Enhanced metabolic health and immune response with bictegravir/emtricitabine/TAF: Insights from a 96‑week retrospective study

  • Authors:
    • Serena Spampinato
    • Giuseppe Nicolò Conti
    • Andrea Marino
    • Vincenzo Raimondo
    • Benedetto Maurizio Celesia
    • Giovanni Francesco Pellicanò
    • Mariangela Valentina Puci
    • Giovanni Sotgiu
    • Roberto Bruno
    • Nunziatina Villari
    • Alessia Mirabile
    • Viviana Agata Maria Coco
    • Michele Salvatore Paternò Raddusa
    • Eugenia Pistarà
    • Vincenzo Boscia
    • Viviana Fisicaro
    • Giorgia Fiorenza
    • Bruno Cacopardo
    • Emmanuele Venanzi Rullo
    • Giuseppe Nunnari
  • View Affiliations

  • Published online on: October 2, 2024     https://doi.org/10.3892/br.2024.1867
  • Article Number: 179
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Bictegravir/emtricitabine/tenofovir alafenamide (B/F/TAF), as a fixed dosed combination, is effective in people living with human immunodeficiency virus (PLWH) previously treated with other therapeutic regimens. The aim of the present retrospective observational real‑life study was to analyze virological suppression and immunological, metabolic and safety profile of B/F/TAF. Data were collected from 127 PLHW who switched from any regimen to B/F/TAF. Viral load and virological suppression (viral load <50 copies/ml) were assessed by using real‑time PCR methodologies; CD4 and CD8 T cell count as well as CD4/CD8 ratio were determined by cytofluorimetric analyses; other metabolic parameters such as total cholesterol, triglycerides, High‑ and Low‑Density Lipoproteins were assessed by using immunoenzymatic assay. All of the aforementioned parameters were assessed at different timepoints (Baseline, 48 and 96 weeks) for the patients switching to B/F/TAF. Of 127 PLHW [96 (75.6%) male and 31 (24.4%) female, with a mean age of 46.8±10.7 years], 107 PLHW were included in the analysis. The percentage of virologically suppressed PLWH increased from 66.4 to 74.8% at 96 weeks. A statistically significant increase in absolute CD4 (P<0.0001) and CD8 T cell count (P=0.002) was observed. Of importance, there was a significant increase in CD4/CD8 ratio from 0.95 (0.52‑1.31) to 1.16 (0.75‑1.39) (P=0.003) after 96 weeks. There was a significant decrease in the median values ​​of triglycerides (P<0.0001) and total cholesterol (P<0.0001). Serum creatinine showed a significant increase (P=0.0001). In real life, switching to B/F/T was safe and highly effective both virologically and immunologically. Decrease in cholesterol and triglyceride levels suggested a favorable metabolic profile, which may decrease inflammation, leading to a healthier state and less organ damage.

Introduction

The management of human immunodeficiency virus (HIV) infection has evolved over the past decades, with antiretroviral therapy (ART) serving a critical role in achieving virological suppression and improving immune function. Despite the efficacy of various ART regimens, the need for optimizing treatment persists to enhance patient outcomes, minimize adverse effects and treat comorbidities and associated infection (1-3).

Novel ART regimens are designed to be highly potent while maintaining a better safety profile. Integrase strand inhibitors (INSTIs) have emerged as a cornerstone in first-line treatment regimens. These include first-generation INSTIs such as raltegravir and elvitegravir and second-generation agents such as dolutegravir, bictegravir and cabotegravir (4,5-7). Bictegravir, available as a fixed-dose combination tablet with the nucleos(t)ide reverse transcriptase inhibitors (NRTIs) emtricitabine and tenofovir alafenamide (B/F/TAF), has been approved both by the U.S. Food and Drug Administration and European Medicines Agency for once-daily treatment. Landmark phase III trials have demonstrated its efficacy in both treatment-naïve people living with HIV (PLWH) and as a switch regimen (4-7). B/F/TAF does not require pre-treatment molecular detection of Human Leukocyte Antigen B allele, which has already been associated with hypersensitivity to the nucleoside-reverse transcriptase inhibitor (NRTI) abacavir (8). The B/F/TAF regimen exhibits activity against hepatitis B virus and HIV-1 non-B subtypes and is suitable for ‘rapid start’ treatment, since it is has a high genetic barrier against resistance development (9). Additionally, it has low drug-drug interactions, comes as a fixed-dose combination and has simple administration guidelines regarding meals, making it a convenient option for PLWH (4,10). As an effective switch strategy, B/F/TAF has shown non-inferiority to continuing a number of regimens, including those with pre-existing NRTI mutations, in several randomized controlled trials (4-7,9,10). Real-world studies further support the virological efficacy and safety of B/F/TAF as a switch regimen (4-7,9,10).

The aim of the present retrospective study was to evaluate the efficacy of B/F/TAF in a real-world setting, focusing on virological, immunological, safety and metabolic profiles.

Materials and methods

Cohort

Data from a cohort of 127 PLWH enrolled from April 2019 to May 2022 at ARNAS Garibaldi in Catania, Italy, were statistically analyzed to determine a safety and efficacy profile of B/F/TAF. Cohort's age ranged from 23 to 72 years, with a mean age of 46.8 (±10.7) years. The majority of the cohort (75,6%) was composed by male individuals, and sexual transmission was the main cause of infection (88.1%).

The inclusion criteria were as follows: i) HIV-1 infection, ii) age >18 years and iii) written informed consent. Exclusion criteria were pregnancy and other ongoing treatments incompatible with B/F/TAF.

Measurements

Epidemiological and clinical data were collected from 107 patients. The epidemiological data gathered at baseline included age, sex, ethnicity, previous regimen, height, weight, comorbidities (such as viral co-infection, hypertension, obesity, diabetes, dyslipidaemia, cancer, osteoporosis, hyperthyroidism, hypothyroidism and psychiatric disease), acquired immune deficiency syndrome (AIDS)-associated diseases (oesophageal candidiasis, Kaposi's sarcoma, cerebral toxoplasmosis, lymphoma, Pneumocystis jirovecii pneumonia, progressive multifocal leukoencephalopathy, atypical mycobacterial infection, cervical carcinoma, hepatitis B or C and other co-infection), time since onset of HIV, CD4 nadir, sexual orientation, transmission mode, recreational habits, cigarette smoking and alcohol consumption. Clinical parameters were assessed using clinical laboratory automated instrumentation: High- and low-density lipoproteins (HDL and LDL), triglycerides, Total cholesterol, creatinine, Glutamate-Pyruvate transaminase (GPT) and Glutamate-Oxalate Transaminase (GOT) were assessed using the DxC 500 AU Chemistry Analyzer by Beckman Coulter; HIV Viral Load (VL) was analyzed using the HIV-1 COBAS test on COBAS 4800 (Roche Diagnostics Corporation); CD4+ and CD8+ T lymphocytes levels were assessed using cytofluorimetric automated instrumentation BD Facs Canto II (BD Biosciences) using the BD Multitest™ CD3 FITC/CD8 PE/CD45 PerCP/CD4 APC with BD Trucount™ Tubes (BD Biosciences), a specific four-color reagent useful to determine the absolute count of CD4+ and CD8+ lymphocytes per microliter by gating all mature lymphocytes (CD3+) using a specific fluorophore (Fluorescein Isothiocyanate, FITC). Mature lymphocytes expressing both CD3+ and CD8+ glycoproteins are gated by assessing the fluorescence of both the respective fluorophores (FITC and Phycoerithrin). The same process is performed also for the CD4+ T lymphocytes, using Allophycocyanin (APC) as a specific fluorophore. The total leukocytes count is performed by targeting CD45+ cluster of differentiation, expressed on all white blood cells. Peridinin-Chlorophyll-Protein (PerCP) was used as the specific fluorophore for CD45+ glycoprotein. This allows to obtain also the relative quantification of both CD4+ and CD8+ cells.

Statistical analysis

Qualitative data were summarized using absolute and relative frequencies. Quantitative variables are presented as either mean and SD or medians and interquartile range. The normal distribution was evaluated using Shapiro-Wilk test. The McNemar test was used to compare qualitative variables between study time-points. Comparisons of biochemical parameters were evaluated using either one-way ANOVA for repeated measures followed by post hoc Bonferroni's test or the non-parametric Friedman test. P<0.05 was considered to indicate a statistically significant difference. STATA17 software (2021. Stata: Release 17; StataCorp LLC.) was used for statistical computations.

Results

Cohort

A total of 127 patients were enrolled, 96 (75.6%) male and 31 (24.4%) female, with a mean age of 46.8±10.7 years (Tables I and II), at the Infectious Disease Unit of the Hospital Azienda di Rilievo Nazionale ed Alta Specializzazione Garibaldi, University of Catania (Catania, Italy). Patients were predominantly Caucasian (88.1%), heterosexual (54.7%), men who wave sex with men (37.6%), with some bisexual (7.7%); thus, the mode of HIV transmission was primarily sexual (88.1%; Table I). A total of 49.6% of patients were smokers, 12.6% used alcohol, 9.5% were drug users and 11% reported substance abuse (Table I). The primary therapeutic regimens were elvitegravir + cobicistat + Emtricitabine (FTC) + TAF (53.54%), dolutegravir + TAF + FTC (7.88%), raltegravir + TAF + FTC (7.09%) and darunavir + cobicistat + FTC + TAF (7.09%). The primary reasons for treatment switch were proactive strategy (34.52%), simplification (27.38%) or adverse events (21.42%).

Table I

Patient characteristics.

Table I

Patient characteristics.

VariableValue
Mean age, years46.8±10.7
Male (%)96 (75.6)
Men who have sex with men (%)44 (37.6)
Ethnicity (%) 
     African10 (7.9)
     Arabic1 (0.8)
     Asian1 (0.8)
     Caucasian111 (88.1)
     Latin American3 (2.4)
Median years since HIV onset (IQR)14.1 (8.8-25.1)
Median CD4 nadir (IQR)283 (122-398)
Sexual orientation (%) 
     Bisexual9 (7.7)
     Heterosexual64 (54.7)
     Homosexual44 (37.6)
Transmission route (%) 
     Sexual103 (88.1)
     Accidental exposure1 (0.8)
     Drug use12 (10.3)
     Transfusion1 (0.8)
Substance abuse (%)14 (11.0)
Smoking status (%) 
     Non-smoker56 (44.1)
     Current smoker63 (49.6)
     Former smoker8 (6.3)
Drug use (%)12 (9.5)
Alcohol use (%)16 (12.6)

[i] HIV, human immunodeficiency virus.

Table II

Patient comorbidities.

Table II

Patient comorbidities.

Variable (%)Value
Comorbidity108 (85.0)
Hypertension27 (21.3)
Dyslipidemia36 (28.4)
Cancer6 (4.7)
Diabetes6 (4.7)
Osteoporosis4 (3.2)
Hypothyroidism3 (2.4)
Hyperthyroidism3 (2.4)
Psychiatric disorder19 (15.0)
AIDS-associated disease76 (59.8)
Co-infection69 (54.3)

[i] AIDS, acquired immune deficiency syndrome.

Virological trends

At baseline, 71 patients had viral load <50 copies/ml (Table III) while 36 had viral load >50 copies/ml. Over time, no statistically significant changes in the number or percentage of virus-suppressed patients were recorded, although the percentage of virologically suppressed PLWH increased from 66.4 to 67.3 after 48 weeks and to 74.8% at 96 weeks. Only one patient had a viral load >200 copies/ml (212 copies/ml) at 96 weeks.

Table III

Number of virologically suppressed patients (<50 copies/ml).

Table III

Number of virologically suppressed patients (<50 copies/ml).

Time pointPatients with <50 copies/ml (%)
Baseline71 (66.4)
48 weeks72 (67.3)
96 weeks80 (74.8)
Immunological profile

A significant increase in absolute CD4 cell count was observed from 658 (459.5-900.5) to 726 (562-974) after 48 weeks and to 882.5 (647.5-1169) cells/µl after 96 weeks (Table IV; Fig. 1A). Furthermore, the CD8 lymphocyte count significantly increased. Baseline values of 760.5 (511.0-1044.5) were recorded, while after 48 weeks the values were 837.6 (641-1066) and after 96 weeks 859.5 (648.5-1181; Fig. 1B) cells/µl. Finally, there was a significant increase in CD4/CD8 ratio from 0.95 (0.52-1.31) to 1.00 (0.61-1.29) after 48 weeks and 1.16 (0.75-1.39) after 96 weeks (Fig. 1C).

Table IV

CD4 and CD8 cell count and CD4/CD8 ratio.

Table IV

CD4 and CD8 cell count and CD4/CD8 ratio.

VariableBaseline48 weeks96 weeksP-value
Median CD4 cell count (IQR)658.00 (459.50-900.50)726.00 (562.00-974.00)882.50 (647.50-1169.00)<0.0001
Median CD8 cell count (IQR)760.50 (511.00-1,044.50)837.60 (641.00-1066.0)859.50 (648.50-1181.00)0.0020
Median CD4/CD8 ratio (IQR)0.95 (0.50-1.30)1.00 (0.6-1.29)1.16 (0.75-1.39)0.0100
Metabolic and clinical trends

At a metabolic level, there was a significant decrease in the median values of triglyceride levels from a median value of 128 (91-200) at baseline to 105 (73-143.5) at 48 and 100 (74-135) mg/dl at the 96 weeks (Table V; Fig. 1C). A significant decrease was also observed for total cholesterol levels from a mean value of 204.2±47.5 at baseline to 188.6±38.5 after 48 weeks and 190.1±38.8 mg/dl after 96 weeks (Fig. 1E). Serum creatinine showed a significant increase from 0.83 (0.73-0.91) to 0.91 (0.81-1.02) after 48 weeks and to 0.90 (0.82-1.02) mg/dl after 96 weeks (Fig. 1F). Finally, significant changes in median GPT values were found from 26.5 (18.5-38.5) at baseline to 29 (20-38.5) U/l after 48 weeks. The value decreased to 25.5 (20-40) U/l after 96 weeks. Median GPT values remained between normal ranges of 5 to 35 U/l.

Table V

Triglyceride, total cholesterol, serum creatinine and GPT levels.

Table V

Triglyceride, total cholesterol, serum creatinine and GPT levels.

VariableBaseline48 weeks96 weeksP-value
Median triglyceride levels, mg/dl (IQR)128.00 (91.00-200.00)105.00 (73.00-143.50)100.00 (74.00-135.00)<0.0001
Mean total cholesterol, mg/dl204.20±47.50188.60±38.50190.10±38.80<0.0001
Median serum creatinine, mg/dl (IQR)0.83 (0.73-0.91)0.91 (0.81-1.02)0.90 (0.82-1.02)0.0001
Median GPT levels, U/l (IQR)26.50 (18.50-38.50)29.00 (20.00-38.50)25.50 (20.00-40.00)0.0200

[i] GPT, glutamate pyruvate transaminase.

Discussion

The present study confirmed the virological and immunological efficacy of B/F/TAF, as well as a favorable metabolic profile. B/F/TAF led to a statistically significant increase in CD4/CD8 ratio >1, which is clinically associated with a lower probability of serious non-AIDS events (11,12).

Success of ART is measured by virological suppression (viral load <50/copies/ml) or the ability to maintain viral load <200 copies/ml (8,13,14). ART can lower the viral load not only in blood, but also in the semen and cervicovaginal and anorectal secretions, reducing the risk of sexual transmission (14,15). Hence, viral suppression is crucial in HIV treatment. Switching to B/F/TAF has been shown to maintain viral load <50 copies in previous studies (16-31). The present study confirmed the aforementioned data, showing a higher percentage of patients who were virus-suppressed at 96 weeks.

HIV exhibits a specific tropism for CD4 lymphocytes, targeting and killing these cells and decreasing their plasma concentration (32,33). ART lowers viral load, which leads to an increase in CD4 count (34,35). A higher CD4 cell count is associated with decreased risk of clinical progression in PLWH (12). In the literature, patients who switch to B/F/TAF maintained the CD4 count induced by previous therapy (18,25,26). Absolute CD4 count increases but to the best of our knowledge, only Chen et al (18), Lazzaro et al (25) and Lazzaro et al (26) found a significant increase in CD4 count in patients who switched to B/F/TAF.

During the asymptomatic stage of HIV infection, naive CD8 T cells are progressively depleted (36,37). Although the total number of CD8 T cells increases during this stage, the number of naive CD8 T cells decreases in parallel with overall CD4 T cell count (36,37). The use of ART during primary HIV infection helps preserve HIV-specific CD8+ T cells both physically and functionally, while sustaining HIV-specific T helper cells (38). Patients who maintain high CD8 lymphocyte count (>1,500 cells/µl) despite ART are at a higher risk of developing adverse events (12). Conditions with increased incidence in these patients include myocardial infarction (39), restenosis following coronary stenting (40), cancer (41) and non-AIDS-associated mortality (42). CD4/CD8 ratio is an important marker of inflammation and associated with serious non-AIDS events (12). As aforementioned, ART typically increases CD4 count and CD4/CD8 ratio (11,12). Low CD4/CD8 ratio is associated with older age in PLWH (43). Chronic HIV infection has been observed to cause immunological changes similar to those seen in natural aging (44). Low CD4/CD8 ratio is associated with impaired vaccine response (45), bacterial (46), fungal (47) and zoonotic infections (48), myocardial infarction (43), cancer (49) and non-AIDS mortality (12). The present study found a significant increase in both CD4 and CD8 T cell count and CD4/CD8 ratio in patients who switched to B/F/TAF. Despite the increase in CD8 T cells, the simultaneous increase in CD4 T cells increased CD4/CD8 ratio. Similar results were observed in studies that measured both CD8 count and CD4/CD8 ratio, with both metrics showing significant increases (25,26).

HIV-1, via its accessory protein Nef, impairs the efflux of cholesterol from macrophages (50). This virus-mediated shift redirects cholesterol transport from normal physiological efflux to virus-controlled transport, thereby decreasing the cell ability to export excess cholesterol (50). ART is not always successful in keeping triglyceride or LDL levels low (51). This is due to several factors, including continuous inflammation and immune activation (as ART achieves viral suppression but not elimination), mitochondrial dysfunction and altered distribution of adipose tissue (52). Frequent dyslipidemia leads to increased cardiovascular risk in PLWH (52,53). Switching to B/F/TAF has been shown to be effective in maintaining lipid levels in the normal range: Most studies have found no significant changes in lipid profile (17,20,27,28,31,54,55). However, certain studies have showed significant decreases in triglyceride (18,19,21,22,56), total cholesterol (18,25,30,55) and LDL levels (18,25,30) and total cholesterol/HDL ratio (19,25,56) and increased HDL levels (18,21,25). Chen et al (18) revealed also a significant increase in patients taking lipid lowering agents. Lazzaro et al (26) confirmed that after 96 weeks, lipid values remained constant. Here, significant decreases in triglyceride and total cholesterol levels were recorded, suggesting that switching to B/F/TAF may lead to an improvement in lipid profile.

Serum creatinine concentration is serves as a measure of the glomerular filtration rate and is commonly used as an index of renal function in clinical practice (57,58). In HIV infection, renal damage can occur due to both the infection and the therapy (59), particularly with the use of TAF (60). Significant increases in serum creatinine were recorded by Lazzaro et al (25,26), a small increase was reported by Kityo et al (22), while Molina et al (28) did not record significant changes. Here, a significant increase in serum creatinine levels was reported in patients switching to B/F/TAF. However, values remained within normal range (0.63-1.16 for male and 0.48-0.93 mg/dl for female patients) (61).

GPT is an enzyme used as a marker of hepatocyte lysis, systemic inflammation and oxidative stress (62). The persistent activation of immune cells by HIV, along with ongoing inflammation and oxidative stress related to HIV infection, leads to increased GPT levels, even in patients on Highly Active Antiretroviral Therapy (HAART) (63). Lazzaro et al (25,26) showed a decrease in GPT levels at 48 weeks and increase at 96 weeks in patients who switched to B/F/TAF, however, these changes were not significant. Another study found that naïve patients on B/F/TAF experienced decrease in GPT levels, while patients who switched to B/F/TAF treatment showed an increase in GPT levels (56). The present study showed significant changes in GPT levels: At 48 weeks there was an increase, but at 96 weeks there was a decrease. However, the median value did not exceed the cut-off threshold of 40 U/l, a value beyond which it becomes an indicator of liver damage (64,65).

One of the primary limitations of the present study is the small samples size (127 patients). This is mainly due to the real-world nature of the study, with patients enrolled and followed by only one center. Additionally, lipid levels showed a significant reduction, but it was not possible to retrieve data on administration of statins and dietary supplements to lower lipid levels, thus is not possible to determine the effects of these molecules on lipid levels. Due to the real-world nature of the cohort, it was not possible to obtain data regarding adherence to treatment, which may have been useful to determine the impact of therapeutic adherence on outcomes.

In real life, switching to B/F/TAF was safe and highly effective both virologically and immunologically. The present study reported a decrease in cholesterol and triglyceride levels, showing a favorable metabolic profile, which may reduce inflammation, leading to a healthier state and less organ damage.

Acknowledgements

Not applicable.

Funding

Funding: No funding was received.

Availability of data and materials

The data generated in the present study may be requested from the corresponding author.

Authors' contributions

BC, SS, GNC and AM conceived the study. VF, GF, VR, BMC and GFP designed the study. MVP, GS and RB analyzed data. EVN, EVR, GN, NV, AM, VC, MSPR, EP and VB performed experiments. EVR and GN wrote the manuscript. VF and GF reviewed the manuscript. BC, EVN and GN supervised the study. All authors have read and approved the final manuscript. SS and GN confirm the authenticity of all the raw data.

Ethics approval and consent to participate

All participants provided written informed consent to partake in the study. The study was conducted in accordance with the Declaration of Helsinki and approved by the Provincial Ethics Committee of Messina (approval no. 34/17 of the 22/03/2017).

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Trickey A, Sabin CA, Burkholder G, Crane H, d'Arminio Monforte A, Egger M, Gill MJ, Grabar S, Guest JL, Jarrin I, et al: Life expectancy after 2015 of adults with HIV on long-term antiretroviral therapy in Europe and North America: A collaborative analysis of cohort studies. Lancet HIV. 10:e295–e307. 2023.PubMed/NCBI View Article : Google Scholar

2 

Marino A, Zafarana G, Ceccarelli M, Cosentino F, Moscatt V, Bruno G, Bruno R, Benanti F, Cacopardo B and Celesia BM: Immunological and clinical impact of DAA-Mediated HCV Eradication in a Cohort of HIV/HCV Coinfected Patients: Monocentric Italian Experience. Diagnostics (Basel). 11(2336)2021.PubMed/NCBI View Article : Google Scholar

3 

D'Andrea F, Venanzi Rullo E, Marino A, Moscatt V, Celesia BM, Cacopardo B, Condorelli F, La Rocca G, Di Rosa M, Pellicanò GF, et al: Hepatitis B virus infection and hepatocellular carcinoma in PLWH: Epidemiology, pathogenesis and treatment. World Cancer Res J. 7(e1537)2020.

4 

Peters E and Iwuji AC: Efficacy, safety and tolerability of Biktarvy in HIV-1 infection: A scoping review. Antivir Ther. 28(13596535231159030)2023.PubMed/NCBI View Article : Google Scholar

5 

Celesia BM, Marino A, del Vecchio RF, Bruno R, Palermo F, Gussio M, Nunnari G and Cacopardo B: Is it Safe and Cost Saving to Defer the CD4+ cell count monitoring in stable patients on art with more than 350 or 500 cells/µl? Mediterr J Hematol Infect Dis. 11(e2019063)2019.PubMed/NCBI View Article : Google Scholar

6 

Celesia BM, Marino A, Borracino S, Arcadipane AF, Pantò G, Gussio M, Coniglio S, Pennisi A, Cacopardo B and Panarello G: Successful extracorporeal membrane oxygenation treatment in an acquired immune deficiency syndrome (AIDS) patient with acute respiratory distress syndrome (ARDS) Complicating Pneumocystis jirovecii Pneumonia: A Challenging Case. Am J Case Rep. 21(e919570)2020.PubMed/NCBI View Article : Google Scholar

7 

Micali C, Russotto Y, Facciolà A, Marino A, Celesia BM, Pistarà E, Caci G, Nunnari G, Pellicanò GF and Venanzi Rullo E: Pulmonary kaposi sarcoma without respiratory symptoms and skin lesions in an HIV-Naïve Patient: A case report and literature review. Infect Dis Rep. 14:228–242. 2022.PubMed/NCBI View Article : Google Scholar

8 

Ambrosioni J, Levi L, Alagaratnam J, Van Bremen K, Mastrangelo A, Waalewijn H, Molina JM, Guaraldi G, Winston A, Boesecke C, et al: Major revision version 12.0 of the European AIDS Clinical Society guidelines 2023. HIV Med. 24:1126–1136. 2023.PubMed/NCBI View Article : Google Scholar

9 

Marino A, Cosentino F, Ceccarelli M, Moscatt V, Pampaloni A, Scuderi D, D'Andrea F, Rullo EV, Nunnari G, Benanti F, et al: Entecavir resistance in a patient with treatment-naïve HBV: A case report. Mol Clin Oncol. 14(113)2021.PubMed/NCBI View Article : Google Scholar

10 

Marino A, Pavone G, Martorana F, Fisicaro V, Motta L, Spampinato S, Celesia BM, Cacopardo B, Vigneri P and Nunnari G: Navigating the Nexus: HIV and Breast Cancer-A Critical Review. Int J Mol Sci. 25(3222)2024.PubMed/NCBI View Article : Google Scholar

11 

Ron R, Martínez-Sanz J, Herrera S, Ramos-Ruperto L, Díez A, Sainz T, Álvarez-Díaz N, Correa-Pérez A, Muriel A, López-Alcalde J, et al: CD4/CD8 ratio and CD8+ T-cell count as prognostic markers for non-AIDS mortality in people living with HIV. A systematic review and meta-analysis. Front Immunol. 15(1343124)2024.PubMed/NCBI View Article : Google Scholar

12 

Serrano-Villar S, Wu K, Hunt PW, Lok JJ, Ron R, Sainz T, Moreno S, Deeks SG and Bosch RJ: Predictive value of CD8+ T cell and CD4/CD8 ratio at two years of successful ART in the risk of AIDS and non-AIDS events. EBioMedicine. 80(104072)2022.PubMed/NCBI View Article : Google Scholar

13 

World Health Organization (WHO): Consolidated guidelines on HIV prevention, testing, treatment, service delivery and monitoring: Recommendations for a public health approach. WHO, Geneva, 2021.

14 

Baeten JM, Donnell D, Ndase P, Mugo NR, Campbell JD, Wangisi J, Tappero JW, Bukusi EA, Cohen CR, Katabira E, et al: Antiretroviral prophylaxis for HIV prevention in heterosexual men and women. N Engl J Med. 367:399–410. 2012.PubMed/NCBI View Article : Google Scholar

15 

Baeten JM, Kahle E, Lingappa JR, Coombs RW, Delany-Moretlwe S, Nakku-Joloba E, Mugo NR, Wald A, Corey L, Donnell D, et al: Genital HIV-1 RNA predicts risk of heterosexual HIV-1 transmission. Sci Transl Med. 3(77ra29)2011.PubMed/NCBI View Article : Google Scholar

16 

Acosta RK, Willkom M, Andreatta K, Liu H, Martin R, Parvangada A, Martin H, Collins S and White KL: Switching to Bictegravir/Emtricitabine/Tenofovir Alafenamide (B/F/TAF) From Dolutegravir (DTG)+F/TAF or DTG+F/Tenofovir Disoproxil Fumarate (TDF) in the Presence of Pre-existing NRTI Resistance. J Acquir Immune Defic Syndr. 85:363–371. 2020.PubMed/NCBI View Article : Google Scholar

17 

Chang HM, Chou PY, Chou CH and Tsai HC: Outcomes After Switching to BIC/FTC/TAF in patients with virological failure to protease inhibitors or non-nucleoside reverse transcriptase inhibitors: A real-world cohort study. Infect Drug Resist. 14:4877–4886. 2021.PubMed/NCBI View Article : Google Scholar

18 

Chen LY, Sun HY, Chuang YC, Huang YS, Liu WD, Lin KY, Chang HY, Luo YZ, Wu PY, Su YC, et al: Patient-reported outcomes among virally suppressed people living with HIV after switching to Co-formulated bictegravir, emtricitabine and tenofovir alafenamide. J Microbiol Immunol Infect. 56:575–585. 2023.PubMed/NCBI View Article : Google Scholar

19 

Daar ES, DeJesus E, Ruane P, Crofoot G, Oguchi G, Creticos C, Rockstroh JK, Molina JM, Koenig E, Liu YP, et al: Efficacy and safety of switching to fixed-dose bictegravir, emtricitabine, and tenofovir alafenamide from boosted protease inhibitor-based regimens in virologically suppressed adults with HIV-1: 48 week results of a randomised, open-label, multicentre, phase 3, non-inferiority trial. Lancet HIV. 5:e347–e356. 2018.PubMed/NCBI View Article : Google Scholar

20 

De Socio GV, Tordi S, Altobelli D, Gidari A, Zoffoli A and Francisci D: Dolutegravir/Lamivudine versus Tenofovir Alafenamide/Emtricitabine/Bictegravir as a Switch Strategy in a Real-Life Cohort of Virogically Suppressed People Living with HIV. J Clin Med. 12(7759)2023.PubMed/NCBI View Article : Google Scholar

21 

Hagins D, Kumar P, Saag M, Wurapa AK, Brar I, Berger D, Osiyemi O, Hileman CO, Ramgopal MN, McDonald C, et al: Switching to Bictegravir/Emtricitabine/Tenofovir Alafenamide in Black Americans With HIV-1: A Randomized Phase 3b, Multicenter, Open-Label Study. J Acquir Immune Defic Syndr. 88:86–95. 2021.PubMed/NCBI View Article : Google Scholar

22 

Kityo C, Hagins D, Koenig E, Avihingsanon A, Chetchotisakd P, Supparatpinyo K, Gankina N, Pokrovsky V, Voronin E, Stephens JL, et al: Switching to Fixed-Dose Bictegravir, Emtricitabine, and Tenofovir Alafenamide (B/F/TAF) in Virologically Suppressed HIV-1 Infected Women: A Randomized, Open-Label, Multicenter, Active-Controlled, Phase 3, Noninferiority Trial. J Acquir Immune Defic Syndr. 82:321–328. 2019.PubMed/NCBI View Article : Google Scholar

23 

Knobel H, Cañas-Ruano E, Guelar A, Knobel P, Villar-García J, González-Mena A, Canepa C, Arrieta-Aldea I, Marcos A, Abalat-Torrres A and Güerri-Fernández R: Switching to Dolutegravir/lamivudine or Bictegravir/Emtricitabine/Tenofovir alafenamide. A comparative real-world study. HIV Res Clin Pract. 24(2239564)2023.PubMed/NCBI

24 

Lake JE, Hyatt AN, Feng H, Debroy P, Kettelhut A, Miao H, Peng L, Bhasin S, Bell S, Rianon N, et al: A randomized clinical trial of transgender women switching to B/F/TAF: The (mo)BETTA Trial. Open Forum Infect Dis. 10(ofad178)2023.PubMed/NCBI View Article : Google Scholar

25 

Lazzaro A, Cacciola EG, Borrazzo C, Innocenti GP, Cavallari EN, Mezzaroma I, Falciano M, Fimiani C, Mastroianni CM, Ceccarelli G and d'Ettorre G: Switching to a Bictegravir Single Tablet Regimen in Elderly People Living with HIV-1: Data Analysis from the BICTEL Cohort. Diagnostics (Basel). 12(76)2021.PubMed/NCBI View Article : Google Scholar

26 

Lazzaro A, Bianchini D, Gentilini Cacciola E, Mezzaroma I, Falciano M, Andreoni C, Fimiani C, Santinelli L, Maddaloni L, Bugani G, et al: Immune Reconstitution and Safe Metabolic Profile after the Switch to Bictegravir/Emtricitabine/Tenofovir Alafenamide Fumarate among Virologically Controlled PLWH: A 96 Week Update from the BICTEL Cohort. Viruses. 15(1222)2023.PubMed/NCBI View Article : Google Scholar

27 

Maggiolo F, Rizzardini G, Molina JM, Pulido F, De Wit S, Vandekerckhove L, Berenguer J, D'Antoni ML, Blair C, Chuck SK, et al: Bictegravir/Emtricitabine/Tenofovir Alafenamide in Virologically Suppressed People with HIV Aged ≥ 65 Years: Week 48 Results of a Phase 3b, Open-Label Trial. Infect Dis Ther. 10:775–788. 2021.PubMed/NCBI View Article : Google Scholar

28 

Molina JM, Ward D, Brar I, Mills A, Stellbrink HJ, López-Cortés L, Ruane P, Podzamczer D, Brinson C, Custodio J, et al: Switching to fixed-dose bictegravir, emtricitabine, and tenofovir alafenamide from dolutegravir plus abacavir and lamivudine in virologically suppressed adults with HIV-1: 48 week results of a randomised, double-blind, multicentre, active-controlled, phase 3, non-inferiority trial. Lancet HIV. 5:e357–e365. 2018.PubMed/NCBI View Article : Google Scholar

29 

Tsai MS, Sun HY, Chen CP, Lee CH, Lee CY, Liu CE, Tang HJ, Hung TC, Li CW, Lee YT, et al: Switching to coformulated bictegravir, emtricitabine, and tenofovir alafenamide maintained viral suppression in adults with historical virological failures and K65N/R mutation. Int J Infect Dis. 126:39–47. 2023.PubMed/NCBI View Article : Google Scholar

30 

Rolle CP, Nguyen V, Patel K, Cruz D, DeJesus E and Hinestrosa F: Real-world efficacy and safety of switching to bictegravir/emtricitabine/tenofovir alafenamide in older people living with HIV. Medicine (Baltimore). 100(e27330)2021.PubMed/NCBI View Article : Google Scholar

31 

Sax PE, Rockstroh JK, Luetkemeyer AF, Yazdanpanah Y, Ward D, Trottier B, Rieger A, Liu H, Acosta R, Collins SE, et al: Switching to bictegravir, emtricitabine, and tenofovir alafenamide in virologically suppressed adults with human immunodeficiency virus. Clin Infect Dis. 73:e485–e493. 2021.PubMed/NCBI View Article : Google Scholar

32 

Klatzmann D, Barré-Sinoussi F, Nugeyre MT, Danquet C, Vilmer E, Griscelli C, Brun-Veziret F, Rouzioux C, Gluckman JC, Chermann JC, et al: Selective tropism of lymphadenopathy associated virus (LAV) for helper-inducer T lymphocytes. Science. 225:59–63. 1984.PubMed/NCBI View Article : Google Scholar

33 

Doitsh G and Greene WC: Dissecting How CD4 T Cells Are Lost During HIV Infection. Cell Host Microbe. 19:280–291. 2016.PubMed/NCBI View Article : Google Scholar

34 

Bishop JD, DeShields S, Cunningham T and Troy SB: CD4 count recovery after initiation of antiretroviral therapy in patients infected with human immunodeficiency virus. Am J Med Sci. 352:239–244. 2016.PubMed/NCBI View Article : Google Scholar

35 

Ho DD, Neumann AU, Perelson AS, Chen W, Leonard JM and Markowitz M: Rapid turnover of plasma virions and CD4 lymphocytes in HIV-1 infection. Nature. 373:123–126. 1995.PubMed/NCBI View Article : Google Scholar

36 

Roederer M, Dubs JG, Anderson MT, Raju PA and Herzenberg LA and Herzenberg LA: CD8 naive T cell counts decrease progressively in HIV-infected adults. J Clin Invest. 95:2061–2066. 1995.PubMed/NCBI View Article : Google Scholar

37 

Favre D, Stoddart CA, Emu B, Hoh R, Martin JN, Hecht FM, Deeks SG and McCune JM: HIV disease progression correlates with the generation of dysfunctional naive CD8(low) T cells. Blood. 117:2189–2199. 2011.PubMed/NCBI View Article : Google Scholar

38 

Oxenius A, Price DA, Easterbrook PJ, O'Callaghan CA, Kelleher AD, Whelan JA, Sontag G, Sewell AK and Phillips RE: Early highly active antiretroviral therapy for acute HIV-1 infection preserves immune function of CD8+ and CD4+ T lymphocytes. Proc Natl Acad Sci USA. 97:3382–3387. 2000.PubMed/NCBI View Article : Google Scholar

39 

Badejo OA, Chang CC, So-Armah KA, Tracy RP, Baker JV, Rimland D, Butt AA, Gordon AJ, Rinaldo CR Jr, Kraemer K, et al: CD8+ T-Cells Count in Acute Myocardial Infarction in HIV disease in a predominantly male cohort. Biomed Res Int. 2015(246870)2015.PubMed/NCBI View Article : Google Scholar

40 

Schneider S, Spinner CD, Cassese S, Promny D, Hapfelmeier A, Byrne RA, Baumann M, Jäger H, Steinlechner E, Laugwitz KL and Kastrati A: Association of increased CD8+ and persisting C-reactive protein levels with restenosis in HIV patients after coronary stenting. AIDS. 30:1413–1421. 2016.PubMed/NCBI View Article : Google Scholar

41 

Borges ÁH, Neuhaus J, Babiker AG, Henry K, Jain MK, Palfreeman A, Mugyenyi P, Domingo P, Hoffmann C, Read TR, et al: Immediate antiretroviral therapy reduces risk of infection-related cancer during early HIV Infection. Clin Infect Dis. 63:1668–1676. 2016.PubMed/NCBI View Article : Google Scholar

42 

Helleberg M, May MT, Ingle SM, Dabis F, Reiss P, Fätkenheuer G, Costagliola D, d'Arminio A, Cavassini M, Smith C, et al: Smoking and life expectancy among HIV-infected individuals on antiretroviral therapy in Europe and North America. AIDS. 29:221–229. 2015.PubMed/NCBI View Article : Google Scholar

43 

Castilho JL, Shepherd BE, Koethe J, Turner M, Bebawy S, Logan J, Rogers WB, Raffanti S and Sterling TR: CD4+/CD8+ ratio, age, and risk of serious non-communicable diseases in HIV-infected adults on antiretroviral therapy. AIDS. 30:899–908. 2016.PubMed/NCBI View Article : Google Scholar

44 

Dock JN and Effros RB: Role of CD8 T cell replicative senescence in human aging and in HIV-mediated Immunosenescence. Aging Dis. 2:382–397. 2011.PubMed/NCBI

45 

Avelino-Silva VI, Miyaji KT, Hunt PW, Huang Y, Simoes M, Lima SB, Freire MS, Caiaffa-Filho HH, Hong MA, Costa DA, et al: CD4/CD8 Ratio and KT Ratio Predict Yellow Fever Vaccine Immunogenicity in HIV-Infected Patients. PLoS Negl Trop Dis. 10(e0005219)2016.PubMed/NCBI View Article : Google Scholar

46 

Collin A, Le Marec F, Vandenhende MA, Lazaro E, Duffau P, Cazanave C, Gérard Y, Dabis F, Bruyand M and Bonnet F: ANRS CO3 Aquitaine Cohort Study Group. Incidence and risk factors for severe bacterial infections in people living with HIV. ANRS CO3 Aquitaine Cohort, 2000-2012. PLoS One. 11(e0152970)2016.PubMed/NCBI View Article : Google Scholar

47 

Vrinceanu D, Dumitru M, Patrascu OM, Costache A, Papacocea T and Cergan R: Current diagnosis and treatment of rhinosinusal aspergilloma (Review). Exp Ther Med. 22(1264)2021.PubMed/NCBI View Article : Google Scholar

48 

Saydam FN, Erdem H, Ankarali H, El-Arab Ramadan ME, El-Sayed NM, Civljak R, Pshenichnaya N, Moroti RV, Mahmuodabad FM, Maduka AV, et al: Vector-borne and zoonotic infections and their relationships with regional and socioeconomic statuses: An ID-IRI survey in 24 countries of Europe, Africa and Asia. Travel Med Infect Dis. 44(102174)2021.PubMed/NCBI View Article : Google Scholar

49 

Sigel K, Wisnivesky J, Crothers K, Gordon K, Brown ST, Rimland D, Rodriguez-Barradas MC, Gibert C, Goetz MB, Bedimo R, et al: Immunological and infectious risk factors for lung cancer in US veterans with HIV: A longitudinal cohort study. Lancet HIV. 4:e67–e73. 2017.PubMed/NCBI View Article : Google Scholar

50 

Mujawar Z, Rose H, Morrow MP, Pushkarsky T, Dubrovsky L, Mukhamedova N, Fu Y, Dart A, Orenstein JM, Bobryshev YV, et al: Human immunodeficiency virus impairs reverse cholesterol transport from macrophages. PLoS Biol. 4(e365)2006.PubMed/NCBI View Article : Google Scholar

51 

Lu L, Yang Y, Yang Z, Wu Y, Liu X, Li X, Chen L, Han Y, Song X, Kong Z, et al: Altered plasma metabolites and inflammatory networks in HIV-1 infected patients with different immunological responses after long-term antiretroviral therapy. Front Immunol. 14(1254155)2023.PubMed/NCBI View Article : Google Scholar

52 

Papantoniou E, Arvanitakis K, Markakis K, Papadakos SP, Tsachouridou O, Popovic DS, Germanidis G, Koufakis T and Kotsa K: Pathophysiology and clinical management of dyslipidemia in people living with HIV: Sailing through Rough Seas. Life (Basel). 14(449)2024.PubMed/NCBI View Article : Google Scholar

53 

Stanley TL and Grinspoon SK: Body composition and metabolic changes in HIV-Infected Patients. J Infect Dis. 205 (Suppl 3):S383–S390. 2012.PubMed/NCBI View Article : Google Scholar

54 

Avihingsanon A, Chetchotisakd P, Kiertiburanakul S, Ratanasuwan W, Siripassorn K, Supparatpinyo K, Martin H, Wang H, Wong T and Wang HY: Efficacy and safety of switching to bictegravir, emtricitabine, and tenofovir alafenamide in virologically suppressed Asian adults living with HIV: A pooled analysis from three international phase III randomized trials. HIV Med. 24:290–300. 2023.PubMed/NCBI View Article : Google Scholar

55 

Maggiolo F, Rizzardini G, Molina JM, Pulido F, De Wit S, Vandekerckhove L, Berenguer J, D'Antoni ML, Blair C, Chuck SK, et al: Bictegravir/emtricitabine/tenofovir alafenamide in older individuals with HIV: Results of a 96-week, phase 3b, open-label, switch trial in virologically suppressed people ≥65 years of age. HIV Med. 24:27–36. 2023.PubMed/NCBI View Article : Google Scholar

56 

Squillace N, Ricci E, Maggi P, Taramasso L, Menzaghi B, De Socio GV, Piconi S, Maurizio Celesia B, Orofino G, Sarchi E, et al: Real-life safety of Emtricitabine/Tenofovir Alafenamide/Bictegravir. PLoS One. 18(e0289132)2023.PubMed/NCBI View Article : Google Scholar

57 

Perrone RD, Madias NE and Levey AS: Serum creatinine as an index of renal function: new insights into old concepts. Clin Chem. 38:1933–1953. 1992.PubMed/NCBI

58 

Kashani K, Rosner MH and Ostermann M: Creatinine: From physiology to clinical application. Eur J Intern Med. 72:9–14. 2020.PubMed/NCBI View Article : Google Scholar

59 

Ando M and Yanagisawa N: Epidemiology, clinical characteristics, and management of chronic kidney disease in human immunodeficiency virus-infected patients. World J Nephrol. 4:388–395. 2015.PubMed/NCBI View Article : Google Scholar

60 

Ryom L, Mocroft A, Kirk O, Worm SW, Kamara DA, Reiss P, Ross M, Fux CA, Morlat P, Moranne O, et al: Association between antiretroviral exposure and renal impairment among HIV-positive persons with normal baseline renal function: The D:A:D study. J Infect Dis. 207:1359–1369. 2013.PubMed/NCBI View Article : Google Scholar

61 

Pottel H, Vrydags N, Mahieu B, Vandewynckele E, Croes K and Martens F: Establishing age/sex related serum creatinine reference intervals from hospital laboratory data based on different statistical methods. Clin Chim Acta. 396:49–55. 2008.PubMed/NCBI View Article : Google Scholar

62 

Yamada J, Tomiyama H, Yambe M, Koji Y, Motobe K, Shiina K, Yamamoto Y and Yamashina A: Elevated serum levels of alanine aminotransferase and gamma glutamyltransferase are markers of inflammation and oxidative stress independent of the metabolic syndrome. Atherosclerosis. 189:198–205. 2006.PubMed/NCBI View Article : Google Scholar

63 

Nixon DE and Landay AL: Biomarkers of immune dysfunction in HIV. Curr Opin HIV AIDS. 5:498–503. 2010.PubMed/NCBI View Article : Google Scholar

64 

Siest G, Schiele F, Galteau MM, Panek E, Steinmetz J, Fagnani F and Gueguen R: Aspartate aminotransferase and alanine aminotransferase activities in plasma: statistical distributions, individual variations, and reference values. Clin Chem. 21:1077–1087. 1975.PubMed/NCBI

65 

Sookoian S and Pirola CJ: Liver enzymes, metabolomics and genome-wide association studies: From systems biology to the personalized medicine. World J Gastroenterol. 21:711–725. 2015.PubMed/NCBI View Article : Google Scholar

Related Articles

Journal Cover

December-2024
Volume 21 Issue 6

Print ISSN: 2049-9434
Online ISSN:2049-9442

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Spampinato S, Conti GN, Marino A, Raimondo V, Celesia BM, Pellicanò GF, Puci MV, Sotgiu G, Bruno R, Villari N, Villari N, et al: Enhanced metabolic health and immune response with bictegravir/emtricitabine/TAF: Insights from a 96‑week retrospective study. Biomed Rep 21: 179, 2024.
APA
Spampinato, S., Conti, G.N., Marino, A., Raimondo, V., Celesia, B.M., Pellicanò, G.F. ... Nunnari, G. (2024). Enhanced metabolic health and immune response with bictegravir/emtricitabine/TAF: Insights from a 96‑week retrospective study. Biomedical Reports, 21, 179. https://doi.org/10.3892/br.2024.1867
MLA
Spampinato, S., Conti, G. N., Marino, A., Raimondo, V., Celesia, B. M., Pellicanò, G. F., Puci, M. V., Sotgiu, G., Bruno, R., Villari, N., Mirabile, A., Coco, V. A., Paternò Raddusa, M. S., Pistarà, E., Boscia, V., Fisicaro, V., Fiorenza, G., Cacopardo, B., Rullo, E. V., Nunnari, G."Enhanced metabolic health and immune response with bictegravir/emtricitabine/TAF: Insights from a 96‑week retrospective study". Biomedical Reports 21.6 (2024): 179.
Chicago
Spampinato, S., Conti, G. N., Marino, A., Raimondo, V., Celesia, B. M., Pellicanò, G. F., Puci, M. V., Sotgiu, G., Bruno, R., Villari, N., Mirabile, A., Coco, V. A., Paternò Raddusa, M. S., Pistarà, E., Boscia, V., Fisicaro, V., Fiorenza, G., Cacopardo, B., Rullo, E. V., Nunnari, G."Enhanced metabolic health and immune response with bictegravir/emtricitabine/TAF: Insights from a 96‑week retrospective study". Biomedical Reports 21, no. 6 (2024): 179. https://doi.org/10.3892/br.2024.1867