MedPath

IBI-321 Advanced Drug Monograph

Published:Oct 9, 2025

Generic Name

IBI-321

Utomilumab (PF-05082566): A Clinical and Scientific Monograph on a Second-Generation 4-1BB Agonist

Introduction: Utomilumab - A Profile of a Second-Generation 4-1BB Agonist

Executive Summary

Utomilumab, also known by its development code PF-05082566, is an investigational, fully human IgG2 monoclonal antibody developed by Pfizer as a novel cancer immunotherapy.[1] Its development represents a pivotal chapter in the complex and challenging history of therapeutic agents targeting the 4-1BB (CD137) costimulatory receptor. This class of drugs is founded on the compelling hypothesis that potent stimulation of 4-1BB on the surface of T cells and natural killer (NK) cells can unleash a powerful and durable anti-tumor immune response.[4] The narrative of Utomilumab is fundamentally one of rational drug design aimed at navigating the treacherous therapeutic window of this pathway—a window defined by the delicate balance between achieving robust immune activation and maintaining an acceptable safety profile for patients.

The development of Utomilumab was a direct and strategic response to the significant clinical setbacks of the first-generation 4-1BB agonist, urelumab. While urelumab demonstrated tantalizing signs of clinical efficacy, its development was severely hampered by dose-limiting and sometimes fatal hepatotoxicity, establishing a clear and urgent need for a safer alternative.[6] Utomilumab was engineered specifically to meet this need. Through a distinct molecular design centered on its IgG2 isotype, it successfully achieved an excellent safety profile, consistently proving to be well-tolerated in clinical trials without the liver toxicity that plagued its predecessor. However, this triumph in safety came at a significant cost. The clinical program for Utomilumab was ultimately discontinued not due to safety concerns, but because of a consistent pattern of insufficient efficacy across both monotherapy and most combination settings.[4] The story of Utomilumab thus serves as a critical case study, offering invaluable lessons on the intricate relationship between antibody structure, mechanism of action, and the clinical balance of risk and benefit in the field of immuno-oncology.

The design and subsequent clinical evaluation of Utomilumab can be understood as a conscious strategic pivot in immuno-oncology development, moving away from a philosophy of "potency at all costs" toward one of "safety-first engineering." The failure of urelumab created a well-defined problem for the field: how to harness the therapeutic potential of 4-1BB without inducing unacceptable toxicity. Pfizer's answer was Utomilumab, a molecule deliberately engineered for a more tempered, conditional agonism. The choice of an IgG2 isotype, which requires secondary cross-linking by Fc gamma receptors (FcγRs) to achieve full agonistic potential, was a key feature of this design.[1] The underlying hypothesis was that this would restrict potent immune activation to the tumor microenvironment, where both target T cells and FcγR-bearing myeloid cells are co-located, thereby sparing the liver and other organs from systemic, off-target effects. Clinical data consistently validated the safety aspect of this hypothesis; trials repeatedly reported that Utomilumab was well-tolerated, with a notable absence of the severe hepatotoxicity associated with the target class.[10] However, this safety came with a trade-off. The efficacy data across these same trials were consistently described as "modest" or "limited," with low objective response rates (ORR) that failed to meet the high bar for advancement in modern oncology.[4] The eventual discontinuation of the program due to this lack of efficacy completed the narrative arc: the attempt to solve the safety problem inadvertently created an efficacy problem. This outcome illustrates a fundamental challenge in modulating the 4-1BB pathway and has profoundly influenced the design of the next generation of agonists that seek to find the elusive "Goldilocks zone" of activity.

Target and Therapeutic Hypothesis

The molecular target of Utomilumab is 4-1BB, also known as CD137 or Tumor Necrosis Factor Receptor Superfamily Member 9 (TNFRSF9).[1] 4-1BB is an inducible costimulatory receptor that is not typically present on resting immune cells but is rapidly expressed on T cells (both CD4+ and CD8+) and NK cells following their activation.[1] This inducible expression makes it an ideal target for cancer immunotherapy, as an agonist drug should theoretically focus its activity at sites of ongoing immune responses, such as within the tumor microenvironment.

The core therapeutic hypothesis behind Utomilumab and other 4-1BB agonists is that providing a potent, targeted costimulatory signal through this receptor can amplify and sustain an anti-tumor immune attack. By binding to and activating 4-1BB, an agonist antibody is expected to promote the proliferation, survival, and cytotoxic function of tumor-specific T cells and NK cells. This mechanism is believed to be particularly effective when combined with other immunotherapies, such as immune checkpoint inhibitors (e.g., anti-PD-1/PD-L1). In this combination paradigm, the checkpoint inhibitor "releases the brakes" on the immune system by blocking inhibitory signals, while the 4-1BB agonist "presses the accelerator," providing a powerful positive signal to drive the immune response forward, potentially leading to synergistic and more durable clinical outcomes.[3]

Molecular and Structural Characteristics

Drug Identification

Utomilumab is a biologic therapeutic agent formally identified through a series of internationally recognized naming conventions and database identifiers. Its proposed non-proprietary name, assigned under the United States Adopted Names (USAN) and International Nonproprietary Name (INN) systems, is Utomilumab.[17] During its development by Pfizer, it was primarily referred to by the development code PF-05082566, with variations such as PF-2566 also appearing in the literature.[1] For comprehensive data archiving and retrieval, it is cataloged in the DrugBank database under the accession ID DB15113.[1] Its unique chemical identity is registered with the Chemical Abstracts Service (CAS) under the number 1417318-27-4.[2]

Structural Profile

Utomilumab is a fully human monoclonal antibody of the Immunoglobulin G2 (IgG2) isotype, with a lambda light chain.[1] The designation "fully human" indicates that its protein sequences are derived entirely from human immunoglobulin genes, a characteristic intended to minimize immunogenicity and the formation of anti-drug antibodies (ADAs) in patients. It is produced using recombinant DNA technology in a mammalian cell expression system, specifically Chinese Hamster Ovary (CHO) cells, which is a standard for the manufacturing of complex therapeutic proteins.[2] The complete antibody has an approximate molecular weight of 142.78 kDa.[20] Analysis of its constituent chains shows a light chain (LC) of approximately 24 kDa and a heavy chain (HC) of approximately 51 kDa.[19]

The specific antigen-binding properties of Utomilumab are determined by the amino acid sequences of its variable domains. The sequences, sourced from the Thera-SAbDab database, are as follows [23]:

  • Heavy Chain Variable Region (): EVQLVQSGAEVKKPGESLRISCKGSGYSFSTYWISWVRQMPGKGLEWMGKIYPGDSYTNYSPSFQGQVTISADKSISTAYLQWSSLKASDTAMYYCARGYGIFDYWGQGTLVTVSS
  • Light Chain Variable Region (): SYELTQPPSVSVSPGQTASITCSGDNIGDQYAHWYQQKPGQSPVLVIYQDKNRPSGIPERFSGSNSGNTATLTISGTQAMDEADYYCATYTGFGSLAVFGGGTKLTVL

The development of this specific antibody was facilitated by advanced protein engineering techniques, notably the MorphoSys Human Combinatorial Antibody Library (HuCAL) phage display technology, which allows for the rapid generation and selection of fully human antibodies against a target of interest.[23]

Table 1: Utomilumab - Key Drug Identifiers and Properties

FeatureDetailSource(s)
Non-proprietary NameUtomilumab17
Development CodePF-050825661
DrugBank IDDB151131
CAS Number1417318-27-42
Drug TypeBiotech / Monoclonal Antibody
IsotypeFully human IgG2 lambda1
SourceChinese Hamster Ovary (CHO) Cells2
Molecular WeightApprox. 143 kDa20
Target4-1BB / CD137 / TNFRSF91

The 4-1BB (CD137) Target: A Cornerstone of T-Cell Costimulation

Biological Context

The therapeutic efficacy of Utomilumab is entirely dependent on its interaction with its molecular target, 4-1BB (also known as CD137 and TNFRSF9). 4-1BB is a transmembrane glycoprotein and a member of the tumor necrosis factor (TNF) receptor superfamily, a group of proteins critical for regulating inflammation, immunity, and apoptosis.[1] A key feature of 4-1BB is that its expression is tightly regulated and inducible. In a resting state, most immune cells do not express 4-1BB on their surface. However, upon activation—typically following T-cell receptor (TCR) engagement with an antigen—its expression is rapidly upregulated on the surface of crucial immune effector cells. These include cytotoxic CD8+ T cells, helper and memory CD4+ T cells, and NK cells.[1] This transient, activation-dependent expression pattern makes 4-1BB an exceptionally attractive target for immunotherapy. It offers the potential for therapeutic intervention to be spatially and temporally focused on sites of active immune responses, such as the tumor microenvironment or draining lymph nodes, thereby minimizing the risk of widespread, non-specific immune activation and associated toxicities.[25]

Signaling and Function

In normal physiology, 4-1BB is activated by its cognate ligand, 4-1BBL (TNFSF9), which is primarily expressed on the surface of professional antigen-presenting cells (APCs) like dendritic cells, macrophages, and B cells.[24] The binding of 4-1BBL to 4-1BB on a T cell provides a critical "signal 2" of costimulation, which acts in concert with the primary TCR signal ("signal 1") to orchestrate a robust immune response. This engagement triggers the recruitment of intracellular adaptor proteins, notably TNF receptor-associated factors (TRAFs), which in turn initiate downstream signaling cascades. The two primary pathways activated are the nuclear factor-kappa B (NF-κB) and mitogen-activated protein kinase (MAPK) pathways.[1]

The activation of these pathways results in a suite of cellular responses that are profoundly beneficial for generating an effective anti-tumor immune attack:

  • Enhanced Proliferation and Survival: 4-1BB signaling strongly promotes the clonal expansion of antigen-specific T cells. It also delivers potent pro-survival signals by upregulating anti-apoptotic proteins like Bcl-xL and Bfl-1, which protects activated T cells from activation-induced cell death and allows for a sustained immune response.[1]
  • Increased Effector Function: The costimulatory signal from 4-1BB significantly boosts the functional capacity of immune cells. It enhances the cytolytic activity of CD8+ T cells and NK cells, making them more efficient at killing target tumor cells.[1]
  • Cytokine Production: 4-1BB activation stimulates the production and secretion of key pro-inflammatory cytokines, most notably interferon-gamma (IFN-γ). IFN-γ plays a multifaceted role in anti-tumor immunity, including the direct inhibition of tumor cell growth, upregulation of MHC molecules on cancer cells to make them more visible to T cells, and the activation of other immune cells like macrophages.[1]
  • Memory Formation: A critical function of 4-1BB signaling is its role in the development and long-term maintenance of memory T-cell populations. This is essential for establishing durable immunity that can prevent tumor recurrence.[4]

The expression of 4-1BB on both effector and regulatory immune cells, however, presents a significant therapeutic challenge. While the primary goal of an agonist like Utomilumab is to stimulate the anti-tumor functions of effector T cells (Teffs), the receptor is also known to be expressed on immunosuppressive regulatory T cells (Tregs).[1] This creates a biological paradox: a non-selective 4-1BB agonist has the potential to amplify both the desired anti-tumor Teff populations and the undesirable pro-tumor Treg populations. The net clinical outcome of such an intervention would depend on the delicate balance between these opposing forces within the patient and the specific tumor microenvironment. This inherent complexity helps to explain the often-inconsistent efficacy observed with 4-1BB agonists and provides a strong scientific rationale for clinical strategies that combine 4-1BB agonism with agents designed to specifically deplete or inhibit Tregs. The clinical trial of Utomilumab in combination with mogamulizumab, an anti-CCR4 antibody that targets and depletes a subset of Tregs, was a direct attempt to test this hypothesis—that by removing the immunosuppressive "brake" applied by Tregs, the "accelerator" of 4-1BB agonism could function more effectively.[15] The failure of this combination to demonstrate meaningful clinical benefit suggests that this paradox is either not the primary barrier to efficacy for a mild agonist like Utomilumab, or that the strategy employed was insufficient to overcome it, further highlighting the deep complexities of modulating this powerful immune pathway.

Mechanism of Action: A Differentiated Approach to 4-1BB Agonism

Receptor Binding and Ligand Competition

Utomilumab is a fully human IgG2 agonist monoclonal antibody engineered to bind with high affinity and specificity to the extracellular domain of human 4-1BB.[1] Its binding site on the 4-1BB receptor is a critical feature that distinguishes it from the first-generation agonist, urelumab. Structural and competitive binding studies have shown that Utomilumab binds to the second and third cysteine-rich domains (CRD2 and CRD3) of 4-1BB.[7] This region of the receptor overlaps with the binding site for its natural ligand, 4-1BBL. As a result, Utomilumab acts as a ligand-blocking antibody, meaning it physically obstructs and competes with the endogenous 4-1BBL for access to the receptor.[1] This contrasts with urelumab, which binds to the membrane-proximal CRD1 domain and does not compete with 4-1BBL.[7] The ligand-blocking nature of Utomilumab may also have the secondary effect of preventing "reverse signaling" through 4-1BBL on the surface of APCs, though the clinical relevance of this is less clear.[1]

The Critical Role of the IgG2 Isotype and Cross-Linking

The most defining characteristic of Utomilumab's mechanism of action, and the cornerstone of its "safety-first" design, is its human IgG2 isotype. Human immunoglobulin isotypes (IgG1, IgG2, IgG3, IgG4) possess distinct structural features, particularly in their hinge regions and Fc domains, which dictate their flexibility and their ability to interact with Fc gamma receptors (FcγRs) on other immune cells. The IgG2 isotype is characterized by a relatively rigid hinge structure and exhibits weak binding to most activating FcγRs.[7]

This structural property has a profound functional consequence: Utomilumab is a relatively weak agonist on its own and requires secondary cross-linking to achieve potent signaling.[1] For Utomilumab to effectively cluster multiple 4-1BB receptors on the surface of a T cell—a necessary step for initiating a strong intracellular signal—the Fc portions of the antibody molecules must simultaneously bind to FcγRs (particularly the inhibitory receptor FcγRIIb) expressed on an adjacent cell, such as a macrophage, dendritic cell, or B cell.[7] This mechanistic requirement was a deliberate engineering choice. The therapeutic hypothesis was that this dependency on cross-linking would spatially restrict potent 4-1BB agonism to tissues where there is a high density of both 4-1BB-expressing T cells and FcγR-expressing cells, such as the tumor microenvironment and secondary lymphoid organs. This was expected to prevent widespread, systemic T-cell activation, which was believed to be the cause of the severe hepatotoxicity observed with the more potent, cross-linking-independent agonist, urelumab. The combination of endogenous ligand blocking and the requirement for in situ cross-linking was therefore designed to deliver a targeted agonist signal with a significantly improved safety margin.[1]

Downstream Cellular Effects

Once potent agonism is achieved through receptor binding and cross-linking, Utomilumab activates the canonical intracellular signaling pathways associated with 4-1BB. This includes the recruitment of TRAF proteins and the subsequent activation of the NF-κB and MAPK signaling cascades.[1] The ultimate biological outcomes are the enhancement of T-cell and NK-cell mediated anti-tumor immunity, driven by increased cellular proliferation, enhanced survival and resistance to apoptosis, and augmented effector functions, including the production of pro-inflammatory cytokines like IFN-γ.[1]

Preclinical Evidence and Therapeutic Rationale

In Vitro Studies

The foundational biological activity of Utomilumab was established through a series of in vitro experiments. In cell-based assays using both engineered reporter cell lines and primary human lymphocytes, Utomilumab demonstrated its ability to bind to 4-1BB and trigger downstream signaling, as measured by the activation of the NF-κB transcription factor and the subsequent production of cytokines.[1] These studies confirmed its intrinsic agonistic properties. Furthermore, competitive binding assays validated its mechanism as a ligand-blocking antibody, showing that it could effectively prevent the interaction between 4-1BB and its natural ligand, 4-1BBL.[1]

In Vivo Animal Models

The therapeutic potential of Utomilumab was further evaluated in preclinical animal models. A particularly relevant model for testing human-specific antibodies is the human peripheral blood lymphocyte (PBL)-reconstituted severe combined immunodeficiency (SCID) mouse model. In these xenograft models, where mice are engrafted with a functional human immune system and human tumors, treatment with single-agent Utomilumab resulted in significant anti-tumor activity, evidenced by a marked inhibition of tumor growth.[1] These in vivo studies also confirmed that Utomilumab could stimulate the proliferation of human leukocytes within the host, providing direct evidence of its immune-stimulatory effects and validating the therapeutic concept before moving into human trials.[1]

Combination Rationale

A substantial body of preclinical work provided a strong scientific rationale for evaluating Utomilumab in combination with other anti-cancer therapies, particularly immune checkpoint inhibitors. In various murine tumor models, the combination of a 4-1BB agonist with an antibody blocking the PD-1/PD-L1 pathway demonstrated additive or even synergistic anti-tumor effects.[3] The mechanistic hypothesis for this synergy is complementary action: checkpoint inhibitors work by "releasing the brakes" on T cells that have become exhausted or suppressed within the tumor microenvironment, while 4-1BB agonists work by "pressing the accelerator," providing a potent costimulatory signal to reinvigorate these and other T cells. This dual-pronged approach was predicted to generate a more robust, comprehensive, and durable anti-tumor immune response than could be achieved with either agent alone, forming the basis for several key clinical trials in the Utomilumab development program.[4]

Clinical Development Program: A Comprehensive Review

The clinical development of Utomilumab was extensive, exploring its potential as a monotherapy and in a wide array of combination regimens across both hematologic malignancies and solid tumors. The program was designed to first establish safety and then to identify a context in which its modest agonistic activity could translate into meaningful clinical benefit. The following table provides a high-level summary of the major clinical trials that defined its trajectory.

Table 2: Summary of Major Clinical Trials for Utomilumab

Trial ID (NCT)PhaseIndication(s)Combination Agent(s)Patient NKey Efficacy OutcomeKey Safety OutcomeSource(s)
NCT01307267IAdvanced Solid Tumors / B-cell LymphomasMonotherapy55ORR: 3.8% (solid tumors)Well-tolerated, no DLTs10
NCT01307267ICD20+ Non-Hodgkin's Lymphoma (NHL)Rituximab67ORR: 21.2%Favorable safety profile, no MTD reached32
NCT02179918IbAdvanced Solid TumorsPembrolizumab (anti-PD-1)23ORR: 26.1% (6 responses)Well-tolerated, no DLTs12
NCT02444793IbAdvanced Solid TumorsMogamulizumab (anti-CCR4)24ORR: 4.2% (1 PR)Safe and tolerable, no DLTs28
NCT03217747I/IIAdvanced GI MalignanciesAvelumab (anti-PD-L1) + Ivuxolimab (anti-OX40)31 (all arms)Modest activity (no ORR, 37.5% irSD)Safe, no Grade 4/5 AEs35
NCT02554812I/IIAdvanced Gynecologic MalignanciesAvelumab ± Ivuxolimab/Radiotherapy40ORR: 2.9% (overall); DCR: 78% in cervical cancer (Avelumab+Utomilumab)Safe, no DLTs36
NCT03414658IIHER2+ Metastatic Breast CancerTrastuzumab + Vinorelbine + Avelumab~100Study in progress during developmentNot reported37

Phase I Monotherapy in Advanced Malignancies (NCT01307267)

The first-in-human evaluation of Utomilumab was a Phase I, open-label, multicenter dose-escalation study involving 55 patients with a range of advanced solid tumors.[10] The primary goal was to assess safety and determine the maximum tolerated dose (MTD). Patients received intravenous infusions of Utomilumab every 4 weeks at doses escalating from 0.006 mg/kg up to 10 mg/kg.[10]

The results of this foundational study established the defining characteristic of Utomilumab: its exceptional safety profile. No dose-limiting toxicities (DLTs) were encountered at any dose level evaluated, and thus an MTD was not reached.[10] The most common treatment-related adverse events were consistently low-grade (Grade 1 or 2) and included fatigue, pyrexia, decreased appetite, dizziness, and rash. Most importantly, there were no clinically significant elevations in liver transaminases, a finding that stood in stark contrast to the known hepatotoxicity of urelumab and validated the "safety-by-design" approach of the IgG2 isotype.[10]

While safety was excellent, the evidence of anti-tumor activity as a monotherapy was modest. The overall objective response rate (ORR) across all solid tumor types was only 3.8% (95% CI, 0.5%–13.0%).[10] However, a potentially promising signal emerged from a small cohort of patients with Merkel cell carcinoma, a highly immunogenic skin cancer. In this subgroup, the ORR was 13.3%, which included one patient with a complete response and another with a partial response.[10] This suggested that while Utomilumab lacked broad monotherapy activity, it might possess some efficacy in tumor types that are inherently more visible to the immune system.

Combination Therapy Regimens: The Search for Synergy

Given the limited monotherapy activity, the clinical development strategy for Utomilumab quickly pivoted to focus on combination therapies, aiming to find a partner agent that could synergize with its immune-stimulatory mechanism.

With Rituximab in CD20+ Non-Hodgkin Lymphomas (NCT01307267)

A cohort of the same Phase I study evaluated Utomilumab in combination with rituximab, a standard-of-care anti-CD20 antibody, in 67 patients with relapsed or refractory CD20+ non-Hodgkin's lymphoma (NHL).[32] The scientific rationale for this combination was compelling: rituximab is known to mediate antibody-dependent cellular cytotoxicity (ADCC), a process carried out by NK cells and other immune effectors that express 4-1BB upon activation. The hypothesis was that Utomilumab could provide a costimulatory signal to these very cells, enhancing their ADCC activity and leading to a more potent anti-lymphoma effect.[31]

The results were among the most promising of the entire Utomilumab program. The combination maintained a favorable safety profile, with no MTD being reached at doses up to 10 mg/kg.[32] In this heavily pretreated patient population, the combination achieved an ORR of 21.2% (95% CI, 12.1%-33.0%), which included four complete responses and ten partial responses.[32] This represented a clinically meaningful signal and suggested that Utomilumab could be effective when paired with an agent that directly engages the innate and adaptive immune cells it is designed to stimulate.

With PD-1 Blockade (Pembrolizumab, NCT02179918)

Following the strong preclinical rationale, a Phase Ib study was initiated to evaluate Utomilumab in combination with the anti-PD-1 checkpoint inhibitor pembrolizumab in 23 patients with advanced solid tumors.[12] This trial tested the central immuno-oncology hypothesis of combining a T-cell accelerator (4-1BB agonist) with a brake release (PD-1 inhibitor).[16]

The combination was again found to be well-tolerated, with no DLTs observed.[12] The efficacy signals were encouraging and represented the high point for Utomilumab in solid tumors. The ORR was 26.1%, with six patients achieving a confirmed response. This included two complete responses (one in a patient with small cell lung cancer and another with renal cell carcinoma) and four partial responses. Importantly, many of these responses were durable, with some patients remaining on treatment for nearly a year.[12] These data provided the strongest clinical evidence to support the IO-IO combination strategy for Utomilumab.

With CCR4 Inhibition (Mogamulizumab, NCT02444793)

To address the therapeutic paradox of 4-1BB expression on both effector and regulatory T cells, a Phase Ib study combined Utomilumab with mogamulizumab, an antibody targeting CCR4, a chemokine receptor expressed on a major subset of Tregs.[28] The goal was to selectively deplete these immunosuppressive cells, thereby creating a more favorable immune microenvironment for Utomilumab to act upon.[15]

The combination proved to be safe and tolerable in the 24 enrolled patients, and pharmacodynamic analyses confirmed the intended biological effect of Treg depletion in the peripheral blood.[15] Despite this, the clinical efficacy was profoundly disappointing. The ORR was a mere 4.2%, consisting of a single partial response in a patient with PD-L1-refractory squamous lung cancer. Nine other patients achieved stable disease as their best response.[15] This result suggested that Treg depletion, at least via this mechanism, was not sufficient to unlock a potent anti-tumor effect for a mild agonist like Utomilumab.

With PD-L1 Blockade (Avelumab) and Other Agonists

Utomilumab was also evaluated in more complex, multi-drug regimens. In a Phase 1/2 trial for patients with advanced gynecologic malignancies, a combination of Utomilumab and the anti-PD-L1 antibody avelumab was tested. In a small, heavily pretreated cohort of patients with cervical cancer, this combination yielded an ORR of 11% but a promising disease control rate (DCR) of 78%. However, across all arms of the study, the multi-drug regimens, while safe, did not produce significant response rates.[36] Similarly, in a trial for advanced gastrointestinal malignancies (NCT03217747), a triplet combination of Utomilumab, avelumab, and the OX40 agonist ivuxolimab was found to be safe but produced only "modest activity," with no objective responses and a best outcome of stable disease in 37.5% of patients.[35]

A holistic view of the clinical data reveals a distinct pattern of context-dependent efficacy and diminishing returns. The most encouraging results for Utomilumab were observed in settings with a clear and potent synergistic partner, such as with rituximab in NHL (leveraging ADCC) or with pembrolizumab in immunologically active tumors (leveraging checkpoint blockade). The ORRs of 21.2% and 26.1% in these trials, respectively, stand in sharp contrast to the 3.8% ORR in solid tumor monotherapy and the 4.2% ORR with mogamulizumab.[11] As the therapeutic setting shifted to less immunologically "hot" solid tumors, like pancreatic and colorectal cancer, or when combined with agents targeting other complex pathways, the efficacy signal largely disappeared, even as the favorable safety profile was maintained. This pattern suggests that Utomilumab was not a potent enough agonist to drive responses on its own or to create synergy in unfavorable biological contexts. It could only provide a modest boost when paired with a powerful, contextually appropriate partner in a receptive patient population.

Pharmacokinetic and Pharmacodynamic Profile

Pharmacokinetics (PK)

The study of Utomilumab's pharmacokinetics—how the body absorbs, distributes, metabolizes, and excretes the drug—revealed a predictable and favorable profile for a monoclonal antibody therapeutic.

  • Linearity and Dose Proportionality: Across its Phase I dose-escalation studies, Utomilumab demonstrated linear pharmacokinetics over a very broad dose range, from 0.006 mg/kg up to 10 mg/kg.[10] Linearity means that its clearance from the body is not saturated within this dose range. Consequently, key exposure parameters such as the maximum serum concentration () and the area under the concentration-time curve (AUC) increased in a dose-proportional manner. This is a highly desirable characteristic, as it allows for predictable and reliable dosing.
  • Consistency in Combination: A crucial finding for its development as a combination agent was that the pharmacokinetic profile of Utomilumab was not meaningfully altered when co-administered with other monoclonal antibodies, including rituximab and pembrolizumab. Likewise, Utomilumab did not affect the PK of its partner drugs.[12] This lack of significant drug-drug interaction simplifies the development of combination regimens, as dose adjustments due to metabolic interference are not required.
  • Immunogenicity: As a fully human monoclonal antibody, Utomilumab was designed to have low immunogenicity. Clinical data confirmed this, showing a very low incidence of treatment-induced anti-drug antibodies (ADAs). For example, in the rituximab combination study, ADAs were detected in only 1.5% of patients.[32] In the patients who did develop ADAs, their presence did not appear to have a clinically relevant impact on the drug's safety, pharmacokinetics, or efficacy.[10]
  • ADME Profile: While detailed studies on its absorption, distribution, metabolism, and excretion (ADME) are not fully detailed in the provided materials, as a large protein administered intravenously, its disposition follows predictable pathways. Absorption is immediate and complete. Distribution occurs primarily within the vascular and interstitial compartments. Metabolism and elimination do not rely on hepatic (e.g., cytochrome P450) or renal pathways but occur through widespread proteolytic catabolism, where the antibody is broken down into constituent amino acids by cells throughout the body. This is the typical clearance mechanism for endogenous and therapeutic immunoglobulins.

Pharmacodynamics (PD)

Pharmacodynamic assessments, which measure the biological effect of a drug on the body, provided evidence that Utomilumab was engaging its target and modulating the immune system as intended.

  • Target Engagement: Analysis of circulating biomarkers in patients from clinical trials provided evidence consistent with the engagement of the 4-1BB/CD137 receptor by Utomilumab.[10]
  • Immune Cell Modulation: The biological consequences of this engagement were observed in some patient cohorts. In the combination study with pembrolizumab, a trend was noted where patients who responded to therapy exhibited higher levels of activated memory and effector CD8+ T cells in their peripheral blood compared to non-responders.[12] This provides a correlative link between clinical benefit and the desired T-cell activation. More direct evidence came from the analysis of paired tumor biopsies from a patient with follicular lymphoma who achieved a complete response in the rituximab combination study. The post-treatment biopsy showed a clear increase in T-cell infiltration into the tumor and enhanced markers of cytotoxic activity, providing a mechanistic snapshot of the drug's anti-tumor effect within the tumor microenvironment.[31]

Consolidated Safety and Tolerability Assessment

Overall Profile

The most consistent and unequivocally positive finding from the entire clinical development program of Utomilumab was its excellent safety and tolerability profile.[6] This favorable profile was maintained whether Utomilumab was administered as a monotherapy or in combination with a diverse range of other anti-cancer agents, including cytotoxic chemotherapy, other monoclonal antibodies, and novel immunotherapies.[12]

Common Adverse Events

The treatment-related adverse events (TRAEs) reported in clinical trials were predominantly mild to moderate in severity (NCI CTCAE Grade 1 or 2).[10] The most frequently observed TRAEs across studies included a constellation of symptoms typical of general immune stimulation:

  • Fatigue [10]
  • Pyrexia (fever) [10]
  • Rash and/or Pruritus (itching) [10]
  • Decreased appetite [10]
  • Nausea [12]

Absence of Severe Toxicity

A hallmark of the Utomilumab safety profile was the remarkable absence of severe toxicity. Across multiple dose-escalation studies, no dose-limiting toxicities (DLTs) were observed, and the maximum tolerated dose (MTD) was frequently not reached, even at doses as high as 10 mg/kg administered every four weeks.[10] The incidence of severe (Grade 3 or 4) TRAEs was very low, and treatment discontinuations due to toxicity were rare.[10]

Lack of Hepatotoxicity

The most critical safety differentiator for Utomilumab was the consistent lack of clinically relevant hepatotoxicity.[6] The severe and sometimes fatal liver inflammation (elevated transaminases) that curtailed the development of the first-generation 4-1BB agonist, urelumab, was not observed in patients treated with Utomilumab. This successful outcome was a direct validation of the "safety-by-design" hypothesis, which posited that the unique properties of the IgG2 isotype—specifically its requirement for secondary cross-linking to achieve potent agonism—would mitigate the risk of systemic immune activation and off-target liver damage.[6] This achievement demonstrated that the severe toxicities of 4-1BB agonism were not an unavoidable on-target effect but could be engineered out of a therapeutic molecule.

Comparative Analysis: Utomilumab within the 4-1BB Agonist Landscape

A Tale of Two Agonists: Utomilumab vs. Urelumab

The clinical development of 4-1BB agonists is best understood through the comparative lens of its two pioneering agents, Utomilumab and urelumab. These two antibodies, while targeting the same receptor, represent fundamentally different philosophies of drug design and resulted in diametrically opposed clinical outcomes, thereby defining the central therapeutic dilemma for this entire class of drugs.

  • Urelumab (BMS-663513): Developed by Bristol Myers Squibb, urelumab is a fully human IgG4 monoclonal antibody. It binds to the CRD1 domain of 4-1BB, a site distinct from the natural ligand binding pocket, and thus acts in a non-ligand-competing manner. Its IgG4 isotype confers potent, intrinsic agonistic activity that can occur without the need for FcγR cross-linking, although cross-linking can further enhance its effects. This potent mechanism led to encouraging early signs of anti-tumor activity in clinical trials. However, this potency was inextricably linked to severe, dose-dependent hepatotoxicity, which included fatal adverse events and ultimately halted its broad development.[4] Urelumab embodied the "high-risk, high-reward" approach.
  • Utomilumab (PF-05082566): Developed by Pfizer, Utomilumab is a fully human IgG2 monoclonal antibody. It binds to the CRD2/CRD3 domains, directly competing with 4-1BBL. Its IgG2 isotype renders it a much weaker agonist, with its activity being highly dependent on FcγR-mediated cross-linking to achieve effective receptor clustering. This design was a deliberate attempt to create a safer, conditionally active agonist. The strategy was successful in terms of safety, as Utomilumab consistently demonstrated an excellent tolerability profile devoid of the hepatotoxicity seen with urelumab. However, this safety came at the cost of efficacy, with the antibody showing only modest and inconsistent anti-tumor activity.[1] Utomilumab embodied the "safety-first" approach.

Table 3: Comparative Profile of First-Generation 4-1BB Agonists: Utomilumab vs. Urelumab

FeatureUtomilumab (PF-05082566)Urelumab (BMS-663513)
IsotypeHuman IgG2Human IgG4
4-1BB Binding DomainCRD2/CRD3CRD1
Ligand (4-1BBL) CompetitionYes (Blocking)No (Non-blocking)
Agonism MechanismWeak; requires FcγR cross-linking for potent activityPotent; FcγR-independent, but enhanced by cross-linking
Clinical Efficacy ProfileModest / LimitedPromising early signals
Key Safety LiabilityLack of compelling efficacySevere Hepatotoxicity
Development StatusDiscontinued (due to efficacy)Severely hampered by toxicity; limited ongoing trials

Lessons for Second-Generation Agonists

The divergent clinical paths of Utomilumab and urelumab provided invaluable, albeit costly, lessons for the field of immuno-oncology. Together, they starkly illustrated that the therapeutic window for systemic 4-1BB agonism is exceptionally narrow. An agonist that is too potent (urelumab) risks unacceptable toxicity, while one that is too mild (Utomilumab) risks therapeutic futility. This realization has driven the development of a diverse and sophisticated second generation of 4-1BB agonists, all of which are designed to circumvent this dilemma by achieving potent, yet tumor-localized, conditional activation.

Current strategies in clinical development are largely based on the lessons learned from these first-generation agents and include:

  • Tumor-Targeted Bispecific Antibodies: These molecules contain one arm that binds to 4-1BB and a second arm that binds to a tumor-associated antigen (e.g., HER2, FAP, PD-L1). This design ensures that the 4-1BB agonistic activity is physically tethered to the tumor cell surface, forcing the cross-linking and activation of T cells to occur only within the tumor microenvironment.[4]
  • Engineered Fc Domains: Some newer antibodies feature Fc domains that have been engineered to preferentially bind to specific FcγRs. For example, enhancing binding to the inhibitory receptor FcγRIIb, which is highly expressed on B cells and myeloid cells in the tumor, while reducing binding to activating FcγRs, is a strategy to promote effective cross-linking with a better safety profile.[7]
  • Novel Formats: The field is also exploring other innovative formats, such as trispecific antibodies that can engage a tumor antigen, 4-1BB, and another immune receptor simultaneously, or antibody-cytokine fusions that deliver an additional pro-inflammatory signal directly to the site of 4-1BB engagement.[4]

Discontinuation and Future Perspectives

Strategic Discontinuation

In its first-quarter pipeline update on May 1, 2018, Pfizer announced that it was discontinuing development of five early-stage oncology programs. This included three separate Phase 1 trials for Utomilumab: the monotherapy study and the combinations with pembrolizumab and mogamulizumab.[9] While the company's public statements did not provide a detailed scientific rationale for the decision, the cumulative evidence from the completed and ongoing clinical trials pointed to an unavoidable conclusion: Utomilumab lacked a sufficiently compelling and differentiated efficacy profile to warrant continued investment.[4]

Despite its outstanding safety record, the anti-tumor activity observed was too modest and inconsistent. The promising signals seen in combination with rituximab in NHL and with pembrolizumab in some solid tumors were not strong enough to justify the immense cost and risk of proceeding to pivotal Phase 3 trials, especially in a fiercely competitive immuno-oncology landscape dominated by highly effective checkpoint inhibitors. The drug had successfully solved the safety problem of its predecessor but had failed to clear the efficacy bar necessary for a viable cancer therapeutic.

Concluding Perspective: The Legacy of Utomilumab

Utomilumab's journey through clinical development should not be viewed as a simple failure, but rather as a crucial and informative scientific experiment conducted on a clinical scale. Its primary legacy is the definitive demonstration that the severe on-target toxicities of 4-1BB agonism, particularly hepatotoxicity, are not an immutable property of the pathway but can be successfully engineered out of a therapeutic molecule. Utomilumab proved that a "safe" 4-1BB agonist was possible.

However, in doing so, it also revealed the other side of the coin: that this safety, achieved through a milder and more conditional mechanism of action, could come at the direct expense of therapeutic potency. The story of Utomilumab powerfully underscores a fundamental principle in immunotherapy: modulating powerful, pleiotropic immune pathways requires a "Goldilocks" approach. The therapeutic signal cannot be too strong, lest it trigger systemic, life-threatening toxicity; nor can it be too weak, lest it fail to overcome the profound immunosuppression within the tumor microenvironment.

The ultimate value of the Utomilumab program lies in the wealth of clinical, pharmacokinetic, and pharmacodynamic data it generated. This body of knowledge has provided a clear benchmark for safety and has directly informed the rational design of the entire next generation of 4-1BB agonists that now populate the clinical pipeline. These novel agents, with their sophisticated tumor-targeting and conditional activation mechanisms, are all, in essence, attempting to find the elusive sweet spot that Utomilumab and urelumab could not: a way to deliver a potent, urelumab-like signal but with the targeted precision and safety profile of Utomilumab. In this way, the legacy of Utomilumab is not an approved drug, but the critical scientific clarity it provided, paving the way for the more advanced and potentially more successful therapies of the future.

Works cited

  1. Utomilumab Overview - Creative Biolabs, accessed October 9, 2025, https://www.creativebiolabs.net/utomilumab-overview.htm
  2. YR1434 Anti-Human CD137 Recombinant Antibody(Utomilumab) DATA SHEET For Research Use Only! - KMD Bioscience, accessed October 9, 2025, https://www.kmdbioscience.com/userfiles/pdf/YR1434%20datasheet.pdf
  3. Utomilumab (PF-05082566) - Pfizer, accessed October 9, 2025, https://cdn.pfizer.com/pfizercom/news/asco/Utomilumab_Fact_Sheet_23MAY2018.PDF
  4. 4-1BB: An Established Target for Cancer Immunotherapy - Crown Bioscience Blog, accessed October 9, 2025, https://blog.crownbio.com/4-1bb-an-established-target-for-cancer-immunotherapy
  5. 4-1BB: A promising target for cancer immunotherapy - Frontiers, accessed October 9, 2025, https://www.frontiersin.org/journals/oncology/articles/10.3389/fonc.2022.968360/full
  6. 1258 PBMC humanized mouse model with clinical relevance in ..., accessed October 9, 2025, https://jitc.bmj.com/content/10/Suppl_2/A1306
  7. (PDF) The emerging landscape of novel 4-1BB (CD137) agonistic ..., accessed October 9, 2025, https://www.researchgate.net/publication/368028118_The_emerging_landscape_of_novel_4-1BB_CD137_agonistic_drugs_for_cancer_immunotherapy
  8. Finding new immunotherapy targets - Immune system agonists - News-Medical.Net, accessed October 9, 2025, https://www.news-medical.net/whitepaper/20230707/Finding-new-immunotherapy-targets-Immune-system-agonists.aspx
  9. Pfizer culls 5 experimental cancer trials and an acne drug in Q1 ..., accessed October 9, 2025, https://www.fiercebiotech.com/biotech/pfizer-culls-5-experimental-cancer-trials-acne-drug-q1-clear-out
  10. Phase I Study of Single-Agent Utomilumab (PF-05082566), a 4-1BB ..., accessed October 9, 2025, https://aacrjournals.org/clincancerres/article/24/8/1816/81312/Phase-I-Study-of-Single-Agent-Utomilumab-PF
  11. Phase I Study of Single-Agent Utomilumab (PF-05082566), a 4-1BB/CD137 Agonist, in Patients with Advanced Cancer - PubMed, accessed October 9, 2025, https://pubmed.ncbi.nlm.nih.gov/29549159/
  12. Phase Ib Study of Utomilumab (PF-05082566), a 4-1BB/CD137 Agonist, in Combination with Pembrolizumab (MK-3475) in Patients with Advanced Solid Tumors | Clinical Cancer Research - AACR Journals, accessed October 9, 2025, https://aacrjournals.org/clincancerres/article/23/18/5349/258640/Phase-Ib-Study-of-Utomilumab-PF-05082566-a-4-1BB
  13. Phase i study of single-agent utomilumab (PF-05082566), a 4-1bb/cd137 agonist, in patients with advanced cancer - Johns Hopkins University, accessed October 9, 2025, https://pure.johnshopkins.edu/en/publications/phase-i-study-of-single-agent-utomilumab-pf-05082566-a-4-1bbcd137
  14. Utomilumab in Patients With Immune Checkpoint Inhibitor-Refractory Melanoma and Non-Small-Cell Lung Cancer, accessed October 9, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC9379324/
  15. A phase Ib study of utomilumab (PF-05082566) in combination with mogamulizumab in patients with advanced solid tumors - PMC - PubMed Central, accessed October 9, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC6894203/
  16. Utomilumab - AWS, accessed October 9, 2025, https://pfe-pfizercom-prod.s3.amazonaws.com/news/asco/Pfizer_IO_41BB_UtomilumabFactSheet.pdf
  17. Utomilumab - Drug Targets, Indications, Patents - Synapse, accessed October 9, 2025, https://synapse.patsnap.com/drug/365f93561d414221b77141835a03e591
  18. Utomilumab Biosimilar (Anti-4-1BB) Antibody (HDBS0037) - Assay Genie, accessed October 9, 2025, https://www.assaygenie.com/anti-4-1bb-utomilumab-biosimilar-mab-hdbs0037/
  19. Buy Utomilumab Biosimilar - Anti-Human TNFRSF9 mAb - Research, accessed October 9, 2025, https://us.proteogenix.science/product/utomilumab-biosimilar-anti-human-tnfrsf9-mab-research-grade/
  20. Utomilumab (Anti-TNFRSF9 / 4-1BB / CD137) CD markers antibody | Read Reviews & Product Use Citations - Selleck Chemicals, accessed October 9, 2025, https://www.selleckchem.com/products/utomilumab-anti-tnfrsf9-4-1bb-cd137.html
  21. HY-P99056 Utomilumab - MedChem Express by Bio-Connect, accessed October 9, 2025, https://shop.bio-connect.nl/utomilumab-1417318-27-4-hy-p99056-m09
  22. Utomilumab (PF 05082566) | 4-1BB/CD137 Agonist | MedChemExpress, accessed October 9, 2025, https://www.medchemexpress.com/utomilumab.html
  23. Utomilumab - Thera-SAbDab - SAbDab: The Structural Antibody ..., accessed October 9, 2025, https://opig.stats.ox.ac.uk/webapps/sabdab-sabpred/therasabdab/therasummary/?INN=Utomilumab
  24. 4-1BB Antibodies in Oncology Clinical Trials: A Review - Allen Press, accessed October 9, 2025, https://meridian.allenpress.com/innovationsjournals-JIPO/article/8/2/121/506216/4-1BB-Antibodies-in-Oncology-Clinical-Trials-A
  25. Agonism of 4-1BB for immune therapy: a perspective on possibilities and complications, accessed October 9, 2025, https://www.frontiersin.org/journals/immunology/articles/10.3389/fimmu.2023.1228486/full
  26. Combination of 4-1BB Agonist and PD-1 Antagonist Promotes Antitumor Effector/Memory CD8 T Cells in a Poorly Immunogenic Tumor Model - AACR Journals, accessed October 9, 2025, https://aacrjournals.org/cancerimmunolres/article/3/2/149/467761/Combination-of-4-1BB-Agonist-and-PD-1-Antagonist
  27. Targeting of 4-1BB by monoclonal antibody PF-05082566 enhances T-cell function and promotes anti-tumor activity - PubMed, accessed October 9, 2025, https://pubmed.ncbi.nlm.nih.gov/22406983/
  28. A phase Ib study of utomilumab (PF-05082566) in combination with mogamulizumab in patients with advanced solid tumors | Journal for ImmunoTherapy of Cancer, accessed October 9, 2025, https://jitc.bmj.com/content/7/1/342
  29. A phase Ib study of utomilumab (PF-05082566) in combination with mogamulizumab in patients with advanced solid tumors - PubMed, accessed October 9, 2025, https://pubmed.ncbi.nlm.nih.gov/31801624/
  30. FcγR requirements and costimulatory capacity of Urelumab, Utomilumab, and Varlilumab, accessed October 9, 2025, https://www.frontiersin.org/journals/immunology/articles/10.3389/fimmu.2023.1208631/full
  31. First-in-Human Study of Utomilumab, a 4-1BB/CD137 Agonist, in Combination with Rituximab in Patients with Follicular and Other CD20 + Non-Hodgkin Lymphomas - AACR Journals, accessed October 9, 2025, https://aacrjournals.org/clincancerres/article/26/11/2524/284532/First-in-Human-Study-of-Utomilumab-a-4-1BB-CD137
  32. First-in-Human Study of Utomilumab, a 4-1BB/CD137 Agonist, in Combination with Rituximab in Patients with Follicular and Other CD20+ Non-Hodgkin Lymphomas - PubMed, accessed October 9, 2025, https://pubmed.ncbi.nlm.nih.gov/32144134/
  33. Phase Ib Study of Utomilumab (PF-05082566), a 4-1BB/CD137 Agonist, in Combination with Pembrolizumab (MK-3475) in Patients with Advanced Solid Tumors - PubMed, accessed October 9, 2025, https://pubmed.ncbi.nlm.nih.gov/28634283/
  34. Pfizer Presents Data from Phase 1b Trial Investigating Utomilumab (a 4-1BB agonist) in Combination with a Checkpoint Inhibitor, accessed October 9, 2025, https://www.pfizer.com/news/press-release/press-release-detail/pfizer_presents_data_from_phase_1b_trial_investigating_utomilumab_a_4_1bb_agonist_in_combination_with_a_checkpoint_inhibitor
  35. phase I/II trial of avelumab combinations with ivuxolimab ..., accessed October 9, 2025, https://academic.oup.com/oncolo/article/30/3/oyaf032/8096320
  36. Phase 1/2 trial of avelumab combined with utomilumab (4-1BB agonist), PF-04518600 (OX40 agonist), or radiotherapy in patients with advanced gynecologic malignancies - PubMed, accessed October 9, 2025, https://pubmed.ncbi.nlm.nih.gov/37864520/
  37. AVIATOR Study: Trastuzumab and Vinorelbine With Avelumab OR Avelumab & Utomilumab in Advanced HER2+ Breast Cancer - UCSF Clinical Trials, accessed October 9, 2025, https://clinicaltrials.ucsf.edu/trial/NCT03414658
  38. Utomilumab for treatment of solid tumours - Action Kidney Cancer, accessed October 9, 2025, https://actionkidneycancer.org/utomilumab-for-treatment-of-solid-tumours/
  39. Phase I Study of Single-Agent Utomilumab (PF ... - AACR Journals, accessed October 9, 2025, https://aacrjournals.org/clincancerres/article-pdf/24/8/1816/2050045/1816.pdf
  40. Phase Ib Study of Utomilumab (PF-05082566), a 4-1BB/CD137 Agonist, in Combination with Pembrolizumab (MK-3475) in Patients with - SciSpace, accessed October 9, 2025, https://scispace.com/pdf/phase-ib-study-of-utomilumab-pf-05082566-a-4-1bb-cd137-1799xrr2vt.pdf
  41. The emerging landscape of novel 4-1BB (CD137) agonistic drugs for cancer immunotherapy - PMC - PubMed Central, accessed October 9, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC9897756/
  42. Final results of urelumab, an anti-CD137 agonist monoclonal antibody, in combination with cetuximab or nivolumab in patients with advanced solid tumors | Journal for ImmunoTherapy of Cancer, accessed October 9, 2025, https://jitc.bmj.com/content/12/3/e007364

Published at: October 9, 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.