1
|
Flores-Gonzalez J, Ramón-Luing LA, Falfán-Valencia R, Batista CVF, Soto-Alvarez S, Huerta-Nuñez L, Chávez-Galán L. The presence of cytotoxic CD4 and exhausted-like CD8+ T-cells is a signature of active tuberculosis. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167219. [PMID: 38734321 DOI: 10.1016/j.bbadis.2024.167219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 04/22/2024] [Accepted: 05/02/2024] [Indexed: 05/13/2024]
Abstract
Chronic infections induce CD4+ T-cells with cytotoxic functions (CD4 CTLs); at present, it is still unknown whether latent tuberculosis (LTB) and active tuberculosis (ATB) induce CD4 CTLs. Plasma and cells from four patient groups-uninfected contact (UC), LTB, and ATB (divided as sensitive [DS-TB]- or resistant [DR-TB]-drug)-were evaluated by flow cytometry, q-PCR, and proteomics. The data showed that ATB patients had an increased frequency of CD4+ T-cells and a decreased frequency of CD8+ T-cells. The latter displays an exhausted-like profile characterized by CD39, CD279, and TIM-3 expression. ATB had a high frequency of CD4 + perforin+ cells, suggesting a CD4 CTL profile. The expression (at the transcriptional level) of granzyme A, granzyme B, granulysin, and perforin, as well as the genes T-bet (Tbx21) and NKG2D (Klrk1), in enriched CD4+ T-cells, confirmed the cytotoxic signature of CD4+ T-cells during ATB (which was stronger in DS-TB than in DR-TB). Moreover, proteomic analysis revealed the presence of HSP70 (in DS-TB) and annexin A5 (in DR-TB), which are molecules that have been associated with favoring the CD4 CTL profile. Finally, we found that lipids from Mycobacterium tuberculosis increased the presence of CD4 CTLs in DR-TB patients. Our data suggest that ATB is characterized by exhausted-like CD8+ T-cells, which, together with a specific microenvironment, favor the presence of CD4 CTLs.
Collapse
Affiliation(s)
- Julio Flores-Gonzalez
- Laboratory of Integrative Immunology, Instituto Nacional de Enfermedades Respiratorias Ismael Cosio Villegas, Mexico City 14080. Mexico
| | - Lucero A Ramón-Luing
- Laboratory of Integrative Immunology, Instituto Nacional de Enfermedades Respiratorias Ismael Cosio Villegas, Mexico City 14080. Mexico
| | - Ramcés Falfán-Valencia
- HLA Laboratory, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City 14080, Mexico
| | - Cesar V F Batista
- Laboratory of Pharmacology, Escuela Militar de Graduados de Sanidad, Universidad del Ejército y Fuerza Aérea Mexicana, Mexico City 11200, Mexico
| | - Silverio Soto-Alvarez
- Laboratory of Pharmacology, Escuela Militar de Graduados de Sanidad, Universidad del Ejército y Fuerza Aérea Mexicana, Mexico City 11200, Mexico
| | - Lidia Huerta-Nuñez
- Laboratory of Pharmacology, Escuela Militar de Graduados de Sanidad, Universidad del Ejército y Fuerza Aérea Mexicana, Mexico City 11200, Mexico
| | - Leslie Chávez-Galán
- Laboratory of Integrative Immunology, Instituto Nacional de Enfermedades Respiratorias Ismael Cosio Villegas, Mexico City 14080. Mexico.
| |
Collapse
|
2
|
Rodrigues RR, Freitas VS, Alves PM, Almeida Freitas RD, Souza LBD, Andrade Santos PPD. Evaluation of the presence of Th1 response through T-bet and IFN-gamma immunohistochemical expression in lower lip and oral tongue squamous cell carcinomas. Pathol Res Pract 2024; 253:155010. [PMID: 38101155 DOI: 10.1016/j.prp.2023.155010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Accepted: 12/02/2023] [Indexed: 12/17/2023]
Abstract
INTRODUCTION Evaluate the immunohistochemical expression of T-bet and IFN-γ in lower lip (LLSCC) and oral tongue squamous cell carcinoma (OTSCC), verifying the presence of Th1 responses in lesions with different clinical conditions. METHODS AND MATERIALS Thirty OTSCC and 30 LLSCC were analyzed by immunohistochemistry. T-bet was quantitatively assessed by parenchyma cell and stroma quantification, and IFN-γ was semi-quantitatively analyzed: 1:0-25%; 2:26-50%; 3:51-75%; 4:> 75% immunopositive cells. Histological differentiation degrees were categorized as well differentiated (WD), moderately differentiated (MD), or poorly differentiated (PD). RESULTS OTSCC presented the highest number of T-bet+, parenchyma (p: 0.006), stroma (p: 0.156), parenchyma/stroma (p: 0.015), with no relationship to histological malignancy grade. IFN-γ higher concentrations in LLSCC were detected in parenchyma, stroma and in parenchyma/stroma (p: 0.000), as well as greater immunoreactivity in WD and MD (p: 0.001). In OTSCC, a positive and statistically significant correlation was observed between T-bet+ in parenchyma and IFN-γ in stroma(r: 0.388; p: 0.034), in addition to a statistically significant positive correlation between T-bet in parenchyma compared to stroma(r: 0.411; p: 0.024) and for IFN-γ in both parenchyma and stroma(r: 0.775; p: 0.000) in LLSCC. Higher T-bet+ was observed in OTSCCs, although higher IFN-γ was detected in LLSCCs. CONCLUSION Thus, we suggest that, even though LLSCC presented lower T-bet+, the favorable microenvironment in these lesions led to an expressive activation of IFN-γ by T-bet+, considerably acting on Th1 differentiation and in antitumor activity, which, admittedly, present less aggressive behavior, reinforcing once again the important role of this cytokine and its use in strategy to fight cancer.
Collapse
Affiliation(s)
| | - Valéria Souza Freitas
- Department of Dentistry, State University of Feira de Santana (UEFS), Feira de Santana, BA, Brazil
| | - Pollianna Muniz Alves
- Department of Dentistry, State University of Paraíba (UEPB), Campina Grande, PB, Brazil
| | | | - Lélia Batista de Souza
- Department of Oral Pathology, Federal University of Rio Grande do Norte (UFRN), Natal, RN, Brazil
| | | |
Collapse
|
3
|
Dabbaghi R, Safaralizadeh R, Rahmani S, Barpour N, Hosseinpourfeizi M, Rajabi A, Baradaran B. The effect of glatiramer acetate, IFNβ-1a, fingolimod, and dimethyl fumarate on the expression of T-bet, IFN-γ, and MEG3 in PBMC of RRMS patients. BMC Res Notes 2023; 16:273. [PMID: 37845751 PMCID: PMC10577903 DOI: 10.1186/s13104-023-06556-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 10/05/2023] [Indexed: 10/18/2023] Open
Abstract
OBJECTIVE Multiple sclerosis (MS) is a progressing neurodegenerative disease marked by chronic central nervous system inflammation and degeneration.This study investigates gene expression profiles of T-box transcription factor TBX21 (T-bet), interferon-gamma (IFN-γ), and long non-coding RNA MEG3 in peripheral blood mononuclear cells (PBMCs) from treatment-naïve Relapsing-Remitting Multiple Sclerosis patients (RRMS), healthy controls, and RRMS patients on different Disease Modifying Therapies (DMTs). The aim is to understand the role of T-bet, IFN-γ, and MEG3 in MS pathogenesis and their potential as diagnostic and therapeutic targets. RESULTS Elevated T-bet expression is observed in treatment-naïve RRMS patients compared to healthy individuals. RRMS patients treated with Interferon beta-1alpha (IFNβ-1a) and fingolimod exhibit downregulated T-bet and MEG3 expression levels, respectively, with more pronounced effects in females. Healthy individuals show a moderate positive correlation between T-bet and MEG3 and between IFN-γ and T-bet. In RRMS patients treated with Glatiramer Acetate (GA), a strong positive correlation is observed between MEG3 and IFN-γ. Remarkably, RRMS patients treated with Dimethyl Fumarate (DMF) exhibit a significant positive correlation between T-bet and MEG3. These findings underscore the diagnostic potential of T-bet in RRMS, warranting further exploration of MEG3, T-bet, and IFN-γ interplay in RRMS patients.
Collapse
Affiliation(s)
- Rozhin Dabbaghi
- Department of Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| | - Reza Safaralizadeh
- Department of Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran.
- Department of Animal Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran.
| | - Shima Rahmani
- Department of Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nesa Barpour
- Department of Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| | | | - Ali Rajabi
- Department of Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
4
|
Hausmann F, Ergen C, Khatri R, Marouf M, Hänzelmann S, Gagliani N, Huber S, Machart P, Bonn S. DISCERN: deep single-cell expression reconstruction for improved cell clustering and cell subtype and state detection. Genome Biol 2023; 24:212. [PMID: 37730638 PMCID: PMC10510283 DOI: 10.1186/s13059-023-03049-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 08/23/2023] [Indexed: 09/22/2023] Open
Abstract
BACKGROUND Single-cell sequencing provides detailed insights into biological processes including cell differentiation and identity. While providing deep cell-specific information, the method suffers from technical constraints, most notably a limited number of expressed genes per cell, which leads to suboptimal clustering and cell type identification. RESULTS Here, we present DISCERN, a novel deep generative network that precisely reconstructs missing single-cell gene expression using a reference dataset. DISCERN outperforms competing algorithms in expression inference resulting in greatly improved cell clustering, cell type and activity detection, and insights into the cellular regulation of disease. We show that DISCERN is robust against differences between batches and is able to keep biological differences between batches, which is a common problem for imputation and batch correction algorithms. We use DISCERN to detect two unseen COVID-19-associated T cell types, cytotoxic CD4+ and CD8+ Tc2 T helper cells, with a potential role in adverse disease outcome. We utilize T cell fraction information of patient blood to classify mild or severe COVID-19 with an AUROC of 80% that can serve as a biomarker of disease stage. DISCERN can be easily integrated into existing single-cell sequencing workflow. CONCLUSIONS Thus, DISCERN is a flexible tool for reconstructing missing single-cell gene expression using a reference dataset and can easily be applied to a variety of data sets yielding novel insights, e.g., into disease mechanisms.
Collapse
Affiliation(s)
- Fabian Hausmann
- Institute of Medical Systems Biology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
- Center for Biomedical AI, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
| | - Can Ergen
- Institute of Medical Systems Biology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
- Center for Biomedical AI, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
- I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
| | - Robin Khatri
- Institute of Medical Systems Biology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
- Center for Biomedical AI, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
| | - Mohamed Marouf
- Institute of Medical Systems Biology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
| | - Sonja Hänzelmann
- Institute of Medical Systems Biology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
- Center for Biomedical AI, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
| | - Nicola Gagliani
- I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
- Hamburg Center for Translational Immunology (HCTI), I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
- Section of Molecular Immunology und Gastroenterology, I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
| | - Samuel Huber
- I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
- Hamburg Center for Translational Immunology (HCTI), I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
| | - Pierre Machart
- Institute of Medical Systems Biology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
- Center for Biomedical AI, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
| | - Stefan Bonn
- Institute of Medical Systems Biology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany.
- Center for Biomedical AI, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany.
- Hamburg Center for Translational Immunology (HCTI), I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany.
| |
Collapse
|
5
|
Venkatesh H, Tracy SI, Farrar MA. Cytotoxic CD4 T cells in the mucosa and in cancer. Front Immunol 2023; 14:1233261. [PMID: 37654482 PMCID: PMC10466411 DOI: 10.3389/fimmu.2023.1233261] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 07/24/2023] [Indexed: 09/02/2023] Open
Abstract
CD4 T cells were initially described as helper cells that promote either the cellular immune response (Th1 cells) or the humoral immune response (Th2 cells). Since then, a plethora of functionally distinct helper and regulatory CD4 T cell subsets have been described. CD4 T cells with cytotoxic function were first described in the setting of viral infections and autoimmunity, and more recently in cancer and gut dysbiosis. Regulatory CD4 T cell subsets such as Tregs and T-regulatory type 1 (Tr1) cells have also been shown to have cytotoxic potential. Indeed, Tr1 cells have been shown to be important for maintenance of stem cell niches in the bone marrow and the gut. This review will provide an overview of cytotoxic CD4 T cell development, and discuss the role of inflammatory and Tr1-like cytotoxic CD4 T cells in maintenance of intestinal stem cells and in anti-cancer immune responses.
Collapse
Affiliation(s)
- Hrishi Venkatesh
- Center for Immunology, Masonic Cancer Center, Minneapolis, MN, United States
- University of Minnesota, Department of Laboratory Medicine and Pathology, Minneapolis, MN, United States
| | - Sean I. Tracy
- Center for Immunology, Masonic Cancer Center, Minneapolis, MN, United States
- Division of Hematology, Oncology and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, MN, United States
| | - Michael A. Farrar
- Center for Immunology, Masonic Cancer Center, Minneapolis, MN, United States
- University of Minnesota, Department of Laboratory Medicine and Pathology, Minneapolis, MN, United States
| |
Collapse
|
6
|
Li L, Wen Q, Ding R. Therapeutic targeting of VEGF and/or TGF-β to enhance anti-PD-(L)1 therapy: The evidence from clinical trials. Front Oncol 2022; 12:905520. [PMID: 35957885 PMCID: PMC9360509 DOI: 10.3389/fonc.2022.905520] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Accepted: 06/30/2022] [Indexed: 11/19/2022] Open
Abstract
Normalizing the tumor microenvironment (TME) is a potential strategy to improve the effectiveness of immunotherapy. Vascular endothelial growth factor (VEGF) and transforming growth factor (TGF)-β pathways play an important role in the development and function of the TME, contributing to the immunosuppressive status of TME. To inhibit VEGF and/or TGF-β pathways can restore TME from immunosuppressive to immune-supportive status and enhance sensitivity to immunotherapy such as programmed death protein-1 (PD-1)/programmed cell death-ligand 1 (PD-L1) inhibitors. In this review, we described the existing preclinical and clinical evidence supporting the use of anti-VEGF and/or anti-TGF-β therapies to enhance cancer immunotherapy. Encouragingly, adopting anti-VEGF and/or anti-TGF-β therapies as a combination treatment with anti-PD-(L)1 therapy have been demonstrated as effective and tolerable in several solid tumors in clinical trials. Although several questions need to be solved, the clinical value of this combination strategy is worthy to be studied further.
Collapse
Affiliation(s)
- Linwei Li
- Department of Oncology, Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Qinglian Wen
- Department of Oncology, Affiliated Hospital of Southwest Medical University, Luzhou, China
- *Correspondence: Qinglian Wen, ; Ruilin Ding,
| | - Ruilin Ding
- Institute of Drug Clinical Trial/GCP Center, Affiliated Hospital of Southwest Medical University, Luzhou, China
- *Correspondence: Qinglian Wen, ; Ruilin Ding,
| |
Collapse
|
7
|
Hoeks C, Duran G, Hellings N, Broux B. When Helpers Go Above and Beyond: Development and Characterization of Cytotoxic CD4+ T Cells. Front Immunol 2022; 13:951900. [PMID: 35903098 PMCID: PMC9320319 DOI: 10.3389/fimmu.2022.951900] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 06/21/2022] [Indexed: 11/26/2022] Open
Abstract
Once regarded as an experimental artefact, cytotoxic CD4+ T cells (CD4 CTL) are presently recognized as a biologically relevant T cell subset with important functions in anti-viral, anti-tumor, and autoimmune responses. Despite the potentially large impact on their micro-environment, the absolute cell counts of CD4 CTL within the peripheral circulation are relatively low. With the rise of single cell analysis techniques, detection of these cells is greatly facilitated. This led to a renewed appraisal of CD4 CTL and an increased insight into their heterogeneous nature and ontogeny. In this review, we summarize the developmental path from naïve CD4+ T cells to terminally differentiated CD4 CTL, and present markers that can be used to detect or isolate CD4 CTL and their precursors. Subsets of CD4 CTL and their divergent functionalities are discussed. Finally, the importance of local cues as triggers for CD4 CTL differentiation is debated, posing the question whether CD4 CTL develop in the periphery and migrate to site of inflammation when called for, or that circulating CD4 CTL reflect cells that returned to the circulation following differentiation at the local inflammatory site they previously migrated to. Even though much remains to be learned about this intriguing T cell subset, it is clear that CD4 CTL represent interesting therapeutic targets for several pathologies.
Collapse
Affiliation(s)
- Cindy Hoeks
- Neuro Immune Connections & Repair Lab, Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Hasselt, Belgium
- University MS Center (UMSC), Hasselt, Belgium
| | - Gayel Duran
- Neuro Immune Connections & Repair Lab, Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Hasselt, Belgium
- University MS Center (UMSC), Hasselt, Belgium
| | - Niels Hellings
- Neuro Immune Connections & Repair Lab, Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Hasselt, Belgium
- University MS Center (UMSC), Hasselt, Belgium
| | - Bieke Broux
- Neuro Immune Connections & Repair Lab, Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Hasselt, Belgium
- University MS Center (UMSC), Hasselt, Belgium
- *Correspondence: Bieke Broux,
| |
Collapse
|
8
|
Preglej T, Ellmeier W. CD4 + Cytotoxic T cells - Phenotype, Function and Transcriptional Networks Controlling Their Differentiation Pathways. Immunol Lett 2022; 247:27-42. [PMID: 35568324 DOI: 10.1016/j.imlet.2022.05.001] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 05/09/2022] [Accepted: 05/10/2022] [Indexed: 11/05/2022]
Abstract
The two major subsets of peripheral T cells are classically divided into the CD4+ T helper cells and the cytotoxic CD8+ T cell lineage. However, the appearance of some effector CD4+ T cell populations displaying cytotoxic activity, in particular during viral infections, has been observed, thus breaking the functional dichotomy of CD4+ and CD8+ T lymphocytes. The strong association of the appearance of CD4+ cytotoxic T lymphocytes (CD4 CTLs) with viral infections suggests an important role of this subset in antiviral immunity by controlling viral replication and infection. Moreover, CD4 CTLs have been linked with anti-tumor activity and might also cause immunopathology in autoimmune diseases. This raises interest into the molecular mechanisms regulating CD4 CTL differentiation, which are poorly understood in comparison to differentiation pathways of other Th subsets. In this review, we provide a brief overview about key features of CD4 CTLs, including their role in viral infections and cancer immunity, and about the link between CD4 CTLs and immune-mediated diseases. Subsequently, we will discuss the current knowledge about transcriptional and epigenetic networks controlling CD4 CTL differentiation and highlight recent data suggesting a role for histone deacetylases in the generation of CD4 CTLs.
Collapse
Affiliation(s)
- Teresa Preglej
- Division of Rheumatology, Department of Internal Medicine III, Medical University of Vienna
| | - Wilfried Ellmeier
- Division of Immunobiology, Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna.
| |
Collapse
|
9
|
Xu C, Marelli B, Qi J, Qin G, Yu H, Wang H, Jenkins MH, Lo KM, Lan Y. NHS-IL12 and bintrafusp alfa combination therapy enhances antitumor activity in preclinical cancer models. Transl Oncol 2021; 16:101322. [PMID: 34954456 PMCID: PMC8718653 DOI: 10.1016/j.tranon.2021.101322] [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: 11/10/2021] [Revised: 12/06/2021] [Accepted: 12/17/2021] [Indexed: 01/22/2023] Open
Abstract
Combination therapy induced both adaptive and innate antitumor immunity. Combination therapy significantly enhanced antitumor activity. Combination therapy generated tumor antigen specific immune memory. Bintrafusp alfa mediated the reduction of lung metastases.
Combinatorial immunotherapy approaches are emerging as viable cancer therapeutic strategies for improving patient responses and outcomes. This study investigated whether two such immunotherapies, with complementary mechanisms of action, could enhance antitumor activity in murine tumor models. The immunocytokine NHS-IL12, and surrogate NHS-muIL12, are designed to deliver IL-12 and muIL-12, respectively, to the tumor microenvironment (TME) to activate NK cells and CD8+ T cells and increase their cytotoxic functions. Bintrafusp alfa (BA) is a bifunctional fusion protein composed of the extracellular domains of the TGF-β receptor II to function as a TGF-β “trap” fused to a human IgG1 antibody blocking PD-L1. With this dual-targeting strategy, BA enhances efficacy over that of monotherapies in preclinical studies. In this study, NHS-muIL12 and BA combination therapy enhanced antitumor activity, prolonged survival, and induced tumor-specific antitumor immunity. This combination therapy increased tumor-specific CD8+ T cells and induced immune profiles, consistent with the activation of both adaptive and innate immune systems. In addition, BA reduced lung metastasis in the 4T1 model. Collectively, these findings could support clinical trials designed to investigate NHS-IL12 and BA combination therapy for patients with advanced solid tumors
Collapse
Affiliation(s)
- Chunxiao Xu
- EMD Serono Research and Development Institute, Inc, 45 Middlesex Turnpike, Billerica, MA 01821, USA.
| | - Bo Marelli
- EMD Serono Research and Development Institute, Inc, 45 Middlesex Turnpike, Billerica, MA 01821, USA
| | - Jin Qi
- EMD Serono Research and Development Institute, Inc, 45 Middlesex Turnpike, Billerica, MA 01821, USA
| | - Guozhong Qin
- EMD Serono Research and Development Institute, Inc, 45 Middlesex Turnpike, Billerica, MA 01821, USA
| | - Huakui Yu
- EMD Serono Research and Development Institute, Inc, 45 Middlesex Turnpike, Billerica, MA 01821, USA
| | - Hong Wang
- EMD Serono Research and Development Institute, Inc, 45 Middlesex Turnpike, Billerica, MA 01821, USA
| | - Molly H Jenkins
- EMD Serono Research and Development Institute, Inc, 45 Middlesex Turnpike, Billerica, MA 01821, USA
| | - Kin-Ming Lo
- EMD Serono Research and Development Institute, Inc, 45 Middlesex Turnpike, Billerica, MA 01821, USA
| | - Yan Lan
- EMD Serono Research and Development Institute, Inc, 45 Middlesex Turnpike, Billerica, MA 01821, USA.
| |
Collapse
|
10
|
TGF-β Signaling: From Tissue Fibrosis to Tumor Microenvironment. Int J Mol Sci 2021; 22:ijms22147575. [PMID: 34299192 PMCID: PMC8303588 DOI: 10.3390/ijms22147575] [Citation(s) in RCA: 91] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 07/09/2021] [Accepted: 07/09/2021] [Indexed: 02/06/2023] Open
Abstract
Transforming growth factor-β (TGF-β) signaling triggers diverse biological actions in inflammatory diseases. In tissue fibrosis, it acts as a key pathogenic regulator for promoting immunoregulation via controlling the activation, proliferation, and apoptosis of immunocytes. In cancer, it plays a critical role in tumor microenvironment (TME) for accelerating invasion, metastasis, angiogenesis, and immunosuppression. Increasing evidence suggest a pleiotropic nature of TGF-β signaling as a critical pathway for generating fibrotic TME, which contains numerous cancer-associated fibroblasts (CAFs), extracellular matrix proteins, and remodeling enzymes. Its pathogenic roles and working mechanisms in tumorigenesis are still largely unclear. Importantly, recent studies successfully demonstrated the clinical implications of fibrotic TME in cancer. This review systematically summarized the latest updates and discoveries of TGF-β signaling in the fibrotic TME.
Collapse
|
11
|
Khanali J, Azangou-Khyavy M, Boroomand-Saboor M, Ghasemi M, Niknejad H. JAK/STAT-Dependent Chimeric Antigen Receptor (CAR) Expression: A Design Benefiting From a Dual AND/OR Gate Aiming to Increase Specificity, Reduce Tumor Escape and Affect Tumor Microenvironment. Front Immunol 2021; 12:638639. [PMID: 34177890 PMCID: PMC8220211 DOI: 10.3389/fimmu.2021.638639] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 05/21/2021] [Indexed: 12/04/2022] Open
Abstract
Recent advances in cancer immunotherapy have attracted great interest due to the natural capacity of the immune system to fight cancer. This field has been revolutionized by the advent of chimeric antigen receptor (CAR) T cell therapy that is utilizing an antigen recognition domain to redirect patients’ T cells to selectively attack cancer cells. CAR T cells are designed with antigen-binding moieties fused to signaling and co-stimulatory intracellular domains. Despite significant success in hematologic malignancies, CAR T cells encounter many obstacles for treating solid tumors due to tumor heterogeneity, treatment-associated toxicities, and immunosuppressive tumor microenvironment. Although the current strategies for enhancing CAR T cell efficacy and specificity are promising, they have their own limitations, making it necessary to develop new genetic engineering strategies. In this article, we have proposed a novel logic gate for recognizing tumor-associated antigens by employing intracellular JAK/STAT signaling pathway to enhance CAR T Cells potency and specificity. Moreover, this new-generation CAR T cell is empowered to secrete bispecific T cell engagers (BiTEs) against cancer-associated fibroblasts (CAFs) to diminish tumor metastasis and angiogenesis and increase T cell infiltration.
Collapse
Affiliation(s)
- Javad Khanali
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | | | - Mobina Ghasemi
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hassan Niknejad
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
12
|
Essential Kinases and Transcriptional Regulators and Their Roles in Autoimmunity. Biomolecules 2019; 9:biom9040145. [PMID: 30974919 PMCID: PMC6523499 DOI: 10.3390/biom9040145] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 04/04/2019] [Accepted: 04/05/2019] [Indexed: 12/11/2022] Open
Abstract
Kinases and transcriptional regulators are fundamental components of cell signaling that are expressed on many types of immune cells which are involved in secretion of cytokines, cell proliferation, differentiation, and apoptosis. Both play important roles in biological responses in health as well as in illnesses such as the autoimmune diseases which comprise at least 80 disorders. These diseases are caused by complex genetic and environmental interactions that lead to a breakage of immunologic tolerance and a disruption of the balance between self-reactive cells and regulatory cells. Kinases or transcriptional regulatory factors often have an abnormal expression in the autoimmune cells that participate in the pathogenesis of autoimmune disease. These abnormally expressed kinases or transcriptional regulators can over-activate the function of self-reactive cells to produce inflammatory cytokines or down-regulate the activity of regulatory cells, thus causing autoimmune diseases. In this review we introduce five kinds of kinase and transcriptional regulator related to autoimmune diseases, namely, members of the Janus kinase (JAK) family (JAK3 and/or tyrosine kinase 2 (TYK2)), fork head box protein 3 (Foxp3), the retinoic acid-related orphan receptor gamma t (RORγt), and T-box expressed in T cells (T-bet) factors. We also provide a mechanistic insight into how these kinases and transcriptional regulators affect the function of the immune cells related to autoimmune diseases, as well as a description of a current drug design targeting these kinases and transcriptional regulators. Understanding their exact role helps offer new therapies for control of the inflammatory responses that could lead to clinical improvement of the autoimmune diseases.
Collapse
|
13
|
Liu Y. A global immune gene expression signature for human cancers. Oncotarget 2019; 10:1993-2005. [PMID: 30956779 PMCID: PMC6443003 DOI: 10.18632/oncotarget.26773] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Accepted: 02/22/2019] [Indexed: 01/22/2023] Open
Abstract
Background Except for a few, the immune gene signatures for most cancer types are not available. We sought to identify a global immune gene signature that is applicable to all human cancers. Results We identified an immune gene signature that was intimately correlated with tumor immune characteristics of human cancers and consisted of 382 genes indicative of different immune cell types. The T helper type 1 and 2 cell activation pathway was most significantly enriched in this global immune gene set, while transcription factors, such as SPI1 and STAT family members, were the top regulators of this gene signature. Skin cutaneous melanoma with higher expression of this immune gene signature had significantly longer survival than those with lower immune gene expression. Breast cancer patients with higher immune gene signature were significantly associated with advanced-stage. Methods We analyzed the gene expression profiles of 10,062 tumor samples from 32 cancer types in The Cancer Genome Atlas Pan-Cancer data set. Hierarchical clustering analysis of previously-defined immune genes was performed to identify a pan-cancer immune gene signature. Pathway and upstream regulator analyses were used to identify significantly enriched signaling pathways and transcription factors. Kaplan-Meier analysis was used to evaluate the survival difference between dichotomic groups with different immune gene signatures. Correlation of immune gene signature with tumor stage was also examined. Conclusions Our identified immune gene signature is applicable in human cancers and can be used to characterize tumor immunogenicity within and across cancer types. Clinical implication of this immune gene set warrants future investigation.
Collapse
Affiliation(s)
- Yuexin Liu
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|