1
|
Mi Y, Jiang P, Luan J, Feng L, Zhang D, Gao X. Peptide‑based therapeutic strategies for glioma: Current state and prospects. Peptides 2025; 185:171354. [PMID: 39922284 DOI: 10.1016/j.peptides.2025.171354] [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: 11/23/2024] [Revised: 01/21/2025] [Accepted: 02/03/2025] [Indexed: 02/10/2025]
Abstract
Glioma is a prevalent form of primary malignant central nervous system tumor, characterized by its cellular invasiveness, rapid growth, and the presence of the blood-brain barrier (BBB)/blood-brain tumor barrier (BBTB). Current therapeutic approaches, such as chemotherapy and radiotherapy, have shown limited efficacy in achieving significant antitumor effects. Therefore, there is an urgent demand for new treatments. Therapeutic peptides represent an innovative class of pharmaceutical agents with lower immunogenicity and toxicity. They are easily modifiable via chemical means and possess deep tissue penetration capabilities which reduce side effects and drug resistance. These unique pharmacokinetic characteristics make peptides a rapidly growing class of new therapeutics that have demonstrated significant progress in glioma treatment. This review outlines the efforts and accomplishments in peptide-based therapeutic strategies for glioma. These therapeutic peptides can be classified into four types based on their anti-tumor function: tumor-homing peptides, inhibitor/antagonist peptides targeting cell surface receptors, interference peptides, and peptide vaccines. Furthermore, we briefly summarize the results from clinical trials of therapeutic peptides in glioma, which shows that peptide-based therapeutic strategies exhibit great potential as multifunctional players in glioma therapy.
Collapse
Affiliation(s)
- Yajing Mi
- Institute of Basic Medical Sciences, School of Basic Medical Science, Xi'an Medical University, Xi'an, China; Shaanxi Key Laboratory of Brain Disorders, School of Basic Medical Science, Xi'an Medical University, Xi'an, China
| | - Pengtao Jiang
- Institute of Basic Medical Sciences, School of Basic Medical Science, Xi'an Medical University, Xi'an, China
| | - Jing Luan
- Institute of Basic and Translational Medicine, Xi'an Medical University, Xi'an, Shaanxi, China
| | - Lin Feng
- Institute of Basic Medical Sciences, School of Basic Medical Science, Xi'an Medical University, Xi'an, China
| | - Dian Zhang
- Institute of Basic Medical Sciences, School of Basic Medical Science, Xi'an Medical University, Xi'an, China
| | - Xingchun Gao
- Institute of Basic Medical Sciences, School of Basic Medical Science, Xi'an Medical University, Xi'an, China; Shaanxi Key Laboratory of Brain Disorders, School of Basic Medical Science, Xi'an Medical University, Xi'an, China.
| |
Collapse
|
2
|
Lu Y, Ma N, Cheng K, Liu G, Liang J, Xu C, Li D, Cao C, Gao X, Chen L, Wang X, Wang Y, Zhao X, Jiang K. An OMV-Based Nanovaccine as Antigen Presentation Signal Enhancer for Cancer Immunotherapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2413392. [PMID: 39811977 DOI: 10.1002/adma.202413392] [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: 09/06/2024] [Revised: 01/07/2025] [Indexed: 01/16/2025]
Abstract
Antigen-presenting cells (APCs) process tumor vaccines and present tumor antigens as the first signals to T cells to activate anti-tumor immunity, which process requires the assistance of co-stimulatory second signals on APCs. The immune checkpoint programmed death ligand 1 (PD-L1) not only mediates the immune escape of tumor cells but also acts as a co-inhibitory second signal on APCs. The serious dysfunction of second signals due to the high expression of PD-L1 on APCs in the tumor body results in the inefficiency of tumor vaccines. To overcome this challenge, a previously established Plug-and-Display tumor vaccine platform based on bacterial outer membrane vesicles (OMVs) is developed into an "Antigen Presentation Signal Enhancer" (APSE) by surface-modifying PD-L1 antibodies (αPD-L1). While delivering tumor antigens, APSE can activate the expression of co-stimulatory second signals in APCs due to the high immunogenicity of OMVs. More importantly, the surface-modified αPD-L1 binds to the co-inhibitory signals PD-L1, potentially restoring CD80 function and ensuring efficient co-stimulatory second signals and activation of anti-tumor immunity. The results reveal the importance of PD-L1 blockage in the initiation process of anti-tumor immunity, and the second signal modulation capability of APSE can expand the application potential of cancer vaccines to less immunogenic malignancies.
Collapse
Affiliation(s)
- Yichao Lu
- Pancreas Center, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, Jiangsu Province, 210029, China
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, No.11 Zhongguancun Beiyitiao, Beijing, 100190, China
- IGDB-NCNST Joint Research Center, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Nana Ma
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, No.11 Zhongguancun Beiyitiao, Beijing, 100190, China
- IGDB-NCNST Joint Research Center, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Keman Cheng
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, No.11 Zhongguancun Beiyitiao, Beijing, 100190, China
- IGDB-NCNST Joint Research Center, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Guangna Liu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, No.11 Zhongguancun Beiyitiao, Beijing, 100190, China
- IGDB-NCNST Joint Research Center, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Jie Liang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, No.11 Zhongguancun Beiyitiao, Beijing, 100190, China
- IGDB-NCNST Joint Research Center, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Chen Xu
- Department of Surgical Oncology and General Surgery, Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, The First Affiliated Hospital of China Medical University, 155 North Nanjing Street, Shenyang, 110001, China
| | - Danrui Li
- Pancreas Center, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, Jiangsu Province, 210029, China
| | - Cheng Cao
- Pancreas Center, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, Jiangsu Province, 210029, China
| | - Xiaoyu Gao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, No.11 Zhongguancun Beiyitiao, Beijing, 100190, China
- IGDB-NCNST Joint Research Center, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Liting Chen
- Department of Surgical Oncology and General Surgery, Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, The First Affiliated Hospital of China Medical University, 155 North Nanjing Street, Shenyang, 110001, China
| | - Xinwei Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, No.11 Zhongguancun Beiyitiao, Beijing, 100190, China
- IGDB-NCNST Joint Research Center, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Yazhou Wang
- Pancreas Center, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, Jiangsu Province, 210029, China
| | - Xiao Zhao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, No.11 Zhongguancun Beiyitiao, Beijing, 100190, China
- IGDB-NCNST Joint Research Center, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Kuirong Jiang
- Pancreas Center, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, Jiangsu Province, 210029, China
| |
Collapse
|
3
|
Ruzzi F, Riccardo F, Conti L, Tarone L, Semprini MS, Bolli E, Barutello G, Quaglino E, Lollini PL, Cavallo F. Cancer vaccines: Target antigens, vaccine platforms and preclinical models. Mol Aspects Med 2025; 101:101324. [PMID: 39631227 DOI: 10.1016/j.mam.2024.101324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Accepted: 11/27/2024] [Indexed: 12/07/2024]
Abstract
This review provides a comprehensive overview of the evolving landscape of cancer vaccines, highlighting their potential to revolutionize tumor prevention. Building on the success of vaccines against virus-related cancers, such as HPV- and HBV-associated cervical and liver cancers, the current challenge is to extend these achievements to the prevention of non-viral tumors and the treatment of preneoplastic or early neoplastic lesions. This review analyzes the critical aspects of preventive anti-cancer vaccination, focusing on the choice of target antigens, the development of effective vaccine platforms and technologies, and the use of various model systems for preclinical testing, from laboratory rodents to companion animals.
Collapse
Affiliation(s)
- Francesca Ruzzi
- Laboratory of Immunology and Biology of Metastasis, Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40126, Bologna, Italy
| | - Federica Riccardo
- Laboratory of OncoImmunology, Department of Molecular Biotechnology and Health Sciences (DMBSS), University of Torino, 10126, Torino, Italy
| | - Laura Conti
- Laboratory of OncoImmunology, Department of Molecular Biotechnology and Health Sciences (DMBSS), University of Torino, 10126, Torino, Italy
| | - Lidia Tarone
- Laboratory of OncoImmunology, Department of Molecular Biotechnology and Health Sciences (DMBSS), University of Torino, 10126, Torino, Italy
| | - Maria Sofia Semprini
- Laboratory of Immunology and Biology of Metastasis, Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40126, Bologna, Italy
| | - Elisabetta Bolli
- Laboratory of OncoImmunology, Department of Molecular Biotechnology and Health Sciences (DMBSS), University of Torino, 10126, Torino, Italy
| | - Giuseppina Barutello
- Laboratory of OncoImmunology, Department of Molecular Biotechnology and Health Sciences (DMBSS), University of Torino, 10126, Torino, Italy
| | - Elena Quaglino
- Laboratory of OncoImmunology, Department of Molecular Biotechnology and Health Sciences (DMBSS), University of Torino, 10126, Torino, Italy
| | - Pier-Luigi Lollini
- Laboratory of Immunology and Biology of Metastasis, Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40126, Bologna, Italy; IRCCS Azienda Ospedaliera Universitaria di Bologna, 40138, Bologna, Italy.
| | - Federica Cavallo
- Laboratory of OncoImmunology, Department of Molecular Biotechnology and Health Sciences (DMBSS), University of Torino, 10126, Torino, Italy.
| |
Collapse
|
4
|
Grant M, Ni Lee L, Chinnakannan S, Tong O, Kwok J, Cianci N, Tillman L, Saha A, Pereira Almeida V, Leung C. Unlocking cancer vaccine potential: What are the key factors? Hum Vaccin Immunother 2024; 20:2331486. [PMID: 38564321 PMCID: PMC11657071 DOI: 10.1080/21645515.2024.2331486] [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: 01/08/2024] [Revised: 03/05/2024] [Accepted: 03/13/2024] [Indexed: 04/04/2024] Open
Abstract
Cancer is a global health challenge, with changing demographics and lifestyle factors producing an increasing burden worldwide. Screening advancements are enabling earlier diagnoses, but current cancer immunotherapies only induce remission in a small proportion of patients and come at a high cost. Cancer vaccines may offer a solution to these challenges, but they have been mired by poor results in past decades. Greater understanding of tumor biology, coupled with the success of vaccine technologies during the COVID-19 pandemic, has reinvigorated cancer vaccine development. With the first signs of efficacy being reported, cancer vaccines may be beginning to fulfill their potential. Solid tumors, however, present different hurdles than infectious diseases. Combining insights from previous cancer vaccine clinical development and contemporary knowledge of tumor immunology, we ask: who are the 'right' patients, what are the 'right' targets, and which are the 'right' modalities to maximize the chances of cancer vaccine success?
Collapse
|
5
|
Xu C. CRISPR/Cas9-mediated knockout strategies for enhancing immunotherapy in breast cancer. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:8561-8601. [PMID: 38907847 DOI: 10.1007/s00210-024-03208-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Accepted: 05/31/2024] [Indexed: 06/24/2024]
Abstract
Breast cancer, a prevalent disease with significant mortality rates, often presents treatment challenges due to its complex genetic makeup. This review explores the potential of combining Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR)/CRISPR-associated protein 9 (Cas9) gene knockout strategies with immunotherapeutic approaches to enhance breast cancer treatment. The CRISPR/Cas9 system, renowned for its precision in inducing genetic alterations, can target and eliminate specific cancer cells, thereby minimizing off-target effects. Concurrently, immunotherapy, which leverages the immune system's power to combat cancer, has shown promise in treating breast cancer. By integrating these two strategies, we can potentially augment the effectiveness of immunotherapies by knocking out genes that enable cancer cells to evade the immune system. However, safety considerations, such as off-target effects and immune responses, necessitate careful evaluation. Current research endeavors aim to optimize these strategies and ascertain the most effective methods to stimulate the immune response. This review provides novel insights into the integration of CRISPR/Cas9-mediated knockout strategies and immunotherapy, a promising avenue that could revolutionize breast cancer treatment as our understanding of the immune system's interplay with cancer deepens.
Collapse
Affiliation(s)
- Chenchen Xu
- Department of Gynecology and Obstetrics, Changzhou Maternal and Child Health Care Hospital, Changzhou Medical Center, Nanjing Medical University, Changzhou, 213000, China.
| |
Collapse
|
6
|
Morales-Martínez M, Andón-García D, Patiño-Santiago KA, Parga-Ortega JM, Hernández-Hernández A, Aquino-Jarquin G, Patino-Lopez G. Identification of potential new T cell activation molecules: a Bioinformatic Approach. Sci Rep 2024; 14:22219. [PMID: 39333573 PMCID: PMC11436975 DOI: 10.1038/s41598-024-73003-9] [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: 04/18/2024] [Accepted: 09/12/2024] [Indexed: 09/29/2024] Open
Abstract
T-cell activation is central for the initiation of T cell mediated adaptive immune response and is the result of the close communication between the Antigen Presenting Cell (APC) and the T lymphocyte. Although T-cell activation is currently well understood, and many intracellular pathways are well characterized, nevertheless new players are constantly identified, and this complements the known protein interactome. In this work we aimed to identify new proteins involved in T cell activation. We reviewed and analyzed results of microarray gene expression datasets reported in the public database GEO-NCBI. Using data from GSE136625, GSE50971, GSE13887, GSE11989 and GSE902 we performed different comparisons using R and other bioinformatic tools including GEO2R and we report here upregulated genes that have no previous reports in immune related functions and with potential participation upon T-cell activation. Our results indicate that RND3, SYT10, IgSF6 and PIN1 are potential new T-cell activation molecules.
Collapse
Affiliation(s)
- Mario Morales-Martínez
- Immunology and Proteomics Laboratory, Children's Hospital of Mexico, Mexico City, 06720, Mexico
| | - David Andón-García
- Immunology and Proteomics Laboratory, Children's Hospital of Mexico, Mexico City, 06720, Mexico
| | | | | | | | - Guillermo Aquino-Jarquin
- RNA Biology and Genome Editing Section, Genomics, Genetics, and Bioinformatics Research Laboratory, 'Federico Gómez' Children's Hospital of Mexico, Mexico City, 06720, Mexico
| | - Genaro Patino-Lopez
- Immunology and Proteomics Laboratory, Children's Hospital of Mexico, Mexico City, 06720, Mexico.
| |
Collapse
|
7
|
Luo Y, Lu C, Huang Y, Liao W, Huang Y. Identification of colon adenocarcinoma necroptosis subtypes and tumor antigens for the development of mRNA vaccines. Heliyon 2024; 10:e32531. [PMID: 38952359 PMCID: PMC11215264 DOI: 10.1016/j.heliyon.2024.e32531] [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: 06/03/2024] [Accepted: 06/05/2024] [Indexed: 07/03/2024] Open
Abstract
Background Colon adenocarcinoma (COAD) is a serious public health issue due to high incidence and mortality rate. This study aimed to identify possible tumor antigens and necroptosis subtypes of COAD for the development of mRNA vaccines and the selection of appropriate patients for precision therapy. Methods Gene expression profiles and clinical information for COAD were obtained from The Cancer Genome Atlas and Gene Expression Omnibus, respectively. We comprehensively studied the alterations in necroptosis-related genes (NRGs) using cBioPortal, and screened the hub NRGs associated with the prognosis of patients with COAD using Gene Expression Profiling Interactive Analysis 2. Consensuses clustering analysis was performed to identify necroptosis subtypes. Weighted gene co-expression network analysis (WGCNA) was used to identify the co-expression modules of the NRGs. The necroptosis landscape of COAD was assessed using graph learning-based dimensionality reduction. Finally, a drug sensitivity analysis of the two necroptosis subtypes was performed. Findings Two tumor antigens, BLC-2-associated X protein (BAX) and interleukin 1 beta (IL1B) were identified based on their associations with prognosis of patients and antigen presenting cell infiltration. Two necroptosis subtypes (N1 and N2) were distinguished in patients with COAD, and they were characterized by their differential survival status and molecular expression levels of immune checkpoint proteins and immunogenetic cell death modulators. Furthermore, the necroptosis landscape of COAD indicated that individual patients had obvious heterogeneity. Co-expression modules were identified using WGCNA, and the hub NRGs were found to be involved in various immune processes. Drug sensitivity analysis indicated that there were significant differences in drug sensitivity between the N1 and N2 subtypes. Cell experiments suggested that both overexpression of BAX and IL1B promoted necroptosis of COAD cells and enhanced the cytotoxicity of CD8+ T cells. Interpretation BAX and IL1B are potential antigens for the development of anti-COAD mRNA vaccines, specifically for patients with the N2 subtype. Consequently, this study will guide the development of more effective immunotherapeutic approaches and the identification of appropriate patients.
Collapse
Affiliation(s)
- Yuqi Luo
- Department of Gastrointestinal and Hepatobiliary Surgery, Shenzhen Longhua District Central Hospital, No. 187, Guanlan Road, Longhua District, Shenzhen 518110, Guangdong Province, China
| | - Caijie Lu
- Department of Gastrointestinal and Hepatobiliary Surgery, Shenzhen Longhua District Central Hospital, No. 187, Guanlan Road, Longhua District, Shenzhen 518110, Guangdong Province, China
| | - Yiwen Huang
- Department of Emergency, Nansha Hospital, Guangzhou First People's Hospital, School of Medicine, Southern China University of Technology, Guangzhou, Guangdong, China
| | - Weihua Liao
- Department of Radiology, Guangzhou Nansha District Maternal and Child Health Hospital, No. 103, Haibang Road, Nansha District, Guangzhou 511457, Guangdong Province, China
| | - Yaoxing Huang
- Department of Gastrointestinal and Hepatobiliary Surgery, Shenzhen Longhua District Central Hospital, No. 187, Guanlan Road, Longhua District, Shenzhen 518110, Guangdong Province, China
- Department of Gastroenterology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| |
Collapse
|
8
|
Kelly AM, McCarthy KN, Claxton TJ, Carlile SR, O'Brien EC, Vozza EG, Mills KH, McLoughlin RM. IL-10 inhibition during immunization improves vaccine-induced protection against Staphylococcus aureus infection. JCI Insight 2024; 9:e178216. [PMID: 38973612 PMCID: PMC11383370 DOI: 10.1172/jci.insight.178216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 05/22/2024] [Indexed: 07/09/2024] Open
Abstract
Staphylococcus aureus is a major human pathogen. An effective anti-S. aureus vaccine remains elusive as the correlates of protection are ill-defined. Targeting specific T cell populations is an important strategy for improving anti-S. aureus vaccine efficacy. Potential bottlenecks that remain are S. aureus-induced immunosuppression and the impact this might have on vaccine-induced immunity. S. aureus induces IL-10, which impedes effector T cell responses, facilitating persistence during both colonization and infection. Thus, it was hypothesized that transient targeting of IL-10 might represent an innovative way to improve vaccine efficacy. In this study, IL-10 expression was elevated in the nares of persistent carriers of S. aureus, and this was associated with reduced systemic S. aureus-specific Th1 responses. This suggests that systemic responses are remodeled because of commensal exposure to S. aureus, which negatively implicates vaccine function. To provide proof of concept that targeting immunosuppressive responses during immunization may be a useful approach to improve vaccine efficacy, we immunized mice with T cell-activating vaccines in combination with IL-10-neutralizing antibodies. Blocking IL-10 during vaccination enhanced effector T cell responses and improved bacterial clearance during subsequent systemic and subcutaneous infection. Taken together, these results reveal a potentially novel strategy for improving anti-S. aureus vaccine efficacy.
Collapse
Affiliation(s)
| | - Karen N McCarthy
- Host-Pathogen Interactions Group and
- Immune Regulation Research Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | | | | | | | | | - Kingston Hg Mills
- Immune Regulation Research Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | | |
Collapse
|
9
|
van Dorst MMAR, Pyuza JJ, Nkurunungi G, Kullaya VI, Smits HH, Hogendoorn PCW, Wammes LJ, Everts B, Elliott AM, Jochems SP, Yazdanbakhsh M. Immunological factors linked to geographical variation in vaccine responses. Nat Rev Immunol 2024; 24:250-263. [PMID: 37770632 DOI: 10.1038/s41577-023-00941-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/24/2023] [Indexed: 09/30/2023]
Abstract
Vaccination is one of medicine's greatest achievements; however, its full potential is hampered by considerable variation in efficacy across populations and geographical regions. For example, attenuated malaria vaccines in high-income countries confer almost 100% protection, whereas in low-income regions these same vaccines achieve only 20-50% protection. This trend is also observed for other vaccines, such as bacillus Calmette-Guérin (BCG), rotavirus and yellow fever vaccines, in terms of either immunogenicity or efficacy. Multiple environmental factors affect vaccine responses, including pathogen exposure, microbiota composition and dietary nutrients. However, there has been variable success with interventions that target these individual factors, highlighting the need for a better understanding of their downstream immunological mechanisms to develop new ways of modulating vaccine responses. Here, we review the immunological factors that underlie geographical variation in vaccine responses. Through the identification of causal pathways that link environmental influences to vaccine responsiveness, it might become possible to devise modulatory compounds that can complement vaccines for better outcomes in regions where they are needed most.
Collapse
Affiliation(s)
- Marloes M A R van Dorst
- Department of Parasitology, Leiden University Center for Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | - Jeremia J Pyuza
- Department of Parasitology, Leiden University Center for Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
- Department of Pathology, Kilimanjaro Christian Medical Centre, Moshi, Tanzania
| | - Gyaviira Nkurunungi
- Immunomodulation and Vaccines Programme, Medical Research Council/Uganda Virus Research Institute and London School of Hygiene and Tropical Medicine Uganda Research Unit, Entebbe, Uganda
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, UK
| | - Vesla I Kullaya
- Kilimanjaro Clinical Research Institute, Kilimanjaro Christian Medical Centre, Moshi, Tanzania
| | - Hermelijn H Smits
- Department of Parasitology, Leiden University Center for Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | | | - Linda J Wammes
- Department of Medical Microbiology, Leiden University Center for Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | - Bart Everts
- Department of Parasitology, Leiden University Center for Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | - Alison M Elliott
- Immunomodulation and Vaccines Programme, Medical Research Council/Uganda Virus Research Institute and London School of Hygiene and Tropical Medicine Uganda Research Unit, Entebbe, Uganda
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, UK
| | - Simon P Jochems
- Department of Parasitology, Leiden University Center for Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | - Maria Yazdanbakhsh
- Department of Parasitology, Leiden University Center for Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands.
| |
Collapse
|
10
|
Shen K, Zhang J, Zhao Z, Ma H, Wang Y, Zheng W, Xu J, Li Y, Wang B, Zhang Z, Wu S, Hou L, Chen W. Microparticulated Polygonatum sibiricum polysaccharide shows potent vaccine adjuvant effect. Int J Pharm 2024; 652:123802. [PMID: 38218508 DOI: 10.1016/j.ijpharm.2024.123802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 12/26/2023] [Accepted: 01/09/2024] [Indexed: 01/15/2024]
Abstract
Adjuvants are necessary for protein vaccines and have been used for nearly 100 years. However, developing safe and effective adjuvants is still urgently needed. Polysaccharides isolated from traditional Chinese medicine are considered novel vaccine adjuvant sources. This study aimed to investigate the adjuvant activity and immune-enhancing mechanisms of the microparticulated Polygonatum sibiricum polysaccharide (MP-PSP) modified by calcium carbonate. PSP demonstrated adjuvant activity, and MP-PSP further showed a higher humoral response compared to PSP. Subsequently, MP-PSP was elucidated to improving the immunity by slowing the rate of antigen release and activating dendritic cells along with interleukin-6 secretion through toll-like receptor 4 signaling, followed by T follicular helper cell and B cell interactions. Moreover, MP-PSP had a good safety profile in vaccinated mice. Thus, MP-PSP may be a promising vaccine adjuvant and warrants further investigation.
Collapse
Affiliation(s)
- Kai Shen
- College of Pharmacy, Nanjing University of Chinese Medicine, 138 Xianlin Avenue, Nanjing 210023, China; Beijing Institute of Biotechnology, 20 Dongdajie Street, Beijing 100071, China; Department of Pharmacy, Affiliated Hospital of Nantong University, 20 West Temple Road, Nantong 226001, China
| | - Jinlong Zhang
- Beijing Institute of Biotechnology, 20 Dongdajie Street, Beijing 100071, China
| | - Zhenghao Zhao
- Beijing Institute of Biotechnology, 20 Dongdajie Street, Beijing 100071, China
| | - Hao Ma
- Beijing Institute of Pharmacology and Toxicology, 27 Taiping Road, Beijing 100850, China
| | - Yudong Wang
- Beijing Institute of Biotechnology, 20 Dongdajie Street, Beijing 100071, China
| | - Wanru Zheng
- Beijing Institute of Biotechnology, 20 Dongdajie Street, Beijing 100071, China
| | - Jinghan Xu
- Beijing Institute of Biotechnology, 20 Dongdajie Street, Beijing 100071, China
| | - Yao Li
- Beijing Institute of Biotechnology, 20 Dongdajie Street, Beijing 100071, China
| | - Busen Wang
- Beijing Institute of Biotechnology, 20 Dongdajie Street, Beijing 100071, China
| | - Zhe Zhang
- Beijing Institute of Biotechnology, 20 Dongdajie Street, Beijing 100071, China
| | - Shipo Wu
- Beijing Institute of Biotechnology, 20 Dongdajie Street, Beijing 100071, China
| | - Lihua Hou
- Beijing Institute of Biotechnology, 20 Dongdajie Street, Beijing 100071, China.
| | - Wei Chen
- Beijing Institute of Biotechnology, 20 Dongdajie Street, Beijing 100071, China.
| |
Collapse
|
11
|
Sacchi de Camargo Correia G, Zhao Y, Manochakian R, Lou Y. The role of immunotherapy sensitizers and novel immunotherapy modalities in the treatment of cancer. Front Oncol 2024; 14:1336546. [PMID: 38476371 PMCID: PMC10928615 DOI: 10.3389/fonc.2024.1336546] [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: 11/15/2023] [Accepted: 02/05/2024] [Indexed: 03/14/2024] Open
Abstract
The importance of the immune system in the response against cancer has always been a subject of intense investigation. The advent of immune checkpoint inhibitors has transformed the landscape of oncologic treatments, while expanding the understanding of this disease's pathophysiology. Consequently, many therapies are being investigated, with interventions directed at different steps and pathways of the immune response. Relevantly, immunotherapy sensitizers have arisen as approaches focused on the synergistic effects of immunotherapy combination, or the combination of immunotherapy and other treatment modalities, such as chemotherapy or radiation therapy. Concomitantly, novel immunotherapy modalities are also in development. Approaches focusing from the tumor intrinsic pathways to the tumor microenvironment and ex-vivo interventions, such as CAR-T cell therapies and tumor-infiltrating lymphocytes are important examples. Although many of those interventions were initially envisioned as standalone options, their combination has demonstrated promising results in early-phase in vitro studies and clinical trials. The possibility of coupling different immunotherapy modalities, as well as with other techniques, further strengthen the concept of sensitizers, allowing for deeper and more robust responses in cancer treatment. This review aims to present an overview of the concepts of these sensitizing mechanisms that are the basis for the synergistic effects of immunotherapy combination, or the combination of immunotherapy and a multitude of therapeutic strategies. Novel immunotherapy modalities are also presented, focusing on the potential of combining them with sensitizer interventions. Understanding the complexity underlying these principles may be the key for future breakthroughs and improved patient outcomes.
Collapse
Affiliation(s)
| | - Yujie Zhao
- Division of Hematology and Medical Oncology, Mayo Clinic, Jacksonville, FL, United States
| | - Rami Manochakian
- Division of Hematology and Medical Oncology, Mayo Clinic, Jacksonville, FL, United States
| | - Yanyan Lou
- Division of Hematology and Medical Oncology, Mayo Clinic, Jacksonville, FL, United States
| |
Collapse
|
12
|
Meulewaeter S, Zhang Y, Wadhwa A, Fox K, Lentacker I, Harder KW, Cullis PR, De Smedt SC, Cheng MHY, Verbeke R. Considerations on the Design of Lipid-based mRNA Vaccines Against Cancer. J Mol Biol 2024; 436:168385. [PMID: 38065276 DOI: 10.1016/j.jmb.2023.168385] [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: 09/15/2023] [Revised: 11/29/2023] [Accepted: 12/02/2023] [Indexed: 12/26/2023]
Abstract
Throughout the last decades, mRNA vaccines have been developed as a cancer immunotherapeutic and the technology recently gained momentum during the COVID-19 pandemic. Recent promising results obtained from clinical trials investigating lipid-based mRNA vaccines in cancer therapy further highlighted the potential of this therapy. Interestingly, while the technologies being used in authorized mRNA vaccines for the prevention of COVID-19 are relatively similar, mRNA vaccines in clinical development for cancer vaccination show marked differences in mRNA modification, lipid carrier, and administration route. In this review, we describe findings on how these factors can impact the potency of mRNA vaccines in cancer therapy and provide insights into the complex interplay between them. We discuss how lipid carrier composition can affect passive targeting to immune cells to improve the efficacy and safety of mRNA vaccines. Finally, we summarize strategies that are established or still being explored to improve the efficacy of mRNA cancer vaccines and include next-generation vaccines that are on the horizon in clinical development.
Collapse
Affiliation(s)
- Sofie Meulewaeter
- Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences, Ghent University, Ghent 9000, Belgium; Cancer Research Institute Ghent (CRIG), Ghent University Hospital, Ghent University, Ghent 9000, Belgium
| | - Yao Zhang
- School of Biomedical Engineering, University of British Columbia, Vancouver, British Columbia V6T 1Z4, Canada; Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, British Columbia V6T 1Z4, Canada
| | - Abishek Wadhwa
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, British Columbia V6T 1Z4, Canada
| | - Kevin Fox
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, British Columbia V6T 1Z4, Canada
| | - Ine Lentacker
- Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences, Ghent University, Ghent 9000, Belgium; Cancer Research Institute Ghent (CRIG), Ghent University Hospital, Ghent University, Ghent 9000, Belgium
| | - Kenneth W Harder
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, British Columbia V6T 1Z4, Canada
| | - Pieter R Cullis
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, British Columbia V6T 1Z4, Canada
| | - Stefaan C De Smedt
- Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences, Ghent University, Ghent 9000, Belgium; Cancer Research Institute Ghent (CRIG), Ghent University Hospital, Ghent University, Ghent 9000, Belgium
| | - Miffy H Y Cheng
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, British Columbia V6T 1Z4, Canada.
| | - Rein Verbeke
- Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences, Ghent University, Ghent 9000, Belgium; Cancer Research Institute Ghent (CRIG), Ghent University Hospital, Ghent University, Ghent 9000, Belgium.
| |
Collapse
|
13
|
Jiménez-Cortegana C, Poveda C, Cabrera G. Editorial: The regulatory immune system as a target to improve adjuvants and novel vaccines. Front Cell Infect Microbiol 2023; 13:1223689. [PMID: 37342243 PMCID: PMC10277800 DOI: 10.3389/fcimb.2023.1223689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 05/30/2023] [Indexed: 06/22/2023] Open
Affiliation(s)
- Carlos Jiménez-Cortegana
- Department of Medical Biochemistry, Molecular Biology and Immunology, Faculty of Medicine, University of Seville, Seville, Spain
| | - Cristina Poveda
- Department of Pediatrics, National School of Tropical Medicine, Baylor College of Medicine, Houston, TX, United States
| | - Gabriel Cabrera
- Laboratorio de Tecnología Inmunológica, Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe Capital, Argentina
| |
Collapse
|
14
|
Batista-Duharte A, Aliño SF. Editorial: Immune-related adverse Events (irAEs) of biopharmaceuticals. FRONTIERS IN ALLERGY 2023; 4:1214914. [PMID: 37292091 PMCID: PMC10246496 DOI: 10.3389/falgy.2023.1214914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Accepted: 05/05/2023] [Indexed: 06/10/2023] Open
Affiliation(s)
- Alexander Batista-Duharte
- Immunology and Allergy Group (GC01), Maimonides Biomedical Research Institute of Cordoba (IMIBIC), University of Cordoba, Reina Sofia University Hospital, Cordoba, Spain
| | - Salvador F. Aliño
- Farmacogenetics and Gene Therapy Group, Instituto de Investigación Sanitaria La Fe, Valencia, Spain
- Gene Therapy and Pharmacogenomics Group, Department of Pharmacology, Faculty of Medicine, University of Valencia, Valencia, Spain
| |
Collapse
|
15
|
Lokhov PG, Balashova EE, Trifonova OP, Maslov DL, Archakov AI. Cell Proteomic Footprinting: Advances in the Quality of Cellular and Cell-Derived Cancer Vaccines. Pharmaceutics 2023; 15:661. [PMID: 36839983 PMCID: PMC9963030 DOI: 10.3390/pharmaceutics15020661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 01/31/2023] [Accepted: 02/13/2023] [Indexed: 02/18/2023] Open
Abstract
In omics sciences, many compounds are measured simultaneously in a sample in a single run. Such analytical performance opens up prospects for improving cellular cancer vaccines and other cell-based immunotherapeutics. This article provides an overview of proteomics technology, known as cell proteomic footprinting. The molecular phenotype of cells is highly variable, and their antigenic profile is affected by many factors, including cell isolation from the tissue, cell cultivation conditions, and storage procedures. This makes the therapeutic properties of cells, including those used in vaccines, unpredictable. Cell proteomic footprinting makes it possible to obtain controlled cell products. Namely, this technology facilitates the cell authentication and quality control of cells regarding their molecular phenotype, which is directly connected with the antigenic properties of cell products. Protocols for cell proteomic footprinting with their crucial moments, footprint processing, and recommendations for the implementation of this technology are described in this paper. The provided footprints in this paper and program source code for their processing contribute to the fast implementation of this technology in the development and manufacturing of cell-based immunotherapeutics.
Collapse
Affiliation(s)
- Petr G. Lokhov
- Institute of Biomedical Chemistry, 10 Building 8, Pogodinskaya Street, 119121 Moscow, Russia
| | | | | | | | | |
Collapse
|
16
|
Nie Z, Zhu S, Wu L, Sun R, Shu J, He Y, Feng H. Progress on innate immune evasion and live attenuated vaccine of pseudorabies virus. Front Microbiol 2023; 14:1138016. [PMID: 36937252 PMCID: PMC10020201 DOI: 10.3389/fmicb.2023.1138016] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Accepted: 02/15/2023] [Indexed: 03/06/2023] Open
Abstract
Pseudorabies virus (PRV) is a highly infectious disease that can infect most mammals, with pigs as the only natural host, has caused considerable economic losses to the pig husbandry of the world. Innate immunity is the first defense line of the host against the attack of pathogens and is essential for the proper establishment of adaptive immunity. The host uses the innate immune response to against the invasion of PRV; however PRV makes use of various strategies to inhibit the innate immunity to promote the virus replication. Currently, live attenuated vaccine is used to prevent pig from infection with the PRV worldwide, such as Bartha K61. However, a growing number of data indicates that these vaccines do not provide complete protection against new PRV variants that have emerged since late 2011. Here we summarized the interactions between PRV and host innate immunity and the current status of live attenuated PRV vaccines to promote the development of novel and more effective PRV vaccines.
Collapse
Affiliation(s)
- Zhenyu Nie
- Department of Biopharmacy, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
- Shaoxing Academy of Biomedicine, Zhejiang Sci-Tech University, Shaoxing, China
| | - Shunfan Zhu
- Department of Biopharmacy, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
- Shaoxing Academy of Biomedicine, Zhejiang Sci-Tech University, Shaoxing, China
| | - Li Wu
- Department of Biology, College of Life Sciences, China Jiliang University, Hangzhou, China
| | - Ruolin Sun
- Department of Biopharmacy, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Jianhong Shu
- Department of Biopharmacy, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Yulong He
- Department of Biopharmacy, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Huapeng Feng
- Department of Biopharmacy, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
- *Correspondence: Huapeng Feng,
| |
Collapse
|
17
|
Tsiakos K, Gavrielatou N, Vathiotis IA, Chatzis L, Chatzis S, Poulakou G, Kotteas E, Syrigos NK. Programmed Cell Death Protein 1 Axis Inhibition in Viral Infections: Clinical Data and Therapeutic Opportunities. Vaccines (Basel) 2022; 10:vaccines10101673. [PMID: 36298538 PMCID: PMC9611078 DOI: 10.3390/vaccines10101673] [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: 08/31/2022] [Revised: 09/29/2022] [Accepted: 10/05/2022] [Indexed: 11/16/2022] Open
Abstract
A vital function of the immune system is the modulation of an evolving immune response. It is responsible for guarding against a wide variety of pathogens as well as the establishment of memory responses to some future hostile encounters. Simultaneously, it maintains self-tolerance and minimizes collateral tissue damage at sites of inflammation. In recent years, the regulation of T-cell responses to foreign or self-protein antigens and maintenance of balance between T-cell subsets have been linked to a distinct class of cell surface and extracellular components, the immune checkpoint molecules. The fact that both cancer and viral infections exploit similar, if not the same, immune checkpoint molecules to escape the host immune response highlights the need to study the impact of immune checkpoint blockade on viral infections. More importantly, the process through which immune checkpoint blockade completely changed the way we approach cancer could be the key to decipher the potential role of immunotherapy in the therapeutic algorithm of viral infections. This review focuses on the effect of programmed cell death protein 1/programmed death-ligand 1 blockade on the outcome of viral infections in cancer patients as well as the potential benefit from the incorporation of immune checkpoint inhibitors (ICIs) in treatment of viral infections.
Collapse
Affiliation(s)
- Konstantinos Tsiakos
- 3rd Department of Internal Medicine, Medical School, National and Kapodistrian University of Athens, 157 72 Athens, Greece
- Correspondence:
| | - Niki Gavrielatou
- Department of Pathology, School of Medicine, Yale University, New Haven, CT 06520, USA
| | - Ioannis A. Vathiotis
- 3rd Department of Internal Medicine, Medical School, National and Kapodistrian University of Athens, 157 72 Athens, Greece
| | - Loukas Chatzis
- Pathophysiology Department, Athens School of Medicine, National and Kapodistrian University of Athens, 157 72 Athens, Greece
| | - Stamatios Chatzis
- Department of Internal Medicine, Medical School, National and Kapodistrian University of Athens, “Hippokration” Hospital, 115 27 Athens, Greece
| | - Garyfallia Poulakou
- 3rd Department of Internal Medicine, Medical School, National and Kapodistrian University of Athens, 157 72 Athens, Greece
| | - Elias Kotteas
- 3rd Department of Internal Medicine, Medical School, National and Kapodistrian University of Athens, 157 72 Athens, Greece
| | - Nikolaos K. Syrigos
- 3rd Department of Internal Medicine, Medical School, National and Kapodistrian University of Athens, 157 72 Athens, Greece
- Dana-Farber Brigham Cancer Center, Boston, MA 02215, USA
- Harvard Medical School, Boston, MA 02215, USA
| |
Collapse
|