Sponsor: Northwestern University (other)
Phase: 2
Start date: Jan. 31, 2024
Planned enrollment: 25
Balstilimab (AGEN2034, BAL) is a fully human monoclonal IgG4 antibody that binds with high affinity to programmed death 1 (PD-1), preventing its interaction with PD-L1 and PD-L2 ligands. The drug is designed to enhance T cell activation and effector function [1]. It is being developed by Agenus Inc. as both a monotherapy and in combination with other immunotherapy agents, particularly botensilimab (an anti-CTLA-4 antibody).
As a monotherapy, balstilimab showed activity in a phase II trial for recurrent/metastatic cervical cancer, with an objective response rate (ORR) of 15% and median duration of response of 15.4 months. In PD-L1 positive patients with squamous cell carcinoma, the ORR was 21%. The drug also showed some activity in PD-L1 negative patients [2]. When combined with zalifrelimab (anti-CTLA-4), the ORR increased to 25.6% in cervical cancer patients, with a disease control rate of 52% [3].
The safety profile appears manageable based on clinical trials. In combination therapy with zalifrelimab for cervical cancer, the most common treatment-related adverse events were hypothyroidism (16.8%), diarrhea (14.2%), fatigue (11.6%), and nausea (9.0%) [3]. Development history includes a withdrawn BLA for cervical cancer in 2021, but the drug continues to be studied in various combinations and indications, particularly with botensilimab in colorectal cancer and other solid tumors [4].
[1] Phase II Trial Results in Journal of Clinical Oncology [2] Clinical Trial Results in Gynecologic Oncology [3] Phase II Combination Trial Results in Journal of Clinical Oncology [4] Recent Development Update from BioSpace
Last updated: Dec 2024
Botensilimab is an investigational Fc-enhanced anti-CTLA-4 monoclonal antibody being developed by Agenus Inc. for cancer immunotherapy. It is designed to boost both innate and adaptive anti-tumor immune responses through multiple mechanisms: enhanced T cell priming and activation, depletion of regulatory T cells (Tregs) in the tumor microenvironment, activation of myeloid cells, and induction of long-term memory responses [1]. The drug's novel Fc-enhanced design allows it to bind more effectively to both high and low-affinity FcγRIIIA variants, potentially extending its benefits to a broader patient population compared to first-generation CTLA-4 inhibitors. Additionally, botensilimab is engineered to reduce complement-mediated toxicity through reduced C1q binding [2].
The drug has shown promising results in clinical trials, particularly in combination with the PD-1 inhibitor balstilimab. In a phase 1 trial of patients with microsatellite stable (MSS) metastatic colorectal cancer without liver metastases, the combination achieved a 23% objective response rate with a median overall survival of 21.2 months [3]. More recently, in the neoadjuvant NEST-1 trial for resectable colorectal cancer, the combination showed impressive pathologic response rates, with tumor shrinkage of ≥50% observed in 67.5% of MSS CRC patients [4]. The drug has demonstrated clinical responses across nine different types of metastatic, late-line cancers, including traditionally immunotherapy-resistant tumors.
The safety profile of botensilimab appears manageable based on clinical trial data. In the phase 1 trial combining botensilimab with balstilimab, the most common treatment-related adverse events were diarrhea/colitis, fatigue, and decreased appetite. Grade 3-4 adverse events occurred in approximately 39% of patients, with diarrhea/colitis being the most frequent serious adverse event. Notably, unlike some other CTLA-4 inhibitors, no cases of hypophysitis were reported in the monotherapy arm, which may be attributed to the drug's reduced complement activation [2]. As of 2024, approximately 1100 patients have been treated with botensilimab in phase 1 and phase 2 clinical trials.
[1] Cancer Discovery Article on Botensilimab Mechanism and Clinical Results [2] OncoDaily Detailed Review of Botensilimab [3] Phase 1 Trial Results in OncoLive [4] NEST-1 Trial Results Press Release
Last updated: Dec 2024
Goal: Evaluate safety, feasibility, and preliminary efficacy of combining immune modulation with repeated ultrasound-mediated, reversible blood–brain barrier (BBB) opening to enhance drug delivery in newly diagnosed glioblastoma.
Patients: Adults (≥18) with newly diagnosed, IDH1/2–wild-type, MGMT promoter–unmethylated glioblastoma who have completed standard radiotherapy (with or without temozolomide), ECOG 0–2, with residual tumor/resection cavity amenable to Sonocloud-9 sonication coverage, adequate organ function, and no prior immunotherapy. Key exclusions include multifocal disease not coverable within a 50-mm field or posterior fossa location, uncontrolled epilepsy, active autoimmune disease/contraindication to checkpoint inhibitors, recent investigational agents, uncontrolled illness, other recent active malignancy, and pregnancy/breastfeeding.
Design: Single-arm, phase 2a, open-label feasibility and safety study with a 6-patient safety run-in through cycle 2; planned enrollment of 25. Device implantation occurs 1–5 weeks post-radiotherapy; treatment is delivered in 21-day cycles for up to 9 cycles with possible extension if beneficial.
Treatments: Ultrasound-mediated BBB opening using the implanted Sonocloud-9 device every 3 weeks, synchronized with intravenous liposomal doxorubicin 30 mg q3w, balstilimab 450 mg q3w, and botensilimab 1 mg/kg q6w. Balstilimab is a human IgG4 anti–PD-1 antibody that blocks PD-1 interaction with PD-L1/PD-L2 to restore T-cell activity; in phase 2 cervical cancer it produced an ORR of about 15% as monotherapy with manageable immune-related AEs, and higher responses when combined with CTLA-4 blockade. Botensilimab is an Fc-enhanced anti–CTLA-4 antibody designed to augment T-cell priming and deplete intratumoral Tregs with reduced complement-mediated toxicity; in early trials, particularly in combination with balstilimab, it has shown activity across multiple solid tumors including MSS colorectal cancer with encouraging response rates and survival signals, with diarrhea/colitis the most common high-grade toxicity. Liposomal doxorubicin is a standard anthracycline formulation used for cytotoxic tumor kill with reduced peak cardiotoxicity compared with conventional doxorubicin.
Outcomes: Primary: unacceptable toxicity rate in cycle 1 (<33%) at 42 days; landmark overall and progression-free survival at 12 and 18 months, plus median PFS and OS. Secondary/other: predictive value of baseline tumor p-ERK expression for immunotherapy benefit; RANO-based response rate; MRI-defined patterns of recurrence relative to the sonication field; correlations of blood-based immune and nucleic acid/exosome profiles with PFS.
Burden on patient: Moderate to high. Patients undergo neurosurgical implantation of the Sonocloud-9 device shortly after radiotherapy, followed by frequent hospital visits every 3 weeks for sonication, IV infusions, and safety assessments over at least 6 months. MRI is performed to document BBB opening and for response/recurrence monitoring, and serial blood draws for ctDNA and immune profiling are collected before and after each sonication. The combination of two checkpoint inhibitors plus liposomal doxorubicin entails risk of immune-related adverse events and chemotherapy toxicities, potentially requiring additional clinic visits, labs, and supportive care. Travel and time commitments are greater than standard adjuvant management due to device activations, imaging, and intensified early safety monitoring in the run-in cohort.
Inclusion Criteria:
* Have newly diagnosed pathologically proven glioblastoma, isocitric dehydrogenase-1/2 wild-type
* Tumor with methyl guanine methyl transferase (MGMT) gene promoter unmethylated
* Available paraffin embedded tumor tissue for the study
* Have completed standard radiotherapy with or without temozolomide
* 18 years of age or older
* Able to undergo contrast-enhanced MRI
* Have an Eastern Cooperative Oncology Group/World Health Organization performance status ≤ 2
* Size and location of the residual tumor and/or resection cavity must allow to be able to be covered by the sonication field
* Have not received any prior treatment with immunotherapeutic agents treatments for glioblastoma or other indications
* Have the ability to understand and willingness to sign a written informed consent prior to registration on study.
* Be willing and able to comply with the protocol.
* Have adequate organ and bone marrow function
* Agree to use adequate contraception if appropriate
Exclusion Criteria: Patients will be ineligible if they have:
* Multifocal tumor (unless all localized in a 50-mm diameter area accessible to ultrasound field) or tumor located in the posterior fossa.
* Uncontrolled epilepsy.
* Received other investigational agents within 2 weeks of registration
* Received prior therapy with or have history of allergic reactions attributed to compounds of similar chemical or biologic composition to agents used in this study.
* Contraindication to checkpoint inhibitor therapy (e.g., history of autoimmune disease)
* Uncontrolled illness
* History of active malignancy other than the brain tumor within 12 months prior to registration.
* Are pregnant or breastfeeding.
Chicago, Illinois, 60611, United States
[email protected] / 13126959124
Status: Recruiting