1
|
Lee J, McClure S, Weichselbaum RR, Mimee M. Designing live bacterial therapeutics for cancer. Adv Drug Deliv Rev 2025; 221:115579. [PMID: 40228606 PMCID: PMC12067981 DOI: 10.1016/j.addr.2025.115579] [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: 01/31/2025] [Revised: 03/26/2025] [Accepted: 04/09/2025] [Indexed: 04/16/2025]
Abstract
Humans are home to a diverse community of bacteria, many of which form symbiotic relationships with their host. Notably, tumors can also harbor their own unique bacterial populations that can influence tumor growth and progression. These bacteria, which selectively colonize hypoxic and acidic tumor microenvironments, present a novel therapeutic strategy to combat cancer. Advancements in synthetic biology enable us to safely and efficiently program therapeutic drug production in bacteria, further enhancing their potential. This review provides a comprehensive guide to utilizing bacteria for cancer treatment. We discuss key considerations for selecting bacterial strains, emphasizing their colonization efficiency, the delicate balance between safety and anti-tumor efficacy, and the availability of tools for genetic engineering. We also delve into strategies for precise spatiotemporal control of drug delivery to minimize adverse effects and maximize therapeutic impact, exploring recent examples of engineered bacteria designed to combat tumors. Finally, we address the underlying challenges and future prospects of bacterial cancer therapy. This review underscores the versatility of bacterial therapies and outlines strategies to fully harness their potential in the fight against cancer.
Collapse
Affiliation(s)
- Jaehyun Lee
- Department of Microbiology, University of Chicago, Chicago, IL 60637, USA
| | - Sandra McClure
- Department of Microbiology, University of Chicago, Chicago, IL 60637, USA; Duchoissois Family Institute, University of Chicago, Chicago, IL 60637, USA; Committee On Molecular Metabolism and Nutrition, University of Chicago, Chicago, IL 60637, USA
| | - Ralph R Weichselbaum
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago 60637, USA; The Ludwig Center for Metastasis Research, University of Chicago, Chicago 60637, USA
| | - Mark Mimee
- Department of Microbiology, University of Chicago, Chicago, IL 60637, USA; Duchoissois Family Institute, University of Chicago, Chicago, IL 60637, USA; Committee On Molecular Metabolism and Nutrition, University of Chicago, Chicago, IL 60637, USA; Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL 60637, USA.
| |
Collapse
|
2
|
Kumar RR, Agarwal N, Shree A, Gorain JK, Rahul E, Ganguly S, Bakhshi S, Sharma U. Decoding the immune landscape in Ewing sarcoma pathogenesis: The role of tumor infiltrating immune cells and immune milieu. J Bone Oncol 2025; 52:100678. [PMID: 40242222 PMCID: PMC12002756 DOI: 10.1016/j.jbo.2025.100678] [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: 02/03/2025] [Revised: 03/24/2025] [Accepted: 03/24/2025] [Indexed: 04/18/2025] Open
Abstract
Ewing sarcoma (EwS) is the second most prevalent pediatric bone malignancy, characterized by its aggressive behavior and unfavorable prognosis. The tumor microenvironment (TME) of EwS is shaped by immunosuppressive components, including myeloid-derived suppressor cells, tumor-associated macrophages, and immune checkpoint molecules such as PD-1/PD-L1 and HLA-G. These elements impair anti-tumor immune responses by modulating the function of tumor-infiltrating immune cells, such as regulatory T cells (Tregs), CD8+ T cells, and natural killer cells. Chemokines, including CXCL9 and CXCL12, and cytokines, such as transforming growth factor-beta and interleukin-10, further contribute to immune suppression and promote metastatic dissemination. Recent advances in immunotherapy have highlighted the therapeutic potential of modulating immune cells and signaling pathways to enhance anti-tumor immunity. This review provides a comprehensive analysis of the complex immune landscape within the EwS TME, focusing on the mechanistic roles of key immune components and their potential as therapeutic targets. Understanding these interactions could pave the way for innovative treatment strategies to improve clinical outcomes in patients with EwS.
Collapse
Affiliation(s)
- Rajiv Ranjan Kumar
- Department of Medical Oncology, Dr. B.R.A. Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, New Delhi, India
| | - Nikita Agarwal
- Department of Medical Oncology, Dr. B.R.A. Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, New Delhi, India
| | - Akshi Shree
- Department of Medical Oncology, Dr. B.R.A. Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, New Delhi, India
- Department of Biomedical Science, Shaheed Rajguru College of Applied Sciences for Women, University of Delhi, Delhi 110096, India
| | - Jaya Kanta Gorain
- Department of Medical Oncology, Dr. B.R.A. Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, New Delhi, India
| | - Ekta Rahul
- Department of Pathology, Atal Bihari Vajpayee Institute of Medical Sciences and Dr. Ram Manohar Lohia Hospital, Delhi 110001, India
| | - Shuvadeep Ganguly
- Department of Medical Oncology, Jawaharlal Institute of Postgraduate Medical Education & Research, Puducherry, India
| | - Sameer Bakhshi
- Department of Medical Oncology, Dr. B.R.A. Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, New Delhi, India
| | - Uttam Sharma
- Department of Medical Oncology, Dr. B.R.A. Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
3
|
Islam MD, Islam MM, Inoue A, Yesmin S, Brindha S, Yoshizue T, Tsurui H, Kurosu T, Kuroda Y. Neutralizing antibodies against the Japanese encephalitis virus are produced by a 12 kDa E. coli- expressed envelope protein domain III (EDIII) tagged with a solubility-controlling peptide. Vaccine 2025; 56:127143. [PMID: 40267616 DOI: 10.1016/j.vaccine.2025.127143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Revised: 04/01/2025] [Accepted: 04/13/2025] [Indexed: 04/25/2025]
Abstract
Escherichia coli is a powerful and cost-effective platform for producing recombinant proteins. However, E. coli- produced proteins lack side-chain glycosylation and may be misfolded due to non-native disulfide bonds, often leading to poor immunogenicity. As a result, they are commonly perceived as unsuitable for use as antiviral vaccine antigens. This study addresses this challenge using the small 12 kDa envelope protein domain III of the Japanese encephalitis virus (JEV-EDIII) as a model. We demonstrate that the low immunogenicity of E. coli- produced proteins can be effectively overcome by employing a solubility-controlling peptide tag (SCP-tag) composed of five isoleucines (C5I). E. coli-produced JEV-EDIII oligomerized into 100 nm (Rh) soluble oligomers upon attachment of the C5I-tag, whereas the untagged JEV-EDIII remained monomeric (Rh ∼ 1.9 nm). The C5I-tag significantly enhanced anti-JEV EDIII IgG titers, as evidenced by ELISA, and increased the population of memory T cells in the spleen, as assessed by flow cytometry. Most notably, the C5I-tagged JEV-EDIII elicited neutralizing antibodies, confirmed by the FRNT50 neutralization assay using live JEV. These findings suggest that oligomerization via SCP-tagging offers a promising, adjuvant-free approach for producing neutralizing antibodies with long-term T cell memory, paving the way for developing E. coli- produced, protein domain-based vaccines.
Collapse
Affiliation(s)
- Md Din Islam
- Department of Biotechnology and Life Science, Faculty of Engineering, Tokyo University of Agriculture and Technology, Nakamachi 2-24-16, Tokyo 184-8588, Japan
| | - M Monirul Islam
- Institute of Global Innovation Research, Tokyo University of Agriculture and Technology, 3-8-1 Harumi-cho, Fuchu-shi, Tokyo 183-8538, Japan; Department of Biochemistry and Molecular Biology, Faculty of Biological Sciences, University of Chittagong, Chittagong 4331, Bangladesh
| | - Ayae Inoue
- Department of Biotechnology and Life Science, Faculty of Engineering, Tokyo University of Agriculture and Technology, Nakamachi 2-24-16, Tokyo 184-8588, Japan
| | - Sanjida Yesmin
- Department of Biotechnology and Life Science, Faculty of Engineering, Tokyo University of Agriculture and Technology, Nakamachi 2-24-16, Tokyo 184-8588, Japan
| | - Subbaian Brindha
- Department of Biotechnology and Life Science, Faculty of Engineering, Tokyo University of Agriculture and Technology, Nakamachi 2-24-16, Tokyo 184-8588, Japan; Institute of Global Innovation Research, Tokyo University of Agriculture and Technology, 3-8-1 Harumi-cho, Fuchu-shi, Tokyo 183-8538, Japan
| | - Takahiro Yoshizue
- Department of Biotechnology and Life Science, Faculty of Engineering, Tokyo University of Agriculture and Technology, Nakamachi 2-24-16, Tokyo 184-8588, Japan
| | - Hiromichi Tsurui
- Department of Biotechnology and Life Science, Faculty of Engineering, Tokyo University of Agriculture and Technology, Nakamachi 2-24-16, Tokyo 184-8588, Japan; Department of Immunological Diagnosis, Juntendo University School of Medicine, Hongo 2-1-1, Tokyo 113-8421, Japan
| | - Takeshi Kurosu
- Department of Virology I, National Institute of Infectious Diseases, Musashimurayama, Gakuen 4-7-1, Tokyo 208-0011, Japan
| | - Yutaka Kuroda
- Department of Biotechnology and Life Science, Faculty of Engineering, Tokyo University of Agriculture and Technology, Nakamachi 2-24-16, Tokyo 184-8588, Japan; Institute of Global Innovation Research, Tokyo University of Agriculture and Technology, 3-8-1 Harumi-cho, Fuchu-shi, Tokyo 183-8538, Japan.
| |
Collapse
|
4
|
Zhu Z, Lou G, Luo Y, Yihunie K, Hoar J, Daniel JA, Evers BM, Yao C, Wu T. Aging Compromises Terminal Differentiation Program of Cytotoxic Effector Lineage and Promotes Exhaustion in CD8 + T Cells Responding to Coronavirus Infection. Aging Cell 2025:e70109. [PMID: 40396260 DOI: 10.1111/acel.70109] [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: 12/26/2024] [Revised: 04/16/2025] [Accepted: 05/07/2025] [Indexed: 05/22/2025] Open
Abstract
T cell aging increases the risk of viral infection-related morbidity and mortality and reduces vaccine efficacy in the elderly. A major hallmark of T cell aging is the loss of quiescence and shift toward terminal differentiation during homeostasis. However, how aging impacts the differentiation program of virus-specific T cells during infection is unclear. Here, in a murine coronavirus (MHV) infection model with age-associated increased mortality, we demonstrate that aging impairs, instead of promoting, the terminal differentiation program of virus-specific CD8+ T cells. Upon infection, CD8+ and CD4+ T cells in old mice showed marked reduction in clonal expansion and upregulation of immune checkpoints associated with T cell exhaustion. Bulk and single-cell transcriptomics showed that aging upregulated the T cell exhaustion transcriptional program associated with TOX in virus-specific CD8+ T cells and shifted the myeloid compartment from immunostimulatory to immunosuppressive phenotype. In addition, aging downregulated the transcriptional program of terminally differentiated effector CD8+ T cells and diminished the CX3CR1+ cytotoxic effector lineage. Mechanistically, virus-specific CD8+ T cells from infected aged mice displayed defects in inducing transcription factors ZEB2 and KLF2, which were required for terminal differentiation of effector CD8+ T cells. Together, our study shows that aging impairs terminal differentiation and promotes exhaustion of virus-specific CD8+ T cells responding to coronavirus infection through dysregulating expression of lineage-defining transcription factors.
Collapse
Affiliation(s)
- Ziang Zhu
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Immunology Ph.D. Program, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Guohua Lou
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Ying Luo
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Kiddist Yihunie
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Cancer Biology Ph.D. Program, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Jonathan Hoar
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Juan A Daniel
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Bret M Evers
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Chen Yao
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Kidney Cancer Program, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Tuoqi Wu
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Peter O'Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
5
|
Lang HP, Almeer FF, Jenkins MK, Friedenberg SG. Rabies vaccination induces a CD4+ TEM and CD4+CD8+ TEMRA TH1 phenotype in dogs. PLoS One 2025; 20:e0323823. [PMID: 40354406 PMCID: PMC12068608 DOI: 10.1371/journal.pone.0323823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Accepted: 04/15/2025] [Indexed: 05/14/2025] Open
Abstract
The canine rabies vaccine consists of the whole killed rabies virus and an alum adjuvant. While it is known to provide immunological protection in dogs, its effects on cell-mediated responses remain largely uncharacterized. Here, we analyzed blood and spleen samples from vaccinated dogs to understand adaptive immune responses ex vivo following restimulation with rabies vaccine antigens. Our results showed that recombinant rabies virus glycoprotein (RABV-G) elicited higher antibody titers and IFNγ production compared to recombinant rabies virus nucleoprotein (RABV-N). CD4+ and CD4+CD8+ double-positive (DP) T cells proliferate robustly after five days of RABV-G stimulation, which was inhibited by an anti-canine MHC class II blocking antibody. Both RABV-G-specific CD4+ and DP T cells demonstrated a polarized TH1 phenotype, with minor subsets showing TH1/TH17 hybrid and pathogenic TH1/TH17 hybrid cell features. CD4+ T cells were primarily effector memory T cells (TEM), while DP T cells exhibited a terminally differentiated effector memory phenotype that re-expressed CD45RA (TEMRA). Both RABV-G-specific CD4+ and DP T cells were detectable up to 1,024 days post-vaccination in spleen samples and their proliferative capacities were unaffected by age. Our results provide the first characterization of canine RABV-G-specific T cell phenotypes in the spleen and blood following rabies vaccination.
Collapse
Affiliation(s)
- Haeree P. Lang
- Center for Immunology, University of Minnesota, Minneapolis, Minnesota, United States of America
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St. Paul, Minnesota, United States of America
| | - Farah F. Almeer
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St. Paul, Minnesota, United States of America
| | - Marc K. Jenkins
- Center for Immunology, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Steven G. Friedenberg
- Center for Immunology, University of Minnesota, Minneapolis, Minnesota, United States of America
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St. Paul, Minnesota, United States of America
| |
Collapse
|
6
|
Niekamp P, Kim RH, Jayaraman A, Klement N, Kostlan R, Kim CH. The Nuclear Receptor NR1B1/RARα Arrests the Differentiation of Anti-Tumor Effector Cytotoxic T Cells. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2410241. [PMID: 40068101 PMCID: PMC12061256 DOI: 10.1002/advs.202410241] [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] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Revised: 02/26/2025] [Indexed: 05/10/2025]
Abstract
NR1B1/RARα expression is dynamically regulated in cytotoxic lymphocytes (CTLs) in tumors, but the importance of its expression in anti-tumor CTLs remains unknown. RARα gene expression is upregulated in CTLs in tumor microenvironments (TME), but its protein expression is downregulated by retinoic acid. The role of RARα expression in regulating anti-tumor effector CTL (Teff) differentiation is reported. Mice that over-express RARα in T cells are defective in early Teff differentiation and fail to populate tumors. In contrast, RARα-deficient CTLs are hyper-active in making tumor-populating Teff cells, suggesting that RARα represses Teff differentiation. Moreover, RARα negatively controls the trafficking receptor switch from the lymphoid to an effector type. Generation of chimeric antigen receptor (CAR) T cells with reduced RARα expression produces highly effective CAR T cells with enhanced anti-tumor cytotoxicity. Mechanistically, upregulated RARα expression decreases the nuclear histone acetylase (HAT) activity, required for TCF1 to BATF transcription factor and trafficking switches during Teff differentiation. Additionally, RARα and BATF closely associate with each other on Teff-associated genes on the chromatin for possible cross-regulation. In sum, T cell-expressed RARα is identified as a novel negative regulator and potential target of intervention in promoting anti-cancer T cell immunity.
Collapse
Affiliation(s)
- Patrick Niekamp
- Department of PathologyUniversity of Michigan School of MedicineAnn ArborMI48109USA
- Mary H. Weiser Food Allergy CenterUniversity of Michigan School of MedicineAnn ArborMI48109USA
| | - Ryun Hee Kim
- Department of PathologyUniversity of Michigan School of MedicineAnn ArborMI48109USA
- Mary H. Weiser Food Allergy CenterUniversity of Michigan School of MedicineAnn ArborMI48109USA
| | - Adithyan Jayaraman
- Department of PathologyUniversity of Michigan School of MedicineAnn ArborMI48109USA
- Mary H. Weiser Food Allergy CenterUniversity of Michigan School of MedicineAnn ArborMI48109USA
| | - Nils Klement
- University of BielefeldFaculty of Physics33615BielefeldGermany
| | - Raymond Kostlan
- Department of PathologyUniversity of Michigan School of MedicineAnn ArborMI48109USA
| | - Chang H. Kim
- Department of PathologyUniversity of Michigan School of MedicineAnn ArborMI48109USA
- Mary H. Weiser Food Allergy CenterUniversity of Michigan School of MedicineAnn ArborMI48109USA
- Immunology Graduate ProgramUniversity of MichiganAnn ArborMI48109USA
- Rogel Cancer CenterUniversity of Michigan School of MedicineAnn ArborMI48109USA
| |
Collapse
|
7
|
Berton RR, Heidarian M, Kannan SK, Shah M, Butler NS, Harty JT, Badovinac VP. Accurate enumeration of pathogen-specific and virtual memory CD8 T cells after infection. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2025; 214:995-1007. [PMID: 40167212 PMCID: PMC12123210 DOI: 10.1093/jimmun/vkaf007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 01/04/2025] [Indexed: 04/02/2025]
Abstract
Establishing the magnitude and kinetics of polyclonal Ag-specific CD8 T-cell responses, in addition to their functional fitness, is critical for evaluating a host's ability to respond to different kinds of infections and/or immunizations. To track CD8 T-cell responses during infection, a surrogate-activation-marker approach (CD8αloCD11ahi) is used to distinguish naïve and Ag-experienced effector/memory CD8 T cells in vivo. However, semidifferentiated virtual memory (Tvm) CD8 T cells have recently been identified in uninfected/unmanipulated mice that display a phenotype similar to Ag-experienced cells. Therefore, magnitude and breadth of CD8 T-cell responses may be overestimated when responses are profiled using only CD8α/CD11a markers. Thus, to precisely define and distinguish Tvm from pathogen-specific CD8 T cells during bacterial, parasitic, and viral infections, pathogen-specific sensor TCR-Tg cells were adoptively transferred prior to challenge. We demonstrate that Tvm CD8 T cells are found in CD8αloCD11ahi-defined Ag-experienced CD8 T cells but can be parsed out in infected host with their CD49d-CD44hiCD122hi expression pattern. However, this approach presents potential limitations as CD49d+ Ag-specific CD8 T cells can lose CD49d expression and adopt a Tvm-like phenotype depending on their Ag-stimulation history, age, and naïve CD8 T-cell precursor frequency before the infection. Importantly, Tvm cells contribute to the breadth of the CD8 T-cell response, and their contribution depends on type of infection, time after infection, and tissue examined. Thus, these data define limitations in our ability to resolve between pathogen/Ag-specific and Tvm CD8 T-cell responses during infection, a notion of direct relevance for experimental murine studies designed to follow CD8 T-cell responses in vivo.
Collapse
Affiliation(s)
- Roger R Berton
- Interdisciplinary Graduate Program in Immunology, University of Iowa, 500 Newton Rd, 1020 ML, Iowa City, IA 52242, USA
- Department of Pathology, University of Iowa, 500 Newton Rd, 1020 ML, Iowa City, IA 52242, USA
| | - Mohammad Heidarian
- Department of Pathology, University of Iowa, 500 Newton Rd, 1020 ML, Iowa City, IA 52242, USA
- Department of Pathology Graduate Programs, University of Iowa, 500 Newton Rd, 1020 ML, Iowa City, IA 52242, USA
| | - Shravan Kumar Kannan
- Interdisciplinary Graduate Program in Immunology, University of Iowa, 500 Newton Rd, 1020 ML, Iowa City, IA 52242, USA
- Department of Pathology, University of Iowa, 500 Newton Rd, 1020 ML, Iowa City, IA 52242, USA
| | - Manan Shah
- Interdisciplinary Graduate Program in Immunology, University of Iowa, 500 Newton Rd, 1020 ML, Iowa City, IA 52242, USA
- Department of Microbiology and Immunology, University of Iowa, 500 Newton Rd, 1020 ML, Iowa City, IA 52242, USA
| | - Noah S Butler
- Interdisciplinary Graduate Program in Immunology, University of Iowa, 500 Newton Rd, 1020 ML, Iowa City, IA 52242, USA
- Department of Microbiology and Immunology, University of Iowa, 500 Newton Rd, 1020 ML, Iowa City, IA 52242, USA
| | - John T Harty
- Interdisciplinary Graduate Program in Immunology, University of Iowa, 500 Newton Rd, 1020 ML, Iowa City, IA 52242, USA
- Department of Pathology, University of Iowa, 500 Newton Rd, 1020 ML, Iowa City, IA 52242, USA
- Department of Pathology Graduate Programs, University of Iowa, 500 Newton Rd, 1020 ML, Iowa City, IA 52242, USA
| | - Vladimir P Badovinac
- Interdisciplinary Graduate Program in Immunology, University of Iowa, 500 Newton Rd, 1020 ML, Iowa City, IA 52242, USA
- Department of Pathology, University of Iowa, 500 Newton Rd, 1020 ML, Iowa City, IA 52242, USA
- Department of Pathology Graduate Programs, University of Iowa, 500 Newton Rd, 1020 ML, Iowa City, IA 52242, USA
| |
Collapse
|
8
|
Ghazizadeh Y, Salehi Shadkami H, Madani F, Niknam S, Adabi M. Advances in cancer nanovaccines: a focus on colorectal cancer. Nanomedicine (Lond) 2025; 20:1029-1041. [PMID: 40186876 PMCID: PMC12051617 DOI: 10.1080/17435889.2025.2486930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Accepted: 03/27/2025] [Indexed: 04/07/2025] Open
Abstract
Nanotechnology has revolutionized cancer treatment by providing innovative solutions through nanocancer therapies, nanovaccines, and nanoparticles. This review focuses on the application of these technologies in colorectal cancer (CRC), highlighting their progression from preclinical studies to clinical trials. Nanoparticles, including liposomes, silica, gold, and lipid nanoparticles, possess unique properties that enhance drug delivery, improve therapeutic efficacy, and minimize systemic toxicity. Additionally, nanovaccines are being developed to elicit robust immune responses against CRC cells. This paper offers a comprehensive overview of the current state of nanotechnology-based treatments for CRC, emphasizing key preclinical studies and clinical trials that demonstrate their potential. Furthermore, the review discusses the challenges faced in this field. It outlines future directions for research, underscoring the need for ongoing efforts to translate these promising technologies into practical clinical applications.
Collapse
Affiliation(s)
- Yalda Ghazizadeh
- Student Research Committee, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Nanomedicine Student Association (NMA), Student’s Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Hossein Salehi Shadkami
- Nanomedicine Student Association (NMA), Student’s Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran
- Department of Medical Science, Tehran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Madani
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Sedigheh Niknam
- Institute of Nano Science and Nano Technology, University of Kashan, Kashan, Iran
| | - Mahdi Adabi
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Food Microbiology Research Center, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
9
|
Sutanto H, Pradana FR, Adytia GJ, Ansharullah BA, Waitupu A, Bramantono B, Fetarayani D. Memory T Cells in Respiratory Virus Infections: Protective Potential and Persistent Vulnerabilities. Med Sci (Basel) 2025; 13:48. [PMID: 40407543 PMCID: PMC12101432 DOI: 10.3390/medsci13020048] [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: 03/13/2025] [Revised: 04/19/2025] [Accepted: 04/25/2025] [Indexed: 05/26/2025] Open
Abstract
Respiratory virus infections, such as those caused by influenza viruses, respiratory syncytial virus (RSV), and coronaviruses, pose a significant global health burden. While the immune system's adaptive components, including memory T cells, are critical for recognizing and combating these pathogens, recurrent infections and variable disease outcomes persist. Memory T cells are a key element of long-term immunity, capable of responding swiftly upon re-exposure to pathogens. They play diverse roles, including cross-reactivity to conserved viral epitopes and modulation of inflammatory responses. However, the protective efficacy of these cells is influenced by several factors, including viral evolution, host age, and immune system dynamics. This review explores the dichotomy of memory T cells in respiratory virus infections: their potential to confer robust protection and the limitations that allow for breakthrough infections. Understanding the underlying mechanisms governing the formation, maintenance, and functional deployment of memory T cells in respiratory mucosa is critical for improving immunological interventions. We highlight recent advances in vaccine strategies aimed at bolstering T cell-mediated immunity and discuss the challenges posed by viral immune evasion. Addressing these gaps in knowledge is pivotal for designing effective therapeutics and vaccines to mitigate the global burden of respiratory viruses.
Collapse
Affiliation(s)
- Henry Sutanto
- Internal Medicine Study Program, Department of Internal Medicine, Faculty of Medicine, Universitas Airlangga, Surabaya 60132, Indonesia; (H.S.); (F.R.P.); (G.J.A.); (B.A.A.); (A.W.)
- Department of Internal Medicine, Dr. Soetomo General Academic Hospital, Surabaya 60286, Indonesia
| | - Febrian Ramadhan Pradana
- Internal Medicine Study Program, Department of Internal Medicine, Faculty of Medicine, Universitas Airlangga, Surabaya 60132, Indonesia; (H.S.); (F.R.P.); (G.J.A.); (B.A.A.); (A.W.)
- Department of Internal Medicine, Dr. Soetomo General Academic Hospital, Surabaya 60286, Indonesia
| | - Galih Januar Adytia
- Internal Medicine Study Program, Department of Internal Medicine, Faculty of Medicine, Universitas Airlangga, Surabaya 60132, Indonesia; (H.S.); (F.R.P.); (G.J.A.); (B.A.A.); (A.W.)
- Department of Internal Medicine, Dr. Soetomo General Academic Hospital, Surabaya 60286, Indonesia
| | - Bagus Aditya Ansharullah
- Internal Medicine Study Program, Department of Internal Medicine, Faculty of Medicine, Universitas Airlangga, Surabaya 60132, Indonesia; (H.S.); (F.R.P.); (G.J.A.); (B.A.A.); (A.W.)
- Department of Internal Medicine, Dr. Soetomo General Academic Hospital, Surabaya 60286, Indonesia
| | - Alief Waitupu
- Internal Medicine Study Program, Department of Internal Medicine, Faculty of Medicine, Universitas Airlangga, Surabaya 60132, Indonesia; (H.S.); (F.R.P.); (G.J.A.); (B.A.A.); (A.W.)
- Department of Internal Medicine, Dr. Soetomo General Academic Hospital, Surabaya 60286, Indonesia
| | - Bramantono Bramantono
- Department of Internal Medicine, Dr. Soetomo General Academic Hospital, Surabaya 60286, Indonesia
- Division of Tropical and Infectious Diseases, Department of Internal Medicine, Faculty of Medicine, Universitas Airlangga, Surabaya 60132, Indonesia
| | - Deasy Fetarayani
- Department of Internal Medicine, Dr. Soetomo General Academic Hospital, Surabaya 60286, Indonesia
- Division of Allergy and Clinical Immunology, Department of Internal Medicine, Faculty of Medicine, Universitas Airlangga, Surabaya 60132, Indonesia
| |
Collapse
|
10
|
Morán-Plata FJ, Muñoz-García N, Barrena S, Yeguas A, Balanzategui A, Carretero-Domínguez S, Pozo J, Lécrevisse Q, González-González M, Bárcena P, Alcoceba M, Herrero-García M, Solano F, López-Parra M, Martín García-Sancho A, de Sá Ferreira-Facio C, Villamor N, Lau C, Dos Anjos Teixeira M, Botafogo V, Orfao A, Almeida J. Maturation-Related and Functional-Associated Phenotypic Profile of Tumor T Cells in Mature/Peripheral T-Cell Neoplasms: Association With the Diagnostic Subtype of the Disease. J Transl Med 2025; 105:104180. [PMID: 40288651 DOI: 10.1016/j.labinv.2025.104180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2024] [Revised: 03/20/2025] [Accepted: 04/17/2025] [Indexed: 04/29/2025] Open
Abstract
T-cell chronic lymphoproliferative disorders (T-CLPD) are a heterogeneous group of mature T-cell malignancies, the classification of which remains challenging. In this study, we classified tumor cells from 86 patients diagnosed with either T-CLPD (n = 81) or T-cell acute lymphoblastic leukemia (n = 5) into precise functional and maturation-associated compartments, based on their phenotypic similarities with their normal maturation-related and functional associated T-cell counterparts. A database was generated using blood samples from 6 sex- and age-matched healthy donors as a template for normal T-cell subset flow cytometric immunophenotypes, to which tumor cells of individual patients were compared. Except for nodal T follicular-helper cell lymphoma and adult T-cell leukemia/lymphoma, which showed phenotypes overlapping with that of T follicular-helper and T regulatory cells, respectively, all other T-CLPD displayed immunophenotypic profiles consistent with conventional T helper (Th) cells, with different maturation-associated profiles per diagnostic category. These included predominant naive/naive-central memory phenotypes in T-cell prolymphocytic leukemia to terminal effector cytotoxic cellular profiles in T-cell large granular lymphocytic leukemia; other T-CLPD diagnostic categories (mostly Sézary syndrome/mycosis fungoides) resembled the diverse memory T-cell subsets. Interestingly, immunophenotypically less-mature tumor cells (T-cell prolymphocytic leukemia) displayed more heterogeneous Th profiles, whereas those with memory T-cell profiles showed more consistent Th-associated patterns (eg, Th2 or Th17 in Sézary syndrome/mycosis fungoides), and the most mature neoplasms (eg, T-cell large granular lymphocytic leukemia) systematically displayed a Th1-like pattern, reflecting progressively lower plasticity for the more advanced tumor-associated maturation stages. These findings confirm the presence of distinct phenotypic patterns resembling specific maturation-associated and Th-related profiles of normal T cells among distinct diagnostic categories of T-CLPD, which might contribute to a more precise classification of T-CLPD.
Collapse
Affiliation(s)
- F Javier Morán-Plata
- Translational and Clinical Research Program, Cancer Research Center, University of Salamanca, Salamanca, Spain; Cytometry Service, NUCLEUS, University of Salamanca, Salamanca, Spain; Departamento de Medicina, Universidad de Salamanca, Salamanca, Spain; Institute of Biomedical Research of Salamanca, Salamanca, Spain
| | - Noemí Muñoz-García
- Translational and Clinical Research Program, Cancer Research Center, University of Salamanca, Salamanca, Spain; Cytometry Service, NUCLEUS, University of Salamanca, Salamanca, Spain; Departamento de Medicina, Universidad de Salamanca, Salamanca, Spain; Institute of Biomedical Research of Salamanca, Salamanca, Spain
| | - Susana Barrena
- Translational and Clinical Research Program, Cancer Research Center, University of Salamanca, Salamanca, Spain; Cytometry Service, NUCLEUS, University of Salamanca, Salamanca, Spain; Departamento de Medicina, Universidad de Salamanca, Salamanca, Spain; Institute of Biomedical Research of Salamanca, Salamanca, Spain
| | - Ana Yeguas
- Service of Hematology, University Hospital of Salamanca, Salamanca, Spain
| | - Ana Balanzategui
- Institute of Biomedical Research of Salamanca, Salamanca, Spain; Service of Hematology, University Hospital of Salamanca, Salamanca, Spain; Biomedical Research Networking Centre Consortium of Oncology, Instituto de Salud Carlos III, Madrid, Spain
| | - Sonia Carretero-Domínguez
- Translational and Clinical Research Program, Cancer Research Center, University of Salamanca, Salamanca, Spain; Cytometry Service, NUCLEUS, University of Salamanca, Salamanca, Spain; Departamento de Medicina, Universidad de Salamanca, Salamanca, Spain; Institute of Biomedical Research of Salamanca, Salamanca, Spain; Biomedical Research Networking Centre Consortium of Oncology, Instituto de Salud Carlos III, Madrid, Spain
| | - Julio Pozo
- Translational and Clinical Research Program, Cancer Research Center, University of Salamanca, Salamanca, Spain; Cytometry Service, NUCLEUS, University of Salamanca, Salamanca, Spain; Departamento de Medicina, Universidad de Salamanca, Salamanca, Spain
| | - Quentin Lécrevisse
- Translational and Clinical Research Program, Cancer Research Center, University of Salamanca, Salamanca, Spain; Cytometry Service, NUCLEUS, University of Salamanca, Salamanca, Spain; Departamento de Medicina, Universidad de Salamanca, Salamanca, Spain; Institute of Biomedical Research of Salamanca, Salamanca, Spain; Biomedical Research Networking Centre Consortium of Oncology, Instituto de Salud Carlos III, Madrid, Spain
| | - María González-González
- Translational and Clinical Research Program, Cancer Research Center, University of Salamanca, Salamanca, Spain; Cytometry Service, NUCLEUS, University of Salamanca, Salamanca, Spain; Departamento de Medicina, Universidad de Salamanca, Salamanca, Spain; Institute of Biomedical Research of Salamanca, Salamanca, Spain
| | - Paloma Bárcena
- Translational and Clinical Research Program, Cancer Research Center, University of Salamanca, Salamanca, Spain; Cytometry Service, NUCLEUS, University of Salamanca, Salamanca, Spain; Departamento de Medicina, Universidad de Salamanca, Salamanca, Spain; Institute of Biomedical Research of Salamanca, Salamanca, Spain; Cell-purification Service, NUCLEUS, University of Salamanca, Salamanca, Spain
| | - Miguel Alcoceba
- Institute of Biomedical Research of Salamanca, Salamanca, Spain; Service of Hematology, University Hospital of Salamanca, Salamanca, Spain; Biomedical Research Networking Centre Consortium of Oncology, Instituto de Salud Carlos III, Madrid, Spain
| | - María Herrero-García
- Translational and Clinical Research Program, Cancer Research Center, University of Salamanca, Salamanca, Spain; Cytometry Service, NUCLEUS, University of Salamanca, Salamanca, Spain; Departamento de Medicina, Universidad de Salamanca, Salamanca, Spain; Institute of Biomedical Research of Salamanca, Salamanca, Spain
| | - Fernando Solano
- Hospital Ntra Sra del Prado, Talavera De La Reina, Toledo, Spain
| | - Miriam López-Parra
- Institute of Biomedical Research of Salamanca, Salamanca, Spain; Service of Hematology, University Hospital of Salamanca, Salamanca, Spain
| | - Alejandro Martín García-Sancho
- Departamento de Medicina, Universidad de Salamanca, Salamanca, Spain; Institute of Biomedical Research of Salamanca, Salamanca, Spain; Service of Hematology, University Hospital of Salamanca, Salamanca, Spain; Biomedical Research Networking Centre Consortium of Oncology, Instituto de Salud Carlos III, Madrid, Spain
| | - Cristiane de Sá Ferreira-Facio
- Internal Medicine Postgraduate Program, Faculty of Medicine, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil; Cytometry Service, Institute of Paediatrics and Puericultura Martagão Gesteira, Faculty of Medicine, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Neus Villamor
- Biomedical Research Networking Centre Consortium of Oncology, Instituto de Salud Carlos III, Madrid, Spain; Department of Pathology, Hematopathology Unit, Hospital Clínic, Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
| | - Catarina Lau
- Laboratory of Cytometry, Unit for Hematology Diagnosis, Department of Hematology, Hospital de Santo António, Centro Hospitalar Universitário do Porto, Unidade Multidisciplinar de Investigação Biomédica, Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Maria Dos Anjos Teixeira
- Laboratory of Cytometry, Unit for Hematology Diagnosis, Department of Hematology, Hospital de Santo António, Centro Hospitalar Universitário do Porto, Unidade Multidisciplinar de Investigação Biomédica, Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Vitor Botafogo
- Translational and Clinical Research Program, Cancer Research Center, University of Salamanca, Salamanca, Spain; Cytometry Service, NUCLEUS, University of Salamanca, Salamanca, Spain; Departamento de Medicina, Universidad de Salamanca, Salamanca, Spain
| | - Alberto Orfao
- Translational and Clinical Research Program, Cancer Research Center, University of Salamanca, Salamanca, Spain; Cytometry Service, NUCLEUS, University of Salamanca, Salamanca, Spain; Departamento de Medicina, Universidad de Salamanca, Salamanca, Spain; Institute of Biomedical Research of Salamanca, Salamanca, Spain; Biomedical Research Networking Centre Consortium of Oncology, Instituto de Salud Carlos III, Madrid, Spain
| | - Julia Almeida
- Translational and Clinical Research Program, Cancer Research Center, University of Salamanca, Salamanca, Spain; Cytometry Service, NUCLEUS, University of Salamanca, Salamanca, Spain; Departamento de Medicina, Universidad de Salamanca, Salamanca, Spain; Institute of Biomedical Research of Salamanca, Salamanca, Spain; Biomedical Research Networking Centre Consortium of Oncology, Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
11
|
Willett BAS, Thompson SB, Chen V, Dareshouri A, Jackson CL, Brunetti T, D'Alessandro A, Klarquist J, Nemkov T, Kedl RM. Mitochondrial protein OPA1 is required for the expansion of effector CD8 T cells. Cell Rep 2025; 44:115610. [PMID: 40261796 DOI: 10.1016/j.celrep.2025.115610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Revised: 03/14/2025] [Accepted: 04/02/2025] [Indexed: 04/24/2025] Open
Abstract
Short-lived effector cells are characterized metabolically by a highly glycolytic state, driving energy and biomass acquisition, whereas memory-fated T cells have historically been described as meeting these requirements through mitochondrial metabolism. Here, we show that the mitochondrial protein optic atrophy 1 (OPA1) is critical for rapidly dividing CD8 T cells in vivo, the requirement for which is most pronounced in effector CD8 T cells. More specifically, OPA1 supports proper cell cycle initiation and progression and the viability and survival of CD8 T cells during clonal expansion. Use of mice deficient in the mitochondrial membrane fusion proteins Mitofusin 1 and 2 (MFN1/2) in both in vivo proliferation/differentiation assays and ex vivo metabolic analysis indicates that the requirement for OPA1 during cell division supersedes its role in mitochondrial fusion. We conclude that OPA1 is critical for the generation and accumulation of short-lived effector cells that arise during the response to infection.
Collapse
Affiliation(s)
- Benjamin A S Willett
- Department of Immunology and Microbiology, University of Colorado-Denver Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Scott B Thompson
- Department of Immunology and Microbiology, University of Colorado-Denver Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Vincent Chen
- Department of Immunology and Microbiology, University of Colorado-Denver Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Anza Dareshouri
- Department of Cell and Developmental Biology, University of Colorado-Denver Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Conner L Jackson
- Department of Biostatistics and Informatics, Colorado School of Public Health, University of Colorado-Denver Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Tonya Brunetti
- Department of Immunology and Microbiology, University of Colorado-Denver Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Angelo D'Alessandro
- Department of Biochemistry & Molecular Genetics, University of Colorado-Denver Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Jared Klarquist
- Department of Immunology and Microbiology, University of Colorado-Denver Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Travis Nemkov
- Department of Biochemistry & Molecular Genetics, University of Colorado-Denver Anschutz Medical Campus, Aurora, CO 80045, USA.
| | - Ross M Kedl
- Department of Immunology and Microbiology, University of Colorado-Denver Anschutz Medical Campus, Aurora, CO 80045, USA.
| |
Collapse
|
12
|
Bonilha CS, Veras FP, Dos Santos Ramos A, Gomes GF, Rodrigues Lemes RM, Arruda E, Alves-Filho JC, Cunha TM, Cunha FQ. PAD4 inhibition impacts immune responses in SARS-CoV-2 infection. Mucosal Immunol 2025:S1933-0219(25)00044-3. [PMID: 40258416 DOI: 10.1016/j.mucimm.2025.04.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Revised: 04/04/2025] [Accepted: 04/14/2025] [Indexed: 04/23/2025]
Abstract
Protein arginine deiminase 4 (PAD4) has emerged as a potential therapeutic target for various diseases due to its role in promoting neutrophil extracellular trap (NET) formation. NETs, composed of DNA and antimicrobial proteins, serve as a defense mechanism against pathogens but can also drive lung injury, particularly in SARS-CoV-2 infection. In this study, we examined the effects of PAD4 inhibition on clinical outcomes and adaptive immunity within the context of SARS-CoV-2 infection. Our results show that PAD4 pharmacological inhibition reduced lung NET concentration and improved clinical outcomes, similar to treatment with recombinant human DNase (rhDNase), which degrades NET structure. However, in contrast to rhDNase, PAD4 targeting diminished virus-specific T cell responses by impairing dendritic cell antigen presentation and reducing IL-2 signaling by affecting its production by T cells. In line with these observations, PAD4 pharmacological inhibition diminished antigen-specific T cell responses in a model of lung inflammation. These findings highlight the importance of carefully evaluating PAD4 as a therapeutic target in COVID-19, given its potential to compromise adaptive immune responses crucial for fighting the virus.
Collapse
Affiliation(s)
- Caio Santos Bonilha
- Center for Research in Inflammatory Diseases, Ribeirao Preto Medical School, University of Sao Paulo, 14049-900, Brazil; Institute of Infection, Immunity and Inflammation, University of Glasgow, G12 8TA, UK; Institute of Developmental & Regenerative Medicine, University of Oxford, OX3 7TY, UK.
| | - Flavio Protasio Veras
- Center for Research in Inflammatory Diseases, Ribeirao Preto Medical School, University of Sao Paulo, 14049-900, Brazil; Institute of Biomedical Sciences, Federal University of Alfenas, 37130-001, Brazil
| | - Anderson Dos Santos Ramos
- Center for Research in Inflammatory Diseases, Ribeirao Preto Medical School, University of Sao Paulo, 14049-900, Brazil
| | - Giovanni Freitas Gomes
- Center for Research in Inflammatory Diseases, Ribeirao Preto Medical School, University of Sao Paulo, 14049-900, Brazil
| | | | - Eurico Arruda
- Virology Research Center, Ribeirao Preto Medical School, University of Sao Paulo 14049-900, Brazil
| | - José Carlos Alves-Filho
- Center for Research in Inflammatory Diseases, Ribeirao Preto Medical School, University of Sao Paulo, 14049-900, Brazil
| | - Thiago Mattar Cunha
- Center for Research in Inflammatory Diseases, Ribeirao Preto Medical School, University of Sao Paulo, 14049-900, Brazil
| | - Fernando Queiroz Cunha
- Center for Research in Inflammatory Diseases, Ribeirao Preto Medical School, University of Sao Paulo, 14049-900, Brazil.
| |
Collapse
|
13
|
Culina S, Commère PH, Turc E, Jouy A, Pellegrini S, Roux T, Hasan M, Monot M, Michel F. MicroRNA signatures of CD4 + T cell subsets in healthy and multiple sclerosis subjects determined by small RNA-sequencing. J Neuroimmunol 2025; 401:578531. [PMID: 40010156 DOI: 10.1016/j.jneuroim.2025.578531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 01/13/2025] [Accepted: 01/19/2025] [Indexed: 02/28/2025]
Abstract
Diverse CD4+ T cell subsets with specialized functions operate at different phases of the immune response. Among these are phenotypically and functionally characterized naïve, central memory (CM), effector memory (EM), and regulatory (Treg) cells. Using small RNA-sequencing, we have profiled miRNAs in these cell subsets from healthy subjects and untreated patients with relapsing-remitting multiple sclerosis (RRMS). MiRNA genomic clustering and abundance were also investigated. From the 60 most differentially expressed miRNAs, broad and highly selective core signatures were determined for naïve and memory cells at homeostasis, while miR-146a-5p was strongly upregulated in Treg cells. In line with other studies, a 5-miRNA core was identified for naïve cells (miR-125b-5p, miR-99a-5p, miR-365a-3p, miR-365b-3p, miR-193b-3p). In memory cells, a number of identical miRNAs were more expressed in EM than CM cells, supporting the progressive T cell differentiation model. This was particularly the case for an 8-miRNA core (members from miR-23a∼27a∼24-2, miR-23b∼27b∼24-1, miR-221∼222 clusters, miR-22-3p, miR-181c-5p) and for the large ChrXq27.3 miR-506∼514 cluster. Interestingly, most of these miRNAs were reported to negatively regulate cell proliferation and survival. Finally, we found that the miRNA core signatures of naïve and memory CD4+ T cells were conserved in RRMS patients. Only few miRNAs were quantitatively modified and, among these, miR-1248 was validated to be downregulated in EM cells. Overall, this study expands and provides novel insights into miRNA profiling of CD4+ T cell subsets that may be useful for further investigations.
Collapse
Affiliation(s)
- Slobodan Culina
- Single Cell Biomarkers UTechS, Paris Cité University, Institut Pasteur, Paris, France
| | | | - Elodie Turc
- Biomics Technological Platform, Paris Cité University, Institut Pasteur, Paris, France
| | - Axel Jouy
- Paris Saclay University, Saclay, France; T cell activation and function DIO3 team, Department of Immunology, Paris Cité University, Institut Pasteur, Paris, France
| | - Sandra Pellegrini
- Unit of Cytokine Signaling, Inserm U1224, Paris Cité University, Institut Pasteur, Paris, France
| | - Thomas Roux
- CRC-SEP, Department of Neurology, Pitié-Salpêtrière Hospital, Paris, France
| | - Milena Hasan
- Single Cell Biomarkers UTechS, Paris Cité University, Institut Pasteur, Paris, France
| | - Marc Monot
- Biomics Technological Platform, Paris Cité University, Institut Pasteur, Paris, France
| | - Frédérique Michel
- Unit of Cytokine Signaling, Inserm U1224, Paris Cité University, Institut Pasteur, Paris, France; T cell activation and function DIO3 team, Department of Immunology, Paris Cité University, Institut Pasteur, Paris, France.
| |
Collapse
|
14
|
Sainz-Mejías M, Ma C, Hou Y, Jurado-Martin I, Romerio A, Franco AR, Shaik MM, Tomás-Cortázar J, Peri F, McClean S. Monosaccharide-Based Synthetic TLR4 Agonist Enhances Vaccine Efficacy against Pseudomonas aeruginosa Challenge. ACS Infect Dis 2025; 11:894-904. [PMID: 40129118 PMCID: PMC11998000 DOI: 10.1021/acsinfecdis.4c00932] [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: 11/19/2024] [Revised: 03/13/2025] [Accepted: 03/14/2025] [Indexed: 03/26/2025]
Abstract
Vaccine adjuvants are critical to improve the immunogenicity, efficacy, and durability of vaccines; however, their development has lagged behind that of vaccine antigens. Monophosphoryl lipid A (MPLA), a clinically approved adjuvant that stimulates Toll-like receptor 4 (TLR4), faces manufacturing challenges due to its complex and long synthesis. With the aim of simplifying the structure of MPLA while retaining its biological activity, we developed monosaccharide-based molecules FP18 and FP20Rha that activate TLR4 signaling. Both TLR4 agonists induced robust antibody activity against the model antigen, ovalbumin. Here, we report the potential of these TLR4 agonists to enhance the protective efficacy of the well-characterized OprF antigen against P. aeruginosa infection. OprF adjuvanted with FP18 showed reduced bacterial loads in lungs and spleens, relative to antigen alone in an acute P. aeruginosa pneumonia model. FP18-adjuvanted OprF also enhanced the production of anti-OprF antibodies and stimulated IFNγ and TNF in CD4+ T cells, suggesting a Th1-skewed cellular immune response. These adjuvants have promise for accelerating the development of effective vaccines against P. aeruginosa and other infectious diseases.
Collapse
Affiliation(s)
- Maite Sainz-Mejías
- School
of Biomolecular and Biomedical Sciences and Conway Institute of Biomolecular
and Biomedical Research, University College
Dublin, Belfield, Dublin 4 D04 V1W8, Ireland
| | - Chaoying Ma
- School
of Biomolecular and Biomedical Sciences and Conway Institute of Biomolecular
and Biomedical Research, University College
Dublin, Belfield, Dublin 4 D04 V1W8, Ireland
| | - Yueran Hou
- School
of Biomolecular and Biomedical Sciences and Conway Institute of Biomolecular
and Biomedical Research, University College
Dublin, Belfield, Dublin 4 D04 V1W8, Ireland
| | - Irene Jurado-Martin
- School
of Biomolecular and Biomedical Sciences and Conway Institute of Biomolecular
and Biomedical Research, University College
Dublin, Belfield, Dublin 4 D04 V1W8, Ireland
| | - Alessio Romerio
- Department
of Biotechnology and Biosciences, University
of Milano-Bicocca, Piazza della Scienza, 2, Milano 20126, Italy
| | - Ana Rita Franco
- Department
of Biotechnology and Biosciences, University
of Milano-Bicocca, Piazza della Scienza, 2, Milano 20126, Italy
| | - Mohammed Monsoor Shaik
- Department
of Biotechnology and Biosciences, University
of Milano-Bicocca, Piazza della Scienza, 2, Milano 20126, Italy
| | - Julen Tomás-Cortázar
- School
of Biomolecular and Biomedical Sciences and Conway Institute of Biomolecular
and Biomedical Research, University College
Dublin, Belfield, Dublin 4 D04 V1W8, Ireland
| | - Francesco Peri
- Department
of Biotechnology and Biosciences, University
of Milano-Bicocca, Piazza della Scienza, 2, Milano 20126, Italy
| | - Siobhán McClean
- School
of Biomolecular and Biomedical Sciences and Conway Institute of Biomolecular
and Biomedical Research, University College
Dublin, Belfield, Dublin 4 D04 V1W8, Ireland
| |
Collapse
|
15
|
Akhtar A, Shakir M, Ansari MS, Divya, Faizan MI, Chauhan V, Singh A, Alam R, Azmi I, Sharma S, Pracha M, Uddin IM, Bashir U, Shahni SN, Chaudhuri R, Albogami S, Ganguly R, Sagar S, Singh VP, Kharya G, Srivastava AK, Mabalirajan U, Roy SS, Rahman I, Ahmad T. Bioengineering the metabolic network of CAR T cells with GLP-1 and Urolithin A increases persistence and long-term anti-tumor activity. Cell Rep Med 2025; 6:102021. [PMID: 40107240 PMCID: PMC11970383 DOI: 10.1016/j.xcrm.2025.102021] [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: 05/17/2024] [Revised: 12/10/2024] [Accepted: 02/18/2025] [Indexed: 03/22/2025]
Abstract
Constant tumor antigen exposure disrupts chimeric antigen receptor (CAR) T cell metabolism, limiting their persistence and anti-tumor efficacy. To address this, we develop metabolically reprogrammed CAR (MCAR) T cells with enhanced autophagy and mitophagy. A compound screening identifies a synergy between GLP-1R agonist (semaglutide [SG]) and Urolithin A (UrA), which activate autophagy through mTOR (mechanistic target of rapamycin) inhibition and mitophagy via Atg4b activation, maintaining mitochondrial metabolism in CAR T cells (MCAR T-1). These changes increase CD8+ T memory cells (Tm), enhancing persistence and anti-tumor activity in vitro and in xenograft models. GLP-1R knockdown in CAR T cells diminishes autophagy/mitophagy induction, confirming its critical role. We further engineer GLP-1-secreting cells (MCAR T-2), which exhibited sustained memory, stemness, and long-term persistence, even under tumor re-challenge. MCAR T-2 cells also reduce cytokine release syndrome (CRS) risks while demonstrating potent anti-tumor effects. This strategy highlights the potential of metabolic reprogramming via targeting autophagy/mitophagy pathways to improve CAR T cell therapy outcomes, ensuring durability and efficacy.
Collapse
Affiliation(s)
- Areej Akhtar
- Multidisciplinary Centre for Advanced Research and Studies, Jamia Millia Islamia, New Delhi, India
| | - Md Shakir
- Multidisciplinary Centre for Advanced Research and Studies, Jamia Millia Islamia, New Delhi, India
| | - Mohammad Sufyan Ansari
- Multidisciplinary Centre for Advanced Research and Studies, Jamia Millia Islamia, New Delhi, India
| | - Divya
- Multidisciplinary Centre for Advanced Research and Studies, Jamia Millia Islamia, New Delhi, India
| | - Md Imam Faizan
- Multidisciplinary Centre for Advanced Research and Studies, Jamia Millia Islamia, New Delhi, India
| | - Varnit Chauhan
- Multidisciplinary Centre for Advanced Research and Studies, Jamia Millia Islamia, New Delhi, India
| | - Aashi Singh
- Multidisciplinary Centre for Advanced Research and Studies, Jamia Millia Islamia, New Delhi, India
| | - Ruquaiya Alam
- Multidisciplinary Centre for Advanced Research and Studies, Jamia Millia Islamia, New Delhi, India
| | - Iqbal Azmi
- Multidisciplinary Centre for Advanced Research and Studies, Jamia Millia Islamia, New Delhi, India
| | - Sheetal Sharma
- Multidisciplinary Centre for Advanced Research and Studies, Jamia Millia Islamia, New Delhi, India
| | - Mehak Pracha
- Multidisciplinary Centre for Advanced Research and Studies, Jamia Millia Islamia, New Delhi, India
| | - Insha Mohi Uddin
- Multidisciplinary Centre for Advanced Research and Studies, Jamia Millia Islamia, New Delhi, India
| | - Uzma Bashir
- Multidisciplinary Centre for Advanced Research and Studies, Jamia Millia Islamia, New Delhi, India
| | - Syeda Najidah Shahni
- Multidisciplinary Centre for Advanced Research and Studies, Jamia Millia Islamia, New Delhi, India
| | - Rituparna Chaudhuri
- Indian Institute of Science, Centre for Brain Research, Bengaluru, Karnataka, India
| | - Sarah Albogami
- Department of Biotechnology, College of Science, Taif University, Taif, Saudi Arabia
| | - Rik Ganguly
- Department of Biotechnology & Bioinformatics, North-Eastern Hill University, Shillong, India
| | - Shakti Sagar
- CSIR-Institute of Genomics & Integrative Biology, New Delhi, India
| | - Vijay Pal Singh
- CSIR-Institute of Genomics & Integrative Biology, New Delhi, India
| | - Gaurav Kharya
- Centre for Bone Marrow Transplant & Cellular Therapy, Indraprastha Apollo Hospital, New Delhi, India
| | | | | | - Soumya Sinha Roy
- CSIR-Institute of Genomics & Integrative Biology, New Delhi, India
| | - Irfan Rahman
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Tanveer Ahmad
- Multidisciplinary Centre for Advanced Research and Studies, Jamia Millia Islamia, New Delhi, India.
| |
Collapse
|
16
|
Pandit JJ, Yassin AAK, Martin CU, Saux GL, Porgador A, Schvartzman M. Effect of binary mechanical environment on T cell function. Acta Biomater 2025; 195:83-93. [PMID: 39952341 DOI: 10.1016/j.actbio.2025.02.029] [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] [Revised: 02/04/2025] [Accepted: 02/11/2025] [Indexed: 02/17/2025]
Abstract
T cells, key players in the immune system, recognize antigens via T-cell receptors (TCRs) and require additional costimulatory and cytokine signals for full activation. Beyond biochemical signals, T cells also respond to mechanical cues such as tissue stiffness. Traditional ex-vivo mechanostimulating platforms, however, present a uniform mechanical environment, unlike the heterogeneous conditions T cells encounter in-vivo. This work introduces a mechanically-biphasic T-cell stimulating surface, with alternating soft and stiff microdomains, to mimic the complex mechanical signals T cells face. Results show that T cells exposed to this biphasic environment do not average the mechanical signals but instead respond similarly to those on a homogeneously soft surface, leading to lower activation compared to those on a stiff surface. Interestingly, long-term exposure to these patterns enhances the proliferation of central memory and effector T cell phenotypes, similar to stiff environments. These findings reveal the non-linear nature of T cell mechanosensing and suggest that mechanical heterogeneity plays a critical role in modulating T cell responses, providing new insights into T cell activation and potential implications for immunotherapies. STATEMENT OF SIGNIFICANCE: This research offers a fresh perspective in T cell mehanosensing, an important yet underexplored aspect of immunity. While previous studies have demonstrated that T cells sense homogeneous mechanical environments ex-vivo, their ability to discern and respond to simultaneous mechanical cues-resembling the complexity of in-vivo conditions-remained unexamined. By designing a mechanically patterned surface with alternating soft and stiff microdomains, this study simulates the diverse mechanical landscape encountered by T cells in-vivo. The findings reveal that T cells predominantly respond to this pattern as they would to a uniformly soft environment. This insight, showing that mechanical signals shape T cell activation and promote specific phenotypes, enhances our understanding of T cell biology and points to new directions for immunotherapy development.
Collapse
Affiliation(s)
- Jatin Jawhir Pandit
- Department of Materials Engineering, Ben-Gurion University of the Negev, Beer-Sheva , Israel; Ilse Katz Institute for Nanoscale Science and Technology, Ben-Gurion University of the Negev, Beer-Sheva , Israel
| | - Abed Al-Kader Yassin
- The Shraga Segal Department of Microbiology, Immunology, and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| | - Carlos Ureña Martin
- Department of Materials Engineering, Ben-Gurion University of the Negev, Beer-Sheva , Israel; Ilse Katz Institute for Nanoscale Science and Technology, Ben-Gurion University of the Negev, Beer-Sheva , Israel
| | - Guillaume Le Saux
- Department of Materials Engineering, Ben-Gurion University of the Negev, Beer-Sheva , Israel; Ilse Katz Institute for Nanoscale Science and Technology, Ben-Gurion University of the Negev, Beer-Sheva , Israel
| | - Angel Porgador
- The Shraga Segal Department of Microbiology, Immunology, and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| | - Mark Schvartzman
- Department of Materials Engineering, Ben-Gurion University of the Negev, Beer-Sheva , Israel; Ilse Katz Institute for Nanoscale Science and Technology, Ben-Gurion University of the Negev, Beer-Sheva , Israel.
| |
Collapse
|
17
|
Gilboa E, Gupta V, Muharemagic D, Ham S, Stelekati E, Clark E. KLF2 inhibition expands tumor-resident T cells and enhances tumor immunity. RESEARCH SQUARE 2025:rs.3.rs-5966555. [PMID: 40162209 PMCID: PMC11952643 DOI: 10.21203/rs.3.rs-5966555/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Tissue resident memory CD8+ T cells (Trm) constitute a distinct population of non-circulating memory T cells1-5 vastly exceeding the number of circulating T cells5, and play a pivotal role in protective immunity against pathogens6-8. How to promote the generation of vaccine specific Trm remains an important challenge. Whether Trm contribute also to immune control of tumors or just correlate with an unrelated process linked to clinical outcome has not been unequivocally established9,10, and phenotypic markers such as co-expression of CD69 and CD103 or CD49a integrins commonly used to monitor tumor infiltrating Trm do not unambiguously define this subset. Here we tested the hypothesis that transient downregulation of KLF2, the most conserved feature of Trm ontogeny4,11,12, will promote the differentiation of vaccine activated CD8+ T cells into Trm and enhance antitumor immunity. We show that 4-1BB antibody targeted delivery of a KLF2 siRNA to tumor bearing mice led to the downregulation of KLF2 in vaccine activated CD8+ T cells and the accumulation of phenotypically defined intratumoral CD69+CD103+ and CD69+CD49a+ CD8+ T cells which correlated with enhanced control of tumor growth. This study could serve as the foundation of a broadly applicable and clinically useful way to promote the generation of vaccine specific Trm and provides direct evidence that intratumoral CD8+CD69+CD103+ and CD8+CD69+CD49a+ cells are indeed Trm and that Trm contribute to tumor immunity.
Collapse
|
18
|
Wang LT, Juang SE, Lan CL, Chang HH, He AC, Chen WA, Huang YW, Van Dyke TE, Chen YW, Ma KSK. Titanium implants trigger extra-periodontal T cell-mediated immunity. Biomater Sci 2025; 13:1543-1553. [PMID: 39957356 DOI: 10.1039/d4bm00246f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/18/2025]
Abstract
Peri-implant inflammation and periodontitis share a common etiology rooted in periodontopathic bacterial invasion, with periodontitis notably linked to systemic inflammatory comorbidities involving T cells. However, the intricate processes within the peri-implant microenvironment and systemic repercussions of implants, particularly related to implant materials, remain inadequately understood. We aim to elucidate the impact of contact with titanium materials, widely employed in dental implants for their high biocompatibility and excellent corrosion resistance, on diverse T cell subpopulations. This study adopts a comprehensive approach, encompassing (1) transcriptomic profiling of peri-implant epithelium in a rat model, (2) examination of phenotypic and functional changes in T cell immunity in human blood cells cultured on titanium discs, and (3) in vivo validation of T cells in implanted mice. Transcriptomic evidence and functional in vitro results revealed that exposure to titanium materials promoted T cell activation and differentiation towards inflammatory subsets, and escalated the secretion of corresponding cytokines. In vivo results showed that most of the gingiva-extracted T cells were activated in both healthy and implanted mice, the latter exhibiting significant lymphadenitis. High-dimensional flow cytometric findings in the in vivo lymphadenitis model indicated titanium-induced T cell immunity, involving preferential activation of Th1, Th17, and Tc1 cells over Tregs in adjacent lymph nodes within three days after implant placement. These findings highlight the pivotal role of T cells in the initiation of peri-implant inflammation, emphasizing the need to understand extra-periodontal inflammatory complications associated with implant surgeries. Our study provides a foundation for future therapeutic strategies targeting T cell responses to enhance the success and longevity of dental implant treatments.
Collapse
Affiliation(s)
- Li-Tzu Wang
- School of Medical Laboratory Science and Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
- Ph.D. Program in Medical Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Sin-Ei Juang
- Department of Anesthesiology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University, College of Medicine, Kaohsiung, Taiwan
| | - Chi-Lun Lan
- Department of Dentistry, National Taiwan University Hospital & Graduate Institute of Clinical Dentistry, School of Dentistry, National Taiwan University, Taipei, Taiwan.
| | - Hsuan-Hao Chang
- Department of Dentistry, National Taiwan University Hospital & Graduate Institute of Clinical Dentistry, School of Dentistry, National Taiwan University, Taipei, Taiwan.
| | - Ai-Chia He
- Graduate Institute of Oral Biology, School of Dentistry, National Taiwan University, Taipei, Taiwan
| | - Wei-An Chen
- Department of Dentistry, National Taiwan University Hospital & Graduate Institute of Clinical Dentistry, School of Dentistry, National Taiwan University, Taipei, Taiwan.
| | - Yu-Wen Huang
- Department of Medical Research, National Taiwan University Hospital, Taipei, Taiwan
| | - Thomas E Van Dyke
- Center for Clinical and Translational Research, ADA Forsyth Institute, Cambridge, Massachusetts, USA.
- Department of Oral Medicine, Infection, and Immunity, Harvard School of Dental Medicine, Boston, Massachusetts, USA
| | - Yi-Wen Chen
- Department of Dentistry, National Taiwan University Hospital & Graduate Institute of Clinical Dentistry, School of Dentistry, National Taiwan University, Taipei, Taiwan.
- Graduate Institute of Oral Biology, School of Dentistry, National Taiwan University, Taipei, Taiwan
| | - Kevin Sheng-Kai Ma
- Center for Global Health, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
19
|
Xiao Q, He S, Wang C, Zhou Y, Zeng C, Liu J, Liu T, Li T, Quan X, Wang L, Zhai L, Liu Y, Li J, Zhang X, Liu Y. Deep Thought on the HIV Cured Cases: Where Have We Been and What Lies Ahead? Biomolecules 2025; 15:378. [PMID: 40149913 PMCID: PMC11940578 DOI: 10.3390/biom15030378] [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: 01/02/2025] [Revised: 02/26/2025] [Accepted: 03/03/2025] [Indexed: 03/29/2025] Open
Abstract
Antiretroviral therapy (ART) can effectively suppress the replication of human immunodeficiency virus (HIV), but it cannot completely eradicate the virus. The persistent existence of the HIV reservoir is a major obstacle in the quest for a cure. To date, there have been a total of seven cured cases of HIV worldwide. These patients all cleared HIV while undergoing allogeneic stem cell transplantation (allo-HSCT) for hematological malignancies. However, in these cases, the specific mechanism by which allo-HSCT leads to the eradication of HIV remains unclear, so it is necessary to conduct an in-depth analysis. Due to the difficulty in obtaining donors and the risks associated with transplantation, this treatment method is not applicable to all HIV patients. There is still a need to explore new treatment strategies. In recent years, emerging therapies such as neutralizing antibody immunotherapy, chimeric antigen receptor T cell (CAR-T) therapy, gene editing, and antiviral therapies targeting the reservoir have attracted wide attention due to their ability to effectively inhibit HIV replication. This article first elaborates on the nature of the HIV reservoir, then deeply explores the treatment modalities and potential success factors of HIV cured cases, and finally discusses the current novel treatment methods, hoping to provide comprehensive and feasible strategies for achieving the cure of HIV.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | - Xiaomei Zhang
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Department of Hematology-Oncology, Chongqing University Cancer Hospital, Chongqing 400030, China
| | - Yao Liu
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Department of Hematology-Oncology, Chongqing University Cancer Hospital, Chongqing 400030, China
| |
Collapse
|
20
|
Trujillo‐Vargas CM, Rendón‐Macías LM, Paredes Guerrero RY, de Paiva CS, Cardona‐Arias JA. Lymphocyte Subpopulations in the Healthy Human Lacrimal Gland and Their Variations With Age and Sex, Systematic Review 1960-2023. Immun Inflamm Dis 2025; 13:e70167. [PMID: 40105662 PMCID: PMC11921470 DOI: 10.1002/iid3.70167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 02/18/2025] [Accepted: 02/23/2025] [Indexed: 03/20/2025] Open
Abstract
BACKGROUND Immunosenescence has been associated with an imbalance in the lacrimal functional unit and histopathological changes in exocrine glands, especially in women. OBJECTIVE To define the main lymphocyte subpopulations in the human lacrimal gland and their variations with age and sex, according to scientific articles published between 1960 and 2023. METHODS A systematic review was performed on PubMed, ScienceDirect, and Google Scholar. The PRISMA 2020 guidelines were applied for the search and selection of studies. The methodological quality was evaluated with the STROBE guidelines. A meta-analysis of three selected articles dichotomizing lymphocytic infiltrates according to age group was also performed. RESULTS We selected 20 observational studies, including 774 healthy individuals (722 cadavers). The articles evaluated the lymphocyte infiltration with hematoxylin and eosin staining, immunohistochemistry and immunofluorescence. There was high variability in the criteria to define the apparently human lacrimal and to quantify the lymphocytic infiltration. There was an underrepresentation of individuals younger than 40 years (12.6%), and female sex (38.9%). Three articles reported an association of age and sex with lymphocytic infiltration in the healthy lacrimal gland, while two articles did not. Plasma cells were the most abundant lymphocyte subpopulation in the healthy lacrimal gland, including IgA-containing plasma cells. B cells were reported to be very scarce in the LG in two articles. In the meta-analysis of three selected articles, no statistical difference in lymphocytic infiltration was found between individuals younger and older than 60. CONCLUSION There is the need of further observational studies, better defining the study design, with similar representation across sex and ages to ascertain what are the changes of lymphocytic composition in the lacrimal gland related to age and sex. Further studies are also needed to assess the dynamics of lymphocytic populations in a more detailed manner using cutting-edge methodologies such as single-cell sequencing or transcriptomics. TRIAL REGISTRATION PROSPERO 2023 CRD42023435653 Available from: https://www.crd.york.ac.uk/prospero/display_record.php?ID=CRD42023435653.
Collapse
Affiliation(s)
- Claudia M. Trujillo‐Vargas
- Grupo de Inmunodeficiencias Primarias, Facultad de Medicina, Universidad de Antioquia UdeAMedellínColombia
| | | | | | - Cinta S. de Paiva
- Department of OphthalmologyBaylor College of MedicineHoustonTexasUSA
| | | |
Collapse
|
21
|
Ma M, Jin C, Dong Q. Intratumoral Heterogeneity and Immune Microenvironment in Hepatoblastoma Revealed by Single-Cell RNA Sequencing. J Cell Mol Med 2025; 29:e70482. [PMID: 40099956 PMCID: PMC11915626 DOI: 10.1111/jcmm.70482] [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: 11/09/2024] [Revised: 02/21/2025] [Accepted: 02/27/2025] [Indexed: 03/20/2025] Open
Abstract
Hepatoblastoma (HB) is a common paediatric liver malignancy characterised by significant intratumoral heterogeneity and a complex tumour microenvironment (TME). Using single-cell RNA sequencing (scRNA-seq), we analysed 43,592 cells from three tumour regions and adjacent normal tissue of an HB patient. Our study revealed distinct cellular compositions and varying degrees of malignancy across different tumour regions, with the T1 region showing the highest malignancy and overexpression of HMGB2 and TOP2A. Survival analysis demonstrated that high HMGB2 expression is associated with poor prognosis and increased recurrence, suggesting its potential as a prognostic marker. Additionally, we identified a diverse immune microenvironment enriched with regulatory T cells (Tregs) and CD8+ effector memory T cells (Tem), indicating potential immune evasion mechanisms. Notably, CTLA-4 and PD-1 were highly expressed in Tregs and Tem cells, highlighting their potential as immunotherapy targets. Myeloid cells, including Kupffer cells and dendritic cells, also exhibited distinct functional roles in different tumour regions. This study provides the first comprehensive single-cell atlas of HB, revealing critical insights into its intratumoral heterogeneity and immune microenvironment. Our findings not only advance the understanding of HB biology but also offer new directions for precision medicine, including the development of targeted therapies and immunotherapeutic strategies to improve patient outcomes.
Collapse
Affiliation(s)
- Mingdi Ma
- Department of Pediatric SurgeryThe Affiliated Hospital of Qingdao UniversityQingdaoChina
| | - Chen Jin
- Department of Pediatric SurgeryThe Affiliated Hospital of Qingdao UniversityQingdaoChina
| | - Qian Dong
- Department of Pediatric SurgeryThe Affiliated Hospital of Qingdao UniversityQingdaoChina
- Shandong Key Laboratory of Digital Medicine and Computer Assisted SurgeryThe Affiliated Hospital of Qingdao UniversityQingdaoChina
| |
Collapse
|
22
|
Zhang J, Jiao D, Qi X, Zhang Y, Liu X, Pan T, Gao H, Liu Z, Ding D, Feng G. An Albumin-Photosensitizer Supramolecular Assembly with Type I ROS-Induced Multifaceted Tumor Cell Deaths for Photodynamic Immunotherapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2410405. [PMID: 39804949 PMCID: PMC11884554 DOI: 10.1002/advs.202410405] [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: 08/28/2024] [Revised: 12/19/2024] [Indexed: 01/16/2025]
Abstract
Photodynamic therapy holds great potentials in cancer treatment, yet its effectiveness in hypoxic solid tumor is limited by the oxygen-dependence and insufficient oxidative potential of conventional type II reactive oxygen species (ROS). Herein, the study reports a supramolecular photosensitizer, BSA@TPE-BT-SCT NPs, through encapsulating aggregation-enhanced emission photosensitizer by bovine serum albumin (BSA) to significantly enhance ROS, particularly less oxygen-dependent type I ROS for photodynamic immunotherapy. The abundant type I ROS generated by BSA@TPE-BT-SCT NPs induce multiple forms of programmed cell death, including apoptosis, pyroptosis, and ferroptosis. These multifaceted cell deaths synergistically facilitate the release of damage-associated molecular patterns and antitumor cytokines, thereby provoking robust antitumor immunity. Both in vitro and in vivo experiments confirmed that BSA@TPE-BT-SCT NPs elicited the immunogenic cell death, enhance dendritic cell maturation, activate T cell, and reduce myeloid-derived suppressor cells, leading to the inhibition of both primary and distant tumors. Additionally, BSA@TPE-BT-SCP NPs also exhibited excellent antitumor performance in a humanized mice model, evidenced by a reduction in senescent T cells among these activated T cells. The findings advance the development of robust type I photosensitizers and unveil the important role of type I ROS in enhancing multifaceted tumor cell deaths and antitumor immunogenicity.
Collapse
Affiliation(s)
- Jingtian Zhang
- Frontiers Science Center for Cell ResponsesState Key Laboratory of Medicinal Chemical BiologyKey Laboratory of Bioactive MaterialsMinistry of Educationand College of Life SciencesNankai UniversityTianjin300071China
| | - Di Jiao
- Frontiers Science Center for Cell ResponsesState Key Laboratory of Medicinal Chemical BiologyKey Laboratory of Bioactive MaterialsMinistry of Educationand College of Life SciencesNankai UniversityTianjin300071China
| | - Xinwen Qi
- Frontiers Science Center for Cell ResponsesState Key Laboratory of Medicinal Chemical BiologyKey Laboratory of Bioactive MaterialsMinistry of Educationand College of Life SciencesNankai UniversityTianjin300071China
| | - Yufan Zhang
- Frontiers Science Center for Cell ResponsesState Key Laboratory of Medicinal Chemical BiologyKey Laboratory of Bioactive MaterialsMinistry of Educationand College of Life SciencesNankai UniversityTianjin300071China
| | - Xiaoang Liu
- Frontiers Science Center for Cell ResponsesState Key Laboratory of Medicinal Chemical BiologyKey Laboratory of Bioactive MaterialsMinistry of Educationand College of Life SciencesNankai UniversityTianjin300071China
| | - Tengwu Pan
- Frontiers Science Center for Cell ResponsesState Key Laboratory of Medicinal Chemical BiologyKey Laboratory of Bioactive MaterialsMinistry of Educationand College of Life SciencesNankai UniversityTianjin300071China
| | - Heqi Gao
- Frontiers Science Center for Cell ResponsesState Key Laboratory of Medicinal Chemical BiologyKey Laboratory of Bioactive MaterialsMinistry of Educationand College of Life SciencesNankai UniversityTianjin300071China
| | - Zhaoyun Liu
- Department of HematologyTianjin Medical University General HospitalTianjin Key Laboratory of Bone Marrow Failure and Malignant Hemopoietic Clone ControlTianjin Institute of HematologyTianjin300052China
| | - Dan Ding
- Frontiers Science Center for Cell ResponsesState Key Laboratory of Medicinal Chemical BiologyKey Laboratory of Bioactive MaterialsMinistry of Educationand College of Life SciencesNankai UniversityTianjin300071China
| | - Guangxue Feng
- Guangdong Provincial Key Laboratory of Luminescence from Molecular AggregatesState Key Laboratory of Luminescent Materials and DevicesSchool of Materials Science and EngineeringAIE InstituteSouth China University of TechnologyGuangzhou510640China
| |
Collapse
|
23
|
Poudel K, Vithiananthan T, Kim JO, Tsao H. Recent progress in cancer vaccines and nanovaccines. Biomaterials 2025; 314:122856. [PMID: 39366184 DOI: 10.1016/j.biomaterials.2024.122856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 09/03/2024] [Accepted: 09/26/2024] [Indexed: 10/06/2024]
Abstract
Vaccine science, nanotechnology, and immunotherapy are at the forefront of cancer treatment strategies, each offering significant potential for enhancing tumor-specific immunity and establishing long-lasting immune memory to prevent tumor recurrence. Despite the promise of these personalized and precision-based anti-cancer approaches, challenges such as immunosuppression, suboptimal immune activation, and T-cell exhaustion continue to hinder their effectiveness. The limited clinical success of cancer vaccines often stems from difficulties in identifying effective antigens, efficiently targeting immune cells, lymphoid organs, and the tumor microenvironment, overcoming immune evasion, enhancing immunogenicity, and avoiding lysosomal degradation. However, numerous studies have demonstrated that integrating nanotechnology with immunotherapeutic strategies in vaccine development can overcome these challenges, leading to potent antitumor immune responses and significant progress in the field. This review highlights the critical components of cancer vaccine and nanovaccine strategies for immunomodulatory antitumor therapy. It covers general vaccine strategies, types of vaccines, antigen forms, nanovaccine platforms, challenges faced, potential solutions, and key findings from preclinical and clinical studies, along with future perspectives. To fully unlock the potential of cancer vaccines and nanovaccines, precise immunological monitoring during early-phase trials is essential. This approach will help identify and address obstacles, ultimately expanding the available options for patients who are resistant to conventional cancer immunotherapies.
Collapse
Affiliation(s)
- Kishwor Poudel
- Wellman Center for Photomedicine and Department of Dermatology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Tulasi Vithiananthan
- Wellman Center for Photomedicine and Department of Dermatology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Jong Oh Kim
- College of Pharmacy, Yeungnam University, Gyeongsan, 38541, Republic of Korea
| | - Hensin Tsao
- Wellman Center for Photomedicine and Department of Dermatology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
24
|
Dobutr T, Jangpromma N, Patramanon R, Daduang J, Kulchat S, Areemit J, Lomthaisong K, Daduang S. Screening of aqueous plant extracts for immunomodulatory effects on immune cells and cytokine production: In vitro and in vivo analyses. Heliyon 2025; 11:e42692. [PMID: 40034324 PMCID: PMC11872543 DOI: 10.1016/j.heliyon.2025.e42692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 02/09/2025] [Accepted: 02/12/2025] [Indexed: 03/05/2025] Open
Abstract
This study investigates the immunomodulatory effects of various aqueous plant extracts on immune cells and cytokine production. In vitro, several extracts, including holy basil (Ocimum sanctum), patawali (Tinospora crispa), and Indian borage (Plectranthus amboinicus L.), significantly increased CD3+ T-cell populations, while soap pod (Acacia concinna), garlic (Allium sativum L.), and neem (Azadirachta indica) also boosted CD45RA+ B-cells. In vivo, the extracts had subtle effects on spleen morphology and Peyer's Patches, with milk bush (Euphorbia tirucalli L.) and Indian borage enhancing T-cell responses, while soap pod stimulated B-cells. Additionally, we observed that Neem and milk bush significantly suppressed B-cell populations. Furthermore, cytokine analysis showed that garlic and patawali reduced IL-2, while soap pod, holy basil, and patawali increased TNF-alpha levels. Soap pod also elevated IL-10 and IL-17A, indicating both anti-inflammatory and pro-inflammatory signaling, while patawali induced an increase in IL-4. In conclusion, Thai medicinal plants show strong potential as both immunostimulants and immunosuppressants. They can enhance lymphocyte proliferation, particularly in T-cells, and modulate B-cell populations. Their aqueous extracts play a key role in regulating Th1, Th2, and Th17 cytokine production. Thus, these plants could serve as natural agents and alternative medicines for boosting or modulating immune function.
Collapse
Affiliation(s)
- Theerawat Dobutr
- Department of Biomedical Sciences and Biomedical Engineering, Faculty of Medicine, Prince of Songkla University, Hat Yai, 90000, Thailand
- Protein and Proteomics Research Center for Commercial and Industrial Purposes (ProCCI), Faculty of Science, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Nisachon Jangpromma
- Protein and Proteomics Research Center for Commercial and Industrial Purposes (ProCCI), Faculty of Science, Khon Kaen University, Khon Kaen 40002, Thailand
- Department of Biochemistry, Faculty of Science, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Rina Patramanon
- Protein and Proteomics Research Center for Commercial and Industrial Purposes (ProCCI), Faculty of Science, Khon Kaen University, Khon Kaen 40002, Thailand
- Department of Biochemistry, Faculty of Science, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Jureerut Daduang
- Centre for Research and Development of Medical Diagnostic Laboratories (CMDL), Faculty of Associated Medical Sciences, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Sirinan Kulchat
- Department of Chemistry, Faculty of Science, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Jringjai Areemit
- Division of Pharmaceutical Technology, Faculty of Pharmaceutical Sciences, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Komsorn Lomthaisong
- Protein and Proteomics Research Center for Commercial and Industrial Purposes (ProCCI), Faculty of Science, Khon Kaen University, Khon Kaen 40002, Thailand
- Department of Biochemistry, Faculty of Science, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Sakda Daduang
- Protein and Proteomics Research Center for Commercial and Industrial Purposes (ProCCI), Faculty of Science, Khon Kaen University, Khon Kaen 40002, Thailand
- Division of Pharmacognosy and Toxicology, Faculty of Pharmaceutical Sciences, Khon Kaen University, Khon Kaen 40002, Thailand
| |
Collapse
|
25
|
Nagy MZ, Plaza-Rojas LB, Boucher JC, Kostenko E, Austin AL, Tarhini AA, Chen Z, Du D, Ojwang' AME, Davis J, Obermayer A, Rejniak KA, Shaw TI, Guevara-Patino JA. Effector T cells under hypoxia have an altered transcriptome similar to tumor-stressed T cells found in non-responsive melanoma patients. J Immunother Cancer 2025; 13:e010153. [PMID: 40010774 PMCID: PMC12086921 DOI: 10.1136/jitc-2024-010153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Accepted: 01/26/2025] [Indexed: 02/28/2025] Open
Abstract
BACKGROUND In the tumor microenvironment (TME), hypoxia stands as a significant factor that modulates immune responses, especially those driven by T cells. As T cell-based therapies often fail to work in solid tumors, this study aims to investigate the effects of hypoxia on T cell topo-distribution in the TME, gene expression association with T cell states, and clinical responses in melanoma. METHODS To generate detailed information on tumor oxygenation and T cell accessibility, we used mathematical modeling of human melanoma tissue microarrays that incorporate oxygen supply from vessels, intratumoral diffusion, and cellular uptake. We created tumor maps and derived plots showing the fraction of CD4 and CD8 T cells against the distance to the nearest vessel and oxygen pressure. To assess their function and transcriptional changes caused by hypoxia, effector T cells were generated and cultured under hypoxia (0.5% oxygen) or normoxia (21% oxygen). The T cell hypoxia-transcriptional signature was compared against datasets from msigDB, iATLAS (clinical trials of melanoma patients treated with immune checkpoint inhibitors (ICIs)), ORIEN AVATAR (real-world melanoma patients treated with ICIs), and a single-cell atlas of tumor-infiltrating lymphocytes. RESULTS We made three specific observations: (1) in melanoma T cells preferentially accumulated in oxygenated areas close to blood vessels (50-100 µm from the vasculature in the regions of high oxygen availability) but not in hypoxic areas far from blood vessels. (2) Our analysis confirmed that under hypoxia, T cell functions were significantly reduced compared with normoxic conditions and accompanied by a unique gene signature. Furthermore, this hypoxic gene signature was prevalent in resting and non-activated T cells. Notably and clinically relevant, the hypoxic T cell gene set was found to correlate with reduced overall survival and reduced progression-free survival in melanoma patients, which was more pronounced in non-responder patients undergoing ICI therapy. (3) Finally, compared with a single-cell atlas of tumor-infiltrating T cells, our hypoxia signature aligned with a population of cells at a state termed stress response state (TSTR). CONCLUSIONS Our study highlights the critical role of hypoxia in shaping T cell distribution and its correlation with clinical outcomes in melanoma. We revealed a preferential accumulation of T cells in oxygenated areas. Moreover, hypoxic T cells develop a distinct hypoxic gene signature prevalent in resting, non-activated T cells and TSTR that was also associated with poorer outcomes, particularly pronounced among non-responders to ICIs.
Collapse
Affiliation(s)
- Mate Z Nagy
- Department of Immunology, H Lee Moffitt Cancer Center & Research Institute, Tampa, Florida, USA
| | - Lourdes B Plaza-Rojas
- Department of Immunology, H Lee Moffitt Cancer Center & Research Institute, Tampa, Florida, USA
| | - Justin C Boucher
- Department of Immunology, H Lee Moffitt Cancer Center & Research Institute, Tampa, Florida, USA
| | - Elena Kostenko
- Department of Immunology, H Lee Moffitt Cancer Center & Research Institute, Tampa, Florida, USA
| | - Anna L Austin
- Department of Immunology, H Lee Moffitt Cancer Center & Research Institute, Tampa, Florida, USA
| | - Ahmad A Tarhini
- Departments of Cutaneous Oncology and Immunology, H Lee Moffitt Cancer Center & Research Institute, Tampa, Florida, USA
| | - Zhihua Chen
- Department of Biostatistics and Bioinformatics, H Lee Moffitt Cancer Center & Research Institute, Tampa, Florida, USA
| | - Dongliang Du
- Department of Biostatistics and Bioinformatics, H Lee Moffitt Cancer Center & Research Institute, Tampa, Florida, USA
| | - Awino Maureiq E Ojwang'
- Department of Biostatistics and Bioinformatics, H Lee Moffitt Cancer Center & Research Institute, Tampa, Florida, USA
| | - Joshua Davis
- Department of Biostatistics and Bioinformatics, H Lee Moffitt Cancer Center & Research Institute, Tampa, Florida, USA
| | - Alyssa Obermayer
- Department of Biostatistics and Bioinformatics, H Lee Moffitt Cancer Center & Research Institute, Tampa, Florida, USA
| | - Katarzyna A Rejniak
- Department of Integrated Mathematical Oncology, H Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA
| | - Timothy I Shaw
- Department of Biostatistics and Bioinformatics, H Lee Moffitt Cancer Center & Research Institute, Tampa, Florida, USA
| | - Jose A Guevara-Patino
- Department of Immunology, H Lee Moffitt Cancer Center & Research Institute, Tampa, Florida, USA
| |
Collapse
|
26
|
Giri S, Batra L. Memory Cells in Infection and Autoimmunity: Mechanisms, Functions, and Therapeutic Implications. Vaccines (Basel) 2025; 13:205. [PMID: 40006751 PMCID: PMC11860616 DOI: 10.3390/vaccines13020205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Revised: 02/11/2025] [Accepted: 02/13/2025] [Indexed: 02/27/2025] Open
Abstract
Memory cells are central to the adaptive immune system's ability to remember and respond effectively to previously encountered pathogens. While memory cells provide robust protection against infections, they can also contribute to autoimmunity when regulation fails. Here, we review the roles of memory T and B cells in infection and autoimmunity, focusing on their differentiation, activation, effector functions, and underlying regulatory mechanisms. We elaborate on the precise mechanisms by which memory cells contribute to autoimmune diseases, highlighting insights from current research on how pathogenic memory responses are formed and sustained in autoimmunity. Finally, we explore potential therapeutic strategies aimed at modulating memory cells to prevent or treat autoimmune disorders, including B cell-depleting therapies (e.g., Rituximab), T cell-targeting agents (e.g., Abatacept), and cytokine inhibitors (e.g., IL-17 or IL-23 blockers) that are currently used in diseases such as rheumatoid arthritis, multiple sclerosis, and psoriasis.
Collapse
Affiliation(s)
- Shilpi Giri
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Lalit Batra
- Center for Predictive Medicine for Biodefence and Emerging Infectious Diseases, School of Medicine, University of Louisville, Louisville, KY 40222, USA;
| |
Collapse
|
27
|
Kalimuddin S, Tham CYL, Chan YFZ, Hang SK, Kunasegaran K, Chia A, Chan CYY, Ng DHL, Sim JXY, Tan HC, Syenina A, Ngoh AQ, Hamis NZ, Chew V, Leong YS, Yee JX, Low JG, Chan KR, Ong EZ, Bertoletti A, Ooi EE. Vaccine-induced T cell responses control Orthoflavivirus challenge infection without neutralizing antibodies in humans. Nat Microbiol 2025; 10:374-387. [PMID: 39794472 PMCID: PMC11790491 DOI: 10.1038/s41564-024-01903-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Accepted: 12/04/2024] [Indexed: 01/13/2025]
Abstract
T cells have been identified as correlates of protection in viral infections. However, the level of vaccine-induced T cells needed and the extent to which they alone can control acute viral infection in humans remain uncertain. Here we conducted a double-blind, randomized controlled trial involving vaccination and challenge in 33 adult human volunteers, using the live-attenuated yellow fever (YF17D) and chimeric Japanese encephalitis-YF17D (JE/YF17D) vaccines. Both Orthoflavivirus vaccines share T cell epitopes but have different neutralizing antibody epitopes. The primary objective was to assess the extent to which vaccine-induced T cell responses, independent of neutralizing antibodies, were able to reduce post-challenge viral RNAaemia levels. Secondary objectives included an assessment of surrogate measures of viral control, including post-challenge antibody titres and symptomatic outcomes. YF17D vaccinees had reduced levels of JE/YF17D challenge viraemia, compared with those without previous YF17D vaccination (mean log10(area under the curve genome copies per ml): 2.23 versus 3.22; P = 0.039). Concomitantly, YF17D vaccinees had lower post-JE/YF17D challenge antibody titres that reduced JE virus plaque number by 50%, or PRNT50 (mean log10(PRNT50 titre): 1.87 versus 2.5; P < 0.0001) and symptomatic rates (6% (n = 1/16) versus 53% (n = 9/17), P = 0.007). There were no unexpected safety events. Importantly, after challenge infection, several vaccinees had undetectable viraemia and no seroconversion, even in the absence of neutralizing antibodies. Indeed, high vaccine-induced T cell responses, specifically against the capsid protein, were associated with a level of viral control conventionally interpreted as sterilizing immunity. Our findings reveal the importance of T cells in controlling acute viral infection and suggests a potential correlate of protection against orthoflaviviral infections. ClinicalTrials.gov registration: NCT05568953 .
Collapse
Affiliation(s)
- Shirin Kalimuddin
- Department of Infectious Diseases, Singapore General Hospital, Singapore, Singapore.
- Program in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore, Singapore.
| | - Christine Y L Tham
- Program in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore, Singapore
- Viral Research and Experimental Medicine Centre, SingHealth Duke-NUS Academic Medical Centre, Singapore, Singapore
| | - Yvonne F Z Chan
- Department of Infectious Diseases, Singapore General Hospital, Singapore, Singapore
| | - Shou Kit Hang
- Program in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore, Singapore
| | - Kamini Kunasegaran
- Program in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore, Singapore
| | - Adeline Chia
- Program in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore, Singapore
| | - Candice Y Y Chan
- Department of Infectious Diseases, Singapore General Hospital, Singapore, Singapore
| | - Dorothy H L Ng
- Department of Infectious Diseases, Singapore General Hospital, Singapore, Singapore
| | - Jean X Y Sim
- Department of Infectious Diseases, Singapore General Hospital, Singapore, Singapore
- Department of Infection Prevention and Epidemiology, Singapore General Hospital, Singapore, Singapore
| | - Hwee-Cheng Tan
- Program in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore, Singapore
| | - Ayesa Syenina
- Program in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore, Singapore
- Viral Research and Experimental Medicine Centre, SingHealth Duke-NUS Academic Medical Centre, Singapore, Singapore
| | - An Qi Ngoh
- Program in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore, Singapore
- Viral Research and Experimental Medicine Centre, SingHealth Duke-NUS Academic Medical Centre, Singapore, Singapore
| | - Noor Zayanah Hamis
- Program in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore, Singapore
- Viral Research and Experimental Medicine Centre, SingHealth Duke-NUS Academic Medical Centre, Singapore, Singapore
| | - Valerie Chew
- Program in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore, Singapore
- Viral Research and Experimental Medicine Centre, SingHealth Duke-NUS Academic Medical Centre, Singapore, Singapore
| | - Yan Shan Leong
- Program in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore, Singapore
- Viral Research and Experimental Medicine Centre, SingHealth Duke-NUS Academic Medical Centre, Singapore, Singapore
| | - Jia Xin Yee
- Program in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore, Singapore
- Viral Research and Experimental Medicine Centre, SingHealth Duke-NUS Academic Medical Centre, Singapore, Singapore
| | - Jenny G Low
- Department of Infectious Diseases, Singapore General Hospital, Singapore, Singapore
- Program in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore, Singapore
- Viral Research and Experimental Medicine Centre, SingHealth Duke-NUS Academic Medical Centre, Singapore, Singapore
| | - Kuan Rong Chan
- Program in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore, Singapore
| | - Eugenia Z Ong
- Program in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore, Singapore
- Viral Research and Experimental Medicine Centre, SingHealth Duke-NUS Academic Medical Centre, Singapore, Singapore
| | - Antonio Bertoletti
- Program in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore, Singapore
- Singapore Immunology Network, A*STAR Singapore, Singapore, Singapore
| | - Eng Eong Ooi
- Program in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore, Singapore.
- Viral Research and Experimental Medicine Centre, SingHealth Duke-NUS Academic Medical Centre, Singapore, Singapore.
- Department of Translational Clinical Research, Singapore General Hospital, Singapore, Singapore.
- Saw Swee Hock School of Public Health, National University of Singapore, Singapore, Singapore.
| |
Collapse
|
28
|
Liu C, Chen Y, Li X, Bai Z, Jiang M, Sheng D, Zou W, Huang R, Huang Q, Wang F, Zhu J, Sun H, Liu B, Li Z, Sun B. Pre-immunotherapy alters stereotactic ablative radiotherapy-induced systemic T cell responses in early-stage NSCLC. Cancer Immunol Immunother 2025; 74:80. [PMID: 39891774 PMCID: PMC11787101 DOI: 10.1007/s00262-024-03935-8] [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: 07/08/2024] [Accepted: 12/27/2024] [Indexed: 02/03/2025]
Abstract
BACKGROUND Stereotactic ablative radiotherapy (SABR) is thought to activate T cell responses in patients with cancer, leading to its combination with immunotherapy and chemotherapy for treatment of non-small-cell lung cancer (NSCLC). Here, we aimed to provide a high-resolution transcriptomic profiling of the systemic T cell response following SABR, with or without preceding immunotherapy/chemotherapy. METHODS We conducted single-cell RNA and T cell receptor (TCR) sequencing of T cells from peripheral blood of seven patients with early-stage NSCLC taken pre- and post-SABR without or with prior immunotherapy and chemotherapy (icSABR). Other flow cytometry, single-cell RNA-seq data and bulk RNA-seq data were used to validate the results. RESULTS We uncovered distinct T cell response patterns induced by these treatments: while terminal effector CD8+ T cells showed increased cytotoxic and inhibitory scores, and upregulated immune-activated pathways post-SABR, the reverse responses occurred post-icSABR. Furthermore, the proportion of large T cell clones increased and single clone decreased post-SABR, while the opposite was seen post-icSABR. Of note, both SABR and icSABR largely changed TCR clonotypes, which were mainly large clones post-SABR but single clone post-icSABR, and predominantly from terminal effector CD8+ T cells and T helper cells, respectively. CONCLUSIONS These findings reveal a complex interplay between SABR and immunotherapy, with potentially valuable implications for treatment strategies involving SABR and immunotherapy to induce systemic T cell responses for tumor eradication in patients with NSCLC.
Collapse
Affiliation(s)
- Chao Liu
- Department of Radiation Oncology, Peking University First Hospital, Beijing, 100034, China
| | - Yanjuan Chen
- Department of Geriatrics and Division of Rheumatology and Research, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen, 518020, China
| | - Xiaohui Li
- Department of Medical Oncology, Peking University First Hospital, Beijing, 100034, China
| | - Zhijie Bai
- State Key Laboratory of Experimental Hematology, Institute of Hematology, Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100071, China
| | - Meilin Jiang
- Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, School of Medicine, Jinan University, Guangzhou, 510632, Guangdong, China
| | - Dongsheng Sheng
- Department of Thoracic Surgery, Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100071, China
| | - Wenxue Zou
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250117, China
| | - Rui Huang
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250117, China
| | - Qingyu Huang
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250117, China
| | - Fuhao Wang
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250117, China
| | - Jingyang Zhu
- Department of Radiation Oncology, Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100071, China
| | - Huiru Sun
- Department of Radiation Oncology, Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100071, China
| | - Bing Liu
- State Key Laboratory of Experimental Hematology, Institute of Hematology, Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100071, China.
| | - Zongcheng Li
- State Key Laboratory of Experimental Hematology, Institute of Hematology, Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100071, China.
| | - Bing Sun
- Department of Radiation Oncology, Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100071, China.
| |
Collapse
|
29
|
Calderón-Pérez B, Núñez-Muñoz LA, Trejo-Ayala LL, Rosales-García VH, Chávez-Álvarez BE, Vargas-Hernández BY, Ramírez-Pool JA, Ruiz-Medrano R, Xoconostle-Cázares B. Immunogenicity of a multivalent protein subunit vaccine based on non-glycosylated RBD antigens of SARS-cov-2 and its variants. Virology 2025; 603:110380. [PMID: 39731906 DOI: 10.1016/j.virol.2024.110380] [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: 07/29/2024] [Revised: 12/09/2024] [Accepted: 12/24/2024] [Indexed: 12/30/2024]
Abstract
COVID-19 infections continue due to accessibility barriers to vaccines and the emergence of SARS-CoV-2 variants. An effective, safe, accessible, and broad-spectrum vaccine is still needed to control the disease. We developed a multivalent protein subunit vaccine comprising antigens designed from a non-N-glycosylated region of the receptor-binding domain of the spike protein of SARS-CoV-2. We combined a previously developed antigen based on the Wuhan original viral strain, and a site-mutated antigen based on several variants including Alpha, Beta, Gamma, Eta, Iota, Theta, Zeta, Mu and Omicron. The recombinant antigens were expressed in a prokaryotic system and the immunogenicity of the multivalent vaccine was tested in a mouse model. The evaluation of the subunit vaccine candidate, incorporating different variant-based multivalent recombinant antigens from non-glycosylated regions of the RBD, demonstrated a favorable safety profile, significant immunogenicity, and potent neutralizing activity, collectively supporting its potential efficacy and safety for further development.
Collapse
MESH Headings
- Spike Glycoprotein, Coronavirus/immunology
- Spike Glycoprotein, Coronavirus/genetics
- Animals
- SARS-CoV-2/immunology
- SARS-CoV-2/genetics
- COVID-19 Vaccines/immunology
- COVID-19 Vaccines/administration & dosage
- COVID-19 Vaccines/genetics
- Mice
- Vaccines, Subunit/immunology
- Vaccines, Subunit/genetics
- Vaccines, Subunit/administration & dosage
- Antibodies, Neutralizing/immunology
- Antibodies, Neutralizing/blood
- Antibodies, Viral/blood
- Antibodies, Viral/immunology
- COVID-19/prevention & control
- COVID-19/immunology
- COVID-19/virology
- Antigens, Viral/immunology
- Antigens, Viral/genetics
- Immunogenicity, Vaccine
- Humans
- Female
- Mice, Inbred BALB C
- Glycosylation
Collapse
Affiliation(s)
- Berenice Calderón-Pérez
- Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV), Departamento de Biotecnología y Bioingeniería, Av. Instituto Politécnico Nacional 2508, Mexico City, 07360, Mexico.
| | - Leandro Alberto Núñez-Muñoz
- Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV), Departamento de Biotecnología y Bioingeniería, Av. Instituto Politécnico Nacional 2508, Mexico City, 07360, Mexico.
| | - Lady Laura Trejo-Ayala
- Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV), Departamento de Biotecnología y Bioingeniería, Av. Instituto Politécnico Nacional 2508, Mexico City, 07360, Mexico.
| | | | | | - Brenda Yazmín Vargas-Hernández
- Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV), Departamento de Biotecnología y Bioingeniería, Av. Instituto Politécnico Nacional 2508, Mexico City, 07360, Mexico.
| | - José Abrahán Ramírez-Pool
- Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV), Departamento de Biotecnología y Bioingeniería, Av. Instituto Politécnico Nacional 2508, Mexico City, 07360, Mexico.
| | - Roberto Ruiz-Medrano
- Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV), Departamento de Biotecnología y Bioingeniería, Av. Instituto Politécnico Nacional 2508, Mexico City, 07360, Mexico; CINVESTAV, Programa de Doctorado Transdisciplinario en Desarrollo Científico y Tecnológico para la Sociedad, Mexico.
| | - Beatriz Xoconostle-Cázares
- Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV), Departamento de Biotecnología y Bioingeniería, Av. Instituto Politécnico Nacional 2508, Mexico City, 07360, Mexico; CINVESTAV, Programa de Doctorado Transdisciplinario en Desarrollo Científico y Tecnológico para la Sociedad, Mexico.
| |
Collapse
|
30
|
Cozac-Szőke AR, Cozac DA, Negovan A, Tinca AC, Vilaia A, Cocuz IG, Sabău AH, Niculescu R, Chiorean DM, Tomuț AN, Cotoi OS. Immune Cell Interactions and Immune Checkpoints in the Tumor Microenvironment of Gastric Cancer. Int J Mol Sci 2025; 26:1156. [PMID: 39940924 PMCID: PMC11818890 DOI: 10.3390/ijms26031156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Revised: 01/26/2025] [Accepted: 01/27/2025] [Indexed: 02/16/2025] Open
Abstract
Gastric cancer (GC) ranks as the fifth most prevalent malignant neoplasm globally, with an increased death rate despite recent advancements in research and therapeutic options. Different molecular subtypes of GC have distinct interactions with the immune system, impacting the tumor microenvironment (TME), prognosis, and reaction to immunotherapy. Tumor-infiltrating lymphocytes (TILs) in the TME are crucial for preventing tumor growth and metastasis, as evidenced by research showing that patients with GC who have a significant density of TILs have better survival rates. But cancer cells have evolved a variety of mechanisms to evade immune surveillance, both sialic acid-binding immunoglobulin-like lectin 15 (Siglec-15) and Programmed Death-Ligand 1 (PD-L1) playing a pivotal role in the development of an immunosuppressive TME. They prevent T cell activation and proliferation resulting in a decrease in the immune system's capacity to recognize and eliminate malignant cells. These immune checkpoint molecules function via different but complementary mechanisms, the expression of Siglec-15 being mutually exclusive with PD-L1 and, therefore, providing a different therapeutic approach. The review explores how TILs affect tumor growth and patient outcomes in GC, with particular emphasis on their interactions within the TME and potential targeting of the PD-L1 and Siglec-15 pathways for immunotherapy.
Collapse
Affiliation(s)
- Andreea-Raluca Cozac-Szőke
- Doctoral School of Medicine and Pharmacy, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Targu Mures, 540142 Targu Mures, Romania; (A.-R.C.-S.); (A.H.S.); (R.N.); (D.M.C.)
- Pathophysiology Department, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures, 540142 Targu Mures, Romania; (A.C.T.); (I.-G.C.); (O.S.C.)
- Pathology Department, Mures Clinical County Hospital, 540011 Targu Mures, Romania
| | - Dan Alexandru Cozac
- Doctoral School of Medicine and Pharmacy, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Targu Mures, 540142 Targu Mures, Romania; (A.-R.C.-S.); (A.H.S.); (R.N.); (D.M.C.)
- Emergency Institute for Cardiovascular Diseases and Transplantation Targu Mures, 540142 Targu Mures, Romania
| | - Anca Negovan
- Department of Clinical Science-Internal Medicine, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures, 540142 Targu Mures, Romania;
| | - Andreea Cătălina Tinca
- Pathophysiology Department, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures, 540142 Targu Mures, Romania; (A.C.T.); (I.-G.C.); (O.S.C.)
- Pathology Department, Mures Clinical County Hospital, 540011 Targu Mures, Romania
| | - Alexandra Vilaia
- Department of Infectious Diseases I, Doctoral School of Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania;
| | - Iuliu-Gabriel Cocuz
- Pathophysiology Department, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures, 540142 Targu Mures, Romania; (A.C.T.); (I.-G.C.); (O.S.C.)
- Pathology Department, Mures Clinical County Hospital, 540011 Targu Mures, Romania
| | - Adrian Horațiu Sabău
- Doctoral School of Medicine and Pharmacy, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Targu Mures, 540142 Targu Mures, Romania; (A.-R.C.-S.); (A.H.S.); (R.N.); (D.M.C.)
- Pathophysiology Department, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures, 540142 Targu Mures, Romania; (A.C.T.); (I.-G.C.); (O.S.C.)
- Pathology Department, Mures Clinical County Hospital, 540011 Targu Mures, Romania
| | - Raluca Niculescu
- Doctoral School of Medicine and Pharmacy, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Targu Mures, 540142 Targu Mures, Romania; (A.-R.C.-S.); (A.H.S.); (R.N.); (D.M.C.)
- Pathophysiology Department, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures, 540142 Targu Mures, Romania; (A.C.T.); (I.-G.C.); (O.S.C.)
- Pathology Department, Mures Clinical County Hospital, 540011 Targu Mures, Romania
| | - Diana Maria Chiorean
- Doctoral School of Medicine and Pharmacy, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Targu Mures, 540142 Targu Mures, Romania; (A.-R.C.-S.); (A.H.S.); (R.N.); (D.M.C.)
- Pathophysiology Department, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures, 540142 Targu Mures, Romania; (A.C.T.); (I.-G.C.); (O.S.C.)
- Pathology Department, Mures Clinical County Hospital, 540011 Targu Mures, Romania
| | - Alexandru Nicușor Tomuț
- Faculty of Medicine, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures, 540142 Targu Mures, Romania;
| | - Ovidiu Simion Cotoi
- Pathophysiology Department, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures, 540142 Targu Mures, Romania; (A.C.T.); (I.-G.C.); (O.S.C.)
- Pathology Department, Mures Clinical County Hospital, 540011 Targu Mures, Romania
| |
Collapse
|
31
|
Viramontes KM, Thone MN, DeRogatis JM, Neubert EN, Henriquez ML, De La Torre JJ, Tinoco R. Prion protein modulation of virus-specific T cell differentiation and function during acute viral infection. Immunohorizons 2025; 9:vlae002. [PMID: 39846843 PMCID: PMC11841969 DOI: 10.1093/immhor/vlae002] [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: 06/03/2024] [Accepted: 10/11/2024] [Indexed: 01/24/2025] Open
Abstract
The differentiation and functionality of virus-specific T cells during acute viral infections are crucial for establishing long-term protective immunity. While numerous molecular regulators impacting T cell responses have been uncovered, the role of cellular prion proteins (PrPc) remains underexplored. Here, we investigated the impact of PrPc deficiency on the differentiation and function of virus-specific T cells using the lymphocytic choriomeningitis virus (LCMV) Armstrong acute infection model. Our findings reveal that Prnp-/- mice exhibit a robust expansion of virus-specific CD8+ T cells, with similar activation profiles as wild-type mice during the early stages of infection. However, Prnp-/- mice had higher frequencies and numbers of virus-specific memory CD8+ T cells, along with altered differentiation profiles characterized by increased central and effector memory subsets. Despite similar proliferation rates early during infection, Prnp-/- memory CD8+ T cells had decreased proliferation compared with their wild-type counterparts. Additionally, Prnp-/- mice had higher numbers of cytokine-producing memory CD8+ T cells, indicating a more robust functional response. Furthermore, Prnp-/- mice had increased virus-specific CD4+ T cell responses, suggesting a broader impact of PrPc deficiency on T cell immunity. These results unveil a previously unrecognized role for PrPc in regulating the differentiation, proliferation, and functionality of virus-specific T cells, providing valuable insights into immune system regulation by prion proteins during viral infections.
Collapse
Affiliation(s)
- Karla M Viramontes
- Center for Virus Research, Chao Family Comprehensive Cancer Center, Department of Molecular Biology and Biochemistry, Charlie Dunlop School of Biological Sciences, University of California, Irvine, Irvine, CA, United States
| | - Melissa N Thone
- Center for Virus Research, Chao Family Comprehensive Cancer Center, Department of Molecular Biology and Biochemistry, Charlie Dunlop School of Biological Sciences, University of California, Irvine, Irvine, CA, United States
| | - Julia M DeRogatis
- Center for Virus Research, Chao Family Comprehensive Cancer Center, Department of Molecular Biology and Biochemistry, Charlie Dunlop School of Biological Sciences, University of California, Irvine, Irvine, CA, United States
| | - Emily N Neubert
- Center for Virus Research, Chao Family Comprehensive Cancer Center, Department of Molecular Biology and Biochemistry, Charlie Dunlop School of Biological Sciences, University of California, Irvine, Irvine, CA, United States
| | - Monique L Henriquez
- Center for Virus Research, Chao Family Comprehensive Cancer Center, Department of Molecular Biology and Biochemistry, Charlie Dunlop School of Biological Sciences, University of California, Irvine, Irvine, CA, United States
| | - Jamie-Jean De La Torre
- Center for Virus Research, Chao Family Comprehensive Cancer Center, Department of Molecular Biology and Biochemistry, Charlie Dunlop School of Biological Sciences, University of California, Irvine, Irvine, CA, United States
| | - Roberto Tinoco
- Center for Virus Research, Chao Family Comprehensive Cancer Center, Department of Molecular Biology and Biochemistry, Charlie Dunlop School of Biological Sciences, University of California, Irvine, Irvine, CA, United States
| |
Collapse
|
32
|
Bhandarkar V, Dinter T, Spranger S. Architects of immunity: How dendritic cells shape CD8 + T cell fate in cancer. Sci Immunol 2025; 10:eadf4726. [PMID: 39823318 PMCID: PMC11970844 DOI: 10.1126/sciimmunol.adf4726] [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: 03/24/2023] [Accepted: 12/16/2024] [Indexed: 01/19/2025]
Abstract
Immune responses against cancer are dominated by T cell exhaustion and dysfunction. Recent advances have underscored the critical role of early priming interactions in establishing T cell fates. In this review, we explore the importance of dendritic cell (DC) signals in specifying CD8+ T cell fates in cancer, drawing on insights from acute and chronic viral infection models. We highlight the role of DCs in lymph nodes and tumors in maintaining stem-like CD8+ T cells, which are critical for durable antitumor immune responses. Understanding how CD8+ T cell fates are determined will enable the rational design of immunotherapies, particularly therapeutic cancer vaccines, that can modulate DC-T cell interactions to generate beneficial CD8+ T cell fates.
Collapse
Affiliation(s)
- Vidit Bhandarkar
- Koch Institute at MIT, Cambridge, MA 02139, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Teresa Dinter
- Koch Institute at MIT, Cambridge, MA 02139, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Stefani Spranger
- Koch Institute at MIT, Cambridge, MA 02139, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| |
Collapse
|
33
|
Pradhan SS, Balena V, Bera BC, Anand T, Khetmalis R, Madhwal A, Kandasamy S, Pavulraj S, Bernela M, Mor P, Tripathi BN, Virmani N. Multiple Gene Deletion Mutants of Equine Herpesvirus 1 Exhibit Strong Protective Efficacy Against Wild Virus Challenge in a Murine Model. Vaccines (Basel) 2025; 13:45. [PMID: 39852824 PMCID: PMC11768829 DOI: 10.3390/vaccines13010045] [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: 10/24/2024] [Revised: 11/22/2024] [Accepted: 11/26/2024] [Indexed: 01/26/2025] Open
Abstract
BACKGROUND Equine herpesvirus type 1 (EHV1) is a ubiquitous viral pathogen infecting the equine population worldwide. EHV1 infection causes respiratory illness, abortion, neonatal foal mortality, and myeloencephalopathy. The currently available modified live EHV1 vaccines have safety and efficacy limitations. The two mutant EHV1 viruses (vToH-DMV (∆IR6/gE) and vToH-QMV (∆IR6/UL43/gE/UL56)), generated by the deletion of genes responsible for virulence (gE and IR6) and immunosuppression (uL43 and uL56), have been previously characterized by our group and found to generate good immune responses. The present study aimed to determine the safety and protective efficacy of the above mutants against a virulent EHV1 challenge in a murine model. METHODS BALB/c mice were intranasally immunized with a live vToH-QMV or vToH-DMV vaccine. Intranasal booster immunization was given at 14 days post-vaccination (dpv). Both mutants induced an optimal level of EHV1-specific humoral and cell-mediated immune responses, as determined by virus neutralization assay, ELISA, and immunophenotyping. At 35 dpv, the mice were intranasally challenged with wild-type EHV1 (vRaj strain). RESULTS Amongst the two mutants, vToH-QMV induced a better immune response than the vToH-DMV vaccine. Furthermore, vToH-QMV provided good protection in mice against the virulent challenge. It specifically exhibited less severe clinical disease in terms of clinical signs, body weight reduction, and gross and histopathological lung lesions accompanied by early virus clearance. CONCLUSIONS These studies are suggestive of vToH-QMV EHV1 being a potential vaccine candidate against EHV1 infection, which needs to be finally tested in the main host, i.e., horses.
Collapse
Affiliation(s)
- Stephanie S. Pradhan
- ICAR-National Research Centre on Equines, Sirsa Road, Hisar 125001, Haryana, India; (S.S.P.); (A.M.); (S.K.); (P.M.)
- Division of Pathology, ICAR-Indian Veterinary Research Institute, Bareilly 243122, Uttar Pradesh, India
| | - Vekataramireddy Balena
- ICAR-National Research Centre on Equines, Sirsa Road, Hisar 125001, Haryana, India; (S.S.P.); (A.M.); (S.K.); (P.M.)
- Division of Pathology, ICAR-Indian Veterinary Research Institute, Bareilly 243122, Uttar Pradesh, India
| | - Bidhan Chandra Bera
- ICAR-National Research Centre on Equines, Sirsa Road, Hisar 125001, Haryana, India; (S.S.P.); (A.M.); (S.K.); (P.M.)
| | - Taruna Anand
- ICAR-National Research Centre on Equines, Sirsa Road, Hisar 125001, Haryana, India; (S.S.P.); (A.M.); (S.K.); (P.M.)
| | - Rhushikesh Khetmalis
- ICAR-National Research Centre on Equines, Sirsa Road, Hisar 125001, Haryana, India; (S.S.P.); (A.M.); (S.K.); (P.M.)
- Division of Pathology, ICAR-Indian Veterinary Research Institute, Bareilly 243122, Uttar Pradesh, India
| | - Aashwina Madhwal
- ICAR-National Research Centre on Equines, Sirsa Road, Hisar 125001, Haryana, India; (S.S.P.); (A.M.); (S.K.); (P.M.)
- Division of Pathology, ICAR-Indian Veterinary Research Institute, Bareilly 243122, Uttar Pradesh, India
| | - Supriya Kandasamy
- ICAR-National Research Centre on Equines, Sirsa Road, Hisar 125001, Haryana, India; (S.S.P.); (A.M.); (S.K.); (P.M.)
- Division of Pathology, ICAR-Indian Veterinary Research Institute, Bareilly 243122, Uttar Pradesh, India
| | - Selvaraj Pavulraj
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, USA
| | - Manju Bernela
- ICAR-National Research Centre on Equines, Sirsa Road, Hisar 125001, Haryana, India; (S.S.P.); (A.M.); (S.K.); (P.M.)
| | - Priya Mor
- ICAR-National Research Centre on Equines, Sirsa Road, Hisar 125001, Haryana, India; (S.S.P.); (A.M.); (S.K.); (P.M.)
| | | | - Nitin Virmani
- ICAR-National Research Centre on Equines, Sirsa Road, Hisar 125001, Haryana, India; (S.S.P.); (A.M.); (S.K.); (P.M.)
| |
Collapse
|
34
|
Hosseini SA, Nasab NK, Kargozar S, Wang AZ. Advanced biomaterials and scaffolds for cancer immunotherapy. BIOMATERIALS FOR PRECISION CANCER MEDICINE 2025:377-424. [DOI: 10.1016/b978-0-323-85661-4.00016-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
35
|
Han C, Xiao S, Xing Z, Xu X, Wang M, Han X, Adeli M, Qiu L, Ye L, Cheng C. NADPH Oxidases-Inspired Reactive Oxygen Biocatalysts with Electron-Rich Pt Sites to Potently Amplify Immune Checkpoint Blockade Therapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2407644. [PMID: 39400421 DOI: 10.1002/adma.202407644] [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: 05/29/2024] [Revised: 07/19/2024] [Indexed: 10/15/2024]
Abstract
Clinical immune checkpoint blockade (ICB)-based immunotherapy of malignant tumors only elicits durable responses in a minority of patients, primarily due to the highly immunosuppressive tumor microenvironment. Although inducing immunogenic cell death (ICD) through reactive oxygen biocatalyst represents an attractive therapeutic strategy to amplify ICB, currently reported biocatalysts encounter insurmountable challenges in achieving high ROS-generating activity to induce potent ICD. Here, inspired by the natural catalytic characteristics of NADPH oxidases, the design of efficient, robust, and electron-rich Pt-based redox centers on the non-stoichiometric W18O49 substrates (Pt─WOx) to serve as bioinspired reactive oxygen biocatalysts to potently activate the ICD, which eventually enhance cancer immune responses and amplifies the ICB-based immunotherapy is reported. These studies demonstrate that the Pt─WOx exhibits rapid electron transfer capability and can promote the formation of electron-rich and low oxophilic Pt redox centers for superior reactive oxygen biocatalysis, which enables the Pt─WOx-based inducers to trigger endoplasmic reticulum stress directly and stimulate immune responses potently for amplifying the anti-PD-L1-based ICB therapy. This bioinspired design provides a straightforward strategy to engineer efficient, robust, and electron-rich reactive oxygen biocatalysts and also opens up a new avenue to create efficient ICD inducers for primary/metastatic tumor treatments.
Collapse
Affiliation(s)
- Chuyi Han
- Department of Endodontics, Department of Orthodontics, State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Sutong Xiao
- Department of Endodontics, Department of Orthodontics, State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
- Department of Medical Ultrasound, West China Hospital, Sichuan University, Chengdu, 610041, China
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, 610065, China
| | - Zhenyu Xing
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, 610065, China
| | - Xiaohui Xu
- Department of Medical Ultrasound, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Mao Wang
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, 610065, China
| | - Xianglong Han
- Department of Endodontics, Department of Orthodontics, State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Mohsen Adeli
- Institute of Chemistry and Biochemistry, Freie Universitat Berlin, Takustr. 3, 14195, Berlin, Germany
- Department of Organic Chemistry, Faculty of Chemistry, Lorestan University, Khorramabad, 68137-17133, Iran
| | - Li Qiu
- Department of Medical Ultrasound, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Ling Ye
- Department of Endodontics, Department of Orthodontics, State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Chong Cheng
- Department of Endodontics, Department of Orthodontics, State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, 610065, China
| |
Collapse
|
36
|
Pichler AS, Amador C, Fujimoto A, Takeuchi K, de Jong D, Iqbal J, Staber PB. Advances in peripheral T cell lymphomas: pathogenesis, genetic landscapes and emerging therapeutic targets. Histopathology 2025; 86:119-133. [PMID: 39679758 DOI: 10.1111/his.15376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
Peripheral T cell lymphomas (PTCLs) are a biologically diverse and aggressive group of non-Hodgkin lymphomas that originate from mature T cells, often presenting with complex clinical and morphological features. This review explores the challenges in diagnosing and classifying PTCLs, focusing on the intricate biology of the more common nodal entities. Advances in molecular diagnostics, such as mutational and gene expression profiling, have improved our understanding. However, the rarity and morphological variability of PTCLs continue to complicate the definition of biologically and clinically meaningful entities, as well as the application of current diagnoses in daily practice; these advancements have not yet translated into improved clinical outcomes. Standard therapies fail in most cases and lead to poor prognoses, highlighting the urgent need for improved therapeutic strategies. Precise characterisation of PTCL advances refined classification and supports the development of more targeted and effective treatments. Recent approaches have focused on biology-based risk stratification, either within specific entities or in an entity-agnostic manner. This development aims for improved treatment selection or even personalised treatment based on genetic, epigenetic and functional profiles.
Collapse
Affiliation(s)
- Alexander S Pichler
- Division of Hematology and Hemostaseology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Catalina Amador
- Department of Pathology and Laboratory Medicine, University of Miami, Miami, Florida, USA
| | - Ayumi Fujimoto
- Division of Pathology, Cancer Institute, Japanese Foundation of Cancer Research, Tokyo, Japan
| | - Kengo Takeuchi
- Division of Pathology, Cancer Institute, Japanese Foundation of Cancer Research, Tokyo, Japan
| | - Daphne de Jong
- Department of Pathology, the Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Javeed Iqbal
- University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Philipp B Staber
- Division of Hematology and Hemostaseology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
37
|
Wang L, Song BS, Poojary R, Xiong X, Ren X, Yang JM, Song J. eEF-2K Deficiency Boosts the Virus-Specific Effector CD8 + T Cell Responses During Viral Infection. Viruses 2024; 17:26. [PMID: 39861816 PMCID: PMC11768472 DOI: 10.3390/v17010026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 12/24/2024] [Accepted: 12/26/2024] [Indexed: 01/27/2025] Open
Abstract
In this study, we revealed a critical role of eukaryotic elongation factor-2 kinase (eEF-2K), a negative regulator of protein synthesis, in regulating T cells during vaccinia virus (VACV) infection. We found that eEF-2K-deficient (eEF-2K⁻/⁻) mice exhibited a significantly higher proportion of VACV-specific effector CD8+ T cells without compromising the development of VACV-specific memory CD8+ T cells. RNA sequencing demonstrated that eEF-2K⁻/⁻ VACV-specific effector CD8+ T cells had enhanced functionality, which improves their capacity to combat viral infection during the effector phase. Moreover, we identified tumor necrosis factor receptor-associated factor 3 (TRAF3) as a critical mediator of the stronger antiviral response observed in eEF-2K⁻/⁻ effector CD8+ T cells. These findings suggest that targeting eEF-2K may provide a novel strategy to augmenting effector CD8+ T cell responses against viral infections.
Collapse
Affiliation(s)
- Liqing Wang
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, Bryan, TX 77807, USA (R.P.); (X.X.)
| | - Benny Shone Song
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, Bryan, TX 77807, USA (R.P.); (X.X.)
| | - Rayansh Poojary
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, Bryan, TX 77807, USA (R.P.); (X.X.)
| | - Xiaofang Xiong
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, Bryan, TX 77807, USA (R.P.); (X.X.)
| | - Xingcong Ren
- Department of Toxicology and Cancer Biology, University of Kentucky College of Medicine, Lexington, KY 40536, USA
| | - Jin-Ming Yang
- Department of Toxicology and Cancer Biology, University of Kentucky College of Medicine, Lexington, KY 40536, USA
| | - Jianxun Song
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, Bryan, TX 77807, USA (R.P.); (X.X.)
| |
Collapse
|
38
|
Eshaq AM, Flanagan TW, Ba Abbad AA, Makarem ZAA, Bokir MS, Alasheq AK, Al Asheikh SA, Almashhor AM, Binyamani F, Al-Amoudi WA, Bawzir AS, Haikel Y, Megahed M, Hassan M. Immune Checkpoint Inhibitor-Associated Cutaneous Adverse Events: Mechanisms of Occurrence. Int J Mol Sci 2024; 26:88. [PMID: 39795946 PMCID: PMC11719825 DOI: 10.3390/ijms26010088] [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: 11/17/2024] [Revised: 12/19/2024] [Accepted: 12/20/2024] [Indexed: 01/13/2025] Open
Abstract
Immunotherapy, particularly that based on blocking checkpoint proteins in many tumors, including melanoma, Merkel cell carcinoma, non-small cell lung cancer (NSCLC), triple-negative breast (TNB cancer), renal cancer, and gastrointestinal and endometrial neoplasms, is a therapeutic alternative to chemotherapy. Immune checkpoint inhibitor (ICI)-based therapies have the potential to target different pathways leading to the destruction of cancer cells. Although ICIs are an effective treatment strategy for patients with highly immune-infiltrated cancers, the development of different adverse effects including cutaneous adverse effects during and after the treatment with ICIs is common. ICI-associated cutaneous adverse effects include mostly inflammatory and bullous dermatoses, as well as severe cutaneous side reactions such as rash or inflammatory dermatitis encompassing erythema multiforme; lichenoid, eczematous, psoriasiform, and morbilliform lesions; and palmoplantar erythrodysesthesia. The development of immunotherapy-related adverse effects is a consequence of ICIs' unique molecular action that is mainly mediated by the activation of cytotoxic CD4+/CD8+ T cells. ICI-associated cutaneous disorders are the most prevalent effects induced in response to anti-programmed cell death 1 (PD-1), anti-cytotoxic T-lymphocyte-associated antigen-4 (CTLA-4), and anti-programmed cell death ligand 1 (PD-L1) agents. Herein, we will elucidate the mechanisms regulating the occurrence of cutaneous adverse effects following treatment with ICIs.
Collapse
Affiliation(s)
- Abdulaziz M. Eshaq
- Department of Epidemiology and Biostatstics, Milken Institute School of Public Health, George Washington University Washington, Washington, DC 20052, USA;
- Research Laboratory of Surgery-Oncology, Department of Surgery, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Thomas W. Flanagan
- Department of Pharmacology and Experimental Therapeutics, LSU Health Sciences Center, New Orleans, LA 70112, USA;
| | - Abdulqader A. Ba Abbad
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (A.A.B.A.); (Z.A.A.M.); (M.S.B.); (A.K.A.); (A.M.A.); (F.B.); (W.A.A.-A.)
| | - Zain Alabden A. Makarem
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (A.A.B.A.); (Z.A.A.M.); (M.S.B.); (A.K.A.); (A.M.A.); (F.B.); (W.A.A.-A.)
| | - Mohammed S. Bokir
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (A.A.B.A.); (Z.A.A.M.); (M.S.B.); (A.K.A.); (A.M.A.); (F.B.); (W.A.A.-A.)
| | - Ahmed K. Alasheq
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (A.A.B.A.); (Z.A.A.M.); (M.S.B.); (A.K.A.); (A.M.A.); (F.B.); (W.A.A.-A.)
| | - Sara A. Al Asheikh
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (A.A.B.A.); (Z.A.A.M.); (M.S.B.); (A.K.A.); (A.M.A.); (F.B.); (W.A.A.-A.)
| | - Abdullah M. Almashhor
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (A.A.B.A.); (Z.A.A.M.); (M.S.B.); (A.K.A.); (A.M.A.); (F.B.); (W.A.A.-A.)
| | - Faroq Binyamani
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (A.A.B.A.); (Z.A.A.M.); (M.S.B.); (A.K.A.); (A.M.A.); (F.B.); (W.A.A.-A.)
| | - Waleed A. Al-Amoudi
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (A.A.B.A.); (Z.A.A.M.); (M.S.B.); (A.K.A.); (A.M.A.); (F.B.); (W.A.A.-A.)
| | - Abdulaziz S. Bawzir
- Department of Radiology, King Saud Medical City, Riyadh 11533, Saudi Arabia;
| | - Youssef Haikel
- Institut National de la Santé et de la Recherche Médicale, University of Strasbourg, 67000 Strasbourg, France;
- Department of Operative Dentistry and Endodontics, Dental Faculty, University of Strasbourg, 67000 Strasbourg, France
- Pôle de Médecine et Chirurgie Bucco-Dentaire, Hôpital Civil, Hôpitaux Universitaire de Strasbourg, 67000 Strasbourg, France
| | - Mossad Megahed
- Clinic of Dermatology, University Hospital of Aachen, 52074 Aachen, Germany;
| | - Mohamed Hassan
- Research Laboratory of Surgery-Oncology, Department of Surgery, Tulane University School of Medicine, New Orleans, LA 70112, USA
- Institut National de la Santé et de la Recherche Médicale, University of Strasbourg, 67000 Strasbourg, France;
- Department of Operative Dentistry and Endodontics, Dental Faculty, University of Strasbourg, 67000 Strasbourg, France
| |
Collapse
|
39
|
Wei Y, Ma HK, Wong ME, Papasavvas E, Konnikova L, Tebas P, Morgenstern R, Montaner LJ, Ho YC. BACH2-driven tissue resident memory programs promote HIV-1 persistence. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.16.628794. [PMID: 39763845 PMCID: PMC11702684 DOI: 10.1101/2024.12.16.628794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/12/2025]
Abstract
Transcription repressor BACH2 redirects short-lived terminally differentiated effector into long-lived memory cells. We postulate that BACH2-mediated long-lived memory programs promote HIV-1 persistence in gut CD4+ T cells. We coupled single-cell DOGMA-seq and TREK-seq to capture chromatin accessibility, transcriptome, surface proteins, T cell receptor, HIV-1 DNA and HIV-1 RNA in 100,744 gut T cells from ten aviremic HIV-1+ individuals and five HIV-1- donors. BACH2 was the leading transcription factor that shaped gut tissue resident memory T cells (TRMs) into long-lived memory with restrained interferon-induced effector function. We found that HIV-1-infected cells were enriched in TRMs (80.8%). HIV-1-infected cells had increased BACH2 transcription factor accessibility, TRM (CD49a, CD69, CD103) and survival (IL7R) gene expression, and Th17 polarization (RORC, CCR6). In vitro gut CD4+ T cell infection revealed preferential infection and persistence of HIV-1 in CCR6+ TRMs. Overall, we found BACH2-driven TRM program promotes HIV-1 persistence and BACH2 as a new therapeutic target.
Collapse
Affiliation(s)
- Yulong Wei
- Department of Microbial Pathogenesis, Yale University School of Medicine, New Haven, CT 06519, USA
| | - Haocong Katherine Ma
- Department of Microbial Pathogenesis, Yale University School of Medicine, New Haven, CT 06519, USA
| | - Michelle E. Wong
- Department of Microbial Pathogenesis, Yale University School of Medicine, New Haven, CT 06519, USA
| | | | - Liza Konnikova
- Departments of Pediatrics, Yale University School of Medicine, New Haven, CT 06519, USA
| | - Pablo Tebas
- Presbyterian Hospital-University of Pennsylvania Hospital, Philadelphia, PA 19104, USA
| | - Ricardo Morgenstern
- Division of Gastroenterology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | | | - Ya-Chi Ho
- Department of Microbial Pathogenesis, Yale University School of Medicine, New Haven, CT 06519, USA
| |
Collapse
|
40
|
Mahapatra B, Singh A, Banerjee A, Sirohi S, Singh S, Dubey VK, Singh RK. A squalene oil emulsified MPL-A and anti-CD200/CD300a antibodies adjuvanted whole-killed Leishmania vaccine provides durable immunity against L. donovani parasites. Vaccine 2024; 42:126373. [PMID: 39288578 DOI: 10.1016/j.vaccine.2024.126373] [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: 06/19/2024] [Revised: 09/11/2024] [Accepted: 09/11/2024] [Indexed: 09/19/2024]
Abstract
Antigenic inefficacy to induce robust immune responses and durable memory are major causes of constantly failing prophylactic approaches in leishmaniasis. Here, we determine the potential of a standardized whole-killed Leishmania vaccine (Leishvacc) adjuvanted with anti-CD200 and anti-CD300a antibodies, either alone or with monophosphoryl lipid A (MPL-SE) emulsified in squalene oil, in restoring the compromised antigen presenting abilities of dendritic cells (DCs), effector properties of CD4+T cells and providing protection against Leishmania donovani parasites. In animals vaccinated with antibodies adjuvanted vaccines, either alone or with MPL-SE, the antigen presenting abilities of CD11c+ DCs against Leishmania antigens, measured in terms of CD80, CD86, MHC-I, and MHC-II surface receptors and intracellular IL-12 were found enhanced than non-adjuvanted vaccine. We observed more proliferative and pro-inflammatory cytokines i.e. IL-2, IFN-γ, IL-23, and IL-12 producing CD4+T cells in antibodies/MPL-SE adjuvanted vaccinated animals further suggesting that this approach helps antigen activated CD4+T cells to acquire pro-inflammatory cytokines producing abilities. In antibodies, either alone or with MPL-SE, vaccinated animals, the number of CD4+ central memory T cells and their longevity were found significantly enhanced that further evidenced the impact of this vaccination approach in inducing long term protective immunity. The animals, receiving antibodies adjuvanted vaccines, either alone or with MPL-SE, exhibited excellent protection against virulent parasites by restricting their growth, which correlated with the significantly reduced parasitemia, splenomegaly, and hepatomegaly, along with fewer numbers of liver granulomas. Our findings provide an insight to a new immunoprophylactic approach against visceral leishmaniasis, which not only satisfies the safety criteria, but also provides a robust immunogenic response with remarkable potential for parasites control. However, further in-depth investigations are needed to ascertain its ability in inducing long-lasting immunity.
Collapse
Affiliation(s)
- Baishakhi Mahapatra
- Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi 221005, India
| | - Abhishek Singh
- Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi 221005, India
| | - Arpita Banerjee
- Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi 221005, India
| | - Shruti Sirohi
- Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi 221005, India
| | - Samer Singh
- Centre of Experimental Medicine and Surgery, Institute of Medical Science, Banaras Hindu University, Varanasi 221005, India
| | - Vikash K Dubey
- Department of Biochemical Engineering, Indian Institute of Technology, Banaras Hindu University, Varanasi 221005, India
| | - Rakesh K Singh
- Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi 221005, India.
| |
Collapse
|
41
|
Gupta A, Rudra A, Reed K, Langer R, Anderson DG. Advanced technologies for the development of infectious disease vaccines. Nat Rev Drug Discov 2024; 23:914-938. [PMID: 39433939 DOI: 10.1038/s41573-024-01041-z] [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] [Accepted: 08/28/2024] [Indexed: 10/23/2024]
Abstract
Vaccines play a critical role in the prevention of life-threatening infectious disease. However, the development of effective vaccines against many immune-evading pathogens such as HIV has proven challenging, and existing vaccines against some diseases such as tuberculosis and malaria have limited efficacy. The historically slow rate of vaccine development and limited pan-variant immune responses also limit existing vaccine utility against rapidly emerging and mutating pathogens such as influenza and SARS-CoV-2. Additionally, reactogenic effects can contribute to vaccine hesitancy, further undermining the ability of vaccination campaigns to generate herd immunity. These limitations are fuelling the development of novel vaccine technologies to more effectively combat infectious diseases. Towards this end, advances in vaccine delivery systems, adjuvants, antigens and other technologies are paving the way for the next generation of vaccines. This Review focuses on recent advances in synthetic vaccine systems and their associated challenges, highlighting innovation in the field of nano- and nucleic acid-based vaccines.
Collapse
Affiliation(s)
- Akash Gupta
- David H Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Arnab Rudra
- David H Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children's Hospital, Boston, MA, USA
| | - Kaelan Reed
- David H Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Robert Langer
- David H Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Harvard and MIT Division of Health Science and Technology, Massachusetts Institute of Technology, Cambridge, MA, USA
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Daniel G Anderson
- David H Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children's Hospital, Boston, MA, USA.
- Harvard and MIT Division of Health Science and Technology, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA.
| |
Collapse
|
42
|
Swan DA, Krantz EM, Byrne C, Okuku F, Nankoma J, Mutyaba I, Phipps W, Schiffer JT. Human Herpes Virus-8 Oral Shedding Heterogeneity is Due to Varying Rates of Reactivation from Latency and Immune Containment. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.26.625350. [PMID: 39651144 PMCID: PMC11623612 DOI: 10.1101/2024.11.26.625350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2024]
Abstract
Human herpesvirus-8 (HHV-8) is a gamma herpesvirus linked to the development of Kaposi sarcoma (KS). KS is more common in persons living with HIV (PLWH), but endemic KS in HIV-negative individuals is also common in sub-Saharan Africa. HHV-8 shedding occurs in the oral mucosa and is likely responsible for transmission. The mechanistic drivers of different HHV-8 shedding patterns in infected individuals are unknown. We applied stochastic mathematical models to a longitudinal study of HHV-8 oral shedding in 295 individuals in Uganda who were monitored daily with oral swabs. Participants were divided into four groups based on whether they were HIV-negative or positive as well as KS-negative or positive. In all groups, we observed a wide variance of shedding patterns, including no shedding, episodic low viral load shedding, and persistent high viral load shedding. Our model closely replicates patterns in individual data and attributes higher shedding rates to increased rates of viral reactivation, and lower median viral load values to more rapid and effective engagement of cytolytic immune responses. Our model provides a framework for understanding different shedding patterns observed in individuals with HHV-8 infection. Keypoints HHV8 shedding rate is mosty determined by rate of reactivation from latency while viral loads is mostly dteremined by peripheral immune responses.DAS performed all mathematical modeling and editied the paper; EMK performed statistical analysis and edited the paper; CB assisted with modeling; FO, JN and IM designed and implemented the clinical protocols; WP designed and implemented the clinical protocols and edited the paper; JTS conceived the study and write the paper.
Collapse
|
43
|
Tian Q, Chen C, Lu J, Zheng X, Zhai X, Yang Y, Zhao Z, Hao J, Yang K, Ye L, Wang Y. Ferroptosis exacerbates the clonal deletion of virus-specific exhausted CD8 + T cells. Front Immunol 2024; 15:1490845. [PMID: 39654902 PMCID: PMC11625764 DOI: 10.3389/fimmu.2024.1490845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Accepted: 10/30/2024] [Indexed: 12/12/2024] Open
Abstract
During chronic infection or tumorigenesis, persistent antigen stimulation contributes to the exhaustion of CD8+ T cells. Nevertheless, exhausted CD8+ T (TEX) cells still preserve certain effector function, and maintaining a reservoir of exhausted cells is of vital importance for virus elimination and tumor eradiation. Despite considerable work interrogating the rejuvenation of TEX cells, mechanisms underpinning the clonal deletion of TEX cells remain largely unexplored over the past decade. In this study, we employed mouse models of LCMV infection to demonstrate that excessive accumulation of lipid peroxidation rendered virus-specific TEX cells to ferroptosis, which may correlate with enhanced mitochondria-derived oxidative stress and compromised activity of glutathione peroxidase 4 (GPX4). In addition, either incomplete or complete ablation of GPX4 resulted in exacerbated ferroptosis and aggravated shrunken population of virus-specific TEX cells. On the other hand, inhibiting ferroptosis via administration of a ferroptosis inhibitor or overexpression of GPX4 greatly rectified the cell loss of virus-specific TEX cells. Collectively, we disclosed ferroptosis as a crucial player in the clonal deletion of virus-specific TEX cells and stressed the intervention of ferroptosis as a promising approach to optimize the longevity of virus-specific TEX cells.
Collapse
Affiliation(s)
- Qin Tian
- Dermatology Hospital, Southern Medical University, Guangzhou, China
| | - Cheng Chen
- Guangdong Province Key Laboratory of Immune Regulation and Immunotherapy, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, Guangdong, China
| | - Jinjin Lu
- Guangdong Province Key Laboratory of Immune Regulation and Immunotherapy, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, Guangdong, China
| | - Xinyu Zheng
- Institute of Immunology, Third Military Medical University, Chongqing, China
| | - Xiuming Zhai
- Institute of Immunological Innovation and Translation, Chongqing Medical University, Chongqing, China
| | - Yanping Yang
- School of Life Science, Chongqing University, Chongqing, China
| | - Ziyao Zhao
- Institute of Immunological Innovation and Translation, Chongqing Medical University, Chongqing, China
| | - Jiangtao Hao
- Institute of Immunological Innovation and Translation, Chongqing Medical University, Chongqing, China
| | - Ke Yang
- Institute of Immunological Innovation and Translation, Chongqing Medical University, Chongqing, China
| | - Lilin Ye
- Guangdong Province Key Laboratory of Immune Regulation and Immunotherapy, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, Guangdong, China
- Institute of Immunology, Third Military Medical University, Chongqing, China
| | - Yifei Wang
- Institute of Immunological Innovation and Translation, Chongqing Medical University, Chongqing, China
| |
Collapse
|
44
|
Huang S, He Y, Madow A, Peng H, Griffin M, Qi J, Huang M, Amoroso H, Abrashoff R, Heldman N, Belcher AM. Reprogramming the genome of M13 bacteriophage for all-in-one personalized cancer vaccine. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.22.624916. [PMID: 39605543 PMCID: PMC11601592 DOI: 10.1101/2024.11.22.624916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Peptide-based vaccines face limitations in immunogenicity and stability, and challenges in co-delivering antigens and adjuvants effectively. Virus-based nanoparticles, particularly M13 bacteriophage, present a promising solution due to their genetic modifiability, intrinsic adjuvanticity, and efficient antigen presentation capabilities. Here we developed a programmable M13 phage-based personalized cancer vaccine enabling single-step antigen-adjuvant assembly. Specifically, we designed a reprogrammed (RP) phage platform that precisely regulates Toll-like receptor 9 activation by programming its genome sequence and modulates antigen density through genetic engineering. Vaccination studies with RP phages demonstrated that the immune response could be modulated by fine-tuning the adjuvanticity and antigen density, revealing an optimal antigen dose and adjuvanticity for maximum vaccine efficacy. The RP phage induced a remarkable 24-fold increase in neoantigen-specific CD8 + T cells and eradicated established MC-38 tumors when combined with anti-PD-1 therapy. These findings highlight the RP phage's potential as a powerful nanovaccine platform for personalized cancer vaccines.
Collapse
|
45
|
LaFleur MW, D’Andrea JM, Patterson DG, Streeter IS, Coxe MA, Osborn JF, Milling LE, Tjokrosurjo Q, Gillis JE, Nguyen TH, Schwartz MA, Hacohen N, Doench JG, Sharpe AH. In Vivo CRISPR Screening Reveals CHD7 as a Positive Regulator of Short-lived Effector Cells. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 213:1528-1541. [PMID: 39373572 PMCID: PMC11578095 DOI: 10.4049/jimmunol.2400213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 09/06/2024] [Indexed: 10/08/2024]
Abstract
CD8+ T cells differentiate into two subpopulations in response to acute viral infection: memory precursor effector cells (MPECs) and short-lived effector cells (SLECs). MPECs and SLECs are epigenetically distinct; however, the epigenetic regulators required for formation of these subpopulations are mostly unknown. In this study, we performed an in vivo CRISPR screen in murine naive CD8+ T cells to identify the epigenetic regulators required for MPEC and SLEC formation, using the acute lymphocytic choriomeningitis virus Armstrong infection model. We identified the ATP-dependent chromatin remodeler CHD7 (chromodomain-helicase DNA-binding protein 7) as a positive regulator of SLEC formation, as knockout (KO) of Chd7 reduced SLECs numerically. In contrast, KO of Chd7 increased the formation of central memory T cells following pathogen clearance yet attenuated memory cell expansion following a rechallenge. These findings establish CHD7 as a novel positive regulator of SLEC and a negative regulator of central memory T cell formation.
Collapse
Affiliation(s)
- Martin W. LaFleur
- Department of Immunology, Blavatnik Institute, Harvard
Medical School, Boston, MA 02115
- Gene Lay Institute of Immunology and Inflammation, Brigham
and Women’s Hospital, Massachusetts General Hospital, and Harvard Medical
School, Boston, MA 02115
| | - Jasmin M. D’Andrea
- Department of Immunology, Blavatnik Institute, Harvard
Medical School, Boston, MA 02115
- Gene Lay Institute of Immunology and Inflammation, Brigham
and Women’s Hospital, Massachusetts General Hospital, and Harvard Medical
School, Boston, MA 02115
| | - Dillon G. Patterson
- Department of Immunology, Blavatnik Institute, Harvard
Medical School, Boston, MA 02115
- Gene Lay Institute of Immunology and Inflammation, Brigham
and Women’s Hospital, Massachusetts General Hospital, and Harvard Medical
School, Boston, MA 02115
| | - Ivy S.L. Streeter
- Department of Immunology, Blavatnik Institute, Harvard
Medical School, Boston, MA 02115
- Gene Lay Institute of Immunology and Inflammation, Brigham
and Women’s Hospital, Massachusetts General Hospital, and Harvard Medical
School, Boston, MA 02115
| | - Matthew A. Coxe
- Department of Immunology, Blavatnik Institute, Harvard
Medical School, Boston, MA 02115
- Gene Lay Institute of Immunology and Inflammation, Brigham
and Women’s Hospital, Massachusetts General Hospital, and Harvard Medical
School, Boston, MA 02115
| | - Jossef F. Osborn
- Department of Immunology, Blavatnik Institute, Harvard
Medical School, Boston, MA 02115
- Gene Lay Institute of Immunology and Inflammation, Brigham
and Women’s Hospital, Massachusetts General Hospital, and Harvard Medical
School, Boston, MA 02115
| | - Lauren E. Milling
- Department of Immunology, Blavatnik Institute, Harvard
Medical School, Boston, MA 02115
- Gene Lay Institute of Immunology and Inflammation, Brigham
and Women’s Hospital, Massachusetts General Hospital, and Harvard Medical
School, Boston, MA 02115
| | - Qin Tjokrosurjo
- Department of Immunology, Blavatnik Institute, Harvard
Medical School, Boston, MA 02115
- Gene Lay Institute of Immunology and Inflammation, Brigham
and Women’s Hospital, Massachusetts General Hospital, and Harvard Medical
School, Boston, MA 02115
| | - Jacob E. Gillis
- Department of Immunology, Blavatnik Institute, Harvard
Medical School, Boston, MA 02115
- Gene Lay Institute of Immunology and Inflammation, Brigham
and Women’s Hospital, Massachusetts General Hospital, and Harvard Medical
School, Boston, MA 02115
| | - Thao H. Nguyen
- Department of Immunology, Blavatnik Institute, Harvard
Medical School, Boston, MA 02115
- Gene Lay Institute of Immunology and Inflammation, Brigham
and Women’s Hospital, Massachusetts General Hospital, and Harvard Medical
School, Boston, MA 02115
| | - Marc A. Schwartz
- Massachusetts General Hospital Cancer Center,
Department of Medicine, Massachusetts General Hospital, Harvard Medical School,
Boston, MA 02114
- Broad Institute of MIT and Harvard, Cambridge, MA
02142
- Division of Hematology/Oncology, Boston Children’s
Hospital, Boston, MA 02115
| | - Nir Hacohen
- Massachusetts General Hospital Cancer Center,
Department of Medicine, Massachusetts General Hospital, Harvard Medical School,
Boston, MA 02114
- Broad Institute of MIT and Harvard, Cambridge, MA
02142
| | | | - Arlene H. Sharpe
- Department of Immunology, Blavatnik Institute, Harvard
Medical School, Boston, MA 02115
- Gene Lay Institute of Immunology and Inflammation, Brigham
and Women’s Hospital, Massachusetts General Hospital, and Harvard Medical
School, Boston, MA 02115
- Broad Institute of MIT and Harvard, Cambridge, MA
02142
| |
Collapse
|
46
|
Geng S, Fang B, Wang K, Wang F, Zhou Y, Hou Y, Iqbal MZ, Chen Y, Yu Z. Polydopamine Nanoformulations Induced ICD and M1 Phenotype Macrophage Polarization for Enhanced TNBC Synergistic Photothermal Immunotherapy. ACS APPLIED MATERIALS & INTERFACES 2024; 16:59814-59832. [PMID: 39450881 DOI: 10.1021/acsami.4c11594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/26/2024]
Abstract
Photothermal therapy (PTT) is a promising technology that can achieve the thermal ablation of tumors and induce immunogenic cell death (ICD). However, relying solely on the antitumor immune responses caused by PTT-induced ICD is insufficient to suppress tumor metastasis and recurrence effectively. Fortunately, multifunctional nanoformulation-based synergistic photothermal immunotherapy can eliminate primary and metastatic tumors and inhibit tumor recurrence for a long time. Herein, we select polydopamine (PDA) nanoparticles to serve as the carrier for our nanomedicine as well as a potent photothermal agent and modulator of macrophage polarization. PDA nanoparticles are loaded with the insoluble immune adjuvant Imiquimod (R837) to construct PDA(R837) nanoformulations. These straightforward yet highly effective nanoformulations demonstrate excellent performance, allowing for successful triple-negative breast cancer (TNBC) treatment through synergistic photothermal immunotherapy. Moreover, experimental results showed that PDA(R837) implementation of PTT is effective in the thermal ablation of primary tumors while causing ICD and releasing R837, further promoting dendritic cell (DC) maturation and activating the systemic antitumor immune response. Furthermore, PDA(R837) nanoformulations inhibit tumor metastasis and recurrence and achieve M1 phenotype macrophage polarization, achieving long-term and excellent antitumor efficacy. Therefore, the structurally simple PDA(R837) nanoformulations provide cancer treatment and have excellent clinical translational application prospects.
Collapse
Affiliation(s)
- Siqi Geng
- School of Life and Environmental Sciences, Shaoxing University, Shaoxing, Zhejiang 312000, P. R. China
- Laboratory of Nanomedicine, Medical Science Research Center, School of Medicine, Shaoxing University, Shaoxing, Zhejiang 312000, P. R. China
| | - Baoru Fang
- School of Life and Environmental Sciences, Shaoxing University, Shaoxing, Zhejiang 312000, P. R. China
- Laboratory of Nanomedicine, Medical Science Research Center, School of Medicine, Shaoxing University, Shaoxing, Zhejiang 312000, P. R. China
| | - Ke Wang
- School of Life and Environmental Sciences, Shaoxing University, Shaoxing, Zhejiang 312000, P. R. China
- Laboratory of Nanomedicine, Medical Science Research Center, School of Medicine, Shaoxing University, Shaoxing, Zhejiang 312000, P. R. China
| | - Fang Wang
- Laboratory of Nanomedicine, Medical Science Research Center, School of Medicine, Shaoxing University, Shaoxing, Zhejiang 312000, P. R. China
- The First Clinical Medical College, Wenzhou Medical University, Wenzhou, Zhejiang 325000, P. R. China
| | - Yiqing Zhou
- Laboratory of Nanomedicine, Medical Science Research Center, School of Medicine, Shaoxing University, Shaoxing, Zhejiang 312000, P. R. China
| | - Yike Hou
- Institute of Smart Biomedical Materials, School of Materials Science and Engineering, Zhejiang Sci-Tech University, Hangzhou, Zhejiang 310018, P. R. China
| | - M Zubair Iqbal
- Institute of Smart Biomedical Materials, School of Materials Science and Engineering, Zhejiang Sci-Tech University, Hangzhou, Zhejiang 310018, P. R. China
| | - Yanping Chen
- School of Life and Environmental Sciences, Shaoxing University, Shaoxing, Zhejiang 312000, P. R. China
- Laboratory of Nanomedicine, Medical Science Research Center, School of Medicine, Shaoxing University, Shaoxing, Zhejiang 312000, P. R. China
| | - Zhangsen Yu
- School of Life and Environmental Sciences, Shaoxing University, Shaoxing, Zhejiang 312000, P. R. China
- Laboratory of Nanomedicine, Medical Science Research Center, School of Medicine, Shaoxing University, Shaoxing, Zhejiang 312000, P. R. China
| |
Collapse
|
47
|
Shoumariyeh K, Csernalabics B, Salimi Alizei E, Reinscheid M, Giese S, Ciminski K, Kochs G, Schwemmle M, Lang-Meli J, Maas M, Roehlen N, Karl V, Graeser A, Sogukpinar O, von Metzler I, Grathwohl D, Rasche L, Hebart H, Kull M, Emmerich F, Waller CF, Duyster J, Engelhardt M, Hartmann TN, Bengsch B, Boettler T, Neumann-Haefelin C, Hofmann M, Thimme R, Luxenburger H. Impaired SARS-CoV-2-Specific CD8+ T Cells After Infection or Vaccination but Robust Hybrid T Cell Immunity in Patients with Multiple Myeloma. Vaccines (Basel) 2024; 12:1249. [PMID: 39591152 PMCID: PMC11598869 DOI: 10.3390/vaccines12111249] [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: 09/24/2024] [Revised: 10/18/2024] [Accepted: 10/31/2024] [Indexed: 11/28/2024] Open
Abstract
BACKGROUND Multiple myeloma (MM) patients are at high risk of severe infections including COVID-19 due to an immune dysregulation affecting both innate and adaptive immune responses. However, our understanding of the immune responses to infection and vaccination in MM patients is limited. To gain more detailed insights into infection- and vaccine-elicited T cell immunity in MM, we studied the CD8+ T cell response on the single-epitope level in SARS-CoV-2 convalescent and mRNA-vaccinated MM patients. METHODS We compared peptide/MHC class I tetramer-enriched SARS-CoV-2-specific CD8+ T cells and antibody responses in MM patients (convalescent: n = 16, fully vaccinated: n = 5, vaccinated convalescent: n = 5) and healthy controls (HCs) (convalescent: n = 58, fully vaccinated: n = 7) either after infection with SARS-CoV-2 alone, complete mRNA vaccination or SARS-CoV-2 infection and single-shot mRNA vaccination (hybrid immunity). RESULTS MM patients have lower frequencies and a lower proportion of fully functional virus-specific CD8+ T cells compared to HCs, after both SARS-CoV-2 infection and vaccination. CD8+ T cell memory subset distribution in MM patients is skewed towards reduced frequencies of central memory (TCM) T cells and higher frequencies of effector memory 1 (TEM1) T cells. In contrast, the humoral immune response was comparable in both cohorts after viral clearance. Notably, CD8+ T cell frequencies as well as the humoral immune response were improved by a single dose of mRNA vaccine in convalescent MM patients. CONCLUSIONS MM patients have relative immunological deficiencies in SARS-CoV-2 immunity but benefit from hybrid immunity. These findings underline the relevance of vaccinations in this vulnerable patient group.
Collapse
Affiliation(s)
- Khalid Shoumariyeh
- Department of Medicine I, Medical Center—University of Freiburg, Faculty of Medicine, University of Freiburg, 79098 Freiburg, Germany
- German Cancer Consortium (DKTK), Partner Site Freiburg, a Partnership Between DKFZ and University Medical Center Freiburg, 79098 Freiburg, Germany
| | - Benedikt Csernalabics
- Department of Medicine II (Gastroenterology, Hepatology, Endocrinology and Infectious Diseases), Freiburg University Medical Center, Faculty of Medicine, University of Freiburg, 79098 Freiburg, Germany
| | - Elahe Salimi Alizei
- Department of Medicine II (Gastroenterology, Hepatology, Endocrinology and Infectious Diseases), Freiburg University Medical Center, Faculty of Medicine, University of Freiburg, 79098 Freiburg, Germany
- Faculty of Chemistry and Pharmacy, University of Freiburg, 79098 Freiburg, Germany
| | - Matthias Reinscheid
- Department of Medicine II (Gastroenterology, Hepatology, Endocrinology and Infectious Diseases), Freiburg University Medical Center, Faculty of Medicine, University of Freiburg, 79098 Freiburg, Germany
- Faculty of Biology, University of Freiburg, 79098 Freiburg, Germany
| | - Sebastian Giese
- Institute of Virology, Freiburg University Medical Center, Faculty of Medicine, University of Freiburg, 79098 Freiburg, Germany
| | - Kevin Ciminski
- Institute of Virology, Freiburg University Medical Center, Faculty of Medicine, University of Freiburg, 79098 Freiburg, Germany
| | - Georg Kochs
- Institute of Virology, Freiburg University Medical Center, Faculty of Medicine, University of Freiburg, 79098 Freiburg, Germany
| | - Martin Schwemmle
- Institute of Virology, Freiburg University Medical Center, Faculty of Medicine, University of Freiburg, 79098 Freiburg, Germany
| | - Julia Lang-Meli
- Department of Medicine II (Gastroenterology, Hepatology, Endocrinology and Infectious Diseases), Freiburg University Medical Center, Faculty of Medicine, University of Freiburg, 79098 Freiburg, Germany
| | - Michelle Maas
- Department of Medicine II (Gastroenterology, Hepatology, Endocrinology and Infectious Diseases), Freiburg University Medical Center, Faculty of Medicine, University of Freiburg, 79098 Freiburg, Germany
- Faculty of Biology, University of Freiburg, 79098 Freiburg, Germany
| | - Natascha Roehlen
- Department of Medicine II (Gastroenterology, Hepatology, Endocrinology and Infectious Diseases), Freiburg University Medical Center, Faculty of Medicine, University of Freiburg, 79098 Freiburg, Germany
| | - Vivien Karl
- Department of Medicine II (Gastroenterology, Hepatology, Endocrinology and Infectious Diseases), Freiburg University Medical Center, Faculty of Medicine, University of Freiburg, 79098 Freiburg, Germany
- Faculty of Biology, University of Freiburg, 79098 Freiburg, Germany
| | - Anne Graeser
- Department of Medicine II (Gastroenterology, Hepatology, Endocrinology and Infectious Diseases), Freiburg University Medical Center, Faculty of Medicine, University of Freiburg, 79098 Freiburg, Germany
| | - Oezlem Sogukpinar
- Department of Medicine II (Gastroenterology, Hepatology, Endocrinology and Infectious Diseases), Freiburg University Medical Center, Faculty of Medicine, University of Freiburg, 79098 Freiburg, Germany
| | - Ivana von Metzler
- Department of Medicine II—Hematology and Oncology, Goethe-University Frankfurt, University Hospital, 60629 Frankfurt am Main, Germany
- Frankfurt Cancer Institute (FCI), 60596 Frankfurt am Main, Germany
- German Cancer Consortium (DKTK), Partner Site Frankfurt/Mainz, a Partnership Between DKFZ and University Hospital Frankfurt, 60596 Frankfurt am Main, Germany
| | - Denise Grathwohl
- Department of Internal Medicine II, University of Würzburg, 97070 Würzburg, Germany
| | - Leo Rasche
- Department of Internal Medicine II, University of Würzburg, 97070 Würzburg, Germany
| | - Holger Hebart
- Clinics Ostalb, Stauferklinikum, 73557 Mutlangen, Germany
| | - Miriam Kull
- Department of Internal Medicine III, Ulm University Hospital, 89081 Ulm, Germany
| | - Florian Emmerich
- Institute for Transfusion Medicine and Gene Therapy, Freiburg University Medical Center, Faculty of Medicine, University of Freiburg, 79098 Freiburg, Germany
| | - Cornelius Florian Waller
- Department of Medicine I, Medical Center—University of Freiburg, Faculty of Medicine, University of Freiburg, 79098 Freiburg, Germany
| | - Justus Duyster
- Department of Medicine I, Medical Center—University of Freiburg, Faculty of Medicine, University of Freiburg, 79098 Freiburg, Germany
- German Cancer Consortium (DKTK), Partner Site Freiburg, a Partnership Between DKFZ and University Medical Center Freiburg, 79098 Freiburg, Germany
| | - Monika Engelhardt
- Department of Medicine I, Medical Center—University of Freiburg, Faculty of Medicine, University of Freiburg, 79098 Freiburg, Germany
| | - Tanja Nicole Hartmann
- Department of Medicine I, Medical Center—University of Freiburg, Faculty of Medicine, University of Freiburg, 79098 Freiburg, Germany
| | - Bertram Bengsch
- German Cancer Consortium (DKTK), Partner Site Freiburg, a Partnership Between DKFZ and University Medical Center Freiburg, 79098 Freiburg, Germany
- Department of Medicine II (Gastroenterology, Hepatology, Endocrinology and Infectious Diseases), Freiburg University Medical Center, Faculty of Medicine, University of Freiburg, 79098 Freiburg, Germany
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, 79098 Freiburg, Germany
| | - Tobias Boettler
- Department of Medicine II (Gastroenterology, Hepatology, Endocrinology and Infectious Diseases), Freiburg University Medical Center, Faculty of Medicine, University of Freiburg, 79098 Freiburg, Germany
| | - Christoph Neumann-Haefelin
- Department of Medicine II (Gastroenterology, Hepatology, Endocrinology and Infectious Diseases), Freiburg University Medical Center, Faculty of Medicine, University of Freiburg, 79098 Freiburg, Germany
| | - Maike Hofmann
- Department of Medicine II (Gastroenterology, Hepatology, Endocrinology and Infectious Diseases), Freiburg University Medical Center, Faculty of Medicine, University of Freiburg, 79098 Freiburg, Germany
| | - Robert Thimme
- Department of Medicine II (Gastroenterology, Hepatology, Endocrinology and Infectious Diseases), Freiburg University Medical Center, Faculty of Medicine, University of Freiburg, 79098 Freiburg, Germany
| | - Hendrik Luxenburger
- Department of Medicine II (Gastroenterology, Hepatology, Endocrinology and Infectious Diseases), Freiburg University Medical Center, Faculty of Medicine, University of Freiburg, 79098 Freiburg, Germany
| |
Collapse
|
48
|
Park LM, Lannigan J, Low Q, Jaimes MC, Bonilla DL. OMIP-109: 45-color full spectrum flow cytometry panel for deep immunophenotyping of the major lineages present in human peripheral blood mononuclear cells with emphasis on the T cell memory compartment. Cytometry A 2024; 105:807-815. [PMID: 39466962 DOI: 10.1002/cyto.a.24900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 09/11/2024] [Accepted: 09/14/2024] [Indexed: 10/30/2024]
Abstract
The need for more in-depth exploration of the human immune system has moved the flow cytometry field forward with advances in instrumentation, reagent development and availability, and user-friendly implementation of data analysis methods. We developed a high-quality human 45-color panel, for comprehensive characterization of major cell lineages present in circulation including T cells, γδ T cells, NKT-like cells, B cells, NK cells, monocytes, basophils, dendritic cells, and ILCs. Assay optimization steps are described in detail to ensure that each marker in the panel was optimally resolved. In addition, we highlight the outstanding discernment of cell activation, exhaustion, memory, and differentiation states of CD4+ and CD8+ T cells using this 45-color panel. The panel enabled an in-depth description of very distinct phenotypes associated with the complexity of the T cell memory response. Furthermore, we present how this panel can be effectively used for cell sorting on instruments with a similar optical layout to achieve the same level of resolution. Functional evaluation of sorted specific rare cell subsets demonstrated significantly different patterns of immunological responses to stimulation, supporting functional and phenotypic differences within the T cell memory subsets. In summary, the combination of full spectrum profiling technology and careful assay design and optimization results in a high resolution multiparametric 45-color assay. This panel offers the opportunity to fully characterize immunological profiles present in peripheral blood in the context of infectious diseases, autoimmunity, neurodegeneration, immunotherapy, and biomarker discovery.
Collapse
Affiliation(s)
- Lily M Park
- Cytek Biosciences, Inc., Scientific Commercialization, Fremont, California, USA
| | - Joanne Lannigan
- Flow Cytometry Support Services, LLC, Alexandria, Virginia, USA
| | - Quentin Low
- Cytek Biosciences, Inc., Scientific Commercialization, Fremont, California, USA
| | - Maria C Jaimes
- Cytek Biosciences, Inc., Scientific Commercialization, Fremont, California, USA
| | - Diana L Bonilla
- Cytek Biosciences, Inc., Scientific Commercialization, Fremont, California, USA
| |
Collapse
|
49
|
Iqbal J, Inghirami G, Chan WC. New insights into the biology of T-cell lymphomas. Blood 2024; 144:1873-1886. [PMID: 39213420 PMCID: PMC11551850 DOI: 10.1182/blood.2023021787] [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: 01/08/2024] [Revised: 07/29/2024] [Accepted: 07/29/2024] [Indexed: 09/04/2024] Open
Abstract
ABSTRACT Peripheral T-cell lymphomas (PTCLs) encompass a heterogeneous group of postthymic T-cell lymphomas with >30 distinct subtypes associated with varied clinicopathological features. Unfortunately, the overall survival of the major PTCL subtypes is dismal and has not improved for decades; thus, there is an urgent unmet clinical need to improve diagnosis, therapies, and clinical outcomes. The diagnosis is often challenging, requiring a combinatorial evaluation of clinical, morphologic, and immunophenotypic features. PTCL pathobiology is difficult to investigate due to enormous intertumor and intratumor heterogeneity, limited tissue availability, and the paucity of authentic T-cell lymphoma cell lines or genetically faithful animal models. The application of transcriptomic profiling and genomic sequencing has markedly accelerated the discovery of new biomarkers, molecular signatures, and genetic lesions, and some of the discoveries have been included in the revised World Health Organization or International Consensus Classification. Genome-wide investigations have revealed the mutational landscape and transcriptomic profiles of PTCL entities, defined the cell of origin as a major determinant of T-cell lymphoma biology, and allowed for the refinement of biologically and clinically meaningful entities for precision therapy. In this review, we prioritize the discussion on common nodal PTCL subtypes together with 2 virus-associated T-cell and natural killer cell lymphomas. We succinctly review normal T-cell development, differentiation, and T-cell receptor signaling as they relate to PTCL pathogenesis and biology. This review will facilitate a better biological understanding of the different PTCL entities and their stratification for additional studies and target-directed clinical trials.
Collapse
Affiliation(s)
- Javeed Iqbal
- Department of Pathology, Microbiology, and Immunology, University of Nebraska Medical Center, Omaha, NE
| | - Giorgio Inghirami
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York, NY
| | - Wing C. Chan
- Department of Pathology, City of Hope National Medical Center, Duarte, CA
| |
Collapse
|
50
|
Abate G, Meza KA, Colbert CG, Eickhoff CS. Assays for Assessing Mycobacterium avium Immunity and Evaluating the Effects of Therapeutics. Pathogens 2024; 13:903. [PMID: 39452774 PMCID: PMC11510112 DOI: 10.3390/pathogens13100903] [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: 08/12/2024] [Revised: 10/02/2024] [Accepted: 10/11/2024] [Indexed: 10/26/2024] Open
Abstract
In Europe and North America, the prevalence of pulmonary nontuberculous mycobacteria (NTM) is increasing. Most pulmonary NTM infections are caused by the Mycobacterium avium complex (MAC). Sadly, the treatment of pulmonary MAC is suboptimal with failure rates ranging from 37% to 58%. Therefore, there is a need to develop new therapeutics. Developing new immunotherapies and studying their interaction with standard or new drugs requires reliable assays. Four different assays including CFSE-based flow cytometry, in vitro protection assays, IFN-γ ELISPOT, and murine infection models were optimized using a reference strain of MAC (ATCC 700898) to help with the development of immunotherapies for MAC. Expansion of proliferating and IFN-γ producing human T cells is optimal after 7 days of stimulation with MAC at a multiplicity of infection (MOI) of 0.1, achieving a stimulation index of 26.5 ± 11.6 (mean ± SE). The in vitro protection assay for MAC works best by co-culturing T cells expanded for 7 days with MAC (MOI 1)-infected autologous macrophages. Aerosol MAC infection of mice allows measurement of the effects of the BCG vaccine and clarithromycin. IFN-γ ELISPOT assays with live MAC (MOI 3) stimulation of splenocytes from mice immunized with BCG help identify differences between unimmunized mice and mice immunized with BCG. In conclusion, multiple assays are available for use to identify MAC-specific effector T cells, which will help in the development of new therapeutics or vaccines against pulmonary MAC.
Collapse
Affiliation(s)
- Getahun Abate
- Division of Infectious Diseases, Allergy and Immunology, Department of Internal Medicine, School of Medicine, Saint Louis University, St. Louis, MO 63104, USA (C.S.E.)
| | | | | | | |
Collapse
|