Next Article in Journal
Enhancing the Functional Properties of Tea Tree Oil: In Vitro Antimicrobial Activity and Microencapsulation Strategy
Next Article in Special Issue
Update and Application of a Deep Learning Model for the Prediction of Interactions between Drugs Used by Patients with Multiple Sclerosis
Previous Article in Journal
Enhancing Dental Applications: A Novel Approach for Incorporating Bioactive Substances into Textile Threads
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Systematic Review

Clinical Relevance of Drug Interactions in People Living with Human Immunodeficiency Virus on Antiretroviral Therapy—Update 2022: Systematic Review

by
Pedro Amariles
1,2,*,
Mónica Rivera-Cadavid
1 and
Mauricio Ceballos
1,3
1
Research Group on Pharmaceutical Promotion and Prevention, University of Antioquia, UdeA, AA 1226, Medellin 050010, Colombia
2
Research Group on Pharmaceutical Care, University of Granada, 18071 Granada, Spain
3
Research Group on Pharmacy Regency Technology, University of Antioquia, Medellin 050010, Colombia
*
Author to whom correspondence should be addressed.
Pharmaceutics 2023, 15(10), 2488; https://doi.org/10.3390/pharmaceutics15102488
Submission received: 7 June 2023 / Revised: 25 September 2023 / Accepted: 29 September 2023 / Published: 18 October 2023
(This article belongs to the Special Issue Drug–Drug Interactions—New Approaches and Perspectives)

Abstract

:
Background: The clinical outcomes of antiretroviral drugs may be modified through drug interactions; thus, it is important to update the drug interactions in people living with HIV (PLHIV). Aim: To update clinically relevant drug interactions in PLHIV on antiretroviral therapy with novel drug interactions published from 2017 to 2022. Methods: A systematic review in Medline/PubMed database from July 2017 to December 2022 using the Mesh terms antiretroviral agents and drug interactions or herb–drug interactions or food–drug interactions. Publications with drug interactions in humans, in English or Spanish, and with full-text access were retrieved. The clinical relevance of drug interactions was grouped into five levels according to the gravity and probability of occurrence. Results: A total of 366 articles were identified, with 219 (including 87 citation lists) were included, which allowed for the identification of 471 drug interaction pairs; among them, 291 were systematically reported for the first time. In total 42 (14.4%) and 137 (47.1%) were level one and two, respectively, and 233 (80.1%) pairs were explained with the pharmacokinetic mechanism. Among these 291 pairs, protease inhibitors (PIs) and ritonavir/cobicistat-boosted PIs, as well as integrase strand transfer inhibitors (InSTIs), with 70 (24.1%) and 65 (22.3%) drug interaction pairs of levels one and two, respectively, were more frequent. Conclusions: In PLHIV on antiretroviral therapy, we identify 291 drug interaction pairs systematically reported for the first time, with 179 (61.5%) being assessed as clinically relevant (levels one and two). The pharmacokinetic mechanism was the most frequently identified. PIs, ritonavir/cobicistat-boosted PIs, and InSTIs were the antiretroviral groups with the highest number of clinically relevant drug interaction pairs (levels one and two).

1. Introduction

Human immunodeficiency virus (HIV) is one of the main public health problems. According to the World Health Organization (WHO), globally, 39.0 million (33.1–45.7 million) people were living with HIV at the end of 2022; additionally, during 2022, 630,000 (480,000–880,000) people died from HIV-related causes and 1.3 million (1.0–1.7 million) persons acquired HIV [1]. In recent years, remarkable advances have been achieved in the treatment of HIV; thus, currently, most people living with HIV (PLHIV) have a life expectancy similar to persons without HIV. According to the latest updated guidelines, it is recommended to start antiretroviral (ARV) therapy as soon as possible after HIV diagnosis, ideally within 7 days. Additionally, if they have an opportunistic infection, ARV therapy should be started shortly after the initiation of the treatment for the infection, being recommended within 2 weeks [2].
Currently, in PLHIV, initial ARV therapy generally consists of two nucleoside/nucleotide reverse transcriptase inhibitors (NRTIs) combined with a third active ARV drug, which may be an integrase strand transfer inhibitor (InSTI), a non-nucleoside reverse transcriptase inhibitor (NNRTI), or a protease inhibitor (PI) boosted with cobicistat (COBI) or ritonavir (RTV). InSTIs such as bictegravir (BIC) or dolutegravir (DTG) are the preferred third ARV drug, mainly due to being associated with a lower risk for drug resistance and for drug–drug interactions [2]. Additionally, the two-drug regimen, DTG plus lamivudine (3TC), may be recommended for the initial option for patients with an initial HIV viral load of <500,000 copies/mL; for patients who have achieved viral suppression, a long-acting injectable regimen of bimonthly injections of long-acting cabotegravir (CAB) and rilpivirine (RPV) may be used. Additionally, advances in ARV therapy have led to the availability of well-tolerated single-tablet regimens that are associated with a lower risk of drug interactions, as well as options for pre-exposure prophylaxis, including daily oral medications, such as tenofovir (TDF)/emtricitabine (FTC), or bimonthly injectable CAB [2].
A drug interaction is an undesirable modification that is quantifiable in the magnitude or duration of effects related to the simultaneous or previous administration of other drugs, phytotherapeutics, foods, or due to pathophysiological (special) conditions of the patient [3]. The identification, prevention, and resolution of clinically relevant drug interactions are a critical aspect of achieving pharmacotherapy goals. Among other methods for evaluating the clinical relevance of interactions, a proposal based on the gravity of the effect on the patient’s health (grave, moderate, and minor) and the probability of occurrence (defined, probable, and possible, according to the type of study supporting the drug interaction) has been considered as appropriate. This proposed classification generates four levels of clinical relevance: level one (very high risk), level two (high risk), level three (medium risk), and level four (low risk) [3,4]. In addition, a new level of clinical relevance (level five: lowest risk) has been proposed, which is characterized by the absence of an effect on the patient’s health (lack of gravity) documented in meta-analyses, systematic reviews, or clinical trials (defined probability), and, therefore, with evidence of the absence of clinically relevant drug interactions [4].
Regarding clinically relevant drug interactions in persons with HIV, from 1995 to 2017, we identified four previously published reviews, which focused on identifying drug interactions between ARV drugs, phytotherapeutics, and foods [5,6,7,8]. The most recent review updated the reported ARV interactions up to June 2017 [8]. However, due to the commercialization of new ARVs, updates to guidelines and expert recommendations and, mainly, both the identification and reporting of new clinically relevant drug interactions or the generation of new knowledge about drug interactions systematically reported previously, this information should be periodically updated. In addition, in 2017, a free software to facilitate the identification and assessment of the clinical relevance of ARV drug interactions (SIMARV®) was developed [9]; then, a free mobile version (InterApp-ARV) was developed and is available for mobile phones and tablets [10]. The development of both SIMARV® and InterApp ARV, used as a graphic reference the free software developed by the University of Liverpool (https://www.hiv-druginteractions.org/ accessed on 25 July 2023), is considered as the most used online source of DDI in HIV [11]. In this context, this systematic review aimed to update clinically relevant drug interactions in PLHIV on antiretroviral therapy, with novel drug interaction pairs between ARVs and other medications, phytotherapeutics, or foods published from 2017 to 2022.

2. Materials and Methods

Similar to previously published reviews, a systematic review was conducted in the Medline/PubMed database from 1 July 2017 to 31 December 2022 using the Mesh terms antiretroviral agents AND drug interactions OR herb–drug interactions OR food–drug interactions. Articles published in English or Spanish and with full-text access were identified.
Inclusion criteria: We included all articles containing clinically relevant information on drug interactions in humans using antiretroviral agents for the treatment of persons living with HIV/AIDS. Additionally, other studies were identified from the reference list of retrieved articles.
Exclusion criteria: We excluded the following types of articles: (a) preclinical or in vitro studies; (b) with theoretical concepts regarding drug interactions; (c) without specific ARV drug interactions; (d) not related to HIV; and (e) without full-text availability.
To ensure a systematic approach, three researchers reviewed the studies identified according to the preferred reporting items for systematic reviews and meta-analysis (PRISMA) flow chart via a predetermined eligibility criteria [12]. The titles and abstracts of all identified articles were screened for eligibility by the three authors, and any discrepancies were resolved by consensus. Subsequently, to allow for the synthesis and analysis of the results, the data were collected in a table with the following information: (a) article title, ARV assessed, (b) drug-related interaction, (c) clinical relevance level (according to the combination of the gravity and probability of occurrence), (d) pharmacodynamic or pharmacokinetic mechanism, (e) comment and recommendation, and (f) reference. The information registered was proofread by the three authors.
The drug interaction pairs of identified ARV agent–drug interactions were classified into five levels according to the gravity (effect on patient’s health) and probability of occurrence (type of study that supports the interaction), following the combination of options, as shown in Table 1 [3,4].
The probability was determined and classified according to the kind of study that supported the interaction found for each pair of drug interactions [3]:
  • Possible: The drug interaction pair was documented with results from less than three case reports or by expert consensus.
  • Probable: The drug interaction pair was documented with results from at least one observational study (cohort or case–control study) or at least three case reports.
  • Defined: The drug interaction pair was documented with results from at least one meta-analysis, systematic review, or randomized or nonrandomized clinical trial.
In the cases of the reviews, including systematic reviews or meta-analyses, the reference list was reviewed and the drug interaction had to be support with a clinical study. In addition, if the study (case reports, observational study, clinical trial, systematic review, or meta-analysis) was identified for the first time, it was included. Therefore, in the current update, the drug interaction pair probability, due to bringing together all the references that supported it, could be (a) systematically identified for the first time (the references identified for the first time generated the probability), (b) increased (the references identified for the first time modified the probability), or conserved (the references systematically identified for the first time reinforced it but did not modify it).
The gravity attributed to the drug interaction was determined and classified according to the effect on the patient’s health [3,4]:
  • Lack of gravity: There was evidence that the drug interaction did not cause harm to the patient.
  • Minor: The drug interaction did not cause or caused minimum harm to the patient (including those that did not require an additional drug treatment nor generated qualitative or quantitative pharmacotherapy changes, neither increasing the patient’s hospitalization), but generated the need for monitoring the patient’s health.
  • Moderate: The drug interaction generated the need for a closer monitoring of the patient’s health (including those that required an additional drug treatment, generated qualitative or quantitative pharmacotherapy changes, or increased the patient’s hospitalization.
  • Grave: The drug interaction could cause harm or injury to the patient (including those that could be life threatening, result in persistent or significant disability or hospitalization, or cause birth defects).

3. Results

From the search in the PubMed/Medline database, 366 records were retrieved; among them, 5 were removed before screening. Then, 110 records were excluded due to the screened title and abstract. Thus, 251 articles were assessed for eligibility; among them, 119 were excluded and, consequently, a total of 132 articles were included in the review. In addition, from the citation list, 87 articles were included; thus, 219 articles were used for this review (Figure 1). However, in the current article, only drug interactions assessed as levels one, two, and five were presented, which were supported by 194 [13,14,15,16,17,18,19,20,21,22,23,24,25,26,27,28,29,30,31,32,33,34,35,36,37,38,39,40,41,42,43,44,45,46,47,48,49,50,51,52,53,54,55,56,57,58,59,60,61,62,63,64,65,66,67,68,69,70,71,72,73,74,75,76,77,78,79,80,81,82,83,84,85,86,87,88,89,90,91,92,93,94,95,96,97,98,99,100,101,102,103,104,105,106,107,108,109,110,111,112,113,114,115,116,117,118,119,120,121,122,123,124,125,126,127,128,129,130,131,132,133,134,135,136,137,138,139,140,141,142,143,144,145,146,147,148,149,150,151,152,153,154,155,156,157,158,159,160,161,162,163,164,165,166,167,168,169,170,171,172,173,174,175,176,177,178,179,180,181,182,183,184,185,186,187,188,189,190,191,192,193,194,195,196,197,198,199,200,201,202,203,204,205,206] of those 219 articles.
A total of 471 drug interaction pairs between antiretroviral agents and other drugs were identified; of them, 291 were interactions systematically reported for the first time, 125 were updates to drug interactions reported previously, and 55 were related to drugs not yet approved or to discontinued drugs in clinal practice (Figure 1). The clinical relevance levels (based on gravity and probability) and the mechanism for the 291 pairs of drug interactions systematically reported for the first time are shown in Table 2.
Among the 291 interactions systematically reported for the first time, 179 (61.5%) were assessed as having a higher risk of causing adverse drug outcomes related to the ineffectiveness or unsafe use of the pharmacotherapy, of which 42 (14.4%) and 137 (47.1%) were assessed as level one and level two, respectively (Table 2). Table 3 and Table 4 provide detailed information for the drug interaction pairs assessed as levels one and two. Among the ARV pharmacologic groups, the protease inhibitors (PIs) and RTV/COBI-boosted PIs, the integrase strand transfer inhibitors (InSTIs), and the non-nucleoside reverse transcriptase inhibitors (NNRTIs) with 70 (24.1%), 65 (22.3%), and 37 (15.9%) drug interaction pairs of levels one and two, respectively, were the more frequent (Table 3 and Table 4). In addition, for 233 (80.1%) of the 291 drug interaction pairs, the pharmacokinetic mechanism was the most frequent, including bioavailability modifications based on P-gp or presystemic enzyme alterations (Table 2).
Globally, among the 291 interactions systematically reported for the first time, 50 (17.2%) were assessed as being level five; thus, with evidence of the absence of clinically relevant drug interactions, 20 (6.1%) for InSTIs, 14 (4.8%) for NNRTIs, and 9 (3.1%) for PIs and RTV/COBI-boosted PIs were the more frequent (Table 5). In addition, information regarding 55 drug interaction pairs was related to drugs not yet approved or to discontinued drugs or with minimum use in clinal practice, with 43 corresponding to PIs (indinavir, nelfinavir, saquinavir, tipranavir, and fosamprenavir), 11 corresponding to NRTIs (didanosine and stavudine), and 1 to a sulfonylurea (chlorpropamide).
Among 179 drug interactions of levels one and two, cation-containing antacids/supplements and iron or zinc products, antiulcer (proton pump inhibitors and anti-H2), with 29, (16.2%) and oral anticoagulants (warfarin and direct-anticoagulants), with 23 (12,8%), were the non-ARV pharmacologic groups with more drug interaction pairs. Additionally, there were six (3.4%) clinically relevant drug interactions (level two) associated with drug or substance misuse, such as amphetamines or ketamine, mainly due to the concomitant use of boosted PIs and elvitegravir/cobicistat (EVG/COBI). Additionally, six (3.4%) drug interaction pairs with benzodiazepines were identified (Table 3 and Table 4).

4. Discussion

Although current ARV therapy is simplified, safe, and effective, the combination of three ARV drugs or two with some recent regimens [2] increases the risk of clinically relevant drug interactions, mainly in patients with one or more long-term disease in addition to HIV. Currently, some systematic reviews, meta-analyses, and randomized clinical trials provide sufficient evidence that some drug interactions may cause negative health outcomes in PLHIV. Overall, clinically relevant drug interactions occur in 20–30% of PLHIVs [207], including recently authorized ARV drugs, mainly explained through pharmacokinetics changes linked to the inhibition or induction of different enzymes and metabolic transporters [3,5,6,7,8,208].
The current review identified 219 articles with information that allowed us to assess and classify the clinical relevance of 471 drug interaction pairs in PLHIVs on antiretroviral therapy. Among these 471 drug interaction pairs, 291 were systematically reported for the first time, which shows the need for conducting a periodic update on this topic through comprehensive reviews [5,6,7,8].
In the current update, we identified 291 drug interaction pairs systematically reported for the first time; among them, 179 (61.1%) were assessed as the most clinically relevant (levels one or two). These figures were lower than reported in the update for 2015–2017 [8], in which 534 drug interaction pairs were systematically reported for the first time and 308 (64.2%) were assessed as levels one or two. This reduction may be due to the current guidelines [2] recommending, as first therapeutic options, regimes with more recent ARV drugs, such as BIC (authorized in 2018) or CAB (authorized in 2021), which have less probability of pharmacokinetic interactions. In this sense, among 50 interaction pairs assessed as level five, 20 (40.0%) were related to InSTIs (Table 5). Overall, second-generation InSTIs (CAB, BIC, and DTG), compared to PIs, have less probability to inhibit the metabolism of CYP450 isoenzymes and, therefore, a lesser risk of clinically relevant drug interactions [2,209]. Similarly, for NNRTIs, 14 (28.0%) drug interaction pairs were assessed as level five, which may be explained due to more recent drugs (second generation), for instance, doravirine (DOR), etravirine (ETR), and rilpivirine (RPV) may have less probability of pharmacokinetic interactions in comparison with nevirapine (NVP) and efavirenz (EFV) [5,6,7,8].
The pharmacokinetic mechanism was the most frequent to explain the drug interaction; thus, it was the mechanism to 233 (80.1%) of the 291 pairs systematically reported for the first time. The pharmacokinetics of ARV involve the isoenzymes of the cytochrome P450 enzyme (CYP) family, such as CYP3A4, CYP2B6, and CYP2C9, enzyme of the glucuronidation pathway, such as UGT1A1, as well as uptake transporters, such as OCT2, MATE1, OATP1B1, and export proteins, such as glycoprotein P (P-gp), which increase the probability of pharmacokinetic drug interactions [210]. Some ARV drugs, especially PIs and NNRTIs, are considered strong inhibitors or inducers of various isoenzymes of the CYP450 family, as well as carrier proteins, which increase the risk of significant clinical drug interactions, thereby increasing the risk of not achieving the therapeutic goals in PLHIVs [205].
In the current review, the ARV groups with more clinically relevant drug interaction pairs were the PIs and RTV/COBI-boosted PIs, the InSTIs, and the NNRTIs, with 70 (24.1%), 65 (22.3%), and 37(15.9%) drug interaction pairs of levels one and two, respectively, for PIs and RTV/COBI-boosted PIs and NNRTIs, likely due to their metabolism through the CYP450 family [210], and for InSTIs, mainly related to EVG/COBI, likely due to both metabolism through the CYP3A4 and the inhibition of CYP3A4 activity through COBI. Similarly, a retrospective study found that the most frequently involved ARVs were RTV/COBI-enhanced PIs (49.3%), followed by NNRTIs (38.3%) [211]. However, these results were slightly different from the findings in the 2015–2017 update, in which PIs were the predominant group with a percentage of 29.2% [8].
Drug substance misuse is an important consideration in PLHIV on ARV therapy, requiring an integrated approach based on evidence [2]. Therefore, there are clinically relevant drug interactions associated with drug substance misuse, such as ketamine, amphetamine, and substitutes (methamphetamine, methylenedioxymethamphetamine—MDMA; ‘ecstasy’) with RTV-boosted PIs [14,75,76,77,78,79,80] and EVG/COBI [14,15,75,76,80,142,143], increasing the risk of toxicity, including a possible fatal serotonergic reaction. Furthermore, clinically relevant interactions were identified between psychotropic drugs, particularly benzodiazepines, which have the potential to cause dependence in patients. In combination with boosted PIs and EVG/COBI, the probability of respiratory depression, sedation, and muscle weakness was increased [13,14,15,93]. It is important to denote that, currently, there is increasing awareness about the probability occurrence of clinical relevance of cannabis–drug interactions [212], particularly with efavirenz (EFV) and COBI or RTV-boosted regimes, which may increase plasma levels of cannabis, prolonging its clinical effects and increasing toxicity [33].
The method for assessing the clinical relevance of drug interactions used in this review was similar to that used in previous reviews [5,6,7,8]; therefore, these results were useful in updating and synthesizing the previous identified information regarding ARV drug interactions. Thereby, the 291 drug interaction pairs systematically reported for the first time should be used to update the mobile application for analyzing the clinical relevance of ARV drug interactions (InterApp ARV) [10], which is an evolution of the SIMARV® software [9]. The application is freely accessible and can be downloaded for Android devices from the Play Store (https://play.google.com/store/apps/details?id=co.com.pypudea.interapparv accessed on 25 July 2023).
Neither the current nor the previous reviews included the specific search for pharmacogenetic interactions [5,6,7,8], mainly, gene–antiretroviral drug interactions. However, this issue emerges as a key explanation for clinically relevant drug interactions. For instance, patient genetics explain the extent of the inductor effect of efavirenz or nevirapine on etonogestrel pharmacokinetics, and show that drug interactions with NNRTIs are influenced by host genetics. Thus, the combination of efavirenz plus etonogestrel/ethinylestradiol (vaginal ring) results in an unfavorable drug–drug interaction regardless of patient genetics (Table 3), but it is most notorious in women with variant alleles for CYP2B6 single-nucleotide polymorphisms (slow metabolizer genotype) [60,61]. As a consequence, this issue should be included in future systematic reviews.
The results of this review may have some limitations; therefore, the results should be interpreted and used with caution. In this context, the main limitation was the search restriction to a single database, since the search was performed only in the PubMed/MEDLINE database, which may not have identified other clinically relevant interactions. However, this situation could be minimized with the inclusion of publications identified as relevant in the reference list of the articles included. In addition, the method for assessing and classifying the level of relevance could be considered a subjective scale of “clinical significance”, which was not subject to a validation process. However, the method was proposed in 2007 [3] and updated in 2021 [4], and among several reviews of clinically relevant drug interactions, it was used for four previous reviews regarding antiretroviral drug interactions [5,6,7,8] for other pharmacologic groups, for instance, cannabis [212], or for hypolipidemic agents [213] and for specific drug–drug interactions, for instance, for nonsteroidal anti-inflammatory drugs and antihypertensives [214]. Additionally, it was used for assessing drug interactions in different settings, for instance, in intensive care [215].

5. Conclusions

From 2017 to 2022, in the PubMed/Medline database, we identified 219 records, including 87 from citation lists, related to 291 drug interaction pairs in PLHIVs in patients living with HIV on antiretroviral therapy. Thus, the clinical relevance of 471 drug interaction pairs was assessed; of them, 291 were pairs systematically reported for the first time, a figure that was lower than the one reported in the update for 2015–2017. Among the 291 drug interaction pairs systematically reported for the first time, 179 (61.5%) were assessed as level one (42) or level two (137), thus, with a high risk of causing adverse drug outcomes linked to ineffectiveness or unsafety of the pharmacotherapy. Pharmacokinetics is the mechanism most frequently identified to explain drug interactions. In addition, PIs and RTV/COBI-boosted PIs and InSTIs were the ARV drugs with a greater number of clinically relevant interactions. Cation-containing antacids/supplements and iron or zinc products, antiulcer (proton pump inhibitors and anti-H2), and oral anticoagulants were drug groups most frequently with drug interactions with ARV, accounting for 16.2% and 12.8% of cases, respectively. In PLHIVs, clinically relevant drug interactions were associated with drug substance misuse, mainly amphetamines and psychotropic drugs, particularly benzodiazepines.

Author Contributions

Database search, screening of articles, and extraction of information: M.R.-C., M.C. and P.A.; writing of the manuscript: M.C., M.R.-C. and P.A. All authors have read and agreed to the published version of the manuscript.

Funding

The Pharmaceutical Promotion and Prevention Group received financial support from the Committee for Development Research (CODI) and sustainability program (2018–2019), Universidad de Antioquia.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Data are contained within this article.

Acknowledgments

The authors acknowledge the members of the Research Group on Pharmaceutical Promotion and Prevention of the University of Antioquia for their comments and suggestions about the manuscript.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. World Health Organization (WHO). HIV and AIDS, July of 2023. Available online: https://www.who.int/news-room/fact-sheets/detail/hiv-aids (accessed on 5 September 2023).
  2. Gandhi, R.T.; Bedimo, R.; Hoy, J.F.; Landovitz, R.J.; Smith, D.M.; Eaton, E.F.; Lehmann, C.; Springer, S.A.; Sax, P.E.; Thompson, M.A.; et al. Antiretroviral Drugs for Treatment and Prevention of HIV Infection in Adults: 2022 Recommendations of the International Antiviral Society-USA Panel. JAMA 2023, 329, 63–84. [Google Scholar] [CrossRef] [PubMed]
  3. Amariles, P.; Giraldo, N.A.; Faus, M.J. Interacciones medicamentosas: Aproximación para establecer y evaluar su relevancia clínica. Med. Clin. 2007, 129, 27–35. [Google Scholar] [CrossRef] [PubMed]
  4. Amariles, P.; Madrigal-Cadavid, J.; Giraldo, N.A. Clinical relevance of drug interactions: Proposal to update the classification based on the severity and probability of its occurrence. Rev. Chil. Infectol. 2021, 38, 304–305. [Google Scholar] [CrossRef]
  5. Amariles, P.; Giraldo, N.A.; Faus, M.J. Interacciones medicamentosas en pacientes infectados con el VIH: Aproximación para establecer y evaluar su relevancia clínica. Farm Hosp. 2007, 31, 283–302. [Google Scholar] [CrossRef]
  6. Giraldo, N.A.; Amariles, P.; Gutiérrez, F.J.; Monsalve, M.; Faus, M.J. Aproximación para establecer y evaluar la relevancia clínica de las interacciones medicamentosas en pacientes infectados con virus de la inmunodeficiencia humana: Actualización 2009. Farm. Hosp. 2010, 34, 90–93. [Google Scholar] [CrossRef] [PubMed]
  7. Giraldo, N.A.; Amariles, P.; Pino Marín, D.E.; Faus, M.J. Relevancia clínica de las interacciones medicamentosas en pacientes infectados con el virus de la inmunodeficiencia humana: Actualización 2009–2014. Rev. Chil. Infectol. 2016, 33, 36–53. [Google Scholar] [CrossRef]
  8. Osorio, T.L.; Rivera, C.M.; Pino-Marín, D.E.; Giraldo, N.A.; Amariles, P. Relevancia clínica de las interacciones medicamentosas en pacientes infectados con el virus de la inmunodeficiencia humana: Actualización 2015–2017. Rev. Chil. Infectol. 2019, 36, 475–489. [Google Scholar] [CrossRef]
  9. Giraldo, N.A.; Amariles, P.; Monsalve, M.; Faus, M.J. Free software to analyse the clinical relevance of drug interactions with antiretroviral agents (SIMARV®) in patients with HIV/AIDS. Res. Soc. Adm. Pharm. RSAP 2017, 13, 831–839. [Google Scholar] [CrossRef]
  10. Amariles, P.; Giraldo, N.; Madrigal-Cadavid, J.; Granados, J. Free InterApp ARV: The subsequent version of SIMARV® to analyse the clinical relevance of drug interactions with antiretroviral agents in patients with HIV/AIDS. Res. Soc. Adm. Pharm. RSAP 2019, 15, 1376. [Google Scholar] [CrossRef]
  11. University of Liverpool. HIV Drug Interactions—Interaction Checker. 2023. Available online: https://www.hiv-druginteractions.org/checker (accessed on 10 July 2023).
  12. 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, n71. [Google Scholar] [CrossRef]
  13. Marzolini, C.; Livio, F. Prescribing issues in elderly individuals living with HIV. Expert Rev. Clin. Pharmacol. 2019, 12, 643–659. [Google Scholar] [CrossRef] [PubMed]
  14. Chary, A.; Nguyen, N.N.; Maiton, K.; Holodniy, M. A review of drug-drug interactions in older HIV-infected patients. Expert Rev. Clin. Pharmacol. 2017, 10, 1329–1352. [Google Scholar] [CrossRef]
  15. Panel on Antiretroviral Guidelines for Adults and Adolescents. Guidelines for the Use of Antiretroviral Agents in Adults and Adolescents with HIV. Department of Health and Human Services. Available online: https://clinicalinfo.hiv.gov/en/guidelines/adult-and-adolescent-arv (accessed on 31 July 2023).
  16. Hsu, A.J.; Carson, K.A.; Yung, R.; Pham, P.A. Severe prolonged sedation associated with coadministration of protease inhibitors and intravenous midazolam during bronchoscopy. Pharmacotherapy 2012, 32, 538–545. [Google Scholar] [CrossRef]
  17. Ali, M.S.; Kooragayalu, S.; Mba, B.I.; Olsen, F.C. Isolated Myopathy: An Unusual Manifestation of Inhaled Fluticasone Propionate and Ritonavir Interaction. Arch. Bronconeumol. 2019, 55, 223–225. [Google Scholar] [CrossRef] [PubMed]
  18. Cattaneo, D.; Baldelli, S.; Cozzi, V.; Fusi, M.; Atzori, C.; Micheli, V.; Filice, C.; Gervasoni, C. Drug-Drug Interactions Between Antiretrovirals and Carbamazepine/Oxcarbazepine: A Real-Life Investigation. Ther. Drug Monit. 2020, 42, 330–334. [Google Scholar] [CrossRef] [PubMed]
  19. Calderón, M.M.; Penzak, S.R.; Pau, A.K.; Kumar, P.; McManus, M.; Alfaro, R.M.; Kovacs, J.A. Efavirenz but Not Atazanavir/Ritonavir Significantly Reduces Atovaquone Concentrations in HIV-Infected Subjects. Clin. Infect. Dis. 2016, 62, 1036–1042. [Google Scholar] [CrossRef]
  20. Van Luin, M.; Van der Ende, M.E.; Richter, C.; Visser, M.; Faraj, D.; Van der Ven, A.; Gelinck, L.; Kroon, F.; Wit, F.W.; Van Schaik, R.H.; et al. Lower atovaquone/proguanil concentrations in patients taking efavirenz, lopinavir/ritonavir or atazanavir/ritonavir. AIDS 2010, 24, 1223–1226. [Google Scholar] [CrossRef]
  21. Tommasi, C.; Bellagamba, R.; Tempestilli, M.; D’Avolio, A.; Gallo, A.L.; Ivanovic, J.; Nicastri, E.; Pucillo, L.P.; Narciso, P. Marked increase in etravirine and saquinavir plasma concentrations during atovaquone/proguanil prophylaxis. Malar J. 2011, 10, 141. [Google Scholar] [CrossRef]
  22. Chastain, D.B.; Franco-Paredes, C.; Stover, K.R. Addressing Antiretroviral Therapy-Associated Drug-Drug Interactions in Patients Requiring Treatment for Opportunistic Infections in Low-Income and Resource-Limited Settings. J. Clin. Pharmacol. 2017, 57, 1387–1399. [Google Scholar] [CrossRef]
  23. Devanathan, A.S.; Anderson, D.J.C.; Cottrell, M.L.; Burgunder, E.M.; Saunders, A.C.; Kashuba, A.D.M. Contemporary Drug–Drug Interactions in HIV Treatment. Clin. Pharmacol. Ther. 2019, 105, 1362–1377. [Google Scholar] [CrossRef]
  24. Soyinka, J.O.; Onyeji, C.O. Alteration of pharmacokinetics of proguanil in healthy volunteers following concurrent administration of efavirenz. Eur. J. Pharm. Sci. 2010, 39, 213–218. [Google Scholar] [CrossRef]
  25. Seden, K.; Gibbons, S.; Marzolini, C.; Schapiro, J.M.; Burger, D.M.; Back, D.J.; Khoo, S.H. Development of an evidence evaluation and synthesis system for drug-drug interactions, and its application to a systematic review of HIV and malaria co-infection. PLoS ONE 2017, 12, e0173509. [Google Scholar] [CrossRef] [PubMed]
  26. Usman, S.O.; Oreagba, I.A.; Busari, A.; Akinyede, A.; Adewumi, O.; Kadri, M.R.; Hassan, O.; Fashina, Y.A.; Agbaje, E.O.; Akanmu, S.A. Evaluation of cardiotoxicity and other adverse effects associated with concomitant administration of artemether/lumefantrine and atazanavir/ritonavir-based antiretroviral regimen in patients living with HIV. Saudi. Pharm. J. 2022, 30, 605–612. [Google Scholar] [CrossRef] [PubMed]
  27. Banda, C.G.; Dzinjalamala, F.; Mukaka, M.; Mallewa, J.; Maiden, V.; Terlouw, D.J.; Lalloo, D.G.; Khoo, S.H.; Mwapasa, V. Pharmacokinetics and Safety Profile of Artesunate-Amodiaquine Coadministered with Antiretroviral Therapy in Malaria-Uninfected HIV-Positive Malawian Adults. Antimicrob. Agents Chemother. 2018, 62, e00412-18. [Google Scholar] [CrossRef] [PubMed]
  28. Haaland, R.E.; Otieno, K.; Martin, A.; Katana, A.; Dinh, C.; Slutsker, L.; Menendez, C.; Gonzalez, R.; Williamson, J.; Heneine, W.; et al. Short Communication: Reduced Nevirapine Concentrations Among HIV-Positive Women Receiving Mefloquine for Intermittent Preventive Treatment for Malaria Control During Pregnancy. AIDS Res. Hum. Retroviruses 2018, 34, 912–915. [Google Scholar] [CrossRef] [PubMed]
  29. Hughes, E.; Mwebaza, N.; Huang, L.; Kajubi, R.; Nguyen, V.; Nyunt, M.M.; Orukan, F.; Mwima, M.W.; Parikh, S.; Aweeka, F. Efavirenz-Based Antiretroviral Therapy Reduces Artemether-Lumefantrine Exposure for Malaria Treatment in HIV-Infected Pregnant Women. J. Acquir. Immune. Defic. Syndr. 2020, 83, 140–147. [Google Scholar] [CrossRef]
  30. Francis, J.; Barnes, K.I.; Workman, L.; Kredo, T.; Vestergaard, L.S.; Hoglund, R.M.; Byakika-Kibwika, P.; Lamorde, M.; Walimbwa, S.I.; Chijioke-Nwauche, I.; et al. An Individual Participant Data Population Pharmacokinetic Meta-analysis of Drug-Drug Interactions between Lumefantrine and Commonly Used Antiretroviral Treatment. Antimicrob. Agents Chemother. 2020, 64, e02394-19. [Google Scholar] [CrossRef]
  31. Zakaria, Z.; Badhan, R.K.S. The impact of CYP2B6 polymorphisms on the interactions of efavirenz with lumefantrine: Implications for paediatric antimalarial therapy. Eur. J. Pharm. Sci. 2018, 119, 90–101. [Google Scholar] [CrossRef]
  32. Maganda, B.A.; Minzi, O.M.; Ngaimisi, E.; Kamuhabwa, A.A.; Aklillu, E. CYP2B6*6 genotype and high efavirenz plasma concentration but not nevirapine are associated with low lumefantrine plasma exposure and poor treatment response in HIV-malaria-coinfected patients. Pharmacogenomics J. 2016, 16, 88–95. [Google Scholar] [CrossRef]
  33. Usman, S.O.; Oreagba, I.A.; Akinyede, A.A.; Agbaje, E.O.; Akinleye, M.O.; Onwujuobi, A.G.; Ken-Owotor, C.; Adeuja, O.; Ogunfowokan, T.; Kogbe, S.; et al. Effect of nevirapine, efavirenz and lopinavir/ritonavir on the therapeutic concentration and toxicity of lumefantrine in people living with HIV at Lagos University Teaching Hospital, Nigeria. J. Pharmacol. Sci. 2020, 144, 95–101. [Google Scholar] [CrossRef]
  34. Naidoo, A.; Chirehwa, M.; McIlleron, H.; Naidoo, K.; Essack, S.; Yende-Zuma, N.; Kimba-Phongi, E.; Adamson, J.; Govender, K.; Padayatchi, N.; et al. Effect of rifampicin and efavirenz on moxifloxacin concentrations when co-administered in patients with drug-susceptible TB. J. Antimicrob. Chemother. 2017, 72, 1441–1449. [Google Scholar] [CrossRef]
  35. Back, D.; Marzolini, C. The challenge of HIV treatment in an era of polypharmacy. J. Int. AIDS Soc. 2020, 23, e25449. [Google Scholar] [CrossRef] [PubMed]
  36. Khalilieh, S.; Yee, K.L.; Sanchez, R.; Stoch, S.A.; Wenning, L.; Iwamoto, M. Clinical Pharmacokinetics of the Novel HIV-1 Non-Nucleoside Reverse Transcriptase Inhibitor Doravirine: An Assessment of the Effect of Patient Characteristics and Drug-Drug Interactions. Clin. Drug Investig. 2020, 40, 927–946. [Google Scholar] [CrossRef] [PubMed]
  37. Boyle, A.; Moss, C.E.; Marzolini, C.; Khoo, S. Clinical Pharmacodynamics, Pharmacokinetics, and Drug Interaction Profile of Doravirine. Clin. Pharmacokinet. 2019, 58, 1553–1565. [Google Scholar] [CrossRef] [PubMed]
  38. Khalilieh, S.G.; Yee, K.L.; Sanchez, R.I.; Liu, R.; Fan, L.; Martell, M.; Jordan, H.; Iwamoto, M. Multiple doses of rifabutin reduce exposure of doravirine in healthy subjects. J. Clin. Pharmacol. 2018, 58, 1044–1052. [Google Scholar] [CrossRef]
  39. Atwine, D.; Bonnet, M.; Taburet, A.M. Pharmacokinetics of efavirenz in patients on antituberculosis treatment in high human immunodeficiency virus and tuberculosis burden countries: A systematic review. Br. J. Clin. Pharmacol. 2018, 84, 1641–1658. [Google Scholar] [CrossRef]
  40. Hodge, D.; Back, D.J.; Gibbons, S.; Khoo, S.H.; Marzolini, C. Pharmacokinetics and Drug–Drug Interactions of Long-Acting Intramuscular Cabotegravir and Rilpivirine. Clin. Pharmacokinet. 2021, 60, 835–853. [Google Scholar] [CrossRef]
  41. Schafer, J.J.; Short, W.R. Rilpivirine, a novel non-nucleoside reverse transcriptase inhibitor for the management of HIV-1 infection: A systematic review. Antivir. Ther. 2012, 17, 1495–1502. [Google Scholar] [CrossRef]
  42. Crauwels, H.; van Heeswijk, R.P.G.; Kestens, D.; Stevens, M.; Buelens, A.; Boven, K.; Hoetelmans, R.M.W. The pharmacokinetic (PK) interaction between rifabutin and TMC278, an investigational non-nucleoside reverse transcriptase inhibitor (NNRTI). In Proceedings of the 17th International AIDS Conference, Mexico City, Mexico, 3–8 August 2008. Abstract TUPE0080. [Google Scholar]
  43. Cerrone, M.; Bracchi, M.; Wasserman, S.; Pozniak, A.; Meintjes, G.; Cohen, K.; Wilkinson, R.J. Safety implications of combined antiretroviral and anti-tuberculosis drugs. Expert Opin. Drug Saf. 2020, 19, 23–41. [Google Scholar] [CrossRef]
  44. Ebrahim, I.; Maartens, G.; Wiesner, L.; Orrell, C.; Smythe, W.; McIlleron, H. Pharmacokinetic profile and safety of adjusted doses of darunavir/ritonavir with rifampicin in people living with HIV. J. Antimicrob. Chemother. 2020, 75, 1019–1025. [Google Scholar] [CrossRef]
  45. Yee, K.L.; Khalilieh, S.G.; Sanchez, R.I.; Liu, R.; Anderson, M.S.; Manthos, H.; Judge, T.; Brejda, J.; Butterton, J.R. The Effect of Single and Multiple Doses of Rifampin on the Pharmacokinetics of Doravirine in Healthy Subjects. Clin. Drug Investig. 2017, 37, 659–667. [Google Scholar] [CrossRef] [PubMed]
  46. Van Heeswijk, R.; Hoetelmans, R.; Kestens, D.; Stevens, M.; Peeters, M.; Boven, K.; Woodfall, B. The effects of CYP3A4 modulation on the pharmacokinetics of TMC278, an investigational non-nucleoside reverse transcriptase inhibitor (NNRTI). In Proceedings of the 7th International Workshop on Clinical Pharmacology of HIV Therapy, Lisbon, Portugal, 20–22 April 2006. Abstract 45. [Google Scholar]
  47. Cerrone, M.; Alfarisi, O.; Neary, M.; Marzinke, M.A.; Parsons, T.L.; Owen, A.; Maartens, G.; Pozniak, A.; Flexner, C.; Boffito, M. Rifampicin effect on intracellular and plasma pharmacokinetics of tenofovir alafenamide. J. Antimicrob. Chemother. 2019, 74, 1670–1678. [Google Scholar] [CrossRef] [PubMed]
  48. Freise, K.J.; Hu, B.; Salem, A.H. Impact of ritonavir dose and schedule on CYP3A inhibition and venetoclax clinical pharmaco-kinetics. Eur. J. Clin. Pharmacol. 2018, 74, 413–421. [Google Scholar] [CrossRef]
  49. Cordova, E.; Morganti, L.; Odzak, A.; Arcondo, F.; Silva, M.; Zylberman, M.; Rodriguez, C. Severe hypokalemia due to a possible drug-drug interaction between vinblastine and antiretrovirals in a HIV-infected patient with Hodgkin’s lymphoma. Int. J. STD AIDS 2017, 28, 1259–1262. [Google Scholar] [CrossRef] [PubMed]
  50. Mutiti, C.S.; Kapungu, N.N.; Kanji, C.R.; Stadler, N.; Stingl, J.; Nhachi, C.; Hakim, J.; Masimirembwa, C.; Thelingwani, R.S. Clinically relevant enantiomer specific R- and S-praziquantel pharmacokinetic drug-drug interactions with efavirenz and ritonavir. Pharmacol. Res. Perspect. 2021, 9, e00769. [Google Scholar] [CrossRef]
  51. Itkonen, M.K.; Tornio, A.; Lapatto-Reiniluoto, O.; Neuvonen, M.; Neuvonen, P.J.; Niemi, M.; Backman, J.T. Clopidogrel increases dasabuvir exposure with or without ritonavir, and ritonavir inhibits the bioactivation of clopidogrel. Clin. Pharmacol. Ther. 2019, 105, 219–228. [Google Scholar] [CrossRef]
  52. Marsousi, N.; Daali, Y.; Fontana, P.; Reny, J.L.; Ancrenaz-Sirot, V.; Calmy, A.; Rudaz, S.; Desmeules, J.A.; Samer, C.F. Impact of boosted antiretroviral therapy on the pharmacokinetics and efficacy of clopidogrel and prasugrel active metabolites. Clin. Pharmacokinet. 2018, 57, 1347–1354. [Google Scholar] [CrossRef]
  53. Bravo, I.; Álvarez, H.; Mariño, A.; Clotet, B.; Moltó, J. Recurrent coronary disease in HIV-infected patients: Role of drug-drug interactions. Br. J. Clin. Pharmacol. 2018, 84, 1617–1619. [Google Scholar] [CrossRef]
  54. Van Heeswijk, R.; Hoetelmans, R.; Kestens, D.; Stevens, M.; Peeters, M.; Williams, P.; Woodfall, B.; Boven, K. The pharmacokinetic interaction between famotidine and TMC278, a next generation non-nucleoside reverse transcriptase inhibitor, in HIV-negative volunteers. In Proceedings of the 4th IAS Conference on HIV Pathogenesis, Treatment and Prevention, Sydney, NSW, Australia, 22–25 July 2007. Abstract TUPDB01. [Google Scholar]
  55. Parant, F.; Miailhes, P.; Brunel, F.; Gagnieu, M.C. Dolutegravir Population Pharmacokinetics in a Real-Life Cohort of People Living with HIV Infection: A Covariate Analysis. Ther. Drug Monit. 2019, 41, 444–451. [Google Scholar] [CrossRef]
  56. Scarsi, K.K.; Darin, K.M.; Nakalema, S.; Back, D.J.; Byakika-Kibwika, P.; Else, L.J.; Dilly Penchala, S.; Buzibye, A.; Cohn, S.E.; Merry, C.; et al. Unintended pregnancies observed with combined use of the levonorgestrel contraceptive implant and efavirenz-based antiretroviral therapy: A three-arm pharmacokinetic evaluation over 48 weeks. Clin. Infect. Dis. 2016, 62, 675–682. [Google Scholar] [CrossRef]
  57. Scarsi, K.K.; Cirrincione, L.; Nakalema, S.; Darin, K.; Musinguzi, I.; Kyohairwe, I.; Byakika-Kibwika, P.; Owen, A.; Winchester, L.; Podany, A.; et al. Double-dose levonorgestrel implant does not fully overcome drug-drug interaction with efavirenz. In Proceedings of the Conference on Retroviruses and Opportunistic Infections, Seattle, WA, USA, 7–9 March 2019. Abstract O51. [Google Scholar]
  58. Patel, R.C.; Stalter, R.M.; Thomas, K.K.; Tamraz, B.; Blue, S.W.; Erikson, D.W.; Kim, C.J.; Kelly, E.J.; Nanda, K.; Kourtis, A.P.; et al. A pharmacokinetic and pharmacogenetic evaluation of contraceptive implants and antiretroviral therapy among women in Kenya and Uganda. AIDS 2019, 33, 1995–2004. [Google Scholar] [CrossRef] [PubMed]
  59. Chappell, C.A.; Lamorde, M.; Nakalema, S.; Chen, B.A.; Mackline, H.; Riddler, S.A.; Cohn, S.E.; Darin, K.M.; Achilles, S.L.; Scarsi, K.K. Efavirenz decreases etonogestrel exposure: A pharmacokinetic evaluation of implantable contraception with antiretroviral therapy. AIDS 2017, 31, 1965–1972. [Google Scholar] [CrossRef]
  60. Scarsi, K.K.; Cramer, Y.S.; Rosenkranz, S.L.; Aweeka, F.; Berzins, B.; Coombs, R.W.; Coughlin, K.; Moran, L.E.; Zorrilla, C.D.; Akelo, V.; et al. Antiretroviral therapy and vaginally administered contraceptive hormones: A three arm, pharmacokinetic study. Lancet HIV 2019, 6, e601–e612. [Google Scholar] [CrossRef]
  61. Haas, D.W.; Cramer, Y.S.; Godfrey, C.; Rosenkranz, S.L.; Aweeka, F.; Berzins, B.; Coombs, R.; Coughlin, K.; Moran, L.E.; Gingrich, D.; et al. Pharmacogenetic interactions between antiretroviral drugs and vaginally administered hormonal contraceptives. Pharmacogenet. Genom. 2020, 30, 45–53. [Google Scholar] [CrossRef] [PubMed]
  62. Haas, D.W.; Mngqibisa, R.; Francis, J.; McIlleron, H.; Robinson, J.A.; Kendall, M.A.; Baker, P.; Mawlana, S.; Badal-Faesen, S.; Angira, F.; et al. Pharmacogenetics of interaction between depot medroxyprogesterone acetate and efavirenz, rifampicin, and isoniazid during treatment of HIV and tuberculosis. Pharmacogenet Genom. 2022, 32, 24–30. [Google Scholar] [CrossRef]
  63. Neary, M.; Chappell, C.A.; Scarsi, K.K.; Nakalema, S.; Matovu, J.; Achilles, S.L.; Chen, B.A.; Siccardi, M.; Owen, A.; Lamorde, M. Effect of patient genetics on etonogestrel pharmacokinetics when combined with efavirenz or nevirapine ART. J. Antimicrob. Chemother. 2019, 74, 3003–3010. [Google Scholar] [CrossRef] [PubMed]
  64. Kosloski, M.P.; Oberoi, R.; Wang, S.; Viani, R.M.; Asatryan, A.; Hu, B.; Ding, B.; Qi, X.; Kim, E.J.; Mensa, F.; et al. Drug-Drug Interactions of Glecaprevir and Pibrentasvir coadministered with Human Immunodeficiency Virus Antiretrovirals. J. Infect. Dis. 2020, 221, 223–231. [Google Scholar] [CrossRef] [PubMed]
  65. Hong, J.; Wright, R.C.; Partovi, N.; Yo-shida, E.M.; Hussaini, T. Review of Clinically Relevant Drug Interactions with Next Generation Hepatitis C Direct-acting Antiviral Agents. J. Clin. Transl. Hepatol. 2020, 8, 322–335. [Google Scholar] [CrossRef]
  66. Garrison, K.L.; Kirby, B.; Stamm, L.M.; Ma, G.; Vu, A.; Ling, J.; Mathias, A. Drug-drug interaction profile of sofosbuvir/velpatasvir/voxilaprevir fixed-dose combination. J. Hepatol. 2017, 66, S492–S493. [Google Scholar] [CrossRef]
  67. Mogalian, E.; Stamm, L.M.; Osinusi, A.; Brainard, D.M.; Shen, G.; Ling, K.H.J.; Mathias, A. Drug-Drug Interaction Studies Be-tween Hepatitis C Virus Antivirals Sofosbuvir/Velpatasvir and Boosted and Unboosted Human Immunodeficiency Virus Antiretroviral Regimens in Healthy Volunteers. Clin. Infect. Dis. 2018, 67, 934–940. [Google Scholar] [CrossRef]
  68. Feng, H.P.; Caro, L.; Fandozzi, C.; Chu, X.; Guo, Z.; Talaty, J.; Panebianco, D.; Dunnington, K.; Du, L.; Hanley, W.D.; et al. Pharmacokinetic Interactions between the Hepatitis C Virus Inhibitors Elbasvir and Grazoprevir and HIV Protease Inhibitors Ritonavir, Atazanavir, Lopinavir, and Darunavir in Healthy Volunteers. Antimicrob. Agents Chemother. 2019, 63, e02142-18. [Google Scholar] [CrossRef]
  69. Tempestilli, M.; Fabbri, G.; Mastrorosa, I.; Timelli, L.; Notari, S.; Bellagamba, R.; Libertone, R.; Lupi, F.; Zaccarelli, M.; Antinori, A.; et al. Plasma trough concentrations of antiretrovirals in HIV-infected persons treated with direct acting antiviral agents for hepatitis C in the real world. J. Antimicrob. Chemother. 2018, 73, 160–164. [Google Scholar] [CrossRef]
  70. Sabourin, A.A.; Patel, T.; Saad, S.; Renner, E.; Mouland, E.; Adie, S.; Ha, N.B. Management of anticoagulation in patients with human immunodeficiency virus/acquired immunodeficiency virus. Thromb. Res. 2021, 200, 102–108. [Google Scholar] [CrossRef] [PubMed]
  71. Nisly, S.A.; Stevens, B.N. Ritonavir- or cobicistat-boosted antiretroviral therapy and direct oral anticoagulants: A case for apixaban. Int. J. STD AIDS 2019, 30, 718–722. [Google Scholar] [CrossRef] [PubMed]
  72. Capiau, A.; Mehuys, E.; De Bolle, L.; Van Tongelen, I.; De Backer, T.; Boussery, K. Drug-drug interactions with direct oral anticoagulants: Development of a consensus list for ambulatory care. Int. J. Clin. Pharm. 2023, 45, 364–374. [Google Scholar] [CrossRef] [PubMed]
  73. Mueck, W.; Kubitza, D.; Becka, M. Co-administration of rivaroxaban with drugs that share its elimination pathways: Pharmacokinetic effects in healthy subjects. Br. J. Clin. Pharmacol. 2013, 76, 455–466. [Google Scholar] [CrossRef] [PubMed]
  74. Corallo, C.E.; Grannell, L.; Tran, H. Post-operative bleeding after administration of a single dose of rivaroxaban to a patient receiving antiretroviral therapy. Drug Saf. Case Rep. 2015, 2, 11. [Google Scholar] [CrossRef] [PubMed]
  75. Chen, G.L.; Lin, S.Y.; Lo, H.Y.; Wu, H.C.; Lin, Y.M.; Chen, T.C.; Sandy Chu, C.Y.; Lee, W.C.; Chen, Y.H.; Lu, P.L. Clinical impact of recreational drug use among people living with HIV in southern Taiwan. J. Microbiol. Immunol. Infect. 2021, 54, 952–962. [Google Scholar] [CrossRef]
  76. Castro-Granell, V.; Garin, N.; Jaén, Á.; Cenoz, S.; Galindo, M.J.; Fuster-RuizdeApodaca, M.J. Prevalence, beliefs and impact of drug-drug interactions between antiretroviral therapy and illicit drugs among people living with HIV in Spain. PLoS ONE 2021, 16, e0260334. [Google Scholar] [CrossRef]
  77. Antoniou, T.; Tseng, A.L. Interactions between recreational drugs and antiretroviral agents. Ann. Pharmacother. 2002, 36, 1598–1613. [Google Scholar] [CrossRef]
  78. Hales, G.; Roth, N.; Smith, D. Possible fatal interaction between protease inhibitors and methamphetamine. Antivir. Ther. 2000, 5, 19. [Google Scholar] [CrossRef] [PubMed]
  79. Henry, J.A.; Hill, I.R. Fatal interaction between ritonavir and MDMA. Lancet 1998, 352, 1751–1752. [Google Scholar] [CrossRef] [PubMed]
  80. De La Mora, L.; Nebot, M.J.; Martinez-Rebollar, M.; De Lazzari, E.; Tuset, M.; Laguno, M.; Ambrosioni, J.; Miquel, L.; Blanch, J.; Ugarte, A.; et al. Do ART and Chemsex Drugs Get Along? Potential Drug-Drug Interactions in a Cohort of People Living with HIV Who Engaged in Chemsex: A Retrospective Observational Study. Infect. Dis. Ther. 2022, 11, 2111–2124. [Google Scholar] [CrossRef]
  81. Cloarec, N.; Solas, C.; Ladaique, A.; Tamalet, C.; Zaegel-Faucher, O.; Bregigeon, S.; Canet, B.; Cano, C.E.; Poizot-Martin, I. Subtherapeutic darunavir con-centration and garlic consumption: A “Mediterranean” drug-food interaction, about 2 cases. Eur. J. Clin. Pharmacol. 2017, 73, 1331–1333. [Google Scholar] [CrossRef]
  82. Cordova, E.; Garibaldi, F.; Bono, L.; Rodriguez, C. Severe hyperkalaemia due to a potential drug-drug interaction between eplerenone and antiretrovirals in a HIV-positive patient after a myocardial infarction. Int. J. STD AIDS 2021, 32, 771–773. [Google Scholar] [CrossRef]
  83. Giguère, P.; Nhean, S.; Tseng, A.L.; Hughes, C.A.; Angel, J.B. Getting to the Heart of the Matter: A Review of Drug Interactions Between HIV Antiretrovirals and Cardiology Medications. Can. J. Cardiol. 2019, 35, 326–340. [Google Scholar] [CrossRef]
  84. Tyrberg, E.; Edén, A.; Eriksen, J.; Nilsson, S.; Treutiger, C.J.; Thalme, A.; Mellgren, Å.; Gisslén, M.; Andersson, L.M. Higher plasma drug levels in elderly people living with HIV treated with darunavir. PLoS ONE 2021, 16, e0246171. [Google Scholar] [CrossRef] [PubMed]
  85. Geletko, S.M.; Erickson, A.D. Decreased methadone effect after ritonavir initiation. Pharmacotherapy 2000, 20, 93–94. [Google Scholar] [CrossRef]
  86. Lozano, R.; Domeque, N.; Frutos, A.J. Methadone dosing in patients on ritonavir-boosted-based highly active antiretroviral therapy. Med. Clin. 2019, 152, 161–162. [Google Scholar] [CrossRef]
  87. Bukkems, V.E.; Colbers, A.; Marzolini, C.; Molto, J.; Burger, D.M. Drug-Drug Interactions with Antiretroviral Drugs in Pregnant Women Living with HIV: Are They Different from Non-Pregnant Individuals? Clin. Pharmacokinet. 2020, 59, 1217–1236. [Google Scholar] [CrossRef]
  88. Cowdell, I.; Beck, K.; Portwood, C.; Sexton, H.; Kumarendran, M.; Brandon, Z.; Kirtley, S.; Hemelaar, J. Adverse perinatal outcomes associated with protease inhibitor-based antiretroviral therapy in pregnant women living with HIV: A systematic review and meta-analysis. eClinicalMedicine 2022, 46, 101368. [Google Scholar] [CrossRef] [PubMed]
  89. Jimenez, H.R.; Natali, K.M.; Zahran, A.A.R. Drug interaction after ritonavir discontinuation: Considerations for antiretroviral therapy changes in renal trans-plant recipients. Int. J. STD AIDS 2019, 30, 710–714. [Google Scholar] [CrossRef] [PubMed]
  90. Cooper, M.; Dunne, I.; Kuten, S.; Curtis, A.; Graviss, E.A.; Nguyen, D.T.; Hobeika, M.; Gaber, A.O. Impact of Protease Inhibitor-Based Antiretroviral Therapy on Tacrolimus Intrapatient Variability in HIV-Positive Kidney Transplant Recipients. Transpl. Proc. 2021, 53, 984–988. [Google Scholar] [CrossRef] [PubMed]
  91. Cope, R.J.; Fischetti, B.S.; Kavanagh, R.K.; Lepa, T.M.; Sorbera, M.A. Safety and Efficacy of Weight-Loss Pharmacotherapy in Persons Living with HIV: A Review of the Literature and Potential Drug-Drug Interactions with Antiretroviral Therapy. Pharmacotherapy 2019, 39, 1204–1215. [Google Scholar] [CrossRef]
  92. Gervasoni, C.; Cattaneo, D.; Di Cristo, V.; Castoldi, S.; Gervasi, E.; Clementi, E.; Riva, A. Orlistat: Weight lost at cost of HIV rebound. J. Antimicrob. Chemother. 2016, 71, 1739–1741. [Google Scholar] [CrossRef]
  93. Kent, S.J. Loss of control of HIV viremia associated with the fat malabsorption drug orlistat. AIDS Res. Hum. Retrov. 2012, 28, 961–962. [Google Scholar] [CrossRef]
  94. Mathias, A.A.; German, P.; Murray, B.P.; Wei, L.; Jain, A.; West, S.; Warren, D.; Hui, J.; Kearney, B.P. Pharmacokinetics and pharmacodynamics of GS-9350: A novel pharmacokinetic enhancer without anti-HIV activity. Clin. Pharmacol. Ther. 2010, 87, 322–329. [Google Scholar] [CrossRef]
  95. Sinxadi, P.Z.; Khoo, S.H.; Boffito, M. Pharmacokinetic interactions of modern antiretroviral therapy. AIDS 2021, 35 (Suppl. S2), S145–S151. [Google Scholar] [CrossRef]
  96. Kiser, J.J.; Bumpass, J.B.; Meditz, A.L.; Anderson, P.L.; Bushman, L.; Ray, M.; Predhomme, J.A.; Rower, J.; Mawhinney, S.; Brundage, R. Effect of antacids on the pharmacokinetics of raltegravir in human immunodeficiency virus-seronegative volunteers. Antimicrob. Agents Chemother. 2010, 54, 4999–5003. [Google Scholar] [CrossRef]
  97. Ramanathan, S.; Mathias, A.; Wei, X.; Shen, G.; Koziara, J.; Cheng, A.; Kearney, B.P. Pharmacokinetics of once-daily boosted elvitegravir when administered in combination with acid-reducing agents. J. Acquir. Immune Defic. Syndr. 2013, 64, 45–50. [Google Scholar] [CrossRef]
  98. Song, I.; Borland, J.; Arya, N.; Wynne, B.; Piscitelli, S. Pharmacokinetics of dolutegravir when administered with mineral supplements in healthy adult subjects. J. Clin. Pharmacol. 2015, 55, 490–496. [Google Scholar] [CrossRef] [PubMed]
  99. Mathias, A.; Lutz, J.; West, S.; Xiao, D.; Chuck, S.; Martin, H.; Quirk, E.; Kearney, B. Pharmacokinetics (PK) of bictegravir (BIC) in combination with polyvalent cation containing (PVCC) antacids and supplements. HIV Med. 2019, 20, S5. [Google Scholar]
  100. Krishna, R.; East, L.; Larson, P.; Valiathan, C.; Butterfield, K.; Teng, Y.; Hernandez-Illas, M. Effect of metal-cation antacids on the pharmacokinetics of 1200 mg raltegravir. J. Pharm. Pharmacol. 2016, 68, 1359–1365. [Google Scholar] [CrossRef]
  101. Kang-Birken, S.L.; El-Sayed, D.; Prichard, J. HIV Viral Rebound Due to a Possible Drug-Drug Interaction between Elvitegravir/Cobicistat/Emtricitabine/Tenofovir Alafenamide and Calcium-Containing Products: Report of 2 Cases. J. Int. Assoc. Provid. AIDS Care 2019, 18, 1–4. [Google Scholar] [CrossRef] [PubMed]
  102. Dougherty, J.A.; Wynn, R.E.; Maarsingh, H. Unrecognized Drug Interaction: Ranolazine Adverse Effects Intensified by Darunavir-Cobicistat. Ann. Pharmacother. 2019, 53, 966–967. [Google Scholar] [CrossRef] [PubMed]
  103. Cattaneo, D.; Giacomelli, A.; Pagani, G.; Filice, C.; Gervasoni, C. Ritonavir/Cobicistat-Induc. Cushing Syndr. HIV Patients Treat. Non-Oral Corticosteroids: A Call Action? Am. J. Med. Sci. 2021, 361, 137–139. [Google Scholar] [CrossRef] [PubMed]
  104. Peyro-Saint-Paul, L.; Besnier, P.; Demessine, L.; Biour, M.; Hillaire-Buys, D.; de Canecaude, C.; Fedrizzi, S.; Parienti, J.J. Cushing’s syndrome due to interaction be-tween ritonavir or cobicistat and corticosteroids: A case-control study in the French Pharmacovigilance Database. J. Antimicrob. Chemother. 2019, 74, 3291–3294. [Google Scholar] [CrossRef] [PubMed]
  105. Seymour, N.; Robinson, M.; Richardson, D.; Mohammed, H.; Williams, D.; McGilligan, J.A. Prescribing intranasal steroids in HIV-positive patients: Systematic review of the literature. J. Laryngol. Otol. 2021, 135, 755–758. [Google Scholar] [CrossRef]
  106. Monge, E.; Colombo, V.; Giacomelli, A. Iatrogenic Cushing syndrome due to drug interaction be-tween inhaled fluticasone and cobicistat. Infez. Med. 2019, 27, 445. [Google Scholar]
  107. Gómez, Á.S.; García, M.J.A.; María Rafael Saponi Cortés, J.; Ruiz, C.M. Cushing’s syndrome secondary to inhaled fluticasone. Endocrinol. Diabetes Nutr. 2022, 69, 442–444. [Google Scholar] [CrossRef]
  108. Burger, D.M.; Calmy, A.; Marzolini, C. Cobicistat: A Case Mislabelled Drug-Drug Interactive Risk? Br. J. Clin. Pharmacol. 2020, 86, 834–836. [Google Scholar] [CrossRef]
  109. Lewis, J.; Turtle, L.; Khoo, S.; Nsutebu, E.N. A case of iatrogenic adrenal suppression after coadministration of cobicistat and fluticasone nasal drops. AIDS 2014, 28, 2636–2637. [Google Scholar] [CrossRef]
  110. Elliot, E.R.; Theodoraki, A.; Jain, L.R.; Marshall, N.J.; Boffito, M.; Baldeweg, S.E.; Waters, L.J. Iatrogenic Cushing’s syndrome due to drug interaction between glucocorticoids and the ritonavir or cobicistat containing HIV therapies. Clin. Med. 2016, 16, 412–418. [Google Scholar] [CrossRef]
  111. Wassner, C.; Maiti, S.; Kodroff, K.; Cohen, H. Iatrogenic Adrenal Insufficiency Secondary to Combination Therapy with Elvitegravir/Cobicistat/Tenofovir Disoproxil Fumarate/Emtricitabine and Interlaminar Triamcinolone Injection in an AIDS Patient. J. Int. Assoc. Provid. AIDS Care 2017, 16, 535–539. [Google Scholar] [CrossRef] [PubMed]
  112. Alidoost, M.; Conte, G.A.; Agarwal, K.; Carson, M.P.; Lann, D.; Marchesani, D. Iatrogenic Cushing’s Syndrome Fol-low-ing Intra-Articular Triamcinolone Injection in an HIV-Infected Patient on Cobicistat Presenting as a Pulmonary Embolism: Case Report and Literature Review. Int. Med. Case Rep. J. 2020, 13, 229–235. [Google Scholar] [CrossRef] [PubMed]
  113. Mathias, P.M.; Epstein, E.J. Iatrogenic Cushing syndrome in an HIV-infected patient secondary to concomitant therapy with Genvoya and epidural triamcinolone. AACE Clin. Case Rep. 2020, 6, e217–e220. [Google Scholar] [CrossRef] [PubMed]
  114. Makaram, N.; Russell, C.D.; Roberts, S.B.; Stevens, J.; Macpherson, G. Exogenous steroid-induced hypoadrenalism in a person living with HIV caused by a drug-drug interaction between cobicistat and intrabursal triamcinolone. BMJ Case Rep. 2018, 11, e226912. [Google Scholar] [CrossRef]
  115. Rosales-Castillo, A.; López-Ruz, M.Á. Cushing’s syndrome due to interaction between topical betamethasone dipropionate and darunavir/cobicistat. Med. Clin. 2020, 155, 466–467. [Google Scholar] [CrossRef]
  116. Sang, N.; Pastakia, S.; Nyanje, S. International Normalized Ratio (INR) Increases amongst Two Patients Living with HIV on Warfarin after Being Switched from a Nevirapine to a Dolutegravir-Based Antiretroviral Regimen. Case Rep. Infect. Dis. 2021, 29, 9384663. [Google Scholar] [CrossRef]
  117. Good, B.L.; Gomes, D.C.; Fulco, P.P. An unexpected interaction between warfarin and cobicistat-boosted elvitegravir. AIDS 2015, 29, 985–986. [Google Scholar] [CrossRef]
  118. Malagnino, V.; Cerva, C.; Teti, E.; Maffongelli, G.; Andreoni, M.; Sarmati, L. Abnormal elevation of international normalized ratio in a patient during the coadministration of elvitegravir/cobicistat/emtricitabine/tenofovir alafenamide and warfarin: A case report. AIDS 2019, 33, 360–361. [Google Scholar] [CrossRef] [PubMed]
  119. Tseng, A.; Hughes, C.A.; Wu, J.; Seet, J.; Phillips, E.J. Cobicistat Versus Ritonavir: Similar Pharmacokinetic Enhancers but Some Important Differences. Ann. Pharmacother. 2017, 51, 1008–1022. [Google Scholar] [CrossRef] [PubMed]
  120. Tseng, A.L.; Luetkehoelter, J.; Walmsley, S.L. Increase in international normalized ratio after switching from atazanavir/ritonavir to darunavir/cobicistat in a patient on warfarin: Boosters are not always equal. AIDS 2017, 31, 175–176. [Google Scholar] [CrossRef]
  121. Hikasa, S.; Sawada, A.; Seino, H.; Shimabukuro, S.; Hideta, K.; Uwa, N.; Higasa, S.; Tokugawa, T.; Kimura, T. A potential drug interaction between phenobarbital and dolutegravir: A case report. J. Infect. Chemother. 2018, 24, 476–478. [Google Scholar] [CrossRef]
  122. Tseng, A.L.; Wong, A.Y.J.; McLelland, C.J.; Walmsley, S.L. Drug interactions are not always predictable: The curious case of valproic acid and dolutegravir and a possible explanation. AIDS 2019, 33, 1677–1679. [Google Scholar] [CrossRef] [PubMed]
  123. Palazzo, A.; Trunfio, M.; Pirriatore, V.; Milesi, M.; De Nicolò, A.; Alcantarini, C.; D’Avolio, A.; Bonora, S.; Di Perri, G.; Calcagno, A. Lower dolutegravir plasma con-centrations in HIV-positive patients receiving valproic acid. J. Antimicrob. Chemother. 2018, 73, 826–827. [Google Scholar] [CrossRef]
  124. Bollen, P.D.J.; Prins, H.A.B.; Colbers, A.; Velthoven-Graafland, K.; Rijnders, B.J.A.; de Vries-Sluijs, T.; van Nood, E.; Nouwen, J.; Bax, H.; de Mendonca Melo, M.; et al. The dolutegravir/valproic acid drug-drug interaction is primarily based on protein displacement. J. Antimicrob. Chemother. 2021, 76, 1273–1276. [Google Scholar] [CrossRef]
  125. Calcagno, A.; Cusato, J.; Ferrara, M.; De Nicolò, A.; Lazzaro, A.; Manca, A.; D’Avolio, A.; Di Perri, G.; Bonora, S. Antiretroviral concentrations in the presence and absence of valproic acid. J. Antimicrob. Chemother. 2020, 75, 1969–1971. [Google Scholar] [CrossRef]
  126. Müller, F.; König, J.; Hoier, E.; Mandery, K.; Fromm, M.F. Role of organic cation transporter OCT2 and multidrug and toxin extrusion proteins MATE1 and MATE2-K for transport and drug interactions of the antiviral lamivudine. Biochem. Pharmacol. 2013, 86, 808–815. [Google Scholar] [CrossRef]
  127. Naidoo, A.; Naidoo, K.; Padayatchi, N.; Dooley, K.E. Use of integrase inhibitors in HIV-associated tuberculosis in high-burden settings: Implementation challenges and research gaps. Lancet HIV 2022, 9, e130–e138. [Google Scholar] [CrossRef]
  128. Custodio, J.M.; West, S.K.; Vu, A.; Xiao, D.; Collins, S.; Martin, H.; Quirk, E.; Kearney, B.P.; Mathias, A. Pharmacokinetics of bictegravir administered twice daily in combination with rifampin. In Proceedings of the 2018 Conference on Retroviruses and Opportunistic Infections, Boston, MA, USA, 4–7 March 2018. Abstract 34. [Google Scholar]
  129. Rockwood, N.; Cerrone, M.; Barber, M.; Hill, A.M.; Pozniak, A.L. Global access of rifabutin for the treatment of tuberculosis—Why should we prioritize this? J. Int. AIDS Soc. 2019, 22, e25333. [Google Scholar] [CrossRef] [PubMed]
  130. Ford, S.L.; Sutton, K.; Lou, Y.; Zhang, Z.; Tenorio, A.; Trezza, C.; Patel, P.; Spreen, W. Effect of Rifampin on the Single-Dose Pharmaco-kinetics of Oral Cabotegravir in Healthy Subjects. Antimicrob. Agents Chemother. 2017, 61, e00487-17. [Google Scholar] [CrossRef] [PubMed]
  131. Clement, M.E.; Kofron, R.; Landovitz, R.J. Long-acting injectable cabotegravir for the prevention of HIV infection. Curr. Opin. HIV AIDS 2020, 15, 19–26. [Google Scholar] [CrossRef]
  132. Hostench-Junoy, N.; Ramírez-Montoya, M.; Arefai-Refai, B.; Estal-Jiménez, J.; Santana-Rodríguez, Z.J.; Costa-Pérez, L. Acute Ischemia of Lower Extremities Caused by Ergotamine Toxicity due to Pharmacologic Interaction with Cobicistat in an HIV-Positive Patient. Ann. Vasc. Surg. 2022, 80, 392.e1–392.e6. [Google Scholar] [CrossRef]
  133. Navarro, J.; Curran, A.; Burgos, J.; Torrella, A.; Ocaña, I.; Falcó, V.; Crespo, M.; Ribera, E. Acute leg ischaemia in an HIV-infected patient receiving antiretroviral treatment. Antivir. Ther. 2017, 22, 89–90. [Google Scholar] [CrossRef] [PubMed]
  134. Charakhanian, S.; De Sabb, R.; Vaseghi, M.; Cardon, B.; Rozenbaum, W. Evaluation of the association of zidovudine (Z) and vinblastine (V) in treatment of AIDS-related Kaposi’s sarcoma (KS). In Proceedings of the 5th International Conference on AIDS, Montreal, QC, Canada, 4–6 June 1989. Abstract MBP368. [Google Scholar]
  135. Bidon, D.; Bauler, S.; Venon, M.D.; Dupont, C. Cobicistat-vinblastine interaction and severe peripheral neuropathy. AIDS 2015, 29, 1120–1121. [Google Scholar] [CrossRef] [PubMed]
  136. Majeed, S.R.; West, S.; Ling, K.H.; Das, M.; Kearney, B.P. Confirmation of the drug-drug interaction potential between cobicistat-boosted antiretroviral regimens and hormonal contraceptives. Antivir. Ther. 2019, 24, 557–566. [Google Scholar] [CrossRef] [PubMed]
  137. Ammar, H.; Govindu, R.R. A Dangerous and Unrecognized Interaction of Apixaban. Cureus 2021, 13, e19688. [Google Scholar] [CrossRef]
  138. Gordon, L.A.; Kumar, P.; Brooks, K.M.; Kellogg, A.; McManus, M.; Alfaro, R.M.; Nghiem, K.; George, J.M.; Lozier, J.; Penzak, S.R.; et al. Antiretroviral Boosting Agent Cobicistat Increases the Pharmacokinetic Exposure and Anticoagulant Effect of Dabigatran in HIV-Negative Healthy Volunteers. Circulation 2016, 134, 1909–1911. [Google Scholar] [CrossRef]
  139. Kumar, P.; Gordon, L.A.; Brooks, K.M.; George, J.M.; Kellogg, A.; McManus, M.; Alfaro, R.M.; Nghiem, K.; Lozier, J.; Hadigan, C.; et al. Differential Influence of the Antiretroviral Pharmacokinetic Enhancers Ritonavir and Cobicistat on Intestinal P-Glycoprotein Transport and the Pharmacokinetic/Pharmacodynamic Disposition of Dabigatran. Antimicrob. Agents Chemother. 2017, 61, e01201-17. [Google Scholar] [CrossRef]
  140. Lingineni, K.; Farhan, N.; Kim, S.; Cristo-foletti, R.; Gordon, L.A.; Kumar, P.; Penzak, S.; Hadigan, C.; George, J.M.; Brown, J.D.; et al. Quantitative Benefit-Risk Assessment of P-gp-Mediated Drug-Drug Interactions of Dabigatran Coadministered with Pharmacokinetic Enhancers in Patients with Renal Impairment. Clin. Pharmacol. Ther. 2021, 109, 193–200. [Google Scholar] [CrossRef] [PubMed]
  141. Yoong, D.; Naccarato, M.; Gough, K. Extensive Bruising and Elevated Rivaroxaban Plasma Concentration in a Patient Receiving Cobicistat-Boosted Elvitegravir. Ann. Pharmacol. 2017, 51, 713–714. [Google Scholar] [CrossRef] [PubMed]
  142. Desai, N.; Burns, L.; Gong, Y.; Zhi, K.; Kumar, A.; Summers, N.; Kumar, S.; Cory, T.J. An update on drug-drug interactions between antiretroviral therapies and drugs of abuse in HIV systems. Expert Opin. Drug Metab. Toxicol. 2020, 16, 1005–1018. [Google Scholar] [CrossRef] [PubMed]
  143. Bracchi, M.; Stuart, D.; Castles, R.; Khoo, S.; Back, D.; Boffito, M. Increasing use of «party drugs» in people living with HIV on antiretrovirals: A concern for patient safety. AIDS 2015, 29, 1585–1592. [Google Scholar] [CrossRef] [PubMed]
  144. Hiransuthikul, A.; Janamnuaysook, R.; Himmad, K.; Kerr, S.J.; Thammajaruk, N.; Pankam, T.; Phanjaroen, K.; Mills, S.; Vannakit, R.; Phanuphak, P.; et al. Drug-drug interactions between feminizing hormone therapy and pre-exposure prophylaxis among transgender women: The iFACT study. J. Int. AIDS Soc. 2019, 22, e25338. [Google Scholar] [CrossRef]
  145. Shieh, E.; Marzinke, M.A.; Fuchs, E.J.; Hamlin, A.; Bakshi, R.; Aung, W.; Breakey, J.; Poteat, T.; Brown, T.; Bumpus, N.N.; et al. Transgender women on oral HIV pre-exposure prophylaxis have significantly lower tenofovir and emtricitabine concentrations when also taking estrogen when com-pared to cisgender men. J. Int. AIDS Soc. 2019, 22, e25405. [Google Scholar] [CrossRef]
  146. Badowski, M.E.; Britt, N.; Huesgen, E.C.; Lewis, M.M.; Miller, M.M.; Nowak, K.; Sherman, E.; Smith, R.O. Pharmacotherapy considerations in transgender individuals living with human immunodeficiency virus. Pharmacotherapy 2021, 41, 299–314. [Google Scholar] [CrossRef]
  147. Cattani, V.B.; Jalil, E.M.; Eksterman, L.; Torres, T.; Cardoso, S.W.; Castro, C.R.V.; Monteiro, L.; Wilson, E.; Bushman, L.; Anderson, P.; et al. Impact of feminizing hormone therapy on tenofovir and emtricitabine plasma pharmacokinetics: A nested drug-drug interaction study in a cohort of Brazilian transgender women using HIV pre-exposure prophylaxis. J. Antimicrob. Chemother. 2022, 77, 2729–2736. [Google Scholar] [CrossRef]
  148. Yager, J.L.; Anderson, P.L. Pharmacology and drug interactions with HIV PrEP in transgender persons receiving gender affirming hormone therapy. Expert Opin. Drug Metab. Toxicol. 2020, 16, 463–474. [Google Scholar] [CrossRef]
  149. Yager, J.; Brooks, K.M.; Brothers, J.; Mulligan, K.; Landovitz, R.; Reirden, D.; Glenny, C.; Malhotra, M.; Anderson, P.L.; Hosek, S. Pharmacokinet-ics of Emtricitabine/Tenofovir Disoproxil Fumarate Among Transgender Adolescents and Young Adults Without HIV Receiving Gender Affirming Hormones. AIDS Res. Hum. Retroviruses 2022, 38, 840–846. [Google Scholar] [CrossRef]
  150. Cottrell, M.L.; Prince, H.M.A.; Schauer, A.P.; Sykes, C.; Maffuid, K.; Poliseno, A.; Chun, T.W.; Huiting, E.; Stanczyk, F.Z.; Peery, A.F.; et al. Decreased Tenofovir Diphosphate Concentrations in a Transgender Female Cohort: Implications for Human Immunodeficiency Virus Preexposure Prophylaxis. Clin. Infect. Dis. 2019, 69, 2201–2204. [Google Scholar] [CrossRef] [PubMed]
  151. Tanaudommongkon, A.; Chaturvedula, A.; Hendrix, C.W.; Fuchs, E.J.; Shieh, E.; Bakshi, R.P.; Marzinke, M.A. Population pharmacokinetics of tenofovir, emtricitabine and intracellular metabolites in transgender women. Br. J. Clin. Pharmacol. 2022, 88, 3674–3682. [Google Scholar] [CrossRef] [PubMed]
  152. Grant, R.M.; Pellegrini, M.; Defechereux, P.A.; Anderson, P.L.; Yu, M.; Glidden, D.V.; O’Neal, J.; Yager, J.; Bhasin, S.; Sevelius, J.; et al. Sex Hormone Therapy and Tenofovir Diphosphate Concentration in Dried Blood Spots: Primary Results of the Interactions Between Antiretrovirals and Transgender Hormones Study. Clin. Infect. Dis. 2021, 73, e2117–e2123. [Google Scholar] [CrossRef] [PubMed]
  153. Cattaneo, D.; Ridolfo, A.; Baldelli, S.; Gervasoni, C. Effects of guggulsterones-containing thermogenic com-plex on elvitegravir plasma concentrations: A case report. Eur. J. Clin. Pharmacol. 2019, 75, 1177–1178. [Google Scholar] [CrossRef] [PubMed]
  154. Adkison, K.; Wolstenholme, A.; Lou, Y.; Zhang, Z.; Eld, A.; Perger, T.; Vangerow, H.; Hayward, K.; Shaefer, M.; McCoig, C. Effect of Sorbitol on the Pharmacokinetic Profile of Lamivudine Oral Solution in Adults: An Open-Label, Randomized Study. Clin. Pharmacol. Ther. 2018, 103, 402–408. [Google Scholar] [CrossRef] [PubMed]
  155. Pecora Fulco, P.; Patel, B. Sildenafil Use for Pulmonary Artery Hypertension with a Cobicistat-Boosted Antiretroviral Regimen. Ann. Pharmacother. 2020, 54, 84–85. [Google Scholar] [CrossRef] [PubMed]
  156. Bukkems, V.E.; Hidalgo-Tenorio, C.; Garcia, C.; van Hulzen, A.G.W.; Richel, O.; Burger, D.M.; Colbers, A.P.; Pharmacokinetics of ANtiretroviral agents in HIV-infected pregNAnt women (PANNA) network. First pharmacokinetic data of bictegravir in pregnant women living with HIV. AIDS 2021, 35, 2405–2406. [Google Scholar] [CrossRef]
  157. Scarsi, K.K.; Havens, J.P.; Podany, A.T.; Avedissian, S.N.; Fletcher, C.V. HIV-1 Integrase Inhibitors: A Comparative Re-view of Efficacy and Safety. Drugs 2020, 80, 1649–1676. [Google Scholar] [CrossRef]
  158. Momper, J.D.; Best, B.M.; Wang, J.; Capparelli, E.V.; Stek, A.; Barr, E.; Badell, M.L.; Acosta, E.P.; Purswani, M.; Smith, E.; et al. Elvitegravir/cobicistat pharmacokinetics in pregnant and postpartum women with HIV. AIDS 2018, 32, 2507–2516. [Google Scholar] [CrossRef]
  159. Patel, P.; Ford, S.L.; Baker, M.; Meyer, C.; Garside, L.; D’Amico, R.; Van Solingen-Ristea, R.; Crauwels, H.; Polli, J.W.; Seal, C.; et al. Pregnancy outcomes and pharmacokinetics in pregnant women living with HIV exposed to long-acting cabotegravir and rilpivirine in clinical trials. Open Forum Infect. Dis. 2021, 8 (Suppl. S1), S534. [Google Scholar] [CrossRef]
  160. Bukkems, V.E.; Smolders, E.J.; Jourdain, G.; Burger, D.M.; Colbers, A.P.; Cressey, T.R.; PANNA Network and iTAP Study Group. Effect of Pregnancy and Concomitant Antiretrovirals on the Pharmacokinetics of Tenofovir in Women with HIV Receiving Tenofovir Disoproxil Fumarate-Based Antiretroviral Therapy Versus Women with HBV Receiving Tenofovir Disoproxil Fumarate Monotherapy. J. Clin. Pharmacol. 2021, 61, 388–393. [Google Scholar] [CrossRef] [PubMed]
  161. Mulligan, N.; Salama, E.; Momper, J.D.; Capparelli, E.V.; Stek, A.; Chakhtoura, N.; Mirochnick, M.; Best, B.M.; IMPAACT P1026s Protocol Team. Lopinavir and tenofovir interaction observed in non-pregnant adults altered during pregnancy. J. Clin. Pharm. Ther. 2021, 46, 1459–1464. [Google Scholar] [CrossRef]
  162. Han, Z.; Kane, B.M.; Petty, L.A.; Josephson, M.A.; Sutor, J.; Pursell, K.J. Cobicistat Significantly Increases Tacrolimus Serum Concentrations in a Renal Transplant Recipient with Human Immunodeficiency Virus Infection. Pharmacotherapy 2016, 36, e50–e53. [Google Scholar] [CrossRef]
  163. Gutierrez, M.D.M.; Mateo, M.G.; Corbacho, N.; Vidal, F.; Domingo, P. Drug-drug interactions when treating HIV-related metabolic disorders. Expert Opin. Drug Metab. Toxicol. 2019, 15, 787–802. [Google Scholar] [CrossRef] [PubMed]
  164. Godinho, R.; Bugnon, S.; Gracin, T.; Tataw, J. Severe rhabdomyolysis-induced acute kidney injury following concomitant use of Genvoya® (EVG/COBI/FTC/TAF) and simvastatin; a case report. BMC Nephrol. 2019, 20, 69. [Google Scholar] [CrossRef] [PubMed]
  165. Suttels, V.; Florence, E.; Leys, J.; Vekemans, M.; Van den Ende, J.; Vlieghe, E.; Kenyon, C. A 68-year old male presenting with rhabdomyolysis-associated acute kidney injury following concomitant use of elvitegravir/cobicistat/emtricitabine/tenofovir disoproxil fumarate and pravastatin/fenofibrate: A case report. J. Med. Case Rep. 2015, 9, 190. [Google Scholar] [CrossRef]
  166. Custodio, J.; West, S.; SenGupta, D.; Zari, A.; Humeniuk, R.; Ling, K.H.J.; Das, M.; Kearney, B.P. Evaluation of the drug-drug interaction potential between cobicistat-boosted protease inhibitors and statins. In Proceedings of the 18th International Workshop on Clinical Pharmacology of Antiviral Therapy, Chicago, IL, USA, 14–16 June 2017. Abstract O_04. [Google Scholar]
  167. Liu, S.N.; Gufford, B.T.; Lu, J.B.L.; Bushman, L.R.; Anderson, P.L.; Bergstrom, R.F.; Desta, Z.; Gupta, S.K. Inhibitory Effects of Probenecid on Pharmaco-kinetics of Tenofovir Disoproxil Fumarate and Emtricitabine for On-Demand HIV Preexposure Prophylaxis. Clin. Pharmacol. Ther. 2020, 107, 1200–1208. [Google Scholar] [CrossRef]
  168. Reese, M.J.; Bowers, G.D.; Humphreys, J.E.; Gould, E.P.; Ford, S.L.; Webster, L.O.; Polli, J.W. Drug interaction profile of the HIV integrase inhibitor cabotegravir: Assessment from in vitro studies and a clinical investigation with midazolam. Xenobiotica 2016, 46, 445–456. [Google Scholar] [CrossRef]
  169. Ippolito, M.M.; Jacobson, J.M.; Lederman, M.M.; Winterberg, M.; Tarning, J.; Shapiro, T.A.; Flexner, C. Effect of Antiretroviral Therapy on Plasma Concentrations of Chloroquine and Desethyl-chloroquine. Clin. Infect. Dis. 2018, 67, 1617–1620. [Google Scholar] [CrossRef]
  170. Ewing, A.C.; King, C.C.; Wiener, J.B.; Chasela, C.S.; Hudgens, M.G.; Kamwendo, D.; Tegha, G.; Hosseinipour, M.C.; Jamieson, D.J.; Van der Horst, C.; et al. Effects of concurrent exposure to antiretrovirals and cotrimoxazole prophylaxis among HIV-exposed, uninfected infants. AIDS 2017, 31, 2455–2463. [Google Scholar] [CrossRef]
  171. Villani, P.; Viale, P.; Signorini, L.; Cadeo, B.; Mar-chetti, F.; Villani, A.; Fiocchi, C.; Regazzi, M.B.; Carosi, G. Pharmacokinetic evaluation of oral levofloxacin in human immunodeficiency virus-infected subjects receiving concomitant antiretroviral therapy. Antimicrob. Agents Chemother. 2001, 45, 2160–2162. [Google Scholar] [CrossRef]
  172. Van Der Laan, L.E.; Garcia-Prats, A.J.; Schaaf, H.S.; Tikiso, T.; Wiesner, L.; De Kock, M.; Winckler, J.; Norman, J.; McIlleron, H.; Denti, P.; et al. Pharmacokinetics and Drug-Drug Interactions of Lopinavir-Ritonavir Administered with First- and Second-Line Antituberculosis Drugs in HIV-Infected Children Treated for Multidrug-Resistant Tuberculosis. Antimicrob. Agents Chemother. 2018, 62, e00420-17. [Google Scholar] [CrossRef] [PubMed]
  173. Ramachandran, G.; Hemanth Kumar, A.K.; Kannan, T.; Sridhar, R.; Guha, S.K.; Kadam, D.; Poorana Gangadevi, N.; Rajapandian, T. Pharmacokinetics of rifabutin during atazanavir/ritonavir co-administration in HIV-infected TB patients. Indian J. Tuberc. 2019, 66, 129–133. [Google Scholar] [CrossRef] [PubMed]
  174. Ford, S.L.; Lou, Y.; Lewis, N.; Kostapanos, M.; D’Amico, R.; Spreen, W.; Patel, P. Effect of rifabutin on the pharmacokinetics of oral cabotegravir in healthy subjects. Antivir. Ther. 2019, 24, 301–308. [Google Scholar] [CrossRef] [PubMed]
  175. Dooley, K.; Kaplan, R.; Mwelase, T.; Grinsztejn, B.; Ticona, E.; Lacerda, M.; Cahn, P.; Belonosova, E.; Ait-Khaled, M.; Angelis, K.; et al. Safety and efficacy of dolutegravir-based art in TB + HIV coinfected adults at week 24. In Proceedings of the 25th Conference on Retroviruses and Opportunistic Infections (CROI), Boston, MA, USA, 4–7 March 2018; pp. 334–347. [Google Scholar]
  176. Dooley, K.E.; Kaplan, R.; Mwelase, N.; Grinsztejn, B.; Ticona, E.; Lacerda, M.; Sued, O.; Belonosova, E.; Ait-Khaled, M.; Angelis, K.; et al. Dolutegravir-based Antiretroviral Therapy for Patients Coinfected with Tuberculosis and Human Immunodeficiency Virus: A Multicenter, Noncomparative, Open-label, Randomized Trial. Clin. Infect Dis. 2020, 70, 549–555. [Google Scholar] [CrossRef]
  177. Havens, J.P.; Podany, A.T.; Scarsi, K.K.; Fletcher, C.V. Clinical Pharmacokinetics and Pharmacodynamics of Etravirine: An Updated Review. Clin. Pharmacokinet. 2020, 59, 137–154. [Google Scholar] [CrossRef]
  178. Thompson, M.; Lalezari, J.P.; Kaplan, R.; Pinedo, Y.; Pena, O.A.S.; Cahn, P.; Stock, D.A.; Joshi, S.R.; Hanna, G.J.; Lataillade, M.; et al. Safety and efficacy of the HIV-1 attachment inhibitor prodrug fostemsavir in antiretroviral-experienced subjects: Week 48 analysis of AI438011, a Phase IIb, randomized controlled trial. Antivir. Ther. 2017, 22, 215–223. [Google Scholar]
  179. Lataillade, M.; Lalezari, J.P.; Kozal, M.; Aberg, J.A.; Pialoux, G.; Cahn, P.; Thompson, M.; Molina, J.M.; Moreno, S.; Grinsztejn, B.; et al. Safety and efficacy of the HIV-1 attachment inhibitor prodrug fostemsavir in heavily treatment-experienced individuals: Week 96 results of the phase 3 BRIGHTE study. Lancet HIV 2020, 7, e740–e751. [Google Scholar] [CrossRef]
  180. Feng, H.P.; Guo, Z.; Ross, L.L.; Fraser, I.; Panebianco, D.; Jumes, P.; Fandozzi, C.; Caro, L.; Talaty, J.; Ma, J.; et al. Assessment of drug interaction potential between the HCV direct-acting antiviral agents elbasvir/grazoprevir and the HIV integrase inhibitors raltegravir and dolutegravir. J. Antimicrob. Chemother. 2019, 74, 710–717. [Google Scholar] [CrossRef]
  181. MacBrayne, C.E.; Castillo-Mancilla, J.; Burton, J.R., Jr.; MaWhinney, S.; Wagner, C.B.; Micke, K.; Fey, J.; Huntley, R.T.; Larson, B.; Bushman, L.R.; et al. Small increase in dolutegravir trough, but equivalent total dolutegravir exposure with simeprevir in HIV/HCV seronegative volunteers. J. Antimicrob. Chemother. 2018, 73, 156–159. [Google Scholar] [CrossRef]
  182. Custodio, J.M.; Chuck, S.K.; Chu, H.; Cao, H.; Ma, G.; Flaherty, J.; Ling, J.; Kearney, B.P. Lack of clinically important PK interaction between coformulated ledipasvir/sofosbuvir and rilpivirine/emtricitabine/tenofovir alafenamide. Pharmacol. Res. Perspect. 2017, 5, e00353. [Google Scholar] [CrossRef] [PubMed]
  183. Podany, A.T.; Scarsi, K.K.; Pham, M.M.; Fletcher, C.V. Comparative Clinical Pharmacokinetics and Pharmacodynamics of HIV-1 Integrase Strand Transfer Inhibitors: An Updated Review. Clin. Pharmacokinet. 2020, 59, 1085–1107. [Google Scholar] [CrossRef] [PubMed]
  184. Zhang, H.; Shao, Y.; Garner, W.; Vu, A.; Martin, H.; SenGupta, D.; Quirk, E.; Kearney, B.P.; Custodio, J.M. The effect of hepatic or renal impairment on bictegravir pharmacokinetics. In Proceedings of the 18th International Workshop on Clinical Pharmacology of Antiviral Therapy, Chicago, IL, USA, 14–16 June 2017. [Google Scholar]
  185. Shaik, J.S.B.; Ford, S.L.; Lou, Y.; Zhang, Z.; Bakshi, K.K.; Tenorio, A.R.; Trezza, C.; Spreen, W.R.; Patel, P. A phase 1 study to evaluate the pharmacokinetics and safety of cabotegravir in patients with hepatic impairment and healthy matched controls. Clin. Pharmacol. Drug Dev. 2019, 8, 664–673. [Google Scholar] [CrossRef]
  186. Song, I.H.; Borland, J.; Savina, P.M.; Chen, S.; Patel, P.; Wajima, T.; Peppercorn, A.F.; Piscitelli, S.C. Pharmacokinetics of single-dose dolutegravir in HIV seronegative subjects with moderate hepatic impairment compared to healthy matched controls. Clin. Pharmacol. Drug Dev. 2013, 2, 342–348. [Google Scholar] [CrossRef]
  187. Custodio, J.M.; Rhee, M.; Shen, G.; Ling, K.H.; Kearney, B.P.; Ramanathan, S. Pharmacokinetics and safety of boosted elvitegravir in subjects with hepatic impairment. Antimicrob. Agents Chemother. 2014, 58, 2564–2569. [Google Scholar] [CrossRef] [PubMed]
  188. Iwamoto, M.; Hanley, W.D.; Petry, A.S.; Friedman, E.J.; Kost, J.T.; Breidinger, S.A.; Lasseter, K.C.; Robson, R.; Lunde, N.M.; Wenning, L.A.; et al. Lack of a clinically important effect of moderate hepatic insufficiency and severe renal insufficiency on raltegravir pharmacokinetics. Antmicrob. Agents Chemother. 2009, 53, 1747–1755. [Google Scholar] [CrossRef]
  189. Parasrampuria, R.; Ford, S.L.; Lou, Y.; Fu, C.; Bakshi, K.K.; Tenorio, A.R.; Trezza, C.; Spreen, W.R.; Patel, P. A phase I study to evaluate the pharmacokinetics and safety of cabotegravir in adults with severe renal impairment and healthy matched control participants. Clin. Pharmacol. Drug Dev. 2019, 8, 674–681. [Google Scholar] [CrossRef]
  190. Weller, S.; Borland, J.; Chen, S.; Johnson, M.; Savina, P.; Wynne, B.; Wajima, T.; Peppercorn, A.F.; Piscitelli, S.C. Pharmacokinetics of dolutegravir in HIV-seronegative subjects with severe renal impairment. Eur. J. Clin. Pharmacol. 2014, 70, 29–35. [Google Scholar] [CrossRef]
  191. German, P.; Wei, X.; Mizuno, V.; Cheng, A.; Kearney, B.; Mathias, A. (Eds.) Pharmacokinetics of elvitegravir and cobicistat in subjects with severe renal impairment. In Proceedings of the 13th International Workshop on Clinical Pharmacology of HIV Therapy, Barcelona, Spain, 16–18 April 2012. [Google Scholar]
  192. Eron, J.J., Jr.; Lelievre, J.D.; Kalayjian, R.; Slim, J.; Wurapa, A.K.; Stephens, J.L.; McDonald, C.; Cua, E.; Wilkin, A.; Schmied, B.; et al. Safety of elvitegravir, cobicistat, emtricitabine, and tenofovir alafenamide in HIV-1-infected adults with end-stage renal disease on chronic haemodialysis: An open-label, single-arm, multicentre, phase 3b trial. Lancet HIV 2019, 6, e15–e24. [Google Scholar] [CrossRef]
  193. Siritientong, T.; Thet, D.; Methaneethorn, J.; Leelakanok, N. Pharmacokinetic Outcomes of the Interactions of Antiretroviral Agents with Food and Supplements: A Systematic Review and Meta-Analysis. Nutrients 2022, 14, 520. [Google Scholar] [CrossRef]
  194. Yamada, H.; Ikushima, I.; Nemoto, T.; Ishikawa, T.; Ninomiya, N.; Irie, S. Effects of a Nutritional Protein-Rich Drink on the Pharmacokinetics of Elvitegravir, Cobicistat, Emtricitabine, Tenofovir Alafenamide, and Tenofovir Compared with a Standard Meal in Healthy Japanese Male Subjects. Clin. Pharmacol. Drug Dev. 2018, 7, 132–142. [Google Scholar] [CrossRef]
  195. Yonemura, T.; Okada, N.; Sagane, K.; Okamiya, K.; Ozaki, H.; Iida, T.; Yamada, H.; Yagura, H. Effects of Milk or Apple Juice Ingestion on the Pharmacokinetics of Elvitegravir and Cobicistat in Healthy Japanese Male Volunteers: A Randomized, Single-Dose, Three-Way Crossover Study. Clin. Pharmacol. Drug Dev. 2018, 7, 737–743. [Google Scholar] [CrossRef] [PubMed]
  196. Thurman, A.R.; Schwartz, J.L.; Brache, V.; Chen, B.A.; Chandra, N.; Kashuba, A.D.M.; Weiner, D.H.; Mauck, C.; Doncel, G.F. Effect of Hormonal Contraception on Pharmacokinetics of Vaginal Tenofovir in Healthy Women: Increased Tenofovir Diphosphate in Injectable Depot Medroxyprogesterone Acetate Users. J. Acquir. Immune. Defic. Syndr. 2019, 80, 79–88. [Google Scholar] [CrossRef] [PubMed]
  197. Ford, S.L.; Gould, E.; Chen, S.; Lou, Y.; Dumont, E.; Spreen, W.; Piscitelli, S. Effects of etravirine on the pharmacokinetics of the integrase inhibitor S/GSK1265744. Antimicrob. Agents Chemother. 2013, 57, 277–280. [Google Scholar] [CrossRef] [PubMed]
  198. Anderson, M.S.; Kakuda, T.N.; Hanley, W.; Miller, J.; Kost, J.T.; Stoltz, R.; Wenning, L.A.; Stone, J.A.; Hoetelmans, R.M.; Wagner, J.A.; et al. Minimal pharmacokinetic interaction between the human immunodeficiency virus nonnucleoside reverse transcriptase inhibitor etravirine and the integrase inhibitor raltegravir in healthy subjects. Antimicrob. Agents Chemother. 2008, 52, 4228–4232. [Google Scholar] [CrossRef] [PubMed]
  199. Lê, M.P.; Valantin, M.A.; Assoumou, L.; Soulie, C.; Le Mestre, S.; Weiss, L.; Yazdanpanah, Y.; Molina, J.M.; Bouchaud, O.; Raffi, F.; et al. ANRS-163 ETRAL study group. Lack of a Clinically Significant Pharmacokinetic Interaction between Etravirine and Raltegravir Using an Original Approach Based on Drug Metabolism, Protein Binding, and Penetration in Seminal Fluid: A Pharmacokinetic Substudy of the ANRS-163 ETRAL Study. Pharmacotherapy 2019, 39, 514–520. [Google Scholar]
  200. Trezza, C.; Ford, S.L.; Gould, E.; Lou, Y.; Huang, C.; Ritter, J.M.; Buchanan, A.M.; Spreen, W.; Patel, P. Lack of effect of oral cabotegravir on the pharmacokinetics of a levonorgestrel/ethinyl oestradiol-containing oral contraceptive in healthy adult women: Effect of cabotegravir on combined oral contraceptives. Br. J. Clin. Pharmacol. 2017, 83, 1499–1505. [Google Scholar] [CrossRef]
  201. Womack, J.A.; Novick, G.; Goulet, J.L. Hormonal contraceptive use in HIV-infected women using antiretroviral therapy: A Systematic review. Open Access J. Contracept. 2015, 6, 37–52. [Google Scholar] [CrossRef] [PubMed]
  202. Kearney, B.P.; Mathias, A. Lack of effect of tenofovir disoproxil fumarate on pharmacokinetics of hormonal contraceptives. Pharmacotherapy 2009, 29, 924–929. [Google Scholar] [CrossRef]
  203. Moore, K.; Magee, M.; Sevinsky, H.; Chang, M.; Lubin, S.; Myers, E.; Ackerman, P.; Llamoso, C. Methadone and buprenorphine pharmacokinetics and pharmacodynamics when coadministered with fostemsavir to opioid-dependent, human immunodeficiency virus seronegative participants. Br. J. Clin. Pharmacol. 2019, 85, 1771–1780. [Google Scholar] [CrossRef]
  204. Maggiolo, F.; Rizzardini, G.; Raffi, F.; Pulido, F.; Mateo-Garcia, M.G.; Molina, J.M.; Ong, E.; Shao, Y.; Piontkowsky, D.; Das, M.; et al. Bone mineral density in virologically suppressed people aged 60 years or older with HIV-1 switching from a regimen containing tenofovir disoproxil fumarate to an elvitegravir, cobicistat, emtricitabine, and tenofovir alafenamide single-tablet regimen: A multicentre, open-label, phase 3b, randomised trial. Lancet HIV 2019, 6, e655–e666. [Google Scholar] [PubMed]
  205. Wang, S.C.; Kaur, G.; Schulman-Marcus, J.; Purga, S.; Mookherjee, S.; Miller, C.; Sidhu, M.S.; Rosenson, R.S. Implementation of Cholesterol-Lowering Therapy to Reduce Cardiovascular Risk in Persons Living with HIV. Cardiovasc. Drugs Ther. 2022, 36, 173–186. [Google Scholar] [CrossRef] [PubMed]
  206. Aberg, J.A.; Sponseller, C.A.; Ward, D.J.; Kryzhanovski, V.A.; Campbell, S.E.; Thompson, M.A. Pitavastatin versus pravastatin in adults with HIV-1 infection and dyslipidaemia (IN-TREPID): 12 week and 52 week results of a phase 4, multicentre, randomised, double-blind, superiority trial. Lancet HIV 2017, 4, e284–e294. [Google Scholar] [CrossRef] [PubMed]
  207. Al Sayed, H.A.H.; Sharif-Askari, N.S.; Rahimi, M.R. Clinically significant drug interactions between antiretroviral and co-prescribed drugs in HIV infected patients: Retrospective cohort study. Med. Pharm. Rep. 2022, 95, 260. [Google Scholar] [CrossRef] [PubMed]
  208. Burgess, S.; Partovi, N.; Yoshida, E.M.; Erb, S.R.; Azalgara, V.M.; Hussaini, T. Drug Interactions with Direct-Acting Antivirals for Hepatitis C: Implications for HIV and Transplant Patients. Ann. Pharmacother. 2015, 49, 674–687. [Google Scholar] [CrossRef] [PubMed]
  209. Zhao, A.V.; Crutchley, R.D.; Guduru, R.C.; Ton, K.; Lam, T.; Min, A.C. A clinical review of HIV integrase strand transfer inhibitors (INSTIs) for the prevention and treatment of HIV-1 infection. Retrovirology 2022, 19, 22. [Google Scholar] [CrossRef]
  210. Rathbun, R.C.; Liedtke, M.D. Antiretroviral drug interactions: Overview of interactions involving new and investigational agents and the role of therapeutic drug monitoring for management. Pharmaceutics 2011, 3, 745–781. [Google Scholar] [CrossRef]
  211. Núñez-Núñez, M.; Castañeda-Macías, I.; Sandoval-Fernández Del Castillo, S. Interacciones potenciales en una cohorte de pacientes VIH positivos de edad avanzada. Farm. Hosp. 2018, 42, 163–167. [Google Scholar]
  212. Lopera, V.; Rodríguez, A.; Amariles, P. Clinical Relevance of Drug Interactions with Cannabis: A Systematic Re-view. J. Clin. Med. 2022, 11, 1154. [Google Scholar] [CrossRef]
  213. Franco, D.; Henao, Y.; Monsalve, M.; Gutiérrez, F.; Hincapie, J.; Amariles, P. Interacciones medicamentosas de agentes hipolipemiantes: Aproximación para establecer y valorar su relevancia clínica. Revisión estructurada [Hypoli-pidemic agents drug interactions: Approach to establish and assess its clinical significance. Structured review]. Farm. Hosp. 2013, 37, 539–557. [Google Scholar]
  214. Villa, J.; Cano, A.; Franco, D.; Monsalve, M.; Hincapié, J.; Amariles, P. Relevancia clínica de las interacciones medicamentosas entre antiinflamatorios no esteroideos y antihipertensivos [Clinical relevance of drug interactions between nonsteroidal antiinflammatory drugs (NSAIDs) and antihypertensives]. Aten. Primaria. 2014, 46, 464–474. [Google Scholar] [CrossRef] [PubMed]
  215. Reis, A.M.; Cassiani, S.H. Prevalence of potential drug interactions in patients in an intensive care unit of a university hospital in Brazil. Clinics 2011, 66, 9–15. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Preferred reporting items for systematic review and meta-analysis (PRISMA) [12] flow diagram for the systematic review of the clinical relevance of drug interactions in people living with human immunodeficiency virus.
Figure 1. Preferred reporting items for systematic review and meta-analysis (PRISMA) [12] flow diagram for the systematic review of the clinical relevance of drug interactions in people living with human immunodeficiency virus.
Pharmaceutics 15 02488 g001
Table 1. Levels of the clinical relevance of drug interactions according to the combination of gravity and probability of occurrence [3,4].
Table 1. Levels of the clinical relevance of drug interactions according to the combination of gravity and probability of occurrence [3,4].
Probability
GravityDefinedProbablePossible
Grave1 (very high risk)1 (very high risk) 2 (high risk)
Moderate2 (high risk)2 (high risk)3 (medium risk)
Minor3 (medium risk)3 (medium risk)4 (low risk)
Lack of gravity5 (lowest risk)N/AN/A
N/A: Not applicable.
Table 2. Summary of 291 antiretroviral–drug interactions systematically reported for the first time.
Table 2. Summary of 291 antiretroviral–drug interactions systematically reported for the first time.
Total of Drug Interaction Pairs, n (%)291 (100%)
Pharmacokinetic mechanism233 (80.1%)
  • Enzyme inhibition
101 (43.3%)
  • Enzyme induction
71 (30.5%)
  • Bioavailability modifications based on pH alteration or chelation
19 (8.2%)
  • Bioavailability modifications based on P-gp or presystemic enzyme alterations
39 (16.7%)
  • Protein displacement
1 (0.4)
  • Bidirectional enzyme inhibition/induction
2 (0.9%)
Pharmacodynamic mechanism5 (1.7%)
  • Synergism (toxicity)
5 (100%)
Pharmacokinetic/pharmacodynamic mechanism3 (1.0%)
  • Enzyme inhibition/synergism
2 (66.7%)
  • Enzyme induction/synergism
1 (33.3%)
Evidence of absence of clinically relevant drug interactions50 (17.2%)
Level of the clinical relevance of drug interaction
Level 1Level 2Level 3Level 4Level 5Total, n (%)
42 (14.4%)137 (47.1%) 61 (21.0%) 1 (0.3%)50 (17.2%) 291 (100.0%)
Table 3. Drug interaction pairs with protease inhibitors and non-nucleoside reverse transcriptase inhibitors, levels 1 and 2, systematically reported for the first time.
Table 3. Drug interaction pairs with protease inhibitors and non-nucleoside reverse transcriptase inhibitors, levels 1 and 2, systematically reported for the first time.
Pharmacological Group or Drugs AffectedAntiretroviral AgentGravity/ProbabilityComments/Suggestions
Anesthetic/anxiolytic/benzodiazepine
Midazolam (oral)
[13,14,15]
ATV/RTV(Grave/Possible): Level 2Boosted PIs may increase both midazolam plasma levels and the probability of respiratory depression, sedation, and muscle weakness. Overall, the use of oral midazolam with PIs is not recommended. Additionally, parenteral midazolam has been associated with the risk of severe prolonged sedation in a cohort of hospitalized HIV patients. Thus, this combination should be closely monitored or alternative sedatives for procedural sedation considered (lorazepam or propofol).
Midazolam (intravenous) [13,14,15,16] (Grave/Probable): Level 1
Triazolam
[13,14,15]
ATV/RTV(Grave/Possible): Level 2Boosted PIs may increase both triazolam plasma levels and the probability of respiratory depression, sedation, and muscle weakness. Overall, the use of oral midazolam with PIs is not recommended. Additionally, consider using other safer alternatives, such as oxazepam, lorazepam, or temazepam.
Antiasthmatic/glucocorticoid
Budesonide [13,14,15,16] ATV(Grave/Possible): Level 2ATV may increase the budesonide plasma levels, enhancing the side effects produced by steroids. Monitoring budesonide safety parameters and a dose adjustment may be necessary.
Fluticasone
[17]
LPV/RTVGrave/Possible): Level 2LPV/RTV may increase the fluticasone plasma levels (due to CIP3A4 inhibition), which can cause isolated myopathy (unusual manifestation of inhaled fluticasone). This combination should be avoided. If it is needing, beclomethasone and budesonide are considered as much safer alternatives. Additionally, similar to the COBI-based regimen, consider switching RTV-boosted regimens to BIC-, DTG-, RAL-, or RPV-based regimens.
Anticonvulsant
Carbamazepine–oxcarbazepine
[14,15,16,18]
ATV(Moderate/Probable): Level 2Carbamazepine or oxcarbazepine may cause a significantly lower trough concentration of ATV by 65% (190 ± 91 vs. 546 ± 380 ng/mL), and may be associated with the induction of CYP3A4 and UGT1A1 (metabolizing enzymes). Thus, these combinations should be avoided, owing to the potential risk of virological failure.
Antimalarial
Atovaquone
[19,20,21,22]
EFV, ETR(Moderate/Defined): Level 2EFV or ETR may decrease the AUC and Cmax of atovaquone by inducing glucuronidation. Monitoring atovaquone effectiveness parameters and a dose adjustment may be necessary.
Proguanil
[20,23,24]
EFV, ETR(Moderate/Defined): Level 2EFV or ETR may decrease proguanil AUC and Cmax. Monitoring proguanil effectiveness parameters and a dose adjustment may be necessary.
Atovaquone/proguanil
[15,19,20,21,22,23,24,25]
EFV(Moderate/Defined): Level 2EFV-based antiretroviral therapy decreases AUC atovaquone/proguanil by 75% and proguanil AUC by 44%, leading to possible atovaquone/proguanil prophylaxis failures in HIV-infected patients. Thus, these combinations should be used with caution or avoided. Additionally, an increase in the dose of atovaquone/proguanil or other alternative drug for malaria prophylaxis may be considered.
Artemether/lumefantrine
[26]
ATV/RTV(Moderate/Defined): Level 2ATV/RTV may significantly increase QTc interval from 0.4079 ± 0.008 to 0.4215 ± 0.007 s, leading to artemether/lumefantrine, potentially being cardiotoxic.
Artesunate/amodiaquine
[27]
NVP(Moderate/Probable): Level 2Coadministration of nevirapine and artesunate/amodiaquine may be associated with hepatoxicity and may be associated with synergic hepatotoxicity effect (pharmacodynamic mechanism). Thus, the use of combination artesunate/amodiaquine and NVP should be avoided or be used with a careful monitoring of liver function.
Mefloquine
[28]
NVP(Moderate/Probable): Level 2Mefloquine may cause a decrease in exposure to NVP (concentrations are significantly lower in both maternal plasma and cord plasma, which may increase the risk of mother-to-child transmission among HIV-positive women receiving this combination). Thus, this combination should be avoided.
Artemether/lumefantrine
[29,30,31,32,33]
EFV(Moderate/Defined): Level 2EFV-based antiretroviral therapy may decrease artemether/lumefantrine exposure (47% for lumefantrine) in pregnant and not pregnant women with malaria, which is more pronounced in CYP2B6 slow metabolizers (CYP2B6*6/*6 genotype, linked to two- or three-fold higher EFV plasma levels). Thus, mainly pregnant women or persons with the CYP2B6*6/*6 genotype, may receive subtherapeutic doses due to a higher EFV effect, and, thereby, a dose adjustment and monitoring of artemether/lumefantrine effectiveness may be necessary. Additionally, the EFV effect may be attenuated by using LPV/TRV in conjunction with the NVP-based ART regimen.
Antimicrobial/antituberculosis
Moxifloxacin
[34]
EFV(Moderate/Defined): Level 2EFV may increase moxifloxacin oral clearance by 42%, resulting in a 30% reduction in moxifloxacin AUC, and increasing the risk of resistance and antimicrobial failure. When coadministered with efavirenz, it is necessary to increase the moxifloxacin doses to maintain moxifloxacin levels within the stablished therapeutic range. Additionally, levofloxacin is suggested instead of moxifloxacin when used concomitantly with EFV.
Rifabutin
[13,15,35,36,37,38]
DOR(Moderate/Defined): Level 2Rifabutin may decrease DOR plasma levels through the induction of CYP3A4. Doravirine 100 mg twice daily may be necessary and also to monitor DOR effectiveness parameters.
Rifabutin
[13,35,39]
EFV, ETR(Moderate/Defined): Level 2EFV or ETR may induce CYP3A4 and increase rifabutin metabolism, causing decrease in their plasma levels and response. Thus, daily rifabutin may be increase by 50% (300 mg once daily).
Rifabutin
[13,15,35,40,41,42]
RPV(Moderate/Defined): Level 2Rifabutin may decrease RPV AUC and Cmax by 46% and 35%, respectively, increasing the risk of virological failure and the development of resistance; therefore, this combination is not recommended, mainly in parenteral RPV. Additionally, rifabutin 300 mg daily and a double dose of oral RPV (50 mg) may be used.
Rifampicin
[13,34,38,43]
EFV(Moderate/Defined): Level 2Rifampicin may induce CYP3A4 and decrease EFV exposure and virologic response; thus, although the coadministration of rifampicin and EFV could be safe and effective, the dose of EFV may be increased from 400 to 600 mg once daily (800 mg once daily if patient weigh is >60 kg); it is important to monitor EFV serum levels and, if needed, to adjusted doses to obtain concentrations within the reference range (1–4 mg/L). In addition, if it is possible, the EFV dose may be defined according to the CYP2B6 metabolizer genotype of the patient (slow or extensive). Additionally, EFV may induce CYP3A4 and decrease rifampicin exposure and microbiologic response; thus, the daily dose of rifampicin may be increased by 50% in the presence of EFV (intervention that has a minor effect on EFV concentrations and is well tolerated).
Rifampicin [13,15,21,35,44]DRV/RTV(Grave/Defined): Level 1Rifampicin may significantly decrease DRV/RTV plasma levels by more than 98% (similar to other PIs), increasing the risk of the development of resistance and virological failure. The DRV/RTV (similar to other PIs/RTV) dose increase does not warrant satisfactory DRV exposures and may cause severe alanine transaminase elevations and excessive risk of hepatotoxicity. Alternatively, consider a rifabutin 150 mg daily dose.
Rifampicin
[13,15,35,36,37,42,45]
DOR(Grave/Defined): Level 1Long-term coadministration of rifampicin may induce CYP3A4, causing a decrease in DOR plasma levels by 88%, and may reduce its efficacy. Administration of rifampicin with DOR is not recommended.
Rifampicin [13,15,35,40,41,43,46]RPV(Grave/Defined): Level 1Rifampicin may decrease RPV AUC and Cmax by 80% and 69%, respectively, increasing the risk of virological failure and the development of resistance; therefore, this combination is contraindicated. As an alternative, daily rifabutin at 300 mg and a double dose of RPV are recommended.
Rifampicin
[15,47]
ETR/TAF (Moderate/Defined): Level 2Rifampicin may decrease TAF exposure when ETR/TAF is coadministered; however, intracellular tenofovir diphosphate (DP) concentrations are 4.21 higher when TAF is coadministered with rifampin than when TDF is administered alone, although clinical outcomes have not been studied. Thus, this combination is not recommended; however, if it is coadministered, monitor virologic response.
Antineoplastic
Venetoclax
[48]
RTV(Moderate/Probable): Level 2RTV (doses of 50 mg and 100 mg) may inhibit CYP3A4, resulting in an increase in venetoclax Cmax 2.3 to 2.4 times compared with venetoclax alone and AUC 6.1 and 8.1 times, respectively. After completing the gradual increase in the dose, venetoclax dose reductions of at least 75% are recommended when administered concomitantly with strong CYP3A inhibitors to maintain venetoclax exposures within the therapeutic range established for the treatment of chronic lymphocytic leukemia.
Vimblastine
[49]
ATZ/RTV(Grave/Possible): Level 2RTV-boosted PIs may inhibit CYP3A4, resulting in an increase in vimblastine plasma levels, which increases the risk of severe hypokalemia (1.92 mEq/L) with electrocardiography fluctuations. Thus, this combination should be avoided; additionally, patients can be switched from RTV-boosted PI therapies to RAL, DTG, or BIC-based regimens.
Antiparasitic/anthelmintic
Praziquantel
[50]
EFV(Moderate/Defined): Level 2EFV may decrease AUC praziquantel by four-fold, increasing the risk of treatment failure. To monitor praziquantel effectiveness parameters, a dose adjustment may be necessary, but not exceeding the maximum.
Antiplatelet agent
Clopidogrel
[13,15,34,51,52,53]
ATV/RTV, DRV/RTV, LPV/RTV(Grave/Defined): Level 1PIs boosted with both RTV or COBI inhibit the bioactivation of clopidogrel to its active metabolite. Thus, these boosters may decrease Cmax and AUC0–4h clopidogrel active metabolite from 48 to 68% and from 51 to 69%, respectively, significantly reducing the antiplatelet effect of clopidogrel and increasing the risk for atherothrombotic events, including the recurrence of acute coronary syndrome. Clopidogrel should not be used as an antiplatelet agent for patients on boosted ARV therapies. Prasugrel as antiplatelet agent or unboosted regimens may be used in this context.
Ticagrelor
[13,15,35]
ATV/RTV, DRV/RTV, LPV/RTV(Grave/Possible): Level 2PIs boosted with RTV may increase ticagrelor plasma levels, increasing the risk of bleeding. The use of ticagrelor with boosted PIs is not recommended. Other antiplatelet agents are suggested as an alternative, for instance, prasugrel.
Ticagrelor
[15,53]
EFV(Grave/Possible): Level 2EFV-based ARV therapy may decrease ticagrelor plasma levels, increasing the risk of the recurrence of acute coronary syndrome. The use of ticagrelor with the EFV-based regimen is not recommended. Other antiplatelet agents are suggested as an alternative, for instance, prasugrel.
Antiulcer/anti-H2
Famotidine
[13,35,40,54]
RPV(Moderate/Defined): Level 2The bioavailability of RPV is pH-dependent; thus, famotidine through increasing the gastric pH may decreased RPV AUC concentrations by 76% when administered 2 h prior to RPV, increasing the risk of virological failure and the development of resistance. This combination may be used if famotidine is administered 4 h after or 12 h before RPV.
Antiulcer/proton pump inhibitor
Lansoprazole
[13,14,35,40,41]
RPV(Grave/Defined): Level 1The bioavailability of RPV is pH-dependent; thus, proton pump inhibitors may increase gastrointestinal pH and affect RPV absorption by decreasing its plasma levels. For instance, RPV administered with 20 mg omeprazole once daily decreased RPV AUC by 40% (if RPV was administered at a supratherapeutic daily dose of 150 mg). Thereby, oral RPV is contraindicated with proton pump inhibitors, and this combination should be avoided. However, spacing the administration of esomeprazole by 10 h could be used with RPV.
Omeprazole
[13,14,35,40,41,42]
Pantoprazole
[13,14,35,40,41]
Rabeprazole
[13,14,35,40,41]
Atypical antipsychotic
Quetiapine
[55]
ATZ/TRV DRV/RTV LPV/RTV(Moderate/Probable): Level 2Boosted PIs increase quetiapine plasma levels, mainly explained through CYP3A4 inhibition, which could lead to excess sedation or coma. The administration of quetiapine with PI-based regimens should be avoided or a quetiapine dose reduction is needing, mainly when a PI-based regimen is initiated in a patient on stable quetiapine therapy (a six-fold dose reduction is recommended).
Contraceptive
Levonorgestrel (subdermal implant contraceptive)
[15,34,56,57,58]
EFV(Grave/Defined): Level 1EFV-based ART therapy may significantly reduce levonorgestrel plasma levels by 61%, and the contraceptive effect can be decreased. In addition, even with the doubling of the dose of the levonorgestrel implants, concentrations remained >30% lower. Therefore, levonorgestrel implant is not recommended for women receiving long-term treatment with EFV. The patient should be counselled to use a barrier method as a complementary method of planning or to seek a safer contraceptive alternative.
Etonogestrel (subdermal implant contraceptive) [15,59]EFV(Grave/Defined): Level 1EFV-based ART therapy may significantly reduce etonogestrel plasma levels by 82% in women after 24 weeks of combined use. Therefore, etonogestrel implant is not recommended for women receiving long-term treatment with EFV-based regimen. The patient should be counselled to use a barrier method as a complementary method of planning or to seek a safer contraceptive alternative.
Etonogestrel/ethinylestradiol (vaginal ring)
[15,60,61]
EFV(Grave/Defined): Level 1EFV-based ART therapy may significantly reduce both the etonogestrel and ethinylestradiol levels of the intravaginal ring administered contraceptive by 79–81% and 56–59, respectively, in women after 21 days of coadministration. This pharmacokinetic interaction is most notorious in patients with the CYP2B6 slow metabolizer genotype (levels decrease by 93% for etonogestrel and by 75% for ethinylestradiol). Therefore, etonogestrel/ethinylestradiol (vaginal ring) is not recommended for women receiving treatment with the EFV-based regimen. The patient should be counselled to use a barrier method as a complementary method of planning or to seek a safer contraceptive alternative.
Depot medroxyprogesterone intramuscular
[62]
EFV(Moderate/defined) Level 2EFV-based regimens in HIV and tuberculosis coinfected women and receiving rifampicin and isoniazid for tuberculosis increased the medroxyprogesterone clearance, leading to a more frequent contraceptive dosing (from 12 weeks to 8–10 weeks).
Etonogestrel (oral and subdermal implant contraceptive)
[63]
NVP(Grave/Defined): Level 1In women with the single-nucleotide polymorphism NR1I2 63396 C>T (in gen CYP2B6), the NVP-based ART treatment may reduce etonogestrel plasma levels (Cmin decrease by 39% and AUC0-24 weeks by 37%), significantly decreasing the contraceptive effect. Thus, the etonogestrel implant is not recommended in women with single-nucleotide polymorphism NR1I2 63396 C>T (in gen CYP2B6) receiving long-term treatment with the NVP-based regimen. The patient should be informed of the need to use a barrier method as a complementary method of planning or to seek a safer contraceptive alternative.
Direct-acting antivirals
Glecaprevir/pibrentasvir
[15,22,64,65]
ATV
ATV/RTV
(Grave/Defined): Level 1ATZ and ATV/RTV increase glecaprevir/pibrentasvir plasma levels (until pibrentasvir AUC 14-fold and until glecaprevir AUC 6.5-fold), which may cause elevations in transaminases, jaundice, and severe hyperbilirubinemia. This increase may be mainly explained due to P-gp, BCRP and/or OATP1B1/3 inhibition due to RTV (also COBI)-boosted regimens. The administration of glecaprevir/pibrentasvir with ATV or ATZ/RTV regimens is contraindicated.
Glecaprevir/pibrentasvir
[15,22,64,65]
DRV/RTV, LPV/RTV(Moderate/Defined): Level 2Boosted PIs increase glecaprevir/pibrentasvir plasma levels, mainly explained through P-gp, BCRP, and/or OATP1B1/3 inhibition. The administration of glecaprevir/pibrentasvir with boosted PI regimens is not recommended.
Glecaprevir/pibrentasvir
[15,22,64,65]
EFV(Moderate/Defined): Level 2EFV may reduce glecaprevir/pibrentasvir plasma levels, CYP3A4, and CYP2B6. The administration of glecaprevir/pibrentasvir with EFV is not recommended (overall, it is recommended to avoid the coadministration of any newer-generation direct-acting antiviral with efavirenz due to decreased plasma levels and the possible loss of efficacy).
Elbasvir/grazoprevir
[15]
NVP(Grave/Possible): Level 2NVP may decrease grazoprevir/elbasvir plasma levels, CYP3A4, and CYP2B6. Consider alternative ARV or HCV treatments. If coadministration is necessary, it is recommended to monitor for HCV treatment efficacy.
Sofosbuvir/velpatasvir/voxilaprevir
[15,65,66,67]
ATV/RTV(Grave/Defined): Level 1ATV/RTV single dose increases voxilaprevir AUC by 331%. Concomitant administration between ATV/RTV and voxilaprevir-containing regimens is not recommended.
Sofosbuvir/velpatasvir/voxilaprevir
[15,65,66,67]
EFV(Moderate/Defined): Level 2EFV may decrease velpatasvir plasma levels. Concomitant use of sofosbuvir/velpatasvir with EFV-containing regimens is not recommended due to a 50% reduction in velpatasvir AUC.
Elbasvir/grazoprevir
[15,22,65,68]
ATV/RTV, LPV/RTV, DRV/RTV(Grave/Defined): Level 1Grazoprevir exposure, mainly through OATP1B1/3 inhibition, is noticeably increased with the coadministration of ATV/RTV, LPV/RTV, and DRV/RTV, with AUC0–24 increasing by 10.58, 12.86, and 7.50, respectively. Similarly, elbasvir exposure is increased with the coadministration of ATV/RTV, LPV/RTV, and DRV/RTV, with AUC0–24 increasing by 4.76, 3.71, and 1.66, respectively. Thus, the coadministration of elbasvir/grazoprevir with PI-boosted RTV regimens is contraindicated, due to a marked increase in grazoprevir exposure. Therefore, PI-RTV-boosted regimens should be not used in patients with HCV/HIV coinfection who are being treated with elbasvir/grazoprevir.
Ombitasvir/paritaprevir/RTV/dasabuvir
[69]
DRV/RTV(Moderate/Probable): Level 2Ombitasvir/paritaprevir/ritonavir plus dasabuvir (2D/3D) may cause a significant reduction in DRV levels by 41%. Thus, DRV plasma levels measurement during this combination may be needed. Additionally, ATV should be administered in the morning together with 2D/3D therapy.
Direct-acting oral anticoagulant
Apixaban
[13,15,22,34,70,71,72]
ATV/RTV, DRV/RTV, LPV/RTV(Grave/Possible): Level 2RTV-boosted PIs regimens may increase apixaban plasma levels, mainly through P-gp/CYP3A4 inhibition, increasing the risk of bleeding. Avoid this combination in patients who require apixaban 2.5 mg twice daily; similarly, in patients who require apixaban 5 mg or 10 mg twice daily, reduce apixaban dose by 50% (overall, a dose of 2.5 mg daily or 2.5 mg twice daily is used). Additionally, consider switching PI-RTV-boosted regimens to BIC-, DTG-, RAL-, or RPV-based regimens.
Apixaban
[15,22,70]
EFV, ETR, NVP (Grave/Possible): Level 2EFV, ETR, and NVP may decrease apixaban plasma levels, via CYP3A4 induction, increasing the risk of thrombotic events. The use of these combinations is not recommended; it is suggested to use other anticoagulants as an alternative.
Edoxaban [13,15,22,35,70,72]ATV/RTV, DRV/RTV, LPV/RTV(Grave/Possible): Level 2RTV-boosted PIs regimens may increase edoxaban plasma levels, mainly through P-gp inhibition. It may be necessary to monitor edoxaban safety parameters. For deep venous thrombosis and pulmonary embolism, it is recommended to reduce the dose of edoxaban from 60 mg to 30 mg daily. However, for stroke prevention in nonvalvular atrial fibrillation indication, no dose adjustment is recommended.
Rivaroxaban
[13,15,35,72,73,74]
ATV/RTV, DRV/RTV, LPV/RTV(Grave/Probable): Level 1RTV-boosted PIs regimens may increase the rivaroxaban AUC and Cmax by 153% and 55%, respectively, and increase the risk of bleeding events, mainly through P-gp/CYP3A4 inhibition. It is not recommended to use these combinations; it is suggested to use other anticoagulants as an alternative or to consider switching PI-RTV-boosted regimens to BIC-, DTG-, RAL-, or RPV-based regimens.
Drug substance misuse
Amphetamine and substituted amphetamines (methamphetamine, methylenedioxymethamphetamin—MDMA; ‘Ecstasy’)
[14,75,76,77,78,79,80]
ATV/RTV, DRV/RTV, LPV/RTV(Grave/Possible): Level 2RTV-boosted (ATV, DRV, or LPV) regimens may inhibit amphetamine metabolism, mainly through RTV CYP2D6 inhibition, leading to increased plasma levels (until 10-fold higher than expected) and toxicity, including fatal serotonergic reaction. Advise patients about the risks of these combinations.
Food/supplements
Garlic-containing products
[81]
DRV/RTV(Grave/Possible):
Level 2
Significant dietary garlic consumption may cause subtherapeutic DRV levels, probably due to the induction of duodenal P-gp, which increases the RTV efflux, decreasing the bioavailability and levels of RTV associated with a lower facility to boost DRV levels. The subtherapeutic DRV levels increase the risk of viral rebound. Thus, garlic consumption should be avoided during DRV-based antiretroviral regimen.
Mineralocorticoid
Eplerenone
[13,15,82,83]
ATV/RTV, DRV/RTV, LPV/RTV(Grave/Possible): Level 2RTV-boosted PIs regimens may increase eplerenone plasma levels, increasing the risk of possible life-threatening hyperkalemia. The use of this combination is not recommended.
Special conditions
Adults > 65 years
[84]
DRV and ATV Moderate/probableHigher steady-state plasma levels of DRV and ATV are found in PLHIV aged ≥ 65 years of age, compared to controls ≤ 49 years of age. Thus, in patients 65 years of age or older, the monitoring of plasma levels and dose adjustment of DRV and ATV are recommended.
Methadone for heroin addiction
[85,86]
ATV/RTV
DRV/RTV, LPV/RTV
(Moderate/Probable): Level 2RTV-boosted PIs decrease (mainly through CYP2B6 induction) methadone plasma levels. The administration of methadone with RTV-boosted PI regimens need to increase the dose of MTD (mg/kg-body weight) by 50%.
Pregnancy
[15,87,88]
ATV,
DRV,
LPV
(Grave/Possible): Level 2Pregnancy may decrease PI exposure (the average AUC is decreased in a range of 5–56% during the first, second, and third trimesters vs. postpartum). Thus, RTV-boosted PIs regimens are needed in women who become pregnant. Overall, the comparisons between RTV-boosted PIs regimens showed no differences, for instance, there are no differences between ATV/RTV, LPV/TRV, or DRV/RTV regimens.
Pregnancy
[15,87]
DOR(Grave/Possible): Level 2Pregnancy may decrease DOR exposure. Currently, its use is not recommended in women who become pregnant while on ART treatment.
Renal transplant recipients
[89,90]
ATV/RTV(Grave/Probable): Level 1Regimen switch from ATV/RTV to DTG/FTC/TAF in a postrenal transplant recipient may cause subtherapeutic tacrolimus levels higher than expected from dolutegravir initiation (via organ cation transporter 2 inhibition), increasing the risk of increased serum creatinine and graft rejection. Thus, postrenal transplant recipients switched from ATV/RTV to DTG regimens need a tacrolimus dose increase (which could be up to 70-fold: 0.5 mg/week to 5 mg twice daily) to achieve the tacrolimus therapeutic range (usually, 4–8 ng/mL in postrenal transplantation) with careful clinical monitoring.
Weight-loss drugs
Orlistat
[91,92]
ATZ/RTV(Grave/Possible): Level 2Overall, the absorption of class II drugs (extensive presystemic metabolism, low water solubility, and high lipid permeability), for instance, atazanavir, is likely to be meaningfully affected by orlistat. Therefore, ATV/RTV-based regimen effectiveness may be affected by orlistat, which could be linked to the risk of virologic failure, loss of control of HIV viremia, and HIV viral rebound. Thus, this combination should be avoided.
Orlistat
[91,92,93]
EFV(Grave/Possible): Level 2Overall, the absorption of class II drugs (extensive presystemic metabolism, low water solubility, and high lipid permeability), for instance, EFV, is likely to be meaningfully affected by orlistat. Therefore, EFV-based regimen effectiveness may be affected by orlistat, which could be linked to the risk of virologic failure, loss of control of HIV viremia, and HIV viral rebound. Thus, this combination should be avoided.
ART: antiretroviral therapy; ATV: atazanavir; ATV/RTV: atazanavir/ritonavir; AUC: area under the curve; BCRP: breast cancer resistance protein; Cmax: maximum concentration; Cmin: minimum concentration; CYP: cytochrome; DRV/COBI: darunavir/cobicistat; DRV/RTV: darunavir/ritonavir; DOR: doravirine; ECG: electrocardiogram; EFV: efavirenz; ETR: etravirine; HIV: human immunodeficiency virus; LPV/RTV: lopinavir/ritonavir; NNRTIs: non-nucleoside reverse transcriptase inhibitors; NVP: nevirapine; OATP: organic anion transporting polypeptide; P-gp: P-glycoprotein; PIs: protease inhibitors; RPV: rilpivirine; RTV: ritonavir.
Table 4. Drug interaction pairs with nucleoside/nucleotide reverse transcriptase inhibitors, integrase inhibitors, and cobicistat-boosted regimes, levels 1 and 2, systematically reported for the first time.
Table 4. Drug interaction pairs with nucleoside/nucleotide reverse transcriptase inhibitors, integrase inhibitors, and cobicistat-boosted regimes, levels 1 and 2, systematically reported for the first time.
Pharmacological Group or Drugs AffectedAntiretroviral AgentGravity/ProbabilityComments/Suggestions
Anesthetic/anxiolytic/benzodiazepine
Midazolam (oral)
[13,14,15,94]
EVG/COBI(Grave/Defined):
Level 1
COBI may reduce midazolam clearance near to 96%, increasing midazolam plasma levels, increasing the probability of respiratory depression, sedation, and muscle weakness. Thus, the use of this combination is not recommended. If necessary, parenteral midazolam should be used with close clinical monitoring or safer alternatives, such as lorazepam or propofol.
Alprazolam, clonazepam
diazepam, and triazolam
[13,14,15]
(Grave/Possible): Level 2EVG/COBI may reduce the clearance of alprazolam, clonazepam, diazepam, and triazolam, increasing plasma levels and the probability of these benzodiazepines’ toxicity (respiratory depression, sedation, and muscle weakness). These combinations should be used with caution, monitoring for adverse effects of benzodiazepines. Additionally, safer alternatives, such as lorazepam, oxazepam, or temazepam, should be used.
Cation-containing antacids/supplements and iron or zinc products
Aluminum hydroxide
[13,14,15,22,95]
EVG/COBI,
RAL,
DTG,
BIC,
CAB
(Moderate/Defined): Level 2Calcium, aluminum, and magnesium (Ca2+, Al3+, Mg2+)-containing antacids/supplements and also iron or zinc (Fe2+, Zn2+)-containing products may form chelates with InSTIs, thereby decreasing their absorption and efficacy. For instance, calcium carbonate may cause a reduction in both DTG and BIC AUC and Cmax by up to 33–42%; ferrous fumarate may cause a decrease in AUC and Cmax by 55–58% for DTG and by 63–70% for BIC. Additionally, BIC coadministered simultaneously with calcium, aluminum, and magnesium antacids under fasted conditions significantly reduced BIC exposure by 63–79%. In this sense:
EVG/COBI: Should be administered at least 4 h separate from antacids or iron/zinc supplements.
RAL: Do not coadminister with aluminum- and magnesium-containing antacids. Coadministration is possible with calcium-containing antacids, but only with 400 mg twice daily. Administer separately by 4 h from iron/zinc supplement administration.
DTG: Administer at least 2 h before or at least 6 h after the administration of antacids containing calcium, magnesium, or aluminum.
BIC: With antacids containing calcium should be administered together with food; do not coadminister simultaneously on an empty stomach.
CAB: Administer at least 2 h before or at least 4 h after the administration of antacids containing calcium, magnesium, or aluminum.
Additionally, it is very important to assess for use of over-the-counter drugs and vitamins/supplements in patients on InSTI-based antiretroviral therapy.
Calcium carbonate
[13,14,15,22,95]
Ferrous fumarate
[13,14,15,22,95]
Magnesium hydroxide
[13,14,15,22,95]
Zinc gluconate
[13,14,15,22,95]
Antianginal
Ranolazine
[15,102]
DRV/COBI(Grave/possible): Level 2DRV/COBI regimens may increase ranolazine plasma levels, mainly through CYP3A4 inhibition, increasing risk of adverse events (persistent episodes of nausea, vomiting, dyspepsia, anorexia, dizziness, syncope, and, potentially, auriculo-ventricular block). Thus, these combinations are not recommended.
Antiarrhythmic
Dofetilide
[14,15]
BIC
DTG
(Grave/possible): Level 2BIC and DTG administration may increase dofetilide plasma levels, for instance, 70–90% by DTG. Thus, these combinations are not recommended.
Antiasthmatic/glucocorticoid
Fluticasone
[13,14,35,103,104,105,106,107,108,109,110]
EVG/COBIGrave/Probable):
Level 1
EVG/COBI causes a significant increase in exposure to fluticasone (due to CIP3A4 inhibition), which can cause Cushing’s syndrome and adrenocortical suppression, leading to secondary adrenal insufficiency. This drug interaction can occur after a brief exposure (10–14 days) and with fluticasone nasal drops. This combination should be avoided. If it is needed, beclomethasone can be used as an alternative, but with caution. Additionally, similar to the RTV-based regimen, consider switching COBI-boosted regimen to BIC-, DTG-, RAL-, or RPV-based regimens.
Budesonide
[13,14,35,103,104,105]
EVG/COBIGrave/Possible):
Level 2
EVG/COBI causes a significant increase in exposure to budesonide (due to CIP3A4 inhibition), which can cause Cushing’s syndrome and adrenocortical suppression, leading to secondary adrenal insufficiency. This combination should be used cautiously, monitoring budesonide toxicity (in acutely unwell, hypotensive, and hyponatremic patients).
Mometasone
[13,14,35,103,104,108]
EVG/COBI(Grave/Possible): Level 2EVG/COBI may increase mometasone plasma levels (due to CIP3A4 inhibition), which can cause Cushing’s syndrome and adrenocortical suppression, leading to secondary adrenal insufficiency. This combination should be used cautiously, monitoring mometasone toxicity (in acutely unwell, hypotensive, and hyponatremic patients).
Triamcinolone (injectable)
[13,35,103,108,111,112,113]
EVG/COBI(Grave/Probable): Level 1EVG/COBI causes a significantly increase in exposure to triamcinolone (due to CIP3A4 inhibition), which can cause Cushing’s syndrome and adrenocortical suppression, leading to secondary adrenal insufficiency. This combination should be avoided. If it is needed, methylprednisolone can be used as an alternative, but with caution. Additionally, similar to the RTV-based regimen, consider switching COBI-boosted regimen to BIC-, DTG-, RAL-, or RPV-based regimens.
Triamcinolone (intrabursal)
[13,15,35,108,114]
DRV/COBI(Grave/possible): Level 2DRV/COBI may increase triamcinolone plasma levels (due to CIP3A4 inhibition), which can cause Cushingoid features and an undetectable cortisol, leading to secondary hypoadrenalism. This combination should be avoided. Additionally, similar to the RTV-based regimen, consider switching COBI-boosted regimen to BIC-, DTG-, RAL-, or RPV-based regimens.
Betamethasone (topical)
[15,115]
DRV/COBI(Grave/possible): Level 2DRV/COBI may increase betamethasone plasma levels (due to CIP3A4 inhibition), if it is used topically and for ≥2 months. This alteration may cause Cushingoid features and an undetectable cortisol, leading to secondary hypoadrenalism. In patients on COBI-based regimen, the use of chronic topical betamethasone should be avoided.
Anticoagulant/antivitamin K
Warfarin
[116]
DTG
(Grave/Possible): Level 2Switching from NEV-based regimen to DTG-based regimen may increase warfarin plasma levels and INR. Close monitoring of INR is mandatory, due to warfarin dosing requirements potentially significantly reducing.
Warfarin
[15,35,70,117]
EVG/COBI(Grave/possible): Level 2EVG/COBI appears to have a strong induction on CYP2C9, leading to a decrease in warfarin plasma levels and INR and a warfarin dosage increase.
Warfarin
[118]
EVG/COBI/TAF/FTC(Grave/possible): Level 2EVG/COBI/TAF/FTC, mainly TAF, may increase warfarin plasma levels and INR from 2.7 to 4.5 (after 7 days) using the usual warfarin dosage, then to 6.5 (after 14 days), despite a reduction in the warfarin dose. This effect could be attributed to TAF; however, the mechanism is not clear.
Warfarin
[13,14,35,119,120]
DRV/COBI(Moderate/Probable: Level 2)DRV/COBI may increase warfarin plasma levels, increasing the risk of bleeding. However, boosted ARVs may cause both inhibiting/inducing effects on CYPs and, therefore, are expected to alter warfarin plasma levels. Therefore, close monitoring of INR to determine appropriate warfarin dose in patients on ARV treatment is recommended.
Anticonvulsant
Carbamazepine–oxcarbazepine
[14,15,18,35,96]
DTG(Moderate/Probable): Level 2Carbamazepine or oxcarbazepine may cause a significantly lower trough concentration of DTG by 83% (191 ± 78 vs. 1096 ± 510 ng/mL), and may be associated with the induction of CYP3A4 and UGT1A1 (metabolizing enzymes). Thus, these combinations should be avoided, owing to the potential risk of virological failure. Additionally, in ART-naive or ART-experienced (except for InSTIs-naive) patients, increase DTG to 50 mg twice daily use. However, in InSTIs-experienced patients with known or suspected InSTIs resistance, this combination should not be used. Additionally, alternatives, such as levetiracetam and/or gabapentin (not CYP metabolized), may be considered.
Phenobarbital
[14,15,35,121]
DTG(Moderate/Probable): Level 2Phenobarbital may cause a significantly lower trough concentration of DTG by 83% (191 ± 78 vs. 1096 ± 510 ng/mL), and may be associated with the induction of CYP3A4 and UGT1A1 (metabolizing enzymes). Thus, these combinations should be avoided, owing to the potential risk of virological failure. Additionally, in ART-naive or ART-experienced (except for InSTIs-naive) patients, increase DTG to 50 mg twice daily use. However, in InSTIs-experienced patients with known or suspected InSTIs resistance, this combination should not be used. Additionally, alternatives, such as levetiracetam and/or gabapentin (not CYP metabolized), may be considered.
Valproic acid
[122,123,124,125]
DTG(Moderate/Probable): Level 2Valproic acid may decrease DTG plasma levels due to a mechanism, including protein displacement, increasing the induction of CYP3A4 and UGT1A1 (metabolizing enzymes) or P-glycoprotein (transporting enzyme). Closely monitor DTG effectiveness parameters, because it may lead to increased resistance and increased viral load. In addition, other anticonvulsant alternatives such as levetiracetam are suggested. However, if both dolutegravir is taken with food and patient is adherent to treatment, this DDI may be not clinically relevant. More clinical information is needed.
Atovaquone/proguanil
[15,20,21,22,24,25]
ATZ/RTV
LPV/RTV
(Moderate/Defined): Level 2ATZ/RTV- or LPV/RTV-based antiretroviral therapy decreases AUC atovaquone/proguanil (with ATV/RTV: atovaquone AUC 46% and proguanil AUC by 41%; with LPV/TRV: atovaquone AUC by 74%), leading to possible atovaquone/proguanil prophylaxis failures in HIV-infected patients. Thus, these combinations should be used with caution or avoided. Additionally, an increase in the dose of atovaquone/proguanil or other alternative drug for malaria prophylaxis may be considered.
Antimicrobial/sulfonamide
Cotrimoxazole (trimethoprim/sulfamethoxazole
[21,126]
3TC(Moderate/Defined): Level 2 Trimethoprim administration may cause 43% increase in AUC and a 35% decrease in renal clearance of 3TC, leading to increase in the plasma levels and in the risk to development metabolic acidosis and hyperlactatemia. The dose of 3TC may be reduced in patients with renal disease; however, in patients with normal renal function, there is no need to adjust the dose.
Antimicrobial/antituberculosis
Rifampicin
[13,15,34,94,123,124]
BIC(Grave/Defined): Level 1Rifampicin may induce BIC metabolism, causing a significant decrease in AUC (60%) and in trough concentrations (80%), resulting in a loss of therapeutic effect. Dose adjustment recommendations for coadministration of BIC and rifampicin have not been established; however, the combination is contraindicated.
Rifabutin
[13,15,35,127,129]
BIC(Moderate/Defined): Level 2Rifabutin may induce BIC metabolism, causing a significant decrease in AUC (38%) and trough concentrations (56%), and might generate ineffectiveness. Dose adjustment recommendations for coadministration of BIC and rifabutin have not been established and the combination is not recommended.
Rifampicin
[15,35,40,96,130,131]
CAB(Grave/Defined): Level 1Rifampicin may induce CAB metabolism, causing a decrease in the CAB AUC and the half-life by 59% and 57%, respectively, while clearance is increased 2.4-fold, resulting in loss of therapeutic effect. Additionally, oral rifampicin may decrease exposure to intramuscular CAB by 44%. Dose recommendations for the coadministration of CAB and rifampin have not been established; however, the combination is not recommended, regardless of the route of CAB administration.
Antimigraine
Ergotamine
[15,132,133]
EVG/COBI(Grave/Possible): Level 2Similar to RTV, COBI may increase plasma ergotamine concentrations by inhibiting CYP3A4, increasing the probability of ergotism, including gastrointestinal and muscle disorders, as well as the sudden onset of severe pain and paresthesias in both legs associated with acute ischemia. To monitor ergotamine safety parameters, it is important to counsel about the risk associated with the use of ergotamine-containing drugs through self-medication.
Antineoplastic
Vinblastine
[134]
AZT(Grave/Possible): Level 2AZT may increase the probability of hematologic toxicity with the use of vinblastine. This combination is not recommended due to the possibility of serious adverse effects.
Vinblastine
[108,135]
EVG/COBI(Grave/Possible): Level 2COBI-boosted regimens may increase vimblastine plasma levels, increasing the risk of peripheral neuropathy (sensory–motor lower and upper limbs). The use of vimblastine with boosted ARV (including both RTV and COBI) is not recommended. Unboosted ARV regimens may be used in this context.
Antiplatelet agents
Clopidogrel
[13,35,51,52]
EVG/COBI(Grave/Defined): Level 1COBI-boosted regimens inhibits the bioactivation of clopidogrel to its active metabolite, significantly reducing the antiplatelet effect of clopidogrel and increasing the risk for atherothrombotic events. Clopidogrel should not be used as an antiplatelet agent for patients on EVG/COBI-based regimens. Prasugrel as an antiplatelet agent or unboosted regimens may be used in this context.
Ticagrelor
[13,15,35]
EVG/COBI(Grave/possible): Level 2EVG/COBI may increase ticagrelor plasma levels, increasing the risk of bleeding. The use of ticagrelor with boosted ARVs is not recommended. Other antiplatelet agents are suggested as an alternative.
Contraceptive
Drospirenone/ethinyl estradiol
[136]
ATV/COBI
DRV/COBI
(Moderate/probable): Level 2 Drospirenone AUC is increased with the coadministration of COBI-containing regimens (1.6-fold with DRV/COBI and 2.3-fold with ATV/COBI), maybe due to COBI-mediated CYP3A inhibition. The increase in drospirenone AUC increases the risk of drospirenone-associated hyperkalemia. Therefore, with DRV+COBI monitoring for hyperkalemia is recommended, and the combination of ATV+COBI with drospirenone should be avoided.
Direct-acting antivirals
Elbasvir/grazoprevir
[15,22,65,84]
EVG/COBI(Moderate/Defined): Level 2Elbasvir and grazoprevir exposure is increased with the coadministration of EVG/COBI; thus, AUC0–24 increases by 2.2 and 5.4, respectively. Due to the increased grazoprevir exposure associated with the coadministration of EVG/COBI, this combination is not recommended in patients with HCV/HIV coinfection, who are being treated with elbasvir/grazoprevir.
Direct oral anticoagulant
Apixaban
[13,15,22,35,70,71,72]
EVG/COBI(Grave/Possible): Level 2EVG/COBI may increase apixaban plasma levels mainly through P-gp/CYP3A4 inhibition. Avoid this combination in patients who require apixaban 2.5 mg twice daily; similarly, in patients who require apixaban 5 mg or 10 mg twice daily, reduce apixaban dose by 50%. Consider switching COBI-boosted regimens (similar to RTV- and RTV-boosted regimens) and PIs-RTV boosted regimens to BIC-, DTG-, RAL-, or RPV-based regimens.
Apixaban [13,15,22,35,70,71,72,137]DRV/COBI(Grave/Possible): Level 2DRV/COBI may increase apixaban plasma levels mainly through P-gp/CYP3A4 inhibition. Avoid this combination in patients with a recent vascular procedure or in those who require apixaban 2.5 mg twice daily; similarly, in patients who require apixaban 5 mg or 10 mg twice daily, reduce apixaban dose by 50%. Consider switching COBI-boosted regimen (similar to RTV and RTV-boosted regimens) to BIC-, DTG-, RAL-, or RPV-based regimens.
Dabigatran
[13,15,34,69,71,138,139,140]
EVG/COBI(Grave/Defined): Level 1COBI may increase AUC and Cmax dabigatran by 127% in both, leading to an increase in thrombin time assessment by 33% for the area-under-the-effect curve, since time zero to 24 h and 51% at 24 h, mainly through P-gp inhibition. Overall avoid this combination. Consider switching COBI-boosted base regimen to BIC, DTG, RAL, RPV-, or inclusive to RTV-boosted regimen. Dabigatran dosing recommendation depends on estimated creatinine clearance (CLCr): (a) Severe renal failure (CLCr < 30 mL/min): coadministration of dabigatran and COBI (and RTV) should be avoided. (b) Moderate renal failure (CLCr 30–59 mL/min): dabigatran and COBI: 75 mg twice daily (dabigatran and RTV: 110 mg twice daily). (c) Normal renal function (CLCr > 60 mL/min): dabigatran and COBI: 110 mg twice daily (dabigatran and RTV: 150 twice daily, usual dose).
Edoxaban
[13,15,35,70,72]
ATV/COBI(Grave/Possible): Level 2ATV/COBI may increase edoxaban plasma levels mainly through P-gp inhibition. It is not recommended to use this combination; it is suggested to use other anticoagulants as an alternative or to switch COBI-boosted regimen (similar to RTV- and RTV-boosted regimens) to BIC-, DTG-, RAL-, or RPV- based regimens.
Edoxaban
[13,15,22,35,70,72]
DRV/COBI(Grave/Possible): Level 2DRV/COBI may increase plasma concentrations of edoxaban through the inhibition of CYP3A4 and P-gp. It is recommended to monitor edoxaban safety parameters, especially in patients with renal disease.
Edoxaban
[13,15,22,35,71,72]
EVG/COBI(Grave/Possible): Level 2EVG/COBI may increase edoxaban plasma levels through the inhibition of CYP3A4 and P-gp. It is recommended to monitor edoxaban safety parameters. It is recommended to reduce the dose of edoxaban from 60 mg to 30 mg in patients with deep venous thrombosis and pulmonary embolism.
Rivaroxaban [13,15,35,73,74,141]EVG/COBI(Grave/Possible): Level 2EVG/COBI may increase rivaroxaban plasma levels and bleeding events mainly through P-gp/CYP3A4 inhibition. It is not recommended to use this combination; it is suggested to use other anticoagulants as an alternative or to switch COBI-boosted regimen (similar to RTV- and RTV-boosted regimens) to BIC-, DTG-, RAL-, or RPV-based regimens.
Drug substance misuse
Amphetamine and substituted amphetamines (methamphetamine, methylenedioxymethamphetamine—MDMA; ‘Ecstasy’)
[14,15,75,76,80,141,142]
EVG/COBI(Grave/Possible):
Level 2
EVG/COBI (and other COBI-boosted regimens), mainly through CYP2D6 inhibition, may increase plasma levels of amphetamines, prolong their clinical effects, and increase toxicity. Overall, concomitant use of CYP2D6 inhibitors such as COBI (RTV) could increase amphetamine toxicity due to drug accumulation. In these cases, it is recommended to use unboosted regimens, and counselling patients about the risks of these combinations.
Ketamine
[14,15,75,76,142,143]
EVG/COBI
DRV/COBI
(Grave/Possible):
Level 2
EVG/COBI (and other boosted regimens), mainly through CYP3A4 inhibition may increase plasma levels of ketamine, prolong its clinical effects, and increase toxicity. Overall, the concomitant use of CYP3A4 inhibitors, such as COBI (RTV), could increase ketamine toxicity due to drug accumulation. In these cases, it is recommended to use unboosted regimens and counselling patients about the risks of these combinations.
Feminizing hormone therapy (FHT)
Estradiol valerate/cyproterone acetate
[144,145,146,147,148,149,150,151,152]
TDF/FTC (on- demand PrEP: 2, 1, 1)(Moderate/Probable): Level 2TDF/FTC exposure may be reduced by 12–32% among transgender women vs. cisgender men in the presence of FHT, suggesting that it may potentially affect the effectiveness of PrEP. Additionally, active metabolites and competing deoxynucleotides in the rectal tissue of transgender women are seven-fold lower in rectal TFVdp:dATP vs. cisgender men. However, some results show no interaction for FHT on tenofovir levels and supporting PrEP use among transgender women using FHT. The clinical significance and the mechanism of this drug interaction is still unclear and this reduction may impact the HIV protective efficacy of a daily PrEP regimen. However, FHT may produce concentrations too low for the consistent prevention of HIV infection on demand in the PrEP regimen: 600 mg TDF/400 mg FTC on day 1 and 300 mg TDF/200 mg FTC on days 2 and 3.
Foods/supplements
Guggulsterone-containing supplements
[153]
EVG/COBI(Grave/Possible):
Level 2
Guggulsterone may be a pregnane X receptor (PXR) agonist, causing the induction of the expression of CYP3A genes. Thus, the concomitant administration of guggulsterone-containing supplements may cause a significant reduction in the EVG plasma levels, increasing the risk of viral rebound after a 4-month treatment. Thus, guggulsterone-containing thermogenic complex consumption should be avoided during the EVG/COBI treatment.
Sorbitol
[154]
3TC(Grave/Possible):
Level 2
Sorbitol 3.2, 10.2, and 13.4 g decrease 3TC Cmax by 28%, 52%, and 55% and AUC0–24 by 20%, 39%, and 44%, respectively, explained due to an absorption-based interaction, which can lead to 3TC-based regimen therapeutic failure. Thus, the coadministration of 3TC-based regimens with sorbitol-containing medicines should be avoided; however, if it is coadministered, monitor the virologic response.
Mineralocorticoid
Eplerenone
[13,15,83]
EVG/COBI(Grave/Possible): Level 2EVG/COBI may increase eplerenone plasma levels, by affecting the hepatic/intestinal enzyme CYP3A4 metabolism, increasing the risk of hyperkalemia. The use of this combination is not recommended.
Phosphodiesterase 5 inhibitors
Sildenafil
[13,14,15,155]
EVG/COBI
DRV/COBI
(Grave/Possible): Level 2COBI-boosted (similar to RTV-boosted) regimens may increase sildenafil plasma levels (10 mg 3 times daily causes concentrations similar to 20 mg 3 times daily, recommended dose for pulmonary arterial hypertension), mainly through CYP3A4 inhibition. Therefore, this combination increases the risk of hypotension and respiratory failure and it is not recommended. Failing that, begin sildenafil at 10 mg 3 times daily (lower doses) and monitor its plasma levels.
Special conditions
Pregnancy
[15,156]
BIC(Grave/Possible): Level 2Pregnancy may decrease BIC exposure. More research and information regarding clinical efficacy and safety of BIC during pregnancy is needed. This drug is not currently recommended in pregnancy.
Pregnancy
[15,87,157,158]
EVG/COBI(Grave/Defined): Level 1Overall, COBI, mainly with the EVG regimen, should be avoided in pregnancy due to lower concentrations of COBI and its boosted drugs—EVG, DRV, and ATV (the average AUC is decreased in a range of 5–56% during the first, second, and third trimester vs. postpartum). For instance, pregnancy may decrease the AUC of EVG and COBI by 24% and 44%, respectively, during the second trimester, increasing the risk of virologic failure and mother-to-child transmission. Additionally, pregnancy may decrease the AUC of COBI by 59% during the third trimester. If women become pregnant during treatment with EVG/COBI, it should be changed to an alternative boosting, such as ritonavir, or the viral load should be monitored every month.
Pregnancy
[15,131,159]
CAB/RPV(Grave/Possible): Level 2CAB/RPV is not recommended for use in pregnancy, due to lack in data existing for people who are trying to conceive or who become pregnant. Overall, CAB is not recommended during pregnancy and there is a need for additional studies regarding the safety of this drug during pregnancy.
Pregnancy
[87,160,161]
TDF monotherapy(Moderate/Defined): Level 2Among women with HIV on TDF with RTV/PIs or without RTV/PIs and women with HBV on treatment with TDF monotherapy, TFV exposure is decreased by nearly 25% during the third trimester of pregnancy, probably associated with an increase because of an increased volume of distribution and renal excretion. However, this does not apply to LPV/RTV, due to some results having showed that tenofovir exposure is comparable between the third trimester of pregnancy and postpartum among women receiving concomitant LPV/RTV.
Renal transplant recipients
[162]
EVG/COBIGrave/possible): Level 2 EVG/COBI may cause supratherapeutic tacrolimus concentrations in postrenal transplant recipients (level of 111.2 ng/mL at 1 week, goal trough level 4–6 ng/mL), mainly via CYP3A inhibition, increasing risk of acute kidney damage (serum creatinine level increasing from 1.5 to 2.3 mg/dL). Thus, in postrenal transplant recipients, this combination should be avoided.
Statins
Lovastatin
[13,14,15,22,35,163]
EVG/COBI(Grave/Possible): Level 2EVG/COBI may increase lovastatin plasma levels, increasing the risk of muscle toxicity, severe muscle pain, rhabdomyolysis, and acute renal injury. This combination is not recommended. Safer alternatives, such as pravastatin or fluvastatin, should be used.
Simvastatin
[13,14,15,22,35,163,164]
EVG/COBI(Grave/Possible): Level 2EVG/COBI may increase simvastatin plasma levels, increasing the risk of muscle toxicity, severe muscle pain, rhabdomyolysis, and acute renal injury. This combination is not recommended. Safer alternatives, such as pravastatin or fluvastatin, should be used.
Pravastatin/fenofibrate [14,15,22,165]EVG/COBI(Grave/Possible): Level 2EVG/COBI may increase pravastatin plasma levels, increasing the risk of muscle toxicity, severe muscle pain, rhabdomyolysis, and acute renal injury. It is possible that COBI increases pravastatin plasma levels through complex mechanisms, including inhibition of BCRP intestinal and biliary efflux pumps; the inhibition of the P-gp intestinal, biliary, and renal efflux pumps; and the inhibition of OATP1B1-mediated entry into hepatocytes. This combination should be used with caution, using the lowest effective pravastatin dose together with monitoring for muscle toxicity signs.
Atorvastatin
[13,14,15,22,35,83,163,166]
EVG/COBI(Grave/Possible): Level 2ATV/COBI and DRV/COBI may increase atorvastatin AUC by 822–853% and 292%, respectively, increasing the risk of muscle toxicity, severe muscle pain, rhabdomyolysis, and acute renal injury. These combinations should be used with caution, atorvastatin doses should not exceed 20 mg daily (10 mg with ATV/COBI), and monitor for typical adverse effects associated with statins. Additionally, safer alternatives, such as pravastatin or fluvastatin, should be used.
Rosuvastatin
[13,14,15,22,35,83,163,166]
ATV/COBI
DRV/COBI
(Moderate/Defined): Level 2ATV/COBI and DRV/COBI may increase rosuvastatin AUC by 218–242% and 89–93%, respectively, increasing the risk of muscle toxicity, severe muscle pain, rhabdomyolysis, and acute renal injury. These combinations should be used with caution, atorvastatin doses should not exceed 20 mg daily (10 mg with ATV/COBI), and monitor for typical adverse effects associated with statins. Additionally, safer alternatives, such as pravastatin or fluvastatin, should be used.
Simvastatin
[13,14,15,22,35,166]
ATV/COBI
DRV/COBI
(Moderate/Defined): Level 2DRV may increase simvastatin plasma levels, increasing the risk of muscle toxicity, severe muscle pain, rhabdomyolysis, and acute renal injury. This combination is not recommended. Safer alternatives, such as pravastatin or fluvastatin, should be used.
Uricosuric agent
Probenecid
[167]
TDF/FTC (on-demand PrEP: 2,1,1)(Moderated/Defined): Level 2Probenecid increases the AUC of TDF and FTC by 61% and 68%, respectively, mainly through the inhibition of OAT1/OAT3, which leads to decreased TDF uptake into renal cells available for elimination. However, the TDF diphosphate (TDF active form) concentrations in peripheral blood mononuclear cells are higher (~30%) at 24 h, but then fall significantly lower by 40% at 72 h. In patients on-demand PrEP with TDF/FTC (2, 1, 1), the risk/benefit of probenecid addition should be assessed.
ARV: antiretroviral; ATV/COBI: atazanavir/cobicistat; AUC: area under the curve; AZT: zidovudine; BCRP: breast cancer resistant protein; BIC: bictegravir; BCRP: breast cancer resistance protein; CAB: cabotegravir; COBI: cobicistat; CYP: cytochrome; DRV/COBI: darunavir/cobicistat; DTG: dolutegravir; ECG: electrocardiogram; EVG: elvitegravir; EVG/COBI: elvitegravir/cobicistat; EVG/COBI/TAF/FTC: elvitegravir/cobicistat/tenofovir alafenamide/emtricitabine; INR: international normalized ratio; InSTIs: integrase strand transfer inhibitors; MATE 1: toxin extrusion proteins and multiple drugs; NRTIs: nucleoside/nucleotide reverse transcriptase inhibitors; OAT1/OAT3: organic anion transporters; OCT2: organic cation transporters; P-gp: P-glycoprotein; PrEP: pre-exposure prophylaxis; RAL: raltegravir; TDF: tenofovir disoproxil-fumarate; TDF/FTC: tenofovir disoproxil-fumarate/emtricitabine; TDF/FTC/EVG/COBI: tenofovir disoproxil-fumarate/emtricitabine/elvitegravir/cobicistat; TDF/FTC/RAL: tenofovir disoproxil-fumarate/emtricitabine/raltegravir; UGT1A1: uridinadifosfatoglucoronosiltransferasa; 3TC: lamivudine.
Table 5. Drug interaction pairs with evidence of absence of clinically relevant drug interactions (Level 5) systematically reported for the first time.
Table 5. Drug interaction pairs with evidence of absence of clinically relevant drug interactions (Level 5) systematically reported for the first time.
Pharmacological Group or Drugs AffectedAntiretroviral AgentComments/Suggestions
Anesthetic/anxiolytic/benzodiazepine
Midazolam
[168]
CABMidazolam 3 mg once daily administered with CAB 30 mg once daily only increases midazolam AUC and Cmax by 10% and 9%, respectively. Thus, CAB does not modify the pharmacokinetics of midazolam. It is a safe combination, therefore, does not require a dose adjustment.
Antimalarial
Chloroquine
[169]
EFVEfavirenz-based ART and chloroquine 150 mg daily is well tolerated and EFV does not change chloroquine plasma levels. Thus, this combination is safe and it does not require a dose adjustment.
Antimicrobial/antituberculosis
Cotrimoxazole (trimethoprim and sulfamethoxazole)
[21,170]
NVPNo significant changes in plasma levels of NVP and clotrimazole are generated. This combination is safe and does not require a dose adjustment.
Levofloxacin
[171]
EFV, NVPNFV or EFV do not affect levofloxacin AUC and Cmax; additionally, there is no significance in the NVP or EFV plasma levels with and without levofloxacin. Therefore, these combinations are safe and dose adjustments are not needed.
Amikacin, ethambutol, ethionamide, pyrazinamide, terizidone [172]LPV/RTVCoadministration of antituberculosis drugs (high-dose isoniazid, pyrazinamide, ethambutol, ethionamide, terizidone, and amikacin) and LPV/RTV in HIV-infected children generates minor changes both in AUC and Cmax of antituberculosis drugs and LPV/TRV. These combinations are safe and dose adjustments are not needed.
Rifabutin
[21,173]
ATV/RTVNo significant changes in plasma levels of ATV/RTV (330/110 mg daily) and rifabutin (300 mg thrice-weekly or 150 mg daily doses) are generated. The combination of ATV/RTV (330/110 mg daily) and rifabutin (300 mg thrice weekly or 150 mg daily doses) is safe.
Rifabutin [21,127,174] CAB (oral)Rifabutin has a minor effect on AUC and Cmax CAB, resulting in CAB plasma levels to maintain viral suppression in HIV-1-infected persons. Thus, rifabutin and oral CAB can be coadministered without dose adjustments.
Rifampicin
[21,175,176]
DTGDTG (50 mg twice daily) in patients on rifampicin-based treatment is effective and safe. Therefore, among ART-naive patients with HIV on rifampicin-based tuberculosis treatment, DTG-based ART (50 mg twice daily) is effective and safe.
Attachment inhibitor
Fostemsavir
[177,178,179]
ETR/DRV/RTVFostemsavir 600 mg twice daily combined with ETR 200 mg twice daily causes a decrease in AUC of temsavir (active drug) by 50%. However, when fostemsavir and ETR are combined with DRV/RTV 600/100 mg twice daily, the temsavir AUC increases by 34%. Thus, the coadministration of fostemsavir/etravirine/DRV/RTV 600/200/600/100 mg twice daily does not need a dose adjustment and it is effective and safe, which has been evidenced from phase IIb/III studies. Thus, this combination has been approved for patients with HIV not able to be treated with other options.
Direct-acting antivirals
Elbasvir/grazoprevir
[180]
DTG or RALThe coadministration of elbasvir/grazoprevir with RAL or DTG does not produce relevant clinical drug interactions and it is overall well tolerated. These results support the use of these combinations without dose adjustments for elbasvir, grazoprevir, RAL, or DGV in HCV/HIV-coinfected persons.
Simeprevir
[181]
DTGSimeprevir causes a slight increase in the AUC by 15% of DTG; however, it is bioequivalent and DTG plasmatic levels are within the safely established therapeutic range. Thus, simeprevir and DTG can be safely coadministered without dose adjustments.
Ledipasvir/sofosbuvir
[182]
FTC/RPV/TAFAlthough the Cmax and AUC of tenofovir (main metabolite of TAF) increase by 62% and 75%, respectively, the resulting absolute tenofovir exposures are noticeably inferior to the historical tenofovir exposures following TDF; as a consequence, this increase in tenofovir is not considered to be clinically relevant. Therefore, fixed-dose combinations of ledipasvir/sofosbuvir 90/400 can be coadministered with fixed-dose combinations of FTC/RPV/TAF 200/25/25 mg for HIV without the need for dosage adjustments.
Disease
Mild to moderate hepatic impairment (a Child–Pugh score of 7–9)
[157,183,184,185,186,187,188]
BIC, CABO, DTG, EVG/COBI, RALModerate liver impairment does not affect plasma InSTIs exposure compared with persons without hepatic impairment. Thus, InSTIs may be administered without a dose adjustment in patients with moderate liver impairment.
Mild to severe renal impairment (estimated creatinine clearance
<30–60 mL/min) [157,183,184,188,189,190,191,192]
BIC, CAB, DTG, EVG/COBI, RALSevere renal impairment does not affect plasma InSTIs exposure compared with persons without severe renal impairment. Thus, these drugs may be administered without a dose adjustment in patients with renal impairment, including patients on chronic hemodialysis (mainly for CAB and EVG/COBI). However, when TDF is coformulated with any InSTIs, its use may be limited due to TDF, caution should be taken in persons with renal insufficiency.
Foods/supplements
Food (standard breakfast and protein-rich drink)
[193,194,195]
EVG/COBI/FTC/TAFEVG/COBI/FTC/TAF administration under fasted conditions is associated with a decrease by 50% and 57% in the mean AUC and Cmax of EVG, respectively. Thus, EVG/COBI/FTC/TAF should be administered orally once daily with food, for instance, a standard breakfast (413 kcal, 10% fat: two slices of bread with strawberry jam, one boiled egg, and 160 g of grape juice). It is important to denote that oral EVG/COBI/FTC/TAF administration once daily with a nutritional protein-rich drink generates similar EVG/COBI exposure as administration with standard breakfast. Regardless, food or a nutritional protein-rich drink did not affect the bioavailability of EVG/COBI/FTC/TAF.
Injectable contraceptive
Depot medroxyprogesterone
[196]
TDF (1% gel)TDF 1% vaginal gel administered topically for HIV-1 prevention in women resulted in high and protective mucosal concentrations with or without use of depot medroxyprogesterone (DMPA) or oral contraceptives. TDF diphosphate (intracellular active metabolite of TDF) reached higher vaginal tissue concentrations several times above estimated protective levels in women using DMPA. This combination is safe and does not require a dose adjustment.
NNRTI
Etravirine
[176,197]
CABIn total, 30 mg of CAB for 10 days followed by the coadministration of ETR 200 mg twice daily for 14 days does not cause significantly changes in plasma levels for either drug. Thus, CAB may be administered with ETR without a dosage adjustment for either agent.
Etravirine
[176,198,199]
RALETR and RAL have no clinically significant pharmacokinetic interactions; therefore, ETR and RAL plasmatic levels are acceptable in most patients. The virologic efficacy and therapeutic success rates at week 48 are 99.4% and 94.5%, respectively; results are similar at week 96. Additionally, the combination is generally well tolerated (most drug-related adverse clinical experiences are minor and generally temporary in nature). Thus, etravirine 200 mg/RAL 400 mg twice daily is an effective and safe dual therapy. However, the coadministration of etravirine with RAL 1200 mg daily should be avoided due to a decrease in the Cmin of RAL near to 34%.
Oral contraceptive
Ethinylestradiol/Levonorgestrel
[40,200]
CABThere are no significant changes in plasma levels of CAB and ethinylestradiol/levonorgestrel containing oral contraceptive. Additionally, repeat doses of oral CAB do not have a significant effect on ethinylestradiol/levonorgestrel plasma levels or effects, which supports that this combination is safe and requires no dose adjustment.
Ethinylestradiol/Levonorgestrel
[196]
TDF (1% gel)TDF 1% vaginal gel administered topically for HIV-1 prevention in women resulted in high and protective mucosal concentrations with or without the use of ethinylestradiol/levonorgestrel as an oral contraceptive. This combination is safe and does not require a dose adjustment.
Ethinylestradiol/norgestimate [201,202]TDFTDF and norgestimate-ethinyl estradiol coadministration shows a lack of clinically significant drug interactions. TDF does not cause significant changes to norgestimate or ethinyl estradiol plasmatic levels; similarly, norgestimate-ethinyl estradiol does not cause significant changes to TDF plasmatic levels. This combination is well tolerated and requires no dose adjustment.
Oral opioid analgesic
Methadone and buprenorphine
[203]
FTRFostemsavir and methadone or buprenorphine coadministration shows a lack of clinically significant drug interactions. Fostemsavir does not cause significant changes to both methadone or buprenorphine plasmatic levels, shown with standardized assessments for opiate withdrawal and overdose scale scores. This combination is well tolerated and can be administered without dose adjustments.
Special conditions
Adults ≥ 60 years
[157,204]
EVG/COBI/FTC/TAFSwitching to EVG/COBI/FTC/TAF, in virologically suppressed adults aged 60 years or older, is effective (virological suppression is maintained and there is a moderate increase in CD4 cell counts) and safe (lack of treatment-related serious adverse events and improved safety, both in bone mineral density and renal biomarkers) after 48 weeks.
Adults ≥ 65 years virologically suppressed
[157,158,159,160,161,162,163,164,165,166,167,168,169,170,171,172,173,174,175,176,177,178,179,180,181,182,183,184,185,186,187,188,189,190,191,192,193,194,195,196,197,198,199,200,201,202,203,204]
BIC/FTC/TAFSwitching to BIC/FTC/TAF, in virologically suppressed adults aged 65 years or older, is effective (97.7% and 90.7% virologic suppression at weeks 24 and 48, respectively) and safe (not grade 3–4, serious drug-related adverse events or deaths) after 48 weeks.
Statin
Pitavastatin
[205,206]
ATV/RTV
DRV/RTV,
LPV/RTV
Due to pitavastatin being marginally metabolized by the hepatic CYP3A4 enzyme, this statin can be used (4 mg daily) in the setting of a complex background ART and no clinically significant pharmacokinetic interactions have been noted with its coadministration with ATV/RTV, LPV/RTV, or DRV/RTV. Therefore, there are no significant changes in the plasma levels of PIs and pitavastatin; these combinations are safe and no dose adjustments are necessary.
Pitavastatin
[205,206]
DOR,
EFV,
ETR,
NVP,
RPV
Due to pitavastatin being marginally metabolized by the hepatic cytochrome P450 enzyme, this statin can be used (4 mg daily) in the setting of a complex background ART, and no clinically significant pharmacokinetic interactions have been noted with its coadministration with DOR, EFV, ETR, NVP, or RPV. Therefore, there are no significant changes in the plasma levels of these ARVs and pitavastatin; these combinations are safe and no dose adjustments are necessary.
Pravastatin
[205,206]
DOR,
NVP,
RPV
Due to pravastatin being marginally metabolized by the hepatic cytochrome P450 enzyme, this statin can be used (40 mg daily) in the setting of a complex background ART, and no clinically significant pharmacokinetic interactions have been noted with its coadministration with DOR, NVP, or RPV. Therefore, there are no significant changes in the plasma levels of these ARVs and pravastatin; these combinations are safe and no dose adjustments are necessary.
ABC: abacavir; ABC/AZT: abacavir/zidovudine; ABC/3TC: abacavir/lamivudine; ARV: antiretroviral; ATV/RTV: atazanavir/ritonavir; AZT: zidovudine; BIC: bictegravir; BIC/TAF/FTC: bictegravir/tenofovir alafenamide/emtricitabine; CAB: cabotegravir; DOR: doravirine; DRV: darunavir; DRV/RTV: darunavir/ritonavir; DTG: dolutegravir; DTG/ABC/3TC: dolutegravir/abacavir/lamivudine; EFV: efavirenz; ETR: etravirine; EVG: elvitegravir; EVG/COBI: elvitegravir/cobicistat; GAHT: gender-affirmation hormone therapy; HIV: human immunodeficiency virus; InSTIs integrase strand transfer inhibitors; LPV/RTV: lopinavir/ritonavir; NNRTIs: non-nucleoside reverse transcriptase inhibitors; NVP: nevirapine; PIs: protease inhibitors; RAL: raltegravir; RPV: rilpivirine; TAF: tenofovir alafenamide; TDF: tenofovir disoproxil-fumarate; TDF/FTC: tenofovir disoproxil-fumarate/emtricitabine; TDF/FTC/RPV: tenofovir disoproxil-fumarate/emtricitabine/rilpivirine; 3TC: lamivudine.
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

Amariles, P.; Rivera-Cadavid, M.; Ceballos, M. Clinical Relevance of Drug Interactions in People Living with Human Immunodeficiency Virus on Antiretroviral Therapy—Update 2022: Systematic Review. Pharmaceutics 2023, 15, 2488. https://doi.org/10.3390/pharmaceutics15102488

AMA Style

Amariles P, Rivera-Cadavid M, Ceballos M. Clinical Relevance of Drug Interactions in People Living with Human Immunodeficiency Virus on Antiretroviral Therapy—Update 2022: Systematic Review. Pharmaceutics. 2023; 15(10):2488. https://doi.org/10.3390/pharmaceutics15102488

Chicago/Turabian Style

Amariles, Pedro, Mónica Rivera-Cadavid, and Mauricio Ceballos. 2023. "Clinical Relevance of Drug Interactions in People Living with Human Immunodeficiency Virus on Antiretroviral Therapy—Update 2022: Systematic Review" Pharmaceutics 15, no. 10: 2488. https://doi.org/10.3390/pharmaceutics15102488

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