101
|
Hsu MA, Okamura SM, De Magalhaes Filho CD, Bergeron DM, Rodriguez A, West M, Yadav D, Heim R, Fong JJ, Garcia-Guzman M. Cancer-targeted photoimmunotherapy induces antitumor immunity and can be augmented by anti-PD-1 therapy for durable anticancer responses in an immunologically active murine tumor model. Cancer Immunol Immunother 2023; 72:151-168. [PMID: 35776159 DOI: 10.1007/s00262-022-03239-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 06/06/2022] [Indexed: 01/07/2023]
Abstract
The complex immunosuppressive nature of solid tumor microenvironments poses a significant challenge to generating efficacious and durable anticancer responses. Photoimmunotherapy is a cancer treatment strategy by which an antibody is conjugated with a non-toxic light-activatable dye. Following administration of the conjugate and binding to the target tumor, subsequent local laser illumination activates the dye, resulting in highly specific target cell membrane disruption. Here we demonstrate that photoimmunotherapy treatment elicited tumor necrosis, thus inducing immunogenic cell death characterized by the release of damage-associated molecular patterns (DAMPs). Photoimmunotherapy-killed tumor cells activated dendritic cells (DC), leading to the production of proinflammatory cytokines, T cell stimulation, priming antigen-specific T cells, and durable memory T cell responses, which led complete responder mice to effectively reject new tumors upon rechallenge. PD-1 blockade in combination with photoimmunotherapy enhanced overall anticancer efficacy, including against anti-PD-1-resistant tumors. The combination treatment also elicited abscopal anticancer activity, as observed by reduction of distal, non-illuminated tumors, further demonstrating the ability of photoimmunotherapy to harness local and peripheral T cell responses. With this work we therefore delineate the immune mechanisms of action for photoimmunotherapy and demonstrate the potential for cancer-targeted photoimmunotherapy to be combined with other immunotherapy approaches for augmented, durable anticancer efficacy. Moreover, we demonstrate responses utilizing various immunocompetent mouse models, as well as in vitro data from human cells, suggesting broad translational potential.
Collapse
Affiliation(s)
- Michelle A Hsu
- Rakuten Medical, Inc., Translational Sciences, 11080 Roselle Street, San Diego, CA, 92121, USA
| | - Stephanie M Okamura
- Rakuten Medical, Inc., Translational Sciences, 11080 Roselle Street, San Diego, CA, 92121, USA
| | | | - Daniele M Bergeron
- Rakuten Medical, Inc., Translational Sciences, 11080 Roselle Street, San Diego, CA, 92121, USA
| | - Ahiram Rodriguez
- Rakuten Medical, Inc., Translational Sciences, 11080 Roselle Street, San Diego, CA, 92121, USA
| | - Melissa West
- Rakuten Medical, Inc., Translational Sciences, 11080 Roselle Street, San Diego, CA, 92121, USA
| | - Deepak Yadav
- Rakuten Medical, Inc., Translational Sciences, 11080 Roselle Street, San Diego, CA, 92121, USA
| | - Roger Heim
- Rakuten Medical, Inc., Translational Sciences, 11080 Roselle Street, San Diego, CA, 92121, USA
| | - Jerry J Fong
- Rakuten Medical, Inc., Translational Sciences, 11080 Roselle Street, San Diego, CA, 92121, USA.
| | - Miguel Garcia-Guzman
- Rakuten Medical, Inc., Translational Sciences, 11080 Roselle Street, San Diego, CA, 92121, USA
| |
Collapse
|
102
|
Najibi AJ, Larkin K, Feng Z, Jeffreys N, Dacus MT, Rustagi Y, Hodi FS, Mooney DJ. Chemotherapy Dose Shapes the Expression of Immune-Interacting Markers on Cancer Cells. Cell Mol Bioeng 2022; 15:535-551. [PMID: 36531864 PMCID: PMC9751245 DOI: 10.1007/s12195-022-00742-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Accepted: 09/21/2022] [Indexed: 11/30/2022] Open
Abstract
Introduction Tumor and immune cells interact through a variety of cell-surface proteins that can either restrain or promote tumor progression. The impacts of cytotoxic chemotherapy dose and delivery route on this interaction profile remain incompletely understood, and could support the development of more effective combination therapies for cancer treatment. Methods and Results Here, we found that exposure to the anthracycline doxorubicin altered the expression of numerous immune-interacting markers (MHC-I, PD-L1, PD-L2, CD47, Fas, and calreticulin) on live melanoma, breast cancer, and leukemia cells in a dose-dependent manner in vitro. Notably, an intermediate dose best induced immunogenic cell death and the expression of immune-activating markers without maximizing expression of markers associated with immune suppression. Bone marrow-derived dendritic cells exposed to ovalbumin-expressing melanoma treated with intermediate doxorubicin dose became activated and best presented tumor antigen. In a murine melanoma model, both the doxorubicin dose and delivery location (systemic infusion versus local administration) affected the expression of these markers on live tumor cells. Particularly, local release of doxorubicin from a hydrogel increased calreticulin expression on tumor cells without inducing immune-suppressive markers, in a manner dependent on the loaded dose. Doxorubicin exposure also altered the expression of immune-interacting markers in patient-derived melanoma cells. Conclusions Together, these results illustrate how standard-of-care chemotherapy, when administered in various manners, can lead to distinct expression of immunogenic markers on cancer cells. These findings may inform development of chemo-immunotherapy combinations for cancer treatment. Supplementary Information The online version contains supplementary material available at 10.1007/s12195-022-00742-y.
Collapse
Affiliation(s)
- Alexander J. Najibi
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138 USA
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA 02115 USA
| | - Kerry Larkin
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138 USA
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA 02115 USA
| | - Zhaoqianqi Feng
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138 USA
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA 02115 USA
| | - Nicholas Jeffreys
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138 USA
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA 02115 USA
| | - Mason T. Dacus
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138 USA
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA 02115 USA
| | - Yashika Rustagi
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215 USA
| | - F. Stephen Hodi
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215 USA
| | - David J. Mooney
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138 USA
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA 02115 USA
| |
Collapse
|
103
|
Gayle S, Paradis T, Jones K, Vasquez J, Paralkar VM. Antigen-independent tumor targeting by CBX-12 (alphalex ™-exatecan) induces long-term antitumor immunity. Immunotherapy 2022; 14:1467-1480. [PMID: 36597724 DOI: 10.2217/imt-2022-0121] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Aims: To determine whether antigen-independent targeting of the TOP1 inhibitor exatecan to tumor with a pH-sensitive peptide (CBX-12) produces superior synergy with immunotherapy compared with unconjugated exatecan. Materials & methods: In vitro and ex vivo functional assays were performed via FACS and ELISA assays. In vivo efficacy was evaluated in the syngeneic CT26 model. Results: CBX-12 combined with anti-PD-1 or anti-CTLA4 results in delayed tumor growth and complete response, with cured animals displaying long-term antitumor immunity. CBX-12 stimulates expression of MHC 1 and PD-L1 and is an inducer of immunogenic cell death, producing long-term immune recognition of tumor cells and resultant antitumor immunity. Conclusion: The authors' data provide the rationale for exploring immunotherapy combinations with CBX-12 in clinical trials.
Collapse
Affiliation(s)
| | | | - Kelli Jones
- Cybrexa Therapeutics, New Haven, CT 06511, USA
| | - Juan Vasquez
- Section of Hematology & Oncology, Department of Pediatrics, Yale University School of Medicine, New Haven, CT 06520, USA
| | | |
Collapse
|
104
|
He M, Gu W, Gao Y, Liu Y, Liu J, Li Z. Molecular subtypes and a prognostic model for hepatocellular carcinoma based on immune- and immunogenic cell death-related lncRNAs. Front Immunol 2022; 13:1043827. [PMID: 36479122 PMCID: PMC9720162 DOI: 10.3389/fimmu.2022.1043827] [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: 09/14/2022] [Accepted: 11/04/2022] [Indexed: 11/22/2022] Open
Abstract
Background Accumulating evidence shows that immunogenic cell death (ICD) enhances immunotherapy effectiveness. In this study, we aimed to develop a prognostic model combining ICD, immunity, and long non-coding RNA biomarkers for predicting hepatocellular carcinoma (HCC) outcomes. Methods Immune- and immunogenic cell death-related lncRNAs (IICDLs) were identified from The Cancer Genome Atlas and Ensembl databases. IICDLs were extracted based on the results of differential expression and univariate Cox analyses and used to generate molecular subtypes using ConsensusClusterPlus. We created a prognostic signature based on IICDLs and a nomogram based on risk scores. Clinical characteristics, immune landscapes, immune checkpoint blocking (ICB) responses, stemness, and chemotherapy responses were also analyzed for different molecular subtypes and risk groups. Result A total of 81 IICDLs were identified, 20 of which were significantly associated with overall survival (OS) in patients with HCC. Cluster analysis divided patients with HCC into two distinct molecular subtypes (C1 and C2), with patients in C1 having a shorter survival time than those in C2. Four IICDLs (TMEM220-AS1, LINC02362, LINC01554, and LINC02499) were selected to develop a prognostic model that was an independent prognostic factor of HCC outcomes. C1 and the high-risk group had worse OS (hazard ratio > 1.5, p < 0.01), higher T stage (p < 0.05), higher clinical stage (p < 0.05), higher pathological grade (p < 0.05), low immune cell infiltration (CD4+ T cells, B cells, macrophages, neutrophils, and myeloid dendritic cells), low immune checkpoint gene expression, poor response to ICB therapy, and high stemness. Different molecular subtypes and risk groups showed significantly different responses to several chemotherapy drugs, such as doxorubicin (p < 0.001), 5-fluorouracil (p < 0.001), gemcitabine (p < 0.001), and sorafenib (p < 0.01). Conclusion Our study identified molecular subtypes and a prognostic signature based on IICDLs that could help predict the clinical prognosis and treatment response in patients with HCC.
Collapse
Affiliation(s)
- Mingang He
- Department of Gastrointestinal Surgery, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Wenchao Gu
- Department of Pulmonary and Critical Care Medicine, Shanghai Pudong New Area People’s Hospital, Shanghai, China
| | - Yang Gao
- Department of Gastrointestinal Surgery, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Ying Liu
- Department of Pathology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Jie Liu
- Cancer Center, Shandong Public Health Clinical Center, Public Health Clinical Center Affiliated to Shandong University, Jinan, China,*Correspondence: Jie Liu, ; Zengjun Li,
| | - Zengjun Li
- Department of Gastrointestinal Surgery, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China,*Correspondence: Jie Liu, ; Zengjun Li,
| |
Collapse
|
105
|
Identification of Immunogenic Cell Death-Related Signature for Glioma to Predict Survival and Response to Immunotherapy. Cancers (Basel) 2022; 14:cancers14225665. [PMID: 36428756 PMCID: PMC9688866 DOI: 10.3390/cancers14225665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 11/16/2022] [Accepted: 11/17/2022] [Indexed: 11/19/2022] Open
Abstract
Immunogenic cell death (ICD) is a type of regulated cell death (RCD) and is correlated with the progression, prognosis, and therapy of tumors, including glioma. Numerous studies have shown that the immunotherapeutic and chemotherapeutic agents of glioma might induce ICD. However, studies on the comprehensive analysis of the role of ICD-related genes and their correlations with overall survival (OS) in glioma are lacking. The genetic, transcriptional, and clinical data of 1896 glioma samples were acquired from five distinct databases and analyzed in terms of genes and transcription levels. The method of consensus unsupervised clustering divided the patients into two disparate molecular clusters: A and B. All of the patients were randomly divided into training and testing groups. Employing the training group data, 14 ICD-related genes were filtered out to develop a risk-score model. The correlations between our risk groups and prognosis, cells in the tumor microenvironment (TME) and immune cells infiltration, chemosensitivity and cancer stem cell (CSC) index were assessed. A highly precise nomogram model was constructed to enhance and optimize the clinical application of the risk score. The results demonstrated that the risk score could independently predict the OS rate and the immunotherapeutic response of glioma patients. This study analyzed the ICD-related genes in glioma and evaluated their role in the OS, clinicopathological characteristics, TME and immune cell infiltration of glioma. Our results may help in assessing the OS of glioma and developing better immunotherapeutic strategies.
Collapse
|
106
|
Florêncio KGD, Edson EA, Fernandes KSDS, Luiz JPM, Pinto FDCL, Pessoa ODL, Cunha FDQ, Machado-Neto JA, Wilke DV. Chromomycin A 5 induces bona fide immunogenic cell death in melanoma. Front Immunol 2022; 13:941757. [PMID: 36439184 PMCID: PMC9682167 DOI: 10.3389/fimmu.2022.941757] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 10/12/2022] [Indexed: 08/27/2023] Open
Abstract
PURPOSE Some first-line cytotoxic chemotherapics, e.g. doxorubicin, paclitaxel and oxaliplatin, induce activation of the immune system through immunogenic cell death (ICD). Tumor cells undergoing ICD function as a vaccine, releasing damage-associated molecular patterns (DAMPs), which act as adjuvants, and neoantigens of the tumor are recognized as antigens. ICD induction is rare, however it yields better and long-lasting antitumor responses to chemotherapy. Advanced metastatic melanoma (AMM) is incurable for more than half of patients. The discovery of ICD inducers against AMM is an interesting drug discovery strategy with high translational potential. Here we evaluated ICD induction of four highly cytotoxic chromomycins A (CA5-8). METHODS ICD features and DAMPs were evaluated using several in vitro techniques with metastatic melanoma cell line (B16-F10) exposed to chromomcins A5-8 such as flow cytometry, western blot, RT-PCR and luminescence. Additionally in vivo vaccination assays with CA5-treated cells in a syngeneic murine model (C57Bl/6) were performed to confirm ICD evaluating the immune cells activation and their antitumor activity. RESULTS B16-F10 treated with CA5-8 and doxorubicin exhibited ICD features such as autophagy and apoptosis, externalization of calreticulin, and releasing of HMGB1. However, CA5-treated cells had the best profile, also inducing ATP release, ERp57 externalization, phosphorylation of eIF2α and altering expression of transcription of genes related to autophagy, endoplasmic reticulum stress, and apoptosis. Bona fide ICD induction by CA5 was confirmed by vaccination of C57BL/6 mice with CA5-treated cells which activated antigen-presenting cells and T lymphocytes and stimulated antitumor activity. CONCLUSION CA5 induces bona fide immunogenic cell death on melanoma.
Collapse
Affiliation(s)
- Katharine Gurgel Dias Florêncio
- Drug Research and Development Center, Department of Physiology and Pharmacology, School of Medicine, Federal University of Ceara, Ceara, Brazil
| | - Evelline Araújo Edson
- Drug Research and Development Center, Department of Physiology and Pharmacology, School of Medicine, Federal University of Ceara, Ceara, Brazil
| | - Keilla Santana da Silva Fernandes
- Drug Research and Development Center, Department of Physiology and Pharmacology, School of Medicine, Federal University of Ceara, Ceara, Brazil
| | - João Paulo Mesquita Luiz
- Center for Research in Inflammatory Diseases (CRID), Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil
| | | | | | - Fernando de Queiroz Cunha
- Center for Research in Inflammatory Diseases (CRID), Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil
| | | | - Diego Veras Wilke
- Drug Research and Development Center, Department of Physiology and Pharmacology, School of Medicine, Federal University of Ceara, Ceara, Brazil
| |
Collapse
|
107
|
Lu Z, Chen J, Yu P, Atherton MJ, Gui J, Tomar VS, Middleton JD, Sullivan NT, Singhal S, George SS, Woolfork AG, Weljie AM, Hai T, Eruslanov EB, Fuchs SY. Tumor factors stimulate lysosomal degradation of tumor antigens and undermine their cross-presentation in lung cancer. Nat Commun 2022; 13:6623. [PMID: 36333297 PMCID: PMC9636202 DOI: 10.1038/s41467-022-34428-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Accepted: 10/25/2022] [Indexed: 11/06/2022] Open
Abstract
Activities of dendritic cells (DCs) that present tumor antigens are often suppressed in tumors. Here we report that this suppression is induced by tumor microenvironment-derived factors, which activate the activating transcription factor-3 (ATF3) transcription factor and downregulate cholesterol 25-hydroxylase (CH25H). Loss of CH25H in antigen presenting cells isolated from human lung tumors is associated with tumor growth and lung cancer progression. Accordingly, mice lacking CH25H in DCs exhibit an accelerated tumor growth, decreased infiltration and impaired activation of intratumoral CD8+ T cells. These mice do not establish measurable long-term immunity against malignant cells that undergo chemotherapy-induced immunogenic cell death. Mechanistically, downregulation of CH25H stimulates membrane fusion between endo-phagosomes and lysosomes, accelerates lysosomal degradation and restricts cross-presentation of tumor antigens in the intratumoral DCs. Administration of STING agonist MSA-2 reduces the lysosomal activity in DCs, restores antigen cross presentation, and increases therapeutic efficacy of PD-1 blockade against tumour challenge in a CH25H-dependent manner. These studies highlight the importance of downregulation of CH25H in DCs for tumor immune evasion and resistance to therapy.
Collapse
Affiliation(s)
- Zhen Lu
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Jinyun Chen
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Pengfei Yu
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Matthew J Atherton
- Department of Clinical Sciences & Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Jun Gui
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Vivek S Tomar
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Justin D Middleton
- Department of Biological Chemistry and Pharmacology, The Ohio State University, Columbus, OH, 43210, USA
| | - Neil T Sullivan
- Division of Thoracic Surgery, Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Sunil Singhal
- Division of Thoracic Surgery, Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Subin S George
- Institute for Biomedical Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Ashley G Woolfork
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Aalim M Weljie
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Tsonwin Hai
- Department of Biological Chemistry and Pharmacology, The Ohio State University, Columbus, OH, 43210, USA
| | - Evgeniy B Eruslanov
- Division of Thoracic Surgery, Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Serge Y Fuchs
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| |
Collapse
|
108
|
Mandula JK, Chang S, Mohamed E, Jimenez R, Sierra-Mondragon RA, Chang DC, Obermayer AN, Moran-Segura CM, Das S, Vazquez-Martinez JA, Prieto K, Chen A, Smalley KSM, Czerniecki B, Forsyth P, Koya RC, Ruffell B, Cubillos-Ruiz JR, Munn DH, Shaw TI, Conejo-Garcia JR, Rodriguez PC. Ablation of the endoplasmic reticulum stress kinase PERK induces paraptosis and type I interferon to promote anti-tumor T cell responses. Cancer Cell 2022; 40:1145-1160.e9. [PMID: 36150390 PMCID: PMC9561067 DOI: 10.1016/j.ccell.2022.08.016] [Citation(s) in RCA: 58] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 06/20/2022] [Accepted: 08/16/2022] [Indexed: 11/18/2022]
Abstract
Activation of unfolded protein responses (UPRs) in cancer cells undergoing endoplasmic reticulum (ER) stress promotes survival. However, how UPR in tumor cells impacts anti-tumor immune responses remains poorly described. Here, we investigate the role of the UPR mediator pancreatic ER kinase (PKR)-like ER kinase (PERK) in cancer cells in the modulation of anti-tumor immunity. Deletion of PERK in cancer cells or pharmacological inhibition of PERK in melanoma-bearing mice incites robust activation of anti-tumor T cell immunity and attenuates tumor growth. PERK elimination in ER-stressed malignant cells triggers SEC61β-induced paraptosis, thereby promoting immunogenic cell death (ICD) and systemic anti-tumor responses. ICD induction in PERK-ablated tumors stimulates type I interferon production in dendritic cells (DCs), which primes CCR2-dependent tumor trafficking of common-monocytic precursors and their intra-tumor commitment into monocytic-lineage inflammatory Ly6C+CD103+ DCs. These findings identify how tumor cell-derived PERK promotes immune evasion and highlight the potential of PERK-targeting therapies in cancer immunotherapy.
Collapse
Affiliation(s)
- Jessica K Mandula
- Department of Immunology, H. Lee Moffitt Cancer Center, Tampa, FL 33612, USA
| | - Shiun Chang
- Department of Immunology, H. Lee Moffitt Cancer Center, Tampa, FL 33612, USA
| | - Eslam Mohamed
- California Northstate University, Elk Grove, CA 95757, USA
| | - Rachel Jimenez
- Department of Immunology, H. Lee Moffitt Cancer Center, Tampa, FL 33612, USA
| | | | - Darwin C Chang
- Department of Immunology, H. Lee Moffitt Cancer Center, Tampa, FL 33612, USA
| | - Alyssa N Obermayer
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center, Tampa, FL 33612, USA
| | | | - Satyajit Das
- Department of Immunology, H. Lee Moffitt Cancer Center, Tampa, FL 33612, USA
| | | | - Karol Prieto
- Department of Immunology, H. Lee Moffitt Cancer Center, Tampa, FL 33612, USA
| | - Ann Chen
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center, Tampa, FL 33612, USA
| | - Keiran S M Smalley
- Department of Tumor Biology, H. Lee Moffitt Cancer Center, Tampa, FL 33612, USA
| | - Brian Czerniecki
- Department of Immunology, H. Lee Moffitt Cancer Center, Tampa, FL 33612, USA
| | - Peter Forsyth
- Department of NeuroOncology, H. Lee Moffitt Cancer Center, Tampa, FL 33612, USA
| | - Richard C Koya
- Department of Obstetrics and Gynecology, University of Chicago, Chicago, IL 60637, USA
| | - Brian Ruffell
- Department of Immunology, H. Lee Moffitt Cancer Center, Tampa, FL 33612, USA
| | - Juan R Cubillos-Ruiz
- Department of Obstetrics and Gynecology, Weill Cornell Medicine, New York, NY 10065, USA; Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY 10065, USA
| | - David H Munn
- Department of Pediatrics, Georgia Cancer Center, Augusta University, Augusta, GA 30912, USA
| | - Timothy I Shaw
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center, Tampa, FL 33612, USA
| | | | - Paulo C Rodriguez
- Department of Immunology, H. Lee Moffitt Cancer Center, Tampa, FL 33612, USA.
| |
Collapse
|
109
|
HIF-α activation by the prolyl hydroxylase inhibitor roxadustat suppresses chemoresistant glioblastoma growth by inducing ferroptosis. Cell Death Dis 2022; 13:861. [PMID: 36209275 PMCID: PMC9547873 DOI: 10.1038/s41419-022-05304-8] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 09/26/2022] [Accepted: 09/27/2022] [Indexed: 11/06/2022]
Abstract
Patients with glioblastoma (GBM) have poor prognosis and limited treatment options, largely due to therapy resistance upon the induction of apoptosis. Ferroptosis emerges as a potential antineoplastic strategy to bypass apoptosis resistance in traditional therapeutics. Hypoxia is a fundamental hallmark of GBM and hypoxia-inducible factor (HIF) is the main regulator of hypoxia response, however, the role of HIF has not been sufficiently explored in GBM. Herein, we first discovered that amplifying HIF signals by the prolyl hydroxylase (PHD) inhibitor roxadustat significantly suppressed GBM cell growth in vitro and in vivo, especially when the cells were resistant to temozolomide (TMZ). The accumulation of lipid peroxidation and cellular iron in GBM cells following roxadustat treatment indicated that the cells underwent ferroptosis, which was also supported by morphological changes in mitochondrial ultrastructure and immunogenic signals release. Moreover, in vivo studies further confirmed the ferroptosis induction and verified that roxadustat significantly prolonged survival of the mice harboring chemoresistant GBM without visible organ toxicity. Finally, we proved that the ferroptosis induction by roxadustat is HIF-α independent, especially activation of HIF-2α upregulating lipid regulatory genes was revealed to be mainly responsible for the enhanced lipid peroxidation. Altogether, our study provided novel evidence that amplifying HIF signals induced ferroptosis in chemoresistant GBM cells and suppressed the tumor growth in vivo, highlighting that ferroptosis induction by targeting HIF-α might provide new approaches to improve GBM treatment.
Collapse
|
110
|
Zhou Y, Hu F, Cui Y, Wu H, Hu S, Wei W. Bibliometric analysis of research on immunogenic cell death in cancer. Front Pharmacol 2022; 13:1029020. [PMID: 36278159 PMCID: PMC9582244 DOI: 10.3389/fphar.2022.1029020] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 09/23/2022] [Indexed: 11/18/2022] Open
Abstract
Background: Immunotherapy is changing the way we treat cancer. Immunogenic cell death (ICD) has received considerable attention in the treatments of various cancer types, due to the long-lasting antitumor responses elicited in human body. However, to date, no relevant bibliometric research has been reported. Methods: Publications related to ICD in cancer research were collected from the Web of Science Core Collection. Using CiteSpace, VOSviewer and an online platform, the analyses of co-author, co-citation, and co-occurrence of terms retrieved from literatures were carried out. Results: A total of 1,577 publications were included in this study. The global research literatures on ICD in cancer research have been increasing from 2005 to 2021. China, the United States and France dominated in this area and had close collaborations with many countries. Six of the top 10 most contributive institutions were from France. When it comes to author analysis, Kroemer G, Zitvogel L, Kepp O, Garg AD and Galluzzi L were in both the top 10 most productive authors and top 10 most co-cited authors lists. The co-occurring author keywords could be grouped into three clusters: “biomarkers of ICD”, “nanoparticles” and “combination therapy”. In terms of promising hotspots, keywords (author keywords and KeyWords Plus) with recent citation bursts could be summarized into two aspects: “tumor microenvironment” and “nanoparticles”. Conclusion: Increased attention has been paid to ICD in cancer treatment. However, there are still many unresolved domains in the field of ICD, such as clinical application and molecular mechanisms of this cell death process. ICD-inducing modalities combined with nanotechnology could potentiate the current immunotherapies, and will be hotspots for future research.
Collapse
Affiliation(s)
- Yan Zhou
- Clinical College of Neurology, Neurosurgery and Neurorehabilitation, Tianjin Medical University, Tianjin, China
| | - Fen Hu
- Department of Oncology, XiangYang Central Hospital, Hubei University of Arts and Science, XiangYang, China
- Institute of Oncology, XiangYang Central Hospital, Hubei University of Arts and Science, XiangYang, China
| | - Yang Cui
- Department of Neurosurgery, Hebei Yanda Hospital, Langfang, China
| | - Haiyang Wu
- Clinical College of Neurology, Neurosurgery and Neurorehabilitation, Tianjin Medical University, Tianjin, China
| | - Shunan Hu
- Department of Neurosurgery, XiangYang Central Hospital, Hubei University of Arts and Science, XiangYang, China
- *Correspondence: Shunan Hu, ; Wei Wei,
| | - Wei Wei
- Department of Oncology, XiangYang Central Hospital, Hubei University of Arts and Science, XiangYang, China
- Institute of Oncology, XiangYang Central Hospital, Hubei University of Arts and Science, XiangYang, China
- *Correspondence: Shunan Hu, ; Wei Wei,
| |
Collapse
|
111
|
Doroshenko A, Tomkova S, Kozar T, Stroffekova K. Hypericin, a potential new BH3 mimetic. Front Pharmacol 2022; 13:991554. [PMID: 36267274 PMCID: PMC9577225 DOI: 10.3389/fphar.2022.991554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Accepted: 08/30/2022] [Indexed: 11/13/2022] Open
Abstract
Many types of cancer such as prostate cancer, myeloid leukemia, breast cancer, glioblastoma display strong chemo resistance, which is supported by enhanced expression of multiple anti-apoptotic Bcl-2, Bcl-XL and Mcl-1 proteins. The viable anti-cancer strategies are based on developing anti-apoptotic Bcl-2 proteins inhibitors, BH3 mimetics. Our focus in past years has been on the investigating a new potential BH3 mimetic, Hypericin (Hyp). Hyp is a naturally occurring photosensitive compound used in photodynamic therapy and diagnosis. We have demonstrated that Hyp can cause substantial effects in cellular ultrastructure, mitochondria function and metabolism, and distribution of Bcl2 proteins in malignant and non-malignant cells. One of the possible mechanisms of Hyp action could be the direct interactions between Bcl-2 proteins and Hyp. We investigated this assumption by in silico computer modelling and in vitro fluorescent spectroscopy experiments with the small Bcl2 peptide segments designed to correspond to Bcl2 BH3 and BH1 domains. We show here that Hyp interacts with BH3 and BH1 peptides in concentration dependent manner, and shows the stronger interactions than known BH3 mimetics, Gossypol (Goss) and ABT-263. In addition, interactions of Hyp, Goss and ABT263, with whole purified proteins Bcl-2 and Mcl-1 by fluorescence spectroscopy show that Hyp interacts stronger with the Bcl-2 and less with Mcl-1 protein than Goss or ABT-263. This suggest that Hyp is comparable to other BH3 mimetics and could be explore as such. Hyp cytotoxicity was low in human U87 MG glioma, similar to that of ABT263, where Goss exerted sufficient cytotoxicity, suggesting that Hyp acts primarily on Bcl-2, but not on Mcl-1 protein. In combination therapy, low doses of Hyp with Goss effectively decreased U87 MG viability, suggesting a possible synergy effect. Overall, we can conclude that Hyp as BH3 mimetic acts primarily on Bcl-2 protein and can be explored to target cells with Bcl-2 over-expression, or in combination with other BH3 mimetics, that target Mcl-1 or Bcl-XL proteins, in dual therapy.
Collapse
Affiliation(s)
- Anastasia Doroshenko
- Department of Biophysics, Faculty of Natural Sciences, PJ Safarik University, Kosice, Slovakia
| | - Silvia Tomkova
- Department of Biophysics, Faculty of Natural Sciences, PJ Safarik University, Kosice, Slovakia
| | - Tibor Kozar
- Center of Interdisciplinary Biosciences, TIP-Safarik University, Kosice, Slovakia
| | - Katarina Stroffekova
- Department of Biophysics, Faculty of Natural Sciences, PJ Safarik University, Kosice, Slovakia
- *Correspondence: Katarina Stroffekova,
| |
Collapse
|
112
|
Curcumin Enhanced Ionizing Radiation-Induced Immunogenic Cell Death in Glioma Cells through Endoplasmic Reticulum Stress Signaling Pathways. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:5424411. [PMID: 36238646 PMCID: PMC9553401 DOI: 10.1155/2022/5424411] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 09/08/2022] [Accepted: 09/13/2022] [Indexed: 11/17/2022]
Abstract
Objective Local radiotherapy may cause distant tumor regression via inducing immunogenic cell death (ICD). Here, we investigated the effect of curcumin on ionizing radiation-induced immunogenic cell death in normoxic or hypoxic glioma cells and its mechanism in vitro and vivo. Methods Hypoxic or normoxic glioma cell apoptosis and the cell surface exposure of calreticulin (CRT) were detected by flow cytometry. Extracellular ATP and HSP70 were measured by chemiluminescence assay and ELISA, respectively. Endoplasmic reticulum (ER) stress protein levels were detected by western blot. Moreover, the induction of bona fide ICD was detected by vaccination assays in mice bearing glioma model. Spleen lymphocytes and tumor-infiltrating lymphocyte subsets were analyzed by flow cytometry and immunohistochemistry. Results Curcumin incubation before X-ray irradiation significantly increased radiation-induced apoptosis rate in normoxic or hypoxic glioma cells. Curcumin enhanced radiation-induced CRT exposure, release of HSP70 and ATP, and ER stress signaling activity. After treatment with ER stress pathway inhibitors, cell apoptosis and CRT exposure induced by the combination treatment of curcumin and X-ray were reduced. In vaccination experiments, glioma cells irradiated by X-ray produced a strong immunogenic response rejecting tumor formation in 70% mice. In comparison, cells treated by curcumin and X-ray produced a stronger immune response rejecting tumor formation in 90% mice. The combination treatment increased the percentage of tumor-infiltrating CD4+, CD8+ T lymphocytes, and CD11c+ dendritic cells compared to X-ray irradiation alone. Conclusion Ionizing radiation-induced normoxic or hypoxic glioma immunogenic cell death could be further enhanced by curcumin through activating the ER stress PERK-eIF2α and IRE1α-XBP1 signaling pathways.
Collapse
|
113
|
Hengst JA, Nduwumwami AJ, Raup-Konsavage WM, Vrana KE, Yun JK. Inhibition of Sphingosine Kinase Activity Enhances Immunogenic Cell Surface Exposure of Calreticulin Induced by the Synthetic Cannabinoid 5-epi-CP-55,940. Cannabis Cannabinoid Res 2022; 7:637-647. [PMID: 34846947 PMCID: PMC9587795 DOI: 10.1089/can.2021.0100] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Background: Endogenous and synthetic cannabinoids have been shown to induce cancer cell death through the accumulation of the sphingolipid, ceramide (Cer). Recently, we have demonstrated that Cer accumulation enhances the induction of immunogenic cell death (ICD). Objectives: The primary objective of this study was to demonstrate that (±) 5-epi CP 55,940 (5-epi), a by-product of the chemical synthesis of the synthetic cannabinoid CP 55,940, induces ICD in colorectal cancer (CRC) cells, and that modulation of the sphingolipid metabolic pathway through inhibition of the sphingosine kinases (SphKs) enhances these effects. Methods: A cell culture model system of human CRC cell lines was employed to measure the cell surface and intracellular production of markers of ICD. The effects of 5-epi, alone and in combination with SphK inhibitors, on production of Cer through the de novo sphingolipid synthesis pathway were measured by Liquid Chromatography - Tandem Mass Spectrometry (LC/MS/MS)-based sphingolipidomic analysis. Cell surface exposure of calreticulin (ectoCRT), a hallmark of ICD, was measured by flow cytometry. Examination of the effects of 5-epi, alone and in combination with SphK inhibitors, on the intracellular signaling pathway associated with ICD was conducted by immunoblot analysis of human CRC cell lines. Results: Sphingolipidomic analysis indicated that 5-epi induces the de novo sphingolipid synthetic pathway. 5-epi dose dependently induces cell surface exposure of ectoCRT, and inhibition of Cer metabolism through inhibition of the SphKs significantly enhances 5-epi-induced ectoCRT exposure in multiple CRC cell lines. 5-epi induces and SphK inhibition enhances activation of the cell death signaling pathway associated with ICD. Conclusions: This study is the first demonstration that cannabinoids can induce the cell surface expression of ectoCRT, and potentially induce ICD. Moreover, this study reinforces our previous observation of a role for Cer accumulation in the induction of ICD and extends this observation to the cannabinoids, agents not typically associated with ICD. Inhibition of SphKs enhanced the 5-epi-induced signaling pathways leading to ICD and production of ectoCRT. Overexpression of SphK1 has previously been associated with chemotherapy resistance. Thus, targeting the SphKs has the potential to reverse chemotherapy resistance and simultaneously enhance the antitumor immune response through enhancement of ICD induction.
Collapse
Affiliation(s)
- Jeremy A. Hengst
- Department of Pharmacology, Pennsylvania State University College of Medicine, Hershey, Pennsylvania, USA
| | - Asvelt J. Nduwumwami
- Department of Pharmacology, Pennsylvania State University College of Medicine, Hershey, Pennsylvania, USA
| | - Wesley M. Raup-Konsavage
- Department of Pharmacology, Pennsylvania State University College of Medicine, Hershey, Pennsylvania, USA
| | - Kent E. Vrana
- Department of Pharmacology, Pennsylvania State University College of Medicine, Hershey, Pennsylvania, USA
| | - Jong K. Yun
- Department of Pharmacology, Pennsylvania State University College of Medicine, Hershey, Pennsylvania, USA
| |
Collapse
|
114
|
Hageman E, Che PP, Dahele M, Slotman BJ, Sminia P. Radiobiological Aspects of FLASH Radiotherapy. Biomolecules 2022; 12:biom12101376. [PMID: 36291585 PMCID: PMC9599153 DOI: 10.3390/biom12101376] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 09/21/2022] [Accepted: 09/22/2022] [Indexed: 11/16/2022] Open
Abstract
Radiotherapy (RT) is one of the primary treatment modalities for cancer patients. The clinical use of RT requires a balance to be struck between tumor effect and the risk of toxicity. Sparing normal tissue is the cornerstone of reducing toxicity. Advances in physical targeting and dose-shaping technology have helped to achieve this. FLASH RT is a promising, novel treatment technique that seeks to exploit a potential normal tissue-sparing effect of ultra-high dose rate irradiation. A significant body of in vitro and in vivo data has highlighted a decrease in acute and late radiation toxicities, while preserving the radiation effect in tumor cells. The underlying biological mechanisms of FLASH RT, however, remain unclear. Three main mechanisms have been hypothesized to account for this differential FLASH RT effect between the tumor and healthy tissue: the oxygen depletion, the DNA damage, and the immune-mediated hypothesis. These hypotheses and molecular mechanisms have been evaluated both in vitro and in vivo. Furthermore, the effect of ultra-high dose rate radiation with extremely short delivery times on the dynamic tumor microenvironment involving circulating blood cells and immune cells in humans is essentially unknown. Therefore, while there is great interest in FLASH RT as a means of targeting tumors with the promise of an increased therapeutic ratio, evidence of a generalized FLASH effect in humans and data to show that FLASH in humans is safe and at least effective against tumors as standard photon RT is currently lacking. FLASH RT needs further preclinical investigation and well-designed in-human studies before it can be introduced into clinical practice.
Collapse
Affiliation(s)
- Eline Hageman
- Amsterdam UMC Location Vrije Universiteit Amsterdam, Radiation Oncology, Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, 1081 HV Amsterdam, The Netherlands
| | - Pei-Pei Che
- Amsterdam UMC Location Vrije Universiteit Amsterdam, Radiation Oncology, Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, 1081 HV Amsterdam, The Netherlands
| | - Max Dahele
- Amsterdam UMC Location Vrije Universiteit Amsterdam, Radiation Oncology, Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
| | - Ben J. Slotman
- Amsterdam UMC Location Vrije Universiteit Amsterdam, Radiation Oncology, Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
| | - Peter Sminia
- Amsterdam UMC Location Vrije Universiteit Amsterdam, Radiation Oncology, Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, 1081 HV Amsterdam, The Netherlands
- Correspondence:
| |
Collapse
|
115
|
Imaging Effector Memory T-Cells Predicts Response to PD1-Chemotherapy Combinations in Colon Cancer. Biomedicines 2022; 10:biomedicines10102343. [PMID: 36289605 PMCID: PMC9598730 DOI: 10.3390/biomedicines10102343] [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: 08/18/2022] [Revised: 09/14/2022] [Accepted: 09/18/2022] [Indexed: 11/16/2022] Open
Abstract
Often, patients fail to respond to immune checkpoint inhibitor (ICI) treatment despite favourable biomarker status. Numerous chemotherapeutic agents have been shown to promote tumour immunogenicity when used in conjunction with ICIs; however, little is known about whether such combination therapies lead to a lasting immune response. Given the potential toxicity of ICI–chemotherapy combinations, identification of biomarkers that accurately predict how individuals respond to specific treatment combinations and whether these responses will be long lasting is of paramount importance. In this study, we explored [18F]AlF-NOTA-KCNA3P, a peptide radiopharmaceutical that targets the Kv1.3 potassium channel overexpressed on T-effector memory (TEM) cells as a PET imaging biomarker for lasting immunological memory response. The first-line colon cancer chemotherapies oxaliplatin and 5-fluorouracil were assessed in a syngeneic colon cancer model, either as monotherapies or in combination with PD1, comparing radiopharmaceutical uptake to memory-associated immune cells in the tumour. [18F]AlF-NOTA-KCNA3P reliably separated tumours with immunological memory responses from non-responding tumours and could be used to measure Kv1.3-expressing TEM cells responsible for durable immunological memory response to combination therapy in vivo.
Collapse
|
116
|
He J, Zheng P, Chen Y, Qi J, Ye C, Li D, Yang Y, Yang Y, Liu Q, Hu Y, Zheng X, Li W, Hua L, Yang Z, Chen H, Huang W, Sun W, Yang X, Long Q, Bai H, Ma Y. A new personalized vaccine strategy based on inducing the pyroptosis of tumor cells in vivo by transgenic expression of a truncated GSDMD N-terminus. Front Immunol 2022; 13:991857. [PMID: 36189310 PMCID: PMC9521720 DOI: 10.3389/fimmu.2022.991857] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 08/29/2022] [Indexed: 12/02/2022] Open
Abstract
The variability and heterogeneity of tumor antigens and the tumor-driven development of immunosuppressive mechanisms leading to tumor escape from established immunological surveillance. Here, the tumor cells were genetically modified to achieve an inducible overexpression of the N-terminal domain of gasdermin D (GSDMD-NT) and effectively cause pyroptosis under a strict control. Pyroptotic tumor cells release damage-associated molecular patterns (DAMPs) and inflammatory cytokines to promote the maturation and migration of bone marrow-derived dendritic cells (BMDCs). Furthermore, local tumor delivery, and preventive or therapeutic subcutaneous immunization of the modified cells, followed by the induction of GSDMD-NT expression, significantly stimulated both the systemic and local responses of antitumor immunity, and reprogrammed the tumor microenvironment, leading to the dramatic suppression of tumor growth in mice. This study has explored the application potency of inducing the pyroptosis of tumor cells in the field of tumor immunotherapy, especially for developing a new and promising personalized tumor vaccine.
Collapse
Affiliation(s)
- Jinrong He
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, China
| | - Peng Zheng
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, China
| | - Yongjun Chen
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, China
| | - Jialong Qi
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, China
- Yunnan Digestive Endoscopy Clinical Medical Center, Department of Gastroenterology, The First People’s Hospital of Yunnan Province, Kunming, China
| | - Chao Ye
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, China
| | - Duo Li
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, China
- Department of Acute Infectious Diseases Control and Prevention, Yunnan Provincial Center for Disease Control and Prevention, Kunming, China
| | - Ying Yang
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, China
- Institute of Medical Biology, Kunming Medical University, Kunming, China
| | - Ying Yang
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, China
| | - Qingwen Liu
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, China
- Institute of Medical Biology, Kunming Medical University, Kunming, China
| | - Yongmao Hu
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, China
- School of Life Sciences, Yunnan University, Kunming, China
| | - Xiao Zheng
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, China
- School of Life Sciences, Yunnan University, Kunming, China
| | - Weiran Li
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, China
| | - Liangqun Hua
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, China
- School of Life Sciences, Yunnan University, Kunming, China
| | - Zhongqian Yang
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, China
| | - Haoqian Chen
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, China
- School of Ethnic Medicine, Yunnan Minzu University, Kunming, China
| | - Weiwei Huang
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, China
| | - Wenjia Sun
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, China
| | - Xu Yang
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, China
| | - Qiong Long
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, China
| | - Hongmei Bai
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, China
| | - Yanbing Ma
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, China
- *Correspondence: Yanbing Ma,
| |
Collapse
|
117
|
Muppala V, Farran B, Nagaraju GP. Pyroptosis-based nanotherapeutics: Possible mechanisms for cancer treatment. Life Sci 2022; 308:120970. [PMID: 36115581 DOI: 10.1016/j.lfs.2022.120970] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 09/08/2022] [Accepted: 09/12/2022] [Indexed: 11/16/2022]
Abstract
Pyroptosis represents an inflammatory cell death form induced by inflammasomes and performed by gasdermins. It is characterized by swelling, pore formation, release of cellular content and the activation of innate immunity leading to inflammation. Hence, pyroptosis contributes to inflammatory conditions like cancer and has emerged as a promising immuno-strategy for treating cancer. The advent of nanotechnology, which overlaps with the discovery of pyroptotic cell death, has enabled the development of nano-based pyroptosis inducing platforms aimed at overcoming resistance to apoptosis and enhancing tumor immunity. In this paper, we will describe the various molecular pathways underlying pyroptosis, such as canonical and non-canonical pyroptosis. We will then explore the advances in the field of pyroptosis-based nanotherapeutics and their future implications.
Collapse
Affiliation(s)
- Veda Muppala
- Department of Hematology and Oncology, Heersink School of Medicine, The University of Alabama, Birmingham, AL 35294, USA
| | - Batoul Farran
- Department of Oncology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Ganji Purnachandra Nagaraju
- Department of Hematology and Oncology, Heersink School of Medicine, The University of Alabama, Birmingham, AL 35294, USA.
| |
Collapse
|
118
|
De Azevedo J, Mourtada J, Bour C, Devignot V, Schultz P, Borel C, Pencreach E, Mellitzer G, Gaiddon C, Jung AC. The EXTREME Regimen Associating Cetuximab and Cisplatin Favors Head and Neck Cancer Cell Death and Immunogenicity with the Induction of an Anti-Cancer Immune Response. Cells 2022; 11:cells11182866. [PMID: 36139440 PMCID: PMC9496761 DOI: 10.3390/cells11182866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 09/09/2022] [Accepted: 09/12/2022] [Indexed: 12/24/2022] Open
Abstract
(1) Background: The first line of treatment for recurrent/metastatic Head and Neck Squamous Cell Carcinoma (HNSCC) has recently evolved with the approval of immunotherapies that target the anti-PD-1 immune checkpoint. However, only about 20% of the patients display a long-lasting objective tumor response. The modulation of cancer cell immunogenicity via a treatment-induced immunogenic cell death is proposed to potentially be able to improve the rate of patients who respond to immune checkpoint blocking immunotherapies. (2) Methods: Using human HNSCC cell line models and a mouse oral cancer syngeneic model, we have analyzed the ability of the EXTREME regimen (combination therapy using the anti-EGFR cetuximab antibody and platinum-based chemotherapy) to modify the immunogenicity of HNSCC cells. (3) Results: We showed that the combination of cetuximab and cisplatin reduces cell growth through both cell cycle inhibition and the induction of apoptotic cell death independently of p53. In addition, different components of the EXTREME regimen were found to induce, to a variable extent, and in a cell-dependent manner, the emission of mediators of immunogenic cell death, including calreticulin, HMGB1, and type I Interferon-responsive chemokines. Interestingly, cetuximab alone or combined with the IC50 dose of cisplatin can induce an antitumor immune response in vivo, but not when combined with a high dose of cisplatin. (4) Conclusions: Our observations suggest that the EXTREME protocol or cetuximab alone are capable, under conditions of moderate apoptosis induction, of eliciting the mobilization of the immune system and an anti-tumor immune response in HNSCC.
Collapse
Affiliation(s)
- Justine De Azevedo
- Laboratory Streinth, Université de Strasbourg-Inserm, UMR_S 1113 IRFAC, 67200 Strasbourg, France
| | - Jana Mourtada
- Laboratory Streinth, Université de Strasbourg-Inserm, UMR_S 1113 IRFAC, 67200 Strasbourg, France
| | - Cyril Bour
- Laboratory Streinth, Université de Strasbourg-Inserm, UMR_S 1113 IRFAC, 67200 Strasbourg, France
- Laboratoire de Biologie Tumorale, Institut de Cancérologie Strasbourg Europe, 67200 Strasbourg, France
| | - Véronique Devignot
- Laboratory Streinth, Université de Strasbourg-Inserm, UMR_S 1113 IRFAC, 67200 Strasbourg, France
| | - Philippe Schultz
- Laboratory Streinth, Université de Strasbourg-Inserm, UMR_S 1113 IRFAC, 67200 Strasbourg, France
- Department of Otorhinolaryngology and Head and Neck Surgery, Hôpitaux Universitaires de Strasbourg, 67200 Strasbourg, France
| | - Christian Borel
- Laboratory Streinth, Université de Strasbourg-Inserm, UMR_S 1113 IRFAC, 67200 Strasbourg, France
- Department of Medical Oncology, Institut de Cancérologie Strasbourg Europe, 67200 Strasbourg, France
| | - Erwan Pencreach
- Laboratory Streinth, Université de Strasbourg-Inserm, UMR_S 1113 IRFAC, 67200 Strasbourg, France
- Laboratoire de Biochimie et Biologie Moléculaire, Hôpitaux Universitaires de Strasbourg, 67200 Strasbourg, France
| | - Georg Mellitzer
- Laboratory Streinth, Université de Strasbourg-Inserm, UMR_S 1113 IRFAC, 67200 Strasbourg, France
| | - Christian Gaiddon
- Laboratory Streinth, Université de Strasbourg-Inserm, UMR_S 1113 IRFAC, 67200 Strasbourg, France
- Correspondence: (C.G.); (A.C.J.)
| | - Alain C. Jung
- Laboratory Streinth, Université de Strasbourg-Inserm, UMR_S 1113 IRFAC, 67200 Strasbourg, France
- Laboratoire de Biologie Tumorale, Institut de Cancérologie Strasbourg Europe, 67200 Strasbourg, France
- Correspondence: (C.G.); (A.C.J.)
| |
Collapse
|
119
|
Alonso-Miguel D, Fiering S, Arias-Pulido H. Proactive Immunotherapeutic Approaches against Inflammatory Breast Cancer May Improve Patient Outcomes. Cells 2022; 11:2850. [PMID: 36139425 PMCID: PMC9497132 DOI: 10.3390/cells11182850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Revised: 09/07/2022] [Accepted: 09/09/2022] [Indexed: 11/16/2022] Open
Abstract
Inflammatory breast cancer (IBC) is highly metastatic at the onset of the disease with no IBC-specific treatments, resulting in dismal patient survival. IBC treatment is a clear unmet clinical need. This commentary highlights findings from a recent seminal approach in which pembrolizumab, a checkpoint inhibitor against programmed cell death protein 1 (PD-1), was provided to a triple-negative IBC patient as a neoadjuvant immune therapy combined with anthracycline-taxane-based chemotherapy. We highlight the findings of the case report and offer a perspective on taking a proactive approach to deploy approved immune checkpoint inhibitors. On the basis of our recently published research study, we propose in situ vaccination with direct injection of immunostimulatory agents into the tumor as an option to improve outcomes safely, effectively, and economically for IBC patients.
Collapse
Affiliation(s)
- Daniel Alonso-Miguel
- Department of Animal Medicine and Surgery, Veterinary Medicine School, Complutense University of Madrid, 28040 Madrid, Spain
| | - Steven Fiering
- Department of Microbiology and Immunology, and Dartmouth Cancer Center, Geisel School of Medicine at Dartmouth and Dartmouth Hitchcock Health, Lebanon, NH 03756, USA
| | - Hugo Arias-Pulido
- Department of Microbiology and Immunology, and Dartmouth Cancer Center, Geisel School of Medicine at Dartmouth and Dartmouth Hitchcock Health, Lebanon, NH 03756, USA
| |
Collapse
|
120
|
Ischemia and reperfusion injury combined with cisplatin induces immunogenic cell death in lung cancer cells. Cell Death Dis 2022; 13:764. [PMID: 36057637 PMCID: PMC9440929 DOI: 10.1038/s41419-022-05176-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Revised: 08/08/2022] [Accepted: 08/10/2022] [Indexed: 01/21/2023]
Abstract
A first-line chemotherapeutic drug for non-small cell lung cancer (NSCLC), cisplatin (CDDP), fails to induce immunogenic cell death (ICD) because it fails to induce calreticulin (CRT) exposure on the cell surface. We investigated the potential of ischemia and reperfusion injury (I/R) combined with CDDP to induce ICD in lung cancer cells. The in vitro model of I/R, oxygen-glucose deprivation and reperfusion (OGD/R), effectively induced CRT exposure, ATP secretion, high mobility group box 1 (HMGB1) release and eIF2α phosphorylation in both Lewis lung carcinoma (LLC) and A549 cells when combined with CDDP. By using a vaccine assay and coculture with bone marrow-derived dendritic cells (BMDCs), we showed that OGD/R restored the immunogenicity of CDDP by phosphorylating eIF2α and demonstrated that OGD/R + CDDP (O + C) is an ICD inducer. Using the inguinal tumor model, we found that I/R significantly enhanced the tumor-killing effect of CDDP and Mitomycin C, and this effect relied on adaptive antitumor immunity. Consistently, I + C altered the ratio of interferon-gamma-secreting T lymphocytes, thus overcoming the immunosuppressive effect induced by CDDP. In conclusion, our research presents a new combination strategy and indicates that I/R is a potential anticancer immunogenic modality when combined with nonimmunogenic chemotherapy.
Collapse
|
121
|
Mussafi O, Mei J, Mao W, Wan Y. Immune checkpoint inhibitors for PD-1/PD-L1 axis in combination with other immunotherapies and targeted therapies for non-small cell lung cancer. Front Oncol 2022; 12:948405. [PMID: 36059606 PMCID: PMC9430651 DOI: 10.3389/fonc.2022.948405] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 07/22/2022] [Indexed: 12/13/2022] Open
Abstract
It has been widely acknowledged that the use of immune checkpoint inhibitors (ICI) is an effective therapeutic treatment in many late-stage cancers. However, not all patients could benefit from ICI therapy. Several biomarkers, such as high expression of PD-L1, high mutational burden, and higher number of tumor infiltration lymphocytes have shown to predict clinical benefit from immune checkpoint therapies. One approach using ICI in combination with other immunotherapies and targeted therapies is now being investigated to enhance the efficacy of ICI alone. In this review, we summarized the use of other promising immunotherapies and targeted therapies in combination with ICI in treatment of lung cancers. The results from multiple animals and clinical trials were reviewed. We also briefly discussed the possible outlooks for future treatment.
Collapse
Affiliation(s)
- Ofek Mussafi
- Department of Cardiothoracic Surgery, The Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi, China
- The Pq Laboratory of BiomeDx/Rx, Department of Biomedical Engineering, Binghamton University-SUNY, Binghamton, NY, United States
| | - Jie Mei
- Department of Oncology, The Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi, China
| | - Wenjun Mao
- Department of Cardiothoracic Surgery, The Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi, China
| | - Yuan Wan
- The Pq Laboratory of BiomeDx/Rx, Department of Biomedical Engineering, Binghamton University-SUNY, Binghamton, NY, United States
| |
Collapse
|
122
|
Li Z, Lai X, Fu S, Ren L, Cai H, Zhang H, Gu Z, Ma X, Luo K. Immunogenic Cell Death Activates the Tumor Immune Microenvironment to Boost the Immunotherapy Efficiency. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2201734. [PMID: 35652198 PMCID: PMC9353475 DOI: 10.1002/advs.202201734] [Citation(s) in RCA: 242] [Impact Index Per Article: 80.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 04/21/2022] [Indexed: 02/05/2023]
Abstract
Tumor immunotherapy is only effective in a fraction of patients due to a low response rate and severe side effects, and these challenges of immunotherapy in clinics can be addressed through induction of immunogenic cell death (ICD). ICD is elicited from many antitumor therapies to release danger associated molecular patterns (DAMPs) and tumor-associated antigens to facilitate maturation of dendritic cells (DCs) and infiltration of cytotoxic T lymphocytes (CTLs). The process can reverse the tumor immunosuppressive microenvironment to improve the sensitivity of immunotherapy. Nanostructure-based drug delivery systems (NDDSs) are explored to induce ICD by incorporating therapeutic molecules for chemotherapy, photosensitizers (PSs) for photodynamic therapy (PDT), photothermal conversion agents for photothermal therapy (PTT), and radiosensitizers for radiotherapy (RT). These NDDSs can release loaded agents at a right dose in the right place at the right time, resulting in greater effectiveness and lower toxicity. Immunotherapeutic agents can also be combined with these NDDSs to achieve the synergic antitumor effect in a multi-modality therapeutic approach. In this review, NDDSs are harnessed to load multiple agents to induce ICD by chemotherapy, PDT, PTT, and RT in combination of immunotherapy to promote the therapeutic effect and reduce side effects associated with cancer treatment.
Collapse
Affiliation(s)
- Zhilin Li
- Department of BiotherapyHuaxi MR Research Center (HMRRC)Day Surgery CenterDepartment of RadiologyCancer CenterResearch Core Facilities of West China HospitalNational Clinical Research Center for GeriatricsFrontiers Science Center for Disease‐Related Molecular NetworkState Key Laboratory of BiotherapyWest China HospitalSichuan UniversityChengdu610041China
| | - Xiaoqin Lai
- Department of BiotherapyHuaxi MR Research Center (HMRRC)Day Surgery CenterDepartment of RadiologyCancer CenterResearch Core Facilities of West China HospitalNational Clinical Research Center for GeriatricsFrontiers Science Center for Disease‐Related Molecular NetworkState Key Laboratory of BiotherapyWest China HospitalSichuan UniversityChengdu610041China
| | - Shiqin Fu
- Department of BiotherapyHuaxi MR Research Center (HMRRC)Day Surgery CenterDepartment of RadiologyCancer CenterResearch Core Facilities of West China HospitalNational Clinical Research Center for GeriatricsFrontiers Science Center for Disease‐Related Molecular NetworkState Key Laboratory of BiotherapyWest China HospitalSichuan UniversityChengdu610041China
| | - Long Ren
- Department of BiotherapyHuaxi MR Research Center (HMRRC)Day Surgery CenterDepartment of RadiologyCancer CenterResearch Core Facilities of West China HospitalNational Clinical Research Center for GeriatricsFrontiers Science Center for Disease‐Related Molecular NetworkState Key Laboratory of BiotherapyWest China HospitalSichuan UniversityChengdu610041China
| | - Hao Cai
- Department of BiotherapyHuaxi MR Research Center (HMRRC)Day Surgery CenterDepartment of RadiologyCancer CenterResearch Core Facilities of West China HospitalNational Clinical Research Center for GeriatricsFrontiers Science Center for Disease‐Related Molecular NetworkState Key Laboratory of BiotherapyWest China HospitalSichuan UniversityChengdu610041China
| | - Hu Zhang
- Department of BiotherapyHuaxi MR Research Center (HMRRC)Day Surgery CenterDepartment of RadiologyCancer CenterResearch Core Facilities of West China HospitalNational Clinical Research Center for GeriatricsFrontiers Science Center for Disease‐Related Molecular NetworkState Key Laboratory of BiotherapyWest China HospitalSichuan UniversityChengdu610041China
- Amgen Bioprocessing CentreKeck Graduate InstituteClaremontCA91711USA
| | - Zhongwei Gu
- Department of BiotherapyHuaxi MR Research Center (HMRRC)Day Surgery CenterDepartment of RadiologyCancer CenterResearch Core Facilities of West China HospitalNational Clinical Research Center for GeriatricsFrontiers Science Center for Disease‐Related Molecular NetworkState Key Laboratory of BiotherapyWest China HospitalSichuan UniversityChengdu610041China
| | - Xuelei Ma
- Department of BiotherapyHuaxi MR Research Center (HMRRC)Day Surgery CenterDepartment of RadiologyCancer CenterResearch Core Facilities of West China HospitalNational Clinical Research Center for GeriatricsFrontiers Science Center for Disease‐Related Molecular NetworkState Key Laboratory of BiotherapyWest China HospitalSichuan UniversityChengdu610041China
| | - Kui Luo
- Department of BiotherapyHuaxi MR Research Center (HMRRC)Day Surgery CenterDepartment of RadiologyCancer CenterResearch Core Facilities of West China HospitalNational Clinical Research Center for GeriatricsFrontiers Science Center for Disease‐Related Molecular NetworkState Key Laboratory of BiotherapyWest China HospitalSichuan UniversityChengdu610041China
- Functional and Molecular Imaging Key Laboratory of Sichuan Provinceand Research Unit of PsychoradiologyChinese Academy of Medical SciencesChengdu610041China
| |
Collapse
|
123
|
Li Y, Palmer A, Lupu L, Huber-Lang M. Inflammatory response to the ischaemia-reperfusion insult in the liver after major tissue trauma. Eur J Trauma Emerg Surg 2022; 48:4431-4444. [PMID: 35831749 DOI: 10.1007/s00068-022-02026-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 05/28/2022] [Indexed: 11/29/2022]
Abstract
BACKGROUND Polytrauma is often accompanied by ischaemia-reperfusion injury to tissues and organs, and the resulting series of immune inflammatory reactions are a major cause of death in patients. The liver is one of the largest organs in the body, a characteristic that makes it the most vulnerable organ after multiple injuries. In addition, the liver is an important digestive organ that secretes a variety of inflammatory mediators involved in local as well as systemic immune inflammatory responses. Therefore, this review considers the main features of post-traumatic liver injury, focusing on the immuno-pathophysiological changes, the interactions between liver organs, and the principles of treatment deduced. METHODS We focus on the local as well as systemic immune response involving the liver after multiple injuries, with emphasis on the pathophysiological mechanisms. RESULTS An overview of the mechanisms underlying the pathophysiology of local as well as systemic immune responses involving the liver after multiple injuries, the latest research findings, and the current mainstream therapeutic approaches. CONCLUSION Cross-reactivity between various organs and cascade amplification effects are among the main causes of systemic immune inflammatory responses after multiple injuries. For the time being, the pathophysiological mechanisms underlying this interaction remain unclear. Future work will continue to focus on identifying potential signalling pathways as well as target genes and intervening at the right time points to prevent more severe immune inflammatory responses and promote better and faster recovery of the patient.
Collapse
Affiliation(s)
- Yang Li
- Institute for Clinical and Experimental Trauma Immunology (ITI), University Hospital Ulm, Helmholtzstr. 8/1, 89081, Ulm, Germany
| | - Annette Palmer
- Institute for Clinical and Experimental Trauma Immunology (ITI), University Hospital Ulm, Helmholtzstr. 8/1, 89081, Ulm, Germany
| | - Ludmila Lupu
- Institute for Clinical and Experimental Trauma Immunology (ITI), University Hospital Ulm, Helmholtzstr. 8/1, 89081, Ulm, Germany
| | - Markus Huber-Lang
- Institute for Clinical and Experimental Trauma Immunology (ITI), University Hospital Ulm, Helmholtzstr. 8/1, 89081, Ulm, Germany.
| |
Collapse
|
124
|
Selvin T, Fasterius E, Jarvius M, Fryknäs M, Larsson R, Andersson CR. Single-cell transcriptional pharmacodynamics of trifluridine in a tumor-immune model. Sci Rep 2022; 12:11960. [PMID: 35831404 PMCID: PMC9279337 DOI: 10.1038/s41598-022-16077-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 07/04/2022] [Indexed: 11/09/2022] Open
Abstract
Understanding the immunological effects of chemotherapy is of great importance, especially now that we have entered an era where ever-increasing pre-clinical and clinical efforts are put into combining chemotherapy and immunotherapy to combat cancer. Single-cell RNA sequencing (scRNA-seq) has proved to be a powerful technique with a broad range of applications, studies evaluating drug effects in co-cultures of tumor and immune cells are however scarce. We treated a co-culture comprised of human colorectal cancer (CRC) cells and peripheral blood mononuclear cells (PBMCs) with the nucleoside analogue trifluridine (FTD) and used scRNA-seq to analyze posttreatment gene expression profiles in thousands of individual cancer and immune cells concurrently. ScRNA-seq recapitulated major mechanisms of action previously described for FTD and provided new insight into possible treatment-induced effects on T-cell mediated antitumor responses.
Collapse
Affiliation(s)
- Tove Selvin
- Department of Medical Sciences, Uppsala University, 75185, Uppsala, Sweden.
| | - Erik Fasterius
- National Bioinformatics Infrastructure Sweden (NBIS), Stockholm University, Stockholm, Sweden
| | - Malin Jarvius
- Department of Medical Sciences, Uppsala University, 75185, Uppsala, Sweden.,Department of Pharmaceutical Biosciences and Science for Life Laboratory, Uppsala University, Box 591, 751 24, Uppsala, Sweden
| | - Mårten Fryknäs
- Department of Medical Sciences, Uppsala University, 75185, Uppsala, Sweden
| | - Rolf Larsson
- Department of Medical Sciences, Uppsala University, 75185, Uppsala, Sweden
| | - Claes R Andersson
- Department of Medical Sciences, Uppsala University, 75185, Uppsala, Sweden.
| |
Collapse
|
125
|
Ganguly K, Kishore U, Metkari SM, Madan T. Immunomodulatory Role of Surfactant Protein-D in a Transgenic Adenocarcinoma of Mouse Prostate (TRAMP) Model. Front Immunol 2022; 13:930449. [PMID: 35874783 PMCID: PMC9302643 DOI: 10.3389/fimmu.2022.930449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 05/23/2022] [Indexed: 11/16/2022] Open
Abstract
Surfactant protein D (SP-D), a pattern recognition molecule, is emerging as a potent anti-tumoural innate immune defense molecule in a range of cancers. Previously, SP-D expression was found to be significantly downregulated at the malignant sites of human prostate adenocarcinoma and associated with an increasing Gleason score and severity. We recently reported selective induction of intrinsic apoptosis by a recombinant fragment of human SP-D (rfhSP-D) in the human Prostate cancer (PCa) biopsy explants and cells with glucose regulated protein of 78 (GRP78) as one of the key interacting partners. The present study evaluated the expression of SP-D in early and advanced stages of PCa using transgenic adenocarcinoma of mouse prostate (TRAMP) model. Both early and late stages of PCa showed significantly decreased SP-D mRNA expression and increased proteolytic degradation of SP-D protein. Systemic and tumoural immunophenotyping of TRAMP model revealed increased serine proteases producing granulocytes and polymorphonuclear myeloid-derived suppressor cells (PMN MDSCs) in the late stage; the serine proteases secreted by these cells could be involved in the degradation of SP-D. Susceptibility of rfhSP-D to elastase-mediated proteolysis provided the rationale to use an elastase-inhibitor to sustain intact rfhSP-D in the tumour microenvironment. The study revealed an immunomodulatory potential of rfhSP-D and elastase inhibitor, sivelestat, to induce macrophage polarization towards M1 with downregulation of PMN MDSCs in ex-vivo cultured TRAMP tumours. Furthermore, rfhSP-D induced immunogenic cell death in murine PCa cells and in TRAMP explants. The findings highlight that SP-D plays an anti-tumourigenic role in PCa by inducing immunogenic cell death and immunomodulation while the prostate tumour milieu adversely impacts SP-D by inhibiting its transcription, and enhancing its proteolytic degradation. Transformation of an immunologically "cold tumour" into a "hot tumour" implicates therapeutic potential of rfhSP-D in PCa.
Collapse
Affiliation(s)
- Kasturi Ganguly
- Department of Innate Immunity, Indian Council of Medical Research (ICMR)- National Institute for Research in Reproductive and Child Health, Mumbai, India
| | - Uday Kishore
- Biosciences, College of Health and Life Sciences, Brunel University London, Uxbridge, United Kingdom
- Department of Veterinary Medicine, United Arab Emirates (U.A.E) University, Al Ain, United Arab Emirates
| | - Siddhanath M. Metkari
- Indian Council of Medical Research (ICMR)- National Institute for Research in Reproductive and Child Health, Mumbai, India
| | - Taruna Madan
- Department of Innate Immunity, Indian Council of Medical Research (ICMR)- National Institute for Research in Reproductive and Child Health, Mumbai, India
| |
Collapse
|
126
|
Ertveldt T, De Beck L, De Ridder K, Locy H, de Mey W, Goyvaerts C, Lecocq Q, Ceuppens H, De Vlaeminck Y, Awad RM, Keyaerts M, Devoogdt N, D'Huyvetter M, Breckpot K, Krasniqi A. Targeted Radionuclide Therapy with Low and High-Dose Lutetium-177-Labeled Single Domain Antibodies Induces Distinct Immune Signatures in a Mouse Melanoma Model. Mol Cancer Ther 2022; 21:1136-1148. [PMID: 35499391 PMCID: PMC9377759 DOI: 10.1158/1535-7163.mct-21-0791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2021] [Revised: 02/16/2022] [Accepted: 04/22/2022] [Indexed: 01/07/2023]
Abstract
Targeted radionuclide therapy (TRT) using probes labeled with Lutetium-177 (177Lu) represents a new and growing type of cancer therapy. We studied immunologic changes in response to TRT with 177Lu labeled anti-human CD20 camelid single domain antibodies (sdAb) in a B16-melanoma model transfected to express human CD20, the target antigen, and ovalbumin, a surrogate tumor antigen. High-dose TRT induced melanoma cell death, calreticulin exposure, and ATP-release in vitro. Melanoma-bearing mice received fractionated low and high-dose TRT via tumor targeting anti-human CD20 sdAbs, as opposed to control sdAbs. Tumor growth was delayed with both doses. Low- and high-dose TRT increased IL10 serum levels. Low-dose TRT also decreased CCL5 serum levels. At the tumor, high-dose TRT induced a type I IFN gene signature, while low-dose TRT induced a proinflammatory gene signature. Low- and high-dose TRT increased the percentage of PD-L1pos and PD-L2pos myeloid cells in tumors with a marked increase in alternatively activated macrophages after high-dose TRT. The percentage of tumor-infiltrating T cells was not changed, yet a modest increase in ovalbumin-specific CD8pos T-cells was observed after low-dose TRT. Contradictory, low and high-dose TRT decreased CD4pos Th1 cells in addition to double negative T cells. In conclusion, these data suggest that low and high-dose TRT induce distinct immunologic changes, which might serve as an anchoring point for combination therapy.
Collapse
Affiliation(s)
- Thomas Ertveldt
- Laboratory for Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel, Brussels, Belgium.,Corresponding Authors: Karine Breckpot, Laboratory for Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel, Laarbeeklaan 103, Brussels 1090, Belgium. Phone: 322-477-4566; Fax: 322-477-4506; E-mail: ; and Thomas Ertveldt, E-mail:
| | - Lien De Beck
- Laboratory for Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Kirsten De Ridder
- Laboratory for Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Hanne Locy
- Laboratory for Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Wout de Mey
- Laboratory for Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Cleo Goyvaerts
- Laboratory for Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Quentin Lecocq
- Laboratory for Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Hannelore Ceuppens
- Laboratory for Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Yannick De Vlaeminck
- Laboratory for Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Robin Maximilian Awad
- Laboratory for Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Marleen Keyaerts
- Laboratory for In Vivo Cellular and Molecular Imaging, Department of Medical Imaging, Vrije Universiteit Brussel, Brussels, Belgium.,Department of Nuclear Medicine, UZ Brussel, Vrije Universiteit Brussel, Brussels, Belgium
| | - Nick Devoogdt
- Laboratory for In Vivo Cellular and Molecular Imaging, Department of Medical Imaging, Vrije Universiteit Brussel, Brussels, Belgium
| | - Matthias D'Huyvetter
- Laboratory for In Vivo Cellular and Molecular Imaging, Department of Medical Imaging, Vrije Universiteit Brussel, Brussels, Belgium
| | - Karine Breckpot
- Laboratory for Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel, Brussels, Belgium.,Corresponding Authors: Karine Breckpot, Laboratory for Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel, Laarbeeklaan 103, Brussels 1090, Belgium. Phone: 322-477-4566; Fax: 322-477-4506; E-mail: ; and Thomas Ertveldt, E-mail:
| | - Ahmet Krasniqi
- Laboratory for In Vivo Cellular and Molecular Imaging, Department of Medical Imaging, Vrije Universiteit Brussel, Brussels, Belgium
| |
Collapse
|
127
|
Catanzaro E, Feron O, Skirtach AG, Krysko DV. Immunogenic Cell Death and Role of Nanomaterials Serving as Therapeutic Vaccine for Personalized Cancer Immunotherapy. Front Immunol 2022; 13:925290. [PMID: 35844506 PMCID: PMC9280641 DOI: 10.3389/fimmu.2022.925290] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 06/02/2022] [Indexed: 07/20/2023] Open
Abstract
Immunogenic cell death (ICD) is a rapidly growing research area representing one of the emerging therapeutic strategies of cancer immunotherapy. ICD is an umbrella term covering several cell death modalities including apoptosis, necroptosis, ferroptosis and pyroptosis, and is the product of a balanced combination of adjuvanticity (damage-associated molecular patterns and chemokines/cytokines) and antigenicity (tumor associated antigens). Only a limited number of anti-cancer therapies are available to induce ICD in experimental cancer therapies and even much less is available for clinical use. To overcome this limitation, nanomaterials can be used to increase the immunogenicity of cancer cells killed by anti-cancer therapy, which in themselves are not necessarily immunogenic. In this review, we outline the current state of knowledge of ICD modalities and discuss achievements in using nanomaterials to increase the immunogenicity of dying cancer cells. The emerging trends in modulating the immunogenicity of dying cancer cells in experimental and translational cancer therapies and the challenges facing them are described. In conclusion, nanomaterials are expected to drive further progress in their use to increase efficacy of anti-cancer therapy based on ICD induction and in the future, it is necessary to validate these strategies in clinical settings, which will be a challenging research area.
Collapse
Affiliation(s)
- Elena Catanzaro
- Cell Death Investigation and Therapy (CDIT) Laboratory, Department of Human Structure and Repair, Ghent University, Ghent, Belgium
- Cancer Research Institute Ghent, Ghent, Belgium
| | - Olivier Feron
- Cancer Translational Research Laboratory, Pole of Pharmacology and Therapeutics, Institut de Recherche Expérimentale et Clinique (IREC), UCLouvain, Brussels, Belgium
| | - André G. Skirtach
- Cancer Research Institute Ghent, Ghent, Belgium
- Nano-BioTechnology Laboratory, Department of Biotechnology, Ghent University, Ghent, Belgium
| | - Dmitri V. Krysko
- Cell Death Investigation and Therapy (CDIT) Laboratory, Department of Human Structure and Repair, Ghent University, Ghent, Belgium
- Cancer Research Institute Ghent, Ghent, Belgium
- Institute of Biology and Biomedicine, National Research Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, Russia
- Department of Pathophysiology, Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| |
Collapse
|
128
|
Alghamri MS, Banerjee K, Mujeeb AA, Mauser A, Taher A, Thalla R, McClellan BL, Varela ML, Stamatovic SM, Martinez-Revollar G, Andjelkovic AV, Gregory JV, Kadiyala P, Calinescu A, Jiménez JA, Apfelbaum AA, Lawlor ER, Carney S, Comba A, Faisal SM, Barissi M, Edwards MB, Appelman H, Sun Y, Gan J, Ackermann R, Schwendeman A, Candolfi M, Olin MR, Lahann J, Lowenstein PR, Castro MG. Systemic Delivery of an Adjuvant CXCR4-CXCL12 Signaling Inhibitor Encapsulated in Synthetic Protein Nanoparticles for Glioma Immunotherapy. ACS NANO 2022; 16:8729-8750. [PMID: 35616289 PMCID: PMC9649873 DOI: 10.1021/acsnano.1c07492] [Citation(s) in RCA: 72] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
Glioblastoma (GBM) is an aggressive primary brain cancer, with a 5 year survival of ∼5%. Challenges that hamper GBM therapeutic efficacy include (i) tumor heterogeneity, (ii) treatment resistance, (iii) immunosuppressive tumor microenvironment (TME), and (iv) the blood-brain barrier (BBB). The C-X-C motif chemokine ligand-12/C-X-C motif chemokine receptor-4 (CXCL12/CXCR4) signaling pathway is activated in GBM and is associated with tumor progression. Although the CXCR4 antagonist (AMD3100) has been proposed as an attractive anti-GBM therapeutic target, it has poor pharmacokinetic properties, and unfavorable bioavailability has hampered its clinical implementation. Thus, we developed synthetic protein nanoparticles (SPNPs) coated with the transcytotic peptide iRGD (AMD3100-SPNPs) to target the CXCL2/CXCR4 pathway in GBM via systemic delivery. We showed that AMD3100-SPNPs block CXCL12/CXCR4 signaling in three mouse and human GBM cell cultures in vitro and in a GBM mouse model in vivo. This results in (i) inhibition of GBM proliferation, (ii) reduced infiltration of CXCR4+ monocytic myeloid-derived suppressor cells (M-MDSCs) into the TME, (iii) restoration of BBB integrity, and (iv) induction of immunogenic cell death (ICD), sensitizing the tumor to radiotherapy and leading to anti-GBM immunity. Additionally, we showed that combining AMD3100-SPNPs with radiation led to long-term survival, with ∼60% of GBM tumor-bearing mice remaining tumor free after rechallenging with a second GBM in the contralateral hemisphere. This was due to a sustained anti-GBM immunological memory response that prevented tumor recurrence without additional treatment. In view of the potent ICD induction and reprogrammed tumor microenvironment, this SPNP-mediated strategy has a significant clinical translation applicability.
Collapse
Affiliation(s)
- Mahmoud S Alghamri
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Kaushik Banerjee
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Anzar A Mujeeb
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Ava Mauser
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Ayman Taher
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Rohit Thalla
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Brandon L McClellan
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Maria L Varela
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Svetlana M Stamatovic
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
| | | | - Anuska V Andjelkovic
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
| | - Jason V Gregory
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Padma Kadiyala
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Alexandra Calinescu
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Jennifer A Jiménez
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI 48109, USA
| | - April A Apfelbaum
- Seattle Children’s Research Institute, University of Washington Seattle, WA, 98101
- Cancer Biology Ph.D. Program, University of Michigan, Ann Arbor, MI 48109, USA
| | - Elizabeth R Lawlor
- Seattle Children’s Research Institute, University of Washington Seattle, WA, 98101
| | - Stephen Carney
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Andrea Comba
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Syed Mohd Faisal
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Marcus Barissi
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Marta B. Edwards
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Henry Appelman
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
| | - Yilun Sun
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, OH 44106
| | - Jingyao Gan
- Department of Pharmaceutical Sciences, University of Michigan College of Pharmacy, Ann Arbor, MI 48109, USA
| | - Rose Ackermann
- Department of Pharmaceutical Sciences, University of Michigan College of Pharmacy, Ann Arbor, MI 48109, USA
| | - Anna Schwendeman
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA
- Department of Pharmaceutical Sciences, University of Michigan College of Pharmacy, Ann Arbor, MI 48109, USA
| | - Marianela Candolfi
- Instituto de Investigaciones Biomédicas (INBIOMED, UBA-CONICET), Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Michael R. Olin
- Department of Pediatrics, University of Minnesota, Minneapolis MN 55455
- Masonic Cancer Center, University of Minnesota, Minneapolis MN 55455
| | - Joerg Lahann
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
- Corresponding Authors:, ,
| | - Pedro R. Lowenstein
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Biosciences Initiative in Brain Cancer, University of Michigan, Ann Arbor, MI 48109, USA
- Corresponding Authors:, ,
| | - Maria G. Castro
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Biosciences Initiative in Brain Cancer, University of Michigan, Ann Arbor, MI 48109, USA
- Corresponding Authors:, ,
| |
Collapse
|
129
|
Miri SM, Pourhossein B, Hosseini SY, Keshavarz M, Shahmahmoodi S, Zolfaghari MR, Mohebbi SR, Gorji A, Ghaemi A. Enhanced synergistic antitumor effect of a DNA vaccine with anticancer cytokine, MDA-7/IL-24, and immune checkpoint blockade. Virol J 2022; 19:106. [PMID: 35752792 PMCID: PMC9233788 DOI: 10.1186/s12985-022-01842-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Accepted: 06/06/2022] [Indexed: 11/22/2022] Open
Abstract
Background MDA-7/IL-24 cytokine has shown potent antitumor properties in various types of cancer without exerting any significant toxicity on healthy cells. It has also been proved to encompass pro-immune Th1 cytokine-like behavior. Several E7 DNA vaccines have developed against human papillomavirus (HPV)-related cervical cancer. However, the restricted immunogenicity has limited their clinical applications individually. To address this deficiency, we investigated whether combining the E7 DNA vaccine with MDA-7/IL-24 as an adjuvant would elicit efficient antitumor responses in tumor-bearing mouse models. Next, we evaluated how suppression of immunosuppressive IL-10 cytokine would enhance the outcome of our candidate adjuvant vaccine.
Methods For this purpose, tumor-bearing mice received either E7 DNA vaccine, MDA-7/IL-24 cytokine or combination of E7 vaccine with MDA-7/IL-24 adjuvant one week after tumor challenge and boosted two times with one-week interval. IL-10 blockade was performed by injection of anti-IL-10 mAb before each immunization. One week after the last immunization, mice were sacrificed and the treatment efficacy was evaluated through immunological and immunohistochemical analysis. Moreover, the condition of tumors was monitored every two days for six weeks intervals from week 2 on, and the tumor volume was measured and compared within different groups. Results A highly significant synergistic relationship was observed between the E7 DNA vaccine and the MDA-7/IL-24 cytokine against HPV-16+ cervical cancer models. An increase in proliferation of lymphocytes, cytotoxicity of CD8+ T cells, the level of Th1 cytokines (IFN-γ, TNF-α) and IL-4, the level of apoptotic markers (TRAIL and caspase-9), and a decrease in the level of immunosuppressive IL-10 cytokine, together with the control of tumor growth and the induction of tumor regression, all prove the efficacy of adjuvant E7&IL-24 vaccine when compared to their individual administration. Surprisingly, vaccination with the DNA E7&IL-24 significantly reduced the population of Regulatory T cells (Treg) in the spleen of immunized mice compared to sole administration and control groups. Moreover, IL-10 blockade enhanced the effect of the co-administration by eliciting higher levels of IFN-γ and caspase-9, reducing Il-10 secretion and provoking the regression of tumor size. Conclusion The synergy between the E7 DNA vaccine and MDA-7/IL-24 suggests that DNA vaccines’ low immunogenicity can be effectively addressed by coupling them with an immunoregulatory agent. Moreover, IL-10 blockade can be considered a complementary treatment to improve the outcome of conventional or novel cancer therapies. Supplementary Information The online version contains supplementary material available at 10.1186/s12985-022-01842-x.
Collapse
Affiliation(s)
- Seyed Mohammad Miri
- Department of Influenza and Other Respiratory Viruses, Pasteur Institute of Iran, P.O. Box: 1316943551, Tehran, Iran
| | - Behzad Pourhossein
- Department of Medical Virology, Hamedan University of Medical Sciences, Hamedan, Iran.,Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Seyed Younes Hosseini
- Department of Bacteriology and Virology, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohsen Keshavarz
- The Persian Gulf Tropical Medicine Research Center, The Persian Gulf Biomedical Sciences Research Institute, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Shohreh Shahmahmoodi
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Seyed Reza Mohebbi
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ali Gorji
- Department of Neurosurgery and Department of Neurology, Westfälische Wilhelms-Universität, Münster, Germany.,Shefa Neuroscience Research Center, Khatam Alanbia Hospital, Tehran, Iran
| | - Amir Ghaemi
- Department of Influenza and Other Respiratory Viruses, Pasteur Institute of Iran, P.O. Box: 1316943551, Tehran, Iran.
| |
Collapse
|
130
|
Liu W, Chen H, Zhu Z, Liu Z, Ma C, Lee YJ, Bartlett DL, Guo ZS. Ferroptosis Inducer Improves the Efficacy of Oncolytic Virus-Mediated Cancer Immunotherapy. Biomedicines 2022; 10:1425. [PMID: 35740445 PMCID: PMC9219720 DOI: 10.3390/biomedicines10061425] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 06/10/2022] [Accepted: 06/13/2022] [Indexed: 12/15/2022] Open
Abstract
Ferroptosis is a type of programmed cell death dependent on iron and characterized by the accumulation of lipid peroxides. In this study, we explore the combination of a ferroptosis activator with an oncolytic vaccinia virus in tumor models. Erastin induced cell death in hepatoma, colon, and ovarian cancer cells, but not in melanoma cancer cells. Erastin, not the oncolytic vaccinia virus (OVV), induced the expression of key marker genes for ferroptosis in cancer cells. In hepatocellular carcinoma and colon cancer models, either erastin or OVV inhibited tumor growth, but a combination of the two yielded the best therapeutic effects, as indicated by inhibited tumor growth or regression and longer host survival. Immunological analyses indicate that erastin alone had little or no effect on systemic immunity or local immunity in the tumor. However, when combined with OV, erastin enhanced the number of activated dendritic cells and the activity of tumor-infiltrating T lymphocytes as indicated by an increase in IFN-γ+CD8+ and PD-1+CD8+ T cells. These results demonstrate that erastin can exert cytotoxicity on cancer cells via ferroptosis, but has little effect on immune activity by itself. However, when combined with an OVV, erastin promoted antitumoral immunity and efficacy by increasing the number of activated dendritic cells and promoting the activities of tumor specific CD8+ T cells in the tumor.
Collapse
Affiliation(s)
- Weilin Liu
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; (W.L.); (H.C.); (Z.Z.); (Z.L.); (C.M.); (Y.J.L.)
- UPMC Hillman Cancer Center, Pittsburgh, PA 15232, USA
- Xiangya Medical College, Central South University, Changsha 410013, China
| | - Hongqi Chen
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; (W.L.); (H.C.); (Z.Z.); (Z.L.); (C.M.); (Y.J.L.)
- UPMC Hillman Cancer Center, Pittsburgh, PA 15232, USA
| | - Zhi Zhu
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; (W.L.); (H.C.); (Z.Z.); (Z.L.); (C.M.); (Y.J.L.)
- UPMC Hillman Cancer Center, Pittsburgh, PA 15232, USA
| | - Zuqiang Liu
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; (W.L.); (H.C.); (Z.Z.); (Z.L.); (C.M.); (Y.J.L.)
- UPMC Hillman Cancer Center, Pittsburgh, PA 15232, USA
- AHN-Cancer Institute, Pittsburgh, PA 15212, USA
| | - Congrong Ma
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; (W.L.); (H.C.); (Z.Z.); (Z.L.); (C.M.); (Y.J.L.)
- UPMC Hillman Cancer Center, Pittsburgh, PA 15232, USA
| | - Yong J. Lee
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; (W.L.); (H.C.); (Z.Z.); (Z.L.); (C.M.); (Y.J.L.)
- UPMC Hillman Cancer Center, Pittsburgh, PA 15232, USA
| | - David L. Bartlett
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; (W.L.); (H.C.); (Z.Z.); (Z.L.); (C.M.); (Y.J.L.)
- UPMC Hillman Cancer Center, Pittsburgh, PA 15232, USA
- AHN-Cancer Institute, Pittsburgh, PA 15212, USA
| | - Zong-Sheng Guo
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; (W.L.); (H.C.); (Z.Z.); (Z.L.); (C.M.); (Y.J.L.)
- UPMC Hillman Cancer Center, Pittsburgh, PA 15232, USA
- Department of Immunology, Roswell Park Cancer Institute, Buffalo, NY 14203, USA
| |
Collapse
|
131
|
Xiong X, Huang KB, Wang Y, Cao B, Luo Y, Chen H, Yang Y, Long Y, Liu M, Chan ASC, Liang H, Zou T. Target Profiling of an Iridium(III)-Based Immunogenic Cell Death Inducer Unveils the Engagement of Unfolded Protein Response Regulator BiP. J Am Chem Soc 2022; 144:10407-10416. [PMID: 35658433 DOI: 10.1021/jacs.2c02435] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Clinical chemotherapeutic drugs have occasionally been observed to induce antitumor immune responses beyond the direct cytotoxicity. Such effects are coined as immunogenic cell death (ICD), representing a "second hit" from the host immune system to tumor cells. Although chemo-immunotherapy is highly promising, ICD inducers remain sparse with vague drug-target mechanisms. Here, we report an endoplasmic reticulum stress-inducing cyclometalated Ir(III)-bisNHC complex (1a) as a new ICD inducer, and based on this compound, a clickable photoaffinity probe was designed for target identification, which unveiled the engagement of the master regulator protein BiP (binding immunoglobulin protein)/GRP78 of the unfolded protein response pathway. This has been confirmed by a series of cellular and biochemical studies including fluorescence microscopy, cellular thermal shift assay, enzymatic assays, and so forth, showing the capability of 1a for BiP destabilization. Notably, besides 1a, the previously reported ICD inducers including KP1339, mitoxantrone, and oxaliplatin were also found to engage BiP interaction, suggesting the important role of BiP in eliciting anticancer immunity. We believe that the ICD-related target information in this work will help to understand the mode of action of ICD that is beneficial to designing new ICD agents with high specificity and improved efficacy.
Collapse
Affiliation(s)
- Xiaolin Xiong
- Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, P. R. China
| | - Ke-Bin Huang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry & Pharmacy, Guangxi Normal University, Guilin, Guangxi 541004, P. R. China
| | - Yuan Wang
- Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, P. R. China
| | - Bei Cao
- Warshel Institute for Computational Biology, and General Education Division, The Chinese University of Hong Kong, Shenzhen 518172, P. R. China
| | - Yunli Luo
- Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, P. R. China
| | - Huowen Chen
- Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, P. R. China
| | - Yan Yang
- Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, P. R. China
| | - Yan Long
- Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, P. R. China
| | - Moyi Liu
- Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, P. R. China
| | - Albert S C Chan
- Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, P. R. China
| | - Hong Liang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry & Pharmacy, Guangxi Normal University, Guilin, Guangxi 541004, P. R. China
| | - Taotao Zou
- Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, P. R. China
| |
Collapse
|
132
|
Tian H, Lin J, Zhu F, Li J, Jiang S, Xie L, Li Y, Wang P, Hou Z, Mi J. 2D graphene oxide-L-arginine-soybean lecithin nanogenerator for synergistic photothermal and NO gas therapy. CHINESE CHEM LETT 2022. [DOI: 10.1016/j.cclet.2022.05.091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
133
|
The Achilles' heel of cancer: targeting tumors via lysosome-induced immunogenic cell death. Cell Death Dis 2022; 13:509. [PMID: 35637197 PMCID: PMC9151667 DOI: 10.1038/s41419-022-04912-8] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Revised: 03/10/2022] [Accepted: 05/04/2022] [Indexed: 12/14/2022]
Abstract
Interest in the lysosome's potential role in anticancer therapies has recently been appreciated in the field of immuno-oncology. Targeting lysosomes triggers apoptotic pathways, inhibits cytoprotective autophagy, and activates a unique form of apoptosis known as immunogenic cell death (ICD). This mechanism stimulates a local and systemic immune response against dead-cell antigens. Stressors that can lead to ICD include an abundance of ROS which induce lysosome membrane permeability (LMP). Dying cells express markers that activate immune cells. Dendritic cells engulf the dying cell and then present the cell's neoantigens to T cells. The discovery of ICD-inducing agents is important due to their potential to trigger autoimmunity. In this review, we discuss the various mechanisms of activating lysosome-induced cell death in cancer cells specifically and the strategies that current laboratories are using to selectively promote LMP in tumors.
Collapse
|
134
|
Min T, Xie X, Ren K, Sun T, Wang H, Dang C, Zhang H. Therapeutic Effects of Cold Atmospheric Plasma on Solid Tumor. Front Med (Lausanne) 2022; 9:884887. [PMID: 35646968 PMCID: PMC9139675 DOI: 10.3389/fmed.2022.884887] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Accepted: 04/13/2022] [Indexed: 12/14/2022] Open
Abstract
Cancer is a devastating disease, and there is no particularly effective treatment at present. Recently, a new treatment, cold atmospheric plasma (CAP), has been proposed. At present, CAP is confirmed to have selective killing effect on tumor by many studies in vitro and in vivo. A targeted literature search was carried out on the study of cold atmospheric plasma. Through analysis and screening, a narrative review approach was selected to describe therapeutic effects of cold atmospheric plasma on solid tumor. According to the recent studies on plasma, some hypothetical therapeutic schemes of CAP are proposed in this paper. The killing mechanism of CAP on solid tumor is expounded in terms of the selectivity of CAP to tumor, the effects of CAP on cells, tumor microenvironment (TME) and immune system. CAP has many effects on solid tumors, and these effects are dose-dependent. The effects of optimal doses of CAP on solid tumors include killing tumor cells, inhibiting non-malignant cells and ECM in TME, affecting the communication between tumor cells, and inducing immunogenic death of tumor cells. In addition, several promising research directions of CAP are proposed in this review, which provide guidance for future research.
Collapse
Affiliation(s)
- Tianhao Min
- Department of Surgical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xin Xie
- Department of Nuclear Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Kaijie Ren
- Department of Surgical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Tuanhe Sun
- Department of Surgical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Haonan Wang
- Department of Surgical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Chengxue Dang
- Department of Surgical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- *Correspondence: Chengxue Dang
| | - Hao Zhang
- Department of Surgical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Hao Zhang
| |
Collapse
|
135
|
Metformin combined with local irradiation provokes abscopal effects in a murine rectal cancer model. Sci Rep 2022; 12:7290. [PMID: 35508498 PMCID: PMC9068771 DOI: 10.1038/s41598-022-11236-2] [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: 12/17/2021] [Accepted: 04/18/2022] [Indexed: 11/20/2022] Open
Abstract
Although preoperative chemoradiation therapy can down-stage locally advanced rectal cancer (LARC), it has little effect on distant metastases. Metformin exerts an anti-cancer effect partly through the activation of host immunity. LuM1, a highly lung metastatic subclone of colon 26, was injected subcutaneously (sc) in BALB/c mice and treated with metformin and/or local radiation (RT). Lung metastases and the primary tumors were evaluated and the phenotypes of immune cells in the spleen and lung metastases were examined with flow cytometry and immunohistochemistry. Local RT, but not metformin, partially delayed the growth of sc tumor which was augmented with metformin. Lung metastases were unchanged in metformin or RT alone, but significantly reduced in the combined therapy. The ratios of splenic T cells tended to be low in the RT group, which were increased by the addition of metformin. IFN-γ production of the splenic CD4(+) and CD8(+) T cells was enhanced and CD49b (+) CD335(+) activated NK cells was increased after combined treatment group. Density of NK cells infiltrating in lung metastases was increased after combination treatment. Metformin effectively enhances local and abscopal effects of RT though the activation of cell-mediated immunity and might be clinically useful for LARC.
Collapse
|
136
|
Kawanabe-Matsuda H, Takeda K, Nakamura M, Makino S, Karasaki T, Kakimi K, Nishimukai M, Ohno T, Omi J, Kano K, Uwamizu A, Yagita H, Boneca IG, Eberl G, Aoki J, Smyth MJ, Okumura K. Dietary Lactobacillus-Derived Exopolysaccharide Enhances Immune-Checkpoint Blockade Therapy. Cancer Discov 2022; 12:1336-1355. [PMID: 35180303 PMCID: PMC9662940 DOI: 10.1158/2159-8290.cd-21-0929] [Citation(s) in RCA: 70] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2021] [Revised: 12/20/2021] [Accepted: 02/15/2022] [Indexed: 01/07/2023]
Abstract
Microbes and their byproducts have been reported to regulate host health and immune functions. Here we demonstrated that microbial exopolysaccharide produced by Lactobacillus delbrueckii subsp. bulgaricus OLL1073R-1 (EPS-R1) induced CCR6+ CD8+ T cells of mice and humans. In mice, ingestion of EPS-R1 augmented antitumor effects of anti-CTLA-4 or anti-PD-1 monoclonal antibody against CCL20-expressing tumors, in which infiltrating CCR6+ CD8+ T cells were increased and produced IFNγ accompanied by a substantial immune response gene expression signature maintaining T-cell functions. Of note, the antitumor adjuvant effect of EPS-R1 was also observed in germ-free mice. Furthermore, the induction of CCR6 expression was mediated through the phosphorylated structure in EPS-R1 and a lysophosphatidic acid receptor on CD8+ T cells. Overall, we find that dietary EPS-R1 consumption induces CCR6+ CD8+ T cells in Peyer's patches, favoring a tumor microenvironment that augments the therapeutic effect of immune-checkpoint blockade depending on CCL20 production by tumors. SIGNIFICANCE Gut microbiota- and probiotic-derived metabolites are attractive agents to augment the efficacy of immunotherapies. Here we demonstrated that dietary consumption of Lactobacillus-derived exopolysaccharide induced CCR6+ CD8+ T cells in Peyer's patches and improved the tumor microenvironment to augment the therapeutic effects of immune-checkpoint blockade against CCL20-producing tumors. See related commentary by Di Luccia and Colonna, p. 1189. This article is highlighted in the In This Issue feature, p. 1171.
Collapse
Affiliation(s)
- Hirotaka Kawanabe-Matsuda
- Department of Biofunctional Microbiota, Graduate School of Medicine, Juntendo University, Tokyo, Japan.,Research Team, Co-Creation Center, Meiji Holdings Co., Ltd., Hachioji, Japan
| | - Kazuyoshi Takeda
- Department of Biofunctional Microbiota, Graduate School of Medicine, Juntendo University, Tokyo, Japan.,Laboratory of Cell Biology, Research Support Center, Graduate School of Medicine, Juntendo University, Tokyo, Japan.,Corresponding Author: Kazuyoshi Takeda, Laboratory of Cell Biology, Research Support Center, Graduate School of Medicine, Juntendo University, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan. Phone: 81-3-5802-1591; E-mail:
| | - Marie Nakamura
- Research Team, Co-Creation Center, Meiji Holdings Co., Ltd., Hachioji, Japan
| | - Seiya Makino
- Department of Biofunctional Microbiota, Graduate School of Medicine, Juntendo University, Tokyo, Japan.,Research Team, Co-Creation Center, Meiji Holdings Co., Ltd., Hachioji, Japan
| | - Takahiro Karasaki
- Department of Immunotherapeutics, The University of Tokyo Hospital, Tokyo, Japan.,Department of Thoracic Surgery, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Kazuhiro Kakimi
- Department of Immunotherapeutics, The University of Tokyo Hospital, Tokyo, Japan
| | - Megumi Nishimukai
- Department of Animal Science, Faculty of Agriculture, Iwate University, Morioka, Japan
| | - Tatsukuni Ohno
- Department of Biofunctional Microbiota, Graduate School of Medicine, Juntendo University, Tokyo, Japan.,Oral Health Science Center, Tokyo Dental College, Tokyo, Japan.,Tokyo Dental College Research Branding Project, Tokyo Dental College, Tokyo, Japan
| | - Jumpei Omi
- Department of Health Chemistry, Graduate School of Pharmaceutical Science, The University of Tokyo, Tokyo, Japan.,Department of Molecular and Cellular Biochemistry, Graduate School of Pharmaceutical Science, Tohoku University, Sendai, Japan.,AMED-LEAP, Japan Science and Technology Corporation, Kawaguchi, Japan
| | - Kuniyuki Kano
- Department of Health Chemistry, Graduate School of Pharmaceutical Science, The University of Tokyo, Tokyo, Japan.,Department of Molecular and Cellular Biochemistry, Graduate School of Pharmaceutical Science, Tohoku University, Sendai, Japan.,AMED-LEAP, Japan Science and Technology Corporation, Kawaguchi, Japan
| | - Akiharu Uwamizu
- Department of Health Chemistry, Graduate School of Pharmaceutical Science, The University of Tokyo, Tokyo, Japan.,Department of Molecular and Cellular Biochemistry, Graduate School of Pharmaceutical Science, Tohoku University, Sendai, Japan.,AMED-LEAP, Japan Science and Technology Corporation, Kawaguchi, Japan
| | - Hideo Yagita
- Department of Immunology, School of Medicine, Juntendo University, Tokyo, Japan
| | - Ivo Gomperts Boneca
- Institut Pasteur, Unit of Biology and Genetics of Bacterial Cell Wall, Paris, France. INSERM, Équipe Avenir, Paris, France
| | - Gérard Eberl
- Microenvironment and Immunity Unit, Institut Pasteur, Paris, France
| | - Junken Aoki
- Department of Health Chemistry, Graduate School of Pharmaceutical Science, The University of Tokyo, Tokyo, Japan.,Department of Molecular and Cellular Biochemistry, Graduate School of Pharmaceutical Science, Tohoku University, Sendai, Japan.,AMED-LEAP, Japan Science and Technology Corporation, Kawaguchi, Japan
| | - Mark J. Smyth
- Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
| | - Ko Okumura
- Department of Biofunctional Microbiota, Graduate School of Medicine, Juntendo University, Tokyo, Japan.,Atopy (Allergy) Research Center, Graduate School of Medicine, Juntendo University, Tokyo, Japan
| |
Collapse
|
137
|
Combining antibody-drug conjugates with immunotherapy in solid tumors: current landscape and future perspectives. Cancer Treat Rev 2022; 106:102395. [DOI: 10.1016/j.ctrv.2022.102395] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 04/12/2022] [Accepted: 04/13/2022] [Indexed: 12/11/2022]
|
138
|
Xu J, Tao P, Lü D, Jiang Y, Xia Q. Role of high-mobility group box 1 in cancer. ZHONG NAN DA XUE XUE BAO. YI XUE BAN = JOURNAL OF CENTRAL SOUTH UNIVERSITY. MEDICAL SCIENCES 2022; 47:505-511. [PMID: 35545346 PMCID: PMC10930161 DOI: 10.11817/j.issn.1672-7347.2022.210679] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Indexed: 06/15/2023]
Abstract
High-mobility group box 1 (HMGB1) is a non-histone nuclear protein in most eukaryocytes. Inside the nucleus, HMGB1 plays an important role in several DNA events such as DNA repair, transcription, telomere maintenance, and genome stability. While outside the nucleus, it fulfils more complicated functions, including promoting cell proliferation, inflammation, angiogenesis, immune tolerance and immune escape, which may play a pro-tumoral role.Meanwhile, HMGB1 acts as an anti-tumoral protein by regulating immune cell recruitment and inducing immunogenic cell death (ICD) during the carcinogenesis process. Therefore, abnormal expression of HMGB1 is associated with oncogenesis, development, and metastasis of cancer, which may play a dual role of pro-tumor and anti-tumor.
Collapse
Affiliation(s)
- Juan Xu
- Second Department of Internal Medicine, People's Hospital of Guandu District, Kunming 650200.
| | - Pengzuo Tao
- Department of Clinical Laboratory, Yunan Cancer Hospital/Third Affiliated Hospital of Kunming Medical University, Kunming 650118
| | - Dongjin Lü
- Third Department of Internal Medicine, Yunan Cancer Hospital/Third Affiliated Hospital of Kunming Medical University, Kunming 650118, China
| | - Yu'e Jiang
- Department of Clinical Laboratory, Yunan Cancer Hospital/Third Affiliated Hospital of Kunming Medical University, Kunming 650118
| | - Quansong Xia
- Department of Clinical Laboratory, Yunan Cancer Hospital/Third Affiliated Hospital of Kunming Medical University, Kunming 650118.
| |
Collapse
|
139
|
Luo W, Wang Y, Zhang T. Win or loss? Combination therapy does improve the oncolytic virus therapy to pancreatic cancer. Cancer Cell Int 2022; 22:160. [PMID: 35443724 PMCID: PMC9022249 DOI: 10.1186/s12935-022-02583-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Accepted: 04/11/2022] [Indexed: 11/10/2022] Open
Abstract
Pancreatic cancer (PC) is a growing global burden, remaining one of the most lethal cancers of the gastrointestinal tract. Moreover, PC is resistant to various treatments such as chemotherapy, radiotherapy, and immunotherapy. New therapies are urgently needed to improve the prognosis of PC. Oncolytic virus (OV) therapy is a promising new treatment option. OV is a genetically modified virus that selectively replicates in tumor cells. It can kill tumor cells without harming normal cells. The activation of tumor-specific T-cells is a unique feature of OV-mediated therapy. However, OV-mediated mono-therapeutic efficacy remains controversial, especially for metastatic or advanced patients who require systemically deliverable therapies. Hence, combination therapies will be critical to improve the therapeutic efficacy of OV-mediated therapy and prevent tumor recurrence. This review aims to investigate novel combinatorial treatments with OV therapy and explore the inner mechanism of those combined therapies, hopefully providing a new direction for a better prognosis of PC.
Collapse
Affiliation(s)
- Wenhao Luo
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1 Shuaifuyuan, Wangfujing Street, Beijing, 100730, China
| | - Yawen Wang
- Department of Endocrinology, State Key Laboratory of Complex Severe and Rare Diseases, Key Laboratory of Endocrinology of National Health Commission of the People's Republic of China, The Translational Medicine Center of Peking Union Medical College Hospital (PUMCH), PUMCH, Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), Beijing, 100730, China
| | - Taiping Zhang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1 Shuaifuyuan, Wangfujing Street, Beijing, 100730, China. .,Clinical Immunology Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
| |
Collapse
|
140
|
Record M, Attia M, Carayon K, Pucheu L, Bunay J, Soulès R, Ayadi S, Payré B, Perrin‐Cocon L, Bourgailh F, Lamazière A, Lotteau V, Poirot M, Silvente‐Poirot S, de Medina P. Targeting the liver X receptor with dendrogenin A differentiates tumour cells to secrete immunogenic exosome-enriched vesicles. J Extracell Vesicles 2022; 11:e12211. [PMID: 35411723 PMCID: PMC9001168 DOI: 10.1002/jev2.12211] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 03/07/2022] [Accepted: 03/21/2022] [Indexed: 01/02/2023] Open
Abstract
Tumour cells are characterized by having lost their differentiation state. They constitutively secrete small extracellular vesicles (sEV) called exosomes when they come from late endosomes. Dendrogenin A (DDA) is an endogenous tumour suppressor cholesterol‐derived metabolite. It is a new class of ligand of the nuclear Liver X receptors (LXR) which regulate cholesterol homeostasis and immunity. We hypothesized that DDA, which induces tumour cell differentiation, inhibition of tumour growth and immune cell infiltration into tumours, could functionally modify sEV secreted by tumour cells. Here, we have shown that DDA differentiates tumour cells by acting on the LXRβ. This results in an increased production of sEV (DDA‐sEV) which includes exosomes. The DDA‐sEV secreted from DDA‐treated cells were characterized for their content and activity in comparison to sEV secreted from control cells (C‐sEV). DDA‐sEV were enriched, relatively to C‐sEV, in several proteins and lipids such as differentiation antigens, “eat‐me” signals, lipidated LC3 and the endosomal phospholipid bis(monoacylglycero)phosphate, which stimulates dendritic cell maturation and a Th1 T lymphocyte polarization. Moreover, DDA‐sEV inhibited the growth of tumours implanted into immunocompetent mice compared to control conditions. This study reveals a pharmacological control through a nuclear receptor of exosome‐enriched tumour sEV secretion, composition and immune function. Targeting the LXR may be a novel way to reprogram tumour cells and sEV to stimulate immunity against cancer.
Collapse
Affiliation(s)
- Michel Record
- Team “Cholesterol Metabolism and Therapeutic Innovations” Cancer Research Centre of Toulouse (CRCT)UMR 1037 INSERMUMR 5071 CNRSUniversité de Toulouse IIIEquipe labellisée par la Ligue Nationale Contre le CancerFrench network for Nutrition And Cancer Research (NACRe network)France
| | - Mehdi Attia
- Team “Cholesterol Metabolism and Therapeutic Innovations” Cancer Research Centre of Toulouse (CRCT)UMR 1037 INSERMUMR 5071 CNRSUniversité de Toulouse IIIEquipe labellisée par la Ligue Nationale Contre le CancerFrench network for Nutrition And Cancer Research (NACRe network)France
| | - Kevin Carayon
- Team “Cholesterol Metabolism and Therapeutic Innovations” Cancer Research Centre of Toulouse (CRCT)UMR 1037 INSERMUMR 5071 CNRSUniversité de Toulouse IIIEquipe labellisée par la Ligue Nationale Contre le CancerFrench network for Nutrition And Cancer Research (NACRe network)France
| | - Laly Pucheu
- Team “Cholesterol Metabolism and Therapeutic Innovations” Cancer Research Centre of Toulouse (CRCT)UMR 1037 INSERMUMR 5071 CNRSUniversité de Toulouse IIIEquipe labellisée par la Ligue Nationale Contre le CancerFrench network for Nutrition And Cancer Research (NACRe network)France
| | - Julio Bunay
- Team “Cholesterol Metabolism and Therapeutic Innovations” Cancer Research Centre of Toulouse (CRCT)UMR 1037 INSERMUMR 5071 CNRSUniversité de Toulouse IIIEquipe labellisée par la Ligue Nationale Contre le CancerFrench network for Nutrition And Cancer Research (NACRe network)France
| | - Régis Soulès
- Team “Cholesterol Metabolism and Therapeutic Innovations” Cancer Research Centre of Toulouse (CRCT)UMR 1037 INSERMUMR 5071 CNRSUniversité de Toulouse IIIEquipe labellisée par la Ligue Nationale Contre le CancerFrench network for Nutrition And Cancer Research (NACRe network)France
| | - Silia Ayadi
- Team “Cholesterol Metabolism and Therapeutic Innovations” Cancer Research Centre of Toulouse (CRCT)UMR 1037 INSERMUMR 5071 CNRSUniversité de Toulouse IIIEquipe labellisée par la Ligue Nationale Contre le CancerFrench network for Nutrition And Cancer Research (NACRe network)France
| | - Bruno Payré
- Team “Cholesterol Metabolism and Therapeutic Innovations” Cancer Research Centre of Toulouse (CRCT)UMR 1037 INSERMUMR 5071 CNRSUniversité de Toulouse IIIEquipe labellisée par la Ligue Nationale Contre le CancerFrench network for Nutrition And Cancer Research (NACRe network)France
| | - Laure Perrin‐Cocon
- Team “ VIRal InfectionMetabolism and ImmunityCIRICentre International de Recherche en InfectiologieUniv LyonInsermU1111Université Claude Bernard Lyon 1CNRSUMR5308ENS de LyonLyonFrance
| | - Florence Bourgailh
- Centre de Microscopie Electronique Appliquée à la BiologieFaculté de Médecine RangueilToulouseFrance
| | - Antonin Lamazière
- Sorbonne UniversitéINSERMCentre de Recherche Saint‐AntoineCRSAAP‐HP.SUHôpital Saint AntoineDépartement de métabobolomique cliniqueParisFrance
| | - Vincent Lotteau
- Team “ VIRal InfectionMetabolism and ImmunityCIRICentre International de Recherche en InfectiologieUniv LyonInsermU1111Université Claude Bernard Lyon 1CNRSUMR5308ENS de LyonLyonFrance
| | - Marc Poirot
- Team “Cholesterol Metabolism and Therapeutic Innovations” Cancer Research Centre of Toulouse (CRCT)UMR 1037 INSERMUMR 5071 CNRSUniversité de Toulouse IIIEquipe labellisée par la Ligue Nationale Contre le CancerFrench network for Nutrition And Cancer Research (NACRe network)France
| | - Sandrine Silvente‐Poirot
- Team “Cholesterol Metabolism and Therapeutic Innovations” Cancer Research Centre of Toulouse (CRCT)UMR 1037 INSERMUMR 5071 CNRSUniversité de Toulouse IIIEquipe labellisée par la Ligue Nationale Contre le CancerFrench network for Nutrition And Cancer Research (NACRe network)France
| | - Philippe de Medina
- Team “Cholesterol Metabolism and Therapeutic Innovations” Cancer Research Centre of Toulouse (CRCT)UMR 1037 INSERMUMR 5071 CNRSUniversité de Toulouse IIIEquipe labellisée par la Ligue Nationale Contre le CancerFrench network for Nutrition And Cancer Research (NACRe network)France
| |
Collapse
|
141
|
Zheng J, Sun Y, Long T, Yuan D, Yue S, Zhang N, Yang Z. Sonosensitizer nanoplatform-mediated sonodynamic therapy induced immunogenic cell death and tumor immune microenvironment variation. Drug Deliv 2022; 29:1164-1175. [PMID: 35393920 PMCID: PMC9004507 DOI: 10.1080/10717544.2022.2058653] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Epithelial ovarian cancer (EOC) is one of the most lethal gynecologic malignancies, and effective treatments are still lacking due to drug tolerance and tumor recurrence. In this study, we aimed to investigate the effects of sonodynamic therapy (SDT) on ovarian cancer and its potential mechanism. Folate receptor-targeted and ultrasound-responsive nanoparticles (NPs) were constructed using PLGA-PEG-FA (PLGA: poly (lactic-co-glycolic) acid, polyethylene glycol (PEG), FA: folate), the reactive oxygen species (ROS)-generating sonosensitizer IR780 and the oxygen-carrying material perfluorohexane (PFH), termed IRO@FA NPs. The antitumor effect of NPs triggered by ultrasound (US) was measured by an apoptosis assay in a C57/BL6 mouse model. Immunochemistry and flow cytometry were used to detect the proportion of CD3+ T, CD4+ T, CD8+ T cells and activated dendritic cells (DCs) in spleens and tumor tissues to assess variation in the immune response. Moreover, endoplasmic reticulum (ER) stress and immunogenic cell death (ICD) markers (high mobility group protein box-1, ATP and calreticulin) were detected to identify potential mechanisms. The results showed that IRO@FA NP-mediated SDT promoted ID8 cell apoptosis both in vitro and in vivo. The densities of CD3+ and CD8+ T lymphocytes and inflammatory markers were upregulated in tumor tissues. IRO@FA NP-mediated SDT prompted DC maturation and T lymphocyte infiltration by inducing ID8 cell ICD.
Collapse
Affiliation(s)
- Jing Zheng
- Department of Gynecology and Obstetrics, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yixuan Sun
- Department of Gynecology and Obstetrics, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Tengfei Long
- Department of Gynecology and Obstetrics, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Dong Yuan
- Department of Gynecology and Obstetrics, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Song Yue
- Department of Gynecology and Obstetrics, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Ni Zhang
- Department of Oncology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zhu Yang
- Department of Gynecology and Obstetrics, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
142
|
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: 5.7] [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
|
143
|
Miguel Cejalvo J, Falato C, Villanueva L, Tolosa P, González X, Pascal M, Canes J, Gavilá J, Manso L, Pascual T, Prat A, Salvador F. Oncolytic Viruses: a new immunotherapeutic approach for breast cancer treatment? Cancer Treat Rev 2022; 106:102392. [DOI: 10.1016/j.ctrv.2022.102392] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 04/03/2022] [Accepted: 04/05/2022] [Indexed: 12/22/2022]
|
144
|
Lee W, Kim DK, Synn CB, Lee HK, Park S, Jung DS, Choi Y, Kim JH, Byeon Y, Kim YS, Lee S, Lee S, Joo Y, Lee EJ, Yun MR, Heo SG, Yang W, Jung JE, Kim EK, Park J, Park JD, Lee DJ, Kim HW, Lim SM, Hong MH, Ahn BC, Lee JB, Pyo KH. Incorporation of SKI-G-801, a Novel AXL Inhibitor, With Anti-PD-1 Plus Chemotherapy Improves Anti-Tumor Activity and Survival by Enhancing T Cell Immunity. Front Oncol 2022; 12:821391. [PMID: 35356198 PMCID: PMC8959645 DOI: 10.3389/fonc.2022.821391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 02/02/2022] [Indexed: 11/13/2022] Open
Abstract
A recently developed treatment strategy for lung cancer that combines immune checkpoint inhibitors with chemotherapy has been applied as a standard treatment for lung adenocarcinoma (LUAD) and lung squamous cell carcinoma (LUSC), and it has improved the outcomes of chemotherapy. Maintenance treatment with anti-PD-1 antibody (aPD-1) enhances the effect of immunochemical combination therapy and improves therapeutic efficacy, which contributes toward a significant improvement in patient survival rates. The AXL receptor tyrosine kinase (AXL), which is expressed in tumor cells, plays an essential role in the resistance of cancers to chemotherapy and immunotherapy, and stimulates signaling associated with epithelial-mesenchymal transition (EMT) in metastatic cancer. AXL is thus an attractive target for controlling resistance to anti-tumor therapies. In this study, we examined the effect of AXL inhibitors on immune activation and tumor growth in TC1 and C3PQ mouse tumor models, in the context of clinical immunotherapy/chemotherapy and maintenance treatment, using an aPD-1 with/without pemetrexed. To determine the optimal timing for administration of SKI-G-801, an AXL inhibitor, we investigated its anti-tumor effects based on inclusion at the immunochemotherapy and maintenance therapy stages. We also performed flow cytometry-based immune profiling of myeloid cells and lymphoid cells at different points in the treatment schedule, to investigate the immune activation and anti-tumor effects of the AXL inhibitor. The addition of SKI-G-801 to the immune checkpoint inhibitor and chemotherapy stage, as well as the maintenance therapy stage, produced the best anti-tumor results, and significant tumor growth inhibition was observed in both the TC1 and C3PQ models. Both models also exhibited increased proportion of effector memory helper T cells and increased expression of CD86+ macrophages. Especially, regulatory T cells were significantly reduced in the TC1 tumor model and there was an increase in central memory cytotoxic T cell infiltration and an increased proportion of macrophages with high CD80 expression in the C3PQ tumor model. These results suggest increased infiltration of T cells, consistent with previous studies using AXL inhibitors. It is expected that the results from this study will serve as a stepping stone for clinical research to improve the existing standard of care.
Collapse
Affiliation(s)
- Wongeun Lee
- JEUK Institute for Cancer Research, JEUK Co., Ltd., Gumi-si, South Korea.,Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, South Korea
| | - Dong Kwon Kim
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, South Korea
| | - Chun-Bong Synn
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, South Korea
| | - Hee Kyu Lee
- Department of Discovery Biology, Oscotec Inc., Seongnam, South Korea
| | - Sungho Park
- Department of Discovery Biology, Oscotec Inc., Seongnam, South Korea
| | - Dong-Sik Jung
- Department of Discovery Biology, Oscotec Inc., Seongnam, South Korea
| | - Yewon Choi
- Department of Discovery Biology, Oscotec Inc., Seongnam, South Korea
| | - Jae Hwan Kim
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, South Korea
| | - Youngseon Byeon
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, South Korea
| | - Young Seob Kim
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, South Korea
| | - Seul Lee
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, South Korea
| | - Soyeon Lee
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, South Korea
| | - Yunjoo Joo
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, South Korea
| | - Eun Ji Lee
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, South Korea
| | - Mi Ran Yun
- JEUK Institute for Cancer Research, JEUK Co., Ltd., Gumi-si, South Korea
| | - Seong Gu Heo
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, South Korea
| | - Wookyeom Yang
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, South Korea
| | - Ji Eun Jung
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, South Korea
| | - Eun Kyung Kim
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, South Korea
| | - Jooyeon Park
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, South Korea
| | - June Dong Park
- Wide River Institute of Immunology, Seoul National University, Hongcheon, South Korea.,Department of Pediatrics, Seoul National University College of Medicine, Seoul, South Korea
| | - Doo Jae Lee
- Wide River Institute of Immunology, Seoul National University, Hongcheon, South Korea
| | - Hyeon-Woo Kim
- Wide River Institute of Immunology, Seoul National University, Hongcheon, South Korea
| | - Sun Min Lim
- Division of Medical Oncology, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, South Korea
| | - Min Hee Hong
- Division of Medical Oncology, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, South Korea
| | - Beung-Chul Ahn
- Division of Medical Oncology, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, South Korea.,Center for Lung Cancer, National Cancer Center, Goyang-si, South Korea
| | - Jii Bum Lee
- Division of Hemato-oncology, Wonju Severance Christian Hospital, Yonsei University Wonju College of Medicine, Wonju, South Korea
| | - Kyoung-Ho Pyo
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, South Korea.,Division of Medical Oncology, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, South Korea
| |
Collapse
|
145
|
Howard FHN, Al-Janabi H, Patel P, Cox K, Smith E, Vadakekolathu J, Pockley AG, Conner J, Nohl JF, Allwood DA, Collado-Rojas C, Kennerley A, Staniland S, Muthana M. Nanobugs as Drugs: Bacterial Derived Nanomagnets Enhance Tumor Targeting and Oncolytic Activity of HSV-1 Virus. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2104763. [PMID: 35076148 DOI: 10.1002/smll.202104763] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 12/18/2021] [Indexed: 06/14/2023]
Abstract
The survival strategies of infectious organisms have inspired many therapeutics over the years. Indeed the advent of oncolytic viruses (OVs) exploits the uncontrolled replication of cancer cells for production of their progeny resulting in a cancer-targeting treatment that leaves healthy cells unharmed. Their success against inaccessible tumors however, is highly variable due to inadequate tumor targeting following systemic administration. Coassembling herpes simplex virus (HSV1716) with biocompatible magnetic nanoparticles derived from magnetotactic bacteria enables tumor targeting from circulation with magnetic guidance, protects the virus against neutralizing antibodies and thereby enhances viral replication within tumors. This approach additionally enhances the intratumoral recruitment of activated immune cells, promotes antitumor immunity and immune cell death, thereby inducing tumor shrinkage and increasing survival in a syngeneic mouse model of breast cancer by 50%. Exploiting the properties of such a nanocarrier, rather than tropism of the virus, for active tumor targeting offers an exciting, novel approach for enhancing the bioavailability and treatment efficacy of tumor immunotherapies for disseminated neoplasms.
Collapse
Affiliation(s)
- Faith H N Howard
- Department of Oncology and Metabolism, Mellanby Centre for Bone Research, University of Sheffield, Beech Hill Road, Sheffield, S10 2RX, UK
| | - Haider Al-Janabi
- Department of Oncology and Metabolism, Mellanby Centre for Bone Research, University of Sheffield, Beech Hill Road, Sheffield, S10 2RX, UK
| | - Priya Patel
- Department of Oncology and Metabolism, Mellanby Centre for Bone Research, University of Sheffield, Beech Hill Road, Sheffield, S10 2RX, UK
| | - Katie Cox
- Department of Oncology and Metabolism, Mellanby Centre for Bone Research, University of Sheffield, Beech Hill Road, Sheffield, S10 2RX, UK
| | - Emily Smith
- NMRC, School of Chemistry, University of Nottingham, Nottingham, NG7 2RD, UK
| | - Jayakumar Vadakekolathu
- John van Geest Cancer Research Centre, Centre for Health, Ageing and Understanding Disease (CHAUD), School of Science and Technology, Nottingham Trent University, Nottingham, NG11 8NS, UK
| | - A Graham Pockley
- John van Geest Cancer Research Centre, Centre for Health, Ageing and Understanding Disease (CHAUD), School of Science and Technology, Nottingham Trent University, Nottingham, NG11 8NS, UK
| | - Joe Conner
- Invizius Ltd, BioCity, Bo'ness road, Newhouse, ML1 5UH, UK
| | - James F Nohl
- Department of Materials Science and Engineering, University of Sheffield, Mappin Street, Sheffield, S1 3JD, UK
| | - Dan A Allwood
- Department of Materials Science and Engineering, University of Sheffield, Mappin Street, Sheffield, S1 3JD, UK
| | - Cristal Collado-Rojas
- Department of Oncology and Metabolism, Mellanby Centre for Bone Research, University of Sheffield, Beech Hill Road, Sheffield, S10 2RX, UK
| | - Aneurin Kennerley
- Department of Chemistry, University of York, Heslington, York, YO10 5DD, UK
| | - Sarah Staniland
- Department of Chemistry, University of Sheffield, Brook Hill, Sheffield, S3 7HF, UK
| | - Munitta Muthana
- Department of Oncology and Metabolism, Mellanby Centre for Bone Research, University of Sheffield, Beech Hill Road, Sheffield, S10 2RX, UK
| |
Collapse
|
146
|
Vergato C, Doshi KA, Roblyer D, Waxman DJ. Type-I interferon signaling is essential for robust metronomic chemo-immunogenic tumor regression in murine breast cancer. CANCER RESEARCH COMMUNICATIONS 2022; 2:246-257. [PMID: 36187936 PMCID: PMC9524291 DOI: 10.1158/2767-9764.crc-21-0148] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Many patients with breast cancer have a poor prognosis with limited therapeutic options. Here, we investigated the potential of chemo-immunogenic therapy as an avenue of treatment. We utilized two syngeneic mouse mammary tumor models, 4T1 and E0771, to examine the chemo-immunogenic potential of cyclophosphamide and the mechanistic contributions of cyclophosphamide-activated type-I interferon (IFN) signaling to therapeutic activity. Chemically-activated cyclophosphamide induced robust IFNα/β receptor-1-dependent signaling linked to hundreds of IFN-stimulated gene responses in both cell lines. Further, in 4T1 tumors, cyclophosphamide given on a medium-dose, 6-day intermittent metronomic schedule induced strong IFN signaling but comparatively weak immune cell infiltration associated with long-term tumor growth stasis. Induction of IFN signaling was somewhat weaker in E0771 tumors but was followed by widespread downstream gene responses, robust immune cell infiltration and extensive, prolonged tumor regression. The immune dependence of these effective anti-tumor responses was established by CD8 T-cell immunodepletion, which blocked cyclophosphamide-induced E0771 tumor regression and led to tumor stasis followed by regrowth. Strikingly, IFNα/β receptor-1 antibody blockade was even more effective in preventing E0771 immune cell infiltration and blocked the major tumor regression induced by cyclophosphamide treatment. Type-I IFN signaling is thus essential for the robust chemo-immunogenic response of these tumors to cyclophosphamide administered on a metronomic schedule.
Collapse
Affiliation(s)
- Cameron Vergato
- Department of Biology, Boston University, Boston, Massachusetts
| | - Kshama A. Doshi
- Department of Biology, Boston University, Boston, Massachusetts
| | - Darren Roblyer
- Department of Biomedical Engineering, Boston University, Boston, Massachusetts
| | - David J. Waxman
- Department of Biology, Boston University, Boston, Massachusetts
- Department of Biomedical Engineering, Boston University, Boston, Massachusetts
- Corresponding Author: David J. Waxman, Department of Biology, Boston University, 5 Cummington Mall, Boston, MA 02215. Phone: 617-353-7401; E-mail:
| |
Collapse
|
147
|
Applying nanotechnology to boost cancer immunotherapy by promoting immunogenic cell death. CHINESE CHEM LETT 2022. [DOI: 10.1016/j.cclet.2021.10.074] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
|
148
|
Chen XY, Yung LYL, Tan PH, Bay BH. Harnessing the Immunogenic Potential of Gold Nanoparticle-Based Platforms as a Therapeutic Strategy in Breast Cancer Immunotherapy: A Mini Review. Front Immunol 2022; 13:865554. [PMID: 35432376 PMCID: PMC9008216 DOI: 10.3389/fimmu.2022.865554] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Accepted: 03/14/2022] [Indexed: 12/24/2022] Open
Abstract
Breast cancer remains the most common malignancy among women worldwide. Although the implementation of mammography has dramatically increased the early detection rate, conventional treatments like chemotherapy, radiation therapy, and surgery, have significantly improved the prognosis for breast cancer patients. However, about a third of treated breast cancer patients are known to suffer from disease recurrences and progression to metastasis. Immunotherapy has recently gained traction due to its ability to establish long-term immune surveillance, and response for the prevention of disease recurrence and extension of patient survival. Current research findings have revealed that gold nanoparticles can enhance the safety and efficacy of cancer immunotherapy, through their unique intrinsic properties of good biocompatibility, durability, convenient surface modification, as well as enhanced permeability and retention effect. Gold nanoparticles are also able to induce innate immune responses through the process of immunogenic cell death, which can lead to the establishment of lasting adaptive immunity. As such gold nanoparticles are considered as good candidates for next generation immunotherapeutic strategies. This mini review gives an overview of gold nanoparticles and their potential applications in breast cancer immunotherapeutic strategies.
Collapse
Affiliation(s)
- Xiao-Yang Chen
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Division of Pathology, Singapore General Hospital, Singapore, Singapore
| | - Lin-Yue Lanry Yung
- Department of Biomolecular and Chemical Engineering, College of Design and Engineering, National University of Singapore, Singapore, Singapore
| | - Puay Hoon Tan
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Division of Pathology, Singapore General Hospital, Singapore, Singapore
- Duke-NUS Medical School, Singapore, Singapore
| | - Boon Huat Bay
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| |
Collapse
|
149
|
Regulation of the release of damage-associated molecular patterns from necroptotic cells. Biochem J 2022; 479:677-685. [PMID: 35293986 DOI: 10.1042/bcj20210604] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 02/23/2022] [Accepted: 02/25/2022] [Indexed: 02/07/2023]
Abstract
Damage-associated molecular patterns (DAMPs) are molecules within living cells that are released when cell membranes are ruptured. Although DAMPs have physiological functions inside the cell, once DAMPs are released extracellularly, they elicit various biological responses, including inflammation, proliferation, tissue damage, and tissue repair, in a context-dependent manner. In past decades, it was assumed that the release of DAMPs was induced by a membrane rupture, caused by passive ATP depletion, or by chemical or mechanical damage to the membrane. However, that concept has been challenged by recent advancements in understanding the regulation of cell death. Necroptosis is a form of regulated cell death, where cells show necrotic morphology. Necroptosis is triggered by death receptors, toll-like receptors, and some viral infections. The membrane rupture is executed by the mixed lineage-like kinase domain-like pseudokinase (MLKL), which forms oligomers that translocate to the plasma membrane during necroptosis. Although the causal relationship between MLKL function and membrane rupture has been extensively investigated, the detailed molecular mechanisms by which oligomerized MLKL induces membrane rupture are not fully understood. This review summarizes recent advances in understanding how MLKL regulates DAMP release and new technologies for visualizing DAMP release at single-cell resolution.
Collapse
|
150
|
Imran KM, Nagai-Singer MA, Brock RM, Alinezhadbalalami N, Davalos RV, Allen IC. Exploration of Novel Pathways Underlying Irreversible Electroporation Induced Anti-Tumor Immunity in Pancreatic Cancer. Front Oncol 2022; 12:853779. [PMID: 35372046 PMCID: PMC8972192 DOI: 10.3389/fonc.2022.853779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 02/28/2022] [Indexed: 12/04/2022] Open
Abstract
Advancements in medical sciences and technologies have significantly improved the survival of many cancers; however, pancreatic cancer remains a deadly diagnosis. This malignancy is often diagnosed late in the disease when metastases have already occurred. Additionally, the location of the pancreas near vital organs limits surgical candidacy, the tumor's immunosuppressive environment limits immunotherapy success, and it is highly resistant to radiation and chemotherapy. Hence, clinicians and patients alike need a treatment paradigm that reduces primary tumor burden, activates systemic anti-tumor immunity, and reverses the local immunosuppressive microenvironment to eventually clear distant metastases. Irreversible electroporation (IRE), a novel non-thermal tumor ablation technique, applies high-voltage ultra-short pulses to permeabilize targeted cell membranes and induce cell death. Progression with IRE technology and an array of research studies have shown that beyond tumor debulking, IRE can induce anti-tumor immune responses possibly through tumor neo-antigen release. However, the success of IRE treatment (i.e. full ablation and tumor recurrence) is variable. We believe that IRE treatment induces IFNγ expression, which then modulates immune checkpoint molecules and thus leads to tumor recurrence. This indicates a co-therapeutic use of IRE and immune checkpoint inhibitors as a promising treatment for pancreatic cancer patients. Here, we review the well-defined and speculated pathways involved in the immunostimulatory effects of IRE treatment for pancreatic cancer, as well as the regulatory pathways that may negate these anti-tumor responses. By defining these underlying mechanisms, future studies may identify improvements to systemic immune system engagement following local tumor ablation with IRE and beyond.
Collapse
Affiliation(s)
- Khan Mohammad Imran
- Graduate Program in Translational Biology, Medicine and Health, Virginia Tech, Roanoke, VA, United States
| | - Margaret A. Nagai-Singer
- Department of Biomedical Sciences and Pathobiology, Virginia Tech, Blacksburg, VA, United States
- Institute for Critical Technology and Applied Sciences, Virginia Tech, Blacksburg, VA, United States
| | - Rebecca M. Brock
- Graduate Program in Translational Biology, Medicine and Health, Virginia Tech, Roanoke, VA, United States
| | - Nastaran Alinezhadbalalami
- Institute for Critical Technology and Applied Sciences, Virginia Tech, Blacksburg, VA, United States
- Department of Biomedical Engineering and Mechanics, Virginia Tech, Blacksburg, VA, United States
| | - Rafael V. Davalos
- Institute for Critical Technology and Applied Sciences, Virginia Tech, Blacksburg, VA, United States
- Department of Biomedical Engineering and Mechanics, Virginia Tech, Blacksburg, VA, United States
- Department of Mechanical Engineering, Virginia Tech, Blacksburg, VA, United States
| | - Irving Coy Allen
- Department of Biomedical Sciences and Pathobiology, Virginia Tech, Blacksburg, VA, United States
- Institute for Critical Technology and Applied Sciences, Virginia Tech, Blacksburg, VA, United States
| |
Collapse
|