C20H19F2N3O5
1051375-16-6
Human Immunodeficiency Virus Type 1 (HIV-1) Infection
Dolutegravir (DrugBank ID: DB08930, CAS Number: 1051375-16-6) is a second-generation integrase strand transfer inhibitor (INSTI) that has become a cornerstone in the contemporary management of Human Immunodeficiency Virus type 1 (HIV-1) infection. As a small molecule antiviral agent, dolutegravir is administered in combination with other antiretroviral drugs to suppress viral replication. Its primary mechanism of action involves blocking the strand transfer step of viral genome integration into the host cell's DNA [User Query]. This targeted viral process has no direct counterpart in human cellular mechanisms, a characteristic that contributes significantly to dolutegravir's generally favorable tolerability and minimal toxicity profile observed in clinical practice.[1] This specificity for a viral enzyme is a highly desirable attribute for antiretroviral agents intended for long-term administration, as is typical in HIV-1 management, potentially reducing the burden of off-target effects often associated with older drug classes.
Dolutegravir was developed by ViiV Healthcare.[3] It received its initial approval from the U.S. Food and Drug Administration (FDA) on August 12, 2013, under the brand name Tivicay [User Query]. This was followed by approvals from other regulatory agencies, including the European Medicines Agency (EMA). A significant milestone in its development was the approval of Juluca (dolutegravir/rilpivirine) on November 21, 2017, which marked the advent of the first complete two-drug regimen for the treatment of adults with HIV-1 [User Query]. This innovation signaled a shift towards simplifying treatment regimens. Further fixed-dose combinations (FDCs) incorporating dolutegravir have since become available, including Dovato (dolutegravir/lamivudine) and Triumeq (dolutegravir/abacavir/lamivudine), underscoring its versatility and central role in HIV therapy.[3] The strategic development from a single agent to its inclusion in multiple FDCs, particularly two-drug regimens, reflects a significant evolution in HIV treatment philosophy, aiming to reduce long-term drug exposure and pill burden while maintaining high efficacy.
Dolutegravir-based regimens are prominently featured in major international HIV treatment guidelines, such as those from the World Health Organization (WHO), often as preferred options for both first-line and second-line antiretroviral therapy (ART).[7] This strong recommendation is founded on a robust body of evidence demonstrating its superior or non-inferior virological efficacy compared to previous standards of care, a favorable safety and tolerability profile, a high genetic barrier to the development of drug resistance, and the convenience of once-daily dosing for many patient populations.[8] The introduction of dolutegravir represented a considerable advancement over earlier-generation INSTIs and other antiretroviral classes, offering improved outcomes and simplified treatment approaches for individuals living with HIV-1.
Dolutegravir exerts its antiviral effect by specifically inhibiting the HIV-1 integrase enzyme. This enzyme is crucial for the viral replication cycle, as it catalyzes the insertion of the viral DNA (reverse transcribed from viral RNA) into the host cell's chromosomal DNA. Dolutegravir binds to the active site of the integrase enzyme, effectively blocking the strand transfer step of this integration process.[1] The mechanism involves the chelation of divalent metal ions, typically magnesium (Mg2+), which are essential cofactors located within the catalytic core of the integrase enzyme. By sequestering these metal ions, dolutegravir prevents the enzyme from properly processing the viral DNA ends and covalently joining them to the host DNA.[2]
A distinguishing feature of dolutegravir, when compared to first-generation INSTIs such as raltegravir and elvitegravir, is its molecular interaction with the target enzyme. Dolutegravir has been shown to have a slower rate of dissociation from the integrase-DNA complex. Furthermore, it appears capable of occupying more space within the catalytic site, particularly the region vacated by the displaced viral DNA end.[2] These distinct binding kinetics and deeper penetration into the active site are thought to be key contributors to dolutegravir's higher genetic barrier to resistance. A prolonged interaction with its target means that the virus would likely need to accumulate multiple or more significant mutations to effectively evade the drug's inhibitory action, compared to drugs with faster off-rates. This sustained enzyme inhibition is a critical factor in its robust antiviral activity, even against some viral strains that have developed resistance to earlier INSTIs. The chemical property of dolutegravir enabling chelation of divalent cations is not only fundamental to its enzymatic inhibition but also underlies its pharmacokinetic interaction with polyvalent cation-containing medications in the gastrointestinal tract, a distinct phenomenon discussed later.
Dolutegravir demonstrates potent in vitro antiviral activity against a range of laboratory strains and clinical isolates of wild-type HIV-1. The mean half-maximal effective concentration (EC50) values are typically observed in the low nanomolar range, for instance, between 0.5 nM and 2.1 nM in peripheral blood mononuclear cells (PBMCs) and MT-4 T-cell lines.[1] The pharmacodynamic profile of dolutegravir is characterized by a clear relationship between drug exposure, particularly trough plasma concentrations (Cmin or Cτ), and virologic response. Clinical studies have established that the achieved trough concentrations with standard dosing regimens are substantially higher than the protein-adjusted EC90 (the concentration required to inhibit 90% of viral replication in vitro, adjusted for plasma protein binding) for wild-type HIV-1.[11] This ensures sustained virologic suppression and contributes to its high efficacy and the difficulty with which resistance emerges.
The pharmacokinetic (PK) properties of dolutegravir have been extensively studied in healthy volunteers and HIV-1-infected individuals, revealing a profile conducive to effective antiretroviral therapy.[13]
Following oral administration, dolutegravir is absorbed with peak plasma concentrations (Tmax) typically reached 2 to 3 hours post-dose.[13] With once-daily dosing, pharmacokinetic steady-state is generally achieved within approximately 5 days, showing an average accumulation ratio for area under the concentration-time curve (AUC), maximum concentration (Cmax), and concentration at 24 hours (C24h) ranging from 1.2 to 1.5.[13] Dolutegravir exhibits less than dose-proportional increases in plasma concentrations at doses above 50 mg.[13] The absolute bioavailability of dolutegravir has not been definitively established.[13] In vitro studies have identified dolutegravir as a substrate for P-glycoprotein (P-gp), a drug efflux transporter.[13]
The presence of food influences dolutegravir absorption. Food intake generally increases the extent of absorption and slows the rate at which it is absorbed. Specifically, low-, moderate-, and high-fat meals have been shown to increase dolutegravir AUC(0−∞) by 33%, 41%, and 66%, respectively. Concurrently, Cmax increases by 46%, 52%, and 67%, respectively, and Tmax is prolonged from approximately 2 hours under fasted conditions to 3, 4, or 5 hours with low-, moderate-, or high-fat meals, respectively.[11] Despite these observed increases in exposure, dolutegravir can generally be administered with or without food, as these changes are not anticipated to adversely impact clinical safety or efficacy in most patients.[11] However, this pharmacokinetic food effect can be strategically utilized; in the presence of certain integrase class resistance mutations, particularly those involving the Q148 pathway, administering dolutegravir with food is preferred to enhance systemic exposure and potentially overcome reduced viral susceptibility.[15] This nuanced recommendation highlights how a generally non-critical PK interaction can become a therapeutic consideration in specific clinical scenarios, aiming to maximize drug concentrations against less sensitive viral strains.
Dolutegravir is extensively bound to human plasma proteins, with a binding percentage of $\geq$98.9% based on in vivo data. This binding is reported to be independent of the plasma concentration of dolutegravir.[13] The apparent volume of distribution (Vd/F) following a 50 mg once-daily administration is estimated to be 17.4 L, based on population pharmacokinetic analyses.[13]
Dolutegravir demonstrates penetration into the cerebrospinal fluid (CSF). In a study involving 11 treatment-naive subjects receiving dolutegravir 50 mg daily in combination with abacavir/lamivudine, the median dolutegravir concentration in CSF was 18 ng/mL (range: 4 ng/mL to 232 ng/mL) when measured 2 to 6 hours post-dose after 2 weeks of treatment.[13] While CSF penetration is confirmed, the direct clinical relevance of these concentrations for controlling viral reservoirs in the central nervous system is not fully established.
The primary metabolic pathway for dolutegravir is glucuronidation, mediated predominantly by the enzyme uridine diphosphate glucuronosyltransferase 1A1 (UGT1A1).[1] The cytochrome P450 (CYP) enzyme system, specifically CYP3A, plays a minor role in its metabolism.[1] In vitro studies also indicate that dolutegravir can be a substrate for UGT1A3 and UGT1A9.[1] This reliance on UGT1A1 rather than extensive CYP450 metabolism contributes to a relatively lower potential for many common drug-drug interactions compared to antiretrovirals that are heavily metabolized by CYP enzymes. Nevertheless, potent inducers or inhibitors of UGT1A1 can still significantly affect dolutegravir concentrations, necessitating dose adjustments in certain situations.[13]
Following a single oral dose of radiolabeled dolutegravir, approximately 53% of the total administered dose is excreted unchanged in the feces.[1] Urinary excretion accounts for about 31% of the total oral dose. This fraction comprises an ether glucuronide of dolutegravir (representing 18.9% of the total dose), a metabolite formed by oxidation at the benzylic carbon (3.0% of the total dose), and its hydrolytic N-dealkylation product (3.6% of the total dose).[1] The renal elimination of unchanged dolutegravir is minimal, accounting for less than 1% of the administered dose.[1] Dolutegravir has a terminal half-life of approximately 14 hours, and its apparent clearance (CL/F) is estimated to be around 1.0 L/h, based on population pharmacokinetic analyses.[13]
Dolutegravir is available in different oral formulations, including film-coated tablets and dispersible tablets for oral suspension. It is critical to note that these formulations are not bioequivalent on a milligram-per-milligram basis due to differences in bioavailability. For instance, for adults, a 50 mg film-coated tablet provides exposure comparable to 30 mg of the dispersible tablet formulation.[15] Therefore, patients switching between these formulations must adhere to the specific dosing recommendations for the particular formulation being used to ensure appropriate drug exposure and avoid under- or over-dosing. This distinction is particularly crucial for pediatric patients or individuals with swallowing difficulties who may rely on the dispersible formulation.
The following table summarizes key pharmacokinetic parameters for dolutegravir:
Table 1: Summary of Dolutegravir Pharmacokinetic Parameters
Parameter | Value | Reference(s) |
---|---|---|
Tmax (fasted) | 2-3 hours | 13 |
Food Effect on AUC(0−∞) (High-Fat Meal) | Increased by 66% | 11 |
Food Effect on Cmax (High-Fat Meal) | Increased by 67% | 11 |
Protein Binding | $\geq$98.9% | 13 |
Apparent Volume of Distribution (Vd/F) | 17.4 L | 13 |
CSF Penetration (Median Conc.) | 18 ng/mL (range 4-232 ng/mL) | 13 |
Primary Metabolic Pathway | Glucuronidation via UGT1A1 | 1 |
Minor Metabolic Pathway | Oxidation via CYP3A | 1 |
Primary Excretion Route (Unchanged Drug) | Feces (~53%) | 1 |
Secondary Excretion Route (Metabolites) | Urine (~31% as metabolites) | 1 |
Terminal Half-life (t1/2) | ~14 hours | 13 |
Apparent Clearance (CL/F) | ~1.0 L/h | 13 |
Dolutegravir has demonstrated robust clinical efficacy across a spectrum of HIV-1 infected patient populations, including treatment-naive individuals, treatment-experienced but INSTI-naive patients, and as a component of maintenance therapy.
Several pivotal Phase 3 trials have established the efficacy of dolutegravir in antiretroviral-naive adults:
The consistent theme of better tolerability leading to fewer discontinuations, as seen in the SINGLE and FLAMINGO trials, is a significant factor in dolutegravir's comparative trial successes. While virologic potency is essential, a drug's ability to be well-tolerated allows for better adherence and persistence on therapy, which translates to superior long-term outcomes in head-to-head comparisons. Furthermore, the robust efficacy observed in patients with high baseline viral loads in trials like FLAMINGO and SAILING suggests an intrinsic potency or a more forgiving profile when the initial virologic challenge is substantial.
The emergence and validation of 2DRs centered around dolutegravir, such as DTG+3TC in the D2ARLING and LAMIDOL trials, represent a significant evolution in HIV treatment paradigms. This shift away from the traditional three-drug backbone is largely enabled by dolutegravir's high potency and robust resistance barrier, which allow for regimen simplification without compromising virologic control in suitable patients, aiming to reduce long-term drug exposure and potential toxicities.
The following table summarizes the key efficacy outcomes from these pivotal clinical trials:
Table 2: Summary of Key Phase 3/4 Clinical Trial Efficacy Outcomes for Dolutegravir
Trial Name | Patient Population | Comparator(s) | Primary Endpoint (% HIV RNA <50 copies/mL at Week X) | Dolutegravir Arm Result | Comparator Arm Result | Key Conclusion (Non-inferiority/Superiority) | Emergent INSTI Resistance (DTG vs. Comparator) | Reference(s) |
---|---|---|---|---|---|---|---|---|
SINGLE | Naive | EFV/TDF/FTC | Week 48 | 88% | 81% | Superiority | None vs. N/A (EFV is NNRTI) | 10 |
SPRING-2 | Naive | Raltegravir (+ dual NRTI) | Week 48 | 88% | 85% | Non-inferiority | None vs. 1 patient (6% of failures) | 8 |
FLAMINGO | Naive | Darunavir/ritonavir (+ dual NRTI) | Week 48 | 90% | 83% | Superiority | None vs. None | 10 |
D2ARLING | Naive (no baseline resistance testing) | DTG + TDF/FTC or TDF/3TC | Week 48 | 92% | 89% | Non-inferiority | None vs. None | 22 |
SAILING | Experienced, INSTI-Naive | Raltegravir (+ OBR) | Week 48 | 71% | 64% | Superiority | 1% (4 patients) vs. 5% (17 patients) | 9 |
LAMIDOL | Suppressed (switch to DTG+3TC) | N/A (single arm, switch from triple therapy) | Week 48 (success rate) | 97% | N/A | Promising maintenance strategy | None detected | 21 |
OBR: Optimized Background Regimen
Dolutegravir is generally recognized for its favorable safety and tolerability profile, a factor that significantly contributes to its widespread use and patient adherence.
In clinical trials involving adult populations, dolutegravir has been shown to be well-tolerated.[1] The most frequently reported adverse events (typically defined as occurring in $\geq$2% of participants in any one adult trial) are generally mild to moderate in intensity. These commonly include insomnia, fatigue, and headache.[1] Gastrointestinal disturbances such as nausea and diarrhea are also among the more common side effects.[1] Other reported events include nasopharyngitis, dizziness, rash, and abnormal dreams.[1] Laboratory abnormalities that have been noted include elevations in cholesterol, triglycerides, lipase, glucose (hyperglycemia), creatine kinase, aspartate aminotransferase (AST), alanine aminotransferase (ALT), and bilirubin.[25] The overall low rates of discontinuation due to adverse events in pivotal clinical trials, often lower than comparator arms, underscore its good tolerability and contribute to its favorable standing in comparative efficacy assessments.[17]
Despite its generally good safety profile, certain serious adverse events and warnings are associated with dolutegravir use, as outlined in regulatory agency labeling and observed in clinical studies:
Dolutegravir is subject to several clinically significant drug-drug interactions, primarily involving polyvalent cations, certain UGT1A1/CYP3A enzyme inducers, and drugs that are substrates of the renal transporter OCT2.
Coadministration of dolutegravir with products containing polyvalent cations, such as antacids (containing magnesium or aluminum), sucralfate, or mineral supplements (containing iron or calcium), can significantly decrease the absorption of dolutegravir. This interaction occurs due to the formation of poorly soluble chelation complexes between dolutegravir and the polyvalent cations in the gastrointestinal tract, leading to reduced dolutegravir plasma concentrations and potentially compromising its antiviral efficacy.[2]
To manage this interaction, specific administration timing is recommended: dolutegravir should be administered at least 2 hours before or 6 hours after taking medications containing these polyvalent cations.[2] An important exception exists for calcium and iron supplements: if dolutegravir (as Tivicay or in the FDC Dovato) is taken with food, these supplements can be co-administered at the same time.[25] This flexibility when taken with food may be due to food mitigating the chelation effect, possibly by altering gastric conditions or by the cations binding to food components. However, this adds a layer of complexity to patient counseling, requiring clear instructions to ensure adherence and maintain therapeutic dolutegravir levels.
The coadministration of dolutegravir with dofetilide, an antiarrhythmic agent, is contraindicated.[4] Dolutegravir is an inhibitor of the renal organic cation transporter 2 (OCT2), which plays a crucial role in the renal clearance of dofetilide. By inhibiting OCT2, dolutegravir can significantly increase dofetilide plasma concentrations.[13] Elevated dofetilide levels pose a substantial risk of serious and potentially life-threatening cardiac arrhythmias, including Torsades de Pointes.[25] This contraindication underscores the critical importance of recognizing transporter-mediated drug interactions, as a pharmacokinetic effect (OCT2 inhibition) can lead to severe pharmacodynamic consequences, especially with drugs like dofetilide that have a narrow therapeutic index.
Dolutegravir has been shown to significantly increase plasma concentrations (both AUC and Cmax) of metformin, an oral antihyperglycemic agent, in a dose-dependent manner.[27] When dolutegravir 50 mg is administered once daily, metformin AUC increases by approximately 79% and Cmax by 66%. With dolutegravir 50 mg twice daily, metformin AUC increases by about 145% and Cmax by 111%.[27]
The primary mechanism for this interaction is the inhibition of OCT2 by dolutegravir, which reduces the renal tubular secretion of metformin.[27] Dolutegravir is a more potent inhibitor of OCT2 (IC$_{50}$ = 1.9 µM) compared to its inhibition of MATE1/2-K (IC$_{50}$ = 6.3-25 µM).[28] While OCT2 inhibition is the main driver, the observed magnitude of the increase in metformin exposure was somewhat higher than might be anticipated based solely on OCT2 inhibition and the known fraction of metformin clearance mediated by active tubular secretion, suggesting that other unidentified processes might also contribute to this interaction.[27]
Given the significant increase in metformin exposure, clinical recommendations include considering dose adjustments of metformin to maintain optimal glycemic control when dolutegravir is initiated or discontinued in a patient taking metformin. Patients should be monitored for metformin-related adverse effects, although the risk of hypoglycemia is not typically increased with metformin monotherapy, and the plasma concentrations observed in interaction studies were generally below those associated with lactic acidosis.[26]
As dolutegravir is primarily metabolized by UGT1A1 with a minor contribution from CYP3A, drugs that induce these enzymes can decrease dolutegravir plasma concentrations, potentially reducing its efficacy. Potent inducers include efavirenz, fosamprenavir/ritonavir, tipranavir/ritonavir, rifampin, carbamazepine, oxcarbazepine, phenytoin, phenobarbital, and the herbal product St. John's wort (Hypericum perforatum).[13]
For treatment-naive or treatment-experienced INSTI-naive patients, when dolutegravir (Tivicay) is coadministered with potent UGT1A/CYP3A inducers such as efavirenz, fosamprenavir/ritonavir, tipranavir/ritonavir, or rifampin, the dolutegravir dose should be increased to 50 mg twice daily.[13] If Dovato (DTG/3TC) is coadministered with carbamazepine or rifampin, an additional 50 mg dose of dolutegravir (as Tivicay) should be taken, separated by 12 hours from the Dovato dose.[26] Coadministration of Dovato with oxcarbazepine, phenobarbital, phenytoin, or St. John's wort should be avoided due to insufficient data to make dosing recommendations.[26] Etravirine, when administered without boosted protease inhibitors, also decreases dolutegravir concentrations, and the recommended adult dose of dolutegravir in this scenario is 50 mg twice daily.[15] Conversely, potent inhibitors of UGT1A1 or CYP3A could theoretically increase dolutegravir concentrations, but this is generally less of a clinical concern requiring dose adjustment for dolutegravir itself, though interactions with other co-administered drugs may still occur.
Several other drug interactions with dolutegravir warrant attention:
The following table summarizes key drug interactions with dolutegravir and provides management recommendations:
Table 3: Clinically Significant Drug Interactions with Dolutegravir and Management Recommendations
Interacting Agent/Class | Mechanism of Interaction | Effect on Dolutegravir or Co-administered Drug | Clinical Recommendation | Reference(s) |
---|---|---|---|---|
Dofetilide | OCT2 inhibition by dolutegravir | Increased dofetilide levels | Contraindicated due to risk of serious/life-threatening arrhythmias | 4 |
Polyvalent Cations (Mg, Al, Ca, Fe antacids/supplements) | Chelation in GI tract | Decreased dolutegravir absorption and plasma concentrations | Administer dolutegravir 2 hours before or 6 hours after polyvalent cations. If taken with food, dolutegravir and Ca/Fe supplements can be taken together (Tivicay, Dovato). | 2 |
Metformin | OCT2 inhibition by dolutegravir | Increased metformin plasma exposure (AUC and Cmax) | Consider metformin dose adjustment to maintain glycemic control when starting/stopping dolutegravir. Refer to metformin PI. | 26 |
Rifampin | Potent UGT1A1/CYP3A induction | Decreased dolutegravir plasma concentrations | Increase dolutegravir (Tivicay) dose to 50 mg BID. For Dovato, add DTG 50 mg 12h apart from Dovato. | 13 |
Efavirenz | Potent UGT1A1/CYP3A induction | Decreased dolutegravir plasma concentrations | Increase dolutegravir (Tivicay) dose to 50 mg BID. | 13 |
Carbamazepine | Potent UGT1A1/CYP3A induction | Decreased dolutegravir plasma concentrations | For Dovato, add DTG 50 mg 12h apart from Dovato. For Tivicay, consider 50 mg BID if INSTI-naive. | 15 |
St. John's wort (Hypericum perforatum) | Potent UGT1A1/CYP3A induction | Decreased dolutegravir plasma concentrations | Avoid coadministration with Dovato. For Tivicay, if INSTI-naive, increase dose to 50 mg BID. | 15 |
Oxcarbazepine, Phenobarbital, Phenytoin | Potent UGT1A1/CYP3A induction | Decreased dolutegravir plasma concentrations | Avoid coadministration with Dovato. For Tivicay, if INSTI-naive, increase dose to 50 mg BID. | 15 |
Dalfampridine | OCT2 and MATE1 inhibition by dolutegravir | Increased dalfampridine levels | Potential for increased risk of seizures. Weigh benefits vs. risks. | 25 |
Sorbitol (repeated use) | Potential to decrease lamivudine concentrations (in Dovato) | Decreased lamivudine exposure | When possible, avoid concomitant use with Dovato. | 26 |
Dolutegravir is characterized by a relatively high genetic barrier to resistance, especially when compared to first-generation INSTIs. This property is a significant clinical advantage, contributing to its durable efficacy.
The term "high genetic barrier" implies that multiple viral mutations, or specific combinations thereof, are typically necessary to confer clinically significant resistance to dolutegravir.[2] This contrasts with some antiretroviral agents where a single mutation can lead to high-level resistance. However, this barrier is not absolute. Resistance to dolutegravir can and does emerge, particularly under conditions that favor viral replication in the presence of suboptimal drug concentrations, such as poor adherence, pre-existing low-level resistance that might not be detected by standard assays, or in situations of functional monotherapy (where other drugs in the regimen are inactive).[7] The observation of resistance emerging even in INSTI-naive individuals, though at low rates in controlled clinical trials, underscores the importance of ongoing surveillance and adherence support in broader clinical application.
Several distinct mutational pathways have been identified that can reduce susceptibility to dolutegravir. The patterns of emergent mutations can differ based on the patient's prior INSTI exposure and the specific clinical context:
Specific mutational pathways have distinct characteristics:
* The R263K mutation often appears as a single primary mutation or in combination with minor accessory mutations. It typically confers a relatively low-level reduction in dolutegravir susceptibility, often around 2-fold.7
* The G118R mutation can also emerge and may be associated with a more significant reduction in dolutegravir susceptibility, generally >5-fold. When G118R is combined with other mutations, such as T66I and E138K, it can lead to high-level resistance and potentially pan-INSTI resistance, meaning resistance to all drugs in the class.7
* The Q148H/R/K mutations are particularly significant. While a Q148 mutation alone may not drastically reduce dolutegravir susceptibility, its combination with one or more secondary mutations (e.g., G140S/A, E138K, L74I) can lead to substantial and high-level resistance to dolutegravir and other INSTIs.29
* The N155H mutation, when present alone or with only one additional INSTI resistance mutation, usually results in less than a 2-fold reduction in dolutegravir susceptibility. However, its impact can be greater when it co-occurs with Q148 pathway mutations or multiple other resistance mutations.29
Understanding these distinct mutational pathways is critical for interpreting genotypic resistance tests. The implications of, for example, an R263K mutation are different from a G118R or a complex Q148 pathway mutation in terms of residual drug activity and options for future therapy.
The degree to which these mutations affect dolutegravir's antiviral activity is quantified by in vitro susceptibility testing, often reported as a "fold-change" (FC) in the EC50 value compared to wild-type virus. For mutations in the Q148 pathway, a 3- to 4-fold reduction in dolutegravir susceptibility has been associated with a measurably reduced virologic response to therapy, while a 10-fold reduction is linked to a markedly diminished response.[29] The precise clinical significance thresholds for fold-changes associated with the R263K or G118R pathways are less clearly defined.[29] Furthermore, some INSTI resistance mutations, notably G118R and R263K, have been shown to reduce the replication capacity of HIV-1 in vitro. This impaired viral fitness might influence their clinical impact and the rate at which they emerge or persist.[29]
Dolutegravir generally retains antiviral activity against many HIV-1 isolates that are resistant to first-generation INSTIs like raltegravir and elvitegravir, especially if the resistance is due to single mutations such as Y143R, T66I, or E92Q.[2] However, high-level cross-resistance can occur. This is particularly true for viruses harboring Q148 pathway mutations when combined with other secondary INSTI resistance mutations.[15] As mentioned, the G118R pathway, if it evolves to include additional mutations like T66I and E138K, can confer broad or pan-INSTI resistance, limiting future treatment options within this class.[7]
While clinical trials provide controlled data, real-world observations are crucial. In clinical trials, the emergence of dolutegravir-selected DRMs in INSTI-naive patients failing dual NRTI + DTG-based ART has been reported in up to 3.8% of ART-experienced individuals and around 0.7% in ART-naive individuals.[7] There are concerns that the rates of resistance emergence might be higher in real-world resource-limited settings. Factors contributing to this could include highly standardized regimens with limited flexibility, challenges in maintaining optimal adherence, interruptions in drug supply, and restricted access to routine viral load monitoring and genotypic resistance testing.[7]
Furthermore, emerging evidence suggests that resistance patterns and their frequencies might vary depending on the HIV-1 subtype. For instance, some data indicate that non-B subtypes might exhibit higher frequencies of G118R and Q148 pathway mutations, whereas R263K might be more predominant in subtype B infections.[7] Given that non-B subtypes are prevalent in many regions where dolutegravir is being extensively deployed, these potential subtype-specific differences in resistance development could have significant global health implications and warrant dedicated surveillance.
The following table summarizes key dolutegravir resistance mutations and their characteristics:
Table 4: Key Dolutegravir Resistance Mutations and Their Impact
Mutation(s) | Typical Clinical Setting of Emergence | Fold-Change in Dolutegravir Susceptibility (Approximate Range) | Impact on Replication Capacity (if known) | Cross-Resistance to other INSTIs | Reference(s) |
---|---|---|---|---|---|
R263K (often alone or +M50I/H51Y/E138K) | INSTI-Naive (primary or monotherapy) | ~2-fold (alone); up to 10-15 fold with G118R/N155H/Q148R | Reduced | Minimal alone; increased with additional mutations | 7 |
G118R (alone or +T66A/L74I/T97A/E138K) | INSTI-Naive (primary or monotherapy) | 5-15 fold (alone/some combos); >15-fold (e.g., +T66I+E138K) | Reduced | Partial to high; G118R+T66I+E138K can lead to pan-INSTI resistance | 7 |
N155H (alone or + other mutations) | INSTI-Naive (esp. monotherapy), INSTI-Experienced | <2-fold (alone); 2 to >15-fold with Q148H/R or multiple others | Variable | Variable; contributes to cross-resistance with other mutations | 29 |
Q148H/R/K + G140S/A and/or E138K/A/T (± L74I) | INSTI-Experienced (common); INSTI-Naive (less common, esp. monotherapy) | 3-fold (Q148H+G140S) to >20-fold (complex patterns) | Variable | High-level cross-resistance to raltegravir, elvitegravir; significant impact on dolutegravir | 15 |
T97A | INSTI-Experienced (often with other mutations) | Minor alone; contributes with other mutations | Not well defined | Accessory mutation, contributes to resistance to other INSTIs | 29 |
The use of dolutegravir in specific populations requires careful consideration of unique physiological states, potential risks, and available data.
The recommendations for dolutegravir use during pregnancy and around the time of conception have evolved significantly based on accumulating pharmacovigilance data.
The following table provides a summary of EMA-recommended dosing for dolutegravir dispersible tablets in pediatric populations without INSTI resistance:
Table 5: Dolutegravir Dosing in Pediatric Populations (Tivicay Dispersible Tablets - EMA Recommendations)
Body Weight (kg) | Age | Once-Daily Dose (mg) | Twice-Daily Dose (mg) | Reference(s) |
---|---|---|---|---|
3 to < 6 kg | ≥ 4 weeks | 5 | Not Applicable | 15 |
6 to < 10 kg | < 6 months | 10 | 5 | 15 |
≥ 6 months | 15 | 10 | 15 | |
10 to < 14 kg | ≥ 6 months | 20 | 10 | 15 |
14 to < 20 kg | ≥ 6 months | 25 | 15 | 15 |
≥ 20 kg | ≥ 6 months | 30 | 15 | 15 |
Note: If certain UGT1A or CYP3A inducers are coadministered, the weight-based once-daily dose should be administered twice daily.[15] These are EMA recommendations; FDA dosing for 10 mg dispersible tablets may vary slightly.
Patients with HIV-1 and co-infection with hepatitis B virus (HBV) or hepatitis C virus (HCV) may be at an increased risk for the development or worsening of transaminase elevations when treated with dolutegravir-containing regimens. Close monitoring of liver function is recommended in these patients.[13]
A critical consideration for HIV/HBV co-infected patients is the HBV activity of NRTIs included in the regimen. Lamivudine (a component of Dovato and Triumeq) and tenofovir (often used as part of a dolutegravir-based regimen) possess activity against HBV. Discontinuation of these HBV-active agents in co-infected patients can lead to HBV reactivation or flares, characterized by sudden increases in HBV DNA and ALT levels.[4] Therefore, fixed-dose combinations like Dovato and Triumeq carry boxed warnings regarding the risk of lamivudine-resistant HBV emergence and exacerbations of HBV upon discontinuation in co-infected individuals. Careful management, including consideration of ongoing anti-HBV therapy, is essential if a dolutegravir-containing FDC with HBV activity is changed or stopped in these patients.
Dolutegravir has received widespread regulatory approval and is available in various formulations to suit different patient needs.
The approval of multiple FDCs containing dolutegravir highlights its central role as a cornerstone agent in modern HIV therapy. This strategy facilitates regimen simplification, which can lead to improved adherence and quality of life for patients. The evolution from a single agent to a 3-drug FDC (Triumeq) and subsequently to 2-drug FDCs (Juluca, Dovato) reflects a significant trend in HIV treatment towards minimizing drug burden while maintaining high efficacy.
Dolutegravir is available in the following formulations:
Dolutegravir has fundamentally transformed the landscape of HIV-1 treatment since its introduction. Its combination of high virological efficacy, a generally favorable tolerability profile, a high genetic barrier to resistance, and the convenience of once-daily dosing for most patients has established it as a preferred agent in global HIV treatment guidelines. Its impact is evident in its widespread use in first-line therapy for treatment-naive individuals, its role in simplifying treatment through effective two-drug regimens, and its utility in managing certain treatment-experienced populations, including those with some prior INSTI exposure, albeit with careful consideration of resistance patterns. The success of dolutegravir has been a key driver in the broader acceptance and investigation of 2DRs as both initial and maintenance therapy, challenging the long-standing paradigm of triple-drug regimens.
Despite the significant advancements offered by dolutegravir, ongoing research and unmet needs persist in HIV-1 management. Continued long-term surveillance of dolutegravir-based regimens, especially 2DRs, is essential to monitor for any unforeseen late-emerging toxicities and to confirm sustained efficacy over decades of use.
A critical area of focus is the continued understanding and management of dolutegravir resistance. While the genetic barrier is high, resistance does emerge. Monitoring these resistance patterns in diverse real-world settings, particularly in regions with high HIV prevalence, varied viral subtypes, and potential challenges in adherence and healthcare access, is crucial.[7] The potential for different resistance pathways to emerge in non-B HIV subtypes, as suggested by some research, requires further investigation to ensure optimal treatment strategies globally.[7] This vigilance is paramount to preserving dolutegravir's long-term utility, especially in resource-limited settings where it forms the backbone of public health ART programs.
Further optimization of dolutegravir use in specific populations, such as the very young, the elderly with increasing comorbidities, and pregnant individuals (despite current favorable data), will continue to be an area of research. While not specifically covered in the provided information, the broader field of HIV research is also exploring the role of potent agents like dolutegravir in long-acting injectable formulations, HIV prevention strategies (Pre-Exposure Prophylaxis, PrEP), and as components of potential HIV cure strategies, areas where its favorable characteristics could be highly advantageous. The journey of dolutegravir from its development to its current prominent role exemplifies the remarkable progress in antiretroviral therapy, yet the ongoing pursuit of even better, safer, and more convenient treatments for all individuals living with HIV remains a global health priority.
Published at: June 9, 2025
This report is continuously updated as new research emerges.
Empowering clinical research with data-driven insights and AI-powered tools.
© 2025 MedPath, Inc. All rights reserved.