduction in BMD within the first 48 weeks of initiation.Citation16 The BMD stabilised for the remainder of the study.Citation16

Tenofovir is a nucleoside reverse transcriptase inhibitor (NRTI) and its effects on bone health have been the best studied of all the antiretroviral drugs. The drug has been associated with the development of osteomalacia due to multiple effects on the calcium, vitamin D and parathyroid hormone (PTH) system, as well as proximal renal tubular toxicity.Citation17,18 In a UK study by Klassen et al in which the effect of tenofovir on vitamin D metabolism was examined, it was shown that tenofovir was associated with an increase in PTH levels in non-white men.Citation19 The increase in PTH levels was confirmed in a Spanish cohort.Citation20 The UK study also illustrated that tenofovir use was associated with increased urinary phosphate excretion and reduced serum phosphate levels. Acquired Fanconi syndrome has also been linked to tenofovir use.Citation17 It was shown in mouse models that tenofovir induced genetic changes that led to reduced osteoblast and osteoclast function, with a negative impact on bone remodelling, and subsequently BMD.Citation21,22 It must be noted that the clinical significance of this was recently questioned by a meta-analysis performed by Cooper et al. The study investigated the renal safety of tenofovir, and found that although there was a significant drop in renal function and serum phosphate levels, the clinical effect was moderate at most.Citation23

EFV, a nonnuceloside reverse transcriptase inhibitor, upregulates the expression of CYP24.Citation24 CYP24, a member of the cytochrome P450 super family of enzymes, initiates the degradation of 1,25-dihydroxyvitamin D3 [1,25(OH)2 D], causing the increased conversion of 1,25(OH)2 D to its inactive form.

3TC is an example of an NRTI.Citation25 Zidovudine (AZT) and didanosine (ddI) are examples of other drugs in this category. It was shown in a mouse model that AZT enhanced osteoclastogenesis in a receptor activation of nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) ligand (RANKL)-dependent manner.Citation26 Similar results were seen with 3TC and ddI.Citation25

The effects of other drugs are outside the scope of this article and can be reviewed elsewhere.Citation27

An important question that arises is whether or not a decrease in BMD in HIV-positive patients on HAART translates into an increase in fracture risk. A population-based cohort study showed that patients with HIV infection have an increased incidence ratio of 1.6 (95% confidence interval: 1.4–1.8) of low-energy fractures, compared to HIV-naïve patients.Citation28 The incidence of fractures was significantly increased after exposure to HAART.Citation28 Similarly, Guaraldi et al compared the prevalence of fractures (traumatic or non-traumatic) in HIV-naïve and HIV-positive patients on HAART, and found that there was a 10-fold increase in fracture risk over all age groups in the HIV-population on HAART.Citation29 Once again, it must be noted that more than 60% of the patients in the trials were males, and the studies were conducted in the Northern Hemisphere. Despite the male ratio that was observed in the above studies, it has been shown that HAART reduces BMD, as well as increasing fracture risk.

Multiple risk factors have been identified as contributing to a decreased BMD and associated fragility fractures in patients with HIV. HIV-related bone disease can be attributed to classical and non-classical risk factors. Classical risk factors for metabolic bone disease, which also occur with HIV infection, include poor nutrition, a low BMI, current tobacco use, alcohol use, increased age and low vitamin D levels.Citation15,30 Non-classical risk factors include concurrent hepatitis C infection and a CD4 count less than 200 cells/mmCitation3 or the diagnosis of AIDS.Citation15,30

HIV infection has direct effects on bone remodelling, and influencing osteoclast and osteoblast function. HIV envelope protein, gp120, increases the receptor activation of the NF-κB ligand and HIV viral protein of regulation increases RANKL expression.Citation7,31 This leads to an increase in osteoclast number and function, which results in increased bone resorption.Citation32 HIV infection is associated with elevated tumour necrosis factor alpha (TNF-α) levels.Citation32 TNF-α is a well known stimulator of NF-κB, leading to increased osteoclast formation, bone resorption and metabolic bone disease. This was recently reviewed by Barkhordarian et al.Citation33 Osteoblasts are negatively affected by HIV.Citation7 HIV infection increases apoptotic stimuli in the mesenchymal stem cells, which are precursors to osteoblasts, leading to decreased pre-osteoblast formation, and subsequently, osteoblast formation.Citation34 HIV envelope protein gp120 interacts with the cell membrane of the osteoblast, leading to an increased rate of apoptosis.Citation7 HIV does not seem to directly infect the osteoblast.

Management

HIV-infected patients should undergo a detailed history and physical examination in order to identify clinical risk factors, a propensity for falls and previous fragility fractures. Traditional risk factors for the development of osteoporosis should be excluded. These must include a family history of hip fractures, alcohol abuse, smoking, vitamin D deficiency, a low BMI (< 20 kg/m2), hyperthyroidism and glucocorticoid use.Citation35

The current National Osteoporosis Foundation of South Africa (NOFSA) guidelines state that bone mass measurement is indicated in female patients aged 65 years and older, and in male patients aged 70 years and older.Citation35 Bone mass measurement is also indicated in patients older than 40 years in whom secondary causes for osteoporosis are present, or in patients who sustain a fragility fracture after the age of 40 years. HIV infection and HAART are not currently listed by NOFSA as risk factors for osteoporosis, and further South African data are necessary to determine whether or not these two risk factors should be included in future guidelines. It must also be kept in mind that the majority of South Africans who are infected with HIV are between the ages of 20 and 50 years.Citation11

Baseline special investigations should include a full blood count, erythrocyte sedimentation rate, creatinine, calcium, magnesium, phosphate, thyroid-stimulating hormone and 25-hydroxyvitamin D levels.Citation35 Additional special investigations should be considered in selected patients.

General measures must include stopping smoking, reducing alcohol intake, participating in weight-bearing exercises, fall prevention and adequate calcium and vitamin D supplementation. The NOFSA guidelines state that a daily elemental calcium intake of up to 1 200 mg in most adults is safe and effective in preventing bone loss.Citation35 Calcium supplementation should be individualised according to dietary intake as excessive calcium intake may have detrimental effects on cardiovascular health.Citation36 Vitamin D levels must be maintained above 30 ng/ml for optimal bone health.Citation35

The prevalence of vitamin D deficiency in the HIV-positive population is more than 50% when using a cut-off point of < 20 ng/ml.Citation8,37 Risk factors for vitamin D deficiency include race (black or Hispanic), poor nutrition, advanced disease and the use of HAART.Citation8,37 A vitamin D intake of between 800 IU and 1 000 IU per day is suggested in patients with adequate vitamin D levels. Vitamin D levels of less than 30 ng/ml might warrant treatment with a weekly dose of 50 000 IU vitamin D2 or D3 for 6–8 weeks in order to obtain a level of more than 30 ng/ml.Citation35 Higher dosages might be needed in patients using HAART because of the negative effects of HAART on vitamin D metabolism.Citation24

Ethnic differences in bone metabolism have been described, which may explain the possible differences in fracture risk between black and white South Africans.Citation38 Increased fracture risk associated with HIV infection has been demonstrated internationally, but once again, South Africa-specific data are needed.Citation28 The specific therapeutic agent (bone-active drug) that is effective and safe in this group of patients still needs to be determined. Anti-osteoporotic drugs are usually investigated in female patients aged 50 years and older. There are data on only a few drugs in the younger population. Bisphosphonates are well known and widely used drugs in the treatment of osteoporosis. The effects of once-weekly oral alendronate, in combination with calcium and vitamin D supplementation, were compared to calcium and vitamin D supplementation alone, in HIV sufferers on HAART, in a trial by Mondy et al. Citation39 There was a statistically significant change in lumbar spine BMD in the alendronate group, compared to that in the non-alendronate group. It must also be noted that there were no serious adverse events. Bolland et al demonstrated that intravenous zoledronic acid was also associated with an increase in BMD, not only at the lumbar spine, but also at the hip and total body.Citation40 However, it must be noted that most of the patients in these trials were males aged approximately 50 years, whereas as previously mentioned, the majority of HIV sufferers in South Africa are females in their reproductive years.

The future

With the advent of effective HAART, HIV infection has undergone a metamorphosis from being a fatal disease characterised by progressive deterioration and associated with opportunistic infections, to a chronic controllable condition. It is now frequently complicated by diseases of lifestyle, including osteoporosis, which increase the burden of disease associated with HIV.

HIV infection is associated with a decrease in BMD and an increased fracture risk in non-South African populations, and it seems that this effect is compounded by HAART, irrespective of the drug class used.Citation9,13,29,30 Current South African data are inconclusive, and further research is warranted in order to determine whether or not the South African HIV population carries the same risk for metabolic bone disease as that of its Northern Hemisphere counterparts.

If the South African HIV population is found to have an increased risk of fragility fractures, disregard could lead to an escalation in the morbidity, mortality and economic burden associated with HIV infection in a country already straining with limited resources. BMD testing is currently expensive, and not freely available to the majority of South African citizens who are dependent on public health services. If data from local studies indicate that HIV is a threat to bone health, BMD testing should be incorporated in the management of HIV patients.

References

  • Joint United Nations Programme on HIV/AIDS (UNAIDS). Global report: UNAIDS report on the global AIDS epidemic. 2012 [cited 2013 August 28]. Available from: http://www.unaids.org/en/dataanalysis/knowyourepidemic/2012
  • Islam FM, Wu J, Jansson J, et al. Relative risk of cardiovascular disease among people living with HIV: a systematic review and meta-analysis. HIV medicine. 2012;13(8):453–68. Epub 2012/03/15.
  • Engels EA, Biggar RJ, Hall HI, et al. Cancer risk in people infected with human immunodeficiency virus in the United States. Int J Cancer. 2008;123(1):187–94. Epub 2008/04/26.10.1002/(ISSN)1097-0215
  • Galli L, Salpietro S, Pellicciotta G, et al. Risk of type 2 diabetes among HIV-infected and healthy subjects in Italy. Eur J Epidemiol. 2012;27(8):657–665. Epub 2012/06/23.10.1007/s10654-012-9707-5
  • Brown TT, Qaqish RB. Antiretroviral therapy and the prevalence of osteopenia and osteoporosis: a meta-analytic review. AIDS. 2006;20(17):2165–74. Epub 2006/11/07.10.1097/QAD.0b013e32801022eb
  • Meintjes G, Boulle A, et al. Guidelines for antiretroviral therapy in adults. S Afr J HIV Med. 2012;13(3):114–33.
  • Gibellini D, De Crignis E, Ponti C, et al. HIV-1 triggers apoptosis in primary osteoblasts and HOBIT cells through TNFα activation. J Med Virol. 2008;80(9):1507–14. Epub 2008/07/24.10.1002/jmv.v80:9
  • Rodríguez M, Daniels B, Gunawardene S, et al. High frequency of vitamin D deficiency in ambulatory HIV-positive patients. AIDS Res Hum Retroviruses. 2009;25(1):9–14. Epub 2008/12/26.10.1089/aid.2008.0183
  • 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(12):1791–801. Epub 2011/05/25.10.1093/infdis/jir188
  • Riggs BL, Melton LJ 3rd. The worldwide problem of osteoporosis: insights afforded by epidemiology. Bone 1995;17(5):S505–11. Epub 1995/11/01.10.1016/8756-3282(95)00258-4
  • Shisana ORT, Simbayi LC, Zuma K, et al. South African national HIV prevalence, incidence, behaviour and communication survey 2008: a turning tide among teenagers? HSRC Press; 2009.
  • Hamill MM, Ward KA, PettiforJM, et al. Bone mass, body composition and vitamin D status of ARV-naive, urban, black South African women with HIV infection, stratified by CD count. Osteoporos Int. 2013;24:2855–61. Epub 2013/05/31.
  • Amiel C, Ostertag A, Slama L, et al. BMD is reduced in HIV-infected men irrespective of treatment. J Bone Miner Res. 2004;19(3):402–9. Epub 2004/03/26.
  • Staa TP, Staa T. P. van, Staa T. P. van. The epidemiology of corticosteroid-induced osteoporosis: a meta-analysis. Osteoporos Int. 2002;13(10):777–87. Epub 2002/10/16.10.1007/s001980200108
  • Cournil A, Eymard-Duvernay S, Diouf A, et al. Reduced quantitative ultrasound bone mineral density in HIV-infected patients on antiretroviral therapy in senegal. PLoS ONE. 2012;7(2):e31726. Epub 2012/02/24.10.1371/journal.pone.0031726
  • 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-naïve HIV-1—infected patients. HIV Clin Trials. 2007;8(3):164–72. Epub 2007/07/11.10.1310/hct0803-164
  • Woodward CL, Hall AM, Williams IG, et al. Tenofovir-associated renal and bone toxicity. HIV Med. 2009;10(8):482–7. Epub 2009/05/23.10.1111/hiv.2009.10.issue-8
  • Parsonage MJ, Wilkins EG, Snowden N, et al. The development of hypophosphataemic osteomalacia with myopathy in two patients with HIV infection receiving tenofovir therapy. HIV Med. 2005;6(5):341–6. Epub 2005/09/15.10.1111/hiv.2005.6.issue-5
  • Klassen K, Martineau AR, Wilkinson RJ et al. The Effect of tenofovir on vitamin D metabolism in HIV-infected adults is dependent on sex and ethnicity. PLoS ONE. 2012;7(9):e44845. Epub 2012/09/18.10.1371/journal.pone.0044845
  • Masiá M, Padilla S, Robledano C, et al. Short communication: early changes in parathyroid hormone concentrations in HIV-infected patients initiating antiretroviral therapy with tenofovir. AIDS Res Hum Retroviruses. 2012;28(3):242–6. Epub 2011/06/07.10.1089/aid.2011.0052
  • Grigsby IF, Pham L, Mansky LM, et al. Tenofovir treatment of primary osteoblasts alters gene expression profiles: implications for bone mineral density loss. Biochem Biophys Res Commun. 2010;394(1):48–53. Epub 2010/02/23.10.1016/j.bbrc.2010.02.080
  • Grigsby IF, Pham L, Gopalakrishnan R, et al. Downregulation of Gnas, Got2 and Snord32a following tenofovir exposure of primary osteoclasts. Biochem Biophys Res Commun. 2010;391(3):1324–1329. Epub 2009/12/23.10.1016/j.bbrc.2009.12.039
  • Cooper RD, Wiebe N, Smith N, et al. Systematic review and meta‐analysis: renal safety of tenofovir disoproxil fumarate in hiv‐infected patients. Clin Infect Dis. 2010;51(5):496–505. Epub 2010/08/03.10.1086/652979
  • Landriscina M, Altamura SA, Roca L, et al. Reverse transcriptase inhibitors induce cell differentiation and enhance the immunogenic phenotype in human renal clear-cell carcinoma. Int J Cancer. 2008;122(12):2842–50. Epub 2008/03/21.10.1002/(ISSN)1097-0215
  • Pan G, Kilby M, McDonald JM. Modulation of osteoclastogenesis induced by nucleoside reverse transcriptase inhibitors. AIDS Res Hum Retroviruses. 2006;22(11):1131–41. Epub 2006/12/07.10.1089/aid.2006.22.1131
  • Pan G, Wu X, McKenna MA, et al. AZT enhances osteoclastogenesis and bone loss. AIDS Res Hum Retroviruses. 2004;20(6):608–20. Epub 2004/07/10.10.1089/0889222041217482
  • Pan G, Yang Z, Ballinger SW, et al. Pathogenesis of osteopenia/osteoporosis induced by highly active anti-retroviral therapy for AIDS. Ann N Y Acad Sci. 2006;1068:297–308. Epub 2006/07/13.10.1196/annals.1346.057
  • Hansen AB, Gerstoft J, Kronborg Get al. Incidence of low and high-energy fractures in persons with and without HIV infection. AIDS. 2012;26(3):285–93. Epub 2011/11/19.10.1097/QAD.0b013e32834ed8a7
  • Guaraldi G, Orlando G, Zona S, et al. Premature age-related comorbidities among HIV-infected persons compared with the general population. Clin Infect Dis. 2011;53(11):1120–6. Epub 2011/10/15.10.1093/cid/cir627
  • Young B, Dao CN, Buchacz K, et al. 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(8):1061–8. Epub 2011/03/15.10.1093/cid/ciq242
  • Crotti TN, Smith MD, Findlay DM, et al. Factors regulating osteoclast formation in human tissues adjacent to peri-implant bone loss: expression of receptor activator NFkappaB, RANK ligand and osteoprotegerin. Biomaterials. 2004;25(4):565–73. Epub 2003/11/11.
  • Vikulina T, Fan X, Yamaguchi M, et al. Alterations in the immuno-skeletal interface drive bone destruction in HIV-1 transgenic rats. Proc Natl Acad Sci. 2010;107(31):13848–53. Epub 2010/07/21.10.1073/pnas.1003020107
  • Barkhordarian A, Ajaj R, Ramchandani MH, et al. Osteoimmunopathology in HIV/AIDS: a translational evidence-based perspective. Pathol Res Int. 2011;2011:359242. Epub 2011/06/11.
  • Cotter EJ, Chew N, Powderly WG. HIV type 1 alters mesenchymal stem cell differentiation potential and cell phenotype ex vivo. AIDS Res Hum Retroviruses. 2011;27(2):187–99. Epub 2010/10/12.10.1089/aid.2010.0114
  • Hough SA-E, Brown S, Cassim B, et al. NOFSA guideline for the diagnosis and management of osteoporosis. JEMDSA. 2010;15(3):107–8.
  • Bolland MJ, Grey A, Avenell A, et al. Calcium supplements with or without vitamin D and risk of cardiovascular events: reanalysis of the Women’s health initiative limited access dataset and meta-analysis. BMJ. 2011;342:d2040. Epub 2011/04/21.10.1136/bmj.d2040
  • Adeyemi OM, Agniel D, French AL, et al. Vitamin D deficiency in HIV-infected and HIV-uninfected women in the United States. J Acquir Immune Defic Syndr. 2011;57(3):197–204. Epub 2011/04/08.10.1097/QAI.0b013e31821ae418
  • Schnitzler CM, Mesquita JM. Cortical bone histomorphometry of the iliac crest in normal black and white South African adults. Calcif Tissue Int. 2006;79(6):373–82. Epub 2006/12/13.10.1007/s00223-006-0053-z
  • Mondy K, Powderly WG, Claxton SA, et al. Alendronate, vitamin D, and calcium for the treatment of osteopenia/osteoporosis associated with HIV infection. J Acquir Immune Defic Syndr. 2005;38(4):426–31. Epub 2005/03/15.10.1097/01.qai.0000145352.04440.1e
  • Bolland MJ, Grey A, Horne AM, et al. Effects of intravenous zoledronate on bone turnover and bone density persist for at least five years in HIV-infected men. J Clin Endocrinol Metab. 2012;97(6):1922–8. Epub 2012/03/16.10.1210/jc.2012-1424