1
|
Sprooten J, Laureano RS, Vanmeerbeek I, Govaerts J, Naulaerts S, Borras DM, Kinget L, Fucíková J, Špíšek R, Jelínková LP, Kepp O, Kroemer G, Krysko DV, Coosemans A, Vaes RD, De Ruysscher D, De Vleeschouwer S, Wauters E, Smits E, Tejpar S, Beuselinck B, Hatse S, Wildiers H, Clement PM, Vandenabeele P, Zitvogel L, Garg AD. Trial watch: chemotherapy-induced immunogenic cell death in oncology. Oncoimmunology 2023; 12:2219591. [PMID: 37284695 PMCID: PMC10240992 DOI: 10.1080/2162402x.2023.2219591] [Citation(s) in RCA: 21] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 05/25/2023] [Accepted: 05/25/2023] [Indexed: 06/08/2023] Open
Abstract
Immunogenic cell death (ICD) refers to an immunologically distinct process of regulated cell death that activates, rather than suppresses, innate and adaptive immune responses. Such responses culminate into T cell-driven immunity against antigens derived from dying cancer cells. The potency of ICD is dependent on the immunogenicity of dying cells as defined by the antigenicity of these cells and their ability to expose immunostimulatory molecules like damage-associated molecular patterns (DAMPs) and cytokines like type I interferons (IFNs). Moreover, it is crucial that the host's immune system can adequately detect the antigenicity and adjuvanticity of these dying cells. Over the years, several well-known chemotherapies have been validated as potent ICD inducers, including (but not limited to) anthracyclines, paclitaxels, and oxaliplatin. Such ICD-inducing chemotherapeutic drugs can serve as important combinatorial partners for anti-cancer immunotherapies against highly immuno-resistant tumors. In this Trial Watch, we describe current trends in the preclinical and clinical integration of ICD-inducing chemotherapy in the existing immuno-oncological paradigms.
Collapse
Affiliation(s)
- Jenny Sprooten
- Cell Stress & Immunity (CSI) Lab, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Raquel S. Laureano
- Cell Stress & Immunity (CSI) Lab, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Isaure Vanmeerbeek
- Cell Stress & Immunity (CSI) Lab, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Jannes Govaerts
- Cell Stress & Immunity (CSI) Lab, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Stefan Naulaerts
- Cell Stress & Immunity (CSI) Lab, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Daniel M. Borras
- Cell Stress & Immunity (CSI) Lab, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Lisa Kinget
- Laboratory of Experimental Oncology, Department of Oncology, Leuven Cancer Institute, KU Leuven, Leuven, Belgium
| | - Jitka Fucíková
- Department of Immunology, Charles University, 2Faculty of Medicine and University Hospital Motol, Prague, Czech Republic
- Sotio Biotech, Prague, Czech Republic
| | - Radek Špíšek
- Department of Immunology, Charles University, 2Faculty of Medicine and University Hospital Motol, Prague, Czech Republic
- Sotio Biotech, Prague, Czech Republic
| | - Lenka Palová Jelínková
- Department of Immunology, Charles University, 2Faculty of Medicine and University Hospital Motol, Prague, Czech Republic
- Sotio Biotech, Prague, Czech Republic
| | - Oliver Kepp
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy Cancer Center, Université Paris Saclay, Villejuif, France
- Centre de Recherche des Cordeliers, Equipe Labellisée Par la Liguecontre le Cancer, Université de Paris, sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France
| | - Guido Kroemer
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy Cancer Center, Université Paris Saclay, Villejuif, France
- Centre de Recherche des Cordeliers, Equipe Labellisée Par la Liguecontre le Cancer, Université de Paris, sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France
- Department of Biology, Hôpital Européen Georges Pompidou, AP-HP, Institut du Cancer Paris CARPEM, Paris, France
| | - Dmitri V. Krysko
- Cell Death Investigation and Therapy (CDIT) Laboratory, Department of Human Structure and Repair, Ghent University, Ghent, Belgium
- Cancer Research Insitute Ghent, Ghent University, Ghent, Belgium
| | - An Coosemans
- Laboratory of Tumor Immunology and Immunotherapy, Department of Oncology, Leuven Cancer Institute, KU Leuven, Leuven, Belgium
| | - Rianne D.W. Vaes
- Department of Radiation Oncology (MAASTRO), GROW School for Oncology and Reproduction, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Dirk De Ruysscher
- Department of Radiation Oncology (MAASTRO), GROW School for Oncology and Reproduction, Maastricht University Medical Center, Maastricht, The Netherlands
- Department of Radiotherapy, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Steven De Vleeschouwer
- Department Neurosurgery, University Hospitals Leuven, Leuven, Belgium
- Department Neuroscience, Laboratory for Experimental Neurosurgery and Neuroanatomy, KU Leuven, Leuven, Belgium
- Leuven Brain Institute (LBI), KU Leuven, Leuven, Belgium
| | - Els Wauters
- Laboratory of Respiratory Diseases and Thoracic Surgery (Breathe), Department of Chronic Diseases and Metabolism, KU Leuven, Leuven, Belgium
| | - Evelien Smits
- Center for Oncological Research (CORE), Integrated Personalized and Precision Oncology Network (IPPON), University of Antwerp, Antwerp, Belgium
- Center for Cell Therapy and Regenerative Medicine, Antwerp University Hospital, Antwerp, Belgium
| | - Sabine Tejpar
- Molecular Digestive Oncology, Department of Oncology, Katholiek Universiteit Leuven, Leuven, Belgium
- Cell Death and Inflammation Unit, VIB-Ugent Center for Inflammation Research (IRC), Ghent, Belgium
| | - Benoit Beuselinck
- Laboratory of Experimental Oncology, Department of Oncology, Leuven Cancer Institute, KU Leuven, Leuven, Belgium
| | - Sigrid Hatse
- Laboratory of Experimental Oncology, Department of Oncology, Leuven Cancer Institute, KU Leuven, Leuven, Belgium
| | - Hans Wildiers
- Laboratory of Experimental Oncology, Department of Oncology, Leuven Cancer Institute, KU Leuven, Leuven, Belgium
| | - Paul M. Clement
- Laboratory of Experimental Oncology, Department of Oncology, Leuven Cancer Institute, KU Leuven, Leuven, Belgium
| | - Peter Vandenabeele
- Cell Death and Inflammation Unit, VIB-Ugent Center for Inflammation Research (IRC), Ghent, Belgium
- Molecular Signaling and Cell Death Unit, Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Laurence Zitvogel
- Tumour Immunology and Immunotherapy of Cancer, European Academy of Tumor Immunology, Gustave Roussy Cancer Center, Inserm, Villejuif, France
| | - Abhishek D. Garg
- Cell Stress & Immunity (CSI) Lab, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| |
Collapse
|
2
|
He Y, Wang C, Liang Q, Guo R, Jiang J, Shen W, Hu K. PKHB1 peptide induces antiviral effects through induction of immunogenic cell death in herpes simplex keratitis. Front Pharmacol 2022; 13:1048978. [PMID: 36532743 PMCID: PMC9751201 DOI: 10.3389/fphar.2022.1048978] [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: 09/20/2022] [Accepted: 11/09/2022] [Indexed: 12/26/2023] Open
Abstract
Herpes simplex keratitis (HSK) is a severe, infectious corneal disease caused by herpes simplex virus type 1 (HSV-1) infection. The increasing prevalence of acyclovir resistance, the side effects of hormonal drugs, and the ease of recurrence after surgery have made it crucial to develop new methods of treating HSK. HSV-1 evades the host immune response through various mechanisms. Therefore, we explored the role of the immunogenic cell death inducer PKHB1 peptide in HSK. After subconjunctival injection of PKHB1 peptide, we observed the ocular surface lesions and survival of HSK mice and detected the virus levels in tear fluid, corneas, and trigeminal ganglions. We found that PKHB1 peptide reduced HSV-1 levels in the eye and alleviated the severity of HSK. Moreover, it increased the number of corneal infiltrating antigen-presenting cells (APCs), such as macrophages and dendritic cells, and CD8+ T cells in ocular draining lymph nodes. We further observed that PKHB1 peptide promoted the exposure of calreticulin, as well as the release of ATP and high-mobility group box 1 in HSV-1-infected cells in vitro. Our findings suggested that PKHB1 peptide promoted the recruitment and maturation of APCs by inducing the release of large amounts of damage-associated molecular patterns from infected cells. APCs then phagocytized antigenic materials and translocated to the lymph nodes, triggering a cytotoxic T lymphocyte-dependent immune response that ultimately alleviated HSK.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Kai Hu
- Department of Ophthalmology, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
| |
Collapse
|
3
|
Jalil AR, Andrechak JC, Hayes BH, Chenoweth DM, Discher DE. Human CD47-Derived Cyclic Peptides Enhance Engulfment of mAb-Targeted Melanoma by Primary Macrophages. Bioconjug Chem 2022; 33:1973-1982. [PMID: 35285229 PMCID: PMC10805119 DOI: 10.1021/acs.bioconjchem.2c00020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
CD47 on healthy cells, cancer cells, and even engineered particles can inhibit phagocytic clearance by binding SIRPα on macrophages. To mimic and modulate this interaction with peptides that could be used as soluble antagonists or potentially as bioconjugates to surfaces, we made cyclic "nano-Self" peptides based on the key interaction loop of human CD47. Melanoma cells were studied as a standard preclinical cancer model and were antibody-opsonized to adhere to and activate engulfment by primary mouse macrophages. Phagocytosis in the presence of soluble peptides showed cyclic > wildtype > scrambled activity, with the same trend observed with human cells. Opsonized cells that were not engulfed adhered tightly to macrophages, with opposite trends to phagocytosis. Peptide activity is nonetheless higher in human versus mouse assays, consistent with species differences in CD47-SIRPα. Small peptides thus function as soluble antagonists of a major macrophage checkpoint.
Collapse
|
4
|
Activation of Cellular Players in Adaptive Immunity via Exogenous Delivery of Tumor Cell Lysates. Pharmaceutics 2022; 14:pharmaceutics14071358. [PMID: 35890254 PMCID: PMC9316852 DOI: 10.3390/pharmaceutics14071358] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 06/19/2022] [Accepted: 06/22/2022] [Indexed: 12/04/2022] Open
Abstract
Tumor cell lysates (TCLs) are a good immunogenic source of tumor-associated antigens. Since whole necrotic TCLs can enhance the maturation and antigen-presenting ability of dendritic cells (DCs), multiple strategies for the exogenous delivery of TCLs have been investigated as novel cancer immunotherapeutic solutions. The TCL-mediated induction of DC maturation and the subsequent immunological response could be improved by utilizing various material-based carriers. Enhanced antitumor immunity and cancer vaccination efficacy could be eventually achieved through the in vivo administration of TCLs. Therefore, (1) important engineering methodologies to prepare antigen-containing TCLs, (2) current therapeutic approaches using TCL-mediated DC activation, and (3) the significant sequential mechanism of DC-based signaling and stimulation in adaptive immunity are summarized in this review. More importantly, the recently reported developments in biomaterial-based exogenous TCL delivery platforms and co-delivery strategies with adjuvants for effective cancer vaccination and antitumor effects are emphasized.
Collapse
|
5
|
Calvillo-Rodríguez KM, Mendoza-Reveles R, Gómez-Morales L, Uscanga-Palomeque AC, Karoyan P, Martínez-Torres AC, Rodríguez-Padilla C. PKHB1, a thrombospondin-1 peptide mimic, induces anti-tumor effect through immunogenic cell death induction in breast cancer cells. Oncoimmunology 2022; 11:2054305. [PMID: 35402082 PMCID: PMC8986196 DOI: 10.1080/2162402x.2022.2054305] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Breast cancer is the most commonly diagnosed cancer and the leading cause of cancer death in women worldwide. Recent advances in the field of immuno-oncology demonstrate the beneficial immunostimulatory effects of the induction of immunogenic cell death (ICD). ICD increases tumor infiltration by T cells and is associated with improved prognosis in patients affected by triple negative breast cancer (TNBC) with residual disease. The aim of this study was to evaluate the antitumoral effect of PKHB1, a thrombospondin-1 peptide mimic, against breast cancer cells, and the immunogenicity of the cell death induced by PKHB1 in vitro, ex vivo, and in vivo. Our results showed that PKHB1 induces mitochondrial alterations, ROS production, intracellular Ca2+ accumulation, as well calcium-dependent cell death in breast cancer cells, including triple negative subtypes. PKHB1 has antitumor effect in vivo leading to a reduction of tumor volume and weight and promotes intratumoral CD8 + T cell infiltration. Furthermore, in vitro, PKHB1 induces calreticulin (CALR), HSP70, and HSP90 exposure and release of ATP and HMGB1. Additionally, the killed cells obtained after treatment with PKHB1 (PKHB1-KC) induced dendritic cell maturation, and T cell antitumor responses, ex vivo. Moreover, PKHB1-KC in vivo were able to induce an antitumor response against breast cancer cells in a prophylactic application, whereas in a therapeutic setting, PKHB1-KC induced tumor regression; both applications induced a long-term antitumor response. Altogether our data shows that PKHB1, a thrombospondin-1 peptide mimic, has in vivo antitumor effect and induce immune system activation through immunogenic cell death induction in breast cancer cells.
Collapse
Affiliation(s)
- Kenny Misael Calvillo-Rodríguez
- Facultad de Ciencias Biologicas, Laboratorio de Inmunología y Virología, Universidad Autónoma de Nuevo León, San Nicolás de los Garza, Mexico
- Sorbonne Université, Ecole Normale Supérieure, PSL University, CNRS, Laboratoire des Biomolécules, LBM, 75005 Paris, France
- Sorbonne Université, Ecole Normale Supérieure, PSL University, CNRS, Laboratoire des Biomolécules, DRUG Lab, Site OncoDesign, 25-27 Avenue du Québec, 91140 Les Ulis, France
| | - Rodolfo Mendoza-Reveles
- Facultad de Ciencias Biologicas, Laboratorio de Inmunología y Virología, Universidad Autónoma de Nuevo León, San Nicolás de los Garza, Mexico
| | - Luis Gómez-Morales
- Facultad de Ciencias Biologicas, Laboratorio de Inmunología y Virología, Universidad Autónoma de Nuevo León, San Nicolás de los Garza, Mexico
- Sorbonne Université, Ecole Normale Supérieure, PSL University, CNRS, Laboratoire des Biomolécules, LBM, 75005 Paris, France
- Sorbonne Université, Ecole Normale Supérieure, PSL University, CNRS, Laboratoire des Biomolécules, DRUG Lab, Site OncoDesign, 25-27 Avenue du Québec, 91140 Les Ulis, France
- Kaybiotix, GmbH, Zugerstrasse 32, 6340 Baar, Switzerland
| | | | - Philippe Karoyan
- Sorbonne Université, Ecole Normale Supérieure, PSL University, CNRS, Laboratoire des Biomolécules, LBM, 75005 Paris, France
- Sorbonne Université, Ecole Normale Supérieure, PSL University, CNRS, Laboratoire des Biomolécules, DRUG Lab, Site OncoDesign, 25-27 Avenue du Québec, 91140 Les Ulis, France
- Kaybiotix, GmbH, Zugerstrasse 32, 6340 Baar, Switzerland
- Kayvisa, AG, Industriestrasse, 44, 6300 Zug, Switzerland
- χ-Pharma, 25 Avenue du Québec, 91140 Les Ulis, France
| | - Ana Carolina Martínez-Torres
- Facultad de Ciencias Biologicas, Laboratorio de Inmunología y Virología, Universidad Autónoma de Nuevo León, San Nicolás de los Garza, Mexico
| | - Cristina Rodríguez-Padilla
- Facultad de Ciencias Biologicas, Laboratorio de Inmunología y Virología, Universidad Autónoma de Nuevo León, San Nicolás de los Garza, Mexico
- LONGEVEDEN SA de CV, Monterrey, Mexico
| |
Collapse
|
6
|
The bovine dialysable leukocyte extract IMMUNEPOTENT CRP induces immunogenic cell death in breast cancer cells leading to long-term antitumour memory. Br J Cancer 2021; 124:1398-1410. [PMID: 33531687 PMCID: PMC8039030 DOI: 10.1038/s41416-020-01256-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 12/01/2020] [Accepted: 12/16/2020] [Indexed: 12/22/2022] Open
Abstract
Background Cancer recurrence is a serious problem in breast cancer (BC) patients, and immunogenic cell death (ICD) has been proposed as a strategy to overcome this recurrence. IMMUNEPOTENT CRP (ICRP) acts as an immunomodulator and can be cytotoxic to cancer cells. Thus, we evaluated if ICRP induces ICD in BC cells. Methods Immunogenicity of ICRP-induced cell death was evaluated in vitro, analysing the principal biochemical characteristics of ICD in MCF-7, MDA-MB-231 and 4T1 cells. Ex vivo, we assessed the ability of killed cancer cells (KCC) obtained from ICRP-treated 4T1 cells (ICRP-KCC) to induce DC maturation, T-cell priming and T-cell-mediated cancer cytotoxicity. In vivo, we evaluated tumour establishment and antitumour immune memory after prophylactic ICRP-KCC vaccination in BALB/c mice. Results ICRP induced caspase-independent, ROS-dependent cell death, autophagosome formation, P-eIF2α, chaperone protein exposure, CD47 loss, ATP and HMBG1 release in BC cells. Additionally, ICRP-KCC promoted DC maturation, which triggered T-cell priming and cancer cytotoxicity. Prophylactic vaccination with ICRP-KCC prevented tumour establishment and induced long-term antitumour memory in BALB/c mice, involving DC maturation in lymph nodes, CD8+ T-cell augmentation in lymph nodes, peripheral blood and tumour site and ex vivo tumour-specific cytotoxicity by splenocytes. Conclusions ICRP induces ICD in BC cells, leading to long-term antitumour memory.
Collapse
|
7
|
Leclair P, Lim CJ. CD47 (Cluster of differentiation 47): an anti-phagocytic receptor with a multitude of signaling functions. Anim Cells Syst (Seoul) 2020; 24:243-252. [PMID: 33224442 PMCID: PMC7654641 DOI: 10.1080/19768354.2020.1818618] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
CD47 is a tumor-associated antigen best known for its ability to bind counter-receptors on the surface of professional phagocytes as an immune-evasion strategy. Recently, CD47 has been shown to play a role as a signaling receptor, involving a number of cell physiological processes. This review provides a comprehensive survey of the signaling pathways triggered by CD47 ligand-mediated cell death in tumor cells. Such an understanding should lead to improvement of CD47-targeted anti-tumor therapeutics able to both neutralize the anti-phagocytic role and trigger autonomous tumor cell death.
Collapse
Affiliation(s)
- Pascal Leclair
- Department of Pediatrics, University of British Columbia, and, Michael Cuccione Childhood Cancer Research Program, B.C. Children's Hospital Research Institute, Vancouver, Canada
| | - Chinten James Lim
- Department of Pediatrics, University of British Columbia, and, Michael Cuccione Childhood Cancer Research Program, B.C. Children's Hospital Research Institute, Vancouver, Canada
| |
Collapse
|
8
|
Eladl E, Tremblay-LeMay R, Rastgoo N, Musani R, Chen W, Liu A, Chang H. Role of CD47 in Hematological Malignancies. J Hematol Oncol 2020; 13:96. [PMID: 32677994 PMCID: PMC7364564 DOI: 10.1186/s13045-020-00930-1] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Accepted: 07/02/2020] [Indexed: 12/12/2022] Open
Abstract
CD47, or integrin-associated protein, is a cell surface ligand expressed in low levels by nearly all cells of the body. It plays an integral role in various immune responses as well as autoimmunity, by sending a potent "don't eat me" signal to prevent phagocytosis. A growing body of evidence demonstrates that CD47 is overexpressed in various hematological malignancies and its interaction with SIRPα on the phagocytic cells prevents phagocytosis of cancer cells. Additionally, it is expressed by different cell types in the tumor microenvironment and is required for establishing tumor metastasis. Overexpression of CD47 is thus often associated with poor clinical outcomes. CD47 has emerged as a potential therapeutic target and is being investigated in various preclinical studies as well as clinical trials to prove its safety and efficacy in treating hematological neoplasms. This review focuses on different therapeutic mechanisms to target CD47, either alone or in combination with other cell surface markers, and its pivotal role in impairing tumor growth and metastatic spread of various types of hematological malignancies.
Collapse
Affiliation(s)
- Entsar Eladl
- Laboratory Medicine Program, Toronto General Hospital, University Health Network, University of Toronto, 11th floor, 200 Elizabeth Street, Toronto, ON, M5G 2C4, Canada
| | - Rosemarie Tremblay-LeMay
- Laboratory Medicine Program, Toronto General Hospital, University Health Network, University of Toronto, 11th floor, 200 Elizabeth Street, Toronto, ON, M5G 2C4, Canada
| | - Nasrin Rastgoo
- Laboratory Medicine Program, Toronto General Hospital, University Health Network, University of Toronto, 11th floor, 200 Elizabeth Street, Toronto, ON, M5G 2C4, Canada
| | - Rumina Musani
- Laboratory Medicine Program, Toronto General Hospital, University Health Network, University of Toronto, 11th floor, 200 Elizabeth Street, Toronto, ON, M5G 2C4, Canada
| | - Wenming Chen
- Department of Hematology, Beijing Chaoyang Hospital, Capital University, Beijing, China
| | - Aijun Liu
- Department of Hematology, Beijing Chaoyang Hospital, Capital University, Beijing, China.
| | - Hong Chang
- Laboratory Medicine Program, Toronto General Hospital, University Health Network, University of Toronto, 11th floor, 200 Elizabeth Street, Toronto, ON, M5G 2C4, Canada.
| |
Collapse
|
9
|
Martínez-Torres AC, Reyes-Ruiz A, Calvillo-Rodriguez KM, Alvarez-Valadez KM, Uscanga-Palomeque AC, Tamez-Guerra RS, Rodríguez-Padilla C. IMMUNEPOTENT CRP induces DAMPS release and ROS-dependent autophagosome formation in HeLa and MCF-7 cells. BMC Cancer 2020; 20:647. [PMID: 32660440 PMCID: PMC7359018 DOI: 10.1186/s12885-020-07124-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Accepted: 07/01/2020] [Indexed: 12/23/2022] Open
Abstract
Background IMMUNEPOTENT CRP (ICRP) can be cytotoxic to cancer cell lines. However, its widespread use in cancer patients has been limited by the absence of conclusive data on the molecular mechanism of its action. Here, we evaluated the mechanism of cell death induced by ICRP in HeLa and MCF-7 cells. Methods Cell death, cell cycle, mitochondrial membrane potential and ROS production were evaluated in HeLa and MCF-7 cell lines after ICRP treatment. Caspase-dependence and ROS-dependence were evaluated using QVD.oph and NAC pre-treatment in cell death analysis. DAMPs release, ER stress (eIF2-α phosphorylation) and autophagosome formation were analyzed as well. Additionally, the role of autophagosomes in cell death induced by ICRP was evaluated using SP-1 pre-treatment in cell death in HeLa and MCF-7 cells. Results ICRP induces cell death, reaching CC50 at 1.25 U/mL and 1.5 U/mL in HeLa and MCF-7 cells, respectively. Loss of mitochondrial membrane potential, ROS production and cell cycle arrest were observed after ICRP CC50 treatment in both cell lines, inducing the same mechanism, a type of cell death independent of caspases, relying on ROS production. Additionally, ICRP-induced cell death involves features of immunogenic cell death such as P-eIF2α and CRT exposure, as well as, ATP and HMGB1 release. Furthermore, ICRP induces ROS-dependent autophagosome formation that acts as a pro-survival mechanism. Conclusions ICRP induces a non-apoptotic cell death that requires an oxidative stress to take place, involving mitochondrial damage, ROS-dependent autophagosome formation, ER stress and DAMPs’ release. These data indicate that ICRP could work together with classic apoptotic inductors to attack cancer cells from different mechanisms, and that ICRP-induced cell death might activate an immune response against cancer cells.
Collapse
Affiliation(s)
- Ana Carolina Martínez-Torres
- Universidad Autonoma de Nuevo Leon, Facultad de Ciencias Biologicas, Laboratorio de Inmunologia y Virologia, San Nicolás de los Garza, Mexico.
| | - Alejandra Reyes-Ruiz
- Universidad Autonoma de Nuevo Leon, Facultad de Ciencias Biologicas, Laboratorio de Inmunologia y Virologia, San Nicolás de los Garza, Mexico
| | - Kenny Misael Calvillo-Rodriguez
- Universidad Autonoma de Nuevo Leon, Facultad de Ciencias Biologicas, Laboratorio de Inmunologia y Virologia, San Nicolás de los Garza, Mexico
| | - Karla Maria Alvarez-Valadez
- Universidad Autonoma de Nuevo Leon, Facultad de Ciencias Biologicas, Laboratorio de Inmunologia y Virologia, San Nicolás de los Garza, Mexico
| | - Ashanti C Uscanga-Palomeque
- Universidad Autonoma de Nuevo Leon, Facultad de Ciencias Biologicas, Laboratorio de Inmunologia y Virologia, San Nicolás de los Garza, Mexico
| | - Reyes S Tamez-Guerra
- Universidad Autonoma de Nuevo Leon, Facultad de Ciencias Biologicas, Laboratorio de Inmunologia y Virologia, San Nicolás de los Garza, Mexico
| | - Cristina Rodríguez-Padilla
- Universidad Autonoma de Nuevo Leon, Facultad de Ciencias Biologicas, Laboratorio de Inmunologia y Virologia, San Nicolás de los Garza, Mexico.,Longeveden, SA de CV, Monterrey, Mexico
| |
Collapse
|
10
|
Martínez-Torres AC, Lorenzo-Anota HY, García-Juárez MG, Zarate-Triviño DG, Rodríguez-Padilla C. Chitosan gold nanoparticles induce different ROS-dependent cell death modalities in leukemic cells. Int J Nanomedicine 2019; 14:7173-7190. [PMID: 31564872 PMCID: PMC6734554 DOI: 10.2147/ijn.s221021] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2019] [Accepted: 08/03/2019] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Nanotechnology proposes the use of gold nanoparticles (AuNPs) for drug delivery, diagnosis, and treatment of cancer. Leukemia is a type of hematopoietic cancer that results from the malignant transformation of white blood cells. Chitosan-coated AuNPs (CH-AuNPs) are cell death inductors in HeLa and MCF-7 cancer cells without affecting peripheral blood mononuclear cells (PBMC). Considering the selectivity and versatile cytotoxicity of CH-AuNPs, we evaluated whether their selectivity is due to the cell lineage or the characteristics of the cancer cells, by assessing its cytotoxicity in leukemic cells. Moreover, we further examined the cell death mechanism and assessed the implication of nuclear damage, autophagosome formation, and the cell death mechanism induced in leukemic cells. MATERIALS AND METHODS We synthesized CH-AuNPs by chemical methods and analyzed their cell death capacity in a T-acute lymphocytic leukemia cell line (CEM), in a chronic myeloid leukemia cell line (K562), and in healthy cells from the same lineage (PBMC and bone marrow, BM, cells). Then, we assessed ROS generation and mitochondrial and nuclear damage. Finally, we evaluated whether cell death occurred by autophagy, apoptosis, or necroptosis, and the role of ROS in this mechanism. RESULTS We found that CH-AuNPs did not affect PBMC and BM cells, whereas they are cytotoxic in a dose-dependent manner in leukemic cells. ROS production leads to mitochondrial and nuclear damage, and cell death. We found that CH-AuNPs induce apoptosis in CEM and necroptosis in K562, both undergoing autophagy as a pro-survival mechanism. CONCLUSION CH-AuNPs are selective cell death inductors in hematologic cancer cells, without affecting their healthy counterparts. Cell death induced by CH-AuNPs is independent of the cancer cell type; however, its mechanism is different depending on the type of leukemic cells.
Collapse
Affiliation(s)
- Ana Carolina Martínez-Torres
- Universidad Autónoma De Nuevo León, Facultad De Ciencias Biológicas, Laboratorio De Inmunología Y Virología, Monterrey, Nuevo Leon, Mexico
| | - Helen Yarimet Lorenzo-Anota
- Universidad Autónoma De Nuevo León, Facultad De Ciencias Biológicas, Laboratorio De Inmunología Y Virología, Monterrey, Nuevo Leon, Mexico
| | - Martín Gerardo García-Juárez
- Universidad Autónoma De Nuevo León, Facultad De Ciencias Biológicas, Laboratorio De Inmunología Y Virología, Monterrey, Nuevo Leon, Mexico
| | - Diana G Zarate-Triviño
- Universidad Autónoma De Nuevo León, Facultad De Ciencias Biológicas, Laboratorio De Inmunología Y Virología, Monterrey, Nuevo Leon, Mexico
| | - Cristina Rodríguez-Padilla
- Universidad Autónoma De Nuevo León, Facultad De Ciencias Biológicas, Laboratorio De Inmunología Y Virología, Monterrey, Nuevo Leon, Mexico
| |
Collapse
|
11
|
Denèfle T, Pramil E, Gómez-Morales L, Levasseur MD, Lardé E, Newton C, Herry K, Herbi L, Lamotte Y, Odile E, Ancellin N, Grondin P, Martinez-Torres AC, Viviani F, Merle-Beral H, Lequin O, Susin SA, Karoyan P. Homotrimerization Approach in the Design of Thrombospondin-1 Mimetic Peptides with Improved Potency in Triggering Regulated Cell Death of Cancer Cells. J Med Chem 2019; 62:7656-7668. [PMID: 31403795 DOI: 10.1021/acs.jmedchem.9b00024] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
In order to optimize the potency of the first serum-stable peptide agonist of CD47 (PKHB1) in triggering regulated cell death of cancer cells, we designed a maturation process aimed to mimic the trimeric structure of the thrombospondin-1/CD47 binding epitope. For that purpose, an N-methylation scan of the PKHB1 sequence was realized to prevent peptide aggregation. Structural and pharmacological analyses were conducted in order to assess the conformational impact of these chemical modifications on the backbone structure and the biological activity. This structure-activity relationship study led to the discovery of a highly soluble N-methylated peptide that we termed PKT16. Afterward, this monomer was used for the design of a homotrimeric peptide mimic that we termed [PKT16]3, which proved to be 10-fold more potent than its monomeric counterpart. A pharmacological evaluation of [PKT16]3 in inducing cell death of adherent (A549) and nonadherent (MEC-1) cancer cell lines was also performed.
Collapse
Affiliation(s)
- Thomas Denèfle
- Sorbonne Université , Ecole Normale Supérieure, PSL University, CNRS, Laboratoire des Biomolécules, LBM , 75005 Paris , France.,Sorbonne Université , Ecole Normale Supérieure, PSL University, CNRS, Laboratoire des Biomolécules, LBM, Site OncoDesign , 25-27 Avenue du Québec , 91140 Les Ulis , France
| | - Elodie Pramil
- Sorbonne Université , Ecole Normale Supérieure, PSL University, CNRS, Laboratoire des Biomolécules, LBM , 75005 Paris , France.,Sorbonne Université , Ecole Normale Supérieure, PSL University, CNRS, Laboratoire des Biomolécules, LBM, Site OncoDesign , 25-27 Avenue du Québec , 91140 Les Ulis , France.,Cell Death and Drug Resistance in Lymphoproliferative Disorders Team , Centre de Recherche des Cordeliers, INSERM UMRS 1138 , 75006 Paris , France
| | - Luis Gómez-Morales
- Sorbonne Université , Ecole Normale Supérieure, PSL University, CNRS, Laboratoire des Biomolécules, LBM , 75005 Paris , France.,Sorbonne Université , Ecole Normale Supérieure, PSL University, CNRS, Laboratoire des Biomolécules, LBM, Site OncoDesign , 25-27 Avenue du Québec , 91140 Les Ulis , France.,Laboratory of Immunology and Virology , Autonomous University of Nuevo Leon , 66451 San Nicolas de los Garza , NL , Mexico
| | - Mikail D Levasseur
- Sorbonne Université , Ecole Normale Supérieure, PSL University, CNRS, Laboratoire des Biomolécules, LBM , 75005 Paris , France.,Sorbonne Université , Ecole Normale Supérieure, PSL University, CNRS, Laboratoire des Biomolécules, LBM, Site OncoDesign , 25-27 Avenue du Québec , 91140 Les Ulis , France
| | - Eva Lardé
- Sorbonne Université , Ecole Normale Supérieure, PSL University, CNRS, Laboratoire des Biomolécules, LBM , 75005 Paris , France.,Sorbonne Université , Ecole Normale Supérieure, PSL University, CNRS, Laboratoire des Biomolécules, LBM, Site OncoDesign , 25-27 Avenue du Québec , 91140 Les Ulis , France
| | - Clara Newton
- Sorbonne Université , Ecole Normale Supérieure, PSL University, CNRS, Laboratoire des Biomolécules, LBM , 75005 Paris , France.,Sorbonne Université , Ecole Normale Supérieure, PSL University, CNRS, Laboratoire des Biomolécules, LBM, Site OncoDesign , 25-27 Avenue du Québec , 91140 Les Ulis , France
| | - Kenny Herry
- OncoDesign , 25 Avenue du Québec , 91140 Les Ulis , France
| | - Linda Herbi
- Cell Death and Drug Resistance in Lymphoproliferative Disorders Team , Centre de Recherche des Cordeliers, INSERM UMRS 1138 , 75006 Paris , France
| | - Yann Lamotte
- OncoDesign , 25 Avenue du Québec , 91140 Les Ulis , France
| | - Estelle Odile
- Sorbonne Université , Ecole Normale Supérieure, PSL University, CNRS, Laboratoire des Biomolécules, LBM , 75005 Paris , France.,Sorbonne Université , Ecole Normale Supérieure, PSL University, CNRS, Laboratoire des Biomolécules, LBM, Site OncoDesign , 25-27 Avenue du Québec , 91140 Les Ulis , France
| | | | - Pascal Grondin
- OncoDesign , 25 Avenue du Québec , 91140 Les Ulis , France
| | - Ana-Carolina Martinez-Torres
- Laboratory of Immunology and Virology , Autonomous University of Nuevo Leon , 66451 San Nicolas de los Garza , NL , Mexico
| | | | - Hélène Merle-Beral
- Cell Death and Drug Resistance in Lymphoproliferative Disorders Team , Centre de Recherche des Cordeliers, INSERM UMRS 1138 , 75006 Paris , France
| | - Olivier Lequin
- Sorbonne Université , Ecole Normale Supérieure, PSL University, CNRS, Laboratoire des Biomolécules, LBM , 75005 Paris , France
| | - Santos A Susin
- Cell Death and Drug Resistance in Lymphoproliferative Disorders Team , Centre de Recherche des Cordeliers, INSERM UMRS 1138 , 75006 Paris , France
| | - Philippe Karoyan
- Sorbonne Université , Ecole Normale Supérieure, PSL University, CNRS, Laboratoire des Biomolécules, LBM , 75005 Paris , France.,Sorbonne Université , Ecole Normale Supérieure, PSL University, CNRS, Laboratoire des Biomolécules, LBM, Site OncoDesign , 25-27 Avenue du Québec , 91140 Les Ulis , France.,SiRIC CURAMUS (CANCER UNITED RESEARCH ASSOCIATING MEDICINE, UNIVERSITY & SOCIETY, Site de Recherche Intégrée sur le Cancer) IUC, AP-HP.6 , Sorbonne Université 75005 Paris , France.,Kayvisa AG , Industriestrasse, 44 , 6300 Zug , Switzerland.,Kaybiotix GmbH , Zugerstrasse 32 , 6340 Baar , Switzerland
| |
Collapse
|