1
|
Paul S, Kaya M, Johnsson O, Grauers Wiktorin H, Törnell A, Arabpour M, Hellstrand K, Martner A. Targeting murine metastatic cancers with cholera toxin A1-adjuvanted peptide vaccines. Hum Vaccin Immunother 2025; 21:2455240. [PMID: 39848921 PMCID: PMC11760229 DOI: 10.1080/21645515.2025.2455240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 12/26/2024] [Accepted: 01/15/2025] [Indexed: 01/25/2025] Open
Abstract
The dissemination of tumor cells with ensuing metastasis is responsible for most cancer-related deaths. Cancer vaccines may, by inducing tumor-specific effector T cells, offer a strategy to eliminate metastasizing tumor cells. However, several obstacles remain in the development of effective cancer vaccines, including the identification of adjuvants that enhance the evolvement and efficacy of tumor-specific T cells. Cholera toxin-based adjuvants have shown efficacy in vaccines for infectious diseases, but their role in cancer vaccine therapies remains to be elucidated. Here, we explored the potential of cholera toxin A1 (CTA1)-based adjuvants to boost anti-tumor T cell responses and protect against metastasis. We report that an adjuvant where CTA1 was fused to a dimer from Staphylococcus aureus protein A (DD) enhanced immune responses against the tumor-associated antigens TRP2 and Twist1 in mice, providing protection against B16F1 melanoma and 4T1 breast cancer metastasis, respectively. Both mucosal (intranasal) and systemic (intraperitoneal) vaccine administration provided effective protection against intravenously injected tumor cells, with intranasal administration leading to superior induction of CD4+ T cells at metastatic sites. When comparing antigens admixed with CTA1-DD to those fused with a CTA1-based adjuvant, the fusion construct elicited the strongest immunogenicity. Nevertheless, by administrating a 20-fold higher antigen dose also the admix formulation provided efficient protection against metastasis.
Collapse
MESH Headings
- Animals
- Cholera Toxin/administration & dosage
- Cholera Toxin/immunology
- Cholera Toxin/genetics
- Cancer Vaccines/immunology
- Cancer Vaccines/administration & dosage
- Adjuvants, Immunologic/administration & dosage
- Female
- Vaccines, Subunit/immunology
- Vaccines, Subunit/administration & dosage
- Vaccines, Subunit/genetics
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Neoplasm Metastasis/prevention & control
- Adjuvants, Vaccine/administration & dosage
- Melanoma, Experimental
- CD4-Positive T-Lymphocytes/immunology
- Administration, Intranasal
- Antigens, Neoplasm/immunology
- Cell Line, Tumor
- Disease Models, Animal
- Protein Subunit Vaccines
Collapse
Affiliation(s)
- Sanchari Paul
- TIMM Laboratory, Sahlgrenska Center for Cancer Research, Department of Microbiology and Immunology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Mustafa Kaya
- TIMM Laboratory, Sahlgrenska Center for Cancer Research, Department of Microbiology and Immunology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Olivia Johnsson
- TIMM Laboratory, Sahlgrenska Center for Cancer Research, Department of Microbiology and Immunology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Hanna Grauers Wiktorin
- TIMM Laboratory, Sahlgrenska Center for Cancer Research, Department of Microbiology and Immunology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Immunology, Genetics and Pathology, Science of Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Andreas Törnell
- TIMM Laboratory, Sahlgrenska Center for Cancer Research, Department of Microbiology and Immunology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Mohammad Arabpour
- TIMM Laboratory, Sahlgrenska Center for Cancer Research, Department of Microbiology and Immunology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Kristoffer Hellstrand
- TIMM Laboratory, Sahlgrenska Center for Cancer Research, Department of Infectious Diseases, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Anna Martner
- TIMM Laboratory, Sahlgrenska Center for Cancer Research, Department of Microbiology and Immunology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
2
|
Marciniak M, Stachowicz-Suhs M, Wagner M. The role of innate immune cells in modulating vascular dynamics in skin malignancies. Biochim Biophys Acta Rev Cancer 2025; 1880:189331. [PMID: 40280501 DOI: 10.1016/j.bbcan.2025.189331] [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: 01/28/2025] [Revised: 04/17/2025] [Accepted: 04/21/2025] [Indexed: 04/29/2025]
Abstract
A developing tumor relies heavily on blood vessels to supply oxygen and nutrients. As a result, angiogenesis, the formation of new blood vessels, supports tumor growth and progression. Similarly, lymphangiogenesis, the formation of new lymphatic vessels, plays a critical role in metastatic dissemination by providing pathways for malignant cells to spread. The tumor microenvironment is crucial for establishing and maintaining these vascular networks, with innate immune cells playing a key regulatory role. Notably, immune cells are specifically enriched in barrier tissues, such as the skin, emphasizing their importance in skin malignancies. Therefore, understanding their role in regulating angiogenesis and lymphangiogenesis is essential for developing novel therapeutic strategies. This review article explores how innate immune cells influence tumor vasculature and highlights the therapeutic potential that may arise from this knowledge.
Collapse
Affiliation(s)
- Mateusz Marciniak
- Innate Immunity Research Group, Life Sciences and Biotechnology Center, Łukasiewicz Research Network - PORT Polish Center for Technology Development, Wrocław, Poland; Department of Biochemistry and Immunochemistry, Wrocław Medical University, Wrocław, Poland
| | - Martyna Stachowicz-Suhs
- Innate Immunity Research Group, Life Sciences and Biotechnology Center, Łukasiewicz Research Network - PORT Polish Center for Technology Development, Wrocław, Poland
| | - Marek Wagner
- Innate Immunity Research Group, Life Sciences and Biotechnology Center, Łukasiewicz Research Network - PORT Polish Center for Technology Development, Wrocław, Poland.
| |
Collapse
|
3
|
Amor NP, Guo K, Zhang S, Xia J, Yang Y, Lin A. Lipid Nanoparticle: Beyond Delivery Vehicle-Unveiling Its Immunological Adjuvant Potential. FASEB J 2025; 39:e70641. [PMID: 40372384 DOI: 10.1096/fj.202500622r] [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: 02/28/2025] [Revised: 04/17/2025] [Accepted: 05/07/2025] [Indexed: 05/16/2025]
Abstract
Lipid nanoparticles (LNPs) have extensively been used in drug delivery over the years, and the perspective of their significance has been well established. Latest findings have demonstrated the inherent adjuvant capacity of some specific lipid components, especially in stimulating immune compartments, which prompted the rational use of lipid-based delivery vehicles in drug R&D. In this concise review, we summarize current knowledge on the adjuvant properties of LNP, with a particular focus on the key components that mediate such effects. Specifically, we describe the vital role of ionizable lipids in triggering innate immune activation and inflammation and highlight the importance of these lipids in determining vaccine effectiveness or safety. Furthermore, the mechanisms by which LNP enhance the immune response are discussed in detail, shedding light on their potential applications in next-generation vaccine design and development. We also present compelling pre-clinical studies that serve as strong evidence for the adjuvant properties of LNP components in enhancing vaccine immunogenicity.
Collapse
Affiliation(s)
- Narh Philip Amor
- Innovation Center for Nucleic Acid Medicine, Institute for Innovative Drug Development and Life Sciences, Wuxi, China Pharmaceutical University, Nanjing, China
- Institute of Translational Medicine, China Pharmaceutical University, Nanjing, China
- Center for Infectious Medicine and Vaccine Research, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Kun Guo
- Innovation Center for Nucleic Acid Medicine, Institute for Innovative Drug Development and Life Sciences, Wuxi, China Pharmaceutical University, Nanjing, China
- Institute of Translational Medicine, China Pharmaceutical University, Nanjing, China
- Center for Infectious Medicine and Vaccine Research, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Shun Zhang
- Ningbo Institute of Life and Health Industry, University of Chinese Academy of Sciences, Ningbo, China
| | - Jun Xia
- Institute of Veterinary Medicine, Xinjiang Academy of Animal Sciences, Urumqi, China
| | - Yong Yang
- Institute of Translational Medicine, China Pharmaceutical University, Nanjing, China
- Center for Infectious Medicine and Vaccine Research, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Ang Lin
- Innovation Center for Nucleic Acid Medicine, Institute for Innovative Drug Development and Life Sciences, Wuxi, China Pharmaceutical University, Nanjing, China
- Institute of Translational Medicine, China Pharmaceutical University, Nanjing, China
- Center for Infectious Medicine and Vaccine Research, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
- Institute of Immunopharmaceutical Sciences, School of Pharmaceutical Sciences, Shandong University, Jinan, China
| |
Collapse
|
4
|
Zaidi N, Jaffee EM, Yarchoan M. Recent advances in therapeutic cancer vaccines. Nat Rev Cancer 2025:10.1038/s41568-025-00820-z. [PMID: 40379970 DOI: 10.1038/s41568-025-00820-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/04/2025] [Indexed: 05/19/2025]
Abstract
The success of cancer prevention vaccines targeting cancer-causing viruses has drastically reduced cancer mortality worldwide. However, the development of therapeutic cancer vaccines, which aim to elicit an immune response directly against cancer cells, has faced notable clinical setbacks. In this Review, we explore lessons learned from past cancer vaccine trials and how the field has progressed into an era of renewed promise. Previous vaccines primarily targeted tumour-associated antigens and were mainly tested as monotherapies in late-stage cancers. In contrast, contemporary vaccines focus on targeting tumour-specific antigens (neoantigens) and are showing initial evidence of clinical efficacy, particularly in early-stage cancers and precancers when combined with immune checkpoint inhibitors. Advances in tumour profiling and novel vaccine platforms have enhanced vaccine specificity and potency. We discuss recent clinical trials of therapeutic cancer vaccines and outline future directions for the field.
Collapse
Affiliation(s)
- Neeha Zaidi
- Johns Hopkins Convergence Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Johns Hopkins Bloomberg Kimmel Institute for Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, USA
| | - Elizabeth M Jaffee
- Johns Hopkins Convergence Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Johns Hopkins Bloomberg Kimmel Institute for Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, USA.
| | - Mark Yarchoan
- Johns Hopkins Convergence Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Johns Hopkins Bloomberg Kimmel Institute for Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
5
|
Tsutsumi C, Ohuchida K, Yamada Y, Shimada Y, Imamura M, Son K, Mochida Y, Katayama N, Iwamoto C, Torata N, Horioka K, Shindo K, Mizuuchi Y, Ikenaga N, Nakata K, Onishi H, Oda Y, Nakamura M. Claudin18.2-positive gastric cancer-specific changes in neoadjuvant chemotherapy-driven immunosuppressive tumor microenvironment. Br J Cancer 2025; 132:793-804. [PMID: 40128286 PMCID: PMC12041497 DOI: 10.1038/s41416-025-02981-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 02/14/2025] [Accepted: 03/10/2025] [Indexed: 03/26/2025] Open
Abstract
BACKGROUND Claudin 18 isoform 2 (CLDN18.2) is a potential therapeutic target in gastric cancer (GC). However, combining chemotherapy with anti-CLDN18.2 antibodies has shown limited efficacy in CLDN18.2-positive GC, and chemotherapy-induced changes in the tumor microenvironment (TME) remain unclear. METHODS This study analyzed 37 GC samples, including 11 CLDN18.2-positive cases, using single-cell RNA sequencing and multiplex immunofluorescence to assess chemotherapy-driven TME changes in CLDN18.2-positive GC. RESULTS In chemotherapy-treated CLDN18.2-positive GC, cytotoxic natural killer (NK) cells displayed antibody-dependent cytotoxicity (ADCC)-related genes at lower levels than in untreated CLDN18.2-positive GC, while regulatory T cells (Tregs) and tumor-associated macrophages (TAMs) showed TGFB1 expression at higher levels. Additionally, NK cells, Tregs, and TAMs were more abundant in chemotherapy-treated than untreated CLDN18.2-positive GC. These chemotherapy-induced changes were absent in CLDN18.2-negative GC. Cell-cell interaction analysis identified unique interactions in chemotherapy-treated CLDN18.2-positive GC, including CCL5-CCR5 signaling between cytotoxic NK cells (Sender) and effector Tregs (Receptor) and TGFB1-TGFBR signaling between effector Tregs (Sender) and TAMs (Receptor). Cytotoxic NK cells expressed CCL5 at higher levels, CCR5-positive Tregs were more prevalent, and TAMs exhibited higher TGF-β receptor signature scores in chemotherapy-treated than untreated CLDN18.2-positive GC. CONCLUSIONS Our findings indicate that chemotherapy can drive immunosuppressive TME modifications specific to CLDN18.2-positive GC.
Collapse
Affiliation(s)
- Chikanori Tsutsumi
- Department of Surgery and Oncology, Graduate School of Medical Sciences; Kyushu University, Fukuoka, Japan
| | - Kenoki Ohuchida
- Department of Surgery and Oncology, Graduate School of Medical Sciences; Kyushu University, Fukuoka, Japan.
- Department of Advanced Medical Initiatives, Graduate School of Medical Sciences; Kyushu University, Fukuoka, Japan.
| | - Yutaka Yamada
- Department of Anatomic Pathology, Graduate School of Medical Sciences; Kyushu University, Fukuoka, Japan
| | - Yuki Shimada
- Department of Anatomic Pathology, Graduate School of Medical Sciences; Kyushu University, Fukuoka, Japan
| | - Masaki Imamura
- Department of Surgery and Oncology, Graduate School of Medical Sciences; Kyushu University, Fukuoka, Japan
| | - Kiwa Son
- Department of Surgery and Oncology, Graduate School of Medical Sciences; Kyushu University, Fukuoka, Japan
| | - Yuki Mochida
- Department of Surgery and Oncology, Graduate School of Medical Sciences; Kyushu University, Fukuoka, Japan
| | - Naoki Katayama
- Department of Surgery and Oncology, Graduate School of Medical Sciences; Kyushu University, Fukuoka, Japan
| | - Chika Iwamoto
- Department of Surgery and Oncology, Graduate School of Medical Sciences; Kyushu University, Fukuoka, Japan
| | - Nobuhiro Torata
- Department of Surgery and Oncology, Graduate School of Medical Sciences; Kyushu University, Fukuoka, Japan
| | - Kohei Horioka
- Department of Surgery and Oncology, Graduate School of Medical Sciences; Kyushu University, Fukuoka, Japan
| | - Koji Shindo
- Department of Surgery and Oncology, Graduate School of Medical Sciences; Kyushu University, Fukuoka, Japan
| | - Yusuke Mizuuchi
- Department of Surgery and Oncology, Graduate School of Medical Sciences; Kyushu University, Fukuoka, Japan
| | - Naoki Ikenaga
- Department of Surgery and Oncology, Graduate School of Medical Sciences; Kyushu University, Fukuoka, Japan
| | - Kohei Nakata
- Department of Surgery and Oncology, Graduate School of Medical Sciences; Kyushu University, Fukuoka, Japan
| | - Hideya Onishi
- Pancreatobiliary Surgery / Kidney & Pancreas Transplantation, Kyushu University Hospital, Fukuoka, Japan
| | - Yoshinao Oda
- Department of Anatomic Pathology, Graduate School of Medical Sciences; Kyushu University, Fukuoka, Japan
| | - Masafumi Nakamura
- Department of Surgery and Oncology, Graduate School of Medical Sciences; Kyushu University, Fukuoka, Japan
| |
Collapse
|
6
|
Ma L, Liu Y, Sun J, Yang X, He Y, Zhang T, Zhao J, Lu Z, Yan X, Qie X. The synthesis of nitric oxide regulated by JNK pathway in the pea aphid to defend against bacterial infection. INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2025; 180:104315. [PMID: 40274239 DOI: 10.1016/j.ibmb.2025.104315] [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: 01/24/2025] [Revised: 04/13/2025] [Accepted: 04/20/2025] [Indexed: 04/26/2025]
Abstract
Compared to other insects, the pea aphid Acyrthosiphon pisum exhibits limited immune responses, particularly due to the absence of many immune genes, including those encoding antimicrobial peptides and key components of the IMD pathway. Prior studies proved that the conserved signaling, Jun N-terminal kinase (JNK) pathway, plays a critical role in the immune system of the pea aphid, and nitric oxide synthase (NOS) is required for the pea aphid's defense against infections. Herein, using in vitro biochemical assays and in vivo bioassays, we demonstrated that the JNK pathway directly regulates the expression of NOS and that the JNK pathway-NOS-NO signal axis is efficient in defending against bacterial infections. The Toll pathway is instrumental for combating bacterial infections, and NO can activate the Toll pathway. The Toll pathway induced by NO regulates the expressions of ROS metabolism, lysosome, and phagocytosis-related genes. NO was identified as a crucial signaling molecule that facilitates communication between the JNK and Toll pathways.
Collapse
Affiliation(s)
- Li Ma
- College of Plant Protection, Shanxi Agricultural University, Taigu, Shanxi, 030801, China
| | - Yaya Liu
- College of Plant Protection, Shanxi Agricultural University, Taigu, Shanxi, 030801, China
| | - Jing Sun
- College of Plant Protection, Shanxi Agricultural University, Taigu, Shanxi, 030801, China
| | - Xiaorong Yang
- College of Plant Protection, Shanxi Agricultural University, Taigu, Shanxi, 030801, China
| | - Yingying He
- Research Institute of Applied Biology, Shanxi University, Taiyuan, Shanxi, 030006, China
| | - Tingting Zhang
- Research Institute of Applied Biology, Shanxi University, Taiyuan, Shanxi, 030006, China
| | - Jingyu Zhao
- College of Plant Protection, Shanxi Agricultural University, Taigu, Shanxi, 030801, China
| | - Zhiqiang Lu
- Department of Entomology, College of Plant Protection, Northwest A&F University, Yangling, Shaanxi, 712100, China.
| | - Xizhong Yan
- College of Plant Protection, Shanxi Agricultural University, Taigu, Shanxi, 030801, China.
| | - Xingtao Qie
- College of Plant Protection, Shanxi Agricultural University, Taigu, Shanxi, 030801, China.
| |
Collapse
|
7
|
Tsutsumi C, Ohuchida K, Imamura M, Tan B, Shimada Y, Son K, Kosai T, Katayama N, Mochida Y, Hayashida S, Iwamoto C, Torata N, Horioka K, Shindo K, Mizuuchi Y, Ikenaga N, Nakata K, Oda Y, Nakamura M. Prognostic nutrition index reveals LAG3 in cytotoxic CD8+ T cells and MHC class II in gastric cancer cells. Cancer Immunol Immunother 2025; 74:176. [PMID: 40252096 PMCID: PMC12009253 DOI: 10.1007/s00262-025-04037-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Accepted: 03/25/2025] [Indexed: 04/21/2025]
Abstract
BACKGROUND The prognostic nutrition index (PNI) has recently been highlighted as a predictor of immune checkpoint (IC) inhibitor efficacy in gastric cancer (GC). Although LAG3, an IC molecule, has gained considerable attention, its association with PNI remains unexplored. MATERIALS AND METHODS We retrospectively analyzed clinical data from 796 GC patients who underwent radical gastrectomy to identify which previously reported nutritional index had the greatest impact on prognosis. Single-cell RNA sequencing was performed on 38 GC tissues, and multiplex immunofluorescence staining was conducted on 59 GC tissues to evaluate the relationship between nutritional indices and IC molecule expression in cytotoxic CD8-positive T cells. RESULTS A low preoperative PNI was identified as the strongest predictor of poor prognosis among the nutritional indices in GC patients. The expression of not only PDCD1 (encoding PD1) but also LAG3 in cytotoxic CD8-positive T cells was significantly higher in GC with low PNI compared to those with high PNI. Among cytotoxic CD8-positive T cells, the proportion of LAG3-positive cells was greater than that of PDCD1-positive cells, particularly in GC with low PNI, and most LAG3-positive cells did not co-express PDCD1. Additionally, the expression of MHC class II, a ligand for LAG3, was higher in GC cells with high levels of epithelial-mesenchymal transition-related molecules in GC with low PNI compared to those with high PNI. CONCLUSIONS PNI can reflect LAG3 expression in cytotoxic CD8-positive T cells and MHC class II expression in GC cells.
Collapse
Affiliation(s)
- Chikanori Tsutsumi
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Kenoki Ohuchida
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan.
- Department of Advanced Medical Initiatives, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.
| | - Masaki Imamura
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Bryan Tan
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Yuki Shimada
- Department of Anatomic Pathology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kiwa Son
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Takaaki Kosai
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Naoki Katayama
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Yuki Mochida
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Sayuri Hayashida
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Chika Iwamoto
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Nobuhiro Torata
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Kohei Horioka
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Koji Shindo
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Yusuke Mizuuchi
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Naoki Ikenaga
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Kohei Nakata
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Yoshinao Oda
- Department of Anatomic Pathology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Masafumi Nakamura
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| |
Collapse
|
8
|
Moon CY, Belabed M, Park MD, Mattiuz R, Puleston D, Merad M. Dendritic cell maturation in cancer. Nat Rev Cancer 2025; 25:225-248. [PMID: 39920276 PMCID: PMC11954679 DOI: 10.1038/s41568-024-00787-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/12/2024] [Indexed: 02/09/2025]
Abstract
Dendritic cells (DCs) are specialized antigen-presenting cells that are present at low abundance in the circulation and tissues; they serve as crucial immune sentinels by continually sampling their environment, migrating to secondary lymphoid organs and shaping adaptive immune responses through antigen presentation. Owing to their ability to orchestrate tolerogenic or immunogenic responses to a specific antigen, DCs have a pivotal role in antitumour immunity and the response to immune checkpoint blockade and other immunotherapeutic approaches. The multifaceted functions of DCs are acquired through a complex, multistage process called maturation. Although the role of inflammatory triggers in driving DC maturation was established decades ago, less is known about DC maturation in non-inflammatory contexts, such as during homeostasis and in cancer. The advent of single-cell technologies has enabled an unbiased, high-dimensional characterization of various DC states, including mature DCs. This approach has clarified the molecular programmes associated with DC maturation and also revealed how cancers exploit these pathways to subvert immune surveillance. In this Review, we discuss the mechanisms by which cancer disrupts DC maturation and highlight emerging therapeutic opportunities to modulate DC states. These insights could inform the development of DC-centric immunotherapies, expanding the arsenal of strategies to enhance antitumour immunity.
Collapse
Affiliation(s)
- Chang Yoon Moon
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Meriem Belabed
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Matthew D Park
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Raphaël Mattiuz
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Daniel Puleston
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Miriam Merad
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
9
|
Li Z, Wen X, Lu S, Zheng Y, Zhao P, Mu S, Wang X, Shi Y, Qu F, Chang H. Ice-pop making inspired photothermal ultra-swelling microneedles to facilitate loading and intradermal vaccination of tumor antigen. J Control Release 2025; 379:77-88. [PMID: 39756684 DOI: 10.1016/j.jconrel.2024.12.076] [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: 09/27/2024] [Revised: 12/10/2024] [Accepted: 12/29/2024] [Indexed: 01/07/2025]
Abstract
Cancer vaccines hold great promise in the fight against cancer. Here, we report an ice-pop making inspired photothermal ultra-swelling microneedle (PUSMN) patch for facilitating and enhancing cancer vaccination. The PUSMN patch consist of an array of microneedles made from photo-crosslinked methacrylated hyaluronic acid and polydopamine, a near-infrared photothermal conversion material, connected to a customized resin handle like an ice-pop stick. Using a fabrication process similar to ice-pop making, the PUSMNs exhibit a rapid swelling ratio of over 2000 %, enabling straightforward and efficient loading of tumor antigen with just a 1-min incubation in the antigen solution, followed by 15 min of drying. The handle not only ensures convenient application but also guarantees full embedding of the PUSMNs in the skin after penetration. Under near-infrared irradiation, PUSMNs efficiently generate local heat, further promoting the activation and maturation of dendritic cells. In vivo vaccination with the model antigen ovalbumin using PUSMNs combined with near-infrared irradiation elicits robust tumor antigen-specific cellular and humoral immune responses, ultimately resulting in delayed tumor growth. Given its ease of use, efficiency, and safety features, this biocompatible PUSMN patch could greatly improve cancer vaccination.
Collapse
Affiliation(s)
- Zhiming Li
- Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Xueyu Wen
- Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Shaojie Lu
- Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China; College of Materials Science and Engineering, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, China
| | - Yanting Zheng
- Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China; College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou 310014, Zhejiang, China
| | - Puxuan Zhao
- Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China; College of Materials Science and Engineering, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, China
| | - Sijia Mu
- Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China; Medical School, Faculty of Medicine, Tianjin University, Tianjin 300072, China
| | - Xin Wang
- Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China; College of Materials Science and Engineering, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, China
| | - Yanan Shi
- Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China; Medical School, Faculty of Medicine, Tianjin University, Tianjin 300072, China
| | - Fengli Qu
- Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Hao Chang
- Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China.
| |
Collapse
|
10
|
Cui X, Song Y, Han J, Yuan Z. The multifaceted role of SMAD4 in immune cell function. Biochem Biophys Rep 2025; 41:101902. [PMID: 39802394 PMCID: PMC11721226 DOI: 10.1016/j.bbrep.2024.101902] [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: 08/30/2024] [Revised: 11/25/2024] [Accepted: 12/14/2024] [Indexed: 01/16/2025] Open
Abstract
The Transforming Growth Factor-beta (TGF-β) signaling pathway, with SMAD4 as its central mediator, plays a pivotal role in regulating cellular functions, including growth, differentiation, apoptosis, and immune responses. While extensive research has elucidated SMAD4's role in tumorigenesis, its functions within immune cells remain underexplored. This review synthesizes current knowledge on SMAD4's diverse roles in various immune cells such as T cells, B cells, dendritic cells, and macrophages, highlighting its impact on immune homeostasis and pathogen response. Understanding SMAD4's role in immune cells is crucial, as its dysregulation can lead to autoimmune disorders, chronic inflammation, and immune deficiencies. The review emphasizes the significance of SMAD4 in immune regulation, proposing that deeper investigation could reveal novel therapeutic targets for immune-mediated conditions. Insights into SMAD4's involvement in processes like T cell differentiation, B cell class switch recombination, and macrophage polarization underscore its potential as a therapeutic target for a range of diseases, including autoimmune disorders and cancer.
Collapse
Affiliation(s)
- Xinmu Cui
- Changchun Medical College, 6177, Jilin Street, Changchun, 130031, China
| | - Yu Song
- Changchun Medical College, 6177, Jilin Street, Changchun, 130031, China
| | - Jianfeng Han
- Changchun Medical College, 6177, Jilin Street, Changchun, 130031, China
- Cellular Biomedicine Group Inc, Shanghai, 201203, China
| | - Zhaoxin Yuan
- Changchun Medical College, 6177, Jilin Street, Changchun, 130031, China
| |
Collapse
|
11
|
Zhao Y, Liu J, Peng C, Guo S, Wang B, Chen L, Wang Y, Tang H, Liu L, Pan Q, Li S, Wang J, Yang D, Du E. Cross-protection against homo and heterologous influenza viruses via intranasal administration of an HA chimeric multiepitope nanoparticle vaccine. J Nanobiotechnology 2025; 23:77. [PMID: 39905416 PMCID: PMC11792681 DOI: 10.1186/s12951-025-03122-6] [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: 08/10/2024] [Accepted: 01/13/2025] [Indexed: 02/06/2025] Open
Abstract
BACKGROUND Influenza A viruses (IAVs) cause seasonal influenza epidemics and pose significant threats to public health. However, seasonal influenza vaccines often elicit strain-specific immune responses and confer little protection against mismatched strains. There is an urgent need to develop universal influenza vaccines against emerging and potentially re-emerging influenza virus infections. Multiepitope vaccines combining multiple conserved epitopes can induce more robust and broader immune responses and provide a potential solution. RESULTS Here, we demonstrated that an HA chimeric multiepitope nanoparticle vaccine, delivered intranasally conferred broad protection against challenges with various influenza viruses in mice. The nanoparticle vaccine co-expresses the ectodomain of haemagglutinin (H), three repeated highly conserved ectodomains of matrix protein 2 (M), and the M-cell-targeting ligand Co4B (C) in a baculovirus-insect cell system. These elements (C, H and M) were presented on the surface of self-assembling ferritin (f) in tandem to generate a nanoparticle denoted as CHM-f. Intranasal vaccination with CHM-f nanoparticles elicited robust humoral and cellular immune responses, conferring complete protection against a variety of IAVs, including the A/PR8/34 H1N1 strain, the swine flu H3N2 strain, the avian flu H5N8 strain, and H9N2. When CHM-f nanoparticles adjuvanted with CpG IAMA-002, the weight loss protective effect, cellular immune responses and mucosal IgA responses were significantly augmented. Compared with controls, mice immunized with CHM-f nanoparticles with or without CpG IAMA-002 showed significant reductions in weight loss, lung viral titres and pathological changes. CONCLUSIONS These results suggest that CHM-f nanoparticle with or without CpG IAMA-002 is a promising candidate as a universal influenza vaccine.
Collapse
Affiliation(s)
- Yongqiang Zhao
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Jia Liu
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Chun Peng
- Chengdu NanoVAX Biotechnology Co., Ltd., Chengdu, Sichuan, 610219, China
| | - Shuangshuang Guo
- Yangling Carey Biotechnology Co., Ltd., Yangling, Shaanxi, 712100, China
| | - Bo Wang
- Yangling Carey Biotechnology Co., Ltd., Yangling, Shaanxi, 712100, China
| | - Longping Chen
- Yangling Carey Biotechnology Co., Ltd., Yangling, Shaanxi, 712100, China
| | - Yating Wang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Haiwen Tang
- Chengdu NanoVAX Biotechnology Co., Ltd., Chengdu, Sichuan, 610219, China
| | - Liming Liu
- Nanjing JSIAMA Biopharmaceuticals Ltd., Nanjing, Jiangsu, 210000, China
| | - Qi Pan
- Nanjing JSIAMA Biopharmaceuticals Ltd., Nanjing, Jiangsu, 210000, China
| | - Shiren Li
- Chengdu NanoVAX Biotechnology Co., Ltd., Chengdu, Sichuan, 610219, China
| | - Jingyu Wang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Dongni Yang
- Chengdu NanoVAX Biotechnology Co., Ltd., Chengdu, Sichuan, 610219, China.
| | - Enqi Du
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, 712100, China.
- Yangling Carey Biotechnology Co., Ltd., Yangling, Shaanxi, 712100, China.
| |
Collapse
|
12
|
Song Z, Jin L, Jiao L, Yu R, Liu H, Zhang S, Hu Y, Sun Y, Li E, Zhao G, Liu Z, Cai T. ALC-0315 Lipid-Based mRNA LNP Induces Stronger Cellular Immune Responses Postvaccination. Mol Pharm 2025; 22:859-870. [PMID: 39813729 DOI: 10.1021/acs.molpharmaceut.4c00995] [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: 01/18/2025]
Abstract
At the end of 2019, SARS-CoV-2 emerged and rapidly spread, having a profound negative impact on human health and socioeconomic conditions. In response to this unprecedented global health crisis, significant advancements were made in the mRNA vaccine technology. In this study, we have compared the difference between two SARS-CoV-2 receptor-binding domain (RBD) mRNA-Lipid nanoparticle (LNP) vaccines prepared from two different ionizable cationic lipids: ALC-0315 and MC3. Characterization of RBD mRNA-LNPs showed that both MC3-LNP and ALC-0315-LNP are highly uniform and stable. Furthermore, we assessed the humoral immune response in mice after immunization; our findings indicated that both vaccine formulations effectively enhanced the formation and differentiation of germinal center (GC). Notably, the mice immunized with the ALC-0315-LNP vaccine elicited higher levels of IgG and its subclasses and significantly enhanced the activation of dendritic cells and T cells in draining lymph nodes (dLNs) compared to those immunized with the MC3-LNP vaccine. Further analysis of the T cell phenotype after splenic restimulation showed that mice injected with both LNP mRNA vaccines had significantly increased activation of the splenic T cells and Th1-type cytokine production. In addition, our finding showed that both LNP mRNA vaccines significantly increased the proportions of follicular helper T cells (Tfh) and long-lasting plasma cells in the dLNs of mice on day 14 postimmunization compared to control. In conclusion, both ALC-0315 and MC3 exhibited good stability and immunogenicity as mRNA-LNP recipes, but the ALC-0315-based mRNA-LNP vaccine showed higher efficacy in humoral and cellular immune responses compared to MC3.
Collapse
Affiliation(s)
- Zuchen Song
- Ningbo No.2 Hospital, Ningbo, Zhejiang 315010, P. R. China
- Guoke Ningbo Life Science and Health Industry Research Institute, Ningbo, Zhejiang 315000, P. R. China
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu 210095, P. R. China
| | - Lan Jin
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu 210095, P. R. China
| | - Lina Jiao
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu 210095, P. R. China
| | - Ruihong Yu
- Ningbo No.2 Hospital, Ningbo, Zhejiang 315010, P. R. China
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu 210095, P. R. China
| | - Huina Liu
- Ningbo No.2 Hospital, Ningbo, Zhejiang 315010, P. R. China
- Guoke Ningbo Life Science and Health Industry Research Institute, Ningbo, Zhejiang 315000, P. R. China
| | - Shun Zhang
- Ningbo No.2 Hospital, Ningbo, Zhejiang 315010, P. R. China
- Guoke Ningbo Life Science and Health Industry Research Institute, Ningbo, Zhejiang 315000, P. R. China
| | - Yaoren Hu
- Ningbo No.2 Hospital, Ningbo, Zhejiang 315010, P. R. China
- Guoke Ningbo Life Science and Health Industry Research Institute, Ningbo, Zhejiang 315000, P. R. China
| | - Yuechao Sun
- Ningbo No.2 Hospital, Ningbo, Zhejiang 315010, P. R. China
- Guoke Ningbo Life Science and Health Industry Research Institute, Ningbo, Zhejiang 315000, P. R. China
| | - Entao Li
- Ningbo No.2 Hospital, Ningbo, Zhejiang 315010, P. R. China
- Guoke Ningbo Life Science and Health Industry Research Institute, Ningbo, Zhejiang 315000, P. R. China
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu 210095, P. R. China
| | - Guofang Zhao
- Ningbo No.2 Hospital, Ningbo, Zhejiang 315010, P. R. China
- Guoke Ningbo Life Science and Health Industry Research Institute, Ningbo, Zhejiang 315000, P. R. China
| | - Zhenguang Liu
- Ningbo No.2 Hospital, Ningbo, Zhejiang 315010, P. R. China
- Guoke Ningbo Life Science and Health Industry Research Institute, Ningbo, Zhejiang 315000, P. R. China
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu 210095, P. R. China
| | - Ting Cai
- Ningbo No.2 Hospital, Ningbo, Zhejiang 315010, P. R. China
- Guoke Ningbo Life Science and Health Industry Research Institute, Ningbo, Zhejiang 315000, P. R. China
| |
Collapse
|
13
|
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
|
14
|
Bosteels V, Janssens S. Striking a balance: new perspectives on homeostatic dendritic cell maturation. Nat Rev Immunol 2025; 25:125-140. [PMID: 39289483 DOI: 10.1038/s41577-024-01079-5] [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: 07/31/2024] [Indexed: 09/19/2024]
Abstract
Dendritic cells (DCs) are crucial gatekeepers of the balance between immunity and tolerance. They exist in two functional states, immature or mature, that refer to an information-sensing versus an information-transmitting state, respectively. Historically, the term DC maturation was used to describe the acquisition of immunostimulatory capacity by DCs following their triggering by pathogens or tissue damage signals. As such, immature DCs were proposed to mediate tolerance, whereas mature DCs were associated with the induction of protective T cell immunity. Later studies have challenged this view and unequivocally demonstrated that two distinct modes of DC maturation exist, homeostatic and immunogenic DC maturation, each with a distinct functional outcome. Therefore, the mere expression of maturation markers cannot be used to predict immunogenicity. How DCs become activated in homeostatic conditions and maintain tolerance remains an area of intense debate. Several recent studies have shed light on the signals driving the homeostatic maturation programme, especially in the conventional type 1 DC (cDC1) compartment. Here, we highlight our growing understanding of homeostatic DC maturation and the relevance of this process for immune tolerance.
Collapse
Affiliation(s)
- Victor Bosteels
- Laboratory for ER Stress and Inflammation, VIB Center for Inflammation Research, Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
| | - Sophie Janssens
- Laboratory for ER Stress and Inflammation, VIB Center for Inflammation Research, Ghent, Belgium.
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium.
| |
Collapse
|
15
|
Kantheti U, Forward TS, Lucas ED, Schafer JB, Tamburini PJ, Burchill MA, Tamburini BAJ. PD-L1-CD80 interactions are required for intracellular signaling necessary for dendritic cell migration. SCIENCE ADVANCES 2025; 11:eadt3044. [PMID: 39879305 PMCID: PMC11777207 DOI: 10.1126/sciadv.adt3044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Accepted: 12/26/2024] [Indexed: 01/31/2025]
Abstract
Programmed cell death protein 1 (PD-1) and programmed death ligand 1 (PD-L1) interactions are targets for immunotherapies aimed to reinvigorate T cell function. Recently, it was documented that PD-L1 regulates dendritic cell (DC) migration through intracellular signaling events. In this study, we find that both preclinical murine and clinically available human PD-L1 antibodies limit DC migration. We show that cis interactions between PD-L1 and CD80 are critical for promoting migration and define specific regions within these proteins necessary for migration. Furthermore, we demonstrate that αPD-L1 significantly impedes DC migration in a B16 melanoma tumor model. Last, we outline how blocking cis PD-L1:CD80 interactions or mutation of the intracellular domain of PD-L1, in an imiquimod-induced murine model of psoriasis, limits DC migration to the lymph node, decreases interleukin-17 production by CD4+ T cells in the lymph node, and reduces epidermal thickening. Therefore, PD-L1 and CD80 interactions are important regulators of DC migration to the draining lymph node.
Collapse
Affiliation(s)
- Uma Kantheti
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Colorado School of Medicine, Aurora, CO, USA
- Immunology Graduate Program, University of Colorado School of Medicine, Aurora, CO, USA
- Medical Scientist Training Program, University of Colorado School of Medicine, Aurora, CO, USA
| | - Tadg S. Forward
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Erin D. Lucas
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Colorado School of Medicine, Aurora, CO, USA
- Immunology Graduate Program, University of Colorado School of Medicine, Aurora, CO, USA
| | - Johnathon B. Schafer
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Pierce J. Tamburini
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Matthew A. Burchill
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Beth Ann Jirón Tamburini
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Colorado School of Medicine, Aurora, CO, USA
- Immunology Graduate Program, University of Colorado School of Medicine, Aurora, CO, USA
- Medical Scientist Training Program, University of Colorado School of Medicine, Aurora, CO, USA
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO, USA
| |
Collapse
|
16
|
Mathew D, Ohri N, Ngiow SF, Halmos B, Wumesh KC, Garg M, Levsky JM, Cheng H, Gucalp RA, Bodner WR, Kabarriti R, Gutierrez-Chavez C, Zakharia A, Kalnicki S, Keler T, Yellin M, Minn AJ, Maity A, Giles JR, Guha C, Wherry EJ. Revitalizing systemic immune responses in advanced NSCLC using FLT3L and SBRT. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.01.29.25321332. [PMID: 39974012 PMCID: PMC11838671 DOI: 10.1101/2025.01.29.25321332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Non-small cell lung cancer (NSCLC) is the leading cause of cancer mortality worldwide. For patients who develop progressive disease after treatment with chemotherapy and immunotherapy, treatment options are limited. Here, we report the results of a Phase II clinical trial where twenty-nine patients with advanced, previously treated NSCLC were treated with stereotactic body radiotherapy (SBRT) targeting a single site of disease along with CDX-301, a recombinant human Fms-like tyrosine kinase 3 ligand (FLT3L). The primary study endpoint was progression-free survival four months after study entry (PFS4), which was achieved for 14 patients (48%). Abscopal responses were observed on fludeoxyglucose-18 positron emission tomography (FDG-PET) in nine patients (31%). The median overall survival for all participants was 18 months, with a median overall survival for participants with abscopal response of 31 months. To identify underlying immune signatures of response, we developed a high dimensional flow cytometric approach that quantified 31 distinct cell subsets from peripheral blood, and performed multiplex proteomic analysis of 92 proteins from plasma. SBRT combined with CDX-301 significantly increased circulating myeloid cells, including monocytes, myeloid derived suppressor cells (MDSCs) and dendritic cells (DCs). Among DCs, FceR1-expressing DC2 and DC3 subsets changed most robustly upon treatment. These dynamic changes in DCs and MDSCs following initiation of SBRT and CDX-301 returned to baseline by 8 weeks. However, these changes were also associated with treatment-induced T cell activation with quantitatively robust activation of CD4 T cells. Abscopal responses were associated with a prolonged increase in DC1 cells, Th1-like CD4 T cells, circulating IL-12 and FLT3L. Integrated analysis of multiple types of immune parameters revealed a strong association between coordinated DC induction, CD4 T cell and CD8 T cell activation, and inflammatory cytokine activity in patients with abscopal responses, compared to a stark lack of immune coordination in patients without abscopal responses. These coordinated immune responses were sustained for over 4 weeks in responders, highlighting a potential therapeutic axis engaged in a systemic immune response against multiple lesions in NSCLC. Overall, these findings underscore the potential of combining in situ vaccination, using SBRT, with strategies to enhance the activation of innate immune cells, such as DCs through FLT3L, to potentiate robust anti-tumor T cell responses.
Collapse
|
17
|
Watanabe T, Hayashi M, Arai M, Matsushita S, Handa H, Kawano M. Interferon-γ production in response to the cytotoxic T lymphocyte epitope within an antigen incorporated in simian virus 40 virus-like particles. Heliyon 2025; 11:e41729. [PMID: 39897799 PMCID: PMC11786666 DOI: 10.1016/j.heliyon.2025.e41729] [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: 05/22/2024] [Revised: 12/05/2024] [Accepted: 01/04/2025] [Indexed: 02/04/2025] Open
Abstract
In a previous study, we showed that administration of antigen-incorporated virus-like particles (VLPs) derived from simian virus 40 (SV40) induces production of cytotoxic T lymphocytes (CTL), as well as antibodies against the incorporated antigen(s), without the need for an adjuvant; however, it remains unclear how immune cells recognize and respond to the SV40 capsid because SV40 VLPs did not upregulate expression of maturation markers on dendritic cells. In this study, administration of chicken ovalbumin (OVA) incorporated within SV40 VLPs induced interferon (IFN)-γ production in response to the OVA CTL epitope. IFN-γ production in response to the OVA CTL epitope was not inhibited in B cell-depleted mice, but it was inhibited in cluster of differentiation (CD)4+ T cell-depleted mice. Administration of SV40 VLPs upregulated expression of CD63/CD68/CD83/CD86/CD196, and induced secretion of C-C chemokine ligand (CCL)3 and CCL4, by B cells, as well as secretion of CCL4 by T cells and tumor necrosis factor-α by bone marrow-derived dendritic cells.
Collapse
Affiliation(s)
- Takeharu Watanabe
- Vaccine Sohyaku Group Modality Laboratories Sohyaku. Innovation Research Division Mitsubishi Tanabe Pharma Corporation, 1000 Kamoshida, Aoba-ku, Yokohama, 227-0033, Japan
| | - Masayuki Hayashi
- Vaccine Sohyaku Group Modality Laboratories Sohyaku. Innovation Research Division Mitsubishi Tanabe Pharma Corporation, 1000 Kamoshida, Aoba-ku, Yokohama, 227-0033, Japan
| | - Masaaki Arai
- Vaccine Sohyaku Group Modality Laboratories Sohyaku. Innovation Research Division Mitsubishi Tanabe Pharma Corporation, 1000 Kamoshida, Aoba-ku, Yokohama, 227-0033, Japan
| | - Sho Matsushita
- Department of Allergy and Immunology, Faculty of Medicine, Saitama Medical University, 38 Morohongo, Moroyama, Saitama, 350-0495, Japan
- Allergy Center, Saitama Medical University, 38 Morohongo, Moroyama, Saitama, 350-0495, Japan
| | - Hiroshi Handa
- Department of Molecular Pharmacology, Tokyo Medical University, Shinjuku, Tokyo, 160-8402, Japan
- Center for Future Medical Research, Institute of Medical Science, Tokyo Medical University, Shinjuku, Tokyo, 160-8402, Japan
| | - Masaaki Kawano
- Department of Allergy and Immunology, Faculty of Medicine, Saitama Medical University, 38 Morohongo, Moroyama, Saitama, 350-0495, Japan
- Allergy Center, Saitama Medical University, 38 Morohongo, Moroyama, Saitama, 350-0495, Japan
- Division of Analytical Science, Biomedical Research Center, Faculty of Medicine, Saitama Medical University, 38 Morohongo, Moroyama, Saitama, 350-0495, Japan
| |
Collapse
|
18
|
Yi K, Sun C, Yuan Y, Luo Z, Luo H, Xie Y. A new weapon: the application of tumor vaccines based on extracellular exosomal heat shock proteins in immunotherapy. Front Immunol 2025; 16:1510650. [PMID: 39911383 PMCID: PMC11794256 DOI: 10.3389/fimmu.2025.1510650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Accepted: 01/08/2025] [Indexed: 02/07/2025] Open
Abstract
Despite the significant advancements in cancer research, innovative approaches are still needed to reduce tumor incidence, progression, and dissemination, as well as for prolonging patient survival. Currently, the development of cancer vaccines is gaining attention as a novel preventative and therapeutic strategy. Although the concept of cancer vaccination is not new, a limited number of vaccines have received approval for tumor therapy. Heat shock protein (HSP)-based vaccination represents a promising strategy that harnesses specific tumor antigens to activate immune responses. Exosomes (Exs) are highly heterogeneous bilayer vesicles capable of transporting various types of molecules through extracellular space. Compared with conventional anticancer drugs, exosomes exhibit low toxicity and good biocompatibility, and they can stimulate the immune system either directly or indirectly. Ex-based vaccines may elicit an antitumor immune response that generates memory cells capable of recognizing cancer antigens, thereby inhibiting disease progression. This paper reviews the potential applications of HSPs and exosomes in the prevention and treatment of solid tumors. Finally, we discuss the advantages of the extracellular exosomal heat shock protein (HSP-Ex) vaccine and future research directions aimed at optimizing heat shock protein-based cancer immunotherapy strategies.
Collapse
Affiliation(s)
- Kexin Yi
- The Second Clinical Medical College, Nanchang University, Nanchang, China
- Huankui Academy, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Chengpeng Sun
- Department of General Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Yalin Yuan
- Huankui Academy, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Zhaowei Luo
- Department of General Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Hongliang Luo
- Department of General Surgery, Jiujiang Hospital of Traditional Chinese Medicine, Jiujiang, Jiangxi, China
| | - Yunhe Xie
- The Second Clinical Medical College, Nanchang University, Nanchang, China
| |
Collapse
|
19
|
Hillemanns P, Zikan M, Forget F, Denys HG, Baurain JF, Rob L, Woelber L, Blecharz P, Bidzinski M, Chovanec J, Marmé F, Link T, Dannecker C, Rosholm A, Berg KCG, Oliveri RS, Lindemann K. Safety and efficacy of the therapeutic DNA-based vaccine VB10.16 in combination with atezolizumab in persistent, recurrent or metastatic HPV16-positive cervical cancer: a multicenter, single-arm phase 2a study. J Immunother Cancer 2025; 13:e010827. [PMID: 39773564 PMCID: PMC11749841 DOI: 10.1136/jitc-2024-010827] [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: 10/14/2024] [Accepted: 12/23/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND Second-line treatment options for persistent, recurrent or metastatic (r/m) cervical cancer are limited. We investigated the safety, efficacy, and immunogenicity of the therapeutic DNA-based vaccine VB10.16 combined with the immune checkpoint inhibitor atezolizumab in patients with human papillomavirus (HPV)16-positive r/m cervical cancer. PATIENTS AND METHODS This multicenter, single-arm, phase 2a study (NCT04405349, registered 26 May 2020) enrolled adult patients with persistent, r/m HPV16-positive cervical cancer. Patients received 3 mg VB10.16 (every 3 weeks (Q3W) for 12 weeks, hereafter every 6 weeks) combined with 1,200 mg atezolizumab (Q3W) for 48 weeks in total with a 12-month follow-up. The primary endpoints were incidence and severity of adverse events (AEs) and objective response rate (ORR; Response Evaluation Criteria in Solid Tumor V.1.1). ORR was assessed in the efficacy population, being all response-evaluable patients who received any administration of VB10.16 and atezolizumab and had at least one post-baseline imaging assessment. RESULTS Between June 16, 2020, and January 25, 2022, 52 patients received at least one administration of study treatment. Of these, 47 patients had a minimum of one post-baseline tumor assessment. The median follow-up time for survival was 11.7 months. AEs related to VB10.16 were non-serious and mainly mild injection site reactions (9 of 52 patients). There were no signs of new toxicities other than what was already described with atezolizumab. ORR was 19.1% (95% CI 9.1% to 33.3%). Median duration of response was not reached (n.r.) (95% CI 2.2 to n.r.), median progression-free survival was 4.1 months (95% CI 2.1 to 6.2), and median overall survival was 21.3 months (95% CI 8.5 to n.r.). In programmed death-ligand 1 (PD-L1)-positive patients (n=24), ORR was 29.2% (95% CI 12.6 to 51.1). HPV16-specific T-cell responses were analyzed in 36 of 47 patients with an increase observed in 22/36 (61%). CONCLUSIONS The therapeutic DNA-based vaccine VB10.16 combined with atezolizumab was safe and well tolerated showing a promising clinically meaningful efficacy with durable responses in patients with persistent, r/m HPV16-positive cervical cancer, especially if PD-L1-positive.
Collapse
Affiliation(s)
- Peter Hillemanns
- Department of Gynecology and Obstetrics, Hannover Medical School, Hannover, Germany
| | - Michal Zikan
- Department of Obstetrics and Gynecology, Bulovka University Hospital Na Bulovce Budinova 67/2, Prague, Czech Republic
| | | | - Hannelore G Denys
- Medical Oncology, University Hospital Ghent, Gent, Flanders, Belgium
| | | | - Lukas Rob
- University Hospital Kralovske Vinohrady, Praha, Czech Republic
| | - Linn Woelber
- Department of Gynecology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Pawel Blecharz
- Gynecologic Oncology Department, Maria Sklodowska-Curie National Research Institute of Oncology in Warsaw Krakow Branch, Krakow, Poland
| | - Mariusz Bidzinski
- Gynecologic Oncology Department, Maria Sklodowska-Curie National Research Institute of Oncology, Warzawa, Poland
| | | | - Frederik Marmé
- Department of Gynecology and Obstetrics, University Hospital Mannheim, Mannheim, Germany
| | - Theresa Link
- Department of Gynecology and Obstetrics, Medical Faculty, Dresden, Germany
- Department of Gynecology and Obstetrics, Technische Universität Dresden, Dresden, Germany
| | - Christian Dannecker
- Gynecology and Obstetrics, Faculty of Medicine, University of Augsburg, Augsburg, Germany
| | | | | | | | - Kristina Lindemann
- Department of gynecological oncology, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
20
|
Lei X, Yang Y, Zheng J, Liang L, Cheng L, Dong Y, Qiu B, Bikker FJ, Forouzanfar T, Cheng B, Wu G, Yang B. The cyclization of human salivary Histatin 1 via click chemistry for skin wound healing. Eur J Pharm Sci 2025; 204:106978. [PMID: 39631632 DOI: 10.1016/j.ejps.2024.106978] [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: 09/09/2024] [Revised: 12/01/2024] [Accepted: 12/01/2024] [Indexed: 12/07/2024]
Abstract
Acute skin injuries can result in the breakdown of the skin barrier, heightening the risk of infections and complications. Histatin 1 (Hst1) promotes the adhesion, spreading, and migration of various skin-related cells, thus encouraging wound healing. However, Hst1 is extensively degraded upon exposure to wound exudates. Cyclized hst1 (Cyclic-hst1) has a much higher resistance to protease degradation than Hst1, thus increasing its stability and half-life. Herein, we synthesized Cyclic-hst1 via a click reaction and explored its efficacy in wound healing via cellular and animal experiments. Cyclic-hst1, at a 100-fold lower concentration than Hst1, effectively promoted acute skin wound healing. In addition, Cyclic-hst1 had a superior effect to Hst1 in terms of its anti-inflammatory, re-epithelialization, collagen deposition, and angiogenic effects, thus significantly promoting skin wound healing. Consequently, Cyclic-hst1 could represent a favorable treatment to manage acute skin wound healing, providing a promising experimental basis for clinical transformation and application.
Collapse
Affiliation(s)
- Xiaoxuan Lei
- Dermatology Hospital, Southern Medical University, Guangzhou, China
| | - Yu Yang
- Department of Burn and Plastic Surgery, General Hospital of Southern Theater Command, Guangzhou, China; Department of Plastic Surgery, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Judun Zheng
- Dermatology Hospital, Southern Medical University, Guangzhou, China
| | - Liwen Liang
- Dermatology Hospital, Southern Medical University, Guangzhou, China
| | - Liuhanghang Cheng
- Department of Burn and Plastic Surgery, General Hospital of Southern Theater Command, Guangzhou, China
| | - Yunqing Dong
- Department of Burn and Plastic Surgery, General Hospital of Southern Theater Command, Guangzhou, China
| | - Biying Qiu
- Dermatology Hospital, Southern Medical University, Guangzhou, China
| | - Floris J Bikker
- Department of Oral Biochemistry, Academic Center for Dentistry Amsterdam (ACTA), University of Amsterdam (UvA) and Vrije Universiteit Amsterdam (VU), Amsterdam, The Netherlands
| | - Tymour Forouzanfar
- Department of Oral and Maxillofacial Surgery/Pathology, Amsterdam UMC and Academic Center for Dentistry Amsterdam (ACTA), Vrije Universiteit Amsterdam, Amsterdam Movement Science, Amsterdam, The Netherlands
| | - Biao Cheng
- Dermatology Hospital, Southern Medical University, Guangzhou, China.
| | - Gang Wu
- Savaid Stomatology School, Hangzhou Medical college, Hangzhou, Zhejiang Province, PR China.
| | - Bin Yang
- Dermatology Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
21
|
Song M, Ivkov R, Korangath P. Dendritic cell activation by iron oxide nanoparticles depends on the extracellular environment. NANOSCALE ADVANCES 2024; 7:209-218. [PMID: 39569333 PMCID: PMC11575603 DOI: 10.1039/d4na00561a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 11/10/2024] [Indexed: 11/22/2024]
Abstract
Nanoparticles can exert immune modulating effects in a host depending on composition, mode of administration, and type of disease. Although the specific mechanisms of nanoparticle-induced immune responses remain unclear, their uptake by macrophages and other phagocytic innate immune cells is considered to be a key event. Our objective here was to ascertain if nanoparticle-mediated activation of dendritic cells (DCs) occurs in vitro or in vivo when exposed to hydroxyethyl starch-coated iron oxide nanoparticles. For the present studies, our choice of nanoparticles, animal model, and experimental design is motivated by our previously published observations that systemic exposure can induce antitumor adaptive immune responses in mouse models of metastatic breast cancer. Here, we began by assessing the potential toxicity of systemic exposure to commercially available starch-coated Bionized Nanoferrite® nanoparticles (BP) by measuring body weight, complete blood count, and enzyme parameters in healthy FVB/NJ mice after repeated BP dosing. We observed no evidence of toxicity at doses up to 25 mg Fe per mouse, five-fold higher than those used in subsequent in vivo experiments. We then measured the expression of surface maturation markers (CD86, MHC II) in DCs incubated with BP in vitro. Although DCs cultured with BP revealed high levels of nanoparticle uptake, neither JAWSII dendritic cells nor bone marrow derived dendritic cells (BMDCs) showed significant changes in marker expression to indicate stimulation of maturation and effector function. To assess whether BP interactions in vivo produced different effects, we analyzed CD80, CD86, and MHC II expression of DCs recovered from the livers, spleens, bone marrows, and lymph nodes of mice injected once with BP (5 mg Fe). Interestingly, only DCs in spleens and bone marrow cells responded to BP exposure. DCs recovered from other organs showed no evidence of increased activation. These findings highlight complex interactions between living systems and nanoparticles, and their potential to mediate context-specific and selective activation of innate immune cells. Our study also emphasizes that results obtained from in vitro experiments must be interpreted with caution, as they may not faithfully represent responses in living systems.
Collapse
Affiliation(s)
- Mason Song
- Department of Biomedical Engineering, Whiting School of Engineering, Johns Hopkins University Baltimore 21218 USA
| | - Robert Ivkov
- Department of Radiation Oncology and Molecular Radiation Sciences, School of Medicine, Johns Hopkins University 1550 Orleans Street, Cancer Research Building - II, Rm 416 Baltimore MD 21231 USA
- Department of Oncology, Sydney Kimmel Comprehensive Cancer Center, School of Medicine, Johns Hopkins University Baltimore MD 21231 USA
- Department of Mechanical Engineering, Whiting School of Engineering, Johns Hopkins University Baltimore 21218 USA
- Department of Materials Science and Engineering, Whiting School of Engineering, Johns Hopkins University Baltimore 21218 USA
| | - Preethi Korangath
- Department of Radiation Oncology and Molecular Radiation Sciences, School of Medicine, Johns Hopkins University 1550 Orleans Street, Cancer Research Building - II, Rm 416 Baltimore MD 21231 USA
- Department of Oncology, Sydney Kimmel Comprehensive Cancer Center, School of Medicine, Johns Hopkins University Baltimore MD 21231 USA
| |
Collapse
|
22
|
Velimirov B, Velimirov BA. Immune Responses Elicited by Outer Membrane Vesicles of Gram-Negative Bacteria: Important Players in Vaccine Development. Life (Basel) 2024; 14:1584. [PMID: 39768292 PMCID: PMC11678573 DOI: 10.3390/life14121584] [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: 10/11/2024] [Revised: 11/12/2024] [Accepted: 11/20/2024] [Indexed: 01/11/2025] Open
Abstract
The attractiveness of OMVs derived from Gram-negative bacteria lies in the fact that they have two biomembranes sandwiching a peptidoglycan layer. It is well known that the envelope of OMVs consists of the outer bacterial membrane [OM] and not of the inner one [IM] of the source bacterium. This implies that all outer membranous molecules found in the OM act as antigens. However, under specific conditions, some of the inner membrane proteins can be exported into the outer membrane layer and perform as antigens. A key information was that the used purification procedures for OMVs, the induction methods to increase the production of OMVs as well as the specific mutant strains obtained via genetic engineering affect the composition of potential antigens on the surface and in the lumen of the OMVs. The available literature allowed us to list the major antigens that could be defined on OMVs. The functions of the antigens within the source bacterium are discussed for a better understanding of the various available hypotheses on the biogenesis of vesicle formation. Also, the impacts of OMV antigens on the immune system using animal models are assessed. Furthermore, information on the pathways of OMVs entering the host cell is presented. An example of a bacterial infection that causes epidemic diseases, namely via Neisseria meningitidis, is used to demonstrate that OMVs derived from this pathogen elicit protective immune responses when administered as a vaccine. Furthermore, information on OMV vaccines under development is presented. The assembled knowledge allowed us to formulate a number of reasons why OMVs are attractive as vaccine platforms, as their undesirable side effects remain small, and to provide an outlook on the potential use of OMVs as a vaccine platform.
Collapse
Affiliation(s)
- Branko Velimirov
- Division of Microbiology and Molecular Biology, Medical Faculty, Private Sigmund Freud University, Freudplatz 3, 1020 Wien, Austria;
| | | |
Collapse
|
23
|
Ahuja S, Zaheer S. The evolution of cancer immunotherapy: a comprehensive review of its history and current perspectives. KOREAN JOURNAL OF CLINICAL ONCOLOGY 2024; 20:51-73. [PMID: 39778508 PMCID: PMC11717579 DOI: 10.14216/kjco.24009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 10/24/2024] [Accepted: 11/19/2024] [Indexed: 01/11/2025]
Abstract
Cancer immunotherapy uses the body's immune system to combat cancer, marking a significant advancement in treatment. This review traces its evolution from the late 19th century to its current status. It began with William Coley's pioneering work using bacterial toxins to stimulate the immune system against cancer cells, establishing the foundational concept of immunotherapy. In the mid-20th century, cytokine therapies like interferons and interleukins emerged, demonstrating that altering the immune response could reduce tumors and highlighting the complex interplay between cancer and the immune system. The discovery of immune checkpoints, regulatory pathways that prevent autoimmunity but are exploited by cancer cells to evade detection, was a pivotal development. Another major breakthrough is CAR-T cell therapy, which involves modifying a patient's T cells to target cancer-specific antigens. This personalized treatment has shown remarkable success in certain blood cancers. Additionally, cancer vaccines aim to trigger immune responses against tumor-specific or associated antigens, and while challenging, ongoing research is improving their efficacy. The historical progression of cancer immunotherapy, from Coley's toxins to modern innovations like checkpoint inhibitors and CAR-T cell therapy, underscores its transformative impact on cancer treatment. As research delves deeper into the immune system's complexities, immunotherapy is poised to become even more crucial in oncology, offering renewed hope to patients globally.
Collapse
Affiliation(s)
- Sana Ahuja
- Department of Pathology, Safdarjung Hospital, Vardhman Mahavir Medical College, New Delhi, India
| | - Sufian Zaheer
- Department of Pathology, Safdarjung Hospital, Vardhman Mahavir Medical College, New Delhi, India
| |
Collapse
|
24
|
Rajsiglova L, Babic M, Krausova K, Lukac P, Kalkusova K, Taborska P, Sojka L, Bartunkova J, Stakheev D, Vannucci L, Smrz D. Immunogenic properties of nickel-doped maghemite nanoparticles and the implication for cancer immunotherapy. J Immunotoxicol 2024; 21:2416988. [PMID: 39484726 DOI: 10.1080/1547691x.2024.2416988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Revised: 09/29/2024] [Accepted: 10/10/2024] [Indexed: 11/03/2024] Open
Abstract
Nanoparticles are commonly used in diagnostics and therapy. They are also increasingly being implemented in cancer immunotherapy because of their ability to deliver drugs and modulate the immune system. However, the effect of nanoparticles on immune cells involved in the anti-tumor immune response is not well understood. The study reported here showed that nickel-doped maghemite nanoparticles (FN NP) are differentially cytotoxic to cultured mouse and human cancer cell lines, causing their death without negatively impacting the subsequent anticancer immune response. It also found that FN NP induced cell death in the mouse colorectal cancer cell line CT26 and human prostate cancer cell line PC-3, but not in the human prostate cancer cell line LNCaP. The induced cancer cell death did not affect the phenotype and responsivity of the isolated mouse peritoneal macrophages, or ex vivo-generated mouse bone marrow-derived, or human monocyte-derived dendritic cells. Additionally, the induced cancer cell death did not prevent the ex vivo-generated mouse or human dendritic cells from stimulating lymphocytes and enriching cell cultures with cancer cell-reactive T-cells. In conclusion, this study shows that FN NP could be a valuable platform for targeting cancer cells without causing immunosuppressive effects on the subsequent anticancer immune response.
Collapse
Affiliation(s)
- Lenka Rajsiglova
- Laboratory of Immunotherapy, Institute of Microbiology of the Czech Academy of Sciences, v.v.i, Prague, Czech Republic
- Department of Cell Biology, Faculty of Science, Charles University, Prague, Czech Republic
| | - Michal Babic
- Institute of Macromolecular Chemistry, Czech Academy of Sciences, Prague, Czech Republic
| | - Katerina Krausova
- Laboratory of Immunotherapy, Institute of Microbiology of the Czech Academy of Sciences, v.v.i, Prague, Czech Republic
- Department of Immunology, Second Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czech Republic
| | - Pavol Lukac
- Laboratory of Immunotherapy, Institute of Microbiology of the Czech Academy of Sciences, v.v.i, Prague, Czech Republic
- Department of Cell Biology, Faculty of Science, Charles University, Prague, Czech Republic
| | - Katerina Kalkusova
- Department of Immunology, Second Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czech Republic
| | - Pavla Taborska
- Department of Immunology, Second Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czech Republic
| | - Ludek Sojka
- Department of Immunology, Second Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czech Republic
- Department of Technical Operations, SOTIO, a.s., Prague, Czech Republic
| | - Jirina Bartunkova
- Department of Immunology, Second Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czech Republic
| | - Dmitry Stakheev
- Laboratory of Immunotherapy, Institute of Microbiology of the Czech Academy of Sciences, v.v.i, Prague, Czech Republic
- Department of Immunology, Second Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czech Republic
| | - Luca Vannucci
- Laboratory of Immunotherapy, Institute of Microbiology of the Czech Academy of Sciences, v.v.i, Prague, Czech Republic
| | - Daniel Smrz
- Laboratory of Immunotherapy, Institute of Microbiology of the Czech Academy of Sciences, v.v.i, Prague, Czech Republic
- Department of Immunology, Second Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czech Republic
| |
Collapse
|
25
|
Huang L, Zhang H, Xia W, Yao N, Xu R, He Y, Yang Q. Structural characteristics, biological activities and market applications of Rehmannia Radix polysaccharides: A review. Int J Biol Macromol 2024; 282:136554. [PMID: 39423982 DOI: 10.1016/j.ijbiomac.2024.136554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 09/14/2024] [Accepted: 10/11/2024] [Indexed: 10/21/2024]
Abstract
Rehmannia Radix Polysaccharides (RRPs) are biopolymers that are isolated and purified from the roots of Rehmannia glutinosa Libosch, which have attracted considerable attention because of their biological activities, such as anti-inflammatory, antioxidant, immunomodulatory, anti-tumor, hypoglycaemic etc. In this manuscript, the composition and structural characteristics of RRPs are reviewed. Moreover, the research progress on the conformational relationships and biological activities of RRPs is systematically summarized. Additionally, this manuscript also analyzes 155 patents using RRPs as the main raw materials to explore the status quo and bottleneck for the development and utilization of RRPs. In summary, this review not only provides a theoretical basis for future research on RRPs but also provides clear guidance for their market applications and innovation.
Collapse
Affiliation(s)
- Lin Huang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, Sichuan, China
| | - Haibo Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, Sichuan, China
| | - Wenrui Xia
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
| | - Nairong Yao
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
| | - Runchun Xu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, Sichuan, China
| | - Yanan He
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, Sichuan, China.
| | - Qiyue Yang
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China.
| |
Collapse
|
26
|
Ma J, Wu Y, Wu S, Fang Z, Chen L, Jiang J, Zheng X. CX3CR1 +CD8 + T cells: Key players in antitumor immunity. Cancer Sci 2024; 115:3838-3845. [PMID: 39377122 PMCID: PMC11611776 DOI: 10.1111/cas.16359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 08/18/2024] [Accepted: 09/17/2024] [Indexed: 10/09/2024] Open
Abstract
CX3CR1 functions as the specific receptor for the chemokine CX3CL1, demonstrating expression across a broad spectrum of immune cells. This underscores its pivotal role in communication and response mechanisms within the immune system. Upon engagement with CX3CL1, CX3CR1 initiates a cascade of downstream signaling pathways that regulate various biological functions. In the context of tumor progression, the intricate and inhibitory nature of the tumor microenvironment presents a significant challenge to current clinical treatment techniques. This review aims to comprehensively explore the tumor-destructive potential shown by CX3CR1+CD8+ T cells. Simultaneously, it investigates the promising prospects of utilizing CX3CR1 in future tumor immunotherapies.
Collapse
Affiliation(s)
- Jiajin Ma
- Department of Tumor Biological TreatmentThe Third Affiliated Hospital of Soochow UniversityChangzhouChina
- Jiangsu Engineering Research Center for Tumor ImmunotherapyChangzhouChina
- Institute for Cell Therapy of Soochow UniversityChangzhouChina
| | - Yue Wu
- Department of Tumor Biological TreatmentThe Third Affiliated Hospital of Soochow UniversityChangzhouChina
- Jiangsu Engineering Research Center for Tumor ImmunotherapyChangzhouChina
- Institute for Cell Therapy of Soochow UniversityChangzhouChina
| | - Shaoxian Wu
- Department of Tumor Biological TreatmentThe Third Affiliated Hospital of Soochow UniversityChangzhouChina
- Jiangsu Engineering Research Center for Tumor ImmunotherapyChangzhouChina
- Institute for Cell Therapy of Soochow UniversityChangzhouChina
| | - Zhang Fang
- Department of Tumor Biological TreatmentThe Third Affiliated Hospital of Soochow UniversityChangzhouChina
- Jiangsu Engineering Research Center for Tumor ImmunotherapyChangzhouChina
- Institute for Cell Therapy of Soochow UniversityChangzhouChina
| | - Lujun Chen
- Department of Tumor Biological TreatmentThe Third Affiliated Hospital of Soochow UniversityChangzhouChina
- Jiangsu Engineering Research Center for Tumor ImmunotherapyChangzhouChina
- Institute for Cell Therapy of Soochow UniversityChangzhouChina
| | - Jingting Jiang
- Department of Tumor Biological TreatmentThe Third Affiliated Hospital of Soochow UniversityChangzhouChina
- Jiangsu Engineering Research Center for Tumor ImmunotherapyChangzhouChina
- Institute for Cell Therapy of Soochow UniversityChangzhouChina
| | - Xiao Zheng
- Department of Tumor Biological TreatmentThe Third Affiliated Hospital of Soochow UniversityChangzhouChina
- Jiangsu Engineering Research Center for Tumor ImmunotherapyChangzhouChina
- Institute for Cell Therapy of Soochow UniversityChangzhouChina
| |
Collapse
|
27
|
He J, Ding X, Zhao J, Zeng J, Zhou Y, Xiao W, Hua D, Liu M, Guo H, Zhang Y, Qiu M, Li J. A novel pan-epitope based nanovaccine self-assembled with CpG enhances immune responses against flavivirus. J Nanobiotechnology 2024; 22:738. [PMID: 39609873 PMCID: PMC11603839 DOI: 10.1186/s12951-024-03031-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 11/21/2024] [Indexed: 11/30/2024] Open
Abstract
BACKGROUND Flavivirus is a highly prevalent and outbreak-prone disease, affecting billions of individuals annually and posing substantial public health challenges. Vaccination is critical to reducing the global impact of flavivirus infections, making the development of a safe and effective vaccine a top priority. The self-assembled pan-epitope vaccine presents key advantages for improving immunogenicity and safety without relying on external vectors or adding immunomodulatory elements, both of which are essential for successful vaccine development. RESULTS In this study, the pan-epitope peptide TBT was combined with adjuvant CpG to form the TBT-CpG nanovaccine (TBT-CpG NaVs), which was found to be spherical, uniform in shape, and demonstrated strong serum stability. In vitro studies showed that the TBT-CpG NaVs were efficiently taken up and internalized by bone marrow-derived dendritic cells (BMDCs). Flow cytometry and transcriptomic analysis indicated that the antigens were effectively presented to antigen-presenting cells (APCs) via the MHC II pathway, which facilitated BMDCs maturation and promoted the release of pro-inflammatory cytokines IL-1β, TNF-α, and IL-6. In vivo studies confirmed that TBT-CpG NaVs enhanced antigen-specific IgG levels, significantly increased IFN-γ and IL-4 expression in spleen cells, and offered protective effects against Dengue virus (DENV) and Zika virus (ZIKV) infections. Safety evaluations revealed no hepatotoxicity and no significant organ damage in immunized mice. CONCLUSION The self-assembled candidate nanovaccine TBT-CpG NaVs effectively activates BMDCs and triggers a targeted immune response, providing antiviral effects against DENV and ZIKV. This vaccine demonstrates good immunogenicity and safety, establishing a promising foundation and a new strategy for the development of safe and effective vaccines.
Collapse
Affiliation(s)
- Jiuxiang He
- College of Basic Medicine, Army Medical University, Gaotanyan str. 30, Chongqing, 400038, China
| | - Xiaoyan Ding
- College of Basic Medicine, Army Medical University, Gaotanyan str. 30, Chongqing, 400038, China
- Department of Pediatrics, Dr. von Hauner Children's Hospital, Ludwig Maximilians University, Munich, Germany
| | - Jing Zhao
- College of Basic Medicine, Army Medical University, Gaotanyan str. 30, Chongqing, 400038, China
| | - Jie Zeng
- College of Basic Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Yuxin Zhou
- College of Basic Medicine, Army Medical University, Gaotanyan str. 30, Chongqing, 400038, China
| | - Wen Xiao
- College of Basic Medicine, Army Medical University, Gaotanyan str. 30, Chongqing, 400038, China
| | - Dong Hua
- College of Basic Medicine, Army Medical University, Gaotanyan str. 30, Chongqing, 400038, China
| | - Minchi Liu
- College of Basic Medicine, Army Medical University, Gaotanyan str. 30, Chongqing, 400038, China
| | - Hongxia Guo
- College of Basic Medicine, Army Medical University, Gaotanyan str. 30, Chongqing, 400038, China
| | - Yu Zhang
- College of Basic Medicine, Army Medical University, Gaotanyan str. 30, Chongqing, 400038, China
| | - Minyue Qiu
- College of Basic Medicine, Army Medical University, Gaotanyan str. 30, Chongqing, 400038, China
| | - Jintao Li
- College of Basic Medicine, Army Medical University, Gaotanyan str. 30, Chongqing, 400038, China.
| |
Collapse
|
28
|
Turner MB, Dalmasso C, Loria AS. The adipose tissue keeps the score: priming of the adrenal-adipose tissue axis by early life stress predisposes women to obesity and cardiometabolic risk. Front Endocrinol (Lausanne) 2024; 15:1481923. [PMID: 39493777 PMCID: PMC11527639 DOI: 10.3389/fendo.2024.1481923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Accepted: 09/26/2024] [Indexed: 11/05/2024] Open
Abstract
Adverse Childhood Experiences (ACEs) refer to early life stress events, including abuse, neglect, and other psychosocial childhood traumas that can have long-lasting effects on a wide range of physiological functions. ACEs provoke sex-specific effects, whereas women have been shown to display a strong positive correlation with obesity and cardiometabolic disease. Notably, rodent models of chronic behavioral stress during postnatal life recapitulate several effects of ACEs in a sex-specific fashion. In this review, we will discuss the potential mechanisms uncovered by models of early life stress that may explain the greater susceptibility of females to obesity and metabolic risk compared with their male counterparts. We highlight the early life stress-induced neuroendocrine shaping of the adrenal-adipose tissue axis as a primary event conferring sex-dependent heightened sensitivity to obesity.
Collapse
Affiliation(s)
| | | | - Analia S. Loria
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, United States
| |
Collapse
|
29
|
Chudnovskiy A, Castro TBR, Nakandakari-Higa S, Cui A, Lin CH, Sade-Feldman M, Phillips BK, Pae J, Mesin L, Bortolatto J, Schweitzer LD, Pasqual G, Lu LF, Hacohen N, Victora GD. Proximity-dependent labeling identifies dendritic cells that drive the tumor-specific CD4 + T cell response. Sci Immunol 2024; 9:eadq8843. [PMID: 39365874 DOI: 10.1126/sciimmunol.adq8843] [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: 06/04/2024] [Accepted: 08/26/2024] [Indexed: 10/06/2024]
Abstract
Dendritic cells (DCs) are uniquely capable of transporting tumor antigens to tumor-draining lymph nodes (tdLNs) and interact with effector T cells in the tumor microenvironment (TME) itself, mediating both natural antitumor immunity and the response to checkpoint blockade immunotherapy. Using LIPSTIC (Labeling Immune Partnerships by SorTagging Intercellular Contacts)-based single-cell transcriptomics, we identified individual DCs capable of presenting antigen to CD4+ T cells in both the tdLN and TME. Our findings revealed that DCs with similar hyperactivated transcriptional phenotypes interact with helper T cells both in tumors and in the tdLN and that checkpoint blockade drugs enhance these interactions. These findings show that a relatively small fraction of DCs is responsible for most of the antigen presentation in the tdLN and TME to both CD4+ and CD8+ tumor-specific T cells and that classical checkpoint blockade enhances CD40-driven DC activation at both sites.
Collapse
Affiliation(s)
- Aleksey Chudnovskiy
- Laboratory of Lymphocyte Dynamics, Rockefeller University, New York, NY, USA
| | - Tiago B R Castro
- Laboratory of Lymphocyte Dynamics, Rockefeller University, New York, NY, USA
| | | | - Ang Cui
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Harvard School of Dental Medicine, Harvard University, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Chia-Hao Lin
- School of Biological Sciences, University of California, San Diego, La Jolla, CA, USA
- Moores Cancer Center, University of California, San Diego, La Jolla, CA, USA
| | | | - Brooke K Phillips
- Laboratory of Lymphocyte Dynamics, Rockefeller University, New York, NY, USA
| | - Juhee Pae
- Laboratory of Lymphocyte Dynamics, Rockefeller University, New York, NY, USA
| | - Luka Mesin
- Laboratory of Lymphocyte Dynamics, Rockefeller University, New York, NY, USA
| | - Juliana Bortolatto
- Laboratory of Lymphocyte Dynamics, Rockefeller University, New York, NY, USA
| | | | - Giulia Pasqual
- Laboratory of Synthetic Immunology, Oncology and Immunology Section, Department of Surgery Oncology and Gastroenterology, University of Padua, Padua, Italy
- Veneto Institute of Oncology IOV-IRCCS, Padua, Italy
| | - Li-Fan Lu
- School of Biological Sciences, University of California, San Diego, La Jolla, CA, USA
- Moores Cancer Center, University of California, San Diego, La Jolla, CA, USA
| | - Nir Hacohen
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Center for Cancer Research, Massachusetts General Hospital, Boston, MA, USA
| | - Gabriel D Victora
- Laboratory of Lymphocyte Dynamics, Rockefeller University, New York, NY, USA
| |
Collapse
|
30
|
Zheng Y, Li Z, Li S, Zhao P, Wang X, Lu S, Shi Y, Chang H. Separable nanocomposite hydrogel microneedles for intradermal and sustained delivery of antigens to enhance adaptive immune responses. Acta Biomater 2024; 185:203-214. [PMID: 39053817 DOI: 10.1016/j.actbio.2024.07.031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 07/03/2024] [Accepted: 07/18/2024] [Indexed: 07/27/2024]
Abstract
Vaccines play a critical role in combating infectious diseases and cancers, yet improving their efficacy remains challenging. Here, we introduce a separable nanocomposite hydrogel microneedle (NHMN) patch designed for intradermal and sustained delivery of ovalbumin (OVA), a model antigen, to enhance adaptive immune responses. The NHMN patch consists of an array of OVA-loaded microneedles made from photo-cross-linked methacrylated hyaluronic acid and laponite (LAP), supported by a hyaluronic acid backing. The incorporation of LAP not only enhances the mechanical strength of the pure hydrogel microneedles but also significantly prolongs OVA release. Furthermore, in vitro cell experiments demonstrate that NHMNs effectively activate dendritic cells without compromising cell viability. Upon skin penetration, NHMNs detach from the backing as the hyaluronic acid rapidly dissolves upon contact with the skin interstitial fluid, thereby acting as antigen reservoirs to release antigens to abundant skin dendritic cells. NHMNs containing 0.5% w/v LAP achieved a 15-day OVA release in vivo. Immunization studies demonstrate that the intradermal and sustained release of OVA via NHMNs elicited stronger and longer-lasting adaptive immune responses compared to conventional bolus injection. Given its easy to use, painless and minimally invasive features, the NHMN patch shows promise in improving vaccination accessibility and efficacy against a range of diseases. STATEMENT OF SIGNIFICANCE: The study introduces a separable nanocomposite hydrogel microneedle (NHMN) patch. This patch consists of an array of ovalbumin (OVA, a model antigen)-loaded microneedles made from photo-cross-linked methacrylated hyaluronic acid and laponite, with a hyaluronic acid backing, designed for intradermal and sustained delivery of antigens. This patch addresses several key challenges in traditional vaccination methods, including poor antigen uptake and presentation, and rapid systematic clearance. The incorporation of laponite enhances mechanical strength of microneedles, promotes dendritic cell activation, and significantly slows down antigen release. NHMN-based vaccination elicits stronger and longer-lasting adaptive immune responses compared to conventional bolus injection. This NHMN patch holds great potential for improving the efficacy, accessibility, and patient comfort of vaccinations against a range of diseases.
Collapse
Affiliation(s)
- Yanting Zheng
- Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou 310018, Zhejiang, China; College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou 310014, Zhejiang, China
| | - Zhiming Li
- Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou 310018, Zhejiang, China
| | - Shaohua Li
- Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou 310018, Zhejiang, China; College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou 310014, Zhejiang, China
| | - Puxuan Zhao
- Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou 310018, Zhejiang, China; College of Materials Science and Engineering, Zhejiang University of Technology, Hangzhou, Zhejiang, 310014, China
| | - Xin Wang
- Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou 310018, Zhejiang, China; College of Materials Science and Engineering, Zhejiang University of Technology, Hangzhou, Zhejiang, 310014, China
| | - Shaojie Lu
- Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou 310018, Zhejiang, China; College of Materials Science and Engineering, Zhejiang University of Technology, Hangzhou, Zhejiang, 310014, China
| | - Yanan Shi
- Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou 310018, Zhejiang, China; Medical School, Faculty of Medicine, Tianjin University, Tianjin 300072, China
| | - Hao Chang
- Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou 310018, Zhejiang, China.
| |
Collapse
|
31
|
Rakoczy K, Kaczor J, Sołtyk A, Jonderko L, Sędzik M, Lizon J, Lewandowska A, Saczko M, Kulbacka J. Pregnancy, abortion, and birth control methods' complicity with breast cancer occurrence. Mol Cell Endocrinol 2024; 590:112264. [PMID: 38705365 DOI: 10.1016/j.mce.2024.112264] [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: 11/24/2023] [Revised: 04/05/2024] [Accepted: 04/30/2024] [Indexed: 05/07/2024]
Abstract
Reproductive factors play significantly important roles in determining the breast cancer (BC) risk. The impact of pregnancy, abortion, and birth control methods on tumor development remains unclear. It has been found that early full-term pregnancies in young women can lower their lifetime risk of developing the type of cancer in question. However, having a first full-term pregnancy at an older age can increase this risk. The relationship between pregnancy and breast cancer (BC) is, however, much more complicated. Both induced and spontaneous abortions lead to sudden changes in hormonal balance, which could cause different effects on sensitive breast epithelial cells, making abortion a potential risk factor for breast cancer. The influence of hormonal contraception on carcinogenesis is not comprehensively understood, and therefore, more exhaustive analysis of existing data and further investigation is needed. This review explores how the mentioned reproductive factors affect the risk of breast cancer (BC), focusing on the molecular mechanisms that contribute to its complexity. By comprehending this intricate network of relationships, we can develop new strategies for predicting and treating the disease.
Collapse
Affiliation(s)
- Katarzyna Rakoczy
- Faculty of Medicine, Wroclaw Medical University, J. Mikulicza-Radeckiego 5, 50-345, Wroclaw, Poland
| | - Justyna Kaczor
- Faculty of Medicine, Wroclaw Medical University, J. Mikulicza-Radeckiego 5, 50-345, Wroclaw, Poland
| | - Adam Sołtyk
- Faculty of Medicine, Wroclaw Medical University, J. Mikulicza-Radeckiego 5, 50-345, Wroclaw, Poland
| | - Laura Jonderko
- Faculty of Medicine, Wroclaw Medical University, J. Mikulicza-Radeckiego 5, 50-345, Wroclaw, Poland
| | - Mikołaj Sędzik
- Faculty of Medicine, Wroclaw Medical University, J. Mikulicza-Radeckiego 5, 50-345, Wroclaw, Poland
| | - Julia Lizon
- Faculty of Medicine, Wroclaw Medical University, J. Mikulicza-Radeckiego 5, 50-345, Wroclaw, Poland
| | - Anna Lewandowska
- Faculty of Medicine, Wroclaw Medical University, J. Mikulicza-Radeckiego 5, 50-345, Wroclaw, Poland
| | - Małgorzata Saczko
- A. Falkiewicz Specialist Hospital in Wroclaw, Warszawska 2, 52-114 Wroclaw, Poland
| | - Julita Kulbacka
- Department of Molecular and Cellular Biology, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211a, 50-556 Wroclaw, Poland; Department of Immunology and Bioelectrochemistry, State Research Institute Centre for Innovative Medicine, Santariškių 5, 08410, Vilnius, Lithuania.
| |
Collapse
|
32
|
Yoshizaki K, Frias DP, Maier K, Smelan J, Correia AT, Oliveira LMDS, Amato-Lourenço LF, Santillo BT, Prado CM, Oshiro TM, Barbuto JAM, Mauad T, Macchione M. Exposure of cinnamyl alcohol in co-culture of BEAS-2B and dendritic cells: Interaction between CYP450 and cytokines. J Appl Toxicol 2024; 44:1317-1328. [PMID: 38715282 DOI: 10.1002/jat.4623] [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: 01/23/2024] [Revised: 04/18/2024] [Accepted: 04/19/2024] [Indexed: 08/16/2024]
Abstract
The prevalence of fragrances in various hygiene products contributes to their sensorial allure. However, fragrances can induce sensitization in the skin or respiratory system, and the mechanisms involved in this process are incompletely understood. This study investigated the intricate mechanisms underlying the fragrance's effects on sensitization response, focusing on the interplay between CYP450 enzymes, a class of drug-metabolizing enzymes, and the adaptive immune system. Specifically, we assessed the expression of CYP450 enzymes and cytokine profiles in culture of BEAS-2B and mature dendritic cells (mDC) alone or in co-culture stimulated with 2 mM of a common fragrance, cinnamyl alcohol (CA) for 20 h. CYP1A1, CYP1A2, CYP1B1, CYP2A6, and CYP2A13 were analyzed by RT-PCR and IL-10, IL-12p70, IL-18, IL-33, and thymic stromal lymphopoietin (TSLP) by Cytometric Bead Array (CBA). Through RT-PCR analysis, we observed that CA increased CYP1A2 and CYP1B1 expression in BEAS-2B, with a further increased in BEAS-2B-mDC co-culture. Additionally, exposure to CA increased IL-12p70 levels in mDC rather than in BEAS-2B-mDC co-culture. In regards to IL-18, level was higher in BEAS-2B than in BEAS-2B-mDC co-culture. A positive correlation between the levels of IL-10 and CYP1B1 was found in mDC-CA-exposed and between IL-12p70 and CYP1A1 was found in BEAS-2B after CA exposure. However, IL-12p70 and CYP1A2 as well as IL-18, IL-33, and CYP1A1 levels were negative, correlated mainly in co-culture control. These correlations highlight potential immunomodulatory interactions and complex regulatory relationships. Overall, exposure to CA enhances CYP450 expression, suggesting that CA can influence immune responses by degrading ligands on xenosensitive transcription factors.
Collapse
Affiliation(s)
- Kelly Yoshizaki
- Laboratory of Experimental Environmental Pathology, Department of Pathology, São Paulo University Medical School, São Paulo, Brazil
| | - Daniela Perroni Frias
- Laboratory of Experimental Environmental Pathology, Department of Pathology, São Paulo University Medical School, São Paulo, Brazil
| | - Kevin Maier
- Laboratory of Experimental Environmental Pathology, Department of Pathology, São Paulo University Medical School, São Paulo, Brazil
| | - Juliana Smelan
- Laboratory of Experimental Environmental Pathology, Department of Pathology, São Paulo University Medical School, São Paulo, Brazil
| | - Aristides Tadeu Correia
- Thoracic Surgery Division, Department of Cardiopneumology, InCor, Clinics Hospital, School of Medicine, University of São Paulo, São Paulo, Brazil
| | - Luanda Mara da Silva Oliveira
- Laboratory of Medical Investigation in Dermatology and Immunodeficiences, Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Luís Fernando Amato-Lourenço
- Laboratory of Experimental Environmental Pathology, Department of Pathology, São Paulo University Medical School, São Paulo, Brazil
- Institute of Advanced Studies (IEA) Global Cities Program, University of São Paulo, São Paulo, Brazil
| | - Bruna Tereso Santillo
- Laboratory of Medical Investigation in Dermatology and Immunodeficiences, Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | | | - Telma Miyuki Oshiro
- Laboratory of Medical Investigation in Dermatology and Immunodeficiences, Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Jose Alexandre M Barbuto
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Thais Mauad
- Laboratory of Experimental Environmental Pathology, Department of Pathology, São Paulo University Medical School, São Paulo, Brazil
| | - Mariangela Macchione
- Laboratory of Experimental Environmental Pathology, Department of Pathology, São Paulo University Medical School, São Paulo, Brazil
| |
Collapse
|
33
|
Yadav P, Vengoji R, Jain M, Batra SK, Shonka N. Pathophysiological role of histamine signaling and its implications in glioblastoma. Biochim Biophys Acta Rev Cancer 2024; 1879:189146. [PMID: 38955315 PMCID: PMC11770814 DOI: 10.1016/j.bbcan.2024.189146] [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: 01/08/2024] [Revised: 06/21/2024] [Accepted: 06/27/2024] [Indexed: 07/04/2024]
Abstract
Glioblastoma (GBM), an extremely aggressive and prevalent malignant brain tumor, remains a challenge to treat. Despite a multimodality treatment approach, GBM recurrence remains inevitable, particularly with the emergence of temozolomide (TMZ) resistance and limited treatment options. Surprisingly, previous studies show that a history of allergies, atopy, or asthma is inversely associated with GBM risk. Further, the electronic medical record at the University Hospital of Lausanne showed that the GBM patients taking antihistamine during treatment had better survival. Histamine is an essential neurotransmitter in the brain and plays a significant role in regulating sleep, hormonal balance, and cognitive functions. Elevated levels of histamine and increased histamine receptor expression have been found in different tumors and their microenvironments, including GBM. High histamine 1 receptor (HRH1) expression is inversely related to overall and progression-free survival in GBM patients, further emphasizing the role of histamine in disease progression. This review aims to provide insights into the challenges of GBM treatment, the role of histamine in GBM progression, and the rationale for considering antihistamines as targeted therapy. The review concludes by encouraging further investigation into antihistamine mechanisms and their impact on the tumor microenvironment.
Collapse
Affiliation(s)
- Poonam Yadav
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198-5870, USA
| | - Raghupathy Vengoji
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198-5870, USA
| | - Maneesh Jain
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198-5870, USA; Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198-5950, USA; Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198-5950, USA
| | - Surinder K Batra
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198-5870, USA; Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198-5950, USA; Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198-5950, USA.
| | - Nicole Shonka
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198-5950, USA; Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68198-6840, USA.
| |
Collapse
|
34
|
Kim SE, Kim YK, Oh KB, Hwang JH. Development of the PD9-9 Monoclonal Antibody for Identifying Porcine Bone Marrow-Derived Dendritic Cells. Life (Basel) 2024; 14:1054. [PMID: 39337839 PMCID: PMC11433566 DOI: 10.3390/life14091054] [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: 07/03/2024] [Revised: 08/03/2024] [Accepted: 08/19/2024] [Indexed: 09/30/2024] Open
Abstract
The purpose of this study was to develop a monoclonal antibody (mAb) that can identify porcine dendritic cells (DCs) that have differentiated from bone marrow progenitor cells. Hybridoma technology was used to obtain mAbs, and bone marrow-derived DCs (BMDCs) were employed as immunogens for producing antibodies. The generated PD9-9 mAbs exhibited considerable reactivity towards porcine BMDCs with applications in flow cytometry and immunostaining. The antibody was composed of heavy immunoglobulin gamma-1 chains and light kappa chains. The PD9-9 mAb recognized fully differentiated porcine BMDCs and cells undergoing DC differentiation. In contrast, bone marrow cells and macrophages were not recognized by PD9-9. In addition, the PD9-9 mAb promoted porcine DC proliferation. Consequently, the PD9-9 mAb may be a biomarker for porcine DCs and will be advantageous for investigating porcine DC biology.
Collapse
Affiliation(s)
- Sang Eun Kim
- Department of Stem Cell and Regenerative Biotechnology, KU Institute of Science and Technology (KIT), Konkuk University, Seoul 05029, Republic of Korea
- Animal Biotechnology Division, National Institute of Animal Science, Rural Development Administration, Wanju-gun 55365, Republic of Korea;
| | - Young Kyu Kim
- Department of Stem Cell and Regenerative Biotechnology, KU Institute of Science and Technology (KIT), Konkuk University, Seoul 05029, Republic of Korea
- Animal Model Research Group, Korea Institute of Toxicology, 30 Baekhak1-gil, Jeongeup-si 56212, Republic of Korea;
| | - Keon Bong Oh
- Animal Biotechnology Division, National Institute of Animal Science, Rural Development Administration, Wanju-gun 55365, Republic of Korea;
| | - Jeong Ho Hwang
- Department of Stem Cell and Regenerative Biotechnology, KU Institute of Science and Technology (KIT), Konkuk University, Seoul 05029, Republic of Korea
- Animal Model Research Group, Korea Institute of Toxicology, 30 Baekhak1-gil, Jeongeup-si 56212, Republic of Korea;
| |
Collapse
|
35
|
Liu D, Liu L, Li X, Wang S, Wu G, Che X. Advancements and Challenges in Peptide-Based Cancer Vaccination: A Multidisciplinary Perspective. Vaccines (Basel) 2024; 12:950. [PMID: 39204073 PMCID: PMC11359700 DOI: 10.3390/vaccines12080950] [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: 06/28/2024] [Revised: 08/09/2024] [Accepted: 08/21/2024] [Indexed: 09/03/2024] Open
Abstract
With the continuous advancements in tumor immunotherapy, researchers are actively exploring new treatment methods. Peptide therapeutic cancer vaccines have garnered significant attention for their potential in improving patient outcomes. Despite its potential, only a single peptide-based cancer vaccine has been approved by the U.S. Food and Drug Administration (FDA). A comprehensive understanding of the underlying mechanisms and current development status is crucial for advancing these vaccines. This review provides an in-depth analysis of the production principles and therapeutic mechanisms of peptide-based cancer vaccines, highlights the commonly used peptide-based cancer vaccines, and examines the synergistic effects of combining these vaccines with immunotherapy, targeted therapy, radiotherapy, and chemotherapy. While some studies have yielded suboptimal results, the potential of combination therapies remains substantial. Additionally, we addressed the management and adverse events associated with peptide-based cancer vaccines, noting their relatively higher safety profile compared to traditional radiotherapy and chemotherapy. Lastly, we also discussed the roles of adjuvants and targeted delivery systems in enhancing vaccine efficacy. In conclusion, this review comprehensively outlines the current landscape of peptide-based cancer vaccination and underscores its potential as a pivotal immunotherapy approach.
Collapse
Affiliation(s)
- Dequan Liu
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China; (D.L.); (L.L.); (S.W.)
| | - Lei Liu
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China; (D.L.); (L.L.); (S.W.)
| | - Xinghan Li
- Department of Stomatology, General Hospital of Northern Theater Command, Shenyang 110016, China;
| | - Shijin Wang
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China; (D.L.); (L.L.); (S.W.)
| | - Guangzhen Wu
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China; (D.L.); (L.L.); (S.W.)
| | - Xiangyu Che
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China; (D.L.); (L.L.); (S.W.)
| |
Collapse
|
36
|
Rogers J, Bajur AT, Salaita K, Spillane KM. Mechanical control of antigen detection and discrimination by T and B cell receptors. Biophys J 2024; 123:2234-2255. [PMID: 38794795 PMCID: PMC11331051 DOI: 10.1016/j.bpj.2024.05.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 05/10/2024] [Accepted: 05/21/2024] [Indexed: 05/26/2024] Open
Abstract
The adaptive immune response is orchestrated by just two cell types, T cells and B cells. Both cells possess the remarkable ability to recognize virtually any antigen through their respective antigen receptors-the T cell receptor (TCR) and B cell receptor (BCR). Despite extensive investigations into the biochemical signaling events triggered by antigen recognition in these cells, our ability to predict or control the outcome of T and B cell activation remains elusive. This challenge is compounded by the sensitivity of T and B cells to the biophysical properties of antigens and the cells presenting them-a phenomenon we are just beginning to understand. Recent insights underscore the central role of mechanical forces in this process, governing the conformation, signaling activity, and spatial organization of TCRs and BCRs within the cell membrane, ultimately eliciting distinct cellular responses. Traditionally, T cells and B cells have been studied independently, with researchers working in parallel to decipher the mechanisms of activation. While these investigations have unveiled many overlaps in how these cell types sense and respond to antigens, notable differences exist. To fully grasp their biology and harness it for therapeutic purposes, these distinctions must be considered. This review compares and contrasts the TCR and BCR, placing emphasis on the role of mechanical force in regulating the activity of both receptors to shape cellular and humoral adaptive immune responses.
Collapse
Affiliation(s)
- Jhordan Rogers
- Department of Chemistry, Emory University, Atlanta, Georgia
| | - Anna T Bajur
- Department of Physics, King's College London, London, United Kingdom; Randall Centre for Cell and Molecular Biophysics, King's College London, London, United Kingdom
| | - Khalid Salaita
- Department of Chemistry, Emory University, Atlanta, Georgia; Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia.
| | - Katelyn M Spillane
- Department of Physics, King's College London, London, United Kingdom; Randall Centre for Cell and Molecular Biophysics, King's College London, London, United Kingdom; Department of Life Sciences, Imperial College London, London, United Kingdom.
| |
Collapse
|
37
|
Quiniou G, Andromaque L, Duclaux-Loras R, Dinet O, Cervantes O, Verdet M, Meunier C, Boschetti G, Viret C, Nancey S, Faure M, Rozières A. Impaired reprogramming of the autophagy flux in maturing dendritic cells from crohn disease patients with core autophagy gene-related polymorphisms. Autophagy 2024; 20:1837-1853. [PMID: 38615686 PMCID: PMC11262231 DOI: 10.1080/15548627.2024.2338574] [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: 03/02/2023] [Revised: 03/16/2024] [Accepted: 03/29/2024] [Indexed: 04/16/2024] Open
Abstract
Crohn disease (CD) is an inflammatory bowel disease whose pathogenesis involves inappropriate immune responses toward gut microbiota on genetically predisposed backgrounds. Notably, CD is associated with single-nucleotide polymorphisms affecting several genes involved in macroautophagy/autophagy, the catabolic process that ensures the degradation and recycling of cytosolic components and microorganisms. In a clinical translation perspective, monitoring the autophagic activity of CD patients will require some knowledge on the intrinsic functional status of autophagy. Here, we focused on monocyte-derived dendritic cells (DCs) to characterize the intrinsic quantitative features of the autophagy flux. Starting with DCs from healthy donors, we documented a reprogramming of the steady state flux during the transition from the immature to mature status: both the autophagosome pool size and the flux were diminished at the mature stage while the autophagosome turnover remained stable. At the cohort level, DCs from CD patients were comparable to control in term of autophagy flux reprogramming capacity. However, the homozygous presence of ATG16L1 rs2241880 A>G (T300A) and ULK1 rs12303764 (G/T) polymorphisms abolished the capacity of CD patient DCs to reprogram their autophagy flux during maturation. This effect was not seen in the case of CD patients heterozygous for these polymorphisms, revealing a gene dose dependency effect. In contrast, the NOD2 rs2066844 c.2104C>T (R702W) polymorphism did not alter the flux reprogramming capacity of DCs. The data, opening new clinical translation perspectives, indicate that polymorphisms affecting autophagy-related genes can differentially influence the capacity of DCs to reprogram their steady state autophagy flux when exposed to proinflammatory challenges.Abbreviation: BAFA1: bafilomycin A1, CD: Crohn disease; DC: dendritic cells; HD: healthy donor; iDCs: immature DCs; IL: interleukin; J: autophagosome flux; LPS: lipopolysaccharide; MHC: major histocompatibility complex; nA: autophagosome pool size; SNPs: single-nucleotide polymorphisms; PCA: principal component analysis; TLR: toll like receptor; τ: transition time; TNF: tumor necrosis factor.
Collapse
Affiliation(s)
- Gaëlle Quiniou
- CIRI, Centre International de Recherche en Infectiologie, Université de Lyon, Inserm U1111, Université Claude Bernard Lyon 1, CNRS, Lyon, France
| | - Leslie Andromaque
- CIRI, Centre International de Recherche en Infectiologie, Université de Lyon, Inserm U1111, Université Claude Bernard Lyon 1, CNRS, Lyon, France
| | - Rémi Duclaux-Loras
- CIRI, Centre International de Recherche en Infectiologie, Université de Lyon, Inserm U1111, Université Claude Bernard Lyon 1, CNRS, Lyon, France
- Department of Pediatric Hepatology, Gastroenterology and Nutrition, Femme-Mère-Enfant Hospital, Hospices Civils de Lyon, Bron, France
| | - Océane Dinet
- CIRI, Centre International de Recherche en Infectiologie, Université de Lyon, Inserm U1111, Université Claude Bernard Lyon 1, CNRS, Lyon, France
| | - Ornella Cervantes
- CIRI, Centre International de Recherche en Infectiologie, Université de Lyon, Inserm U1111, Université Claude Bernard Lyon 1, CNRS, Lyon, France
| | - Mallorie Verdet
- CIRI, Centre International de Recherche en Infectiologie, Université de Lyon, Inserm U1111, Université Claude Bernard Lyon 1, CNRS, Lyon, France
| | - Camille Meunier
- CIRI, Centre International de Recherche en Infectiologie, Université de Lyon, Inserm U1111, Université Claude Bernard Lyon 1, CNRS, Lyon, France
- Department of Gastroenterology, Lyon-Sud university hospital, Lyon, France
| | - Gilles Boschetti
- CIRI, Centre International de Recherche en Infectiologie, Université de Lyon, Inserm U1111, Université Claude Bernard Lyon 1, CNRS, Lyon, France
- Department of Gastroenterology, Lyon-Sud university hospital, Lyon, France
| | - Christophe Viret
- CIRI, Centre International de Recherche en Infectiologie, Université de Lyon, Inserm U1111, Université Claude Bernard Lyon 1, CNRS, Lyon, France
| | - Stéphane Nancey
- CIRI, Centre International de Recherche en Infectiologie, Université de Lyon, Inserm U1111, Université Claude Bernard Lyon 1, CNRS, Lyon, France
- Department of Gastroenterology, Lyon-Sud university hospital, Lyon, France
| | - Mathias Faure
- CIRI, Centre International de Recherche en Infectiologie, Université de Lyon, Inserm U1111, Université Claude Bernard Lyon 1, CNRS, Lyon, France
- Equipe Labellisée par la Fondation pour la Recherche Médicale, FRM, France
| | - Aurore Rozières
- CIRI, Centre International de Recherche en Infectiologie, Université de Lyon, Inserm U1111, Université Claude Bernard Lyon 1, CNRS, Lyon, France
| |
Collapse
|
38
|
Ancuţa DL, Alexandru DM, Ţucureanu C, Coman C. A Comparative Analysis of the Efficacy of Bacterial Lysate versus Antibiotic Therapy in the Treatment of Experimental Peri-Implantitis in Rats. Microorganisms 2024; 12:1537. [PMID: 39203379 PMCID: PMC11356466 DOI: 10.3390/microorganisms12081537] [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: 06/27/2024] [Revised: 07/25/2024] [Accepted: 07/25/2024] [Indexed: 09/03/2024] Open
Abstract
Peri-implantitis (PI) is a current concern whose understanding and resolution are ongoing. We aimed to evaluate in vivo a new treatment with antibacterial properties, based on bacterial lysates obtained from the strains of Aggregatibacter actinomycetemcomitans, Streptococcus oralis, and Fusobacterium nucleatum. This research was conducted on 30 rats with PI which were divided into three groups and treated with antibiotic and anti-inflammatory (AAi) drugs, bacterial lysates (BLs), and saline (C), respectively. The monitoring period included the clinical and paraclinical examination where hematological, immunological, imaging, and histopathological analysis were performed. No particular clinical signs were observed, but the radiological examination showed the loss of all implants in group C, in contrast to group BL which had the highest survival rate of devices. White cells showed a decrease from the PI period, as did the immunological analysis. Only IL-6 showed an increase in the AAi and BL groups. Histopathologically, the C group presented a high degree of bone destruction, and in the BL group, many attenuated inflammatory phenomena appeared compared to the AAi animals. Bacterial lysates have similar effects to antibiotic-based therapeutic regimens for PI, and their future use may help to improve the current therapeutic management of the disease.
Collapse
Affiliation(s)
- Diana Larisa Ancuţa
- Cantacuzino National Medical Military Institute for Research and Development, 050096 Bucharest, Romania; (D.L.A.); (C.Ţ.)
- Faculty of Veterinary Medicine, University of Agronomic Sciences and Veterinary Medicine, 050097 Bucharest, Romania
| | - Diana Mihaela Alexandru
- Faculty of Veterinary Medicine, University of Agronomic Sciences and Veterinary Medicine, 050097 Bucharest, Romania
| | - Cătălin Ţucureanu
- Cantacuzino National Medical Military Institute for Research and Development, 050096 Bucharest, Romania; (D.L.A.); (C.Ţ.)
| | - Cristin Coman
- Cantacuzino National Medical Military Institute for Research and Development, 050096 Bucharest, Romania; (D.L.A.); (C.Ţ.)
- Center of Excellence in Translational Medicine, Fundeni Clinical Institute, 022328 Bucharest, Romania
| |
Collapse
|
39
|
Li S, Luo X, Sun M, Wang Y, Zhang Z, Jiang J, Hu D, Zhang J, Wu Z, Wang Y, Huang W, Xia L. Context-dependent T-BOX transcription factor family: from biology to targeted therapy. Cell Commun Signal 2024; 22:350. [PMID: 38965548 PMCID: PMC11225425 DOI: 10.1186/s12964-024-01719-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 06/17/2024] [Indexed: 07/06/2024] Open
Abstract
T-BOX factors belong to an evolutionarily conserved family of transcription factors. T-BOX factors not only play key roles in growth and development but are also involved in immunity, cancer initiation, and progression. Moreover, the same T-BOX molecule exhibits different or even opposite effects in various developmental processes and tumor microenvironments. Understanding the multiple roles of context-dependent T-BOX factors in malignancies is vital for uncovering the potential of T-BOX-targeted cancer therapy. We summarize the physiological roles of T-BOX factors in different developmental processes and their pathological roles observed when their expression is dysregulated. We also discuss their regulatory roles in tumor immune microenvironment (TIME) and the newly arising questions that remain unresolved. This review will help in systematically and comprehensively understanding the vital role of the T-BOX transcription factor family in tumor physiology, pathology, and immunity. The intention is to provide valuable information to support the development of T-BOX-targeted therapy.
Collapse
Affiliation(s)
- Siwen Li
- Department of Gastroenterology, Institute of Liver and Gastrointestinal Diseases, Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, 430030, China
| | - Xiangyuan Luo
- Department of Gastroenterology, Institute of Liver and Gastrointestinal Diseases, Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, 430030, China
| | - Mengyu Sun
- Department of Gastroenterology, Institute of Liver and Gastrointestinal Diseases, Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, 430030, China
| | - Yijun Wang
- Department of Gastroenterology, Institute of Liver and Gastrointestinal Diseases, Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, 430030, China
| | - Zerui Zhang
- Department of Gastroenterology, Institute of Liver and Gastrointestinal Diseases, Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, 430030, China
| | - Junqing Jiang
- Department of Gastroenterology, Institute of Liver and Gastrointestinal Diseases, Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, 430030, China
| | - Dian Hu
- Department of Gastroenterology, Institute of Liver and Gastrointestinal Diseases, Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, 430030, China
| | - Jiaqian Zhang
- Department of Gastroenterology, Institute of Liver and Gastrointestinal Diseases, Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, 430030, China
| | - Zhangfan Wu
- Department of Gastroenterology, Institute of Liver and Gastrointestinal Diseases, Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, 430030, China
| | - Yufei Wang
- Department of Gastroenterology, Institute of Liver and Gastrointestinal Diseases, Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, 430030, China
| | - Wenjie Huang
- Department of Gastroenterology, Institute of Liver and Gastrointestinal Diseases, Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, 430030, China.
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Clinical Medicine Research Center for Hepatic Surgery of Hubei Province, Key Laboratory of Organ Transplantation, Huazhong University of Science and Technology, Ministry of Education and Ministry of Public Health, Wuhan, Hubei, 430030, China.
| | - Limin Xia
- Department of Gastroenterology, Institute of Liver and Gastrointestinal Diseases, Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, 430030, China.
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, 710032, China.
| |
Collapse
|
40
|
Yang C, Jiang Y, Zhang K, Zhu X, Li J, Yu H, Chen J, Gu X, Gan Z, Yu Q. Photodynamic Therapy Derived Personalized Whole Cell Tumor Vaccine Prevents Postsurgery Tumor Recurrence and Metastasis. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2308456. [PMID: 38342675 DOI: 10.1002/smll.202308456] [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: 09/22/2023] [Revised: 01/03/2024] [Indexed: 02/13/2024]
Abstract
In order to avoid the time-consuming and laborious identification of tumor-specific antigens (TSAs) during the traditional vaccine fabrication process, a versatile photodynamic therapy (PDT)-based method is developed to construct a whole-tumor antigen tumor vaccine (TV) from surgically resected tumor tissues for personalized immunotherapy. Mucoadhesive nanoparticles containing small-molecular photosensitizer are fabricated and directly co-incubated with suspended tumor cells obtained after cytoreduction surgery. After irradiation with a 405 nm laser, potent immunogenic cell death of cancer cells could be induced. Along with the release of TSAs, the as-prepared TV could activate safe and robust tumor-specific immune responses, leading to efficient suppression of postsurgery tumor recurrence and metastasis. The as-prepared TV cannot only be applied alone through various administration routes but also synergize with immunoadjuvant, chemotherapeutics, and immune checkpoint blockers to exert more potent immune responses. This work provides an alternative way to promote the clinical translation of PDT, which is generally restricted by the limited penetration of light. Moreover, the versatile strategy of vaccine fabrication also facilitates the clinical application of personalized whole-cell tumor vaccines.
Collapse
Affiliation(s)
- Chunyu Yang
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, State Key Laboratory of Chemical Resource Engineering, College of Materials Science and Engineering, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Yitong Jiang
- The State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Kaixin Zhang
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, State Key Laboratory of Chemical Resource Engineering, College of Materials Science and Engineering, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Xianqi Zhu
- The State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Jianlin Li
- The State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Haiwang Yu
- The State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Jiawei Chen
- The State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Xinggui Gu
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, State Key Laboratory of Chemical Resource Engineering, College of Materials Science and Engineering, Beijing University of Chemical Technology, Beijing, 100029, China
- Beijing National Laboratory for Molecular Sciences, Beijing, 100190, China
| | - Zhihua Gan
- The State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Qingsong Yu
- The State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, China
| |
Collapse
|
41
|
Lusta KA, Summerhill VI, Khotina VA, Sukhorukov VN, Glanz VY, Orekhov AN. The Role of Bacterial Extracellular Membrane Nanovesicles in Atherosclerosis: Unraveling a Potential Trigger. Curr Atheroscler Rep 2024; 26:289-304. [PMID: 38805145 DOI: 10.1007/s11883-024-01206-6] [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] [Accepted: 04/30/2024] [Indexed: 05/29/2024]
Abstract
PURPOSE OF REVIEW In this review, we explore the intriguing and evolving connections between bacterial extracellular membrane nanovesicles (BEMNs) and atherosclerosis development, highlighting the evidence on molecular mechanisms by which BEMNs can promote the athero-inflammatory process that is central to the progression of atherosclerosis. RECENT FINDINGS Atherosclerosis is a chronic inflammatory disease primarily driven by metabolic and lifestyle factors; however, some studies have suggested that bacterial infections may contribute to the development of both atherogenesis and inflammation in atherosclerotic lesions. In particular, the participation of BEMNs in atherosclerosis pathogenesis has attracted special attention. We provide some general insights into how the immune system responds to potential threats such as BEMNs during the development of atherosclerosis. A comprehensive understanding of contribution of BEMNs to atherosclerosis pathogenesis may lead to the development of targeted interventions for the prevention and treatment of the disease.
Collapse
Affiliation(s)
- Konstantin A Lusta
- Laboratory of Cellular and Molecular Pathology of Cardiovascular System, Petrovsky Russian National Center of Surgery, Moscow, 119991, Russia
| | - Volha I Summerhill
- Department of Research and Development, Institute for Atherosclerosis Research, Moscow, 121609, Russia.
| | - Victoria A Khotina
- Laboratory of Cellular and Molecular Pathology of Cardiovascular System, Petrovsky Russian National Center of Surgery, Moscow, 119991, Russia
| | - Vasily N Sukhorukov
- Laboratory of Cellular and Molecular Pathology of Cardiovascular System, Petrovsky Russian National Center of Surgery, Moscow, 119991, Russia
| | - Victor Y Glanz
- Laboratory of Cellular and Molecular Pathology of Cardiovascular System, Petrovsky Russian National Center of Surgery, Moscow, 119991, Russia
| | - Alexander N Orekhov
- Laboratory of Cellular and Molecular Pathology of Cardiovascular System, Petrovsky Russian National Center of Surgery, Moscow, 119991, Russia.
- Department of Research and Development, Institute for Atherosclerosis Research, Moscow, 121609, Russia.
| |
Collapse
|
42
|
Alraies Z, Rivera CA, Delgado MG, Sanséau D, Maurin M, Amadio R, Maria Piperno G, Dunsmore G, Yatim A, Lacerda Mariano L, Kniazeva A, Calmettes V, Sáez PJ, Williart A, Popard H, Gratia M, Lamiable O, Moreau A, Fusilier Z, Crestey L, Albaud B, Legoix P, Dejean AS, Le Dorze AL, Nakano H, Cook DN, Lawrence T, Manel N, Benvenuti F, Ginhoux F, Moreau HD, P F Nader G, Piel M, Lennon-Duménil AM. Cell shape sensing licenses dendritic cells for homeostatic migration to lymph nodes. Nat Immunol 2024; 25:1193-1206. [PMID: 38834865 PMCID: PMC11224020 DOI: 10.1038/s41590-024-01856-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 04/25/2024] [Indexed: 06/06/2024]
Abstract
Immune cells experience large cell shape changes during environmental patrolling because of the physical constraints that they encounter while migrating through tissues. These cells can adapt to such deformation events using dedicated shape-sensing pathways. However, how shape sensing affects immune cell function is mostly unknown. Here, we identify a shape-sensing mechanism that increases the expression of the chemokine receptor CCR7 and guides dendritic cell migration from peripheral tissues to lymph nodes at steady state. This mechanism relies on the lipid metabolism enzyme cPLA2, requires nuclear envelope tensioning and is finely tuned by the ARP2/3 actin nucleation complex. We also show that this shape-sensing axis reprograms dendritic cell transcription by activating an IKKβ-NF-κB-dependent pathway known to control their tolerogenic potential. These results indicate that cell shape changes experienced by immune cells can define their migratory behavior and immunoregulatory properties and reveal a contribution of the physical properties of tissues to adaptive immunity.
Collapse
Affiliation(s)
- Zahraa Alraies
- INSERM U932, Immunity and Cancer, Institut Curie, PSL University, Paris, France
| | - Claudia A Rivera
- INSERM U932, Immunity and Cancer, Institut Curie, PSL University, Paris, France
| | | | - Doriane Sanséau
- INSERM U932, Immunity and Cancer, Institut Curie, PSL University, Paris, France
| | - Mathieu Maurin
- INSERM U932, Immunity and Cancer, Institut Curie, PSL University, Paris, France
| | - Roberto Amadio
- Cellular Immunology, International Centre for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy
| | - Giulia Maria Piperno
- Cellular Immunology, International Centre for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy
| | - Garett Dunsmore
- INSERM U1015, Gustave Roussy Cancer Campus, Villejuif, France
| | - Aline Yatim
- INSERM U932, Immunity and Cancer, Institut Curie, PSL University, Paris, France
| | | | - Anna Kniazeva
- INSERM U932, Immunity and Cancer, Institut Curie, PSL University, Paris, France
| | - Vincent Calmettes
- INSERM U932, Immunity and Cancer, Institut Curie, PSL University, Paris, France
| | - Pablo J Sáez
- Cell Communication and Migration Laboratory, Institute of Biochemistry and Molecular Cell Biology, Center for Experimental Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Alice Williart
- CNRS UMR144, Institut Curie, PSL Research University, Paris, France
| | - Henri Popard
- CNRS UMR144, Institut Curie, PSL Research University, Paris, France
| | - Matthieu Gratia
- INSERM U932, Immunity and Cancer, Institut Curie, PSL University, Paris, France
| | | | - Aurélie Moreau
- Center for Research in Transplantation and Translational Immunology, UMR 1064, INSERM, Nantes Université, Nantes, France
| | - Zoé Fusilier
- INSERM U932, Immunity and Cancer, Institut Curie, PSL University, Paris, France
- INSERM U932, Immunity and Cancer, Institut Curie, Paris-Cité University, Paris, France
| | - Lou Crestey
- INSERM U932, Immunity and Cancer, Institut Curie, PSL University, Paris, France
| | | | - Patricia Legoix
- INSERM U932, Immunity and Cancer, Institut Curie, PSL University, Paris, France
| | - Anne S Dejean
- INSERM UMR1291, CNRS UMR5051, Institut Toulousain des Maladies Infectieuses et Inflammatoires (INFINITy), Université Toulouse III, Toulouse, France
| | - Anne-Louise Le Dorze
- INSERM UMR1291, CNRS UMR5051, Institut Toulousain des Maladies Infectieuses et Inflammatoires (INFINITy), Université Toulouse III, Toulouse, France
| | - Hideki Nakano
- Immunity, Inflammation, and Disease Laboratory, National Institute of Environmental Health Sciences (NIEHS), National Institutes of Health (NIH), Research Triangle Park, NC, USA
| | - Donald N Cook
- Immunity, Inflammation, and Disease Laboratory, National Institute of Environmental Health Sciences (NIEHS), National Institutes of Health (NIH), Research Triangle Park, NC, USA
| | - Toby Lawrence
- Centre d'Immunologie de Marseille-Luminy, INSERM, CNRS, Université Aix-Marseille, Marseille, France
- Centre for Inflammation Biology and Cancer Immunology, School of Immunology and Microbial Sciences, King's College London, London, UK
- Henan Key Laboratory of Immunology and Targeted Therapy, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, China
| | - Nicolas Manel
- INSERM U932, Immunity and Cancer, Institut Curie, PSL University, Paris, France
| | - Federica Benvenuti
- Cellular Immunology, International Centre for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy
| | - Florent Ginhoux
- INSERM U1015, Gustave Roussy Cancer Campus, Villejuif, France
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), 8A Biomedical Grove, Immunos, Singapore, Singapore
- Department of Immunology and Microbiology, Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Translational Immunology Institute, SingHealth Duke-NUS Academic Medical Centre, Singapore, Singapore
| | - Hélène D Moreau
- INSERM U932, Immunity and Cancer, Institut Curie, PSL University, Paris, France
| | - Guilherme P F Nader
- CNRS UMR144, Institut Curie, PSL Research University, Paris, France
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia and University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Matthieu Piel
- CNRS UMR144, Institut Curie, PSL Research University, Paris, France.
| | | |
Collapse
|
43
|
Fowler JWM, Song L, Tam K, Roth Flach RJ. Targeting lymphatic function in cardiovascular-kidney-metabolic syndrome: preclinical methods to analyze lymphatic function and therapeutic opportunities. Front Cardiovasc Med 2024; 11:1412857. [PMID: 38915742 PMCID: PMC11194411 DOI: 10.3389/fcvm.2024.1412857] [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: 04/05/2024] [Accepted: 05/24/2024] [Indexed: 06/26/2024] Open
Abstract
The lymphatic vascular system spans nearly every organ in the body and serves as an important network that maintains fluid, metabolite, and immune cell homeostasis. Recently, there has been a growing interest in the role of lymphatic biology in chronic disorders outside the realm of lymphatic abnormalities, lymphedema, or oncology, such as cardiovascular-kidney-metabolic syndrome (CKM). We propose that enhancing lymphatic function pharmacologically may be a novel and effective way to improve quality of life in patients with CKM syndrome by engaging multiple pathologies at once throughout the body. Several promising therapeutic targets that enhance lymphatic function have already been reported and may have clinical benefit. However, much remains unclear of the discreet ways the lymphatic vasculature interacts with CKM pathogenesis, and translation of these therapeutic targets to clinical development is challenging. Thus, the field must improve characterization of lymphatic function in preclinical mouse models of CKM syndrome to better understand molecular mechanisms of disease and uncover effective therapies.
Collapse
Affiliation(s)
| | | | | | - Rachel J. Roth Flach
- Internal Medicine Research Unit, Pfizer Research and Development, Cambridge, MA, United States
| |
Collapse
|
44
|
Troise D, Infante B, Mercuri S, Catalano V, Ranieri E, Stallone G. Dendritic Cells: A Bridge between Tolerance Induction and Cancer Development in Transplantation Setting. Biomedicines 2024; 12:1240. [PMID: 38927447 PMCID: PMC11200833 DOI: 10.3390/biomedicines12061240] [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: 04/29/2024] [Revised: 05/23/2024] [Accepted: 05/30/2024] [Indexed: 06/28/2024] Open
Abstract
Dendritic cells (DCs) are a heterogeneous group of antigen-presenting cells crucial for fostering allograft tolerance while simultaneously supporting host defense against infections and cancer. Within the tumor microenvironment, DCs can either mount an immune response against cancer cells or foster immunotolerance, presenting a dual role. In immunocompromised individuals, posttransplant malignancies pose a significant health concern, with DCs serving as vital players in immune responses against cancer cells. Both recipient- and donor-derived DCs play a critical role in the rejection process, infiltrating the transplanted organ and sustaining T-cell responses. The use of immunosuppressive drugs represents the predominant approach to control this immunological barrier in transplanted organs. Evidence has shed light on the immunopharmacology of these drugs and novel strategies for manipulating DCs to promote allograft survival. Therefore, comprehending the mechanisms underlying this intricate microenvironment and the effects of immunosuppressive therapy on DCs is crucial for developing targeted therapies to reduce graft failure rates. This review will delve into the fundamental immunobiology of DCs and provide a detailed exploration of their clinical significance concerning alloimmune responses and posttransplant malignancies.
Collapse
Affiliation(s)
- Dario Troise
- Nephrology, Dialysis and Transplantation Unit, Advanced Research Center on Kidney Aging (A.R.K.A.), Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy
- Renal Medicine and Baxter Novum, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, 141 52 Stockholm, Sweden
| | - Barbara Infante
- Nephrology, Dialysis and Transplantation Unit, Advanced Research Center on Kidney Aging (A.R.K.A.), Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy
| | - Silvia Mercuri
- Nephrology, Dialysis and Transplantation Unit, Advanced Research Center on Kidney Aging (A.R.K.A.), Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy
| | - Valeria Catalano
- Unit of Clinical Pathology, Advanced Research Center on Kidney Aging (A.R.K.A.), Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy
| | - Elena Ranieri
- Unit of Clinical Pathology, Advanced Research Center on Kidney Aging (A.R.K.A.), Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy
| | - Giovanni Stallone
- Nephrology, Dialysis and Transplantation Unit, Advanced Research Center on Kidney Aging (A.R.K.A.), Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy
| |
Collapse
|
45
|
Benmelech S, Le T, McKay M, Nam J, Subramaniam K, Tellez D, Vlasak G, Mak M. Biophysical and biochemical aspects of immune cell-tumor microenvironment interactions. APL Bioeng 2024; 8:021502. [PMID: 38572312 PMCID: PMC10990568 DOI: 10.1063/5.0195244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Accepted: 03/19/2024] [Indexed: 04/05/2024] Open
Abstract
The tumor microenvironment (TME), composed of and influenced by a heterogeneous set of cancer cells and an extracellular matrix, plays a crucial role in cancer progression. The biophysical aspects of the TME (namely, its architecture and mechanics) regulate interactions and spatial distributions of cancer cells and immune cells. In this review, we discuss the factors of the TME-notably, the extracellular matrix, as well as tumor and stromal cells-that contribute to a pro-tumor, immunosuppressive response. We then discuss the ways in which cells of the innate and adaptive immune systems respond to tumors from both biochemical and biophysical perspectives, with increased focus on CD8+ and CD4+ T cells. Building upon this information, we turn to immune-based antitumor interventions-specifically, recent biophysical breakthroughs aimed at improving CAR-T cell therapy.
Collapse
Affiliation(s)
- Shoham Benmelech
- Department of Biomedical Engineering, Yale University, New Haven, Connecticut 06511, USA
| | - Thien Le
- Department of Biomedical Engineering, Yale University, New Haven, Connecticut 06511, USA
| | - Maggie McKay
- Department of Biomedical Engineering, Yale University, New Haven, Connecticut 06511, USA
| | - Jungmin Nam
- Department of Biomedical Engineering, Yale University, New Haven, Connecticut 06511, USA
| | - Krupakar Subramaniam
- Department of Molecular, Cellular, and Developmental Biology, Yale University, New Haven, Connecticut 06511, USA
| | - Daniela Tellez
- Department of Biomedical Engineering, Yale University, New Haven, Connecticut 06511, USA
| | - Grace Vlasak
- Department of Biomedical Engineering, Yale University, New Haven, Connecticut 06511, USA
| | - Michael Mak
- Department of Biomedical Engineering, Yale University, New Haven, Connecticut 06511, USA
| |
Collapse
|
46
|
Rodi M, de Lastic AL, Panagoulias I, Aggeletopoulou I, Kelaidonis K, Matsoukas J, Apostolopoulos V, Mouzaki A. Myelin Oligodendrocyte Glycoprotein (MOG)35-55 Mannan Conjugate Induces Human T-Cell Tolerance and Can Be Used as a Personalized Therapy for Multiple Sclerosis. Int J Mol Sci 2024; 25:6092. [PMID: 38892275 PMCID: PMC11172913 DOI: 10.3390/ijms25116092] [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: 04/24/2024] [Revised: 05/29/2024] [Accepted: 05/30/2024] [Indexed: 06/21/2024] Open
Abstract
We have previously performed preclinical studies with the oxidized mannan-conjugated peptide MOG35-55 (OM-MOG35-55) in vivo (EAE mouse model) and in vitro (human peripheral blood) and demonstrated that OM-MOG35-55 suppresses antigen-specific T cell responses associated with autoimmune demyelination. Based on these results, we developed different types of dendritic cells (DCs) from the peripheral blood monocytes of patients with multiple sclerosis (MS) or healthy controls presenting OM-MOG35-55 or MOG-35-55 to autologous T cells to investigate the tolerogenic potential of OM-MOG35-55 for its possible use in MS therapy. To this end, monocytes were differentiated into different DC types in the presence of IL-4+GM-CSF ± dexamethasone (DEXA) ± vitamin D3 (VITD3). At the end of their differentiation, the DCs were loaded with peptides and co-cultured with T cells +IL-2 for 4 antigen presentation cycles. The phenotypes of the DC and T cell populations were analyzed using flow cytometry and the secreted cytokines using flow cytometry or ELISA. On day 8, the monocytes had converted into DCs expressing the typical markers of mature or immature phenotypes. Co-culture of T cells with all DC types for 4 antigen presentation cycles resulted in an increase in memory CD4+ T cells compared to memory CD8+ T cells and a suppressive shift in secreted cytokines, mainly due to increased TGF-β1 levels. The best tolerogenic effect was obtained when patient CD4+ T cells were co-cultured with VITD3-DCs presenting OM-MOG35-55, resulting in the highest levels of CD4+PD-1+ T cells and CD4+CD25+Foxp3+ Τ cells. In conclusion, the tolerance induction protocols presented in this work demonstrate that OM-MOG35-55 could form the basis for the development of personalized therapeutic vaccines or immunomodulatory treatments for MS.
Collapse
Affiliation(s)
- Maria Rodi
- Laboratory of Immunohematology, Medical School, University of Patras, 26500 Patras, Greece; (M.R.); (A.-L.d.L.); (I.P.); (I.A.)
| | - Anne-Lise de Lastic
- Laboratory of Immunohematology, Medical School, University of Patras, 26500 Patras, Greece; (M.R.); (A.-L.d.L.); (I.P.); (I.A.)
| | - Ioannis Panagoulias
- Laboratory of Immunohematology, Medical School, University of Patras, 26500 Patras, Greece; (M.R.); (A.-L.d.L.); (I.P.); (I.A.)
| | - Ioanna Aggeletopoulou
- Laboratory of Immunohematology, Medical School, University of Patras, 26500 Patras, Greece; (M.R.); (A.-L.d.L.); (I.P.); (I.A.)
| | - Kostas Kelaidonis
- NewDrug P.C., Patras Science Park, 26504 Patras, Greece; (K.K.); (J.M.)
| | - John Matsoukas
- NewDrug P.C., Patras Science Park, 26504 Patras, Greece; (K.K.); (J.M.)
- Immunology and Translational Research, Institute for Health and Sport, Victoria University, Melbourne, VIC 3030, Australia;
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N1N4, Canada
- Department of Chemistry, University of Patras, 26504 Patras, Greece
| | - Vasso Apostolopoulos
- Immunology and Translational Research, Institute for Health and Sport, Victoria University, Melbourne, VIC 3030, Australia;
- Immunology Program, Australian Institute for Musculoskeletal Science (AIMSS), Melbourne, VIC 3021, Australia
| | - Athanasia Mouzaki
- Laboratory of Immunohematology, Medical School, University of Patras, 26500 Patras, Greece; (M.R.); (A.-L.d.L.); (I.P.); (I.A.)
| |
Collapse
|
47
|
Obare LM, Temu T, Mallal SA, Wanjalla CN. Inflammation in HIV and Its Impact on Atherosclerotic Cardiovascular Disease. Circ Res 2024; 134:1515-1545. [PMID: 38781301 PMCID: PMC11122788 DOI: 10.1161/circresaha.124.323891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/25/2024]
Abstract
People living with HIV have a 1.5- to 2-fold increased risk of developing cardiovascular disease. Despite treatment with highly effective antiretroviral therapy, people living with HIV have chronic inflammation that makes them susceptible to multiple comorbidities. Several factors, including the HIV reservoir, coinfections, clonal hematopoiesis of indeterminate potential (CHIP), microbial translocation, and antiretroviral therapy, may contribute to the chronic state of inflammation. Within the innate immune system, macrophages harbor latent HIV and are among the prominent immune cells present in atheroma during the progression of atherosclerosis. They secrete inflammatory cytokines such as IL (interleukin)-6 and tumor necrosis-α that stimulate the expression of adhesion molecules on the endothelium. This leads to the recruitment of other immune cells, including cluster of differentiation (CD)8+ and CD4+ T cells, also present in early and late atheroma. As such, cells of the innate and adaptive immune systems contribute to both systemic inflammation and vascular inflammation. On a molecular level, HIV-1 primes the NLRP3 (NLR family pyrin domain containing 3) inflammasome, leading to an increased expression of IL-1β, which is important for cardiovascular outcomes. Moreover, activation of TLRs (toll-like receptors) by HIV, gut microbes, and substance abuse further activates the NLRP3 inflammasome pathway. Finally, HIV proteins such as Nef (negative regulatory factor) can inhibit cholesterol efflux in monocytes and macrophages through direct action on the cholesterol transporter ABCA1 (ATP-binding cassette transporter A1), which promotes the formation of foam cells and the progression of atherosclerotic plaque. Here, we summarize the stages of atherosclerosis in the context of HIV, highlighting the effects of HIV, coinfections, and antiretroviral therapy on cells of the innate and adaptive immune system and describe current and future interventions to reduce residual inflammation and improve cardiovascular outcomes among people living with HIV.
Collapse
Affiliation(s)
- Laventa M. Obare
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN (L.M.O., S.A.M., C.N.W.)
| | - Tecla Temu
- Department of Pathology, Harvard Medical School, Boston, MA (T.T.)
| | - Simon A. Mallal
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN (L.M.O., S.A.M., C.N.W.)
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN (S.A.M.)
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN (S.A.M.)
- Institute for Immunology and Infectious Diseases, Murdoch University, WA, Western Australia (S.A.M.)
| | - Celestine N. Wanjalla
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN (L.M.O., S.A.M., C.N.W.)
| |
Collapse
|
48
|
Li X, Gao J, Wu C, Wang C, Zhang R, He J, Xia ZJ, Joshi N, Karp JM, Kuai R. Precise modulation and use of reactive oxygen species for immunotherapy. SCIENCE ADVANCES 2024; 10:eadl0479. [PMID: 38748805 PMCID: PMC11095489 DOI: 10.1126/sciadv.adl0479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 04/10/2024] [Indexed: 05/19/2024]
Abstract
Reactive oxygen species (ROS) play an important role in regulating the immune system by affecting pathogens, cancer cells, and immune cells. Recent advances in biomaterials have leveraged this mechanism to precisely modulate ROS levels in target tissues for improving the effectiveness of immunotherapies in infectious diseases, cancer, and autoimmune diseases. Moreover, ROS-responsive biomaterials can trigger the release of immunotherapeutics and provide tunable release kinetics, which can further boost their efficacy. This review will discuss the latest biomaterial-based approaches for both precise modulation of ROS levels and using ROS as a stimulus to control the release kinetics of immunotherapeutics. Finally, we will discuss the existing challenges and potential solutions for clinical translation of ROS-modulating and ROS-responsive approaches for immunotherapy, and provide an outlook for future research.
Collapse
Affiliation(s)
- Xinyan Li
- School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, China
- Tsinghua-Peking Center for Life Sciences, Beijing 100084, China
| | - Jingjing Gao
- Department of Anesthesiology, Perioperative, and Pain Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- Department of Biomedical Engineering, Material Science and Engineering Graduate Program and The Center for Bioactive Delivery-Institute for Applied Life Sciences, University of Massachusetts, Amherst, MA 01003, USA
| | - Chengcheng Wu
- School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, China
- Tsinghua-Peking Center for Life Sciences, Beijing 100084, China
| | - Chaoyu Wang
- School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, China
- Tsinghua-Peking Center for Life Sciences, Beijing 100084, China
| | - Ruoshi Zhang
- School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, China
- Tsinghua-Peking Center for Life Sciences, Beijing 100084, China
| | - Jia He
- School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, China
- Tsinghua-Peking Center for Life Sciences, Beijing 100084, China
| | - Ziting Judy Xia
- Department of Anesthesiology, Perioperative, and Pain Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Nitin Joshi
- Department of Anesthesiology, Perioperative, and Pain Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Jeffrey M. Karp
- Department of Anesthesiology, Perioperative, and Pain Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Rui Kuai
- School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, China
- Tsinghua-Peking Center for Life Sciences, Beijing 100084, China
| |
Collapse
|
49
|
Su G, Chen Y, Li X, Shao JW. Virus versus host: influenza A virus circumvents the immune responses. Front Microbiol 2024; 15:1394510. [PMID: 38817972 PMCID: PMC11137263 DOI: 10.3389/fmicb.2024.1394510] [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: 03/01/2024] [Accepted: 04/23/2024] [Indexed: 06/01/2024] Open
Abstract
Influenza A virus (IAV) is a highly contagious pathogen causing dreadful losses to humans and animals around the globe. As is known, immune escape is a strategy that benefits the proliferation of IAVs by antagonizing, blocking, and suppressing immune surveillance. The HA protein binds to the sialic acid (SA) receptor to enter the cytoplasm and initiate viral infection. The conserved components of the viral genome produced during replication, known as the pathogen-associated molecular patterns (PAMPs), are thought to be critical factors for the activation of effective innate immunity by triggering dependent signaling pathways after recognition by pattern recognition receptors (PRRs), followed by a cascade of adaptive immunity. Viral infection-induced immune responses establish an antiviral state in the host to effectively inhibit virus replication and enhance viral clearance. However, IAV has evolved multiple mechanisms that allow it to synthesize and transport viral components by "playing games" with the host. At its heart, this review will describe how host and viral factors interact to facilitate the viral evasion of host immune responses.
Collapse
Affiliation(s)
- Guanming Su
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, Guangzhou, China
| | - Yiqun Chen
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Xiaowen Li
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou, China
| | - Jian-Wei Shao
- School of Life Science and Engineering, Foshan University, Foshan, China
| |
Collapse
|
50
|
Zhang M, Xu W, Yang N, Li Z, Zhou S, Liu X, Wang J, Li H. PCV2 Induced Endothelial Derived IL-8 Affects MoDCs Maturation Mainly via NF-κB Signaling Pathway. Viruses 2024; 16:646. [PMID: 38675986 PMCID: PMC11053600 DOI: 10.3390/v16040646] [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: 02/05/2024] [Revised: 04/16/2024] [Accepted: 04/17/2024] [Indexed: 04/28/2024] Open
Abstract
Porcine circovirus type 2 (PCV2) infection can cause immunosuppressive diseases in pigs. Vascular endothelial cells (VECs), as the target cells for PCV2, play an important role in the immune response and inflammatory regulation. Endothelial IL-8, which is produced by porcine hip artery endothelial cells (PIECs) infected with PCV2, can inhibit the maturation of monocyte-derived dendritic cells (MoDCs). Here, we established a co-culture system of MoDCs and different groups of PIECs to further investigate the PCV2-induced endothelial IL-8 signaling pathway that drives the inhibition of MoDC maturation. The differentially expressed genes related to MoDC maturation were mainly enriched in the NF-κB and JAK2-STAT3 signaling pathways. Both the NF-κB related factor RELA and JAK2-STAT3 signaling pathway related factors (IL2RA, JAK, STAT2, STAT5, IL23A, IL7, etc.) decreased significantly in the IL-8 up-regulated group, and increased significantly in the down-regulated group. The expression of NF-κB p65 in the IL-8 up-regulated group was reduced significantly, and the expression of IκBα was increased significantly. Nuclear translocation of NF-κB p65 was inhibited, while the nuclear translocation of p-STAT3 was increased in MoDCs in the PCV2-induced endothelial IL-8 group. The results of treatment with NF-κB signaling pathway inhibitors showed that the maturation of MoDCs was inhibited and the expression of IL-12 and GM-CSF at mRNA level were lower. Inhibition of the JAK2-STAT3 signaling pathway had no significant effect on maturation, and the expression of IL-12 and GM-CSF at mRNA level produced no significant change. In summary, the NF-κB signaling pathway is the main signaling pathway of MoDC maturation, and is inhibited by the PCV2-induced up-regulation of endothelial-derived IL-8.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Jianfang Wang
- College of Animal Science and Technology, Beijing University of Agriculture, No. 7 Beinong Road, Beijing 102206, China; (M.Z.); (W.X.); (N.Y.); (Z.L.); (S.Z.); (X.L.)
| | - Huanrong Li
- College of Animal Science and Technology, Beijing University of Agriculture, No. 7 Beinong Road, Beijing 102206, China; (M.Z.); (W.X.); (N.Y.); (Z.L.); (S.Z.); (X.L.)
| |
Collapse
|