1
|
Liu WN, Harden SL, Tan SLW, Tan RJR, Fong SY, Tan SY, Liu M, Karnik I, Shuen TWH, Toh HC, Fan Y, Lim SG, Chan JKY, Chen Q. Single-cell RNA sequencing reveals anti-tumor potency of CD56 + NK cells and CD8 + T cells in humanized mice via PD-1 and TIGIT co-targeting. Mol Ther 2024; 32:3895-3914. [PMID: 39318093 PMCID: PMC11573594 DOI: 10.1016/j.ymthe.2024.09.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 08/16/2024] [Accepted: 09/19/2024] [Indexed: 09/26/2024] Open
Abstract
In solid tumors, the exhaustion of natural killer (NK) cells and cytotoxic T cells in the immunosuppressive tumor microenvironment poses challenges for effective tumor control. Conventional humanized mouse models of hepatocellular carcinoma patient-derived xenografts (HCC-PDX) encounter limitations in NK cell infiltration, hindering studies on NK cell immunobiology. Here, we introduce an improved humanized mouse model with restored NK cell reconstitution and infiltration in HCC-PDX, coupled with single-cell RNA sequencing (scRNA-seq) to identify potential anti-HCC treatments. A single administration of adeno-associated virus carrying human interleukin-15 reinstated persistent NK cell reconstitution and infiltration in HCC-PDX in humanized mice. scRNA-seq revealed NK cell and T cell subpopulations with heightened PDCD1 and TIGIT levels. Notably, combination therapy with anti-PD-1 and anti-TIGIT antibodies alleviated HCC burden in humanized mice, demonstrating NK cell-dependent efficacy. Bulk-RNA sequencing analysis also revealed significant alterations in the tumor transcriptome that may contribute to further resistance after combination therapy, warranting further investigations. As an emerging strategy, ongoing clinical trials with anti-PD-1 and anti-TIGIT antibodies provide limited data. The improved humanized mouse HCC-PDX model not only sheds light on the pivotal role of NK cells but also serves as a robust platform for evaluating safety and anti-tumor efficacy of combination therapies and other potential regimens, complementing clinical insights.
Collapse
MESH Headings
- Animals
- Killer Cells, Natural/immunology
- Killer Cells, Natural/metabolism
- Humans
- Mice
- Programmed Cell Death 1 Receptor/antagonists & inhibitors
- Programmed Cell Death 1 Receptor/metabolism
- Receptors, Immunologic/metabolism
- Receptors, Immunologic/genetics
- CD56 Antigen/metabolism
- CD56 Antigen/genetics
- Carcinoma, Hepatocellular/therapy
- Carcinoma, Hepatocellular/immunology
- Carcinoma, Hepatocellular/genetics
- Carcinoma, Hepatocellular/pathology
- Liver Neoplasms/therapy
- Liver Neoplasms/immunology
- Liver Neoplasms/genetics
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/metabolism
- Interleukin-15/metabolism
- Interleukin-15/genetics
- Xenograft Model Antitumor Assays
- Single-Cell Analysis/methods
- Tumor Microenvironment/immunology
- Disease Models, Animal
- Cell Line, Tumor
- Sequence Analysis, RNA/methods
- Dependovirus/genetics
Collapse
Affiliation(s)
- Wai Nam Liu
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A∗STAR), 61 Biopolis Drive, Proteos, Singapore 138673, Republic of Singapore
| | - Sarah L Harden
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A∗STAR), 61 Biopolis Drive, Proteos, Singapore 138673, Republic of Singapore
| | - Shawn Lu Wen Tan
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A∗STAR), 61 Biopolis Drive, Proteos, Singapore 138673, Republic of Singapore
| | - Rachel Jun Rou Tan
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A∗STAR), 61 Biopolis Drive, Proteos, Singapore 138673, Republic of Singapore
| | - Shin Yie Fong
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A∗STAR), 61 Biopolis Drive, Proteos, Singapore 138673, Republic of Singapore
| | - Sue Yee Tan
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A∗STAR), 61 Biopolis Drive, Proteos, Singapore 138673, Republic of Singapore
| | - Min Liu
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A∗STAR), 61 Biopolis Drive, Proteos, Singapore 138673, Republic of Singapore
| | - Isha Karnik
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A∗STAR), 61 Biopolis Drive, Proteos, Singapore 138673, Republic of Singapore
| | - Timothy Wai Ho Shuen
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore 168583, Republic of Singapore
| | - Han Chong Toh
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore 168583, Republic of Singapore
| | - Yong Fan
- Key Laboratory for Major Obstetric Diseases of Guangdong Province, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Seng Gee Lim
- Division of Gastroenterology and Hepatology, National University Hospital, Singapore 119228, Republic of Singapore; Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Republic of Singapore
| | - Jerry Kok Yen Chan
- Department of Reproductive Medicine, KK Women's and Children's Hospital, Singapore 229899, Republic of Singapore; Experimental Fetal Medicine Group, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117593, Republic of Singapore
| | - Qingfeng Chen
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A∗STAR), 61 Biopolis Drive, Proteos, Singapore 138673, Republic of Singapore; Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117593, Republic of Singapore; Singapore Immunology Network (SIgN), A∗STAR, 8A Biomedical Grove, Immunos, Singapore 138648, Republic of Singapore.
| |
Collapse
|
2
|
Baharom F, Hermans D, Delamarre L, Seder RA. Vax-Innate: improving therapeutic cancer vaccines by modulating T cells and the tumour microenvironment. Nat Rev Immunol 2024:10.1038/s41577-024-01091-9. [PMID: 39433884 DOI: 10.1038/s41577-024-01091-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/02/2024] [Indexed: 10/23/2024]
Abstract
T cells have a critical role in mediating antitumour immunity. The success of immune checkpoint inhibitors (ICIs) for cancer treatment highlights how enhancing endogenous T cell responses can mediate tumour regression. However, mortality remains high for many cancers, especially in the metastatic setting. Based on advances in the genetic characterization of tumours and identification of tumour-specific antigens, individualized therapeutic cancer vaccines targeting mutated tumour antigens (neoantigens) are being developed to generate tumour-specific T cells for improved therapeutic responses. Early clinical trials using individualized neoantigen vaccines for patients with advanced disease had limited clinical efficacy despite demonstrated induction of T cell responses. Therefore, enhancing T cell activity by improving the magnitude, quality and breadth of T cell responses following vaccination is one current goal for improving outcome against metastatic tumours. Another major consideration is how T cells can be further optimized to function within the tumour microenvironment (TME). In this Perspective, we focus on neoantigen vaccines and propose a new approach, termed Vax-Innate, in which vaccination through intravenous delivery or in combination with tumour-targeting immune modulators may improve antitumour efficacy by simultaneously increasing the magnitude, quality and breadth of T cells while transforming the TME into a largely immunostimulatory environment for T cells.
Collapse
Affiliation(s)
| | - Dalton Hermans
- Vaccine Research Center, National Institutes of Health, Bethesda, MD, USA
| | | | - Robert A Seder
- Vaccine Research Center, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
3
|
Skapinker E, Aucoin EB, Kombargi HL, Yaish AM, Li Y, Baghaie L, Szewczuk MR. Contemporaneous Inflammatory, Angiogenic, Fibrogenic, and Angiostatic Cytokine Profiles of the Time-to-Tumor Development by Cancer Cells to Orchestrate Tumor Neovascularization, Progression, and Metastasis. Cells 2024; 13:1739. [PMID: 39451257 PMCID: PMC11506673 DOI: 10.3390/cells13201739] [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: 09/21/2024] [Revised: 10/15/2024] [Accepted: 10/16/2024] [Indexed: 10/26/2024] Open
Abstract
Cytokines can promote various cancer processes, such as angiogenesis, epithelial to mesenchymal transition (EMT), invasion, and tumor progression, and maintain cancer stem-cell-like (CSCs) cells. The mechanism(s) that continuously promote(s) tumors to progress in the TME still need(s) to be investigated. The data in the present study analyzed the inflammatory, angiogenic, fibrogenic, and angiostatic cytokine profiles in the host serum during tumor development in a mouse model of human pancreatic cancer. Pancreatic MiaPaCa-2-eGFP cancer cells were subcutaneously implanted in RAG2xCγ double mutant mice. Blood samples were collected before cancer cell implantation and every week until the end point of the study. The extracted serum from the blood of each mouse at different time points during tumor development was analyzed using a Bio-Plex microarray analysis and a Bio-Plex 200 system for proinflammatory (IL-1β, IL-10, IFN-γ, and TNF-α) and angiogenic and fibrogenic (IL-15, IL-18, basic FGF, LIF, M-CSF, MIG, MIP-2, PDGF-BB, and VEGF) cytokines. Here, we find that during cancer cell colonization for tumor development, host angiogenic, fibrogenic, and proinflammatory cytokine profiling in the tumor-bearing mice has been shown to significantly reduce host angiostatic and proinflammatory cytokines that restrain tumor development and increase those for tumor growth. The proinflammatory cytokines IL-15, IL-18, and IL-1β profiles reveal a significant host serum increase after day 35 when the tumor began to progress in growth. In contrast, the angiostatic cytokine profiles of TNFα, MIG, M-CSF, IL-10, and IFNγ in the host serum revealed a dramatic and significant decrease after day 5 post-implantation of cancer cells. OP treatment of tumor-bearing mice on day 35 maintained high levels of angiostatic and fibrogenic cytokines. The data suggest an entirely new regulation by cancer cells for tumor development. The findings identify for the first time how pancreatic cancer cells use host cytokine profiling to orchestrate the initiation of tumor development.
Collapse
Affiliation(s)
- Elizabeth Skapinker
- Faculty of Arts and Science, Queen’s University, Kingston, ON K7L 3N9, Canada; (E.S.); (Y.L.)
| | - Emilyn B. Aucoin
- Faculty of Science, Biology (Biomedical Science), York University, Toronto, ON M3J 1P3, Canada;
| | - Haley L. Kombargi
- Faculty of Health Sciences, Queen’s University, Kingston, ON K7L 3N9, Canada; (H.L.K.); (A.M.Y.)
| | - Abdulrahman M. Yaish
- Faculty of Health Sciences, Queen’s University, Kingston, ON K7L 3N9, Canada; (H.L.K.); (A.M.Y.)
| | - Yunfan Li
- Faculty of Arts and Science, Queen’s University, Kingston, ON K7L 3N9, Canada; (E.S.); (Y.L.)
| | - Leili Baghaie
- Department of Biomedical and Molecular Sciences, Queen’s University, Kingston, ON K7L 3N9, Canada;
| | - Myron R. Szewczuk
- Department of Biomedical and Molecular Sciences, Queen’s University, Kingston, ON K7L 3N9, Canada;
| |
Collapse
|
4
|
Fischer A, Albert TK, Moreno N, Interlandi M, Mormann J, Glaser S, Patil P, de Faria FW, Richter M, Verma A, Balbach ST, Wagener R, Bens S, Dahlum S, Göbel C, Münter D, Inserte C, Graf M, Kremer E, Melcher V, Di Stefano G, Santi R, Chan A, Dogan A, Bush J, Hasselblatt M, Cheng S, Spetalen S, Fosså A, Hartmann W, Herbrüggen H, Robert S, Oyen F, Dugas M, Walter C, Sandmann S, Varghese J, Rossig C, Schüller U, Tzankov A, Pedersen MB, d'Amore FA, Mellgren K, Kontny U, Kancherla V, Veloza L, Missiaglia E, Fataccioli V, Gaulard P, Burkhardt B, Soehnlein O, Klapper W, de Leval L, Siebert R, Kerl K. Lack of SMARCB1 expression characterizes a subset of human and murine peripheral T-cell lymphomas. Nat Commun 2024; 15:8571. [PMID: 39362842 PMCID: PMC11452211 DOI: 10.1038/s41467-024-52826-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Accepted: 09/23/2024] [Indexed: 10/05/2024] Open
Abstract
Peripheral T-cell lymphoma, not otherwise specified (PTCL-NOS) is a heterogeneous group of malignancies with poor outcome. Here, we identify a subgroup, PTCL-NOSSMARCB1-, which is characterized by the lack of the SMARCB1 protein and occurs more frequently in young patients. Human and murine PTCL-NOSSMARCB1- show similar DNA methylation profiles, with hypermethylation of T-cell-related genes and hypomethylation of genes involved in myeloid development. Single-cell analyses of human and murine tumors revealed a rich and complex network of interactions between tumor cells and an immunosuppressive and exhausted tumor microenvironment (TME). In a drug screen, we identified histone deacetylase inhibitors (HDACi) as a class of drugs effective against PTCL-NOSSmarcb1-. In vivo treatment of mouse tumors with SAHA, a pan-HDACi, triggered remodeling of the TME, promoting replenishment of lymphoid compartments and reversal of the exhaustion phenotype. These results provide a rationale for further exploration of HDACi combination therapies targeting PTCL-NOSSMARCB1- within the TME.
Collapse
MESH Headings
- Animals
- SMARCB1 Protein/genetics
- SMARCB1 Protein/metabolism
- Humans
- Lymphoma, T-Cell, Peripheral/genetics
- Lymphoma, T-Cell, Peripheral/drug therapy
- Lymphoma, T-Cell, Peripheral/metabolism
- Lymphoma, T-Cell, Peripheral/pathology
- Mice
- Histone Deacetylase Inhibitors/pharmacology
- Tumor Microenvironment/genetics
- Tumor Microenvironment/drug effects
- DNA Methylation
- Gene Expression Regulation, Neoplastic
- Female
- Cell Line, Tumor
- Male
- Vorinostat/pharmacology
- Single-Cell Analysis
Collapse
Affiliation(s)
- Anja Fischer
- Institute of Human Genetics, Ulm University Medical Center, Ulm, Germany
| | - Thomas K Albert
- Department of Pediatric Hematology and Oncology, University Children's Hospital Münster, Münster, Germany
| | - Natalia Moreno
- Department of Pediatric Hematology and Oncology, University Children's Hospital Münster, Münster, Germany
| | - Marta Interlandi
- Department of Pediatric Hematology and Oncology, University Children's Hospital Münster, Münster, Germany
- Institute of Medical Informatics, University of Münster, 48149, Münster, Germany
| | - Jana Mormann
- Department of Pediatric Hematology and Oncology, University Children's Hospital Münster, Münster, Germany
| | - Selina Glaser
- Institute of Human Genetics, Ulm University Medical Center, Ulm, Germany
| | - Paurnima Patil
- Institute of Human Genetics, Ulm University Medical Center, Ulm, Germany
| | - Flavia W de Faria
- Department of Pediatric Hematology and Oncology, University Children's Hospital Münster, Münster, Germany
| | - Mathis Richter
- Institute for Experimental Pathology, Center for Molecular Biology of Inflammation, University of Münster, Münster, Germany
| | - Archana Verma
- Department of Pediatric Hematology and Oncology, University Children's Hospital Münster, Münster, Germany
| | - Sebastian T Balbach
- Department of Pediatric Hematology and Oncology, University Children's Hospital Münster, Münster, Germany
| | - Rabea Wagener
- Institute of Human Genetics, Ulm University Medical Center, Ulm, Germany
| | - Susanne Bens
- Institute of Human Genetics, Ulm University Medical Center, Ulm, Germany
| | - Sonja Dahlum
- Institute of Human Genetics, Ulm University Medical Center, Ulm, Germany
| | - Carolin Göbel
- Department of Pediatric Hematology and Oncology, University Medical Center Hamburg, Eppendorf (UKE), 20251, Hamburg, Germany
- Research Institute Children's Cancer Center, 20251, Hamburg, Germany
| | - Daniel Münter
- Department of Pediatric Hematology and Oncology, University Children's Hospital Münster, Münster, Germany
| | - Clara Inserte
- Institute of Medical Informatics, University of Münster, 48149, Münster, Germany
| | - Monika Graf
- Department of Pediatric Hematology and Oncology, University Children's Hospital Münster, Münster, Germany
| | - Eva Kremer
- Department of Pediatric Hematology and Oncology, University Children's Hospital Münster, Münster, Germany
| | - Viktoria Melcher
- Department of Pediatric Hematology and Oncology, University Children's Hospital Münster, Münster, Germany
| | - Gioia Di Stefano
- Pathological Anatomy Section, Careggi University Hospital, Florence, Italy
| | - Raffaella Santi
- Pathological Anatomy Section, Careggi University Hospital, Florence, Italy
| | - Alexander Chan
- Department of Pathology, Hematopathology Service, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | - Ahmet Dogan
- Department of Pathology, Hematopathology Service, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | - Jonathan Bush
- Division of Anatomical Pathology, British Columbia Children's Hospital and Women's Hospital and Health Center, Vancouver, BC, Canada
| | - Martin Hasselblatt
- Institute of Neuropathology, University Hospital Münster, 48149, Münster, Germany
| | - Sylvia Cheng
- Division of Pediatric Hematology/Oncology/BMT, Department of Pediatrics, British Columbia Children's Hospital, University of British Columbia, Vancouver, BC, Canada
| | - Signe Spetalen
- Department of Pathology, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Alexander Fosså
- Department of Oncology, Oslo University Hospital-Norwegian Radium Hospital, Oslo, Norway
| | - Wolfgang Hartmann
- Division of Translational Pathology, Gerhard-Domagk-Institut für Pathologie, Universitätsklinikum Münster, Albert-Schweitzer-Campus 1, Gebäude D17, 48149, Münster, Germany
| | - Heidi Herbrüggen
- Department of Pediatric Hematology and Oncology, University Children's Hospital Münster, Münster, Germany
| | - Stella Robert
- Department of Medicine A, Hematology, Oncology, and Pneumology, University Hospital Münster, Münster, Germany
| | - Florian Oyen
- Department of Pediatric Hematology and Oncology, University Medical Center Hamburg, Eppendorf (UKE), 20251, Hamburg, Germany
| | - Martin Dugas
- Institute of Medical Informatics, University of Münster, 48149, Münster, Germany
- Institute of Medical Informatics, Heidelberg University Hospital, Heidelberg, Germany
| | - Carolin Walter
- Institute of Medical Informatics, University of Münster, 48149, Münster, Germany
| | - Sarah Sandmann
- Institute of Medical Informatics, University of Münster, 48149, Münster, Germany
| | - Julian Varghese
- Institute of Medical Informatics, University of Münster, 48149, Münster, Germany
| | - Claudia Rossig
- Department of Pediatric Hematology and Oncology, University Children's Hospital Münster, Münster, Germany
| | - Ulrich Schüller
- Department of Pediatric Hematology and Oncology, University Medical Center Hamburg, Eppendorf (UKE), 20251, Hamburg, Germany
- Research Institute Children's Cancer Center, 20251, Hamburg, Germany
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf (UKE), 20251, Hamburg, Germany
| | - Alexandar Tzankov
- Institute of Medical Genetics and Pathology, University Hospital Basel, Basel, Switzerland
| | - Martin B Pedersen
- Department of Hematology, Aarhus University Hospital, Aarhus, Denmark
| | - Francesco A d'Amore
- Department of Hematology, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Karin Mellgren
- Department of Pediatric Oncology and Hematology, Sahlgrenska University Hospital, The Queen Silvia Children's Hospital, Gothenburg, Sweden
| | - Udo Kontny
- Section of Pediatric Hematology, Oncology, and Stem Cell Transplantation, Department of Pediatric and Adolescent Medicine, RWTH Aachen University Hospital, Aachen, Germany
| | - Venkatesh Kancherla
- Institute of Pathology, Department of Laboratory Medicine and Pathology, Lausanne University Hospital, Lausanne, Switzerland
| | - Luis Veloza
- Institute of Pathology, Department of Laboratory Medicine and Pathology, Lausanne University Hospital, Lausanne, Switzerland
| | - Edoardo Missiaglia
- Institute of Pathology, Department of Laboratory Medicine and Pathology, Lausanne University Hospital, Lausanne, Switzerland
| | - Virginie Fataccioli
- INSERM U955, Université Paris-Est, Créteil, France
- Département de Pathologie, Hôpitaux Universitaires Henri Mondor, AP-HP, INSERM U955, Université Paris Est Créteil, Créteil, France
| | - Philippe Gaulard
- Département de Pathologie, Hôpitaux Universitaires Henri Mondor, AP-HP, INSERM U955, Université Paris Est Créteil, Créteil, France
| | - Birgit Burkhardt
- Department of Pediatric Hematology and Oncology, University Children's Hospital Münster, Münster, Germany
| | - Oliver Soehnlein
- Institute for Experimental Pathology, Center for Molecular Biology of Inflammation, University of Münster, Münster, Germany
| | - Wolfram Klapper
- Department of Pathology, Haematopathology Section and Lymph Node Registry, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Laurence de Leval
- Institute of Pathology, Department of Laboratory Medicine and Pathology, Lausanne University Hospital, Lausanne, Switzerland
| | - Reiner Siebert
- Institute of Human Genetics, Ulm University Medical Center, Ulm, Germany
| | - Kornelius Kerl
- Department of Pediatric Hematology and Oncology, University Children's Hospital Münster, Münster, Germany.
| |
Collapse
|
5
|
Norgard RJ, Budhani P, O'Brien SA, Xia Y, Egan JN, Flynn B, Tagore JR, Seco J, Peet GW, Mikucka A, Wasti R, Chan LC, Hinkel M, Martinez-Morilla S, Pignatelli J, Trapani F, Corse E, Feng D, Kostyrko K, Hofmann MH, Liu K, Kashyap AS. Reshaping the Tumor Microenvironment of KRASG12D Pancreatic Ductal Adenocarcinoma with Combined SOS1 and MEK Inhibition for Improved Immunotherapy Response. CANCER RESEARCH COMMUNICATIONS 2024; 4:1548-1560. [PMID: 38727236 PMCID: PMC11191876 DOI: 10.1158/2767-9764.crc-24-0172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Revised: 05/02/2024] [Accepted: 05/07/2024] [Indexed: 06/23/2024]
Abstract
KRAS inhibitors have demonstrated exciting preclinical and clinical responses, although resistance occurs rapidly. Here, we investigate the effects of KRAS-targeting therapies on the tumor microenvironment using a library of KrasG12D, p53-mutant, murine pancreatic ductal adenocarcinoma-derived cell lines (KPCY) to leverage immune-oncology combination strategies for long-term tumor efficacy. Our findings show that SOS1 and MEK inhibitors (SOS1i+MEKi) suppressed tumor growth in syngeneic models and increased intratumoral CD8+ T cells without durable responses. Single-cell RNA sequencing revealed an increase in inflammatory cancer-associated fibroblasts (iCAF), M2 macrophages, and a decreased dendritic cell (DC) quality that ultimately resulted in a highly immunosuppressive microenvironment driven by IL6+ iCAFs. Agonist CD40 treatment was effective to revert macrophage polarization and overcome the lack of mature antigen-presenting DCs after SOS1i+MEKi therapy. Treatment increased the overall survival of KPCY tumor-bearing mice. The addition of checkpoint blockade to SOS1i+MEKi combination resulted in tumor-free mice with established immune memory. Our data suggest that KRAS inhibition affects myeloid cell maturation and highlights the need for combining KRAS cancer-targeted therapy with myeloid activation to enhance and prolong antitumor effects. SIGNIFICANCE Combination of SOS1 and MEK inhibitors increase T cell infiltration while blunting pro-immune myeloid cell maturation and highlights the need for combining KRAS cancer-targeted therapy with myeloid activation to enhance and prolong anti-tumor effects.
Collapse
Affiliation(s)
- Robert J. Norgard
- Cancer Immunology and Immune Modulation, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut
| | - Pratha Budhani
- Cancer Immunology and Immune Modulation, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut
| | - Sarah A. O'Brien
- Cancer Immunology and Immune Modulation, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut
| | - Youli Xia
- Global Computational Biology and Digital Sciences, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut
| | - Jessica N. Egan
- Cancer Immunology and Immune Modulation, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut
| | - Brianna Flynn
- Cancer Immunology and Immune Modulation, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut
| | - Joshua R. Tagore
- Cancer Immunology and Immune Modulation, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut
| | - Joseph Seco
- Cancer Immunology and Immune Modulation, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut
| | - Gregory W. Peet
- Cancer Immunology and Immune Modulation, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut
| | - Ania Mikucka
- Cancer Immunology and Immune Modulation, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut
| | - Ruby Wasti
- Cancer Immunology and Immune Modulation, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut
| | - Li-Chuan Chan
- Cancer Immunology and Immune Modulation, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut
| | - Melanie Hinkel
- Late Stage Cancer Research, Boehringer Ingelheim RCV GmbH & Co KG, Vienna, Austria
| | - Sandra Martinez-Morilla
- Translational Medicine and Clinical Pharmacology, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut
| | - Jeanine Pignatelli
- Cancer Immunology and Immune Modulation, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut
| | - Francesca Trapani
- Translational Medicine and Clinical Pharmacology, Boehringer Ingelheim RCV GmbH & Co KG, Vienna, Austria
| | - Emily Corse
- Cancer Immunology and Immune Modulation, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut
| | - Di Feng
- Global Computational Biology and Digital Sciences, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut
| | - Kaja Kostyrko
- Late Stage Cancer Research, Boehringer Ingelheim RCV GmbH & Co KG, Vienna, Austria
| | - Marco H. Hofmann
- Cancer Pharmacology and Disease Positioning Department, Boehringer Ingelheim RCV GmbH & Co KG, Vienna, Austria
| | - Kang Liu
- Cancer Immunology and Immune Modulation, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut
| | - Abhishek S. Kashyap
- Cancer Immunology and Immune Modulation, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut
| |
Collapse
|
6
|
Song N, Cui K, Zeng L, Li M, Fan Y, Shi P, Wang Z, Su W, Wang H. Advance in the role of chemokines/chemokine receptors in carcinogenesis: Focus on pancreatic cancer. Eur J Pharmacol 2024; 967:176357. [PMID: 38309677 DOI: 10.1016/j.ejphar.2024.176357] [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: 09/13/2023] [Revised: 01/17/2024] [Accepted: 01/23/2024] [Indexed: 02/05/2024]
Abstract
The chemokines/chemokine receptors pathway significantly influences cell migration, particularly in recruiting immune cells to the tumor microenvironment (TME), impacting tumor progression and treatment outcomes. Emerging research emphasizes the involvement of chemokines in drug resistance across various tumor therapies, including immunotherapy, chemotherapy, and targeted therapy. This review focuses on the role of chemokines/chemokine receptors in pancreatic cancer (PC) development, highlighting their impact on TME remodeling, immunotherapy, and relevant signaling pathways. The unique immunosuppressive microenvironment formed by the interaction of tumor cells, stromal cells and immune cells plays an important role in the tumor proliferation, invasion, migration and therapeutic resistance. Chemokines/chemokine receptors, such as chemokine ligand (CCL) 2, CCL3, CCL5, CCL20, CCL21, C-X-C motif chemokine ligand (CXCL) 1, CXCL2, CXCL3, CXCL4, CXCL5, CXCL8, CXCL9, CXCL10, CXCL11, CXCL12, CXCL13, CXCL14, CXCL16, CXCL17, and C-X3-C motif chemokine ligand (CX3CL)1, derived mainly from leukocyte cells, cancer-related fibroblasts (CAFs), pancreatic stellate cells (PSCs), and tumor-associated macrophages (TAMs), contribute to PC progression and treatment resistance. Chemokines recruit myeloid-derived suppressor cells (MDSC), regulatory T cells (Tregs), and M2 macrophages, inhibiting the anti-tumor activity of immune cells. Simultaneously, they enhance pathways like epithelial-mesenchymal transition (EMT), Akt serine/threonine kinase (AKT), extracellular regulated protein kinases (ERK) 1/2, and nuclear factor kappa-B (NF-κB), etc., elevating the risk of PC metastasis and compromising the efficacy of radiotherapy, chemotherapy, and anti-PD-1/PD-L1 immunotherapy. Notably, the CCLx-CCR2 and CXCLx-CXCR2/4 axis emerge as potential therapeutic targets in PC. This review integrates recent findings on chemokines and receptors in PC treatment, offering valuable insights for innovative therapeutic approaches.
Collapse
Affiliation(s)
- Na Song
- Department of Pathology, Xinxiang Key Laboratory of Precision Medicine, The First Affiliated Hospital of Xinxiang Medical University, China; Department of Pathology, Xinxiang Medical University, Xinxiang, 453000, China
| | - Kai Cui
- Department of Pathology, Xinxiang Medical University, Xinxiang, 453000, China
| | - Liqun Zeng
- Department of Pathology, Xinxiang Medical University, Xinxiang, 453000, China
| | - Mengxiao Li
- Department of Pathology, Xinxiang Key Laboratory of Precision Medicine, The First Affiliated Hospital of Xinxiang Medical University, China
| | - Yanwu Fan
- Department of Pathology, Xinxiang Medical University, Xinxiang, 453000, China
| | - Pingyu Shi
- Department of Pathology, Xinxiang Medical University, Xinxiang, 453000, China
| | - Ziwei Wang
- Department of Pathology, Xinxiang Medical University, Xinxiang, 453000, China
| | - Wei Su
- Department of Pathology, Xinxiang Key Laboratory of Precision Medicine, The First Affiliated Hospital of Xinxiang Medical University, China.
| | - Haijun Wang
- Department of Pathology, Xinxiang Key Laboratory of Precision Medicine, The First Affiliated Hospital of Xinxiang Medical University, China; Department of Pathology, Xinxiang Medical University, Xinxiang, 453000, China.
| |
Collapse
|
7
|
Zhang J, Wang L, Jiang M. Diagnostic value of sphingolipid metabolism-related genes CD37 and CXCL9 in nonalcoholic fatty liver disease. Medicine (Baltimore) 2024; 103:e37185. [PMID: 38394483 PMCID: PMC11309649 DOI: 10.1097/md.0000000000037185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Accepted: 01/17/2024] [Indexed: 02/25/2024] Open
Abstract
The development of nonalcoholic fatty liver disease (NAFLD) has been reported to be caused by sphingolipid family inducing insulin resistance, mitochondrial dysfunction, and inflammation, which can be regulated by multiple sphingolipid metabolic pathways. This study aimed to explore the molecular mechanism of crucial sphingolipid metabolism related genes (SMRGs) in NAFLD. Firstly, the datasets (GSE48452, GSE126848, and GSE63067) from the Gene Expression Omnibus database and sphingolipid metabolism genes (SMGs) from previous research were collected for this study. The differentially expressed genes (DEGs) between different NAFLD and controls were acquired through "limma," and the SMRGs were authenticated via weighted gene co-expression network analysis (WGCNA). After overlapping the DEGs and SMRGs, the causality between the intersection genes (DE-SMRGs) and NAFLD was explored to sort out the candidate biomarkers by Mendelian randomization (MR) study. The receiver operating characteristic (ROC) curves of candidate biomarkers in GSE48452 and GSE126848 were yielded to determine the biomarkers, followed by the nomogram construction and enrichment analysis. Finally, the immune infiltration analysis, the prediction of transcription factors (TFs) and drugs targeting biomarkers were put into effect. A total of 23 DE-SMRGs were acquired based on the differential analysis and weighted gene co-expression network analysis (WGCNA), of which 3 DE-SMRGs (CD37, CXCL9 and IL7R) were picked out for follow-up analysis through univariate and multivariate MR analysis. The values of area under ROC curve of CD37 and CXCL9 were >0.7 in GSE48452 and GSE126848, thereby being regarded as biomarkers, which were mainly enriched in amino acid metabolism. With respect to the Spearman analysis between immune cells and biomarkers, CD37 and CXCL9 were significantly positively associated with M1 macrophages (P < .001), whose proportion was observably higher in NAFLD patients compared with controls. At last, TFs (ZNF460 and ZNF384) of CD37 and CXCL9 and a total of 79 chemical drugs targeting CD37 and CXCL9 were predicted. This study mined the pivotal SMRGs, CD37 and CXCL9, and systematically explored the mechanism of action of both biomarkers based on the public databases, which could tender a fresh reference for the clinical diagnosis and therapy of NAFLD.
Collapse
Affiliation(s)
- Jiayi Zhang
- Queen Mary School, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Lingfang Wang
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Meixiu Jiang
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| |
Collapse
|
8
|
Zhang Y, Zhang G, Wang Y, Ye L, Peng L, Shi R, Guo S, He J, Yang H, Dai Q. Current treatment strategies targeting histone deacetylase inhibitors in acute lymphocytic leukemia: a systematic review. Front Oncol 2024; 14:1324859. [PMID: 38450195 PMCID: PMC10915758 DOI: 10.3389/fonc.2024.1324859] [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: 10/20/2023] [Accepted: 01/29/2024] [Indexed: 03/08/2024] Open
Abstract
Acute lymphocytic leukemia is a hematological malignancy that primarily affects children. Long-term chemotherapy is effective, but always causes different toxic side effects. With the application of a chemotherapy-free treatment strategy, we intend to demonstrate the most recent results of using one type of epigenetic drug, histone deacetylase inhibitors, in ALL and to provide preclinical evidence for further clinical trials. In this review, we found that panobinostat (LBH589) showed positive outcomes as a monotherapy, whereas vorinostat (SAHA) was a better choice for combinatorial use. Preclinical research has identified chidamide as a potential agent for investigation in more clinical trials in the future. In conclusion, histone deacetylase inhibitors play a significant role in the chemotherapy-free landscape in cancer treatment, particularly in acute lymphocytic leukemia.
Collapse
Affiliation(s)
- Yingjun Zhang
- Department of Laboratory Medicine, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, Sichuan, China
| | - Ge Zhang
- Department of Laboratory Medicine, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, Sichuan, China
| | - Yuefang Wang
- Department of Laboratory Medicine, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, Sichuan, China
| | - Lei Ye
- Department of Laboratory Medicine, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, Sichuan, China
| | - Luyun Peng
- Department of Laboratory Medicine, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, Sichuan, China
| | - Rui Shi
- Department of Laboratory Medicine, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, Sichuan, China
| | - Siqi Guo
- Department of Laboratory Medicine, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, Sichuan, China
| | - Jiajing He
- Department of Laboratory Medicine, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, Sichuan, China
| | - Hao Yang
- Department of Laboratory Medicine, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, Sichuan, China
| | - Qingkai Dai
- Department of Laboratory Medicine, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, Sichuan, China
| |
Collapse
|
9
|
Zou Z, Chen S, Wu Y, Ji S. The USP35-CXCR3 Axis plays an oncogenic role in JeKo-1 mantle cell lymphoma cells. Integr Biol (Camb) 2024; 16:zyae021. [PMID: 39591978 DOI: 10.1093/intbio/zyae021] [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: 07/09/2024] [Revised: 11/04/2024] [Accepted: 11/21/2024] [Indexed: 11/28/2024]
Abstract
In B cells, the chemokine receptor CXCR3 is expressed only by a subset of B cells. However, CXCR3 is highly expressed in a rare type of B-cell lymphoma known as Mantle Cell Lymphoma (MCL) and CXCR3 inhibitor impairs proliferation and induces apoptosis in the MCL cell line JeKo-1. Despite this, the mechanism responsible for maintaining high levels of CXCR3 in MCL cells remains unclear. In this study, we assessed CXCR3 expression and amplification in MCL samples and confirmed that CXCR3 is overexpressed in MCL tissues. We also observed that CXCR3 amplification is present in a small portion of MCL patients and is associated with MCL classification. We then screened ubiquitin-specific proteases (USPs) that might control the degradation of CXCR3 protein. Our investigation revealed that USP35 acts as a potent stabilizer of CXCR3 protein. Knockdown of USP35 substantially reduced the CXCR3 protein levels in JeKo-1 cells, resulting in reduced cell viability, cell cycle arrest, increased apoptosis, and mitigated migration and invasion in these cells. At the molecular level, USP35 deubiquitinates and stabilizes CXCR3. USP35 deficiency attenuated the activation of the JAK1/STAT1 pathway and reduced the expression of β-catenin and c-Myc in JeKo-1 cells. Furthermore, we observed that overexpression of CXCR3 rescued the impaired tumorigenicity of USP35-deficient JeKo-1 cells, and the mechanism may be related to the fact that USP35 promotes CXCR3 deubiquitination to stabilize its expression. These findings collectively demonstrate the oncogenic role of the USP35-CXCR3 axis in JeKo-1 MCL cells.
Collapse
Affiliation(s)
- Zongkai Zou
- Department of Pathology, Zhangzhou Affiliated Hospital of Fujian Medical University, No. 59, Shengli West Road, Xiangcheng District, Zhangzhou 363000, Fujian, China
| | - Shumin Chen
- Department of Basic Medicine, Zhangzhou Health Vocational College; Collaborative Innovation Center for Translation Medical Testing and Application Technology, No. 29, Shengli West Road, Xiangcheng District, Zhangzhou 363000, Fujian, China
| | - Yonghe Wu
- Department of Pathology, Zhangzhou Affiliated Hospital of Fujian Medical University, No. 59, Shengli West Road, Xiangcheng District, Zhangzhou 363000, Fujian, China
| | - Siling Ji
- Department of Pathology, Zhangzhou Affiliated Hospital of Fujian Medical University, No. 59, Shengli West Road, Xiangcheng District, Zhangzhou 363000, Fujian, China
| |
Collapse
|
10
|
Hu S, Duan H, Lu Y, Huang S. The genes regulating sensitivity of tumor cells to T cell-mediated killing: could they be potential personalized immunotherapeutic targets in head and neck squamous cell carcinoma? Discov Oncol 2023; 14:199. [PMID: 37926766 PMCID: PMC10625926 DOI: 10.1007/s12672-023-00806-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Accepted: 10/09/2023] [Indexed: 11/07/2023] Open
Abstract
OBJECTIVE To identify the pivotal genes, specifically the STTK genes, that govern the sensitivity of tumor cells to T cell-mediated killing in Head and Neck Squamous Cell Carcinoma (HNSC). METHODS The differentially expressed genes (DEGs) in HNSC and STTK genes were overlapped to obtain the DE-STTK genes. Univariate and LASSO regression analyses were conducted to identify the pivotal DE-STTK genes that serve as hubs in HNSC (i.e., hub DE-STTK genes). The risk model was established to divide HNSC tumor samples into high- and low-risk groups based on the hub DE-STTK genes. Further investigations were carried out by examing the expression level, prognostic values, diagnostic values, enriched signaling pathways, correlation with tumor mutation burden (TMB), and association with tumor immune infiltration cells (TIICs). RESULTS A total of 71 genes were found to be overlapped between DEGs in HNSC and STTK genes. Lasso regression analysis identified 9 hub genes which were MYF6, AATF, AURKA, CXCL9, DPM2, MYO1B, NCBP2, TNFRSF12A, and TRAF1. The network analysis of hub DE-STTK genes-pathway reveals that these 9 hub genes exhibit enrichment in multiple signaling pathways, including toll-like receptor signaling, TNF signaling, NF-kappa B signaling, cytokine-cytokine receptor interaction, spliceosome, mRNA surveillance pathway, nucleocytoplasmic transport, GPI-anchor biosynthesis, as well as N-Glycan biosynthesis. The Pearson correlation analysis showed that the majority of correlations between 9 hub DE-STTK genes and immune cells were positive. CONCLUSION The 9 identified hub DE-STTK genes (MYF6, AATF, AURKA, CXCL9, DPM2, MYO1B, NCBP2, TNFRSF12A, and TRAF1) are presumptively implicated in the modulation of tumor immunity in HNSC. These genes, along with their enriched pathways, hold promise as potential personalized immunotherapeutic targets for the treatment of HNSC, thereby offering novel avenues for therapeutic intervention in this malignancy.
Collapse
Affiliation(s)
- Shaonan Hu
- Stomatological Hospital, School of Stomatology, Southern Medical University, 366 Jiangnan South Avenue, Haizhu District, Guangzhou, 510280, Guangdong, China
| | - Heng Duan
- Stomatological Hospital, School of Stomatology, Southern Medical University, 366 Jiangnan South Avenue, Haizhu District, Guangzhou, 510280, Guangdong, China
| | - Yongtao Lu
- Stomatological Hospital, School of Stomatology, Southern Medical University, 366 Jiangnan South Avenue, Haizhu District, Guangzhou, 510280, Guangdong, China.
| | - Shaohong Huang
- Stomatological Hospital, School of Stomatology, Southern Medical University, 366 Jiangnan South Avenue, Haizhu District, Guangzhou, 510280, Guangdong, China.
| |
Collapse
|
11
|
Varisli L, Dancik GM, Tolan V, Vlahopoulos S. Critical Roles of SRC-3 in the Development and Progression of Breast Cancer, Rendering It a Prospective Clinical Target. Cancers (Basel) 2023; 15:5242. [PMID: 37958417 PMCID: PMC10648290 DOI: 10.3390/cancers15215242] [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: 10/06/2023] [Revised: 10/27/2023] [Accepted: 10/30/2023] [Indexed: 11/15/2023] Open
Abstract
Breast cancer (BCa) is the most frequently diagnosed malignant tumor in women and is also one of the leading causes of cancer-related death. Most breast tumors are hormone-dependent and estrogen signaling plays a critical role in promoting the survival and malignant behaviors of these cells. Estrogen signaling involves ligand-activated cytoplasmic estrogen receptors that translocate to the nucleus with various co-regulators, such as steroid receptor co-activator (SRC) family members, and bind to the promoters of target genes and regulate their expression. SRC-3 is a member of this family that interacts with, and enhances, the transcriptional activity of the ligand activated estrogen receptor. Although SRC-3 has important roles in normal homeostasis and developmental processes, it has been shown to be amplified and overexpressed in breast cancer and to promote malignancy. The malignancy-promoting potential of SRC-3 is diverse and involves both promoting malignant behavior of tumor cells and creating a tumor microenvironment that has an immunosuppressive phenotype. SRC-3 also inhibits the recruitment of tumor-infiltrating lymphocytes with effector function and promotes stemness. Furthermore, SRC-3 is also involved in the development of resistance to hormone therapy and immunotherapy during breast cancer treatment. The versatility of SRC-3 in promoting breast cancer malignancy in this way makes it a good target, and methodical targeting of SRC-3 probably will be important for the success of breast cancer treatment.
Collapse
Affiliation(s)
- Lokman Varisli
- Department of Molecular Biology and Genetics, Science Faculty, Dicle University, Diyarbakir 21280, Turkey;
| | - Garrett M. Dancik
- Department of Computer Science, Eastern Connecticut State University, Willimantic, CT 06226, USA;
| | - Veysel Tolan
- Department of Molecular Biology and Genetics, Science Faculty, Dicle University, Diyarbakir 21280, Turkey;
| | - Spiros Vlahopoulos
- First Department of Pediatrics, National and Kapodistrian University of Athens, Thivon & Levadeias 8, Goudi, 11527 Athens, Greece
| |
Collapse
|
12
|
Cui L, Zhu L, Chen J, Li C, Yu Y, Xu S. Systematic Pan-Cancer Analysis Reveals X-C Motif Chemokine Receptor 1 as a Prognostic and Immunological Biomarker. Genes (Basel) 2023; 14:1961. [PMID: 37895310 PMCID: PMC10606244 DOI: 10.3390/genes14101961] [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: 09/17/2023] [Revised: 10/13/2023] [Accepted: 10/17/2023] [Indexed: 10/29/2023] Open
Abstract
Chemokines and their receptors play an important role in immune monitoring and immune defense during tumor growth and metastasis. However, their prognostic roles in pan-cancer have not been elucidated. In this work, we screened all chemokine receptors in pan-cancer and discovered X-C Motif Chemokine Receptor 1 (XCR1) as a reliable immunological and prognostic biomarker in pan-cancer using bioinformation. The TCGA database served as the foundation for the primary research database analysis in this work. XCR1 was downregulated in tumors. Patients with reduced XCR1 showed worse prognoses and a concomitant decrease in immune cell infiltration (DCs and CD8+ T cells). According to a gene enrichment study, XCR1 enhanced immune system performance by promoting T-cell infiltration through the C-X-C Motif Chemokine Ligand 9 (CXCL9)- C-X-C Motif Chemokine Receptor 3 (CXCR3) axis. In addition, XCR1 is mainly expressed in infiltrated DCs and some malignant cells in tumor tissues. Our data revealed the important role of XCR1 in remodeling the tumor microenvironment and predicting the survival prognosis, which could also be used as a sensitive biomarker for tumor immunotherapy.
Collapse
Affiliation(s)
- Likun Cui
- National Key Laboratory of Medical Immunology and Institute of Immunology, Naval Medical University, Shanghai 200433, China; (L.C.); (L.Z.); (J.C.); (C.L.)
| | - Liye Zhu
- National Key Laboratory of Medical Immunology and Institute of Immunology, Naval Medical University, Shanghai 200433, China; (L.C.); (L.Z.); (J.C.); (C.L.)
| | - Jie Chen
- National Key Laboratory of Medical Immunology and Institute of Immunology, Naval Medical University, Shanghai 200433, China; (L.C.); (L.Z.); (J.C.); (C.L.)
| | - Chunzhen Li
- National Key Laboratory of Medical Immunology and Institute of Immunology, Naval Medical University, Shanghai 200433, China; (L.C.); (L.Z.); (J.C.); (C.L.)
| | - Yizhi Yu
- National Key Laboratory of Medical Immunology and Institute of Immunology, Naval Medical University, Shanghai 200433, China; (L.C.); (L.Z.); (J.C.); (C.L.)
| | - Sheng Xu
- National Key Laboratory of Medical Immunology and Institute of Immunology, Naval Medical University, Shanghai 200433, China; (L.C.); (L.Z.); (J.C.); (C.L.)
- Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai 200120, China
| |
Collapse
|
13
|
Tshering LF, Luo F, Russ S, Szenk M, Rubel D, Tutuska K, Rail JG, Balázsi G, Shen MM, Talos F. Immune mechanisms shape the clonal landscape during early progression of prostate cancer. Dev Cell 2023; 58:1071-1086.e8. [PMID: 37148881 PMCID: PMC10330387 DOI: 10.1016/j.devcel.2023.04.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Revised: 12/18/2022] [Accepted: 04/14/2023] [Indexed: 05/08/2023]
Abstract
Understanding the role of the immune microenvironment in modulating intratumor heterogeneity is essential for effective cancer therapies. Using multicolor lineage tracing in genetically engineered mouse models and single-cell transcriptomics, we show that slowly progressing tumors contain a multiclonal landscape of relatively homogeneous subpopulations within a well-organized tumor microenvironment. In more advanced and aggressive tumors, however, the multiclonal landscape develops into competing dominant and minor clones accompanied by a disordered microenvironment. We demonstrate that this dominant/minor landscape is associated with differential immunoediting, in which minor clones are marked by an increased expression of IFNγ-response genes and the T cell-activating chemokines Cxcl9 and Cxcl11. Furthermore, immunomodulation of the IFNγ pathway can rescue minor clones from elimination. Notably, the immune-specific gene signature of minor clones exhibits a prognostic value for biochemical recurrence-free survival in human prostate cancer. These findings suggest new immunotherapy approaches for modulating clonal fitness and tumor progression in prostate cancer.
Collapse
Affiliation(s)
- Lara F Tshering
- Department of Urology, Stony Brook University, Stony Brook, NY 11794, USA; Department of Pathology, Stony Brook University, Stony Brook, NY 11794, USA
| | - Fu Luo
- Department of Urology, Stony Brook University, Stony Brook, NY 11794, USA; Department of Pathology, Stony Brook University, Stony Brook, NY 11794, USA
| | - Savanah Russ
- Department of Urology, Stony Brook University, Stony Brook, NY 11794, USA; Department of Pathology, Stony Brook University, Stony Brook, NY 11794, USA
| | - Mariola Szenk
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY 11794, USA; Laufer Center for Physical and Quantitative Biology, Stony Brook University, Stony Brook, NY 11794, USA
| | - Diana Rubel
- Department of Urology, Stony Brook University, Stony Brook, NY 11794, USA; Department of Pathology, Stony Brook University, Stony Brook, NY 11794, USA
| | - Karis Tutuska
- Department of Urology, Stony Brook University, Stony Brook, NY 11794, USA; Department of Pathology, Stony Brook University, Stony Brook, NY 11794, USA
| | - James G Rail
- Department of Urology, Stony Brook University, Stony Brook, NY 11794, USA; Department of Pathology, Stony Brook University, Stony Brook, NY 11794, USA
| | - Gábor Balázsi
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY 11794, USA; Laufer Center for Physical and Quantitative Biology, Stony Brook University, Stony Brook, NY 11794, USA
| | - Michael M Shen
- Departments of Medicine, Genetics & Development, Urology, and Systems Biology, Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY 10032, USA.
| | - Flaminia Talos
- Department of Urology, Stony Brook University, Stony Brook, NY 11794, USA; Department of Pathology, Stony Brook University, Stony Brook, NY 11794, USA; Departments of Medicine, Genetics & Development, Urology, and Systems Biology, Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY 10032, USA; Stony Brook Cancer Center, Stony Brook University, Stony Brook, NY 11794, USA.
| |
Collapse
|
14
|
Xue S, Su XM, Ke LN, Huang YG. CXCL9 correlates with antitumor immunity and is predictive of a favorable prognosis in uterine corpus endometrial carcinoma. Front Oncol 2023; 13:1077780. [PMID: 36845675 PMCID: PMC9945585 DOI: 10.3389/fonc.2023.1077780] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Accepted: 01/17/2023] [Indexed: 02/11/2023] Open
Abstract
Background The C-X-C motif chemokine ligand-9 (CXCL9) is related to the progression of multiple neoplasms. Yet, its biological functions in uterine corpus endometrioid carcinoma (UCEC) remain shrouded in confusion. Here, we assessed the prognostic significance and potential mechanism of CXCL9 in UCEC. Methods Firstly, bioinformatics analysis of the public cancer database, including the Cancer Genome Atlas / the Genotype-Tissue Expression project (TCGA+ GTEx, n=552) and Gene Expression Omnibus (GEO): GSE63678 (n=7), were utilized for the CXCL9 expression-related analysis in UCEC. Then, the survival analysis of TCGA-UCEC was performed. Futher, the gene set enrichment analysis (GSEA) was carried out to reveal the potential molecular signaling pathway in UCEC associated with CXCL9 expression. Moreover, the immunohistochemistry (IHC) assay of our validation cohort (n=124) from human specimens were used to demonstrate the latent significance of CXCL9 in UCEC. Results The bioinformatics analysis suggested that CXCL9 expression was significantly upregulated in UCEC patients; and hyper-expression of CXCL9 was related to prolonged survival. the GSEA enrichment analysis showed various immune response-related pathways, including T/NK cell, lymphocyte activation, cytokine-cytokine receptor interaction network, and chemokine signaling pathway, mediated by CXCL9. In addition, the cytotoxic molecules (IFNG, SLAMF7, JCHAIN, NKG7, GBP5, LYZ, GZMA, GZMB, and TNF3F9) and the immunosuppressive genes (including PD-L1) were positively related to the expression of CXCL9. Further, the IHC assay indicated that the CXCL9 protein expression was mainly located in intertumoral and significantly upregulated in the UCEC patients; UCEC with high intertumoral CXCL9 cell abundance harbored an improved prognosis; a higher ratio of anti-tumor immune cells (CD4+, CD8+, and CD56+ cell) and PD-L1 was found in UCEC with CXCL9 high expression. Conclusion Overexpressed CXCL9 correlates with antitumor immunity and is predictive of a favorable prognosis in UCEC. It hinted that CXCL9 may serve as an independent prognostic biomarker or therapeutic target in UCEC patients, which augmented anti-tumor immune effects to furnish survival benefits.
Collapse
Affiliation(s)
- Shen Xue
- Department of obstetrics and gynecology, Sinopharm Dongfeng General Hospital, Hubei University of Medicine, Shiyan, China
| | - Xiao-min Su
- Department of Immunology, Nankai University School of Medicine, Tianjin, China
| | - Li-na Ke
- Department of obstetrics and gynecology, Sinopharm Dongfeng General Hospital, Hubei University of Medicine, Shiyan, China,*Correspondence: Yu-gang Huang, ; Li-na Ke,
| | - Yu-gang Huang
- Department of Pathology, Taihe Hospital, Hubei University of Medicine, Shiyan, China,*Correspondence: Yu-gang Huang, ; Li-na Ke,
| |
Collapse
|
15
|
Dey M, Kim MH, Nagamine M, Karhan E, Kozhaya L, Dogan M, Unutmaz D, Ozbolat IT. Biofabrication of 3D breast cancer models for dissecting the cytotoxic response of human T cells expressing engineered MAIT cell receptors. Biofabrication 2022; 14:10.1088/1758-5090/ac925a. [PMID: 36108605 PMCID: PMC9556424 DOI: 10.1088/1758-5090/ac925a] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 09/15/2022] [Indexed: 11/12/2022]
Abstract
Immunotherapy has revolutionized cancer treatment with the advent of advanced cell engineering techniques aimed at targeted therapy with reduced systemic toxicity. However, understanding the underlying immune-cancer interactions require development of advanced three-dimensional (3D) models of human tissues. In this study, we fabricated 3D tumor models with increasing complexity to study the cytotoxic responses of CD8+T cells, genetically engineered to express mucosal-associated invariant T (MAIT) cell receptors, towards MDA-MB-231 breast cancer cells. Homotypic MDA-MB-231 and heterotypic MDA-MB-231/human dermal fibroblast tumor spheroids were primed with precursor MAIT cell ligand 5-amino-6-D-ribitylaminouracil (5-ARU). Engineered T cells effectively eliminated tumors after a 3 d culture period, demonstrating that the engineered T cell receptor recognized major histocompatibility complex class I-related (MR1) protein expressing tumor cells in the presence of 5-ARU. Tumor cell killing efficiency of engineered T cells were also assessed by encapsulating these cells in fibrin, mimicking a tumor extracellular matrix microenvironment. Expression of proinflammatory cytokines such as interferon gamma, interleukin-13, CCL-3 indicated immune cell activation in all tumor models, post immunotherapy. Further, in corroborating the cytotoxic activity, we found that granzymes A and B were also upregulated, in homotypic as well as heterotypic tumors. Finally, a 3D bioprinted tumor model was employed to study the effect of localization of T cells with respect to tumors. T cells bioprinted proximal to the tumor had reduced invasion index and increased cytokine secretion, which indicated a paracrine mode of immune-cancer interaction. Development of 3D tumor-T cell platforms may enable studying the complex immune-cancer interactions and engineering MAIT cells for cell-based cancer immunotherapies.
Collapse
Affiliation(s)
- Madhuri Dey
- Department of Chemistry, Penn State University; University Park, PA, 16802, USA
- The Huck Institutes of the Life Sciences, Penn State University; University Park, PA 16802, USA
| | - Myong Hwan Kim
- The Huck Institutes of the Life Sciences, Penn State University; University Park, PA 16802, USA
- Biomedical Engineering Department, Penn State University; University Park, PA 16802, USA
| | - Momoka Nagamine
- Department of Chemistry, Penn State University; University Park, PA, 16802, USA
- The Huck Institutes of the Life Sciences, Penn State University; University Park, PA 16802, USA
| | - Ece Karhan
- The Jackson Laboratory for Genomic Medicine; Farmington, CT 06032, USA
| | - Lina Kozhaya
- The Jackson Laboratory for Genomic Medicine; Farmington, CT 06032, USA
| | - Mikail Dogan
- The Jackson Laboratory for Genomic Medicine; Farmington, CT 06032, USA
| | - Derya Unutmaz
- The Jackson Laboratory for Genomic Medicine; Farmington, CT 06032, USA
- University of Connecticut Health Center; Farmington, CT 06032, USA
| | - Ibrahim T. Ozbolat
- The Huck Institutes of the Life Sciences, Penn State University; University Park, PA 16802, USA
- Biomedical Engineering Department, Penn State University; University Park, PA 16802, USA
- Engineering Science and Mechanics Department, Penn State University; University Park, PA 16802, USA
- Materials Research Institute, Penn State University; University Park, PA 16802, USA
- Department of Neurosurgery, Penn State College of Medicine, Hershey, PA 17033, USA
- Penn State Cancer Institute, Penn State University, Hershey, PA 17033, USA
| |
Collapse
|
16
|
Landeira-Viñuela A, Arias-Hidalgo C, Juanes-Velasco P, Alcoceba M, Navarro-Bailón A, Pedreira CE, Lecrevisse Q, Díaz-Muñoz L, Sánchez-Santos JM, Hernández ÁP, García-Vaquero ML, Góngora R, De Las Rivas J, González M, Orfao A, Fuentes M. Unravelling soluble immune checkpoints in chronic lymphocytic leukemia: Physiological immunomodulators or immune dysfunction. Front Immunol 2022; 13:965905. [PMID: 36248816 PMCID: PMC9554405 DOI: 10.3389/fimmu.2022.965905] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 08/24/2022] [Indexed: 11/13/2022] Open
Abstract
Chronic lymphocytic leukemia (CLL) is a lymphoid neoplasm characterized by the accumulation of mature B cells. The diagnosis is established by the detection of monoclonal B lymphocytes in peripheral blood, even in early stages [monoclonal B-cell lymphocytosis (MBLhi)], and its clinical course is highly heterogeneous. In fact, there are well-characterized multiple prognostic factors that are also related to the observed genetic heterogenicity, such as immunoglobulin heavy chain variable region (IGHV) mutational status, del17p, and TP53 mutations, among others. Moreover, a dysregulation of the immune system (innate and adaptive immunity) has been observed in CLL patients, with strong impact on immune surveillance and consequently on the onset, evolution, and therapy response. In addition, the tumor microenvironment is highly complex and heterogeneous (i.e., matrix, fibroblast, endothelial cells, and immune cells), playing a critical role in the evolution of CLL. In this study, a quantitative profile of 103 proteins (cytokines, chemokines, growth/regulatory factors, immune checkpoints, and soluble receptors) in 67 serum samples (57 CLL and 10 MBLhi) has been systematically evaluated. Also, differential profiles of soluble immune factors that discriminate between MBLhi and CLL (sCD47, sCD27, sTIMD-4, sIL-2R, and sULBP-1), disease progression (sCD48, sCD27, sArginase-1, sLAG-3, IL-4, and sIL-2R), or among profiles correlated with other prognostic factors, such as IGHV mutational status (CXCL11/I-TAC, CXCL10/IP-10, sHEVM, and sLAG-3), were deciphered. These results pave the way to explore the role of soluble immune checkpoints as a promising source of biomarkers in CLL, to provide novel insights into the immune suppression process and/or dysfunction, mostly on T cells, in combination with cellular balance disruption and microenvironment polarization leading to tumor escape.
Collapse
Affiliation(s)
- Alicia Landeira-Viñuela
- Department of Medicine and General Service of Cytometry, Centro de Investigación Biomédica en Red Cáncer (CIBERONC)- CB16/12/00400, Cancer Research Centre-Instituto Universitario de Biología Molecular y Celular del Cáncer (IBMCC), Consejo Superior de Investigaciones Científicas - Universidad de Salamanca (CSIC-USAL), Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain
| | - Carlota Arias-Hidalgo
- Department of Medicine and General Service of Cytometry, Centro de Investigación Biomédica en Red Cáncer (CIBERONC)- CB16/12/00400, Cancer Research Centre-Instituto Universitario de Biología Molecular y Celular del Cáncer (IBMCC), Consejo Superior de Investigaciones Científicas - Universidad de Salamanca (CSIC-USAL), Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain
| | - Pablo Juanes-Velasco
- Department of Medicine and General Service of Cytometry, Centro de Investigación Biomédica en Red Cáncer (CIBERONC)- CB16/12/00400, Cancer Research Centre-Instituto Universitario de Biología Molecular y Celular del Cáncer (IBMCC), Consejo Superior de Investigaciones Científicas - Universidad de Salamanca (CSIC-USAL), Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain
| | - Miguel Alcoceba
- Department of Hematology, University Hospital of Salamanca, Centro de Investigación Biomédica en Red Cáncer (CIBERONC)- CB16/12/00233, Center Research-Centre Instituto Universitario de Biología Molecular y Celular del Cáncer (IBMCC) Consejo Superior de Investigaciones Científicas - Universidad de Salamanca, Instituto de Investigación Biomédica de Salamanca (CSIC-USAL, IBSAL), Salamanca, Spain
| | - Almudena Navarro-Bailón
- Department of Hematology, University Hospital of Salamanca, Centro de Investigación Biomédica en Red Cáncer (CIBERONC)- CB16/12/00233, Center Research-Centre Instituto Universitario de Biología Molecular y Celular del Cáncer (IBMCC) Consejo Superior de Investigaciones Científicas - Universidad de Salamanca, Instituto de Investigación Biomédica de Salamanca (CSIC-USAL, IBSAL), Salamanca, Spain
| | - Carlos Eduardo Pedreira
- Systems and Computing Department Instituto Alberto Luiz Coimbra de Pós-Graduação e Pesquisa de Engenharia-Programa de Engenharia de Sistemas e Computação (COPPE-PESC), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| | - Quentin Lecrevisse
- Department of Medicine and General Service of Cytometry, Centro de Investigación Biomédica en Red Cáncer (CIBERONC)- CB16/12/00400, Cancer Research Centre-Instituto Universitario de Biología Molecular y Celular del Cáncer (IBMCC), Consejo Superior de Investigaciones Científicas - Universidad de Salamanca (CSIC-USAL), Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain
| | - Laura Díaz-Muñoz
- Department of Medicine and General Service of Cytometry, Centro de Investigación Biomédica en Red Cáncer (CIBERONC)- CB16/12/00400, Cancer Research Centre-Instituto Universitario de Biología Molecular y Celular del Cáncer (IBMCC), Consejo Superior de Investigaciones Científicas - Universidad de Salamanca (CSIC-USAL), Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain
| | | | - Ángela-Patricia Hernández
- Department of Medicine and General Service of Cytometry, Centro de Investigación Biomédica en Red Cáncer (CIBERONC)- CB16/12/00400, Cancer Research Centre-Instituto Universitario de Biología Molecular y Celular del Cáncer (IBMCC), Consejo Superior de Investigaciones Científicas - Universidad de Salamanca (CSIC-USAL), Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain
- Department of Pharmaceutical Sciences, Organic Chemistry Section, Faculty of Pharmacy, University of Salamanca, Salamanca, Spain
| | - Marina L. García-Vaquero
- Department of Medicine and General Service of Cytometry, Centro de Investigación Biomédica en Red Cáncer (CIBERONC)- CB16/12/00400, Cancer Research Centre-Instituto Universitario de Biología Molecular y Celular del Cáncer (IBMCC), Consejo Superior de Investigaciones Científicas - Universidad de Salamanca (CSIC-USAL), Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain
| | - Rafael Góngora
- Department of Medicine and General Service of Cytometry, Centro de Investigación Biomédica en Red Cáncer (CIBERONC)- CB16/12/00400, Cancer Research Centre-Instituto Universitario de Biología Molecular y Celular del Cáncer (IBMCC), Consejo Superior de Investigaciones Científicas - Universidad de Salamanca (CSIC-USAL), Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain
| | - Javier De Las Rivas
- Bioinformatics and Functional Genomics Group, Cancer Research Center Instituto Universitario de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas/Universidad de Salamanca (CIC-IBMCC, CSIC/USAL), Consejo Superior de Investigaciones Científicas (CSIC) and University of Salamanca (USAL), Salamanca, Spain
| | - Marcos González
- Department of Hematology, University Hospital of Salamanca, Centro de Investigación Biomédica en Red Cáncer (CIBERONC)- CB16/12/00233, Center Research-Centre Instituto Universitario de Biología Molecular y Celular del Cáncer (IBMCC) Consejo Superior de Investigaciones Científicas - Universidad de Salamanca, Instituto de Investigación Biomédica de Salamanca (CSIC-USAL, IBSAL), Salamanca, Spain
| | - Alberto Orfao
- Department of Medicine and General Service of Cytometry, Centro de Investigación Biomédica en Red Cáncer (CIBERONC)- CB16/12/00400, Cancer Research Centre-Instituto Universitario de Biología Molecular y Celular del Cáncer (IBMCC), Consejo Superior de Investigaciones Científicas - Universidad de Salamanca (CSIC-USAL), Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain
| | - Manuel Fuentes
- Department of Medicine and General Service of Cytometry, Centro de Investigación Biomédica en Red Cáncer (CIBERONC)- CB16/12/00400, Cancer Research Centre-Instituto Universitario de Biología Molecular y Celular del Cáncer (IBMCC), Consejo Superior de Investigaciones Científicas - Universidad de Salamanca (CSIC-USAL), Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain
- Proteomics Unit, Cancer Research Centre-IBMCC, Instituto de Investigación Biomédica de Salamanca (IBSAL), University of Salamanca-Consejo Superior de Investigaciones Científicas (CSIC), Salamanca, Spain
- *Correspondence: Manuel Fuentes,
| |
Collapse
|
17
|
Jiang VC, Hao D, Jain P, Li Y, Cai Q, Yao Y, Nie L, Liu Y, Jin J, Wang W, Lee HH, Che Y, Dai E, Han G, Wang R, Rai K, Futreal A, Flowers C, Wang L, Wang M. TIGIT is the central player in T-cell suppression associated with CAR T-cell relapse in mantle cell lymphoma. Mol Cancer 2022; 21:185. [PMID: 36163179 PMCID: PMC9513944 DOI: 10.1186/s12943-022-01655-0] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 09/15/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Chimeric antigen receptor (CAR) T-cell therapy using brexucabtagene autoleucel (BA) induces remission in many patients with mantle cell lymphoma (MCL), and BA is the only CAR T-cell therapy approved by the FDA for MCL. However, development of relapses to BA is recognized with poor patient outcomes. Multiple CAR T-cell therapies have been approved for other lymphomas and the resistance mechanisms have been investigated. However, the mechanisms underlying BA relapse in MCL have not been investigated and whether any previously reported resistance mechanisms apply to BA-relapsed patients with MCL is unknown. METHODS To interrogate BA resistance mechanisms in MCL, we performed single-cell RNA sequencing on 39 longitudinally collected samples from 15 BA-treated patients, and multiplex cytokine profiling on 80 serial samples from 20 patients. RESULTS We demonstrate that after BA relapse, the proportion of T cells, especially cytotoxic T cells (CTLs), decreased among non-tumor cells, while the proportion of myeloid cells correspondingly increased. TIGIT, LAG3, and CD96 were the predominant checkpoint molecules expressed on exhausted T cells and CTLs; only TIGIT was significantly increased after relapse. CTLs expanded during remission, and then contracted during relapse with upregulated TIGIT expression. Tumor cells also acquired TIGIT expression after relapse, leading to the enhanced interaction of tumor cell TIGIT with monocyte CD155/PVR. In myeloid cells, post-relapse HLA-II expression was reduced relative to pretreatment and during remission. Myeloid-derived suppressor cells (MDSCs) were enriched after relapse with elevated expression of activation markers, including CLU (clusterin) and VCAN (versican). Extracellular chemokines (CCL4, CXCL9, CXCL13), soluble checkpoint inhibitors (sPD-L1, sTIM3, s4-1BB), and soluble receptors (sIL-2R, sTNFRII) were decreased during remission but elevated after relapse. CONCLUSIONS Our data demonstrate that multiple tumor-intrinsic and -extrinsic factors are associated with T-cell suppression and BA relapse. Among these, TIGIT appears to be the central player given its elevated expression after BA relapse in not only CTLs but also MCL cells. The acquisition of TIGIT expression on tumor cells is MCL-specific and has not been reported in other CAR T-treated diseases. Together, our data suggest that co-targeting TIGIT may prevent CAR T relapses and thus promote long-term progression-free survival in MCL patients.
Collapse
Affiliation(s)
- Vivian Changying Jiang
- Department of Lymphoma and Myeloma, the University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Dapeng Hao
- Department of Genomic Medicine, the University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Preetesh Jain
- Department of Lymphoma and Myeloma, the University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Yijing Li
- Department of Lymphoma and Myeloma, the University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Qingsong Cai
- Department of Lymphoma and Myeloma, the University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Yixin Yao
- Department of Lymphoma and Myeloma, the University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Lei Nie
- Department of Lymphoma and Myeloma, the University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Yang Liu
- Department of Lymphoma and Myeloma, the University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Jingling Jin
- Department of Lymphoma and Myeloma, the University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Wei Wang
- Department of Lymphoma and Myeloma, the University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Heng-Huan Lee
- Department of Lymphoma and Myeloma, the University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Yuxuan Che
- Department of Lymphoma and Myeloma, the University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Enyu Dai
- Department of Genomic Medicine, the University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Guangchun Han
- Department of Genomic Medicine, the University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Ruiping Wang
- Department of Genomic Medicine, the University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Kunal Rai
- Department of Genomic Medicine, the University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Andrew Futreal
- Department of Genomic Medicine, the University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Christopher Flowers
- Department of Lymphoma and Myeloma, the University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Linghua Wang
- Department of Genomic Medicine, the University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA. .,The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences (GSBS), Houston, TX, 77030, USA.
| | - Michael Wang
- Department of Lymphoma and Myeloma, the University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA. .,Department of Stem Cell Transplantation and Cellular Therapy, the University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
| |
Collapse
|
18
|
Chen R, Wu W, Liu T, Zhao Y, Wang Y, Zhang H, Wang Z, Dai Z, Zhou X, Luo P, Zhang J, Liu Z, Zhang LY, Cheng Q. Large-scale bulk RNA-seq analysis defines immune evasion mechanism related to mast cell in gliomas. Front Immunol 2022; 13:914001. [PMID: 36159780 PMCID: PMC9492887 DOI: 10.3389/fimmu.2022.914001] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 07/14/2022] [Indexed: 11/13/2022] Open
Abstract
Accumulating evidence has demonstrated that the immune cells have an emerging role in controlling anti-tumor immune responses and tumor progression. The comprehensive role of mast cell in glioma has not been illustrated yet. In this study, 1,991 diffuse glioma samples were collected from The Cancer Genome Atlas (TCGA) and the Chinese Glioma Genome Atlas (CGGA). xCell algorithm was employed to define the mast cell-related genes. Based on mast cell-related genes, gliomas were divided into two clusters with distinct clinical and immunological characteristics. The survival probability of cluster 1 was significantly lower than that of cluster 2 in the TCGA dataset, three CGGA datasets, and the Xiangya cohort. Meanwhile, the hypoxic and metabolic pathways were active in cluster 1, which were beneficial to the proliferation of tumor cells. A potent prognostic model based on mast cell was constructed. Via machine learning, DRG2 was screened out as a characteristic gene, which was demonstrated to predict treatment response and predict survival outcome in the Xiangya cohort. In conclusion, mast cells could be used as a potential effective prognostic factor for gliomas.
Collapse
Affiliation(s)
- Rui Chen
- Department of Neurosurgery, Affiliated Nanhua Hospital, University of South China, Hengyang, China
| | - Wantao Wu
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
| | - Tao Liu
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Yihan Zhao
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Yifan Wang
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Hao Zhang
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
| | - Zeyu Wang
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
| | - Ziyu Dai
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
| | - Xiaoxi Zhou
- Department of Neurosurgery, Affiliated Nanhua Hospital, University of South China, Hengyang, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Peng Luo
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Jian Zhang
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Zaoqu Liu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou, Zhengzhou, China
| | - Li-Yang Zhang
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Xiangya School of Medicine, Central South University, Changsha, China
- *Correspondence: Quan Cheng, ; Liyang Zhang,
| | - Quan Cheng
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Xiangya School of Medicine, Central South University, Changsha, China
- *Correspondence: Quan Cheng, ; Liyang Zhang,
| |
Collapse
|
19
|
Zhang Y, Liu Z, Wei W, Li Y. TCR engineered T cells for solid tumor immunotherapy. Exp Hematol Oncol 2022; 11:38. [PMID: 35725570 PMCID: PMC9210724 DOI: 10.1186/s40164-022-00291-0] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 06/09/2022] [Indexed: 11/26/2022] Open
Abstract
T cell immunotherapy remains an attractive approach for cancer immunotherapy. T cell immunotherapy mainly employs chimeric antigen receptor (CAR)- and T cell receptor (TCR)-engineered T cells. CAR-T cell therapy has been an essential breakthrough in treating hematological malignancies. TCR-T cells can recognize antigens expressed both on cell surfaces and in intracellular compartments. Although TCR-T cells have not been approved for clinical application, a number of clinical trials have been performed, particularly for solid tumors. In this article, we summarized current TCR-T cell advances and their potential advantages for solid tumor immunotherapy.
Collapse
Affiliation(s)
- Yikai Zhang
- Guangzhou Municipality Tianhe Nuoya Bio-engineering Co. Ltd, Guangzhou, 510663, China.,Guangdong Cord blood bank, Guangzhou, 510663, China.,Department of Hematology, First Affiliated Hospital, Jinan University, Guangzhou, 510632, China.,Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, School of Medicine, Jinan University, 601 Huang Pu Da Dao Xi, Guangzhou, 510632, China
| | - Zhipeng Liu
- Guangzhou Municipality Tianhe Nuoya Bio-engineering Co. Ltd, Guangzhou, 510663, China.,Guangdong Cord blood bank, Guangzhou, 510663, China
| | - Wei Wei
- Guangzhou Municipality Tianhe Nuoya Bio-engineering Co. Ltd, Guangzhou, 510663, China. .,Guangdong Cord blood bank, Guangzhou, 510663, China.
| | - Yangqiu Li
- Department of Hematology, First Affiliated Hospital, Jinan University, Guangzhou, 510632, China. .,Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, School of Medicine, Jinan University, 601 Huang Pu Da Dao Xi, Guangzhou, 510632, China.
| |
Collapse
|
20
|
Li H, Wu M, Zhao X. Role of chemokine systems in cancer and inflammatory diseases. MedComm (Beijing) 2022; 3:e147. [PMID: 35702353 PMCID: PMC9175564 DOI: 10.1002/mco2.147] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 05/10/2022] [Accepted: 05/11/2022] [Indexed: 12/12/2022] Open
Abstract
Chemokines are a large family of small secreted proteins that have fundamental roles in organ development, normal physiology, and immune responses upon binding to their corresponding receptors. The primary functions of chemokines are to coordinate and recruit immune cells to and from tissues and to participate in regulating interactions between immune cells. In addition to the generally recognized antimicrobial immunity, the chemokine/chemokine receptor axis also exerts a tumorigenic function in many different cancer models and is involved in the formation of immunosuppressive and protective tumor microenvironment (TME), making them potential prognostic markers for various hematologic and solid tumors. In fact, apart from its vital role in tumors, almost all inflammatory diseases involve chemokines and their receptors in one way or another. Modulating the expression of chemokines and/or their corresponding receptors on tumor cells or immune cells provides the basis for the exploitation of new drugs for clinical evaluation in the treatment of related diseases. Here, we summarize recent advances of chemokine systems in protumor and antitumor immune responses and discuss the prevailing understanding of how the chemokine system operates in inflammatory diseases. In this review, we also emphatically highlight the complexity of the chemokine system and explore its potential to guide the treatment of cancer and inflammatory diseases.
Collapse
Affiliation(s)
- Hongyi Li
- Department of Gynecology and Obstetrics, Development and Related Disease of Women and Children Key Laboratory of Sichuan Province, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of EducationWest China Second HospitalSichuan UniversityChengduChina
| | - Min Wu
- Department of Biomedical Sciences, School of Medicine and Health SciencesUniversity of North DakotaGrand ForksNorth DakotaUSA
| | - Xia Zhao
- Department of Gynecology and Obstetrics, Development and Related Disease of Women and Children Key Laboratory of Sichuan Province, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of EducationWest China Second HospitalSichuan UniversityChengduChina
| |
Collapse
|
21
|
Pham NB, Abraham N, Velankar KY, Schueller NR, Philip EJ, Jaber Y, Gawalt ES, Fan Y, Pal SK, Meng WS. Localized PD-1 Blockade in a Mouse Model of Renal Cell Carcinoma. FRONTIERS IN DRUG DELIVERY 2022; 2. [PMID: 36132332 PMCID: PMC9486680 DOI: 10.3389/fddev.2022.838458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Herein we report the impact of localized delivery of an anti-mouse PD-1-specific monoclonal antibody (aPD1) on Renca tumors in the resulting T cell responses and changes in broader immune gene expression profiles. Renca is a BALB/c mice syngeneic tumor that has been used to model human renal cell carcinoma In this study, T cell subsets were examined in tumors and draining lymph nodes of mice treated with localized PD-1 with and without the addition of adenosine deaminase (ADA), an enzyme that catabolizes adenosine (ADO), identified as an immune checkpoint in several types of human cancers. The biologics, aPD1, or aPD1 with adenosine deaminase (aPD1/ADA), were formulated with the self-assembling peptides Z15_EAK to enhance retention near the tumor inoculation site. We found that both aPD1 and aPD1/ADA skewed the local immune milieu towards an immune stimulatory phenotype by reducing Tregs, increasing CD8 T cell infiltration, and upregulating IFNɣ. Analysis of tumor specimens using bulk RNA-Seq confirmed the impact of the localized aPD1 treatment and revealed differential gene expressions elicited by the loco-regional treatment. The effects of ADA and Z15_EAK were limited to tumor growth delay and lymph node enlargement. These results support the notion of expanding the use of locoregional PD-1 blockade in solid tumors.
Collapse
Affiliation(s)
- Ngoc B. Pham
- Graduate School of Pharmaceutical Sciences, Duquesne University, Pittsburgh, PA, United States
| | - Nevil Abraham
- Graduate School of Pharmaceutical Sciences, Duquesne University, Pittsburgh, PA, United States
| | - Ketki Y. Velankar
- Graduate School of Pharmaceutical Sciences, Duquesne University, Pittsburgh, PA, United States
| | - Nathan R. Schueller
- Graduate School of Pharmaceutical Sciences, Duquesne University, Pittsburgh, PA, United States
| | - Errol J. Philip
- School of Medicine, University of California, San Francisco, San Francisco, CA, United States
| | - Yasmeen Jaber
- Department of Medical Oncology and Developmental Therapeutics, City of Hope Comprehensive Cancer Center, Duarte, CA, United States
| | - Ellen S. Gawalt
- Department of Chemistry and Biochemistry, Duquesne University, Pittsburgh, PA, United States
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Yong Fan
- Institute of Cellular Therapeutics, Allegheny-Singer Research Institute, Pittsburgh, PA, United States
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA, United States
| | - Sumanta K. Pal
- Department of Medical Oncology and Developmental Therapeutics, City of Hope Comprehensive Cancer Center, Duarte, CA, United States
| | - Wilson S. Meng
- Graduate School of Pharmaceutical Sciences, Duquesne University, Pittsburgh, PA, United States
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, United States
- Correspondence: Wilson S. Meng,
| |
Collapse
|
22
|
Wu Z, Wan J, Wang J, Meng X, Qian H. Identification of prognostic biomarkers for breast cancer brain metastases based on the bioinformatics analysis. Biochem Biophys Rep 2022; 29:101203. [PMID: 35059509 PMCID: PMC8760394 DOI: 10.1016/j.bbrep.2022.101203] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 01/05/2022] [Accepted: 01/05/2022] [Indexed: 12/12/2022] Open
Abstract
Purpose The prognosis of breast cancer (BC) patients who develop into brain metastases (BMs) is very poor. Thus, it is of great significance to explore the etiology of BMs in BC and identify the key genes involved in this process to improve the survival of BC patients with BMs. Patients and methods The gene expression data and the clinical information of BC patients were downloaded from TCGA and GEO database. Differentially expressed genes (DEGs) in TCGA-BRCA and GSE12276 were overlapped to find differentially expressed metastatic genes (DEMGs). The protein-protein interaction (PPI) network of DEMGs was constructed via STRING database. ClusterProfiler R package was applied to perform the gene ontology (GO) enrichment analysis of DEMGs. The univariate Cox regression analysis and the Kaplan-Meier (K-M) curves were plotted to screen DEMGs associated with the overall survival and the metastatic recurrence survival, which were identified as the key genes associated with the BMs in BC. The immune infiltration and the expressions of immune checkpoints for BC patients with brain relapses and BC patients with other relapses were analyzed respectively. The correlations among the expressions of key genes and the differently infiltrated immune cells or the differentially expressed immune checkpoints were calculated. The gene set enrichment analysis (GSEA) of each key gene was conducted to investigate the potential mechanisms of key genes involved in BC patients with BMs. Moreover, CTD database was used to predict the drug-gene interaction network of key genes. Results A total of 154 DEGs were identified in BC patients at M0 and M1 in TCGA database. A total of 667 DEGs were identified in BC patients with brain relapses and with other relapses. By overlapping these DEGs, 17 DEMGs were identified, which were enriched in the cell proliferation related biological processes and the immune related molecular functions. The univariate Cox regression analysis and the Kaplan-Meier curves revealed that CXCL9 and GPR171 were closely associated with the overall survival and the metastatic recurrence survival and were identified as key genes associated with BMs in BC. The analyses of immune infiltration and immune checkpoint expressions showed that there was a significant difference of the immune microenvironment between brain relapses and other relapses in BC. GSEA indicated that CXCL9 and GPR171 may regulate BMs in BC via the immune-related pathways. Conclusion Our study identified the key genes associated with BMs in BC patients and explore the underlying mechanisms involved in the etiology of BMs in BC. These findings may provide a promising approach for the treatments of BC patients with BMs. CXCL9 and GPR171, as the key genes, were closely associated with the prognosis of brain metastases in breast cancer. There was a significant difference of the immune microenvironment between brain and other metastases in breast cancer. We revealed candidate drugs which associated with the key genes of breast cancer patients with brain metastases. A series of bioinformatic analysis methods were used in this article.
Collapse
Affiliation(s)
| | - Jinghai Wan
- Corresponding author. National Cancer Center/ National Clinical Research Center for Cancer/ Cancer Hospital, Neurosurgery, Panjiayuan 17#, Chaoyang District, Beijing, China.
| | | | | | | |
Collapse
|
23
|
Powles T, Sridhar SS, Loriot Y, Bellmunt J, Mu XJ, Ching KA, Pu J, Sternberg CN, Petrylak DP, Tambaro R, Dourthe LM, Alvarez-Fernandez C, Aarts M, di Pietro A, Grivas P, Davis CB. Avelumab maintenance in advanced urothelial carcinoma: biomarker analysis of the phase 3 JAVELIN Bladder 100 trial. Nat Med 2021; 27:2200-2211. [PMID: 34893775 DOI: 10.1038/s41591-021-01579-0] [Citation(s) in RCA: 80] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 10/13/2021] [Indexed: 12/20/2022]
Abstract
In a recent phase 3 randomized trial of 700 patients with advanced urothelial cancer (JAVELIN Bladder 100; NCT02603432 ), avelumab/best supportive care (BSC) significantly prolonged overall survival relative to BSC alone as maintenance therapy after first-line chemotherapy. Exploratory biomarker analyses were performed to identify biological pathways that might affect survival benefit. Tumor molecular profiling by immunohistochemistry, whole-exome sequencing and whole-transcriptome sequencing revealed that avelumab survival benefit was positively associated with PD-L1 expression by tumor cells, tumor mutational burden, APOBEC mutation signatures, expression of genes underlying innate and adaptive immune activity and the number of alleles encoding high-affinity variants of activating Fcγ receptors. Pathways connected to tissue growth and angiogenesis might have been associated with reduced survival benefit. Individual biomarkers did not comprehensively identify patients who could benefit from therapy; however, multi-parameter models incorporating genomic alteration, immune responses and tumor growth showed promising predictive utility. These results characterize the complex biologic pathways underlying survival benefit from immune checkpoint inhibition in advanced urothelial cancer and suggest that multiple biomarkers might be needed to identify patients who would benefit from treatment.
Collapse
Affiliation(s)
- Thomas Powles
- Barts Cancer Institute, Experimental Cancer Medicine Centre, Queen Mary University of London, St. Bartholomew's Hospital, London, UK.
| | - Srikala S Sridhar
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Yohann Loriot
- Gustave Roussy, INSERMU981, Université Paris-Saclay, Villejuif, France
| | - Joaquim Bellmunt
- Department of Medical Oncology, Beth Israel Deaconess Medical Center and IMIM-PSMAR Lab, Harvard Medical School, Boston, MA, USA
| | - Xinmeng Jasmine Mu
- Computational Biology, Oncology Research and Development, Pfizer, La Jolla, CA, USA
| | - Keith A Ching
- Computational Biology, Oncology Research and Development, Pfizer, La Jolla, CA, USA
| | - Jie Pu
- Statistics, Global Biometrics and Data Management, Pfizer, La Jolla, CA, USA
| | - Cora N Sternberg
- Englander Institute for Precision Medicine, Weill Cornell Medicine, Hematology/Oncology, Meyer Cancer Center, New York, NY, USA
| | | | - Rosa Tambaro
- Istituto Nazionale per lo Studio e la Cura dei Tumori, IRCCS Fondazione Giovanni Pascale, Naples, Italy
| | - Louis M Dourthe
- Service d'Oncologie Médicale, Clinique St Anne, Strasbourg, France
| | - Carlos Alvarez-Fernandez
- Department of Medical Oncology, Hospital Universitario Central de Asturias. Instituto de Investigación Sanitaria del Principado de Asturias, Oviedo, Spain
| | - Maureen Aarts
- Department of Medical Oncology, GROW School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, Netherlands
| | | | - Petros Grivas
- Department of Medicine, Division of Medical Oncology, University of Washington; Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle Cancer Care Alliance, Seattle, WA, USA
| | | |
Collapse
|
24
|
Yi J, Shuang Z, Zhong W, Wu H, Feng J, Zouxu X, Huang X, Li S, Wang X. Identification of Immune-Related Risk Characteristics and Prognostic Value of Immunophenotyping in TNBC. Front Genet 2021; 12:730442. [PMID: 34777466 PMCID: PMC8586457 DOI: 10.3389/fgene.2021.730442] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Accepted: 09/23/2021] [Indexed: 11/13/2022] Open
Abstract
Background: Triple-negative breast cancer (TNBC) is not sensitive to targeted therapy with HER-2 monoclonal antibody and endocrine therapy due to lack of ER, PR, and HER-2 receptors. TNBC is a breast cancer subtype with the worst prognosis and the highest mortality rate compared with other subtypes. Materials and Methods: Breast cancer-related data were retrieved from The Cancer Genome Atlas (TCGA) database, and 116 cases of triple-negative breast cancer were identified from the data. GSE31519 dataset was retrieved from Gene Expression Omnibus (GEO) database, comprising a total of 68 cases with TNBC. Survival analysis was performed based on immune score, infiltration score and mutation score to explore differences in prognosis of different immune types. Analysis of differentially expressed genes was conducted and GSEA analysis based on these genes was conducted to explore the potential mechanism. Results: The findings showed that comprehensive immune typing is highly effective and accurate in assessing prognosis of TNBC patients. Analysis showed that MMP9, CXCL9, CXCL10, CXCL11 and CD7 are key genes that may affect immune typing of TNBC patients and play an important role in prediction of prognosis in TNBC patients. Conclusion: The current study presents an evaluation system based on immunophenotyping, which provides a more accurate prognostic evaluation tool for TNBC patients. Differentially expressed genes can be targeted to improve treatment of TNBC.
Collapse
Affiliation(s)
- Jiarong Yi
- Department of Breast Oncology, Sun Yat-sen University Cancer Center, The State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Zeyu Shuang
- Department of Breast Oncology, Sun Yat-sen University Cancer Center, The State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Wenjing Zhong
- Department of Breast Oncology, Sun Yat-sen University Cancer Center, The State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Haoming Wu
- Department of Breast Oncology, Sun Yat-sen University Cancer Center, The State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Jikun Feng
- Department of Breast Oncology, Sun Yat-sen University Cancer Center, The State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Xiazi Zouxu
- Department of Breast Oncology, Sun Yat-sen University Cancer Center, The State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Xinjian Huang
- Department of Breast Oncology, Sun Yat-sen University Cancer Center, The State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Siqi Li
- Department of Breast Oncology, Sun Yat-sen University Cancer Center, The State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Xi Wang
- Department of Breast Oncology, Sun Yat-sen University Cancer Center, The State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| |
Collapse
|
25
|
Olkowska-Truchanowicz J, Białoszewska A, Zwierzchowska A, Sztokfisz-Ignasiak A, Janiuk I, Dąbrowski F, Korczak-Kowalska G, Barcz E, Bocian K, Malejczyk J. Peritoneal Fluid from Patients with Ovarian Endometriosis Displays Immunosuppressive Potential and Stimulates Th2 Response. Int J Mol Sci 2021; 22:ijms22158134. [PMID: 34360900 PMCID: PMC8347337 DOI: 10.3390/ijms22158134] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Revised: 07/26/2021] [Accepted: 07/27/2021] [Indexed: 11/16/2022] Open
Abstract
Endometriosis is a common gynaecological disorder characterized by the ectopic growth of endometrial tissue outside the uterine cavity. It is associated with chronic pelvic inflammation and autoimmune reactivity manifesting by autoantibody production and abrogated cellular immune responses. Endometriotic peritoneal fluid contains various infiltrating leucocyte populations and a bulk of proinflammatory and immunoregulatory cytokines. However, the nature and significance of the peritoneal milieu in women with endometriosis still remains obscure. Therefore, the aim of the present study was to investigate the immunoregulatory activity of the peritoneal fluid (PF) from women with endometriosis. The peritoneal fluid samples were collected during laparoscopic surgery from 30 women with and without endometriosis. Immunoregulatory cytokines (IL-2, IL-4, IL-6, IL-10, IL-17A, IFN-γ and TNF) and chemokines (CCL2, CCL5, CXCL8 and CXCL9) were evaluated in PF and culture supernatants generated by unstimulated and CD3/CD28/IL-2-stimulated CD4+ T cells cultured in the presence of PF. The effect of PF on the generation of Treg and Th17 cells in CD4+ T cell cultures, as well as the natural cytotoxic activity of peripheral blood mononuclear cells, was also investigated. Concentrations of IL-6, IL-10, CCL2, CXCL8 and CXCL9 were significantly upregulated in the PF from women with endometriosis when compared to control women, whereas concentrations of other cytokines and chemokines were unaffected. The culturing of unstimulated and CD3/CD28/IL-2-stimulated CD4+ T cells in the presence of endometriotic PF resulted in the downregulation of their IL-2, IFN-γ, IL-17A and TNF production as compared to culture medium alone. On the other side, endometriotic PF significantly stimulated the production of IL-4 and IL-10. Endometriotic PF also stimulated the release of CCL2 and CXCL8, whereas the production of CCL5 and CXCL9 was downregulated. Endometriotic PF stimulated the generation of Treg cells and had an inhibitory effect on the generation of Th17 cells in cultures of CD4+ T cells. It also inhibited the NK cell cytotoxic activity of the peripheral blood lymphocytes. These results strongly imply that the PF from patients with endometriosis has immunoregulatory/immunosuppressive activity and shifts the Th1/Th2 cytokine balance toward the Th2 response, which may account for deviation of local and systemic immune responses. However, a similar trend, albeit not a statistically significant one, was also observed in case of PF from women without endometriosis, thus suggesting that peritoneal milieu may in general display some immunoregulatory/immunosuppressive properties. It should be stressed, however, that our present observations were made on a relatively small number of PF samples and further studies are needed to reveal possible mechanism(s) responsible for this phenomenon.
Collapse
Affiliation(s)
- Joanna Olkowska-Truchanowicz
- Department of Transplantology and Central Tissue Bank, Centre of Biostructure Research, Medical University of Warsaw, 02-004 Warsaw, Poland;
| | - Agata Białoszewska
- Department of Histology and Embryology, Centre of Biostructure Research, Medical University of Warsaw, 02-004 Warsaw, Poland; (A.B.); (A.S.-I.); (I.J.)
| | - Aneta Zwierzchowska
- 1st Department of Obstetrics and Gynecology, Medical University of Warsaw, 02-015 Warsaw, Poland; (A.Z.); (F.D.); (E.B.)
- Department of Obstetrics and Gynecology, Multidisciplinary Hospital Warsaw-Miedzylesie, 04-749 Warsaw, Poland
| | - Alicja Sztokfisz-Ignasiak
- Department of Histology and Embryology, Centre of Biostructure Research, Medical University of Warsaw, 02-004 Warsaw, Poland; (A.B.); (A.S.-I.); (I.J.)
| | - Izabela Janiuk
- Department of Histology and Embryology, Centre of Biostructure Research, Medical University of Warsaw, 02-004 Warsaw, Poland; (A.B.); (A.S.-I.); (I.J.)
| | - Filip Dąbrowski
- 1st Department of Obstetrics and Gynecology, Medical University of Warsaw, 02-015 Warsaw, Poland; (A.Z.); (F.D.); (E.B.)
- Department of Gynecology and Obstetrics, Medical University of Silesia, 40-778 Katowice, Poland
| | | | - Ewa Barcz
- 1st Department of Obstetrics and Gynecology, Medical University of Warsaw, 02-015 Warsaw, Poland; (A.Z.); (F.D.); (E.B.)
- Department of Obstetrics and Gynecology, Multidisciplinary Hospital Warsaw-Miedzylesie, 04-749 Warsaw, Poland
| | - Katarzyna Bocian
- Department of Immunology, Faculty of Biology, University of Warsaw, 02-096 Warsaw, Poland;
- Correspondence: (K.B.); (J.M.)
| | - Jacek Malejczyk
- Department of Histology and Embryology, Centre of Biostructure Research, Medical University of Warsaw, 02-004 Warsaw, Poland; (A.B.); (A.S.-I.); (I.J.)
- Laboratory of Experimental Immunology, Military Institute of Hygiene and Epidemiology, 01-163 Warsaw, Poland
- Correspondence: (K.B.); (J.M.)
| |
Collapse
|
26
|
Lv Y, Lv D, Lv X, Xing P, Zhang J, Zhang Y. Immune Cell Infiltration-Based Characterization of Triple-Negative Breast Cancer Predicts Prognosis and Chemotherapy Response Markers. Front Genet 2021; 12:616469. [PMID: 33815462 PMCID: PMC8017297 DOI: 10.3389/fgene.2021.616469] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Accepted: 02/23/2021] [Indexed: 12/24/2022] Open
Abstract
Breast cancer represents the number one cause of cancer-associated mortality globally. The most aggressive molecular subtype is triple negative breast cancer (TNBC), of which limited therapeutic options are available. It is well known that breast cancer prognosis and tumor sensitivity toward immunotherapy are dictated by the tumor microenvironment. Breast cancer gene expression profiles were extracted from the METABRIC dataset and two TNBC clusters displaying unique immune features were identified. Activated immune cells formed a large proportion of cells in the high infiltration cluster, which correlated to a good prognosis. Differentially expressed genes (DEGs) extracted between two heterogeneous subtypes were used to further explore the underlying immune mechanism and to identify prognostic biomarkers. Functional enrichment analysis revealed that the DEGs were predominately related to some processes involved in activation and regulation of innate immune signaling. Using network analysis, we identified two modules in which genes were selected for further prognostic investigation. Validation by independent datasets revealed that CXCL9 and CXCL13 were good prognostic biomarkers for TNBC. We also performed comparisons between the above two genes and immune markers (CYT, APM, TILs, and TIS), as well as cell checkpoint marker expressions, and found a statistically significant correlation between them in both METABRIC and TCGA datasets. The potential of CXCL9 and CXCL13 to predict chemotherapy sensitivity was also evaluated. We found that the CXCL9 and CXCL13 were good predictors for chemotherapy and their expressions were higher in chemotherapy-responsive patients in contrast to those who were not responsive. In brief, immune infiltrate characterization on TNBC revealed heterogeneous subtypes with unique immune features allowed for the identification of informative and reliable characteristics representative of the local immune tumor microenvironment and were potential candidates to guide the management of TNBC patients.
Collapse
Affiliation(s)
- Yufei Lv
- Department of Anatomy, Harbin Medical University, Harbin, China
| | - Dongxu Lv
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Xiaohong Lv
- Department of Anatomy, Harbin Medical University, Harbin, China
| | - Ping Xing
- Department of Ultrasound, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jianguo Zhang
- Department of Breast Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yafang Zhang
- Department of Anatomy, Harbin Medical University, Harbin, China
| |
Collapse
|
27
|
Wang N, Wang S, Wang X, Zheng Y, Yang B, Zhang J, Pan B, Gao J, Wang Z. Research trends in pharmacological modulation of tumor-associated macrophages. Clin Transl Med 2021; 11:e288. [PMID: 33463063 PMCID: PMC7805405 DOI: 10.1002/ctm2.288] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 12/27/2020] [Accepted: 12/29/2020] [Indexed: 02/06/2023] Open
Abstract
As one of the most abundant immune cell populations in the tumor microenvironment (TME), tumor-associated macrophages (TAMs) play important roles in multiple solid malignancies, including breast cancer, prostate cancer, liver cancer, lung cancer, ovarian cancer, gastric cancer, pancreatic cancer, and colorectal cancer. TAMs could contribute to carcinogenesis, neoangiogenesis, immune-suppressive TME remodeling, cancer chemoresistance, recurrence, and metastasis. Therefore, reprogramming of the immune-suppressive TAMs by pharmacological approaches has attracted considerable research attention in recent years. In this review, the promising pharmaceutical targets, as well as the existing modulatory strategies of TAMs were summarized. The chemokine-chemokine receptor signaling, tyrosine kinase receptor signaling, metabolic signaling, and exosomal signaling have been highlighted in determining the biological functions of TAMs. Besides, both preclinical research and clinical trials have suggested the chemokine-chemokine receptor blockers, tyrosine kinase inhibitors, bisphosphonates, as well as the exosomal or nanoparticle-based targeting delivery systems as the promising pharmacological approaches for TAMs deletion or reprogramming. Lastly, the combined therapies of TAMs-targeting strategies with traditional treatments or immunotherapies as well as the exosome-like nanovesicles for cancer therapy are prospected.
Collapse
Affiliation(s)
- Neng Wang
- The Research Center for Integrative MedicineSchool of Basic Medical SciencesGuangzhou University of Chinese MedicineGuangzhouGuangdongChina
- The Research Center of Integrative Cancer MedicineDiscipline of Integrated Chinese and Western MedicineThe Second Clinical College of Guangzhou University of Chinese MedicineGuangzhouGuangdongChina
- Guangdong‐Hong Kong‐Macau Joint Lab on Chinese Medicine and Immune Disease ResearchGuangzhou University of Chinese MedicineGuangzhouGuangdongChina
| | - Shengqi Wang
- The Research Center of Integrative Cancer MedicineDiscipline of Integrated Chinese and Western MedicineThe Second Clinical College of Guangzhou University of Chinese MedicineGuangzhouGuangdongChina
- Guangdong‐Hong Kong‐Macau Joint Lab on Chinese Medicine and Immune Disease ResearchGuangzhou University of Chinese MedicineGuangzhouGuangdongChina
- Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine SyndromeGuangdong Provincial Hospital of Chinese MedicineGuangdong Provincial Academy of Chinese Medical SciencesGuangzhouGuangdongChina
| | - Xuan Wang
- The Research Center of Integrative Cancer MedicineDiscipline of Integrated Chinese and Western MedicineThe Second Clinical College of Guangzhou University of Chinese MedicineGuangzhouGuangdongChina
- Guangdong‐Hong Kong‐Macau Joint Lab on Chinese Medicine and Immune Disease ResearchGuangzhou University of Chinese MedicineGuangzhouGuangdongChina
- Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine SyndromeGuangdong Provincial Hospital of Chinese MedicineGuangdong Provincial Academy of Chinese Medical SciencesGuangzhouGuangdongChina
| | - Yifeng Zheng
- The Research Center of Integrative Cancer MedicineDiscipline of Integrated Chinese and Western MedicineThe Second Clinical College of Guangzhou University of Chinese MedicineGuangzhouGuangdongChina
- Guangdong‐Hong Kong‐Macau Joint Lab on Chinese Medicine and Immune Disease ResearchGuangzhou University of Chinese MedicineGuangzhouGuangdongChina
- Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine SyndromeGuangdong Provincial Hospital of Chinese MedicineGuangdong Provincial Academy of Chinese Medical SciencesGuangzhouGuangdongChina
| | - Bowen Yang
- The Research Center of Integrative Cancer MedicineDiscipline of Integrated Chinese and Western MedicineThe Second Clinical College of Guangzhou University of Chinese MedicineGuangzhouGuangdongChina
- Guangdong‐Hong Kong‐Macau Joint Lab on Chinese Medicine and Immune Disease ResearchGuangzhou University of Chinese MedicineGuangzhouGuangdongChina
- Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine SyndromeGuangdong Provincial Hospital of Chinese MedicineGuangdong Provincial Academy of Chinese Medical SciencesGuangzhouGuangdongChina
| | - Juping Zhang
- The Research Center of Integrative Cancer MedicineDiscipline of Integrated Chinese and Western MedicineThe Second Clinical College of Guangzhou University of Chinese MedicineGuangzhouGuangdongChina
- Guangdong‐Hong Kong‐Macau Joint Lab on Chinese Medicine and Immune Disease ResearchGuangzhou University of Chinese MedicineGuangzhouGuangdongChina
- Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine SyndromeGuangdong Provincial Hospital of Chinese MedicineGuangdong Provincial Academy of Chinese Medical SciencesGuangzhouGuangdongChina
| | - Bo Pan
- The Research Center of Integrative Cancer MedicineDiscipline of Integrated Chinese and Western MedicineThe Second Clinical College of Guangzhou University of Chinese MedicineGuangzhouGuangdongChina
- Guangdong‐Hong Kong‐Macau Joint Lab on Chinese Medicine and Immune Disease ResearchGuangzhou University of Chinese MedicineGuangzhouGuangdongChina
- Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine SyndromeGuangdong Provincial Hospital of Chinese MedicineGuangdong Provincial Academy of Chinese Medical SciencesGuangzhouGuangdongChina
| | - Jianli Gao
- Academy of Traditional Chinese MedicineZhejiang Chinese Medical UniversityHangzhouZhejiangChina
| | - Zhiyu Wang
- The Research Center for Integrative MedicineSchool of Basic Medical SciencesGuangzhou University of Chinese MedicineGuangzhouGuangdongChina
- The Research Center of Integrative Cancer MedicineDiscipline of Integrated Chinese and Western MedicineThe Second Clinical College of Guangzhou University of Chinese MedicineGuangzhouGuangdongChina
- Guangdong‐Hong Kong‐Macau Joint Lab on Chinese Medicine and Immune Disease ResearchGuangzhou University of Chinese MedicineGuangzhouGuangdongChina
- Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine SyndromeGuangdong Provincial Hospital of Chinese MedicineGuangdong Provincial Academy of Chinese Medical SciencesGuangzhouGuangdongChina
| |
Collapse
|
28
|
Lu C, Zhu J, Chen X, Hu Y, Xie W, Yao J, Huang S. Risk Stratification in Acute Myeloid Leukemia Using CXCR Gene Signatures: A Bioinformatics Analysis. Front Oncol 2020; 10:584766. [PMID: 33381455 PMCID: PMC7769120 DOI: 10.3389/fonc.2020.584766] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Accepted: 10/09/2020] [Indexed: 12/12/2022] Open
Abstract
The role of CXC chemokine receptors in tumors has been an increasingly researched focus in recent years. However, significant prognostic values of CXCR members in acute myeloid leukemia are yet to be explored profoundly. In this study, we firstly made an analysis of the relationship of CXCR family members and AML using samples from TCGA. Our results suggested that transcriptional expressions of CXCRs serve an important role in AML. CXCR transcript expressions, except CXCR1 expression, were significantly increased in AML. It displayed the expression pattern of CXCR members in different AML subtypes according to FAB classification. The correlations of CXCR transcript expression with different genotypes and karyotypes were also present. High CXCR2 expression was found to have a significantly worse prognosis compared with that of low CXCR2 expression, and CXCR2 was also found to be an independent prognostic factor. We also established a CXCR signature to identify high-risk subgroups of patients with AML. It was an independent prognostic factor and could become a powerful method to predict the survival rate of patients.
Collapse
Affiliation(s)
- Cong Lu
- Center for Stem Cell Research and Application, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Biological Targeted Therapy Key Laboratory in Hubei, Wuhan, China
| | - Jiang Zhu
- Center for Stem Cell Research and Application, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Biological Targeted Therapy Key Laboratory in Hubei, Wuhan, China
| | - Xiangjun Chen
- Center for Stem Cell Research and Application, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Biological Targeted Therapy Key Laboratory in Hubei, Wuhan, China
| | - Yanjie Hu
- Center for Stem Cell Research and Application, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Biological Targeted Therapy Key Laboratory in Hubei, Wuhan, China
| | - Wei Xie
- Center for Stem Cell Research and Application, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Biological Targeted Therapy Key Laboratory in Hubei, Wuhan, China
| | - Junxia Yao
- Center for Stem Cell Research and Application, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Biological Targeted Therapy Key Laboratory in Hubei, Wuhan, China
| | - Shiang Huang
- Center for Stem Cell Research and Application, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Biological Targeted Therapy Key Laboratory in Hubei, Wuhan, China
| |
Collapse
|