1
|
Trautmann T, Yakobian N, Nguyen R. CAR T-cells for pediatric solid tumors: where to go from here? Cancer Metastasis Rev 2024; 43:1445-1461. [PMID: 39317919 PMCID: PMC11554711 DOI: 10.1007/s10555-024-10214-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Accepted: 09/13/2024] [Indexed: 09/26/2024]
Abstract
Despite the great success that chimeric antigen receptor (CAR) T-cells have had in patients with B-cell malignancies and multiple myeloma, they continue to have limited efficacy against most solid tumors. Especially in the pediatric population, pre- and post-treatment biopsies are rarely performed due to ethical reasons, and thus, our understanding is still very limited regarding the mechanisms in the tumor microenvironment by which tumor cells exclude effectors and attract immune-suppressive cells. Nevertheless, based on the principles that are known, current T-cell engineering has leveraged some of these processes and created more potent CAR T-cells. The recent discovery of new oncofetal antigens and progress made in CAR design have expanded the potential pool of candidate antigens for therapeutic development. The most promising approaches to enhance CAR T-cells are novel CAR gating strategies, creative ways of cytokine delivery to the TME without enhancing systemic toxicity, and hijacking the chemokine axis of tumors for migratory purposes. With these new modifications, the next step in the era of CAR T-cell development will be the clinical validation of these promising preclinical findings.
Collapse
Affiliation(s)
- Tina Trautmann
- Pediatric Oncology Branch, NCI, NIH, NCI, 10 Center Drive, 1W-5832, Bethesda, MD, 20892, USA
| | - Natalia Yakobian
- Pediatric Oncology Branch, NCI, NIH, NCI, 10 Center Drive, 1W-5832, Bethesda, MD, 20892, USA
| | - Rosa Nguyen
- Pediatric Oncology Branch, NCI, NIH, NCI, 10 Center Drive, 1W-5832, Bethesda, MD, 20892, USA.
| |
Collapse
|
2
|
Lin KR, Li PX, Zhu XH, Mao XF, Peng JL, Chen XP, SiTu CY, Zhang LF, Luo W, Han YB, Yu SF. Peripheral immune characteristics and subset disorder in reproductive females with endometriosis. Front Immunol 2024; 15:1431175. [PMID: 39669572 PMCID: PMC11634862 DOI: 10.3389/fimmu.2024.1431175] [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: 05/13/2024] [Accepted: 11/11/2024] [Indexed: 12/14/2024] Open
Abstract
Pathogenesis of endometriosis (EN) is still unknown, but growing evidence suggests that immune regulation may be important, and the pattern of peripheral immune changes in reproductive women with EN has yet to be fully explored. In this study, we conducted a comprehensive and systematic analysis of immune cell subsets within T cells, B cells, NK cells, and γδ T cells in peripheral blood (PB) samples from women with EN, women with uterine fibroids (UF) but without EN (UF-alone), and healthy controls using multi-parameter flow cytometry. Our findings revealed that UF, a common comorbidity of EN, exhibited similar peripheral immune features to EN, particularly in T cell and B cell immunity. Compared to healthy controls, we constructed the peripheral immune profile of EN. This profile highlighted that the immunopathogenic factors in EN predominantly relate to the immune disorder of B cells and their subsets, as well as the functional abnormalities within immune cell subsets of CD4+ T cells, CD8+ T cells, and γδ T cells. Moreover, using the random forest (RF) machine-learning method, we developed a diagnostic model that can effectively identify the patients with EN from healthy controls. The immune factors identified within this model could be pivotal for unraveling the immune pathogenic mechanisms of EN. Our study is the first to present a comprehensive depiction of the circulating immune features in EN, although the detailed roles and underlying mechanisms of these immune factors in the context of EN require further investigation.
Collapse
Affiliation(s)
- Kai-Rong Lin
- Institute of Translational Medicine, The First People'sHospital of Foshan, Foshan, Guangdong, China
| | - Pei-Xian Li
- Institute of Translational Medicine, The First People'sHospital of Foshan, Foshan, Guangdong, China
| | - Xiao-hong Zhu
- Department of Gynecology, The First People’s Hospital of Foshan, Foshan, Guangdong, China
| | - Xiao-fan Mao
- Institute of Translational Medicine, The First People'sHospital of Foshan, Foshan, Guangdong, China
| | - Jia-Li Peng
- Department of Gynecology, The First People’s Hospital of Foshan, Foshan, Guangdong, China
| | - Xiang-Ping Chen
- Institute of Translational Medicine, The First People'sHospital of Foshan, Foshan, Guangdong, China
| | - Cui-Yao SiTu
- Department of Gynecology, The First People’s Hospital of Foshan, Foshan, Guangdong, China
| | - Li-Fang Zhang
- Institute of Translational Medicine, The First People'sHospital of Foshan, Foshan, Guangdong, China
| | - Wei Luo
- Institute of Translational Medicine, The First People'sHospital of Foshan, Foshan, Guangdong, China
| | - Yu-Bin Han
- Department of Gynecology, The First People’s Hospital of Foshan, Foshan, Guangdong, China
| | - Si-Fei Yu
- Institute of Translational Medicine, The First People'sHospital of Foshan, Foshan, Guangdong, China
| |
Collapse
|
3
|
Chuensirikulchai K, Pata S, Laopajon W, Takheaw N, Kotemul K, Jindaphun K, Khummuang S, Kasinrerk W. Identification of different functions of CD8 + T cell subpopulations by a novel monoclonal antibody. Immunology 2024; 173:321-338. [PMID: 38922845 DOI: 10.1111/imm.13826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 06/05/2024] [Indexed: 06/28/2024] Open
Abstract
The explicit identification of CD8+ T cell subpopulation is important for deciphering the role of CD8+ T cells for protecting our body against invading pathogens and cancer. Our generated monoclonal antibody (mAb), named FE-1H10, recognized two novel subpopulations of peripheral blood CD8+ T cells, FE-1H10+ and FE-1H10- CD8+ T cells. The molecule recognized by mAb FE-1H10 (FE-1H10 molecules) had a higher distribution on effector memory CD8+ T cell subsets. The functions of FE-1H10- and FE-1H10+ CD8+ T cells were investigated. T cell proliferation assays revealed that FE-1H10- CD8+ T cells exhibited a higher proliferation rate than FE-1H10+ CD8+ T cells, whereas FE-1H10+ CD8+ T cells produced higher levels of IFN-γ and TNF-α than FE-1H10- CD8+ T cells. In T cell cytotoxicity assays, FE-1H10+ CD8+ T cells were able to kill target cells better than FE-1H10- CD8+ T cells. RNA-sequencing analysis confirmed that these subpopulations were distinct: FE-1H10+ CD8+ T cells have higher expression of genes involved in effector functions (IFNG, TNF, GZMB, PRF1, GNLY, FASL, CX3CR1) while FE-1H10- CD8+ T cells have greater expression of genes related to memory CD8+ T cell populations (CCR7, SELL, TCF7, CD40LG). The results suggested that mAb FE-1H10 identifies two novel distinctive CD8+ T cell subpopulations. The FE-1H10+ CD8+ T cells carried a superior functionality in response to tumour cells. The uncover of these novel CD8+ T cell subpopulations may be the basis knowledge of an optional immunotherapy for the selection of potential CD8+ T cells in cancer treatment.
Collapse
Affiliation(s)
| | - Supansa Pata
- Division of Clinical Immunology, Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
- Biomedical Technology Research Center, National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency at the Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
| | - Witida Laopajon
- Division of Clinical Immunology, Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
- Biomedical Technology Research Center, National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency at the Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
| | - Nuchjira Takheaw
- Division of Clinical Immunology, Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
- Biomedical Technology Research Center, National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency at the Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
| | - Kamonporn Kotemul
- Division of Clinical Immunology, Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
| | - Kanyaruck Jindaphun
- Division of Clinical Immunology, Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
| | - Saichit Khummuang
- Division of Clinical Immunology, Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
| | - Watchara Kasinrerk
- Division of Clinical Immunology, Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
- Biomedical Technology Research Center, National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency at the Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
| |
Collapse
|
4
|
Loffredo LF, Kaiser KA, Kornberg A, Rao S, de Los Santos-Alexis K, Han A, Arpaia N. An amphiregulin reporter mouse enables transcriptional and clonal expansion analysis of reparative lung Treg cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.26.615245. [PMID: 39386607 PMCID: PMC11463663 DOI: 10.1101/2024.09.26.615245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Regulatory T (Treg) cells are known to play critical roles in tissue repair via provision of growth factors such as amphiregulin (Areg). Areg-producing Treg cells have previously been difficult to study because of an inability to isolate live Areg-producing cells. In this report, we created a novel reporter mouse to detect Areg expression in live cells ( Areg Thy1.1 ). We employed influenza A and bleomycin models of lung damage to sort Areg-producing and -non-producing Treg cells for transcriptomic analyses. Single cell RNA-seq revealed distinct subpopulations of Treg cells and allowed transcriptomic comparisons of damage-induced populations. Single cell TCR sequencing showed that Treg cell clonal expansion is biased towards Areg-producing Treg cells, and largely occurs within damage-induced subgroups. Gene module analysis revealed functional divergence of Treg cells into immunosuppression-oriented and tissue repair-oriented groups, leading to identification of candidate receptors for induction of repair activity in Treg cells. We tested these using an ex vivo assay for Treg cell-mediated tissue repair, identifying 4-1BB agonism as a novel mechanism for reparative activity induction. Overall, we demonstrate that the Areg Thy1.1 mouse is a promising tool for investigating tissue repair activity in leukocytes.
Collapse
|
5
|
Chen X, Soma L, Murphy C, Tretiakova M, Naresh KN, Fromm JR. Utility of CCR7 to differentiate classic Hodgkin lymphoma and other B-cell lymphomas by flow cytometry and immunohistochemistry. Am J Clin Pathol 2024:aqae119. [PMID: 39288406 DOI: 10.1093/ajcp/aqae119] [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: 05/06/2024] [Accepted: 08/16/2024] [Indexed: 09/19/2024] Open
Abstract
OBJECTIVES Classic Hodgkin lymphoma (CHL) is characterized by infrequent neoplastic Hodgkin and Reed-Sternberg (HRS) cells in an inflammatory background. The diagnostic utility of CC-chemokine receptor 7 (CCR7) in CHL was explored using flow cytometry and immunohistochemistry (IHC). METHODS Neoplastic specimens and non-neoplastic lymph nodes were immunophenotyped and CCR7 expression was measured semiquantitatively by flow cytometry (clone 3D12) and IHC (clone 150503). RESULTS Our results showed that CCR7 was expressed on HRS cells in the vast majority of CHL cases (45/48 by flow cytometry, 57/59 by IHC) but rarely expressed in neoplastic cells in diffuse large B-cell lymphoma, not otherwise specified (1/25 by flow cytometry, 2/40 by IHC) and nodular lymphocyte predominant Hodgkin lymphoma (0/4 by flow cytometry, 1/13 by IHC). Primary mediastinal large B-cell lymphoma (PMLBCL) revealed weak CCR7 expression by flow cytometry in most cases (8/10) but only occasionally by IHC (2/12). Both cases (2/2) of T-cell/histiocyte-rich large B-cell lymphoma (THRLBCL) also showed CCR7 expression detected by flow cytometry compared with IHC (0/7). The HRS cells demonstrated a greater percentage of positive cells and greater antigen intensity than the other B-cell lymphomas by IHC. The expression identified by flow cytometry in PMLBCL and THRLBCL but not by IHC suggests that there may be differences in the detection capabilities of the 2 techniques or the 2 CCR7 clones used. CONCLUSIONS The expression of CCR7 in HRS cells suggests its potential utility in differentiating CHL from other B-cell lymphomas. Incorporating CCR7 into flow cytometry and IHC panels may further enhance the diagnostic sensitivity of CHL.
Collapse
Affiliation(s)
- Xueyan Chen
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, US
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, US
| | - Lori Soma
- Department of Pathology, City of Hope, Duarte, CA, US
| | - Claire Murphy
- Pathology Consultants, PC, Eugene/Springfield Lab, Springfield, OR, US
| | - Maria Tretiakova
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, US
| | - Kikkeri N Naresh
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, US
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, US
| | - Jonathan R Fromm
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, US
| |
Collapse
|
6
|
Krakow EF, Brault M, Summers C, Cunningham TM, Biernacki MA, Black RG, Woodward KB, Vartanian N, Kanaan SB, Yeh AC, Dossa RG, Bar M, Cassaday RD, Dahlberg A, Till BG, Denker AE, Yeung CCS, Gooley TA, Maloney DG, Riddell SR, Greenberg PD, Chapuis AG, Newell EW, Furlan SN, Bleakley M. HA-1-targeted T-cell receptor T-cell therapy for recurrent leukemia after hematopoietic stem cell transplantation. Blood 2024; 144:1069-1082. [PMID: 38683966 PMCID: PMC11406181 DOI: 10.1182/blood.2024024105] [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: 01/29/2024] [Revised: 03/27/2024] [Accepted: 04/10/2024] [Indexed: 05/02/2024] Open
Abstract
ABSTRACT Relapse is the leading cause of death after allogeneic hematopoietic stem cell transplantation (HCT) for leukemia. T cells engineered by gene transfer to express T cell receptors (TCR; TCR-T) specific for hematopoietic-restricted minor histocompatibility (H) antigens may provide a potent selective antileukemic effect post-HCT. We conducted a phase 1 clinical trial using a novel TCR-T product targeting the minor H antigen, HA-1, to treat or consolidate treatment of persistent or recurrent leukemia and myeloid neoplasms. The primary objective was to evaluate the feasibility and safety of administration of HA-1 TCR-T after HCT. CD8+ and CD4+ T cells expressing the HA-1 TCR and a CD8 coreceptor were successfully manufactured from HA-1-disparate HCT donors. One or more infusions of HA-1 TCR-T following lymphodepleting chemotherapy were administered to 9 HCT recipients who had developed disease recurrence after HCT. TCR-T cells expanded and persisted in vivo after adoptive transfer. No dose-limiting toxicities occurred. Although the study was not designed to assess efficacy, 4 patients achieved or maintained complete remissions following lymphodepletion and HA-1 TCR-T, with 1 patient still in remission at >2 years. Single-cell RNA sequencing of relapsing/progressive leukemia after TCR-T therapy identified upregulated molecules associated with T-cell dysfunction or cancer cell survival. HA-1 TCR-T therapy appears feasible and safe and shows preliminary signals of efficacy. This clinical trial was registered at ClinicalTrials.gov as #NCT03326921.
Collapse
Affiliation(s)
- Elizabeth F. Krakow
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA
- Department of Medicine, University of Washington School of Medicine, Seattle, WA
| | - Michelle Brault
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Corinne Summers
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA
- Cancer and Blood Disorders Center, Seattle Children's Hospital, Seattle, WA
- Department of Pediatrics, University of Washington School of Medicine, Seattle, WA
| | - Tanya M. Cunningham
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Melinda A. Biernacki
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - R. Graeme Black
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Kyle B. Woodward
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Nicole Vartanian
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Sami B. Kanaan
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Albert C. Yeh
- Department of Medicine, University of Washington School of Medicine, Seattle, WA
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Robson G. Dossa
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Merav Bar
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Ryan D. Cassaday
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA
- Department of Medicine, University of Washington School of Medicine, Seattle, WA
| | - Ann Dahlberg
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA
- Cancer and Blood Disorders Center, Seattle Children's Hospital, Seattle, WA
- Department of Pediatrics, University of Washington School of Medicine, Seattle, WA
| | - Brian G. Till
- Department of Medicine, University of Washington School of Medicine, Seattle, WA
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA
| | | | - Cecilia C. S. Yeung
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, WA
| | - Ted A. Gooley
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - David G. Maloney
- Department of Medicine, University of Washington School of Medicine, Seattle, WA
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Stanley R. Riddell
- Department of Medicine, University of Washington School of Medicine, Seattle, WA
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Philip D. Greenberg
- Department of Medicine, University of Washington School of Medicine, Seattle, WA
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA
- Department of Immunology, University of Washington School of Medicine, Seattle, WA
| | - Aude G. Chapuis
- Department of Medicine, University of Washington School of Medicine, Seattle, WA
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Evan W. Newell
- Vaccine and Infection Disease Division, Fred Hutchinson Cancer Center, Seattle, WA
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Scott N. Furlan
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA
- Cancer and Blood Disorders Center, Seattle Children's Hospital, Seattle, WA
- Department of Pediatrics, University of Washington School of Medicine, Seattle, WA
| | - Marie Bleakley
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA
- Cancer and Blood Disorders Center, Seattle Children's Hospital, Seattle, WA
- Department of Pediatrics, University of Washington School of Medicine, Seattle, WA
| |
Collapse
|
7
|
Van der Vreken A, Vanderkerken K, De Bruyne E, De Veirman K, Breckpot K, Menu E. Fueling CARs: metabolic strategies to enhance CAR T-cell therapy. Exp Hematol Oncol 2024; 13:66. [PMID: 38987856 PMCID: PMC11238373 DOI: 10.1186/s40164-024-00535-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 07/02/2024] [Indexed: 07/12/2024] Open
Abstract
CAR T cells are widely applied for relapsed hematological cancer patients. With six approved cell therapies, for Multiple Myeloma and other B-cell malignancies, new insights emerge. Profound evidence shows that patients who fail CAR T-cell therapy have, aside from antigen escape, a more glycolytic and weakened metabolism in their CAR T cells, accompanied by a short lifespan. Recent advances show that CAR T cells can be metabolically engineered towards oxidative phosphorylation, which increases their longevity via epigenetic and phenotypical changes. In this review we elucidate various strategies to rewire their metabolism, including the design of the CAR construct, co-stimulus choice, genetic modifications of metabolic genes, and pharmacological interventions. We discuss their potential to enhance CAR T-cell functioning and persistence through memory imprinting, thereby improving outcomes. Furthermore, we link the pharmacological treatments with their anti-cancer properties in hematological malignancies to ultimately suggest novel combination strategies.
Collapse
Affiliation(s)
- Arne Van der Vreken
- Translational Oncology Research Center, Team Hematology and Immunology, Vrije Universiteit Brussel, Laarbeeklaan 103, Brussels, 1090, Belgium
| | - Karin Vanderkerken
- Translational Oncology Research Center, Team Hematology and Immunology, Vrije Universiteit Brussel, Laarbeeklaan 103, Brussels, 1090, Belgium
| | - Elke De Bruyne
- Translational Oncology Research Center, Team Hematology and Immunology, Vrije Universiteit Brussel, Laarbeeklaan 103, Brussels, 1090, Belgium
| | - Kim De Veirman
- Translational Oncology Research Center, Team Hematology and Immunology, Vrije Universiteit Brussel, Laarbeeklaan 103, Brussels, 1090, Belgium
| | - Karine Breckpot
- Translational Oncology Research Center, Team Laboratory of Cellular and Molecular Therapy, Vrije Universiteit Brussel, Laarbeeklaan 103, Brussels, 1090, Belgium
| | - Eline Menu
- Translational Oncology Research Center, Team Hematology and Immunology, Vrije Universiteit Brussel, Laarbeeklaan 103, Brussels, 1090, Belgium.
| |
Collapse
|
8
|
Lin CP, Levy PL, Alflen A, Apriamashvili G, Ligtenberg MA, Vredevoogd DW, Bleijerveld OB, Alkan F, Malka Y, Hoekman L, Markovits E, George A, Traets JJH, Krijgsman O, van Vliet A, Poźniak J, Pulido-Vicuña CA, de Bruijn B, van Hal-van Veen SE, Boshuizen J, van der Helm PW, Díaz-Gómez J, Warda H, Behrens LM, Mardesic P, Dehni B, Visser NL, Marine JC, Markel G, Faller WJ, Altelaar M, Agami R, Besser MJ, Peeper DS. Multimodal stimulation screens reveal unique and shared genes limiting T cell fitness. Cancer Cell 2024; 42:623-645.e10. [PMID: 38490212 PMCID: PMC11003465 DOI: 10.1016/j.ccell.2024.02.016] [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: 07/07/2023] [Revised: 01/03/2024] [Accepted: 02/22/2024] [Indexed: 03/17/2024]
Abstract
Genes limiting T cell antitumor activity may serve as therapeutic targets. It has not been systematically studied whether there are regulators that uniquely or broadly contribute to T cell fitness. We perform genome-scale CRISPR-Cas9 knockout screens in primary CD8 T cells to uncover genes negatively impacting fitness upon three modes of stimulation: (1) intense, triggering activation-induced cell death (AICD); (2) acute, triggering expansion; (3) chronic, causing dysfunction. Besides established regulators, we uncover genes controlling T cell fitness either specifically or commonly upon differential stimulation. Dap5 ablation, ranking highly in all three screens, increases translation while enhancing tumor killing. Loss of Icam1-mediated homotypic T cell clustering amplifies cell expansion and effector functions after both acute and intense stimulation. Lastly, Ctbp1 inactivation induces functional T cell persistence exclusively upon chronic stimulation. Our results functionally annotate fitness regulators based on their unique or shared contribution to traits limiting T cell antitumor activity.
Collapse
Affiliation(s)
- Chun-Pu Lin
- Division of Molecular Oncology and Immunology, Oncode Institute, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, the Netherlands
| | - Pierre L Levy
- Division of Molecular Oncology and Immunology, Oncode Institute, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, the Netherlands; Tumor Immunology and Immunotherapy Group, Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron Barcelona Hospital Campus, 08035 Barcelona, Spain
| | - Astrid Alflen
- Division of Molecular Oncology and Immunology, Oncode Institute, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, the Netherlands; Department of Hematology and Medical Oncology, University Medical Center, Johannes Gutenberg-University, 55131 Mainz, Germany; Research Center for Immunotherapy (FZI), University Medical Center, Johannes Gutenberg-University, 55131 Mainz, Germany
| | - Georgi Apriamashvili
- Division of Molecular Oncology and Immunology, Oncode Institute, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, the Netherlands
| | - Maarten A Ligtenberg
- Division of Molecular Oncology and Immunology, Oncode Institute, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, the Netherlands
| | - David W Vredevoogd
- Division of Molecular Oncology and Immunology, Oncode Institute, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, the Netherlands
| | - Onno B Bleijerveld
- Proteomics Facility, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, the Netherlands
| | - Ferhat Alkan
- Division of Oncogenomics, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, the Netherlands
| | - Yuval Malka
- Division of Oncogenomics, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, the Netherlands
| | - Liesbeth Hoekman
- Proteomics Facility, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, the Netherlands
| | - Ettai Markovits
- Ella Lemelbaum Institute for Immuno-oncology and Melanoma, Sheba Medical Center, Ramat Gan 52612, Israel; Department of Clinical Microbiology and Immunology, Faculty of Medicine, Tel Aviv University, Tel-Aviv 6997801, Israel
| | - Austin George
- Division of Molecular Oncology and Immunology, Oncode Institute, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, the Netherlands
| | - Joleen J H Traets
- Division of Molecular Oncology and Immunology, Oncode Institute, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, the Netherlands; Division of Tumor Biology and Immunology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, the Netherlands
| | - Oscar Krijgsman
- Division of Molecular Oncology and Immunology, Oncode Institute, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, the Netherlands
| | - Alex van Vliet
- Division of Molecular Oncology and Immunology, Oncode Institute, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, the Netherlands
| | - Joanna Poźniak
- Laboratory for Molecular Cancer Biology, VIB Center for Cancer Biology, 3000 Leuven, Belgium; Laboratory for Molecular Cancer Biology, Department of Oncology, KU Leuven, 3000 Leuven, Belgium
| | - Carlos Ariel Pulido-Vicuña
- Laboratory for Molecular Cancer Biology, VIB Center for Cancer Biology, 3000 Leuven, Belgium; Laboratory for Molecular Cancer Biology, Department of Oncology, KU Leuven, 3000 Leuven, Belgium
| | - Beaunelle de Bruijn
- Division of Molecular Oncology and Immunology, Oncode Institute, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, the Netherlands
| | - Susan E van Hal-van Veen
- Division of Molecular Oncology and Immunology, Oncode Institute, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, the Netherlands
| | - Julia Boshuizen
- Division of Molecular Oncology and Immunology, Oncode Institute, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, the Netherlands
| | - Pim W van der Helm
- Division of Molecular Oncology and Immunology, Oncode Institute, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, the Netherlands
| | - Judit Díaz-Gómez
- Division of Molecular Oncology and Immunology, Oncode Institute, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, the Netherlands
| | - Hamdy Warda
- Division of Molecular Oncology and Immunology, Oncode Institute, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, the Netherlands
| | - Leonie M Behrens
- Division of Molecular Oncology and Immunology, Oncode Institute, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, the Netherlands
| | - Paula Mardesic
- Division of Molecular Oncology and Immunology, Oncode Institute, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, the Netherlands
| | - Bilal Dehni
- Division of Molecular Oncology and Immunology, Oncode Institute, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, the Netherlands
| | - Nils L Visser
- Division of Molecular Oncology and Immunology, Oncode Institute, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, the Netherlands
| | - Jean-Christophe Marine
- Laboratory for Molecular Cancer Biology, VIB Center for Cancer Biology, 3000 Leuven, Belgium; Laboratory for Molecular Cancer Biology, Department of Oncology, KU Leuven, 3000 Leuven, Belgium
| | - Gal Markel
- Department of Clinical Microbiology and Immunology, Faculty of Medicine, Tel Aviv University, Tel-Aviv 6997801, Israel; Davidoff Cancer Center and Samueli Integrative Cancer Pioneering Institute, Rabin Medical Center, Petach Tikva 4941492, Israel
| | - William J Faller
- Division of Oncogenomics, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, the Netherlands
| | - Maarten Altelaar
- Proteomics Facility, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, the Netherlands; Biomolecular Mass Spectrometry and Proteomics, Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Padualaan 8, 3584 CH Utrecht, the Netherlands
| | - Reuven Agami
- Division of Oncogenomics, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, the Netherlands
| | - Michal J Besser
- Ella Lemelbaum Institute for Immuno-oncology and Melanoma, Sheba Medical Center, Ramat Gan 52612, Israel; Department of Clinical Microbiology and Immunology, Faculty of Medicine, Tel Aviv University, Tel-Aviv 6997801, Israel; Davidoff Cancer Center and Samueli Integrative Cancer Pioneering Institute, Rabin Medical Center, Petach Tikva 4941492, Israel; Felsenstein Medical Research Center, Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Daniel S Peeper
- Division of Molecular Oncology and Immunology, Oncode Institute, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, the Netherlands; Department of Pathology, VU University Amsterdam, 1081 HV Amsterdam, the Netherlands.
| |
Collapse
|
9
|
Mitra S, Malik R, Wong W, Rahman A, Hartemink AJ, Pritykin Y, Dey KK, Leslie CS. Single-cell multi-ome regression models identify functional and disease-associated enhancers and enable chromatin potential analysis. Nat Genet 2024; 56:627-636. [PMID: 38514783 PMCID: PMC11018525 DOI: 10.1038/s41588-024-01689-8] [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: 06/13/2023] [Accepted: 02/14/2024] [Indexed: 03/23/2024]
Abstract
We present a gene-level regulatory model, single-cell ATAC + RNA linking (SCARlink), which predicts single-cell gene expression and links enhancers to target genes using multi-ome (scRNA-seq and scATAC-seq co-assay) sequencing data. The approach uses regularized Poisson regression on tile-level accessibility data to jointly model all regulatory effects at a gene locus, avoiding the limitations of pairwise gene-peak correlations and dependence on peak calling. SCARlink outperformed existing gene scoring methods for imputing gene expression from chromatin accessibility across high-coverage multi-ome datasets while giving comparable to improved performance on low-coverage datasets. Shapley value analysis on trained models identified cell-type-specific gene enhancers that are validated by promoter capture Hi-C and are 11× to 15× and 5× to 12× enriched in fine-mapped eQTLs and fine-mapped genome-wide association study (GWAS) variants, respectively. We further show that SCARlink-predicted and observed gene expression vectors provide a robust way to compute a chromatin potential vector field to enable developmental trajectory analysis.
Collapse
Affiliation(s)
- Sneha Mitra
- Computational and Systems Biology Program, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | | | - Wilfred Wong
- Computational and Systems Biology Program, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
- Tri-Institutional Training Program in Computational Biology and Medicine, New York City, NY, USA
| | - Afsana Rahman
- Hunter College, City University of New York, New York City, NY, USA
| | - Alexander J Hartemink
- Department of Computer Science, Duke University, Durham, NC, USA
- Program in Computational Biology and Bioinformatics, Duke University, Durham, NC, USA
- Center for Genomic and Computational Biology, Duke University, Durham, NC, USA
| | - Yuri Pritykin
- Computational and Systems Biology Program, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
- Department of Computer Science, Princeton University, Princeton, NJ, USA
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA
| | - Kushal K Dey
- Computational and Systems Biology Program, Memorial Sloan Kettering Cancer Center, New York City, NY, USA.
| | - Christina S Leslie
- Computational and Systems Biology Program, Memorial Sloan Kettering Cancer Center, New York City, NY, USA.
| |
Collapse
|
10
|
Weller S, Li X, Petersen LR, Kempen P, Clergeaud G, Andresen TL. Influence of different conjugation methods for activating antibodies on polymeric nanoparticles: Effects for polyclonal expansion of human CD8+ T cells. Int Immunopharmacol 2024; 129:111643. [PMID: 38340420 DOI: 10.1016/j.intimp.2024.111643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 01/31/2024] [Accepted: 01/31/2024] [Indexed: 02/12/2024]
Abstract
Particle-based systems have become a state-of-the-art method for in vitro expanding cytotoxic T cells by tailoring their surface with activating molecules. However, commonly used methods utilize facile carbodiimide chemistry leading to uncontrolled orientation of the immobilized antibodies on the particle surface that can lead to poor binding to target cells. To address this, selective coupling strategies utilizing regioselective chemical groups such as disulfide bridges offer a simple approach. In this work we present a set of methods to investigate the effect of polymeric nanoparticles, conjugated with either regioselective- or randomly-immobilized antiCD3 and antiCD28 antibodies, on the activation potential, expansion and expression of activation markers in T cells. We show that nanoparticles with well-oriented monovalent antibodies conjugated via maleimide require fewer ligands on the surface to efficiently expand T cells compared to bivalent antibodies randomly-immobilized via carbodiimide conjugation. Analysis of the T cell expression markers reveal that the T cell phenotype can be fine-tuned by adjusting the surface density of well-oriented antibodies, while randomly immobilized antibodies showed no differences despite their ligand density. Both conjugation techniques induced cytotoxic T cells, evidenced by analyzing their Granzyme B secretion. Furthermore, antibody orientation affects the immunological synapse and T cell activation by changing the calcium influx profile upon activation. Nanoparticles with well-oriented antibodies showed lower calcium influx compared to their bivalent randomly-immobilized counterparts. These results highlight the importance of controlling the antibody density and orientation on the nanoparticle surface via controlled coupling chemistries, helping to develop improved particle-based expansion protocols to enhance T cell therapies.
Collapse
Affiliation(s)
- Sven Weller
- Department of Health Technology, Technical University of Denmark, Kongens Lyngby, Denmark.
| | - Xin Li
- Department of Health Technology, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Lars R Petersen
- Department of Health Technology, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Paul Kempen
- Department of Health Technology, Technical University of Denmark, Kongens Lyngby, Denmark; DTU Nanolab, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Gael Clergeaud
- Department of Health Technology, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Thomas L Andresen
- Department of Health Technology, Technical University of Denmark, Kongens Lyngby, Denmark
| |
Collapse
|
11
|
Orozco RC, Marquardt K, Pratumchai I, Shaikh AF, Mowen K, Domissy A, Teijaro JR, Sherman LA. Autoimmunity-associated allele of tyrosine phosphatase gene PTPN22 enhances anti-viral immunity. PLoS Pathog 2024; 20:e1012095. [PMID: 38512979 PMCID: PMC10987006 DOI: 10.1371/journal.ppat.1012095] [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: 04/26/2023] [Revised: 04/02/2024] [Accepted: 03/04/2024] [Indexed: 03/23/2024] Open
Abstract
The 1858C>T allele of the tyrosine phosphatase PTPN22 is present in 5-10% of the North American population and is strongly associated with numerous autoimmune diseases. Although research has been done to define how this allele potentiates autoimmunity, the influence PTPN22 and its pro-autoimmune allele has in anti-viral immunity remains poorly defined. Here, we use single cell RNA-sequencing and functional studies to interrogate the impact of this pro-autoimmune allele on anti-viral immunity during Lymphocytic Choriomeningitis Virus clone 13 (LCMV-cl13) infection. Mice homozygous for this allele (PEP-619WW) clear the LCMV-cl13 virus whereas wildtype (PEP-WT) mice cannot. This is associated with enhanced anti-viral CD4 T cell responses and a more immunostimulatory CD8α- cDC phenotype. Adoptive transfer studies demonstrated that PEP-619WW enhanced anti-viral CD4 T cell function through virus-specific CD4 T cell intrinsic and extrinsic mechanisms. Taken together, our data show that the pro-autoimmune allele of Ptpn22 drives a beneficial anti-viral immune response thereby preventing what is normally a chronic virus infection.
Collapse
Affiliation(s)
- Robin C. Orozco
- Department of Immunology and Microbiology, Scripps Research, La Jolla, California, United States of America
- Department of Molecular Biosciences, University of Kansas, Lawrence, Kansas, United States of America
| | - Kristi Marquardt
- Department of Immunology and Microbiology, Scripps Research, La Jolla, California, United States of America
| | - Isaraphorn Pratumchai
- Department of Immunology and Microbiology, Scripps Research, La Jolla, California, United States of America
| | - Anam Fatima Shaikh
- Department of Molecular Biosciences, University of Kansas, Lawrence, Kansas, United States of America
| | - Kerri Mowen
- Department of Immunology and Microbiology, Scripps Research, La Jolla, California, United States of America
| | - Alain Domissy
- Genomics Core, Scripps Research, La Jolla, California, United States of America
| | - John R. Teijaro
- Department of Immunology and Microbiology, Scripps Research, La Jolla, California, United States of America
| | - Linda A. Sherman
- Department of Immunology and Microbiology, Scripps Research, La Jolla, California, United States of America
| |
Collapse
|
12
|
Dailah HG, Hommdi AA, Koriri MD, Algathlan EM, Mohan S. Potential role of immunotherapy and targeted therapy in the treatment of cancer: A contemporary nursing practice. Heliyon 2024; 10:e24559. [PMID: 38298714 PMCID: PMC10828696 DOI: 10.1016/j.heliyon.2024.e24559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Revised: 01/09/2024] [Accepted: 01/10/2024] [Indexed: 02/02/2024] Open
Abstract
Immunotherapy and targeted therapy have emerged as promising therapeutic options for cancer patients. Immunotherapies induce a host immune response that mediates long-lived tumor destruction, while targeted therapies suppress molecular mechanisms that are important for tumor maintenance and growth. In addition, cytotoxic agents and targeted therapies regulate immune responses, which increases the chances that these therapeutic approaches may be efficiently combined with immunotherapy to ameliorate clinical outcomes. Various studies have suggested that combinations of therapies that target different stages of anti-tumor immunity may be synergistic, which can lead to potent and more prolonged responses that can achieve long-lasting tumor destruction. Nurses associated with cancer patients should have a better understanding of the immunotherapies and targeted therapies, such as their efficacy profiles, mechanisms of action, as well as management and prophylaxis of adverse events. Indeed, this knowledge will be important in establishing care for cancer patients receiving immunotherapies and targeted therapies for cancer treatment. Moreover, nurses need a better understanding regarding targeted therapies and immunotherapies to ameliorate outcomes in patients receiving these therapies, as well as management and early detection of possible adverse effects, especially adverse events associated with checkpoint inhibitors and various other therapies that control T-cell activation causing autoimmune toxicity. Nurses practice in numerous settings, such as hospitals, home healthcare agencies, radiation therapy facilities, ambulatory care clinics, and community agencies. Therefore, as compared to other members of the healthcare team, nurses often have better opportunities to develop the essential rapport in providing effective nurse-led patient education, which is important for effective therapeutic outcomes and continuance of therapy. In this article, we have particularly focused on providing a detailed overview on targeted therapies and immunotherapies used in cancer treatment, management of their associated adverse events, and the impact as well as strategies of nurse-led patient education.
Collapse
Affiliation(s)
- Hamad Ghaleb Dailah
- Research and Scientific Studies Unit, College of Nursing, Jazan University, Jazan, 45142, Saudi Arabia
| | - Abdullah Abdu Hommdi
- Research and Scientific Studies Unit, College of Nursing, Jazan University, Jazan, 45142, Saudi Arabia
| | - Mahdi Dafer Koriri
- Research and Scientific Studies Unit, College of Nursing, Jazan University, Jazan, 45142, Saudi Arabia
| | - Essa Mohammed Algathlan
- Research and Scientific Studies Unit, College of Nursing, Jazan University, Jazan, 45142, Saudi Arabia
| | - Syam Mohan
- Substance Abuse and Toxicology Research Centre, Jazan University, Jazan, Saudi Arabia
- Center for Global Health Research, Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, India
- School of Health Sciences, University of Petroleum and Energy Studies, Dehradun, Uttarakhand, India
| |
Collapse
|
13
|
Albelda SM. CAR T cell therapy for patients with solid tumours: key lessons to learn and unlearn. Nat Rev Clin Oncol 2024; 21:47-66. [PMID: 37904019 DOI: 10.1038/s41571-023-00832-4] [Citation(s) in RCA: 84] [Impact Index Per Article: 84.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/09/2023] [Indexed: 11/01/2023]
Abstract
Chimeric antigen receptor (CAR) T cells have been approved for use in patients with B cell malignancies or relapsed and/or refractory multiple myeloma, yet efficacy against most solid tumours remains elusive. The limited imaging and biopsy data from clinical trials in this setting continues to hinder understanding, necessitating a reliance on imperfect preclinical models. In this Perspective, I re-evaluate current data and suggest potential pathways towards greater success, drawing lessons from the few successful trials testing CAR T cells in patients with solid tumours and the clinical experience with tumour-infiltrating lymphocytes. The most promising approaches include the use of pluripotent stem cells, co-targeting multiple mechanisms of immune evasion, employing multiple co-stimulatory domains, and CAR ligand-targeting vaccines. An alternative strategy focused on administering multiple doses of short-lived CAR T cells in an attempt to pre-empt exhaustion and maintain a functional effector pool should also be considered.
Collapse
Affiliation(s)
- Steven M Albelda
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Pulmonary and Critical Care Division, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
14
|
Samborska I, Maievskyi O, Podzihun L, Lavrynenko V. Features of immune reactivity of the spleen and mechanisms of organ damage under the influence of animal venom toxins including scorpions (review). WIADOMOSCI LEKARSKIE (WARSAW, POLAND : 1960) 2024; 77:120-125. [PMID: 38431816 DOI: 10.36740/wlek202401115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/05/2024]
Abstract
OBJECTIVE Aim: To establish features of immune reactivity of the spleen and mechanisms of organ damage under the influence of animal venom toxins including scorpions. PATIENTS AND METHODS Materials and Methods: A thorough literature analysis was conducted on the basis of PubMed, Google Scholar, Web of Science, and Scopus databases. When processing the search results, we chose the newest publications up to 5 years old or the most thorough publications that vividly described the essence of our topic. CONCLUSION Conclusions: Spleen plays a leading role in the implementation of the body's defense processes, the elimination of structural elements affected by toxins, and the restoration of immune homeostasis. Its participation in the formation of the immune response can be accompanied by qualitative and quantitative changes in histological organization. Morpho-functional changes in the spleen under the action of animal venom toxins currently require careful study, because from the information available in the literature today, it is not possible to clearly construct a complete picture of lesions of certain components of the organ at the microscopic or submicroscopic levels. Therefore, this direction of research in the medical field is currently relevant, taking into account the existence of a large number of poisonous animals, including scorpions.
Collapse
Affiliation(s)
- Inha Samborska
- NATIONAL PIROGOV MEMORIAL MEDICAL UNIVERSITY, VINNYTSIA, UKRAINE
| | - Oleksandr Maievskyi
- EDUCATIONAL AND SCIENTIFIC CENTER "INSTITUTE OF BIOLOGY AND MEDICINE" OF TARAS SHEVCHENKO NATIONAL UNIVERSITY OF KYIV, KYIV, UKRAINE
| | | | - Victoriia Lavrynenko
- EDUCATIONAL AND SCIENTIFIC CENTER "INSTITUTE OF BIOLOGY AND MEDICINE" OF TARAS SHEVCHENKO NATIONAL UNIVERSITY OF KYIV, KYIV, UKRAINE
| |
Collapse
|
15
|
Riihimäki M, Fegraeus K, Nordlund J, Waern I, Wernersson S, Akula S, Hellman L, Raine A. Single-cell transcriptomics delineates the immune cell landscape in equine lower airways and reveals upregulation of FKBP5 in horses with asthma. Sci Rep 2023; 13:16261. [PMID: 37758813 PMCID: PMC10533524 DOI: 10.1038/s41598-023-43368-4] [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: 04/02/2023] [Accepted: 09/22/2023] [Indexed: 09/29/2023] Open
Abstract
Equine asthma (EA) is a heterogenous, complex disease, with a significant negative impact on horse welfare and performance. EA and human asthma share fundamental similarities, making EA a useful model for studying the disease. One relevant sample type for investigating chronic lung inflammation is bronchoalveolar lavage fluid (BALF), which provides a snapshot of the immune cells present in the alveolar space. To investigate the immune cell landscape of the respiratory tract in horses with mild-to-moderate equine asthma (mEA) and healthy controls, single-cell RNA sequencing was conducted on equine BALF cells. We characterized the major immune cell populations present in equine BALF, as well as subtypes thereof. Interestingly, the most significantly upregulated gene discovered in cases of mEA was FKBP5, a chaperone protein involved in regulating the activity of the glucocorticoid receptor.
Collapse
Affiliation(s)
- Miia Riihimäki
- Department of Clinical Sciences, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - Kim Fegraeus
- Department of Medical Sciences, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Jessica Nordlund
- Department of Medical Sciences, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Ida Waern
- Department of Anatomy, Physiology and Biochemistry, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - Sara Wernersson
- Department of Anatomy, Physiology and Biochemistry, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - Srinivas Akula
- Department of Anatomy, Physiology and Biochemistry, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - Lars Hellman
- Department of Cell and Molecular Biology, Uppsala University, Uppsala, Sweden
| | - Amanda Raine
- Department of Medical Sciences, Science for Life Laboratory, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
16
|
Diupotex M, Zamora-Chimal J, Gajón JA, Bonifaz LC, Becker I. CXCR5 and TIM-3 expressions define distinct exhausted T cell subsets in experimental cutaneous infection with Leishmania mexicana. Front Immunol 2023; 14:1231836. [PMID: 37691941 PMCID: PMC10485697 DOI: 10.3389/fimmu.2023.1231836] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 08/08/2023] [Indexed: 09/12/2023] Open
Abstract
T-cell exhaustion is a key stage in chronic infections since it limits immunopathology, but also hinders the elimination of pathogens. Exhausted T (Tex) cells encompass dynamic subsets, including progenitor cells that sustain long-term immunity through their memory/stem like properties, and terminally-differentiated cells, resembling the so-called Tex cells. The presence of Tex cells in chronic leishmaniasis has been reported in humans and murine models, yet their heterogeneity remains unexplored. Using flow cytometry, we identified Tex cells subtypes based on PD-1, CXCR5 and TIM-3 expressions in draining lymph nodes (dLNs) and lesion sites of C57BL/6 mice infected with L. mexicana at 30-, 60- and 90-days post-infection. We showed that infected mice developed a chronic infection characterized by non-healing lesions with a high parasite load and impaired Th1/Th2 cytokine production. Throughout the infection, PD-1+ cells were observed in dLNs, in addition to an enhanced expression of PD-1 in both CD4+ and CD8+ T lymphocytes. We demonstrated that CD4+ and CD8+ T cells were subdivided into PD-1+CXCR5+TIM-3- (CXCR5+), PD-1+CXCR5+TIM-3+ (CXCR5+TIM-3+), and PD-1+CXCR5-TIM-3+ (TIM-3+) subsets. CXCR5+ Tex cells were detected in dLNs during the whole course of the infection, whereas TIM-3+ cells were predominantly localized in the infection sites at day 90. CXCR5+TIM-3+ cells only increased at 30 and 60 days of infection in dLNs, whereas no increase was observed in the lesions. Phenotypic analysis revealed that CXCR5+ cells expressed significantly higher levels of CCR7 and lower levels of CX3CR1, PD-1, TIM-3, and CD39 compared to the TIM-3+ subset. CXCR5+TIM-3+ cells expressed the highest levels of all exhaustion-associated markers and of CX3CR1. In agreement with a less exhausted phenotype, the frequency of proliferating Ki-67 and IFN-γ expressing cells was significantly higher in the CXCR5+ subset within both CD4+ and CD8+ T cells compared to their respective TIM-3+ subsets, whereas CD8+CXCR5+TIM-3+ and CD8+TIM-3+ subsets showed an enhanced frequency of degranulating CD107a+ cells. In summary, we identified a novel, less-differentiated CXCR5+ Tex subset in experimental cutaneous leishmaniasis caused by L. mexicana. Targeting these cells through immune checkpoint inhibitors such as anti-PD-1 or anti PD-L1 might improve the current treatment for patients with the chronic forms of leishmaniasis.
Collapse
Affiliation(s)
- Mariana Diupotex
- Unidad de Investigación en Medicina Experimental, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Jaime Zamora-Chimal
- Unidad de Investigación en Medicina Experimental, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Julián A. Gajón
- Unidad de Investigación Médica en Inmunoquímica, Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Ciudad de México, Mexico
| | - Laura C. Bonifaz
- Unidad de Investigación Médica en Inmunoquímica, Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Ciudad de México, Mexico
- Coordinación de Investigación en Salud, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Ciudad de México, Mexico
| | - Ingeborg Becker
- Unidad de Investigación en Medicina Experimental, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| |
Collapse
|
17
|
Gillette JS, Wang EJ, Dowd RS, Toms SA. Barriers to overcoming immunotherapy resistance in glioblastoma. Front Med (Lausanne) 2023; 10:1175507. [PMID: 37275361 PMCID: PMC10232794 DOI: 10.3389/fmed.2023.1175507] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 04/12/2023] [Indexed: 06/07/2023] Open
Abstract
Glioblastoma multiforme (GBM) is the most common malignant primary brain tumor, known for its poor prognosis and high recurrence rate. Current standard of care includes surgical resection followed by combined radiotherapy and chemotherapy. Although immunotherapies have yielded promising results in hematological malignancies, their successful application in GBM remains limited due to a host of immunosuppressive factors unique to GBM. As a result of these roadblocks, research efforts have focused on utilizing combinatorial immunotherapies that target networks of immune processes in GBM with promising results in both preclinical and clinical trials, although limitations in overcoming the immunosuppressive factors within GBM remain. In this review, we aim to discuss the intrinsic and adaptive immune resistance unique to GBM and to summarize the current evidence and outcomes of engineered and non-engineered treatments targeted at overcoming GBM resistance to immunotherapy. Additionally, we aim to highlight the most promising strategies of targeted GBM immunotherapy combinatorial treatments and the insights that may directly improve the current patient prognosis and clinical care.
Collapse
|
18
|
Choi H, Kim Y, Jung YW. The Function of Memory CD8+ T Cells in Immunotherapy for Human Diseases. Immune Netw 2023; 23:e10. [PMID: 36911798 PMCID: PMC9995995 DOI: 10.4110/in.2023.23.e10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 02/13/2023] [Accepted: 02/13/2023] [Indexed: 03/07/2023] Open
Abstract
Memory T (Tm) cells protect against Ags that they have previously contacted with a fast and robust response. Therefore, developing long-lived Tm cells is a prime goal for many vaccines and therapies to treat human diseases. The remarkable characteristics of Tm cells have led scientists and clinicians to devise methods to make Tm cells more useful. Recently, Tm cells have been highlighted for their role in coronavirus disease 2019 vaccines during the ongoing global pandemic. The importance of Tm cells in cancer has been emerging. However, the precise characteristics and functions of Tm cells in these diseases are not completely understood. In this review, we summarize the known characteristics of Tm cells and their implications in the development of vaccines and immunotherapies for human diseases. In addition, we propose to exploit the beneficial characteristics of Tm cells to develop strategies for effective vaccines and overcome the obstacles of immunotherapy.
Collapse
Affiliation(s)
- Hanbyeul Choi
- Department of Pharmacy, Korea University, Sejong 30019, Korea
| | - Yeaji Kim
- Department of Pharmacy, Korea University, Sejong 30019, Korea
| | - Yong Woo Jung
- Department of Pharmacy, Korea University, Sejong 30019, Korea
| |
Collapse
|
19
|
Wan S, Zhao E, Freeman D, Weissinger D, Krantz BA, Werba G, Khanna LG, Siolas D, Oberstein PE, Chattopadhyay PK, Simeone DM, Welling TH. Tumor infiltrating T cell states and checkpoint inhibitor expression in hepatic and pancreatic malignancies. Front Immunol 2023; 14:1067352. [PMID: 36798126 PMCID: PMC9927010 DOI: 10.3389/fimmu.2023.1067352] [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: 10/11/2022] [Accepted: 01/18/2023] [Indexed: 02/04/2023] Open
Abstract
Hepato-pancreatico-biliary (HPB) malignancies are difficult-to-treat and continue to to have a high mortality and significant therapeutic resistance to standard therapies. Immune oncology (IO) therapies have demonstrated efficacy in several solid malignancies when combined with chemotherapy, whereas response rates in pancreatic ductal adenocarcinoma (PDA) are poor. While promising in hepatocellular carcinoma (HCC) and cholangiocarcinoma (CCA), there remains an unmet need to fully leverage IO therapies to treat HPB tumors. We therefore defined T cell subsets in the tumor microenvironment of HPB patients utilizing a novel, multiparameter flow cytometry and bioinformatics analysis. Our findings quantify the T cell phenotypic states in relation to checkpoint receptor expression. We demonstrate the presence of CD103+ tissue resident memory T cells (TRM), CCR7+ central memory T cells, and CD57+ terminally differentiated effector cells across all HPB cancers, while the anti-tumor function was dampened by expression of multiple co-inhibitory checkpoint receptors. Terminally exhausted T cells lacking co-stimulatory receptors were more prevalent in PDA, whereas partially exhausted T cells expressing both co-inhibitory and co-stimulatory receptors were most prevalent in HCC, especially in early stage. HCC patients had significantly higher TRM with a phenotype that could confer restored activation in response to immune checkpoint therapies. Further, we found a lack of robust alteration in T cell activation state or checkpoint expression in response to chemotherapy in PDA patients. These results support that HCC patients might benefit most from combined checkpoint therapies, whereas efforts other than cytotoxic chemotherapy will likely be necessary to increase overall T cell activation in CCA and PDA for future clinical development.
Collapse
Affiliation(s)
- Shanshan Wan
- Department of Surgery, NYU Langone Health, New York, NY, United States
| | - Ende Zhao
- Department of Surgery, NYU Langone Health, New York, NY, United States
- Perlmutter Cancer Center, NYU Langone Health, New York, NY, United States
| | - Daniel Freeman
- Pathology, NYU Langone Health, New York, NY, United States
| | - Daniel Weissinger
- Perlmutter Cancer Center, NYU Langone Health, New York, NY, United States
| | - Benjamin A. Krantz
- Perlmutter Cancer Center, NYU Langone Health, New York, NY, United States
- Internal Medicine, NYU Langone Health, New York, NY, United States
| | - Gregor Werba
- Perlmutter Cancer Center, NYU Langone Health, New York, NY, United States
| | - Lauren G. Khanna
- Internal Medicine, NYU Langone Health, New York, NY, United States
| | - Despina Siolas
- Perlmutter Cancer Center, NYU Langone Health, New York, NY, United States
- Internal Medicine, NYU Langone Health, New York, NY, United States
| | - Paul E. Oberstein
- Perlmutter Cancer Center, NYU Langone Health, New York, NY, United States
- Internal Medicine, NYU Langone Health, New York, NY, United States
| | - Pratip K. Chattopadhyay
- Perlmutter Cancer Center, NYU Langone Health, New York, NY, United States
- Pathology, NYU Langone Health, New York, NY, United States
- Talon Biomarkers, Mendham, NJ, United States
| | - Diane M. Simeone
- Department of Surgery, NYU Langone Health, New York, NY, United States
- Perlmutter Cancer Center, NYU Langone Health, New York, NY, United States
- Pathology, NYU Langone Health, New York, NY, United States
| | - Theodore H. Welling
- Department of Surgery, NYU Langone Health, New York, NY, United States
- Perlmutter Cancer Center, NYU Langone Health, New York, NY, United States
| |
Collapse
|
20
|
Co-expression of a PD-L1-specific chimeric switch receptor augments the efficacy and persistence of CAR T cells via the CD70-CD27 axis. Nat Commun 2022; 13:6051. [PMID: 36229619 PMCID: PMC9561169 DOI: 10.1038/s41467-022-33793-w] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 09/29/2022] [Indexed: 12/24/2022] Open
Abstract
Co-expression of chimeric switch receptors (CSRs) specific for PD-L1 improves the antitumor effects of chimeric antigen receptor (CAR) T cells. However, the effects of trans-recognition between CSRs and PD-L1 expressed by activated CAR T cells remain unclear. Here, we design a CSR specific for PD-L1 (CARP), containing the transmembrane and cytoplasmic signaling domains of CD28 but not the CD3 ζ chain. We show that CARP T cells enhance the antitumor activity of anti-mesothelin CAR (CARMz) T cells in vitro and in vivo. In addition, confocal microscopy indicates that PD-L1 molecules on CARMz T cells accumulate at cell-cell contacts with CARP T cells. Using single-cell RNA-sequencing analysis, we reveal that CARP T cells promote CARMz T cells differentiation into central memory-like T cells, upregulate genes related to Th1 cells, and downregulate Th2-associated cytokines through the CD70-CD27 axis. Moreover, these effects are not restricted to PD-L1, as CAR19 T cells expressing anti-CD19 CSR exhibit similar effects on anti-PSCA CAR T cells with truncated CD19 expression. These findings suggest that target trans-recognition by CSRs on CAR T cells may improve the efficacy and persistence of CAR T cells via the CD70-CD27 axis.
Collapse
|
21
|
Altendorfer B, Unger MS, Poupardin R, Hoog A, Asslaber D, Gratz IK, Mrowetz H, Benedetti A, de Sousa DMB, Greil R, Egle A, Gate D, Wyss-Coray T, Aigner L. Transcriptomic Profiling Identifies CD8 + T Cells in the Brain of Aged and Alzheimer's Disease Transgenic Mice as Tissue-Resident Memory T Cells. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 209:1272-1285. [PMID: 36165202 PMCID: PMC9515311 DOI: 10.4049/jimmunol.2100737] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 07/20/2022] [Indexed: 12/13/2022]
Abstract
Peripheral immune cell infiltration into the brain is a prominent feature in aging and various neurodegenerative diseases such as Alzheimer's disease (AD). As AD progresses, CD8+ T cells infiltrate into the brain parenchyma, where they tightly associate with neurons and microglia. The functional properties of CD8+ T cells in the brain are largely unknown. To gain further insights into the putative functions of CD8+ T cells in the brain, we explored and compared the transcriptomic profile of CD8+ T cells isolated from the brain and blood of transgenic AD (APPswe/PSEN1dE9, line 85 [APP-PS1]) and age-matched wild-type (WT) mice. Brain CD8+ T cells of APP-PS1 and WT animals had similar transcriptomic profiles and substantially differed from blood circulating CD8+ T cells. The gene signature of brain CD8+ T cells identified them as tissue-resident memory (Trm) T cells. Gene Ontology enrichment and Kyoto Encyclopedia of Genes and Genomes pathway analysis on the significantly upregulated genes revealed overrepresentation of biological processes involved in IFN-β signaling and the response to viral infections. Furthermore, brain CD8+ T cells of APP-PS1 and aged WT mice showed similar differentially regulated genes as brain Trm CD8+ T cells in mouse models with acute virus infection, chronic parasite infection, and tumor growth. In conclusion, our profiling of brain CD8+ T cells suggests that in AD, these cells exhibit similar adaptive immune responses as in other inflammatory diseases of the CNS, potentially opening the door for immunotherapy in AD.
Collapse
Affiliation(s)
- Barbara Altendorfer
- Institute of Molecular Regenerative Medicine, Paracelsus Medical University, Salzburg, Austria
- Spinal Cord Injury and Tissue Regeneration Center Salzburg, Paracelsus Medical University, Salzburg, Austria
| | - Michael Stefan Unger
- Institute of Molecular Regenerative Medicine, Paracelsus Medical University, Salzburg, Austria
- Spinal Cord Injury and Tissue Regeneration Center Salzburg, Paracelsus Medical University, Salzburg, Austria
| | - Rodolphe Poupardin
- Spinal Cord Injury and Tissue Regeneration Center Salzburg, Paracelsus Medical University, Salzburg, Austria
- Experimental and Clinical Cell Therapy Institute, Paracelsus Medical University, Salzburg, Austria
| | - Anna Hoog
- Spinal Cord Injury and Tissue Regeneration Center Salzburg, Paracelsus Medical University, Salzburg, Austria
- Experimental and Clinical Cell Therapy Institute, Paracelsus Medical University, Salzburg, Austria
| | - Daniela Asslaber
- IIIrd Medical Department with Hematology and Medical Oncology, Oncologic Center, Paracelsus Medical University, Salzburg, Austria
- Salzburg Cancer Research Institute with Laboratory of Immunological and Molecular Cancer Research and Center for Clinical Cancer and Immunology Trials, Salzburg, Austria
- Cancer Cluster Salzburg, Salzburg, Austria
| | - Iris Karina Gratz
- Department of Biosciences, University of Salzburg, Salzburg, Austria
| | - Heike Mrowetz
- Institute of Molecular Regenerative Medicine, Paracelsus Medical University, Salzburg, Austria
- Spinal Cord Injury and Tissue Regeneration Center Salzburg, Paracelsus Medical University, Salzburg, Austria
| | - Ariane Benedetti
- Spinal Cord Injury and Tissue Regeneration Center Salzburg, Paracelsus Medical University, Salzburg, Austria
- Institute of Experimental Neuroregeneration, Paracelsus Medical University, Salzburg, Austria
| | - Diana Marisa Bessa de Sousa
- Institute of Molecular Regenerative Medicine, Paracelsus Medical University, Salzburg, Austria
- Spinal Cord Injury and Tissue Regeneration Center Salzburg, Paracelsus Medical University, Salzburg, Austria
| | - Richard Greil
- IIIrd Medical Department with Hematology and Medical Oncology, Oncologic Center, Paracelsus Medical University, Salzburg, Austria
- Salzburg Cancer Research Institute with Laboratory of Immunological and Molecular Cancer Research and Center for Clinical Cancer and Immunology Trials, Salzburg, Austria
- Cancer Cluster Salzburg, Salzburg, Austria
| | - Alexander Egle
- IIIrd Medical Department with Hematology and Medical Oncology, Oncologic Center, Paracelsus Medical University, Salzburg, Austria
- Salzburg Cancer Research Institute with Laboratory of Immunological and Molecular Cancer Research and Center for Clinical Cancer and Immunology Trials, Salzburg, Austria
- Cancer Cluster Salzburg, Salzburg, Austria
| | - David Gate
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA
- Veterans Administration Palo Alto Healthcare System, Palo Alto, CA; and
| | - Tony Wyss-Coray
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA
- Veterans Administration Palo Alto Healthcare System, Palo Alto, CA; and
| | - Ludwig Aigner
- Institute of Molecular Regenerative Medicine, Paracelsus Medical University, Salzburg, Austria;
- Spinal Cord Injury and Tissue Regeneration Center Salzburg, Paracelsus Medical University, Salzburg, Austria
- Austrian Cluster for Tissue Regeneration, Vienna, Austria
| |
Collapse
|
22
|
The Memory T Cell “Communication Web” in Context with Gastrointestinal Disorders—How Memory T Cells Affect Their Surroundings and How They Are Influenced by It. Cells 2022; 11:cells11182780. [PMID: 36139354 PMCID: PMC9497182 DOI: 10.3390/cells11182780] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 08/30/2022] [Accepted: 09/03/2022] [Indexed: 11/17/2022] Open
Abstract
Gut-related diseases like ulcerative colitis, Crohn’s disease, or colorectal cancer affect millions of people worldwide. It is an ongoing process finding causes leading to the development and manifestation of those disorders. This is highly relevant since understanding molecular processes and signalling pathways offers new opportunities in finding novel ways to interfere with and apply new pharmaceuticals. Memory T cells (mT cells) and their pro-inflammatory properties have been proven to play an important role in gastrointestinal diseases and are therefore increasingly spotlighted. This review focuses on mT cells and their subsets in the context of disease pathogenesis and maintenance. It illustrates the network of regulatory proteins and metabolites connecting mT cells with other cell types and tissue compartments. Furthermore, the crosstalk with various microbes will be a subject of discussion. Characterizing mT cell interactions will help to further elucidate the sophisticated molecular and cellular networking system in the intestine and may present new ideas for future research approaches to control gut-related diseases.
Collapse
|
23
|
Zhou WH, Wang Y, Yan C, Du WD, Al-Aroomi MA, Zheng L, Lin SF, Gao JX, Jiang S, Wang ZX, Sun CF, Liu FY. CC chemokine receptor 7 promotes macrophage recruitment and induces M2-polarization through CC chemokine ligand 19&21 in oral squamous cell carcinoma. Discov Oncol 2022; 13:67. [PMID: 35904690 PMCID: PMC9338204 DOI: 10.1007/s12672-022-00533-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 07/13/2022] [Indexed: 12/14/2022] Open
Abstract
PURPOSE This study aimed to investigate the impact of CC chemokine receptor 7 (CCR7) on the recruitment and polarization of tumor-associated macrophages (TAMs) in oral squamous cell carcinoma (OSCC). METHODS We analyzed CCR7 expression pattern, clinicopathological significance, and its association with M2 macrophage infiltration in OSCC by bioinformatic methods. Small interfering RNA (siRNA) was utilized to silence CCR7 in OSCC cells. Conditioned media (CM) was harvested from transfected OSCC cells to establish a co-culture model of THP-1 derived macrophages and OSCC cells. Transwell assay and cell adhesion assay were performed to examine the effect of CCR7 on macrophages recruitment and adhesion. Cytoskeleton was labelled by phalloidin to observe macrophage morphological changes. Moreover, phenotypic alteration of macrophages was measured using quantitative real-time PCR (qRT-PCR), flow cytometry, and immunofluorescence (IF) staining. Ultimately, recombinant human CCL19 and CCL21 were added into the medium of THP-1 derived macrophages to explore their effects on polarization in vitro. RESULTS In OSCC patients, the overexpression of CCR7 positively correlated with lymph node metastasis and M2 macrophage infiltration. Macrophage not only exhibited enhanced migration, invasion and adhesion abilities, but also appeared more spindle and branched in vitro when treated with CM from OSCC cells. However, these phenomena were abrogated with knockdown of CCR7. We also discovered that inhibition of CCR7 in OSCC cells suppressed TAMs polarization to an M2 phenotype. In addition, recombinant human CCL19 and CCL21 promoted macrophage M2-polarization in vitro. CONCLUSION CCR7 in OSCC cells promoted recruitment and M2-polarization of THP-1 derived macrophages in vitro by regulating production of CCL19 and CCL21.
Collapse
Affiliation(s)
- Wan-Hang Zhou
- Department of Oral Maxillofacial-Head and Neck Surgery, School of Stomatology, China Medical University, Oral Diseases Laboratory of Liaoning, 117 Nanjing North Road, Heping District, Shenyang, 110000, Liaoning, China
| | - Yao Wang
- Department of Oral Maxillofacial-Head and Neck Surgery, School of Stomatology, China Medical University, Oral Diseases Laboratory of Liaoning, 117 Nanjing North Road, Heping District, Shenyang, 110000, Liaoning, China
| | - Cong Yan
- Department of Oral Maxillofacial-Head and Neck Surgery, School of Stomatology, China Medical University, Oral Diseases Laboratory of Liaoning, 117 Nanjing North Road, Heping District, Shenyang, 110000, Liaoning, China
| | - Wei-Dong Du
- Department of Oral Maxillofacial-Head and Neck Surgery, School of Stomatology, China Medical University, Oral Diseases Laboratory of Liaoning, 117 Nanjing North Road, Heping District, Shenyang, 110000, Liaoning, China
| | - Maged Ali Al-Aroomi
- Department of Oral Maxillofacial-Head and Neck Surgery, School of Stomatology, China Medical University, Oral Diseases Laboratory of Liaoning, 117 Nanjing North Road, Heping District, Shenyang, 110000, Liaoning, China
| | - Li Zheng
- Department of Oral Maxillofacial-Head and Neck Surgery, School of Stomatology, China Medical University, Oral Diseases Laboratory of Liaoning, 117 Nanjing North Road, Heping District, Shenyang, 110000, Liaoning, China
| | - Shan-Feng Lin
- Department of Oral Maxillofacial-Head and Neck Surgery, School of Stomatology, China Medical University, Oral Diseases Laboratory of Liaoning, 117 Nanjing North Road, Heping District, Shenyang, 110000, Liaoning, China
| | - Jia-Xing Gao
- Department of Oral Maxillofacial-Head and Neck Surgery, School of Stomatology, China Medical University, Oral Diseases Laboratory of Liaoning, 117 Nanjing North Road, Heping District, Shenyang, 110000, Liaoning, China
| | - Sheng Jiang
- Department of Oral Maxillofacial-Head and Neck Surgery, School of Stomatology, China Medical University, Oral Diseases Laboratory of Liaoning, 117 Nanjing North Road, Heping District, Shenyang, 110000, Liaoning, China
| | - Zeng-Xu Wang
- Department of Oral Maxillofacial-Head and Neck Surgery, School of Stomatology, China Medical University, Oral Diseases Laboratory of Liaoning, 117 Nanjing North Road, Heping District, Shenyang, 110000, Liaoning, China
| | - Chang-Fu Sun
- Department of Oral Maxillofacial-Head and Neck Surgery, School of Stomatology, China Medical University, Oral Diseases Laboratory of Liaoning, 117 Nanjing North Road, Heping District, Shenyang, 110000, Liaoning, China
| | - Fa-Yu Liu
- Department of Oral Maxillofacial-Head and Neck Surgery, School of Stomatology, China Medical University, Oral Diseases Laboratory of Liaoning, 117 Nanjing North Road, Heping District, Shenyang, 110000, Liaoning, China.
| |
Collapse
|
24
|
Jang MS, Ismail NSB, Yu YG. Development of a human antibody that exhibits antagonistic activity toward CC chemokine receptor 7. Antib Ther 2022; 5:192-201. [PMID: 35967907 PMCID: PMC9372883 DOI: 10.1093/abt/tbac016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 07/08/2022] [Accepted: 07/12/2022] [Indexed: 11/13/2022] Open
Abstract
Abstract
Background
CC chemokine receptor 7 (CCR7) is a member of G-protein-coupled receptor family and mediates chemotactic migration of immune cells and different cancer cells induced via chemokine (C-C motif) ligand 19 (CCL19) or chemokine (C-C motif) ligand 21 (CCL21). Hence, the identification of blockade antibodies against CCR7 could lead to the development of therapeutics targeting metastatic cancer.
Methods
CCR7 was purified and stabilized in its active conformation, and antibodies specific to purified CCR7 were screened from the synthetic M13 phage library displaying humanized scFvs. The in vitro characterization of selected scFvs identified two scFvs that exhibited CCL19-competitive binding to CCR7. IgG4’s harboring selected scFv sequences were characterized for binding activity in CCR7+ cells, inhibitory activity toward CCR7-dependent cAMP attenuation, and the CCL19 or CCL21-dependent migration of CCR7+ cells.
Results
Antibodies specifically binding to purified CCR7 and CCR7+ cells were isolated and characterized. Two antibodies, IgG4(6RG11) and IgG4(72C7), showed ligand-dependent competitive binding to CCR7 with KD values of 40 nM and 50 nM, respectively. Particularly, IgG4(6RG11) showed antagonistic activity against CCR7, whereas both antibodies significantly blocked the ligand-induced migration and invasion activity of CCR7+ cancer cells.
Conclusions
Two antibody clones were successfully identified from a synthetic scFv-displaying phage library using purified recombinant CCR7 as an antigen. Antibodies specifically bound to the surface of CCR7+ cells and blocked CCR7+ cell migration. Particularly, 6RG11 showed antagonist activity against CCR7-dependent cAMP attenuation.
Statement of Significance
Antibodies targeting CCR7 were identified and could serve as therapeutic reagents against cancer metastasis.
Collapse
Affiliation(s)
- Moon-Sung Jang
- Department of Biopharmaceutical Chemistry , Kookmin University, Seoul, 02707 , Republic of Korea
- Antibody Research Institute , Kookmin University, Seoul 02707 , Republic of Korea
| | - Nurain Syahirah Binti Ismail
- Department of Biopharmaceutical Chemistry , Kookmin University, Seoul, 02707 , Republic of Korea
- Antibody Research Institute , Kookmin University, Seoul 02707 , Republic of Korea
| | - Yeon Gyu Yu
- Department of Biopharmaceutical Chemistry , Kookmin University, Seoul, 02707 , Republic of Korea
- Antibody Research Institute , Kookmin University, Seoul 02707 , Republic of Korea
| |
Collapse
|
25
|
Damian L, Login CC, Solomon C, Belizna C, Encica S, Urian L, Jurcut C, Stancu B, Vulturar R. Inclusion Body Myositis and Neoplasia: A Narrative Review. Int J Mol Sci 2022; 23:ijms23137358. [PMID: 35806366 PMCID: PMC9266341 DOI: 10.3390/ijms23137358] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 06/28/2022] [Accepted: 06/28/2022] [Indexed: 02/04/2023] Open
Abstract
Inclusion body myositis (IBM) is an acquired, late-onset inflammatory myopathy, with both inflammatory and degenerative pathogenesis. Although idiopathic inflammatory myopathies may be associated with malignancies, IBM is generally not considered paraneoplastic. Many studies of malignancy in inflammatory myopathies did not include IBM patients. Indeed, IBM is often diagnosed only after around 5 years from onset, while paraneoplastic myositis is generally defined as the co-occurrence of malignancy and myopathy within 1 to 3 years of each other. Nevertheless, a significant association with large granular lymphocyte leukemia has been recently described in IBM, and there are reports of cancer-associated IBM. We review the pathogenic mechanisms supposed to be involved in IBM and outline the common mechanisms in IBM and malignancy, as well as the therapeutic perspectives. The terminally differentiated, CD8+ highly cytotoxic T cells expressing NK features are central in the pathogenesis of IBM and, paradoxically, play a role in some cancers as well. Interferon gamma plays a central role, mostly during the early stages of the disease. The secondary mitochondrial dysfunction, the autophagy and cell cycle dysregulation, and the crosstalk between metabolic and mitogenic pathways could be shared by IBM and cancer. There are intermingled subcellular mechanisms in IBM and neoplasia, and probably their co-existence is underestimated. The link between IBM and cancers deserves further interest, in order to search for efficient therapies in IBM and to improve muscle function, life quality, and survival in both diseases.
Collapse
Affiliation(s)
- Laura Damian
- Centre for Rare Autoimmune and Autoinflammatory Diseases (ERN-ReCONNET), Department of Rheumatology, Emergency Clinical County Hospital Cluj, 400347 Cluj-Napoca, Romania;
- CMI Reumatologie Dr. Damian, 6-8 Petru Maior St., 400002 Cluj-Napoca, Romania
| | - Cristian Cezar Login
- Department of Physiology, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400006 Cluj-Napoca, Romania
- Correspondence:
| | - Carolina Solomon
- Radiology Department, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400006 Cluj-Napoca, Romania;
- Radiology Department, Emergency Clinical County Hospital Cluj, 400006 Cluj-Napoca, Romania
| | - Cristina Belizna
- UMR CNRS 6015—INSERM U1083, University of Angers, 49100 Angers, France;
- Internal Medicine Department Clinique de l’Anjou, Angers and Vascular and Coagulation Department, University Hospital Angers, 49100 Angers, France
| | - Svetlana Encica
- Department of Pathology, “Niculae Stancioiu” Heart Institute Cluj-Napoca, 19-21 Calea Moților St., 400001 Cluj-Napoca, Romania;
| | - Laura Urian
- Department of Hematology, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400004 Cluj-Napoca, Romania;
- Department of Hematology, Ion Chiricuta Clinical Cancer Center, 400014 Cluj-Napoca, Romania
| | - Ciprian Jurcut
- Department of Internal Medicine, “Carol Davila” Central Military Emergency University Hospital, Calea Plevnei No 134, 010825 Bucharest, Romania;
| | - Bogdan Stancu
- 2nd Surgical Department, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania;
| | - Romana Vulturar
- Department of Molecular Sciences, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania;
- Cognitive Neuroscience Laboratory, University “Babes-Bolyai” Cluj-Napoca, 400294 Cluj-Napoca, Romania
| |
Collapse
|
26
|
Skurikhin EG, Pershina O, Ermakova N, Pakhomova A, Widera D, Zhukova M, Pan E, Sandrikina L, Kogai L, Kushlinskii N, Morozov SG, Kubatiev A, Dygai A. Reprogrammed CD8 + T-Lymphocytes Isolated from Bone Marrow Have Anticancer Potential in Lung Cancer. Biomedicines 2022; 10:biomedicines10061450. [PMID: 35740471 PMCID: PMC9219954 DOI: 10.3390/biomedicines10061450] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 06/08/2022] [Accepted: 06/17/2022] [Indexed: 12/16/2022] Open
Abstract
CD8+ T-lymphocytes play a key role in antitumor immune response. Patients with lung cancer often suffer from T-lymphocyte dysfunction and low T-cell counts. The exhaustion of effector T-lymphocytes largely limits the effectiveness of therapy. In this study, reprogrammed T-lymphocytes used MEK inhibitors and PD-1 blockers to increase their antitumor activity. Antitumor effects of reprogrammed T-lymphocytes were shown in vitro and in vivo in the Lewis lung carcinoma model. The population of T- lymphocytes with persistent expression of CCR7 was formed as a result of reprogramming. Reprogrammed T-lymphocytes were resistant to apoptosis and characterized by high cytotoxicity against Lewis lung carcinoma (LLC) cells in vitro. Administration of reprogrammed T-lymphocytes to C57BL/6 mice with LLC reduced the number of lung metastases. The antitumor effect resulted from the elimination of tumor cells and cancer stem cells, and the effect of therapy on cytotoxic T-lymphocyte counts. Thus, reprogramming of T-lymphocytes using MEK inhibitors is a promising approach for targeted therapy of lung cancer.
Collapse
Affiliation(s)
- Evgenii G. Skurikhin
- Laboratory of Regenerative Pharmacology, Goldberg ED Research Institute of Pharmacology and Regenerative Medicine, Tomsk National Research Medical Centre of the Russian Academy of Sciences, Lenin, 3, 634028 Tomsk, Russia; (O.P.); (N.E.); (A.P.); (M.Z.); (E.P.); (L.S.); (L.K.); (A.D.)
- Correspondence: ; Tel.: +7-3822-418-375
| | - Olga Pershina
- Laboratory of Regenerative Pharmacology, Goldberg ED Research Institute of Pharmacology and Regenerative Medicine, Tomsk National Research Medical Centre of the Russian Academy of Sciences, Lenin, 3, 634028 Tomsk, Russia; (O.P.); (N.E.); (A.P.); (M.Z.); (E.P.); (L.S.); (L.K.); (A.D.)
| | - Natalia Ermakova
- Laboratory of Regenerative Pharmacology, Goldberg ED Research Institute of Pharmacology and Regenerative Medicine, Tomsk National Research Medical Centre of the Russian Academy of Sciences, Lenin, 3, 634028 Tomsk, Russia; (O.P.); (N.E.); (A.P.); (M.Z.); (E.P.); (L.S.); (L.K.); (A.D.)
| | - Angelina Pakhomova
- Laboratory of Regenerative Pharmacology, Goldberg ED Research Institute of Pharmacology and Regenerative Medicine, Tomsk National Research Medical Centre of the Russian Academy of Sciences, Lenin, 3, 634028 Tomsk, Russia; (O.P.); (N.E.); (A.P.); (M.Z.); (E.P.); (L.S.); (L.K.); (A.D.)
| | - Darius Widera
- Stem Cell Biology and Regenerative Medicine Group, School of Pharmacy, Whiteknights Campus, Reading RG6 6AP, UK;
| | - Mariia Zhukova
- Laboratory of Regenerative Pharmacology, Goldberg ED Research Institute of Pharmacology and Regenerative Medicine, Tomsk National Research Medical Centre of the Russian Academy of Sciences, Lenin, 3, 634028 Tomsk, Russia; (O.P.); (N.E.); (A.P.); (M.Z.); (E.P.); (L.S.); (L.K.); (A.D.)
| | - Edgar Pan
- Laboratory of Regenerative Pharmacology, Goldberg ED Research Institute of Pharmacology and Regenerative Medicine, Tomsk National Research Medical Centre of the Russian Academy of Sciences, Lenin, 3, 634028 Tomsk, Russia; (O.P.); (N.E.); (A.P.); (M.Z.); (E.P.); (L.S.); (L.K.); (A.D.)
| | - Lubov Sandrikina
- Laboratory of Regenerative Pharmacology, Goldberg ED Research Institute of Pharmacology and Regenerative Medicine, Tomsk National Research Medical Centre of the Russian Academy of Sciences, Lenin, 3, 634028 Tomsk, Russia; (O.P.); (N.E.); (A.P.); (M.Z.); (E.P.); (L.S.); (L.K.); (A.D.)
| | - Lena Kogai
- Laboratory of Regenerative Pharmacology, Goldberg ED Research Institute of Pharmacology and Regenerative Medicine, Tomsk National Research Medical Centre of the Russian Academy of Sciences, Lenin, 3, 634028 Tomsk, Russia; (O.P.); (N.E.); (A.P.); (M.Z.); (E.P.); (L.S.); (L.K.); (A.D.)
| | - Nikolai Kushlinskii
- Blokhin National Medical Research Center of Oncology, 115522 Moscow, Russia;
| | - Sergey G. Morozov
- Institute of General Pathology and Pathophysiology, 125315 Moscow, Russia; (S.G.M.); (A.K.)
| | - Aslan Kubatiev
- Institute of General Pathology and Pathophysiology, 125315 Moscow, Russia; (S.G.M.); (A.K.)
| | - Alexander Dygai
- Laboratory of Regenerative Pharmacology, Goldberg ED Research Institute of Pharmacology and Regenerative Medicine, Tomsk National Research Medical Centre of the Russian Academy of Sciences, Lenin, 3, 634028 Tomsk, Russia; (O.P.); (N.E.); (A.P.); (M.Z.); (E.P.); (L.S.); (L.K.); (A.D.)
- Institute of General Pathology and Pathophysiology, 125315 Moscow, Russia; (S.G.M.); (A.K.)
| |
Collapse
|
27
|
Liu Y, Wang H, Taylor M, Cook C, Martínez-Berdeja A, North JP, Harirchian P, Hailer AA, Zhao Z, Ghadially R, Ricardo-Gonzalez RR, Grekin RC, Mauro TM, Kim E, Choi J, Purdom E, Cho RJ, Cheng JB. Classification of human chronic inflammatory skin disease based on single-cell immune profiling. Sci Immunol 2022; 7:eabl9165. [PMID: 35427179 PMCID: PMC9301819 DOI: 10.1126/sciimmunol.abl9165] [Citation(s) in RCA: 71] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Inflammatory conditions represent the largest class of chronic skin disease, but the molecular dysregulation underlying many individual cases remains unclear. Single-cell RNA sequencing (scRNA-seq) has increased precision in dissecting the complex mixture of immune and stromal cell perturbations in inflammatory skin disease states. We single-cell-profiled CD45+ immune cell transcriptomes from skin samples of 31 patients (7 atopic dermatitis, 8 psoriasis vulgaris, 2 lichen planus (LP), 1 bullous pemphigoid (BP), 6 clinical/histopathologically indeterminate rashes, and 7 healthy controls). Our data revealed active proliferative expansion of the Treg and Trm components and universal T cell exhaustion in human rashes, with a relative attenuation of antigen-presenting cells. Skin-resident memory T cells showed the greatest transcriptional dysregulation in both atopic dermatitis and psoriasis, whereas atopic dermatitis also demonstrated recurrent abnormalities in ILC and CD8+ cytotoxic lymphocytes. Transcript signatures differentiating these rash types included genes previously implicated in T helper cell (TH2)/TH17 diatheses, segregated in unbiased functional networks, and accurately identified disease class in untrained validation data sets. These gene signatures were able to classify clinicopathologically ambiguous rashes with diagnoses consistent with therapeutic response. Thus, we have defined major classes of human inflammatory skin disease at the molecular level and described a quantitative method to classify indeterminate instances of pathologic inflammation. To make this approach accessible to the scientific community, we created a proof-of-principle web interface (RashX), where scientists and clinicians can visualize their patient-level rash scRNA-seq-derived data in the context of our TH2/TH17 transcriptional framework.
Collapse
Affiliation(s)
- Yale Liu
- Department of Dermatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, ShaanXi 710004, P. R. China
- Department of Dermatology, University of California, San Francisco, San Francisco, CA 94107, USA
- Dermatology, Veterans Affairs Medical Center, San Francisco, CA 94121, USA
| | - Hao Wang
- Department of Statistics, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Mark Taylor
- Department of Dermatology, University of California, San Francisco, San Francisco, CA 94107, USA
| | - Christopher Cook
- Department of Dermatology, University of California, San Francisco, San Francisco, CA 94107, USA
- Dermatology, Veterans Affairs Medical Center, San Francisco, CA 94121, USA
| | | | - Jeffrey P North
- Department of Dermatology, University of California, San Francisco, San Francisco, CA 94107, USA
| | - Paymann Harirchian
- Department of Dermatology, University of California, San Francisco, San Francisco, CA 94107, USA
- Dermatology, Veterans Affairs Medical Center, San Francisco, CA 94121, USA
| | - Ashley A Hailer
- Department of Dermatology, University of California, San Francisco, San Francisco, CA 94107, USA
- Dermatology, Veterans Affairs Medical Center, San Francisco, CA 94121, USA
| | - Zijun Zhao
- Santa Clara Valley Medical Center, Santa Clara, CA 95128, USA
| | - Ruby Ghadially
- Department of Dermatology, University of California, San Francisco, San Francisco, CA 94107, USA
- Dermatology, Veterans Affairs Medical Center, San Francisco, CA 94121, USA
| | - Roberto R Ricardo-Gonzalez
- Department of Dermatology, University of California, San Francisco, San Francisco, CA 94107, USA
- Department of Immunology and Microbiology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Roy C Grekin
- Department of Dermatology, University of California, San Francisco, San Francisco, CA 94107, USA
| | - Theodora M Mauro
- Department of Dermatology, University of California, San Francisco, San Francisco, CA 94107, USA
- Dermatology, Veterans Affairs Medical Center, San Francisco, CA 94121, USA
| | - Esther Kim
- Department of Plastic Surgery, University of California, San Francisco, San Francisco, CA 94107, USA
| | - Jaehyuk Choi
- Department of Dermatology, Northwestern School of Medicine, Chicago, IL 60611, USA
| | - Elizabeth Purdom
- Department of Statistics, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Raymond J Cho
- Department of Dermatology, University of California, San Francisco, San Francisco, CA 94107, USA
| | - Jeffrey B Cheng
- Department of Dermatology, University of California, San Francisco, San Francisco, CA 94107, USA
- Dermatology, Veterans Affairs Medical Center, San Francisco, CA 94121, USA
| |
Collapse
|
28
|
Xu J, Ren Z, Cao K, Li X, Yang J, Luo X, Zhu L, Wang X, Ding L, Liang J, Jin D, Yuan T, Li L, Xu J. Boosting Vaccine-Elicited Respiratory Mucosal and Systemic COVID-19 Immunity in Mice With the Oral Lactobacillus plantarum. Front Nutr 2022; 8:789242. [PMID: 35004816 PMCID: PMC8733898 DOI: 10.3389/fnut.2021.789242] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 11/16/2021] [Indexed: 12/21/2022] Open
Abstract
Boosting and prolonging SARS-CoV-2 vaccine-elicited immunity is paramount for containing the COVID-19 pandemic, which wanes substantially within months after vaccination. Here we demonstrate that the unique strain of probiotic Lactobacillus plantarum GUANKE (LPG) could promote SARS-CoV-2-specific immune responses in both effective and memory phases through enhancing interferon signaling and suppressing apoptotic and inflammatory pathways. Interestingly, oral LPG administration promoted SARS-CoV-2 neutralization antibodies even 6 months after immunization. Furthermore, when LPG was given immediately after SARS-CoV-2 vaccine inoculation, specific neutralization antibodies could be boosted >8-fold in bronchoalveolar lavage (BAL) and >2-fold in sera, T-cell responses were persistent and stable for a prolonged period both in BAL and the spleen. Transcriptional analyses showed that oral application of LPG mobilized immune responses in the mucosal and systemic compartments; in particular, gut-spleen and gut-lung immune axes were observed. These results suggest that LPG could be applied in combination with SARS-CoV-2 vaccines to boost and prolong both the effective and memory immune responses in mucosal and systemic compartments, thereby improving the efficacy of SARS-CoV-2 vaccination.
Collapse
Affiliation(s)
- Jianqing Xu
- Zhongshan Hospital, Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China.,Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Zhihong Ren
- Shanghai Public Health Clinical Center, Fudan University, Shanghai, China.,State Key Laboratory for Infectious Disease Prevention and Control, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Research Units of Discovery of Unknown Bacteria and Function (2018 RU010), Chinese Academy of Medical Sciences, Beijing, China
| | - Kangli Cao
- Zhongshan Hospital, Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xianping Li
- State Key Laboratory for Infectious Disease Prevention and Control, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Research Units of Discovery of Unknown Bacteria and Function (2018 RU010), Chinese Academy of Medical Sciences, Beijing, China
| | - Jing Yang
- State Key Laboratory for Infectious Disease Prevention and Control, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Research Units of Discovery of Unknown Bacteria and Function (2018 RU010), Chinese Academy of Medical Sciences, Beijing, China
| | - Xuelian Luo
- State Key Laboratory for Infectious Disease Prevention and Control, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Research Units of Discovery of Unknown Bacteria and Function (2018 RU010), Chinese Academy of Medical Sciences, Beijing, China
| | - Lingyan Zhu
- Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Xiangwei Wang
- Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Longfei Ding
- Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Junrong Liang
- State Key Laboratory for Infectious Disease Prevention and Control, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Research Units of Discovery of Unknown Bacteria and Function (2018 RU010), Chinese Academy of Medical Sciences, Beijing, China
| | - Dong Jin
- State Key Laboratory for Infectious Disease Prevention and Control, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Research Units of Discovery of Unknown Bacteria and Function (2018 RU010), Chinese Academy of Medical Sciences, Beijing, China
| | - Tingting Yuan
- Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Lianfeng Li
- Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Jianguo Xu
- State Key Laboratory for Infectious Disease Prevention and Control, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Research Units of Discovery of Unknown Bacteria and Function (2018 RU010), Chinese Academy of Medical Sciences, Beijing, China.,Institute of Public Health, Nankai University, Tianjing, China
| |
Collapse
|
29
|
Kim KH, Choi A, Kim SH, Song H, Jin S, Kim K, Jang J, Choi H, Jung YW. Neural-Cadherin Influences the Homing of Terminally Differentiated Memory CD8 T Cells to the Lymph Nodes and Bone Marrow. Mol Cells 2021; 44:795-804. [PMID: 34819396 PMCID: PMC8627834 DOI: 10.14348/molcells.2021.0137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Revised: 09/07/2021] [Accepted: 09/27/2021] [Indexed: 11/27/2022] Open
Abstract
Memory T (TM) cells play an important role in the long-term defense against pathogen reinvasion. However, it is still unclear how these cells receive the crucial signals necessary for their longevity and homeostatic turnover. To understand how TM cells receive these signals, we infected mice with lymphocytic choriomeningitis virus (LCMV) and examined the expression sites of neural cadherin (N-cadherin) by immunofluorescence microscopy. We found that N-cadherin was expressed in the surroundings of the white pulps of the spleen and medulla of lymph nodes (LNs). Moreover, TM cells expressing high levels of killer cell lectin-like receptor G1 (KLRG1), a ligand of N-cadherin, were co-localized with N-cadherin+ cells in the spleen but not in LNs. We then blocked N-cadherin in vivo to investigate whether it regulates the formation or function of TM cells. The numbers of CD127hiCD62Lhi TM cells in the spleen of memory P14 chimeric mice declined when N-cadherin was blocked during the contraction phase, without functional impairment of these cells. In addition, when CD127loKLRG1hi TM cells were adoptively transferred into anti-N-cadherin-treated mice compared with control mice, the number of these cells was reduced in the bone marrow and LNs, without functional loss. Taken together, our results suggest that N-cadherin participates in the development of CD127hiCD62Lhi TM cells and homing of CD127loKLRG1hi TM cells to lymphoid organs.
Collapse
Affiliation(s)
- Kyong Hoon Kim
- Department of Pharmacy, Korea University, Sejong 30019, Korea
| | - Aryeong Choi
- Department of Pharmacy, Korea University, Sejong 30019, Korea
| | - Sang Hoon Kim
- Department of Pharmacy, Korea University, Sejong 30019, Korea
| | - Heonju Song
- Department of Pharmacy, Korea University, Sejong 30019, Korea
| | - Seohoon Jin
- Department of Applied Statistics, Korea University, Sejong 30019, Korea
| | - Kyungim Kim
- Department of Pharmacy, Korea University, Sejong 30019, Korea
| | - Jaebong Jang
- Department of Pharmacy, Korea University, Sejong 30019, Korea
| | - Hanbyeul Choi
- Department of Pharmacy, Korea University, Sejong 30019, Korea
| | - Yong Woo Jung
- Department of Pharmacy, Korea University, Sejong 30019, Korea
| |
Collapse
|
30
|
Ren H, Cao K, Wang M. A Correlation Between Differentiation Phenotypes of Infused T Cells and Anti-Cancer Immunotherapy. Front Immunol 2021; 12:745109. [PMID: 34603332 PMCID: PMC8479103 DOI: 10.3389/fimmu.2021.745109] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 08/25/2021] [Indexed: 12/30/2022] Open
Abstract
T-cell therapy, usually with ex-vivo expansion, is very promising to treat cancer. Differentiation status of infused T cells is a crucial parameter for their persistence and antitumor immunity. Key phenotypic molecules are effective and efficient to analyze differentiation status. Differentiation status is crucial for T cell exhaustion, in-vivo lifespan, antitumor immunity, and even antitumor pharmacological interventions. Strategies including cytokines, Akt, Wnt and Notch signaling, epigenetics, and metabolites have been developed to produce less differentiated T cells. Clinical trials have shown better clinical outcomes from infusion of T cells with less differentiated phenotypes. CD27+, CCR7+ and CD62L+ have been the most clinically relevant phenotypic molecules, while Tscm and Tcm the most clinically relevant subtypes. Currently, CD27+, CD62L+ and CCR7+ are recommended in the differentiation phenotype to evaluate strategies of enhancing stemness. Future studies may discover highly clinically relevant differentiation phenotypes for specific T-cell production methods or specific subtypes of cancer patients, with the advantages of precision medicine.
Collapse
Affiliation(s)
- Hao Ren
- Department of Research and Development, Shenzhen Institute for Innovation and Translational Medicine, Shenzhen, China
| | - Kunkun Cao
- Department of Research and Development, Shenzhen Institute for Innovation and Translational Medicine, Shenzhen, China
| | - Mingjun Wang
- Department of Research and Development, Shenzhen Institute for Innovation and Translational Medicine, Shenzhen, China
| |
Collapse
|
31
|
Tian WJ, Feng PH, Wang J, Yan T, Qin QF, Li DL, Liang WT. CCR7 Has Potential to Be a Prognosis Marker for Cervical Squamous Cell Carcinoma and an Index for Tumor Microenvironment Change. Front Mol Biosci 2021; 8:583028. [PMID: 33869272 PMCID: PMC8047428 DOI: 10.3389/fmolb.2021.583028] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 03/05/2021] [Indexed: 12/30/2022] Open
Abstract
The tumor microenvironment (TME) has an essential role in the development of cervical squamous cell carcinoma (CSCC); however, the dynamic role of the stromal and immune cells is still unclear in TME. We downloaded data from The Cancer Genome Atlas (TCGA) database and applied ESTIMATE and CIBERSORT algorithms to measure the quantity of stromal and immune cells and the composition of tumor-infiltrating immune cell (TIC) in 253 CSCC cases. The protein-protein interaction (PPI) network and Cox regression analysis presented the differentially expressed genes (DEGs). Then, C-C chemokine receptor type 7 (CCR7) was screened out as a prognostic marker by the univariate Cox and intersection analysis of PPI. Further analysis showed a positive correlation between the expression of CCR7 and the survival of CSCC patients. The result of the Gene Set Enrichment Analysis (GSEA) of genes in the high CCR7 expression group displayed a predominant enrichment in immune-related pathways. An enrichment in metabolic activities was observed in the low CCR7 expression group. CIBERSORT analysis showed a positive correlation between Plasma cells, CD8+ T cells, and regulatory T cells and the CCR7 expression, suggesting that CCR7 might play a crucial role in maintaining the immunological dominance status for TME. Therefore, the expression level of CCR7 might help predict the survival of CSCC cases and be an index that the status of TME transitioned from immunological dominance to metabolic activation, which presented a new insight into the treatment of CSCC.
Collapse
Affiliation(s)
- Wei-Jie Tian
- Department of Gynecology, Guizhou Provincial People's Hospital, Medical College of Guizhou University, Guiyang, China
| | - Peng-Hui Feng
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Jun Wang
- Department of Gynecology, Guizhou Provincial People's Hospital, Medical College of Guizhou University, Guiyang, China
| | - Ting Yan
- Department of Gynecology, Guizhou Provincial People's Hospital, Medical College of Guizhou University, Guiyang, China
| | - Qing-Feng Qin
- Department of Gynecology, Guizhou Provincial People's Hospital, Medical College of Guizhou University, Guiyang, China
| | - Dong-Lin Li
- Department of Gynecology, Guizhou Provincial People's Hospital, Medical College of Guizhou University, Guiyang, China
| | - Wen-Tong Liang
- Department of Gynecology, Guizhou Provincial People's Hospital, Medical College of Guizhou University, Guiyang, China
| |
Collapse
|
32
|
Otvos B, Alban TJ, Grabowski MM, Bayik D, Mulkearns-Hubert EE, Radivoyevitch T, Rabljenovic A, Johnson S, Androjna C, Mohammadi AM, Barnett GH, Ahluwalia MS, Vogelbaum MA, Fecci PE, Lathia JD. Preclinical Modeling of Surgery and Steroid Therapy for Glioblastoma Reveals Changes in Immunophenotype that are Associated with Tumor Growth and Outcome. Clin Cancer Res 2021; 27:2038-2049. [PMID: 33542075 DOI: 10.1158/1078-0432.ccr-20-3262] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 12/08/2020] [Accepted: 02/02/2021] [Indexed: 11/16/2022]
Abstract
PURPOSE Glioblastoma (GBM) immunotherapy clinical trials are generally initiated after standard-of-care treatment-including surgical resection, perioperative high-dose steroid therapy, chemotherapy, and radiation treatment-has either begun or failed. However, the impact of these interventions on the antitumoral immune response is not well studied. While discoveries regarding the impact of chemotherapy and radiation on immune response have been made and translated into clinical trial design, the impact of surgical resection and steroids on the antitumor immune response has yet to be determined. EXPERIMENTAL DESIGN We developed a murine model integrating tumor resection and steroid treatment and used flow cytometry to analyze systemic and local immune changes. These mouse model findings were validated in a cohort of 95 patients with primary GBM. RESULTS Using our murine resection model, we observed a systemic reduction in lymphocytes corresponding to increased tumor volume and decreased circulating lymphocytes that was masked by dexamethasone treatment. The reduction in circulating T cells was due to reduced CCR7 expression, resulting in T-cell sequestration in lymphoid organs and the bone marrow. We confirmed these findings in a cohort of patients with primary GBM and found that prior to steroid treatment, circulating lymphocytes inversely correlated with tumor volume. Finally, we demonstrated that peripheral lymphocyte content varies with progression-free survival and overall survival, independent of tumor volume, steroid use, or molecular profiles. CONCLUSIONS These data reveal that prior to intervention, increased tumor volume corresponds with reduced systemic immune function and that peripheral lymphocyte counts are prognostic when steroid treatment is taken into account.
Collapse
Affiliation(s)
- Balint Otvos
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio.,Department of Neurosurgery, Cleveland Clinic, Cleveland, Ohio
| | - Tyler J Alban
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio.,Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio.,Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, Ohio
| | - Matthew M Grabowski
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio.,Department of Neurosurgery, Cleveland Clinic, Cleveland, Ohio
| | - Defne Bayik
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio.,Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio
| | - Erin E Mulkearns-Hubert
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
| | - Tomas Radivoyevitch
- Department of Quantitative Health Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
| | - Anja Rabljenovic
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
| | - Sarah Johnson
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
| | - Charlie Androjna
- Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
| | - Alireza M Mohammadi
- Department of Neurosurgery, Cleveland Clinic, Cleveland, Ohio.,Rose Ella Burkhardt Brain Tumor and Neuro-Oncology Center, Cleveland Clinic, Cleveland, Ohio.,Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, Ohio
| | - Gene H Barnett
- Department of Neurosurgery, Cleveland Clinic, Cleveland, Ohio.,Rose Ella Burkhardt Brain Tumor and Neuro-Oncology Center, Cleveland Clinic, Cleveland, Ohio.,Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, Ohio
| | - Manmeet S Ahluwalia
- Department of Neurosurgery, Cleveland Clinic, Cleveland, Ohio.,Rose Ella Burkhardt Brain Tumor and Neuro-Oncology Center, Cleveland Clinic, Cleveland, Ohio.,Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, Ohio
| | | | - Peter E Fecci
- Department of Neurosurgery, Duke University Hospital, Durham, North Carolina
| | - Justin D Lathia
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio. .,Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio.,Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, Ohio.,Rose Ella Burkhardt Brain Tumor and Neuro-Oncology Center, Cleveland Clinic, Cleveland, Ohio.,Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, Ohio
| |
Collapse
|