1
|
Rastogi I, Mannone JA, Gibadullin R, Moseman JE, Sidney J, Sette A, McNeel DG, Gellman SH. β-amino acid substitution in the SIINFEKL antigen alters immunological recognition. Cancer Biol Ther 2025; 26:2486141. [PMID: 40200635 PMCID: PMC11988276 DOI: 10.1080/15384047.2025.2486141] [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: 02/18/2025] [Revised: 03/20/2025] [Accepted: 03/24/2025] [Indexed: 04/10/2025] Open
Abstract
BACKGROUND Peptide vaccines offer a direct way to initiate an immunogenic response to a defined antigen epitope. However, peptide vaccines are unstable in vivo, subject to rapid enzymatic proteolysis. Replacement of an α-amino acid residue with a homologous β-amino acid residue (native side chain, but backbone extended by a single CH2 unit) impairs proteolysis at nearby amide bonds. Therefore, antigen analogues containing α-to-β replacements have been examined for functional mimicry of native all-α antigens. Another group previously took this approach in the ovalbumin (OVA) antigen model by evaluating single α-to-β analogues of the murine major histocompatibility complex (MHC) I-restricted peptide SIINFEKL. METHODS We re-examined this set of α/β SIINFEKL antigens. We tested the susceptibility to proteolysis in mouse serum and their ability to activate OVA-antigen-specific CD8 T cells in vitro. Additionally, we tested the α/β antigens in vivo for their ability to induce an antigen-specific immunogenic response in naïve mice and in OVA-expressing tumor-bearing mice. RESULTS The α/β antigens were comparable to the native antigen in their susceptibility to proteolysis in serum. Each α/β antigen was capable of activating antigen-specific CD8 T cells in vitro. However, antigen-specific CD8 T cells induced against α/β antigens in vivo were not cross-reactive to the native antigen. Moreover, immunization with α/β analogues did not elicit anti-tumor effects in tumor-bearing mice. CONCLUSIONS We conclude that even though α/β analogues of the SIINFEKL antigen can elicit a T cell-based response, this class of backbone-modified peptides is not promising from the perspective of antitumor vaccine development.
Collapse
Affiliation(s)
- Ichwaku Rastogi
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI, USA
| | - John A. Mannone
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI, USA
| | - Ruslan Gibadullin
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI, USA
| | - Jena E. Moseman
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI, USA
| | - John Sidney
- Division of Vaccine Discovery, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Alessandro Sette
- Division of Vaccine Discovery, La Jolla Institute for Immunology, La Jolla, CA, USA
- Department of Medicine, University of California, San Diego, CA, USA
| | - Douglas G. McNeel
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI, USA
| | - Samuel H. Gellman
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI, USA
| |
Collapse
|
2
|
Hu J, Arvejeh PM, Bone S, Hett E, Marincola FM, Roh KH. Nanocarriers for cutting-edge cancer immunotherapies. J Transl Med 2025; 23:447. [PMID: 40234928 PMCID: PMC12001629 DOI: 10.1186/s12967-025-06435-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2025] [Accepted: 03/26/2025] [Indexed: 04/17/2025] Open
Abstract
Cancer immunotherapy aims to harness the body's own immune system for effective and long-lasting elimination of malignant neoplastic tissues. Owing to the advance in understanding of cancer pathology and immunology, many novel strategies for enhancing immunological responses against various cancers have been successfully developed, and some have translated into excellent clinical outcomes. As one promising strategy for the next generation of immunotherapies, activating the multi-cellular network (MCN) within the tumor microenvironment (TME) to deploy multiple mechanisms of action (MOAs) has attracted significant attention. To achieve this effectively and safely, delivering multiple or pleiotropic therapeutic cargoes to the targeted sites of cancerous tissues, cells, and intracellular organelles is critical, for which numerous nanocarriers have been developed and leveraged. In this review, we first introduce therapeutic payloads categorized according to their predicted functions in cancer immunotherapy and their physicochemical structures and forms. Then, various nanocarriers, along with their unique characteristics, properties, advantages, and limitations, are introduced with notable recent applications in cancer immunotherapy. Following discussions on targeting strategies, a summary of each nanocarrier matching with suitable therapeutic cargoes is provided with comprehensive background information for designing cancer immunotherapy regimens.
Collapse
Affiliation(s)
- Joyce Hu
- Translational and Advanced Medicine (TAM) Biosciences, Nashville, TN, 37011, USA
| | - Pooria M Arvejeh
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Sydney Bone
- Department of Chemical and Materials Engineering, The University of Alabama in Huntsville, Huntsville, AL, 35899, USA
| | - Erik Hett
- Translational and Advanced Medicine (TAM) Biosciences, Nashville, TN, 37011, USA
| | | | - Kyung-Ho Roh
- Department of Chemical and Materials Engineering, The University of Alabama in Huntsville, Huntsville, AL, 35899, USA.
- Biotechnology Science and Engineering Program, The University of Alabama in Huntsville, Huntsville, AL, 35899, USA.
| |
Collapse
|
3
|
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] [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
MESH Headings
- Animals
- Antigens, Neoplasm/immunology
- Cancer Vaccines/immunology
- Cancer Vaccines/therapeutic use
- Mice
- Carcinoma, Lewis Lung/immunology
- Carcinoma, Lewis Lung/therapy
- mRNA Vaccines/immunology
- Lung Neoplasms/immunology
- Lung Neoplasms/therapy
- Mice, Inbred C57BL
- Disease Models, Animal
- Peptides/immunology
- Immunotherapy/methods
- Female
- Humans
- Vaccines, Synthetic/immunology
- Vaccines, Subunit/immunology
- Vaccines, Subunit/therapeutic use
- RNA, Messenger/genetics
- RNA, Messenger/immunology
- Cell Line, Tumor
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
|
4
|
Che J, Liu Y, Liu Y, Song J, Cui H, Feng D, Tian A, Zhang Z, Xu Y. The application of emerging immunotherapy in the treatment of prostate cancer: progress, dilemma and promise. Front Immunol 2025; 16:1544882. [PMID: 40145100 PMCID: PMC11937122 DOI: 10.3389/fimmu.2025.1544882] [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: 12/13/2024] [Accepted: 02/25/2025] [Indexed: 03/28/2025] Open
Abstract
In recent years, there has been a growing trend towards the utilization of immunotherapy techniques for the treatment of cancer. Some malignancies have acquired significant progress with the use of cancer vaccines, immune checkpoint inhibitors, and adoptive cells therapy. Scholars are exploring the aforementioned methods as potential treatments for advanced prostate cancer (PCa) due to the absence of effective adjuvant therapy to improve the prognosis of metastatic castration-resistant prostate cancer (mCRPC). Immunotherapy strategies have yet to achieve significant advancements in the treatment of PCa, largely attributed to the inhibitory tumor microenvironment and low mutation load characteristic of this malignancy. Hence, researchers endeavor to address these challenges by optimizing the design and efficacy of immunotherapy approaches, as well as integrating them with other therapeutic modalities. To date, studies have also shown potential clinical benefits. This comprehensive review analyzed the utilization of immunotherapy techniques in the treatment of PCa, assessing their advantages and obstacles, with the aim of providing healthcare professionals and scholars with a comprehensive understanding of the progress in this field.
Collapse
Affiliation(s)
- Jizhong Che
- Department of Urology, Yantai Affiliated Hospital of Binzhou Medical University, The Second Clinical Medical College of Binzhou Medical University, Yantai, Shandong, China
| | - Yuanyuan Liu
- Department of Urology, Yantai Affiliated Hospital of Binzhou Medical University, The Second Clinical Medical College of Binzhou Medical University, Yantai, Shandong, China
| | - Yangyang Liu
- Department of Urology, Yantai Affiliated Hospital of Binzhou Medical University, The Second Clinical Medical College of Binzhou Medical University, Yantai, Shandong, China
| | - Jingheng Song
- Department of Urology, Yantai Affiliated Hospital of Binzhou Medical University, The Second Clinical Medical College of Binzhou Medical University, Yantai, Shandong, China
| | - Hongguo Cui
- Department of Urology, Yantai Affiliated Hospital of Binzhou Medical University, The Second Clinical Medical College of Binzhou Medical University, Yantai, Shandong, China
| | - Dongdong Feng
- Department of Urology, Haiyang City People’s Hospital, Yantai, Shandong, China
| | - Aimin Tian
- Department of Urology, Yantai Affiliated Hospital of Binzhou Medical University, The Second Clinical Medical College of Binzhou Medical University, Yantai, Shandong, China
| | - Zhengchao Zhang
- Department of Urology, Yantai Affiliated Hospital of Binzhou Medical University, The Second Clinical Medical College of Binzhou Medical University, Yantai, Shandong, China
| | - Yankai Xu
- Department of Urology, Yantai Affiliated Hospital of Binzhou Medical University, The Second Clinical Medical College of Binzhou Medical University, Yantai, Shandong, China
| |
Collapse
|
5
|
Ying F, Zhang Y, Li X, Meng Z, Li J, Lo CY, Peng W, Tian X, Yao X. Active Immunization Using TRPM2 Peptide Vaccine Attenuates Atherosclerotic Progression in a Mouse Model of Atherosclerosis. Vaccines (Basel) 2025; 13:241. [PMID: 40266108 DOI: 10.3390/vaccines13030241] [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: 01/06/2025] [Revised: 02/22/2025] [Accepted: 02/24/2025] [Indexed: 04/24/2025] Open
Abstract
Background/Objective: Atherosclerosis is one of the leading causes of cardiovascular diseases and mortality around the world. One exciting strategy for atherosclerosis treatment is immunotherapy, especially active immunization. Active immunization relies on the delivery of antigens in a vaccine platform to introduce humoral and cellular immunity, alleviating atherosclerotic progression. Transient receptor potential channel isoform M2 (TRPM2) is an ROS-activated Ca2+-permeable ion channel that can promote atherosclerosis via stimulating vascular inflammation. In the present study, we developed a strategy of active immunization with the TRPM2 E3 domain peptide in a vaccine platform, aiming to induce the endogenous production of anti-TRPM2 blocking antibody in mice in vivo, consequently inhibiting TRPM2 channel activity to alleviate atherosclerotic progression. Methods: ApoE knockout mice were fed with a high cholesterol diet to develop atherosclerosis. The mice were injected with or without the E3 peptide vaccines, followed by analysis of atherosclerotic lesion by en face Oil Red O staining of the whole aorta and histologic analysis of thin tissue sections from aortic roots. Results: The results show that immunization with a pig TRPM2 E3 region-based peptide (P1) could effectively alleviate high cholesterol diet-induced atherosclerosis in ApoE knockout mice. We worked out the best vaccine formulation for the most effective atheroprotection, namely P1 at the dose of 67.5 µg per mouse (2.5 mg/kg body weight) with aluminum salts as adjuvant. Conclusions: The present study provides a novel target TRPM2 for peptide vaccine-based anti-atherosclerotic strategy and lays the foundation for future preclinical/clinical trials using TRPM2 E3 P1 vaccine for a potential therapeutic option against atherosclerosis.
Collapse
Affiliation(s)
- Fan Ying
- School of Biomedical Sciences, Heart and Vascular Institute, Li Ka Shing Institute of Health Science, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Yunting Zhang
- School of Biomedical Sciences, Heart and Vascular Institute, Li Ka Shing Institute of Health Science, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
- Department of Laboratory Medicine, The Second Affiliated Hospital of Chongqing Medical University, No. 74 Linjing Road, Yuzhong District, Chongqing 400010, China
| | - Xiao Li
- School of Biomedical Sciences, Heart and Vascular Institute, Li Ka Shing Institute of Health Science, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
- Mengniu Institute of Nutrition Science, Global R&D Innovation Center, Inner Mongolia Mengniu Dairy (Group) Co., Ltd., Hohhot 011500, China
| | - Zhaoyue Meng
- School of Biomedical Sciences, Heart and Vascular Institute, Li Ka Shing Institute of Health Science, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Jingxuan Li
- School of Biomedical Sciences, Heart and Vascular Institute, Li Ka Shing Institute of Health Science, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Chun-Yin Lo
- School of Biomedical Sciences, Heart and Vascular Institute, Li Ka Shing Institute of Health Science, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Wentao Peng
- School of Biomedical Sciences, Heart and Vascular Institute, Li Ka Shing Institute of Health Science, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Xiaoyu Tian
- School of Biomedical Sciences, Heart and Vascular Institute, Li Ka Shing Institute of Health Science, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Xiaoqiang Yao
- School of Biomedical Sciences, Heart and Vascular Institute, Li Ka Shing Institute of Health Science, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
- School of Medicine, The Chinese University of Hong Kong, Shenzhen 518172, China
| |
Collapse
|
6
|
Talpin A, Maia A, Carpier JM, Kulakowski G, Aubergeon L, Kervevan J, Gaal C, Strozzi F, Billerey C, Amable L, Mersceman T, Garnier A, Oliveira C, Calderon C, Bachrouche D, Ventujol C, Bernard L, Manteau A, Martinez J, Bonnet M, Noguerol J, Laviolette K, Boullerot L, Malfroy M, Chevalier G, Adotevi O, Joffre O, Idbaih A, Vieito M, Ghiringhelli F, Stradella A, Tabatabai G, Burger MC, Mildenberger I, Herrlinger U, Reardon DA, Wick W, Gouttefangeas C, Bonny C, Chene L, Gamelas Magalhaes J. Mimicry-based strategy between human and commensal antigens for the development of a new family of immune therapies for cancer. J Immunother Cancer 2025; 13:e010192. [PMID: 39979071 PMCID: PMC11842988 DOI: 10.1136/jitc-2024-010192] [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: 07/25/2024] [Accepted: 01/28/2025] [Indexed: 02/22/2025] Open
Abstract
BACKGROUND Molecular mimicry between commensal bacterial antigens and tumor-associated antigens (TAAs) has shown potential in enhancing antitumor immune responses. This study leveraged this concept using commensal bacterial antigens, termed OncoMimics, to induce TAA-derived peptide (TAAp)-specific cross-reactive cytotoxic T cells and improve the efficacy of peptide-based immunotherapies. METHODS The discovery of OncoMimics primarily relied on a bioinformatics approach to identify commensal bacteria-derived peptide sequences mimicking TAAps. Several OncoMimics peptide (OMP) candidates were selected in silico based on multiple key parameters to assess their potential to elicit and ameliorate immune responses against TAAs. Selected OMPs were synthesized and tested for their affinity and stability on the major histocompatibility complex (MHC) in vitro and for their capacity to elicit cross-reactive OMP-specific/TAAp-specific CD8+T cell responses in human leukocyte antigen (HLA)-A2-humanized mice, human peripheral blood mononuclear cells (PBMC) and patients with cancer. RESULTS Selected OMPs demonstrated superior HLA-A2 binding affinities and stabilities compared with homologous TAAps. Vaccination of HLA-A2-humanized mice with OMPs led to the expansion of OMP-specific CD8+T cells that recognize both OMPs and homologous TAAps, exhibiting cytotoxic capacities towards tumor antigens and resulting in tumor protection in a prophylactic setting. Using PBMCs from HLA-A2+healthy donors, we confirmed the ability of OMPs to elicit potent cross-reactive OMP-specific/TAAp-specific CD8+ T-cell responses. Interestingly, we observed a high prevalence of OMP-specific T cells across donors. Cytotoxicity assays revealed that OMP-stimulated human T cells specifically targeted and killed tumor cells loaded with OMPs or TAAps. Preliminary data from an ongoing clinical trial (NCT04116658) support these findings, indicating that OMPs elicit robust OMP-specific/TAAp-specific CD8+T cell responses in patients. Initial immunomonitoring data revealed sustained T-cell responses over time, with T cells maintaining a polyfunctional, cytotoxic and memory phenotype, which is critical for effective antitumor activity and long-term immune surveillance. CONCLUSIONS These findings suggest that leveraging naturally occurring commensal-derived antigens through OMPs could significantly remodel the tumor immune landscape, offering guidance for a promising strategy for cancer peptide-based immunotherapies.
Collapse
Affiliation(s)
| | - Ana Maia
- Institute for Immunology and Cluster of Excellence iFIT (EXC2180), Image-Guided and Functionally Instructed Tumor Therapies, Eberhard-Karls-University Tübingen, Tübingen, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Julie Noguerol
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), INSERM UMR1291 - CNRS UMR5051 - University Toulouse III, Toulouse, France
| | - Karl Laviolette
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), INSERM UMR1291 - CNRS UMR5051 - University Toulouse III, Toulouse, France
| | - Laura Boullerot
- Université de Franche-Comté, EFS, INSERM, UMR 1098 RIGHT, F-25000 Besançon, France
| | - Marine Malfroy
- Université de Franche-Comté, EFS, INSERM, UMR 1098 RIGHT, F-25000 Besançon, France
| | | | - Olivier Adotevi
- Université de Franche-Comté, EFS, INSERM, UMR 1098 RIGHT, F-25000 Besançon, France
| | - Olivier Joffre
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), INSERM UMR1291 - CNRS UMR5051 - University Toulouse III, Toulouse, France
| | - Ahmed Idbaih
- Sorbonne Université, AP-HP, ICM, Hôpital Universitaire La Pitié-Salpêtrière, Paris, France
| | - Maria Vieito
- Hospital Universitari Vall d'Hebron, Barcelona, Catalunya, Spain
| | | | - Agostina Stradella
- Institut Catala D'Oncologia - Hospital Duran i Reynals, Barcelona, Spain
| | - Ghazaleh Tabatabai
- Department of Neurology & Interdisciplinary Neuro-Oncology, University Hospital Tübingen, Hertie Institute for Clinical Brain Research, Center for Neuro-Oncology, Comprehensive Cancer Center, Stuttgart, Germany
| | - Michael C Burger
- Dr. Senckenberg Institute of Neurooncology, Goethe University Hospital, Frankfurt, Germany
| | - Iris Mildenberger
- Universitat Heidelberg Medizinische Fakultat Mannheim, Mannheim, Baden-Württemberg, Germany
| | - Ulrich Herrlinger
- Division of Clinical Neurooncology, Department of Neurology and Center of Integrated Oncology, University Hospital Bonn, Bonn, Nordrhein-Westfalen, Germany
| | | | - Wolfgang Wick
- Universitätsklinikum Heidelberg and German Cancer Research Center, Heidelberg, Baden-Württemberg, Germany
| | - Cecile Gouttefangeas
- Institute for Immunology and Cluster of Excellence iFIT (EXC2180), Image-Guided and Functionally Instructed Tumor Therapies, Eberhard-Karls-University Tübingen, Tübingen, Germany
| | | | | | | |
Collapse
|
7
|
Moore JA, Ali U, Vungarala S, Young-Seigler A, Tiriveedhi V. Conjugation with S4 protein transduction domain enhances the immunogenicity of the peptide vaccine against breast cancer. Mol Clin Oncol 2025; 22:20. [PMID: 39776941 PMCID: PMC11706339 DOI: 10.3892/mco.2024.2815] [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: 07/18/2024] [Accepted: 11/26/2024] [Indexed: 01/11/2025] Open
Abstract
Although peptide vaccines offer a novel venue for cancer immunotherapy, clinical success has been rather limited. Cell-penetrating peptides, due to their ability to translocate through the cell membrane, could be conjugated to the peptide vaccine to2 enhance therapeutic efficiency. The S4 transduction domain of the shaker-potassium channel was conjugated to mammaglobin-A (MamA) immunodominant epitope (MamA2.1) to verify its anticancer immunogenicity. S4-MamA2.1 peptide has demonstrated significantly higher epitope loading and stable membrane expression of HLA-A2 antigen-presenting molecules on T2 cell lines. Further, these S4-MamA2.1 treated T2 cells were able to activate naïve CD8+ T cells to induce MamA-specific cytotoxicity against breast cancer cells. Conjugation of the S4 domain has also demonstrated a slight increase in immunogenicity of lesser immunodominant MamA epitopes. The conjugation of the S4 domain to N-terminus of MamA2.1 demonstrated significantly higher immunogenicity over C-terminus conjugation. Taken together, the results of the present study suggest that conjugation of the S4 cell-penetrating peptide domain to MamA2.1 epitope enhances the peptide vaccine immunogenicity against MamA-expressing breast cancers.
Collapse
Affiliation(s)
- Jayla A. Moore
- Department of Biological Sciences, Tennessee State University, Nashville, TN 37209, USA
| | - Umer Ali
- Department of Biological Sciences, Tennessee State University, Nashville, TN 37209, USA
| | - Sunitha Vungarala
- Department of Population Sciences, Meharry-Vanderbilt Alliance, Nashville, TN 37208, USA
| | | | - Venkataswarup Tiriveedhi
- Department of Biological Sciences, Tennessee State University, Nashville, TN 37209, USA
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA
| |
Collapse
|
8
|
Graciotti M, Kandalaft LE. Vaccines for cancer prevention: exploring opportunities and navigating challenges. Nat Rev Drug Discov 2025; 24:134-150. [PMID: 39622986 DOI: 10.1038/s41573-024-01081-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/24/2024] [Indexed: 02/06/2025]
Abstract
Improved understanding of cancer immunology has gradually brought increasing attention towards cancer-preventive vaccines as an important tool in the fight against cancer. The aim of this approach is to reduce cancer occurrence by inducing a specific immune response targeting tumours at an early stage before they can fully develop. The great advantage of preventive cancer vaccines lies in the potential to harness a less-compromised immune system in vaccine recipients before their immune responses become affected by the advanced status of the disease itself or by aggressive treatments such as chemotherapy. Successful implementation of immunoprevention against oncogenic viruses such as hepatitis B and papillomavirus has led to a dramatic decrease in virally induced cancers. Extending this approach to other cancers holds great promise but remains a major challenge. Here, we provide a comprehensive review of preclinical evidence supporting this approach, encouraging results from pioneering clinical studies as well as a discussion on the key aspects and open questions to address in order to design potent prophylactic cancer vaccines in the near future.
Collapse
Affiliation(s)
- Michele Graciotti
- Center of Experimental Therapeutics, Department of Oncology, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland
- Ludwig Institute for Cancer Research, Lausanne Branch, Lausanne, Switzerland
| | - Lana E Kandalaft
- Ludwig Institute for Cancer Research, Lausanne Branch, Lausanne, Switzerland.
- Department of Oncology, University of Lausanne (UNIL), Lausanne, Switzerland.
- AGORA Cancer Research Center, Lausanne, Lausanne, Switzerland.
- Swiss Medical Network, Genolier Innovation Network, Genolier Clinic, Genolier, Switzerland.
| |
Collapse
|
9
|
Kessler AL, Pieterman RFA, Doff WAS, Bezstarosti K, Bouzid R, Klarenaar K, Jansen DTSL, Luijten RJ, Demmers JAA, Buschow SI. HLA I immunopeptidome of synthetic long peptide pulsed human dendritic cells for therapeutic vaccine design. NPJ Vaccines 2025; 10:12. [PMID: 39827205 PMCID: PMC11742953 DOI: 10.1038/s41541-025-01069-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 01/03/2025] [Indexed: 01/22/2025] Open
Abstract
Synthetic long peptides (SLPs) are a promising vaccine modality that exploit dendritic cells (DC) to treat chronic infections or cancer. Currently, the design of SLPs relies on in silico prediction and multifactorial T cells assays to determine which SLPs are best cross-presented on DC human leukocyte antigen class I (HLA-I). Furthermore, it is unknown how TLR ligand-based adjuvants affect DC cross-presentation. Here, we generated a unique, high-quality immunopeptidome dataset of human DCs pulsed with 12 hepatitis B virus (HBV)-based SLPs combined with either a TLR1/2 (Amplivant®) or TLR3 (PolyI:C) ligand. The obtained immunopeptidome reflected adjuvant-induced differences, but no differences in cross-presentation of SLPs. We uncovered dominant (cross-)presentation on B-alleles, and identified 33 unique SLP-derived HLA-I peptides, several of which were not in silico predicted and some were consistently found across donors. Our work puts forward DC immunopeptidomics as a valuable tool for therapeutic vaccine design.
Collapse
Affiliation(s)
- Amy L Kessler
- Department of Gastroenterology & Hepatology, Erasmus MC University Medical Center Rotterdam, Rotterdam, The Netherlands
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical sciences, University of Utrecht, Utrecht, The Netherlands
| | - Roel F A Pieterman
- Department of Gastroenterology & Hepatology, Erasmus MC University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Wouter A S Doff
- Proteomics Center, Department of Biochemistry, Erasmus MC University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Karel Bezstarosti
- Proteomics Center, Department of Biochemistry, Erasmus MC University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Rachid Bouzid
- Department of Gastroenterology & Hepatology, Erasmus MC University Medical Center Rotterdam, Rotterdam, The Netherlands
- Merus N.V., Utrecht, The Netherlands
| | - Kim Klarenaar
- Department of Gastroenterology & Hepatology, Erasmus MC University Medical Center Rotterdam, Rotterdam, The Netherlands
- Division of Laboratories, Pharmacy and Biomedical Genetics, UMC Utrecht, Utrecht, The Netherlands
| | - Diahann T S L Jansen
- Department of Gastroenterology & Hepatology, Erasmus MC University Medical Center Rotterdam, Rotterdam, The Netherlands
- Department of Internal Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Robbie J Luijten
- Department of Gastroenterology & Hepatology, Erasmus MC University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Jeroen A A Demmers
- Proteomics Center, Department of Biochemistry, Erasmus MC University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Sonja I Buschow
- Department of Gastroenterology & Hepatology, Erasmus MC University Medical Center Rotterdam, Rotterdam, The Netherlands.
| |
Collapse
|
10
|
Zhao W, Li X, Guan J, Yan S, Teng L, Sun X, Dong Y, Wang H, Tao W. Potential and development of cellular vesicle vaccines in cancer immunotherapy. Discov Oncol 2025; 16:48. [PMID: 39812959 PMCID: PMC11735706 DOI: 10.1007/s12672-025-01781-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Accepted: 01/06/2025] [Indexed: 01/16/2025] Open
Abstract
Cancer vaccines are promising as an effective means of stimulating the immune system to clear tumors as well as to establish immune surveillance. In this paper, we discuss the main platforms and current status of cancer vaccines and propose a new cancer vaccine platform, the cytosolic vesicle vaccine. This vaccine has a unique structure that can integrate antigen and adjuvant carriers to improve the delivery efficiency and immune activation ability, which brings new ideas for cancer vaccine design. Tumor exosomes carry antigens and MHC-peptide complexes, which can provide tumor antigens to antigen-processing cells and increase the chances of recognition of tumor antigens by immune cells. DEVs play a role in amplifying the immune response by acting as carriers for the dissemination of antigenic substances in dendritic cells. OMVs, with their natural adjuvant properties, are one of the advantages for the preparation of antitumor vaccines. This paper presents the advantages of these three bacteria/extracellular vesicles as cancer vaccines and discusses the potential applications of functionally modified extracellular vesicles as cancer vaccines after cellular engineering or genetic engineering, as well as current clinical trials of extracellular vesicle vaccines. In summary, extracellular vesicle vaccines are a promising direction for cancer vaccine research.
Collapse
Affiliation(s)
- Wenxi Zhao
- Department of Breast Surgery, The First Affiliated Hospital of Harbin Medical University, No. 23, Youzheng Street, Nangang District, Harbin, 150001, China
| | - Xianjun Li
- Department of Breast Surgery, The First Affiliated Hospital of Harbin Medical University, No. 23, Youzheng Street, Nangang District, Harbin, 150001, China
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin, 150081, China
| | - Jialu Guan
- Department of Breast Surgery, The First Affiliated Hospital of Harbin Medical University, No. 23, Youzheng Street, Nangang District, Harbin, 150001, China
| | - Shuai Yan
- Department of Breast Surgery, The First Affiliated Hospital of Harbin Medical University, No. 23, Youzheng Street, Nangang District, Harbin, 150001, China
| | - Lizhi Teng
- Department of Breast Surgery, The First Affiliated Hospital of Harbin Medical University, No. 23, Youzheng Street, Nangang District, Harbin, 150001, China
| | - Xitong Sun
- Department of Breast Surgery, The First Affiliated Hospital of Harbin Medical University, No. 23, Youzheng Street, Nangang District, Harbin, 150001, China
| | - Yuhan Dong
- Department of Breast Surgery, The First Affiliated Hospital of Harbin Medical University, No. 23, Youzheng Street, Nangang District, Harbin, 150001, China
| | - Hongyue Wang
- Department of Breast Surgery, The First Affiliated Hospital of Harbin Medical University, No. 23, Youzheng Street, Nangang District, Harbin, 150001, China
| | - Weiyang Tao
- Department of Breast Surgery, The First Affiliated Hospital of Harbin Medical University, No. 23, Youzheng Street, Nangang District, Harbin, 150001, China.
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, China.
| |
Collapse
|
11
|
Wang S, Jiang S, Li X, Huang H, Qiu X, Yu M, Yang X, Liu F, Wang C, Shen W, Wang Y, Wang B. FGL2 172-220 peptides improve the antitumor effect of HCMV-IE1mut vaccine against glioblastoma by modulating immunosuppressive cells in the tumor microenvironment. Oncoimmunology 2024; 13:2423983. [PMID: 39508842 PMCID: PMC11542393 DOI: 10.1080/2162402x.2024.2423983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2024] Open
Abstract
Glioblastoma multiforme (GBM) is a highly aggressive primary brain tumor characterized by poor prognosis and lack of effective treatments. In recent years, peptide vaccines that use sequences based on tumor-specific or tumor-associated antigens to activate immune responses against tumor cells have emerged as a new therapeutic strategy. In this study, we developed a novel therapeutic polypeptide vaccine targeting the tumor-associated antigen Fibrinogen-Like Protein 2 (FGL2), whose dominant epitope peptide was tandemly linked to the C-terminus of HCMV-IE1mut via a linker. We used this vaccine to compare the therapeutic efficacy of HCMV-IE1mut alone versus HCMV-IE1mut-FGL2172-220 and investigate the potential mechanism of action of HCMV-IE1mut-FGL2172-220 in glioma treatment. An in situ GBM model (GL261-IE1-luc cells) was used to determine the efficacy of the vaccine. Treatment with HCMV-IE1mut-FGL2172-220 exerted antitumor effects and extended the survival of the GL261 animal model. We observed reduced proportions of microglia, regulatory T cells (Treg), and myeloid-derived suppressor cells (MDSC) in the tumor microenvironment (TME) by immunofluorescence. Flow cytometry showed that compared to HCMV-IE1mut alone, treatment with HCMV-IE1mut-FGL2172-220 increased the proportion of CD8+ T cells and tissue-resident memory T cells (TRM). ELISA analysis showed that it improved the secretion of tumor-specific IFN-γ and TNF-α by these cells and downregulated the expression of IL-6 and IL-10. Our study demonstrates that the long-peptide FGL2172-220 improves the antitumor efficacy of HCMV-IE1mut, possibly by reshaping immune cells in the glioma microenvironment. These findings lay the groundwork for the development of therapeutic antigenic peptide vaccines to improve antitumor effects for cancer.
Collapse
Affiliation(s)
- Shan Wang
- Department of Pathogenic Biology, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Shasha Jiang
- Department of Clinical Laboratory, Honghui Hospital, Xi’an Jiaotong University, Xi’an, China
| | - Xu Li
- Department of Pathogenic Biology, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Huan Huang
- Department of Pathogenic Biology, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Xu Qiu
- Department of Pathogenic Biology, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Meng Yu
- Department of Pathogenic Biology, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Xiaoli Yang
- Department of Pathogenic Biology, School of Basic Medicine, Qingdao University, Qingdao, China
| | | | - Chen Wang
- Department of Pathogenic Biology, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Wen Shen
- Department of Pathogenic Biology, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Yunyang Wang
- Department of Endocrinology and Metabolism, the Affiliated Hospital of Qingdao University, Qingdao, China
| | - Bin Wang
- Department of Pathogenic Biology, School of Basic Medicine, Qingdao University, Qingdao, China
| |
Collapse
|
12
|
Zeng YY, Gu Q, Li D, Li AX, Liu RM, Liang JY, Liu JY. Immunocyte membrane-derived biomimetic nano-drug delivery system: a pioneering platform for tumour immunotherapy. Acta Pharmacol Sin 2024; 45:2455-2473. [PMID: 39085407 PMCID: PMC11579519 DOI: 10.1038/s41401-024-01355-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 07/03/2024] [Indexed: 08/02/2024]
Abstract
Tumor immunotherapy characterized by its high specificity and minimal side effects has achieved revolutionary progress in the field of cancer treatment. However, the complex mechanisms of tumor immune microenvironment (TIME) and the individual variability of patients' immune system still present significant challenges to its clinical application. Immunocyte membrane-coated nanocarrier systems, as an innovative biomimetic drug delivery platform, exhibit remarkable advantages in tumor immunotherapy due to their high targeting capability, good biocompatibility and low immunogenicity. In this review we summarize the latest research advances in biomimetic delivery systems based on immune cells for tumor immunotherapy. We outline the existing methods of tumor immunotherapy including immune checkpoint therapy, adoptive cell transfer therapy and cancer vaccines etc. with a focus on the application of various immunocyte membranes in tumor immunotherapy and their prospects and challenges in drug delivery and immune modulation. We look forward to further exploring the application of biomimetic delivery systems based on immunocyte membrane-coated nanoparticles, aiming to provide a new framework for the clinical treatment of tumor immunity.
Collapse
Affiliation(s)
- Yuan-Ye Zeng
- School of Pharmacy, Fudan University, Shanghai, 201203, China
- Department of Pharmacy, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Qing Gu
- Department of Pharmacy, Jingan District Zhabei Central Hospital, Shanghai, 200070, China
| | - Dan Li
- Department of Pharmacy, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Ai-Xue Li
- Department of Pharmacy, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Rong-Mei Liu
- Department of Pharmacy, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Jian-Ying Liang
- School of Pharmacy, Fudan University, Shanghai, 201203, China.
| | - Ji-Yong Liu
- Department of Pharmacy, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
13
|
Han H, Santos HA. Nano- and Micro-Platforms in Therapeutic Proteins Delivery for Cancer Therapy: Materials and Strategies. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2409522. [PMID: 39263818 DOI: 10.1002/adma.202409522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 08/26/2024] [Indexed: 09/13/2024]
Abstract
Proteins have emerged as promising therapeutics in oncology due to their great specificity. Many treatment strategies are developed based on protein biologics, such as immunotherapy, starvation therapy, and pro-apoptosis therapy, while some protein biologics have entered the clinics. However, clinical translation is severely impeded by instability, short circulation time, poor transmembrane transportation, and immunogenicity. Micro- and nano-particles-based drug delivery platforms are designed to solve those problems and enhance protein therapeutic efficacy. This review first summarizes the different types of therapeutic proteins in clinical and research stages, highlighting their administration limitations. Next, various types of micro- and nano-particles are described to demonstrate how they can overcome those limitations. The potential of micro- and nano-particles are then explored to enhance the therapeutic efficacy of proteins by combinational therapies. Finally, the challenges and future directions of protein biologics carriers are discussed for optimized protein delivery.
Collapse
Affiliation(s)
- Huijie Han
- Department of Biomaterials and Biomedical Technology, The Personalized Medicine Research Institute (PRECISION), University Medical Center Groningen (UMCG), University of Groningen, Ant. Deusinglaan 1, Groningen, 9713 AV, The Netherlands
| | - Hélder A Santos
- Department of Biomaterials and Biomedical Technology, The Personalized Medicine Research Institute (PRECISION), University Medical Center Groningen (UMCG), University of Groningen, Ant. Deusinglaan 1, Groningen, 9713 AV, The Netherlands
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, FI-00014, Finland
| |
Collapse
|
14
|
Pollack IF, Felker J, Frederico SC, Raphael I, Kohanbash G. Immunotherapy for pediatric low-grade gliomas. Childs Nerv Syst 2024; 40:3263-3275. [PMID: 38884777 DOI: 10.1007/s00381-024-06491-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 06/01/2024] [Indexed: 06/18/2024]
Abstract
Pediatric low-grade gliomas (pLGGs) are the most common brain tumor types affecting children. Although gross-total resection remains the treatment of choice, many tumors are not amenable to complete removal, because they either involve midline structures, such as the optic chiasm or hypothalamus, and are not conducive to aggressive resection, or have diffuse biological features and blend with the surrounding brain. Historically, radiation therapy was used as the second-line option for disease control, but with the recognition that this often led to adverse long-term sequelae, particularly in young children, conventional chemotherapy assumed a greater role in initial therapy for unresectable tumors. A variety of agents demonstrated activity, but long-term disease control was suboptimal, with more than 50% of tumors exhibiting disease progression within 5 years. More recently, it has been recognized that a high percentage of these tumors in children exhibit constitutive activation of the mitogen-activated protein kinase (MAPK) pathway because of BRAF translocations or mutations, NFI mutations, or a host of other anomalies that converged on MAPK. This led to phase 1, 2, and 3 trials that explored the activity of blocking this signaling pathway, and the efficacy of this approach compared to conventional chemotherapy. Despite initial promise of these strategies, not all children tolerate this therapy, and many tumors resume growth once MAPK inhibition is stopped, raising concern that long-term and potentially life-long treatment will be required to maintain tumor control, even among responders. This observation has led to interest in other treatments, such as immunotherapy, that may delay or avoid the need for additional treatments. This chapter will summarize the place of immunotherapy in the current armamentarium for these tumors and discuss prior results and future options to improve disease control, with a focus on our prior efforts and experience in this field.
Collapse
Affiliation(s)
- Ian F Pollack
- Department of Neurosurgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
- University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
- Department of Neurosurgery, UPMC Children's Hospital of Pittsburgh, 4401 Penn Avenue, Pittsburgh, PA, 15224, USA.
| | - James Felker
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Neurosurgery, UPMC Children's Hospital of Pittsburgh, 4401 Penn Avenue, Pittsburgh, PA, 15224, USA
| | - Stephen C Frederico
- University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Neurosurgery, UPMC Children's Hospital of Pittsburgh, 4401 Penn Avenue, Pittsburgh, PA, 15224, USA
| | - Itay Raphael
- Department of Neurosurgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Neurosurgery, UPMC Children's Hospital of Pittsburgh, 4401 Penn Avenue, Pittsburgh, PA, 15224, USA
| | - Gary Kohanbash
- University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Neurosurgery, UPMC Children's Hospital of Pittsburgh, 4401 Penn Avenue, Pittsburgh, PA, 15224, USA
| |
Collapse
|
15
|
Bojarska J, Wolf WM. Short Peptides as Powerful Arsenal for Smart Fighting Cancer. Cancers (Basel) 2024; 16:3254. [PMID: 39409876 PMCID: PMC11476321 DOI: 10.3390/cancers16193254] [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: 08/19/2024] [Revised: 09/18/2024] [Accepted: 09/23/2024] [Indexed: 10/20/2024] Open
Abstract
Short peptides have been coming around as a strong weapon in the fight against cancer on all fronts-in immuno-, chemo-, and radiotherapy, and also in combinatorial approaches. Moreover, short peptides have relevance in cancer imaging or 3D culture. Thanks to the natural 'smart' nature of short peptides, their unique structural features, as well as recent progress in biotechnological and bioinformatics development, short peptides are playing an enormous role in evolving cutting-edge strategies. Self-assembling short peptides may create excellent structures to stimulate cytotoxic immune responses, which is essential for cancer immunotherapy. Short peptides can help establish versatile strategies with high biosafety and effectiveness. Supramolecular short peptide-based cancer vaccines entered clinical trials. Peptide assemblies can be platforms for the delivery of antigens, adjuvants, immune cells, and/or drugs. Short peptides have been unappreciated, especially in the vaccine aspect. Meanwhile, they still hide the undiscovered unlimited potential. Here, we provide a timely update on this highly active and fast-evolving field.
Collapse
Affiliation(s)
- Joanna Bojarska
- Chemistry Department, Institute of Inorganic and Ecological Chemistry, Łódź University of Technology, S. Żeromskiego Str. 116, 90-924 Łódź, Poland;
| | | |
Collapse
|
16
|
Kandaswamy K, Guru A. Harnessing virulence factor-derived peptides for innovative oral cancer treatment. Nat Prod Res 2024:1-2. [PMID: 39105452 DOI: 10.1080/14786419.2024.2387250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 07/29/2024] [Indexed: 08/07/2024]
Affiliation(s)
- Karthikeyan Kandaswamy
- Department of Cariology, Saveetha Dental College and Hospital, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, Tamil Nadu, India
| | - Ajay Guru
- Department of Cariology, Saveetha Dental College and Hospital, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, Tamil Nadu, India
| |
Collapse
|
17
|
Sun H, Wei M, Guo A, Zhang C, Wang Y, Huang R, Li X, Zhan J, Wu J, Jiang B. Shrimp hemocyanin elicits a potent humoral response in mammals and is favorable to hapten conjugation. Sci Rep 2024; 14:16771. [PMID: 39039159 PMCID: PMC11263335 DOI: 10.1038/s41598-024-67715-1] [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: 03/13/2024] [Accepted: 07/15/2024] [Indexed: 07/24/2024] Open
Abstract
Conjugation to a carrier protein is essential to give rise to the antigenicity of hapten. Three carrier proteins e.g. KLH (Keyhole Limpet hemocyanin), BSA (bovine serum albumin), and OVA (Ovalbumin) were used mostly. KLH is advantageous to the others, majorly owing to its strong immunogenicity and limited usage in other biological assays. However, the cost of obtaining Keyhole Limpet is high and the solubility of KLH is not as well as the other carriers, especially after hapten conjugation. Here, we extracted the shrimp hemocyanin (SHC) from Litopenaeus vannamei (L. vannamei), which is a commonly sea product worldwide. The high pure SHC could be acquired by two-step purification, with a production yield of > 1 g proteins (98% pure) per 1 kg shrimp. Compared to KLH, the peptide-SHC conjugates exhibit higher solubility after hapten conjugation. Meanwhile, compared with KLH, SHC induces comparable antibody production efficiency in mammals, with or without conjugation. Furthermore, rabbit polyclonal antibodies or mouse monoclonal antibodies were generated by immunizing SHC-peptide conjugates, and the subsequent antibodies were confirmed to be used in western blot, immunofluorescence and immunohistochemistry. Therefore, we demonstrated that SHC may be used as a substitute for KLH in future antibody and vaccine development.
Collapse
Affiliation(s)
- Huiwen Sun
- Shanghai Epizyme BioMedical, Shanghai, China
| | - Moris Wei
- Shanghai Epizyme BioMedical, Shanghai, China
| | - Amber Guo
- Shanghai Epizyme BioMedical, Shanghai, China
| | - Ci Zhang
- Shanghai Epizyme BioMedical, Shanghai, China
| | - Yuefeng Wang
- Shanghai Epizyme BioMedical, Shanghai, China
- Shanghai Yilawo Biotech., Shanghai, China
| | - Renhui Huang
- Shanghai Epizyme BioMedical, Shanghai, China
- Tongling Epizyme BioMedical, Tongling, China
| | - Xiaoxiao Li
- Shanghai Epizyme BioMedical, Shanghai, China
| | | | - Jonny Wu
- Shanghai Epizyme BioMedical, Shanghai, China.
- Tongling Epizyme BioMedical, Tongling, China.
| | - Bruce Jiang
- Shanghai Epizyme BioMedical, Shanghai, China.
- Tongling Epizyme BioMedical, Tongling, China.
| |
Collapse
|
18
|
Azzi L, Celesti F, Chiaravalli AM, Shaik AKB, Shallak M, Gatta A, Battaglia P, La Rosa S, Tagliabue A, Accolla RS, Forlani G. Novel vaccination strategies based on optimal stimulation of CD4 + T helper cells for the treatment of oral squamous cell carcinoma. Front Immunol 2024; 15:1387835. [PMID: 39035008 PMCID: PMC11257872 DOI: 10.3389/fimmu.2024.1387835] [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: 02/18/2024] [Accepted: 06/13/2024] [Indexed: 07/23/2024] Open
Abstract
Oral Squamous Cell Carcinoma (OSCC) is the most common malignant tumor of the oral cavity. Despite recent advances in the field of oral cancer therapy, including the introduction of immunotherapeutic approaches, the 5-year survival rate remains steadily assessed around 50%. Thus, there is an urgent need for new therapeutic strategies. After the characterization of the immune phenotype of three human OSCC cell lines (CAL-27, SCC-25, and SCC-4) and one mouse OSCC cell line (MOC2) showing their similarities to resected patient tumors, we explored for the first time an experimental preclinical model of therapeutic vaccination with mouse OSCC MOC2 cell line stably expressing MHC class II antigens after CIITA gene transfection (MOC2-CIITA). Mice injected with MOC2-CIITA reject or strongly retard tumor growth; more importantly, vaccinated animals that fully reject MOC2-CIITA tumors display anti-tumor immunological memory protective against challenge with parental MOC2 tumor cells. Further experiments of adoptive cell transfer or in vivo cell depletion show that both CD4+ and CD8+ T lymphocytes prove fundamental in tumor rejection. This unprecedented approach for oral cancer opens the way for possible future translation of novel immunotherapeutic strategies to the human setting for the treatment of this tumor.
Collapse
Affiliation(s)
- Lorenzo Azzi
- Department of Medicine and Technological Innovation, University of Insubria, Varese, Italy
- Azienda Socio-Sanitaria Territoriale (ASST) dei Sette Laghi, Varese, Italy
| | - Fabrizio Celesti
- Center for Immuno-Oncology, Department of Medicine, Surgery, and Neurosciences, University of Siena, Siena, Italy
| | | | | | - Mariam Shallak
- Department of Medicine and Technological Innovation, University of Insubria, Varese, Italy
| | - Andrea Gatta
- Department of Medicine and Technological Innovation, University of Insubria, Varese, Italy
| | - Paolo Battaglia
- Department of Biotechnology and Life Sciences, University of Insubria, Varese, Italy
- Azienda Socio-Sanitaria Territoriale (ASST) Lariana, San Fermo della Battaglia, CO, Italy
| | - Stefano La Rosa
- Department of Medicine and Technological Innovation, University of Insubria, Varese, Italy
- Azienda Socio-Sanitaria Territoriale (ASST) dei Sette Laghi, Varese, Italy
| | - Angelo Tagliabue
- Azienda Socio-Sanitaria Territoriale (ASST) dei Sette Laghi, Varese, Italy
- Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Roberto Sergio Accolla
- Department of Medicine and Technological Innovation, University of Insubria, Varese, Italy
| | - Greta Forlani
- Department of Medicine and Technological Innovation, University of Insubria, Varese, Italy
| |
Collapse
|
19
|
Cen L, Zhang Z, Sun Y, Wu N, Shao J, Qian Z, Tian M, Ke Y, Liu B. Efficacy of MAGE-A4 long peptide as a universal immunoprevention cancer vaccine. Cancer Cell Int 2024; 24:232. [PMID: 38961429 PMCID: PMC11223347 DOI: 10.1186/s12935-024-03421-2] [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: 01/03/2024] [Accepted: 06/22/2024] [Indexed: 07/05/2024] Open
Abstract
BACKGROUND The clinical application of peptide vaccines in tumor immunotherapy holds significant promise. Peptide-based tumor vaccines are currently subject to certain limitations in clinical trials, including the challenge of inducing a sustained response from CD4+ T helper cells and cytotoxic T lymphocytes (CTL), as well as human leukocyte antigen (HLA) restrictions. METHODS Through the utilization of biological information methodology, a screening process was conducted to identify three potential long peptides that are specifically targeted by the MAGE-A4 antigen. The candidate long peptides were subjected to in vitro testing using human peripheral blood lymphocytes as samples to evaluate their immunogenicity and immune function. The antitumor properties and preliminary mechanism of the long peptide vaccine were investigated through the use of a mouse model designed for the prevention of triple negative breast cancer (TNBC). RESULTS Three predicted multi-epitope long peptides targeting MAGE-A4 have shown to have a strong immunogenicity, with a total positive rate of 72% across different HLA subtypes in Chinese populations. they can also increase the levels of the costimulatory factor CD137 and tumor necrosis factor-alpha (TNF-α), activate T cells, and boost the cytotoxic activity. Results from an animal study have revealed that the long-peptide vaccine, both on its own and in combination with R848, has displayed impressive anti-tumor and target-specific capabilities. Moreover, it has the ability to increase the expression of effector memory T cells and central memory T cells. CONCLUSIONS This study was the first to screen three multi-epitope long peptides targeting MAGE-A4 and assess their immunogenicity, immune function, and potential as adjuvant peptides. The results showed that the MAGE-A4 long peptide vaccine can be used as a novel immunoprophylaxis method to prevent TNBC. Moreover, the proposed development model is capable of screening multiple target antigens, which lead to its clinical application.
Collapse
Affiliation(s)
- Lanqi Cen
- Department of Oncology, China Pharmaceutical University Nanjing Drum Tower Hospital, Nanjing, 210000, China
| | - Zhe Zhang
- Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221000, China
| | - Yi Sun
- Department of Oncology, Affiliated Hospital of Medical School, Nanjing Drum Tower Hospital, Nanjing University, Nanjing, 210000, China
| | - Nandie Wu
- Department of Oncology, Affiliated Hospital of Medical School, Nanjing Drum Tower Hospital, Nanjing University, Nanjing, 210000, China
| | - Jie Shao
- Department of Oncology, Affiliated Hospital of Medical School, Nanjing Drum Tower Hospital, Nanjing University, Nanjing, 210000, China
| | - Zhaoye Qian
- Department of Oncology, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, 210000, China
| | - Manman Tian
- Department of Oncology, Affiliated Hospital of Medical School, Nanjing Drum Tower Hospital, Nanjing University, Nanjing, 210000, China
| | - Yaohua Ke
- Department of Oncology, Affiliated Hospital of Medical School, Nanjing Drum Tower Hospital, Nanjing University, Nanjing, 210000, China
| | - Baorui Liu
- Department of Oncology, China Pharmaceutical University Nanjing Drum Tower Hospital, Nanjing, 210000, China.
- Department of Oncology, Affiliated Hospital of Medical School, Nanjing Drum Tower Hospital, Nanjing University, Nanjing, 210000, China.
- Department of Oncology, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, 210000, China.
| |
Collapse
|
20
|
Lei X, Ban J, Wu Z, Cao S, Zhou M, Zhang L, Zhu R, Lu H, Zhu S. Boosting PRRSV-Specific Cellular Immunity: The Immunological Profiling of an Fc-Fused Multi-CTL Epitope Vaccine in Mice. Vet Sci 2024; 11:274. [PMID: 38922021 PMCID: PMC11209284 DOI: 10.3390/vetsci11060274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Revised: 06/06/2024] [Accepted: 06/14/2024] [Indexed: 06/27/2024] Open
Abstract
The continuously evolving PRRSV has been plaguing pig farms worldwide for over 30 years, with conventional vaccines suffering from insufficient protection and biosecurity risks. To address these challenges, we identified 10 PRRSV-specific CTL epitopes through enzyme-linked immunospot assay (ELISPOT) and constructed a multi-epitope peptide (PTE) by linking them in tandem. This PTE was then fused with a modified porcine Fc molecule to create the recombinant protein pFc-PTE. Our findings indicate that pFc-PTE effectively stimulates PRRSV-infected specific splenic lymphocytes to secrete high levels of interferon-gamma (IFN-γ) and is predicted to be non-toxic and non-allergenic. Compared to PTE alone, pFc-PTE not only induced a comparable cellular immune response in mice but also extended the duration of the immune response to at least 10 weeks post-immunization. Additionally, pFc-PTE predominantly induced a Th1 immune response, suggesting its potential advantage in enhancing cellular immunity. Consequently, pFc-PTE holds promise as a novel, safe, and potent candidate vaccine for PRRSV and may also provide new perspectives for vaccine design against other viral diseases.
Collapse
Affiliation(s)
- Xinnuo Lei
- Jiangsu Key Laboratory for High-Tech Research and Development of Veterinary Biopharmaceuticals, Engineering Technology Research Center for Modern Animal Science and Novel Veterinary Pharmaceutic Development, Jiangsu Agri-Animal Husbandry Vocational College, Taizhou 225300, China; (X.L.); (J.B.); (Z.W.); (S.C.); (M.Z.); (L.Z.); (R.Z.); (H.L.)
| | - Jinzhao Ban
- Jiangsu Key Laboratory for High-Tech Research and Development of Veterinary Biopharmaceuticals, Engineering Technology Research Center for Modern Animal Science and Novel Veterinary Pharmaceutic Development, Jiangsu Agri-Animal Husbandry Vocational College, Taizhou 225300, China; (X.L.); (J.B.); (Z.W.); (S.C.); (M.Z.); (L.Z.); (R.Z.); (H.L.)
- Ministry of Agriculture Key Laboratory of Animal Bacteriology, International Joint Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Zhi Wu
- Jiangsu Key Laboratory for High-Tech Research and Development of Veterinary Biopharmaceuticals, Engineering Technology Research Center for Modern Animal Science and Novel Veterinary Pharmaceutic Development, Jiangsu Agri-Animal Husbandry Vocational College, Taizhou 225300, China; (X.L.); (J.B.); (Z.W.); (S.C.); (M.Z.); (L.Z.); (R.Z.); (H.L.)
| | - Shinuo Cao
- Jiangsu Key Laboratory for High-Tech Research and Development of Veterinary Biopharmaceuticals, Engineering Technology Research Center for Modern Animal Science and Novel Veterinary Pharmaceutic Development, Jiangsu Agri-Animal Husbandry Vocational College, Taizhou 225300, China; (X.L.); (J.B.); (Z.W.); (S.C.); (M.Z.); (L.Z.); (R.Z.); (H.L.)
| | - Mo Zhou
- Jiangsu Key Laboratory for High-Tech Research and Development of Veterinary Biopharmaceuticals, Engineering Technology Research Center for Modern Animal Science and Novel Veterinary Pharmaceutic Development, Jiangsu Agri-Animal Husbandry Vocational College, Taizhou 225300, China; (X.L.); (J.B.); (Z.W.); (S.C.); (M.Z.); (L.Z.); (R.Z.); (H.L.)
| | - Li Zhang
- Jiangsu Key Laboratory for High-Tech Research and Development of Veterinary Biopharmaceuticals, Engineering Technology Research Center for Modern Animal Science and Novel Veterinary Pharmaceutic Development, Jiangsu Agri-Animal Husbandry Vocational College, Taizhou 225300, China; (X.L.); (J.B.); (Z.W.); (S.C.); (M.Z.); (L.Z.); (R.Z.); (H.L.)
| | - Rui Zhu
- Jiangsu Key Laboratory for High-Tech Research and Development of Veterinary Biopharmaceuticals, Engineering Technology Research Center for Modern Animal Science and Novel Veterinary Pharmaceutic Development, Jiangsu Agri-Animal Husbandry Vocational College, Taizhou 225300, China; (X.L.); (J.B.); (Z.W.); (S.C.); (M.Z.); (L.Z.); (R.Z.); (H.L.)
| | - Huipeng Lu
- Jiangsu Key Laboratory for High-Tech Research and Development of Veterinary Biopharmaceuticals, Engineering Technology Research Center for Modern Animal Science and Novel Veterinary Pharmaceutic Development, Jiangsu Agri-Animal Husbandry Vocational College, Taizhou 225300, China; (X.L.); (J.B.); (Z.W.); (S.C.); (M.Z.); (L.Z.); (R.Z.); (H.L.)
| | - Shanyuan Zhu
- Jiangsu Key Laboratory for High-Tech Research and Development of Veterinary Biopharmaceuticals, Engineering Technology Research Center for Modern Animal Science and Novel Veterinary Pharmaceutic Development, Jiangsu Agri-Animal Husbandry Vocational College, Taizhou 225300, China; (X.L.); (J.B.); (Z.W.); (S.C.); (M.Z.); (L.Z.); (R.Z.); (H.L.)
| |
Collapse
|
21
|
Strum S, Andersen MH, Svane IM, Siu LL, Weber JS. State-Of-The-Art Advancements on Cancer Vaccines and Biomarkers. Am Soc Clin Oncol Educ Book 2024; 44:e438592. [PMID: 38669611 DOI: 10.1200/edbk_438592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/28/2024]
Abstract
The origins of cancer vaccines date back to the 1800s. Since then, there have been significant efforts to generate vaccines against solid and hematologic malignancies using a variety of platforms. To date, these efforts have generally been met with minimal success. However, in the era of improved methods and technological advancements, supported by compelling preclinical and clinical data, a wave of renewed interest in the field offers the promise of discovering field-changing paradigms in the management of established and resected disease using cancer vaccines. These include novel approaches to personalized neoantigen vaccine development, as well as innovative immune-modulatory vaccines (IMVs) that facilitate activation of antiregulatory T cells to limit immunosuppression caused by regulatory immune cells. This article will introduce some of the limitations that have affected cancer vaccine development over the past several decades, followed by an introduction to the latest advancements in neoantigen vaccine and IMV therapy, and then conclude with a discussion of some of the newest technologies and progress that are occurring across the cancer vaccine space. Cancer vaccines are among the most promising frontiers for breakthrough innovations and strategies poised to make a measurable impact in the ongoing fight against cancer.
Collapse
Affiliation(s)
- Scott Strum
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, Canada
| | - Mads Hald Andersen
- National Center for Cancer Immune Therapy, Department of Oncology, Copenhagen University Hospital - Herlev and Gentofte, Herlev, Denmark
| | - Inge Marie Svane
- National Center for Cancer Immune Therapy, Department of Oncology, Copenhagen University Hospital - Herlev and Gentofte, Herlev, Denmark
| | - Lillian L Siu
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, Canada
| | - Jeffrey S Weber
- Laura and Isaac Perlmutter Cancer Center, NYU Langone Health, New York, NY
| |
Collapse
|
22
|
Moon DO. Deciphering the Role of BCAR3 in Cancer Progression: Gene Regulation, Signal Transduction, and Therapeutic Implications. Cancers (Basel) 2024; 16:1674. [PMID: 38730626 PMCID: PMC11083344 DOI: 10.3390/cancers16091674] [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: 04/10/2024] [Revised: 04/24/2024] [Accepted: 04/24/2024] [Indexed: 05/13/2024] Open
Abstract
This review comprehensively explores the gene BCAR3, detailing its regulation at the gene, mRNA, and protein structure levels, and delineating its multifunctional roles in cellular signaling within cancer contexts. The discussion covers BCAR3's involvement in integrin signaling and its impact on cancer cell migration, its capability to induce anti-estrogen resistance, and its significant functions in cell cycle regulation. Further highlighted is BCAR3's modulation of immune responses within the tumor microenvironment, a novel area of interest that holds potential for innovative cancer therapies. Looking forward, this review outlines essential future research directions focusing on transcription factor binding studies, isoform-specific expression profiling, therapeutic targeting of BCAR3, and its role in immune cell function. Each segment builds towards a holistic understanding of BCAR3's operational mechanisms, presenting a critical evaluation of its therapeutic potential in oncology. This synthesis aims to not only extend current knowledge but also catalyze further research that could pivotally influence the development of targeted cancer treatments.
Collapse
Affiliation(s)
- Dong Oh Moon
- Department of Biology Education, Daegu University, 201 Daegudae-ro, Gyeongsan-si 38453, Gyeongsangbuk-do, Republic of Korea
| |
Collapse
|
23
|
Sheikhlary S, Lopez DH, Moghimi S, Sun B. Recent Findings on Therapeutic Cancer Vaccines: An Updated Review. Biomolecules 2024; 14:503. [PMID: 38672519 PMCID: PMC11048403 DOI: 10.3390/biom14040503] [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: 02/23/2024] [Revised: 04/06/2024] [Accepted: 04/16/2024] [Indexed: 04/28/2024] Open
Abstract
Cancer remains one of the global leading causes of death and various vaccines have been developed over the years against it, including cell-based, nucleic acid-based, and viral-based cancer vaccines. Although many vaccines have been effective in in vivo and clinical studies and some have been FDA-approved, there are major limitations to overcome: (1) developing one universal vaccine for a specific cancer is difficult, as tumors with different antigens are different for different individuals, (2) the tumor antigens may be similar to the body's own antigens, and (3) there is the possibility of cancer recurrence. Therefore, developing personalized cancer vaccines with the ability to distinguish between the tumor and the body's antigens is indispensable. This paper provides a comprehensive review of different types of cancer vaccines and highlights important factors necessary for developing efficient cancer vaccines. Moreover, the application of other technologies in cancer therapy is discussed. Finally, several insights and conclusions are presented, such as the possibility of using cold plasma and cancer stem cells in developing future cancer vaccines, to tackle the major limitations in the cancer vaccine developmental process.
Collapse
Affiliation(s)
- Sara Sheikhlary
- Department of Biomedical Engineering, College of Engineering, The University of Arizona, Tucson, AZ 85721, USA
| | - David Humberto Lopez
- Department of Pharmacology and Toxicology, College of Pharmacy, The University of Arizona, Tucson, AZ 85721, USA; (D.H.L.); (S.M.)
| | - Sophia Moghimi
- Department of Pharmacology and Toxicology, College of Pharmacy, The University of Arizona, Tucson, AZ 85721, USA; (D.H.L.); (S.M.)
| | - Bo Sun
- Department of Pharmacology and Toxicology, College of Pharmacy, The University of Arizona, Tucson, AZ 85721, USA; (D.H.L.); (S.M.)
| |
Collapse
|
24
|
Gambirasi M, Safa A, Vruzhaj I, Giacomin A, Sartor F, Toffoli G. Oral Administration of Cancer Vaccines: Challenges and Future Perspectives. Vaccines (Basel) 2023; 12:26. [PMID: 38250839 PMCID: PMC10821404 DOI: 10.3390/vaccines12010026] [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: 11/23/2023] [Revised: 12/19/2023] [Accepted: 12/22/2023] [Indexed: 01/23/2024] Open
Abstract
Cancer vaccines, a burgeoning strategy in cancer treatment, are exploring innovative administration routes to enhance patient and medical staff experiences, as well as immunological outcomes. Among these, oral administration has surfaced as a particularly noteworthy approach, which is attributed to its capacity to ignite both humoral and cellular immune responses at systemic and mucosal tiers, thereby potentially bolstering vaccine efficacy comprehensively and durably. Notwithstanding this, the deployment of vaccines through the oral route in a clinical context is impeded by multifaceted challenges, predominantly stemming from the intricacy of orchestrating effective oral immunogenicity and necessitating strategic navigation through gastrointestinal barriers. Based on the immunogenicity of the gastrointestinal tract, this review critically analyses the challenges and recent advances and provides insights into the future development of oral cancer vaccines.
Collapse
Affiliation(s)
- Marta Gambirasi
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS National Cancer Institute, 33081 Aviano, Italy; (M.G.); (I.V.); (F.S.)
- Department of Life Sciences, University of Trieste, 34127 Trieste, Italy;
| | - Amin Safa
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS National Cancer Institute, 33081 Aviano, Italy; (M.G.); (I.V.); (F.S.)
- Doctoral School in Pharmacological Sciences, University of Padua, 35131 Padova, Italy
- Department of Immunology, School of Medicine, Zabol University of Medical Sciences, Zabol 98616-15881, Iran
| | - Idris Vruzhaj
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS National Cancer Institute, 33081 Aviano, Italy; (M.G.); (I.V.); (F.S.)
- Doctoral School in Pharmacological Sciences, University of Padua, 35131 Padova, Italy
| | - Aurora Giacomin
- Department of Life Sciences, University of Trieste, 34127 Trieste, Italy;
| | - Franca Sartor
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS National Cancer Institute, 33081 Aviano, Italy; (M.G.); (I.V.); (F.S.)
| | - Giuseppe Toffoli
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS National Cancer Institute, 33081 Aviano, Italy; (M.G.); (I.V.); (F.S.)
| |
Collapse
|
25
|
Hargrave A, Mustafa AS, Hanif A, Tunio JH, Hanif SNM. Recent Advances in Cancer Immunotherapy with a Focus on FDA-Approved Vaccines and Neoantigen-Based Vaccines. Vaccines (Basel) 2023; 11:1633. [PMID: 38005965 PMCID: PMC10675687 DOI: 10.3390/vaccines11111633] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 09/30/2023] [Accepted: 10/10/2023] [Indexed: 11/26/2023] Open
Abstract
Cancer immunotherapies refer to the concept of retraining the immune system to target malignant cells. Multiple immunotherapeutic options exist including immune modulating antibodies, immune stimulating cytokines, chimeric antigen receptor T cell therapy, and vaccines. Overall, this field has advanced rapidly as knowledge of the tumor microenvironment, immunological pathways, and biotechnology expands. Specifically, advancements in neoantigen identification, characterization, and formulation into a vaccine show promise. This review is focused on previously United States Food and Drug Administration-approved cancer therapeutic vaccines and neoantigen-based vaccine developments along with the associated relevant clinical trials.
Collapse
Affiliation(s)
- Anna Hargrave
- Department of Biomedical Sciences, University of Pikeville, Pikeville, KY 41501, USA;
| | - Abu Salim Mustafa
- Department of Microbiology, Kuwait University, Kuwait City 12037, Kuwait;
| | - Asma Hanif
- Department of Restorative Sciences, Kuwait University, Kuwait City 12037, Kuwait;
| | - Javed H. Tunio
- Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA;
| | | |
Collapse
|