MedPath

PF-08046037 Advanced Drug Monograph

Published:May 30, 2025

Generic Name

PF-08046037

Comprehensive Report on PF-08046037: An Investigational PD-L1 Targeted TLR7 Agonist Immune-Stimulating Antibody Conjugate

1. Introduction and Overview of PF-08046037

1.1. Identification and Nomenclature

PF-08046037 is an investigational therapeutic agent currently undergoing clinical evaluation. Its primary development code, PF-08046037, is consistently utilized in recent scientific communications and clinical trial registries.[1] The compound was formerly designated SGN-PDL1iT, a nomenclature that indicates its origin and initial development phase by Seagen.[1] Additional synonyms found in various databases include PDL1iT.[2] The consistent cross-referencing of SGN-PDL1iT with PF-08046037 is essential for tracing the molecule's developmental history and acknowledging its technological provenance from Seagen's research pipeline.

1.2. Originator and Current Developer

PF-08046037 was conceived and underwent its initial stages of development at Seagen, Inc., a biotechnology firm recognized for its pioneering work and extensive expertise in the field of antibody-drug conjugate (ADC) technology.[2] Subsequent to Pfizer Inc.'s strategic acquisition of Seagen, PF-08046037 has been integrated into Pfizer's oncology portfolio and is now under active development by Pfizer.[1] This transition is a significant marker of Pfizer's intensified commitment to oncology, particularly through the incorporation of Seagen's advanced ADC and Immune-Stimulating Antibody Conjugate (ISAC) platforms.[4] The acquisition has endowed Pfizer with promising pipeline candidates like PF-08046037 and the specialized expertise crucial for their continued advancement.

1.3. Drug Classification

PF-08046037 is categorized as an Immune-Stimulating Antibody Conjugate (ISAC).[1] More precisely, it is defined as a Programmed Death-Ligand 1 (PD-L1) targeted Toll-like Receptor 7 (TLR7) agonist ISAC.[1] This classification is pivotal. Unlike conventional ADCs, which typically deliver cytotoxic payloads to directly kill cancer cells, ISACs such as PF-08046037 are engineered to deliver immunomodulatory agents. Their principal objective is to activate and redirect the patient's own immune system to combat the malignancy, thereby representing an innovative therapeutic modality within the expanding landscape of antibody-based treatments.[6]

1.4. Therapeutic Rationale and Significance

The core therapeutic strategy underpinning PF-08046037 is the induction of a targeted and potent anti-cancer immune response. This is accomplished by the specific delivery of a powerful TLR7 agonist to tumor-associated antigen-presenting cells (APCs) that express PD-L1.[1] The intended biological sequelae include the repolarization of the typically immunosuppressive tumor microenvironment (TME), the enhancement of antigen presentation by activated APCs, and the consequent establishment of robust, adaptive anti-tumor immunity.[1] PF-08046037 seeks to address the considerable unmet medical need for novel immuno-oncology (IO) strategies, especially for patients whose cancers are refractory to existing therapies or for tumor types that are traditionally non-responsive to established IO agents. The therapeutic rationale is founded on a sophisticated understanding of tumor immunology: PD-L1 serves not only as an immune checkpoint but also as a convenient surface marker for targeted delivery to immune cells within the TME. Localized TLR7 agonism, when precisely delivered, can potently activate innate immunity, which subsequently bridges to and amplifies adaptive T-cell mediated anti-tumor responses. This approach aims to overcome tumor-induced immune suppression at multiple, interconnected levels.

The active clinical development of PF-08046037 by Pfizer, following its acquisition from Seagen, highlights a strategic imperative to lead in next-generation cancer therapies, especially those leveraging innovative ADC and ISAC platforms. Pfizer, having not previously achieved a major market success with a standalone PD-(L)1 monoclonal antibody comparable to some competitors, appears to be pursuing a differentiated strategy by targeting PD-L1 with a payload-delivering mechanism.[4] This is evidenced by Pfizer's multi-billion dollar investment in Seagen to acquire its ADC technology and pipeline [4], the Seagen-originated nature of PF-08046037 [2], and Pfizer's decision to advance it into Phase 1 clinical trials.[2] This suggests a strategic direction to compete in the immuno-oncology space not by replicating existing checkpoint inhibitors, but by innovating with targeted delivery of immune activators.

Furthermore, the design and mechanism of PF-08046037 represent a significant evolution in the field of immunotherapy. It transcends the concept of merely "releasing the brakes" on the immune system, characteristic of standard checkpoint inhibitors. Instead, it embodies a more proactive strategy of "stepping on the gas" in a highly targeted manner. This involves the direct activation and reprogramming of key innate immune cells within the tumor microenvironment. Standard anti-PD-1/PD-L1 therapies function by blocking inhibitory signals, thereby relying on a pre-existing, albeit suppressed, anti-tumor immune response. In contrast, PF-08046037, through the delivery of a TLR7 agonist to APCs [1], aims to initiate or substantially amplify an immune response by directly stimulating these critical orchestrators of immunity. This localized activation is intended to convert an immunologically "cold" or unresponsive TME into an inflamed, "hot" TME, potentially expanding the cohort of patients who can derive benefit from immunotherapy. This approach reflects a sophisticated understanding of the TME's complexity and the necessity for multi-faceted interventions, marking a progression from broad immune checkpoint blockade to precision immune stimulation.[6]

2. Molecular Profile and Mechanism of Action

PF-08046037 is an intricately engineered molecule, comprising three key structural elements designed to work in concert for targeted immune activation.

2.1. Structural Components

2.1.1. Anti-PD-L1 Monoclonal Antibody

The targeting component of PF-08046037 is a monoclonal antibody (mAb) specifically developed to bind to human Programmed Death-Ligand 1 (PD-L1).[1] PD-L1 is a transmembrane protein expressed on various tumor cells and, crucially for the mechanism of this ISAC, on multiple types of antigen-presenting cells (APCs) within the tumor microenvironment (TME). A significant engineering feature of this antibody is the incorporation of specific mutations within its Fc (fragment crystallizable) region. These mutations are designed to substantially reduce the antibody's binding affinity to Fcγ receptors (FcγRs) present on various immune effector cells.[1] The selection of an anti-PD-L1 mAb provides the necessary specificity for delivering the immunomodulatory payload. The reduction in FcγR binding is a critical design element for both safety and efficacy. It serves to prevent off-target activation of other immune cell populations (such as NK cells, macrophages, and neutrophils) via FcγR engagement, which could otherwise lead to undesirable systemic toxicities or antibody-dependent cellular cytotoxicity (ADCC) against normal, PD-L1-expressing tissues. This engineering enhances the selective delivery of the TLR7 agonist payload to the intended PD-L1-positive APCs and minimizes unintended systemic immune perturbations.

2.1.2. Toll-like Receptor 7 (TLR7) Agonist Payload

The immunomodulatory "warhead" of PF-08046037 is a potent, synthetic small-molecule agonist of Toll-like Receptor 7 (TLR7). This agonist belongs to the imidazoquinoline chemical class.[1] A key characteristic of this payload is its intentional design for low membrane permeability.[1] TLR7 is an endosomally located pattern recognition receptor, primarily expressed by APCs such as dendritic cells (DCs) and macrophages. Its activation triggers robust innate immune responses, including the production of Type I interferons (IFN-α/β) and other pro-inflammatory cytokines, and promotes APC maturation and enhanced antigen presentation capabilities. The imidazoquinoline class includes well-established synthetic TLR7 agonists known for their immune-stimulatory properties. The low membrane permeability of the payload is a deliberate design feature. It ensures that once the agonist is released from the ISAC within the target cell's endosomal system, it remains localized within these TLR7-containing compartments. This maximizes on-target activity by concentrating the agonist at the site of its receptor and minimizes its potential to leak into the cytoplasm or extracellular space, which could lead to off-target effects or diminished efficacy. This targeted intracellular delivery contrasts with the challenges faced by systemically administered TLR agonists, which can cause widespread, non-specific immune activation.[6]

2.1.3. Linker Technology

The anti-PD-L1 antibody is covalently conjugated to the TLR7 agonist payload through a specialized linker system. This linker is identified as a mannosidase-cleavable maleimidopropionyl-mannose linker, often abbreviated as mp-mann.[1] The linker's properties are paramount for the overall performance and therapeutic index of the ISAC. It must exhibit high stability in the systemic circulation to prevent premature release of the potent TLR7 agonist, which could otherwise lead to systemic toxicity and a reduction in the amount of active payload reaching the target cells. The mp-mann linker is specifically designed to be susceptible to cleavage by mannosidase enzymes. These enzymes are characteristically abundant within the endosomal and lysosomal compartments of APCs. This enzymatic targeting strategy ensures that the TLR7 agonist is preferentially and efficiently released at the site of its intracellular target (TLR7, located in endosomes) following the binding and internalization of the ISAC into PD-L1-expressing APCs.

2.2. Detailed Pharmacodynamics and Mechanism of Action (MoA)

The mechanism of action of PF-08046037 is a carefully orchestrated multi-step process designed to achieve highly targeted and localized immune activation:

  • Step 1: Targeted Delivery to PD-L1 Expressing APCs: Following systemic administration, PF-08046037 circulates and, by virtue of its anti-PD-L1 antibody component, selectively binds to PD-L1 molecules expressed on the cell surface of APCs (e.g., macrophages, dendritic cells). These target cells are typically located within the tumor microenvironment and associated immune compartments, such as tumor-draining lymph nodes (tdLNs).[1]
  • Step 2: Internalization, Linker Cleavage, and Payload Release: Upon binding to PD-L1, the entire ISAC-PD-L1 complex is internalized by the APC, typically via receptor-mediated endocytosis, into endosomal vesicles. Within the acidic and enzyme-rich environment of these endosomes, the mannosidase-cleavable mp-mann linker encounters mannosidase enzymes. These enzymes catalyze the cleavage of the linker, leading to the precise and localized release of the imidazoquinoline-based TLR7 agonist payload directly into the TLR7-containing endosomes of the APC.[1]
  • Step 3: TLR7 Activation and Subsequent Innate Immune Stimulation: The liberated TLR7 agonist is now free to bind to and activate its cognate receptor, TLR7, which is expressed on the endosomal membrane within the APC. This receptor engagement initiates downstream intracellular signaling cascades, primarily mediated through the MyD88-dependent pathway. This signaling culminates in a range of innate immune responses, most notably:
  • The robust production and secretion of pro-inflammatory cytokines (such as TNF-α, IL-6, and IL-12) and, critically, Type I interferons (IFN-α/β).[1]
  • The enhanced maturation of the APCs themselves. This maturation process is characterized by the upregulation of co-stimulatory molecules (e.g., CD80, CD86, CD40) and Major Histocompatibility Complex (MHC) class I and class II molecules on the APC surface.[1]
  • Step 4: Repolarization of the Tumor Microenvironment (TME) and Enhanced Adaptive Anti-Tumor Immunity: The localized activation of APCs by PF-08046037 is intended to fundamentally alter the TME, shifting it from an immunosuppressive state to an immunostimulatory one. This reprogramming involves several key changes:
  • A shift in macrophage polarization, favoring the development of pro-inflammatory, anti-tumor M1-like macrophages over immunosuppressive M2-like macrophages. Preclinical data specifically demonstrate a reduction in CD206+ (an M2 marker) suppressive macrophages within the TME.[1]
  • The promotion of dendritic cell maturation and activation, significantly enhancing their capacity to process tumor-associated antigens and present them effectively to T cells.
  • Ultimately, these effects converge to facilitate more effective priming, activation, clonal expansion, and recruitment of tumor-specific CD8+ cytotoxic T lymphocytes (CTLs) and CD4+ helper T cells. This fosters a robust and potentially durable adaptive anti-tumor immune response.[1] Preclinical evidence supports an observed increase in the activity of cytotoxic cells following PF-08046037 treatment.[1]

The sophisticated multi-component architecture of PF-08046037—comprising a PD-L1 specific antibody, an engineered Fc domain for diminished off-target interactions, a highly specific mannosidase-cleavable linker, and a low membrane permeability TLR7 agonist payload—reflects an advanced engineering strategy. This design aims to achieve an unparalleled level of precision in delivering immune stimulation directly to target cells within the TME. The overarching goal is to maximize therapeutic efficacy through potent local immune activation while concurrently minimizing the systemic immune-related adverse events (irAEs) that have historically limited the utility of less targeted immunotherapies or the systemic administration of potent immune agonists.[1] Each component of PF-08046037 is meticulously tailored to address specific challenges in immunotherapy. The antibody ensures targeting specificity to PD-L1+ APCs.[1] Fc engineering and the low permeability of the payload are critical safety features designed to curtail off-target immune cell activation and systemic exposure of the potent agonist.[1] The mannosidase-cleavable linker guarantees that the TLR7 agonist is released predominantly within the endosomes of target APCs, where TLR7 is situated, thereby maximizing on-target activity.[1] This targeted approach is fundamentally different from the systemic administration of immune-stimulating agents like cytokines or free TLR agonists, which often lead to dose-limiting systemic toxicities due to widespread, non-specific immune activation.[6] Consequently, PF-08046037's design endeavors to fundamentally improve the therapeutic index by concentrating the immune-stimulatory effect precisely where it is most needed—within the tumor.

While PD-L1 is widely recognized as an immune checkpoint molecule, PF-08046037 primarily leverages PD-L1 expression as a "molecular address" for the targeted delivery of its immunomodulatory payload to APCs. Although the antibody's principal function in this construct is delivery, its binding to PD-L1 on APCs might also subtly modulate the PD-1/PD-L1 inhibitory axis in the immediate tumor microenvironment. This interaction could add a layer of complexity to its overall effect, even with blunted Fc effector functions. The dominant therapeutic action stems from the released TLR7 agonist; however, the physical engagement of the antibody with PD-L1 on an APC could sterically hinder PD-L1's interaction with PD-1 on adjacent T cells to some degree. While not the primary mechanism, this incidental interference could contribute, albeit likely minorly, to the overall immune modulation.

The selection of a mannosidase-cleavable linker (mp-mann) is a highly specific and astute design feature, tailored to the unique enzymatic milieu of APCs, which are notably rich in mannosidases within their endo-lysosomal compartments. This enzymatic specificity is crucial for ensuring that the potent TLR7 agonist payload is released predominantly within the target APCs and, more precisely, within the endosomal compartment where TLR7 receptors are located. This maximizes the on-target effect and significantly minimizes the risk of premature payload release in systemic circulation or within non-target cells, which could precipitate systemic toxicity. The efficacy and safety of any ADC or ISAC are critically dependent on linker stability in circulation and efficient cleavage at the target site. The mp-mann linker's reliance on mannosidase activity for cleavage [1] ensures agonist liberation precisely where its molecular target resides. This contrasts with other linker strategies, such as those cleaved by more ubiquitously active enzymes like cathepsins, which might lead to less specific payload release. The high degree of specificity afforded by the mannosidase-cleavable linker is, therefore, central to achieving the desired therapeutic index for PF-08046037.

The following table summarizes the key components of PF-08046037 and their respective functions:

Table 1: Key Components of PF-08046037 and Their Functions

ComponentDescription/Key FeaturesPrimary Function in PF-08046037
Anti-PD-L1 Monoclonal AntibodyHumanized or human antibody targeting human PD-L1.Targets PD-L1 on APCs for selective ISAC delivery into the tumor microenvironment.
Fc Domain MutationsSpecific amino acid alterations in the antibody's Fc region.Engineered to reduce FcγR binding, thereby minimizing off-target immune cell activation and enhancing safety and specificity.
Imidazoquinoline-based TLR7 Agonist PayloadPotent, synthetic small-molecule agonist of Toll-like Receptor 7.Activates endosomal TLR7 in APCs upon release, initiating innate immune responses and promoting adaptive anti-tumor immunity.
Low Membrane Permeability of PayloadPhysicochemical property of the TLR7 agonist.Designed to keep the agonist localized within the target cell's endosomes post-release, reducing off-target effects.
Mannosidase-cleavable mp-mann LinkerConnects the antibody to the payload; specifically cleaved by mannosidase enzymes.Provides stability in systemic circulation and ensures specific release of the TLR7 agonist payload within the mannosidase-rich endosomes of target APCs.

3. Preclinical Evidence Base

The preclinical development of PF-08046037 has yielded a substantial body of evidence supporting its proposed mechanism of action and anti-tumor potential. These studies span in vitro cellular assays and in vivo animal models.

3.1. In Vitro Activity

Laboratory studies have demonstrated that PF-08046037 can effectively engage its molecular and cellular targets. The compound induces potent and selective activation of TLR7 reporter cells.[1] This foundational experiment confirms that the TLR7 agonist payload, once released from the ISAC, is biologically active and capable of stimulating the TLR7 signaling pathway in a controlled cellular system. The "selective" nature of this activation likely implies dependency on appropriate ISAC processing or delivery, as a free agonist might elicit broader activation.

More importantly, PF-08046037 has been shown to elicit potent and selective activation of primary human macrophages and dendritic cells, provided these cells express PD-L1.[1] This finding is of greater clinical relevance as it validates that the ISAC can successfully target bona fide human APCs—the intended cellular targets in patients—in a PD-L1-dependent manner. The process involves internalization, payload release, and the subsequent triggering of the desired TLR7-mediated activation pathways within these key immune cells. This provides direct in vitro evidence for the intended cellular mechanism of action.

3.2. In Vivo Efficacy and Pharmacodynamics

Studies conducted in syngeneic mouse tumor models, specifically human PD-L1 knock-in models (hPD-L-1.CT26 colon carcinoma and hPD-L-1.MC38 colon adenocarcinoma, implanted in mice engineered to express human PD-L1), have produced significant findings regarding PF-08046037's efficacy and its effects on the immune system:

  • Anti-tumor Efficacy: PF-08046037 exhibits single-agent anti-tumor efficacy in these well-characterized syngeneic models.[1] The demonstration of monotherapy efficacy in immunocompetent models is a critical preclinical milestone. It suggests that the ISAC, by itself, can orchestrate a sufficiently potent immune response to control or eradicate established tumors. The use of models expressing human PD-L1 ensures that the antibody component of the ISAC engages its target with clinically relevant specificity and affinity.
  • Induction of Immunological Memory: Treatment with PF-08046037 has been observed to provide anamnestic protection against subsequent tumor rechallenge in these preclinical models.[1] This is a particularly compelling finding. Anamnestic protection, or immunological memory, implies that the initial immune response generated by PF-08046037 is not merely transient but leads to the development of a long-lasting adaptive immune memory. If this phenomenon translates to human patients, it could signify the potential for durable, long-term remissions or control of cancer, a primary objective of cancer immunotherapy.
  • Pharmacodynamic Effects (Consistent with the Intended Mechanism of Action): A range of in vivo pharmacodynamic effects have been documented, aligning with the proposed mechanism of PF-08046037:
  • Induction of acute tumor inflammation, indicative of an active immune response being mounted within the tumor site.[1]
  • A quantifiable reduction in CD206+ suppressive macrophages within the TME. CD206 is a marker often associated with M2-like, immunosuppressive macrophages; its reduction suggests a repolarization towards an M1-like, immunostimulatory macrophage phenotype.[1]
  • Upregulation of cellular activation markers and co-stimulatory molecules on APCs. This effect was observed both on APCs located within the tumor itself and on those in the tumor-draining lymph nodes (tdLNs), which are critical sites for the priming of adaptive immune responses.[1]
  • An observed increase in the activity of cytotoxic T cells, the primary effector cells responsible for killing tumor cells.[1]

These pharmacodynamic observations provide strong mechanistic support for PF-08046037's in vivo activity. They collectively paint a picture of comprehensive immune modulation: the drug effectively reaches the tumor, activates local APCs, reverses key immunosuppressive features of the TME (such as the prevalence of M2-like macrophages), enhances APC function in crucial immune priming sites (tdLNs), and ultimately boosts the effector phase of the anti-tumor T cell response.

The preclinical demonstration of anamnestic protection is of high significance.[1] It suggests that PF-08046037 does more than transiently stimulate the immune system; it appears capable of inducing a robust adaptive immune response leading to long-term immunological memory. This is a cornerstone of successful cancer immunotherapies. If this capacity for generating lasting immunity translates from preclinical models to human patients, PF-08046037 could offer more than temporary tumor shrinkage; it could fundamentally alter the patient's long-term immune surveillance against their cancer.

The diverse array of pharmacodynamic effects observed in vivo—including acute tumor inflammation, reduction of CD206+ suppressive macrophages, and enhanced activation of APCs in both tumors and tdLNs [1]—strongly indicates that the anti-tumor efficacy of PF-08046037 is driven by its ability to comprehensively reprogram the TME. This holistic approach to TME modulation, by "rewiring" the local immune landscape, is hypothesized to be more potent and capable of overcoming multiple resistance mechanisms than therapies with a narrower mode of action.

The use of syngeneic mouse tumor models genetically engineered to express human PD-L1 (hPD-L-1.CT26 and hPD-L-1.MC38) [1] substantially enhances the translational relevance of these preclinical findings. This approach provides a more accurate bridge to human clinical trials compared to studies relying solely on murine surrogate antibodies or targets. Using human PD-L1 knock-in mice ensures that the antibody component of PF-08046037 engages its human target with clinically relevant affinity and specificity. This mitigates a common pitfall in drug development where promising results with surrogate molecules in mice do not translate to humans due to species-specific differences. The positive data from these more sophisticated models thus lend greater credence to the potential clinical utility of PF-08046037.

The following table summarizes significant preclinical findings for PF-08046037:

Table 2: Summary of Significant Preclinical Findings for PF-08046037

Finding CategorySpecific ObservationSource Snippet(s)Significance/Implication for Clinical Development
In Vitro Cellular ActivityPotent and selective activation of TLR7 reporter cells.1Confirms the fundamental biological activity of the released TLR7 agonist payload on its intended molecular target.
In Vitro Cellular ActivityPotent and selective activation of PD-L1-expressing human macrophages & dendritic cells.1Validates the ISAC's ability to target and activate primary human APCs, the intended target cells in patients, in a PD-L1 dependent manner.
In Vivo Anti-Tumor EfficacySingle-agent efficacy in syngeneic hPD-L-1.CT26 & hPD-L-1.MC38 tumor models.1Suggests potential for monotherapy anti-tumor activity in humans; use of human PD-L1 models increases translational relevance.
In Vivo Immunological MemoryProvides anamnestic protection against tumor rechallenge in preclinical models.1Indicates the potential for inducing durable immune responses and long-term immunological memory, a key goal for cancer immunotherapies.
In Vivo Pharmacodynamic Effects - TME ModulationInduction of acute tumor inflammation.1Provides evidence of an active immune response being mounted within the tumor site.
In Vivo Pharmacodynamic Effects - TME ModulationLoss of CD206+ suppressive macrophages.1Suggests reversal of M2 macrophage-mediated immunosuppression and repolarization towards an M1-like (immunostimulatory) macrophage phenotype.
In Vivo Pharmacodynamic Effects - APC ActivationUpregulation of activation & co-stimulatory molecules on APCs in tumors & tdLNs.1Shows enhanced antigen presentation capacity and APC maturation in both the tumor and critical immune priming sites (tdLNs).
In Vivo Pharmacodynamic Effects - T Cell ActivityIncreased activity of cytotoxic T cells.1Links the ISAC-induced APC activation and TME modulation to an enhanced effector T cell response, crucial for tumor cell killing.

4. Clinical Development Program: Focus on NCT06974734

Following promising preclinical evaluation, PF-08046037 has transitioned into early-stage clinical development to assess its safety, tolerability, and preliminary efficacy in human subjects.

4.1. Current Clinical Phase and Overall Strategy

PF-08046037 is currently being investigated in a Phase 1 clinical trial.[2] The primary first-in-human (FIH) study, registered under the identifier NCT06974734, is designed to evaluate PF-08046037 both as a monotherapy and, significantly, in combination with sasanlimab (PF-06801591), Pfizer's investigational anti-PD-1 monoclonal antibody.[9] A Phase 1 trial is the standard initial step for introducing an investigational drug into humans. Its core objectives typically include evaluating safety, tolerability, and pharmacokinetic profiles, and identifying a dose and schedule suitable for further development (Phase 2). The early incorporation of a combination arm with a PD-1 inhibitor (sasanlimab) reflects a common and often accelerated strategy in contemporary immuno-oncology development, driven by a strong mechanistic rationale for potential synergy between the two agents—TLR7-mediated APC activation enhancing the efficacy of PD-1 blockade.

4.2. In-Depth Analysis of Clinical Trial NCT06974734

The clinical trial NCT06974734 is central to the current development phase of PF-08046037.

  • Official Title: "AN OPEN-LABEL PHASE 1 STUDY TO EVALUATE PF-08046037 AS MONOTHERAPY AND PART OF COMBINATION THERAPY IN PARTICIPANTS WITH ADVANCED MALIGNANCIES".[2]
  • Identifiers: ClinicalTrials.gov Identifier: NCT06974734; EU Clinical Trials Information System (CTIS) Number: 2025-521499-69-00; Pfizer Study Protocol Number: C5941001.[8]
  • Sponsor: Pfizer Inc..[8]
  • Study Design: This is a Phase 1, multicenter, open-label, non-randomized study that employs a sequential assignment, multi-part design.[8] The study is structured into several distinct parts to systematically evaluate the investigational agent:
  • Part 1a: PF-08046037 Monotherapy Dose Escalation.
  • Part 2a: PF-08046037 Monotherapy Dose Optimization.
  • Part 3a: PF-08046037 Monotherapy Dose Expansion.
  • Part 1b: PF-08046037 in Combination with Sasanlimab - Dose Escalation (primarily for safety evaluation of the combination).
  • Part 2b: PF-08046037 in Combination with Sasanlimab - Dose Optimization.
  • Part 3b: PF-08046037 in Combination with Sasanlimab - Dose Expansion. This adaptive, multi-part design is characteristic of modern early-phase oncology trials. It allows for the efficient determination of the Maximum Tolerated Dose (MTD) and/or Recommended Phase 2 Dose (RP2D) for both monotherapy and combination therapy. Dose escalation cohorts typically enroll a small number of patients per dose level, while dose expansion cohorts enroll larger numbers at the selected dose(s) to gather more robust safety data and preliminary signals of efficacy in specific tumor types or patient populations.
  • Primary Objectives: While the primary purpose is broadly stated as "Treatment" [8], for a Phase 1 dose-escalation and optimization study, the primary objectives inherently focus on:
  • Assessing the safety and tolerability profile of PF-08046037 administered as monotherapy and when administered in combination with sasanlimab.
  • Identifying any dose-limiting toxicities (DLTs) associated with PF-08046037 monotherapy and the combination regimen.
  • Determining the MTD and/or the RP2D for PF-08046037 as a monotherapy and in combination with sasanlimab. (These are standard primary objectives for Phase 1 oncology trials; specific outcome measures for NCT06974734 are not fully detailed in the provided materials [8], although examples of typical oncology trial outcomes can be seen in unrelated trial descriptions [12]).
  • Secondary Objectives: These are anticipated to include:
  • Characterization of the pharmacokinetic (PK) profile of PF-08046037 (and of sasanlimab when used in combination arms).
  • Evaluation of preliminary anti-tumor activity, likely including measures such as Objective Response Rate (ORR) based on Response Evaluation Criteria in Solid Tumors version 1.1 (RECIST v1.1), Duration of Response (DOR), and Progression-Free Survival (PFS). The eligibility criterion of measurable disease per RECIST v1.1 supports this.[8]
  • Assessment of pharmacodynamic (PD) markers of immune activation in peripheral blood and tumor tissue samples to confirm the mechanism of action in human subjects and explore potential biomarkers of response.
  • Target Patient Populations and Key Eligibility Criteria:
  • Estimated Total Enrollment: Approximately 399 participants are planned to be enrolled across all parts of the study.[8]
  • Age: Eligible participants must be adults, aged 18 years and older.[11]
  • General Inclusion Criteria: Patients must have a diagnosis of advanced or metastatic malignancies, be willing and able to provide tumor tissue samples (which are crucial for correlative scientific studies), and have measurable disease as defined by RECIST v1.1 criteria.[8]
  • Specific Tumor Types and Prior Therapy Requirements by Cohort [8]:
  • Monotherapy Dose Escalation (Part 1a) & Optimization (Part 2a): Patients with advanced or metastatic Non-Small Cell Lung Cancer (NSCLC), Head and Neck Squamous Cell Carcinoma (HNSCC), Melanoma, or Pancreatic Ductal Adenocarcinoma (PDAC). These patients must have experienced disease progression following at least one prior line of approved systemic therapy for their malignancy.
  • Monotherapy Dose Expansion (Part 3a): Patients with advanced or metastatic NSCLC or PDAC.
  • Combination Safety Evaluation (Part 1b) & Dose Optimization (Part 2b): Patients with advanced or metastatic NSCLC or HNSCC. These patients may either be immunotherapy-naïve for their specific tumor type OR may have relapsed or become refractory after receiving prior immunotherapy.
  • Combination Dose Expansion (Part 3b): Patients with unresectable locally advanced or metastatic HNSCC or NSCLC. Critically, these patients must be in the first-line treatment setting (i.e., they must not have received prior systemic cytotoxic therapy for their locally advanced or metastatic disease) AND they must be naïve to any prior immunotherapy. Furthermore, for NSCLC patients to be eligible for this cohort, their tumors must exhibit PD-L1 expression with a Tumor Proportion Score (TPS) of ≥50%; for HNSCC patients, their tumors must demonstrate PD-L1 expression with a Combined Positive Score (CPS) of ≥1.
  • Tumor Tissue Requirements: Specific requirements for the provision of archival or de novo (fresh) tumor biopsies vary depending on the study part. For instance, Part 1a at lower dose levels may accept archival tissue collected within 12 months of enrollment, whereas higher dose levels in Part 1a, as well as Parts 2a, 3a, 1b, 2b, and 3b, may necessitate de novo baseline biopsies or more recently collected archival tissue. Mandatory on-treatment tumor biopsies are also stipulated for certain study parts to assess pharmacodynamic effects.[8]
  • Key Exclusion Criteria [8]: These include, but are not limited to, a history of Grade ≥3 immune-mediated adverse events related to prior immune modulatory therapy that required immunosuppressive treatment; active or prior autoimmune disease that might deteriorate when receiving an immunostimulatory agent; a history of uveitis within the preceding 6 months; clinically significant Grade ≥3 neurodegenerative disease; Grade 3 or higher pulmonary disease unrelated to the underlying malignancy; and previous exposure to an investigational immunostimulatory antibody conjugate or a systemic TLR agonist.
  • Intervention Details:
  • PF-08046037: Administered as an intravenous (IV) infusion.[8]
  • Sasanlimab (PF-06801591): Administered as a subcutaneous (SQ) injection.[9] Sasanlimab is an anti-PD-1 monoclonal antibody.[9]
  • Dosing Schedule: When used in combination, both PF-08046037 and sasanlimab are administered once every 3 weeks (Q3W).[8]
  • Treatment Duration: Participants will continue to receive the study drug(s) until their cancer is no longer responding to treatment (disease progression) or if they experience unacceptable toxicity.[8]
  • Current Status and Key Timelines:
  • Study Status: As of May 2025, the trial was listed as "Not yet recruiting".[2]
  • Planned Study Start Date: May 6, 2025.[11]
  • Estimated Primary Completion Date: February 29, 2028.[14]
  • Estimated Study Completion Date: Not explicitly stated in the provided materials, but will occur after the primary completion date.
  • Known Study Locations:
  • One initial site identified is START Midwest, located in Grand Rapids, Michigan, United States.[11] As a multicenter trial, additional sites in the US and potentially other countries are anticipated.

The inclusion of combination arms with sasanlimab (a PD-1 inhibitor) from Part 1b onwards in this first-in-human trial [8] signifies a robust hypothesis for synergistic activity and an ambition for accelerated development of the combination regimen. While Phase 1 trials traditionally prioritize monotherapy safety, introducing combination arms relatively early, as seen here, suggests high confidence in the mechanistic rationale for synergy—specifically, that TLR7-mediated APC activation by PF-08046037 will enhance the efficacy of PD-1 blockade by sasanlimab. This concurrent evaluation can potentially shorten overall development timelines if the combination proves safe and demonstrates promising activity early on. Pfizer's strategy to combine PF-08046037 with its proprietary PD-1 asset, sasanlimab [8], also allows for greater control over the combination's development pathway.

The requirement for specific PD-L1 expression levels (TPS ≥50% for NSCLC, CPS ≥1 for HNSCC) in the first-line combination expansion cohort (Part 3b) [8] indicates an early intent to identify a targeted patient population where the combination might yield the most substantial benefit. PD-L1 is an established biomarker for predicting response to PD-1/PD-L1 inhibitors. Applying this criterion to a first-line cohort for an ISAC plus PD-1 combination suggests an effort to enrich the study population for potential responders. This could be predicated on preclinical data suggesting that PD-L1 expression correlates with ISAC efficacy or synergy, or it may be a pragmatic approach to align with existing indications for PD-1 inhibitors. This biomarker-driven selection in later cohorts contrasts with the broader eligibility in earlier dose-finding cohorts (e.g., Parts 1a, 2a, 1b, 2b), which reflects their exploratory nature. Such an approach could streamline the design of subsequent, larger-phase trials.

The mandatory tumor tissue requirements, including archival samples, de novo baseline biopsies, and on-treatment biopsies across various trial parts [8], underscore a significant investment in translational research. These biological samples are vital for understanding the mechanism of action of PF-08046037 in humans, identifying potential predictive biomarkers of response or resistance, and characterizing the pharmacodynamic effects of the drug on the tumor microenvironment and systemic immunity. Biopsies allow for direct assessment of changes within the TME, such as immune cell infiltration patterns, gene expression profiles, and protein levels. This is crucial for validating the preclinical MoA (e.g., APC activation, M2 macrophage reduction) in patients. The early identification of predictive biomarkers can de-risk later stages of development and optimize patient selection strategies. This is particularly important for a novel ISAC like PF-08046037, where the full spectrum of human biological effects is yet to be comprehensively characterized.

The trial's design to enroll patients with several types of advanced cancers—NSCLC, HNSCC, Melanoma, and PDAC [8]—many of whom have progressed on prior therapies, reflects an exploratory approach. This breadth allows for the investigation of PF-08046037's activity across different histological and immunological contexts. These tumor types exhibit varying baseline levels of PD-L1 expression and diverse immune infiltration patterns, offering a range of environments in which to test the ISAC's potential. The inclusion of patients with prior disease progression, including on immunotherapy for some cohorts, directly addresses significant unmet medical needs. Positive signals of activity in any of these initial tumor types could lead to the design of focused expansion cohorts or inform the selection of indications for subsequent Phase 2 trials.

The following table provides a structured overview of the multi-part NCT06974734 trial:

Table 3: Detailed Structure of NCT06974734: Cohorts, Interventions, and Key Objectives/Populations

Trial PartIntervention(s)Key Objective(s)Target Tumor TypesKey Population Characteristics
Part 1aPF-08046037 MonotherapyDose Escalation, MTD/RP2D Determination, SafetyNSCLC, HNSCC, Melanoma, PDACAdvanced/metastatic, progressed on ≥1 prior systemic therapy.
Part 2aPF-08046037 MonotherapyDose Optimization, Safety, Preliminary EfficacyNSCLC, HNSCC, Melanoma, PDACAdvanced/metastatic, progressed on ≥1 prior systemic therapy.
Part 3aPF-08046037 MonotherapyMonotherapy Expansion, Safety, EfficacyNSCLC, PDACAdvanced/metastatic.
Part 1bPF-08046037 + SasanlimabCombination Dose Escalation, Safety, MTD/RP2DNSCLC, HNSCCAdvanced/metastatic; may be immunotherapy-naïve OR relapsed/refractory to prior immunotherapy.
Part 2bPF-08046037 + SasanlimabCombination Dose Optimization, Safety, EfficacyNSCLC, HNSCCAdvanced/metastatic; may be immunotherapy-naïve OR relapsed/refractory to prior immunotherapy.
Part 3bPF-08046037 + SasanlimabCombination Expansion (First-Line)NSCLC, HNSCCUnresectable locally advanced/metastatic; No prior systemic cytotoxic therapy in advanced setting; Immunotherapy-naïve; NSCLC: PD-L1 TPS ≥50%; HNSCC: PD-L1 CPS ≥1.

5. Strategic Considerations and Future Outlook

The development of PF-08046037 occurs within a dynamic and competitive landscape of oncology therapeutics, particularly in immuno-oncology and antibody-drug conjugates.

5.1. Positioning within Pfizer's Immuno-Oncology and ADC/ISAC Portfolio

PF-08046037, likely a legacy asset from Seagen's innovative pipeline [4], is a component of Pfizer's significantly expanded focus on ADCs and novel immunotherapeutics following the acquisition of Seagen.[15] This ISAC complements Pfizer's other PD-L1 targeted ADC, PF-08046054 (also known as SGN-PDL1V), which notably carries a cytotoxic monomethyl auristatin E (MMAE) payload.[4] The concurrent development of these two agents, both targeting PD-L1 but employing distinct payload mechanisms (immune stimulation via TLR7 agonism versus direct cytotoxicity via MMAE), suggests a sophisticated and comprehensive strategy. This dual approach could allow Pfizer to address diverse tumor biologies or patterns of resistance to other therapies. For instance, an ISAC like PF-08046037 might be more suited for immunologically "cold" tumors requiring immune activation, whereas a cytotoxic ADC like PF-08046054 might be effective in "hot" tumors that have developed resistance to checkpoint inhibitors but remain targetable for direct cell killing. This represents a significant investment in maximizing the therapeutic utility of the PD-L1 target beyond standard checkpoint blockade. Furthermore, the planned combination of PF-08046037 with Pfizer's own PD-1 inhibitor, sasanlimab [8], allows for the controlled development of proprietary combination regimens, potentially offering strategic advantages in terms of clinical development and market positioning.

5.2. Differentiation from other PD-L1 Targeting Agents and ISACs

PF-08046037 is designed to offer a distinct mechanism of action compared to standard anti-PD-L1 antibodies. While conventional anti-PD-L1 mAbs primarily function by blocking the PD-1/PD-L1 inhibitory checkpoint, PF-08046037 utilizes the PD-L1 molecule predominantly as a "docking site" or "molecular address" to deliver its immune-activating TLR7 agonist payload directly to PD-L1-expressing APCs within the TME.[1] The specific combination of an anti-PD-L1 antibody, an imidazoquinoline-based TLR7 agonist, and a mannosidase-cleavable linker constitutes a unique molecular construct. Other ISACs currently in development may target different antigens or employ alternative immune-stimulating payloads (e.g., STING agonists, other TLR agonists like TLR8 agonists, or cytokine mimetics) or different linker-payload chemistries.[6] The ultimate clinical success of PF-08046037 will depend on the robust clinical validation of this specific tripartite approach, demonstrating a favorable efficacy and safety profile.

5.3. Potential Therapeutic Niches and Unmet Needs Addressed

PF-08046037 is being investigated for its potential to address several unmet medical needs in oncology:

  • Patients with advanced malignancies who have experienced disease progression on, or are refractory to, standard-of-care therapies, including existing checkpoint inhibitors. The eligibility criteria for certain cohorts in the NCT06974734 trial specifically include such patient populations.[8]
  • Tumors characterized by an immunosuppressive TME, particularly those with dysfunctional or poorly activated APCs, or those with low T-cell infiltration (often termed "cold" tumors). In such scenarios, the in-situ immune activation mediated by PF-08046037 could be particularly beneficial by converting these non-immunogenic tumors into ones that can be recognized and attacked by the immune system.
  • There is also potential for PF-08046037 to be used in the first-line treatment setting, in combination with PD-1 inhibitors like sasanlimab, for biomarker-selected patient populations. This is being explored in Part 3b of the NCT06974734 trial for PD-L1 high NSCLC and HNSCC.[8]

5.4. Anticipated Challenges and Opportunities

The clinical development of PF-08046037, while promising, is not without its challenges and opportunities:

  • Challenges:
  • Managing Immune-Related Adverse Events (irAEs): A primary challenge will be the careful management of potential irAEs and cytokine release syndrome (CRS) that can be associated with TLR7 agonism, even with a targeted delivery approach.[6] The preclinical observation of "acute tumor inflammation" [1] warrants vigilant clinical monitoring for both local and systemic inflammatory responses. While the sophisticated design of PF-08046037 (Fc mutations, cleavable linker, low permeability payload) aims to widen the therapeutic window [1], the historical difficulty in balancing efficacy and toxicity with immune-stimulating agents, including TLR agonists [6], remains a key consideration. The Phase 1 trial's primary focus on safety and DLTs [8] will be critical in determining if this balance has been successfully achieved in humans.
  • Biomarker Identification: Identifying optimal patient populations and robust predictive biomarkers beyond PD-L1 expression will be crucial for maximizing the benefit-risk ratio and guiding future development.
  • Immunogenicity: As with all biologic therapies, there is a potential for the development of anti-drug antibodies (ADAs) against the antibody component or the linker-payload complex of PF-08046037, which could impact efficacy or safety.[6]
  • Competitive Landscape: The immuno-oncology field is highly competitive, with numerous novel therapies and other ADC/ISAC approaches in development.[15] PF-08046037 will need to demonstrate a clear clinical advantage.
  • Opportunities:
  • Converting "Cold" Tumors: A significant opportunity lies in PF-08046037's potential to convert immunologically "cold" tumors (those lacking significant T-cell infiltration or with a highly suppressive TME) into "hot" tumors that are more responsive to immunotherapy.
  • Synergistic Combinations: The strong mechanistic rationale for synergy with PD-1/PD-L1 inhibitors (like sasanlimab) and potentially other cancer therapies (e.g., chemotherapy, targeted agents) offers broad opportunities for combination strategies.
  • Novel Mechanism of Action: The development of a novel, first-in-class ISAC with a distinct MoA provides an opportunity to overcome resistance to existing therapies.
  • Durable Responses: The robust preclinical data, particularly the induction of anamnestic protection [1], offers the prospect of achieving durable clinical responses and long-term disease control in patients.

The immuno-oncology landscape is indeed crowded. PF-08046037's differentiation lies in its specific ISAC mechanism, targeting PD-L1 for delivery of a TLR7 agonist. However, it will face competition from a multitude of other novel IO agents, cell therapies, and alternative ADC/ISAC strategies. Its clinical data will need to demonstrate a clear advantage in terms of efficacy, safety, or applicability to specific, well-defined patient populations to successfully carve out a therapeutic niche. Demonstrating benefit in refractory settings or through unique synergistic combinations will be key for the future trajectory of PF-08046037.

6. Conclusion

PF-08046037 (formerly SGN-PDL1iT) emerges as a highly innovative investigational agent, classified as a PD-L1 targeted Toll-like Receptor 7 (TLR7) agonist Immune-Stimulating Antibody Conjugate (ISAC). Its sophisticated molecular design, featuring an anti-PD-L1 antibody with engineered Fc mutations, an imidazoquinoline-based TLR7 agonist payload with low membrane permeability, and a mannosidase-cleavable linker, is aimed at achieving highly localized immune activation within the tumor microenvironment.[1] This strategy seeks to harness the potent immunostimulatory effects of TLR7 agonism while minimizing systemic toxicities often associated with untargeted immune activators.

Preclinical studies have provided a strong rationale for its clinical development. In vitro, PF-08046037 has demonstrated potent and selective activation of TLR7 reporter cells and, more importantly, PD-L1-expressing primary human macrophages and dendritic cells.[1] In vivo, in syngeneic mouse models engineered to express human PD-L1, PF-08046037 has shown single-agent anti-tumor efficacy and, remarkably, the ability to induce anamnestic protection against tumor rechallenge. These efficacy findings are supported by pharmacodynamic data indicating acute tumor inflammation, a favorable repolarization of tumor-associated macrophages, enhanced activation of antigen-presenting cells in both tumors and tumor-draining lymph nodes, and increased cytotoxic T cell activity.[1] These preclinical results collectively suggest a potent ability to remodel the tumor microenvironment and generate durable anti-tumor immunity.

Currently, PF-08046037 is advancing through Phase 1 clinical development with the NCT06974734 trial. This multi-part study is designed to evaluate its safety, tolerability, pharmacokinetics, and preliminary efficacy both as a monotherapy and in combination with Pfizer's anti-PD-1 antibody, sasanlimab, across a range of advanced malignancies including NSCLC, HNSCC, melanoma, and PDAC.[8] The trial's design incorporates dose escalation, optimization, and expansion cohorts, with specific attention to biomarker assessment (e.g., PD-L1 expression in certain cohorts) and intensive translational research through mandatory tissue sampling.[8]

Strategically, PF-08046037 represents a key asset in Pfizer's expanded oncology pipeline following the acquisition of Seagen, reflecting a commitment to pioneering ADC and ISAC technologies.[4] It offers a differentiated approach to PD-L1 targeting, moving beyond simple checkpoint blockade to active immune stimulation. The potential to convert immunologically "cold" tumors, overcome resistance to existing therapies, and achieve durable responses presents significant opportunities. However, challenges related to managing immune-related adverse events, identifying optimal patient populations, and navigating a competitive therapeutic landscape must be addressed.

In summary, PF-08046037 is a promising, mechanistically novel ISAC with a strong preclinical foundation. Its journey from a preclinical candidate to a clinically impactful therapy will depend on the successful validation of its efficacy and safety profile in ongoing and future human trials. The ability to demonstrate a clear clinical benefit, particularly in well-defined patient populations and potentially in combination regimens, will determine its ultimate role in the evolving paradigm of cancer immunotherapy. The initial data from the Phase 1 NCT06974734 trial will be critical in shaping its future development pathway and realizing its potential to address unmet needs in patients with advanced cancers.

Works cited

  1. Abstract 5840: PF-08046037, a PD-L1 targeted TLR7 agonist ISAC, drives innate immune activation and anti-tumor efficacy in preclinical in vitro and in vivo models - AACR Journals, accessed May 30, 2025, https://aacrjournals.org/cancerres/article/85/8_Supplement_1/5840/759151/Abstract-5840-PF-08046037-a-PD-L1-targeted-TLR7?searchresult=1
  2. PF-08046037 - Drug Targets, Indications, Patents - Patsnap Synapse, accessed May 30, 2025, https://synapse.patsnap.com/drug/afe931473a664884bf5cef0ce107afae
  3. Inflammatory Tumor Research Articles - R Discovery, accessed May 30, 2025, https://discovery.researcher.life/topic/inflammatory-tumour/3696222?page=2
  4. Pfizer makes haste to catch up in PD-L1 | ApexOnco - Oncology Pipeline, accessed May 30, 2025, https://www.oncologypipeline.com/apexonco/pfizer-makes-haste-catch-pd-l1
  5. Pfizer doubles down on PD-L1 conjugates | ApexOnco - Oncology Pipeline, accessed May 30, 2025, https://www.oncologypipeline.com/apexonco/pfizer-doubles-down-pd-l1-conjugates
  6. Innovative payloads for ADCs in cancer treatment: moving beyond the selective delivery of chemotherapy - ResearchGate, accessed May 30, 2025, https://www.researchgate.net/publication/387665554_Innovative_payloads_for_ADCs_in_cancer_treatment_moving_beyond_the_selective_delivery_of_chemotherapy
  7. Innovative payloads for ADCs in cancer treatment: moving beyond the selective delivery of chemotherapy - PMC, accessed May 30, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC11694294/
  8. A Clinical Trial of PF-08046037 Alone or With Sasanlimab in Patients With Advanced or Metastatic Malignancies - ClinicalTrials.Veeva, accessed May 30, 2025, https://ctv.veeva.com/study/a-clinical-trial-of-pf-08046037-alone-or-with-sasanlimab-in-patients-with-advanced-or-metastatic-mal
  9. Sasanlimab - Drug Targets, Indications, Patents - Patsnap Synapse, accessed May 30, 2025, https://synapse.patsnap.com/drug/70d140b74aca4e5caceb3dda28a96e49
  10. Clinical Digest: A Comprehensive Review of Recent Pfizer Clinical Trials, accessed May 30, 2025, https://resources.clinicaldigest.org/2025/05/20/clinical-digest-a-comprehensive-review-of-recent-pfizer-clinical-trials/
  11. A Clinical Trial of PF-08046037 Alone or With Sasanlimab in Patients With Advanced or Metastatic Malignancies, accessed May 30, 2025, https://www.pfizerclinicaltrials.com/find-a-trial/nct06974734-carcinoma-trial
  12. Select Clinical Trials Currently Enrolling Patients with Leukemia, accessed May 30, 2025, https://oncpracticemanagement.com/issues/2017/july-2017-vol-7-no-7/select-clinical-trials-currently-enrolling-patients-with-leukemia
  13. Select Ongoing Trials Currently Enrolling Patients with Multiple Myeloma, accessed May 30, 2025, https://jons-online.com/issues/2016/may-2016-vol-7-no-4/select-ongoing-trials-currently-enrolling-patients-with-multiple-myeloma
  14. A Clinical Trial of PF-08046037 Alone or With Sasanlimab in Patients With Advanced or Metastatic Malignancies - LarvolClin, accessed May 30, 2025, https://clin.larvol.com/trial-detail/NCT06974734
  15. Pfizer Pflash - Antibody Drug Conjugates (ADCs), accessed May 30, 2025, https://s28.q4cdn.com/781576035/files/doc_events/2025/Feb/27/Pfizer-Pflash_ADCs-Presentation_FINAL.pdf
  16. Pfizer Advances Bold Vision for Future of Cancer Care at the ASCO 2025 Annual Meeting, accessed May 30, 2025, https://www.pfizer.com/news/press-release/press-release-detail/pfizer-advances-bold-vision-future-cancer-care-asco-2025

Published at: May 30, 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.