Next Article in Journal
Experimental Setups for In Vitro Studies on Radon Exposure in Mammalian Cells—A Critical Overview
Next Article in Special Issue
Community Perceptions of HIV Stigma, Discriminatory Attitudes, and Disclosure Concerns: A Health Facility-Based Study in Selected Health Districts of South Africa
Previous Article in Journal / Special Issue
Barriers and Facilitators to Pre-Exposure Prophylaxis by Men Who Have Sex with Men and Community Stakeholders in Malaysia
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Systematic Review

A Systematic Review and Meta-Analysis on the Impact of Statin Treatment in HIV Patients on Antiretroviral Therapy

by
Kabelo Mokgalaboni
1,*,
Wendy Nokhwezi Phoswa
1,
Samantha Yates
1,
Sogolo Lucky Lebelo
1,
Sphiwe Madiba
2 and
Perpetua Modjadji
3
1
Department of Life and Consumer Sciences, College of Agriculture and Environmental Sciences, University of South Africa, Florida Campus, Johannesburg 1709, South Africa
2
Faculty of Health Sciences, University of Limpopo, Polokwane 0700, South Africa
3
Non-Communicable Disease Research Unit, South African Medical Research Council, Cape Town 7505, South Africa
*
Author to whom correspondence should be addressed.
Int. J. Environ. Res. Public Health 2023, 20(9), 5668; https://doi.org/10.3390/ijerph20095668
Submission received: 10 March 2023 / Revised: 21 April 2023 / Accepted: 25 April 2023 / Published: 27 April 2023

Abstract

:
The rate of new human immunodeficiency virus (HIV) infections globally is alarming. Although antiretroviral therapy (ART) improves the quality of life among this group of patients, ARTs are associated with risk of cardiovascular diseases (CVD). Moreover, virally suppressed patients still experience immune activation associated with HIV migration from reservoir sites. Statins are widely recommended as therapeutic agents to control ART-related CVD; however, their impacts on the cluster of differentiation (CD)4 count and viral load are inconsistent. To assess the effect of statins on markers of HIV infections, immune activation and cholesterol, we thoroughly reviewed evidence from randomised controlled trials. We found 20 relevant trials from three databases with 1802 people living with HIV (PLHIV) on statin–placebo treatment. Our evidence showed no significant effect on CD4 T-cell count standardised mean difference (SMD): (−0.59, 95% confidence intervals (CI): (−1.38, 0.19), p = 0.14) following statin intervention in PLHIV on ART. We also found no significant difference in baseline CD4 T-cell count (SD: (−0.01, 95%CI: (−0.25, 0.23), p = 0.95). Our findings revealed no significant association between statins and risk of viral rebound in PLHIV with undetectable viral load risk ratio (RR): (1.01, 95% CI: (0.98, 1.04), p = 0.65). Additionally, we found a significant increase in CD8+CD38+HLA-DR+ T-cells (SMD (1.10, 95% CI: (0.93, 1.28), p < 0.00001) and CD4+CD38+HLA-DR+ T-cells (SMD (0.92, 95% CI: (0.32, 1.52), p = 0.003). Finally, compared to placebo, statins significantly reduced total cholesterol (SMD: (−2.87, 95% CI: (−4.08, −1.65), p < 0.0001)). Our results suggest that the statin lipid-lowering effect in PLHIV on ART may elevate immune activation without influencing the viral load and CD4 count. However, due to the limited evidence synthesised in this meta-analysis, we recommend that future powered trials with sufficient sample sizes evaluate statins’ effect on CD4 count and viral load, especially in virally suppressed patients.

1. Introduction

Human immunodeficiency virus (HIV) is a public health concern; according to the 2021 UNAIDS report, about 38.4 million people were living with HIV globally and 1.5 million new infections and 650,000 deaths from AIDS-related illnesses were recorded [1]. About 7.5 million children and adults in South Africa were living with HIV in 2021, with 210,000 new infections and 51,000 AIDS-related deaths reported [2]. This significant rate of new infections suggests an increasing need to combat this virus through prevention and treatment, respectively. This virus attacks the individual immune system, also known as cluster of differentiation 4 (CD4) counts, and pre-disposes infected individuals to various infections, including metabolic and acquired immune deficiency virus syndrome (AIDS) [3]. HIV has received enormous research attention since its outbreak; most recently, researchers have been exploring the bone marrow transplantation approach, which seems to be associated with the elimination of HIV reservoirs [4]. However, this is risky as it involves replacing individual bone marrow with HIV-resistant stem cells from a donor, implying that the patients will be susceptible to opportunistic infections during this procedure.
Nevertheless, the introduction and progress made with antiretroviral therapy (ART) have improved the quality of life for people living with HIV (PLHIV); the number of AIDS-related complications and deaths has since decreased in those that adhere to ART [5,6]. The fundamental goal of ART is to improve the immune system through increasing CD4 count and successfully suppressing HIV. This method stops the disease’s progression and reduces the risk and vulnerability to opportunistic infections. However, ART has reportedly been linked to an increased risk of cardiovascular disease (CVD) [7], as demonstrated by impaired lipid profile [8].
Additionally, kidney malfunctioning has been noted in other PLHIV on ART [9]. Therefore, due to the above secondary complications other patients are still reluctant to take ART as they show adversity in other HIV individuals. It is worth noting that PLHIV currently on ART are prescribed statins, a lipid-lowering treatment to control cholesterol and HIV-ART-related dyslipidaemia [10]. However, their impact, especially on viral load and CD4 count in PLHIV on ART, is unclear. This problem has prompted other researchers to conduct clinical trials investigating this paradigm. Surprisingly, researchers have reported contradicting findings, with some showing consistently improved HIV surrogate markers [11,12,13,14,15,16,17,18] and others showing negative findings [19,20,21,22,23]. HIV as a chronic infection is closely associated with inflammation due to the continuously rising viral load [24,25]. Interestingly, this seems to be controlled in PLHIV who are virally suppressed [26]. Therefore, any potential pharmaceutical agent that can target inflammatory pathways may be of interest when it comes to the development of drugs that can curb inflammation in PLHIV.
Although the effect of statins on inflammation is well documented in diabetes [27], this aspect is not clear in PLHIV who are prescribed ART [12,16,20,21]. We believe that amelioration of inflammation by statins may partly contribute to reducing the risk of CVD and, thus, improving the overall quality of health. Disappointingly, Nachega et al., 2012 [28] have highlighted a serious concern regarding the administration of ART with polypharmacy as this is associated with drug–drug interactions. This subsequently results in drug toxicity, poorer ART adherence, and loss of efficacy of the used ART in older PLHIV. The same sentiments were expressed by Potentelo et al. [29]. Therefore, all these concerns make us question whether statins are safe for PLHIV taking ART. We have noted a few literature reviews that evaluated data on the beneficial effects of statins against secondary complications in PLHIV [30,31,32]; however, the impact on surrogate markers of HIV infections and cholesterol is inconsistent. Therefore, the current meta-analysis sought to evaluate whether statin treatment in HIV patients improves or worsens the surrogate markers of HIV infection, focusing on CD4 count, viral load and markers of immune activation. Furthermore, we sought to evaluate the effect of statin treatment on cholesterol in virally suppressed patients.

2. Materials and Methods

2.1. Study Design and Registration

The current study was conducted and reported following the formatting guidelines recommended by preferred reporting items for systematic reviews and meta-analysis (PRISMA) guidelines [33]. Due to the study design, no institutional ethics approval and patient informed consent were required. Only evidence from randomised controlled trials (RCTs) was sought and analysed. The protocol that accompanies this review has been registered with the PROSPERO registry: CRD42023397964.

2.2. Search Strategy and Information Sources

Relevant RCTs were searched from inception until February 2023 and retrieved from MEDLINE, Scopus, and Cochrane Library, irrespective of the language, using the medical subject headings (MesH) terms or text words. The following search terms were applied: “statin” and synonyms such as “hydroxyl glutaryl (HMG-CoA) reductase inhibitors”, “atorvastatin”, “rosuvastatin”, “fluvastatin”, “lovastatin”, “pitavastatin”, “simvastatin” using OR as a Boolean AND “HIV” OR “AIDS” OR “human AND immunodeficiency” AND “Antiretroviral therapy” OR “ART”. Exact search strategies adapted to these databases are presented in Supplementary Table S1A–C.

2.3. Eligibility Criteria and Study Selection

All records obtained from these databases were exported to Mendeley Desktop software and duplicate records were excluded. Two researchers (K.M. and W.N.P.) screened the remaining records independently and disagreements were resolved by consensus. Retrieved full-text articles were thoroughly evaluated for eligibility and subjected to meta-analysis if they met all the following inclusion criteria: (i) human studies; (ii) PLHIV on ART; and (iii) treated with any form of statin. In contrast, the following types of studies were all excluded: (i) animal models; (ii) without control; (iii) comparative trials; (iv) PLHIV not on ART or ART treated PLHIV not on statin; and (v) studies not reporting any of the primary markers of HIV infections and cholesterol.

2.4. Data Extraction and Quality Assessment

Two researchers (K.M. and W.N.P.) independently extracted information from relevant studies using a pre-defined data extraction form. Briefly, the following data items were extracted from each of the studies: author’s surname and year of publication; study design, population states, and sample size in each group; the country where the study was conducted; dosage form of statin and duration of intervention; age and sex of study participants; surrogate markers evaluated; and general findings. Disagreements in data extractions were resolved by discussion. K.M. and W.N.P. independently evaluated the quality of included RCTs using a modified JADAD scale [34]. This tool focuses on these four main domains: (i) description of randomisation; (ii) accuracy of double blinding; (iii) description of patient loss or exclusion; and (iv) adequacy of randomisation. Any disagreement was resolved by inviting the third researcher, P.M. and re-evaluating the study and domains in dispute.

2.5. Data Synthesis and Analysis

All analyses were conducted using RevMan 5.4 software (Review Manager (RevMan), version 5.4.1, The Cochrane Collaboration, 2020) and metaHUN, a web tool accessible at http://softmed.hacettepe.edu.tr/metaHUN/ (accessed on 24 February 2023). Effect size from all outcomes for continuous data was estimated by computing each study’s mean, standard deviation (SD), and sample size. Mean and SD were estimated following guidelines [29] in cases where the included study reported only mean and interquartile range (IQR); when the standard error of the mean (SER) was given, SD was estimated using SD = SER × √n [35]. For other outcomes, we extracted or estimated a change in mean and SD from baseline and post-test results using the equation (∆mean = (mf − mb)). We further estimated change in SD using the following formula: Δ SD = ( SD b ) 2 + ( SD f ) 2 2 ( r × SD b × SD f ) ; this formula was outlined by Cochrane guidelines and other researchers’ findings, with a correlation coefficient (r) of 0.7 used in both groups [36,37]. Statistical heterogeneity was estimated using the I2 and Cochran Q tests [38]. I2 values below 50% and above 75% were classified as minimal and substantial statistical heterogeneity, respectively. Random-effect and fixed models were used in the presence and absence of statistical heterogeneity, respectively. We performed sub-group analyses to determine the sources of clinical heterogeneity across studies [39]. Our sub-group analyses explored the impact of statin and statin doses. The funnel plots were used to assess publication bias. We further conducted a sensitivity analysis to evaluate the stability of our result [40].

3. Results

3.1. Search, General Characteristics and Quality of Included Trials

The comprehensive search for the literature from these databases yielded 795 records that were screened and evaluated at the title, abstract and keywords level (Figure 1). Finally, 20 studies [12,13,14,16,19,20,21,22,23,41,42,43,44,45,46,47,48,49,50,51,52] were included, of which 12 trials used rosuvastatin of either 10, 20 or 80 mg, four used atorvastatin of 10, 30 and 40 mg, four used pravastatin at 40 mg and one used pitavastatin of 2 mg. These studies were published in peer-reviewed journals between 2004 and 2021, thus tracing old and advanced evidence on statins in PLHIV taking ART. Of 1802 PLHIV taking ART, 953 people taking a statin and 871 taking a placebo treatment were included. About 760 (80%) participants on statin treatment were males. The overall mean and SD ages of participants on statin treatment were 47.48 ± 4.96 years (Table 1). Publications of these trials were distributed across seven countries. Briefly, 12 studies [14,22,23,42,43,45,46,47,49,50,52,53] were conducted in the United States of America, three studies [12,21,44] were conducted in Australia, two [16,20] were conducted in Switzerland, and one study was conducted in each of the following countries, China [19], France [41] and Uganda [48]. For all four domains, we found the quality to be good: domain one received an overall score of 20 out of a possible score of 21, domain two scored 18 out of 21, and domains three and four received at least 14 out of 21. Across individual studies, about two were rated fair as they scored at least two points, ten rated good with a score of about three and, lastly, eight studies scored four points and were rated excellent (Supplementary Table S2).

3.2. Effect of Statin on Markers of HIV Infection

3.2.1. The Effect of Statin on Baseline CD4 T-Cell Count in HIV Patients on Antiretroviral Therapy (ART)

All included trials [12,13,14,16,19,20,21,22,23,41,42,43,44,45,46,47,48,49,50,51,52] investigated baseline CD4 counts in PLHIV taking both ART and statin treatments. Following our meta-analysis of the collected data, we found no significant difference in CD4 counts between PLHIV taking a statin and those taking a placebo, with a standardised mean difference (SMD) (−0.01), 95% CI: (−0.25, 0.23), p = 0.95). Notably, statistical heterogeneity was high (I2 = 83%) (Supplementary Figure S1).
We also collected data on CD4 T-cell counts to find the effect of statins following the intervention period. Only three trials [12,20,41] reported sufficient data on CD4 T-cells or data to estimate the change in CD4 T-cell counts. The overall pooled effect estimates showed comparable results between PLHIV taking a statin and those taking a placebo (SMD (−0.59), 95% CI: (−1.38, 0.19), p = 0.14). Similarly, there was a high statistical heterogeneity (I2 = 72%) among the analysed evidence (Figure 2). We further assessed the magnitude of the effect of statin and found that, according to Cohens d interpretation, the statin in PLHIV on ART might have a medium suppressive effect (SMD > 0.5) on CD4 T-cell count.

3.2.2. Effect of Statin on CD8+, and CD4+CD38+HLA-DR+ Cells

We evaluated the effect of statins on markers of immune activation. Evidence from five trials revealed a significant increase in CD8+CD38+HLA-DR+ T-cells in PLHIV taking ART who were also treated with a statin (SMD (1.10), 95%CI: (0.93, 1.28), p < 0.00001). Interestingly, no evidence of heterogeneity was observed (I2 = 0%) (Figure 3A). Similarly, there was a significant increase in CD4+CD38+HLA-DR+ T-cells (SMD (0.92), 95% CI: (0.32, 1.52), p = 0.003) (Figure 3B).

3.2.3. Effect of Statin on Undetectable HIV Load (<50 HIV RNA Copy)

Sixteen RCTs [13,14,16,19,21,22,23,42,44,45,46,47,49,50,51,52] evaluated the effect of statin on undetectable viral copies in HIV patients taking ART. Our overall effect estimates showed a non-significant risk of experiencing a viral rebound in PLHIV-ART with an undetectable viral load on statin treatment compared to placebo, with a risk ratio (RR) of (1.01, 95% CI: (0.98, 1.04), p = 0.65). This suggests that statin treatment in PLHIV with ART-suppressed viremia does not present a risk of viral duplication. Interestingly, there was no evidence of heterogeneity (I2 = 0%) among the analysed studies (Supplementary Figure S2).

3.3. Effect of Statin on Total Cholesterol in HIV Patients on ART

Total cholesterol (TC) data were extracted from nine studies [12,16,19,20,41,42,44,47,50] with 200 PLHIV taking statin treatments. The pooled effect estimates revealed a significant reduction in TC following statin treatment compared to placebo treatment (SMD (−2.87), 95% CI: (−4.08, −1.65), p < 0.00001) (Figure 4). However, we observed substantial heterogeneity amongst these studies (I2 = 95%).

3.4. Test for Sub-Group Differences

Due to substantial statistical heterogeneity observed in our effect estimates, we conducted a sub-group analysis to explore the sources of heterogeneity based on the doses and form of statins across all outcomes. Data supporting these are presented in a Supplementary File (Supplementary Figures S1–S6). Briefly, a test for sub-grouping differences according to the form of statin on baseline CD4 T-cell count showed an (SMD (−0.01), 95% CI:(−0.25, 0.21), I2 = 0%) (Supplementary Figure S3). This suggests that the observed heterogeneity may arise from the use of different forms of statin treatment. Due to the presence of only three studies, no sub-group was conducted on change in CD4 T-cell counts and CD4+CD38+HLA-DR+ T-cells. A sub-group difference according to statin doses on TC revealed an (SMD (−3.06), 95% CI: (−4.15, −1.98), I2 = 0%) (Supplementary Figure S4), while 40 mg of statin showed a pronounced decrease in TC (SMD (3.48), 95% CI: (−5.35, −1.62), I2 = 93%. Consistently, a decrease in TC was observed when the sub-group was performed according to the form of statin (SMD (−2.73) 95% CI: (−3.77, −1.70), I2 = 40.5%) (Supplementary Figure S5). This suggests that the statistical heterogeneity observed might not be due to the forms of statin used but associated with different doses used.

3.5. Sensitivity Analysis

Sensitivity analysis was used to evaluate the robustness of our findings and was conducted following a one-study exclusion approach. This method was based on the sample size of the study. After removing a study [41] with a small sample size, we found that the baseline CD4 T-cell count effect size changed to SMD (0.03), 95% CI: (−0.22, 0.27). However, this was statistically not significant (p = 0.82). For a change in CD4 T-cell count, we removed a study [20] with a large sample size and found that the effect size increased significantly SMD (−0.96), 95% CI: (−1.90, −0.03), p = 0.04; this finding was about 0.37 (37%) increase from the original effect size. Lastly, for TC the exclusion of the study by Bonnet et al. [41] due to the small sample size resulted in a minimal increase in TC, as demonstrated by SMD (−2.83), 95% CI: (−4.13, −1.53), which was a 5% decrease from original effect size. Removing a study by Nakanjako et al. [48] due to small sample size, our effect size changes significantly from original to CD8+CD38+HLA-DR+T-cells SMD (1.14), 95% CI (0.96, 1.32), p < 0.00001 and CD4+CD38+HLA-DR+ T-cell SMD (0.70), 95% CI: (0.10, 1.29), p = 0.02.

3.6. Publication Bias

We also evaluated publications using visual inspection of funnel plots and QQ-normal plots. At least four studies provided sufficient data about the change in the CD4 T-cell count level, while visually the funnel plot revealed a symmetrical shape, suggesting no evidence of publication bias (Figure 5A). Additionally, our Q-Q plot also showed that all studies were closer to the straight line, thus suggesting no publication bias (Figure 5B). On the other hand, we noticed evidence of publication bias across studies that reported on the level of total cholesterol, as indicated by an asymmetrically shaped funnel plot coupled with Egger’s regression test (Q = 22.3847, p = 0.0000) (Supplementary Figure S6).

4. Discussion

According to our knowledge, no meta-analysis explored the effect of statin on an ART-enhanced CD4 T-cell count, undetectable HIV-RNA, and markers of T-cell activation amongst ART-treated PLHIV co-supplemented with statins. In this study, we have explored the RCT evidence investigating the impact of statin on the CD4 T-cell count, undetectable HIV-RNA copy, markers of T-cell activation and TC level in PLHIV on ART. We found that in about 1802 PLHIV on ART, there were no significant effects of statin on either baseline or post-treatment CD4 T-cell count compared to placebo. We also found a significant elevation in markers of T-cell activation, with a focus on both CD8+ CD38+ and CD4+CD38+HLA-DR+ T-cells. Furthermore, we showed that statins in PLHIV treated with ART significantly reduce TC levels.
Although we had many trials in this analysis, pooled evidence from three studies with sufficient data on post-treatment CD4 T-cell count suggests that statin does not significantly impact CD4 T-cell count in virally suppressed patients. Our findings are supported by results obtained from previous trials that reported no significant increase in CD4 T-cell count following statin treatment in PLHIV on ART [22,48]. In contrast to our overall findings, a study by (Rodriguez et al., 2007) reported reduced CD4 T-cell gain following statin treatment in viremic-suppressed patients [11]. Long-term ART in HIV is associated with improved CD4 count; however, this depends on the level of CD4 count at the time of ART initiation, with low CD4 count failing to normalise even after prolonged ART administration [54]. On the other hand, in PLHIV who are ART-naïve rosuvastatin administration also revealed no significant increase in CD4 T-cell count [55]. This overall trend in CD4 T-cell counts suggests that statin may not effectively offer beneficial effects in improving CD4 count, especially in virally suppressed patients. Statins are known to have anti-inflammatory properties, while it has been reported that rosuvastatin significantly reduces markers of inflammation in HIV patients on ART [22]. Hearps et al. [56] support the sentiments of Funderberg [22]; however, they reported no normalisation of CD4 T-cell counts in the virally suppressed patients despite amelioration of inflammation. On the other hand, results from ART naïve PLHIV indicate that low serum cholesterol in children and adults is associated with reduced production of interleukin-2 and CD4 T-cell counts [57,58]. This suggests that the cholesterol-lowering effect of these statin therapies may induce CD4 T-cell reduction.
As demonstrated by a comparable level of suppressed viral load, our findings suggest that statin administration in virally suppressed patients is not associated with the risk of viral rebound. These results are corroborated by other researchers who have shown no effect of statin on viral load in HIV on stable ART [19,47]. In disagreement, Do et al. [52] reported that about 81% of PLHIV with a viral load below 50 maintained the undetectable state compared to 83% on placebo. Similarly, 87% and 92% were reported to have a viral load below 50 copies in statin and placebo, respectively [16]. PLHIV on ART with undetectable HIV-RNA copies sometimes experience viral blips; this generally reflects on the failures of the ART if the blips continue and the viral load is consecutively above 50 HIV-RNA copies [59]. These viral blips are reportedly associated with high levels of baseline viral load prior to initiation of ART [60].
One reason for a continuous viral rebound in PLHIV taking ART is an elevated immune activation associated with the continuous production of viral particles from reservoir sites [61]. Our results revealed increased immune activation among virally suppressed patients on statin treatment. This pre-disposes patients to secondary complications and adversity. Previous evidence has revealed that even in successful ART functionality, demonstrated via low viral load and increased CD4 T-cell count, HIV in the reservoir still induces immune activation [62]. This partly explains increased immune activation in our current analysis, as demonstrated via increased presence of CD4+CD38+HLA-DR+ and CD8+CD38+HLA-DR+ T-cells. According to Overton et al., statins can reduce markers of immune activation, which are CD38 on CD8+ T cells in PLHIV on ART [63]. Immune activation is a chronic inflammatory state often observed in virally suppressed patients. One of the mechanisms by which HIV reservoirs cause immune activation is through the activation of glycoprotein 120 (gp120), which promotes the release of IL-1β from resident macrophages by binding to the chemokine receptor CCR5 [64]. On the other hand, HIV-1, via its proteins such as p17, p24, and gp41, act as a viral pathogen-associated molecular pattern (PAMP) that signals through toll-like receptor-2 (TLR2) and further promotes immunological activation via the nuclear factor-kappa-β (NF-κβ) signalling pathway [65].
Conversely, single-stranded (ssRNA) HIV-1 can interact with plasmacytoid dendritic cells, including TLR-7 and TLR-9, resulting in the release of interferon-alpha (IFN-α), which may contribute to persistent immunological activation [66]. Although atorvastatin has been shown to reduce immune activation in PLHIV, these results are observed in ART-naïve PLHIV [67]. These results suggest that there might be drug–drug interaction when statins are co-administered with ART in PLHIV. Therefore, any pharmacological approach that can effectively reduce immune activation in PLHIV may potentially reduce secondary complications associated with HIV. Due to the multifaceted nature of HIV, numerous pathways are implicated in the reduction in immune activation mediated by statin. For example, statins may reduce cyclo-oxygenase activity and prostaglandin synthesis [68]. They may also inhibit the activation of immune cells, including monocytes and macrophages, which contribute to inflammation [22,67,69].
Interestingly, atorvastatin and pravastatin are reportedly safe and effective for PLHIV on a protease inhibitor or non-nucleoside reverse transcriptase inhibitor-based ART. While rosuvastatin is safe at a lower dose, it should not be prescribed for PLHIV on any PI-based ART. Although there is data to support the use of pitavastatin, its pharmacokinetic features and few drug–drug interactions make it a safe agent in PLHIV. Due to potential drug interactions, side effects, and the paucity of clinical data, patients receiving ART should avoid taking fluvastatin, lovastatin, and simvastatin [70].
A significant reduction in TC levels, which is a primary biomarker of hyperlipidaemia and predictor of CVD, demonstrated the cholesterol-lowering effect of statins. PLHIV have an increased risk of ART-induced CVD, including dyslipidaemia; however, the administration of statins seems to alleviate these complications [71]. For instance, previous meta-analyses of cohorts and RCTs conducted in 2016 [30,31] showed a significant effect of statin on TC; however, these studies analysed data from ART-naïve and ART-treated PLHIV. Therefore, the combined results might not necessarily show the effect of statin on ART-enhanced immune function in PLHIV. Most importantly, our results are corroborated by evidence that shows a significant decrease in TC following statin treatment [12,16,19,20,41,42,44,47,50,72,73,74,75]. While such positive effects are acknowledged, Baker et al. reported conflicting results in 2012, finding no change in TC when pravastatin was administered to PLHIV on ART [76]. Such results potentially reflect the limitations of the statin lipid-lowering effect and a need for further investigations. The mechanism by which statins lower cholesterol is inhibiting the activity of 3-hydroxy-3-methylglutaryl co-enzyme A reductase, resulting in decreased cholesterol synthesis in the liver [77,78].

5. Strength and Limitations

This study is the first quantitative analysis to explore the effect of statin on CD4 T-cell count, viral load and immune activation in ART-controlled PLHIV. The evidence may be reliable as evidence was gathered from high-quality RCT across different databases. Other outcomes were analysed using evidence from a few studies with small sample sizes; therefore, caution must be made when interpreting the analysed evidence. Although heterogeneity was high, sub-group analysis was conducted to explore the source, which was suggested by evidence to be arising from different doses. Through sub-group analysis, we also established that pravastatin and pitavastatin effectively reduced TC amongst virally suppressed patients. Most importantly, the baseline and post-treatment CD4 T-cell counts were meta-analysed.

6. Conclusions

This meta-analysis highlights the importance of statin in PLHIV whose viral load is maintained below a detectable level using ART. Our findings revealed no significant differences in CD4 count (baseline and post-treatment), suggesting no beneficial effect of statins on these parameters. Notably, viral load and CD4 count are primarily used to monitor ART’s efficacy amongst PLHIV on ART. Moreover, our findings revealed no statistical association between statin and risk of viral rebounds. Moreover, we found that statin was significantly associated with an increased marker of immune activation, suggesting the risk of developing HIV-related complications. Nevertheless, statins effectively lowered TC among PLHIV on ART, suggesting that ART treatments did not overshadow the effect of statins. However, more trials may be necessary to ascertain the effect of statin on CD4 T-cell counts as this remains an essential biomarker that reflects ART efficacy in PLHIV.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/ijerph20095668/s1, Figure S1: Effect of statin on baseline CD4 count in PLHIV on ART; Figure S2: Effect of statin on undetectable viral load; Figure S3: Subgroup analysis for CD4 count baseline according to statins; Figure S4: Subgroup total cholesterol according to doses of statin; Figure S5: Subgroup total cholesterol according to the form of statin; Figure S6: Publication bias for total cholesterol using a funnel plot; Table S1A: Search strategy adapted on PubMed on the 18th February, 2023; Table S1B: Search strategy adapted on Scopus; Table S1C: Search strategy adapted on Cochrane Library; Table S2: Quality assessment according to a modified JADAD score guideline.

Author Contributions

K.M.: Conceptualisation, methodology; investigation; project management; formal analysis; software; resources; validation; original draft; and writing—review and editing. W.N.P.: methodology; formal analysis; software; and writing—review and editing. S.L.L.: Validation and writing—review and editing. P.M.: Conceptualisation; formal analysis; validation; and writing—review and editing. S.M. and S.Y.: Writing—review and editing. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable as this study involved the use of already published studies.

Informed Consent Statement

Not applicable.

Data Availability Statement

The additional information supporting this manuscript is provided as a Supplementary File.

Conflicts of Interest

The authors declare no conflict of interest. Perpetua Modjadji and Sphiwe Madiba are Guest Editors for the IJERPH Special Issue on “HIV Prevention: Maximizing Access and Uptake of Prevention Services amidst Societal and Structural Barriers and Intersecting Societal Stigmas” and declare that they have no conflicts of interest associated with this study and did not influence the review process.

References

  1. Global HIV Statistics. Available online: https://www.unaids.org/en/resources/fact-sheet (accessed on 23 January 2023).
  2. UNAIDS HIV and AIDS Estimates. Available online: https://www.unaids.org/en/resources/fact-sheet (accessed on 24 January 2023).
  3. Vijayan, K.V.; Karthigeyan, K.P.; Tripathi, S.P.; Hanna, L.E. Pathophysiology of CD4+ T-Cell Depletion in HIV-1 and HIV-2 Infections. Front. Immunol. 2017, 8, 580. [Google Scholar] [CrossRef]
  4. Jensen, B.E.O.; Knops, E.; Cords, L.; Lübke, N.; Salgado, M.; Busman-Sahay, K.; Estes, J.D.; Huyveneers, L.E.P.; Perdomo-Celis, F.; Wittner, M.; et al. In-Depth Virological and Immunological Characterization of HIV-1 Cure after CCR5Δ32/Δ32 Allogeneic Hematopoietic Stem Cell Transplantation. Nat. Med. 2023, 615, 13–14. [Google Scholar] [CrossRef]
  5. Schramm, B.; Temfack, E.; Descamps, D.; Nicholas, S.; Peytavin, G.; Bitilinyu-Bangoh, J.E.; Storto, A.; Lê, M.P.; Abdi, B.; Ousley, J.; et al. Viral Suppression and HIV-1 Drug Resistance 1 Year after Pragmatic Transitioning to Dolutegravir First-Line Therapy in Malawi: A Prospective Cohort Study. Lancet HIV 2022, 9, e544–e553. [Google Scholar] [CrossRef] [PubMed]
  6. Mahlangu, K.; Modjadji, P.; Madiba, S. The Nutritional Status of Adult Antiretroviral Therapy Recipients with a Recent Hiv Diagnosis; a Cross-Sectional Study in Primary Health Facilities in Gauteng, South Africa. Healthcare 2020, 8, 290. [Google Scholar] [CrossRef] [PubMed]
  7. Hyle, E.P.; Mayosi, B.M.; Middelkoop, K.; Mosepele, M.; Martey, E.B.; Walensky, R.P.; Bekker, L.G.; Triant, V.A. The Association between HIV and Atherosclerotic Cardiovascular Disease in Sub-Saharan Africa: A Systematic Review. BMC Public Health 2017, 17, 954. [Google Scholar] [CrossRef] [PubMed]
  8. Kishabongo, A.S.; Shabani, C.U.; Bisangamo, C.K.; Shindano, T.A.; Takaisi-Kikuni, N.B. Changes of Lipid Profile and Other Biological Parameters in People Living with Human Immunodeficiency Virus on Highly Active Antiretroviral Therapy in the General Referral Provincial Hospital of Bukavu, Eastern of the Democratic Republic of Congo. J. HIV AIDS 2020, 6, 1–9. [Google Scholar] [CrossRef]
  9. Schaefer, R.; Amparo da Costa Leite, P.H.; Silva, R.; Abdool Karim, Q.; Akolo, C.; Cáceres, C.F.; Dourado, I.; Green, K.; Hettema, A.; Hoornenborg, E.; et al. Kidney Function in Tenofovir Disoproxil Fumarate-Based Oral Pre-Exposure Prophylaxis Users: A Systematic Review and Meta-Analysis of Published Literature and a Multi-Country Meta-Analysis of Individual Participant Data. Lancet HIV 2022, 9, e242–e253. [Google Scholar] [CrossRef]
  10. Myerson, M.; Malvestutto, C.; Aberg, J.A. Management of Lipid Disorders in Patients Living with HIV. J. Clin. Pharmacol. 2015, 55, 957–974. [Google Scholar] [CrossRef]
  11. Rodriguez, B.; Valdez, H.; Mijch, A.; Watson, K.; Lederman, M.M.; McComsey, G.A.; Loupa, C.V.; Woolley, I. Statins Blunt HAART-Induced CD4 T-Cell Gains but Have No Long-Term Effect on Virologic Response to HAART. J. Int. Assoc. Physicians. AIDS Care 2007, 6, 198–202. [Google Scholar] [CrossRef]
  12. Mallon, P.W.G.; Miller, J.; Kovacic, J.C.; Kent-Hughes, J.; Norris, R.; Samaras, K.; Feneley, M.P.; Cooper, D.A.; Carr, A. Effect of Pravastatin on Body Composition and Markers of Cardiovascular Disease in HIV-Infected Men-a Randomized, Placebo-Controlled Study. AIDS 2006, 20, 1003–10101. [Google Scholar] [CrossRef]
  13. Hileman, C.O.; Turner, R.; Funderburg, N.T.; Semba, R.D.; McComsey, G.A. Changes in Oxidized Lipids Drive the Improvement in Monocyte Activation and Vascular Disease after Statin Therapy in HIV. AIDS 2016, 30, 65–73. [Google Scholar] [CrossRef]
  14. El Kamari, V.; Hileman, C.O.; Gholam, P.M.; Kulkarni, M.; Funderburg, N.; McComsey, G.A. Statin Therapy Does Not Reduce Liver Fat Scores in Patients Receiving Antiretroviral Therapy for HIV Infection. Clin. Gastroenterol. Hepatol. 2019, 17, 536–542.e1. [Google Scholar] [CrossRef]
  15. Calza, L.; Colangeli, V.; Magistrelli, E.; Contadini, I.; Bon, I.; Re, M.C.; Conti, M.; Mancini, R.; Viale, P. Significant Decrease in Plasma Levels of D-Dimer, Interleukin-8, and Interleukin-12 after a 12-Month Treatment with Rosuvastatin in HIV-Infected Patients under Antiretroviral Therapy. AIDS Res. Hum. Retrovir. 2017, 33, 126–132. [Google Scholar] [CrossRef]
  16. Hearps, A.C.; Angelovich, T.A.; Trevillyan, J.M.; Wong, M.E.; Calmy, A.; Hoy, J.F.; Jaworowski, A. Effect of Rosuvastatin Therapy on Biomarkers of Inflammation and Immune Activation in People with Human Immunodeficiency Virus at Intermediate Cardiovascular Risk. J. Infect. Dis. 2021, 224, 667–672. [Google Scholar] [CrossRef]
  17. Bittar, R.; Giral, P.; Aslangul, E.; Assoumou, L.; Valantin, M.A.; Kalmykova, O.; Federspiel, M.C.; Cherfils, C.; Costagliola, D.; Bonnefont-Rousselot, D. Effects of Rosuvastatin versus Pravastatin on Low-Density Lipoprotein Diameter in HIV-1-Infected Patients Receiving Ritonavir-Boosted Protease Inhibitor. AIDS 2012, 26, 1801–1805. [Google Scholar] [CrossRef] [PubMed]
  18. Bedimo, R.J.; Mar, H.; Bosch, R.J.; Drechsler, H.; Cyktor, J.C.; Macatangay, B.J.C.; Lalama, C.; Rinaldo, C.; Collier, A.; Godfrey, C.; et al. No Evidence for an Association Between Statin Use and Lower Biomarkers of HIV Persistence or Immune Activation/Inflammation During Effective ART. J. Acquir. Immune. Defic. Syndr. 2019, 82, 27–31. [Google Scholar] [CrossRef]
  19. Wongprikorn, A.; Sukasem, C.; Puangpetch, A.; Numthavej, P.; Thakkinstian, A.; Kiertiburanakul, S. Effects of Pitavastatin on Lipid Profiles in HIV-Infected Patients with Dyslipidemia and Receiving Atazanavir/Ritonavir: A Randomized, Double-Blind, Crossover Study. PLoS ONE 2016, 11, e0157531. [Google Scholar] [CrossRef] [PubMed]
  20. Hürlimann, D.; Chenevard, R.; Ruschitzka, F.; Flepp, M.; Enseleit, F.; Béchir, M.; Kobza, R.; Muntwyler, J.; Ledergerber, B.; Lüscher, T.F.; et al. Effects of Statins on Endothelial Function and Lipid Profile in HIV Infected Persons Receiving Protease Inhibitor-Containing Anti-Retroviral Combination Therapy: A Randomised Double Blind Crossover Trial. Heart 2006, 92, 110–112. [Google Scholar] [CrossRef] [PubMed]
  21. Longenecker, C.T.; Sattar, A.; Gilkeson, R.; Mccomsey, G.A. Rosuvastatin Slows Progression of Subclinical Atherosclerosis in Patients with Treated HIV Infection. AIDS 2016, 30, 2195–2203. [Google Scholar] [CrossRef] [PubMed]
  22. Funderburg, N.T.; Jiang, Y.; Debanne, S.M.; Storer, N.; Labbato, D.; Clagett, B.; Robinson, J.; Lederman, M.M.; McComsey, G.A. Rosuvastatin Treatment Reduces Markers of Monocyte Activation in HIV-Infected Subjects on Antiretroviral Therapy. Clin. Infect. Dis. 2014, 58, 588–595. [Google Scholar] [CrossRef]
  23. Eckard, A.R.; Jiang, Y.; Debanne, S.M.; Funderburg, N.T.; Mccomsey, G.A. Effect of 24 Weeks of Statin Therapy on Systemic and Vascular Inflammation in HIV-Infected Subjects Receiving Antiretroviral Therapy. J. Infect. Dis. 2014, 209, 1156–1164. [Google Scholar] [CrossRef] [PubMed]
  24. Cromarty, R.; Archary, D. Inflammation, HIV, and Immune Quiescence: Leveraging on Immunomodulatory Products to Reduce HIV Susceptibility. AIDS Res. Treat. 2020, 2020, 8672850. [Google Scholar] [CrossRef] [PubMed]
  25. Deeks, S.G.; Tracy, R.; Douek, D.C. Systemic Effects of Inflammation on Health during Chronic HIV Infection. Immunity 2013, 39, 633–645. [Google Scholar] [CrossRef] [PubMed]
  26. Lombardi, F.; Belmonti, S.; Moschese, D.; Fabbiani, M.; Borghetti, A.; Ciccullo, A.; Visconti, E.; di Giambenedetto, S. Inflammation Markers in Virologically Suppressed HIV-Infected Patients after Switching to Dolutegravir plus Lamivudine vs Continuing Triple Therapy: 48-Week Results in Real-Life Setting. HIV Res. Clin. Pract. 2022, 23, 28–36. [Google Scholar] [CrossRef]
  27. Werida, R.; Khairat, I.; Khedr, N.F. Effect of Atorvastatin versus Rosuvastatin on Inflammatory Biomarkers and LV Function in Type 2 Diabetic Patients with Dyslipidemia. Biomed. Pharmacother. 2021, 135, 111179. [Google Scholar] [CrossRef]
  28. Nachega, J.B.; Hsu, A.J.; Uthman, O.A.; Spinewine, A.; Pham, P.A. Antiretroviral Therapy Adherence and Drug-Drug Interactions in the Aging HIV Population. AIDS 2012, 26, S39–S53. [Google Scholar] [CrossRef]
  29. Pontelo, B.M.; Greco, D.B.; Guimarães, N.S.; Rotsen, N.; Braga, V.A.R.; Pimentel, P.H.N.; Barbosa, H.; Barroso, T.M.; Tupinambás, U. Profile of Drug–Drug Interactions and Impact on the Effectiveness of Antiretroviral Therapy among Patients Living with HIV Followed at an Infectious Diseases Referral Center in Belo Horizonte, Brazil. Braz. J. Infect. Dis. 2020, 24, 104–109. [Google Scholar] [CrossRef]
  30. Gili, S.; Marra, W.G.; D’Ascenzo, F.; Lonni, E.; Calcagno, A.; Cannillo, M.; Ballocca, F.; Cerrato, E.; Pianelli, M.; Barbero, U.; et al. Comparative Safety and Efficacy of Statins for Primary Prevention in Human Immunodeficiency Virus-Positive Patients: A Systematic Review and Meta-Analysis. Eur. Heart J. 2016, 37, 3600–3609. [Google Scholar] [CrossRef]
  31. Banach, M.; Dinca, M.; Ursoniu, S.; Serban, M.C.; Howard, G.; Mikhailidis, D.P.; Nicholls, S.; Lip, G.Y.H.; Glasser, S.; Martin, S.S.; et al. A PRISMA-Compliant Systematic Review and Meta-Analysis of Randomized Controlled Trials Investigating the Effects of Statin Therapy on Plasma Lipid Concentrations in HIV-Infected Patients. Pharmacol. Res. 2016, 111, 343–356. [Google Scholar] [CrossRef]
  32. Vigny, N.N.; Bonsu, K.O.; Kadirvelu, A. Effectiveness and Safety of Statins on Outcomes in Patients with HIV Infection: A Systematic Review and Meta-Analysis. Sci. Rep. 2022, 12, 18121. [Google Scholar] [CrossRef]
  33. Page, M.J.; McKenzie, J.E.; Bossuyt, P.M.; Boutron, I.; Hoffmann, T.C.; Mulrow, C.D.; Shamseer, L.; Tetzlaff, J.M.; Akl, E.A.; Brennan, S.E.; et al. The PRISMA 2020 Statement: An Updated Guideline for Reporting Systematic Reviews. BMJ 2021, 372, 18121. [Google Scholar] [CrossRef]
  34. Jadad, A.R.; Andrew Moore, R.; Carroll, D.; Jenkinson, C.; John Reynolds, D.M.; Gavaghan, D.J.; McQuay DM, H.J. Assessing the Quality of Reports of Randomised Clinical Trials: Is Blinding Necessary? Control. Clin Trials 1996, 17, 1–12. [Google Scholar] [CrossRef] [PubMed]
  35. Hassani, H.; Ghodsi, M.; Howell, G. A Note on Standard Deviation and Standard Error. Teach. Math. Its Appl. 2010, 29, 108–112. [Google Scholar] [CrossRef]
  36. Cochrane Handbook 6.1.3.2 Imputing Standard Deviations for Changes from Baseline. Available online: https://handbook-5-1.cochrane.org/chapter_16/16_1_3_2_imputing_standard_deviations_for_changes_from_baseline.htm (accessed on 28 January 2023).
  37. Yagiz, G.; Akaras, E.; Kubis, H.P.; Owen, J.A. The Effects of Resistance Training on Architecture and Volume of the Upper Extremity Muscles: A Systematic Review of Randomised Controlled Trials and Meta-Analyses. Appl. Sci. 2022, 12, 1593. [Google Scholar] [CrossRef]
  38. Bowden, J.; Tierney, J.F.; Copas, A.J.; Burdett, S. Quantifying, Displaying and Accounting for Heterogeneity in the Meta-Analysis of RCTs Using Standard and Generalised Q Statistics. BMC Med. Res. Methodol. 2011, 11, 41. [Google Scholar] [CrossRef] [PubMed]
  39. Sedgwick, P. Meta-Analyses: Heterogeneity and Subgroup Analysis. BMJ 2013, 346, f4040. [Google Scholar] [CrossRef]
  40. Mathur, M.B.; VanderWeele, T.J. Sensitivity Analysis for Publication Bias in Meta-Analyses. J. R Stat. Soc. Ser. C Appl. Stat. 2020, 69, 1091–1119. [Google Scholar] [CrossRef]
  41. Bonnet, F.; Aurillac-Lavignolle, V.; Breilh, D.; Thiébaut, R.; Peuchant, E.; Bernard, N.; Lacoste, D.; Dabis, F.; Beylot, J.; Chêne, G.; et al. Pravastatin in HIV-Infected Patients Treated with Protease Inhibitors: A Placebo-Controlled Randomised Study. HIV Clin. Trials 2007, 8, 53–60. [Google Scholar] [CrossRef]
  42. Stein, J.H.; Merwood, M.A.; Bellehumeur, J.L.; Aeschlimann, S.E.; Korcarz, C.E.; Underbakke, G.L.; Mays, M.E.; Sosman, J.M. Effects of Pravastatin on Lipoproteins and Endothelial Function in Patients Receiving Human Immunodeficiency Virus Protease Inhibitors. Am. Heart J. 2004, 147, 713–717. [Google Scholar] [CrossRef]
  43. Hileman, C.O.; McComsey, G.A. Short Communication: The Effect of Rosuvastatin on Vascular Disease Differs by Smoking Status in Treated HIV Infection. AIDS Res. Hum. Retrovir. 2018, 34, 282–285. [Google Scholar] [CrossRef]
  44. Trevillyan, J.M.; Dart, A.; Paul, E.; Cavassini, M.; Fehr, J.; Staehelin, C.; Dewar, E.M.; Hoy, J.F.; Calmy, A. Impact of Rosuvastatin on Atherosclerosis in People with HIV at Moderate Cardiovascular Risk: A Randomised, Controlled Trial. AIDS 2021, 35, 619–624. [Google Scholar] [CrossRef] [PubMed]
  45. Nixon, D.E.; Bosch, R.J.; Chan, E.S.; Funderburg, N.T.; Hodder, S.; Lake, J.E.; Lederman, M.M.; Klingman, K.L.; Aberg, J.A.; Bergstrom, K.; et al. Effects of Atorvastatin on Biomarkers of Immune Activation, Inflammation, and Lipids in Virologically Suppressed, Human Immunodeficiency Virus-1–Infected Individuals with Low-Density Lipoprotein Cholesterol <130 Mg/DL (AIDS Clinical Trials Group Study A5275). J. Clin. Lipidol. 2017, 11, 61–69. [Google Scholar] [CrossRef] [PubMed]
  46. Morrison, J.T.; Longenecker, C.T.; Mittelsteadt, A.; Jiang, Y.; Debanne, S.M.; McComsey, G.A. Effect of Rosuvastatin on Plasma Coenzyme Q10 in HIV-Infected Individuals on Antiretroviral Therapy. HIV Clin. Trials 2016, 17, 140–146. [Google Scholar] [CrossRef] [PubMed]
  47. Nou, E.; Lu, M.T.; Looby, S.E.; Fitch, K.V.; Kim, E.A.; Lee, H.; Hoffmann, U.; Grinspoon, S.K.; Lo, J. Serum Oxidized Low-Density Lipoprotein Decreases in Response to Statin Therapy and Relates Independently to Reductions in Coronary Plaque in Patients with HIV. AIDS 2016, 30, 583–590. [Google Scholar] [CrossRef]
  48. Nakanjako, D.; Ssinabulya, I.; Nabatanzi, R.; Bayigga, L.; Kiragga, A.; Joloba, M.; Kaleebu, P.; Kambugu, A.D.; Kamya, M.R.; Sekaly, R.; et al. Atorvastatin Reduces T-Cell Activation and Exhaustion among HIV-Infected CART-Treated Suboptimal Immune Responders in Uganda: A Randomised Crossover Placebo-Controlled Trial. Trop. Med. Int. Health 2015, 20, 380–390. [Google Scholar] [CrossRef]
  49. Hileman, C.O.; Tangpricha, V.; Sattar, A.; McComsey, G.A. Baseline Vitamin D Deficiency Decreases the Effectiveness of Statins in HIV-Infected Adults on Antiretroviral Therapy. J. Acquired. Defic. Syndr. 2017, 74, 539–547. [Google Scholar] [CrossRef]
  50. Defilippi, C.; Christenson, R.; Joyce, J.; Park, E.A.; Wu, A.; Fitch, K.V.; Looby, S.E.; Lu, M.T.; Hoffmann, U.; Grinspoon, S.K.; et al. Statin Effects on Myocardial Fibrosis Markers in People Living With HIV. J. Acquir. Immune. Defic. Syndr. 2018, 78, 105–110. [Google Scholar] [CrossRef]
  51. Erlandson, K.M.; Jiang, Y.; Debanne, S.M.; McComsey, G.A. Rosuvastatin Worsens Insulin Resistance in HIV-Infected Adults on Antiretroviral Therapy. Clin. Infect. Dis. 2015, 61, 1566–1572. [Google Scholar] [CrossRef]
  52. Dirajlal-Fargo, S.; Kinley, B.; Jiang, Y.; Longenecker, C.T.; Hileman, C.O.; Debanne, S.; McComsey, G.A. Statin Therapy Decreases N-Terminal pro-B-Type Natriuretic Peptide in HIV: Randomized Placebo-Controlled Trial. AIDS 2014, 29, 313–321. [Google Scholar] [CrossRef]
  53. Erlandson, K.M.; Jiang, Y.; Debanne, S.M.; Mccomsey, G.A. Effects of 96 Weeks of Rosuvastatin on Bone, Muscle, and Fat in HIV-Infected Adults on Effective Antiretroviral Therapy. AIDS Res. Hum. Retrovir. 2016, 32, 311–316. [Google Scholar] [CrossRef]
  54. Lok, J.J.; Bosch, R.J.; Benson, C.A.; Collier, A.C.; Robbins, G.K.; Shafer, R.W.; Hughes, M.D. Long-Term Increase in CD4+ T-Cell Counts during Combination Antiretroviral Therapy for HIV-1 Infection. AIDS 2010, 24, 1867–1876. [Google Scholar] [CrossRef] [PubMed]
  55. Weijma, R.G.M.; Vos, E.R.A.; Oever, J.T.; Van Schilfgaarde, M.; Dijksman, L.M.; Van Der Ven, A.; Van Den Berk, G.E.L.; Brinkman, K.; Frissen, J.P.H.J.; Leyte, A.; et al. The Effect of Rosuvastatin on Markers of Immune Activation in Treatment-Naive Human Immunodeficiency Virus-Patients. Open Forum. Infect Dis. 2016, 3, ofv201. [Google Scholar] [CrossRef] [PubMed]
  56. Hearps, A.C.; Maisa, A.; Cheng, W.J.; Angelovich, T.A.; Lichtfuss, G.F.; Palmer, C.S.; Landay, A.L.; Jaworowski, A.; Crowe, S.M. HIV Infection Induces Age-Related Changes to Monocytes and Innate Immune Activation in Young Men That Persist despite Combination Antiretroviral Therapy. AIDS 2012, 26, 843–853. [Google Scholar] [CrossRef] [PubMed]
  57. Muldoon, M.F.; Marsland, A.; Flory, J.D.; Rabin, B.S.; Whiteside, T.L.; Manuck, S.B. Immune System Differences in Men with Hypo- or Hypercholesterolemia. Clin. Immunol. Immunopathol. 1997, 84, 145–149. [Google Scholar] [CrossRef] [PubMed]
  58. Motkowski, R.; Alifier, M.; Abramowicz, P.; Konstantynowicz, J.; Mikołuć, B.; Stasiak-Barmuta, A. Innate and Acquired Cellular Immunity in Children with Familial Hypercholesterolemia Treated with Simvastatin. J. Clin. Med. 2022, 11, 2924. [Google Scholar] [CrossRef] [PubMed]
  59. Suzuki, K.; Levert, A.; Yeung, J.; Starr, M.; Cameron, J.; Williams, R.; Rismanto, N.; Stark, T.; Druery, D.; Prasad, S.; et al. HIV-1 Viral Blips Are Associated with Repeated and Increasingly High Levels of Cell-Associated HIV-1 RNA Transcriptional Activity. AIDS 2021, 35, 2095–2103. [Google Scholar] [CrossRef]
  60. Sörstedt, E.; Nilsson, S.; Blaxhult, A.; Gisslén, M.; Flamholc, L.; Sönnerborg, A.; Yilmaz, A. Viral Blips during Suppressive Antiretroviral Treatment Are Associated with High Baseline HIV-1 RNA Levels. BMC Infect Dis. 2016, 16, 305. [Google Scholar] [CrossRef]
  61. Corbeau, P.; Reynes, J. Immune Reconstitution under Antiretroviral Therapy: The New Challenge in HIV-1 Infection. Blood 2011, 117, 5582–5590. [Google Scholar] [CrossRef]
  62. Bourgeois, C.; Gorwood, J.; Olivo, A.; Le Pelletier, L.; Capeau, J.; Lambotte, O.; Béréziat, V.; Lagathu, C. Contribution of Adipose Tissue to the Chronic Immune Activation and Inflammation Associated With HIV Infection and Its Treatment. Front. Immunol. 2021, 12, 2222. [Google Scholar] [CrossRef]
  63. Overton, E.T.; Sterrett, S.; Westfall, A.O.; Kahan, S.M.; Burkholder, G.; Zajac, A.J.; Goepfert, P.A.; Bansal, A. Effects of Atorvastatin and Pravastatin on Immune Activation and T-Cell Function in Antiretroviral Therapy-Suppressed HIV-1-Infected Patients. AIDS 2014, 28, 2627–2631. [Google Scholar] [CrossRef]
  64. Cheung, R.; Ravyn, V.; Wang, L.; Ptasznik, A.; Collman, R.G. Signaling Mechanism of HIV-1 Gp120 and Virion-Induced IL-1 Release in Primary Human Macrophages 1. J. Immunol. 2008, 180, 6675–6684. [Google Scholar] [CrossRef] [PubMed]
  65. Henrick, B.M.; Yao, X.D.; Rosenthal, K.L. HIV-1 Structural Proteins Serve as PAMPs for TLR2 Heterodimers Significantly Increasing Infection and Innate Immune Activation. Front. Immunol. 2015, 6, 426. [Google Scholar] [CrossRef] [PubMed]
  66. Younas, M.; Psomas, C.; Reynes, J.; Corbeau, P. Immune Activation in the Course of HIV-1 Infection: Causes, Phenotypes and Persistence under Therapy. HIV Med. 2016, 17, 89–105. [Google Scholar] [CrossRef] [PubMed]
  67. Ganesan, A.; Crum-Cianflone, N.; Higgins, J.; Qin, J.; Rehm, C.; Metcalf, J.; Brandt, C.; Vita, J.; Decker, C.F.; Sklar, P.; et al. High Dose Atorvastatin Decreases Cellular Markers of Immune Activation without Affecting HIV-1 RNA Levels: Results of a Double-Blind Randomized Placebo Controlled Clinical Trial. J. Infect. Dis. 2011, 203, 756–764. [Google Scholar] [CrossRef]
  68. Massaro, M.; Zampolli, A.; Scoditti, E.; Carluccio, M.A.; Storelli, C.; Distante, A.; De Caterina, R. Statins Inhibit Cyclooxygenase-2 and Matrix Metalloproteinase-9 in Human Endothelial Cells: Anti-Angiogenic Actions Possibly Contributing to Plaque Stability. Cardiovasc. Res. 2010, 86, 311–320. [Google Scholar] [CrossRef]
  69. Hussain, S.K.; Golozar, A.; Widney, D.P.; Rappocciolo, G.; Penugonda, S.; Bream, J.H.; Martínez-Maza, O.; Jacobson, L.P. Effect of Statin Use on Inflammation and Immune Activation Biomarkers in HIV-Infected Persons on Effective Antiretroviral Therapy. AIDS Res. Hum. Retrovir. 2021, 37, 357–367. [Google Scholar] [CrossRef]
  70. Chastain, D.B.; Stover, K.R.; Riche, D.M. Evidence-Based Review of Statin Use in Patients with HIV on Antiretroviral Therapy. J. Clin. Transl. Endocrinol. 2017, 8, 6–14. [Google Scholar] [CrossRef] [PubMed]
  71. Fitch, K.V.; Fulda, E.S.; Grinspoon, S.K. Statins for Primary Cardiovascular Disease Prevention among People with HIV: Emergent Directions. Curr. Opin. HIV AIDS 2022, 17, 293–300. [Google Scholar] [CrossRef]
  72. Baldini, F.; di Giambenedetto, S.; Cingolani, A.; Murri, R.; Ammassari, A.; Luca, A. de Efficacy and Tolerability of Pravastatin for the Treatment of HIV-1 Protease Inhibitor-Associated Hyperlipidaemia: A Pilot Study. AIDS 2000, 14, 1660–1662. [Google Scholar] [CrossRef] [PubMed]
  73. Calza, L.; Manfredi, R.; Chiodo, F. Statins and Fibrates for the Treatment of Hyperlipidaemia in HIV-Infected Patients Receiving HAART. AIDS 2003, 17, 851–859. [Google Scholar] [CrossRef]
  74. Calza, L.; Manfredi, R.; Colangeli, V.; Trapani, F.F.; Salvadori, C.; Magistrelli, E.; Danese, I.; Verucchi, G.; Serra, C.; Viale, P. Two-Year Treatment with Rosuvastatin Reduces Carotid Intima-Media Thickness in HIV Type 1-Infected Patients Receiving Highly Active Antiretroviral Therapy with Asymptomatic Atherosclerosis and Moderate Cardiovascular Risk. AIDS Res. Hum. Retrovir. 2013, 29, 547–556. [Google Scholar] [CrossRef]
  75. Grandi, A.M.; Nicolini, E.; Rizzi, L.; Caputo, S.; Annoni, F.; Cremona, A.M.; Marchesi, C.; Guasti, L.; Maresca, A.M.; Grossi, P. Dyslipidemia in HIV-Positive Patients: A Randomized, Controlled, Prospective Study on Ezetimibe+fenofibrate versus Pravastatin Monotherapy. J. Int. AIDS Soc. 2014, 17, 19004. [Google Scholar] [CrossRef] [PubMed]
  76. Baker, J.V.; Huppler Hullsiek, K.; Prosser, R.; Duprez, D.; Grimm, R.; Tracy, R.P.; Rhame, F.; Henry, K.; Neaton, J.D. Angiotensin Converting Enzyme Inhibitor and HMG-CoA Reductase Inhibitor as Adjunct Treatment for Persons with HIV Infection: A Feasibility Randomised Trial. PLoS ONE 2012, 7, e46894. [Google Scholar] [CrossRef] [PubMed]
  77. Goldstein, J.L.; Brown, M.S. A Century of Cholesterol and Coronaries: From Plaques to Genes to Statins. Cell 2015, 161, 161–172. [Google Scholar] [CrossRef] [PubMed]
  78. Buhaescu, I.; Izzedine, H. Mevalonate Pathway: A Review of Clinical and Therapeutical Implications. Clin. Biochem. 2007, 40, 575–584. [Google Scholar] [CrossRef]
Figure 1. PRISMA flow diagram presenting information sources and study selection procedure followed.
Figure 1. PRISMA flow diagram presenting information sources and study selection procedure followed.
Ijerph 20 05668 g001
Figure 2. Effect of statin on follow-up CD4 T-cells in PLHIV on ART. I2 statistic was used to assess statistical heterogeneity. The probability value of less than 0.05 was considered statistically significant. Hürlimann et al., 2006 [20]; Mallon et al., 2006 [12]; Bonnet et al., 2007 [41].
Figure 2. Effect of statin on follow-up CD4 T-cells in PLHIV on ART. I2 statistic was used to assess statistical heterogeneity. The probability value of less than 0.05 was considered statistically significant. Hürlimann et al., 2006 [20]; Mallon et al., 2006 [12]; Bonnet et al., 2007 [41].
Ijerph 20 05668 g002
Figure 3. Effect of statin on markers of immune activation in PLHIV on ART. (A) CD8+ CD38+ HLA-DR+ T-cells; and (B) CD4+CD38+HLA-DR+ T-cells. I2 statistic was used to assess statistical heterogeneity. The probability value of less than 0.05 was considered statistically significant. Funderburg et al., 2013 [22]; Do et al., 2014 [52]; Eckard et al., 2014 [23]; Nakanjako et al., 2015 [48]; Longenecker et al., 2016 [21].
Figure 3. Effect of statin on markers of immune activation in PLHIV on ART. (A) CD8+ CD38+ HLA-DR+ T-cells; and (B) CD4+CD38+HLA-DR+ T-cells. I2 statistic was used to assess statistical heterogeneity. The probability value of less than 0.05 was considered statistically significant. Funderburg et al., 2013 [22]; Do et al., 2014 [52]; Eckard et al., 2014 [23]; Nakanjako et al., 2015 [48]; Longenecker et al., 2016 [21].
Ijerph 20 05668 g003
Figure 4. Effect of statin on total cholesterol in PLHIV on ART. I2 statistic was used to assess statistical heterogeneity. The probability value of less than 0.05 was considered statistically significant. Bonnet et al., 2007 [41]; Defilippi et al., 2018 [50]; Hearps et al., 2021 [16]; Hürlimann et al., 2006 [20]; Mallon et al., 2006 [12]; Nou et al., 2016 [47]; Stein et al., 2004 [42]; Trevillyan et al., 2021 [44]; Wongprikorn et al., 2016 [19].
Figure 4. Effect of statin on total cholesterol in PLHIV on ART. I2 statistic was used to assess statistical heterogeneity. The probability value of less than 0.05 was considered statistically significant. Bonnet et al., 2007 [41]; Defilippi et al., 2018 [50]; Hearps et al., 2021 [16]; Hürlimann et al., 2006 [20]; Mallon et al., 2006 [12]; Nou et al., 2016 [47]; Stein et al., 2004 [42]; Trevillyan et al., 2021 [44]; Wongprikorn et al., 2016 [19].
Ijerph 20 05668 g004
Figure 5. Visualisation of publication bias graphically on CD4 count. (A) Funnel plot; and (B) Q-Q normal plot.
Figure 5. Visualisation of publication bias graphically on CD4 count. (A) Funnel plot; and (B) Q-Q normal plot.
Ijerph 20 05668 g005
Table 1. Characteristics of included RCTs (n = 20) evaluating effect of statin in PLHIV on antiretroviral therapy.
Table 1. Characteristics of included RCTs (n = 20) evaluating effect of statin in PLHIV on antiretroviral therapy.
Study, Year, CountryStudy DesignPopulation StatusDuration of HIV
(Years/Months)
Duration of ART (Years/Months)Sample Size Statin Group, nForm of Statin, Dosage, and DurationAge (Years)Gender,
Male (%)
Hearps et al., 2021 [16]
Switzerland
Post hoc analysis of the RCT.Sixty-nine virologically suppressed patients.19.1 ± 3.65NR3320 mg rosuvastatin for 96 weeks.54 ± 6.532 (97)
Trevillyan et al., 2021 [44]
Australia
Post hoc analysis of the RCT.Eighty-eight controlled PLHIV on ART.17.2 ± 8.5NR4420 mg rosuvastatin or 10 mg on antiretroviral therapy for 96 weeks.53.9 ± 5.942 (95.5)
Kamari et al., 2019 [14]
USA
Post hoc analysis of the RCT.One hundred and forty-seven PLHIV on ART.NR7.0 ± 5.3 7210 mg rosuvastatin for 96 weeks.45 ± 9.058 (81)
deFillippi et al., 2018 [50]
USA
Post hoc analysis of the RCT.Forty PLHIV on stable ART.16.8 ± 5.1 12.4 ± 3.71920 mg to 40 mg atorvastatin for three months.52.2 ± 3.815 (79)
Hileman et al., 2017 [49]
USA
Post hoc analysis of the RCT.One hundred and forty-seven PLHIV on ART.11.71 ± 6.6 7.0 ± 5.37210 mg rosuvastatin for 96 weeks.45.37 ± 9.1358 (81)
Nixon et al., 2017 [45]
USA
Post hoc analysis of the RCT.Ninety-four PLHIV on boosted PI-based ART.6 ± 1.3 NR9410 mg of atorvastatin for 4 weeks, followed by 20 mg for 16 weeks.48 ± 2.364 (68)
Hileman et al., 2016 [13]
USA
Post hoc analysis of the RCT.One hundred and forty-seven PLHIV on stable ART.NRNR7210 mg rosuvastatin for 48 weeks.45.4 ± 9.158 (81)
Longenecker et al., 2016 [21]
Australia
Post hoc analysis of the RCT.One hundred and forty-seven PLHIV on stable ART.11 ± 1.835.2 ± 1.137210 mg rosuvastatin for 96 weeks.45 ± 1.758 (81)
Morrison et al., 2016 [46]
USA
Post hoc analysis of the RCT.One hundred and forty-seven PLHIV on ART.11 ± 1.83 3.9 ± 1.23 7210 mg rosuvastatin for 24 weeks.45 ± 1.6758 (81)
Nou et al., 2016 [47]
USA
Post hoc analysis of the RCT.Forty PLHIV with sub-clinical coronary atherosclerosis on stable ART.16.8 ± 5.1 12.4 ± 3.71940 atorvastatin for 12 months.52.2 ± 3.815 (79)
Wongprikorn et al., 2016 [19]
China
Post hoc analysis of the RCT.Twenty-four PLHIV on ART.NR42 ± 6.9312 2 mg pitavastatin for 12 weeks.49.6 ± 10.68 (66.7)
Do et al., 2014 [52]
USA
Post hoc analysis of the RCT.One hundred and forty-seven PLHIV on stable ART.122 ± 31.556 ± 21.25 6410 mg rosuvastatin for 96 weeks46 ± 2.553 (83)
Erlandson et al., 2015 [51]
USA
Post hoc analysis of the RCT.One hundred and forty-seven PLHIV on stable ART.NR63 ± 13.67 7210 mg of rosuvastatin for 96 weeks.45.6 ± 1.7258 (81)
Nakanjako et al., 2015 [48]
Uganda
Post hoc analysis of the RCT.Thirty PLHIV on a suppressive cART.NRNR1580 mg of rosuvastatin for 12 weeks.43 ± 8.54 8 (53)
Eckard et al., 2014 [23]
USA
Post hoc analysis of the RCT.One hundred and forty-seven PLHIV on stable ART.133 ± 20.763 ± 13.7 7210 mg rosuvastatin 24 weeks45.6 ± 1.7258 (81)
Funderburg et al., 2013 [22]
USA
Post hoc analysis of the RCT.One hundred and forty-seven PLHIV on ART.133 ± 20.763 ± 13.77210 mg rosuvastatin for 24 weeks.45.6 ± 1.7158 (81)
Bonnet et al., 2007 [41]
France
Post hoc analysis of the RCT.Twenty PLHIV on stable ART.NRNR1240 mg of pravastatin for three months.42.5 ± 2.31 11 (92)
Hürlimann et al., 2006 [20]
Switzerland
Post hoc analysis of the RCT.Twenty-nine PLHIV on a PI-containing cART.NRNR2940 mg pravastatin for 8 weeks.4323 (79)
Mallon et al., 2006 [12]
Australia
Post hoc analysis of the RCT.Thirty-one PLHIV with hypercholesterolaemia on PI-containing therapy.13.5 ± 4.5 NR1640 mg pravastatin for 12 weeks.52 ± 1216 (100)
Stein et al., 2004 [42]
USA
Post hoc analysis of the RCT.Twenty PLHIV on HIV PI-based ART.11.8 ± 0.5 8 ± 3.72040 mg pravastatin for 8 weeks.44.1 ± 1.618 (90)
HIV—human immune deficiency virus; RCT—randomised controlled trials; PLHIV—people living with HIV; PI—protease inhibitor; ARV—antiretroviral therapy; cART—combined antiretroviral therapy, NR—not reported.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Mokgalaboni, K.; Phoswa, W.N.; Yates, S.; Lebelo, S.L.; Madiba, S.; Modjadji, P. A Systematic Review and Meta-Analysis on the Impact of Statin Treatment in HIV Patients on Antiretroviral Therapy. Int. J. Environ. Res. Public Health 2023, 20, 5668. https://doi.org/10.3390/ijerph20095668

AMA Style

Mokgalaboni K, Phoswa WN, Yates S, Lebelo SL, Madiba S, Modjadji P. A Systematic Review and Meta-Analysis on the Impact of Statin Treatment in HIV Patients on Antiretroviral Therapy. International Journal of Environmental Research and Public Health. 2023; 20(9):5668. https://doi.org/10.3390/ijerph20095668

Chicago/Turabian Style

Mokgalaboni, Kabelo, Wendy Nokhwezi Phoswa, Samantha Yates, Sogolo Lucky Lebelo, Sphiwe Madiba, and Perpetua Modjadji. 2023. "A Systematic Review and Meta-Analysis on the Impact of Statin Treatment in HIV Patients on Antiretroviral Therapy" International Journal of Environmental Research and Public Health 20, no. 9: 5668. https://doi.org/10.3390/ijerph20095668

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop