1
|
Abedi A, Moosazadeh Moghaddam M, Kachuei R, Imani Fooladi AA. Exosomes as a Therapeutic Strategy in Cancer: Potential Roles as Drug Carriers and Immune Modulators. Biochim Biophys Acta Rev Cancer 2024; 1880:189238. [PMID: 39674417 DOI: 10.1016/j.bbcan.2024.189238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 12/04/2024] [Accepted: 12/05/2024] [Indexed: 12/16/2024]
Abstract
Exosome-based cancer immunotherapy is advancing quickly on the concept of artificially activating the immune system to combat cancer. They can mechanistically change the tumor microenvironment, increase immune responses, and function as efficient drug delivery vehicles because of their inherent bioactivity, low toxicity, and immunogenicity. Accurate identification of the mechanisms of action of exosomes in tumor environments, along with optimization of their isolation, purification, and characterization methods, is necessary to increase clinical applications. Exosomes can be modified through cargo loading and surface modification to enhance their therapeutic applications, either before or after the donor cells' isolation. These engineered exosomes can directly target tumor cells at the tumor site or indirectly activate innate and adaptive immune responses in the tumor microenvironment. This approach is particularly effective when combined with traditional cancer immunotherapy techniques such as vaccines, immune checkpoints, and CAR-T cells. It can improve anti-tumor responses, induce long-term immunity, and address the limitations of traditional therapies, such as poor penetration in solid tumors and immunosuppressive environments. This review aims to provide a comprehensive and detailed overview of the direct role of engineered exosomes as drug delivery systems and their immunomodulatory effects on tumors as an indirect approach to fighting cancer. Additionally, it will discuss novel immunotherapy options.
Collapse
Affiliation(s)
- Azam Abedi
- Tissue Engineering and Regenerative Medicine Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Mehrdad Moosazadeh Moghaddam
- Tissue Engineering and Regenerative Medicine Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Reza Kachuei
- Molecular Biology Research Center, Biomedicine Technologies Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Abbas Ali Imani Fooladi
- Applied Microbiology Research Center, Biomedicine Technologies Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
2
|
Adam K, Butler SC, Workman CJ, Vignali DAA. Advances in LAG3 cancer immunotherapeutics. Trends Cancer 2024:S2405-8033(24)00248-6. [PMID: 39603977 DOI: 10.1016/j.trecan.2024.10.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 10/28/2024] [Accepted: 10/29/2024] [Indexed: 11/29/2024]
Abstract
Cancer treatment has entered the age of immunotherapy. Immune checkpoint inhibitor (ICI) therapy has shown robust therapeutic potential in clinical practice, with significant improvements in progression-free survival (PFS) and overall survival (OS). Recently, checkpoint blockade of the lymphocyte activation gene 3 (LAG3) inhibitory receptor (IR) in combination with programmed death protein 1 (PD1) inhibition has been FDA approved in patients with advanced melanoma. This has encouraged the clinical evaluation of new LAG3-directed biologics in combination with other checkpoint inhibitors. Several of these studies are evaluating bispecific antibodies that target exhausted T (TEX) cells expressing multiple IRs. This review discusses the current understanding of LAG3 in regulating antitumor immunity and the ongoing clinical testing of LAG3 inhibition in cancer.
Collapse
Affiliation(s)
- Kieran Adam
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA, USA; Program in Microbiology and Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Samuel C Butler
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA, USA; Program in Microbiology and Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Creg J Workman
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
| | - Dario A A Vignali
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA, USA; Cancer Immunology and Immunotherapy Program, UPMC Hillman Cancer Center, Pittsburgh, PA, USA.
| |
Collapse
|
3
|
Thomas CJ, Delgado K, Sawant K, Roy J, Gupta U, Song CS, Poojary R, de Figueiredo P, Song J. Harnessing Bacterial Agents to Modulate the Tumor Microenvironment and Enhance Cancer Immunotherapy. Cancers (Basel) 2024; 16:3810. [PMID: 39594765 PMCID: PMC11593222 DOI: 10.3390/cancers16223810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 10/22/2024] [Accepted: 10/26/2024] [Indexed: 11/28/2024] Open
Abstract
Cancer immunotherapy has revolutionized cancer treatment by leveraging the immune system to attack tumors. However, its effectiveness is often hindered by the immunosuppressive tumor microenvironment (TME), where a complex interplay of tumor, stromal, and immune cells undermines antitumor responses and allows tumors to evade immune detection. This review explores innovative strategies to modify the TME and enhance immunotherapy outcomes, focusing on the therapeutic potential of engineered bacteria. These bacteria exploit the unique characteristics of the TME, such as abnormal vasculature and immune suppression, to selectively accumulate in tumors. Genetically modified bacteria can deliver therapeutic agents, including immune checkpoint inhibitors and cytokines, directly to tumor sites. This review highlights how bacterial therapeutics can target critical immune cells within the TME, such as myeloid-derived suppressor cells and tumor-associated macrophages, thereby promoting antitumor immunity. The combination of bacterial therapies with immune checkpoint inhibitors or adoptive cell transfer presents a promising strategy to counteract immune suppression. Continued research in this area could position bacterial agents as a powerful new modality to reshape the TME and enhance the efficacy of cancer immunotherapy, particularly for tumors resistant to conventional treatments.
Collapse
Affiliation(s)
- Christina James Thomas
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, MREB II, Room 3344, 8447 John Sharp Parkway, Bryan, TX 77807, USA; (C.J.T.); (K.D.)
| | - Kaylee Delgado
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, MREB II, Room 3344, 8447 John Sharp Parkway, Bryan, TX 77807, USA; (C.J.T.); (K.D.)
| | - Kamlesh Sawant
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, MREB II, Room 3344, 8447 John Sharp Parkway, Bryan, TX 77807, USA; (C.J.T.); (K.D.)
| | - Jacob Roy
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, MREB II, Room 3344, 8447 John Sharp Parkway, Bryan, TX 77807, USA; (C.J.T.); (K.D.)
| | - Udit Gupta
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, MREB II, Room 3344, 8447 John Sharp Parkway, Bryan, TX 77807, USA; (C.J.T.); (K.D.)
| | - Carly Shaw Song
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, MREB II, Room 3344, 8447 John Sharp Parkway, Bryan, TX 77807, USA; (C.J.T.); (K.D.)
| | - Rayansh Poojary
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, MREB II, Room 3344, 8447 John Sharp Parkway, Bryan, TX 77807, USA; (C.J.T.); (K.D.)
| | - Paul de Figueiredo
- Department of Molecular Microbiology and Immunology, The University of Missouri School of Medicine, Columbia, MO 65212, USA
| | - Jianxun Song
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, MREB II, Room 3344, 8447 John Sharp Parkway, Bryan, TX 77807, USA; (C.J.T.); (K.D.)
| |
Collapse
|
4
|
Ceuppens H, Pombo Antunes AR, Navarro L, Ertveldt T, Berdal M, Nagachinta S, De Ridder K, Lahoutte T, Keyaerts M, Devoogdt N, Goyvaerts C, D'Huyvetter M, Breckpot K. Efficient α and β - radionuclide therapy targeting fibroblast activation protein-α in an aggressive preclinical mouse tumour model. Eur J Nucl Med Mol Imaging 2024:10.1007/s00259-024-06914-4. [PMID: 39237746 DOI: 10.1007/s00259-024-06914-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Accepted: 09/01/2024] [Indexed: 09/07/2024]
Abstract
PURPOSE Targeted radionuclide therapy (TRT) is a cancer treatment with relative therapeutic efficacy across various cancer types. We studied the therapeutic potential of TRT using fibroblast activation protein-α (FAP) targeting sdAbs (4AH29) labelled with 225Ac or 131I in immunocompetent mice in a human FAP (hFAP) expressing lung cancer mouse model. We further explored the combination of TRT with programmed cell death ligand 1 (PD-L1) immune checkpoint blockade (ICB). METHODS We studied the biodistribution and tumour uptake of [131I]I-GMIB-4AH29 and [225Ac]Ac-DOTA-4AH29 by ex vivo γ-counting. Therapeutic efficacy of [131I]I-GMIB-4AH29 and [225Ac]Ac-DOTA-4AH29 was evaluated in an immunocompetent mouse model. Flow cytometry analysis of tumours from [225Ac]Ac-DOTA-4AH29 treated mice was performed. Treatment with [225Ac]Ac-DOTA-4AH29 was repeated in combination with PD-L1 ICB. RESULTS The biodistribution showed high tumour uptake of [131I]I-GMIB-4AH29 with 3.5 ± 0.5% IA/g 1 h post-injection (p.i.) decreasing to 0.9 ± 0.1% IA/g after 24 h. Tumour uptake of [225Ac]Ac-DOTA-4AH29 was also relevant with 2.1 ± 0.5% IA/g 1 h p.i. with a less steep decrease to 1.7 ± 0.2% IA/g after 24 h. Survival was significantly improved after treatment with low and high doses [131I]I-GMIB-4AH29 or [225Ac]Ac-DOTA-4AH29 compared to vehicle solution. Moreover, we observed significantly higher PD-L1 expression in tumours of mice treated with [225Ac]Ac-DOTA-4AH29 compared to vehicle solution. Therefore, we combined high dose [225Ac]Ac-DOTA-4AH29 with PD-L1 ICB showing therapeutic synergy. CONCLUSION [225Ac]Ac-DOTA-4AH29 and [131I]I-GMIB-4AH29 exhibit high and persistent tumour targeting, translating into prolonged survival in mice bearing aggressive tumours. Moreover, we demonstrate that the combination of PD-L1 ICB with [225Ac]Ac-DOTA-4AH29 TRT enhances its therapeutic efficacy.
Collapse
Affiliation(s)
- Hannelore Ceuppens
- Vrije Universiteit Brussel, Department of Biomedical Sciences, Translational Oncology Research Center, Laboratory for Molecular and Cellular Therapy, Laarbeeklaan 103. Building E, Brussels, 1090, Belgium.
| | | | - Laurent Navarro
- Precirix NV/SA, Burg. Etienne Demunterlaan 3, Brussels, B-1090, Belgium
| | - Thomas Ertveldt
- Vrije Universiteit Brussel, Molecular Imaging and Therapy Research Group, Laarbeeklaan 103/K, Brussels, 1090, Belgium
| | - Marion Berdal
- Precirix NV/SA, Burg. Etienne Demunterlaan 3, Brussels, B-1090, Belgium
| | - Surasa Nagachinta
- Precirix NV/SA, Burg. Etienne Demunterlaan 3, Brussels, B-1090, Belgium
| | - Kirsten De Ridder
- Vrije Universiteit Brussel, Department of Biomedical Sciences, Translational Oncology Research Center, Laboratory for Molecular and Cellular Therapy, Laarbeeklaan 103. Building E, Brussels, 1090, Belgium
| | - Tony Lahoutte
- Vrije Universiteit Brussel, Molecular Imaging and Therapy Research Group, Laarbeeklaan 103/K, Brussels, 1090, Belgium
- Vrije Universiteit Brussel, Universitair Ziekenhuis Brussel (UZ Brussel), Nuclear Medicine Department, Laarbeeklaan 101, Brussels, 1090, Belgium
| | - Marleen Keyaerts
- Vrije Universiteit Brussel, Molecular Imaging and Therapy Research Group, Laarbeeklaan 103/K, Brussels, 1090, Belgium
- Vrije Universiteit Brussel, Universitair Ziekenhuis Brussel (UZ Brussel), Nuclear Medicine Department, Laarbeeklaan 101, Brussels, 1090, Belgium
| | - Nick Devoogdt
- Vrije Universiteit Brussel, Molecular Imaging and Therapy Research Group, Laarbeeklaan 103/K, Brussels, 1090, Belgium
| | - Cleo Goyvaerts
- Vrije Universiteit Brussel, Molecular Imaging and Therapy Research Group, Laarbeeklaan 103/K, Brussels, 1090, Belgium
| | - Matthias D'Huyvetter
- Precirix NV/SA, Burg. Etienne Demunterlaan 3, Brussels, B-1090, Belgium.
- Vrije Universiteit Brussel, Molecular Imaging and Therapy Research Group, Laarbeeklaan 103/K, Brussels, 1090, Belgium.
| | - Karine Breckpot
- Vrije Universiteit Brussel, Department of Biomedical Sciences, Translational Oncology Research Center, Laboratory for Molecular and Cellular Therapy, Laarbeeklaan 103. Building E, Brussels, 1090, Belgium.
| |
Collapse
|
5
|
Zhang X, Wu Y, Lin J, Lu S, Lu X, Cheng A, Chen H, Zhang W, Luan X. Insights into therapeutic peptides in the cancer-immunity cycle: Update and challenges. Acta Pharm Sin B 2024; 14:3818-3833. [PMID: 39309492 PMCID: PMC11413705 DOI: 10.1016/j.apsb.2024.05.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 03/05/2024] [Accepted: 04/12/2024] [Indexed: 09/25/2024] Open
Abstract
Immunotherapies hold immense potential for achieving durable potency and long-term survival opportunities in cancer therapy. As vital biological mediators, peptides with high tissue penetration and superior selectivity offer significant promise for enhancing cancer immunotherapies (CITs). However, physicochemical peptide features such as conformation and stability pose challenges to their on-target efficacy. This review provides a comprehensive overview of recent advancements in therapeutic peptides targeting key steps of the cancer-immunity cycle (CIC), including tumor antigen presentation, immune cell regulation, and immune checkpoint signaling. Particular attention is given to the opportunities and challenges associated with these peptides in boosting CIC within the context of clinical progress. Furthermore, possible future developments in this field are also discussed to provide insights into emerging CITs with robust efficacy and safety profiles.
Collapse
Affiliation(s)
- Xiaokun Zhang
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research and Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Ye Wu
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research and Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Jiayi Lin
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research and Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Shengxin Lu
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research and Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Xinchen Lu
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research and Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Aoyu Cheng
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research and Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Hongzhuan Chen
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research and Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Weidong Zhang
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research and Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
- Institute of Medicinal Plant Development, Chinese Academy of Medical Science &, Peking Union Medical College, Beijing 100193, China
- School of Pharmacy, Second Military Medical University, Shanghai 200433, China
| | - Xin Luan
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research and Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| |
Collapse
|
6
|
La-Beck NM, Owoso J. Updates and emerging trends in the management of immune-related adverse events associated with immune checkpoint inhibitor therapy. Asia Pac J Oncol Nurs 2024; 11:100549. [PMID: 39234578 PMCID: PMC11372807 DOI: 10.1016/j.apjon.2024.100549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 06/26/2024] [Indexed: 09/06/2024] Open
Abstract
The rapidly expanding class of therapies targeting immune checkpoints for the treatment of various cancers now includes 8 clinically approved agents: a lymphocyte-activation gene 3 (LAG-3) inhibitor (relatlimab), a cytotoxic T lymphocyte associated protein 4 (CTLA-4) inhibitor (ipilimumab), three programmed cell death protein 1 (PD-1) inhibitors (nivolumab, pembrolizumab and cemiplimab), and three programmed cell death ligand-1 (PD-L1) inhibitors (atezolizumab, durvalumab, and avelumab). Previously, we reviewed the mechanisms of immune-related adverse events (irAEs), strategies for management of irAEs, and highlighted similarities as well as differences amongst clinical guidelines from the National Comprehensive Cancer Network (NCCN), American Society of Clinical Oncology (ASCO), Society for Immunotherapy of Cancer (SITC), and European Society for Medical Oncology (ESMO). Herein, we provide an update that includes discussion of changes to these clinical guidelines since our last review, the new LAG-3 targeted agents, emerging patterns of irAEs, and new directions for improved monitoring and treatment of irAEs that could incorporate interdisciplinary pharmacist-led teams, artificial intelligence, and pharmacogenomics.
Collapse
Affiliation(s)
- Ninh M La-Beck
- Department of Immunotherapeutics and Biotechnology, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Abilene, TX, USA
| | - Jesuwapelumi Owoso
- Department of Immunotherapeutics and Biotechnology, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Abilene, TX, USA
| |
Collapse
|
7
|
Lopresti L, Tatangelo V, Baldari CT, Patrussi L. Rewiring the T cell-suppressive cytokine landscape of the tumor microenvironment: a new frontier for precision anti-cancer therapy. Front Immunol 2024; 15:1418527. [PMID: 39281678 PMCID: PMC11392891 DOI: 10.3389/fimmu.2024.1418527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Accepted: 08/06/2024] [Indexed: 09/18/2024] Open
Abstract
T lymphocytes that infiltrate the tumor microenvironment (TME) often fail to function as effective anti-cancer agents. Within the TME, cell-to-cell inhibitory interactions play significant roles in dampening their anti-tumor activities. Recent studies have revealed that soluble factors released in the TME by immune and non-immune cells, as well as by tumor cells themselves, contribute to the exacerbation of T cell exhaustion. Our understanding of the cytokine landscape of the TME, their interrelationships, and their impact on cancer development is still at its early stages. In this review, we aim to shed light on Interleukin (IL) -6, IL-9, and IL-10, a small group of JAK/STAT signaling-dependent cytokines harboring T cell-suppressive effects in the TME and summarize their mechanisms of action. Additionally, we will explore how advancements in scientific research can help us overcoming the obstacles posed by cytokines that suppress T cells in tumors, with the ultimate objective of stimulating further investigations for the development of novel therapeutic strategies to counteract their tumor-promoting activities.
Collapse
Affiliation(s)
| | | | | | - Laura Patrussi
- Department of Life Sciences, University of Siena, Siena, Italy
| |
Collapse
|
8
|
Luo Y, Cai X, Yang B, Lu F, Yi C, Wu G. Advances in understanding the role of immune checkpoint LAG-3 in tumor immunity: a comprehensive review. Front Oncol 2024; 14:1402837. [PMID: 39252941 PMCID: PMC11381248 DOI: 10.3389/fonc.2024.1402837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 07/26/2024] [Indexed: 09/11/2024] Open
Abstract
Lymphocyte activation gene 3 (LAG-3), also known as CD223, is an emerging immune checkpoint that follows PD-1 and CTLA-4. Several LAG-3 targeting inhibitors in clinical trials and the combination of relatlimab (anti-LAG-3) and nivolumab (anti-PD-1) have been approved for treating - unresectable or metastatic melanoma. Despite the encouraging clinical potential of LAG-3, the physiological function and mechanism of action in tumors are still not well understood. In this review, we systematically summarized the structure of LAG-3, ligands of LAG-3, cell-specific functions and signaling of LAG-3, and the current status of LAG-3 inhibitors under development.
Collapse
Affiliation(s)
- Yingzhe Luo
- Department of Oncology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Xuebin Cai
- Department of Abdominal Oncology, Division of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Biao Yang
- Department of Abdominal Oncology, Division of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Facheng Lu
- Department of Abdominal Oncology, Division of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Cheng Yi
- Department of Oncology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Guoyu Wu
- Department of Oncology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| |
Collapse
|
9
|
Yuan P, Long Y, Wei N, Wang Y, Zhu Z, Han J, Jiang D, Lan X, Gai Y. Peptide-based PET tracer targeting LAG-3 for evaluating the efficacy of immunotherapy in melanoma. J Immunother Cancer 2024; 12:e009010. [PMID: 39043603 DOI: 10.1136/jitc-2024-009010] [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] [Accepted: 07/12/2024] [Indexed: 07/25/2024] Open
Abstract
BACKGROUND Lymphocyte activation gene 3 (LAG-3) is expressed on activated immune cells and has emerged as a promising target for immune checkpoints blockade. However, conflicting findings have been reported regarding the association between LAG-3 expression in tumors and patient prognosis, indicating the need for further investigation into the significance of LAG-3 expression levels in tumor therapies. In this study, 68Ga-NOTA-XH05, a novel peptide-based positron emission tomography (PET) tracer targeting LAG-3, was constructed to non-invasively detect LAG-3 expression in melanoma after CpG oligonucleotide (CpG) treatment and explore the relationship between LAG-3 expression and therapeutic effect. METHODS The tracer 68Ga-NOTA-XH05 was identified by high-performance liquid chromatography after being prepared and purified. Cell uptake and blocking essays were performed to verify the specificity of the tracer in vitro. The expression of LAG-3 in B16-F10 subcutaneous tumors was monitored by flow cytometry, and its correlation with the tracer uptake was analyzed to evaluate the tracer specificity. PET imaging and biodistribution studies were conducted after CpG treatment of unilateral or bilateral B16-F10 subcutaneous tumor models to assess the ability of 68Ga-NOTA-XH05 in monitoring immunotherapy efficacy and the abscopal effect of CpG. RESULTS Following purification, 68Ga-NOTA-XH05 exhibited high radiochemical purity and specificity. Flow cytometry analysis revealed a positive correlation between LAG-3 expression in tumors and the uptake of 68Ga-NOTA-XH05. In B16-F10 bearing mice treated with CpG, PET imaging using 68Ga-NOTA-XH05 demonstrated a higher tumor to blood ratio (TBR) compared with the control group. Furthermore, TBR values obtained from CpG-treated mice allowed for differentiation between responders and non-responders. In a bilateral subcutaneous tumor model where only right-sided tumors were treated with intratumoral injection of CpG, TBR values of left-sided tumors were significantly higher than those in the control group, indicating that 68Ga-NOTA-XH05 could effectively monitor the systemic effect of local CpG injection. CONCLUSION Our findings highlight the detection capability of 68Ga-NOTA-XH05 in assessing LAG-3 expression levels within tumors and evaluating response to immunotherapy, thereby suggesting promising clinical translational prospects.
Collapse
Affiliation(s)
- Peizhe Yuan
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
- Key Laboratory of Biological Targeted Therapy, the Ministry of Education, Wuhan, People's Republic of China
| | - Yu Long
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
- Key Laboratory of Biological Targeted Therapy, the Ministry of Education, Wuhan, People's Republic of China
| | - Nannan Wei
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
- Key Laboratory of Biological Targeted Therapy, the Ministry of Education, Wuhan, People's Republic of China
| | - Yan Wang
- Department of Oncology, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ziyang Zhu
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
- Key Laboratory of Biological Targeted Therapy, the Ministry of Education, Wuhan, People's Republic of China
| | | | - Dawei Jiang
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
- Key Laboratory of Biological Targeted Therapy, the Ministry of Education, Wuhan, People's Republic of China
| | - Xiaoli Lan
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
- Key Laboratory of Biological Targeted Therapy, the Ministry of Education, Wuhan, People's Republic of China
| | - Yongkang Gai
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
- Key Laboratory of Biological Targeted Therapy, the Ministry of Education, Wuhan, People's Republic of China
| |
Collapse
|
10
|
Olejarz W, Sadowski K, Szulczyk D, Basak G. Advancements in Personalized CAR-T Therapy: Comprehensive Overview of Biomarkers and Therapeutic Targets in Hematological Malignancies. Int J Mol Sci 2024; 25:7743. [PMID: 39062986 PMCID: PMC11276786 DOI: 10.3390/ijms25147743] [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: 06/30/2024] [Revised: 07/12/2024] [Accepted: 07/12/2024] [Indexed: 07/28/2024] Open
Abstract
Chimeric antigen receptor T-cell (CAR-T) therapy is a novel anticancer therapy using autologous or allogeneic T-cells. To date, six CAR-T therapies for specific B-cell acute lymphoblastic leukemia (B-ALL), non-Hodgkin lymphomas (NHL), and multiple myeloma (MM) have been approved by the Food and Drug Administration (FDA). Significant barriers to the effectiveness of CAR-T therapy include cytokine release syndrome (CRS), neurotoxicity in the case of Allogeneic Stem Cell Transplantation (Allo-SCT) graft-versus-host-disease (GVHD), antigen escape, modest antitumor activity, restricted trafficking, limited persistence, the immunosuppressive microenvironment, and senescence and exhaustion of CAR-Ts. Furthermore, cancer drug resistance remains a major problem in clinical practice. CAR-T therapy, in combination with checkpoint blockades and bispecific T-cell engagers (BiTEs) or other drugs, appears to be an appealing anticancer strategy. Many of these agents have shown impressive results, combining efficacy with tolerability. Biomarkers like extracellular vesicles (EVs), cell-free DNA (cfDNA), circulating tumor (ctDNA) and miRNAs may play an important role in toxicity, relapse assessment, and efficacy prediction, and can be implicated in clinical applications of CAR-T therapy and in establishing safe and efficacious personalized medicine. However, further research is required to fully comprehend the particular side effects of immunomodulation, to ascertain the best order and combination of this medication with conventional chemotherapy and targeted therapies, and to find reliable predictive biomarkers.
Collapse
Affiliation(s)
- Wioletta Olejarz
- Department of Biochemistry and Pharmacogenomics, Faculty of Pharmacy, Medical University of Warsaw, 02-097 Warsaw, Poland;
- Centre for Preclinical Research, Medical University of Warsaw, 02-097 Warsaw, Poland
| | - Karol Sadowski
- Department of Biochemistry and Pharmacogenomics, Faculty of Pharmacy, Medical University of Warsaw, 02-097 Warsaw, Poland;
- Centre for Preclinical Research, Medical University of Warsaw, 02-097 Warsaw, Poland
- Department of Hematology, Transplantation and Internal Medicine, Medical University of Warsaw, 02-097 Warsaw, Poland;
| | - Daniel Szulczyk
- Chair and Department of Biochemistry, The Medical University of Warsaw, 02-097 Warsaw, Poland;
| | - Grzegorz Basak
- Department of Hematology, Transplantation and Internal Medicine, Medical University of Warsaw, 02-097 Warsaw, Poland;
| |
Collapse
|
11
|
Pan Y, Yao Z, Huang L, Xu M, Chen R, Li D, Wang X, Wu J, Li M, Liang X, Tan J. Overexpression of LAG-3: a potential indicator of low immune function in tuberculosis. Front Cell Infect Microbiol 2024; 14:1410015. [PMID: 38957797 PMCID: PMC11217189 DOI: 10.3389/fcimb.2024.1410015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Accepted: 06/04/2024] [Indexed: 07/04/2024] Open
Abstract
Background Tuberculosis (TB) persists as a global health challenge, with its treatment hampered by the side effects of long-term combination drug therapies and the growing issue of drug resistance. Therefore, the development of novel therapeutic strategies is critical. This study focuses on the role of immune checkpoint molecules (ICs) and functions of CD8+ T cells in the search for new potential targets against TB. Methods We conducted differential expression genes analysis and CD8+ T cell functional gene analysis on 92 TB samples and 61 healthy individual (HI) samples from TB database GSE83456, which contains data on 34,603 genes. The GSE54992 dataset was used to validated the findings. Additionally, a cluster analysis on single-cell data from primates infected with mycobacterium tuberculosis and those vaccinated with BCG was performed. Results The overexpression of LAG-3 gene was found as a potentially important characteristic of both pulmonary TB (PTB) and extrapulmonary TB (EPTB). Further correlation analysis showed that LAG-3 gene was correlated with GZMB, perforin, IL-2 and IL-12. A significant temporal and spatial variation in LAG-3 expression was observed in T cells and macrophages during TB infection and after BCG vaccination. Conclusion LAG-3 was overexpressed in TB samples. Targeting LAG-3 may represent a potential therapeutic target for tuberculosis.
Collapse
Affiliation(s)
- Yun Pan
- The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Zengxi Yao
- The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Lifen Huang
- The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Meina Xu
- College of Pharmacy, Jinan University, Guangzhou, China
| | - Ruichang Chen
- The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Dengsheng Li
- The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Xinyuan Wang
- The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Jianchao Wu
- The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Minran Li
- The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Xujing Liang
- The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Jiaxiong Tan
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
- Pediatric Oncology, Tianjin’s Clinical Research Center for Cancer, Tianjin, China
| |
Collapse
|
12
|
El Dosoky W, Aref S, El Menshawy N, Ramez A, Abou Zaid T, Aref M, Atia D. Prognostic effect of CTLA4/LAG3 Expression by T-Cells Subsets on Acute Myeloid Leukemia Patients. Asian Pac J Cancer Prev 2024; 25:1777-1785. [PMID: 38809650 PMCID: PMC11318815 DOI: 10.31557/apjcp.2024.25.5.1777] [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: 01/17/2024] [Accepted: 05/18/2024] [Indexed: 05/31/2024] Open
Abstract
BACKGROUND Deregulation of immune checkpoint is an important point in cancer evolution as well as patients outcome. T-cells is an important arm in immunity against cancer. This study aimed to assess CTLA4/LAG3 expression on different T-cell subsets and its effect on disease outcome. METHODS This study included 81 newly diagnosed Egyptian adult AML patients. For each one of the patients CTLA4/ LAG3 expression on T-cell subsets was identified by flowcytometry before start of induction chemotherapy. RESULTS Total CD3 count in AML patients was lower than control. LAG3 expression were significantly higher in total CD3, T-cell subsets (CD4, CD8) as compared to healthy control. Moreover, co-expression of LAG3/CTLA4 on T-cell subsets were significantly higher in AML as compared to healthy control . NPM-/ FLT3+ was significantly associated with high LAG3 expression in T-cells subsets as compared to other molecular subtypes. Shorter OS, DFS were significantly associated with higher expression of LAG3 on T-cells subsets as compared to patients harbor low expression. COX regression analysis revealed that high expression of CD3/LAG3, CD4/LAG3, CD8/LAG4, CD3/CTLA4/LAG3 were considered a poor prognostic risk factor. CONCLUSION High LAG3/CTLA4 expression could predict AML Patients' outcome Conclusion: Our findings indicated that high expression of LAG3/CTL4 on T cells subsets identify a subgroup of AML patients with poor prognosis.
Collapse
Affiliation(s)
- Wesam El Dosoky
- Hematology Unit, Mansoura University Oncology Center, Mansoura University, Egypt.
| | - Salah Aref
- Hematology Unit, Mansoura University Oncology Center, Mansoura University, Egypt.
| | - Nadia El Menshawy
- Hematology Unit, Mansoura University Oncology Center, Mansoura University, Egypt.
| | - Ahmed Ramez
- Mansoura University Oncology Center, Mansoura University, Egypt.
| | - Tarek Abou Zaid
- Mansoura University Oncology Center, Mansoura Faculty of Medicine, Mansoura University, Egypt.
| | - Mohamed Aref
- Mansoura University Oncology Center, Mansoura Faculty of Medicine, Mansoura University, Egypt.
| | - Doaa Atia
- Mansoura University Oncology Center, Mansoura Faculty of Medicine, Mansoura University, Egypt.
| |
Collapse
|
13
|
Guo W, Peng D, Liao Y, Lou L, Guo M, Li C, Yu W, Tian X, Wang G, Lv P, Zuo J, Shen H, Li Y. Upregulation of HLA-II related to LAG-3 +CD4 + T cell infiltration is associated with patient outcome in human glioblastoma. Cancer Sci 2024; 115:1388-1404. [PMID: 38480275 PMCID: PMC11093187 DOI: 10.1111/cas.16128] [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: 11/19/2022] [Revised: 02/01/2024] [Accepted: 02/17/2024] [Indexed: 05/15/2024] Open
Abstract
Glioblastoma (GBM) is the most common malignant diffuse glioma of the brain. Although immunotherapy with immune checkpoint inhibitors (ICIs), such as programmed cell death protein (PD)-1/PD ligand-1 inhibitors, has revolutionized the treatment of several cancers, the clinical benefit in GBM patients has been limited. Lymphocyte-activation gene 3 (LAG-3) binding to human leukocyte antigen-II (HLA-II) plays an essential role in triggering CD4+ T cell exhaustion and could interfere with the efficiency of anti-PD-1 treatment; however, the value of LAG-3-HLA-II interactions in ICI immunotherapy for GBM patients has not yet been analyzed. Therefore, we aimed to investigate the expression and regulation of HLA-II in human GBM samples and the correlation with LAG-3+CD4+ T cell infiltration. Human leukocyte antigen-II was highly expressed in GBM and correlated with increased LAG-3+CD4+ T cell infiltration in the stroma. Additionally, HLA-IIHighLAG-3High was associated with worse patient survival. Increased interleukin-10 (IL-10) expression was observed in GBM, which was correlated with high levels of HLA-II and LAG-3+ T cell infiltration in stroma. HLA-IIHighIL-10High GBM associated with LAG-3+ T cells infiltration synergistically showed shorter overall survival in patients. Combined anti-LAG-3 and anti-IL-10 treatment inhibited tumor growth in a mouse brain GL261 tumor model. In vitro, CD68+ macrophages upregulated HLA-II expression in GBM cells through tumor necrosis factor-α (TNF-α). Blocking TNF-α-dependent inflammation inhibited tumor growth in a mouse GBM model. In summary, T cell-tumor cell interactions, such as LAG-3-HLA-II, could confer an immunosuppressive environment in human GBM, leading to poor prognosis in patients. Therefore, targeting the LAG-3-HLA-II interaction could be beneficial in ICI immunotherapy to improve the clinical outcome of GBM patients.
Collapse
Affiliation(s)
- Wenli Guo
- Department of PathologyThe Second Hospital, Hebei Medical UniversityShijiazhuangChina
- Laboratory of PathologyHebei Medical UniversityShijiazhuangChina
| | - Daijun Peng
- Department of PathologyThe Second Hospital, Hebei Medical UniversityShijiazhuangChina
| | - Yuee Liao
- Department of PathologyThe Second Hospital, Hebei Medical UniversityShijiazhuangChina
| | - Lei Lou
- Department of PathologyThe Second Hospital, Hebei Medical UniversityShijiazhuangChina
| | - Moran Guo
- Department of NeurologySecond Hospital of Hebei Medical UniversityShijiazhuangChina
| | - Chen Li
- Department of NeurosurgerySecond Hospital of Hebei Medical UniversityShijiazhuangChina
| | - Wangyang Yu
- Department of NeurosurgerySecond Hospital of Hebei Medical UniversityShijiazhuangChina
| | - Xiaoxi Tian
- Department of PathologyThe Second Hospital, Hebei Medical UniversityShijiazhuangChina
| | - Guohui Wang
- Department of PathologyThe Second Hospital, Hebei Medical UniversityShijiazhuangChina
| | - Ping Lv
- Department of PharmacologyHebei Medical UniversityShijiazhuangChina
| | - Jing Zuo
- Department of OncologyThe Fourth Hospital of Hebei Medical UniversityShijiazhuangChina
| | - Haitao Shen
- Laboratory of PathologyHebei Medical UniversityShijiazhuangChina
- Hebei Collaborative Innovation Center of Tumor Microecological Metabolism RegulationHebei UniversityBaodingChina
| | - Yuehong Li
- Department of PathologyThe Second Hospital, Hebei Medical UniversityShijiazhuangChina
- Laboratory of PathologyHebei Medical UniversityShijiazhuangChina
| |
Collapse
|
14
|
Gorgulho J, Roderburg C, Beier F, Bokemeyer C, Brümmendorf TH, Loosen SH, Luedde T. Soluble lymphocyte activation gene-3 (sLAG3) and CD4/CD8 ratio dynamics as predictive biomarkers in patients undergoing immune checkpoint blockade for solid malignancies. Br J Cancer 2024; 130:1013-1022. [PMID: 38233492 PMCID: PMC10951205 DOI: 10.1038/s41416-023-02558-7] [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: 02/05/2023] [Revised: 12/05/2023] [Accepted: 12/13/2023] [Indexed: 01/19/2024] Open
Abstract
BACKGROUND The search for biomarkers to identify suitable candidates for immune checkpoint inhibitor (ICI) therapy remains ongoing. We evaluate how soluble levels of the next generation immune checkpoint Lymphocyte Activation Gene-3 (sLAG-3) and its association with circulating T lymphocyte subsets could pose as a novel biomarker to predict outcome to ICI therapy. METHODS Circulating levels of sLAG3 were analyzed using multiplex immunoassay in n = 84 patients undergoing ICI therapy for advanced solid cancer, accompanied by flow cytometry analyses of peripheral blood mononuclear cells (PBMCs). RESULTS Uni- and multivariate analysis shows that patients with higher sLAG3 concentrations before ICI therapy had a significantly impaired progression-free (PFS) and overall survival (OS) (HRPFS: 1.005 [95%CI: 1.000-1.009], p = 0.039; HROS: 1.006 [95%CI: 1.001-1.011], p = 0.015). The CD4/CD8 cell ratio and its dynamics during therapy were strong predictors of PFS and OS with patients with a decreasing ratio between baseline and after 1-2 cycles having an improved median OS compared to patients with increasing values (p = 0.012, HR: 3.32). An immunological score combining sLAG3 and the CD4/CD8 ratio showed the highest predictive potential (HROS: 10.3). CONCLUSION Pending prospective validation, sLAG3 and correlating circulating T-cell subsets can be used as a non-invasive predictive marker to predict outcome to ICI therapy to help identifying ideal ICI candidates in the future.
Collapse
Affiliation(s)
- Joao Gorgulho
- Department of Oncology, Hematology and Bone Marrow Transplantation with Section of Pneumology, University Medical Centre Hamburg-Eppendorf, Martinistraße 52, 20251, Hamburg, Germany
- Mildred Scheel Cancer Career Center, University Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Christoph Roderburg
- Department of Gastroenterology, Hepatology and Infectious Diseases, University Hospital Düsseldorf, Medical Faculty of Heinrich Heine University Düsseldorf, 40225, Düsseldorf, Germany
- Center for Integrated Oncology Aachen-Bonn-Cologne-Düsseldorf (CIOABCD), Aachen, Germany
| | - Fabian Beier
- Center for Integrated Oncology Aachen-Bonn-Cologne-Düsseldorf (CIOABCD), Aachen, Germany
- Department of Medicine IV, University Hospital RWTH Aachen, Pauwelsstrasse 30, 52074, Aachen, Germany
| | - Carsten Bokemeyer
- Department of Oncology, Hematology and Bone Marrow Transplantation with Section of Pneumology, University Medical Centre Hamburg-Eppendorf, Martinistraße 52, 20251, Hamburg, Germany
| | - Tim H Brümmendorf
- Center for Integrated Oncology Aachen-Bonn-Cologne-Düsseldorf (CIOABCD), Aachen, Germany
- Department of Medicine IV, University Hospital RWTH Aachen, Pauwelsstrasse 30, 52074, Aachen, Germany
| | - Sven H Loosen
- Department of Gastroenterology, Hepatology and Infectious Diseases, University Hospital Düsseldorf, Medical Faculty of Heinrich Heine University Düsseldorf, 40225, Düsseldorf, Germany.
- Center for Integrated Oncology Aachen-Bonn-Cologne-Düsseldorf (CIOABCD), Aachen, Germany.
| | - Tom Luedde
- Department of Gastroenterology, Hepatology and Infectious Diseases, University Hospital Düsseldorf, Medical Faculty of Heinrich Heine University Düsseldorf, 40225, Düsseldorf, Germany.
- Center for Integrated Oncology Aachen-Bonn-Cologne-Düsseldorf (CIOABCD), Aachen, Germany.
| |
Collapse
|
15
|
Fang Q, Liu Q, Song Z, Zhang X, Du Y. A NAD(P)H oxidase mimic for catalytic tumor therapy via a deacetylase SIRT7-mediated AKT/GSK3β pathway. NANOSCALE 2024; 16:6585-6595. [PMID: 38465774 DOI: 10.1039/d3nr06538c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
Nicotinamide adenine dinucleotide (NADH) and its phosphorylated form, NADPH, are essential cofactors that play critical roles in cell functions, influencing antioxidation, reductive biosynthesis, and cellular pathways involved in tumor cell apoptosis and tumorigenesis. However, the use of nanomaterials to consume NAD(P)H and thus bring an impact on signaling pathways in cancer treatment remains understudied. In this study, we employed a salt template method to synthesize a carbon-coated-cobalt composite (C@Co) nanozyme, which exhibited excellent NAD(P)H oxidase (NOX)-like activity and mimicked the reaction mechanism of natural NOX. The C@Co nanozyme efficiently consumed NAD(P)H within cancer cells, leading to increased production of reactive oxygen species (ROS) and a reduction in mitochondrial membrane potential. Meanwhile, the generation of the biologically active cofactor NAD(P)+ promoted the expression of the deacetylase SIRT7, which in turn inhibited the serine/threonine kinase AKT signaling pathway, ultimately promoting apoptosis. This work sheds light on the influence of nanozymes with NOX-like activity on cellular signaling pathways in tumor therapy and demonstrates their promising antitumor effects in a tumor xenograft mouse model. These findings contribute to a better understanding of NAD(P)H manipulation in cancer treatment and suggest the potential of nanozymes as a therapeutic strategy for cancer therapy.
Collapse
Affiliation(s)
- Qi Fang
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, P. R. China.
- School of Applied Chemistry and Engineering, University of Science & Technology of China, Hefei, Anhui 230026, P. R. China
| | - Quanyi Liu
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, P. R. China.
- School of Applied Chemistry and Engineering, University of Science & Technology of China, Hefei, Anhui 230026, P. R. China
| | - Zhimin Song
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, P. R. China.
- School of Applied Chemistry and Engineering, University of Science & Technology of China, Hefei, Anhui 230026, P. R. China
| | - Xiaojun Zhang
- School of Applied Chemistry and Engineering, University of Science & Technology of China, Hefei, Anhui 230026, P. R. China
| | - Yan Du
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, P. R. China.
- School of Applied Chemistry and Engineering, University of Science & Technology of China, Hefei, Anhui 230026, P. R. China
| |
Collapse
|
16
|
Wang W, Liu Y, He Z, Li L, Liu S, Jiang M, Zhao B, Deng M, Wang W, Mi X, Sun Z, Ge X. Breakthrough of solid tumor treatment: CAR-NK immunotherapy. Cell Death Discov 2024; 10:40. [PMID: 38245520 PMCID: PMC10799930 DOI: 10.1038/s41420-024-01815-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 01/04/2024] [Accepted: 01/09/2024] [Indexed: 01/22/2024] Open
Abstract
As the latest and most anticipated method of tumor immunotherapy, CAR-NK therapy has received increasing attention in recent years, and its safety and high efficiency have irreplaceable advantages over CAR-T. Current research focuses on the application of CAR-NK in hematological tumors, while there are fewer studies on solid tumor. This article reviews the process of constructing CAR-NK, the effects of hypoxia and metabolic factors, NK cell surface receptors, cytokines, and exosomes on the efficacy of CAR-NK in solid tumor, and the role of CAR-NK in various solid tumor. The mechanism of action and the research status of the potential of CAR-NK in the treatment of solid tumor in clinical practice, and put forward the advantages, limitations and future problems of CAR-NK in the treatment of solid tumor.
Collapse
Affiliation(s)
- Wenkang Wang
- Department of Breast Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| | - Yang Liu
- Department of Radiotherapy, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China
| | - Zhen He
- Department of Internal Medicine, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China
| | - Lifeng Li
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Senbo Liu
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Mingqiang Jiang
- Department of Breast Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Bing Zhao
- Department of Breast Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Meng Deng
- Department of Breast Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Wendong Wang
- Department of Breast Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xuefang Mi
- Department of Breast Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhenqiang Sun
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| | - Xin Ge
- Department of Breast Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| |
Collapse
|
17
|
Adhikary S, Pathak S, Palani V, Acar A, Banerjee A, Al-Dewik NI, Essa MM, Mohammed SGAA, Qoronfleh MW. Current Technologies and Future Perspectives in Immunotherapy towards a Clinical Oncology Approach. Biomedicines 2024; 12:217. [PMID: 38255322 PMCID: PMC10813720 DOI: 10.3390/biomedicines12010217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 01/08/2024] [Accepted: 01/12/2024] [Indexed: 01/24/2024] Open
Abstract
Immunotherapy is now established as a potent therapeutic paradigm engendering antitumor immune response against a wide range of malignancies and other diseases by modulating the immune system either through the stimulation or suppression of immune components such as CD4+ T cells, CD8+ T cells, B cells, monocytes, macrophages, dendritic cells, and natural killer cells. By targeting several immune checkpoint inhibitors or blockers (e.g., PD-1, PD-L1, PD-L2, CTLA-4, LAG3, and TIM-3) expressed on the surface of immune cells, several monoclonal antibodies and polyclonal antibodies have been developed and already translated clinically. In addition, natural killer cell-based, dendritic cell-based, and CAR T cell therapies have been also shown to be promising and effective immunotherapeutic approaches. In particular, CAR T cell therapy has benefited from advancements in CRISPR-Cas9 genome editing technology, allowing the generation of several modified CAR T cells with enhanced antitumor immunity. However, the emerging SARS-CoV-2 infection could hijack a patient's immune system by releasing pro-inflammatory interleukins and cytokines such as IL-1β, IL-2, IL-6, and IL-10, and IFN-γ and TNF-α, respectively, which can further promote neutrophil extravasation and the vasodilation of blood vessels. Despite the significant development of advanced immunotherapeutic technologies, after a certain period of treatment, cancer relapses due to the development of resistance to immunotherapy. Resistance may be primary (where tumor cells do not respond to the treatment), or secondary or acquired immune resistance (where tumor cells develop resistance gradually to ICIs therapy). In this context, this review aims to address the existing immunotherapeutic technologies against cancer and the resistance mechanisms against immunotherapeutic drugs, and explain the impact of COVID-19 on cancer treatment. In addition, we will discuss what will be the future implementation of these strategies against cancer drug resistance. Finally, we will emphasize the practical steps to lay the groundwork for enlightened policy for intervention and resource allocation to care for cancer patients.
Collapse
Affiliation(s)
- Subhamay Adhikary
- Medical Biotechnology, Faculty of Allied Health Sciences, Chettinad Academy of Research and Education (CARE), Chettinad Hospital and Research Institute (CHRI), Chennai 603103, India
| | - Surajit Pathak
- Medical Biotechnology, Faculty of Allied Health Sciences, Chettinad Academy of Research and Education (CARE), Chettinad Hospital and Research Institute (CHRI), Chennai 603103, India
| | - Vignesh Palani
- Faculty of Medicine, Chettinad Hospital and Research Institute (CHRI), Chennai 603103, India
| | - Ahmet Acar
- Department of Biological Sciences, Middle East Technical University, 06800 Ankara, Türkiye;
| | - Antara Banerjee
- Medical Biotechnology, Faculty of Allied Health Sciences, Chettinad Academy of Research and Education (CARE), Chettinad Hospital and Research Institute (CHRI), Chennai 603103, India
| | - Nader I. Al-Dewik
- Department of Pediatrics, Women’s Wellness and Research Center, Hamad Medical Corporation, Doha 00974, Qatar;
| | - Musthafa Mohamed Essa
- College of Agricultural and Marine Sciences, Sultan Qaboos University, Muscat 123, Oman
| | | | - M. Walid Qoronfleh
- Research & Policy Division, Q3 Research Institute (QRI), Ypsilanti, MI 48917, USA
| |
Collapse
|
18
|
Su J, Fu Y, Cui Z, Abidin Z, Yuan J, Zhang X, Li R, Zhao C. Relatlimab: a novel drug targeting immune checkpoint LAG-3 in melanoma therapy. Front Pharmacol 2024; 14:1349081. [PMID: 38269271 PMCID: PMC10806167 DOI: 10.3389/fphar.2023.1349081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 12/27/2023] [Indexed: 01/26/2024] Open
Abstract
Relatlimab is a type of human immunoglobulin G4 monoclonal blocking antibody. It is the world's first Lymphocyte-Activation Gene-3 (LAG-3) inhibitor and the third immune checkpoint inhibitor with clinical application, following PD-1 and CTLA-4. Relatlimab can bind to the LAG-3 receptor which blocks the interaction between LAG-3 and its ligand to reduce LAG-3 pathway-mediated immunosuppression and promote T-cell proliferation, inducing tumor cell death. On 18 March 2022, the U.S. FDA approved the fixed-dose combination of relatlimab developed by Bristol Myers Squibb with nivolumab, under the brand name Opdualag for the treatment of unresectable or metastatic melanoma in adult and pediatric patients aged 12 and older. This study comprehensively describes the mechanism of action and clinical trials of relatlimab and a brief overview of immune checkpoint drugs currently used for the treatment of melanoma.
Collapse
Affiliation(s)
- Jingjing Su
- Key Laboratory of Molecular Pharmacology and Translational Medicine and Department of Pharmacology, College of Pharmacy, Weifang Medical University, Weifang, China
| | - Yiting Fu
- Key Laboratory of Molecular Pharmacology and Translational Medicine and Department of Pharmacology, College of Pharmacy, Weifang Medical University, Weifang, China
| | - Zitong Cui
- Key Laboratory of Molecular Pharmacology and Translational Medicine and Department of Pharmacology, College of Pharmacy, Weifang Medical University, Weifang, China
| | - Zain Abidin
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John’s University, Queens, NY, United States
| | - Jingsong Yuan
- Key Laboratory of Molecular Pharmacology and Translational Medicine and Department of Pharmacology, College of Pharmacy, Weifang Medical University, Weifang, China
| | - Xinmiao Zhang
- Key Laboratory of Molecular Pharmacology and Translational Medicine and Department of Pharmacology, College of Pharmacy, Weifang Medical University, Weifang, China
| | - Runmin Li
- Key Laboratory of Molecular Pharmacology and Translational Medicine and Department of Pharmacology, College of Pharmacy, Weifang Medical University, Weifang, China
| | - Chunzhen Zhao
- Key Laboratory of Molecular Pharmacology and Translational Medicine and Department of Pharmacology, College of Pharmacy, Weifang Medical University, Weifang, China
| |
Collapse
|
19
|
Al-Kaif LAIK, Al-Ameri H, Alfatlawi WRO, Mahdi AE, Al-Khafaji YAK, Al-Saadi MAK, Al-Charrakh AH, Al-Mammori RT, Akkaif MA. Detection of CTLA-4 level and humeral immune response after the second dose of COVID-19 vaccine in certain Iraqi provinces participants. PLoS One 2024; 19:e0296521. [PMID: 38180994 PMCID: PMC10769031 DOI: 10.1371/journal.pone.0296521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Accepted: 12/14/2023] [Indexed: 01/07/2024] Open
Abstract
BACKGROUND Evaluating immune responses following COVID-19 vaccination is paramount to understanding vaccine effectiveness and optimizing public health interventions. This study seeks to elucidate individuals' immune status after administering a second dose of diverse COVID-19 vaccines. By analyzing immune responses through serological markers, we aim to contribute valuable insights into the uniformity of vaccine performance. METHODS A total of 80 participants were enrolled in this study, with demographic and COVID-19 infection-related data collected for categorization. Serum samples were acquired within a specified timeframe, and SARS-CoV-2 IgM/IgG rapid tests were conducted. Moreover, CTLA-4 levels were measured through ELISA assays, allowing us to assess the immune responses comprehensively. The participants were divided into eight groups based on various factors, facilitating a multifaceted analysis. RESULTS The outcomes of our investigation demonstrated consistent immune responses across the diverse types of COVID-19 vaccines administered in Iraq. Statistical analysis revealed no significant distinctions among the vaccine categories. In contrast, significant differences were observed in CTLA-4 among the control group (non-infected/non-vaccinated, infected/non-vaccinated) and infected/Pfizer, non-infected/Pfizer, and infected/Sinopharm, non-infected/sinopharm (P = 0.001, < 0.001, 0.023, respectively). This suggests that these vaccines exhibit comparable effectiveness in eliciting an immune response among the study participants. CONCLUSIONS In conclusion, our study's results underscore the lack of discriminatory variations between different COVID-19 vaccine types utilized in Iraq. The uniform immune responses observed signify the equitable efficacy and performance of these vaccines. Despite minor quantitative discrepancies, these variations do not hold statistical significance, reaffirming the notion that the various vaccines serve a similar purpose in conferring protection against COVID-19.
Collapse
Affiliation(s)
- Laith A. I. K. Al-Kaif
- Department of Medical Microbiology, Hammurabi College of Medicine, University of Babylon, Hillah, Babylon, Iraq
- Department of Medical Laboratory Techniques, Al-Mustaqbal University, Hillah, Babylon, Iraq
| | - Hussain Al-Ameri
- Department of Medical Laboratory Techniques, Al-Mustaqbal University, Hillah, Babylon, Iraq
| | | | - Ammar Eesa Mahdi
- Basic Science Department, College of Dentistry, University of Babylon, Hillah, Babylon, Iraq
| | | | | | - Alaa H. Al-Charrakh
- Basic Science Department, College of Dentistry, University of Babylon, Hillah, Babylon, Iraq
| | | | - Mohammed Ahmed Akkaif
- Department of Cardiology, Qingpu Branch of Zhongshan Hospital Affiliated to Fudan University, Shanghai, China
| |
Collapse
|
20
|
Chagan-Yasutan H, He N, Arlud S, Fang J, Hattori T. The elevation of plasma galectin-9 levels in patients with psoriasis and its associations with inflammatory and immune checkpoint molecules in skin tissues. Hum Immunol 2024; 85:110741. [PMID: 38092632 DOI: 10.1016/j.humimm.2023.110741] [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: 10/08/2023] [Revised: 11/29/2023] [Accepted: 12/04/2023] [Indexed: 01/24/2024]
Abstract
Psoriasis is a chronic, immune-mediated disorder that mainly affects the skin, with an estimated global prevalence of 2-3%. Galectin-9 (Gal-9) is a β-galactoside-binding lectin capable of promoting or suppressing the progression of infectious and immune-mediated diseases. Here, we determined if the expression of Gal-9 is observed in psoriasis. Gal-9 levels were measured in plasma of psoriasis (n = 62) and healthy control (HC) (n = 31) using an enzyme-linked immunosorbent assay. In addition, skin samples from seven patients were screened for RNA transcriptomes and the expression of Gal-9 was compared with inflammatory, immune checkpoint molecules (ICMs) and Foxp3. The plasma Gal-9 levels in patients with psoriasis were significantly higher (841 pg/mL) than in HCs (617 pg/mL) (P < 0.0001) and were associated with white blood cell numbers, eosinophils (%) and alanine transaminase. The levels of inflammatory molecules IL-36B, IL-17RA, IL-6R, IL-10, IRF8, TGFb1, and IL-37, and those of ICMs of Tim-3, CTLA-4, CD86, CD80, PD-1LG2, CLEC4G, and Foxp3 were significantly correlated with Gal-9 (LGALS9) in skin. However, HMGB1, CD44, CEACAM1 and PDL1-known to be associated with a variety of Gal-9 biological functions were not correlated with LGALS9. Thus, it is likely that Gal-9 expression affects the disease state of PS.
Collapse
Affiliation(s)
- Haorile Chagan-Yasutan
- Mongolian Psychosomatic Medicine Department, Inner Mongolia International Mongolian Medicine Hospital, Hohhot 010065, China; Research Institute of Health and Welfare, Kibi International University, 8-Iga-machi, Takahashi, Okayama 716-8508, Japan.
| | - Nagongbilige He
- Mongolian Psychosomatic Medicine Department, Inner Mongolia International Mongolian Medicine Hospital, Hohhot 010065, China; The Inner Mongolia Institute of Chinese and Mongolian Medicine, Hohhot 010010, China.
| | - Sarnai Arlud
- Mongolian Psychosomatic Medicine Department, Inner Mongolia International Mongolian Medicine Hospital, Hohhot 010065, China
| | - Jun Fang
- Mongolian Psychosomatic Medicine Department, Inner Mongolia International Mongolian Medicine Hospital, Hohhot 010065, China; The Inner Mongolia Institute of Chinese and Mongolian Medicine, Hohhot 010010, China
| | - Toshio Hattori
- Research Institute of Health and Welfare, Kibi International University, 8-Iga-machi, Takahashi, Okayama 716-8508, Japan; Shizuoka Graduate University of Public Health, 4-27-2 Kita Ando Aoi-ku, Shizuoka City 420-0881, Japan.
| |
Collapse
|
21
|
Li L, Li A, Jin H, Li M, Jia Q. Inhibitory receptors and checkpoints on NK cells: Implications for cancer immunotherapy. Pathol Res Pract 2024; 253:155003. [PMID: 38042093 DOI: 10.1016/j.prp.2023.155003] [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: 11/08/2023] [Revised: 11/23/2023] [Accepted: 11/27/2023] [Indexed: 12/04/2023]
Abstract
With the success of immunosuppressive checkpoint in tumor therapy, the corresponding adverse response and drug resistance defects have been exposed. T cells and NK cells are the body's immune system of the two substantial main forces. in recent years, study of T cell checkpoints appeared a certain block, such as PD-1 the effect not benign, on the distribution of NK cell surface excitatory and inhibitory receptors under normal conditions to maintain steady, could be targeted in the tumor treatment blockade have therapeutic effect. This paper reviews the function of NK cells and the effects of corresponding receptors in various types of tumors, providing a direction for the selection of appropriate gate control sites for future treatment.
Collapse
Affiliation(s)
- Lingfei Li
- State Key Laboratory of Cancer Biology, Department of Pathology, Xijing Hospital and School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Ang Li
- Department of Cardiology, 2nd Medical Center of PLA General Hospital, Beijing, China
| | - Hai Jin
- Department of Neurosurgery, General Hospital of Northern Theater Command, Shenyang, China.
| | - Mingyang Li
- State Key Laboratory of Cancer Biology, Department of Pathology, Xijing Hospital and School of Basic Medicine, Fourth Military Medical University, Xi'an, China.
| | - Qingge Jia
- Department of Reproductive Medicine, Xi'an International Medical Center Hospital, Northwest University, Xi'an, China.
| |
Collapse
|
22
|
Mejía-Guarnizo LV, Monroy-Camacho PS, Turizo-Smith AD, Rodríguez-García JA. The role of immune checkpoints in antitumor response: a potential antitumor immunotherapy. Front Immunol 2023; 14:1298571. [PMID: 38162657 PMCID: PMC10757365 DOI: 10.3389/fimmu.2023.1298571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Accepted: 11/29/2023] [Indexed: 01/03/2024] Open
Abstract
Immunotherapy aims to stimulate the immune system to inhibit tumor growth or prevent metastases. Tumor cells primarily employ altered expression of human leukocyte antigen (HLA) as a mechanism to avoid immune recognition and antitumor immune response. The antitumor immune response is primarily mediated by CD8+ cytotoxic T cells (CTLs) and natural killer (NK) cells, which plays a key role in the overall anti-tumor immune response. It is crucial to comprehend the molecular events occurring during the activation and subsequent regulation of these cell populations. The interaction between antigenic peptides presented on HLA-I molecules and the T-cell receptor (TCR) constitutes the initial signal required for T cell activation. Once activated, in physiologic circumstances, immune checkpoint expression by T cells suppress T cell effector functions when the antigen is removed, to ensures the maintenance of self-tolerance, immune homeostasis, and prevention of autoimmunity. However, in cancer, the overexpression of these molecules represents a common method through which tumor cells evade immune surveillance. Numerous therapeutic antibodies have been developed to inhibit immune checkpoints, demonstrating antitumor activity with fewer side effects compared to traditional chemotherapy. Nevertheless, it's worth noting that many immune checkpoint expressions occur after T cell activation and consequently, altered HLA expression on tumor cells could diminish the clinical efficacy of these antibodies. This review provides an in-depth exploration of immune checkpoint molecules, their corresponding blocking antibodies, and their clinical applications.
Collapse
Affiliation(s)
- Lidy Vannessa Mejía-Guarnizo
- Cancer Biology Research Group, Instituto Nacional de Cancerología, Bogotá, Colombia
- Sciences Faculty, Master in Microbiology, Universidad Nacional de Colombia, Bogotá, Colombia
| | | | | | | |
Collapse
|
23
|
Cheng X, Shen J, Xu J, Zhu J, Xu P, Wang Y, Gao M. In vivo clinical molecular imaging of T cell activity. Trends Immunol 2023; 44:1031-1045. [PMID: 37932176 DOI: 10.1016/j.it.2023.10.002] [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: 09/21/2023] [Revised: 10/07/2023] [Accepted: 10/09/2023] [Indexed: 11/08/2023]
Abstract
Tumor immunotherapy is refashioning traditional treatments in the clinic for certain tumors, especially by relying on the activation of T cells. However, the safety and effectiveness of many antitumor immunotherapeutic agents are suboptimal due to difficulties encountered in assessing T cell responses and adjusting treatment regimens accordingly. Here, we review advances in the clinical visualization of T cell activity in vivo, and focus particularly on molecular imaging probes and biomarkers of T cell activation. Current challenges and prospects are also discussed that aim to achieve a better strategy for real-time monitoring of T cell activity, predicting prognoses and responses to tumor immunotherapy, and assessing disease management.
Collapse
Affiliation(s)
- Xiaju Cheng
- Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, PR China
| | - Jiahao Shen
- Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, PR China
| | - Jingwei Xu
- Department of Cardiothoracic Surgery, Suzhou Municipal Hospital Institution, Suzhou 215000, PR China.
| | - Jinfeng Zhu
- Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, PR China
| | - Pei Xu
- Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, PR China
| | - Yong Wang
- Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, PR China.
| | - Mingyuan Gao
- Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, PR China.
| |
Collapse
|
24
|
Lamas NJ, Lassalle S, Martel A, Nahon-Estève S, Macocco A, Zahaf K, Lalvee S, Fayada J, Lespinet-Fabre V, Bordone O, Pedeutour F, Baillif S, Hofman P. Characterisation of the protein expression of the emerging immunotherapy targets VISTA, LAG-3 and PRAME in primary uveal melanoma: insights from a southern French patient cohort. Pathology 2023; 55:929-944. [PMID: 37863710 DOI: 10.1016/j.pathol.2023.08.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 07/02/2023] [Accepted: 08/04/2023] [Indexed: 10/22/2023]
Abstract
Uveal melanoma (UM) is the most common intraocular tumour in adults, with dismal prognosis once metastases develop, since therapeutic options for the metastatic disease are ineffective. Over the past decade, novel cancer therapies based on immunotherapy have changed the landscape of treatment of different forms of cancer leading to many hopes of improvement in patient overall survival (OS). VISTA, LAG-3 and PRAME are novel promising targets of immunotherapy that have recently gained attention in different solid tumours, but whose relevance in UM remained to be comprehensively evaluated until now. Here, we studied the protein expression of VISTA, LAG-3 and PRAME using immunohistochemistry in representative whole tissue sections from primary UM cases in a cohort of 30 patients from a single centre (Nice University Hospital, Nice, France). The expression of each of these markers was correlated with different clinical and pathological parameters, including onset of metastases and OS. We demonstrated the protein expression of VISTA and LAG-3 in small lymphocytes infiltrating the tumour, while no expression of the proteins was detected in UM cells. For PRAME, nuclear expression was observed in UM cells, but no expression in tumour infiltrating immune cells was identified. Increased levels of VISTA expression in tumour infiltrating lymphocytes (TILs) were associated with nuclear BAP1 expression and better prognosis. Higher levels of LAG-3 in TILs were associated with higher levels of CD8-positive TILs. PRAME nuclear positivity in melanoma cells was associated with epithelioid cell dominant (>90%) UM histological subtype, higher mitotic numbers and a higher percentage of chromosome 8q gain. This study proposes VISTA as a novel relevant immune checkpoint molecule in primary UM and contributes to confirm LAG-3 and PRAME as potentially important immunotherapy targets in the treatment of UM patients, helping to expand the number of immunotherapy candidate molecules that are relevant to modulate in this aggressive cancer.
Collapse
Affiliation(s)
- Nuno Jorge Lamas
- Université Côte d'Azur, Laboratory of Clinical and Experimental Pathology, Biobank BB-0033-00025, Pasteur Hospital, Centre Hospitalier Universitaire de Nice, Nice, France; Anatomic Pathology Service, Pathology Department, Centro Hospitalar Universitário de Santo António (CHUdSA), Porto, Largo Professor Abel Salazar, Porto, Portugal; Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, Braga, Portugal; ICVS/3B's, PT Government Associate Laboratory, University of Minho, Braga/Guimarães, Portugal
| | - Sandra Lassalle
- Université Côte d'Azur, Laboratory of Clinical and Experimental Pathology, Biobank BB-0033-00025, Pasteur Hospital, Centre Hospitalier Universitaire de Nice, Nice, France; IRCAN Team 4, Inserm U1081/CNRS 7284, Centre de Lutte contre le Cancer Antoine Lacassagne, Nice, France; FHU OncoAge, Centre Hospitalier Universitaire de Nice, Nice, France
| | - Arnaud Martel
- Université Côte d'Azur, Department of Ophthalmology, Pasteur Hospital, Centre Hospitalier Universitaire de Nice, Nice, France
| | - Sacha Nahon-Estève
- Université Côte d'Azur, Department of Ophthalmology, Pasteur Hospital, Centre Hospitalier Universitaire de Nice, Nice, France
| | - Adam Macocco
- Université Côte d'Azur, Laboratory of Clinical and Experimental Pathology, Biobank BB-0033-00025, Pasteur Hospital, Centre Hospitalier Universitaire de Nice, Nice, France
| | - Katia Zahaf
- Université Côte d'Azur, Laboratory of Clinical and Experimental Pathology, Biobank BB-0033-00025, Pasteur Hospital, Centre Hospitalier Universitaire de Nice, Nice, France
| | - Salome Lalvee
- Université Côte d'Azur, Laboratory of Clinical and Experimental Pathology, Biobank BB-0033-00025, Pasteur Hospital, Centre Hospitalier Universitaire de Nice, Nice, France
| | - Julien Fayada
- Université Côte d'Azur, Laboratory of Clinical and Experimental Pathology, Biobank BB-0033-00025, Pasteur Hospital, Centre Hospitalier Universitaire de Nice, Nice, France
| | - Virginie Lespinet-Fabre
- Université Côte d'Azur, Laboratory of Clinical and Experimental Pathology, Biobank BB-0033-00025, Pasteur Hospital, Centre Hospitalier Universitaire de Nice, Nice, France; IRCAN Team 4, Inserm U1081/CNRS 7284, Centre de Lutte contre le Cancer Antoine Lacassagne, Nice, France; FHU OncoAge, Centre Hospitalier Universitaire de Nice, Nice, France
| | - Olivier Bordone
- Université Côte d'Azur, Laboratory of Clinical and Experimental Pathology, Biobank BB-0033-00025, Pasteur Hospital, Centre Hospitalier Universitaire de Nice, Nice, France; IRCAN Team 4, Inserm U1081/CNRS 7284, Centre de Lutte contre le Cancer Antoine Lacassagne, Nice, France; FHU OncoAge, Centre Hospitalier Universitaire de Nice, Nice, France
| | - Florence Pedeutour
- Laboratory of Solid Tumour Genetics, Centre Hospitalier Universitaire de Nice, Université Côte d'Azur, Nice, France
| | - Stéphanie Baillif
- Université Côte d'Azur, Department of Ophthalmology, Pasteur Hospital, Centre Hospitalier Universitaire de Nice, Nice, France
| | - Paul Hofman
- Université Côte d'Azur, Laboratory of Clinical and Experimental Pathology, Biobank BB-0033-00025, Pasteur Hospital, Centre Hospitalier Universitaire de Nice, Nice, France; IRCAN Team 4, Inserm U1081/CNRS 7284, Centre de Lutte contre le Cancer Antoine Lacassagne, Nice, France; FHU OncoAge, Centre Hospitalier Universitaire de Nice, Nice, France.
| |
Collapse
|
25
|
He W, Chen J, Zhou Y, Deng T, Feng Y, Luo X, Zhang C, Huang H, Liu J. Mitophagy genes in ovarian cancer: a comprehensive analysis for improved immunotherapy. Discov Oncol 2023; 14:221. [PMID: 38038814 PMCID: PMC10692064 DOI: 10.1007/s12672-023-00750-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 07/07/2023] [Indexed: 12/02/2023] Open
Abstract
BACKGROUND Mitophagy is a process of selectively degrading damaged mitochondria, which has been found to be related to immunity, tumorigenesis, tumor progression, and metastasis. However, the role of mitophagy-related genes (MRGs) in the tumor microenvironment (TME) of ovarian cancer (OV) remains largely unexplored. METHODS We analyzed the expression, prognosis, and genetic alterations of 29 MRGs in 480 OV samples. Unsupervised clustering was used to classify OV into two subtypes (clusters A and B) based on MRG changes. We compared the clinical features, differential expressed genes (DEGs), pathways, and immune cell infiltration between the two clusters. We constructed a mitophagy scoring system (MRG_score) based on the DEGs and validated its ability to predict overall survival of OV patients. RESULTS We found that patients with high MRG_scores had better survival status and increased infiltration by immune cells. Further analysis showed that these patients may be more sensitive to immune checkpoint inhibitor (ICI) treatment. Additionally, the MRG_score significantly correlated with the sensitivity of chemotherapeutic drugs and targeted inhibitors. CONCLUSION Our comprehensive analysis of MRGs in the TME, clinical features, and patient prognosis revealed that the MRG_score is a potentially effective prognostic biomarker and predictor of treatment. This study provides new insights into the role of MRGs in OV and identifies patients who may benefit from ICI treatment, chemotherapy, or targeted treatment.
Collapse
Affiliation(s)
- Wenting He
- Department of Gynecologic Oncology, State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
| | - Jieping Chen
- Department of Gynecologic Oncology, State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
| | - Yun Zhou
- Department of Gynecologic Oncology, State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
| | - Ting Deng
- Department of Gynecologic Oncology, State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
| | - Yanling Feng
- Department of Gynecologic Oncology, State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
| | - Xiaolin Luo
- Department of Gynecologic Oncology, State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
| | - Chuyao Zhang
- Department of Gynecologic Oncology, State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
| | - He Huang
- Department of Gynecologic Oncology, State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China.
| | - Jihong Liu
- Department of Gynecologic Oncology, State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China.
| |
Collapse
|
26
|
Lu Y, Wang S, Chi T, Zhao Y, Guo H, Wang H, Feng L. DNA damage repair-related gene signature for identifying the immune status and predicting the prognosis of hepatocellular carcinoma. Sci Rep 2023; 13:18978. [PMID: 37923899 PMCID: PMC10624694 DOI: 10.1038/s41598-023-45999-z] [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: 07/31/2023] [Accepted: 10/26/2023] [Indexed: 11/06/2023] Open
Abstract
The heterogeneity of hepatocellular carcinoma (HCC) poses a challenge for accurate prognosis prediction. DNA damage repair genes (DDRGs) have an impact on a wide range of malignancies. However, the relevance of these genes in HCC prognosis has received little attention. In this study, we aimed to develop a prognostic signature to identify novel therapy options for HCC. We acquired mRNA expression profiles and clinical data for HCC patients from The Cancer Genome Atlas (TCGA) database. A polygenic prognostic model for HCC was constructed using selection operator Cox analysis and least absolute shrinkage. The model was validated using International Cancer Genome Consortium (ICGC) data. Overall survival (OS) between the high-risk and low-risk groups was compared using Kaplan‒Meier analysis. Independent predictors of OS were identified through both univariate and multivariate Cox analyses. To determine immune cell infiltration scores and activity in immune-related pathways, a single-sample gene set enrichment analysis was performed. The protein and mRNA expression levels of the prognostic genes between HCC and normal liver tissues were also examined by immunohistochemistry (IHC), immunofluorescence (IF) and quantitative real-time PCR (qRT-PCR). A novel ten-gene signature (CHD1L, HDAC1, KPNA2, MUTYH, PPP2R5B, NEIL3, POLR2L, RAD54B, RUVBL1 and SPP1) was established for HCC prognosis prediction. Patients in the high-risk group had worse OS than those in the low-risk group. Receiver operating characteristic curve analysis confirmed the predictive ability of this prognostic gene signature. Multivariate Cox analysis showed that the risk score was an independent predictor of OS. Functional analysis revealed a strong association with cell cycle and antigen binding pathways, and the risk score was highly correlated with tumor grade, tumor stage, and types of immune infiltrate. High expression levels of the prognostic genes were significantly correlated with increased sensitivity of cancer cells to antitumor drugs. IHC, IF and qRT-PCR all indicated that the prognostic genes were highly expressed in HCC relative to normal liver tissue, consistent with the results of bioinformatics analysis. Ten DDRGs were utilized to create a new signature for identifying the immunological state of HCC and predicting prognosis. In addition, blocking these genes could represent a promising treatment.
Collapse
Affiliation(s)
- Yongpan Lu
- Department of Plastic Surgery, Shandong University of Traditional Chinese Medicine, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qian Foshan Hospital, Jingshi Road, Jinan, 250014, Shandong, China
| | - Sen Wang
- Department of Medical Ultrasound, Shandong Medicine and Health Key Laboratory of Abdominal Medical Imaging, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qian Foshan Hospital, Shandong First Medical University, No. 16766, Jingshi Road, Jinan, 250014, Shandong, China
| | - Tingting Chi
- Department of Acupuncture and Rehabilitation, The Affiliated Qingdao Hai Ci Hospital of Qingdao University (West Hospital Area), Qingdao, 266000, Shandong, China
| | - Yuli Zhao
- Department of Medical Ultrasound, Shandong Medicine and Health Key Laboratory of Abdominal Medical Imaging, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qian Foshan Hospital, Shandong First Medical University, No. 16766, Jingshi Road, Jinan, 250014, Shandong, China
| | - Huimin Guo
- Department of Medical Ultrasound, Shandong Medicine and Health Key Laboratory of Abdominal Medical Imaging, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qian Foshan Hospital, Jining Medical College, No. 16766, Jingshi Road, Jinan, 250014, Shandong, China
| | - Haizheng Wang
- Department of Medical Ultrasound, Shandong Medicine and Health Key Laboratory of Abdominal Medical Imaging, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qian Foshan Hospital, Shandong First Medical University, No. 16766, Jingshi Road, Jinan, 250014, Shandong, China
| | - Li Feng
- Department of Medical Ultrasound, Shandong Medicine and Health Key Laboratory of Abdominal Medical Imaging, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qian Foshan Hospital, Shandong First Medical University, No. 16766, Jingshi Road, Jinan, 250014, Shandong, China.
| |
Collapse
|
27
|
Li Y, Ju M, Miao Y, Zhao L, Xing L, Wei M. Advancement of anti-LAG-3 in cancer therapy. FASEB J 2023; 37:e23236. [PMID: 37846808 DOI: 10.1096/fj.202301018r] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Revised: 08/25/2023] [Accepted: 09/20/2023] [Indexed: 10/18/2023]
Abstract
Immune checkpoint inhibitors have effectively transformed the treatment of many cancers, particularly those highly devastating malignancies. With their widespread popularity, the drawbacks of immune checkpoint inhibitors are also recognized, such as drug resistance and immune-related systematic side effects. Thus, it never stops investigating novel immune checkpoint inhibitors. Lymphocyte Activation Gene-3 (LAG-3) is a well-established co-inhibitory receptor that performs negative regulation on immune responses. Recently, a novel FDA-approved LAG-3 blocking agent, together with nivolumab as a new combinational immunotherapy for metastatic melanoma, brought LAG-3 back into focus. Clinical data suggests that anti-LAG-3 agents can amplify the therapeutic response of other immune checkpoint inhibitors with manageable side effects. In this review, we elucidate the intercellular and intracellular mechanisms of LAG-3, clarify the current understanding of LAG-3 in the tumor microenvironment, identify present LAG-3-associated therapeutic agents, discuss current LAG-3-involving clinical trials, and eventually address future prospects for LAG-3 inhibitors.
Collapse
Affiliation(s)
- Yunong Li
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, P.R. China
| | - Mingyi Ju
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, P.R. China
| | - Yuxi Miao
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, P.R. China
| | - Lin Zhao
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, P.R. China
| | - Lijuan Xing
- Precision Laboratory, Panjin Central Hospital, Panjin, P.R. China
| | - Minjie Wei
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, P.R. China
- Shenyang Kangwei Medical Laboratory Analysis Co. Ltd, Shenyang, P.R. China
| |
Collapse
|
28
|
Taghiloo S, Asgarian-Omran H. Current Approaches of Immune Checkpoint Therapy in Chronic Lymphocytic Leukemia. Curr Treat Options Oncol 2023; 24:1408-1438. [PMID: 37561383 DOI: 10.1007/s11864-023-01129-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/07/2023] [Indexed: 08/11/2023]
Abstract
OPINION STATEMENT Increasing understanding of the complex interaction between leukemic and immune cells, which is responsible for tumor progression and immune evasion, has paved the way for the development of novel immunotherapy approaches in chronic lymphocytic leukemia (CLL). One of the well-known immune escape mechanisms of tumor cells is the up-regulation of immune checkpoint molecules. In recent years, targeting immune checkpoint receptors is the most clinically effective immunotherapeutic strategy for cancer treatment. In this regard, various immune checkpoint blockade (ICB) drugs are currently been investigating for their potential effects on improving anti-tumor immune response and clinical efficacy in the hematological malignancies; however, their effectiveness in patients with CLL has shown less remarkable success, and ongoing research is focused on identifying strategies to enhance the efficacy of ICB in CLL.
Collapse
Affiliation(s)
- Saeid Taghiloo
- Department of Immunology, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Hossein Asgarian-Omran
- Department of Immunology, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran.
- Gastrointestinal Cancer Research Center, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran.
| |
Collapse
|
29
|
Han L, Zhang Y, Li L, Zhang Q, Liu Z, Niu H, Hu J, Ding Z, Shi X, Qian X. Exploring the Expression and Prognosis of Mismatch Repair Proteins and PD-L1 in Colorectal Cancer in a Chinese Cohort. Cancer Manag Res 2023; 15:791-801. [PMID: 37575316 PMCID: PMC10417781 DOI: 10.2147/cmar.s417470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Accepted: 07/14/2023] [Indexed: 08/15/2023] Open
Abstract
Purpose Exploring the expression and prognosis of mismatch repair proteins and PD-L1 in colorectal cancer. Patients and Methods A total of 272 patients with surgically resected CRC were enrolled in the study from January 2018 to May 2022 at Nanjing Drum Tower Hospital (The Affiliated Hospital of Nanjing University Medical School). Surgically resected samples were collected from patients along with general, clinicopathological, and imaging data for each patient. Immunohistochemistry (IHC) was used to detect expression of MSH2, MSH6, MLH1, and PMS2 proteins in tumor tissue. X-squared (X2) testing was performed to investigate the correlation between expression of MMR proteins and PD-L1 in CRC tumor tissues and clinicopathological characteristics. Correlation analysis was also used to compare the deletion of four MMR proteins in CRC tumor tissues. A survival curve and Log rank test were used to investigate the relationship between the expression of MMR proteins and PD-L1 with regard to CRC patient prognosis and survival. Results MMR protein expression deletion was correlated with tumor location, the degree of tissue differentiation, and TNM stage (P<0.05). PD-L1 expression was correlated with TNM stage (P<0.05). Correlation analysis of deletion of MMR protein isoform expression found that PMS2 deletion was significantly correlated with MLH1 deletion (P<0.05). Similarly, MSH2 deletion was significantly correlated with MSH6 deletion (P<0.05). PMS2 deletion was also found to be correlated with PD-L1 expression (P<0.05). Progression-free survival was found to be significantly longer in mismatch repair-proficient (pMMR) patients compared with mismatch repair-deficient (dMMR) patients. Conclusion Deletion of MMR proteins and expression of PD-L1 are closely related to clinicopathological characteristics and overall prognosis of CRC patients. This suggests the relevance of MMR and PD-L1 as potential biomarkers for treatment of CRC patients.
Collapse
Affiliation(s)
- Lu Han
- Department of Oncology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, People’s Republic of China
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University, Nanjing, People’s Republic of China
| | - Yaping Zhang
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University, Nanjing, People’s Republic of China
- Department of Oncology, Nanjing Drum Tower Hospital Clinical College of Xuzhou Medical University, Nanjing, People’s Republic of China
- Department of Pathology, The First People’s Hospital of Yangzhou, Yangzhou, People’s Republic of China
| | - Li Li
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University, Nanjing, People’s Republic of China
| | - Qun Zhang
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University, Nanjing, People’s Republic of China
| | - Zhihao Liu
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University, Nanjing, People’s Republic of China
- Department of Oncology, Nanjing Drum Tower Hospital Clinical College of Xuzhou Medical University, Nanjing, People’s Republic of China
| | - Haiqing Niu
- Department of Oncology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, People’s Republic of China
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University, Nanjing, People’s Republic of China
| | - Jing Hu
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University, Nanjing, People’s Republic of China
| | - Zhou Ding
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University, Nanjing, People’s Republic of China
| | - Xiao Shi
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University, Nanjing, People’s Republic of China
| | - Xiaoping Qian
- Department of Oncology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, People’s Republic of China
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University, Nanjing, People’s Republic of China
| |
Collapse
|
30
|
Xu J, Liu C, Wu X, Ma J. Current immune therapeutic strategies in advanced or metastatic non-small cell lung cancer. Chin Med J (Engl) 2023; 136:1765-1782. [PMID: 37257112 PMCID: PMC10405997 DOI: 10.1097/cm9.0000000000002536] [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: 09/28/2022] [Indexed: 06/02/2023] Open
Abstract
ABSTRACT Immune escape mechanisms in non-small cell lung cancer (NSCLC) can disrupt every step of the anti-cancer immune response. In recent years, an increased understanding of the specific mechanisms fueling immune escape has allowed for the development of numerous immunotherapeutic treatments that have been introduced into the clinical practice. The advent of immunotherapy has dramatically changed the current treatment landscape of advanced or metastatic NSCLC because of its durable efficacy and manageable toxicity. In this review, we will first present a brief overview of recent evidence on immune escape mechanisms in NSCLC. We will then discuss the current promising immunotherapeutic strategies in advanced or metastatic NSCLC tumors.
Collapse
Affiliation(s)
- Jing Xu
- Center of Biotherapy, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Caixia Liu
- Center of Biotherapy, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Xiaonan Wu
- Department of Medical Oncology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Jie Ma
- Center of Biotherapy, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing 100730, China
| |
Collapse
|
31
|
Ellison JM, Nohria A. An Increased Understanding of the Association Between Atherosclerosis and Immune Checkpoint Inhibitors. Curr Cardiol Rep 2023; 25:879-887. [PMID: 37395892 DOI: 10.1007/s11886-023-01908-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/14/2023] [Indexed: 07/04/2023]
Abstract
PURPOSE OF REVIEW Immune checkpoint inhibitor (ICI) therapy activates the host immune system to promote tumor cell death. This activation of the immune system can lead to off-target immune-related adverse events (irAEs). There is an established link between inflammation and atherosclerosis. The purpose of this manuscript is to review the existing body of literature discussing the potential association between ICI treatment and atherosclerosis. RECENT FINDINGS Pre-clinical studies suggest that ICI therapy may lead to T-cell-mediated progression of atherosclerosis. Recent retrospective clinical studies have shown higher rates of myocardial infarction and stroke with ICI therapy, particularly in patients with pre-existing cardiovascular risk factors. Additionally, small observational cohort studies have used imaging modalities to demonstrate higher rates of atherosclerotic progression with ICI treatment. Early pre-clinical and clinical evidence suggests an association between ICI treatment and the progression of atherosclerosis. However, these findings are preliminary, and adequately powered prospective studies are needed to demonstrate a conclusive association. As ICI therapy is increasingly used to treat a variety of solid tumors, it is important to evaluate and mitigate the potential adverse atherosclerotic effects of ICI treatment.
Collapse
Affiliation(s)
- Judah M Ellison
- Cardiovascular Division, Brigham and Women's Hospital, Boston, MA, 02115, USA
| | - Anju Nohria
- Cardiovascular Division, Brigham and Women's Hospital, Boston, MA, 02115, USA.
| |
Collapse
|
32
|
Li S, Hao L, Zhang J, Deng J, Hu X. Focus on T cell exhaustion: new advances in traditional Chinese medicine in infection and cancer. Chin Med 2023; 18:76. [PMID: 37355637 DOI: 10.1186/s13020-023-00785-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 06/16/2023] [Indexed: 06/26/2023] Open
Abstract
In chronic infections and cancers, T lymphocytes (T cells) are exposed to persistent antigen or inflammatory signals. The condition is often associated with a decline in T-cell function: a state called "exhaustion". T cell exhaustion is a state of T cell dysfunction characterized by increased expression of a series of inhibitory receptors (IRs), decreased effector function, and decreased cytokine secretion, accompanied by transcriptional and epigenetic changes and metabolic defects. The rise of immunotherapy, particularly the use of immune checkpoint inhibitors (ICIs), has dramatically changed the clinical treatment paradigm for patients. However, its low response rate, single target and high immunotoxicity limit its clinical application. The multiple immunomodulatory potential of traditional Chinese medicine (TCM) provides a new direction for improving the treatment of T cell exhaustion. Here, we review recent advances that have provided a clearer molecular understanding of T cell exhaustion, revealing the characteristics and causes of T cell exhaustion in persistent infections and cancers. In addition, this paper summarizes recent advances in improving T cell exhaustion in infectious diseases and cancer with the aim of providing a comprehensive and valuable source of information on TCM as an experimental study and their role in collaboration with ICIs therapy.
Collapse
Affiliation(s)
- Shenghao Li
- Hospital of Chengdu University of Traditional Chinese Medicine, No. 39 Shi-Er-Qiao Road, Chengdu, 610072, Sichuan Province, People's Republic of China
- Chengdu University of Traditional Chinese Medicine, No. 37 Shi-Er-Qiao Road, Chengdu, 610075, Sichuan Province, People's Republic of China
| | - Liyuan Hao
- Hospital of Chengdu University of Traditional Chinese Medicine, No. 39 Shi-Er-Qiao Road, Chengdu, 610072, Sichuan Province, People's Republic of China
- Chengdu University of Traditional Chinese Medicine, No. 37 Shi-Er-Qiao Road, Chengdu, 610075, Sichuan Province, People's Republic of China
| | - Junli Zhang
- Hospital of Chengdu University of Traditional Chinese Medicine, No. 39 Shi-Er-Qiao Road, Chengdu, 610072, Sichuan Province, People's Republic of China
- Chengdu University of Traditional Chinese Medicine, No. 37 Shi-Er-Qiao Road, Chengdu, 610075, Sichuan Province, People's Republic of China
| | - Jiali Deng
- Hospital of Chengdu University of Traditional Chinese Medicine, No. 39 Shi-Er-Qiao Road, Chengdu, 610072, Sichuan Province, People's Republic of China
- Chengdu University of Traditional Chinese Medicine, No. 37 Shi-Er-Qiao Road, Chengdu, 610075, Sichuan Province, People's Republic of China
| | - Xiaoyu Hu
- Hospital of Chengdu University of Traditional Chinese Medicine, No. 39 Shi-Er-Qiao Road, Chengdu, 610072, Sichuan Province, People's Republic of China.
| |
Collapse
|
33
|
Miedema IHC, Huisman MC, Zwezerijnen GJC, Grempler R, Pitarch AP, Thiele A, Hesse R, Elgadi M, Peltzer A, Vugts DJ, van Dongen GAMS, de Gruijl TD, Menke-van der Houven van Oordt CW, Bahce I. 89Zr-immuno-PET using the anti-LAG-3 tracer [ 89Zr]Zr-BI 754111: demonstrating target specific binding in NSCLC and HNSCC. Eur J Nucl Med Mol Imaging 2023; 50:2068-2080. [PMID: 36859619 PMCID: PMC10199858 DOI: 10.1007/s00259-023-06164-w] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 02/18/2023] [Indexed: 03/03/2023]
Abstract
PURPOSE Although lymphocyte activation gene-3 (LAG-3) directed therapies demonstrate promising clinical anti-cancer activity, only a subset of patients seems to benefit and predictive biomarkers are lacking. Here, we explored the potential use of the anti-LAG-3 antibody tracer [89Zr]Zr-BI 754111 as a predictive imaging biomarker and investigated its target specific uptake as well as the correlation of its tumor uptake and the tumor immune infiltration. METHODS Patients with head and neck (N = 2) or lung cancer (N = 4) were included in an imaging substudy of a phase 1 trial with BI 754091 (anti-PD-1) and BI 754111 (anti-LAG-3). After baseline tumor biopsy and [18F]FDG-PET, patients were given 240 mg of BI 754091, followed 8 days later by administration of [89Zr]Zr-BI 754111 (37 MBq, 4 mg). PET scans were performed 2 h, 96 h, and 144 h post-injection. To investigate target specificity, a second tracer administration was given two weeks later, this time with pre-administration of 40 (N = 3) or 600 mg (N = 3) unlabeled BI 754111, followed by PET scans at 96 h and 144 h post-injection. Tumor immune cell infiltration was assessed by immunohistochemistry and RNA sequencing. RESULTS Tracer uptake in tumors was clearly visible at the 4-mg mass dose (tumor-to-plasma ratio 1.63 [IQR 0.37-2.89]) and could be saturated by increasing mass doses (44 mg: 0.67 [IQR 0.50-0.85]; 604 mg: 0.56 [IQR 0.42-0.75]), demonstrating target specificity. Tumor uptake correlated to immune cell-derived RNA signatures. CONCLUSIONS [89Zr]Zr-BI-754111 PET imaging shows favorable technical and biological characteristics for developing a potential predictive imaging biomarker for LAG-3-directed therapies. TRIAL REGISTRATION ClinicalTrials.gov , NCT03780725. Registered 19 December 2018.
Collapse
Affiliation(s)
- Iris H C Miedema
- Department of Medical Oncology, Amsterdam UMC location Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV, Amsterdam, the Netherlands
- Imaging and Biomarkers, Cancer Center Amsterdam, De Boelelaan 1117, 1081 HV, Amsterdam, the Netherlands
| | - Marc C Huisman
- Imaging and Biomarkers, Cancer Center Amsterdam, De Boelelaan 1117, 1081 HV, Amsterdam, the Netherlands
- Department of Radiology and Nuclear Medicine, Amsterdam UMC location Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV, Amsterdam, the Netherlands
| | - Gerben J C Zwezerijnen
- Imaging and Biomarkers, Cancer Center Amsterdam, De Boelelaan 1117, 1081 HV, Amsterdam, the Netherlands
- Department of Radiology and Nuclear Medicine, Amsterdam UMC location Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV, Amsterdam, the Netherlands
| | - Rolf Grempler
- Department of Translational Medicine & Clinical Pharmacology, Boehringer Ingelheim Pharmaceuticals, 900 Ridgebury Road, Ridgefield, CT, 06877, USA
| | - Alejandro Perez Pitarch
- Department of Translational Medicine & Clinical Pharmacology, Boehringer Ingelheim Pharma GmbH & Co. KG, Birkendorfer Strasse 65, 88400, Biberach and der Riss, Germany
| | - Andrea Thiele
- Department of Translational Medicine & Clinical Pharmacology, Boehringer Ingelheim Pharma GmbH & Co. KG, Birkendorfer Strasse 65, 88400, Biberach and der Riss, Germany
| | - Raphael Hesse
- Department of Translational Medicine & Clinical Pharmacology, Boehringer Ingelheim Pharma GmbH & Co. KG, Birkendorfer Strasse 65, 88400, Biberach and der Riss, Germany
| | - Mabrouk Elgadi
- Department of Translational Medicine & Clinical Pharmacology, Boehringer Ingelheim Pharmaceuticals, 900 Ridgebury Road, Ridgefield, CT, 06877, USA
| | - Alexander Peltzer
- Department of Translational Medicine & Clinical Pharmacology, Boehringer Ingelheim Pharma GmbH & Co. KG, Birkendorfer Strasse 65, 88400, Biberach and der Riss, Germany
| | - Danielle J Vugts
- Imaging and Biomarkers, Cancer Center Amsterdam, De Boelelaan 1117, 1081 HV, Amsterdam, the Netherlands
- Department of Radiology and Nuclear Medicine, Amsterdam UMC location Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV, Amsterdam, the Netherlands
| | - Guus A M S van Dongen
- Imaging and Biomarkers, Cancer Center Amsterdam, De Boelelaan 1117, 1081 HV, Amsterdam, the Netherlands
- Department of Radiology and Nuclear Medicine, Amsterdam UMC location Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV, Amsterdam, the Netherlands
| | - Tanja D de Gruijl
- Department of Medical Oncology, Amsterdam UMC location Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV, Amsterdam, the Netherlands
- Cancer Biology and Immunology, Cancer Center Amsterdam, De Boelelaan 1117, 1018 HV, Amsterdam, the Netherlands
| | - C Willemien Menke-van der Houven van Oordt
- Department of Medical Oncology, Amsterdam UMC location Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV, Amsterdam, the Netherlands.
- Imaging and Biomarkers, Cancer Center Amsterdam, De Boelelaan 1117, 1081 HV, Amsterdam, the Netherlands.
| | - Idris Bahce
- Imaging and Biomarkers, Cancer Center Amsterdam, De Boelelaan 1117, 1081 HV, Amsterdam, the Netherlands
- Department of Pulmonary Medicine, Amsterdam UMC location Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV, Amsterdam, the Netherlands
| |
Collapse
|
34
|
Koukourakis IM, Platoni K, Tiniakos D, Kouloulias V, Zygogianni A. Immune Response and Immune Checkpoint Molecules in Patients with Rectal Cancer Undergoing Neoadjuvant Chemoradiotherapy: A Review. Curr Issues Mol Biol 2023; 45:4495-4517. [PMID: 37232754 DOI: 10.3390/cimb45050285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 05/17/2023] [Accepted: 05/18/2023] [Indexed: 05/27/2023] Open
Abstract
It is well-established that tumor antigens and molecules expressed and secreted by cancer cells trigger innate and adaptive immune responses. These two types of anti-tumor immunity lead to the infiltration of the tumor's microenvironment by immune cells with either regulatory or cytotoxic properties. Whether this response is associated with tumor eradication after radiotherapy and chemotherapy or regrowth has been a matter of extensive research through the years, mainly focusing on tumor-infiltrating lymphocytes and monocytes and their subtypes, and the expression of immune checkpoint and other immune-related molecules by both immune and cancer cells in the tumor microenvironment. A literature search has been conducted on studies dealing with the immune response in patients with rectal cancer treated with neoadjuvant radiotherapy or chemoradiotherapy, assessing its impact on locoregional control and survival and underlying the potential role of immunotherapy in the treatment of this cancer subtype. Here, we provide an overview of the interactions between local/systemic anti-tumor immunity, cancer-related immune checkpoint, and other immunological pathways and radiotherapy, and how these affect the prognosis of rectal cancer patients. Chemoradiotherapy induces critical immunological changes in the tumor microenvironment and cancer cells that can be exploited for therapeutic interventions in rectal cancer.
Collapse
Affiliation(s)
- Ioannis M Koukourakis
- Radiation Oncology Unit, 1st Department of Radiology, School of Medicine, Aretaieion University Hospital, National and Kapodistrian University of Athens (NKUOA), 11528 Athens, Greece
| | - Kalliopi Platoni
- Medical Physics Unit, 2nd Department of Radiology, School of Medicine, Attikon University Hospital, National and Kapodistrian University of Athens, 12462 Athens, Greece
| | - Dina Tiniakos
- Department of Pathology, School of Medicine, Aretaieion University Hospital, National and Kapodistrian University of Athens, 11528 Athens, Greece
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - Vassilis Kouloulias
- Radiotherapy Unit, 2nd Department of Radiology, School of Medicine, National and Kapodistrian University of Athens, 12462 Athens, Greece
| | - Anna Zygogianni
- Radiation Oncology Unit, 1st Department of Radiology, School of Medicine, Aretaieion University Hospital, National and Kapodistrian University of Athens (NKUOA), 11528 Athens, Greece
| |
Collapse
|
35
|
Villaruz LC, Blumenschein GR, Otterson GA, Leal TA. Emerging therapeutic strategies for enhancing sensitivity and countering resistance to programmed cell death protein 1 or programmed death-ligand 1 inhibitors in non-small cell lung cancer. Cancer 2023; 129:1319-1350. [PMID: 36848319 PMCID: PMC11234508 DOI: 10.1002/cncr.34683] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 10/27/2022] [Accepted: 12/13/2022] [Indexed: 03/01/2023]
Abstract
The availability of agents targeting the programmed cell death protein 1 (PD-1)/programmed death-ligand 1 (PD-L1) immune checkpoint has transformed treatment of advanced and/or metastatic non-small cell lung cancer (NSCLC). However, a substantial proportion of patients treated with these agents do not respond or experience only a brief period of clinical benefit. Even among those whose disease responds, many subsequently experience disease progression. Consequently, novel approaches are needed that enhance antitumor immunity and counter resistance to PD-(L)1 inhibitors, thereby improving and/or prolonging responses and patient outcomes, in both PD-(L)1 inhibitor-sensitive and inhibitor-resistant NSCLC. Mechanisms contributing to sensitivity and/or resistance to PD-(L)1 inhibitors in NSCLC include upregulation of other immune checkpoints and/or the presence of an immunosuppressive tumor microenvironment, which represent potential targets for new therapies. This review explores novel therapeutic regimens under investigation for enhancing responses to PD-(L)1 inhibitors and countering resistance, and summarizes the latest clinical evidence in NSCLC.
Collapse
Affiliation(s)
- Liza C Villaruz
- Division of Hematology/Oncology, Department of Medicine, UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
| | - George R Blumenschein
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Gregory A Otterson
- The Ohio State University-James Comprehensive Cancer Center, Columbus, Ohio, USA
| | - Ticiana A Leal
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, Georgia, USA
| |
Collapse
|
36
|
Silva-Pilipich N, Covo-Vergara Á, Smerdou C. Local Delivery of Immunomodulatory Antibodies for Gastrointestinal Tumors. Cancers (Basel) 2023; 15:cancers15082352. [PMID: 37190279 DOI: 10.3390/cancers15082352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 04/12/2023] [Accepted: 04/13/2023] [Indexed: 05/17/2023] Open
Abstract
Cancer therapy has experienced a breakthrough with the use of immune checkpoint inhibitors (ICIs) based on monoclonal antibodies (mAbs), which are able to unleash immune responses against tumors refractory to other therapies. Despite the great advancement that ICIs represent, most patients with gastrointestinal tumors have not benefited from this therapy. In addition, ICIs often induce adverse effects that are related to their systemic use. Local administration of ICIs in tumors could concentrate their effect in the malignant tissue and provide a higher safety profile. A new and attractive approach for local delivery of ICIs is the use of gene therapy vectors to express these blocking antibodies in tumor cells. Several vectors have been evaluated in preclinical models of gastrointestinal tumors to express ICIs against PD-1, PD-L1, and CTLA-4, among other immune checkpoints, with promising results. Vectors used in these settings include oncolytic viruses, self-replicating RNA vectors, and non-replicative viral and non-viral vectors. The use of viral vectors, especially when they have replication capacity, provides an additional adjuvant effect that has been shown to enhance antitumor responses. This review covers the most recent studies involving the use of gene therapy vectors to deliver ICIs to gastrointestinal tumors.
Collapse
Affiliation(s)
- Noelia Silva-Pilipich
- Division of Gene Therapy and Regulation of Gene Expression, Cima Universidad de Navarra, 31008 Pamplona, Spain
- Instituto de Investigación Sanitaria de Navarra (IdISNA), Cancer Center Clínica Universidad de Navarra (CCUN), 31008 Pamplona, Spain
| | - Ángela Covo-Vergara
- Division of Gene Therapy and Regulation of Gene Expression, Cima Universidad de Navarra, 31008 Pamplona, Spain
- Instituto de Investigación Sanitaria de Navarra (IdISNA), Cancer Center Clínica Universidad de Navarra (CCUN), 31008 Pamplona, Spain
| | - Cristian Smerdou
- Division of Gene Therapy and Regulation of Gene Expression, Cima Universidad de Navarra, 31008 Pamplona, Spain
- Instituto de Investigación Sanitaria de Navarra (IdISNA), Cancer Center Clínica Universidad de Navarra (CCUN), 31008 Pamplona, Spain
| |
Collapse
|
37
|
Zhang C, Zhang C, Wang H. Immune-checkpoint inhibitor resistance in cancer treatment: Current progress and future directions. Cancer Lett 2023; 562:216182. [PMID: 37076040 DOI: 10.1016/j.canlet.2023.216182] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 04/07/2023] [Accepted: 04/11/2023] [Indexed: 04/21/2023]
Abstract
Cancer treatment has been advanced with the advent of immune checkpoint inhibitors (ICIs) exemplified by anti-cytotoxic T-lymphocyte-associated protein 4 (CTLA-4), anti-programmed cell death protein 1 (PD-1) and programmed cell death ligand 1 (PD-L1) drugs. Patients have reaped substantial benefit from ICIs in many cancer types. However, few patients benefit from ICIs whereas the vast majority undergoing these treatments do not obtain survival benefit. Even for patients with initial responses, they may encounter drug resistance in their subsequent treatments, which limits the efficacy of ICIs. Therefore, a deepening understanding of drug resistance is critically important for the explorations of approaches to reverse drug resistance and to boost ICI efficacy. In the present review, different mechanisms of ICI resistance have been summarized according to the tumor intrinsic, tumor microenvironment (TME) and host classifications. We further elaborated corresponding strategies to battle against such resistance accordingly, which include targeting defects in antigen presentation, dysregulated interferon-γ (IFN-γ) signaling, neoantigen depletion, upregulation of other T cell checkpoints as well as immunosuppression and exclusion mediated by TME. Moreover, regarding the host, several additional approaches that interfere with diet and gut microbiome have also been described in reversing ICI resistance. Additionally, we provide an overall glimpse into the ongoing clinical trials that utilize these mechanisms to overcome ICI resistance. Finally, we summarize the challenges and opportunities that needs to be addressed in the investigation of ICI resistance mechanisms, with the aim to benefit more patients with cancer.
Collapse
Affiliation(s)
- Chenyue Zhang
- Department of Integrated Therapy, Fudan University Shanghai Cancer Center, Shanghai Medical College, Shanghai, China
| | - Chenxing Zhang
- Department of Nephrology, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Haiyong Wang
- Department of Internal Medicine-Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China.
| |
Collapse
|
38
|
Awad RM, Breckpot K. Novel technologies for applying immune checkpoint blockers. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2023; 382:1-101. [PMID: 38225100 DOI: 10.1016/bs.ircmb.2023.03.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/03/2023]
Abstract
Cancer cells develop several ways to subdue the immune system among others via upregulation of inhibitory immune checkpoint (ICP) proteins. These ICPs paralyze immune effector cells and thereby enable unfettered tumor growth. Monoclonal antibodies (mAbs) that block ICPs can prevent immune exhaustion. Due to their outstanding effects, mAbs revolutionized the field of cancer immunotherapy. However, current ICP therapy regimens suffer from issues related to systemic administration of mAbs, including the onset of immune related adverse events, poor pharmacokinetics, limited tumor accessibility and immunogenicity. These drawbacks and new insights on spatiality prompted the exploration of novel administration routes for mAbs for instance peritumoral delivery. Moreover, novel ICP drug classes that are adept to novel delivery technologies were developed to circumvent the drawbacks of mAbs. We therefore review the state-of-the-art and novel delivery strategies of ICP drugs.
Collapse
Affiliation(s)
- Robin Maximilian Awad
- Laboratory for Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Karine Breckpot
- Laboratory for Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel, Brussels, Belgium.
| |
Collapse
|
39
|
Chen J, Xu J, Niu Y, Yao L, Liu X, Chen H, Chen S, Wu M, Yu X, Xu P. The elevated expression of LAG-3 on CD8+T cells correlates with disease severity of pulmonary TB. Microb Pathog 2023; 179:106089. [PMID: 37004963 DOI: 10.1016/j.micpath.2023.106089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 03/11/2023] [Accepted: 03/26/2023] [Indexed: 04/03/2023]
Abstract
OBJECTIVE Lymphocyte-activation gene 3 (LAG-3) plays an important role in regulating T-cell responses and inducing peripheral tolerance. Our aim in this study was to investigate the relationship between LAG-3 and active tuberculosis (ATB) and the impact of LAG-3 blockade on CD8+T cells. METHODS Flow cytometry was used to detect the expression of LAG-3 on CD4+T and CD8+T cells in the peripheral blood and bronchoalveolar lavage fluid from ATB patients and to explore the relationship between LAG-3 and ATB. RESULTS The expression of LAG-3 on CD4+T and CD8+T cells in ATB patients was increased (P < 0.001), and CD8+T cells with high expression of LAG-3 were associated with sputum culture results (P < 0.05). We further analyzed the relationship between the expression of LAG-3 in CD8+T cells and the severity of tuberculosis and found that the expression of LAG-3 on CD8+T cells in smear-positive tuberculosis patients was significantly higher than that in sputum smear-negative tuberculosis patients (P < 0.05). LAG-3 expression on CD8+T cells was negatively correlated with the presence of lung lesions (P < 0.05). After stimulation with a tuberculosis-specific antigen, the expression of LAG-3 on tuberculosis-specific CD8+T cells was also upregulated, and LAG-3-expressing CD8+T cells showed reduced production of IFN-γ, decreased activation, and lower proliferation, while the function of CD8+T cells was restored when LAG-3 signaling was blocked. CONCLUSIONS This study further explored the relationship between immune exhaustion caused by LAG-3 and immune escape of Mycobacterium tuberculosis and revealed that the elevated expression of LAG-3 on CD8+T cells correlates with functional defects of CD8+T cells and the severity of pulmonary TB.
Collapse
Affiliation(s)
- Jie Chen
- The Affiliation Infections Diseases Hospital, Suzhou Medical College of Soochow University, SuZhou, China
| | - Junchi Xu
- Department of Clinical Laboratory, The Fifth People's Hospital of Suzhou, Suzhou, China.
| | - Yayan Niu
- Department of Clinical Laboratory, The Fifth People's Hospital of Suzhou, Suzhou, China
| | - Lin Yao
- Department of Clinical Laboratory, The Fifth People's Hospital of Suzhou, Suzhou, China
| | - Xuanmiao Liu
- The Affiliation Infections Diseases Hospital, Suzhou Medical College of Soochow University, SuZhou, China
| | - Hui Chen
- Department of Clinical Laboratory, The Fifth People's Hospital of Suzhou, Suzhou, China
| | - Siyi Chen
- Department of Clinical Laboratory, The Fifth People's Hospital of Suzhou, Suzhou, China
| | - Meiying Wu
- Department of Clinical Laboratory, The Fifth People's Hospital of Suzhou, Suzhou, China.
| | - Xin Yu
- Department of Clinical Laboratory, The Fifth People's Hospital of Suzhou, Suzhou, China.
| | - Ping Xu
- The Affiliation Infections Diseases Hospital, Suzhou Medical College of Soochow University, SuZhou, China.
| |
Collapse
|
40
|
Khadela A, Chavda VP, Postwala H, Ephraim R, Apostolopoulos V, Shah Y. Configuring Therapeutic Aspects of Immune Checkpoints in Lung Cancer. Cancers (Basel) 2023; 15:543. [PMID: 36672492 PMCID: PMC9856297 DOI: 10.3390/cancers15020543] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 12/25/2022] [Accepted: 01/13/2023] [Indexed: 01/18/2023] Open
Abstract
Immune checkpoints are unique components of the body's defense mechanism that safeguard the body from immune responses that are potent enough to harm healthy body cells. When proteins present on the surface of T cells recognize and bind to the proteins present on other tumor cells, immune checkpoints are triggered. These proteins are called immunological checkpoints. The T cells receive an on/off signal when the checkpoints interact with companion proteins. This might avert the host's immune system from eliminating cancer cells. The standard care plan for the treatment of non-small cell lung cancer (NSCLC) has been revolutionized with the use of drugs targeting immune checkpoints, in particular programmed cell death protein 1. These drugs are now extended for their potential to manage SCLC. However, it is acknowledged that these drugs have specific immune related adverse effects. Herein, we discuss the use of immune checkpoint inhibitors in patients with NSCLC and SCLC, their outcomes, and future perspectives.
Collapse
Affiliation(s)
- Avinash Khadela
- Department of Pharmacology, L. M. College of Pharmacy, Navrangpura, Ahmedabad 380009, Gujarat, India
| | - Vivek P. Chavda
- Department of Pharmaceutics and Pharmaceutical Technology, L. M. College of Pharmacy, Navrangpura, Ahmedabad 380009, Gujarat, India
| | - Humzah Postwala
- Pharm. D Section, L. M. College of Pharmacy, Navrangpura, Ahmedabad 380009, Gujarat, India
| | - Ramya Ephraim
- Institute for Health and Sport, Victoria University, Melbourne, VIC 3030, Australia
| | - Vasso Apostolopoulos
- Institute for Health and Sport, Victoria University, Melbourne, VIC 3030, Australia
- Australian Institute for Musculoskeletal Science, Melbourne, VIC 3021, Australia
| | - Yesha Shah
- Pharm. D Section, L. M. College of Pharmacy, Navrangpura, Ahmedabad 380009, Gujarat, India
| |
Collapse
|
41
|
Review to Understand the Crosstalk between Immunotherapy and Tumor Metabolism. MOLECULES (BASEL, SWITZERLAND) 2023; 28:molecules28020862. [PMID: 36677919 PMCID: PMC9863813 DOI: 10.3390/molecules28020862] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/12/2022] [Revised: 01/11/2023] [Accepted: 01/13/2023] [Indexed: 01/19/2023]
Abstract
Immune checkpoint inhibitors have ushered in a new era of cancer treatment by increasing the likelihood of long-term survival for patients with metastatic disease and by introducing fresh therapeutic indications in cases where the disease is still in its early stages. Immune checkpoint inhibitors that target the proteins cytotoxic T-lymphocyte-associated antigen-4 (CTLA-4) or programmed death-1/programmed death ligand-1 have significantly improved overall survival in patients with certain cancers and are expected to help patients achieve complete long-lasting remissions and cures. Some patients who receive immune checkpoint inhibitors, however, either experience therapeutic failure or eventually develop immunotherapy resistance. Such individuals are common, which necessitates a deeper understanding of how cancer progresses, particularly with regard to nutritional regulation in the tumor microenvironment (TME), which comprises metabolic cross-talk between metabolites and tumor cells as well as intracellular metabolism in immune and cancer cells. Combination of immunotherapy with targeted metabolic regulation might be a focus of future cancer research despite a lack of existing clinical evidence. Here, we reviewed the significance of the tumor microenvironment and discussed the most significant immunological checkpoints that have recently been identified. In addition, metabolic regulation of tumor immunity and immunological checkpoints in the TME, including glycolysis, amino acid metabolism, lipid metabolism, and other metabolic pathways were also incorporated to discuss the possible metabolism-based treatment methods being researched in preclinical and clinical settings. This review will contribute to the identification of a relationship or crosstalk between tumor metabolism and immunotherapy, which will shed significant light on cancer treatment and cancer research.
Collapse
|
42
|
Meggyes M, Feik T, Nagy DU, Polgar B, Szereday L. CD8 and CD4 Positive NKT Subpopulations and Immune-Checkpoint Pathways in Early-Onset Preeclampsia and Healthy Pregnancy. Int J Mol Sci 2023; 24:ijms24021390. [PMID: 36674905 PMCID: PMC9863229 DOI: 10.3390/ijms24021390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 01/06/2023] [Accepted: 01/07/2023] [Indexed: 01/13/2023] Open
Abstract
Although many studies have investigated the clinical aspect of early-onset preeclampsia, our knowledge about the immunological consequences of improper placenta development is scarce. The maternal immunotolerance against the fetus is greatly influenced by the Th1 predominance developed by the mother's immune system. Thirty-two early-onset preeclamptic and fifty-one healthy pregnant women with appropriately matched gestational age were involved in our study. Mononuclear cells were separated from peripheral venous blood and the frequency of CD8⁺, CD4⁺, double positive (DP), and double negative (DN) NKT cell subpopulations was determined using multicolor flow cytometry. Following the characterization, the expression levels of different immune checkpoint receptors and ligands were also defined. Soluble CD226 levels were quantified by ELISA. Novel and significant differences were revealed among the ratios of the investigated NKT subsets and in the expression patterns of PD-1, LAG-3, TIGIT and CD226 receptors. Further differences were determined in the expression of CD112, PD-1, LAG-3 and CD226 MFI values between the early-onset preeclamptic and the healthy pregnant groups. Our results suggest that the investigated NKT subpopulations act differently in the altered immune condition characteristic of early-onset preeclampsia and indicate that the different subsets may contribute to the compensation or maintenance of Th1 predominance.
Collapse
Affiliation(s)
- Matyas Meggyes
- Department of Medical Microbiology and Immunology, Medical School, University of Pecs, 12 Szigeti Street, 7624 Pecs, Hungary
- Janos Szentagothai Research Centre, 20 Ifjusag Street, 7624 Pecs, Hungary
- Correspondence:
| | - Timoteus Feik
- Department of Medical Microbiology and Immunology, Medical School, University of Pecs, 12 Szigeti Street, 7624 Pecs, Hungary
| | - David U. Nagy
- Institute of Geobotany/Plant Ecology, Martin-Luther-University, Große Steinstraße 79/80, 06108 Halle (Saale), Germany
| | - Beata Polgar
- Department of Medical Microbiology and Immunology, Medical School, University of Pecs, 12 Szigeti Street, 7624 Pecs, Hungary
- Janos Szentagothai Research Centre, 20 Ifjusag Street, 7624 Pecs, Hungary
| | - Laszlo Szereday
- Department of Medical Microbiology and Immunology, Medical School, University of Pecs, 12 Szigeti Street, 7624 Pecs, Hungary
- Janos Szentagothai Research Centre, 20 Ifjusag Street, 7624 Pecs, Hungary
| |
Collapse
|
43
|
Gao J, Wang D, Yang Q, Tang M, Du J, He L, Liu W. The signature of pyroptosis-related gene prognostic and immune microenvironment in adrenocortical carcinoma. Front Mol Biosci 2023; 10:1131402. [PMID: 36911522 PMCID: PMC9998516 DOI: 10.3389/fmolb.2023.1131402] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2022] [Accepted: 02/06/2023] [Indexed: 02/26/2023] Open
Abstract
Adrenocortical carcinoma (ACC) has a low incidence but a poor prognosis. And ACC has complex clinical manifestations and limited treatment. Pyroptosis has a dual character and has both positive and negative effects on cancer. However, the role of pyroptosis-related genes (PRGs) in ACC and the impact on ACC progression remains unelucidated. This study performed systematic bioinformatics analysis and basic experimental validation to enable the establishment of prognostic models and demonstrate levels of immune infiltration. Pearson's correlation analysis was used to assess the association of PRGs with tumor immune infiltration, tumor mutation burden (TMB), microsatellite instability (MSI), and immune checkpoints. There 4 PRGs were upregulated, and 25 PRGs were downregulated in ACC. At the same time, we analyzed and reviewed the genetic mutation variation landscape of PRGs. Functional enrichment analysis was also performed to clarify the function of PRGs. Pyroptosis, the inflammatory response, the Toll-like receptor signaling pathway, and the NOD-like receptor signaling pathway are the functions and pathways mainly involved and exerted effects by these 33 PRGs. The results of the prognosis analysis revealed high expression of CASP3, CASP9, GSDMB, GSDMD, NLRC4, PRKACA, and SCAF11 caused a poor survival rate for ACC patients. The above seven PRGs were screened by the optimal λ value of LASSO Cox analysis, and the five selected genes (CASP3, CASP9, GSDMB, GSDMD, NLRC4) were involved in constructing a prognostic PRGs model which enables the overall survival in ACC patients can be predicted with moderate to high accuracy. Prognostic PRGs, especially CASP9, which is the independent factor of ACC prognosis, may be closely correlated with immune-cell infiltration, tumor mutation burden, microsatellite instability, and immune checkpoints. Quantitative Real-Time PCR (qRT-PCR), Western blot and immunohistochemical were performed to validate the mRNA expression levels of CASP9 in adjacent normal tissues and ACC tissues. According to the result of immune checkpoints analysis, NLRC4 and GSDMB may be identified as potential therapeutic targets. In conclusion, we established a prognostic model of PRG characteristics in ACC and analyzed the relationship between PRGs and immune infiltration. Through our study, it may be helpful to find the mechanism of pyroptosis in ACC.
Collapse
Affiliation(s)
- Jun Gao
- Department of Urology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Dai Wang
- Xiangya School of Pharmacy, Central South University, Changsha, China
| | - Qingping Yang
- Department of Pharmacy, Zunyi Medical University, Zunyi, China
| | - Mengjie Tang
- Department of Pathology, Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Jiayi Du
- Department of Pharmacy, Fuqing City Hospital of Fujian, Fuqing, China
| | - Leye He
- Department of Urology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Wei Liu
- Department of Pharmacy, The Third Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
44
|
Vatankhah F, Salimi N, Khalaji A, Baradaran B. Immune checkpoints and their promising prospect in cholangiocarcinoma treatment in combination with other therapeutic approaches. Int Immunopharmacol 2023; 114:109526. [PMID: 36481527 DOI: 10.1016/j.intimp.2022.109526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 11/21/2022] [Accepted: 11/27/2022] [Indexed: 12/12/2022]
Abstract
Cholangiocarcinoma (CCA) is one of the malignant tumors that has shown rapid development in incidence and mortality in recent years. Like other types of cancer, patients with CCA experience alterations in the expression of immune checkpoints, indicating the importance of immune checkpoint inhibitors in treating CCA. The results of TCGA analysis in this study revealed a marginal difference in the expression of important immune checkpoints, Programmed cell death 1 (PD-1) and Cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) and their ligands in CCA samples compared to normal ones. This issue showed the importance of combination therapy in this cancer. This review considers CCA treatment and covers several therapeutic modalities or combined treatment strategies. We also cover the most recent developments in the field and outline the important areas of immune checkpoint molecules as prognostic variables and therapeutic targets in CCA.
Collapse
Affiliation(s)
- Fatemeh Vatankhah
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Neda Salimi
- School of Pharmacy, Guilan University of Medical Sciences, Rasht, Iran
| | - Amirreza Khalaji
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
45
|
Suero-Abreu GA, Zanni MV, Neilan TG. Atherosclerosis With Immune Checkpoint Inhibitor Therapy: Evidence, Diagnosis, and Management: JACC: CardioOncology State-of-the-Art Review. JACC CardioOncol 2022; 4:598-615. [PMID: 36636438 PMCID: PMC9830225 DOI: 10.1016/j.jaccao.2022.11.011] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2022] [Accepted: 11/13/2022] [Indexed: 12/24/2022] Open
Abstract
As the clinical applications of immune checkpoint inhibitors (ICIs) expand, our knowledge of the potential adverse effects of these drugs continues to broaden. Emerging evidence supports the association between ICI therapy with accelerated atherosclerosis and atherosclerotic cardiovascular (CV) events. We discuss the biological plausibility and the clinical evidence supporting an effect of inhibition of these immune checkpoints on atherosclerotic CV disease. Further, we provide a perspective on potential diagnostic and pharmacological strategies to reduce atherosclerotic risk in ICI-treated patients. Our understanding of the pathophysiology of ICI-related atherosclerosis is in its early stages. Further research is needed to identify the mechanisms linking ICI therapy to atherosclerosis, leverage the insight that ICI therapy provides into CV biology, and develop robust approaches to manage the expanding cohort of patients who may be at risk for atherosclerotic CV disease.
Collapse
Affiliation(s)
| | - Markella V. Zanni
- Metabolism Unit, Division of Endocrinology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Tomas G. Neilan
- Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA,Cardiovascular Imaging Research Center, Department of Radiology and Department of Cardiology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA,Address for correspondence: Dr Tomas G. Neilan, Cardio-Oncology Program and Cardiovascular Imaging Research Center (CIRC), Massachusetts General Hospital, 165 Cambridge Street, Suite 400, Boston, Massachusetts 02114, USA. @TomasNeilan
| |
Collapse
|
46
|
Andrews LP, Cillo AR, Karapetyan L, Kirkwood JM, Workman CJ, Vignali DAA. Molecular Pathways and Mechanisms of LAG3 in Cancer Therapy. Clin Cancer Res 2022; 28:5030-5039. [PMID: 35579997 PMCID: PMC9669281 DOI: 10.1158/1078-0432.ccr-21-2390] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 03/29/2022] [Accepted: 05/05/2022] [Indexed: 01/24/2023]
Abstract
Immunotherapy targeting coinhibitory receptors has been highly successful in treating a wide variety of malignancies; however, only a subset of patients exhibits durable responses. The first FDA-approved immunotherapeutics targeting coinhibitory receptors PD1 and CTLA4, alone or in combination, significantly improved survival but were also accompanied by substantial toxicity in combination. The third FDA-approved immune checkpoint inhibitor targets LAG3, a coinhibitory receptor expressed on activated CD4+ and CD8+ T cells, especially in settings of long-term antigenic stimulation, such as chronic viral infection or cancer. Mechanistically, LAG3 expression limits both the expansion of activated T cells and the size of the memory pool, suggesting that LAG3 may be a promising target for immunotherapy. Importantly, the mechanism(s) by which LAG3 contributes to CD8+ T-cell exhaustion may be distinct from those governed by PD1, indicating that the combination of anti-LAG3 and anti-PD1 may synergistically enhance antitumor immunity. Clinical studies evaluating the role of anti-LAG3 in combination with anti-PD1 are underway, and recent phase III trial results in metastatic melanoma demonstrate both the efficacy and safety of this combination. Further ongoing clinical trials are evaluating this combination across multiple tumor types and the adjuvant setting, with accompanying translational and biomarker-focused studies designed to elucidate the molecular pathways that lead to improved antitumor T-cell responses following dual blockade of PD1 and LAG3. Overall, LAG3 plays an important role in limiting T-cell activation and has now become part of the repertoire of combinatorial immunotherapeutics available for the treatment of metastatic melanoma.
Collapse
Affiliation(s)
- Lawrence P Andrews
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
- Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania
| | - Anthony R Cillo
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
- Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania
| | - Lilit Karapetyan
- Division of Hematology and Oncology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - John M Kirkwood
- Division of Hematology and Oncology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
- Cancer Immunology and Immunotherapy Program, UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania
| | - Creg J Workman
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
- Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania
| | - Dario A A Vignali
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
- Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania
- Cancer Immunology and Immunotherapy Program, UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania
| |
Collapse
|
47
|
Perez-Santos M, Anaya-Ruiz M, Villafaña-Diaz L, Sánchez Esgua G. Approaches for development of LAG-3 inhibitors and the promise they hold as anticancer agents. Expert Opin Drug Discov 2022; 17:1341-1355. [PMID: 36399656 DOI: 10.1080/17460441.2022.2148652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
INTRODUCTION LAG-3 is considered to be the third point of immunological control in relation to clinical trials that address cancer treatment, only behind PD-1 and CTLA-4, due to its role as a suppressor of the immune response and enhancer of differentiation of Treg cells. AREAS COVERED The authors focus on emphasizing the strategy of development of LAG-3 inhibitors to develop anticancer therapeutics, especially from the perspective of designing new monoclonal and bispecific antibodies against LAG-3. This article also covers details of patents and clinical trials of LAG-3 inhibitors reported in the literature. In addition, we highlight as future research challenges the design and development of peptides and small molecules as inhibitors of LAG-3 function. EXPERT OPINION Three approaches have been used for the development of LAG-3 inhibitors, and they include inhibitory LAG-3 binding peptides and antagonist monoclonal and multispecific antibodies. These approaches include more than 100 clinical trials of 21 molecules that bind to LAG-3 and block its binding to MHC II. However, these approaches do not cover the design and development of peptides and small molecules that could inhibit the function of LAG-3, for which it is necessary to develop new alternatives that cover this gap.
Collapse
Affiliation(s)
- Martin Perez-Santos
- Dirección de Innovación y Transferencia de Conocimiento, Benemérita Universidad Autónoma de Puebla, Puebla CP, México
| | - Maricruz Anaya-Ruiz
- Laboratorio de Biología Celular, Centro de Investigación Biomédica de Oriente, Instituto Mexicano del Seguro Social, Metepec, Puebla CP, México
| | - Luis Villafaña-Diaz
- Centro de Investigación en Inteligencia de Negocios, Universidad Popular Autónoma del Estado de Puebla, Puebla, México
| | - Gabriela Sánchez Esgua
- Dirección de Innovación y Transferencia de Conocimiento, Benemérita Universidad Autónoma de Puebla, Puebla CP, México
| |
Collapse
|
48
|
MUTYH is a potential prognostic biomarker and correlates with immune infiltrates in hepatocellular carcinoma. LIVER RESEARCH 2022. [DOI: 10.1016/j.livres.2022.12.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
49
|
Shi N, Zhou Y, Liu Y, Zhang R, Jiang X, Ren C, Gao X, Luo L. PD-1/LAG-3 bispecific antibody potentiates T cell activation and increases antitumor efficacy. Front Immunol 2022; 13:1047610. [PMID: 36518768 PMCID: PMC9742559 DOI: 10.3389/fimmu.2022.1047610] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Accepted: 11/14/2022] [Indexed: 11/29/2022] Open
Abstract
Several clinical studies demonstrate that there exist other immune checkpoints overexpressed in some PD-1 inhibitor-resistant tumor patients. Among them, Lymphocyte-activation gene 3 (LAG-3) is one of the important immune checkpoint molecules and has been clinically demonstrated to have synergistic anti-tumor effects in combination with PD-1 antibody. In this study, we designed a novel 'knob-in-hole' PD-1/LAG-3 bispecific antibody (BsAb) YG-003D3. In conclusion, the BsAb maintained the similar affinity and thermal stability to the parental antibody, and the BsAb structure can be independent of each other in the process of double-target recognition, and the recognition activity will not be affected. Moreover, the BsAb can not only target PD-1 and LAG-3 on single cell simultaneously, but also bridge the two kinds of cells expressing PD-1 and LAG-3, so as to release the 'brake system of immune checkpoints' and activate immune cells to exert anti-tumor effects more effectively. Especially in the PBMCs activation assay, YG-003D3 induced stronger IFN-γ, IL-6, and TNF-α secretion compared to anti-PD-1 or anti-LAG-3 single drug group or even combined drug group. In the tumor killing experiment of PBMC in vitro, YG-003D3 has a better ability to activate PBMC to kill tumor cells than anti-PD-1 or anti-LAG-3 single drug group or even combined drug group, and the killing rate is as high as 20%. In a humanized PD-1/LAG-3 transgenic mouse subcutaneous tumor-bearing model, YG-003D3 showed good anti-tumor activity, even better than that of the combination group at the same molar concentration. Further studies have shown that YG-003D3 could significantly alter the proportion of immune cells in the tumor microenvironment. In particular, the proportion of CD45+, CD3+ T, CD8+ T cells in tumor tissue and the proportion of CD3+ T, CD8+ T, CD4+ T cells in peripheral blood were significantly increased. These results suggest that YG-003D3 exerts a potent antitumor effect by activating the body 's immune system. In summary, the BsAb YG-003D3 has good anti-tumor activity, which is expected to become a novel drug candidate for cancer immunotherapy.
Collapse
Affiliation(s)
- Ning Shi
- National Health Commission (NHC) Key Laboratory of Carcinogenesis, Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China,State Key Laboratory of Toxicology and Medical Countermeasures, Institute of Pharmacology and Toxicology, Beijing, China,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China,Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China,Cancer Research Institute, School of Basic Medicine, Central South University, Changsha, Hunan, China
| | - Yangyihua Zhou
- State Key Laboratory of Toxicology and Medical Countermeasures, Institute of Pharmacology and Toxicology, Beijing, China,Hunan Normal University School of Medicine, Changsha, Hunan, China
| | - Yujun Liu
- State Key Laboratory of Toxicology and Medical Countermeasures, Institute of Pharmacology and Toxicology, Beijing, China
| | - Ran Zhang
- Hunan Normal University School of Medicine, Changsha, Hunan, China
| | - Xingjun Jiang
- National Health Commission (NHC) Key Laboratory of Carcinogenesis, Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China,Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Caiping Ren
- National Health Commission (NHC) Key Laboratory of Carcinogenesis, Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China,Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China,Cancer Research Institute, School of Basic Medicine, Central South University, Changsha, Hunan, China,*Correspondence: Caiping Ren, ; Xiang Gao, ; Longlong Luo,
| | - Xiang Gao
- State Key Laboratory of Toxicology and Medical Countermeasures, Institute of Pharmacology and Toxicology, Beijing, China,*Correspondence: Caiping Ren, ; Xiang Gao, ; Longlong Luo,
| | - Longlong Luo
- State Key Laboratory of Toxicology and Medical Countermeasures, Institute of Pharmacology and Toxicology, Beijing, China,*Correspondence: Caiping Ren, ; Xiang Gao, ; Longlong Luo,
| |
Collapse
|
50
|
Litière S, Bogaerts J. Imaging endpoints for clinical trial use: a RECIST perspective. J Immunother Cancer 2022; 10:jitc-2022-005092. [PMID: 36424032 PMCID: PMC9693866 DOI: 10.1136/jitc-2022-005092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/11/2022] [Indexed: 11/27/2022] Open
Abstract
Twenty years after its initial introduction, Response Evaluation Criteria in Solid Tumors (RECIST) remains today a unique standardized tool allowing uniform objective evaluation of response in solid tumors in clinical trials across different treatment indications. Several attempts have been made to update or replace RECIST, but none have realized the general traction or uptake seen with RECIST. This communication provides an overview of some challenges faced by RECIST in the rapidly changing oncology landscape, including the incorporation of PET with 18F-fluorodeoxyglucose tracer as a tool for response assessment and the validation of criteria for use in trials involving immunotherapeutics. The latter has mainly been slow due to lack of data sharing. Work is ongoing to try to address this.We also aim to share our view as statistician representatives on the RECIST Working Group on what would be needed to validate new imaging endpoints for clinical trial use, with a specific focus on RECIST. Whether this could lead to an update of RECIST or replace RECIST altogether, depends on the changes being proposed. The ultimate goal remains to have a well defined, repeatable, confirmable and objective standard as provided by RECIST today.
Collapse
Affiliation(s)
- Saskia Litière
- European Organisation for Research and Treatment of Cancer, Brussels, Belgium
| | - Jan Bogaerts
- European Organisation for Research and Treatment of Cancer, Brussels, Belgium
| |
Collapse
|