Complement factor H targeting antibody GT103 in refractory non-small cell lung cancer: a phase 1b dose escalation trial.
Date
2025-01
Journal Title
Journal ISSN
Volume Title
Repository Usage Stats
views
downloads
Citation Stats
Attention Stats
Abstract
GT103 is a first-in-class, fully human, IgG3 monoclonal antibody targeting complement factor H that kills tumor cells and promotes anti-cancer immunity in preclinical models. We conducted a first-in-human phase 1b study dose escalation trial of GT103 in refractory non-small cell lung cancer to assess the safety of GT103 (NCT04314089). Dose escalation was performed using a "3 + 3" schema with primary objectives of determining safety, tolerability, PK profile and maximum tolerated dose (MTD) of GT103. Secondary objectives included describing objective response rate, progression-free survival and overall survival. Dose escalation cohorts included GT103 given intravenously at 0.3, 1, 3, 10, and 15 mg/kg every 3 weeks, and 10 mg/kg every 2 weeks. Thirty one patients were enrolled across 3 institutions. Two dose-limiting adverse events were reported: grade 3 acute kidney injury (0.3 mg/kg) and grade 2 colitis (1 mg/kg). No dose-limiting toxicities were noted at the highest dose levels and the MTD was not reached. No objective responses were seen. Stable disease occurred in 9 patients (29%) and the median overall survival was 25.7 weeks (95% confidence interval [CI], 19.1-30.6). Pharmacokinetic analysis confirmed an estimated half life of 6.5 days. The recommended phase 2 dose of GT103 was 10 mg/kg every 3 weeks, however further dose optimization is needed given the absence of an MTD. The study achieved its primary objective of demonstrating safety and tolerability of GT103 in refractory NSCLC.
Type
Department
Description
Provenance
Subjects
Citation
Permalink
Published Version (Please cite this version)
Publication Info
Clarke, Jeffrey M, George R Simon, Hirva Mamdani, Lin Gu, James E Herndon, Thomas E Stinchcombe, Neal Ready, Jeffrey Crawford, et al. (2025). Complement factor H targeting antibody GT103 in refractory non-small cell lung cancer: a phase 1b dose escalation trial. Nature communications, 16(1). p. 93. 10.1038/s41467-024-55092-2 Retrieved from https://hdl.handle.net/10161/32057.
This is constructed from limited available data and may be imprecise. To cite this article, please review & use the official citation provided by the journal.
Collections
Scholars@Duke

Jeffrey Melson Clarke

James Emmett Herndon
Current research interests have application to the design and analysis of cancer clinical trials. Specifically, interests include the use of time-dependent covariables within survival models, the design of phase II cancer clinical trials which minimize some of the logistical problems associated with their conduct, and the analysis of longitudinal studies with informative censoring (in particular, quality of life studies of patients with advanced cancer).

Neal Edward Ready

Jeffrey Crawford
- Lung cancer/new treatment approaches.
2. Clinical trials of hematopoietic growth factors, biological agents and targeted drug development.
3. Cancer in the elderly and supportive care
Accomplishments
1. Lead Investigator of the U. S. multicenter, randomized trial of Filgrastim (G-CSF, Neupogen) to reduce the morbidity of chemotherapy-related neutropenia, leading to FDA approval 2/91.
2. Lead Investigator of the U. S. multicenter, randomized trial of Vinorelbine (Navelbine) in treatment of patients with advanced non small cell carcinoma of lung (NSCLC), leading to FDA approval 12/94.
3. Principal Investigator in initial phase I clinical trials of stem cell factor (SCF), megakaryocyte growth and development factor (MGDF), pegylated granulocyte-colony-stimulating factor and other novel hematopoietic growth factors.

Stephen Joseph Balevic
I am an Adult and Pediatric Rheumatologist and care for patients with a wide variety of autoimmune and rheumatic diseases, including: systemic lupus, rheumatoid arthritis, juvenile arthritis, vasculitis, and sarcoidosis, among others. I have a special interest in using musculoskeletal ultrasound to optimize diagnosis and treatment decisions at the bedside.
I am also a clinical researcher at the Duke Clinical Research Institute (DCRI). My research interests are in clinical trials and precision medicine through population pharmacokinetic/pharmacodynamic modeling. I obtained my PhD in Pharmaceutical Sciences from the UNC Eshelman School of Pharmacy. I serve as the principal investigator on several grants studying hydroxychloroquine and azathioprine pharmacokinetics and exposure-response in lupus, as well as principal investigator or co-investigator for several clinical trials at the DCRI. Additionally, I am an Assistant Scientific Director for the Childhood Arthritis and Rheumatology Research Alliance (CARRA) Registry- the largest registry of children with rheumatic diseases in North America.

Andrew Benjamin Nixon
Dr. Andrew Nixon is Professor of Medicine in the Department of Medicine/Division of Medical Oncology at Duke University. He holds a BS in chemistry from Miami University, a PhD in biochemistry from Wake Forest University, and an MBA with a focus on healthcare management from Duke University/Fuqua School of Business. He is a nationally recognized expert in the development of cancer biomarkers and correlative science, with extensive experience leading large multi-center biomarker analyses. Dr. Nixon serves in various leadership roles within the National Cancer Institute (NCI), working with diverse multi-disciplinary teams focused on improving cancer patient outcomes through biomarker science. He serves as the national co-chair for the NCI Core Correlative Sciences Committee which adjudicates the use of biospecimens collected throughout the NCI National Clinical Trial Network (NCTN). Within the NCI-NCTN Alliance cooperative group, Dr. Nixon has multiple leadership positions including serving on the Alliance Board of Directors, co-chair of the Immuno-Oncology Committee, co-chair for Gastrointestinal Correlative Research, and has been an executive member of the Translational Research Program since its inception. Additionally, within the NCI-NCTN NRG cooperative group, he serves as a member of the Gynecologic Translational Science Committee and on the Gynecological Oncology Group - Partners Investigator Council Translational Research Subcommittee. Dr. Nixon is an American Society for Clinical Oncology (ASCO) Ambassador and has chaired various committees and led scientific and educational sessions at national conferences. Recently, Dr. Nixon has focused his research on cellular senescence and biomarkers of aging and early carcinogenesis. He serves as Principal Investigator for a large multi-center NIH grant to develop high-resolution tissue maps and biomarkers of cellular senescence as a part of the Senescence Network (SenNet) Consortium.

Huihua Li

You-Wen He
We study T cell biology in health and disease. Our current study is divided into two parts. Part I is to investigate T lymphocyte-mediated anti-caner immunity. We have found that host complement inhibits the cytokine IL-10 production in CD8+ tumor infiltrating lymphocytes through complement receptors C3aR and C5aR. Complement-deficient animals are resistant to tumor development in a T cell- and IL-10-dependent manner. CD8+ tumor infiltrating T cells express IL-10 when complement signaling is disabled. We found that tumor infiltrating lymphocytes from human cancers expanded with IL-2 plus IL-10 are potent tumor killers. Complement-mediated inhibition on antitumor immunity is independent of the PD-1/PD-L1 immune checkpoint pathway. Our findings suggest that complement receptors C3aR and C5aR expressed on CD8+ tumor infiltrating lymphocytes represent a novel class of immune checkpoints that needs to be targeted for tumor immunotherapy. Our current effort is to enhance cancer immunotherapy through several strategies. First, we investigate a combined blockade of complement signaling and anti-PD-1 to enhance the antitumor efficacy; second, we are studying the antitumor efficacy of a targeted delivery of IL-10 to antitumor CD8+ T cells by using anti-PD1-IL-10 or anti-CTLA-4-IL-10 fusion proteins; third, we are studying the tumor killing efficacy of addition of IL-10 in the expansion protocol of tumor infiltrating lymphocytes for adaptive cellular therapy.
Part II is to investigate the intracellular process termed autophagy in T lymphocyte function. Autophagy is a highly conserved self-digestion pathway that plays essential roles in maintaining the homeostasis of organelles, degrading long-lived proteins and recycling amino acids under starvation conditions. We have found that autophagy related molecules are expressed in T lymphocytes and autophagy occurs inside T lymphocytes. We have generated autophagy-deficient T lymphocytes in multiple genetic models and investigated the roles of autophagy in T lymphocytes. We found that autophagy plays a critical role in T lymphocyte function. Our current effort is to elucidate the molecular pathways by which TCR signal induces autophagy and the impact of autophagy on intracellular organelle homeostasis in dividing T cells.
Unless otherwise indicated, scholarly articles published by Duke faculty members are made available here with a CC-BY-NC (Creative Commons Attribution Non-Commercial) license, as enabled by the Duke Open Access Policy. If you wish to use the materials in ways not already permitted under CC-BY-NC, please consult the copyright owner. Other materials are made available here through the author’s grant of a non-exclusive license to make their work openly accessible.