51
|
Jiang W, Li T, Guo J, Wang J, Jia L, Shi X, Yang T, Jiao R, Wei X, Feng Z, Tang Q, Ji G. Bispecific c-Met/PD-L1 CAR-T Cells Have Enhanced Therapeutic Effects on Hepatocellular Carcinoma. Front Oncol 2021; 11:546586. [PMID: 33777728 PMCID: PMC7987916 DOI: 10.3389/fonc.2021.546586] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Accepted: 01/25/2021] [Indexed: 12/15/2022] Open
Abstract
T cells expressing chimeric antigen receptors, especially CD19 CAR-T cells have exhibited effective antitumor activities in B cell malignancies, but due to several factors such as antigen escape effects and tumor microenvironment, their curative potential in hepatocellular carcinoma has not been encouraging. To reduce the antigen escape risk of hepatocellular carcinoma, this study was to design and construct a bispecific CAR targeting c-Met and PD-L1. c-Met/PD-L1 CAR-T cells were obtained by lentiviral transfection, and the transfection efficiency was monitored by flow cytometry analysis. LDH release assays were used to elucidate the efficacy of c-Met/PD-L1 CAR-T cells on hepatocellular carcinoma cells. In addition, xenograft models bearing human hepatocellular carcinoma were constructed to detect the antitumor effect of c-Met/PD-L1 CAR-T cells in vivo. The results shown that this bispecific CAR was manufactured successfully, T cells modified with this bispecific CAR demonstrated improved antitumor activities against c-Met and PD-L1 positive hepatocellular carcinoma cells when compared with those of monovalent c-Met CAR-T cells or PD-L1 CAR-T cells but shown no distinct cytotoxicity on hepatocytes in vitro. In vivo experiments shown that c-Met/PD-L1 CAR-T cells significantly inhibited tumor growth and improve survival persistence compared with other groups. These results suggested that the design of single-chain, bi-specific c-Met/PD-L1 CAR-T is more effective than that of monovalent c-Met CAR-T for the treatment of hepatocellular carcinoma., and this bi-specific c-Met/PD-L1 CAR is rational and implementable with current T-cell engineering technology.
Collapse
Affiliation(s)
- Wei Jiang
- Department of Gastroenterology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China
| | - Tao Li
- Department of Gastroenterology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China
| | - Jiaojiao Guo
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China.,Key Laboratory of Antibody Technique of National Health Commission, Nanjing Medical University, Nanjing, China.,Department of Pathology, Nanjing Medical University, Nanjing, China
| | - Jingjing Wang
- Department of Gastroenterology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China
| | - Lizhou Jia
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China.,Key Laboratory of Antibody Technique of National Health Commission, Nanjing Medical University, Nanjing, China.,Department of Pathology, Nanjing Medical University, Nanjing, China
| | - Xiao Shi
- Department of Gastroenterology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China
| | - Tingting Yang
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China.,Key Laboratory of Antibody Technique of National Health Commission, Nanjing Medical University, Nanjing, China.,Department of Pathology, Nanjing Medical University, Nanjing, China
| | - Ruonan Jiao
- Department of Gastroenterology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China
| | - Xin Wei
- Department of Gastroenterology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China
| | - Zhenqing Feng
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China.,Key Laboratory of Antibody Technique of National Health Commission, Nanjing Medical University, Nanjing, China.,Department of Pathology, Nanjing Medical University, Nanjing, China
| | - Qi Tang
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China.,Key Laboratory of Antibody Technique of National Health Commission, Nanjing Medical University, Nanjing, China.,Department of Pathology, Nanjing Medical University, Nanjing, China
| | - Guozhong Ji
- Department of Gastroenterology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China
| |
Collapse
|
52
|
Pang B, Quan F, Ping Y, Hu J, Lan Y, Pang L. Dissecting the Invasion-Associated Long Non-coding RNAs Using Single-Cell RNA-Seq Data of Glioblastoma. Front Genet 2021; 11:633455. [PMID: 33505440 PMCID: PMC7831882 DOI: 10.3389/fgene.2020.633455] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Accepted: 12/14/2020] [Indexed: 12/25/2022] Open
Abstract
Glioblastoma (GBM) is characterized by rapid and lethal infiltration of brain tissue, which is the primary cause of treatment failure and deaths for GBM. Therefore, understanding the molecular mechanisms of tumor cell invasion is crucial for the treatment of GBM. In this study, we dissected the single-cell RNA-seq data of 3345 cells from four patients and identified dysregulated genes including long non-coding RNAs (lncRNAs), which were involved in the development and progression of GBM. Based on co-expression network analysis, we identified a module (M1) that significantly overlapped with the largest number of dysregulated genes and was confirmed to be associated with GBM invasion by integrating EMT signature, experiment-validated invasive marker and pseudotime trajectory analysis. Further, we denoted invasion-associated lncRNAs which showed significant correlations with M1 and revealed their gradually increased expression levels along the tumor cell invasion trajectory, such as VIM-AS1, WWTR1-AS1, and NEAT1. We also observed the contribution of higher expression of these lncRNAs to poorer survival of GBM patients. These results were mostly recaptured in another validation data of 7930 single cells from 28 GBM patients. Our findings identified lncRNAs that played critical roles in regulating or controlling cell invasion and migration of GBM and provided new insights into the molecular mechanisms underlying GBM invasion as well as potential targets for the treatment of GBM.
Collapse
Affiliation(s)
- Bo Pang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Fei Quan
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Yanyan Ping
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Jing Hu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Yujia Lan
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Lin Pang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| |
Collapse
|
53
|
Li L, Zhu X, Qian Y, Yuan X, Ding Y, Hu D, He X, Wu Y. Chimeric Antigen Receptor T-Cell Therapy in Glioblastoma: Current and Future. Front Immunol 2020; 11:594271. [PMID: 33224149 PMCID: PMC7669545 DOI: 10.3389/fimmu.2020.594271] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 10/12/2020] [Indexed: 02/06/2023] Open
Abstract
Glioblastoma (GBM) is a highly aggressive glioma with an extremely poor prognosis after conventional treatment. Recent advances in immunotherapy offer hope for these patients with incurable GBM. Our present review aimed to provide an overview of immunotherapy for GBM, especially chimeric antigen receptor T-cell (CAR T) therapy. CAR T-cell immunotherapy, which involves the engineering of T cells to kill tumors by targeting cell surface-specific antigens, has been successful in eliminating B-cell leukemia by targeting CD19. IL-13Rα2, EGFRvIII, and HER2-targeted CAR T cells have shown significant clinical efficacy and safety in phase 1 or 2 clinical trials conducted in patients with GBM; these findings support the need for further studies to examine if this therapy can ultimately benefit this patient group. However, local physical barriers, high tumor heterogeneity, and antigen escape make the use of CAR T therapy, as a treatment for GBM, challenging. The potential directions for improving the efficacy of CAR T in GBM are to combine the existing traditional therapies and the construction of multi-target CAR T cells.
Collapse
MESH Headings
- Animals
- Antigens, Neoplasm/genetics
- Antigens, Neoplasm/immunology
- Brain Neoplasms/etiology
- Brain Neoplasms/therapy
- Combined Modality Therapy/methods
- Genetic Engineering
- Glioblastoma/etiology
- Glioblastoma/therapy
- Humans
- Immunotherapy, Adoptive/methods
- Immunotherapy, Adoptive/trends
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/immunology
- Receptors, Antigen, T-Cell/metabolism
- Receptors, Chimeric Antigen/immunology
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- Treatment Outcome
Collapse
Affiliation(s)
- Long Li
- Department of Oncology, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Xiqun Zhu
- Department of Surgical Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yu Qian
- Department of Medical Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiangling Yuan
- Department of Oncology, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Yi Ding
- Department of Radiation Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Desheng Hu
- Department of Radiation Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xin He
- Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Yuan Wu
- Department of Radiation Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
54
|
Ponterio E, De Maria R, Haas TL. Identification of Targets to Redirect CAR T Cells in Glioblastoma and Colorectal Cancer: An Arduous Venture. Front Immunol 2020; 11:565631. [PMID: 33101285 PMCID: PMC7555836 DOI: 10.3389/fimmu.2020.565631] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Accepted: 08/25/2020] [Indexed: 12/11/2022] Open
Abstract
The chimeric antigen receptor (CAR) is an artificial molecule engineered to induce cytolytic T cell reactions in tumors. Generally, this molecule combines an extracellular single-chain variable fragment (scFv) able to recognize tumor-associated epitopes together with the intracellular signaling domains that are required for T cell activation. When expressed by T cells, the CAR enables the recognition and subsequent destruction of cancer cells expressing the complementary antigen on their surface. Although the clinical application for CAR T cells is currently limited to some hematological malignancies, researchers are trying to develop CAR T cell-based therapies for the treatment of solid tumors. However, while in the case of CD19, or other targets restricted to the hematopoietic compartment, the toxicity is limited and manageable, the scarcity of specific antigens expressed by solid tumors and not by healthy cells from vital organs makes the clinical development of CAR T cells in this context particularly challenging. Here we summarize relevant research and clinical trials conducted to redirect CAR T cells to surface antigens in solid tumors and cancer stem cells with a focus on colorectal cancer and glioblastoma. Finally, we will discuss current knowledge of altered glycosylation of CSCs and cancer cells and how these novel epitopes may help to target CAR T cell-based immunotherapy in the future.
Collapse
Affiliation(s)
- Eleonora Ponterio
- Fondazione Policlinico Universitario "A. Gemelli" -Istituti di Ricovero e Cura a Carattere Scientifico, Rome, Italy.,Istituto di Patologia Generale, Università Cattolica del Sacro Cuore Rome, Rome, Italy
| | - Ruggero De Maria
- Fondazione Policlinico Universitario "A. Gemelli" -Istituti di Ricovero e Cura a Carattere Scientifico, Rome, Italy.,Istituto di Patologia Generale, Università Cattolica del Sacro Cuore Rome, Rome, Italy
| | - Tobias Longin Haas
- Istituto di Patologia Generale, Università Cattolica del Sacro Cuore Rome, Rome, Italy.,IIGM-Italian Institute for Genomic Medicine, IRCCS, Candiolo, Italy.,Candiolo Cancer Institute, Fondazione del Piemonte per l'Oncologia-Istituti di Ricovero e Cura a Carattere Scientifico, Candiolo, Italy
| |
Collapse
|
55
|
T Cells Expressing NKG2D CAR with a DAP12 Signaling Domain Stimulate Lower Cytokine Production While Effective in Tumor Eradication. Mol Ther 2020; 29:75-85. [PMID: 32956627 DOI: 10.1016/j.ymthe.2020.08.016] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 07/08/2020] [Accepted: 08/21/2020] [Indexed: 12/11/2022] Open
Abstract
Cytokine-related toxicity associated with the use of highly active chimeric antigen receptor T cells (CAR-T cells) is a significant clinical problem. By fusing the natural killer group 2D (NKG2D) ectodomain to 4-1BB and the DAP12 cytoplasmic domain containing only one immunoreceptor tyrosine-based activation motif, we have developed a 2nd-generation (2nd-Gen) NKG2D CAR for stable expression in human T cells. When compared to T cells modified with NKG2D CAR containing the commonly used CD3ζ activation domain, T cells expressing the NKG2D-DAP12 CAR stimulated lower level release of interferon gamma (IFN-γ), tumor necrosis factor alpha (TNF-α), and interleukin (IL)-2 during tumor cell lysis and their proliferative activity was lower upon repeated antigen stimulation, although no difference between the two CARs was observed in mediating in vitro tumor cell lysis. In tumor-bearing NSG mice, both types of CAR-T cells displayed similar anti-tumor activity, being able to completely eradicate established solid tumor xenografts. However, treatment with the NKG2D-CD3ζ CAR-T cells led to the death of most mice from xenogeneic graft versus host disease starting 30 days post-CAR-T cell injection, which was associated with a higher level of cytokine release, whereas all the mice treated with the NKG2D-DAP12 CAR-T cells survived well. Thus, the incorporation of the DAP12 activation domain in a CAR design may possibly provide a potential clinical advantage in mitigating the risk of cytokine release syndrome (CRS).
Collapse
|
56
|
Obajdin J, Davies DM, Maher J. Engineering of chimeric natural killer cell receptors to develop precision adoptive immunotherapies for cancer. Clin Exp Immunol 2020; 202:11-27. [PMID: 32544282 PMCID: PMC7488126 DOI: 10.1111/cei.13478] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 06/04/2020] [Accepted: 06/07/2020] [Indexed: 12/15/2022] Open
Abstract
Natural killer (NK) cells are innate immune effectors which play a crucial role in recognizing and eliminating virally infected and cancerous cells. They effectively distinguish between healthy and distressed self through the integration of signals delivered by germline‐encoded activating and inhibitory cell surface receptors. The frequent up‐regulation of stress markers on genetically unstable cancer cells has prompted the development of novel immunotherapies that exploit such innate receptors. One prominent example entails the development of chimeric antigen receptors (CAR) that detect cell surface ligands bound by NK receptors, coupling this engagement to the delivery of tailored immune activating signals. Here, we review strategies to engineer CARs in which specificity is conferred by natural killer group 2D (NKG2D) or other NK receptor types. Multiple preclinical studies have demonstrated the remarkable ability of chimeric NK receptor‐targeted T cells and NK cells to effectively and specifically eliminate cancer cells and to reject established tumour burdens. Importantly, such systems act not only acutely but, in some cases, they also incite immunological memory. Moreover, CARs targeted with the NKG2D ligand binding domain have also been shown to disrupt the tumour microenvironment, through the targeting of suppressive T regulatory cells, myeloid‐derived suppressor cells and tumour vasculature. Collectively, these findings have led to the initiation of early‐phase clinical trials evaluating both autologous and allogeneic NKG2D‐targeted CAR T cells in the haematological and solid tumour settings.
Collapse
Affiliation(s)
- J Obajdin
- School of Cancer and Pharmaceutical Sciences, CAR Mechanics Laboratory, Guy's Cancer Centre, King's College London, London, UK
| | - D M Davies
- School of Cancer and Pharmaceutical Sciences, CAR Mechanics Laboratory, Guy's Cancer Centre, King's College London, London, UK
| | - J Maher
- School of Cancer and Pharmaceutical Sciences, CAR Mechanics Laboratory, Guy's Cancer Centre, King's College London, London, UK.,Department of Clinical Immunology and Allergy, King's College Hospital NHS Foundation Trust, London, UK.,Department of Immunology, Eastbourne Hospital, Eastbourne, UK.,Leucid Bio Ltd, Guy's Hospital, London, UK
| |
Collapse
|
57
|
Salinas RD, Durgin JS, O'Rourke DM. Potential of Glioblastoma-Targeted Chimeric Antigen Receptor (CAR) T-Cell Therapy. CNS Drugs 2020; 34:127-145. [PMID: 31916100 DOI: 10.1007/s40263-019-00687-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Despite the established efficacy of chimeric antigen receptor (CAR) T-cell therapy in hematologic malignancies, translating CAR T therapy to solid tumors has remained investigational. Glioblastoma, the most aggressive and lethal form of primary brain tumor, has recently been among the malignancies being trialed clinically with CAR T cells. Glioblastoma in particular holds several unique features that have hindered clinical translation, including its vast intertumoral and intratumoral heterogeneity, associated immunosuppressive environment, and lack of clear experimental models to predict response and analyze resistant phenotypes. Here, we review the history of CAR T therapy development, its current progress in treating glioblastoma, as well as the current challenges and future directions in establishing CAR T therapy as a viable alternative to the current standard of care. Tremendous efforts are currently ongoing to identify novel CAR targets and target combinations for glioblastoma, to modify T cells to enhance their efficacy and to enable them to resist tumor-mediated immunosuppression, and to utilize adjunct therapies such as lymphodepletion, checkpoint inhibition, and bi-specific engagers to improve CAR T persistence. Furthermore, new preclinical models of CAR T therapy are being developed that better reflect the clinical features seen in human trials. Current clinical trials that rapidly incorporate key preclinical findings to patient translation are emerging.
Collapse
Affiliation(s)
- Ryan D Salinas
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Joseph S Durgin
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Donald M O'Rourke
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA. .,Glioblastoma Translational Center of Excellence, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| |
Collapse
|
58
|
Quaglino E, Conti L, Cavallo F. Breast cancer stem cell antigens as targets for immunotherapy. Semin Immunol 2020; 47:101386. [PMID: 31932198 DOI: 10.1016/j.smim.2020.101386] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Accepted: 01/01/2020] [Indexed: 12/13/2022]
Abstract
The great success of immunotherapy is paving the way for a new era in cancer treatment and is driving major improvements in the therapy of patients suffering from a range of solid tumors. However, the choice of the appropriate tumor antigens to be targeted with cancer vaccines and T-cell therapies is still a challenge. Most antigens targeted so far have been identified on the tumor bulk and are expressed on differentiated cancer cells. The discovery of a small population of cancer stem cells (CSC), which is refractory to most current therapies and responsible for the development of metastasis and recurrence, has made it clear that the ideal targets for immunotherapies are the antigens that are expressed in CSC and play a key role in their function. Indeed, their immunotargeting would enable the eradication of CSC to be performed, thus eliminating the tumor source. We call these antigens "CSC oncoantigens". Herein, we summarize the controversial nature of breast CSC, discuss why they represent good candidates for cancer immunotherapy, and review the CSC antigens that have been used as targets for CSC immunotargeting this far. Moreover, we describe the pipeline that we have developed for the identification of fresh CSC oncoantigens, and present the pre-clinical results obtained with vaccines that target some of these antigens.
Collapse
Affiliation(s)
- Elena Quaglino
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy.
| | - Laura Conti
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy.
| | - Federica Cavallo
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy.
| |
Collapse
|
59
|
Ravindran S, Rasool S, Maccalli C. The Cross Talk between Cancer Stem Cells/Cancer Initiating Cells and Tumor Microenvironment: The Missing Piece of the Puzzle for the Efficient Targeting of these Cells with Immunotherapy. CANCER MICROENVIRONMENT 2019; 12:133-148. [PMID: 31758404 PMCID: PMC6937350 DOI: 10.1007/s12307-019-00233-1] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/02/2019] [Accepted: 10/17/2019] [Indexed: 12/14/2022]
Abstract
Cancer Stem Cells/Cancer Initiating Cells (CSCs/CICs) is a rare sub-population within a tumor that is responsible for tumor formation, progression and resistance to therapies. The interaction between CSCs/CICs and tumor microenvironment (TME) can sustain “stemness” properties and promote their survival and plasticity. This cross-talk is also pivotal in regulating and modulating CSC/CIC properties. This review will provide an overview of the mechanisms underlying the mutual interaction between CSCs/CICs and TME. Particular focus will be dedicated to the immunological profile of CSCs/CICs and its role in orchestrating cancer immunosurveillance. Moreover, the available immunotherapy strategies that can target CSCs/CICs and of their possible implementation will be discussed. Overall, the dissection of the mechanisms regulating the CSC/CIC-TME interaction is warranted to understand the plasticity and immunoregulatory properties of stem-like tumor cells and to achieve complete eradications of tumors through the optimization of immunotherapy.
Collapse
Affiliation(s)
- Shilpa Ravindran
- Research Department, Sidra Medicine, Al Luqta Street, PO Box 26999, Doha, Qatar
| | - Saad Rasool
- Research Department, Sidra Medicine, Al Luqta Street, PO Box 26999, Doha, Qatar
| | - Cristina Maccalli
- Research Department, Sidra Medicine, Al Luqta Street, PO Box 26999, Doha, Qatar.
| |
Collapse
|
60
|
Giannone G, Attademo L, Scotto G, Genta S, Ghisoni E, Tuninetti V, Aglietta M, Pignata S, Valabrega G. Endometrial Cancer Stem Cells: Role, Characterization and Therapeutic Implications. Cancers (Basel) 2019; 11:E1820. [PMID: 31752447 PMCID: PMC6896186 DOI: 10.3390/cancers11111820] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 11/08/2019] [Accepted: 11/13/2019] [Indexed: 02/06/2023] Open
Abstract
Endometrial cancer (EC) is the most frequent gynecological cancer. In patients with relapsed and advanced disease, prognosis is still dismal and development of resistance is common. In this context, endometrial Cancer Stem Cells (eCSC), stem-like cells capable to self-renewal and differentiation in mature cancer cells, represent a potential field of expansion for drug development. The aim of this review is to characterize the role of eCSC in EC, their features and how they could be targeted. CSC are involved in progression, invasiveness and metastasis (though epithelial to mesenchimal transition, EMT), as well as chemoresistance in EC. Nevertheless, isolation of eCSC is still controversial. Indeed, CD133, Aldheyde dehydrogenase (ALDH), CD117, CD55 and CD44 are enriched in CSCs but there is no universal marker nowadays. The most frequently activated pathways in eCSC are Wingless-INT (Wnt)/β-catenin, Notch1, and Hedghog, with a high expression of self-renewal transcription factors like Octamer binding transcription factor 4 (OCT), B Lymphoma Mo-MLV Insertion Region 1 Homolog (BMI1), North American Network Operations Group Homebox protein (NANOG), and SRY-Box 2 (SOX2). These pathways have been targeted with selective drugs alone or in combination with chemotherapy and immunotherapy. Unfortunately, although preclinical results are encouraging, few clinical data are available.
Collapse
Affiliation(s)
- Gaia Giannone
- Department of Oncology, University of Torino, 10124 Torino, Italy; (G.S.); (S.G.); (E.G.); (V.T.); (M.A.); (G.V.)
- Candiolo Cancer Institute, FPO - IRCCS - Str. Prov.le 142, km. 3,95, 10060 Candiolo (TO), Italy
| | - Laura Attademo
- Department of Urology and Gynecology, Istituto Nazionale Tumori IRCCS Fondazione G. Pascale Napoli, 80131 Napoli, Italy; (L.A.); (S.P.)
| | - Giulia Scotto
- Department of Oncology, University of Torino, 10124 Torino, Italy; (G.S.); (S.G.); (E.G.); (V.T.); (M.A.); (G.V.)
- Candiolo Cancer Institute, FPO - IRCCS - Str. Prov.le 142, km. 3,95, 10060 Candiolo (TO), Italy
| | - Sofia Genta
- Department of Oncology, University of Torino, 10124 Torino, Italy; (G.S.); (S.G.); (E.G.); (V.T.); (M.A.); (G.V.)
- Candiolo Cancer Institute, FPO - IRCCS - Str. Prov.le 142, km. 3,95, 10060 Candiolo (TO), Italy
| | - Eleonora Ghisoni
- Department of Oncology, University of Torino, 10124 Torino, Italy; (G.S.); (S.G.); (E.G.); (V.T.); (M.A.); (G.V.)
- Candiolo Cancer Institute, FPO - IRCCS - Str. Prov.le 142, km. 3,95, 10060 Candiolo (TO), Italy
| | - Valentina Tuninetti
- Department of Oncology, University of Torino, 10124 Torino, Italy; (G.S.); (S.G.); (E.G.); (V.T.); (M.A.); (G.V.)
- Candiolo Cancer Institute, FPO - IRCCS - Str. Prov.le 142, km. 3,95, 10060 Candiolo (TO), Italy
| | - Massimo Aglietta
- Department of Oncology, University of Torino, 10124 Torino, Italy; (G.S.); (S.G.); (E.G.); (V.T.); (M.A.); (G.V.)
- Candiolo Cancer Institute, FPO - IRCCS - Str. Prov.le 142, km. 3,95, 10060 Candiolo (TO), Italy
| | - Sandro Pignata
- Department of Urology and Gynecology, Istituto Nazionale Tumori IRCCS Fondazione G. Pascale Napoli, 80131 Napoli, Italy; (L.A.); (S.P.)
| | - Giorgio Valabrega
- Department of Oncology, University of Torino, 10124 Torino, Italy; (G.S.); (S.G.); (E.G.); (V.T.); (M.A.); (G.V.)
- Candiolo Cancer Institute, FPO - IRCCS - Str. Prov.le 142, km. 3,95, 10060 Candiolo (TO), Italy
| |
Collapse
|