Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2017 May 1.
Published in final edited form as: Curr Opin HIV AIDS. 2016 May;11(3):333–342. doi: 10.1097/COH.0000000000000260

The Protease Inhibitors and HIV Associated Bone Loss

Caitlin A Moran 1,2, M Neale Weitzmann 3,4, Ighovwerha Ofotokun 1,2,#
PMCID: PMC4838480  NIHMSID: NIHMS777571  PMID: 26918650

Abstract

Purpose of review

HIV infection is an established risk factor for osteoporosis and bone fracture. Combination antiretroviral therapy (cART) increases bone resorption leading to an additional 2–6% bone mineral density (BMD) loss within the first 1–2 years of therapy. While tenofovir disoproxil fumarate is often blamed for antiretroviral drug-associated bone loss, evidence abounds to suggest that other agents, including the protease inhibitors (PI) have adverse bone effects. In the current review, we examine bone loss associated with PI use, describing the relative magnitude of bone loss reported for individual PIs. We also review the potential mechanisms associated with PI-induced bone loss.

Recent findings

As a class, PIs contribute to a greater degree of bone loss than other anchor drugs. HIV disease reversal and the associated immune reconstitution following cART initiation play an important role in PI-mediated bone loss in addition to plausible direct effects of PIs on bone cells.

Summary

PIs remain an important component of cART despite their adverse effects on bone. A better understanding of factors that drive HIV/cART-induced bone loss is needed to stem the rising rate of fracture in the HIV-infected population.

Keywords: Protease inhibitors, cART-induced bone loss, osteopenia, osteoporosis, fracture, HIV

Introduction

Combination antiretroviral therapy (cART) is now recommended for all HIV-infected patients regardless of CD4 T cell counts [1**3], and those treated with cART can expect to attain a near-normal life expectancy [4]. However, many will experience age-related comorbidities including musculoskeletal abnormalities, cardiovascular diseases, renal impairment, and certain non-AIDS associated malignancies with greater frequency and at younger ages than their HIV-uninfected counterparts [5, 6*, 7*]. Indeed, HIV infection is now an established risk factor for osteopenia and osteoporosis [8] as defined by the World Health Organization criteria [femoral neck or lumbar spine T-score as measured by dual energy X-ray absorptiometry (DXA) between −1.0 and −2.5 (osteopenia) and less than or equal to −2.5 (osteoporosis)] [9]. cART further aggravates rather than alleviates HIV-associated bone loss by inducing an additional 2% to 6% loss in bone mineral density (BMD) within the first two years of therapy, a rate of bone loss comparable to that seen in post-menopausal osteoporosis, the archetype of fragility bone disease [7*, 9, 10]. The rate of BMD loss decreases after 1–2 years of cART [1113], but whether bone resorption returns to baseline (already elevated in HIV-infected subjects) or to levels associated with uninfected subjects remains unclear. Importantly, the bone effects of cART appear to be universal across all regimens, although the magnitude of the effect may vary by regimen [12, 14, 15]. In one meta-analysis, the odds ratio (OR) of osteoporosis among HIV-infected individuals compared with HIV-uninfected controls was 3.7 [95% confidence interval (CI) 2.3 – 5.9]; cART use conferred an additional 2.5 fold increased odds of low BMD among HIV-infected patients [16].

Despite the relatively young age of the HIV/AIDS population, the high prevalence of fragility bone disease in this group is accompanied by an increasing rate of bone fractures [17]. In the landmark study of Triant et al. [17] involving 8,525 HIV-infected patients and 2,208,792 HIV-negative controls, an increase in fracture prevalence of up to 4 fold was observed in both sexes over a wide age range. Importantly, while fracture rates in HIV-negative men historically have been low until advanced age, fracture rates in HIV-infected men have risen dramatically even at young ages. For example, age-adjusted fracture rates were 2–4 fold higher in the HOPS cohort of 5,826 HIV-infected patients compared with HIV-uninfected adults in the U.S. general population [18]. In the Veterans Aging Cohort Study Virtual Cohort (VASC-VC) comprising 40,115 HIV-infected patients, fracture rates were 24% – 32% higher compared with HIV-negative controls, and in the Women’s Interagency HIV Study (WIHS) cohort, HIV-infected patients were found to have a greater incidence of fragility fractures than HIV-uninfected patients [19, 20*]. In addition, a recent Spanish study with 2,489 HIV-infected and 1,115,667 HIV-uninfected participants revealed a 5-fold higher hip fracture rate among participants with HIV infection [21], and a large Danish case-control study using nationwide health registry data found a 9-fold higher risk of fracture at the hip in HIV-infected patients compared with HIV-uninfected patients [22**]. Of note, data from two prospective cohorts of HIV-infected patients with a median age of 43 years in the U.S. demonstrated that osteoporosis is associated with a four times greater risk of fracture compared with normal BMD, thus linking osteoporosis with the rising fracture rates observed in the HIV/AIDS population [23*], which is in contrast to the general population in which BMD is not a predictor of fracture in younger patients [24]. Taken together, these data suggest that an understanding of the factors underlying HIV/cART-induced bone loss is needed to guide effective preventive and therapeutic strategies to stem the looming epidemic of bone fracture in the aging HIV/AIDS population. In the current review, we examine the role of the PIs in HIV/cART-induced bone loss, describing the magnitude of bone loss reported for individual PIs relative to other PIs and non-PI based cART. We also review the potential mechanisms associated with PI-induced bone loss.

The bone effects of the HIV protease inhibitors

While the effects of the nucleotide reverse transcriptase inhibitor (NRTI) tenofovir disoproxil fumarate (TDF) on bone resorption and BMD are well established [2528*, 29, 30*], the effects of the PIs on BMD are less clear. However, most studies suggest an association between PI use and BMD loss [15, 31**35*]. The magnitude of this effect was examined in a meta-analysis of cross-sectional studies by Brown and Qaqish, who found an OR for osteoporosis of 1.57 (95% CI 1.05 – 2.34) in HIV-infected patients treated with PIs compared with those on non-PI containing cART regimens [16]. In a Japanese cohort study, each year of PI exposure was associated with an OR of 1.100 [95% CI 1.003 – 1.207] and 1.187 (95% CI 1.043 – 1.351) for low BMD in the spine and femoral neck, respectively, independent of TDF exposure [31**]. Furthermore, data from the Veterans Affairs’ Clinical Case Registry showed that the cumulative exposure to ritonavir-boosted lopinavir (LPV/r) [but not ritonavir-boosted atazanavir (ATV/r), ritonavir-boosted indinavir (IDV/r) or ritonavir (RTV) alone] was independently predictive of osteoporotic fracture [36]. Randomized clinical trials have also provided mixed evidence regarding the effects of PI use on BMD loss in treatment-naïve HIV-infected individuals (Table 1). The AIDS Clinical Trials Group (ACTG) substudy A5005s found no significant difference in total body mean percent BMD change between patients randomized to efavirenz (EFV) vs. ritonavir-boosted nelfinavir (NFV/r) [42]. Brown et al. likewise found no difference in the total body mean percent BMD change in patients receiving EFV vs. LPV/r [14]. However, these studies were limited by their reliance on BMD measurements from whole body DXA rather than measurements obtained at specific body sites.

Table 1.

Randomized clinical trials: PIs compared with other anchor drugs

Author (1st)/year NRTI backbone PIs Comparison Duration Findings
Treatment naive
Rockstroh et al. 2013 [37] TDF/FTC ATV/r EVG/c/
FTC/TDF
96 weeks Fracture:
1.1% EVG/c/FTC/TDF, 3.9% ATV/r*
Spine BMD mean % change:
−1.96% EVG/c/FTC/TDF, −3.54% ATV/r
Hip BMD mean % change:***
−3.16% EVG/c/FTC/TDF, −4.19% ATV/r
Brown TT et al. 2015 [38**] TDF/FTC DRV/r
ATV/r
RAL 96 weeks Spine BMD mean % change:+
−3.1% DRV/r, −4.0% ATV/r, −1.6% RAL
−3.8% pooled PI, 1.8% RAL
Hip BMD mean % change:+
−3.3% DRV/r, −3.7% ATV/r, −2.2% RAL
−3.7% pooled PI, −2.4% RAL
Total body BMD mean % change:
−2.9% ATV/r vs. −1.6% DRV/r
−2.9% ATV/r vs. −1.7% RAL
DRV/r vs. RAL NS
McComsey et al. 2011 [26] TDF/FTC
ABC/3TC
ATV/r EFV 96 weeks Spine BMD mean % change:
TDF/FTC: −3.3%, ABC/3TC −1.3%
EFV −1.7%, ATV/r 3.1%
Hip BMD mean % change:
TDF/FTC −4.0%, ABC/3TC −2.6%
EFV −3.1%, ATV/r 3.4%, NS
Bonnet et al. 2013 [39] ZDV/3TC
TDF/FTC
ABC/3TC
ddI/3TC
PI/r:
fAPV/r
ATV/r
SQV/r
IDV/r
LPV/r
NNRTI:
EFV
NVP
21 months L2–L4 BMD mean % change:
−1.5% NNRTI vs. −4.0 PI/r
Duvivier et al. 2009 [40] ZDV/3TC
ddI/3TC
TDF/3TC
LPV/r
IDV/r
EFV
NVP
48 weeks Spine BMD mean % change:++
PI/r+NNRTI −4.4%,
PI/r +NRTI −5.8%,
NNRTI + NRTI −1.5%
Hip BMD mean % change: NS between all 3 groups
Moyle et al. 2015 [41*] TDF/FTC LPV/r
ATV/r
- 96 weeks Trunk BMD mean % change:
ATV/r −2.8%, LPV/r −4.6%
Total body BMD mean % change:
ATV/r −2.65%, LPV/r −3.7% (p=0.077)**
Brown TT et al. 2009 [14] ZDV/3TC LPV/r EFV 96 weeks Total body BMD mean % change:***
−2.5% LPV/r, −2.3% EFV NS (p=0.86)
Tebas et al. 2007 [42] ddI/d4T
ZDV/3TC
NFV/r EFV 96 weeks Total body BMD median % change:***
−2.25% average, NS between groups
Huang et al. 2013 [27] ZDV/3TC
d4T/3TC
TDF/3TC
LPV/r EFV 96 weeks Total body BMD mean % change:
NRTI-sparing: −2.21%
EFV/ZDV+3TC: −1.59%
LPV/ZDV+3TC: −1.89%
EFV/d4T+3TC: −1.66%
LPV/d4T+3TC: −2.35%
EFV/TDF+3TC: −2.31%
LPV/TDF+3TC: −3.98%
Hansen et al. 2011 [12] ZDV/3TC LPV/r EFV 144 weeks Spine BMD mean % change:***
PI-sparing: −3.2%
NRTI-sparing: −2.7%
Hip BMD mean % change:
PI-sparing: −5.0%
NRTI-sparing: −4.5%
Treatment experienced
Curran et al. 2012 AIDS SPIRAL-LIP substudy [43] TDF
ABC
ZDV
ddI
d4T
3TC/FTC
PI/r
ATV/r
fAPV/r
LPV/r
SQV/r
RAL 48 weeks Total body BMD (g/cm2):
RAL: +0.01, PI/r: 0
PI/r 0 vs. RAL +0.01***
Spine BMD (g/cm2):
PI/r +0.02, RAL: +0.0***
Total hip BMD (g/cm2):
PI/r: 0, RAL: +0.01
PI/r vs. RAL: +0.01***

NRTI, nucleos(t)ide reverse transcriptase inhibitor; PI, protease inhibitor; PI/r, ritonavir-boosted protease inhibitor; TDF, tenofovir disoproxil fumarate; FTC, emtricitabine; ATV, atazanavir; EVG, elvitegravir; c, cobicistat; BMD, bone mineral density; DRV, darunavir; RAL, raltegravir; ABC, abacavir; 3TC lamivudine; EFV, efavirenz; ZDV, zidovudine; ddI, didanosine; fAPV, fosamprenavir; SQV, saquinavir; IDV, indinavir; LPV, lopinavir; NNRTI, non-nucleoside reverse transcriptase inhibitor; NVP, nevirapine; d4T, stavudine; NS, non-significant

*

Most fractures were traumatic. 3 non-traumatic fractures were seen in the ATV/r arm.

**

Differences in mean % change of both trunk and total body BMD mean % change were statistically significant for men, and were not statistically significant for women

***

Differences are not statistically significant

+

NS between PI arms; statistically significant for each PI vs. RAL comparison

++

p=0.007 PI/r + NNRTI vs. NNRTI + NRTI; p= 0.001 PI/r + NRTI vs. NNRTI + NRTI

In studies in which BMD loss at specific body sites was compared, significant differences were seen between PIs and other anchor drugs. In the ACTG substudy A5224s, there was greater mean percent BMD loss in the spine among patients receiving ATV/r vs. those receiving EFV, although no statistically significant difference was seen in BMD loss in the hip [26]. Duvivier et al. also reported a greater loss of BMD in the spine, but not the hip, in patients on a ritonavir-boosted PI (PI/r) vs. those on a non-nucleoside reverse transcriptase inhibitor (NNRTI) in the Hippocampe-ANRS 121 study [40]. Finally, Rockstroh et al. found that patients receiving ritonavir-boosted darunavir (DRV/r) with NRTI backbone tenofovir-emtricitabine (TDF-FTC) had significantly greater loss of spine, but not hip, BMD than those receiving elvitegravir/cobicistat/emtricitabine/tenofovir (EVG/c/FTC/TDF) [37]. The mechanisms accounting for this difference in body site measurements are unclear, but may be attributable in part to the weaker correlation between site-specific DXA and whole body DXA, particularly at the hip [44]. In addition, it has been speculated that this effect may be due to the faster turnover of trabecular vertebral bone compared with the relatively slow turnover of cortical bone in the hip [26].

To date, only two randomized clinical trials have compared the BMD loss associated with two different PIs. In the ACTG substudy 5260s, no difference was observed in the mean percent BMD loss in both the spine and hip in patients receiving ATV/r vs. DRV/r; however, patients in both PI arms had a greater percent BMD loss than patients in the integrase strand transfer inhibitor (INSTI) raltegravir (RAL) arm in both the spine and hip [38**]. Interestingly, while there was no difference in total body mean percent BMD loss in the DRV/r and RAL arms, patients in the ATV/r arm experienced more total body BMD loss than those in either of the other two arms [38**]. Investigators in the CASTLE substudy reported a greater loss of trunk mean percent BMD in patients on LPV/r compared with those on ATV/r. While no statistically significant difference was seen overall in total body mean percent change between groups, when stratified by sex, greater total body mean percent BMD loss was seen in men on LPV/r compared with men on ATV/r, while no significant difference was seen among women [41*]. Although the evidence regarding the effects of PIs on bone health in treatment experienced patients is less robust, the SPIRAL-LIP study found a 0.01 g/cm2 increase in the femoral neck BMD of virologically suppressed patients switched from a PI/r-based regimen to a RAL-based regimen, with no statistically significant difference in the total body or total hip BMD [43].

While the above results argue that bone loss especially soon after cART initiation is attributable to PIs, results from other clinical trials suggest that maintaining a PI while removing the NRTI backbone also results in less loss of BMD both in viremic and virologically suppressed patients (Table 2). Treatment experienced patients failing first line therapy who were randomized to second line therapy with either LPV/r + RAL or LPV/r + 2NRTIs experienced less bone loss in the LPV/r + RAL arm; the greatest effect was seen after 48 weeks with subsequent stabilization by 96 weeks [47**, 49]. Treatment naïve patients enrolled in the RADAR study who were randomized to RAL + DRV/r experienced a smaller increase in markers of bone turnover as well as an increase in total and subtotal BMD from baseline compared with the greater increase in bone turnover markers and a decrease in BMD experienced by patients treated with TDF-FTC + DRV/r [48*]. Virologically suppressed patients in the Monarch RCT substudy who were randomized to DRV/r monotherapy experienced an increase in spine and hip BMD compared with those who were maintained on a 2NRTI + DRV/r regimen [45*]. Similarly, results from the MIDAS study demonstrate an improvement in BMD in patients switched from TDF/FTC/EFV to DRV/r monotherapy [46*]. Similar results were seen in a nonrandomized study in which cART experienced, virologically suppressed patients on a PI/r based regimen containing TDF were switched from TDF to RAL [50].

Table 2.

Randomized clinical trials: PIs with and without NRTIs

Author (1st)/year Baseline regimen Comparison Study duration Findings
Virologically suppressed
Guaraldi et al. 2014 [45*] TDF/FTC + DRV/r or ABC/3TC + DRV/r DRV/r 48 weeks Lumbar spine BMD (g/cm2):
+0.01 DRV/r, 0.0 DRV/r + 2NRTI
Femur BMD (g/cm2):
+0.02 DRV/r, 0.0 DRV/r + 2NRTI
Total body BMD (g/cm2):
+0.01 DRV/r, +0.01 DRV/r + 2NRTI
Hamzah et al. 2015 [46*] TDF/FTC/EFV DRV/r 48 weeks Hip BMD mean % change:
TDF/FTC/EFV −0.002%, DRV/r +1.8%
Femoral neck BMD mean % change:
TDF/FTC/EFV −0.003%, DRV/r +2.9%
Lumbar spine BMD mean % change:
TDF/FTC/EFV +0.008%, DRV/r +2.6%
Viremic
Haskelberg et al. 2014 [47**] 2NRTI + LPV/r LPV/r + RAL 96 weeks Hip BMD mean % change:
2NRTI + LPV/r −4.1%, LPV/r + RAL −2.2%
Lumbar spine BMD mean % change:
2NRTI + LPV/r −4.9%, LPV/r + RAL −3.5%
Bedimo et al. 2014 [48*] TDF/FTC + DRV/r DRV/r + RAL 48 weeks Subtotal BMD change (g/cm2):
TDF/FTC −7.0, RAL +9.2

TDF, tenofovir disoproxil fumarate; FTC, emtricitabine; DRV, darunavir;/r, ritonavir-boosted; ABC, abacavir; 3TC, lamivudine; EFV, efavirenz

These data suggest that a proportion of the bone loss observed with PI use may be attributed to concomitant TDF use. Indeed, RTV has been shown to increase plasma tenofovir (TFV) concentrations by 32 – 50% [51, 52*, 53], via inhibition of active TFV secretion by the proximal convoluted tubule [54, 55]. Similarly, cobicistat (COBI), a CYP 3A4 inhibitor that acts similarly to RTV to boost PI and EVG levels, has been shown to increase plasma TFV concentrations by 24 – 30%, possibly via the inhibition of gastrointestinal efflux transporter P-glycoprotein (P-gp), resulting in greater TDF absorption [55, 56]. However, as seen in the study by Rockstroh et al., PIs have an effect on bone loss beyond what can be explained by increased TFV levels by COBI [37].

Although these studies provide significant evidence that PIs contribute to bone loss, the choice of a cART regimen is complex, involving many biologic and psychosocial factors. The benefits of cART far outweigh any risks of future bone disease, and PIs remain part of recommended first-line and salvage regimens [57, 58**]. For treatment naïve patients with known osteoporosis, we concur with the recommendation that the clinician and patient weigh the risks and benefits of initiating a PI-based regimen and continue to monitor the patient’s bone health [58**]. The data are less clear on whether patients with osteoporosis who are virologically suppressed for more than 12 – 24 months on a PI-based regimen would benefit from switching to an alternative regimen.

Proposed mechanisms of PI-associated bone loss

Despite the abundance of evidence from clinical studies for PI-associated bone loss, the underlying mechanism remains unclear. The maintenance of skeletal health and bone homeostasis are complex processes mediated by a balance between osteoblastic bone formation and osteoclastic bone resorption [8, 59]. Osteoblasts are derived from mesenchymal stem cells (MSCs), while osteoclasts are cells of monocyte-macrophage origin whose differentiation is regulated by the receptor activator of nuclear factor-kappa B ligand (RANKL) and its decoy receptor, osteoprotegerin (OPG) [60, 61]. Therefore, processes that increase osteoclastic bone resorption relative to osteoblastic bone formation will lead to BMD loss.

Direct effects of PI on bone cells

Previously, it was thought that the bone effects of the antiretroviral drugs were mediated by direct toxicity of the drugs on bone cells. However, establishing this phenomenon in vivo has been challenging since these drugs are used together in cART. While certain data suggest that antiretroviral drugs do have effects on osteoclasts and osteoblasts in vitro, and in animal models in vivo, results from these experiments generally have failed to recapitulate the in vivo bone effects observed in the clinical setting. For example, in in vitro experiments, the PI fosamprenavir (FPV) increases OPG expression and decreases RANKL production, while PIs RTV and saquinavir (SQV) were found to abrogate a physiologic block to RANKL [62, 63], effects that should result in an increase in BMD rather than the clinically observed loss of BMD. As another example, RTV, long considered a major protagonist of bone loss in humans, was shown in one study to inhibit osteoclast function and suppress osteoclastogenesis in vitro and in vivo by impairing RANKL-induced signaling [60], although RTV concentrations in that study were greater than normal pharmacologic concentrations [64]. Of note, the related PI indinavir (IDV) had no effect on osteoclastogenesis [60]. In contrast, other in vitro data have suggested a potential mechanism for PI-associated bone loss. NFV and IDV have been shown to alter osteoblast gene expression leading to a decrease in osteoblastic phenotype including a decrease in bone alkaline phosphatase activity and calcium deposition [65], while an increase in senescence of human MSCs when exposed to ATV and LPV leads to a decrease in differentiation to osteoblasts [59], which is consistent with clinical observations [26, 40]. Additional studies have demonstrated that RTV, at serum concentrations achieved with standard dosing for PI-boosting, increases the differentiation of peripheral blood mononuclear cells (PBMCs) into osteoclasts by upregulating growth factors and suppressing transcripts of antagonists in vitro [64, 66, 67]. A greater effect was observed in bone turnover markers (BTMs) and osteoclast differentiation from PBMCs in sera obtained from women on RTV-containing regimens compared with sera from HIV-infected women on other cART regimens as well as from HIV-uninfected women [66]. Taken together, these results demonstrate that PIs have the potential to reduce the RANKL/OPG ratio, inhibit osteoblastic activity, and enhance osteoclast formation. The clinical significance of these effects remains unclear, and further research is needed.

HIV disease reversal and immune reconstitution

Because cART-induced bone loss is universal across all antiretroviral drug classes, it has been speculated that this effect may be due to drug-induced HIV disease reversal and T-cell restoration. Recently, our group examined bone turnover in treatment naïve HIV-infected patients initiating cART. We observed a surge in bone resorption, starting as early as 2 weeks after cART initiation and lasting through 24 weeks [68**]. Because T-cell recovery with cART reaches a significant magnitude by 12 weeks [69], the time point at which we observed a peak in bone resorption, we speculated that there was a link between immune reconstitution and cART-induced bone loss [68**]. Using an animal model of immune reconstitution created by adoptive transfer of T-cells into T-cell knock-out mice, we demonstrated that immune reconstitution did indeed result in a profound loss in BMD via activation of T-cells and/or other immune cells leading to RANKL and/or tumor necrosis factor-alpha (TNF-α) production [70*].

Altered vitamin D metabolism by protease inhibitors

Vitamin D is important for bone metabolism and for maintaining serum calcium levels. Vitamin D insufficiency and deficiency result in in some cases in secondary hyperparathyroidism, which in turn stimulates osteoclastogenesis via production of RANKL [71]. Vitamin D deficiency may further lead to osteomalacia (poorly mineralized bone matrix). Both Vitamin D insufficiency and deficiency are highly prevalent in HIV-infected individuals [72*, 73]. They are generally worsened by cART regimens that contain TDF and EFV [73, 74], and Vitamin D and calcium supplementation have been shown to mitigate the bone loss seen with initiation of an EFV-based regimen [75**]. The effect of PIs on vitamin D levels is less clear. Conversion of 25-hydroxyvitamin D [25-(OH)D] to the active metabolite 1,25-dihydroxyvitamin D [1,25-(OH)2D] is impaired by PIs in vitro via suppression of 25- and 1α-hydroxylase in hepatocyte and monocyte cultures, although the clinical significance of this inhibition remains unclear [76]. Most observational studies have evaluated the association between PIs and 25-(OH)D levels rather than 1,25-(OH)2D levels, and suggest an increase in 25-(OH)D levels with initiation of PI therapy, which may be due to inhibition of the conversion of 25-(OH)D [7779]. In a small clinical trial of vitamin D-deficient HIV-infected postmenopausal women on cART, supplementation with high- dose cholecalciferol was shown to increase both 25-(OH)D and 1,25-(OH)2D levels with concurrent decrease in parathyroid hormone (PTH) levels regardless of PI therapy, suggesting that PI-induced 25- and 1α-hydroxylase suppression can be overcome, although the kinetics have not been described [80]. Further study of the clinical effects of PIs on vitamin D metabolism is warranted.

Conclusions

The HIV PIs contribute to bone loss in the setting of HIV infection, which appears to be a class effect that is observed with all PIs that have been studied. HIV disease reversal and the associated immune reconstitution following cART initiation may play a central role in cART-mediated bone loss. Other potential mechanisms specific to PIs include the direct effect of PIs on the RANKL/OPG axis and on osteoblasts and osteoclasts, as well as the inhibitory effect of PIs on vitamin D metabolism. The PIs remain an important component of cART, and future research is warranted to investigate both the pathophysiology of PI-induced bone loss and prevention strategies in order to impact the long-term health of an aging HIV-infected population.

Key points.

  • HIV infection and cART use are established risk factors for osteoporosis, and protease inhibitors as a class contribute to cART-induced bone loss.

  • Protease inhibitors are associated with a greater degree of bone loss than NNRTIs and INSTIs.

  • The mechanisms of PI-associated bone loss are not fully elucidated; however, the effects of immune reconstitution and T cell recovery after cART initiation likely play a central role.

Acknowledgments

Financial support and sponsorship

The authors’ research activities are supported by the National Institute on Aging (NIA) under Award Number R01AG040013 and the National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS) under Award Numbers R01AR059364 and R01AR068157 to M.N.W. and I.O. M.N.W. is also supported by a grant from the Biomedical Laboratory Research & Development Service of the VA Office of Research and Development (5I01BX000105) and by NIAMS grant (R01AR056090). C.A.M. is also supported by National Center for Advancing Translational Sciences of the National Institutes of Health under Award Number UL1TR000454. The authors gratefully acknowledge services provided by the Emory Center for AIDS Research (CFAR) funded though NIAID (P30AI050409) and the Atlanta Clinical and Translational Science Institute (ACTSI), funded though the National Center for Advancing Translational Sciences (UL1TR000454).

References

  • **1.Initiation of Antiretroviral Therapy in Early Asymptomatic HIV Infection. New England Journal of Medicine. 2015;373(9):795–807. doi: 10.1056/NEJMoa1506816. The START study demonstrated that initiation of cART regardless of CD4 cell count was beneficial over delayed initiation of therapy, and has significant implications for clinical practice. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Gunthard HF, Aberg JA, Eron JJ, et al. Antiretroviral treatment of adult HIV infection: 2014 recommendations of the International Antiviral Society-USA Panel. Jama. 2014;312(4):410–25. doi: 10.1001/jama.2014.8722. [DOI] [PubMed] [Google Scholar]
  • 3.World Health Orgaization. Guideline on when to start antiretroviral therapy and on pre-exposure prophylaxis for HIV. 2015 [updated September 2015; cited 2015 October 30, 2015]. Available from: http://www.who.int/hiv/pub/guidelines/earlyrelease-arv/en/ [PubMed]
  • 4.Harrison KM, Song R, Zhang X. Life expectancy after HIV diagnosis based on national HIV surveillance data from 25 states, United States. Journal of acquired immune deficiency syndromes. 2010;53(1):124–30. doi: 10.1097/QAI.0b013e3181b563e7. [DOI] [PubMed] [Google Scholar]
  • 5.Aberg JA. Aging, inflammation, and HIV infection. Topics in antiviral medicine. 2012;20(3):101–5. [PMC free article] [PubMed] [Google Scholar]
  • *6.Smit M, Brinkman K, Geerlings S, et al. Future challenges for clinical care of an ageing population infected with HIV: a modelling study. The Lancet Infectious diseases. 2015;15(7):810–8. doi: 10.1016/S1473-3099(15)00056-0. This informative modelling study that looks at the challenges that will be faced in caring for an ageing HIV-infected population including non-AIDS comorbidities and polypharmacy. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • *7.Rey D, Treger M, Sibilia J, et al. Bone mineral density changes after 2 years of ARV treatment, compared to naive HIV-1-infected patients not on HAART. Infectious diseases (London, England) 2015;47(2):88–95. doi: 10.3109/00365548.2014.968610. This prospective study comparing HIV-infected patients initiating cART with those not on therapy demonstrates the impact of cART on bone mineral density loss in HIV-infected patients. [DOI] [PubMed] [Google Scholar]
  • 8.Ofotokun I, McIntosh E, Weitzmann MN. HIV: inflammation and bone. Current HIV/AIDS reports. 2012;9(1):16–25. doi: 10.1007/s11904-011-0099-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.McComsey GA, Tebas P, Shane E, et al. Bone disease in HIV infection: a practical review and recommendations for HIV care providers. Clinical infectious diseases: an official publication of the Infectious Diseases Society of America. 2010;51(8):937–46. doi: 10.1086/656412. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Finkelstein JS, Brockwell SE, Mehta V, et al. Bone mineral density changes during the menopause transition in a multiethnic cohort of women. The Journal of clinical endocrinology and metabolism. 2008;93(3):861–8. doi: 10.1210/jc.2007-1876. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Bolland MJ, Wang TK, Grey A, et al. Stable bone density in HAART-treated individuals with HIV: a meta-analysis. The Journal of clinical endocrinology and metabolism. 2011;96(9):2721–31. doi: 10.1210/jc.2011-0591. [DOI] [PubMed] [Google Scholar]
  • 12.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 medicine. 2011;12(3):157–65. doi: 10.1111/j.1468-1293.2010.00864.x. [DOI] [PubMed] [Google Scholar]
  • 13.Yin MT, Kendall MA, Wu X, et al. Fractures after antiretroviral initiation. AIDS (London, England) 2012;26(17):2175–84. doi: 10.1097/QAD.0b013e328359a8ca. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Brown TT, McComsey GA, King MS, et al. Loss of bone mineral density after antiretroviral therapy initiation, independent of antiretroviral regimen. Journal of acquired immune deficiency syndromes (1999) 2009;51(5):554–61. doi: 10.1097/QAI.0b013e3181adce44. [DOI] [PubMed] [Google Scholar]
  • 15.Briot K, Kolta S, Flandre P, et al. Prospective one-year bone loss in treatment-naive HIV+ men and women on single or multiple drug HIV therapies. Bone. 2011;48(5):1133–9. doi: 10.1016/j.bone.2011.01.015. [DOI] [PubMed] [Google Scholar]
  • 16.Brown TT, Qaqish RB. Antiretroviral therapy and the prevalence of osteopenia and osteoporosis: a meta-analytic review. AIDS (London, England) 2006;20(17):2165–74. doi: 10.1097/QAD.0b013e32801022eb. [DOI] [PubMed] [Google Scholar]
  • 17.Triant VA, Brown TT, Lee H, et al. Fracture prevalence among human immunodeficiency virus (HIV)-infected versus non-HIV-infected patients in a large U.S. healthcare system. The Journal of clinical endocrinology and metabolism. 2008;93(9):3499–504. doi: 10.1210/jc.2008-0828. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.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. Clinical infectious diseases: an official publication of the Infectious Diseases Society of America. 2011;52(8):1061–8. doi: 10.1093/cid/ciq242. [DOI] [PubMed] [Google Scholar]
  • 19.Womack JA, Goulet JL, Gibert C, et al. Increased risk of fragility fractures among HIV infected compared to uninfected male veterans. PloS one. 2011;6(2):e17217. doi: 10.1371/journal.pone.0017217. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • *20.Sharma A, Shi Q, Hoover DR, et al. Increased Fracture Incidence in Middle-Aged HIV-Infected and HIV-Uninfected Women: Updated Results From the Women’s Interagency HIV Study. Journal of acquired immune deficiency syndromes (1999) 2015;70(1):54–61. doi: 10.1097/QAI.0000000000000674. This follow-up study of the WIHS cohort, of which a prior study did not demonstrate an increased risk of fracture among HIV-infected women despite lower BMD, has now demonstrated that these women are at increased risk of fracture. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Guerri-Fernandez R, Vestergaard P, Carbonell C, et al. HIV infection is strongly associated with hip fracture risk, independently of age, gender, and comorbidities: a population-based cohort study. Journal of bone and mineral research: the official journal of the American Society for Bone and Mineral Research. 2013;28(6):1259–63. doi: 10.1002/jbmr.1874. [DOI] [PubMed] [Google Scholar]
  • **22.Prieto-Alhambra D, Guerri-Fernandez R, De Vries F, et al. HIV infection and its association with an excess risk of clinical fractures: a nationwide case-control study. Journal of acquired immune deficiency syndromes (1999) 2014;66(1):90–5. doi: 10.1097/QAI.0000000000000112. This large case-control study clearly demonstrates that HIV infection is an independent risk factor for fracture when controlling for traditional risk factors. [DOI] [PubMed] [Google Scholar]
  • *23.Battalora L, Buchacz K, Armon C, et al. Low bone mineral density and risk of incident fracture in HIV-infected adults. Antiviral therapy. 2015 doi: 10.3851/IMP2979. This study associates osteoporosis as diagnosed with DXA scan with an increased risk for fracture in a young population of HIV-infected adults. [DOI] [PubMed] [Google Scholar]
  • 24.Hui SL, Slemenda CW, Johnston CC., Jr Age and bone mass as predictors of fracture in a prospective study. The Journal of clinical investigation. 1988;81(6):1804–9. doi: 10.1172/JCI113523. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Stellbrink HJ, Orkin C, Arribas JR, 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. Clinical infectious diseases: an official publication of the Infectious Diseases Society of America. 2010;51(8):963–72. doi: 10.1086/656417. [DOI] [PubMed] [Google Scholar]
  • 26.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. Journal of Infectious Diseases. 2011;203(12):1791–801. doi: 10.1093/infdis/jir188. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Huang JS, Hughes MD, Riddler SA, et al. Bone Mineral Density Effects of Randomized Regimen and Nucleoside Reverse Transcriptase Inhibitor (NRTI) Selection from ACTG A5142. HIV clinical trials. 2013;14(5):224–34. doi: 10.1310/hct1405-224. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • *28.Mulligan K, Glidden DV, Anderson PL, et al. Effects of Emtricitabine/Tenofovir on Bone Mineral Density in HIV-Negative Persons in a Randomized, Double-Blind, Placebo-Controlled Trial. Clinical infectious diseases: an official publication of the Infectious Diseases Society of America. 2015;61(4):572–80. doi: 10.1093/cid/civ324. This study showed that tenofovir use is associated with BMD loss in the absence of HIV infection. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Brown TT, Ross AC, Storer N, et al. Bone turnover, osteoprotegerin/RANKL and inflammation with antiretroviral initiation: tenofovir versus non-tenofovir regimens. Antiviral therapy. 2011;16(7):1063–72. doi: 10.3851/IMP1874. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • *30.Wohl DA, Bhatti L, Small CB, et al. The ASSURE study: HIV-1 suppression is maintained with bone and renal biomarker improvement 48 weeks after ritonavir discontinuation and randomized switch to abacavir/lamivudine + atazanavir. HIV medicine. 2016;17(2):106–17. doi: 10.1111/hiv.12281. Switching from tenofovir to abacavir results in improved BMD in virologically suppressed HIV-infected patients. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • **31.Kinai E, Nishijima T, Mizushima D, et al. Long-term use of protease inhibitors is associated with bone mineral density loss. AIDS research and human retroviruses. 2014;30(6):553–9. doi: 10.1089/aid.2013.0252. This cohort study associates duration of PI use with low BMD, and discontinuation of PI with improvement in BMD, indicating that discontinuation of a PI may be a useful strategy for HIV-infected patients with osteoporosis. [DOI] [PubMed] [Google Scholar]
  • 32.Tebas P, Powderly WG, Claxton S, et al. Accelerated bone mineral loss in HIV-infected patients receiving potent antiretroviral therapy. AIDS (London, England) 2000;14(4):F63–7. doi: 10.1097/00002030-200003100-00005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Zuccotti G, Vigano A, Gabiano C, et al. Antiretroviral therapy and bone mineral measurements in HIV-infected youths. Bone. 2010;46(6):1633–8. doi: 10.1016/j.bone.2010.02.029. [DOI] [PubMed] [Google Scholar]
  • 34.Grant PM, Kitch D, McComsey GA, et al. Low baseline CD4+ count is associated with greater bone mineral density loss after antiretroviral therapy initiation. Clinical infectious diseases: an official publication of the Infectious Diseases Society of America. 2013;57(10):1483–8. doi: 10.1093/cid/cit538. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • *35.Kooij KW, Wit FW, Bisschop PH, et al. Low bone mineral density in patients with well-suppressed HIV infection: association with body weight, smoking, and prior advanced HIV disease. The Journal of infectious diseases. 2015;211(4):539–48. doi: 10.1093/infdis/jiu499. This cohort study suggests that among older, well-controlled HIV-infected patients, HIV infection is no longer an independent predictor of low BMD when controlling for low BMI and smoking. [DOI] [PubMed] [Google Scholar]
  • 36.Bedimo R, Maalouf NM, Zhang S, et al. Osteoporotic fracture risk associated with cumulative exposure to tenofovir and other antiretroviral agents. AIDS (London, England) 2012;26(7):825–31. doi: 10.1097/QAD.0b013e32835192ae. [DOI] [PubMed] [Google Scholar]
  • 37.Rockstroh JK, DeJesus E, Henry K, et al. A randomized, double-blind comparison of coformulated elvitegravir/cobicistat/emtricitabine/tenofovir DF vs ritonavir-boosted atazanavir plus coformulated emtricitabine and tenofovir DF for initial treatment of HIV-1 infection: analysis of week 96 results. Journal of acquired immune deficiency syndromes (1999) 2013;62(5):483–6. doi: 10.1097/QAI.0b013e318286415c. [DOI] [PubMed] [Google Scholar]
  • **38.Brown TT, Moser C, Currier JS, 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. doi: 10.1093/infdis/jiv194. The ACTG substudy 5260s demonstrated loss of BMD in all regimens studied, but a greater loss in PI-containing arms compared with the raltegravir-containing arm, further implicating PIs in cART-induced bone loss. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Bonnet E, Ruidavets JB, Genoux A, et al. Early loss of bone mineral density is correlated with a gain of fat mass in patients starting a protease inhibitor containing regimen: the prospective Lipotrip study. BMC infectious diseases. 2013;13:293. doi: 10.1186/1471-2334-13-293. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Duvivier C, Kolta S, Assoumou L, 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 (London, England) 2009;23(7):817–24. doi: 10.1097/QAD.0b013e328328f789. [DOI] [PubMed] [Google Scholar]
  • *41.Moyle GJ, Hardy H, Farajallah A, et al. Changes in bone mineral density after 96 weeks of treatment with atazanavir/ritonavir or lopinavir/ritonavir plus tenofovir DF/emtricitabine in treatment-naive patients with HIV-1 infection: the CASTLE body composition substudy. Journal of acquired immune deficiency syndromes (1999) 2015;68(1):40–5. doi: 10.1097/QAI.0000000000000383. A greater BMD loss was observed in men treated with lopinavir/ritonavir compared with men treated with atazanavir/ritonavir, indicating that there differences among PIs with regard to their effects on bone, as well as possible sex differences in bone effects of PI therapy. [DOI] [PubMed] [Google Scholar]
  • 42.Tebas P, Umbleja T, Dube MP, et al., editors. Initiation of ART is associated with bone loss independent of the specific ART regimen. The results of ACTG A5005s. 14th Conference on Retroviruses and Opportunistic Infections; 2007; Los Angeles, CA. [Google Scholar]
  • 43.Curran A, Martinez E, Saumoy M, et al. Body composition changes after switching from protease inhibitors to raltegravir: SPIRAL-LIP substudy. AIDS (London, England) 2012;26(4):475–81. doi: 10.1097/QAD.0b013e32834f3507. [DOI] [PubMed] [Google Scholar]
  • 44.Melton LJ, 3rd, Looker AC, Shepherd JA, et al. Osteoporosis assessment by whole body region vs. site-specific DXA. Osteoporosis international: a journal established as result of cooperation between the European Foundation for Osteoporosis and the National Osteoporosis Foundation of the USA. 2005;16(12):1558–64. doi: 10.1007/s00198-005-1871-y. [DOI] [PubMed] [Google Scholar]
  • *45.Guaraldi G, Zona S, Cossarizza A, et al. Switching to darunavir/ritonavir monotherapy vs. triple-therapy on body fat redistribution and bone mass in HIV-infected adults: the Monarch randomized controlled trial. International journal of STD & AIDS. 2014;25(3):207–12. doi: 10.1177/0956462413497701. This study shows that elimination of NRTIs in virologically suppressed patients previously treated with triple-therapy cART results in improvement in bone density despite continuation of a PI. [DOI] [PubMed] [Google Scholar]
  • *46.Hamzah L, Tiraboschi JM, Iveson H, et al. Effects on vitamin D, bone and the kidney of switching from fixed-dose tenofovir disoproxil fumarate/emtricitabine/efavirenz to darunavir/ritonavir monotherapy: a randomized, controlled trial (MIDAS) Antiviral therapy. 2015 doi: 10.3851/IMP3000. Switching from TDF/FTC/EFV to DRV/r monotherpay results in improved serum vitamin D levels, improved biomarkers of bone turnover, and improved BMD. [DOI] [PubMed] [Google Scholar]
  • **47.Haskelberg H, Mallon PW, Hoy J, et al. Bone mineral density over 96 weeks in adults failing first-line therapy randomized to raltegravir/lopinavir/ritonavir compared with standard second-line therapy. Journal of acquired immune deficiency syndromes (1999) 2014;67(2):161–8. doi: 10.1097/QAI.0000000000000288. This study shows that among viremic patients who have failed first line theray and are initiating second line therapy with lopinavir/ritonavir, replacement of tenofovir with raltegravir results in less BMD loss, suggesting that PIs interact with tenofovir in causing bone loss. [DOI] [PubMed] [Google Scholar]
  • *48.Bedimo RJ, Drechsler H, Jain M, et al. The RADAR study: week 48 safety and efficacy of RAltegravir combined with boosted DARunavir compared to tenofovir/emtricitabine combined with boosted darunavir in antiretroviral-naive patients. Impact on bone health. PloS one. 2014;9(8):e106221. doi: 10.1371/journal.pone.0106221. The RADAR study demonstrated less BMD loss in the NRTI-sparing arm, although there were more virologic failures in the darunavir/ritonavir + raltegravir arm. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Martin A, Moore C, Mallon PW, et al. Bone mineral density in HIV participants randomized to raltegravir and lopinavir/ritonavir compared with standard second line therapy. AIDS (London, England) 2013;27(15):2403–11. doi: 10.1097/01.aids.0000432534.47217.b4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Bloch M, Tong WW, Hoy J, et al. Switch from tenofovir to raltegravir increases low bone mineral density and decreases markers of bone turnover over 48 weeks. HIV medicine. 2014;15(6):373–80. doi: 10.1111/hiv.12123. [DOI] [PubMed] [Google Scholar]
  • 51.Kearney BP, Mathias A, Mittan A, et al. Pharmacokinetics and safety of tenofovir disoproxil fumarate on coadministration with lopinavir/ritonavir. Journal of acquired immune deficiency syndromes (1999) 2006;43(3):278–83. doi: 10.1097/01.qai.0000243103.03265.2b. [DOI] [PubMed] [Google Scholar]
  • *52.Baxi SM, Greenblatt RM, Bacchetti P, et al. Common clinical conditions - age, low BMI, ritonavir use, mild renal impairment - affect tenofovir pharmacokinetics in a large cohort of HIV-infected women. AIDS (London, England) 2014;28(1):59–66. doi: 10.1097/QAD.0000000000000033. This study using the WIHS cohort demonstrated the real-world impact of the combining tenofovir and ritonavir on tenofovir pharmacokinetics. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Pruvost A, Negredo E, Theodoro F, et al. Pilot pharmacokinetic study of human immunodeficiency virus-infected patients receiving tenofovir disoproxil fumarate (TDF): investigation of systemic and intracellular interactions between TDF and abacavir, lamivudine, or lopinavir-ritonavir. Antimicrobial agents and chemotherapy. 2009;53(5):1937–43. doi: 10.1128/AAC.01064-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Cihlar T, Ray AS, Laflamme G, et al. Molecular assessment of the potential for renal drug interactions between tenofovir and HIV protease inhibitors. Antiviral therapy. 2007;12(2):267–72. [PubMed] [Google Scholar]
  • 55.Gutierrez F, Fulladosa X, Barril G, et al. Renal tubular transporter-mediated interactions of HIV drugs: implications for patient management. AIDS reviews. 2014;16(4):199–212. [PubMed] [Google Scholar]
  • 56.German P, Warren D, West S, et al. Pharmacokinetics and bioavailability of an integrase and novel pharmacoenhancer-containing single-tablet fixed-dose combination regimen for the treatment of HIV. Journal of acquired immune deficiency syndromes (1999) 2010;55(3):323–9. doi: 10.1097/QAI.0b013e3181eb376b. [DOI] [PubMed] [Google Scholar]
  • 57.Panel on Antiretroviral Guidelines for Adults and Adolescents. Guidelines for the use of antiretroviral agents in HIV-1-infected adults and adolescents. Department of Health and Human Services; 2015. [cited 2016 January 27, 2016]. Available from: http://aidsinfo.nih.gov/contentfiles/lvguidelines/AdultandAdolescentGL.pdf. [Google Scholar]
  • **58.Brown TT, Hoy J, Borderi M, et al. Recommendations for evaluation and management of bone disease in HIV. Clinical infectious diseases: an official publication of the Infectious Diseases Society of America. 2015;60(8):1242–51. doi: 10.1093/cid/civ010. This paper provides evidence-based guidance for the evaluation and management of HIV-infected patients with bone disease, including recommendations for choosing cART regimens in this population. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Hernandez-Vallejo SJ, Beaupere C, Larghero J, et al. HIV protease inhibitors induce senescence and alter osteoblastic potential of human bone marrow mesenchymal stem cells: beneficial effect of pravastatin. Aging cell. 2013;12(6):955–65. doi: 10.1111/acel.12119. [DOI] [PubMed] [Google Scholar]
  • 60.Wang MWH, Shi W, Faccio R, et al. The HIV protease inhibitor ritonavir blocks osteoclastogenesis and function by impairing RANKL-induced signaling. Journal of Clinical Investigation. 2004;114(2):206–13. doi: 10.1172/JCI15797. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Cotter AG, Mallon PW. The effects of untreated and treated HIV infection on bone disease. Current opinion in HIV and AIDS. 2014;9(1):17–26. doi: 10.1097/COH.0000000000000028. [DOI] [PubMed] [Google Scholar]
  • 62.Gibellini D, Borderi M, de Crignis E, et al. Analysis of the effects of specific protease inhibitors on OPG/RANKL regulation in an osteoblast-like cell line. The new microbiologica. 2010;33(2):109–15. [PubMed] [Google Scholar]
  • 63.Fakruddin JM, Laurence J. HIV envelope gp120-mediated regulation of osteoclastogenesis via receptor activator of nuclear factor kappa B ligand (RANKL) secretion and its modulation by certain HIV protease inhibitors through interferon-gamma/RANKL cross-talk. The Journal of biological chemistry. 2003;278(48):48251–8. doi: 10.1074/jbc.M304676200. [DOI] [PubMed] [Google Scholar]
  • 64.Modarresi R, Xiang Z, Yin M, et al. WNT/beta-catenin signaling is involved in regulation of osteoclast differentiation by human immunodeficiency virus protease inhibitor ritonavir: relationship to human immunodeficiency virus-linked bone mineral loss. The American journal of pathology. 2009;174(1):123–35. doi: 10.2353/ajpath.2009.080484. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Malizia AP, Cotter E, Chew N, et al. HIV protease inhibitors selectively induce gene expression alterations associated with reduced calcium deposition in primary human osteoblasts. AIDS research and human retroviruses. 2007;23(2):243–50. doi: 10.1089/aid.2006.0084. [DOI] [PubMed] [Google Scholar]
  • 66.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. Osteoporosis international: a journal established as result of cooperation between the European Foundation for Osteoporosis and the National Osteoporosis Foundation of the USA. 2011;22(5):1459–68. doi: 10.1007/s00198-010-1363-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Santiago F, Oguma J, Brown AM, et al. Noncanonical Wnt signaling promotes osteoclast differentiation and is facilitated by the human immunodeficiency virus protease inhibitor ritonavir. Biochemical and biophysical research communications. 2012;417(1):223–30. doi: 10.1016/j.bbrc.2011.11.089. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • **68.Ofotokun I, Titanji K, Vunnava A, et al. Antiretroviral therapy induces a rapid increase in bone resorption that is positively associated with the magnitude of immune reconstitution in HIV infection. AIDS (London, England) 2016;30(3):405–14. doi: 10.1097/QAD.0000000000000918. This clinical trial provides evidence that immune reconstitution is at least partly responsible for the decrease in BMD observed after initiation of cART, and provides a mechanism that explains why this observation is seen across all cART regimens. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Franco JM, Rubio A, Martinez-Moya M, et al. T-cell repopulation and thymic volume in HIV-1-infected adult patients after highly active antiretroviral therapy. Blood. 2002;99(10):3702–6. doi: 10.1182/blood.v99.10.3702. [DOI] [PubMed] [Google Scholar]
  • *70.Ofotokun I, Titanji K, Vikulina T, et al. Role of T-cell reconstitution in HIV-1 antiretroviral therapy-induced bone loss. Nature communications. 2015;6:8282. doi: 10.1038/ncomms9282. This animal model provides evidence that T-cell reconstitution results in decreased BMD and may explain the mechanism by which cART results in bone loss. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Reid IR, Bolland MJ, Grey A. Effects of vitamin D supplements on bone mineral density: a systematic review and meta-analysis. Lancet (London, England) 2014;383(9912):146–55. doi: 10.1016/S0140-6736(13)61647-5. [DOI] [PubMed] [Google Scholar]
  • *72.Hidron AI, Hill B, Guest JL, et al. Risk factors for vitamin D deficiency among veterans with and without HIV infection. PloS one. 2015;10(4):e0124168. doi: 10.1371/journal.pone.0124168. This study demonstrates that HIV infection is a risk factor for vitamin D deficiency beyond traditional risk factors among U.S. veterans. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Cervero M, Agud JL, Garcia-Lacalle C, et al. Prevalence of vitamin D deficiency and its related risk factor in a Spanish cohort of adult HIV-infected patients: effects of antiretroviral therapy. AIDS research and human retroviruses. 2012;28(9):963–71. doi: 10.1089/AID.2011.0244. [DOI] [PubMed] [Google Scholar]
  • 74.Havens PL, Stephensen CB, Hazra R, et al. Vitamin D3 decreases parathyroid hormone in HIV-infected youth being treated with tenofovir: a randomized, placebo-controlled trial. Clinical infectious diseases: an official publication of the Infectious Diseases Society of America. 2012;54(7):1013–25. doi: 10.1093/cid/cir968. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • **75.Overton ET, Chan ES, Brown TT, et al. Vitamin D and Calcium Attenuate Bone Loss With Antiretroviral Therapy Initiation: A Randomized Trial. Annals of internal medicine. 2015;162(12):815–24. doi: 10.7326/M14-1409. This trial demonstrates that supplementation with high-dose vitamin D and calcium can attenuate bone loss in cART-naïve patients initiating tenofovir/emtricitabine/efavirenz, and provides a possible clinical strategy for mitigating bone loss in HIV-infected patients. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Cozzolino M, Vidal M, Arcidiacono MV, et al. HIV-protease inhibitors impair vitamin D bioactivation to 1,25-dihydroxyvitamin D. AIDS (London, England) 2003;17(4):513–20. doi: 10.1097/00002030-200303070-00006. [DOI] [PubMed] [Google Scholar]
  • 77.Kim JH, Gandhi V, Psevdos G, Jr, et al. Evaluation of vitamin D levels among HIV-infected patients in New York City. AIDS research and human retroviruses. 2012;28(3):235–41. doi: 10.1089/AID.2011.0040. [DOI] [PubMed] [Google Scholar]
  • 78.Cervero M, Agud JL, Torres R, et al. Higher vitamin D levels in HIV-infected out-patients on treatment with boosted protease inhibitor monotherapy. HIV medicine. 2013;14(9):556–62. doi: 10.1111/hiv.12049. [DOI] [PubMed] [Google Scholar]
  • 79.Koga I, Seo K, Yoshino Y, et al. Increase of 25-hydroxyvitamin D levels after initiation of combination antiretroviral therapy. Journal of infection and chemotherapy: official journal of the Japan Society of Chemotherapy. 2015;21(10):737–41. doi: 10.1016/j.jiac.2015.07.003. [DOI] [PubMed] [Google Scholar]
  • 80.Pepe J, Mezzaroma I, Fantauzzi A, et al. An oral high dose of cholecalciferol restores vitamin D status in deficient postmenopausal HIV-1-infected women independently of protease inhibitors therapy: a pilot study. Endocrine. 2015 doi: 10.1007/s12020-015-0693-8. [DOI] [PubMed] [Google Scholar]

RESOURCES