1
|
Song MY, Cho J, Park H, Song Y, Kim K, Ahn JH, Lee CM, Kim DH, Ko HJ. Discovery and functional characterization of canine PD-L1-targeted antibodies for evaluating antitumor efficacy in a canine osteosarcoma xenograft model. Sci Rep 2025; 15:7574. [PMID: 40038403 PMCID: PMC11880529 DOI: 10.1038/s41598-025-90770-1] [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: 09/02/2024] [Accepted: 02/17/2025] [Indexed: 03/06/2025] Open
Abstract
Targeting the programmed cell death protein 1 (PD-1)/programmed death ligand 1 (PD-L1) pathway is promising in treating cancer in humans and offers potential for veterinary applications. However, no PD-L1 antibodies have been approved specifically for treating canine cancer. We aimed to develop PD-L1-specific antibodies using phage display technology for treating canine cancer. A synthetic antibody library was screened, and 18 high-affinity single-chain variable fragment clones were subsequently converted to the IgG format for enhancing binding affinity and functional stability. The clone #15 exhibited the highest binding affinity and most pronounced antitumor effects. The PD-1/PD-L1 interaction was inhibited by antibody #15. The binding and thermal stabilities of the antibodies were validated by flow cytometry and thermal stability assays, respectively. In NOG mice xenografted with canine osteosarcoma cells and treated with canine peripheral blood mononuclear cells and antibody #15, the tumor size and weight were reduced. Antibody #15 significantly increased apoptosis of tumor cells and lymphocyte populations. Therefore, anti-PD-L1 antibodies, particularly antibody #15, have substantial potential as novel immunotherapeutic agents against canine osteosarcoma. This study represents a significant advancement in veterinary oncology, with the potential of improving treatment outcomes for canine cancers and providing insights into similar strategies in human cancer therapy.
Collapse
Affiliation(s)
- Min-Young Song
- Department of Pharmacy, Kangwon National University, Chuncheon, 24341, Republic of Korea
- Innovative Drug Development Research Team for Intractable Diseases (BK21 Plus), Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Jaewon Cho
- Department of Pharmacy, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Hyosung Park
- Department of Pharmacy, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Yujeong Song
- Department of Pharmacy, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Keon Kim
- Department of Veterinary Internal Medicine, College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Jae-Hee Ahn
- Department of Pharmacy, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Chang-Min Lee
- Department of Veterinary Internal Medicine, College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Dae Hee Kim
- Department of Pharmacy, Kangwon National University, Chuncheon, 24341, Republic of Korea.
- Kangwon Institute of Inclusive Technology, Kangwon National University, Chuncheon, 24341, Republic of Korea.
- Global/Gangwon Innovative Biologics-Regional Leading Research Center (GIB-RLRC), Kangwon National University, Chuncheon, 24341, Republic of Korea.
| | - Hyun-Jeong Ko
- Department of Pharmacy, Kangwon National University, Chuncheon, 24341, Republic of Korea.
- Kangwon Institute of Inclusive Technology, Kangwon National University, Chuncheon, 24341, Republic of Korea.
- Innovative Drug Development Research Team for Intractable Diseases (BK21 Plus), Kangwon National University, Chuncheon, 24341, Republic of Korea.
- Global/Gangwon Innovative Biologics-Regional Leading Research Center (GIB-RLRC), Kangwon National University, Chuncheon, 24341, Republic of Korea.
| |
Collapse
|
2
|
Cho J, Tae N, Song Y, Kim CW, Lee SJ, Ahn JH, Lee KH, Lee BH, Kim BS, Chang SY, Kim DH, Ko HJ. The expression of PD-L1 on tumor-derived exosomes enhances infiltration and anti-tumor activity of αCD3 × αPD-L1 bispecific antibody-armed T cells. Cancer Immunol Immunother 2024; 73:196. [PMID: 39105814 PMCID: PMC11303351 DOI: 10.1007/s00262-024-03785-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Accepted: 07/17/2024] [Indexed: 08/07/2024]
Abstract
Anti-cluster of differentiation (CD) 3 × α programmed death-ligand 1 (PD-L1) bispecific T-cell engager (BsTE)-bound T-cells (BsTE:T) are a promising new cancer treatment agent. However, the mechanisms of action of bispecific antibody-armed activated T-cells are poorly understood. Therefore, this study aimed to investigate the anti-tumor mechanism and efficacy of BsTE:T. The BsTE:T migration was assessed in vivo and in vitro using syngeneic and xenogeneic tumor models, flow cytometry, immunofluorescence staining, transwell migration assays, microfluidic chips, Exo View R100, western blotting, and clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated protein 9 technology. In murine B16 melanoma, MC38 colon cancer, and human multiple myeloma cells, BsTE:T exhibited superior tumor elimination relative to that of T-cells or BsTE alone. Moreover, BsTE:T migration into tumors was significantly enhanced owing to the presence of PD-L1 in tumor cells and secretion of PD-L1-containing exosomes. Furthermore, increased infiltration of CD44highCD62Llow effector memory CD8+ T-cells into tumors was closely associated with the anti-tumor effect of BsTE:T. Therefore, BsTE:T is an innovative potential anti-tumor therapy, and exosomal PD-L1 plays a crucial role both in vitro and in vivo in the anti-tumor activity of BsTE:T.
Collapse
Affiliation(s)
- Jaewon Cho
- Department of Pharmacy, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Nara Tae
- Kangwon Institute of Inclusive Technology, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Yujeong Song
- Department of Pharmacy, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Chae-Won Kim
- Department of Pharmacy, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Seung-Joo Lee
- Department of Pharmacy, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Jae-Hee Ahn
- Department of Pharmacy, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Kwang-Ho Lee
- Department of Advanced Material Science and Engineering, College of Engineering, Kangwon National University, Chuncheon, 25561, Korea
| | - Byung-Hyun Lee
- Department of Internal Medicine, Korea University College of Medicine, Seoul, 02841, Republic of Korea
| | - Byung Soo Kim
- Department of Internal Medicine, Korea University College of Medicine, Seoul, 02841, Republic of Korea
| | - Sun-Young Chang
- Laboratory of Microbiology, College of Pharmacy, and Research Institute of Pharmaceutical Science and Technology (RIPST), Ajou University, Suwon, 16499, Korea
| | - Dae Hee Kim
- Department of Pharmacy, Kangwon National University, Chuncheon, 24341, Republic of Korea.
- Kangwon Institute of Inclusive Technology, Kangwon National University, Chuncheon, 24341, Republic of Korea.
- Global/Gangwon Innovative Biologics-Regional Leading Research Center (GIB-RLRC), Kangwon National University, Chuncheon, 24341, Republic of Korea.
| | - Hyun-Jeong Ko
- Department of Pharmacy, Kangwon National University, Chuncheon, 24341, Republic of Korea.
- Kangwon Institute of Inclusive Technology, Kangwon National University, Chuncheon, 24341, Republic of Korea.
- Innovative Drug Development Research Team for Intractable Diseases (BK21 plus), Kangwon National University, Chuncheon, 24341, Republic of Korea.
- Global/Gangwon Innovative Biologics-Regional Leading Research Center (GIB-RLRC), Kangwon National University, Chuncheon, 24341, Republic of Korea.
| |
Collapse
|
3
|
Kiem D, Ocker M, Greil R, Neureiter D, Melchardt T. Enhancing anti-CD274 (PD-L1) targeting through combinatorial immunotherapy with bispecific antibodies and fusion proteins: from preclinical to phase II clinical trials. Expert Opin Investig Drugs 2024; 33:229-242. [PMID: 38354028 DOI: 10.1080/13543784.2024.2319317] [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: 12/03/2023] [Accepted: 02/12/2024] [Indexed: 02/24/2024]
Abstract
INTRODUCTION Immune checkpoint inhibitors have achieved great success in the treatment of many different types of cancer. Programmed cell death protein ligand 1 (PD-L1, CD274) is a major immunosuppressive immune checkpoint and a target for several already approved monoclonal antibodies. Despite this, novel strategies are under development, as the overall response remains low. AREAS COVERED In this review, an overview of the current biomarkers for response to PD-L1 inhibitor treatment is given, followed by a discussion of potential novel biomarkers, including tumor mutational burden and circulating tumor DNA. Combinatorial immunotherapy is a potential novel strategy to increase the response to PD-L1 inhibitor treatment and currently, several interesting bispecific antibodies as well as bispecific fusion proteins are undergoing early clinical investigation. We focus on substances targeting PD-L1 and a secondary target, and a secondary immunomodulatory target like CTLA-4, TIGIT, or CD47. EXPERT OPINION Overall, the presented studies show anti-tumor activity of these combinatorial immunotherapeutic approaches. However, still relatively low response rates suggest a need for better biomarkers.
Collapse
Affiliation(s)
- Dominik Kiem
- III Medical Department, Paracelsus Medical University, Salzburg, Austria
| | - Matthias Ocker
- Medical Department, Division of Hematology, Oncology, and Cancer Immunology, Campus, Charité Mitte, Charité University Medicine Berlin, Berlin, Germany
- EO Translational Insights Consulting GmbH, Berlin, Germany
- Tacalyx GmbH, Berlin, Germany
| | - Richard Greil
- III Medical Department, Paracelsus Medical University, Salzburg, Austria
- Cancer Cluster Salzburg, Salzburg, Austria
| | - Daniel Neureiter
- Cancer Cluster Salzburg, Salzburg, Austria
- Institute of Pathology, Paracelsus Medical University, University Hospital Salzburg (SALK), Salzburg, Austria
| | - Thomas Melchardt
- III Medical Department, Paracelsus Medical University, Salzburg, Austria
- Cancer Cluster Salzburg, Salzburg, Austria
| |
Collapse
|
4
|
Duwa R, Pokhrel RH, Banstola A, Pandit M, Shrestha P, Jeong JH, Chang JH, Yook S. T-cell engaging poly(lactic-co-glycolic acid) nanoparticles as a modular platform to induce a potent cytotoxic immunogenic response against PD-L1 overexpressing cancer. Biomaterials 2022; 291:121911. [DOI: 10.1016/j.biomaterials.2022.121911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 10/19/2022] [Accepted: 11/10/2022] [Indexed: 11/15/2022]
|