151
|
Mabrouk N, Tran T, Sam I, Pourmir I, Gruel N, Granier C, Pineau J, Gey A, Kobold S, Fabre E, Tartour E. CXCR6 expressing T cells: Functions and role in the control of tumors. Front Immunol 2022; 13:1022136. [PMID: 36311728 PMCID: PMC9597613 DOI: 10.3389/fimmu.2022.1022136] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 09/27/2022] [Indexed: 11/13/2022] Open
Abstract
CXCR6 is a receptor for the chemokine CXCL16, which exists as a membrane or soluble form. CXCR6 is a marker for resident memory T (TRM) cells that plays a role in immunosurveillance through their interaction with epithelial cells. The interaction of CXCR6 with CXCL16 expressed at the membrane of certain subpopulations of intratumor dendritic cells (DC) called DC3, ideally positions these CXCR6+ T cells to receive a proliferation signal from IL-15 also presented by DC3. Mice deficient in cxcr6 or blocking the interaction of CXCR6 with its ligand, experience a poorer control of tumor proliferation by CD8+ T cells, but also by NKT cells especially in the liver. Intranasal vaccination induces CXCL16 production in the lungs and is associated with infiltration by TRM expressing CXCR6, which are then required for the efficacy of anti-tumor vaccination. Therapeutically, the addition of CXCR6 to specific CAR-T cells enhances their intratumoral accumulation and prolongs survival in animal models of pancreatic, ovarian and lung cancer. Finally, CXCR6 is part of immunological signatures that predict response to immunotherapy based on anti-PD-(L)1 in various cancers. In contrast, a protumoral role of CXCR6+T cells has also been reported mainly in Non-alcoholic steatohepatitis (NASH) due to a non-antigen specific mechanism. The targeting and amplification of antigen-specific TRM expressing CXCR6 and its potential use as a biomarker of response to immunotherapy opens new perspectives in cancer treatment.
Collapse
Affiliation(s)
| | - Thi Tran
- Université ParisCité, INSERM, PARCC, Paris, France
| | - Ikuan Sam
- Université ParisCité, INSERM, PARCC, Paris, France
| | - Ivan Pourmir
- Université ParisCité, INSERM, PARCC, Paris, France
| | - Nadège Gruel
- Institut Curie, PSL Research University, Department of Translational Research, Paris, France
- INSERM U830, Equipe labellisée LNCC, Siredo Oncology Centre, Institut Curie, Paris, France
| | - Clémence Granier
- Université ParisCité, INSERM, PARCC, Paris, France
- Immunology, APHP, Hôpital Europeen Georges Pompidou and Hôpital Necker, Paris, France
| | - Joséphine Pineau
- Université ParisCité, INSERM, PARCC, Paris, France
- Immunology, APHP, Hôpital Europeen Georges Pompidou and Hôpital Necker, Paris, France
| | - Alain Gey
- Université ParisCité, INSERM, PARCC, Paris, France
- Immunology, APHP, Hôpital Europeen Georges Pompidou and Hôpital Necker, Paris, France
| | - Sebastian Kobold
- Division of Clinical Pharmacology, Department of Medicine IV, Klinikum der Universität München, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
| | - Elizabeth Fabre
- Université ParisCité, INSERM, PARCC, Paris, France
- Lung Oncology Unit, APHP, Hôpital Européen Georges Pompidou, Paris, France
| | - Eric Tartour
- Université ParisCité, INSERM, PARCC, Paris, France
- Immunology, APHP, Hôpital Europeen Georges Pompidou and Hôpital Necker, Paris, France
- Equipe Labellisée Ligue contre le Cancer, Paris, France
- *Correspondence: Eric Tartour,
| |
Collapse
|
152
|
Li Y, Chen C, Liu HL, Zhang ZF, Wang CL. LARRPM restricts lung adenocarcinoma progression and M2 macrophage polarization through epigenetically regulating LINC00240 and CSF1. Cell Mol Biol Lett 2022; 27:91. [PMID: 36221069 PMCID: PMC9552444 DOI: 10.1186/s11658-022-00376-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Accepted: 08/17/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Long non-coding RNAs (lncRNAs) are critical regulators in lung adenocarcinoma (LUAD). M2-type tumor-associated macrophages (TAMs) also play oncogenic roles in LUAD. However, the involvement of lncRNAs in TAM activation is still largely unknown. METHODS The expressions of LARRPM, LINC00240 and CSF1 were determined by RT-qPCR. The regulation of LINC00240 and CSF1 by LARRPM was investigated by RNA-protein pull-down, RNA immunoprecipitation, chromatin immunoprecipitation and bisulfite DNA sequencing. In vitro and in vivo gain- and loss-of-function assays were performed to investigate the roles of LARRPM. RESULTS The lncRNA LARRPM was expressed at low levels in LUAD tissues and cells. The low expression of LARRPM was correlated with advanced stage and poor survival of patients with LUAD. Functional experiments revealed that LARRPM suppressed LUAD cell proliferation, migration and invasion, and promoted apoptosis. LARRPM also repressed macrophage M2 polarization and infiltration. Taken together, LARRPM significantly restricted LUAD progression in vivo. Mechanistically, LARRPM bound and recruited DNA demethylase TET1 to the promoter of its anti-sense strand gene LINC00240, leading to a decrease in DNA methylation level of the LINC00240 promoter and transcriptional activation of LINC00240. Functional rescue assays suggested that the lncRNA LINC00240 was responsible for the roles of LARRPM in the malignant behavior of LUAD cells. LARRPM decreased the binding of TET1 to the CSF1 promoter, resulting in increased DNA methylation of the CSF1 promoter and transcriptional repression of CSF1, which is responsible for the roles of LARRPM in macrophage M2 polarization and infiltration. The TAMs educated by LUAD cells exerted oncogenic roles, which was negatively regulated by LARRPM expressed in LUAD cells. CONCLUSIONS LARRPM restricts LUAD progression through repressing both LUAD cell and macrophages. These data shed new insights into the regulation of LUAD progression by lncRNAs and provide data on the potential utility of LARRPM as a target for LUAD treatment.
Collapse
Affiliation(s)
- Yue Li
- Department of Lung Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin Lung Cancer Center, Tianjin, 300060, China
| | - Chen Chen
- Department of Lung Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin Lung Cancer Center, Tianjin, 300060, China
| | - Hai-Lin Liu
- Department of Lung Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin Lung Cancer Center, Tianjin, 300060, China
| | - Zhen-Fa Zhang
- Department of Lung Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin Lung Cancer Center, Tianjin, 300060, China
| | - Chang-Li Wang
- Department of Lung Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin Lung Cancer Center, Tianjin, 300060, China.
| |
Collapse
|
153
|
Bai KH, Zhang YY, Li XP, Tian XP, Pan MM, Wang DW, Dai YJ. Comprehensive analysis of tumor necrosis factor-α-inducible protein 8-like 2 (TIPE2): A potential novel pan-cancer immune checkpoint. Comput Struct Biotechnol J 2022; 20:5226-5234. [PMID: 36187930 PMCID: PMC9508481 DOI: 10.1016/j.csbj.2022.09.021] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 09/07/2022] [Accepted: 09/13/2022] [Indexed: 11/03/2022] Open
Abstract
Tumor necrosis factor-α-inducible protein 8-like 2 (TIPE2) is encoded by TNFAIP8L2 and is a newly identified negative regulator of natural and acquired immunity that plays a critical function in maintaining immune homeostasis. Recently, CAR-NK immune cell therapy has been a focus of major research efforts as a novel cancer therapeutic strategy. TIPE2 is a potential checkpoint molecule for immune cell maturation and antitumor immunity that could be used as a novel NK cell-based immunotherapeutic approach. In this study, we explored the expression of TNFAIP8L2 across various tumor types and found that TNFAIP8L2 was highly expressed in most tumor types and correlated with prognosis. Survival analysis showed that TNFAIP8L2 expression was predictive of improved survival in cervical-squamous-cell-carcinoma (CESC), sarcoma (SARC) and skin-cutaneous-melanoma (SKCM). Conversely, TNFAIP8L2 expression predicted poorer survival in acute myeloid leukemia (LAML), lower-grade-glioma (LGG), kidney-renal-clear-cell-carcinoma (KIRC) and uveal-melanoma (UVM). Analysis of stemness features and immune cell infiltration indicated that TNFAIP8L2 was significantly associated with cancer stem cell index and increased macrophage and dendritic cell infiltration. Our data suggest that TNFAIP8L2 may be a novel immune checkpoint biomarker across different tumor types, particularly in LAML, LGG, KIRC and UVM, and may have further utility as a potential target for immunotherapy.
Collapse
Key Words
- BLCA, Bladder Urothelial Carcinoma
- BRCA, Breast invasive carcinoma
- CESC, Cervical squamous cell carcinoma and endocervical adenocarcinoma
- CHOL, Cholangiocarcinoma
- COAD, Colon adenocarcinoma
- ESCA, Esophageal carcinoma
- GBM, Glioblastoma multiforme
- HNSC, Head and Neck squamous cell carcinoma
- Immune cell infiltration
- KICH, Kidney Chromophobe
- KIRC, Kidney renal clear cell carcinoma
- KIRP, Kidney renal papillary cell carcinoma
- LAML, Acute Myeloid Leukemia
- LGG, Lower Grade Glioma
- LIHC, Liver hepatocellular carcinoma
- LUAD, Lung adenocarcinoma
- LUSC, Lung squamous cell carcinoma
- MESO, Mesothelioma
- New immune checkpoint
- OV, Ovarian serous cystadenocarcinoma
- PAAD, Pancreatic adenocarcinoma
- PCPG, Pheochromocytoma and Paraganglioma
- PRAD, Prostate adenocarcinoma
- Pan-cancer
- READ, Rectum adenocarcinoma
- SARC, Sarcoma
- SKCM, Skin Cutaneous Melanoma
- STAD, Stomach adenocarcinoma
- Stemness feature
- TGCT, Testicular Germ Cell Tumor
- THCA, Thyroid carcinoma
- THYM, Thymoma
- TIPE2
- UCEC, Uterine Corpus Endometrial Carcinoma
- UCS, Uterine Carcinosarcoma
- UVM, Uveal Melanoma
Collapse
Affiliation(s)
- Kun-Hao Bai
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng East Road, Guangzhou 500020, Guangdong, China
- Department of Endoscopy, Sun Yat-sen University Cancer Center, Guangzhou, 500026, China
| | - Yi-Yang Zhang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng East Road, Guangzhou 500020, Guangdong, China
- Department of Endoscopy, Sun Yat-sen University Cancer Center, Guangzhou, 500026, China
| | - Xue-Ping Li
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng East Road, Guangzhou 500020, Guangdong, China
- Department of Hematologic Oncology, Sun Yat-sen University Cancer Center, Guangzhou 500020, Guangdong, China
| | - Xiao-Peng Tian
- Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu, China
| | - Meng-Meng Pan
- National Research Center for Translational Medicine, Ruijin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, 197, Ruijin Road II, Shanghai 200025, China
| | - Da-Wei Wang
- National Research Center for Translational Medicine, Ruijin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, 197, Ruijin Road II, Shanghai 200025, China
| | - Yu-Jun Dai
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng East Road, Guangzhou 500020, Guangdong, China
- Department of Hematologic Oncology, Sun Yat-sen University Cancer Center, Guangzhou 500020, Guangdong, China
| |
Collapse
|
154
|
Katoh M, Katoh M. WNT signaling and cancer stemness. Essays Biochem 2022; 66:319-331. [PMID: 35837811 PMCID: PMC9484141 DOI: 10.1042/ebc20220016] [Citation(s) in RCA: 83] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 07/07/2022] [Accepted: 07/08/2022] [Indexed: 12/11/2022]
Abstract
Cancer stemness, defined as the self-renewal and tumor-initiation potential of cancer stem cells (CSCs), is a cancer biology property featuring activation of CSC signaling networks. Canonical WNT signaling through Frizzled and LRP5/6 receptors is transmitted to the β-catenin-TCF/LEF-dependent transcription machinery to up-regulate MYC, CCND1, LGR5, SNAI1, IFNG, CCL28, CD274 (PD-L1) and other target genes. Canonical WNT signaling causes expansion of rapidly cycling CSCs and modulates both immune surveillance and immune tolerance. In contrast, noncanonical WNT signaling through Frizzled or the ROR1/2 receptors is transmitted to phospholipase C, Rac1 and RhoA to control transcriptional outputs mediated by NFAT, AP-1 and YAP-TEAD, respectively. Noncanonical WNT signaling supports maintenance of slowly cycling, quiescent or dormant CSCs and promotes epithelial-mesenchymal transition via crosstalk with TGFβ (transforming growth factor-β) signaling cascades, while the TGFβ signaling network induces immune evasion. The WNT signaling network orchestrates the functions of cancer-associated fibroblasts, endothelial cells and immune cells in the tumor microenvironment and fine-tunes stemness in human cancers, such as breast, colorectal, gastric and lung cancers. Here, WNT-related cancer stemness features, including proliferation/dormancy plasticity, epithelial-mesenchymal plasticity and immune-landscape plasticity, will be discussed. Porcupine inhibitors, β-catenin protein-protein interaction inhibitors, β-catenin proteolysis targeting chimeras, ROR1 inhibitors and ROR1-targeted biologics are investigational drugs targeting WNT signaling cascades. Mechanisms of cancer plasticity regulated by the WNT signaling network are promising targets for therapeutic intervention; however, further understanding of context-dependent reprogramming trajectories might be necessary to optimize the clinical benefits of WNT-targeted monotherapy and applied combination therapy for patients with cancer.
Collapse
Affiliation(s)
| | - Masaru Katoh
- M & M Precision Medicine
- Department of Omics Network, National Cancer Center, Japan
- Department of Clinical Genomics, National Cancer Center, Japan
| |
Collapse
|
155
|
Karam A, Mjaess G, Martinez Chanza N, Aoun F, Bou Kheir G, Younes H, Kazzi H, Albisinni S, Roumeguère T. CAR-T cell therapy for solid tumors: are we still that far? A systematic review of literature. Cancer Invest 2022; 40:923-937. [DOI: 10.1080/07357907.2022.2125004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Affiliation(s)
- Aya Karam
- Hotel-Dieu de France, University of Saint Joseph, Faculty of Medicine, Beirut, Lebanon
| | - Georges Mjaess
- Department of Urology, Hôpital Universitaire de Bruxelles, Hôpital Érasme, Université Libre de Bruxelles, Brussels, Belgium
| | - Nieves Martinez Chanza
- Department of Medical Oncology, Hôpital Universitaire de Bruxelles, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | - Fouad Aoun
- Hotel-Dieu de France, University of Saint Joseph, Faculty of Medicine, Beirut, Lebanon
| | - George Bou Kheir
- Department of Urology, Hôpital Universitaire de Bruxelles, Hôpital Érasme, Université Libre de Bruxelles, Brussels, Belgium
| | - Hadi Younes
- Hotel-Dieu de France, University of Saint Joseph, Faculty of Medicine, Beirut, Lebanon
| | - Hanane Kazzi
- Department of Radiology, Saint Joseph Medical Center, Beirut, Lebanon
| | - Simone Albisinni
- Department of Urology, Hôpital Universitaire de Bruxelles, Hôpital Érasme, Université Libre de Bruxelles, Brussels, Belgium
| | - Thierry Roumeguère
- Department of Urology, Hôpital Universitaire de Bruxelles, Hôpital Érasme, Université Libre de Bruxelles, Brussels, Belgium
- Department of Urology, Hôpital Universitaire de Bruxelles, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| |
Collapse
|
156
|
Adoptive cell therapies in thoracic malignancies. Cancer Immunol Immunother 2022; 71:2077-2098. [PMID: 35129636 DOI: 10.1007/s00262-022-03142-3] [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] [Received: 09/04/2021] [Accepted: 12/27/2021] [Indexed: 12/22/2022]
Abstract
Immunotherapy has gained great interest in thoracic malignancies in the last decade, first in non-small cell lung cancer (NSCLC), but also more recently in small-cell lung cancer (SCLC) and malignant pleural mesothelioma (MPM). However, while 15-20% of patients will greatly benefit from immune checkpoint blockers (ICBs), a vast majority will rapidly exhibit resistance. Reasons for this are multiple: non-immunogenic tumors, immunosuppressive tumor microenvironment or defects in immune cells trafficking to the tumor sites being some of the most frequent. Current progress in adoptive cell therapies could offer a way to overcome these hurdles and bring effective immune cells to the tumor site. In this review, we discuss advantages, limits and future perspectives of adoptive cell therapy (ACT) in thoracic malignancies from lymphokine-activated killer cells (LAK), cytokine-induced killer cells (CIK), natural killer cells (NK), dendritic cells (DC) vaccines and tumor-infiltrating lymphocytes (TILs) to TCR engineering and CARs. Trials are still in their early phases, and while there may still be many limitations to overcome, a combination of these different approaches with ICBs, chemotherapy and/or radiotherapy could vastly improve the way we treat thoracic cancers.
Collapse
|
157
|
Lv W, Shi L, Pan J, Wang S. Comprehensive prognostic and immunological analysis of CCT2 in pan-cancer. Front Oncol 2022; 12:986990. [PMID: 36119498 PMCID: PMC9476648 DOI: 10.3389/fonc.2022.986990] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 08/01/2022] [Indexed: 11/18/2022] Open
Abstract
CCT2 acts as a molecular chaperone protein that assists in the proper folding of proteins, thus ensuring a dynamic balance of cellular homeostasis. Despite increasing evidence supporting the important role of CCT2 in the tumorigenesis of certain cancers, few articles that provide a systematic pan-cancer analysis of CCT2 have been published. Hence, to evaluate the expression status and prognostic significance of CCT2 in pan-cancers, an analysis of the relationship between CCT2 and different tumor immune cell infiltrations was conducted using datasets from the Cancer Genome Atlas, Cancer Cell Lineage Encyclopedia, and so on. In most cancers, CCT2 expression was high and was associated with poor prognosis. Moreover, CCT2 gene expression was negatively correlated with infiltration of most immune cells in 10 cancer types, and CCT2 expression was related to tumor mutation burden and microsatellite instability. The role that CCT2 plays in tumorigenesis and tumor immunity suggests that it can serve as a prognostic marker in many cancers.
Collapse
Affiliation(s)
- Wenming Lv
- Department of Neurology, Second Hospital of Lanzhou University, Lanzhou, China
| | - Lin Shi
- Department of Hematology, Peking University International Hospital, Beijing, China
| | - Jiebing Pan
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, China
| | - Shengbao Wang
- Emergency Center of the Second Hospital of Lanzhou University, Lanzhou, China
| |
Collapse
|
158
|
Füchsl F, Krackhardt AM. Paving the Way to Solid Tumors: Challenges and Strategies for Adoptively Transferred Transgenic T Cells in the Tumor Microenvironment. Cancers (Basel) 2022; 14:4192. [PMID: 36077730 PMCID: PMC9454442 DOI: 10.3390/cancers14174192] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Revised: 08/22/2022] [Accepted: 08/25/2022] [Indexed: 01/10/2023] Open
Abstract
T cells are important players in the antitumor immune response. Over the past few years, the adoptive transfer of genetically modified, autologous T cells-specifically redirected toward the tumor by expressing either a T cell receptor (TCR) or a chimeric antigen receptor (CAR)-has been adopted for use in the clinic. At the moment, the therapeutic application of CD19- and, increasingly, BCMA-targeting-engineered CAR-T cells have been approved and have yielded partly impressive results in hematologic malignancies. However, employing transgenic T cells for the treatment of solid tumors remains more troublesome, and numerous hurdles within the highly immunosuppressive tumor microenvironment (TME) need to be overcome to achieve tumor control. In this review, we focused on the challenges that these therapies must face on three different levels: infiltrating the tumor, exerting efficient antitumor activity, and overcoming T cell exhaustion and dysfunction. We aimed to discuss different options to pave the way for potent transgenic T cell-mediated tumor rejection by engineering either the TME or the transgenic T cell itself, which responds to the environment.
Collapse
Affiliation(s)
- Franziska Füchsl
- Klinik und Poliklinik für Innere Medizin III, School of Medicine, Technische Universität München, Klinikum rechts der Isar, Ismaningerstr. 22, 81675 Munich, Germany
| | - Angela M. Krackhardt
- Klinik und Poliklinik für Innere Medizin III, School of Medicine, Technische Universität München, Klinikum rechts der Isar, Ismaningerstr. 22, 81675 Munich, Germany
- German Cancer Consortium of Translational Cancer Research (DKTK) and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- Center for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, 81675 Munich, Germany
| |
Collapse
|
159
|
Peng H, Nerreter T, Mestermann K, Wachter J, Chang J, Hudecek M, Rader C. ROR1-targeting switchable CAR-T cells for cancer therapy. Oncogene 2022; 41:4104-4114. [PMID: 35859167 PMCID: PMC9398970 DOI: 10.1038/s41388-022-02416-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 07/07/2022] [Accepted: 07/08/2022] [Indexed: 02/03/2023]
Abstract
The success of chimeric antigen receptor T cell (CAR-T) therapy in the treatment of hematologic malignancies has prompted the development of numerous CAR-T technologies, including switchable CAR-T (sCAR-T) systems that combine a universal CAR-T with bispecific adapter proteins. Owing to their controllability and versatility, sCAR-Ts have received considerable attention. To explore the therapeutic utility of sCAR-Ts targeting the receptor tyrosine kinase ROR1, which is expressed in hematologic and solid malignancies, and to identify bispecific adaptor proteins that efficiently mediate universal CAR-T engagement, a panel of switches based on ROR1-targeting Fabs with different epitopes and affinities was compared in in vitro and in vivo models of ROR1-expressing cancers. For switches targeting overlapping or identical epitopes, potency correlated with affinity. Surprisingly, however, we identified a switch targeting a unique epitope with low affinity but mediating potent and selective antitumor activity in vitro and in vivo. Converted to a conventional CAR-T, the same anti-ROR1 mAb (324) outperformed a clinically investigated conventional CAR-T that is based on an anti-ROR1 mAb (R12) with ~200-fold higher affinity. Thus, demonstrating therapeutic utility on their own, sCAR-Ts also facilitate higher throughput screening for the identification of conventional CAR-T candidates for preclinical and clinical studies.
Collapse
Affiliation(s)
- Haiyong Peng
- Department of Immunology and Microbiology, UF Scripps Biomedical Research, University of Florida, Jupiter, FL, 33458, USA.
| | - Thomas Nerreter
- Medizinische Klinik und Poliklinik II, Universitätsklinikum Würzburg, Oberdürrbacher Strasse 6, 97080, Würzburg, Germany
| | - Katrin Mestermann
- Medizinische Klinik und Poliklinik II, Universitätsklinikum Würzburg, Oberdürrbacher Strasse 6, 97080, Würzburg, Germany
| | - Jakob Wachter
- Medizinische Klinik und Poliklinik II, Universitätsklinikum Würzburg, Oberdürrbacher Strasse 6, 97080, Würzburg, Germany
| | - Jing Chang
- Department of Immunology and Microbiology, UF Scripps Biomedical Research, University of Florida, Jupiter, FL, 33458, USA
| | - Michael Hudecek
- Medizinische Klinik und Poliklinik II, Universitätsklinikum Würzburg, Oberdürrbacher Strasse 6, 97080, Würzburg, Germany
| | - Christoph Rader
- Department of Immunology and Microbiology, UF Scripps Biomedical Research, University of Florida, Jupiter, FL, 33458, USA.
| |
Collapse
|
160
|
Lv B, Wang Y, Ma D, Cheng W, Liu J, Yong T, Chen H, Wang C. Immunotherapy: Reshape the Tumor Immune Microenvironment. Front Immunol 2022; 13:844142. [PMID: 35874717 PMCID: PMC9299092 DOI: 10.3389/fimmu.2022.844142] [Citation(s) in RCA: 193] [Impact Index Per Article: 64.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Accepted: 06/13/2022] [Indexed: 12/12/2022] Open
Abstract
Tumor immune microenvironment (TIME) include tumor cells, immune cells, cytokines, etc. The interactions between these components, which are divided into anti-tumor and pro-tumor, determine the trend of anti-tumor immunity. Although the immune system can eliminate tumor through the cancer-immune cycle, tumors appear to eventually evade from immune surveillance by shaping an immunosuppressive microenvironment. Immunotherapy reshapes the TIME and restores the tumor killing ability of anti-tumor immune cells. Herein, we review the function of immune cells within the TIME and discuss the contribution of current mainstream immunotherapeutic approaches to remolding the TIME. Changes in the immune microenvironment in different forms under the intervention of immunotherapy can shed light on better combination treatment strategies.
Collapse
Affiliation(s)
- Bingzhe Lv
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, China.,The Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Yunpeng Wang
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, China.,The Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Dongjiang Ma
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, China.,The Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Wei Cheng
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, China.,The Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Jie Liu
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, China.,The Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Tao Yong
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, China.,The Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Hao Chen
- Key Laboratory of Digestive System Tumors of Gansu Province, Lanzhou University Second Hospital, Lanzhou, China.,Department of Surgical Oncology, Lanzhou University Second Hospital, Lanzhou, China
| | - Chen Wang
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, China.,Key Laboratory of Digestive System Tumors of Gansu Province, Lanzhou University Second Hospital, Lanzhou, China
| |
Collapse
|
161
|
Developing ROR1 Targeting CAR-T Cells against Solid Tumors in Preclinical Studies. Cancers (Basel) 2022; 14:cancers14153618. [PMID: 35892876 PMCID: PMC9331269 DOI: 10.3390/cancers14153618] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 07/18/2022] [Accepted: 07/21/2022] [Indexed: 11/16/2022] Open
Abstract
Chimeric antigen receptor (CAR)-modified T-cells (CAR-T) have demonstrated promising clinical benefits against B-cell malignancies. Yet, its application for solid tumors is still facing challenges. Unlike haematological cancers, solid tumors often lack good targets, which are ideally expressed on the tumor cells, but not by the normal healthy cells. Fortunately, receptor tyrosine kinase-like orphan receptor 1 (ROR1) is among a few good cancer targets that is aberrantly expressed on various tumors but has a low expression on normal tissue, suggesting it as a good candidate for CAR-T therapy. Here, we constructed two ROR1 CARs with the same antigen recognition domain that was derived from Zilovertamab but differing in hinge regions. Both CARs target ROR1+ cancer cells specifically, but CAR with a shorter IgG4 hinge exhibits a higher surface expression and better in vitro functionality. We further tested the ROR1 CAR-T in three human solid tumor xenografted mouse models. Our ROR1 CAR-T cells controlled the solid tumor growth without causing any severe toxicity. Our results demonstrated that ROR1 CAR-T derived from Zilovertamab is efficacious and safe to suppress ROR1+ solid tumors in vitro and in vivo, providing a promising therapeutic option for future clinical application.
Collapse
|
162
|
Zhang H, Zhu S, Deng W, Li R, Zhou H, Xiong H. The landscape of chimeric antigen receptor T cell therapy in breast cancer: Perspectives and outlook. Front Immunol 2022; 13:887471. [PMID: 35935930 PMCID: PMC9354605 DOI: 10.3389/fimmu.2022.887471] [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: 03/01/2022] [Accepted: 07/01/2022] [Indexed: 11/25/2022] Open
Abstract
Chimeric antigen receptor-T (CAR-T) cell therapy is a revolutionary adoptive cell therapy, which could modify and redirect T cells to specific tumor cells. Since CAR-T cell therapy was first approved for B cell-derived malignancies in 2017, it has yielded unprecedented progress in hematological tumors and has dramatically reshaped the landscape of cancer therapy in recent years. Currently, cumulative evidence has demonstrated that CAR-T cell therapy could be a viable therapeutic strategy for solid cancers. However, owing to the immunosuppressive tumor microenvironment (TME) and heterogenous tumor antigens, the application of CAR-T cell therapy against solid cancers requires circumventing more challenging obstacles. Breast cancer is characterized by a high degree of invasiveness, malignancy, and poor prognosis. The review highlights the underlying targets of CAR-T cell therapy in breast cancer, summarizes the challenges associated with CAR-T cell therapy, and proposes the strategies to overcome these challenges, which provides a novel approach to breast cancer treatment.
Collapse
|
163
|
Chen L, Chen F, Niu H, Li J, Pu Y, Yang C, Wang Y, Huang R, Li K, Lei Y, Huang Y. Chimeric Antigen Receptor (CAR)-T Cell Immunotherapy Against Thoracic Malignancies: Challenges and Opportunities. Front Immunol 2022; 13:871661. [PMID: 35911706 PMCID: PMC9334018 DOI: 10.3389/fimmu.2022.871661] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Accepted: 06/14/2022] [Indexed: 11/13/2022] Open
Abstract
Different from surgery, chemical therapy, radio-therapy and target therapy, Chimeric antigen receptor-modified T (CAR-T) cells, a novel adoptive immunotherapy strategy, have been used successfully against both hematological tumors and solid tumors. Although several problems have reduced engineered CAR-T cell therapeutic outcomes in clinical trials for the treatment of thoracic malignancies, including the lack of specific antigens, an immunosuppressive tumor microenvironment, a low level of CAR-T cell infiltration into tumor tissues, off-target toxicity, and other safety issues, CAR-T cell treatment is still full of bright future. In this review, we outline the basic structure and characteristics of CAR-T cells among different period, summarize the common tumor-associated antigens in clinical trials of CAR-T cell therapy for thoracic malignancies, and point out the current challenges and new strategies, aiming to provide new ideas and approaches for preclinical experiments and clinical trials of CAR-T cell therapy for thoracic malignancies.
Collapse
Affiliation(s)
- Long Chen
- Department of PET/CT Center, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Cancer Center of Yunnan Province, Kunming, China
| | - Fukun Chen
- Department of Nuclear Medicine, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Cancer Center of Yunnan Province, Kunming, China
| | - Huatao Niu
- Department of Neurosurgery, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Cancer Center of Yunnan Province, Kunming, China
| | - Jindan Li
- Department of PET/CT Center, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Cancer Center of Yunnan Province, Kunming, China
| | - Yongzhu Pu
- Department of PET/CT Center, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Cancer Center of Yunnan Province, Kunming, China
| | - Conghui Yang
- Department of PET/CT Center, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Cancer Center of Yunnan Province, Kunming, China
| | - Yue Wang
- Department of PET/CT Center, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Cancer Center of Yunnan Province, Kunming, China
| | - Rong Huang
- Department of PET/CT Center, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Cancer Center of Yunnan Province, Kunming, China
| | - Ke Li
- Department of Cancer Biotherapy Center, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Cancer Center of Yunnan Province, Kunming, China
| | - Yujie Lei
- Department of Thoracic Surgery I, Key Laboratory of Lung Cancer of Yunnan Province, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Cancer Center of Yunnan Province, Kunming, China
| | - Yunchao Huang
- Department of Thoracic Surgery I, Key Laboratory of Lung Cancer of Yunnan Province, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Cancer Center of Yunnan Province, Kunming, China
| |
Collapse
|
164
|
Liu P, Chen J, Zhao L, Hollebecque A, Kepp O, Zitvogel L, Kroemer G. PD-1 blockade synergizes with oxaliplatin-based, but not cisplatin-based, chemotherapy of gastric cancer. Oncoimmunology 2022; 11:2093518. [PMID: 35769948 PMCID: PMC9235886 DOI: 10.1080/2162402x.2022.2093518] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Preclinical experimentation revealed that established cancers treated with the immunogenic cell death (ICD) inducer oxaliplatin are sensitized to immune checkpoint inhibitors targeting PD-1. In contrast, no such sensitizing effect is observed when cisplatin, a non-immunogenic cell death inducer is used. Two randomized phase III clinical trials targeting unresectable gastric and gastro-esophageal junction carcinomas apparently validate this observation. Thus, oxaliplatin-based chemotherapy (together with capecitabine or 5-fluorouracil plus leucovorin) favorably interacted with nivolumab, yielding improved outcome. In contrast, the outcome of cisplatin-based chemotherapy (together with capecitabine or 5-fluorouracil) failed to be improved by concomitant treatment with pembrolizumab. These clinical findings underscore the importance of choosing appropriate ICD-inducing cytotoxicants for the development of chemoimmunotherapeutic regimens. Unfortunately, the FDA and EMA have approved PD-1 blockade in combination with “platinum-based chemotherapy” without specifying the precise nature of the platinum-containing drug. This is a non sequitur. Based on the available clinical data, such approvals should be restricted to the use of oxaliplatin.
Collapse
Affiliation(s)
- Peng Liu
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Center, Université Paris Saclay, Villejuif, France.,Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, Inserm U1138 and CNRS SNC 5096, Institut Universitaire de France, Paris, France
| | - Jianzhou Chen
- INSERM U1015, Equipe Labellisée - Ligue Nationale contre le Cancer, Villejuif, France
| | - Liwei Zhao
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Center, Université Paris Saclay, Villejuif, France.,Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, Inserm U1138 and CNRS SNC 5096, Institut Universitaire de France, Paris, France
| | - Antoine Hollebecque
- Department of Medical Oncology, Gustave Roussy Cancer Campus, Villejuif, France
| | - Oliver Kepp
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Center, Université Paris Saclay, Villejuif, France.,Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, Inserm U1138 and CNRS SNC 5096, Institut Universitaire de France, Paris, France
| | - Laurence Zitvogel
- INSERM U1015, Equipe Labellisée - Ligue Nationale contre le Cancer, Villejuif, France.,Department of Medical Oncology, Gustave Roussy Cancer Campus, Villejuif, France.,Gustave Roussy, ClinicObiome, Villejuif, France.,Center of Clinical Investigations in Biotherapies of Cancer (CICBT) 1428, Villejuif, France
| | - Guido Kroemer
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Center, Université Paris Saclay, Villejuif, France.,Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, Inserm U1138 and CNRS SNC 5096, Institut Universitaire de France, Paris, France.,Institut du Cancer Paris CARPEM, Department of Biology, APHP, Hôpital Européen Georges Pompidou, Paris, France
| |
Collapse
|
165
|
Li ZL, Wu H, Zhu JQ, Sun LY, Tong XM, Huang DS, Yang T. Novel Strategy for Optimized Nanocatalytic Tumor Therapy: From an Updated View. SMALL SCIENCE 2022; 2:2200024. [PMID: 40213113 PMCID: PMC11936048 DOI: 10.1002/smsc.202200024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Indexed: 11/08/2022] Open
Abstract
Nanozyme has been experiencing rapid development in biomedical applications involving biosensors, immunoassays, and antitumor agents in recent years due to its tunable catalytic performance and desirable biocompatibility. Since the first exploration of nanozyme-based Fenton reaction for nanocatalytic therapy (NCT) against tumor, a variety of Fenton (and Fenton-like) nanozymes, such as Fe3O4, transition metal ions (Co2+, Cu2+, and Mn2+), and metal-organic frameworks (MOFs), have been proved as desirable candidates for tumor therapy, and the modulation of the tumor microenvironment (TME) is determined to be a feasible approach to improve the catalytic efficiency for in situ tumor suppression. At present, increasing studies have focused on improving the therapeutic efficiency of NCT by formulating multifunctional nanozyme-based systems to satisfy the demand for versatile and optimized applications. Herein, updated insights into the novel strategies of 1) achieving highly effective nanocatalytic reactions, including the modification of nanocatalysts and TME-modulating approaches, are provided and 2) the design and formulation of multifunctional nanozyme-based systems which achieve targeted, synergistic therapy, and theranostic applications are analyzed and concluded. Concise and concentrated comments and outlooks are illuminated at the end to outline the perspectives and the remaining challenges for the next-step explorations on further biomedical translation of NCT.
Collapse
Affiliation(s)
- Zhen-Li Li
- Department of General Surgery, Cancer Center, Division of Hepatobiliary and Pancreatic SurgeryZhejiang Provincial People's Hospital (People's Hospital of Hangzhou Medical College)HangzhouZhejiang310014China
- School of Public HealthHangzhou Medical CollegeHangzhouZhejiang310014China
- Department of Hepatobiliary SurgeryEastern Hepatobiliary Surgery HospitalSecond Military Medical University (Naval Medical University)Shanghai200438China
- Eastern Hepatobiliary Clinical Research InstituteThird Affiliated Hospital of Naval Medical UniversityShanghai200438China
| | - Han Wu
- Department of General Surgery, Cancer Center, Division of Hepatobiliary and Pancreatic SurgeryZhejiang Provincial People's Hospital (People's Hospital of Hangzhou Medical College)HangzhouZhejiang310014China
- School of Public HealthHangzhou Medical CollegeHangzhouZhejiang310014China
- Department of Hepatobiliary SurgeryEastern Hepatobiliary Surgery HospitalSecond Military Medical University (Naval Medical University)Shanghai200438China
- Eastern Hepatobiliary Clinical Research InstituteThird Affiliated Hospital of Naval Medical UniversityShanghai200438China
| | - Jia-Qi Zhu
- College of Biotechnology and BioengineeringZhejiang University of TechnologyHangzhouZhejiang310014China
| | - Li-Yang Sun
- Department of General Surgery, Cancer Center, Division of Hepatobiliary and Pancreatic SurgeryZhejiang Provincial People's Hospital (People's Hospital of Hangzhou Medical College)HangzhouZhejiang310014China
- School of Public HealthHangzhou Medical CollegeHangzhouZhejiang310014China
| | - Xiang-Min Tong
- Department of General Surgery, Cancer Center, Division of Hepatobiliary and Pancreatic SurgeryZhejiang Provincial People's Hospital (People's Hospital of Hangzhou Medical College)HangzhouZhejiang310014China
- School of Public HealthHangzhou Medical CollegeHangzhouZhejiang310014China
| | - Dong-Sheng Huang
- Department of General Surgery, Cancer Center, Division of Hepatobiliary and Pancreatic SurgeryZhejiang Provincial People's Hospital (People's Hospital of Hangzhou Medical College)HangzhouZhejiang310014China
- School of Public HealthHangzhou Medical CollegeHangzhouZhejiang310014China
| | - Tian Yang
- Department of General Surgery, Cancer Center, Division of Hepatobiliary and Pancreatic SurgeryZhejiang Provincial People's Hospital (People's Hospital of Hangzhou Medical College)HangzhouZhejiang310014China
- School of Public HealthHangzhou Medical CollegeHangzhouZhejiang310014China
- Department of Hepatobiliary SurgeryEastern Hepatobiliary Surgery HospitalSecond Military Medical University (Naval Medical University)Shanghai200438China
- Eastern Hepatobiliary Clinical Research InstituteThird Affiliated Hospital of Naval Medical UniversityShanghai200438China
| |
Collapse
|
166
|
Oncofetal proteins and cancer stem cells. Essays Biochem 2022; 66:423-433. [PMID: 35670043 DOI: 10.1042/ebc20220025] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 05/17/2022] [Accepted: 05/20/2022] [Indexed: 12/12/2022]
Abstract
Abstract
Cancer stem cells (CSCs) are considered as a small population of cells with stem-like properties within the tumor bulk, and are largely responsible for tumor recurrence, metastasis, and therapy resistance. CSCs share critical features with embryonic stem cells (ESCs). The pluripotent transcription factors (TFs) and developmental signaling pathways of ESCs are invariably hijacked by CSCs termed ‘oncofetal drivers’ in many cancers, which are rarely detectable in adult tissues. The unique expression pattern makes oncofetal proteins ideal therapeutic targets in cancer treatment. Therefore, elucidation of oncofetal drivers in cancers is critical for the development of effective CSCs-directed therapy. In this review, we summarize the common pluripotent TFs such as OCT4, SOX2, NANOG, KLF4, MYC, SALL4, and FOXM1, as well as the development signaling including Wnt/β-catenin, Hedgehog (Hh), Hippo, Notch, and TGF-β pathways of ESCs and CSCs. We also describe the newly identified oncofetal proteins that drive the self-renewal, plasticity, and therapy-resistance of CSCs. Finally, we explore how the clinical implementation of targeting oncofetal drivers, including small-molecule inhibitors, vaccines, antibodies, and CAR-T (chimeric antigen receptor T cell) can facilitate the development of CSCs-directed therapy.
Collapse
|
167
|
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: 46] [Impact Index Per Article: 15.3] [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
|
168
|
Yamaguchi Y, Gibson J, Ou K, Lopez LS, Ng RH, Leggett N, Jonsson VD, Zarif JC, Lee PP, Wang X, Martinez C, Dorff TB, Forman SJ, Priceman SJ. PD-L1 blockade restores CAR T cell activity through IFN-γ-regulation of CD163+ M2 macrophages. J Immunother Cancer 2022; 10:e004400. [PMID: 35738799 PMCID: PMC9226933 DOI: 10.1136/jitc-2021-004400] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/08/2022] [Indexed: 11/04/2022] Open
Abstract
BACKGROUND The immune suppressive tumor microenvironment (TME) that inhibits T cell infiltration, survival, and antitumor activity has posed a major challenge for developing effective immunotherapies for solid tumors. Chimeric antigen receptor (CAR)-engineered T cell therapy has shown unprecedented clinical response in treating patients with hematological malignancies, and intense investigation is underway to achieve similar responses with solid tumors. Immunologically cold tumors, including prostate cancers, are often infiltrated with abundant tumor-associated macrophages (TAMs), and infiltration of CD163+ M2 macrophages correlates with tumor progression and poor responses to immunotherapy. However, the impact of TAMs on CAR T cell activity alone and in combination with TME immunomodulators is unclear. METHODS To model this in vitro, we utilized a novel co-culture system with tumor cells, CAR T cells, and polarized M1 or M2 macrophages from CD14+ peripheral blood mononuclear cells collected from healthy human donors. Tumor cell killing, T cell activation and proliferation, and macrophage phenotypes were evaluated by flow cytometry, cytokine production, RNA sequencing, and functional blockade of signaling pathways using antibodies and small molecule inhibitors. We also evaluated the TME in humanized mice following CAR T cell therapy for validation of our in vitro findings. RESULTS We observed inhibition of CAR T cell activity with the presence of M2 macrophages, but not M1 macrophages, coinciding with a robust induction of programmed death ligand-1 (PD-L1) in M2 macrophages. We observed similar PD-L1 expression in TAMs following CAR T cell therapy in the TME of humanized mice. PD-L1, but not programmed cell death protein-1, blockade in combination with CAR T cell therapy altered phenotypes to more M1-like subsets and led to loss of CD163+ M2 macrophages via interferon-γ signaling, resulting in improved antitumor activity of CAR T cells. CONCLUSION This study reveals an alternative mechanism by which the combination of CAR T cells and immune checkpoint blockade modulates the immune landscape of solid tumors to enhance therapeutic efficacy of CAR T cells.
Collapse
Affiliation(s)
- Yukiko Yamaguchi
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope, Duarte, California, USA
| | - Jackson Gibson
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope, Duarte, California, USA
| | - Kevin Ou
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope, Duarte, California, USA
| | - Lupita S Lopez
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope, Duarte, California, USA
| | - Rachel H Ng
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope, Duarte, California, USA
- Department of Bioengineering, University of Washington, Seattle, Washington, USA
| | - Neena Leggett
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope, Duarte, California, USA
| | - Vanessa D Jonsson
- Department of Applied Mathematics, University of California, Santa Cruz, California, USA
- Department of Biomolecular Engineering, University of California, Santa Cruz, California, USA
| | - Jelani C Zarif
- Department of Oncology, Johns Hopkins University School of Medicine and The Sidney Kimmel Comprehensive Cancer Center, Baltimore, Maryland, USA
| | - Peter P Lee
- Department of Immuno-Oncology, Beckman Research Institute of City of Hope, Duarte, California, USA
| | - Xiuli Wang
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope, Duarte, California, USA
| | - Catalina Martinez
- Department of Clinical and Translational Project Development, City of Hope, Duarte, CA, USA
| | - Tanya B Dorff
- Department of Medical Oncology and Therapeutics Research, City of Hope National Medical Center, Duarte, California, USA
| | - Stephen J Forman
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope, Duarte, California, USA
- Department of Immuno-Oncology, Beckman Research Institute of City of Hope, Duarte, California, USA
| | - Saul J Priceman
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope, Duarte, California, USA
- Department of Immuno-Oncology, Beckman Research Institute of City of Hope, Duarte, California, USA
| |
Collapse
|
169
|
Luo Z, Yao X, Li M, Fang D, Fei Y, Cheng Z, Xu Y, Zhu B. Modulating tumor physical microenvironment for fueling CAR-T cell therapy. Adv Drug Deliv Rev 2022; 185:114301. [PMID: 35439570 DOI: 10.1016/j.addr.2022.114301] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 04/07/2022] [Accepted: 04/12/2022] [Indexed: 02/06/2023]
Abstract
Chimeric antigen receptor (CAR) T cell therapy has achieved unprecedented clinical success against hematologic malignancies. However, the transition of CAR-T cell therapies for solid tumors is limited by heterogenous antigen expression, immunosuppressive microenvironment (TME), immune adaptation of tumor cells and impeded CAR-T-cell infiltration/transportation. Recent studies increasingly reveal that tumor physical microenvironment could affect various aspects of tumor biology and impose profound impacts on the antitumor efficacy of CAR-T therapy. In this review, we discuss the critical roles of four physical cues in solid tumors for regulating the immune responses of CAR-T cells, which include solid stress, interstitial fluid pressure, stiffness and microarchitecture. We highlight new strategies exploiting these features to enhance the therapeutic potency of CAR-T cells in solid tumors by correlating with the state-of-the-art technologies in this field. A perspective on the future directions for developing new CAR-T therapies for solid tumor treatment is also provided.
Collapse
|
170
|
Abstract
With the outstanding achievement of chimeric antigen receptor (CAR)-T cell therapy in the clinic, cell-based medicines have attracted considerable attention for biomedical applications and thus generated encouraging progress. As the basic construction unit of organisms, cells harbor low immunogenicity, desirable compatibility, and a strong capability of crossing various biological barriers. However, there is still a long way to go to fix significant bottlenecks for their clinical translation, such as facile preparation, strict stability requirements, scale-up manufacturing, off-target toxicity, and affordability. The rapid development of biotechnology and engineering approaches in materials sciences has provided an ideal platform to assist cell-based therapeutics for wide application in disease treatments by overcoming these issues. Herein, we survey the most recent advances of various cells as bioactive ingredients and outline the roles of biomaterials in developing cell-based therapeutics. Besides, a perspective of cell therapies is offered with a particular focus on biomaterial-involved development of cell-based biopharmaceuticals.
Collapse
Affiliation(s)
- Yu Chen
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, USA. .,Carbone Cancer Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA.,Wisconsin Center for NanoBioSystems, School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Samira Pal
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, USA.
| | - Quanyin Hu
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, USA. .,Carbone Cancer Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA.,Wisconsin Center for NanoBioSystems, School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, USA
| |
Collapse
|
171
|
Hu J, Yu X, Yin P, Du B, Cai X. Intravoxel Incoherent Motion Diffusion-Weighted MR Imaging for Monitoring the Immune Response of Immunogenic Chemotherapy. Front Oncol 2022; 12:796936. [PMID: 35646652 PMCID: PMC9136146 DOI: 10.3389/fonc.2022.796936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 04/19/2022] [Indexed: 11/13/2022] Open
Abstract
ObjectiveTo evaluate the predictive value of intravoxel incoherent motion (IVIM) diffusion-weighted imaging (DWI) in the quantitative assessment of conventional chemotherapy-activated immune responses in mouse tumor models and clinics.MethodsA total of 19 subcutaneous tumor-bearing mice were randomly divided into treated and control groups. Both groups had orderly IVIM DWI examinations before and on days 6 and 12 after the administration of cyclophosphamide (CPA) or saline. Pathologic examinations were performed, including HE staining and immunohistochemistry (IHC). The expressions of immune-related genes in the tumor were measured by qPCR. In addition, six patients with breast cancer requiring neoadjuvant chemotherapy (NACT) also underwent functional MRI examinations and IHC to determine potential antitumor immune response.ResultsAt the end of the study, the CPA treatment group showed the lowest tumor volume compared to the control group. For pathological examinations, the CPA treatment group showed a lower percentage of CD31 staining (P < 0.01) and Ki-67 staining (P<0.01), and a higher percentage of TUNEL staining (P < 0.01). The tumoral pseudodiffusion coefficient (D*) value showed a positive correlation with the CD31-positive staining rate (r = 0.729, P < 0.0001). The diffusion related parameters (D) value was positively correlated with TUNEL (r = 0.858, P < 0.0001) and negatively correlated with Ki-67 (r = -0.904, P < 0.0001). Moreover, a strong induction of the expression of the immune responses in the CPA treatment group was observed on day 12. D values showed a positive correlation with the Ifnb1-, CD8a-, Mx1-, Cxcl10- (r = 0.868, 0.864, 0.874, and 0.885, respectively, P < 0.0001 for all). Additionally, the functional MRI parameters and IHC results in patients with breast cancer after NACT also showed a close correlation between D value and CD8a (r = 0.631, P = 0.028).ConclusionsThe treatment response induced by immunogenic chemotherapy could be effectively evaluated using IVIM-DWI. The D values could be potential, sensitive imaging marker for identifying the antitumor immune response initiated by immunogenic chemotherapy.
Collapse
Affiliation(s)
- Junjiao Hu
- Medical Imaging Center, First Affiliated Hospital of Jinan University, Guangzhou, China
- Medical Imaging Center, Shenzhen Hospital, Southern Medical University, Shenzhen, China
| | - Xin Yu
- Medical Imaging Center, First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Peidi Yin
- Department of Pathology, First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Bin Du
- Department of Pathology, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, China
- *Correspondence: Xiangran Cai, ; Bin Du,
| | - Xiangran Cai
- Medical Imaging Center, First Affiliated Hospital of Jinan University, Guangzhou, China
- *Correspondence: Xiangran Cai, ; Bin Du,
| |
Collapse
|
172
|
Guedan S, Luu M, Ammar D, Barbao P, Bonini C, Bousso P, Buchholz CJ, Casucci M, De Angelis B, Donnadieu E, Espie D, Greco B, Groen R, Huppa JB, Kantari-Mimoun C, Laugel B, Mantock M, Markman JL, Morris E, Quintarelli C, Rade M, Reiche K, Rodriguez-Garcia A, Rodriguez-Madoz JR, Ruggiero E, Themeli M, Hudecek M, Marchiq I. Time 2EVOLVE: predicting efficacy of engineered T-cells - how far is the bench from the bedside? J Immunother Cancer 2022; 10:jitc-2021-003487. [PMID: 35577501 PMCID: PMC9115015 DOI: 10.1136/jitc-2021-003487] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/07/2022] [Indexed: 12/13/2022] Open
Abstract
Immunotherapy with gene engineered CAR and TCR transgenic T-cells is a transformative treatment in cancer medicine. There is a rich pipeline with target antigens and sophisticated technologies that will enable establishing this novel treatment not only in rare hematological malignancies, but also in common solid tumors. The T2EVOLVE consortium is a public private partnership directed at accelerating the preclinical development of and increasing access to engineered T-cell immunotherapies for cancer patients. A key ambition in T2EVOLVE is to assess the currently available preclinical models for evaluating safety and efficacy of engineered T cell therapy and developing new models and test parameters with higher predictive value for clinical safety and efficacy in order to improve and accelerate the selection of lead T-cell products for clinical translation. Here, we review existing and emerging preclinical models that permit assessing CAR and TCR signaling and antigen binding, the access and function of engineered T-cells to primary and metastatic tumor ligands, as well as the impact of endogenous factors such as the host immune system and microbiome. Collectively, this review article presents a perspective on an accelerated translational development path that is based on innovative standardized preclinical test systems for CAR and TCR transgenic T-cell products.
Collapse
Affiliation(s)
- Sonia Guedan
- Department of Hematology and Oncology, Hospital Clinic, IDIBAPS, Barcelona, Spain
| | - Maik Luu
- 19 Lehrstuhl für Zelluläre Immuntherapie, Medizinische Klinik und Poliklinik II, Universitätsklinikum Würzburg, Wurzburg, Germany
| | | | - Paula Barbao
- Department of Hematology and Oncology, Hospital Clinic, IDIBAPS, Barcelona, Spain
| | - Chiara Bonini
- Experimental Hematology Unit, IRCCS San Raffaele Scientific Institute, Milano, Italy
| | - Philippe Bousso
- Institut Pasteur, Université de Paris Cité, Inserm U1223, Paris, France
| | | | - Monica Casucci
- Innovative Immunotherapies Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Biagio De Angelis
- Department Onco-Haematology, and Cell and Gene Therapy, Bambino Gesù Children Hospital, IRCCS, Rome, Italy
| | - Emmanuel Donnadieu
- Université Paris Cité, CNRS, INSERM, Equipe Labellisée Ligue Contre le Cancer, Institut Cochin, F-75014 Paris, France
| | - David Espie
- Université Paris Cité, CNRS, INSERM, Equipe Labellisée Ligue Contre le Cancer, Institut Cochin, F-75014 Paris, France.,CAR-T Cells Department, Invectys, Paris, France
| | - Beatrice Greco
- Innovative Immunotherapies Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Richard Groen
- Amsterdam University Medical Centers at Vrije Universiteit, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Johannes B Huppa
- Medical University of Vienna, Center for Pathophysiology, Infectiology and Immunology, Institute for Hygiene and Applied Immunolgy, Vienna, Austria
| | | | - Bruno Laugel
- Institut de Recherches internationales Servier (IRIS), Suresnes, France
| | | | - Janet L Markman
- Takeda Development Centers Americas, Inc. Lexington, Massachusetts, USA
| | - Emma Morris
- Institute of Immunity & Transplantation, University College London Medical School - Royal Free Campus, London, UK
| | - Concetta Quintarelli
- Department Onco-Haematology, and Cell and Gene Therapy, Bambino Gesù Children Hospital, IRCCS, Rome, Italy
| | - Michael Rade
- Fraunhofer Institute for Cell Therapy and Immunology IZI, Leipzig, Germany
| | - Kristin Reiche
- Fraunhofer Institute for Cell Therapy and Immunology IZI, Leipzig, Germany
| | | | | | - Eliana Ruggiero
- Experimental Hematology Unit, IRCCS San Raffaele Scientific Institute, Milano, Italy
| | - Maria Themeli
- Amsterdam University Medical Centers at Vrije Universiteit, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Michael Hudecek
- 19 Lehrstuhl für Zelluläre Immuntherapie, Medizinische Klinik und Poliklinik II, Universitätsklinikum Würzburg, Wurzburg, Germany
| | - Ibtissam Marchiq
- Institut de Recherches internationales Servier (IRIS), Suresnes, France
| |
Collapse
|
173
|
Li Z, Ma R, Ma S, Tian L, Lu T, Zhang J, Mundy-Bosse BL, Zhang B, Marcucci G, Caligiuri MA, Yu J. ILC1s control leukemia stem cell fate and limit development of AML. Nat Immunol 2022; 23:718-730. [PMID: 35487987 PMCID: PMC9106917 DOI: 10.1038/s41590-022-01198-y] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 03/23/2022] [Indexed: 12/13/2022]
Abstract
Type I innate lymphoid cells (ILC1s) are critical regulators of inflammation and immunity in mammalian tissues. However, their function in cancer is mostly undefined. Here, we show that a high density of ILC1s induces leukemia stem cell (LSC) apoptosis in mice. At a lower density, ILC1s prevent LSCs from differentiating into leukemia progenitors and promote their differentiation into non-leukemic cells, thus blocking the production of terminal myeloid blasts. All of these effects, which require ILC1s to produce interferon-γ after cell-cell contact with LSCs, converge to suppress leukemogenesis in vivo. Conversely, the antileukemia potential of ILC1s wanes when JAK-STAT or PI3K-AKT signaling is inhibited. The relevant antileukemic properties of ILC1s are also functional in healthy individuals and impaired in individuals with acute myeloid leukemia (AML). Collectively, these findings identify ILC1s as anticancer immune cells that might be suitable for AML immunotherapy and provide a potential strategy to treat AML and prevent relapse of the disease.
Collapse
Affiliation(s)
- Zhenlong Li
- Department of Hematology & Hematopoietic Cell Transplantation, City of Hope National Medical Center, Los Angeles, CA, USA
- Hematologic Malignancies Research Institute, City of Hope National Medical Center, Los Angeles, CA, USA
| | - Rui Ma
- Department of Hematology & Hematopoietic Cell Transplantation, City of Hope National Medical Center, Los Angeles, CA, USA
- Hematologic Malignancies Research Institute, City of Hope National Medical Center, Los Angeles, CA, USA
| | - Shoubao Ma
- Department of Hematology & Hematopoietic Cell Transplantation, City of Hope National Medical Center, Los Angeles, CA, USA
- Hematologic Malignancies Research Institute, City of Hope National Medical Center, Los Angeles, CA, USA
| | - Lei Tian
- Department of Hematology & Hematopoietic Cell Transplantation, City of Hope National Medical Center, Los Angeles, CA, USA
- Hematologic Malignancies Research Institute, City of Hope National Medical Center, Los Angeles, CA, USA
| | - Ting Lu
- Department of Hematology & Hematopoietic Cell Transplantation, City of Hope National Medical Center, Los Angeles, CA, USA
- Hematologic Malignancies Research Institute, City of Hope National Medical Center, Los Angeles, CA, USA
| | - Jianying Zhang
- Department of Computational and Quantitative Medicine, City of Hope National Medical Center, Los Angeles, CA, USA
| | - Bethany L Mundy-Bosse
- Division of Hematology, Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Bin Zhang
- Gehr Family Center for Leukemia Research, Department of Hematological Malignancies Translational Science, City of Hope National Medical Center, Los Angeles, CA, USA
| | - Guido Marcucci
- Gehr Family Center for Leukemia Research, Department of Hematological Malignancies Translational Science, City of Hope National Medical Center, Los Angeles, CA, USA
| | - Michael A Caligiuri
- Department of Hematology & Hematopoietic Cell Transplantation, City of Hope National Medical Center, Los Angeles, CA, USA.
- Hematologic Malignancies Research Institute, City of Hope National Medical Center, Los Angeles, CA, USA.
- City of Hope Comprehensive Cancer Center, Los Angeles, CA, USA.
| | - Jianhua Yu
- Department of Hematology & Hematopoietic Cell Transplantation, City of Hope National Medical Center, Los Angeles, CA, USA.
- Hematologic Malignancies Research Institute, City of Hope National Medical Center, Los Angeles, CA, USA.
- City of Hope Comprehensive Cancer Center, Los Angeles, CA, USA.
- Department of Immuno-Oncology, City of Hope, Los Angeles, CA, USA.
| |
Collapse
|
174
|
Liu J, Tu X, Liu L, Fang W. Advances in CAR-T cell therapy for malignant solid tumors. Zhejiang Da Xue Xue Bao Yi Xue Ban 2022; 51:175-184. [PMID: 36161290 DOI: 10.3724/zdxbyxb-2022-0044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
T cells modified by chimeric antigen receptor (CAR) have the advantage of major histocompatibility complex-independent recognition of tumor-associated antigens, so can achieve efficient response to tumor targets. Chimeric antigen receptor (CAR) T cell therapy has shown a good therapeutic effect in hematological malignancies; however, its efficacy is generally not satisfactory for solid tumors. The reasons include the lack of tumor specific antigen target on solid tumors, the uncertainty of homing ability of engineered T cells and the inhibitory immune microenvironment of tumors. In clinical trials, the targets of CAR-T cell therapy for solid tumors are mainly disialoganglioside (GD2), claudin-18 isoform 2 (CLDN18.2), mesenchymal, B7 homolog 3 (B7H3), glypican (GPC) 3 and epidermal growth factor receptor variant Ш (EGFRvШ)Ⅲ. Combination of CAR-T cells with oncolytic viruses, tyrosine kinase inhibitors, and programmed death ligand-1 monoclonal antibodies may increase its efficacy. The CAR-T cell therapy for solid tumors can be optimized through gene editing to enhance the activity of CAR-T cells, adding corresponding regulatory components to make the activation of CAR-T cells safer and more controllable, and enhancing the persistence of CAR-T cells. In this article, we review the latest advances of CAR-T cell therapy in solid tumors to provide new insights for clinical application.
Collapse
Affiliation(s)
- Jiao Liu
- 1. Department of General Medicine, People's Hospital of Changshan County, Quzhou 324200, Zhejiang Province, China
| | - Xiaoxuan Tu
- 2. Department of Medical Oncology, the First Affiliated Hospital, Zhejiang University School of Medicine, Key Laboratory of Malignant Tumor Early Warning and Intervention of Ministry of Education, Hangzhou 310003, China
| | - Lulu Liu
- 2. Department of Medical Oncology, the First Affiliated Hospital, Zhejiang University School of Medicine, Key Laboratory of Malignant Tumor Early Warning and Intervention of Ministry of Education, Hangzhou 310003, China
| | - Weijia Fang
- 2. Department of Medical Oncology, the First Affiliated Hospital, Zhejiang University School of Medicine, Key Laboratory of Malignant Tumor Early Warning and Intervention of Ministry of Education, Hangzhou 310003, China
| |
Collapse
|
175
|
Yin J, Wu Y, Yang X, Gan L, Xue J. Checkpoint Inhibitor Pneumonitis Induced by Anti-PD-1/PD-L1 Therapy in Non-Small-Cell Lung Cancer: Occurrence and Mechanism. Front Immunol 2022; 13:830631. [PMID: 35464480 PMCID: PMC9021596 DOI: 10.3389/fimmu.2022.830631] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 03/16/2022] [Indexed: 02/05/2023] Open
Abstract
Immune checkpointty inhibitors (ICIs), particularly those targeting programmed death 1 (PD-1) and anti-programmed death ligand 1 (PD-L1), enhance the antitumor effect by restoring the function of the inhibited effector T cells and produce durable responses in a large variety of metastatic and late patients with non-small-cell lung cancer. Although often well tolerated, the activation of the immune system results in side effects known as immune-related adverse events (irAEs), which can affect multiple organ systems, including the lungs. The occurrence of severe pulmonary irAEs, especially checkpoint inhibitor pneumonitis (CIP), is rare but has extremely high mortality and often overlaps with the respiratory symptoms and imaging of primary tumors. The development of CIP may be accompanied by radiation pneumonia and infectious pneumonia, leading to the simultaneous occurrence of a mixture of several types of inflammation in the lungs. However, there is a lack of authoritative diagnosis, grading criteria and clarified mechanisms of CIP. In this article, we review the incidence and median time to onset of CIP in patients with non-small-cell lung cancer treated with PD-1/PD-L1 blockade in clinical studies. We also summarize the clinical features, potential mechanisms, management and predictive biomarkers of CIP caused by PD-1/PD-L1 blockade in non-small-cell lung cancer treatment.
Collapse
Affiliation(s)
- Jianqiong Yin
- Department of Thoracic Oncology, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Yuanjun Wu
- Department of Thoracic Oncology, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Xue Yang
- Department of Thoracic Oncology, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Lu Gan
- Research Laboratory of Emergency Medicine, Department of Emergency Medicine, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Jianxin Xue
- Department of Thoracic Oncology, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
- Department of Radiation Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Clinical Cell Therapy, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
176
|
Liu J, Hong M, Li Y, Chen D, Wu Y, Hu Y. Programmed Cell Death Tunes Tumor Immunity. Front Immunol 2022; 13:847345. [PMID: 35432318 PMCID: PMC9005769 DOI: 10.3389/fimmu.2022.847345] [Citation(s) in RCA: 164] [Impact Index Per Article: 54.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2022] [Accepted: 02/28/2022] [Indexed: 12/14/2022] Open
Abstract
The demise of cells in various ways enables the body to clear unwanted cells. Studies over the years revealed distinctive molecular mechanisms and functional consequences of several key cell death pathways. Currently, the most intensively investigated programmed cell death (PCD) includes apoptosis, necroptosis, pyroptosis, ferroptosis, PANoptosis, and autophagy, which has been discovered to play crucial roles in modulating the immunosuppressive tumor microenvironment (TME) and determining clinical outcomes of the cancer therapeutic approaches. PCD can play dual roles, either pro-tumor or anti-tumor, partly depending on the intracellular contents released during the process. PCD also regulates the enrichment of effector or regulatory immune cells, thus participating in fine-tuning the anti-tumor immunity in the TME. In this review, we focused primarily on apoptosis, necroptosis, pyroptosis, ferroptosis, PANoptosis, and autophagy, discussed the released molecular messengers participating in regulating their intricate crosstalk with the immune response in the TME, and explored the immunological consequence of PCD and its implications in future cancer therapy developments.
Collapse
Affiliation(s)
- Jing Liu
- Guangdong Provincial Key Laboratory of Tumour Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai People’s Hospital Affiliated with Jinan University, Jinan University, Zhuhai, China
- The Biomedical Translational Research Institute, Faculty of Medical Science, Jinan University, Guangzhou, China
| | - Minjing Hong
- Guangdong Provincial Key Laboratory of Tumour Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai People’s Hospital Affiliated with Jinan University, Jinan University, Zhuhai, China
| | - Yijia Li
- Guangdong Provincial Key Laboratory of Tumour Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai People’s Hospital Affiliated with Jinan University, Jinan University, Zhuhai, China
- The Biomedical Translational Research Institute, Faculty of Medical Science, Jinan University, Guangzhou, China
| | - Dan Chen
- Guangdong Provincial Key Laboratory of Tumour Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai People’s Hospital Affiliated with Jinan University, Jinan University, Zhuhai, China
| | - Yangzhe Wu
- Guangdong Provincial Key Laboratory of Tumour Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai People’s Hospital Affiliated with Jinan University, Jinan University, Zhuhai, China
| | - Yi Hu
- Microbiology and Immunology Department, School of Medicine, Faculty of Medical Science, Jinan University, Guangzhou, China
| |
Collapse
|
177
|
Heyman BM, Tzachanis D, Kipps TJ. Recent Advances in CAR T-Cell Therapy for Patients with Chronic Lymphocytic Leukemia. Cancers (Basel) 2022; 14:1715. [PMID: 35406490 PMCID: PMC8996890 DOI: 10.3390/cancers14071715] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 03/21/2022] [Accepted: 03/22/2022] [Indexed: 11/17/2022] Open
Abstract
Chimeric antigen receptor T cells (CAR T cells) have resulted in dramatic treatment responses for patients with hematologic malignancies, resulting in improved survival for patients with intractable disease. The first patient treated with CD19 directed CAR T cell therapy had chronic lymphocytic leukemia (CLL) and achieved a complete remission. Subsequent clinical trials have focused largely on patients with other B-cell hematologic malignancies, owing to the fact that CAR T cell therapy for patients with CLL has met with challenges. More recent clinical trials have demonstrated CAR T cell therapy can be well tolerated and effective for patients with CLL, making it a potential treatment option for patients with this disease. In this article we review the background on CAR T cells for the treatment of patients with CLL, focusing on the unique obstacles that patients with CLL present for the development of adoptive T cell therapy, and the novel approaches currently under development to overcome these hurdles.
Collapse
Affiliation(s)
- Benjamin M. Heyman
- Division of Regenerative Medicine, Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Dimitrios Tzachanis
- Division of Blood and Marrow Transplantation, Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA;
| | - Thomas J. Kipps
- Center for Novel Therapeutics, Department of Medicine, University of California San Diego, La Jolla, CA 92037, USA;
| |
Collapse
|
178
|
Miao L, Zhang J, Zhang Z, Wang S, Tang F, Teng M, Li Y. A Bibliometric and Knowledge-Map Analysis of CAR-T Cells From 2009 to 2021. Front Immunol 2022; 13:840956. [PMID: 35371087 PMCID: PMC8971369 DOI: 10.3389/fimmu.2022.840956] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 03/01/2022] [Indexed: 12/15/2022] Open
Abstract
ObjectivesA bibliometric and knowledge-map analysis is used to explore hotspots’ evolution and development trends in the CAR-T cell field. By looking for research hotspots and new topics, we can provide new clues and ideas for researchers in this field.MethodsThe articles and reviews regarding CAR-T cells were retrieved and obtained from the Web of Science Core Collection (WOSCC) on October 28th, 2021. CtieSpace [version 5.8.R3 (64-bit)] and VOSviewer (version 1.6.17) were used to conduct the bibliometric and knowledge-map analysis.Results660 authors from 488 institutions in 104 countries/regions published 6,867 papers in 1,212 academic journals. The United States was absolutely in the leading position in this research field. The institution that contributed the most publications was the University of Pennsylvania. Carl H June published the most articles, while Shannon L Maude had the most co-citations. However, there was little cooperation between countries. After 2012, cooperation among various institutions was also small. The journals that published the most CAR-T cell-related papers were Frontiers in immunology and Cancers. Nevertheless, Blood and The New England Journal of Medicine were the most commonly co-cited journals. The most influential research hotspots were the research of CAR-T cells in hematological malignancies, the related research of cytokine release syndrome (CRS), CD19, and the anti-tumor activity and efficacy of CAR-T cells. The latest hotspots and topics included the study of CAR-T cells in solid tumors, universal CAR-T cells, CAR-NK cells, CD22, and anakinra (the IL-1 receptor antagonist). The research of CAR-T cells in solid tumors was a rapidly developing hot field. Emerging topics in this field mainly included the study of CAR-T cells in glioblastoma (related targets: IL13Rα2, EGFRvIII, and HER2), neuroblastoma (related target: GD2), sarcoma (related target: HER2), and pancreatic cancer (related target: mesothelin), especially glioblastoma.ConclusionAs an anti-tumor therapy with great potential and clinical application prospects, CAR-T cell therapy is still in a stage of rapid development. The related field of CAR-T cells will remain a research hotspot in the future.
Collapse
Affiliation(s)
- Lele Miao
- Department of General Surgery, Second Hospital of Lanzhou University, Lanzhou, China
- Key Laboratory of the Digestive System Tumors of Gansu Province, Lanzhou, China
| | - Juan Zhang
- Department of Hematology, Fifth Medical Center, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Zhengchao Zhang
- Department of General Surgery, Second Hospital of Lanzhou University, Lanzhou, China
- Key Laboratory of the Digestive System Tumors of Gansu Province, Lanzhou, China
| | - Song Wang
- Department of General Surgery, Second Hospital of Lanzhou University, Lanzhou, China
- Key Laboratory of the Digestive System Tumors of Gansu Province, Lanzhou, China
| | - Futian Tang
- Key Laboratory of the Digestive System Tumors of Gansu Province, Lanzhou, China
| | - Muzhou Teng
- Key Laboratory of the Digestive System Tumors of Gansu Province, Lanzhou, China
- Lanzhou University, Lanzhou, China
- *Correspondence: Yumin Li, ; Muzhou Teng,
| | - Yumin Li
- Department of General Surgery, Second Hospital of Lanzhou University, Lanzhou, China
- Key Laboratory of the Digestive System Tumors of Gansu Province, Lanzhou, China
- Lanzhou University, Lanzhou, China
- *Correspondence: Yumin Li, ; Muzhou Teng,
| |
Collapse
|
179
|
Chen L, Chen F, Li J, Pu Y, Yang C, Wang Y, Lei Y, Huang Y. CAR-T cell therapy for lung cancer: Potential and perspective. Thorac Cancer 2022; 13:889-899. [PMID: 35289077 PMCID: PMC8977151 DOI: 10.1111/1759-7714.14375] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 02/18/2022] [Accepted: 02/20/2022] [Indexed: 12/24/2022] Open
Abstract
Lung cancer is the highest incidence and mortality of all cancers around the world. In the present immunotherapy era, an increasing number of immunotherapeutic agents including monoclonal antibody‐targeted drugs have been used in the clinical treatment of malignancy, but it still has many limitations. Chimeric antigen receptor‐modified T (CAR‐T) cells, a novel adoptive immunotherapy strategy, have not only been used successfully against hematological tumors, but have also opened up new avenues for immunotherapy of solid tumors, including lung cancer. However, targeting lung cancer‐specific antigens using engineered CAR‐T cells is complicated by the lack of proper tumor‐specific antigens, an immunosuppressive tumor microenvironment, a low level of CAR‐T cell infiltration into tumor tissues, along with off‐target effect, etc. Simultaneously, the clinical application of CAR‐T cells remains limited because of many challenges such as tumor lysis syndrome, neurotoxicity syndrome, and cytokine release syndrome. In this review, we outline the basic structure and generation characteristic of CAR‐T cells and summarize the common tumor‐associated antigens in clinical trials of CAR‐T cell therapy for lung cancer, and point out the current challenges and new strategies, aiming to provide new ideas and approaches for the pre‐clinical experiments and clinical trials of CAR‐T cell therapy in lung cancer.
Collapse
Affiliation(s)
- Long Chen
- Department of PET/CT Center, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Cancer Center of Yunnan Province, Kunming, China
| | - Fukun Chen
- Department of Nuclear Medicine, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Cancer Center of Yunnan Province, Kunming, China
| | - Jindan Li
- Department of PET/CT Center, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Cancer Center of Yunnan Province, Kunming, China
| | - Yongzhu Pu
- Department of PET/CT Center, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Cancer Center of Yunnan Province, Kunming, China
| | - Conghui Yang
- Department of PET/CT Center, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Cancer Center of Yunnan Province, Kunming, China
| | - Yue Wang
- Department of PET/CT Center, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Cancer Center of Yunnan Province, Kunming, China
| | - Yujie Lei
- Department of Thoracic Surgery I, Key Laboratory of Lung Cancer of Yunnan Province, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Cancer Center of Yunnan Province, Kunming, China
| | - Yunchao Huang
- Department of Thoracic Surgery I, Key Laboratory of Lung Cancer of Yunnan Province, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Cancer Center of Yunnan Province, Kunming, China
| |
Collapse
|
180
|
Dong J, Huang Y, Zhou Z, Sun M. Breaking Immunosuppressive Barriers by Engineered Nanoplatforms for Turning Cold Tumor to Hot. ADVANCED THERAPEUTICS 2022. [DOI: 10.1002/adtp.202200020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Affiliation(s)
- Jingwen Dong
- State Key Laboratory of Natural Medicines Department of Pharmaceutics China Pharmaceutical University Nanjing 210009 P. R. China
| | - Ying Huang
- State Key Laboratory of Natural Medicines Department of Pharmaceutics China Pharmaceutical University Nanjing 210009 P. R. China
| | - Zhanwei Zhou
- State Key Laboratory of Natural Medicines Department of Pharmaceutics China Pharmaceutical University Nanjing 210009 P. R. China
| | - Minjie Sun
- State Key Laboratory of Natural Medicines Department of Pharmaceutics China Pharmaceutical University Nanjing 210009 P. R. China
| |
Collapse
|
181
|
Nguyen A, Johanning G, Shi Y. Emerging Novel Combined CAR-T Cell Therapies. Cancers (Basel) 2022; 14:cancers14061403. [PMID: 35326556 PMCID: PMC8945996 DOI: 10.3390/cancers14061403] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 03/03/2022] [Accepted: 03/07/2022] [Indexed: 12/08/2022] Open
Abstract
Simple Summary As a result of FDA approval of CAR-T cell treatments in the last few years, this immunotherapy has provided further direction to precision medicine through its combination with other therapeutic approaches. In the past year, several review articles have been published focusing on advances in this fast-developing field, especially with respect to efforts to overcome hurdles associated with applying CAR-T cells in solid tumors. This review paper focuses on combining CAR-T cell therapy with small molecule drugs, up-to-date progress in CAR-T cell therapy research, and advances in combined CAR-T immunotherapy with other treatments targeting solid tumors. Abstract Chimeric antigen receptors (CAR) T cells are T cells engineered to express membrane receptors with high specificity to recognize specific target antigens presented by cancer cells and are co-stimulated with intracellular signals to increase the T cell response. CAR-T cell therapy is emerging as a novel therapeutic approach to improve T cell specificity that will lead to advances in precision medicine. CAR-T cells have had impressive outcomes in hematological malignancies. However, there continue to be significant limitations of these therapeutic responses in targeting solid malignancies such as heterogeneous antigens in solid tumors, tumor immunosuppressive microenvironment, risk of on-target/off-tumor, infiltrating CAR-T cells, immunosuppressive checkpoint molecules, and cytokines. This review paper summarizes recent approaches and innovations through combination therapies of CAR-T cells and other immunotherapy or small molecule drugs to counter the above disadvantages to potentiate the activity of CAR-T cells.
Collapse
Affiliation(s)
- Anh Nguyen
- College of Graduate Studies, California Northstate University, Elk Grove, CA 95757, USA;
| | | | - Yihui Shi
- College of Medicine, California Northstate University, Elk Grove, CA 95757, USA
- Correspondence:
| |
Collapse
|
182
|
Villa NY, Rahman MM, Mamola J, Sharik ME, de Matos AL, Kilbourne J, Lowe K, Daggett-Vondras J, D'Isabella J, Goras E, Chesi M, Bergsagel PL, McFadden G. Transplantation of autologous bone marrow pre-loaded ex vivo with oncolytic myxoma virus is efficacious against drug-resistant Vk*MYC mouse myeloma. Oncotarget 2022; 13:490-504. [PMID: 35251496 PMCID: PMC8893797 DOI: 10.18632/oncotarget.28205] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Accepted: 02/14/2022] [Indexed: 11/25/2022] Open
Abstract
Multiple myeloma (MM) is a hematological malignancy of plasma cells that remains incurable despite significant progress with myeloablative regimens and autologous stem cell transplantation for eligible patients and, more recently with T cell redirected immunotherapy. Recently, we reported that ex vivo virotherapy with oncolytic myxoma virus (MYXV) improved MM-free survival in an autologous-transplant Balb/c mouse model. Here, we tested the Vk*MYC transplantable C57BL/6 mouse MM model that more closely recapitulates human disease. In vitro, the murine bortezomib-resistant Vk12598 cell line is fully susceptible to MYXV infection. In vivo results demonstrate: (i) autologous bone marrow (BM) leukocytes armed ex vivo with MYXV exhibit moderate therapeutic effects against MM cells pre-seeded into recipient mice; (ii) Cyclophosphamide in combination with BM/MYXV delays the onset of myeloma in mice seeded with Vk12598 cells; (iii) BM/MYXV synergizes with the Smac-mimetics LCL161 and with immune checkpoint inhibitor α-PD-1 to control the progression of established MM in vivo, resulting in significant improvement of survival rates and decreased of tumor burden; (iv) Survivor mice from (ii) and (iii), when re-challenged with fresh Vk12598 cells, developed acquired anti-MM immunity. These results highlight the utility of autologous BM grafts armed ex vivo with oncolytic MYXV alone or in combination with chemotherapy/immunotherapy to treat drug-resistant MM in vivo.
Collapse
Affiliation(s)
- Nancy Y. Villa
- Biodesign Institute, Center for Immunotherapy, Vaccines and Virotherapy (CIVV), Arizona State University, Tempe, AZ 85281, USA
- Division of Hematology/Oncology, School of Medicine, Emory University, Atlanta, GA 32322, USA
| | - Masmudur M. Rahman
- Biodesign Institute, Center for Immunotherapy, Vaccines and Virotherapy (CIVV), Arizona State University, Tempe, AZ 85281, USA
| | - Joseph Mamola
- Biodesign Institute, Center for Immunotherapy, Vaccines and Virotherapy (CIVV), Arizona State University, Tempe, AZ 85281, USA
| | | | - Ana Lemos de Matos
- Biodesign Institute, Center for Immunotherapy, Vaccines and Virotherapy (CIVV), Arizona State University, Tempe, AZ 85281, USA
| | - Jacquelyn Kilbourne
- Biodesign Institute, Center for Immunotherapy, Vaccines and Virotherapy (CIVV), Arizona State University, Tempe, AZ 85281, USA
| | - Kenneth Lowe
- Biodesign Institute, Center for Immunotherapy, Vaccines and Virotherapy (CIVV), Arizona State University, Tempe, AZ 85281, USA
| | - Juliane Daggett-Vondras
- Biodesign Institute, Center for Immunotherapy, Vaccines and Virotherapy (CIVV), Arizona State University, Tempe, AZ 85281, USA
| | - Julia D'Isabella
- Biodesign Institute, Center for Immunotherapy, Vaccines and Virotherapy (CIVV), Arizona State University, Tempe, AZ 85281, USA
| | - Elizabeth Goras
- Biodesign Institute, Center for Immunotherapy, Vaccines and Virotherapy (CIVV), Arizona State University, Tempe, AZ 85281, USA
| | - Marta Chesi
- Department of Medicine, Mayo Clinic, Scottsdale, AZ 85259, USA
| | | | - Grant McFadden
- Biodesign Institute, Center for Immunotherapy, Vaccines and Virotherapy (CIVV), Arizona State University, Tempe, AZ 85281, USA
| |
Collapse
|
183
|
Huang X, Zhang G, Tang T, Gao X, Liang T. One shoot, three birds: Targeting NEK2 orchestrates chemoradiotherapy, targeted therapy, and immunotherapy in cancer treatment. Biochim Biophys Acta Rev Cancer 2022; 1877:188696. [PMID: 35157980 DOI: 10.1016/j.bbcan.2022.188696] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 02/02/2022] [Accepted: 02/04/2022] [Indexed: 12/16/2022]
Abstract
Combinational therapy has improved the cancer therapeutic landscape but is associated with a concomitant increase in adverse side reactions. Emerging evidence proposes that targeting one core target with multiple critical roles in tumors can achieve combined anti-tumor effects. This review focuses on NEK2, a member of serine/threonine kinases, with broad sequence identity to the mitotic regulator NIMA of the filamentous fungus Aspergillus nidulans. Elevated expression of NEK2 was initially found to promote tumorigeneses through abnormal regulation of the cell cycle. Subsequent studies report that NEK2 is overexpressed in a broad spectrum of tumor types and is associated with tumor progression and therapeutic resistance. Intriguingly, NEK2 has recently been revealed to mediate tumor immune escape by stabilizing the expression of PD-L1. Targeting NEK2 is thus becoming a promising approach for cancer treatment by orchestrating chemoradiotherapy, targeted therapy, and immunotherapy. It represents a novel strategy for inducing combined anti-cancer effects using a mono-agent.
Collapse
Affiliation(s)
- Xing Huang
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, Zhejiang, China; Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang, China; Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic Diseases, Hangzhou 310003, Zhejiang, China; The Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province, Hangzhou 310009, Zhejiang, China; Cancer Center, Zhejiang University, Hangzhou 310058, Zhejiang, China.
| | - Gang Zhang
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, Zhejiang, China; Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang, China; Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic Diseases, Hangzhou 310003, Zhejiang, China; The Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province, Hangzhou 310009, Zhejiang, China; Cancer Center, Zhejiang University, Hangzhou 310058, Zhejiang, China
| | - Tianyu Tang
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, Zhejiang, China; Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang, China; Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic Diseases, Hangzhou 310003, Zhejiang, China; The Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province, Hangzhou 310009, Zhejiang, China; Cancer Center, Zhejiang University, Hangzhou 310058, Zhejiang, China
| | - Xiang Gao
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, Zhejiang, China; Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang, China; Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic Diseases, Hangzhou 310003, Zhejiang, China; The Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province, Hangzhou 310009, Zhejiang, China; Cancer Center, Zhejiang University, Hangzhou 310058, Zhejiang, China
| | - Tingbo Liang
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, Zhejiang, China; Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang, China; Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic Diseases, Hangzhou 310003, Zhejiang, China; The Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province, Hangzhou 310009, Zhejiang, China; Cancer Center, Zhejiang University, Hangzhou 310058, Zhejiang, China.
| |
Collapse
|
184
|
Liang Y, Lei Y, Liang M, Du M, Liu Z, Li X, Meng X, Zhou B, Gao Y. GBE1 Is an Independent Prognostic Marker and Associated With CD163 + Tumor-Associated Macrophage Infiltration in Lung Adenocarcinoma. Front Oncol 2022; 11:781344. [PMID: 35155189 PMCID: PMC8828580 DOI: 10.3389/fonc.2021.781344] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Accepted: 12/20/2021] [Indexed: 12/18/2022] Open
Abstract
Glycogen branching enzyme (GBE1) is a critical gene that participates in regulating glycogen metabolism. However, the correlations between GBE1 expression and the prognosis and tumor-associated macrophages in lung adenocarcinoma (LUAD) also remain unclear. Herein, we firstly analyzed the expression level of GBE1 in LUAD tissues and adjacent lung tissues via The Cancer Genome Atlas (TCGA) database. The effect of GBE1 on prognosis was estimated by utilizing TCGA database and the PrognoScan database. The relationships between the clinical characteristics and GBE1 expression were evaluated via TCGA database. We then investigated the relationships between GBE1 and infiltration of immune cells in LUAD by utilizing the CIBERSORT algorithm and Tumor Immune Estimation Resource (TIMER) database. In addition, we used a tissue microarray (TMA) containing 92 LUAD tissues and 88 adjacent lung tissues with immunohistochemistry staining to verify the association between GBE1 expression and clinical characteristics, as well as the immune cell infiltrations. We found the expression level of GBE1 was significantly higher in LUAD tissues. High expression of GBE1 was associated with poorer overall survival (OS) in LUAD. In addition, high expression of GBE1 was correlated with advanced T classification, N classification, M classification, TNM stage, and lower grade. Moreover, GBE1 was positively correlated with infiltrating levels of CD163+ tumor-associated macrophages in LUAD. In conclusion, the expression of GBE1 is associated with the prognosis and CD163+ tumor-associated macrophage infiltration in LUAD, suggesting that it has potential to be prognostic and immunological biomarkers in LUAD.
Collapse
Affiliation(s)
- Yicheng Liang
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yangyang Lei
- Department of Interventional Radiology, Shanghai Institute of Medical Imaging, Shanghai, China.,Department of Interventional Radiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Mei Liang
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Minjun Du
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zixu Liu
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xingkai Li
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiangzhi Meng
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Boxuan Zhou
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yushun Gao
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
185
|
Andrea AE, Chiron A, Mallah S, Bessoles S, Sarrabayrouse G, Hacein-Bey-Abina S. Advances in CAR-T Cell Genetic Engineering Strategies to Overcome Hurdles in Solid Tumors Treatment. Front Immunol 2022; 13:830292. [PMID: 35211124 PMCID: PMC8861853 DOI: 10.3389/fimmu.2022.830292] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 01/18/2022] [Indexed: 12/15/2022] Open
Abstract
During this last decade, adoptive transfer of T lymphocytes genetically modified to express chimeric antigen receptors (CARs) emerged as a valuable therapeutic strategy in hematological cancers. However, this immunotherapy has demonstrated limited efficacy in solid tumors. The main obstacle encountered by CAR-T cells in solid malignancies is the immunosuppressive tumor microenvironment (TME). The TME impedes tumor trafficking and penetration of T lymphocytes and installs an immunosuppressive milieu by producing suppressive soluble factors and by overexpressing negative immune checkpoints. In order to overcome these hurdles, new CAR-T cells engineering strategies were designed, to potentiate tumor recognition and infiltration and anti-cancer activity in the hostile TME. In this review, we provide an overview of the major mechanisms used by tumor cells to evade immune defenses and we critically expose the most optimistic engineering strategies to make CAR-T cell therapy a solid option for solid tumors.
Collapse
Affiliation(s)
- Alain E. Andrea
- Laboratoire de Biochimie et Thérapies Moléculaires, Faculté de Pharmacie, Université Saint Joseph de Beyrouth, Beirut, Lebanon
| | - Andrada Chiron
- Université de Paris, CNRS, INSERM, UTCBS, Unité des technologies Chimiques et Biologiques pour la Santé, Paris, France
- Clinical Immunology Laboratory, Groupe Hospitalier Universitaire Paris-Sud, Hôpital Kremlin-Bicêtre, Assistance Publique-Hôpitaux de Paris, Le-Kremlin-Bicêtre, France
| | - Sarah Mallah
- Faculty of Arts and Sciences, Lebanese American University, Beirut, Lebanon
| | - Stéphanie Bessoles
- Université de Paris, CNRS, INSERM, UTCBS, Unité des technologies Chimiques et Biologiques pour la Santé, Paris, France
| | - Guillaume Sarrabayrouse
- Université de Paris, CNRS, INSERM, UTCBS, Unité des technologies Chimiques et Biologiques pour la Santé, Paris, France
| | - Salima Hacein-Bey-Abina
- Université de Paris, CNRS, INSERM, UTCBS, Unité des technologies Chimiques et Biologiques pour la Santé, Paris, France
- Clinical Immunology Laboratory, Groupe Hospitalier Universitaire Paris-Sud, Hôpital Kremlin-Bicêtre, Assistance Publique-Hôpitaux de Paris, Le-Kremlin-Bicêtre, France
| |
Collapse
|
186
|
Zhao J, Ye H, Lu Q, Wang K, Chen X, Song J, Wang H, Lu Y, Cheng M, He Z, Zhai Y, Zhang H, Sun J. Inhibition of post-surgery tumour recurrence via a sprayable chemo-immunotherapy gel releasing PD-L1 antibody and platelet-derived small EVs. J Nanobiotechnology 2022; 20:62. [PMID: 35109878 PMCID: PMC8812025 DOI: 10.1186/s12951-022-01270-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Accepted: 01/17/2022] [Indexed: 12/28/2022] Open
Abstract
Background Melanoma is the most serious type of skin cancer, and surgery is an effective method to treat melanoma. Unfortunately, local residual micro-infiltrated tumour cells and systemic circulating tumour cells (CTCs) are significant causes of treatment failure, leading to tumour recurrence and metastasis. Methods Small EVs were isolated from platelets by differential centrifugation, and doxorubicin-loaded small EVs (PexD) was prepared by mixing small EVs with doxorubicin (DOX). PexD and an anti-PD-L1 monoclonal antibody (aPD-L1) were co-encapsulated in fibrin gel. The synergistic antitumour efficacy of the gel containing PexD and aPD-L1 was assessed both in vitro and in vivo. Results Herein, we developed an in situ-formed bioresponsive gel combined with chemoimmunotherapeutic agents as a drug reservoir that could effectively inhibit both local tumour recurrence and tumour metastasis. In comparison with a DOX solution, PexD could better bind to tumour cells, induce more tumour immunogenic cell death (ICD) and promote a stronger antitumour immune response. PexD could enter the blood circulation through damaged blood vessels to track and eliminate CTCs. The concurrent release of aPD-L1 at the tumour site could impair the PD-1/PD-L1 pathway and restore the tumour-killing effect of cytotoxic T cells. This chemoimmunotherapeutic strategy triggered relatively strong T cell immune responses, significantly improving the tumour immune microenvironment. Conclusion Our findings indicated that the immunotherapeutic fibrin gel could “awaken” the host innate immune system to inhibit both local tumour recurrence post-surgery and metastatic potential, thus, it could serve as a promising approach to prevent tumour recurrence. Graphical Abstract ![]()
Supplementary Information The online version contains supplementary material available at 10.1186/s12951-022-01270-7.
Collapse
Affiliation(s)
- Jian Zhao
- College of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, 110016, Liaoning, People's Republic of China
| | - Hao Ye
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, 110016, Liaoning, People's Republic of China.,Multi-Scale Robotics Lab (MSRL), Institute of Robotics & Intelligent Systems (IRIS), ETH Zurich, 8092, Zurich, Switzerland
| | - Qi Lu
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, 110016, Liaoning, People's Republic of China
| | - Kaiyuan Wang
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, 110016, Liaoning, People's Republic of China
| | - Xiaofeng Chen
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, 110016, Liaoning, People's Republic of China
| | - Jiaxuan Song
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, 110016, Liaoning, People's Republic of China
| | - Helin Wang
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, 110016, Liaoning, People's Republic of China
| | - Yutong Lu
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, 110016, Liaoning, People's Republic of China
| | - Maosheng Cheng
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Zhonggui He
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, 110016, Liaoning, People's Republic of China
| | - Yinglei Zhai
- Department of Biomedical Engineering, School of Medical Devices, Shenyang Pharmaceutical University, Shenyang, 110016, Liaoning, China
| | - Haotian Zhang
- School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, 110016, Liaoning, People's Republic of China
| | - Jin Sun
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, 110016, Liaoning, People's Republic of China.
| |
Collapse
|
187
|
Ma J, Zhang C, Shi G, Yue D, Shu Y, Hu S, Qi Z, Chen Y, Zhang B, Zhang Y, Huang A, Su C, Zhang Y, Deng H, Cheng P. High-dose VitC plus oncolytic adenoviruses enhance immunogenic tumor cell death and reprogram tumor immune microenvironment. Mol Ther 2022; 30:644-661. [PMID: 34547462 PMCID: PMC8821933 DOI: 10.1016/j.ymthe.2021.09.015] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 08/25/2021] [Accepted: 09/14/2021] [Indexed: 02/08/2023] Open
Abstract
Preclinical and clinical studies have validated the antitumor effects of several oncolytic viruses (OVs). However, the efficacy of OVs is limited when they are administered as monotherapies. Combination therapy is a promising direction for oncolytic virotherapy in the future. A high dose of vitamin C (VitC) exerts anticancer effects by triggering the accretion of substantial amounts of reactive oxygen species (ROS). OVs can induce immunogenic tumor cell death and elicit an antitumor immune response. ROS play an important role in immunogenic cell death (ICD). This study aimed to explore whether high-dose VitC in combination with oncolytic adenoviruses (oAds) exhibited a synergistic antitumor effect. High-dose VitC synergized with oAds against tumor by enhancing immunogenic tumor cell death. Combination therapy with high-dose VitC and oAds significantly increased the number of T cells in the tumor microenvironment (TME) and promoted the activation of T cells. Furthermore, the antitumor effect of the combination therapy was CD8+ T cell dependent. In addition, combination therapy with high-dose VitC and oAds reprogramed the immunosuppressive TME. Our study provides a new strategy for combination therapy of OVs.
Collapse
Affiliation(s)
- Jinhu Ma
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, 17 People’s South Road, Chengdu 610041, PR China
| | - Chunxue Zhang
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, 17 People’s South Road, Chengdu 610041, PR China
| | - Gang Shi
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, 17 People’s South Road, Chengdu 610041, PR China
| | - Dan Yue
- School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, PR China
| | - Yongheng Shu
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, 17 People’s South Road, Chengdu 610041, PR China
| | - Shichuan Hu
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, 17 People’s South Road, Chengdu 610041, PR China
| | - Zhongbing Qi
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, 17 People’s South Road, Chengdu 610041, PR China
| | - Yanwei Chen
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, 17 People’s South Road, Chengdu 610041, PR China
| | - Bin Zhang
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, 17 People’s South Road, Chengdu 610041, PR China
| | - Yong Zhang
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, 17 People’s South Road, Chengdu 610041, PR China
| | - Anliang Huang
- Department of Pathology, Chengdu Fifth People’s Hospital, Chengdu, PR China
| | - Chao Su
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, 17 People’s South Road, Chengdu 610041, PR China
| | - Yan Zhang
- Department of Thoracic Oncology, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Hongxin Deng
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, 17 People’s South Road, Chengdu 610041, PR China
| | - Ping Cheng
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, 17 People’s South Road, Chengdu 610041, PR China,Corresponding author: Prof. Ping Cheng, State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, 17 People’s South Road, Chengdu 610041, PR China.
| |
Collapse
|
188
|
Paterson K, Paterson S, Mulholland T, Coffelt S, Zagnoni M. Assessment of CAR-T cell-mediated cytotoxicity in 3D microfluidic cancer co-culture models for combination therapy. IEEE OPEN JOURNAL OF ENGINEERING IN MEDICINE AND BIOLOGY 2022; 3:86-95. [PMID: 35813488 PMCID: PMC9252335 DOI: 10.1109/ojemb.2022.3178302] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 05/16/2022] [Accepted: 05/23/2022] [Indexed: 12/01/2022] Open
Abstract
Chimeric antigen receptor (CAR)-T cell therapy is efficacious against many haematological malignancies, but challenges remain when using this cellular immunotherapy for treating solid tumours. Classical 2D in vitro models fail to recapitulate the complexity of the tumour microenvironment, whilst in vivo models, such as patient-derived xenografts, are costly and labour intensive. Microfluidic technologies can provide miniaturized solutions to assess CAR-T therapies in 3D complex preclinical models of solid tumours. Here, we present a novel microfluidic immunoassay for the evaluation of CAR-T cell cytotoxicity and targeting specificity on 3D spheroids containing cancer cells and stromal cells. Monitoring the interaction between CAR-T cells and spheroid co-cultures, we show that CAR-T cells home towards target-expressing cancer cells and elicit a cytotoxic effect. Testing CAR-T cells in combination therapies, we show that CAR-T cell cytotoxicity is enhanced with anti-PD-L1 therapy and carboplatin chemotherapy. We propose this proof-of-concept microfluidic immunoassay as a material-saving, pre-clinical screening tool for quantification of cell therapy efficacy.
Collapse
Affiliation(s)
- Karla Paterson
- Centre for Microsystems and Photonics, EEE Department, University of Strathclyde, Glasgow, UK
| | - Sarah Paterson
- ScreenIn3D Limited, Technology and Innovation Centre, Glasgow, UK
| | | | - Seth Coffelt
- Cancer Research UK Beatson Institute, Glasgow, UK
| | - Michele Zagnoni
- EEE, Univ Strathclyde, Glasgow, United Kingdom of Great Britain and Northern Ireland, G1 1XW
| |
Collapse
|
189
|
Zhang K, Wu J, Zhao X, Qin J, Xue Y, Zheng W, Wang L, Wang H, Shen H, Niu T, Luo Y, Tang R, Wang B. Prussian Blue/Calcium Peroxide Nanocomposites-Mediated Tumor Cell Iron Mineralization for Treatment of Experimental Lung Adenocarcinoma. ACS NANO 2021; 15:19838-19852. [PMID: 34851083 DOI: 10.1021/acsnano.1c07308] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Current lung cancer diagnosis methods encounter delayed visual confirmation of tumor foci and low-resolution metrics in imaging findings, which delays the early treatment of tumors. Here, we developed a potent lung cancer imaging and treatment strategy centered around a nanotransformational concept of tumor iron mineralization in situ, which employs Prussian blue/calcium peroxide nanocomposites as a precursor. The resultant iron mineralization in tumor cells greatly facilitates the early and differential diagnosis of lung carcinoma from benign nodules via medical imaging, meanwhile introducing oxidative stress to activate the cellular apoptosis and ferroptosis pathways, resulting in inhibition of the malignant behavior of tumor cells. Tumor-microenvironment-triggered iron mineralization enables integration of the detection and prevention of tumor metastasis at its early stages with no assistance of toxic drugs, which offers a potential solution for the precise management of lung cancer with ideal outcomes.
Collapse
Affiliation(s)
- Kaixin Zhang
- Institute of Translational Medicine, Zhejiang University, Hangzhou 310029, China
| | - Jicheng Wu
- Institute of Translational Medicine, Zhejiang University, Hangzhou 310029, China
| | - Xiaoxiong Zhao
- Institute of Translational Medicine, Zhejiang University, Hangzhou 310029, China
| | | | - Yi Xue
- Institute of Translational Medicine, Zhejiang University, Hangzhou 310029, China
| | | | | | - Haoran Wang
- Institute of Translational Medicine, Zhejiang University, Hangzhou 310029, China
| | | | - Tianye Niu
- Nuclear & Radiological Engineering and Medical Physics Programs, Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332, United States
| | - Yan Luo
- Department of Biochemistry, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Ruikang Tang
- Department of Chemistry, Zhejiang University, Hangzhou 310027, China
| | - Ben Wang
- Institute of Translational Medicine, Zhejiang University, Hangzhou 310029, China
| |
Collapse
|
190
|
Safarzadeh Kozani P, Safarzadeh Kozani P, Rahbarizadeh F. Addressing the obstacles of CAR T cell migration in solid tumors: wishing a heavy traffic. Crit Rev Biotechnol 2021; 42:1079-1098. [PMID: 34957875 DOI: 10.1080/07388551.2021.1988509] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Chimeric antigen receptor T cell (CAR-T) therapy has been recognized as one of the most prosperous treatment options against certain blood-based malignancies. However, the same clinical and commercial success have been out of range in the case of solid tumors. The main contributing factor in this regard is the hostile environment the tumor cells impose that results in the exhaustion of immune effector cells alongside the abrogation of their infiltration capacity. The discovery of the underlying mechanisms and the development of reliable counterstrategies to overcome the inaccessibility of CAR-Ts to their target cells might correlate with encouraging clinical outcomes in advanced solid tumors. Here, we highlight the successive physical and metabolic barriers that systemically administered CAR-Ts face on their journey toward their target cells. Moreover, we propose meticulously-devised countertactics and combination therapies that can be applied to maximize the therapeutic benefits of CAR-T therapies against solid tumors.
Collapse
Affiliation(s)
- Pooria Safarzadeh Kozani
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Pouya Safarzadeh Kozani
- Department of Medical Biotechnology, Faculty of Paramedicine, Guilan University of Medical Sciences, Rasht, Iran.,Student Research Committee, Medical Biotechnology Research Center, School of Nursing, Midwifery, and Paramedicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Fatemeh Rahbarizadeh
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran.,Research and Development Center of Biotechnology, Tarbiat Modares University, Tehran, Iran
| |
Collapse
|
191
|
Leruste A, Beccaria K, Doz F. CAR-T cells for pediatric brain tumors: Present and future. Bull Cancer 2021; 108:S109-S116. [PMID: 34920793 DOI: 10.1016/j.bulcan.2021.06.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 06/02/2021] [Accepted: 06/07/2021] [Indexed: 10/19/2022]
Abstract
Chimeric Antigen Receptor T (CAR-T) cells are currently approved for B cell malignancies only, in children and adults. Despite a lack of robust evidence to approve such cellular immunotherapy for pediatric solid tumors, there is a growing interest for this approach in the treatment of pediatric brain tumors. Following the identification of tumor antigens as targets, the first clinical trials demonstrated some degree of clinical and biological responses to CAR-T cells for such tumor types. Additionaly, several preclinical studies have recently identified new attractive targets and antigen combination strategies, along with a superior tumor trafficking following locoregional administration. We review here the preclinical and clinical knowledge at the basis of the current clinical development of CAR-T cells for pediatric brain tumors.
Collapse
Affiliation(s)
- Amaury Leruste
- PSL Research University, SIREDO Oncology Center (Care, Innovation and Research for Children and AYA with Cancer), Institut Curie, Paris, France.
| | - Kevin Beccaria
- Université de Paris, AP-HP, Necker Hospital, Department of Pediatric Neurosurgery, 149, rue de Sèvres, 75015 Paris, France
| | - François Doz
- PSL Research University, SIREDO Oncology Center (Care, Innovation and Research for Children and AYA with Cancer), Institut Curie, Paris, France
| |
Collapse
|
192
|
Aiba T, Hattori C, Sugisaka J, Shimizu H, Ono H, Domeki Y, Saito R, Kawana S, Kawashima Y, Terayama K, Toi Y, Nakamura A, Yamanda S, Kimura Y, Suzuki Y, Niida A, Sugawara S. Gene expression signatures as candidate biomarkers of response to PD-1 blockade in non-small cell lung cancers. PLoS One 2021; 16:e0260500. [PMID: 34843570 PMCID: PMC8629226 DOI: 10.1371/journal.pone.0260500] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Accepted: 11/10/2021] [Indexed: 11/18/2022] Open
Abstract
Although anti-PD-1/PD-L1 monotherapy has achieved clinical success in non-small cell lung cancer (NSCLC), definitive predictive biomarkers remain to be elucidated. In this study, we performed whole-transcriptome sequencing of pretreatment tumor tissue samples and pretreatment and on-treatment whole blood samples (WB) samples obtained from a clinically annotated cohort of NSCLC patients (n = 40) treated with nivolumab (anti-PD-1) monotherapy. Using a single-sample gene set enrichment scoring method, we found that the tumors of responders with lung adenocarcinoma (LUAD, n = 20) are inherently immunogenic to promote antitumor immunity, whereas those with lung squamous cell carcinoma (LUSC, n = 18) have a less immunosuppressive tumor microenvironment. These findings suggested that nivolumab may function as a molecular targeted agent in LUAD and as an immunomodulating agent in LUSC. In addition, our study explains why the reliability of PD-L1 expression on tumor cells as a predictive biomarker for the response to nivolumab monotherapy is quite different between LUAD and LUSC.
Collapse
Affiliation(s)
- Tomoiki Aiba
- Department of Pulmonary Medicine, Sendai Kousei Hospital, Sendai, Japan
- * E-mail:
| | - Chieko Hattori
- Department of Pulmonary Medicine, Sendai Kousei Hospital, Sendai, Japan
| | - Jun Sugisaka
- Department of Pulmonary Medicine, Sendai Kousei Hospital, Sendai, Japan
| | - Hisashi Shimizu
- Department of Pulmonary Medicine, Sendai Kousei Hospital, Sendai, Japan
| | - Hirotaka Ono
- Department of Pulmonary Medicine, Sendai Kousei Hospital, Sendai, Japan
| | - Yutaka Domeki
- Department of Pulmonary Medicine, Sendai Kousei Hospital, Sendai, Japan
| | - Ryohei Saito
- Department of Pulmonary Medicine, Sendai Kousei Hospital, Sendai, Japan
| | - Sachiko Kawana
- Department of Pulmonary Medicine, Sendai Kousei Hospital, Sendai, Japan
| | - Yosuke Kawashima
- Department of Pulmonary Medicine, Sendai Kousei Hospital, Sendai, Japan
| | - Keisuke Terayama
- Department of Pulmonary Medicine, Sendai Kousei Hospital, Sendai, Japan
| | - Yukihiro Toi
- Department of Pulmonary Medicine, Sendai Kousei Hospital, Sendai, Japan
| | - Atsushi Nakamura
- Department of Pulmonary Medicine, Sendai Kousei Hospital, Sendai, Japan
| | - Shinsuke Yamanda
- Department of Pulmonary Medicine, Sendai Kousei Hospital, Sendai, Japan
| | - Yuichiro Kimura
- Department of Pulmonary Medicine, Sendai Kousei Hospital, Sendai, Japan
| | - Yutaka Suzuki
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Chiba, Japan
| | - Atsushi Niida
- Human Genome Center, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Shunichi Sugawara
- Department of Pulmonary Medicine, Sendai Kousei Hospital, Sendai, Japan
| |
Collapse
|
193
|
Long non-coding RNA HOMER3-AS1 drives hepatocellular carcinoma progression via modulating the behaviors of both tumor cells and macrophages. Cell Death Dis 2021; 12:1103. [PMID: 34815380 PMCID: PMC8611033 DOI: 10.1038/s41419-021-04309-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 10/07/2021] [Accepted: 10/07/2021] [Indexed: 12/21/2022]
Abstract
The crosstalk between cancer cells and tumor microenvironment plays critical roles in hepatocellular carcinoma (HCC). The identification of long non-coding RNAs (lncRNAs) mediating the crosstalk might promote the development of new therapeutic strategies against HCC. Here, we identified a lncRNA, HOMER3-AS1, which is over-expressed in HCC and correlated with poor survival of HCC patients. HOMER3-AS1 promoted HCC cellular proliferation, migration, and invasion, and reduced HCC cellular apoptosis. Furthermore, HOMER3-AS1 promoted macrophages recruitment and M2-like polarization. In vivo, HOMER3-AS1 significantly facilitated HCC progression. Mechanism investigations revealed that HOMER3-AS1 activated Wnt/β-catenin signaling via upregulating HOMER3. Functional rescue experiments revealed that HOMER3/Wnt/β-catenin axis mediated the roles of HOMER3-AS1 in promoting HCC cellular malignant phenotypes. Furthermore, colony stimulating factor-1 (CSF-1) was also identified as a critical downstream target of HOMER3-AS1. HOMER3-AS1 increased CSF-1 expression and secretion. Blocking CSF-1 reversed the roles of HOMER3-AS1 in inducing macrophages recruitment and M2 polarization. Furthermore, positive correlations between HOMER3-AS1 and HOMER3 expression, HOMER3-AS1 and CSF-1 expression, and HOMER3-AS1 expression and M2-like macrophages infiltration were found in human HCC tissues. In summary, our findings demonstrated that HOMER3-AS1 drives HCC progression via modulating the behaviors of both tumor cells and macrophages, which are dependent on the activation of HOMER3/Wnt/β-catenin axis and CSF-1, respectively. HOMER3-AS1 might be a promising prognostic and therapeutic target for HCC.
Collapse
|
194
|
Peng H. Perspectives on the development of antibody-drug conjugates targeting ROR1 for hematological and solid cancers. Antib Ther 2021; 4:222-227. [PMID: 34805745 PMCID: PMC8597957 DOI: 10.1093/abt/tbab023] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 09/13/2021] [Accepted: 10/10/2021] [Indexed: 02/07/2023] Open
Abstract
Antibody–drug conjugates (ADCs) are targeted therapeutics generated by conjugation of cytotoxic small molecules to monoclonal antibodies (mAbs) via chemical linkers. Due to their selective delivery of toxic payloads to antigen-positive cancer cells, ADCs demonstrate wider therapeutic indexes compared with conventional chemotherapy. After decades of intensive research and development, significant advances have been made in the field, leading to a total of 10 U.S. food and drug administration (FDA)-approved ADCs to treat cancer patients. Currently, ~80 ADCs targeting different antigens are under clinical evaluation for treatment of either hematological or solid malignancies. Notably, three ADCs targeting the same oncofetal protein, receptor tyrosine kinase like orphan receptor 1 (ROR1), have attracted considerable attention when they were acquired or licensed successively in the fourth quarter of 2020 by three major pharmaceutical companies. Apparently, ROR1 has emerged as an attractive target for cancer therapy. Since all the components of ADCs, including the antibody, linker and payload, as well as the conjugation method, play critical roles in ADC’s efficacy and performance, their choice and combination will determine how far they can be advanced. This review summarizes the design and development of current anti-ROR1 ADCs and highlights an emerging trend to target ROR1 for cancer therapy.
Collapse
Affiliation(s)
- Haiyong Peng
- Department of Immunology and Microbiology, The Scripps Research Institute, 130 Scripps Way, C278, Jupiter, FL 33458, USA
| |
Collapse
|
195
|
Xiao BF, Zhang JT, Zhu YG, Cui XR, Lu ZM, Yu BT, Wu N. Chimeric Antigen Receptor T-Cell Therapy in Lung Cancer: Potential and Challenges. Front Immunol 2021; 12:782775. [PMID: 34790207 PMCID: PMC8591168 DOI: 10.3389/fimmu.2021.782775] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Accepted: 10/13/2021] [Indexed: 12/21/2022] Open
Abstract
Chimeric antigen receptor T (CAR-T) cell therapy has exhibited a substantial clinical response in hematological malignancies, including B-cell leukemia, lymphoma, and multiple myeloma. Therefore, the feasibility of using CAR-T cells to treat solid tumors is actively evaluated. Currently, multiple basic research projects and clinical trials are being conducted to treat lung cancer with CAR-T cell therapy. Although numerous advances in CAR-T cell therapy have been made in hematological tumors, the technology still entails considerable challenges in treating lung cancer, such as on−target, of−tumor toxicity, paucity of tumor-specific antigen targets, T cell exhaustion in the tumor microenvironment, and low infiltration level of immune cells into solid tumor niches, which are even more complicated than their application in hematological tumors. Thus, progress in the scientific understanding of tumor immunology and improvements in the manufacture of cell products are advancing the clinical translation of these important cellular immunotherapies. This review focused on the latest research progress of CAR-T cell therapy in lung cancer treatment and for the first time, demonstrated the underlying challenges and future engineering strategies for the clinical application of CAR-T cell therapy against lung cancer.
Collapse
Affiliation(s)
- Bu-Fan Xiao
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Thoracic Surgery II, Peking University Cancer Hospital & Institute, Beijing, China
| | - Jing-Tao Zhang
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yu-Ge Zhu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Thoracic Surgery II, Peking University Cancer Hospital & Institute, Beijing, China
| | - Xin-Run Cui
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Thoracic Surgery II, Peking University Cancer Hospital & Institute, Beijing, China
| | - Zhe-Ming Lu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Biochemistry and Molecular Biology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Ben-Tong Yu
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Nan Wu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Thoracic Surgery II, Peking University Cancer Hospital & Institute, Beijing, China
| |
Collapse
|
196
|
Daikuzono H, Yamazaki M, Sato Y, Takahashi T, Yamagata K. Development of a DELFIA method to detect oncofetal antigen ROR1-positive exosomes. Biochem Biophys Res Commun 2021; 578:170-176. [PMID: 34597914 DOI: 10.1016/j.bbrc.2021.08.054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 08/03/2021] [Accepted: 08/23/2021] [Indexed: 10/20/2022]
Abstract
Receptor tyrosine kinase-like orphan receptor 1 (ROR1) is highly expressed in a wide variety of hematological and solid cancers, but is low or absent in adult tissues. Here, we show that ROR1 is released with exosomes from ROR1-positive cancer cells. We also developed a simple dissociation-enhanced lanthanide fluorescence immunoassay (DELFIA) to detect cancer-derived ROR1-positive exosomes, which are captured by two anti-ROR1 antibodies and detected by the fluorescence of free chelating europium. This new DELFIA method can detect cancer-derived ROR1-positive exosomes in the cell supernatant and serum with a wide range and rapidly compared with the conventional Western blot assay. This method may be useful as a companion diagnostics for ROR1-positive cancers.
Collapse
Affiliation(s)
- Hina Daikuzono
- Department of Cancer Biology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, 860-8556, Japan
| | - Masaya Yamazaki
- Department of Cancer Biology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, 860-8556, Japan; Department of Medical Biochemistry, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, 860-8556, Japan.
| | - Yoshifumi Sato
- Department of Medical Biochemistry, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, 860-8556, Japan
| | | | - Kazuya Yamagata
- Department of Medical Biochemistry, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, 860-8556, Japan
| |
Collapse
|
197
|
Pocaterra A, Catucci M, Mondino A. Adoptive T cell therapy of solid tumors: time to team up with immunogenic chemo/radiotherapy. Curr Opin Immunol 2021; 74:53-59. [PMID: 34743069 DOI: 10.1016/j.coi.2021.10.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 10/13/2021] [Accepted: 10/19/2021] [Indexed: 12/20/2022]
Abstract
Adoptive T cell therapy (ACT) with tumor-reactive lymphocytes can overcome the immune desert of poorly immunogenic tumors and instruct tumor eradication. Several hurdles limit the efficacy of this strategy against solid tumor including, but not limited to, sub optimal T cell engraftment, tumor infiltration, poor tumor antigenicity/immunogenicity, and immunosuppressive or resistance mechanisms. Recent advances indicate that concomitant treatments can be set in place to offset such barriers. In this review, we highlight the beneficial effects of combining ACT with conventional chemo and/or radiotherapy. While originally classified as immunosuppressive, these methodologies can also promote the engraftment of ACT products, immunogenic cell death, and the reprogramming of more favorable microenvironments. Data indicates that systemic and local chemo/radiotherapy regimens promote intratumoral cytokine and chemokine upregulation, tumor antigen presentation and cross presentation, infiltration and in situ T cells reactivation. Here we review the most recent contributions supporting these notions and discuss further developments.
Collapse
Affiliation(s)
- Arianna Pocaterra
- Lymphocyte Activation Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Via Olgettina, 58, 20132, Milan, Italy
| | - Marco Catucci
- Lymphocyte Activation Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Via Olgettina, 58, 20132, Milan, Italy
| | - Anna Mondino
- Lymphocyte Activation Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Via Olgettina, 58, 20132, Milan, Italy.
| |
Collapse
|
198
|
Wan G, Chen X, Wang H, Hou S, Wang Q, Cheng Y, Chen Q, Lv Y, Chen H, Zhang Q. Gene augmented nuclear-targeting sonodynamic therapy via Nrf2 pathway-based redox balance adjustment boosts peptide-based anti-PD-L1 therapy on colorectal cancer. J Nanobiotechnology 2021; 19:347. [PMID: 34715867 PMCID: PMC8555306 DOI: 10.1186/s12951-021-01094-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Accepted: 10/18/2021] [Indexed: 12/14/2022] Open
Abstract
Background Colorectal cancer is known to be resistant to immune checkpoint blockade (ICB) therapy. Sonodynamic therapy (SDT) has been reported to improve the efficacy of immunotherapy by inducing immunogenic cell death (ICD) of cancer. However, the SDT efficacy is extremely limited by Nrf2-based natural redox balance regulation pathway in cancer cells in response to the increased contents of reactive oxygen species (ROS). Nuclear-targeting strategy has shown unique advantages in tumor therapy by directly destroying the DNA. Thus it can be seen that Nrf2-siRNA augmented nuclear-targeting SDT could boost ICB therapy against colorectal cancer. Results The nuclear-targeting delivery system TIR@siRNA (TIR was the abbreviation of assembled TAT-IR780) with great gene carrier capacity and smaller diameter (< 60 nm) was designed to achieve the gene augmented nuclear-targeting SDT facilitating the anti-PD-L1 (programmed cell death-ligand-1) therapy against colorectal cancer. In CT26 cells, TIR@siRNA successfully delivered IR780 (the fluorescent dye used as sonosensitizer) into cell nucleus and Nrf2-siRNA into cytoplasm. Under US (utrasound) irradiation, TIR@siRNA notably increased the cytotoxicity and apoptosis-inducing activity of SDT through down-regulating the Nrf2, directly damaging the DNA, activating mitochondrial apoptotic pathway while remarkably inducing ICD of CT26 cells. In CT26 tumor-bearing mice, TIR@siRNA mediated gene enhanced nuclear-targeting SDT greatly inhibited tumor growth, noticeably increased the T cell infiltration and boosted DPPA-1 peptide-based anti-PD-L1 therapy to ablate the primary CT26 tumors and suppress the intestinal metastases. Conclusions All results demonstrate that TIR@siRNA under US irradiation can efficiently inhibit the tumor progression toward colorectal CT26 cancer in vitro and in vivo by its mediated gene augmented nuclear-targeting sonodynamic therapy. Through fully relieving the immunosuppressive microenvironment of colorectal cancer by this treatment, this nanoplatform provides a new synergistic strategy for enhancing the anti-PD-L1 therapy to ablate colorectal cancer and inhibit its metastasis. Graphical Abstract ![]()
Supplementary Information The online version contains supplementary material available at 10.1186/s12951-021-01094-x.
Collapse
Affiliation(s)
- Guoyun Wan
- The Key Laboratory of Biomedical Material, School of Life Science and Technology, Xinxiang Medical University, 453003, Xinxiang, China.
| | - Xuheng Chen
- The Key Laboratory of Biomedical Material, School of Life Science and Technology, Xinxiang Medical University, 453003, Xinxiang, China
| | - Haijiao Wang
- The Key Laboratory of Biomedical Material, School of Life Science and Technology, Xinxiang Medical University, 453003, Xinxiang, China
| | - Shenglei Hou
- Institute of Biomedical Engineering, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), 518020, Shenzhen, China.,Post-doctoral Scientific Research Station of Basic Medicine, Jinan Unviersity, 510632, Guangzhou, China
| | - Qian Wang
- The Key Laboratory of Biomedical Material, School of Life Science and Technology, Xinxiang Medical University, 453003, Xinxiang, China
| | - Yuanyuan Cheng
- School of Pharmacy, Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics), Tianjin Medical University, 300070, Tianjin, China
| | - Qian Chen
- School of Pharmacy, Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics), Tianjin Medical University, 300070, Tianjin, China
| | - Yingge Lv
- The Key Laboratory of Biomedical Material, School of Life Science and Technology, Xinxiang Medical University, 453003, Xinxiang, China
| | - Hongli Chen
- The Key Laboratory of Biomedical Material, School of Life Science and Technology, Xinxiang Medical University, 453003, Xinxiang, China.
| | - Qiqing Zhang
- The Key Laboratory of Biomedical Material, School of Life Science and Technology, Xinxiang Medical University, 453003, Xinxiang, China. .,Institute of Biomedical Engineering, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), 518020, Shenzhen, China. .,Post-doctoral Scientific Research Station of Basic Medicine, Jinan Unviersity, 510632, Guangzhou, China.
| |
Collapse
|
199
|
Lamb LS, Pereboeva L, Youngblood S, Gillespie GY, Nabors LB, Markert JM, Dasgupta A, Langford C, Spencer HT. A combined treatment regimen of MGMT-modified γδ T cells and temozolomide chemotherapy is effective against primary high grade gliomas. Sci Rep 2021; 11:21133. [PMID: 34702850 PMCID: PMC8548550 DOI: 10.1038/s41598-021-00536-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 10/07/2021] [Indexed: 01/13/2023] Open
Abstract
Chemotherapeutic drugs such as the alkylating agent Temozolomide (TMZ), in addition to reducing tumor mass, can also sensitize tumors to immune recognition by transient upregulation of multiple stress induced NKG2D ligands (NKG2DL). However, the potential for an effective response by innate lymphocyte effectors such as NK and γδ T cells that recognize NKG2DL is limited by the drug's concomitant lymphodepleting effects. We have previously shown that modification of γδ T cells with a methylguanine DNA methyltransferase (MGMT) transgene confers TMZ resistance via production of O6-alkylguanine DNA alkyltransferase (AGT) thereby enabling γδ T cell function in therapeutic concentrations of TMZ. In this study, we tested this strategy which we have termed Drug Resistant Immunotherapy (DRI) to examine whether combination therapy of TMZ and MGMT-modified γδ T cells could improve survival outcomes in four human/mouse xenograft models of primary and refractory GBM. Our results confirm that DRI leverages the innate response of γδ T cells to chemotherapy-induced stress associated antigen expression and achieves synergies that are significantly greater than either individual approach.
Collapse
Affiliation(s)
- Lawrence S Lamb
- Department of Medicine, Division of Hematology and Oncology, University of Alabama at Birmingham, Birmingham, AL, USA.
| | - Larisa Pereboeva
- Department of Medicine, Division of Hematology and Oncology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Samantha Youngblood
- Department of Medicine, Division of Hematology and Oncology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - G Yancey Gillespie
- Department of Neurosurgery, University of Alabama at Birmingham, Birmingham, AL, USA
| | - L Burton Nabors
- Department of Neurology, Division of Neuro-Oncology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - James M Markert
- Department of Neurosurgery, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Anindya Dasgupta
- Department of Pediatrics, Aflac Cancer and Blood Disorders Center, Emory University, Atlanta, GA, USA
| | - Catherine Langford
- Department of Neurosurgery, University of Alabama at Birmingham, Birmingham, AL, USA
| | - H Trent Spencer
- Department of Pediatrics, Aflac Cancer and Blood Disorders Center, Emory University, Atlanta, GA, USA
| |
Collapse
|
200
|
Decalf J, Tom J, Mai E, Hernandez-Barry H, Noland CL, Vollmar BS, Li A, Li H, Xie D, Zhu L, Payandeh J, Wu C, Comps-Agrar L, Moussion C, Albert ML, Song A. A novel method to produce synthetic murine CXCL10 for efficient screening of functional variants. Bioorg Chem 2021; 116:105376. [PMID: 34560560 DOI: 10.1016/j.bioorg.2021.105376] [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] [Received: 05/26/2021] [Revised: 09/13/2021] [Accepted: 09/14/2021] [Indexed: 11/25/2022]
Abstract
Antitumor immune responses depend on the infiltration of solid tumors by effector T cells, a process guided by chemokines. In particular, the chemokine CXCL10 has been shown to play a critical role in mediating recruitment of CXCR3 + cytolytic T and NK cells in tumors, though its use as a therapeutic agent has not been widely explored. One of the limitations is due to the rapid inactivation of CXCL10 by dipeptidyl peptidase 4 (DPP4), a broadly expressed enzyme that is active in plasma and other bodily fluids. In the present study, we describe a novel method to produce synthetic CXCL10 that is resistant to DPP4 N-terminal truncation. Using a Fmoc solid-phase peptide synthesis approach, synthetic murine WT CXCL10 was produced, showing similar biochemical and biological properties to the recombinant protein. This synthesis method supported production of natural (amino acid substitution, insertion or deletion) and non-natural (chemical modifications) variants of CXCL10. In association with a functional screening cascade that assessed DPP4-mediated cleavage, CXCR3 signaling potency and chemotactic activity, we successfully generated 20 murine CXCL10 variants. Among those, two non-natural variants with N-methylated Leu3 (MeLeu3) and a reduced amide bond between Pro2 and Leu3 (rLeu3), respectively, showed resistance to DPP4 truncation but decreased CXCR3 signaling and chemotactic activity. Interestingly, MeLeu3 and rLeu3 CXCL10 behaved as DPP4 inhibitors, preventing the truncation of WT CXCL10. This study highlights the potential of using Fmoc solid-phase chemistry in association with biochemical and biological characterization to rapidly identify CXCL10 variants with desired properties. These novel methods unlock the opportunity to develop DPP4 resistant CXCL10 variants, as well as other chemokine substrates, while maintaining chemotactic properties.
Collapse
Affiliation(s)
- Jérémie Decalf
- Department of Cancer Immunology, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA.
| | - Jeffrey Tom
- Department of Early Discovery Biochemistry, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Elaine Mai
- Department of Biochemical and Cellular Pharmacology, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Hilda Hernandez-Barry
- Department of Biochemical and Cellular Pharmacology, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Cameron L Noland
- Department of Structural Biology, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Breanna S Vollmar
- Department of Protein Chemistry, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Alice Li
- Department of Protein Chemistry, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Hong Li
- Department of Protein Chemistry, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Daniel Xie
- TideMed Pharma, Room 1-1115, No. 291, Fucheng Road, Hangzhou Qiantang New Area, Hangzhou 310018, China
| | - Lunchao Zhu
- TideMed Pharma, Room 1-1115, No. 291, Fucheng Road, Hangzhou Qiantang New Area, Hangzhou 310018, China
| | - Jian Payandeh
- Department of Structural Biology, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Cong Wu
- Department of Biochemical and Cellular Pharmacology, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Laetitia Comps-Agrar
- Department of Biochemical and Cellular Pharmacology, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Christine Moussion
- Department of Cancer Immunology, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Matthew L Albert
- Department of Cancer Immunology, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA; Department of Immunology and Infectious Diseases, Insitro, South San Francisco, CA 94080, USA
| | - Aimin Song
- Department of Early Discovery Biochemistry, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA.
| |
Collapse
|