1
|
Wu S, Jiang H, Fang Z, Wu Y, Jiao J, Fang W, Wu Y, Lang Y, Chen N, Zhong Z, Chen L, Zheng X, Lu B, Jiang J. Enhanced abscopal anti-tumor response via a triple combination of thermal ablation, IL-21, and PD-1 inhibition therapy. Cancer Immunol Immunother 2024; 73:138. [PMID: 38833177 PMCID: PMC11150342 DOI: 10.1007/s00262-024-03718-1] [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: 02/19/2024] [Accepted: 04/24/2024] [Indexed: 06/06/2024]
Abstract
Despite the success of immune checkpoint inhibitors (ICIs) in treating solid tumors, lots of patients remain unresponsive to this therapy. Microwave ablation (MWA) stimulates systemic adaptive immunity against tumor cells by releasing tumor antigens. Additionally, IL-21 has demonstrated importance in stimulating T-cell effector function. The combination of these three therapies-MWA, IL-21, and anti-PD-1 monoclonal antibodies (mAbs)-has yet to be explored in the context of cancer treatment.In this study, we explored the impact of thermal ablation on IL-21R expression in tumor-infiltrating lymphocytes (TILs). Subsequently, we assessed alterations in the tumor microenvironment (TME) and peripheral lymphoid organs. Additionally, we conducted a thorough examination of tumor-infiltrating CD45+ immune cells across various treatment groups using single-cell RNA sequencing (scRNA-seq). Moreover, we determined the potential anti-tumor effects of the triple combination involving MWA, IL-21, and anti-PD-1 mAbs.Our findings revealed that MWA upregulated the expression of IL-21R on various immune cells in the untreated tumors. The combination of MWA with IL-21 exhibited a robust abscopal anti-tumor effect, enhancing the effector function of CD8+ T cells and facilitating dendritic cells' maturation and antigen presentation in the untreated tumor. Notably, the observed abscopal anti-tumor effect resulting from the combination is contingent upon T-cell recirculation, indicating the reliance of systemic adaptive immunity for this treatment regimen. Additionally, the combination of MWA, IL-21, and PD-1 mAbs demonstrated profound abscopal anti-tumor efficacy. Our findings provide support for further clinical investigation into a triple combination therapy involving MWA, IL-21, and ICIs for the treatment of metastatic cancer.
Collapse
Affiliation(s)
- Shaoxian Wu
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, Jiangsu, China
- Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, Jiangsu, China
- Institute of Cell Therapy, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, Jiangsu, China
| | - Hongwei Jiang
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, Jiangsu, China
- Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, Jiangsu, China
- Institute of Cell Therapy, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, Jiangsu, China
| | - Zhang Fang
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, Jiangsu, China
- Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, Jiangsu, China
- Institute of Cell Therapy, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, Jiangsu, China
| | - You Wu
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, Jiangsu, China
- Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, Jiangsu, China
- Institute of Cell Therapy, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, Jiangsu, China
| | - Jing Jiao
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, Jiangsu, China
- Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, Jiangsu, China
- Institute of Cell Therapy, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, Jiangsu, China
| | - Weiwei Fang
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, Jiangsu, China
- Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, Jiangsu, China
- Institute of Cell Therapy, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, Jiangsu, China
| | - Yue Wu
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, Jiangsu, China
- Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, Jiangsu, China
- Institute of Cell Therapy, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, Jiangsu, China
| | - Yanyan Lang
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, Jiangsu, China
- Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, Jiangsu, China
- Institute of Cell Therapy, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, Jiangsu, China
| | - Ning Chen
- Shanghai Junshi Biosciences Co.,Ltd., Shanghai, 201206, China
| | - Ziyang Zhong
- Anwita Biosciences Inc, San Carlos, CA, 94070, USA
| | - Lujun Chen
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, Jiangsu, China
- Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, Jiangsu, China
- Institute of Cell Therapy, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, Jiangsu, China
| | - Xiao Zheng
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, Jiangsu, China
- Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, Jiangsu, China
- Institute of Cell Therapy, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, Jiangsu, China
| | - Binfeng Lu
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ, 07110, USA
| | - Jingting Jiang
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, Jiangsu, China.
- Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, Jiangsu, China.
- Institute of Cell Therapy, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, Jiangsu, China.
| |
Collapse
|
3
|
Hou K, Xu X, Ge X, Jiang J, Ouyang F. Blockade of PD-1 and CTLA-4: A potent immunotherapeutic approach for hepatocellular carcinoma. Biofactors 2024; 50:250-265. [PMID: 37921427 DOI: 10.1002/biof.2012] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Accepted: 09/07/2023] [Indexed: 11/04/2023]
Abstract
Immune checkpoints (ICPs) can promote tumor growth and prevent immunity-induced cancer cell apoptosis. Fortunately, targeting ICPs, such as programmed cell death 1 (PD-1) or cytotoxic T lymphocyte associated protein 4 (CTLA-4), has achieved great success in the past few years and has gradually become an effective treatment for cancers, including hepatocellular carcinoma (HCC). However, many patients do not respond to ICP therapy due to acquired resistance and recurrence. Therefore, clarifying the specific mechanisms of ICP in the development of HCC is very important for enhancing the efficacy of anti-PD-1 and anti-CTLA-4 therapy. In particular, antigen presentation and interferon-γ (IFN-γ) signaling were reported to be involved in the development of resistance. In this review, we have explained the role and regulatory mechanisms of ICP therapy in HCC pathology. Moreover, we have also elaborated on combinations of ICP inhibitors and other treatments to enhance the antitumor effect. Collectively, recent advances in the pharmacological targeting of ICPs provide insights for the development of a novel alternative treatment for HCC.
Collapse
Affiliation(s)
- Kai Hou
- Clinical Research Center of the Second Affiliated Hospital, University of South China, Hengyang, Hunan, PR China
| | - Xiaohui Xu
- Department of Medicine of the Second Affiliated Hospital, University of South China, Hengyang, Hunan, PR China
| | - Xin Ge
- Clinical Research Center of the Second Affiliated Hospital, University of South China, Hengyang, Hunan, PR China
| | - Jiacen Jiang
- Department of Medicine of the Second Affiliated Hospital, University of South China, Hengyang, Hunan, PR China
| | - Fan Ouyang
- Department of Cardiology, Zhuzhou Hospital, the Affiliated Hospital of Xiangya Medical College of Central South University, Zhuzhou, Hunan, PR China
| |
Collapse
|
4
|
Zhao Y, Wang H, Jin L, Zhang Z, Liu L, Zhou M, Zhang X, Zhang L. Targeting fusion proteins of the interleukin family: A promising new strategy for the treatment of autoinflammatory diseases. Eur J Pharm Sci 2024; 192:106647. [PMID: 37984595 DOI: 10.1016/j.ejps.2023.106647] [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: 10/05/2023] [Accepted: 11/16/2023] [Indexed: 11/22/2023]
Abstract
As a means of communication between immune cells and non-immune cells, Interleukins (ILs) has the main functions of stimulating the proliferation and activation of inflammatory immune cells such as dendritic cells and lymphocytes, promote the development of blood cells and so on. However, dysregulation of ILs expression is a major feature of autoinflammatory diseases. The drugs targeting ILs or IL-like biologics have played an important role in the clinical treatment of autoinflammatory diseases. Nevertheless, the widespread use of IL products may result in significant off-target adverse reactions. Thus, there is a clear need to develop next-generation ILs products in the biomedical field. Fusion proteins are proteins created through the joining of two or more genes that originally coded for separate proteins. Over the last 30 years, there has been increasing interest in the use of fusion protein technology for developing anti-inflammatory drugs. In comparison to single-target drugs, fusion proteins, as multiple targets drugs, have the ability to enhance the cytokine therapeutic index, resulting in improved efficacy over classical drugs. The strategy of preparing ILs or their receptors as fusion proteins is increasingly used in the treatment of autoimmune and chronic inflammation. This review focuses on the efficacy of several fusion protein drugs developed with ILs or their receptors in the treatment of autoinflammatory diseases, in order to illustrate the prospects of this new technology as an anti-inflammatory drug development protocol in the future.
Collapse
Affiliation(s)
- Yuchen Zhao
- Institute of Clinical Pharmacology, Anhui Medical University, Hefei, Anhui 230032, China; Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Hefei, Anhui 230032, China; Anti-inflammatory Immune Drugs Collaborative Innovation Center, Hefei, Anhui 230032, China
| | - Han Wang
- Institute of Clinical Pharmacology, Anhui Medical University, Hefei, Anhui 230032, China; Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Hefei, Anhui 230032, China; Anti-inflammatory Immune Drugs Collaborative Innovation Center, Hefei, Anhui 230032, China
| | - Lin Jin
- Institute of Clinical Pharmacology, Anhui Medical University, Hefei, Anhui 230032, China; Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Hefei, Anhui 230032, China; Anti-inflammatory Immune Drugs Collaborative Innovation Center, Hefei, Anhui 230032, China
| | - Ziwei Zhang
- Institute of Clinical Pharmacology, Anhui Medical University, Hefei, Anhui 230032, China; Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Hefei, Anhui 230032, China; Anti-inflammatory Immune Drugs Collaborative Innovation Center, Hefei, Anhui 230032, China
| | - Lianghu Liu
- Institute of Clinical Pharmacology, Anhui Medical University, Hefei, Anhui 230032, China; Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Hefei, Anhui 230032, China; Anti-inflammatory Immune Drugs Collaborative Innovation Center, Hefei, Anhui 230032, China
| | - Mengqi Zhou
- Institute of Clinical Pharmacology, Anhui Medical University, Hefei, Anhui 230032, China; Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Hefei, Anhui 230032, China; Anti-inflammatory Immune Drugs Collaborative Innovation Center, Hefei, Anhui 230032, China
| | - Xianzheng Zhang
- Institute of Clinical Pharmacology, Anhui Medical University, Hefei, Anhui 230032, China; Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Hefei, Anhui 230032, China; Anti-inflammatory Immune Drugs Collaborative Innovation Center, Hefei, Anhui 230032, China.
| | - Lingling Zhang
- Institute of Clinical Pharmacology, Anhui Medical University, Hefei, Anhui 230032, China; Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Hefei, Anhui 230032, China; Anti-inflammatory Immune Drugs Collaborative Innovation Center, Hefei, Anhui 230032, China.
| |
Collapse
|
5
|
Abstract
Therapeutic fusion proteins are a class of hybrid constructs that combine distinct biomolecules into a single platform with the additive effects of the components. The ability to fuse two unrelated proteins provides a means to localize mechanisms to better treat a range of diseases. Fusion proteins can be designed to impart diverse functions, including increasing half-life, providing targeting, and enabling sustained signaling. Of these, half-life extenders, which are fused to a therapeutic protein to increase exposure, are the most established group of fusion proteins, with many clinical successes. Rapid advances in antibody and antibody-derivative technology have enabled the fusion of targeting domains with therapeutic proteins. An emerging group of therapeutic fusion proteins has two separate active functions. Although most research for therapeutic fusion proteins focuses on cancer, prior successes provide a foundation for studies into other diseases as well. The exponential emergence of biopharmaceuticals gives precedence for increased research into therapeutic fusion proteins for a multitude of diseases.
Collapse
Affiliation(s)
- Morgan C Marsh
- Department of Molecular Pharmaceutics, University of Utah, 30 South 2000 East, Room 301, Salt Lake City, Utah, 84112, USA
| | - Shawn C Owen
- Department of Molecular Pharmaceutics, University of Utah, 30 South 2000 East, Room 301, Salt Lake City, Utah, 84112, USA.
- Department of Medicinal Chemistry, University of Utah, Salt Lake City, Utah, 84112, USA.
- Department of Biomedical Engineering, University of Utah, Salt Lake City, Utah, 84112, USA.
| |
Collapse
|