Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters










Database
Language
Publication year range
1.
Cancer Res Commun ; 3(8): 1564-1579, 2023 08.
Article in English | MEDLINE | ID: mdl-37593752

ABSTRACT

In recent years, there has been considerable interest in mAb-based induction of costimulatory receptor signaling as an approach to combat cancer. However, promising nonclinical data have yet to translate to a meaningful clinical benefit. Inducible T-cell costimulator (ICOS) is a costimulatory receptor important for immune responses. Using a novel clinical-stage anti-ICOS immunoglobulin G4 mAb (feladilimab), which induces but does not deplete ICOS+ T cells and their rodent analogs, we provide an end-to-end evaluation of the antitumor potential of antibody-mediated ICOS costimulation alone and in combination with programmed cell death protein 1 (PD-1) blockade. We demonstrate, consistently, that ICOS is expressed in a range of cancers, and its induction can stimulate growth of antitumor reactive T cells. Furthermore, feladilimab, alone and with a PD-1 inhibitor, induced antitumor activity in mouse and humanized tumor models. In addition to nonclinical evaluation, we present three patient case studies from a first-time-in-human, phase I, open-label, dose-escalation and dose-expansion clinical trial (INDUCE-1; ClinicalTrials.gov: NCT02723955), evaluating feladilimab alone and in combination with pembrolizumab in patients with advanced solid tumors. Preliminary data showing clinical benefit in patients with cancer treated with feladilimab alone or in combination with pembrolizumab was reported previously; with example cases described here. Additional work is needed to further validate the translation to the clinic, which includes identifying select patient populations that will benefit from this therapeutic approach, and randomized data with survival endpoints to illustrate its potential, similar to that shown with CTLA-4 and PD-1 blocking antibodies. Significance: Stimulation of the T-cell activation marker ICOS with the anti-ICOS agonist mAb feladilimab, alone and in combination with PD-1 inhibition, induces antitumor activity across nonclinical models as well as select patients with advanced solid tumors.


Subject(s)
Ambulatory Care Facilities , Antibodies, Monoclonal , Humans , Animals , Mice , Antibodies, Monoclonal/pharmacology , Immune Checkpoint Inhibitors , Immunoglobulin G , Inhibition, Psychological
2.
Mol Cancer Ther ; 20(10): 1941-1955, 2021 10.
Article in English | MEDLINE | ID: mdl-34253590

ABSTRACT

B-cell maturation antigen (BCMA) is an attractive therapeutic target highly expressed on differentiated plasma cells in multiple myeloma and other B-cell malignancies. GSK2857916 (belantamab mafodotin, BLENREP) is a BCMA-targeting antibody-drug conjugate approved for the treatment of relapsed/refractory multiple myeloma. We report that GSK2857916 induces immunogenic cell death in BCMA-expressing cancer cells and promotes dendritic cell activation in vitro and in vivo GSK2857916 treatment enhances intratumor immune cell infiltration and activation, delays tumor growth, and promotes durable complete regressions in immune-competent mice bearing EL4 lymphoma tumors expressing human BCMA (EL4-hBCMA). Responding mice are immune to rechallenge with EL4 parental and EL4-hBCMA cells, suggesting engagement of an adaptive immune response, immunologic memory, and tumor antigen spreading, which are abrogated upon depletion of endogenous CD8+ T cells. Combinations with OX40/OX86, an immune agonist antibody, significantly enhance antitumor activity and increase durable complete responses, providing a strong rationale for clinical evaluation of GSK2857916 combinations with immunotherapies targeting adaptive immune responses, including T-cell-directed checkpoint modulators.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacology , B-Cell Maturation Antigen/antagonists & inhibitors , CD8-Positive T-Lymphocytes/immunology , Immunoconjugates/pharmacology , Immunogenic Cell Death , Lymphoma/drug therapy , Multiple Myeloma/drug therapy , Animals , Antibodies, Monoclonal/chemistry , Apoptosis , B-Cell Maturation Antigen/immunology , Cell Proliferation , Female , Humans , Lymphoma/immunology , Lymphoma/metabolism , Lymphoma/pathology , Mice , Mice, Inbred C57BL , Multiple Myeloma/immunology , Multiple Myeloma/metabolism , Multiple Myeloma/pathology , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
3.
Oncotarget ; 7(26): 39861-39871, 2016 Jun 28.
Article in English | MEDLINE | ID: mdl-27223434

ABSTRACT

Fibroblast growth factor (FGF) ligand-dependent signaling has a fundamental role in cancer development and tumor maintenance. GSK3052230 (also known as FP-1039) is a soluble decoy receptor that sequesters FGFs and inhibits FGFR signaling. Herein, the efficacy of this molecule was tested in models of mesothelioma, a tumor type shown to express high levels of FGF2 and FGFR1. GSK3052230 demonstrated antiproliferative activity across a panel of mesothelioma cell lines and inhibited growth of tumor xenografts in mice. High expression of FGF2 and FGFR1 correlated well with response to FGF pathway inhibition. GSK3052230 inhibited MAPK signaling as evidenced by decreased phospho-ERK and phospho-S6 levels in vitro and in vivo. Additionally, dose-dependent and statistically-significant reductions in tumor vessel density were observed in GSK3052230-treated tumors compared to vehicle-treated tumors. These data support the role of GSK3052230 in effectively targeting FGF-FGFR autocrine signaling in mesothelioma, demonstrate its impact on tumor growth and angiogenesis, and provide a rationale for the current clinical evaluation of this molecule in mesothelioma patients.


Subject(s)
Fibroblast Growth Factors/metabolism , Mesothelioma/metabolism , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Signal Transduction , Animals , Autocrine Communication , Cell Line , Cell Proliferation , Female , Gene Expression Regulation, Neoplastic , Humans , Immunoglobulin G/chemistry , Ligands , Magnetic Resonance Imaging , Mice , Mice, SCID , Neoplasm Transplantation , Neovascularization, Pathologic , Oncogene Proteins, Fusion/chemistry , Receptor, Fibroblast Growth Factor, Type 1/chemistry , Recombinant Fusion Proteins
SELECTION OF CITATIONS
SEARCH DETAIL