1
|
Vízkeleti L, Papp O, Doma V, Gil J, Markó-Varga G, Kovács SA, Győrffy B, Kárpáti S, Tímár J. Identification of genetic fingerprint of type I interferon therapy in visceral metastases of melanoma. Sci Rep 2024; 14:26540. [PMID: 39489756 PMCID: PMC11532416 DOI: 10.1038/s41598-024-77285-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Accepted: 10/21/2024] [Indexed: 11/05/2024] Open
Abstract
Malignant melanoma is a difficult-to-treat skin cancer with increasing incidence worldwide. Although type-I interferon (IFN) is no longer part of guidelines, several melanoma patients are treated with type-I interferon (IFN) at some point of the disease, potentially affecting its genetic progression. We run genome-wide copy number variation (CNV) analysis on previously type-I IFN-treated (n = 17) and control (n = 11) visceral metastases of melanoma patients. Results were completed with data from the TCGA and MM500 databases. We identified metastasis- and brain metastasis-specific gene signatures mostly affected by CN gains. Some cases were genetically resistant to IFN showing characteristic gene alterations (e.g. ABCA4 or ZEB2 gain and alterations of DNA repair genes). Analysis of a previously identified type-I IFN resistance gene set indicates that only a proportion of these genes was exclusive for the IFN-treated metastases reflecting a possible selective genomic pressure of endogenous IFNs during progression. Our data suggest that previous type-I IFN treatment and/or endogenous IFN production by immune response affect genomic progression of melanoma which may have clinical relevance, potentially influence immune checkpoint regulation in the tumor microenvironment.
Collapse
Affiliation(s)
- Laura Vízkeleti
- Department of Bioinformatics, Faculty of Medicine, Semmelweis University, 1094, Budapest, Hungary
- Department of Pathology, Forensic and Insurance Medicine, Faculty of Medicine, Semmelweis University, Üllői Str. 93., 1091, Budapest, Hungary
| | - Orsolya Papp
- Department of Pathology, Forensic and Insurance Medicine, Faculty of Medicine, Semmelweis University, Üllői Str. 93., 1091, Budapest, Hungary
- Turbine Simulated Cell Technologies, Budapest, 1027, Hungary
| | - Viktória Doma
- Department of Pathology, Forensic and Insurance Medicine, Faculty of Medicine, Semmelweis University, Üllői Str. 93., 1091, Budapest, Hungary
- Department of Dermatology, Venerology and Dermato-Oncology, Faculty of Medicine, Semmelweis University, 1085, Budapest, Hungary
| | - Jeovanis Gil
- Clinical Protein Science & Imaging, Biomedical Centre, Department of Biomedical Engineering, Lund University, 223 63, Lund, Sweden
| | - György Markó-Varga
- Clinical Protein Science & Imaging, Biomedical Centre, Department of Biomedical Engineering, Lund University, 223 63, Lund, Sweden
- Chemical Genomics Global Research Lab, Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, South Korea
- 1St Department of Surgery, Tokyo Medical University, Tokyo, 160-8582, Japan
| | - Szonja A Kovács
- Department of Bioinformatics, Faculty of Medicine, Semmelweis University, 1094, Budapest, Hungary
- Doctoral School of Pathological Sciences, Semmelweis University, 1085, Budapest, Hungary
- National Laboratory for Drug Research and Development, 1117, Budapest, Hungary
| | - Balázs Győrffy
- Department of Bioinformatics, Faculty of Medicine, Semmelweis University, 1094, Budapest, Hungary
| | - Sarolta Kárpáti
- Department of Dermatology, Venerology and Dermato-Oncology, Faculty of Medicine, Semmelweis University, 1085, Budapest, Hungary
| | - József Tímár
- Department of Pathology, Forensic and Insurance Medicine, Faculty of Medicine, Semmelweis University, Üllői Str. 93., 1091, Budapest, Hungary.
| |
Collapse
|
2
|
Liu Z, Fan Y, Cui M, Wang X, Zhao P. Investigation of tumour environments through advancements in microtechnology and nanotechnology. Biomed Pharmacother 2024; 178:117230. [PMID: 39116787 DOI: 10.1016/j.biopha.2024.117230] [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: 06/05/2024] [Revised: 07/28/2024] [Accepted: 07/30/2024] [Indexed: 08/10/2024] Open
Abstract
Cancer has a significant negative social and economic impact on both developed and developing countries. As a result, understanding the onset and progression of cancer is critical for developing therapies that can improve the well-being and health of individuals with cancer. With time, study has revealed, the tumor microenvironment has great influence on this process. Micro and nanoscale engineering techniques can be used to study the tumor microenvironment. Nanoscale and Microscale engineering use Novel technologies and designs with small dimensions to recreate the TME. Knowing how cancer cells interact with one another can help researchers develop therapeutic approaches that anticipate and counteract cancer cells' techniques for evading detection and fighting anti-cancer treatments, such as microfabrication techniques, microfluidic devices, nanosensors, and nanodevices used to study or recreate the tumor microenvironment. Nevertheless, a complicated action just like the growth and in cancer advancement, and their intensive association along the environment around it that has to be studied in more detail.
Collapse
Affiliation(s)
- Zhen Liu
- Department of Radiology, Shengjing Hospital of China Medical University, China
| | - Yan Fan
- Department of Pediatrics, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Mengyao Cui
- Department of Surgical Oncology, Breast Surgery, General Surgery, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Xu Wang
- Department of Surgical Oncology, Breast Surgery, General Surgery, The First Hospital of China Medical University, Shenyang, Liaoning, China.
| | - Pengfei Zhao
- Department of Radiology, Shengjing Hospital of China Medical University, China.
| |
Collapse
|
3
|
Karras F, Kunz M. Patient-derived melanoma models. Pathol Res Pract 2024; 259:155231. [PMID: 38508996 DOI: 10.1016/j.prp.2024.155231] [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: 11/30/2023] [Revised: 02/15/2024] [Accepted: 02/26/2024] [Indexed: 03/22/2024]
Abstract
Melanoma is a very aggressive, rapidly metastasizing tumor that has been studied intensively in the past regarding the underlying genetic and molecular mechanisms. More recently developed treatment modalities have improved response rates and overall survival of patients. However, the majority of patients suffer from secondary treatment resistance, which requires in depth analyses of the underlying mechanisms. Here, melanoma models based on patients-derived material may play an important role. Consequently, a plethora of different experimental techniques have been developed in the past years. Among these are 3D and 4D culture techniques, organotypic skin reconstructs, melanoma-on-chip models and patient-derived xenografts, Every technique has its own strengths but also weaknesses regarding throughput, reproducibility, and reflection of the human situation. Here, we provide a comprehensive overview of currently used techniques and discuss their use in different experimental settings.
Collapse
Affiliation(s)
- Franziska Karras
- Institute of Pathology, Otto-von-Guericke University Magdeburg, Leipziger Str. 44, Magdeburg 39120, Germany.
| | - Manfred Kunz
- Department of Dermatology, Venereology and Allergology, University Medical Center Leipzig, Philipp-Rosenthal-Str. 23, Leipzig 04103, Germany
| |
Collapse
|
4
|
Zhang Z, Wang T, Fang G, Xiao X, Zhang Z, Zhao J. Decitabine suppresses MDSC-induced immunosuppression through dual functional mechanism and inhibits melanoma metastasis. Med Oncol 2024; 41:165. [PMID: 38819590 DOI: 10.1007/s12032-024-02320-w] [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: 11/23/2023] [Accepted: 02/01/2024] [Indexed: 06/01/2024]
Abstract
Myeloid-derived suppressor cells (MDSCs) play a crucial role in promoting melanoma metastasis. Reprogramming MDSCs into mature M1 macrophages has emerged as a strategy to inhibit metastasis. Decitabine (Dec) is known to eradicate MDSCs and suppress tumor growth. In this study, we provide evidence that Dec not only reduces the MDSC population by inducing apoptosis, arresting cell cycle, and impairing recruitment, but also suppresses their immunosuppressive function by downregulating related genes and facilitating differentiation into M1 macrophages. Transcriptomic analysis of Dec-treated MDSCs revealed a marked downregulation of immunosuppressive genes including S100a9, S100a8, Vegf, Cxcr2, and Nos2. Meanwhile, M1 macrophage-associated genes involved in immune activation were upregulated, such as Ddx58, Isg15, Tap1, Ccl5, Cxcl9, and Cxcl10. Further bioinformatic analysis indicated that Dec promotes MDSC-to-M1 macrophage differentiation and activates innate immune pathways including NOD-like signaling to enhance anti-tumor immunity. Time-course studies implied that Dec upregulates myeloid transcription factor Irf7 to initiate MDSC differentiation and orchestrate the anti-tumor immune response. Collectively, our study unveils a novel dual-functional mechanism of Dec as both a cytotoxic agent reducing MDSCs and an inducer of their differentiation into M1 macrophages, thereby alleviating immunosuppression. This highlights Dec's potential for clinical melanoma metastasis suppression.
Collapse
Affiliation(s)
- Zhonghai Zhang
- Jiangsu Key Laboratory of Phylogenomics and Comparative Genomics, Jiangsu International Joint Center of Genomics, School of Life Sciences, Jiangsu Normal University, Xuzhou, 221116, Jiangsu, China
- Department of Physiology, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Tianlong Wang
- Jiangsu Key Laboratory of Phylogenomics and Comparative Genomics, Jiangsu International Joint Center of Genomics, School of Life Sciences, Jiangsu Normal University, Xuzhou, 221116, Jiangsu, China
| | - Gaochuan Fang
- Jiangsu Key Laboratory of Phylogenomics and Comparative Genomics, Jiangsu International Joint Center of Genomics, School of Life Sciences, Jiangsu Normal University, Xuzhou, 221116, Jiangsu, China
| | - Xufeng Xiao
- Jiangsu Key Laboratory of Phylogenomics and Comparative Genomics, Jiangsu International Joint Center of Genomics, School of Life Sciences, Jiangsu Normal University, Xuzhou, 221116, Jiangsu, China
| | - Zhengkui Zhang
- Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, 221002, Jiangsu, China.
| | - Jiaojiao Zhao
- Jiangsu Key Laboratory of Phylogenomics and Comparative Genomics, Jiangsu International Joint Center of Genomics, School of Life Sciences, Jiangsu Normal University, Xuzhou, 221116, Jiangsu, China.
| |
Collapse
|
5
|
Giannitelli SM, Peluzzi V, Raniolo S, Roscilli G, Trombetta M, Mozetic P, Rainer A. On-chip recapitulation of the tumor microenvironment: A decade of progress. Biomaterials 2024; 306:122482. [PMID: 38301325 DOI: 10.1016/j.biomaterials.2024.122482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 01/17/2024] [Accepted: 01/20/2024] [Indexed: 02/03/2024]
Abstract
One of the hurdles to the development of new anticancer therapies is the lack of in vitro models which faithfully reproduce the in vivo tumor microenvironment (TME). Understanding the dynamic relationships between the components of the TME in a controllable, scalable, and reliable setting would indeed support the discovery of biological targets impacting cancer diagnosis and therapy. Cancer research is increasingly shifting from traditional two-dimensional (2D) cell culture toward three-dimensional (3D) culture models, which have been demonstrated to increase the significance and predictive value of in vitro data. In this scenario, microphysiological systems (also known as organs-on-chip) have emerged as a relevant technological platform enabling more predictive investigation of cell-cell and cell-ECM interplay in cancer, attracting a significant research effort in the last years. This review illustrates one decade of progress in the field of tumor-microenvironment-on-chip (TMOC) approaches, exploiting either cell-laden microfluidic chambers or microfluidic confined tumor spheroids to model the TME. TMOCs have been designed to recapitulate several aspects of the TME, including tumor cells, the tumor-associated stroma, the immune system, and the vascular component. Significantly, the last aspect has emerged for its pivotal role in orchestrating cellular interactions and modulating drug pharmacokinetics on-chip. A further advancement has been represented by integration of TMOCs into multi-organ microphysiological systems, with the final aim to follow the metastatic cascade to target organs and to study the effects of chemotherapies at a systemic level. We highlight that the increased degree of complexity achieved by the most advanced TMOC models has enabled scientists to shed new light on the role of microenvironmental factors in tumor progression, metastatic cascade, and response to drugs.
Collapse
Affiliation(s)
- S M Giannitelli
- Department of Science and Technology for Sustainable Development and One Health, Università Campus Bio-Medico di Roma, via Álvaro del Portillo, 21, 00128, Rome, Italy.
| | - V Peluzzi
- Department of Engineering, Università Campus Bio-Medico di Roma, via Álvaro del Portillo 21, 00128, Rome, Italy.
| | - S Raniolo
- Department of Science and Technology for Sustainable Development and One Health, Università Campus Bio-Medico di Roma, via Álvaro del Portillo, 21, 00128, Rome, Italy.
| | - G Roscilli
- Takis s.r.l., Via di Castel Romano 100, 00128, Rome, Italy.
| | - M Trombetta
- Department of Science and Technology for Sustainable Development and One Health, Università Campus Bio-Medico di Roma, via Álvaro del Portillo, 21, 00128, Rome, Italy.
| | - P Mozetic
- Institute of Nanotechnology (NANOTEC), National Research Council, via Monteroni, 73100, Lecce, Italy.
| | - A Rainer
- Department of Engineering, Università Campus Bio-Medico di Roma, via Álvaro del Portillo 21, 00128, Rome, Italy; Fondazione Policlinico Universitario Campus Bio-Medico di Roma, via Álvaro del Portillo 200, 00128, Rome, Italy.
| |
Collapse
|
6
|
Bertani FR, Moghaddam FD, Panella C, Giannitelli SM, Peluzzi V, Gerardino A, Rainer A, Roscilli G, De Ninno A, Businaro L. Kinetic Detection of Apoptosis Events Via Caspase 3/7 Activation in a Tumor-Immune Microenvironment on a Chip. Methods Mol Biol 2024; 2748:109-118. [PMID: 38070111 DOI: 10.1007/978-1-0716-3593-3_9] [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] [Indexed: 12/18/2023]
Abstract
The development of advanced biological models like microphysiological systems, able to rebuild the complexity of the physiological and/or pathological environments at a single-cell detail level in an in-vivo-like approach, is proving to be a promising tool to understand the mechanisms of interactions between different cell populations and main features of several diseases. In this frame, the tumor-immune microenvironment on a chip represents a powerful tool to profile key aspects of cancer progression, immune activation, and response to therapy in several immuno-oncology applications. In the present chapter, we provide a protocol to identify and characterize the time evolution of apoptosis by time-lapse fluorescence and confocal imaging in a 3D microfluidic coculture murine model including cancer and spleen cells.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Adele De Ninno
- CNR-IFN Institute for Photonics and Nanotechnologies, Rome, Italy
| | - Luca Businaro
- CNR-IFN Institute for Photonics and Nanotechnologies, Rome, Italy.
| |
Collapse
|
7
|
Haynes C, Graham L, Bear HD. Adoptive immunotherapy with cells from tumor-draining lymph nodes activated and expanded in vitro. Methods Cell Biol 2023; 183:355-380. [PMID: 38548419 DOI: 10.1016/bs.mcb.2023.04.002] [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] [Indexed: 04/02/2024]
Abstract
Tumor-draining lymph nodes (tumor-DLNs) provide a rich source of tumor-reactive lymphocytes which can be used in adoptive immunotherapy (AIT) and that circumvent the need to resect autologous tumor, without the challenges and shortcomings associated with using autologous tumor or anti-CD3 monoclonal antibody. Bryostatin/Ionomycin (Bryo/Io) provide a useful method of activating tumor-DLNs such that they can readily be expanded to sufficient numbers to be used in AIT, and growing the tumor-DLN lymphocytes in the gamma chain cytokines IL-7 plus IL-15 is superior to IL-2 in terms of T cell numbers and phenotype. AIT with these cells induces tumor regression and provides protection against metastases and future tumor challenge. Here, we provide a stepwise protocol to sensitize tumor-DLN cells in donor mice, activate tumor-DLN T cells ex vivo using Bryo/Io, expansion of these cells in gamma chain cytokines and adoptive transfer of the expanded cells back into tumor-bearing hosts. Methods relevant to these experiments, such as injecting tumor cells intravenously and monitoring for pulmonary metastases, tumor volume measurement and resection, and use of luciferase-expressing tumor cells to monitor for metastases following resection, are described in detail. The methods outlined herein can be easily adapted to suit similar experiments across multiple tumor cell lines and syngeneic mouse models.
Collapse
Affiliation(s)
- Carolyn Haynes
- School of Medicine, Virginia Commonwealth University, Richmond, VA, United States; The Massey Cancer Center at Virginia Commonwealth University, Richmond, VA, United States
| | - Laura Graham
- School of Medicine, Virginia Commonwealth University, Richmond, VA, United States; The Massey Cancer Center at Virginia Commonwealth University, Richmond, VA, United States; Division of Surgical Oncology, Department of Surgery and the Massey Cancer Center at Virginia Commonwealth University, Richmond, VA, United States
| | - Harry D Bear
- School of Medicine, Virginia Commonwealth University, Richmond, VA, United States; The Massey Cancer Center at Virginia Commonwealth University, Richmond, VA, United States; Division of Surgical Oncology, Department of Surgery and the Massey Cancer Center at Virginia Commonwealth University, Richmond, VA, United States.
| |
Collapse
|
8
|
Mattei F, George JT, Jolly MK. Editorial: Organoids, organs-on-chip, nanoparticles and in silico approaches to dissect the tumor-immune dynamics and to unveil the drug resistance mechanisms to therapy in the tumor microenvironment. Front Immunol 2023; 14:1253551. [PMID: 37533861 PMCID: PMC10392942 DOI: 10.3389/fimmu.2023.1253551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 07/10/2023] [Indexed: 08/04/2023] Open
Affiliation(s)
- Fabrizio Mattei
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Jason T. George
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, United States
| | - Mohit Kumar Jolly
- Center for BioSystems Science and Engineering, Indian Institute of Science, Bangalore, India
| |
Collapse
|
9
|
Zhong F, Lin Y, Zhao L, Yang C, Ye Y, Shen Z. Reshaping the tumour immune microenvironment in solid tumours via tumour cell and immune cell DNA methylation: from mechanisms to therapeutics. Br J Cancer 2023; 129:24-37. [PMID: 37117649 PMCID: PMC10307880 DOI: 10.1038/s41416-023-02292-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Revised: 04/18/2023] [Accepted: 04/20/2023] [Indexed: 04/30/2023] Open
Abstract
In recent years, the tumour microenvironment (TME) of solid tumours has attracted more and more attention from researchers, especially those non-tumour components such as immune cells. Infiltration of various immune cells causes tumour immune microenvironment (TIME) heterogeneity, and results in different therapeutic effects. Accumulating evidence showed that DNA methylation plays a crucial role in remodelling TIME and is associated with the response towards immune checkpoint inhibitors (ICIs). During carcinogenesis, DNA methylation profoundly changes, specifically, there is a global loss of DNA methylation and increased DNA methylation at the promoters of suppressor genes. Immune cell differentiation is disturbed, and exclusion of immune cells from the TME occurs at least in part due to DNA methylation reprogramming. Therefore, pharmaceutical interventions targeting DNA methylation are promising. DNA methyltransferase inhibitors (DNMTis) enhance antitumor immunity by inducing transcription of transposable elements and consequent viral mimicry. DNMTis upregulate the expression of tumour antigens, mediate immune cells recruitment and reactivate exhausted immune cells. In preclinical studies, DNMTis have shown synergistic effect when combined with immunotherapies, suggesting new strategies to treat refractory solid tumours.
Collapse
Affiliation(s)
- Fengyun Zhong
- Department of Gastroenterological Surgery, Peking University People's Hospital, 100044, Beijing, P. R. China
- Laboratory of Surgical Oncology, Beijing Key Laboratory of Colorectal Cancer Diagnosis and Treatment Research, Peking University People's Hospital, 100044, Beijing, P. R. China
| | - Yilin Lin
- Department of Gastroenterological Surgery, Peking University People's Hospital, 100044, Beijing, P. R. China
- Laboratory of Surgical Oncology, Beijing Key Laboratory of Colorectal Cancer Diagnosis and Treatment Research, Peking University People's Hospital, 100044, Beijing, P. R. China
| | - Long Zhao
- Department of Gastroenterological Surgery, Peking University People's Hospital, 100044, Beijing, P. R. China
- Laboratory of Surgical Oncology, Beijing Key Laboratory of Colorectal Cancer Diagnosis and Treatment Research, Peking University People's Hospital, 100044, Beijing, P. R. China
| | - Changjiang Yang
- Department of Gastroenterological Surgery, Peking University People's Hospital, 100044, Beijing, P. R. China
- Laboratory of Surgical Oncology, Beijing Key Laboratory of Colorectal Cancer Diagnosis and Treatment Research, Peking University People's Hospital, 100044, Beijing, P. R. China
| | - Yingjiang Ye
- Department of Gastroenterological Surgery, Peking University People's Hospital, 100044, Beijing, P. R. China
- Laboratory of Surgical Oncology, Beijing Key Laboratory of Colorectal Cancer Diagnosis and Treatment Research, Peking University People's Hospital, 100044, Beijing, P. R. China
| | - Zhanlong Shen
- Department of Gastroenterological Surgery, Peking University People's Hospital, 100044, Beijing, P. R. China.
- Laboratory of Surgical Oncology, Beijing Key Laboratory of Colorectal Cancer Diagnosis and Treatment Research, Peking University People's Hospital, 100044, Beijing, P. R. China.
| |
Collapse
|
10
|
El Harane S, Zidi B, El Harane N, Krause KH, Matthes T, Preynat-Seauve O. Cancer Spheroids and Organoids as Novel Tools for Research and Therapy: State of the Art and Challenges to Guide Precision Medicine. Cells 2023; 12:cells12071001. [PMID: 37048073 PMCID: PMC10093533 DOI: 10.3390/cells12071001] [Citation(s) in RCA: 48] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 03/14/2023] [Accepted: 03/20/2023] [Indexed: 04/14/2023] Open
Abstract
Spheroids and organoids are important novel players in medical and life science research. They are gradually replacing two-dimensional (2D) cell cultures. Indeed, three-dimensional (3D) cultures are closer to the in vivo reality and open promising perspectives for academic research, drug screening, and personalized medicine. A large variety of cells and tissues, including tumor cells, can be the starting material for the generation of 3D cultures, including primary tissues, stem cells, or cell lines. A panoply of methods has been developed to generate 3D structures, including spontaneous or forced cell aggregation, air-liquid interface conditions, low cell attachment supports, magnetic levitation, and scaffold-based technologies. The choice of the most appropriate method depends on (i) the origin of the tissue, (ii) the presence or absence of a disease, and (iii) the intended application. This review summarizes methods and approaches for the generation of cancer spheroids and organoids, including their advantages and limitations. We also highlight some of the challenges and unresolved issues in the field of cancer spheroids and organoids, and discuss possible therapeutic applications.
Collapse
Affiliation(s)
- Sanae El Harane
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, 1206 Geneva, Switzerland
| | - Bochra Zidi
- Department of Medicine, Faculty of Medicine, University of Geneva, 1206 Geneva, Switzerland
| | - Nadia El Harane
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, 1206 Geneva, Switzerland
| | - Karl-Heinz Krause
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, 1206 Geneva, Switzerland
| | - Thomas Matthes
- Department of Medicine, Faculty of Medicine, University of Geneva, 1206 Geneva, Switzerland
| | - Olivier Preynat-Seauve
- Department of Medicine, Faculty of Medicine, University of Geneva, 1206 Geneva, Switzerland
- Laboratory of Experimental Cell Therapy, Department of Diagnostics, Geneva University Hospitals, 1206 Geneva, Switzerland
| |
Collapse
|
11
|
Pre-Clinical Evaluation of the Hypomethylating Agent Decitabine for the Treatment of T-Cell Lymphoblastic Lymphoma. Cancers (Basel) 2023; 15:cancers15030647. [PMID: 36765607 PMCID: PMC9913791 DOI: 10.3390/cancers15030647] [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: 11/30/2022] [Revised: 01/13/2023] [Accepted: 01/16/2023] [Indexed: 01/24/2023] Open
Abstract
T-cell lymphoblastic lymphoma (T-LBL) is a rare and aggressive lymphatic cancer, often diagnosed at a young age. Patients are treated with intensive chemotherapy, potentially followed by a hematopoietic stem cell transplantation. Although prognosis of T-LBL has improved with intensified treatment protocols, they are associated with side effects and 10-20% of patients still die from relapsed or refractory disease. Given this, the search toward less toxic anti-lymphoma therapies is ongoing. Here, we targeted the recently described DNA hypermethylated profile in T-LBL with the DNA hypomethylating agent decitabine. We evaluated the anti-lymphoma properties and downstream effects of decitabine, using patient derived xenograft (PDX) models. Decitabine treatment resulted in prolonged lymphoma-free survival in all T-LBL PDX models, which was associated with downregulation of the oncogenic MYC pathway. However, some PDX models showed more benefit of decitabine treatment compared to others. In more sensitive models, differentially methylated CpG regions resulted in more differentially expressed genes in open chromatin regions. This resulted in stronger downregulation of cell cycle genes and upregulation of immune response activating transcripts. Finally, we suggest a gene signature for high decitabine sensitivity in T-LBL. Altogether, we here delivered pre-clinical proof of the potential use of decitabine as a new therapeutic agent in T-LBL.
Collapse
|
12
|
Gordy JT, Sandhu AK, Fessler K, Luo K, Kapoor AR, Ayeh SK, Hui Y, Schill C, Chen F, Wang T, Karanika S, Sunshine JC, Karakousis PC, Markham RB. IFNα and 5-Aza-2'-deoxycytidine combined with a dendritic-cell targeting DNA vaccine alter tumor immune cell infiltration in the B16F10 melanoma model. Front Immunol 2023; 13:1074644. [PMID: 36741387 PMCID: PMC9892704 DOI: 10.3389/fimmu.2022.1074644] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 12/29/2022] [Indexed: 01/20/2023] Open
Abstract
Introduction DNA vaccines containing a fusion of the gene encoding chemokine MIP-3α (CCL20), the ligand for CCR6 on immature dendritic cells (DCs), to melanoma-associated antigen genes have enhanced anti-tumor immunity and efficacy compared to those lacking the chemokine gene. Previous work has shown that type-I interferon (IFNα or IFN) and 5-Aza-2'-deoxycytidine (5Aza) significantly enhance the therapeutic benefit of DNA vaccines as measured by reduced tumor burden and improved mouse survival. Methods Here, we explored mouse intratumoral immune correlates underlying the therapeutic benefit of this combination regimen (vaccine, IFN, and 5Aza) as compared to vaccine alone and IFN and 5Aza without vaccine, focusing on chemokine mRNA expression by qRT-PCR and inflammatory cellular infiltration into the tumor microenvironment (TME) by flow cytometry and immunohistochemistry (IHC). Results The combination group significantly upregulated intratumoral mRNA expression of key immune infiltration chemokines XCL1 and CXCL10. Flow cytometric analyses of tumor suspensions exhibited greater tumor infiltration of CD8+ DCs, CCR7+ DCs, and NK cells in the combination group, as well as reduced levels of myeloid-derived suppressor cells (MDSCs) in vaccinated groups. The mice receiving combination therapy also had greater proportions of effector/memory T-cells (Tem), in addition to showing an enhanced infiltration of Tem and central memory CD8+ T-cells, (Tcm). Tem and Tcm populations both correlated with smaller tumor size. Immunohistochemical analysis of tumors confirmed that CD8+ cells were more abundant overall and especially in the tumor parenchyma with combination therapy. Discussion Efficient targeting of antigen to immature DCs with a chemokine-fusion vaccine offers a potential alternative approach to classic and dendritic cell-based vaccines. Combining this approach with IFNα and 5Aza treatments significantly improved vaccine efficacy. This treatment creates an environment of increased inflammatory chemokines that facilitates the trafficking of CD8+ DCs, NK cells, and CD8+ T-cells, especially memory cells, while reducing the number of MDSCs. Importantly, in the combination group, CD8+ cells were more able to penetrate the tumor mass in addition to being more numerous. Further analysis of the pathways engaged by our combination therapy is expected to provide additional insights into melanoma pathogenesis and facilitate the development of novel treatment strategies.
Collapse
Affiliation(s)
- James T. Gordy
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | - Avinaash K. Sandhu
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | - Kaitlyn Fessler
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | - Kun Luo
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | - Aakanksha R. Kapoor
- Division of Infectious Diseases, Center for Tuberculosis Research, Department of Medicine, The Johns Hopkins Hospital, Baltimore, MD, United States
| | - Samuel K. Ayeh
- Division of Infectious Diseases, Center for Tuberculosis Research, Department of Medicine, The Johns Hopkins Hospital, Baltimore, MD, United States
| | - Yinan Hui
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | - Courtney Schill
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | - Fengyixin Chen
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | - Tianyin Wang
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | - Styliani Karanika
- Division of Infectious Diseases, Center for Tuberculosis Research, Department of Medicine, The Johns Hopkins Hospital, Baltimore, MD, United States
| | - Joel C. Sunshine
- The Departments of Dermatology, Pathology, and Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Petros C. Karakousis
- Division of Infectious Diseases, Center for Tuberculosis Research, Department of Medicine, The Johns Hopkins Hospital, Baltimore, MD, United States
| | - Richard B. Markham
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States,*Correspondence: Richard B. Markham,
| |
Collapse
|
13
|
Hargadon KM. Genetic dysregulation of immunologic and oncogenic signaling pathways associated with tumor-intrinsic immune resistance: a molecular basis for combination targeted therapy-immunotherapy for cancer. Cell Mol Life Sci 2023; 80:40. [PMID: 36629955 PMCID: PMC11072992 DOI: 10.1007/s00018-023-04689-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Revised: 01/02/2023] [Accepted: 01/04/2023] [Indexed: 01/12/2023]
Abstract
Since the turn of the century, advances in targeted therapy and immunotherapy have revolutionized the treatment of cancer. Although these approaches have far outperformed traditional therapies in various clinical settings, both remain plagued by mechanisms of innate and acquired resistance that limit therapeutic efficacy in many patients. With a focus on tumor-intrinsic resistance to immunotherapy, this review highlights our current understanding of the immunologic and oncogenic pathways whose genetic dysregulation in cancer cells enables immune escape. Emphasis is placed on genomic, epigenomic, transcriptomic, and proteomic aberrations that influence the activity of these pathways in the context of immune resistance. Specifically, the role of pathways that govern interferon signaling, antigen processing and presentation, and immunologic cell death as determinants of tumor immune susceptibility are discussed. Likewise, mechanisms of tumor immune resistance mediated by dysregulated RAS-MAPK, WNT, PI3K-AKT-mTOR, and cell cycle pathways are described. Finally, this review highlights the ways in which recent insight into genetic dysregulation of these immunologic and oncogenic signaling pathways is informing the design of combination targeted therapy-immunotherapy regimens that aim to restore immune susceptibility of cancer cells by overcoming resistance mechanisms that often limit the success of monotherapies.
Collapse
Affiliation(s)
- Kristian M Hargadon
- Hargadon Laboratory, Department of Biology, Hampden-Sydney College, Hampden-Sydney, VA, 23943, USA.
| |
Collapse
|
14
|
Zhang Z, Wang G, Li Y, Lei D, Xiang J, Ouyang L, Wang Y, Yang J. Recent progress in DNA methyltransferase inhibitors as anticancer agents. Front Pharmacol 2022; 13:1072651. [PMID: 37077808 PMCID: PMC10107375 DOI: 10.3389/fphar.2022.1072651] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 11/28/2022] [Indexed: 12/23/2022] Open
Abstract
DNA methylation mediated by DNA methyltransferase is an important epigenetic process that regulates gene expression in mammals, which plays a key role in silencing certain genes, such as tumor suppressor genes, in cancer, and it has become a promising therapeutic target for cancer treatment. Similar to other epigenetic targets, DNA methyltransferase can also be modulated by chemical agents. Four agents have already been approved to treat hematological cancers. In order to promote the development of a DNA methyltransferase inhibitor as an anti-tumor agent, in the current review, we discuss the relationship between DNA methylation and tumor, the anti-tumor mechanism, the research progress and pharmacological properties of DNA methyltransferase inhibitors, and the future research trend of DNA methyltransferase inhibitors.
Collapse
Affiliation(s)
- Zhixiong Zhang
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, Innovation Center of Nursing Research, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu, China
- Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Guan Wang
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, Innovation Center of Nursing Research, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu, China
| | - Yuyan Li
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, Innovation Center of Nursing Research, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu, China
- Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Dongsheng Lei
- School of Physical Science and Technology, Electron Microscopy Center of Lanzhou University, Lanzhou University, Lanzhou, China
| | - Jin Xiang
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, Innovation Center of Nursing Research, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu, China
| | - Liang Ouyang
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, Innovation Center of Nursing Research, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu, China
- Science and Technology Department, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Yanyan Wang
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, Innovation Center of Nursing Research, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu, China
- Science and Technology Department, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
- *Correspondence: Yanyan Wang, ; Jinliang Yang,
| | - Jinliang Yang
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, Innovation Center of Nursing Research, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu, China
- *Correspondence: Yanyan Wang, ; Jinliang Yang,
| |
Collapse
|
15
|
Anestopoulos I, Kyriakou S, Tragkola V, Paraskevaidis I, Tzika E, Mitsiogianni M, Deligiorgi MV, Petrakis G, Trafalis DT, Botaitis S, Giatromanolaki A, Koukourakis MI, Franco R, Pappa A, Panayiotidis MI. Targeting the epigenome in malignant melanoma: Facts, challenges and therapeutic promises. Pharmacol Ther 2022; 240:108301. [PMID: 36283453 DOI: 10.1016/j.pharmthera.2022.108301] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 10/03/2022] [Accepted: 10/19/2022] [Indexed: 11/16/2022]
Abstract
Malignant melanoma is the most lethal type of skin cancer with high rates of mortality. Although current treatment options provide a short-clinical benefit, acquired-drug resistance highlights the low 5-year survival rate among patients with advanced stage of the disease. In parallel, the involvement of an aberrant epigenetic landscape, (e.g., alterations in DNA methylation patterns, histone modifications marks and expression of non-coding RNAs), in addition to the genetic background, has been also associated with the onset and progression of melanoma. In this review article, we report on current therapeutic options in melanoma treatment with a focus on distinct epigenetic alterations and how their reversal, by specific drug compounds, can restore a normal phenotype. In particular, we concentrate on how single and/or combinatorial therapeutic approaches have utilized epigenetic drug compounds in being effective against malignant melanoma. Finally, the role of deregulated epigenetic mechanisms in promoting drug resistance to targeted therapies and immune checkpoint inhibitors is presented leading to the development of newly synthesized and/or improved drug compounds capable of targeting the epigenome of malignant melanoma.
Collapse
Affiliation(s)
- I Anestopoulos
- Department of Cancer Genetics, Therapeutics & Ultrastructural Pathology, The Cyprus Institute of Neurology & Genetics, Nicosia, Cyprus
| | - S Kyriakou
- Department of Cancer Genetics, Therapeutics & Ultrastructural Pathology, The Cyprus Institute of Neurology & Genetics, Nicosia, Cyprus
| | - V Tragkola
- Department of Cancer Genetics, Therapeutics & Ultrastructural Pathology, The Cyprus Institute of Neurology & Genetics, Nicosia, Cyprus
| | - I Paraskevaidis
- Department of Cancer Genetics, Therapeutics & Ultrastructural Pathology, The Cyprus Institute of Neurology & Genetics, Nicosia, Cyprus
| | - E Tzika
- Department of Cancer Genetics, Therapeutics & Ultrastructural Pathology, The Cyprus Institute of Neurology & Genetics, Nicosia, Cyprus
| | | | - M V Deligiorgi
- Laboratory of Pharmacology, Medical School, National & Kapodistrian University of Athens, Athens, Greece
| | - G Petrakis
- Saint George Hospital, Chania, Crete, Greece
| | - D T Trafalis
- Laboratory of Pharmacology, Medical School, National & Kapodistrian University of Athens, Athens, Greece
| | - S Botaitis
- Department of Surgery, Alexandroupolis University Hospital, Democritus University of Thrace School of Medicine, Alexandroupolis, Greece
| | - A Giatromanolaki
- Department of Pathology, Democritus University of Thrace, University General Hospital of Alexandroupolis, Alexandroupolis, Greece
| | - M I Koukourakis
- Radiotherapy / Oncology, Radiobiology & Radiopathology Unit, Department of Medicine, School of Health Sciences, Democritus University of Thrace, Alexandroupolis, Greece
| | - R Franco
- Redox Biology Centre, University of Nebraska-Lincoln, Lincoln, NE, USA; School of Veterinary Medicine & Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - A Pappa
- Department of Molecular Biology & Genetics, Democritus University of Thrace, Alexandroupolis, Greece
| | - M I Panayiotidis
- Department of Cancer Genetics, Therapeutics & Ultrastructural Pathology, The Cyprus Institute of Neurology & Genetics, Nicosia, Cyprus.
| |
Collapse
|
16
|
Michielon E, de Gruijl TD, Gibbs S. From simplicity to complexity in current melanoma models. Exp Dermatol 2022; 31:1818-1836. [PMID: 36103206 PMCID: PMC10092692 DOI: 10.1111/exd.14675] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 08/30/2022] [Accepted: 09/11/2022] [Indexed: 12/14/2022]
Abstract
Despite the recent impressive clinical success of immunotherapy against melanoma, development of primary and adaptive resistance against immune checkpoint inhibitors remains a major issue in a large number of treated patients. This highlights the need for melanoma models that replicate the tumor's intricate dynamics in the tumor microenvironment (TME) and associated immune suppression to study possible resistance mechanisms in order to improve current and test novel therapeutics. While two-dimensional melanoma cell cultures have been widely used to perform functional genomics screens in a high-throughput fashion, they are not suitable to answer more complex scientific questions. Melanoma models have also been established in a variety of experimental (humanized) animals. However, due to differences in physiology, such models do not fully represent human melanoma development. Therefore, fully human three-dimensional in vitro models mimicking melanoma cell interactions with the TME are being developed to address this need for more physiologically relevant models. Such models include melanoma organoids, spheroids, and reconstructed human melanoma-in-skin cultures. Still, while major advances have been made to complement and replace animals, these in vitro systems have yet to fully recapitulate human tumor complexity. Lastly, technical advancements have been made in the organ-on-chip field to replicate functions and microstructures of in vivo human tissues and organs. This review summarizes advancements made in understanding and treating melanoma and specifically aims to discuss the progress made towards developing melanoma models, their applications, limitations, and the advances still needed to further facilitate the development of therapeutics.
Collapse
Affiliation(s)
- Elisabetta Michielon
- Department of Molecular Cell Biology and Immunology, Amsterdam University Medical Center, Vrije Universiteit, Amsterdam, The Netherlands.,Amsterdam Institute for Infection and Immunity, Amsterdam University Medical Center, Vrije Universiteit, Amsterdam, The Netherlands.,Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
| | - Tanja D de Gruijl
- Amsterdam Institute for Infection and Immunity, Amsterdam University Medical Center, Vrije Universiteit, Amsterdam, The Netherlands.,Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands.,Department of Medical Oncology, Amsterdam University Medical Center, Vrije Universiteit, Amsterdam, The Netherlands
| | - Susan Gibbs
- Department of Molecular Cell Biology and Immunology, Amsterdam University Medical Center, Vrije Universiteit, Amsterdam, The Netherlands.,Amsterdam Institute for Infection and Immunity, Amsterdam University Medical Center, Vrije Universiteit, Amsterdam, The Netherlands.,Department of Oral Cell Biology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit, Amsterdam, The Netherlands
| |
Collapse
|
17
|
Lucarini V, Melaiu O, D’Amico S, Pastorino F, Tempora P, Scarsella M, Pezzullo M, De Ninno A, D’Oria V, Cilli M, Emionite L, Infante P, Di Marcotullio L, De Ioris MA, Barillari G, Alaggio R, Businaro L, Ponzoni M, Locatelli F, Fruci D. Combined mitoxantrone and anti-TGFβ treatment with PD-1 blockade enhances antitumor immunity by remodelling the tumor immune landscape in neuroblastoma. J Exp Clin Cancer Res 2022; 41:326. [PMID: 36397148 PMCID: PMC9670422 DOI: 10.1186/s13046-022-02525-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Accepted: 10/23/2022] [Indexed: 11/18/2022] Open
Abstract
Background Poor infiltration of functioning T cells renders tumors unresponsive to checkpoint-blocking immunotherapies. Here, we identified a combinatorial in situ immunomodulation strategy based on the administration of selected immunogenic drugs and immunotherapy to sensitize poorly T-cell-infiltrated neuroblastoma (NB) to the host antitumor immune response. Methods 975A2 and 9464D NB cell lines derived from spontaneous tumors of TH-MYCN transgenic mice were employed to study drug combinations able of enhancing the antitumor immune response using in vivo and ex vivo approaches. Migration of immune cells towards drug-treated murine-derived organotypic tumor spheroids (MDOTS) were assessed by microfluidic devices. Activation status of immune cells co-cultured with drug-treated MDOTS was evaluated by flow cytometry analysis. The effect of drug treatment on the immune content of subcutaneous or orthotopic tumors was comprehensively analyzed by flow-cytometry, immunohistochemistry and multiplex immunofluorescence. The chemokine array assay was used to detect soluble factors released into the tumor microenvironment. Patient-derived organotypic tumor spheroids (PDOTS) were generated from human NB specimens. Migration and activation status of autologous immune cells to drug-treated PDOTS were performed. Results We found that treatment with low-doses of mitoxantrone (MTX) recalled immune cells and promoted CD8+ T and NK cell activation in MDOTS when combined with TGFβ and PD-1 blockade. This combined immunotherapy strategy curbed NB growth resulting in the enrichment of a variety of both lymphoid and myeloid immune cells, especially intratumoral dendritic cells (DC) and IFNγ- and granzyme B-expressing CD8+ T cells and NK cells. A concomitant production of inflammatory chemokines involved in remodelling the tumor immune landscape was also detected. Interestingly, this treatment induced immune cell recruitment against PDOTS and activation of CD8+ T cells and NK cells. Conclusions Combined treatment with low-dose of MTX and anti-TGFβ treatment with PD-1 blockade improves antitumor immunity by remodelling the tumor immune landscape and overcoming the immunosuppressive microenvironment of aggressive NB. Supplementary Information The online version contains supplementary material available at 10.1186/s13046-022-02525-9.
Collapse
|
18
|
Seliger B, Al-Samadi A, Yang B, Salo T, Wickenhauser C. In vitro models as tools for screening treatment options of head and neck cancer. Front Med (Lausanne) 2022; 9:971726. [PMID: 36160162 PMCID: PMC9489836 DOI: 10.3389/fmed.2022.971726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 08/09/2022] [Indexed: 12/24/2022] Open
Abstract
Various in vitro models using primary and established 2- and 3-dimensional cultures, multicellular tumor spheroids, standardized tumor slice cultures, tumor organoids, and microfluidic systems obtained from tumor lesions/biopsies of head and neck cancer (HNC) have been employed for exploring and monitoring treatment options. All of these in vitro models are to a different degree able to capture the diversity of tumors, recapitulate the disease genetically, histologically, and functionally and retain their tumorigenic potential upon xenotransplantation. The models were used for the characterization of the malignant features of the tumors and for in vitro screens of drugs approved for the treatment of HNC, including chemotherapy and radiotherapy as well as recently developed targeted therapies and immunotherapies, or for novel treatments not yet licensed for these tumor entities. The implementation of the best suitable model will enlarge our knowledge of the oncogenic properties of HNC, expand the drug repertoire and help to develop individually tailored treatment strategies resulting in the translation of these findings into the clinic. This review summarizes the different approaches using preclinical in vitro systems with their advantages and disadvantages and their implementation as preclinical platforms to predict disease course, evaluate biomarkers and test therapy efficacy.
Collapse
Affiliation(s)
- Barbara Seliger
- Institute of Medical Immunology, Martin Luther University Halle-Wittenberg, Halle, Germany
- Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany
- *Correspondence: Barbara Seliger,
| | - Ahmed Al-Samadi
- Department of Oral and Maxillofacial Diseases, Clinicum, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Translational Immunology Research Program, Research Program Unit, University of Helsinki, Helsinki, Finland
| | - Bo Yang
- Institute of Medical Immunology, Martin Luther University Halle-Wittenberg, Halle, Germany
| | - Tuula Salo
- Department of Oral and Maxillofacial Diseases, Clinicum, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Translational Immunology Research Program, Research Program Unit, University of Helsinki, Helsinki, Finland
- Cancer Research and Translational Medicine Research Unit, University of Oulu, Oulu, Finland
- Medical Research Center Oulu, Oulu University Hospital, University of Oulu, Oulu, Finland
| | - Claudia Wickenhauser
- Institute of Pathology, Martin Luther University Halle-Wittenberg, Halle, Germany
| |
Collapse
|
19
|
Musella M, Guarracino A, Manduca N, Galassi C, Ruggiero E, Potenza A, Maccafeo E, Manic G, Mattiello L, Soliman Abdel Rehim S, Signore M, Pietrosanto M, Helmer-Citterich M, Pallocca M, Fanciulli M, Bruno T, De Nicola F, Corleone G, Di Benedetto A, Ercolani C, Pescarmona E, Pizzuti L, Guidi F, Sperati F, Vitale S, Macchia D, Spada M, Schiavoni G, Mattei F, De Ninno A, Businaro L, Lucarini V, Bracci L, Aricò E, Ziccheddu G, Facchiano F, Rossi S, Sanchez M, Boe A, Biffoni M, De Maria R, Vitale I, Sistigu A. Type I IFNs promote cancer cell stemness by triggering the epigenetic regulator KDM1B. Nat Immunol 2022; 23:1379-1392. [PMID: 36002648 PMCID: PMC9477743 DOI: 10.1038/s41590-022-01290-3] [Citation(s) in RCA: 68] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Accepted: 07/17/2022] [Indexed: 12/14/2022]
Abstract
Cancer stem cells (CSCs) are a subpopulation of cancer cells endowed with high tumorigenic, chemoresistant and metastatic potential. Nongenetic mechanisms of acquired resistance are increasingly being discovered, but molecular insights into the evolutionary process of CSCs are limited. Here, we show that type I interferons (IFNs-I) function as molecular hubs of resistance during immunogenic chemotherapy, triggering the epigenetic regulator demethylase 1B (KDM1B) to promote an adaptive, yet reversible, transcriptional rewiring of cancer cells towards stemness and immune escape. Accordingly, KDM1B inhibition prevents the appearance of IFN-I-induced CSCs, both in vitro and in vivo. Notably, IFN-I-induced CSCs are heterogeneous in terms of multidrug resistance, plasticity, invasiveness and immunogenicity. Moreover, in breast cancer (BC) patients receiving anthracycline-based chemotherapy, KDM1B positively correlated with CSC signatures. Our study identifies an IFN-I → KDM1B axis as a potent engine of cancer cell reprogramming, supporting KDM1B targeting as an attractive adjunctive to immunogenic drugs to prevent CSC expansion and increase the long-term benefit of therapy.
Collapse
Affiliation(s)
- Martina Musella
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Andrea Guarracino
- Department of Biology, University of Rome 'Tor Vergata', Rome, Italy
- Genomics Research Centre, Human Technopole, Milan, Italy
| | - Nicoletta Manduca
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Claudia Galassi
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
| | - Eliana Ruggiero
- Experimental Hematology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Alessia Potenza
- Experimental Hematology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Ester Maccafeo
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Gwenola Manic
- Italian Institute for Genomic Medicine (IIGM), Candiolo, Italy
- Candiolo Cancer Institute, FPO - IRCCS, Candiolo, Italy
| | - Luca Mattiello
- Italian Institute for Genomic Medicine (IIGM), Candiolo, Italy
- Candiolo Cancer Institute, FPO - IRCCS, Candiolo, Italy
| | - Sara Soliman Abdel Rehim
- Department of Biology, University of Rome 'Tor Vergata', Rome, Italy
- Italian Institute for Genomic Medicine (IIGM), Candiolo, Italy
| | - Michele Signore
- RPPA Unit, Proteomics Area, Core Facilities, Istituto Superiore di Sanità, Rome, Italy
| | - Marco Pietrosanto
- Department of Biology, University of Rome 'Tor Vergata', Rome, Italy
| | | | - Matteo Pallocca
- UOSD Clinical Trial Center, Biostatistics and Bioinformatics, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | | | - Tiziana Bruno
- SAFU Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | | | - Giacomo Corleone
- SAFU Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Anna Di Benedetto
- Pathology Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Cristiana Ercolani
- Pathology Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Edoardo Pescarmona
- Pathology Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Laura Pizzuti
- Division of Medical Oncology 2, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Francesco Guidi
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy
- Fondazione Policlinico Universitario 'A. Gemelli' - IRCCS, Rome, Italy
| | - Francesca Sperati
- UOSD Clinical Trial Center, Biostatistics and Bioinformatics, IRCCS San Gallicano Dermatological Institute, Rome, Italy
| | - Sara Vitale
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Daniele Macchia
- Center of Animal Research and Welfare, Istituto Superiore di Sanità, Rome, Italy
| | - Massimo Spada
- Center of Animal Research and Welfare, Istituto Superiore di Sanità, Rome, Italy
| | - Giovanna Schiavoni
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Fabrizio Mattei
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Adele De Ninno
- Institute for Photonics and Nanotechnologies, Italian National Research Council, Rome, Italy
| | - Luca Businaro
- Institute for Photonics and Nanotechnologies, Italian National Research Council, Rome, Italy
| | - Valeria Lucarini
- Department of Paediatric Haematology/Oncology and of Cell and Gene Therapy, Ospedale Pediatrico Bambino Gesù, IRCCS, Rome, Italy
| | - Laura Bracci
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Eleonora Aricò
- FaBioCell, Core Facilities, Istituto Superiore di Sanità, Rome, Italy
| | - Giovanna Ziccheddu
- Oncogenomics and Epigenetics, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Francesco Facchiano
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Stefania Rossi
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Massimo Sanchez
- Cytometry Unit, Core Facilities, Istituto Superiore di Sanità, Rome, Italy
| | - Alessandra Boe
- Cytometry Unit, Core Facilities, Istituto Superiore di Sanità, Rome, Italy
| | - Mauro Biffoni
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Ruggero De Maria
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy.
- Fondazione Policlinico Universitario 'A. Gemelli' - IRCCS, Rome, Italy.
| | - Ilio Vitale
- Italian Institute for Genomic Medicine (IIGM), Candiolo, Italy.
- Candiolo Cancer Institute, FPO - IRCCS, Candiolo, Italy.
| | - Antonella Sistigu
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy.
- Fondazione Policlinico Universitario 'A. Gemelli' - IRCCS, Rome, Italy.
- Tumor Immunology and Immunotherapy Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy.
| |
Collapse
|
20
|
Zhang J, Tavakoli H, Ma L, Li X, Han L, Li X. Immunotherapy discovery on tumor organoid-on-a-chip platforms that recapitulate the tumor microenvironment. Adv Drug Deliv Rev 2022; 187:114365. [PMID: 35667465 DOI: 10.1016/j.addr.2022.114365] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Revised: 04/17/2022] [Accepted: 05/25/2022] [Indexed: 02/06/2023]
Abstract
Cancer immunotherapy has achieved remarkable success over the past decade by modulating patients' own immune systems and unleashing pre-existing immunity. However, only a minority of cancer patients across different cancer types are able to benefit from immunotherapy treatment; moreover, among those small portions of patients with response, intrinsic and acquired resistance remains a persistent challenge. Because the tumor microenvironment (TME) is well recognized to play a critical role in tumor initiation, progression, metastasis, and the suppression of the immune system and responses to immunotherapy, understanding the interactions between the TME and the immune system is a pivotal step in developing novel and efficient cancer immunotherapies. With unique features such as low reagent consumption, dynamic and precise fluid control, versatile structures and function designs, and 3D cell co-culture, microfluidic tumor organoid-on-a-chip platforms that recapitulate key factors of the TME and the immune contexture have emerged as innovative reliable tools to investigate how tumors regulate their TME to counteract antitumor immunity and the mechanism of tumor resistance to immunotherapy. In this comprehensive review, we focus on recent advances in tumor organoid-on-a-chip platforms for studying the interaction between the TME and the immune system. We first review different factors of the TME that recent microfluidic in vitro systems reproduce to generate advanced tools to imitate the crosstalk between the TME and the immune system. Then, we discuss their applications in the assessment of different immunotherapies' efficacy using tumor organoid-on-a-chip platforms. Finally, we present an overview and the outlook of engineered microfluidic platforms in investigating the interactions between cancer and immune systems, and the adoption of patient-on-a-chip models in clinical applications toward personalized immunotherapy.
Collapse
Affiliation(s)
- Jie Zhang
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing 100029, China; Department of Chemistry and Biochemistry, University of Texas at El Paso, 500 W University Ave., El Paso, TX 79968, USA
| | - Hamed Tavakoli
- Department of Chemistry and Biochemistry, University of Texas at El Paso, 500 W University Ave., El Paso, TX 79968, USA
| | - Lei Ma
- Department of Chemistry and Biochemistry, University of Texas at El Paso, 500 W University Ave., El Paso, TX 79968, USA
| | - Xiaochun Li
- College of Biomedical Engineering, Taiyuan University of Technology, Taiyuan 030024, China
| | - Lichun Han
- Xi'an Daxing Hospital, Xi'an 710016, China
| | - XiuJun Li
- Department of Chemistry and Biochemistry, University of Texas at El Paso, 500 W University Ave., El Paso, TX 79968, USA; Border Biomedical Research Center, Forensic Science, & Environmental Science and Engineering, University of Texas at El Paso, 500 West University Ave., El Paso, TX 79968, USA.
| |
Collapse
|
21
|
Sieviläinen M, Saavalainen J, Adnan-Awad S, Salo T, Al-Samadi A. IDO1 Inhibition Reduces Immune Cell Exclusion Through Inducing Cell Migration While PD-1 Blockage Increases IL-6 and -8 Secretion From T Cells in Head and Neck Cancer. Front Immunol 2022; 13:812822. [PMID: 35359980 PMCID: PMC8963946 DOI: 10.3389/fimmu.2022.812822] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 02/14/2022] [Indexed: 12/13/2022] Open
Abstract
BackgroundImmune checkpoint inhibitors (ICIs), primarily anti-PD-1, are currently used to treat patients with recurrent or metastatic head and neck squamous cell carcinoma (HNSCC). However, only a minority of patients benefit from these costly therapies. Therefore, there is an unmet need to better understand the effect of ICIs on immune effector cells. This study aimed to investigate the effect of a PD-1 antibody and an IDO1 inhibitor on different lymphocyte populations (NK, CD4+, and CD8+ T cells) in term of migration, cytotoxicity, and cytokine release in the presence of HNSCC cells.MethodsUsing a microfluidic chip, we injected HSC-3 cells (an oral tongue squamous cell carcinoma cell line) embedded in a human tumor-derived matrix “myogel/fibrin” together with NK, CD4+, and CD8+ T cells in separate channels. The two channels were connected with microchannels. The PD-1 antibody nivolumab and IDO1 inhibitor epacadostat were added to the microfluidic chips. Lymphocyte migration and cytotoxicity were examined under fluorescent microscopy and cytokine release was measured using a FirePlex Human Discovery Cytokines Immunoassay.ResultsEpacadostat significantly increased the migration and infiltration of NK and CD4+ T cells, but not CD8+ T cells, towards the cancer cells. Nivolumab did not exhibit a similar effect. While CD8+ T cells alone showed near to no migration, adding CD4+ T cells enhanced migration towards the cancer cells. There was a mild nonsignificant increase in apoptosis of HSC-3 cells after adding epacadostat to lymphocytes. In contrast, HSC-3 proliferation was not affected by lymphocytes regardless of ICIs. Nivolumab significantly increased release of MIP1-α, IL-6, and IL-8 from NK, CD4+, and CD8+ T cells, respectively.ConclusionsThis study revealed that each subpopulation of lymphocytes respond differently to ICIs. We also revealed the subpopulation of lymphocytes responsible for the increases in specific serum cytokines after ICI treatment.
Collapse
Affiliation(s)
- Meri Sieviläinen
- Department of Oral and Maxillofacial Diseases, Clinicum, University of Helsinki, Helsinki, Finland
- Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- *Correspondence: Meri Sieviläinen,
| | - Jordan Saavalainen
- Department of Oral and Maxillofacial Diseases, Clinicum, University of Helsinki, Helsinki, Finland
- School of Biological Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Shady Adnan-Awad
- Clinical Pathology Department, National Cancer Institute, Cairo University, Cairo, Egypt
- Hematology Research Unit, Department of Hematology, University of Helsinki and Helsinki University Central Hospital Comprehensive Cancer Center, Helsinki, Finland
| | - Tuula Salo
- Department of Oral and Maxillofacial Diseases, Clinicum, University of Helsinki, Helsinki, Finland
- Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Cancer and Translational Medicine Research Unit, University of Oulu, Oulu, Finland
- Medical Research Center, Oulu University Hospital, Oulu, Finland
- Department of Pathology, Helsinki University Hospital (HUS), Helsinki, Finland
| | - Ahmed Al-Samadi
- Department of Oral and Maxillofacial Diseases, Clinicum, University of Helsinki, Helsinki, Finland
- Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| |
Collapse
|
22
|
Liu Z, Ren Y, Weng S, Xu H, Li L, Han X. A New Trend in Cancer Treatment: The Combination of Epigenetics and Immunotherapy. Front Immunol 2022; 13:809761. [PMID: 35140720 PMCID: PMC8818678 DOI: 10.3389/fimmu.2022.809761] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 01/03/2022] [Indexed: 12/15/2022] Open
Abstract
In recent years, immunotherapy has become a hot spot in the treatment of tumors. As an emerging treatment, it solves many problems in traditional cancer treatment and has now become the main method for cancer treatment. Although immunotherapy is promising, most patients do not respond to treatment or develop resistance. Therefore, in order to achieve a better therapeutic effect, combination therapy has emerged. The combination of immune checkpoint inhibition and epigenetic therapy is one such strategy. In this review, we summarize the current understanding of the key mechanisms of how epigenetic mechanisms affect cancer immune responses and reveal the key role of epigenetic processes in regulating immune cell function and mediating anti-tumor immunity. In addition, we highlight the outlook of combined epigenetic and immune regimens, particularly the combination of immune checkpoint blockade with epigenetic agents, to address the limitations of immunotherapy alone.
Collapse
Affiliation(s)
- Zaoqu Liu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Interventional Institute of Zhengzhou University, Zhengzhou, China
- Interventional Treatment and Clinical Research Center of Henan Province, Zhengzhou, China
| | - Yuqing Ren
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Siyuan Weng
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Interventional Institute of Zhengzhou University, Zhengzhou, China
- Interventional Treatment and Clinical Research Center of Henan Province, Zhengzhou, China
| | - Hui Xu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Interventional Institute of Zhengzhou University, Zhengzhou, China
- Interventional Treatment and Clinical Research Center of Henan Province, Zhengzhou, China
| | - Lifeng Li
- Internet Medical and System Applications of National Engineering Laboratory, Zhengzhou, China
- Medical School, Huanghe Science and Technology University, Zhengzhou, China
- *Correspondence: Xinwei Han, ; Lifeng Li,
| | - Xinwei Han
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Interventional Institute of Zhengzhou University, Zhengzhou, China
- Interventional Treatment and Clinical Research Center of Henan Province, Zhengzhou, China
- *Correspondence: Xinwei Han, ; Lifeng Li,
| |
Collapse
|
23
|
Niu Y, Chen J, Qiao Y. Epigenetic Modifications in Tumor-Associated Macrophages: A New Perspective for an Old Foe. Front Immunol 2022; 13:836223. [PMID: 35140725 PMCID: PMC8818998 DOI: 10.3389/fimmu.2022.836223] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 01/06/2022] [Indexed: 12/11/2022] Open
Abstract
Tumorigenesis is frequently accompanied by chronic inflammation, and the tumor microenvironment (TME) can be considered an ecosystem that consists of tumor cells, endotheliocytes, fibroblasts, immune cells and acellular components such as extracellular matrix. For tumor cells, their survival advantages are dependent on both genetic and epigenetic alterations, while other cells mainly present epigenetic modifications. Macrophages are the most plastic type of immune cells and undergo diverse epigenetic alterations in the TME. Some of these epigenetic modifications mitigate against cancer progression, and others accelerate this process. Due to the complex roles of macrophages in the TME, it is urgent to understand their epigenetic modifications associated with the TME. Here, we mainly summarize recent findings on TME-associated epigenetic alterations of tumor-associated macrophages (TAMs), including DNA methylation, posttranslational modifications of histone proteins, chromatin remodeling, and noncoding RNA-mediated epigenetic regulation. At the end of this review, we also discuss the translational potential of these epigenetic modifications for developing novel cancer therapies targeting TAMs.
Collapse
Affiliation(s)
- Yuqin Niu
- The First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, China
| | - Jianxiang Chen
- School of Pharmacy, Department of Hepatology, the Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou, China
- *Correspondence: Yiting Qiao, ; Jianxiang Chen,
| | - Yiting Qiao
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, China
- *Correspondence: Yiting Qiao, ; Jianxiang Chen,
| |
Collapse
|
24
|
Adeshakin AO, Adeshakin FO, Yan D, Wan X. Regulating Histone Deacetylase Signaling Pathways of Myeloid-Derived Suppressor Cells Enhanced T Cell-Based Immunotherapy. Front Immunol 2022; 13:781660. [PMID: 35140716 PMCID: PMC8818783 DOI: 10.3389/fimmu.2022.781660] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 01/03/2022] [Indexed: 12/12/2022] Open
Abstract
Immunotherapy has emerged as a promising approach to combat immunosuppressive tumor microenvironment (TME) for improved cancer treatment. FDA approval for the clinical use of programmed death receptor 1/programmed death-ligand 1 (PD-1/PD-L1) inhibitors revolutionized T cell-based immunotherapy. Although only a few cancer patients respond to this treatment due to several factors including the accumulation of immunosuppressive cells in the TME. Several immunosuppressive cells within the TME such as regulatory T cells, myeloid cells, and cancer-associated fibroblast inhibit the activation and function of T cells to promote tumor progression. The roles of epigenetic modifiers such as histone deacetylase (HDAC) in cancer have long been investigated but little is known about their impact on immune cells. Recent studies showed inhibiting HDAC expression on myeloid-derived suppressor cells (MDSCs) promoted their differentiation to less suppressive cells and reduced their immunosuppressive effect in the TME. HDAC inhibitors upregulated PD-1 or PD-L1 expression level on tumor or immune cells sensitizing tumor-bearing mice to anti-PD-1/PD-L1 antibodies. Herein we discuss how inhibiting HDAC expression on MDSCs could circumvent drawbacks to immune checkpoint inhibitors and improve cancer immunotherapy. Furthermore, we highlighted current challenges and future perspectives of HDAC inhibitors in regulating MDSCs function for effective cancer immunotherapy.
Collapse
Affiliation(s)
- Adeleye O. Adeshakin
- Guangdong Immune Cell Therapy Engineering and Technology Research Center, Center for Protein and Cell-Based Drugs, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- University of Chinese Academy of Sciences, Beijing , China
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children’s Research Hospital, Memphis, TN, United States
| | - Funmilayo O. Adeshakin
- Guangdong Immune Cell Therapy Engineering and Technology Research Center, Center for Protein and Cell-Based Drugs, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- University of Chinese Academy of Sciences, Beijing , China
| | - Dehong Yan
- Guangdong Immune Cell Therapy Engineering and Technology Research Center, Center for Protein and Cell-Based Drugs, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- University of Chinese Academy of Sciences, Beijing , China
- *Correspondence: Dehong Yan, ; Xiaochun Wan,
| | - Xiaochun Wan
- Guangdong Immune Cell Therapy Engineering and Technology Research Center, Center for Protein and Cell-Based Drugs, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- University of Chinese Academy of Sciences, Beijing , China
- *Correspondence: Dehong Yan, ; Xiaochun Wan,
| |
Collapse
|
25
|
Fukuda K, Okamura K, Riding RL, Fan X, Afshari K, Haddadi NS, McCauley SM, Guney MH, Luban J, Funakoshi T, Yaguchi T, Kawakami Y, Khvorova A, Fitzgerald KA, Harris JE. AIM2 regulates anti-tumor immunity and is a viable therapeutic target for melanoma. J Exp Med 2021; 218:212521. [PMID: 34325468 PMCID: PMC8329870 DOI: 10.1084/jem.20200962] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 05/24/2021] [Accepted: 07/09/2021] [Indexed: 12/14/2022] Open
Abstract
The STING and absent in melanoma 2 (AIM2) pathways are activated by the presence of cytosolic DNA, and STING agonists enhance immunotherapeutic responses. Here, we show that dendritic cell (DC) expression of AIM2 within human melanoma correlates with poor prognosis and, in contrast to STING, AIM2 exerts an immunosuppressive effect within the melanoma microenvironment. Vaccination with AIM2-deficient DCs improves the efficacy of both adoptive T cell therapy and anti–PD-1 immunotherapy for “cold tumors,” which exhibit poor therapeutic responses. This effect did not depend on prolonged survival of vaccinated DCs, but on tumor-derived DNA that activates STING-dependent type I IFN secretion and subsequent production of CXCL10 to recruit CD8+ T cells. Additionally, loss of AIM2-dependent IL-1β and IL-18 processing enhanced the treatment response further by limiting the recruitment of regulatory T cells. Finally, AIM2 siRNA-treated mouse DCs in vivo and human DCs in vitro enhanced similar anti-tumor immune responses. Thus, targeting AIM2 in tumor-infiltrating DCs is a promising new treatment strategy for melanoma.
Collapse
Affiliation(s)
- Keitaro Fukuda
- Department of Dermatology, University of Massachusetts Medical School, Worcester, MA.,Department of Dermatology, Keio University School of Medicine, Tokyo, Japan
| | - Ken Okamura
- Department of Dermatology, University of Massachusetts Medical School, Worcester, MA
| | - Rebecca L Riding
- Department of Dermatology, University of Massachusetts Medical School, Worcester, MA
| | - Xueli Fan
- Department of Dermatology, University of Massachusetts Medical School, Worcester, MA
| | - Khashayar Afshari
- Department of Dermatology, University of Massachusetts Medical School, Worcester, MA
| | - Nazgol-Sadat Haddadi
- Department of Dermatology, University of Massachusetts Medical School, Worcester, MA
| | - Sean M McCauley
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA
| | - Mehmet H Guney
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA
| | - Jeremy Luban
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA.,Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, MA
| | - Takeru Funakoshi
- Department of Dermatology, Keio University School of Medicine, Tokyo, Japan
| | - Tomonori Yaguchi
- Division of Cellular Signaling, Institute for Advanced Medical Research, Keio University School of Medicine, Tokyo, Japan
| | - Yutaka Kawakami
- Division of Cellular Signaling, Institute for Advanced Medical Research, Keio University School of Medicine, Tokyo, Japan
| | - Anastasia Khvorova
- RNA Therapeutics Institute, University of Massachusetts Medical School, Worcester, MA.,Department of Molecular Medicine, University of Massachusetts Medical School, Worcester, MA
| | - Katherine A Fitzgerald
- Department of Infectious Diseases and Immunology, University of Massachusetts Medical School, Worcester, MA
| | - John E Harris
- Department of Dermatology, University of Massachusetts Medical School, Worcester, MA
| |
Collapse
|
26
|
Paterson K, Zanivan S, Glasspool R, Coffelt SB, Zagnoni M. Microfluidic technologies for immunotherapy studies on solid tumours. LAB ON A CHIP 2021; 21:2306-2329. [PMID: 34085677 PMCID: PMC8204114 DOI: 10.1039/d0lc01305f] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Accepted: 05/09/2021] [Indexed: 05/10/2023]
Abstract
Immunotherapy is a powerful and targeted cancer treatment that exploits the body's immune system to attack and eliminate cancerous cells. This form of therapy presents the possibility of long-term control and prevention of recurrence due to the memory capabilities of the immune system. Various immunotherapies are successful in treating haematological malignancies and have dramatically improved outcomes in melanoma. However, tackling other solid tumours is more challenging, mostly because of the immunosuppressive tumour microenvironment (TME). Current in vitro models based on traditional 2D cell monolayers and animal models, such as patient-derived xenografts, have limitations in their ability to mimic the complexity of the human TME. As a result, they have inadequate translational value and can be poorly predictive of clinical outcome. Thus, there is a need for robust in vitro preclinical tools that more faithfully recapitulate human solid tumours to test novel immunotherapies. Microfluidics and lab-on-a-chip technologies offer opportunities, especially when performing mechanistic studies, to understand the role of the TME in immunotherapy, and to expand the experimental throughput when using patient-derived tissue through its miniaturization capabilities. This review first introduces the basic concepts of immunotherapy, presents the current preclinical approaches used in immuno-oncology for solid tumours and then discusses the underlying challenges. We provide a rationale for using microfluidic-based approaches, highlighting the most recent microfluidic technologies and methodologies that have been used for studying cancer-immune cell interactions and testing the efficacy of immunotherapies in solid tumours. Ultimately, we discuss achievements and limitations of the technology, commenting on potential directions for incorporating microfluidic technologies in future immunotherapy studies.
Collapse
Affiliation(s)
- K Paterson
- Centre for Microsystems and Photonics, EEE Department, University of Strathclyde, Glasgow, UK.
| | - S Zanivan
- Institute of Cancer Sciences, University of Glasgow, Glasgow, UK and Cancer Research UK Beatson Institute, Glasgow, UK
| | - R Glasspool
- Institute of Cancer Sciences, University of Glasgow, Glasgow, UK and Beatson West of Scotland Cancer Centre, Glasgow, UK
| | - S B Coffelt
- Institute of Cancer Sciences, University of Glasgow, Glasgow, UK and Cancer Research UK Beatson Institute, Glasgow, UK
| | - M Zagnoni
- Centre for Microsystems and Photonics, EEE Department, University of Strathclyde, Glasgow, UK.
| |
Collapse
|
27
|
Abstract
Tumor metastasis is a singularly important determinant of survival in most cancers. Historically, radiation therapy (RT) directed at a primary tumor mass was associated infrequently with remission of metastasis outside the field of irradiation. This away-from-target or "abscopal effect" received fringe attention because of its rarity. With the advent of immunotherapy, there are now increasing reports of abscopal effects upon RT in combination with immune checkpoint inhibition. This sparked investigation into underlying mechanisms and clinical trials aimed at enhancement of this effect. While these studies clearly attribute the abscopal effect to an antitumor immune response, the initial molecular triggers for its onset and specificity remain enigmatic. Here, we propose that DNA damage-induced inflammation coupled with neoantigen generation is essential during this intriguing phenomenon of systemic tumor regression and discuss the implications of this model for treatment aimed at triggering the abscopal effect in metastatic cancer.
Collapse
|
28
|
Ciccolella M, Andreone S, Mancini J, Sestili P, Negri D, Pacca AM, D’Urso MT, Macchia D, Canese R, Pang K, SaiYing Ko T, Decadt Y, Schiavoni G, Mattei F, Belardelli F, Aricò E, Bracci L. Anticancer Effects of Sublingual Type I IFN in Combination with Chemotherapy in Implantable and Spontaneous Tumor Models. Cells 2021; 10:845. [PMID: 33917958 PMCID: PMC8068355 DOI: 10.3390/cells10040845] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 03/31/2021] [Accepted: 04/01/2021] [Indexed: 02/06/2023] Open
Abstract
Salivary gland tumors are a heterogeneous group of neoplasms representing less than 10% of all head and neck tumors. Among salivary gland tumors, salivary duct carcinoma (SDC) is a rare, but highly aggressive malignant tumor resembling ductal breast carcinoma. Sublingual treatments are promising for SDC due to the induction of both local and systemic biological effects and to reduced systemic toxicity compared to other administration routes. In the present study, we first established that the sublingual administration of type I IFN (IFN-I) is safe and feasible, and exerts antitumor effects both as monotherapy and in combination with chemotherapy in transplantable tumor models, i.e., B16-OVA melanoma and EG.7-OVA lymphoma. Subsequently, we proved that sublingual IFN-I in combination with cyclophosphamide (CTX) induces a long-lasting reduction of tumor mass in NeuT transgenic mice that spontaneously develop SDC. Most importantly, tumor shrinkage in NeuT transgenic micewas accompanied by the emergence of tumor-specific cellular immune responses both in the blood and in the tumor tissue. Altogether, these results provide evidence that sublingual IFN holds promise in combination with chemotherapy for the treatment of cancer.
Collapse
Affiliation(s)
- Maria Ciccolella
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (M.C.); (S.A.); (J.M.); (G.S.); (F.M.)
| | - Sara Andreone
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (M.C.); (S.A.); (J.M.); (G.S.); (F.M.)
| | - Jacopo Mancini
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (M.C.); (S.A.); (J.M.); (G.S.); (F.M.)
| | - Paola Sestili
- National Center for the Control and Evaluation of Medicines, 00161 Rome, Italy;
| | - Donatella Negri
- Department of Infectious Diseases, Istituto Superiore di Sanità, 00161 Rome, Italy;
| | - Anna Maria Pacca
- Animal Research and Welfare Centre, Istituto Superiore di Sanità, 00161 Rome, Italy; (A.M.P.); (M.T.D.); (D.M.)
| | - Maria Teresa D’Urso
- Animal Research and Welfare Centre, Istituto Superiore di Sanità, 00161 Rome, Italy; (A.M.P.); (M.T.D.); (D.M.)
| | - Daniele Macchia
- Animal Research and Welfare Centre, Istituto Superiore di Sanità, 00161 Rome, Italy; (A.M.P.); (M.T.D.); (D.M.)
| | - Rossella Canese
- Core Facilities, Istituto Superiore di Sanità, 00161 Rome, Italy;
| | - Ken Pang
- Biolingus AG, CH-6052 Hergiswil NW, Switzerland; (K.P.); (T.S.K.); (Y.D.)
- Murdoch Children’s Research Institute, Parkville 3052, Australia
- The Walter and Eliza Hall Institute of Medical Research, Parkville 3052, Australia
- Department of Paediatrics, University of Melbourne, Parkville 3010, Australia
| | - Thomas SaiYing Ko
- Biolingus AG, CH-6052 Hergiswil NW, Switzerland; (K.P.); (T.S.K.); (Y.D.)
| | - Yves Decadt
- Biolingus AG, CH-6052 Hergiswil NW, Switzerland; (K.P.); (T.S.K.); (Y.D.)
| | - Giovanna Schiavoni
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (M.C.); (S.A.); (J.M.); (G.S.); (F.M.)
| | - Fabrizio Mattei
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (M.C.); (S.A.); (J.M.); (G.S.); (F.M.)
| | - Filippo Belardelli
- Institute of Translational Pharmacology, Consiglio Nazionale delle Ricerche, 00133 Rome, Italy;
| | - Eleonora Aricò
- Core Facilities, Istituto Superiore di Sanità, 00161 Rome, Italy;
| | - Laura Bracci
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (M.C.); (S.A.); (J.M.); (G.S.); (F.M.)
| |
Collapse
|
29
|
Mattei F, Andreone S, Mencattini A, De Ninno A, Businaro L, Martinelli E, Schiavoni G. Oncoimmunology Meets Organs-on-Chip. Front Mol Biosci 2021; 8:627454. [PMID: 33842539 PMCID: PMC8032996 DOI: 10.3389/fmolb.2021.627454] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 03/04/2021] [Indexed: 01/04/2023] Open
Abstract
Oncoimmunology represents a biomedical research discipline coined to study the roles of immune system in cancer progression with the aim of discovering novel strategies to arm it against the malignancy. Infiltration of immune cells within the tumor microenvironment is an early event that results in the establishment of a dynamic cross-talk. Here, immune cells sense antigenic cues to mount a specific anti-tumor response while cancer cells emanate inhibitory signals to dampen it. Animals models have led to giant steps in this research context, and several tools to investigate the effect of immune infiltration in the tumor microenvironment are currently available. However, the use of animals represents a challenge due to ethical issues and long duration of experiments. Organs-on-chip are innovative tools not only to study how cells derived from different organs interact with each other, but also to investigate on the crosstalk between immune cells and different types of cancer cells. In this review, we describe the state-of-the-art of microfluidics and the impact of OOC in the field of oncoimmunology underlining the importance of this system in the advancements on the complexity of tumor microenvironment.
Collapse
Affiliation(s)
- Fabrizio Mattei
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Sara Andreone
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Arianna Mencattini
- Department of Electronic Engineering, University of Rome Tor Vergata, Rome, Italy.,Interdisciplinary Center for Advanced Studies on Lab-on-Chip and Organ-on-Chip Applications (ICLOC), University of Rome Tor Vergata, Rome, Italy
| | - Adele De Ninno
- Institute for Photonics and Nanotechnologies, Italian National Research Council, Rome, Italy
| | - Luca Businaro
- Institute for Photonics and Nanotechnologies, Italian National Research Council, Rome, Italy
| | - Eugenio Martinelli
- Department of Electronic Engineering, University of Rome Tor Vergata, Rome, Italy.,Interdisciplinary Center for Advanced Studies on Lab-on-Chip and Organ-on-Chip Applications (ICLOC), University of Rome Tor Vergata, Rome, Italy
| | - Giovanna Schiavoni
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| |
Collapse
|
30
|
Mass-Preserving Approximation of a Chemotaxis Multi-Domain Transmission Model for Microfluidic Chips. MATHEMATICS 2021. [DOI: 10.3390/math9060688] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The present work is inspired by the recent developments in laboratory experiments made on chips, where the culturing of multiple cell species was possible. The model is based on coupled reaction-diffusion-transport equations with chemotaxis and takes into account the interactions among cell populations and the possibility of drug administration for drug testing effects. Our effort is devoted to the development of a simulation tool that is able to reproduce the chemotactic movement and the interactions between different cell species (immune and cancer cells) living in a microfluidic chip environment. The main issues faced in this work are the introduction of mass-preserving and positivity-preserving conditions, involving the balancing of incoming and outgoing fluxes passing through interfaces between 2D and 1D domains of the chip and the development of mass-preserving and positivity preserving numerical conditions at the external boundaries and at the interfaces between 2D and 1D domains.
Collapse
|
31
|
Chen W, Cheng P, Jiang J, Ren Y, Wu D, Xue D. Epigenomic and genomic analysis of transcriptome modulation in skin cutaneous melanoma. Aging (Albany NY) 2020; 12:12703-12725. [PMID: 32639949 PMCID: PMC7377867 DOI: 10.18632/aging.103115] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Accepted: 03/31/2020] [Indexed: 04/10/2023]
Abstract
Skin cutaneous melanoma (SKCM) is characterized by both epigenetic DNA methylation (MET) abnormalities and genomic copy number variations (CNVs). The resulting transcriptome dysregulation promotes progression of many cancers. In this study, DNA copy numbers and MET, as well as mRNA expression, were examined in 466 SKCM samples from The Cancer Genome Atlas. Our results indicate that CNVs-correlated (CNVcor) genes and MET-correlated (METcor) genes are coregulated to a remarkable degree. In addition, integrative multi-omics analysis of both METcor and CNVcor genes revealed four SKCM subtypes with differing prognoses; these subtypes were validated with independent data. Immune cell scores were markedly elevated in the iC1 subtype, which had the best prognosis. Immune cell infiltration correlated with DNA MET or CNV level in SKCM. In the iC3 subtype, which was associated with the most aggressive SKCM cases, FAM135B gene mutation frequencies were increased, while CD8A, GBP5, KIAA0040, and SAMHD1 expression were downregulated, suggesting that these genes play important roles in cancer development and immune responses. Taken together, the results of our epigenetic and genomic transcriptome modulation analysis improve our understanding of SKCM pathobiology and may aid in the development of more effective therapies.
Collapse
Affiliation(s)
- Wuzhen Chen
- Department of Surgical Oncology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou Zhejiang, P.R. China
- Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, Hangzhou, China
| | - Pu Cheng
- Department of Gynecology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou Zhejiang, P.R. China
- Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, Hangzhou, China
| | - Jingxin Jiang
- Department of Surgical Oncology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou Zhejiang, P.R. China
- Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, Hangzhou, China
| | - Yunqing Ren
- Department of Gynecology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou Zhejiang, P.R. China
- Department of Dermatology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou Zhejiang, P.R. China
| | - Dang Wu
- Department of Radiation Oncology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou Zhejiang, P.R. China
- Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, Hangzhou, China
| | - Dan Xue
- Department of Plastic Surgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou Zhejiang, P.R. China
- Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, Hangzhou, China
| |
Collapse
|
32
|
Dahn ML, Cruickshank BM, Jackson AJ, Dean C, Holloway RW, Hall SR, Coyle KM, Maillet H, Waisman DM, Goralski KB, Giacomantonio CA, Weaver ICG, Marcato P. Decitabine Response in Breast Cancer Requires Efficient Drug Processing and Is Not Limited by Multidrug Resistance. Mol Cancer Ther 2020; 19:1110-1122. [PMID: 32156786 DOI: 10.1158/1535-7163.mct-19-0745] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 01/30/2020] [Accepted: 03/05/2020] [Indexed: 11/16/2022]
Abstract
Dysregulation of DNA methylation is an established feature of breast cancers. DNA demethylating therapies like decitabine are proposed for the treatment of triple-negative breast cancers (TNBC) and indicators of response need to be identified. For this purpose, we characterized the effects of decitabine in a panel of 10 breast cancer cell lines and observed a range of sensitivity to decitabine that was not subtype specific. Knockdown of potential key effectors demonstrated the requirement of deoxycytidine kinase (DCK) for decitabine response in breast cancer cells. In treatment-naïve breast tumors, DCK was higher in TNBCs, and DCK levels were sustained or increased post chemotherapy treatment. This suggests that limited DCK levels will not be a barrier to response in patients with TNBC treated with decitabine as a second-line treatment or in a clinical trial. Methylome analysis revealed that genome-wide, region-specific, tumor suppressor gene-specific methylation, and decitabine-induced demethylation did not predict response to decitabine. Gene set enrichment analysis of transcriptome data demonstrated that decitabine induced genes within apoptosis, cell cycle, stress, and immune pathways. Induced genes included those characterized by the viral mimicry response; however, knockdown of key effectors of the pathway did not affect decitabine sensitivity suggesting that breast cancer growth suppression by decitabine is independent of viral mimicry. Finally, taxol-resistant breast cancer cells expressing high levels of multidrug resistance transporter ABCB1 remained sensitive to decitabine, suggesting that the drug could be used as second-line treatment for chemoresistant patients.
Collapse
Affiliation(s)
- Margaret L Dahn
- Department of Pathology, Dalhousie University, Halifax, Nova Scotia, Canada
| | | | - Ainsleigh J Jackson
- Department of Microbiology and Immunology, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Cheryl Dean
- Department of Pathology, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Ryan W Holloway
- Department of Pathology, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Steven R Hall
- Department of Pharmacology, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Krysta M Coyle
- Department of Pathology, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Hillary Maillet
- Department of Psychology and Neuroscience, Dalhousie University, Halifax, Nova Scotia, Canada
| | - David M Waisman
- Department of Pathology, Dalhousie University, Halifax, Nova Scotia, Canada.,Department of Biochemistry and Molecular Biology, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Kerry B Goralski
- Department of Pharmacology, Dalhousie University, Halifax, Nova Scotia, Canada.,College of Pharmacy, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Carman A Giacomantonio
- Department of Pathology, Dalhousie University, Halifax, Nova Scotia, Canada.,Department of Surgery, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Ian C G Weaver
- Department of Pathology, Dalhousie University, Halifax, Nova Scotia, Canada.,Department of Psychology and Neuroscience, Dalhousie University, Halifax, Nova Scotia, Canada.,Department of Psychiatry, Dalhousie University, Halifax, Nova Scotia, Canada.,Brain Repair Centre, Halifax, Nova Scotia, Canada
| | - Paola Marcato
- Department of Pathology, Dalhousie University, Halifax, Nova Scotia, Canada. .,Department of Microbiology and Immunology, Dalhousie University, Halifax, Nova Scotia, Canada
| |
Collapse
|
33
|
Luker AJ, Graham LJ, Smith TM, Camarena C, Zellner MP, Gilmer JJS, Damle SR, Conrad DH, Bear HD, Martin RK. The DNA methyltransferase inhibitor, guadecitabine, targets tumor-induced myelopoiesis and recovers T cell activity to slow tumor growth in combination with adoptive immunotherapy in a mouse model of breast cancer. BMC Immunol 2020; 21:8. [PMID: 32106810 PMCID: PMC7045411 DOI: 10.1186/s12865-020-0337-5] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Accepted: 02/14/2020] [Indexed: 12/14/2022] Open
Abstract
Background Myeloid derived suppressor cells (MDSCs) present a significant obstacle to cancer immunotherapy because they dampen anti-tumor cytotoxic T cell responses. Previous groups, including our own, have reported on the myelo-depletive effects of certain chemotherapy agents. We have shown previously that decitabine increased tumor cell Class I and tumor antigen expression, increased ability of tumor cells to stimulate T lymphocytes, depleted tumor-induced MDSC in vivo and augmented immunotherapy of a murine mammary carcinoma. Results In this study, we expand upon this observation by testing a next-generation DNA methyltransferase inhibitor (DNMTi), guadecitabine, which has increased stability in the circulation. Using the 4 T1 murine mammary carcinoma model, in BALB/cJ female mice, we found that guadecitabine significantly reduces tumor burden in a T cell-dependent manner by preventing excessive myeloid proliferation and systemic accumulation of MDSC. The remaining MDSC were shifted to an antigen-presenting phenotype. Building upon our previous publication, we show that guadecitabine enhances the therapeutic effect of adoptively transferred antigen-experienced lymphocytes to diminish tumor growth and improve overall survival. We also show guadecitabine’s versatility with similar tumor reduction and augmentation of immunotherapy in the C57BL/6 J E0771 murine breast cancer model. Conclusions Guadecitabine depleted and altered MDSC, inhibited growth of two different murine mammary carcinomas in vivo, and augmented immunotherapeutic efficacy. Based on these findings, we believe the immune-modulatory effects of guadecitabine can help rescue anti-tumor immune response and contribute to the overall effectiveness of current cancer immunotherapies.
Collapse
Affiliation(s)
- Andrea J Luker
- Department of Microbiology and Immunology, School of Medicine, VCU, Box 980678, Richmond, VA, 23298, USA.,Massey Cancer Center, VCU, Box 980678, Richmond, VA, 23298, USA
| | - Laura J Graham
- Department of Microbiology and Immunology, School of Medicine, VCU, Box 980678, Richmond, VA, 23298, USA.,Massey Cancer Center, VCU, Box 980678, Richmond, VA, 23298, USA
| | - Timothy M Smith
- Department of Microbiology and Immunology, School of Medicine, VCU, Box 980678, Richmond, VA, 23298, USA.,Massey Cancer Center, VCU, Box 980678, Richmond, VA, 23298, USA
| | - Carmen Camarena
- Department of Microbiology and Immunology, School of Medicine, VCU, Box 980678, Richmond, VA, 23298, USA
| | - Matt P Zellner
- Department of Microbiology and Immunology, School of Medicine, VCU, Box 980678, Richmond, VA, 23298, USA
| | - Jamie-Jean S Gilmer
- Department of Biology, College of Humanities and Sciences, VCU, Richmond, VA, USA
| | - Sheela R Damle
- Department of Microbiology and Immunology, School of Medicine, VCU, Box 980678, Richmond, VA, 23298, USA.,Massey Cancer Center, VCU, Box 980678, Richmond, VA, 23298, USA
| | - Daniel H Conrad
- Department of Microbiology and Immunology, School of Medicine, VCU, Box 980678, Richmond, VA, 23298, USA.,Massey Cancer Center, VCU, Box 980678, Richmond, VA, 23298, USA
| | - Harry D Bear
- Department of Microbiology and Immunology, School of Medicine, VCU, Box 980678, Richmond, VA, 23298, USA.,Massey Cancer Center, VCU, Box 980678, Richmond, VA, 23298, USA.,Division of Surgical Oncology, Department of Surgery, VCU, Richmond, VA, USA
| | - Rebecca K Martin
- Department of Microbiology and Immunology, School of Medicine, VCU, Box 980678, Richmond, VA, 23298, USA. .,Massey Cancer Center, VCU, Box 980678, Richmond, VA, 23298, USA.
| |
Collapse
|
34
|
Gordy JT, Luo K, Kapoor A, Kim ES, Ayeh SK, Karakousis PC, Markham RB. Treatment with an immature dendritic cell-targeting vaccine supplemented with IFN-α and an inhibitor of DNA methylation markedly enhances survival in a murine melanoma model. Cancer Immunol Immunother 2020; 69:569-580. [PMID: 31980915 DOI: 10.1007/s00262-019-02471-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Accepted: 12/31/2019] [Indexed: 12/14/2022]
Abstract
BACKGROUND The chemokine MIP-3α (CCL20) binds to CCR6 on immature dendritic cells. DNA vaccines fusing MIP-3α to melanoma-associated antigens have shown improved efficacy and immunogenicity in the B16F10 mouse melanoma model. Here, we report that the combination of type-I interferon therapy (IFNα) with 5-Aza-2'-deoxycitidine (5Aza) profoundly enhanced the therapeutic efficacy of a MIP-3α-Gp100-Trp2 DNA vaccine. METHODS Beginning on day 5 post-transplantation of B16F10 melanoma, vaccine was administered intramuscularly (i.m.) by electroporation. CpG adjuvant was given 2 days later. 5Aza was given intraperitoneally at 1 mg/kg and IFNα therapy either intratumorally or i.m. as noted. Tumor sizes, tumor growth, and mouse survival were assessed. Tumor lysate gene expression levels and tumor-infiltrating lymphocytes (TILs) were assessed by qRT-PCR and flow cytometry, respectively. RESULTS Adding IFNα and 5Aza treatments to mice vaccinated with MIP-3α-Gp100-Trp2 leads to reduced tumor burden and increased median survival (39% over vaccine and 95% over controls). Tumor lysate expression of CCL19 and CCR7 were upregulated ten and fivefold over vaccine, respectively. Vaccine-specific and overall CD8+ TILs were increased over vaccine (sevenfold and fourfold, respectively), as well as the proportion of TILs that were CD8+ (twofold). CONCLUSIONS Efficient targeting of antigen to immature dendritic cells with a chemokine-fusion vaccine offers an alternative to classic and dendritic cell vaccines. Combining this approach with IFNα and 5Aza treatment significantly improved vaccine efficacy. This improved efficacy correlated with changes in chemokine gene expression and CD8+ TIL infiltration and was dependent on the presence of all therapeutic components.
Collapse
Affiliation(s)
- James T Gordy
- The W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, 615 N. Wolfe Street, Baltimore, MD, 21205, USA
| | - Kun Luo
- The W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, 615 N. Wolfe Street, Baltimore, MD, 21205, USA
| | - Aakanksha Kapoor
- Department of Medicine, Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Emily S Kim
- Department of Medicine, Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Samuel K Ayeh
- Department of Medicine, Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Petros C Karakousis
- Department of Medicine, Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Richard B Markham
- The W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, 615 N. Wolfe Street, Baltimore, MD, 21205, USA.
| |
Collapse
|
35
|
Dan H, Zhang S, Zhou Y, Guan Q. DNA Methyltransferase Inhibitors: Catalysts For Antitumour Immune Responses. Onco Targets Ther 2019; 12:10903-10916. [PMID: 31849494 PMCID: PMC6913319 DOI: 10.2147/ott.s217767] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Accepted: 10/02/2019] [Indexed: 12/15/2022] Open
Abstract
Epigenetics is a kind of heritable change that involves the unaltered DNA sequence and can have effects on gene expression. The regulatory mechanism mainly includes DNA methylation, histone modification and non-coding RNA regulation. DNA methylation is currently the most studied aspect of epigenetics. It is widely present in eukaryotic cells and is the most important epigenetic mark in the regulation of gene expression in the cell. DNA methyltransferase inhibitors (DNMTi) have been increasingly recognized in the field of cancer immunotherapy, have been approved for the treatment of acute myeloid leukaemia (AML) and are widely being used in clinical trials of cancer immunotherapies. DNMTi promote the reactivation of tumour suppressor genes, enhance tumour immunogenicity, and stimulate a variety of immune cells to secrete cytokines that exert cytotoxic effects, promote tumour cell death, including macrophages, natural killer (NK) cells and CD8+ T cells, and upregulate major histocompatibility complex (MHC) class I expression levels. Here, we mainly summarize the epigenetics related to DNMTi and their regulation of the antitumour immune response and DNMTi combined with immuno-therapeutics or histone deacetylase inhibitors to demonstrate the great development potential and clinical application value of DNMTi.
Collapse
Affiliation(s)
- Huimin Dan
- Gansu Province Key Laboratory of Gastrointestinal Diseases, The First Hospital of Lanzhou University, Lanzhou University, Lanzhou, Gansu Province, People's Republic of China
| | - Shanshan Zhang
- Gansu Province Key Laboratory of Gastrointestinal Diseases, The First Hospital of Lanzhou University, Lanzhou University, Lanzhou, Gansu Province, People's Republic of China
| | - Yongning Zhou
- Gansu Province Key Laboratory of Gastrointestinal Diseases, The First Hospital of Lanzhou University, Lanzhou University, Lanzhou, Gansu Province, People's Republic of China
| | - Quanlin Guan
- Gansu Province Key Laboratory of Gastrointestinal Diseases, The First Hospital of Lanzhou University, Lanzhou University, Lanzhou, Gansu Province, People's Republic of China
| |
Collapse
|
36
|
Nguyen M, De Ninno A, Mencattini A, Mermet-Meillon F, Fornabaio G, Evans SS, Cossutta M, Khira Y, Han W, Sirven P, Pelon F, Di Giuseppe D, Bertani FR, Gerardino A, Yamada A, Descroix S, Soumelis V, Mechta-Grigoriou F, Zalcman G, Camonis J, Martinelli E, Businaro L, Parrini MC. Dissecting Effects of Anti-cancer Drugs and Cancer-Associated Fibroblasts by On-Chip Reconstitution of Immunocompetent Tumor Microenvironments. Cell Rep 2019; 25:3884-3893.e3. [PMID: 30590056 DOI: 10.1016/j.celrep.2018.12.015] [Citation(s) in RCA: 106] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Revised: 08/06/2018] [Accepted: 12/03/2018] [Indexed: 01/16/2023] Open
Abstract
A major challenge in cancer research is the complexity of the tumor microenvironment, which includes the host immunological setting. Inspired by the emerging technology of organ-on-chip, we achieved 3D co-cultures in microfluidic devices (integrating four cell populations: cancer, immune, endothelial, and fibroblasts) to reconstitute ex vivo a human tumor ecosystem (HER2+ breast cancer). We visualized and quantified the complex dynamics of this tumor-on-chip, in the absence or in the presence of the drug trastuzumab (Herceptin), a targeted antibody therapy directed against the HER2 receptor. We uncovered the capacity of the drug trastuzumab to specifically promote long cancer-immune interactions (>50 min), recapitulating an anti-tumoral ADCC (antibody-dependent cell-mediated cytotoxicity) immune response. Cancer-associated fibroblasts (CAFs) antagonized the effects of trastuzumab. These observations constitute a proof of concept that tumors-on-chip are powerful platforms to study ex vivo immunocompetent tumor microenvironments, to characterize ecosystem-level drug responses, and to dissect the roles of stromal components.
Collapse
Affiliation(s)
- Marie Nguyen
- Institut Curie, Centre de Recherche, Paris Sciences et Lettres Research University, 75005 Paris, France; ART Group, INSERM U830, 75005 Paris, France
| | - Adele De Ninno
- Institute for Photonics and Nanotechnology, Italian National Research Council, 00156 Rome, Italy; Department of Civil Engineering and Computer Science, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Arianna Mencattini
- Department of Electronic Engineering, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Fanny Mermet-Meillon
- Institut Curie, Centre de Recherche, Paris Sciences et Lettres Research University, 75005 Paris, France; ART Group, INSERM U830, 75005 Paris, France
| | - Giulia Fornabaio
- Institut Curie, Centre de Recherche, Paris Sciences et Lettres Research University, 75005 Paris, France; ART Group, INSERM U830, 75005 Paris, France
| | - Sophia S Evans
- Institut Curie, Centre de Recherche, Paris Sciences et Lettres Research University, 75005 Paris, France; ART Group, INSERM U830, 75005 Paris, France
| | - Mélissande Cossutta
- Institut Curie, Centre de Recherche, Paris Sciences et Lettres Research University, 75005 Paris, France; ART Group, INSERM U830, 75005 Paris, France
| | - Yasmine Khira
- Institut Curie, Centre de Recherche, Paris Sciences et Lettres Research University, 75005 Paris, France; ART Group, INSERM U830, 75005 Paris, France
| | - Weijing Han
- Institut Curie, Centre de Recherche, Paris Sciences et Lettres Research University, 75005 Paris, France; ART Group, INSERM U830, 75005 Paris, France
| | - Philémon Sirven
- Institut Curie, Centre de Recherche, Paris Sciences et Lettres Research University, 75005 Paris, France; Immunity and Cancer, INSERM U932, INSERM Center of Clinical Investigations, CIC IGR Curie, 75005 Paris, France
| | - Floriane Pelon
- Institut Curie, Centre de Recherche, Paris Sciences et Lettres Research University, 75005 Paris, France; Stress and Cancer Team, labelized by Ligue Nationale Contre le Cancer, INSERM U830, 75005 Paris, France
| | - Davide Di Giuseppe
- Department of Electronic Engineering, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Francesca Romana Bertani
- Institute for Photonics and Nanotechnology, Italian National Research Council, 00156 Rome, Italy
| | - Annamaria Gerardino
- Institute for Photonics and Nanotechnology, Italian National Research Council, 00156 Rome, Italy
| | - Ayako Yamada
- Institut Curie, Centre de Recherche, Paris Sciences et Lettres Research University, 75005 Paris, France; Laboratoire Physico Chimie Curie, CNRS UMR168, 75005 Paris, France; Institut Pierre-Gilles de Gennes, 75005 Paris, France
| | - Stéphanie Descroix
- Institut Curie, Centre de Recherche, Paris Sciences et Lettres Research University, 75005 Paris, France; Laboratoire Physico Chimie Curie, CNRS UMR168, 75005 Paris, France; Institut Pierre-Gilles de Gennes, 75005 Paris, France
| | - Vassili Soumelis
- Institut Curie, Centre de Recherche, Paris Sciences et Lettres Research University, 75005 Paris, France; Immunity and Cancer, INSERM U932, INSERM Center of Clinical Investigations, CIC IGR Curie, 75005 Paris, France
| | - Fatima Mechta-Grigoriou
- Institut Curie, Centre de Recherche, Paris Sciences et Lettres Research University, 75005 Paris, France; Stress and Cancer Team, labelized by Ligue Nationale Contre le Cancer, INSERM U830, 75005 Paris, France
| | - Gérard Zalcman
- Institut Curie, Centre de Recherche, Paris Sciences et Lettres Research University, 75005 Paris, France; ART Group, INSERM U830, 75005 Paris, France; Centre d'Investigation Clinique (CIC) 1425, Hôpital Bichat-Claude Bernard, Université Paris-Diderot, Paris, France
| | - Jacques Camonis
- Institut Curie, Centre de Recherche, Paris Sciences et Lettres Research University, 75005 Paris, France; ART Group, INSERM U830, 75005 Paris, France
| | - Eugenio Martinelli
- Department of Electronic Engineering, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Luca Businaro
- Institute for Photonics and Nanotechnology, Italian National Research Council, 00156 Rome, Italy
| | - Maria Carla Parrini
- Institut Curie, Centre de Recherche, Paris Sciences et Lettres Research University, 75005 Paris, France; ART Group, INSERM U830, 75005 Paris, France.
| |
Collapse
|
37
|
Abstract
Over the past decade, preclinical and clinical research have confirmed the essential role of interferons for effective host immunological responses to malignant cells. Type I interferons (IFNα and IFNβ) directly regulate transcription of >100 downstream genes, which results in a myriad of direct (on cancer cells) and indirect (through immune effector cells and vasculature) effects on the tumour. New insights into endogenous and exogenous activation of type I interferons in the tumour and its microenvironment have given impetus to drug discovery and patient evaluation of interferon-directed strategies. When combined with prior observations or with other effective modalities for cancer treatment, modulation of the interferon system could contribute to further reductions in cancer morbidity and mortality. This Review discusses new interferon-directed therapeutic opportunities, ranging from cyclic dinucleotides to genome methylation inhibitors, angiogenesis inhibitors, chemoradiation, complexes with neoantigen-targeted monoclonal antibodies, combinations with other emerging therapeutic interventions and associations of interferon-stimulated gene expression with patient prognosis - all of which are strategies that have or will soon enter translational clinical evaluation.
Collapse
|
38
|
Andreone S, Spadaro F, Buccione C, Mancini J, Tinari A, Sestili P, Gambardella AR, Lucarini V, Ziccheddu G, Parolini I, Zanetti C, D’Urso MT, De Ninno A, Businaro L, Afferni C, Mattei F, Schiavoni G. IL-33 Promotes CD11b/CD18-Mediated Adhesion of Eosinophils to Cancer Cells and Synapse-Polarized Degranulation Leading to Tumor Cell Killing. Cancers (Basel) 2019; 11:cancers11111664. [PMID: 31717819 PMCID: PMC6895824 DOI: 10.3390/cancers11111664] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 10/15/2019] [Accepted: 10/21/2019] [Indexed: 12/15/2022] Open
Abstract
Eosinophils are major effectors of Th2-related pathologies, frequently found infiltrating several human cancers. We recently showed that eosinophils play an essential role in anti-tumor responses mediated by immunotherapy with the ‘alarmin’ intereukin-33 (IL-33) in melanoma mouse models. Here, we analyzed the mechanisms by which IL-33 mediates tumor infiltration and antitumor activities of eosinophils. We show that IL-33 recruits eosinophils indirectly, via stimulation of tumor cell-derived chemokines, while it activates eosinophils directly, up-regulating CD69, the adhesion molecules ICAM-1 and CD11b/CD18, and the degranulation marker CD63. In co-culture experiments with four different tumor cell lines, IL-33-activated eosinophils established large numbers of stable cell conjugates with target tumor cells, with the polarization of eosinophil effector proteins (ECP, EPX, and granzyme-B) and CD11b/CD18 to immune synapses, resulting in efficient contact-dependent degranulation and tumor cell killing. In tumor-bearing mice, IL-33 induced substantial accumulation of degranulating eosinophils within tumor necrotic areas, indicating cytotoxic activity in vivo. Blocking of CD11b/CD18 signaling significantly reduced IL-33-activated eosinophils’ binding and subsequent killing of tumor cells, indicating a crucial role for this integrin in triggering degranulation. Our findings provide novel mechanistic insights for eosinophil-mediated anti-tumoral function driven by IL-33. Treatments enabling tumor infiltration and proper activation of eosinophils may improve therapeutic response in cancer patients.
Collapse
Affiliation(s)
- Sara Andreone
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (S.A.); (C.B.); (J.M.); (A.R.G.); (V.L.); (G.Z.); (I.P.); (C.Z.); (M.T.D.); (F.M.)
| | - Francesca Spadaro
- Microscopy Unit, Core Facilities, Istituto Superiore di Sanità, 00161 Rome, Italy; (F.S.); (P.S.)
| | - Carla Buccione
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (S.A.); (C.B.); (J.M.); (A.R.G.); (V.L.); (G.Z.); (I.P.); (C.Z.); (M.T.D.); (F.M.)
| | - Jacopo Mancini
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (S.A.); (C.B.); (J.M.); (A.R.G.); (V.L.); (G.Z.); (I.P.); (C.Z.); (M.T.D.); (F.M.)
| | - Antonella Tinari
- Center for Gender Medicine, Istituto Superiore di Sanità, 00161, Rome, Italy;
| | - Paola Sestili
- Microscopy Unit, Core Facilities, Istituto Superiore di Sanità, 00161 Rome, Italy; (F.S.); (P.S.)
| | - Adriana Rosa Gambardella
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (S.A.); (C.B.); (J.M.); (A.R.G.); (V.L.); (G.Z.); (I.P.); (C.Z.); (M.T.D.); (F.M.)
| | - Valeria Lucarini
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (S.A.); (C.B.); (J.M.); (A.R.G.); (V.L.); (G.Z.); (I.P.); (C.Z.); (M.T.D.); (F.M.)
| | - Giovanna Ziccheddu
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (S.A.); (C.B.); (J.M.); (A.R.G.); (V.L.); (G.Z.); (I.P.); (C.Z.); (M.T.D.); (F.M.)
| | - Isabella Parolini
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (S.A.); (C.B.); (J.M.); (A.R.G.); (V.L.); (G.Z.); (I.P.); (C.Z.); (M.T.D.); (F.M.)
| | - Cristiana Zanetti
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (S.A.); (C.B.); (J.M.); (A.R.G.); (V.L.); (G.Z.); (I.P.); (C.Z.); (M.T.D.); (F.M.)
| | - Maria Teresa D’Urso
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (S.A.); (C.B.); (J.M.); (A.R.G.); (V.L.); (G.Z.); (I.P.); (C.Z.); (M.T.D.); (F.M.)
| | - Adele De Ninno
- Institute for Photonics and Nanotechnologies, National Research Council (CNR), 00156 Rome, Italy; (A.D.N.); (L.B.)
| | - Luca Businaro
- Institute for Photonics and Nanotechnologies, National Research Council (CNR), 00156 Rome, Italy; (A.D.N.); (L.B.)
| | - Claudia Afferni
- National Center for Drug Research and Evaluation, Istituto Superiore di Sanità, 00161 Rome, Italy;
| | - Fabrizio Mattei
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (S.A.); (C.B.); (J.M.); (A.R.G.); (V.L.); (G.Z.); (I.P.); (C.Z.); (M.T.D.); (F.M.)
| | - Giovanna Schiavoni
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (S.A.); (C.B.); (J.M.); (A.R.G.); (V.L.); (G.Z.); (I.P.); (C.Z.); (M.T.D.); (F.M.)
- Correspondence: ; Tel.: +39-0649906099
| |
Collapse
|
39
|
Mencattini A, De Ninno A, Mancini J, Businaro L, Martinelli E, Schiavoni G, Mattei F. High-throughput analysis of cell-cell crosstalk in ad hoc designed microfluidic chips for oncoimmunology applications. Methods Enzymol 2019; 632:479-502. [PMID: 32000911 DOI: 10.1016/bs.mie.2019.06.012] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Understanding the interactions between immune and cancer cells occurring within the tumor microenvironment is a prerequisite for successful and personalized anti-cancer therapies. Microfluidic devices, coupled to advanced microscopy systems and automated analytical tools, can represent an innovative approach for high-throughput investigations on immune cell-cancer interactions. In order to study such interactions and to evaluate how therapeutic agents can affect this crosstalk, we employed two ad hoc fabricated microfluidic platforms reproducing advanced 2D or 3D tumor immune microenvironments. In the first type of chip, we confronted the capacity of tumor cells embedded in Matrigel containing one drug or Matrigel containing a combination of two drugs to attract differentially immune cells, by fluorescence microscopy analyses. In the second chip, we investigated the migratory/interaction response of naïve immune cells to danger signals emanated from tumor cells treated with an immunogenic drug, by time-lapse microscopy and automated tracking analysis. We demonstrate that microfluidic platforms and their associated high-throughput computed analyses can represent versatile and smart systems to: (i) monitor and quantify the recruitment and interactions of the immune cells with cancer in a controlled environment, (ii) evaluate the immunogenic effects of anti-cancer therapeutic agents and (iii) evaluate the immunogenic efficacy of combinatorial regimens with respect to single agents.
Collapse
Affiliation(s)
- Arianna Mencattini
- Department of Electronic Engineering, University of Rome Tor Vergata, Rome, Italy
| | - Adele De Ninno
- Institute for Photonics and Nanotechnology, Italian National Research Council, Rome, Italy; Department of Civil Engineering and Computer Science, University of Rome Tor Vergata, Rome, Italy
| | - Jacopo Mancini
- Department of Oncology and Molecular Medicine, Tumor Immunology Unit, Istituto Superiore di Sanità, Rome, Italy
| | - Luca Businaro
- Institute for Photonics and Nanotechnology, Italian National Research Council, Rome, Italy
| | - Eugenio Martinelli
- Department of Electronic Engineering, University of Rome Tor Vergata, Rome, Italy
| | - Giovanna Schiavoni
- Department of Oncology and Molecular Medicine, Tumor Immunology Unit, Istituto Superiore di Sanità, Rome, Italy.
| | - Fabrizio Mattei
- Department of Oncology and Molecular Medicine, Tumor Immunology Unit, Istituto Superiore di Sanità, Rome, Italy.
| |
Collapse
|
40
|
Al-Samadi A, Poor B, Tuomainen K, Liu V, Hyytiäinen A, Suleymanova I, Mesimaki K, Wilkman T, Mäkitie A, Saavalainen P, Salo T. In vitro humanized 3D microfluidic chip for testing personalized immunotherapeutics for head and neck cancer patients. Exp Cell Res 2019; 383:111508. [PMID: 31356815 DOI: 10.1016/j.yexcr.2019.111508] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Revised: 07/18/2019] [Accepted: 07/22/2019] [Indexed: 02/07/2023]
Abstract
OBJECTIVES Immunotherapy and personalized medicine therapeutics are emerging as promising approaches in the management of head and neck squamous cell carcinoma (HNSCC). In spite of that, there is yet no assay that could predict individual response to immunotherapy. METHODS We manufactured an in vitro 3D microfluidic chip to test the efficacy of immunotherapy. The assay was first tested using a tongue cancer cell line (HSC-3) embedded in a human tumour-derived matrix "Myogel/fibrin" and immune cells from three healthy donors. Next, the chips were used with freshly isolated cancer cells, patients' serum and immune cells. Chips were loaded with different immune checkpoint inhibitors, PD-L1 antibody and IDO 1 inhibitor. Migration of immune cells towards cancer cells and the cancer cell proliferation rate were evaluated. RESULTS Immune cell migration towards HSC-3 cells was cancer cell density dependent. IDO 1 inhibitor induced immune cells to migrate towards cancer cells both in HSC-3 and in two HNSCC patient samples. Efficacy of PD-L1 antibody and IDO 1 inhibitor was patient dependent. CONCLUSION We introduced the first humanized in vitro microfluidic chip assay to test immunotherapeutic drugs against HNSCC patient samples. This assay could be used to predict the efficacy of immunotherapeutic drugs for individual patients.
Collapse
Affiliation(s)
- Ahmed Al-Samadi
- Department of Oral and Maxillofacial Diseases, Clinicum, Faculty of Medicine, University of Helsinki, Helsinki, Finland; Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland.
| | - Benedek Poor
- Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland; Department of Medical and Clinical Genetics, Medicum, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Katja Tuomainen
- Department of Oral and Maxillofacial Diseases, Clinicum, Faculty of Medicine, University of Helsinki, Helsinki, Finland; Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Ville Liu
- Department of Oral and Maxillofacial Diseases, Clinicum, Faculty of Medicine, University of Helsinki, Helsinki, Finland; Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Aini Hyytiäinen
- Department of Oral and Maxillofacial Diseases, Clinicum, Faculty of Medicine, University of Helsinki, Helsinki, Finland; Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Ilida Suleymanova
- Department of Oral and Maxillofacial Diseases, Clinicum, Faculty of Medicine, University of Helsinki, Helsinki, Finland; Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Karri Mesimaki
- Department of Oral and Maxillofacial Surgery, HUS Helsinki University Hospital, Finland
| | - Tommy Wilkman
- Department of Oral and Maxillofacial Surgery, HUS Helsinki University Hospital, Finland
| | - Antti Mäkitie
- Department of Otorhinolaryngology - Head and Neck Surgery, HUS Helsinki University Hospital and University of Helsinki, Helsinki, Finland; Division of Ear, Nose and Throat Diseases, Department of Clinical Sciences, Intervention and Technology, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden; Research Program in Systems Oncology, Faculty of Medicine, University of Helsinki, Finland
| | - Päivi Saavalainen
- Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland; Department of Medical and Clinical Genetics, Medicum, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Tuula Salo
- Department of Oral and Maxillofacial Diseases, Clinicum, Faculty of Medicine, University of Helsinki, Helsinki, Finland; Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland; Cancer and Translational Medicine Research Unit, University of Oulu, Oulu, Finland; Medical Research Centre, Oulu University Hospital, Oulu, Finland; Helsinki University Hospital, Helsinki, Finland
| |
Collapse
|
41
|
Lenvatinib plus anti-PD-1 antibody combination treatment activates CD8+ T cells through reduction of tumor-associated macrophage and activation of the interferon pathway. PLoS One 2019. [PMID: 30811474 DOI: 10.1371/journal.pone.0212513.] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Lenvatinib is a multiple receptor tyrosine kinase inhibitor targeting mainly vascular endothelial growth factor (VEGF) and fibroblast growth factor (FGF) receptors. We investigated the immunomodulatory activities of lenvatinib in the tumor microenvironment and its mechanisms of enhanced antitumor activity when combined with a programmed cell death-1 (PD-1) blockade. Antitumor activity was examined in immunodeficient and immunocompetent mouse tumor models. Single-cell analysis, flow cytometric analysis, and immunohistochemistry were used to analyze immune cell populations and their activation. Gene co-expression network analysis and pathway analysis using RNA sequencing data were used to identify lenvatinib-driven combined activity with anti-PD-1 antibody (anti-PD-1). Lenvatinib showed potent antitumor activity in the immunocompetent tumor microenvironment compared with the immunodeficient tumor microenvironment. Antitumor activity of lenvatinib plus anti-PD-1 was greater than that of either single treatment. Flow cytometric analysis revealed that lenvatinib reduced tumor-associated macrophages (TAMs) and increased the percentage of activated CD8+ T cells secreting interferon (IFN)-γ+ and granzyme B (GzmB). Combination treatment further increased the percentage of T cells, especially CD8+ T cells, among CD45+ cells and increased IFN-γ+ and GzmB+ CD8+ T cells. Transcriptome analyses of tumors resected from treated mice showed that genes specifically regulated by the combination were significantly enriched for type-I IFN signaling. Pretreatment with lenvatinib followed by anti-PD-1 treatment induced significant antitumor activity compared with anti-PD-1 treatment alone. Our findings show that lenvatinib modulates cancer immunity in the tumor microenvironment by reducing TAMs and, when combined with PD-1 blockade, shows enhanced antitumor activity via the IFN signaling pathway. These findings provide a scientific rationale for combination therapy of lenvatinib with PD-1 blockade to improve cancer immunotherapy.
Collapse
|
42
|
Kato Y, Tabata K, Kimura T, Yachie-Kinoshita A, Ozawa Y, Yamada K, Ito J, Tachino S, Hori Y, Matsuki M, Matsuoka Y, Ghosh S, Kitano H, Nomoto K, Matsui J, Funahashi Y. Lenvatinib plus anti-PD-1 antibody combination treatment activates CD8+ T cells through reduction of tumor-associated macrophage and activation of the interferon pathway. PLoS One 2019; 14:e0212513. [PMID: 30811474 PMCID: PMC6392299 DOI: 10.1371/journal.pone.0212513] [Citation(s) in RCA: 346] [Impact Index Per Article: 57.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Accepted: 02/04/2019] [Indexed: 12/13/2022] Open
Abstract
Lenvatinib is a multiple receptor tyrosine kinase inhibitor targeting mainly vascular endothelial growth factor (VEGF) and fibroblast growth factor (FGF) receptors. We investigated the immunomodulatory activities of lenvatinib in the tumor microenvironment and its mechanisms of enhanced antitumor activity when combined with a programmed cell death-1 (PD-1) blockade. Antitumor activity was examined in immunodeficient and immunocompetent mouse tumor models. Single-cell analysis, flow cytometric analysis, and immunohistochemistry were used to analyze immune cell populations and their activation. Gene co-expression network analysis and pathway analysis using RNA sequencing data were used to identify lenvatinib-driven combined activity with anti-PD-1 antibody (anti-PD-1). Lenvatinib showed potent antitumor activity in the immunocompetent tumor microenvironment compared with the immunodeficient tumor microenvironment. Antitumor activity of lenvatinib plus anti-PD-1 was greater than that of either single treatment. Flow cytometric analysis revealed that lenvatinib reduced tumor-associated macrophages (TAMs) and increased the percentage of activated CD8+ T cells secreting interferon (IFN)-γ+ and granzyme B (GzmB). Combination treatment further increased the percentage of T cells, especially CD8+ T cells, among CD45+ cells and increased IFN-γ+ and GzmB+ CD8+ T cells. Transcriptome analyses of tumors resected from treated mice showed that genes specifically regulated by the combination were significantly enriched for type-I IFN signaling. Pretreatment with lenvatinib followed by anti-PD-1 treatment induced significant antitumor activity compared with anti-PD-1 treatment alone. Our findings show that lenvatinib modulates cancer immunity in the tumor microenvironment by reducing TAMs and, when combined with PD-1 blockade, shows enhanced antitumor activity via the IFN signaling pathway. These findings provide a scientific rationale for combination therapy of lenvatinib with PD-1 blockade to improve cancer immunotherapy.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/administration & dosage
- Antineoplastic Agents/administration & dosage
- CD8-Positive T-Lymphocytes/drug effects
- CD8-Positive T-Lymphocytes/immunology
- Cell Line, Tumor
- Gene Expression/drug effects
- Gene Expression/immunology
- Immunologic Factors/administration & dosage
- Interferons/metabolism
- Lymphocyte Activation/drug effects
- Macrophages/drug effects
- Macrophages/immunology
- Melanoma, Experimental/genetics
- Melanoma, Experimental/immunology
- Melanoma, Experimental/therapy
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mice, Nude
- Neoplasms, Experimental/genetics
- Neoplasms, Experimental/immunology
- Neoplasms, Experimental/therapy
- Phenylurea Compounds/administration & dosage
- Programmed Cell Death 1 Receptor/antagonists & inhibitors
- Protein Kinase Inhibitors/administration & dosage
- Quinolines/administration & dosage
- Signal Transduction/drug effects
- Tumor Microenvironment/drug effects
- Tumor Microenvironment/immunology
Collapse
Affiliation(s)
- Yu Kato
- Tsukuba Research Laboratories, Eisai Co., Ltd., Tsukuba, Ibaraki, Japan
| | - Kimiyo Tabata
- Tsukuba Research Laboratories, Eisai Co., Ltd., Tsukuba, Ibaraki, Japan
| | - Takayuki Kimura
- Tsukuba Research Laboratories, Eisai Co., Ltd., Tsukuba, Ibaraki, Japan
| | | | - Yoichi Ozawa
- Tsukuba Research Laboratories, Eisai Co., Ltd., Tsukuba, Ibaraki, Japan
| | - Kazuhiko Yamada
- Tsukuba Research Laboratories, Eisai Co., Ltd., Tsukuba, Ibaraki, Japan
| | - Junichi Ito
- Tsukuba Research Laboratories, Eisai Co., Ltd., Tsukuba, Ibaraki, Japan
| | - Sho Tachino
- Tsukuba Research Laboratories, Eisai Co., Ltd., Tsukuba, Ibaraki, Japan
| | - Yusaku Hori
- Tsukuba Research Laboratories, Eisai Co., Ltd., Tsukuba, Ibaraki, Japan
| | - Masahiro Matsuki
- Tsukuba Research Laboratories, Eisai Co., Ltd., Tsukuba, Ibaraki, Japan
| | | | - Samik Ghosh
- The Systems Biology Institute, Shinagawa, Tokyo, Japan
| | | | - Kenichi Nomoto
- Oncology Business Group, Eisai Inc., Woodcliff Lake, New Jersey, United States of America
| | - Junji Matsui
- Tsukuba Research Laboratories, Eisai Co., Ltd., Tsukuba, Ibaraki, Japan
- Oncology Business Group, Eisai Inc., Woodcliff Lake, New Jersey, United States of America
| | | |
Collapse
|
43
|
Shang M, Soon RH, Lim CT, Khoo BL, Han J. Microfluidic modelling of the tumor microenvironment for anti-cancer drug development. LAB ON A CHIP 2019; 19:369-386. [PMID: 30644496 DOI: 10.1039/c8lc00970h] [Citation(s) in RCA: 152] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
Cancer is the leading cause of death worldwide. The complex and disorganized tumor microenvironment makes it very difficult to treat this disease. The most common in vitro drug screening method now is based on 2D culture models which poorly represent actual tumors. Therefore, many 3D tumor models which are more physiologically relevant have been developed to conduct in vitro drug screening and alleviate this situation. Among all these models, the microfluidic tumor model has the unique advantage of recapitulating the tumor microenvironment in a comparatively easier and representative fashion. While there are many review papers available on the related topic of microfluidic tumor models, in this review we aim to focus more on the possibility of generating "clinically actionable information" from these microfluidic systems, besides scientific insight. Our topics cover the tumor microenvironment, conventional 2D and 3D cultures, animal models, and microfluidic tumor models, emphasizing their link to anti-cancer drug discovery and personalized medicine.
Collapse
Affiliation(s)
- Menglin Shang
- BioSystems and Micromechanics (BioSyM) IRG, Singapore-MIT Alliance for Research and Technology (SMART) Centre, 1, Create Way, Enterprise Wing, 138602, Singapore.
| | | | | | | | | |
Collapse
|
44
|
Desjobert C, Carrier A, Delmas A, Marzese DM, Daunay A, Busato F, Pillon A, Tost J, Riond J, Favre G, Etievant C, Arimondo PB. Demethylation by low-dose 5-aza-2'-deoxycytidine impairs 3D melanoma invasion partially through miR-199a-3p expression revealing the role of this miR in melanoma. Clin Epigenetics 2019; 11:9. [PMID: 30651148 PMCID: PMC6335767 DOI: 10.1186/s13148-018-0600-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Accepted: 12/17/2018] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Efficient treatments against metastatic melanoma dissemination are still lacking. Here, we report that low-cytotoxic concentrations of 5-aza-2'-deoxycytidine, a DNA demethylating agent, prevent in vitro 3D invasiveness of metastatic melanoma cells and reduce lung metastasis formation in vivo. RESULTS We unravelled that this beneficial effect is in part due to MIR-199A2 re-expression by promoter demethylation. Alone, this miR showed an anti-invasive and anti-metastatic effect. Throughout integration of micro-RNA target prediction databases with transcriptomic analysis after 5-aza-2'-deoxycytidine treatments, we found that miR-199a-3p downregulates set of genes significantly involved in invasion/migration processes. In addition, analysis of data from melanoma patients showed a stage- and tissue type-dependent modulation of MIR-199A2 expression by DNA methylation. CONCLUSIONS Thus, our data suggest that epigenetic- and/or miR-based therapeutic strategies can be relevant to limit metastatic dissemination of melanoma.
Collapse
Affiliation(s)
- Cécile Desjobert
- FRE no. 3600 CNRS, Epigenetic Targeting of Cancer (ETaC), Toulouse, France
| | - Arnaud Carrier
- FRE no. 3600 CNRS, Epigenetic Targeting of Cancer (ETaC), Toulouse, France
| | - Audrey Delmas
- Cancer Research Center of Toulouse, CRCT, Toulouse, France
| | - Diego M Marzese
- Department of Translational Molecular Medicine, John Wayne Cancer Institute, Providence Saint John's Health Center, Santa Monica, CA, USA
| | - Antoine Daunay
- Laboratory for Functional Genomics, Fondation Jean Dausset - CEPH, Paris, France
| | - Florence Busato
- Laboratory for Epigenetics and Environment, Centre National de la Recherche en Génomique Humaine, CEA, Evry, France
| | - Arnaud Pillon
- Institut de Recherche Pierre Fabre, CRDPF, Toulouse, France
| | - Jörg Tost
- Laboratory for Epigenetics and Environment, Centre National de la Recherche en Génomique Humaine, CEA, Evry, France
| | - Joëlle Riond
- FRE no. 3600 CNRS, Epigenetic Targeting of Cancer (ETaC), Toulouse, France.,UMR 1037 INSERM/Université Toulouse III, CRCT, Toulouse, France
| | - Gilles Favre
- Cancer Research Center of Toulouse, CRCT, Toulouse, France
| | | | - Paola B Arimondo
- FRE no. 3600 CNRS, Epigenetic Targeting of Cancer (ETaC), Toulouse, France. .,Institut Pasteur CNRS UMR3523, Epigenetic Chemical Biology, Paris, France.
| |
Collapse
|
45
|
Chen P, Chen F, Zhou B. Systematic review and meta-analysis of prevalence of dermatological toxicities associated with vemurafenib treatment in patients with melanoma. Clin Exp Dermatol 2018; 44:243-251. [PMID: 30280426 DOI: 10.1111/ced.13751] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/27/2017] [Indexed: 12/12/2022]
Affiliation(s)
- P. Chen
- Department of Pharmacy; Renmin Hospital of Wuhan University; Wuhan China
| | - F. Chen
- Department of Pharmacy; Dongfeng Hospital; Hubei University of Medicine; Shiyan China
| | - B. Zhou
- Department of Pharmacy; Renmin Hospital of Wuhan University; Wuhan China
- School of Pharmaceutical Sciences; Wuhan University; Wuhan China
| |
Collapse
|
46
|
Frey B, Rückert M, Deloch L, Rühle PF, Derer A, Fietkau R, Gaipl US. Immunomodulation by ionizing radiation-impact for design of radio-immunotherapies and for treatment of inflammatory diseases. Immunol Rev 2018; 280:231-248. [PMID: 29027224 DOI: 10.1111/imr.12572] [Citation(s) in RCA: 132] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Ionizing radiation is often regarded as an element of danger. But, danger responses on the cellular and molecular level are often beneficial with regard to the induction of anti-tumor immunity and for amelioration of inflammation. We outline how in dependence of radiation dose and fraction, radiation itself-and especially in combination with immune modulators-impacts on the innate and adaptive immune system. Focus is set on radiation-induced changes of the tumor cell phenotype and the cellular microenvironment including immunogenic cancer cell death. Mechanisms how anti-tumor immune responses are triggered by radiotherapy in combination with hyperthermia, inhibition of apoptosis, the adjuvant AnnexinA5, or vaccination with high hydrostatic pressure-killed autologous tumor cells are discussed. Building on this, feasible multimodal radio-immunotherapy concepts are reviewed including overcoming immune suppression by immune checkpoint inhibitors and by targeting TGF-β. Since radiation-induced tissue damage, inflammation, and anti-tumor immune responses are interconnected, the impact of lower doses of radiation on amelioration of inflammation is outlined. Closely meshed immune monitoring concepts based on the liquid biopsy blood are suggested for prognosis and prediction of cancer and non-cancer inflammatory diseases. Finally, challenges and visions for the design of cancer radio-immunotherapies and for treatment of benign inflammatory diseases are given.
Collapse
Affiliation(s)
- Benjamin Frey
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Michael Rückert
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Lisa Deloch
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Paul F Rühle
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Anja Derer
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Rainer Fietkau
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Udo S Gaipl
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| |
Collapse
|
47
|
Anti-IL-10-mediated Enhancement of Antitumor Efficacy of a Dendritic Cell-targeting MIP3α-gp100 Vaccine in the B16F10 Mouse Melanoma Model Is Dependent on Type I Interferons. J Immunother 2018; 41:181-189. [PMID: 29334492 PMCID: PMC5891382 DOI: 10.1097/cji.0000000000000212] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
The chemokine MIP3α (CCL20) binds to CCR6 on immature dendritic cells. Vaccines fusing MIP3α to gp100 have been shown to be effective in therapeutically reducing melanoma tumor burden and prolonging survival in a mouse model. Other studies have provided evidence that interleukin-10 (IL-10) neutralizing antibodies (αIL-10) enhance immunologic melanoma therapies by modulating the tolerogenic tumor microenvironment. In the current study, we have utilized the B16F10 syngeneic mouse melanoma model to demonstrate for the first time that a therapy neutralizing IL-10 enhances the antitumor efficacy of a MIP3α-gp100 DNA vaccine, leading to significantly smaller tumors, slower growing tumors, and overall increases in mouse survival. The additive effects of αIL-10 were not shown to be correlated to vaccine-specific tumor-infiltrating lymphocytes (TILs), total TILs, or regulatory T cells. However, we discovered an upregulation of IFNα-4 transcripts in tumors and a correlation of increased plasmacytoid dendritic cell numbers with reduced tumor burden in αIL-10-treated mice. Interferon α receptor knockout (IFNαR1) mice received no benefit from αIL-10 treatment, demonstrating that the additional therapeutic value of αIL-10 is primarily mediated by type I IFNs. Efficient targeting of antigen to immature dendritic cells with a chemokine-fusion vaccine provides an effective anticancer therapeutic. Combining this approach with an IL-10 neutralizing antibody therapy enhances the antitumor efficacy of the therapy in a manner dependent upon the activity of type I IFNs. This combination of a vaccine and immunomodulatory agent provides direction for future optimization of a novel cancer vaccine therapy.
Collapse
|
48
|
Niu C, Li M, Zhu S, Chen Y, Zhou L, Xu D, Li W, Cui J, Liu Y, Chen J. Decitabine Inhibits Gamma Delta T Cell Cytotoxicity by Promoting KIR2DL2/3 Expression. Front Immunol 2018; 9:617. [PMID: 29632540 PMCID: PMC5879086 DOI: 10.3389/fimmu.2018.00617] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Accepted: 03/12/2018] [Indexed: 12/25/2022] Open
Abstract
Gamma delta (γδ) T cells, which possess potent cytotoxicity against a wide range of cancer cells, have become a potential avenue for adoptive immunotherapy. Decitabine (DAC) has been reported to enhance the immunogenicity of tumor cells, thereby reinstating endogenous immune recognition and tumor lysis. However, DAC has also been demonstrated to have direct effects on immune cells. In this study, we report that DAC inhibits γδ T cell proliferation. In addition, DAC increases the number of KIR2DL2/3-positive γδ T cells, which are less cytotoxic than the KIR2DL2/3-negative γδ T cells. We found that DAC upregulated KIR2DL2/3 expression in KIR2DL2/3-negative γδ T cells by inhibiting KIR2DL2/3 promoter methylation, which enhances the binding of KIR2DL2/3 promoter to Sp-1 and activates KIR2DL2/3 gene expression. Our data demonstrated that DAC can inhibit the function of human γδ T cells at both cellular and molecular levels, which confirms and extrapolates the results of previous studies showing that DAC can negatively regulate the function of NK cells and αβ T cells of the immune system.
Collapse
Affiliation(s)
- Chao Niu
- Department of Translational Medicine, The First Hospital of Jilin University, Changchun, China.,Department of Cancer Center, The First Hospital of Jilin University, Changchun, China
| | - Min Li
- Department of Cancer Center, The First Hospital of Jilin University, Changchun, China
| | - Shan Zhu
- Department of Translational Medicine, The First Hospital of Jilin University, Changchun, China
| | - Yongchong Chen
- Department of Cancer Center, The First Hospital of Jilin University, Changchun, China
| | - Lei Zhou
- Department of Cancer Center, The First Hospital of Jilin University, Changchun, China
| | - Dongsheng Xu
- Department of Cancer Center, The First Hospital of Jilin University, Changchun, China
| | - Wei Li
- Department of Cancer Center, The First Hospital of Jilin University, Changchun, China
| | - Jiuwei Cui
- Department of Cancer Center, The First Hospital of Jilin University, Changchun, China
| | - Yongjun Liu
- Department of Translational Medicine, The First Hospital of Jilin University, Changchun, China.,Sanofi Research and Development, Cambridge, MA, United States
| | - Jingtao Chen
- Department of Translational Medicine, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
49
|
Bai J, Gao Z, Li X, Dong L, Han W, Nie J. Regulation of PD-1/PD-L1 pathway and resistance to PD-1/PD-L1 blockade. Oncotarget 2017; 8:110693-110707. [PMID: 29299180 PMCID: PMC5746415 DOI: 10.18632/oncotarget.22690] [Citation(s) in RCA: 112] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2017] [Accepted: 11/08/2017] [Indexed: 12/31/2022] Open
Abstract
Immune checkpoint blockades, such as inhibitors against programmed death 1 (PD-1) and its ligand (PD-L1), have received extensive attention in the past decade because of their dramatic clinical outcomes in advanced malignancies. However, both primary and acquired resistance becomes one of the major obstacles, which greatly limits the long-lasting effects and wide application of PD-1/PD-L1 blockade therapy. PD-1/PD-L1 both regulates and is regulated by cellular signaling pathways and epigenetic modification, thus inhibiting the proliferation and effector function of T and B cells. The lack of tumor antigens and effective antigen presentation, aberrant activation of oncogenic pathways, mutations in IFN-γ signaling, immunosuppressive tumor microenvironment such as regulatory T cells, myeloid-derived suppressor cells, M2 macrophages, and immunoinhibitory cytokines can lead to resistance to PD-1/PD-L1 blockade. In this review, we describe PD-1 related signaling pathways, essential factors contributing to the resistance of PD-1 blockade, and discuss strategies to increase the efficacy of immunotherapy. Furthermore, we discuss the possibility of combined epigenetic therapy with PD-1 blockade as a potential promising approach for cancer treatment.
Collapse
Affiliation(s)
- Jie Bai
- Department of Molecular Biology and Bio-Therapeutic, School of Life Science, Chinese PLA General Hospital, Beijing 100853, China
| | - Zhitao Gao
- Department of Molecular Biology and Bio-Therapeutic, School of Life Science, Chinese PLA General Hospital, Beijing 100853, China
| | - Xiang Li
- Department of Molecular Biology and Bio-Therapeutic, School of Life Science, Chinese PLA General Hospital, Beijing 100853, China
| | - Liang Dong
- Department of Molecular Biology and Bio-Therapeutic, School of Life Science, Chinese PLA General Hospital, Beijing 100853, China
| | - Weidong Han
- Department of Molecular Biology and Bio-Therapeutic, School of Life Science, Chinese PLA General Hospital, Beijing 100853, China
| | - Jing Nie
- Department of Molecular Biology and Bio-Therapeutic, School of Life Science, Chinese PLA General Hospital, Beijing 100853, China
| |
Collapse
|
50
|
Organs on chip approach: a tool to evaluate cancer -immune cells interactions. Sci Rep 2017; 7:12737. [PMID: 28986543 PMCID: PMC5630614 DOI: 10.1038/s41598-017-13070-3] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Accepted: 07/27/2017] [Indexed: 11/08/2022] Open
Abstract
In this paper we discuss the applicability of numerical descriptors and statistical physics concepts to characterize complex biological systems observed at microscopic level through organ on chip approach. To this end, we employ data collected on a microfluidic platform in which leukocytes can move through suitably built channels toward their target. Leukocyte behavior is recorded by standard time lapse imaging. In particular, we analyze three groups of human peripheral blood mononuclear cells (PBMC): heterozygous mutants (in which only one copy of the FPR1 gene is normal), homozygous mutants (in which both alleles encoding FPR1 are loss-of-function variants) and cells from 'wild type' donors (with normal expression of FPR1). We characterize the migration of these cells providing a quantitative confirmation of the essential role of FPR1 in cancer chemotherapy response. Indeed wild type PBMC perform biased random walks toward chemotherapy-treated cancer cells establishing persistent interactions with them. Conversely, heterozygous mutants present a weaker bias in their motion and homozygous mutants perform rather uncorrelated random walks, both failing to engage with their targets. We next focus on wild type cells and study the interactions of leukocytes with cancerous cells developing a novel heuristic procedure, inspired by Lyapunov stability in dynamical systems.
Collapse
|