Dolutegravir

Dolutegravir/Lamivudine Single‑Tablet Regimen: A Review in HIV‑1 Infection
Lesley J. Scott1

© Springer Nature Switzerland AG 2019

Abstract
The oral once-daily, fixed-dose single-tablet regimen (STR) of dolutegravir/lamivudine (Dovato®), combining a second gen- eration integrase single-strand transfer inhibitor (INSTI) and a nucleoside reverse transcriptase inhibitor (NRTI), is indicated as a complete regimen for the treatment of HIV-1 infection in adults and adolescents (> 12 years of age weighing at least 40 kg) with no known or suspected resistance to the INSTI class or lamivudine. In GEMINI trials in antiretroviral therapy (ART)-naïve HIV-1-infected adults, treatment with dolutegravir plus lamivudine provided rapid and sustained virological suppression and was noninferior to dolutegravir plus tenofovir disoproxil fumarate/emtricitabine at 48 weeks, irrespective of baseline patient or disease characteristics. Virological suppression was sustained at 96 weeks in these ongoing trials. In patients with HIV-1 with sustained virological suppression on their current tenofovir alafenamide (AF)-based ART regimen (≥ 3 drugs), switching to treatment with dolutegravir/lamivudine was noninferior to continuing on a tenofovir AF-based regimen at 48 weeks in the ongoing TANGO trial. No resistance mutations to dolutegravir or lamivudine were detected in patients who met criteria for confirmed virological withdrawal in GEMINI and TANGO trials. Hence, the dolutegravir/ lamivudine STR is an effective, generally well tolerated and convenient initial and subsequent ART option for adolescents and adults with HIV-1 infection with no known or suspected resistance to the INSTI class or lamivudine.

⦁ Introduction
Antiretroviral combination therapy (ART) has revolution- ized the management of and clinical outcomes in patients with HIV-infection, transforming HIV infection into a manageable chronic condition and significantly reducing

Enhanced material for this Adis Drug Evaluation can be found at https://doi.org/10.6084/m9.figshare.9947255

The manuscript was reviewed by: J. Arribas, Infectious Diseases Unit, Department of Internal Medicine, La Paz University Hospital, Madrid, Spain; R. de Miguel, Infectious Diseases Unit, Department of Internal Medicine, La Paz University Hospital, Madrid, Spain; F. Gutierrez, Department of Internal Medicine,
Division of Infectious Diseases and HIV Unit, Hospital General of Elche and University of Miguel Hernández, Alicante, Spain;
R. Montejano, Infectious Diseases Unit, Department of Internal Medicine, La Paz University Hospital, Madrid, Spain; M. Nelson, Chelsea and Westminster Hospital, London, UK.

 Lesley J. Scott [email protected]
1 Springer Nature, Private Bag 65901, Mairangi Bay, Auckland 0754, New Zealand
HIV-associated morbidity and mortality [1–4]. Indeed, ART has meant achievement of sustained virological suppression (primary goal of therapy) is attainable by nearly all patients infected with HIV-1, with virological suppression central to recovery of immune function, optimization of health and decreasing the risk of HIV transmission and emergence of resistance [1–4].
Given the current requirement for life-time treatment, optimization and simplification of ART regimens utiliz- ing single-tablet regimens (STRs) combining two or more antiretroviral drug classes is an important tool for achieving sustained virological suppression, whilst improving health- related quality of life and potentially reducing ART-related toxicity and costs [3, 5, 6]. Adherence to ART is a critical factor in achieving and sustaining virological suppression, with STRs reducing the pill burden and complexity of regi- mens, and probably improving adherence [5–7]. Further de- intensification of ART from a three-drug to a complete two- drug regimen represents a recent advance in ART therapy [3, 5, 8].
Integrase single-strand transfer inhibitors (INSTIs) rep- resent the latest class of ART, with the first generation INSTIs raltegravir and elvitegravir providing highly effective

virological suppression but exhibiting a low genetic barrier to resistance. Most resistance mutations confer a high-degree of cross-resistance amongst other first-generation INSTIs, but not to second-generation INSTIs (e.g. dolutegravir and bictegravir) [9]. With its high genetic barrier to resistance, favourable pharmacokinetic profile and generally good tol- erability profile, dolutegravir is an option for use in STRs. The oral once-daily, fixed-dose dolutegravir/lamivudine STR (Dovato®) is a complete two-drug regimen approved in several countries, including those of the EU [10] and in the USA [11], for the treatment of HIV-1 infection in adolescents and adults with no known or suspected resistance to the INSTI class or lamivudine; specific indications may vary between individual countries. This article, written from EU perspective, reviews the clinical efficacy and tolerability of dolutegravir plus lami- vudine in the management of HIV-1 infection and summarizes relevant pharmacology.

⦁ Pharmacodynamic Properties of Dolutegravir/Lamivudine
Dolutegravir, a second generation HIV-1 INSTI, binds to the active site of HIV integrase, which inhibits the strand transfer step of retroviral DNA integration (an essential step in the HIV replication cycle) [10, 12, 13]. Lamivudine (a nucleoside reverse transcriptase inhibitor; NRTI), via its active metabolite 5′-triphosphate, inhibits the reverse tran- scriptase of HIV-1 and HIV-2 through incorporation of the monophosphate form into the viral DNA, resulting in chain termination [10]. Lamivudine triphosphate exhibits signifi- cantly less affinity for host cell DNA polymerases than HIV reverse transcriptase [10].
⦁ Antiretroviral Activity

The antiretroviral activity of dolutegravir and lamivudine against HIV-1 is well established, with both drugs shown to inhibit replication of laboratory strains and clinical isolates of HIV in a number of cell types, including transformed T cells, monocyte/macrophage-derived lines and primary cultures of activated peripheral blood mononuclear cells (PMBCs) and monocytes/macrophages [10, 11, 13–15]. The concentration of active drug at which 50% of viral replication is inhibited (i.e. IC50) varied according to virus and host cell type [10]. The IC50 for dolutegravir against various laboratory strains using PBMCs was 0.5 nmol/L, with IC50s of 0.7–2 nmol/L in MT-4 cells. Similar IC50s for dolutegravir were observed against clinical isolates with no major difference between subtypes; in a panel of 24 HIV-1 isolates of clades A-G and group O, the mean IC50 was 0.2 nmol/L and that against three HIV-2 isolates was 0.18 nmol/L. For lamivudine, the mean or median IC50s against laboratory strains of HIV-1 ranged from 0.007 to 2.3 μmol/L and the mean IC50s against labora- tory strains of HIV-2 (LAV2 and EHO) ranged from 0.16 to
0.51 μmol/L. In PBMCs, the range of IC50s for lamivudine
against HIV-1 isolates of clades A-G, group O and HIV-2 isolates were 0.001–0.170 μmol/L, 0.030–0.160 μmol/L and 0.002–0.120 μmol/L, respectively [10].
The shift in potency for dolutegravir in the presence of 100% human serum was 75-fold, resulting in a protein- adjusted concentration at which 90% of replication was inhibited of 0.064 μg/mL [10].
An indirect comparison of clinical trials indicated that there was no difference in the rates of viral decay in ART- naïve patients receiving dolutegravir plus lamivudine (ATG A5353; Sect. 4.1.1) compared with patients receiving a dolutegravir-based three-drug regimen (SPRING-1 and SIN- GLE trials), including in those with a baseline viral load of
> 100,000 copies/mL [16].
Dissociation of dolutegravir from the wild-type integrase- DNA complex was significantly slower than for the first generation INSTIs raltegravir and elvitegravir (mean dis- sociation rate constant 2.7 × 10−6 vs 22 × 10−6 and 71 × 10−6, respectively) [17]. The rate of dissociation for dolutegravir was also slower (5- to 40-fold) than first-generation INSTIs for mutant integrases with single amino acid substitutions. For wild-type integrase, the calculated dissociative half-lives for dolutegravir, raltegravir and elvitegravir were 71 h, 8.8 h and 2.7 h, respectively [17]. Based on molecular structural modelling studies, it has been suggested that this prolonged binding of dolutegravir to the integrase-DNA complex may contribute to its high genetic barrier to resistance and its distinct in vitro drug resistance profile versus raltegravir and elvitegravir (Sect. 2.2) [18].

Neither dolutegravir or lamivudine were antagonist to all tested ART drugs [11]. In vitro, dolutegravir exhibited mostly synergistic activity in combination with representa- tive agents of other antiretroviral drug classes [15]. The activity of dolutegravir was not affected by combination with adefovir or ribavirin [drugs coadministered with other antiretroviral agents in HIV patients with concomitant hepa- titis B virus (HBV) and hepatitis C virus (HCV) infection, respectively] [15].
⦁ Resistance

Dolutegravir has a high genetic barrier to resistance devel- opment in vitro (reviewed previously in Drugs [12]) [15]. In vitro in serial passage experiments with wild-type HIV-1 IIIB, the presence of dolutegravir did not result in the emer- gence of mutations that reduced susceptibility to the drug more than fivefold. The only integrase amino acid substitu- tion mutations (single or combination) to occur were T124A, S153Y, T124A/S153F, T124A/S153Y and L110I/T124A/
S153F, with the S133Y and S153F mutations associated with highest fold change in IC50 (fourfold) [15]. These muta- tions were not selected in patients treated with dolutegravir in clinical studies [19]. The key resistance mutations for raltegravir (Y143C/H/R, Q148H/R/K and N155H/T) were not selected by serial passage with dolutegravir [15].
R236K, G118R, N155H and Q148H/R/K were the four most commonly selected INSTI resistance mutations in vitro studies and clinical isolates from dolutegravir recipients, based on a systematic review [20]. In INSTI-naïve patients receiving dolutegravir, R236K and G118R were the most common of these mutations and reduced susceptibility to dolutegravir approximately twofold and >fivefold, respec- tively. The highest levels of resistance to dolutegravir occurred in isolates with N155H plus Q148H/R or ≥ 2 addi- tional mutations (reduced susceptibility by 2- to > 15-fold), R236K plus G118R, N155H or Q148R (by 10- to 15-fold), Q148R plus G140A/S (by 5- to 10-fold) and Q148K plus E138K (by 10- to 20-fold) [20].
HIV-1 resistance to lamivudine involves the development of a M184V or M184I amino acid substitution close to the active site of the viral reverse transcriptase, which arises both in vitro and in HIV-1 patients treated with lamivudine- containing ART [11]. Both of these mutations confer high- level resistance to lamivudine.
In phase 3 trials of dolutegravir plus lamivudine dis- cussed in Sect. 4.1, there were no treatment-emergent INSTI or NRTI resistance mutations detected in patients experi- encing protocol-defined confirmed virological withdrawal (CVW) in ART-naïve (GEMINI trials at week 48 [21] and 96 [22]) or ART-experienced (TANGO at week 48 [23]) patients, based on genotypic testing of samples at the time of CVW versus baseline samples. In GEMINI trials, there
were six CVW occurrences in the dolutegravir plus lami- vudine group (vs four patients in the three-drug regimen group) during the initial 48 weeks of treatment [21], with CVW occurring in 11 and 7 patients by week 96 [22]. One 48-week plasma sample collected at the time of CVW was not evaluable in the integrase gene assay. Participants met CVW criteria if a second and consecutive HIV-1 RNA value met any of the following definitions: decrease from baseline of < 1 log10 copies/mL, unless the HIV-1RNA was < 200 copies/mL, by week 12; confirmed plasma HIV-1 RNA of
≥ 200 copies/mL at or after week 24; or confirmed rebound (HIV-1 RNA ≥ 200 copies/mL after confirmed consecutive HIV-1 RNA < 200 copies/mL) [21]. In ART-experienced patients, there were no CVW occurrences in the dolutegra- vir/lamivudine group (vs one patient in the tenofovir alafena- mide (AF)-based 3–4 drug regimen group) during the initial 48 weeks of treatment in TANGO [23]. CVW was defined as one assessment with HIV-1 RNA ≥ 200 copies/mL after day 1 with an immediately prior HIV-1 RNA of ≥ 50 copies/mL [23]. In ART-naïve patients with HIV-1 receiving dolutegra- vir plus two NRTIs in phase 2b and 3 trials, no development of resistance to the INSTI class or NRTI class was observed (n = 1118, with a follow-up of 48–96 weeks) [10].

⦁ Cross‑Resistance

In vitro, dolutegravir was highly active against most HIV-1 strains displaying phenotypic and/or genotypic resistance to raltegravir, with fold increases in the IC50 of dolutegravir against raltegravir-resistant strains very low except for cer- tain strains with multiple integrase mutations [11, 13–15, 17, 19]. Against 60 INSTI-resistant site-directed mutant HIV-1 viruses (28 with a single substitution and 32 with two or more substitutions), single INSTI-resistance substi- tutions T66K, I151L and S153Y conferred a greater than twofold decrease in dolutegravir susceptibility (range 2.3- to 3.6-fold) and combination substitutions T66K/L74M, E92Q/N155H, G140C/Q148R, G140S/Q148H or R or K,
Q148R/N155H, T97A/G140S/Q148; substitutions at E138/ G140/Q148 also conferred a greater than twofold decrease in dolutegravir susceptibility (range 2.5- to 21-fold) [11]. Of 705 raltegravir resistant clinical isolates, dolutegravir exhibited a ≤ 10 fold-change in susceptibility against 94% of these isolates [19].
In a French national study of 502 HIV-1 infected patients failing a raltegravir-containing regimen, a high proportion of viruses remained susceptible to dolutegravir, with resist- ance to dolutegravir, elvitegravir and raltegravir predicted in 13.9%, 34% and 38.6% of patients, respectively [24].
Cross-resistance conferred by the M184V or M184I (confers high resistance to lamivudine; Sect. 2.2) has been observed within the NRTI class of antiretroviral agents

[11]. These substitutions confer cross-resistance to emtric- itabine, which selects M184V/M184I, and to abacavir when additional reverse transcriptase substitutions K65R, L74V and Y115F are present. Zidovudine maintains its antiviral activity against lamivudine-resistant HIV-1, with abacavir and tenofovir maintaining viral activity against lamivudine- resistant HIV-1 harbouring only the M184V or I substitution [11].
⦁ Other Effects

At a supratherapeutic doses (≈ 3-fold higher), dolutegravir had no clinically relevant effects on the corrected QT inter- val [10]. A similar study was not conducted with lamivudine.

⦁ Pharmacokinetic Properties of Dolutegravir/Lamivudine
Bioequivalence was established for the single-tablet, fixed- dose combination of dolutegravir/lamivudine and corre- sponding strength tablets of dolutegravir and lamivudine administered concurrently in healthy adults under fasted conditions [10].
Dolutegravir and lamivudine were rapidly absorbed after oral administration, with the absolute bioavailability of dolutegravir not yet established and lamivudine having an absolute bioavailability of ≈ 80–85% [10]. In the fasted state, respective maximum plasma concentrations of dolute- gravir and lamivudine were attained in a median of 2.5 h and 1.0 h. Systemic exposure to dolutegravir was generally similar between healthy adults and patients infected with HIV-1. There were no clinically relevant effects of food on the pharmacokinetics of dolutegravir/lamivudine [10].
The apparent volume of distribution of oral dolutegravir was 17–20 L and that for intravenous lamivudine was 1.3 L/ kg [10]. Dolutegravir is highly (> 99%) bound to human plasma protein in vitro; it was not substantially associated with blood cellular components. Lamivudine displayed lim- ited protein binding in vitro (< 16–36%). Dolutegravir and lamivudine distribute into cerebrospinal fluid; the concentra- tion of dolutegravir is above the IC50 and comparable to the unbound plasma concentration. The extent of lamivudine penetration into the CNS and its relationship to the efficacy of the drug is unknown. Dolutegravir is present in the female and male genital tract, with exposure in cervicovaginal fluid, cervical tissue and vaginal tissue 6–10% of the correspond- ing steady-state plasma exposure. In semen and rectal tis- sue, exposure was 7% and 17% of the corresponding plasma exposure at steady state [10].
Dolutegravir is primarily metabolized by UGT1A1, with a minor contribution by CYP3A4 (in humans, 9.7% of the total dose) [10]. Of the total dolutegravir dose, 53% was
excreted as unchanged drug in the faeces, with renal elimi- nation of the unchanged drug accounting for < 1% of the dose. Metabolism is a minor route of elimination of lami- vudine (hepatic metabolism accounts for 5–10%), with the drug primarily eliminated as parent compound in the urine. Respective terminal elimination half-lives of dolutegravir and lamivudine were ≈ 14 h and 18–19 h, with a terminal intracellular half-life of lamivudine-triphosphate of 16–19 h [10].
Pharmacokinetic data for dolutegravir and lamivudine in special patient populations were obtained separately [10]. The pharmacokinetics of dolutegravir and lamivudine were not altered to a clinically relevant extent by age (adolescents and adults; data in patients aged > 65 years are limited), gender or race. With dolutegravir, the presence of HCV co- infection or renal impairment also had no clinically relevant impact on its pharmacokinetics. Although the pharmacoki- netics of dolutegravir has not been studied in patients on dialysis, no differences in exposure are expected. Given its renal elimination, exposure to lamivudine is increased in patients with renal impairment; hence, dolutegravir/lami- vudine is not recommended for patients with a creatinine clearance (CRCL) of < 50 mL/min. There is no clinically relevant impact of hepatic dysfunction on the pharmacoki- netics of lamivudine. The pharmacokinetics of dolutegravir in patients with moderate hepatic impairment (Child Pugh Class B) were not significantly different from those in sub- jects with normal hepatic function. No data are available on the pharmacokinetics of dolutegravir in patients with severe hepatic impairment (Child Pugh Class C); therefore, dolutegravir/lamivudine should be used with caution in these patients [10].
⦁ Potential Drug Interactions of Dolutegravir/ Lamivudine

No drug interaction studies have been conducted with dolutegravir/lamivudine; any interactions identified for the individual components are relevant to dolutegravir/lami- vudine [10]. No clinically significant drug interactions are expected between dolutegravir and lamivudine. Local pre- scribing information, including those for the individual com- ponents of dolutegravir/lamivudine, should be consulted for detailed information relating to potential drug interactions, including dosage adjustments and contraindications.
In vitro, dolutegravir did not directly or weakly (IC50
> 50 μmol/L) inhibited CYP1A2, CYP2A6, CYP2B6, CYP2C8, CYP2C9, CYP2C19, CYP2D6, CYP3A4,
UGT1A1 or UGT2B7, or the transporters, P-glycopro- tein (P-gp), BCRP, BSEP, OATP1B1, OATP1B3, OCT1,
MATE2-K, multidrug resistance-associated (MRP) 2 or MRP4 [10]. In vitro, dolutegravir did not induce CYP1A2, CYP2B6 or CYP3A4. Based on this in vitro

data, dolutegravir is not expected to affect the plasma concentrations of medicinal products that are substrates of these major enzymes or transporters [10].
Lamivudine did not inhibit or induce CYP enzymes such as CYP3A4, CYP2C9 or CYP2D6 in vitro, and dem- onstrated weak or no inhibition of OATP1B1, OATP1B3, OCT3, BCRP, P-gp, MATE1 or MATE2-K [10]. Several
properties/features of lamivudine mean that the drug has a low potential for drug-drug interactions and is not expected to affect the plasma concentrations of medicinal products that are substrates of these enzymes or transporters [10].
Dolutegravir is a substrate of UGT1A3, UGT1A9, CYP3A4, P-gp, and BCRP; hence, coadministration of dolutegravir/lamivudine and other medicinal products that inhibit these enzymes may increase dolutegravir plasma concentrations [10]. Conversely, medicinal products that induce these enzymes (including etravirine, efavirenz, nevirapine, boosted tipranavir, rifampicin, phenobarbital, phenytoin, oxcarbazepine, St. John’s wort) or transport- ers may decrease dolutegravir plasma concentrations and reduce its therapeutic effect.
Absorption of dolutegravir is reduced by certain metal cation-containing antacids and supplements (e.g. calcium supplements, iron supplements or multivitamins). There- fore, these preparations should be taken a minimum of 6 h before or 2 h after the administration of dolutegravir. When taken with food, dolutegravir/lamivudine and sup- plements or multivitamins containing calcium, iron and magnesium may be taken together [10].
Dolutegravir inhibits OCT2 and MATE1, and may increase the plasma concentrations of drugs that require these renal transporters for excretion (e.g. metformin) [10]. Exposure to metformin is increased when it is co- administered with dolutegravir; to maintain glycaemic control, dosage adjustment of metformin should be con- sidered when starting or stopping concomitant adminis- tration with dolutegravir [10]. In patients with moder- ate renal impairment, a dosage adjustment of metformin should be considered when coadministered with dolute- gravir, because of the increased risk of lactic acidosis in this patient population due to increased metformin con- centrations [10].

⦁ Therapeutic Efficacy of Dolutegravir/ Lamivudine
⦁ In Clinical Trials
⦁ In Antiretroviral Therapy (ART)‑Naïve Adults

The efficacy of once-daily dolutegravir 50 mg plus lami- vudine 300 mg (administered as separate tablets) in
ART-naïve patients with HIV-1 infection was evaluated in two identically designed, ongoing, 144-week, randomized, double-blind, multinational, noninferiority trials (GEM- INI-1 and GEMINI-2 [21, 22]), with data available at 48 weeks (primary analysis) [21] and 96 weeks (abstract) [22]. These pivotal phase 3 trials are supported by data from the 48-week proof-of-concept PADDLE trial (n = 20)
[25] and the 52-week, single-arm, phase 2 ACTG A5353 trial (n = 120) [26, 27], which are not discussed further.
In GEMINI trials, key eligibility criteria included aged ≥ 18 years, plasma HIV-1 RNA loads of 1000–500,000 copies/mL at screening, received ≤ 10 days of prior ART, no evidence of pre-existing viral resistance based on the presence of any major resistance mutation to a protease inhibitor (PI), non-nucleoside reverse tran- scriptase inhibitor (NNRTI) or NRTI, and no HBV infec- tion or need for HCV therapy [21]. Randomization of patients to study regimen was stratified by HIV-1 RNA load (≤ 100,000 vs > 100,000 copies/mL) and CD4+ cell counts (≤ 200 vs > 200 cells/μL). In the intent-to- treat-exposed (ITTe) pooled population, the median age of patients was 33 years, 80% of patients had an HIV-1 RNA load of ≤ 100,000 copies/mL and 8% had a CD4+ cell count of ≤ 200 cells/μL. There were no significant between-group differences in terms of key demographic and baseline clinical characteristics. The primary endpoint was the proportion of patients with plasma HIV-1 RNA of < 50 copies/mL at 48 weeks in the ITTe population, assessed using the FDA Snapshot algorithm [21].
In GEMINI-1 and -2, dolutegravir plus lamivudine
treatment was noninferior to a triple regimen of dolute- gravir plus tenofovir disoproxil fumarate (DF)/emtricit- abine in terms of establishing virological suppression at 48 weeks, with a high proportion of patients in both treat- ment groups achieving a viral load of < 50 copies/mL in ITTe analyses (primary endpoint; Table 1) [21]. Virologi- cal efficacy in terms of this outcome in the pooled analysis of the ITTe population was consistent with those in the individual trials (Table 1). Results from per-protocol (PP) analyses were also consistent with those in ITTe analyses, with most patients (92–95%) in the two-drug and three- drug regimen groups in individual trials and the pooled analysis achieving a viral load of < 50 copies/mL. The greatest rate of HIV-1 RNA decline occurred in the first 4 weeks of treatment, irrespective of the study regimen, with the majority of patients achieving an HIV-1 RNA load of < 50 copies by this timepoint (median time to viral suppression 29 days in both groups). These findings were not influenced by baseline patient or disease characteris- tics, including HIV-1 RNA load, HCV serostatus, age, sex and ethnicity. In patients with baseline CD4+ counts of
≤ 200 cells/μL, virological response rates in the two-drug and three-drug groups were 79% and 93% (n = 63 and 55)

Table 1 Efficacy of the dolutegravir plus lamivudine in pivotal phase 3 trials in treatment-naïve adults with HIV-1 infection

Study Regimena (no. of ITTeb pts) Plasma HIV-1 RNA < 50 copies/mLc (% of pts) [TDd; 95% CI]

48 weekse [21] 96 weeksf [22]
GEMINI-1 DTG + 3TC (356) 90 [− 2.6; − 6.7 to 1.5] NI 84 [− 4.9; − 9.8 to 0.0] NI
DTG + TDF/FTC (358) 93 89
GEMINI-2 DTG + 3TC (360) 93 [− 0.7; − 4.3 to 2.9] NI 88 [− 1.8; − 6.4 to 2.7] NI
DTG + TDF/FTC (359) 94 90
Pooled analysis DTG + 3TC (716) 91 [− 1.7; − 4.4 to 1.1] NI 86 [− 3.4; − 6.7 to 0.0] NI
DTC + TDF/FTC (717) 93 90
3TC lamivudine 300 mg, DTG dolutegravir 50 mg, FTC emtricitabine 200 mg, ITTe intent-to-treat-exposed population, NI noninferior, pts
patients, TD adjusted treatment difference, TDF tenofovir disoproxil fumarate 300 mg
aAdministered orally once daily; 3TC and TDF/FTC tablets were over-encapsulated to match each other and maintain blinding
bAll pts who received ≥ 1 dose of study medication
cUsing the FDA Snapshot algorithm (missing, switch or discontinuation = failure)
dCochran–Mantel–Haenszel-stratified analysis adjusted for baseline viral load (≤ 100,000 vs > 100,000 copies/mL) and CD4+ cell counts (≤ 200 vs > 200 cells/μL) in individual trials (predefined noninferiority margin − 10%); pooled analysis stratified by trial (no predefined margin) ePrimary endpoint
fAbstract presentation

and those in patients with a CD4+ count of > 200 cells/μL were both 93% (n = 653 and 662). The reasons for Snap- shot-defined virological failure in those with a baseline CD4+ counts of ≤ 200 cells/μL did not relate to efficacy or treatment failure [21]. In pooled post hoc analyses, there was also no between-group difference in the incidence of blips during 48 weeks’ treatment (14% of patients in both groups); a blip was defined as a viral load between 50–200 copies/mL and no viral load of ≥ 200 copies/mL after viral suppression to < 50 copies/mL [28].
Viral suppression (i.e. HIV RNA load < 50 copies/mL) was sustained at week 96 in ITTe analyses of the individual trials and pooled data (Table 1), with no treatment-emergent resistance observed [22].
The virological efficacy of dolutegravir plus lamivudine in ART-naïve adults with HIV-1 is supported by evidence from a network meta-analysis of 14 randomized controlled trials (RCTs; n = 10,043) [29]. At 48 weeks, the efficacy of dolutegravir plus lamivudine was generally similar to that of traditional three-drug regimens, including a regimen of dolutegravir plus tenofovir DF/emtricitabine. However, dolutegravir plus lamivudine provided significantly better virological efficacy than a triple regimen of efavirenz plus tenofovir DF/emtricitabine at 48 weeks, with a mean treat- ment difference for the proportion of patients achieving a viral load of < 50 copies/mL of − 7.3% (95% CI − 13.8 to
− 0.8) [29].
⦁ In ART‑Experienced Adults

The ongoing, 200-week, randomized, open-label, multina- tional, phase 3, noninferiority TANGO trial is evaluating
the efficacy of switching adults with HIV-1 infection with virological suppression (HIV-RNA < 50 copies/mL) for
≥ 6 months on a ≥ 3-drug tenofovir alafenamide (AF) regi- men to once-daily dolutegravir/lamivudine (50/300 mg) [23, 30]. These data are supported by evidence from earlier phase 1 and 2 trials (DOLAM [31], LAMIDOL [32], ASPIRE
[33]), which are not discussed further.
TANGO includes a screening phase of up to 28 days, a randomized early switch phase (day 1 to week 148), a late switch phase (week 148–200) and a continuation phase (post week 200) [23]. Randomization of patients to study regi- men was stratified by baseline third agent class (PI, INSTI or NNRTI) in the tenofovir AF-based regimen [30]. Key eligibility criteria were ≥ 2 documented HIV-1 RNA meas- urements of < 50 copies/mL, no HBV infection or need for HCV therapy, no prior virological failure and no docu- mented NRTI or INSTI resistance mutations, and receiving tenofovir AF/emtricitabine plus a PI or INSTI or NNRTI as their initial regimen (participants with initial tenofovir DF treatment who switched to tenofovir AF ≥ 3 months before screening, with no changes to other drugs in their regimen, were also eligible) [23]. At baseline in the ITTe population, the median age of patients in the dolutegravir/lamivudine and tenofovir AF-based regimen groups was 40 and 39 years (n = 369 and 372), the mean CD4+ cell count was 682 cells/ μL and 720 cells/μL (91% and 92% of patients had counts of
≥ 350 cells/μL), the third class of agent was an INSTI (78% and 80%), NNRTI (14% and 13%) or PI (8% and 8%), and the median duration of ART before day 1 was 33.8 months and 35.1 months. The primary endpoint was the proportion of patients with plasma HIV-1 RNA of ≥ 50 copies/mL (i.e. FDA Snapshot virological failure) at 48 weeks in the ITTe

population, assessed using the FDA Snapshot algorithm [23].
At 48 weeks, switching to dolutegravir/lamivudine was noninferior to continuing on a tenofovir AF-based regi- men in terms of the virological failure rate, with very few patients (< 1%) in either group having an HIV RNA load of
≥ 50 copies/mL (primary endpoint; Table 2) [23]. Dolute- gravir/lamivudine treatment was also noninferior to con- tinuing a tenofovir AF-based regimen for the proportion of patients with HIV RNA load of < 50 copies/mL at 48 weeks (key secondary endpoint), with the vast majority of patients (93%) in both groups maintaining virological suppression (Table 2) [23]. Findings at the interim 24-week analysis also demonstrated that switching to dolutegravir/lamivudine was noninferior to remaining on a tenofovir AF-based regimen for these two virological efficacy outcomes (Table 2) [30].
⦁ In Real‑World Studies

The virological efficacy of switching to dolutegravir plus lamivudine in HIV-1 infected adults with durable viro- logical suppression on ≥ 3-drug ART regimens in several real-world studies (n = 70–494) [34–39] was consistent with that observed in phase 3 trials in ART-experienced patients (Sect. 4.1.2). These real-world studies were of ret- rospective [34, 36, 39] or prospective [35, 37, 38] design,
single-centre [34, 36] or multicentre [35, 37–39], and
conducted in Belgium [34], Italy [36–38], Spain [39] or
Switzerland [35].
For example, in the largest retrospective study in ART- experienced patients, switching from a ≥ 3 drug ART regi- men (mean duration of ART 8 years) to dolutegravir plus lamivudine (n = 183) provided similar maintenance of virological efficacy at week 48 and 96 as switching to dual
regimens of lamivudine plus a boosted PI (atazanavir/r or darunavir/r; n = 141 and 170) in ART-experienced patients with virological suppression [36]. Of note, switching to dolutegravir plus lamivudine was associated with a sig- nificantly (p < 0.001) lower risk of treatment discontinua- tion than switching to lamivudine plus atazanavir/r [haz- ard ratio (HR) 0.19; 95% CI 0.10–0.39) or lamivudine plus darunavir/r (adjusted HR 0.15; 95% CI 0.08–0.27).
In the largest prospective study (Italian multicentre cohort) in virologically suppressed adults with HIV-1, there was no significant difference in the estimated probability of remaining free from virological failure between patients who switched to dolutegravir plus lamivudine (n = 229) and those who switched to dolutegravir plus rilpivirine (n = 187) [37]. After a median of 22.3 months’ follow-up, the overall incidence of virological failure in the dolutegra- vir plus lamivudine group was 2.9 virological failures/100 person-years of follow-up (PYFU) and that in the dolute- gravir plus rilpivirine group was 1.3 virological failures/100 PYFU. There was no evidence of new mutations occurring in patients experiencing virological failure. There was also no between-group difference in the estimated probability of remaining on study treatment after adjustment for potential confounders [37].
⦁ Pharmacoeconomic Considerations

As is the case with all pharmacoeconomic studies, these data should be interpreted with caution given the inherent limi- tations of this type of study and the individual limitations/ assumptions of each study.
In a UK cost-minimization study conducted from a healthpayer perspective, dolutegravir plus lamivudine was estimated to be associated with a reduction in lifetime costs

Table 2 Efficacy of switching to single-tablet, once-daily dolutegravir/lamivudine in HIV-1 infected adults with current virological sup- pression on a ≥ 3-drug tenofovir alafenamide regimen in the phase 3 TANGO trial

Endpoint Regimen (no. of pts) 24 weeksa [30] (TDb; 95% CI) 48 weeksa [23]

HIV-1 RNA ≥ 50 copies/ mLc (% of pts)
HIV-1 RNA < 50 copies/ mLc,f (% of pts)
DTG/3TC (369) 0.3 (− 0.5; − 1.6 to 0.5) NI 0.3d (− 0.3e; − 1.2 to 0.7) NI

TAF-based regimen (372) 0.8 0.5d
DTG/3TC (369) 95 (− 1.4; − 4.4 to 1.6) NI 93.2 (0.2e; − 3.4 to 3.9) NI

TAF-based regimen (372) 96 93.0

An ongoing 200-week, randomized, open-label, multinational, noninferiority trial; analyses in the intent-to-treat-exposed population [23] 3TC lamivudine 300 mg, DTG dolutegravir 50 mg, NI noninferior, pts patients, TAF tenofovir alafenamide, TD adjusted treatment difference aAbstract [30] or oral [23] presentation
bCochran–Mantel–Haenszel-stratified analysis adjusted for baseline third agent class in TAF regimen [predefined noninferiority margin for viro- logical failure (HIV-1 RNA ≥ 50 copies/mL) of 4% and for virological success (HIV-1 RNA < 50 copies/mL) of − 8%] [23]
cUsing the FDA snapshot algorithm
dPrimary endpoint
eValue estimated from graph
fKey secondary endpoint

of £27,320 compared with dolutegravir plus tenofovir DF/ emtricitabine in ART-naïve HIV-infected patients (efficacy data from the GEMINI trials; Sect. 4.1.1) (abstract) [40]. This reduction in costs mostly reflects lower drug acquisition costs and to a lesser extent, reduced costs associated with adverse event and disease management [40].
In a Spanish cost-minimization study in virologically suppressed HIV-1-infected patients (DOLAMA study), switching to dolutegravir plus lamivudine was estimated to be associated with respective cost savings versus elvitegra- vir/cobicistat/emtricitabine/tenofovir AF or dolutegravir/ abacavir/lamivudine of €4164/year and €2741/year [39]. In a cost-effectiveness analysis, relative to these other ART regimens, switching to dolutegravir plus lamivudine was predicted be the more cost-effective approach [39].

⦁ Tolerability of Dolutegravir/Lamivudine
Treatment with dolutegravir plus lamivudine (adminis- tered separately or as an STR) for up 96 weeks was gen- erally well tolerated in ART-naïve or -experienced adults with HIV-1 infection participating in phase 3 trials [21–23] and real-world studies [37, 39] discussed in Sect. 4, with most adverse events (AEs) of mild to moderate severity, where specified [21–23, 39]. Based on clinical trials and postmarketing experience, very common (frequency ≥ 10%) adverse reactions (ARs) were headache, nausea and diar- rhoea, and common (≥ 1 to < 10%) ARs were psychiatric disorders (depression, anxiety, insomnia, abnormal dream), nervous system disorders (dizziness and somnolence, gas- trointestinal disorders (vomiting, flatulence, abdominal pain/ discomfort), skin and subcutaneous tissue disorders (rash, pruritus, alopecia), musculoskeletal and connective tissue disorders (arthralgia, muscle disorders), fatigue and liver enzyme abnormalities [10]
During the initial 48 weeks’ treatment in GEMINI trials (ART-naïve patients), a similar proportion of patients in the dolutegravir plus lamivudine and dolutegravir plus tenofovir DF/emtricitabine groups experienced ≥ 1 treatment-emer- gent AE (TEAE; 76 vs 81% of patients), most of which were not considered treatment-related (TR; any grade TRAEs 18 vs 24%; grade 2–5 TRAEs 6 vs 7%) and very few patients (1 vs 1%) discontinued therapy because of TRAEs [21]. In TANGO (ART-experienced patients), a similar proportion of patients in the dolutegravir plus lamivudine and tenofo- vir AF-based regimen group experienced ≥ 1 TEAE (80 vs 79%), most of which were not considered to be TRAEs (any grade TRAEs 5 vs 1%) and very few patients (2 vs < 1%) discontinued therapy because of TRAEs [23].
In GEMINI and TANGO trials, the most common TEAEs of any grade occurring in the dolutegravir plus lamivudine and the three-drug ART comparator arms were headache
(10 vs 10% in GEMINI [21]; 7 vs 5% in TANGO [23]),
diarrhoea (9 vs 11% [21]; 8 vs 7% [23]), nasopharyngitis
(8 vs 11% [21]; 12 vs 11% [23]), upper respiratory tract
infection (8 vs 6% [21]; 8 vs 9% [23]), syphilis (7 vs 4%
[23]), back pain (5 vs 4% [21]; 6 vs 8% [23]), fatigue (5 vs
1% [23]), pharyngitis (5 vs 4% [21]), nausea (4 vs 7% [21]),
insomnia (4 vs 6% [21]) and bronchitis (2 vs 5% [23]). No TRAE occurred with a frequency of more than 1% in either treatment group, with the most common grade 2–5 TRAEs being headache (1% in the dolutegravir + lamivudine group vs 1% in comparator group [21]; 1 vs 0% [23]), insomnia 1
vs 0% [23]), constipation (1 vs < 1% [23]) and flatulence (1
vs 0%) [23].
Serious TRAEs occurred in 1% of ART-naïve patients in each treatment group in the GEMINI trials [21], with no such events occurring in ART-experienced patients par- ticipating in TANGO [23]. No TR deaths occurred in the GEMINI and TANGO trials [21, 23].
Changes in renal biomarkers over 48 weeks generally favoured dolutegravir plus lamivudine treatment compared with a 3- or 4-drug ART regimen in phase 3 trials. In GEM- INI, adjusted mean changes at week 48 in the dolutegravir plus lamivudine group were significantly (all p ≤ 0.0004) better than in the dolutegravir plus tenofovir DF/emtricit- abine group, including adjusted mean changes from baseline in serum creatinine (10.4 vs 13.5 μmol/L), estimated glo- merular filtration (eGFR) calculated using cystatin C (6.3 vs 4.1 mL/min/1.73 m2) and eGFR calculated using creati- nine (− 12.1 vs − 15.5 mL/min/1.73 m2) [21]. This between- group difference may, at least in part, reflect the known adverse renal effects of tenofovir DF, a component of the three-drug regimen [21]. In TANGO, plasma/serum renal biomarkers also favoured (all p < 0.001) dolutegravir plus lamivudine over a tenofovir AF-based regimen (≥ 3 drugs), including adjusted mean changes from baseline in creatinine (+ 6.67 vs + 2.19 μmol/L), eGFR calculated using cystatin C (+ 0.1 vs − 1.6 mL/min/1.73 m2) and eGFR calculated using creatinine (− 7.8 vs − 3.0 mL/min/1.73 m2) [23]. Mean changes in serum creatinine over 48 weeks are associated with the inhibiting effect of dolutegravir on renal tubular transporters of creatinine; the changes are not considered clinically relevant and do not reflect a change in GFR [10]. Changes in bone turnover biomarkers at 48 weeks also generally favoured dolutegravir plus lamivudine over a 3- or 4-drug regimens [21, 23], with between-group differ- ences in GEMINI trials potentially reflecting the tenofovir DF (known association with impaired bone health) compo- nent of the three-drug regimen. For instance, in GEMINI, adjusted mean changes from baseline to week 48 were sig- nificantly lower (p < 0.0001) in the dolutegravir plus lamivu- dine group than in the three-drug regimen group for serum bone-specific alkaline phosphatase (1.22 vs 4.07 μg/L), serum osteocalcin (0.6 vs 6.17 μg/L), serum procollagen-1

N-terminal propeptide (0.40 vs 13.1 μg/L) and serum type 1 collagen C-telopeptide (0.14 vs 0.33 μg/L) [21].
Real-world studies in a limited number of ART-expe- rienced HIV-positive patients suggested that switching to dolutegravir plus lamivudine improved lumbar spine bone mineral density (BMD) [41, 42]. In a retrospective, obser- vational study in 20 ART-experienced patients with viro- logically suppressed HIV-1 who switched from two- or three-drug ART regimens to dolutegravir plus lamivudine, lumbar spine and femoral neck BMD and T-score increased after 12 months’ treatment [42]. These data are supported by evidence from the retrospective DOLUTILITY study (n = 1039) in HIV-1 patients switching to dolutegravir-based triple or dual regimens [41]. Based on data from 21 patients who had DEXA scans within 6 months prior to switching and 96 weeks after switching to dolutegravir plus lamivu- dine, this 2-drug regimen was associated with significant (p < 0.001) improvements in lumbar spine BMD, T-score and Z-score at 96 weeks, although changes at the hip were not significant [41].
In GEMINI trials, adjusted mean changes over 48 weeks in serum or plasma lipids from baseline were generally less favourable in the dolutegravir plus lamivudine than with dolutegravir plus tenofovir DF/emtricitabine group. Adjusted mean changes for assessed parameters were: serum or plasma total cholesterol (+ 0.32 vs − 0.15 mmol/L; p < 0.0001), high-density lipoprotein cholesterol (HDL-C;
+ 0.15 vs + 0.02 mmol/L; p < 0.0001), low-density lipo- protein cholesterol (+ 0.17 vs − 0.14 mmol/L; p < 0.0001), triglycerides (+ 0.03 vs − 0.08 mmol/L; p = 0.0457) and the ratio of total cholesterol to HDL-C (− 0.12 vs
− 0.24 mmol/L; p = 0.0182). These between-group differ- ences reflected the known impact of tenofovir DF (compo- nent of the three-drug regimen) on lipid parameters [21]. In TANGO, changes in serum lipids from baseline to week 48 week were similar with dolutegravir plus lamivudine to those with a 3- or 4-drug tenofovir AF-based regimen [23]. Dolutegravir lacks activity against HBV, whereas lami- vudine is active against HBV; however, lamivudine mono- therapy is not generally considered an adequate treatment for HBV. Hence, if dolutegravir/lamivudine is used in patients co-infected with HBV, an additional antiviral is generally needed. If dolutegravir/lamivudine is discontin- ued in patients co-infected with HBV, periodic monitoring of both liver function tests and markers of HBV replication is recommended, as withdrawal of lamivudine may result in acute exacerbation of hepatitis. Patients with pre-existing liver dysfunction, including chronic active hepatitis, have an increased frequency of liver function abnormalities during combination therapy, and should be monitored according to standard practice. If there is evidence of worsening liver disease in such patients, interruption or discontinuation of treatment must be considered. Liver chemistry elevations
consistent with immune reconstitution syndrome were observed in some HBV and/or HCV co-infected patients at the start of dolutegravir therapy. Monitoring of liver chem- istries is recommended in patients with HBV and/or HCV co-infection [10].

⦁ Dosage and Administration of Dolutegravir/Lamivudine
Oral dolutegravir/lamivudine is approved in several coun- tries, including those of the EU [10] and in the USA [11], for the treatment of HIV-1 infection; the specific indication may vary between individual countries. In the EU, it is recom- mended for the treatment of HIV-1 infection in adults and adolescents above 12 years of age weighing at least 40 kg, with no known or suspected resistance to the INSTI class or lamivudine [10]. The recommended dosage of dolutegravir/ lamivudine is 50/300 mg once daily. A separate prepara- tion of dolutegravir is available where a dose adjustment is indicated due to drug-drug interactions (Sect. 3.1); the physician should refer to the individual product information for dolutegravir for specific dosage adjustments [10].
Consult local prescribing information for detailed infor- mation regarding the use of dolutegravir/lamivudine, includ- ing specific indications, warnings, contraindications, drug interactions and use in special patient populations.

⦁ Place of Dolutegravir/Lamivudine
in the Management of HIV‑1 Infection
Several effective antiretroviral agents targeting different steps in the viral replication pathway are available for the treatment of HIV-1 infection, including STRs combining at least two drugs from at least two antiretroviral drug classes. Selection of an ART regimen requires an individualized approach based on regimen-specific factors such as com- plexity, costs, tolerability, drug interactions and genetic bar- riers to resistance, along with patient-specific factors such as baseline viral load, CD4+ count, HIV genotypic resist- ance, presence of comorbidities (including pregnancy, co- infections such as tuberculosis, HBV or HCV) and renal or hepatic dysfunction [4, 43–45]. Second generation INSTIs (dolutegravir, bictegravir) with their convenient once-daily regimens and noninferior efficacy, lower potential for drug interactions and high genetic barrier to resistance relative to PIs, provide options for a non-PI based ART regimen [46]. Current EACS [43], WHO [44], British HIVA [45] and US DHHS [4] treatment guidelines typically recommend INSTI-based regimens (preferred option) or ART regimens that include two NRTIs, plus a third drug from the NNRTI, boosted PI or INSTI class as an initial treatment option for

most individuals infected with HIV, including dolutegravir/ abacavir/lamivudine, bictegravir/emtricitabine/tenofovir AF, combination therapy with dolutegravir plus emtric- itabine plus tenofovir AF or tenofovir DF and raltegravir plus emtricitabine plus tenofovir DF or tenofovir AF. Of note, the updated November 2019 EACS guidelines are the first major HIV guidelines to include dual therapy with dolutegravir plus lamivudine as one of the preferred ini- tial treatment options [43]. WHO guidelines recommend a dolutegravir-based regimen as the preferred first-line ART for people living with HIV initiating ART and, in those with HIV infection for whom non-dolutegravir-based regimens are failing, dolutegravir in combination with an optimized NRTI backbone is recommended as a preferred second-line ART regimen [44]. STRs, including dual STRs such as dolutegravir/lamivudine, provide a complete regi- men with proven virological efficacy in ART-naïve and
-experienced patients with HIV-1 infection and offer the advantage of further simplification and de-intensification of ART regimens [3, 5, 8]. This also potentially spares the use of other ART drugs for subsequent treatment if viro- logical failure occurs.
In the pivotal GEMINI trials in ART-naïve patients with HIV-1 infection, treatment with dolutegravir plus lamivudine provided rapid and sustained virological sup- pression and was noninferior to the three-drug regimen of dolutegravir plus tenofovir DF/emtricitabine at 48 weeks, irrespective of baseline patient or disease characteristics (Sect. 4.1.1). Virological suppression was sustained at 96 weeks in these ongoing phase 3 trials (Sect. 4.1.1). Indi- rect evidence from a network meta-analysis supports the efficacy of dolutegravir plus lamivudine compared with traditional three-drug regimens such as dolutegravir plus tenofovir DF/emtricitabine (Sect. 4.1.1). In patients with HIV-1 with sustained virological suppression on their current tenofovir AF-based ART regimen (≥ 3 drugs), switching treatment to dolutegravir/lamivudine was noninferior to continuing on a tenofovir AF-based regi- men at 48 weeks in the ongoing phase 3 TANGO trial (Sect. 4.1.2). Results from retrospective or prospective real-world studies in patients with virological suppression switching from a ≥ 3-drug ART regimen to dolutegravir/ lamivudine STR were consistent with those observed in TANGO (Sect. 4.2). Importantly, no resistance mutations to either of the component drugs of dolutegravir/lamivu- dine were detected in patients with CVW in the GEMINI and TANGO trials, providing clinical support for the high genetic barrier to resistance observed with dolutegravir in vitro (Sect. 2.2).
Dolutegravir/lamivudine (administered as separate
agents in ART-naïve patients) was generally well toler- ated in RCTs and/or real-world studies in ART-naïve and -experienced patients, with most TEAEs of mild to
moderate severity, and very few patients discontinuing treatment because of these events (≤ 2% of patients) or experiencing serious TRAEs (≤ 1%) (Sect. 5). Changes in renal and bone biomarkers generally favoured dolutegravir plus lamivudine over three-drug ART regimens in RCTs; this may, at least in part, reflect the known adverse renal and bone effects of tenofovir DF (and to a lesser extent tenofovir AF), a component of the three-drug regimens (Sect. 5).
Given the lifetime commitment to ART in HIV-infected patients and the increasing number of patients living with HIV infection and taking ART, costs are an important consideration in ART selection [4, 44, 45]. Relative to no treatment, ART has been shown to be very cost effective in the EU and USA using both older and newer ART regi- mens, including in ART-experienced patients with drug- resistant HIV [4, 45]. Recent pharmacoeconomic analyses estimated that dolutegravir plus lamivudine reduced life- time costs compared with the three-drug dolutegravir plus tenofovir DF/emtricitabine regimen in ART-naive patients with HIV-1 (UK healthpayer perspective) (Sect. 4.3). In ART-experienced patients, switching to the two-drug dolutegravir plus lamivudine regimen was predicted to be cost-effective compared with a three- or four-drug ART regimen (Spanish DOLAMA study) (Sect. 4.3).
In conclusion, the STR dolutegravir/lamivudine offers the advantages of a complete two-drug regimen, with a convenient once-daily regimen and high genetic barrier to resistance. Current clinical data from ongoing RCTs and the real-world setting indicate that dolutegravir/lamivudine is an effective and generally well tolerated initial and sub- sequent treatment option for patients with HIV-1 infection with no known or suspected resistance to the INSTI class or lamivudine.

Data Selection Dolutegravir/Lamivudine: 275 records
Duplicates removed 31
Excluded during initial screening (e.g. press releases; news reports; not relevant drug/indication; preclinical study; reviews; case reports; not randomized trial) 153
Excluded during writing (e.g. reviews; duplicate data; small patient number; nonrandomized/phase I/II trials) 45
Cited efficacy/tolerability articles 21
Cited articles not efficacy/tolerability 25
Search Strategy: EMBASE, MEDLINE and PubMed from 1946 to present. Clinical trial registries/databases and websites were also searched for relevant data. Key words were HIV-1, dolute- gravir, lamivudine, DTG, 3TC. Records were limited to those in English language. Searches last updated 10 December 2019.

Acknowledgements During the peer review process, the manufacturer of dolutegravir/lamivudine was also offered an opportunity to review this article. Changes resulting from comments received were made on the basis of scientific and editorial merit.
Compliance with Ethical Standards
Funding The preparation of this review was not supported by any external funding.

Conflict of interest Lesley Scott is a salaried employee of Adis Inter- national Ltd/Springer Nature, is responsible for the article content and declares no relevant conflicts of interest.

References
⦁ Collins SE, Grant PM, Shafer RW. Modifying antiretroviral ther- apy in virologically suppressed HIV-1-infected patients. Drugs. 2016;76(1):75–98.
⦁ Sebaaly JC, Kelley D. HIV clinical updates: new single-tablet regimens. Ann Pharmacol. 2019;53(1):82–94.
⦁ Boswell R, Foisy MM, Hughes CA. Dolutegravir dual therapy as maintenance treatment in HIV-infected patients: a review. Ann Pharmacol. 2018;52(7):681–9.
⦁ US Department of Health and Human Services Panel on Antiret- roviral Guidelines for Adults and Adolescents. Guidelines for the use of antiretroviral agents in adults and adolescents with HIV; 2019. http://adisinfo.nih.gov/guidelines. Accessed 9 Sep 2019.
⦁ de Miguel Buckley R, Montejano R, Stella-Ascariz N, et al. New strategies of ARV: the road to simplification. Curr HIV/AIDS Rep. 2018;15:11–9.
⦁ Kangethe A, Polson M, Lord TC, et al. Real-world healthplan data analysis: key trends in medication adherence and overall costs in patients with HIV. J Manag Care Spec Pharm. 2019;25(1):88–93.
⦁ Altice F, Evuarherhe O, Shina S, et al. Adherence to HIV treat- ment regimens: systematic literature review and meta-analysis. Patient Prefer Adherence. 2019;13:475–90.
⦁ Cento V, Perno CF. Two-drug regimens with dolutegravir plus rilpivirine or lamivudine in HIV-1 treatment-naive, virologically suppressed patients: latest evidences from the literature on their efficacy and safety. J Glob Antimicrob Resist. 2019. ⦁ https://doi. ⦁ o⦁ rg/10.1016/j.jgar.2019.08.010.
⦁ Anstett K, Brenner B, Mesplede T, et al. HIV drug resistance against strand transfer integrase inhibitors. Retrovirology. 2017;14:36.
⦁ European Medicines Agency. Dovato (dolutegravir/lamivudine): summary of product characteristics; 2019. ⦁ http://www.ema.europ ⦁ a.eu/. Accessed 9 Sep 2019.
⦁ GlaxoSmithKline. DOVATO (dolutegravir and lamivudine tablets, for oral use): US prescribing information; 2019. ⦁ http://www.gskso ⦁ ur⦁ ce.com/. Accessed 9 Sep 2019.
⦁ McCormack PL. Dolutegravir: a review of its use in the man- agement of HIV-1 infection in adolescents and adults. Drugs. 2014;74(11):1241–52.
⦁ Hare S, Smith SJ, Métifiot M, et al. Structural and functional analyses of the second-generation integrase strand inhibitor dolutegravir (S/GSK1349572). Mol Pharmacol. 2011;80:565–72.
⦁ Pollicita M, Surdo M, Di Santo F, et al. Comparative replica- tion capacity of raltegravir-resistant strains and antiviral activity of the new generation integrase inhibitor dolutegravir in human primary macrophages and lymphocytes. J Antimicrob Chemother. 2014;69:2412–9.
⦁ Kobayashi M, Yoshinaga T, Seki T, et al. In vitro antiretroviral properties of S/GSK1349572, a next-generation HIV integrase inhibitor. Antimicrob Agents Chem. 2011;55(2):813–21.
⦁ Gillman J, Janulis P, Gulick R, et al. Comparable viral decay with initial dolutegravir plus lamivudine versus dolutegravir-based tri- ple therapy. J Antimicrob Chemother. 2019;74(8):2365–9.
⦁ Hightower KE, Wang R, DeAnda F, et al. Dolutegravir (S/ GSK1349572) exhibits significantly slower dissociation than raltegravir or elvitegravir from wild-type and integrase inhibitor- resistant HIV-1 integrase-DNA complexes. Antimicrob Agents Chem. 2011;35(10):4552–9.
⦁ DeAnda F, Hightower KE, Nolte RT, et al. Dolutegravir interactions with HIV-1 integrase-DNA: structural ration- ale for drug resistance and dissociation kinetics. PLoS One. 2013;8(10):e77448.
⦁ European Medicines Agency. Trivacy (dolutegravir): summary of product characteristics; 2019. http://www.ema.europa.eu/. Accessed 23 Sep 2019.
⦁ Rhees S-Y, Grant PM, Tzou PL, et al. A systematic review of the genetic mechanisms of dolutegravir resistance. J Antimicrob Chemother. 2019;74:3135–49.
⦁ Cahn P, Madero JS, Arribas JR, et al. Dolutegravir plus lamivu- dine versus dolutegravir plus tenofovir disoproxil fumarate and emtricitabine in antiretroviral-naive adults with HIV-1 infection (GEMINI-1 and GEMINI-2): week 48 results from two multi- centre, double-blind, randomised, non-inferiority, phase 3 trials. Lancet. 2019;393(10167):143–55.
⦁ Cahn P, Sierra Madero J, Arribas J, et al. Durable efficacy of dolutegravir (DTG) plus lamivudine (3TC) in antiretroviral treat- ment-naïve adults with HIV-1 infection—96-week results from the GEMINI studies [abstract no. WEAB0404LB]. J Int AIDS Soc. 2019;22(Suppl 5):103.
⦁ van Wyk J, Ajana F, Bisshop F, et al. Switching to DTG/3TC fixed dose combination (FDC) is non-inferior to continuing a TAF-based regimen in maintaining virologic suppression through 48 weeks (TANGO Study) [oral presentation]. In: 10th IAS con- ference on HIV science; 2019. ⦁ http://www.ias2019.org/Progr ⦁ amme. Accessed 11 Sep 2019.
⦁ Fourati S, Charpentier C, Amiel C, et al. Cross-resistance to elvitegravir and dolutegravir in 502 patients failing on raltegravir: a French national study of raltegravir-experienced HIV-1 infected patients. J Antimicrob Chemother. 2015;70:1507–12.
⦁ Cahn P, Rolon MJ, Figueroa MI, et al. Dolutegravir-lamivudine as initial therapy in HIV-1 infected, ARV-naive patients, 48-week results of the PADDLE (Pilot Antiretroviral Design with Dolute- gravir LamivudinE) study. J Int AIDS Soc. 2017;20(21678):1–7.
⦁ Taiwo BO, Zheng L, Stefanescu A, et al. ACTG A5353: a pilot study of dolutegravir plus lamivudine for initial treatment of human immunodeficiency virus-1 (HIV-1)-infected partici- pants with HIV-1 RNA <500000 copies/mL. Clin Infect Dis. 2018;66(11):1689–97.
⦁ Nyaku AN, Zheng L, Gulick RM, et al. Dolutegravir plus lami- vudine for initial treatment of HIV-1-infected participants with HIV-1 RNA <500 000 copies/mL: week 48 outcomes from ACTG 5353. J Antimicrob Chemother. 2019;74(5):1376–80.
⦁ Underwood M, Wang R, Horton J, et al. Dolutegravir (DTG) plus lamivudine (3TC) versus DTG plus tenofovir/emtricitabine (TDF/FTC) fixed-dose combination in the GEMINI studies: viral load rebound including‚ blips’ through 48 weeks [abstract no. MOPEB231]. In: 10th IAS conference on HIV science; 2019.
⦁ Radford M, Parks DC, Ferrante S, et al. Comparative efficacy and safety and dolutegravir and lamivudine in treatment naive HIV patients. AIDS. 2019;33(11):1739–49.
⦁ van Wyk J, Ajana F, Bisshop F, et al. Switching to DTG+3TC fixed dose combination (FDC) is non-inferior to continuing a

TAF-based regimen (TBR) in maintaining virologic suppression through 24 weeks (TANGO Study) [abstract no. WEAB0403LB]. J Int AIDS Soc. 2019;22(Suppl 5):102–3.
⦁ Blanco JL, Rojas J, Paredes R, et al. Dolutegravir-based main- tenance monotherapy versus dual therapy with lamivudine: a planned 24 week analysis of the DOLAM randomized clinical trial. J Antimicrob Chemother. 2018;73(7):1965–71.
⦁ Joly V, Burdet C, Landman R, et al. Dolutegravir and lamivudine maintenance therapy in HIV-1 virologically suppressed patients: results of the ANRS 167 trial (LAMIDOL). J Antimicrob Chem- other. 2019;74(3):739–45.
⦁ Taiwo BO, Marconi VC, Berzins B, et al. Dolutegravir plus lami- vudine maintains human immunodeficiency virus-1 suppression through week 48 in a pilot randomized trial. Clin Infect Dis. 2018;66(11):1794–7.
⦁ Baldin G, Ciccullo A, Borghetti A, et al. Virological efficacy of dual therapy with lamivudine and dolutegravir in HIV-1-infected virologically suppressed patients: long-term data from clinical practice. J Antimicrob Chemother. 2019;74(5):1461–3.
⦁ Diaco ND, Strickler C, Giezendanner S, et al. Systematic de-esca- lation of successful triple antiretroviral therapy to dual therapy with dolutegravir plus emtricitabine or lamivudine in Swiss HIV- positive persons. EClinicalMedicine. 2018;6:21–5.
⦁ Borghetti A, Lombardi F, Gagliardini R, et al. Efficacy and toler- ability of lamivudine plus dolutegravir compared with lamivudine plus boosted PIs in HIV-1 positive individuals with virologic sup- pression: a retrospective study from the clinical practice. BMC Infect Dis. 2019;19(59):1–9.
⦁ Ciccullo A, Baldin G, Capetti A, et al. A comparison between two dolutegravir-based two-drug regimens as switch strategies in a multicentre cohort of HIV-1-infected patients. Antivir Ther. 2019;24(1):63–7.
⦁ Maggiolo F, Gulminetti R, Pagnucco L, et al. Lamivudine/dolute- gravir dual therapy in HIV-infected, virologically suppressed patients. BMC Infect Dis. 2017;17(215):1–7.
⦁ Hidalgo-Tenorio C, Cortes LL, Gutierrez A, et al. DOLAMA study: effectiveness, safety and pharmacoeconomic analy- sis of dual therapy with dolutegravir and lamivudine in viro- logically suppressed HIV-1 patients. Medicine (Baltimore). 2019;98(32):e16813:1–7.
⦁ Ward T, Punekar YS, Darlington O, et al. Dolutegravir plus lami- vudine for the treatment of naive adults living with HIV-1: a UK cost-minimization analysis [abstract no. PIN64]. Value Health. 2018;21(Suppl 3):S231–2.
⦁ Piscaglia M, Gallazzi I, Restelli S, et al. Impact on bone min- eral density after 2 years of switching to four dolutegravir-based triple or dual regimens [abstract no. P162]. J Int AIDS Soc. 2018;21(Suppl 8):113–4.
⦁ Ciccullo A, D’Avino A, Lassandro AP, et al. Changes in bone mineral density in HIV-positive, virologically suppressed patients switching to lamivudine/dolutegravir dual therapy: preliminary results from clinical practice. Infez Med. 2018;26(4):336–40.
⦁ European AIDS Clinical Society. EACS guidelines version 10.0 November 2019; 2019. ⦁ http://eacsociety.org. Accessed 10 Dec 2019.
⦁ World Health Organization. Updated recommendations on first- line and second-line antiretroviral regimens and post-exposure prophylaxis and recommendations on early infant diagnosis of HIV; 2018. http://www.who.int/. Accessed 9 Sep 2019.
⦁ British HIV Association. BHIVA treatment guidelines: 2019 interim statement on two-drug regimens; 2019. ⦁ http://www.bhiva
.org/HIV-1-treatment-guidelines. Accessed 22 Nov 2019.
⦁ Boffito M, Waters L, Cahn P, et al. Perspectives on the barrier to resistance for dolutegravir + lamivudine, a 2-drug antiretroviral therapy for HIV-1 infection. AIDS Res Hum Retroviruses. 2019. https://doi.org/10.1089/AID.2019.0171.