1
|
Zhang H, Huo Y, Zheng W, Li P, Li H, Zhang L, Sa L, He Y, Zhao Z, Shi C, Shan L, Yang A, Wang T. Silencing of SIRPα enhances the antitumor efficacy of CAR-M in solid tumors. Cell Mol Immunol 2024; 21:1335-1349. [PMID: 39379603 PMCID: PMC11527885 DOI: 10.1038/s41423-024-01220-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 09/14/2024] [Indexed: 10/10/2024] Open
Abstract
The potential of macrophage-mediated phagocytosis as a cancer treatment is promising. Blocking the CD47-SIRPα interaction with a CD47-specific antibody significantly enhances macrophage phagocytosis. However, concerns regarding their toxicity to nontumor cells remain substantial. Here, we engineered chimeric antigen receptor macrophages (CAR-Ms) by fusing a humanized single-chain variable fragment with FcγRIIa and integrating short hairpin RNA to silence SIRPα, thereby disrupting the CD47-SIRPα signaling pathway. These modified CAR-shSIRPα-M cells exhibited an M1-like phenotype, superior phagocytic function, substantial cytotoxic effects on HER2-positive tumor cells, and the ability to eliminate patient-derived organoids. In vivo, CAR-M cells significantly inhibited tumor growth and prolonged survival in tumor-bearing mice. Notably, CAR-shSIRPα-M cells enhanced cytotoxic T-cell infiltration into tumors, thereby enhancing the antitumor response in both the humanized immune system mouse model and immunocompetent mice. Mechanistically, SIRPα inhibition activated inflammatory pathways and the cGAS-STING signaling cascade in CAR-M cells, leading to increased production of proinflammatory cytokines, reactive oxygen species, and nitric oxide, thereby enhancing their antitumor effects. These findings underscore the potential of SIRPα inhibition as a novel strategy to increase the antitumor efficacy of CAR-M cells in cancer immunotherapy, particularly against solid tumors.
Collapse
MESH Headings
- Animals
- Receptors, Immunologic/metabolism
- Receptors, Immunologic/genetics
- Humans
- Mice
- Receptors, Chimeric Antigen/metabolism
- Receptors, Chimeric Antigen/genetics
- Receptors, Chimeric Antigen/immunology
- CD47 Antigen/metabolism
- Cell Line, Tumor
- Antigens, Differentiation
- Macrophages/immunology
- Macrophages/metabolism
- Neoplasms/therapy
- Neoplasms/immunology
- Phagocytosis
- Signal Transduction
- Gene Silencing
- Receptors, IgG/metabolism
- Receptors, IgG/genetics
- Immunotherapy, Adoptive/methods
- Female
Collapse
Affiliation(s)
- Han Zhang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Department of Medical Genetics and Developmental Biology, Fourth Military Medical University, Xi'an, China
- Department of Spine Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Yi Huo
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Department of Immunology, Fourth Military Medical University, Xi'an, China
| | - Wenjing Zheng
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Department of Medical Genetics and Developmental Biology, Fourth Military Medical University, Xi'an, China
| | - Peng Li
- Division of Cancer Biology, Laboratory Animal Center, Fourth Military Medical University, Xi'an, China
| | - Hui Li
- Division of Cancer Biology, Laboratory Animal Center, Fourth Military Medical University, Xi'an, China
| | - Lingling Zhang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Department of Medical Genetics and Developmental Biology, Fourth Military Medical University, Xi'an, China
| | - Longqi Sa
- Department of Spine Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Yang He
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Department of Medical Genetics and Developmental Biology, Fourth Military Medical University, Xi'an, China
| | - Zihao Zhao
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Department of Medical Genetics and Developmental Biology, Fourth Military Medical University, Xi'an, China
| | - Changhong Shi
- Division of Cancer Biology, Laboratory Animal Center, Fourth Military Medical University, Xi'an, China.
| | - Lequn Shan
- Department of Spine Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, China.
| | - Angang Yang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Department of Immunology, Fourth Military Medical University, Xi'an, China.
| | - Tao Wang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Department of Medical Genetics and Developmental Biology, Fourth Military Medical University, Xi'an, China.
| |
Collapse
|
2
|
Balachandran Pillai A, Yousef M, Yousef A, Alfaro-Munoz KD, Smaglo BG, Willis J, Wolff RA, Pant S, Hurd MW, Maitra A, Wang H, Katz MHG, Prakash LR, Tzeng CWD, Snyder R, Castelnovo LF, Chen A, Kravets A, Kudriavtseva K, Tarasov A, Kryukov K, Ying H, Shen JP, Zhao D. Molecular and Clinical Features of Pancreatic Acinar Cell Carcinoma: A Single-Institution Case Series. Cancers (Basel) 2024; 16:3421. [PMID: 39410042 PMCID: PMC11475689 DOI: 10.3390/cancers16193421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 09/18/2024] [Accepted: 09/24/2024] [Indexed: 10/20/2024] Open
Abstract
Objectives: Acinar cell carcinoma (ACC) accounts for about 1% of pancreatic cancers. The molecular and clinical features of ACC are less characterized than those of pancreatic ductal adenocarcinoma. Methods: We retrospectively evaluated the clinical and molecular features of ACC patients who underwent germline and/or somatic molecular testing at The University of Texas MD Anderson Cancer Center from 2008 to 2022 and two cases from 2023-2024 who underwent RNA and TME analysis by Boston Gene. Patient information was extracted from our institutional database with the approval of the Institutional Review Board. Results: We identified 16 patients with available molecular testing results. Fourteen patients had metastatic disease, one had borderline resectable disease, and one had localized resectable disease at diagnosis. Fifteen patients were wild type for KRAS (one patient had unknown KRAS status). Somatic/germline mutations of DNA damage repair genes (BRCA1/2, PALB2, and ATM) were present in 5 of 12 patients tested for these genes. One patient was found to have RET fusion and responded favorably to selpercatinib for over 42 months. The median overall survival (OS) was 24 months for patients with metastatic disease. One of the additional two cases who underwent BostonGene testing was found to have NTRK1 fusion. RNA and TME analysis by Boston Gene of the two cases reported immune desert features and relatively lower RNA levels of CEACAM5, CD47, CD74, and MMP1 and higher RNA levels of CDH6 compared with PDAC.
Collapse
Affiliation(s)
| | - Mahmoud Yousef
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA (A.Y.); (K.D.A.-M.); (B.G.S.); (J.W.); (R.A.W.); (S.P.); (M.W.H.); (L.F.C.); (A.C.); (J.P.S.)
| | - Abdelrahman Yousef
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA (A.Y.); (K.D.A.-M.); (B.G.S.); (J.W.); (R.A.W.); (S.P.); (M.W.H.); (L.F.C.); (A.C.); (J.P.S.)
| | - Kristin D. Alfaro-Munoz
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA (A.Y.); (K.D.A.-M.); (B.G.S.); (J.W.); (R.A.W.); (S.P.); (M.W.H.); (L.F.C.); (A.C.); (J.P.S.)
| | - Brandon G. Smaglo
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA (A.Y.); (K.D.A.-M.); (B.G.S.); (J.W.); (R.A.W.); (S.P.); (M.W.H.); (L.F.C.); (A.C.); (J.P.S.)
| | - Jason Willis
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA (A.Y.); (K.D.A.-M.); (B.G.S.); (J.W.); (R.A.W.); (S.P.); (M.W.H.); (L.F.C.); (A.C.); (J.P.S.)
| | - Robert A. Wolff
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA (A.Y.); (K.D.A.-M.); (B.G.S.); (J.W.); (R.A.W.); (S.P.); (M.W.H.); (L.F.C.); (A.C.); (J.P.S.)
| | - Shubham Pant
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA (A.Y.); (K.D.A.-M.); (B.G.S.); (J.W.); (R.A.W.); (S.P.); (M.W.H.); (L.F.C.); (A.C.); (J.P.S.)
| | - Mark W. Hurd
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA (A.Y.); (K.D.A.-M.); (B.G.S.); (J.W.); (R.A.W.); (S.P.); (M.W.H.); (L.F.C.); (A.C.); (J.P.S.)
- Ahmed Center for Pancreatic Cancer Research, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Anirban Maitra
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (A.M.); (H.W.)
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Huamin Wang
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (A.M.); (H.W.)
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Matthew Harold G. Katz
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (M.H.G.K.); (L.R.P.); (C.-W.D.T.); (R.S.)
| | - Laura R. Prakash
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (M.H.G.K.); (L.R.P.); (C.-W.D.T.); (R.S.)
| | - Ching-Wei D. Tzeng
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (M.H.G.K.); (L.R.P.); (C.-W.D.T.); (R.S.)
| | - Rebecca Snyder
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (M.H.G.K.); (L.R.P.); (C.-W.D.T.); (R.S.)
| | - Luca F. Castelnovo
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA (A.Y.); (K.D.A.-M.); (B.G.S.); (J.W.); (R.A.W.); (S.P.); (M.W.H.); (L.F.C.); (A.C.); (J.P.S.)
| | - Anthony Chen
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA (A.Y.); (K.D.A.-M.); (B.G.S.); (J.W.); (R.A.W.); (S.P.); (M.W.H.); (L.F.C.); (A.C.); (J.P.S.)
| | - Andrey Kravets
- BostonGene Corporation, 100 Beaver St, Waltham, MA 02453, USA; (A.K.); (K.K.); (A.T.); (K.K.)
| | - Kseniia Kudriavtseva
- BostonGene Corporation, 100 Beaver St, Waltham, MA 02453, USA; (A.K.); (K.K.); (A.T.); (K.K.)
| | - Artem Tarasov
- BostonGene Corporation, 100 Beaver St, Waltham, MA 02453, USA; (A.K.); (K.K.); (A.T.); (K.K.)
| | - Kirill Kryukov
- BostonGene Corporation, 100 Beaver St, Waltham, MA 02453, USA; (A.K.); (K.K.); (A.T.); (K.K.)
| | - Haoqiang Ying
- Department of Molecular and Cellular Oncology, Division of Basic Science Research, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA;
| | - John Paul Shen
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA (A.Y.); (K.D.A.-M.); (B.G.S.); (J.W.); (R.A.W.); (S.P.); (M.W.H.); (L.F.C.); (A.C.); (J.P.S.)
| | - Dan Zhao
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA (A.Y.); (K.D.A.-M.); (B.G.S.); (J.W.); (R.A.W.); (S.P.); (M.W.H.); (L.F.C.); (A.C.); (J.P.S.)
| |
Collapse
|
3
|
Luo W, Hoang H, Zhu H, Miller K, Mo X, Eguchi S, Tian M, Liao Y, Ayello J, Rosenblum JM, Marcondes M, Currier M, Mardis E, Cripe T, Lee D, Cairo MS. Circumventing resistance within the Ewing sarcoma microenvironment by combinatorial innate immunotherapy. J Immunother Cancer 2024; 12:e009726. [PMID: 39266215 PMCID: PMC11404285 DOI: 10.1136/jitc-2024-009726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/27/2024] [Indexed: 09/14/2024] Open
Abstract
BACKGROUND Pediatric patients with recurrent/metastatic Ewing sarcoma (ES) have a dismal 5-year survival. Novel therapeutic approaches are desperately needed. Natural killer (NK) cell number and function are low in ES patient tumors, in large part due to the immunosuppressive tumor microenvironment (TME). Melanoma cell adhesion molecule (MCAM) is highly expressed on ES and associated with ES metastasis. NKTR-255 is a polymer-conjugated recombinant human interleukin-15 (IL-15) agonist improving NK cell activity and persistence. Magrolimab (MAG) is a CD47 blockade that reactivates the phagocytic activity of macrophages. METHODS Transcriptome profiling coupled with CIBERSORT analyses in both ES mouse xenografts and human patient tumors were performed to identify mechanisms of NK resistance in ES TME. A chimeric antigen receptor (CAR) NK cell targeting MCAM was engineered by CAR mRNA electroporation into ex vivo expanded NK cells. In vitro cytotoxicity assays were performed to investigate the efficacy of anti-MCAM-CAR-NK cell alone or combined with NKTR-255 against ES cells. Interferon-γ and perforin levels were measured by ELISA. The effect of MAG on macrophage phagocytosis of ES cells was evaluated by in vitro phagocytosis assays. Cell-based and patient-derived xenograft (PDX)-based xenograft mouse models of ES were used to investigate the antitumor efficacy of CAR-NK alone and combined with NKTR-255 and MAG in vivo. RESULTS We found that NK cell infiltration and activity were negatively regulated by tumor-associated macrophages (TAM) in ES TME. Expression of anti-MCAM CAR significantly and specifically enhanced NK cytotoxic activity against MCAMhigh but not MCAM-knockout ES cells in vitro, and significantly reduced lung metastasis and extended animal survival in vivo. NKTR-255 and MAG significantly enhanced in vitro CAR-NK cytotoxicity and macrophage phagocytic activity against ES cells, respectively. By combining with NKTR-255 and MAG, the anti-MCAM-CAR-NK cell significantly decreased primary tumor growth and prolonged animal survival in both cell- and PDX-based ES xenograft mouse models. CONCLUSIONS Our preclinical studies demonstrate that immunotherapy via the innate immune system by combining tumor-targeting CAR-NK cells with an IL-15 agonist and a CD47 blockade is a promising novel therapeutic approach to targeting MCAMhigh malignant metastatic ES.
Collapse
Affiliation(s)
- Wen Luo
- Department of Pediatrics, New York Medical College, Valhalla, New York, USA
- Department of Pathology, Immunology and Microbiology, New York Medical College, Valhalla, New York, USA
| | - Hai Hoang
- Department of Pediatrics, New York Medical College, Valhalla, New York, USA
| | - Hongwen Zhu
- Department of Pediatrics, New York Medical College, Valhalla, New York, USA
| | - Katherine Miller
- The Steve and Cindy Rasmussen Institute for Genomic Medicine, Columbus, Ohio, USA
- Pediatric Hem/Onc/BMT, Nationwide Children's Hospital Hematology Oncology and Blood and Marrow Transplant, Columbus, Ohio, USA
| | - Xiaokui Mo
- Biomedical Informatics, The Ohio State University, Columbus, Ohio, USA
| | - Shiori Eguchi
- Department of Pediatrics, New York Medical College, Valhalla, New York, USA
| | - Meijuan Tian
- Department of Pediatrics, New York Medical College, Valhalla, New York, USA
| | - Yanling Liao
- Department of Pediatrics, New York Medical College, Valhalla, New York, USA
| | - Janet Ayello
- Department of Pediatrics, New York Medical College, Valhalla, New York, USA
| | - Jeremy M Rosenblum
- Department of Pediatrics, New York Medical College, Valhalla, New York, USA
| | | | - Mark Currier
- The Steve and Cindy Rasmussen Institute for Genomic Medicine, Columbus, Ohio, USA
| | - Elaine Mardis
- The Steve and Cindy Rasmussen Institute for Genomic Medicine, Columbus, Ohio, USA
- Pediatric Hem/Onc/BMT, Nationwide Children's Hospital Hematology Oncology and Blood and Marrow Transplant, Columbus, Ohio, USA
- Department of Neurosurgery, The Ohio State University, Columbus, Ohio, USA
| | - Timothy Cripe
- Pediatric Hem/Onc/BMT, Nationwide Children's Hospital Hematology Oncology and Blood and Marrow Transplant, Columbus, Ohio, USA
| | - Dean Lee
- Pediatric Hem/Onc/BMT, Nationwide Children's Hospital Hematology Oncology and Blood and Marrow Transplant, Columbus, Ohio, USA
| | - Mitchell S Cairo
- Department of Pediatrics, New York Medical College, Valhalla, New York, USA
- Departments of Pathology, Immunology and Microbiology, Medicine, Cell Biology and Anatomy, New York Medical College, Valhalla, New York, USA
| |
Collapse
|
4
|
Brichkina A, Ems M, Suezov R, Singh R, Lutz V, Picard FSR, Nist A, Stiewe T, Graumann J, Daude M, Diederich WE, Finkernagel F, Chung HR, Bartsch DK, Roth K, Keber C, Denkert C, Huber M, Gress TM, Lauth M. DYRK1B blockade promotes tumoricidal macrophage activity in pancreatic cancer. Gut 2024; 73:1684-1701. [PMID: 38834297 PMCID: PMC11420735 DOI: 10.1136/gutjnl-2023-331854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 05/15/2024] [Indexed: 06/06/2024]
Abstract
OBJECTIVE Highly malignant pancreatic ductal adenocarcinoma (PDAC) is characterised by an abundant immunosuppressive and fibrotic tumour microenvironment (TME). Future therapeutic attempts will therefore demand the targeting of tumours and stromal compartments in order to be effective. Here we investigate whether dual specificity and tyrosine phosphorylation-regulated kinase 1B (DYRK1B) fulfil these criteria and represent a promising anticancer target in PDAC. DESIGN We used transplantation and autochthonous mouse models of PDAC with either genetic Dyrk1b loss or pharmacological DYRK1B inhibition, respectively. Mechanistic interactions between tumour cells and macrophages were studied in direct or indirect co-culture experiments. Histological analyses used tissue microarrays from patients with PDAC. Additional methodological approaches included bulk mRNA sequencing (transcriptomics) and proteomics (secretomics). RESULTS We found that DYRK1B is mainly expressed by pancreatic epithelial cancer cells and modulates the influx and activity of TME-associated macrophages through effects on the cancer cells themselves as well as through the tumour secretome. Mechanistically, genetic ablation or pharmacological inhibition of DYRK1B strongly attracts tumoricidal macrophages and, in addition, downregulates the phagocytosis checkpoint and 'don't eat me' signal CD24 on cancer cells, resulting in enhanced tumour cell phagocytosis. Consequently, tumour cells lacking DYRK1B hardly expand in transplantation experiments, despite their rapid growth in culture. Furthermore, combining a small-molecule DYRK1B-directed therapy with mammalian target of rapamycin inhibition and conventional chemotherapy stalls the growth of established tumours and results in a significant extension of life span in a highly aggressive autochthonous model of PDAC. CONCLUSION In light of DYRK inhibitors currently entering clinical phase testing, our data thus provide a novel and clinically translatable approach targeting both the cancer cell compartment and its microenvironment.
Collapse
Affiliation(s)
- Anna Brichkina
- Department of Gastroenterology Endocrinology and Metabolism, Center for Tumor and Immune Biology, Marburg, Germany
- Present address: Institute of Systems Immunology, Center for Tumor and Immune Biology, Marburg, Germany
| | - Miriam Ems
- Department of Gastroenterology Endocrinology and Metabolism, Center for Tumor and Immune Biology, Marburg, Germany
| | - Roman Suezov
- Department of Gastroenterology Endocrinology and Metabolism, Center for Tumor and Immune Biology, Marburg, Germany
| | - Rajeev Singh
- Department of Gastroenterology Endocrinology and Metabolism, Center for Tumor and Immune Biology, Marburg, Germany
| | - Veronika Lutz
- Institute of Systems Immunology, Philipps-Universitat Marburg, Marburg, Hessen, Germany
| | - Felix S R Picard
- Institute of Systems Immunology, Philipps-Universitat Marburg, Marburg, Hessen, Germany
| | - Andrea Nist
- Genomics Core Facility, Philipps University Marburg, Marburg, Germany
| | - Thorsten Stiewe
- Genomics Core Facility, Philipps University Marburg, Marburg, Germany
- Institute for Molecular Oncology, German Center for Lung Research (DZL), Marburg, Germany
| | - Johannes Graumann
- Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
- Institute of Translational Proteomics, Philipps University, Marburg, Germany
| | - Michael Daude
- Medicinal Chemistry Core Facility, Philipps University Marburg, Marburg, Germany
| | - Wibke E Diederich
- Medicinal Chemistry Core Facility, Philipps University Marburg, Marburg, Germany
- Department of Medicinal chemistry, Center for Tumor and Immune Biology, Marburg, Germany
| | - Florian Finkernagel
- Bioinformatics Core Facility, Center for Tumor and Immune Biology, Marburg, Germany
| | - Ho-Ryun Chung
- Institute for Medical Bioinformatics and Biostatistics, Institute for Molecular Biology and Tumor Research, Marburg, Germany
| | - Detlef K Bartsch
- Department of Visceral, Thoracic and Vascular Surgery, Philipps-University Marburg, Marburg, Germany
| | - Katrin Roth
- Cell Imaging Core Facility, Center for Tumor Biology and Immunology, Philipps-University Marburg, Marburg, Hessen, Germany
| | - Corinna Keber
- Institute of Pathology, University Hospital of Giessen-Marburg, Marburg, Germany
| | - Carsten Denkert
- Institute of Pathology, University Hospital of Giessen-Marburg, Marburg, Germany
| | - Magdalena Huber
- Institute of Systems Immunology, Philipps-Universitat Marburg, Marburg, Hessen, Germany
| | - Thomas M Gress
- Department of Gastroenterology, Endocrinology and Metabolism, Center for Tumor and Immune Biology, Marburg, Germany
| | - Matthias Lauth
- Department of Gastroenterology Endocrinology and Metabolism, Center for Tumor and Immune Biology, Marburg, Germany
| |
Collapse
|
5
|
Liu Y, Weng L, Wang Y, Zhang J, Wu Q, Zhao P, Shi Y, Wang P, Fang L. Deciphering the role of CD47 in cancer immunotherapy. J Adv Res 2024; 63:129-158. [PMID: 39167629 PMCID: PMC11380025 DOI: 10.1016/j.jare.2023.10.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2023] [Revised: 10/05/2023] [Accepted: 10/18/2023] [Indexed: 08/23/2024] Open
Abstract
BACKGROUND Immunotherapy has emerged as a novel strategy for cancer treatment following surgery, radiotherapy, and chemotherapy. Immune checkpoint blockade and Chimeric antigen receptor (CAR)-T cell therapies have been successful in clinical trials. Cancer cells evade immune surveillance by hijacking inhibitory pathways via overexpression of checkpoint genes. The Cluster of Differentiation 47 (CD47) has emerged as a crucial checkpoint for cancer immunotherapy by working as a "don't eat me" signal and suppressing innate immune signaling. Furthermore, CD47 is highly expressed in many cancer types to protect cancer cells from phagocytosis via binding to SIRPα on phagocytes. Targeting CD47 by either interrupting the CD47-SIRPα axis or combing with other therapies has been demonstrated as an encouraging therapeutic strategy in cancer immunotherapy. Antibodies and small molecules that target CD47 have been explored in pre- and clinical trials. However, formidable challenges such as the anemia and palate aggregation cannot be avoided because of the wide presentation of CD47 on erythrocytes. AIM OF VIEW This review summarizes the current knowledge on the regulation and function of CD47, and provides a new perspective for immunotherapy targeting CD47. It also highlights the clinical progress of targeting CD47 and discusses challenges and potential strategies. KEY SCIENTIFIC CONCEPTS OF REVIEW This review provides a comprehensive understanding of targeting CD47 in cancer immunotherapy, it also augments the concept of combination immunotherapy strategies by employing both innate and adaptive immune responses.
Collapse
Affiliation(s)
- Yu'e Liu
- Tongji University Cancer Center, Shanghai Tenth People's Hospital of Tongji University, School of Medicine, Tongji University, Shanghai 200092, China
| | - Linjun Weng
- Tongji University Cancer Center, Shanghai Tenth People's Hospital of Tongji University, School of Medicine, Tongji University, Shanghai 200092, China
| | - Yanjin Wang
- Department of Nephrology, Shanghai East Hospital, Tongji University, School of Medicine, Shanghai, China
| | - Jin Zhang
- Department of Pharmacology and Toxicology, University of Mississippi, Medical Center, 39216 Jackson, MS, USA
| | - Qi Wu
- Tongji University Cancer Center, Shanghai Tenth People's Hospital of Tongji University, School of Medicine, Tongji University, Shanghai 200092, China
| | - Pengcheng Zhao
- School of Life Sciences and Medicine, Shandong University of Technology, No.266 Xincun West Road, Zibo 255000, Shandong Province, China
| | - Yufeng Shi
- Tongji University Cancer Center, Shanghai Tenth People's Hospital of Tongji University, School of Medicine, Tongji University, Shanghai 200092, China; Clinical Center for Brain and Spinal Cord Research, Tongji University, Shanghai 200092, China.
| | - Ping Wang
- Tongji University Cancer Center, Shanghai Tenth People's Hospital of Tongji University, School of Medicine, Tongji University, Shanghai 200092, China.
| | - Lan Fang
- Tongji University Cancer Center, Shanghai Tenth People's Hospital of Tongji University, School of Medicine, Tongji University, Shanghai 200092, China.
| |
Collapse
|
6
|
Wang Y, Lih TM, Lee JW, Ohtsuka T, Hozaka Y, Mino-Kenudson M, Adsay NV, Luchini C, Scarpa A, Maker AV, Kim GE, Paulino J, Chen L, Jiao L, Sun Z, Goodman D, Pflüger MJ, Roberts NJ, Matthaei H, Wood LD, Furukawa T, Zhang H, Hruban RH. Multi-omic profiling of intraductal papillary neoplasms of the pancreas reveals distinct expression patterns and potential markers of progression. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.07.602385. [PMID: 39005476 PMCID: PMC11245086 DOI: 10.1101/2024.07.07.602385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
In order to advance our understanding of precancers in the pancreas, 69 pancreatic intraductal papillary neoplasms (IPNs), including 64 intraductal papillary mucinous neoplasms (IPMNs) and 5 intraductal oncocytic papillary neoplasms (IOPNs), 32 pancreatic cyst fluid samples, 104 invasive pancreatic ductal adenocarcinomas (PDACs), 43 normal adjacent tissues (NATs), and 76 macro-dissected normal pancreatic ducts (NDs) were analyzed by mass spectrometry. A total of 10,246 proteins and 22,284 glycopeptides were identified in all tissue samples, and 756 proteins with more than 1.5-fold increase in abundance in IPMNs relative to NDs were identified, 45% of which were also identified in cyst fluids. The over-expression of selected proteins was validated by immunolabeling. Proteins and glycoproteins overexpressed in IPMNs included those involved in glycan biosynthesis and the immune system. In addition, multiomics clustering identified two subtypes of IPMNs. This study provides a foundation for understanding tumor progression and targets for earlier detection and therapies. Significance This multilevel characterization of intraductal papillary neoplasms of the pancreas provides a foundation for understanding the changes in protein and glycoprotein expression during the progression from normal duct to intraductal papillary neoplasm, and to invasive pancreatic carcinoma, providing a foundation for informed approaches to earlier detection and treatment.
Collapse
|
7
|
Liu J, Jiang Y, Chen L, Qian Z, Zhang Y. Associations between HIFs and tumor immune checkpoints: mechanism and therapy. Discov Oncol 2024; 15:2. [PMID: 38165484 PMCID: PMC10761656 DOI: 10.1007/s12672-023-00836-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Accepted: 11/21/2023] [Indexed: 01/03/2024] Open
Abstract
Hypoxia, which activates a variety of signaling pathways to enhance tumor cell growth and metabolism, is among the primary features of tumor cells. Hypoxia-inducible factors (HIFs) have a substantial impact on a variety of facets of tumor biology, such as epithelial-mesenchymal transition, metabolic reprogramming, angiogenesis, and improved radiation resistance. HIFs induce hypoxia-adaptive responses in tumor cells. Many academics have presented preclinical and clinical research targeting HIFs in tumor therapy, highlighting the potential applicability of targeted HIFs. In recent years, the discovery of numerous pharmacological drugs targeting the regulatory mechanisms of HIFs has garnered substantial attention. Additionally, HIF inhibitors have attained positive results when used in conjunction with traditional oncology radiation and/or chemotherapy, as well as with the very promising addition of tumor immunotherapy. Immune checkpoint inhibitors (CPIs), which are employed in a range of cancer treatments over the past decades, are essential in tumor immunotherapy. Nevertheless, the use of immunotherapy has been severely hampered by tumor resistance and treatment-related toxicity. According to research, HIF inhibitors paired with CPIs may be game changers for multiple malignancies, decreasing malignant cell plasticity and cancer therapy resistance, among other things, and opening up substantial new pathways for immunotherapy drug development. The structure, activation mechanisms, and pharmacological sites of action of the HIF family are briefly reviewed in this work. This review further explores the interactions between HIF inhibitors and other tumor immunotherapy components and covers the potential clinical use of HIF inhibitors in combination with CPIs.
Collapse
Affiliation(s)
- Jiayu Liu
- Department of Oncology, Wuxi Maternal and Child Health Hospital, Wuxi School of Medicine, Jiangnan University, Wuxi, 214002, Jiangsu, China
| | - Ying Jiang
- Department of Oncology, Wuxi Maternal and Child Health Hospital, Wuxi School of Medicine, Jiangnan University, Wuxi, 214002, Jiangsu, China
| | - Lingyan Chen
- Wuxi Maternal and Child Health Hospital, Nanjing Medical University, Nanjing, 214000, Jiangsu, China
| | - Zhiwen Qian
- Wuxi Maternal and Child Health Hospital, Nanjing Medical University, Nanjing, 214000, Jiangsu, China
| | - Yan Zhang
- Department of Oncology, Wuxi Maternal and Child Health Hospital, Wuxi School of Medicine, Jiangnan University, Wuxi, 214002, Jiangsu, China.
- Wuxi Maternal and Child Health Hospital, Nanjing Medical University, Nanjing, 214000, Jiangsu, China.
| |
Collapse
|
8
|
Matsuoka T, Sugiyama A, Miyawaki Y, Hidaka Y, Okuno Y, Sakai H, Tanaka H, Yoshikawa K, Fukui T, Mizuno K, Sumiyoshi T, Goto T, Inoue T, Akamatsu S, Kobayashi T, Nakamura E. Newly developed preclinical models reveal broad-spectrum CDK inhibitors as potent drugs for CRPC exhibiting primary resistance to enzalutamide. Cancer Sci 2024; 115:283-297. [PMID: 37923364 PMCID: PMC10823279 DOI: 10.1111/cas.15984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 08/30/2023] [Accepted: 09/14/2023] [Indexed: 11/07/2023] Open
Abstract
Androgen-deprivation therapy is a standard treatment for advanced prostate cancer. However, most patients eventually acquire resistance and progress to castration-resistant prostate cancer (CRPC). In this study, we established new CRPC cell lines, AILNCaP14 and AILNCaP15, from LNCaP cells under androgen-deprived conditions. Unlike most pre-existing CRPC cell lines, both cell lines expressed higher levels of androgen receptor (AR) and prostate-specific antigen (PSA) than parental LNCaP cells. Moreover, these cells exhibited primary resistance to enzalutamide. Since AR signaling plays a significant role in the development of CRPC, PSA promoter sequences fused with GFP were introduced into AILNCaP14 cells to conduct GFP fluorescence-based chemical screening. We identified flavopiridol, a broad-spectrum CDK inhibitor, as a candidate drug that could repress AR transactivation of CRPC cells, presumably through the inhibition of phosphorylation of AR on the serine 81 residue (pARSer81 ). Importantly, this broad-spectrum CDK inhibitor inhibited the proliferation of AILNCaP14 cells both in vitro and in vivo. Moreover, a newly developed liver metastatic model using AILNCaP15 cells revealed that the compound attenuated tumor growth of CRPC harboring highly metastatic properties. Finally, we developed a patient-derived xenograft (PDX) model of CRPC and DCaP CR from a patient presenting therapeutic resistance to enzalutamide, abiraterone, and docetaxel. Flavopiridol successfully suppressed the tumor growth of CRPC in this PDX model. Since ARSer81 was found to be phosphorylated in clinical CRPC samples, our data suggested that broad-spectrum CDK inhibitors might be a potent candidate drug for the treatment of CRPC, including those exhibiting primary resistance to enzalutamide.
Collapse
Grants
- 15K21115 Ministry of Education, Culture, Sports, Science and Technology
- 16K15686 Ministry of Education, Culture, Sports, Science and Technology
- 20K18112 Ministry of Education, Culture, Sports, Science and Technology
- 26670700 Ministry of Education, Culture, Sports, Science and Technology
- Ministry of Education, Culture, Sports, Science and Technology
Collapse
Affiliation(s)
- Takashi Matsuoka
- Department of UrologyKyoto University Graduate School of MedicineKyotoJapan
| | - Aiko Sugiyama
- DSK Project, Medical Innovation CenterKyoto University Graduate School of MedicineKyotoJapan
| | - Yoshifumi Miyawaki
- DSK Project, Medical Innovation CenterKyoto University Graduate School of MedicineKyotoJapan
| | - Yusuke Hidaka
- DSK Project, Medical Innovation CenterKyoto University Graduate School of MedicineKyotoJapan
| | - Yukiko Okuno
- Medical Research Support Center, Graduate School of MedicineKyoto UniversityKyotoJapan
| | - Hiroaki Sakai
- DSK Project, Medical Innovation CenterKyoto University Graduate School of MedicineKyotoJapan
| | - Hiroki Tanaka
- DSK Project, Medical Innovation CenterKyoto University Graduate School of MedicineKyotoJapan
| | - Kiyotsugu Yoshikawa
- Laboratory of Pharmacotherapy, Department of Clinical Pharmacy, Faculty of Pharmaceutical SciencesDoshisha Women's College of Liberal ArtsKyotoJapan
| | - Tomohiro Fukui
- Department of UrologyKyoto University Graduate School of MedicineKyotoJapan
| | - Kei Mizuno
- Department of UrologyKyoto University Graduate School of MedicineKyotoJapan
| | - Takayuki Sumiyoshi
- Department of UrologyKyoto University Graduate School of MedicineKyotoJapan
| | - Takayuki Goto
- Department of UrologyKyoto University Graduate School of MedicineKyotoJapan
| | - Takahiro Inoue
- Department of Nephro‐Urologic Surgery and AndrologyMie University Graduate School of MedicineTsuJapan
| | - Shusuke Akamatsu
- Department of UrologyKyoto University Graduate School of MedicineKyotoJapan
| | - Takashi Kobayashi
- Department of UrologyKyoto University Graduate School of MedicineKyotoJapan
| | - Eijiro Nakamura
- Department of UrologyNational Cancer Center HospitalTokyoJapan
| |
Collapse
|
9
|
Zaarour RF, Ribeiro M, Azzarone B, Kapoor S, Chouaib S. Tumor microenvironment-induced tumor cell plasticity: relationship with hypoxic stress and impact on tumor resistance. Front Oncol 2023; 13:1222575. [PMID: 37886168 PMCID: PMC10598765 DOI: 10.3389/fonc.2023.1222575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2023] [Accepted: 09/27/2023] [Indexed: 10/28/2023] Open
Abstract
The role of tumor interaction with stromal components during carcinogenesis is crucial for the design of efficient cancer treatment approaches. It is widely admitted that tumor hypoxic stress is associated with tumor aggressiveness and thus impacts susceptibility and resistance to different types of treatments. Notable biological processes that hypoxia functions in include its regulation of tumor heterogeneity and plasticity. While hypoxia has been reported as a major player in tumor survival and dissemination regulation, the significance of hypoxia inducible factors in cancer stem cell development remains poorly understood. Several reports indicate that the emergence of cancer stem cells in addition to their phenotype and function within a hypoxic tumor microenvironment impacts cancer progression. In this respect, evidence showed that cancer stem cells are key elements of intratumoral heterogeneity and more importantly are responsible for tumor relapse and escape to treatments. This paper briefly reviews our current knowledge of the interaction between tumor hypoxic stress and its role in stemness acquisition and maintenance. Our review extensively covers the influence of hypoxia on the formation and maintenance of cancer stem cells and discusses the potential of targeting hypoxia-induced alterations in the expression and function of the so far known stem cell markers in cancer therapy approaches. We believe that a better and integrated understanding of the effect of hypoxia on stemness during carcinogenesis might lead to new strategies for exploiting hypoxia-associated pathways and their targeting in the clinical setting in order to overcome resistance mechanisms. More importantly, at the present time, efforts are oriented towards the design of innovative therapeutical approaches that specifically target cancer stem cells.
Collapse
Affiliation(s)
- RF. Zaarour
- Thumbay Research Institute for Precision Medicine, Gulf Medical University, Ajman, United Arab Emirates
| | - M. Ribeiro
- Thumbay Research Institute for Precision Medicine, Gulf Medical University, Ajman, United Arab Emirates
| | - B. Azzarone
- Tumor Immunology Unit, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - S. Kapoor
- Thumbay Research Institute for Precision Medicine, Gulf Medical University, Ajman, United Arab Emirates
| | - S. Chouaib
- Thumbay Research Institute for Precision Medicine, Gulf Medical University, Ajman, United Arab Emirates
- INSERM UMR 1186, Integrative Tumor Immunology and Immunotherapy, Gustave Roussy, Faculty of Medicine, University Paris-Saclay, Villejuif, France
| |
Collapse
|
10
|
Liu J, Wu W, Zhu Q, Zhu H. Hydrogel-Based Therapeutics for Pancreatic Ductal Adenocarcinoma Treatment. Pharmaceutics 2023; 15:2421. [PMID: 37896181 PMCID: PMC10610350 DOI: 10.3390/pharmaceutics15102421] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 09/20/2023] [Accepted: 09/28/2023] [Indexed: 10/29/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC), one of the deadliest malignancies worldwide, is characteristic of the tumor microenvironments (TME) comprising numerous fibroblasts and immunosuppressive cells. Conventional therapies for PDAC are often restricted by limited drug delivery efficiency, immunosuppressive TME, and adverse effects. Thus, effective and safe therapeutics are urgently required for PDAC treatment. In recent years, hydrogels, with their excellent biocompatibility, high drug load capacity, and sustainable release profiles, have been developed as effective drug-delivery systems, offering potential therapeutic options for PDAC. This review summarizes the distinctive features of the immunosuppressive TME of PDAC and discusses the application of hydrogel-based therapies in PDAC, with a focus on how these hydrogels remodel the TME and deliver different types of cargoes in a controlled manner. Furthermore, we also discuss potential drug candidates and the challenges and prospects for hydrogel-based therapeutics for PDAC. By providing a comprehensive overview of hydrogel-based therapeutics for PDAC treatment, this review seeks to serve as a reference for researchers and clinicians involved in developing therapeutic strategies targeting the PDAC microenvironment.
Collapse
Affiliation(s)
- Jinlu Liu
- Division of Abdominal Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China; (J.L.); (Q.Z.)
| | - Wenbi Wu
- Department of Biotherapy, Cancer Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China;
| | - Qing Zhu
- Division of Abdominal Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China; (J.L.); (Q.Z.)
| | - Hong Zhu
- Division of Abdominal Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China; (J.L.); (Q.Z.)
| |
Collapse
|
11
|
Xu J, Shi Q, Wang B, Ji T, Guo W, Ren T, Tang X. The role of tumor immune microenvironment in chordoma: promising immunotherapy strategies. Front Immunol 2023; 14:1257254. [PMID: 37720221 PMCID: PMC10502727 DOI: 10.3389/fimmu.2023.1257254] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 08/14/2023] [Indexed: 09/19/2023] Open
Abstract
Chordoma is a rare malignant bone tumor with limited therapeutic options, which is resistant to conventional chemotherapy and radiotherapy, and targeted therapy is also shown with little efficacy. The long-standing delay in researching its mechanisms of occurrence and development has resulted in the dilemma of no effective treatment targets and no available drugs in clinical practice. In recent years, the role of the tumor immune microenvironment in driving tumor growth has become a hot and challenging topic in the field of cancer research. Immunotherapy has shown promising results in the treatment of various tumors. However, the study of the immune microenvironment of chordoma is still in its infancy. In this review, we aim to present a comprehensive reveal of previous exploration on the chordoma immune microenvironment and propose promising immunotherapy strategies for chordoma based on these characteristics.
Collapse
Affiliation(s)
- Jiuhui Xu
- Department of Musculoskeletal Tumor, Peking University People’s Hospital, Beijing, China
- Beijing Key Laboratory of Musculoskeletal Tumor, Peking University People’s Hospital, Beijing, China
| | - Qianyu Shi
- Department of Musculoskeletal Tumor, Peking University People’s Hospital, Beijing, China
- Beijing Key Laboratory of Musculoskeletal Tumor, Peking University People’s Hospital, Beijing, China
| | - Boyang Wang
- Department of Musculoskeletal Tumor, Peking University People’s Hospital, Beijing, China
- Beijing Key Laboratory of Musculoskeletal Tumor, Peking University People’s Hospital, Beijing, China
| | - Tao Ji
- Department of Musculoskeletal Tumor, Peking University People’s Hospital, Beijing, China
- Beijing Key Laboratory of Musculoskeletal Tumor, Peking University People’s Hospital, Beijing, China
| | - Wei Guo
- Department of Musculoskeletal Tumor, Peking University People’s Hospital, Beijing, China
- Beijing Key Laboratory of Musculoskeletal Tumor, Peking University People’s Hospital, Beijing, China
| | - Tingting Ren
- Department of Musculoskeletal Tumor, Peking University People’s Hospital, Beijing, China
- Beijing Key Laboratory of Musculoskeletal Tumor, Peking University People’s Hospital, Beijing, China
| | - Xiaodong Tang
- Department of Musculoskeletal Tumor, Peking University People’s Hospital, Beijing, China
- Beijing Key Laboratory of Musculoskeletal Tumor, Peking University People’s Hospital, Beijing, China
| |
Collapse
|
12
|
Liang C, Huang W, Zhang Y, Zhang D, An S, Wu Q, Zhao H, Wang C, Huang G, Wei W, Liu J. ImmunoPET Imaging of CD47 with VHH-Derived Tracers in Pancreatic Cancers. Mol Pharm 2023; 20:4184-4195. [PMID: 37403817 DOI: 10.1021/acs.molpharmaceut.3c00311] [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: 07/06/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a malignant tumor with insidious onset, rapid progression, and a very poor prognosis. CD47 is a transmembrane protein associated with the development and poor prognosis of pancreatic cancer. The aim of this study was to evaluate the diagnostic value of novel immunoPET tracers targeting CD47 in preclinical pancreatic cancer models. The association of CD47 expression with pancreatic cancer was analyzed using the Gene Expression Profiling Interactive Analysis platform. Immunohistochemical analysis of tissue microarrays was performed to detect CD47 expression in PDAC. CD47 expression levels on BxPC-3 and AsPC-1 cell membranes were compared using flow cytometry. A VHH (C2)-targeting human CD47 and its albumin-binding derivative (ABDC2) were labeled with 68Ga or 89Zr, respectively. The developed tracers were evaluated by immuno-positron emission tomography (immunoPET) imaging in tumor-bearing nude and CD47-humanized mice. [68Ga]Ga-NOTA-C2 effectively detected tumor lesions in nude mice models and further showed confirmative imaging capacity in CD47-humanized PDAC models. Compared with [68Ga]Ga-NOTA-C2, [89Zr]Zr-DFO-ABDC2 had a significantly prolonged circulation time, increased tumor uptake, and reduced accumulation in the kidneys. Finally, biodistribution and histological staining confirmed the results of the immunoPET imaging studies. In this study, we validated that two novel VHH-derived molecular imaging tracers for immunoPET imaging ([68Ga]Ga-NOTA-C2 and [89Zr]Zr-DFO-ABDC2) can effectively annotate CD47 expression and diagnose PDAC in a target-specific manner. Clinical application of the imaging strategies may help select patients for CD47-targeted therapies and assess the response thereafter.
Collapse
Affiliation(s)
- Chenyi Liang
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Wei Huang
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - You Zhang
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Di Zhang
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Shuxian An
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Qianyun Wu
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Haitao Zhao
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Cheng Wang
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Gang Huang
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Weijun Wei
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Jianjun Liu
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| |
Collapse
|
13
|
Huntoon K, Lee D, Dong S, Antony A, Kim BYS, Jiang W. Targeting phagocytosis to enhance antitumor immunity. Trends Cancer 2023; 9:650-665. [PMID: 37150626 DOI: 10.1016/j.trecan.2023.04.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 04/10/2023] [Accepted: 04/14/2023] [Indexed: 05/09/2023]
Abstract
Many patients with metastatic or treatment-resistant cancer have experienced improved outcomes after immunotherapy that targets adaptive immune checkpoints. However, innate immune checkpoints, which can hinder the detection and clearance of malignant cells, are also crucial in tumor-mediated immune escape and may also serve as targets in cancer immunotherapy. In this review, we discuss the current understanding of immune evasion by cancer cells via disruption of phagocytic clearance, and the potential effects of blocking phagocytosis checkpoints on the activation of antitumor immune responses. We propose that a more effective combination immunotherapy strategy could be to exploit tumor-intrinsic processes that inhibit key innate immune surveillance processes, such as phagocytosis, and incorporate both innate and adaptive immune responses for treating patients with cancer.
Collapse
Affiliation(s)
- Kristin Huntoon
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - DaeYong Lee
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Shiyan Dong
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Abin Antony
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Betty Y S Kim
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Wen Jiang
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
14
|
Ferencz B, Megyesfalvi Z, Csende K, Fillinger J, Poór V, Lantos A, Pipek O, Sólyom-Tisza A, Rényi-Vámos F, Schelch K, Lang C, Schwendenwein A, Boettiger K, László V, Hoetzenecker K, Döme B, Berta J. Comparative expression analysis of immune-related markers in surgically resected lung neuroendocrine neoplasms. Lung Cancer 2023; 181:107263. [PMID: 37270937 DOI: 10.1016/j.lungcan.2023.107263] [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: 01/04/2023] [Revised: 05/20/2023] [Accepted: 05/25/2023] [Indexed: 06/06/2023]
Abstract
BACKGROUND Although immunotherapy has led to a paradigm shift in the treatment of lung cancer, the therapeutic approaches for lung neuroendocrine neoplasms (LNENs) are still limited. Our aim was to explore the immunological landscape and the expression of immune checkpoint markers in LNENs. METHODS Surgically removed tumor samples of 26 atypical carcinoid (AC), 30 large cell neuroendocrine carcinoma (LCNEC) and 29 small cell lung cancer (SCLC) patients were included. The immune phenotype of each tumor type was assessed by using a panel of 15 immune-related markers. As these markers are potentially expressed by immune cells and/or tumor cells, they might serve as putative targets for immunotherapy. Expression patterns were measured by immunohistochemistry and correlated with clinicopathological parameters and prognosis. RESULTS Unsupervised hierarchical clustering revealed distinct immunologic profiles across tumor types. Specifically, AC tumors were characterized by high tumor cell CD40 expression and low levels of immune infiltrates whereas SCLC samples had a high CD47 and Inducible T Cell Costimulator (ICOS) expression in tumor cells and immune cells, respectively. High CD70 and CD137 expression by tumor cells as well as elevated expression of CD27, Lymphocyte Activation Gene 3 (LAG3), and CD40 by immune cells were characteristic for LCNEC samples. Overall, SCLC and LCNEC tumors had a more immunogenic phenotype than AC samples. High tumor cell CD47 and CD40 expressions were associated with impaired and improved survival outcomes, respectively. CONCLUSIONS By providing insights into the widely divergent immunologic profiles of LNENs, our results might serve as a basis for the development of novel immunotherapy-related approaches in these devastating malignancies.
Collapse
Affiliation(s)
- Bence Ferencz
- Department of Thoracic Surgery, Semmelweis University and National Institute of Oncology, Budapest, Hungary; National Korányi Institute of Pulmonology, Budapest, Hungary
| | - Zsolt Megyesfalvi
- Department of Thoracic Surgery, Semmelweis University and National Institute of Oncology, Budapest, Hungary; National Korányi Institute of Pulmonology, Budapest, Hungary; Department of Thoracic Surgery, Comprehensive Cancer Center Vienna, Medical University of Vienna, Vienna, Austria.
| | - Kristóf Csende
- Department of Thoracic Surgery, Semmelweis University and National Institute of Oncology, Budapest, Hungary
| | - János Fillinger
- National Korányi Institute of Pulmonology, Budapest, Hungary
| | - Valentin Poór
- Department of Thoracic Surgery, Semmelweis University and National Institute of Oncology, Budapest, Hungary
| | - András Lantos
- National Korányi Institute of Pulmonology, Budapest, Hungary
| | - Orsolya Pipek
- Department of Physics of Complex Systems, Eötvös Loránd University, Budapest, Hungary
| | | | - Ferenc Rényi-Vámos
- Department of Thoracic Surgery, Semmelweis University and National Institute of Oncology, Budapest, Hungary; National Korányi Institute of Pulmonology, Budapest, Hungary
| | - Karin Schelch
- Department of Thoracic Surgery, Comprehensive Cancer Center Vienna, Medical University of Vienna, Vienna, Austria; Center for Cancer Research, Medical University of Vienna, Vienna, Austria
| | - Christian Lang
- Department of Thoracic Surgery, Comprehensive Cancer Center Vienna, Medical University of Vienna, Vienna, Austria; Division of Pulmonology, Department of Medicine II, Medical University of Vienna, Vienna, Austria
| | - Anna Schwendenwein
- Department of Thoracic Surgery, Comprehensive Cancer Center Vienna, Medical University of Vienna, Vienna, Austria
| | - Kristiina Boettiger
- Department of Thoracic Surgery, Comprehensive Cancer Center Vienna, Medical University of Vienna, Vienna, Austria
| | - Viktória László
- National Korányi Institute of Pulmonology, Budapest, Hungary; Department of Thoracic Surgery, Comprehensive Cancer Center Vienna, Medical University of Vienna, Vienna, Austria
| | - Konrad Hoetzenecker
- Department of Thoracic Surgery, Comprehensive Cancer Center Vienna, Medical University of Vienna, Vienna, Austria
| | - Balázs Döme
- Department of Thoracic Surgery, Semmelweis University and National Institute of Oncology, Budapest, Hungary; National Korányi Institute of Pulmonology, Budapest, Hungary; Department of Thoracic Surgery, Comprehensive Cancer Center Vienna, Medical University of Vienna, Vienna, Austria; Department of Translational Medicine, Lund University, Lund, Sweden.
| | - Judit Berta
- National Korányi Institute of Pulmonology, Budapest, Hungary
| |
Collapse
|
15
|
Liu T, Zhu M, Chang X, Tang X, Yuan P, Tian R, Zhu Z, Zhang Y, Chen X. Tumor-Specific Photothermal-Therapy-Assisted Immunomodulation via Multiresponsive Adjuvant Nanoparticles. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2300086. [PMID: 36782382 DOI: 10.1002/adma.202300086] [Citation(s) in RCA: 25] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 02/05/2023] [Indexed: 05/05/2023]
Abstract
Multiresponsive adjuvant nanoparticles (RMmAGL) are fabricated to perform tumor-specific photothermal therapy while regulating the behavior of tumor-associated immune cells for primary tumor eradication and metastasis inhibition. Core-satellite-like RMmAGL have a core of mannose-functionalized mesoporous silica nanoparticles loaded with the TLR7 agonist imiquimod (R837@MSN-mannose) connected via hydrazone bonds to satellites of glutamine (Glu)- and lysine (Lys)-comodified gold nanoparticles (AuNPs-Glu/Lys). During therapy, the acidic environment in tumor tissue cleaves the hydrazone bonds to release AuNPs-Glu/Lys, which further accumulate in tumor cells. After internalization, photothermal agents (aggregated AuNPs-Glu/Lys) are generated in situ through the intratumoral enzyme-catalyzed reaction between Glu and Lys, resulting in tumor-specific photothermal therapy. The detachment of AuNPs-Glu/Lys also triggers the release of R837, which matured dendritic cells (DCs) via a vaccine-like mechanism along with the tumor-associated antigens generated by photothermal therapy. These matured DCs further activates surrounding T cells for immunotherapy. Moreover, the resulting free MSN-mannose serves as an artificial glycocalyx to continuously induce the polarization of tumor-associated macrophages from an immunosuppressive phenotype to an inflammatory phenotype, thus further enhancing immunotherapy. Both in vivo and in vitro experiments demonstrate significant inhibition of malignant tumors after therapy.
Collapse
Affiliation(s)
- Tao Liu
- School of Chemical Engineering and Technology, Shaanxi Key Laboratory of Energy Chemical Process Intensification, Institute of Polymer Science in Chemical Engineering, Xi'an Jiaotong University, Xi'an, 710049, P. R. China
| | - Man Zhu
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, P. R. China
| | - Xiaowei Chang
- School of Chemical Engineering and Technology, Shaanxi Key Laboratory of Energy Chemical Process Intensification, Institute of Polymer Science in Chemical Engineering, Xi'an Jiaotong University, Xi'an, 710049, P. R. China
| | - Xiaoyu Tang
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, P. R. China
| | - Pingyun Yuan
- School of Chemical Engineering and Technology, Shaanxi Key Laboratory of Energy Chemical Process Intensification, Institute of Polymer Science in Chemical Engineering, Xi'an Jiaotong University, Xi'an, 710049, P. R. China
| | - Ran Tian
- School of Chemical Engineering and Technology, Shaanxi Key Laboratory of Energy Chemical Process Intensification, Institute of Polymer Science in Chemical Engineering, Xi'an Jiaotong University, Xi'an, 710049, P. R. China
| | - Zeren Zhu
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, P. R. China
| | - Yanmin Zhang
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, P. R. China
| | - Xin Chen
- School of Chemical Engineering and Technology, Shaanxi Key Laboratory of Energy Chemical Process Intensification, Institute of Polymer Science in Chemical Engineering, Xi'an Jiaotong University, Xi'an, 710049, P. R. China
| |
Collapse
|
16
|
Wang F, Pu K, Li J. Activating Nanomedicines with Electromagnetic Energy for Deep-Tissue Induction of Immunogenic Cell Death in Cancer Immunotherapy. SMALL METHODS 2023; 7:e2201083. [PMID: 36316270 DOI: 10.1002/smtd.202201083] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 10/12/2022] [Indexed: 05/17/2023]
Abstract
Immunotherapy is an attractive approach for cancer therapy, while its antitumor efficacy is still limited, especially for non-immunogenic tumors. Nanomedicines can be utilized to convert the non-immunogenic "cold" tumors to immunogenic "hot" tumors via inducing immunogenic cell death (ICD), thereby promoting the antitumor immune response. Some nanomedicines that can produce local heat and reactive oxygen species upon the stimulation of electromagnetic energy are the main candidates for inducing the ICD effect. However, their applications are often restricted due to the poor tissue penetration depths of electromagnetic energy, such as light. By contrast, ultrasound, X-ray, alternating magnetic field, and microwave show excellent tissue penetration depths and thereby can be used for sonodynamic therapy, radiotherapy, magnetic hyperthermia therapy, and microwave ablation therapy, all of which can effectively induce ICD. Herein, the combination of deep-tissue electromagnetic energy with nanomedicines for inducing ICD and cancer immunotherapy are summarized. In particular, the designs of nanomedicines to amplify ICD effect in the presence of deep-tissue electromagnetic energy and sensitize tumors to various immunotherapies will be discussed. At the end of this review, a brief conclusion and discussion of current challenges and further perspectives in this subfield are provided.
Collapse
Affiliation(s)
- Fengshuo Wang
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Biological Science and Medical Engineering, Donghua University, Shanghai, 201620, China
| | - Kanyi Pu
- School of Chemistry, Chemical Engineering and Biotechnology, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, 637457, Singapore
| | - Jingchao Li
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Biological Science and Medical Engineering, Donghua University, Shanghai, 201620, China
| |
Collapse
|
17
|
Emerging phagocytosis checkpoints in cancer immunotherapy. Signal Transduct Target Ther 2023; 8:104. [PMID: 36882399 PMCID: PMC9990587 DOI: 10.1038/s41392-023-01365-z] [Citation(s) in RCA: 78] [Impact Index Per Article: 78.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 01/31/2023] [Accepted: 02/14/2023] [Indexed: 03/09/2023] Open
Abstract
Cancer immunotherapy, mainly including immune checkpoints-targeted therapy and the adoptive transfer of engineered immune cells, has revolutionized the oncology landscape as it utilizes patients' own immune systems in combating the cancer cells. Cancer cells escape immune surveillance by hijacking the corresponding inhibitory pathways via overexpressing checkpoint genes. Phagocytosis checkpoints, such as CD47, CD24, MHC-I, PD-L1, STC-1 and GD2, have emerged as essential checkpoints for cancer immunotherapy by functioning as "don't eat me" signals or interacting with "eat me" signals to suppress immune responses. Phagocytosis checkpoints link innate immunity and adaptive immunity in cancer immunotherapy. Genetic ablation of these phagocytosis checkpoints, as well as blockade of their signaling pathways, robustly augments phagocytosis and reduces tumor size. Among all phagocytosis checkpoints, CD47 is the most thoroughly studied and has emerged as a rising star among targets for cancer treatment. CD47-targeting antibodies and inhibitors have been investigated in various preclinical and clinical trials. However, anemia and thrombocytopenia appear to be formidable challenges since CD47 is ubiquitously expressed on erythrocytes. Here, we review the reported phagocytosis checkpoints by discussing their mechanisms and functions in cancer immunotherapy, highlight clinical progress in targeting these checkpoints and discuss challenges and potential solutions to smooth the way for combination immunotherapeutic strategies that involve both innate and adaptive immune responses.
Collapse
|
18
|
Abstract
C-Myc overexpression is a common finding in pancreatic cancer and predicts the aggressive behavior of cancer cells. It binds to the promoter of different genes, thereby regulating their transcription. C-Myc is downstream of KRAS and interacts with several oncogenic and proliferative pathways in pancreatic cancer. C-Myc enhances aerobic glycolysis in cancer cells and regulates glutamate biosynthesis from glutamine. It provides enough energy for cancer cells' metabolism and sufficient substrate for the synthesis of organic molecules. C-Myc overexpression is associated with chemoresistance, intra-tumor angiogenesis, epithelial-mesenchymal transition (EMT), and metastasis in pancreatic cancer. Despite its title, c-Myc is not "undruggable" and recent studies unveiled that it can be targeted, directly or indirectly. Small molecules that accelerate c-Myc ubiquitination and degradation have been effective in preclinical studies. Small molecules that hinder c-Myc-MAX heterodimerization or c-Myc/MAX/DNA complex formation can functionally inhibit c-Myc. In addition, c-Myc can be targeted through transcriptional, post-transcriptional, and translational modifications.
Collapse
Affiliation(s)
- Moein Ala
- School of Medicine, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| |
Collapse
|
19
|
Gupta G, Merhej G, Saravanan S, Chen H. Cancer resistance to immunotherapy: What is the role of cancer stem cells? CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2022; 5:981-994. [PMID: 36627890 PMCID: PMC9771758 DOI: 10.20517/cdr.2022.19] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 08/08/2022] [Accepted: 09/19/2022] [Indexed: 11/06/2022]
Abstract
Immunotherapy is an emerging form of cancer therapy that is associated with promising outcomes. However, most cancer patients either do not respond to immunotherapy or develop resistance to treatment. The resistance to immunotherapy is poorly understood compared to chemotherapy and radiotherapy. Since immunotherapy targets cells within the tumor microenvironment, understanding the behavior and interactions of different cells within that environment is essential to adequately understand both therapy options and therapy resistance. This review focuses on reviewing and analyzing the special features of cancer stem cells (CSCs), which we believe may contribute to cancer resistance to immunotherapy. The mechanisms are classified into three main categories: mechanisms related to surface markers which are differentially expressed on CSCs and help CSCs escape from immune surveillance and immune cells killing; mechanisms related to CSC-released cytokines which can recruit immune cells and tame hostile immune responses; and mechanisms related to CSC metabolites which modulate the activities of infiltrated immune cells in the tumor microenvironment. This review also discusses progress made in targeting CSCs with immunotherapy and the prospect of developing novel cancer therapies.
Collapse
Affiliation(s)
| | | | | | - Hexin Chen
- Correspondence to: Dr. Hexin Chen, Department of Biological Science, University of South Carolina, 715 Sumter Street, PSC621, Columbia, SC 29205, USA. E-mail:
| |
Collapse
|
20
|
Cheng K, Cai N, Zhu J, Yang X, Liang H, Zhang W. Tumor-associated macrophages in liver cancer: From mechanisms to therapy. CANCER COMMUNICATIONS (LONDON, ENGLAND) 2022; 42:1112-1140. [PMID: 36069342 DOI: 10.1002/cac2.12345] [Citation(s) in RCA: 121] [Impact Index Per Article: 60.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/09/2022] [Revised: 06/28/2022] [Accepted: 07/26/2022] [Indexed: 12/19/2022]
Abstract
Multidimensional analyses have demonstrated the presence of a unique tumor microenvironment (TME) in liver cancer. Tumor-associated macrophages (TAMs) are among the most abundant immune cells infiltrating the TME and are present at all stages of liver cancer progression, and targeting TAMs has become one of the most favored immunotherapy strategies. In addition, macrophages and liver cancer cells have distinct origins. At the early stage of liver cancer, macrophages can provide a niche for the maintenance of liver cancer stem cells. In contrast, cancer stem cells (CSCs) or poorly differentiated tumor cells are key factors modulating macrophage activation. In the present review, we first propose the origin connection between precursor macrophages and liver cancer cells. Macrophages undergo dynamic phenotypic transition during carcinogenesis. In this course of such transition, it is critical to determine the appropriate timing for therapy and block specific markers to suppress pro-tumoral TAMs. The present review provides a more detailed discussion of transition trends of such surface markers than previous reviews. Complex crosstalk occurs between TAMs and liver cancer cells. TAMs play indispensable roles in tumor progression, angiogenesis, and autophagy due to their heterogeneity and robust plasticity. In addition, macrophages in the TME interact with other immune cells by directing cell-to-cell contact or secreting various effector molecules. Similarly, tumor cells combined with other immune cells can drive macrophage recruitment and polarization. Despite the latest achievements and the advancements in treatment strategies following TAMs studies, comprehensive discussions on the communication between macrophages and cancer cells or immune cells in liver cancer are currently lacking. In this review, we discussed the interactions between TAMs and liver cancer cells (from cell origin to maturation), the latest therapeutic strategies (including chimeric antigen receptor macrophages), and critical clinical trials for hepatocellular carcinoma (HCC) and intrahepatic cholangiocarcinoma (iCCA) to provide a rationale for further clinical investigation of TAMs as a potential target for treating patients with liver cancer.
Collapse
Affiliation(s)
- Kun Cheng
- Hepatic Surgery Centre, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, P. R. China
| | - Ning Cai
- Hepatic Surgery Centre, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, P. R. China
| | - Jinghan Zhu
- Hepatic Surgery Centre, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, P. R. China
| | - Xing Yang
- Hepatic Surgery Centre, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, P. R. China
| | - Huifang Liang
- Hepatic Surgery Centre, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, P. R. China
| | - Wanguang Zhang
- Hepatic Surgery Centre, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, P. R. China
| |
Collapse
|
21
|
Janji B, Chouaib S. The Promise of Targeting Hypoxia to Improve Cancer Immunotherapy: Mirage or Reality? Front Immunol 2022; 13:880810. [PMID: 35795658 PMCID: PMC9251545 DOI: 10.3389/fimmu.2022.880810] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Accepted: 05/10/2022] [Indexed: 01/02/2023] Open
Abstract
Almost all solid tumors display hypoxic areas in the tumor microenvironment associated with therapeutic failure. It is now well established that the abnormal growth of malignant solid tumors exacerbates their susceptibility to hypoxia. Therefore, targeting hypoxia remains an attractive strategy to sensitize tumors to various therapies. Tumor cell adaptions to hypoxia are primarily mediated by hypoxia-inducible factor-1 alpha (HIF-1α). Sensing hypoxia by HIF-1α impairs the apoptotic potential of tumor cells, thus increasing their proliferative capacity and contributing to the development of a chaotic vasculature in the tumor microenvironment. Therefore, in addition to the negative impact of hypoxia on tumor response to chemo- and radio-therapies, hypoxia has also been described as a major hijacker of the tumor response by impairing the tumor cell susceptibility to immune cell killing. This review is not intended to provide a comprehensive overview of the work published by several groups on the multiple mechanisms by which hypoxia impairs the anti-tumor immunity and establishes the immunosuppressive tumor microenvironment. There are several excellent reviews highlighting the value of targeting hypoxia to improve the benefit of immunotherapy. Here, we first provide a brief overview of the mechanisms involved in the establishment of hypoxic stress in the tumor microenvironment. We then discuss our recently published data on how targeting hypoxia, by deleting a critical domain in HIF-1α, contributes to the improvement of the anti-tumor immune response. Our aim is to support the current dogma about the relevance of targeting hypoxia in cancer immunotherapy.
Collapse
Affiliation(s)
- Bassam Janji
- Tumor Immunotherapy and Microenvironment (TIME) group, Department of Cancer Research. Luxembourg Institute of Health (LIH), Luxembourg City, Luxembourg
- *Correspondence: Salem Chouaib, ; Bassam Janji,
| | - Salem Chouaib
- Institut National de la Santé et de la Recherche Médicale (INSERM) Unités Mixtes de Recherche (UMR) 1186, Integrative Tumor Immunology and Genetic Oncology, Gustave Roussy, Villejuif, France
- Thumbay Research Institute of Precision Medicine, Gulf Medical University, Ajman, United Arab Emirates
- *Correspondence: Salem Chouaib, ; Bassam Janji,
| |
Collapse
|
22
|
Agostini A, Orlacchio A, Carbone C, Guerriero I. Understanding Tricky Cellular and Molecular Interactions in Pancreatic Tumor Microenvironment: New Food for Thought. Front Immunol 2022; 13:876291. [PMID: 35711414 PMCID: PMC9193393 DOI: 10.3389/fimmu.2022.876291] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 04/29/2022] [Indexed: 12/16/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) represents 90% of all pancreatic cancer cases and shows a high mortality rate among all solid tumors. PDAC is often associated with poor prognosis, due to the late diagnosis that leads to metastasis development, and limited efficacy of available treatments. The tumor microenvironment (TME) represents a reliable source of novel targets for therapy, and even if many of the biological interactions among stromal, immune, and cancer cells that populate the TME have been studied, much more needs to be clarified. The great limitation in the efficacy of current standard chemoterapy is due to both the dense fibrotic inaccessible TME barrier surrounding cancer cells and the immunological evolution from a tumor-suppressor to an immunosuppressive environment. Nevertheless, combinatorial therapies may prove more effective at overcoming resistance mechanisms and achieving tumor cell killing. To achieve this result, a deeper understanding of the pathological mechanisms driving tumor progression and immune escape is required in order to design rationale-based therapeutic strategies. This review aims to summarize the present knowledge about cellular interactions in the TME, with much attention on immunosuppressive functioning and a specific focus on extracellular matrix (ECM) contribution.
Collapse
Affiliation(s)
- Antonio Agostini
- Medical Oncology, Department of Medical and Surgical Sciences, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
- Medical Oncology, Department of Translational Medicine, Catholic University of the Sacred Heart, Rome, Italy
| | - Arturo Orlacchio
- NYU Grossman School of Medicine, NYU Langone Health, New York, NY, United States
| | - Carmine Carbone
- Medical Oncology, Department of Medical and Surgical Sciences, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Ilaria Guerriero
- Biogem, Biology and Molecular Genetics Institute, Ariano Irpino, Italy
| |
Collapse
|
23
|
Alausa A, Lawal KA, Babatunde OA, Obiwulu ENO, Oladokun OC, Fadahunsi OS, Celestine UO, Moses EU, Rejoice AI, Adegbola PI. Overcoming Immunotherapeutic Resistance in PDAC: SIRPα-CD47 blockade. Pharmacol Res 2022; 181:106264. [PMID: 35597384 DOI: 10.1016/j.phrs.2022.106264] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 05/15/2022] [Indexed: 11/25/2022]
Abstract
A daily increase in the number of new cases of pancreatic ductal adenocarcinoma remains an issue of contention in cancer research. The data revealed that a global cumulated case of about 500, 000 have been reported. This has made PDAC the fourteenth most occurring tumor case in cancer research. Furthermore, PDAC is responsible for about 466,003 deaths annually, representing the seventh prevalent type of cancer mortality. PDAC has no salient symptoms in its early stages. This has exasperated several attempts to produce a perfect therapeutic agent against PDAC. Recently, immunotherapeutic research has shifted focus to the blockade of checkpoint proteins in the management and of some cancers. Investigations have centrally focused on developing therapeutic agents that could at least to a significant extent block the SIRPα-CD47 signaling cascade (a cascade which prevent phagocytosis of tumors by dendritic cells, via the deactivation of innate immunity and subsequently resulting in tumor regression) with minimal side effects. The concept on the blockade of this interaction as a possible mechanism for inhibiting the progression of PDAC is currently being debated. This review examined the structure--function activity of SIRPα-CD47 interaction while discussing in detail the mechanism of tumor resistance in PDAC. Further, this review details how the blockade of SIRPα-CD47 interaction serve as a therapeutic option in the management of PDAC.
Collapse
Affiliation(s)
- Abdullahi Alausa
- Department of Biochemistry, Ladoke Akintola University of Technology, Ogbomoso, Oyo state.
| | - Khadijat Ayodeji Lawal
- Heamtalogy and Blood Transfusion Unit, Department of Medical Laboratory Science, Ladoke Akintola University of Technology, Ogbomoso, Nigeria
| | | | - E N O Obiwulu
- Department of Chemical Science, University of Delta, Agbor, Delta State
| | | | | | - Ugwu Obiora Celestine
- Department of Pharmacology, Faculty of Pharmaceutical Sciences, Enugu State University of Science and Technology
| | | | | | | |
Collapse
|
24
|
Sally Á, McGowan R, Finn K, Moran BM. Current and Future Therapies for Pancreatic Ductal Adenocarcinoma. Cancers (Basel) 2022; 14:cancers14102417. [PMID: 35626020 PMCID: PMC9139531 DOI: 10.3390/cancers14102417] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 04/28/2022] [Accepted: 05/11/2022] [Indexed: 01/02/2023] Open
Abstract
Simple Summary Pancreatic cancer is the fourth leading cause of cancer-related mortality worldwide. The poor survival associated with this disease is due to delayed diagnosis, a lack of reliable biomarkers, and tumour resistance to treatment. Currently, surgery is the only curative treatment option, but few patients are eligible for this procedure. Developing resistance to current chemotherapies such as gemcitabine has led to a reduction in effective therapy options for patients and an urgent requirement for the development of novel therapeutic avenues. Potential success has been noted in therapeutic approaches such as synthetic lethality and immunotherapy. An array of clinical trials are currently recruiting, primarily in the area of monoclonal antibodies in combination with other therapies such as chemotherapy and immune checkpoint inhibitors. This review article aims to highlight the potential these therapies have to improve patient prognosis and survival. Abstract Pancreatic cancer is one of the leading causes of cancer-related death worldwide. This is due to delayed diagnosis and resistance to traditional chemotherapy. Delayed diagnosis is often due to the broad range of non-specific symptoms that are associated with the disease. Resistance to current chemotherapies, such as gemcitabine, develops due to genetic mutations that are either intrinsic or acquired. This has resulted in poor patient prognosis and, therefore, justifies the requirement for new targeted therapies. A synthetic lethality approach, that targets specific loss-of-function mutations in cancer cells, has shown great potential in pancreatic ductal adenocarcinoma (PDAC). Immunotherapies have also yielded promising results in the development of new treatment options, with several currently undergoing clinical trials. The utilisation of monoclonal antibodies, immune checkpoint inhibitors, adoptive cell transfer, and vaccines have shown success in several neoplasms such as breast cancer and B-cell malignancies and, therefore, could hold the same potential in PDAC treatment. These therapeutic strategies could have the potential to be at the forefront of pancreatic cancer therapy in the future. This review focuses on currently approved therapies for PDAC, the challenges associated with them, and future directions of therapy including synthetically lethal approaches, immunotherapy, and current clinical trials.
Collapse
Affiliation(s)
- Áine Sally
- Department of Analytical, Biopharmaceutical and Medical Sciences, School of Science and Computing, Atlantic Technological University Galway City, Dublin Road, H91 T8NW Galway, Ireland; (Á.S.); (R.M.); (K.F.)
| | - Ryan McGowan
- Department of Analytical, Biopharmaceutical and Medical Sciences, School of Science and Computing, Atlantic Technological University Galway City, Dublin Road, H91 T8NW Galway, Ireland; (Á.S.); (R.M.); (K.F.)
- Department of Life Sciences, School of Science, Atlantic Technological University Sligo, Ash Lane, Ballytivnan, F91 YW50 Sligo, Ireland
| | - Karen Finn
- Department of Analytical, Biopharmaceutical and Medical Sciences, School of Science and Computing, Atlantic Technological University Galway City, Dublin Road, H91 T8NW Galway, Ireland; (Á.S.); (R.M.); (K.F.)
| | - Brian Michael Moran
- Department of Analytical, Biopharmaceutical and Medical Sciences, School of Science and Computing, Atlantic Technological University Galway City, Dublin Road, H91 T8NW Galway, Ireland; (Á.S.); (R.M.); (K.F.)
- Correspondence:
| |
Collapse
|
25
|
To SKY, Tang MKS, Tong Y, Zhang J, Chan KKL, Ip PPC, Shi J, Wong AST. A Selective β-Catenin-Metadherin/CEACAM1-CCL3 Axis Mediates Metastatic Heterogeneity upon Tumor-Macrophage Interaction. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2103230. [PMID: 35403834 PMCID: PMC9165500 DOI: 10.1002/advs.202103230] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 01/31/2022] [Indexed: 05/23/2023]
Abstract
Tumor heterogeneity plays a key role in cancer relapse and metastasis, however, the distinct cellular behaviors and kinetics of interactions among different cancer cell subclones and the tumor microenvironment are poorly understood. By profiling an isogenic model that resembles spontaneous human ovarian cancer metastasis with an highly metastatic (HM) and non-metastatic (NM) tumor cell pair, one finds an upregulation of Wnt/β-catenin signaling uniquely in HM. Using humanized immunocompetent mice, one shows for the first time that activated β-catenin acts nonautonomously to modulate the immune microenvironment by enhancing infiltrating tumor-associated macrophages (TAM) at the metastatic site. Single-cell time-lapse microscopy further reveals that upon contact with macrophages, a significant subset of HM, but not NM, becomes polyploid, a phenotype pivotal for tumor aggressiveness and therapy resistance. Moreover, HM, but not NM, polarizes macrophages to a TAM phenotype. Mechanistically, β-catenin upregulates cancer cell surface metadherin, which communicates through CEACAM1 expressed on macrophages to produce CCL3. Tumor xenografts in humanized mice and clinical patient samples both corroborate the relevance of enhanced metastasis, TAM activation, and polyploidy in vivo. The results thus suggest that targeting the β-catenin-metadherin/CEACAM1-CCL3 positive feedback cascade holds great therapeutic potential to disrupt polyploidization of the cancer subclones that drive metastasis.
Collapse
Affiliation(s)
- Sally K. Y. To
- School of Biological SciencesThe University of Hong KongPokfulam RoadHong KongChina
| | - Maggie K. S. Tang
- School of Biological SciencesThe University of Hong KongPokfulam RoadHong KongChina
- Laboratory for Synthetic Chemistry and Chemical Biology Limited17W, Hong Kong Science and Technology Parks, New TerritoriesHong KongChina
| | - Yin Tong
- Department of PathologyThe University of Hong KongQueen Mary HospitalPokfulam RoadHong Kong
| | - Jiangwen Zhang
- School of Biological SciencesThe University of Hong KongPokfulam RoadHong KongChina
| | - Karen K. L. Chan
- Department of Obstetrics & GynaecologyThe University of Hong KongQueen Mary HospitalPokfulam RoadHong KongChina
| | - Philip P. C. Ip
- Department of PathologyThe University of Hong KongQueen Mary HospitalPokfulam RoadHong Kong
| | - Jue Shi
- Centre for Quantitative Systems Biology and Department of PhysicsHong Kong Baptist UniversityHong KongChina
| | - Alice S. T. Wong
- School of Biological SciencesThe University of Hong KongPokfulam RoadHong KongChina
| |
Collapse
|
26
|
Lu Y, Gong Y, Zhu X, Dong X, Zhu D, Ma G. Design of Light-Activated Nanoplatform through Boosting "Eat Me" Signals for Improved CD47-Blocking Immunotherapy. Adv Healthc Mater 2022; 11:e2102712. [PMID: 34981660 DOI: 10.1002/adhm.202102712] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Indexed: 01/01/2023]
Abstract
Here, the authors propose a light-activated reactive oxygen species (ROS)-responsive nanoplatform that can boost immunogenic cell death (ICD) to release "eat me" signals, and improve CD47-blocking immunotherapy by tumor-targeted codelivery of photosensitizer IR820 and anti-CD47 antibody (αCD47). Human serum albumin and αCD47 are first constructed into a single nanoparticle using ROS-responsive linkers, which are further conjugated with photosensitizer IR820 via a matrix metalloproteinase-sensitive peptide as linker and then modified with poly(ethylene glycol) on the surface of the obtained nanoparticles. When exposed to the first wave of near-infrared (NIR) laser irradiation, the obtained nanoplatform (M-IR820/αCD47@NP) can generate ROS, which triggers nanoparticles dissociation and thus, facilitates the release of αCD47 and IR820. The second wave of NIR laser irradiation is subsequently used to perform phototherapy and induce ICD of tumor cells. An in vitro cellular study shows that M-IR820/αCD47@NP can stimulate dendritic cells activation while simultaneously enhancing the phagocytic activity of macrophage against tumor cells. In 4T1 tumor-bearing mice, M-IR820/αCD47@NP-mediated combination of phototherapy and CD47 blockade can effectively induce the synergistic antitumor immune responses to inhibit the growth of tumors and prevent local tumor recurrence. This work offers a promising strategy to improve the CD47-blocking immunotherapy efficacy using αCD47 nanomedicine.
Collapse
Affiliation(s)
- Yan Lu
- Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy The Tianjin Key Laboratory of Biomaterials Institute of Biomedical Engineering Peking Union Medical College & Chinese Academy of Medical Sciences 236# Baidi Road, Nankai District Tianjin 300192 China
| | - Yonghua Gong
- Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy The Tianjin Key Laboratory of Biomaterials Institute of Biomedical Engineering Peking Union Medical College & Chinese Academy of Medical Sciences 236# Baidi Road, Nankai District Tianjin 300192 China
| | - Xianghui Zhu
- Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy The Tianjin Key Laboratory of Biomaterials Institute of Biomedical Engineering Peking Union Medical College & Chinese Academy of Medical Sciences 236# Baidi Road, Nankai District Tianjin 300192 China
| | - Xia Dong
- Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy The Tianjin Key Laboratory of Biomaterials Institute of Biomedical Engineering Peking Union Medical College & Chinese Academy of Medical Sciences 236# Baidi Road, Nankai District Tianjin 300192 China
| | - Dunwan Zhu
- Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy The Tianjin Key Laboratory of Biomaterials Institute of Biomedical Engineering Peking Union Medical College & Chinese Academy of Medical Sciences 236# Baidi Road, Nankai District Tianjin 300192 China
| | - Guilei Ma
- Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy The Tianjin Key Laboratory of Biomaterials Institute of Biomedical Engineering Peking Union Medical College & Chinese Academy of Medical Sciences 236# Baidi Road, Nankai District Tianjin 300192 China
| |
Collapse
|
27
|
Qu T, Li B, Wang Y. Targeting CD47/SIRPα as a therapeutic strategy, where we are and where we are headed. Biomark Res 2022; 10:20. [PMID: 35418166 PMCID: PMC9009010 DOI: 10.1186/s40364-022-00373-5] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 03/31/2022] [Indexed: 02/08/2023] Open
Abstract
Immunotherapy using PD-1 and CTLA4 inhibitors to stimulate T cell immunity has achieved significant clinical success. However, only a portion of patients benefit from T cell-based immunotherapy. Macrophages, the most abundant type of innate immune cells in the body, play an important role in eliminating tumor cells and infectious microbes. The phagocytic check point protein CD47 inhibits the phagocytic activity of macrophages through binding to SIRPα expressed on macrophages. Blockade of the interaction between CD47 and SIRPα could restore phagocytic activity and eliminate tumor cells in vitro and in vivo. In this manuscript, we review the mechanism of action and development status of agents (antibodies targeting CD47 and SIRPα, SIRPα-Fc fusion proteins, and bi-specific antibodies) that block CD47/SIRPα interaction in preclinical studies and in the clinical setting. In addition, small molecules, mRNA, and CAR-T/M that target the CD47/SIRPα axis are also reviewed in this article.
Collapse
Affiliation(s)
- Tailong Qu
- College of life Science and Technology, Jinan University, No.601, West Huangpu Avenue, Guangzhou, Guangdong 510632 People’s Republic of China
- Department of Antibody Discovery, Akeso Biopharma, No.6 of Shennong Road, Torch Development District, Zhongshan, 528437 People’s Republic of China
| | - Baiyong Li
- Department of Antibody Discovery, Akeso Biopharma, No.6 of Shennong Road, Torch Development District, Zhongshan, 528437 People’s Republic of China
| | - Yifei Wang
- College of life Science and Technology, Jinan University, No.601, West Huangpu Avenue, Guangzhou, Guangdong 510632 People’s Republic of China
| |
Collapse
|
28
|
Hei Y, Chen Y, Li Q, Mei Z, Pan J, Zhang S, Xiong C, Su X, Wei S. Multifunctional Immunoliposomes Enhance the Immunotherapeutic Effects of PD-L1 Antibodies against Melanoma by Reprogramming Immunosuppressive Tumor Microenvironment. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2105118. [PMID: 34915595 DOI: 10.1002/smll.202105118] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 11/24/2021] [Indexed: 06/14/2023]
Abstract
The immunosuppressive tumor microenvironment (TME) can significantly limit the immunotherapeutic effects of the PD-L1 antibody (aPDL1) by inhibiting the infiltration of CD8+ cytotoxic T cells (CTLs) into the tumor tissues. However, how to reprogram the immunosuppressive TME and promote the infiltration of CTLs remains a huge challenge for aPDL1 to achieve the maximum benefits. Herein, the authors design a multifunctional immunoliposome that encapsulates the adrenergic receptor blocker carvedilol (CAR) and connects the "don't eat me" signal antibody (aCD47) and aPDL1 in series via a reactive oxygen species (ROS)-sensitive linker on the surface. In ROS-enriched immunosuppressive TME, the multifunctional immunoliposome (CAR@aCD47/aPDL1-SSL) can first release the outer aCD47 to block the "do not eat me" pathway, promote the phagocytosis of tumor cells by phagocytic cells, and activate CTLs. Then, the aPDL1 on the liposome surface is exposed to block the PD-1/PD-L1 signaling pathway, thereby inducing CTLs to kill tumor cells. CAR encapsulated in CAR@aCD47/aPDL1-SSL can block the adrenergic nerves in the tumor tissues and reduce their densities, thereby inhibiting angiogenesis in the tumor tissues and reprogramming the immunosuppressive TME. According to the results, CAR@aCD47/aPDL1-SSL holds an effective way to reprogram the immunosuppressive TME and significantly enhance immunotherapeutic efficiency of aPDL1 against the primary cancer and metastasis.
Collapse
Affiliation(s)
- Yu Hei
- Central Laboratory, and Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Peking University, Beijing, 100081, P. R. China
- Department of Mechanics and Engineering Science, College of Engineering, Peking University, Beijing, 100871, P. R. China
| | - Yang Chen
- Laboratory of Biomaterials and Regenerative Medicine, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100871, P. R. China
| | - Qian Li
- Laboratory of Biomaterials and Regenerative Medicine, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100871, P. R. China
| | - Zi Mei
- Central Laboratory, and Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Peking University, Beijing, 100081, P. R. China
| | - Jijia Pan
- Laboratory of Biomaterials and Regenerative Medicine, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100871, P. R. China
| | - Siqi Zhang
- Institute of molecular medicine, Peking University, Beijing, 100871, P. R. China
| | - Chunyang Xiong
- Department of Mechanics and Engineering Science, College of Engineering, Peking University, Beijing, 100871, P. R. China
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang, 325000, P. R. China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou, Zhejiang, 325001, P. R. China
| | - Xiaodong Su
- Biomedical Pioneering Innovation Center (BIOPIC), State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking University, Beijing, 100871, P. R. China
| | - Shicheng Wei
- Central Laboratory, and Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Peking University, Beijing, 100081, P. R. China
- Laboratory of Biomaterials and Regenerative Medicine, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100871, P. R. China
| |
Collapse
|
29
|
Lappano R, Todd LA, Stanic M, Cai Q, Maggiolini M, Marincola F, Pietrobon V. Multifaceted Interplay between Hormones, Growth Factors and Hypoxia in the Tumor Microenvironment. Cancers (Basel) 2022; 14:539. [PMID: 35158804 PMCID: PMC8833523 DOI: 10.3390/cancers14030539] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 01/17/2022] [Accepted: 01/18/2022] [Indexed: 02/07/2023] Open
Abstract
Hormones and growth factors (GFs) are signaling molecules implicated in the regulation of a variety of cellular processes. They play important roles in both healthy and tumor cells, where they function by binding to specific receptors on target cells and activating downstream signaling cascades. The stages of tumor progression are influenced by hormones and GF signaling. Hypoxia, a hallmark of cancer progression, contributes to tumor plasticity and heterogeneity. Most solid tumors contain a hypoxic core due to rapid cellular proliferation that outgrows the blood supply. In these circumstances, hypoxia-inducible factors (HIFs) play a central role in the adaptation of tumor cells to their new environment, dramatically reshaping their transcriptional profile. HIF signaling is modulated by a variety of factors including hormones and GFs, which activate signaling pathways that enhance tumor growth and metastatic potential and impair responses to therapy. In this review, we summarize the role of hormones and GFs during cancer onset and progression with a particular focus on hypoxia and the interplay with HIF proteins. We also discuss how hypoxia influences the efficacy of cancer immunotherapy, considering that a hypoxic environment may act as a determinant of the immune-excluded phenotype and a major hindrance to the success of adoptive cell therapies.
Collapse
Affiliation(s)
- Rosamaria Lappano
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy;
| | - Lauren A. Todd
- Department of Biology, University of Waterloo, Waterloo, ON N2L 3G1, Canada;
| | - Mia Stanic
- Department of Laboratory Medicine & Pathobiology, University of Toronto, Toronto, ON M5S 1A8, Canada;
| | - Qi Cai
- Kite Pharma Inc., Santa Monica, CA 90404, USA; (Q.C.); (F.M.)
| | - Marcello Maggiolini
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy;
| | | | | |
Collapse
|
30
|
Perfluorocarbon loaded fluorinated covalent organic polymers with effective sonosensitization and tumor hypoxia relief enable synergistic sonodynamic-immunotherapy. Biomaterials 2021; 280:121250. [PMID: 34823883 DOI: 10.1016/j.biomaterials.2021.121250] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Revised: 11/05/2021] [Accepted: 11/08/2021] [Indexed: 12/14/2022]
Abstract
Relieving tumor hypoxia has recently been found to be a promising approach to reverse tumor immunosuppression and thus enhance the treatment outcomes of diverse cancer treatments. Herein, we prepared a type of fluorinated covalent conjugate polymers (COPs) with sonosensitizer meso-5, 10, 15, 20-tetra (4-hydroxylphenyl) porphyrin (THPP) and perfluorosebacic acid (PFSEA) as cross-linkers, yielding THPPpf-COPs with efficient sonodynamic efficacy and loading capacity towards perfluoro-15-crown-5-ether (PFCE), a model perfluorocarbon molecule. Upon intratumoral injection, such PFCE@THPPpf-COPs could not only attenuate tumor hypoxia, but also exhibit the most effective suppression effect on tumor growth in the presence of ultrasound exposure by inducing immunogenic cell death of cancer cells. Furthermore, we found that the sonodynamic therapy of PFCE@THPPpf-COPs together with anti-CD47 immunotherapy would synergistically suppress tumor growth by increasing the tumor-infiltrating frequencies of phagocytic M1 macrophages and cytotoxic CD3+CD8+ T cells, while reducing the frequency of immunosuppressive regulatory T cells. Moreover, such combination treatment could also elicit potent protective memory antitumor immunity to prevent tumor challenge. Therefore, this work presents PFCE@THPPpf-COPs are a type of multifunctional nano-sonosensitizers potent in removing negative impacts of inherent tumor hypoxia and immunosuppression, and suppressing tumor growth and tumor recurrence by priming host's antitumor immunity, particularly in synergizing with anti-CD47 immunotherapy.
Collapse
|
31
|
Shi B, Chu J, Huang T, Wang X, Li Q, Gao Q, Xia Q, Luo S. The Scavenger Receptor MARCO Expressed by Tumor-Associated Macrophages Are Highly Associated With Poor Pancreatic Cancer Prognosis. Front Oncol 2021; 11:771488. [PMID: 34778091 PMCID: PMC8586414 DOI: 10.3389/fonc.2021.771488] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Accepted: 10/12/2021] [Indexed: 12/31/2022] Open
Abstract
Macrophage-targeting therapies have become attractive strategies for immunotherapy. Deficiency of MARCO significantly inhibits tumor progression and metastasis in murine models of pancreatic cancer. However, the role of MARCO in patients with pancreatic cancer remains unclear. In the present study, we analyzed tumor-associated macrophage (TAM)-related changes using the Cancer Genome Atlas database. We observed a significant enrichment of M2 macrophages in pancreatic cancer tissues. We found that several pro-tumor markers are increased in cancer tissues, including CD163, CD206, SIRPα, LILRB1, SIGLEC10, AXL, MERTK, and MARCO. Crucially, MARCO is highly or exclusively expressed in pancreatic cancer across many types of solid tumors, suggesting its significant role in pancreatic cancer. Next, we investigated the expression of MARCO in relation to the macrophage marker CD163 in a treatment-naïve pancreatic cancer cohort after surgery (n = 65). MARCO and CD163 were analyzed using immunohistochemistry. We observed increased expression of CD163 and MARCO in pancreatic cancer tissues compared with paracancerous tissues. Furthermore, we observed a large variation in CD163 and MARCO expression in pancreatic cancer tissues among cases, suggesting the heterogeneous expression of these two markers among patients. Correlation to clinical data indicated a strong trend toward worse survival for patients with high CD163 and MARCO macrophage infiltration. Moreover, high CD163 and MARCO expression negatively affected the disease-free survival and overall survival rates of patients with pancreatic cancer. Univariate and multivariate analysis revealed that CD163 and MARCO expression was an independent indicator of pancreatic cancer prognosis. In conclusion, high CD163 and MARCO expression in cancer tissues is a negative prognostic marker for pancreatic cancer after surgery. Furthermore, anti-MARCO may be a novel therapy that is worth studying in depth.
Collapse
Affiliation(s)
- Bian Shi
- Department of Integrated Traditional Chinese and Western Medicine, Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, China
| | - Junfeng Chu
- Department of Oncology, Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, China
| | - Tao Huang
- Department of Hepatopancreatobiliary Surgery, Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, China
| | - Xiaoqian Wang
- Department of Hepatopancreatobiliary Surgery, Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, China
| | - Qiujian Li
- Department of Integrated Traditional Chinese and Western Medicine, Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, China
| | - Qilong Gao
- Department of Integrated Traditional Chinese and Western Medicine, Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, China
| | - Qingxin Xia
- Department of Pathology, Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, China
| | - Suxia Luo
- Department of Oncology, Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
32
|
Kunk PR, Dougherty SC, Lynch K, Whitehair R, Meneveau M, Obeid JM, Winters K, Ju JY, Stelow EB, Bauer TW, Slingluff CL, Rahma OE. Myeloid Cell Infiltration Correlates With Prognosis in Cholangiocarcinoma and Varies Based on Tumor Location. J Immunother 2021; 44:254-263. [PMID: 34191790 PMCID: PMC8373662 DOI: 10.1097/cji.0000000000000378] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Accepted: 05/03/2021] [Indexed: 01/04/2023]
Abstract
Cholangiocarcinoma (CC) is an uncommon malignancy with increasing incidence and dismal prognosis. We conducted a comprehensive analysis of the CC tumor immune microenvironment (TIME) based on tumor location to identify therapeutic targets. We hypothesized that the TIME of CC would vary by primary tumor location and that high tumor infiltration by CD8+ T cells and low infiltration by M2 macrophages would be associated with improved survival. A retrospective analysis was conducted of 99 CC tumor samples surgically resected between 2000 and 2014. Tissue microarrays were constructed from each tumor and stained by immunohistochemistry for 24 markers of immune cells, immune activation or inhibition, programmed cell death-ligand 1, and mesothelin. Most tumors were amply infiltrated with by CD4+, CD8+, and FoxP3+ T cells, as well as by myeloid cells. Mesothelin expression ≥1+ by immunohistochemistry was found in 68% of tumors. We identified higher densities of M1 macrophages in primary distal extrahepatic CC, as well as metastatic lesions. Mesothelin expression was also significantly higher in distal extrahepatic CC. There was no association with survival of infiltration by CD4+, CD8+, or FoxP3+ T cells, mesothelin expression, or programmed cell death-ligand 1 percentage expression, however, high CD14+ myeloid cells and high CD163+ M2 macrophages were associated with worse survival. In conclusion, the CC TIME is a heterogenous milieu highly infiltrated by innate and adaptive immune cells, which differs based on primary tumor location and between primary tumors and metastatic lesions. The correlation of intratumoral M2 macrophages and myeloid cells with a worse prognosis may suggest promising immunotherapeutic targets in CC.
Collapse
Affiliation(s)
- Paul R. Kunk
- Department of Medicine, Division of Hematology-Oncology, University of Virginia Health System, Charlottesville, VA, United States
| | - Sean C. Dougherty
- Department of Medicine, Division of Hematology-Oncology, University of Virginia Health System, Charlottesville, VA, United States
| | - Kevin Lynch
- Department of Surgery, University of Virginia Health System, Charlottesville, VA, United States
| | - Rachel Whitehair
- Department of Pathology, Division of Anatomic Pathology, University of Virginia Health System, Charlottesville, VA, United States
| | - Max Meneveau
- Department of Surgery, University of Virginia Health System, Charlottesville, VA, United States
| | - Joseph M. Obeid
- Department of Surgery, Stony Brook University Hospital, Stony Brook, NY, United States
| | - Kevin Winters
- Department of Medicine, Division of Hematology-Oncology, University of Virginia Health System, Charlottesville, VA, United States
| | - Jennifer Y. Ju
- Department of Pathology, Division of Anatomic Pathology, University of Virginia Health System, Charlottesville, VA, United States
| | - Edward B. Stelow
- Department of Pathology, Division of Anatomic Pathology, University of Virginia Health System, Charlottesville, VA, United States
| | - Todd W. Bauer
- Department of Surgery, University of Virginia Health System, Charlottesville, VA, United States
| | - Craig L. Slingluff
- Department of Surgery, University of Virginia Health System, Charlottesville, VA, United States
| | - Osama E. Rahma
- Departement of Medical Oncology, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
33
|
Beizavi Z, Gheibihayat SM, Moghadasian H, Zare H, Yeganeh BS, Askari H, Vakili S, Tajbakhsh A, Savardashtaki A. The regulation of CD47-SIRPα signaling axis by microRNAs in combination with conventional cytotoxic drugs together with the help of nano-delivery: a choice for therapy? Mol Biol Rep 2021; 48:5707-5722. [PMID: 34275112 DOI: 10.1007/s11033-021-06547-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 07/02/2021] [Indexed: 12/12/2022]
Abstract
CD47, a member of the immunoglobulin superfamily, is an important "Don't Eat-Me" signal in phagocytosis process [clearance of apoptotic cells] as well as a regulator of the adaptive immune response. The lower level of CD47 on the cell surface leads to the clearance of apoptotic cells. Dysregulation of CD47 plays a critical role in the development of disorders, particularly cancers. In cancers, recognition of CD47 overexpression on the surface of cancer cells by its receptor, SIRPα on the phagocytic cells, inhibits phagocytosis of cancer cells. Thus, blocking of CD47-SIRPα signaling axis might be as a promising therapeutic target, which promotes phagocytosis of cancer cells, antigen-presenting cell function as well as adaptive T cell-mediated anti-cancer immunity. In this respect, it has been reported that CD47 expression can be regulated by microRNAs (miRNAs). MiRNAs can regulate phagocytosis of macrophages apoptotic process, drug resistance, relapse of disease, radio-sensitivity, and suppress cell proliferation, migration, and invasion through post-transcriptional regulation of CD47-SIRPα signaling axis. Moreover, the regulation of CD47 expression by miRNAs and combination with conventional cytotoxic drugs together with the help of nano-delivery represent a valuable opportunity for effective cancer treatment. In this review, we review studies that evaluate the role of miRNAs in the regulation of CD47-SIRPα in disorders to achieve a novel preventive, diagnostic, and therapeutic strategy.Please confirm if the author names are presented accurately and in the correct sequence (given name, middle name/initial, family name). Also, kindly confirm the details in the metadata are correct. Confirmed.Journal standard instruction requires a structured abstract; however, none was provided. Please supply an Abstract with subsections..Not confirmed. This is a review article. According to submission guidelines: "The abstract should be presented divided into subheadings (unless it is a mini or full review article)". Kindly check and confirm whether the corresponding authors and mail ID are correctly identified. Confirmed.
Collapse
Affiliation(s)
- Zahra Beizavi
- Department of General Surgery, Shiraz University of Medical Science, Shiraz, Iran
| | - Seyed Mohammad Gheibihayat
- Department of Medical Biotechnology, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Hadis Moghadasian
- Laboratory of Common Basic Sciences, Mohammad Rasool Allah Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Hossein Zare
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Babak Shirazi Yeganeh
- Department of Pathology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Hassan Askari
- Gastroenterohepatology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Sina Vakili
- Infertility Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Amir Tajbakhsh
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Amir Savardashtaki
- Epilepsy Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
34
|
Clinicopathological and Prognostic Significance of CD47 Expression in Lung Neuroendocrine Tumors. J Immunol Res 2021; 2021:6632249. [PMID: 34195295 PMCID: PMC8214491 DOI: 10.1155/2021/6632249] [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: 10/27/2020] [Revised: 04/09/2021] [Accepted: 05/26/2021] [Indexed: 12/22/2022] Open
Abstract
Background Lung neuroendocrine tumors account for approximately 15% of all lung cancer cases. LNET are subdivided into typical carcinoid (TC), atypical carcinoid (AC), large cell neuroendocrine carcinoma (LCNEC), and small-cell lung cancer (SCLC). The Ki-67 index has been used for decades to evaluate mitotic counts however, the role of Ki-67 as a biomarker for assessing prognosis and guiding therapy in metastatic LNET still lacks feasible clinical validation. Recent clinical trials have indicated that inhibition of CD47 with anti-CD47 antibodies exerts a promising antitumor effect against several human malignancies, including NSCLC, melanoma, and hematologic malignancies. However, the clinical relevance of CD47 expression in LNET has remained unclear. Methods We performed a retrospective study in which we analyzed tumor biopsies from 51 patients with a confirmed diagnosis of LNET that received treatment at our hospital. Then, we analyzed if there was any correlation between CD47 expression with any clinical or pathological characteristic. We also analyzed the prognostic significance of CD47, assessed as progression-free survival and overall survival. Results A total of 51 patients with LNET were enrolled in our study. The mean age at diagnosis was 57.6 (±11.6) years; 30 patients were women (59%). 27.5% of patients were positive for CD47 expression, and 72.5% of patients showed a CD47 expression of less than 1% and were considered as negatives. In patients with high-grade tumors (this time defined as Ki-67 > 40%), the positive expression of CD47 was strongly associated with an increased PFS. Albeit, these differences did not reach statistical significance when analyzing OS. Conclusion Contrary to what happens in a wide range of hematologic and solid tumors, a higher expression of CD47 in patients with LNET is associated with a better progression-free survival, especially in patients with a Ki-67 ≥ 40%. This "paradox" remains to be confirmed and explained by larger studies.
Collapse
|
35
|
Poh AR, Ernst M. Tumor-Associated Macrophages in Pancreatic Ductal Adenocarcinoma: Therapeutic Opportunities and Clinical Challenges. Cancers (Basel) 2021; 13:cancers13122860. [PMID: 34201127 PMCID: PMC8226457 DOI: 10.3390/cancers13122860] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 06/03/2021] [Accepted: 06/06/2021] [Indexed: 02/06/2023] Open
Abstract
Simple Summary Macrophages are a major component of the pancreatic tumor microenvironment, and their increased abundance is associated with poor patient survival. Given the multi-faceted role of macrophages in promoting pancreatic tumor development and progression, these cells represent promising targets for anti-cancer therapy. Abstract Pancreatic ductal adenocarcinoma (PDAC) is an aggressive malignant disease with a 5-year survival rate of less than 10%. Macrophages are one of the earliest infiltrating cells in the pancreatic tumor microenvironment, and are associated with an increased risk of disease progression, recurrence, metastasis, and shorter overall survival. Pre-clinical studies have demonstrated an unequivocal role of macrophages in PDAC by contributing to chronic inflammation, cancer cell stemness, desmoplasia, immune suppression, angiogenesis, invasion, metastasis, and drug resistance. Several macrophage-targeting therapies have also been investigated in pre-clinical models, and include macrophage depletion, inhibiting macrophage recruitment, and macrophage reprogramming. However, the effectiveness of these drugs in pre-clinical models has not always translated into clinical trials. In this review, we discuss the molecular mechanisms that underpin macrophage heterogeneity within the pancreatic tumor microenvironment, and examine the contribution of macrophages at various stages of PDAC progression. We also provide a comprehensive update of macrophage-targeting therapies that are currently undergoing clinical evaluation, and discuss clinical challenges associated with these treatment modalities in human PDAC patients.
Collapse
|
36
|
Wang C, Sun C, Li M, Xia B, Wang Y, Zhang L, Zhang Y, Wang J, Sun F, Lu S, Zhu J, Huang J, Zhang Y. Novel fully human anti-CD47 antibodies stimulate phagocytosis and promote elimination of AML cells. J Cell Physiol 2021; 236:4470-4481. [PMID: 33206395 DOI: 10.1002/jcp.30163] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 11/02/2020] [Accepted: 11/04/2020] [Indexed: 01/03/2023]
Abstract
Although most patients with acute myeloid leukemia (AML) enter remission after induction chemotherapy, the risk of relapse remains considerable. Therefore, some novel therapeutic strategies are still required. This study found that the overexpression of CD47 on AML cells was at least twofold more than that on normal bone marrow (NBM) cells in 81% (17/21) of the investigated patients; no patients had lower expression level of CD47 compared with healthy donors. The study also demonstrated that blocking the CD47/SIRPα (signal regulatory protein α) signal with the established novel fully human anti-CD47 monoclonal antibodies increased the phagocytosis of AML cells by macrophages in vitro. Furthermore, in vivo experiments showed that the novel fully human anti-CD47 monoclonal antibodies could significantly prolong the survival time of mice. Overall, the novel fully human anti-CD47 antibodies could block CD47/SIRPα interaction, increase macrophage-mediated phagocytosis, and enhance the elimination of AML cells.
Collapse
MESH Headings
- Adolescent
- Adult
- Animals
- Antibodies, Monoclonal, Humanized/pharmacology
- Antibody Specificity
- Antigens, Differentiation/metabolism
- Antineoplastic Agents, Immunological/pharmacology
- Binding Sites, Antibody
- CD47 Antigen/antagonists & inhibitors
- CD47 Antigen/immunology
- CD47 Antigen/metabolism
- Case-Control Studies
- Female
- HL-60 Cells
- Humans
- K562 Cells
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/immunology
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/pathology
- Macrophages/drug effects
- Macrophages/immunology
- Macrophages/metabolism
- Male
- Mice, Inbred C57BL
- Mice, Inbred NOD
- Mice, SCID
- Middle Aged
- Phagocytosis/drug effects
- Receptors, Immunologic/metabolism
- THP-1 Cells
- U937 Cells
- Xenograft Model Antitumor Assays
- Mice
Collapse
Affiliation(s)
- Chaoyu Wang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, China
- Department of Pediatric Oncology, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, Department of Hematology, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing, China
| | - Chengtao Sun
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, China
- Department of Pediatric Oncology, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, Department of Hematology, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Mengzhen Li
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, China
- Department of Pediatric Oncology, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, Department of Hematology, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Bing Xia
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, Department of Hematology, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Yi Wang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, China
- Department of Pediatric Oncology, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, Department of Hematology, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Li Zhang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, China
- Department of Pediatric Oncology, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, China
| | - Yanyan Zhang
- INSERM Unité Mixte de Recherche (UMR), Villejuif, France
- Université Paris-Saclay, Gustave Roussy, Villejuif, France
- Gustave Roussy, Villejuif, France
| | - Juan Wang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, China
- Department of Pediatric Oncology, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, China
| | - Feifei Sun
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, China
- Department of Pediatric Oncology, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, China
| | - Suying Lu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, China
- Department of Pediatric Oncology, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, China
| | - Jia Zhu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, China
- Department of Pediatric Oncology, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, China
| | - Junting Huang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, China
- Department of Pediatric Oncology, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, China
| | - Yizhuo Zhang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, China
- Department of Pediatric Oncology, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, Department of Hematology, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| |
Collapse
|
37
|
Kelley SM, Ravichandran KS. Putting the brakes on phagocytosis: "don't-eat-me" signaling in physiology and disease. EMBO Rep 2021; 22:e52564. [PMID: 34041845 DOI: 10.15252/embr.202152564] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 03/12/2021] [Accepted: 04/29/2021] [Indexed: 12/12/2022] Open
Abstract
Timely removal of dying or pathogenic cells by phagocytes is essential to maintaining host homeostasis. Phagocytes execute the clearance process with high fidelity while sparing healthy neighboring cells, and this process is at least partially regulated by the balance of "eat-me" and "don't-eat-me" signals expressed on the surface of host cells. Upon contact, eat-me signals activate "pro-phagocytic" receptors expressed on the phagocyte membrane and signal to promote phagocytosis. Conversely, don't-eat-me signals engage "anti-phagocytic" receptors to suppress phagocytosis. We review the current knowledge of don't-eat-me signaling in normal physiology and disease contexts where aberrant don't-eat-me signaling contributes to pathology.
Collapse
Affiliation(s)
- Shannon M Kelley
- Center for Cell Clearance, University of Virginia, Charlottesville, VA, USA.,Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA, USA
| | - Kodi S Ravichandran
- Center for Cell Clearance, University of Virginia, Charlottesville, VA, USA.,Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA, USA.,VIB-UGent Center for Inflammation Research, Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| |
Collapse
|
38
|
Yoon JH, Jung YJ, Moon SH. Immunotherapy for pancreatic cancer. World J Clin Cases 2021; 9:2969-2982. [PMID: 33969083 PMCID: PMC8080736 DOI: 10.12998/wjcc.v9.i13.2969] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 02/03/2021] [Accepted: 03/09/2021] [Indexed: 02/06/2023] Open
Abstract
Pancreatic cancer, a highly lethal cancer, has the lowest 5-year survival rate for several reasons, including its tendency for the late diagnosis, a lack of serologic markers for screening, aggressive local invasion, its early metastatic dissemination, and its resistance to chemotherapy/radiotherapy. Pancreatic cancer evades immunologic elimination by a variety of mechanisms, including induction of an immunosuppressive microenvironment. Cancer-associated fibroblasts interact with inhibitory immune cells, such as tumor-associated macrophages and regulatory T cells, to form an inflammatory shell-like desmoplastic stroma around tumor cells. Immunotherapy has the potential to mobilize the immune system to eliminate cancer cells. Nevertheless, although immunotherapy has shown brilliant results across a wide range of malignancies, only anti-programmed cell death 1 antibodies have been approved for use in patients with pancreatic cancer who test positive for microsatellite instability or mismatch repair deficiency. Some patients treated with immunotherapy who show progression based on conventional response criteria may prove to have a durable response later. Continuation of immune-based treatment beyond disease progression can be chosen if the patient is clinically stable. Immunotherapeutic approaches for pancreatic cancer treatment deserve further exploration, given the plethora of combination trials with other immunotherapeutic agents, targeted therapy, stroma-modulating agents, chemotherapy, and multi-way combination therapies.
Collapse
Affiliation(s)
- Jai Hoon Yoon
- Department of Internal Medicine, Hanyang University College of Medicine, Seoul 04763, South Korea
| | - Ye-Ji Jung
- Department of Internal Medicine, Hallym University, Anyang 14068, South Korea
| | - Sung-Hoon Moon
- Department of Internal Medicine, University of Hallym College of Medicine, Hallym University Sacred Heart Hospital, Anyang 14068, South Korea
| |
Collapse
|
39
|
Cruz-Bermúdez A, Laza-Briviesca R, Casarrubios M, Sierra-Rodero B, Provencio M. The Role of Metabolism in Tumor Immune Evasion: Novel Approaches to Improve Immunotherapy. Biomedicines 2021; 9:361. [PMID: 33807260 PMCID: PMC8067102 DOI: 10.3390/biomedicines9040361] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 03/25/2021] [Accepted: 03/28/2021] [Indexed: 12/16/2022] Open
Abstract
The tumor microenvironment exhibits altered metabolic properties as a consequence of the needs of tumor cells, the natural selection of the most adapted clones, and the selfish relationship with other cell types. Beyond its role in supporting uncontrolled tumor growth, through energy and building materials obtention, metabolism is a key element controlling tumor immune evasion. Immunotherapy has revolutionized the treatment of cancer, being the first line of treatment for multiple types of malignancies. However, many patients either do not benefit from immunotherapy or eventually relapse. In this review we overview the immunoediting process with a focus on the metabolism-related elements that are responsible for increased immune evasion, either through reduced immunogenicity or increased resistance of tumor cells to the apoptotic action of immune cells. Finally, we describe the main molecules to modulate these immune evasion processes through the control of the metabolic microenvironment as well as their clinical developmental status.
Collapse
Affiliation(s)
- Alberto Cruz-Bermúdez
- Medical Oncology Department, Health Research Institute Puerta de Hierro–Segovia de Arana (IDIPHISA) & Puerta de Hierro Hospital, Manuel de Falla Street #1, 28222 Madrid, Spain; (R.L.-B.); (M.C.); (B.S.-R.)
| | - Raquel Laza-Briviesca
- Medical Oncology Department, Health Research Institute Puerta de Hierro–Segovia de Arana (IDIPHISA) & Puerta de Hierro Hospital, Manuel de Falla Street #1, 28222 Madrid, Spain; (R.L.-B.); (M.C.); (B.S.-R.)
- PhD Programme in Molecular Biosciences, Faculty of Medicine Doctoral School, Universidad Autónoma de Madrid, 28222 Madrid, Spain
| | - Marta Casarrubios
- Medical Oncology Department, Health Research Institute Puerta de Hierro–Segovia de Arana (IDIPHISA) & Puerta de Hierro Hospital, Manuel de Falla Street #1, 28222 Madrid, Spain; (R.L.-B.); (M.C.); (B.S.-R.)
- PhD Programme in Molecular Biosciences, Faculty of Medicine Doctoral School, Universidad Autónoma de Madrid, 28222 Madrid, Spain
| | - Belén Sierra-Rodero
- Medical Oncology Department, Health Research Institute Puerta de Hierro–Segovia de Arana (IDIPHISA) & Puerta de Hierro Hospital, Manuel de Falla Street #1, 28222 Madrid, Spain; (R.L.-B.); (M.C.); (B.S.-R.)
- PhD Programme in Molecular Biosciences, Faculty of Medicine Doctoral School, Universidad Autónoma de Madrid, 28222 Madrid, Spain
| | - Mariano Provencio
- Medical Oncology Department, Health Research Institute Puerta de Hierro–Segovia de Arana (IDIPHISA) & Puerta de Hierro Hospital, Manuel de Falla Street #1, 28222 Madrid, Spain; (R.L.-B.); (M.C.); (B.S.-R.)
| |
Collapse
|
40
|
Imam R, Chang Q, Black M, Yu C, Cao W. CD47 expression and CD163 + macrophages correlated with prognosis of pancreatic neuroendocrine tumor. BMC Cancer 2021; 21:320. [PMID: 33765961 PMCID: PMC7992939 DOI: 10.1186/s12885-021-08045-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2021] [Accepted: 03/11/2021] [Indexed: 12/12/2022] Open
Abstract
Background Recent studies have suggested the important roles of CD47 and tumor-associated macrophages in the prognosis and immunotherapy of various human malignancies. However, the clinical significance of CD47 expression and CD163+ TAMs in pancreatic neuroendocrine tumor (PanNET) remains unclear. Methods In this study, 47 well-differentiated PanNET resection specimens were collected. CD47 expression and CD163+ macrophages were evaluated using immunohistochemistry and correlated with clinicopathologic properties. Results Positive CD47 staining was seen in all PanNETs as well as adjacent normal islets. Compared to normal islets, CD47 overexpressed in PanNETs (p = 0.0015). In the cohort, lymph node metastasis (LNM), lymphovascular invasion (LVI), and perineural invasion (PNI) were found in 36.2, 59.6, and 48.9% of the cases, respectively. Interestingly, PanNETs with LNM, LVI, or PNI had significantly lower H-score of CD47 than those without LNM (p = 0.035), LVI (p = 0.0005), or PNI (p = 0.0035). PanNETs in patients with disease progression (recurrence/death) also showed a significantly lower expression of CD47 than those without progression (p = 0.022). In contrast, CD163+ macrophage counts were significantly higher in cases with LNM, LVI, and PNI. Conclusions Our data suggest relative low CD47 expression and high CD163+ TAMs may act as indicators for poor prognosis of PanNETs. Supplementary Information The online version contains supplementary material available at 10.1186/s12885-021-08045-7.
Collapse
Affiliation(s)
- Rami Imam
- Department of Pathology, New York University Grossman School of Medicine, 560 1st Ave, Tisch 4-15I, New York, NY, 10016, USA
| | - Qing Chang
- Department of Pathology, New York University Grossman School of Medicine, 560 1st Ave, Tisch 4-15I, New York, NY, 10016, USA
| | - Margaret Black
- Department of Pathology, New York University Grossman School of Medicine, 560 1st Ave, Tisch 4-15I, New York, NY, 10016, USA
| | - Caroline Yu
- Department of Pathology, New York University Grossman School of Medicine, 560 1st Ave, Tisch 4-15I, New York, NY, 10016, USA
| | - Wenqing Cao
- Department of Pathology, New York University Grossman School of Medicine, 560 1st Ave, Tisch 4-15I, New York, NY, 10016, USA.
| |
Collapse
|
41
|
Xi Q, Zhang J, Yang G, Zhang L, Chen Y, Wang C, Zhang Z, Guo X, Zhao J, Xue Z, Li Y, Zhang Q, Da Y, Liu L, Yao Z, Zhang R. Restoration of miR-340 controls pancreatic cancer cell CD47 expression to promote macrophage phagocytosis and enhance antitumor immunity. J Immunother Cancer 2021; 8:jitc-2019-000253. [PMID: 32503944 PMCID: PMC7279671 DOI: 10.1136/jitc-2019-000253] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/09/2020] [Indexed: 01/15/2023] Open
Abstract
Background Immune checkpoint blockade has emerged as a potential cancer immunotherapy. The “don’t eat me” signal CD47 in cancer cells binds signal regulatory protein-α on macrophages and prevents their phagocytosis. The role of miR-340 in pancreatic ductal adenocarcinoma (PDAC), especially in tumor immunity, has not been explored. Here, we examined the clinical and biological relevance of miR-340 and the molecular pathways regulated by miR-340 in PDAC. Methods CD47 and miR-340 expression and the relationship with cancer patient survival were analyzed by bioinformatics. The mechanism of miR-340 action was explored through bioinformatics, luciferase reporter, qRT-PCR and western blot analyses. The effects of miR-340 on cancer cells were analyzed in terms of apoptosis, proliferation, migration and phagocytosis by macrophages. In vivo tumorigenesis was studied in orthotopic and subcutaneous models, and immune cells from the peripheral and tumor immune microenvironments were analyzed by flow cytometry. Depletion of macrophages was used to verify the role of macrophages in impacting the function of miR-340 in tumor progression. Results miR-340 directly regulates and inversely correlates with CD47, and it predicts patient survival in PDAC. The restoration of miR-340 expression in pancreatic cancer cells was sufficient to downregulate CD47 and promote phagocytosis of macrophages, further inhibiting tumor growth. The overexpression of miR-340 promoted macrophages to become M1-like phenotype polarized in peripheral and tumor immune microenvironments and increased T cells, especially CD8+ T cells, contributing to the antitumor effect of miR-340. Conclusions miR-340 is a key regulator of phagocytosis and antitumor immunity, and it could offer a new opportunity for immunotherapy for PDAC.
Collapse
Affiliation(s)
- Qing Xi
- Department of Immunology and Research Center of Basic Medical Sciences, Key Laboratory of Immune Microenvironment and Diseases of Educational Ministry of China, Tianjin Key Laboratory of Cellular and Molecular Immunology, Tianjin Medical University, Tianjin, China
| | - Jieyou Zhang
- Department of Immunology and Research Center of Basic Medical Sciences, Key Laboratory of Immune Microenvironment and Diseases of Educational Ministry of China, Tianjin Key Laboratory of Cellular and Molecular Immunology, Tianjin Medical University, Tianjin, China
| | - Guangze Yang
- Department of Immunology and Research Center of Basic Medical Sciences, Key Laboratory of Immune Microenvironment and Diseases of Educational Ministry of China, Tianjin Key Laboratory of Cellular and Molecular Immunology, Tianjin Medical University, Tianjin, China
| | - Lijuan Zhang
- Department of Immunology and Research Center of Basic Medical Sciences, Key Laboratory of Immune Microenvironment and Diseases of Educational Ministry of China, Tianjin Key Laboratory of Cellular and Molecular Immunology, Tianjin Medical University, Tianjin, China
| | - Ying Chen
- Guangdong Province Key Laboratory for Biotechnology Drug Candidates, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| | - Chengzhi Wang
- Department of Immunology and Research Center of Basic Medical Sciences, Key Laboratory of Immune Microenvironment and Diseases of Educational Ministry of China, Tianjin Key Laboratory of Cellular and Molecular Immunology, Tianjin Medical University, Tianjin, China
| | - Zimu Zhang
- Department of Immunology and Research Center of Basic Medical Sciences, Key Laboratory of Immune Microenvironment and Diseases of Educational Ministry of China, Tianjin Key Laboratory of Cellular and Molecular Immunology, Tianjin Medical University, Tianjin, China
| | - Xiangdong Guo
- Department of Immunology and Research Center of Basic Medical Sciences, Key Laboratory of Immune Microenvironment and Diseases of Educational Ministry of China, Tianjin Key Laboratory of Cellular and Molecular Immunology, Tianjin Medical University, Tianjin, China
| | - Jingyi Zhao
- Guangdong Province Key Laboratory for Biotechnology Drug Candidates, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| | - Zhenyi Xue
- Department of Immunology and Research Center of Basic Medical Sciences, Key Laboratory of Immune Microenvironment and Diseases of Educational Ministry of China, Tianjin Key Laboratory of Cellular and Molecular Immunology, Tianjin Medical University, Tianjin, China
| | - Yan Li
- Department of Immunology and Research Center of Basic Medical Sciences, Key Laboratory of Immune Microenvironment and Diseases of Educational Ministry of China, Tianjin Key Laboratory of Cellular and Molecular Immunology, Tianjin Medical University, Tianjin, China
| | - Qi Zhang
- Tianjin Key Laboratory of A cute Abdomen Disease Associated Organ Injury and ITCWM Repair, Institute of Integrative Medicines for Acute Abdominal Diseases, Nankai Hospital, Tianjin, China
| | - Yurong Da
- Department of Immunology and Research Center of Basic Medical Sciences, Key Laboratory of Immune Microenvironment and Diseases of Educational Ministry of China, Tianjin Key Laboratory of Cellular and Molecular Immunology, Tianjin Medical University, Tianjin, China
| | - Li Liu
- Department of Radiology, The University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Zhi Yao
- Department of Immunology and Research Center of Basic Medical Sciences, Key Laboratory of Immune Microenvironment and Diseases of Educational Ministry of China, Tianjin Key Laboratory of Cellular and Molecular Immunology, Tianjin Medical University, Tianjin, China
| | - Rongxin Zhang
- Guangdong Province Key Laboratory for Biotechnology Drug Candidates, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China; Department of Immunology and Research Center of Basic Medical Sciences, Key Laboratory of Immune Microenvironment and Diseases of Educational Ministry of China, Tianjin Key Laboratory of Cellular and Molecular Immunology, Tianjin Medical University, Tianjin, China
| |
Collapse
|
42
|
Role of targeted immunotherapy for pancreatic ductal adenocarcinoma (PDAC) treatment: An overview. Int Immunopharmacol 2021; 95:107508. [PMID: 33725635 DOI: 10.1016/j.intimp.2021.107508] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 01/18/2021] [Accepted: 02/12/2021] [Indexed: 12/15/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the deadliest solid tumors with a high mortality rate and poor survival rate. Depending on the tumor stage, PDAC is either treated by resection surgery, chemotherapies, or radiotherapies. Various chemotherapeutic agents have been used to treat PDAC, alone or in combination. Despite the combinations, chemotherapy exhibits many side-effects leading to an increase in the toxicity profile amongst the PDAC patients. Additionally, these standard chemotherapeutic agents have only a modest impact on patient survival due to their limited efficacy. PDAC was previously considered as an immunologically silent malignancy, but recent findings have demonstrated that effective immune-mediated tumor cell death can be used for its treatment. PDAC is characterized by an immunosuppressive tumor microenvironment accompanied by the major expression of myeloid-derived suppressor cells (MDSC) and M2 tumor-associated macrophages. In contrast, the expression of CD8+ T cells is significantly low. Additionally, infiltration of mast cells in PDAC correlates with the poor prognosis. Immunotherapeutic agents target the immunity mediators and empower them to suppress the tumor and effectively treat PDAC. Different targets are studied and exploited to induce an antitumor immune response in PDAC patients. In recent times, site-specific delivery of immunotherapeutics also gained attention among researchers to effectively treat PDAC. In the present review, existing immunotherapies for PDAC treatment along with their limitations are addressed in detail. The review also includes the pathophysiology, traditional strategies and significance of targeted immunotherapies to combat PDAC effectively. Separately, the identification of ideal targets for the targeted therapy of PDAC is also reviewed exhaustively. Additionally, the review also addresses the applications of targeted immunotherapeutics like checkpoint inhibitors, adoptive T-cell therapy etc.
Collapse
|
43
|
Hou Z, Pan Y, Fei Q, Lin Y, Zhou Y, Liu Y, Guan H, Yu X, Lin X, Lu F, Huang H. Prognostic significance and therapeutic potential of the immune checkpoint VISTA in pancreatic cancer. J Cancer Res Clin Oncol 2021; 147:517-531. [PMID: 33237432 PMCID: PMC7817580 DOI: 10.1007/s00432-020-03463-9] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 11/09/2020] [Indexed: 12/11/2022]
Abstract
OBJECTIVE V-domain Ig suppressor of T cell activation (VISTA) is a novel immune checkpoint protein that belongs to the B7 family. The aim of this study was to investigate the prognostic significance and therapeutic potential of VISTA in patients with pancreatic cancer. METHODS Using immunohistochemistry (IHC), we examined the expression of VISTA and demonstrated the associations between the VISTA and overall survival in 223 PDAC patients from 2 different unrelated retrospective cohorts. The multiplex immunofluorescence was performed to illuminate the relationship between VISTA expression and tumor-infiltrating immune cell subclusters of PDAC. We also verified the findings in The Cancer Genome Atlas (TCGA) dataset. The anti-tumor effect of anti-VISTA therapy was studied by the mouse model with liver metastases of PDAC. RESULTS The VISTA protein was highly expressed in 25.6% of tumor cells (TCs), 38.1% of immune cells, and 26.0% of endothelial cells in 223 PDAC tumor tissues. VISTA expression in TCs was significantly associated with prolonged overall survival. Multiplex immunofluorescence analysis revealed that VISTA level was positively correlated with CD68+ macrophages, CD3+ T cells, and CD19+ B cells in PDAC. However, a higher expression level of VISTA was detected in tumor-infiltrating CD68+ macrophages than in CD3+ T and CD19+ B cells. Furthermore, anti-VISTA antibody treatment significantly reduced the number of metastatic nodules in livers of mouse models of PDAC with liver metastases. CONCLUSION VISTA expressed in TCs is associated with a favorable prognosis in PDAC. Moreover, immunotherapy with anti-VISTA antibodies may potentially be an effective treatment strategy against PDAC.
Collapse
Affiliation(s)
- Zelin Hou
- Department of General Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, 350001 People’s Republic of China
| | - Yu Pan
- Department of General Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, 350001 People’s Republic of China
| | - Qinglin Fei
- Department of General Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, 350001 People’s Republic of China
- The Cancer Center, Fujian Medical University Union Hospital, Fuzhou, 350001 China
| | - Yali Lin
- Department of General Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, 350001 People’s Republic of China
| | - Yuanyuan Zhou
- Department of Geriatrics, Fujian Medical University Union Hospital, Fuzhou, 350001 China
| | - Ying Liu
- Department of Endocrinology, Quanzhou Hospital of Traditional Chinese Medicine, Quanzhou, 362000 China
| | - Hongdan Guan
- Department of Radiation Oncology, Fujian Medical University Union Hospital, Fuzhou, 350001 China
| | - Xunbin Yu
- Department of Pathology, Fujian Provincialial Hospital, Fuzhou, 350001 China
| | - Xianchao Lin
- Department of General Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, 350001 People’s Republic of China
| | - Fengchun Lu
- Department of General Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, 350001 People’s Republic of China
| | - Heguang Huang
- Department of General Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, 350001 People’s Republic of China
| |
Collapse
|
44
|
Yang S, Liu Q, Liao Q. Tumor-Associated Macrophages in Pancreatic Ductal Adenocarcinoma: Origin, Polarization, Function, and Reprogramming. Front Cell Dev Biol 2021; 8:607209. [PMID: 33505964 PMCID: PMC7829544 DOI: 10.3389/fcell.2020.607209] [Citation(s) in RCA: 100] [Impact Index Per Article: 33.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 11/19/2020] [Indexed: 12/19/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly lethal malignancy. PDAC is only cured by surgical resection in its early stage, but there remains a relatively high possibility of recurrence. The development of PDAC is closely associated with the tumor microenvironment. Tumor-associated macrophages (TAMs) are one of the most abundant immune cell populations in the pancreatic tumor stroma. TAMs are inclined to M2 deviation in the tumor microenvironment, which promotes and supports tumor behaviors, including tumorigenesis, immune escape, metastasis, and chemotherapeutic resistance. Herein, we comprehensively reviewed the latest researches on the origin, polarization, functions, and reprogramming of TAMs in PDAC.
Collapse
Affiliation(s)
- Sen Yang
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Qiaofei Liu
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Quan Liao
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
45
|
Wang B, Zhao Q, Zhang Y, Liu Z, Zheng Z, Liu S, Meng L, Xin Y, Jiang X. Targeting hypoxia in the tumor microenvironment: a potential strategy to improve cancer immunotherapy. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2021; 40:24. [PMID: 33422072 PMCID: PMC7796640 DOI: 10.1186/s13046-020-01820-7] [Citation(s) in RCA: 134] [Impact Index Per Article: 44.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 12/20/2020] [Indexed: 12/14/2022]
Abstract
With the success of immune checkpoint inhibitors (ICIs), significant progress has been made in the field of cancer immunotherapy. Despite the long-lasting outcomes in responders, the majority of patients with cancer still do not benefit from this revolutionary therapy. Increasing evidence suggests that one of the major barriers limiting the efficacy of immunotherapy seems to coalesce with the hypoxic tumor microenvironment (TME), which is an intrinsic property of all solid tumors. In addition to its impact on shaping tumor invasion and metastasis, the hypoxic TME plays an essential role in inducing immune suppression and resistance though fostering diverse changes in stromal cell biology. Therefore, targeting hypoxia may provide a means to enhance the efficacy of immunotherapy. In this review, the potential impact of hypoxia within the TME, in terms of key immune cell populations, and the contribution to immune suppression are discussed. In addition, we outline how hypoxia can be manipulated to tailor the immune response and provide a promising combinational therapeutic strategy to improve immunotherapy.
Collapse
Affiliation(s)
- Bin Wang
- Department of Radiation Oncology, The First Hospital of Jilin University, 71 Xinmin Street, Changchun, 130021, China.,Jilin Provincial Key Laboratory of Radiation Oncology & Therapy, The First Hospital of Jilin University, Changchun, 130021, China.,NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun, 130021, China
| | - Qin Zhao
- Department of Radiation Oncology, The First Hospital of Jilin University, 71 Xinmin Street, Changchun, 130021, China.,Jilin Provincial Key Laboratory of Radiation Oncology & Therapy, The First Hospital of Jilin University, Changchun, 130021, China.,NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun, 130021, China
| | - Yuyu Zhang
- Department of Radiation Oncology, The First Hospital of Jilin University, 71 Xinmin Street, Changchun, 130021, China.,Jilin Provincial Key Laboratory of Radiation Oncology & Therapy, The First Hospital of Jilin University, Changchun, 130021, China.,NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun, 130021, China
| | - Zijing Liu
- Department of Radiation Oncology, The First Hospital of Jilin University, 71 Xinmin Street, Changchun, 130021, China.,Jilin Provincial Key Laboratory of Radiation Oncology & Therapy, The First Hospital of Jilin University, Changchun, 130021, China.,NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun, 130021, China
| | - Zhuangzhuang Zheng
- Department of Radiation Oncology, The First Hospital of Jilin University, 71 Xinmin Street, Changchun, 130021, China.,Jilin Provincial Key Laboratory of Radiation Oncology & Therapy, The First Hospital of Jilin University, Changchun, 130021, China.,NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun, 130021, China
| | - Shiyu Liu
- Department of Radiation Oncology, The First Hospital of Jilin University, 71 Xinmin Street, Changchun, 130021, China.,Jilin Provincial Key Laboratory of Radiation Oncology & Therapy, The First Hospital of Jilin University, Changchun, 130021, China.,NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun, 130021, China
| | - Lingbin Meng
- Department of Hematology and Medical Oncology, Moffitt Cancer Center, Tampa, FL, 33612, USA
| | - Ying Xin
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, 126 Xinmin Street, Changchun, 130021, China.
| | - Xin Jiang
- Department of Radiation Oncology, The First Hospital of Jilin University, 71 Xinmin Street, Changchun, 130021, China. .,Jilin Provincial Key Laboratory of Radiation Oncology & Therapy, The First Hospital of Jilin University, Changchun, 130021, China. .,NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun, 130021, China.
| |
Collapse
|
46
|
Abstract
Over the last few years, cancer immunotherapy experienced tremendous developments and it is nowadays considered a promising strategy against many types of cancer. However, the exclusion of lymphocytes from the tumor nest is a common phenomenon that limits the efficiency of immunotherapy in solid tumors. Despite several mechanisms proposed during the years to explain the immune excluded phenotype, at present, there is no integrated understanding about the role played by different models of immune exclusion in human cancers. Hypoxia is a hallmark of most solid tumors and, being a multifaceted and complex condition, shapes in a unique way the tumor microenvironment, affecting gene transcription and chromatin remodeling. In this review, we speculate about an upstream role for hypoxia as a common biological determinant of immune exclusion in solid tumors. We also discuss the current state of ex vivo and in vivo imaging of hypoxic determinants in relation to T cell distribution that could mechanisms of immune exclusion and discover functional-morphological tumor features that could support clinical monitoring.
Collapse
|
47
|
Macrophages in multiple myeloma: key roles and therapeutic strategies. Cancer Metastasis Rev 2021; 40:273-284. [PMID: 33404860 DOI: 10.1007/s10555-020-09943-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Accepted: 11/24/2020] [Indexed: 12/20/2022]
Abstract
Macrophages are a vital component of the tumour microenvironment and crucial mediators of tumour progression. In the last decade, significant strides have been made in understanding the crucial functional roles played by macrophages in the development of the plasma cell (PC) malignancy, multiple myeloma (MM). Whilst the interaction between MM PC and stromal cells within the bone marrow (BM) microenvironment has been extensively studied, we are only just starting to appreciate the multifaceted roles played by macrophages in disease progression. Accumulating evidence demonstrates that macrophage infiltration is associated with poor overall survival in MM. Indeed, macrophages influence numerous pathways critical for the initiation and progression of MM, including homing of malignant cells to BM, tumour cell growth and survival, drug resistance, angiogenesis and immune suppression. As such, therapeutic strategies aimed at targeting macrophages within the BM niche have promise in the clinical setting. This review will discuss the functions elicited by macrophages throughout different stages of MM and provide a comprehensive evaluation of potential macrophage-targeted therapies.
Collapse
|
48
|
Cai Y, Zheng C, Xiong F, Ran W, Zhai Y, Zhu HH, Wang H, Li Y, Zhang P. Recent Progress in the Design and Application of Supramolecular Peptide Hydrogels in Cancer Therapy. Adv Healthc Mater 2021; 10:e2001239. [PMID: 32935937 DOI: 10.1002/adhm.202001239] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 09/04/2020] [Indexed: 12/15/2022]
Abstract
Supramolecular peptide hydrogel (SPH) is a class of biomaterials self-assembled from peptide-based gelators through non-covalent interactions. Among many of its biomedical applications, the potential of SPH in cancer therapy has been vastly explored in the past decade, taking advantage of its good biocompatibility, multifunctionality, and injectability. SPHs can exert localized cancer therapy and induce systemic anticancer immunity to prevent tumor recurrence, depending on the design of SPH. This review first gives a brief introduction to SPH and then outlines the major types of peptide-based gelators that have been developed so far. The methodologies to tune the physicochemical properties and biological activities are summarized. The recent advances of SPH in cancer therapy as carriers, prodrugs, or drugs are highlighted. Finally, the clinical translation potential and main challenges in this field are also discussed.
Collapse
Affiliation(s)
- Ying Cai
- State Key Laboratory of Drug Research and Center of Pharmaceutics Shanghai Institute of Materia Medica Chinese Academy of Sciences Shanghai 201203 China
- University of Chinese Academy of Sciences Beijing 100049 China
| | - Chao Zheng
- State Key Laboratory of Drug Research and Center of Pharmaceutics Shanghai Institute of Materia Medica Chinese Academy of Sciences Shanghai 201203 China
- China State Institute of Pharmaceutical Industry Shanghai 200040 China
| | - Fengqin Xiong
- State Key Laboratory of Drug Research and Center of Pharmaceutics Shanghai Institute of Materia Medica Chinese Academy of Sciences Shanghai 201203 China
- China State Institute of Pharmaceutical Industry Shanghai 200040 China
| | - Wei Ran
- State Key Laboratory of Drug Research and Center of Pharmaceutics Shanghai Institute of Materia Medica Chinese Academy of Sciences Shanghai 201203 China
- University of Chinese Academy of Sciences Beijing 100049 China
| | - Yihui Zhai
- State Key Laboratory of Drug Research and Center of Pharmaceutics Shanghai Institute of Materia Medica Chinese Academy of Sciences Shanghai 201203 China
- University of Chinese Academy of Sciences Beijing 100049 China
| | - Helen H. Zhu
- State Key Laboratory of Oncogenes and Related Genes Renji‐Med‐X Stem Cell Research Center Department of Urology Ren Ji Hospital School of Medicine and School of Biomedical Engineering Shanghai Jiao Tong University Shanghai 200127 China
| | - Hao Wang
- China State Institute of Pharmaceutical Industry Shanghai 200040 China
| | - Yaping Li
- State Key Laboratory of Drug Research and Center of Pharmaceutics Shanghai Institute of Materia Medica Chinese Academy of Sciences Shanghai 201203 China
- University of Chinese Academy of Sciences Beijing 100049 China
| | - Pengcheng Zhang
- State Key Laboratory of Drug Research and Center of Pharmaceutics Shanghai Institute of Materia Medica Chinese Academy of Sciences Shanghai 201203 China
- University of Chinese Academy of Sciences Beijing 100049 China
- Yantai Key Laboratory of Nanomedicine and Advanced Preparations Yantai Institute of Materia Medica Shandong 264000 China
| |
Collapse
|
49
|
Characteristics of the Tumor Microenvironment That Influence Immune Cell Functions: Hypoxia, Oxidative Stress, Metabolic Alterations. Cancers (Basel) 2020; 12:cancers12123802. [PMID: 33348579 PMCID: PMC7765870 DOI: 10.3390/cancers12123802] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Revised: 12/06/2020] [Accepted: 12/10/2020] [Indexed: 12/20/2022] Open
Abstract
Immunotherapy (IMT) is now a core component of cancer treatment, however, many patients do not respond to these novel therapies. Investigating the resistance mechanisms behind this differential response is now a critical area of research. Immune-based therapies, particularly immune checkpoint inhibitors (ICI), rely on a robust infiltration of T-cells into the tumor microenvironment (TME) for an effective response. While early efforts relied on quantifying tumor infiltrating lymphocytes (TIL) in the TME, characterizing the functional quality and degree of TIL exhaustion correlates more strongly with ICI response. Even with sufficient TME infiltration, immune cells face a harsh metabolic environment that can significantly impair effector function. These tumor-mediated metabolic perturbations include hypoxia, oxidative stress, and metabolites of cellular energetics. Primarily through HIF-1-dependent processes, hypoxia invokes an immunosuppressive phenotype via altered molecular markers, immune cell trafficking, and angiogenesis. Additionally, oxidative stress can promote lipid peroxidation, ER stress, and Treg dysfunction, all associated with immune dysregulation. Finally, the metabolic byproducts of lipids, amino acids, glucose, and cellular energetics are associated with immunosuppression and ICI resistance. This review will explore these biochemical pathways linked to immune cell dysfunction in the TME and highlight potential adjunctive therapies to be used alongside current IMT.
Collapse
|
50
|
Zhang YR, Luo JQ, Zhang JY, Miao WM, Wu JS, Huang H, Tong QS, Shen S, Leong KW, Du JZ, Wang J. Nanoparticle-Enabled Dual Modulation of Phagocytic Signals to Improve Macrophage-Mediated Cancer Immunotherapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2020; 16:e2004240. [PMID: 33107142 DOI: 10.1002/smll.202004240] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 09/07/2020] [Indexed: 05/13/2023]
Abstract
Activation of the phagocytosis of macrophages to tumor cells is an attractive strategy for cancer immunotherapy, but the effectiveness is limited by the fact that many tumor cells express an increased level of anti-phagocytic signals (e.g., CD47 molecules) on their surface. To promote phagocytosis of macrophages, a pro-phagocytic nanoparticle (SNPACALR&aCD47 ) that concurrently carries CD47 antibody (aCD47) and a pro-phagocytic molecule calreticulin (CALR) is constructed to simultaneously modulate the phagocytic signals of macrophages. SNPACALR&aCD47 can achieve targeted delivery to tumor cells by specifically binding to the cell-surface CD47 and block the CD47-SIRPα pathway to inhibit the "don't eat me" signal. Tumor cell-targeted delivery increases the exposure of recombinant CALR on the cell surface and stimulates an "eat me" signal. Simultaneous modulation of the two signals enhances the phagocytosis of 4T1 tumor cells by macrophages, which leads to significantly improved anti-tumor efficacy in vivo. The findings demonstrate that the concurrent blockade of anti-phagocytic signals and activation of pro-phagocytic signals can be effective in macrophage-mediated cancer immunotherapy.
Collapse
Affiliation(s)
- Ya-Ru Zhang
- Guangzhou First People's Hospital, and Institutes for Life Sciences, School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, 510006, China
| | - Jia-Qi Luo
- Guangzhou First People's Hospital, and Institutes for Life Sciences, School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, 510006, China
| | - Jing-Yang Zhang
- Guangzhou First People's Hospital, and Institutes for Life Sciences, School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, 510006, China
| | - Wei-Min Miao
- Guangzhou First People's Hospital, and Institutes for Life Sciences, School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, 510006, China
| | - Jia-Si Wu
- Guangzhou First People's Hospital, and Institutes for Life Sciences, School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, 510006, China
| | - Hua Huang
- Guangzhou First People's Hospital, and Institutes for Life Sciences, School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, 510006, China
| | - Qi-Song Tong
- Guangzhou First People's Hospital, and Institutes for Life Sciences, School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, 510006, China
| | - Song Shen
- Guangzhou First People's Hospital, and Institutes for Life Sciences, School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, 510006, China
- National Engineering Research Center for Tissue Restoration and Reconstruction, Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, Key Laboratory of Biomedical Engineering of Guangdong Province, and Innovation Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, China
| | - Kam W Leong
- Department of Biomedical Engineering, Columbia University, New York, NY, 10027, USA
| | - Jin-Zhi Du
- Guangzhou First People's Hospital, and Institutes for Life Sciences, School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, 510006, China
- National Engineering Research Center for Tissue Restoration and Reconstruction, Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, Key Laboratory of Biomedical Engineering of Guangdong Province, and Innovation Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, China
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, 510005, China
| | - Jun Wang
- Guangzhou First People's Hospital, and Institutes for Life Sciences, School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, 510006, China
- National Engineering Research Center for Tissue Restoration and Reconstruction, Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, Key Laboratory of Biomedical Engineering of Guangdong Province, and Innovation Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, China
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, 510005, China
| |
Collapse
|