Relatlimab is a fully human immunoglobulin G4 (IgG4) monoclonal antibody that specifically binds to and inhibits lymphocyte-activation gene 3 (LAG-3), an immune checkpoint receptor expressed on activated T cells and tumor-infiltrating lymphocytes, thereby preventing LAG-3 from interacting with major histocompatibility complex class II (MHC-II) molecules on antigen-presenting cells.[1] This blockade reduces LAG-3-mediated suppression of T-cell function, enhancing antitumor immune responses, particularly when combined with programmed death-1 (PD-1) inhibitors like nivolumab.[2] Developed by Bristol Myers Squibb, relatlimab is approved by the U.S. Food and Drug Administration (FDA) as part of the fixed-dose combination product Opdualag (nivolumab and relatlimab-rmbw) for the treatment of unresectable or metastatic melanoma in adults and pediatric patients aged 12 years and older weighing at least 40 kg.[3] It represents the first approved LAG-3-targeted therapy and the third major class of immune checkpoint inhibitors after cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) and PD-1/PD-L1 inhibitors.[2]The mechanism of relatlimab involves disrupting the LAG-3/MHC-II interaction, which otherwise dampens T-cell activation and proliferation in the tumor microenvironment, leading to improved cytotoxic activity against cancer cells.[1] In combination with nivolumab, which blocks PD-1 signaling to further release T-cell brakes, the dual inhibition synergistically reactivates exhausted T cells, promotes tumor infiltration, and inhibits tumor growth more effectively than PD-1 monotherapy alone.[4] Preclinical studies demonstrated this synergy, showing enhanced antitumor effects in melanoma models, which paved the way for clinical development.[2]Relatlimab's development accelerated through the RELATIVITY clinical trial program, with the pivotal phase 2/3 RELATIVITY-047 trial (NCT03470922) evaluating nivolumab plus relatlimab versus nivolumab monotherapy in patients with previously untreated advanced melanoma.[5] The trial, involving 714 patients, met its primary endpoint of progression-free survival (PFS), with a median PFS of 10.2 months for the combination versus 4.6 months for nivolumab alone (hazard ratio [HR] 0.78; 95% CI, 0.65-0.93).[6] FDA approval of Opdualag occurred on March 18, 2022, based on these results, marking it as a first-line option regardless of BRAF mutation status and including it in National Comprehensive Cancer Network (NCCN) guidelines for metastatic melanoma.[3] As of November 2025, it remains a preferred regimen for eligible patients.[7]Long-term data from RELATIVITY-047, updated in July 2025 with 4 years of follow-up, confirmed sustained benefits, including a median overall survival (OS) of 53.3 months (95% CI, 34.0–NR) for the combination versus 33.2 months (95% CI, 25.2–45.8) for nivolumab alone (HR 0.77; 95% CI, 0.64–0.94), with 4-year OS rates of 52.0% (95% CI, 46.6–57.1) versus 42.8% (95% CI, 37.5–47.9).[6] The objective response rate was 43.9% (95% CI, 38.7–49.3) with the combination versus 33.4% (95% CI, 28.6–38.6), and no new safety concerns emerged, though grade 3/4 treatment-related adverse events occurred in 22.3% of combination recipients compared to 12.0% with nivolumab monotherapy.[6] Administered as an intravenous infusion every 4 weeks (fixed dose of 480 mg nivolumab and 160 mg relatlimab), Opdualag has transformed frontline therapy for advanced melanoma by offering improved outcomes with a manageable safety profile.[3] Ongoing trials explore its potential in other solid tumors, such as non-small cell lung cancer and hepatocellular carcinoma.[2]
Medical uses
Approved indications
Relatlimab is approved solely in fixed-dose combination with nivolumab under the brand name Opdualag for the treatment of unresectable or metastatic melanoma in adults and pediatric patients 12 years of age or older who weigh at least 40 kg.[8] This indication encompasses use as first-line therapy or in subsequent lines following prior anti-PD-1 therapy.[9]The U.S. Food and Drug Administration (FDA) approved Opdualag on March 18, 2022, under accelerated approval based on the phase 3 RELATIVITY-047 trial (NCT03470922), which enrolled 714 patients with previously untreated advanced melanoma.[8] In this randomized, double-blind study, the nivolumab-relatlimab combination significantly improved progression-free survival compared to nivolumab monotherapy, with a median of 10.1 months versus 4.6 months (hazard ratio 0.75; 95% confidence interval, 0.62 to 0.92; P=0.0055).[4] Updated analyses from the trial, with follow-up extending to 33.8 months as of 2024, confirmed durable benefits, including an objective response rate of 43.7% for the combination arm.[10] The pediatric inclusion in the approval relied on extrapolation from adultefficacydata, supported by pharmacokinetic modeling and safety profiles in adolescents.[8]The European Medicines Agency (EMA) authorized Opdualag on September 15, 2022, for the same patient population and indication as first-line treatment of unresectable or metastatic melanoma.[11] As of November 2025, no regulatory approvals exist for relatlimab monotherapy or for indications beyond advanced melanoma in major jurisdictions, including the FDA and EMA.[12]
Investigational uses
Relatlimab, approved in combination with nivolumab for unresectable or metastatic melanoma, is under investigation in several other cancer types, primarily in combination regimens to enhance immune checkpoint inhibition and address resistance mechanisms.In non-small cell lung cancer (NSCLC), the phase 3 RELATIVITY-1093 trial (NCT06561386) is evaluating nivolumab plus high-dose relatlimab combined with platinum-doublet chemotherapy as first-line treatment for metastatic non-squamous NSCLC, compared to pembrolizumab plus chemotherapy; the study remains ongoing as of November 2025 with no interim efficacy data reported.[13][14]For hepatocellular carcinoma (HCC), the phase 2 RELATIVITY-073 trial (NCT04567615) assesses nivolumab plus relatlimab versus nivolumab monotherapy in patients with advanced HCC who progressed on tyrosine kinase inhibitors but are immuno-oncology naive; preliminary safety and efficacy data have not been publicly disclosed, though the regimen aims to improve outcomes in this IO-resistant population.[15]01305-3/fulltext) Additionally, the phase 1/2 RELATIVITY-106 trial is exploring nivolumab plus relatlimab with bevacizumab as first-line therapy for unresectable HCC.[16]In the adjuvant setting for high-risk resected melanoma, the phase 3 RELATIVITY-098 trial (NCT05002569) compared nivolumab plus relatlimab to nivolumab alone but failed to meet its primary endpoint of improved recurrence-free survival, with topline results reported in early 2025; subgroup analyses suggest potential benefits in patients with higher LAG-3 expression, though no disease-free survival advantage was demonstrated overall.[17][18][19]Early-phase investigations include a phase 2 trial (NCT03607890) of nivolumab plus relatlimab in microsatellite instability-high (MSI-H) advanced solid tumors, showing promising activity in previously treated patients.[20] In head and neck squamous cell carcinoma, a phase 2 study (NCT04080804) is examining the safety and tolerability of nivolumab alone or with relatlimab or ipilimumab in resectable or recurrent disease.[21] Potential applications in triple-negative breast cancer are being explored in phase 1b trials, such as NCT06963905 combining sacituzumab govitecan with nivolumab plus or minus relatlimab.[22]As of November 2025, relatlimab has no additional regulatory approvals beyond melanoma, with research emphasizing combinations to overcome immunotherapy resistance across tumor types.
Adverse effects
Common adverse effects
In clinical trials, particularly the pivotal RELATIVITY-047 study, the most common adverse reactions (occurring in ≥20% of patients) to relatlimab combined with nivolumab were musculoskeletal pain (45%), fatigue (39%), rash (28%), pruritus (25%), and diarrhea (24%).[9] These effects were generally mild to moderate in severity, with grade 3 or 4 events reported in less than 5% of cases for each.[9]Mild immune-related adverse effects were also frequently observed, including hypothyroidism in 17% of patients and elevations in liver enzymes such as alanine aminotransferase (ALT) in 26% and aspartate aminotransferase (AST) in 30%.[9] Hypothyroidism typically required hormone replacement therapy and rarely led to treatment discontinuation (0.3%).[23] Elevated transaminases were often asymptomatic and managed through monitoring, with most cases resolving upon withholding therapy or using corticosteroids if needed.[23]These common adverse effects usually emerged within the first few treatment cycles, often during or shortly after infusion, and were manageable with supportive care such as analgesics for pain, topical treatments for skin reactions, or antidiarrheal agents.[23] The majority resolved without long-term sequelae, though some persisted beyond treatment discontinuation.[9]Overall, the profile of these common effects aligns closely with that seen in PD-1 inhibitor monotherapy, without introducing novel toxicities specific to LAG-3 inhibition.[4]
Serious adverse effects
Serious adverse effects of relatlimab, typically in combination with nivolumab as Opdualag, primarily involve immune-mediated reactions due to enhanced T-cell activation, occurring in approximately 10-15% of patients as grade 3-4 events across categories.[9] These high-grade reactions can lead to hospitalization, intervention, or discontinuation, with overall serious adverse reactions reported in 36% of patients and fatal outcomes in 0.8%. Long-term follow-up data from 2024 confirmed no new safety concerns, with grade 3/4 treatment-related adverse events occurring in 22% of patients receiving the combination compared to 12% with nivolumab monotherapy.[10] Key examples include colitis (7% all grades, 1.1% grade 3-4), pneumonitis (3.7% all grades, 0.6% grade 3-4), hepatitis (6% all grades, 4% grade 3-4), and endocrinopathies such as adrenal insufficiency (4.2% all grades, 1.4% grade 3-4) or type 1 diabetes mellitus (<1% all grades, 0.3% grade 3).[9][4]Rarer but potentially life-threatening events encompass myocarditis or pericarditis (<1% grade 3-4, with some fatal cases) and Guillain-Barré syndrome (rare, <1%).[9] Infusion-related reactions occur in 5.9-7% of patients overall, with grade 3-4 events in about 0.5%; symptoms may include fever, chills, hypotension, itching, or shortness of breath, requiring immediate attention during administration.[4][9]Management follows standardized guidelines for immune checkpoint inhibitors: initiate systemic corticosteroids (1-2 mg/kg/day prednisone or equivalent) for grade 2 or higher immune-mediated reactions, with withholding of therapy for grade 2 and permanent discontinuation for grade 4 or recurrent grade 3 events; endocrine disorders may require hormone replacement therapy.[9] For infusion reactions, interrupt or slow the infusion for mild to moderate symptoms and permanently discontinue for severe or life-threatening cases.[9] Monitoring includes baseline assessments of liver enzymes, creatinine, and thyroid function, along with periodic testing (e.g., monthly during therapy) and evaluation for autoantibodies if clinically indicated to detect early signs of toxicity.[9]
Pharmacology
Mechanism of action
Relatlimab is a human IgG4-kappa monoclonal antibody that specifically binds to lymphocyte activation gene-3 (LAG-3), an immune checkpoint receptor expressed on activated T cells, with high affinity (apparent KD of approximately 0.12 nM for the bivalent antibody).[24] This binding sterically blocks the interaction between LAG-3 and major histocompatibility complex (MHC) class II molecules on antigen-presenting cells and tumor cells, with an IC50 of 0.67 nM, thereby preventing LAG-3-mediated inhibitory signaling on tumor-infiltrating lymphocytes.[24] Due to its IgG4 isotype, modified with an S228P mutation to stabilize the hinge region, relatlimab exhibits minimal antibody-dependent cellular cytotoxicity (ADCC) and complement-dependent cytotoxicity (CDC) activity, ensuring its primary function is targeted blockade rather than effector-mediated cell depletion.[24]LAG-3 ligation by MHC class II normally downregulates T-cell receptor (TCR) signaling through intracellular immunoreceptor tyrosine-based inhibitory motifs (ITIMs) in its cytoplasmic tail, which recruit phosphatases like SHP-1 and SHP-2 to inhibit proximal TCR events such as ZAP-70 phosphorylation and downstream activation pathways.[25] By disrupting this interaction, relatlimab prevents LAG-3-mediated inhibition of CD8+ T-cell activation, leading to enhanced proliferation, cytokine secretion—including increased interferon-gamma (IFN-γ)—and cytotoxic granule release, such as granzyme B, in response to tumor antigens.[24][26]In the tumor microenvironment, LAG-3 is frequently co-expressed with programmed cell death protein 1 (PD-1) on exhausted CD8+ T cells in solid tumors, including melanoma, where it contributes to T-cell dysfunction and tumor immune escape by amplifying inhibitory signals.[27][28] Relatlimab synergizes with PD-1 blockade to reverse this exhaustion, restoring robust anti-tumor immunity through augmented T-cell effector functions without overlapping mechanisms.[29]
Pharmacokinetics
Relatlimab is administered by intravenous infusion over 30 minutes, achieving 100% bioavailability with maximum plasma concentrations reached immediately at the end of infusion. Steady-state concentrations are attained after approximately 16 weeks of every-4-week dosing.[9]The volume of distribution at steady state for relatlimab is 6.6 L (CV% 20%), with a central compartment volume estimated at approximately 3.5 L based on population pharmacokinetic modeling; this limited extravascular distribution is attributable to its large molecular weight of 148 kDa, which restricts tissue penetration primarily to the vascular and interstitial spaces.[9]As a monoclonal antibody, relatlimab undergoes catabolism via proteolytic degradation in the reticuloendothelial system to generate peptides and amino acids, without involvement of cytochrome P450 enzymes or other phase I/II metabolic pathways.[30]Relatlimab exhibits an effective elimination half-life of 26.2 days (CV% 37%), with steady-state clearance of 5.5 mL/h (CV% 41%), representing a 10% reduction from the initial dose clearance of 6 mL/h (CV% 39%); at doses below 160 mg every 4 weeks, pharmacokinetics are nonlinear due to target-mediated drug disposition, while exposures increase proportionally at higher doses.[9][31]Pharmacokinetics of relatlimab are unaffected by age (range 17–92 years), sex, race/ethnicity, or mild to moderate renal (eGFR 30–89 mL/min/1.73 m²) or hepatic impairment, though data for severe impairment are lacking. Anti-relatlimab antibodies developed in 5.6% of patients and neutralizing antibodies in 0.3%, with no observed impact on exposure. Co-administration with nivolumab does not alter relatlimab pharmacokinetics.[9]
Chemistry
Molecular structure
Relatlimab is a fully human immunoglobulin G4 (IgG4) kappa monoclonal antibody, consisting of two heavy chains of approximately 440 amino acids each and two kappa light chains of 214 amino acids, linked by interchain disulfide bonds.[32][33] It is produced recombinantly in Chinese hamster ovary (CHO) cells, a common expression system for therapeutic monoclonal antibodies that ensures proper folding and post-translational modifications.[32][9]The predominant molecular isoform of relatlimab features a calculated molecular formula of C6584H10106N1718O2102S38 (accounting for heavy chain N-terminal pyroglutamate, absence of C-terminal lysine, and the G0F/G0F glycoform), with a molecular weight of approximately 148 kDa.[32] It exhibits typical N-linked glycosylation at the Fc region, primarily the biantennary G0F glycoform (core-fucosylated, agalactosylated complex-type glycans at Asn297), which is characteristic of IgG4 antibodies produced in CHO cells and contributes to stability without an aglycosylated variant in clinical formulations.[32][33]Relatlimab was generated by immunizing HuMAb Mouse and KM Mouse transgenic mice with recombinant LAG-3 protein, followed by hybridoma screening to select clone 25F7, which was then expressed recombinantly with human IgG4 kappa constant regions.[34] It binds to the domain 1 (D1) of the extracellular region of lymphocyte activation gene-3 (LAG-3), specifically targeting the insertion loop encompassing residues H63–W70 to block ligand interactions.[24]
Physical properties
Relatlimab is provided as a component of the fixed-dose combination product Opdualag, a sterile, preservative-free injection supplied as a clear to opalescent, colorless to slightly yellow solution that may contain a few translucent-to-white particles.[35] Each single-dose 20 mL vial contains 80 mg of relatlimab (at a concentration of 4 mg/mL) co-formulated with 240 mg of nivolumab (12 mg/mL).[35]The formulation of Opdualag includes the excipients histidine (1.1 mg/mL), L-histidine hydrochloride monohydrate (2.7 mg/mL), sucrose (85.6 mg/mL), pentetic acid (0.008 mg/mL), and polysorbate 80 (0.5 mg/mL) in water for injection to support stability and prevent aggregation.[35] The solution has a pH of 5.8 and is designed for stability in aqueous buffers within a pH range of approximately 5.5 to 6.5.[35]For storage, unopened vials must be refrigerated at 2°C to 8°C in the original carton to protect from light, with no freezing permitted.[35] Once prepared for infusion (diluted or undiluted), the solution remains stable for up to 8 hours at room temperature under room light or up to 24 hours if refrigerated and protected from light.[35]
Clinical development
Preclinical research
Preclinical research on relatlimab, a human IgG4 monoclonal antibody targeting lymphocyte activation gene-3 (LAG-3), began with foundational studies elucidating LAG-3's role in suppressing antitumor immunity. In the early 2010s, researchers utilized LAG-3 knockout (Lag3^{-/-}) mice to demonstrate that LAG-3 deficiency enhances T-cell responses against tumors. Specifically, Lag3^{-/-}Pdcd1^{-/-} double-knockout mice exhibited significantly reduced tumor growth in B16 melanoma and MC38 adenocarcinoma models compared to single knockouts or wild-type controls, with 80% tumor clearance in high-dose B16 challenges and 83% survival in MC38 models, attributed to increased IFN-γ production by CD4^{+} and CD8^{+} T cells in tumor-draining lymph nodes.[36]Subsequent in vitro studies characterized relatlimab's binding and functional properties. Relatlimab binds human LAG-3 with high affinity, achieving an EC{50} of 0.49 nmol/L in ELISA assays and a K_D of 0.12 nmol/L via surface plasmon resonance, while showing no binding to murine LAG-3. It potently blocks LAG-3 interactions with its ligands, including fibrinogen-like protein 1 (FGL1) with an IC{50} of 0.019 nmol/L (<1 nM) and MHC class II with an IC_{50} of 0.67 nmol/L. Functionally, relatlimab restored IL-2 production in human peripheral blood mononuclear cells (PBMCs) stimulated with superantigen, with an IC_{50} of 1.05 nmol/L in T-cell hybridoma assays, thereby alleviating LAG-3-mediated T-cell suppression.[37]In vivo efficacy was evaluated using surrogate anti-LAG-3 antibodies in syngeneic mouse tumor models. In the MC38 colon adenocarcinoma model, the surrogate antibody alone achieved approximately 60% tumor growth inhibition (TGI), while combination with anti-PD-1 blockade resulted in up to 90-100% TGI and increased tumor-free survival (7-8/10 mice), demonstrating synergistic antitumor effects beyond PD-1 monotherapy. Similar enhancements were observed in the SA1N sarcoma model, where dual blockade reduced tumor burden more effectively than single agents. These findings supported relatlimab's potential to augment checkpoint inhibition in immunocompetent hosts.[30][37]Safety profiling included repeat-dose toxicology studies in cynomolgus monkeys, which express cross-reactive LAG-3 (though with lower affinity, EC_{50} 29 nmol/L). In a 13-week (3-month) study, relatlimab at doses up to 100 mg/kg weekly, alone or combined with nivolumab (50 mg/kg), was generally well-tolerated, with a no-observed-adverse-effect level (NOAEL) of ≥30 mg/kg—exceeding clinical exposures by about 9-fold. Target-related effects included reversible lymphocyte modulation, such as reduced LAG-3^{+} T cells, and mild, non-adverse changes in lymphoid tissues; one instance of reversible CNS vasculitis occurred in the combination arm but was not dose-limiting. No off-target toxicities were noted.[30][37]Relatlimab demonstrated high selectivity, binding exclusively to the D1 domain of human LAG-3 (epitope H63-W70) without cross-reactivity to related immune checkpoints like PD-1 or TIM-3, or other unrelated proteins, as confirmed by binding assays and tissue cross-reactivity studies limited to expected lymphoid expression patterns.[37]
Clinical trials
The clinical development of relatlimab has progressed through several key trials evaluating its safety, dosing, and efficacy primarily in combination with nivolumab for advanced melanoma and other solid tumors. The phase 1/2a RELATIVITY-020 trial (NCT01968109), an open-label dose-escalation and cohort-expansion study, assessed relatlimab monotherapy and in combination with nivolumab in patients with advanced solid tumors. Doses of relatlimab ranged from 20 mg to 800 mg every 2 weeks for monotherapy and up to 1440 mg every 4 weeks in combination with nivolumab (80 mg to 480 mg). Within this trial, part E evaluated the fixed-dose combination, confirming the recommended phase 2 dose of 160 mg relatlimab every 4 weeks with 480 mg nivolumab every 4 weeks based on favorable safety and pharmacokinetic profiles. The maximum tolerated dose was not reached.[30]The expansion cohorts of RELATIVITY-020 evaluated the combination in 518 patients with advanced melanoma that had progressed on prior anti-PD-1/PD-L1 therapy. In the cohort with one prior anti-PD-1 regimen (n=351 evaluable), the objective response rate was 12.0% (95% CI, 8.8-15.8), indicating modest activity in PD-1-refractory settings. The regimen demonstrated a manageable safety profile, with grade 3-4 treatment-related adverse events occurring in 15.0% of these patients.[38]The pivotal phase 3 RELATIVITY-047 trial (NCT03470922) randomized 714 previously untreated patients with unresectable or metastatic melanoma in a 1:1 ratio to receive nivolumab plus relatlimab or nivolumab alone. The primary endpoint of progression-free survival was met, with a hazard ratio of 0.75 (95% CI, 0.62-0.92; P=0.006) favoring the combination. At 2 years, overall survival rates were 63.7% (95% CI, 58.1-68.7) with the combination versus 58.3% (95% CI, 52.7-63.4) with nivolumab monotherapy.[4][39]Supportive analyses from RELATIVITY-047 highlighted consistent benefits across subgroups, with stronger responses observed in BRAF wild-type patients compared to BRAF-mutant subgroups. Safety data indicated that 94% of treatment-related adverse events were manageable, with grade 3-4 events in 22.0% of patients on the combination versus 12.0% on nivolumab alone; no new safety signals emerged. Updates as of July 2025, including 4-year follow-up data, confirmed the durability of these outcomes, with 4-year overall survival rates of approximately 52% versus 43%.[10][40]
History
Discovery and development
The foundational research on lymphocyte activation gene-3 (LAG-3), the immune checkpoint targeted by relatlimab, was advanced by Dr. Lieping Chen and his team at Johns Hopkins University during the 1990s and 2010s, establishing LAG-3's role in suppressing T-cell responses and its potential as a target for cancer immunotherapy.[41]Bristol-Myers Squibb (BMS) developed relatlimab (BMS-986016), a fully human immunoglobulin G4 monoclonal antibody against LAG-3, internally as part of its immuno-oncology efforts. The candidate entered clinical development with the initiation of a phase 1 trial (NCT01968109) in November 2013, marking the first administration to patients to evaluate safety, tolerability, and pharmacokinetics as monotherapy and in combination with nivolumab.[42]Early phase 1/2 data supported accelerated progression, leading to the launch of the pivotal phase 2/3 RELATIVITY-047 trial (NCT03470922) in April 2018, which evaluated relatlimab plus nivolumab versus nivolumab alone in untreated advanced melanoma. In November 2019, BMS completed its $74 billion acquisition of Celgene Corporation, incorporating relatlimab into an expanded immuno-oncology pipeline that emphasized combination therapies.[43][5]Development gained momentum with presentations at key conferences, including ASCO 2024, where interim data on relatlimab-containing triplets (nivolumab, relatlimab, and ipilimumab) in advanced solid tumors highlighted promising safety and activity profiles, further fueling industry interest. No significant development setbacks, such as halts or major safety signals beyond expected immune-related events, were reported during this period.[44]
Regulatory approvals
The U.S. Food and Drug Administration (FDA) accepted the Biologics License Application for Opdualag (nivolumab and relatlimab-rmbw), a fixed-dose combination, for priority review on September 20, 2021, with a target action date of March 19, 2022.[45] The FDA granted accelerated approval to Opdualag on March 18, 2022, for the treatment of adult and pediatric patients 12 years of age or older with unresectable or metastatic melanoma, based on demonstrated improvement in progression-free survival as a surrogate endpoint from the phase 2/3 RELATIVITY-047 trial.[8] Prior to approval, the combination received fast track designation on November 20, 2019, and orphan drug designation on August 18, 2017, for the treatment of stage IIb to IV melanoma.[46][47] As of November 2025, the approval remains accelerated, with confirmatory overall survival data from RELATIVITY-047 anticipated in 2026 to support potential conversion to full approval.[39]In the European Union, the European Medicines Agency (EMA) validated the marketing authorization application for Opdualag in October 2021, following a positive opinion from the Committee for Medicinal Products for Human Use in July 2022.[48] The European Commission granted marketing authorization on September 15, 2022, for the first-line treatment of advanced melanoma in adults and adolescents aged 12 years and older whose tumors express PD-L1 <1% tumor cell expression.[48] This authorization was based on progression-free survival data from the RELATIVITY-047 trial, with overall survival as a key confirmatory endpoint.Health Canada issued a Notice of Compliance for Opdualag on September 13, 2023, authorizing its use for adult and pediatric patients 12 years of age or older with unresectable or metastatic melanoma who have not received prior systemic therapy.[49] The Therapeutic Goods Administration (TGA) in Australia registered Opdualag on the Australian Register of Therapeutic Goods on October 7, 2022, for the same indication in patients 12 years and older.[50]Opdualag was approved by Japan's Pharmaceuticals and Medical Devices Agency (PMDA) in 2025, with submission of phase 3 data from RELATIVITY-047 supporting evaluation for approval in unresectable or metastatic melanoma.[51]
Society and culture
Brand names
Relatlimab is the international nonproprietary name (INN) assigned to this monoclonal antibody targeting lymphocyte-activation gene 3 (LAG-3), as proposed by the World Health Organization in 2018.[52] In the United States, it holds the United States Adopted Name (USAN) of relatlimab-rmbw, where the suffix -rmbw denotes a recombinant monoclonal antibody produced with a human immunoglobulin framework.[8] This nomenclature adheres to the WHO guidelines for biological and biotechnological substances, which standardize naming for monoclonal antibodies to ensure clarity in scientific and regulatory contexts.[52]The primary trade name associated with relatlimab is Opdualag, which refers to the fixed-dose combination product containing relatlimab and nivolumab approved for treating unresectable or metastatic melanoma.[8] Relatlimab is not marketed under a separate brand name as a standalone agent, as its regulatory approvals have been limited to this dual-immunotherapy formulation.[53]In scientific literature and clinical studies, relatlimab is commonly abbreviated as an anti-LAG-3 monoclonal antibody (mAb) due to its mechanism of action in blocking the LAG-3 immune checkpoint.[54] It was also known by its developmental code BMS-986016 during early research and trials conducted by Bristol Myers Squibb.[54]
Availability and access
Relatlimab is available as part of the fixed-dose combination Opdualag (nivolumab and relatlimab-rmbw), which launched in the United States in April 2022 following FDA approval for adult and pediatric patients aged 12 years and older with unresectable or metastatic melanoma.[53] In the European Union, EU-wide availability commenced in 2023 after European Commission approval in September 2022 for the same indication in adults and adolescents aged 12 years and older.[48] Opdualag has also received approvals in Canada (March 2023), Japan (September 2023), and Australia (November 2023) for similar indications.[55][56][57] As of 2025, access remains primarily limited to high-income countries, with distribution focused on regions with established reimbursement frameworks.[58]In the United States, the wholesale acquisition cost (WAC) for the full dose of Opdualag (480 mg nivolumab and 160 mg relatlimab, administered as two 20 mL vials every 4 weeks) is approximately $30,844 (as of July 2025), resulting in an estimated annual cost of approximately $400,972 based on 13 doses per year.[59] Discounts are available through the 340B Drug Pricing Program, which provides eligible safety-net providers with reductions of around 50% off the wholesale price.[60]Medicare Part B covers Opdualag for eligible patients, often at 100% after applicable deductibles when combined with supplemental insurance or assistance programs.[61] In the United Kingdom, the National Institute for Health and Care Excellence (NICE) approved reimbursement in 2024 following a cost-effectiveness analysis that supported its use in untreated advanced melanoma.[62]Bristol Myers Squibb offers patient support programs, including the BMS Access Support Co-Pay Assistance Program, which provides financial assistance with out-of-pocket costs for eligible commercially insured patients.[63]Expanded access programs for investigational use concluded in 2022 upon commercial availability.[30]Access challenges persist due to the high cost, which restricts availability in low- and middle-income countries where alternative therapies like chemotherapy remain more feasible.[58] No generic or biosimilar equivalents exist as of 2025, given the recent approval and biologic nature of the drug.[48] Supply chains have remained stable, with no reported shortages impacting distribution in approved markets.[64]