1
|
Amrane K, Le Noac'h P, Hemon P, Abgral R, Le Meur C, Pradier O, Misery L, Legoupil D, Berthou C, Uguen A. MHC class II: a predictor of outcome in melanoma treated with immune checkpoint inhibitors. Melanoma Res 2025; 35:176-186. [PMID: 39945603 DOI: 10.1097/cmr.0000000000001022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/03/2025]
Abstract
This study aimed to evaluate the predictive value of MHC class II (MHC-II) expression by melanoma cells in a large cohort of metastatic cutaneous melanoma patients treated with immune checkpoint inhibitors (ICIs). We conducted a single-center, retrospective study involving stage IV cutaneous melanoma patients who received ICI as first-line therapy. MHC-II expression in melanoma cells was quantified using dual-color anti-SOX10 and anti-MHC-II immunohistochemistry on tumor samples from 95 patients. The primary endpoint was event-free survival (EFS), with secondary endpoints including 1-year EFS, 1-year overall survival (OS), disease control rate (DCR), and the correlation between MHC-II expression and clinico-biological characteristics. The cohort had a median age of 67 years (range, 33-90), with a male-to-female ratio of 50 : 45. Thirty-three percent of patients received the ipilimumab-nivolumab combination. The median follow-up was 16.8 months. Disease progression occurred in 58 patients (61%), with a median time to progression of 4.8 months. Forty-six patients (48.4%) experienced an event within the first year, and 52 patients (54.7%) died during follow-up. MHC-II positivity was observed in ≥10% of melanoma cells in 6.3% of patients. MHC-II expression was significantly associated with 1-year EFS ( P = 0.037) and DCR ( P = 0.032), but not with EFS or 1-year OS. Age, phototype, and brain metastases were correlated with MHC-II expression status. Our findings suggest that MHC-II expression by melanoma cells may serve as a favorable predictive biomarker for survival in metastatic cutaneous melanoma patients treated with ICIs.
Collapse
Affiliation(s)
- Karim Amrane
- Department of Oncology, Regional Hospital of Morlaix, Morlaix
- Inserm, UMR1227, Lymphocytes B et Autoimmunité, Univ Brest, Inserm, LabEx IGO
| | - Pierre Le Noac'h
- Inserm, UMR1227, Lymphocytes B et Autoimmunité, Univ Brest, Inserm, LabEx IGO
- Department of Pathology, University Hospital of Brest
| | - Patrice Hemon
- Inserm, UMR1227, Lymphocytes B et Autoimmunité, Univ Brest, Inserm, LabEx IGO
| | - Ronan Abgral
- Department of Nuclear Medicine, University Hospital of Brest
- UMR Inserm 1304 GETBO, IFR 148, University of Western Brittany
| | - Coline Le Meur
- Department of Radiotherapy, University Hospital of Brest
| | - Olivier Pradier
- Department of Radiotherapy, University Hospital of Brest
- Inserm, UMR1101, LaTIM, University of Western Brittany
| | - Laurent Misery
- Department of Dermatology, University Hospital of Brest
- Laboratoire sur les Interactions Épithéliums-Neurones (LIEN-EA4685), Université de Bretagne Occidentale
| | - Delphine Legoupil
- Department of Dermatology, University Hospital of Brest
- Laboratoire sur les Interactions Épithéliums-Neurones (LIEN-EA4685), Université de Bretagne Occidentale
| | - Christian Berthou
- Inserm, UMR1227, Lymphocytes B et Autoimmunité, Univ Brest, Inserm, LabEx IGO
- Department of Hematology, University Hospital of Brest, Brest, France
| | - Arnaud Uguen
- Inserm, UMR1227, Lymphocytes B et Autoimmunité, Univ Brest, Inserm, LabEx IGO
- UMR Inserm 1304 GETBO, IFR 148, University of Western Brittany
| |
Collapse
|
2
|
Fan X, Brunetti TM, Jackson K, Roop DR. Single-Cell Profiling Reveals Global Immune Responses During the Progression of Murine Epidermal Neoplasms. Cancers (Basel) 2025; 17:1379. [PMID: 40282557 PMCID: PMC12025564 DOI: 10.3390/cancers17081379] [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: 03/10/2025] [Revised: 04/01/2025] [Accepted: 04/11/2025] [Indexed: 04/29/2025] Open
Abstract
BACKGROUND/OBJECTIVES Immune cells determine the role of the tumor microenvironment during tumor progression, either suppressing tumor formation or promoting tumorigenesis. This study aimed to fully characterize immune cell responses during skin tumor progression. METHODS Using single-cell RNA sequencing, we analyzed the profile of immune cells in the tumor microenvironment of control mouse skins and skin tumors at the single-cell level. RESULTS We identified 15 CD45+ immune cell clusters, which broadly represent the most functionally characterized immune cell types including macrophages, Langerhans cells (LC), conventional type 1 dendritic cells (cDC1), conventional type 2 dendritic cells (cDC2), migratory/mature dendritic cells (mDC), dendritic epidermal T cells (DETC), dermal γδ T cells (γδT), T cells, regulatory T cells (Tregs), natural killer cells (NK), type 2 innate lymphoid cells (ILC2), neutrophils (Neu), mast cells (Mast), and two proliferating populations (Prolif.1 and Prolif.2). Skin tumor progression reprogramed immune cells and led to a marked increase in the relative percentages of macrophages, cDC2, mDC, Tregs, and Neu. Macrophages, the largest cell cluster of immune cells in skin tumors. In addition, macrophages emerged as the predominant communication 'hub' in skin tumors, highlighting the importance of macrophages during skin tumor progression. In contrast, other immune cell clusters decreased during skin tumor progression, including DETC, γδT, ILC2, and LC. In addition, skin tumor progression dramatically upregulated Jak2/Stat3 expression and the interferon response across various immune cell clusters. Further, skin tumor progression activated T cells and NK cells indicated by elevated expression of IFN-γ and Granzyme B in skin tumors. Meanwhile, a pronounced infiltration of M2-macrophages and Tregs in skin tumors created an immunosuppressive microenvironment, consistent with the elevated expression of the Stat3 pathway in skin tumors. CONCLUSIONS Our study elucidates the immune cell landscape of epidermal neoplasms, offering a comprehensive understanding of the immune response during skin tumor progression and providing new insights into cancer immune evasion mechanisms.
Collapse
Affiliation(s)
- Xiying Fan
- Department of Dermatology, University of Colorado Anschutz Medical Campus, 12700 E. 19th Ave., Room 4007, Aurora, CO 80045, USA;
- Gates Institute, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Tonya M. Brunetti
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA;
| | - Kelsey Jackson
- Department of Dermatology, University of Colorado Anschutz Medical Campus, 12700 E. 19th Ave., Room 4007, Aurora, CO 80045, USA;
- Gates Institute, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Dennis R. Roop
- Department of Dermatology, University of Colorado Anschutz Medical Campus, 12700 E. 19th Ave., Room 4007, Aurora, CO 80045, USA;
- Gates Institute, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| |
Collapse
|
3
|
Cao R, Jiao P, Zhang S, Li J, Liu Q. Predicting the Efficacy of Immune Checkpoint Inhibitors in Esophageal Cancer: Changes in Peripheral Blood Lymphocyte Subsets Before and After Immunotherapy. Cancer Manag Res 2025; 17:815-825. [PMID: 40256769 PMCID: PMC12009565 DOI: 10.2147/cmar.s503171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Accepted: 04/04/2025] [Indexed: 04/22/2025] Open
Abstract
Background Immunotherapy has demonstrated potential in the treatment of esophageal cancer (EC); however, the overall response rate (ORR) remains below 30% among EC patients. Herein, the use of peripheral blood lymphocyte subsets as biomarkers was explored to evaluate the efficacy of immunotherapy in this patient population. Methods Sixty-three patients were enrolled. The patients were diagnosed with EC and treated with immune checkpoint inhibitors (ICIs) at The Fourth Hospital of Hebei Medical University from December 2019 to June 2023. Kaplan-Meier (KM) survival curves were used to reflect differences in survival benefit. The prognostic factors of survival were investigated using the Cox proportional hazards regression model for both univariate and multivariate analyses. Two-tailed P values were reported and statistical significance was defined as P < 0.05. Results The results of univariate and multifactorial Cox regression analysis for progression-free survival (PFS) revealed that only CD8+ T lymphocytes demonstrated a significant association with PFS (P = 0.034, P = 0.020). Additionally, the multifactorial Cox regression analysis results for overall survival (OS) revealed a significant association between natural killer (NK) cells and OS (P=0.049). Further, a systematic analysis was conducted on the CD8+ T cell biomarker. The KM survival curves indicated that the group with low CD8+ T cell levels experienced a significantly greater PFS benefit compared to the high CD8+ T cell group (P = 0.030). Conclusion The present study reveals that the reduction of both CD8+ T lymphocytes and NK cells in peripheral blood lymphocyte subsets after immunotherapy can serve as superior predictors for the effectiveness of ICIs in patients diagnosed with EC.
Collapse
Affiliation(s)
- Ruijie Cao
- Department of Immunology and Rheumatology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, People’s Republic of China
| | - Pengqing Jiao
- Department of Immunology and Rheumatology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, People’s Republic of China
| | - Shasha Zhang
- Department of Immunology and Rheumatology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, People’s Republic of China
| | - Jiasong Li
- Department of Immunology and Rheumatology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, People’s Republic of China
| | - Qingyi Liu
- Department of Cardiothoracic Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, People’s Republic of China
| |
Collapse
|
4
|
Shu Q, Wang Q, Yang X, Li B. ARIH2 serves as a potential prognostic biomarker for hepatocellular carcinoma associated with immune infiltration and ferroptosis. Front Immunol 2025; 16:1548691. [PMID: 40260250 PMCID: PMC12009847 DOI: 10.3389/fimmu.2025.1548691] [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: 12/20/2024] [Accepted: 03/17/2025] [Indexed: 04/23/2025] Open
Abstract
Background Ariadne homolog 2 (ARIH2) has been demonstrated to be upregulated in various human cancer tissues. Nevertheless, the underlying biological function of ARIH2 in the progression of hepatocellular carcinoma (HCC) remains ambiguous. Hence, we conducted a comprehensive bioinformatics analysis on the liver hepatocellular carcinoma (LIHC) dataset to explore the role of ARIH2 in tumorigenesis. Methods The mRNA and protein expression of ARIH2 was analyzed by using data from public databases and verified through immunohistochemical staining and Western blot. Logistic regression, Cox regression, receiver operating characteristic curve (ROC), Kaplan-Meier analysis and nomogram model were employed to assess the association between ARIH2 and the clinicopathological characteristics of HCC. We utilized functional enrichment analysis to investigate the potential pathways of ARIH2 in the progression of HCC. The association of ARIH2 with immune infiltration, ferroptosis and immune checkpoint genes was further evaluated. Finally, the correlation between ARIH2 and the IC50 of chemotherapeutic drugs was analyzed in HCC. Results Our study discovered that ARIH2 was up-regulated in HCC tumor tissues compared with the control group. ARIH2 expression could effectively distinguish tumor tissues from normal liver tissues. The genes related to ARIH2 showed differential expression in pathways involving immune system-related pathways and ion channels. We identified a significant association between the expression level of ARIH2 in HCC tissues and immune infiltration, immune checkpoint genes and ferroptosis. The expression level of ARIH2 was significantly correlated with the clinical stage, histological pathological grade and clinical characteristics of HCC, and could independently predict overall survival. Conclusions The expression level of ARIH2 may serve as a promising biomarker for the diagnosis and prognosis of HCC, as well as a potential drug target, which holds great significance for the development of targeted therapy for HCC.
Collapse
Affiliation(s)
| | | | - Xiaoli Yang
- Department of Hepatobiliary Surgery, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Bo Li
- Department of Hepatobiliary Surgery, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| |
Collapse
|
5
|
Palli E, Lavigne M, Verginis P, Alissafi T, Anastasopoulou A, Lyrarakis G, Kirkwood JM, Gogas H, Ziogas DC. Transcriptomic signatures in peripheral CD4 +T-lymphocytes may reflect melanoma staging and immunotherapy responsiveness prior to ICI initiation. Front Immunol 2025; 16:1529707. [PMID: 40226614 PMCID: PMC11986426 DOI: 10.3389/fimmu.2025.1529707] [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: 11/17/2024] [Accepted: 03/10/2025] [Indexed: 04/15/2025] Open
Abstract
Background and purpose Promoting adaptive immunity with ICIs has drastically improved melanoma prognosis, but not for all patients. Some cases relapse in the first few months, while others keep durable benefit, even after immunotherapy discontinuation. To identify cellular/molecular signatures in peripheral blood that could differentiate advanced from metastatic melanoma and predict dynamics for primary/secondary immune escape, we examined 100 consecutive patients with stage III/IV melanoma scheduled to start ICIs. Materials and methods At melanoma diagnosis, a multiparameter flow cytometric analysis and purification scheme using standard conjugated antibodies were performed for all individuals prior to ICI initiation. In each stage(III/IV) according to their RFS/PFS, we retrospectively selected the cases with the clearest clinical outcomes and focused our analysis on the extreme responders(n=7) and non-responders(n=7) to characterize the transcriptomes of circulating CD4+T-cells by bulk RNA-seq, Differential Expression Analysis(DEA)and Gene Ontology(GO)enrichment analysis. Based on our selected patient cohort, we examined for differentially expressed genes(DEGs)and key-pathways that appear preferentially activated in stage III vs. IV melanoma, and in long vs. short immunotherapy responders. Results Although circulating immune-cells did not numerically differ in both sets of analysis(staging and ICI responsiveness), DEA and GO data showed that patients could be clustered separately, identifying 189vs.92 DEGs in stage IV/III and 101vs.47 DEGs in early progressors/long responders. These DEGs were functionally implicated in distinct pathways. For metastatic cases: inflammatory response(logp-value=-9.2:ADGRE5/2,CYBA,GRN,HMOX1,IRF5,ITGAM), adaptive immunity(logp-value=-7.7:CD1C,CD74,CYBB,NCF2,CTSA,S100A8/9,BCL3,FCER1G), T-cell activation(logp-value=-6.3:BCL3,CD1C,CD74,FCER1G,FGL2)and lipid metabolism/catabolism(logp-value=-2.5/-2.6:ARF3,GPX1,MVD,OCRL,PCCB,CTSA,PNPLA2,NAGLU,GBA2,ABHD4); while in early-progressors to ICIs: immune effector processing(logp-value=-13.7:BCL6,FGR,HLA-DQA1/DQB1,HLA-DRA,HLA-DRB1/DRB5,NKG7,SLC11A1,TYROBP,SPON2,HAVCR2),PD-1(logp-value=-10.2:HLA-DQA1/DQB1,HLA-DRA,HLA-DRB1/DRB5)and IFN signaling(logp-value=-8.5: HLA-DQA1/DQB1,HLA-DRA,HLA-DRB1/DRB5,NCAM1,IFITM3),positive regulation of T-cell activation(logp-value=-7.7:BCL6,HLA-DQA1/DQB1,HLA-DRA,HLA-DRB1/DRB5,SASH3,HAVCR2)and CD28 co-stimulation(logp-value=-10.3:HLA-DQA1/DQB1,HLA-DRA,HLA-DRB1/DRB5), supporting an immune-mediated behavior. Conclusions Specific pathways and marker genes in the peripheral CD4+T-cells may predetermine melanoma staging and immunotherapy resistance.
Collapse
Affiliation(s)
- Eleni Palli
- First Department of Internal Medicine, Laikon General Hospital, National and Kapodistrian University of Athens - School of Medicine, Athens, Greece
| | - Matthieu Lavigne
- Institute of Molecular Biology and Biotechnology of the Foundation for Research and Technology - Biology Department, University of Crete, School of Medicine, Heraklion, Greece
| | - Panagiotis Verginis
- Institute of Molecular Biology and Biotechnology of the Foundation for Research and Technology - Biology Department, University of Crete, School of Medicine, Heraklion, Greece
| | - Themis Alissafi
- Laboratory of Biology, National and Kapodistrian University of Athens - School of Medicine, Athens, Greece
| | - Amalia Anastasopoulou
- First Department of Internal Medicine, Laikon General Hospital, National and Kapodistrian University of Athens - School of Medicine, Athens, Greece
| | - Georgios Lyrarakis
- First Department of Internal Medicine, Laikon General Hospital, National and Kapodistrian University of Athens - School of Medicine, Athens, Greece
| | - John M. Kirkwood
- Division of Hematology/Oncology, University of Pittsburgh, School of Medicine, Pittsburgh, PA, United States
| | - Helen Gogas
- First Department of Internal Medicine, Laikon General Hospital, National and Kapodistrian University of Athens - School of Medicine, Athens, Greece
| | - Dimitrios C. Ziogas
- First Department of Internal Medicine, Laikon General Hospital, National and Kapodistrian University of Athens - School of Medicine, Athens, Greece
| |
Collapse
|
6
|
Dhital R, Kim Y, Kim D, Hernandez-Aguirre I, Hedberg J, Martin A, Cassady KA. Ruxolitinib and oHSV combination therapy increases CD4 T cell activity and germinal center B cell populations in murine sarcoma. MOLECULAR THERAPY. ONCOLOGY 2025; 33:200929. [PMID: 39895689 PMCID: PMC11787636 DOI: 10.1016/j.omton.2024.200929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 12/04/2024] [Accepted: 12/18/2024] [Indexed: 02/04/2025]
Abstract
Malignant peripheral nerve sheath tumors (MPNSTs) are a highly aggressive neoplasm of the peripheral nervous system and are resistant to most conventional cancer therapies. We previously showed that pretreatment with ruxolitinib (RUX) enhanced the efficacy of oncolytic herpes simplex virus (oHSV) virotherapy in this murine sarcoma model. A low abundance of tumor-infiltrating leukocytes and limitations in conventional flow cytometry restrict analyses to a narrow subset of immune cells, potentially introducing a confirmation bias. To address these limitations, we developed a 46-color spectral flow cytometry panel for the detailed analysis of immune cell dynamics following repeated oHSV dosing. Beyond the cytotoxic T lymphocyte (CTL) and regulatory T cell (Treg) changes reported in our earlier studies, RUX+oHSV treatment modulates myeloid and other lymphoid compartments, including germinal center B cell populations with enhanced activation. RUX+oHSV therapy also increased cytokine-expressing CD4(+) populations, predominantly granzyme B(+) cytotoxic-like, interferon (IFN)-γ(+) T helper type 1 (Th1)-like, and interleukin (IL)-21(+) T follicular helper (Tfh)-like phenotypes, within the tumor infiltrates, suggestive of potential tertiary lymphoid structure development in the treated tumors. Here, we illustrate the utility of a high-dimensional spectral flow cytometry panel that permits simultaneous evaluation of intratumoral CD4/CD8 T cell, Treg, γδ-T cell, natural killer T (NKT) cell, B cell, NK cell, monocyte, macrophage, granulocyte, myeloid-derived suppressor cell (MDSC), and dendritic cell functional changes from RUX+oHSV-treated MPNSTs.
Collapse
Affiliation(s)
- Ravi Dhital
- Center for Childhood Cancer Research, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH, USA
| | - Yeaseul Kim
- Center for Childhood Cancer Research, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH, USA
| | - Doyeon Kim
- Center for Childhood Cancer Research, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH, USA
| | - Ilse Hernandez-Aguirre
- Center for Childhood Cancer Research, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH, USA
- Department of Biomedical Informatics, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Jack Hedberg
- Center for Childhood Cancer Research, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH, USA
- Department of Biomedical Informatics, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Alexia Martin
- Center for Childhood Cancer Research, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH, USA
- Department of Biomedical Informatics, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Kevin A. Cassady
- Center for Childhood Cancer Research, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH, USA
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH, USA
- Department of Pediatrics, Division of Pediatric Infectious Diseases, Nationwide Children’s Hospital, Columbus, OH, USA
| |
Collapse
|
7
|
Engelbrecht E, Stamp BF, Chew L, Sarkar OS, Harter P, Waigel SJ, Rouchka EC, Chariker J, Smolenkov A, Chesney J, McMasters K, Watson CT, Yaddanapudi K. Single-cell transcriptomics of melanoma sentinel lymph nodes identifies immune cell signatures associated with metastasis. JCI Insight 2025; 10:e183080. [PMID: 40048259 PMCID: PMC11981627 DOI: 10.1172/jci.insight.183080] [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: 05/16/2024] [Accepted: 02/25/2025] [Indexed: 04/09/2025] Open
Abstract
The sentinel lymph node (SLN) is the first lymph node encountered by a metastatic cancer cell and serves as a predictor of poor prognosis, as cases with clinically occult SLN metastases are classified as stage III with elevated rates of recurrence and diminished overall survival. However, the dynamics of immune infiltrates in SLNs remain poorly characterized. Here, using an unbiased cellular indexing of transcriptomes and epitopes by sequencing technique, we profiled 97,777 cells from SLN tissues obtained from patients with stages I/II and III cutaneous melanoma. We described the transcriptional programs of a multitude of T, B, and myeloid cell subtypes in SLNs. Based on the proportions of cell types, we determined that SLN subtypes stratified along a naive → activated axis; patients with a "high activated" signature score appeared to be undergoing a robust melanoma antigen-driven adaptive immune response and, thus, could be responsive to immunotherapy. Additionally, we identified transcriptomic signatures of SLN-infiltrating dendritic cell subsets that compromise antitumor immune responses. Our analyses provide valuable insights into tumor-driven immune changes in the SLN tissue, offering a powerful tool for the informed design of immune therapies for patients with high-risk melanoma.
Collapse
Affiliation(s)
| | | | - Lewis Chew
- Immuno-Oncology Group, UofL-Health Brown Cancer Center
- Department of Microbiology/Immunology
| | - Omar Sadi Sarkar
- Immuno-Oncology Group, UofL-Health Brown Cancer Center
- Department of Microbiology/Immunology
| | - Phillip Harter
- Immuno-Oncology Group, UofL-Health Brown Cancer Center
- Department of Microbiology/Immunology
| | | | - Eric C. Rouchka
- Department of Biochemistry and Molecular Genetics
- Department of Computer Science and Engineering
| | | | | | - Jason Chesney
- Immuno-Oncology Group, UofL-Health Brown Cancer Center
- UofL-Health Brown Cancer Center
| | | | | | - Kavitha Yaddanapudi
- Immuno-Oncology Group, UofL-Health Brown Cancer Center
- Department of Microbiology/Immunology
- Division of Immunotherapy, Department of Surgery, University of Louisville, Louisville, Kentucky, USA
| |
Collapse
|
8
|
Lingel H, Fischer L, Remstedt S, Kuropka B, Philipsen L, Han I, Sander JE, Freund C, Arra A, Brunner-Weinzierl MC. SLAMF7 (CD319) on activated CD8 + T cells transduces environmental cues to initiate cytotoxic effector cell responses. Cell Death Differ 2025; 32:561-572. [PMID: 39390117 PMCID: PMC11893764 DOI: 10.1038/s41418-024-01399-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 09/10/2024] [Accepted: 10/02/2024] [Indexed: 10/12/2024] Open
Abstract
CD8+ T-cell responses are meticulously orchestrated processes regulated by intercellular receptor:ligand interactions. These interactions critically control the dynamics of CD8+ T-cell populations that is crucial to overcome threats such as viral infections or cancer. Yet, the mechanisms governing these dynamics remain incompletely elucidated. Here, we identified a hitherto unknown T-cell referred function of the self-ligating surface receptor SLAMF7 (CD319) on CD8+ T cells during initiation of cytotoxic T-cell responses. According to its cytotoxicity related expression on T effector cells, we found that CD8+ T cells could utilize SLAMF7 to transduce environmental cues into cellular interactions and information exchange. Indeed, SLAMF7 facilitated a dose-dependent formation of stable homotypic contacts that ultimately resulted in stable cell-contacts, quorum populations and commitment to expansion and differentiation. Using pull-down assays and network analyses, we identified novel SLAMF7-binding intracellular signaling molecules including the CRK, CRKL, and Nck adaptors, which are involved in T-cell contact formation and may mediate SLAMF7 functions in sensing and adhesion. Hence, providing SLAMF7 signals during antigen recognition of CD8+ T cells enhanced their overall magnitude, particularly in responses towards low-affinity antigens, resulting in a significant boost in their proliferation and cytotoxic capacity. Overall, we have identified and characterized a potent initiator of the cytotoxic T lymphocyte response program and revealed advanced mechanisms to improve CD8+ T-cell response decisions against weak viral or tumor-associated antigens, thereby strengthening our defense against such adversaries.
Collapse
Affiliation(s)
- Holger Lingel
- Department of Experimental Paediatrics, University Hospital, Otto-von-Guericke-University, Magdeburg, Germany
- Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke-University, Magdeburg, Germany
| | - Laura Fischer
- Department of Experimental Paediatrics, University Hospital, Otto-von-Guericke-University, Magdeburg, Germany
| | - Sven Remstedt
- Department of Experimental Paediatrics, University Hospital, Otto-von-Guericke-University, Magdeburg, Germany
- Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke-University, Magdeburg, Germany
| | - Benno Kuropka
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany
| | - Lars Philipsen
- Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke-University, Magdeburg, Germany
- Multi-parametric bioimaging and cytometry (MPBIC) core facility, University Hospital, Otto-von-Guericke-University, Magdeburg, Germany
- Institute of Cellular and Molecular Immunology, University Hospital, Otto-von-Guericke-University, Magdeburg, Germany
| | - Irina Han
- Department of Experimental Paediatrics, University Hospital, Otto-von-Guericke-University, Magdeburg, Germany
- Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke-University, Magdeburg, Germany
| | - Jan-Erik Sander
- Department of Experimental Paediatrics, University Hospital, Otto-von-Guericke-University, Magdeburg, Germany
- Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke-University, Magdeburg, Germany
| | - Christian Freund
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany
| | - Aditya Arra
- Department of Experimental Paediatrics, University Hospital, Otto-von-Guericke-University, Magdeburg, Germany
- Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke-University, Magdeburg, Germany
| | - Monika C Brunner-Weinzierl
- Department of Experimental Paediatrics, University Hospital, Otto-von-Guericke-University, Magdeburg, Germany.
- Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke-University, Magdeburg, Germany.
| |
Collapse
|
9
|
TIAN KE, LI ZHIPENG, ZHAI XIANGYU, ZHOU HUAXIN, YAO HUI. A novel prognostic scoring model based on cuproptosis identifies COMMD1 as a novel therapy target for liver hepatocellular carcinoma. Oncol Res 2025; 33:617-630. [PMID: 40109870 PMCID: PMC11915051 DOI: 10.32604/or.2024.049772] [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: 01/17/2024] [Accepted: 05/16/2024] [Indexed: 03/22/2025] Open
Abstract
Background Primary liver cancer poses a significant global health burden, with projections indicating a surpassing of one million cases by 2025. Cuproptosis, a copper-dependent mechanism of cell death, plays a crucial role in the pathogenesis, progression, and prognosis of various cancers, including hepatocellular carcinoma (HCC). Purpose This study aimed to develop a prognostic model for HCC based on cuproptosis-related genes, utilizing clinical data and gene expression profiles from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) databases. Materials and Methods Clinical features and gene expression data of HCC patients were collected from publicly available databases. Patients from TCGA were randomly divided into training and testing sets, and Lasso Cox regression was applied to develop a predictive model using cuproptosis-related genes. Results The analysis identified Copper Metabolism Domain Containing 1 (COMMD1) as a potential prognostic marker for HCC, with deletion of this gene impacting disease progression. Cellular functional experiments validated the role of COMMD1 in HCC. Conclusions COMMD1 emerges as a promising candidate for HCC treatment, with implications for prognosis prediction and therapeutic targeting.
Collapse
Affiliation(s)
- KE TIAN
- General Surgery Department 2, The No. 2 People’s Hospital of Lanzhou, Lanzhou, 730030, China
| | - ZHIPENG LI
- The Hepatobiliary Surgery Department, The Second Hospital of Shandong University, Jinan, 250000, China
| | - XIANGYU ZHAI
- The Hepatobiliary Surgery Department, The Second Hospital of Shandong University, Jinan, 250000, China
- Organ Transplant Department, Qilu Hospital of Shandong University, Jinan, 250000, China
| | - HUAXIN ZHOU
- The Hepatobiliary Surgery Department, The Second Hospital of Shandong University, Jinan, 250000, China
| | - HUI YAO
- General Surgery Department 2, The No. 2 People’s Hospital of Lanzhou, Lanzhou, 730030, China
| |
Collapse
|
10
|
Tamura Y, Ohki S, Nagai H, Yoshizato R, Nishi S, Jin Y, Kitajima Y, Guo Y, Ichinohe T, Okada S, Kawano Y, Yasuda T. Co-expression of B7-H3 and LAG3 represents cytotoxicity of CD4 + T cells in humans. Front Immunol 2025; 16:1560383. [PMID: 40070836 PMCID: PMC11893609 DOI: 10.3389/fimmu.2025.1560383] [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: 01/14/2025] [Accepted: 02/10/2025] [Indexed: 03/14/2025] Open
Abstract
Recent studies have highlighted the potential contribution of CD4+ T cells with cytotoxic activity (CD4 CTLs) to anti-tumor immunity. However, their precise roles remain elusive, partly due to the absence of specific markers defining CD4 CTLs with target-killing potential in humans. We previously demonstrated that Epstein-Barr virus (EBV)-driven immortalized B cell lines efficiently induce human CD4 CTLs with cytotoxic functions comparable to cytotoxic CD8+ T cells (CD8 CTLs). Here we show that EBV-driven CD4 CTLs exhibit prolonged proliferation and sustained cytotoxicity compared with CD8 CTLs, although their cytotoxic function markedly decreased during long-term culture. Comparative transcriptomic analysis of CD4 CTLs with varying cytotoxic activities identified B7-H3 and LAG3 as surface molecules associated with highly cytotoxic CD4 CTLs. Co-expression of B7-H3 and LAG3 correlated with CD107a expression and was observed on CD4+ T cells with enhanced cytotoxic potential in a target-dependent manner but not on CD8 CTLs. Furthermore, B7-H3+LAG3+ CD4+ T cells were induced during co-culture with bone marrow cells from pediatric patients with B-cell acute lymphoblastic leukemia (B-ALL). These findings suggest that B7-H3 and LAG3 co-expression represents a characteristic feature of functional CD4 CTLs in humans, providing valuable insights into the role of CD4 CTLs in tumor immunity.
Collapse
Affiliation(s)
- Yumi Tamura
- Department of Immunology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Shun Ohki
- Department of Immunology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Haruna Nagai
- Department of Immunology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Rin Yoshizato
- Department of Immunology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Shizuki Nishi
- Department of Immunology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Yuqi Jin
- Department of Immunology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Yasuo Kitajima
- Department of Immunology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Yun Guo
- Department of Immunology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Tatsuo Ichinohe
- Department of Hematology and Oncology, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
| | - Satoshi Okada
- Department of Pediatrics, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Yohei Kawano
- Department of Immunology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Tomoharu Yasuda
- Department of Immunology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| |
Collapse
|
11
|
Yoon JW, Kim KM, Cho S, Cho MJ, Park S, Hwang D, Kim HR, Park SH, Cho JH, Jeong H, Choi JM. Th1-poised naive CD4 T cell subpopulation reflects anti-tumor immunity and autoimmune disease. Nat Commun 2025; 16:1962. [PMID: 40000667 PMCID: PMC11861895 DOI: 10.1038/s41467-025-57237-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Accepted: 02/13/2025] [Indexed: 02/27/2025] Open
Abstract
Naïve CD4 T cells are traditionally viewed as a quiescent, homogeneous, resting population, but emerging evidence reveals their heterogeneity, which can be crucial for understanding disease contexts and therapeutic outcomes. In this study, we identify distinct subpopulations within both murine and human naïve CD4 T cells by single cell-RNA-sequencing (scRNA-seq), particularly focusing on a subpopulation that expresses super-high levels of interleukin-7 receptor (IL-7Rsup-hi), along with CD97, IL-18R, and Ly6C. This subpopulation, absent in the thymus and peripherally induced, exhibits type 1 helper T cell (Th1)-poised characteristics and contributes to the inhibition of cancer progression in B16F10 tumor-bearing mice. In humans, this IL-7Rsup-hi subpopulation expressing CD97 correlates with the responsiveness to anti-PD-1 therapy in cancer patients and the disease state of multiple sclerosis. By elucidating the heterogeneity of naive CD4 T cells and identifying a Th1-poised subpopulation capable of robust type 1 responses, we highlight the importance of this heterogeneity in inflammatory conditions for defining the disease states and predicting drug responsiveness.
Collapse
Affiliation(s)
- Jae-Won Yoon
- Department of Life Science, College of Natural Sciences, Hanyang University, Seoul, 04763, Republic of Korea
| | - Kyung Min Kim
- Department of Biological Sciences, Seoul National University, Seoul, Korea
| | - Sookyung Cho
- Department of Life Science, College of Natural Sciences, Hanyang University, Seoul, 04763, Republic of Korea
| | - Min-Ji Cho
- Department of Life Science, College of Natural Sciences, Hanyang University, Seoul, 04763, Republic of Korea
| | - Seonjun Park
- Department of Biological Sciences, Ulsan National Institute of Science & Technology (UNIST), Ulsan, Republic of Korea
| | - Daehee Hwang
- Department of Biological Sciences, Seoul National University, Seoul, Korea
| | - Hye Ryun Kim
- Division of Medical Oncology, Department of Internal Medicine, Yonsei Cancer Center Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Sung Ho Park
- Department of Biological Sciences, Ulsan National Institute of Science & Technology (UNIST), Ulsan, Republic of Korea
| | - Jae-Ho Cho
- Medical Research Center for Combinatorial Tumor Immunotherapy, Department of Microbiology and Immunology, Chonnam National University Medical School, Hwasun, 58128, Korea
| | - Hyobin Jeong
- Department of Systems Biology, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, South Korea.
| | - Je-Min Choi
- Department of Life Science, College of Natural Sciences, Hanyang University, Seoul, 04763, Republic of Korea.
- Hanyang Institute of Bioscience and Biotechnology, Hanyang University, Seoul, Republic of Korea.
- Research Institute for Natural Sciences, Hanyang University, Seoul, Republic of Korea.
- Research Institute for Convergence of Basic Sciences, Hanyang University, Seoul, 04763, Republic of Korea.
| |
Collapse
|
12
|
Bertrand L, Nelde A, Ramirez BC, Hatin I, Arbes H, François P, Demais S, Labaronne E, Decimo D, Guiguettaz L, Grégoire S, Bet A, Beauclair G, Gross A, Ziegler MC, Pereira M, Jeger-Madiot R, Verdier Y, Vinh J, Cardinaud S, Graff-Dubois S, Esclatine A, Gouttefangeas C, Altfeld M, Hocqueloux L, Samri A, Autran B, Lambotte O, Rammensee HG, Ricci EP, Walz J, Namy O, Moris A. Unveiling conserved HIV-1 open reading frames encoding T cell antigens using ribosome profiling. Nat Commun 2025; 16:1707. [PMID: 39966340 PMCID: PMC11836469 DOI: 10.1038/s41467-025-56773-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 01/29/2025] [Indexed: 02/20/2025] Open
Abstract
The development of ribosomal profiling (Riboseq) revealed the immense coding capacity of human and viral genomes. Here, we used Riboseq to delineate the translatome of HIV-1 in infected CD4+ T cells. In addition to canonical viral protein coding sequences (CDSs), we identify 98 alternative open reading frames (ARFs), corresponding to small Open Reading Frames (sORFs) that are distributed across the HIV genome including the UTR regions. Using a database of HIV genomes, we observe that most ARF amino-acid sequences are likely conserved among clade B and C of HIV-1, with 8 ARF-encoded amino-acid sequences being more conserved than the overlapping CDSs. Using T cell-based assays and mass spectrometry-based immunopeptidomics, we demonstrate that ARFs encode viral polypeptides. In the blood of people living with HIV, ARF-derived peptides elicit potent poly-functional T cell responses mediated by both CD4+ and CD8+ T cells. Our discovery expands the list of conserved viral polypeptides that are targets for vaccination strategies and might reveal the existence of viral microproteins or pseudogenes.
Collapse
Affiliation(s)
- Lisa Bertrand
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), 91190, Gif-sur-Yvette, France
- Sorbonne Université, Inserm U1135, CNRS ERL 8255, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), 75013, Paris, France
| | - Annika Nelde
- Department of Peptide-based Immunotherapy, University and University Hospital Tübingen, 72076, Tübingen, Germany
- Institute of Immunology, University of Tübingen, 72076, Tübingen, Germany
- Cluster of Excellence iFIT (EXC2180) "Image-Guided and Functionally Instructed Tumor Therapies", University of Tübingen, Tübingen, Germany
| | - Bertha Cecilia Ramirez
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), 91190, Gif-sur-Yvette, France
| | - Isabelle Hatin
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), 91190, Gif-sur-Yvette, France
| | - Hugo Arbes
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), 91190, Gif-sur-Yvette, France
| | - Pauline François
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), 91190, Gif-sur-Yvette, France
| | - Stéphane Demais
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), 91190, Gif-sur-Yvette, France
| | - Emmanuel Labaronne
- Laboratoire de Biologie et Modélisation de la Cellule, Ecole Normale Supérieure de Lyon, CNRS, UMR 5239, Inserm, U1293, Université Claude Bernard Lyon 1, 46 allée d'Italie F-69364, Lyon, France
- ADLIN Science, Evry-Courcouronnes, France
| | - Didier Decimo
- CIRI, Centre International de Recherche en Infectiologie, Univ Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, F-69007, Lyon, France
| | - Laura Guiguettaz
- Laboratoire de Biologie et Modélisation de la Cellule, Ecole Normale Supérieure de Lyon, CNRS, UMR 5239, Inserm, U1293, Université Claude Bernard Lyon 1, 46 allée d'Italie F-69364, Lyon, France
| | - Sylvie Grégoire
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), 91190, Gif-sur-Yvette, France
- Sorbonne Université, Inserm U1135, CNRS ERL 8255, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), 75013, Paris, France
| | - Anne Bet
- Sorbonne Université, Inserm U1135, CNRS ERL 8255, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), 75013, Paris, France
| | - Guillaume Beauclair
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), 91190, Gif-sur-Yvette, France
| | - Antoine Gross
- IRIM, UMR 9004, CNRS, Université de Montpellier, Montpellier, France
| | | | - Mathias Pereira
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), 91190, Gif-sur-Yvette, France
- Sorbonne Université, Inserm U1135, CNRS ERL 8255, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), 75013, Paris, France
| | - Raphaël Jeger-Madiot
- Sorbonne Université, Inserm U1135, CNRS ERL 8255, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), 75013, Paris, France
| | - Yann Verdier
- ESPCI Paris, PSL University, Spectrométrie de Masse Biologique et Protéomique, CNRS UAR2051, Paris, France
| | - Joelle Vinh
- ESPCI Paris, PSL University, Spectrométrie de Masse Biologique et Protéomique, CNRS UAR2051, Paris, France
| | - Sylvain Cardinaud
- Sorbonne Université, Inserm U1135, CNRS ERL 8255, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), 75013, Paris, France
- Vaccine Research Institute (VRI), INSERM-U955 (IMRB) Équipe 16, Université Paris-Est Créteil (UPEC), Créteil, France
| | - Stéphanie Graff-Dubois
- Sorbonne Université, Inserm U1135, CNRS ERL 8255, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), 75013, Paris, France
| | - Audrey Esclatine
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), 91190, Gif-sur-Yvette, France
| | - Cécile Gouttefangeas
- Institute of Immunology, University of Tübingen, 72076, Tübingen, Germany
- Cluster of Excellence iFIT (EXC2180) "Image-Guided and Functionally Instructed Tumor Therapies", University of Tübingen, Tübingen, Germany
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), partner site Tübingen, 72076, Tübingen, Germany
| | | | | | - Assia Samri
- Sorbonne Université, Inserm U1135, CNRS ERL 8255, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), 75013, Paris, France
| | - Brigitte Autran
- Sorbonne Université, Inserm U1135, CNRS ERL 8255, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), 75013, Paris, France
| | - Olivier Lambotte
- Université Paris Saclay, Inserm, CEA, AP-HP, UMR1184 IDMIT, Department of Internal Medicine & Clinical Immunology, Bicêtre Hospital, Le Kremlin-Bicêtre, Bicêtre, France
| | - Hans-Georg Rammensee
- Institute of Immunology, University of Tübingen, 72076, Tübingen, Germany
- Cluster of Excellence iFIT (EXC2180) "Image-Guided and Functionally Instructed Tumor Therapies", University of Tübingen, Tübingen, Germany
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), partner site Tübingen, 72076, Tübingen, Germany
| | - Emiliano P Ricci
- Laboratoire de Biologie et Modélisation de la Cellule, Ecole Normale Supérieure de Lyon, CNRS, UMR 5239, Inserm, U1293, Université Claude Bernard Lyon 1, 46 allée d'Italie F-69364, Lyon, France
| | - Juliane Walz
- Department of Peptide-based Immunotherapy, University and University Hospital Tübingen, 72076, Tübingen, Germany
- Institute of Immunology, University of Tübingen, 72076, Tübingen, Germany
- Cluster of Excellence iFIT (EXC2180) "Image-Guided and Functionally Instructed Tumor Therapies", University of Tübingen, Tübingen, Germany
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), partner site Tübingen, 72076, Tübingen, Germany
- Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK), Department of Internal Medicine, University Hospital Tübingen, 72076, Tübingen, Germany
| | - Olivier Namy
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), 91190, Gif-sur-Yvette, France.
| | - Arnaud Moris
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), 91190, Gif-sur-Yvette, France.
- Sorbonne Université, Inserm U1135, CNRS ERL 8255, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), 75013, Paris, France.
| |
Collapse
|
13
|
Mohamed AO, Boone DT, Ferry SL, Peck MC, Santos AM, Soderholm HE, Wittling MC, Paulos C, Turk MJ, Huang YH. CD4 T cell depletion increases memory differentiation of endogenous and CAR T cells and enhances the efficacy of Super2 and IL-33-armored CAR T cells against solid tumors. J Immunother Cancer 2025; 13:e009994. [PMID: 39933839 PMCID: PMC11815418 DOI: 10.1136/jitc-2024-009994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 01/23/2025] [Indexed: 02/13/2025] Open
Abstract
BACKGROUND Responsiveness to chimeric antigen receptor (CAR) T cell therapy correlates with CAR T cell expansion and persistence in vivo. Multiple strategies improve persistence by increasing stem-like properties or sustaining CAR T cell activity with combination therapies. Here, we describe the intrinsic ability of CAR T cells to differentiate into memory T cells, the effect of cytokine armoring, and neoadjuvant CD4 depletion therapy on CAR and tumor-specific endogenous memory T cells. METHODS TRP1-specific or NKG2D CAR T cells alone or with Super2+IL-33 (S233) armoring and/or CD4 depletion were evaluated in immunocompetent B16F10 melanoma or MC38 colon cell carcinoma models without preconditioning. We characterized CAR and endogenous tumor-specific memory T cell precursors, establishment of circulating (TCIRC) and resident (TRM) memory T cell subsets, and ability to protect against secondary tumors. RESULTS TRP1-specific or NKG2D CAR T cells had no effect on primary tumor growth in immunocompetent mice unless they were combined with S233 armoring or CD4 depletion. Unarmored CAR T cells expressed a stem-like phenotype in the tumor-draining lymph node and differentiated into CAR TCIRC memory cells in lymphoid organs and CAR TRM cells in the skin. In contrast, S233-armored CAR T cells exhibited an activated effector phenotype and differentiated inefficiently into CAR effector and central memory T cells. Combining CD4 therapy with unarmored CAR T cells increased CAR TCIRC and TRM memory T cells. Either CD4 depletion therapy or S233-armored CAR T cells induced activation of tumor-specific endogenous T cells that differentiated into both TCIRC and TRM memory T cells. CD4 depletion and S233-armored CAR T cell combination therapy synergized to increase endogenous memory T cells. CONCLUSIONS Unarmored TRP-1-specific or NKG2D CAR T cells have intrinsic stem-like properties and differentiate into memory T cell subsets but are non-protective against primary or secondary tumors. S233 cytokine armoring alone or with CD4 depletion improved effector responses but limited CAR memory T cell generation. S233-armored CAR T cells or CD4 depletion therapy induced endogenous tumor-specific TCIRC and TRM T cells, but the combination potentiated endogenous memory T cell generation and resulted in improved protection against B16F10 rechallenge.
Collapse
Affiliation(s)
- Asmaa O Mohamed
- Department of Microbiology & Immunology, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire, USA
| | - David Tyler Boone
- Department of Microbiology & Immunology, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire, USA
| | - Shannon L Ferry
- Department of Microbiology & Immunology, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire, USA
| | - Melanie C Peck
- Department of Microbiology & Immunology, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire, USA
| | - Alicia M Santos
- Department of Microbiology & Immunology, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire, USA
| | - Haille E Soderholm
- Department of Microbiology & Immunology, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire, USA
| | | | - Chrystal Paulos
- Department of Surgery, Emory University, Atlanta, Georgia, USA
- Emory University Winship Cancer Institute, Atlanta, Georgia, USA
| | - Mary Jo Turk
- Department of Microbiology & Immunology, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire, USA
- Dartmouth Cancer Center, Lebanon, New Hampshire, USA
| | - Yina H Huang
- Department of Microbiology & Immunology, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire, USA
- Dartmouth Cancer Center, Lebanon, New Hampshire, USA
- Pathology and Laboratory Medicine, Dartmouth Health, Lebanon, New Hampshire, USA
| |
Collapse
|
14
|
Fusagawa M, Tokita S, Murata K, Mariya T, Umemoto M, Sugita S, Matsuo K, Hirohashi Y, Saito T, Kanaseki T, Torigoe T. Identification and Phenotypic Characterization of Neoantigen-Specific Cytotoxic CD4+ T Cells in Endometrial Cancer. Cancer Immunol Res 2025; 13:171-184. [PMID: 39655805 DOI: 10.1158/2326-6066.cir-24-0514] [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: 05/30/2024] [Revised: 08/14/2024] [Accepted: 11/12/2024] [Indexed: 02/04/2025]
Abstract
Tumor-reactive CD4+ T cells often accumulate in the tumor microenvironment (TME) in human cancer, but their functions and roles in antitumor responses remain elusive. Here, we investigated the immunopeptidome of HLA class II-positive (HLA-II+) endometrial cancer with an inflamed TME using a proteogenomic approach. We identified HLA-II neoantigens, one of which induced polyclonal CD4+ tumor-infiltrating lymphocyte responses. We then experimentally demonstrated that neoantigen-specific CD4+ tumor-infiltrating lymphocytes lyse target cells in an HLA-II-dependent manner. Single-cell transcriptomic analysis of the TME coupled with T-cell receptor sequencing revealed the presence of CD4+ T-cell clusters characterized by CXCL13 expression. The CXCL13+ clusters contained two subclusters with distinct cytotoxic gene expression patterns. The identified neoantigen-specific CD4+ T cells were found exclusively in one of the CXCL13+ subclusters characterized by granzyme B and CCL5 expression. These results demonstrate the involvement of tumor-reactive CD4+ T cells with cytotoxic function in immune surveillance of endometrial cancer and reveal their transcriptomic signature.
Collapse
Affiliation(s)
- Minami Fusagawa
- Department of Pathology, Sapporo Medical University, Sapporo, Japan
| | - Serina Tokita
- Department of Pathology, Sapporo Medical University, Sapporo, Japan
- Joint Research Center for Immunoproteogenomics, Sapporo Medical University, Sapporo, Japan
| | - Kenji Murata
- Department of Pathology, Sapporo Medical University, Sapporo, Japan
- Joint Research Center for Immunoproteogenomics, Sapporo Medical University, Sapporo, Japan
| | - Tasuku Mariya
- Department of Obstetrics and Gynecology, Sapporo Medical University, Sapporo, Japan
| | - Mina Umemoto
- Department of Obstetrics and Gynecology, Sapporo Medical University, Sapporo, Japan
| | - Shintaro Sugita
- Department of Surgical Pathology, Sapporo Medical University, Sapporo, Japan
| | | | | | - Tsuyoshi Saito
- Department of Obstetrics and Gynecology, Sapporo Medical University, Sapporo, Japan
| | - Takayuki Kanaseki
- Department of Pathology, Sapporo Medical University, Sapporo, Japan
- Joint Research Center for Immunoproteogenomics, Sapporo Medical University, Sapporo, Japan
| | | |
Collapse
|
15
|
Hiraga T. Immune microenvironment of cancer bone metastasis. Bone 2025; 191:117328. [PMID: 39549899 DOI: 10.1016/j.bone.2024.117328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 11/06/2024] [Accepted: 11/08/2024] [Indexed: 11/18/2024]
Abstract
Bone is a common and frequent site of metastasis in cancer patients, leading to a significant reduction in quality of life and increased mortality. Bone marrow, the primary site of hematopoiesis, also serves as both a primary and secondary lymphoid organ. It harbors and supports a diverse array of immune cells, thereby creating a distinct immune microenvironment. These immune cells engage in a range of activities, including anti-tumor, pro-tumor, or a combination of both, which influence the development and progression of bone metastases. Rapid advances in cancer immunotherapy have underscored its potential to eradicate bone metastases. However, clinical outcomes have not yet met expectations. To improve the efficacy of immunotherapy, it is crucial to gain a comprehensive and in-depth understanding of the immune microenvironment within bone metastases. This review provides an overview of the current understanding of the role of different immune cells, their anti-tumor and pro-tumor activities, and their overall contribution to bone metastasis.
Collapse
Affiliation(s)
- Toru Hiraga
- Department of Histology and Cell Biology, Matsumoto Dental University, Shiojiri, Nagano, Japan.
| |
Collapse
|
16
|
Zhong Y, Chen G, Chen M, Cui J, Tan Q, Xiao Z. Gene prediction of immune cells association between gut microbiota and colorectal cancer: a Mendelian randomization study. Front Immunol 2025; 16:1460936. [PMID: 39958359 PMCID: PMC11825486 DOI: 10.3389/fimmu.2025.1460936] [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: 07/07/2024] [Accepted: 01/06/2025] [Indexed: 02/18/2025] Open
Abstract
Background An increasing number of studies have revealed that gut microbiota influences the development and progression of Colorectal cancer (CRC). However, whether a causal relationship exists between the two remains unclear, and the role of immune cells in this context is not well understood. Objective To elucidate the causal relationship between gut microbiota and CRC and to explore the potential mediating role of circulating immune cells. Materials and methods To analyze the causal relationship between gut microbiota and CRC, we employed a univariable Mendelian randomization (UVMR) approach. Subsequently, a two-step multivariable Mendelian randomization (MVMR) to assess the potential mediating role of circulating immune cells. Primarily, applied the Inverse-Variance Weighted method to evaluate the causal relationship between exposure and outcome. To ensure the robustness of the results linking gut microbiota and CRC, we validated the findings using Robust Inverse-Variance Weighted, Penalized Inverse-Variance Weighted, and Penalized Robust Inverse-Variance Weighted methods. Additionally, we employed MR-Egger Intercept to mitigate the influence of horizontal pleiotropy. MR-PRESSO was used to detect and correct outliers by excluding anomalous instrumental variables. Finally, we supplemented our analysis with methods such as Bayesian Weighted Mendelian Randomization (BWMR), Maximum-Likelihood, Lasso, Debiased Inverse Variance Weighted, and Contamination Mixture to establish a robust and compelling causal relationship. Results After accounting for reverse causality, horizontal pleiotropy, and various methodological corrections, Bifidobacterium kashiwanohense, GCA-900066755 sp900066755, Geminocystis, and Saccharofermentanaceae exhibited strong and robust causal effects on CRC. Specifically, CD40 on monocytes (2.82%) and CD45 on CD33+HLA-DR+CD14- cells (12.87%) mediated the causal relationship between Bifidobacterium kashiwanohense and CRC risk. Furthermore, CD45 on CD33-HLA-DR+ (3.94%) mediated the causal relationship between GCA-900066755 sp900066755 and CRC risk. Additionally, terminally differentiated CD4+T cells (11.55%) mediated the causal relationship between Geminocystis and CRC risk. Lastly, CD40 on monocytes (2.35%), central memory CD4+T cells (5.76%), and CD28 on CD28+CD45RA+CD8+T cells (5.00%) mediated the causal relationship between Saccharofermentanaceae and CRC risk. Conclusion Our mediation MR analysis provides genetic evidence suggesting that circulating immune cells may mediate the causal relationship between gut microbiota and CRC. The identified associations and mediation effects offer new insights into potential therapeutic avenues for CRC.
Collapse
Affiliation(s)
- Yan Zhong
- Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, China
| | - Guanglei Chen
- Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, China
| | - Menglu Chen
- Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, China
| | - Junsong Cui
- The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, China
| | - Qianren Tan
- The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, China
| | - Zhenghua Xiao
- The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, China
| |
Collapse
|
17
|
Martini E, Cremonesi M, Felicetta A, Serio S, Puccio S, Pelamatti E, van Beek JJP, Papadopoulou V, Catalano C, Fanuele F, Giuliano D, Basso G, Bonfiglio CA, Panico C, Vacchiano M, Carullo P, Papa L, D'Andrea C, Tuzger N, Marchini S, Magistroni P, Deaglio S, Amoroso A, Lugli E, Condorelli G, Kallikourdis M. Autoimmune-Like Mechanism in Heart Failure Enables Preventive Vaccine Therapy. Circ Res 2025; 136:4-25. [PMID: 39629560 DOI: 10.1161/circresaha.124.324999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 10/31/2024] [Accepted: 11/20/2024] [Indexed: 01/04/2025]
Abstract
BACKGROUND Heart failure (HF) is strongly associated with inflammation. In pressure overload (PO)-induced HF, cardiac stress triggers adaptive immunity, ablation or inhibition of which blocks disease progression. We hypothesized that PO-HF might fulfill the often-used criteria of autoimmunity: if so, the associated adaptive immune response would be not only necessary but also sufficient to induce HF; it should also be possible to identify self-antigens driving the autoimmune response. Finally, we hypothesized that such an antigen-specific response can be manipulated to preventively reduce the severity of PO-HF in a tolerizing vaccine. METHODS We used the transfer of lymphocytes or serum from PO-HF mice into healthy recipients to assess whether the adaptive response is sufficient to induce disease. We devised a novel pipeline to identify self-antigens driving the response. We immunized healthy mice with novel antigens to assess whether they induce disease. To determine whether these antigens could be present in human patients, we sought to detect existing responses against these antigens in patients with HF. Finally, we used the antigens in an oral tolerance protocol to preventively protect mice from subsequently induced PO-HF, analyzing the results with next-generation sequencing. RESULTS We found that PO-HF fulfills the criteria of an autoimmune disease, albeit partially, and identified novel cardiac self-antigens, capable of inducing cardiac dysfunction. The novel antigens in a tolerizing vaccine formulation preemptively reduced the severity of disease triggered by subsequent application of PO, via induction of effector regulatory T cells, enabling a potent reduction of PO-driven loss of systolic function, cardiac inflammation, and proinflammatory CD4+ T-cell clonal expansion. CONCLUSIONS We demonstrate that PO-HF is triggered by hemodynamic stress and then sets off an autoimmune-like response against cardiac self-antigens. The antigens can be used to reduce the severity of future-onset disease, via oral tolerization, effectively acting as a protective vaccine.
Collapse
Affiliation(s)
- Elisa Martini
- Adaptive Immunity Laboratory, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Humanitas Research Hospital, Rozzano, Italy (E.M., M.C., E.P., V.P., C.C., F.F., C.A.B., N.T., M.K.)
| | - Marco Cremonesi
- Adaptive Immunity Laboratory, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Humanitas Research Hospital, Rozzano, Italy (E.M., M.C., E.P., V.P., C.C., F.F., C.A.B., N.T., M.K.)
| | - Arianna Felicetta
- Cardio Center, IRCCS Humanitas Research Hospital, Rozzano, Italy (A.F., S.S., C.P., M.V., P.C., L.P., C.D., G.C.)
| | - Simone Serio
- Cardio Center, IRCCS Humanitas Research Hospital, Rozzano, Italy (A.F., S.S., C.P., M.V., P.C., L.P., C.D., G.C.)
- Institute of Genetics and Biomedical Research, Milan Unit, Consiglio Nazionale delle Ricerche, Italy (S.S., S.P.)
| | - Simone Puccio
- Laboratory of Translational Immunology, IRCCS Humanitas Research Hospital, Rozzano, Italy (S.P., J.J.P.v.B., E.L.)
- Institute of Genetics and Biomedical Research, Milan Unit, Consiglio Nazionale delle Ricerche, Italy (S.S., S.P.)
| | - Erica Pelamatti
- Adaptive Immunity Laboratory, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Humanitas Research Hospital, Rozzano, Italy (E.M., M.C., E.P., V.P., C.C., F.F., C.A.B., N.T., M.K.)
| | - Jasper J P van Beek
- Laboratory of Translational Immunology, IRCCS Humanitas Research Hospital, Rozzano, Italy (S.P., J.J.P.v.B., E.L.)
| | - Vasiliki Papadopoulou
- Adaptive Immunity Laboratory, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Humanitas Research Hospital, Rozzano, Italy (E.M., M.C., E.P., V.P., C.C., F.F., C.A.B., N.T., M.K.)
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy (V.P., C.C., F.F., C.P., C.D., N.T., G.C., M.K.)
| | - Chiara Catalano
- Adaptive Immunity Laboratory, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Humanitas Research Hospital, Rozzano, Italy (E.M., M.C., E.P., V.P., C.C., F.F., C.A.B., N.T., M.K.)
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy (V.P., C.C., F.F., C.P., C.D., N.T., G.C., M.K.)
| | - Francesca Fanuele
- Adaptive Immunity Laboratory, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Humanitas Research Hospital, Rozzano, Italy (E.M., M.C., E.P., V.P., C.C., F.F., C.A.B., N.T., M.K.)
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy (V.P., C.C., F.F., C.P., C.D., N.T., G.C., M.K.)
| | - Desirée Giuliano
- Humanitas Genomics Facility, IRCCS Humanitas Research Hospital, Rozzano, Italy (D.G., G.B., S.M.)
| | - Gianluca Basso
- Humanitas Genomics Facility, IRCCS Humanitas Research Hospital, Rozzano, Italy (D.G., G.B., S.M.)
| | - Cecilia Assunta Bonfiglio
- Adaptive Immunity Laboratory, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Humanitas Research Hospital, Rozzano, Italy (E.M., M.C., E.P., V.P., C.C., F.F., C.A.B., N.T., M.K.)
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-Universität München, Germany (C.A.B.)
| | - Cristina Panico
- Cardio Center, IRCCS Humanitas Research Hospital, Rozzano, Italy (A.F., S.S., C.P., M.V., P.C., L.P., C.D., G.C.)
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy (V.P., C.C., F.F., C.P., C.D., N.T., G.C., M.K.)
| | - Marco Vacchiano
- Cardio Center, IRCCS Humanitas Research Hospital, Rozzano, Italy (A.F., S.S., C.P., M.V., P.C., L.P., C.D., G.C.)
| | - Pierluigi Carullo
- Cardio Center, IRCCS Humanitas Research Hospital, Rozzano, Italy (A.F., S.S., C.P., M.V., P.C., L.P., C.D., G.C.)
| | - Laura Papa
- Cardio Center, IRCCS Humanitas Research Hospital, Rozzano, Italy (A.F., S.S., C.P., M.V., P.C., L.P., C.D., G.C.)
| | - Carla D'Andrea
- Cardio Center, IRCCS Humanitas Research Hospital, Rozzano, Italy (A.F., S.S., C.P., M.V., P.C., L.P., C.D., G.C.)
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy (V.P., C.C., F.F., C.P., C.D., N.T., G.C., M.K.)
| | - Naz Tuzger
- Adaptive Immunity Laboratory, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Humanitas Research Hospital, Rozzano, Italy (E.M., M.C., E.P., V.P., C.C., F.F., C.A.B., N.T., M.K.)
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy (V.P., C.C., F.F., C.P., C.D., N.T., G.C., M.K.)
| | - Sergio Marchini
- Humanitas Genomics Facility, IRCCS Humanitas Research Hospital, Rozzano, Italy (D.G., G.B., S.M.)
| | - Paola Magistroni
- Immunogenetics and Transplant Biology Service, Città della Salute e della Scienza Hospital, Turin, Italy (P.M., S.D., A.A.)
| | - Silvia Deaglio
- Immunogenetics and Transplant Biology Service, Città della Salute e della Scienza Hospital, Turin, Italy (P.M., S.D., A.A.)
- Department of Medical Sciences, University of Turin, Italy (S.D., A.A.)
| | - Antonio Amoroso
- Immunogenetics and Transplant Biology Service, Città della Salute e della Scienza Hospital, Turin, Italy (P.M., S.D., A.A.)
- Department of Medical Sciences, University of Turin, Italy (S.D., A.A.)
| | - Enrico Lugli
- Laboratory of Translational Immunology, IRCCS Humanitas Research Hospital, Rozzano, Italy (S.P., J.J.P.v.B., E.L.)
| | - Gianluigi Condorelli
- Cardio Center, IRCCS Humanitas Research Hospital, Rozzano, Italy (A.F., S.S., C.P., M.V., P.C., L.P., C.D., G.C.)
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy (V.P., C.C., F.F., C.P., C.D., N.T., G.C., M.K.)
| | - Marinos Kallikourdis
- Adaptive Immunity Laboratory, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Humanitas Research Hospital, Rozzano, Italy (E.M., M.C., E.P., V.P., C.C., F.F., C.A.B., N.T., M.K.)
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy (V.P., C.C., F.F., C.P., C.D., N.T., G.C., M.K.)
| |
Collapse
|
18
|
Yang GH, Ma XD, Wei XF, Liu RL, Wang C. A Novel KIF4A-related Model for Predicting Immunotherapy Response and Prognosis in Kidney Renal Clear Cell Carcinoma. Comb Chem High Throughput Screen 2025; 28:691-710. [PMID: 38357945 DOI: 10.2174/0113862073296897240212114403] [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: 12/22/2023] [Revised: 01/23/2024] [Accepted: 01/30/2024] [Indexed: 02/16/2024]
Abstract
BACKGROUND The efficacy of chemotherapy in treating Kidney Renal Clear Cell Carcinoma (KIRC) is limited, whereas immunotherapy has shown some promising clinical outcomes. In this context, KIF4A is considered a potential therapeutic target for various cancers. Therefore, identifying the mechanism of KIF4A that can predict the prognosis and immunotherapy response of KIRC would be of significant importance. METHODS Based on the TCGA Pan-Cancer dataset, the prognostic significance of the KIF4A expression across 33 cancer types was analyzed by univariate Cox algorithm. Furthermore, overlapping differentially expressed genes (DEGs1) between the KIF4A high- and lowexpression groups and DEGs2 between the KIRC and normal groups were also analyzed. Machine learning and Cox regression algorithms were performed to obtain biomarkers and construct a prognostic model. Finally, the role of KIF4A in KIRC was analyzed using quantitative real-time PCR, transwell assay, and EdU experiment. RESULTS Our analysis revealed that KIF4A was significant for the prognosis of 13 cancer types. The highest correlation with KIF4A was found for KICH among the tumour mutation burden (TMB) indicators. Subsequently, a prognostic model developed with UBE2C, OTX1, PPP2R2C, and RFLNA was obtained and verified with the Renal Cell Cancer-EU/FR dataset. There was a positive correlation between risk score and immunotherapy. Furthermore, the experiment results indicated that KIF4A expression was considerably increased in the KIRC group. Besides, the proliferation, migration, and invasion abilities of KIRC tumor cells were significantly weakened after KIF4A was knocked out. CONCLUSION We identified four KIF4A-related biomarkers that hold potential for prognostic assessment in KIRC. Specifically, early implementation of immunotherapy targeting these biomarkers may yield improved outcomes for patients with KIRC.
Collapse
Affiliation(s)
- Guang Hua Yang
- Department of Urology, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, People's Republic of China
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Xu Dong Ma
- Department of Urology, Baotou Central Hospital, Inner Mongolia Medical University, Baotou, China
| | - Xi Feng Wei
- Department of Urology, People's Hospital of Ningxia Hui Autonomous Region, Yinchuan, China
| | - Ran Lu Liu
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Chao Wang
- Department of Urology, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, People's Republic of China
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| |
Collapse
|
19
|
Nurlaila I, Rajabto W, Ningsih FN, Listiyaningsih E, Ratih US. Impaired Granularity in T cell Subsets but not in B cell Favors the Carcinogenesis of the Breast: A Preliminary Study in Indonesian Women Cohort. Asian Pac J Cancer Prev 2025; 26:225-231. [PMID: 39874005 DOI: 10.31557/apjcp.2025.26.1.225] [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: 08/20/2024] [Indexed: 01/30/2025] Open
Abstract
OBJECTIVE The progress made in cancer immunology has led to the development of innovative therapeutic strategies. However, despite these advances, the superficial characteristics of immune cells have been frequently overlooked: This oversight may be attributed to a limited understanding of the intricate relationships between immune cells and their microenvironment. This study seeks to address this limitation by comprehensively examining cell size and granularity in breast cancer (BC) patients and healthy donors (HD). METHODS Peripheral blood mononuclear cell (PBMC) samples were isolated from BC patients and HD. We examined the size (FSC-A%rCV) and granularity (SSC-A%rCV) of immune cell subsets in both patient groups and HD using flowcytometry. RESULTS Despite the absence of statistically significant variations in cell size between BC and HD, visual examination reveals noticeable discrepancies. There is a substantial decrease in granularity in CD8 and CD4 T-cell populations in BC compared to HD which is not observed in B cells. CONCLUSION Our analysis shows that while the size of immune cells may not be significantly altered in breast cancer patients compared to healthy donors, a closer examination of cell granularity reveals a distinct pattern. Specifically, the T-cell populations, including CD8 and CD4 cells, exhibit a substantial decrease in granularity in BC compared to HD. In contrast, B cells remain unaffected, suggesting that the granularity of T cells is uniquely susceptible to perturbations in breast cancer. This observation highlights the importance of considering cell granularity as a critical aspect of immune cell function, particularly in the context of cancer development.
Collapse
Affiliation(s)
- Ika Nurlaila
- Research Center for Vaccine and Drugs, The National Research and Innovation Agency (BRIN), South Tangerang 15310, Indonesia
| | - Wulyo Rajabto
- Hematology-Medical Oncology, Department of Internal Medicine, Dr. Cipto Mangunkusumo Hospital/Faculty of Medicine Universitas Indonesia, Jakarta, 10340, Indonesia
| | - Febby Nurdiya Ningsih
- Research Center for Vaccine and Drugs, The National Research and Innovation Agency (BRIN), South Tangerang 15310, Indonesia
| | - Erlin Listiyaningsih
- Faculty of Medicine, Universitas Muhammadiyah Prof. Dr. HAMKA, Jakarta, 12130, Indonesia
| | - Udani Sari Ratih
- Research Center for Vaccine and Drugs, The National Research and Innovation Agency (BRIN), South Tangerang 15310, Indonesia
| |
Collapse
|
20
|
Kared H, Tan C, Narang V, Tan SW, Xian CH, Wei ATS, Lum J, Ruiz-Mateos E, Rajasuriar R, Kamarulzaman A, Ng TP, Larbi A. SLAMF7 defines subsets of human effector CD8 T cells. Sci Rep 2024; 14:30779. [PMID: 39730488 DOI: 10.1038/s41598-024-80971-5] [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: 07/08/2024] [Accepted: 11/22/2024] [Indexed: 12/29/2024] Open
Abstract
Long-term control of viral replication relies on the efficient differentiation of memory T cells into effector T cells during secondary immune responses. Recent findings have identified T cell precursors for both memory and exhausted T cells, suggesting the existence of progenitor-like effector T cells. These cells can persist without antigenic challenge but expand and acquire effector functions upon recall immune responses. In this study, we demonstrate that the combination of SLAMF7 with either CD27 or TCF-1 effectively identifies progenitor-like effector CD8 T cells, while SLAMF7 with GPR56 or TOX defines effector CD8 T cells. These markers allow for the clear segregation of these distinct cell subsets. SLAMF7+ CD8T cells are dynamically modulated during viral infections, including HIV, HCV, CMV, and SARS-CoV-2, as well as during aging. We further characterize the SLAMF7 signature at both phenotypic and transcriptional levels. Notably, during aging, the SLAMF7 pathway becomes dysregulated, resulting in persistent phosphorylation of STAT1. Additionally, SLAMF7 ligation in the presence of IL-15 induces TCF-1 expression, which promotes the homeostatic proliferation of progenitor-like effector CD8 T cells.
Collapse
Affiliation(s)
- Hassen Kared
- Singapore Immunology Network (SIgN), Agency for Science Technology and Research (A*STAR), Immunos Building, 8A Biomedical Grove, Biopolis, Republic of Singapore.
- Department of Immunology, Oslo University Hospital, Oslo, Norway.
- Precision Immunotherapy Alliance, University of Oslo, Oslo, Norway.
| | - Crystal Tan
- Singapore Immunology Network (SIgN), Agency for Science Technology and Research (A*STAR), Immunos Building, 8A Biomedical Grove, Biopolis, Republic of Singapore
| | - Vipin Narang
- Singapore Immunology Network (SIgN), Agency for Science Technology and Research (A*STAR), Immunos Building, 8A Biomedical Grove, Biopolis, Republic of Singapore
| | - Shu Wen Tan
- Singapore Immunology Network (SIgN), Agency for Science Technology and Research (A*STAR), Immunos Building, 8A Biomedical Grove, Biopolis, Republic of Singapore
| | - Chin Hui Xian
- Singapore Immunology Network (SIgN), Agency for Science Technology and Research (A*STAR), Immunos Building, 8A Biomedical Grove, Biopolis, Republic of Singapore
| | - Alicia Tay Seok Wei
- Singapore Immunology Network (SIgN), Agency for Science Technology and Research (A*STAR), Immunos Building, 8A Biomedical Grove, Biopolis, Republic of Singapore
| | - Josephine Lum
- Singapore Immunology Network (SIgN), Agency for Science Technology and Research (A*STAR), Immunos Building, 8A Biomedical Grove, Biopolis, Republic of Singapore
| | - Ezequiel Ruiz-Mateos
- Clinical Unit of Infectious Diseases, Microbiology and Preventive Medicine, Institute of Biomedicine of Seville (IBiS), Virgen del Rocío University Hospital, CSIC, University of Seville, Seville, Spain
| | - Reena Rajasuriar
- Centre of Excellence for Research in AIDS (CERiA), University of Malaya, Kuala Lumpur, Malaysia
- The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Australia
- Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Adeeba Kamarulzaman
- Centre of Excellence for Research in AIDS (CERiA), University of Malaya, Kuala Lumpur, Malaysia
- Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Tze Pin Ng
- Gerontology Research Programme and Department of Psychological Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Anis Larbi
- Singapore Immunology Network (SIgN), Agency for Science Technology and Research (A*STAR), Immunos Building, 8A Biomedical Grove, Biopolis, Republic of Singapore
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Republic of Singapore
| |
Collapse
|
21
|
Fan X, Brunetti TM, Jackson K, Roop DR. Single-Cell Profiling Reveals Global Immune Responses during the Progression of Murine Epidermal Neoplasms. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.24.630251. [PMID: 39763798 PMCID: PMC11703249 DOI: 10.1101/2024.12.24.630251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
Immune cells determine the role of the tumor microenvironment during tumor progression, either suppressing tumor formation or promoting tumorigenesis. We analyzed the profile of immune cells in the tumor microenvironment of control mouse skins and skin tumors at the single-cell level. We identified 15 CD45 + immune cell clusters, which broadly represent the most functionally characterized immune cell types including macrophages, Langerhans cells (LC), conventional type 1 dendritic cells (cDC1), conventional type 2 dendritic cells (cDC2), migratory/mature dendritic cells (mDC), dendritic epidermal T cells (DETC), dermal γδ T cells (γδT), T cells, regulatory T cells (Tregs), natural killer cells (NK), type 2 innate lymphoid cells (ILC2), neutrophils (Neu), mast cells (Mast), and two proliferating populations (Prolif.1 and Prolif.2). Skin tumor progression reprogramed immune cells and led to a marked increase in the relative percentages of macrophages, cDC2, mDC, Tregs, and Neu. Macrophages, the largest cell cluster of immune cells in skin tumors. In addition, macrophages emerged as the predominant communication 'hub' in skin tumors, highlighting the importance of macrophages during skin tumor progression. In contrast, other immune cell clusters decreased during skin tumor progression, including DETC, γδT, ILC2, and LC. In addition, skin tumor progression dramatically upregulated Jak2/Stat3 expression and the interferon response across various immune cell clusters. Further, skin tumor progression activated T cells and NK cells indicated by elevated expression of IFN-γ and Granzyme B in skin tumors. Meanwhile, a pronounced infiltration of M2-macrophages and Tregs in skin tumors created an immunosuppressive microenvironment, consistent with the elevated expression of the Stat3 pathway in skin tumors. In summary, our study elucidates the immune cell landscape of epidermal neoplasms, offering a comprehensive understanding of the immune response during skin tumor progression and providing new insights into cancer immune evasion mechanisms.
Collapse
|
22
|
Ma YY, Zhou WY, Qian Y, Mu YY, Zhang W. SOX13 as a potential prognostic biomarker linked to immune infiltration and ferroptosis inhibits the proliferation, migration, and metastasis of thyroid cancer cells. Front Immunol 2024; 15:1478395. [PMID: 39726600 PMCID: PMC11670200 DOI: 10.3389/fimmu.2024.1478395] [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: 08/09/2024] [Accepted: 11/25/2024] [Indexed: 12/28/2024] Open
Abstract
Background SOX13 is a transcription factor belonging to the SOX family. SOX proteins are critical regulators of multiple cancer progression, and some are known to control carcinogenesis. Nevertheless, the functional and clinical significance of SOX13 in human thyroid cancer (THCA) remain largely unelucidated. Methods Data on SOX13 expression were obtained through The Cancer Genome Atlas together with Gene Expression Omnibus. Co-expression, differential expression, and functional analyses of genes were investigated by databases. Associations between SOX13 levels, immune infiltration, ferroptosis, and immune checkpoint gene levels were analyzed. Genetic changes in SOX13 were investigated using CBioPortal. Associations between SOX13 levels and THCA clinicopathological features were analyzed and nomogram modeling for diagnostic and prognostic prediction. The influence of SOX13 on proliferation, migration, and metastasis was determined in KTC-1 and TPC-1 cell lines. Results SOX13 was significantly lower in THCA tumors compared to controls. In addition, upregulated SOX13 gene mutation were evident in thyroid cancer. SOX13-associated genes exhibited differential expression in pathways associated with thyroid cancer development. Significant associations were found between SOX13 levels, immune infiltration, ferroptosis, and immune checkpoint genes in THCA tissue. SOX13 levels correlated with THCA stage, histologic grade, and primary neoplasm focus types, and independently predicted overall and progression-free intervals. SOX13 expression effectively distinguished between tumor and normal thyroid tissue. Spearman correlations highlighted a significant relationship between SOX13 and ferroptosis-associated genes. Overexpression of SOX13 enhances the inhibition of RSL3 (iron death activator) on the cell viability of TPC-1. Higher SOX13 levels in Thyroid cancer cells may lead to reduced proliferation, migration, and metastasis by regulating ferroptosis. Conclusion Reduced SOX13 expression inversely impacts patient prognosis. In addition, SOX13 strongly regulates cancer immunity and Ferroptosis. Hence, SOX13 has great promise as a bioindicator for both thyroid cancer prognosis and immune cell invasion.
Collapse
Affiliation(s)
- Yan-yan Ma
- Department of Rehabilitation Medicine, Beijing Jishuitan Hospital Guizhou Hospital, Guiyang, Guizhou, China
| | - Wei-ye Zhou
- Cell Biology Department, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China
| | - Yue Qian
- Department of Pathogen Biology, Guizhou Nursing Vocational College, Guiyang, Guizhou, China
| | - Ying-ying Mu
- Department of Pathology, Zunyi Hospital of Traditional Chinese Medicine, Zunyi, Guizhou, China
| | - Wei Zhang
- Cell Biology Department, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China
- Department of Pathogen Biology, Guizhou Nursing Vocational College, Guiyang, Guizhou, China
| |
Collapse
|
23
|
Yang X, Zhen C, Huang H, Jiao Y, Fan X, Zhang C, Song J, Wang S, Zhou C, Yang X, Yuan J, Zhang J, Xu R, Wang FS. Implications of accumulation of clonally expanded and senescent CD4 +GNLY + T cells in immunological non-responders of HIV-1 infection. Emerg Microbes Infect 2024; 13:2396868. [PMID: 39239709 PMCID: PMC11441045 DOI: 10.1080/22221751.2024.2396868] [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: 04/01/2024] [Revised: 08/02/2024] [Accepted: 08/22/2024] [Indexed: 09/07/2024]
Abstract
Increased CD4+GNLY+ T cells have been confirmed to be inversely associated with CD4+ T cell count in immunological non-responders (INRs), however, the underlying mechanisms are unknown. This study aimed to elucidate the characteristics of CD4+GNLY+ T cells and their relationship with immune restoration. Single-cell RNA sequencing, single-cell TCR sequencing, and flow cytometry were used to analyze the frequency, phenotypes, and function of CD4+GNLY+ T cells. Moreover, Enzyme linked immunosorbent assay was performed to detect plasma cytokines production in patients. CD4+GNLY+ T cells were found to be highly clonally expanded, characterized by higher levels of cytotoxicity, senescence, P24, and HIV-1 DNA than CD4+GNLY- T cells. Additionally, the frequency of CD4+GNLY+ T cells increased after ART, and further increased in INRs, and were positively associated with the antiretroviral therapy duration in INR. Furthermore, increased IL-15 levels in INRs positively correlated with the frequency and senescence of CD4+GNLY+ T cells, suggesting that CD4+GNLY+ T cells may provide new insights for understanding the poor immune reconstitution of INRs. In conclusion, increased, highly clonally expanded, and senescent CD4+GNLY+ T cells may contribute to poor immune reconstitution in HIV-1 infection.
Collapse
Affiliation(s)
- Xiuhan Yang
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, People’s Republic of China
- Senior Department of Infectious Diseases, The Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, People’s Republic of China
| | - Cheng Zhen
- Senior Department of Infectious Diseases, The Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, People’s Republic of China
| | - Huihuang Huang
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, People’s Republic of China
- Senior Department of Infectious Diseases, The Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, People’s Republic of China
| | - Yanmei Jiao
- Senior Department of Infectious Diseases, The Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, People’s Republic of China
| | - Xing Fan
- Senior Department of Infectious Diseases, The Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, People’s Republic of China
| | - Chao Zhang
- Senior Department of Infectious Diseases, The Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, People’s Republic of China
| | - Jinwen Song
- Senior Department of Infectious Diseases, The Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, People’s Republic of China
| | - Songshan Wang
- Senior Department of Infectious Diseases, The Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, People’s Republic of China
| | - Chunbao Zhou
- Senior Department of Infectious Diseases, The Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, People’s Republic of China
| | - XinXin Yang
- Senior Department of Infectious Diseases, The Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, People’s Republic of China
| | - Jinhong Yuan
- Senior Department of Infectious Diseases, The Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, People’s Republic of China
| | - Jiyuan Zhang
- Senior Department of Infectious Diseases, The Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, People’s Republic of China
| | - Ruonan Xu
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, People’s Republic of China
- Senior Department of Infectious Diseases, The Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, People’s Republic of China
| | - Fu-Sheng Wang
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, People’s Republic of China
- Senior Department of Infectious Diseases, The Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, People’s Republic of China
| |
Collapse
|
24
|
Lawton ML, Inge MM, Blum BC, Smith-Mahoney EL, Bolzan D, Lin W, McConney C, Porter J, Moore J, Youssef A, Tharani Y, Varelas X, Denis GV, Wong WW, Padhorny D, Kozakov D, Siggers T, Wuchty S, Snyder-Cappione J, Emili A. Multiomic profiling of chronically activated CD4+ T cells identifies drivers of exhaustion and metabolic reprogramming. PLoS Biol 2024; 22:e3002943. [PMID: 39689157 DOI: 10.1371/journal.pbio.3002943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 01/06/2025] [Accepted: 11/15/2024] [Indexed: 12/19/2024] Open
Abstract
Repeated antigen exposure leads to T-cell exhaustion, a transcriptionally and epigenetically distinct cellular state marked by loss of effector functions (e.g., cytotoxicity, cytokine production/release), up-regulation of inhibitory receptors (e.g., PD-1), and reduced proliferative capacity. Molecular pathways underlying T-cell exhaustion have been defined for CD8+ cytotoxic T cells, but which factors drive exhaustion in CD4+ T cells, that are also required for an effective immune response against a tumor or infection, remains unclear. Here, we utilize quantitative proteomic, phosphoproteomic, and metabolomic analyses to characterize the molecular basis of the dysfunctional cell state induced by chronic stimulation of CD4+ memory T cells. We identified a dynamic response encompassing both known and novel up-regulated cell surface receptors, as well as dozens of unexpected transcriptional regulators. Integrated causal network analysis of our combined data predicts the histone acetyltransferase p300 as a driver of aspects of this phenotype following chronic stimulation, which we confirmed via targeted small molecule inhibition. While our integrative analysis also revealed large-scale metabolic reprogramming, our independent investigation confirmed a global remodeling away from glycolysis to a dysfunctional fatty acid oxidation-based metabolism coincident with oxidative stress. Overall, these data provide both insights into the mechanistic basis of CD4+ T-cell exhaustion and serve as a valuable resource for future interventional studies aimed at modulating T-cell dysfunction.
Collapse
Affiliation(s)
- Matthew L Lawton
- Center for Network Systems Biology, Boston University School of Medicine, Boston, Massachusetts, United States of America
- Department of Biochemistry, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Melissa M Inge
- Depart of Biology, Boston University, Boston, Massachusetts, United States of America
| | - Benjamin C Blum
- Center for Network Systems Biology, Boston University School of Medicine, Boston, Massachusetts, United States of America
- Department of Biochemistry, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Erika L Smith-Mahoney
- Department of Microbiology, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Dante Bolzan
- Department of Computer Science, University of Miami, Miami, Florida, United States of America
| | - Weiwei Lin
- Center for Network Systems Biology, Boston University School of Medicine, Boston, Massachusetts, United States of America
- Department of Biochemistry, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Christina McConney
- Department of Microbiology, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Jacob Porter
- Knight Cancer Institute, Oregon Health and Science University, Portland, Oregon, United States of America
| | - Jarrod Moore
- Center for Network Systems Biology, Boston University School of Medicine, Boston, Massachusetts, United States of America
- Department of Biochemistry, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Ahmed Youssef
- Center for Network Systems Biology, Boston University School of Medicine, Boston, Massachusetts, United States of America
- Department of Biochemistry, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Yashasvi Tharani
- Center for Network Systems Biology, Boston University School of Medicine, Boston, Massachusetts, United States of America
- Depart of Biology, Boston University, Boston, Massachusetts, United States of America
| | - Xaralabos Varelas
- Department of Biochemistry, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Gerald V Denis
- Hematology and Medical Oncology, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Wilson W Wong
- Department of Biomedical Engineering, Boston University, Boston, Massachusetts, United States of America
| | - Dzmitry Padhorny
- Department of Applied Mathematics and Statistics, Stony Brook University, Stony Brook, New York, United States of America
- Laufer Center for Physical and Quantitative Biology, Stony Brook University, Stony Brook, New York, United States of America
| | - Dima Kozakov
- Department of Applied Mathematics and Statistics, Stony Brook University, Stony Brook, New York, United States of America
- Laufer Center for Physical and Quantitative Biology, Stony Brook University, Stony Brook, New York, United States of America
| | - Trevor Siggers
- Depart of Biology, Boston University, Boston, Massachusetts, United States of America
| | - Stefan Wuchty
- Department of Computer Science, University of Miami, Miami, Florida, United States of America
- Miami Institute of Data Science and Computing, Miami, Florida, United States of America
| | - Jennifer Snyder-Cappione
- Department of Microbiology, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Andrew Emili
- Center for Network Systems Biology, Boston University School of Medicine, Boston, Massachusetts, United States of America
- Department of Biochemistry, Boston University School of Medicine, Boston, Massachusetts, United States of America
- Depart of Biology, Boston University, Boston, Massachusetts, United States of America
- Knight Cancer Institute, Oregon Health and Science University, Portland, Oregon, United States of America
| |
Collapse
|
25
|
Lei X, Xiao R, Chen Z, Ren J, Zhao W, Tang W, Wen K, Zhu Y, Li X, Ouyang S, Xu A, Hu Y, Bi E. Augmenting antitumor efficacy of Th17-derived Th1 cells through IFN-γ-induced type I interferon response network via IRF7. Proc Natl Acad Sci U S A 2024; 121:e2412120121. [PMID: 39541355 PMCID: PMC11588128 DOI: 10.1073/pnas.2412120121] [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: 06/19/2024] [Accepted: 10/14/2024] [Indexed: 11/16/2024] Open
Abstract
The importance of CD4+ T cells in cancer immunotherapy has gained increasing recognition. Particularly, a specific subset of CD4+ T cells coexpressing the T helper type 1 (Th1) and Th17 markers has demonstrated remarkable antitumor potential. However, the underlying mechanisms governing the differentiation of these cells and their subsequent antitumor responses remain incompletely understood. Single-cell RNA sequencing (scRNA-seq) data reanalysis demonstrated the presence of Th171 cells within tumors. Subsequent trajectory analysis found that these Th171 cells are initially primed under Th17 conditions and then converted into IFN-γ-producing cells. Following the in vivo differentiation trajectory of Th171 cells, we successfully established in vitro Th171 cell culture. Transcriptomic profiling has unveiled a substantial resemblance between in vitro-generated Th171 cells and their tumor-infiltrating counterparts. Th171 cells exhibit more potent antitumor responses than Th1 or Th17 cells. Additionally, Th171chimeric antigen receptor T (CAR-T) cells eradicate solid tumors more efficiently. Importantly, Th171 cells display an early exhaustion phenotype while retaining stemness. Mechanistically, Th171 cells migrate faster and accumulate more in tumors in an extracellular matrix protein 1 (ECM1)-dependent manner. Furthermore, we show that IFN-γ up-regulated IRF7 to promote the type I interferon response network and ECM1 expression but decreased the exhaustion status in Th171 cells. Taken together, our findings position Th171 cells as a great candidate for improving targeted immunotherapies in solid malignancies.
Collapse
Affiliation(s)
- Xiaoyi Lei
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Guangdong Provincial Key Laboratory of Single Cell Technology and Application, Southern Medical University, Guangzhou, Guangdong510515, China
| | - Ruipei Xiao
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Guangdong Provincial Key Laboratory of Single Cell Technology and Application, Southern Medical University, Guangzhou, Guangdong510515, China
| | - Zhe Chen
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Guangdong Provincial Key Laboratory of Single Cell Technology and Application, Southern Medical University, Guangzhou, Guangdong510515, China
| | - Jie Ren
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Guangdong Provincial Key Laboratory of Single Cell Technology and Application, Southern Medical University, Guangzhou, Guangdong510515, China
| | - Wenli Zhao
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Guangdong Provincial Key Laboratory of Single Cell Technology and Application, Southern Medical University, Guangzhou, Guangdong510515, China
| | - Wenting Tang
- Division of Nephrology, State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Nanfang Hospital, Southern Medical University, Guangzhou510515, China
| | - Kang Wen
- Department of Urology, Zhujiang Hospital, Southern Medical University, Guangzhou510280, China
| | - Yihan Zhu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Guangdong Provincial Key Laboratory of Single Cell Technology and Application, Southern Medical University, Guangzhou, Guangdong510515, China
| | - Xinru Li
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Guangdong Provincial Key Laboratory of Single Cell Technology and Application, Southern Medical University, Guangzhou, Guangdong510515, China
| | - Suidong Ouyang
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan523808, China
| | - Abai Xu
- Department of Urology, Zhujiang Hospital, Southern Medical University, Guangzhou510280, China
| | - Yu Hu
- Division of Nephrology, State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Nanfang Hospital, Southern Medical University, Guangzhou510515, China
| | - Enguang Bi
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Guangdong Provincial Key Laboratory of Single Cell Technology and Application, Southern Medical University, Guangzhou, Guangdong510515, China
- Department of Urology, Zhujiang Hospital, Southern Medical University, Guangzhou510280, China
| |
Collapse
|
26
|
Tokita S, Kanaseki T, Torigoe T. Neoantigen prioritization based on antigen processing and presentation. Front Immunol 2024; 15:1487378. [PMID: 39569190 PMCID: PMC11576432 DOI: 10.3389/fimmu.2024.1487378] [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: 08/28/2024] [Accepted: 10/21/2024] [Indexed: 11/22/2024] Open
Abstract
Somatic mutations in tumor cells give rise to mutant proteins, fragments of which are often presented by MHC and serve as neoantigens. Neoantigens are tumor-specific and not expressed in healthy tissues, making them attractive targets for T-cell-based cancer immunotherapy. On the other hand, since most somatic mutations differ from patient to patient, neoantigen-targeted immunotherapy is personalized medicine and requires their identification in each patient. Computational algorithms and machine learning methods have been developed to prioritize neoantigen candidates. In fact, since the number of clinically relevant neoantigens present in a patient is generally limited, this process is like finding a needle in a haystack. Nevertheless, MHC presentation of neoantigens is not random but follows certain rules, and the efficiency of neoantigen detection may be further improved with technological innovations. In this review, we discuss current approaches to the detection of clinically relevant neoantigens, with a focus on antigen processing and presentation.
Collapse
Affiliation(s)
- Serina Tokita
- Department of Pathology, Sapporo Medical University, Sapporo, Japan
- Joint Research Center for Immunoproteogenomics, Sapporo Medical University, Sapporo, Japan
| | - Takayuki Kanaseki
- Department of Pathology, Sapporo Medical University, Sapporo, Japan
- Joint Research Center for Immunoproteogenomics, Sapporo Medical University, Sapporo, Japan
| | | |
Collapse
|
27
|
Zhang C, Ren T, Zhao X, Su Y, Wang Q, Zhang T, He B, Chen Y, Wu LY, Sun L, Zhang B, Xia Z. Biologically informed machine learning modeling of immune cells to reveal physiological and pathological aging process. Immun Ageing 2024; 21:74. [PMID: 39449067 PMCID: PMC11515583 DOI: 10.1186/s12979-024-00479-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Accepted: 10/17/2024] [Indexed: 10/26/2024]
Abstract
The immune system undergoes progressive functional remodeling from neonatal stages to old age. Therefore, understanding how aging shapes immune cell function is vital for precise treatment of patients at different life stages. Here, we constructed the first transcriptomic atlas of immune cells encompassing human lifespan, ranging from newborns to supercentenarians, and comprehensively examined gene expression signatures involving cell signaling, metabolism, differentiation, and functions in all cell types to investigate immune aging changes. By comparing immune cell composition among different age groups, HLA highly expressing NK cells and CD83 positive B cells were identified with high percentages exclusively in the teenager (Tg) group, whereas unknown_T cells were exclusively enriched in the supercentenarian (Sc) group. Notably, we found that the biological age (BA) of pediatric COVID-19 patients with multisystem inflammatory syndrome accelerated aging according to their chronological age (CA). Besides, we proved that inflammatory shift- myeloid abundance and signature correlate with the progression of complications in Kawasaki disease (KD). The shift- myeloid signature was also found to be associated with KD treatment resistance, and effective therapies improve treatment outcomes by reducing this signaling. Finally, based on those age-related immune cell compositions, we developed a novel BA prediction model PHARE ( https://xiazlab.org/phare/ ), which can apply to both scRNA-seq and bulk RNA-seq data. Using this model, we found patients with coronary artery disease (CAD) also exhibit accelerated aging compared to healthy individuals. Overall, our study revealed changes in immune cell proportions and function associated with aging, both in health and disease, and provided a novel tool for successfully capturing features that accelerate or delay aging.
Collapse
Affiliation(s)
- Cangang Zhang
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, China
- Institute of Infection and Immunity, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China
- Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education, Xi'an, Shaanxi, China
| | - Tao Ren
- Academy of Mathematics and Systems Science, Chinese Academy of Sciences, Beijing, China
- School of Mathematical Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Xiaofan Zhao
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR, USA
| | - Yanhong Su
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, China
- Institute of Infection and Immunity, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China
- Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education, Xi'an, Shaanxi, China
| | - Qianhao Wang
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, China
- Institute of Infection and Immunity, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China
- Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education, Xi'an, Shaanxi, China
| | - Tianzhe Zhang
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, China
- Institute of Infection and Immunity, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China
- Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education, Xi'an, Shaanxi, China
| | - Boxiao He
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, China
| | - Yabing Chen
- Department of Pathology and Laboratory Medicine, Oregon Health & Science University, Portland, OR, USA
| | - Ling-Yun Wu
- Academy of Mathematics and Systems Science, Chinese Academy of Sciences, Beijing, China
- School of Mathematical Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Lina Sun
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, China.
- Institute of Infection and Immunity, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China.
- Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education, Xi'an, Shaanxi, China.
| | - Baojun Zhang
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, China.
- Institute of Infection and Immunity, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China.
- Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education, Xi'an, Shaanxi, China.
| | - Zheng Xia
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR, USA.
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA.
- Center for Biomedical Data Science, Oregon Health & Science University, Portland, OR, USA.
| |
Collapse
|
28
|
Zhang J, Cao J, Wang L, Li S, Meng F, Liang X, Jiang H, Luo R, Zhu D, Zhang F, Zhang L, Zhang X, Mei L. Neoantigen sequestrated autophagosomes as therapeutic cancer vaccines. J Control Release 2024; 376:369-381. [PMID: 39413847 DOI: 10.1016/j.jconrel.2024.10.019] [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: 05/17/2024] [Revised: 09/29/2024] [Accepted: 10/12/2024] [Indexed: 10/18/2024]
Abstract
Neoantigens serve as ideal personalized cancer vaccines because of their high immunogenicity, ability to evade central thymic tolerance, and minimal risk of eliciting autoimmune responses. Herein, we describe a genetically engineered autophagosome-based neoantigen vaccine (APNV) in combination with an immune checkpoint inhibitor (anti-PD-1 antibody) for cancer immunotherapy. The APNV was derived from engineered NIH 3T3 cells, which co-express melanoma neoantigens and autophagosome maker microtubule-associated proteins 1 A/1B light chain 3B (LC3), from which the LC3-labeled neoantigen-autophagosomes were isolated. These purified autophagosomes, in conjunction with vaccine adjuvants high-mobility group box 1 (HMGB1) and granulocyte-macrophage colony-stimulating factor (GM-CSF), were integrated into a hydrogel to create an APNV. The APNV effectively activated dendritic cells both in vitro and in vivo. Moreover, APNV, in combination with checkpoint blockade therapy, significantly hampered post-surgical tumor recurrence in a subcutaneous melanoma tumor model and effectively impeded metastatic progression in a melanoma lung metastasis model. This APNV may be conducive to making personalized therapeutic neoantigen vaccines for cancer immunotherapy.
Collapse
Affiliation(s)
- Jinxie Zhang
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, PR China
| | - Jiahui Cao
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, PR China
| | - Liuchang Wang
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, PR China
| | - Sitong Li
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, PR China
| | - Fanqiang Meng
- Shenzhen Key Laboratory for Systems Medicine in Inflammatory Diseases, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, Sun Yat-Sen University, Shenzhen 518107, Guangdong, China
| | - Xin Liang
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Key Laboratory of Stem Cell and Regenerative Tissue Engineering, School of Basic Medical Sciences, Guangdong Medical University, Dongguan 523808, China
| | - Hanyu Jiang
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Key Laboratory of Stem Cell and Regenerative Tissue Engineering, School of Basic Medical Sciences, Guangdong Medical University, Dongguan 523808, China; Guangdong Second Provincial General Hospital, Guangdong Medical University, Guangzhou 510317, PR China
| | - Ran Luo
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, PR China
| | - Dunwan Zhu
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, PR China
| | - Fan Zhang
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, PR China
| | - Linhua Zhang
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, PR China.
| | - Xudong Zhang
- Shenzhen Key Laboratory for Systems Medicine in Inflammatory Diseases, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, Sun Yat-Sen University, Shenzhen 518107, Guangdong, China.
| | - Lin Mei
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, PR China.
| |
Collapse
|
29
|
Montauti E, Oh DY, Fong L. CD4 + T cells in antitumor immunity. Trends Cancer 2024; 10:969-985. [PMID: 39242276 PMCID: PMC11464182 DOI: 10.1016/j.trecan.2024.07.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 07/24/2024] [Accepted: 07/25/2024] [Indexed: 09/09/2024]
Abstract
Advances in cancer immunotherapy have transformed cancer care and realized unprecedented responses in many patients. The growing arsenal of novel therapeutics - including immune checkpoint inhibition (ICI), adoptive T cell therapies (ACTs), and cancer vaccines - reflects the success of cancer immunotherapy. The therapeutic benefits of these treatment modalities are generally attributed to the enhanced quantity and quality of antitumor CD8+ T cell responses. Nevertheless, CD4+ T cells are now recognized to play key roles in both the priming and effector phases of the antitumor immune response. In addition to providing T cell help through co-stimulation and cytokine production, CD4+ T cells can also possess cytotoxicity either directly on MHC class II-expressing tumor cells or to other cells within the tumor microenvironment (TME). The presence of specific populations of CD4+ T cells, and their intrinsic plasticity, within the TME can represent an important determinant of clinical response to immune checkpoint inhibitors, vaccines, and chimeric antigen receptor (CAR) T cell therapies. Understanding how the antitumor functions of specific CD4+ T cell types are induced while limiting their protumorigenic attributes will enable more successful immunotherapies.
Collapse
Affiliation(s)
- Elena Montauti
- Division of Hematology/Oncology, Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - David Y Oh
- Division of Hematology/Oncology, Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Lawrence Fong
- Division of Hematology/Oncology, Department of Medicine, University of California San Francisco, San Francisco, CA, USA; Immunotherapy Integrated Research Center, Fred Hutchison Cancer Center, Seattle, WA, USA.
| |
Collapse
|
30
|
Zhu J, Wang Y, Chang WY, Malewska A, Napolitano F, Gahan JC, Unni N, Zhao M, Yuan R, Wu F, Yue L, Guo L, Zhao Z, Chen DZ, Hannan R, Zhang S, Xiao G, Mu P, Hanker AB, Strand D, Arteaga CL, Desai N, Wang X, Xie Y, Wang T. Mapping cellular interactions from spatially resolved transcriptomics data. Nat Methods 2024; 21:1830-1842. [PMID: 39227721 DOI: 10.1038/s41592-024-02408-1] [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: 01/24/2024] [Accepted: 08/02/2024] [Indexed: 09/05/2024]
Abstract
Cell-cell communication (CCC) is essential to how life forms and functions. However, accurate, high-throughput mapping of how expression of all genes in one cell affects expression of all genes in another cell is made possible only recently through the introduction of spatially resolved transcriptomics (SRT) technologies, especially those that achieve single-cell resolution. Nevertheless, substantial challenges remain to analyze such highly complex data properly. Here, we introduce a multiple-instance learning framework, Spacia, to detect CCCs from data generated by SRTs, by uniquely exploiting their spatial modality. We highlight Spacia's power to overcome fundamental limitations of popular analytical tools for inference of CCCs, including losing single-cell resolution, limited to ligand-receptor relationships and prior interaction databases, high false positive rates and, most importantly, the lack of consideration of the multiple-sender-to-one-receiver paradigm. We evaluated the fitness of Spacia for three commercialized single-cell resolution SRT technologies: MERSCOPE/Vizgen, CosMx/NanoString and Xenium/10x. Overall, Spacia represents a notable step in advancing quantitative theories of cellular communications.
Collapse
Affiliation(s)
- James Zhu
- Quantitative Biomedical Research Center, Peter O'Donnell Jr. School of Public Health, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Yunguan Wang
- Quantitative Biomedical Research Center, Peter O'Donnell Jr. School of Public Health, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Division of Pediatric Gastroenterology, Hepatology and Nutrition, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati, Cincinnati, OH, USA
| | - Woo Yong Chang
- Quantitative Biomedical Research Center, Peter O'Donnell Jr. School of Public Health, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Alicia Malewska
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Fabiana Napolitano
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Jeffrey C Gahan
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Nisha Unni
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Min Zhao
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Rongqing Yuan
- Quantitative Biomedical Research Center, Peter O'Donnell Jr. School of Public Health, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Fangjiang Wu
- Quantitative Biomedical Research Center, Peter O'Donnell Jr. School of Public Health, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Lauren Yue
- Quantitative Biomedical Research Center, Peter O'Donnell Jr. School of Public Health, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Lei Guo
- Quantitative Biomedical Research Center, Peter O'Donnell Jr. School of Public Health, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Zhuo Zhao
- Department of Computer Science and Engineering, University of Notre Dame, Notre Dame, IN, USA
| | - Danny Z Chen
- Department of Computer Science and Engineering, University of Notre Dame, Notre Dame, IN, USA
| | - Raquibul Hannan
- Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Siyuan Zhang
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Guanghua Xiao
- Quantitative Biomedical Research Center, Peter O'Donnell Jr. School of Public Health, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Ping Mu
- Department of Molecular Biology, UT Southwestern Medical Center, Dallas, TX, USA
- Hamon Center for Regenerative Science and Medicine, UT Southwestern Medical Center, Dallas, TX, USA
| | - Ariella B Hanker
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Douglas Strand
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Carlos L Arteaga
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Neil Desai
- Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Xinlei Wang
- Department of Mathematics, University of Texas at Arlington, Arlington, TX, USA.
- Division of Data Science, College of Science, University of Texas at Arlington, Arlington, TX, USA.
| | - Yang Xie
- Quantitative Biomedical Research Center, Peter O'Donnell Jr. School of Public Health, University of Texas Southwestern Medical Center, Dallas, TX, USA.
- Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| | - Tao Wang
- Quantitative Biomedical Research Center, Peter O'Donnell Jr. School of Public Health, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
31
|
Radhakrishnan H, Newmyer SL, Javitz HS, Bhatnagar P. Engineered CD4 T cells for in vivo delivery of therapeutic proteins. Proc Natl Acad Sci U S A 2024; 121:e2318687121. [PMID: 39312667 PMCID: PMC11459198 DOI: 10.1073/pnas.2318687121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 08/12/2024] [Indexed: 09/25/2024] Open
Abstract
The CD4 T cell, when engineered with a chimeric antigen receptor (CAR) containing specific intracellular domains, has been transformed into a zero-order drug-delivery platform. This introduces the capability of prolonged, disease-specific engineered protein biologics production, at the disease site. Experimental findings demonstrate that CD4 T cells offer a solution when modified with a CAR that includes 4-1BB but excludes CD28 intracellular domain. In this configuration, they achieve ~3X transduction efficiency of CD8 T cells, ~2X expansion rates, generating ~5X more biologic, and exhibit minimal cytolytic activity. Cumulatively, this addresses two main hurdles in the translation of cell-based drug delivery: scaling the production of engineered T cell ex vivo and generating sufficient biologics in vivo. When programmed to induce IFNβ upon engaging the target antigen, the CD4 T cells outperforms CD8 T cells, effectively suppressing cancer cell growth in vitro and in vivo. In summary, this platform enables precise targeting of disease sites with engineered protein-based therapeutics while minimizing healthy tissue exposure. Leveraging CD4 T cells' persistence could enhance disease management by reducing drug administration frequency, addressing critical challenges in cell-based therapy.
Collapse
|
32
|
Liu YC, Ansaryan S, Tan J, Broguiere N, Lorenzo-Martín LF, Homicsko K, Coukos G, Lütolf MP, Altug H. Nanoplasmonic Single-Tumoroid Microarray for Real-Time Secretion Analysis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2401539. [PMID: 38924371 PMCID: PMC11425908 DOI: 10.1002/advs.202401539] [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: 02/14/2024] [Revised: 04/30/2024] [Indexed: 06/28/2024]
Abstract
Organoid tumor models have emerged as a powerful tool in the fields of biology and medicine as such 3D structures grown from tumor cells recapitulate better tumor characteristics, making these tumoroids unique for personalized cancer research. Assessment of their functional behavior, particularly protein secretion, is of significant importance to provide comprehensive insights. Here, a label-free spectroscopic imaging platform is presented with advanced integrated optofluidic nanoplasmonic biosensor that enables real-time secretion analysis from single tumoroids. A novel two-layer microwell design isolates tumoroids, preventing signal interference, and the microarray configuration allows concurrent analysis of multiple tumoroids. The dual imaging capability combining time-lapse plasmonic spectroscopy and bright-field microscopy facilitates simultaneous observation of secretion dynamics, motility, and morphology. The integrated biosensor is demonstrated with colorectal tumoroids derived from both cell lines and patient samples to investigate their vascular endothelial growth factor A (VEGF-A) secretion, growth, and movement under various conditions, including normoxia, hypoxia, and drug treatment. This platform, by offering a label-free approach with nanophotonics to monitor tumoroids, can pave the way for new applications in fundamental biological studies, drug screening, and the development of therapies.
Collapse
Affiliation(s)
- Yen-Cheng Liu
- Bionanophotonic Systems Laboratory, Institute of Bioengineering, School of Engineering, École Polytechnique Fédérale de Lausanne, Lausanne, 1015, Switzerland
| | - Saeid Ansaryan
- Bionanophotonic Systems Laboratory, Institute of Bioengineering, School of Engineering, École Polytechnique Fédérale de Lausanne, Lausanne, 1015, Switzerland
| | - Jiayi Tan
- Bionanophotonic Systems Laboratory, Institute of Bioengineering, School of Engineering, École Polytechnique Fédérale de Lausanne, Lausanne, 1015, Switzerland
| | - Nicolas Broguiere
- Laboratory of Stem Cell Bioengineering, Institute of Bioengineering, School of Life Sciences and School of Engineering, École Polytechnique Fédérale de Lausanne, Lausanne, 1015, Switzerland
| | - Luis Francisco Lorenzo-Martín
- Laboratory of Stem Cell Bioengineering, Institute of Bioengineering, School of Life Sciences and School of Engineering, École Polytechnique Fédérale de Lausanne, Lausanne, 1015, Switzerland
| | - Krisztian Homicsko
- Department of Oncology, Centre Hospitalier Universitaire Vaudois, Rue du Bugnon 46, Lausanne, 1005, Switzerland
- Ludwig Institute for Cancer Research, Ludwig Lausanne Branch, Chem. des Boveresses 155, Epalinges, 1066, Switzerland
- Swiss Cancer Center Leman, Rue du Bugnon 25A, Lausanne, 1011, Switzerland
- Agora Translational Research Center, Rue du Bugnon 25A, Lausanne, 1011, Switzerland
| | - George Coukos
- Department of Oncology, Centre Hospitalier Universitaire Vaudois, Rue du Bugnon 46, Lausanne, 1005, Switzerland
- Ludwig Institute for Cancer Research, Ludwig Lausanne Branch, Chem. des Boveresses 155, Epalinges, 1066, Switzerland
- Swiss Cancer Center Leman, Rue du Bugnon 25A, Lausanne, 1011, Switzerland
- Agora Translational Research Center, Rue du Bugnon 25A, Lausanne, 1011, Switzerland
| | - Matthias P Lütolf
- Laboratory of Stem Cell Bioengineering, Institute of Bioengineering, School of Life Sciences and School of Engineering, École Polytechnique Fédérale de Lausanne, Lausanne, 1015, Switzerland
| | - Hatice Altug
- Bionanophotonic Systems Laboratory, Institute of Bioengineering, School of Engineering, École Polytechnique Fédérale de Lausanne, Lausanne, 1015, Switzerland
| |
Collapse
|
33
|
Lang HP, Osum KC, Friedenberg SG. A review of CD4 + T cell differentiation and diversity in dogs. Vet Immunol Immunopathol 2024; 275:110816. [PMID: 39173398 PMCID: PMC11421293 DOI: 10.1016/j.vetimm.2024.110816] [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: 06/19/2024] [Revised: 08/12/2024] [Accepted: 08/13/2024] [Indexed: 08/24/2024]
Abstract
CD4+ T cells are an integral component of the adaptive immune response, carrying out many functions to combat a diverse range of pathogenic challenges. These cells exhibit remarkable plasticity, differentiating into specialized subsets such as T helper type 1 (TH1), TH2, TH9, TH17, TH22, regulatory T cells (Tregs), and follicular T helper (TFH) cells. Each subset is capable of addressing a distinct immunological need ranging from pathogen eradication to regulation of immune homeostasis. As the immune response subsides, CD4+ T cells rest down into long-lived memory phenotypes-including central memory (TCM), effector memory (TEM), resident memory (TRM), and terminally differentiated effector memory cells (TEMRA) that are localized to facilitate a swift and potent response upon antigen re-encounter. This capacity for long-term immunological memory and rapid reactivation upon secondary exposure highlights the role CD4+ T cells play in sustaining both adaptive defense mechanisms and maintenance. Decades of mouse, human, and to a lesser extent, pig T cell research has provided the framework for understanding the role of CD4+ T cells in immune responses, but these model systems do not always mimic each other. Although our understanding of pig immunology is not as extensive as mouse or human research, we have gained valuable insight by studying this model. More akin to pigs, our understanding of CD4+ T cells in dogs is much less complete. This disparity exists in part because canine immunologists depend on paradigms from mouse and human studies to characterize CD4+ T cells in dogs, with a fraction of available lineage-defining antibody markers. Despite this, every major CD4+ T cell subset has been described to some extent in dogs. These subsets have been studied in various contexts, including in vitro stimulation, homeostatic conditions, and across a range of disease states. Canine CD4+ T cells have been categorized according to lineage-defining characteristics, trafficking patterns, and what cytokines they produce upon stimulation. This review addresses our current understanding of canine CD4+ T cells from a comparative perspective by highlighting both the similarities and differences from mouse, human, and pig CD4+ T cell biology. We also discuss knowledge gaps in our current understanding of CD4+ T cells in dogs that could provide direction for future studies in the field.
Collapse
Affiliation(s)
- Haeree P Lang
- Center for Immunology, University of Minnesota, Minneapolis, MN 55414, USA; Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St. Paul, MN 55108, USA.
| | - Kevin C Osum
- Center for Immunology, University of Minnesota, Minneapolis, MN 55414, USA.
| | - Steven G Friedenberg
- Center for Immunology, University of Minnesota, Minneapolis, MN 55414, USA; Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St. Paul, MN 55108, USA.
| |
Collapse
|
34
|
Chen H, Ao Q, Wang Y, Qian Y, Cheng Q, Zhang W. SOX11 as a potential prognostic biomarker in hepatocellular carcinoma linked to immune infiltration and ferroptosis. Chin J Cancer Res 2024; 36:378-397. [PMID: 39246708 PMCID: PMC11377886 DOI: 10.21147/j.issn.1000-9604.2024.04.03] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 07/25/2024] [Indexed: 09/10/2024] Open
Abstract
Objective SOX11 is expressed in numerous malignancies, including hepatocellular carcinomas (HCC), but its oncogenic function has not been elucidated. Here, we performed a comprehensive bioinformatics analysis of the Liver Hepatocellular Carcinoma (LIHC) dataset to investigate the function of SOX11 in tumorgenesis. Methods SOX11 expression data from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) were validated by immunohistochemistry (IHC). Co-expression, differential expression, and functional analyses utilized TCGA-LIHC, Timer 2.0, Metascape, GTEx, and LinkedOmics databases. Associations with immune infiltration, ferroptosis, and immune checkpoint genes were assessed. Genetic changes were explored via CBioPortal. Logistic regression, receiver operating characteristic curve (ROC), Kaplan-Meier analysis, and nomogram modeling evaluated associations with HCC clinicopathological features. SOX11's impact on proliferation and migration was studied in HepG2 and HuH7 cell lines. Results SOX11 was significantly elevated in HCC tumors compared to controls. SOX11-associated genes exhibited differential expression in pathways involving extracellular membrane ion channels. Significant associations were found between SOX11 levels, immune infiltration, ferroptosis, and immune checkpoint genes in HCC tissue. SOX11 levels correlated with HCC stage, histologic grade, and tumor status, and independently predicted overall and disease-specific survival. SOX11 expression effectively distinguished between tumor and normal liver tissue. Spearman correlations highlighted a significant relationship between SOX11 and ferroptosis-associated genes. Decreased SOX11 levels in HepG2 and HuH7 cells resulted in reduced proliferation and migration. Conclusions SOX11 was found to represent a promising biomarker within HCC diagnosis and prognosis together with being a possible drug-target.
Collapse
Affiliation(s)
- Hongyu Chen
- Department of Nephrology, the Second Medical Center of PLA General Hospital, National Clinical Research Center for Geriatric Diseases, Beijing 100853, China
| | - Qiangguo Ao
- Department of Nephrology, the Second Medical Center of PLA General Hospital, National Clinical Research Center for Geriatric Diseases, Beijing 100853, China
| | - Yueling Wang
- Clinical Laboratory, the First Affiliated Hospital of Henan University, Kaifeng 475000, China
| | - Yue Qian
- Cell Biology Department, Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China
| | - Qingli Cheng
- Department of Nephrology, the Second Medical Center of PLA General Hospital, National Clinical Research Center for Geriatric Diseases, Beijing 100853, China
| | - Wei Zhang
- Cell Biology Department, Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China
- Department of Pathogen Biology, Guizhou Nursing Vocational College, Guiyang 550000, China
| |
Collapse
|
35
|
Jiménez-Reinoso A, Molero-Abraham M, Cirauqui C, Blanco B, Garrido-Martin EM, Nehme-Álvarez D, Domínguez-Alonso C, Ramírez-Fernández Á, Díez-Alonso L, Nuñez-Buiza Á, González-Murillo Á, Tobes R, Pareja E, Ramírez-Orellana M, Rodriguez-Peralto JL, Ferrer I, Zugazagoitia J, Paz-Ares L, Álvarez-Vallina L. CD4 + tumor-infiltrating lymphocytes secreting T cell-engagers induce regression of autologous patient-derived non-small cell lung cancer xenografts. Oncoimmunology 2024; 13:2392897. [PMID: 39206095 PMCID: PMC11352715 DOI: 10.1080/2162402x.2024.2392897] [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: 06/23/2024] [Revised: 08/11/2024] [Accepted: 08/12/2024] [Indexed: 09/04/2024] Open
Abstract
Adoptive transfer of tumor-infiltrating lymphocytes (TIL) has shown remarkable results in melanoma, but only modest clinical benefits in other cancers, even after TIL have been genetically modified to improve their tumor homing, cytotoxic potential or overcome cell exhaustion. The required ex vivo TIL expansion process may induce changes in the T cell clonal composition, which could likely compromise the tumor reactivity of TIL preparations and ultimately the success of TIL therapy. A promising approach based on the production of bispecific T cell-engagers (TCE) by engineered T cells (STAb-T therapy) improves the efficacy of current T cell redirection strategies against tumor-associated antigens in hematological tumors. We studied the TCRβ repertoire in non-small cell lung cancer (NSCLC) tumors and in ex vivo expanded TIL from two unrelated patients. We generated TIL secreting anti-epidermal growth factor receptor (EGFR) × anti-CD3 TCE (TILSTAb) and tested their antitumor efficacy in vitro and in vivo using a NSCLC patient-derived xenograft (PDX) model in which tumor fragments and TIL from the same patient were transplanted into hIL-2 NOG mice. We confirmed that the standard TIL expansion protocol promotes the loss of tumor-dominant T cell clones and the overgrowth of virus-reactive TCR clonotypes that were marginally detectable in primary tumors. We demonstrated the antitumor activity of TILSTAb both in vitro and in vivo when administered intratumorally and systemically in an autologous immune-humanized PDX EGFR+ NSCLC mouse model, where tumor regression was mediated by TCE-redirected CD4+ TIL bearing non-tumor dominant clonotypes.
Collapse
Affiliation(s)
- Anaïs Jiménez-Reinoso
- Cancer Immunotherapy Unit (UNICA), Department of Immunology, Hospital Universitario 12 de Octubre, Madrid, Spain
- Immuno-Oncology and Immunotherapy Group, Instituto de Investigación Sanitaria 12 de Octubre (imas12), Madrid, Spain
- H12O-CNIO Cancer Immunotherapy Clinical Research Unit, Centro Nacional de Investigaciones Oncológicas (CNIO), Madrid, Spain
| | - Magdalena Molero-Abraham
- Tumor Microenvironment and Immunotherapy Research Group, Madrid, Spain
- H12O-CNIO Lung Cancer Clinical Research Unit, Instituto de Investigación Sanitaria 12 de Octubre (imas12), CNIO, Madrid, Spain
| | - Cristina Cirauqui
- H12O-CNIO Lung Cancer Clinical Research Unit, Instituto de Investigación Sanitaria 12 de Octubre (imas12), CNIO, Madrid, Spain
| | - Belén Blanco
- Cancer Immunotherapy Unit (UNICA), Department of Immunology, Hospital Universitario 12 de Octubre, Madrid, Spain
- Immuno-Oncology and Immunotherapy Group, Instituto de Investigación Sanitaria 12 de Octubre (imas12), Madrid, Spain
- H12O-CNIO Cancer Immunotherapy Clinical Research Unit, Centro Nacional de Investigaciones Oncológicas (CNIO), Madrid, Spain
| | - Eva M. Garrido-Martin
- H12O-CNIO Lung Cancer Clinical Research Unit, Instituto de Investigación Sanitaria 12 de Octubre (imas12), CNIO, Madrid, Spain
- Spanish Center for Biomedical Research Network in Oncology (CIBERONC), Madrid, Spain
| | - Daniel Nehme-Álvarez
- Cancer Immunotherapy Unit (UNICA), Department of Immunology, Hospital Universitario 12 de Octubre, Madrid, Spain
- Immuno-Oncology and Immunotherapy Group, Instituto de Investigación Sanitaria 12 de Octubre (imas12), Madrid, Spain
| | - Carmen Domínguez-Alonso
- Cancer Immunotherapy Unit (UNICA), Department of Immunology, Hospital Universitario 12 de Octubre, Madrid, Spain
- Immuno-Oncology and Immunotherapy Group, Instituto de Investigación Sanitaria 12 de Octubre (imas12), Madrid, Spain
- H12O-CNIO Cancer Immunotherapy Clinical Research Unit, Centro Nacional de Investigaciones Oncológicas (CNIO), Madrid, Spain
| | - Ángel Ramírez-Fernández
- Cancer Immunotherapy Unit (UNICA), Department of Immunology, Hospital Universitario 12 de Octubre, Madrid, Spain
- Immuno-Oncology and Immunotherapy Group, Instituto de Investigación Sanitaria 12 de Octubre (imas12), Madrid, Spain
- H12O-CNIO Cancer Immunotherapy Clinical Research Unit, Centro Nacional de Investigaciones Oncológicas (CNIO), Madrid, Spain
| | - Laura Díez-Alonso
- Cancer Immunotherapy Unit (UNICA), Department of Immunology, Hospital Universitario 12 de Octubre, Madrid, Spain
- Immuno-Oncology and Immunotherapy Group, Instituto de Investigación Sanitaria 12 de Octubre (imas12), Madrid, Spain
- H12O-CNIO Cancer Immunotherapy Clinical Research Unit, Centro Nacional de Investigaciones Oncológicas (CNIO), Madrid, Spain
| | - Ángel Nuñez-Buiza
- H12O-CNIO Lung Cancer Clinical Research Unit, Instituto de Investigación Sanitaria 12 de Octubre (imas12), CNIO, Madrid, Spain
| | - África González-Murillo
- Advanced Therapies Unit, Oncology, Hospital Infantil Universitario Niño Jesús, Madrid, Spain
- Fundación de Investigación Biomédica, Hospital Infantil Universitario Niño Jesús, Madrid, Spain
- Instituto de Investigación Sanitaria La Princesa, Madrid, Spain
| | - Raquel Tobes
- Alamo blanco Research Association, Granada, Spain
| | | | - Manuel Ramírez-Orellana
- Advanced Therapies Unit, Oncology, Hospital Infantil Universitario Niño Jesús, Madrid, Spain
- Fundación de Investigación Biomédica, Hospital Infantil Universitario Niño Jesús, Madrid, Spain
- Instituto de Investigación Sanitaria La Princesa, Madrid, Spain
| | - José Luis Rodriguez-Peralto
- Spanish Center for Biomedical Research Network in Oncology (CIBERONC), Madrid, Spain
- Department of Pathology, Hospital Universitario 12 de Octubre, Madrid, Spain
- Department of Pathology, Complutense University, Madrid, Spain
- Cutaneous Oncology Group, Madrid, Spain
| | - Irene Ferrer
- H12O-CNIO Lung Cancer Clinical Research Unit, Instituto de Investigación Sanitaria 12 de Octubre (imas12), CNIO, Madrid, Spain
- Spanish Center for Biomedical Research Network in Oncology (CIBERONC), Madrid, Spain
| | - Jon Zugazagoitia
- Tumor Microenvironment and Immunotherapy Research Group, Madrid, Spain
- H12O-CNIO Lung Cancer Clinical Research Unit, Instituto de Investigación Sanitaria 12 de Octubre (imas12), CNIO, Madrid, Spain
- Spanish Center for Biomedical Research Network in Oncology (CIBERONC), Madrid, Spain
- Department of Medical Oncology, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Luis Paz-Ares
- H12O-CNIO Lung Cancer Clinical Research Unit, Instituto de Investigación Sanitaria 12 de Octubre (imas12), CNIO, Madrid, Spain
- Spanish Center for Biomedical Research Network in Oncology (CIBERONC), Madrid, Spain
- Department of Medical Oncology, Hospital Universitario 12 de Octubre, Madrid, Spain
- Department of Medicine, Complutense University, Madrid, Spain
| | - Luis Álvarez-Vallina
- Cancer Immunotherapy Unit (UNICA), Department of Immunology, Hospital Universitario 12 de Octubre, Madrid, Spain
- Immuno-Oncology and Immunotherapy Group, Instituto de Investigación Sanitaria 12 de Octubre (imas12), Madrid, Spain
- H12O-CNIO Cancer Immunotherapy Clinical Research Unit, Centro Nacional de Investigaciones Oncológicas (CNIO), Madrid, Spain
| |
Collapse
|
36
|
Yang C, Trivedi V, Dyson K, Gu T, Candelario KM, Yegorov O, Mitchell DA. Identification of tumor rejection antigens and the immunologic landscape of medulloblastoma. Genome Med 2024; 16:102. [PMID: 39160595 PMCID: PMC11331754 DOI: 10.1186/s13073-024-01363-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 07/12/2024] [Indexed: 08/21/2024] Open
Abstract
BACKGROUND The current standard of care treatments for medulloblastoma are insufficient as these do not take tumor heterogeneity into account. Newer, safer, patient-specific treatment approaches are required to treat high-risk medulloblastoma patients who are not cured by the standard therapies. Immunotherapy is a promising treatment modality that could be key to improving survival and avoiding morbidity. For an effective immune response, appropriate tumor antigens must be targeted. While medulloblastoma patients with subgroup-specific genetic substitutions have been previously reported, the immunogenicity of these genetic alterations remains unknown. The aim of this study is to identify potential tumor rejection antigens for the development of antigen-directed cellular therapies for medulloblastoma. METHODS We developed a cancer immunogenomics pipeline and performed a comprehensive analysis of medulloblastoma subgroup-specific transcription profiles (n = 170, 18 WNT, 46 SHH, 41 Group 3, and 65 Group 4 patient tumors) available through International Cancer Genome Consortium (ICGC) and European Genome-Phenome Archive (EGA). We performed in silico antigen prediction across a broad array of antigen classes including neoantigens, tumor-associated antigens (TAAs), and fusion proteins. Furthermore, we evaluated the antigen processing and presentation pathway in tumor cells and the immune infiltrating cell landscape using the latest computational deconvolution methods. RESULTS Medulloblastoma patients were found to express multiple private and shared immunogenic antigens. The proportion of predicted TAAs was higher than neoantigens and gene fusions for all molecular subgroups, except for sonic hedgehog (SHH), which had a higher neoantigen burden. Importantly, cancer-testis antigens, as well as previously unappreciated neurodevelopmental antigens, were found to be expressed by most patients across all medulloblastoma subgroups. Despite being immunologically cold, medulloblastoma subgroups were found to have distinct immune cell gene signatures. CONCLUSIONS Using a custom antigen prediction pipeline, we identified potential tumor rejection antigens with important implications for the development of immunotherapy for medulloblastoma.
Collapse
Affiliation(s)
- Changlin Yang
- UF Brain Tumor Immunotherapy Program, Preston A. Wells Center for Brain Tumor Therapy, Lillian S. Wells Department of Neurosurgery, University of Florida, 1333 Center Drive, BSB B1-118, Gainesville, FL, 32610, USA
| | - Vrunda Trivedi
- UF Brain Tumor Immunotherapy Program, Preston A. Wells Center for Brain Tumor Therapy, Lillian S. Wells Department of Neurosurgery, University of Florida, 1333 Center Drive, BSB B1-118, Gainesville, FL, 32610, USA
| | - Kyle Dyson
- UF Brain Tumor Immunotherapy Program, Preston A. Wells Center for Brain Tumor Therapy, Lillian S. Wells Department of Neurosurgery, University of Florida, 1333 Center Drive, BSB B1-118, Gainesville, FL, 32610, USA
| | - Tongjun Gu
- Department of Biostatistics, University of Florida, Gainesville, FL, USA
| | - Kate M Candelario
- UF Brain Tumor Immunotherapy Program, Preston A. Wells Center for Brain Tumor Therapy, Lillian S. Wells Department of Neurosurgery, University of Florida, 1333 Center Drive, BSB B1-118, Gainesville, FL, 32610, USA
| | - Oleg Yegorov
- UF Brain Tumor Immunotherapy Program, Preston A. Wells Center for Brain Tumor Therapy, Lillian S. Wells Department of Neurosurgery, University of Florida, 1333 Center Drive, BSB B1-118, Gainesville, FL, 32610, USA
| | - Duane A Mitchell
- UF Brain Tumor Immunotherapy Program, Preston A. Wells Center for Brain Tumor Therapy, Lillian S. Wells Department of Neurosurgery, University of Florida, 1333 Center Drive, BSB B1-118, Gainesville, FL, 32610, USA.
| |
Collapse
|
37
|
Najar TA, Hao Y, Hao Y, Romero-Meza G, Dolynuk A, Littman DR. Microbiota-induced plastic T cells enhance immune control of antigen-sharing tumors. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.12.607605. [PMID: 39185202 PMCID: PMC11343098 DOI: 10.1101/2024.08.12.607605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/27/2024]
Abstract
Therapies that harness the immune system to target and eliminate tumor cells have revolutionized cancer care. Immune checkpoint blockade (ICB), which boosts the anti-tumor immune response by inhibiting negative regulators of T cell activation1-3, is remarkably successful in a subset of cancer patients, yet a significant proportion do not respond to treatment, emphasizing the need to understand factors influencing the therapeutic efficacy of ICB4-9. The gut microbiota, consisting of trillions of microorganisms residing in the gastrointestinal tract, has emerged as a critical determinant of immune function and response to cancer immunotherapy, with multiple studies demonstrating association of microbiota composition with clinical response10-16. However, a mechanistic understanding of how gut commensal bacteria influence the efficacy of ICB remains elusive. Here we utilized a gut commensal microorganism, segmented filamentous bacteria (SFB), which induces an antigen-specific Th17 cell effector program17, to investigate how colonization with it affects the efficacy of ICB in restraining distal growth of tumors sharing antigen with SFB. We find that anti-PD-1 treatment effectively inhibits the growth of implanted SFB antigen-expressing melanoma only if mice are colonized with SFB. Through T cell receptor clonal lineage tracing, fate mapping, and peptide-MHC tetramer staining, we identify tumor-associated SFB-specific Th1-like cells derived from the homeostatic Th17 cells induced by SFB colonization in the small intestine lamina propria. These gut-educated ex-Th17 cells produce high levels of the pro-inflammatory cytokines IFN-γ and TNF-α, and promote expansion and effector functions of CD8+ tumor-infiltrating cytotoxic lymphocytes, thereby controlling tumor growth. A better understanding of how distinct intestinal commensal microbes can promote T cell plasticity-dependent responses against antigen-sharing tumors may allow for the design of novel cancer immunotherapeutic strategies.
Collapse
Affiliation(s)
- Tariq A. Najar
- Department of Cell Biology, New York University School of Medicine, New York, NY 10016, USA
| | - Yuan Hao
- Applied Bioinformatics Laboratories, Division of Advanced Research Technologies, New York, New York University School of Medicine, New York, NY 10016, USA
- Perlmutter Cancer Center, New York University Langone Health, New York, NY 10016, USA
| | - Yuhan Hao
- Center for Genomics and Systems Biology, New York University, New York, NY 10003, USA
- New York Genome Center, New York, NY 10013, USA
| | - Gabriela Romero-Meza
- Department of Cell Biology, New York University School of Medicine, New York, NY 10016, USA
- Howard Hughes Medical Institute, New York, NY 10016, USA
| | - Alexandra Dolynuk
- Department of Cell Biology, New York University School of Medicine, New York, NY 10016, USA
- Howard Hughes Medical Institute, New York, NY 10016, USA
| | - Dan R. Littman
- Department of Cell Biology, New York University School of Medicine, New York, NY 10016, USA
- Perlmutter Cancer Center, New York University Langone Health, New York, NY 10016, USA
- Howard Hughes Medical Institute, New York, NY 10016, USA
| |
Collapse
|
38
|
Lee CR, Kim MJ, Park SH, Kim S, Kim SY, Koh SJ, Lee S, Choi M, Chae JH, Park SG, Moon J. Recurrent fever of unknown origin and unexplained bacteremia in a patient with a novel 4.5 Mb microdeletion in Xp11.23-p11.22. Sci Rep 2024; 14:17801. [PMID: 39090138 PMCID: PMC11294525 DOI: 10.1038/s41598-024-65341-5] [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: 01/24/2024] [Accepted: 06/19/2024] [Indexed: 08/04/2024] Open
Abstract
Fever of unknown origin (FUO) remains a formidable diagnostic challenge in the field of medicine. Numerous studies suggest an association between FUO and genetic factors, including chromosomal abnormalities. Here, we report a female patient with a 4.5 Mb Xp microdeletion, who presented with recurrent FUO, bacteremia, colitis, and hematochezia. To elucidate the underlying pathogenic mechanism, we employed a comprehensive approach involving single cell RNA sequencing, T cell receptor sequencing, and flow cytometry to evaluate CD4 T cells. Analysis of peripheral blood mononuclear cells revealed augmented Th1, Th2, and Th17 cell populations, and elevated levels of proinflammatory cytokines in serum. Notably, the patient exhibited impaired Treg cell function, possibly related to deletion of genes encoding FOPX3 and WAS. Single cell analysis revealed specific expansion of cytotoxic CD4 T lymphocytes, characterized by upregulation of various signature genes associated with cytotoxicity. Moreover, interferon-stimulated genes were upregulated in the CD4 T effector memory cluster. Further genetic analysis confirmed maternal inheritance of the Xp microdeletion. The patient and her mother exhibited X chromosome-skewed inactivation, a potential protective mechanism against extensive X chromosome deletions; however, the mother exhibited complete skewing and the patient exhibited incomplete skewing (85:15), which may have contributed to emergence of immunological symptoms. In summary, this case report describes an exceptional instance of FUO stemming from an incompletely inactivated X chromosome microdeletion, thereby increasing our understanding of the genetics underpinning FUO.
Collapse
Affiliation(s)
- Cho-Rong Lee
- College of Pharmacy, Institute of Pharmaceutical Sciences, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Republic of Korea
| | - Man Jin Kim
- Department of Genomic Medicine, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
- Department of Laboratory Medicine, Seoul National University Hospital, Seoul, 03080, Republic of Korea
| | - Sang-Heon Park
- College of Pharmacy, Institute of Pharmaceutical Sciences, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Republic of Korea
| | - Sheehyun Kim
- Department of Genomic Medicine, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
| | - Soo Yeon Kim
- Department of Genomic Medicine, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
- Department of Pediatrics, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
| | - Seong-Joon Koh
- Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
| | - Seungbok Lee
- Department of Genomic Medicine, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
- Department of Pediatrics, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
| | - Murim Choi
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
| | - Jong Hee Chae
- Department of Genomic Medicine, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
- Department of Pediatrics, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
| | - Sung-Gyoo Park
- College of Pharmacy, Institute of Pharmaceutical Sciences, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Republic of Korea.
| | - Jangsup Moon
- Department of Genomic Medicine, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea.
- Department of Neurology, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea.
| |
Collapse
|
39
|
Zhu M, Wu Y, Zhu T, Chen J, Chen Z, Ding H, Tan S, He J, Zeng Q, Huang X. Multifunctional Bispecific Nanovesicles Targeting SLAMF7 Trigger Potent Antitumor Immunity. Cancer Immunol Res 2024; 12:1007-1021. [PMID: 38819238 DOI: 10.1158/2326-6066.cir-23-1102] [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: 12/27/2023] [Revised: 04/03/2024] [Accepted: 05/29/2024] [Indexed: 06/01/2024]
Abstract
The effectiveness of immune checkpoint inhibitor (ICI) therapy is hindered by the ineffective infiltration and functioning of cytotoxic T cells and the immunosuppressive tumor microenvironment (TME). Signaling lymphocytic activation molecule family member 7 (SLAMF7) is a pivotal co-stimulatory receptor thought to simultaneously trigger NK-cell, T-cell, and macrophage antitumor cytotoxicity. However, the potential of this collaborative immune stimulation in antitumor immunity for solid tumors is underexplored due to the exclusive expression of SLAMF7 by hematopoietic cells. Here, we report the development and characterization of multifunctional bispecific nanovesicles (NVs) targeting SLAMF7 and glypican-3-a hepatocellular carcinoma (HCC)-specific tumor antigen. We found that by effectively "decorating" the surfaces of solid tumors with SLAMF7, these NVs directly induced potent and specific antitumor immunity and remodeled the immunosuppressive TME, sensitizing the tumors to programmed cell death protein 1 (PD1) blockade. Our findings highlight the potential of SLAMF7-targeted multifunctional bispecific NVs as an anticancer strategy with implications for designing next-generation targeted cancer therapies.
Collapse
Affiliation(s)
- Manman Zhu
- Center for Infection and Immunity and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
| | - Yongjian Wu
- Center for Infection and Immunity and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
| | - Tianchuan Zhu
- Center for Infection and Immunity and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
| | - Jian Chen
- Center for Infection and Immunity and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
| | - Zhenxing Chen
- Center for Infection and Immunity and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
- Kingcell Regenerative Medicine (Guangdong) Co., Zhuhai, China
| | - Hanxi Ding
- Center for Infection and Immunity and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
- Cancer Center, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, China
| | - Siyi Tan
- Center for Infection and Immunity and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
| | - Jianzhong He
- Department of Pathology, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
| | - Qi Zeng
- Cancer Center, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, China
| | - Xi Huang
- Center for Infection and Immunity and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
| |
Collapse
|
40
|
Zhivaki D, Kennedy SN, Park J, Boriello F, Devant P, Cao A, Bahleda KM, Murphy S, McCabe C, Evavold CL, Chapman KL, Zanoni I, Ashenberg O, Xavier RJ, Kagan JC. Correction of age-associated defects in dendritic cells enables CD4 + T cells to eradicate tumors. Cell 2024; 187:3888-3903.e18. [PMID: 38870946 PMCID: PMC11283364 DOI: 10.1016/j.cell.2024.05.026] [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: 11/02/2022] [Revised: 04/02/2024] [Accepted: 05/13/2024] [Indexed: 06/15/2024]
Abstract
Defective host defenses later in life are associated with changes in immune cell activities, suggesting that age-specific considerations are needed in immunotherapy approaches. In this study, we found that PD-1 and CTLA4-based cancer immunotherapies are unable to eradicate tumors in elderly mice. This defect in anti-tumor activity correlated with two known age-associated immune defects: diminished abundance of systemic naive CD8+ T cells and weak migratory activities of dendritic cells (DCs). We identified a vaccine adjuvant, referred to as a DC hyperactivator, which corrects DC migratory defects in the elderly. Vaccines containing tumor antigens and DC hyperactivators induced T helper type 1 (TH1) CD4+ T cells with cytolytic activity that drive anti-tumor immunity in elderly mice. When administered early in life, DC hyperactivators were the only adjuvant identified that elicited anti-tumor CD4+ T cells that persisted into old age. These results raise the possibility of correcting age-associated immune defects through DC manipulation.
Collapse
Affiliation(s)
- Dania Zhivaki
- Harvard Medical School and Division of Gastroenterology, Boston Children's Hospital, Boston, MA, USA
| | - Stephanie N Kennedy
- Harvard Medical School and Division of Gastroenterology, Boston Children's Hospital, Boston, MA, USA
| | - Josh Park
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Francesco Boriello
- Harvard Medical School and Division of Immunology, Boston Children's Hospital, Boston, MA, USA
| | - Pascal Devant
- Harvard Medical School and Division of Gastroenterology, Boston Children's Hospital, Boston, MA, USA
| | - Anh Cao
- Harvard Medical School and Division of Gastroenterology, Boston Children's Hospital, Boston, MA, USA
| | - Kristin M Bahleda
- Harvard Medical School and Division of Gastroenterology, Boston Children's Hospital, Boston, MA, USA
| | - Shane Murphy
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Cristin McCabe
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Charles L Evavold
- Harvard Medical School and Division of Gastroenterology, Boston Children's Hospital, Boston, MA, USA
| | - Kate L Chapman
- Harvard Medical School and Division of Gastroenterology, Boston Children's Hospital, Boston, MA, USA
| | - Ivan Zanoni
- Harvard Medical School and Division of Immunology, Boston Children's Hospital, Boston, MA, USA
| | - Orr Ashenberg
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Ramnik J Xavier
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Jonathan C Kagan
- Harvard Medical School and Division of Gastroenterology, Boston Children's Hospital, Boston, MA, USA.
| |
Collapse
|
41
|
Yang X, Wu J, Fan L, Chen B, Zhang S, Zheng W. Single-Cell Analysis Identifies Distinct Populations of Cytotoxic CD4 + T Cells Linked to the Therapeutic Efficacy of Immune Checkpoint Inhibitors in Metastatic Renal Cell Carcinoma. J Inflamm Res 2024; 17:4505-4523. [PMID: 39006494 PMCID: PMC11246657 DOI: 10.2147/jir.s457570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Accepted: 06/18/2024] [Indexed: 07/16/2024] Open
Abstract
Background The involvement of cytotoxic CD4+ T cells (CD4+ CTLs) and their potential role in dictating the response to immune checkpoint inhibitors (ICIs) in patients with metastatic renal cell carcinoma (mRCC) remains an unexplored area of research. Methods Utilizing single-cell RNA sequencing, we analyzed the immunophenotype and expression patterns of CD4+ T lymphocyte subtypes in mRCC patients, followed by preliminary validation via multi-immunofluorescent staining. In addition, we obtained a comprehensive immunotherapy dataset encompassing single-cell RNA sequencing datasets and bulk RNA-seq cohorts from the European Genome-Phenome Archive and ArrayExpress database. Utilizing the CIBERSORTx deconvolution algorithms, we derived a signature score for CD4+ CTLs from the bulk-RNA-seq datasets of the CheckMate 009/025 clinical trials. Results Single-cell analysis of CD4+ T lymphocytes in mRCC reveals several cancer-specific states, including diverse phenotypes of regulatory T cells. Remarkably, we observe that CD4+ CTLs cells constitute a substantial proportion of all CD4+ T lymphocyte sub-clusters in mRCC patients, highlighting their potential significance in the disease. Furthermore, within mRCC patients, we identify two distinct cytotoxic states of CD4+ T cells: CD4+GZMK+ T cells, which exhibit a weaker cytotoxic potential, and CD4+GZMB+ T cells, which demonstrate robust cytotoxic activity. Both regulatory T cells and CD4+ CTLs originate from proliferating CD4+ T cells within mRCC tissues. Intriguingly, our trajectory analysis indicates that the weakly cytotoxic CD4+GZMK+ T cells differentiate from their more cytotoxic CD4+GZMB+ counterparts. In comparing patients with lower CD4+ CTLs levels to those with higher CD4+ CTLs abundance in the CheckMate 009 and 25 immunotherapy cohorts, the latter group exhibited significantly improved OS and PFS probability. Conclusion Our study underscores the pivotal role that intratumoral CD4+ CTLs may play in bolstering anti-tumor immunity, suggesting their potential as a promising biomarker for predicting response to ICIs in patients with mRCC.
Collapse
Affiliation(s)
- Xu Yang
- Department of Urology, Fujian Medical University Union Hospital, Fuzhou, People’s Republic of China
| | - Jianwei Wu
- Department of Urology, Fujian Medical University Union Hospital, Fuzhou, People’s Republic of China
| | - Longlong Fan
- Department of Urology, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, People’s Republic of China
| | - Binghua Chen
- Department of Urology, Pingtan Branch of Fujian Medical University Union Hospital, Fuzhou, People’s Republic of China
| | - Shiqiang Zhang
- Department of Urology, Kidney and Urology Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, People’s Republic of China
| | - Wenzhong Zheng
- Department of Urology, Fujian Medical University Union Hospital, Fuzhou, People’s Republic of China
| |
Collapse
|
42
|
Oguchi A, Suzuki A, Komatsu S, Yoshitomi H, Bhagat S, Son R, Bonnal RJP, Kojima S, Koido M, Takeuchi K, Myouzen K, Inoue G, Hirai T, Sano H, Takegami Y, Kanemaru A, Yamaguchi I, Ishikawa Y, Tanaka N, Hirabayashi S, Konishi R, Sekito S, Inoue T, Kere J, Takeda S, Takaori-Kondo A, Endo I, Kawaoka S, Kawaji H, Ishigaki K, Ueno H, Hayashizaki Y, Pagani M, Carninci P, Yanagita M, Parrish N, Terao C, Yamamoto K, Murakawa Y. An atlas of transcribed enhancers across helper T cell diversity for decoding human diseases. Science 2024; 385:eadd8394. [PMID: 38963856 DOI: 10.1126/science.add8394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2022] [Accepted: 05/01/2024] [Indexed: 07/06/2024]
Abstract
Transcribed enhancer maps can reveal nuclear interactions underpinning each cell type and connect specific cell types to diseases. Using a 5' single-cell RNA sequencing approach, we defined transcription start sites of enhancer RNAs and other classes of coding and noncoding RNAs in human CD4+ T cells, revealing cellular heterogeneity and differentiation trajectories. Integration of these datasets with single-cell chromatin profiles showed that active enhancers with bidirectional RNA transcription are highly cell type-specific and that disease heritability is strongly enriched in these enhancers. The resulting cell type-resolved multimodal atlas of bidirectionally transcribed enhancers, which we linked with promoters using fine-scale chromatin contact maps, enabled us to systematically interpret genetic variants associated with a range of immune-mediated diseases.
Collapse
Affiliation(s)
- Akiko Oguchi
- RIKEN-IFOM Joint Laboratory for Cancer Genomics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
- Institute for the Advanced Study of Human Biology, Kyoto University, Kyoto, Japan
- Department of Nephrology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Akari Suzuki
- Laboratory for Autoimmune Diseases, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Shuichiro Komatsu
- RIKEN-IFOM Joint Laboratory for Cancer Genomics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
- IFOM ETS - the AIRC Institute of Molecular Oncology, Milan, Italy
| | - Hiroyuki Yoshitomi
- Institute for the Advanced Study of Human Biology, Kyoto University, Kyoto, Japan
- Department of Immunology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Shruti Bhagat
- Institute for the Advanced Study of Human Biology, Kyoto University, Kyoto, Japan
| | - Raku Son
- RIKEN-IFOM Joint Laboratory for Cancer Genomics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
- Institute for the Advanced Study of Human Biology, Kyoto University, Kyoto, Japan
- Department of Nephrology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | | | - Shohei Kojima
- Genome Immunobiology RIKEN Hakubi Research Team, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Masaru Koido
- Division of Molecular Pathology, Department of Cancer Biology, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- Laboratory of Complex Trait Genomics, Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Tokyo, Japan
- Laboratory for Statistical and Translational Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Kazuhiro Takeuchi
- RIKEN-IFOM Joint Laboratory for Cancer Genomics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
- Institute for the Advanced Study of Human Biology, Kyoto University, Kyoto, Japan
- Department of Medical Systems Genomics, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Keiko Myouzen
- Laboratory for Autoimmune Diseases, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Gyo Inoue
- Laboratory for Autoimmune Diseases, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Tomoya Hirai
- RIKEN-IFOM Joint Laboratory for Cancer Genomics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
- Department of Gastroenterological Surgery, Yokohama City University Graduate School of Medicine, Yokohama City University, Yokohama, Japan
| | - Hiromi Sano
- RIKEN-IFOM Joint Laboratory for Cancer Genomics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | | | | | | | - Yuki Ishikawa
- Laboratory for Statistical and Translational Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Nao Tanaka
- Laboratory for Statistical and Translational Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Shigeki Hirabayashi
- RIKEN-IFOM Joint Laboratory for Cancer Genomics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
- Department of Hematology and Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
- Division of Precision Medicine, Kyushu University Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Riyo Konishi
- Inter-Organ Communication Research Team, Institute for Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Sho Sekito
- Institute for the Advanced Study of Human Biology, Kyoto University, Kyoto, Japan
- Department of Nephro-Urologic Surgery and Andrology, Mie University Graduate School of Medicine, Mie University, Tsu, Japan
| | - Takahiro Inoue
- Department of Nephro-Urologic Surgery and Andrology, Mie University Graduate School of Medicine, Mie University, Tsu, Japan
| | - Juha Kere
- Department of Biosciences and Nutrition, Karolinska Institutet, Stockholm, Sweden
- Stem Cells and Metabolism Research Program, University of Helsinki, Helsinki, Finland
- Folkhalsan Research Center, Helsinki, Finland
| | - Shunichi Takeda
- Department of Radiation Genetics, Graduate School of Medicine, Kyoto University, Kyoto, Japan
- Shenzhen University School of Medicine, Shenzhen, Guangdong, China
| | - Akifumi Takaori-Kondo
- Department of Hematology and Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Itaru Endo
- Department of Gastroenterological Surgery, Yokohama City University Graduate School of Medicine, Yokohama City University, Yokohama, Japan
| | - Shinpei Kawaoka
- Inter-Organ Communication Research Team, Institute for Life and Medical Sciences, Kyoto University, Kyoto, Japan
- Department of Integrative Bioanalytics, Institute of Development, Aging and Cancer, Tohoku University, Sendai, Japan
| | - Hideya Kawaji
- Research Center for Genome & Medical Sciences, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
- Preventive Medicine and Applied Genomics Unit, RIKEN Center for Integrative Medical Science, Yokohama, Japan
- RIKEN Preventive Medicine and Diagnosis Innovation Program, Wako, Japan
| | - Kazuyoshi Ishigaki
- Laboratory for Human Immunogenetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Hideki Ueno
- Institute for the Advanced Study of Human Biology, Kyoto University, Kyoto, Japan
- Department of Immunology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Yoshihide Hayashizaki
- K.K. DNAFORM, Yokohama, Japan
- RIKEN Preventive Medicine and Diagnosis Innovation Program, Wako, Japan
| | - Massimiliano Pagani
- IFOM ETS - the AIRC Institute of Molecular Oncology, Milan, Italy
- Department of Medical Biotechnology and Translational Medicine, Università degli Studi, Milan, Italy
| | - Piero Carninci
- Laboratory for Transcriptome Technology, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
- Human Technopole, Milan, Italy
| | - Motoko Yanagita
- Institute for the Advanced Study of Human Biology, Kyoto University, Kyoto, Japan
- Department of Nephrology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Nicholas Parrish
- Genome Immunobiology RIKEN Hakubi Research Team, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Chikashi Terao
- Laboratory for Statistical and Translational Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
- Clinical Research Center, Shizuoka General Hospital, Shizuoka, Japan
- Department of Applied Genetics, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan
| | - Kazuhiko Yamamoto
- Laboratory for Autoimmune Diseases, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Yasuhiro Murakawa
- RIKEN-IFOM Joint Laboratory for Cancer Genomics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
- Institute for the Advanced Study of Human Biology, Kyoto University, Kyoto, Japan
- IFOM ETS - the AIRC Institute of Molecular Oncology, Milan, Italy
- Department of Medical Systems Genomics, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| |
Collapse
|
43
|
Zemek RM, Anagnostou V, Pires da Silva I, Long GV, Lesterhuis WJ. Exploiting temporal aspects of cancer immunotherapy. Nat Rev Cancer 2024; 24:480-497. [PMID: 38886574 DOI: 10.1038/s41568-024-00699-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/25/2024] [Indexed: 06/20/2024]
Abstract
Many mechanisms underlying an effective immunotherapy-induced antitumour response are transient and critically time dependent. This is equally true for several immunological events in the tumour microenvironment induced by other cancer treatments. Immune checkpoint therapy (ICT) has proven to be very effective in the treatment of some cancers, but unfortunately, with many cancer types, most patients do not experience a benefit. To improve outcomes, a multitude of clinical trials are testing combinations of ICT with various other treatment modalities. Ideally, those combination treatments should take time-dependent immunological events into account. Recent studies have started to map the dynamic cellular and molecular changes that occur during treatment with ICT, in the tumour and systemically. Here, we overlay the dynamic ICT response with the therapeutic response following surgery, radiotherapy, chemotherapy and targeted therapies. We propose that by combining treatments in a time-conscious manner, we may optimally exploit the interactions between the individual therapies.
Collapse
Affiliation(s)
- Rachael M Zemek
- Telethon Kids Institute, University of Western Australia, Perth, Western Australia, Australia
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Valsamo Anagnostou
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Inês Pires da Silva
- Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia
- Faculty of Medicine & Health, The University of Sydney, Sydney, New South Wales, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia
- Crown Princess Mary Cancer Centre Westmead, Blacktown Hospital, Sydney, New South Wales, Australia
| | - Georgina V Long
- Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia
- Faculty of Medicine & Health, The University of Sydney, Sydney, New South Wales, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia
- Royal North Shore and Mater Hospitals, Sydney, New South Wales, Australia
| | - Willem Joost Lesterhuis
- Telethon Kids Institute, University of Western Australia, Perth, Western Australia, Australia.
| |
Collapse
|
44
|
Kirkpatrick C, Lu YCW. Deciphering CD4 + T cell-mediated responses against cancer. Mol Carcinog 2024; 63:1209-1220. [PMID: 38725218 PMCID: PMC11166516 DOI: 10.1002/mc.23730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 04/05/2024] [Indexed: 05/15/2024]
Abstract
It's been long thought that CD8+ cytotoxic T cells play a major role in T cell-mediated antitumor responses, whereas CD4+ T cells merely provide some assistance to CD8+ T cells as the "helpers." In recent years, numerous studies support the notion that CD4+ T cells play an indispensable role in antitumor responses. Here, we summarize and discuss the current knowledge regarding the roles of CD4+ T cells in antitumor responses and immunotherapy, with a focus on the molecular and cellular mechanisms behind these observations. These new insights on CD4+ T cells may pave the way to further optimize cancer immunotherapy.
Collapse
Affiliation(s)
- Catherine Kirkpatrick
- Department of Pathology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
- Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Yong-Chen William Lu
- Department of Pathology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
- Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| |
Collapse
|
45
|
Wittling MC, Knochelmann HM, Wyatt MM, Rangel Rivera GO, Cole AC, Lesinski GB, Paulos CM. Distinct host preconditioning regimens differentially impact the antitumor potency of adoptively transferred Th17 cells. J Immunother Cancer 2024; 12:e008715. [PMID: 38945552 PMCID: PMC11216073 DOI: 10.1136/jitc-2023-008715] [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] [Accepted: 06/02/2024] [Indexed: 07/02/2024] Open
Abstract
BACKGROUND How distinct methods of host preconditioning impact the efficacy of adoptively transferred antitumor T helper cells is unknown. METHODS CD4+ T cells with a transgenic T-cell receptor that recognize tyrosinase-related peptide (TRP)-1 melanoma antigen were polarized to the T helper 17 (Th17) phenotype and then transferred into melanoma-bearing mice preconditioned with either total body irradiation or chemotherapy. RESULTS We found that preconditioning mice with a non-myeloablative dose of total body irradiation (TBI of 5 Gy) was more effective than using an equivalently dosed non-myeloablative chemotherapy (cyclophosphamide (CTX) of 200 mg/kg) at augmenting therapeutic activity of antitumor TRP-1 Th17 cells. Antitumor Th17 cells engrafted better following preconditioning with TBI and regressed large established melanoma in all animals. Conversely, only half of mice survived long-term when preconditioned with CTX and infused with anti-melanoma Th17 cells. Interleukin (IL)-17 and interferon-γ, produced by the infused Th17 cells, were detected in animals given either TBI or CTX preconditioning. Interestingly, inflammatory cytokines (granulocyte colony stimulating factor, IL-6, monocyte chemoattractant protein-1, IL-5, and keratinocyte chemoattractant) were significantly elevated in the serum of mice preconditioned with TBI versus CTX after Th17 therapy. The addition of fludarabine (FLU, 200 mg/kg) to CTX (200 mg/kg) improved the antitumor response to the same degree mediated by TBI, whereas FLU alone with Th17 therapy was ineffective. CONCLUSIONS Our results indicate, for the first time, that the antitumor response, persistence, and cytokine profiles resulting from Th17 therapy are impacted by the specific regimen of host preconditioning. This work is important for understanding mechanisms that promote long-lived responses by adoptive cellular therapy, particularly as CD4+ based T-cell therapies are now emerging in the clinic.
Collapse
Affiliation(s)
- Megen C Wittling
- Surgery/Oncology & Microbiology/Immunology, Emory University, Atlanta, Georgia, USA
- School of Medicine, Emory University, Atlanta, Georgia, USA
| | - Hannah M Knochelmann
- Surgery/Oncology & Microbiology/Immunology, Emory University, Atlanta, Georgia, USA
- Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Megan M Wyatt
- Surgery/Oncology & Microbiology/Immunology, Emory University, Atlanta, Georgia, USA
| | - Guillermo O Rangel Rivera
- Surgery/Oncology & Microbiology/Immunology, Emory University, Atlanta, Georgia, USA
- Microbiology and Immunology, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Anna C Cole
- Surgery/Oncology & Microbiology/Immunology, Emory University, Atlanta, Georgia, USA
| | | | - Chrystal M Paulos
- Surgery/Oncology & Microbiology/Immunology, Emory University, Atlanta, Georgia, USA
| |
Collapse
|
46
|
Benmelech S, Le T, McKay M, Nam J, Subramaniam K, Tellez D, Vlasak G, Mak M. Biophysical and biochemical aspects of immune cell-tumor microenvironment interactions. APL Bioeng 2024; 8:021502. [PMID: 38572312 PMCID: PMC10990568 DOI: 10.1063/5.0195244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Accepted: 03/19/2024] [Indexed: 04/05/2024] Open
Abstract
The tumor microenvironment (TME), composed of and influenced by a heterogeneous set of cancer cells and an extracellular matrix, plays a crucial role in cancer progression. The biophysical aspects of the TME (namely, its architecture and mechanics) regulate interactions and spatial distributions of cancer cells and immune cells. In this review, we discuss the factors of the TME-notably, the extracellular matrix, as well as tumor and stromal cells-that contribute to a pro-tumor, immunosuppressive response. We then discuss the ways in which cells of the innate and adaptive immune systems respond to tumors from both biochemical and biophysical perspectives, with increased focus on CD8+ and CD4+ T cells. Building upon this information, we turn to immune-based antitumor interventions-specifically, recent biophysical breakthroughs aimed at improving CAR-T cell therapy.
Collapse
Affiliation(s)
- Shoham Benmelech
- Department of Biomedical Engineering, Yale University, New Haven, Connecticut 06511, USA
| | - Thien Le
- Department of Biomedical Engineering, Yale University, New Haven, Connecticut 06511, USA
| | - Maggie McKay
- Department of Biomedical Engineering, Yale University, New Haven, Connecticut 06511, USA
| | - Jungmin Nam
- Department of Biomedical Engineering, Yale University, New Haven, Connecticut 06511, USA
| | - Krupakar Subramaniam
- Department of Molecular, Cellular, and Developmental Biology, Yale University, New Haven, Connecticut 06511, USA
| | - Daniela Tellez
- Department of Biomedical Engineering, Yale University, New Haven, Connecticut 06511, USA
| | - Grace Vlasak
- Department of Biomedical Engineering, Yale University, New Haven, Connecticut 06511, USA
| | - Michael Mak
- Department of Biomedical Engineering, Yale University, New Haven, Connecticut 06511, USA
| |
Collapse
|
47
|
Zhang C, Li D, Zhang X, Dai R, Kang W, Li Y, Liu Q, Gao M, Zheng Z, Zhang R, Wen Z. Dual regulation of osteosarcoma hypoxia microenvironment by a bioinspired oxygen nanogenerator for precise single-laser synergistic photodynamic/photothermal/induced antitumor immunity therapy. Mater Today Bio 2024; 26:101054. [PMID: 38633865 PMCID: PMC11021954 DOI: 10.1016/j.mtbio.2024.101054] [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: 01/28/2024] [Revised: 04/03/2024] [Accepted: 04/09/2024] [Indexed: 04/19/2024] Open
Abstract
The hypoxic tumor microenvironment (TME) of osteosarcoma (OS) is the Achilles' heel of oxygen-dependent photodynamic therapy (PDT), and tremendous challenges are confronted to reverse the hypoxia. Herein, we proposed a "reducing expenditure of O2 and broadening sources" dual-strategy and constructed ultrasmall IrO2@BSA-ATO nanogenerators (NGs) for decreasing the O2-consumption and elevating the O2-supply simultaneously. As O2 NGs, the intrinsic catalase (CAT) activity could precisely decompose the overexpressed H2O2 to produce O2 in situ, enabling exogenous O2 infusion. Moreover, the cell respiration inhibitor atovaquone (ATO) would be at the tumor sites, effectively inhibiting cell respiration and elevating oxygen content for endogenous O2 conservation. As a result, IrO2@BSA-ATO NGs systematically increase tumor oxygenation in dual ways and significantly enhance the antitumor efficacy of PDT. Moreover, the extraordinary photothermal conversion efficiency allows the implementation of precise photothermal therapy (PTT) under photoacoustic guidance. Upon a single laser irradiation, this synergistic PDT, PTT, and the following immunosuppression regulation performance of IrO2@BSA-ATO NGs achieved a superior tumor cooperative eradicating capability both in vitro and in vivo. Taken together, this study proposes an innovative dual-strategy to address the serious hypoxia problem, and this microenvironment-regulable IrO2@BSA-ATO NGs as a multifunctional theranostics platform shows great potential for OS therapy.
Collapse
Affiliation(s)
- Chongqing Zhang
- Department of Neurology, Brain Ultrasound, First Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang Province, China
- Medical Imaging Department, Shanxi Province Cancer Hospital (Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University), Taiyuan, 030001, China
| | - Dongsheng Li
- Department of Radiology, Fifth Hospital of Shanxi Medical University (Shanxi Provincial People's Hospital), Taiyuan, 030000, China
| | - Xin Zhang
- Medical Imaging Department, Shanxi Province Cancer Hospital (Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University), Taiyuan, 030001, China
| | - Rong Dai
- Department of Radiology, Fifth Hospital of Shanxi Medical University (Shanxi Provincial People's Hospital), Taiyuan, 030000, China
| | - Weiwei Kang
- Department of Radiology, Fifth Hospital of Shanxi Medical University (Shanxi Provincial People's Hospital), Taiyuan, 030000, China
| | - Yao Li
- Department of Radiology, Fifth Hospital of Shanxi Medical University (Shanxi Provincial People's Hospital), Taiyuan, 030000, China
| | - Qin Liu
- Department of Radiology, Fifth Hospital of Shanxi Medical University (Shanxi Provincial People's Hospital), Taiyuan, 030000, China
| | - Mengting Gao
- Department of Radiology, Fifth Hospital of Shanxi Medical University (Shanxi Provincial People's Hospital), Taiyuan, 030000, China
| | - Ziliang Zheng
- Department of Radiology, Fifth Hospital of Shanxi Medical University (Shanxi Provincial People's Hospital), Taiyuan, 030000, China
| | - Ruiping Zhang
- Department of Radiology, Fifth Hospital of Shanxi Medical University (Shanxi Provincial People's Hospital), Taiyuan, 030000, China
| | - Zhaohui Wen
- Department of Neurology, Brain Ultrasound, First Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang Province, China
| |
Collapse
|
48
|
Ye Z, Cheng P, Huang Q, Hu J, Huang L, Hu G. Immunocytes interact directly with cancer cells in the tumor microenvironment: one coin with two sides and future perspectives. Front Immunol 2024; 15:1388176. [PMID: 38840908 PMCID: PMC11150710 DOI: 10.3389/fimmu.2024.1388176] [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: 02/19/2024] [Accepted: 05/07/2024] [Indexed: 06/07/2024] Open
Abstract
The tumor microenvironment is closely linked to the initiation, promotion, and progression of solid tumors. Among its constitutions, immunologic cells emerge as critical players, facilitating immune evasion and tumor progression. Apart from their indirect impact on anti-tumor immunity, immunocytes directly influence neoplastic cells, either bolstering or impeding tumor advancement. However, current therapeutic modalities aimed at alleviating immunosuppression from regulatory cells on effector immune cell populations may not consistently yield satisfactory results in various solid tumors, such as breast carcinoma, colorectal cancer, etc. Therefore, this review outlines and summarizes the direct, dualistic effects of immunocytes such as T cells, innate lymphoid cells, B cells, eosinophils, and tumor-associated macrophages on tumor cells within the tumor microenvironment. The review also delves into the underlying mechanisms involved and presents the outcomes of clinical trials based on these direct effects, aiming to propose innovative and efficacious therapeutic strategies for addressing solid tumors.
Collapse
Affiliation(s)
- Zhiyi Ye
- Department of General Surgery (Breast and Thyroid Surgery), Shaoxing People’s Hospital; Shaoxing Hospital, Zhejiang University School of Medicine, Zhejiang, China
| | - Pu Cheng
- Department of Gynecology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Qi Huang
- Department of Oncology, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Department of Oncology, Anhui Medical University, Hefei, Anhui, China
| | - Jingjing Hu
- School of Medicine, Shaoxing University, Zhejiang, China
| | - Liming Huang
- Department of General Surgery (Breast and Thyroid Surgery), Shaoxing People’s Hospital; Shaoxing Hospital, Zhejiang University School of Medicine, Zhejiang, China
| | - Guoming Hu
- Department of General Surgery (Breast and Thyroid Surgery), Shaoxing People’s Hospital, Shaoxing Hospital, Zhejiang University School of Medicine, Shaoxing, Zhejiang, China
- Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, Hangzhou, Zhejiang, China
| |
Collapse
|
49
|
Rustamadji P, Wiyarta E, Pramono M, Maulanisa SC. Response to Neoadjuvant Chemotherapy in Invasive Breast Cancer Predicted by CD4+, CD8+, and FOXP3+ Tumor-Infiltrating Lymphocytes. Asian Pac J Cancer Prev 2024; 25:1607-1613. [PMID: 38809632 PMCID: PMC11318808 DOI: 10.31557/apjcp.2024.25.5.1607] [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: 11/22/2023] [Accepted: 05/04/2023] [Indexed: 05/31/2024] Open
Abstract
BACKGROUND Response to neoadjuvant chemotherapy (NC) in individuals with invasive breast cancer (IBC) must be monitored, and biomarkers are needed. NC can activate an anti-tumour immune response in its microenvironment, known as Tumor-infiltrating Lymphocytes (TIL). TIL components believed to have great potential as predictors are CD4+, CD8+, and FOXP3+ TIL. This study aims to explore TIL components that can potentially be predictive biomarkers of NC pathological responses. METHODS A sample size of 40 were analyzed based on the relationship between CD4+, CD8+, and FOXP3+ TIL expression with the Miller-Payne (MP) grading system. Age, tumour grade, PR, ER, Ki-67, and HER2 were also evaluated. CD4+, CD8+, and FOXP3+ TIL expressions were analayzed by IHC staining, while other data were collected from archives. Data was analyzed using univariate and multivariate analysis. RESULTS Univariate analysis showed a significant relationship between CD4+ TIL and MP (p<0.001), CD8+ and MP (p=0.004), and FOXP3 with MP (p<0.001). The simultaneous integration of the three biomarkers in one model was not good enough to be a predictive model. Therefore, an exploratory analysis was conducted by testing several alternative models that combined two of the three existing biomarkers. It turned out that CD4+ TIL in model 2 (CD4+CD8+) and FOXP3+ TIL in model 4 (CD8+FOXP3+) showed significant coefficient values. Moreover, all of the threshold coefficients in model 4 are significant. CONCLUSION This study shows that CD4+, CD8+, and FOXP3+ TIL have promising potential as predictive biomarkers. In particular, FOXP3+ is dominant in predictive models of pathological response in patients with IBC.
Collapse
MESH Headings
- Humans
- Lymphocytes, Tumor-Infiltrating/immunology
- Lymphocytes, Tumor-Infiltrating/metabolism
- Female
- Forkhead Transcription Factors/metabolism
- Breast Neoplasms/drug therapy
- Breast Neoplasms/pathology
- Breast Neoplasms/immunology
- Breast Neoplasms/metabolism
- Neoadjuvant Therapy/methods
- Middle Aged
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/metabolism
- Biomarkers, Tumor/metabolism
- Prognosis
- CD4-Positive T-Lymphocytes/immunology
- CD4-Positive T-Lymphocytes/metabolism
- Adult
- Follow-Up Studies
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Tumor Microenvironment/immunology
- Neoplasm Invasiveness
- Aged
- Carcinoma, Ductal, Breast/drug therapy
- Carcinoma, Ductal, Breast/pathology
- Carcinoma, Ductal, Breast/immunology
- Carcinoma, Ductal, Breast/metabolism
Collapse
Affiliation(s)
- Primariadewi Rustamadji
- Department of Anatomic Pathology, Faculty of Medicine Universitas Indonesia-Dr. Cipto Mangunkusumo National Hospital, Jakarta, Indonesia.
| | | | | | | |
Collapse
|
50
|
Kannen V, Grant DM, Matthews J. The mast cell-T lymphocyte axis impacts cancer: Friend or foe? Cancer Lett 2024; 588:216805. [PMID: 38462035 DOI: 10.1016/j.canlet.2024.216805] [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: 08/24/2023] [Revised: 02/01/2024] [Accepted: 03/06/2024] [Indexed: 03/12/2024]
Abstract
Crosstalk between mast cells (MCs) and T lymphocytes (TLs) releases specific signals that create an environment conducive to tumor development. Conversely, they can protect against cancer by targeting tumor cells for destruction. Although their role in immunity and cancer is complex, their potential in anticancer strategies is often underestimated. When peripheral MCs are activated, they can affect cancer development. Tumor-infiltrating TLs may malfunction and contribute to aggressive cancer and poor prognoses. One promising approach for cancer patients is TL-based immunotherapies. Recent reports suggest that MCs modulate TL activity in solid tumors and may be a potential therapeutic layer in multitargeting anticancer strategies. Pharmacologically modulating MC activity can enhance the anticancer cytotoxic TL response in tumors. By identifying tumor-specific targets, it has been possible to genetically alter patients' cells into fully humanized anticancer cellular therapies for autologous transplantation, including the engineering of TLs and MCs to target and kill cancer cells. Hence, recent scientific evidence provides a broader understanding of MC-TL activity in cancer.
Collapse
Affiliation(s)
- Vinicius Kannen
- Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada.
| | - Denis M Grant
- Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada
| | - Jason Matthews
- Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada; Nutrition, University of Oslo, Oslo, Norway
| |
Collapse
|