Skip to main content

The effect of a treatment switch to integrase Strand transfer inhibitor–based regimens on weight gain and other metabolic syndrome-related conditions

Abstract

Objective

This study aimed to assess weight gain associated with treatment switching to INSTI-based regimens in people living with HIV (PLWH) and to determine whether it is accompanied by worsening features of hypertension, dyslipidemia, or hyperglycemia.

Methods

In this two-center retrospective observational study, we assessed weight gain and metabolic features in PLWH who switched to an INSTI-based regimen (study group) as compared to patients who remained on a non-INSTI regimen (control group) over a 24-month follow-up period.

Results

One-hundred seventy-four PLWH were included in the study group, and 175 were included in the control group. The study group gained 2.51 kg ± 0.31 (mean ± standard deviation) over the 2 years of follow-up, while the control group gained 1.1 ± 0.31 kg over the same time course (p < 0.001). INSTI treatment, Caucasian origin, and lower BMI were risk factors associated with excessive weight gain during the 2 years of follow-up. Among metabolic parameters, only glucose levels increased after initiating INSTI-based regimens, although limited to males of African origin (p = 0.009).

Conclusions

We observed a mild weight gain after switching to INSTI-based regimens, with no major impact on metabolic parameters over 2 years of follow-up. Longer follow-up might be needed to observe the adverse metabolic effects of INSTI-based regimens. The impact on weight gain should be discussed with every patient before the treatment switch to ensure a balanced diet and physical activity to prevent excessive weight gain that might hamper compliance with ART.

Peer Review reports

Introduction

Integrase inhibitors (Integrase Strand Transfer Inhibitor – INSTI) [1] revolutionized antiretroviral treatment. They were proven to achieve superior viral suppression as compared to protease inhibitors (PIs) and have an improved drug-drug interaction and safety profile, leading to their inclusion as core agents in most guidelines of antiretroviral treatment (ART) regimens [2,3,4]. Nevertheless, these drugs are not free from adverse effects. INSTIs are associated with side effects impacting the central nervous system, including headaches, insomnia, mood disturbances, abnormal dreaming, and dizziness [5]. Other adverse effects include nausea, diarrhea, rash, and increased serum creatinine levels. Some studies [6, 7] have suggested that weight gain may be a consequence of treatment with INSTI, while this was not observed in other studies [8]. In addition, there were substantial discrepancies in the extent of weight gain noted between different studies [9,10,11,12,13,14,15].

ART-associated weight gain is a known phenomenon commonly observed in the first year of antiretroviral treatment and is often ascribed to a general improvement of the patient’s condition, a term known as “return to health” [16,17,18]. There is evidence indicating that weight gain may also be an adverse effect of combined antiretroviral treatment (cART), which may exacerbate metabolic disturbances such as dyslipidemia and insulin resistance and contribute to the development of cardiovascular complications [18,19,20,21,22]. Additionally, drug-related weight gain may have a negative effect on patients’ compliance with their life-saving medical treatment.

In this study, we evaluated whether the transition to INSTI-based regimens leads to weight gain and worsening of metabolic syndrome-related conditions. To this end, we compared PLWH who were switched to INSTI-based regimens with those who continued treatment with non-INSTI-based regimens, namely non-nucleoside reverse transcriptase inhibitors (NNRTIs) and protease inhibitors (PIs). We further examined the effects exerted by different medications within the INSTIs family and the accompanying nucleoside reverse transcriptase inhibitors (NRTIs) on metabolic features in ART-treated PLWH, and studied whether demographic characteristics such as age, gender, or ethnicity play a role in INSTI-associated weight gain.

Methods

This retrospective observational clinical study included adult PLWH treated with PI or NNRTI-based cART for at least a year in two HIV clinics in Israel. Two groups of PLWH were compared: PLWH who switched to an INSTI-based regimen and a control group that maintained a non INSTI-based ART regimen. Clinical parameters were retrieved from medical records at three different time points. “Time 0” was defined for participants in the study group as the day of treatment change to an INSTI-based regimen, after successfully being treated for at least 1 year with a non- INSTI-based regimen. In the control group, “Time 0” was defined as an arbitrary day, at least 1 year after being successfully treated with a non-INSTI based regimen. Additional data was collected at 12 ± 2 months and 24 ± 2 months after time 0. The parameters retrieved from the medical records included gender, origin, date of birth, weight (kg, at diagnosis, treatment initiation and study follow-up), height (cm), BMI (kg/m2), blood pressure (mmHg) and biochemical tests, including fasting triglycerides, total cholesterol, LDL, HDL, and fasting glucose levels (all in mg/dL). All parameters were evaluated during routine clinic follow-up visits at 0, 12 ± 2 months and 24 ± 2 months. HIV-related parameters were date of HIV diagnosis, date of treatment initiation, CD4 cell counts and HIV viral load (VL) at diagnosis, treatment initiation, and study follow-up (time 0, 12 ± 2 months and 24 ± 2 months), ART history and duration of viral suppression until time 0 (months). Patients were excluded from the study in cases of HBV or HCV cross-infections, opportunistic infections, hospitalization, or pregnancy during the period of follow-up.

Statistical analysis

Patient characteristics were reported by descriptive statistics using all available information. Demographic data were analyzed by independent sample t-test or by Chi-square test (or likelihood ratio test, G-test) according to the scale of the variable, and descriptive statistics are given as mean (M) with standard deviation (SD) or as frequency (n) with percentage (%). Two-order interactions: study group*time and three-order interactions: risk factor (gender, origin or age)*study group*time were assessed by repeated measures mixed-effect models with a random intercept at the participant level,after adjusting for covariates. These models were followed by post-hoc analysis to test the differences in time between all levels of risk factors and study groups. The analysis was done using the Benjamini-Hochberg procedure to control the type I error. The models were adjusted for CD4 and BMI at index time, which were included as they were influential in the model. We also performed a multivariate analysis to identify risk factors for gaining excessive weight, defined as more than 5% weight gain during 2 years of follow-up. Variables included in the analysis were study group (control or switch), origin, gender, age, BMI and CD4 levels at index time (time 0). A sensitivity analysis introduced an alternative criterion for defining excessive weight gain: individuals in the highest quintile of the relative weight difference, either within a one-year follow-up or 2 years (gained more than 5.6 kg during 1 year of follow-up or 7.3 kg during 2 years). P-value < 0.05 was considered statistically significant. All reported p-values are two-tailed. Analysis was done with IBM SPSS STATISTICS version 25.0, Stata/SE version 15.0 (StataCorp) and R statistical software version 3.5.0 (R Project for Statistical Computing).

Ethics

This research was approved by the Hadassah-Hebrew University Medical Center Institutional Review Board (IRB) (0259–19-HMO) and the Kaplan Medical Center ethics committee (KMC-0036-10). Due to the retrospective design of the research, a waiver of informed consent of the participants in the study was provided by both ethics committees.

Results

Approximately two thousand files of PLWH were screened at two AIDS centers: Neve Or (Kaplan Medical Center, Rehovot, Israel) and the Hadassah AIDS Center (Hadassah - Hebrew University Medical Center, Jerusalem, Israel). Three hundred and forty nine PLWH were included: 174 participants were included in the study group and 175 in the control group. The two groups were comparable in age, gender, risk groups, time from HIV diagnosis to treatment initiation, CD4 T-cell counts, viral load, and weight at diagnosis and Time 0 (Table 1). BMI differed only at the time of HIV diagnosis but was comparable at Time 0. Viral load suppression time until Time 0 was longer in the study group (71 months vs. 56 months in the control group, p = 0.0028). Almost half of the participants (84/174, 48.3%) who switched to INSTI were prescribed Dolutegravir. Tenofovir disoproxil fumarate/Emtricitabine (TDF/FTC) was the most prevalent backbone used in the control and study groups before the treatment switch (295/349, 84.5%). Usage of TDF/FTC as a backbone decreased to 54% following the switch, as patients were switched to INSTI regimens combined with abacavir/lamivudine (ABC/3TC) or Tenofovir alafenamide/Emtricitabine (TAF/FTC) (Table 1).

Table 1 Basic characteristics

Weight and BMI

Participants in both groups gained weight and their BMI increased over the 2 years of follow-up. While PLWH in the control group gained 1.10 ± 0.31 kg during the 2 years of follow-up, participants who switched to an INSTI-based regimen gained 2.51 ± 0.31 kg during the same period (p < 0.001). The main effect was noticed in the first year of follow-up, in which the control group gained 0.65 ± 0.27 kg, while the study group gained 1.91 ± 0.27 kg (p < 0.001, Fig. 1A). As expected, a similar difference was noted for BMI, as the control and the study groups BMI increased by 0.40 ± 0.12 kg/m2 and 0.92 ± 0.12 kg/m2 over 2 years, respectively (p < 0.001). The rate of weight gain and BMI change in the study group weakened in the second year of follow-up and became similar to the one observed in the control group (Fig. 1A).

Fig. 1
figure 1

Weight gain following the switch to INSTI-based regimens. Results are presented as mean values ± standard error. Weight gain in study vs. control group (A), stratified by ethnicity (B), gender (C) and age (D). Differences in weight (kg) were calculated after 12 and 24 months of follow-up; P-values in each panel related to the difference between the control and study groups at 24 months; NS: not significant. Models were adjusted for CD4 and BMI at index time

To explore risk factors, we assessed the effect of ethnicity, gender, age, and the impact of the specific INSTI types and backbone agents prescribed.

Ethnicity

Our cohort included 110 Caucasians and 239 African descendants. Regardless of their group, Caucasians gained significantly more weight than African descendants did (2.55 ± 0.40 vs 1.46 ± 0.27 kg, respectively, p < 0.009). During the first year, both ethnicities gained weight similarly in their respective study groups. However, while the African descendants moderated the rate of weight gain in the second year of follow-up, Caucasians continued to demonstrate an increased rate similar to the one observed in the first year (Fig. 1B). Weight gain patterns differed in the two groups based on ethnicity: African descendants in the study group gained 2.43 ± 0.37 kg, while in the control group they gained only 0.45 ± 0.38 kg (p < 0.001). In contrast, we found no difference in weight gain between the study and control group among Caucasians (2.71 ± 0.57 kg vs 2.40 ± 0.54 kg, respectively, p = 0.649, Fig. 1B).

Gender

Our cohort included 196 males and 153 females. Males and females gained 1.80 ± 0.34 kg and 1.81 ± 0.30 kg, respectively (p = 0.985). During the 2 years of follow-up, males’ weight increased by 2.60 ± 0.44 kg in the study group and by 1.12 ± 0.41 kg in the control group (p = 0.004). Among females the difference was also significant: females gained 2.42 ± 0.46 and 1.06 ± 0.50 kg in the study and control groups, respectively (p = 0.002). The difference between genders within the study group was not significant (2.60 ± 0.44 vs. 2.42 ± 0.46 kg among males and females, respectively, p = 0.743 Fig. 1C).

Age

To assess the impact of age as a risk factor for weight gain after switching to an INSTI-based regimen, we divided the participants into three age subgroups of ≤42, 43–51 and ≥ 52 years old at time 0 and compared the weight gain in the study group vs. the control group (Fig. 1D). In the youngest subgroup, patients gained weight similarly in the control and the study group (p = 0.271). A significant difference in weight gain between the study and the control group was observed in the two other age subgroups. While the control group gained 0.28 ± 0.56 kg in the 43–51 age subgroup and 0.61 ± 0.56 kg in the above 52 age subgroup over 2 years, a significantly higher weight gain was observed in both age subgroups within the study group (2.19 ± 0.53 kg in the 43–51 age subgroup and 2.56 ± 0.56 kg in the above 52 age subgroup (p = 0.004 in both comparisons, Fig. 1D)).

INSTIs type

Participants included in the study group were switched to one of the following INSTIs: Dolutegravir (DTG, n = 84), Elvitegravir/Cobicistat (EVG, n = 49) or Raltegravir (Ral, n = 41, Table 1). Participants gained 2.39 ± 0.47, 3.16 ± 0.61, and 2.096 ± 0.67 kg during the 2 years of treatment with DTG, EVG, and RAL, respectively; the differences were not statistically significant.

Backbone agent

We identified four main groups of PLWH within the study group according to the backbone drug they received before and after the switch to an INSTI-based regimen. Participants who were treated with Tenofovir disoproxil/Emtricitabine (TDF/FTC) before and after the switch (n = 70) gained 2.01 ± 0.51 kg; those who were treated with Abacavir/Lamivudine (ABC/3TC) before and after the switch (n = 24) gained 3.87 ± 0.87 kg. Participants who changed from TDF/FTC to Tenofovir-Alafenamide/Emtricitabine (TAF/FTC, n = 29) gained 2.91 ± 0.79 kg, and those who switched from TDF/FTC to ABC/3TC, (n = 16) gained 1.54 ± 1.07 kg. The differences between the groups were statistically non-significant (p = 0.396).

We also performed a multivariate logistic regression analysis for ≥5% weight gain during 24 months of follow-up. Switching to INSTI treatment (the study group), Caucasian origin, and initial low BMI were found to be significant risk factors for excessive weight gain (Fig. 2). These parameters were confirmed by a sensitivity analysis of the upper quintile of the relative difference in weight either within 1 year of follow-up or 2 years. We performed the same analysis to investigate whether other risk factors for excessive weight gain are significant after switching to INSTI, including only patients from the study group. This analysis indicated higher initial BMI at the switch was associated with a smaller weight gain after the switch (OR 0.87, CI 0.79–0.96; p = 0.0005).

Fig. 2
figure 2

Multivariate regression analysis for weight gain of ≥5% of initial body weight during 24 months. The width of the confidence interval (CI) line is proportional to the CI (a smaller CI is expressed in a wider line)

Metabolic parameters

To explore the effect of INSTIs on parameters related to metabolic syndromes such as hyperglycemia, hyperlipidemia, and hypertension, we examined whether the groups were comparable in terms of antidiabetic medications, lipid-lowering agents, and antihypertensive treatment status during the 2 years of follow-up. The proportion of participants who were already receiving these treatments at baseline, those who initiated such treatment during follow-up and those who did not necessitate such treatment was similar between the groups (p = 0.678, p = 0.493, and p = 0.298 for antihypertensive, antihyperglycemic and antihyperlipidemic treatment, respectively).

Glucose

Blood glucose levels were comparable between the two groups at baseline: 97.1 ± 28.0 mg/dL in the control group and 97.7 ± 28.2 mg/dL in the study group (p = 0.836). The average glucose levels in the control group were stable during the first year of follow-up (96.7 ± 28 mg/dL, p = 1.000), and by the end of the second year of follow-up, a slight decrease was observed (95.6 ± 25.4 mg/dL, p = 0.962). In contrast, the study group experienced a constant increase in glucose levels throughout the 2 years of follow-up, which reached 98.6 ± 33.8 mg/dL at the end of the follow-up period (p = 0.865). Generally, exploring the effect of race or gender, none of these parameters had an independent impact upon glucose levels changes. When we analyzed only the subgroup of African-origin males, there was a significantly higher increase in glucose levels in the study group (9.05 ± 3.38 mg/dL) during 2 years of follow-up after the switch compared to the control group, where glucose levels decreased by 1.6 + 3.38 mg/dL (p = 0.0009).

Mean blood pressure (MPB)

There was no difference between the groups in the change of MBP (p = 0.522).

Lipids’ profile

No statistically significant differences were noted between the groups regarding the measured fasting lipids: total cholesterol, HDL, LDL, and triglycerides over the two-years follow-up period. (p = 0.899, p = 0.269 p = 0.827 and p = 0.420, respectively).

Discussion

Our study adds to the existing data on INSTI-based ART and short-term weight gain. We report that PLWH who switched to an INSTI-based regimen gained more weight over 24 months than PLWH remaining on a non-INSTI-based regimen. Most of the difference was observed during the first year of follow-up; both groups demonstrated a similar rate of weight gain after that. Our results suggest that weight gain following the switch to an INSTI-based regimen, although statistically significant, is relatively moderate and may be limited only to the first year after the switch. A direct comparison between the groups showed a higher weight gain among older patients and African descendants in the study group. However, a multivariate logistic regression analysis did not confirm these results. Instead, it indicated that lower initial weight constitutes a risk factor for a significant weight gain (≥5%) among PLWH after a switch to an INSTI- based regimen.

Previous studies performed in different populations resulted in conflicting results, with some noting weight gain [7, 10, 13,14,15, 22,23,24], which was not confirmed by others [8]. The main differences between the studies relate to the magnitude of weight gain, the impact of weight gain on the development of comorbidities, and the differential effect of specific ART combinations.

Substantial weight gain has been reported in studies of treatment-naïve PLWH. Two open-label randomized African studies assessed the effect of Dolutegravir (DTG) as compared to Efavirenz (EFV, an NNRTI) in treatment-naïve PLWH. They showed an excessive weight gain induced by DTG of 5–8 kg vs. 2–3 kg induced by EFV [11, 23]. In a retrospective observational cohort study from the USA, DTG was associated with a 6 kg increase in weight compared to 2.6 kg with NNRTI treatment; 84% of the last group were treated with EFV (p < 0.05) [9]. In a pooled analysis of randomized controlled studies of treatment-naïve PLWH, INSTI-based regimens resulted in a more minor but still significant weight gain of 3.24 kg, in comparison to 1.93 kg with NNRTIs (p < 0.001) and 1.72 kg with PIs (PIs vs. NNRTIs, p = 0.6) over 96 weeks [13]. A minor degree of weight gain was reported by Norwood et al. and Saber et al. [14, 24], who enrolled ART-experienced and virologically suppressed PLWH who switched to INSTI-based regimens and showed a 2.7 kg [14] and 2 kg [24] weight gain in comparison to PLWH who continued NNRTI or PI-based regimens. Recently, pooled data from randomized controlled trials reported a 1.6 kg weight gain over 48 weeks following a switch to INSTI (EVG, DTG, or Bictegravir), in comparison to a 0.4 kg weight gain in patients remaining on their previous treatment, including PI-based regimens and NNRTI-based regimens (EFV, Rilpivarin or Nevirapine), [15].

Identifying risk factors associated with weight gain in PLWH treated with INSTI is a growing research topic. Sax et al. identified INSTI initiation, low initial CD4 count, high viral load, female gender, and black race as risk factors for weight gain among over 5000 patients who initiated ART in a pooled analysis [13]. The ADVANCE trial, which almost exclusively enrolled African patients, identified similar risk factors [23]. In contrast, Bourgi et al., who identified DTG as associated with the highest weight gain in ART-naïve PLWH, did not support any other risk factors, as neither gender nor race significant effected weight gain [9]. In a switch study, Lake et al. [25] identified African ethnicity, female gender, and age > 60 as risk factors for weight gain. Erlandson used pooled data of PLWH from switch studies [15] and observed that younger age and underweight represented risk factors for weight gain greater than 10% of initial weight, which is considered clinically significant. Unfortunately, the discrepancies between the studies and the analyses, including ours, stemming from different study designs (RCTs or observational), different patient characteristics (naive, treatment experienced and diverse populations), different diets and heterogeneity of ART make it challenging to identify the patients that are prone to weight gain following a treatment switch.

Our study did not identify an association between weight gain and specific ART regimens, probably due to the high heterogeneity of the ART regimens used and the limitation of a small sample size. Previous studies showed that 2nd generation INSTI, such as DTG or Bictegravir (BIC), as well as Rilpivirine (NNRTI) or TAF/FTC (NRTIs), are associated with the most pronounced weight gain effect [9, 13, 24].

Rebeiro et al. observed a higher risk of diabetes mellitus (Hazard ratio 1.17) in patients who initiated INSTI-based ART in comparison to NNRTIs or PIs [26]. Summers et al. found a tendency to develop hyperglycemia in virally controlled women who used INSTIs [27], which was not confirmed by others [28, 29]. Recently, Millic et al. observed improved glycemic control with the use of INSTI, except for those with a weight gain of ≥5% [22]. The only metabolic effect observed in our cohort other than weight gain was an increase in glucose levels following a switch from NNRTIs or PIs to INSTI, solely in the subgroup of African descendant males. Similarly to others [30], we did not identify a worsening of other metabolic parameters such as hypertension or dyslipidemia, following the treatment switch.

The mechanism driving weight gain in this setting remains elusive, and some explanations were suggested. A rapid reduction of HIV RNA by INSTI’s correlates with the lower patients resting energy, thus causing more weight gain after treatment initiation in naive PLWH [31]. The impaired adipogenesis and adipocyte metabolism elicited by EFV occurred in higher concentrations of EVL, while RAL had neutral actions of adipogenesis [32, 33]; RAL was shown to increase zonulin, a marker of integrity of intercellular tight junctions, thus probably reducing bacterial translocation from the gut and inflammation, another driving force of weight reduction [34]. Another suggested mechanism is the interference of some INSTIs with the melanocortin signaling system, which is involved in the regulation of energy homeostasis, food intake, and satiety [35]. Variations in weight gain may also derive from unique adverse effects caused by different regimens [36]. Common side effects of PIs treatment are severe nausea and diarrhea [37], and NNRTIs have a more prominent neuro-depressant effect, that can result in depression and reduced appetite [38, 39]. This explanation could be supported by the short-term excessive weight gain noted in our study as PLWH suffer less from the side effects of PIs and NNRTIs noted after ART switch to INSTI, thus gaining excessive weight mainly shortly after the switch. It is likely that a combination of these mechanisms may explain weight gain in treated patients.

The limitations of our study include a relatively small sample size, and heterogeneity of ART medications used in both study and control groups. Furthermore, our data is retrospective; therefore, patients whose weight was not consistently recorded were excluded. Notably, the retrospective observational nature of the study does not allow to make assumptions regarding causality.

In conclusion, switching to INSTI-based antiretroviral treatment resulted in a moderate weight gain over 2 years of follow-up. This obesogenic effect mainly developed in the first year after the treatment switch. In contrast, a diabetogenic effect noted in the second year after the switch to INSTIs was observed only in a subgroup of African origin males. Although our results did not show a clinically significant weight gain following the switch to INSTI in most of the participants, the potential risk of weight gain should be discussed with patients before starting INSTI-based regimens. Both patients and their caretakers should be encouraged to closely monitor weight and long-term related metabolic features, while enhancing physical activity.

Availability of data and materials

All data is available and will be provided upon request from the last author, Dr. Hila Elinav, hilaelinav@gmail.com.

References

  1. Drug and food administration. Application number 22–145 approval letter 2007. Available from: https://www.accessdata.fda.gov/drugsatfda_docs/nda/2007/022145_Isentress.cfm.

  2. National institue of Health (.gov). Guidelines for the Use of Antiretroviral Agents in Adults and Adolescents Living with HIV 2022 [cited 2022]. Available from: https://clinicalinfo.hiv.gov/en/guidelines/adult-and-adolescent-arv/what-start-initial-combination-regimens-antiretroviral-naive.

  3. Arribas J MJ, Buckely RDM,Antinori A,Bracchi M,Calmy A,Dedes N,Horban A, Katlama C,Latysheva I,Lundgren JD, McCormack S,Mussini C,Pozniak A,Pulido F,Raffi F,Stelbrink HJ,Van der Vallk M, Vasylyev M. European AIDS clinical Society Guidlines 2021 [updated 10.2021; cited 2021 20.11.2021]. 11:[Available from: https://www.eacsociety.org/guidelines/eacs-guidelines/.

  4. Saag MS, Gandhi RT, Hoy JF, Landovitz RJ, Thompson MA, Sax PE, et al. Antiretroviral drugs for treatment and prevention of HIV infection in adults: 2020 recommendations of the international antiviral society-USA panel. JAMA. 2020;324(16):1651–69. Epub 2020/10/15. https://doi.org/10.1001/jama.2020.17025.

    Article  CAS  PubMed  Google Scholar 

  5. Kolakowska A, Maresca AF, Collins IJ, Cailhol J. Update on Adverse Effects of HIV Integrase Inhibitors. Curr Treat Options Infect Dis. 2019;11(4):372–87. https://doi.org/10.1007/s40506-019-00203-7.

    Article  PubMed  PubMed Central  Google Scholar 

  6. Bakal DRCL, Luz PM, Clark JL, Boni RBD, Cardoso SW, Veloso VG, et al. Obesity following ART initiation is common and influenced by both traditional and HIV−/ART-specific risk factors. J Antimicrob Chemother. 2018;73(8):2177–85. https://doi.org/10.1093/jac/dky145.

    Article  PubMed  PubMed Central  Google Scholar 

  7. McComsey GAEJ, Santiago S, Mounzer K, Moyle G, Vantig T, Sax PE, et al. Weight gain during treatment among 3,468 treatment-experienced adults with HIV. Seattle, Washington: Conference on retroviruses and opportunistic infections; 2019.

    Google Scholar 

  8. Burns JE, Stirrup OT, Dunn D, Runcie-Unger I, Milinkovic A, Candfield S, et al. No overall change in the rate of weight gain after switching to an integrase-inhibitor in virologically suppressed adults with HIV. AIDS. 2020;34(1):109–14. https://doi.org/10.1097/QAD.0000000000002379.

    Article  CAS  PubMed  Google Scholar 

  9. Bourgi K, Rebeiro PF, Turner M, Castilho JL, Hulgan T, Raffanti SP, et al. Greater Weight Gain in Treatment-naive Persons Starting Dolutegravir-based Antiretroviral Therapy. Clin Infect Dis. 2020;70(7):1267–74. https://doi.org/10.1093/cid/ciz407. PubMed PMID: 31100116; PubMed Central PMCID: PMCPMC8205610

    Article  CAS  PubMed  Google Scholar 

  10. Group NAS, Kouanfack C, Mpoudi-Etame M, Omgba Bassega P, Eymard-Duvernay S, Leroy S, et al. Dolutegravir-based or low-dose Efavirenz-based regimen for the treatment of HIV-1. N Engl J Med. 2019;381(9):816–26. https://doi.org/10.1056/NEJMoa1904340.

    Article  Google Scholar 

  11. Hill AVF, Delaporte E, Sokhela S, Kouanfack C, Moorhouse M, McCann K, et al. Progressive rises in weight and clinical obesity for TAF/FTC+DTG and TDF/FTC+DTG versus TDF/FTC/EFV: ADVANCE and NAMSAL trials. Mexico City: IAS 2019: conference on HIV pathogenesis; 2019.

    Google Scholar 

  12. Kerchberger AM, Sheth AN, Angert CD, Mehta CC, Summers NA, Ofotokun I, et al. Weight Gain Associated With Integrase Stand Transfer Inhibitor Use in Women. Clin Infect Dis. 2020;71(3):593–600. https://doi.org/10.1093/cid/ciz853.

    Article  CAS  PubMed  Google Scholar 

  13. Sax PE, Erlandson KM, Lake JE, McComsey GA, Orkin C, Esser S, et al. Weight Gain Following Initiation of Antiretroviral Therapy: Risk Factors in Randomized Comparative Clinical Trials. Clin Infect Dis. 2020;71(6):1379–89. https://doi.org/10.1093/cid/ciz999.

    Article  CAS  PubMed  Google Scholar 

  14. Saber SBA, Sparks A, Siegel M. Weight change associated with antiretroviral therapy switch to integrase Strand transfer inhibitor-based regimens. Open forum. Infect Dis Ther. 2019:6. https://doi.org/10.1093/ofid/ofz360.407.

  15. Erlandson KM, Carter CC, Melbourne K, Brown TT, Cohen C, Das M, et al. Weight change following antiretroviral therapy switch in people with viral suppression: pooled data from randomized clinical trials. Clin Infect Dis. 2021; https://doi.org/10.1093/cid/ciab444. PubMed PMID: 33987636

  16. Taramasso L, Ricci E, Menzaghi B, Orofino G, Passerini S, Madeddu G, et al. Weight Gain: A Possible Side Effect of All Antiretrovirals. Open Forum Infect Dis. 2017;4(4):ofx239. https://doi.org/10.1093/ofid/ofx239.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Achhra AC, Mocroft A, Reiss P, Sabin C, Ryom L, de Wit S, et al. Short-term weight gain after antiretroviral therapy initiation and subsequent risk of cardiovascular disease and diabetes: the D:a:D study. HIV Med. 2016;17(4):255–68. Epub 2015/07/29. https://doi.org/10.1111/hiv.12294.

    Article  CAS  PubMed  Google Scholar 

  18. Samaras KWH, Law M, Emery S, Cooper D, Carr A. Prevalence of metabolic syndrome in HIV-infected patients receiving highly active antiretroviral therapy using international Diabetes Foundation and adult treatment panel III criteria: associations with insulin resistance, disturbed body fat compartmentalization, elevated C-reactive protein, and [corrected] hypoadiponectinemia. Diabetes Care. 2007;30(1):113–9. https://doi.org/10.2337/dc06-1075.

    Article  CAS  PubMed  Google Scholar 

  19. Herrin M, Tate JP, Akgun KM, Butt AA, Crothers K, Freiberg MS, et al. Weight Gain and Incident Diabetes Among HIV-Infected Veterans Initiating Antiretroviral Therapy Compared With Uninfected Individuals. J Acquir Immune Defic Syndr. 2016;73(2):228–36. https://doi.org/10.1097/QAI.0000000000001071.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Kumar S, Samaras K. The Impact of Weight Gain During HIV Treatment on Risk of Pre-diabetes, Diabetes Mellitus, Cardiovascular Disease, and Mortality. Front Endocrinol (Lausanne). 2018;9:705. https://doi.org/10.3389/fendo.2018.00705.

    Article  PubMed  Google Scholar 

  21. Mankal PK, Kotler DP. From wasting to obesity, changes in nutritional concerns in HIV/AIDS. Endocrinol Metab Clin N Am. 2014;43(3):647–63. Epub 2014/08/30. https://doi.org/10.1016/j.ecl.2014.05.004.

    Article  Google Scholar 

  22. Milic J, Renzetti S, Ferrari D, Barbieri S, Menozzi M, Carli F, et al. Relationship between weight gain and insulin resistance in people living with HIV switching to integrase strand transfer inhibitors-based regimens. AIDS. 2022;36(12):1643–53. Epub 2022/06/22. https://doi.org/10.1097/QAD.0000000000003289.

    Article  CAS  PubMed  Google Scholar 

  23. Venter WDF, Moorhouse M, Sokhela S, Fairlie L, Mashabane N, Masenya M, et al. Dolutegravir plus two different prodrugs of Tenofovir to treat HIV. N Engl J Med. 2019;381(9):803–15. Epub 2019/07/25. https://doi.org/10.1056/NEJMoa1902824.

    Article  CAS  PubMed  Google Scholar 

  24. Norwood J, Turner M, Bofill C, Rebeiro P, Shepherd B, Bebawy S, et al. Brief Report: Weight Gain in Persons With HIV Switched From Efavirenz-Based to Integrase Strand Transfer Inhibitor-Based Regimens. J Acquir Immune Defic Syndr. 2017;76(5):527–31. https://doi.org/10.1097/QAI.0000000000001525.

    Article  PubMed  PubMed Central  Google Scholar 

  25. Lake JE, Wu K, Bares SH, Debroy P, Godfrey C, Koethe JR, et al. Risk Factors for Weight Gain Following Switch to Integrase Inhibitor-Based Antiretroviral Therapy. Clin Infect Dis. 2020;71(9):e471–e7. https://doi.org/10.1093/cid/ciaa177.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. JC RRF, Bian A, Lake JE, Bourgi K, Moore RD, Horberg MA, et al. Risk of Incident Diabetes Mellitus, Weight Gain, and Their Relationships With Integrase Inhibitor-Based Initial Antiretroviral Therapy Among Persons With Human Immunodeficiency Virus in the United States and Canada. Clin Infect Dis. 2021;73(7):e2234–e42. https://doi.org/10.1093/cid/ciaa1403.

    Article  CAS  Google Scholar 

  27. Summers NA, Lahiri CD, Angert CD, Aldredge A, Mehta CC, Ofotokun I, et al. Metabolic Changes Associated With the Use of Integrase Strand Transfer Inhibitors Among Virally Controlled Women. J Acquir Immune Defic Syndr. 2020;85(3):355–62. https://doi.org/10.1097/QAI.0000000000002447.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Zimmerman MDJ, Schafer JJ. Exploring the prevalence and characteristics of weight gain and other metabolic changes in patients with HIV infection switching to integrase inhibitor containing ART. Open Forum Infect Dis. 2019;6:S176–7. https://doi.org/10.1093/ofid/ofz360.405.

    Article  PubMed Central  Google Scholar 

  29. Hsu RBL, Fusco JS, Mounzer K, Vannappagari V, Henegar C, van Wyk JA, et al. Risk of type 2 diabetes mellitus after antiretroviral therapy initiation in individuals living with HIV in the United States. Open Forun Infectious Diseases. 2019;6(S2):S181–S2. https://doi.org/10.1093/ofid/ofz360.414.

    Article  Google Scholar 

  30. Aldredge ALC, Summers NA, Mehta CC, Angert CD, Kerchberger AM, Sheri Weiser S, et al. Effects of Integrase Strand-Transfer Inhibitor Use on Lipids, Glycemic Control, and Insulin Resistance in the Women’s Interagency HIV Study (WIHS). Open Forum Infect Dis. 2019;supp 2:S38. https://doi.org/10.1093/ofid/ofz359.082.

    Article  Google Scholar 

  31. Mulligan K, Tai VW, Schambelan M. Energy expenditure in human immunodeficiency virus infection. N Engl J Med. 1997;336(1):70–1. Epub 1997/01/02. 8984340. https://doi.org/10.1056/NEJM199701023360115.

    Article  CAS  PubMed  Google Scholar 

  32. Moure R, Domingo P, Gallego-Escuredo JM, Villarroya J, Gutierrez Mdel M, Mateo MG, et al. Impact of elvitegravir on human adipocytes: alterations in differentiation, gene expression and release of adipokines and cytokines. Antivir Res. 2016;132:59–65. Epub 2016/05/25. https://doi.org/10.1016/j.antiviral.2016.05.013.

    Article  CAS  PubMed  Google Scholar 

  33. Gorwood J, Bourgeois C, Pourcher V, Pourcher G, Charlotte F, Mantecon M, et al. The integrase inhibitors Dolutegravir and Raltegravir exert Proadipogenic and Profibrotic effects and induce insulin resistance in human/simian adipose tissue and human adipocytes. Clin Infect Dis. 2020;71(10):e549–e60. Epub 2020/03/14. https://doi.org/10.1093/cid/ciaa259.

    Article  CAS  PubMed  Google Scholar 

  34. El Kamari V, Moser C, Hileman CO, Currier JS, Brown TT, Johnston L, et al. Lower Pretreatment Gut Integrity Is Independently Associated With Fat Gain on Antiretroviral Therapy. Clin Infect Dis. 2019;68(8):1394–401. https://doi.org/10.1093/cid/ciy716.

    Article  CAS  PubMed  Google Scholar 

  35. Domingo PVF, Giralt M, Domingo JC. Potential role of the melanocortin signaling system interference in the excess weight gain associated to some antiretroviral drugs in people living with HIV. Int J Obes. 2020;44:1970–3.

    Article  CAS  Google Scholar 

  36. Raffi F, Esser S, Nunnari G, Perez-Valero I, Waters L. Switching regimens in virologically suppressed HIV-1-infected patients: evidence base and rationale for integrase strand transfer inhibitor (INSTI)-containing regimens. HIV Med. 2016;17(Suppl 5):3–16. Epub 2016/10/08. https://doi.org/10.1111/hiv.12440.

    Article  CAS  PubMed  Google Scholar 

  37. Brites C, Nobrega I, Luz E, Travassos AG, Lorenzo C, Netto EM. Raltegravir versus lopinavir/ritonavir for treatment of HIV-infected late-presenting pregnant women. HIV Clin Trials. 2018;19(3):94–100. Epub 2018/04/10. https://doi.org/10.1080/15284336.2018.1459343.

    Article  CAS  PubMed  Google Scholar 

  38. Munoz-Moreno JA, Fumaz CR, Ferrer MJ, Gonzalez-Garcia M, Molto J, Negredo E, et al. Neuropsychiatric symptoms associated with efavirenz: prevalence, correlates, and management. A neurobehavioral review AIDS Rev. 2009;11(2):103–9.

    PubMed  Google Scholar 

  39. Lennox JL, DeJesus E, Lazzarin A, Pollard RB, Madruga JV, Berger DS, et al. Safety and efficacy of raltegravir-based versus efavirenz-based combination therapy in treatment-naive patients with HIV-1 infection: a multicentre, double-blind randomised controlled trial. Lancet. 2009;374(9692):796–806. Epub 2009/08/04. https://doi.org/10.1016/S0140-6736(09)60918-1.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank Professor Eran Elinav, for the scientific review and linguistic editing.

Funding

Dr. Maman is the first author, and the work was part of the requirements of the Hebrew University for a medical doctor degree. The study was not funded by any grant, organization, or company.

Author information

Authors and Affiliations

Authors

Contributions

All authors contributed to the manuscript. Dr. Mamam collected the data, and wrote the manuscript, Dr. Wiessam Abu Ahmad performed the statistical analysis. Both have equally contributed to the work. Dr. Ofer Perzon performed the statistical analysis. Dr. Keren Mahlab-Guri collected data and reviewed the manuscript. Dr. Daniel Elbirt conceived and designed the study, collected data, and reviewed the manuscript. Dr. Hila Elinav collected data, supervised the analyses, and wrote the manuscript. She is listed as the corresponding and the last author. Dr. Elbirt and Dr. Elinav have equally contributed to the work.

Corresponding author

Correspondence to Hila Elinav.

Ethics declarations

Ethics approval and consent to participate

This research was approved by the Hadassah-Hebrew University Medical Center Institutional Review Board (IRB) (0259–19-HMO) and the Kaplan Medical Center ethics committee (KMC-0036-10). Due to the retrospective design of the research, a waiver of informed consent of the participants in the study was provided by both ethics committees.

Consent for publication

Not applicable. Due to the retrospective design of the research, and the usage of anonymized data, a waiver of informed consent of the participants was approved by the both institutions ethics committees, thus approval for publication by the participants is not requested as well.

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Maman, O., Ahmad, W.A., Perzon, O. et al. The effect of a treatment switch to integrase Strand transfer inhibitor–based regimens on weight gain and other metabolic syndrome-related conditions. BMC Infect Dis 24, 221 (2024). https://doi.org/10.1186/s12879-024-09120-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12879-024-09120-7

Keywords