1
|
Sonar S, Das A, Kalele K, Subramaniyan V. Exosome-based cancer vaccine: a cell-free approach. Mol Biol Rep 2025; 52:421. [PMID: 40272645 DOI: 10.1007/s11033-025-10519-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Accepted: 04/15/2025] [Indexed: 05/04/2025]
|
2
|
Fines C, McCarthy H, Buckley N. The search for a TNBC vaccine: the guardian vaccine. Cancer Biol Ther 2025; 26:2472432. [PMID: 40089851 PMCID: PMC11913391 DOI: 10.1080/15384047.2025.2472432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Revised: 02/18/2025] [Accepted: 02/19/2025] [Indexed: 03/17/2025] Open
Abstract
Nearly 20 million people are diagnosed with cancer each year with breast cancer being the most common among women. Triple negative breast cancer (TNBC), defined by its no/low expression of ER and PR and lack of amplification of HER2, makes up 15-20% of all breast cancer cases. While patients overall have a higher response to chemotherapy, this subgroup is associated with the lowest survival rate indicating significant clinical and molecular heterogeneity demanding alternate treatment options. Therefore, new therapies have been explored, with a large focus on utilizing the immune system. A whole host of immunotherapies have been studied including immune checkpoint inhibitors, now standard of care for eligible patients, and possibly the most exciting and promising is that of a TNBC vaccine. While currently there are no approved TNBC vaccines, this review highlights many promising studies and points to an antigen, p53, which we believe is highly relevant for TNBC.
Collapse
Affiliation(s)
- Cory Fines
- School of Pharmacy, Queen’s University Belfast, Belfast, UK
| | - Helen McCarthy
- School of Pharmacy, Queen’s University Belfast, Belfast, UK
| | - Niamh Buckley
- School of Pharmacy, Queen’s University Belfast, Belfast, UK
| |
Collapse
|
3
|
Pojar K, Reckendorfer D, Strauss J, Szaffich S, Ahmadi-Erber S, Schippers T, Berraondo P, Orlinger KK, Raguz J, Lauterbach H. Combining local cytokine delivery and systemic immunization with recombinant artLCMV boosts antitumor efficacy in several preclinical tumor models. Oncoimmunology 2025; 14:2514040. [PMID: 40492380 DOI: 10.1080/2162402x.2025.2514040] [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: 03/12/2025] [Revised: 05/22/2025] [Accepted: 05/23/2025] [Indexed: 06/12/2025] Open
Abstract
Among the plethora of cancer immune evasion mechanisms, T-cell-inhibiting factors within the tumor microenvironment impose a major challenge for the development of novel immunotherapies. Strategies to overcome immunosuppression and remodel the TME are therefore urgently needed. Therapeutic cancer vaccines based on engineered arenaviruses have been proven to generate potent tumor specific CD8+ T-cell responses in preclinical models and cancer patients. Despite signs of clinical activity as monotherapy, combination therapies are needed to further increase the therapeutic effect. To address this need, we evaluated the efficacy of recombinant vectors based on lymphocytic choriomeningitis virus encoding the T-cell stimulating cytokines IL-7, IL-12 and IL-15 with or without tumor-associated antigens. These vectors were tested in three different mouse tumor models (TC-1, MC-38 and B16.F10). Our results demonstrate that only IL-12 encoding vectors led to increased immunogenicity and efficacy, which, after systemic administration, was associated with adverse events. The safest and most potent regimen consisted of systemic vaccination with tumor antigen encoding vectors and local injection of IL-12-encoding vectors. A single round of this treatment regimen resulted in 86-100% tumor-free mice and warrants further investigation.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Pedro Berraondo
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain
| | | | | | | |
Collapse
|
4
|
Zhang P, Pan J, Lin S, Peng B, An C, Zhang J, Xu L, Lai Y, Yu H, Xu Z. Smart drug delivery platforms reprogramming cancer immune cycle to mitigate immune resistance of pancreatic tumors. Adv Drug Deliv Rev 2025; 224:115620. [DOI: 10.1016/j.addr.2025.115620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/12/2025]
|
5
|
Dabo-Trubelja A. Considerations for Care of the Cancer Patient. Int Anesthesiol Clin 2025; 63:42-52. [PMID: 40353581 DOI: 10.1097/aia.0000000000000487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/14/2025]
Affiliation(s)
- Anahita Dabo-Trubelja
- Department of Anesthesiology and Critical Care, Director, Perioperative Point of Care Ultrasound, Memorial Sloan Kettering Cancer Center, New York
| |
Collapse
|
6
|
Tang Y, Yi X, Ai J. mRNA vaccines for prostate cancer: A novel promising immunotherapy. Biochim Biophys Acta Rev Cancer 2025; 1880:189333. [PMID: 40288658 DOI: 10.1016/j.bbcan.2025.189333] [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: 01/21/2025] [Revised: 04/21/2025] [Accepted: 04/21/2025] [Indexed: 04/29/2025]
Abstract
The treatment of advanced prostate cancer (PCa) primarily based on androgen deprivation therapy (ADT); however, patients inevitably progress to the castration-resistant prostate cancer (CRPC) stage. Despite the recent advancements in CRPC treatment with novel endocrine drugs that further inhibit androgen receptor signaling, resistance ultimately develops, underscoring the urgent need for new effective therapeutic strategies. Therapeutic cancer vaccines, a form of immunotherapy, exert anti-cancer effects by activating the host's immune system. Over the past few decades, various conventional therapeutic PCa vaccines based on cells, microbes, proteins, peptides, or DNA have been developed and tested in patients with advanced PCa. These attempts have largely failed to improve survival, with the sole exception of sipuleucel-T, which extended the median overall survival of asymptomatic or minimally symptomatic metastatic CRPC (mCRPC) patients by four months. The rapid development and high efficacy of mRNA vaccines during the COVID-19 pandemic have garnered worldwide attention. Compared to conventional vaccines, mRNA vaccines offer several unique advantages, including high production efficiency, low cost, high safety, strong immune response induction, and high adaptability and precision. These attributes make mRNA vaccines a promising frontier in the treatment of advanced PCa.
Collapse
Affiliation(s)
- Yaxiong Tang
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, 88 South Keyuan Road, Chengdu 610041, China
| | - Xianyanling Yi
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, 88 South Keyuan Road, Chengdu 610041, China
| | - Jianzhong Ai
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, 88 South Keyuan Road, Chengdu 610041, China.
| |
Collapse
|
7
|
Sultan MH, Zhan Q, Wang Y, Xia Y, Jia X. Precision oncolytic viral therapy in colorectal cancer: Genetic targeting and immune modulation for personalized treatment (Review). Int J Mol Med 2025; 56:104. [PMID: 40342021 PMCID: PMC12081034 DOI: 10.3892/ijmm.2025.5545] [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: 12/13/2024] [Accepted: 04/09/2025] [Indexed: 05/11/2025] Open
Abstract
Colorectal cancer (CRC) is a leading health issue and treatments to eradicate it, such as conventional chemotherapy, are non‑selective and come with a number of complications. The present review focuses on the relatively new area of precision oncolytic viral therapy (OVT), with genetic targeting and immune modifications that offer a new future for CRC treatment. In the present review, an overview of the selection factors that are considered optimal for an oncolytic virus, mechanisms of oncolysis and immunomodulation applied to the OVT, as well as new strategies to improve the efficacy of this method are described. Additionally, cause‑and‑effect relationships are examined for OVT efficacy, mediated by the tumor microenvironment, and directions for genetic manipulation of viral specificity are explored. The possibility of synergy between OVT and immune checkpoint inhibitors and other treatment approaches are demonstrated. Incorporating the details of the present review, biomarker‑guided combination therapies in precision OVT for individualized CRC care, significant issues and future trends in this required area of medicine are highlighted. Increasingly, OVT is leaving the experimental stage and may become routine practice; it provides a new perspective on overcoming CRC and highlights the importance of further research and clinical work.
Collapse
Affiliation(s)
- Muhammad Haris Sultan
- College of Life Sciences and Medicine, Xinyuan Institute of Medicine and Biotechnology, Zhejiang Sci-Tech University, Hangzhou, Zhejiang 310018, P.R. China
- Center for Translational Medicine and Precision Medicine, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325000, P.R. China
| | - Qi Zhan
- College of Life Sciences and Medicine, Xinyuan Institute of Medicine and Biotechnology, Zhejiang Sci-Tech University, Hangzhou, Zhejiang 310018, P.R. China
| | - Yigang Wang
- College of Life Sciences and Medicine, Xinyuan Institute of Medicine and Biotechnology, Zhejiang Sci-Tech University, Hangzhou, Zhejiang 310018, P.R. China
| | - Yulong Xia
- Center for Translational Medicine and Precision Medicine, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325000, P.R. China
| | - Xiaoyuan Jia
- College of Life Sciences and Medicine, Xinyuan Institute of Medicine and Biotechnology, Zhejiang Sci-Tech University, Hangzhou, Zhejiang 310018, P.R. China
| |
Collapse
|
8
|
Freitas R, Peixoto A, Santos LL, Ferreira JA. Glycan-based therapeutic approaches for bladder cancer: Overcoming clinical barriers. Biochim Biophys Acta Rev Cancer 2025; 1880:189327. [PMID: 40274080 DOI: 10.1016/j.bbcan.2025.189327] [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: 01/03/2025] [Revised: 04/15/2025] [Accepted: 04/16/2025] [Indexed: 04/26/2025]
Abstract
Bladder cancer (BLCA) remains a significant global health concern, being characterized by high incidence, recurrence, and mortality rates. Disease heterogeneity and rapid progression pose major challenges for effective management and identification of actionable biomarkers. Conventional therapies often fail to successfully achieve disease control, urging the development of novel, personalized approaches. In recent years, anti-tumour immunotherapy approaches in both pre-clinical and clinical settings have boomed. However, the efficacy of these strategies has been limited by the low mutational burden in some tumours, which hinders neoantigen presentation and the identification of BLCA-specific signatures. Cancer-associated aberrant glycosylation presents a unique opportunity for identifying BLCA-specific glycosignatures and developing innovative targeted therapeutics. This review provides a comprehensive overview of the clinical challenges in BLCA management and emerging novel therapies. Furthermore, it highlights the potential of glycosylation alterations as a unique opportunity for developing glycan-based therapies, potentially revolutionizing BLCA treatment strategies.
Collapse
Affiliation(s)
- Rui Freitas
- Experimental Pathology and Therapeutics Group, Research Center of IPO-Porto (CI-IPOP), 4200-072 Porto, Portugal; CI-IPOP@RISE (Health Research Network), Portuguese Oncology Institute of Porto (IPO-Porto)/Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC Raquel Seruca), 4200-072 Porto, Portugal; ICBAS - School of Medicine and Biomedical Sciences, University of Porto, 4050-313 Porto, Portugal; i3S - Institute for Research and Innovation in Health (i3S), University of Porto, 4200-135 Porto, Portugal
| | - Andreia Peixoto
- Experimental Pathology and Therapeutics Group, Research Center of IPO-Porto (CI-IPOP), 4200-072 Porto, Portugal; CI-IPOP@RISE (Health Research Network), Portuguese Oncology Institute of Porto (IPO-Porto)/Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC Raquel Seruca), 4200-072 Porto, Portugal; i3S - Institute for Research and Innovation in Health (i3S), University of Porto, 4200-135 Porto, Portugal
| | - Lúcio Lara Santos
- Experimental Pathology and Therapeutics Group, Research Center of IPO-Porto (CI-IPOP), 4200-072 Porto, Portugal; CI-IPOP@RISE (Health Research Network), Portuguese Oncology Institute of Porto (IPO-Porto)/Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC Raquel Seruca), 4200-072 Porto, Portugal; ICBAS - School of Medicine and Biomedical Sciences, University of Porto, 4050-313 Porto, Portugal; i3S - Institute for Research and Innovation in Health (i3S), University of Porto, 4200-135 Porto, Portugal; INEB - Institute for Biomedical Engineering, University of Porto, 4200-135 Porto, Portugal; Health School of University Fernando Pessoa, 4249-004 Porto, Portugal; Department of Surgical Oncology, Portuguese Oncology Institute of Porto (IPO Porto), 4200-072 Porto, Portugal; GlycoMatters Biotech, 4500-162 Espinho, Portugal
| | - José Alexandre Ferreira
- Experimental Pathology and Therapeutics Group, Research Center of IPO-Porto (CI-IPOP), 4200-072 Porto, Portugal; CI-IPOP@RISE (Health Research Network), Portuguese Oncology Institute of Porto (IPO-Porto)/Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC Raquel Seruca), 4200-072 Porto, Portugal; GlycoMatters Biotech, 4500-162 Espinho, Portugal..
| |
Collapse
|
9
|
Xie Y, Guo J, Hu J, Li Y, Zhang Z, Zhu Y, Deng F, Qi J, Zhou Y, Chen W. A factorial design-optimized microfluidic LNP vaccine elicits potent magnesium-adjuvating cancer immunotherapy. Mater Today Bio 2025; 32:101703. [PMID: 40230646 PMCID: PMC11994397 DOI: 10.1016/j.mtbio.2025.101703] [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: 11/13/2024] [Revised: 03/19/2025] [Accepted: 03/23/2025] [Indexed: 04/16/2025] Open
Abstract
Human papillomavirus (HPV)-associated cancers remain a critical health challenge, prompting the development of effective therapeutic vaccines. This study presents a lipid nanoparticle (LNP)-based vaccine co-loading E7 antigen peptide and magnesium ions as the adjuvant. Microfluidic technology was employed to optimize LNP preparation and formulation, ensuring efficient co-delivery of antigen and adjuvant. Magnesium ions were chosen over conventional aluminum-based adjuvants, which often suffer from limited efficacy and adverse effects, particularly for cancer immunotherapy. Compared to aluminum, magnesium ions exhibited superior capabilities in enhancing T-cell activation and promoting cellular immune response. Mechanistic insights suggest that magnesium ions facilitate dendritic cell maturation and antigen presentation via a collagen-CD36 axis, contributing to the adjuvant activity of magnesium. Through design of experiments (DoE) optimization, the LNP formulation was tailored for enhanced encapsulation and stability, positioning it as a targeted system for immune activation. These findings support the promise of magnesium ions as effective and safer adjuvants in LNP-based vaccines, marking a potential advancement for therapeutic cancer vaccination.
Collapse
Affiliation(s)
- Yongyi Xie
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, 511436, PR China
| | - Jiaxin Guo
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, 511436, PR China
| | - Jialin Hu
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, 511436, PR China
| | - Yuan Li
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, 511436, PR China
| | - Zhongqian Zhang
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, 511436, PR China
| | - Yongcheng Zhu
- Department of Emergency, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510260, PR China
| | - Fei Deng
- Graduate School of Biomedical Engineering, ARC Centre of Excellence in Nanoscale Biophotonics, Faculty of Engineering, UNSW Sydney, NSW, 2052, Australia
| | - Jialong Qi
- Yunnan Digestive Endoscopy Clinical Medical Center, Department of Gastroenterology, The First People's Hospital of Yunnan Province, Affiliated Hospital of Kunming University of Science and Technology, Kunming, 650032, PR China
| | - You Zhou
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, 511436, PR China
| | - Wenjie Chen
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, 511436, PR China
| |
Collapse
|
10
|
Chen E, Zhou W. Immunotherapy in microsatellite-stable colorectal cancer: Strategies to overcome resistance. Crit Rev Oncol Hematol 2025; 212:104775. [PMID: 40409481 DOI: 10.1016/j.critrevonc.2025.104775] [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: 02/18/2025] [Revised: 05/04/2025] [Accepted: 05/19/2025] [Indexed: 05/25/2025] Open
Abstract
Colorectal cancer (CRC) is among the foremost causes of cancer-related mortality worldwide; however, individuals with microsatellite-stable (MSS) disease-who constitute most CRC diagnoses-derive limited benefit from existing immunotherapeutic approaches. Here, we outline emerging methods designed to address the inherent resistance of MSS CRC to immune checkpoint inhibitors (ICIs). Recent findings emphasize how the immunosuppressive tumor microenvironment (TME) in MSS CRC, marked by diminished immunogenicity and high levels of regulatory T cells and myeloid-derived suppressor cells, restricts effective antitumor immune activity. Combination regimens that merge ICIs with chemotherapy, anti-angiogenic agents, or targeted blockade of pathways such as TGF-β and VEGF have shown encouraging early outcomes, including enhanced antigen presentation and T-cell penetration. Novel immunomodulatory platforms-such as epigenetic modifiers, oncolytic viruses, and engineered probiotic vaccines-are under assessment to further reprogram the TME and boost therapeutic efficacy. Concurrently, progress in adoptive cell therapies (for example, chimeric antigen receptor (CAR) T cells) and the development of cancer vaccines targeting tumor-associated and neoantigens promise to extend immune control over MSS CRC. In parallel, improving patient selection through predictive biomarkers-from circulating tumor DNA (ctDNA) to gene expression signatures and specific molecular subtypes-could refine individualized treatment strategies. Finally, interventions that alter the gut microbiome, including probiotics and fecal transplantation, serve as complementary tools to strengthen ICI responses. Taken together, these insights and combined treatment strategies lay the foundation for more successful immunotherapeutic interventions in MSS CRC, ultimately aiming to provide sustained clinical benefits to a broader spectrum of patients.
Collapse
Affiliation(s)
- Engeng Chen
- Department of Colorectal Surgery, Sir Run Run Shaw Hospital of Zhejiang University, Hangzhou 310016, China
| | - Wei Zhou
- Department of Colorectal Surgery, Sir Run Run Shaw Hospital of Zhejiang University, Hangzhou 310016, China.
| |
Collapse
|
11
|
Obata A, Katanosaka R, Taharabaru T, Arita-Morioka KI, Motozono C, Motoyama K, Higashi T. Cyclodextrin-Based Supramolecular Carrier for Cancer Proteinaceous Antigen. Mol Pharm 2025. [PMID: 40388578 DOI: 10.1021/acs.molpharmaceut.5c00115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/21/2025]
Abstract
Protein-based vaccines are gaining attention as a promising platform for vaccines because they are highly safe and induce humoral and cellular immunity. However, antigenic proteins have the disadvantages of low cell membrane permeability and easy degradability by endo/lysosomes. Therefore, the development of carriers that can overcome these challenges is essential. We recently developed a supramolecular carrier for the intracellular delivery of genome-editing molecules using cyclodextrin-based aminated polyrotaxanes (amino-PRX). The amino-PRX deformed its structure in response to the complicated shape and charge distribution of the genome-editing molecule, forming a complex with high efficiency. Moreover, by optimizing its structure, a second-generation amino-PRX (2G) possessing endosomal escape ability was constructed. Considering 2G deformability, it is applicable to antigenic proteins and could be an excellent antigen carrier. Therefore, here, we aimed to evaluate the utility of 2G as a protein-based cancer vaccine carrier. The results showed that 2G formed complexes with antigenic proteins efficiently. In addition, the 2G/antigenic protein complex activated immune cells with high efficiency and exhibited excellent antitumor effects. These results suggest that 2G is a promising protein-based cancer vaccine carrier.
Collapse
Affiliation(s)
- Airi Obata
- Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto 862-0973, Japan
| | - Reina Katanosaka
- Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto 862-0973, Japan
| | - Toru Taharabaru
- Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto 862-0973, Japan
| | - Ken-Ichi Arita-Morioka
- Kumamoto Innovative Development Organization, Kumamoto University, 2-39-1 Kurokami, Chuo-ku, Kumamoto 860-8555, Japan
| | - Chihiro Motozono
- Division of Infection and Immunity, Joint Research Center for Human Retrovirus Infection, Kumamoto University, 2-2-1 Honjo, Chuo-ku, Kumamoto 860-0811, Japan
| | - Keiichi Motoyama
- Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto 862-0973, Japan
| | - Taishi Higashi
- Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto 862-0973, Japan
| |
Collapse
|
12
|
Hushmandi K, Imani Fooladi AA, Reiter RJ, Farahani N, Liang L, Aref AR, Nabavi N, Alimohammadi M, Liu L, Sethi G. Next-generation immunotherapeutic approaches for blood cancers: Exploring the efficacy of CAR-T and cancer vaccines. Exp Hematol Oncol 2025; 14:75. [PMID: 40382583 DOI: 10.1186/s40164-025-00662-3] [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: 02/07/2025] [Accepted: 04/25/2025] [Indexed: 05/20/2025] Open
Abstract
Recent advancements in immunotherapy, particularly Chimeric antigen receptor (CAR)-T cell therapy and cancer vaccines, have significantly transformed the treatment landscape for leukemia. CAR-T cell therapy, initially promising in hematologic cancers, faces notable obstacles in solid tumors due to the complex and immunosuppressive tumor microenvironment. Challenges include the heterogeneous immune profiles of tumors, variability in antigen expression, difficulties in therapeutic delivery, T cell exhaustion, and reduced cytotoxic activity at the tumor site. Additionally, the physical barriers within tumors and the immunological camouflage used by cancer cells further complicate treatment efficacy. To overcome these hurdles, ongoing research explores the synergistic potential of combining CAR-T cell therapy with cancer vaccines and other therapeutic strategies such as checkpoint inhibitors and cytokine therapy. This review describes the various immunotherapeutic approaches targeting leukemia, emphasizing the roles and interplay of cancer vaccines and CAR-T cell therapy. In addition, by discussing how these therapies individually and collectively contribute to tumor regression, this article aims to highlight innovative treatment paradigms that could enhance clinical outcomes for leukemia patients. This integrative approach promises to pave the way for more effective and durable treatment strategies in the oncology field. These combined immunotherapeutic strategies hold great promise for achieving more complete and lasting remissions in leukemia patients. Future research should prioritize optimizing treatment sequencing, personalizing therapeutic combinations based on individual patient and tumor characteristics, and developing novel strategies to enhance T cell persistence and function within the tumor microenvironment. Ultimately, these efforts will advance the development of more effective and less toxic immunotherapeutic interventions, offering new hope for patients battling this challenging disease.
Collapse
Affiliation(s)
- Kiavash Hushmandi
- Nephrology and Urology Research Center, Clinical Sciences Institute, Baqiyatallah University of Medical Sciences, Tehran, Islamic Republic of Iran.
| | - Abbas Ali Imani Fooladi
- Applied Microbiology Research Center, Biomedicine Technologies Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Russel J Reiter
- Department of Cell Systems and Anatomy, UT Health San Antonio, San Antonio, TX, 78229, USA
| | - Najma Farahani
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Liping Liang
- Guangzhou Key Laboratory of Digestive Diseases, Department of Gastroenterology and Hepatology, Guangzhou Digestive Disease Center, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, 510180, China
| | - Amir Reza Aref
- Department of Vitro Vision, DeepkinetiX, Inc, Boston, MA, USA
| | | | - Mina Alimohammadi
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Le Liu
- Integrated Clinical Microecology Center, Shenzhen Hospital, Southern Medical University, Shenzhen, 518000, China.
- Department of Gastroenterology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China.
| | - Gautam Sethi
- Department of Pharmacology and NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore.
| |
Collapse
|
13
|
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
|
14
|
de Moura IA, Silva AJD, de Macêdo LS, de Melo KMTB, Leal LRS, Espinoza BCF, Invenção MDCV, de Pinho SS, de Freitas AC. Advances in the Functionalization of Vaccine Delivery Systems: Innovative Strategies and Translational Perspectives. Pharmaceutics 2025; 17:640. [PMID: 40430931 PMCID: PMC12115142 DOI: 10.3390/pharmaceutics17050640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2025] [Revised: 04/28/2025] [Accepted: 05/06/2025] [Indexed: 05/29/2025] Open
Abstract
The development of effective vaccines requires a rational design that considers the interaction between antigens, their vectors, and the immune system in addition to the activation of pathways that induce a safe and specific immune response. The efficacy of a vaccine formulation depends on the nature of the antigen, the protection offered by the delivery system, the ability to potentiate the immune response, and the precise release of the immunogen. Carrier systems such as lipid nanoparticles, polymers, exosomes, and microorganisms can be functionalized by chemical, physical, or biological methods to generate selective and improved biodistribution profiles. These methods enhance interaction with target cells, thereby improving immunological efficacy. The conjugation of specific ligands or the modification of parameters such as shape, charge, and size of vectors can enhance the specificity, stability, and efficiency of antigen transport to cellular compartments, thereby facilitating a robust immune response. This study examines modifications in vaccine delivery systems, focusing on biomolecules and physicochemical changes that enhance antigen presentation. Additionally, we examine innovative methods, including microneedles, electroporation, and needle-free systems that show potential for enhancing the immune response.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Antonio Carlos de Freitas
- Laboratory of Molecular Studies and Experimental Therapy—LEMTE, Department of Genetics, Federal University of Pernambuco, Avenida da Engenharia S/N, Recife 50740-600, Pernambuco, Brazil; (I.A.d.M.); (A.J.D.S.); (L.S.d.M.); (K.M.T.B.d.M.); (L.R.S.L.); (B.C.F.E.); (M.d.C.V.I.); (S.S.d.P.)
| |
Collapse
|
15
|
Yoon J, Moon H, Jeon Y, Choe S, Yoon H. Signature Gene Mutations in Colorectal Cancer: Potential Neoantigens for Cancer Vaccines. Int J Mol Sci 2025; 26:4559. [PMID: 40429703 PMCID: PMC12111162 DOI: 10.3390/ijms26104559] [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: 04/08/2025] [Revised: 05/07/2025] [Accepted: 05/08/2025] [Indexed: 05/29/2025] Open
Abstract
Colorectal cancer (CRC), the third most common cancer worldwide, is one of the deadliest cancers. CRC is known as a cold tumor, characterized by a low immune response that makes it difficult for immune cells to infiltrate and exhibits strong resistance to immunotherapy with checkpoint inhibition. This restricted response is largely attributed to signature gene mutations including mismatch repair (MMR) genes, KRAS, BRAF, APC, and TP53, which are also the main oncogenes in CRC. Mutated signature genes continuously upregulate abnormal signaling pathways, leading to excessive proliferation, cancer progression, and metastasis. Furthermore, it reorganizes the tumor microenvironment (TME) by recruiting immunosuppressive cells. However, the mutation can produce neoantigens that can provoke an immune response, making it a potential target for immunotherapy. In particular, cancer vaccines that leverage the strong neoantigenic properties of these mutations are considered promising for overcoming immune resistance and eliciting anti-tumor responses. In this review, we will describe signature gene mutations in CRC and focus on cancer vaccines targeting these mutations as potential therapies for CRC.
Collapse
Affiliation(s)
- Jaegoo Yoon
- Department of Medical and Biological Sciences, The Catholic University of Korea, Bucheon 14662, Republic of Korea; (J.Y.); (H.M.); (Y.J.); (S.C.)
| | - Haeun Moon
- Department of Medical and Biological Sciences, The Catholic University of Korea, Bucheon 14662, Republic of Korea; (J.Y.); (H.M.); (Y.J.); (S.C.)
| | - Yuna Jeon
- Department of Medical and Biological Sciences, The Catholic University of Korea, Bucheon 14662, Republic of Korea; (J.Y.); (H.M.); (Y.J.); (S.C.)
| | - Soohyun Choe
- Department of Medical and Biological Sciences, The Catholic University of Korea, Bucheon 14662, Republic of Korea; (J.Y.); (H.M.); (Y.J.); (S.C.)
- Department of Biotechnology, The Catholic University of Korea, Bucheon 14662, Republic of Korea
| | - Hyunho Yoon
- Department of Medical and Biological Sciences, The Catholic University of Korea, Bucheon 14662, Republic of Korea; (J.Y.); (H.M.); (Y.J.); (S.C.)
- Department of Biotechnology, The Catholic University of Korea, Bucheon 14662, Republic of Korea
| |
Collapse
|
16
|
Seta T, Nakamura S, Oura M, Yokoyama K, Nishikawa Y, Hoshino N, Ninomiya K, Shimoi T, Hotta K, Nakayama T. Efficacy and safety of cancer vaccine therapy in malignant melanoma: a systematic review. Int J Clin Oncol 2025:10.1007/s10147-025-02753-x. [PMID: 40329122 DOI: 10.1007/s10147-025-02753-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Accepted: 03/23/2025] [Indexed: 05/08/2025]
Abstract
INTRODUCTION Malignant melanoma is a cancer that develops from melanocytes in the skin and mucous membranes. Surgery, and chemotherapy, radiation therapy, or immunotherapy are also used in cases of distant metastasis. Immunotherapy includes immune checkpoint inhibitors and cancer vaccine therapy; however, their efficacy remains limited. METHODS A systematic review and meta-analysis of cancer vaccine therapies were conducted. PubMed was used to search the literature up to December 2023, and clinical trials were also identified for the same period. RCTs involving patients with resectable or unresectable malignant melanoma were included. The primary outcome was OS, and secondary outcomes were PFS, DFS, and RFS. Safety was assessed based on AEs. Integrated results were presented as risk ratio and risk difference. RESULTS The initial search identified 418 studies on cancer vaccine therapy and 44 studies on effector T-cell therapy. We supplemented this with our PubMed search, extracting 149 studies. After database searches and screening, 611 studies were initially considered. Following exclusions based on eligibility criteria, 20 RCTs using cancer vaccines remained. For the primary outcome, OS, the pooled RR was 1.11 (95% CI, 0.97-1.32, I2 = 59.0%), and the pooled RD was 0.02 (-0.01 to 0.06, I2 = 74.0%) at 12 months. PFS, DFS, and RFS did not show statistically significant differences, and AEs did not increase significantly. CONCLUSION Cancer vaccine therapy, neither alone, or in combination with other agents for malignant melanoma did not show a significant improvement in OS, PFS, DFS, or RFS nor did it significantly increase AEs.
Collapse
Affiliation(s)
- Takeshi Seta
- Department of Gastroenterology and Hepatology and Gastrointestinal Cancer Center, JRC Wakayama Medical Center, 4-20, Komatsubara-Dori, Wakayama City, Wakayama, 640-8558, Japan
| | - Shohei Nakamura
- Department of Medical Oncology, Tokyo Metropolitan Cancer and Infectious Diseases Center Komagome Hospital, 3-18-22 Honkomagome, Bunkyo-Ku, Tokyo, 113-8677, Japan
| | - Mitsuaki Oura
- Department of Hematology and Oncology, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-Ku, Tokyo, 113-8655, Japan
| | - Kazuki Yokoyama
- Department of Head and Neck, Esophageal Medical Oncology, National Cancer Center Hospital, 5-1-1, Tsukiji, Chuo-Ku, Tokyo, 104-0045, Japan
| | - Yoshitaka Nishikawa
- Department of Health Informatics, Kyoto University School of Public Health, Yoshidakonoe-Cho, Sakyo-Ku, Kyoto, 606-8501, Japan
- Takemi Program in International Health, Harvard T.H. Chan School of Public Health, 665 Huntington Avenue, Boston, MA, 02115, USA
| | - Nobuaki Hoshino
- Department of Surgery, Kyoto University Graduate School of Medicine, 54 Shogoin-Kawahara-Cho, Sakyo-Ku, Kyoto, 606-8507, Japan
| | - Kiichiro Ninomiya
- Center for Comprehensive Genomic Medicine, Okayama University Hospital, 2-5-1 Kitaka-Cho, Kita-Ku, Okayama, 700-8558, Japan
| | - Tatsunori Shimoi
- Department of Medical Oncology, National Cancer Center Hospital, 5-1-1, Tsukiji, Chuo-Ku, Tokyo, 104-0045, Japan.
| | - Katsuyuki Hotta
- Center for Innovative Clinical Medicine, Okayama University Hospital, Okayama, Japan
| | - Takeo Nakayama
- Department of Health Informatics, Kyoto University School of Public Health, Yoshidakonoe-Cho, Sakyo-Ku, Kyoto, 606-8501, Japan
| |
Collapse
|
17
|
Ying S, Liu H, Zhang Y, Mei Y. Harnessing Dendritic Cell Function in Hepatocellular Carcinoma: Advances in Immunotherapy and Therapeutic Strategies. Vaccines (Basel) 2025; 13:496. [PMID: 40432108 PMCID: PMC12115466 DOI: 10.3390/vaccines13050496] [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: 03/21/2025] [Revised: 04/29/2025] [Accepted: 05/01/2025] [Indexed: 05/29/2025] Open
Abstract
Hepatocellular carcinoma (HCC) is a major cause of cancer-related mortality worldwide. Conventional therapies are frequently limited by tumor heterogeneity and the immunosuppressive tumor microenvironment (TME). Dendritic cells (DCs), central to orchestrating antitumor immunity, have become key targets for HCC immunotherapy. This review examines the biological functions of DC subsets (cDC1, cDC2, pDC, and moDC) and their roles in initiating and modulating immune responses against HCC. We detail the mechanisms underlying DC impairment within the TME, including suppression by regulatory T cells (Tregs), myeloid-derived suppressor cells (MDSCs), tumor-associated macrophages (TAMs), and cancer-associated fibroblasts (CAFs). Additionally, we discuss novel DC-based therapeutic strategies, such as DC-based vaccines designed to enhance antigen presentation and T cell activation. Combining DC vaccines with immune checkpoint inhibitors (ICIs), including PD-1/PD-L1 and CTLA-4 blockers, demonstrates synergistic effects that can overcome immune evasion and improve clinical outcomes. Despite progress, challenges related to DC subset heterogeneity, TME complexity, and patient variability require the further optimization and personalization of DC-based therapies. Future research should focus on refining these strategies, leveraging advanced technologies like genomic profiling and artificial intelligence, to maximize therapeutic efficacy and revolutionize HCC treatment. By restoring DC function and reprogramming the TME, DC-based immunotherapy holds immense potential to transform the management of HCC and improve patient survival.
Collapse
Affiliation(s)
- Shiding Ying
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China;
| | - Haiyan Liu
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117545, Singapore;
- NUSMED Immunology Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117456, Singapore
- Immunology Programme, Life Science Institute, National University of Singapore, Singapore 117456, Singapore
| | - Yongliang Zhang
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117545, Singapore;
- NUSMED Immunology Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117456, Singapore
- Immunology Programme, Life Science Institute, National University of Singapore, Singapore 117456, Singapore
| | - Yu Mei
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117545, Singapore;
- NUSMED Immunology Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117456, Singapore
- Immunology Programme, Life Science Institute, National University of Singapore, Singapore 117456, Singapore
| |
Collapse
|
18
|
Gomari MM, Ghantabpour T, Pourgholam N, Rostami N, Hatfield SM, Namazifar F, Abkhiz S, Eslami SS, Ramezanpour M, Darestanifarahani M, Astsaturov I, Bencherif SA. Breaking barriers: Smart vaccine platforms for cancer immunomodulation. Cancer Commun (Lond) 2025; 45:529-571. [PMID: 39901621 PMCID: PMC12067400 DOI: 10.1002/cac2.70002] [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: 07/23/2024] [Revised: 01/18/2025] [Accepted: 01/21/2025] [Indexed: 02/05/2025] Open
Abstract
Despite significant advancements in cancer treatment, current therapies often fail to completely eradicate malignant cells. This shortfall underscores the urgent need to explore alternative approaches such as cancer vaccines. Leveraging the immune system's natural ability to target and kill cancer cells holds great therapeutic potential. However, the development of cancer vaccines is hindered by several challenges, including low stability, inadequate immune response activation, and the immunosuppressive tumor microenvironment, which limit their efficacy. Recent progress in various fields, such as click chemistry, nanotechnology, exosome engineering, and neoantigen design, offer innovative solutions to these challenges. These achievements have led to the emergence of smart vaccine platforms (SVPs), which integrate protective carriers for messenger ribonucleic acid (mRNA) with functionalization strategies to optimize targeted delivery. Click chemistry further enhances SVP performance by improving the encapsulation of mRNA antigens and facilitating their precise delivery to target cells. This review highlights the latest developments in SVP technologies for cancer therapy, exploring both their opportunities and challenges in advancing these transformative approaches.
Collapse
Affiliation(s)
- Mohammad Mahmoudi Gomari
- Department of Medical BiotechnologyFaculty of Allied MedicineIran University of Medical SciencesTehranIran
| | - Taha Ghantabpour
- Department of AnatomySchool of MedicineQazvin University of Medical SciencesQazvinIran
| | - Nima Pourgholam
- School of Nursing and MidwiferyIran University of Medical ScienceTehranIran
| | - Neda Rostami
- Department of Chemical EngineeringArak UniversityArakIran
| | - Stephen M. Hatfield
- New England Inflammation and Tissue Protection InstituteDepartment of Pharmaceutical SciencesNortheastern UniversityBostonMassachusettsUSA
- Department of Chemical EngineeringNortheastern UniversityBostonMassachusettsUSA
| | | | - Shadi Abkhiz
- Department of Medical BiotechnologyFaculty of Allied MedicineIran University of Medical SciencesTehranIran
| | - Seyed Sadegh Eslami
- Department of Medical BiotechnologyFaculty of Allied MedicineIran University of Medical SciencesTehranIran
- Molecular Proteomics LaboratoryBaker Heart and Diabetes InstituteMelbourneVictoriaAustralia
| | - Mahsa Ramezanpour
- Department of Medical BiotechnologyFaculty of Allied MedicineIran University of Medical SciencesTehranIran
| | | | - Igor Astsaturov
- Marvin and Concetta Greenberg Pancreatic Cancer InstituteFox Chase Cancer CenterPhiladelphiaPennsylvaniaUSA
| | - Sidi A. Bencherif
- Department of Chemical EngineeringNortheastern UniversityBostonMassachusettsUSA
- Harvard John A. Paulson School of Engineering and Applied SciencesHarvard UniversityCambridgeMassachusettsUSA
- Polymers, Biopolymers Surfaces (PBS) LaboratoryNational Center for Scientific Research (CNRS) Mixed Research Unit (UMR) 6270University Rouen NormandieRouenFrance
| |
Collapse
|
19
|
Zhao S, Qu Y, Sun Z, Zhang S, Xia M, Shi Y, Wang J, Wang Y, Zhong Z, Meng F. Glioblastoma Cell Lysate and Adjuvant Nanovaccines via Strategic Vaccination Completely Regress Established Murine Tumors. Adv Healthc Mater 2025; 14:e2500911. [PMID: 40270217 DOI: 10.1002/adhm.202500911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2025] [Revised: 04/02/2025] [Indexed: 04/25/2025]
Abstract
Tumor vaccines have shown great promise for treating various malignancies; however, glioblastoma (GBM), characterized by its immunosuppressive tumor microenvironment, high heterogeneity, and limited accessibility, has achieved only modest clinical benefits. Here, it is reported that GBM cell lysate nanovaccines boosted with TLR9 agonist CpG ODN (GlioVac) via a strategic vaccination regimen achieve complete regression of malignant murine GBM tumors. Subcutaneous administration of GlioVac promotes uptake by cervical lymph nodes and antigen presentation cells, bolstering antigen cross-presentation and infiltration of GBM-specific CD8+ T cells into the tumor. Notably, a regimen involving two subcutaneous and three intravenous vaccinations not only activates systemic anti-GBM immunity but also further enhances the tumor infiltration of cytotoxic T lymphocytes, effectively reshaping the "cold" GBM tumor into a "hot" tumor. This approach led to a state of tumor-free survival in 5 out of 7 mice bearing the established GL261 GBM model with complete protection from tumor rechallenge. In an orthotopic hRas-GBM model induced by a lentiviral plasmid, GlioVac resulted in ≈100% complete tumor regression. These findings suggest that GlioVac provides a personalized therapeutic vaccine strategy for glioblastoma.
Collapse
Affiliation(s)
- Songsong Zhao
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, 215123, P. R. China
| | - Yanyi Qu
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, 215123, P. R. China
| | - Zhiwei Sun
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, 215123, P. R. China
| | - Shuo Zhang
- College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, P. R. China
| | - Mingyu Xia
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, 215123, P. R. China
| | - Yan Shi
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, 215123, P. R. China
| | - Jingyi Wang
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, 215123, P. R. China
| | - Yuan Wang
- College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, P. R. China
| | - Zhiyuan Zhong
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, 215123, P. R. China
- College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, P. R. China
| | - Fenghua Meng
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, 215123, P. R. China
| |
Collapse
|
20
|
Zhang Y, Li Y, Xu Z, Xu L, Wang Y, Li N, Solek NC, Wang Y, Li B, Liu H. PPS-TLR7/8 agonist nanoparticles equip robust anticancer immunity by selectively prolonged activation of dendritic cells. Biomaterials 2025; 316:123032. [PMID: 39705927 DOI: 10.1016/j.biomaterials.2024.123032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 12/14/2024] [Accepted: 12/15/2024] [Indexed: 12/23/2024]
Abstract
Checkpoint inhibitor therapies do not benefit all patients, and adjuvants play a critical role in boosting immune responses for effective cancer immunotherapy. However, their systemic toxicity and suboptimal activation kinetics pose significant challenges. Here, this study presented a linker-based strategy to modulate the activation kinetics of Toll-like receptor 7/8 (TLR7/8) agonists delivered via poly (propylene sulfide) nanoparticles (PPS NPs). By covalently binding small molecule TLR7/8 agonists to PPS NPs with different linkers, enhanced therapeutic efficacy is achieved while abrogating systemic toxicity. These results showed that an alkyl linker selectively prolong the activation of DCs. It avoided the extensive activation of other APCs, favoring the limitation of immune-related toxicities. This strategy exhibited significant anti-tumor activity in alkyl linked nano-TLR7/8 agonists treatment alone, and cytokine and immune cell profiling provided evidence of prolonged immune cell activation in the tumor microenvironment, with evidence of an increase in the frequency of tumor antigen-specific CD8+ T cells. This linker-based approach offers a promising strategy to optimize the delivery of nano-TLR7/8 agonists for cancer immunotherapy, potentially advancing the field toward improved clinical outcomes.
Collapse
Affiliation(s)
- Yingxi Zhang
- Wuya college of innovation, Shenyang Pharmaceutical University, No.103, Wenhua Road, Shenyang, 110016, China
| | - Yicheng Li
- Wuya college of innovation, Shenyang Pharmaceutical University, No.103, Wenhua Road, Shenyang, 110016, China
| | - Zhaochu Xu
- Wuya college of innovation, Shenyang Pharmaceutical University, No.103, Wenhua Road, Shenyang, 110016, China
| | - Linyi Xu
- Wuya college of innovation, Shenyang Pharmaceutical University, No.103, Wenhua Road, Shenyang, 110016, China
| | - Yue Wang
- Wuya college of innovation, Shenyang Pharmaceutical University, No.103, Wenhua Road, Shenyang, 110016, China
| | - Ning Li
- Wuya college of innovation, Shenyang Pharmaceutical University, No.103, Wenhua Road, Shenyang, 110016, China
| | - Nicholas C Solek
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON, M5S 3M2, Canada; Institute of Biomedical Engineering, University of Toronto, Toronto, ON, M5S 3G9, Canada
| | - Yongjun Wang
- Wuya college of innovation, Shenyang Pharmaceutical University, No.103, Wenhua Road, Shenyang, 110016, China; Joint International Research Laboratory of Intelligent Drug Delivery Systems, Ministry of Education, Shenyang Pharmaceutical University, Shenyang, Liaoning, 110016, China.
| | - Bowen Li
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON, M5S 3M2, Canada; Institute of Biomedical Engineering, University of Toronto, Toronto, ON, M5S 3G9, Canada; Department of Chemistry, University of Toronto, Toronto, ON, M5S 3H6, Canada; Princess Margaret Cancer Center, University Health Network, Toronto, ON M5G 2C1, Canada.
| | - Hongzhuo Liu
- Wuya college of innovation, Shenyang Pharmaceutical University, No.103, Wenhua Road, Shenyang, 110016, China; Joint International Research Laboratory of Intelligent Drug Delivery Systems, Ministry of Education, Shenyang Pharmaceutical University, Shenyang, Liaoning, 110016, China.
| |
Collapse
|
21
|
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
|
22
|
Wang J, Chen Q, Shan Q, Liang T, Forde P, Zheng L. Clinical development of immuno-oncology therapeutics. Cancer Lett 2025; 617:217616. [PMID: 40054657 PMCID: PMC11930610 DOI: 10.1016/j.canlet.2025.217616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 03/03/2025] [Accepted: 03/05/2025] [Indexed: 03/15/2025]
Abstract
Immuno-oncology (IO) is one of the fastest growing therapeutic areas within oncology. IO agents work indirectly via the host's adaptive and innate immune system to recognize and eradicate tumor cells. Despite checkpoint inhibitors being only introduced to the market since 2011, they have become the second most approved product category. Current Food and Drug Administration (FDA)-approved classes of IO agents include: immune checkpoint inhibitors (ICIs), chimeric antigen receptor T-cell therapy (CAR-T), bi-specific T-cell engager (BiTE) antibody therapy, T-cell receptor (TCR) engineered T cell therapy, tumor-infiltrating lymphocyte (TIL) therapy, cytokine therapy, cancer vaccine therapy, and oncolytic virus therapy. Cancer immunotherapy has made progress in multiple cancer types including melanoma, non-small cell lung cancer (NSCLC), renal cell carcinoma (RCC), and urothelial carcinoma; however, several cancers remain refractory to immunotherapy. Future directions of IO include exploration in the neoadjuvant/perioperative setting, combination strategies, and optimizing patient selection through improved biomarkers.
Collapse
Affiliation(s)
- Jianxin Wang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China; Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China; Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic Diseases, Hangzhou, 310003, China; The Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province, Hangzhou, 310003, China
| | - Qi Chen
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China; Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China; Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic Diseases, Hangzhou, 310003, China; The Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province, Hangzhou, 310003, China
| | - Qiang Shan
- Department of General Surgery, Haining People's Hospital, Haining, 314400, China
| | - Tingbo Liang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China; Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China; Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic Diseases, Hangzhou, 310003, China; The Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province, Hangzhou, 310003, China
| | - Patrick Forde
- Department of Oncology and the Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, 1650 Orleans St, Baltimore, MD, 21287, USA; The Pancreatic Cancer Precision Medicine Center of Excellence Program, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA; The Bloomberg Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA; Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA; Mays Cancer Center at the University of Texas Health San Antonio, San Antonio, TX, 78229, USA
| | - Lei Zheng
- Department of Oncology and the Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, 1650 Orleans St, Baltimore, MD, 21287, USA; The Pancreatic Cancer Precision Medicine Center of Excellence Program, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA; The Bloomberg Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA; Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA; Mays Cancer Center at the University of Texas Health San Antonio, San Antonio, TX, 78229, USA.
| |
Collapse
|
23
|
Xia J, Chen X, Dong M, Liu S, Zhang L, Pan J, Wang J. Antigen self-presenting dendrosomes swallowing nanovaccines boost antigens and STING agonists codelivery for cancer immunotherapy. Biomaterials 2025; 316:122998. [PMID: 39657509 DOI: 10.1016/j.biomaterials.2024.122998] [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: 08/04/2024] [Revised: 11/24/2024] [Accepted: 12/04/2024] [Indexed: 12/12/2024]
Abstract
Cancer vaccines show promise by eliciting tumor-specific cytotoxic T lymphocytes (CTL) responses. Efficient cytosolic co-delivery of antigens and adjuvants to dendritic cells (DCs) is crucial for vaccines to induce anti-tumor immunity. However, peptide- or nucleic acid-based biomolecules like tumor antigens and STING agonist cyclic-di-GMP (cdGMP) are prone to endosomal degradation, resulting in low cytosolic delivery and CTL response rates. Cationic nanocarriers can improve cytosolic delivery, but their positive charges induce off-target effects. Here, we develop cationic poly(ester amide) based nanoparticles co-loaded with antigens and adjuvant cdGMP (NP(cG, OVA)) for efficient cytosolic delivery and swallow them within antigen self-presenting DCs-derived dendrosomes (ODs) for lymph nodes (LNs) homing. The constructed dendrosomes swallowing nanovaccines ODs/NP(cG, OVA) demonstrated significantly reduced liver accumulation and enhanced LNs and DCs targeting compared to NP(cG, OVA). ODs/NP(cG, OVA) effectively cross-dressed the antigen epitopes on the shell to DCs and facilitated internalization of NP(cG, OVA), realizing DCs cytosolic co-delivery of antigens and adjuvants, thereby promoting antigen presentation, maturation and inflammatory cytokines secretion of DCs. Consequently, DCs stimulated by ODs/NP(cG, OVA) effectively induced activation, proliferation, and differentiation of antigen-specific CTLs that provided robust immune protection against tumor invasion. This work presents a powerful vaccine strategy for cancer immunotherapy.
Collapse
Affiliation(s)
- Jiaxuan Xia
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai Institute of Infectious Disease and Biosecurity, Shanghai, 201203, China
| | - Xing Chen
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai Institute of Infectious Disease and Biosecurity, Shanghai, 201203, China
| | - Meichen Dong
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai Institute of Infectious Disease and Biosecurity, Shanghai, 201203, China
| | - Shengyao Liu
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai Institute of Infectious Disease and Biosecurity, Shanghai, 201203, China
| | - Longlong Zhang
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai Institute of Infectious Disease and Biosecurity, Shanghai, 201203, China
| | - Junjie Pan
- Department of Cardiology, Huashan Hospital, Fudan University, Shanghai, 200040, China.
| | - Jianxin Wang
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai Institute of Infectious Disease and Biosecurity, Shanghai, 201203, China; Quzhou Fudan Institute, Quzhou, Zhejiang Province, 324000, China.
| |
Collapse
|
24
|
Ghazizadeh Y, Salehi Shadkami H, Madani F, Niknam S, Adabi M. Advances in cancer nanovaccines: a focus on colorectal cancer. Nanomedicine (Lond) 2025; 20:1029-1041. [PMID: 40186876 PMCID: PMC12051617 DOI: 10.1080/17435889.2025.2486930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Accepted: 03/27/2025] [Indexed: 04/07/2025] Open
Abstract
Nanotechnology has revolutionized cancer treatment by providing innovative solutions through nanocancer therapies, nanovaccines, and nanoparticles. This review focuses on the application of these technologies in colorectal cancer (CRC), highlighting their progression from preclinical studies to clinical trials. Nanoparticles, including liposomes, silica, gold, and lipid nanoparticles, possess unique properties that enhance drug delivery, improve therapeutic efficacy, and minimize systemic toxicity. Additionally, nanovaccines are being developed to elicit robust immune responses against CRC cells. This paper offers a comprehensive overview of the current state of nanotechnology-based treatments for CRC, emphasizing key preclinical studies and clinical trials that demonstrate their potential. Furthermore, the review discusses the challenges faced in this field. It outlines future directions for research, underscoring the need for ongoing efforts to translate these promising technologies into practical clinical applications.
Collapse
Affiliation(s)
- Yalda Ghazizadeh
- Student Research Committee, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Nanomedicine Student Association (NMA), Student’s Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Hossein Salehi Shadkami
- Nanomedicine Student Association (NMA), Student’s Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran
- Department of Medical Science, Tehran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Madani
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Sedigheh Niknam
- Institute of Nano Science and Nano Technology, University of Kashan, Kashan, Iran
| | - Mahdi Adabi
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Food Microbiology Research Center, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
25
|
Li Y, Wang W, Xu J, Zhao B, Xiong L, Ge D, Wu Y, Dou X, Fu Y, Wang L, Zhao C, Chen M. Tumor signal amplification and immune decoy strategy using bacterial membrane-coated nanoparticles for immunotherapy. Biomater Sci 2025; 13:2368-2380. [PMID: 40104961 DOI: 10.1039/d4bm01535e] [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: 03/20/2025]
Abstract
In cancer therapy, tumor cells can diminish their signals through mechanisms such as immune escape, thereby evading recognition and elimination by the immune system. Providing tumor signals to enhance the recognition of tumor sites is considered a crucial approach in cancer treatment. Inspired by the decoy-induced directed feeding of fish, we propose a biomimetic nanoparticle system for tumor signal amplification. This biomimetic system comprises magnetically responsive nanoparticles and immune-inducing bacterial membranes. These designs work together to create a baiting effect at the tumor site, attracting and activating immune cells to attack. It has been demonstrated that the generated nanoparticles have the potential to be targeted and delivered to the tumor site under the influence of an external magnetic field, as demonstrated in preliminary in vitro and in vivo studies. Moreover, the nanoparticles utilize the bacterial membrane and cell membrane-translocated calreticulin to induce an immune response, simulating a decoy mechanism to recruit immune cells. The nanoparticles were proved to be effective in recruiting macrophages and neutrophils and reducing tumor size in animal experiments. These features make the nanoparticles an ideal candidate for treating tumors.
Collapse
Affiliation(s)
- Yifan Li
- Department of Gastroenterology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, 210008, China.
| | - Weiwei Wang
- Department of Gastroenterology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, 210008, China.
| | - Jiale Xu
- Department of Gastroenterology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, 210008, China.
| | - Bei Zhao
- Department of Gastroenterology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China.
| | - Longying Xiong
- Department of Gastroenterology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China.
| | - Dan Ge
- Department of Gastroenterology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China.
| | - Yanping Wu
- Department of Gastroenterology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China.
| | - Xiaotan Dou
- Department of Gastroenterology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China.
| | - Yuping Fu
- Department of Pharmacy, Zhangjiajie People's Hospital, Hunan, 427099, China
| | - Lei Wang
- Department of Gastroenterology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China.
| | - Cheng Zhao
- Department of Gastroenterology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China.
| | - Min Chen
- Department of Gastroenterology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, 210008, China.
- Department of Gastroenterology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China.
| |
Collapse
|
26
|
Tejedor-Tejada E, Gómez Pérez B, Lizeaga Cundin G. [Translated article] Messenger RNA as a new strategy against cancer. FARMACIA HOSPITALARIA 2025:S1130-6343(25)00042-X. [PMID: 40307087 DOI: 10.1016/j.farma.2025.03.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Accepted: 10/14/2024] [Indexed: 05/02/2025] Open
|
27
|
Chen M, Ji S, Liu X, Zheng X, Zhou M, Wang W. Porphyrins and Their Derivatives in Cancer Therapy: Current Advances, Mechanistic Insights, and Prospective Directions. Mol Pharm 2025. [PMID: 40294305 DOI: 10.1021/acs.molpharmaceut.4c01359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
Porphyrin and its derivatives are widely used in cancer therapy due to their strong photon absorption capabilities and moderate light stability. Due to their hydrophobic nature, porphyrins with tetrapyrrolic macrocycles ease self-aggregation in physiological conditions. Instead, exploiting the C4 symmetry structure for self-assembly is beneficial to improve the bioavailability of porphyrin and its derivatives. Herein, this Review outlines porphyrin-based nanoformulations for therapeutic applications in cancer treatment. The typical pharmaceutical application of the integrated porphyrinic structure is systematically summarized, focusing on the typical synthetic methodologies and structure-functionality relationship. Additionally, therapeutic modalities (e.g., photothermal, photodynamic, and sonodynamic) and their synergy mechanism in regulated cell death are overviewed. Special attention is given to emerging technologies in nanocatalytic therapy, therapeutic vaccines, and proteolysis-targeting chimeras, which align with the trend toward personalization and minimal invasiveness in healthcare. Finally, we discuss the challenges and limitations of porphyrinic nanoformulations and explore their future directions in the healthcare sector, aiming to bridge the gap between research and practical clinical application.
Collapse
Affiliation(s)
- Min Chen
- School of Pharmacy, Nantong University, Nantong, Jiangsu 226001, China
| | - Shuying Ji
- School of Pharmacy, Nantong University, Nantong, Jiangsu 226001, China
| | - Xingxing Liu
- School of Pharmacy, Nantong University, Nantong, Jiangsu 226001, China
| | - Xiaohua Zheng
- School of Pharmacy, Nantong University, Nantong, Jiangsu 226001, China
| | - Mengjiao Zhou
- School of Pharmacy, Nantong University, Nantong, Jiangsu 226001, China
| | - Weiqi Wang
- School of Pharmacy, Nantong University, Nantong, Jiangsu 226001, China
| |
Collapse
|
28
|
Li H, Min L, Du H, Wei X, Tong A. Cancer mRNA vaccines: clinical application progress and challenges. Cancer Lett 2025; 625:217752. [PMID: 40306545 DOI: 10.1016/j.canlet.2025.217752] [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: 04/13/2025] [Accepted: 04/26/2025] [Indexed: 05/02/2025]
Abstract
Messenger RNA (mRNA) vaccines have emerged as one of the most promising and rapidly evolving immunotherapeutic approaches due to their ease of production, demonstrated clinical efficacy, and high safety. The coronavirus disease 2019(COVID-19) pandemic has showcased the remarkable therapeutic potential of mRNA vaccines, prompting researchers to explore their use for cancer treatment. Preclinical studies and human clinical trials have indicated their substantial clinical applicability. However, current research faces several challenges, including the complexity of tumor antigen selection, vaccine stability, and the development of resistance. This review summarizes the optimization strategies for cancer mRNA vaccines in preclinical settings, the progress of clinical trials, and the challenges encountered while analyzing various delivery vehicle types, infusion methods, and application cases across different cancer types, highlighting key factors in vaccine design. The findings demonstrate that mRNA vaccines elicit specific immune responses and exhibit favorable safety and tolerability in clinical trials. Moreover, developing personalized neoantigen vaccines offers a novel direction for cancer immunotherapy. The unique contribution of this review lies in its comprehensive overview of the latest advancements in therapeutic mRNA vaccines for cancer treatment while identifying critical areas for future research to propel the field forward.
Collapse
Affiliation(s)
- Hang Li
- State Key Laboratory of Biotherapy and Cancer Center, Research Unit of Gene and Immunotherapy, Chinese Academy of Medical Sciences, Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan Province, China
| | - Lang Min
- Department of Hematology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Haotian Du
- State Key Laboratory of Biotherapy and Cancer Center, Research Unit of Gene and Immunotherapy, Chinese Academy of Medical Sciences, Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan Province, China
| | - Xiawei Wei
- Laboratory of Aging Research and Cancer Drug Target, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Aiping Tong
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
29
|
Jwo SH, Ng SK, Li CT, Chen SP, Chen LY, Liu PJ, Wang HJ, Lin JS, Ko CJ, Lee CF, Wang CH, Ouyang X, Wang L, Wei TT. Dual prophylactic and therapeutic potential of iPSC-based vaccines and neoantigen discovery in colorectal cancer. Theranostics 2025; 15:5890-5908. [PMID: 40365296 PMCID: PMC12068288 DOI: 10.7150/thno.111400] [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: 02/01/2025] [Accepted: 04/11/2025] [Indexed: 05/15/2025] Open
Abstract
Rationale: Induced pluripotent stem cells (iPSCs) share transcriptomic similarities with cancer cells and express tumor-specific and tumor-associated antigens, highlighting their potential as cancer vaccines. Our previous study demonstrated that an iPSC-based vaccine effectively prevented tumor growth in various mouse models, including melanoma, breast, lung, and pancreatic cancers. However, the underlying mechanisms and the therapeutic efficacy of the iPSC-based vaccine remain unclear. Colorectal cancer (CRC), the third most common cancer with a rising incidence worldwide, presents an urgent need for novel strategies to prevent and treat CRC. Methods: Allograft mouse models were established to evaluate the antitumor effects of the iPSC-based vaccine. CpG oligonucleotide (ODN) 1826 served as a vaccine adjuvant. Bulk RNA-Sequencing (RNA-Seq) and the Microenvironment Cell Population counter (MCP-Counter) algorithm were performed to analyze transcriptomic changes. Liquid chromatography-mass spectrometry (LC-MS) combined with in silico strategies was employed to identify potential antigen proteins. Chinese Hamster Ovary (CHO-K1) models were utilized to express candidate neoantigen proteins. Mouse bone marrow-derived dendritic cells (BMDCs) were used to investigate T cell priming in response to iPSC-associated proteins. Immune cell profiles were characterized by flow cytometry. Results: The combination of CpG and iPSC vaccination demonstrated both prophylactic and therapeutic efficacy in reducing tumor growth in CRC mouse models. Vaccination significantly increased CD8+ T cell infiltration within tumor regions, while T cell depletion abrogated the antitumor effects, underscoring the critical role of T cells in mediating these responses. Proteomic analysis identified two iPSC-associated proteins, heterogeneous nuclear ribonucleoprotein U (HNRNPU) and nucleolin (NCL), as key drivers of the observed immune responses. Vaccination with HNRNPU or NCL, in combination with CpG, enhanced dendritic cell activation, induced antigen-specific CD8+ T cell cytotoxicity, and promoted the formation of central memory CD8+ T cells, collectively leading to significant CRC tumor shrinkage. Conclusions: Our findings reveal potential mechanisms underlying the efficacy of iPSC-based vaccines in cancer immunotherapy. Additionally, HNRNPU and NCL were identified as key antigen proteins in iPSC, demonstrating promise for the development of peptide-based vaccines for both the prevention and treatment of CRC.
Collapse
Affiliation(s)
- Si-Han Jwo
- Department and Graduate Institute of Pharmacology, College of Medicine, National Taiwan University, Taipei 10051, Taiwan
| | - Shang-Kok Ng
- Department and Graduate Institute of Pharmacology, College of Medicine, National Taiwan University, Taipei 10051, Taiwan
| | - Chin-Tzu Li
- Department and Graduate Institute of Pharmacology, College of Medicine, National Taiwan University, Taipei 10051, Taiwan
| | - Shao-Peng Chen
- Department and Graduate Institute of Pharmacology, College of Medicine, National Taiwan University, Taipei 10051, Taiwan
| | - Li-Yu Chen
- Department and Graduate Institute of Pharmacology, College of Medicine, National Taiwan University, Taipei 10051, Taiwan
| | - Pin-Jung Liu
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei 11031, Taiwan
| | - Huai-Jie Wang
- Department and Graduate Institute of Pharmacology, College of Medicine, National Taiwan University, Taipei 10051, Taiwan
| | - Jr-Shiuan Lin
- Graduate Institute of Immunology, College of Medicine, National Taiwan University, Taipei 10051, Taiwan
| | - Chun-Jung Ko
- Graduate Institute of Immunology, College of Medicine, National Taiwan University, Taipei 10051, Taiwan
| | - Cheng-Fan Lee
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Chun-Hao Wang
- Department of Internal Medicine, National Taiwan University Hospital, Taipei 100225, Taiwan
| | - Xiaoming Ouyang
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA 94305, USA
| | - Lin Wang
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA 94305, USA
| | - Tzu-Tang Wei
- Department and Graduate Institute of Pharmacology, College of Medicine, National Taiwan University, Taipei 10051, Taiwan
- Chemical Biology and Molecular Biophysics, Taiwan International Graduate Program in Chemical Biology and Molecular Biophysics (TIGP-CBMB), Academia Sinica, Taipei 11529, Taiwan
| |
Collapse
|
30
|
Naffaa MM, Al-Ewaidat OA, Gogia S, Begiashvili V. Neoantigen-based immunotherapy: advancing precision medicine in cancer and glioblastoma treatment through discovery and innovation. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2025; 6:1002313. [PMID: 40309350 PMCID: PMC12040680 DOI: 10.37349/etat.2025.1002313] [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: 01/29/2025] [Accepted: 04/07/2025] [Indexed: 05/02/2025] Open
Abstract
Neoantigen-based immunotherapy has emerged as a transformative approach in cancer treatment, offering precision medicine strategies that target tumor-specific antigens derived from genetic, transcriptomic, and proteomic alterations unique to cancer cells. These neoantigens serve as highly specific targets for personalized therapies, promising more effective and tailored treatments. The aim of this article is to explore the advances in neoantigen-based therapies, highlighting successful treatments such as vaccines, tumor-infiltrating lymphocyte (TIL) therapy, T-cell receptor-engineered T cells therapy (TCR-T), and chimeric antigen receptor T cells therapy (CAR-T), particularly in cancer types like glioblastoma (GBM). Advances in technologies such as next-generation sequencing, RNA-based platforms, and CRISPR gene editing have accelerated the identification and validation of neoantigens, moving them closer to clinical application. Despite promising results, challenges such as tumor heterogeneity, immune evasion, and resistance mechanisms persist. The integration of AI-driven tools and multi-omic data has refined neoantigen discovery, while combination therapies are being developed to address issues like immune suppression and scalability. Additionally, the article discusses the ongoing development of personalized immunotherapies targeting tumor mutations, emphasizing the need for continued collaboration between computational and experimental approaches. Ultimately, the integration of cutting-edge technologies in neoantigen research holds the potential to revolutionize cancer care, offering hope for more effective and targeted treatments.
Collapse
Affiliation(s)
- Moawiah M Naffaa
- Department of Psychology and Neuroscience, Duke University, Durham, NC 27708, USA
- Department of Cell Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Ola A Al-Ewaidat
- Department of Internal Medicine, Ascension Saint Francis Hospital, Evanston, IL 60202, USA
| | - Sopiko Gogia
- Department of Internal Medicine, Ascension Saint Francis Hospital, Evanston, IL 60202, USA
| | - Valiko Begiashvili
- Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS 66103, USA
| |
Collapse
|
31
|
Hussain A, Moxley-Wyles B, Bryan M, Gordon-Weeks A, Al-Obaidi I, Sandhu C, Lee L. Cancer vaccine trial evaluations: immunobridging and potential immunological endpoints. IMMUNOTHERAPY ADVANCES 2025; 5:ltaf016. [PMID: 40438385 PMCID: PMC12116883 DOI: 10.1093/immadv/ltaf016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Accepted: 04/12/2025] [Indexed: 06/01/2025] Open
Abstract
Therapeutic cancer vaccines are an emerging class of immunotherapy, but challenges remain in effectively adapting approved vaccines to a growing number of adjuvants, combination therapies, and antigen-selection methods. Phase III clinical trials remain the gold standard in determining clinical benefit, but are slow and resource intensive, whilst radiological surrogates often fail to reliably predict clinical benefit. Using immunological surrogates of efficacy, deployed in 'immunobridging trials', could present a viable alternative, safely speeding up cancer vaccine development in a cost-effective manner. Whilst this approach has proven successful in infectious disease vaccines, identifying reliable immunological correlates of protection has proven difficult for cancer vaccines. Most clinical trials, which present the richest source of data to establish a correlate, rely on peripheral blood samples and standard immunoassays that are ill-equipped to capture the complexity of the vaccine-induced anti-tumour response. This review is the first to outline the importance and challenges of establishing immunological surrogates for cancer vaccines in the context of immunobridging trials, evaluating current immunoassay methods, and highlighting the need for techniques that can characterize tumour-infiltrating lymphocytes and the suppressive tumour microenvironment across a range of patients. The authors propose adapting trial designs for surrogate discovery, including combining phase I/II trials and the use of multi-omics approaches. Successful immunological surrogate development could enable future immunobridging trials to accelerate the optimization of approved cancer vaccines without requiring new phase III trials, promoting faster clinical implementation of scientific advances and patient benefits.
Collapse
Affiliation(s)
- Ahmed Hussain
- Medical Sciences Division, University of Oxford, Oxford, United Kingdom
| | - Benjamin Moxley-Wyles
- Department of Cellular Pathology, Oxford University Hospitals NHS Foundation Trust, University of Oxford, Oxford, United Kingdom
| | - Michael Bryan
- Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Alex Gordon-Weeks
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, United Kingdom
| | - Ibrahem Al-Obaidi
- Medical Sciences Division, University of Oxford, Oxford, United Kingdom
| | - Ciaran Sandhu
- Medical Sciences Division, University of Oxford, Oxford, United Kingdom
| | - Lennard Lee
- Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
32
|
Xue F, Liu J, Wu J, Li X, Zhu N, Tang S, Zhang M, Duan H, Wang R, Zhang J. Combination therapy with alisertib enhances the anti-tumor immunity induced by a liver cancer vaccine. iScience 2025; 28:112120. [PMID: 40230537 PMCID: PMC11995041 DOI: 10.1016/j.isci.2025.112120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 12/26/2024] [Accepted: 02/24/2025] [Indexed: 04/16/2025] Open
Abstract
Alisertib is a potent aurora A kinase inhibitor in clinical trials for cancer treatment, but its efficacy on cancer vaccines remains unclear. Here, we developed a DNA vaccine targeting glypican-3 (pGPC3) and evaluated its efficacy with alisertib in hepatocellular carcinoma (HCC) models. The combination therapy of pGPC3 vaccine and alisertib significantly inhibited subcutaneous tumor growth, enhanced the induction and maturation of CD11c+ and CD8+CD11c+ dendritic cells (DCs), and expanded tumor-specific CD8+ T cell responses. CD8+ T cell depletion abolished the anti-tumor effects, underscoring the essential role of functional CD8+ T cell responses. Moreover, the combined treatment promoted memory CD8+ T cell induction, providing long-term protection. In liver orthotopic tumor models, the combination of pGPC3 vaccine and alisertib demonstrated potent therapeutic efficacy through CD8+ T cell responses. These results indicate that alisertib enhances the pGPC3 vaccine's therapeutic effect, offering a promising strategy for HCC treatment.
Collapse
Affiliation(s)
- Fang Xue
- School of Nursing, Bengbu Medical University, Bengbu, Anhui 233030, China
| | - Jing Liu
- Surgical Oncology, First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui 233004, China
| | - Jinyan Wu
- School of Nursing, Bengbu Medical University, Bengbu, Anhui 233030, China
| | - Xueer Li
- School of Nursing, Bengbu Medical University, Bengbu, Anhui 233030, China
| | - Ningning Zhu
- School of Nursing, Bengbu Medical University, Bengbu, Anhui 233030, China
| | - Shuting Tang
- School of Nursing, Bengbu Medical University, Bengbu, Anhui 233030, China
| | - Mengli Zhang
- School of Nursing, Bengbu Medical University, Bengbu, Anhui 233030, China
| | - Huan Duan
- Intensive Care Unit, First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui 233004, China
| | - Rui Wang
- Department of Medical Oncology, First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui 233004, China
- Anhui Provincial Key Laboratory of Tumor Evolution and Intelligent Diagnosis and Treatment, Bengbu Medical University, Bengbu 233004, Anhui, China
| | - Jing Zhang
- School of Mental Health, Bengbu Medical University, Bengbu, Anhui 233030, China
| |
Collapse
|
33
|
Deng Y, Hui Z, Liu L, Gao F, Chen X, Huang L, Liu S, Yin Y, Zhang Y, Peng S, Chen X, Chen D, Yuan Y, Chen J. Modular assembly and evaluation of a TROP2-targeting immunotoxin for cancer therapy. Int J Biol Macromol 2025; 310:143311. [PMID: 40253026 DOI: 10.1016/j.ijbiomac.2025.143311] [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: 01/27/2025] [Revised: 04/15/2025] [Accepted: 04/16/2025] [Indexed: 04/21/2025]
Abstract
Trophoblast cell surface antigen 2 (TROP2) is a transmembrane glycoprotein overexpressed in various cancers and associated with poor prognosis. Despite the success of TROP2-targeting antibody-drug conjugates (ADCs), challenges such as high molecular weight, linker instability, and complex manufacturing limit their broader application. To address these limitations, we developed a novel immunotoxin, Fab-PE24, by conjugating the Fab fragment of a humanized anti-TROP2 monoclonal antibody with a truncated Pseudomonas aeruginosa exotoxin A (PE24KDEL) using SpyCatcher-SpyTag technology. Fab-PE24 demonstrated high binding affinity to both recombinant and native TROP2 antigens, efficient internalization, and potent cytotoxicity, inducing apoptosis and inhibiting proliferation in TROP2-positive cancer cells. In vivo, Fab-PE24 significantly suppressed tumor growth in gastric and lung cancer xenograft models. Notably, Fab-PE24 exhibited synergistic antitumor effects when combined with cisplatin, suggesting its potential for overcoming chemotherapy resistance. This study presents a novel modular approach for constructing PE-based immunotoxins and highlights Fab-PE24 as a promising therapeutic candidate for TROP2-positive malignancies.
Collapse
Affiliation(s)
- Yajie Deng
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Zi Hui
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Lu Liu
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Fei Gao
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Xuan Chen
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Lulu Huang
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Shuaiyuan Liu
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Yu Yin
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Yu Zhang
- College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Nanjing 211816, Jiangsu, China
| | - Sen Peng
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Xuting Chen
- Department of Neonatology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Daijie Chen
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Yunsheng Yuan
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China.
| | - Junsheng Chen
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China.
| |
Collapse
|
34
|
Yang H, Feng R, Heng X, Shan F, Wang Y, Yao L, Wang S, Chen G, Chen H. Enhanced Whole Tumor Cell-Based Vaccines by a RAFT and Protein Fusion Strategy for Tumor Immunotherapy. Biomacromolecules 2025; 26:2690-2699. [PMID: 40117506 DOI: 10.1021/acs.biomac.5c00115] [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: 03/23/2025]
Abstract
Inactivated whole tumor cell-based vaccines (WTVs) are a promising strategy for tumor immunotherapy, but have exhibited limited antitumor effects clinically. Aiming at constructing enhanced WTVs, we developed glycopolymer-engineered WTVs (G-WTVs) using a Halo-Tag protein (HTP) fusion technique and reversible addition-fragmentation chain transfer (RAFT) polymerization. In our study, G-WTVs with varying molecular weights of glycopolymers were constructed. Compared to unmodified tumor cells, all G-WTVs effectively induced the polarization of macrophages toward the M1 phenotype and promoted the secretion of pro-inflammatory cytokines. This enhanced immune response was attributed to the improved interactions between G-WTVs and the macrophages. Among the G-WTVs, the medium molecular weight variant demonstrated the most pronounced enhancement of antitumor immune responses. Notably, the administration of optimized G-WTVs effectively inhibited the growth of B16 melanoma in mice. Our findings provide a new approach to enhance the antitumor efficacy of WTVs via cell membrane glycopolymer engineering, offering a promising strategy for tumor immunotherapy.
Collapse
Affiliation(s)
- He Yang
- State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials College of Chemistry, Chemical Engineering and Materials Science Soochow University, 199 Ren'ai Road, Suzhou, Jiangsu 215123, P. R. China
| | - Ruyan Feng
- School of Chemical and Environmental Engineering, Hunan Institute of Technology, Hengyang 421002, P. R. China
| | - Xingyu Heng
- State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials College of Chemistry, Chemical Engineering and Materials Science Soochow University, 199 Ren'ai Road, Suzhou, Jiangsu 215123, P. R. China
| | - Fangjian Shan
- State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials College of Chemistry, Chemical Engineering and Materials Science Soochow University, 199 Ren'ai Road, Suzhou, Jiangsu 215123, P. R. China
| | - Yichen Wang
- State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials College of Chemistry, Chemical Engineering and Materials Science Soochow University, 199 Ren'ai Road, Suzhou, Jiangsu 215123, P. R. China
| | - Lihua Yao
- State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials College of Chemistry, Chemical Engineering and Materials Science Soochow University, 199 Ren'ai Road, Suzhou, Jiangsu 215123, P. R. China
| | - Sujian Wang
- State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials College of Chemistry, Chemical Engineering and Materials Science Soochow University, 199 Ren'ai Road, Suzhou, Jiangsu 215123, P. R. China
| | - Gaojian Chen
- State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials College of Chemistry, Chemical Engineering and Materials Science Soochow University, 199 Ren'ai Road, Suzhou, Jiangsu 215123, P. R. China
| | - Hong Chen
- State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials College of Chemistry, Chemical Engineering and Materials Science Soochow University, 199 Ren'ai Road, Suzhou, Jiangsu 215123, P. R. China
- Jiangsu Biosurf Biotech Co., Ltd., Suzhou 215123, P. R. China
| |
Collapse
|
35
|
Notaro M, Borghetti M, Bresesti C, Giacca G, Kerzel T, Mercado CM, Beretta S, Monti M, Merelli I, Iaia S, Genua M, Annoni A, Canu T, Cristofori P, Degl'Innocenti S, Sanvito F, Rancoita PMV, Ostuni R, Gregori S, Naldini L, Squadrito ML. In vivo armed macrophages curb liver metastasis through tumor-reactive T-cell rejuvenation. Nat Commun 2025; 16:3471. [PMID: 40216735 PMCID: PMC11992024 DOI: 10.1038/s41467-025-58369-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Accepted: 03/18/2025] [Indexed: 04/14/2025] Open
Abstract
Despite recent progress in cancer treatment, liver metastases persist as an unmet clinical need. Here, we show that arming liver and tumor-associated macrophages in vivo to co-express tumor antigens (TAs), IFNα, and IL-12 unleashes robust anti-tumor immune responses, leading to the regression of liver metastases. Mechanistically, in vivo armed macrophages expand tumor reactive CD8+ T cells, which acquire features of progenitor exhausted T cells and kill cancer cells independently of CD4+ T cell help. IFNα and IL-12 produced by armed macrophages reprogram antigen presenting cells and rewire cellular interactions, rescuing tumor reactive T cell functions. In vivo armed macrophages trigger anti-tumor immunity in distinct liver metastasis mouse models of colorectal cancer and melanoma, expressing either surrogate tumor antigens, naturally occurring neoantigens or tumor-associated antigens. Altogether, our findings support the translational potential of in vivo armed liver macrophages to expand and rejuvenate tumor reactive T cells for the treatment of liver metastases.
Collapse
Affiliation(s)
- Marco Notaro
- Vector Engineering and In vivo Tumor Targeting Unit, San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Maristella Borghetti
- Vector Engineering and In vivo Tumor Targeting Unit, San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Chiara Bresesti
- Vector Engineering and In vivo Tumor Targeting Unit, San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Giovanna Giacca
- Vector Engineering and In vivo Tumor Targeting Unit, San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Thomas Kerzel
- Vector Engineering and In vivo Tumor Targeting Unit, San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Carl Mirko Mercado
- Vector Engineering and In vivo Tumor Targeting Unit, San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Stefano Beretta
- BioInformatics Core, San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Marco Monti
- BioInformatics Core, San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Ivan Merelli
- BioInformatics Core, San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Silvia Iaia
- Mechanisms of Peripheral Tolerance Unit and Immune Core, San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Marco Genua
- Genomics of the Innate Immune System Unit, San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Andrea Annoni
- Mechanisms of Peripheral Tolerance Unit and Immune Core, San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Tamara Canu
- Preclinical Imaging Facility, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Patrizia Cristofori
- GLP Test Facility, San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Sara Degl'Innocenti
- GLP Test Facility, San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Francesca Sanvito
- GLP Test Facility, San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Pathology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | | | - Renato Ostuni
- Vita-Salute San Raffaele University, Milan, Italy
- Genomics of the Innate Immune System Unit, San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Silvia Gregori
- Mechanisms of Peripheral Tolerance Unit and Immune Core, San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Luigi Naldini
- Vita-Salute San Raffaele University, Milan, Italy
- Targeted Cancer Gene Therapy Unit, San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Mario Leonardo Squadrito
- Vector Engineering and In vivo Tumor Targeting Unit, San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy.
- Vita-Salute San Raffaele University, Milan, Italy.
| |
Collapse
|
36
|
Bhojnagarwala PS, Jose J, Zhao S, Weiner DB. DNA-based immunotherapy for cancer: In vivo approaches for recalcitrant targets. Mol Ther 2025:S1525-0016(25)00282-5. [PMID: 40211538 DOI: 10.1016/j.ymthe.2025.04.008] [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: 01/07/2025] [Revised: 04/01/2025] [Accepted: 04/07/2025] [Indexed: 05/10/2025] Open
Abstract
Immunotherapy has revolutionized cancer treatment and complements traditional therapies, including surgery, chemotherapy, radiation therapy, and targeted therapies. Immunotherapy redirects the patient's immune system against tumors via several immune-mediated approaches. Over the past few years, therapeutic immunization, which enable the patient's T cells to better recognize and kill tumors, have been increasingly tested in the clinic, with several approaches demonstrating treatment improvements. There has been a renewed interest in cancer vaccines due to advances in tumor antigen identification, immune response optimization, novel adjuvants, next-generation vaccine delivery platforms, and antigen designs. The COVID-19 pandemic accelerated progress in nucleic acid-based vaccine manufacturing, which spurred broader interest in mRNA or plasmid platforms. Enhanced DNA vaccine designs, including optimized leader sequences and RNA and codon optimizations, improved formulations, and delivery via adaptive electroporation using stereotactic intramuscular/intradermal methods have improved T cell responses to plasmid-delivered tumor antigens. Additionally, advancements for direct in vivo delivery of DNA-encoded monospecific/bispecific antibodies offer novel tumor-targeting strategies. This review summarizes the recent clinical data for therapeutic cancer vaccines utilizing the DNA platform, including vaccines targeting common tumor-associated and viral antigens and neoantigen vaccines using nucleic acid technologies. We also summarize preclinical data using DNA-launched monoclonal/bispecific antibodies, underscoring their potential as a novel cancer therapy tool.
Collapse
Affiliation(s)
- Pratik S Bhojnagarwala
- Vaccine and Immunotherapy Center, The Wistar Institute, 3601 Spruce Street, Philadelphia, PA, USA
| | - Joshua Jose
- Vaccine and Immunotherapy Center, The Wistar Institute, 3601 Spruce Street, Philadelphia, PA, USA
| | - Shushu Zhao
- Vaccine and Immunotherapy Center, The Wistar Institute, 3601 Spruce Street, Philadelphia, PA, USA
| | - David B Weiner
- Vaccine and Immunotherapy Center, The Wistar Institute, 3601 Spruce Street, Philadelphia, PA, USA.
| |
Collapse
|
37
|
Jamal A, Aldreiwish AD, Banawas SS, Alqurashi YE, Kamal MA, Ahmad F. The paths toward immunotherapy of esophageal cancer: An overview of clinical trials. Int Immunopharmacol 2025; 151:114261. [PMID: 40015204 DOI: 10.1016/j.intimp.2025.114261] [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: 12/30/2024] [Revised: 01/26/2025] [Accepted: 02/06/2025] [Indexed: 03/01/2025]
Abstract
As the seventh-leading contributor to global cancer-related deaths, esophageal cancer (EC) is one of the most challenging types of cancer. Despite advancements in conventional therapies, including surgery, chemotherapy, and radiotherapy, the five-year survival rate remains low, underscoring the need for the development of more efficacious treatment approaches. Immunotherapy has emerged as a promising treatment approach, offering new hope for EC patients. This review provides an in-depth examination of the latest immunotherapeutic strategies for EC, focusing on immune checkpoint inhibitors, adoptive cell therapy, cancer vaccines, and oncolytic virotherapy. We critically analyze the current clinical data to highlight the progress and pitfalls of each immunotherapeutic approach for EC. Additionally, we explore the potential for combination therapies, which could overcome the resistance often seen with monotherapies. Finally, we discuss the limitations of current treatments and outline key areas for future research to improve patient outcomes and survival.
Collapse
Affiliation(s)
- Azfar Jamal
- Department of Biology, College of Science Al-Zulfi, Majmaah University, Al-Majmaah 11952, Saudi Arabia; Health and Basic Science Research Centre, Majmaah University, Al-Majmaah 11952, Saudi Arabia.
| | - Allolo D Aldreiwish
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Majmaah University, Majmaah 11952, Saudi Arabia
| | - Saeed S Banawas
- Health and Basic Science Research Centre, Majmaah University, Al-Majmaah 11952, Saudi Arabia; Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Majmaah University, Majmaah 11952, Saudi Arabia
| | - Yaser E Alqurashi
- Department of Biology, College of Science Al-Zulfi, Majmaah University, Al-Majmaah 11952, Saudi Arabia
| | - Mohammad Azhar Kamal
- Department of Pharmaceutics, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Alkharj 11942, Saudi Arabia
| | - Fuzail Ahmad
- Respiratory Care Department, College of Applied Sciences, Almaarefa University, Diriya, Riyadh 13713, Saudi Arabia
| |
Collapse
|
38
|
Ma S, Lu Y, Sui S, Yang JS, Fu BB, Tan PX, Chai Y, Lv J, Kong L, Wu X, Gao YB, Yan T. Unraveling the triad of immunotherapy, tumor microenvironment, and skeletal muscle biomechanics in oncology. Front Immunol 2025; 16:1572821. [PMID: 40242775 PMCID: PMC12000078 DOI: 10.3389/fimmu.2025.1572821] [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: 02/07/2025] [Accepted: 03/03/2025] [Indexed: 04/18/2025] Open
Abstract
The intricate interaction between skeletal muscle biomechanics, the tumor microenvironment, and immunotherapy constitutes a pivotal research focus oncology. This work provides a comprehensive review of methodologies for evaluating skeletal muscle biomechanics, including handheld dynamometry, advanced imaging techniques, electrical impedance myography, elastography, and single-fiber experiments to assess muscle quality and performance. Furthermore, it elucidates the mechanisms, applications, and limitations of various immunotherapy modalities, including immune checkpoint inhibitors, adoptive cell therapy, cancer vaccines, and combined chemoimmunotherapy, while examining their effects on skeletal muscle function and systemic immune responses. Key findings indicate that although immunotherapy is effective in augmenting antitumor immunity, it frequently induces muscle-related adverse effects such as weakness, fatigue, or damage, primarily mediated by cytokine release and immune activation. This work underscores the significance of immune niches within the tumor microenvironment in influencing treatment outcomes and proposes strategies to optimize therapy through personalized regimens and combinatorial approaches. This review highlights the need for further research on the formation of immune niches and interactions muscle-tumor. Our work is crucial for advancing the efficacy of immunotherapy, reducing adverse effects, and ultimately improving survival rates and quality of life of patients with cancer.
Collapse
Affiliation(s)
- Shuang Ma
- School of Information Science and Engineering, Shenyang Ligong University, Shenyang, China
| | - Ying Lu
- School of Information Science and Engineering, Shenyang Ligong University, Shenyang, China
| | - Shang Sui
- St. John’s Kilmarnock School, Breslau, ON, Canada
| | - Jia-shuo Yang
- Department of Anesthesiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Bing-bing Fu
- Department of Anesthesiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Pei-xin Tan
- Department of Anesthesiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yicheng Chai
- School of Information Science and Engineering, Shenyang Ligong University, Shenyang, China
| | - Jiaqi Lv
- School of Information Science and Engineering, Shenyang Ligong University, Shenyang, China
| | - Lingyu Kong
- School of Information Science and Engineering, Shenyang Ligong University, Shenyang, China
| | - Xiaolin Wu
- School of Mathematics and Statistics, Liaoning University, Shenyang, China
| | - Yi-bo Gao
- Department of Oral and Maxillofacial Surgery, Taikang Bybo Dental, Beijing, China
| | - Tao Yan
- Department of Anesthesiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
39
|
Cho YB, Ko IY, Kim HJ, Kim JW, Heo K, Na HJ, Woo JR, Lee S. Functional epitope mapping of cell surface glucose-regulated protein 94: A combinatorial approach for therapeutic targeting. Int J Biol Macromol 2025; 301:140374. [PMID: 39880265 DOI: 10.1016/j.ijbiomac.2025.140374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 01/14/2025] [Accepted: 01/25/2025] [Indexed: 01/31/2025]
Abstract
Glucose-regulated protein 94 (GRP94) overexpression plays a critical role in tumor cell survival across various cancers. Previously, we developed K101.1, a fully human antibody targeting cell surface GRP94, which effectively inhibits tumor angiogenesis in colorectal cancer (CRC). This study aims to identify K101.1's interaction site and further elucidate GRP94's role in tumor angiogenesis. In an HT29 CRC xenograft mouse model, K101.1 reduced tumor growth and angiogenesis by 30 % and 69 %, respectively, without inducing severe toxicity. Using hydrogen‑deuterium exchange mass spectrometry, we identified the binding site of K101.1 on GRP94, analyzing 225 peptides with 94.5 % sequence coverage and a redundancy score of 4.20. This revealed a linear epitope (Glu26-Ala34) as the specific binding site. The binding was further validated via enzyme-linked immunosorbent assay, and human umbilical vein endothelial cell tube formation assays using a synthetic epitope peptide corresponding to the identified epitope. Our findings suggest that this epitope is functionally involved in GRP94-mediated angiogenesis and is integral to its proangiogenic activity. By integrating epitope profiling with complementary experimental approaches, this study advances the understanding of GRP94's role in CRC angiogenesis and provides new insights for developing targeted cancer therapies and vaccines, offering promising avenues for CRC prevention and treatment.
Collapse
Affiliation(s)
- Yea Bin Cho
- Department of Chemistry, Kookmin University, Seoul 02707, Republic of Korea
| | - In Young Ko
- New Drug Development Center, KBIOHealth, Cheongju 28160, Republic of Korea
| | - Hyun Jung Kim
- Department of Biopharmaceutical Chemistry, Kookmin University, Seoul 02707, Republic of Korea
| | - Ji Woong Kim
- Department of Biopharmaceutical Chemistry, Kookmin University, Seoul 02707, Republic of Korea
| | - Kyun Heo
- Department of Chemistry, Kookmin University, Seoul 02707, Republic of Korea; Department of Biopharmaceutical Chemistry, Kookmin University, Seoul 02707, Republic of Korea; Antibody Research Institute, Kookmin University, Seoul 02707, Republic of Korea
| | - Hee Jun Na
- HauulBio, Chuncheon, Gangwon 24398, Republic of Korea
| | - Ju Rang Woo
- New Drug Development Center, KBIOHealth, Cheongju 28160, Republic of Korea.
| | - Sukmook Lee
- Department of Chemistry, Kookmin University, Seoul 02707, Republic of Korea; Department of Biopharmaceutical Chemistry, Kookmin University, Seoul 02707, Republic of Korea; Antibody Research Institute, Kookmin University, Seoul 02707, Republic of Korea.
| |
Collapse
|
40
|
Sprooten J, Garg AD. Next-generation DC vaccines with an immunogenic trajectory against cancer: therapeutic opportunities vs. resistance mechanisms. Genes Immun 2025; 26:166-168. [PMID: 39191999 DOI: 10.1038/s41435-024-00294-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/29/2024]
Affiliation(s)
- Jenny Sprooten
- Department of Cellular & Molecular Medicine, Laboratory of Cell Stress & Immunity, KU Leuven, Leuven, Belgium
| | - Abhishek D Garg
- Department of Cellular & Molecular Medicine, Laboratory of Cell Stress & Immunity, KU Leuven, Leuven, Belgium.
| |
Collapse
|
41
|
Yang Y, Wu Y. Potential of bacterial outer membrane vesicles in tumor vaccine: characteristics, advancements, and future directions. Essays Biochem 2025; 69:EBC20253004. [PMID: 40159726 DOI: 10.1042/ebc20253004] [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: 01/03/2025] [Accepted: 03/02/2025] [Indexed: 04/02/2025]
Abstract
Bacterial outer membrane vesicles (OMVs), naturally released by Gram-negative bacteria, are a type of lipid bilayer nanoparticles containing many components found within the parent bacterium. Despite OMVs were first considered mere by-products of bacterial growth, recent studies have shown them as a highly adaptable platform for tumor vaccine. Here, we first demonstrate the biogenesis of OMVs, then review the strong immunogenicity of OMVs as an immune adjuvant in tumor vaccine and its excellent vaccine delivery capability, and finally discuss OMVs' engineering potentials through summarizing recent scientific advancements in genetic engineering, chemical modification, and nanotechnology. We also point out the clinical trials and future challenges of OMV-based vaccine. Overall, this review offers valuable insights into cancer immunotherapy, providing a roadmap for leveraging OMVs as a versatile platform for next-generation cancer vaccines.
Collapse
Affiliation(s)
- Yizhe Yang
- College of Nano Science & Technology (CNST), Soochow University, Suzhou, Jiangsu 215123, China
| | - Yumin Wu
- Institute of Functional Nano & Soft Materials (FUNSOM), Soochow University, Suzhou, Jiangsu 215123, China
| |
Collapse
|
42
|
Zheng Y, Wang B, Cai Z, Lai Z, Yu H, Wu M, Liu X, Zhang D. Tailoring nanovectors for optimal neoantigen vaccine efficacy. J Mater Chem B 2025; 13:4045-4058. [PMID: 40042164 DOI: 10.1039/d4tb02547d] [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: 03/27/2025]
Abstract
The primary objective of neoantigen vaccines is to elicit a robust anti-tumor immune response by generating neoantigen-specific T cells that can eradicate tumor cells. Despite substantial advancements in personalized neoantigen prediction using next-generation sequencing, machine learning, and mass spectrometry, challenges remain in efficiently expanding neoantigen-specific T cell populations in vivo. This challenge impedes the widespread clinical application of neoantigen vaccines. Nanovector-based neoantigen delivery systems have emerged as a promising solutions to this challenge. These nanovectors offer several advantages, such as enhanced stability, targeted intracellular delivery, sustained release, and improved antigen-presenting cell (APC) activation. Notably, they effectively deliver various neoantigen vaccine formulations (DC cell-based, synthetic long peptide (SLP)-based or DNA/mRNA-based) to APCs or T cells, thereby activating both CD4+ T and CD8+ T cells. This ultimately induces a specific anti-tumor immune response. This review focuses on recent innovations in neoantigen vaccine delivery vectors. We aim to identify optimal design parameters for vectors tailored to different neoantigen vaccine types, with an emphasis on enhancing the tumor microenvironment and stimulating the production of neoantigen-specific cytotoxic T cells. By maximizing the potential of these delivery systems, we aim to accelerate the clinical translation of neoantigen nanovaccines and advance cancer immunotherapy.
Collapse
Affiliation(s)
- Youshi Zheng
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, P. R. China.
- Fujian Agriculture and Forestry University, Fuzhou 350002, P. R. China
- Mengchao Med-X Center, Fuzhou University, Fuzhou 350116, P. R. China
| | - Bing Wang
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, P. R. China.
- Fujian Agriculture and Forestry University, Fuzhou 350002, P. R. China
| | - Zhixiong Cai
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, P. R. China.
- Fujian Agriculture and Forestry University, Fuzhou 350002, P. R. China
- Mengchao Med-X Center, Fuzhou University, Fuzhou 350116, P. R. China
| | - Zisen Lai
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, P. R. China.
- Fujian Agriculture and Forestry University, Fuzhou 350002, P. R. China
| | - Haijun Yu
- Mengchao Med-X Center, Fuzhou University, Fuzhou 350116, P. R. China
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Ming Wu
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, P. R. China.
- Fujian Agriculture and Forestry University, Fuzhou 350002, P. R. China
- Mengchao Med-X Center, Fuzhou University, Fuzhou 350116, P. R. China
| | - Xiaolong Liu
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, P. R. China.
- Fujian Agriculture and Forestry University, Fuzhou 350002, P. R. China
- CAS Key Laboratory of Design and Assembly of Functional Nanostructures, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou 350002, P. R. China
- Mengchao Med-X Center, Fuzhou University, Fuzhou 350116, P. R. China
| | - Da Zhang
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, P. R. China.
- Fujian Agriculture and Forestry University, Fuzhou 350002, P. R. China
- Mengchao Med-X Center, Fuzhou University, Fuzhou 350116, P. R. China
| |
Collapse
|
43
|
Chan LL, Chan SL. Future perspectives on immunotherapy for hepatocellular carcinoma. Ther Adv Med Oncol 2025; 17:17588359251323199. [PMID: 40144682 PMCID: PMC11938898 DOI: 10.1177/17588359251323199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2024] [Accepted: 02/05/2025] [Indexed: 03/28/2025] Open
Abstract
In recent years, several global phase III trials have shown that combinations of immune checkpoint inhibitors (ICIs) offer superior efficacy and survival compared to multi-kinase inhibitors, establishing them as the gold standard for treating patients with advanced hepatocellular carcinoma (HCC). This success has led to investigations into expanding the use of immunotherapy into various other settings and populations, including neoadjuvant and adjuvant therapies, patients with decompensated liver function and those awaiting liver transplantation. Despite its proven efficacy, a significant number of patients still develop resistance to immunotherapy, highlighting the need for innovative strategies to address this challenge. Approaches aimed at enhancing tumour immunogenicity, such as combining immunotherapy with transarterial chemoembolization or radiation therapies, show significant promise. Additionally, novel immunotherapeutics - such as triplet therapy, bispecific antibodies, adoptive T-cell therapy and cancer vaccines - are in early development for HCC. These agents have demonstrated potential for synergistic effects with existing ICIs, with initial studies yielding positive outcomes. In this review, we offer our future perspective on immunotherapy, emphasizing emerging indications, novel combination strategies and the development of new immunotherapeutic agents. Overall, the future of immunotherapy in HCC is brimming with extraordinary potential, set to transform the treatment landscape and redefine the possibilities for managing this challenging disease.
Collapse
Affiliation(s)
- Landon L. Chan
- State Key Laboratory of Translational Oncology, Department of Clinical Oncology, Sir YK Pao Centre for Cancer, SIRT, Hong Kong Cancer Institute, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Stephen L. Chan
- State Key Laboratory of Translational Oncology, Department of Clinical Oncology, Sir YK Pao Centre for Cancer, SIRT, Hong Kong Cancer Institute, Prince of Wales Hospital, The Chinese University of Hong Kong, 30-32 Ngan Shing Street, Shatin, Hong Kong, China
| |
Collapse
|
44
|
Thiruppathi J, Vijayan V, Hwang HS, Bang YJ, Loeurng V, Hong SH, Sundaram A, Park IK, Lee SE, Rhee JH. Thermoresistant flagellin-adjuvanted cancer vaccine combined with photothermal therapy synergizes with anti-PD-1 treatment. J Immunother Cancer 2025; 13:e010272. [PMID: 40118497 PMCID: PMC11931959 DOI: 10.1136/jitc-2024-010272] [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: 08/02/2024] [Accepted: 03/04/2025] [Indexed: 03/23/2025] Open
Abstract
BACKGROUND Cancer immunotherapy, leveraging the immune system to target and eradicate cancer cells, has transformed cancer treatment paradigms. Immune checkpoint inhibitors (ICIs) are used in a wide array of cancers, but only a limited fraction of patients are responding. Cancer vaccines could elicit antigen-specific immune responses and establish long-term immune memory, preventing recurrence and metastasis. Despite their promising profiles, ICIs and cancer vaccines by themselves are often insufficient to overcome the immunosuppressive tumor microenvironment (TME) and recurrence/metastasis. Addressing these challenges is crucial for improving cancer immunotherapy outcomes. METHODS The targeted liposomal formulation (TLIF), displaying Cyclic RGD (cRGD) peptide on the surface and encapsulating ICG and thermoresistant flagellin (FlaB) inside, was used for photothermal therapy (PTT), which was designed to induce robust immunogenic cell death (ICD) and release tumor antigens (TAs). We employed a mouse breast cancer model amenable to PTT. Utilizing a bilateral DD-Her2/neu tumor implantation model, we evaluated local and abscopal effects of combinatorial approaches employing PTT, FlaB-adjuvanted peptide vaccine (FlaB-Vax), and anti-PD-1 treatment. FlaB-Vax was designed to trigger tumor-associated antigen (TAA)-specific immune responses, which will trigger specific anti-tumor immunity. TLIF-PTT aimed to reduce tumor burden and induce ICD-mediated TA liberation for epitope spreading. Sustained anti-tumor immune memory was assessed by orthotopic rechallenging cured mice with the DD-Her2/neu tumor cells. RESULTS The combination of TLIF-PTT and FlaB-Vax provided significantly enhanced primary tumor suppression, with strong abscopal effects and long-lasting immune memory. The addition of anti-PD-1 therapy further improved long-term relapse-free survival, highlighting the potential of this combinatorial approach to induce durable antitumor immunity and sustainably prevent cancer recurrence and metastasis. CONCLUSION This study demonstrates that the combination of TLIF-PTT and FlaB-Vax synergistically induced synergistic anti-tumor immune responses, which were efficaciously potentiated by anti-PD-1 treatment for recurrence-free long-term survival.
Collapse
Affiliation(s)
| | - Veena Vijayan
- Chonnam National University, Hwasun, Korea (the Republic of)
| | - Hye Suk Hwang
- Chonnam National University, Hwasun, Korea (the Republic of)
- Department of Biomedical Science,College of Life Science and Industry, Sunchon National University, Sunchon 57922, South Korea
| | - Yong Jun Bang
- Chonnam National University, Hwasun, Korea (the Republic of)
| | - Vandara Loeurng
- Chonnam National University, Hwasun, Korea (the Republic of)
| | - Seol Hee Hong
- Chonnam National University, Gwangju, Korea (the Republic of)
| | | | - In-Kyu Park
- Chonnam National University Medical School, Gwangju, Korea (the Republic of)
| | - Shee Eun Lee
- Chonnam National University, Gwangju, Korea (the Republic of)
| | - Joon Haeng Rhee
- Chonnam National University, Hwasun, Korea (the Republic of)
| |
Collapse
|
45
|
Kwon WA, Joung JY. Immunotherapy in Prostate Cancer: From a "Cold" Tumor to a "Hot" Prospect. Cancers (Basel) 2025; 17:1064. [PMID: 40227610 PMCID: PMC11987915 DOI: 10.3390/cancers17071064] [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: 02/17/2025] [Revised: 03/18/2025] [Accepted: 03/20/2025] [Indexed: 04/15/2025] Open
Abstract
Immunotherapy has shown limited efficacy in prostate cancer, largely due to low tumor immunogenicity, sparse tumor-infiltrating lymphocytes, and a suppressive microenvironment. Recent therapeutic strategies aim to boost immune responses and counteract immunosuppressive factors through interventions such as immune checkpoint inhibitors, immunogenic cell death-inducing therapies, and the targeted blockade of pathways like that of transforming growth factor-β. Vaccine-based approaches, potent immune adjuvants, and engineered chimeric antigen receptor (CAR) T cells are also being investigated to overcome local immune inhibitory signals. Advancements in imaging, multi-omic profiling, and liquid biopsies offer promising avenues for real-time monitoring, better patient selection, and precision treatment. This review provides an overview of the key immunosuppressive features of prostate cancer, current immunotherapeutic modalities, and emerging strategies to transform "cold" tumors into more responsive "hot" targets. By integrating these approaches, we may achieve more durable clinical benefits for patients with advanced or metastatic prostate cancer.
Collapse
Affiliation(s)
- Whi-An Kwon
- Department of Urology, Hanyang University College of Medicine, Myongji Hospital, Goyang 10475, Republic of Korea
| | - Jae Young Joung
- Department of Urology, Urological Cancer Center, National Cancer Center, Goyang 10408, Republic of Korea
| |
Collapse
|
46
|
Stojchevski R, Sutanto EA, Sutanto R, Hadzi-Petrushev N, Mladenov M, Singh SR, Sinha JK, Ghosh S, Yarlagadda B, Singh KK, Verma P, Sengupta S, Bhaskar R, Avtanski D. Translational Advances in Oncogene and Tumor-Suppressor Gene Research. Cancers (Basel) 2025; 17:1008. [PMID: 40149342 PMCID: PMC11940485 DOI: 10.3390/cancers17061008] [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: 02/10/2025] [Revised: 03/10/2025] [Accepted: 03/15/2025] [Indexed: 03/29/2025] Open
Abstract
Cancer, characterized by the uncontrolled proliferation of cells, is one of the leading causes of death globally, with approximately one in five people developing the disease in their lifetime. While many driver genes were identified decades ago, and most cancers can be classified based on morphology and progression, there is still a significant gap in knowledge about genetic aberrations and nuclear DNA damage. The study of two critical groups of genes-tumor suppressors, which inhibit proliferation and promote apoptosis, and oncogenes, which regulate proliferation and survival-can help to understand the genomic causes behind tumorigenesis, leading to more personalized approaches to diagnosis and treatment. Aberration of tumor suppressors, which undergo two-hit and loss-of-function mutations, and oncogenes, activated forms of proto-oncogenes that experience one-hit and gain-of-function mutations, are responsible for the dysregulation of key signaling pathways that regulate cell division, such as p53, Rb, Ras/Raf/ERK/MAPK, PI3K/AKT, and Wnt/β-catenin. Modern breakthroughs in genomics research, like next-generation sequencing, have provided efficient strategies for mapping unique genomic changes that contribute to tumor heterogeneity. Novel therapeutic approaches have enabled personalized medicine, helping address genetic variability in tumor suppressors and oncogenes. This comprehensive review examines the molecular mechanisms behind tumor-suppressor genes and oncogenes, the key signaling pathways they regulate, epigenetic modifications, tumor heterogeneity, and the drug resistance mechanisms that drive carcinogenesis. Moreover, the review explores the clinical application of sequencing techniques, multiomics, diagnostic procedures, pharmacogenomics, and personalized treatment and prevention options, discussing future directions for emerging technologies.
Collapse
Affiliation(s)
- Radoslav Stojchevski
- Friedman Diabetes Institute, Lenox Hill Hospital, Northwell Health, New York, NY 10022, USA;
- Feinstein Institutes for Medical Research, Manhasset, NY 11030, USA
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY 11549, USA
| | - Edward Agus Sutanto
- CUNY School of Medicine, The City College of New York, 160 Convent Avenue, New York, NY 10031, USA;
| | - Rinni Sutanto
- New York Institute of Technology College of Osteopathic Medicine, Glen Head, NY 11545, USA;
| | - Nikola Hadzi-Petrushev
- Faculty of Natural Sciences and Mathematics, Institute of Biology, Ss. Cyril and Methodius University, 1000 Skopje, North Macedonia; (N.H.-P.)
| | - Mitko Mladenov
- Faculty of Natural Sciences and Mathematics, Institute of Biology, Ss. Cyril and Methodius University, 1000 Skopje, North Macedonia; (N.H.-P.)
| | - Sajal Raj Singh
- GloNeuro, Sector 107, Vishwakarma Road, Noida 201301, Uttar Pradesh, India (J.K.S.)
| | - Jitendra Kumar Sinha
- GloNeuro, Sector 107, Vishwakarma Road, Noida 201301, Uttar Pradesh, India (J.K.S.)
| | - Shampa Ghosh
- GloNeuro, Sector 107, Vishwakarma Road, Noida 201301, Uttar Pradesh, India (J.K.S.)
| | | | - Krishna Kumar Singh
- Symbiosis Centre for Information Technology (SCIT), Rajiv Gandhi InfoTech Park, Hinjawadi, Pune 411057, Maharashtra, India;
| | - Prashant Verma
- School of Management, BML Munjal University, NH8, Sidhrawali, Gurugram 122413, Haryana, India
| | - Sonali Sengupta
- Department of Gastroenterology, All India Institute of Medical Sciences (AIIMS), New Delhi 110029, India
| | - Rakesh Bhaskar
- School of Chemical Engineering, Yeungnam University, Gyeongsan 38541, Republic of Korea
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan 38541, Republic of Korea
| | - Dimiter Avtanski
- Friedman Diabetes Institute, Lenox Hill Hospital, Northwell Health, New York, NY 10022, USA;
- Feinstein Institutes for Medical Research, Manhasset, NY 11030, USA
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY 11549, USA
| |
Collapse
|
47
|
Yuan Z, Wang JH, Cui H, Wang SY, Wei B, Cui JX. Mapping the landscape of gastric cancer immunotherapy: Bibliometric insights into advances and hotspots. World J Gastrointest Oncol 2025; 17:100997. [PMID: 40092931 PMCID: PMC11866247 DOI: 10.4251/wjgo.v17.i3.100997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Revised: 12/11/2024] [Accepted: 12/31/2024] [Indexed: 02/14/2025] Open
Abstract
BACKGROUND Immunotherapy has surfaced as a promising therapeutic modality for gastric cancer (GC). A comprehensive review of advancements, current status, and research trends in GC immunotherapy is essential to inform future investigative efforts. AIM To delineate the trends, advancements, and focal points in immunotherapy for GC. METHODS We performed a bibliometric analysis of 2906 articles in English concerning GC immunotherapy published from 2000 to December 20, 2023, indexed in the Web of Science Core Collection. Data analysis and visualization were facilitated by CiteSpace (6.1.6R), VOSviewer v.1.6.17, and GraphPad Prism v8.0.2. RESULTS There has been an increase in the annual publication rate of GC immunotherapy research. China leads in publication volume, while the United States demonstrates the highest citation impact. Fudan University is notable for its citation frequency and publication output. Co-citation analysis and keyword frequency revealed and highlighted a focus on GC prognosis, the tumor microenvironment (TME), and integrative immunotherapy with targeted therapy. Emerging research areas include gastroesophageal junction cancer, adoptive immunotherapy, and the role of Treg cell in immunotherapy. CONCLUSION GC immunotherapy research is an expanding field attracting considerable scientific interest. With the clinical adoption of immunotherapy in GC, the primary goals are to enhance treatment efficacy and patient outcomes. Unlike hematological malignancies, GC's solid TME presents distinct immunological challenges that may attenuate the cytotoxic effects of immune cells on cancer cells. For instance, although CAR-T therapy is effective in hematological malignancies, it has underperformed in GC settings. Current research is centered on overcoming immunosuppression within the TME, with a focus on combinations of targeted therapy, adoptive immunotherapy, Treg cell dynamics, and precise prognosis prediction in immunotherapy. Additionally, immunotherapy's role in treating gastroesophageal junction cancer has become a novel research focus.
Collapse
Affiliation(s)
- Zhen Yuan
- School of Medicine, Nankai University, Tianjin 300071, China
- Department of General Surgery, The First Medical Center, Chinese PLA General Hospital, Beijing 100853, China
| | - Jing-Hang Wang
- School of Medicine, Nankai University, Tianjin 300071, China
- Department of General Surgery, The First Medical Center, Chinese PLA General Hospital, Beijing 100853, China
| | - Hao Cui
- School of Medicine, Nankai University, Tianjin 300071, China
- Department of General Surgery, The First Medical Center, Chinese PLA General Hospital, Beijing 100853, China
| | - Shu-Yuan Wang
- School of Medicine, Nankai University, Tianjin 300071, China
| | - Bo Wei
- Department of General Surgery, The First Medical Center, Chinese PLA General Hospital, Beijing 100853, China
| | - Jian-Xin Cui
- Department of General Surgery, The First Medical Center, Chinese PLA General Hospital, Beijing 100853, China
| |
Collapse
|
48
|
Haghmorad D, Eslami M, Orooji N, Halabitska I, Kamyshna I, Kamyshnyi O, Oksenych V. mRNA vaccine platforms: linking infectious disease prevention and cancer immunotherapy. Front Bioeng Biotechnol 2025; 13:1547025. [PMID: 40144393 PMCID: PMC11937095 DOI: 10.3389/fbioe.2025.1547025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Accepted: 02/24/2025] [Indexed: 03/28/2025] Open
Abstract
The advent of mRNA vaccines, accelerated by the global response to the COVID-19 pandemic, marks a transformative shift in vaccine technology. In this article, we discuss the development, current applications, and prospects of mRNA vaccines for both the prevention and treatment of infectious diseases and oncology. By leveraging the capacity to encode antigens within host cells directly, mRNA vaccines provide a versatile and scalable platform suitable for addressing a broad spectrum of pathogens and tumor-specific antigens. We highlight recent advancements in mRNA vaccine design, innovative delivery mechanisms, and ongoing clinical trials, with particular emphasis on their efficacy in combating infectious diseases, such as COVID-19, Zika, and influenza, as well as their emerging potential in cancer immunotherapy. We also address critical challenges, including vaccine stability, optimization of immune responses, and the broader issue of global accessibility. Finally, we review potential strategies for advancing next-generation mRNA vaccines, with the aim of overcoming current limitations in vaccine technology and enhancing both preventive and therapeutic approaches for infectious and oncological diseases.
Collapse
Affiliation(s)
- Dariush Haghmorad
- Department of Immunology, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Majid Eslami
- Department of Bacteriology and Virology, Semnan University of Medical Sciences, Semnan, Iran
| | - Niloufar Orooji
- Department of Immunology, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran
- Student Research Committee, Semnan University of Medical Sciences, Semnan, Iran
| | - Iryna Halabitska
- Department of Therapy and Family Medicine, I. Horbachevsky Ternopil National Medical University, Ternopil, Ukraine
| | - Iryna Kamyshna
- Department of Medical Rehabilitation, I. Horbachevsky Ternopil National Medical University, Ternopil, Ukraine
| | - Oleksandr Kamyshnyi
- Department of Microbiology, Virology, and Immunology, I. Horbachevsky Ternopil National Medical University, Ternopil, Ukraine
| | - Valentyn Oksenych
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| |
Collapse
|
49
|
Nguyen CM, Vu TT, Nguyen MN, Tran-Nguyen TS, Huynh CT, Ha QT, Nguyen HN, Tran LS. Neoantigen-based mRNA vaccine exhibits superior anti-tumor activity compared to synthetic long peptides in an in vivo lung carcinoma model. Cancer Immunol Immunother 2025; 74:145. [PMID: 40072566 PMCID: PMC11949242 DOI: 10.1007/s00262-025-03992-7] [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: 10/27/2024] [Accepted: 02/21/2025] [Indexed: 03/14/2025]
Abstract
Neoantigen vaccines hold great promise in cancer immunotherapy, but the comparative efficacy of different vaccine platforms, particularly in the context of tumor burden (TB), remains insufficiently studied. In this research, we evaluated the safety and therapeutic efficacy of synthetic long peptide and mRNA-based vaccines, both designed to target identical neoantigens across different Lewis Lung Carcinoma (LLC) tumor burdens. We employed the LLC syngeneic mouse model, a widely used preclinical model for aggressive and immunosuppressive tumors. Our findings demonstrated that the mRNA-based vaccine significantly outperformed the peptide-based vaccine in preventing tumor growth in mice with low TB. These results underscore the potential of mRNA vaccines as a more effective approach for treating aggressive tumors, contributing valuable insights for the future development of neoantigen-based cancer vaccines.
Collapse
Affiliation(s)
| | - Trung T Vu
- Medical Genetics Institute, Ho Chi Minh City, Vietnam
| | | | | | - Chi Thien Huynh
- Biotechnology Center of Ho Chi Minh City, Ho Chi Minh City, Vietnam
| | - Quang Thanh Ha
- Biotechnology Center of Ho Chi Minh City, Ho Chi Minh City, Vietnam
| | | | - Le Son Tran
- Medical Genetics Institute, Ho Chi Minh City, Vietnam.
| |
Collapse
|
50
|
Cui R, Luo Z, Zhang X, Yu X, Yuan G, Li X, Xie F, Jiang O. Targeting PI3K Signaling to Overcome Tumor Immunosuppression: Synergistic Strategies to Enhance Cancer Vaccine Efficacy. Vaccines (Basel) 2025; 13:292. [PMID: 40266213 PMCID: PMC11946485 DOI: 10.3390/vaccines13030292] [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: 01/31/2025] [Revised: 03/01/2025] [Accepted: 03/06/2025] [Indexed: 04/24/2025] Open
Abstract
Phosphoinositide 3-kinases (PI3Ks), members of the lipid kinase family, play a significant role in modulating immune cell functions, including activation, proliferation, and differentiation. Recent studies have identified the PI3K signaling pathway as a key regulator in tumor biology and the immune microenvironment. This pathway enhances the activity of regulatory T cells (Tregs) and myeloid-derived suppressor cells (MDSCs), contributing to an immunosuppressive tumor microenvironment that impairs the effectiveness of cancer vaccines and immunotherapies. The present study explores PI3K isoforms, particularly p110γ and p110δ, and their associated signaling pathways. The therapeutic potential of selective PI3K inhibitors and their capacity to act synergistically with immunization strategies are analyzed. Targeting the PI3K signaling pathway represents a promising approach to counteract tumor-induced immune suppression and improve the efficacy of immune checkpoint inhibitors and vaccines, ultimately leading to better clinical outcomes.
Collapse
Affiliation(s)
- Ran Cui
- Department of Oncology, The First People’s Hospital of Neijiang, Neijiang 641000, China; (R.C.); (X.L.); (F.X.)
- Department of Oncology, Southwest Medical University, Luzhou 646000, China; (Z.L.); (X.Z.)
| | - Zhongxiang Luo
- Department of Oncology, Southwest Medical University, Luzhou 646000, China; (Z.L.); (X.Z.)
| | - Xialin Zhang
- Department of Oncology, Southwest Medical University, Luzhou 646000, China; (Z.L.); (X.Z.)
| | - Xinlin Yu
- Department of Oncology, Affiliated Hospital of Chengdu University, Chengdu 610000, China;
| | - Gang Yuan
- Department of Interventional & Vascular, Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou 646000, China;
| | - Xingming Li
- Department of Oncology, The First People’s Hospital of Neijiang, Neijiang 641000, China; (R.C.); (X.L.); (F.X.)
| | - Fei Xie
- Department of Oncology, The First People’s Hospital of Neijiang, Neijiang 641000, China; (R.C.); (X.L.); (F.X.)
| | - Ou Jiang
- Department of Oncology, The First People’s Hospital of Neijiang, Neijiang 641000, China; (R.C.); (X.L.); (F.X.)
- Department of Oncology, Southwest Medical University, Luzhou 646000, China; (Z.L.); (X.Z.)
| |
Collapse
|