1
|
Nguyen QTT, Kim J, Yoo HC, Lee EJ. Strategies to overcome chemoresistance in epithelial ovarian cancer: Navigating beyond challenges. Crit Rev Oncol Hematol 2025; 210:104706. [PMID: 40127787 DOI: 10.1016/j.critrevonc.2025.104706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 02/04/2025] [Accepted: 03/14/2025] [Indexed: 03/26/2025] Open
Abstract
Epithelial ovarian cancer (EOC) is the most fetal gynecological malignancy. The main causes of treatment failure are primary and acquired chemoresistance that remains a major therapeutic challenge. The mechanisms underlying chemoresistance in EOC are complex and not fully understood. This review explores novel therapeutic strategies targeting chemoresistant EOC, including advanced drug delivery systems, targeting non-coding RNAs, peptide-based therapies, immunotherapy, and the use of poly-ADP ribose polymerase inhibitors. By summarizing the latest research and potential treatments, this review aims to contribute to the development of more effective therapies for patients with chemoresistant EOC.
Collapse
Affiliation(s)
- Que Thanh Thanh Nguyen
- Department of Obstetrics and Gynecology, School of Medicine, Chung-Ang University, Seoul 06974, Republic of Korea; Organoid Medical Research Center, Chung-Ang University, Seoul 06974, Republic of Korea
| | - Jihye Kim
- Department of Obstetrics and Gynecology, School of Medicine, Chung-Ang University, Seoul 06974, Republic of Korea
| | - Hee Chan Yoo
- Organoid Medical Research Center, Chung-Ang University, Seoul 06974, Republic of Korea; College of Pharmacy, Chung-Ang University, Seoul 06974, Republic of Korea.
| | - Eun-Ju Lee
- Department of Obstetrics and Gynecology, School of Medicine, Chung-Ang University, Seoul 06974, Republic of Korea; Organoid Medical Research Center, Chung-Ang University, Seoul 06974, Republic of Korea.
| |
Collapse
|
2
|
Giram P, Md Mahabubur Rahman K, Aqel O, You Y. In Situ Cancer Vaccines: Redefining Immune Activation in the Tumor Microenvironment. ACS Biomater Sci Eng 2025; 11:2550-2583. [PMID: 40223683 DOI: 10.1021/acsbiomaterials.5c00121] [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] [Indexed: 04/15/2025]
Abstract
Cancer is one of the leading causes of mortality worldwide. Nanomedicines have significantly improved life expectancy and survival rates for cancer patients in current standard care. However, recurrence of cancer due to metastasis remains a significant challenge. Vaccines can provide long-term protection and are ideal for preventing bacterial and viral infections. Cancer vaccines, however, have shown limited therapeutic efficacy and raised safety concerns despite extensive research. Cancer vaccines target and stimulate responses against tumor-specific antigens and have demonstrated great potential for cancer treatment in preclinical studies. However, tumor-associated immunosuppression and immune tolerance driven by immunoediting pose significant challenges for vaccine design. In situ vaccination represents an alternative approach to traditional cancer vaccines. This strategy involves the intratumoral administration of immunostimulants to modulate the growth and differentiation of innate immune cells, such as dendritic cells, macrophages, and neutrophils, and restore T-cell activity. Currently approved in situ vaccines, such as T-VEC, have demonstrated clinical promise, while ongoing clinical trials continue to explore novel strategies for broader efficacy. Despite these advancements, failures in vaccine research highlight the need to address tumor-associated immune suppression and immune escape mechanisms. In situ vaccination strategies combine innate and adaptive immune stimulation, leveraging tumor-associated antigens to activate dendritic cells and cross-prime CD8+ T cells. Various vaccine modalities, such as nucleotide-based vaccines (e.g., RNA and DNA vaccines), peptide-based vaccines, and cell-based vaccines (including dendritic, T-cell, and B-cell approaches), show significant potential. Plant-based viral approaches, including cowpea mosaic virus and Newcastle disease virus, further expand the toolkit for in situ vaccination. Therapeutic modalities such as chemotherapy, radiation, photodynamic therapy, photothermal therapy, and Checkpoint blockade inhibitors contribute to enhanced antigen presentation and immune activation. Adjuvants like CpG-ODN and PRR agonists further enhance immune modulation and vaccine efficacy. The advantages of in situ vaccination include patient specificity, personalization, minimized antigen immune escape, and reduced logistical costs. However, significant barriers such as tumor heterogeneity, immune evasion, and logistical challenges remain. This review explores strategies for developing potent cancer vaccines, examines ongoing clinical trials, evaluates immune stimulation methods, and discusses prospects for advancing in situ cancer vaccination.
Collapse
Affiliation(s)
- Prabhanjan Giram
- Department of Pharmaceutical Sciences, University at Buffalo, The State University of New York, Buffalo, New York 14214, United States
| | - Kazi Md Mahabubur Rahman
- Department of Pharmaceutical Sciences, University at Buffalo, The State University of New York, Buffalo, New York 14214, United States
| | - Osama Aqel
- Department of Pharmaceutical Sciences, University at Buffalo, The State University of New York, Buffalo, New York 14214, United States
| | - Youngjae You
- Department of Pharmaceutical Sciences, University at Buffalo, The State University of New York, Buffalo, New York 14214, United States
| |
Collapse
|
3
|
Afrashteh F, Seyedpour S, Rezaei N. The therapeutic effect of mRNA vaccines in glioma: a comprehensive review. Expert Rev Clin Immunol 2025; 21:603-615. [PMID: 40249391 DOI: 10.1080/1744666x.2025.2494656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Revised: 03/18/2025] [Accepted: 04/14/2025] [Indexed: 04/19/2025]
Abstract
INTRODUCTION Glioma is the most common primary brain tumor, with glioblastoma being the most lethal type due to its heterogeneous and invasive nature of the cancer. Current therapies have low curative success and are limited to surgery, radiotherapy, and chemotherapy. More than 50% of patients become resistant to chemotherapy, and tumor recurrence occurs in most patients following an initial course of therapy. Therefore, developing novel, effective strategies for glioma treatment is essential. Cancer vaccines are novel therapies that demonstrate advantages over conventional methods and, therefore, may be promising options for treating glioma. AREAS COVERED This article provided a critical review of pre-clinical and clinical studies that explored appropriate tumor antigen candidates for developing mRNA vaccines and discussed their clinical application in glioma patients. Medline database, PubMed, and ClinicalTrials.gov were searched for glioma vaccine studies published before 2025 using related keywords. EXPERT OPINION mRNA vaccines are promising strategies for treating glioma because they are efficient, cost-beneficial, and have lower side effects than other types such as peptide or DNA-based vaccines.
Collapse
Affiliation(s)
- Fatemeh Afrashteh
- Student Research Committee, School of Medicine, Iran University of Medical Sciences (IUMS), Tehran, Iran
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Simin Seyedpour
- Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
- Nanomedicine Research Association (NRA), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Nima Rezaei
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran
- Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
4
|
Zhang W, Wang C, Meng Y, He L, Dong M. EBV Vaccines in the Prevention and Treatment of Nasopharyngeal Carcinoma. Vaccines (Basel) 2025; 13:478. [PMID: 40432090 DOI: 10.3390/vaccines13050478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2025] [Revised: 04/21/2025] [Accepted: 04/22/2025] [Indexed: 05/29/2025] Open
Abstract
Epstein-Barr virus (EBV), a ubiquitous human herpesvirus, has been robustly linked to the pathogenesis of nasopharyngeal carcinoma (NPC). The mechanism of EBV-induced NPC involves complex interactions between viral proteins and host cell pathways. This review aims to comprehensively outline the mechanism of EBV-induced NPC and the latest advances in targeted EBV vaccines for prophylaxis and treatment. This review explores the intricate molecular mechanisms by which EBV contributes to NPC pathogenesis, highlighting viral latency, genetic and epigenetic alterations, and immune evasion strategies. It emphasizes the pivotal role of key viral proteins, including EBNA1, LMP1, and LMP2A, in carcinogenesis. Subsequently, the discussion shifts towards the development of targeted EBV vaccines, including preventive vaccines aimed at preventing primary EBV infection and therapeutic vaccines aimed at treating diagnosed EBV-related NPC. The review underscores the challenges and future directions in the field, stressing the importance of developing innovative vaccine strategies and combination therapies to improve efficacy. This review synthesizes current insights into the molecular mechanisms of EBV-induced NPC and the development of EBV-targeted vaccines, highlighting the potential use of mRNA vaccines for NPC treatment.
Collapse
Affiliation(s)
- Weiwei Zhang
- Department of Oncology, Cancer Prevention and Treatment Institute of Chengdu, Chengdu Fifth People's Hospital/The Second Clinical Medical College, Affiliated Fifth People's Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 611130, China
| | - Chuang Wang
- Chengdu Yunce Medical Biotechnology Co., Ltd., Chengdu 611135, China
| | - Yousheng Meng
- Department of Oncology, Cancer Prevention and Treatment Institute of Chengdu, Chengdu Fifth People's Hospital/The Second Clinical Medical College, Affiliated Fifth People's Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 611130, China
| | - Lang He
- Department of Oncology, Cancer Prevention and Treatment Institute of Chengdu, Chengdu Fifth People's Hospital/The Second Clinical Medical College, Affiliated Fifth People's Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 611130, China
| | - Mingqing Dong
- Division of Pulmonary Medicine, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou Key Laboratory of Interdiscipline and Translational Medicine, Wenzhou Key Laboratory of Heart and Lung, Wenzhou 325000, China
| |
Collapse
|
5
|
Ding S, Alexander E, Liang H, Kulchar RJ, Singh R, Herzog RW, Daniell H, Leong KW. Synthetic and Biogenic Materials for Oral Delivery of Biologics: From Bench to Bedside. Chem Rev 2025; 125:4009-4068. [PMID: 40168474 DOI: 10.1021/acs.chemrev.4c00482] [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: 04/03/2025]
Abstract
The development of nucleic acid and protein drugs for oral delivery has lagged behind their production for conventional nonoral routes. Over the past decade, the evolution of DNA- and RNA-based technologies combined with the innovation of state-of-the-art delivery vehicles for nucleic acids has brought rapid advancements to the biopharmaceutical field. Nucleic acid therapies have the potential to achieve long-lasting effects, or even cures, by inhibiting or editing genes, which is not possible with conventional small-molecule drugs. However, challenges and limitations must be addressed before these therapies can provide cures for chronic conditions and rare diseases, rather than only offering temporary relief. Nucleic acids and proteins face premature degradation in the acidic, enzyme-rich stomach environment and are rapidly cleared by the liver. To overcome these challenges, various delivery vehicles have been developed to transport therapeutic compounds to the intestines, where the active compounds are released and gut microbiota and mucosal immune system also play an important role. This review provides a comprehensive overview of the promises and pitfalls associated with the oral route of administration of biologics, current delivery systems, applications of orally delivered therapeutics, and the challenges and considerations for translation of nucleic acid and protein therapeutics into clinical practice.
Collapse
Affiliation(s)
- Suwan Ding
- Department of Biomedical Engineering, Columbia University, 500 West 120th Street, New York, New York 10027, United States
| | - Elena Alexander
- Department of Biomedical Engineering, Columbia University, 500 West 120th Street, New York, New York 10027, United States
| | - Huiyi Liang
- Department of Biomedical Engineering, Columbia University, 500 West 120th Street, New York, New York 10027, United States
| | - Rachel J Kulchar
- Department of Basic and Translational Sciences, School of Dental Medicine, University of Pennsylvania, 240 South 40th Street, Philadelphia, Pennsylvania 19104, United States
| | - Rahul Singh
- Department of Basic and Translational Sciences, School of Dental Medicine, University of Pennsylvania, 240 South 40th Street, Philadelphia, Pennsylvania 19104, United States
| | - Roland W Herzog
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana 46202, United States
| | - Henry Daniell
- Department of Basic and Translational Sciences, School of Dental Medicine, University of Pennsylvania, 240 South 40th Street, Philadelphia, Pennsylvania 19104, United States
| | - Kam W Leong
- Department of Biomedical Engineering, Columbia University, 500 West 120th Street, New York, New York 10027, United States
| |
Collapse
|
6
|
Posa A. Spike protein-related proteinopathies: A focus on the neurological side of spikeopathies. Ann Anat 2025; 260:152662. [PMID: 40254264 DOI: 10.1016/j.aanat.2025.152662] [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: 02/24/2025] [Revised: 04/07/2025] [Accepted: 04/09/2025] [Indexed: 04/22/2025]
Abstract
BACKGROUND The spike protein (SP) is an outward-projecting transmembrane glycoprotein on viral surfaces. SARS-CoV-2 (Severe Acute Respiratory Syndrome Coronavirus 2), responsible for COVID-19 (Coronavirus Disease 2019), uses SP to infect cells that express angiotensin converting enzyme 2 (ACE2) on their membrane. Remarkably, SP has the ability to cross the blood-brain barrier (BBB) into the brain and cause cerebral damage through various pathomechanisms. To combat the COVID-19 pandemic, novel gene-based products have been used worldwide to induce human body cells to produce SP to stimulate the immune system. This artificial SP also has a harmful effect on the human nervous system. STUDY DESIGN Narrative review. OBJECTIVE This narrative review presents the crucial role of SP in neurological complaints after SARS-CoV-2 infection, but also of SP derived from novel gene-based anti-SARS-CoV-2 products (ASP). METHODS Literature searches using broad terms such as "SARS-CoV-2", "spike protein", "COVID-19", "COVID-19 pandemic", "vaccines", "COVID-19 vaccines", "post-vaccination syndrome", "post-COVID-19 vaccination syndrome" and "proteinopathy" were performed using PubMed. Google Scholar was used to search for topic-specific full-text keywords. CONCLUSIONS The toxic properties of SP presented in this review provide a good explanation for many of the neurological symptoms following SARS-CoV-2 infection and after injection of SP-producing ASP. Both SP entities (from infection and injection) interfere, among others, with ACE2 and act on different cells, tissues and organs. Both SPs are able to cross the BBB and can trigger acute and chronic neurological complaints. Such SP-associated pathologies (spikeopathies) are further neurological proteinopathies with thrombogenic, neurotoxic, neuroinflammatory and neurodegenerative potential for the human nervous system, particularly the central nervous system. The potential neurotoxicity of SP from ASP needs to be critically examined, as ASPs have been administered to millions of people worldwide.
Collapse
Affiliation(s)
- Andreas Posa
- University Clinics and Outpatient Clinics for Radiology, Neuroradiology and Neurology, Martin Luther University Halle-Wittenberg, Ernst-Grube-Straße 40, Halle 06120, Germany.
| |
Collapse
|
7
|
Su WC, Lieu R, Fu Y, Kempen T, Yu Z, Zhang K, Chen T, Fan Y. A platform method for simultaneous quantification of lipid and nucleic acid components in lipid nanoparticles. J Chromatogr A 2025; 1746:465788. [PMID: 39987694 DOI: 10.1016/j.chroma.2025.465788] [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: 12/21/2024] [Revised: 02/14/2025] [Accepted: 02/16/2025] [Indexed: 02/25/2025]
Abstract
Nucleic acid-based medicines have achieved significant advancements in recent years, with lipid nanoparticles (LNPs) being a pivotal platform for their delivery. However, the complexity of LNP presents significant challenges, requiring analytical methods to identify and quantify individual components to guide formulation development and ensure quality and safety. Current approaches often perform nucleic acid and lipid analysis separately and focus on a single type of formulation, highlighting the need for a simple platform method that can be applied to diverse formulations. We present a platform ion-pair reversed-phase HPLC method with UV and charged aerosol detection (CAD) to simultaneously separate and quantify lipid and nucleic acid components in LNPs. The method separated and quantified 12 lipid species and three types of nucleic acids (antisense oligonucleotide, single-guide RNA, and mRNA), covering a broad range of therapeutic cargoes. Notably, this can be achieved for the first time by one HPLC run with one-step facile sample preparation. Specifically, we used a simple buffer containing Triton and heparin to enable the single-step, simultaneous extraction of both nucleic acid and lipid components from LNPs, achieving quantification recoveries of 90-110 %. We further applied this method and addressed process and quality control challenges of LNPs, including the recovery rate of individual LNP components after purification and simultaneous quantification of co-loaded, different nucleic acid species for potential gene editing applications. This new platform method offers a robust and widely applicable tool to assess the quality of lipid-based nucleic acid therapies.
Collapse
Affiliation(s)
- Wan-Chih Su
- Synthetic Molecule Pharmaceutical Sciences, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA.
| | - Raymond Lieu
- Synthetic Molecule Pharmaceutical Sciences, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Yige Fu
- Synthetic Molecule Pharmaceutical Sciences, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Trevor Kempen
- Synthetic Molecule Pharmaceutical Sciences, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Zhixin Yu
- Synthetic Molecule Pharmaceutical Sciences, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Kelly Zhang
- Synthetic Molecule Pharmaceutical Sciences, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Tao Chen
- Synthetic Molecule Pharmaceutical Sciences, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA.
| | - Yuchen Fan
- Synthetic Molecule Pharmaceutical Sciences, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA.
| |
Collapse
|
8
|
Suri A, Satani S, Dobrovolny HM. Analyzing Differences in Viral Dynamics Between Vaccinated and Unvaccinated RSV Patients. EPIDEMIOLOGIA 2025; 6:16. [PMID: 40265347 PMCID: PMC12015914 DOI: 10.3390/epidemiologia6020016] [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: 01/24/2025] [Revised: 03/15/2025] [Accepted: 03/19/2025] [Indexed: 04/24/2025] Open
Abstract
Background: Respiratory syncytial virus (RSV) is a common respiratory virus that can cause serious illness in infants and the elderly. Vaccines for RSV have recently been introduced and have been shown to reduce the severity of the disease. However, there has been limited examination of how viral dynamics differ between vaccinated and unvaccinated individuals. Methods: Here, we use data from the MVA-BN-RSV Phase II vaccine study to quantify the dynamical differences between vaccinated and unvaccinated patients challenged with RSV. We use an ordinary differential equation model of within host viral dynamics to fit viral load data. Results: We find statistically significant differences in viral clearance rate and basic reproduction number. We also find that vaccinated patients experience a higher response variance than the placebo group. Conclusions: While the differences in viral clearance and basic reproduction number are promising, the high variability in response to the vaccine could leave many vaccinated patients without adequate protection.
Collapse
Affiliation(s)
| | | | - Hana M. Dobrovolny
- Department of Physics & Astronomy, Texas Christian University, Fort Worth, TX 76129, USA
| |
Collapse
|
9
|
Palma M. Advancing Breast Cancer Treatment: The Role of Immunotherapy and Cancer Vaccines in Overcoming Therapeutic Challenges. Vaccines (Basel) 2025; 13:344. [PMID: 40333213 PMCID: PMC12030785 DOI: 10.3390/vaccines13040344] [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: 02/11/2025] [Revised: 03/20/2025] [Accepted: 03/21/2025] [Indexed: 05/09/2025] Open
Abstract
Breast cancer (BC) remains a significant global health challenge due to its complex biology, which complicates both diagnosis and treatment. Immunotherapy and cancer vaccines have emerged as promising alternatives, harnessing the body's immune system to precisely target and eliminate cancer cells. However, several key factors influence the selection and effectiveness of these therapies, including BC subtype, tumor mutational burden (TMB), tumor-infiltrating lymphocytes (TILs), PD-L1 expression, HER2 resistance, and the tumor microenvironment (TME). BC subtypes play a critical role in shaping treatment responses. Triple-negative breast cancer (TNBC) exhibits the highest sensitivity to immunotherapy, while HER2-positive and hormone receptor-positive (HR+) subtypes often require combination strategies for optimal outcomes. High TMB enhances immune responses by generating neoantigens, making tumors more susceptible to immune checkpoint inhibitors (ICIs); whereas, low TMB may indicate resistance. Similarly, elevated TIL levels are associated with better immunotherapy efficacy, while PD-L1 expression serves as a key predictor of checkpoint inhibitor success. Meanwhile, HER2 resistance and an immunosuppressive TME contribute to immune evasion, highlighting the need for multi-faceted treatment approaches. Current breast cancer immunotherapies encompass a range of targeted treatments. HER2-directed therapies, such as trastuzumab and pertuzumab, block HER2 dimerization and enhance antibody-dependent cellular cytotoxicity (ADCC), while small-molecule inhibitors, like lapatinib and tucatinib, suppress HER2 signaling to curb tumor growth. Antibody-drug conjugates (ADCs) improve tumor targeting by coupling monoclonal antibodies with cytotoxic agents, minimizing off-target effects. Meanwhile, ICIs, including pembrolizumab, restore T-cell function, and CAR-macrophage (CAR-M) therapy leverages macrophages to reshape the TME and overcome immunotherapy resistance. While immunotherapy, particularly in TNBC, has demonstrated promise by eliciting durable immune responses, its efficacy varies across subtypes. Challenges such as immune-related adverse events, resistance mechanisms, high costs, and delayed responses remain barriers to widespread success. Breast cancer vaccines-including protein-based, whole-cell, mRNA, dendritic cell, and epitope-based vaccines-aim to stimulate tumor-specific immunity. Though clinical success has been limited, ongoing research is refining vaccine formulations, integrating combination therapies, and identifying biomarkers for improved patient stratification. Future advancements in BC treatment will depend on optimizing immunotherapy through biomarker-driven approaches, addressing tumor heterogeneity, and developing innovative combination therapies to overcome resistance. By leveraging these strategies, researchers aim to enhance treatment efficacy and ultimately improve patient outcomes.
Collapse
Affiliation(s)
- Marco Palma
- Institute for Globally Distributed Open Research and Education (IGDORE), 03181 Torrevieja, Spain
| |
Collapse
|
10
|
Kayesh MEH, Kohara M, Tsukiyama-Kohara K. Toll-like receptor response to Zika virus infection: progress toward infection control. NPJ VIRUSES 2025; 3:20. [PMID: 40295746 PMCID: PMC11906774 DOI: 10.1038/s44298-025-00102-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/27/2024] [Accepted: 02/19/2025] [Indexed: 04/30/2025]
Abstract
Infection with the Zika virus (ZIKV) poses a threat to human health. An improved understanding of the host Toll-like receptor response, disease onset, and viral clearance in vivo and in vitro may lead to the development of therapeutic or prophylactic interventions against viral infections. Currently, no clinically approved ZIKV vaccine is available, highlighting the need for its development. In this study, we discuss the progress in the Zika vaccine, including advances in the use of Toll-like receptor agonists as vaccine adjuvants to enhance vaccine efficacy.
Collapse
Affiliation(s)
- Mohammad Enamul Hoque Kayesh
- Department of Microbiology and Public Health, Faculty of Animal Science and Veterinary Medicine, Patuakhali Science and Technology University, Barishal, Bangladesh.
| | - Michinori Kohara
- Department of Microbiology and Cell Biology, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Kyoko Tsukiyama-Kohara
- Transboundary Animal Diseases Center, Joint Faculty of Veterinary Medicine, Kagoshima University, Kagoshima, Japan.
| |
Collapse
|
11
|
Ao D, Peng D, He C, Ye C, Hong W, Huang X, Lu Y, Shi J, Zhang Y, Liu J, Wei X, Wei Y. A promising mRNA vaccine derived from the JN.1 spike protein confers protective immunity against multiple emerged Omicron variants. MOLECULAR BIOMEDICINE 2025; 6:13. [PMID: 40035925 PMCID: PMC11880457 DOI: 10.1186/s43556-025-00258-7] [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: 12/06/2024] [Revised: 02/17/2025] [Accepted: 02/18/2025] [Indexed: 03/06/2025] Open
Abstract
Despite the declared end of the COVID-19 pandemic, SARS-CoV-2 continues to evolve, with emerging JN.1-derived subvariants (e.g., KP.2, KP.3) compromising the efficacy of current XBB.1.5-based vaccines. To address this, we developed an mRNA vaccine encoding the full-length spike protein of JN.1, incorporating GSAS and 2P mutations and encapsulated in lipid nanoparticles (LNPs). The JN.1-mRNA vaccine elicited robust humoral and cellular immune responses in mice, including high JN.1-specific IgG titers, cross-neutralizing antibodies, and increased T follicular helper (Tfh) cells, germinal center (GC) B cells, and T cell cytokines. Importantly, immunity persisted for up to six months and induced RBD-specific long-lived plasma cells. We also compared the immune responses induced by homologous and heterologous vaccination regimens, and our results demonstrated that the heterologous regimen-combining JN.1-mRNA with a recombinant protein vaccine (RBDJN.1-HR)-induced stronger responses. These findings highlight the JN.1-mRNA vaccine constitutes an effective prophylactic approach against JN.1-related variants, as it induces potent neutralizing antibody responses across all tested lineages. This enhanced immunogenicity is expected to significantly reduce hospitalization rates and mitigate post-COVID complications associated with JN.1 and KP.3 infections. This study emphasizes the need for timely vaccine updates and the adaptability of mRNA vaccines in addressing emerging pathogens, providing a framework for combating future infectious diseases. Collectively, these results offer critical insights for vaccine design and public health strategies in response to emerging SARS-CoV-2 variants.
Collapse
Affiliation(s)
- Danyi Ao
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041, Sichuan, China
| | - Dandan Peng
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041, Sichuan, China
| | - Cai He
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041, Sichuan, China
| | - Chunjun Ye
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041, Sichuan, China
| | - Weiqi Hong
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041, Sichuan, China
| | - Xiya Huang
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041, Sichuan, China
| | - Yishan Lu
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041, Sichuan, China
| | - Jie Shi
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041, Sichuan, China
| | - Yu Zhang
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041, Sichuan, China
| | - Jian Liu
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041, Sichuan, China
| | - Xiawei Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041, Sichuan, China.
- WestVac Biopharma Co. Ltd., Chengdu, China.
| | - Yuquan Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
12
|
Adelusi TI, Ogunlana AT, Oyewole MP, Ojo TO, Olaoba OT, Oladipo EK, Akash S, Ibenmoussa S, Bourhia M, Jardan YAB, Sitotaw B. Designing of an innovative conserved multiepitope subunit vaccine targeting SARS-CoV-2 glycoprotein and nucleoprotein through immunoinformatic. Sci Rep 2025; 15:2563. [PMID: 39833186 PMCID: PMC11747174 DOI: 10.1038/s41598-024-72495-9] [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: 11/23/2023] [Accepted: 09/09/2024] [Indexed: 01/22/2025] Open
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has imposed substantial challenges on our society due to the COVID-19 pandemic. This virus relies heavily on its surface glycoprotein (S-glycoprotein) to facilitate attachment, fusion, and entry into host cells. While the nucleoprotein (N) in the ribonucleoprotein core binds to the viral RNA genome. Therefore, our objective is to develop a novel vaccine candidate targeting the dominant T-cell and B-cell epitopes of the immune system. On the S-glycoprotein and nucleoprotein. Employing an immunoinformatic approach, we constructed a vaccine candidate with 13 highly antigenic B-cell epitopes, 19 HTL antigenic epitopes, and 18 CTL epitopes following a rigorous assessment. The multi-epitope construct successfully passed three-fold toxicity, allergenicity, and antigenicity tests, affirming its non-toxic, non-allergenic, and antigenic nature. This demonstrates the potentiality of the vaccine design to trigger an immunological response. Furthermore, the vaccine-ACE-2 receptor complex was tested, confirming its ability to interact with ACE-2's core pocket and induce an immunological response. Additionally, the vaccine's binding prowess for human toll-like receptors (TLR) (1, 3, 4, and 8) was investigated. According to the Ramachandran plot, 77.46% of the construct's amino acid residues fall within a favorable zone, establishing it as a viable vaccine candidate.
Collapse
Affiliation(s)
- Temitope Isaac Adelusi
- Computational Biology and Drug Discovery Laboratory, Department of Biochemistry, Ladoke Akintola University of Technology, Ogbomoso, Oyo State, Nigeria.
- Department of Surgery, School of Medicine, University of Connecticut Health, Farmington Ave, Connecticut, 06030, United States of America.
- Genomics Unit, Helix Biogen Institute, Ogbomoso, Oyo State, Nigeria.
| | - Abdeen Tunde Ogunlana
- Computational Biology and Drug Discovery Laboratory, Department of Biochemistry, Ladoke Akintola University of Technology, Ogbomoso, Oyo State, Nigeria
| | - Moyosoluwa Precious Oyewole
- Computational Biology and Drug Discovery Laboratory, Department of Biochemistry, Ladoke Akintola University of Technology, Ogbomoso, Oyo State, Nigeria
| | - Taiwo Ooreoluwa Ojo
- Computational Biology and Drug Discovery Laboratory, Department of Biochemistry, Ladoke Akintola University of Technology, Ogbomoso, Oyo State, Nigeria
- Genomics Unit, Helix Biogen Institute, Ogbomoso, Oyo State, Nigeria
| | - Olamide Tosin Olaoba
- Department of Surgery, School of Medicine, University of Connecticut Health, Farmington Ave, Connecticut, 06030, United States of America
| | - Elijah Kolawole Oladipo
- Genomics Unit, Helix Biogen Institute, Ogbomoso, Oyo State, Nigeria
- Department of Microbiology, Laboratory of Molecular Biology, Immunology and Bioinformatics, Adeleke University, Osun State, Nigeria
| | - Shopnil Akash
- Computational Biology Research Laboratory, Department of Pharmacy, Daffodil International University, Dhaka, Bangladesh
| | - Samir Ibenmoussa
- Laboratory of Therapeutic and Organic Chemistry, Faculty of Pharmacy, University of Montpellier, 34000, Montpellier, France
| | - Mohammed Bourhia
- Laboratory of Biotechnology and Natural Resources Valorization, Faculty of Sciences, Ibn Zohr University, 80060, Agadir, Morocco
| | - Yousef A Bin Jardan
- Department of Pharmaceutics, College of Pharmacy, King Saud University, P.O. Box 11451, Riyadh, Saudi Arabia
| | - Baye Sitotaw
- Department of Biology, Bahir Dar University, P.O.Box 79, Bahir Dar, Ethiopia.
| |
Collapse
|
13
|
Fu Q, Zhao X, Hu J, Jiao Y, Yan Y, Pan X, Wang X, Jiao F. mRNA vaccines in the context of cancer treatment: from concept to application. J Transl Med 2025; 23:12. [PMID: 39762875 PMCID: PMC11702060 DOI: 10.1186/s12967-024-06033-6] [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: 07/29/2024] [Accepted: 12/24/2024] [Indexed: 01/11/2025] Open
Abstract
Immuno-oncology has witnessed remarkable advancements in the past decade, revolutionizing the landscape of cancer therapeutics in an encouraging manner. Among the diverse immunotherapy strategies, mRNA vaccines have ushered in a new era for the therapeutic management of malignant diseases, primarily due to their impressive impact on the COVID-19 pandemic. In this comprehensive review, we offer a systematic overview of mRNA vaccines, focusing on the optimization of structural design, the crucial role of delivery materials, and the administration route. Additionally, we summarize preclinical studies and clinical trials to provide valuable insights into the current status of mRNA vaccines in cancer treatment. Furthermore, we delve into a systematic discussion on the significant challenges facing the current development of mRNA tumor vaccines. These challenges encompass both intrinsic and external factors that are closely intertwined with the successful application of this innovative approach. To pave the way for a more promising future in cancer treatments, a deeper understanding of immunological mechanisms, an increasing number of high-quality clinical trials, and a well-established manufacturing platform are crucial. Collaborative efforts between scientists, clinicians, and industry engineers are essential to achieving these goals.
Collapse
Affiliation(s)
- Qiang Fu
- School of Pharmacology, Institute of Aging Medicine, Binzhou Medical University, Yantai, 264003, P. R. China
| | - Xiaoming Zhao
- Center of Physical Examination, Binzhou Medical University Affiliated 970 Hospital of the PLA Joint Logistic Support Force, Yantai, 264002, P. R. China
| | - Jinxia Hu
- Department of Biochemistry and Molecular Biology, Binzhou Medical University, 346 Guanhai Road, Yantai, 264003, P. R. China
| | - Yang Jiao
- Department of Biomedical Engineering, Faculty of Engineering, The Hong Kong Polytechnic University, Hong Kong, 999077, P. R. China
| | - Yunfei Yan
- Department of Biochemistry and Molecular Biology, Binzhou Medical University, 346 Guanhai Road, Yantai, 264003, P. R. China
| | - Xuchen Pan
- Department of Clinical Laboratory & Health Service Training, Binzhou Medical University Affiliated 970 Hospital of the PLA Joint Logistic Support Force, Yantai, 264002, P. R. China
| | - Xin Wang
- Department of Clinical Laboratory & Health Service Training, Binzhou Medical University Affiliated 970 Hospital of the PLA Joint Logistic Support Force, Yantai, 264002, P. R. China.
| | - Fei Jiao
- Department of Biochemistry and Molecular Biology, Binzhou Medical University, 346 Guanhai Road, Yantai, 264003, P. R. China.
| |
Collapse
|
14
|
Sira EMJS, Fajardo LE, Banico EC, Odchimar NMO, Orosco FL. Design of a Multiepitope Pan-Proteomic mRNA Vaccine Construct Against African Swine Fever Virus: A Reverse Vaccinology Approach. Vet Med Int 2025; 2025:2638167. [PMID: 39803351 PMCID: PMC11724734 DOI: 10.1155/vmi/2638167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 12/13/2024] [Indexed: 01/16/2025] Open
Abstract
African swine fever (ASF), caused by African swine fever virus (ASFV), is a highly contagious disease with devastating effects on the global pig industry. This warrants the development of effective control strategies, such as vaccines. However, previously developed inactivated vaccines have proven ineffective, while live-attenuated vaccines carry inherent safety risks. The use of mRNA vaccines eliminates these risks offering a safe, cost-effective, and efficient vaccine strategy against ASFV. In this study, a reverse vaccinology approach was used to design a multiepitope pan-proteomic mRNA vaccine against ASFV. Various bioinformatics tools were employed to predict epitopes for cytotoxic T lymphocytes, helper T lymphocytes, and B lymphocytes. A 50S ribosomal L7/L12 protein adjuvant, 5' cap, poly(A) tail, signal peptide, and MHC-I-targeting domain were incorporated into the design using appropriate linkers to increase immunogenicity, stability, and recognition efficiency. The physicochemical properties of the final construct were evaluated, and docking analyses were done with Toll-like receptors (TLRs) 3, 4, and 7 to evaluate binding affinity. A molecular dynamics simulation was then performed to determine binding stability, while immune simulations evaluated host's immune response. Based on 100 ASFV proteomes, six epitopes that induce cytotoxic T-cell responses, five epitopes that induce helper T-cell responses, and four epitopes that induce antibody production were predicted. The designed vaccine construct was found to be nonallergenic, antigenic, and stable when bound to TLR4 while the binding pocket analyses of the vaccine construct to TLR3 and TLR7 indicate high translation efficiency. Immune simulations demonstrated successful induction of immune responses and generation of antigen-specific memory cells. In conclusion, this study introduces an mRNA vaccine construct as a potential disease control strategy against ASF for in vitro confirmation.
Collapse
Affiliation(s)
- Ella Mae Joy S. Sira
- Department of Science and Technology, Virology and Vaccine Research Program, Industrial Technology Development Institute, Bicutan, Taguig 1634, Philippines
| | - Lauren Emily Fajardo
- Department of Science and Technology, Virology and Vaccine Research Program, Industrial Technology Development Institute, Bicutan, Taguig 1634, Philippines
| | - Edward C. Banico
- Department of Science and Technology, Virology and Vaccine Research Program, Industrial Technology Development Institute, Bicutan, Taguig 1634, Philippines
| | - Nyzar Mabeth O. Odchimar
- Department of Science and Technology, Virology and Vaccine Research Program, Industrial Technology Development Institute, Bicutan, Taguig 1634, Philippines
| | - Fredmoore L. Orosco
- Department of Science and Technology, Virology and Vaccine Research Program, Industrial Technology Development Institute, Bicutan, Taguig 1634, Philippines
- Department of Biology, College of Arts and Sciences, University of the Philippines Manila, Manila 1000, Philippines
- Department of Science and Technology, S&T Fellows Program, Bicutan, Taguig 1634, Philippines
| |
Collapse
|
15
|
Yu H, Wen Y, Yu W, Lu L, Yang Y, Liu C, Hu Z, Fang Z, Huang S. Optimized circular RNA vaccines for superior cancer immunotherapy. Theranostics 2025; 15:1420-1438. [PMID: 39816687 PMCID: PMC11729565 DOI: 10.7150/thno.104698] [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: 10/05/2024] [Accepted: 12/10/2024] [Indexed: 01/18/2025] Open
Abstract
Rationale: Circular RNA (circRNA) has gained attention as a promising platform for mRNA vaccines due to its stability, sustained protein expression, and intrinsic immunostimulatory properties. This study aimed to design and optimize a circRNA cancer vaccine platform by screening for efficient internal ribosome entry sites (IRES) and enhancing circRNA translation efficiency for improved cancer immunotherapy. Methods: We screened 29 IRES elements to identify the most efficient one for immune cell translation, ultimately discovering the Enterovirus A (EV-A) IRES. Using SHAPE-MaP technology, we analyzed the secondary structure of circRNA and introduced targeted mutations and deletions to optimize translation efficiency. Additionally, we investigated the regulatory roles of spacer sequences and microRNA recognition sites in circRNA design and examined the mechanisms behind IRES-mediated translation initiation. Results: The EV-A IRES was identified as the most efficient for immune cell translation. Structural modifications and optimization of spacer sequences enhanced the translation efficiency of circRNA. Comparative studies demonstrated that circRNA vaccines induced stronger T cell immune responses and exhibited superior tumor prevention and therapeutic efficacy compared to traditional linear mRNA vaccines. Conclusion: The optimized tumor antigen circRNA vaccine platform offers a stable, efficient alternative to conventional mRNA vaccines for cancer immunotherapy, with enhanced immune responses and improved therapeutic outcomes. This work lays the foundation for developing circRNA-based vaccines as a novel strategy for cancer treatment.
Collapse
Affiliation(s)
- Hongwu Yu
- Department of Integrative Oncology, Fudan University Shanghai Cancer Center, and Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Yifan Wen
- Department of Integrative Oncology, Fudan University Shanghai Cancer Center, and Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Wenqian Yu
- Department of Integrative Oncology, Fudan University Shanghai Cancer Center, and Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Liang Lu
- Department of Integrative Oncology, Fudan University Shanghai Cancer Center, and Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Yu Yang
- Department of Integrative Oncology, Fudan University Shanghai Cancer Center, and Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Chengye Liu
- Department of Integrative Oncology, Fudan University Shanghai Cancer Center, and Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Zhixiang Hu
- Department of Integrative Oncology, Fudan University Shanghai Cancer Center, and Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Zhuting Fang
- Department of Oncology and Vascular Interventional Therapy, Clinical Oncology School of Fujian Medical University, Fujian Key Laboratory of Translational Cancer Medicine, Fujian Cancer Hospital (Fujian Branch of Fudan University Shanghai Cancer Center), Fuzhou, 350014, China
- Department of Interventional Radiology, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou University Afliated Provincial Hospital, Fuzhou 350001, China
| | - Shenglin Huang
- Department of Integrative Oncology, Fudan University Shanghai Cancer Center, and Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| |
Collapse
|
16
|
Smith OA, Fujimoto B, Wong TAS, To A, Odo T, Ball A, Haun BK, Muramatsu H, Tam YK, Pardi N, Lehrer AT. Impact of Metabolic States on SARS-CoV-2 Vaccine Responses in Mouse Models of Obesity and Diabetes. COVID 2025; 5:2. [PMID: 40406709 PMCID: PMC12097603 DOI: 10.3390/covid5010002] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/25/2025]
Abstract
The emergence of SARS-CoV-2 has resulted in a significant impact on public health, particularly for individuals with underlying health conditions such as obesity and diabetes. While vaccination efforts have played a crucial role in reducing hospitalizations, it remains unclear whether the effectiveness of these vaccines varies among different population groups. In this study, we investigated the immune responses generated by various SARS-CoV-2 vaccine platforms in mouse models with obesity and diabetes, focusing on both cell-mediated and humoral immune responses. Our findings revealed diminished immune responses in diabetic and obese mice compared to healthy counterparts. After vaccination with adjuvanted subunit or mRNA lipid nanoparticle (LNP) vaccines, both humoral and cell-mediated responses were significantly reduced in diabetic mice. Obese mice also exhibited decreased immunogenicity, albeit to a lesser extent. However, it should be noted that mRNA vaccines demonstrated strong neutralizing responses across all metabolic states, while adjuvanted subunit vaccines elicited higher antibody avidity in mice with type 2 diabetes (T2D) and obesity compared to healthy mice. These results suggest that the impaired humoral and cell-mediated responses observed in altered metabolic states may be linked to chronic inflammation associated with obesity and suboptimal glycemic control in diabetes. Understanding the impact of these metabolic disturbances on vaccine immunogenicity is crucial for developing optimized vaccines that can effectively enhance immune responses and provide long-lasting protection against SARS-CoV-2, even in individuals with obesity and diabetes. By contributing these findings, we support efforts to improve vaccine efficacy in populations affected by metabolic disorders, advancing effective immunization against SARS-CoV-2.
Collapse
Affiliation(s)
- Olivia A. Smith
- Department of Tropical Medicine, Medical Microbiology, and Pharmacology, University of Hawaii Manoa, Honolulu, HI 96813, USA
| | - Brent Fujimoto
- Department of Tropical Medicine, Medical Microbiology, and Pharmacology, University of Hawaii Manoa, Honolulu, HI 96813, USA
| | - Teri Ann S. Wong
- Department of Tropical Medicine, Medical Microbiology, and Pharmacology, University of Hawaii Manoa, Honolulu, HI 96813, USA
| | - Albert To
- Department of Tropical Medicine, Medical Microbiology, and Pharmacology, University of Hawaii Manoa, Honolulu, HI 96813, USA
| | - Troy Odo
- Department of Tropical Medicine, Medical Microbiology, and Pharmacology, University of Hawaii Manoa, Honolulu, HI 96813, USA
| | - Aquena Ball
- Department of Tropical Medicine, Medical Microbiology, and Pharmacology, University of Hawaii Manoa, Honolulu, HI 96813, USA
| | - Brien K. Haun
- Department of Tropical Medicine, Medical Microbiology, and Pharmacology, University of Hawaii Manoa, Honolulu, HI 96813, USA
- Cell and Molecular Biology Graduate Program, University of Hawaii Manoa, Honolulu, HI 96813, USA
| | - Hiromi Muramatsu
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ying K Tam
- Acuitas Therapeutics, Vancouver, BC V6T 1Z3, Canada
| | - Norbert Pardi
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Axel T. Lehrer
- Department of Tropical Medicine, Medical Microbiology, and Pharmacology, University of Hawaii Manoa, Honolulu, HI 96813, USA
| |
Collapse
|
17
|
Rai S, Tripathi S. Comparative efficacy of leading COVID-19 vaccines: A network meta-analysis. Indian J Med Res 2025; 161:9-20. [PMID: 40036106 PMCID: PMC11878698 DOI: 10.25259/ijmr_750_2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Accepted: 01/10/2025] [Indexed: 03/06/2025] Open
Abstract
In the fight against the COVID-19 virus, various vaccines using different technologies such as mRNA, viral vectors, protein subunits, and inactivated whole viruses have become primary defence strategies. This study aims to compare their effectiveness in controlling the spread of the pandemic. Using the comprehensive resources from three major databases-PubMed, EMBASE, and the Cochrane Library-we conducted an extensive literature review up to April 30, 2023. By employing a frequentist network meta-analysis, we analysed both direct and indirect estimates of vaccine efficacy, providing a clear comparison of the leading candidates in the global fight against COVID-19. Fifteen vaccines from 26 articles were used in our network meta-analysis. The statistically significant direct estimates were obtained by Spikevax [VE: 93.29 (91.31, 95.27); P<0.05], Pfizer BioNTech [VE: 92.07 (90.03, 94.12); P<0.05], Sputnik [VE: 91.60 (85.60, 97.60); P<0.05], Novavax [VE: 88.99 (83.55, 94.42); P<0.05], Sinovac [VE: 83.50 (65.40, 101.60); P<0.05], Covifenz [VE: 77.27 (68.48, 86.06); P<0.05], Zifivax [VE: 75.94 (70.86, 81.02); P<0.05], Covishield [VE: 72.34 (67.12, 77.56); P<0.05], S-Trimer [VE: 71.61 (56.23, 86.98); P<0.05], Covaxin [VE: 70.81 (65.33, 76.29); P<0.05], Soberna [VE: 69.70 (56.50, 82.90); P<0.05], Zydus Cadila [VE: 66.60 (47.60, 85.60); P<0.05], CVnCoV [VE: 63.70 (52.20, 75.20); P<0.05], Convidecia [VE: 57.50 (39.70, 75.30); P <0.05], and Jcovden [VE : 52.42 (47.28, 57.57); P<0.05]. Spikevax emerged triumphant with an unparalleled P score of 0.95, solidifying its status as a top ranking prevention tool against the COVID-19 in our investigation. Our analysis reveals a ranking of vaccine efficacy, with Spikevax emerging as the most effective, followed closely by Comirnaty, Sputnik, and others, collectively providing strong protection against the ongoing threat of COVID-19.
Collapse
Affiliation(s)
- Sanjay Rai
- Department of Centre of Community Medicine, All India Institute of Medical Sciences, New Delhi, India
| | - Shashank Tripathi
- Department of Biostatistics and Medical Informatics, University College of Medical Sciences and GTB Hospital, New Delhi, India
| |
Collapse
|
18
|
Shi R, Liu X, Wang Y, Pan M, Wang S, Shi L, Ni B. Long-term stability and immunogenicity of lipid nanoparticle COVID-19 mRNA vaccine is affected by particle size. Hum Vaccin Immunother 2024; 20:2342592. [PMID: 38714327 PMCID: PMC11085994 DOI: 10.1080/21645515.2024.2342592] [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] [Accepted: 04/09/2024] [Indexed: 05/09/2024] Open
Abstract
Messenger ribonucleic acid (mRNA) technology has been rapidly applied for the development of the COVID-19 vaccine. However, naked mRNA itself is inherently unstable. Lipid nanoparticles (LNPs) protect mRNAs from extracellular ribonucleases and facilitate mRNA trafficking. For mRNA vaccines, antigen-presenting cells utilize LNPs through uptake to elicit antigen-specific immunity. There are reports on the impact of various physical characteristics of LNPs, particularly those with sizes less than 200 nm, especially 50 to 150 nm, on the overall stability and protective efficacy of mRNA vaccines. To address this, a single change in the size of LNPs using the same mRNA stock solution was assessed for the physicochemical characterization of the resulting mRNA-LNPs vaccine, along with the evaluation of their protective efficacy. Particles of smaller sizes generally disperse more effectively in solutions, with minimized occurrence of particle precipitation and aggregation. Here, we demonstrate that the vaccine containing 80-100 nm mRNA-LNPs showed the best stability and protection at 4°C and -20°C. Furthermore, we can conclude that freezing the vaccine at -20°C is more appropriate for maintaining stability over the long term. This effort is poised to provide a scientific basis for improving the quality of ongoing mRNA vaccine endeavors and providing information on the development of novel products.
Collapse
Affiliation(s)
- Ruimeng Shi
- School of Pharmacy, Hebei Medical University, Shijiazhuang, PR China
| | - Xueli Liu
- Pharmacology Laboratory, Hebei Research Institute of Pharmaceutical and Medical Device Inspection, Shijiazhuang, PR China
| | - Yajuan Wang
- Research and Development Department, CSPC Pharmaceutical Group Co., Ltd., Shijiazhuang, PR China
| | - Meilu Pan
- Pharmacology Laboratory, Hebei Research Institute of Pharmaceutical and Medical Device Inspection, Shijiazhuang, PR China
| | - Shaoqin Wang
- School of Pharmacy, Hebei Medical University, Shijiazhuang, PR China
| | - Lin Shi
- Pharmacology Laboratory, Hebei Research Institute of Pharmaceutical and Medical Device Inspection, Shijiazhuang, PR China
| | - Beibei Ni
- Research and Development Department, CSPC Pharmaceutical Group Co., Ltd., Shijiazhuang, PR China
| |
Collapse
|
19
|
Shariati A, Khani P, Nasri F, Afkhami H, Khezrpour A, Kamrani S, Shariati F, Alavimanesh S, Modarressi MH. mRNA cancer vaccines from bench to bedside: a new era in cancer immunotherapy. Biomark Res 2024; 12:157. [PMID: 39696625 DOI: 10.1186/s40364-024-00692-9] [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: 09/10/2024] [Accepted: 11/15/2024] [Indexed: 12/20/2024] Open
Abstract
Harnessing the power of the immune system to target cancer cells is one of the most appealing approaches for cancer therapy. Among these immunotherapies, messenger ribonucleic acid (mRNA) cancer vaccines are worthy of consideration, as they have demonstrated promising results in clinical trials. These vaccines have proven to be safe and well-tolerated. They can be easily mass-produced in a relatively short time and induce a systemic immune response effective against both the primary tumor and metastases. Transcripts encoding immunomodulatory molecules can also be incorporated into the mRNA, enhancing its efficacy. On the other hand, there are some challenges associated with their application, including mRNA instability, insufficient uptake by immune cells, and intrinsic immunogenicity, which can block mRNA translation. Many innovations have been suggested to overcome these obstacles, including structural modification (such as 5' cap modification), optimizing delivery vehicles (especially dendritic cells (DCs) and nanoparticles), and using antigens that can enhance immunogenicity by circumventing tolerance mechanisms. A popular approach is to combine mRNA cancer vaccines with traditional and novel cancer treatments like chemotherapy, radiotherapy, and immune checkpoint blockade (ICB). They are most efficacious when combined with other therapies like ICBs. There is still a long way to go before these vaccines enter the standard of care for cancer patients, but with the incredible pace of development in this field, their clinical application will soon be witnessed. This review highlights the recent advances and challenges of mRNA cancer vaccines. Finally, some of the most prominent clinical applications of these vaccines will be reviewed.
Collapse
Affiliation(s)
- Alireza Shariati
- School of Medicine, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Pouria Khani
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Farzad Nasri
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Hamed Afkhami
- Cellular and Molecular Research Center, Qom University of Medical Sciences, Qom, Iran
- Nervous System Stem Cells Research Center, Semnan University of Medical Sciences, Semnan, Iran
- Department of Medical Microbiology, Faculty of Medicine, Shahed University, Tehran, Iran
| | - Arya Khezrpour
- School of Medicine, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Sina Kamrani
- Department of Orthopedic, Faculty of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Fatemeh Shariati
- Department of Genetics, North Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Sajad Alavimanesh
- Student Research Committee, Shahrekord University of Medical Sciences, Shahrekord, Iran.
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran.
| | - Mohammad Hossein Modarressi
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences (TUMS), Tehran, Iran.
| |
Collapse
|
20
|
Kafle U, Truong HQ, Nguyen CTG, Meng F. Development of Thermally Stable mRNA-LNP Delivery Systems: Current Progress and Future Prospects. Mol Pharm 2024; 21:5944-5959. [PMID: 39529245 DOI: 10.1021/acs.molpharmaceut.4c00826] [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] [Indexed: 11/16/2024]
Abstract
The success of mRNA-LNP-based COVID-19 vaccines opens a new era for mRNA-LNP-based therapy. This breakthrough is expected to catalyze the development of more mRNA-LNP-based medicines, not only for preventive vaccines but also for therapeutic purposes. Despite the promising outlook, there are fundamental challenges impeding the progress and widespread application of mRNA-LNP formulations. One of the significant challenges is their thermal instability, requiring these products to be stored at ultralow temperatures for long-term stability. The specific requirements present significant challenges for the storage, transportation, and distribution of mRNA-LNP formulations. To effectively prepare for future infectious disease outbreaks and broaden the application of mRNA-LNP-based therapies for other illnesses, improving the thermostability of mRNA-LNP formulations is critical. In this review, we discuss the potential factors contributing to the thermal instability of mRNA-LNP formulations and examine the roles of key components such as ionizable lipids, cholesterol, pH, buffers, and stabilizing agents like sugars in maintaining their thermal stability, with the goal of providing insights that can guide the future development of thermally stable mRNA-LNP formulations.
Collapse
Affiliation(s)
- Urmila Kafle
- Department of Biomedical and Nutritional Sciences, University of Massachusetts Lowell, 3 Solomont Way, Lowell, Massachusetts 01854, United States
| | - Hoang Quan Truong
- Department of Biomedical and Nutritional Sciences, University of Massachusetts Lowell, 3 Solomont Way, Lowell, Massachusetts 01854, United States
| | - Cao Thuy Giang Nguyen
- Department of Biomedical and Nutritional Sciences, University of Massachusetts Lowell, 3 Solomont Way, Lowell, Massachusetts 01854, United States
| | - Fanfei Meng
- Department of Biomedical and Nutritional Sciences, University of Massachusetts Lowell, 3 Solomont Way, Lowell, Massachusetts 01854, United States
| |
Collapse
|
21
|
Baig MMFA, Wong LY, Wu H. Development of mRNA nano-vaccines for COVID-19 prevention and its biochemical interactions with various disease conditions and age groups. J Drug Target 2024; 32:21-32. [PMID: 38010097 DOI: 10.1080/1061186x.2023.2288996] [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: 06/24/2023] [Accepted: 11/18/2023] [Indexed: 11/29/2023]
Abstract
This review has focused on the development of mRNA nano-vaccine and the biochemical interactions of anti-COVID-19 mRNA vaccines with various disease conditions and age groups. It studied five major groups of individuals with different disease conditions and ages, including allergic background, infarction background, adolescent, and adult (youngsters), pregnant women, and elderly. All five groups had been reported to have background-related adverse effects. Allergic background individuals were observed to have higher chances of experiencing allergic reactions and even anaphylaxis. Individuals with an infarction background had a higher risk of vaccine-induced diseases, e.g. pneumonitis and interstitial lung diseases. Pregnant women were seen to suffer from obstetric and gynecological adverse effects after receiving vaccinations. However, interestingly, the elderly individuals (> 65 years old) had experienced milder and less frequent adverse effects compared to the adolescent (<19 and >9 years old) and young adulthood (19-39 years old), or middle adulthood (40-59 years old) age groups, while middle to late adolescent (14-17 years old) was the riskiest age group to vaccine-induced cardiovascular manifestations.
Collapse
Affiliation(s)
- Mirza Muhammad Faran Ashraf Baig
- Department of Chemistry and the Hong Kong Branch of Chinese National Engineering Research Centre for Tissue Restoration, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Lok Yin Wong
- Department of Chemistry and the Hong Kong Branch of Chinese National Engineering Research Centre for Tissue Restoration, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Hongkai Wu
- Department of Chemistry and the Hong Kong Branch of Chinese National Engineering Research Centre for Tissue Restoration, The Hong Kong University of Science and Technology, Hong Kong, China
| |
Collapse
|
22
|
Freppel W, Silva LA, Stapleford KA, Herrero LJ. Pathogenicity and virulence of chikungunya virus. Virulence 2024; 15:2396484. [PMID: 39193780 PMCID: PMC11370967 DOI: 10.1080/21505594.2024.2396484] [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: 02/27/2024] [Revised: 08/09/2024] [Accepted: 08/14/2024] [Indexed: 08/29/2024] Open
Abstract
Chikungunya virus (CHIKV) is a mosquito-transmitted, RNA virus that causes an often-severe musculoskeletal illness characterized by fever, joint pain, and a range of debilitating symptoms. The virus has re-emerged as a global health threat in recent decades, spreading from its origin in Africa across Asia and the Americas, leading to widespread outbreaks impacting millions of people. Despite more than 50 years of research into the pathogenesis of CHIKV, there is still no curative treatment available. Current management of CHIKV infections primarily involves providing supportive care to alleviate symptoms and improve the patient's quality of life. Given the ongoing threat of CHIKV, there is an urgent need to better understand its pathogenesis. This understanding is crucial for deciphering the mechanisms underlying the disease and for developing effective strategies for both prevention and management. This review aims to provide a comprehensive overview of CHIKV and its pathogenesis, shedding light on the complex interactions of viral genetics, host factors, immune responses, and vector-related factors. By exploring these intricate connections, the review seeks to contribute to the knowledge base surrounding CHIKV, offering insights that may ultimately lead to more effective prevention and management strategies for this re-emerging global health threat.
Collapse
Affiliation(s)
- Wesley Freppel
- Institute for Biomedicine and Glycomics, Gold Coast Campus, Griffith University, Southport, Australia
| | - Laurie A. Silva
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Kenneth A. Stapleford
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY, USA
| | - Lara J. Herrero
- Institute for Biomedicine and Glycomics, Gold Coast Campus, Griffith University, Southport, Australia
| |
Collapse
|
23
|
Fardoost A, Karimi K, Govindaraju H, Jamali P, Javanmard M. Applications of microfluidics in mRNA vaccine development: A review. BIOMICROFLUIDICS 2024; 18:061502. [PMID: 39553921 PMCID: PMC11567697 DOI: 10.1063/5.0228447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Accepted: 10/30/2024] [Indexed: 11/19/2024]
Abstract
The transformative potential of microfluidics in the development of mRNA vaccines is explored in this review, highlighting its pivotal role in enhancing easy-to-use functionality, efficacy, and production efficiency. Moreover, we examine the innovative applications of microfluidics in biomedical research, including its contribution to the rapid and cost-effective synthesis of lipid nanoparticles for mRNA delivery and delve into the advantages of mRNA vaccines, such as targeted delivery and controlled expression. Furthermore, it outlines the future prospects of microfluidic devices, their cutting-edge examples in both research and industry, and the potential to revolutionize vaccine formulation and production. The integration of microfluidics with mRNA vaccine development represents a significant advancement in public health and disease prevention strategies.
Collapse
Affiliation(s)
- Ali Fardoost
- Department of Electrical and Computer Engineering, Rutgers, The State University of New Jersey, Piscataway, New Jersey 08854, USA
| | - Koosha Karimi
- Department of Electrical and Computer Engineering, Rutgers, The State University of New Jersey, Piscataway, New Jersey 08854, USA
| | - Harshitha Govindaraju
- Department of Electrical and Computer Engineering, Rutgers, The State University of New Jersey, Piscataway, New Jersey 08854, USA
| | - Pegah Jamali
- Department of Electrical and Computer Engineering, Rutgers, The State University of New Jersey, Piscataway, New Jersey 08854, USA
| | - Mehdi Javanmard
- Department of Electrical and Computer Engineering, Rutgers, The State University of New Jersey, Piscataway, New Jersey 08854, USA
| |
Collapse
|
24
|
Niazi SK, Magoola M. Advancing Therapeutic and Vaccine Proteins: Switching from Recombinant to Ribosomal Delivery-A Humanitarian Cause. Int J Mol Sci 2024; 25:12797. [PMID: 39684504 DOI: 10.3390/ijms252312797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 11/25/2024] [Accepted: 11/25/2024] [Indexed: 12/18/2024] Open
Abstract
Recombinant therapeutic and vaccine proteins have revolutionized healthcare, but there remain challenges, as many are awaiting development due to their slow development speed and high development cost. Cell-free in vivo ribosomes offer one choice, but they come with similar constraints. The validation of in vivo messenger RNA (mRNA) technology has been accomplished for COVID-19 vaccines. The bioreactors inside the body, the ribosomes, deliver these proteins at a small cost, since these are chemical products and do not require extensive analytical and regulatory exercises. In this study, we test and validate the final product. A smaller fraction of the recombinant protein cost is needed, removing both constraints. Although thousands of in vivo mRNA products are under development, their regulatory classification remains unresolved: do they qualify as chemical drugs, biological drug, or gene therapy items? These questions will soon be resolved. Additionally, how would the copies of approved in vivo mRNA protein products be brought in, and how would they be treated: as new drugs, generic drugs, or new biological drugs? Researchers are currently working to answer these questions. Regardless, these products' cost of goods (COGs) remains much smaller than that of ex vivo mRNA or recombinant products. This is necessary to meet the needs of the approximately 6.5 billion people around the world who do not have access to biological drugs; these products will indeed serve the dire needs of humanity. Given the minor cost of establishing the manufacturing of these products, it will also prove financially attractive to investors.
Collapse
Affiliation(s)
- Sarfaraz K Niazi
- College of Pharmacy, University of Illinois, Chicago, IL 60612, USA
| | | |
Collapse
|
25
|
O'Hare M, Miller WP, Arevalo-Alquichire S, Amarnani D, Apryani E, Perez-Corredor P, Marino C, Shu DY, Vanderleest TE, Muriel-Torres A, Gordon HB, Gunawan AL, Kaplan BA, Barake KW, Bejjani RP, Doan TH, Lin R, Delgado-Tirado S, Gonzalez-Buendia L, Rossin EJ, Zhao G, Eliott D, Weinl-Tenbruck C, Chevessier-Tünnesen F, Rejman J, Montrasio F, Kim LA, Arboleda-Velasquez JF. An mRNA-encoded dominant-negative inhibitor of transcription factor RUNX1 suppresses vitreoretinal disease in experimental models. Sci Transl Med 2024; 16:eadh0994. [PMID: 39602510 DOI: 10.1126/scitranslmed.adh0994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 02/06/2024] [Accepted: 11/06/2024] [Indexed: 11/29/2024]
Abstract
Messenger RNA (mRNA)-based therapies are a promising approach to medical treatment. Except for infectious diseases, no other disease has mRNA-based therapies available. The eye is an ideal model for mRNA therapeutic development because it requires limited dosing. Proliferative vitreoretinopathy (PVR) is a blinding condition caused by retinal detachment that now lacks available medical treatment, with surgery as the only treatment option. We previously implicated runt-related transcription factor-1 (RUNX1) as a driver of epithelial-to-mesenchymal transition (EMT) in PVR and as a critical mediator of aberrant ocular angiogenesis when up-regulated. On the basis of these findings, an mRNA was designed to express a dominant-negative inhibitor of RUNX1 (RUNX1-Trap). We show that RUNX1-Trap delivered in polymer-lipidoid complexes or lipid nanoparticles sequestered RUNX1 in the cytosol and strongly reduced proliferation in primary cell cultures established from fibrotic membranes derived from patients with PVR. We assessed the preclinical efficacy of intraocular delivery of mRNA-encoded RUNX1-Trap in a rabbit model of PVR and in a laser-induced mouse model of aberrant angiogenesis often used to study wet age-related macular degeneration. mRNA-encoded RUNX1-Trap suppressed ocular pathology, measured as pathological scores in the rabbit PVR model and leakage and lesion size in the laser-induced choroidal neovascularization mouse model. mRNA-encoded RUNX1-Trap also strongly reduced proliferation in a human ex vivo explant model of PVR. These data demonstrate the therapeutic potential of mRNA-encoded therapeutic molecules with dominant-negative properties, highlighting the potential of mRNA-based therapies beyond standard gene supplementation approaches.
Collapse
Affiliation(s)
- Michael O'Hare
- Schepens Eye Research Institute of Mass Eye and Ear and the Department of Ophthalmology at Harvard Medical School, Boston, MA 02114, USA
- Mass Eye and Ear and the Department of Ophthalmology at Harvard Medical School, Boston, MA 02114, USA
| | - William P Miller
- Schepens Eye Research Institute of Mass Eye and Ear and the Department of Ophthalmology at Harvard Medical School, Boston, MA 02114, USA
- Mass Eye and Ear and the Department of Ophthalmology at Harvard Medical School, Boston, MA 02114, USA
| | - Said Arevalo-Alquichire
- Schepens Eye Research Institute of Mass Eye and Ear and the Department of Ophthalmology at Harvard Medical School, Boston, MA 02114, USA
- Mass Eye and Ear and the Department of Ophthalmology at Harvard Medical School, Boston, MA 02114, USA
| | - Dhanesh Amarnani
- Schepens Eye Research Institute of Mass Eye and Ear and the Department of Ophthalmology at Harvard Medical School, Boston, MA 02114, USA
| | - Evhy Apryani
- Schepens Eye Research Institute of Mass Eye and Ear and the Department of Ophthalmology at Harvard Medical School, Boston, MA 02114, USA
- Mass Eye and Ear and the Department of Ophthalmology at Harvard Medical School, Boston, MA 02114, USA
| | - Paula Perez-Corredor
- Schepens Eye Research Institute of Mass Eye and Ear and the Department of Ophthalmology at Harvard Medical School, Boston, MA 02114, USA
- Mass Eye and Ear and the Department of Ophthalmology at Harvard Medical School, Boston, MA 02114, USA
| | - Claudia Marino
- Schepens Eye Research Institute of Mass Eye and Ear and the Department of Ophthalmology at Harvard Medical School, Boston, MA 02114, USA
- Mass Eye and Ear and the Department of Ophthalmology at Harvard Medical School, Boston, MA 02114, USA
| | - Daisy Y Shu
- Schepens Eye Research Institute of Mass Eye and Ear and the Department of Ophthalmology at Harvard Medical School, Boston, MA 02114, USA
- Mass Eye and Ear and the Department of Ophthalmology at Harvard Medical School, Boston, MA 02114, USA
| | - Timothy E Vanderleest
- Schepens Eye Research Institute of Mass Eye and Ear and the Department of Ophthalmology at Harvard Medical School, Boston, MA 02114, USA
- Mass Eye and Ear and the Department of Ophthalmology at Harvard Medical School, Boston, MA 02114, USA
| | - Andres Muriel-Torres
- Schepens Eye Research Institute of Mass Eye and Ear and the Department of Ophthalmology at Harvard Medical School, Boston, MA 02114, USA
- Mass Eye and Ear and the Department of Ophthalmology at Harvard Medical School, Boston, MA 02114, USA
| | - Harper B Gordon
- Schepens Eye Research Institute of Mass Eye and Ear and the Department of Ophthalmology at Harvard Medical School, Boston, MA 02114, USA
- Mass Eye and Ear and the Department of Ophthalmology at Harvard Medical School, Boston, MA 02114, USA
| | - Audrey L Gunawan
- Schepens Eye Research Institute of Mass Eye and Ear and the Department of Ophthalmology at Harvard Medical School, Boston, MA 02114, USA
- Mass Eye and Ear and the Department of Ophthalmology at Harvard Medical School, Boston, MA 02114, USA
| | - Bryan A Kaplan
- Schepens Eye Research Institute of Mass Eye and Ear and the Department of Ophthalmology at Harvard Medical School, Boston, MA 02114, USA
- Mass Eye and Ear and the Department of Ophthalmology at Harvard Medical School, Boston, MA 02114, USA
| | - Karim W Barake
- Schepens Eye Research Institute of Mass Eye and Ear and the Department of Ophthalmology at Harvard Medical School, Boston, MA 02114, USA
- Mass Eye and Ear and the Department of Ophthalmology at Harvard Medical School, Boston, MA 02114, USA
| | - Romy P Bejjani
- Schepens Eye Research Institute of Mass Eye and Ear and the Department of Ophthalmology at Harvard Medical School, Boston, MA 02114, USA
- Mass Eye and Ear and the Department of Ophthalmology at Harvard Medical School, Boston, MA 02114, USA
| | - Tri H Doan
- Schepens Eye Research Institute of Mass Eye and Ear and the Department of Ophthalmology at Harvard Medical School, Boston, MA 02114, USA
- Mass Eye and Ear and the Department of Ophthalmology at Harvard Medical School, Boston, MA 02114, USA
| | - Rose Lin
- Schepens Eye Research Institute of Mass Eye and Ear and the Department of Ophthalmology at Harvard Medical School, Boston, MA 02114, USA
- Mass Eye and Ear and the Department of Ophthalmology at Harvard Medical School, Boston, MA 02114, USA
| | - Santiago Delgado-Tirado
- Schepens Eye Research Institute of Mass Eye and Ear and the Department of Ophthalmology at Harvard Medical School, Boston, MA 02114, USA
| | - Lucia Gonzalez-Buendia
- Schepens Eye Research Institute of Mass Eye and Ear and the Department of Ophthalmology at Harvard Medical School, Boston, MA 02114, USA
| | - Elizabeth J Rossin
- Schepens Eye Research Institute of Mass Eye and Ear and the Department of Ophthalmology at Harvard Medical School, Boston, MA 02114, USA
- Mass Eye and Ear and the Department of Ophthalmology at Harvard Medical School, Boston, MA 02114, USA
| | - Guannan Zhao
- Schepens Eye Research Institute of Mass Eye and Ear and the Department of Ophthalmology at Harvard Medical School, Boston, MA 02114, USA
| | - Dean Eliott
- Mass Eye and Ear and the Department of Ophthalmology at Harvard Medical School, Boston, MA 02114, USA
| | | | | | | | | | - Leo A Kim
- Schepens Eye Research Institute of Mass Eye and Ear and the Department of Ophthalmology at Harvard Medical School, Boston, MA 02114, USA
- Mass Eye and Ear and the Department of Ophthalmology at Harvard Medical School, Boston, MA 02114, USA
| | - Joseph F Arboleda-Velasquez
- Schepens Eye Research Institute of Mass Eye and Ear and the Department of Ophthalmology at Harvard Medical School, Boston, MA 02114, USA
- Mass Eye and Ear and the Department of Ophthalmology at Harvard Medical School, Boston, MA 02114, USA
| |
Collapse
|
26
|
Zhu J, He C, Liu Y, Chen M, Zhang J, Chen D, Ni H, Wen J. An engineered Japanese encephalitis virus mRNA-lipid nanoparticle immunization induces protective immunity in mice. Front Microbiol 2024; 15:1472824. [PMID: 39588106 PMCID: PMC11586386 DOI: 10.3389/fmicb.2024.1472824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 10/21/2024] [Indexed: 11/27/2024] Open
Abstract
Introduction Japanese encephalitis virus (JEV) and Zika virus (ZIKV) pose a severe threat to human health. Our previous research results, as well as those of other research groups, indicated that antibodies (Abs) induced by JEV infection or JEV vaccine vaccination could enhance ZIKV infection in vitro and exacerbate the mortality of ZIKV-infected mice, vice versa, which is known as antibody-dependent enhancement (ADE). Although studies on other flaviviruses revealed that altering the amino acid residues located in the fusion loop (FL) of envelope (E) protein can reduce the level of flavivirus-cross-reactive Abs, thereby abating the ADE of heterologous flavivirus infection, it is unclear whether this strategy is equally applicable to JEV. Methods In this study, we constructed recombinant adenoviruses and nucleotide-modified mRNA-lipid nanoparticle (LNP) encoding JEV wild-type E protein or E protein mutant (designated as Ad5-JEV-EWT and Ad5-JEV-Emut; JEV-EWT mRNA-LNP, and JEV-Emut mRNA-LNP). We evaluated the immunogenicity of these vaccine candidates in mice and the capacity of vaccine-immune mouse sera to neutralize JEV infection or mediate ADE of ZIKV infection in vitro and in vivo. Results Ad5-JEV-Emut or JEV-Emut mRNA-LNP immunization induced ZIKV-cross-reactive Ab response which is dramatically lower than that induced by Ad5-JEV-EWT and JEV-EWT mRNA-LNP, respectively. The levels of JEV-neutralizing Abs induced by Ad5-JEV-Emut or JEV-Emut mRNA-LNP are comparable to that induced by Ad5-JEV-EWT and JEV-EWT mRNA-LNP, respectively. The ability of Abs induced by Ad5-JEV-Emut to enhance ZIKV infection in vitro is attenuated as compared with that induced by Ad5-JEV-EWT. Moreover, JEV-Emut mRNA-LNP immunization elicited potent T cell response similar to JEV-EWT mRNA-LNP in mice. Mice immunized with each mRNA-LNP exhibited lower level of serum viral load than the mock-immunized mice post JEV challenge. Mice receiving JEV-EWT mRNA-LNP-immune mouse sera exhibited ADE post ZIKV challenge whereas passively transferred JEV-Emut mRNA-LNP-immune mouse sera did not lead to obvious ADE of ZIKV infection in recipient mice. Most importantly, maternally acquired Abs did not enhance the mortality of 1-day-old neonates born to JEV-Emut mRNA-LNP-immunized mice post ZIKV challenge. Discussion These results suggest that optimizing the FL sequence of JEV could significantly reduce the level of JEV/ZIKV-cross-reactive Abs and abrogate the ADE of ZIKV infection, providing a promising strategy to develop effective and safety JEV vaccine.
Collapse
Affiliation(s)
- Jiayang Zhu
- School of Basic Medical Sciences, Health Science Center, Ningbo University, Ningbo, China
| | - Caiying He
- School of Basic Medical Sciences, Health Science Center, Ningbo University, Ningbo, China
| | - Yusha Liu
- School of Basic Medical Sciences, Health Science Center, Ningbo University, Ningbo, China
| | - Min Chen
- School of Basic Medical Sciences, Health Science Center, Ningbo University, Ningbo, China
| | - Jiayi Zhang
- School of Basic Medical Sciences, Health Science Center, Ningbo University, Ningbo, China
- Wenzhou Central Blood Station, Wenzhou, China
- Key Laboratory of Laboratory Medicine, Ministry of Education, Zhejiang Provincial Key Laboratory of Medical Genetics, College of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, China
| | - Dong Chen
- School of Basic Medical Sciences, Health Science Center, Ningbo University, Ningbo, China
- Wenzhou Central Blood Station, Wenzhou, China
- Key Laboratory of Laboratory Medicine, Ministry of Education, Zhejiang Provincial Key Laboratory of Medical Genetics, College of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, China
| | - Hongxia Ni
- Ningbo Municipal Center for Disease Control and Prevention, Ningbo, China
| | - Jinsheng Wen
- School of Basic Medical Sciences, Health Science Center, Ningbo University, Ningbo, China
| |
Collapse
|
27
|
Chen Z, Shu J, Hu Y, Mei H. Synergistic integration of mRNA-LNP with CAR-engineered immune cells: Pioneering progress in immunotherapy. Mol Ther 2024; 32:3772-3792. [PMID: 39295145 PMCID: PMC11573621 DOI: 10.1016/j.ymthe.2024.09.019] [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: 04/26/2024] [Revised: 08/18/2024] [Accepted: 09/13/2024] [Indexed: 09/21/2024] Open
Abstract
Chimeric antigen receptor T cell (CAR-T) therapy has emerged as a revolutionary approach in the treatment of malignancies. Despite its remarkable successes, this field continues to grapple with challenges such as scalability, safety concerns, limited therapeutic effect, in vivo persistence, and the need for precise control over CAR expression. In the post-pandemic era of COVID-19 vaccine immunization, the application of messenger RNA (mRNA) encapsulated within lipid nanoparticles (LNPs) has recently garnered significant attention as a potential solution to address these challenges. This review delves into the dynamic landscape of mRNA-LNP technology and its potential implications for CAR-engineered immune cell-based immunotherapy.
Collapse
Affiliation(s)
- Zhaozhao Chen
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, Hubei, China; Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, Wuhan 430022, China
| | - Jinhui Shu
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, Hubei, China; Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, Wuhan 430022, China
| | - Yu Hu
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, Hubei, China; Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, Wuhan 430022, China
| | - Heng Mei
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, Hubei, China; Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, Wuhan 430022, China.
| |
Collapse
|
28
|
Kumar R, Krishnaperumal G, Vellapandian C. Innovative mRNA Vaccine Approaches in Targeting Atherosclerosis: A New Era in Cardiovascular Therapy. Cureus 2024; 16:e74141. [PMID: 39712846 PMCID: PMC11662511 DOI: 10.7759/cureus.74141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Accepted: 11/21/2024] [Indexed: 12/24/2024] Open
Abstract
Atherosclerosis, a major cause of cardiovascular disease (CVD), involves plaque buildup in arteries driven by inflammation, endothelial dysfunction, and lipid metabolism disturbances. Current therapies aim to reduce cholesterol through statins and proprotein convertase subtilisin/kexin type 9 (PCSK9) inhibitors, prevent blood clots with antiplatelet drugs like aspirin, and control inflammation, alongside lifestyle modifications. However, these approaches often fall short due to patient non-compliance and residual risks. This review explores emerging mRNA vaccine strategies targeting the complex mechanisms of atherosclerosis. These vaccines could produce therapeutic proteins to modulate inflammation by encoding sequences that inhibit pro-inflammatory cytokines such as interleukin-1β (IL-1β), interleukin-6 (IL-6), and tumor necrosis factor-alpha (TNF-α), stabilizing plaques. Key targets include interleukin-10 (IL-10) for plaque stability, PCSK9 for cholesterol regulation, and vascular endothelial growth factor (VEGF) for endothelial repair. Addressing these unmet needs, mRNA-based approaches offer the potential for more effective and personalized treatments for atherosclerosis. However, challenges remain, including difficulty replicating human atherosclerosis in preclinical models, regulatory concerns about long-term safety, and ensuring accessibility in low-resource settings. In addition, large and diverse clinical trials are needed to confirm the efficacy of these vaccines in reducing cardiovascular events.
Collapse
Affiliation(s)
- Rahul Kumar
- Pharmacy/Pharmacology, SRM College of Pharmacy, SRM Institute of Science and Technology (SRMIST), Chengalpattu, IND
| | - Gowri Krishnaperumal
- Pharmacology, SRM College of Pharmacy, SRM Institute of Science and Technology (SRMIST), Chengalpattu, IND
| | - Chitra Vellapandian
- Pharmacy/Pharmacology, SRM College of Pharmacy, SRM Institute of Science and Technology (SRMIST), Chengalpattu, IND
| |
Collapse
|
29
|
Garcia-Atutxa I, Mondragon-Teran P, Huerta-Saquero A, Villanueva-Flores F. Advancements in monkeypox vaccines development: a critical review of emerging technologies. Front Immunol 2024; 15:1456060. [PMID: 39464881 PMCID: PMC11502315 DOI: 10.3389/fimmu.2024.1456060] [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: 06/27/2024] [Accepted: 09/20/2024] [Indexed: 10/29/2024] Open
Abstract
Monkeypox (mpox) is a zoonotic illness caused by the monkeypox virus (MPXV), with higher health concerns among people who are pregnant, children, and persons who are immunocompromised, including people with untreated and advanced HIV disease. Significant progress has been made in developing vaccines against mpox, yet critical challenges and limitations persist in ensuring their effectiveness, safety, and accessibility. The pertinence of this review is highlighted by the World Health Organization's declaration of a global health emergency on August 14, 2024, due to the recent mpox outbreak, underscoring the critical necessity for effective vaccine solutions in the face of a rapidly evolving virus. Here, we comprehensively analyze various vaccine platforms utilized in mpox prevention, including attenuated and non-replicating virus vaccines, viral vector-based vaccines, recombinant protein vaccines, and DNA and mRNA vaccines. We evaluate the advantages and limitations of each platform, highlighting the urgent need for ongoing research and innovation to enhance vaccine efficacy and safety. Recent advancements, such as incorporating immunostimulatory sequences, improved delivery systems, and developing polyvalent vaccines, are explored for their potential to offer broader protection against diverse orthopoxvirus strains. This work underscores the need to optimize currently available vaccines and investigate novel vaccination strategies to address future public health emergencies effectively. By focusing on these advanced methodologies, we aim to contribute to the development of robust and adaptable vaccine solutions for mpox and other related viral threats.
Collapse
Affiliation(s)
- Igor Garcia-Atutxa
- Computer Science Department, Universidad Católica de Murcia (UCAM), Murcia, Spain
| | - Paul Mondragon-Teran
- Centro de Investigación en Ciencia Aplicada y Tecnología Avanzada (CICATA) Unidad Morelos del Instituto Politécnico Nacional (IPN), Xochitepec, Morelos, Mexico
| | - Alejandro Huerta-Saquero
- Departamento de Bionanotecnología, Centro de Nanociencias y Nanotecnología, Universidad Nacional Autónoma de México (UNAM), Ensenada, Mexico
| | - Francisca Villanueva-Flores
- Centro de Investigación en Ciencia Aplicada y Tecnología Avanzada (CICATA) Unidad Morelos del Instituto Politécnico Nacional (IPN), Xochitepec, Morelos, Mexico
| |
Collapse
|
30
|
Almanaa TN. Design of an Epitope-Based Vaccine Against MERS-CoV. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:1632. [PMID: 39459420 PMCID: PMC11509718 DOI: 10.3390/medicina60101632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 10/01/2024] [Accepted: 10/04/2024] [Indexed: 10/28/2024]
Abstract
Background and Objectives: Middle East Respiratory Syndrome (MERS) is a viral respiratory illness caused by a coronavirus called Middle East respiratory syndrome. In the current study, immunoinformatics studies were applied to design an epitope-based vaccine construct against Middle East Respiratory Syndrome. Materials and Methods: In this study, epitopes base vaccine construct was designed against MERS using immunoinformatics approach. Results: In this approach, the targeted proteins were screened, and probable antigenic, non-allergenic, and good water-soluble epitopes were selected for vaccine construction. In vaccine construction, the selected epitopes were joined by GPGPG linkers, and a linear multi-epitope vaccine was constructed. The vaccine construct underwent a physiochemical property analysis. The 3D structure of the vaccine construct was predicted and subjected to refinement. After the refinement, the 3D model was subjected to a molecular docking analysis, TLRs (TLR-3 and TLR-9) were selected as receptors for vaccine construct, and the molecular docking analysis study determined that the vaccine construct has binding ability with the targeted receptor. Conclusions: The docking analysis also unveils that the vaccine construct can properly activate immune system against the target virus however experimental validation is needed to confirm the in silico findings further.
Collapse
Affiliation(s)
- Taghreed N Almanaa
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh 11451, Saudi Arabia
| |
Collapse
|
31
|
Basnotra R, Philip J, Mallhi RS, Shrotriya A. Naturally Occurring Anti-D in a Male Blood Donor, Unresolved Mystery. Indian J Hematol Blood Transfus 2024; 40:715-718. [PMID: 39469151 PMCID: PMC11512962 DOI: 10.1007/s12288-024-01771-5] [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: 09/26/2023] [Accepted: 04/05/2024] [Indexed: 10/30/2024] Open
Abstract
In accordance with numerous national and international criteria, screening donated blood for abnormal antibodies against red cell antigens is crucial for patient safety. Antibodies can be formed by three mechanisms: by immune responses, naturally occurring, or passively acquired. In accordance with departmental policy, ICT was performed on a 30-yearold male blood donor's sample in addition to ABO Rh grouping. He tested A Rh Dnegative and had a positive ICT (+ 2). Anti-D was identified by using the antibody screening and identification panel. Retesting revealed similar results, so we called the donor. We were unable to find a source for the Rh isoimmunization during the course of his history. On further work-up, we found that antibodies reacted only in the antiglobulin phase with no reaction in the saline phase, suggesting an IgG type of antibody. Also, the anti-D titers (IgG) of the donor were up to 1:32. All potential sources of Rh isoimmunization have been ruled out for a male donor. Therefore, we assume that this donor has naturally occurring anti-D, perhaps created by certain environmental factors, bacteria, or components of the COVID vaccine that have cross-reactivity with the D antigen. This instance emphasizes how crucial the ICT test on the donor blood is. In our case, all potential causes of alloimmunization were ruled out, leaving only exposure to antigens with antigenic similarity to D antigens as a viable explanation.
Collapse
Affiliation(s)
- Ritika Basnotra
- Department of Immunohematology and Blood Transfusion, Medical College, Bharati Vidyapeeth Deemed University, Pune Satara Road, 411043 Dhankawadi, Pune, Maharashtra India
| | - Joseph Philip
- Department of Immunohematology and Blood Transfusion, Medical College, Bharati Vidyapeeth Deemed University, Pune Satara Road, 411043 Dhankawadi, Pune, Maharashtra India
| | - R. S. Mallhi
- Department of Immunohematology and Blood Transfusion, Medical College, Bharati Vidyapeeth Deemed University, Pune Satara Road, 411043 Dhankawadi, Pune, Maharashtra India
| | - Abhipsa Shrotriya
- Department of Immunohematology and Blood Transfusion, Medical College, Bharati Vidyapeeth Deemed University, Pune Satara Road, 411043 Dhankawadi, Pune, Maharashtra India
| |
Collapse
|
32
|
Chandra S, Wilson JC, Good D, Wei MQ. mRNA vaccines: a new era in vaccine development. Oncol Res 2024; 32:1543-1564. [PMID: 39308511 PMCID: PMC11413818 DOI: 10.32604/or.2024.043987] [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: 07/18/2023] [Accepted: 07/02/2024] [Indexed: 09/25/2024] Open
Abstract
The advent of RNA therapy, particularly through the development of mRNA cancer vaccines, has ushered in a new era in the field of oncology. This article provides a concise overview of the key principles, recent advancements, and potential implications of mRNA cancer vaccines as a groundbreaking modality in cancer treatment. mRNA cancer vaccines represent a revolutionary approach to combatting cancer by leveraging the body's innate immune system. These vaccines are designed to deliver specific mRNA sequences encoding cancer-associated antigens, prompting the immune system to recognize and mount a targeted response against malignant cells. This personalized and adaptive nature of mRNA vaccines holds immense potential for addressing the heterogeneity of cancer and tailoring treatments to individual patients. Recent breakthroughs in the development of mRNA vaccines, exemplified by the success of COVID-19 vaccines, have accelerated their application in oncology. The mRNA platform's versatility allows for the rapid adaptation of vaccine candidates to various cancer types, presenting an agile and promising avenue for therapeutic intervention. Clinical trials of mRNA cancer vaccines have demonstrated encouraging results in terms of safety, immunogenicity, and efficacy. Pioneering candidates, such as BioNTech's BNT111 and Moderna's mRNA-4157, have exhibited promising outcomes in targeting melanoma and solid tumors, respectively. These successes underscore the potential of mRNA vaccines to elicit robust and durable anti-cancer immune responses. While the field holds great promise, challenges such as manufacturing complexities and cost considerations need to be addressed for widespread adoption. The development of scalable and cost-effective manufacturing processes, along with ongoing clinical research, will be pivotal in realizing the full potential of mRNA cancer vaccines. Overall, mRNA cancer vaccines represent a cutting-edge therapeutic approach that holds the promise of transforming cancer treatment. As research progresses, addressing challenges and refining manufacturing processes will be crucial in advancing these vaccines from clinical trials to mainstream oncology practice, offering new hope for patients in the fight against cancer.
Collapse
Affiliation(s)
- Shubhra Chandra
- School of Pharmacy & Medical Sciences, Gold Coast campus, Griffith University, Brisbane, QLD-4222, Australia
- Menzies Health Institute Queensland (MHIQ), Gold Coast Campus, Griffith University, Brisbane, QLD-4215, Australia
| | - Jennifer C Wilson
- School of Pharmacy & Medical Sciences, Gold Coast campus, Griffith University, Brisbane, QLD-4222, Australia
- Menzies Health Institute Queensland (MHIQ), Gold Coast Campus, Griffith University, Brisbane, QLD-4215, Australia
| | - David Good
- School of Allied Health, Australian Catholic University, Brisbane, QLD-4014, Australia
| | - Ming Q Wei
- School of Pharmacy & Medical Sciences, Gold Coast campus, Griffith University, Brisbane, QLD-4222, Australia
- Menzies Health Institute Queensland (MHIQ), Gold Coast Campus, Griffith University, Brisbane, QLD-4215, Australia
| |
Collapse
|
33
|
Li J, Xiao L, Chen Z, Fan L, Wang W, Guo R, He Z, Hu H, Jiang J, Zhao L, Zhong T, Fan B, Zhu X, Li B. A spike-based mRNA vaccine that induces durable and broad protection against porcine deltacoronavirus in piglets. J Virol 2024; 98:e0053524. [PMID: 39158273 PMCID: PMC11406889 DOI: 10.1128/jvi.00535-24] [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: 03/21/2024] [Accepted: 07/16/2024] [Indexed: 08/20/2024] Open
Abstract
Coronaviruses (CoVs) are important pathogens for humans and other vertebrates, causing severe respiratory and intestinal infections that have become a threat to public health because of the potential for interspecies transmission between animals and humans. Therefore, the development of safe, effective vaccines remains a top priority for the control of CoV infection. The unique immunological characteristics of vaccines featuring messenger RNA (mRNA) present an advantageous tool for coronavirus vaccine development. Here, we designed two lipid nanoparticle (LNP)-encapsulated mRNA (mRNA-LNP) vaccines: one encoding full-length spike (S) protein and the other encoding the spike ectodomain (Se) from porcine deltacoronavirus (PDCoV). Fourteen days after primary immunization, both mRNA vaccines induced high levels of immunoglobulin G and neutralizing antibodies in mice, with the S vaccine showing better performance than the Se vaccine. Passive immune protection of the S mRNA vaccine in suckling piglets was confirmed by the induction of robust PDCoV-specific humoral and cellular immune responses. The S mRNA vaccine also showed better protective effects than the inactivated vaccine. Our results suggest that the novel PDCoV-S mRNA-LNP vaccine may have the potential to combat PDCoV infection. IMPORTANCE As an emerging porcine enteropathogenic coronavirus, porcine deltacoronavirus (PDCoV) has the potential for cross-species transmission, attracting extensive attention. Messenger RNA (mRNA) vaccines are a promising option for combating emerging and re-emerging infectious diseases, as evidenced by the demonstrated efficacy of the COVID-19 mRNA vaccine. Here, we first demonstrated that PDCoV-S mRNA-lipid nanoparticle (LNP) vaccines could induce potent humoral and cellular immune responses in mice. An evaluation of passive immune protection of S mRNA vaccines in suckling piglets confirmed that the protective effect of mRNA vaccine was better than that of inactivated vaccine. This study suggests that the PDCoV-S mRNA-LNP vaccine may serve as a potential and novel vaccine candidate for combating PDCoV infection.
Collapse
MESH Headings
- Animals
- Swine
- Coronavirus Infections/prevention & control
- Coronavirus Infections/immunology
- Coronavirus Infections/veterinary
- Coronavirus Infections/virology
- Antibodies, Neutralizing/immunology
- Antibodies, Neutralizing/blood
- Spike Glycoprotein, Coronavirus/immunology
- Spike Glycoprotein, Coronavirus/genetics
- Mice
- Swine Diseases/prevention & control
- Swine Diseases/virology
- Swine Diseases/immunology
- Antibodies, Viral/immunology
- Antibodies, Viral/blood
- Viral Vaccines/immunology
- Viral Vaccines/administration & dosage
- mRNA Vaccines
- Deltacoronavirus/immunology
- Deltacoronavirus/genetics
- Nanoparticles
- RNA, Messenger/genetics
- RNA, Messenger/immunology
- Immunoglobulin G/blood
- Immunoglobulin G/immunology
- Mice, Inbred BALB C
- Female
- Immunity, Humoral
- Liposomes
Collapse
Affiliation(s)
- Jizong Li
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Biological Engineering and Technology Ministry of Agriculture, Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base of Ministry of Science and Technology, Nanjing, China
- Institute of Life Sciences, School of Food and Biological Engineering, Jiangsu University, Zhenjiang, China
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
- Jiangsu Co-Innovation Center for the Prevention and Control of Important Animal Infectious Disease and Zoonose, Yangzhou University, Yangzhou, China
- Guotai (Taizhou) Center of Technology Innovation for Veterinary Biologicals, Taizhou, China
| | - Li Xiao
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Biological Engineering and Technology Ministry of Agriculture, Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base of Ministry of Science and Technology, Nanjing, China
- College of Animal Science, Guizhou University, Guiyang, China
| | - Zhuoqi Chen
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Biological Engineering and Technology Ministry of Agriculture, Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base of Ministry of Science and Technology, Nanjing, China
| | - Liyuan Fan
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Biological Engineering and Technology Ministry of Agriculture, Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base of Ministry of Science and Technology, Nanjing, China
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Wei Wang
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Biological Engineering and Technology Ministry of Agriculture, Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base of Ministry of Science and Technology, Nanjing, China
| | - Rongli Guo
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Biological Engineering and Technology Ministry of Agriculture, Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base of Ministry of Science and Technology, Nanjing, China
| | - Zhaoming He
- Suzhou Huiliao Biomedical Technology Co., Ltd., Suzhou, China
| | - Hongpeng Hu
- Suzhou Huiliao Biomedical Technology Co., Ltd., Suzhou, China
| | - Jianhao Jiang
- Suzhou Huiliao Biomedical Technology Co., Ltd., Suzhou, China
| | - Lixiang Zhao
- Suzhou Huiliao Biomedical Technology Co., Ltd., Suzhou, China
| | - Tianyi Zhong
- Suzhou Huiliao Biomedical Technology Co., Ltd., Suzhou, China
| | - Baochao Fan
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Biological Engineering and Technology Ministry of Agriculture, Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base of Ministry of Science and Technology, Nanjing, China
- Institute of Life Sciences, School of Food and Biological Engineering, Jiangsu University, Zhenjiang, China
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
- Jiangsu Co-Innovation Center for the Prevention and Control of Important Animal Infectious Disease and Zoonose, Yangzhou University, Yangzhou, China
- Guotai (Taizhou) Center of Technology Innovation for Veterinary Biologicals, Taizhou, China
| | - Xing Zhu
- College of Animal Science, Guizhou University, Guiyang, China
| | - Bin Li
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Biological Engineering and Technology Ministry of Agriculture, Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base of Ministry of Science and Technology, Nanjing, China
- Institute of Life Sciences, School of Food and Biological Engineering, Jiangsu University, Zhenjiang, China
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
- Jiangsu Co-Innovation Center for the Prevention and Control of Important Animal Infectious Disease and Zoonose, Yangzhou University, Yangzhou, China
- Guotai (Taizhou) Center of Technology Innovation for Veterinary Biologicals, Taizhou, China
| |
Collapse
|
34
|
Shah SK, Bhandari K, Shah A, Chaurasiya G. COVID-19: vaccination, therapeutics and a review of the science and public health. Ann Med Surg (Lond) 2024; 86:5343-5353. [PMID: 39239001 PMCID: PMC11374161 DOI: 10.1097/ms9.0000000000002374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 07/06/2024] [Indexed: 09/07/2024] Open
Abstract
COVID-19, stemming from the SARS-CoV-2 virus, has initiated a worldwide respiratory pandemic. Remarkable headway has been made in the realm of vaccination, as nearly every nation has initiated COVID-19 vaccine deployment. However, a mere 32.6% of individuals in low-income countries have received only a single vaccine dose. Unprecedented research and development endeavors have yielded over 170 COVID-19 vaccines, several of which are now in practical use. These vaccines have demonstrated remarkable efficacy in averting severe illness, hospitalization, and fatalities from COVID-19, even against emerging variants. Research pursuits persist, concentrating on novel vaccine technologies, oral and nasal vaccines, broader coronavirus protection, and vaccine combinations. In the realm of therapeutics, there have been significant strides in developing oral antiviral medications and monoclonal antibodies. Nonetheless, challenges in COVID-19 vaccination persist, encompassing issues of hesitancy, accessibility, financial barriers, knowledge gaps, and logistical hindrances. Robust monitoring via global agencies and reporting systems remains pivotal. Strategies for enhancing vaccination efficacy are rooted in fostering trust, countering misinformation, and expanding access. As for therapeutics, the approach involves dedicated research, clinical trials, regulatory streamlining, stockpiling, and international collaboration. Telemedicine and public awareness campaigns play integral roles in this effort, with coordination being the linchpin for preserving lives and mitigating the disease's impact. The global campaign against COVID-19 has witnessed substantial advancements, with an ongoing research focus on developing vaccines and therapeutics that are not only more accessible and affordable but also more effective, particularly for populations in low-income countries and vulnerable communities.
Collapse
Affiliation(s)
| | | | - Avish Shah
- Kist Medical College and Teaching Hospital, Imadol, Lalitpur
- Everest Hospital, New Baneshwor, Kathmandu, Nepal
| | | |
Collapse
|
35
|
Coll De Peña A, Gutterman-Johns E, Gautam GP, Rutberg J, Frej MB, Mehta DR, Shah S, Tripathi A. Assessment of pDNA isoforms using microfluidic electrophoresis. Electrophoresis 2024; 45:1525-1534. [PMID: 38571381 DOI: 10.1002/elps.202300293] [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: 12/17/2023] [Revised: 03/11/2024] [Accepted: 03/15/2024] [Indexed: 04/05/2024]
Abstract
The recent rise in nucleic acid-based vaccines and therapies has resulted in an increased demand for plasmid DNA (pDNA). As a result, there is added pressure to streamline the manufacturing of these vectors, particularly their design and construction, which is currently considered a bottleneck. A significant challenge in optimizing pDNA production is the lack of high-throughput and rapid analytical methods to support the numerous samples produced during the iterative plasmid construction step and for batch-to-batch purity monitoring. pDNA is generally present as one of three isoforms: supercoiled, linear, or open circular. Depending on the ultimate use, the desired isoform may be supercoiled in the initial stages for cell transfection or linear in the case of mRNA synthesis. Here, we present a high-throughput microfluidic electrophoresis method capable of detecting the three pDNA isoforms and determining the size and concentration of the predominant supercoiled and linear isoforms from 2 to 7 kb. The limit of detection of the method is 0.1 ng/µL for the supercoiled and linear isoforms and 0.5 ng/µL for the open circular isoform, with a maximum loading capacity of 10-15 ng/µL. The turnaround time is 1 min/sample, and the volume requirement is 10 µL, making the method suitable for process optimization and batch-to-batch analysis. The results presented in this study will enhance the understanding of electrophoretic transport in microscale systems dependent on molecular conformations and potentially aid technological advances in diverse areas relevant to microfluidic devices.
Collapse
Affiliation(s)
- Adriana Coll De Peña
- Center for Biomedical Engineering, School of Engineering, Brown University, Providence, Rhode Island, USA
| | - Everett Gutterman-Johns
- Department of Molecular Biology, Cell Biology, and Biochemistry, Division of Biology and Medicine, Brown University, Providence, Rhode Island, USA
| | | | - Jenna Rutberg
- Center for Biomedical Engineering, School of Engineering, Brown University, Providence, Rhode Island, USA
| | - Menel Ben Frej
- Applied Genomics, Revvity, Hopkinton, Massachusetts, USA
| | - Dipti R Mehta
- Applied Genomics, Revvity, Hopkinton, Massachusetts, USA
| | - Shreyas Shah
- Applied Genomics, Revvity, Hopkinton, Massachusetts, USA
| | - Anubhav Tripathi
- Center for Biomedical Engineering, School of Engineering, Brown University, Providence, Rhode Island, USA
| |
Collapse
|
36
|
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
|
37
|
Li S, Moayedpour S, Li R, Bailey M, Riahi S, Kogler-Anele L, Miladi M, Miner J, Pertuy F, Zheng D, Wang J, Balsubramani A, Tran K, Zacharia M, Wu M, Gu X, Clinton R, Asquith C, Skaleski J, Boeglin L, Chivukula S, Dias A, Strugnell T, Montoya FU, Agarwal V, Bar-Joseph Z, Jager S. CodonBERT large language model for mRNA vaccines. Genome Res 2024; 34:1027-1035. [PMID: 38951026 PMCID: PMC11368176 DOI: 10.1101/gr.278870.123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 06/25/2024] [Indexed: 07/03/2024]
Abstract
mRNA-based vaccines and therapeutics are gaining popularity and usage across a wide range of conditions. One of the critical issues when designing such mRNAs is sequence optimization. Even small proteins or peptides can be encoded by an enormously large number of mRNAs. The actual mRNA sequence can have a large impact on several properties, including expression, stability, immunogenicity, and more. To enable the selection of an optimal sequence, we developed CodonBERT, a large language model (LLM) for mRNAs. Unlike prior models, CodonBERT uses codons as inputs, which enables it to learn better representations. CodonBERT was trained using more than 10 million mRNA sequences from a diverse set of organisms. The resulting model captures important biological concepts. CodonBERT can also be extended to perform prediction tasks for various mRNA properties. CodonBERT outperforms previous mRNA prediction methods, including on a new flu vaccine data set.
Collapse
Affiliation(s)
- Sizhen Li
- Digital R&D, Sanofi, Cambridge, Massachusetts 02141, USA
| | | | - Ruijiang Li
- Digital R&D, Sanofi, Cambridge, Massachusetts 02141, USA
| | - Michael Bailey
- Digital R&D, Sanofi, Cambridge, Massachusetts 02141, USA
| | - Saleh Riahi
- Digital R&D, Sanofi, Cambridge, Massachusetts 02141, USA
| | | | - Milad Miladi
- mRNA Center of Excellence, Sanofi, Waltham, Massachusetts 02451, USA
| | - Jacob Miner
- mRNA Center of Excellence, Sanofi, Waltham, Massachusetts 02451, USA
| | - Fabien Pertuy
- mRNA Center of Excellence, Sanofi, Waltham, Massachusetts 02451, USA
| | - Dinghai Zheng
- mRNA Center of Excellence, Sanofi, Waltham, Massachusetts 02451, USA
| | - Jun Wang
- mRNA Center of Excellence, Sanofi, Waltham, Massachusetts 02451, USA
| | | | - Khang Tran
- mRNA Center of Excellence, Sanofi, Waltham, Massachusetts 02451, USA
| | - Minnie Zacharia
- mRNA Center of Excellence, Sanofi, Waltham, Massachusetts 02451, USA
| | - Monica Wu
- mRNA Center of Excellence, Sanofi, Waltham, Massachusetts 02451, USA
| | - Xiaobo Gu
- mRNA Center of Excellence, Sanofi, Waltham, Massachusetts 02451, USA
| | - Ryan Clinton
- mRNA Center of Excellence, Sanofi, Waltham, Massachusetts 02451, USA
| | - Carla Asquith
- mRNA Center of Excellence, Sanofi, Waltham, Massachusetts 02451, USA
| | - Joseph Skaleski
- mRNA Center of Excellence, Sanofi, Waltham, Massachusetts 02451, USA
| | - Lianne Boeglin
- mRNA Center of Excellence, Sanofi, Waltham, Massachusetts 02451, USA
| | - Sudha Chivukula
- mRNA Center of Excellence, Sanofi, Waltham, Massachusetts 02451, USA
| | - Anusha Dias
- mRNA Center of Excellence, Sanofi, Waltham, Massachusetts 02451, USA
| | - Tod Strugnell
- mRNA Center of Excellence, Sanofi, Waltham, Massachusetts 02451, USA
| | | | - Vikram Agarwal
- mRNA Center of Excellence, Sanofi, Waltham, Massachusetts 02451, USA
| | - Ziv Bar-Joseph
- Digital R&D, Sanofi, Cambridge, Massachusetts 02141, USA;
| | - Sven Jager
- Digital R&D, Sanofi, Cambridge, Massachusetts 02141, USA
| |
Collapse
|
38
|
Isaac I, Bhatia M, Bhattacharya C. Recent Advances in Biomaterials for mRNA Delivery to Immune Cells. ACS APPLIED BIO MATERIALS 2024; 7:5136-5146. [PMID: 39058246 DOI: 10.1021/acsabm.4c00734] [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] [Indexed: 07/28/2024]
Abstract
Modulating the function of immune cells by targeting the cells themselves has become a key strategy in immunotherapy for combating various diseases such as cancer, autoimmune disorders, and infectious ailments. The use of mRNA (mRNA) is a powerful tool for transiently inducing protein expression, which is often used for genetic manipulation. However, its inherent instability and inability to precisely reach target cells necessitate the use of biomaterials for safe and effective delivery. Additionally, transfecting immune cells is difficult and complex due to their resistance mechanisms, signaling pathways, and cellular interactions. This review focuses on the recent development of biomaterials for mRNA delivery to immune cells, including lipid nanoparticles and polymeric carriers. It also outlines the challenges of targeting and delivering therapeutic payloads to immune cells, providing commentary and outlook on the design of next-generation materials. Finally, this approach has the potential to significantly enhance the precision and effectiveness of therapeutic interventions for various diseases, shaping the future of mRNA delivery for immune conditions.
Collapse
Affiliation(s)
- Ivan Isaac
- Department of Chemistry and Biochemistry, University of Nevada─Las Vegas 4505 S. Maryland Parkway, Las Vegas, Nevada 89154, United States
| | - Mayurakkhi Bhatia
- Department of Chemistry and Biochemistry, University of Nevada─Las Vegas 4505 S. Maryland Parkway, Las Vegas, Nevada 89154, United States
| | - Chandrabali Bhattacharya
- Department of Chemistry and Biochemistry, University of Nevada─Las Vegas 4505 S. Maryland Parkway, Las Vegas, Nevada 89154, United States
- Interdisciplinary Biomedical Engineering Program, University of Nevada─Las Vegas, Las Vegas, Nevada 89154, United States
| |
Collapse
|
39
|
Mujahid K, Rana I, Suliman IH, Li Z, Wu J, He H, Nam J. Biomaterial-Based Sustained-Release Drug Formulations for Localized Cancer Immunotherapy. ACS APPLIED BIO MATERIALS 2024; 7:4944-4961. [PMID: 38050811 DOI: 10.1021/acsabm.3c00808] [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] [Indexed: 12/07/2023]
Abstract
Cancer immunotherapy has revolutionized clinical cancer treatments by taking advantage of the immune system to selectively and effectively target and kill cancer cells. However, clinical cancer immunotherapy treatments often have limited efficacy and/or present severe adverse effects associated primarily with their systemic administration. Localized immunotherapy has emerged to overcome these limitations by directly targeting accessible tumors via local administration, reducing potential systemic drug distribution that hampers drug efficacy and safety. Sustained-release formulations can prolong drug activity at target sites, which maximizes the benefits of localized immunotherapy to increase the therapeutic window using smaller dosages than those used for systemic injection, avoiding complications of frequent dosing. The performance of sustained-release formulations for localized cancer immunotherapy has been validated preclinically using various implantable and injectable scaffold platforms. This review introduces the sustained-release formulations developed for localized cancer immunotherapy and highlights their biomaterial-based platforms for representative classes, including inorganic scaffolds, natural hydrogels, synthetic hydrogels, and microneedle patches. The design rationale and other considerations are summarized for further development of biomaterials for the construction of optimal sustained-release formulations.
Collapse
Affiliation(s)
- Khizra Mujahid
- College of Pharmacy, Chonnam National University, Gwangju 61186, South Korea
| | - Isra Rana
- College of Pharmacy, Chonnam National University, Gwangju 61186, South Korea
| | | | - Zhen Li
- College of Pharmacy, Chonnam National University, Gwangju 61186, South Korea
| | - Jiang Wu
- School of Pharmaceutical Sciences, Key Laboratory of Biotechnology and Pharmaceutical Engineering, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Huacheng He
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou, Zhejiang 325000, P. R. China
| | - Jutaek Nam
- College of Pharmacy, Chonnam National University, Gwangju 61186, South Korea
| |
Collapse
|
40
|
Wimalawansa SJ. Unlocking insights: Navigating COVID-19 challenges and Emulating future pandemic Resilience strategies with strengthening natural immunity. Heliyon 2024; 10:e34691. [PMID: 39166024 PMCID: PMC11334859 DOI: 10.1016/j.heliyon.2024.e34691] [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: 03/06/2024] [Revised: 06/17/2024] [Accepted: 07/15/2024] [Indexed: 08/22/2024] Open
Abstract
The original COVID-19 vaccines, developed against SARS-CoV-2, initially mitigated hospitalizations. Bivalent vaccine boosters were used widely during 2022-23, but the outbreaks persisted. Despite this, hospitalizations, mortality, and outbreaks involving dominant mutants like Alpha and Delta increased during winters when the population's vitamin D levels were at their lowest. Notably, 75 % of human immune cell/system functions, including post-vaccination adaptive immunity, rely on adequate circulatory vitamin D levels. Consequently, hypovitaminosis compromises innate and adaptive immune responses, heightening susceptibility to infections and complications. COVID-19 vaccines primarily target SARS-CoV-2 Spike proteins, thus offering only a limited protection through antibodies. mRNA vaccines, such as those for COVID-19, fail to generate secretory/mucosal immunity-like IgG responses, rendering them ineffective in halting viral spread. Additionally, mutations in the SARS-CoV-2 binding domain reduce immune recognition by vaccine-derived antibodies, leading to immune evasion by mutant viruses like Omicron variants. Meanwhile, the repeated administration of bivalent boosters intended to enhance efficacy resulted in the immunoparesis of recipients. As a result, relying solely on vaccines for outbreak prevention, it became less effective. Dominant variants exhibit increased affinity to angiotensin-converting enzyme receptor-2, enhancing infectivity but reducing virulence. Meanwhile, spike protein-related viral mutations do not impact the potency of widely available, repurposed early therapies, like vitamin D and ivermectin. With the re-emergence of COVID-19 and impending coronaviral pandemics, regulators and health organizations should proactively consider approval and strategic use of cost-effective adjunct therapies mentioned above to counter the loss of vaccine efficacy against emerging variants and novel coronaviruses and eliminate vaccine- and anti-viral agents-related serious adverse effects. Timely implementation of these strategies could reduce morbidity, mortality, and healthcare costs and provide a rational approach to address future epidemics and pandemics. This perspective critically reviews relevant literature, providing insights, justifications, and viewpoints into how the scientific community and health authorities can leverage this knowledge cost-effectively.
Collapse
Affiliation(s)
- Sunil J. Wimalawansa
- Medicine, Endocrinology, and Nutrition, B14 G2, De Soyza Flats, Moratuwa, Sri Lanka
| |
Collapse
|
41
|
Hussain A, Wang M, Yu D, Zhang J, Naseer QA, Ullah A, Milon Essola J, Zhang X. Medical and molecular biophysical techniques as substantial tools in the era of mRNA-based vaccine technology. Biomater Sci 2024; 12:4117-4135. [PMID: 39016519 DOI: 10.1039/d4bm00561a] [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: 07/18/2024]
Abstract
The COVID-19 pandemic prompted the advancement of vaccine technology using mRNA delivery into the host cells. Consequently, mRNA-based vaccines have emerged as a practical approach against SARS-CoV-2 owing to their inherent properties, such as cost-effectiveness, rapid manufacturing, and preservation. These features are vital, especially in resource-constrained regions. Nevertheless, the design of mRNA-based vaccines is intricately intertwined with the refinement of biophysical technologies, thereby establishing their high potential. The preparation of mRNA-based vaccines involves a sequence of phases combining medical and molecular biophysical technologies. Furthermore, their efficiency depends on the capability to optimize their positive attributes, thus paving the way for their subsequent preclinical and clinical evaluations. Using biophysical techniques, the characterization of nucleic acids extends from their initial formulation to their cellular internalization abilities and encapsulation in biomolecule complexes, such as lipid nanoparticles (LNPs), for designing mRNA-based LNPs. Furthermore, nanoparticles are subjected to a series of careful screening steps to assess their physical and chemical characteristics before achieving an optimum formulation suitable for preclinical and clinical studies. This review provides a comprehensive understanding of the fundamental role of biophysical techniques in the complex development of mRNA-based vaccines and their role in the recent success during the COVID-19 pandemic.
Collapse
Affiliation(s)
- Abid Hussain
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, 212013, Jiangsu, China.
| | - Maoye Wang
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, 212013, Jiangsu, China.
| | - Dan Yu
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, 212013, Jiangsu, China.
| | - Jiahui Zhang
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, 212013, Jiangsu, China.
| | - Qais Ahmad Naseer
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, China
| | - Aftab Ullah
- School of Medicine, Huaqiao University, No. 269 Chenghua North Rd., Quanzhou, Fujian 362021, China.
| | - Julien Milon Essola
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Centre for Nanoscience and Technology of China, No. 11, First North Road, Zhongguancun, Beijing, 100190, P.R. China.
- University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Xu Zhang
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, 212013, Jiangsu, China.
| |
Collapse
|
42
|
Oladipo EK, Oyelakin OD, Aiyelabegan AO, Olajide EO, Olatayo VO, Owolabi KP, Shittu YB, Olugbodi RO, Ajala HA, Rukayat RA, Olayiwola DO, Irewolede BA, Jimah EM, Oloke JK, Ojo TO, Ajani OF, Iwalokun BA, Kolawole OM, Ariyo OE, Adediran DA, Olufemi SE, Onyeaka H. Exploring computational approaches to design mRNA Vaccine against vaccinia and Mpox viruses. Immun Inflamm Dis 2024; 12:e1360. [PMID: 39150224 PMCID: PMC11328121 DOI: 10.1002/iid3.1360] [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: 08/02/2023] [Revised: 07/15/2024] [Accepted: 07/19/2024] [Indexed: 08/17/2024] Open
Abstract
BACKGROUND Messenger RNA (mRNA) vaccines emerged as a powerful tool in the fight against infections. Unlike traditional vaccines, this unique type of vaccine elicits robust and persistent innate and humoral immune response with a unique host cell-mediated pathogen gene expression and antigen presentation. METHODS This offers a novel approach to combat poxviridae infections. From the genome of vaccinia and Mpox viruses, three key genes (E8L, E7R, and H3L) responsible for virus attachment and virulence were selected and employed for designing the candidate mRNA vaccine against vaccinia and Mpox viral infection. Various bioinformatics tools were employed to generate (B cell, CTL, and HTL) epitopes, of which 28 antigenic and immunogenic epitopes were selected and are linked to form the mRNA vaccine construct. Additional components, including a 5' cap, 5' UTR, adjuvant, 3' UTR, and poly(A) tail, were incorporated to enhance stability and effectiveness. Safety measures such as testing for human homology and in silico immune simulations were implemented to avoid autoimmunity and to mimics the immune response of human host to the designed mRNA vaccine, respectively. The mRNA vaccine's binding affinity was evaluated by docking it with TLR-2, TLR-3, TLR-4, and TLR-9 receptors which are subsequently followed by molecular dynamics simulations for the highest binding one to predict the stability of the binding complex. RESULTS With a 73% population coverage, the mRNA vaccine looks promising, boasting a molecular weight of 198 kDa and a molecular formula of C8901H13609N2431O2611S48 and it is said to be antigenic, nontoxic and nonallergic, making it safe and effective in preventing infections with Mpox and vaccinia viruses, in comparison with other insilico-designed vaccine for vaccinia and Mpox viruses. CONCLUSIONS However, further validation through in vivo and in vitro techniques is underway to fully assess its potential.
Collapse
Affiliation(s)
- Elijah K. Oladipo
- Division of Vaccine Design and DevelopmentHelix Biogen InstituteOgbomosoOyo StateNigeria
- Laboratory of Molecular Biology, Immunology and Bioinformatics, Department of MicrobiologyAdeleke UniversityEdeOsun StateNigeria
| | - Olanrewaju D. Oyelakin
- Division of Vaccine Design and DevelopmentHelix Biogen InstituteOgbomosoOyo StateNigeria
| | | | - Elizabeth O. Olajide
- Division of Vaccine Design and DevelopmentHelix Biogen InstituteOgbomosoOyo StateNigeria
- Molecular Biology and Biotechnology DepartmentNigeria Institute of Medical ResearchLagosNigeria
| | - Victoria O. Olatayo
- Division of Vaccine Design and DevelopmentHelix Biogen InstituteOgbomosoOyo StateNigeria
| | - Kaothar P. Owolabi
- Division of Vaccine Design and DevelopmentHelix Biogen InstituteOgbomosoOyo StateNigeria
| | - Yewande B. Shittu
- Division of Vaccine Design and DevelopmentHelix Biogen InstituteOgbomosoOyo StateNigeria
| | - Rhoda O. Olugbodi
- Division of Vaccine Design and DevelopmentHelix Biogen InstituteOgbomosoOyo StateNigeria
| | - Hezekiah A. Ajala
- Division of Vaccine Design and DevelopmentHelix Biogen InstituteOgbomosoOyo StateNigeria
| | - Raji A. Rukayat
- Division of Vaccine Design and DevelopmentHelix Biogen InstituteOgbomosoOyo StateNigeria
| | - Deborah O. Olayiwola
- Division of Vaccine Design and DevelopmentHelix Biogen InstituteOgbomosoOyo StateNigeria
| | | | - Esther M. Jimah
- Division of Vaccine Design and DevelopmentHelix Biogen InstituteOgbomosoOyo StateNigeria
| | - Julius K. Oloke
- Department of Natural SciencesPrecious Cornerstone UniversityIbadanOyo StateNigeria
| | - Taiwo O. Ojo
- Division of Vaccine Design and DevelopmentHelix Biogen InstituteOgbomosoOyo StateNigeria
| | | | - Bamidele A. Iwalokun
- Molecular Biology and Biotechnology DepartmentNigeria Institute of Medical ResearchLagosNigeria
| | | | - Olumuyiwa E. Ariyo
- Department of Medicine, Infectious Disease and Tropical Medicine UnitFederal Teaching HospitalIdo EkitiEkiti StateNigeria
| | - Daniel A. Adediran
- Division of Vaccine Design and DevelopmentHelix Biogen InstituteOgbomosoOyo StateNigeria
- Department of BiochemistryLadoke Akintola University of TechnologyOgbomosoOyo StateNigeria
| | - Seun E. Olufemi
- Division of Vaccine Design and DevelopmentHelix Biogen InstituteOgbomosoOyo StateNigeria
- Department of BiochemistryLadoke Akintola University of TechnologyOgbomosoOyo StateNigeria
| | - Helen Onyeaka
- School of Chemical EngineeringUniversity of BirminghamBirminghamUK
| |
Collapse
|
43
|
Chen B, Yang Y, Wang X, Yang W, Lu Y, Wang D, Zhuo E, Tang Y, Su J, Tang G, Shao S, Gu K. mRNA vaccine development and applications: A special focus on tumors (Review). Int J Oncol 2024; 65:81. [PMID: 38994758 PMCID: PMC11251742 DOI: 10.3892/ijo.2024.5669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Accepted: 05/20/2024] [Indexed: 07/13/2024] Open
Abstract
Cancer is characterized by unlimited proliferation and metastasis, and traditional therapeutic strategies usually result in the acquisition of drug resistance, thus highlighting the need for more personalized treatment. mRNA vaccines transfer the gene sequences of exogenous target antigens into human cells through transcription and translation to stimulate the body to produce specific immune responses against the encoded proteins, so as to enable the body to obtain immune protection against said antigens; this approach may be adopted for personalized cancer therapy. Since the recent coronavirus pandemic, the development of mRNA vaccines has seen substantial progress and widespread adoption. In the present review, the development of mRNA vaccines, their mechanisms of action, factors influencing their function and the current clinical applications of the vaccine are discussed. A focus is placed on the application of mRNA vaccines in cancer, with the aim of highlighting unique advances and the remaining challenges of this novel and promising therapeutic approach.
Collapse
Affiliation(s)
- Bangjie Chen
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, P.R. China
| | - Yipin Yang
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, P.R. China
| | - Xinyi Wang
- Department of Radiation Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, P.R. China
| | - Wenzhi Yang
- First Clinical Medical College, Anhui Medical University, Hefei, Anhui 230022, P.R. China
| | - You Lu
- First Clinical Medical College, Anhui Medical University, Hefei, Anhui 230022, P.R. China
| | - Daoyue Wang
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, P.R. China
| | - Enba Zhuo
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, P.R. China
| | - Yanchao Tang
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, P.R. China
| | - Junhong Su
- Department of Rehabilitation, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, P.R. China
| | - Guozheng Tang
- Department of Orthopedics, Lu'an Hospital of Anhui Medical University, Lu'an, Anhui 237008, P.R. China
| | - Song Shao
- Department of Orthopedics, Lu'an Hospital of Anhui Medical University, Lu'an, Anhui 237008, P.R. China
| | - Kangsheng Gu
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, P.R. China
| |
Collapse
|
44
|
Chang LA, Schotsaert M. Ally, adversary, or arbitrator? The context-dependent role of eosinophils in vaccination for respiratory viruses and subsequent breakthrough infections. J Leukoc Biol 2024; 116:224-243. [PMID: 38289826 PMCID: PMC11288382 DOI: 10.1093/jleuko/qiae010] [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: 10/29/2023] [Revised: 12/12/2023] [Accepted: 12/26/2023] [Indexed: 02/01/2024] Open
Abstract
Eosinophils are a critical type of immune cell and central players in type 2 immunity. Existing literature suggests that eosinophils also can play a role in host antiviral responses, typically type 1 immune events, against multiple respiratory viruses, both directly through release of antiviral mediators and indirectly through activation of other effector cell types. One way to prime host immune responses toward effective antiviral responses is through vaccination, where typically a type 1-skewed immunity is desirable in the context of intracellular pathogens like respiratory viruses. In the realm of breakthrough respiratory viral infection in vaccinated hosts, an event in which virus can still establish productive infection despite preexisting immunity, eosinophils are most prominently known for their link to vaccine-associated enhanced respiratory disease upon natural respiratory syncytial virus infection. This was observed in a pediatric cohort during the 1960s following vaccination with formalin-inactivated respiratory syncytial virus. More recent research has unveiled additional roles of the eosinophil in respiratory viral infection and breakthrough infection. The specific contribution of eosinophils to the quality of vaccine responses, vaccine efficacy, and antiviral responses to infection in vaccinated hosts remains largely unexplored, especially regarding their potential roles in protection. On the basis of current findings, we will speculate upon the suggested function of eosinophils and consider the many potential ways by which eosinophils may exert protective and pathological effects in breakthrough infections. We will also discuss how to balance vaccine efficacy with eosinophil-related risks, as well as the use of eosinophils and their products as potential biomarkers of vaccine efficacy or adverse events.
Collapse
Affiliation(s)
- Lauren A Chang
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, United States
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1124, New York, NY 10029, United States
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1124, New York, NY 10029, United States
| | - Michael Schotsaert
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1124, New York, NY 10029, United States
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1124, New York, NY 10029, United States
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, 1425 Madison Avenue, Box 1630, New York, NY 10029, United States
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, United States
| |
Collapse
|
45
|
Duan M, Dev I, Lu A, Ayrapetyan G, You MY, Shapiro MG. SEMPER: Stoichiometric expression of mRNA polycistrons by eukaryotic ribosomes for compact, ratio-tunable multi-gene expression. Cell Syst 2024; 15:597-609.e4. [PMID: 38971149 PMCID: PMC11298409 DOI: 10.1016/j.cels.2024.06.001] [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: 06/07/2023] [Revised: 04/01/2024] [Accepted: 06/12/2024] [Indexed: 07/08/2024]
Abstract
Here, we present a method for expressing multiple open reading frames (ORFs) from single transcripts using the leaky scanning model of translation initiation. In this approach termed "stoichiometric expression of mRNA polycistrons by eukaryotic ribosomes" (SEMPER), adjacent ORFs are translated from a single mRNA at tunable ratios determined by their order in the sequence and the strength of their translation initiation sites. We validate this approach by expressing up to three fluorescent proteins from one plasmid in two different cell lines. We then use it to encode a stoichiometrically tuned polycistronic construct encoding gas vesicle acoustic reporter genes that enables efficient formation of the multi-protein complex while minimizing cellular toxicity. We also demonstrate that SEMPER enables polycistronic expression of recombinant monoclonal antibodies from plasmid DNA and of two fluorescent proteins from single mRNAs made through in vitro transcription. Finally, we provide a probabilistic model to elucidate the mechanisms underlying SEMPER. A record of this paper's transparent peer review process is included in the supplemental information.
Collapse
Affiliation(s)
- Mengtong Duan
- Division of Biology and Biological Engineering, Caltech, Pasadena, CA 91125, USA
| | - Ishaan Dev
- Division of Chemistry and Chemical Engineering, Caltech, Pasadena, CA 91125, USA
| | - Andrew Lu
- Division of Biology and Biological Engineering, Caltech, Pasadena, CA 91125, USA; UCLA-Caltech Medical Scientist Training Program, UCLA, Los Angeles, CA 90095, USA
| | - Goar Ayrapetyan
- Division of Chemistry and Chemical Engineering, Caltech, Pasadena, CA 91125, USA
| | - Mei Yi You
- Division of Biology and Biological Engineering, Caltech, Pasadena, CA 91125, USA
| | - Mikhail G Shapiro
- Division of Chemistry and Chemical Engineering, Caltech, Pasadena, CA 91125, USA; Andrew and Peggy Cherng Department of Medical Engineering, Caltech, Pasadena, CA 91125, USA; Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA.
| |
Collapse
|
46
|
Masum MHU, Wajed S, Hossain MI, Moumi NR, Talukder A, Rahman MM. An mRNA vaccine for pancreatic cancer designed by applying in silico immunoinformatics and reverse vaccinology approaches. PLoS One 2024; 19:e0305413. [PMID: 38976715 PMCID: PMC11230540 DOI: 10.1371/journal.pone.0305413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Accepted: 05/30/2024] [Indexed: 07/10/2024] Open
Abstract
Pancreatic ductal adenocarcinoma is the most prevalent pancreatic cancer, which is considered a significant global health concern. Chemotherapy and surgery are the mainstays of current pancreatic cancer treatments; however, a few cases are suitable for surgery, and most of the cases will experience recurrent episodes. Compared to DNA or peptide vaccines, mRNA vaccines for pancreatic cancer have more promise because of their delivery, enhanced immune responses, and lower proneness to mutation. We constructed an mRNA vaccine by analyzing S100 family proteins, which are all major activators of receptors for advanced glycation end products. We applied immunoinformatic approaches, including physicochemical properties analysis, structural prediction and validation, molecular docking study, in silico cloning, and immune simulations. The designed mRNA vaccine was estimated to have a molecular weight of 165023.50 Da and was highly soluble (grand average of hydropathicity of -0.440). In the structural assessment, the vaccine seemed to be a well-stable and functioning protein (Z score of -8.94). Also, the docking analysis suggested that the vaccine had a high affinity for TLR-2 and TLR-4 receptors. Additionally, the molecular mechanics with generalized Born and surface area solvation analysis of the "Vaccine-TLR-2" (-141.07 kcal/mol) and "Vaccine-TLR-4" (-271.72 kcal/mol) complexes also suggests a strong binding affinity for the receptors. Codon optimization also provided a high expression level with a GC content of 47.04% and a codon adaptation index score 1.0. The appearance of memory B-cells and T-cells was also observed over a while, with an increased level of helper T-cells and immunoglobulins (IgM and IgG). Moreover, the minimum free energy of the mRNA vaccine was predicted at -1760.00 kcal/mol, indicating the stability of the vaccine following its entry, transcription, and expression. This hypothetical vaccine offers a groundbreaking tool for future research and therapeutic development of pancreatic cancer.
Collapse
Affiliation(s)
- Md Habib Ullah Masum
- Department of Microbiology, Noakhali Science and Technology University, Noakhali, Bangladesh
- Microbiology, Cancer and Bioinformatics Research Group, Noakhali Science and Technology University, Noakhali, Bangladesh
| | - Shah Wajed
- Department of Microbiology, Noakhali Science and Technology University, Noakhali, Bangladesh
- Microbiology, Cancer and Bioinformatics Research Group, Noakhali Science and Technology University, Noakhali, Bangladesh
- Infectiology: Biology of Infectious Diseases, Universite Paris-Saclay, Gif-sur-Yvette, France
| | - Md Imam Hossain
- Department of Microbiology, Noakhali Science and Technology University, Noakhali, Bangladesh
| | - Nusrat Rahman Moumi
- Medical Sciences, University of Central Lancashire, Preston, Lancashire, United Kingdom
| | - Asma Talukder
- Microbiology, Cancer and Bioinformatics Research Group, Noakhali Science and Technology University, Noakhali, Bangladesh
- Department of Biotechnology and Genetic Engineering, Noakhali Science and Technology University, Noakhali, Bangladesh
- School of Pharmacy and Medical Sciences, and Menzies Health Institute Queensland, Griffith University, Brisbane, Queensland, Australia
| | - Md Mijanur Rahman
- Department of Microbiology, Noakhali Science and Technology University, Noakhali, Bangladesh
- Microbiology, Cancer and Bioinformatics Research Group, Noakhali Science and Technology University, Noakhali, Bangladesh
- School of Pharmacy and Medical Sciences, and Menzies Health Institute Queensland, Griffith University, Brisbane, Queensland, Australia
| |
Collapse
|
47
|
Sayour EJ, Boczkowski D, Mitchell DA, Nair SK. Cancer mRNA vaccines: clinical advances and future opportunities. Nat Rev Clin Oncol 2024; 21:489-500. [PMID: 38760500 DOI: 10.1038/s41571-024-00902-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/25/2024] [Indexed: 05/19/2024]
Abstract
mRNA vaccines have been revolutionary in terms of their rapid development and prevention of SARS-CoV-2 infections during the COVID-19 pandemic, and this technology has considerable potential for application to the treatment of cancer. Compared with traditional cancer vaccines based on proteins or peptides, mRNA vaccines reconcile the needs for both personalization and commercialization in a manner that is unique to each patient but not beholden to their HLA haplotype. A further advantage of mRNA vaccines is the availability of engineering strategies to improve their stability while retaining immunogenicity, enabling the induction of complementary innate and adaptive immune responses. Thus far, no mRNA-based cancer vaccines have received regulatory approval, although several phase I-II trials have yielded promising results, including in historically poorly immunogenic tumours. Furthermore, many early phase trials testing a wide range of vaccine designs are currently ongoing. In this Review, we describe the advantages of cancer mRNA vaccines and advances in clinical trials using both cell-based and nanoparticle-based delivery methods, with discussions of future combinations and iterations that might optimize the activity of these agents.
Collapse
Affiliation(s)
- Elias J Sayour
- Preston A. Wells Jr. Center for Brain Tumour Therapy, University of Florida, Gainesville, FL, USA
- Lillian S. Wells Department of Neurosurgery, University of Florida, Gainesville, FL, USA
| | - David Boczkowski
- Department of Surgery, Duke University School of Medicine, Durham, NC, USA
| | - Duane A Mitchell
- Preston A. Wells Jr. Center for Brain Tumour Therapy, University of Florida, Gainesville, FL, USA
- Lillian S. Wells Department of Neurosurgery, University of Florida, Gainesville, FL, USA
| | - Smita K Nair
- Department of Surgery, Duke University School of Medicine, Durham, NC, USA.
- Department of Neurosurgery, Duke University School of Medicine, Durham, NC, USA.
- Department of Pathology, Duke University School of Medicine, Durham, NC, USA.
- Duke Cancer Institute, Duke University School of Medicine, Durham, NC, USA.
| |
Collapse
|
48
|
Wang C, Yuan F. A comprehensive comparison of DNA and RNA vaccines. Adv Drug Deliv Rev 2024; 210:115340. [PMID: 38810703 PMCID: PMC11181159 DOI: 10.1016/j.addr.2024.115340] [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: 03/28/2024] [Revised: 05/06/2024] [Accepted: 05/18/2024] [Indexed: 05/31/2024]
Abstract
Nucleic acid technology has revolutionized vaccine development, enabling rapid design and production of RNA and DNA vaccines for prevention and treatment of diseases. The successful deployment of mRNA and plasmid DNA vaccines against COVID-19 has further validated the technology. At present, mRNA platform is prevailing due to its higher efficacy, while DNA platform is undergoing rapid evolution because it possesses unique advantages that can potentially overcome the problems associated with the mRNA platform. To help understand the recent performances of the two vaccine platforms and recognize their clinical potentials in the future, this review compares the advantages and drawbacks of mRNA and DNA vaccines that are currently known in the literature, in terms of development timeline, financial cost, ease of distribution, efficacy, safety, and regulatory approval of products. Additionally, the review discusses the ongoing clinical trials, strategies for improvement, and alternative designs of RNA and DNA platforms for vaccination.
Collapse
Affiliation(s)
- Chunxi Wang
- Department of Biomedical Engineering, Duke University, Durham, NC 27705, United States
| | - Fan Yuan
- Department of Biomedical Engineering, Duke University, Durham, NC 27705, United States.
| |
Collapse
|
49
|
Lin Y, Chen X, Wang K, Liang L, Zhang H. An Overview of Nanoparticle-Based Delivery Platforms for mRNA Vaccines for Treating Cancer. Vaccines (Basel) 2024; 12:727. [PMID: 39066365 PMCID: PMC11281455 DOI: 10.3390/vaccines12070727] [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: 05/10/2024] [Revised: 06/16/2024] [Accepted: 06/21/2024] [Indexed: 07/28/2024] Open
Abstract
With its unique properties and potential applications, nanoparticle-based delivery platforms for messenger RNA (mRNA) vaccines have gained significant attention in recent years. Nanoparticles have the advantages of enhancing immunogenicity, targeting delivery, and improving stability, providing a new solution for drug and vaccine delivery. In some clinical studies, a variety of nanoparticle delivery platforms have been gradually applied to a wide range of vaccine applications. Current research priorities are exploring various types of nanoparticles as vaccine delivery systems to enhance vaccine stability and immunogenicity. Lipid nanoparticles (LNPs) have shown promising potential in preclinical and clinical studies on the efficient delivery of antigens to immune cells. Moreover, lipid nanoparticles and other nanoparticles for nucleic acids, especially for mRNA delivery systems, have shown vast potential for vaccine development. In this review, we present various vaccine platforms with an emphasis on nanoparticles as mRNA vaccine delivery vehicles. We describe several novel nanoparticle delivery platforms for mRNA vaccines, such as lipid-, polymer-, and protein-based nanoparticles. In addition, we provide an overview of the anti-tumor immunity of nanovaccines against different tumors in cancer immunotherapy. Finally, we outline future perspectives and remaining challenges for this promising technology of nanoparticle-based delivery platforms for vaccines.
Collapse
Affiliation(s)
- Yang Lin
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China; (Y.L.); (X.C.); (K.W.)
- Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou 510515, China
| | - Xuehua Chen
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China; (Y.L.); (X.C.); (K.W.)
- Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou 510515, China
| | - Ke Wang
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China; (Y.L.); (X.C.); (K.W.)
| | - Li Liang
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China; (Y.L.); (X.C.); (K.W.)
- Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou 510515, China
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
- Jinfeng Laboratory, Chongqing Science and Technology Innovation Center, Chongqing 401329, China
| | - Hongxia Zhang
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China; (Y.L.); (X.C.); (K.W.)
- Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou 510515, China
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
50
|
Parveen A, Elkordy AA. Brief Insights into mRNA Vaccines: Their Successful Production and Nanoformulation for Effective Response against COVID-19 and Their Potential Success for Influenza A and B. Pathogens 2024; 13:500. [PMID: 38921798 PMCID: PMC11206352 DOI: 10.3390/pathogens13060500] [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: 05/01/2024] [Revised: 06/06/2024] [Accepted: 06/07/2024] [Indexed: 06/27/2024] Open
Abstract
A mRNA vaccine is a type of vaccine that induces an immune response. Antigen-encoding mRNA is delivered via vaccine carriers into the immune cells, which are produced because of antigen-encoding mRNA translation, a protein. For example, COVID-19 mRNA vaccines produce the spike protein of the COVID-19 virus, whereas for influenza virus, mRNA vaccines target the haemagglutinin protein to treat the flu, and it requires modifications depending on the pandemic or seasonal viruses as it is capable of adapting the immune response, which makes the development of vaccines arduous. The protein molecule promotes an adaptive immune response that eliminates and terminates the corresponding virus or pathogen. There are many challenges to delivering an mRNA vaccine into the body; hence, the encapsulation of the mRNA (usually within lipid nanoparticles) is necessary to protect the mRNA from the body's surrounding environment. In this review article, we focus mainly on the production, formulation, and stabilization of mRNA vaccines in general, elaborating more on and focusing more on SARS-CoV-2, or COVID-19, and influenza viruses, which have become a major concern as these viruses have turned into life-threatening diseases.
Collapse
Affiliation(s)
| | - Amal Ali Elkordy
- School of Pharmacy and Pharmaceutical Sciences, Faculty of Health Sciences and Wellbeing, University of Sunderland, Sunderland SR1 3SD, UK;
| |
Collapse
|