101
|
Topchyan P, Lin S, Cui W. The Role of CD4 T Cell Help in CD8 T Cell Differentiation and Function During Chronic Infection and Cancer. Immune Netw 2023; 23:e41. [PMID: 37970230 PMCID: PMC10643329 DOI: 10.4110/in.2023.23.e41] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 09/29/2023] [Accepted: 10/17/2023] [Indexed: 11/17/2023] Open
Abstract
CD4 and CD8 T cells are key players in the immune response against both pathogenic infections and cancer. CD4 T cells provide help to CD8 T cells via multiple mechanisms, including licensing dendritic cells (DCs), co-stimulation, and cytokine production. During acute infection and vaccination, CD4 T cell help is important for the development of CD8 T cell memory. However, during chronic viral infection and cancer, CD4 helper T cells are critical for the sustained effector CD8 T cell response, through a variety of mechanisms. In this review, we focus on T cell responses in conditions of chronic Ag stimulation, such as chronic viral infection and cancer. In particular, we address the significant role of CD4 T cell help in promoting effector CD8 T cell responses, emerging techniques that can be utilized to further our understanding of how these interactions may take place in the context of tertiary lymphoid structures, and how this key information can be harnessed for therapeutic utility against cancer.
Collapse
Affiliation(s)
- Paytsar Topchyan
- Blood Research Institute, Versiti Wisconsin, Milwaukee, WI 53213, USA
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Siying Lin
- Blood Research Institute, Versiti Wisconsin, Milwaukee, WI 53213, USA
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Department of Pathology, Northwestern University, Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Weiguo Cui
- Blood Research Institute, Versiti Wisconsin, Milwaukee, WI 53213, USA
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Department of Pathology, Northwestern University, Feinberg School of Medicine, Chicago, IL 60611, USA
| |
Collapse
|
102
|
Xue W, Qiu K, Dong B, Guo D, Fu J, Zhu C, Niu Z. Disulfidptosis-associated long non-coding RNA signature predicts the prognosis, tumor microenvironment, and immunotherapy and chemotherapy options in colon adenocarcinoma. Cancer Cell Int 2023; 23:218. [PMID: 37759294 PMCID: PMC10523716 DOI: 10.1186/s12935-023-03065-8] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Accepted: 09/13/2023] [Indexed: 09/29/2023] Open
Abstract
BACKGROUND Disulfidptosis is independent of apoptosis, ferroptosis, and cuproptosis and is associated with cancer progression, treatment response, and prognosis. However, the predictive potential of disulfidptosis-associated lncRNAs in colon adenocarcinoma (COAD) and their features in the tumor immune microenvironment (TIME) require further elucidation. METHODS RNA transcriptome, clinical information, and mutation data of COAD samples were obtained from the TCGA database. The risk model was first constructed by co-expression analysis of disulfidptosis genes and lncRNAs, and prognostic lncRNAs were screened using Cox regression, followed by least absolute shrinkage and selection operator analysis. Enrichment analyses were performed to explore the underlying biological functions and signaling of model-associated differentially expressed genes (MADEGs). Moreover, TIME of MADEGs was analyzed to assess the immunotherapy. Finally, the expression levels of the lncRNAs were verified by taking specimens of patients with COAD from the Affiliated Hospital of Qingdao University. RESULTS We constructed a prognosis-related risk model based on four disulfidptosis-associated lncRNAs (ZEB1-AS1, SNHG16, SATB2-AS1, and ALMS1-IT1). By analyzing the survival of patients in the whole, training, and test groups, we found that patients with COAD in the low-risk group had better overall survival than those in the high-risk group. Validation of the model via Cox analysis and clinical indicators demonstrated that the model had a decent potential for predicting the prognosis of patients with COAD. Enrichment analyses revealed that the MADEGs were related to disulfidptosis-associated biological functions and cancer pathways. Furthermore, patients with COAD in the high-risk group had more positive responses to immune checkpoint inhibitors (ICIs) than those in the low-risk group, as confirmed by TIME analysis. ZEB1-AS1, SNHG16, and ALMS1-IT1 were expressed at higher levels in tumor samples than those in the corresponding paracancerous samples (p < 0.05), whereas SATB2-AS1 was upregulated in the paracancerous samples (p < 0.05). CONCLUSIONS This signature may guide prognosis, molecular mechanisms, and treatment strategies, including ICIs and chemotherapy, in patients with COAD.
Collapse
Affiliation(s)
- Weijie Xue
- Kumamoto University Graduate School of Medical Sciences, Kumamoto, Japan
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Qingdao University, No.16 Jiangsu Road, Qingdao, 266003, China
| | - Kang Qiu
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Hospital of Qingdao University, No. 16 Jiangsu Road, Qingdao, 266003, China
| | - Bingzi Dong
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao, 266003, China
| | - Dong Guo
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Qingdao University, No.16 Jiangsu Road, Qingdao, 266003, China
| | - Junhua Fu
- Department of Operation Room, The Affiliated Hospital of Qingdao University, No.16 Jiangsu Road, Qingdao, 266003, China
| | - Chengzhan Zhu
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Hospital of Qingdao University, No. 16 Jiangsu Road, Qingdao, 266003, China.
| | - Zhaojian Niu
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Qingdao University, No.16 Jiangsu Road, Qingdao, 266003, China.
| |
Collapse
|
103
|
Khanal S, Wieland A, Gunderson AJ. Mechanisms of tertiary lymphoid structure formation: cooperation between inflammation and antigenicity. Front Immunol 2023; 14:1267654. [PMID: 37809103 PMCID: PMC10551175 DOI: 10.3389/fimmu.2023.1267654] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Accepted: 09/04/2023] [Indexed: 10/10/2023] Open
Abstract
To mount an effective anti-tumor immune response capable of controlling or eliminating disease, sufficient numbers of lymphocytes must be recruited to malignant tissue and allowed to sustain their effector functions. Indeed, higher infiltration of T and B cells in tumor tissue, often referred to as "hot tumors", is prognostic for patient survival and predictive of response to immunotherapy in almost all cancer types. The organization of tertiary lymphoid structures (TLS) in solid tumors is a unique example of a hot tumor whereby T and B lymphocytes aggregate with antigen presenting cells and high endothelial venules reflecting the cellular organization observed in lymphoid tissue. Many groups have reported that the presence of preexisting TLS in tumors is associated with a superior adaptive immune response, response to immunotherapy, and improved survivorship over those without TLS. Accordingly, there is significant interest into understanding the mechanisms of how and why TLS organize so that they can be elicited therapeutically in patients with few or no TLS. Unfortunately, the most commonly used mouse models of cancer do not spontaneously form TLS, thus significantly restricting our understanding of TLS biology. This brief review will summarize our current state of knowledge of TLS neogenesis and address the current gaps in the field.
Collapse
Affiliation(s)
- Shrijan Khanal
- Division of Surgical Oncology, Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States
| | - Andreas Wieland
- Department of Otolaryngology, Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States
| | - Andrew J. Gunderson
- Division of Surgical Oncology, Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States
| |
Collapse
|
104
|
Kohlmeyer JL, Lingo JJ, Kaemmer CA, Scherer A, Warrier A, Voigt E, Garay JAR, McGivney GR, Brockman QR, Tang A, Calizo A, Pollard K, Zhang X, Hirbe AC, Pratilas CA, Leidinger M, Breheny P, Chimenti MS, Sieren JC, Monga V, Tanas MR, Meyerholz DK, Darbro BW, Dodd RD, Quelle DE. CDK4/6-MEK Inhibition in MPNSTs Causes Plasma Cell Infiltration, Sensitization to PD-L1 Blockade, and Tumor Regression. Clin Cancer Res 2023; 29:3484-3497. [PMID: 37410426 PMCID: PMC10528807 DOI: 10.1158/1078-0432.ccr-23-0749] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 05/22/2023] [Accepted: 07/03/2023] [Indexed: 07/07/2023]
Abstract
PURPOSE Malignant peripheral nerve sheath tumors (MPNST) are lethal, Ras-driven sarcomas that lack effective therapies. We investigated effects of targeting cyclin-dependent kinases 4 and 6 (CDK4/6), MEK, and/or programmed death-ligand 1 (PD-L1) in preclinical MPNST models. EXPERIMENTAL DESIGN Patient-matched MPNSTs and precursor lesions were examined by FISH, RNA sequencing, IHC, and Connectivity-Map analyses. Antitumor activity of CDK4/6 and MEK inhibitors was measured in MPNST cell lines, patient-derived xenografts (PDX), and de novo mouse MPNSTs, with the latter used to determine anti-PD-L1 response. RESULTS Patient tumor analyses identified CDK4/6 and MEK as actionable targets for MPNST therapy. Low-dose combinations of CDK4/6 and MEK inhibitors synergistically reactivated the retinoblastoma (RB1) tumor suppressor, induced cell death, and decreased clonogenic survival of MPNST cells. In immune-deficient mice, dual CDK4/6-MEK inhibition slowed tumor growth in 4 of 5 MPNST PDXs. In immunocompetent mice, combination therapy of de novo MPNSTs caused tumor regression, delayed resistant tumor outgrowth, and improved survival relative to monotherapies. Drug-sensitive tumors that regressed contained plasma cells and increased cytotoxic T cells, whereas drug-resistant tumors adopted an immunosuppressive microenvironment with elevated MHC II-low macrophages and increased tumor cell PD-L1 expression. Excitingly, CDK4/6-MEK inhibition sensitized MPNSTs to anti-PD-L1 immune checkpoint blockade (ICB) with some mice showing complete tumor regression. CONCLUSIONS CDK4/6-MEK inhibition induces a novel plasma cell-associated immune response and extended antitumor activity in MPNSTs, which dramatically enhances anti-PD-L1 therapy. These preclinical findings provide strong rationale for clinical translation of CDK4/6-MEK-ICB targeted therapies in MPNST as they may yield sustained antitumor responses and improved patient outcomes.
Collapse
Affiliation(s)
- Jordan L Kohlmeyer
- Molecular Medicine Graduate Program, Carver College of Medicine, University of Iowa, Iowa City, Iowa
- Department of Neuroscience and Pharmacology, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | - Joshua J Lingo
- Cancer Biology Graduate Program, University of Iowa, Iowa City, Iowa
- Holden Comprehensive Cancer Center, University of Iowa, Iowa City, Iowa
| | - Courtney A Kaemmer
- Department of Neuroscience and Pharmacology, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | - Amanda Scherer
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | - Akshaya Warrier
- Cancer Biology Graduate Program, University of Iowa, Iowa City, Iowa
- Medical Scientist Training Program, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | - Ellen Voigt
- Cancer Biology Graduate Program, University of Iowa, Iowa City, Iowa
- Holden Comprehensive Cancer Center, University of Iowa, Iowa City, Iowa
- Medical Scientist Training Program, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | | | - Gavin R McGivney
- Cancer Biology Graduate Program, University of Iowa, Iowa City, Iowa
| | - Qierra R Brockman
- Molecular Medicine Graduate Program, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | - Amy Tang
- Department of Microbiology and Molecular Cell Biology, Leroy T. Canoles Jr. Cancer Center, Eastern Virginia Medical School, Norfolk, Virginia
| | - Ana Calizo
- Department of Oncology, Johns Hopkins University, Sidney Kimmel Comprehensive Cancer Center, Baltimore, Maryland
| | - Kai Pollard
- Department of Oncology, Johns Hopkins University, Sidney Kimmel Comprehensive Cancer Center, Baltimore, Maryland
| | - Xiaochun Zhang
- Division of Medical Oncology, Washington University, St. Louis, Missouri
| | - Angela C Hirbe
- Division of Medical Oncology, Washington University, St. Louis, Missouri
| | - Christine A Pratilas
- Department of Oncology, Johns Hopkins University, Sidney Kimmel Comprehensive Cancer Center, Baltimore, Maryland
| | - Mariah Leidinger
- Department of Pathology, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | - Patrick Breheny
- Holden Comprehensive Cancer Center, University of Iowa, Iowa City, Iowa
- Department of Biostatistics, College of Public Health, University of Iowa, Iowa City, Iowa
| | - Michael S Chimenti
- Iowa Institute of Human Genetics, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | - Jessica C. Sieren
- Holden Comprehensive Cancer Center, University of Iowa, Iowa City, Iowa
- Department of Radiation, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | - Varun Monga
- Holden Comprehensive Cancer Center, University of Iowa, Iowa City, Iowa
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | - Munir R Tanas
- Molecular Medicine Graduate Program, Carver College of Medicine, University of Iowa, Iowa City, Iowa
- Cancer Biology Graduate Program, University of Iowa, Iowa City, Iowa
- Holden Comprehensive Cancer Center, University of Iowa, Iowa City, Iowa
- Department of Pathology, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | - David K Meyerholz
- Department of Pathology, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | - Benjamin W Darbro
- Holden Comprehensive Cancer Center, University of Iowa, Iowa City, Iowa
- Medical Scientist Training Program, Carver College of Medicine, University of Iowa, Iowa City, Iowa
- Department of Pediatrics, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | - Rebecca D Dodd
- Molecular Medicine Graduate Program, Carver College of Medicine, University of Iowa, Iowa City, Iowa
- Cancer Biology Graduate Program, University of Iowa, Iowa City, Iowa
- Holden Comprehensive Cancer Center, University of Iowa, Iowa City, Iowa
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, Iowa
- Medical Scientist Training Program, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | - Dawn E Quelle
- Molecular Medicine Graduate Program, Carver College of Medicine, University of Iowa, Iowa City, Iowa
- Department of Neuroscience and Pharmacology, Carver College of Medicine, University of Iowa, Iowa City, Iowa
- Cancer Biology Graduate Program, University of Iowa, Iowa City, Iowa
- Holden Comprehensive Cancer Center, University of Iowa, Iowa City, Iowa
- Medical Scientist Training Program, Carver College of Medicine, University of Iowa, Iowa City, Iowa
- Department of Pathology, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| |
Collapse
|
105
|
Bonaud A, Larraufie P, Khamyath M, Szachnowski U, Flint SM, Brunel-Meunier N, Delhommeau F, Munier A, Lönnberg T, Toffano-Nioche C, Gautheret D, Balabanian K, Espéli M. Transinteractome analysis reveals distinct niche requirements for isotype-based plasma cell subsets in the bone marrow. Eur J Immunol 2023; 53:e2250334. [PMID: 37377335 DOI: 10.1002/eji.202250334] [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: 12/13/2022] [Revised: 05/09/2023] [Accepted: 05/22/2023] [Indexed: 06/29/2023]
Abstract
Bone marrow (BM) long-lived plasma cells (PCs) are essential for long-term protection against infection, and their persistence within this organ relies on interactions with Cxcl12-expressing stromal cells that are still not clearly identified. Here, using single cell RNAseq and in silico transinteractome analyses, we identified Leptin receptor positive (LepR+ ) mesenchymal cells as the stromal cell subset most likely to interact with PCs within the BM. Moreover, we demonstrated that depending on the isotype they express, PCs may use different sets of integrins and adhesion molecules to interact with these stromal cells. Altogether, our results constitute an unprecedented characterization of PC subset stromal niches and open new avenues for the specific targeting of BM PCs based on their isotype.
Collapse
Affiliation(s)
- Amélie Bonaud
- Université Paris Cité, Institut de Recherche Saint-Louis, INSERM U1160, Paris, France
- OPALE Carnot Institute, Hôpital St-Louis, Paris, France
| | - Pierre Larraufie
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France
| | - Mélanie Khamyath
- Université Paris Cité, Institut de Recherche Saint-Louis, INSERM U1160, Paris, France
- OPALE Carnot Institute, Hôpital St-Louis, Paris, France
| | - Ugo Szachnowski
- Université Paris-Saclay, INSERM, Inflammation, Microbiome and Immunosurveillance, Clamart, France
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), Gif-sur-Yvette, France
| | - Shaun M Flint
- Université Paris-Saclay, INSERM, Inflammation, Microbiome and Immunosurveillance, Clamart, France
| | - Nadège Brunel-Meunier
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine (CRSA), AP-HP, Saint-Antoine Hospital, Paris, France
| | - François Delhommeau
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine (CRSA), AP-HP, Saint-Antoine Hospital, Paris, France
| | - Annie Munier
- Sorbonne Université-INSERM UMRS_938, Centre de Recherche Saint-Antoine (CRSA), Plateforme de Cytométrie CISA, Paris, France
| | - Tapio Lönnberg
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Hinxton, Cambridge, UK
- Wellcome Trust Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Claire Toffano-Nioche
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), Gif-sur-Yvette, France
| | - Daniel Gautheret
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), Gif-sur-Yvette, France
| | - Karl Balabanian
- Université Paris Cité, Institut de Recherche Saint-Louis, INSERM U1160, Paris, France
- OPALE Carnot Institute, Hôpital St-Louis, Paris, France
| | - Marion Espéli
- Université Paris Cité, Institut de Recherche Saint-Louis, INSERM U1160, Paris, France
- OPALE Carnot Institute, Hôpital St-Louis, Paris, France
| |
Collapse
|
106
|
Heinze K, Cairns ES, Thornton S, Harris B, Milne K, Grube M, Meyer C, Karnezis AN, Fereday S, Garsed DW, Leung SC, Chiu DS, Moubarak M, Harter P, Heitz F, McAlpine JN, DeFazio A, Bowtell DD, Goode EL, Pike M, Ramus SJ, Pearce CL, Staebler A, Köbel M, Kommoss S, Talhouk A, Nelson BH, Anglesio MS. The Prognostic Effect of Immune Cell Infiltration Depends on Molecular Subtype in Endometrioid Ovarian Carcinomas. Clin Cancer Res 2023; 29:3471-3483. [PMID: 37339172 PMCID: PMC10472107 DOI: 10.1158/1078-0432.ccr-22-3815] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 04/14/2023] [Accepted: 06/14/2023] [Indexed: 06/22/2023]
Abstract
PURPOSE Endometrioid ovarian carcinoma (ENOC) is the second most-common type of ovarian carcinoma, comprising 10%-20% of cases. Recently, the study of ENOC has benefitted from comparisons to endometrial carcinomas including defining ENOC with four prognostic molecular subtypes. Each subtype suggests differential mechanisms of progression, although tumor-initiating events remain elusive. There is evidence that the ovarian microenvironment may be critical to early lesion establishment and progression. However, while immune infiltrates have been well studied in high-grade serous ovarian carcinoma, studies in ENOC are limited. EXPERIMENTAL DESIGN We report on 210 ENOC, with clinical follow-up and molecular subtype annotation. Using multiplex IHC and immunofluorescence, we examine the prevalence of T-cell lineage, B-cell lineage, macrophages, and populations with programmed cell death protein 1 or programmed death-ligand 1 across subtypes of ENOC. RESULTS Immune cell infiltrates in tumor epithelium and stroma showed higher densities in ENOC subtypes with known high mutation burden (POLEmut and MMRd). While molecular subtypes were prognostically significant, immune infiltrates were not (overall survival P > 0.2). Analysis by molecular subtype revealed that immune cell density was prognostically significant in only the no specific molecular profile (NSMP) subtype, where immune infiltrates lacking B cells (TILB minus) had inferior outcome (disease-specific survival: HR, 4.0; 95% confidence interval, 1.1-14.7; P < 0.05). Similar to endometrial carcinomas, molecular subtype stratification was generally superior to immune response in predicting outcomes. CONCLUSIONS Subtype stratification is critical for better understanding of ENOC, in particular the distribution and prognostic significance of immune cell infiltrates. The role of B cells in the immune response within NSMP tumors warrants further study.
Collapse
Affiliation(s)
- Karolin Heinze
- Department of Obstetrics and Gynecology, University of British Columbia, Vancouver, Canada
- OVCARE - British Columbia's Gynecological Cancer Research Program, BC Cancer, Vancouver General Hospital, and the University of British Columbia, Vancouver, British Columbia, Canada
| | - Evan S. Cairns
- Department of Obstetrics and Gynecology, University of British Columbia, Vancouver, Canada
| | - Shelby Thornton
- OVCARE - British Columbia's Gynecological Cancer Research Program, BC Cancer, Vancouver General Hospital, and the University of British Columbia, Vancouver, British Columbia, Canada
- Molecular and Cellular Immunology Core (MCIC), Deeley Research Centre, BC Cancer, Victoria, Canada
| | - Bronwyn Harris
- Molecular and Cellular Immunology Core (MCIC), Deeley Research Centre, BC Cancer, Victoria, Canada
| | - Katy Milne
- Molecular and Cellular Immunology Core (MCIC), Deeley Research Centre, BC Cancer, Victoria, Canada
| | - Marcel Grube
- Department of Women's Health, Tübingen University Hospital, Tübingen, Germany
| | - Charlotte Meyer
- OVCARE - British Columbia's Gynecological Cancer Research Program, BC Cancer, Vancouver General Hospital, and the University of British Columbia, Vancouver, British Columbia, Canada
- Department of Women's Health, Tübingen University Hospital, Tübingen, Germany
| | - Anthony N. Karnezis
- Department of Pathology and Laboratory, UC Davis Medical Center, Sacramento, California
| | - Sian Fereday
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia
| | - Dale W. Garsed
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia
| | - Samuel C.Y. Leung
- OVCARE - British Columbia's Gynecological Cancer Research Program, BC Cancer, Vancouver General Hospital, and the University of British Columbia, Vancouver, British Columbia, Canada
| | - Derek S. Chiu
- OVCARE - British Columbia's Gynecological Cancer Research Program, BC Cancer, Vancouver General Hospital, and the University of British Columbia, Vancouver, British Columbia, Canada
| | - Malak Moubarak
- Kliniken Essen Mitte, Department of Gynecology and Gynecologic Oncology, Essen, Germany
| | - Philipp Harter
- Kliniken Essen Mitte, Department of Gynecology and Gynecologic Oncology, Essen, Germany
| | - Florian Heitz
- Kliniken Essen Mitte, Department of Gynecology and Gynecologic Oncology, Essen, Germany
- Department for Gynecology with the Center for Oncologic Surgery Charité Campus Virchow-Klinikum, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Jessica N. McAlpine
- Department of Obstetrics and Gynecology, University of British Columbia, Vancouver, Canada
- OVCARE - British Columbia's Gynecological Cancer Research Program, BC Cancer, Vancouver General Hospital, and the University of British Columbia, Vancouver, British Columbia, Canada
| | - Anna DeFazio
- Centre for Cancer Research, The Westmead Institute for Medical Research, Sydney, New South Wales, Australia
- Department of Gynaecological Oncology, Westmead Hospital, Sydney, New South Wales, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
- The Daffodil Centre, The University of Sydney, a joint venture with Cancer Council NSW, Sydney, New South Wales, Australia
| | - David D.L. Bowtell
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia
| | - Ellen L. Goode
- Mayo Clinic, Department of Health Science Research, Division of Epidemiology, Rochester, Minnesota
| | - Malcolm Pike
- Department of Epidemiology & Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Susan J. Ramus
- School of Clinical Medicine, UNSW Medicine and Health, University of New South Wales Sydney, Sydney, Australia
- Multidisciplinary Ovarian Cancer Outcomes Group (Consortium)
| | - C. Leigh Pearce
- Multidisciplinary Ovarian Cancer Outcomes Group (Consortium)
- School of Public Health, University of Michigan, Ann Arbor, Michigan
| | - Annette Staebler
- Institute of Pathology, University Hospital of Tübingen, Tübingen, Germany
| | - Martin Köbel
- Department of Pathology, University of Calgary, Calgary, Alberta, Canada
| | - Stefan Kommoss
- Department of Women's Health, Tübingen University Hospital, Tübingen, Germany
| | - Aline Talhouk
- Department of Obstetrics and Gynecology, University of British Columbia, Vancouver, Canada
- OVCARE - British Columbia's Gynecological Cancer Research Program, BC Cancer, Vancouver General Hospital, and the University of British Columbia, Vancouver, British Columbia, Canada
| | - Brad H. Nelson
- OVCARE - British Columbia's Gynecological Cancer Research Program, BC Cancer, Vancouver General Hospital, and the University of British Columbia, Vancouver, British Columbia, Canada
- Molecular and Cellular Immunology Core (MCIC), Deeley Research Centre, BC Cancer, Victoria, Canada
- Multidisciplinary Ovarian Cancer Outcomes Group (Consortium)
| | - Michael S. Anglesio
- Department of Obstetrics and Gynecology, University of British Columbia, Vancouver, Canada
- OVCARE - British Columbia's Gynecological Cancer Research Program, BC Cancer, Vancouver General Hospital, and the University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
107
|
Zhang Q, Liu Y, Wang X, Zhang C, Hou M, Liu Y. Integration of single-cell RNA sequencing and bulk RNA transcriptome sequencing reveals a heterogeneous immune landscape and pivotal cell subpopulations associated with colorectal cancer prognosis. Front Immunol 2023; 14:1184167. [PMID: 37675100 PMCID: PMC10477986 DOI: 10.3389/fimmu.2023.1184167] [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: 03/11/2023] [Accepted: 07/27/2023] [Indexed: 09/08/2023] Open
Abstract
Introduction Colorectal cancer (CRC) is a highly heterogeneous cancer. The molecular and cellular characteristics differ between the colon and rectal cancer type due to the differences in their anatomical location and pathological properties. With the advent of single-cell sequencing, it has become possible to analyze inter- and intra-tumoral tissue heterogeneities. Methods A comprehensive CRC immune atlas, comprising 62,398 immune cells, was re-structured into 33 immune cell clusters at the single-cell level. Further, the immune cell lineage heterogeneity of colon, rectal, and paracancerous tissues was explored. Simultaneously, we characterized the TAM phenotypes and analyzed the transcriptomic factor regulatory network of each macrophage subset using SCENIC. In addition, monocle2 was used to elucidate the B cell developmental trajectory. The crosstalk between immune cells was explored using CellChat and the patterns of incoming and outgoing signals within the overall immune cell population were identified. Afterwards, the bulk RNA-sequencing data from The Cancer Genome Atlas (TCGA) were combined and the relative infiltration abundance of the identified subpopulations was analyzed using CIBERSORT. Moreover, cell composition patterns could be classified into five tumor microenvironment (TME) subtypes by employing a consistent non-negative matrix algorithm. Finally, the co-expression and interaction between SPP1+TAMs and Treg cells in the tumor microenvironment were analyzed by multiplex immunohistochemistry. Results In the T cell lineage, we found that CXCL13+T cells were more widely distributed in colorectal cancer tissues, and the proportion of infiltration was increased. In addition, Th17 was found accounted for the highest proportion in CD39+CD101+PD1+T cells. Mover, Ma1-SPP1 showed the characteristics of M2 phenotypes and displayed an increased proportion in tumor tissues, which may promote angiogenesis. Plasma cells (PCs) displayed a significantly heterogeneous distribution in tumor as well as normal tissues. Specifically, the IgA+ PC population could be shown to be decreased in colorectal tumor tissues whereas the IgG+ PC one was enriched. In addition, information flow mediated by SPP1 and CD44, regulate signaling pathways of tumor progression. Among the five TME subtypes, the TME-1 subtype displayed a markedly reduced proportion of T-cell infiltration with the highest proportion of macrophages which was correlated to the worst prognosis. Finally, the co-expression and interaction between SPP1+TAMs and Treg cells were observed in the CD44 enriched region. Discussion The heterogeneity distribution and phenotype of immune cells were analyzed in colon cancer and rectal cancer at the single-cell level. Further, the prognostic role of major tumor-infiltrating lymphocytes and TME subtypes in CRC was evaluated by integrating bulk RNA. These findings provide novel insight into the immunotherapy of CRC.
Collapse
Affiliation(s)
- Qian Zhang
- College of Medicine and Biological Information Engineering, Northeastern University, Shenyang, Liaoning, China
- Shuren International College, Shenyang Medical College, Shenyang, Liaoning, China
| | - Yang Liu
- Shuren International College, Shenyang Medical College, Shenyang, Liaoning, China
| | - Xinyu Wang
- Department of General Surgery, General Hospital of Northern Theater Command, Shenyang, China
| | - Cheng Zhang
- Department of General Surgery, General Hospital of Northern Theater Command, Shenyang, China
| | - Mingxiao Hou
- College of Medicine and Biological Information Engineering, Northeastern University, Shenyang, Liaoning, China
- Shuren International College, Shenyang Medical College, Shenyang, Liaoning, China
- The Second Affiliated Hospital of Shenyang Medical College, The Veterans General Hospital of Liaoning Province, Shenyang, Liaoning, China
| | - Yunen Liu
- Shuren International College, Shenyang Medical College, Shenyang, Liaoning, China
| |
Collapse
|
108
|
Sabahi M, Salehipour A, Bazl MSY, Rezaei N, Mansouri A, Borghei-Razavi H. Local immunotherapy of glioblastoma: A comprehensive review of the concept. J Neuroimmunol 2023; 381:578146. [PMID: 37451079 DOI: 10.1016/j.jneuroim.2023.578146] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Revised: 05/24/2023] [Accepted: 07/04/2023] [Indexed: 07/18/2023]
Abstract
Despite advancements in standard treatments, the prognosis of Glioblastoma (GBM) remains poor, prompting research for novel therapies. Immunotherapy is a promising treatment option for GBM, and many immunotherapeutic agents are currently under investigation. Chimeric antigen receptor (CAR) T cells are rapidly evolving in immunotherapy of GBM with many clinical trials showing efficacy of CAR T cells exerting anti-tumor activity following recognition of tumor-associated antigens (TAAs). Exhaustion in CAR T cells can reduce their capacity for long-term persistence and anti-tumor action. Local immunotherapy, which targets the tumor microenvironment and creates a more hospitable immunological environment for CAR T cells, has the potential to reduce CAR T cell exhaustion and increase immunity. Tertiary lymphoid structures (TLS) are ectopic lymphoid-like formations that can develop within the tumor microenvironment or in other non-lymphoid tissues. As a comprehensive local immunotherapy tool, the incorporation of TLS into an implanted biodegradable scaffold has amazing immunotherapeutic potential. The immune response to GBM can be improved even further by strategically inserting a stimulator of interferon genes (STING) agonist into the scaffold. Additionally, the scaffold's addition of glioma stem cells (GSC), which immunotherapeutic approaches may use to target, enhances the removal of cancer cells from their source. Furthermore, it has been demonstrated that GSCs have an impact on TLS formation, which helps to create a favorable tumor microenvironment. Herein, we overview local delivery of a highly specific tandem AND-gate CAR T cell along with above mentioned components. A multifaceted approach that successfully engages the immune system to mount an efficient targeted immune response against GBM is provided by the integration of CAR T cells, TLS, STING agonists, and GSCs within an implantable biodegradable scaffold. This approach offers a promising therapeutic approach for patients with GBM.
Collapse
Affiliation(s)
- Mohammadmahdi Sabahi
- Department of Neurological Surgery, Pauline Braathen Neurological Center, Cleveland Clinic Florida, Weston, FL, USA.
| | - Arash Salehipour
- Neurosurgery Research Group (NRG), Student Research Committee, Hamadan University of Medical Sciences, Hamadan, Iran; Brain and Spinal Cord Injury Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran.
| | - Mohammad Sajjad Yavari Bazl
- Neurosurgery Research Group (NRG), Student Research Committee, Hamadan University of Medical Sciences, Hamadan, Iran; Brain and Spinal Cord Injury Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran.
| | - Nima Rezaei
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran; Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran; Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| | - Alireza Mansouri
- Department of Neurosurgery, Penn State Milton S. Hershey Medical Center, Hershey, PA, USA.
| | - Hamid Borghei-Razavi
- Department of Neurological Surgery, Pauline Braathen Neurological Center, Cleveland Clinic Florida, Weston, FL, USA.
| |
Collapse
|
109
|
Zhang D, Ni Y, Wang Y, Feng J, Zhuang N, Li J, Liu L, Shen W, Zheng J, Zheng W, Qian C, Shan J, Zhou Z. Spatial heterogeneity of tumor microenvironment influences the prognosis of clear cell renal cell carcinoma. J Transl Med 2023; 21:489. [PMID: 37474942 PMCID: PMC10360235 DOI: 10.1186/s12967-023-04336-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2023] [Accepted: 07/09/2023] [Indexed: 07/22/2023] Open
Abstract
BACKGROUND Clear cell renal cell carcinoma (ccRCC) is an immunologically and histologically diverse tumor. However, how the structural heterogeneity of tumor microenvironment (TME) affects cancer progression and treatment response remains unclear. Hence, we characterized the TME architectures of ccRCC tissues using imaging mass cytometry (IMC) and explored their associations with clinical outcome and therapeutic response. METHODS Using IMC, we profiled the TME landscape of ccRCC and paracancerous tissue by measuring 17 markers involved in tissue architecture, immune cell and immune activation. In the ccRCC tissue, we identified distinct immune architectures of ccRCC tissue based on the mix score and performed cellular neighborhood (CN) analysis to subdivide TME phenotypes. Moreover, we assessed the relationship between the different TME phenotypes and ccRCC patient survival, clinical features and treatment response. RESULTS We found that ccRCC tissues had higher levels of CD8+ T cells, CD163- macrophages, Treg cells, endothelial cells, and fibroblasts than paracancerous tissues. Immune infiltrates in ccRCC tissues distinctly showed clustered and scattered patterns. Within the clustered pattern, we identified two subtypes with different clinical outcomes based on CN analysis. The TLS-like phenotype had cell communities resembling tertiary lymphoid structures, characterized by cell-cell interactions of CD8+ T cells-B cells and GZMB+CD8+ T cells-B cells, which exhibited anti-tumor features and favorable outcomes, while the Macrophage/T-clustered phenotype with macrophage- or T cell-dominated cell communities had a poor prognosis. Patients with scattered immune architecture could be further divided into scattered-CN-hot and scattered-CN-cold phenotypes based on the presence or absence of immune CNs, but both had a better prognosis than the macrophage/T-clustered phenotype. We further analyzed the relationship between the TME phenotypes and treatment response in five metastatic ccRCC patients treated with sunitinib, and found that all three responders were scattered-CN-hot phenotype while both non-responders were macrophage/T-clustered phenotype. CONCLUSION Our study revealed the structural heterogeneity of TME in ccRCC and its impact on clinical outcome and personalized treatment. These findings highlight the potential of IMC and CN analysis for characterizing TME structural units in cancer research.
Collapse
Affiliation(s)
- Dawei Zhang
- Department of Urology, The Southwest Hospital, Army Medical University, No. 30 Gaotanyan Street, Shapingba District, Chongqing, 400038, China
| | - Yuanli Ni
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, No. 181 Hanyu Road, Shapingba District, Chongqing, 400030, China
| | - Yongquan Wang
- Department of Urology, The Southwest Hospital, Army Medical University, No. 30 Gaotanyan Street, Shapingba District, Chongqing, 400038, China
| | - Juan Feng
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, No. 181 Hanyu Road, Shapingba District, Chongqing, 400030, China
| | - Na Zhuang
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, No. 181 Hanyu Road, Shapingba District, Chongqing, 400030, China
| | - Jiatao Li
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, No. 181 Hanyu Road, Shapingba District, Chongqing, 400030, China
| | - Limei Liu
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, No. 181 Hanyu Road, Shapingba District, Chongqing, 400030, China
| | - Wenhao Shen
- Department of Urology, The Southwest Hospital, Army Medical University, No. 30 Gaotanyan Street, Shapingba District, Chongqing, 400038, China
| | - Ji Zheng
- Department of Urology, The Southwest Hospital, Army Medical University, No. 30 Gaotanyan Street, Shapingba District, Chongqing, 400038, China
| | - Wei Zheng
- Anesthesiology Department, The 80th Army Hospital of Chinese PLA, Weifang, 261021, Shandong, China
| | - Cheng Qian
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, No. 181 Hanyu Road, Shapingba District, Chongqing, 400030, China.
| | - Juanjuan Shan
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, No. 181 Hanyu Road, Shapingba District, Chongqing, 400030, China.
| | - Zhansong Zhou
- Department of Urology, The Southwest Hospital, Army Medical University, No. 30 Gaotanyan Street, Shapingba District, Chongqing, 400038, China.
| |
Collapse
|
110
|
Pi Y, Sun F, Zhang Z, Liu X, Lou G. A Novel Notch-Related Gene Signature for Prognosis and Immune Response Prediction in Ovarian Cancer. MEDICINA (KAUNAS, LITHUANIA) 2023; 59:1277. [PMID: 37512088 PMCID: PMC10385113 DOI: 10.3390/medicina59071277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Revised: 06/07/2023] [Accepted: 06/19/2023] [Indexed: 07/30/2023]
Abstract
Background and Objectives: Notch is a fascinating signaling pathway. It is extensively involved in tumor growth, cancer stem cells, metastasis, and treatment resistance and plays important roles in metabolic regulation, tumor microenvironment, and tumor immunity. However, the role of Notch in ovarian cancer (OC) has yet to be fully understood. Therefore, this study systematically described the expression, mutation, and copy number variation of genes in the Notch signaling pathway in OC and evaluated the relationship between gene mutation and Overall Survival (OS) prognosis. Materials and Methods: Notch risk score (NTRS) was established by univariate Cox regression analysis combined with Lasso regression analysis, and the efficacy of NTRS in predicting prognosis and immunotherapy response in patients with OC was verified. We further assessed the correlations of NTRS with clinical features, immune infiltration level, immune checkpoint expression, and immune characteristics. Additionally, differential expression and functions of the fourteen signature genes were confirmed via vitro assays. Results: The results showed that Notch genes (NTGs) were markedly differentiated between tumor and normal tissues, which may help to explain the high heterogeneity in the biological characteristics and therapeutic outcomes of human OC. A Notch risk (NTR) prognostic model based on 11 key NTGs was successfully constructed. Tumors with high Notch risk scores (NTRS) were independently associated with shorter overall survival and poorer immunotherapy outcomes. We further assessed the correlations of NTRS with immune characteristics. The results showed that NTGs play a key role in regulating the tumor immune microenvironment. Additionally, we validated the baseline and induced expressions of 14 prognosis-related NTGs in our own OC samples. In vitro assays confirmed that the knockdown of NCOR2 and APH1B and overexpression of HEY2 and SKP2 could inhibit the proliferation, invasion, and migration of OC cells. Conclusions: These findings emphasize that Notch multilayer changes are associated with the prognosis of patients with OC and the characteristics of immune cell infiltration. Our predictive signature may predict the prognosis and immunotherapy response of OC patients in an independent manner. NCOR2, APH1B, HEY2, and SKP2 may more prominently represent important indicators to improve patient prognosis.
Collapse
Affiliation(s)
- Yanan Pi
- Department of Gynecology, Harbin Medical University Cancer Hospital, Harbin 150086, China
| | - Fusheng Sun
- Department of Gynecology, Harbin Medical University Cancer Hospital, Harbin 150086, China
| | - Zhaocong Zhang
- Department of Gynecology, Harbin Medical University Cancer Hospital, Harbin 150086, China
| | - Xiaoli Liu
- Harbin Obstetrics and Gynecology Hospital, Harbin Medical University, Harbin 150086, China
| | - Ge Lou
- Department of Gynecology, Harbin Medical University Cancer Hospital, Harbin 150086, China
| |
Collapse
|
111
|
Li H, Ding JY, Zhang MJ, Yu HJ, Sun ZJ. Tertiary lymphoid structures and cytokines interconnections: The implication in cancer immunotherapy. Cancer Lett 2023:216293. [PMID: 37392991 DOI: 10.1016/j.canlet.2023.216293] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 06/26/2023] [Accepted: 06/28/2023] [Indexed: 07/03/2023]
Abstract
Tertiary lymphoid structures (TLSs) are organized aggregates of lymphocytes and antigen-presenting cells that develop in non-lymphoid tissues during chronic inflammation, resembling the structure and features of secondary lymphoid organs. Numerous studies have shown that TLSs may be an important source of antitumor immunity within solid tumors, facilitating T cell and B cell differentiation and the subsequent production of antitumor antibodies, which are beneficial for cancer prognosis and responses to immunotherapy. The formation of TLS relies on the cytokine signaling network between heterogeneous cell populations, such as stromal cells, lymphocytes and cancer cells. The coordinated action of various cytokines drives the complex process of TLS development. In this review, we will comprehensively describe the mechanisms by which various cytokines regulate TLS formation and function, and the recent advancements and therapeutic potential of exploiting these mechanisms to induce TLS as an emerging immunotherapeutic approach or to enhance existing immunotherapy.
Collapse
Affiliation(s)
- Hao Li
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine, Ministry of Education, School and Hospital of Stomatology, Wuhan University, 237 Luoyu Road, Wuhan, 430079, China; Department of Oral Maxillofacial-Head Neck Oncology, School and Hospital of Stomatology, Wuhan University, 237 Luoyu Road, Wuhan, 430079, China.
| | - Jia-Yi Ding
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine, Ministry of Education, School and Hospital of Stomatology, Wuhan University, 237 Luoyu Road, Wuhan, 430079, China.
| | - Meng-Jie Zhang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine, Ministry of Education, School and Hospital of Stomatology, Wuhan University, 237 Luoyu Road, Wuhan, 430079, China.
| | - Hai-Jun Yu
- Department of Radiation and Medical Oncology, Hubei Province Cancer Clinical Study Center, Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital of Wuhan University, 430071, Wuhan, China.
| | - Zhi-Jun Sun
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine, Ministry of Education, School and Hospital of Stomatology, Wuhan University, 237 Luoyu Road, Wuhan, 430079, China; Department of Oral Maxillofacial-Head Neck Oncology, School and Hospital of Stomatology, Wuhan University, 237 Luoyu Road, Wuhan, 430079, China.
| |
Collapse
|
112
|
Ye W, Li M, Luo K. Therapies Targeting Immune Cells in Tumor Microenvironment for Non-Small Cell Lung Cancer. Pharmaceutics 2023; 15:1788. [PMID: 37513975 PMCID: PMC10384189 DOI: 10.3390/pharmaceutics15071788] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 06/02/2023] [Accepted: 06/15/2023] [Indexed: 07/30/2023] Open
Abstract
The tumor microenvironment (TME) plays critical roles in immune modulation and tumor malignancies in the process of cancer development. Immune cells constitute a significant component of the TME and influence the migration and metastasis of tumor cells. Recently, a number of therapeutic approaches targeting immune cells have proven promising and have already been used to treat different types of cancer. In particular, PD-1 and PD-L1 inhibitors have been used in the first-line setting in non-small cell lung cancer (NSCLC) with PD-L1 expression ≥1%, as approved by the FDA. In this review, we provide an introduction to the immune cells in the TME and their efficacies, and then we discuss current immunotherapies in NSCLC and scientific research progress in this field.
Collapse
Affiliation(s)
- Wei Ye
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510091, China
| | - Meiye Li
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510091, China
| | - Kewang Luo
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510091, China
- People's Hospital of Longhua, Affiliated Longhua People's Hospital, Southern Medical University, Shenzhen 518109, China
| |
Collapse
|
113
|
Lu H, Lou H, Wengert G, Paudel R, Patel N, Desai S, Crum B, Linton-Reid K, Chen M, Li D, Ip J, Mauri F, Pinato DJ, Rockall A, Copley SJ, Ghaem-Maghami S, Aboagye EO. Tumor and local lymphoid tissue interaction determines prognosis in high-grade serous ovarian cancer. Cell Rep Med 2023:101092. [PMID: 37348499 PMCID: PMC10394173 DOI: 10.1016/j.xcrm.2023.101092] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 03/29/2023] [Accepted: 05/30/2023] [Indexed: 06/24/2023]
Abstract
Tertiary lymphoid structure (TLS) is associated with prognosis in copy-number-driven tumors, including high-grade serous ovarian cancer (HGSOC), although the function of TLS and its interaction with copy-number alterations in HGSOC are not fully understood. In the current study, we confirm that TLS-high HGSOC patients show significantly better progression-free survival (PFS). We show that the presence of TLS in HGSOC tumors is associated with B cell maturation and cytotoxic tumor-specific T cell activation and proliferation. In addition, the copy-number loss of IL15 and CXCL10 may limit TLS formation in HGSOC; a list of genes that may dysregulate TLS function is also proposed. Last, a radiomics-based signature is developed to predict the presence of TLS, which independently predicts PFS in both HGSOC patients and immune checkpoint inhibitor (ICI)-treated non-small cell lung cancer (NSCLC) patients. Overall, we reveal that TLS coordinates intratumoral B cell and T cell response to HGSOC tumor, while the cancer genome evolves to counteract TLS formation and function.
Collapse
Affiliation(s)
- Haonan Lu
- Department of Surgery and Cancer, Imperial College, Hammersmith Campus, The Commonwealth Building, Du Cane Road, W12 0NN London, UK
| | - Hantao Lou
- Ludwig Cancer Research, Nuffield Department of Medicine, University of Oxford, OX3 7DQ Oxford, UK
| | - Georg Wengert
- Department of Surgery and Cancer, Imperial College, Hammersmith Campus, The Commonwealth Building, Du Cane Road, W12 0NN London, UK
| | - Reema Paudel
- Department of Surgery and Cancer, Imperial College, Hammersmith Campus, The Commonwealth Building, Du Cane Road, W12 0NN London, UK
| | - Naina Patel
- Department of Surgery and Cancer, Imperial College, Hammersmith Campus, The Commonwealth Building, Du Cane Road, W12 0NN London, UK
| | - Saral Desai
- Imperial College Healthcare NHS Trust, Du Cane Road, W12 0HS London, UK
| | - Bill Crum
- Department of Surgery and Cancer, Imperial College, Hammersmith Campus, The Commonwealth Building, Du Cane Road, W12 0NN London, UK
| | - Kristofer Linton-Reid
- Department of Surgery and Cancer, Imperial College, Hammersmith Campus, The Commonwealth Building, Du Cane Road, W12 0NN London, UK
| | - Mitchell Chen
- Department of Surgery and Cancer, Imperial College, Hammersmith Campus, The Commonwealth Building, Du Cane Road, W12 0NN London, UK; Imperial College Healthcare NHS Trust, Du Cane Road, W12 0HS London, UK
| | - Dongyang Li
- Department of Surgery and Cancer, Imperial College, Hammersmith Campus, The Commonwealth Building, Du Cane Road, W12 0NN London, UK
| | - Jacey Ip
- Department of Surgery and Cancer, Imperial College, Hammersmith Campus, The Commonwealth Building, Du Cane Road, W12 0NN London, UK; Imperial College Healthcare NHS Trust, Du Cane Road, W12 0HS London, UK
| | - Francesco Mauri
- Department of Surgery and Cancer, Imperial College, Hammersmith Campus, The Commonwealth Building, Du Cane Road, W12 0NN London, UK
| | - David J Pinato
- Department of Surgery and Cancer, Imperial College, Hammersmith Campus, The Commonwealth Building, Du Cane Road, W12 0NN London, UK; Division of Oncology, Department of Translational Medicine, University of Piemonte Orientale, Novara, Italy
| | - Andrea Rockall
- Department of Surgery and Cancer, Imperial College, Hammersmith Campus, The Commonwealth Building, Du Cane Road, W12 0NN London, UK
| | - Susan J Copley
- Department of Surgery and Cancer, Imperial College, Hammersmith Campus, The Commonwealth Building, Du Cane Road, W12 0NN London, UK; Imperial College Healthcare NHS Trust, Du Cane Road, W12 0HS London, UK
| | - Sadaf Ghaem-Maghami
- Department of Surgery and Cancer, Imperial College, Hammersmith Campus, The Commonwealth Building, Du Cane Road, W12 0NN London, UK; Imperial College Healthcare NHS Trust, Du Cane Road, W12 0HS London, UK
| | - Eric O Aboagye
- Department of Surgery and Cancer, Imperial College, Hammersmith Campus, The Commonwealth Building, Du Cane Road, W12 0NN London, UK.
| |
Collapse
|
114
|
Kumar P, Ranmale S, Mehta S, Tongaonkar H, Patel V, Singh AK, Mania-Pramanik J. Immune profile of primary and recurrent epithelial ovarian cancer cases indicates immune suppression, a major cause of progression and relapse of ovarian cancer. J Ovarian Res 2023; 16:114. [PMID: 37322531 DOI: 10.1186/s13048-023-01192-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Accepted: 05/24/2023] [Indexed: 06/17/2023] Open
Abstract
BACKGROUND Ovarian cancer is the third most prevalent cancer in Indian women. Relative frequency of High grade serous epithelial ovarian cancer (HGSOC) and its associated deaths are highest in India which suggests the importance of understanding their immune profiles for better treatment modality. Hence, the present study investigated the NK cell receptor expression, their cognate ligands, serum cytokines, and soluble ligands in primary and recurrent HGSOC patients. We have used multicolor flow cytometry for immunophenotyping of tumor infiltrated and circulatory lymphocytes. Procartaplex, and ELISA were used to measure soluble ligands and cytokines of HGSOC patients. RESULTS Among the enrolled 51 EOC patients, 33 were primary high grade serous epithelial ovarian cancer (pEOC) and 18 were recurrent epithelial ovarian cancer (rEOC) patients. Blood samples from 46 age matched healthy controls (HC) were used for comparative analysis. Results revealed, frequency of circulatory CD56Bright NK, CD56Dim NK, NKT-like, and T cells was reduced with activating receptors while alterations in immune subsets with inhibitory receptors were observed in both groups. Study also highlights differential immune profile of primary and recurrent ovarian cancer patients. We have found increased soluble MICA which might have acted as "decoy" molecule and could be a reason of decrease in NKG2D positive subsets in both groups of patients. Furthermore, elevated level of serum cytokines IL-2, IL-5, IL-6, IL-10, and TNF-α in ovarian cancer patients, might be associated with ovarian cancer progression. Profiling of tumor infiltrated immune cells revealed the reduced level of DNAM-1 positive NK and T cells in both groups than their circulatory counterpart, which might have led to decrease in NK cell's ability of synapse formation. CONCLUSIONS The study brings out differential receptor expression profile on CD56BrightNK, CD56DimNK, NKT-like, and T cells, cytokines levels and soluble ligands which may be exploited to develop alternate therapeutic approaches for HGSOC patients. Further, few differences in the circulatory immune profiles between pEOC and rEOC cases, indicates the immune signature of pEOC undergoes some changes in circulation that might facilitated the disease relapse. They also maintains some common immune signatures such as reduced expression of NKG2D, high level of MICA as well as IL-6, IL10 and TNF-α, which indicates irreversible immune suppression of ovarian cancer patients. It is also emphasized that a restoration of cytokines level, NKG2D and DNAM-1on tumor infiltrated immune cells may be targeted to develop specific therapeutic approaches for high-grade serous epithelial ovarian cancer.
Collapse
Affiliation(s)
- Pavan Kumar
- ICMR-National Institute for Research in Reproductive and Child Health, Jehangir Merwanji Street, Parel, Mumbai, 400012, India
| | - Samruddhi Ranmale
- ICMR-National Institute for Research in Reproductive and Child Health, Jehangir Merwanji Street, Parel, Mumbai, 400012, India
| | | | - Hemant Tongaonkar
- P. D. Hinduja National Hospital & Medical Research Centre, Mumbai, 400016, India
| | - Vainav Patel
- ICMR-National Institute for Research in Reproductive and Child Health, Jehangir Merwanji Street, Parel, Mumbai, 400012, India
| | - Amit Kumar Singh
- ICMR-National Institute for Research in Reproductive and Child Health, Jehangir Merwanji Street, Parel, Mumbai, 400012, India
| | - Jayanti Mania-Pramanik
- ICMR-National Institute for Research in Reproductive and Child Health, Jehangir Merwanji Street, Parel, Mumbai, 400012, India.
| |
Collapse
|
115
|
Handley KF, Mehta S, Martin AL, Biswas S, Maharaj K, Nagy MZ, Mine JA, Cortina C, Yu X, Sprenger K, Mandal G, Innamarato P, Powers JJ, Harro CM, Chaurio RA, Anadon CM, Shahzad MM, Flores I, Conejo-Garcia JR. Actionable spontaneous antibody responses antagonize malignant progression in ovarian carcinoma. Gynecol Oncol 2023; 173:114-121. [PMID: 37121178 PMCID: PMC10701373 DOI: 10.1016/j.ygyno.2023.03.020] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Revised: 03/21/2023] [Accepted: 03/25/2023] [Indexed: 05/02/2023]
Abstract
OBJECTIVE To demonstrate that shared antibody responses in endometriosis and endometriosis-associated ovarian cancer spontaneously antagonize malignant progression and can be leveraged to develop future immunotherapies. METHODS B cells from cyopreserved clear cell ovarian carcinoma (CCC, n = 2), endometrioid ovarian carcinoma (EC, n = 2), and endometriomas (n = 2) were isolated, activated, and EBV-immortalized. Antibodies were purified from B cell supernatants and used for screening arrays containing most of the human proteome. Targets were prioritized based on accessibility (transmembrane or secreted proteins), expression in endometriosis and cancer, and concurrent IgA and IgG responses. We focused on antibodies targeting tumor-promoting syndecan binding protein (SDCBP) to demonstrate anti-tumor activity. Immunoblots and qPCR were performed to assess SDCBP expression in ovarian cancer and endometriosis cell lines and tumor samples. Recombinant IgG4 was generated using the variable heavy and light chains of dominant B cell receptors (BCRs) reacting against the extracellular domain of SDCBP, and used in in vivo studies in human CCC- and high-grade serous ovarian carcinoma (HGSOC)-bearing immunodeficient mice. RESULTS Nine accessible proteins detected by both IgA and IgG were identified in all samples - including SDCBP, which is expressed in ovarian carcinomas of multiple histologies. Administration of α-SDCBP IgG4 in OVCAR3 (HGSOC), TOV21G and RMG-I (CCC) tumor-bearing mice significantly decreased tumor volume compared to control irrelevant IgG4. CONCLUSIONS Spontaneous antibody responses exert suboptimal but measurable immune pressure against malignant progression in ovarian carcinomas. Using tumor-derived antibodies for developing novel immunotherapeutics warrants further investigation.
Collapse
Affiliation(s)
- Katelyn F Handley
- Department of Gynecologic Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA; Division of Gynecologic Oncology, Department of Obstetrics & Gynecology, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA.
| | - Sumit Mehta
- Department of Gynecologic Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA; Division of Gynecologic Oncology, Department of Obstetrics & Gynecology, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| | - Alexandra L Martin
- Department of Clinical Science, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA; University of Tennessee Health Science Center/West Cancer Clinic, Memphis, TN 38138, USA
| | - Subir Biswas
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA; Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar, Navi Mumbai-410210, India
| | - Kamira Maharaj
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| | - Mate Z Nagy
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| | - Jessica A Mine
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA; Department of Immunology, Duke School of Medicine, Durham, NC 27710, USA; Duke Cancer Institute, Duke School of Medicine, Durham, NC 27710, USA
| | - Carla Cortina
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| | - Xiaoqing Yu
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| | - Kimberly Sprenger
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| | - Gunjan Mandal
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA; Division of Cancer Biology, DBT-Institute of Life Sciences, Bhubaneswar- 751023, India
| | - Patrick Innamarato
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| | - John J Powers
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| | - Carly M Harro
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| | - Ricardo A Chaurio
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA; Department of Immunology, Duke School of Medicine, Durham, NC 27710, USA; Duke Cancer Institute, Duke School of Medicine, Durham, NC 27710, USA
| | - Carmen M Anadon
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA; Department of Immunology, Duke School of Medicine, Durham, NC 27710, USA; Duke Cancer Institute, Duke School of Medicine, Durham, NC 27710, USA
| | - Mian M Shahzad
- Department of Gynecologic Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA; Department of Oncologic Sciences, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| | - Idhaliz Flores
- Departments of Basic Sciences and Obstetrics & Gynecology, Ponce Health Sciences University, Ponce, PR 00716, USA
| | - José R Conejo-Garcia
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA; Department of Immunology, Duke School of Medicine, Durham, NC 27710, USA; Duke Cancer Institute, Duke School of Medicine, Durham, NC 27710, USA
| |
Collapse
|
116
|
Hayashi Y, Makino T, Sato E, Ohshima K, Nogi Y, Kanemura T, Honma K, Yamashita K, Saito T, Tanaka K, Yamamoto K, Takahashi T, Kurokawa Y, Miyata H, Nakajima K, Wada H, Morii E, Eguchi H, Doki Y. Density and maturity of peritumoral tertiary lymphoid structures in oesophageal squamous cell carcinoma predicts patient survival and response to immune checkpoint inhibitors. Br J Cancer 2023; 128:2175-2185. [PMID: 37016103 PMCID: PMC10241865 DOI: 10.1038/s41416-023-02235-9] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Revised: 03/06/2023] [Accepted: 03/14/2023] [Indexed: 04/06/2023] Open
Abstract
BACKGROUND Tertiary lymphoid structures (TLSs) are ectopic lymphoid aggregates in non-lymphoid tissues, which are associated with improved prognosis in some cancer types. This study aimed to investigate the clinical significance of TLSs in oesophageal cancer (EC). METHODS In a series of 316 EC surgical specimens from two different institutes, we evaluated the density and maturity of peritumoral TLSs using haematoxylin/eosin, immunohistochemistry, and multiplex immunofluorescence staining. We analysed the association between TLSs and clinicopathological parameters. The clinical significance of TLSs was further evaluated in a different cohort of 34 patients with recurrent EC treated with anti-PD-1 antibody. RESULTS Tumours with high TLS density predominantly consisted of matured TLSs. High TLS density was significantly associated with less advanced tumour stage, absence of lymphatic/vascular invasion, better serum nutrition parameters (neutrophils count, albumin, neutrophil-to-lymphocyte ratio, and prognostic nutritional index), and prolonged survival. This survival trend was more remarkable in cases with matured TLSs, which represented an increased population of CD138+ plasma cells. In the second EC cohort, TLS density predicted the clinical response to anti-PD-1 antibody and patient survival. CONCLUSION The density and maturity of peritumoral TLSs are useful parameters for predicting long-term survival and response to anti-PD-1 antibody treatment in EC patients.
Collapse
Affiliation(s)
- Yoshinori Hayashi
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan
- Department of Clinical Research in Tumor Immunology, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Tomoki Makino
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan.
| | - Eiichi Sato
- Department of Pathology, Institute of Medical Science (Medical Research Center), Tokyo Medical University, Tokyo, Japan
| | - Kenji Ohshima
- Department of Pathology, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Yuya Nogi
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Takashi Kanemura
- Department of Gastroenterological Surgery, Osaka International Cancer Institute, Osaka, Japan
| | - Keiichiro Honma
- Department of Pathology, Osaka International Cancer Institute, Osaka, Japan
| | - Kotaro Yamashita
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Takuro Saito
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Koji Tanaka
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Kazuyoshi Yamamoto
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Tsuyoshi Takahashi
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Yukinori Kurokawa
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Hiroshi Miyata
- Department of Gastroenterological Surgery, Osaka International Cancer Institute, Osaka, Japan
| | - Kiyokazu Nakajima
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Hisashi Wada
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan
- Department of Clinical Research in Tumor Immunology, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Eiichi Morii
- Department of Pathology, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Hidetoshi Eguchi
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Yuichiro Doki
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan
| |
Collapse
|
117
|
Pankowska KA, Będkowska GE, Chociej-Stypułkowska J, Rusak M, Dąbrowska M, Osada J. Crosstalk of Immune Cells and Platelets in an Ovarian Cancer Microenvironment and Their Prognostic Significance. Int J Mol Sci 2023; 24:ijms24119279. [PMID: 37298230 DOI: 10.3390/ijms24119279] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 05/17/2023] [Accepted: 05/23/2023] [Indexed: 06/12/2023] Open
Abstract
Ovarian cancer (OC) is one of the deadliest gynecological cancers, largely due to the fast development of metastasis and drug resistance. The immune system is a critical component of the OC tumor microenvironment (TME) and immune cells such as T cells, NK cells, and dendritic cells (DC) play a key role in anti-tumor immunity. However, OC tumor cells are well known for evading immune surveillance by modulating the immune response through various mechanisms. Recruiting immune-suppressive cells such as regulatory T cells (Treg cells), macrophages, or myeloid-derived suppressor cells (MDSC) inhibit the anti-tumor immune response and promote the development and progression of OC. Platelets are also involved in immune evasion by interaction with tumor cells or through the secretion of a variety of growth factors and cytokines to promote tumor growth and angiogenesis. In this review, we discuss the role and contribution of immune cells and platelets in TME. Furthermore, we discuss their potential prognostic significance to help in the early detection of OC and to predict disease outcome.
Collapse
Affiliation(s)
- Katarzyna Aneta Pankowska
- Department of Haematological Diagnostics, Medical University of Bialystok, Waszyngtona 15A Street, 15-269 Bialystok, Poland
| | - Grażyna Ewa Będkowska
- Department of Haematological Diagnostics, Medical University of Bialystok, Waszyngtona 15A Street, 15-269 Bialystok, Poland
| | - Joanna Chociej-Stypułkowska
- Department of Haematological Diagnostics, Medical University of Bialystok, Waszyngtona 15A Street, 15-269 Bialystok, Poland
| | - Małgorzata Rusak
- Department of Haematological Diagnostics, Medical University of Bialystok, Waszyngtona 15A Street, 15-269 Bialystok, Poland
| | - Milena Dąbrowska
- Department of Haematological Diagnostics, Medical University of Bialystok, Waszyngtona 15A Street, 15-269 Bialystok, Poland
| | - Joanna Osada
- Department of Haematological Diagnostics, Medical University of Bialystok, Waszyngtona 15A Street, 15-269 Bialystok, Poland
| |
Collapse
|
118
|
Quesada S, Lebreton C, Caux C, Italiano A, Dubois B. [Tertiary lymphoid structures in cancer: From biology to therapeutic guides]. Bull Cancer 2023:S0007-4551(23)00205-9. [PMID: 37150731 DOI: 10.1016/j.bulcan.2023.04.010] [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: 03/09/2023] [Revised: 04/06/2023] [Accepted: 04/17/2023] [Indexed: 05/09/2023]
Abstract
Tertiary lymphoid structures (TLS) are inducible ectopic lymphoid aggregates, which form in response to various inflammatory situations, including cancer. TLS are notably composed of B lymphocytes, T lymphocytes, mature dendritic cells and other key players such as high endothelial venules. Furthermore, TLS can present different levels of organization and maturation, from simple T/B lymphocyte aggregates to authentic mature B cell follicles with germinal centers adjacent to T cell rich areas. While over the past decade, TLS may have been associated with a favorable prognosis in various cancers, the year 2022 was marked by the first prospective trial (PEMBROSARC) that reported the interest of TLS as predictive biomarkers of pembrolizumab efficacy for the treatment of soft-tissue sarcomas. All along this review, we will first address the molecular and cellular bases of TLS as well as the different strategies for identifying them in clinical practice, then discuss the prognostic/predictive impact of their presence and finally, we will elaborate on the current limitations and perspectives in translational research.
Collapse
Affiliation(s)
- Stanislas Quesada
- Institut régional du cancer de Montpellier - ICM (UNICANCER), département d'oncologie médicale, Montpellier, France.
| | - Coriolan Lebreton
- Institut Bergonié (UNICANCER), département d'oncologie médicale, Bordeaux, France; ARTiSt Lab, Inserm U1312, université de Bordeaux, Bordeaux, France
| | - Christophe Caux
- Centre Léon Bérard, CNRS 5286, Inserm 1052, université Claude Bernard Lyon 1, université de Lyon, centre de recherche en cancérologie de Lyon (CRCL), Équipe "Surveillance immunitaire des tumeurs et ciblage thérapeutique", Lyon, France; Laboratoire d'immunothérapie du cancer de Lyon (LICL), Lyon, France
| | - Antoine Italiano
- Institut Bergonié (UNICANCER), département d'oncologie médicale, Bordeaux, France; Université de Bordeaux, faculté de médecine, Bordeaux, France; DITEP, institut Gustave Roussy - IGR (UNICANCER), Villejuif, France
| | - Bertrand Dubois
- Centre Léon Bérard, CNRS 5286, Inserm 1052, université Claude Bernard Lyon 1, université de Lyon, centre de recherche en cancérologie de Lyon (CRCL), Équipe "Surveillance immunitaire des tumeurs et ciblage thérapeutique", Lyon, France; Laboratoire d'immunothérapie du cancer de Lyon (LICL), Lyon, France
| |
Collapse
|
119
|
Ferri-Borgogno S, Zhu Y, Sheng J, Burks JK, Gomez JA, Wong KK, Wong ST, Mok SC. Spatial Transcriptomics Depict Ligand-Receptor Cross-talk Heterogeneity at the Tumor-Stroma Interface in Long-Term Ovarian Cancer Survivors. Cancer Res 2023; 83:1503-1516. [PMID: 36787106 PMCID: PMC10159916 DOI: 10.1158/0008-5472.can-22-1821] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 12/06/2022] [Accepted: 02/10/2023] [Indexed: 02/15/2023]
Abstract
Advanced high-grade serous ovarian cancer (HGSC) is an aggressive disease that accounts for 70% of all ovarian cancer deaths. Nevertheless, 15% of patients diagnosed with advanced HGSC survive more than 10 years. The elucidation of predictive markers of these long-term survivors (LTS) could help identify therapeutic targets for the disease, and thus improve patient survival rates. To investigate the stromal heterogeneity of the tumor microenvironment (TME) in ovarian cancer, we used spatial transcriptomics to generate spatially resolved transcript profiles in treatment-naïve advanced HGSC from LTS and short-term survivors (STS) and determined the association between cancer-associated fibroblasts (CAF) heterogeneity and survival in patients with advanced HGSC. Spatial transcriptomics and single-cell RNA-sequencing data were integrated to distinguish tumor and stroma regions, and a computational method was developed to investigate spatially resolved ligand-receptor interactions between various tumor and CAF subtypes in the TME. A specific subtype of CAFs and its spatial location relative to a particular ovarian cancer cell subtype in the TME correlated with long-term survival in patients with advanced HGSC. Also, increased APOE-LRP5 cross-talk occurred at the stroma-tumor interface in tumor tissues from STS compared with LTS. These findings were validated using multiplex IHC. Overall, this spatial transcriptomics analysis revealed spatially resolved CAF-tumor cross-talk signaling networks in the ovarian TME that are associated with long-term survival of patients with HGSC. Further studies to confirm whether such cross-talk plays a role in modulating the malignant phenotype of HGSC and could serve as a predictive biomarker of patient survival are warranted. SIGNIFICANCE Generation of spatially resolved gene expression patterns in tumors from patients with ovarian cancer surviving more than 10 years allows the identification of novel predictive biomarkers and therapeutic targets for better patient management. See related commentary by Kelliher and Lengyel, p. 1383.
Collapse
Affiliation(s)
- Sammy Ferri-Borgogno
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Ying Zhu
- Systems Medicine and Bioengineering Department, Houston Methodist Cancer Center, Houston Methodist Hospital, Houston, TX 77030, USA
- Departments of Pathology and Laboratory Medicine and Radiology, Houston Methodist Hospital, Weill Cornell Medicine, Houston, TX 77030, USA
| | - Jianting Sheng
- Systems Medicine and Bioengineering Department, Houston Methodist Cancer Center, Houston Methodist Hospital, Houston, TX 77030, USA
- Departments of Pathology and Laboratory Medicine and Radiology, Houston Methodist Hospital, Weill Cornell Medicine, Houston, TX 77030, USA
| | - Jared K. Burks
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Javier A. Gomez
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Kwong Kwok Wong
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Stephen T.C. Wong
- Systems Medicine and Bioengineering Department, Houston Methodist Cancer Center, Houston Methodist Hospital, Houston, TX 77030, USA
- Departments of Pathology and Laboratory Medicine and Radiology, Houston Methodist Hospital, Weill Cornell Medicine, Houston, TX 77030, USA
| | - Samuel C. Mok
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
120
|
Zorea J, Motro Y, Mazor RD, Carmi YK, Shulman Z, Mahajna J, Moran-Gilad J, Elkabets M. TRAF3 suppression encourages B cell recruitment and prolongs survival of microbiome-intact mice with ovarian cancer. J Exp Clin Cancer Res 2023; 42:107. [PMID: 37121997 PMCID: PMC10150478 DOI: 10.1186/s13046-023-02680-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 04/22/2023] [Indexed: 05/02/2023] Open
Abstract
BACKGROUND Ovarian cancer (OC) is known for exhibiting low response rates to immune checkpoint inhibitors that activate T cells. However, immunotherapies that activate B cells have not yet been extensively explored and may be a potential target, as B cells that secrete immunoglobulins have been associated with better outcomes in OC. Although the secretion of immunoglobulins is often mediated by the microbiome, it is still unclear what role they play in limiting the progression of OC. METHODS We conducted an in-vivo CRISPR screen of immunodeficient (NSG) and immune-intact wild type (WT) C57/BL6 mice to identify tumor-derived immune-escape mechanisms in a BRAC1- and TP53-deficient murine ID8 OC cell line (designated ITB1). To confirm gene expression and signaling pathway activation in ITB1 cells, we employed western blot, qPCR, immunofluorescent staining, and flow cytometry. Flow cytometry was also used to identify immune cell populations in the peritoneum of ITB1-bearing mice. To determine the presence of IgA-coated bacteria in the peritoneum of ITB1-bearing mice and the ascites of OC patients, we employed 16S sequencing. Testing for differences was done by using Deseq2 test and two-way ANOVA test. Sequence variants (ASVs) were produced in Qiime2 and analyzed by microeco and phyloseq R packages. RESULTS We identified tumor necrosis factor receptor-associated factor 3 (TRAF3) as a tumor-derived immune suppressive mediator in ITB1 cells. Knockout of TRAF3 (TRAF3KO) activated the type-I interferon pathway and increased MHC-I expression. TRAF3KO tumors exhibited a growth delay in WT mice vs. NSG mice, which was correlated with increased B cell infiltration and activation compared to ITB1 tumors. B cells were found to be involved in the progression of TRAF3KO tumors, and B-cell surface-bound and secreted IgA levels were significantly higher in the ascites of TRAF3KO tumors compared to ITB1. The presence of commensal microbiota was necessary for B-cell activation and for delaying the progression of TRAF3KO tumors in WT mice. Lastly, we observed unique profiles of IgA-coated bacteria in the ascites of OC-bearing mice or the ascites of OC patients. CONCLUSIONS TRAF3 is a tumor-derived immune-suppressive modulator that influences B-cell infiltration and activation, making it a potential target for enhancing anti-tumor B-cell responses in OC.
Collapse
Affiliation(s)
- Jonathan Zorea
- Shraga Segal Department of Microbiology, Immunology, and Genetics, Ben-Gurion University of the Negev, 8410501, Beer-Sheva, Israel
- Department of Health Policy and Management, School of Public Health, Faculty of Health Sciences, Ben-Gurion University of the Negev, 8410501, Beer-Sheva, Israel
| | - Yair Motro
- Department of Health Policy and Management, School of Public Health, Faculty of Health Sciences, Ben-Gurion University of the Negev, 8410501, Beer-Sheva, Israel
| | - Roei D Mazor
- Department of Systems Immunology, Weizmann Institute of Science, 7610001, Rehovot, Israel
| | - Yifat Koren Carmi
- Department of Nutrition and Natural Products, Migal-Galilee Research Institute, 11016, Kiryat Shemona, Israel
| | - Ziv Shulman
- Department of Systems Immunology, Weizmann Institute of Science, 7610001, Rehovot, Israel
| | - Jamal Mahajna
- Department of Nutrition and Natural Products, Migal-Galilee Research Institute, 11016, Kiryat Shemona, Israel
| | - Jacob Moran-Gilad
- Department of Health Policy and Management, School of Public Health, Faculty of Health Sciences, Ben-Gurion University of the Negev, 8410501, Beer-Sheva, Israel
| | - Moshe Elkabets
- Shraga Segal Department of Microbiology, Immunology, and Genetics, Ben-Gurion University of the Negev, 8410501, Beer-Sheva, Israel.
- Department of Health Policy and Management, School of Public Health, Faculty of Health Sciences, Ben-Gurion University of the Negev, 8410501, Beer-Sheva, Israel.
| |
Collapse
|
121
|
Shapiro DD, Dolan B, Laklouk IA, Rassi S, Lozar T, Emamekhoo H, Wentland AL, Lubner MG, Abel EJ. Understanding the Tumor Immune Microenvironment in Renal Cell Carcinoma. Cancers (Basel) 2023; 15:cancers15092500. [PMID: 37173966 PMCID: PMC10177515 DOI: 10.3390/cancers15092500] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 04/24/2023] [Accepted: 04/24/2023] [Indexed: 05/15/2023] Open
Abstract
Scientific understanding of how the immune microenvironment interacts with renal cell carcinoma (RCC) has substantially increased over the last decade as a result of research investigations and applying immunotherapies, which modulate how the immune system targets and eliminates RCC tumor cells. Clinically, immune checkpoint inhibitor therapy (ICI) has revolutionized the treatment of advanced clear cell RCC because of improved outcomes compared to targeted molecular therapies. From an immunologic perspective, RCC is particularly interesting because tumors are known to be highly inflamed, but the mechanisms underlying the inflammation of the tumor immune microenvironment are atypical and not well described. While technological advances in gene sequencing and cellular imaging have enabled precise characterization of RCC immune cell phenotypes, multiple theories have been suggested regarding the functional significance of immune infiltration in RCC progression. The purpose of this review is to describe the general concepts of the anti-tumor immune response and to provide a detailed summary of the current understanding of the immune response to RCC tumor development and progression. This article describes immune cell phenotypes that have been reported in the RCC microenvironment and discusses the application of RCC immunophenotyping to predict response to ICI therapy and patient survival.
Collapse
Affiliation(s)
- Daniel D Shapiro
- Department of Urology, The University of Wisconsin School of Medicine and Public Health, Madison, WI 53726, USA
- Division of Urology, William S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA
| | - Brendan Dolan
- Department of Urology, The University of Wisconsin School of Medicine and Public Health, Madison, WI 53726, USA
| | - Israa A Laklouk
- Department of Pathology, The University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
| | - Sahar Rassi
- Department of Urology, The University of Wisconsin School of Medicine and Public Health, Madison, WI 53726, USA
| | - Taja Lozar
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53726, USA
| | - Hamid Emamekhoo
- Department of Medical Oncology, The University of Wisconsin School of Medicine and Public Health, Madison, WI 53726, USA
| | - Andrew L Wentland
- Department of Radiology, The University of Wisconsin School of Medicine and Public Health, Madison, WI 53726, USA
| | - Meghan G Lubner
- Department of Radiology, The University of Wisconsin School of Medicine and Public Health, Madison, WI 53726, USA
| | - Edwin Jason Abel
- Department of Urology, The University of Wisconsin School of Medicine and Public Health, Madison, WI 53726, USA
| |
Collapse
|
122
|
Wen Z, Liu H, Qiao D, Chen H, Li L, Yang Z, Zhu C, Zeng Z, Chen Y, Liu L. Nanovaccines Fostering Tertiary Lymphoid Structure to Attack Mimicry Nasopharyngeal Carcinoma. ACS NANO 2023; 17:7194-7206. [PMID: 37057967 DOI: 10.1021/acsnano.2c09619] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/09/2023]
Abstract
Tertiary lymphoid structures (TLSs) are formed in inflamed tissues, and recent studies demonstrated that the appearance of TLSs in tumor sites is associated with a good prognosis for tumor patients. However, the process of natural TLSs' formation was slow and uncontrollable. Herein, we developed a nanovaccine consisting of Epstein-Barr virus nuclear antigen 1 (EBNA1) and a bi-adjuvant of Mn2+ and cytosine-phosphate-guanine (CpG) formulated with tannic acid that significantly inhibited the development of mimicry nasopharyngeal carcinoma by fostering TLS formation. The nanovaccine activated LT-α and LT-β pathways, subsequently enhancing the expression of downstream chemokines, CCL19/CCL21, CXCL10 and CXCL13, in the tumor microenvironment. In turn, normalized blood and lymph vessels were detected in the tumor tissues of the nanovaccine group, correlated with increased infiltration of lymphocytes. Especially, the proportion of the B220+ CD8+ T, which was produced via trogocytosis between T and B cells during activation of T cells, was increased in tumors of the nanovaccine group. Furthermore, the intratumoral effector memory T cells (Tem), CD45+, CD3+, CD8+, CD44+, and CD62L-, did not decrease after blocking the egress of T cells from tumor-draining lymph nodes by FTY-720. These results demonstrated that the nanovaccine can foster TLS formation, which thus enhances local immune responses significantly, delays tumor outgrowth, and prolongs the median survival time of murine models of mimicry nasopharyngeal carcinoma, demonstrating a promising strategy for nanovaccine development.
Collapse
Affiliation(s)
- Zhenfu Wen
- School of Materials Science and Engineering, Key Laboratory for Polymeric Composite and Functional Materials of Ministry of Education, Sun Yat-sen University, Guangzhou 510275, China
| | - Hong Liu
- School of Materials Science and Engineering, Key Laboratory for Polymeric Composite and Functional Materials of Ministry of Education, Sun Yat-sen University, Guangzhou 510275, China
| | - Dongdong Qiao
- School of Materials Science and Engineering, Key Laboratory for Polymeric Composite and Functional Materials of Ministry of Education, Sun Yat-sen University, Guangzhou 510275, China
| | - Haolin Chen
- School of Materials Science and Engineering, Key Laboratory for Polymeric Composite and Functional Materials of Ministry of Education, Sun Yat-sen University, Guangzhou 510275, China
| | - Liyan Li
- School of Materials Science and Engineering, Key Laboratory for Polymeric Composite and Functional Materials of Ministry of Education, Sun Yat-sen University, Guangzhou 510275, China
| | - Zeyu Yang
- School of Materials Science and Engineering, Key Laboratory for Polymeric Composite and Functional Materials of Ministry of Education, Sun Yat-sen University, Guangzhou 510275, China
| | - Chenxu Zhu
- School of Materials Science and Engineering, Key Laboratory for Polymeric Composite and Functional Materials of Ministry of Education, Sun Yat-sen University, Guangzhou 510275, China
| | - Zhipeng Zeng
- School of Materials Science and Engineering, Key Laboratory for Polymeric Composite and Functional Materials of Ministry of Education, Sun Yat-sen University, Guangzhou 510275, China
| | - Yongming Chen
- School of Materials Science and Engineering, Key Laboratory for Polymeric Composite and Functional Materials of Ministry of Education, Sun Yat-sen University, Guangzhou 510275, China
- State Key Laboratory of Oncology in Southern China, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Lixin Liu
- School of Materials Science and Engineering, Key Laboratory for Polymeric Composite and Functional Materials of Ministry of Education, Sun Yat-sen University, Guangzhou 510275, China
- State Key Laboratory of Oncology in Southern China, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| |
Collapse
|
123
|
Huang R, Zhao B, Hu S, Zhang Q, Su X, Zhang W. Adoptive neoantigen-reactive T cell therapy: improvement strategies and current clinical researches. Biomark Res 2023; 11:41. [PMID: 37062844 PMCID: PMC10108522 DOI: 10.1186/s40364-023-00478-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 03/21/2023] [Indexed: 04/18/2023] Open
Abstract
Neoantigens generated by non-synonymous mutations of tumor genes can induce activation of neoantigen-reactive T (NRT) cells which have the ability to resist the growth of tumors expressing specific neoantigens. Immunotherapy based on NRT cells has made preeminent achievements in melanoma and other solid tumors. The process of manufacturing NRT cells includes identification of neoantigens, preparation of neoantigen expression vectors or peptides, induction and activation of NRT cells, and analysis of functions and phenotypes. Numerous improvement strategies have been proposed to enhance the potency of NRT cells by engineering TCR, promoting infiltration of T cells and overcoming immunosuppressive factors in the tumor microenvironment. In this review, we outline the improvement of the preparation and the function assessment of NRT cells, and discuss the current status of clinical trials related to NRT cell immunotherapy.
Collapse
Affiliation(s)
- Ruichen Huang
- Department of Respiratory and Critical Care Medicine, the First Affiliated Hospital of Second Military Medical University, Shanghai, 200433, People's Republic of China
| | - Bi Zhao
- Department of Respiratory and Critical Care Medicine, the First Affiliated Hospital of Second Military Medical University, Shanghai, 200433, People's Republic of China
| | - Shi Hu
- Department of Biophysics, College of Basic Medical Sciences, Second Military Medical University, 800 Xiangyin Road, Shanghai, 200433, People's Republic of China
| | - Qian Zhang
- National Key Laboratory of Medical Immunology, Institute of Immunology, Second Military Medical University, 800 Xiangyin Road, Shanghai, 200433, People's Republic of China
| | - Xiaoping Su
- School of Basic Medicine, Wenzhou Medical University, Wenzhou, 325000, People's Republic of China.
| | - Wei Zhang
- Department of Respiratory and Critical Care Medicine, the First Affiliated Hospital of Second Military Medical University, Shanghai, 200433, People's Republic of China.
| |
Collapse
|
124
|
Zeng L, Koh VCY, Chen XY, Tan PH. Tertiary lymphoid structures in breast ductal carcinoma in situ correlate with adverse pathological parameters. Histopathology 2023; 82:779-788. [PMID: 36635954 DOI: 10.1111/his.14865] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 12/17/2022] [Accepted: 01/07/2023] [Indexed: 01/14/2023]
Abstract
AIMS To investigate tertiary lymphoid structures (TLSs) in ductal carcinoma in situ (DCIS) of the breast and their correlation with pathological features, immune cell markers and clinical outcomes. METHODS AND RESULTS Morphological identification of TLSs in 198 DCIS cases incorporated B and T cell zones with high endothelial venules. TLS positivity was defined as ≥ 1 TLSs in lesional areas, while TLS area percentage was divided into two categories: low (TLSs < 5%) and high (TLSs ≥ 5%). Previously reported biomarkers included ER, PR, HER2, CD68, CD163, CD4, CD8 and PD-L1. TLSs were observed in 24.7% (49 of 198) of cases, with a mean diameter of 0.44 mm (median = 0.4 mm, range = 0.12-1.43 mm). TLSs were significantly associated with higher nuclear grade, presence of necrosis, hormone receptor negativity/HER2 positivity, triple negativity, tumour infiltrating lymphocytes (TILs) and immune related biomarkers such as FOXP3, CD163, CD4 and CD4/CD8 ratio (all P < 0.05). There were no significant associations between TLSs and recurrence, but a combination of TLSshigh with FOXP3+ , CD4high , CD4/CD8 ratiohigh and CD68high individually, compared with all other combinations, disclosed significantly poorer disease-free survival (DFS) for ipsilateral invasive recurrence (IIR) on both Kaplan-Meier and multivariable Cox regression analyses (all P < 0.05). CONCLUSIONS TLSs in DCIS were associated with unfavourable prognostic features, TILs and immune cell markers in our study. TLSshigh /FoxP3+ , TLSshigh /CD4high , TLSshigh /(CD4/CD8) ratiohigh and TLSshigh /CD68high were independent factors for poorer DFS for IIR. Further exploration of the pathological significance of TLSs may provide a clinical basis for their recognition as an important structure and functional unit in the tumour immune microenvironment.
Collapse
Affiliation(s)
- Lixia Zeng
- Department of Anatomical Pathology, Singapore General Hospital, Singapore, Singapore.,Department of Pathology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, China
| | - Valerie Cui Yun Koh
- Department of Anatomical Pathology, Singapore General Hospital, Singapore, Singapore
| | - Xiao-Yang Chen
- Department of Anatomical Pathology, Singapore General Hospital, Singapore, Singapore.,Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Puay Hoon Tan
- Luma Medical Centre, Singapore, Singapore.,Department of Pathology and Laboratory Medicine, KK Women's and Children's Hospital, Singapore, Singapore.,Department of Pathology, University of Western Sydney, Sydney, Australia
| |
Collapse
|
125
|
He M, He Q, Cai X, Liu J, Deng H, Li F, Zhong R, Lu Y, Peng H, Wu X, Chen Z, Lao S, Li C, Li J, He J, Liang W. Intratumoral tertiary lymphoid structure (TLS) maturation is influenced by draining lymph nodes of lung cancer. J Immunother Cancer 2023; 11:jitc-2022-005539. [PMID: 37072348 PMCID: PMC10124324 DOI: 10.1136/jitc-2022-005539] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/14/2023] [Indexed: 04/20/2023] Open
Abstract
BACKGROUND Tertiary lymphoid structure (TLS) is an organized infiltration of immune cells, showing features of germinal center (GC) commonly seen in secondary lymphoid organs. However, its relationship with tumor-draining lymph nodes (TDLNs) has not been studied and we hypothesized that TDLN may influence maturation of intratumoral TLS in non-small cell lung cancer (NSCLC). METHODS Tissue slides of 616 patients that had undergone surgeries were examined. Cox proportional hazard regression model was used to assess risk factors of patients' survival, and logistic regression model was used for their relationship with TLS. Single-cell RNA-sequencing (scRNA-seq) was employed to explore transcriptomic features of TDLNs. Immunohistochemistry, multiplex immunofluorescence and flow cytometry were performed to analyze cellular composition. Cellular components of NSCLC samples from The Cancer Genome Atlas database were inferred with Microenvironment Cell Populations-counter (MCP-counter) method. Murine NSCLC models were used to dissect underlying mechanisms for relationship between TDLN and TLS maturation. RESULTS While GC+ TLS was associated with better prognosis, GC- TLS was not. TDLN metastasis reduced the prognostic relevance of TLS, and was associated with less GC formation. Primary tumor sites showed reduced B cell infiltration in TDLN-positive patients, and scRNA-seq revealed diminished memory B cell formation in tumor-invaded TDLNs, together with an emphasis on weakened interferon (IFN)-γ response. Murine NSCLC models revealed that IFN-γ signaling is involved in memory B cell differentiation in TDLNs and GC formation in primary tumors. CONCLUSIONS Our research emphasizes the influence of TDLN on intratumoral TLS maturation and suggests a role of memory B cells and IFN-γ signaling in this communication.
Collapse
Affiliation(s)
- Miao He
- Department of Thoracic Surgery and Oncology, The First Affiliated Hospital of Guangzhou Medical University, China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou, Guangdong, China
| | - Qihua He
- Department of Thoracic Surgery and Oncology, The First Affiliated Hospital of Guangzhou Medical University, China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou, Guangdong, China
| | - Xiuyu Cai
- Department of General Internal Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Jun Liu
- Department of Thoracic Surgery and Oncology, The First Affiliated Hospital of Guangzhou Medical University, China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou, Guangdong, China
| | - Hongshen Deng
- Department of Thoracic Surgery and Oncology, The First Affiliated Hospital of Guangzhou Medical University, China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou, Guangdong, China
| | - Feng Li
- Department of Thoracic Surgery and Oncology, The First Affiliated Hospital of Guangzhou Medical University, China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou, Guangdong, China
| | - Ran Zhong
- Department of Thoracic Surgery and Oncology, The First Affiliated Hospital of Guangzhou Medical University, China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou, Guangdong, China
| | - Yi Lu
- Department of Thoracic Surgery and Oncology, The First Affiliated Hospital of Guangzhou Medical University, China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou, Guangdong, China
| | - Haoxin Peng
- Department of Thoracic Surgery and Oncology, The First Affiliated Hospital of Guangzhou Medical University, China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou, Guangdong, China
| | - Xiangrong Wu
- Department of Thoracic Surgery and Oncology, The First Affiliated Hospital of Guangzhou Medical University, China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou, Guangdong, China
| | - Zisheng Chen
- Department of Thoracic Surgery and Oncology, The First Affiliated Hospital of Guangzhou Medical University, China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou, Guangdong, China
| | - Shen Lao
- Department of Thoracic Surgery and Oncology, The First Affiliated Hospital of Guangzhou Medical University, China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou, Guangdong, China
| | - Caichen Li
- Department of Thoracic Surgery and Oncology, The First Affiliated Hospital of Guangzhou Medical University, China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou, Guangdong, China
| | - Jianfu Li
- Department of Thoracic Surgery and Oncology, The First Affiliated Hospital of Guangzhou Medical University, China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou, Guangdong, China
| | - Jianxing He
- Department of Thoracic Surgery and Oncology, The First Affiliated Hospital of Guangzhou Medical University, China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou, Guangdong, China
- Department of surgery, Southern Medical University, Guangzhou, Guangdong, China
| | - Wenhua Liang
- Department of Thoracic Surgery and Oncology, The First Affiliated Hospital of Guangzhou Medical University, China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou, Guangdong, China
- Department of Medical Oncology, The First People Hospital of Zhaoqing, Zhaoqing, Guangdong, China
| |
Collapse
|
126
|
Baier E, Kluge IA, Hakroush S, Tampe D, Tampe B. Leukocyturia and hematuria enable non-invasive differentiation of Bowman's capsule rupture severity in PR3-ANCA glomerulonephritis. J Nephrol 2023; 36:799-808. [PMID: 36542276 PMCID: PMC10090024 DOI: 10.1007/s40620-022-01486-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2022] [Accepted: 10/01/2022] [Indexed: 12/24/2022]
Abstract
BACKGROUND Renal involvement is a common and severe complication of anti-neutrophil cytoplasmic antibody-(ANCA)-associated vasculitis potentially resulting in pauci-immune necrotizing and crescentic ANCA glomerulonephritis (GN) with rapid deterioration of kidney function, progression to end stage kidney disease or, if left untreated, lethal exitus. Analysis of the urinary sediment routinely supports clinical management of ANCA GN, but histopathological implications of aberrancies in the urinary sediment mostly remain elusive. Therefore, we aimed to systematically assess the correlation of aberrancies in the urinary sediment and clinico-pathologic findings. METHODS A total of 42 kidney biopsies with ANCA GN were retrospectively analyzed in a single-center observational study. Laboratory and histopathological parameters were systematically analyzed and correlated with findings of the urinary sediment. RESULTS In the overall ANCA GN cohort, leukocyturia and hematuria were associated among each other, and with markers for non-selective glomerular damage, respectively. Non-invasive measurement of leukocyturia indicated focal (but not extensive) Bowman's capsule rupture (BCR) specifically in proteinase-3 (PR3)-ANCA GN, whereas hematuria correlated with extensive (but not focal) BCR. Concerning intrarenal immune cell infiltration, leukocyturia was associated with tubulointerstitial plasma cell infiltration in PR3-ANCA GN. Finally, none of these associations were detectable in myeloperoxidase-ANCA GN, implying different modes of kidney damage. CONCLUSION We herein expand our current knowledge by providing evidence that leukocyturia and hematuria enable non-invasive differentiation of BCR severity specifically in PR3-ANCA GN.
Collapse
Affiliation(s)
- Eva Baier
- Department of Nephrology and Rheumatology, University Medical Center Göttingen, Göttingen, Germany
| | | | - Samy Hakroush
- Institute of Pathology, University Medical Center Göttingen, Göttingen, Germany
- SYNLAB Pathology Hannover, SYNLAB Holding Germany, Augsburg, Germany
| | - Désirée Tampe
- Department of Nephrology and Rheumatology, University Medical Center Göttingen, Göttingen, Germany
| | - Björn Tampe
- Department of Nephrology and Rheumatology, University Medical Center Göttingen, Göttingen, Germany.
| |
Collapse
|
127
|
Laumont CM, Nelson BH. B cells in the tumor microenvironment: Multi-faceted organizers, regulators, and effectors of anti-tumor immunity. Cancer Cell 2023; 41:466-489. [PMID: 36917951 DOI: 10.1016/j.ccell.2023.02.017] [Citation(s) in RCA: 51] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 02/11/2023] [Accepted: 02/12/2023] [Indexed: 03/14/2023]
Abstract
Our understanding of tumor-infiltrating lymphocytes (TILs) is rapidly expanding beyond T cell-centric perspectives to include B cells and plasma cells, collectively referred to as TIL-Bs. In many cancers, TIL-Bs carry strong prognostic significance and are emerging as key predictors of response to immune checkpoint inhibitors. TIL-Bs can perform multiple functions, including antigen presentation and antibody production, which allow them to focus immune responses on cognate antigen to support both T cell responses and innate mechanisms involving complement, macrophages, and natural killer cells. In the stroma of the most immunologically "hot" tumors, TIL-Bs are prominent components of tertiary lymphoid structures, which resemble lymph nodes structurally and functionally. Additionally, TIL-Bs participate in a variety of other lympho-myeloid aggregates and engage in dynamic interactions with the tumor stroma. Here, we summarize our current understanding of TIL-Bs in human cancer, highlighting the compelling therapeutic opportunities offered by their unique tumor recognition and effector mechanisms.
Collapse
Affiliation(s)
- Céline M Laumont
- Deeley Research Centre, BC Cancer, Victoria, BC V8R 6V5, Canada; Department of Medical Genetics, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Brad H Nelson
- Deeley Research Centre, BC Cancer, Victoria, BC V8R 6V5, Canada; Department of Medical Genetics, University of British Columbia, Vancouver, BC V6T 1Z3, Canada; Department of Biochemistry and Microbiology, University of Victoria, Victoria, BC V8P 3E6, Canada.
| |
Collapse
|
128
|
Vella G, Hua Y, Bergers G. High endothelial venules in cancer: Regulation, function, and therapeutic implication. Cancer Cell 2023; 41:527-545. [PMID: 36827979 DOI: 10.1016/j.ccell.2023.02.002] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 01/12/2023] [Accepted: 02/01/2023] [Indexed: 02/25/2023]
Abstract
The lack of sufficient intratumoral CD8+ T lymphocytes is a significant obstacle to effective immunotherapy in cancer. High endothelial venules (HEVs) are organ-specific and specialized postcapillary venules uniquely poised to facilitate the transmigration of lymphocytes to lymph nodes (LNs) and other secondary lymphoid organs (SLOs). HEVs can also form in human and murine cancer (tumor HEVs [TU-HEVs]) and contribute to the generation of diffuse T cell-enriched aggregates or tertiary lymphoid structures (TLSs), which are commonly associated with a good prognosis. Thus, therapeutic induction of TU-HEVs may provide attractive avenues to induce and sustain the efficacy of immunotherapies by overcoming the major restriction of T cell exclusion from the tumor microenvironment. In this review, we provide current insight into the commonalities and discrepancies of HEV formation and regulation in LNs and tumors and discuss the specific function and significance of TU-HEVs in eliciting, predicting, and aiding anti-tumoral immunity.
Collapse
Affiliation(s)
- Gerlanda Vella
- Laboratory of Tumor Microenvironment and Therapeutic Resistance, Department of Oncology, VIB-Center for Cancer Biology, KU Leuven, Leuven, Belgium
| | - Yichao Hua
- Laboratory of Tumor Microenvironment and Therapeutic Resistance, Department of Oncology, VIB-Center for Cancer Biology, KU Leuven, Leuven, Belgium
| | - Gabriele Bergers
- Laboratory of Tumor Microenvironment and Therapeutic Resistance, Department of Oncology, VIB-Center for Cancer Biology, KU Leuven, Leuven, Belgium.
| |
Collapse
|
129
|
Zhang E, Ding C, Li S, Zhou X, Aikemu B, Fan X, Sun J, Zheng M, Yang X. Roles and mechanisms of tumour-infiltrating B cells in human cancer: a new force in immunotherapy. Biomark Res 2023; 11:28. [PMID: 36890557 PMCID: PMC9997025 DOI: 10.1186/s40364-023-00460-1] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 01/28/2023] [Indexed: 03/10/2023] Open
Abstract
Immune checkpoint inhibitors (ICIs) targeting PD-1 or PD-L1 have emerged as a revolutionary treatment strategy for human cancer patients. However, as the response rate to ICI therapy varies widely among different types of tumours, we are beginning to gain insight into the mechanisms as well as biomarkers of therapeutic response and resistance. Numerous studies have highlighted the dominant role of cytotoxic T cells in determining the treatment response to ICIs. Empowered by recent technical advances, such as single-cell sequencing, tumour-infiltrating B cells have been identified as a key regulator in several solid tumours by affecting tumour progression and the response to ICIs. In the current review, we summarized recent advances regarding the role and underlying mechanisms of B cells in human cancer and therapy. Some studies have shown that B-cell abundance in cancer is positively associated with favourable clinical outcomes, while others have indicated that they are tumour-promoting, implying that the biological function of B cells is a complex landscape. The molecular mechanisms involved multiple aspects of the functions of B cells, including the activation of CD8+ T cells, the secretion of antibodies and cytokines, and the facilitation of the antigen presentation process. In addition, other crucial mechanisms, such as the functions of regulatory B cells (Bregs) and plasma cells, are discussed. Here, by summarizing the advances and dilemmas of recent studies, we depicted the current landscape of B cells in cancers and paved the way for future research in this field.
Collapse
Affiliation(s)
- Enkui Zhang
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.,Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Chengsheng Ding
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.,Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Shuchun Li
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.,Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Xueliang Zhou
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.,Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Batuer Aikemu
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.,Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Xiaodong Fan
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.,Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Jing Sun
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China. .,Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Minhua Zheng
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China. .,Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China. .,Department of General Surgery & Carson International Cancer Research Center, Shenzhen University General Hospital and Shenzhen University Clinical Medical Academy, Shenzhen, 518055, China.
| | - Xiao Yang
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China. .,Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China. .,Department of General Surgery & Carson International Cancer Research Center, Shenzhen University General Hospital and Shenzhen University Clinical Medical Academy, Shenzhen, 518055, China.
| |
Collapse
|
130
|
Sun K, Zhang Z, Wang D, Huang Y, Zhang J, Lian C. B cell-related tertiary lymphoid structure may exert inhibitory effects on lung adenocarcinoma and SARS-COV-2. Heliyon 2023; 9:e14334. [PMID: 36942234 PMCID: PMC10008815 DOI: 10.1016/j.heliyon.2023.e14334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 02/26/2023] [Accepted: 02/28/2023] [Indexed: 03/14/2023] Open
Abstract
Background The prognosis of lung adenocarcinoma (LUAD) is poor. Infection with coronavirus disease 2019 (COVID-19) may further worsen the outcome of LUAD. This study utilized the immune model and the COVID-19 receptor signal to identify the potential immune structure affecting the prognosis of COVID-19 and LUAD. Methods A prognostic model was established and verified. The correlation between immune cells and risk score was examined through a variety of immune calculation methods. Gene set variation analysis (GSVA) was used to explore the correlation between the immune signaling pathway, risk model, COVID-19 binding receptor (CO19ORS) signal, and different clinicopathological factors. Results The analysis showed that the prognosis of patients was better in the low-risk group versus the high-risk group. The tertiary lymphoid structure dominated by T and B cells (TLS1) can improve the prognosis of patients in the low-risk group. Interestingly, the CO19ORS was enriched only in females and aged >65 years. The age group >65 years is closely related to the tertiary lymphatic structure of the newborn (TLS2), while the female sex is closely related to the TLS2 and TLS1 signature. The two groups exhibited a high level of inflammation-related signal distribution. In the near future, I will collect LUAD and COVID-19 related organizations to verify the changes of 8 risk protein. Conclusion TLS1 structure may improve the prognosis of patients with LUAD and SARS-COV-2 (Severe acute respiratory syndrome coronavirus 2). This unexpected discovery provides new insight into the comprehensive treatment of patients with LUAD and SARS-COV-2.
Collapse
Affiliation(s)
- Kang Sun
- Research Center of Clinical Laboratory Science, Bengbu Medical College, Bengbu, 233030, China
| | - Zhiqiang Zhang
- Research Center of Clinical Laboratory Science, Bengbu Medical College, Bengbu, 233030, China
| | - Dongqin Wang
- Research Center of Clinical Laboratory Science, Bengbu Medical College, Bengbu, 233030, China
| | - Yinlong Huang
- Department of Genetics, School of Life Sciences, Bengbu Medical College, Bengbu, 233030, China
| | - Jing Zhang
- Department of Genetics, School of Life Sciences, Bengbu Medical College, Bengbu, 233030, China
| | - Chaoqun Lian
- Research Center of Clinical Laboratory Science, Bengbu Medical College, Bengbu, 233030, China
- Department of Biochemistry and Molecular Biology, School of Laboratory Medicine, Bengbu Medical College, Bengbu, 233030, China
| |
Collapse
|
131
|
Body Composition and Metabolic Dysfunction Really Matter for the Achievement of Better Outcomes in High-Grade Serous Ovarian Cancer. Cancers (Basel) 2023; 15:cancers15041156. [PMID: 36831500 PMCID: PMC9953877 DOI: 10.3390/cancers15041156] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 02/02/2023] [Accepted: 02/08/2023] [Indexed: 02/16/2023] Open
Abstract
Although obesity-associated metabolic disorders have a negative impact on various cancers, such evidence remains controversial for ovarian cancer. Here, we aimed to evaluate the impact of body composition (BC) and metabolism disorders on outcomes in high-grade serous ovarian cancer (HGSOC). METHODS We analyzed clinical/genomic data from two cohorts (PUC n = 123/TCGA-OV n = 415). BC was estimated using the measurement of adiposity/muscle mass by a CT scan. A list of 425 genes linked to obesity/lipid metabolism was used to cluster patients using non-negative matrix factorization. Differential expression, gene set enrichment analyses, and Ecotyper were performed. Survival curves and Cox-regression models were also built-up. RESULTS We identified four BC types and two clusters that, unlike BMI, effectively correlate with survival. High adiposity and sarcopenia were associated with worse outcomes. We also found that recovery of a normal BC and drug interventions to correct metabolism disorders had a positive impact on outcomes. Additionally, we showed that immune-cell-depleted microenvironments predominate in HGSOC, which was more evident among the BC types and the obesity/lipid metabolism cluster with worse prognosis. CONCLUSIONS We have demonstrated the relevance of BC and metabolism disorders as determinants of outcomes in HGSOC. We have shone a spotlight on the relevance of incorporating corrective measures addressing these disorders to obtain better results.
Collapse
|
132
|
Tertiary Lymphoid Structures Are Associated with a Favorable Prognosis in High-Grade Serous Ovarian Cancer Patients. Reprod Sci 2023:10.1007/s43032-023-01188-x. [PMID: 36759495 DOI: 10.1007/s43032-023-01188-x] [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: 10/28/2022] [Accepted: 01/30/2023] [Indexed: 02/11/2023]
Abstract
There was accumulating evidence indicating that tertiary lymphoid structures (TLSs) were strongly associated with improved survival and clinical outcome in several solid tumors. In this study, we intended to assess the presence of TLSs and their potential clinical significance in high-grade serous ovarian cancer (HGSOC). TCGA (The Cancer Genome Atlas) cohort included RNA-seq data of 376 HGSOC patients, of which 74 patients included available hematoxylin-eosin (H&E) sections; GEO (Gene Expression Omnibus) cohort, GSE140082, included microarray data of 212 HGSOC patients. TLSs were counted by pathological sections, and the relative abundance of TLSs was assessed by the unsupervised consensus clustering of 12-chemokine transcriptome signatures. The potential associations between TLSs and clinical prognosis, tumor microenvironment (TME), and immunotherapy response of HGSOC were further performed based on transcriptome data. In the H&E sections of HGSOC, TLSs were predominantly located in the stroma and invasive margin of the tumor. Pathological counting results suggested that the expression of 12 chemokines was significantly higher in samples abundant with TLSs than that in the lack of TLSs. Consensus clustering of both TCGA and GEO cohorts divided HGSOC patients into two clusters with different TLSs abundance: low- and high-TLSs. Based on transcriptome analysis, the high-TLS cluster was characterized by better clinical prognosis, a higher degree of immune infiltration, more biological pathways, higher tumor mutational burden score, and higher expression of immune checkpoints. In conclusion, TLSs strongly correlated with the immune-responsive microenvironment and remained a favorable prognostic factor independent of other clinical characteristics in HGSOC. The presence of TLSs was also associated with a potentially favorable response to immune checkpoint blockade (ICB) therapy in HGSOC.
Collapse
|
133
|
Kendal JK, Shehata MS, Lofftus SY, Crompton JG. Cancer-Associated B Cells in Sarcoma. Cancers (Basel) 2023; 15:cancers15030622. [PMID: 36765578 PMCID: PMC9913500 DOI: 10.3390/cancers15030622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 01/16/2023] [Accepted: 01/16/2023] [Indexed: 01/20/2023] Open
Abstract
Despite being one of the first types of cancers studied that hinted at a major role of the immune system in pro- and anti-tumor biology, little is known about the immune microenvironment in sarcoma. Few types of sarcoma have shown major responses to immunotherapy, and its rarity and heterogeneity makes it challenging to study. With limited systemic treatment options, further understanding of the underlying mechanisms in sarcoma immunity may prove crucial in advancing sarcoma care. While great strides have been made in the field of immunotherapy over the last few decades, most of these efforts have focused on harnessing the T cell response, with little attention on the role B cells may play in the tumor microenvironment. A growing body of evidence suggests that B cells have both pro- and anti-tumoral effects in a large variety of cancers, and in the age of bioinformatics and multi-omic analysis, the complexity of the humoral response is just being appreciated. This review explores what is currently known about the role of B cells in sarcoma, including understanding the various B cell populations associated with sarcoma, the organization of intra-tumoral B cells in tertiary lymphoid structures, recent trials in immunotherapy in sarcoma, intra-tumoral immunoglobulin, the pro-tumor effects of B cells, and exciting future areas for research.
Collapse
Affiliation(s)
- Joseph K. Kendal
- Department of Orthopaedic Surgery, University of California, Los Angeles, CA 90404, USA
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA 90024, USA
| | - Michael S. Shehata
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA 90024, USA
| | - Serena Y. Lofftus
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA 90024, USA
- Division of Surgical Oncology, Department of Surgery, University of California, Los Angeles, CA 90095, USA
| | - Joseph G. Crompton
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA 90024, USA
- Division of Surgical Oncology, Department of Surgery, University of California, Los Angeles, CA 90095, USA
- Correspondence: ; Tel.: +1-310-825-2644
| |
Collapse
|
134
|
Zhang Q, Wu S. Tertiary lymphoid structures are critical for cancer prognosis and therapeutic response. Front Immunol 2023; 13:1063711. [PMID: 36713409 PMCID: PMC9875059 DOI: 10.3389/fimmu.2022.1063711] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Accepted: 12/22/2022] [Indexed: 01/12/2023] Open
Abstract
Tertiary lymphoid structures (TLSs) are ectopic lymphocyte aggregates that form at sites of chronic inflammation, including cancers, in non-lymphoid tissues. Although the formation of TLSs is similar to that of secondary lymphoid organs, the pathogenic factors leading to TLS formation in cancerous tissues and the mechanisms underlying the role of these structures in the intra-tumoral adaptive antitumor immune response are not fully understood. The presence of TLSs may impact patient prognosis and treatment outcomes. This review examines the current understanding of TLSs in cancers, including their composition and formation as well as their potential to predict prognosis and therapeutic efficacy. We also summarize strategies to induce TLS formation for cancer treatment.
Collapse
Affiliation(s)
| | - Suhui Wu
- Department of Obstetrics and Gynecology, Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| |
Collapse
|
135
|
Hou Y, Qiao S, Li M, Han X, Wei X, Pang Y, Mao H. The gene signature of tertiary lymphoid structures within ovarian cancer predicts the prognosis and immunotherapy benefit. Front Genet 2023; 13:1090640. [PMID: 36704336 PMCID: PMC9871364 DOI: 10.3389/fgene.2022.1090640] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Accepted: 12/27/2022] [Indexed: 01/12/2023] Open
Abstract
Ovarian cancer (OC) has the lowest survival rate among gynecologic malignancies. Ectopic lymphocyte aggregates, namely tertiary lymphoid structures (TLSs), have been reported as positive biomarkers for tumor prognosis. However, the related gene signature of tertiary lymphoid structure in ovarian cancer was less understood. Therefore, this study first exhibited the organizational patterns of tertiary lymphoid structure by H&E staining and immunohistochemistry (IHC), and confirmed the improved survival values of tertiary lymphoid structure and quantified tumor-infiltrating lymphocytes (CD20+ B cells and CD8+ T cells) in ovarian cancer patients. Secondly, we collected the genes involved in tertiary lymphoid structure from databases. By the univariate regression analysis, the tertiary lymphoid structure gene signature (CETP, CCR7, SELL, LAMP3, CCL19, CXCL9, CXCL10, CXCL11, and CXCL13) with prognostic value, characteristically of ovarian cancer, was constructed in the TCGA dataset and validated in the GSE140082 dataset. Thirdly, by performing CIBERSORT and Tumor Immune Dysfunction and Exclusion (TIDE) analysis, we found that the high expression of this gene signature was positively correlated with developed immune infiltration and reduced immune escape. The improved IPS score and application in the IMvigor210 dataset received PD-L1 proved the predictive value of immunotherapy for this gene signature. Furthermore, this signature showed a better correlation between tumor mutation burden and classical checkpoint genes. In conclusion, Tertiary lymphoid structure plays important role in tumor immunity and the gene signature can be evaluated as a biomarker for predicting prognosis and guiding immunotherapy in ovarian cancer.
Collapse
Affiliation(s)
- Yue Hou
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, Shandong, China,Division of Gynecology Oncology, Qilu Hospital of Shandong University, Jinan, Shandong, China,Key Laboratory of Gynecology Oncology of Shandong Province, Qilu Hospital of Shandong University, Jinan, Shandong, China,Shandong Engineering Laboratory for Urogynecology, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Sijing Qiao
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, Shandong, China,Division of Gynecology Oncology, Qilu Hospital of Shandong University, Jinan, Shandong, China,Key Laboratory of Gynecology Oncology of Shandong Province, Qilu Hospital of Shandong University, Jinan, Shandong, China,Shandong Engineering Laboratory for Urogynecology, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Miao Li
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, Shandong, China,Division of Gynecology Oncology, Qilu Hospital of Shandong University, Jinan, Shandong, China,Key Laboratory of Gynecology Oncology of Shandong Province, Qilu Hospital of Shandong University, Jinan, Shandong, China,Shandong Engineering Laboratory for Urogynecology, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Xue Han
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, Shandong, China,Division of Gynecology Oncology, Qilu Hospital of Shandong University, Jinan, Shandong, China,Key Laboratory of Gynecology Oncology of Shandong Province, Qilu Hospital of Shandong University, Jinan, Shandong, China,Shandong Engineering Laboratory for Urogynecology, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Xuan Wei
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, Shandong, China,Division of Gynecology Oncology, Qilu Hospital of Shandong University, Jinan, Shandong, China,Key Laboratory of Gynecology Oncology of Shandong Province, Qilu Hospital of Shandong University, Jinan, Shandong, China,Shandong Engineering Laboratory for Urogynecology, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Yingxin Pang
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, Shandong, China,Division of Gynecology Oncology, Qilu Hospital of Shandong University, Jinan, Shandong, China,Key Laboratory of Gynecology Oncology of Shandong Province, Qilu Hospital of Shandong University, Jinan, Shandong, China,Shandong Engineering Laboratory for Urogynecology, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Hongluan Mao
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, Shandong, China,Division of Gynecology Oncology, Qilu Hospital of Shandong University, Jinan, Shandong, China,Key Laboratory of Gynecology Oncology of Shandong Province, Qilu Hospital of Shandong University, Jinan, Shandong, China,Shandong Engineering Laboratory for Urogynecology, Qilu Hospital of Shandong University, Jinan, Shandong, China,*Correspondence: Hongluan Mao,
| |
Collapse
|
136
|
Bernitsa S, Dayan R, Stephanou A, Tzvetanova ID, Patrikios IS. Natural biomolecules and derivatives as anticancer immunomodulatory agents. Front Immunol 2023; 13:1070367. [PMID: 36700235 PMCID: PMC9868674 DOI: 10.3389/fimmu.2022.1070367] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 12/05/2022] [Indexed: 01/11/2023] Open
Abstract
Despite advancements in chemotherapy, the issue of resistance and non-responsiveness to many chemotherapeutic drugs that are currently in clinical use still remains. Recently, cancer immunotherapy has gathered attention as a novel treatment against select cancers. Immunomodulation is also emerging as an effective strategy to improve efficacy. Natural phytochemicals, with known anticancer properties, been reported to mediate their effects by modulating both traditional cancer pathways and immunity. The mechanism of phytochemical mediated-immunomodulatory activity may be attributed to the remodeling of the tumor immunosuppressive microenvironment and the sensitization of the immune system. This allows for improved recognition and targeting of cancer cells by the immune system and synergy with chemotherapeutics. In this review, we will discuss several well-known plant-derived biomolecules and examine their potential as immunomodulators, and therefore, as novel immunotherapies for cancer treatment.
Collapse
Affiliation(s)
| | - Rotem Dayan
- School of Medicine, European University Cyprus, Nicosia, Cyprus
| | | | | | | |
Collapse
|
137
|
Xu X, Wu Y, Jia G, Zhu Q, Li D, Xie K. A signature based on glycosyltransferase genes provides a promising tool for the prediction of prognosis and immunotherapy responsiveness in ovarian cancer. J Ovarian Res 2023; 16:5. [PMID: 36611197 PMCID: PMC9826597 DOI: 10.1186/s13048-022-01088-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 12/22/2022] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Ovarian cancer (OC) is the most fatal gynaecological malignancy and has a poor prognosis. Glycosylation, the biosynthetic process that depends on specific glycosyltransferases (GTs), has recently attracted increasing importance due to the vital role it plays in cancer. In this study, we aimed to determine whether OC patients could be stratified by glycosyltransferase gene profiles to better predict the prognosis and efficiency of immune checkpoint blockade therapies (ICBs). METHODS We retrieved transcriptome data across 420 OC and 88 normal tissue samples using The Cancer Genome Atlas (TCGA) and Genotype-Tissue Expression (GTEx) databases, respectively. An external validation dataset containing 185 OC samples was downloaded from the Gene Expression Omnibus (GEO) database. Knockdown and pathway prediction of B4GALT5 were conducted to investigate the function and mechanism of B4GALT5 in OC proliferation, migration and invasion. RESULTS A total of 50 differentially expressed GT genes were identified between OC and normal ovarian tissues. Two clusters were stratified by operating consensus clustering, but no significant prognostic value was observed. By applying the least absolute shrinkage and selection operator (LASSO) Cox regression method, a 6-gene signature was built that classified OC patients in the TCGA cohort into a low- or high-risk group. Patients with high scores had a worse prognosis than those with low scores. This risk signature was further validated in an external GEO dataset. Furthermore, the risk score was an independent risk predictor, and a nomogram was created to improve the accuracy of prognostic classification. Notably, the low-risk OC patients exhibited a higher degree of antitumor immune cell infiltration and a superior response to ICBs. B4GALT5, one of six hub genes, was identified as a regulator of proliferation, migration and invasion in OC. CONCLUSION Taken together, we established a reliable GT-gene-based signature to predict prognosis, immune status and identify OC patients who would benefit from ICBs. GT genes might be a promising biomarker for OC progression and a potential therapeutic target for OC.
Collapse
Affiliation(s)
- Xuyao Xu
- grid.459791.70000 0004 1757 7869Department of Gynecology, Nanjing Maternity and Child Health Care Hospital, Women’s Hospital of Nanjing Medical University, Nanjing, 210004 China
| | - Yue Wu
- grid.459791.70000 0004 1757 7869Department of Gynecology, Nanjing Maternity and Child Health Care Hospital, Women’s Hospital of Nanjing Medical University, Nanjing, 210004 China
| | - Genmei Jia
- grid.459791.70000 0004 1757 7869Department of Women Health Care, Nanjing Maternity and Child Health Care Hospital, Women’s Hospital of Nanjing Medical University, Nanjing, 210004 China
| | - Qiaoying Zhu
- grid.459791.70000 0004 1757 7869Department of Gynecology, Nanjing Maternity and Child Health Care Hospital, Women’s Hospital of Nanjing Medical University, Nanjing, 210004 China
| | - Dake Li
- grid.459791.70000 0004 1757 7869Department of Gynecology, Nanjing Maternity and Child Health Care Hospital, Women’s Hospital of Nanjing Medical University, Nanjing, 210004 China
| | - Kaipeng Xie
- grid.459791.70000 0004 1757 7869Department of Women Health Care, Nanjing Maternity and Child Health Care Hospital, Women’s Hospital of Nanjing Medical University, Nanjing, 210004 China ,grid.459791.70000 0004 1757 7869Department of Public Health, Nanjing Maternity and Child Health Care Hospital, Women’s Hospital of Nanjing Medical University, Nanjing, 210004 China
| |
Collapse
|
138
|
Yang J, Wang C, Cheng S, Zhang Y, Jin Y, Zhang N, Wang Y. Construction and validation of a novel ferroptosis-related signature for evaluating prognosis and immune microenvironment in ovarian cancer. Front Genet 2023; 13:1094474. [PMID: 36685851 PMCID: PMC9849594 DOI: 10.3389/fgene.2022.1094474] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 12/20/2022] [Indexed: 01/07/2023] Open
Abstract
Ovarian cancer (OV) is the most lethal form of gynecological malignancy worldwide, with limited therapeutic options and high recurrence rates. However, research focusing on prognostic patterns of ferroptosis-related genes (FRGs) in ovarian cancer is still lacking. From the 6,406 differentially expressed genes (DEGs) between TCGA-OV (n = 376) and GTEx cohort (n = 180), we identified 63 potential ferroptosis-related genes. Through the LASSO-penalized Cox analysis, 3 prognostic genes, SLC7A11, ZFP36, and TTBK2, were finally distinguished. The time-dependent ROC curves and K-M survival analysis performed powerful prognostic ability of the 3-gene signature. Stepwise, we constructed and validated the nomogram based on the 3-gene signature and clinical features, with promising prognostic value in both TCGA (p-value < .0001) and ICGC cohort (p-value = .0064). Gene Set Enrichment Analysis elucidated several potential pathways between the groups stratified by 3-gene signature, while the m6A gene analysis implied higher m6A level in the high-risk group. We applied the CIBERSORT algorithm to distinct tumor immune microenvironment between two groups, with less activated dendritic cells (DCs) and plasma cells, more M0 macrophages infiltration, and higher expression of key immune checkpoint molecules (CD274, CTLA4, HAVCR2, and PDCD1LG2) in the high-risk group. In addition, the low-risk group exhibited more favorable immunotherapy and chemotherapy responses. Collectively, our findings provided new prospects in the role of ferroptosis-related genes, as a promising prediction tool for prognosis and immune responses, in order to assist personalized treatment decision-making among ovarian cancer patients.
Collapse
Affiliation(s)
- Jiani Yang
- Department of Obstetrics and Gynecology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China,Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Chao Wang
- Department of Obstetrics and Gynecology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China,Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Shanshan Cheng
- Department of Obstetrics and Gynecology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Yue Zhang
- Department of Obstetrics and Gynecology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China,Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yue Jin
- Department of Obstetrics and Gynecology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Nan Zhang
- Department of Obstetrics and Gynecology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Yu Wang
- Department of Obstetrics and Gynecology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China,Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China,*Correspondence: Yu Wang,
| |
Collapse
|
139
|
Kadota H, Gono T, Kunugi S, Ota Y, Takeno M, Seike M, Shimizu A, Kuwana M. Tertiary lymphoid structures in the primary tumor site of patients with cancer-associated myositis: A case-control study. Front Med (Lausanne) 2023; 9:1066858. [PMID: 36687449 PMCID: PMC9845936 DOI: 10.3389/fmed.2022.1066858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 12/12/2022] [Indexed: 01/05/2023] Open
Abstract
Objective To investigate histologic features of immunological components in the primary tumor site of patients with cancer-associated myositis (CAM) by focusing on tumor-infiltrating lymphocytes (TILs) and tertiary lymphoid structures (TLSs), which play major roles in antitumor immunity. Methods Cancer-associated myositis patients were selected from the single-center idiopathic inflammatory myopathy cohort based on the availability of primary tumor specimens obtained before the introduction of immunomodulatory agents. Control cancer subjects without CAM were selected from the cancer tissue repository at a ratio of 1:2 matched for demographics and cancer characteristics of CAM cases. A series of immunohistochemical analyses was conducted using sequential tumor sections. TLS was defined as an ectopic lymphoid-like structure composed of DC-LAMP+ mature dendritic cells, CD23+ follicular dendritic cells (FDCs) and PNAd+ high endothelial venules. TLS distribution was classified into the tumor center, invasive margin, and peritumoral area. Results Six CAM patients and 12 matched non-CAM controls were eligible for the study. There was no apparent difference in the density or distribution of TILs between the groups. TLSs were found in 3 CAM patients (50%) and 4 non-CAM controls (33%). TLSs were exclusively located at the tumor center or invasive margin in CAM cases but were mainly found in the peritumoral area in non-CAM controls. FDCs and class-switched B cells colocalized with follicular helper T cells were abundantly found in the germinal center-like area of TLSs from CAM patients compared with those from non-CAM controls. Conclusion The adaptive immune response within TLSs in the primary tumor site might contribute to the pathogenic process of CAM.
Collapse
Affiliation(s)
- Hiroko Kadota
- Department of Allergy and Rheumatology, Graduate School of Medicine, Nippon Medical School, Tokyo, Japan
| | - Takahisa Gono
- Department of Allergy and Rheumatology, Graduate School of Medicine, Nippon Medical School, Tokyo, Japan,Scleroderma/Myositis Center of Excellence, Nippon Medical School Hospital, Tokyo, Japan
| | - Shinobu Kunugi
- Department of Analytic Human Pathology, Graduate School of Medicine, Nippon Medical School, Tokyo, Japan
| | - Yuko Ota
- Department of Allergy and Rheumatology, Graduate School of Medicine, Nippon Medical School, Tokyo, Japan,Scleroderma/Myositis Center of Excellence, Nippon Medical School Hospital, Tokyo, Japan
| | - Mitsuhiro Takeno
- Department of Allergy and Rheumatology, Graduate School of Medicine, Nippon Medical School, Tokyo, Japan,Department of Allergy and Rheumatology, Nippon Medical School Musashi Kosugi Hospital, Kawasaki, Japan
| | - Masahiro Seike
- Department of Pulmonary Medicine and Oncology, Graduate School of Medicine, Nippon Medical School, Tokyo, Japan
| | - Akira Shimizu
- Department of Analytic Human Pathology, Graduate School of Medicine, Nippon Medical School, Tokyo, Japan
| | - Masataka Kuwana
- Department of Allergy and Rheumatology, Graduate School of Medicine, Nippon Medical School, Tokyo, Japan,Scleroderma/Myositis Center of Excellence, Nippon Medical School Hospital, Tokyo, Japan,*Correspondence: Masataka Kuwana,
| |
Collapse
|
140
|
Abstract
Immuno-oncology has traditionally focused on the cellular arm of the adaptive immune response, while attributing tumor-promoting activity to humoral responses in tumor-bearing hosts. This view stems from mouse models that do not necessarily recapitulate the antibody response process consistently observed in most human cancers. In recent years, the field has reconsidered the coordinated action of T and B cell responses in the context of anti-tumor immunity, as in any other immune response. Thus, recent studies in human cancer identify B cell responses with better outcome, typically in association with superior T cell responses. An area of particular interest is tertiary lymphoid structures, where germinal centers produce isotype switched antibodies and B cells and T lymphocytes interact with other immune cell types. The presence of these lymphoid structures is associated with better immunotherapeutic responses and remain poorly understood. Here, we discuss recent discoveries on how coordination between humoral and cellular responses is required for effective immune pressure against malignant progression, providing a perspective on the role of tertiary lymphoid structures and interventions to elicit their formation in unresectable tumors.
Collapse
Affiliation(s)
- Jose R Conejo-Garcia
- Department of Immunology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA.
| | - Subir Biswas
- Department of Immunology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Ricardo Chaurio
- Department of Immunology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Paulo C Rodriguez
- Department of Immunology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| |
Collapse
|
141
|
Multiparameter single-cell proteomic technologies give new insights into the biology of ovarian tumors. Semin Immunopathol 2023; 45:43-59. [PMID: 36635516 PMCID: PMC9974728 DOI: 10.1007/s00281-022-00979-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 12/11/2022] [Indexed: 01/13/2023]
Abstract
High-grade serous ovarian cancer (HGSOC) is the most lethal gynecological malignancy. Its diagnosis at advanced stage compounded with its excessive genomic and cellular heterogeneity make curative treatment challenging. Two critical therapeutic challenges to overcome are carboplatin resistance and lack of response to immunotherapy. Carboplatin resistance results from diverse cell autonomous mechanisms which operate in different combinations within and across tumors. The lack of response to immunotherapy is highly likely to be related to an immunosuppressive HGSOC tumor microenvironment which overrides any clinical benefit. Results from a number of studies, mainly using transcriptomics, indicate that the immune tumor microenvironment (iTME) plays a role in carboplatin response. However, in patients receiving treatment, the exact mechanistic details are unclear. During the past decade, multiplex single-cell proteomic technologies have come to the forefront of biomedical research. Mass cytometry or cytometry by time-of-flight, measures up to 60 parameters in single cells that are in suspension. Multiplex cellular imaging technologies allow simultaneous measurement of up to 60 proteins in single cells with spatial resolution and interrogation of cell-cell interactions. This review suggests that functional interplay between cell autonomous responses to carboplatin and the HGSOC immune tumor microenvironment could be clarified through the application of multiplex single-cell proteomic technologies. We conclude that for better clinical care, multiplex single-cell proteomic technologies could be an integral component of multimodal biomarker development that also includes genomics and radiomics. Collection of matched samples from patients before and on treatment will be critical to the success of these efforts.
Collapse
|
142
|
Lima CAD, Jammal MP, Etchebehere RM, Murta EFC, Nomelini RS. Immunostaining of stromal CD56 cells in ovarian malignancies. REVISTA DA ASSOCIACAO MEDICA BRASILEIRA (1992) 2023; 69:e20220992. [PMID: 37194901 DOI: 10.1590/1806-9282.20220992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 02/23/2023] [Indexed: 05/18/2023]
Abstract
OBJECTIVES The aim of this study was to evaluate CD56 immunostaining in the stroma of benign and malignant ovarian epithelial neoplasms and associate the CD56 immunostaining with prognostic factors and survival in ovarian cancer. METHODS Patients with ovarian epithelial neoplasia (n=77) were studied with a prospective cohort. The CD56 immunostaining was evaluated in the peritumoral stroma. Two groups were evaluated: benign ovarian neoplasms (n=40) and malignant ovarian neoplasms (n=37). Data were recorded for histological type and grade, International Federation of Gynecology and Obstetrics staging, molecular subtype, and lymph node metastases. Fisher's exact test and Kaplan-Meier survival curves were used, with a significance level of ≤0.05. RESULTS We found greater CD56 stromal immunostaining in malignant neoplasms when compared to the group of benign neoplasms (p=0.00001). There was no significant difference in relation to the prognostic factors and survival. CONCLUSION Malignant ovarian neoplasms showed higher stromal CD56 immunostaining. As the prognostic value of natural killer in ovarian cancer is controversial, knowing the specific function of each cell present both in the tumor tissue and systemically may help guide successful immunotherapies in the near future.
Collapse
Affiliation(s)
- Cid Almeida de Lima
- Universidade Federal do Triângulo Mineiro, Department of Gynecology and Obstetrics - Uberaba (MG), Brazil
| | - Millena Prata Jammal
- Universidade Federal do Triângulo Mineiro, Department of Gynecology and Obstetrics - Uberaba (MG), Brazil
| | | | | | - Rosekeila Simões Nomelini
- Universidade Federal do Triângulo Mineiro, Department of Gynecology and Obstetrics - Uberaba (MG), Brazil
| |
Collapse
|
143
|
Guo Z, Zhang R, Yang AG, Zheng G. Diversity of immune checkpoints in cancer immunotherapy. Front Immunol 2023; 14:1121285. [PMID: 36960057 PMCID: PMC10027905 DOI: 10.3389/fimmu.2023.1121285] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Accepted: 02/23/2023] [Indexed: 03/09/2023] Open
Abstract
Finding effective treatments for cancer remains a challenge. Recent studies have found that the mechanisms of tumor evasion are becoming increasingly diverse, including abnormal expression of immune checkpoint molecules on different immune cells, in particular T cells, natural killer cells, macrophages and others. In this review, we discuss the checkpoint molecules with enhanced expression on these lymphocytes and their consequences on immune effector functions. Dissecting the diverse roles of immune checkpoints in different immune cells is crucial for a full understanding of immunotherapy using checkpoint inhibitors.
Collapse
Affiliation(s)
- Zhangyan Guo
- State Key Laboratory of Cancer Biology, Department of Immunology, Fourth Military Medical University, Xi’an, China
| | - Rui Zhang
- State Key Laboratory of Cancer Biology, Department of Immunology, Fourth Military Medical University, Xi’an, China
- State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi’an, China
| | - An-Gang Yang
- State Key Laboratory of Cancer Biology, Department of Immunology, Fourth Military Medical University, Xi’an, China
- *Correspondence: Guoxu Zheng, ; An-Gang Yang,
| | - Guoxu Zheng
- State Key Laboratory of Cancer Biology, Department of Immunology, Fourth Military Medical University, Xi’an, China
- *Correspondence: Guoxu Zheng, ; An-Gang Yang,
| |
Collapse
|
144
|
Giatromanolaki A, Chatzipantelis P, Contrafouris CA, Koukourakis MI. Tertiary Lymphoid Structures, Immune Response, and Prognostic Relevance in Non-Small Cell Lung Cancer. Cancer Invest 2023; 41:48-57. [PMID: 36239379 DOI: 10.1080/07357907.2022.2136684] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
We assessed the presence of 'tertiary lymphoid structures' (TLS) in a series of surgically treated non-small cell lung carcinomas (NSCLC). The TLS-density in the tumor periphery (pTLS) ranged from 0 to 1.8 (median 0.45), while in inner tumor areas (iTLS) ranged from 0 to 1.0 (median 0); (p < 0.0001). High pTLS-density was linked with early stage of the disease. Glycolysis-related enzyme expression (MCT1, Hexokinase 2) was linked with high pTLS-density (p < 0.05). High pTLS and iTLS densities were linked with better postoperative prognosis (p = 0.02 and p = 0.01, respectively). Assessment of TLS is a useful prognostic marker in NSCLC.
Collapse
Affiliation(s)
- Alexandra Giatromanolaki
- Department of Pathology, University Hospital of Alexandroupolis, Democritus University of Thrace, Greece
| | - Paschalis Chatzipantelis
- Department of Pathology, University Hospital of Alexandroupolis, Democritus University of Thrace, Greece
| | | | - Michael I Koukourakis
- Department of Radiotherapy/Oncology, University Hospital of Alexandroupolis, Democritus University of Thrace, Alexandroupolis, Greece
| |
Collapse
|
145
|
Fridman WH, Sibéril S, Pupier G, Soussan S, Sautès-Fridman C. Activation of B cells in Tertiary Lymphoid Structures in cancer: Anti-tumor or anti-self? Semin Immunol 2023; 65:101703. [PMID: 36481358 DOI: 10.1016/j.smim.2022.101703] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 11/28/2022] [Indexed: 12/12/2022]
Abstract
Whereas T cells in the tumor microenvironment have been the main focus as cancer controlling cells and targets of immunotherapies, B cells have recently gained strong attention. Being associated to Tertiary Lymphoid Structures (TLS) located at the vicinity of tumor nests, the fate of B cell depends on TLS maturity. In immature TLS they may evolve as regulatory B cells producing immunosuppressive cytokines and promote tumor growth. In mature TLS with a germinal center, B cells are selected, amplified, undergo affinity maturation and isotypic switching, resulting in plasma cell generation and production of anti-tumor antibodies. In that case, they are associated with longer patient's survival and therapeutic response to immunotherapy. Identification of tumor specific, or tumor overexpressed, antigens recognized by "in situ" produced antibodies and their discrimination from self-antigens induced by ICI treatments is a major challenge to develop novel antibody-based immunotherapies.
Collapse
Affiliation(s)
- Wolf H Fridman
- Centre de recherche des Cordeliers, INSERM, Sorbonne Université, Université Paris Cité, F-75006 Paris, France; Equipe labellisée Ligue Contre le Cancer, 75006 Paris, France.
| | - Sophie Sibéril
- Centre de recherche des Cordeliers, INSERM, Sorbonne Université, Université Paris Cité, F-75006 Paris, France
| | - Guilhem Pupier
- Centre de recherche des Cordeliers, INSERM, Sorbonne Université, Université Paris Cité, F-75006 Paris, France; Equipe labellisée Ligue Contre le Cancer, 75006 Paris, France
| | - Sarah Soussan
- Centre de recherche des Cordeliers, INSERM, Sorbonne Université, Université Paris Cité, F-75006 Paris, France
| | - Catherine Sautès-Fridman
- Centre de recherche des Cordeliers, INSERM, Sorbonne Université, Université Paris Cité, F-75006 Paris, France; Equipe labellisée Ligue Contre le Cancer, 75006 Paris, France
| |
Collapse
|
146
|
Mark J, Fisher DT, Kim M, Emmons T, Khan ANMN, Alqassim E, Singel K, Mistarz A, Lugade A, Zhan H, Yu H, Segal B, Lele S, Frederick P, Kozbor D, Skitzki J, Odunsi K. Carboplatin enhances lymphocyte-endothelial interactions to promote CD8 + T cell trafficking into the ovarian tumor microenvironment. Gynecol Oncol 2023; 168:92-99. [PMID: 36410228 PMCID: PMC11236086 DOI: 10.1016/j.ygyno.2022.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2022] [Revised: 10/16/2022] [Accepted: 11/01/2022] [Indexed: 11/19/2022]
Abstract
OBJECTIVES Standard chemotherapy agents, including carboplatin, have known immunogenic properties. We sought to determine how carboplatin may influence lymphocyte trafficking to tumor sites. METHODS Murine models of ovarian cancer were utilized to examine lymphocyte trafficking with common clinically used agents including carboplatin, anti-PD-1 antibody, or anti-VEGFR-2 antibody. Adhesion interactions of lymphocytes with tumor vasculature were measured using intravital microscopy, lymphocyte homing with immunohistochemistry, and treatment groups followed for overall survival. RESULTS Carboplatin chemotherapy profoundly alters the tumor microenvironment to promote lymphocyte adhesive interactions with tumor vasculature and resultant improvement in lymphocyte trafficking. The measured results seen with carboplatin in the tumor microenvironment were superior to anti-PD-1 treatment or anti-VEGFR-2 which may have contributed to increased overall survival in carboplatin treated groups. CONCLUSIONS These novel findings suggest a role for chemotherapeutic agents to broadly influence anti-tumor immune responses beyond the induction of immunogenic tumor cell death.
Collapse
Affiliation(s)
- Jaron Mark
- Department of Gynecologic Oncology, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY 14263, United States
| | - Dan T Fisher
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY 14263, United States
| | - Minhyung Kim
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY 14263, United States; Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY 14263, United States
| | - Tiffany Emmons
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY 14263, United States
| | - A N M Nazmul Khan
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY 14263, United States
| | - Emad Alqassim
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY 14263, United States
| | - Kelly Singel
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY 14263, United States
| | - Anna Mistarz
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY 14263, United States
| | - Amit Lugade
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY 14263, United States
| | - Haiying Zhan
- Department of Pathology, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY 14263, United States
| | - Han Yu
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY 14263, United States
| | - Brahm Segal
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY 14263, United States
| | - Shashikant Lele
- Department of Gynecologic Oncology, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY 14263, United States
| | - Peter Frederick
- Department of Gynecologic Oncology, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY 14263, United States
| | - Danuta Kozbor
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY 14263, United States
| | - Joseph Skitzki
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY 14263, United States; Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY 14263, United States.
| | - Kunle Odunsi
- Department of Gynecologic Oncology, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY 14263, United States; Department of Immunology, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY 14263, United States; University of Chicago Comprehensive Cancer Center, 5841 S. Maryland Avenue, Chicago, IL 60637, United States.
| |
Collapse
|
147
|
Lacombe RV, Sibéril S, Dimitrov JD. Immature and mature antibodies as defenders against cancer. Cell Mol Immunol 2023; 20:3-5. [PMID: 36369369 PMCID: PMC9794785 DOI: 10.1038/s41423-022-00951-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Accepted: 11/01/2022] [Indexed: 11/13/2022] Open
Affiliation(s)
- Robin V Lacombe
- Centre de Recherche des Cordeliers, INSERM, CNRS, Sorbonne Université, Université Paris Cité, Paris, France
| | - Sophie Sibéril
- Centre de Recherche des Cordeliers, INSERM, CNRS, Sorbonne Université, Université Paris Cité, Paris, France
| | - Jordan D Dimitrov
- Centre de Recherche des Cordeliers, INSERM, CNRS, Sorbonne Université, Université Paris Cité, Paris, France.
| |
Collapse
|
148
|
D’Angelo A, Kilili H, Chapman R, Generali D, Tinhofer I, Luminari S, Donati B, Ciarrocchi A, Giannini R, Moretto R, Cremolini C, Pietrantonio F, Sobhani N, Bonazza D, Prins R, Song SG, Jeon YK, Pisignano G, Cinelli M, Bagby S, Urrutia AO. Immune-related pan-cancer gene expression signatures of patient survival revealed by NanoString-based analyses. PLoS One 2023; 18:e0280364. [PMID: 36649303 PMCID: PMC9844904 DOI: 10.1371/journal.pone.0280364] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Accepted: 12/28/2022] [Indexed: 01/18/2023] Open
Abstract
The immune system plays a central role in the onset and progression of cancer. A better understanding of transcriptional changes in immune cell-related genes associated with cancer progression, and their significance in disease prognosis, is therefore needed. NanoString-based targeted gene expression profiling has advantages for deployment in a clinical setting over RNA-seq technologies. We analysed NanoString PanCancer Immune Profiling panel gene expression data encompassing 770 genes, and overall survival data, from multiple previous studies covering 10 different cancer types, including solid and blood malignancies, across 515 patients. This analysis revealed an immune gene signature comprising 39 genes that were upregulated in those patients with shorter overall survival; of these 39 genes, three (MAGEC2, SSX1 and ULBP2) were common to both solid and blood malignancies. Most of the genes identified have previously been reported as relevant in one or more cancer types. Using Cibersort, we investigated immune cell levels within individual cancer types and across groups of cancers, as well as in shorter and longer overall survival groups. Patients with shorter survival had a higher proportion of M2 macrophages and γδ T cells. Patients with longer overall survival had a higher proportion of CD8+ T cells, CD4+ T memory cells, NK cells and, unexpectedly, T regulatory cells. Using a transcriptomics platform with certain advantages for deployment in a clinical setting, our multi-cancer meta-analysis of immune gene expression and overall survival data has identified a specific transcriptional profile associated with poor overall survival.
Collapse
Affiliation(s)
- Alberto D’Angelo
- Department of Life Sciences, University of Bath, Bath, United Kingdom
- Oncology Department, Royal United Hospital, Bath, United Kingdom
- * E-mail:
| | - Huseyin Kilili
- Milner Centre, Department of Life Sciences, University of Bath, Bath, United Kingdom
| | - Robert Chapman
- Department of Medicine, The Princess Alexandra Hospital, Harlow, United Kingdom
| | - Daniele Generali
- Multidisciplinary Unit of Breast Pathology and Translational Research, Cremona Hospital, Cremona, Italy
| | - Ingeborg Tinhofer
- Department of Radiooncology and Radiotherapy, Charite´ University Hospital, Berlin, Germany
| | - Stefano Luminari
- Hematology Unit, Azienda USL-IRCCS, Reggio Emilia, Italy
- Surgical, Medical and Dental Department of Morphological Sciences Related to Transplant, Oncology and Regenerative Medicine, University of Modena and Reggio Emilia, Reggio Emilia, Italy
| | - Benedetta Donati
- Translational Research Laboratory, Azienda USL-IRCCS, Reggio Emilia, Italy
| | - Alessia Ciarrocchi
- Translational Research Laboratory, Azienda USL-IRCCS, Reggio Emilia, Italy
| | - Riccardo Giannini
- Department of Surgery, Clinical, Molecular and Critical Care Pathology, University of Pisa, Pisa, Italy
| | - Roberto Moretto
- Unit of Medical Oncology 2, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy
| | - Chiara Cremolini
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | | | - Navid Sobhani
- Section of Epidemiology and Population Science, Department of Medicine, Baylor College of Medicine, Houston, Texas, United States of America
| | - Debora Bonazza
- Department of Medical, Surgical and Health Sciences, Cattinara Hospital, University of Trieste, Trieste, Italy
| | - Robert Prins
- Department of Neurosurgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California, United States of America
| | - Seung Geun Song
- Department of Pathology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Yoon Kyung Jeon
- Department of Pathology, Seoul National University College of Medicine, Seoul, Republic of Korea
- Cancer Research Institute, Seoul National University, Seoul, Republic of Korea
| | | | - Mattia Cinelli
- Department of Life Sciences, University of Bath, Bath, United Kingdom
| | - Stefan Bagby
- Department of Life Sciences, University of Bath, Bath, United Kingdom
| | - Araxi O. Urrutia
- Milner Centre, Department of Life Sciences, University of Bath, Bath, United Kingdom
- Instituto de Ecologia, UNAM, Ciudad de Mexico, Mexico
| |
Collapse
|
149
|
Dowling JP, Nikitin PA, Shen F, Shukla H, Finn JP, Patel N, Swider C, Bingaman-Steele JL, Nicolescu C, Sikorski EL, Greenawalt EJ, Morin MJ, Robinson MK, Lundgren K, Harman BC. IL-38 blockade induces anti-tumor immunity by abrogating tumor-mediated suppression of early immune activation. MAbs 2023; 15:2212673. [PMID: 37216961 DOI: 10.1080/19420862.2023.2212673] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 05/02/2023] [Accepted: 05/08/2023] [Indexed: 05/24/2023] Open
Abstract
Immune checkpoint inhibitors that overcome T cell suppressive mechanisms in tumors have revolutionized the treatment of cancer but are only efficacious in a small subset of patients. Targeting suppressive mechanisms acting on innate immune cells could significantly improve the incidence of clinical response by facilitating a multi-lineage response against the tumor involving both adaptive and innate immune systems. Here, we show that intra-tumoral interleukin (IL)-38 expression is a feature of a large frequency of head and neck, lung and cervical squamous cancers and correlates with reduced immune cell numbers. We generated IMM20324, an antibody that binds human and mouse IL-38 proteins and inhibits the binding of IL-38 to its putative receptors, interleukin 1 receptor accessory protein-like 1 (IL1RAPL) and IL-36R. In vivo, IMM20324 demonstrated a good safety profile, delayed tumor growth in a subset of mice in an EMT6 syngeneic model of breast cancer, and significantly inhibited tumor expansion in a B16.F10 melanoma model. Notably, IMM20324 treatment resulted in the prevention of tumor growth following re-implantation of tumor cells, indicating the induction of immunological memory. Furthermore, exposure of IMM20324 correlated with decreased tumor volume and increased levels of intra-tumoral chemokines. Together, our data suggest that IL-38 is expressed in a high frequency of cancer patients and allows tumor cells to suppress anti-tumor immunity. Blockade of IL-38 activity using IMM20324 can re-activate immunostimulatory mechanisms in the tumor microenvironment leading to immune infiltration, the generation of tumor-specific memory and abrogation of tumor growth.
Collapse
Affiliation(s)
| | | | - Fang Shen
- Research & Development, Immunome Inc, Exton, PA, USA
| | - Halley Shukla
- Research & Development, Immunome Inc, Exton, PA, USA
| | - James P Finn
- Research & Development, Immunome Inc, Exton, PA, USA
| | - Nirja Patel
- Research & Development, Immunome Inc, Exton, PA, USA
| | - Cezary Swider
- Research & Development, Immunome Inc, Exton, PA, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
150
|
Palomero J, Panisello C, Lozano-Rabella M, Tirtakasuma R, Díaz-Gómez J, Grases D, Pasamar H, Arregui L, Dorca Duch E, Guerra Fernández E, Vivancos A, de Andrea CE, Melero I, Ponce J, Vidal A, Piulats JM, Matias-Guiu X, Gros A. Biomarkers of tumor-reactive CD4 + and CD8 + TILs associate with improved prognosis in endometrial cancer. J Immunother Cancer 2022; 10:jitc-2022-005443. [PMID: 36581331 PMCID: PMC9806064 DOI: 10.1136/jitc-2022-005443] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/25/2022] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Despite the growing interest in immunotherapeutic interventions for endometrial cancer (EC), the prevalence, phenotype, specificity and prognostic value of tumor infiltrating lymphocytes (TILs) in this tumor type remains unclear. METHODS To better understand the role of TILs in EC, we analyzed the phenotypic traits of CD8+ and CD4+ EC-resident T cells from 47 primary tumors by high-dimensional flow cytometry. In addition, CD8+ and CD4+ TIL subpopulations were isolated based on the differential expression of programmed cell death protein-1 (PD-1) (negative, dim and high) and CD39 (positive or negative) by fluorescence activated cell sorting (FACS), expanded in vitro, and screened for autologous tumor recognition. We further investigated whether phenotypic markers preferentially expressed on CD8+ and CD4+ tumor-reactive TIL subsets were associated with the four distinct molecular subtypes of EC, tumor mutational burden and patient survival. RESULTS We found that CD8+TILs expressing high levels of PD-1 (PD-1hi) co-expressed CD39, TIM-3, HLA-DR and CXCL13, as compared with TILs lacking or displaying intermediate levels of PD-1 expression (PD-1- and PD-1dim, respectively). Autologous tumor reactivity of sorted and in vitro expanded CD8+ TILs demonstrated that the CD8+PD-1dimCD39+ and PD-1hiCD39+ T cell subsets both contained tumor-reactive TILs and that a higher level of PD-1 expression was associated with increased CD39 and a superior frequency of tumor reactivity. With respect to CD4+ T conventional (Tconv) TILs, co-expression of inhibitory and activation markers was more apparent on PD-1hi compared with PD-1- or PD-1dim T cells, and in fact, it was the CD4+PD-1hi subpopulation that accumulated the antitumor T cells irrespective of CD39 expression. Most importantly, detection of CD8+PD-1hiCD39+ and CD4+PD-1hi tumor-reactive T-cell subsets, but also markers specifically expressed by these subpopulations of TILs, that is, PD-1hi, CD39, CXCL13 and CD103 by CD8+ TILs and PD-1hi and CXCL13 by CD4+ Tconv TILs, correlated with prolonged survival of patients with EC. CONCLUSIONS Our results demonstrate that EC are frequently infiltrated by tumor-reactive TILs, and that expression of PD-1hi and CD39 or PD-1hi can be used to select and expand CD8+ and CD4+ tumor-reactive TILs, respectively. In addition, biomarkers preferentially expressed on tumor-reactive TILs, rather than the frequency of CD3+, CD8+ and CD4+ lymphocytes, hold prognostic value suggesting their protective role in antitumor immunity.
Collapse
Affiliation(s)
- Jara Palomero
- Tumor Immunology and Immunotherapy, Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Carla Panisello
- Tumor Immunology and Immunotherapy, Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Maria Lozano-Rabella
- Tumor Immunology and Immunotherapy, Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Ricky Tirtakasuma
- Tumor Immunology and Immunotherapy, Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Judit Díaz-Gómez
- Tumor Immunology and Immunotherapy, Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Daniela Grases
- Tumor Immunology and Immunotherapy, Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Helena Pasamar
- Tumor Immunology and Immunotherapy, Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Laura Arregui
- HUB-ICO-IDIBELL Biobank, Bellvitge Institute for Biomedical Research, L'Hospitalet de Llobregat, Spain
| | - Eduard Dorca Duch
- Pathology, Bellvitge University Hospital, IDIBELL, L'Hospitalet de Llobregat, Spain
| | | | - Ana Vivancos
- Cancer Genomics, Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Carlos E de Andrea
- Pathology, Clinica Universidad de Navarra, Pamplona, Spain,Centro de Investigación Biomedica en Red de Cáncer (CIBERONC), Madrid, Spain
| | - Ignacio Melero
- Centro de Investigación Biomedica en Red de Cáncer (CIBERONC), Madrid, Spain,Program of Immunology and Immunotherapy, CIMA Universidad de Navarra, Pamplona, Spain,Navarra Institute for Health Research IDISNA, Pamplona, Spain
| | - Jordi Ponce
- Department of Gynaecology, Bellvitge University Hospital, L'Hospitalet de Llobregat, Spain
| | - August Vidal
- Pathology, Bellvitge University Hospital, IDIBELL, L'Hospitalet de Llobregat, Spain,Centro de Investigación Biomedica en Red de Cáncer (CIBERONC), Madrid, Spain
| | - Josep Maria Piulats
- Medical Oncology, Catalan Institute of Oncology (ICO), IDIBELL-OncoBell, L'Hospitalet de Llobregat, Spain
| | - Xavier Matias-Guiu
- Pathology, Bellvitge University Hospital, IDIBELL, L'Hospitalet de Llobregat, Spain,Centro de Investigación Biomedica en Red de Cáncer (CIBERONC), Madrid, Spain,Pathology, Arnau de Vilanova University Hospital, University of LLeida, IRBLLEIDA, Lleida, Spain
| | - Alena Gros
- Tumor Immunology and Immunotherapy, Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| |
Collapse
|