1
|
Vázquez-Marmolejo AV, Duarte-Mata DI, Ruiz-De La Cruz ML, López-López N, Salinas-Carmona MC, Mejía-Torres M. Increased expression of the PD-1/PD-L1 regulatory axis in tissue leukocytes from experimental actinomycetoma by Nocardia brasiliensis in BALB/c mice. Microb Pathog 2025; 205:107697. [PMID: 40368069 DOI: 10.1016/j.micpath.2025.107697] [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: 11/01/2024] [Revised: 05/08/2025] [Accepted: 05/12/2025] [Indexed: 05/16/2025]
Abstract
The development of experimental actinomycetoma in mice pioneered the study of infective mechanisms in nocardiosis. However, the understanding of lymphocyte activation in actinomycetoma remains incomplete. In this study, we used flow cytometry to evaluate the cellularity and expression of regulatory receptors on leukocytes from spleen and infected tissue in a mice model of experimental actinomycetoma induced by Nocardia brasiliensis. Our results indicate that neutrophils dominate cellularity in infected tissue, representing >90% of infiltrated leukocytes. Among lymphocytes, the percentage of Th1 and Tc1 cells decreases in spleen and infected tissue during chronic infection. Likewise, both tissues had similar changes in leukocyte expression of Programmed Cell Death Protein 1 (PD-1), T-cell immunoglobulin and mucin-domain containing-3 (TIM-3), and Programmed death-ligand 1 (PD-L1), suggesting the delayed systemic involvement of an initially local disease.
Collapse
Affiliation(s)
- Anna Velia Vázquez-Marmolejo
- Universidad Autónoma de Nuevo León, Service of Immunology, Hospital Universitario "Dr. José Eleuterio González", Monterrey, NL, México
| | - Diana Ivonne Duarte-Mata
- Universidad Autónoma de Nuevo León, Service of Immunology, Hospital Universitario "Dr. José Eleuterio González", Monterrey, NL, México
| | - María Luisa Ruiz-De La Cruz
- Universidad Autónoma de Nuevo León, Service of Immunology, Hospital Universitario "Dr. José Eleuterio González", Monterrey, NL, México
| | - Nallely López-López
- Universidad Autónoma de Nuevo León, Service of Immunology, Hospital Universitario "Dr. José Eleuterio González", Monterrey, NL, México
| | - Mario César Salinas-Carmona
- Universidad Autónoma de Nuevo León, Service of Immunology, Hospital Universitario "Dr. José Eleuterio González", Monterrey, NL, México
| | - Manuel Mejía-Torres
- Universidad Autónoma de Nuevo León, Service of Immunology, Hospital Universitario "Dr. José Eleuterio González", Monterrey, NL, México.
| |
Collapse
|
2
|
Xie Y, Peng H, Hu Y, Jia K, Yuan J, Liu D, Li Y, Feng X, Li J, Zhang X, Sun Y, Shen L, Chen Y. Immune microenvironment spatial landscapes of tertiary lymphoid structures in gastric cancer. BMC Med 2025; 23:59. [PMID: 39901202 PMCID: PMC11792408 DOI: 10.1186/s12916-025-03889-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Accepted: 01/22/2025] [Indexed: 02/05/2025] Open
Abstract
BACKGROUND Tertiary lymphoid structures (TLS) correlate with tumour prognosis and immunotherapy responses in gastric cancer (GC) studies. However, understanding the complex and diverse immune microenvironment within TLS requires comprehensive analysis. METHODS We examined the prognostic impact of TLS within the tumour core (TC) of 59 GC patients undergoing immunotherapy. Multispectral fluorescence imaging was employed to evaluate variations in immune cell infiltration across different TLS sites among 110 GC patients, by quantifying immune cell density and spatial characteristics. We also generated a single-cell transcriptomic atlas of TLS-positive (n = 4) and TLS-negative (n = 8) microenvironments and performed spatial transcriptomics (ST) analysis on two samples. RESULTS TLS presence in the TC significantly correlated with improved immune-related overall survival (P = 0.049). CD8+LAG-3-PD-1+TIM-3-, CD4+PD-L1+, and CD4+FoxP3- T cell densities were significantly higher in the TLS within TC compared to tumour and stromal regions. Immune cells within TLS exhibited closer intercellular proximity than those outside TLS. Five key density and spatial characteristics of immune cells within TLS in the TC were selected to develop the Density and Spatial Score risk model. Single-cell RNA sequencing revealed strong intercellular interactions in the presence of TLS within the microenvironment. However, TLS-absent environment facilitated tumour cell interactions with immune cells through MIF- and galectin-dependent pathways, recruiting immunosuppressive cells. ST analysis confirmed that T and B cells co-localise within TLS, enhancing immune response activation compared to cancer nests and exerting a strong anti-tumour effect. CONCLUSIONS TLS presence facilitates frequent cell-to-cell communication, forming an active immune microenvironment, highlighting the prognostic value of TLS.
Collapse
Affiliation(s)
- Yi Xie
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Peking University Cancer Hospital and Institute, Beijing, 100142, China
| | - Haoxin Peng
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Peking University Cancer Hospital and Institute, Beijing, 100142, China
| | - Yajie Hu
- Department of Pathology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing, 100142, China
| | - Keren Jia
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Peking University Cancer Hospital and Institute, Beijing, 100142, China
| | - Jiajia Yuan
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Peking University Cancer Hospital and Institute, Beijing, 100142, China
| | - Dan Liu
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Peking University Cancer Hospital and Institute, Beijing, 100142, China
| | - Yanyan Li
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Peking University Cancer Hospital and Institute, Beijing, 100142, China
| | - Xujiao Feng
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Peking University Cancer Hospital and Institute, Beijing, 100142, China
| | - Jian Li
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Peking University Cancer Hospital and Institute, Beijing, 100142, China
| | - Xiaotian Zhang
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Peking University Cancer Hospital and Institute, Beijing, 100142, China
| | - Yu Sun
- Department of Pathology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing, 100142, China.
| | - Lin Shen
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Peking University Cancer Hospital and Institute, Beijing, 100142, China.
| | - Yang Chen
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Peking University Cancer Hospital and Institute, Beijing, 100142, China.
- Department of Gastrointestinal Cancer, Beijing GoBroad Hospital, Beijing, 102200, China.
| |
Collapse
|
3
|
Arafat Hossain M. A comprehensive review of immune checkpoint inhibitors for cancer treatment. Int Immunopharmacol 2024; 143:113365. [PMID: 39447408 DOI: 10.1016/j.intimp.2024.113365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Revised: 09/28/2024] [Accepted: 10/05/2024] [Indexed: 10/26/2024]
Abstract
Immunology-based therapies are emerging as an effective cancer treatment, using the body's immune system to target tumors. Immune checkpoints, which regulate immune responses to prevent tissue damage and autoimmunity, are often exploited by cancer cells to avoid destruction. The discovery of checkpoint proteins like PD-1/PD-L1 and CTLA-4 was pivotal in developing cancer immunotherapy. Immune checkpoint inhibitors (ICIs) have shown great success, with FDA-approved drugs like PD-1 inhibitors (Nivolumab, Pembrolizumab, Cemiplimab), PD-L1 inhibitors (Atezolizumab, Durvalumab, Avelumab), and CTLA-4 inhibitors (Ipilimumab, Tremelimumab), alongside LAG-3 inhibitor Relatlimab. Research continues on new checkpoints like TIM-3, VISTA, B7-H3, BTLA, and TIGIT. Biomarkers like PDL-1 expression, tumor mutation burden, interferon-γ presence, microbiome composition, and extracellular matrix characteristics play a crucial role in predicting responses to immunotherapy with checkpoint inhibitors. Despite their effectiveness, not all patients experience the same level of benefit, and organ-specific immune-related adverse events (irAEs) such as rash or itching, colitis, diarrhea, hyperthyroidism, and hypothyroidism may occur. Given the rapid advancements in this field and the variability in patient outcomes, there is an urgent need for a comprehensive review that consolidates the latest findings on immune checkpoint inhibitors, covering their clinical status, biomarkers, resistance mechanisms, strategies to overcome resistance, and associated adverse effects. This review aims to fill this gap by providing an analysis of the current clinical status of ICIs, emerging biomarkers, mechanisms of resistance, strategies to enhance therapeutic efficacy, and assessment of adverse effects. This review is crucial to furthering our understanding of ICIs and optimizing their application in cancer therapy.
Collapse
Affiliation(s)
- Md Arafat Hossain
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj 8100, Bangladesh.
| |
Collapse
|
4
|
Maymí VI, Zhu H, Jager M, Johnson S, Getchell R, Casey JW, Grenier JK, Wherry EJ, Smith NL, Grimson A, Rudd BD. Neonatal CD8+ T Cells Resist Exhaustion during Chronic Infection. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 212:834-843. [PMID: 38231127 PMCID: PMC11298781 DOI: 10.4049/jimmunol.2300396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 01/01/2024] [Indexed: 01/18/2024]
Abstract
Chronic viral infections, such as HIV and hepatitis C virus, represent a major public health problem. Although it is well understood that neonates and adults respond differently to chronic viral infections, the underlying mechanisms remain unknown. In this study, we transferred neonatal and adult CD8+ T cells into a mouse model of chronic infection (lymphocytic choriomeningitis virus clone 13) and dissected out the key cell-intrinsic differences that alter their ability to protect the host. Interestingly, we found that neonatal CD8+ T cells preferentially became effector cells early in chronic infection compared with adult CD8+ T cells and expressed higher levels of genes associated with cell migration and effector cell differentiation. During the chronic phase of infection, the neonatal cells retained more immune functionality and expressed lower levels of surface markers and genes related to exhaustion. Because the neonatal cells protect from viral replication early in chronic infection, the altered differentiation trajectories of neonatal and adult CD8+ T cells is functionally significant. Together, our work demonstrates how cell-intrinsic differences between neonatal and adult CD8+ T cells influence key cell fate decisions during chronic infection.
Collapse
Affiliation(s)
- Viviana I. Maymí
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY, USA
| | - Hongya Zhu
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY, USA
| | - Mason Jager
- Department of Population Medicine and Diagnostic Sciences, Cornell University, Ithaca, NY 14853, USA
| | - Shawn Johnson
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY, USA
| | - Rodman Getchell
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY, USA
| | - James W. Casey
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY, USA
| | - Jennifer K. Grenier
- Transcriptional Regulation and Expression Facility, Department of Biomedical Sciences, Cornell University, Ithaca, NY 14853, USA
| | - E. John Wherry
- Institute for Immunology and Immune Health, and Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Norah L. Smith
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY, USA
| | - Andrew Grimson
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY, USA
| | - Brian D. Rudd
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY, USA
| |
Collapse
|
5
|
Manandhar P, Szymczak-Workman AL, Kane LP. Tim-3 Is Not Required for Establishment of CD8+ T Cell Memory to Lymphocytic Choriomeningitis Virus. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 212:466-474. [PMID: 38108417 PMCID: PMC10906969 DOI: 10.4049/jimmunol.2300401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 11/21/2023] [Indexed: 12/19/2023]
Abstract
Tim-3 is a transmembrane protein that is best known for being highly expressed on terminally exhausted CD8+ T cells associated with chronic infection and tumors, although its expression is not limited to those settings. Tim-3 is also expressed by CD8+ T cells during acute infection and by multiple other immune cell types, including CD4+ Th1 and regulatory T cells, dendritic cells, and mast cells. In this study, we investigated the role of Tim-3 signaling on CD8+ T cell memory using a Tim-3 conditional knockout mouse model and mice lacking the signaling portion of the Tim-3 cytoplasmic domain. Together, our results indicate that Tim-3 has at most a modest effect on the formation and function of CD8+ memory T cells.
Collapse
Affiliation(s)
- Priyanka Manandhar
- Dept. of Immunology, University of Pittsburgh, Pittsburgh, PA 15213
- Graduate Program in Microbiology and Immunology, University of Pittsburgh, Pittsburgh, PA 15213
| | | | - Lawrence P. Kane
- Dept. of Immunology, University of Pittsburgh, Pittsburgh, PA 15213
- Graduate Program in Microbiology and Immunology, University of Pittsburgh, Pittsburgh, PA 15213
| |
Collapse
|
6
|
Rastin F, Javid H, Oryani MA, Rezagholinejad N, Afshari AR, Karimi-Shahri M. Immunotherapy for colorectal cancer: Rational strategies and novel therapeutic progress. Int Immunopharmacol 2024; 126:111055. [PMID: 37992445 DOI: 10.1016/j.intimp.2023.111055] [Citation(s) in RCA: 31] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Revised: 09/30/2023] [Accepted: 10/09/2023] [Indexed: 11/24/2023]
Abstract
There are increasing incidences and mortality rates for colorectal cancer in the world. It is common for chemotherapy and radiation given to patients with colorectal cancer to cause toxicities that limit their effectiveness and cause cancer cells to become resistant to these treatments. Additional targeted treatments are needed to improve patient's quality of life and outcomes. Immunotherapy has rapidly emerged as an incredibly exciting and promising avenue for cancer treatment in recent years. This innovative approach provides novel options for tackling solid tumors, effectively establishing itself as a new cornerstone in cancer treatment. Specifically, in the realm of colorectal cancer (CRC), there is great promise in developing new drugs that target immune checkpoints, offering a hopeful and potentially transformative solution. While immunotherapy of CRC has made significant advances, there are still obstacles and limitations. CRC patients have a poor response to treatment because of the immune-suppressing function of their tumor microenvironment (TME). In addition to blocking inhibitory immune checkpoints, checkpoint-blocking antibodies may also boost immune responses against tumors. The review summarizes recent advances in immune checkpoint inhibitors (ICIs) for CRC, including CTLA-4, PD-1, PD-L1, LAG-3, and TIM-3.
Collapse
Affiliation(s)
- Farangis Rastin
- Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Hossein Javid
- Department of Medical Laboratory Sciences, Varastegan Institute for Medical Sciences, Mashhad, Iran; Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Surgical Oncology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Mahsa Akbari Oryani
- Department of Pathology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Amir-R Afshari
- Department of Physiology and Pharmacology, School of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Mehdi Karimi-Shahri
- Department of Pathology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pathology, School of Medicine, Gonabad University of Medical Sciences, Gonabad, Iran.
| |
Collapse
|
7
|
Mahanti K, Bhattacharyya S. Rough neighborhood: Intricacies of cancer stem cells and infiltrating immune cell interaction in tumor microenvironment and potential in therapeutic targeting. Transl Res 2023; 265:S1931-5244(23)00176-7. [PMID: 39491179 DOI: 10.1016/j.trsl.2023.10.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 10/25/2023] [Accepted: 10/25/2023] [Indexed: 11/05/2024]
Abstract
Ongoing research on cellular heterogeneity of Cancer stem cells (CSCs) and its synergistic involvement with tumor milieu reveals enormous complexity, resulting in diverse hindrance in immune therapy. CSCs has captured attention for their contribution in shaping of tumor microenvironment and as target for therapeutic intervention. Recent studies have highlighted cell-extrinsic and intrinsic mechanisms of reciprocal interaction between tumor stroma constituents and CSCs. Therapeutic targeting requires an in-depth understanding of the underlying mechanisms involved with the rate limiting factors in tumor aggressiveness and pinpoint role of CSCs. Some of the major constituents of tumor microenvironment includes resident and infiltrating immune cell, both innate and adaptive. Some of these immune cells play crucial role as adjustors of tumor immune response. Tumor-adjustor immune cell interaction confer plasticity and features enabling tumor growth and metastasis in one hand and on the other hand blunts anti-tumor immunity. Detail understanding of CSC and TME resident immune cells interaction can shape new avenues for cancer immune therapy. In this review, we have tried to summarize the development of knowledge on cellular, molecular and functional interaction between CSCs and tumor microenvironment immune cells, highlighting immune-mediated therapeutic strategies aimed at CSCs. We also discussed developing a potential CSC and TME targeted therapeutic avenue.
Collapse
Affiliation(s)
- Krishna Mahanti
- Immunobiology and Translational medicine laboratory, Department of Zoology, Sidho Kanho Birsha University, Purulia, 723104, West Bengal India
| | - Sankar Bhattacharyya
- Immunobiology and Translational medicine laboratory, Department of Zoology, Sidho Kanho Birsha University, Purulia, 723104, West Bengal India.
| |
Collapse
|
8
|
Liu Z, Liang W, Zhu Q, Cheng X, Qian R, Gao Y. PSRC1 Regulated by DNA Methylation Is a Novel Target for LGG Immunotherapy. J Mol Neurosci 2023; 73:516-528. [PMID: 37326762 DOI: 10.1007/s12031-023-02133-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 05/28/2023] [Indexed: 06/17/2023]
Abstract
Proline and serine-rich coiled-coil 1 (PSRC1) has been reported to function as an oncogene in several cancers by regulating mitosis, while there are few reports on the role of PSRC1 in lower-grade glioma (LGG). Thus, this study collected 22 samples and 1126 samples from our institution and several databases, respectively, to explore the function of PSRC1 in LGG. First, the analysis of clinical characteristics showed that PSRC1 was always highly expressed in more malignant clinical characteristics of LGG, such as higher WHO grade, recurrence type, and IDH wild type. Second, the prognosis analysis revealed that the high expression of PSRC1 was an independent risk factor contributing to the shorter overall survival of LGG patients. Third, the analysis of DNA methylation showed that the expression of PSRC1 was associated with its 8 DNA methylation sites, overall negatively regulated by its DNA methylation level in LGG. Fourth, the analysis of immune correlation revealed that the expression of PSRC1 was positively correlated with the infiltration of 6 immune cells and the expression of 4 well-known immune checkpoints in LGG, respectively. Finally, co-expression analysis and KEGG analysis showed the 10 genes most related to PSRC1 and the signaling pathways involved by PSRC1 in LGG, respectively, such as MAPK signaling pathway and focal adhesion. In conclusion, this study identified the pathogenic role of PSRC1 in the pathological progression of LGG, expanding the molecular understanding of PSRC1, and provided a biomarker and potential immunotherapeutic target for the treatment of LGG.
Collapse
Affiliation(s)
- Zhendong Liu
- Department of Surgery of Spine and Spinal Cord, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, No.7 Weiwu Road, Jinshui District, Zhengzhou, 450003, Henan Province, China
| | - Wenjia Liang
- People's Hospital of Henan University, Henan Provincial People's Hospital, , Microbiome Laboratory, Zhengzhou, 450003, Henan Province, China
| | - Qingyun Zhu
- Henan University School of Clinical Medicine, Henan Provincial People's Hospital, Microbiome Laboratory, Zhengzhou, 450003, Henan Province, China
| | - Xingbo Cheng
- Department of Surgery of Spine and Spinal Cord, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, No.7 Weiwu Road, Jinshui District, Zhengzhou, 450003, Henan Province, China
| | - Rongjun Qian
- Department of Neurosurgery, Henan Provincial People's Hospital, People's Hospital of Henan University, People's Hospital of Zhengzhou University, , Zhengzhou, 450003, Henan Province, China.
| | - Yanzheng Gao
- Department of Surgery of Spine and Spinal Cord, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, No.7 Weiwu Road, Jinshui District, Zhengzhou, 450003, Henan Province, China.
| |
Collapse
|
9
|
Gaißler A, Bochem J, Spreuer J, Ottmann S, Martens A, Amaral T, Wagner NB, Claassen M, Meier F, Terheyden P, Garbe C, Eigentler T, Weide B, Pawelec G, Wistuba-Hamprecht K. Early decrease of blood myeloid-derived suppressor cells during checkpoint inhibition is a favorable biomarker in metastatic melanoma. J Immunother Cancer 2023; 11:e006802. [PMID: 37286306 PMCID: PMC10254874 DOI: 10.1136/jitc-2023-006802] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/28/2023] [Indexed: 06/09/2023] Open
Abstract
BACKGROUND The need for reliable clinical biomarkers to predict which patients with melanoma will benefit from immune checkpoint blockade (ICB) remains unmet. Several different parameters have been considered in the past, including routine differential blood counts, T cell subset distribution patterns and quantification of peripheral myeloid-derived suppressor cells (MDSC), but none has yet achieved sufficient accuracy for clinical utility. METHODS Here, we investigated potential cellular biomarkers from clinical routine blood counts as well as several myeloid and T cell subsets, using flow cytometry, in two independent cohorts of a total of 141 patients with stage IV M1c melanoma before and during ICB. RESULTS Elevated baseline frequencies of monocytic MDSCs (M-MDSC) in the blood were confirmed to predict shorter overall survival (OS) (HR 2.086, p=0.030) and progression-free survival (HR 2.425, p=0.001) in the whole patient cohort. However, we identified a subgroup of patients with highly elevated baseline M-MDSC frequencies that fell below a defined cut-off during therapy and found that these patients had a longer OS that was similar to that of patients with low baseline M-MDSC frequencies. Importantly, patients with high M-MDSC frequencies exhibited a skewed baseline distribution of certain other immune cells but these did not influence patient survival, illustrating the paramount utility of MDSC assessment. CONCLUSION We confirmed that in general, highly elevated frequencies of peripheral M-MDSC are associated with poorer outcomes of ICB in metastatic melanoma. However, one reason for an imperfect correlation between high baseline MDSCs and outcome for individual patients may be the subgroup of patients identified here, with rapidly decreasing M-MDSCs on therapy, in whom the negative effect of high M-MDSC frequencies was lost. These findings might contribute to developing more reliable predictors of late-stage melanoma response to ICB at the individual patient level. A multifactorial model seeking such markers yielded only MDSC behavior and serum lactate dehydrogenase as predictors of treatment outcome.
Collapse
Affiliation(s)
- Andrea Gaißler
- Department of Dermatology, University Hospital Tübingen, Eberhard Karls University of Tübingen, Tübingen, Germany
- Internal Medicine I, University Hospital Tübingen, Eberhard Karls University of Tübingen, Tübingen, Germany
| | - Jonas Bochem
- Department of Dermatology, University Hospital Tübingen, Eberhard Karls University of Tübingen, Tübingen, Germany
- Internal Medicine I, University Hospital Tübingen, Eberhard Karls University of Tübingen, Tübingen, Germany
| | - Janine Spreuer
- Department of Dermatology, University Hospital Tübingen, Eberhard Karls University of Tübingen, Tübingen, Germany
- Internal Medicine I, University Hospital Tübingen, Eberhard Karls University of Tübingen, Tübingen, Germany
| | - Shannon Ottmann
- Department of Dermatology, University Hospital Tübingen, Eberhard Karls University of Tübingen, Tübingen, Germany
| | - Alexander Martens
- Department of Dermatology, University Hospital Tübingen, Eberhard Karls University of Tübingen, Tübingen, Germany
| | - Teresa Amaral
- Department of Dermatology, University Hospital Tübingen, Eberhard Karls University of Tübingen, Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) "Image Guided and Functionally Instructed Tumor Therapies", Tübingen, Germany
| | - Nikolaus Benjamin Wagner
- Department of Dermatology, University Hospital Tübingen, Eberhard Karls University of Tübingen, Tübingen, Germany
- Department of Dermatology, Venereology and Allergology, Kantonsspital St Gallen, Sankt Gallen, Switzerland
| | - Manfred Claassen
- Internal Medicine I, University Hospital Tübingen, Eberhard Karls University of Tübingen, Tübingen, Germany
- Department of Computer Science, Eberhard Karls University of Tübingen, Tübingen, Germany
| | - Friedegund Meier
- Skin Cancer Center at the University Cancer Centre and National Center for Tumor Diseases Dresden; Department of Dermatology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | | | - Claus Garbe
- Department of Dermatology, University Hospital Tübingen, Eberhard Karls University of Tübingen, Tübingen, Germany
| | - Thomas Eigentler
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Dermatology, Venereology and Allergology, Berlin, Germany
| | - Benjamin Weide
- Department of Dermatology, University Hospital Tübingen, Eberhard Karls University of Tübingen, Tübingen, Germany
| | - Graham Pawelec
- Department of Immunology, Interfaculty Institute for Cell Biology, Eberhard Karls University Tübingen, Tübingen, Germany
- Health Sciences North Research Institute, Sudbury, Ontario, Canada
| | - Kilian Wistuba-Hamprecht
- Department of Dermatology, University Hospital Tübingen, Eberhard Karls University of Tübingen, Tübingen, Germany
- Internal Medicine I, University Hospital Tübingen, Eberhard Karls University of Tübingen, Tübingen, Germany
- Department of Immunology, Interfaculty Institute for Cell Biology, Eberhard Karls University Tübingen, Tübingen, Germany
| |
Collapse
|
10
|
Zhang C, Xu L, Ma Y, Huang Y, Zhou L, Le H, Chen Z. Increased TIM-3 expression in tumor-associated macrophages predicts a poorer prognosis in non-small cell lung cancer: a retrospective cohort study. J Thorac Dis 2023; 15:1433-1444. [PMID: 37065598 PMCID: PMC10089863 DOI: 10.21037/jtd-23-227] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Accepted: 03/22/2023] [Indexed: 03/29/2023]
Abstract
Background T-cell immunoglobulin and mucin domain-containing molecule 3 (TIM-3) is considered a key negative regulator in T-cell-mediated response. However, few studies have been reported on the relationship between TIM-3 expression in tumor-associated macrophages (TAMs) and clinicopathological characteristics of patients. This study evaluated the correlation between the expression of TIM-3 on the surface of TAMs macrophages in tumor matrix and the clinical outcome of patients with non-small cell lung cancer (NSCLC). Methods The expression of CD68, CD163 and TIM-3 in 248 NSCLC patients who underwent surgery in Zhoushan Hospital from January 2010 to January 2013 was detected by immunohistochemistry (IHC). From the date of operation to the date of death, overall survival (OS) was measured to analyze the relationship between the expression of Tim-3 and the prognosis of NSCLC patients. Results The study assessed 248 patients with NSCLC. TIM-3 expression in TAMs was more frequently identified in patients with higher carcinoembryonic antigen (CEA) levels, lymph node metastasis, higher grade, high CD68 expression, and high CD163 expression (P<0.05). The OS of the high TIM-3 expression groups was shorter than that of the low TIM-3 expression groups (P=0.01). Patients with high TIM-3 and CD68/CD163 expressions had the worst prognosis, whereas patients with low expressions of both TIM-3 and CD68/CD163 had the best prognosis (P<0.05). In NSCLC, the OS of the high TIM-3 expression groups was shorter than that of the low TIM-3 expression groups (P=0.01). In lung adenocarcinoma, the OS of the high TIM-3 expression groups was shorter than that of the low TIM-3 expression groups(P=0.03). Conclusions TIM-3 expression in TAMs may be a promising prognostic biomarker for NSCLC or adenocarcinoma. Our results demonstrated that high TIM-3 expression in TAMs was an independent predictor of worse prognosis in patients.
Collapse
Affiliation(s)
- Chi Zhang
- Department of Cell Molecular Biology Laboratory, Zhoushan Hospital, Zhoushan, China
- Department of Clinical Medicine, Jiamusi University, Jiamusi, China
| | - Liyun Xu
- Department of Cell Molecular Biology Laboratory, Zhoushan Hospital, Zhoushan, China
| | - Yongbin Ma
- Department of Cell Molecular Biology Laboratory, Zhoushan Hospital, Zhoushan, China
| | - Yanyan Huang
- Department of Cell Molecular Biology Laboratory, Zhoushan Hospital, Zhoushan, China
| | - Lu Zhou
- Department of Cell Molecular Biology Laboratory, Zhoushan Hospital, Zhoushan, China
| | - Hanbo Le
- Department of Cardiothoracic Surgery, Zhoushan Hospital, Zhoushan, China
| | - Zhijun Chen
- Department of Clinical Medicine, Jiamusi University, Jiamusi, China
- Department of Cardiothoracic Surgery, Zhoushan Hospital, Zhoushan, China
| |
Collapse
|
11
|
Wu B, Shi X, Jiang M, Liu H. Cross-talk between cancer stem cells and immune cells: potential therapeutic targets in the tumor immune microenvironment. Mol Cancer 2023; 22:38. [PMID: 36810098 PMCID: PMC9942413 DOI: 10.1186/s12943-023-01748-4] [Citation(s) in RCA: 84] [Impact Index Per Article: 42.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 02/15/2023] [Indexed: 02/24/2023] Open
Abstract
Ongoing research has revealed that the existence of cancer stem cells (CSCs) is one of the biggest obstacles in the current cancer therapy. CSCs make an influential function in tumor progression, recurrence and chemoresistance due to their typical stemness characteristics. CSCs are preferentially distributed in niches, and those niche sites exhibit characteristics typical of the tumor microenvironment (TME). The complex interactions between CSCs and TME illustrate these synergistic effects. The phenotypic heterogeneity within CSCs and the spatial interactions with the surrounding tumor microenvironment led to increased therapeutic challenges. CSCs interact with immune cells to protect themselves against immune clearance by exploiting the immunosuppressive function of multiple immune checkpoint molecules. CSCs also can protect themselves against immune surveillance by excreting extracellular vesicles (EVs), growth factors, metabolites and cytokines into the TME, thereby modulating the composition of the TME. Therefore, these interactions are also being considered for the therapeutic development of anti-tumor agents. We discuss here the immune molecular mechanisms of CSCs and comprehensively review the interplay between CSCs and the immune system. Thus, studies on this topic seem to provide novel ideas for reinvigorating therapeutic approaches to cancer.
Collapse
Affiliation(s)
- Bo Wu
- grid.459742.90000 0004 1798 5889Department of General Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, 110042 China
| | - Xiang Shi
- grid.459742.90000 0004 1798 5889Department of Thoracic Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, 110042 China
| | - Meixi Jiang
- grid.412644.10000 0004 5909 0696Department of Neurology, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032 China
| | - Hongxu Liu
- Department of Thoracic Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, 110042, China.
| |
Collapse
|
12
|
Nazeri SA, Rezayat SM, Amani A, Hadjati J, Partoazar A, Zamani P, Mashreghi M, Jaafari MR. A novel formulation of cyclosporine A/phosphatidylserine-containing liposome using remote loading method: Potential product for immunosuppressive effects. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.103902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
13
|
Todosenko N, Yurova K, Khaziakhmatova O, Malashchenko V, Khlusov I, Litvinova L. Heparin and Heparin-Based Drug Delivery Systems: Pleiotropic Molecular Effects at Multiple Drug Resistance of Osteosarcoma and Immune Cells. Pharmaceutics 2022; 14:pharmaceutics14102181. [PMID: 36297616 PMCID: PMC9612132 DOI: 10.3390/pharmaceutics14102181] [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: 07/28/2022] [Revised: 09/29/2022] [Accepted: 10/10/2022] [Indexed: 11/23/2022] Open
Abstract
One of the main problems of modern health care is the growing number of oncological diseases both in the elderly and young population. Inadequately effective chemotherapy, which remains the main method of cancer control, is largely associated with the emergence of multidrug resistance in tumor cells. The search for new solutions to overcome the resistance of malignant cells to pharmacological agents is being actively pursued. Another serious problem is immunosuppression caused both by the tumor cells themselves and by antitumor drugs. Of great interest in this context is heparin, a biomolecule belonging to the class of glycosaminoglycans and possessing a broad spectrum of biological activity, including immunomodulatory and antitumor properties. In the context of the rapid development of the new field of “osteoimmunology,” which focuses on the collaboration of bone and immune cells, heparin and delivery systems based on it may be of intriguing importance for the oncotherapy of malignant bone tumors. Osteosarcoma is a rare but highly aggressive, chemoresistant malignant tumor that affects young adults and is characterized by constant recurrence and metastasis. This review describes the direct and immune-mediated regulatory effects of heparin and drug delivery systems based on it on the molecular mechanisms of (multiple) drug resistance in (onco) pathological conditions of bone tissue, especially osteosarcoma.
Collapse
Affiliation(s)
- Natalia Todosenko
- Center for Immunology and Cellular Biotechnology, Immanuel Kant Baltic Federal University, 236001 Kaliningrad, Russia
| | - Kristina Yurova
- Center for Immunology and Cellular Biotechnology, Immanuel Kant Baltic Federal University, 236001 Kaliningrad, Russia
| | - Olga Khaziakhmatova
- Center for Immunology and Cellular Biotechnology, Immanuel Kant Baltic Federal University, 236001 Kaliningrad, Russia
| | - Vladimir Malashchenko
- Center for Immunology and Cellular Biotechnology, Immanuel Kant Baltic Federal University, 236001 Kaliningrad, Russia
| | - Igor Khlusov
- Department of Morphology and General Pathology, Siberian State Medical University, 634050 Tomsk, Russia
| | - Larisa Litvinova
- Center for Immunology and Cellular Biotechnology, Immanuel Kant Baltic Federal University, 236001 Kaliningrad, Russia
- Correspondence:
| |
Collapse
|
14
|
Regulatory T cells induce a suppressive immune milieu and promote lymph node metastasis in intrahepatic cholangiocarcinoma. Br J Cancer 2022; 127:757-765. [PMID: 35597869 PMCID: PMC9381563 DOI: 10.1038/s41416-022-01838-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 04/24/2022] [Accepted: 04/27/2022] [Indexed: 12/04/2022] Open
Abstract
Background Emerging evidence indicates that immunogenicity plays an important role in intrahepatic cholangiocarcinoma (ICC). Herein, we systematically evaluated the clinical relevance of immunogenicity in ICC. Methods Highly immunogenic ICCs identified in the public dataset and the Cancer Immunome Atlas (TCIA) were assessed to determine the prognostic impact of immunogenicity in ICC and key components after curative resection. We also investigated the clinical relevance of the immune milieu in ICC. Results Using the Gene Expression Omnibus dataset 89749 and TCIA, we identified CD8+/forkhead box P3 (FoxP3)+ tumour-infiltrating lymphocytes (TILs), T-cell immunoglobulin and mucin domain 3 (TIM-3) and human leukocyte antigen-A (HLA-A) in highly immunogenic ICCs. Immunohistochemical analysis of the in-house cohort showed that intratumoral FoxP3+ TILs correlated with CD8+ TILs (P = 0.045, Fisher’s exact test) and that high FoxP3+/CD8+ ratio (FCR) was an important marker for poor survival (P < 0.001, log-rank test). Furthermore, the FCR was higher in tumour-free lymph nodes in ICCs with lymph node metastases than in those without lymph node metastases (P = 0.003, Mann–Whitney U test). Conclusions FCR should be considered an important biomarker that represents the immune environment of ICC based on its potentially important role in tumour progression, especially lymph node metastasis.
Collapse
|
15
|
Checkpoints and Immunity in Cancers: Role of GNG12. Pharmacol Res 2022; 180:106242. [DOI: 10.1016/j.phrs.2022.106242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 04/25/2022] [Accepted: 04/28/2022] [Indexed: 12/24/2022]
|
16
|
Abbaszadegan MR, Mojarrad M, Rahimi HR, Moghbeli M. Genetic and molecular biology of gastric cancer among Iranian patients: an update. EGYPTIAN JOURNAL OF MEDICAL HUMAN GENETICS 2022. [DOI: 10.1186/s43042-022-00232-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Abstract
Background
There is a declining trend of gastric cancer (GC) incidence in the world during recent years that is related to the development of novel diagnostic methods. However, there is still a high ratio of GC mortality among the Iranian population that can be associated with late diagnosis. Despite various reports about the novel diagnostic markers, there is not any general and standard diagnostic panel marker for Iranian GC patients. Therefore, it is required to determine an efficient and general panel of molecular markers for early detection.
Main body of the abstract
In the present review, we summarized all of the reported markers until now among Iranian GC patients to pave the way for the determination of a population-based diagnostic panel of markers. In this regard, we categorized these markers in different groups based on their involved processes to know which molecular process is more frequent during the GC progression among Iranians.
Conclusion
We observed that the non-coding RNAs are the main factors involved in GC tumorigenesis in this population.
Collapse
|
17
|
Chen X, Zhang Y, Wang F, Zhou X, Fu Q, Yang X, Lin J, Jin X. A Novel Assessment Model Based on Molecular Subtypes of Hypoxia-Related LncRNAs for Prognosis of Bladder Cancer. Front Cell Dev Biol 2021; 9:718991. [PMID: 34869309 PMCID: PMC8634255 DOI: 10.3389/fcell.2021.718991] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 10/11/2021] [Indexed: 12/19/2022] Open
Abstract
Hypoxia is a common feature in various tumors that regulates aggressiveness. Previous studies have demonstrated that some dysregulated long non-coding RNAs (lncRNAs) are correlated with tumor progression, including bladder cancer (BCa). However, the prognostic effect of hypoxia-related lncRNAs (HRLs) and their clinical relevance, as well as their regulatory effect on the tumor immune microenvironment, are largely unknown in BCa. A co-expression analysis between hypoxia genes and lncRNA expression, which was downloaded from the TCGA database, was performed to identify HRLs. Univariate Cox regression analysis was performed to select the most desirable lncRNAs for molecular subtype, and further LASSO analysis was performed to develop a prognostic model. This molecular subtype based on four HRLs (AC104653, AL136084, AL139393, and LINC00892) showed good performance in the tumor microenvironment and tumor mutation burden. The prognostic risk model suggested better performance in predicting BCa patients' prognosis and obtained a close correlation with clinicopathologic features. Furthermore, four of five first-line clinical chemotherapies showed different sensitivities to this model, and nine immune checkpoints showed different expression in the molecular subtypes or the risk model. In conclusion, this study indicates that this molecular subtype and risk model based on HRLs may be useful in improving the prognostic prediction of BCa patients with different clinical situations and may help to find a useful target for tumor therapy.
Collapse
Affiliation(s)
- Xianwu Chen
- Department of Urology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yan Zhang
- Department of Urology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Feifan Wang
- Department of Urology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xuejian Zhou
- Department of Urology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Qinghe Fu
- Department of Urology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xintao Yang
- Department of Urology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Juntao Lin
- Department of Urology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaodong Jin
- Department of Urology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
18
|
Makaremi S, Asadzadeh Z, Hemmat N, Baghbanzadeh A, Sgambato A, Ghorbaninezhad F, Safarpour H, Argentiero A, Brunetti O, Bernardini R, Silvestris N, Baradaran B. Immune Checkpoint Inhibitors in Colorectal Cancer: Challenges and Future Prospects. Biomedicines 2021; 9:1075. [PMID: 34572263 PMCID: PMC8467932 DOI: 10.3390/biomedicines9091075] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 08/19/2021] [Accepted: 08/19/2021] [Indexed: 12/12/2022] Open
Abstract
Immunotherapy is a new pillar of cancer therapy that provides novel opportunities to treat solid tumors. In this context, the development of new drugs targeting immune checkpoints is considered a promising approach in colorectal cancer (CRC) treatment because it can be induce specific and durable anti-cancer effects. Despite many advances in the immunotherapy of CRC, there are still limitations and obstacles to successful treatment. The immunosuppressive function of the tumor microenvironment (TME) is one of the causes of poor response to treatment in CRC patients. For this reason, checkpoint-blocking antibodies have shown promising outcomes in CRC patients by blocking inhibitory immune checkpoints and enhancing immune responses against tumors. This review summarizes recent advances in immune checkpoint inhibitors (ICIs), such as CTLA-4, PD-1, PD-L1, LAG-3, and TIM-3 in CRC, and it discusses various therapeutic strategies with ICIs, including the double blockade of ICIs, combination therapy of ICIs with other immunotherapies, and conventional treatments. This review also delineates a new hopeful path in the combination of anti-PD-1/anti-PD-L1 with other ICIs such as anti-CTLA-4, anti-LAG-3, and anti-TIM-3 for CRC treatment.
Collapse
Affiliation(s)
- Shima Makaremi
- Department of Immunology & Microbiology, School of Medicine, Arak University of Medical Sciences, Arak 3848176941, Iran;
| | - Zahra Asadzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 5166/15731, Iran; (Z.A.); (N.H.); (A.B.); (F.G.)
| | - Nima Hemmat
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 5166/15731, Iran; (Z.A.); (N.H.); (A.B.); (F.G.)
| | - Amir Baghbanzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 5166/15731, Iran; (Z.A.); (N.H.); (A.B.); (F.G.)
| | - Alessandro Sgambato
- Istituto di Ricovero e Cura a Carattere Scientifico Centro di Riferimento Oncologico della Basilicata (IRCCS-CROB), 5972362 Rome, Italy;
- Area of Pathology, Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario A. Gemelli-IRCCS, 5972362 Rome, Italy
| | - Farid Ghorbaninezhad
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 5166/15731, Iran; (Z.A.); (N.H.); (A.B.); (F.G.)
- Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz 5166/15731, Iran
| | - Hossein Safarpour
- Cellular & Molecular Research Center, Birjand University of Medical Sciences, Birjand 9717853577, Iran;
| | - Antonella Argentiero
- IRCCS Istituto Tumori “Giovanni Paolo II” of Bari, 70124 Bari, Italy; (A.A.); (O.B.)
| | - Oronzo Brunetti
- IRCCS Istituto Tumori “Giovanni Paolo II” of Bari, 70124 Bari, Italy; (A.A.); (O.B.)
| | - Renato Bernardini
- Department of Biomedical and Biotechnological Sciences, University of Catania, Via S. Sofia 97, 95121 Catania, Italy;
| | - Nicola Silvestris
- IRCCS Istituto Tumori “Giovanni Paolo II” of Bari, 70124 Bari, Italy; (A.A.); (O.B.)
- Department of Biomedical Sciences and Human Oncology (DIMO), University of Bari, 70124 Bari, Italy
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 5166/15731, Iran; (Z.A.); (N.H.); (A.B.); (F.G.)
- Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz 5166/15731, Iran
- Pharmaceutical Analysis Research Center, Tabriz University of Medical Sciences, Tabriz 5166/15731, Iran
| |
Collapse
|
19
|
Files JK, Sarkar S, Fram TR, Boppana S, Sterrett S, Qin K, Bansal A, Long DM, Sabbaj S, Kobie JJ, Goepfert PA, Erdmann N. Duration of post-COVID-19 symptoms is associated with sustained SARS-CoV-2-specific immune responses. JCI Insight 2021; 6:151544. [PMID: 34143754 PMCID: PMC8410022 DOI: 10.1172/jci.insight.151544] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Accepted: 06/17/2021] [Indexed: 11/17/2022] Open
Abstract
A subset of COVID-19 patients exhibit post-acute sequelae of COVID-19 (PASC), but little is known about the immune signatures associated with these syndromes. We investigated longitudinal peripheral blood samples in 50 individuals with previously confirmed SARS-CoV-2 infection, including 20 who experienced prolonged duration of COVID-19 symptoms (lasting more than 30 days; median = 74 days) compared with 30 who had symptom resolution within 20 days. Individuals with prolonged symptom duration maintained antigen-specific T cell response magnitudes to SARS-CoV-2 spike protein in CD4+ and circulating T follicular helper cell populations during late convalescence, while those without persistent symptoms demonstrated an expected decline. The prolonged group also displayed increased IgG avidity to SARS-CoV-2 spike protein. Significant correlations between symptom duration and both SARS-CoV-2-specific T cells and antibodies were observed. Activation and exhaustion markers were evaluated in multiple immune cell types, revealing few phenotypic differences between prolonged and recovered groups, suggesting that prolonged symptom duration is not due to persistent systemic inflammation. These findings demonstrate that SARS-CoV-2-specific immune responses are maintained in patients suffering from prolonged post-COVID-19 symptom duration in contrast to those with resolved symptoms and may suggest the persistence of viral antigens as an underlying etiology.
Collapse
Affiliation(s)
- Jacob K. Files
- Division of Infectious Diseases, Department of Medicine, School of Medicine
| | - Sanghita Sarkar
- Division of Infectious Diseases, Department of Medicine, School of Medicine
| | - Tim R. Fram
- Division of Infectious Diseases, Department of Medicine, School of Medicine
| | - Sushma Boppana
- Division of Infectious Diseases, Department of Medicine, School of Medicine
| | - Sarah Sterrett
- Division of Infectious Diseases, Department of Medicine, School of Medicine
| | - Kai Qin
- Division of Infectious Diseases, Department of Medicine, School of Medicine
| | - Anju Bansal
- Division of Infectious Diseases, Department of Medicine, School of Medicine
| | - Dustin M. Long
- Department of Biostatistics, School of Public Health, and
| | - Steffanie Sabbaj
- Division of Infectious Diseases, Department of Medicine, School of Medicine
- Department of Microbiology, School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - James J. Kobie
- Division of Infectious Diseases, Department of Medicine, School of Medicine
| | - Paul A. Goepfert
- Division of Infectious Diseases, Department of Medicine, School of Medicine
- Department of Microbiology, School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Nathan Erdmann
- Division of Infectious Diseases, Department of Medicine, School of Medicine
| |
Collapse
|
20
|
Kataoka S, Manandhar P, Lee J, Workman CJ, Banerjee H, Szymczak-Workman AL, Kvorjak M, Lohmueller J, Kane LP. The costimulatory activity of Tim-3 requires Akt and MAPK signaling and its recruitment to the immune synapse. Sci Signal 2021; 14:eaba0717. [PMID: 34131021 PMCID: PMC9741863 DOI: 10.1126/scisignal.aba0717] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Expression of the transmembrane protein Tim-3 is increased on dysregulated T cells undergoing chronic activation, including during chronic infection and in solid tumors. Thus, Tim-3 is generally thought of as an inhibitory protein. We and others previously reported that under some circumstances, Tim-3 exerts paradoxical costimulatory activity in T cells (and other cells), including enhancement of the phosphorylation of ribosomal S6 protein. Here, we examined the upstream signaling pathways that control Tim-3-mediated increases in phosphorylated S6 in T cells. We also defined the localization of Tim-3 relative to the T cell immune synapse and its effects on downstream signaling. Recruitment of Tim-3 to the immune synapse was mediated exclusively by the transmembrane domain, replacement of which impaired the ability of Tim-3 to costimulate T cell receptor (TCR)-dependent S6 phosphorylation. Furthermore, enforced localization of the Tim-3 cytoplasmic domain to the immune synapse in a chimeric antigen receptor still enabled T cell activation. Together, our findings are consistent with a model whereby Tim-3 enhances TCR-proximal signaling under acute conditions.
Collapse
Affiliation(s)
- Shunsuke Kataoka
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA 15261, USA
- Asahi Kasei Pharma Corporation, Shizuoka, Japan
- Graduate Program in Microbiology and Immunology, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Priyanka Manandhar
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA 15261, USA
- Graduate Program in Microbiology and Immunology, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Judong Lee
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Creg J Workman
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Hridesh Banerjee
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | | | - Michael Kvorjak
- Department of Surgery, Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Jason Lohmueller
- Department of Surgery, Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Lawrence P Kane
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA 15261, USA.
| |
Collapse
|
21
|
Tsotridou E, Vasileiou E, Mantadakis E, Tragiannidis A. Safety and Efficacy of Immune Checkpoint Inhibitors in Children and Young Adults with Haematological Malignancies: Review and Future Perspectives. Cardiovasc Hematol Agents Med Chem 2021; 20:20-33. [PMID: 33970848 DOI: 10.2174/1871525719666210510171132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Revised: 03/18/2021] [Accepted: 03/29/2021] [Indexed: 11/22/2022]
Abstract
Despite the marked improvement in overall survival rates of paediatric patients with haematological malignancies that has been achieved during the last decades, there is still a pressing need for novel therapeutic approaches for the subset of patients with relapsed or refractory disease. Immune checkpoint inhibitors aim to induce potent anti-tumour immune responses by targeted blockade of inhibitory receptors and have shown great promise in preclinical models and studies in the adult population. However, paediatric malignancies present unique features and so far, experience with these agents remains limited. In the current review we present an overview of efficacy and safety data from case reports, case series and clinical trials employing the use of immune checkpoint inhibitors in children, adolescents and young adults with haematological malignancies. We also discuss new possibilities involving novel targets and combination treatments and provide a summary of the currently registered clinical trials.
Collapse
Affiliation(s)
- Eleni Tsotridou
- 2nd Department of Paediatrics, Childhood and Adolescent Hematology Oncology Unit, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, AHEPA Hospital, Thessaloniki, Greece
| | - Eleni Vasileiou
- 2nd Department of Paediatrics, Childhood and Adolescent Hematology Oncology Unit, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, AHEPA Hospital, Thessaloniki, Greece
| | - Elpis Mantadakis
- Department of Paediatrics, Paediatric Hematology/Oncology Unit, Democritus University of Thrace Faculty of Medicine, Alexandroupolis, Thrace, Greece
| | - Athanasios Tragiannidis
- 2nd Department of Paediatrics, Childhood and Adolescent Hematology Oncology Unit, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, AHEPA Hospital, Thessaloniki, Greece
| |
Collapse
|
22
|
Gong L, Kwong DLW, Dai W, Wu P, Li S, Yan Q, Zhang Y, Zhang B, Fang X, Liu L, Luo M, Liu B, Chow LKY, Chen Q, Huang J, Lee VHF, Lam KO, Lo AWI, Chen Z, Wang Y, Lee AWM, Guan XY. Comprehensive single-cell sequencing reveals the stromal dynamics and tumor-specific characteristics in the microenvironment of nasopharyngeal carcinoma. Nat Commun 2021; 12:1540. [PMID: 33750785 PMCID: PMC7943808 DOI: 10.1038/s41467-021-21795-z] [Citation(s) in RCA: 119] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 02/08/2021] [Indexed: 12/19/2022] Open
Abstract
The tumor microenvironment (TME) of nasopharyngeal carcinoma (NPC) harbors a heterogeneous and dynamic stromal population. A comprehensive understanding of this tumor-specific ecosystem is necessary to enhance cancer diagnosis, therapeutics, and prognosis. However, recent advances based on bulk RNA sequencing remain insufficient to construct an in-depth landscape of infiltrating stromal cells in NPC. Here we apply single-cell RNA sequencing to 66,627 cells from 14 patients, integrated with clonotype identification on T and B cells. We identify and characterize five major stromal clusters and 36 distinct subpopulations based on genetic profiling. By comparing with the infiltrating cells in the non-malignant microenvironment, we report highly representative features in the TME, including phenotypic abundance, genetic alternations, immune dynamics, clonal expansion, developmental trajectory, and molecular interactions that profoundly influence patient prognosis and therapeutic outcome. The key findings are further independently validated in two single-cell RNA sequencing cohorts and two bulk RNA-sequencing cohorts. In the present study, we reveal the correlation between NPC-specific characteristics and progression-free survival. Together, these data facilitate the understanding of the stromal landscape and immune dynamics in NPC patients and provides deeper insights into the development of prognostic biomarkers and therapeutic targets in the TME.
Collapse
Affiliation(s)
- Lanqi Gong
- Department of Clinical Oncology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
- Department of Clinical Oncology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Dora Lai-Wan Kwong
- Department of Clinical Oncology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
- Department of Clinical Oncology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Wei Dai
- Department of Clinical Oncology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
- Department of Clinical Oncology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Pingan Wu
- Department of Surgery, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Shanshan Li
- Department of Clinical Oncology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Qian Yan
- Department of Clinical Oncology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
- Department of Clinical Oncology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Yu Zhang
- Department of Clinical Oncology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Baifeng Zhang
- Department of Clinical Oncology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Xiaona Fang
- Department of Clinical Oncology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Li Liu
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
- The AIDS Institute, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
- State Key Laboratory of Emerging Infectious Disease, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Min Luo
- Department of Clinical Oncology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Beilei Liu
- Department of Clinical Oncology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Larry Ka-Yue Chow
- Department of Clinical Oncology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Qingyun Chen
- State Key Laboratory of Oncology in Southern China, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Jinlin Huang
- Department of Clinical Oncology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Victor Ho-Fun Lee
- Department of Clinical Oncology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
- Department of Clinical Oncology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Ka-On Lam
- Department of Clinical Oncology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
- Department of Clinical Oncology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Anthony Wing-Ip Lo
- Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Zhiwei Chen
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
- The AIDS Institute, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
- State Key Laboratory of Emerging Infectious Disease, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Yan Wang
- Department of Pathology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Anne Wing-Mui Lee
- Department of Clinical Oncology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
- Department of Clinical Oncology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Xin-Yuan Guan
- Department of Clinical Oncology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China.
- Department of Clinical Oncology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China.
- State Key Laboratory of Oncology in Southern China, Sun Yat-sen University Cancer Center, Guangzhou, China.
| |
Collapse
|
23
|
Low Distribution of TIM-3 + Cytotoxic Tumor-Infiltrating Lymphocytes Predicts Poor Outcomes in Gastrointestinal Stromal Tumors. J Immunol Res 2021; 2021:6647292. [PMID: 33681387 PMCID: PMC7907748 DOI: 10.1155/2021/6647292] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Revised: 12/13/2020] [Accepted: 02/02/2021] [Indexed: 02/07/2023] Open
Abstract
There are multiple tumor-infiltrating lymphocytes (TILs) and relevant immune checkpoints existing in gastrointestinal stromal tumor (GIST), which provides opportunities and rationales for developing effective immunotherapies. Recent studies have suggested that checkpoint TIM-3/Gal-9 plays a pivotal role on immune response in multiple tumors, similar to the PD-1/PD-L1, emerging as a potential therapeutic target. However, their functions in GIST are unrevealed. Hence, the expression of immune checkpoints TIM-3 and Gal-9, as well as the infiltration of CD8+ T cells and NK cells, is described in 299 cases of GIST specimens. The results showed that TIM-3 and Gal-9 are mainly expressed in TILs, rarely in tumor cells. Expression levels of TIM-3 and Gal-9 significantly differ in varying risks of GIST and exert opposite distribution trends. Indicated by prognosis analysis, high TIM-3 expression of TILs was associated with improved outcome, while low expression levels of TIM-3 in combination with low amounts of CD8+ and CD56+ TILs predict extremely poor survival. The integrated analysis of TIM-3+, CD8+, and CD56+ TILs as one biomarker is a reliable independent predictor of prognosis. In conclusion, low densities of TIM-3+ TILs are associated with poor survival, and integrated immune biomarkers lead to superior predictors of GIST prognosis.
Collapse
|
24
|
Immunological status of peripheral blood is associated with prognosis in patients with bone and soft-tissue sarcoma. Oncol Lett 2021; 21:212. [PMID: 33510813 PMCID: PMC7836390 DOI: 10.3892/ol.2021.12473] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Accepted: 10/23/2020] [Indexed: 12/22/2022] Open
Abstract
Immune-checkpoint inhibitors have shown promising antitumor effects against certain types of cancer. However, specific immune-checkpoint inhibitors for patients with sarcoma have yet to be identified, whereas the immunological status of peripheral blood in patients with bone sarcoma and soft-tissue sarcoma (STS) remains unknown. In addition, it is unclear whether the immunological status from the peripheral blood could be used as a prognostic indicator. Therefore, the present study aimed to clarify the immunological status of peripheral blood samples derived from patients with bone sarcoma and STS. Immune monitoring was performed using the peripheral blood samples of 61 patients with no metastasis of high-grade sarcoma. A total of 25 patients with metastatic sarcoma were used for comparison. A total of 41 immune cell subsets were analyzed using multicolor-flow cytometry. The patients that did not have metastasis demonstrated higher quantities of monocytic myeloid-derived suppressor cells (M-MDSCs) and T cell immunoglobulin and mucin domain-3 (Tim-3)+ CD8+ T cells, which were significantly associated with poor disease-free survival (DFS) time, while higher quantities of NKG2D+ CD8+ T cells were significantly associated with improved DFS time. Multivariate Cox regression analysis demonstrated that the number of Tim-3+ CD8+ T cells was associated with lower DFS time. A significant association was also found between the number of M-MDSCs and progression-free survival (PFS) time in patients with metastasis. The results suggested the occurrence of immune surveillance, which indicated that the host immune reaction against cancer existed in patients with bone sarcoma and STS. Notably, a high number of M-MDSCs was associated with both DFS and PFS time, suggesting a strong prognostic value. The data suggested that the immune status of peripheral blood was associated with the prognosis in patients with sarcoma, as previously reported in patients with other cancer types. In summary, the results may assist with the development of novel strategies for sarcoma treatment, based on the use of biomarkers or immunotherapy.
Collapse
|
25
|
Files JK, Boppana S, Perez MD, Sarkar S, Lowman KE, Qin K, Sterrett S, Carlin E, Bansal A, Sabbaj S, Long DM, Kutsch O, Kobie J, Goepfert PA, Erdmann N. Sustained cellular immune dysregulation in individuals recovering from SARS-CoV-2 infection. J Clin Invest 2021; 131:140491. [PMID: 33119547 PMCID: PMC7773371 DOI: 10.1172/jci140491] [Citation(s) in RCA: 99] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 09/15/2020] [Indexed: 01/08/2023] Open
Abstract
SARS-CoV-2 causes a wide spectrum of clinical manifestations and significant mortality. Studies investigating underlying immune characteristics are needed to understand disease pathogenesis and inform vaccine design. In this study, we examined immune cell subsets in hospitalized and nonhospitalized individuals. In hospitalized patients, many adaptive and innate immune cells were decreased in frequency compared with those of healthy and convalescent individuals, with the exception of an increase in B lymphocytes. Our findings show increased frequencies of T cell activation markers (CD69, OX40, HLA-DR, and CD154) in hospitalized patients, with other T cell activation/exhaustion markers (PD-L1 and TIGIT) remaining elevated in hospitalized and nonhospitalized individuals. B cells had a similar pattern of activation/exhaustion, with increased frequency of CD69 and CD95 during hospitalization followed by an increase in PD1 frequencies in nonhospitalized individuals. Interestingly, many of these changes were found to increase over time in nonhospitalized longitudinal samples, suggesting a prolonged period of immune dysregulation after SARS-CoV-2 infection. Changes in T cell activation/exhaustion in nonhospitalized patients were found to positively correlate with age. Severely infected individuals had increased expression of activation and exhaustion markers. These data suggest a prolonged period of immune dysregulation after SARS-CoV-2 infection, highlighting the need for additional studies investigating immune dysregulation in convalescent individuals.
Collapse
Affiliation(s)
- Jacob K. Files
- Division of Infectious Diseases, Department of Medicine, School of Medicine
| | - Sushma Boppana
- Division of Infectious Diseases, Department of Medicine, School of Medicine
| | - Mildred D. Perez
- Division of Infectious Diseases, Department of Medicine, School of Medicine
| | - Sanghita Sarkar
- Division of Infectious Diseases, Department of Medicine, School of Medicine
| | - Kelsey E. Lowman
- Division of Infectious Diseases, Department of Medicine, School of Medicine
| | - Kai Qin
- Division of Infectious Diseases, Department of Medicine, School of Medicine
| | - Sarah Sterrett
- Division of Infectious Diseases, Department of Medicine, School of Medicine
| | - Eric Carlin
- Division of Infectious Diseases, Department of Medicine, School of Medicine
| | - Anju Bansal
- Division of Infectious Diseases, Department of Medicine, School of Medicine
| | - Steffanie Sabbaj
- Division of Infectious Diseases, Department of Medicine, School of Medicine
| | - Dustin M. Long
- Department of Biostatistics, School of Public Health, and
| | - Olaf Kutsch
- Division of Infectious Diseases, Department of Medicine, School of Medicine
- Department of Microbiology, School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - James Kobie
- Division of Infectious Diseases, Department of Medicine, School of Medicine
| | - Paul A. Goepfert
- Division of Infectious Diseases, Department of Medicine, School of Medicine
- Department of Microbiology, School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Nathan Erdmann
- Division of Infectious Diseases, Department of Medicine, School of Medicine
| |
Collapse
|
26
|
Cao Y, Wang X, Jin T, Tian Y, Dai C, Widarma C, Song R, Xu F. Immune checkpoint molecules in natural killer cells as potential targets for cancer immunotherapy. Signal Transduct Target Ther 2020; 5:250. [PMID: 33122640 PMCID: PMC7596531 DOI: 10.1038/s41392-020-00348-8] [Citation(s) in RCA: 92] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 09/13/2020] [Accepted: 09/21/2020] [Indexed: 12/12/2022] Open
Abstract
Recent studies have demonstrated the potential of natural killer (NK) cells in immunotherapy to treat multiple types of cancer. NK cells are innate lymphoid cells that play essential roles in tumor surveillance and control that efficiently kill the tumor and do not require the major histocompatibility complex. The discovery of the NK's potential as a promising therapeutic target for cancer is a relief to oncologists as they face the challenge of increased chemo-resistant cancers. NK cells show great potential against solid and hematologic tumors and have progressively shown promise as a therapeutic target for cancer immunotherapy. The effector role of these cells is reliant on the balance of inhibitory and activating signals. Understanding the role of various immune checkpoint molecules in the exhaustion and impairment of NK cells when their inhibitory receptors are excessively expressed is particularly important in cancer immunotherapy studies and clinical implementation. Emerging immune checkpoint receptors and molecules have been found to mediate NK cell dysfunction in the tumor microenvironment; this has brought up the need to explore further additional NK cell-related immune checkpoints that may be exploited to enhance the immune response to refractory cancers. Accordingly, this review will focus on the recent findings concerning the roles of immune checkpoint molecules and receptors in the regulation of NK cell function, as well as their potential application in tumor immunotherapy.
Collapse
Affiliation(s)
- Yuqing Cao
- Department of General Surgery, Shengjing Hospital of China Medical University, 110004, Shenyang, China
| | - Xiaoyu Wang
- College of Life and Health Science, Northeastern University, 110819, Shenyang, China
| | - Tianqiang Jin
- Department of General Surgery, Shengjing Hospital of China Medical University, 110004, Shenyang, China
| | - Yu Tian
- Department of General Surgery, Shengjing Hospital of China Medical University, 110004, Shenyang, China
| | - Chaoliu Dai
- Department of General Surgery, Shengjing Hospital of China Medical University, 110004, Shenyang, China
| | - Crystal Widarma
- Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, 92350, USA
| | - Rui Song
- Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, 92350, USA.
| | - Feng Xu
- Department of General Surgery, Shengjing Hospital of China Medical University, 110004, Shenyang, China.
| |
Collapse
|
27
|
Mechanisms of resistance to immune checkpoint inhibitors and strategies to reverse drug resistance in lung cancer. Chin Med J (Engl) 2020; 133:2444-2455. [PMID: 32969861 PMCID: PMC7575183 DOI: 10.1097/cm9.0000000000001124] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
In recent years, the research of immune checkpoint inhibitors has made a great breakthrough in lung cancer treatment. Currently, a variety of immune checkpoint inhibitors have been applied into clinical practice, including antibodies targeting the programmed cell death-1, programmed cell death-ligand 1, and cytotoxic T-lymphocyte antigen 4, and so on. However, not all patients can benefit from the treatment. Abnormal antigen presentation, functional gene mutation, tumor microenvironment, and other factors can lead to primary or secondary resistance. In this paper, we reviewed the molecular mechanism of immune checkpoint inhibitor resistance and various combination strategies to overcome resistance, in order to expand the beneficial population and enable precision medicine.
Collapse
|
28
|
De Sousa Linhares A, Kellner F, Jutz S, Zlabinger GJ, Gabius H, Huppa JB, Leitner J, Steinberger P. TIM-3 and CEACAM1 do not interact in cis and in trans. Eur J Immunol 2020; 50:1126-1141. [PMID: 32222966 PMCID: PMC7496933 DOI: 10.1002/eji.201948400] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Revised: 01/29/2020] [Accepted: 03/24/2020] [Indexed: 01/13/2023]
Abstract
TIM-3 has been considered as a target in cancer immunotherapy. In T cells, inhibitory as well as activating functions have been ascribed to this molecule. Its role may therefore depend on the state of T cells and on the presence of interaction partners capable to perform functional pairing. Carcinoembryonic antigen-related cell adhesion molecule (CEACAM1) has been proposed to bind TIM-3 and to regulate its function. Using a T cell reporter platform we confirmed CEACAM1-mediated inhibition, but CEACAM1 did not functionally engage TIM-3. TIM-3 and CEACAM1 coexpression was limited to a small subset of activated T cells. Moreover, results obtained in extensive binding studies were not in support of an interaction between TIM-3 and CEACAM1. Cytoplasmic sequences derived from TIM-3 induced inhibitory signaling in our human T cell reporter system. Our results indicate that TIM-3 functions are independent of CEACAM1 and that this receptor has the capability to promote inhibitory signaling pathways in human T cells.
Collapse
Affiliation(s)
- Annika De Sousa Linhares
- Division of Immune Receptors and T Cell ActivationCenter for PathophysiologyInfectiology, and ImmunologyInstitute of ImmunologyMedical University of ViennaViennaAustria
| | - Florian Kellner
- Institute for Hygiene and Applied ImmunologyCenter for PathophysiologyInfectiology and ImmunologyMedical University of ViennaViennaAustria
| | - Sabrina Jutz
- Division of Immune Receptors and T Cell ActivationCenter for PathophysiologyInfectiology, and ImmunologyInstitute of ImmunologyMedical University of ViennaViennaAustria
| | - Gerhard J. Zlabinger
- Division of Clinical and Experimental ImmunologyCenter for Pathophysiology, Infectiology, and ImmunologyInstitute of ImmunologyMedical University of ViennaViennaAustria
| | - Hans‐Joachim Gabius
- Faculty of Veterinary MedicineInstitute for Physiological ChemistryLudwig‐Maximilians‐UniversityMunichGermany
| | - Johannes B. Huppa
- Institute for Hygiene and Applied ImmunologyCenter for PathophysiologyInfectiology and ImmunologyMedical University of ViennaViennaAustria
| | - Judith Leitner
- Division of Immune Receptors and T Cell ActivationCenter for PathophysiologyInfectiology, and ImmunologyInstitute of ImmunologyMedical University of ViennaViennaAustria
| | - Peter Steinberger
- Division of Immune Receptors and T Cell ActivationCenter for PathophysiologyInfectiology, and ImmunologyInstitute of ImmunologyMedical University of ViennaViennaAustria
| |
Collapse
|
29
|
Huang P, Lv C, Zhang C, Feng H, Li C, Zhang H, Zhao X, Li M. Expression and significance of T-cell immunoglobulin mucin molecule 3 and its ligand galectin-9 in patients with adenomyosis. Gynecol Endocrinol 2020; 36:605-610. [PMID: 32319832 DOI: 10.1080/09513590.2020.1754788] [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] [Indexed: 10/24/2022] Open
Abstract
The T cell immunoglobulin mucin molecule 3 (TIM-3), as a negative regulatory immune checkpoint, plays an important role in cellular immunity by binding to its ligand galectin-9 (Gal-9). Under abnormal conditions, this pathway can regulate the depletion of T cells and suppress the immune response. Abnormal expression of TIM-3 and Gal-9 has been confirmed in a variety of cancers, recurrent miscarriages, chronic infectious diseases and autoimmune diseases. More and more studies have shown that immune factors play an important role in the pathogenesis of adenomyosis. However, few studies have attempted to explore their expression in adenomyosis. The ectopic and eutopic endometrium were collected from 15 women with adenomyosis and 13 women without adenomyosis. TIM-3/Gal-9 expression in endometrial tissue was detected using immunohistochemistry, western blot analysis and real-time PCR. We observed TIM-3/Gal-9 expression in both eutopic and ectopic endometria, with elevated expression in adenomyosis. These data suggest that TIM-3/Gal-9 may be involved in the development and progression of adenomyosis.
Collapse
Affiliation(s)
- Pu Huang
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, People's Republic of China
| | - Chunzi Lv
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, People's Republic of China
| | - Cheng Zhang
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, People's Republic of China
| | - Hairong Feng
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, People's Republic of China
| | - Chunyan Li
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, People's Republic of China
| | - Hui Zhang
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, People's Republic of China
| | - Xingbo Zhao
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, People's Republic of China
| | - Mingjiang Li
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, People's Republic of China
| |
Collapse
|
30
|
Khosravi N, Mokhtarzadeh A, Baghbanzadeh A, Hajiasgharzadeh K, Shahgoli VK, Hemmat N, Safarzadeh E, Baradaran B. Immune checkpoints in tumor microenvironment and their relevance to the development of cancer stem cells. Life Sci 2020; 256:118005. [PMID: 32593711 DOI: 10.1016/j.lfs.2020.118005] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 06/14/2020] [Accepted: 06/22/2020] [Indexed: 02/06/2023]
Abstract
Cancer is the second cause of mortality in the world after cardiovascular disease. Various studies attribute the emergence of therapeutic resistance in tumors to the presence of cancer stem cells or cancer-initiating cells (CSC/CIC). These relatively rare cells because of their typical stemness features, are responsible for tumor cell progression and recurrence. Moreover, CSCs have immunomodulatory capabilities and through orchestrating, some immunological profiles can stay safe from host anticancer immunity, and provide immunotherapy resistance in cancer patients. Many studies have shown that CSCs by producing immune system inhibitory factors and interacting with immune checkpoint molecules like CD47, PDL-1, CTLA4, Tim3, and LAG3, are able to communicate with tumor microenvironment (TME) components and protect cancer cells from immune clearance. In this review, we summarize the CSCs immunological mechanisms and comprehensively discuss interactions between these cells and factors that are present in the TME to repress immune system responses and enhance tumor survival. Therefore, it seems that further studies on this topic will open new doors to improve the therapeutic approaches of malignant cancers.
Collapse
Affiliation(s)
- Neda Khosravi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ahad Mokhtarzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amir Baghbanzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Vahid Khaze Shahgoli
- Department of Cancer and Inflammation Research, Institute for Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Nima Hemmat
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Elham Safarzadeh
- Department of Microbiology & Immunology, Faculty of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran.
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
31
|
Hu S, Liu X, Li T, Li Z, Hu F. LAG3 (CD223) and autoimmunity: Emerging evidence. J Autoimmun 2020; 112:102504. [PMID: 32576412 DOI: 10.1016/j.jaut.2020.102504] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 06/07/2020] [Accepted: 06/10/2020] [Indexed: 12/31/2022]
Abstract
Immune checkpoint molecules play pivotal roles in maintaining the immune homeostasis. Targeting these molecules, such as the classical Cytotoxic T-Lymphocyte Antigen 4 (CTLA4) and Programmed Cell Death Protein 1 (PD1), achieves great success in treating cancers. However, not all the patients respond well. This urges the immunologists to identify novel immune checkpoint molecules. Lymphocyte activation gene-3 (LAG3; CD223) is a newly identified inhibitory receptor. It is expressed on a variety of immune cells, including CD4+ T cells, CD8+ T cells, Tregs, B cells, and NK cells. Its unique intracellular domains, signaling patterns as well as the striking synergy observed in its targeted therapy with anti-PD1 indicate the important role of LAG3 in maintaining immune tolerance. Currently, a variety of agents targeting LAG3 are in clinical trials, revealing great perspectives in the future immunotherapy. In this review, we briefly summarize the studies on LAG3, including its structure, isoforms, ligands, signaling, function, roles in multiple diseases, as well as the latest targeted therapeutic advances, with particular concern on the potential association of LAG3 with autoimmune diseases.
Collapse
Affiliation(s)
- Suiyuan Hu
- Department of Rheumatology and Immunology, Peking University People's Hospital & Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China
| | - Xu Liu
- Department of Rheumatology and Immunology, Peking University People's Hospital & Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China
| | - Tianding Li
- Software Center, Bank of China, Beijing, China
| | - Zhanguo Li
- Department of Rheumatology and Immunology, Peking University People's Hospital & Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China; State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China; Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| | - Fanlei Hu
- Department of Rheumatology and Immunology, Peking University People's Hospital & Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China; State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China.
| |
Collapse
|
32
|
Huang R, Meng T, Zhu R, Zhao L, Song D, Yin H, Huang Z, Cheng L, Zhang J. The Integrated Transcriptome Bioinformatics Analysis Identifies Key Genes and Cellular Components for Spinal Cord Injury-Related Neuropathic Pain. Front Bioeng Biotechnol 2020; 8:101. [PMID: 32140464 PMCID: PMC7042182 DOI: 10.3389/fbioe.2020.00101] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Accepted: 02/03/2020] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Spinal cord injury (SCI) is one of the most devastating diseases with a high incidence rate around the world. SCI-related neuropathic pain (NeP) is a common complication, whereas its pathomechanism is still unclear. The purpose of this study is to identify key genes and cellular components for SCI-related NeP by an integrated transcriptome bioinformatics analysis. METHODS The gene expression profile of 25 peripheral blood samples from chronic phase SCI patients (E-GEOD-69901) and 337 normal peripheral blood samples were downloaded from ArrayExpress and Genotype-Tissue Expression Portal (GTEx), respectively. A total of 3,368 normal peripheral blood mononuclear cells (PBMC) were download from Sequence Read Archive (SRA713577). Non-parametric tests were used to evaluate the association between all of differential expression genes (DEGs) and SCI-related NeP. CellPhoneDB algorithm was performed to identify the ligand-receptor interactions and their cellular localization among single PBMCs. Transcription factor (TF) enrichment analysis and Gene Set Variation Analysis (GSVA) were used to identify the potential upstream regulatory TFs and downstream signaling pathways, respectively. Co-expression analysis among significantly enriched TFs, key cellular communication genes and differentially expressed signaling pathways were performed to identify key genes and cellular components for SCI-related NeP. RESULTS A total of 2,314 genes were identified as DEGs between the experimental and the control group. Five proteins (ADRB2, LGALS9, PECAM1, HAVCR2, LRP1) were identified in the overlap of proteins in the significant ligand-receptor interactions of PBMCs and protein-protein interaction (PPI) network based on the DEGs. Only HAVCR2 was significantly associated with NeP (P = 0.005). Besides, the co-expression analysis revealed that TF YY1 had significantly co-expression pattern with cellular communication receptor HAVCR2 (R = -0.54, P < 0.001) in NK cells while HAVCR2 was also co-expressed with mTOR signaling pathway (R = 0.57, P < 0.001). The results of RT-qPCR and external dataset validation supported the signaling axis with the most significant co-expression patterns. CONCLUSION In peripheral blood of chronic SCI, HAVCR2 might act as a key receptor on the surface of NK cells and interact with ligand LGALS9 secreted by CD14+ monocytes, inhibiting NK cells through mTOR signaling pathway and ultimately predicting the occurrence of SCI-related NeP. This hypothetical signaling axis may provide prognostic biomarkers and therapeutic targets for SCI-related NeP.
Collapse
Affiliation(s)
- Runzhi Huang
- Division of Spine, Department of Orthopedics, Tongji Hospital Affiliated to Tongji University School of Medicine, Shanghai, China
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Orthopaedic Department of Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Tong Meng
- Division of Spine, Department of Orthopedics, Tongji Hospital Affiliated to Tongji University School of Medicine, Shanghai, China
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Orthopaedic Department of Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Rui Zhu
- Division of Spine, Department of Orthopedics, Tongji Hospital Affiliated to Tongji University School of Medicine, Shanghai, China
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Orthopaedic Department of Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Lijuan Zhao
- Division of Spine, Department of Orthopedics, Tongji Hospital Affiliated to Tongji University School of Medicine, Shanghai, China
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Orthopaedic Department of Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Dianwen Song
- Department of Orthopedics, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Huabin Yin
- Department of Orthopedics, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Zongqiang Huang
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Liming Cheng
- Division of Spine, Department of Orthopedics, Tongji Hospital Affiliated to Tongji University School of Medicine, Shanghai, China
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Orthopaedic Department of Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Jie Zhang
- Division of Spine, Department of Orthopedics, Tongji Hospital Affiliated to Tongji University School of Medicine, Shanghai, China
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Orthopaedic Department of Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
- Department of Prevention, Tongji University School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
33
|
Sun B, Liu M, Cui M, Li T. Granzyme B-expressing treg cells are enriched in colorectal cancer and present the potential to eliminate autologous T conventional cells. Immunol Lett 2019; 217:7-14. [PMID: 31669380 DOI: 10.1016/j.imlet.2019.10.007] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 09/11/2019] [Accepted: 10/05/2019] [Indexed: 12/20/2022]
Abstract
In addition to expressing inhibitory cytokines and suppressive molecules, Treg cells could downplay inflammation by releasing cytotoxic molecules and eliminating proinflammatory immune cells. Colorectal cancer (CRC) is a common malignancy that has led to many cancer-related deaths. In this study, we investigated the cytotoxic aspect of Treg cells in CRC patients. Data showed that tumor-infiltrating FOXP3+ Treg cells expressed granzyme B immediately following resection, indicating that granzyme B-expressing Treg cells were present directly ex vivo. In the tumor-associated lymph nodes (LNs) and circulating lymphocytes, however, granzyme B-expressing Treg cells were only scarcely found. We then attempted to stimulate granzyme B expression in circulating Treg cells. Granzyme B upregulation in Treg cells could not be activated by standard T cell receptor (TCR) activation through anti-CD3/CD28 and IL-2 but required stimulation with bacterial products, such as with heat-killed Staphylococcus aureus. Interestingly, granzyme B expression was highly concentrated in TIM-3+ Treg cells, a Treg subset previously shown to be enriched in the tumor microenvironment and presented increased suppressive capacity. These TIM-3+ Treg cells presented higher cytolytic capacity toward autologous T conventional cells than the TIM-3- Treg cells, in a manner that was dependent on granzyme B but not TIM-3. Overall, we found that granzyme B-expressing Treg cells were enriched in the tumors from CRC patients and had the potential to eliminate autologous T conventional cells.
Collapse
Affiliation(s)
- Bing Sun
- Department of Colorectal Surgery, Shandong Provincial Qianfoshan Hospital, The First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong, China
| | - Mingtao Liu
- Department of Colorectal Surgery, People's Hospital of Xiajin, Shandong, China
| | - Meng Cui
- Department of Colorectal Surgery, Shandong Provincial Qianfoshan Hospital, The First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong, China
| | - Tao Li
- Department of Colorectal Surgery, Shandong Provincial Qianfoshan Hospital, The First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong, China.
| |
Collapse
|
34
|
Park M, Kang KW. Phosphatidylserine receptor-targeting therapies for the treatment of cancer. Arch Pharm Res 2019; 42:617-628. [PMID: 31172440 DOI: 10.1007/s12272-019-01167-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Accepted: 06/01/2019] [Indexed: 02/07/2023]
Abstract
Asymmetric distribution of phospholipids across the plasma membrane is a unique characteristic of eukaryotic cells. Phosphatidylcholine and sphingomyelin are exposed in the outer leaflet, and phosphatidylserine (PS) is predominantly located in the inner leaflet. Redistribution of PS to the cell surface can be observed in several physiological conditions, such as apoptosis and platelet activation, or in pathological conditions, such as the release of microvesicles/exosomes from tumor tissues. PS binding to the phosphatidylserine receptor (PSR) on immune cells initiates immunosuppressive pathways that can lead to immune evasion by cancer cells. Conversely, PSR activation of cancer cells plays an important role in their survival, proliferation and metastasis. Herein, we briefly summarize both recent advances in our understanding of the pathological roles of PS and its receptor in cancer biology, as well as relevant pharmacological approaches.
Collapse
Affiliation(s)
- Miso Park
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - Keon Wook Kang
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, 08826, Republic of Korea.
| |
Collapse
|
35
|
Nibona E, Xu G, Wu K, Shen H, Zhang R, Ke X, Al Hafiz A, Wang Z, Qi C, Zhao H. Identification, characterization, expression profiles of OlHavcr2 in medaka (Oryzias latipes). Gen Comp Endocrinol 2019; 277:30-37. [PMID: 30395804 DOI: 10.1016/j.ygcen.2018.10.023] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 10/10/2018] [Accepted: 10/30/2018] [Indexed: 12/13/2022]
Abstract
Hepatitis A virus cellular receptor2 (Havcr2) also named T-cell immunoglobulin and mucin domain containing-3 (Tim-3) was initially described as a T helper 1-specific cell surface protein, a member of Tim family implicated in the regulating process of adaptive and innate immune responses. Here, medaka (Oryzias latipes) Havcr2 (OlHavcr2) was isolated and characterized. Unlike other Havcr2 proteins, OlHavcr2 possesses two Ig-like domains but lacks cytoplasmic and transmembrane domains. RT-PCR results revealed that OlHavcr2 mRNA was expressed strongly in the liver, moderately in the intestine, heart and ovary, and weakly in the muscle, gill, brain, eye, spleen, and testis. OlHavcr2 expression begun from gastrula stage and was maintained until hatching. The signal of OlHavcr2 was mainly identified in the blood system in the yolk sac by in situ hybridization. These results indicated that OlHavcr2 is expressed ubiquitously in adult tissues, and is a zygotic gene expressed from gastrula onwards in embryogenesis. OlHavcr2 may play a significant role in the blood system of medaka. In the immune organs, OlHavcr2 expression was affected by the immune stimulants, lipopolysaccharide and poly I:C, suggesting that OlHavcr2 was involved in innate immunity and adaptive immunity in medaka.
Collapse
Affiliation(s)
- Emile Nibona
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan 430079, Hubei, China
| | - Gongyu Xu
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan 430079, Hubei, China
| | - Kongyue Wu
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan 430079, Hubei, China
| | - Hao Shen
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan 430079, Hubei, China
| | - Runshuai Zhang
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan 430079, Hubei, China
| | - Xiaomei Ke
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan 430079, Hubei, China
| | - Abdullah Al Hafiz
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan 430079, Hubei, China
| | - Zequn Wang
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan 430079, Hubei, China
| | - Chao Qi
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan 430079, Hubei, China
| | - Haobin Zhao
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan 430079, Hubei, China.
| |
Collapse
|
36
|
Piancone F, Saresella M, Marventano I, La Rosa F, Caputo D, Mendozzi L, Rovaris M, Clerici M. A Deficit of CEACAM-1–Expressing T Lymphocytes Supports Inflammation in Primary Progressive Multiple Sclerosis. THE JOURNAL OF IMMUNOLOGY 2019; 203:76-83. [DOI: 10.4049/jimmunol.1801625] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Accepted: 04/30/2019] [Indexed: 11/19/2022]
|
37
|
Rotte A, Jin JY, Lemaire V. Mechanistic overview of immune checkpoints to support the rational design of their combinations in cancer immunotherapy. Ann Oncol 2019; 29:71-83. [PMID: 29069302 DOI: 10.1093/annonc/mdx686] [Citation(s) in RCA: 238] [Impact Index Per Article: 39.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Checkpoint receptor blockers, known to act by blocking the pathways that inhibit immune cell activation and stimulate immune responses against tumor cells, have been immensely successful in the treatment of cancer. Among several checkpoint receptors of immune cells, cytotoxic T-lymphocyte-associated protein-4 (CTLA-4), programmed cell death protein-1 (PD-1), T-cell immunoglobulin and ITIM domain (TIGIT), T-cell immunoglobulin-3 (TIM-3) and lymphocyte activation gene 3 (LAG-3) are the most commonly targeted checkpoints for cancer immunotherapy. Six drugs including one CTLA-4 blocker (ipilimumab), two PD-1 blockers (nivolumab and pembrolizumab) and three PD-L1 blockers (atezolizumab, avelumab and durvalumab) are approved for the treatment of different types of cancers including both solid tumors such as melanoma, lung cancer, head and neck cancer, bladder cancer and Merkel cell cancer as well as hematological tumors such as classic Hodgkin's lymphoma. The main problem with checkpoint blockers is that only a fraction of patients respond to the therapy. Insufficient immune activation is considered as one of the main reason for low response rates and combination of checkpoint blockers has been proposed to increase the response rates. The combination of checkpoint blockers was successful in melanoma but had significant adverse events. A combination that is selected based on the mechanistic differences between checkpoints and the differences in expression of checkpoints and their ligands in the tumor microenvironment could have a synergistic effect in a given cancer subtype and also have a manageable safety profile. This review aims to help in design of optimal checkpoint blocker combinations by discussing the mechanistic details and outlining the subtle differences between major checkpoints targeted for cancer immunotherapy.
Collapse
Affiliation(s)
- A Rotte
- Department of Clinical Pharmacology, Genentech Research and Early Development, South San Francisco, USA
| | - J Y Jin
- Department of Clinical Pharmacology, Genentech Research and Early Development, South San Francisco, USA
| | - V Lemaire
- Department of Clinical Pharmacology, Genentech Research and Early Development, South San Francisco, USA
| |
Collapse
|
38
|
Margolis N, Markovits E, Markel G. Reprogramming lymphocytes for the treatment of melanoma: From biology to therapy. Adv Drug Deliv Rev 2019; 141:104-124. [PMID: 31276707 DOI: 10.1016/j.addr.2019.06.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2019] [Revised: 05/31/2019] [Accepted: 06/24/2019] [Indexed: 12/15/2022]
Abstract
This decade has introduced drastic changes in melanoma therapy, predominantly due to the materialization of the long promise of immunotherapy. Cytotoxic T cells are the chief component of the immune system, which are targeted by different strategies aimed to increase their capacity against melanoma cells. To this end, reprogramming of T cells occurs by T cell centered manipulation, targeting the immunosuppressive tumor microenvironment or altering the whole patient. These are enabled by delivery of small molecules, functional monoclonal antibodies, different subunit vaccines, as well as living lymphocytes, native or genetically engineered. Current FDA-approved therapies are focused on direct T cell manipulation, such as immune checkpoint inhibitors blocking CTLA-4 and/or PD-1, which paves the way for an effective immunotherapy backbone available for combination with other modalities. Here we review the biology and clinical developments that enable melanoma immunotherapy today and in the future.
Collapse
|
39
|
Interaction of Breast Cancer and Insulin Resistance on PD1 and TIM3 Expression in Peripheral Blood CD8 T Cells. Pathol Oncol Res 2019; 25:1233-1243. [PMID: 30759303 DOI: 10.1007/s12253-019-00610-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Accepted: 01/23/2019] [Indexed: 12/18/2022]
Abstract
Epidemiological evidence points to a link between insulin resistance (IR) and breast cancer (BrCA). Insulin plays a role in CD8+ T cells (CD8T) differentiation and function and affects adipocytokines levels. CD8T activity in BrCA is associated with favorable outcome; while PD1 and TIM3 are markers of CD8T exhaustion and play critical roles in the negative regulation of T cell responses. Patients with (BrCA) have high expression levels of PD1 on circulating. Therefore, we hypothesized that BrCA and IR could affect PD1 and/or TIM3 expression on circulating CD8T. We determine PD1 and TIM3 expression on CD8T and analyze the relationship of CD8T phenotype with serum insulin and plasma adipocytokines levels in the different groups. We enrolled four groups of treatment-naive patients: women without neoplasms (Neo-)/without IR (IR-), Neo-/with IR (IR+), BrCa/IR- and BrCa/IR+. We found interactions between BrCA and IR with respect to TIM3 on naïve and central memory (CM) CD8T subsets. Furthermore, BrCA had a greater PD1 + TIM3- CD8T frequency in CD8T subsets than Neo-. IR+ presented a significantly lower PD1 + TIM3- frequency in CD8T subsets compare to Non-IR. In addition, we found a negative correlation between insulin levels, HOMA and frequency of PD1 + TIM3- in CD8T and a positive correlation between adiponectin levels and the frequency PD1 + TIM3- in CD8T. The increased expression of PD1 on different subsets of CD8T from BrCa patients is consistent with immunological tolerance, whereas IR has a contrary effect. IR could have a deleterious role in the activation of CD8T that can be relevant to new BrCa immunotherapy.
Collapse
|
40
|
Qin A, Zhong T, Zou H, Wan X, Yao B, Zheng X, Yin D. Critical role of Tim-3 mediated autophagy in chronic stress induced immunosuppression. Cell Biosci 2019; 9:13. [PMID: 30680089 PMCID: PMC6341633 DOI: 10.1186/s13578-019-0275-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Accepted: 01/09/2019] [Indexed: 01/02/2023] Open
Abstract
Background Psychological and physical stress can either enhance or suppress immune functions depending on a variety of factors such as duration and severity of stressful situation. Chronic stress exerts a significantly suppressive effect on immune functions. However, the mechanisms responsible for this phenomenon remain to be elucidated. Autophagy plays an essential role in modulating cellular homeostasis and immune responses. However, it is not known yet whether autophagy contributes to chronic stress-induced immunosuppression. T cell immunoglobulin and mucin domain 3 (Tim-3) has shown immune-suppressive effects and obviously positive regulation on cell apoptosis. Tim-3 combines with Tim-3 ligand galectin-9 to modulate apoptosis. However, its impact on autophagy and chronic stress-induced immunosuppression is not yet identified. Results We found remarkably higher autophagy level in the spleens of mice that were subjected to chronic restraint stress compared with the control group. We also found that inhibition of autophagy by the autophagy inhibitor 3-methyladenine (3-MA) significantly attenuated chronic stress-induced alterations of pro-inflammatory and anti-inflammatory cytokine levels. We further elucidated that 3-MA dramatically inhibited the reduction of lymphocyte numbers. Moreover, chronic stress dramatically enhanced the expression of Tim-3 and galectin-9. Inhibition of Tim-3 by small interfering RNA against Tim-3 significantly decreased the level of autophagy and immune suppression in isolated primary splenocytes from stressed mice. In addition, α-lactose, a blocker for the interaction of Tim-3 and galectin-9, also decreased the autophagy level and immune suppression. Conclusion Chronic stress induces autophagy, resulting with suppression of immune system. Tim-3 and galectin-9 play a crucial regulatory role in chronic stress-induced autophagy. These studies suggest that Tim-3 mediated autophagy may offer a novel therapeutic strategy against the deleterious effects of chronic stress on the immune system.
Collapse
Affiliation(s)
- Anna Qin
- 1Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410008 Hunan China
| | - Ting Zhong
- 1Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410008 Hunan China
| | - Huajiao Zou
- 1Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410008 Hunan China
| | - Xiaoya Wan
- 1Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410008 Hunan China
| | - Bifeng Yao
- 1Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410008 Hunan China
| | - Xinbin Zheng
- 1Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410008 Hunan China
| | - Deling Yin
- 1Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410008 Hunan China.,2Department of Internal Medicine, College of Medicine, East Tennessee State University, Johnson City, TN 37614 USA
| |
Collapse
|
41
|
Bhandaru M, Rotte A. Monoclonal Antibodies for the Treatment of Melanoma: Present and Future Strategies. Methods Mol Biol 2019; 1904:83-108. [PMID: 30539467 DOI: 10.1007/978-1-4939-8958-4_4] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Metastatic melanoma is a dreadful type of skin cancer arising due to uncontrolled proliferation of melanocytes. It has very poor prognosis, low 5-year survival rates and until recently there were only handful of treatment options for metastatic melanoma patients. The drugs that were approved for the treatment had low response rates and were associated with severe adverse events. With the introduction of monoclonal antibodies against inhibitory immune checkpoints the treatment landscape for metastatic melanoma has changed dramatically. Ipilimumab, the first monoclonal antibody to be approved for the treatment of metastatic melanoma, showed significant improvements in durable response rates in patients and paved the way for next class of monoclonal antibodies. Nivolumab and pembrolizumab, the anti-PD-1 antibodies that were approved 3-years after the approval of ipilimumab, had decent response rates, low relapse rates and showed manageable safety profile. Antibodies against ligands for PD-1 receptors were then developed to overcome the adverse effects of anti-PD-1 antibodies and combination of monoclonal antibodies (ipilimumab plus nivolumab) was tested to increase the response rates. Additional target receptors that regulate T cell activity were identified on T cells and monoclonal antibodies against potential targets such as TIGIT, TIM-3, and LAG-3 were developed. This chapter discusses the details of monoclonal antibodies used for the treatment of melanoma along with the ones that could be introduced in the near future with emphasis on mechanisms by which antibodies stimulate anti-tumor immune response and the specifics of target molecules of the antibodies.
Collapse
Affiliation(s)
- Madhuri Bhandaru
- Department of Dermatology and Skin Science, University of British Columbia, Vancouver, BC, Canada.
| | - Anand Rotte
- Department of Clinical Pharmacology, Genentech Research and Early Development, South San Francisco, CA, USA. .,Department of Clinical and Regulatory Affairs, Nevro Corp., Redwood City, CA, USA.
| |
Collapse
|
42
|
TIM-3 rs1036199 polymorphism increases susceptibility to autoimmune diseases: evidence based on 4200 subjects. Biosci Rep 2018; 38:BSR20181235. [PMID: 30377229 PMCID: PMC6250810 DOI: 10.1042/bsr20181235] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Revised: 10/23/2018] [Accepted: 10/29/2018] [Indexed: 01/10/2023] Open
Abstract
Conflicting results have been reported regarding differing studies on the association between T-cell immunoglobulin and mucin domain 3 polymorphisms and autoimmune disease. The purpose of the present study was to evaluate the association of TIM-3 rs1036199 (4259 G/T) polymorphism with autoimmune disease susceptibility. A meta-analysis was performed to obtain a more precise evaluation of the association. Ten eligible studies were retrieved by searching PubMed, Embase and Web of Science databases, and statistical analyses were performed using STATA software. The pooled results indicated that TIM-3 rs1036199 polymorphism was significantly associated with an increased risk of overall autoimmune disease in allele comparison (G versus T: OR = 1.59, 95%CI: 1.17-2.17) and heterozygous comparison (GT versus TT: OR = 1.68, 95%CI: 1.37-2.06). Subgroup analyses based on disease type demonstrated that TIM-3 rs1036199 polymorphism was associated with an increased risk of rheumatic arthritis (G versus T: OR = 1.88, 95%CI: 1.45-2.44; GT versus TT: OR = 2.02, 95%CI: 1.53-2.65), especially in Asian populations.
Collapse
|
43
|
Agrawal B, Gupta N, Konowalchuk JD. MUC1 Mucin: A Putative Regulatory (Checkpoint) Molecule of T Cells. Front Immunol 2018; 9:2391. [PMID: 30405607 PMCID: PMC6204366 DOI: 10.3389/fimmu.2018.02391] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Accepted: 09/26/2018] [Indexed: 01/30/2023] Open
Abstract
T lymphocytes are at the center of inducing an effective adaptive immune response and maintaining homeostasis. T cell responses are initiated through interactions between antigen presenting cells (APCs) and T cells. The type and strength of signals delivered through the T cell receptor (TCR) may modulate how the cells respond. The TCR-MHC (T cell receptor-major histocompatibility complex molecules) complex dictates the specificity, whereas co-stimulatory signals induced by interaction of various accessory cell surface molecules strengthen and optimize T cell responses. Multiple immune regulatory mechanisms brought about by co-inhibitory molecules expressed on T cells play a key role in orchestrating successful and non-damaging immunity. These co-inhibitory molecules are also referred to as initiators of immune check-points or co-inhibitory pathways. Knowledge of co-inhibitory pathways associated with activated T lymphocytes has allowed a better understanding of (a) the inflammatory and anti-inflammatory processes associated with infectious diseases and autoimmune diseases, and (b) mechanisms by which tumors evade immune attack. Many of these regulatory pathways are non-redundant and function in a highly concerted manner. Targeting them has provided effective approaches in treating cancer and autoimmune diseases. For this reason, it is valuable to identify any co-inhibitory molecules that affect these pathways. MUC1 mucin (CD227) has long been known to be expressed by epithelial cells and overexpressed by a multitude of adenocarcinomas. As long ago as 1998 we made a surprising discovery that MUC1 is also expressed by activated human T cells and we provided the first evidence of the role of MUC1 as a novel T cell regulator. Subsequent studies from different laboratories, as well as ours, supported an immuno-regulatory role of MUC1 in infections, inflammation, and autoimmunity that corroborated our original findings establishing MUC1 as a novel T cell regulatory molecule. In this article, we will discuss the experimental evidence supporting MUC1 as a putative regulatory molecule or a “checkpoint molecule” of T cells with implications as a novel biomarker and therapeutic target in chronic diseases such as autoimmunity, inflammation and cancer, and possibly infections.
Collapse
Affiliation(s)
- Babita Agrawal
- Department of Surgery, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Nancy Gupta
- Department of Surgery, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Jeffrey D Konowalchuk
- Department of Surgery, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
44
|
Zhou K, Guo S, Tong S, Sun Q, Li F, Zhang X, Qiao Y, Liang G. Immunosuppression of Human Adipose-Derived Stem Cells on T Cell Subsets via the Reduction of NF-kappaB Activation Mediated by PD-L1/PD-1 and Gal-9/TIM-3 Pathways. Stem Cells Dev 2018; 27:1191-1202. [PMID: 29978730 DOI: 10.1089/scd.2018.0033] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Affiliation(s)
- Kaijian Zhou
- Department of Plastic Surgery, The First Affiliated Hospital, China Medical University, Shenyang, Liaoning, China
| | - Shu Guo
- Department of Plastic Surgery, The First Affiliated Hospital, China Medical University, Shenyang, Liaoning, China
| | - Shuang Tong
- Department of Plastic Surgery, The First Affiliated Hospital, China Medical University, Shenyang, Liaoning, China
| | - Qiang Sun
- Department of Plastic Surgery, The First Affiliated Hospital, China Medical University, Shenyang, Liaoning, China
| | - Fei Li
- Department of Pharmaceutical Science, The First Affiliated Hospital, China Medical University, Shenyang, Liaoning, China
| | - Xiaowei Zhang
- Central Laboratory, The First Affiliated Hospital, China Medical University, Shenyang, Liaoning, China
| | - Ying Qiao
- Central Laboratory, The First Affiliated Hospital, China Medical University, Shenyang, Liaoning, China
| | - Guoxin Liang
- Institute of AIDS Research, The First Affiliated Hospital, China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
45
|
Sperk M, Domselaar RV, Neogi U. Immune Checkpoints as the Immune System Regulators and Potential Biomarkers in HIV-1 Infection. Int J Mol Sci 2018; 19:ijms19072000. [PMID: 29987244 PMCID: PMC6073446 DOI: 10.3390/ijms19072000] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Revised: 06/29/2018] [Accepted: 07/06/2018] [Indexed: 11/21/2022] Open
Abstract
Immune checkpoints are several co-stimulatory and inhibitory pathways that regulate T cell immune responses. Most of the discoveries about immune checkpoints were made in cancer research where inhibitory immune checkpoints cause immune exhaustion and down-regulate anti-tumor responses. In addition to cancer, immune checkpoints are exploited in chronic infectious diseases. In human immunodeficiency virus (HIV) infection, the immune checkpoint molecule called programmed cell death protein 1 (PD-1) has been determined as being a major regulatory factor for T cell exhaustion. Recent studies with antibodies blocking either PD-1 ligand 1 (PD-L1) or PD-1 show not only promising results in the enhancement of HIV-specific immune responses but even in reducing the latent HIV reservoir. Apart from the therapeutic target for a functional cure of HIV-1, immune checkpoint molecules might be used as biomarkers for monitoring disease progression and therapeutic response. In this review, we will summarize and discuss the inhibitory immune checkpoint molecules PD-1, cytotoxic T-lymphocyte-associated protein 4 (CTLA4), lymphocyte-activation gene 3 (LAG3), and T cell immunoglobulin and mucin-domain-containing-3 (TIM3) as well as the co-stimulatory molecules CD40L and CD70, including their role in immunity, with a particular focus on HIV infection, and being potential targets for a functional HIV cure.
Collapse
Affiliation(s)
- Maike Sperk
- Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institutet, 141 86 Stockholm, Sweden.
| | - Robert van Domselaar
- Department of Medicine Huddinge, Unit of Infectious Diseases, Karolinska Institutet, Karolinska University Hospital, 141 86 Stockholm, Sweden.
| | - Ujjwal Neogi
- Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institutet, 141 86 Stockholm, Sweden.
| |
Collapse
|
46
|
Wang J, Ti Y, Wang Y, Guo G, Jiang H, Chang M, Qian H, Zhao J, Sun G. LAG-3 Represents a Marker of CD4+ T Cells with Regulatory Activity in Patients with Bone Fracture. Immunol Invest 2018; 47:492-503. [PMID: 29671649 DOI: 10.1080/08820139.2018.1458107] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The lymphocyte activation gene 3 (LAG-3) is a CD4 homolog with binding affinity to MHC class II molecules. It is thought that LAG-3 exerts a bimodal function, such that co-ligation of LAG-3 and CD3 could deliver an inhibitory signal in conventional T cells, whereas, on regulatory T cells, LAG-3 expression could promote their inhibitory function. In this study, we investigated the role of LAG-3 expression on CD4+ T cells in patients with long bone fracture. We found that LAG-3+ cells represented approximately 13% of peripheral blood CD4+ T cells on average. Compared to LAG-3- CD4+ T cells, LAG-3+ CD4+ T cells presented significantly higher Foxp3 and CTLA-4 expression. Directly ex vivo or with TCR stimulation, LAG-3+ CD4+ T cells expressed significantly higher levels of IL-10 and TGF-β than LAG-3- CD4+ T cells. Interestingly, blocking the LAG-3-MHC class II interaction actually increased the IL-10 expression by LAG-3+ CD4+ T cells. The frequency of LAG-3+ CD4+ T cell was positively correlated with restoration of healthy bone function in long bone fracture patients. These results together suggested that LAG-3 is a marker of CD4+ T cells with regulatory function; at the same time, LAG-3 might have limited the full suppressive potential of Treg cells.
Collapse
Affiliation(s)
- Jun Wang
- a Department of Orthopedics , Jinling Hospital, Nanjing University School of Medicine , Nanjing , Jiangsu Province , China
| | - Yunfan Ti
- a Department of Orthopedics , Jinling Hospital, Nanjing University School of Medicine , Nanjing , Jiangsu Province , China
| | - Yicun Wang
- a Department of Orthopedics , Jinling Hospital, Nanjing University School of Medicine , Nanjing , Jiangsu Province , China
| | - Guodong Guo
- a Department of Orthopedics , Jinling Hospital, Nanjing University School of Medicine , Nanjing , Jiangsu Province , China
| | - Hui Jiang
- a Department of Orthopedics , Jinling Hospital, Nanjing University School of Medicine , Nanjing , Jiangsu Province , China
| | - Menghan Chang
- a Department of Orthopedics , Jinling Hospital, Nanjing University School of Medicine , Nanjing , Jiangsu Province , China
| | - Hongbo Qian
- a Department of Orthopedics , Jinling Hospital, Nanjing University School of Medicine , Nanjing , Jiangsu Province , China
| | - Jianning Zhao
- a Department of Orthopedics , Jinling Hospital, Nanjing University School of Medicine , Nanjing , Jiangsu Province , China
| | - Guojing Sun
- a Department of Orthopedics , Jinling Hospital, Nanjing University School of Medicine , Nanjing , Jiangsu Province , China
| |
Collapse
|
47
|
Functional variants of TIM-3/HAVCR2 3'UTR in lymphoblastoid cell lines. Future Sci OA 2018; 4:FSO298. [PMID: 29796301 PMCID: PMC5961449 DOI: 10.4155/fsoa-2017-0121] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Accepted: 01/29/2018] [Indexed: 01/10/2023] Open
Abstract
Aim Variants of TIM-3/HAVCR2 3'UTR miRNA binding sites are significantly associated with cancer; however, roles in post-transcriptional regulation have not been elucidated. Methods The regulatory and coding region single nucleotide polymorphisms (SNPs) of TIM-3/HAVCR2 were identified using an online database. Single nucleotide polymorphism Function Prediction was used to predict potential functional relevance of miRNA binding sites. Results The analysis indicated rs9313439, rs4704846, rs3087616 and rs1036199 affect possible miRNA binding sites in TIM-3/HAVCR2 3'UTR. We used additional data on genotypes and limited minor allele frequency >5% in the HapMap populations. Only rs3087616 and rs4704846 were significantly associated with TIM-3/HAVCR2. Conclusion Both rs3087616 and rs4704846 could be putative variants mediating post-transcriptional regulation of the TIM-3/HAVCR2. Deeper understanding of how 3'UTR variants influence the activity by TIM-3/HAVCR2 for therapy against cancer.
Collapse
|
48
|
Tim-3 co-stimulation promotes short-lived effector T cells, restricts memory precursors, and is dispensable for T cell exhaustion. Proc Natl Acad Sci U S A 2018; 115:2455-2460. [PMID: 29463725 DOI: 10.1073/pnas.1712107115] [Citation(s) in RCA: 124] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Tim-3 is highly expressed on a subset of T cells during T cell exhaustion in settings of chronic viral infection and tumors. Using lymphocytic choriomeningitis virus (LCMV) Clone 13, a model for chronic infection, we found that Tim-3 was neither necessary nor sufficient for the development of T cell exhaustion. Nonetheless, expression of Tim-3 was sufficient to drive resistance to PD-L1 blockade therapy during chronic infection. Strikingly, expression of Tim-3 promoted the development of short-lived effector T cells, at the expense of memory precursor development, after acute LCMV infection. These effects were accompanied by increased Akt/mTOR signaling in T cells expressing endogenous or ectopic Tim-3. Conversely, Akt/mTOR signaling was reduced in effector T cells from Tim-3-deficient mice. Thus, Tim-3 is essential for optimal effector T cell responses, and may also contribute to exhaustion by restricting the development of long-lived memory T cells. Taken together, our results suggest that Tim-3 is actually more similar to costimulatory receptors that are up-regulated after T cell activation than to a dominant inhibitory protein like PD-1. These findings have significant implications for the development of anti-Tim-3 antibodies as therapeutic agents.
Collapse
|
49
|
Gorman JV, Colgan JD. Acute stimulation generates Tim-3-expressing T helper type 1 CD4 T cells that persist in vivo and show enhanced effector function. Immunology 2018; 154:418-433. [PMID: 29315553 DOI: 10.1111/imm.12890] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Revised: 12/14/2017] [Accepted: 12/22/2017] [Indexed: 12/20/2022] Open
Abstract
T-cell immunoglobulin and mucin domain 3 (Tim-3) is a surface receptor expressed by T helper type 1 (Th1) effector CD4 T cells, which are critical for defence against intracellular pathogens and have been implicated in autoimmune disease. Previous studies showed that Tim-3 expression makes Th1 cells more susceptible to apoptosis and also marks functionally impaired T cells that arise due to chronic stimulation. However, other studies suggested that Tim-3-expressing Th1 cells do not always have these properties. To further define the relationship between Tim-3 and Th1 cell function, we analysed the characteristics of Th1 cells that expressed Tim-3 in response to brief stimulation in vitro or an acute viral infection in vivo. As expected, cultured CD4 T cells began expressing Tim-3 during Th1 differentiation and secondary stimulation generated Tim-3- and Tim-3+ fractions that were separated and further analysed. When injected into naive mice, Tim-3+ cells down-regulated Tim-3 and survived equally well compared with Tim-3- cells. Further, Tim-3- and Tim-3+ Th1 cells had similar functional responses when transferred into naive mice that were subsequently infected with lymphocytic choriomeningitis virus (LCMV). Cultured Th1 cells that expressed Tim-3 following T-cell receptor stimulation had a greater capacity to express signature Th1 cytokines than their Tim-3- counterparts and showed differential expression of genes that regulate CD4 T-cell function. Consistent with these findings, Tim-3+ Th1 cells generated in response to LCMV infection displayed augmented effector function relative to Tim-3- cells. These results suggest that Tim-3 expression by Th1 cells responding to acute stimulation can mark cells that are functionally competent and have an augmented ability to produce cytokines.
Collapse
Affiliation(s)
- Jacob V Gorman
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA, USA
| | - John D Colgan
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA, USA.,Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA, USA.,Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| |
Collapse
|
50
|
Knaus HA, Kanakry CG, Luznik L, Gojo I. Immunomodulatory Drugs: Immune Checkpoint Agents in Acute Leukemia. Curr Drug Targets 2017; 18:315-331. [PMID: 25981611 DOI: 10.2174/1389450116666150518095346] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2015] [Revised: 01/15/2015] [Accepted: 02/27/2015] [Indexed: 12/20/2022]
Abstract
Intrinsic immune responses to acute leukemia are inhibited by a variety of mechanisms, such as aberrant antigen expression by leukemia cells, secretion of immunosuppressive cytokines and expression of inhibitory enzymes in the tumor microenvironment, expansion of immunoregulatory cells, and activation of immune checkpoint pathways, all leading to T cell dysfunction and/or exhaustion. Leukemic cells, similar to other tumor cells, hijack these inhibitory pathways to evade immune recognition and destruction by cytotoxic T lymphocytes. Thus, blockade of immune checkpoints has emerged as a highly promising approach to augment innate anti-tumor immunity in order to treat malignancies. Most evidence for the clinical efficacy of this immunotherapeutic strategy has been seen in patients with metastatic melanoma, where anti-CTLA-4 and anti-PD-1 antibodies have recently revolutionized treatment of this lethal disease with otherwise limited treatment options. To meet the high demand for new treatment strategies in acute leukemia, clinical testing of these promising therapies is commencing. Herein, we review the biology of multiple inhibitory checkpoints (including CTLA-4, PD-1, TIM-3, LAG-3, BTLA, and CD200R) and their contribution to immune evasion by acute leukemias. In addition, we discuss the current state of preclinical and clinical studies of immune checkpoint inhibition in acute leukemia, which seek to harness the body's own immune system to fight leukemic cells.
Collapse
Affiliation(s)
| | | | | | - Ivana Gojo
- Cancer Research Building I, Room 346, 1650 Orleans Street, Baltimore, MD 21287, United States
| |
Collapse
|