Gotistobart (also known as ONC-392 and BNT-316) is an investigational, humanized IgG1 monoclonal antibody representing a next-generation approach to immune checkpoint inhibition by targeting the cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4). Its development is predicated on a novel, pH-sensitive mechanism of action designed to overcome the significant toxicity limitations of first-generation anti-CTLA-4 therapies. By binding to CTLA-4 at neutral pH and dissociating in the acidic tumor microenvironment (TME), gotistobart aims to preserve the CTLA-4 receptor, allowing it to be recycled to the T-cell surface. This mechanism is hypothesized to selectively deplete immunosuppressive regulatory T-cells (Tregs) within the tumor while maintaining systemic immune tolerance, thereby improving the therapeutic index.
The clinical development program, designated PRESERVE, has explored gotistobart as both a monotherapy and in combination regimens across a range of advanced, treatment-refractory solid tumors. Early data from the Phase 1/2 PRESERVE-001 trial demonstrated encouraging single-agent activity in patients with metastatic non-small cell lung cancer (NSCLC) that had progressed on prior PD-(L)1 inhibitors, with an objective response rate of 29.6%. Further studies have shown promising efficacy signals in combination with pembrolizumab for platinum-resistant ovarian cancer (PRESERVE-004) and with lutetium Lu 177 vipivotide tetraxetan for metastatic castration-resistant prostate cancer (PRESERVE-006).
However, the safety profile, while manageable in monotherapy, has proven more complex in combination settings, with high rates of Grade 3 or higher treatment-emergent adverse events, challenging the initial hypothesis of a universally superior safety profile. The most significant challenge to the program arose in October 2024, when the U.S. Food and Drug Administration (FDA) placed a partial clinical hold on the pivotal Phase 3 PRESERVE-003 trial in NSCLC. The hold was prompted by divergent efficacy results between patients with squamous and non-squamous histologies. While the hold was subsequently lifted, the trial's scope was narrowed to exclusively enroll patients with squamous NSCLC. This regulatory event represents a critical inflection point, significantly altering the drug's development trajectory and potential market. Gotistobart remains a scientifically compelling agent with demonstrated activity in difficult-to-treat cancers, but its path to approval is now more focused, with its ultimate clinical value contingent on the outcomes of its revised and ongoing clinical program.
Gotistobart is an investigational therapeutic agent classified as a humanized, pH-sensitive immunoglobulin (Ig) G1-kappa isotype monoclonal antibody.[1] It functions as an immune checkpoint inhibitor by targeting the cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4; CD152), a critical negative regulator of T-cell activation.[1] As a next-generation anti-CTLA-4 antibody, it is being developed for its potential antineoplastic activities across a spectrum of solid tumors.[1] The drug is identified by several code names and synonyms in scientific literature and clinical trial registries, including ONC-392, BNT-316, and anti-CTLA-4 monoclonal antibody ONC-392.[1]
The development and commercialization of gotistobart are managed through a strategic corporate partnership. The agent was originated by OncoImmune and is now being co-developed clinically by OncoC4, Inc. and BioNTech SE.[3] In a significant transaction in March 2023, BioNTech acquired exclusive global licensing rights to the asset from OncoC4 for an upfront payment of $200 million, underscoring the perceived value and potential of this next-generation antibody.[7] Under the terms of the agreement, BioNTech holds the rights for commercialization, while the two companies jointly pursue clinical development in various solid tumor indications.[7]
The advent of immune checkpoint inhibitors, particularly those targeting the programmed death 1 (PD-1) and programmed death-ligand 1 (PD-L1) axis, has revolutionized the treatment of many advanced cancers. However, a substantial proportion of patients either do not respond to these therapies (primary resistance) or experience disease progression after an initial response (acquired resistance).[10] This population of patients with immunotherapy-resistant disease represents a major and growing unmet medical need, as subsequent treatment options are often limited and confer modest benefit.[11]
The clinical development program for gotistobart has been strategically designed to address this specific therapeutic gap. The target populations for its most prominent clinical trials consist of patients with advanced cancers that have progressed on standard-of-care treatments, including prior immunotherapy. For example, the pivotal PRESERVE-003 trial is enrolling patients with metastatic non-small cell lung cancer (NSCLC) whose disease has progressed on anti-PD-1/PD-L1 antibody-based therapy.[2] Similarly, the PRESERVE-004 trial is focused on patients with platinum-resistant ovarian cancer, another notoriously difficult-to-treat condition.[14] This deliberate focus positions gotistobart not as a direct competitor to established first-line immunotherapies but as a potential second- or third-line "salvage" therapy for whom current options have failed.
This strategic decision to target immunotherapy-resistant populations represents a high-risk, high-reward approach. Success in this setting would establish gotistobart in a distinct and significant market with a profound unmet need, creating a protected therapeutic niche. However, this patient population is inherently more challenging to treat. These patients often have more aggressive tumor biology, exhausted T-cell populations, and complex resistance mechanisms that make them less responsive to any form of therapy. The high-risk nature of this strategy was starkly illustrated by the partial clinical hold placed on the PRESERVE-003 trial, which was triggered by differential efficacy outcomes within the study population.[8] This event suggests that the complex biology of these resistant tumors may be exposing limitations in the drug's mechanism of action that might not have been apparent in a less heavily pre-treated, first-line patient population.
CTLA-4 is a protein receptor and a member of the immunoglobulin superfamily that is expressed on the surface of T-cells, with particularly high expression on immunosuppressive regulatory T-cells (Tregs).[1] It functions as a critical inhibitory immune checkpoint, playing a key role in the downregulation of the immune system.[1] When T-cells are activated, CTLA-4 is upregulated and competes with the co-stimulatory receptor CD28 for binding to ligands B7-1 (CD80) and B7-2 (CD86) on antigen-presenting cells. Because CTLA-4 binds these ligands with much higher affinity than CD28, it effectively outcompetes the stimulatory signal, leading to the inhibition of T-cell proliferation and cytokine production.[1]
This regulatory function makes CTLA-4 a "double-edged sword".[3] On one hand, it is essential for maintaining self-tolerance and preventing the development of autoimmune diseases by reining in excessive immune responses. On the other hand, this same mechanism can be co-opted by cancer cells to evade immune surveillance. By promoting an immunosuppressive tumor microenvironment (TME) rich in Tregs and by directly inhibiting the activation of tumor-specific cytotoxic T-lymphocytes (CTLs), the CTLA-4 pathway allows tumors to escape destruction by the host immune system.[3]
The discovery of CTLA-4's role in immune suppression led to the development of the first generation of immune checkpoint inhibitors, such as ipilimumab. These antibodies work by blocking the interaction between CTLA-4 and its B7 ligands, thereby removing the inhibitory "brake" on T-cell activation and unleashing an anti-tumor immune response. While these agents have demonstrated significant anti-cancer activity, particularly in melanoma, their clinical utility has been hampered by a narrow therapeutic window. The non-specific, systemic blockade of CTLA-4 leads to widespread immune disinhibition, resulting in a high incidence of severe, and sometimes life-threatening, immune-related adverse events (irAEs).[2] These toxicities, which can affect multiple organ systems, often limit the dose and duration of therapy that patients can tolerate.[2]
A key mechanistic flaw identified with these first-generation antibodies is their effect on the CTLA-4 receptor itself. After the antibody binds to CTLA-4 on the T-cell surface, the entire complex is internalized. The antibody then traffics the receptor to the lysosome, where it is degraded.[3] This process of antibody-induced lysosomal degradation permanently removes the CTLA-4 checkpoint from the cell surface. This not only contributes to systemic toxicity by destroying a key mechanism of immune tolerance but may also paradoxically limit anti-tumor efficacy. By reducing the density of the CTLA-4 target on Tregs within the TME, it diminishes the potential for antibody-dependent cell-mediated cytotoxicity (ADCC) against these immunosuppressive cells, a key proposed mechanism for anti-tumor effect.[17]
Gotistobart was engineered to address the fundamental limitations of first-generation anti-CTLA-4 antibodies through an innovative, pH-sensitive design.[1] The core of this innovation lies in the antibody's binding characteristics. Gotistobart binds with high affinity to the CTLA-4 receptor at the neutral physiological pH (
pH≈7.4) found in blood and peripheral tissues. However, after the antibody-receptor complex is internalized into the endosome, where the environment becomes acidic (pH≈5.5−6.0), the antibody's conformation changes, causing it to dissociate from CTLA-4.[1]
This pH-dependent dissociation is a critical mechanistic differentiator. Because the antibody unbinds from the receptor within the endosome, it does not traffic CTLA-4 to the lysosome for degradation. Instead, the freed CTLA-4 receptor is recycled back to the T-cell surface, preserving its presence and function.[1] This mechanism has two profound, hypothesized consequences. First, within the characteristically acidic TME, the high density of recycled CTLA-4 on tumor-infiltrating Tregs makes them highly susceptible to selective depletion via ADCC. This enhances the local anti-tumor immune response. Second, in peripheral tissues where the pH is neutral, the preservation of CTLA-4 function on T-cells helps to maintain systemic immune tolerance, thereby reducing the risk of off-target irAEs.[1] This dual action—enhancing local efficacy while reducing systemic toxicity—is the central hypothesis for gotistobart's improved therapeutic index.
This design represents a conceptual shift from "brute-force" systemic checkpoint blockade to a more nuanced, "location-aware" form of immunomodulation. The drug's activity is theoretically coupled to the unique biochemical properties of the TME. This dependency on local acidity implies that the efficacy of gotistobart could vary between different tumor types, or even between individual patients, based on the metabolic state and resulting acidity of their specific TME. This factor, which is not typically considered for standard immunotherapies, may provide a biological rationale for the "varying results" observed between different NSCLC histologies in the PRESERVE-003 trial, as different tumor subtypes may foster distinct microenvironments.
The development of gotistobart was guided by preclinical studies in humanized CTLA-4 mouse models, which are engineered to express the human CTLA-4 protein. In these in vivo screening models, the parental clone of gotistobart was specifically selected for its ability to induce high anti-tumor efficacy while demonstrating low autoimmune toxicity.[2] These preclinical findings provided the initial proof-of-concept for the pH-sensitive recycling mechanism.
Based on this mechanism and the supporting preclinical data, several key therapeutic advantages for gotistobart have been hypothesized. The primary advantage is an improved safety profile, which is expected to allow for the administration of higher and more sustained doses compared to first-generation agents.[3] Preclinical data suggested that gotistobart could be dosed up to 10 mg/kg with greater efficacy and fewer adverse effects, a significant increase over the approximately 3 mg/kg dose limit for ipilimumab.[10] This wider therapeutic window is anticipated to translate into reduced systemic toxicity and potent single-agent anti-tumor activity, even in cancers that are typically resistant to immunotherapy.[3]
A robust understanding of a drug's pharmacokinetic profile is essential for optimizing its clinical development. A population PK analysis for gotistobart was conducted using data from the foundational PRESERVE-001 study. The model was built upon 420 measurable PK observations collected from 70 patients with advanced solid tumors, including 57 who received gotistobart as a monotherapy and 13 who received it in combination with pembrolizumab.[19]
Gotistobart is administered via intravenous (IV) infusion, ensuring 100% bioavailability.[2] The analysis of its concentration-time data revealed that its pharmacokinetic behavior is best described by a two-compartment model with first-order elimination.[19] This model suggests that after administration, the drug distributes from a central compartment (primarily the bloodstream) into a peripheral compartment (tissues) before being eliminated from the central compartment.
The population PK model estimated the systemic clearance (CL) of gotistobart to be approximately 182 mL/day.[19] The terminal elimination half-life (
t1/2) was estimated to be 25.7 days.[19] These parameters are highly favorable for a therapeutic monoclonal antibody. The slow clearance and long half-life indicate that the drug remains in the body for an extended period, allowing for sustained target engagement with less frequent dosing (e.g., every 3-4 weeks), which is a desirable characteristic for chronic cancer therapy.
The PK analysis also investigated the influence of various patient-specific factors (covariates) on the drug's disposition. The model identified only two significant covariates: serum albumin levels and body weight. Increased albumin levels were associated with a decrease in drug clearance, while increased body weight was associated with larger central and peripheral volumes of distribution (V1 and V2).[19] The relationship with body weight is expected for monoclonal antibodies and is the rationale for the standard practice of weight-based (mg/kg) dosing.
Crucially, the analysis found that a range of other common clinical variables had no significant effect on gotistobart's PK profile. These included age, sex, race, baseline liver function (as measured by AST and bilirubin), and creatinine clearance.[19] Furthermore, the model showed that the concurrent administration of the anti-PD-1 antibody pembrolizumab did not significantly alter the pharmacokinetics of gotistobart.[19] This predictable and robust PK profile is a significant asset for clinical development. The lack of influence from common patient factors simplifies dosing protocols and broadens the drug's potential applicability without the need for complex dose adjustments. The finding that pembrolizumab does not impact gotistobart's PK is particularly important, as it de-risks the combination strategy from a pharmacokinetic perspective. It suggests that any increase in toxicity observed in combination trials is likely due to additive pharmacodynamic effects (i.e., enhanced immune stimulation) rather than a pharmacokinetic drug-drug interaction where one drug alters the clearance of the other.
| Parameter | Value | Source | 
|---|---|---|
| Pharmacokinetic Model | 2-compartment with first-order elimination | 19 | 
| Systemic Clearance (CL) | 182 mL/day | 19 | 
| Terminal Half-Life (t1/2) | 25.7 days | 19 | 
| Significant Covariates | - Increased albumin associated with decreased CL - Increased body weight associated with increased V1 and V2 | 19 | 
| Non-Significant Covariates | Age, sex, race, AST, bilirubin, creatinine clearance, cancer type, concurrent pembrolizumab | 19 | 
| Table 3. Summary of Gotistobart Pharmacokinetic Parameters | 
Specific absorption, distribution, metabolism, and excretion (ADME) studies for gotistobart have not been detailed in the available documentation.[4] However, as a monoclonal antibody, its metabolic fate is expected to follow the well-established pathways for this class of therapeutic proteins. Unlike small-molecule drugs, which are typically metabolized by the cytochrome P450 (CYP) enzyme system in the liver, mAbs are too large to be processed by these enzymes.[21] Instead, they are cleared from the body through catabolism. This process involves non-specific endocytosis by various cells throughout the body, where they are broken down by proteolytic enzymes into smaller peptides and constituent amino acids.[21] These resulting amino acids can then be re-utilized by the body for the synthesis of new proteins.[23]
Similarly, the excretion of intact monoclonal antibodies is negligible. Their large molecular size prevents them from being filtered by the glomerulus in the kidney and excreted in the urine.[22] Therefore, clearance is driven primarily by cellular uptake and catabolism, as well as target-mediated drug disposition (TMDD), where binding to the CTLA-4 target can also lead to internalization and degradation of the antibody-receptor complex.[22]
The available data does not specify any formal drug-drug interaction studies for gotistobart.[5] Based on its metabolic profile, the potential for pharmacokinetic interactions with drugs metabolized by the CYP450 system is considered very low, a general characteristic of monoclonal antibodies.[26] This is supported by the population PK analysis, which found that concurrent administration of pembrolizumab did not significantly affect gotistobart's clearance or half-life.[19]
The primary concern for drug interactions with gotistobart, as with other immunotherapies, lies in the realm of pharmacodynamics. When combined with other immune-activating agents, there is a potential for additive or synergistic effects on the immune system. While this can lead to enhanced anti-tumor efficacy, it can also result in an increased incidence and severity of irAEs. This was observed in the PRESERVE-004 trial, where the combination of gotistobart and pembrolizumab led to high rates of Grade 3 or higher adverse events.[14] Therefore, while metabolic interactions are unlikely, careful monitoring for overlapping and enhanced pharmacodynamic toxicities is critical when gotistobart is used in combination regimens.
The clinical evaluation of gotistobart is being conducted through a comprehensive series of studies collectively known as the PRESERVE program. This program is designed to assess the safety and efficacy of gotistobart as a monotherapy and in various combination regimens across multiple advanced solid tumor types, with a particular focus on treatment-resistant populations.
| Trial Identifier | Phase | Indication(s) | Treatment Arm(s) | Status | 
|---|---|---|---|---|
| PRESERVE-001 (NCT04140526) | 1/2 | Advanced Solid Tumors, including NSCLC | - Gotistobart Monotherapy - Gotistobart + Pembrolizumab | Active, Not Recruiting | 
| PRESERVE-003 (NCT05671510) | 3 | Metastatic NSCLC (post-PD-(L)1) | - Gotistobart Monotherapy - Docetaxel (Active Control) | Active (Enrollment limited to squamous histology) | 
| PRESERVE-004 (NCT05446298) | 2 | Platinum-Resistant Ovarian Cancer | - Gotistobart + Pembrolizumab | Active | 
| PRESERVE-006 (NCT05682443) | 1/2 | Metastatic Castration-Resistant Prostate Cancer | - Gotistobart + Lutetium Lu 177 - Lutetium Lu 177 (Active Control) | Active | 
| Table 1. Overview of the Gotistobart Clinical Trial Program | 
The PRESERVE-001 trial served as the first-in-human study for gotistobart. It was designed as an open-label, dose-escalation and cohort-expansion study to establish the safety, tolerability, pharmacokinetics, and preliminary efficacy of the drug, both as a single agent and in combination with the anti-PD-1 antibody pembrolizumab.[3]
A key cohort within this trial focused on patients with metastatic NSCLC who were resistant to prior PD-(L)1 inhibitor therapy, representing a significant unmet need. The data from this monotherapy cohort provided the foundational evidence of gotistobart's clinical activity. In an analysis of 27 evaluable patients who received a regimen of two loading doses of 10 mg/kg followed by 6 mg/kg every three weeks, gotistobart demonstrated substantial anti-tumor effects.[11] The confirmed objective response rate (ORR) was 29.6%, which included one patient with a complete response (CR) and seven patients with partial responses (PRs).[11] The disease control rate (DCR), which includes patients with stable disease, was 70.4%.[11] The responses appeared to be durable, with a median duration of response (DOR) exceeding six months.[20] This level of single-agent activity in a heavily pre-treated, immunotherapy-resistant population was highly encouraging and provided the primary rationale for advancing gotistobart into pivotal Phase 3 testing. Anecdotal case studies from the program also highlighted its potential, with reports of single doses inducing regression of large metastatic lesions in lung cancer patients.[3]
Building on the promising results from PRESERVE-001, the PRESERVE-003 trial was initiated as a pivotal, potentially registrational study. It is a large-scale, two-stage, randomized, open-label, active-controlled Phase 3 trial designed to enroll approximately 600 patients globally.[2] The trial's primary objective is to determine whether gotistobart monotherapy can prolong overall survival (OS) compared to the standard-of-care second-line chemotherapy agent, docetaxel.[2] The study population is precisely defined as patients with metastatic NSCLC whose disease has progressed during or after treatment with a PD-1/PD-L1 inhibitor-based regimen.[2]
The trial employed a seamless two-stage design. Stage 1 served as a dose-confirmation phase, where patients were randomized to one of two gotistobart dosing regimens (a lower dose of 3 mg/kg every 3 weeks, or a higher dose involving two 10 mg/kg loading doses followed by 6 mg/kg every 3 weeks) or to the docetaxel control arm.[12] An analysis of the safety, efficacy, and exposure-response data from Stage 1 supported the selection of the high-dose regimen to move forward into the pivotal Stage 2 portion of the trial.[31] Stage 2 involves a direct 1:1 randomization between the selected gotistobart dose and docetaxel.[2] The status of this critical trial is active but has been significantly impacted by a partial clinical hold, which is discussed in detail in Section 6.0.
To explore its potential in other tumor types and in combination settings, the PRESERVE-004 trial was initiated. This is a randomized, open-label Phase 2 study evaluating the combination of gotistobart with pembrolizumab in patients with platinum-resistant high-grade serous ovarian cancer, a population with a poor prognosis and limited effective treatment options.[3]
Preliminary efficacy data from this trial were presented for two different gotistobart dose levels, both in combination with a standard dose of pembrolizumab. In the cohort of 32 evaluable patients receiving 1 mg/kg of gotistobart, the ORR was 25.0%, which included a 3.1% CR rate and a 21.9% PR rate.[14] In the cohort of 29 evaluable patients receiving a higher dose of 2 mg/kg of gotistobart, the ORR was slightly higher at 27.6%, with a 6.9% CR rate and a 20.7% PR rate.[14] These response rates were considered clinically meaningful and encouraging in this heavily pre-treated, refractory patient population, providing support for the continued development of this combination.
The PRESERVE-006 trial explores a novel therapeutic synergy by combining immunotherapy with radioligand therapy. This Phase 1/2, open-label, randomized study is evaluating gotistobart in combination with PLUVICTO® (lutetium Lu 177 vipivotide tetraxetan) in patients with metastatic castration-resistant prostate cancer (mCRPC) who have progressed on prior lines of therapy.[3] The scientific rationale for this combination is based on preclinical evidence suggesting that radiotherapy can induce an influx of immunosuppressive Tregs into the TME. Gotistobart, with its mechanism of selective Treg depletion, is hypothesized to counteract this effect and potentiate the anti-tumor activity of the radioligand therapy.[34]
Preliminary findings from the Phase 1 dose-escalation portion of the study, reported as of December 2024, have shown promising signs of efficacy. The study measured the rate of confirmed PSA50 response, defined as a decline in prostate-specific antigen (PSA) levels of 50% or more from baseline. In the combination arm receiving 3 mg/kg of gotistobart, a PSA50 response was observed in 4 of 6 patients (67%). In the 10 mg/kg arm, 3 of 6 patients (50%) achieved a PSA50 response. This contrasts sharply with the control arm, where only 1 of 6 patients (17%) receiving Lu 177 monotherapy achieved a PSA50 response.[34] These early data suggest that the combination may be substantially more active than radioligand therapy alone, supporting the underlying biological hypothesis.
| Trial | Indication | Treatment | Key Efficacy Endpoint | Result | 
|---|---|---|---|---|
| PRESERVE-001 | PD-(L)1 Resistant NSCLC | Gotistobart Monotherapy | ORR | 29.6% | 
| PRESERVE-004 | Platinum-Resistant Ovarian Cancer | Gotistobart + Pembrolizumab | ORR (1 mg/kg cohort) | 25.0% | 
| ORR (2 mg/kg cohort) | 27.6% | |||
| PRESERVE-006 | mCRPC | Gotistobart + Lu 177 | PSA50 Response (3 mg/kg cohort) | 67% | 
| (vs. Lu 177 alone) | PSA50 Response (Control) | 17% | ||
| Table 2. Summary of Key Efficacy Outcomes from the PRESERVE Program | 
A central tenet of gotistobart's development is the hypothesis that its unique, pH-sensitive mechanism will translate into an improved safety profile compared to first-generation anti-CTLA-4 antibodies. The clinical data gathered from the PRESERVE program provides a nuanced and detailed picture of its tolerability, both as a single agent and in combination with other anti-cancer therapies.
The most comprehensive safety dataset available comes from the PRESERVE-004 trial, which evaluated gotistobart in combination with pembrolizumab in patients with ovarian cancer.[14] In this combination setting, the incidence of TEAEs was high. Any-grade TEAEs were reported in virtually all patients: 100% of those in the 1 mg/kg gotistobart cohort and 96.6% in the 2 mg/kg cohort.[14]
More clinically significant were the rates of severe adverse events. Grade 3 or higher TEAEs occurred in 75.8% of patients in the 1 mg/kg group and 86.2% of patients in the 2 mg/kg group.[14] Serious TEAEs, defined as events that are life-threatening, require hospitalization, or result in significant disability, were also frequent, reported in 54.5% of patients in the 1 mg/kg cohort and 72.4% of those in the 2 mg/kg cohort.[14] The frequency and severity of these events had a direct impact on treatment continuation, with TEAEs leading to the discontinuation of gotistobart in 21.2% and 27.6% of patients in the low- and high-dose cohorts, respectively.[14]
As an immunotherapy, a key focus of the safety assessment is on irAEs, which result from the drug's mechanism of immune stimulation. The safety profile of gotistobart in this regard appears to differ between monotherapy and combination therapy.
In the PRESERVE-001 trial, where gotistobart was administered as a single agent to patients with NSCLC, the high-dose regimen was associated with a Grade 3-4 irAE rate of 30%.[20] While substantial, this rate is generally considered manageable and potentially favorable within the context of the anti-CTLA-4 drug class. Notably, no Grade 5 (fatal) treatment-related adverse events were observed in this cohort.[20]
In contrast, the rates of irAEs were higher in the combination setting of the PRESERVE-004 trial. Grade 3 or higher irAEs were experienced by 24.2% of patients in the 1 mg/kg cohort and rose to 37.9% in the 2 mg/kg cohort.[14] The most common irAEs reported in the trial included fatigue, elevations in liver enzymes (AST/ALT), diarrhea, pruritus (itching), and colitis (inflammation of the colon).[14]
In the PRESERVE-006 trial, which combined gotistobart with radioligand therapy, the safety profile was deemed manageable overall, but a clear dose-dependent toxicity signal emerged. The most significant irAE was Grade 3 colitis, which was observed in two of six patients (33%) at the highest dose level of 10 mg/kg, leading to treatment discontinuation in both cases. This event was not observed at the lower dose levels. No Grade 4 or 5 treatment-related AEs were reported in the study's dose-escalation phase.[34]
The collective safety data indicates a clear relationship between the dose of gotistobart, the use of combination therapy, and the incidence of severe toxicity. The experience in the PRESERVE-006 trial, where the 10 mg/kg dose led to an unacceptable rate of severe colitis, directly informed the decision to proceed with lower doses for the Phase 2 portion of the study.[34] Similarly, the high rates of TEAEs and discontinuations in the PRESERVE-004 trial (21-28%) underscore that even with a novel mechanism, mitigating toxicity remains a significant clinical challenge in dual checkpoint inhibitor regimens.[14]
The clinical data, when viewed holistically, presents a complex and somewhat contradictory assessment of gotistobart's core safety proposition. The monotherapy data from PRESERVE-001 appears to support the hypothesis of an improved therapeutic index, with a manageable rate of severe irAEs. However, this safety advantage seems to erode significantly when gotistobart is combined with other agents like pembrolizumab or high-dose radioligand therapy. This suggests that the pharmacodynamic synergy that enhances anti-tumor efficacy may also create a level of immune activation that overwhelms the protective effect of the pH-sensitive mechanism, negating some of the anticipated safety benefits. This has important implications for the drug's future development, suggesting that its path as a monotherapy may be more straightforward, while its role as a combination partner will require careful dose selection and patient management to balance efficacy and toxicity.
| Trial | Treatment | Grade ≥3 TEAEs | Grade ≥3 irAEs | Discontinuation due to AEs | 
|---|---|---|---|---|
| PRESERVE-001 | Monotherapy (NSCLC) | Not Reported | 30% | Not Reported | 
| PRESERVE-004 | Combo w/ Pembrolizumab (Ovarian Cancer) | 75.8% (1 mg/kg) 86.2% (2 mg/kg) | 24.2% (1 mg/kg) 37.9% (2 mg/kg) | 21.2% (1 mg/kg) 27.6% (2 mg/kg) | 
| PRESERVE-006 | Combo w/ Lu 177 (Prostate Cancer) | Not Reported (Grade 3 Colitis in 33% at 10 mg/kg) | Not Reported | 33% (at 10 mg/kg) | 
| Table 3. Consolidated Safety Profile: Grade ≥3 Adverse Events and Discontinuations | 
Gotistobart is currently a new molecular entity in late-stage clinical development. It has not received marketing authorization from any major global regulatory agency, including the United States Food and Drug Administration (FDA), the European Medicines Agency (EMA), or Australia's Therapeutic Goods Administration (TGA).[4] The available documentation contains no information regarding regulatory submissions or reviews by the EMA or TGA, indicating that its development is most advanced in the United States.[33]
In recognition of its potential to address a serious unmet medical need, gotistobart was granted Fast Track Designation by the FDA in 2022.[6] This designation was for its development as a monotherapy for patients with metastatic, immunotherapy-resistant NSCLC and was based on the encouraging preliminary safety and efficacy data from the Phase 1/2 PRESERVE-001 trial. The Fast Track program is designed to facilitate the development and expedite the review of drugs intended to treat serious conditions, allowing for more frequent interactions with the FDA and eligibility for accelerated approval and priority review if relevant criteria are met.
The development trajectory of gotistobart encountered a significant hurdle in October 2024, when the FDA placed a partial clinical hold on the pivotal PRESERVE-003 trial.[4] This regulatory action was not due to an unexpected safety signal but was prompted by an efficacy-related issue. Following a routine review of accumulating data, the trial's independent data monitoring committee identified a "possible variance in population results," specifically noting "varying results between the squamous and non-squamous non-small cell lung cancer (NSCLC) patient populations".[7] In response to this finding, the developers, OncoC4 and BioNTech, proactively paused the enrollment of new patients and informed the FDA, which subsequently formalized the pause with a partial clinical hold.[7]
The impact of the hold was confined to new patient enrollment in the PRESERVE-003 study; patients already enrolled were permitted to continue receiving treatment, and all other ongoing clinical trials of gotistobart in different indications (ovarian cancer, prostate cancer, etc.) were unaffected.[7]
After further discussion and alignment with the FDA, the partial clinical hold was lifted. However, this resolution came with a critical and transformative amendment to the trial protocol: future enrollment in PRESERVE-003 would be restricted exclusively to patients with the squamous histology of NSCLC.[44]
This sequence of events represents a major pivot in the drug's development strategy. The initial design of PRESERVE-003, which included both major NSCLC subtypes, was aimed at securing a broad indication in the immunotherapy-resistant setting. The divergence in efficacy signals strong enough to warrant a clinical hold implies that the underlying tumor biology of non-squamous NSCLC may be inherently less responsive to gotistobart's mechanism of action. This could be due to a variety of factors, such as a less acidic TME, different compositions of Treg populations, or the presence of alternative immune escape pathways in non-squamous tumors. The FDA's decision to allow the trial to proceed only for the squamous cohort effectively closes the development path for gotistobart in non-squamous NSCLC for the foreseeable future. This critically undermines the initial hypothesis that the drug's novel mechanism would be universally applicable across immunotherapy-resistant tumors and has fundamentally reshaped its clinical and commercial potential.
The path forward for gotistobart in NSCLC is now significantly narrowed, with the registrational potential of the PRESERVE-003 trial hinging entirely on its ability to demonstrate a survival benefit in the squamous NSCLC subpopulation.[31] This places immense pressure on the outcome of this specific cohort.
Simultaneously, the development programs in other indications, which were unaffected by the hold, have gained in strategic importance. The ongoing trials in platinum-resistant ovarian cancer (PRESERVE-004) and metastatic castration-resistant prostate cancer (PRESERVE-006) represent viable alternative pathways to a first regulatory approval.[3] Positive results from these studies would not only open up new therapeutic areas for gotistobart but would also help to validate its mechanism of action in tumor types beyond NSCLC, potentially reigniting interest in exploring its activity in other cancer settings.
Gotistobart entered the clinical development landscape as a promising, scientifically rational, second-generation anti-CTLA-4 antibody. Its novel pH-sensitive mechanism, designed to improve the therapeutic index by localizing its effect to the tumor microenvironment, represents a thoughtful and innovative approach to overcoming the well-documented limitations of its predecessors. The early monotherapy data generated in the PRESERVE-001 trial remains a significant achievement. Demonstrating a nearly 30% objective response rate in a highly refractory population of PD-(L)1-resistant NSCLC patients is a notable accomplishment and a key point of differentiation for the agent, providing a strong proof-of-concept for its single-agent activity.
Despite its elegant preclinical hypothesis, the clinical development of gotistobart has underscored the immense challenges of translating laboratory concepts into consistent clinical benefit. The journey of the PRESERVE-003 trial serves as a powerful case study in the complexities of modern oncology drug development. The partial clinical hold, triggered by efficacy differences between histological subtypes, illustrates that even a well-designed mechanism can encounter unforeseen biological limitations when faced with the heterogeneity of human cancer. It is a stark reminder that the tumor microenvironment is not a monolith, and factors like histology can profoundly influence therapeutic response.
Furthermore, the comprehensive safety data has painted a more complicated picture than initially hoped. While the irAE profile in monotherapy appears manageable and potentially improved, the safety advantage is less clear in combination settings. The high rates of severe adverse events and discontinuations seen when combined with pembrolizumab highlight the persistent difficulty of safely combining multiple immune-activating agents. This suggests that while the pH-sensitive mechanism may mitigate some toxicities, it does not fully abrogate the risks inherent in potent, multi-agent immunotherapy regimens.
The overall outlook for gotistobart has been reshaped by its clinical and regulatory journey. It has transitioned from a candidate with broad potential in NSCLC to a more specialized, histology-specific agent. Its success is now heavily contingent on the outcome of the pivotal PRESERVE-003 trial in the squamous NSCLC subpopulation. A positive result in this setting would still represent a major clinical advance for a patient group with few good options and would validate the drug's core mechanism.
The ongoing trials in ovarian and prostate cancer have become critically important, as they offer alternative pathways to market and could demonstrate the applicability of its mechanism in other solid tumors. Ultimately, gotistobart remains a promising and important investigational agent. Its story is one of scientific innovation confronting clinical reality. While its path to approval is now narrower and more challenging than initially envisioned, its demonstrated activity in difficult-to-treat cancers ensures that it will remain a significant focus of clinical research as its value in the therapeutic armamentarium continues to be defined.
Published at: September 28, 2025
This report is continuously updated as new research emerges.
Empowering clinical research with data-driven insights and AI-powered tools.
© 2025 MedPath, Inc. All rights reserved.