1
|
Li YJ, Chien SH, Huang R, Herrmann A, Zhao Q, Li PC, Zhang C, Martincuks A, Santiago NL, Zong K, Swiderski P, Okimoto RA, Song M, Rodriguez L, Forman SJ, Wang X, Yu H. A platform to deliver single and bi-specific Cas9/guide RNA to perturb genes in vitro and in vivo. Mol Ther 2024; 32:3629-3649. [PMID: 39091030 PMCID: PMC11489542 DOI: 10.1016/j.ymthe.2024.07.025] [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: 11/29/2023] [Revised: 06/20/2024] [Accepted: 07/29/2024] [Indexed: 08/04/2024] Open
Abstract
Although CRISPR-Cas9 technology is poised to revolutionize the treatment of diseases with underlying genetic mutations, it faces some significant issues limiting clinical entry. They include low-efficiency in vivo systemic delivery and undesired off-target effects. Here, we demonstrate, by modifying Cas9 with phosphorothioate-DNA oligos (PSs), that one can efficiently deliver single and bi-specific CRISPR-Cas9/guide RNA (gRNA) dimers in vitro and in vivo with reduced off-target effects. We show that PS-Cas9/gRNA-mediated gene knockout preserves chimeric antigen receptor T cell viability and expansion in vitro and in vivo. PS-Cas9/gRNA mediates gene perturbation in patient-derived tumor organoids and mouse xenograft tumors, leading to potent tumor antitumor effects. Further, HER2 antibody-PS-Cas9/gRNA conjugate selectively perturbs targeted genes in HER2+ ovarian cancer xenografts in vivo. Moreover, we created bi-specific PS-Cas9 with two gRNAs to target two adjacent sequences of the same gene, leading to efficient targeted gene disruption ex vivo and in vivo with markedly reduced unintended gene perturbation. Thus, the cell-penetrating PS-Cas9/gRNA can achieve efficient systemic delivery and precision in gene disruption.
Collapse
Affiliation(s)
- Yi-Jia Li
- Department of Immuno-Oncology, Beckman Research Institute and City of Hope Medical Center, Duarte, CA 91010, USA.
| | - Sheng-Hsuan Chien
- Cellular Immunotherapy Center, Department of Hematology and Hematopoietic Cell Transplantation, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA; Division of Transfusion Medicine, Department of Medicine, Taipei Veterans General Hospital, and Institute of Clinical Medicine, National Yang-Ming Chiao Tung University, Taipei 11201, Taiwan
| | - Rui Huang
- Department of Immuno-Oncology, Beckman Research Institute and City of Hope Medical Center, Duarte, CA 91010, USA
| | - Andreas Herrmann
- Department of Immuno-Oncology, Beckman Research Institute and City of Hope Medical Center, Duarte, CA 91010, USA
| | - Qianqian Zhao
- Department of Immuno-Oncology, Beckman Research Institute and City of Hope Medical Center, Duarte, CA 91010, USA
| | - Pei-Chuan Li
- Department of Immuno-Oncology, Beckman Research Institute and City of Hope Medical Center, Duarte, CA 91010, USA
| | - Chunyan Zhang
- Cellular Immunotherapy Center, Department of Hematology and Hematopoietic Cell Transplantation, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
| | - Antons Martincuks
- Department of Immuno-Oncology, Beckman Research Institute and City of Hope Medical Center, Duarte, CA 91010, USA
| | - Nicole Lugo Santiago
- Department of Surgery, Division of Gynecologic Oncology, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Katherine Zong
- Department of Immuno-Oncology, Beckman Research Institute and City of Hope Medical Center, Duarte, CA 91010, USA
| | - Piotr Swiderski
- DNA/RNA Synthesis Laboratory, Beckman Research Institute at City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA
| | - Ross A Okimoto
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA; Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94115, USA
| | - Mihae Song
- Department of Surgery, Division of Gynecologic Oncology, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Lorna Rodriguez
- Department of Surgery, Division of Gynecologic Oncology, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Stephen J Forman
- Cellular Immunotherapy Center, Department of Hematology and Hematopoietic Cell Transplantation, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
| | - Xiuli Wang
- Cellular Immunotherapy Center, Department of Hematology and Hematopoietic Cell Transplantation, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
| | - Hua Yu
- Department of Immuno-Oncology, Beckman Research Institute and City of Hope Medical Center, Duarte, CA 91010, USA.
| |
Collapse
|
2
|
Li M, Xie Y, Zhang J, Zhou X, Gao L, He M, Liu X, Miao X, Liu Y, Cao R, Jia Y, Zeng Z, Liu L. Intratumoral injection of mRNA encoding survivin in combination with STAT3 inhibitor stattic enhances antitumor effects. Cancer Lett 2024; 598:217111. [PMID: 38972347 DOI: 10.1016/j.canlet.2024.217111] [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: 06/14/2024] [Accepted: 07/04/2024] [Indexed: 07/09/2024]
Abstract
Intratumoral delivery of mRNA encoding immunostimulatory molecules can initiate a robust, global antitumor response with little side effects by enhancing local antigen presentation in the tumor and the tumor draining lymph node. Neoantigen-based mRNA nanovaccine can inhibit melanoma growth in mice by intratumoral injection. Myeloid-derived suppressor cells (MDSCs) suppress antitumor immune responses by secreting immunosuppressive agents, such as reactive oxygen species (ROS). Suppression of STAT3 activity by stattic may reduce MDSC-mediated immunosuppression in the TME and promote the antitumor immune responses. In this study, in vitro transcribed mRNA encoding tumor antigen survivin was prepared and injected intratumorally in BALB/c mice bearing subcutaneous colon cancer tumors. In vivo studies demonstrated that intratumoral survivin mRNA therapy could induce antitumor T cell response and inhibit tumor growth of colon cancer. Depletion of CD8+ T cells could significantly inhibit survivin mRNA-induced antitumor effects. RT-qPCR and ELISA analysis indicated that survivin mRNA treatment led to increased expression of receptor activator nuclear factor-κB ligand (RANKL). In vitro experiment showed that MDSCs could be induced from mouse bone marrow cells by RANKL and RANKL-induced MDSCs could produce high level of ROS. STAT3 inhibitor stattic suppressed activation of STAT3 and NF-κB signals, thereby inhibiting expansion of RANKL-induced MDSCs. Combination therapy of survivin mRNA and stattic could significantly enhance antitumor T cell response, improve long-term survival and reduce immunosuppressive tumor microenvironment compared to each monotherapy. In addition, combined therapy resulted in a significantly reduced level of tumor cell proliferation and an obviously increased level of tumor cell apoptosis in CT26 colon cancer-bearing mice, which could be conducive to inhibit the tumor growth and lead to immune responses to released tumor-associated antigens. These studies explored intratumoral mRNA therapy and mRNA-based combined therapy to treat colon cancer and provide a new idea for cancer therapy.
Collapse
Affiliation(s)
- Min Li
- Engineering Research Center of Cellular Immunotherapy of Guizhou Province, Key Laboratory of Infectious Immune and Antibody Engineering of Guizhou Province, School of Basic Medical Science/School of Biology and Engineering (School of Health Medicine Modern Industry), Guizhou Medical University, Guiyang, Guizhou, China
| | - Ying Xie
- Engineering Research Center of Cellular Immunotherapy of Guizhou Province, Key Laboratory of Infectious Immune and Antibody Engineering of Guizhou Province, School of Basic Medical Science/School of Biology and Engineering (School of Health Medicine Modern Industry), Guizhou Medical University, Guiyang, Guizhou, China
| | - Jincheng Zhang
- Engineering Research Center of Cellular Immunotherapy of Guizhou Province, Key Laboratory of Infectious Immune and Antibody Engineering of Guizhou Province, School of Basic Medical Science/School of Biology and Engineering (School of Health Medicine Modern Industry), Guizhou Medical University, Guiyang, Guizhou, China
| | - Xue Zhou
- Engineering Research Center of Cellular Immunotherapy of Guizhou Province, Key Laboratory of Infectious Immune and Antibody Engineering of Guizhou Province, School of Basic Medical Science/School of Biology and Engineering (School of Health Medicine Modern Industry), Guizhou Medical University, Guiyang, Guizhou, China
| | - Lei Gao
- Engineering Research Center of Cellular Immunotherapy of Guizhou Province, Key Laboratory of Infectious Immune and Antibody Engineering of Guizhou Province, School of Basic Medical Science/School of Biology and Engineering (School of Health Medicine Modern Industry), Guizhou Medical University, Guiyang, Guizhou, China
| | - Mengmeng He
- Engineering Research Center of Cellular Immunotherapy of Guizhou Province, Key Laboratory of Infectious Immune and Antibody Engineering of Guizhou Province, School of Basic Medical Science/School of Biology and Engineering (School of Health Medicine Modern Industry), Guizhou Medical University, Guiyang, Guizhou, China
| | - Xianmei Liu
- Engineering Research Center of Cellular Immunotherapy of Guizhou Province, Key Laboratory of Infectious Immune and Antibody Engineering of Guizhou Province, School of Basic Medical Science/School of Biology and Engineering (School of Health Medicine Modern Industry), Guizhou Medical University, Guiyang, Guizhou, China
| | - Xinyi Miao
- Engineering Research Center of Cellular Immunotherapy of Guizhou Province, Key Laboratory of Infectious Immune and Antibody Engineering of Guizhou Province, School of Basic Medical Science/School of Biology and Engineering (School of Health Medicine Modern Industry), Guizhou Medical University, Guiyang, Guizhou, China
| | - Yu Liu
- Engineering Research Center of Cellular Immunotherapy of Guizhou Province, Key Laboratory of Infectious Immune and Antibody Engineering of Guizhou Province, School of Basic Medical Science/School of Biology and Engineering (School of Health Medicine Modern Industry), Guizhou Medical University, Guiyang, Guizhou, China
| | - Rong Cao
- Engineering Research Center of Cellular Immunotherapy of Guizhou Province, Key Laboratory of Infectious Immune and Antibody Engineering of Guizhou Province, School of Basic Medical Science/School of Biology and Engineering (School of Health Medicine Modern Industry), Guizhou Medical University, Guiyang, Guizhou, China
| | - Yi Jia
- Engineering Research Center of Cellular Immunotherapy of Guizhou Province, Key Laboratory of Infectious Immune and Antibody Engineering of Guizhou Province, School of Basic Medical Science/School of Biology and Engineering (School of Health Medicine Modern Industry), Guizhou Medical University, Guiyang, Guizhou, China; Key Laboratory of Biological and Medical Engineering/Immune Cells and Antibody Engineering Research Center of Guizhou Province/Engineering Research Center of Health Medicine Biotechnology of Institution of Higher Education of Guizhou Province, Guizhou Medical University, Guiyang, Guizhou, China.
| | - Zhu Zeng
- Engineering Research Center of Cellular Immunotherapy of Guizhou Province, Key Laboratory of Infectious Immune and Antibody Engineering of Guizhou Province, School of Basic Medical Science/School of Biology and Engineering (School of Health Medicine Modern Industry), Guizhou Medical University, Guiyang, Guizhou, China; Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, Guizhou, China; Key Laboratory of Biological and Medical Engineering/Immune Cells and Antibody Engineering Research Center of Guizhou Province/Engineering Research Center of Health Medicine Biotechnology of Institution of Higher Education of Guizhou Province, Guizhou Medical University, Guiyang, Guizhou, China.
| | - Lina Liu
- Engineering Research Center of Cellular Immunotherapy of Guizhou Province, Key Laboratory of Infectious Immune and Antibody Engineering of Guizhou Province, School of Basic Medical Science/School of Biology and Engineering (School of Health Medicine Modern Industry), Guizhou Medical University, Guiyang, Guizhou, China; Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, Guizhou, China; Key Laboratory of Biological and Medical Engineering/Immune Cells and Antibody Engineering Research Center of Guizhou Province/Engineering Research Center of Health Medicine Biotechnology of Institution of Higher Education of Guizhou Province, Guizhou Medical University, Guiyang, Guizhou, China.
| |
Collapse
|
3
|
Neill B, Romero AR, Fenton OS. Advances in Nonviral mRNA Delivery Materials and Their Application as Vaccines for Melanoma Therapy. ACS APPLIED BIO MATERIALS 2024; 7:4894-4913. [PMID: 37930174 PMCID: PMC11220486 DOI: 10.1021/acsabm.3c00721] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2023]
Abstract
Messenger RNA (mRNA) vaccines are promising platforms for cancer immunotherapy because of their potential to encode for a variety of tumor antigens, high tolerability, and capacity to induce strong antitumor immune responses. However, the clinical translation of mRNA cancer vaccines can be hindered by the inefficient delivery of mRNA in vivo. In this review, we provide an overview of mRNA cancer vaccines by discussing their utility in treating melanoma. Specifically, we begin our review by describing the barriers that can impede mRNA delivery to target cells. We then review native mRNA structure and discuss various modification methods shown to enhance mRNA stability and transfection. Next, we outline the advantages and challenges of three nonviral carrier platforms (lipid nanoparticles, polymeric nanoparticles, and lipopolyplexes) frequently used for mRNA delivery. Last, we summarize preclinical and clinical studies that have investigated nonviral mRNA vaccines for the treatment of melanoma. In writing this review, we aim to highlight innovative nonviral strategies designed to address mRNA delivery challenges while emphasizing the exciting potential of mRNA vaccines as next-generation therapies for the treatment of cancers.
Collapse
Affiliation(s)
- Bevin Neill
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Adriana Retamales Romero
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Owen S. Fenton
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| |
Collapse
|
4
|
Desai N, Chavda V, Singh TRR, Thorat ND, Vora LK. Cancer Nanovaccines: Nanomaterials and Clinical Perspectives. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2401631. [PMID: 38693099 DOI: 10.1002/smll.202401631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 03/30/2024] [Indexed: 05/03/2024]
Abstract
Cancer nanovaccines represent a promising frontier in cancer immunotherapy, utilizing nanotechnology to augment traditional vaccine efficacy. This review comprehensively examines the current state-of-the-art in cancer nanovaccine development, elucidating innovative strategies and technologies employed in their design. It explores both preclinical and clinical advancements, emphasizing key studies demonstrating their potential to elicit robust anti-tumor immune responses. The study encompasses various facets, including integrating biomaterial-based nanocarriers for antigen delivery, adjuvant selection, and the impact of nanoscale properties on vaccine performance. Detailed insights into the complex interplay between the tumor microenvironment and nanovaccine responses are provided, highlighting challenges and opportunities in optimizing therapeutic outcomes. Additionally, the study presents a thorough analysis of ongoing clinical trials, presenting a snapshot of the current clinical landscape. By curating the latest scientific findings and clinical developments, this study aims to serve as a comprehensive resource for researchers and clinicians engaged in advancing cancer immunotherapy. Integrating nanotechnology into vaccine design holds immense promise for revolutionizing cancer treatment paradigms, and this review provides a timely update on the evolving landscape of cancer nanovaccines.
Collapse
Affiliation(s)
- Nimeet Desai
- Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Kandi, Telangana, 502285, India
| | - Vivek Chavda
- Department of Pharmaceutics and Pharmaceutical Technology, L M College of Pharmacy, Ahmedabad, 380009, India
| | | | - Nanasaheb D Thorat
- Limerick Digital Cancer Research Centre (LDCRC), University of Limerick, Castletroy, Limerick, V94T9PX, Ireland
- Department of Physics, Bernal Institute, Castletroy, Limerick, V94T9PX, Ireland
- Nuffield Department of Women's & Reproductive Health, Medical Science Division, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DU, UK
| | - Lalitkumar K Vora
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, Belfast, BT9 7BL, UK
| |
Collapse
|
5
|
Jiang Z, Xu Y, Du G, Sun X. Emerging advances in delivery systems for mRNA cancer vaccines. J Control Release 2024; 370:287-301. [PMID: 38679162 DOI: 10.1016/j.jconrel.2024.04.039] [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: 02/27/2024] [Revised: 04/18/2024] [Accepted: 04/23/2024] [Indexed: 05/01/2024]
Abstract
The success of lipid nanoparticles (LNPs) in treating COVID-19 promotes further research of mRNA vaccines for cancer vaccination. Aiming at overcoming the constraints of currently available mRNA carriers, various alternative nano-vectors have been developed for delivering tumor antigen encoding mRNA and showed versatility to induce potent anti-tumor immunity. The rationally designed nano-vaccines increase the immune activation capacity of the mRNA vaccines by promoting crucial aspects including mRNA stability, cellular uptake, endosomal escape and targeting of immune cells or organs. Herein, we summarized the research progress of various mRNA based nano-vaccines that have been reported for cancer vaccination, including LNPs, lipid enveloped hybrid nanoparticles, polymeric nanoparticles etc. Several strategies that have been reported for further enhancing the immune stimulation efficacy of mRNA nano-vaccines, including developing nano-vaccines for co-delivering adjuvants, combination of immune checkpoint inhibitors, and optimizing the injection routes for boosting immune responses, have been reviewed. The progress of mRNA nano-vaccines in clinical trials and the prospect of the mRNA vaccines for cancer vaccination are also discussed.
Collapse
Affiliation(s)
- Zhimei Jiang
- Department of Pharmacy, Evidence-Based Pharmacy Center, West China Second University Hospital, Sichuan University, Chengdu 610041, China; Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University, Chengdu 610041, China
| | - Yanhua Xu
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Guangsheng Du
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Xun Sun
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
6
|
Huang X, Zhu X, Yang H, Li Q, Gai L, Sui X, Lu H, Feng J. Nanomaterial Delivery Vehicles for the Development of Neoantigen Tumor Vaccines for Personalized Treatment. Molecules 2024; 29:1462. [PMID: 38611742 PMCID: PMC11012694 DOI: 10.3390/molecules29071462] [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: 02/22/2024] [Revised: 03/16/2024] [Accepted: 03/22/2024] [Indexed: 04/14/2024] Open
Abstract
Tumor vaccines have been considered a promising therapeutic approach for treating cancer in recent years. With the development of sequencing technologies, tumor vaccines based on neoantigens or genomes specifically expressed in tumor cells, mainly in the form of peptides, nucleic acids, and dendritic cells, are beginning to receive widespread attention. Therefore, in this review, we have introduced different forms of neoantigen vaccines and discussed the development of these vaccines in treating cancer. Furthermore, neoantigen vaccines are influenced by factors such as antigen stability, weak immunogenicity, and biosafety in addition to sequencing technology. Hence, the biological nanomaterials, polymeric nanomaterials, inorganic nanomaterials, etc., used as vaccine carriers are principally summarized here, which may contribute to the design of neoantigen vaccines for improved stability and better efficacy.
Collapse
Affiliation(s)
- Xiaoyu Huang
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China; (X.H.); (X.Z.); (H.Y.); (Q.L.); (X.S.)
| | - Xiaolong Zhu
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China; (X.H.); (X.Z.); (H.Y.); (Q.L.); (X.S.)
| | - Huan Yang
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China; (X.H.); (X.Z.); (H.Y.); (Q.L.); (X.S.)
| | - Qinyi Li
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China; (X.H.); (X.Z.); (H.Y.); (Q.L.); (X.S.)
| | - Lizhi Gai
- College of Material, Chemistry and Chemical Engineering, Key Laboratory of Organosilicon Chemistry and Material Technology of Ministry of Education, and Key Laboratory of Organosilicon Material Technology of Zhejiang Province, Hangzhou Normal University, Hangzhou 311121, China;
| | - Xinbing Sui
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China; (X.H.); (X.Z.); (H.Y.); (Q.L.); (X.S.)
| | - Hua Lu
- College of Material, Chemistry and Chemical Engineering, Key Laboratory of Organosilicon Chemistry and Material Technology of Ministry of Education, and Key Laboratory of Organosilicon Material Technology of Zhejiang Province, Hangzhou Normal University, Hangzhou 311121, China;
| | - Jiao Feng
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China; (X.H.); (X.Z.); (H.Y.); (Q.L.); (X.S.)
| |
Collapse
|
7
|
Lin F, Lin EZ, Anekoji M, Ichim TE, Hu J, Marincola FM, Jones LD, Kesari S, Ashili S. Advancing personalized medicine in brain cancer: exploring the role of mRNA vaccines. J Transl Med 2023; 21:830. [PMID: 37978542 PMCID: PMC10656921 DOI: 10.1186/s12967-023-04724-0] [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: 08/25/2023] [Accepted: 11/13/2023] [Indexed: 11/19/2023] Open
Abstract
Advancing personalized medicine in brain cancer relies on innovative strategies, with mRNA vaccines emerging as a promising avenue. While the initial use of mRNA vaccines was in oncology, their stunning success in COVID-19 resulted in widespread attention, both positive and negative. Regardless of politically biased opinions, which relate more to the antigenic source than form of delivery, we feel it is important to objectively review this modality as relates to brain cancer. This class of vaccines trigger robust immune responses through MHC-I and MHC-II pathways, in both prophylactic and therapeutic settings. The mRNA platform offers advantages of rapid development, high potency, cost-effectiveness, and safety. This review provides an overview of mRNA vaccine delivery technologies, tumor antigen identification, combination therapies, and recent therapeutic outcomes, with a particular focus on brain cancer. Combinatorial approaches are vital to maximizing mRNA cancer vaccine efficacy, with ongoing clinical trials exploring combinations with adjuvants and checkpoint inhibitors and even adoptive cell therapy. Efficient delivery, neoantigen identification, preclinical studies, and clinical trial results are highlighted, underscoring mRNA vaccines' potential in advancing personalized medicine for brain cancer. Synergistic combinatorial therapies play a crucial role, emphasizing the need for continued research and collaboration in this area.
Collapse
Affiliation(s)
- Feng Lin
- CureScience Institute, 5820 Oberlin Drive Ste 202, San Diego, CA, 92121, USA.
| | - Emma Z Lin
- University of California San Diego, La Jolla, CA, 92093, USA
| | - Misa Anekoji
- CureScience Institute, 5820 Oberlin Drive Ste 202, San Diego, CA, 92121, USA
| | - Thomas E Ichim
- Therapeutic Solutions International, Oceanside, CA, 92056, USA
| | - Joyce Hu
- Sonata Therapeutics, Watertown, MA, 02472, USA
| | | | - Lawrence D Jones
- CureScience Institute, 5820 Oberlin Drive Ste 202, San Diego, CA, 92121, USA
| | - Santosh Kesari
- Saint John's Cancer Institute, Santa Monica, CA, 90404, USA
| | - Shashaanka Ashili
- CureScience Institute, 5820 Oberlin Drive Ste 202, San Diego, CA, 92121, USA
| |
Collapse
|
8
|
Liu X, Huang P, Yang R, Deng H. mRNA Cancer Vaccines: Construction and Boosting Strategies. ACS NANO 2023; 17:19550-19580. [PMID: 37819640 DOI: 10.1021/acsnano.3c05635] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/13/2023]
Abstract
In late 2020, the U.S. Food and Drug Administration (FDA) approved a lipid-based mRNA vaccine for the prevention of COVID-19, which has pushed this field to be more closely studied and motivated researchers to delve deeper into mRNA therapeutics. To date, the research on mRNA cancer vaccines has been developed rapidly, and substantial hopeful therapeutic results have been achieved against various solid tumors in clinical trials. In this review, we first introduce three main components of mRNA cancer vaccines, including mRNA antigens, adjuvants, and delivery vectors. Engineering these components can optimize the therapeutic effects of mRNA cancer vaccines. For instance, appropriate modification of mRNA structure can alleviate the poor stability and innate immunogenicity of mRNA, and the use of mRNA delivery vectors can address the issues of low delivery efficiency in vivo. Second, we emphatically discuss some strategies to further improve the efficacy of mRNA cancer vaccines, namely modulating the immunosuppressive tumor environment, optimizing administration routes, achieving targeting delivery to intended tissues or organs, and employing combination therapy. These strategies can strengthen the tumor inhibitory ability of mRNA cancer vaccines and increase the possibility of tumor elimination. Finally, we point out some challenges in the clinical practice of mRNA cancer vaccines and offer our perspectives on future developments in this rapidly evolving field. It is anticipated that mRNA cancer vaccines will be rapidly developed for clinical cancer therapy in the near future.
Collapse
Affiliation(s)
- Xiaoqing Liu
- School of Advanced Materials and Nanotechnology, Xidian University, Xi'an 710126 China
- Ministry of Education, School of Life Science and Technology, Xidian University & Engineering Research Center of Molecular and Neuro Imaging, Xi'an, Shaanxi 710126, China
- International Joint Research Center for Advanced Medical Imaging and Intelligent Diagnosis and Treatment & Xi'an Key Laboratory of Intelligent Sensing and Regulation of trans-Scale Life Information, School of Life Science and Technology, Xidian University, Xi'an, Shaanxi 710126, China
| | - Pei Huang
- Ministry of Education, School of Life Science and Technology, Xidian University & Engineering Research Center of Molecular and Neuro Imaging, Xi'an, Shaanxi 710126, China
- International Joint Research Center for Advanced Medical Imaging and Intelligent Diagnosis and Treatment & Xi'an Key Laboratory of Intelligent Sensing and Regulation of trans-Scale Life Information, School of Life Science and Technology, Xidian University, Xi'an, Shaanxi 710126, China
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and Faculty of Engineering, National University of Singapore, Singapore 119074, Singapore
| | - Rusen Yang
- School of Advanced Materials and Nanotechnology, Xidian University, Xi'an 710126 China
| | - Hongzhang Deng
- Ministry of Education, School of Life Science and Technology, Xidian University & Engineering Research Center of Molecular and Neuro Imaging, Xi'an, Shaanxi 710126, China
- International Joint Research Center for Advanced Medical Imaging and Intelligent Diagnosis and Treatment & Xi'an Key Laboratory of Intelligent Sensing and Regulation of trans-Scale Life Information, School of Life Science and Technology, Xidian University, Xi'an, Shaanxi 710126, China
| |
Collapse
|
9
|
Ni L. Advances in mRNA-Based Cancer Vaccines. Vaccines (Basel) 2023; 11:1599. [PMID: 37897001 PMCID: PMC10611059 DOI: 10.3390/vaccines11101599] [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: 09/06/2023] [Revised: 10/08/2023] [Accepted: 10/13/2023] [Indexed: 10/29/2023] Open
Abstract
Cancer is a leading cause of death worldwide, accounting for millions of deaths every year. Immunotherapy is a groundbreaking approach for treating cancer through harnessing the power of the immune system to target and eliminate cancer cells. Cancer vaccines, one immunotherapy approach, have shown promise in preclinical settings, but researchers have struggled to reproduce these results in clinical settings. However, with the maturity of mRNA technology and its success in tackling the recent coronavirus disease 2019 (COVID-19) pandemic, cancer vaccines are expected to regain attention. In this review, we focused on the recent progress made in mRNA-based cancer vaccines over the past five years. The mechanism of action of mRNA vaccines, advancements in neoantigen discovery, adjuvant identification, and delivery materials are summarized and reviewed. In addition, we also provide a detailed overview of current clinical trials involving mRNA cancer vaccines. Lastly, we offer an insight into future considerations for the application of mRNA vaccines in cancer immunotherapy. This review will help researchers to understand the advances in mRNA-based cancer vaccines and explore new dimensions for potential immunotherapy approaches.
Collapse
Affiliation(s)
- Ling Ni
- Institute for Immunology and School of Medicine, Tsinghua University, Medical Research Building, No. 30 Haidian Shuangqing Road, Beijing 100084, China
| |
Collapse
|
10
|
Abstract
RNA modification is manifested as chemically altered nucleotides, widely exists in diverse natural RNAs, and is closely related to RNA structure and function. Currently, mRNA-based vaccines have received great attention and rapid development as novel and mighty fighters against various diseases including cancer. The achievement of RNA vaccines in clinical application is largely attributed to some methodological innovations including the incorporation of modified nucleotides into the synthetic RNA. The selection of optimal RNA modifications aimed at reducing the instability and immunogenicity of RNA molecules is a very critical task to improve the efficacy and safety of mRNA vaccines. This review summarizes the functions of RNA modifications and their application in mRNA vaccines, highlights recent advances of mRNA vaccines in cancer immunotherapy, and provides perspectives for future development of mRNA vaccines in the context of personalized tumor therapy.
Collapse
Affiliation(s)
- Yingxue Mei
- Institute of Cancer Stem Cell, Cancer Center, Dalian Medical University, Dalian, 116044, Liaoning, People's Republic of China
| | - Xiang Wang
- Institute of Cancer Stem Cell, Cancer Center, Dalian Medical University, Dalian, 116044, Liaoning, People's Republic of China.
| |
Collapse
|
11
|
Zhang A, Ji Q, Sheng X, Wu H. mRNA vaccine in gastrointestinal tumors: Immunomodulatory effects and immunotherapy. Biomed Pharmacother 2023; 166:115361. [PMID: 37660645 DOI: 10.1016/j.biopha.2023.115361] [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: 07/03/2023] [Revised: 08/04/2023] [Accepted: 08/22/2023] [Indexed: 09/05/2023] Open
Abstract
Gastrointestinal tumors remain a significant healthcare burden worldwide, necessitating the development of innovative therapeutic strategies. mRNA vaccines have emerged as a promising approach in cancer immunotherapy, harnessing the immune system's potential to recognize and eliminate tumor cells. mRNA vaccines offer several advantages, including their ability to elicit both innate and adaptive immune responses, ease of production, and adaptability to different tumor types. In the context of gastrointestinal tumors, mRNA vaccines hold great potential as a therapeutic strategy. In this review, we will delve into the immunomodulatory mechanisms and immunotherapy strategies of mRNA vaccines in gastrointestinal tumors. Additionally, we will discuss the challenges and ongoing research efforts in optimizing mRNA vaccine development, delivery, and stability. By understanding the potential of mRNA vaccines in addressing the unmet medical need of gastrointestinal tumors, we aim to pave the way for improved treatment strategies and better patient outcomes.
Collapse
Affiliation(s)
- Ao Zhang
- Department of Neurology, The First Hospital of Jilin University, Changchun 130012, China
| | - Qingming Ji
- Department of Intensive Care Medicine, The First Hospital of Jilin University, Changchun 130012, China
| | - Xia Sheng
- Department of Cadre Ward, The First Hospital of Jilin University, Changchun 130012, China
| | - Hui Wu
- Department of Ophthalmology, The First Hospital of Jilin University, Changchun 130012, China.
| |
Collapse
|
12
|
Jackson Hoffman BA, Pumford EA, Enueme AI, Fetah KL, Friedl OM, Kasko AM. Engineered macromolecular Toll-like receptor agents and assemblies. Trends Biotechnol 2023; 41:1139-1154. [PMID: 37068999 DOI: 10.1016/j.tibtech.2023.03.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 03/06/2023] [Accepted: 03/13/2023] [Indexed: 04/19/2023]
Abstract
Macromolecular Toll-like receptor (TLR) agents have been utilized as agonists and inhibitors in preclinical and clinical settings. These agents interface with the TLR class of innate immune receptors which recognize macromolecular ligands that are characteristic of pathogenic material. As such, many agents that have been historically investigated are derived from the natural macromolecules which activate or inhibit TLRs. This review covers recent research and clinically available TLR agents that are macromolecular or polymeric. Synthetic materials that have been found to interface with TLRs are also discussed. Assemblies of these materials are investigated in the context of improving stability or efficacy of ligands. Attention is given to strategies which modify or enhance the current agents and to future outlooks on the development of these agents.
Collapse
Affiliation(s)
| | - Elizabeth A Pumford
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Amaka I Enueme
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Kirsten L Fetah
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Olivia M Friedl
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Andrea M Kasko
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA 90095, USA; California Nanosystems Institute, University of California, Los Angeles, CA 90095, USA.
| |
Collapse
|
13
|
Wang QT, Liu YX, Wang J, Wang H. Advances in Cancer Nanovaccines: Harnessing Nanotechnology for Broadening Cancer Immune Response. ChemMedChem 2023; 18:e202200673. [PMID: 37088719 DOI: 10.1002/cmdc.202200673] [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: 02/03/2023] [Revised: 04/20/2023] [Accepted: 04/20/2023] [Indexed: 04/25/2023]
Abstract
Many advances have been made recently in the field of cancer immunotherapy, particularly with the development of treatments such as immune checkpoint inhibitors and adoptive cellular immunotherapy. The efficacy of immunotherapy is limited, however, owing to high levels of tumor heterogeneity and the immunosuppressive environments of advanced malignant tumors. Therefore, therapeutic anticancer vaccines have gradually become powerful tools for inducing valid antitumor immune responses and regulating the immune microenvironment. Tumor vaccines loaded in nanocarriers have become an indispensable delivery platform for tumor treatment because of their enhanced stability, targeting capability, and high level of safety. Through a unique design, cancer nanovaccines activate innate immunity and tumor-specific immunity simultaneously. For example, the design of cancer vaccines can incorporate strategies such as enhancing the stability and targeting of tumor antigens, combining effective adjuvants, cytokines, and immune microenvironment regulators, and promoting the maturation and cross-presentation of antigen-presenting cells (APCs). In this review, we discuss the design and preparation of nanovaccines for remodeling tumor antigen immunogenicity and regulating the immunosuppressive microenvironment.
Collapse
Affiliation(s)
- Qian-Ting Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology (NCNST), Beijing, 100190, P. R. China
| | - Yi-Xuan Liu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology (NCNST), Beijing, 100190, P. R. China
- University of the Chinese Academy of Sciences (UCAS), Beijing, 100049, P. R. China
| | - Jie Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology (NCNST), Beijing, 100190, P. R. China
| | - Hao Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology (NCNST), Beijing, 100190, P. R. China
| |
Collapse
|
14
|
Chen X, Zhang P, Li P, Wang G, Li J, Wu Y, Cao Z, Zhou Y, Sun Y. CpG ODN 1668 as TLR9 agonist mediates humpback grouper (Cromileptes altivelis) antibacterial immune responses. FISH & SHELLFISH IMMUNOLOGY 2023; 138:108839. [PMID: 37207883 DOI: 10.1016/j.fsi.2023.108839] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 05/16/2023] [Accepted: 05/17/2023] [Indexed: 05/21/2023]
Abstract
Cromileptes altivelis (humpback grouper) is the main farmed species in the southern coastal area of China owing to its important economic value. Toll-like receptor 9 (TLR9) belongs to the toll-like receptor (TLR) family and functions as a pattern recognition receptor, recognising unmethylated oligodeoxynucleotides containing the CpG motif (CpG ODNs) in bacterial and viral genomes, thereby activating host immune response. In this study, the C. altivelis TLR9 (CaTLR9) ligand CpG ODN 1668 was screened and found to significantly enhance the antibacterial immunity of humpback grouper in vivo and head kidney lymphocytes (HKLs) in vitro. In addition, CpG ODN 1668 also promoted the cell proliferation and immune gene expression of HKLs and strengthened the phagocytosis activity of head kidney macrophages. However, when the CaTLR9 expression was knocked down in the humpback group, the expression levels of TLR9, myeloid differentiation factor 88 (Myd88), tumour necrosis factor-α (TNF-α), interferon γ (IFN-γ), interleukin-1β (IL-1β), IL-6, and IL-8 were significantly reduced, and the antibacterial immune effects induced by CpG ODN 1668 were mostly abolished. Therefore, CpG ODN 1668 induced antibacterial immune responses in a CaTLR9-dependent pathway. These results enhance the knowledge of the antibacterial immunity of fish TLR signalling pathways and have important implications for exploring natural antibacterial molecules in fish.
Collapse
Affiliation(s)
- Xiaojuan Chen
- Collaborative Innovation Center of Marine Science and Technology, State Key Laboratory of Marine Resource Utilization in South China Sea, Hainan University, Haikou, 570228, China; Hainan Provincial Key Laboratory for Tropical Hydrobiology and Biotechnology, College of Marine Science, Hainan University, Haikou, 570228, China
| | - Panpan Zhang
- Sanya Nanfan Research Institute, Hainan University, Sanya, 572022, China; Collaborative Innovation Center of Marine Science and Technology, State Key Laboratory of Marine Resource Utilization in South China Sea, Hainan University, Haikou, 570228, China
| | - Pengshuo Li
- Sanya Nanfan Research Institute, Hainan University, Sanya, 572022, China; Hainan Provincial Key Laboratory for Tropical Hydrobiology and Biotechnology, College of Marine Science, Hainan University, Haikou, 570228, China
| | - Guotao Wang
- Sanya Nanfan Research Institute, Hainan University, Sanya, 572022, China; Collaborative Innovation Center of Marine Science and Technology, State Key Laboratory of Marine Resource Utilization in South China Sea, Hainan University, Haikou, 570228, China
| | - Jianlong Li
- Collaborative Innovation Center of Marine Science and Technology, State Key Laboratory of Marine Resource Utilization in South China Sea, Hainan University, Haikou, 570228, China; Hainan Provincial Key Laboratory for Tropical Hydrobiology and Biotechnology, College of Marine Science, Hainan University, Haikou, 570228, China
| | - Ying Wu
- Collaborative Innovation Center of Marine Science and Technology, State Key Laboratory of Marine Resource Utilization in South China Sea, Hainan University, Haikou, 570228, China; Hainan Provincial Key Laboratory for Tropical Hydrobiology and Biotechnology, College of Marine Science, Hainan University, Haikou, 570228, China
| | - Zhenjie Cao
- Collaborative Innovation Center of Marine Science and Technology, State Key Laboratory of Marine Resource Utilization in South China Sea, Hainan University, Haikou, 570228, China; Hainan Provincial Key Laboratory for Tropical Hydrobiology and Biotechnology, College of Marine Science, Hainan University, Haikou, 570228, China
| | - Yongcan Zhou
- Collaborative Innovation Center of Marine Science and Technology, State Key Laboratory of Marine Resource Utilization in South China Sea, Hainan University, Haikou, 570228, China; Hainan Provincial Key Laboratory for Tropical Hydrobiology and Biotechnology, College of Marine Science, Hainan University, Haikou, 570228, China
| | - Yun Sun
- Sanya Nanfan Research Institute, Hainan University, Sanya, 572022, China; Collaborative Innovation Center of Marine Science and Technology, State Key Laboratory of Marine Resource Utilization in South China Sea, Hainan University, Haikou, 570228, China; Hainan Provincial Key Laboratory for Tropical Hydrobiology and Biotechnology, College of Marine Science, Hainan University, Haikou, 570228, China.
| |
Collapse
|
15
|
Bafaloukos D, Gazouli I, Koutserimpas C, Samonis G. Evolution and Progress of mRNA Vaccines in the Treatment of Melanoma: Future Prospects. Vaccines (Basel) 2023; 11:vaccines11030636. [PMID: 36992220 DOI: 10.3390/vaccines11030636] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 03/01/2023] [Accepted: 03/09/2023] [Indexed: 03/14/2023] Open
Abstract
mRNA vaccines encoding tumor antigens may be able to sensitize the immune system of the host against cancer cells, enhancing antigen presentation and immune response. Since the breakout of the COVID19 pandemic, interest in mRNA vaccines has been accelerating, as vaccination against the virus served as a measure to limit disease spread. Given that immunotherapy has been the cornerstone of melanoma treatment over the last several decades, further innate immunity enhancement by targeted mRNA vaccines could be the next pivotal achievement in melanoma treatment. Preclinical data coming from murine cancer models have already provided evidence of mRNA vaccines’ ability to induce host immune responses against cancer. Moreover, specific immune responses have been observed in melanoma patients receiving mRNA vaccines, while the recent KEYNOTE-942 trial may establish the incorporation of the mRNA-4157/V940 vaccine into the melanoma treatment algorithm, in combination with immune checkpoint inhibition. As the existing data are further tested and reviewed, investigators are already gaining enthusiasm about this novel, promising pathway in cancer therapy.
Collapse
Affiliation(s)
- Dimitrios Bafaloukos
- First Department of Medical Oncology, "Metropolitan" Hospital, Neon Faliron, 18547 Attica, Greece
| | - Ioanna Gazouli
- First Department of Medical Oncology, "Metropolitan" Hospital, Neon Faliron, 18547 Attica, Greece
| | - Christos Koutserimpas
- Department of Orthopaedics and Traumatology, "251" Hellenic Air Force General Hospital of Athens, 11525 Athens, Greece
| | - George Samonis
- First Department of Medical Oncology, "Metropolitan" Hospital, Neon Faliron, 18547 Attica, Greece
- Department of Medicine, University of Crete, 71500 Heraklion, Greece
| |
Collapse
|
16
|
Bahreyni A, Mohamud Y, Luo H. Recent advancements in immunotherapy of melanoma using nanotechnology-based strategies. Biomed Pharmacother 2023; 159:114243. [PMID: 36641926 DOI: 10.1016/j.biopha.2023.114243] [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: 12/07/2022] [Revised: 01/07/2023] [Accepted: 01/10/2023] [Indexed: 01/14/2023] Open
Abstract
Melanoma is a malignant tumor that accounts for the deadliest form of skin cancers. Despite the significant efforts made recently for development of immunotherapeutic strategies including using immune checkpoint inhibitors and cancer vaccines, the clinical outcomes are unsatisfying. Different factors affect efficient cancer immunotherapy such as side-effects, immunosuppressive tumor microenvironment, and tumor heterogeneity. In the past decades, various nanotechnology-based approaches have been developed to enhance the efficacy of cancer immunotherapy, in addition to diminishing the toxicity associated with it. Several studies have shown that proper application of nanomaterials can revolutionize the outcome of immunotherapy in diverse melanoma models. This review summarizes the recent advancement in the integration of nanotechnology and cancer immunotherapy in melanoma treatment. The importance of nanomaterials and their therapeutic advantages for patients with melanoma are also discussed.
Collapse
Affiliation(s)
- Amirhossein Bahreyni
- Centre for Heart Lung Innovation, St Paul's Hospital, Vancouver, BC V6Z 1Y6, Canada; Department of Pathology and Laboratory of Medicine, University of British Columbia, Vancouver, BC V6Z 1Y6, Canada
| | - Yasir Mohamud
- Centre for Heart Lung Innovation, St Paul's Hospital, Vancouver, BC V6Z 1Y6, Canada; Department of Pathology and Laboratory of Medicine, University of British Columbia, Vancouver, BC V6Z 1Y6, Canada
| | - Honglin Luo
- Centre for Heart Lung Innovation, St Paul's Hospital, Vancouver, BC V6Z 1Y6, Canada; Department of Pathology and Laboratory of Medicine, University of British Columbia, Vancouver, BC V6Z 1Y6, Canada.
| |
Collapse
|
17
|
Li X, Ma S, Gao T, Mai Y, Song Z, Yang J. The main battlefield of mRNA vaccine – Tumor immune microenvironment. Int Immunopharmacol 2022; 113:109367. [DOI: 10.1016/j.intimp.2022.109367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 10/03/2022] [Accepted: 10/15/2022] [Indexed: 11/05/2022]
|
18
|
Li Y, Fang H, Zhang T, Wang Y, Qi T, Li B, Jiao H. Lipid-mRNA nanoparticles landscape for cancer therapy. Front Bioeng Biotechnol 2022; 10:1053197. [PMID: 36394007 PMCID: PMC9659646 DOI: 10.3389/fbioe.2022.1053197] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Accepted: 10/18/2022] [Indexed: 09/19/2023] Open
Abstract
Intracellular delivery of message RNA (mRNA) technique has ushered in a hopeful era with the successive authorization of two mRNA vaccines for the Coronavirus disease-19 (COVID-19) pandemic. A wide range of clinical studies are proceeding and will be initiated in the foreseeable future to treat and prevent cancers. However, efficient and non-toxic delivery of therapeutic mRNAs maintains the key limited step for their widespread applications in human beings. mRNA delivery systems are in urgent demand to resolve this difficulty. Recently lipid nanoparticles (LNPs) vehicles have prospered as powerful mRNA delivery tools, enabling their potential applications in malignant tumors via cancer immunotherapy and CRISPR/Cas9-based gene editing technique. This review discusses formulation components of mRNA-LNPs, summarizes the latest findings of mRNA cancer therapy, highlights challenges, and offers directions for more effective nanotherapeutics for cancer patients.
Collapse
Affiliation(s)
- Yin Li
- College of Animal Science, Jilin University, Changchun, Jilin, China
| | - Hengtong Fang
- College of Animal Science, Jilin University, Changchun, Jilin, China
| | - Tao Zhang
- College of Animal Science, Jilin University, Changchun, Jilin, China
| | - Yu Wang
- College of Animal Science, Jilin University, Changchun, Jilin, China
| | - Tingting Qi
- College of Animal Science, Jilin University, Changchun, Jilin, China
| | - Bai Li
- Department of Colorectal and Anal Surgery, General Surgery Center, First Hospital of Jilin University, Changchun, Jilin, China
| | - Huping Jiao
- College of Animal Science, Jilin University, Changchun, Jilin, China
| |
Collapse
|
19
|
Kumar M, Dogra R, Mandal UK. Nanomaterial-based delivery of vaccine through nasal route: Opportunities, challenges, advantages, and limitations. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.103533] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
20
|
Fang X, Guo Z, Liang J, Wen J, Liu Y, Guan X, Li H. Neoantigens and their potential applications in tumor immunotherapy. Oncol Lett 2022; 23:88. [PMID: 35126730 PMCID: PMC8805178 DOI: 10.3892/ol.2022.13208] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 01/04/2022] [Indexed: 12/23/2022] Open
Abstract
The incidence of malignant tumors is increasing, the majority of which are associated with high morbidity and mortality rates worldwide. The traditional treatment method for malignant tumors is surgery, coupled with radiotherapy or chemotherapy. However, these therapeutic strategies are frequently accompanied with adverse side effects. Over recent decades, tumor immunotherapy shown promise in demonstrating notable efficacy for the treatment of cancer. With the development of sequencing technology and bioinformatics algorithms, neoantigens have become compelling targets for cancer immunotherapy due to high levels of immunogenicity. In addition, neoantigen-based vaccines have demonstrated potential for cancer therapy, primarily by augmenting T-cell responses. Neoantigens have also been shown to be effective in immune checkpoint blockade therapy. Therefore, neoantigens may serve to be predictive biomarkers and synergistic treatment targets in cancer immunotherapy. The aim of the present review was to provide an overview of the recent progress in the classification, screening and clinical application of neoantigens for cancer therapy.
Collapse
Affiliation(s)
- Xianzhu Fang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Weifang Medical University, Weifang, Shandong 261053, P.R. China
| | - Zhiliang Guo
- Department of Orthopedic, The 80th Group Army Hospital of Chinese People's Liberation Army, Weifang, Shandong 261021, P.R. China
| | - Jinqing Liang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Weifang Medical University, Weifang, Shandong 261053, P.R. China
| | - Jiao Wen
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Weifang Medical University, Weifang, Shandong 261053, P.R. China
| | - Yuanyuan Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Weifang Medical University, Weifang, Shandong 261053, P.R. China
| | - Xiumei Guan
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Weifang Medical University, Weifang, Shandong 261053, P.R. China
| | - Hong Li
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Weifang Medical University, Weifang, Shandong 261053, P.R. China
| |
Collapse
|
21
|
Tang J, Cai L, Xu C, Sun S, Liu Y, Rosenecker J, Guan S. Nanotechnologies in Delivery of DNA and mRNA Vaccines to the Nasal and Pulmonary Mucosa. NANOMATERIALS 2022; 12:nano12020226. [PMID: 35055244 PMCID: PMC8777913 DOI: 10.3390/nano12020226] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 01/03/2022] [Accepted: 01/05/2022] [Indexed: 02/07/2023]
Abstract
Recent advancements in the field of in vitro transcribed mRNA (IVT-mRNA) vaccination have attracted considerable attention to such vaccination as a cutting-edge technique against infectious diseases including COVID-19 caused by SARS-CoV-2. While numerous pathogens infect the host through the respiratory mucosa, conventional parenterally administered vaccines are unable to induce protective immunity at mucosal surfaces. Mucosal immunization enables the induction of both mucosal and systemic immunity, efficiently removing pathogens from the mucosa before an infection occurs. Although respiratory mucosal vaccination is highly appealing, successful nasal or pulmonary delivery of nucleic acid-based vaccines is challenging because of several physical and biological barriers at the airway mucosal site, such as a variety of protective enzymes and mucociliary clearance, which remove exogenously inhaled substances. Hence, advanced nanotechnologies enabling delivery of DNA and IVT-mRNA to the nasal and pulmonary mucosa are urgently needed. Ideal nanocarriers for nucleic acid vaccines should be able to efficiently load and protect genetic payloads, overcome physical and biological barriers at the airway mucosal site, facilitate transfection in targeted epithelial or antigen-presenting cells, and incorporate adjuvants. In this review, we discuss recent developments in nucleic acid delivery systems that target airway mucosa for vaccination purposes.
Collapse
Affiliation(s)
- Jie Tang
- Department of Pediatrics, Ludwig-Maximilians University of Munich, 80337 Munich, Germany;
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane 4072, Australia;
| | - Larry Cai
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane 4072, Australia;
| | - Chuanfei Xu
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, Third Military Medical University, Chongqing 400038, China; (C.X.); (S.S.); (Y.L.)
| | - Si Sun
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, Third Military Medical University, Chongqing 400038, China; (C.X.); (S.S.); (Y.L.)
| | - Yuheng Liu
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, Third Military Medical University, Chongqing 400038, China; (C.X.); (S.S.); (Y.L.)
| | - Joseph Rosenecker
- Department of Pediatrics, Ludwig-Maximilians University of Munich, 80337 Munich, Germany;
- Correspondence: (J.R.); (S.G.); Tel.: +49-89-440057713 (J.R.); +86-23-68771645 (S.G.)
| | - Shan Guan
- Department of Pediatrics, Ludwig-Maximilians University of Munich, 80337 Munich, Germany;
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, Third Military Medical University, Chongqing 400038, China; (C.X.); (S.S.); (Y.L.)
- Correspondence: (J.R.); (S.G.); Tel.: +49-89-440057713 (J.R.); +86-23-68771645 (S.G.)
| |
Collapse
|