1
|
Deng W, Wu L, Chen L, Wang K, Lin N, Zhu L, Chen J. Development of B7-H3 targeted CAR-T cells for renal cell carcinoma therapy: in vitro and in vivo efficacy. Clin Transl Oncol 2025; 27:2667-2678. [PMID: 39560834 DOI: 10.1007/s12094-024-03792-y] [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: 08/21/2024] [Accepted: 11/08/2024] [Indexed: 11/20/2024]
Abstract
PURPOSE This study aims to develop chimeric antigen receptor (CAR)-T cells specifically targeting B7-H3-expressing renal cell carcinoma (RCC) and to evaluate the feasibility of B7-H3 CAR-T therapy for RCC. METHODS We analyzed B7-H3 expression in RCC using bioinformatics approaches and confirmed it in tissues and cell lines through immunohistochemical staining and Western blot analysis. A lentiviral vector containing a B7-H3 specific CAR was constructed and transfected into human T cells, with CAR expression verified by flow cytometry. Cytotoxic efficacy was evaluated in co-culture experiments, measuring the production of interferon-gamma (IFN-γ), interleukin-2 (IL-2), granzyme B, and lactate dehydrogenase (LDH) release. Xenograft models in nude mice were used to evaluate tumor growth inhibition by B7-H3 CAR-T cells. RESULTS B7-H3 was significantly expressed in RCC and associated with poor prognosis. Elevated levels of B7-H3 expression were validated in both RCC tissues and cell lines. A B7-H3-specific CAR-T cell was developed, achieving a CAR transduction efficiency of 39.85%, as assessed by flow cytometry. In vitro co-culture assays demonstrated that the CAR-T cells exhibited substantial cytotoxic activity against RCC cell lines, with this activity positively correlating with the effector-to-target ratio. Furthermore, the secretion levels of IFN-γ, IL-2, granzyme B, and LDH were significantly increased compared to the control groups. In vivo experiments further confirmed that B7-H3 CAR-T cells significantly inhibited tumor growth. CONCLUSION The current study suggests that B7-H3 CAR-T cells exhibit significant efficacy in targeting and eliminating RCC cells, indicating a promising cellular immunotherapy approach for RCC treatment.
Collapse
Affiliation(s)
- Wenyi Deng
- Institute of Clinical Medicine, the Second Affiliated Hospital of Hainan Medical University, 368 Yehai Avenue, Haikou, 570311, Hainan, China
| | - Lvying Wu
- Institute of Clinical Medicine, the Second Affiliated Hospital of Hainan Medical University, 368 Yehai Avenue, Haikou, 570311, Hainan, China
| | - Liuyan Chen
- Institute of Clinical Medicine, the Second Affiliated Hospital of Hainan Medical University, 368 Yehai Avenue, Haikou, 570311, Hainan, China
| | - Kuanyin Wang
- Minimally Invasive Urology and Translational Medicine Center, Fuzhou First General Hospital Affiliated With Fujian Medical University, 190th Dadao Road, Fuzhou, 350009, Fujian, China
- Department of Urology, the Second Affiliated Hospital of Hainan Medical University, Haikou, 570311, Hainan, China
| | - Na Lin
- Minimally Invasive Urology and Translational Medicine Center, Fuzhou First General Hospital Affiliated With Fujian Medical University, 190th Dadao Road, Fuzhou, 350009, Fujian, China
| | - Lingfeng Zhu
- Minimally Invasive Urology and Translational Medicine Center, Fuzhou First General Hospital Affiliated With Fujian Medical University, 190th Dadao Road, Fuzhou, 350009, Fujian, China.
- Department of Urology, the Second Affiliated Hospital of Hainan Medical University, Haikou, 570311, Hainan, China.
| | - Jin Chen
- Institute of Clinical Medicine, the Second Affiliated Hospital of Hainan Medical University, 368 Yehai Avenue, Haikou, 570311, Hainan, China.
- Minimally Invasive Urology and Translational Medicine Center, Fuzhou First General Hospital Affiliated With Fujian Medical University, 190th Dadao Road, Fuzhou, 350009, Fujian, China.
| |
Collapse
|
2
|
Habibi S, Bahramian S, Saeedeh ZJ, Mehri S, Ababzadeh S, Kavianpour M. Novel strategies in breast cancer management: From treatment to long-term remission. Crit Rev Oncol Hematol 2025; 211:104715. [PMID: 40187709 DOI: 10.1016/j.critrevonc.2025.104715] [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: 03/02/2025] [Revised: 03/22/2025] [Accepted: 03/26/2025] [Indexed: 04/07/2025] Open
Abstract
Breast cancer (BC) is the most common malignancy among women and a leading cause of cancer-related mortality worldwide. Although improvements in early detection and therapy have been made, metastatic breast cancer (mBC) continues to be an incurable disease. Although existing treatments can prolong survival and enhance quality of life, they do not provide a definitive cure. Targeted therapies have significantly improved outcomes, particularly for subtypes such as human epidermal growth factor receptor 2 (HER2)-positive and hormone receptor (HR)-positive (HR+) BC. Key innovations include antibodydrug conjugates (ADCs) and next-generation endocrine therapies. ADCs combine monoclonal antibodies with cytotoxic agents, allowing targeted delivery to tumor cells while minimizing systemic toxicity. Immunotherapy is emerging as a promising approach for aggressive subtypes, such as triple-negative breast cancer (TNBC). Strategies under investigation include chimeric antigen receptor T-cell (CAR-T) therapy, tumor-infiltrating lymphocyte (TIL) therapies, and natural killer (NK) cell treatments, all aimed at enhancing the ability of the immune system to target and eliminate resistant tumor cells. Tissue engineering, particularly hydrogel-based delivery systems, offers the potential for localized treatment. These systems enable the controlled release of therapeutic agents or immune cells directly to the tumor site, supporting tissue regeneration and enhancing immune surveillance to reduce recurrence. Despite these advancements, challenges remain, including treatment resistance, the immunosuppressive tumor microenvironment, and high costs. Overcoming these barriers requires further innovation in drug delivery systems and a deeper understanding of tumor biology.
Collapse
Affiliation(s)
- Sina Habibi
- Department of Hematology and Blood Banking, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Shabbou Bahramian
- Department of Tissue Engineering and Applied Cell Sciences, School of Medicine, Qom University of Medical Sciences, Qom, Iran
| | - Zare Jalise Saeedeh
- Department of Tissue Engineering and Applied Cell Sciences, School of Medicine, Qom University of Medical Sciences, Qom, Iran
| | - Sara Mehri
- Department of Biology, Faculty of Sciences, University of Guilan, Rasht, Guilan, Iran
| | - Shima Ababzadeh
- Department of Tissue Engineering and Applied Cell Sciences, School of Medicine, Qom University of Medical Sciences, Qom, Iran; Cellular and Molecular Research Center, Qom University of Medical Sciences, Qom, Iran
| | - Maria Kavianpour
- Department of Tissue Engineering and Applied Cell Sciences, School of Medicine, Qom University of Medical Sciences, Qom, Iran; Cellular and Molecular Research Center, Qom University of Medical Sciences, Qom, Iran.
| |
Collapse
|
3
|
Heinze CM, Pichon TJ, Wu AY, Baldwin M, Matthaei J, Song K, Sylvestre M, Gustafson J, White NJ, Jensen MC, Pun SH. Spatial Control of CAR T Cell Activation Using Tumor-Homing Polymers. J Am Chem Soc 2025; 147:5149-5161. [PMID: 39902740 PMCID: PMC11995850 DOI: 10.1021/jacs.4c15442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2025]
Abstract
CAR T cell therapies often lack specificity, leading to issues ranging from inadequate antigen targeting to off-tumor toxicities. To counter that lack of specificity, we expanded tumor targeting capabilities with universal CAR and spatially defined CAR T cell engagement with targets through a combination of synthetic biology and biomaterial approaches. We developed a novel framework, called "In situ Mobilization: Polymer Activated Cell Therapies" (IMPACT) for polymer-mediated, anatomical control of IF-THEN gated CAR T cells. With IMPACT, a regulated payload such as a BiTE or tumor-targeting CAR will only be expressed after engineered cells engage a tumor-localizing polymer ("IF" condition). In this first demonstration of IMPACT, we engineered CAR T cells to respond to fluorescein that is displayed by an injectable polymer that binds to and is retained in fibrin deposits in tumor microenvironments. This interaction then drives selective and conditional expression of a protein within tumors ("THEN" condition). Here, we develop the polymer and CAR T cell infrastructure of IMPACT and demonstrate tumor-localized CAR T cell activation in a murine tumor model after the intravenous administration of polymer and engineered T cells.
Collapse
Affiliation(s)
- Clinton M Heinze
- Department of Bioengineering and Molecular Engineering and Sciences Institute, University of Washington, 3720 15th Avenue NE, Box 355061, Seattle, Washington 98195, United States
- Seattle Children's Therapeutics, Seattle Children's Research Institute, 1920 Terry Ave, Seattle, Washington 98101, United States
| | - Trey J Pichon
- Department of Bioengineering and Molecular Engineering and Sciences Institute, University of Washington, 3720 15th Avenue NE, Box 355061, Seattle, Washington 98195, United States
- Resuscitation Engineering Science Unit (RESCU), Harborview Research and Training Building, Seattle, Washington 98104, United States
| | - Abe Y Wu
- Department of Bioengineering and Molecular Engineering and Sciences Institute, University of Washington, 3720 15th Avenue NE, Box 355061, Seattle, Washington 98195, United States
| | - Michael Baldwin
- Seattle Children's Therapeutics, Seattle Children's Research Institute, 1920 Terry Ave, Seattle, Washington 98101, United States
| | - James Matthaei
- Seattle Children's Therapeutics, Seattle Children's Research Institute, 1920 Terry Ave, Seattle, Washington 98101, United States
| | - Kefan Song
- Department of Bioengineering and Molecular Engineering and Sciences Institute, University of Washington, 3720 15th Avenue NE, Box 355061, Seattle, Washington 98195, United States
| | - Meilyn Sylvestre
- Department of Bioengineering and Molecular Engineering and Sciences Institute, University of Washington, 3720 15th Avenue NE, Box 355061, Seattle, Washington 98195, United States
| | - Joshua Gustafson
- Seattle Children's Therapeutics, Seattle Children's Research Institute, 1920 Terry Ave, Seattle, Washington 98101, United States
| | - Nathan J White
- Resuscitation Engineering Science Unit (RESCU), Harborview Research and Training Building, Seattle, Washington 98104, United States
- Department of Emergency Medicine, University of Washington School of Medicine, Seattle, Washington 98105, United States
| | - Michael C Jensen
- Seattle Children's Therapeutics, Seattle Children's Research Institute, 1920 Terry Ave, Seattle, Washington 98101, United States
| | - Suzie H Pun
- Department of Bioengineering and Molecular Engineering and Sciences Institute, University of Washington, 3720 15th Avenue NE, Box 355061, Seattle, Washington 98195, United States
- Resuscitation Engineering Science Unit (RESCU), Harborview Research and Training Building, Seattle, Washington 98104, United States
| |
Collapse
|
4
|
Li S, Shi L, Zhao L, Guo Q, Li J, Liu ZL, Guo Z, Cao YJ. Split-design approach enhances the therapeutic efficacy of ligand-based CAR-T cells against multiple B-cell malignancies. Nat Commun 2024; 15:9751. [PMID: 39528513 PMCID: PMC11555413 DOI: 10.1038/s41467-024-54150-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Accepted: 11/02/2024] [Indexed: 11/16/2024] Open
Abstract
To address immune escape, multi-specific CAR-T-cell strategies use natural ligands that specifically bind multiple receptors on malignant cells. In this context, we propose a split CAR design comprising a universal receptor expressed on T cells and ligand-based switch molecules, which preserves the natural trimeric structure of ligands like APRIL and BAFF. Following optimization of the hinges and switch labeling sites, the split-design CAR-T cells ensure the native conformation of ligands, facilitating the optimal formation of immune synapses between target cancer cells and CAR-T cells. Our CAR-T-cell strategy demonstrates antitumor activities against various B-cell malignancy models in female mice, potentially preventing immune escape following conventional CAR-T-cell therapies in the case of antigen loss or switching. This ligand-based split CAR design introduces an idea for optimizing CAR recognition, enhancing efficacy and potentially improving safety in clinical translation, and may be broadly applicable to cellular therapies based on natural receptors or ligands.
Collapse
Affiliation(s)
- Shuhong Li
- State Key Laboratory of Chemical Oncogenomics, Shenzhen Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, Guangdong, China
| | - Licai Shi
- State Key Laboratory of Chemical Oncogenomics, Shenzhen Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, Guangdong, China
| | - Lijun Zhao
- State Key Laboratory of Chemical Oncogenomics, Shenzhen Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, Guangdong, China
| | - Qiaoru Guo
- State Key Laboratory of Chemical Oncogenomics, Shenzhen Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, Guangdong, China
| | - Jun Li
- Fundamenta Therapeutics Co., Ltd, Suzhou, Jiangsu, China
| | - Ze-Lin Liu
- Department of Hematology, Huazhong University of Science and Technology Union Shenzhen Hospital (Nanshan Hospital), Shenzhen, Guangdong, China
| | - Zhi Guo
- Department of Hematology, Huazhong University of Science and Technology Union Shenzhen Hospital (Nanshan Hospital), Shenzhen, Guangdong, China
| | - Yu J Cao
- State Key Laboratory of Chemical Oncogenomics, Shenzhen Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, Guangdong, China.
- Institute of Chemical Biology, Shenzhen Bay Laboratory, Shenzhen, China.
| |
Collapse
|
5
|
Zhang B, Wang M, Sun L, Liu J, Yin L, Xia M, Zhang L, Liu X, Cheng Y. Recent Advances in Targeted Cancer Therapy: Are PDCs the Next Generation of ADCs? J Med Chem 2024; 67:11469-11487. [PMID: 38980167 DOI: 10.1021/acs.jmedchem.4c00106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Antibody-drug conjugates (ADCs) comprise antibodies, cytotoxic payloads, and linkers, which can integrate the advantages of antibodies and small molecule drugs to achieve targeted cancer treatment. However, ADCs also have some shortcomings, such as non-negligible drug resistance, a low therapeutic index, and payload-related toxicity. Many studies have focused on changing the composition of ADCs, and some have even further extended the concept and types of targeted conjugated drugs by replacing the targeted antibodies in ADCs with peptides, revolutionarily introducing peptide-drug conjugates (PDCs). This Perspective summarizes the current research status of ADCs and PDCs and highlights the structural innovations of ADC components. In particular, PDCs are regarded as the next generation of potential targeted drugs after ADCs, and the current challenges of PDCs are analyzed. Our aim is to offer fresh insights for the efficient design and expedited development of innovative targeted conjugated drugs.
Collapse
Affiliation(s)
- Baochen Zhang
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, Hebei Anti-Tumor Molecular Target Technology Innovation Center, Hebei Research Center of the Basic Discipline of Cell Biology, College of Life Science, Hebei Normal University, Shijiazhuang 050024, P.R. China
| | - Mo Wang
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, Hebei Anti-Tumor Molecular Target Technology Innovation Center, Hebei Research Center of the Basic Discipline of Cell Biology, College of Life Science, Hebei Normal University, Shijiazhuang 050024, P.R. China
| | - Li Sun
- School of Chemical Technology, Shijiazhuang University, Shijiazhuang 050035, P.R. China
| | - Jiawei Liu
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, Hebei Anti-Tumor Molecular Target Technology Innovation Center, Hebei Research Center of the Basic Discipline of Cell Biology, College of Life Science, Hebei Normal University, Shijiazhuang 050024, P.R. China
| | - Libinghan Yin
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, Hebei Anti-Tumor Molecular Target Technology Innovation Center, Hebei Research Center of the Basic Discipline of Cell Biology, College of Life Science, Hebei Normal University, Shijiazhuang 050024, P.R. China
| | - Mingjing Xia
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, Hebei Anti-Tumor Molecular Target Technology Innovation Center, Hebei Research Center of the Basic Discipline of Cell Biology, College of Life Science, Hebei Normal University, Shijiazhuang 050024, P.R. China
| | - Ling Zhang
- School of Chemical Technology, Shijiazhuang University, Shijiazhuang 050035, P.R. China
| | - Xifu Liu
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, Hebei Anti-Tumor Molecular Target Technology Innovation Center, Hebei Research Center of the Basic Discipline of Cell Biology, College of Life Science, Hebei Normal University, Shijiazhuang 050024, P.R. China
| | - Yu Cheng
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, Hebei Anti-Tumor Molecular Target Technology Innovation Center, Hebei Research Center of the Basic Discipline of Cell Biology, College of Life Science, Hebei Normal University, Shijiazhuang 050024, P.R. China
| |
Collapse
|
6
|
Guo QR, Cao YJ. Applications of genetic code expansion technology in eukaryotes. Protein Cell 2024; 15:331-363. [PMID: 37847216 PMCID: PMC11074999 DOI: 10.1093/procel/pwad051] [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: 07/04/2023] [Accepted: 09/26/2023] [Indexed: 10/18/2023] Open
Abstract
Unnatural amino acids (UAAs) have gained significant attention in protein engineering and drug development owing to their ability to introduce new chemical functionalities to proteins. In eukaryotes, genetic code expansion (GCE) enables the incorporation of UAAs and facilitates posttranscriptional modification (PTM), which is not feasible in prokaryotic systems. GCE is also a powerful tool for cell or animal imaging, the monitoring of protein interactions in target cells, drug development, and switch regulation. Therefore, there is keen interest in utilizing GCE in eukaryotic systems. This review provides an overview of the application of GCE in eukaryotic systems and discusses current challenges that need to be addressed.
Collapse
Affiliation(s)
- Qiao-ru Guo
- State Key Laboratory of Chemical Oncogenomic, Guangdong Provincial Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen 518055, China
| | - Yu J Cao
- State Key Laboratory of Chemical Oncogenomic, Guangdong Provincial Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen 518055, China
- Institute of Chemical Biology, Shenzhen Bay Laboratory, Shenzhen 518132, China
| |
Collapse
|
7
|
Yu T, Lu Y, Fang J, Jiang X, Lu Y, Zheng J, Shang X, Shen H, Fu P. Chimeric antigen receptor-based immunotherapy in breast cancer: Recent progress in China. Cancer 2024; 130:1378-1391. [PMID: 37950749 DOI: 10.1002/cncr.35096] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 09/11/2023] [Accepted: 10/11/2023] [Indexed: 11/13/2023]
Abstract
Breast cancer (BC) is the fourth most prevalent cancer in China. Despite conventional treatment strategies, BC patients often have poor therapeutic outcomes, leading to significant global cancer mortality rates. Chimeric antigen receptor (CAR)-based immunotherapy is a promising and innovative approach for cancer treatment that redirects immune cells to attack tumor cells expressing selected tumor antigens (TAs). T cells, natural killer (NK) cells, and macrophages, key components of the immune system, are used in CAR-based immunotherapies. Although remarkable progress has been made with CAR-T cells in hematologic malignancies, the application of CAR-based immunotherapy to BC has lagged. This is partly due to obstacles such as tumor heterogeneity, which is further associated with the TA and BC subtypes, and the immunosuppressive tumor microenvironment (TME). Several combinatorial approaches, including the use of immune checkpoint inhibitors, oncolytic viruses, and antitumor drugs, have been proposed to overcome these obstacles in BC treatment. Furthermore, several CAR-based immunotherapies for BC have been translated into clinical trials. This review provides an overview of the recent progress in CAR-based immunotherapy for BC treatment, including targeting of TAs, consideration of BC subtypes, assessment of the TME, and exploration of combinatorial therapies. The authors focused on preclinical studies and clinical trials of CAR-T cells, CAR-NK cells, and CAR-macrophages especially conducted in China, followed by an internal comparison and discussion of current limits. In conclusion, this review elucidates China's contribution to CAR-based immunotherapies for BC and provides inspiration for further research. PLAIN LANGUAGE SUMMARY: Despite conventional treatment strategies, breast cancer (BC) patients in China often have poor therapeutic outcomes. Chimeric antigen receptor (CAR)-based immunotherapy, a promising approach, can redirect immune cells to kill tumor cells expressing selected tumor antigens (TAs). However, obstacles such as TA selection, BC subtypes, and immunosuppressive tumor microenvironment still exist. Therefore, various combinatorial approaches have been proposed. This article elucidates several Chinese CAR-based preclinical and clinical studies in BC treatment with comparisons of foreign research, and CAR-immune cells are analyzed, providing inspiration for further research.
Collapse
Affiliation(s)
- Tianze Yu
- Department of Breast Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- School of Medicine, Zhejiang University, Hangzhou, China
| | - Yuexin Lu
- Department of Breast Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- School of Medicine, Zhejiang University, Hangzhou, China
| | - Jianwen Fang
- Department of Breast Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- School of Medicine, Zhejiang University, Hangzhou, China
| | - Xiaocong Jiang
- Department of Breast Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- School of Medicine, Zhejiang University, Hangzhou, China
| | - Yue Lu
- Department of Breast and Thyroid Surgery, First Affiliated Hospital of Huzhou University, Huzhou, China
| | - Jingyan Zheng
- Department of Breast and Thyroid Surgery, Lishui People's Hospital, The Sixth Affiliated Hospital of Wenzhou Medical University, Lishui, China
| | - Xi Shang
- Department of Breast and Thyroid Surgery, Taizhou Hospital, Zhejiang University, Taizhou, China
| | - Haixing Shen
- Department of Breast and Thyroid Surgery, Cixi People's Hospital, Cixi, China
| | - Peifen Fu
- Department of Breast Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
8
|
Zhang AQ, Hostetler A, Chen LE, Mukkamala V, Abraham W, Padilla LT, Wolff AN, Maiorino L, Backlund CM, Aung A, Melo M, Li N, Wu S, Irvine DJ. Universal redirection of CAR T cells against solid tumours via membrane-inserted ligands for the CAR. Nat Biomed Eng 2023; 7:1113-1128. [PMID: 37291434 PMCID: PMC10504084 DOI: 10.1038/s41551-023-01048-8] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Accepted: 05/01/2023] [Indexed: 06/10/2023]
Abstract
The effectiveness of chimaeric antigen receptor (CAR) T cell therapies for solid tumours is hindered by difficulties in the selection of an effective target antigen, owing to the heterogeneous expression of tumour antigens and to target antigen expression in healthy tissues. Here we show that T cells with a CAR specific for fluorescein isothiocyanate (FITC) can be directed against solid tumours via the intratumoural administration of a FITC-conjugated lipid-poly(ethylene)-glycol amphiphile that inserts itself into cell membranes. In syngeneic and human tumour xenografts in mice, 'amphiphile tagging' of tumour cells drove tumour regression via the proliferation and accumulation of FITC-specific CAR T cells in the tumours. In syngeneic tumours, the therapy induced the infiltration of host T cells, elicited endogenous tumour-specific T cell priming and led to activity against distal untreated tumours and to protection against tumour rechallenge. Membrane-inserting ligands for specific CARs may facilitate the development of adoptive cell therapies that work independently of antigen expression and of tissue of origin.
Collapse
Affiliation(s)
- Angela Q Zhang
- Koch Institute for Integrative Cancer Research, Cambridge, MA, USA
- Department of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Biophysics, Harvard University, Cambridge, MA, USA
| | - Alexander Hostetler
- Koch Institute for Integrative Cancer Research, Cambridge, MA, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Laura E Chen
- Koch Institute for Integrative Cancer Research, Cambridge, MA, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Vainavi Mukkamala
- Koch Institute for Integrative Cancer Research, Cambridge, MA, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Wuhbet Abraham
- Koch Institute for Integrative Cancer Research, Cambridge, MA, USA
| | - Lucia T Padilla
- Koch Institute for Integrative Cancer Research, Cambridge, MA, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Alexandra N Wolff
- Koch Institute for Integrative Cancer Research, Cambridge, MA, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Laura Maiorino
- Koch Institute for Integrative Cancer Research, Cambridge, MA, USA
| | | | - Aereas Aung
- Koch Institute for Integrative Cancer Research, Cambridge, MA, USA
| | - Mariane Melo
- Koch Institute for Integrative Cancer Research, Cambridge, MA, USA
| | - Na Li
- Koch Institute for Integrative Cancer Research, Cambridge, MA, USA
| | - Shengwei Wu
- Koch Institute for Integrative Cancer Research, Cambridge, MA, USA
| | - Darrell J Irvine
- Koch Institute for Integrative Cancer Research, Cambridge, MA, USA.
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Ragon Institute of MIT, MGH, and Harvard, Cambridge, MA, USA.
- Howard Hughes Medical Institute, Chevy Chase, MD, USA.
| |
Collapse
|
9
|
Katiyar V, Chesney J, Kloecker G. Cellular Therapy for Lung Cancer: Focusing on Chimeric Antigen Receptor T (CAR T) Cells and Tumor-Infiltrating Lymphocyte (TIL) Therapy. Cancers (Basel) 2023; 15:3733. [PMID: 37509394 PMCID: PMC10377757 DOI: 10.3390/cancers15143733] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 07/03/2023] [Accepted: 07/13/2023] [Indexed: 07/30/2023] Open
Abstract
Lung cancer is a leading cause of morbidity and mortality in the United States and worldwide. The introduction of immune checkpoint inhibitors has led to a marked improvement in the outcomes of lung cancer patients. Despite these advances, there is a huge unmet need for therapeutic options in patients who are not candidates for targeted or immunotherapy or those who progress after first-line treatment. With its high mutational burden, lung cancer appears to be an attractive target for novel personalized treatment approaches. In this review, we provide an overview of two adoptive cell therapy approaches-chimeric antigen receptors (CAR) T-cell therapy and Tumor-infiltrating lymphocytes (TILs) in lung cancer with an emphasis on current challenges and future perspectives. While both these therapies are still in the early phases of development in lung cancer and need more refinement, they harbor the potential to be effective treatment options for this group of patients with otherwise poor prognoses.
Collapse
Affiliation(s)
- Vatsala Katiyar
- James Graham Brown Cancer Center, University of Louisville, Louisville, KY 40202, USA
| | - Jason Chesney
- James Graham Brown Cancer Center, University of Louisville, Louisville, KY 40202, USA
| | - Goetz Kloecker
- James Graham Brown Cancer Center, University of Louisville, Louisville, KY 40202, USA
| |
Collapse
|
10
|
He Q, Hu H, Yang F, Song D, Zhang X, Dai X. Advances in chimeric antigen receptor T cells therapy in the treatment of breast cancer. Biomed Pharmacother 2023; 162:114609. [PMID: 37001182 DOI: 10.1016/j.biopha.2023.114609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 03/21/2023] [Accepted: 03/24/2023] [Indexed: 03/31/2023] Open
Abstract
Breast cancer (BC) is the most frequently occurring cancer type seriously threatening the lives of women worldwide. Clinically, the high frequency of diverse resistance to current therapeutic strategies advocates a demand to develop novel and effective approaches for the efficient treatment of BC. The chimeric antigen receptor T (CAR-T) cells therapy, one of the immunotherapies, has displayed powerful capacity to specifically kill and eliminate tumors. Due to the success of CAR-T therapy achieved in treating hematological malignancy, the effect of CAR-T cells therapy has been tested in various human diseases including breast cancer. This review summarized and discussed the landscape of the CAR-T therapy for breast cancer, including the advances, challenge and countermeasure of CAR-T therapy in research and clinical application. The roles of potential antigen targets, tumor microenvironment, immune escape in regulating CAR-T therapy, the combination of CAR-T therapy with other therapeutic strategies to further enhance therapeutic efficacy of CAR-T treatment were also highlighted. Therefore, our review provided a comprehensive understanding of CAR-T cell therapy in breast cancer which will awake huge interests for future in-depth investigation of CAR-T based therapy in cancer treatment.
Collapse
|
11
|
Mercogliano MF, Bruni S, Mauro FL, Schillaci R. Emerging Targeted Therapies for HER2-Positive Breast Cancer. Cancers (Basel) 2023; 15:cancers15071987. [PMID: 37046648 PMCID: PMC10093019 DOI: 10.3390/cancers15071987] [Citation(s) in RCA: 60] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 03/23/2023] [Accepted: 03/24/2023] [Indexed: 03/29/2023] Open
Abstract
Breast cancer is the most common cancer in women and the leading cause of death. HER2 overexpression is found in approximately 20% of breast cancers and is associated with a poor prognosis and a shorter overall survival. Tratuzumab, a monoclonal antibody directed against the HER2 receptor, is the standard of care treatment. However, a third of the patients do not respond to therapy. Given the high rate of resistance, other HER2-targeted strategies have been developed, including monoclonal antibodies such as pertuzumab and margetuximab, trastuzumab-based antibody drug conjugates such as trastuzumab-emtansine (T-DM1) and trastuzumab-deruxtecan (T-DXd), and tyrosine kinase inhibitors like lapatinib and tucatinib, among others. Moreover, T-DXd has proven to be of use in the HER2-low subtype, which suggests that other HER2-targeted therapies could be successful in this recently defined new breast cancer subclassification. When patients progress to multiple strategies, there are several HER2-targeted therapies available; however, treatment options are limited, and the potential combination with other drugs, immune checkpoint inhibitors, CAR-T cells, CAR-NK, CAR-M, and vaccines is an interesting and appealing field that is still in development. In this review, we will discuss the highlights and pitfalls of the different HER2-targeted therapies and potential combinations to overcome metastatic disease and resistance to therapy.
Collapse
|
12
|
Celichowski P, Turi M, Charvátová S, Radhakrishnan D, Feizi N, Chyra Z, Šimíček M, Jelínek T, Bago JR, Hájek R, Hrdinka M. Tuning CARs: recent advances in modulating chimeric antigen receptor (CAR) T cell activity for improved safety, efficacy, and flexibility. J Transl Med 2023; 21:197. [PMID: 36922828 PMCID: PMC10015723 DOI: 10.1186/s12967-023-04041-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Accepted: 03/07/2023] [Indexed: 03/17/2023] Open
Abstract
Cancer immunotherapies utilizing genetically engineered T cells have emerged as powerful personalized therapeutic agents showing dramatic preclinical and clinical results, particularly in hematological malignancies. Ectopically expressed chimeric antigen receptors (CARs) reprogram immune cells to target and eliminate cancer. However, CAR T cell therapy's success depends on the balance between effective anti-tumor activity and minimizing harmful side effects. To improve CAR T cell therapy outcomes and mitigate associated toxicities, scientists from different fields are cooperating in developing next-generation products using the latest molecular cell biology and synthetic biology tools and technologies. The immunotherapy field is rapidly evolving, with new approaches and strategies being reported at a fast pace. This comprehensive literature review aims to provide an up-to-date overview of the latest developments in controlling CAR T cell activity for improved safety, efficacy, and flexibility.
Collapse
Affiliation(s)
- Piotr Celichowski
- Department of Haematooncology, Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic
- Department of Haematooncology, University Hospital Ostrava, Ostrava, Czech Republic
| | - Marcello Turi
- Department of Haematooncology, Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic
- Department of Haematooncology, University Hospital Ostrava, Ostrava, Czech Republic
- Faculty of Science, University of Ostrava, Ostrava, Czech Republic
| | - Sandra Charvátová
- Department of Haematooncology, Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic
- Department of Haematooncology, University Hospital Ostrava, Ostrava, Czech Republic
- Faculty of Science, University of Ostrava, Ostrava, Czech Republic
| | - Dhwani Radhakrishnan
- Department of Haematooncology, Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic
- Department of Haematooncology, University Hospital Ostrava, Ostrava, Czech Republic
- Faculty of Science, University of Ostrava, Ostrava, Czech Republic
| | - Neda Feizi
- Department of Internal Clinical Sciences, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Rome, Italy
| | - Zuzana Chyra
- Department of Haematooncology, Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic
- Department of Haematooncology, University Hospital Ostrava, Ostrava, Czech Republic
| | - Michal Šimíček
- Department of Haematooncology, Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic
- Department of Haematooncology, University Hospital Ostrava, Ostrava, Czech Republic
| | - Tomáš Jelínek
- Department of Haematooncology, Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic
- Department of Haematooncology, University Hospital Ostrava, Ostrava, Czech Republic
| | - Juli Rodriguez Bago
- Department of Haematooncology, Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic
- Department of Haematooncology, University Hospital Ostrava, Ostrava, Czech Republic
| | - Roman Hájek
- Department of Haematooncology, Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic
- Department of Haematooncology, University Hospital Ostrava, Ostrava, Czech Republic
| | - Matouš Hrdinka
- Department of Haematooncology, Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic.
- Department of Haematooncology, University Hospital Ostrava, Ostrava, Czech Republic.
| |
Collapse
|
13
|
CAR-NK as a Rapidly Developed and Efficient Immunotherapeutic Strategy against Cancer. Cancers (Basel) 2022; 15:cancers15010117. [PMID: 36612114 PMCID: PMC9817948 DOI: 10.3390/cancers15010117] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 12/14/2022] [Accepted: 12/20/2022] [Indexed: 12/28/2022] Open
Abstract
Chimeric antigen receptor (CAR)-modified T cell therapy has been rapidly developing in recent years, ultimately revolutionizing immunotherapeutic strategies and providing significant anti-tumor potency, mainly in treating hematological neoplasms. However, graft-versus-host disease (GVHD) and other adverse effects, such as cytokine release syndromes (CRS) and neurotoxicity associated with CAR-T cell infusion, have raised some concerns about the broad application of this therapy. Natural killer (NK) cells have been identified as promising alternative platforms for CAR-based therapies because of their unique features, such as a lack of human leukocyte antigen (HLA)-matching restriction, superior safety, and better anti-tumor activity when compared with CAR-T cells. The lack of CRS, neurotoxicity, or GVHD, in the case of CAR-NK therapy, in addition to the possibility of using allogeneic NK cells as a CAR platform for "off-the-shelf" therapy, opens new windows for strategic opportunities. This review underlines recent design achievements in CAR constructs and summarizes preclinical studies' results regarding CAR-NK therapies' safety and anti-tumor potency. Additionally, new approaches in CAR-NK technology are briefly described, and currently registered clinical trials are listed.
Collapse
|
14
|
A novel CD19/CD22/CD3 trispecific antibody enhances therapeutic efficacy and overcomes immune escape against B-ALL. Blood 2022; 140:1790-1802. [PMID: 35981465 DOI: 10.1182/blood.2022016243] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 08/03/2022] [Indexed: 11/20/2022] Open
Abstract
The bispecific T-cell engager (BiTE) blinatumomab against CD19 and CD3 has emerged as the most successful bispecific antibody (bsAb) to date; however, a significant proportion of patients do not respond to the treatments or eventually experience relapse after an initial response, and the recurrence rate increases significantly due to escape or downregulation of the CD19 antigen. To enhance antitumor efficacy and overcome potential immune escape, we developed a novel approach to design a CD19/CD22/CD3 trispecific antibody (tsAb) by site-specifically fusing anti-CD19 scFv (FMC63) and anti-CD22 nanobody (Nb25) to the defined sites of the CD3 antigen-binding fragment (Fab, SP34). This strategy allows for the optimal formation of immune synapses mediated by CD19/CD22/CD3 between target cells and T cells. Optimized tsAb can be superior for inducing T-cell-specific cytotoxicity and cytokine production against CD19+ and/or CD22+ tumor cells compared to other tsAb formats, and demonstrated significantly enhanced antitumor efficacy and the ability to overcome immune escape compared with the corresponding bsAbs alone or in combination, as well as with blinatumomab. In addition, tsAb treatment can lead to the long-term elimination of primary B-ALL patient samples in the PDX model and significantly prolong survival. This novel approach provides unique insight into the structural optimization of T-cell-redirected multispecific antibodies using site-specific recombination, and may be broadly applicable to heterogeneous and resistant tumor populations as well as solid tumors.
Collapse
|
15
|
Qu C, Zhang H, Cao H, Tang L, Mo H, Liu F, Zhang L, Yi Z, Long L, Yan L, Wang Z, Zhang N, Luo P, Zhang J, Liu Z, Ye W, Liu Z, Cheng Q. Tumor buster - where will the CAR-T cell therapy 'missile' go? Mol Cancer 2022; 21:201. [PMID: 36261831 PMCID: PMC9580202 DOI: 10.1186/s12943-022-01669-8] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Accepted: 09/26/2022] [Indexed: 11/10/2022] Open
Abstract
Chimeric antigen receptor (CAR) T cell (CAR-T cell) therapy based on gene editing technology represents a significant breakthrough in personalized immunotherapy for human cancer. This strategy uses genetic modification to enable T cells to target tumor-specific antigens, attack specific cancer cells, and bypass tumor cell apoptosis avoidance mechanisms to some extent. This method has been extensively used to treat hematologic diseases, but the therapeutic effect in solid tumors is not ideal. Tumor antigen escape, treatment-related toxicity, and the immunosuppressive tumor microenvironment (TME) limit their use of it. Target selection is the most critical aspect in determining the prognosis of patients receiving this treatment. This review provides a comprehensive summary of all therapeutic targets used in the clinic or shown promising potential. We summarize CAR-T cell therapies' clinical trials, applications, research frontiers, and limitations in treating different cancers. We also explore coping strategies when encountering sub-optimal tumor-associated antigens (TAA) or TAA loss. Moreover, the importance of CAR-T cell therapy in cancer immunotherapy is emphasized.
Collapse
Affiliation(s)
- Chunrun Qu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
- XiangYa School of Medicine, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Hao Zhang
- 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, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Hui Cao
- Department of Psychiatry, The Second People's Hospital of Hunan Province, The Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
- The School of Clinical Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Lanhua Tang
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Haoyang Mo
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
- XiangYa School of Medicine, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Fangkun Liu
- 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
| | - Liyang Zhang
- 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
| | - Zhenjie Yi
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
- XiangYa School of Medicine, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Lifu Long
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
- XiangYa School of Medicine, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Luzhe Yan
- XiangYa School of Medicine, Central South University, Changsha, Hunan, China
| | - Zeyu Wang
- 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
| | - Nan Zhang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
- One-third Lab, College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang, China
| | - Peng Luo
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Jian Zhang
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Zaoqu Liu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou, Zhengzhou, Henan, China
| | - Weijie Ye
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zhixiong Liu
- 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.
| | - Quan Cheng
- 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.
| |
Collapse
|
16
|
Kandra P, Nandigama R, Eul B, Huber M, Kobold S, Seeger W, Grimminger F, Savai R. Utility and Drawbacks of Chimeric Antigen Receptor T Cell (CAR-T) Therapy in Lung Cancer. Front Immunol 2022; 13:903562. [PMID: 35720364 PMCID: PMC9201083 DOI: 10.3389/fimmu.2022.903562] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 05/06/2022] [Indexed: 11/23/2022] Open
Abstract
The present treatments for lung cancer include surgical resection, radiation, chemotherapy, targeted therapy, and immunotherapy. Despite advances in therapies, the prognosis of lung cancer has not been substantially improved in recent years. Chimeric antigen receptor (CAR)-T cell immunotherapy has attracted growing interest in the treatment of various malignancies. Despite CAR-T cell therapy emerging as a novel potential therapeutic option with promising results in refractory and relapsed leukemia, many challenges limit its therapeutic efficacy in solid tumors including lung cancer. In this landscape, studies have identified several obstacles to the effective use of CAR-T cell therapy including antigen heterogeneity, the immunosuppressive tumor microenvironment, and tumor penetration by CAR-T cells. Here, we review CAR-T cell design; present the results of CAR-T cell therapies in preclinical and clinical studies in lung cancer; describe existing challenges and toxicities; and discuss strategies to improve therapeutic efficacy of CAR-T cells.
Collapse
Affiliation(s)
- Prameela Kandra
- Department of Biotechnology, Gandhi Institute of Technology and Management (GITAM) Institute of Technology, Gandhi Institute of Technology and Management (GITAM) Deemed to be University, Visakhapatnam, India
| | - Rajender Nandigama
- Max Planck Institute for Heart and Lung Research, Member of the German Center for Lung Research Deutsches Zentrum für Lungenforschung (DZL), Member of the Cardio-Pulmonary Institute (CPI), Bad Nauheim, Germany
| | - Bastian Eul
- Department of Internal Medicine, Member of the Deutsches Zentrum für Lungenforschung (DZL), Member of Cardio-Pulmonary Institute (CPI), Justus Liebig University, Giessen, Germany
| | - Magdalena Huber
- Institute for Medical Microbiology and Hygiene, Philipps-University Marburg, Marburg, Germany
| | - Sebastian Kobold
- Division of Clinical Pharmacology, Department of Medicine IV, Member of the Deutsches Zentrum für Lungenforschung (DZL), University Hospital Munich, Munich, Germany.,German Cancer Consortium Deutsches Konsortium für Translationale Krebsforschung (DKTK), Partner site Munich, Munich, Germany
| | - Werner Seeger
- Max Planck Institute for Heart and Lung Research, Member of the German Center for Lung Research Deutsches Zentrum für Lungenforschung (DZL), Member of the Cardio-Pulmonary Institute (CPI), Bad Nauheim, Germany.,Department of Internal Medicine, Member of the Deutsches Zentrum für Lungenforschung (DZL), Member of Cardio-Pulmonary Institute (CPI), Justus Liebig University, Giessen, Germany.,Institute for Lung Health (ILH), Justus Liebig University, Giessen, Germany
| | - Friedrich Grimminger
- Department of Internal Medicine, Member of the Deutsches Zentrum für Lungenforschung (DZL), Member of Cardio-Pulmonary Institute (CPI), Justus Liebig University, Giessen, Germany.,Institute for Lung Health (ILH), Justus Liebig University, Giessen, Germany
| | - Rajkumar Savai
- Max Planck Institute for Heart and Lung Research, Member of the German Center for Lung Research Deutsches Zentrum für Lungenforschung (DZL), Member of the Cardio-Pulmonary Institute (CPI), Bad Nauheim, Germany.,Department of Internal Medicine, Member of the Deutsches Zentrum für Lungenforschung (DZL), Member of Cardio-Pulmonary Institute (CPI), Justus Liebig University, Giessen, Germany.,Institute for Lung Health (ILH), Justus Liebig University, Giessen, Germany
| |
Collapse
|
17
|
Zhang Y, Li Y, Cao W, Wang F, Xie X, Li Y, Wang X, Guo R, Jiang Z, Guo R. Single-Cell Analysis of Target Antigens of CAR-T Reveals a Potential Landscape of "On-Target, Off-Tumor Toxicity". Front Immunol 2022; 12:799206. [PMID: 34975912 PMCID: PMC8716389 DOI: 10.3389/fimmu.2021.799206] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 11/19/2021] [Indexed: 01/09/2023] Open
Abstract
Cellular immunotherapy represented by CD19-directed chimeric antigen receptor T (CAR-T) cells has achieved great success in recent years. An increasing number of CAR-T therapies are being developed for cancer treatment, but the frequent and varied adverse events, such as “on-target, off-tumor toxicity”, limit CAR-T application. Here, we identify the target antigen expression patterns of CAR therapies in 18 tissues and organs (peripheral blood mononuclear cells, bone marrow, lymph nodes, spleen, heart, ascending aortic tissue, trachea, lung, skin, kidney, bladder, esophagus, stomach, small intestine, rectum, liver, common bile duct, and pancreas) from healthy human samples. The atlas determines target antigens expressed on some normal cell types, which facilitates elucidating the cause of “on-target, off-tumor toxicity” in special tissues and organs by targeting some antigens, but not others. Moreover, we describe the target antigen expression patterns of B-lineage-derived malignant cells, acute myeloid leukemia (AML), and solid tumors. Overall, the present study indicates the pathogenesis of “on-target, off-tumor toxicity” during CAR therapies and provides guidance on taking preventive measures during CAR treatment.
Collapse
Affiliation(s)
- Yinyin Zhang
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yingmei Li
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Weijie Cao
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Fang Wang
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xinsheng Xie
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yadan Li
- The Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China
| | - Xiaoyi Wang
- Department of Pediatric Hematology and Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Rong Guo
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhongxing Jiang
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Rongqun Guo
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
18
|
Budi HS, Ahmad FN, Achmad H, Ansari MJ, Mikhailova MV, Suksatan W, Chupradit S, Shomali N, Marofi F. Human epidermal growth factor receptor 2 (HER2)-specific chimeric antigen receptor (CAR) for tumor immunotherapy; recent progress. Stem Cell Res Ther 2022; 13:40. [PMID: 35093187 PMCID: PMC8800342 DOI: 10.1186/s13287-022-02719-0] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2021] [Accepted: 01/11/2022] [Indexed: 12/17/2022] Open
Abstract
Due to the overexpression or amplification of human epidermal growth factor receptor 2 (HER2) with poor prognosis in a myriad of human tumors, recent studies have focused on HER2-targeted therapies. Deregulation in HER2 signaling pathways is accompanied by sustained tumor cells growth concomitant with their migration and also tumor angiogenesis and metastasis by stimulation of proliferation of a network of blood vessels. A large number of studies have provided clear evidence that the emerging HER2-directed treatments could be the outcome of patients suffering from HER2 positive breast and also gastric/gastroesophageal cancers. Thanks to its great anti-tumor competence, immunotherapy using HER2-specific chimeric antigen receptor (CAR) expressing immune cell has recently attracted increasing attention. Human T cells and also natural killer (NK) cells can largely be found in the tumor microenvironment, mainly contributing to the tumor immune surveillance. Such properties make them perfect candidate for genetically modification to express constructed CARs. Herein, we will describe the potential targets of the HER2 signaling in tumor cells to clarify HER2-mediated tumorigenesis and also discuss recent findings respecting the HER2-specific CAR-expressing immune cells (CAR T and CAR NK cell) for the treatment of HER2-expressing tumors.
Collapse
Affiliation(s)
- Hendrik Setia Budi
- Department of Oral Biology, Faculty of Dental Medicine, Universitas Airlangga, Surabaya, 60132 Indonesia
| | | | - Harun Achmad
- Department of Pediatric Dentistry, Faculty of Dentistry, Hasanuddin University, Makassar, Indonesia
| | - Mohammad Javed Ansari
- Department of Pharmaceutics, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | | | - Wanich Suksatan
- Faculty of Nursing, HRH Princess Chulabhorn College of Medical Science, Chulabhorn Royal Academy, Bangkok, 10210 Thailand
| | - Supat Chupradit
- Department of Occupational Therapy, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, 50200 Thailand
| | - Navid Shomali
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Faroogh Marofi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
19
|
Simmons C, Rayson D, Joy AA, Henning JW, Lemieux J, McArthur H, Card PB, Dent R, Brezden-Masley C. Current and future landscape of targeted therapy in HER2-positive advanced breast cancer: redrawing the lines. Ther Adv Med Oncol 2022; 14:17588359211066677. [PMID: 35035535 PMCID: PMC8753087 DOI: 10.1177/17588359211066677] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Accepted: 11/26/2021] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Evidence to date supports continued human epidermal growth factor receptor 2 (HER2) suppression beyond progression on HER2-directed therapy for advanced HER2-positive breast cancer. Data from several phase II and III trials evaluating HER2-directed therapy following second-line T-DM1 have recently become available. METHODS We performed a systematic search of the published and presented literature to identify phase II and phase III trials assessing novel HER2-targeted agents as third-line therapy or beyond for HER2-positive advanced breast cancer using search terms 'breast cancer' AND 'HER2' AND 'advanced' AND ('phase II' OR 'phase III'). RESULTS Eight clinical trials reporting efficacy outcomes on third-line or greater HER2-directed therapy for HER2-positive advanced breast cancer were identified. In phase III trials, margetuximab and neratinib combinations demonstrated significant 1.3-month (hazard ratio, HR = 0.71, p < 0.001) and 0.1-month (HR = 0.76, p = 0.006) net improvements in median progression-free survival (PFS), respectively, with no significant improvements in overall survival (OS). Tucatinib added to trastuzumab and capecitabine demonstrated a significant 2.7-month improvement in median PFS (HR = 0.57, p < 0.00001) and a 5.5-month improvement in median OS (HR = 0.73, p = 0.004) in a randomized phase II trial, including significant clinical benefit for patients with brain metastases. Finally, trastuzumab-deruxtecan, zenocutuzumab, and poziotinib demonstrated benefit in phase II trials with the most robust overall response rate (62.0%) and median duration of response (18.2 months) observed for trastuzumab-deruxtecan among heavily pretreated patients. CONCLUSION Tucatinib plus trastuzumab and capecitabine significantly prolongs OS, and promising preliminary response outcomes for trastuzumab-deruxtecan suggest that sequencing of these regimens following second-line therapy is reasonable.
Collapse
Affiliation(s)
- Christine Simmons
- Medical Oncology, British Columbia Cancer
Agency – Vancouver Centre, University of British Columbia, 600 West 10th
Avenue, Vancouver, BC V5Z 4E6, Canada
| | - Daniel Rayson
- Queen Elizabeth II Health Sciences Centre,
Dalhousie University, Halifax, NS, Canada
| | - Anil Abraham Joy
- Cross Cancer Institute, University of Alberta,
Edmonton, AB, Canada
| | | | - Julie Lemieux
- Centre hospitalier universitaire de Québec,
Université Laval, Quebec, QC, Canada
| | | | - Paul B. Card
- Kaleidoscope Strategic, Inc., Toronto, ON,
Canada
| | - Rebecca Dent
- National Cancer Centre Singapore, Duke-NUS
Medical School, Singapore
| | | |
Collapse
|