MedPath

Tebotelimab Advanced Drug Monograph

Published:Sep 8, 2025

Generic Name

Tebotelimab

Drug Type

Biotech

CAS Number

2245725-04-4

Tebotelimab (MGD013): A Comprehensive Profile of a PD-1 x LAG-3 Bispecific DART® Molecule in Oncology

Section 1: Executive Summary

Tebotelimab, also known by its code name MGD013, is an investigational, first-in-class biologic agent developed by MacroGenics.[1] It is engineered as a humanized, Fc-bearing, bispecific, tetravalent Dual-Affinity Re-Targeting (DART®) molecule designed to simultaneously bind and inhibit two distinct immune checkpoint proteins: Programmed Cell Death 1 (PD-1) and Lymphocyte Activation Gene-3 (LAG-3).[1] The core scientific premise behind Tebotelimab is that dual blockade of these non-redundant inhibitory pathways can more effectively restore the function of exhausted T-cells and potentiate a robust anti-tumor immune response than targeting either pathway alone.[5]

The development of Tebotelimab presents a compelling case study in modern immuno-oncology, illustrating a significant gap between a strong preclinical rationale and the subsequent clinical reality. Preclinical studies suggested that the bispecific DART® format of Tebotelimab could induce a more potent synergistic T-cell activation compared to the co-administration of separate anti-PD-1 and anti-LAG-3 antibodies.[7] This finding provided a powerful justification for its development. In clinical trials, Tebotelimab has consistently demonstrated a predictable and manageable safety profile across multiple studies, both as a monotherapy and in various combination regimens. Its adverse event profile is largely consistent with the established anti-PD-1 class of agents, and a maximum tolerated dose was not reached in dose-escalation studies, indicating good tolerability.[3]

Despite this acceptable safety profile, the clinical efficacy of Tebotelimab has been modest in most settings investigated. Key trials in advanced hepatocellular carcinoma (aHCC) and metastatic gastric cancer yielded low objective response rates (ORRs), leading to the conclusion that the drug's activity was primarily limited to disease stabilization.[5] Consequently, the developmental trajectory of Tebotelimab has been characterized by the systematic termination and discontinuation of clinical programs across a wide spectrum of indications, including liver, gastric, biliary, and head and neck cancers, as well as melanoma.[2] For many of these indications, the developers have explicitly stated that no further clinical trials are planned.[5] A notable exception to this trend was an encouraging preliminary efficacy signal observed in a Phase 1 study combining Tebotelimab with margetuximab, an Fc-engineered anti-HER2 antibody, in patients with advanced HER2-positive malignancies.[3]

Ultimately, the combination of underwhelming efficacy data across most of its clinical program and the successful market entry of a competitor dual checkpoint blockade—the co-formulation of nivolumab (anti-PD-1) and relatlimab (anti-LAG-3)—has rendered Tebotelimab's development path commercially challenging. The drug's innovative molecular design and sound preclinical hypothesis did not translate into the superior clinical benefit required to displace or compete with established and emerging therapeutic options.

Section 2: Scientific Foundation, Molecular Architecture, and Mechanism of Action

The Immunological Rationale for Dual PD-1 and LAG-3 Blockade

The foundation of modern cancer immunotherapy rests on overcoming the mechanisms that tumors use to evade the host immune system. A primary mechanism of this evasion is the upregulation of immune checkpoint pathways, which act as natural "brakes" to prevent excessive immune responses but can be co-opted by cancer cells to induce T-cell tolerance. The most well-characterized of these is the PD-1 pathway. Programmed Cell Death 1 (PD-1; CD279) is an inhibitory receptor expressed on the surface of activated T-cells. When it binds to its ligands, PD-L1 or PD-L2, which are often expressed on tumor cells, it transmits an inhibitory signal that suppresses T-cell proliferation, cytokine production, and cytotoxic activity, leading to a state of T-cell "exhaustion".[1]

Lymphocyte Activation Gene-3 (LAG-3; CD223) is another critical inhibitory receptor that contributes to T-cell exhaustion.[1] Like PD-1, it is upregulated on T-cells following chronic antigen stimulation, a common feature within the tumor microenvironment. LAG-3's primary ligand is Major Histocompatibility Complex (MHC) Class II, and its engagement also delivers a negative regulatory signal to the T-cell.[4] Importantly, PD-1 and LAG-3 represent distinct and non-redundant inhibitory pathways.[17] Their co-expression on tumor-infiltrating lymphocytes (TILs) is a hallmark of profound T-cell dysfunction, and studies have shown that these two pathways can synergistically suppress anti-tumor immunity.[15]

This understanding forms the basis for the therapeutic hypothesis of dual checkpoint blockade. If inhibiting PD-1 alone is insufficient to fully restore T-cell function due to concurrent inhibition via the LAG-3 pathway, then simultaneously blocking both checkpoints should lead to a more complete and potent reactivation of the anti-tumor immune response.[5] This strategy aims to overcome both primary and acquired resistance to single-agent PD-1 inhibitors by targeting a key compensatory resistance mechanism.[21] The expected outcome of successful dual blockade is a robust enhancement of cytotoxic T-lymphocyte (CTL) proliferation, increased production of effector cytokines like interferon-gamma (IFN-γ), and ultimately, more effective tumor cell lysis.[1]

Molecular Architecture of Tebotelimab (MGD013): A DART® Molecule

To execute this dual blockade strategy with a single therapeutic agent, Tebotelimab was engineered using MacroGenics' proprietary Dual-Affinity Re-Targeting (DART®) platform.[4] The DART® technology enables the creation of antibody-like proteins that can simultaneously bind two different target antigens with high affinity and specificity.

Tebotelimab is specifically characterized as a humanized, Fc-bearing, bispecific, tetravalent DART® molecule.[1] Its key structural features include:

  • Bispecificity: It is engineered with two distinct antigen-binding domains, one targeting human PD-1 and the other targeting human LAG-3.[1]
  • Tetravalency: The molecule is designed to be bivalent for each of its targets, meaning it has two binding sites for PD-1 and two for LAG-3, which can enhance avidity and receptor engagement.[3]
  • Fc Domain: It incorporates a humanized IgG4κ crystallizable fragment (Fc) domain.[7] The IgG4 isotype is specifically chosen for its reduced effector functions, such as antibody-dependent cell-mediated cytotoxicity (ADCC), which minimizes the risk of depleting the target T-cells. The primary role of the Fc domain in this context is to engage the neonatal Fc receptor (FcRn), thereby extending the molecule's serum half-life and allowing for less frequent dosing.[7]
  • Molecular Weight and Composition: Tebotelimab has an approximate molecular weight of 166.7 kDa.[11] It is produced as a recombinant protein in Chinese Hamster Ovary (CHO) cells.[25]

The molecule demonstrates high-affinity binding to both of its targets. In vitro assays using NS0 cells showed a half-maximal effective concentration (EC50​) of 1.65 nM for binding to cell-surface expressed PD-1 and 0.41 nM for binding to cell-surface expressed LAG-3.[24]

Pharmacodynamics: Translating Structure to Function

Upon intravenous administration, Tebotelimab is designed to engage PD-1 and LAG-3 expressed on the surface of T-cells within the tumor microenvironment and peripheral lymphoid organs.[1] By physically occupying the binding sites on these receptors, it prevents their interaction with their respective ligands: PD-1 is blocked from engaging with PD-L1 and PD-L2, and LAG-3 is blocked from engaging with MHC Class II.[4] This dual blockade effectively removes two simultaneous inhibitory signals, thereby lowering the threshold for T-cell activation and restoring the capacity of exhausted CTLs to recognize and eliminate tumor cells.[1]

A central element of the scientific rationale for developing Tebotelimab as a single bispecific molecule, rather than simply co-administering two separate antibodies, was derived from compelling preclinical evidence. In vitro experiments using human peripheral blood mononuclear cells (PBMCs) demonstrated that Tebotelimab induced a greater synergistic T-cell activation, as measured by the secretion of IFN-γ, when compared directly to the combination of its individual constituent anti-PD-1 and anti-LAG-3 monoclonal antibodies.[7] This finding suggested that the specific architecture of the DART® molecule, which physically links the two binding domains, might facilitate more efficient co-ligation or blockade on the T-cell surface, leading to a qualitatively superior immune response.

This preclinical observation, however, stands in stark contrast to the eventual clinical outcomes observed in the field of dual checkpoint inhibition. The development of Tebotelimab was predicated on the hypothesis that its bispecific format was inherently superior to a co-administration approach. Yet, its clinical program was largely marked by limited efficacy and widespread discontinuation. Concurrently, the combination of two separate monoclonal antibodies—nivolumab (anti-PD-1) and relatlimab (anti-LAG-3)—demonstrated a significant clinical benefit in a pivotal Phase 3 trial (RELATIVITY-047), leading to its regulatory approval as the combination product Opdualag.[6] This presents a fundamental paradox where the therapeutic strategy deemed inferior in preclinical models ultimately proved successful in late-stage clinical development, while the supposedly superior bispecific agent did not. This disconnect suggests that

in vitro measures of potency, such as IFN-γ secretion, may not be reliable predictors of clinical success for complex immunotherapies. It is plausible that factors not fully captured by these assays, such as in vivo pharmacokinetics, differential tissue penetration, receptor occupancy dynamics over time, or subtle conformational effects of the DART® structure, played unforeseen roles that negated the theoretical advantage of the bispecific format. This outcome serves as a critical lesson for the future development of multi-targeting immunotherapies, emphasizing the limitations of preclinical models in predicting complex in vivo biology.

Section 3: Clinical Development as a Monotherapy

The Cornerstone Trial: Phase 1 First-in-Human Study (NCT03219268)

The clinical journey of Tebotelimab began with a comprehensive Phase 1, first-in-human, open-label study (NCT03219268) designed to establish its foundational safety, tolerability, and pharmacokinetic profile.[31] This multi-part study enrolled patients with unresectable or metastatic solid tumors and hematologic malignancies who had progressed on or were refractory to standard therapies.[10]

The primary objectives of the study were to characterize the safety and tolerability of Tebotelimab and to determine its maximum tolerated dose (MTD) and the recommended Phase 2 dose (RP2D) for both monotherapy and combination therapy settings.[11] The study employed a standard 3+3 dose-escalation design, evaluating a wide range of intravenous doses from 1 mg up to 1200 mg, typically administered once every two weeks (Q2W).[7] A key finding from this phase was that Tebotelimab was well-tolerated across all dose levels tested. No dose-limiting toxicities were observed that would define an MTD. Based on an integrated assessment of safety, pharmacokinetics, and pharmacodynamics, the RP2D for Tebotelimab monotherapy was established as 600 mg Q2W.[10] Following the dose-escalation phase, the study proceeded to enroll multiple disease-specific expansion cohorts to further evaluate safety and gather preliminary signals of anti-tumor activity.[10]

Monotherapy Safety and Tolerability Profile

The safety profile of Tebotelimab monotherapy was characterized in a population of 269 treated patients from the NCT03219268 study.[10] The overall safety data demonstrated that the drug was generally well-tolerated, with a profile consistent with that of other anti-PD-1/PD-L1 checkpoint inhibitors.

Treatment-related adverse events (TRAEs) of any grade were reported in 68% (184 of 269) of patients.[10] Severe (Grade ≥3) TRAEs occurred in 22% of patients.[10] The most common TRAEs reported were typical of the drug class and included fatigue, pyrexia, nausea, diarrhea, and rash.[3] Immune-related adverse events (irAEs), such as hypothyroidism and hyperthyroidism, were also observed and were considered manageable with standard supportive care and treatment algorithms, such as hormone replacement or corticosteroids.[3] Treatment discontinuation due to an adverse event was necessary in 13.7% of patients, and treatment-related deaths were infrequent.[10] Overall, the safety data did not reveal any unexpected toxicities and supported the continued development of Tebotelimab at the 600 mg Q2W dose.

Monotherapy Antitumor Activity

While the primary focus of the Phase 1 study was safety, secondary endpoints included preliminary assessments of anti-tumor activity. The results were mixed, showing modest activity overall but with intriguing signals in specific, highly refractory patient populations. In the dose-escalation cohorts, evidence of biological activity was observed, with tumor size reductions reported in 34% (59 of 172) of response-evaluable patients.[10]

The most compelling efficacy signals for monotherapy emerged from cohorts of patients with immune systems already heavily challenged by prior therapies. Objective responses were documented in patients with solid tumors that were refractory to prior anti-PD-1 therapy, suggesting that the dual blockade of LAG-3 could potentially overcome one mechanism of resistance to single-agent PD-1 inhibition.[10] Even more notably, significant activity was seen in patients with relapsed/refractory diffuse large B-cell lymphoma (DLBCL), including those who had previously failed chimeric antigen receptor (CAR)-T cell therapy.[7] In this heavily pre-treated DLBCL population, a preliminary ORR of 53.8% (7 of 13 evaluable patients) was reported, including two complete responses in post-CAR-T patients.[17]

Exploratory biomarker analyses provided further context for these responses. Clinical activity appeared to be associated with higher baseline expression of LAG-3 on tumor-infiltrating lymphocytes as measured by immunohistochemistry (IHC) and the presence of a pre-existing inflammatory tumor microenvironment, as indicated by an IFN-γ gene signature.[7] This suggests that Tebotelimab's activity is dependent on a T-cell-inflamed tumor phenotype, where its target receptors are present and functional.

These findings reveal a critical dichotomy in Tebotelimab's clinical profile. On one hand, its failure to generate compelling activity in broader, less selected patient populations ultimately led to the discontinuation of most of its development programs. On the other hand, the clear signals of activity in extremely difficult-to-treat, immune-exhausted niches—such as post-PD-1 and post-CAR-T settings—confirm that the drug is biologically active and can mediate clinically meaningful responses under the right immunological conditions. The molecule's mechanism appears to be valid, but the specific conditions required for that mechanism to translate into a robust clinical response may be confined to a therapeutic window that is too narrow to support a broad development strategy. This suggests a potential strategic mismatch, where the drug was tested in wide-ranging indications rather than being focused on a biomarker-driven approach in the specific refractory niches where it showed the most promise. The ultimate decision to halt development implies that the commercial opportunity within these potential niches was deemed insufficient to justify the continued investment.

Section 4: Evaluation in Combination Therapy Regimens

Recognizing the potential for synergistic activity and the established paradigm of combination therapy in oncology, Tebotelimab was evaluated in multiple clinical trials alongside other anti-cancer agents. These studies explored its utility in various tumor types and settings, yielding a range of outcomes from encouraging preliminary signals to definitive program terminations.

4.1. Advanced Hepatocellular Carcinoma (aHCC) - Monotherapy (NCT04212221)

Although technically a monotherapy trial, the study in aHCC is best understood in the context of combination strategies, as it specifically evaluated Tebotelimab in patients who had failed prior therapies, including other immunotherapies. This open-label, single-arm, Phase 1/2 study (NCT04212221) was conducted in patients with advanced hepatocellular carcinoma who had received at least one prior line of systemic treatment.[5] The dose expansion phase was critically divided into two cohorts: one for patients previously treated with immune checkpoint inhibitors (CPI-experienced) and one for those who had not received prior CPIs (CPI-naïve).[5]

The efficacy results were modest and highlighted the challenges of treating this patient population.

  • In the CPI-experienced cohort (n=30 evaluable), the ORR was 3.3%, with one confirmed partial response (PR). The disease control rate (DCR), which includes stable disease, was 50.0%, and the median progression-free survival (PFS) was 2.4 months.[5]
  • In the CPI-naïve cohort (n=30 evaluable), the results were slightly better but still limited, with an ORR of 13.3% (four confirmed PRs), a DCR of 46.7%, and a median PFS of 3.1 months.[5]

The safety profile was considered manageable, with 18.8% of patients in the expansion phase experiencing Grade ≥3 TRAEs. However, one treatment-related death (1.4%) was reported.[5] The study's conclusion was stark: while Tebotelimab demonstrated a manageable safety profile, its antitumor activity was primarily limited to disease stabilization. Consequently, the investigators and sponsor concluded that "No additional clinical trials are planned at this time," effectively marking the end of Tebotelimab's development in aHCC.[5]

4.2. HER2+ Malignancies - Combination with Margetuximab (NCT03219268)

One of the most promising clinical signals for Tebotelimab came from a combination cohort within the main Phase 1 study (NCT03219268), which paired it with margetuximab.[3] Margetuximab is an anti-human epidermal growth factor receptor 2 (HER2) monoclonal antibody that has been Fc-engineered to enhance binding to activating Fcγ receptors, thereby increasing ADCC. The scientific rationale for this combination was compelling:

in vitro studies showed that exposure to margetuximab led to the upregulation of LAG-3 and PD-L1 on immune cells. The hypothesis was that margetuximab could initiate an immune attack on HER2-positive tumor cells, and Tebotelimab could then sustain and deepen this response by preventing the subsequent T-cell exhaustion signaled through PD-1 and LAG-3.[3]

In this study, patients with various advanced HER2-positive malignancies who had received a median of two prior lines of therapy were treated with Tebotelimab (at doses of 300 mg or 600 mg Q3W) plus margetuximab (15 mg/kg Q3W).[3] The preliminary efficacy results were encouraging:

  • Among 20 response-evaluable patients, a preliminary ORR of 40% (8 objective responses) was observed, with 6 of these responses being confirmed.[3]
  • The responses were seen across a range of difficult-to-treat HER2-positive tumors, including a confirmed complete response in cholangiocarcinoma and partial responses in breast cancer, microsatellite stable colorectal cancer, ovarian cancer, and gastroesophageal junction carcinoma.[3]
  • Notably, this activity was observed even in a patient population with predominantly low PD-L1 expression, suggesting the combination could be effective in "cold" tumor microenvironments.[3]

The combination demonstrated an acceptable safety profile, with Grade 3 TRAEs occurring in 19.4% of patients and no Grade 4 or 5 TRAEs reported.[3] Despite these promising early results, a clear path forward for this combination has not been publicly announced, and MacroGenics has since stated it has no active, ongoing studies for Tebotelimab.[38]

4.3. Advanced Gastric Cancer - Combination with Niraparib (NCT04178460)

Based on preclinical data suggesting potential synergy, a Phase 1 study (NCT04178460) was initiated to evaluate Tebotelimab in combination with niraparib, a poly (ADP-ribose) polymerase (PARP) inhibitor, in patients with locally advanced or metastatic gastric cancer who had failed at least two prior lines of therapy.[8]

However, the clinical results did not bear out the preclinical hypothesis. In 19 evaluable patients treated at the RP2D, the combination demonstrated "limited antitumor activity," with a confirmed ORR of only 5.3% (one PR) and a DCR of 52.6%.[8] The median PFS was 2.7 months, and median overall survival was 6.5 months.[8] While the safety profile was deemed acceptable, the lack of meaningful efficacy led to the termination of the trial and the conclusion that "No further clinical trials are planned" for this combination in gastric cancer.[8]

4.4. Head and Neck Squamous Cell Carcinoma (SCCHN) - Combination with Enoblituzumab (NCT04634825)

Another combination strategy involved pairing Tebotelimab with enoblituzumab, an investigational anti-B7-H3 antibody. A Phase 2 trial (NCT04634825) was designed to evaluate this combination as a first-line treatment for patients with recurrent or metastatic SCCHN whose tumors were PD-L1 negative (Combined Positive Score < 1).[22] The rationale was to use a novel immunotherapy combination in a patient population less likely to respond to standard anti-PD-1 monotherapy. However, this trial was ultimately

terminated, as documented in clinical trial registries.[14] The termination of a Phase 2 study typically signals either a lack of efficacy, unacceptable toxicity, or a strategic decision by the sponsor to halt development in that indication, further contributing to the pattern of discontinuation for Tebotelimab.

Section 5: Consolidated Clinical Development Status and Regulatory Trajectory

The clinical development program for Tebotelimab was broad in scope, investigating the agent as both a monotherapy and in multiple combination regimens across a diverse range of solid tumors and hematologic malignancies. Despite its innovative design and strong preclinical rationale, the program has been largely curtailed due to a consistent pattern of limited clinical efficacy. The following table provides a consolidated overview of the key clinical trials and their ultimate status, illustrating the drug's challenging developmental journey.

IndicationNCT IdentifierPhaseSettingHighest Development StatusKey Outcome / Reason for Discontinuation
Advanced Solid Tumors & Hematologic MalignanciesNCT032192681Monotherapy & Combination (Margetuximab)CompletedEstablished RP2D and manageable safety profile. Showed modest monotherapy efficacy but encouraging preliminary signals with margetuximab. 31
Advanced Hepatocellular Carcinoma (aHCC)NCT042122211/2MonotherapyCompletedEfficacy was limited, primarily disease stabilization. No further clinical trials planned. 5
HER2+ Gastric/GEJ Cancer (MAHOGANY study)NCT040823642/3Combination (Tebotelimab + Chemo)CompletedEnrollment in the tebotelimab-containing arm (Module B) was discontinued by partner Zai Lab based on clinical data and changing treatment landscape. 16
Head and Neck Squamous Cell Carcinoma (SCCHN)NCT046348252Combination (Enoblituzumab)TerminatedTrial was stopped prematurely, halting development in this indication. 14
Unresectable or Metastatic MelanomaNCT046530381MonotherapyTerminatedTrial was stopped prematurely, halting development in this indication. 13
Advanced Solid Tumors (incl. Gastric, TNBC)NCT041784601Combination (Niraparib)TerminatedCombination demonstrated limited antitumor activity. No further clinical trials planned. 8
Various Cancers (Biliary, Endometrial, etc.)N/A2N/ADiscontinuedDevelopment was reported as discontinued for multiple other indications, including biliary cancer, cholangiocarcinoma, endometrial cancer, and triple-negative breast cancer. 2

Regulatory Status

As an investigational agent, Tebotelimab has been studied under Investigational New Drug (IND) applications with the U.S. Food and Drug Administration (FDA) and equivalent regulatory filings in other regions, such as the European Economic Area (EEA).[41] However, its clinical development has not progressed to a point of marketing authorization.

  • Marketing Approvals: Tebotelimab has not received marketing approval from the FDA, the European Medicines Agency (EMA), or any other major global regulatory authority. It remains an investigational drug.[40]
  • Special Designations: There is no evidence that Tebotelimab has been granted any expedited regulatory pathway designations. It does not hold an Orphan Drug Designation from the FDA.[2] Furthermore, the available documentation does not indicate that it has received Breakthrough Therapy Designation, Fast Track, or Priority Review status for any indication.[50]

The widespread discontinuation of its clinical programs makes it highly unlikely that Tebotelimab will be the subject of a Biologics License Application (BLA) or Marketing Authorisation Application (MAA) in the foreseeable future.

Section 6: Expert Analysis, Competitive Landscape, and Future Perspective

Interpreting the Pattern of Discontinuation

The clinical development history of Tebotelimab is a narrative of a promising scientific concept confronting the high bar of clinical translation. The consistent pattern across multiple terminated and discontinued trials reveals a recurring theme: the drug demonstrated an acceptable and manageable safety profile, but this was coupled with limited or insufficient antitumor efficacy to justify continued development.[5] This outcome, repeated across different tumor types and combination strategies, points to a portfolio-level strategic decision by MacroGenics and its partner Zai Lab. The conclusion appears to be that Tebotelimab's therapeutic index—the balance between its efficacy and toxicity—was not compelling enough to warrant the substantial investment required for late-stage clinical trials, particularly in a highly competitive immuno-oncology landscape. The one area of encouraging preliminary activity, in combination with margetuximab, has not yet led to a publicly disclosed pivotal development plan, and recent corporate updates indicate no active studies are ongoing, suggesting even this niche opportunity may not be pursued.[3]

The Bispecific Burden of Proof

A critical factor influencing the fate of Tebotelimab is the inherently higher burden of proof faced by a novel bispecific antibody compared to a combination of two separate monoclonal antibodies. A bispecific agent is a single, new molecular entity, requiring a complex and costly development and manufacturing process. To justify this, it must demonstrate a clear and substantial advantage over the more straightforward approach of co-administering two individual drugs. This advantage could manifest as significantly superior efficacy, a markedly improved safety profile, or a major improvement in convenience that drives clinical adoption.

Tebotelimab failed to clear this high bar. The clinical data generated did not show it to be significantly more effective than what might be expected from standard immunotherapies. Its safety profile, while acceptable, was not demonstrably superior to the class. While a single infusion is more convenient than two, this is a minor benefit in the absence of compelling efficacy. This challenge was amplified by the emergence of a successful competitor that validated the co-administration strategy. The approval of Opdualag (nivolumab plus relatlimab) established a new clinical benchmark. Tebotelimab's modest results were therefore not evaluated in a vacuum but against a proven, successful competitor, making the strategic decision to discontinue its development a logical and financially prudent one.

Competitive Landscape: Tebotelimab vs. Nivolumab + Relatlimab (Opdualag)

The most direct competitor to Tebotelimab is Opdualag, a fixed-dose combination of the anti-PD-1 antibody nivolumab and the anti-LAG-3 antibody relatlimab. The success of Opdualag provides a stark contrast to the trajectory of Tebotelimab and effectively closed the most viable market opportunity for a dual PD-1/LAG-3 inhibitor.

In the pivotal Phase 2/3 RELATIVITY-047 trial, Opdualag was compared directly against nivolumab monotherapy in over 700 patients with previously untreated, advanced melanoma.[29] The results were practice-changing:

  • The combination nearly doubled the median progression-free survival, achieving 10.1 months compared to 4.6 months for nivolumab alone, representing a 25% reduction in the risk of disease progression or death.[29]
  • While Grade 3/4 treatment-related adverse events were more frequent with the combination (18.9% vs. 9.7%), the overall safety profile was considered manageable and notably less toxic than the approved combination of nivolumab plus ipilimumab (anti-CTLA-4).[29]

Based on these robust Phase 3 data, Opdualag received FDA approval in March 2022 for the treatment of advanced melanoma, becoming the first approved LAG-3-targeted therapy.[19] This successful outcome for the co-administration strategy contrasts sharply with Tebotelimab's terminated Phase 1 trial in melanoma (NCT04653038) and its complete lack of late-stage data.[13] The competitor's success validated the biological target but simultaneously rendered Tebotelimab's path to market in this key indication untenable.

Concluding Remarks and Outlook

Tebotelimab represents a pioneering but, to date, unsuccessful effort in the development of bispecific immune checkpoint inhibitors. Its journey from a molecule with a superior preclinical profile to one with a largely discontinued clinical program underscores the profound challenges of translating in vitro synergy into in vivo clinical benefit. The program has generated valuable data on the safety of dual PD-1/LAG-3 blockade and has provided intriguing, albeit isolated, signals of activity in highly refractory immune-exhausted disease states.

While Tebotelimab itself is unlikely to advance to market, the knowledge gained from its development is invaluable. It highlights the critical importance of rational combination strategies, as seen in the promising but undeveloped pairing with margetuximab. It also serves as a cautionary tale about the limitations of preclinical models and the formidable competitive and regulatory hurdles that novel platforms must overcome. The lessons learned from the Tebotelimab program will undoubtedly inform the design, strategy, and development of the next generation of multi-targeting immunotherapies as the field continues to seek more effective ways to harness the power of the immune system against cancer.

Works cited

  1. Definition of tebotelimab - NCI Drug Dictionary - NCI, accessed September 8, 2025, https://www.cancer.gov/publications/dictionaries/cancer-drug/def/tebotelimab
  2. Tebotelimab - MacroGenics - AdisInsight - Springer, accessed September 8, 2025, https://adisinsight.springer.com/drugs/800050138
  3. 313 A phase 1 evaluation of tebotelimab, a bispecific PD-1 x LAG-3 DART® molecule, in combination with margetuximab in patients with advanced HER2+ neoplasms | Journal for ImmunoTherapy of Cancer, accessed September 8, 2025, https://jitc.bmj.com/content/8/Suppl_3/A193.1
  4. Phase 2 Trial of Enoblituzumab Plus Retifanlimab or Tebotelimab in First-Line Treatment of Patients with Recurrent or Metastatic Head and Neck Squamous Cell Carcinoma - MacroGenics, Inc., accessed September 8, 2025, https://ir.macrogenics.com/static-files/3c84050f-03c6-4f71-bf49-d3f7837d7af0
  5. Tebotelimab, a PD-1/LAG-3 bispecific antibody, in patients with advanced hepatocellular carcinoma who had failed prior targeted therapy and/or immunotherapy: An open-label, single-arm, phase 1/2 dose-escalation and expansion study. | Journal of Clinical Oncology - ASCO Publications, accessed September 8, 2025, https://ascopubs.org/doi/10.1200/JCO.2023.41.4_suppl.578
  6. Tebotelimab: a bispecific PD-1xLAG-3 DART checkpoint inhibitor molecule... - ResearchGate, accessed September 8, 2025, https://www.researchgate.net/figure/Tebotelimab-a-bispecific-PD-1xLAG-3-DART-checkpoint-inhibitor-molecule-and-binding_fig1_374845429
  7. A Phase 1, Open-Label Study of MGD013 ... - MacroGenics, accessed September 8, 2025, https://macrogenics.com/wp-content/wbuploads/wb_publications/ASH%202020%20-%20Phase%201%20Study%20of%20MGD013%20(Tebotelimab,%20PD-1%20x%20LAG-3)%20in%20Patients%20with%20RR%20DLBCL%20(Wang)%20(1).pdf
  8. An open-label, single-arm, dose escalation and expansion phase 1 study of tebotelimab (MGD013) plus niraparib in patients with locally advanced or metastatic gastric cancer who failed prior treatments. - ResearchGate, accessed September 8, 2025, https://www.researchgate.net/publication/367767125_An_open-label_single-arm_dose_escalation_and_expansion_phase_1_study_of_tebotelimab_MGD013_plus_niraparib_in_patients_with_locally_advanced_or_metastatic_gastric_cancer_who_failed_prior_treatments
  9. Clinical landscape of LAG-3-targeted therapy - PMC - PubMed Central, accessed September 8, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC9216443/
  10. The PD-1- and LAG-3-targeting bispecific molecule tebotelimab in solid tumors and hematologic cancers: a phase 1 trial - PubMed Central, accessed September 8, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC10667103/
  11. (PDF) The PD-1- and LAG-3-targeting bispecific molecule tebotelimab in solid tumors and hematologic cancers: a phase 1 trial - ResearchGate, accessed September 8, 2025, https://www.researchgate.net/publication/374845429_The_PD-1-_and_LAG-3-targeting_bispecific_molecule_tebotelimab_in_solid_tumors_and_hematologic_cancers_a_phase_1_trial
  12. Tebotelimab Terminated Phase 1 Trials for Endometrial Carcinoma / Biliary Tract Carcinoma / Triple-Negative Breast Cancer / Gastric Cancer Treatment - DrugBank, accessed September 8, 2025, https://go.drugbank.com/drugs/DB16685/clinical_trials?conditions=DBCOND0037503%2CDBCOND0031833%2CDBCOND0028483%2CDBCOND0068542&phase=1&purpose=treatment&status=terminated
  13. Tebotelimab Terminated Phase 1 Trials for Unresectable, Recurrent or Metastatic Melanoma / Untreated Mucosal or Acral Lentiginous Melanoma Treatment - DrugBank, accessed September 8, 2025, https://go.drugbank.com/drugs/DB16685/clinical_trials?conditions=DBCOND0134950%2CDBCOND0134951&phase=1&purpose=treatment&status=terminated
  14. Tebotelimab Terminated Phase 2 Trials for Squamous Cell Carcinoma of the Head and Neck (SCCHN) / Head And Neck Cancer / Head and Neck Neoplasms Treatment - DrugBank, accessed September 8, 2025, https://go.drugbank.com/drugs/DB16685/clinical_trials?conditions=DBCOND0042625%2CDBCOND0029278%2CDBCOND0035874&phase=2&purpose=treatment&status=terminated
  15. PD-1 and LAG-3 dual blockade: emerging mechanisms and potential therapeutic prospects in cancer - PMC, accessed September 8, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC11667783/
  16. Tebotelimab - Drug Targets, Indications, Patents - Patsnap Synapse, accessed September 8, 2025, https://synapse.patsnap.com/drug/a5b79df1b2ec40aebd331c1aec5a27d2
  17. MacroGenics Presents Tebotelimab Data in Patients with Relapsed ..., accessed September 8, 2025, https://ir.macrogenics.com/news-releases/news-release-details/macrogenics-presents-tebotelimab-data-patients
  18. PD-1 and LAG-3 Dominate Checkpoint Receptor–Mediated T-cell Inhibition in Renal Cell Carcinoma - AACR Journals, accessed September 8, 2025, https://aacrjournals.org/cancerimmunolres/article/7/11/1891/469469/PD-1-and-LAG-3-Dominate-Checkpoint-Receptor
  19. PD-1 and LAG-3 dual blockade: emerging mechanisms and potential therapeutic prospects in cancer, accessed September 8, 2025, https://www.cancerbiomed.org/content/cbm/21/11/970.full.pdf
  20. The World of Immunotherapy Needs More Than PD-1/PD-L1—Two of the New Kids on the Block: LAG-3 and TIGIT - MDPI, accessed September 8, 2025, https://www.mdpi.com/2673-7523/4/3/10
  21. LAG3-PD-1 Combo Overcome the Disadvantage of Drug Resistance - Frontiers, accessed September 8, 2025, https://www.frontiersin.org/journals/oncology/articles/10.3389/fonc.2022.831407/full
  22. Phase 2 trial of enoblituzumab plus retifanlimab or tebotelimab in first-line treatment of patients (pts) with recurrent or metastatic squamous cell carcinoma of the head and neck (R/M SCCHN). | Journal of Clinical Oncology - ASCO Publications, accessed September 8, 2025, https://ascopubs.org/doi/10.1200/JCO.2022.40.16_suppl.TPS6102
  23. Bispecific Antibodies for the Treatment of Cancer - Athena Publishing, accessed September 8, 2025, https://www.athena-publishing.com/journals/mahig/articles/26/view
  24. Tebotelimab (MGD-013) | Anti-PD-1/LAG-3 DART Antibody | MedChemExpress, accessed September 8, 2025, https://www.medchemexpress.com/tebotelimab.html
  25. Tebotelimab (Anti-LAG-3 & PD-1) CD markers inhibitor | Read Reviews & Product Use Citations - Selleck Chemicals, accessed September 8, 2025, https://www.selleckchem.com/products/tebotelimab.html
  26. Tebotelimab | Abbexa Ltd, accessed September 8, 2025, https://www.abbexa.com/tebotelimab
  27. Tebotelimab - Research Grade Biosimilar - ProSci Incorporated, accessed September 8, 2025, https://www.prosci-inc.com/product/tebotelimab-research-grade-biosimilar-21-972/
  28. The PD-1- and LAG-3-targeting bispecific molecule tebotelimab in solid tumors and hematologic cancers - NET, accessed September 8, 2025, https://rcastoragev2.blob.core.windows.net/97d7566cc7819c0ced0fd4d9a405feea/41591_2023_2593_MOESM1_ESM.pdf
  29. Relatlimab plus nivolumab improves progression-free survival in metastatic melanoma, accessed September 8, 2025, https://www.mdanderson.org/newsroom/relatlimab-plus-nivolumab-improves-progression-free-survival-in-metastatic-melanoma.h00-159536589.html
  30. Opdualag (Nivolumab + Relatlimab) Combination Treatment for Melanoma - MRA, accessed September 8, 2025, https://www.curemelanoma.org/patient-eng/melanoma-treatment/options/opdualag-nivolumab-relatlimab
  31. A Phase 1, First-in-Human, Open-Label, Dose Escalation Study of MGD013, A Bispecific DART Protein Binding PD-1 and LAG-3 In Patients with Unresectable or Metastatic Neoplasms - MD Anderson Cancer Center, accessed September 8, 2025, https://www.mdanderson.org/patients-family/diagnosis-treatment/clinical-trials/clinical-trials-index/clinical-trials-detail.ID2017-0696.html
  32. Tebotelimab Completed Phase 1 Trials for Cervical Cancer / Advanced Solid Tumors / Neoplasms, Hematologic / Triple-Negative Breast Cancer / Squamous Cell Carcinoma of the Head and Neck (SCCHN) / Ovarian Cancer / HER-2 Positive Advanced Solid Tumors / Non-Small Cell Lung Cancer (NSCLC) / Small Cell Lung Cancer (SCLC) / Cholangiocarcinoma Treatment - DrugBank, accessed September 8, 2025, https://go.drugbank.com/drugs/DB16685/clinical_trials?conditions=DBCOND0030110%2CDBCOND0028450%2CDBCOND0012992%2CDBCOND0159523%2CDBCOND0056760%2CDBCOND0034130%2CDBCOND0028213%2CDBCOND0048944%2CDBCOND0035874%2CDBCOND0068542&phase=1&purpose=treatment&status=completed
  33. A Study of MGD013 in Patients With Unresectable or Metastatic Neoplasms - NCI, accessed September 8, 2025, https://www.cancer.gov/research/participate/clinical-trials-search/v?id=NCI-2018-00122
  34. PK/PD and safety profile of tebotelimab monotherapy a, Top 20 TRAEs vs... - ResearchGate, accessed September 8, 2025, https://www.researchgate.net/figure/PK-PD-and-safety-profile-of-tebotelimab-monotherapy-a-Top-20-TRAEs-vs-AEs-irrespective_fig2_374845429
  35. The PD-1- and LAG-3-targeting bispecific molecule ... - MacroGenics, accessed September 8, 2025, https://macrogenics.com/wp-content/uploads/2023/10/Luke-Nat-Med-2023_Tebotelimab.pdf
  36. The PD-1- and LAG-3-targeting bispecific molecule tebotelimab in solid tumors and hematologic cancers: a phase 1 trial - Johns Hopkins University, accessed September 8, 2025, https://pure.johnshopkins.edu/en/publications/the-pd-1-and-lag-3-targeting-bispecific-molecule-tebotelimab-in-s
  37. The PD-1- and LAG-3-targeting bispecific molecule tebotelimab in solid tumors and hematologic cancers: a phase 1 trial - PubMed, accessed September 8, 2025, https://pubmed.ncbi.nlm.nih.gov/37857711/
  38. Corporate Update - MacroGenics, Inc., accessed September 8, 2025, https://ir.macrogenics.com/static-files/18b424ad-6c43-4539-bc5a-9a5ac984e611
  39. An open-label, single-arm, dose escalation and expansion phase 1 study of tebotelimab (MGD013) plus niraparib in patients with locally advanced or metastatic gastric cancer who failed prior treatments. | Journal of Clinical Oncology - ASCO Publications, accessed September 8, 2025, https://ascopubs.org/doi/10.1200/JCO.2023.41.4_suppl.402
  40. Study Details | NCT04634825 | Enoblituzumab Plus Retifanlimab or Tebotelimab in Head and Neck Cancer | ClinicalTrials.gov, accessed September 8, 2025, https://clinicaltrials.gov/study/NCT04634825
  41. EU Clinical Trials Register, accessed September 8, 2025, https://www.clinicaltrialsregister.eu/ctr-search/trial/2020-004918-35/BG
  42. Tebotelimab Completed Phase 2 Trials for HER2-positive Gastric Cancer Treatment, accessed September 8, 2025, https://go.drugbank.com/drugs/DB16685/clinical_trials?conditions=DBCOND0092590&phase=2&purpose=treatment&status=completed
  43. MacroGenics Provides Update on Corporate Progress and 2021 Financial Results, accessed September 8, 2025, https://ir.macrogenics.com/news-releases/news-release-details/macrogenics-provides-update-corporate-progress-and-2021
  44. EU Clinical Trials Register, accessed September 8, 2025, https://www.clinicaltrialsregister.eu/ctr-search/trial/2020-004918-35/ES
  45. FDA Approval Timeline of Active Immunotherapies | CRI - Cancer Research Institute, accessed September 8, 2025, https://www.cancerresearch.org/regulatory-approval-timeline-of-active-immunotherapies
  46. OncLive's August EMA Regulatory Recap: Key EU Approvals in Oncology, accessed September 8, 2025, https://www.onclive.com/view/onclive-s-august-ema-regulatory-recap-key-eu-approvals-in-oncology
  47. Ebvallo | European Medicines Agency (EMA), accessed September 8, 2025, https://www.ema.europa.eu/en/medicines/human/EPAR/ebvallo
  48. BioInvent Receives FDA Orphan Drug Designation for BI-1808 for the Treatment of T-cell Lymphoma, accessed September 8, 2025, https://www.bioinvent.com/en/press/bioinvent-receives-fda-orphan-drug-designation-bi-1808-treatment-t-cell-lymphoma-2310616
  49. Alector Receives Orphan Drug Designation from the U.S. FDA for AL001 for the Treatment of Frontotemporal Dementia, accessed September 8, 2025, https://investors.alector.com/news-releases/news-release-details/alector-receives-orphan-drug-designation-us-fda-al001-treatment/
  50. Izalontamab Brengitecan (EGFRxHER3 ADC) Granted Breakthrough Therapy Designation by U.S. FDA for Patients with Previously Treated Advanced EGFR-Mutated Non-Small Cell Lung Cancer - Bristol Myers Squibb - Press Releases, accessed September 8, 2025, https://news.bms.com/news/corporate-financial/2025/Izalontamab-Brengitecan-EGFRxHER3-ADC-Granted-Breakthrough-Therapy-Designation-by-U-S--FDA-for-Patients-with-Previously-Treated-Advanced-EGFR-Mutated-Non-Small-Cell-Lung-Cancer/default.aspx
  51. Breakthrough Therapy - FDA, accessed September 8, 2025, https://www.fda.gov/patients/fast-track-breakthrough-therapy-accelerated-approval-priority-review/breakthrough-therapy
  52. NCT03470922 | A Study of Relatlimab Plus Nivolumab Versus Nivolumab Alone in Participants With Advanced Melanoma | ClinicalTrials.gov, accessed September 8, 2025, https://clinicaltrials.gov/study/NCT03470922
  53. Nivolumab and Relatlimab for Advanced Melanoma - National Cancer Institute, accessed September 8, 2025, https://www.cancer.gov/news-events/cancer-currents-blog/2021/melanoma-nivolumab-relatlimab-immunotherapy

Published at: September 8, 2025

This report is continuously updated as new research emerges.

MedPath

Empowering clinical research with data-driven insights and AI-powered tools.

© 2025 MedPath, Inc. All rights reserved.