1
|
Sato S, Rancourt A, Satoh MS. Cell fate simulation reveals cancer cell features in the tumor microenvironment. J Biol Chem 2024; 300:107697. [PMID: 39173950 PMCID: PMC11419826 DOI: 10.1016/j.jbc.2024.107697] [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: 02/28/2024] [Revised: 07/26/2024] [Accepted: 08/06/2024] [Indexed: 08/24/2024] Open
Abstract
To elucidate the dynamic evolution of cancer cell characteristics within the tumor microenvironment (TME), we developed an integrative approach combining single-cell tracking, cell fate simulation, and 3D TME modeling. We began our investigation by analyzing the spatiotemporal behavior of individual cancer cells in cultured pancreatic (MiaPaCa2) and cervical (HeLa) cancer cell lines, with a focus on the α2-6 sialic acid (α2-6Sia) modification on glycans, which is associated with cell stemness. Our findings revealed that MiaPaCa2 cells exhibited significantly higher levels of α2-6Sia modification, correlating with enhanced reproductive capabilities, whereas HeLa cells showed less prevalence of this modification. To accommodate the in vivo variability of α2-6Sia levels, we employed a cell fate simulation algorithm that digitally generates cell populations based on our observed data while varying the level of sialylation, thereby simulating cell growth patterns. Subsequently, we performed a 3D TME simulation with these deduced cell populations, considering the microenvironment that could impact cancer cell growth. Immune cell landscape information derived from 193 cervical and 172 pancreatic cancer cases was used to estimate the degree of the positive or negative impact. Our analysis suggests that the deduced cells generated based on the characteristics of MiaPaCa2 cells are less influenced by the immune cell landscape within the TME compared to those of HeLa cells, highlighting that the fate of cancer cells is shaped by both the surrounding immune landscape and the intrinsic characteristics of the cancer cells.
Collapse
Affiliation(s)
- Sachiko Sato
- Glycobiology and Bioimaging Laboratory of Research Center for Infectious Diseases and Axe of Infectious and Immunological Diseases, Research Centre of CHU de Quebec, Faculty of Medicine, Laval University, Quebec, Canada
| | - Ann Rancourt
- Glycobiology and Bioimaging Laboratory of Research Center for Infectious Diseases and Axe of Infectious and Immunological Diseases, Research Centre of CHU de Quebec, Faculty of Medicine, Laval University, Quebec, Canada; Laboratory of DNA Damage Responses and Bioimaging, Research Centre of CHU de Quebec, Faculty of Medicine, Laval University, Quebec, Canada
| | - Masahiko S Satoh
- Laboratory of DNA Damage Responses and Bioimaging, Research Centre of CHU de Quebec, Faculty of Medicine, Laval University, Quebec, Canada.
| |
Collapse
|
2
|
Farrell R, Pascuzzi N, Chen YL, Kim M, Torres M, Gollahon L, Chen KHE. Prolactin Drives Iron Release from Macrophages and Uptake in Mammary Cancer Cells through CD44. Int J Mol Sci 2024; 25:8941. [PMID: 39201626 PMCID: PMC11354873 DOI: 10.3390/ijms25168941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Revised: 08/13/2024] [Accepted: 08/15/2024] [Indexed: 09/02/2024] Open
Abstract
Iron is an essential element for human health. In humans, dysregulated iron homeostasis can result in a variety of disorders and the development of cancers. Enhanced uptake, redistribution, and retention of iron in cancer cells have been suggested as an "iron addiction" pattern in cancer cells. This increased iron in cancer cells positively correlates with rapid tumor growth and the epithelial-to-mesenchymal transition, which forms the basis for tumor metastasis. However, the source of iron and the mechanisms cancer cells adopt to actively acquire iron is not well understood. In the present study, we report, for the first time, that the peptide hormone, prolactin, exhibits a novel function in regulating iron distribution, on top of its well-known pro-lactating role. When stimulated by prolactin, breast cancer cells increase CD44, a surface receptor mediating the endocytosis of hyaluronate-bound iron, resulting in the accumulation of iron in cancer cells. In contrast, macrophages, when treated by prolactin, express more ferroportin, the only iron exporter in cells, giving rise to net iron output. Interestingly, when co-culturing macrophages with pre-stained labile iron pools and cancer cells without any iron staining, in an iron free condition, we demonstrate direct iron flow from macrophages to cancer cells. As macrophages are one of the major iron-storage cells and it is known that macrophages infiltrate tumors and facilitate their progression, our work therefore presents a novel regulatory role of prolactin to drive iron flow, which provides new information on fine-tuning immune responses in tumor microenvironment and could potentially benefit the development of novel therapeutics.
Collapse
Affiliation(s)
- Reagan Farrell
- Department of Biological Sciences, Texas Tech University, Lubbock, TX 79409, USA; (R.F.); (N.P.); (M.T.); (L.G.)
| | - Nicholas Pascuzzi
- Department of Biological Sciences, Texas Tech University, Lubbock, TX 79409, USA; (R.F.); (N.P.); (M.T.); (L.G.)
| | - Yi-Ling Chen
- Department of Electronic Engineering, National Kaohsiung University of Science and Technology, Kaohsiung 80778, Taiwan
| | - Mary Kim
- Department of Biological Sciences, Texas Tech University, Lubbock, TX 79409, USA; (R.F.); (N.P.); (M.T.); (L.G.)
| | - Miguel Torres
- Department of Biological Sciences, Texas Tech University, Lubbock, TX 79409, USA; (R.F.); (N.P.); (M.T.); (L.G.)
| | - Lauren Gollahon
- Department of Biological Sciences, Texas Tech University, Lubbock, TX 79409, USA; (R.F.); (N.P.); (M.T.); (L.G.)
| | - Kuan-Hui Ethan Chen
- Department of Biological Sciences, Texas Tech University, Lubbock, TX 79409, USA; (R.F.); (N.P.); (M.T.); (L.G.)
| |
Collapse
|
3
|
Bakhtiyaridovvombaygi M, Yazdanparast S, Kheyrandish S, Safdari SM, Amiri Samani F, Sohani M, Jaafarian AS, Damirchiloo F, Izadpanah A, Parkhideh S, Mikanik F, Roshandel E, Hajifathali A, Gharehbaghian A. Harnessing natural killer cells for refractory/relapsed non-Hodgkin lymphoma: biological roles, clinical trials, and future prospective. Biomark Res 2024; 12:66. [PMID: 39020411 PMCID: PMC11253502 DOI: 10.1186/s40364-024-00610-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 06/28/2024] [Indexed: 07/19/2024] Open
Abstract
Non-Hodgkin lymphomas (NHLs) are heterogeneous and are among the most common hematological malignancies worldwide. Despite the advances in the treatment of patients with NHLs, relapse or resistance to treatment is anticipated in several patients. Therefore, novel therapeutic approaches are needed. Recently, natural killer (NK) cell-based immunotherapy alone or in combination with monoclonal antibodies, chimeric antigen receptors, or bispecific killer engagers have been applied in many investigations for NHL treatment. The functional defects of NK cells and the ability of cancerous cells to escape NK cell-mediated cytotoxicity within the tumor microenvironment of NHLs, as well as the beneficial results from previous studies in the context of NK cell-based immunotherapy in NHLs, direct our attention to this therapeutic strategy. This review aims to summarize clinical studies focusing on the applications of NK cells in the immunotherapy of patients with NHL.
Collapse
Affiliation(s)
- Mehdi Bakhtiyaridovvombaygi
- Student Research Committee, Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Hematopoietic Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Somayeh Yazdanparast
- Department of Hematology and Blood Banking, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Setare Kheyrandish
- Student Research Committee, Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Seyed Mehrab Safdari
- Departments of Hematology and Blood Transfusion, School of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Fateme Amiri Samani
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Iranian Blood Transfusion Organization (IBTO), Tehran, Iran
| | - Mahsa Sohani
- Student Research Committee, Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Akram Sadat Jaafarian
- Departments of Hematology and Blood Transfusion, School of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Fateme Damirchiloo
- Departments of Hematology and Blood Transfusion, School of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Amirhossein Izadpanah
- Hematopoietic Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sahar Parkhideh
- Hematopoietic Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fatemeh Mikanik
- Laboratory Hematology and Blood Bank Department, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Elham Roshandel
- Hematopoietic Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Abbas Hajifathali
- Hematopoietic Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Ahmad Gharehbaghian
- Laboratory Hematology and Blood Bank Department, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
- Pediatric Congenital Hematologic Disorders Research Center, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
4
|
Shi H, Sun X, Wu Y, Cui Q, Sun S, Ji N, Liu Y. Targeting the tumor microenvironment in primary central nervous system lymphoma: Implications for prognosis. J Clin Neurosci 2024; 124:36-46. [PMID: 38642434 DOI: 10.1016/j.jocn.2024.04.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 03/06/2024] [Accepted: 04/07/2024] [Indexed: 04/22/2024]
Abstract
Primary central nervous system lymphoma (PCNSL) is a rare extranodal non-Hodgkin lymphoma, and there is limited research on its tumor microenvironment (TME). Nevertheless, more and more studies have evidence that TME has essential effects on tumor cell proliferation, immune escape, and drug resistance. Thus, it is critical to elucidate the role of TME in PCNSL. The understanding of the PCNSL TME is gradually unfolding, including factors that distinguish it from systemic diffuse large B-cell lymphoma (DLBCL). The TME in PCNSL exhibits both transcriptional and spatial intratumor heterogeneity. Cellular interactions between tumor cells and stroma cells reveal immune evasion signaling. The comparative analysis between PCNSL and DLBCL suggests that PCNSL is more likely to be an immunologically deficient tumor. In PCNSL, T cell exhaustion and downregulation of macrophage immune function are accompanied by suppressive microenvironmental factors such as M2 polarized macrophages, endothelin B receptor, HLA depletion, PD-L1, and TIM-3. MMP-9, Integrin-β1, and ICAM-1/LFA-1 play crucial roles in transendothelial migration towards the CNS, while CXCL13/CXCR5, CD44, MAG, and IL-8 are essential for brain parenchymal invasion. Further, macrophages, YKL-40, CD31, CD105, PD-1/PD-L1 axis, osteopontin, galectin-3, aggregative perivascular tumor cells, and HLA deletion may contribute to poor outcomes in patients with PCNSL. This article reviews the effect of various components of TME on the progression and prognosis of PCNSL patients to identify novel therapeutic targets.
Collapse
Affiliation(s)
- Han Shi
- Department of Hematology, Beijing Tiantan Hospital, Capital Medical University, 100070 Beijing, China
| | - Xuefei Sun
- Department of Hematology, Beijing Tiantan Hospital, Capital Medical University, 100070 Beijing, China
| | - Yuchen Wu
- Department of Hematology, Beijing Tiantan Hospital, Capital Medical University, 100070 Beijing, China
| | - Qu Cui
- Department of Hematology, Beijing Tiantan Hospital, Capital Medical University, 100070 Beijing, China
| | - Shengjun Sun
- Department of Hematology, Beijing Tiantan Hospital, Capital Medical University, 100070 Beijing, China
| | - Nan Ji
- Department of Hematology, Beijing Tiantan Hospital, Capital Medical University, 100070 Beijing, China
| | - Yuanbo Liu
- Department of Hematology, Beijing Tiantan Hospital, Capital Medical University, 100070 Beijing, China.
| |
Collapse
|
5
|
Farris F, Elhagh A, Vigorito I, Alongi N, Pisati F, Giannattasio M, Casagrande F, Veghini L, Corbo V, Tripodo C, Di Napoli A, Matafora V, Bachi A. Unveiling the mechanistic link between extracellular amyloid fibrils, mechano-signaling and YAP activation in cancer. Cell Death Dis 2024; 15:28. [PMID: 38199984 PMCID: PMC10781709 DOI: 10.1038/s41419-024-06424-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 12/20/2023] [Accepted: 01/02/2024] [Indexed: 01/12/2024]
Abstract
The tumor microenvironment is a complex ecosystem that plays a critical role in cancer progression and treatment response. Recently, extracellular amyloid fibrils have emerged as novel components of the tumor microenvironment; however, their function remains elusive. In this study, we establish a direct connection between the presence of amyloid fibrils in the secretome and the activation of YAP, a transcriptional co-activator involved in cancer proliferation and drug resistance. Furthermore, we uncover a shared mechano-signaling mechanism triggered by amyloid fibrils in both melanoma and pancreatic ductal adenocarcinoma cells. Our findings highlight the crucial role of the glycocalyx protein Agrin which binds to extracellular amyloid fibrils and acts as a necessary factor in driving amyloid-dependent YAP activation. Additionally, we reveal the involvement of the HIPPO pathway core kinase LATS1 in this signaling cascade. Finally, we demonstrate that extracellular amyloid fibrils enhance cancer cell migration and invasion. In conclusion, our research expands our knowledge of the tumor microenvironment by uncovering the role of extracellular amyloid fibrils in driving mechano-signaling and YAP activation. This knowledge opens up new avenues for developing innovative strategies to modulate YAP activation and mitigate its detrimental effects during cancer progression.
Collapse
Affiliation(s)
- Francesco Farris
- IFOM ETS - The AIRC Institute of Molecular Oncology, 20139, Milan, Italy
| | - Alice Elhagh
- IFOM ETS - The AIRC Institute of Molecular Oncology, 20139, Milan, Italy
| | - Ilaria Vigorito
- IFOM ETS - The AIRC Institute of Molecular Oncology, 20139, Milan, Italy
| | - Nicoletta Alongi
- IFOM ETS - The AIRC Institute of Molecular Oncology, 20139, Milan, Italy
- IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Federica Pisati
- Histopathology Unit, Cogentech S.C.a.R.L, 20139, Milan, Italy
| | - Michele Giannattasio
- IFOM ETS - The AIRC Institute of Molecular Oncology, 20139, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, 20122, Milan, Italy
| | - Francesca Casagrande
- IFOM ETS - The AIRC Institute of Molecular Oncology, 20139, Milan, Italy
- Human Technopole, Milan, Italy
| | - Lisa Veghini
- Department of Engineering for Innovation Medicine (DIMI), University of Verona, 37134, Verona, Italy
| | - Vincenzo Corbo
- Department of Engineering for Innovation Medicine (DIMI), University of Verona, 37134, Verona, Italy
- ARC-Net Centre for Applied Research on Cancer, University of Verona, 37134, Verona, Italy
| | - Claudio Tripodo
- IFOM ETS - The AIRC Institute of Molecular Oncology, 20139, Milan, Italy
- Tumor Immunology Unit, Department of Health Sciences, University of Palermo, 90133, Palermo, Italy
| | - Arianna Di Napoli
- Pathology Unit, Department of Clinical and Molecular Medicine, Sant'Andrea University Hospital, Sapienza University of Rome, 00189, Rome, Italy
| | - Vittoria Matafora
- IFOM ETS - The AIRC Institute of Molecular Oncology, 20139, Milan, Italy.
| | - Angela Bachi
- IFOM ETS - The AIRC Institute of Molecular Oncology, 20139, Milan, Italy.
| |
Collapse
|
6
|
Yadav M, Uikey BN, Rathore SS, Gupta P, Kashyap D, Kumar C, Shukla D, Vijayamahantesh, Chandel AS, Ahirwar B, Singh AK, Suman SS, Priyadarshi A, Amit A. Role of cytokine in malignant T-cell metabolism and subsequent alternation in T-cell tumor microenvironment. Front Oncol 2023; 13:1235711. [PMID: 37746258 PMCID: PMC10513393 DOI: 10.3389/fonc.2023.1235711] [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: 06/06/2023] [Accepted: 08/14/2023] [Indexed: 09/26/2023] Open
Abstract
T cells are an important component of adaptive immunity and T-cell-derived lymphomas are very complex due to many functional sub-types and functional elasticity of T-cells. As with other tumors, tissues specific factors are crucial in the development of T-cell lymphomas. In addition to neoplastic cells, T- cell lymphomas consist of a tumor micro-environment composed of normal cells and stroma. Numerous studies established the qualitative and quantitative differences between the tumor microenvironment and normal cell surroundings. Interaction between the various component of the tumor microenvironment is crucial since tumor cells can change the microenvironment and vice versa. In normal T-cell development, T-cells must respond to various stimulants deferentially and during these courses of adaptation. T-cells undergo various metabolic alterations. From the stage of quiescence to attention of fully active form T-cells undergoes various stage in terms of metabolic activity. Predominantly quiescent T-cells have ATP-generating metabolism while during the proliferative stage, their metabolism tilted towards the growth-promoting pathways. In addition to this, a functionally different subset of T-cells requires to activate the different metabolic pathways, and consequently, this regulation of the metabolic pathway control activation and function of T-cells. So, it is obvious that dynamic, and well-regulated metabolic pathways are important for the normal functioning of T-cells and their interaction with the microenvironment. There are various cell signaling mechanisms of metabolism are involved in this regulation and more and more studies have suggested the involvement of additional signaling in the development of the overall metabolic phenotype of T cells. These important signaling mediators include cytokines and hormones. The impact and role of these mediators especially the cytokines on the interplay between T-cell metabolism and the interaction of T-cells with their micro-environments in the context of T-cells lymphomas are discussed in this review article.
Collapse
Affiliation(s)
- Megha Yadav
- Department of Forensic Science, Guru Ghasidas Vishwavidyalaya, Bilaspur, India
| | - Blessi N. Uikey
- Department of Forensic Science, Guru Ghasidas Vishwavidyalaya, Bilaspur, India
| | | | - Priyanka Gupta
- Department of Forensic Science, Guru Ghasidas Vishwavidyalaya, Bilaspur, India
| | - Diksha Kashyap
- Department of Forensic Science, Guru Ghasidas Vishwavidyalaya, Bilaspur, India
| | - Chanchal Kumar
- Department of Forensic Science, Guru Ghasidas Vishwavidyalaya, Bilaspur, India
| | - Dhananjay Shukla
- Department of Biotechnology, Guru Ghasidas Vishwavidyalaya, Bilaspur, India
| | - Vijayamahantesh
- Department of Immunology and Microbiology, University of Missouri, Columbia, SC, United States
| | - Arvind Singh Chandel
- Center for Disease Biology and Integrative Medicine, Faculty of Medicine, The University of Tokyo, Bunkyo, Japan
| | - Bharti Ahirwar
- Department of Pharmacy, Guru Ghasidas Vishwavidyalaya, Bilaspur, India
| | | | - Shashi Shekhar Suman
- Department of Zoology, Udayana Charya (UR) College, Lalit Narayan Mithila University, Darbhanga, India
| | - Amit Priyadarshi
- Department of Zoology, Veer Kunwar Singh University, Arrah, India
| | - Ajay Amit
- Department of Forensic Science, Guru Ghasidas Vishwavidyalaya, Bilaspur, India
| |
Collapse
|
7
|
Naik A, Leask A. Tumor-Associated Fibrosis Impairs the Response to Immunotherapy. Matrix Biol 2023; 119:125-140. [PMID: 37080324 DOI: 10.1016/j.matbio.2023.04.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 04/16/2023] [Accepted: 04/17/2023] [Indexed: 04/22/2023]
Abstract
Previously, impaired responses to immunotherapy in cancer had been attributed mainly to inherent tumor characteristics (tumor cell intrinsic factors) such as low immunogenicity, (low) mutational burden, weak host immune system, etc. However, mapping the responses of immunotherapeutic regimes in clinical trials for different types of cancer has pointed towards an obvious commonality - that tumors with a rich fibrotic stroma respond poorly or not at all. This has prompted a harder look on tumor cell extrinsic factors such as the surrounding tumor microenvironment (TME), and specifically, the fibrotic stroma as a potential enabler of immunotherapy failure. Indeed, the role of cancer-associated fibrosis in impeding efficacy of immunotherapy is now well-established. In fact, recent studies reveal a complex interconnection between fibrosis and treatment efficacy. Accordingly, in this review we provide a general overview of what a tumor associated fibrotic reaction is and how it interacts with the members of immune system that are frequently seen to be modulated in a failed immunotherapeutic regime.
Collapse
Affiliation(s)
- Angha Naik
- University of Saskatchewan, College of Dentistry, 105 Wiggins Road, Saskatoon, SK, Canada
| | - Andrew Leask
- University of Saskatchewan, College of Dentistry, 105 Wiggins Road, Saskatoon, SK, Canada.
| |
Collapse
|
8
|
Moeckel C, Bakhl K, Georgakopoulos-Soares I, Zaravinos A. The Efficacy of Tumor Mutation Burden as a Biomarker of Response to Immune Checkpoint Inhibitors. Int J Mol Sci 2023; 24:ijms24076710. [PMID: 37047684 PMCID: PMC10095310 DOI: 10.3390/ijms24076710] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 03/21/2023] [Accepted: 03/31/2023] [Indexed: 04/14/2023] Open
Abstract
Cancer is one of the leading causes of death in the world; therefore, extensive research has been dedicated to exploring potential therapeutics, including immune checkpoint inhibitors (ICIs). Initially, programmed-death ligand-1 was the biomarker utilized to predict the efficacy of ICIs. However, its heterogeneous expression in the tumor microenvironment, which is critical to cancer progression, promoted the exploration of the tumor mutation burden (TMB). Research in various cancers, such as melanoma and lung cancer, has shown an association between high TMB and response to ICIs, increasing its predictive value. However, the TMB has failed to predict ICI response in numerous other cancers. Therefore, future research is needed to analyze the variations between cancer types and establish TMB cutoffs in order to create a more standardized methodology for using the TMB clinically. In this review, we aim to explore current research on the efficacy of the TMB as a biomarker, discuss current approaches to overcoming immunoresistance to ICIs, and highlight new trends in the field such as liquid biopsies, next generation sequencing, chimeric antigen receptor T-cell therapy, and personalized tumor vaccines.
Collapse
Affiliation(s)
- Camille Moeckel
- Department of Biochemistry and Molecular Biology, Institute for Personalized Medicine, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Katrina Bakhl
- Department of Biochemistry and Molecular Biology, Institute for Personalized Medicine, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Ilias Georgakopoulos-Soares
- Department of Biochemistry and Molecular Biology, Institute for Personalized Medicine, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Apostolos Zaravinos
- Department of Life Sciences, European University Cyprus, Diogenis Str., 6, Nicosia 2404, Cyprus
- Cancer Genetics, Genomics and Systems Biology Laboratory, Basic and Translational Cancer Research Center (BTCRC), Nicosia 1516, Cyprus
| |
Collapse
|
9
|
Baude J, Limagne E, Ladjohounlou R, Mirjolet C. Combining radiotherapy and NK cell-based therapies: The time has come. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2023; 378:31-60. [PMID: 37438020 DOI: 10.1016/bs.ircmb.2023.02.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/14/2023]
Abstract
Natural killer (NK) cells are innate lymphoid cells that play an essential role in the anti-tumor response through immunosurveillance, multiple mechanisms of cytotoxicity and the synthesis of cytokines modulating the immune tumor microenvironment (TME). After the dramatic advances in immunotherapy targeting T cells including the success of checkpoint inhibitors or autologous chimeric antigen receptor (CAR) expressing T cells in clinical practice, NK cells have gained growing interest for the development of new therapies. Although NK cells have shown promising responses in leukemia patients, the effects of NK-targeted therapies are currently limited in the treatment of solid tumors. Thus, radiotherapy could provide a valuable solution to improve treatments targeting NK cells. Indeed, ionizing radiations represent a powerful immuno-modulator that can either induce a pro-inflammatory and anti-tumor TME, or conversely lead to immunosuppression of effector immune cells in favor of tumor growth and therapeutic escape, depending on how it is delivered and tumor models. However, the effects of ionizing radiation on NK cells are only partially understood. Therefore, we review the effects of radiotherapy on the NK cell-mediated anti-tumor response, and propose potential strategies to reinvigorate NK cells by combining radiotherapy with NK cell-targeted therapies.
Collapse
Affiliation(s)
- Jérémy Baude
- Radiation Oncology Department, Preclinical Radiation Therapy and Radiobiology Unit, Centre Georges-François Leclerc, Unicancer, Dijon, France
| | - Emeric Limagne
- TIReCS Team, UMR INSERM 1231, Dijon, France; Cancer Biology Transfer Platform, Centre Georges-François Leclerc, Equipe Labellisée Ligue Contre le Cancer, Dijon, France; University of Bourgogne Franche-Comté, Dijon, France
| | - Riad Ladjohounlou
- Radiation Oncology Department, Preclinical Radiation Therapy and Radiobiology Unit, Centre Georges-François Leclerc, Unicancer, Dijon, France; TIReCS Team, UMR INSERM 1231, Dijon, France
| | - Céline Mirjolet
- Radiation Oncology Department, Preclinical Radiation Therapy and Radiobiology Unit, Centre Georges-François Leclerc, Unicancer, Dijon, France; TIReCS Team, UMR INSERM 1231, Dijon, France.
| |
Collapse
|
10
|
Monoclonal antibodies in breast cancer: A critical appraisal. Crit Rev Oncol Hematol 2023; 183:103915. [PMID: 36702424 DOI: 10.1016/j.critrevonc.2023.103915] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 12/01/2022] [Accepted: 01/20/2023] [Indexed: 01/24/2023] Open
Abstract
In breast cancer, mAbs can play multifunctional roles like targeting cancer cells, sometimes directly attacking them, helping in locating and delivering therapeutic drugs to targets, inhibiting cell growth and blocking immune system inhibitors, etc. Monoclonal antibodies are also one of the important successful treatment strategies especially against HER2 but they have not been explored much for other types of breast cancers especially in triple negative breast cancers. Monoclonal antibodies impact the feasibility of antigen specificity, bispecific and trispecific mAbs have opened new doors for more targeted specific efficacy. Monoclonal antibodies can be used diversly and with efficacy as compared to other methods of treatment thus maining it a suitable candidate for breast cancer treatment. However, mAbs treatment also causes various side effects such as fever, trembling, fatigue, headache and muscle pain, nausea/vomiting, difficulty in breathing, rashes and bleeding. Understanding the pros and cons of this strategy, we have explored in this review, the current and future potential capabilities of monoclonal antibodies with respect to diagnosis and treatment of breast cancer. DATA AVAILABILITY: Not applicable.
Collapse
|
11
|
Schiebout C, Lust HE, Huang YH, Frost HR. Cell type-specific Interaction Analysis using Doublets in scRNA-seq (CIcADA). BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.13.528326. [PMID: 36824707 PMCID: PMC9949061 DOI: 10.1101/2023.02.13.528326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/16/2023]
Abstract
Motivation Doublets are usually considered an unwanted artifact of single-cell RNA-sequencing (scRNA-seq) and are only identified in datasets for the sake of removal. However, if cells have a juxtacrine attachment to one another in situ and maintain this association through an scRNA-seq processing pipeline that only partially dissociates the tissue, these doublets can provide meaningful biological information regarding the interactions and cell processes occurring in the analyzed tissue. This is especially true for cases such as the immune compartment of the tumor microenvironment, where the frequency and type of immune cell juxtacrine interactions can be a prognostic indicator. Results We developed Cell type-specific Interaction Analysis using Doublets in scRNA-seq (CIcADA) as a pipeline for identifying and analyzing biological doublets in scRNA-seq data. CIcADA identifies putative doublets using multi-label cell type scores and characterizes interaction dynamics through a comparison against synthetic doublets of the same cell type composition. In performing CIcADA on several scRNA-seq tumor datasets, we found that the identified doublets were consistently upregulating expression of immune response genes. Contact Courtney.T.Schiebout.GR@Dartmouth.edu , Hildreth.R.Frost@Dartmouth.edu.
Collapse
Affiliation(s)
- Courtney Schiebout
- Department of Biomedical Data Science, Geisel School of Medicine, Dartmouth College, Hanover, NH USA
| | | | - Yina H Huang
- Department of Microbiology and Immunology, Geisel School of Medicine, Dartmouth College, Hanover, NH USA
| | - H Robert Frost
- Department of Biomedical Data Science, Geisel School of Medicine, Dartmouth College, Hanover, NH USA
| |
Collapse
|
12
|
Xie Y, Wang M, Sun Q, Wang D, Li C. Recent Advances in Tetrakis (4‐Carboxyphenyl) Porphyrin‐Based Nanocomposites for Tumor Therapy. ADVANCED NANOBIOMED RESEARCH 2022. [DOI: 10.1002/anbr.202200136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Affiliation(s)
- Yulin Xie
- Institute of Molecular Sciences and Engineering Institute of Frontier and Interdisciplinary Science Shandong University Qingdao 266237 P.R. China
| | - Man Wang
- Institute of Molecular Sciences and Engineering Institute of Frontier and Interdisciplinary Science Shandong University Qingdao 266237 P.R. China
| | - Qianqian Sun
- Institute of Molecular Sciences and Engineering Institute of Frontier and Interdisciplinary Science Shandong University Qingdao 266237 P.R. China
| | - Dongmei Wang
- Key Laboratory of the Ministry of Education for Advanced Catalysis Materials College of Chemistry and Life Sciences Zhejiang Normal University Jinhua 321004 P.R. China
| | - Chunxia Li
- Institute of Molecular Sciences and Engineering Institute of Frontier and Interdisciplinary Science Shandong University Qingdao 266237 P.R. China
| |
Collapse
|
13
|
Wu G, Ruben MD, Francey LJ, Lee YY, Anafi RC, Hogenesch JB. An in silico genome-wide screen for circadian clock strength in human samples. Bioinformatics 2022; 38:5375-5382. [PMID: 36321857 PMCID: PMC9750125 DOI: 10.1093/bioinformatics/btac686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 10/06/2022] [Indexed: 11/16/2022] Open
Abstract
MOTIVATION Years of time-series gene expression studies have built a strong understanding of clock-controlled pathways across species. However, comparatively little is known about how 'non-clock' pathways influence clock function. We need a strong understanding of clock-coupled pathways in human tissues to better appreciate the links between disease and clock function. RESULTS We developed a new computational approach to explore candidate pathways coupled to the clock in human tissues. This method, termed LTM, is an in silico screen to infer genetic influences on circadian clock function. LTM uses natural variation in gene expression in human data and directly links gene expression variation to clock strength independent of longitudinal data. We applied LTM to three human skin and one melanoma datasets and found that the cell cycle is the top candidate clock-coupled pathway in healthy skin. In addition, we applied LTM to thousands of tumor samples from 11 cancer types in the TCGA database and found that extracellular matrix organization-related pathways are tightly associated with the clock strength in humans. Further analysis shows that clock strength in tumor samples is correlated with the proportion of cancer-associated fibroblasts and endothelial cells. Therefore, we show both the power of LTM in predicting clock-coupled pathways and classify factors associated with clock strength in human tissues. AVAILABILITY AND IMPLEMENTATION LTM is available on GitHub (https://github.com/gangwug/LTMR) and figshare (https://figshare.com/articles/software/LTMR/21217604) to facilitate its use. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Gang Wu
- Divisions of Human Genetics and Immunobiology, Center for Circadian Medicine, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Marc D Ruben
- Divisions of Human Genetics and Immunobiology, Center for Circadian Medicine, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Lauren J Francey
- Divisions of Human Genetics and Immunobiology, Center for Circadian Medicine, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Yin Yeng Lee
- Divisions of Human Genetics and Immunobiology, Center for Circadian Medicine, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Ron C Anafi
- Department of Medicine, Chronobiology and Sleep Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | |
Collapse
|
14
|
Mulas O, Mola B, Madeddu C, Caocci G, Macciò A, Nasa GL. Prognostic Role of Cell Blood Count in Chronic Myeloid Neoplasm and Acute Myeloid Leukemia and Its Possible Implications in Hematopoietic Stem Cell Transplantation. Diagnostics (Basel) 2022; 12:2493. [PMID: 36292182 PMCID: PMC9600993 DOI: 10.3390/diagnostics12102493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 10/01/2022] [Accepted: 10/12/2022] [Indexed: 11/17/2022] Open
Abstract
Numerous prognostic indexes have been developed in hematological diseases based on patient characteristics and genetic or molecular assessment. However, less attention was paid to more accessible parameters, such as neutrophils, lymphocytes, monocytes, and platelet counts. Although many studies have defined the role of neutrophil-to-lymphocyte or platelet-to-lymphocyte in lymphoid malignancies, few applications exist for myeloid neoplasm or hematopoietic stem cell transplantation procedures. In this review, we synthesized literature data on the prognostic value of count blood cells in myeloid malignancies and hematopoietic stem cell transplantation in the context of classical prognostic factors and clinical outcomes.
Collapse
Affiliation(s)
- Olga Mulas
- Hematology Unit, Businco Hospital, ARNAS G. Brotzu, 09124 Cagliari, Italy
- Department of Medical Sciences and Public Health, University of Cagliari, S554, km 4500, 09042 Monserrato, Italy
| | - Brunella Mola
- Hematology Unit, Businco Hospital, ARNAS G. Brotzu, 09124 Cagliari, Italy
- Department of Medical Sciences and Public Health, University of Cagliari, S554, km 4500, 09042 Monserrato, Italy
| | - Clelia Madeddu
- Department of Medical Sciences and Public Health, University of Cagliari, S554, km 4500, 09042 Monserrato, Italy
| | - Giovanni Caocci
- Hematology Unit, Businco Hospital, ARNAS G. Brotzu, 09124 Cagliari, Italy
- Department of Medical Sciences and Public Health, University of Cagliari, S554, km 4500, 09042 Monserrato, Italy
| | - Antonio Macciò
- Department of Gynecologic Oncology, Businco Hospital, ARNAS G. Brotzu, 09124 Cagliari, Italy
- Department of Surgical Sciences, University of Cagliari, 09124 Cagliari, Italy
| | - Giorgio La Nasa
- Hematology Unit, Businco Hospital, ARNAS G. Brotzu, 09124 Cagliari, Italy
- Department of Medical Sciences and Public Health, University of Cagliari, S554, km 4500, 09042 Monserrato, Italy
| |
Collapse
|
15
|
Yuan X, Zhou J, Zhou L, Huang Z, Wang W, Qiu J, Yang Q, Zhang C, Ma M. Apoptosis-Related Gene-Mediated Cell Death Pattern Induces Immunosuppression and Immunotherapy Resistance in Gastric Cancer. Front Genet 2022; 13:921163. [PMID: 35865012 PMCID: PMC9295743 DOI: 10.3389/fgene.2022.921163] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 05/23/2022] [Indexed: 11/13/2022] Open
Abstract
Background: Apoptosis is a type of cell death, which can produce abundant mediators to modify the tumor microenvironment. However, relationships between apoptosis, immunosuppression, and immunotherapy resistance of gastric cancer (GC) remain unclear. Methods: Gene expression data and matching clinical information were extracted from TCGA-STAD, GSE84437, GSE34942, GSE15459, GSE57303, ACRG/GSE62254, GSE29272, GSE26253, and IMvigor210 datasets. A consensus clustering analysis based on six apoptosis-related genes (ARGs) was performed to determine the molecular subtypes, and then an apoptosisScore was constructed based on differentially expressed and prognostic genes between molecular subtypes. Estimate R package was utilized to calculate the tumor microenvironment condition. Kaplan-Meier analysis and ROC curves were performed to further confirm the apoptosisScore efficacy. Results: Based on six ARGs, two molecular subgroups with significantly distinct survival and immune cell infiltration were identified. Then, an apoptosisScore was built to quantify the apoptosis index of each GC patient. Next, we investigated the correlations between the clinical characteristics and apoptosisScore using logistic regression. Multivariate Cox analysis shows that low apoptosisScore was an independent predictor of poor overall survival in TCGA and ACRG datasets, and was associated with the higher pathological stage. Meanwhile, low apoptosisScore was associated with higher immune cell, higher ESTIMATEScore, higher immuneScore, higher stromalScore, higher immune checkpoint, and lower tumorpurity, which was consistent with the “immunity tidal model theory”. Importantly, low apoptosisScore was sensitive to immunotherapy. In addition, GSEA indicated that several gene ontology and Kyoto Encyclopedia of Genes and Genomes items associated with apoptosis, several immune-related pathways, and JAK–STAT signal pathway were considerably enriched in the low apoptosisScore phenotype pathway. Conclusion: Our findings propose that low apoptosisScore is a prognostic biomarker, correlated with immune infiltrates, and sensitivity to immunotherapy in GC.
Collapse
Affiliation(s)
- Xiaolu Yuan
- Department of Pathology, Maoming People’s Hospital, Maoming, China
| | - Jun Zhou
- Department of Gastrointestinal Surgery, Maoming People’s Hospital, Maoming, China
| | - Liping Zhou
- Department of Endoscopy Center, The No.6 People’s Hospital of Benxi, Liaoning, China
| | - Zudong Huang
- Department of Gastrointestinal Surgery, Maoming People’s Hospital, Maoming, China
| | - Weiwei Wang
- Department of Gastrointestinal Surgery, Maoming People’s Hospital, Maoming, China
| | - Jiasheng Qiu
- Department of Gastrointestinal Surgery, Maoming People’s Hospital, Maoming, China
| | - Qiangbang Yang
- Department of Gastrointestinal Surgery, Maoming People’s Hospital, Maoming, China
| | - Chaohao Zhang
- Department of Pathology, Maoming People’s Hospital, Maoming, China
| | - MingHui Ma
- Department of Gastrointestinal Surgery, Maoming People’s Hospital, Maoming, China
- *Correspondence: MingHui Ma, ,
| |
Collapse
|
16
|
Ruiz-Martinez A, Gong C, Wang H, Sové RJ, Mi H, Kimko H, Popel AS. Simulations of tumor growth and response to immunotherapy by coupling a spatial agent-based model with a whole-patient quantitative systems pharmacology model. PLoS Comput Biol 2022; 18:e1010254. [PMID: 35867773 PMCID: PMC9348712 DOI: 10.1371/journal.pcbi.1010254] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 08/03/2022] [Accepted: 05/26/2022] [Indexed: 12/23/2022] Open
Abstract
Quantitative systems pharmacology (QSP) models and spatial agent-based models (ABM) are powerful and efficient approaches for the analysis of biological systems and for clinical applications. Although QSP models are becoming essential in discovering predictive biomarkers and developing combination therapies through in silico virtual trials, they are inadequate to capture the spatial heterogeneity and randomness that characterize complex biological systems, and specifically the tumor microenvironment. Here, we extend our recently developed spatial QSP (spQSP) model to analyze tumor growth dynamics and its response to immunotherapy at different spatio-temporal scales. In the model, the tumor spatial dynamics is governed by the ABM, coupled to the QSP model, which includes the following compartments: central (blood system), tumor, tumor-draining lymph node, and peripheral (the rest of the organs and tissues). A dynamic recruitment of T cells and myeloid-derived suppressor cells (MDSC) from the QSP central compartment has been implemented as a function of the spatial distribution of cancer cells. The proposed QSP-ABM coupling methodology enables the spQSP model to perform as a coarse-grained model at the whole-tumor scale and as an agent-based model at the regions of interest (ROIs) scale. Thus, we exploit the spQSP model potential to characterize tumor growth, identify T cell hotspots, and perform qualitative and quantitative descriptions of cell density profiles at the invasive front of the tumor. Additionally, we analyze the effects of immunotherapy at both whole-tumor and ROI scales under different tumor growth and immune response conditions. A digital pathology computational analysis of triple-negative breast cancer specimens is used as a guide for modeling the immuno-architecture of the invasive front.
Collapse
Affiliation(s)
- Alvaro Ruiz-Martinez
- Department of Biomedical Engineering, Johns Hopkins, University School of Medicine, Baltimore, Maryland, United States of America
| | - Chang Gong
- Department of Biomedical Engineering, Johns Hopkins, University School of Medicine, Baltimore, Maryland, United States of America
| | - Hanwen Wang
- Department of Biomedical Engineering, Johns Hopkins, University School of Medicine, Baltimore, Maryland, United States of America
| | - Richard J. Sové
- Department of Biomedical Engineering, Johns Hopkins, University School of Medicine, Baltimore, Maryland, United States of America
| | - Haoyang Mi
- Department of Biomedical Engineering, Johns Hopkins, University School of Medicine, Baltimore, Maryland, United States of America
| | - Holly Kimko
- Clinical Pharmacology & Quantitative Pharmacology, AstraZeneca, Gaithersburg, Maryland, United States of America
| | - Aleksander S. Popel
- Department of Biomedical Engineering, Johns Hopkins, University School of Medicine, Baltimore, Maryland, United States of America
- Department of Oncology and Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland, United States of America
| |
Collapse
|
17
|
Zhao Y, Xu H, Zhang M, Li L. Single-Cell RNA-Seq and Bulk RNA-Seq Reveal Intratumoral Heterogeneity and Tumor Microenvironment Characteristics in Diffuse Large B-Cell Lymphoma. Front Genet 2022; 13:881345. [PMID: 35601491 PMCID: PMC9116505 DOI: 10.3389/fgene.2022.881345] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 04/22/2022] [Indexed: 12/02/2022] Open
Abstract
Background: Diffuse large B-cell lymphoma (DLBCL) is the most common histologic subtype of non-Hodgkin’s lymphoma (NHL) with highly heterogeneous genetic and phenotypic features. Therefore, a comprehensive understanding of cellular diversity and intratumoral heterogeneity is essential to elucidate the mechanisms driving DLBCL progression and to develop new therapeutic approaches. Methods: We analyzed single-cell transcriptomic data from 2 reactive lymph node tissue samples and 2 DLBCL lymph node biopsy tissue samples to explore the transcriptomic landscape of DLBCL. In addition, we constructed a prognostic model based on the genes obtained from differential analysis. Results: Based on gene expression profiles at the single cell level, we identified and characterized different subpopulations of malignant and immune cells. Malignant cells exhibited a high degree of inter-tumor heterogeneity. Tumor-infiltrating regulatory CD4+ T cells showed highly immunosuppressive properties and exhausted cytotoxic CD8+ T cells were highly expressed with markers of exhaustion. Cell communication analysis identified complex interactions between malignant cells and other cell subpopulations. In addition, the prognostic model we constructed allows for monitoring the prognosis of DLBCL patients. Conclusion: This study provides an in-depth dissection of the transcriptional features of malignant B cells and tumor microenvironment (TME) in DLBCL and provides new insights into the tumor heterogeneity of DLBCL.
Collapse
|
18
|
Immunosuppressive Signaling Pathways as Targeted Cancer Therapies. Biomedicines 2022. [DOI: 10.3390/biomedicines10030682
expr 829797163 + 949875436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2023] Open
Abstract
Immune response has been shown to play an important role in defining patient prognosis and response to cancer treatment. Tumor-induced immunosuppression encouraged the recent development of new chemotherapeutic agents that assists in the augmentation of immune responses. Molecular mechanisms that tumors use to evade immunosurveillance are attributed to their ability to alter antigen processing/presentation pathways and the tumor microenvironment. Cancer cells take advantage of normal molecular and immunoregulatory machinery to survive and thrive. Cancer cells constantly adjust their genetic makeup using several mechanisms such as nucleotide excision repair as well as microsatellite and chromosomal instability, thus giving rise to new variants with reduced immunogenicity and the ability to continue to grow without restrictions. This review will focus on the central molecular signaling pathways involved in immunosuppressive cells and briefly discuss how cancer cells evade immunosurveillance by manipulating antigen processing cells and related proteins. Secondly, the review will discuss how these pathways can be utilized for the implementation of precision medicine and deciphering drug resistance.
Collapse
|
19
|
Setlai BP, Hull R, Bida M, Durandt C, Mulaudzi TV, Chatziioannou A, Dlamini Z. Immunosuppressive Signaling Pathways as Targeted Cancer Therapies. Biomedicines 2022; 10:682. [PMID: 35327484 PMCID: PMC8945019 DOI: 10.3390/biomedicines10030682] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 03/08/2022] [Accepted: 03/11/2022] [Indexed: 12/23/2022] Open
Abstract
Immune response has been shown to play an important role in defining patient prognosis and response to cancer treatment. Tumor-induced immunosuppression encouraged the recent development of new chemotherapeutic agents that assists in the augmentation of immune responses. Molecular mechanisms that tumors use to evade immunosurveillance are attributed to their ability to alter antigen processing/presentation pathways and the tumor microenvironment. Cancer cells take advantage of normal molecular and immunoregulatory machinery to survive and thrive. Cancer cells constantly adjust their genetic makeup using several mechanisms such as nucleotide excision repair as well as microsatellite and chromosomal instability, thus giving rise to new variants with reduced immunogenicity and the ability to continue to grow without restrictions. This review will focus on the central molecular signaling pathways involved in immunosuppressive cells and briefly discuss how cancer cells evade immunosurveillance by manipulating antigen processing cells and related proteins. Secondly, the review will discuss how these pathways can be utilized for the implementation of precision medicine and deciphering drug resistance.
Collapse
Affiliation(s)
- Botle Precious Setlai
- Department of Surgery, Faculty of Health Sciences, University of Pretoria, Private Bag X323, Arcadia 0007, South Africa;
| | - Rodney Hull
- SAMRC Precision Oncology Research Unit (PORU), DSI/NRF SARChI Chair in Precision Oncology and Cancer Prevention (POCP), Pan African Cancer Research Institute (PACRI), University of Pretoria, Hatfield 0028, South Africa;
| | - Meshack Bida
- Department of Anatomical Pathology, National Health Laboratory Service (NHLS), University of Pretoria, Hatfield 0028, South Africa;
| | - Chrisna Durandt
- Institute for Cellular and Molecular Medicine, Department of Immunology, SAMRC Extramural Unit for Stem Cell Research and Therapy, Faculty of Health Sciences, University of Pretoria, Pretoria 0001, South Africa;
| | - Thanyani Victor Mulaudzi
- Department of Surgery, Faculty of Health Sciences, University of Pretoria, Private Bag X323, Arcadia 0007, South Africa;
| | - Aristotelis Chatziioannou
- Center of Systems Biology, Biomedical Research Foundation Academy of Athens, 4 Soranou Ephessiou Str., 115 27 Athens, Greece;
| | - Zodwa Dlamini
- SAMRC Precision Oncology Research Unit (PORU), DSI/NRF SARChI Chair in Precision Oncology and Cancer Prevention (POCP), Pan African Cancer Research Institute (PACRI), University of Pretoria, Hatfield 0028, South Africa;
| |
Collapse
|
20
|
Abstract
The therapeutic armamentarium has significantly expanded since the approval of various CD19-targeting chimeric antigen receptor T cell (CAR-T) therapies in non-Hodgkin lymphoma (NHL). These CAR-Ts are patient-specific and require a complex, resource, and time-consuming process. While this appears promising, autologous CAR-Ts are limited due to the lack of accessibility, manufacturing delays, and variable product quality. To overcome these, allogeneic (allo) CARs from healthy donors appear appealing. These can be immediately available as “off the shelf” ready-to-use products of standardized and superior quality exempt from the effects of an immunosuppressive tumor microenvironment and prior treatments, and potentially with lower healthcare utilization using industrialized scale production. Allogeneic CARs, however, are not devoid of complications and require genomic editing, especially with αβ T cells to avoid graft versus host disease (GvHD) and allo-rejection by the recipient’s immune system. Tools for genomic editing such as TALEN and CRISPR provide promise to develop truly “off the shelf” universal CARs and further advance the field of cellular immunotherapy. Several allogeneic CARs are currently in early phase clinical trials, and preliminary data is encouraging. Longer follow-up is required to truly assess the feasibility and safety of these techniques in the patients. This review focuses on the strategies for developing allogeneic CARs along with cell sources and clinical experience thus far in lymphoma.
Collapse
|
21
|
An overview of current therapeutic strategies for glioblastoma and the role of CD73 as an alternative curative approach. Clin Transl Oncol 2021; 24:742-756. [PMID: 34792724 DOI: 10.1007/s12094-021-02732-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 11/01/2021] [Indexed: 10/19/2022]
Abstract
Glioblastoma multiforme (GBM) is a complicated and heterogeneous brain tumor with short-term survival outcomes. Commercial therapies are not practical due to cell infiltration capacity, high proliferative rate, and blood-brain barrier. In this context, recognition of the molecular mechanism of tumor progression might help the development of new cancer therapeutics. Recently, more evidence has supported CD73 and downstream adenosine A2A/A2B receptor signaling playing a crucial role in glioblastoma pathogenesis; therefore, targeting CD73 in murine tumor models can reduce tumor development. CD73 is an ecto-enzyme inducing tumor metastasis, angiogenesis, and immune escape via the production of extracellular adenosine in the tumor microenvironment. In this review, we provided information about clinical characteristics as well as the therapeutic management of glioblastoma. Then, we focused on newly available experimental evidence distinguishing between the essential role of CD73 on this tumor growth and a new method for the treatment of GBM patients.
Collapse
|
22
|
Zhang W, Torres-Rojas C, Yue J, Zhu BM. Adipose-derived stem cells in ovarian cancer progression, metastasis, and chemoresistance. Exp Biol Med (Maywood) 2021; 246:1810-1815. [PMID: 34229470 DOI: 10.1177/15353702211023846] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Ovarian cancer is the deadliest gynecological malignancy due to its symptomless early stage, metastasis, and high recurrence rate. The tumor microenvironment contributes to the ovarian cancer progression, metastasis, and chemoresistance. Adipose-derived stem cell in the tumor microenvironment of ovarian cancer, as a key player, interacts with ovarian cancer cells to form the cancer-associated fibroblasts and cancer-associated adipocytes, and secretes soluble factors to activate tumor cell signaling, which can promote ovarian cancer metastasis and chemoresistance. We summarize in this review the recent progress in the studies of interactions between adipose-derived stem cell and ovarian cancer, thus, to provide some insight for ovarian cancer therapy through targeting adipose-derived stem cell.
Collapse
Affiliation(s)
- Wenjing Zhang
- Department of Genetics, Genomics & Informatics, College of Medicine, the University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Carolina Torres-Rojas
- Department of Genetics, Genomics & Informatics, College of Medicine, the University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Junming Yue
- Department of Pathology and Laboratory Medicine, College of Medicine, the University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Bing-Mei Zhu
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu 610041, People's Republic of China
| |
Collapse
|
23
|
Serrero G. Progranulin/GP88, A Complex and Multifaceted Player of Tumor Growth by Direct Action and via the Tumor Microenvironment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1329:475-498. [PMID: 34664252 DOI: 10.1007/978-3-030-73119-9_22] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Investigation of the role of progranulin/GP88 on the proliferation and survival of a wide variety of cells has been steadily increasing. Several human diseases stem from progranulin dysregulation either through its overexpression in cancer or its absence as in the case of null mutations in some form of frontotemporal dementia. The present review focuses on the role of progranulin/GP88 in cancer development, progression, and drug resistance. Various aspects of progranulin identification, biology, and signaling pathways will be described. Information will be provided about its direct role as an autocrine growth and survival factor and its paracrine effect as a systemic factor as well as via interaction with extracellular matrix proteins and with components of the tumor microenvironment to influence drug resistance, migration, angiogenesis, inflammation, and immune modulation. This chapter will also describe studies examining progranulin/GP88 tumor tissue expression as well as circulating level as a prognostic factor for several cancers. Due to the wealth of publications in progranulin, this review does not attempt to be exhaustive but rather provide a thread to lead the readers toward more in-depth exploration of this fascinating and unique protein.
Collapse
|
24
|
Derbal Y. Modeling Cancer Dynamics. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2020; 2020:2455-2458. [PMID: 33018503 DOI: 10.1109/embc44109.2020.9175155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Cancer is a complex disease that continues to pose formidable challenges to therapeutic interventions. An increased understanding of cancer complexity and in particular tumor growth dynamics is critical to the development of more effective therapies. In this respect, an agent-based model of tumor growth is explored with the consideration of cancer reprograming of metabolism and the immune response, to seek insight about the coupling between these two key determinants of tumor growth dynamics. Ultimately, this exploration is intended to inform the development of therapies that can induce a more effective immune response despite the metabolic constraints of the tumor microenvironment.
Collapse
|
25
|
Briand J, Garnier D, Nadaradjane A, Clément-Colmou K, Potiron V, Supiot S, Bougras-Cartron G, Frenel JS, Heymann D, Vallette FM, Cartron PF. Radiotherapy-induced overexpression of exosomal miRNA-378a-3p in cancer cells limits natural killer cells cytotoxicity. Epigenomics 2020; 12:397-408. [PMID: 32267172 DOI: 10.2217/epi-2019-0193] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Aim: We here hypothesized that tumor-derived exosomal miRNA (TexomiR) released from irradiated tumors may play a role in the tumor cells escape to natural killer (NK) cells. Materials & methods: Our study included the use of different cancer cell lines, blood biopsies of xenograph mice model and patients treated with radiotherapy. Results: The irradiation of cancer cells promotes the TET2-mediated demethylation of miR-378 promoter, miR-378a-3p overexpression and its loading in exosomes, inducing the decrease of granzyme-B (GZMB) secretion by NK cells. An inverse correlation between TexomiR-378a-3p and GZMB was observed in murine and human blood samples. Conclusion: Our work identifies TexomiR-378a-3p as a molecular signature associated with the loss of NK cells cytotoxicity via the decrease of GZMB expression upon radiotherapy.
Collapse
Affiliation(s)
- Joséphine Briand
- CRCINA, INSERM, Université de Nantes, Nantes 44000, France.,LaBCT, Institut de Cancérologie de l'Ouest, Saint Herblain 44800, France.,Cancéropole Grand-Ouest, réseau NET, Nantes 44000, France.,EpiSAVMEN Consortium (Région Pays de la Loire), Nantes 44000, France
| | - Delphine Garnier
- CRCINA, INSERM, Université de Nantes, Nantes 44000, France.,LaBCT, Institut de Cancérologie de l'Ouest, Saint Herblain 44800, France
| | - Arulraj Nadaradjane
- CRCINA, INSERM, Université de Nantes, Nantes 44000, France.,LaBCT, Institut de Cancérologie de l'Ouest, Saint Herblain 44800, France.,Cancéropole Grand-Ouest, réseau NET, Nantes 44000, France.,EpiSAVMEN Consortium (Région Pays de la Loire), Nantes 44000, France
| | - Karen Clément-Colmou
- CRCINA, INSERM, Université de Nantes, Nantes 44000, France.,LaBCT, Institut de Cancérologie de l'Ouest, Saint Herblain 44800, France
| | - Vincent Potiron
- CRCINA, INSERM, Université de Nantes, Nantes 44000, France.,LaBCT, Institut de Cancérologie de l'Ouest, Saint Herblain 44800, France.,EpiSAVMEN Consortium (Région Pays de la Loire), Nantes 44000, France
| | - Stéphane Supiot
- CRCINA, INSERM, Université de Nantes, Nantes 44000, France.,LaBCT, Institut de Cancérologie de l'Ouest, Saint Herblain 44800, France
| | - Gwenola Bougras-Cartron
- CRCINA, INSERM, Université de Nantes, Nantes 44000, France.,LaBCT, Institut de Cancérologie de l'Ouest, Saint Herblain 44800, France.,Cancéropole Grand-Ouest, réseau NET, Nantes 44000, France.,EpiSAVMEN Consortium (Région Pays de la Loire), Nantes 44000, France
| | - Jean-Sébastien Frenel
- CRCINA, INSERM, Université de Nantes, Nantes 44000, France.,LaBCT, Institut de Cancérologie de l'Ouest, Saint Herblain 44800, France
| | - Dominique Heymann
- CRCINA, INSERM, Université de Nantes, Nantes 44000, France.,LaBCT, Institut de Cancérologie de l'Ouest, Saint Herblain 44800, France
| | - François M Vallette
- CRCINA, INSERM, Université de Nantes, Nantes 44000, France.,LaBCT, Institut de Cancérologie de l'Ouest, Saint Herblain 44800, France.,LabEX IGO, Université de Nantes 44000, France
| | - Pierre-François Cartron
- CRCINA, INSERM, Université de Nantes, Nantes 44000, France.,LaBCT, Institut de Cancérologie de l'Ouest, Saint Herblain 44800, France.,Cancéropole Grand-Ouest, réseau NET, Nantes 44000, France.,EpiSAVMEN Consortium (Région Pays de la Loire), Nantes 44000, France.,LabEX IGO, Université de Nantes 44000, France
| |
Collapse
|
26
|
Ribeiro Franco PI, Rodrigues AP, de Menezes LB, Pacheco Miguel M. Tumor microenvironment components: Allies of cancer progression. Pathol Res Pract 2019; 216:152729. [PMID: 31735322 DOI: 10.1016/j.prp.2019.152729] [Citation(s) in RCA: 137] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 11/06/2019] [Accepted: 11/10/2019] [Indexed: 12/12/2022]
Abstract
Cancer is a disease that affects millions of individuals worldwide and has a great impact on public health. Therefore, the study of tumor biology and an understanding of how the components of the tumor microenvironment behave and interact is extremely important for cancer research. Factors expressed by the components of the tumor microenvironment and induce angiogenesis have important roles in the onset and progression of the tumor. These components are represented by the extracellular matrix, fibroblasts, adipocytes, immune cells, and macrophages, besides endothelial cells, which modulate tumor cells and the tumor microenvironment to favor survival and the progression of cancer. The characteristics and function of the main stromal components and their mechanisms of interaction with the tumor cells that contribute to progression, tumor invasion, and tumor spread will be addressed in this review. Furthermore, reviewing these components is expected to indicate their importance as possible prognostic markers and therapeutic targets.
Collapse
Affiliation(s)
- Pablo Igor Ribeiro Franco
- Escola de Veterinária e Zootecnia, Programa de Pós-Graduação em Ciência Animal, Universidade Federal de Goiás, Goiânia, GO, Brazil.
| | - Arthur Perillo Rodrigues
- Escola de Veterinária e Zootecnia, Programa de Pós-Graduação em Ciência Animal, Universidade Federal de Goiás, Goiânia, GO, Brazil
| | | | - Marina Pacheco Miguel
- Escola de Veterinária e Zootecnia, Programa de Pós-Graduação em Ciência Animal, Universidade Federal de Goiás, Goiânia, GO, Brazil
| |
Collapse
|
27
|
Chemoresistance mechanisms of breast cancer and their countermeasures. Biomed Pharmacother 2019; 114:108800. [PMID: 30921705 DOI: 10.1016/j.biopha.2019.108800] [Citation(s) in RCA: 204] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Revised: 03/13/2019] [Accepted: 03/18/2019] [Indexed: 12/12/2022] Open
Abstract
Chemoresistance is one of the major challenges for the breast cancer treatment. Owing to its heterogeneous nature, the chemoresistance mechanisms of breast cancer are complicated, and not been fully elucidated. The existing treatments fall short of offering adequate solution to drug resistance, so more effective approaches are desperately needed to improve existing therapeutic regimens. To overcome this hurdle, a number of strategies are being investigated, such as novel agents or drug carriers and combination treatment. In addition, some new therapeutics including gene therapy and immunotherapy may be promising for dealing with the resistance. In this article, we review the mechanisms of chemoresistance in breast cancer. Furthermore, the potential therapeutic methods to overcome the resistance were discussed.
Collapse
|
28
|
Chimote AA, Hajdu P, Sfyris AM, Gleich BN, Wise-Draper T, Casper KA, Conforti L. Kv1.3 Channels Mark Functionally Competent CD8+ Tumor-Infiltrating Lymphocytes in Head and Neck Cancer. Cancer Res 2016; 77:53-61. [PMID: 27815390 DOI: 10.1158/0008-5472.can-16-2372] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2016] [Revised: 10/14/2016] [Accepted: 10/26/2016] [Indexed: 12/20/2022]
Abstract
Tumor-infiltrating lymphocytes (TIL) are potent mediators of an antitumor response. However, their function is attenuated in solid tumors. CD8+ T-cell effector functions, such as cytokine and granzyme production, depend on cytoplasmic Ca2+, which is controlled by ion channels. In particular, Kv1.3 channels regulate the membrane potential and Ca2+ influx in human effector memory T (TEM) cells. In this study, we assessed the contribution of reduced Kv1.3 and Ca2+ flux on TIL effector function in head and neck cancer (HNC). We obtained tumor samples and matched peripheral blood from 14 patients with HNC. CD3+ TILs were composed of 57% CD4+ (82% TEM and 20% Tregs) and 36% CD8+ cells. Electrophysiology revealed a 70% reduction in functional Kv1.3 channels in TILs as compared with peripheral blood T cells from paired patients, which was accompanied by a decrease in Ca2+ influx. Immunofluorescence analysis showed that CD8+ TILs expressing high Kv1.3 preferentially localized in the stroma. Importantly, high expression of Kv1.3 correlated with high Ki-67 and granzyme B expression. Overall, these data indicate that defective Kv1.3 channels and Ca2+ fluxes in TILs may contribute to reduced immune surveillance in HNC. Cancer Res; 77(1); 53-61. ©2016 AACR.
Collapse
Affiliation(s)
- Ameet A Chimote
- Department of Internal Medicine, Division of Nephrology, University of Cincinnati, Cincinnati, Ohio
| | - Peter Hajdu
- Department of Internal Medicine, Division of Nephrology, University of Cincinnati, Cincinnati, Ohio
| | - Alexandros M Sfyris
- Department of Internal Medicine, Division of Nephrology, University of Cincinnati, Cincinnati, Ohio
| | - Brittany N Gleich
- Department of Internal Medicine, Division of Nephrology, University of Cincinnati, Cincinnati, Ohio
| | - Trisha Wise-Draper
- Department of Internal Medicine, Division of Hematology/Oncology, University of Cincinnati, Cincinnati, Ohio
| | - Keith A Casper
- Department of Otolaryngology, University of Michigan, Ann Arbor, Michigan
| | - Laura Conforti
- Department of Internal Medicine, Division of Nephrology, University of Cincinnati, Cincinnati, Ohio.
| |
Collapse
|
29
|
Zhang Y, Gallastegui N, Rosenblatt JD. Regulatory B cells in anti-tumor immunity. Int Immunol 2015; 27:521-30. [PMID: 25999597 DOI: 10.1093/intimm/dxv034] [Citation(s) in RCA: 86] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2015] [Accepted: 05/18/2015] [Indexed: 12/19/2022] Open
Abstract
Advances in understanding of the immune microenvironment have highlighted the role of immunosuppressive T cell, myeloid, dendritic and monocytic sub-populations in inhibition of the anti-tumor immune response. The role of B cells in modulating the immune response to solid tumors as well as lymphoid malignancies is less well understood. Murine models of autoimmune disease have defined B regulatory cell (Breg) subsets with immune suppressive activity, including B cell subsets that express IL-10, and transforming growth factor-β, which can facilitate T regulatory cell recruitment and expansion. Multiple murine tumor models point to the existence of similar immune suppressive B cell sub-populations that can migrate into tumor deposits and acquire an immune suppressive phenotype, which then leads to attenuation of the local anti-tumor immune response. Other murine models of viral or chemically induced skin carcinogenesis have identified a pivotal role for B cells in promoting inflammation and carcinogenesis. While many human solid tumors demonstrate significant B cell infiltration and/or tertiary lymphoid structure formation, the functional properties of tumor-infiltrating B cells and their effects on immunity are poorly understood. Recent successes in early Phase I/II trials using anti-checkpoint inhibitor antibodies such as nivolumab or pidilizumab directed against PD-1 in the setting of Hodgkin's and non-Hodgkin's lymphomas validate the therapeutic utility of reversing B cell-mediated immune suppression. Further studies to define Breg subsets, and mechanisms of suppression, may provide new avenues for modulation of the immune response and meaningful therapeutic intervention in both lymphoid and solid tumors.
Collapse
Affiliation(s)
- Yu Zhang
- Division of Hematology/Oncology, Department of Medicine, University of Miami Miller School of Medicine and Sylvester Comprehensive Cancer Center, Miami, FL 33136, USA
| | - Nicolas Gallastegui
- Division of Hematology/Oncology, Department of Medicine, University of Miami Miller School of Medicine and Sylvester Comprehensive Cancer Center, Miami, FL 33136, USA
| | - Joseph D Rosenblatt
- Division of Hematology/Oncology, Department of Medicine, University of Miami Miller School of Medicine and Sylvester Comprehensive Cancer Center, Miami, FL 33136, USA
| |
Collapse
|