1
|
Arias-Badia M, Pai CCS, Lwin YM, Chen P, Srinath A, Tanaka M, Musser E, Goodearl A, Gorman JV, Ritacco W, Fong L. Impact of tumor localization on antitumor immunity with conditionally activated CTLA-4 blockade. J Immunother Cancer 2025; 13:e010566. [PMID: 40180419 PMCID: PMC11966968 DOI: 10.1136/jitc-2024-010566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Accepted: 03/17/2025] [Indexed: 04/05/2025] Open
Abstract
BACKGROUND Immune checkpoint blockade (ICB) can induce antitumor efficacy but also induces immune-related adverse events. Systemically administered ICB can activate immune cells throughout the host. Conditionally active ICB with proteolytically cleaved masking domains can potentially reduce the adverse events seen with anti-cytotoxic T-lymphocyte associated protein 4 (CTLA-4) antibody. METHODS We examined how different formats of a conditionally activated dual variable domain IgG (DVD) that binds CTLA-4 and the tumor-associated antigen prostate stem cell antigen (PSCA) can lead to efficacy in syngeneic subcutaneous and metastatic murine tumor models. We also defined the capacity of these DVDs to modulate immune responses by multiparameter flow cytometry. RESULTS Conditionally active DVDs can uncouple antitumor efficacy from toxicity. A fully cleavable construct (symmetric DVD, sDVD), which can be released from the target tumor cells, showed superior antitumor efficacy compared with asymmetric DVD, which retains its tumor antigen binding. The sDVD elicited the highest tumor-antigen-specific T-cell responses detected in tumors and tumor-draining lymph nodes, as well as presenting the highest rate of intratumoral and splenic "non-exhausted" antigen-specific CD8 T cells. SDVD also induced the highest degrees of T-cell memory and self-renewal potential. These effects were dependent on PSCA expression by the tumors. CONCLUSIONS These findings support the notion that ICB modulation of antitumor immunity away from the tumor cells is critically important for optimal antitumor immunity. The bispecific sDVD antibody design may enable improved systemic antitumor responses than traditional ICB in both primary tumors and metastases.
Collapse
Affiliation(s)
- Marcel Arias-Badia
- Department of Medicine, University of California San Francisco, San Francisco, California, USA
| | - Chien-Chun Steven Pai
- Department of Medicine, University of California San Francisco, San Francisco, California, USA
| | - Yee May Lwin
- Department of Medicine, University of California San Francisco, San Francisco, California, USA
| | - PeiXi Chen
- Department of Medicine, University of California San Francisco, San Francisco, California, USA
| | - Aahir Srinath
- Department of Medicine, University of California San Francisco, San Francisco, California, USA
| | - Miho Tanaka
- Department of Medicine, University of California San Francisco, San Francisco, California, USA
| | - Emily Musser
- AbbVie Bioresearch Center, Worcester, Massachusetts, USA
| | | | | | - Wendy Ritacco
- AbbVie Bioresearch Center, Worcester, Massachusetts, USA
| | - Lawrence Fong
- Department of Medicine, University of California San Francisco, San Francisco, California, USA
- Immunology Integrated Research Center, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| |
Collapse
|
2
|
Drum DL, Jallorina AG, Wan LS, Chang VT, Lee-Wong MF. Non-Genetic Biomarkers in Merkel Cell Carcinoma: Prognostic Implications and Predictive Utility for Response to Anti-PD-(L)1 Immune Checkpoint Inhibitors. Exp Dermatol 2025; 34:e70030. [PMID: 39791602 DOI: 10.1111/exd.70030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 11/11/2024] [Accepted: 12/30/2024] [Indexed: 01/12/2025]
Abstract
Merkel cell carcinoma (MCC) is a skin cancer that arises due to either Merkel cell polyomavirus infection (MCPyV) or ultraviolet (UV) radiation exposure, presenting primarily in the head and neck region of fair-skinned males. The recent success of PD-(L)1 immune checkpoint inhibitors (ICIs) in locally advanced/metastatic MCC, with an objective response rate (ORR) around 50% and improved survival, as a first-line treatment has moved ICIs to the forefront of therapy for MCC and generated interest in identifying biomarkers to predict clinical response. The MCC tumour microenvironment (TME) contains various components of the adaptive and innate immune system. These components can contribute to tumour immune escape through immunosuppression by preventing entrance of other immune cells or by aiding in the cytotoxic clearance of tumour cells. We aim to combine information from studies of baseline and on-treatment monitoring of the TME to help predict the success of ICIs in MCC. This review enhances the understanding of how CD8 T cells, γδ T cells and macrophages may impact predictions of response rates to ICIs in MCC patients. These immune cells are non-genetic biomarkers that can also be used to determine prognosis in MCC treatment.
Collapse
Affiliation(s)
- David L Drum
- Department of Medicine, California University of Science and Medicine, Colton, California, USA
| | - Anika G Jallorina
- Department of Medicine, California University of Science and Medicine, Colton, California, USA
| | - Leo S Wan
- Department of Medicine, West Virginia School of Osteopathic Medicine, Lewisburg, West Virginia, USA
| | - Victor T Chang
- Department of Medicine, Hematology/Oncology, Rutgers New Jersey School of Medicine, Newark, New Jersey, USA
- Section of Hematology/Oncology, Veterans Administration New Jersey Health Care System, East Orange, New Jersey, USA
| | - Mary F Lee-Wong
- Department of Medicine and Division of Clinical Immunology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Division of Allergy and Immunology, Maimonides Medical Center, Brooklyn, New York, USA
| |
Collapse
|
3
|
Shen S, Hong Y, Huang J, Qu X, Sooranna SR, Lu S, Li T, Niu B. Targeting PD-1/PD-L1 in tumor immunotherapy: Mechanisms and interactions with host growth regulatory pathways. Cytokine Growth Factor Rev 2024; 79:16-28. [PMID: 39179486 DOI: 10.1016/j.cytogfr.2024.08.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 08/06/2024] [Accepted: 08/07/2024] [Indexed: 08/26/2024]
Abstract
Tumor immunotherapy has garnered considerable attention, emerging as a new standard of care in cancer treatment. The conventional targets, such as VEGF and EGFR, have been extended to others including BRAF and PD-1/PD-L1, which have shown significant potential in recent cancer treatments. This review aims to succinctly overview the impact and mechanisms of therapies that modulate PD-1/PD-L1 expression by targeting VEGF, EGFR, LAG-3, CTLA-4 and BRAF. We investigated how modulation of PD-1/PD-L1 expression impacts growth factor signaling, shedding light on the interplay between immunomodulatory pathways and growth factor networks within the tumor microenvironment. By elucidating these interactions, we aim to provide insights into novel potential synergistic therapeutic strategies for cancer immunotherapy.
Collapse
Affiliation(s)
- Songyu Shen
- School of life Science, Shanghai University, 99 Shangda Road, 200444, China
| | - Yihan Hong
- School of life Science, Shanghai University, 99 Shangda Road, 200444, China
| | - Jiajun Huang
- School of life Science, Shanghai University, 99 Shangda Road, 200444, China
| | - Xiaosheng Qu
- Guangxi Botanical Garden of Medicinal Plants, Nanning, Guangxi 530023, China
| | - Suren Rao Sooranna
- Department of Metabolism, Digestion and Reproduction, Imperial College London, 369 Fulham Road, London SW10 9NH, United Kingdom
| | - Sheng Lu
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
| | - Tian Li
- School of Basic Medicine, Fourth Military Medical University, 169 Changle West Rd, Xi'an 710032, China.
| | - Bing Niu
- School of life Science, Shanghai University, 99 Shangda Road, 200444, China.
| |
Collapse
|
4
|
Roskoski R. Combination immune checkpoint and targeted protein kinase inhibitors for the treatment of renal cell carcinomas. Pharmacol Res 2024; 203:107181. [PMID: 38614375 DOI: 10.1016/j.phrs.2024.107181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 04/08/2024] [Indexed: 04/15/2024]
Abstract
Kidney cancers comprise about 3% of all new malignancies in the United States. Renal cell carcinomas (RCCs) are the most common type of renal malignancy making up about 85% of kidney cancer cases. Signs and symptoms of renal cell carcinomas can result from local tumor growth, paraneoplastic syndromes, or distant metastases. The classic triad of presentation with flank pain, hematuria, and a palpable abdominal mass occurs in fewer than 10% of patients. Most diagnoses result from incidental imaging findings (ultrasonography or abdominal CT imaging) performed for another reason. Localized disease is treated by partial nephrectomy, total nephrectomy, or ablation (tumor destruction with heat or cold). When the tumors have metastasized, systemic therapy with protein-tyrosine kinase antagonists including sorafenib, sunitinib, pazopanib, and tivozanib that target vascular endothelial, platelet-derived, fibroblast, hepatocyte, and stem cell factor growth factor receptors (VEGFR, PDGFR, FGFR, MET, and Kit) were prescribed after 2005. The monoclonal antibody immune checkpoint inhibitor nivolumab (targeting programed cell death protein 1, PD1) was approved for the treatment of RCCs in 2015. It is usually used now in combination with ipilimumab (targeting CTLA-4) or cabozantinib (a multikinase blocker). Other combination therapies include pembrolizumab (targeting PD1) and axitinib (a VEGFR and PDGFR blocker) or lenvatinib (a multikinase inhibitor). Since the KEYNOTE-426 clinical trial, the use of immune checkpoint inhibitors in combination with protein-tyrosine kinase inhibitors is now the standard of care for most patients with metastatic renal cell carcinomas and monotherapies are used only in those individuals who cannot receive or tolerate immune checkpoint inhibitors.
Collapse
Affiliation(s)
- Robert Roskoski
- Blue Ridge Institute for Medical Research, 221 Haywood Knolls Drive, Hendersonville, NC 28791, United States.
| |
Collapse
|
5
|
Spiliopoulou P, Kaur P, Hammett T, Di Conza G, Lahn M. Targeting T regulatory (T reg) cells in immunotherapy-resistant cancers. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2024; 7:2. [PMID: 38318526 PMCID: PMC10838381 DOI: 10.20517/cdr.2023.46] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Revised: 12/11/2023] [Accepted: 01/09/2024] [Indexed: 02/07/2024]
Abstract
Primary or secondary (i.e., acquired) resistance is a common occurrence in cancer patients and is often associated with high numbers of T regulatory (Treg) cells (CD4+CD25+FOXP3+). The approval of ipilimumab and the development of similar pharmacological agents targeting cell surface proteins on Treg cells demonstrates that such intervention may overcome resistance in cancer patients. Hence, the clinical development and subsequent approval of Cytotoxic T Lymphocyte Antigen-4 (CTLA-4) targeting agents can serve as a prototype for similar agents. Such new agents aspire to be highly specific and have a reduced toxicity profile while increasing effector T cell function or effector T/T regulatory (Teff/Treg) ratio. While clinical development with large molecules has shown the greatest advancement, small molecule inhibitors that target immunomodulation are increasingly entering early clinical investigation. These new small molecule inhibitors often target specific intracellular signaling pathways [e.g., phosphoinositide-3-kinase delta (PI3K-δ)] that play an important role in regulating the function of Treg cells. This review will summarize the lessons currently applied to develop novel clinical agents that target Treg cells.
Collapse
Affiliation(s)
- Pavlina Spiliopoulou
- Department of Drug Development Program, Phase I Unit, Beatson West of Scotland Cancer Center, Glasgow G12 0YN, UK
- School of Cancer Sciences, University of Glasgow, Glasgow G61 1BD, UK
| | - Paramjit Kaur
- Department of Oncology Clinical Development, iOnctura SA, Geneva 1202, Switzerland
| | - Tracey Hammett
- Department of Oncology Clinical Development, iOnctura SA, Geneva 1202, Switzerland
| | - Giusy Di Conza
- Department of Oncology Clinical Development, iOnctura SA, Geneva 1202, Switzerland
| | - Michael Lahn
- Department of Oncology Clinical Development, iOnctura SA, Geneva 1202, Switzerland
| |
Collapse
|