1
|
De Lucia A, Mazzotti L, Gaimari A, Zurlo M, Maltoni R, Cerchione C, Bravaccini S, Delmonte A, Crinò L, Borges de Souza P, Pasini L, Nicolini F, Bianchi F, Juan M, Calderon H, Magnoni C, Gazzola L, Ulivi P, Mazza M. Non-small cell lung cancer and the tumor microenvironment: making headway from targeted therapies to advanced immunotherapy. Front Immunol 2025; 16:1515748. [PMID: 39995659 PMCID: PMC11847692 DOI: 10.3389/fimmu.2025.1515748] [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: 10/23/2024] [Accepted: 01/20/2025] [Indexed: 02/26/2025] Open
Abstract
Over the past decades, significant progress has been made in the understanding of non-small cell lung cancer (NSCLC) biology and tumor progression mechanisms, resulting in the development of novel strategies for early detection and wide-ranging care approaches. Since their introduction, over 20 years ago, targeted therapies with tyrosine kinase inhibitors (TKIs) have revolutionized the treatment landscape for NSCLC. Nowadays, targeted therapies remain the gold standard for many patients, but still they suffer from many adverse effects, including unexpected toxicity and intrinsic acquired resistance mutations, which lead to relapse. The adoption of immune checkpoint inhibitors (ICIs) in 2015, has offered exceptional survival benefits for patients without targetable alterations. Despite this notable progress, challenges remain, as not all patients respond favorably to ICIs, and resistance to therapy can develop over time. A crucial factor influencing clinical response to immunotherapy is the tumor microenvironment (TME). The TME is pivotal in orchestrating the interactions between neoplastic cells and the immune system, influencing tumor growth and treatment outcomes. In this review, we discuss how the understanding of this intricate relationship is crucial for the success of immunotherapy and survey the current state of immunotherapy intervention, with a focus on forthcoming and promising chimeric antigen receptor (CAR) T cell therapies in NSCLC. The TME sets major obstacles for CAR-T therapies, creating conditions that suppress the immune response, inducing T cell exhaustion. To enhance treatment efficacy, specific efforts associated with CAR-T cell therapy in NSCLC, should definitely focus TME-related immunosuppression and antigen escape mechanisms, by combining CAR-T cells with immune checkpoint blockades.
Collapse
Affiliation(s)
- Anna De Lucia
- Advanced Cellular Therapies and Rare Tumors Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Lucia Mazzotti
- Advanced Cellular Therapies and Rare Tumors Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Anna Gaimari
- Advanced Cellular Therapies and Rare Tumors Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Matteo Zurlo
- Advanced Cellular Therapies and Rare Tumors Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Roberta Maltoni
- Healthcare Administration, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Claudio Cerchione
- Hematology Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Sara Bravaccini
- Department of Medicine and Surgery, “Kore” University of Enna, Enna, Italy
| | - Angelo Delmonte
- Medical Oncology Department, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Lucio Crinò
- Medical Oncology Department, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Patricia Borges de Souza
- Advanced Cellular Therapies and Rare Tumors Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Luigi Pasini
- Advanced Cellular Therapies and Rare Tumors Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Fabio Nicolini
- Advanced Cellular Therapies and Rare Tumors Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Fabrizio Bianchi
- Unit of Cancer Biomarker, Fondazione IRCCS Casa Sollievo Della Sofferenza, San Giovanni Rotondo, FG, Italy
| | - Manel Juan
- Department of Immunology, Institut D’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Hugo Calderon
- Department of Immunology, Institut D’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Chiara Magnoni
- Advanced Cellular Therapies and Rare Tumors Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Luca Gazzola
- Advanced Cellular Therapies and Rare Tumors Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Paola Ulivi
- Translational Oncology Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Massimiliano Mazza
- Advanced Cellular Therapies and Rare Tumors Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| |
Collapse
|
2
|
Arjmand B, Alavi-Moghadam S, Khorsand G, Sarvari M, Arjmand R, Rezaei-Tavirani M, Rajaeinejad M, Mosaed R. Cell-Based Vaccines: Frontiers in Medical Technology for Cancer Treatment. REGENERATIVE ENGINEERING AND TRANSLATIONAL MEDICINE 2024; 10:480-499. [DOI: 10.1007/s40883-024-00338-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Revised: 01/13/2024] [Accepted: 02/17/2024] [Indexed: 01/03/2025]
|
3
|
Pessoa J, Nóbrega-Pereira S, de Jesus BB. Senescent cell-derived vaccines: a new concept towards an immune response against cancer and aging? Aging (Albany NY) 2024; 16:10657-10665. [PMID: 38942604 PMCID: PMC11236300 DOI: 10.18632/aging.205975] [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: 11/20/2023] [Accepted: 03/13/2024] [Indexed: 06/30/2024]
Abstract
Two recent seminal works have untangled the intricate role of tumor-associated senescent cells in cancer progression, or regression, by guiding our immune system against cancer cells. The characterization of these unique, yet diverse cell populations, should be considered, particularly when contemplating the use of senolytics, which are drugs that selectively eliminate senescent cells, in a cancer framework. Here, we will describe the current knowledge in this field. In particular, we will discuss how the presence of senescent cells in tumors could be used as a therapeutic target in immunogenic cancers and how we may hypothetically design an adaptive anti-aging vaccine.
Collapse
Affiliation(s)
- João Pessoa
- Department of Medical Sciences and Institute of Biomedicine - iBiMED, University of Aveiro, Aveiro 3810-193, Portugal
| | - Sandrina Nóbrega-Pereira
- Department of Medical Sciences and Institute of Biomedicine - iBiMED, University of Aveiro, Aveiro 3810-193, Portugal
| | - Bruno Bernardes de Jesus
- Department of Medical Sciences and Institute of Biomedicine - iBiMED, University of Aveiro, Aveiro 3810-193, Portugal
| |
Collapse
|
4
|
Huang KCY, Chen WTL, Chen JY, Lee CY, Wu CH, Lai CY, Yang PC, Liang JA, Shiau AC, Chao KSC, Ke TW. Neoantigen-augmented iPSC cancer vaccine combined with radiotherapy promotes antitumor immunity in poorly immunogenic cancers. NPJ Vaccines 2024; 9:95. [PMID: 38821980 PMCID: PMC11143272 DOI: 10.1038/s41541-024-00881-5] [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: 12/22/2023] [Accepted: 04/19/2024] [Indexed: 06/02/2024] Open
Abstract
Although irradiated induced-pluripotent stem cells (iPSCs) as a prophylactic cancer vaccine elicit an antitumor immune response, the therapeutic efficacy of iPSC-based cancer vaccines is not promising due to their insufficient antigenicity and the immunosuppressive tumor microenvironment. Here, we found that neoantigen-engineered iPSC cancer vaccines can trigger neoantigen-specific T cell responses to eradicate cancer cells and increase the therapeutic efficacy of RT in poorly immunogenic colorectal cancer (CRC) and triple-negative breast cancer (TNBC). We generated neoantigen-augmented iPSCs (NA-iPSCs) by engineering AAV2 vector carrying murine neoantigens and evaluated their therapeutic efficacy in combination with radiotherapy. After administration of NA-iPSC cancer vaccine and radiotherapy, we found that ~60% of tumor-bearing mice achieved a complete response in microsatellite-stable CRC model. Furthermore, splenocytes from mice treated with NA-iPSC plus RT produced high levels of IFNγ secretion in response to neoantigens and had a greater cytotoxicity to cancer cells, suggesting that the NA-iPSC vaccine combined with radiotherapy elicited a superior neoantigen-specific T-cell response to eradicate cancer cells. The superior therapeutic efficacy of NA-iPSCs engineered by mouse TNBC neoantigens was also observed in the syngeneic immunocompetent TNBC mouse model. We found that the risk of spontaneous lung and liver metastasis was dramatically decreased by NA-iPSCs plus RT in the TNBC animal model. Altogether, these results indicated that autologous iPSC cancer vaccines engineered by neoantigens can elicit a high neoantigen-specific T-cell response, promote tumor regression, and reduce the risk of distant metastasis in combination with local radiotherapy.
Collapse
Affiliation(s)
- Kevin Chih-Yang Huang
- Department of Biomedical Imaging and Radiological Science, China Medical University, Taichung, 406040, Taiwan, ROC.
- Translation Research Core, China Medical University Hospital, China Medical University, Taichung, 404327, Taiwan, ROC.
- Cancer Biology and Precision Therapeutics Center, China Medical University, Taichung, 406040, Taiwan, ROC.
| | - William Tzu-Liang Chen
- Department of Surgery, School of Medicine, China Medical University, Taichung, 406040, Taiwan, ROC
- Department of Colorectal Surgery, China Medical University HsinChu Hospital, China Medical University, HsinChu, 302, Taiwan, ROC
- Department of Colorectal Surgery, China Medical University Hospital, China Medical University, Taichung, 404327, Taiwan, ROC
| | - Jia-Yi Chen
- Proton Therapy and Science Center, China Medical University Hospital, China Medical University, Taichung, 404327, Taiwan, ROC
| | - Chien-Yueh Lee
- Innovation Frontier Institute of Research for Science and Technology, National Taipei University of Technology, Taipei, 106344, Taiwan, ROC
- Department of Electrical Engineering, National Taipei University of Technology, Taipei, 106344, Taiwan, ROC
- Department of Biomedical Engineering, China Medical University, Taichung, 406040, Taiwan, ROC
| | - Chia-Hsin Wu
- Proton Therapy and Science Center, China Medical University Hospital, China Medical University, Taichung, 404327, Taiwan, ROC
- Bioinformatics and Biostatistics Core, Centers of Genomic and Precision Medicine, National Taiwan University, Taipei, 10055, Taiwan, ROC
| | - Chia-Ying Lai
- Department of Biomedical Imaging and Radiological Science, China Medical University, Taichung, 406040, Taiwan, ROC
- Proton Therapy and Science Center, China Medical University Hospital, China Medical University, Taichung, 404327, Taiwan, ROC
| | - Pei-Chen Yang
- Proton Therapy and Science Center, China Medical University Hospital, China Medical University, Taichung, 404327, Taiwan, ROC
| | - Ji-An Liang
- Department of Radiation Oncology, China Medical University Hospital, China Medical University, Taichung, 404327, Taiwan, ROC
- Department of Radiotherapy, School of Medicine, China Medical University, Taichung, 406040, Taiwan, ROC
| | - An-Cheng Shiau
- Department of Biomedical Imaging and Radiological Science, China Medical University, Taichung, 406040, Taiwan, ROC
- Proton Therapy and Science Center, China Medical University Hospital, China Medical University, Taichung, 404327, Taiwan, ROC
- Department of Radiation Oncology, China Medical University Hospital, China Medical University, Taichung, 404327, Taiwan, ROC
| | - K S Clifford Chao
- Proton Therapy and Science Center, China Medical University Hospital, China Medical University, Taichung, 404327, Taiwan, ROC.
- Department of Radiation Oncology, China Medical University Hospital, China Medical University, Taichung, 404327, Taiwan, ROC.
- Department of Radiotherapy, School of Medicine, China Medical University, Taichung, 406040, Taiwan, ROC.
| | - Tao-Wei Ke
- Department of Colorectal Surgery, China Medical University Hospital, China Medical University, Taichung, 404327, Taiwan, ROC.
- School of Chinese Medicine and Graduate Institute of Chinese Medicine, China Medical University, Taichung, 406040, Taiwan, ROC.
| |
Collapse
|
5
|
Chehelgerdi M, Behdarvand Dehkordi F, Chehelgerdi M, Kabiri H, Salehian-Dehkordi H, Abdolvand M, Salmanizadeh S, Rashidi M, Niazmand A, Ahmadi S, Feizbakhshan S, Kabiri S, Vatandoost N, Ranjbarnejad T. Exploring the promising potential of induced pluripotent stem cells in cancer research and therapy. Mol Cancer 2023; 22:189. [PMID: 38017433 PMCID: PMC10683363 DOI: 10.1186/s12943-023-01873-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 09/27/2023] [Indexed: 11/30/2023] Open
Abstract
The advent of iPSCs has brought about a significant transformation in stem cell research, opening up promising avenues for advancing cancer treatment. The formation of cancer is a multifaceted process influenced by genetic, epigenetic, and environmental factors. iPSCs offer a distinctive platform for investigating the origin of cancer, paving the way for novel approaches to cancer treatment, drug testing, and tailored medical interventions. This review article will provide an overview of the science behind iPSCs, the current limitations and challenges in iPSC-based cancer therapy, the ethical and social implications, and the comparative analysis with other stem cell types for cancer treatment. The article will also discuss the applications of iPSCs in tumorigenesis, the future of iPSCs in tumorigenesis research, and highlight successful case studies utilizing iPSCs in tumorigenesis research. The conclusion will summarize the advancements made in iPSC-based tumorigenesis research and the importance of continued investment in iPSC research to unlock the full potential of these cells.
Collapse
Affiliation(s)
- Matin Chehelgerdi
- Novin Genome (NG) Lab, Research and Development Center for Biotechnology, Shahrekord, Iran
- Young Researchers and Elite Club, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran
| | - Fereshteh Behdarvand Dehkordi
- Novin Genome (NG) Lab, Research and Development Center for Biotechnology, Shahrekord, Iran
- Young Researchers and Elite Club, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran
| | - Mohammad Chehelgerdi
- Novin Genome (NG) Lab, Research and Development Center for Biotechnology, Shahrekord, Iran.
- Young Researchers and Elite Club, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran.
| | - Hamidreza Kabiri
- Novin Genome (NG) Lab, Research and Development Center for Biotechnology, Shahrekord, Iran
- Young Researchers and Elite Club, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran
| | | | - Mohammad Abdolvand
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Science, Isfahan, Iran
| | - Sharareh Salmanizadeh
- Department of Cell and Molecular Biology and Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Hezar-Jereeb Street, Isfahan, 81746-73441, Iran
| | - Mohsen Rashidi
- Department Pharmacology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
- The Health of Plant and Livestock Products Research Center, Mazandaran University of Medical Sciences, Sari, Iran
| | - Anoosha Niazmand
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Science, Isfahan, Iran
| | - Saba Ahmadi
- Department of Molecular and Medical Genetics, Tbilisi State Medical University, Tbilisi, Georgia
| | - Sara Feizbakhshan
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Science, Isfahan, Iran
| | - Saber Kabiri
- Novin Genome (NG) Lab, Research and Development Center for Biotechnology, Shahrekord, Iran
- Young Researchers and Elite Club, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran
| | - Nasimeh Vatandoost
- Pediatric Inherited Diseases Research Center, Research Institute for Primordial Prevention of Non-Communicable Disease, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Tayebeh Ranjbarnejad
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Science, Isfahan, Iran
| |
Collapse
|
6
|
Wang R, Zhu T, Hou B, Huang X. An iPSC-derived exosome-pulsed dendritic cell vaccine boosts antitumor immunity in melanoma. Mol Ther 2023; 31:2376-2390. [PMID: 37312452 PMCID: PMC10422017 DOI: 10.1016/j.ymthe.2023.06.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 03/23/2023] [Accepted: 06/08/2023] [Indexed: 06/15/2023] Open
Abstract
Induced pluripotent stem cells (iPSCs) express a broad spectrum of tumor-associated antigens and exert prophylactic effects on various tumors. However, some problems remain, such as potential tumorigenicity, challenges in transport to the lymph nodes and spleen, and limited antitumor effects. Thus, designing a safe and effective iPSC-based tumor vaccine is necessary. We prepared iPSC-derived exosomes and incubated them with DCs (dendritic cells) for pulsing to explore their antitumor effects in murine melanoma models. The antitumor immune response induced by the DC vaccine pulsed with iPSC exosomes (DC + EXO) was assessed in vitro and in vivo. After DC + EXO vaccination, extracted spleen T cells effectively killed a variety of tumor cells (melanoma, lung cancer, breast cancer, and colorectal cancer) in vitro. In addition, DC + EXO vaccination significantly inhibited melanoma growth and lung metastasis in mouse models. Furthermore, DC + EXO vaccination induced long-term T cell responses and prevented melanoma rechallenge. Finally, biocompatibility studies showed that the DC vaccine did not significantly alter the viability of normal cells and mouse viscera. Hence, our research may provide a prospective strategy of a safe and effective iPSC-based tumor vaccine for clinical use.
Collapse
Affiliation(s)
- Ronghao Wang
- Center for Infection and Immunity and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai 519000, Guangdong, China; Department of Gastrointestinal Surgery, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai 519000, Guangdong, China
| | - Tianchuan Zhu
- Center for Infection and Immunity and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai 519000, Guangdong, China
| | - Bingzong Hou
- Department of Gastrointestinal Surgery, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai 519000, Guangdong, China.
| | - Xi Huang
- Center for Infection and Immunity and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai 519000, Guangdong, China; Southern Marine Science and Engineering Guangdong Laboratory, Zhuhai 519000, Guangdong, China.
| |
Collapse
|
7
|
Krog RT, de Miranda NFCC, Vahrmeijer AL, Kooreman NG. The Potential of Induced Pluripotent Stem Cells to Advance the Treatment of Pancreatic Ductal Adenocarcinoma. Cancers (Basel) 2021; 13:cancers13225789. [PMID: 34830945 PMCID: PMC8616212 DOI: 10.3390/cancers13225789] [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: 10/11/2021] [Revised: 11/12/2021] [Accepted: 11/13/2021] [Indexed: 11/16/2022] Open
Abstract
Simple Summary Despite improvements in the treatment of several cancer types, the extremely poor prognosis of pancreatic cancer patients has remained unchanged over the last decades. Therefore, new therapeutic regimens for pancreatic cancer are highly needed. In this review, we will discuss the potential of induced pluripotent stem cells (iPSCs) to generate representative pancreatic cancer models that can aid the development of novel diagnostics and therapeutic strategies. Furthermore, the potential of iPSCs as pancreatic cancer vaccines or as a basis for cellular therapies will be discussed. With promising preclinical results and ongoing clinical trials, the potential of iPSCs to further the treatment of pancreatic cancer is being explored and, in turn, will hopefully provide additional therapies to increase the poor survival rates of this patient population. Abstract Advances in the treatment of pancreatic ductal adenocarcinoma (PDAC) using neoadjuvant chemoradiotherapy, chemotherapy, and immunotherapy have had minimal impact on the overall survival of patients. A general lack of immunogenic features and a complex tumor microenvironment (TME) are likely culprits for therapy refractoriness in PDAC. Induced pluripotent stem cells (iPSCs) should be explored as a means to advance the treatment options for PDAC, by providing representative in vitro models of pancreatic cancer development. In addition, iPSCs could be used for tailor-made cellular immunotherapies or as a source of tumor-associated antigens in the context of vaccination.
Collapse
Affiliation(s)
- Ricki T. Krog
- Department of Surgery, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (R.T.K.); (A.L.V.)
- Department of Pathology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands;
| | | | - Alexander L. Vahrmeijer
- Department of Surgery, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (R.T.K.); (A.L.V.)
| | - Nigel G. Kooreman
- Department of Surgery, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (R.T.K.); (A.L.V.)
- Correspondence:
| |
Collapse
|
8
|
Zhai Y, He X, Li Y, Han R, Ma Y, Gao P, Qian Z, Gu Y, Li S. A splenic-targeted versatile antigen courier: iPSC wrapped in coalescent erythrocyte-liposome as tumor nanovaccine. SCIENCE ADVANCES 2021; 7:eabi6326. [PMID: 34433569 PMCID: PMC8386930 DOI: 10.1126/sciadv.abi6326] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 07/01/2021] [Indexed: 05/05/2023]
Abstract
The major obstacles for tumor vaccine to be surmounted are the lack of versatile property and immunity-inducing effectiveness. Induced pluripotent stem cells (iPSCs) expressed various antigens the same as multiple types of tumors, providing a promising source of wide-spectrum cancer vaccines. The damaged erythrocyte membrane entrapped by spleen could be developed as antigen deliverer for enhancing acquired immunity. Here, the modified lipid materials were used to dilate erythrocyte membrane to fabricate coalescent nanovector, which not only preserved the biological characteristics of erythrocyte membrane but also remedied the defect of insufficient drug loading capacity. After wrapping iPSC protein, the nanovaccine iPSC@RBC-Mlipo exhibited obvious splenic accumulation, systemic specific antitumor immunity evocation, and effective tumor expansion and metastasis inhibition in mice. Hence, our research may provide a prospective strategy of efficient tumor vaccine for clinical practice.
Collapse
Affiliation(s)
- Yuewen Zhai
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Screening, Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, No. 639 Longmian Avenue, Jiangning District, Nanjing 211198, Jiangsu Province, China
| | - Xiaorong He
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Screening, Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, No. 639 Longmian Avenue, Jiangning District, Nanjing 211198, Jiangsu Province, China
| | - Ying Li
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Screening, Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, No. 639 Longmian Avenue, Jiangning District, Nanjing 211198, Jiangsu Province, China
| | - Ran Han
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Screening, Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, No. 639 Longmian Avenue, Jiangning District, Nanjing 211198, Jiangsu Province, China
| | - Yuying Ma
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Screening, Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, No. 639 Longmian Avenue, Jiangning District, Nanjing 211198, Jiangsu Province, China
| | - Peng Gao
- Chia Tai Tianqing Pharmaceutical Group Co. Ltd., No.1099, Fuying Road, Jiangning District, Jiangsu Province, Nanjing, China
| | - Zhiyu Qian
- Department of Biomedical Engineering, School of Automation, Nanjing University of Aeronautics and Astronautics, 29th JiangJun Street, Jiangsu Province, Nanjing 211106, China
| | - Yueqing Gu
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Screening, Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, No. 639 Longmian Avenue, Jiangning District, Nanjing 211198, Jiangsu Province, China.
| | - Siwen Li
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Screening, Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, No. 639 Longmian Avenue, Jiangning District, Nanjing 211198, Jiangsu Province, China.
| |
Collapse
|