MedPath

Garlic Advanced Drug Monograph

Published:Oct 14, 2025

Generic Name

Garlic

Drug Type

Biotech

CAS Number

8008-99-9

Vibostolimab (MK-7684): A Comprehensive Report on the Scientific Rationale, Clinical Development, and Discontinuation of an Anti-TIGIT Immunotherapy

1.0 Executive Summary

Vibostolimab, also known as MK-7684, was a humanized IgG1 monoclonal antibody developed by Merck & Co., Inc., as an investigational immunotherapy targeting the T-cell immunoreceptor with immunoglobulin and ITIM domains (TIGIT). Positioned as a next-generation immune checkpoint inhibitor, its development was underpinned by a strong preclinical rationale suggesting it could overcome tumor-induced immune suppression by blocking the inhibitory TIGIT pathway. A central component of Merck's strategy was the development of vibostolimab as a fixed-dose co-formulation with its cornerstone anti-PD-1 therapy, pembrolizumab (KEYTRUDA®), a combination intended to offer synergistic anti-tumor activity and administrative convenience.

Preclinical and structural analyses revealed that vibostolimab possessed potentially advantageous characteristics compared to other anti-TIGIT antibodies, such as tiragolumab. These included a larger binding footprint on the TIGIT protein, complete competition with the primary ligand CD155, and distinct binding kinetics that translated to superior T-cell activation in in vitro assays. Based on these promising early signals, Merck launched the ambitious KEYVIBE clinical development program, a comprehensive series of nine trials designed to evaluate vibostolimab-based therapies in over 4,000 patients across a wide spectrum of solid tumors and hematological malignancies, including non-small cell lung cancer (NSCLC), small cell lung cancer (SCLC), and melanoma.

Despite the robust scientific rationale and significant investment, the KEYVIBE program ultimately resulted in a series of high-profile clinical failures. Across multiple pivotal Phase 2 and Phase 3 trials, the combination of vibostolimab and pembrolizumab consistently failed to demonstrate a significant clinical benefit over established standards of care. The program was systematically dismantled between 2023 and 2024 due to a recurring pattern of negative outcomes. In some trials, such as the KEYVIBE-010 study in adjuvant melanoma, the combination led to an unacceptable increase in immune-mediated toxicities, resulting in a high rate of treatment discontinuation and an unfavorable risk-benefit profile. In other key studies, including the pivotal KEYVIBE-003 and KEYVIBE-007 trials in first-line metastatic NSCLC, the combination failed to meet primary endpoints for overall survival, leading to their termination for futility.

The comprehensive failure of the vibostolimab program serves as a significant and cautionary data point in the challenging development landscape for TIGIT-targeted therapies. It highlights a fundamental disconnect between promising in vitro functional data and in vivo clinical efficacy, and underscores the profound difficulty in achieving a therapeutic window where the benefits of dual checkpoint blockade outweigh the risks of cumulative toxicity. The discontinuation of vibostolimab, alongside similar setbacks for other anti-TIGIT agents, has tempered enthusiasm for this target and has shifted the scientific focus toward understanding the critical nuances of antibody engineering, such as Fc-receptor interactions, that may be required for this therapeutic modality to succeed.

2.0 Introduction to Vibostolimab and the TIGIT Immune Checkpoint

2.1 Drug Profile and Development History

Vibostolimab, known by its development code MK-7684, is a biologic therapeutic agent discovered and developed by Merck Sharp & Dohme, a subsidiary of Merck & Co., Inc..[1] It is classified as a humanized monoclonal antibody of the immunoglobulin G1 (IgG1) kappa isotype, produced in Chinese Hamster Ovary (CHO) cells.[3] As a new molecular entity, it is identified by the DrugBank accession number DB16420 and the Chemical Abstracts Service (CAS) Registry Number 2231305-30-7.[3] Its therapeutic classification encompasses several categories, including antineoplastics, immunotherapies, T lymphocyte stimulants, and, most specifically, TIGIT protein inhibitors.[1]

A defining feature of vibostolimab's clinical development strategy was its primary evaluation as a fixed-dose co-formulation with pembrolizumab (KEYTRUDA®), Merck's highly successful anti-programmed cell death protein-1 (PD-1) antibody. This co-formulated product, designated MK-7684A, contained fixed doses of both monoclonal antibodies and was designed to simplify intravenous administration and capitalize on the hypothesized synergistic effects of dual immune checkpoint blockade.[10] This approach represented a significant strategic commitment by Merck, aiming not only to develop a novel anti-TIGIT agent but also to create a proprietary, next-generation immuno-oncology product that could build upon and extend the clinical franchise of pembrolizumab. This strategy, however, relied on the unproven assumption that the combination would yield a superior therapeutic index—a greater increase in efficacy than in toxicity—an assumption that would ultimately be challenged by late-stage clinical data.

2.2 The TIGIT/CD226 Axis: A Critical Pathway in Tumor Immune Evasion

The scientific foundation for vibostolimab lies in the biology of TIGIT, a critical inhibitory immune checkpoint receptor. TIGIT is a member of the immunoglobulin superfamily and is expressed on the surface of key immune effector cells, most notably activated CD8+ cytotoxic T lymphocytes, CD4+ helper T cells, regulatory T cells (Tregs), and Natural Killer (NK) cells.[5] Its expression is often upregulated on tumor-infiltrating lymphocytes (TILs) within the tumor microenvironment (TME), particularly on "exhausted" T cells that also co-express other inhibitory receptors like PD-1.[10]

TIGIT exerts its immunosuppressive function through a competitive molecular interaction. It competes with a co-stimulatory receptor, CD226 (also known as DNAM-1), for binding to a shared set of ligands that are frequently overexpressed on the surface of tumor cells and antigen-presenting cells (APCs). These principal ligands are CD155 (also known as the poliovirus receptor, PVR) and CD112 (or PVRL2).[10] The TIGIT/CD226 axis thus functions as a molecular switch. When CD226 binds to CD155 or CD112, it delivers an activating signal that promotes T-cell and NK-cell function. Conversely, when TIGIT—which has a higher affinity for CD155 than CD226 does—binds to these same ligands, it delivers a potent inhibitory signal directly into the immune cell. This signal actively suppresses lymphocyte proliferation, cytokine production (such as interleukin-2 and interferon-gamma), and cytotoxic activity.[15] By upregulating CD155 and exploiting this pathway, tumors effectively hijack a natural regulatory mechanism to create an immunosuppressive TME, allowing them to evade detection and destruction by the immune system.[5]

2.3 Scientific Rationale for TIGIT Blockade in Oncology

The therapeutic strategy for anti-TIGIT antibodies like vibostolimab is based on the principle of "releasing the brakes" on the immune system. The central hypothesis is that an antagonistic antibody that binds to TIGIT can physically prevent it from interacting with its ligands, CD155 and CD112.[17] This blockade is intended to achieve two complementary goals. First, it prevents the transmission of direct inhibitory signals into T cells and NK cells. Second, by blocking TIGIT's access to its ligands, it allows the co-stimulatory receptor CD226 to bind more freely, thereby shifting the balance of signaling from inhibition toward activation.[15] The expected outcome is the restoration and enhancement of the anti-tumor functions of these critical immune cells, enabling them to more effectively recognize and eliminate cancer cells.[15]

Furthermore, a compelling rationale exists for combining TIGIT blockade with PD-1 blockade. The TIGIT and PD-1 pathways represent two distinct, non-redundant mechanisms of immune suppression that are often concurrently active within the TME.[10] TILs that are functionally exhausted frequently co-express both TIGIT and PD-1. Therefore, blocking only one of these pathways may be insufficient to fully reinvigorate an anti-tumor immune response. The combination of vibostolimab (anti-TIGIT) and pembrolizumab (anti-PD-1) was designed to simultaneously address multiple immune escape pathways. The dual blockade was theorized to produce a synergistic effect, leading to a more comprehensive and durable reactivation of the immune system than could be achieved with either agent alone, potentially overcoming resistance mechanisms and improving outcomes for patients.[10] This powerful biological hypothesis drove the extensive clinical investigation of the vibostolimab/pembrolizumab combination.

3.0 Pharmacological and Structural Characterization

3.1 Mechanism of Action: Reversing Immune Suppression

Vibostolimab is a humanized IgG1 monoclonal antibody engineered to bind with high specificity and affinity to the TIGIT receptor on human and cynomolgus monkey immune cells.[18] The primary pharmacological action of vibostolimab is to function as a competitive antagonist. By binding to TIGIT, it physically obstructs the receptor's interaction with its ligands, CD155 and CD112, which are commonly expressed on cancer cells.[2] This ligand blockade effectively disarms the TIGIT inhibitory pathway, thereby reactivating the cytotoxic functions of T cells and NK cells that have been suppressed within the tumor microenvironment.[15]

Beyond this direct mechanism of T-cell and NK-cell disinhibition, preclinical evidence points to a second, complementary mode of action. As an IgG1 isotype antibody, vibostolimab possesses an intact Fc region that can engage with Fc-gamma receptors (FcγR) expressed on the surface of myeloid cells, such as macrophages and dendritic cells (which function as APCs).[5] In vivo studies in mouse models demonstrated that treatment with a surrogate anti-TIGIT antibody led to the activation of myeloid cells within the TME. This was evidenced by the upregulation of key surface markers associated with enhanced antigen presentation, including Major Histocompatibility Complex (MHC) class II, CD86, and CD40.[18] This suggests a dual mechanism whereby vibostolimab not only directly relieves the suppression of cytotoxic lymphocytes but also indirectly enhances the initiation and amplification of the anti-tumor immune response by improving antigen presentation. This functional enhancement of APCs could, in theory, further potentiate the effects of T-cell-mediated therapies like PD-1 inhibitors.[18]

3.2 Molecular and Structural Analysis of Vibostolimab-TIGIT Binding

The interaction between vibostolimab and its target has been characterized at a high molecular resolution. Detailed structural analyses employing solution Nuclear Magnetic Resonance (NMR) and X-ray crystallography have precisely identified the binding epitope of vibostolimab on the TIGIT protein. Vibostolimab binds to a specific region known as the CC'C''FG loop interface, which is the same interface that TIGIT uses to bind its natural ligand, CD155.[18] The atomic-level details of this interaction have been further elucidated by the resolution of the co-crystal structure of the vibostolimab antigen-binding fragment (Fab) in complex with the TIGIT protein. This structure was determined by X-ray diffraction to a resolution of 2.29 Ångströms, providing a definitive molecular blueprint of how the antibody engages its target.[21] This structural information confirms that vibostolimab's mechanism of action is direct, steric hindrance of the TIGIT-CD155 interaction.

3.3 Comparative Analysis: Vibostolimab versus Tiragolumab

In the competitive landscape of anti-TIGIT antibody development, Genentech's tiragolumab emerged as a key comparator. Extensive biochemical and structural studies were conducted to differentiate vibostolimab from tiragolumab, revealing several key distinctions.[18]

Structural and Binding Differences:

Structural analyses demonstrated that while both antibodies target the ligand-binding domain of TIGIT, they do so with different physical properties. X-ray crystallography revealed that vibostolimab binds to the TIGIT surface with a significantly larger contact area, or "footprint," compared to tiragolumab. This larger coverage area allows vibostolimab to completely compete for the CD155 binding site.18 While both antibodies were shown to have a comparable overall binding affinity (KD​) to human TIGIT, their binding kinetics were markedly different. Surface plasmon resonance (Biacore) analysis indicated that vibostolimab is characterized by statistically significant faster association rates (ka​ or "on-rates") and faster dissociation rates (kd​ or "off-rates").18 This kinetic profile suggests a more dynamic interaction with the TIGIT receptor compared to the slower, more prolonged binding of tiragolumab.

In Vitro Functional Differences:

These structural and kinetic differences appeared to translate into a functional advantage in in vitro assays designed to measure T-cell activation. In an engineered Jurkat T-cell co-culture system, where the production of Interleukin-2 (IL-2) serves as a surrogate for T-cell activation, vibostolimab demonstrated a significantly greater maximal effect (Emax​). It induced higher levels of IL-2 production compared to tiragolumab, a difference that was attributed to its distinct binding properties.18 This finding formed a key part of the preclinical argument for vibostolimab's potential superiority.

Preclinical In Vivo Equivalence:

A critical observation, however, emerged from subsequent in vivo studies. Despite the demonstrable in vitro advantages in binding and T-cell activation, these differences failed to translate into superior anti-tumor efficacy in mouse tumor models. When mouse surrogate antibodies designed to mimic the properties of vibostolimab and tiragolumab were tested, no significant difference in tumor shrinkage was observed between the two.18 Both antibodies did, however, confirm the dual mechanism of action by inducing the activation of myeloid cells in addition to enhancing T-cell cytolytic activity.18 This disconnect between the in vitro functional assay (IL-2 production) and the ultimate therapeutic goal (in vivo tumor control) represented a crucial early indicator of translational difficulty. It suggested that the greater T-cell activation observed in the simplified lab setting might not be the decisive factor for efficacy within the complex, multifactorial environment of a living tumor, a finding that foreshadowed the challenges the program would later face in clinical trials.

Table 1: Comparative Profile of Vibostolimab and Tiragolumab

ParameterVibostolimabTiragolumab
Binding EpitopeCC'C''FG loop interface of TIGIT 18CC'C''FG loop interface of TIGIT 18
Surface Coverage on TIGITLarger surface contact area 18Smaller surface contact area 18
Competition with CD155Complete competition 18Partial/Overlapping competition 18
Binding Affinity ()Comparable 18Comparable 18
Binding Kinetics (On/Off Rates)Faster on-rate () and off-rate () 18Slower on-rate () and off-rate () 18
In Vitro IL-2 Production ()Significantly greater 18Lower 18

4.0 The KEYVIBE Clinical Development Program

4.1 Program Strategy and Scope

Building on the preclinical rationale and early clinical signals, Merck & Co. launched the KEYVIBE program, a large-scale, comprehensive clinical development initiative designed to rigorously evaluate the safety and efficacy of vibostolimab.[10] This ambitious program ultimately planned to enroll over 4,000 patients, reflecting a significant investment and a strong belief in the potential of TIGIT blockade.[2] The central strategic thrust of the program was the investigation of vibostolimab in combination with the anti-PD-1 antibody pembrolizumab, most often as the fixed-dose co-formulation MK-7684A.[10] The overarching goal was to establish this dual checkpoint blockade as a new and improved standard of care, leveraging the established efficacy of KEYTRUDA® as a foundation.

The scope of the KEYVIBE program was exceptionally broad, encompassing a wide array of oncologic indications and treatment settings. Clinical trials were initiated in multiple solid tumor types, including non-small cell lung cancer (NSCLC), small cell lung cancer (SCLC), melanoma, gastric cancer, metastatic urothelial carcinoma, and renal cell carcinoma (RCC).[10] The program also extended into hematological malignancies.[23] The trials were designed to assess vibostolimab-based therapies across the entire patient journey, from the neoadjuvant setting for resectable disease to first-line and later-line treatment for advanced and metastatic cancer.[13]

4.2 Overview of the Nine Constituent Clinical Trials

The KEYVIBE program was formally composed of nine distinct clinical trials, spanning all phases of clinical development: one Phase I study, three Phase II studies, and five pivotal Phase III studies.[10] Each trial was designed to answer specific questions about the utility of vibostolimab in different patient populations and therapeutic combinations. The table below provides a consolidated overview of the entire program, outlining the design and ultimate status of each study. This summary illustrates the breadth of the investigation and foreshadows the widespread challenges that led to the program's eventual termination.

Table 2: Summary of the KEYVIBE Clinical Trial Program

Trial IDNCT NumberPhasePrimary Indication(s)Key Intervention(s)Final Status
KEYVIBE-001NCT02964013IAdvanced Solid TumorsVibostolimab mono; Vibo + PemboCompleted 10
KEYVIBE-002NCT04725188IIMetastatic NSCLC (previously treated)Vibo/Pembo + Docetaxel; Vibo/Pembo monoCompleted 23
KEYVIBE-003NCT04738487IIIMetastatic NSCLC (1L, PD-L1+)Vibo/Pembo vs. Pembrolizumab monoDiscontinued (Futility) 23
KEYVIBE-004NCT05005442IIRelapsed/Refractory Hematological MalignanciesVibo/Pembo co-formulationActive, not recruiting 10
KEYVIBE-005NCT05007106IIAdvanced Solid Tumors (Basket Trial)Vibostolimab-based therapiesActive, not recruiting 10
KEYVIBE-006NCT05298423IIIStage III NSCLCVibo/Pembo + CRT vs. Durvalumab + CRTDiscontinued 23
KEYVIBE-007NCT05226598IIIMetastatic NSCLC (1L)Vibo/Pembo + Chemo vs. Pembo + ChemoDiscontinued (Futility) 23
KEYVIBE-008NCT05224141IIIExtensive-Stage SCLC (1L)Vibo/Pembo + Chemo vs. Atezolizumab + ChemoDiscontinued (Futility) 23
KEYVIBE-010NCT05665595IIIResected High-Risk Melanoma (Adjuvant)Vibo/Pembo vs. Pembrolizumab monoDiscontinued (Unfavorable Risk/Benefit) 20
Vibo = Vibostolimab; Pembo = Pembrolizumab; Chemo = Chemotherapy; CRT = Chemoradiotherapy; 1L = First-line

5.0 Analysis of Key Clinical Trials: Efficacy and Safety

5.1 Phase 1 Foundation: The KEYVIBE-001 Study (NCT02964013)

The KEYVIBE-001 trial served as the foundation for the entire vibostolimab development program. It was a first-in-human, multi-part, open-label study designed to assess the safety, tolerability, pharmacokinetics (PK), and preliminary anti-tumor activity of vibostolimab, both as a monotherapy and in combination with pembrolizumab, in patients with advanced solid tumors.[14] The dose-escalation portion of the study (Part A) enrolled 76 patients and reported no dose-limiting toxicities, establishing a manageable safety profile.[35] Treatment-related adverse events (TRAEs) occurred in 56% of patients on monotherapy and 62% on combination therapy, with Grade 3-4 TRAEs being relatively infrequent (9% and 17%, respectively).[5]

In terms of efficacy, the early signals were modest but considered encouraging enough to proceed. Across all tumor types in the dose-escalation phase, the confirmed objective response rate (ORR) was 0% for monotherapy and 7% for the combination.[5] A more promising signal emerged from a specific expansion cohort of patients with metastatic NSCLC who had not previously received anti-PD-1/PD-L1 therapy. In this population of 41 patients, the combination of vibostolimab and pembrolizumab demonstrated an ORR of 29% (95% CI, 16-46) and a median progression-free survival (PFS) of 5.4 months.[36] Conversely, in patients whose cancer was refractory to prior PD-1/PD-L1 blockade, the combination showed minimal activity, with an ORR of only 5%.[36] It was the promising activity in the anti-PD-1/L1-naive NSCLC cohort that provided the primary justification for launching the broader and more ambitious later-phase KEYVIBE program.[10]

5.2 Phase 2 Assessment in NSCLC: The KEYVIBE-002 Study (NCT04725188)

The KEYVIBE-002 trial was a critical test of the vibostolimab/pembrolizumab combination in a challenging patient population: metastatic NSCLC that had already progressed following standard-of-care platinum-based chemotherapy and at least one prior anti-PD-1/L1 immunotherapy.[2] This randomized, partially-blind, three-arm Phase 2 study compared (1) the vibostolimab/pembrolizumab co-formulation plus docetaxel chemotherapy, (2) the co-formulation alone, and (3) placebo plus docetaxel (the active comparator arm).[2]

The trial ultimately failed to meet its dual primary endpoints of improving PFS.[2]

  • Progression-Free Survival (PFS): The triplet combination of vibostolimab/pembrolizumab plus docetaxel resulted in a median PFS of 5.6 months, compared to 3.2 months for docetaxel alone. While this represented a numerical improvement, the result did not achieve statistical significance (Hazard Ratio = 0.77; 95% CI, 0.53-1.13; ).[2] More concerningly, the vibostolimab/pembrolizumab doublet performed worse than the standard of care, with a median PFS of only 2.7 months (HR=1.40 vs. docetaxel).[2]
  • Overall Survival (OS): The secondary endpoint of OS showed a similar pattern. The triplet combination demonstrated a non-statistically significant trend toward improved survival (median OS 10.2 months vs. 8.8 months for docetaxel; HR=0.76; 95% CI, 0.50-1.15).[2] The doublet again showed no benefit (median OS 7.5 months; HR=1.05 vs. docetaxel).[2]
  • Objective Response Rate (ORR): The ORR was highest in the triplet arm at 29.9%, compared to 15.3% for docetaxel alone and a very low 6.0% for the doublet arm.[2]

The safety profile was consistent with the known toxicities of the individual agents, with no new safety signals identified.[2] However, the addition of the co-formulation to chemotherapy substantially increased the burden of adverse events. The rate of any-grade TRAEs was 96.5% in the triplet arm, compared to 89.2% in the docetaxel arm and 60.2% in the doublet arm.[2] The most common severe (Grade 3-5) TRAEs in the triplet arm were hematologic, including neutropenia (16.5%) and anemia (7.1%).[2] The results of KEYVIBE-002 were a major setback, demonstrating that in this heavily pre-treated population, the addition of vibostolimab and pembrolizumab to chemotherapy added toxicity without providing a statistically significant efficacy benefit.

Table 3: Efficacy and Safety Outcomes of the KEYVIBE-002 Trial

EndpointVibo/Pembo + Docetaxel (n=87)Vibo/Pembo Alone (n=83)Docetaxel Alone (n=85)
Median PFS (months)5.62.73.2
HR vs. Docetaxel (95% CI)0.77 (0.53-1.13)1.40 (0.96-2.02)-
p-value0.09100.9622-
Median OS (months)10.27.58.8
HR vs. Docetaxel (95% CI)0.76 (0.50-1.15)1.05 (0.70-1.58)-
ORR (%)29.96.015.3
Any-Grade TRAEs (%)96.560.289.2
Grade 3-5 TRAEs (%)Not specified, but common events listedNot specified, but common events listedNot specified, but common events listed
Most Common Grade 3-5 TRAEsNeutropenia (16.5%), Anemia (7.1%)Asthenia (2.4%), Diarrhea (2.4%)Neutropenia (14.5%), Anemia (6.0%)
Data sourced from 2

5.3 Phase 3 Disappointments in Melanoma and SCLC

The negative signals from Phase 2 were amplified by subsequent failures in large Phase 3 trials in other indications.

The KEYVIBE-010 trial (NCT05665595) evaluated the vibostolimab/pembrolizumab co-formulation as an adjuvant (post-surgery) treatment for patients with resected high-risk melanoma.[20] This trial was unblinded and effectively discontinued not because of a direct finding of futility, but based on the recommendation of an independent Data Monitoring Committee (DMC) due to an unfavorable risk-benefit balance. The DMC observed a significantly higher rate of treatment discontinuation in the combination arm compared to the pembrolizumab-only arm, driven primarily by immune-mediated adverse experiences. This high dropout rate made it "highly unlikely" that the trial could ever demonstrate a statistically significant improvement in its primary endpoint of recurrence-free survival (RFS).[20] This outcome highlighted a critical problem: the additive toxicity of the dual blockade was too high for the adjuvant setting, where patients are clinically free of disease.

The KEYVIBE-008 trial (NCT05224141) investigated the vibostolimab/pembrolizumab co-formulation plus chemotherapy as a first-line treatment for extensive-stage small cell lung cancer (ES-SCLC), comparing it against the standard of care, atezolizumab plus chemotherapy.[32] This study was also stopped early after a pre-planned analysis determined that it had met pre-specified futility criteria for its primary endpoint of OS. Furthermore, the analysis revealed that patients in the vibostolimab arm experienced a higher rate of both general and immune-related adverse events compared to the control arm, confirming a negative therapeutic index.[33]

5.4 The Final Blow: Futility in First-Line NSCLC (KEYVIBE-003 & -007)

The definitive end of the vibostolimab program came with the failure of its two largest and most commercially important pivotal trials in first-line metastatic NSCLC. The KEYVIBE-003 trial (NCT04738487) was a head-to-head comparison of the vibostolimab/pembrolizumab co-formulation versus pembrolizumab monotherapy in patients whose tumors expressed PD-L1.[29] The KEYVIBE-007 trial (NCT05226598) evaluated the co-formulation plus chemotherapy against pembrolizumab plus chemotherapy in a broader NSCLC population.[29]

In a conclusive announcement, Merck stated that it was discontinuing both of these Phase 3 trials based on the recommendation of a DMC.[29] In separate pre-planned analyses, both trials met their pre-specified criteria for futility for the primary endpoint of OS.[29] This meant that there was no evidence that adding vibostolimab to the pembrolizumab backbone was providing any survival benefit. This result, in the core target indication for which the combination was developed, was unequivocal. The consistent pattern of failure—ranging from excessive toxicity in melanoma, to a poor therapeutic index in pre-treated NSCLC, to a complete lack of efficacy in first-line SCLC and NSCLC—demonstrated a fundamental flaw in the therapeutic hypothesis of this specific dual checkpoint combination, leading Merck to terminate the entire clinical development program.[29]

6.0 Pharmacokinetics and Pharmacodynamics

6.1 Pharmacokinetic (PK) Profile

The characterization of vibostolimab's pharmacokinetic profile was a key secondary objective in its early-phase clinical trials, particularly the foundational KEYVIBE-001 (NCT02964013) and KEYVIBE-004 (NCT05005442) studies.[14] The dose-escalation phase of KEYVIBE-001 was designed to determine the recommended Phase 2 dose (RPTD) for vibostolimab. This was established as 200 mg administered as an intravenous infusion every 3 weeks (Q3W).[10] This dosing schedule was strategically chosen to align perfectly with the approved 200 mg Q3W dosing for pembrolizumab, a critical prerequisite for the development and administration of the fixed-dose co-formulation, MK-7684A.[29]

The KEYVIBE-001 study protocol included intensive PK sampling during the initial treatment cycles (pre-dose, end of infusion, and multiple post-dose timepoints) to characterize the drug's absorption, distribution, and elimination after a single dose and to assess its accumulation over multiple doses to reach a steady state.[35] Available data from the study confirmed that, as expected, serum exposures of vibostolimab increased with escalating doses from 2.1 mg up to 700 mg Q3W.[35] However, detailed public reporting on specific PK parameters such as clearance rate, volume of distribution, elimination half-life, and potential drug-drug interactions with pembrolizumab is limited. The primary focus of published results has been on the clinical safety and efficacy outcomes.

6.2 Pharmacodynamic (PD) and Target Engagement

The pharmacodynamic effects of vibostolimab are the direct biological consequences of its binding to and blocking of the TIGIT receptor. The intended PD effect is the functional restoration of anti-tumor immunity, driven by the reactivation of T-cells and NK cells through the prevention of TIGIT-mediated inhibitory signaling and the concurrent promotion of the co-stimulatory CD226 pathway.[15] Preclinical studies identified increased IL-2 production by T-cells as a potential biomarker of this pharmacodynamic activity.[18]

While clinical trial protocols included exploratory objectives to evaluate pharmacodynamic markers, such as receptor occupancy on peripheral immune cells and immune correlates of response within blood and tumor tissue, specific data from these assessments are not widely available in the provided materials.[36] The most telling pharmacodynamic readout from the clinical program is the overall balance of efficacy and toxicity. The repeated observation of increased immune-mediated adverse events (imAEs) in the vibostolimab combination arms across multiple trials (e.g., KEYVIBE-010 in melanoma and KEYVIBE-008 in SCLC) is, in itself, a powerful pharmacodynamic signal.[20] It indicates that the dual blockade of TIGIT and PD-1 was indeed biologically active and capable of augmenting immune responses. However, this augmented immune activity manifested primarily as heightened auto-reactivity and toxicity without a corresponding, or sufficient, increase in anti-tumor efficacy. This demonstrates a potent but ultimately unfavorable clinical pharmacodynamic effect, where the combination failed to achieve a viable therapeutic window.

7.0 Discontinuation of Development and Regulatory Status

7.1 A Cascade of Futility: The Rationale for Termination

The termination of the vibostolimab clinical development program was not the result of a single trial failure but rather a cumulative and consistent pattern of negative or unfavorable results that emerged across its late-stage portfolio throughout 2023 and 2024. This cascade of disappointments systematically dismantled the clinical and commercial case for the drug.

The key events leading to the program's discontinuation were:

  • KEYVIBE-010 (Melanoma): The trial in the adjuvant melanoma setting was the first major Phase 3 setback. It was discontinued due to an unfavorable risk-benefit profile, where the combination arm experienced a significantly higher rate of discontinuation due to immune-mediated adverse events, rendering the primary endpoint unattainable.[20]
  • KEYVIBE-008 (SCLC): The next failure occurred in first-line small cell lung cancer, where the trial was stopped early after a planned analysis showed it met pre-specified criteria for futility on the primary endpoint of overall survival. The combination arm was also associated with a higher rate of adverse events.[33]
  • KEYVIBE-003 & KEYVIBE-007 (NSCLC): The final and most decisive blow came from the two pivotal trials in first-line metastatic non-small cell lung cancer. Both studies were halted based on a DMC recommendation after interim analyses concluded they had met futility criteria for overall survival.[29]

Following these conclusive failures in its most important target indications, Merck announced the discontinuation of the entire vibostolimab clinical development program, including the ongoing KEYVIBE-006 trial, citing the "totality of the data" from the Phase 3 studies as the basis for its decision.[29]

7.2 Regulatory History with the FDA and EMA

Vibostolimab's journey with regulatory agencies reflects its investigational nature and eventual discontinuation. It never received marketing approval from any major regulatory body.

  • U.S. Food and Drug Administration (FDA): On November 8, 2022, the FDA granted Orphan Drug Designation to the combination of vibostolimab and pembrolizumab for the treatment of Small Cell Lung Cancer (SCLC).[42] This designation is intended to encourage the development of drugs for rare diseases. However, following the failure of the KEYVIBE-008 trial in this indication, the designation was officially withdrawn or revoked on January 24, 2025.[42] Vibostolimab is not, and has never been, an FDA-approved drug.[42]
  • European Medicines Agency (EMA): The EMA's Paediatric Committee (PDCO) agreed to a Paediatric Investigation Plan (PIP) for the vibostolimab/pembrolizumab combination for the treatment of Hodgkin lymphoma on May 15, 2023 (Plan number: EMEA-003063-PIP02-22).[45] A PIP is a mandatory development plan for studying a medicine's use in children. In line with the termination of the overall program, a subsequent notification was published confirming the discontinuation of this paediatric development plan.[45] Vibostolimab is not approved for use in the European Union.[46]

8.0 Concluding Analysis and Future Perspective

8.1 Vibostolimab in the Context of the Broader Anti-TIGIT Field

The failure of vibostolimab was not an isolated event but rather a reflection of broader, class-wide challenges facing TIGIT-targeted immunotherapies. The initial excitement for TIGIT as the "next PD-1" has been significantly tempered by a series of high-profile clinical trial failures involving multiple drug candidates from different pharmaceutical companies.

  • Tiragolumab (Genentech/Roche): As the most advanced competitor, tiragolumab's trajectory has closely mirrored that of vibostolimab. Despite showing early promise in the Phase 2 CITYSCAPE trial, its extensive "SKYSCRAPER" Phase 3 program has yielded a string of negative results, failing to show a survival benefit in NSCLC and SCLC. These repeated failures led Roche to ultimately remove tiragolumab from its late-stage pipeline, a major blow to the field.[50]
  • Ociperlimab (BeiGene): Another prominent candidate, ociperlimab, also faced a definitive setback. Its pivotal Phase 3 AdvanTIG-302 trial in lung cancer was terminated for futility after an interim analysis showed it was unlikely to meet its primary endpoint of overall survival. This led BeiGene to discontinue the entire clinical program for the drug.[53]
  • Domvanalimab (Arcus Biosciences/Gilead Sciences): In stark contrast to the failures of vibostolimab, tiragolumab, and ociperlimab, the anti-TIGIT antibody domvanalimab has maintained a positive development trajectory. It has produced encouraging Phase 2 data in studies such as EDGE-Gastric and ARC-10, showing improvements in survival endpoints in gastroesophageal and lung cancers.[57] A key differentiating feature of domvanalimab is its "Fc-silent" design. Vibostolimab and tiragolumab are both standard IgG1 antibodies with active Fc regions, which can lead to the depletion of TIGIT-expressing T cells (including beneficial effector T cells) via antibody-dependent cell-mediated cytotoxicity (ADCC). Domvanalimab's engineered Fc region is designed to prevent this, potentially preserving the pool of T cells that the therapy is intended to activate. This structural difference has emerged as the leading hypothesis to explain the divergent clinical outcomes within the anti-TIGIT class.

Table 4: Comparative Overview of Key Anti-TIGIT Antibodies

Antibody NameDeveloper(s)Fc DesignKey Indication(s) StudiedCurrent Development Status / Key Outcomes
VibostolimabMerck & Co.IgG1 (Fc-active)NSCLC, SCLC, MelanomaDiscontinued. Failed multiple Phase 3 trials due to futility and/or unfavorable risk-benefit.20
TiragolumabGenentech/RocheIgG1 (Fc-active)NSCLC, SCLC, Esophageal CancerDiscontinued. Failed multiple Phase 3 trials (SKYSCRAPER series) for lack of efficacy.50
OciperlimabBeiGeneIgG1 (Fc-active)NSCLCDiscontinued. Phase 3 trial (AdvanTIG-302) terminated for futility.53
DomvanalimabArcus Biosciences / Gilead SciencesFc-silentNSCLC, Gastroesophageal CancerIn active Phase 3 development. Has shown positive Phase 2 data; Fc-silent design is a key differentiator.57

8.2 Insights Gleaned from the Failure of the KEYVIBE Program

The comprehensive collapse of the KEYVIBE program offers several critical lessons for the field of immuno-oncology. The most significant is the clear demonstration of the perils of additive toxicity. The program's results consistently showed that combining two potent immunomodulatory agents, vibostolimab and pembrolizumab, did not produce a reliable synergistic efficacy but carried a high risk of synergistic or cumulative toxicity. The combination repeatedly failed to establish a favorable therapeutic window where the clinical benefit clearly outweighed the harm from immune-mediated adverse events.

Secondly, the program serves as a stark example of translational failure. The promising preclinical data for vibostolimab, including its superior ability to induce IL-2 production in in vitro assays compared to tiragolumab, proved to be a poor and misleading predictor of clinical success. This highlights the profound limitations of simplified laboratory models in capturing the complex interplay of immune cells, tumor cells, and stromal factors within the human tumor microenvironment.

Finally, the widespread failures of Fc-active IgG1 anti-TIGIT antibodies like vibostolimab and tiragolumab have cast serious doubt on the viability of this specific molecular approach. It raises fundamental questions about whether TIGIT is a sufficiently potent target to yield a clinical benefit when combined with PD-1 blockade, at least when using an antibody format that may inadvertently deplete the very cells it aims to activate.

8.3 The Future of TIGIT as a Therapeutic Target in Oncology

While the failures of vibostolimab and other first-generation anti-TIGIT antibodies have significantly dampened the initial widespread enthusiasm, the biological rationale for targeting the TIGIT pathway remains intact. The field has not been abandoned but has been forced to pivot toward more nuanced and differentiated strategies. The future of TIGIT as a therapeutic target now hinges on answering several key questions that have emerged from these setbacks.

The most immediate focus is on the role of antibody engineering. The relative success of the Fc-silent domvanalimab suggests that minimizing Fc-receptor engagement and avoiding ADCC-mediated depletion of TIGIT-positive effector T cells may be critical for therapeutic success. Ongoing and future trials with domvanalimab and other Fc-engineered antibodies will be closely watched to validate this hypothesis. Beyond antibody format, research will likely explore whether TIGIT blockade is more effective in specific, biomarker-defined patient populations or in tumor types outside of the major indications where it has already failed. Furthermore, the exploration of novel combination partners beyond PD-1 inhibitors may unlock the potential of TIGIT blockade. The failure of vibostolimab, while a costly and disappointing outcome for Merck, provides an invaluable dataset for the scientific community, guiding the next wave of research toward a more sophisticated and potentially successful approach to targeting this challenging but still compelling immune checkpoint.

9.0 References

[1]

Works cited

  1. Vibostolimab - Merck Sharp & Dohme - AdisInsight - Springer, accessed October 14, 2025, https://adisinsight.springer.com/drugs/800048295
  2. Merck Announces Findings from Phase 2 KeyVibe-002 Trial Evaluating an Investigational Coformulation of Vibostolimab and Pembrolizumab in Previously Treated Patients with Metastatic Non-Small Cell Lung Cancer (NSCLC), accessed October 14, 2025, https://www.merck.com/news/merck-announces-findings-from-phase-2-keyvibe-002-trial-evaluating-an-investigational-coformulation-of-vibostolimab-and-pembrolizumab-in-previously-treated-patients-with-metastatic-non-small-cell-lung/
  3. Vibostolimab (anti-TIGIT) Datasheet - Selleck Chemicals, accessed October 14, 2025, https://www.selleckchem.com/datasheet/vibostolimab-anti-tigit-A2033-DataSheet.html
  4. Vibostolimab (anti-TIGIT) - Selleck Chemicals, accessed October 14, 2025, https://www.selleckchem.com/products/vibostolimab-anti-tigit.html
  5. First-in-human phase 1 study of the anti–TIGIT antibody ..., accessed October 14, 2025, https://www.researchgate.net/publication/356349852_First-in-human_phase_1_study_of_the_anti-TIGIT_antibody_vibostolimab_as_monotherapy_or_with_pembrolizumab_for_advanced_solid_tumors_including_non-small_cell_lung_cancer
  6. Vibostolimab | MedPath, accessed October 14, 2025, https://trial.medpath.com/drug/f059844dafa3b9a7
  7. Vibostolimab - 5 mg - Tebubio, accessed October 14, 2025, https://www.tebubio.com/en_fr_eur/vibostolimab-494t35391-5-mg.html
  8. Vibostolimab | CAS 2231305-30-7 - AbMole BioScience, accessed October 14, 2025, https://www.abmole.com/products/vibostolimab.html
  9. Vibostolimab, CAS [[2231305-30-7]] | BIOZOL, accessed October 14, 2025, https://www.biozol.de/en/product/tgm-t35391
  10. The KEYVIBE program: vibostolimab and pembrolizumab for the ..., accessed October 14, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC11497960/
  11. Definition of vibostolimab/pembrolizumab MK-7684A - NCI Drug Dictionary, accessed October 14, 2025, https://www.cancer.gov/publications/dictionaries/cancer-drug/def/vibostolimab-pembrolizumab-mk-7684a
  12. Study Details | NCT04725188 | Pembrolizumab/Vibostolimab Coformulation (MK-7684A) or Pembrolizumab/Vibostolimab Coformulation Plus Docetaxel Versus Docetaxel for Metastatic Non Small Cell Lung Cancer (NSCLC) With Progressive Disease After Platinum Doublet Chemotherapy and Immunotherapy (MK-7684A-002, KEYVIBE-002) | ClinicalTrials.gov, accessed October 14, 2025, https://www.clinicaltrials.gov/study/NCT04725188
  13. The KEYVIBE program: vibostolimab and pembrolizumab for the treatment of advanced malignancies - Taylor & Francis Online, accessed October 14, 2025, https://www.tandfonline.com/doi/abs/10.1080/14796694.2024.2343272
  14. NCT02964013 | Study of Vibostolimab Alone and in Combination With Pembrolizumab in Advanced Solid Tumors (MK-7684-001) ( KEYVIBE-001) | ClinicalTrials.gov, accessed October 14, 2025, https://clinicaltrials.gov/study/NCT02964013
  15. Vibostolimab: Mechanism, Clinical Applications, and Research Potential in Oncology, accessed October 14, 2025, https://www.assaygenie.com/blog/vibostolimab-mechanism-clinical-applications-and-research-potential-in-oncology
  16. Safety and Efficacy of Vibostolimab and Pembrolizumab in Patients with Relapsed or Refractory Hematologic Malignancies: A Multicohort, Open-Label, Phase 2 Study, accessed October 14, 2025, https://ashpublications.org/blood/article/138/Supplement%201/2484/478848/Safety-and-Efficacy-of-Vibostolimab-and
  17. Definition of vibostolimab - NCI Drug Dictionary, accessed October 14, 2025, https://www.cancer.gov/publications/dictionaries/cancer-drug/def/vibostolimab
  18. Pharmacological and structural characterization of vibostolimab, a ..., accessed October 14, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC12366625/
  19. Pharmacological and structural characterization of vibostolimab, a novel anti-TIGIT blocking antibody for cancer immunotherapy - PubMed, accessed October 14, 2025, https://pubmed.ncbi.nlm.nih.gov/40829901/
  20. Merck Provides Update on Phase 3 KeyVibe-010 Trial Evaluating an Investigational Coformulation of Vibostolimab and Pembrolizumab as Adjuvant Treatment for Patients With Resected High-Risk Melanoma, accessed October 14, 2025, https://www.merck.com/news/merck-provides-update-on-phase-3-keyvibe-010-trial-evaluating-an-investigational-coformulation-of-vibostolimab-and-pembrolizumab-as-adjuvant-treatment-for-patients-with-resected-high-risk-melanoma/
  21. 8VTE: Co-structure of the Fab of the anti-TIGIT Vibostolimab antibody with its antigen - RCSB PDB, accessed October 14, 2025, https://www.rcsb.org/structure/8vte
  22. Induction of immune activation by vibostolimab in in vitro stimulation.... - ResearchGate, accessed October 14, 2025, https://www.researchgate.net/figure/nduction-of-immune-activation-by-vibostolimab-in-in-vitro-stimulation-Human_fig4_394675378
  23. The KEYVIBE program: vibostolimab and pembrolizumab for the treatment of advanced malignancies - PubMed, accessed October 14, 2025, https://pubmed.ncbi.nlm.nih.gov/39230120/
  24. Clinical Trials Using Vibostolimab - NCI, accessed October 14, 2025, https://www.cancer.gov/research/participate/clinical-trials/intervention/vibostolimab
  25. RCC (Renal Cell Carinoma) Active Not Recruiting Phase 1 Trials for Vibostolimab (DB16420) - DrugBank, accessed October 14, 2025, https://go.drugbank.com/indications/DBCOND0103675/clinical_trials/DB16420?phase=1&status=active_not_recruiting
  26. Metastatic Urothelial Carcinoma Active Not Recruiting Phase 1 Trials for Vibostolimab (DB16420) | DrugBank Online, accessed October 14, 2025, https://go.drugbank.com/indications/DBCOND0073854/clinical_trials/DB16420?phase=1&status=active_not_recruiting
  27. Vibostolimab - Drug Targets, Indications, Patents - Patsnap Synapse, accessed October 14, 2025, https://synapse.patsnap.com/drug/e0642281fe4749fb838e4f7eade3f11a
  28. Vibostolimab Completed Phase 2 Trials for Metastatic Non-Small Cell Lung Cancer Treatment - DrugBank, accessed October 14, 2025, https://go.drugbank.com/drugs/DB16420/clinical_trials?conditions=DBCOND0043377&phase=2&purpose=treatment&status=completed
  29. Merck Provides Update on KeyVibe and KEYFORM Clinical Development Programs Evaluating Investigational Vibostolimab and Favezelimab Fixed-Dose Combinations with Pembrolizumab, accessed October 14, 2025, https://www.merck.com/news/merck-provides-update-on-keyvibe-and-keyform-clinical-development-programs-evaluating-investigational-vibostolimab-and-favezelimab-fixed-dose-combinations-with-pembrolizumab/
  30. NCT05005442 | A Study of Pembrolizumab/Vibostolimab (MK-7684A) in Relapsed/Refractory Hematological Malignancies (MK-7684A-004, KEYVIBE-004) | ClinicalTrials.gov, accessed October 14, 2025, https://clinicaltrials.gov/study/NCT05005442
  31. NCT05226598 | Study of Pembrolizumab/Vibostolimab Coformulation (MK-7684A) in Combination With Chemotherapy Versus Pembrolizumab Plus Chemotherapy in Participants With Metastatic Non-Small Cell Lung Cancer (MK-7684A-007/KEYVIBE-007) | ClinicalTrials.gov, accessed October 14, 2025, https://clinicaltrials.gov/study/NCT05226598
  32. Study Details | NCT05224141 | Pembrolizumab/Vibostolimab (MK-7684A) or Atezolizumab in Combination With Chemotherapy in First Line Treatment of Extensive-Stage Small Cell Lung Cancer (MK-7684A-008/KEYVIBE-008) - Clinical Trials, accessed October 14, 2025, https://clinicaltrials.gov/study/NCT05224141
  33. Merck Provides Update on Phase 3 KeyVibe-008 Trial Evaluating an Investigational Fixed-Dose Combination of Vibostolimab and Pembrolizumab in Patients With Extensive-Stage Small Cell Lung Cancer, accessed October 14, 2025, https://www.merck.com/news/merck-provides-update-on-phase-3-keyvibe-008-trial-evaluating-an-investigational-fixed-dose-combination-of-vibostolimab-and-pembrolizumab-in-patients-with-extensive-stage-small-cell-lung-cancer/
  34. Merck Axes Two Cancer Candidates After Disappointing Late-Stage Results - BioSpace, accessed October 14, 2025, https://www.biospace.com/drug-development/merck-axes-two-cancer-candidates-after-disappointing-late-stage-results
  35. First-in-human phase 1 study of the anti-TIGIT antibody vibostolimab ..., accessed October 14, 2025, https://pubmed.ncbi.nlm.nih.gov/34800678/
  36. Merck Presents Promising New Data for Three Investigational Medicines From Diverse and Expansive Oncology Pipeline at ESMO Virtual Congress 2020, accessed October 14, 2025, https://www.merck.com/news/merck-presents-promising-new-data-for-three-investigational-medicines-from-diverse-and-expansive-oncology-pipeline-at-esmo-virtual-congress-2020/
  37. The KEYVIBE program: vibostolimab and pembrolizumab for the treatment of advanced malignancies - ResearchGate, accessed October 14, 2025, https://www.researchgate.net/publication/383754909_The_KEYVIBE_program_vibostolimab_and_pembrolizumab_for_the_treatment_of_advanced_malignancies
  38. The KEYVIBE program: vibostolimab and pembrolizumab for the treatment of advanced malignancies - Taylor & Francis Group - Figshare, accessed October 14, 2025, https://tandf.figshare.com/articles/journal_contribution/The_KEYVIBE_program_vibostolimab_and_pembrolizumab_for_the_treatment_of_advanced_malignancies/26936392
  39. Vibostolimab/Pembrolizumab Fails to Reach Statistical Significance in NSCLC | Targeted Oncology - Immunotherapy, Biomarkers, and Cancer Pathways, accessed October 14, 2025, https://www.targetedonc.com/view/vibostolimab-pembrolizumab-fails-to-reach-statistical-significance-in-nsclc
  40. Vibostolimab Plus Pembrolizumab Coformulation ± Docetaxel Did ..., accessed October 14, 2025, https://www.onclive.com/view/vibostolimab-plus-pembrolizumab-coformulation-docetaxel-did-not-significantly-improve-pfs-in-nsclc
  41. Late-Stage Vibostolimab, Pembrolizumab Plus Chemotherapy Trial Discontinued Due to Adverse Events | AJMC, accessed October 14, 2025, https://www.ajmc.com/view/late-stage-trial-discontinued-due-to-adverse-events
  42. Search Orphan Drug Designations and Approvals - accessdata.fda ..., accessed October 14, 2025, https://www.accessdata.fda.gov/scripts/opdlisting/oopd/detailedIndex.cfm?cfgridkey=893722
  43. FDA approves pembrolizumab and berahyaluronidase alfa-pmph for subcutaneous injection, accessed October 14, 2025, https://www.fda.gov/drugs/resources-information-approved-drugs/fda-approves-pembrolizumab-and-berahyaluronidase-alfa-pmph-subcutaneous-injection
  44. Oncology (Cancer)/Hematologic Malignancies Approval Notifications - FDA, accessed October 14, 2025, https://www.fda.gov/drugs/resources-information-approved-drugs/oncology-cancerhematologic-malignancies-approval-notifications
  45. EMEA-003063-PIP02-22 - paediatric investigation plan | European ..., accessed October 14, 2025, https://www.ema.europa.eu/en/medicines/human/paediatric-investigation-plans/emea-003063-pip02-22
  46. OncLive's September EMA Regulatory Recap: Key EU Approvals in Oncology, accessed October 14, 2025, https://www.onclive.com/view/onclive-s-september-ema-regulatory-recap-key-eu-approvals-in-oncology
  47. YAbS database - The Antibody Society, accessed October 14, 2025, https://db.antibodysociety.org/db0/results_cmpny?page=11&sort=-cmpny_addrss
  48. Pembrolizumab receives positive EMA CHMP opinion for extended approval in patients with R/R cHL - Lymphoma Hub, accessed October 14, 2025, https://lymphomahub.com/medical-information/pembrolizumab-receives-positive-ema-chmp-opinion-for-extended-approval-in-patients-with-rr-chl
  49. Keytruda - opinion on variation to marketing authorisation - European Medicines Agency, accessed October 14, 2025, https://www.ema.europa.eu/en/medicines/human/variation/keytruda-1
  50. 5000 patients later Roche scraps its TIGIT | ApexOnco - Oncology Pipeline, accessed October 14, 2025, https://www.oncologypipeline.com/apexonco/5000-patients-later-roche-scraps-its-tigit
  51. Tiragolumab (Anti-TIGIT) in SCLC: Skyscraper-02, a Towering Inferno - PMC, accessed October 14, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC9831070/
  52. Tiragolumab Plus Atezolizumab Fails to Meet OS End Point in PD-L1–High NSCLC, accessed October 14, 2025, https://www.onclive.com/view/tiragolumab-plus-atezolizumab-fails-to-meet-os-end-point-in-pd-l1-high-nsclc
  53. Developers Terminate Clinical Program for Ociperlimab in Lung Cancer - CancerNetwork, accessed October 14, 2025, https://www.cancernetwork.com/view/developers-terminate-clinical-program-for-ociperlimab-in-lung-cancer
  54. Phase III Lung Cancer Trial Terminated Due to Futility - EMJ, accessed October 14, 2025, https://www.emjreviews.com/oncology/news/phase-iii-lung-cancer-trial-terminated-due-to-futility/
  55. BeiGene abandons ociperlimab over poor phase 3 prospects in latest blow to TIGITs, accessed October 14, 2025, https://www.fiercebiotech.com/biotech/beigene-abandons-ociperlimab-over-poor-phase-3-prospects-latest-blow-tigits
  56. BeiGene Provides Update on the Ociperlimab (BGB-A1217) Clinical Development Program, accessed October 14, 2025, https://ir.beonemedicines.com/news/beigene-provides-update-on-the-ociperlimab-bgb-a1217-clinical-development-program/4f7a71c1-cb84-4fa6-b562-65c239f04bc6
  57. Anti-TIGIT Domvanalimab Plus Anti-PD-1 Zimberelimab and Chemotherapy Showed 26.7 Months of Median Overall Survival as First-Line Treatment of Unresectable or Advanced Gastroesophageal Adenocarcinomas in the Phase 2 EDGE-Gastric Study - Investors & Media, accessed October 14, 2025, https://investors.arcusbio.com/investors-and-media/press-releases/press-release-details/2025/Anti-TIGIT-Domvanalimab-Plus-Anti-PD-1-Zimberelimab-and-Chemotherapy-Showed-26-7-Months-of-Median-Overall-Survival-as-First-Line-Treatment-of-Unresectable-or-Advanced-Gastroesophageal-Adenocarcinomas-in-the-Phase-2-EDGE-Gastric-Study/default.aspx
  58. Gilead and Arcus Announce Anti-TIGIT Domvanalimab Continues to Demonstrate Consistent Improvement in Progression-Free Survival in Non-Small Cell Lung Cancer Study, accessed October 14, 2025, https://www.gilead.com/news/news-details/2023/gilead-and-arcus-announce-anti-tigit-domvanalimab-continues-to-demonstrate-consistent-improvement-in-progression-free-survival-in-non-small-cell-lung-cancer-study
  59. Domvanalimab Combo Improves Survival in Advanced/Metastatic NSCLC - CancerNetwork, accessed October 14, 2025, https://www.cancernetwork.com/view/domvanalimab-combo-improves-survival-in-advanced-metastatic-nsclc
  60. Gilead, Arcus keep TIGIT dream alive with 27-month OS data - Fierce Biotech, accessed October 14, 2025, https://www.fiercebiotech.com/biotech/gilead-arcus-keep-tigit-dream-alive-27-month-os-phase-2-cancer-cohort
  61. Metastasis Withdrawn Phase 1 Trials for Vibostolimab (DB16420) | DrugBank Online, accessed October 14, 2025, https://go.drugbank.com/indications/DBCOND0017341/clinical_trials/DB16420?phase=1&status=withdrawn
  62. Merck stops anti-TIGIT combo in melanoma trial - Patsnap Synapse, accessed October 14, 2025, https://synapse.patsnap.com/article/merck-stops-anti-tigit-combo-in-melanoma-trial
  63. Basic and Translational Cancer Immunology Archives, accessed October 14, 2025, https://jitc.bmj.com/pages/collection/basic-and-translational-cancer-immunology
  64. Tiragolumab and TIGIT: pioneering the next era of cancer immunotherapy - Frontiers, accessed October 14, 2025, https://www.frontiersin.org/journals/pharmacology/articles/10.3389/fphar.2025.1568664/full
  65. (PDF) 1410P Safety and efficacy of vibostolimab, an anti-TIGIT antibody, plus pembrolizumab in patients with anti-PD-1/PD-L1-naive NSCLC - ResearchGate, accessed October 14, 2025, https://www.researchgate.net/publication/345368293_1410P_Safety_and_efficacy_of_vibostolimab_an_anti-TIGIT_antibody_plus_pembrolizumab_in_patients_with_anti-PD-1PD-L1-naive_NSCLC
  66. Vibostolimab: Uses, Interactions, Mechanism of Action | DrugBank Online, accessed October 14, 2025, https://go.drugbank.com/drugs/DB16420
  67. Study Details | NCT05224141 | Pembrolizumab/Vibostolimab (MK-7684A) or Atezolizumab in Combination With Chemotherapy in First Line Treatment of Extensive-Stage Small Cell Lung Cancer (MK-7684A-008/KEYVIBE-008) | ClinicalTrials.gov, accessed October 14, 2025, https://www.clinicaltrials.gov/study/NCT05224141
  68. EU Clinical Trials Register, accessed October 14, 2025, https://www.clinicaltrialsregister.eu/ctr-search/trial/2021-004564-94/AT
  69. List of anti-TIGIT molecules in clinical development. - ResearchGate, accessed October 14, 2025, https://www.researchgate.net/figure/List-of-anti-TIGIT-molecules-in-clinical-development_tbl2_354740589
  70. List of anti-TIGIT antibodies currently in clinical trials, according... - ResearchGate, accessed October 14, 2025, https://www.researchgate.net/figure/List-of-anti-TIGIT-antibodies-currently-in-clinical-trials-according-to-the-official_tbl1_373420269
  71. Next Generation Anti TIGIT Antibodies In Cancer Immunotherapy ..., accessed October 14, 2025, https://www.biospace.com/press-releases/next-generation-anti-tigit-antibodies-in-cancer-immunotherapy
  72. Study of Vibostolimab Alone and in Combination With Pembrolizumab in Advanced Solid Tumors (MK-7684-001) ( KEYVIBE-001) - larvol clin, accessed October 14, 2025, https://clin.larvol.com/trial-detail/NCT02964013
  73. NCT04746924 | A Study of Ociperlimab With Tislelizumab Compared to Pembrolizumab in Participants With Untreated Lung Cancer | ClinicalTrials.gov, accessed October 14, 2025, https://www.clinicaltrials.gov/study/NCT04746924
  74. AdvanTIG-105: a phase I dose escalation study of the anti-TIGIT monoclonal antibody ociperlimab in combination with tislelizumab in patients with advanced solid tumors - PMC, accessed October 14, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC10603446/
  75. Clinical Trials Using Domvanalimab - NCI, accessed October 14, 2025, https://www.cancer.gov/research/participate/clinical-trials/intervention/domvanalimab?pn=1
  76. Study Details | NCT05502237 | Zimberelimab and Domvanalimab in Combination With Chemotherapy Versus Pembrolizumab With Chemotherapy in Patients With Untreated Metastatic Non-Small Cell Lung Cancer | ClinicalTrials.gov, accessed October 14, 2025, https://clinicaltrials.gov/study/NCT05502237
  77. Phase I study of the anti-TIGIT antibody tiragolumab in combination with atezolizumab in Japanese patients with advanced or metastatic solid tumors - PubMed Central, accessed October 14, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC11258096/
  78. SKYSCRAPER-02: Tiragolumab in Combination With Atezolizumab Plus Chemotherapy in Untreated Extensive-Stage Small-Cell Lung Cancer | Journal of Clinical Oncology - ASCO Publications, accessed October 14, 2025, https://ascopubs.org/doi/10.1200/JCO.23.01363
  79. Clinical Trials Using Tiragolumab - NCI, accessed October 14, 2025, https://www.cancer.gov/research/participate/clinical-trials/intervention/tiragolumab?pn=1

Published at: October 14, 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.