MedPath

Budigalimab Advanced Drug Monograph

Published:Sep 2, 2025

Generic Name

Budigalimab

Drug Type

Biotech

CAS Number

2098225-93-3

Budigalimab (ABBV-181): A Comprehensive Clinical, Pharmacological, and Strategic Analysis

Executive Summary

Budigalimab (ABBV-181) is an investigational, humanized immunoglobulin G1 (IgG1) monoclonal antibody developed by AbbVie, targeting the programmed cell death 1 (PD-1) receptor. As an immune checkpoint inhibitor, its fundamental mechanism of action aligns with a well-established and transformative class of oncology therapeutics. It functions by blocking the interaction between PD-1 on T-cells and its ligands, primarily PD-L1, thereby preventing tumor-induced immune suppression and reactivating a cytotoxic T-cell response against malignant cells. A distinguishing feature of Budigalimab's molecular architecture is its IgG1 backbone, which has been specifically engineered with point mutations (L234A, L235A) in the Fc region to abrogate effector functions, such as antibody-dependent cell-mediated cytotoxicity. This design choice suggests a strategic focus on creating a highly specific and immunologically inert checkpoint blockade agent, optimized for safety and predictability in combination regimens.

The clinical development program for Budigalimab is extensive, encompassing numerous Phase 1 and Phase 2 trials across a wide spectrum of solid tumors. Data from monotherapy expansion cohorts in non-small cell lung cancer (NSCLC) and head and neck squamous cell carcinoma (HNSCC) have demonstrated modest objective response rates of 19% and 13%, respectively.[1] These efficacy signals, while confirming biological activity, are not superior to those of established anti-PD-1 agents. Consequently, the core of AbbVie's strategy for Budigalimab is not as a standalone therapy but as a foundational immunotherapy backbone for a diverse pipeline of proprietary combination agents. It is being systematically evaluated with novel antibody-drug conjugates (ADCs), other monoclonal antibodies, and standard chemotherapy across indications such as gastric cancer, urothelial carcinoma, and small cell lung cancer (SCLC).[3] This positions Budigalimab as a critical internal asset intended to unlock the full potential of AbbVie's next-generation oncology portfolio.

Pharmacologically, Budigalimab exhibits a favorable and predictable profile. Its pharmacokinetics are approximately dose-proportional, and early clinical data facilitated a rapid transition to convenient flat-dosing regimens, leveraging established knowledge from the therapeutic class.[7] Pharmacodynamic studies confirm rapid, complete, and sustained saturation of the PD-1 receptor even at low doses, alongside downstream biomarker evidence of immune activation.[7] The safety profile of Budigalimab is manageable and consistent with the known toxicities of PD-1 inhibitors, characterized primarily by immune-related adverse events (irAEs) such as thyroid dysfunction, pruritus, and rash.[7] No novel or unexpected safety signals have been identified, a crucial attribute for a drug intended for widespread use in combination therapies.

In a novel exploratory venture, Budigalimab is also being investigated for the treatment of Human Immunodeficiency Virus (HIV), with early data suggesting it may delay viral rebound following the interruption of antiretroviral therapy.[10] While this represents a potential area of differentiation, it remains a high-risk, early-stage program. The overarching strategic outlook for Budigalimab is that of an enabling platform rather than a direct market competitor to established agents like pembrolizumab and nivolumab. Its primary value to AbbVie lies in providing strategic independence, controlling the cost and development of combination trials, and ultimately creating proprietary, patent-protected regimens that can define new standards of care in oncology.

Introduction: Budigalimab in the Context of Immune Checkpoint Inhibition

The advent of immuno-oncology has fundamentally reshaped the landscape of cancer treatment over the past decade, shifting the therapeutic paradigm from direct cytotoxic or targeted attacks on tumor cells to the potentiation of the host's own immune system. Central to this revolution is the understanding and therapeutic manipulation of immune checkpoints—a network of regulatory pathways that maintain self-tolerance and modulate the duration and amplitude of physiological immune responses.

The PD-1/PD-L1 Axis as a Cornerstone of Immuno-Oncology

Among the most critical of these pathways is the axis formed by the Programmed cell death 1 (PD-1) receptor (also known as PDCD1 or CD279) and its ligands, Programmed death-ligand 1 (PD-L1) and Programmed death-ligand 2 (PD-L2).[1] PD-1 is a transmembrane protein expressed on the surface of activated T-cells, B-cells, and natural killer (NK) cells.[11] When PD-1 engages with its ligands, which are expressed on various cell types including antigen-presenting cells and, crucially, many types of tumor cells, it transmits an inhibitory signal into the immune cell.[7] This signal dampens T-cell receptor signaling, curtails cytokine production, and ultimately suppresses the cytotoxic activity of T-cells.[12]

While this mechanism is vital for preventing autoimmunity under normal physiological conditions, many cancers have co-opted this pathway as a primary mechanism of immune evasion.[7] By upregulating the expression of PD-L1 on their surface, tumor cells can effectively engage PD-1 on tumor-infiltrating lymphocytes, creating an immunosuppressive tumor microenvironment and functionally "switching off" the anti-tumor immune response.[7] The development of monoclonal antibodies designed to block this interaction represents one of the most significant therapeutic breakthroughs in modern medicine. By physically preventing the binding of PD-1 to PD-L1, these agents release the "brakes" on the immune system, restoring the ability of cytotoxic T-lymphocytes to recognize and eliminate cancer cells.[12] The clinical success of the first-generation PD-1 inhibitors, such as nivolumab and pembrolizumab, has been profound, leading to durable responses and improved survival across a multitude of malignancies, including melanoma, non-small cell lung cancer (NSCLC), and renal cell carcinoma.[1]

Rationale for Development

It is within this established and highly competitive therapeutic landscape that AbbVie is developing Budigalimab (ABBV-181), a monoclonal antibody directed against the PD-1 receptor.[11] The development of a new anti-PD-1 agent in a market with several entrenched, blockbuster incumbents immediately raises questions regarding its strategic rationale and potential for differentiation. The primary mechanism of action—blocking the PD-1/PD-L1 interaction to restore T-cell function—is not novel. However, the rationale for Budigalimab's development appears to be multifaceted, extending beyond the pursuit of a standalone therapy. It is rooted in a combination of specific molecular engineering choices and, more significantly, a broad strategic vision for its role within AbbVie's burgeoning oncology pipeline. Budigalimab is being positioned not merely as another entrant into the PD-1 inhibitor class, but as a foundational, internally controlled immunotherapy backbone upon which a new generation of proprietary combination therapies can be built.

Molecular Architecture and Mechanism of Action

The therapeutic activity and strategic utility of Budigalimab are direct consequences of its specific molecular design, which incorporates features common to its class while also including deliberate engineering choices to refine its biological function.

Drug Identification and Physicochemical Properties

Budigalimab is a biologic drug classified as a humanized, recombinant monoclonal antibody.[14] Its fundamental identifiers and properties are summarized in Table 1, providing a clear reference for its molecular identity.

Table 1: Key Identifiers and Physicochemical Properties of Budigalimab

PropertyDescriptionSource(s)
International Nonproprietary NameBudigalimab11
Synonyms / Code NamesABBV-181, abbv 181, abbv-181, PR 16488171
DrugBank IDDB16668
CAS Number2098225-93-316
Drug TypeBiotech, Monoclonal Antibody14
Molecular WeightApproximately 144.76–145.84 kDa16
IsotypeHumanized, recombinant Immunoglobulin G1 (IgG1), kappa light chain1
Source / HostChinese Hamster Ovary (CHO) cells17
Fc Region MutationsL234A, L235A1
Molecular TargetProgrammed cell death 1 receptor (PD-1, PDCD1, CD279)11
AppearanceColorless to light yellow liquid16

Structural Design and Engineering

The structure of Budigalimab has been carefully selected and modified to optimize its function as a specific and safe immune checkpoint inhibitor.

Humanized IgG1 Isotype

Budigalimab is built on a humanized IgG1 scaffold.[1] This distinguishes it from the two most prominent approved PD-1 inhibitors, nivolumab and pembrolizumab, which are both of the IgG4 subclass.[1] The IgG1 isotype is naturally the most abundant immunoglobulin in human serum and is typically associated with potent effector functions, including Antibody-Dependent Cell-Mediated Cytotoxicity (ADCC) and Complement-Dependent Cytotoxicity (CDC). In the context of an anti-PD-1 antibody, these effector functions could theoretically lead to the depletion of the very PD-1-expressing T-cells that the drug is intended to activate, a counterproductive effect. This makes the specific engineering of Budigalimab's Fc region a critical design element.

Fc Region Mutation (L234A, L235A)

To address the potential for unwanted effector activity, Budigalimab incorporates two specific point mutations in the heavy chain of its Fc (Fragment, crystallizable) region: Leucine to Alanine at position 234 (L234A) and Leucine to Alanine at position 235 (L235A).[1] This "LALA" mutation is a well-characterized engineering strategy known to significantly reduce the binding affinity of the Fc region to Fc-gamma receptors (FcγRs) on immune cells like NK cells and macrophages.[1] By disrupting this interaction, the mutations effectively "silence" the antibody's ability to trigger ADCC and other Fc-mediated effector functions.[19]

The decision to use an IgG1 backbone and then engineer it to be functionally silent, rather than simply using a naturally low-effector IgG4 backbone, is a sophisticated choice. This approach may offer advantages in terms of manufacturing, stability, or pharmacokinetic properties compared to an IgG4 isotype, while ensuring that the therapeutic effect is exclusively derived from checkpoint blockade. This design ensures that Budigalimab acts as a pure antagonist, minimizing the potential for complex or unpredictable biological activities, a particularly desirable feature for an agent intended for broad use in combination therapies.

Mechanism of Action

Budigalimab's therapeutic effect is achieved through a precise, multi-step process of immune reactivation.

Target Binding

The primary molecular event is the high-affinity, specific binding of Budigalimab to the human PD-1 receptor, which is expressed on the surface of activated lymphocytes.[11] Preclinical experiments have demonstrated that Budigalimab exhibits a binding affinity for PD-1 that is similar to that of nivolumab and pembrolizumab, confirming its potency at the target level.[1]

Checkpoint Blockade

Upon binding to PD-1, Budigalimab acts as a competitive antagonist, physically obstructing the receptor's interaction with its ligands, PD-L1 and PD-L2.[7] This blockade prevents the delivery of the co-inhibitory signal that would normally be triggered upon ligand engagement, thereby interrupting the downstream signaling cascade that leads to T-cell anergy and exhaustion.[11]

Restoration of Anti-Tumor Immunity

By negating this dominant inhibitory signal, Budigalimab restores the functional capacity of tumor-specific cytotoxic T-lymphocytes (CTLs).[11] The reactivated T-cells can once again recognize tumor antigens presented by cancer cells and execute their effector functions, leading to tumor cell lysis. This immune reactivation is further supported by pharmacodynamic data showing that Budigalimab administration leads to an increase in the expression of multiple cytokines and chemokines, including Interferon-gamma (IFN-γ), CXCL9 (MIG), and CXCL10 (IP-10).[9] These molecules are hallmarks of a Type 1 T-helper (Th1) immune response and are instrumental in the recruitment and activation of additional T-cells and other immune effectors into the tumor microenvironment, further amplifying the anti-tumor response. The drug's mechanism is therefore not to kill cancer cells directly, but to empower the immune system to do so. This universal mechanism of immune potentiation underpins its broad applicability as a combination partner for other anticancer agents that rely on a competent immune system for maximal efficacy.

Comprehensive Pharmacological Profile

The clinical utility of a monoclonal antibody is defined not only by its mechanism but also by its pharmacological behavior—how it is absorbed, distributed, and eliminated by the body (pharmacokinetics), and the time course and intensity of its biological effects (pharmacodynamics). Budigalimab has been characterized by a profile that is both predictable and consistent with the goals of its development program.

Pharmacokinetics (PK)

The pharmacokinetic profile of Budigalimab has been evaluated in Phase 1 studies, revealing characteristics that support convenient and effective clinical administration.

Dose Proportionality

Across a dose range of 1, 3, and 10 mg/kg administered intravenously every two weeks, the pharmacokinetics of Budigalimab were found to be approximately dose-proportional.[7] This means that as the dose is increased, the resulting drug exposure in the body (as measured by metrics like peak concentration and area under the curve) increases in a predictable, linear fashion. This is a highly desirable property, as it simplifies dosing and reduces the risk of unexpected accumulation or underexposure at different dose levels.

Modeling and Transition to Flat Dosing

A key aspect of Budigalimab's clinical pharmacology program has been the strategic and early application of pharmacokinetic and pharmacodynamic (PK/PD) modeling.[7] Data gathered from the initial dose-escalation cohorts, which used traditional body-weight-based dosing (mg/kg), were fed into sophisticated models. These models were used to simulate and predict the exposures that would be achieved with fixed, or "flat," dosing regimens. This modeling validated that flat doses, such as 250 mg every two weeks (Q2W) or 500 mg every four weeks (Q4W), would produce drug exposures and biological effects comparable to the effective weight-based doses.[1]

This model-informed approach allowed for a confident and rapid transition to flat dosing in the expansion phases of the clinical program. This is a significant practical advantage in the clinical setting, as flat dosing increases convenience for prescribers, reduces the potential for calculation errors, streamlines pharmacy preparation, and minimizes drug wastage from partially used vials.[7] This efficient adoption of a market-standard dosing approach, leveraging extensive knowledge from previously approved anti-PD-1 agents, is indicative of a well-executed "fast-follower" strategy designed to accelerate development and ensure practical parity with competitors.

Population Consistency

Importantly, clinical studies demonstrated that the PK/PD profiles and safety of Budigalimab were comparable between Japanese and Western patient populations.[7] This finding suggests that intrinsic ethnic factors do not significantly alter the drug's disposition or effects, obviating the need for region-specific dosing adjustments and simplifying the design and interpretation of global clinical trials.

Pharmacodynamics (PD)

Pharmacodynamic assessments confirm that Budigalimab effectively engages its target and elicits the expected downstream biological responses.

Target Engagement and Receptor Saturation

The primary pharmacodynamic effect of Budigalimab is its ability to bind to and occupy PD-1 receptors on peripheral T-cells. Studies have shown that the drug achieves rapid, complete, and sustained target engagement. Across all tested dose levels, including the lowest dose of 1 mg/kg, Budigalimab produced greater than 99% saturation of peripheral PD-1 receptors.[7] This maximal receptor occupancy was observed as early as two hours after the infusion, indicating that the drug quickly reaches and binds to its target to exert its biological effect.[7] The achievement of full receptor saturation even at low doses, combined with a flat exposure-safety relationship where higher doses do not correlate with increased toxicity, suggests a wide therapeutic window. This provides a substantial margin of safety and dosing flexibility, which is a critical advantage when developing combination therapies with other agents that have their own toxicity profiles.

Biomarker Evidence of Immune Activation

Beyond simple receptor binding, treatment with Budigalimab leads to measurable changes in biomarkers that are indicative of immune system activation. Clinical data show that administration of Budigalimab results in increased circulating levels of interferon-gamma (IFN-γ)-induced chemokines, specifically Monokine Induced by Gamma Interferon (MIG, or CXCL9) and Interferon-gamma-inducible Protein 10 (IP-10, or CXCL10).[1] These chemokines are crucial for trafficking activated T-cells and other immune cells into tissues, including the tumor microenvironment. This increase in pro-inflammatory chemokines serves as direct evidence that Budigalimab is not just blocking a receptor but is successfully initiating the downstream cascade of events required for a productive anti-tumor immune response.

Clinical Development and Efficacy Analysis

The clinical development program for Budigalimab is notable for its breadth and its clear strategic focus on combination therapies. While monotherapy activity has been established, the vast majority of investigational efforts are directed at evaluating Budigalimab as a foundational element in conjunction with other novel anticancer agents.

Overview of Clinical Program

Budigalimab has been investigated in at least 12 clinical trials, the majority of which are Phase 1 or Phase 2 studies.[11] The program has explored a wide range of advanced solid tumors, with a particular focus on non-small cell lung cancer (NSCLC), head and neck squamous cell carcinoma (HNSCC), small cell lung cancer (SCLC), urothelial carcinoma, hepatocellular carcinoma (HCC), and various gastrointestinal cancers.[3] A key feature across these trials is the frequent use of Budigalimab as the constant immunotherapy backbone while testing various combination partners, effectively positioning the program as a screening platform to identify synergistic pairings within AbbVie's broader oncology pipeline. A summary of major clinical trials is presented in Table 2.

Table 2: Summary of Major Clinical Trials Investigating Budigalimab

NCT IdentifierPhaseStatusConditions / IndicationsIntervention(s)
NCT030002571CompletedAdvanced Solid Tumors, NSCLC, SCLC, HNSCCBudigalimab monotherapy; Budigalimab + Rovalpituzumab Tesirine; Budigalimab + Venetoclax
NCT038219351Active, not recruitingLocally Advanced or Metastatic Solid Tumors, HCC, Pancreatic Cancer, MSS-CRCLivmoniplimab (ABBV-151) +/- Budigalimab
NCT055999841ActiveAdvanced Solid Tumors, SCLC, High-Grade CNS Tumors, Neuroendocrine CarcinomasABBV-706 +/- Budigalimab, Cisplatin, or Carboplatin
NCT062364382RecruitingUntreated Metastatic Non-Squamous NSCLCLivmoniplimab + Budigalimab + Chemotherapy vs. Pembrolizumab + Chemotherapy
NCT066283102RecruitingGastric, Gastroesophageal Junction, or Esophageal AdenocarcinomaABBV-400 + Fluorouracil + Leucovorin + Budigalimab
NCT067726231b/2ActiveAdvanced or Metastatic Non-Squamous NSCLCTelisotuzumab Adizutecan (ABBV-400) + Budigalimab
NCT061589581ActiveAdvanced Solid TumorsABBV-303 +/- Budigalimab
NCT050054031ActiveNSCLC, HNSCC, Other Solid TumorsAzirkitug (ABBV-514) +/- Budigalimab or Bevacizumab
NCT038939551Active, not recruitingMetastatic Solid Neoplasm, TNBC, NSCLCABBV-927 + ABBV-368, Budigalimab, and/or Chemotherapy
NCT042238041CompletedHuman Immunodeficiency Virus (HIV)-1Budigalimab

Monotherapy Efficacy in Solid Tumors (NCT03000257)

The first-in-human Phase 1 study, NCT03000257, provided the primary data on Budigalimab's activity as a single agent.[1] The study included dose-escalation and dose-expansion cohorts. The most detailed efficacy results come from the expansion cohorts in patients with HNSCC and NSCLC who were naïve to prior PD-1/PD-L1 inhibitors.[1]

  • Study Design: Patients in these cohorts received Budigalimab intravenously at a flat dose of 250 mg every two weeks (Q2W) or 500 mg every four weeks (Q4W) until disease progression or unacceptable toxicity.[1] The primary endpoints were safety and pharmacokinetics, with efficacy as a secondary endpoint.[1]
  • Efficacy Results: In a cohort of 41 patients with HNSCC, the confirmed objective response rate (ORR) was 13% (90% Confidence Interval [CI], 5.1–24.5), with a median progression-free survival (PFS) of 3.6 months (95% CI, 1.7–4.7).[1] In a cohort of 40 patients with NSCLC, the ORR was 19% (90% CI, 9.2–32.6), with a median PFS of 1.9 months (95% CI, 1.7–3.7).[1]
  • Analysis: These results confirm that Budigalimab has anti-tumor activity as a monotherapy. However, the response rates are modest and do not suggest superiority over existing PD-1 inhibitors in similar, heavily pre-treated patient populations. This reinforces the assessment that Budigalimab's primary strategic value is not as a standalone agent but as a component of more effective combination regimens.

Combination Therapy Strategy and Emerging Efficacy

The core of the Budigalimab clinical program lies in its evaluation as a combination partner. This strategy aims to create synergistic anti-tumor effects by pairing the broad immune activation of PD-1 blockade with agents that have distinct mechanisms of action.

  • With Livmoniplimab (ABBV-151, anti-GARP-TGFβ1): Transforming growth factor-beta (TGF-β1) is a potent immunosuppressive cytokine in the tumor microenvironment. Livmoniplimab targets the GARP-TGFβ1 complex, preventing the release of active TGF-β1.[4] Preclinical models suggested that combining this mechanism with PD-1 inhibition could enhance anti-tumor activity.[4]
  • In the Phase 1 trial NCT03821935, the combination of Budigalimab and Livmoniplimab was found to be well-tolerated and demonstrated durable anti-tumor activity in heavily pretreated patients.[4] Notably, in a cohort of PD-1–naïve patients with hepatocellular carcinoma (HCC), the combination achieved an ORR of 33% (95% CI, 9.9%-65.1%).[4] A Phase 2 trial (NCT06236438) is actively recruiting patients with untreated metastatic non-squamous NSCLC to compare Livmoniplimab plus Budigalimab and chemotherapy against the standard-of-care, pembrolizumab plus chemotherapy.[22] Another Phase 2 study is evaluating the combination in metastatic urothelial carcinoma.[26]
  • With Novel Antibody-Drug Conjugates (ADCs) and Biologics: AbbVie is systematically combining Budigalimab with its pipeline of next-generation ADCs to pair targeted cytotoxic payload delivery with immune activation.
  • Telisotuzumab Adizutecan (ABBV-400): This is an ADC targeting c-Met. A Phase 1b/2 trial (NCT06772623) is evaluating its combination with Budigalimab in the first-line setting for advanced non-squamous NSCLC.[3] Another Phase 2 trial (NCT06628310) is studying a similar combination with chemotherapy in gastric, gastroesophageal junction, and esophageal adenocarcinoma.[3]
  • ABBV-706: This ADC targets SEZ6, a protein expressed on certain neuroendocrine tumors. A Phase 1 trial (NCT05599984) is assessing its safety and efficacy alone and in combination with Budigalimab in patients with SCLC, high-grade CNS tumors, and high-grade neuroendocrine carcinomas.[5]
  • Other Novel Agents: Budigalimab is also being studied in Phase 1 trials with Azirkitug (ABBV-514) in NSCLC and HNSCC (NCT05005403) and with ABBV-303 in advanced solid tumors (NCT06158958).[3]

Exploratory Indication: Human Immunodeficiency Virus (HIV)

In a significant departure from its oncology focus, Budigalimab is also being explored as a potential therapy for HIV.

  • Rationale: Chronic HIV infection is characterized by T-cell exhaustion, a state of immune dysfunction marked by high levels of PD-1 expression on T-cells.[10] The hypothesis is that blocking PD-1 with Budigalimab could reverse this exhaustion, reinvigorate HIV-specific T-cell responses, and potentially act as a latency-reversing agent to "kick" the virus out of its dormant cellular reservoirs.[10] The ultimate goal is to achieve a "functional cure," defined as durable, ART-free control of the virus.[10]
  • Clinical Data: Several Phase 1 studies (e.g., NCT04223804, NCT04799353) have evaluated the safety, PK, and PD of Budigalimab in people living with HIV-1.[31] In a small pilot study presented at EACS 2023, participants on stable ART received a short course of Budigalimab and then underwent an analytical treatment interruption (ATI).[10] The results showed that Budigalimab was well-tolerated. In 6 of 9 participants who completed treatment, it was associated with either a delayed viral rebound (median time to rebound of 29 days vs. 21 days for placebo) or sustained viral control at a low level (below 1000 copies/mL).[10] This represents an intriguing, albeit very early, signal of biological activity in a non-oncology setting. This investigation represents a high-risk, high-reward "flanker" strategy, attempting to create an entirely new and uncontested market for Budigalimab should it prove successful.

Safety and Tolerability Assessment

A thorough understanding of a drug's safety profile is paramount, particularly for an agent intended for broad use in combination with other therapies. The clinical data for Budigalimab indicate a safety and tolerability profile that is manageable and largely predictable for the anti-PD-1 class.

Overall Adverse Event Profile

Across its clinical program, Budigalimab has demonstrated a consistent pattern of adverse events (AEs).

  • Incidence: As is common with immunotherapies in advanced cancer populations, the incidence of AEs is high. In the foundational Phase 1 study in 59 patients with advanced solid tumors, 97% experienced at least one AE of any grade, and 64% experienced a Grade 3 or higher AE.[7] In the HNSCC and NSCLC expansion cohorts of the NCT03000257 study, 100% of the 81 patients reported at least one treatment-emergent AE (TEAE).[1]
  • Most Common AEs: The most frequently reported AEs of any grade across studies include fatigue, pruritus (itching), nausea, diarrhea, and anemia.[1] In the HNSCC and NSCLC expansion cohorts, anemia was the most common Grade ≥3 TEAE, occurring in 22% of HNSCC patients and 13% of NSCLC patients.[1]

Immune-Related Adverse Events (irAEs)

As a modulator of the immune system, Budigalimab's most characteristic toxicities are immune-related adverse events, which can affect a wide range of organ systems.

  • Profile and Consistency: The safety profile of Budigalimab has been repeatedly described as consistent with that of other approved PD-1 targeting agents.[2] Crucially, no novel or unexpected safety signals have been associated with its use, which provides a high degree of confidence for clinicians and regulators.[7] This predictable safety profile is a significant strategic asset. For a drug primarily intended as a combination backbone, a well-understood and manageable set of side effects simplifies the design of complex clinical trials, as the main safety variable becomes the novel partner agent. It de-risks AbbVie's broader development program by providing a stable, known foundation.
  • Incidence and Type: In the initial Phase 1 study, irAEs were reported in 11 of 59 patients (18.6%), with only one patient (1.7%) experiencing a Grade ≥3 irAE.[7] The most common irAEs observed were related to endocrine dysfunction, particularly hypothyroidism and hyperthyroidism.[7] Other reported irAEs include rash, pruritus, and diarrhea.[4] One serious Grade 3 irAE reported was a case of new-onset type 1 diabetes mellitus that presented as diabetic ketoacidosis, a known but rare complication of PD-1 inhibition.[7]

Discontinuation and Serious Adverse Events

  • Discontinuation Rates: Treatment discontinuation due to AEs provides a measure of a drug's overall tolerability. In the initial Phase 1 solid tumor study, 16 of 59 patients (27.1%) discontinued treatment due to an AE; however, the AE was considered related to Budigalimab in only two of those patients (3.3%).[7] In a combination trial with ABBV-151, treatment-related AEs led to discontinuation in 12% of patients.[4]
  • Fatal AEs: Across the key monotherapy studies, no treatment-related Grade 5 AEs (deaths) were reported.[7] Deaths that did occur were attributed to disease progression.[7]

Contraindications and Drug Interactions

As Budigalimab is an investigational agent, a formal list of contraindications and drug-drug interactions has not been established and is not available in the provided documentation.[1] However, the exclusion criteria from its clinical trials offer insight into patient populations where caution is warranted. These criteria are standard for the immune checkpoint inhibitor class and generally exclude patients with pre-existing conditions that could be exacerbated by systemic immune activation, such as:

  • Active autoimmune diseases (e.g., lupus, rheumatoid arthritis) [1]
  • A history of inflammatory bowel disease [1]
  • A history of immune-mediated pneumonitis [1]
  • Primary immunodeficiency or receipt of a solid organ transplant [1]

Table 3: Consolidated Safety Profile of Budigalimab in Key Monotherapy Cohorts

Adverse Event CategoryMonotherapy - Advanced Solid Tumors (N=59) 7Monotherapy - HNSCC (N=41) 1Monotherapy - NSCLC (N=40) 1
Any Grade TEAE97%100%100%
Grade ≥3 TEAE64%61%68%
Most Common Grade ≥3 TEAEN/AAnemia (22%)Anemia (13%)
Any Grade irAE18.6%N/AN/A
Grade ≥3 irAE1.7%N/AN/A
Common irAEsThyroid dysfunction, diabetic ketoacidosisHypothyroidism (20%), Diarrhea (15%), Pruritus (15%)Hypothyroidism (15%), Fatigue (13%)
Discontinuation due to TRAE3.3%7% (due to any AE)18% (due to any AE)
Treatment-Related Deaths0%0%0%

Strategic Assessment and Future Outlook

The development of Budigalimab is best understood not as an attempt to launch a superior monotherapy into a crowded market, but as a calculated, long-term strategic play by AbbVie to fortify and enhance its entire oncology pipeline.

Regulatory Status and Development Stage

As of the latest available information, Budigalimab is an investigational drug that has not received marketing approval from the U.S. Food and Drug Administration (FDA), the European Medicines Agency (EMA), or any other major global regulatory authority.[14] Its development is concentrated in Phase 1 and Phase 2 clinical trials.[11] The absence of any reported pivotal Phase 3 monotherapy trials is conspicuous. This is not likely an oversight but a deliberate strategic choice. Embarking on a large, expensive Phase 3 trial to challenge established agents like pembrolizumab or nivolumab head-to-head would be a high-risk, low-reward proposition, given that Budigalimab's monotherapy efficacy profile appears to be similar, not superior.[1] AbbVie appears to be wisely conceding the broad monotherapy market to focus resources on combination trials where a stronger case for clinical superiority and market differentiation can be made.

Competitive Landscape

The market for PD-1/PD-L1 inhibitors is mature and intensely competitive, dominated by a few blockbuster products that have set a high bar for efficacy and have been approved across dozens of indications.[36] Multiple analyses of Budigalimab's clinical profile conclude that its safety, efficacy, and biomarker effects are "similar to other PD-1 inhibitors".[1] With a comparable binding affinity to its main competitors and no clear evidence of superior clinical performance as a single agent, Budigalimab is not positioned to displace the current market leaders through direct competition.

Value Proposition and AbbVie's Strategy

The true value proposition of Budigalimab is internal and strategic, serving both defensive and offensive purposes for AbbVie.

  • The "Backbone" Strategy: The central pillar of the Budigalimab program is its role as a proprietary immunotherapy backbone.
  • Defensive Value: By developing its own in-house anti-PD-1 antibody, AbbVie gains complete strategic independence. It avoids the need to partner with direct competitors like Merck or Bristol Myers Squibb to conduct combination studies with its own novel assets (e.g., ADCs, bispecifics). This insulates AbbVie from paying substantial royalties to a competitor, thereby protecting future profit margins, and gives it full control over clinical trial design, execution, and timing.
  • Offensive Value: This strategy allows AbbVie to create fully-owned, proprietary combination regimens. If a trial demonstrates that "Budigalimab + Novel Agent X" is superior to the standard of care, AbbVie owns the entire patented regimen. This creates a formidable barrier to entry for competitors and allows the company to capture the full value of the combination, rather than just the value of the novel agent. Budigalimab is thus a key instrument for maximizing the commercial potential of AbbVie's entire oncology pipeline.

Future Trajectory

The future of Budigalimab is inextricably tied to the success of its combination partners. The most promising development paths are the ongoing and planned trials in indications where there remains a high unmet need, even in the immunotherapy era. The Phase 2 trial in first-line NSCLC (NCT06236438), which pits a Budigalimab-based combination directly against a pembrolizumab-based standard of care, will be a critical test of this strategy.[22] Similarly, the success of combinations with novel ADCs like telisotuzumab adizutecan and ABBV-706 will ultimately determine the clinical and commercial fate of Budigalimab.[3] The exploratory HIV program, while scientifically intriguing, remains a long-term, high-risk venture that is separate from the core oncology strategy.[10] In conclusion, Budigalimab should be viewed not as a product in itself, but as a critical enabler of AbbVie's broader, long-term ambition to become a leader in oncology through innovative, proprietary combination therapies.

Works cited

  1. First-in-human phase 1 study of budigalimab, an anti-PD-1 inhibitor, in patients with non-small cell lung cancer and head and neck squamous cell carcinoma - PMC - PubMed Central, accessed September 2, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC8783908/
  2. First-in-human phase 1 study of budigalimab, an anti-PD-1 inhibitor, in patients with non-small cell lung cancer and head and neck squamous cell carcinoma, accessed September 2, 2025, https://research-repository.uwa.edu.au/en/publications/first-in-human-phase-1-study-of-budigalimab-an-anti-pd-1-inhibito
  3. Clinical Trials Using Budigalimab - NCI, accessed September 2, 2025, https://www.cancer.gov/research/participate/clinical-trials/intervention/budigalimab?pn=1
  4. Budigalimab Plus ABBV-151 Demonstrates Variable Activity Across Locally Advanced or Metastatic Solid Tumors - OncLive, accessed September 2, 2025, https://www.onclive.com/view/budigalimab-plus-abbv-151-demonstrates-variable-activity-across-locally-advanced-or-metastatic-solid-tumors
  5. A phase 1 first-in-human study evaluating safety, pharmacokinetics and efficacy of ABBV-706 as monotherapy and in combination with budigalimab (ABBV-181), carboplatin, or cisplatin in adult subjects with advanced solid tumors. - MD Anderson Cancer Center, accessed September 2, 2025, https://www.mdanderson.org/patients-family/diagnosis-treatment/clinical-trials/clinical-trials-index/clinical-trials-detail.ID2022-1070.html
  6. CLINICAL TRIAL / NCT05599984 - UChicago Medicine, accessed September 2, 2025, https://www.uchicagomedicine.org/find-a-clinical-trial/clinical-trial/irb231757
  7. Model Informed Dosing Regimen and Phase I Results of the Anti‐PD‐1 Antibody Budigalimab (ABBV‐181) - PMC - PubMed Central, accessed September 2, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC7877859/
  8. Model Informed Dosing Regimen and Phase I Results of the Anti-PD-1 Antibody Budigalimab (ABBV-181) - PubMed, accessed September 2, 2025, https://pubmed.ncbi.nlm.nih.gov/32770720/
  9. (PDF) First-in-human phase 1 study of budigalimab, an anti-PD-1 inhibitor, in patients with non-small cell lung cancer and head and neck squamous cell carcinoma - ResearchGate, accessed September 2, 2025, https://www.researchgate.net/publication/352967367_First-in-human_phase_1_study_of_budigalimab_an_anti-PD-1_inhibitor_in_patients_with_non-small_cell_lung_cancer_and_head_and_neck_squamous_cell_carcinoma
  10. PD-1 inhibitor budigalimab may delay viral rebound - Aidsmap, accessed September 2, 2025, https://www.aidsmap.com/news/oct-2023/pd-1-inhibitor-budigalimab-may-delay-viral-rebound
  11. budigalimab - My Cancer Genome, accessed September 2, 2025, https://www.mycancergenome.org/content/drugs/budigalimab/
  12. Definition of budigalimab - NCI Drug Dictionary, accessed September 2, 2025, https://www.cancer.gov/publications/dictionaries/cancer-drug/def/budigalimab
  13. Pembrolizumab vs. nivolumab: How do they compare? - Drugs.com, accessed September 2, 2025, https://www.drugs.com/medical-answers/pembrolizumab-nivolumab-compare-3554067/
  14. Budigalimab - AbbVie - AdisInsight, accessed September 2, 2025, https://adisinsight.springer.com/drugs/800048662
  15. Budigalimab - Drug Targets, Indications, Patents - Patsnap Synapse, accessed September 2, 2025, https://synapse.patsnap.com/drug/1cb725147eb9430ca416dffcd5e841fe
  16. Budigalimab (ABBV 181) | PD-1 Receptor Inhibitor | MedChemExpress, accessed September 2, 2025, https://www.medchemexpress.com/budigalimab.html
  17. Budigalimab (Anti-PDCD1 / PD-1 / CD279) - Selleck Chemicals, accessed September 2, 2025, https://www.selleckchem.com/products/budigalimab-anti-pdcd1-pd-1-cd279.html
  18. Anti-PDCD1/PD-1/CD279 Antibody (Budigalimab), accessed September 2, 2025, https://www.apexbt.com/downloader/document/F1323/Datasheet.pdf
  19. pmc.ncbi.nlm.nih.gov, accessed September 2, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC7877859/#:~:text=Budigalimab%2C%20also%20known%20as%20ABBV,interactions%20and%20limit%20effector%20function.
  20. First-in-human phase 1 study of budigalimab, an anti-PD-1 inhibitor, in patients with non-small cell lung cancer and head and ne, accessed September 2, 2025, https://d-nb.info/1243158840/34
  21. Budigalimab Recruiting Phase 2 Trials for Gastroesophageal Junction Adenocarcinoma / Locally Advanced Unresectable or Metastatic Gastric Adenocarcinoma / Esophagus Adenocarcinoma Treatment - DrugBank, accessed September 2, 2025, https://go.drugbank.com/drugs/DB16668/clinical_trials?conditions=DBCOND0083226%2CDBCOND0038020%2CDBCOND0169298&phase=2&purpose=treatment&status=recruiting
  22. NSCLC, Squamous or Non-Squamous Recruiting Phase 2 Trials for Budigalimab (DB16668) | DrugBank Online, accessed September 2, 2025, https://go.drugbank.com/indications/DBCOND0130187/clinical_trials/DB16668?phase=2&status=recruiting
  23. Clinical Trial: NCT03000257 - My Cancer Genome, accessed September 2, 2025, https://www.mycancergenome.org/content/clinical_trials/NCT03000257/
  24. 770 Safety, efficacy, and pharmacokinetic results from a phase I first-in-human study of ABBV-151 with or without anti-PD1 mAb (budigalimab) in patients with locally advanced or metastatic solid tumors | Journal for ImmunoTherapy of Cancer, accessed September 2, 2025, https://jitc.bmj.com/content/10/Suppl_2/A801
  25. Non-Small Cell Lung Cancer Recruiting Phase 2 Trials for Budigalimab (DB16668), accessed September 2, 2025, https://go.drugbank.com/indications/DBCOND0132865/clinical_trials/DB16668?phase=2&status=recruiting
  26. Study to Evaluate Adverse Events and Change in Disease Activity When Intravenously (IV) Infused Livmoniplimab is Used in Combination With IV Infused Budigalimab in Adult Participants With Urothelial Carcinoma (UC) - ClinicalTrials.Veeva, accessed September 2, 2025, https://ctv.veeva.com/study/study-to-evaluate-adverse-events-and-change-in-disease-activity-when-intravenously-iv-infused-livm
  27. A phase 2, open-label, randomized study of livmoniplimab in combination with budigalimab versus chemotherapy in patients with metastatic urothelial carcinoma. - ASCO Publications, accessed September 2, 2025, https://ascopubs.org/doi/10.1200/JCO.2025.43.16_suppl.TPS4618
  28. Phase 1b/2 study evaluating telisotuzumab adizutecan (ABBV-400; Temab-A) in combination with budigalimab in patients (pts) with advanced non-squamous (NSQ) non-small cell lung cancer (NSCLC) with no prior treatment for advanced disease and no actionable genomic alterations. | Journal of Clinical Oncology - ASCO Publications, accessed September 2, 2025, https://ascopubs.org/doi/10.1200/JCO.2025.43.16_suppl.TPS8655
  29. ABBV-303 + Budigalimab for Solid Tumors - Clinical Trials, accessed September 2, 2025, https://www.withpower.com/trial/phase-1-tumors-solid-1-2024-a9b6f
  30. A global first-in-human study in NSCLC, HNSCC and solid tumors with ABBV-514 as a single agent and in combination with budigalimab, accessed September 2, 2025, https://www.mdanderson.org/patients-family/diagnosis-treatment/clinical-trials/clinical-trials-index/clinical-trials-detail.ID2024-1457.html
  31. Budigalimab Completed Phase 1 Trials for Human Immunodeficiency Virus (HIV) Infections ... - DrugBank, accessed September 2, 2025, https://go.drugbank.com/drugs/DB16668/clinical_trials?conditions=DBCOND0035016%2CDBCOND0042885&phase=1&purpose=treatment&status=completed
  32. Budigalimab | MedPath, accessed September 2, 2025, https://trial.medpath.com/drug/cbbb460727a49564/budigalimab?tab=clinical_trials&page=2
  33. Model Informed Dosing Regimen and Phase I Results of the Anti‐PD‐1 Antibody Budigalimab (ABBV‐181) - ResearchGate, accessed September 2, 2025, https://www.researchgate.net/publication/343533233_Model_Informed_Dosing_Regimen_and_Phase_I_Results_of_the_Anti-PD-1_Antibody_Budigalimab_ABBV-181
  34. A Study of Budigalimab (ABBV-181) in Participants With Advanced Solid Tumors - NCI, accessed September 2, 2025, https://www.cancer.gov/research/participate/clinical-trials-search/v?id=NCI-2017-01370
  35. Budigalimab by AbbVie for Metastatic Adenocarcinoma of The Pancreas: Likelihood of Approval - Pharmaceutical Technology, accessed September 2, 2025, https://www.pharmaceutical-technology.com/data-insights/budigalimab-abbvie-metastatic-adenocarcinoma-of-the-pancreas-likelihood-of-approval/
  36. PD1 Inhibitors Market And Pipeline Insights 2023, accessed September 2, 2025, https://www.precisionbusinessinsights.com/market-reports/pd1-inhibitors-market
  37. Keytruda vs Opdivo: Differences Between Immunotherapy Drugs - GoodRx, accessed September 2, 2025, https://www.goodrx.com/classes/pd-1-pd-l1-inhibitors/keytruda-vs-opdivo
  38. Association of Baseline and Pharmacodynamic Biomarkers With Outcomes in Patients Treated With the PD-1 Inhibitor Budigalimab - PubMed Central, accessed September 2, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC8906246/

Published at: September 2, 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.