1
|
Krenz B, Lee J, Kannan T, Eilers M. Immune evasion: An imperative and consequence of MYC deregulation. Mol Oncol 2024; 18:2338-2355. [PMID: 38957016 PMCID: PMC11459038 DOI: 10.1002/1878-0261.13695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 05/08/2024] [Accepted: 06/19/2024] [Indexed: 07/04/2024] Open
Abstract
MYC has been implicated in the pathogenesis of a wide range of human tumors and has been described for many years as a transcription factor that regulates genes with pleiotropic functions to promote tumorigenic growth. However, despite extensive efforts to identify specific target genes of MYC that alone could be responsible for promoting tumorigenesis, the field is yet to reach a consensus whether this is the crucial function of MYC. Recent work shifts the view on MYC's function from being a gene-specific transcription factor to an essential stress resilience factor. In highly proliferating cells, MYC preserves cell integrity by promoting DNA repair at core promoters, protecting stalled replication forks, and/or preventing transcription-replication conflicts. Furthermore, an increasing body of evidence demonstrates that MYC not only promotes tumorigenesis by driving cell-autonomous growth, but also enables tumors to evade the host's immune system. In this review, we summarize our current understanding of how MYC impairs antitumor immunity and why this function is evolutionarily hard-wired to the biology of the MYC protein family. We show why the cell-autonomous and immune evasive functions of MYC are mutually dependent and discuss ways to target MYC proteins in cancer therapy.
Collapse
Affiliation(s)
- Bastian Krenz
- Department of Biochemistry and Molecular BiologyTheodor Boveri Institute, Biocenter, University of WürzburgWürzburgGermany
- Mildred Scheel Early Career CenterWürzburgGermany
| | - Jongkuen Lee
- Department of Biochemistry and Molecular BiologyTheodor Boveri Institute, Biocenter, University of WürzburgWürzburgGermany
| | - Toshitha Kannan
- Department of Biochemistry and Molecular BiologyTheodor Boveri Institute, Biocenter, University of WürzburgWürzburgGermany
| | - Martin Eilers
- Department of Biochemistry and Molecular BiologyTheodor Boveri Institute, Biocenter, University of WürzburgWürzburgGermany
- Comprehensive Cancer Center MainfrankenWürzburgGermany
| |
Collapse
|
2
|
Luo Q, Shen F, Zhao S, Dong L, Wei J, Hu H, Huang Q, Wang Q, Yang P, Liang W, Li W, He F, Cao J. LINC00460/miR-186-3p/MYC feedback loop facilitates colorectal cancer immune escape by enhancing CD47 and PD-L1 expressions. J Exp Clin Cancer Res 2024; 43:225. [PMID: 39135122 PMCID: PMC11321182 DOI: 10.1186/s13046-024-03145-1] [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: 01/28/2024] [Accepted: 07/29/2024] [Indexed: 08/15/2024] Open
Abstract
BACKGROUND Long non-coding RNAs (LncRNAs) have been implicated as critical regulators of cancer tumorigenesis and progression. However, their functions and molecular mechanisms in colorectal cancer (CRC) still remain to be further elucidated. METHODS LINC00460 was identified by differential analysis between human CRC and normal tissues and verified by in situ hybridization (ISH) and qRT-PCR. We investigated the biological functions of LINC00460 in CRC by in vitro and in vivo experiments. We predicted the mechanism and downstream functional molecules of LINC00460 by bioinformatics analysis, and confirmed them by dual luciferase reporter gene assay, RNA immunoprecipitation (RIP), RNA pull-down, etc. RESULTS: LINC00460 was found to be significantly overexpressed in CRC and associated with poor prognosis. Overexpression of LINC00460 promoted CRC cell immune escape and remodeled a suppressive tumor immune microenvironment, thereby promoting CRC proliferation and metastasis. Mechanistic studies showed that LINC00460 served as a molecular sponge for miR-186-3p, and then promoted the expressions of MYC, CD47 and PD-L1 to facilitate CRC cell immune escape. We also demonstrated that MYC upregulated LINC00460 expression at the transcriptional level and formed a positive feedback loop. CONCLUSIONS The LINC00460/miR-186-3p/MYC feedback loop promotes CRC cell immune escape and subsequently facilitates CRC proliferation and metastasis. Our findings provide novel insight into LINC00460 as a CRC immune regulator, and provide a potential therapeutic target for CRC patients.
Collapse
Affiliation(s)
- Qingqing Luo
- Department of Gastroenterology and Hepatology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, 510180, China
- Guangzhou Key Laboratory of Digestive Diseases, Guangzhou Digestive Disease Center, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, 510180, China
| | - Fei Shen
- Department of General Surgery, The First Affiliated Hospital, Jinan University, Guangzhou, Guangdong, 510630, China
- Department of Thyroid surgery, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, 510180, China
| | - Sheng Zhao
- Department of Nephrology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, 510180, China
| | - Lan Dong
- Guangzhou Key Laboratory of Digestive Diseases, Guangzhou Digestive Disease Center, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, 510180, China
- Guangzhou Digestive Disease Center, Department of Gastrointestinal Surgery, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, 510180, China
| | - Jianchang Wei
- Guangzhou Key Laboratory of Digestive Diseases, Guangzhou Digestive Disease Center, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, 510180, China
- Guangzhou Digestive Disease Center, Department of Gastrointestinal Surgery, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, 510180, China
| | - He Hu
- Guangzhou Key Laboratory of Digestive Diseases, Guangzhou Digestive Disease Center, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, 510180, China
- Guangzhou Digestive Disease Center, Department of Gastrointestinal Surgery, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, 510180, China
| | - Qing Huang
- Guangzhou Key Laboratory of Digestive Diseases, Guangzhou Digestive Disease Center, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, 510180, China
- Guangzhou Digestive Disease Center, Department of Gastrointestinal Surgery, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, 510180, China
| | - Qiang Wang
- Guangzhou Key Laboratory of Digestive Diseases, Guangzhou Digestive Disease Center, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, 510180, China
- Guangzhou Digestive Disease Center, Department of Gastrointestinal Surgery, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, 510180, China
| | - Ping Yang
- Guangzhou Key Laboratory of Digestive Diseases, Guangzhou Digestive Disease Center, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, 510180, China
- Guangzhou Digestive Disease Center, Department of Gastrointestinal Surgery, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, 510180, China
| | - Wenlong Liang
- Guangzhou Key Laboratory of Digestive Diseases, Guangzhou Digestive Disease Center, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, 510180, China
- Guangzhou Digestive Disease Center, Department of Gastrointestinal Surgery, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, 510180, China
| | - Wanglin Li
- Guangzhou Key Laboratory of Digestive Diseases, Guangzhou Digestive Disease Center, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, 510180, China
- Guangzhou Digestive Disease Center, Department of Gastrointestinal Surgery, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, 510180, China
| | - Feng He
- Department of Nephrology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, 510180, China.
| | - Jie Cao
- Guangzhou Key Laboratory of Digestive Diseases, Guangzhou Digestive Disease Center, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, 510180, China.
- Guangzhou Digestive Disease Center, Department of Gastrointestinal Surgery, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, 510180, China.
| |
Collapse
|
3
|
Zhu J, Wang F, Wang L, Dai B, Xu G, Zhao L, Jiang H, Gao W, Zhang T, Zhao C, Li YX, Hu J, Li K. HDAC Inhibition Increases CXCL12 Secretion to Recruit Natural Killer Cells in Peripheral T-cell Lymphoma. Cancer Res 2024; 84:2450-2467. [PMID: 38718305 DOI: 10.1158/0008-5472.can-23-3250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 03/13/2024] [Accepted: 05/01/2024] [Indexed: 08/02/2024]
Abstract
Peripheral T-cell lymphoma (PTCL) is a heterogeneous and aggressive disease with a poor prognosis. Histone deacetylase (HDAC) inhibitors have shown inhibitory effects on PTCL. A better understanding of the therapeutic mechanism underlying the effects of HDAC inhibitors could help improve treatment strategies. Herein, we found that high expression of HDAC3 is associated with poor prognosis in PTCL. HDAC3 inhibition suppressed lymphoma growth in immunocompetent mice but not in immunodeficient mice. HDAC3 deletion delayed the progression of lymphoma, reduced the lymphoma burden in the thymus, spleen, and lymph nodes, and prolonged the survival of mice bearing N-methyl-N-nitrosourea-induced lymphoma. Furthermore, inhibiting HDAC3 promoted the infiltration and enhanced the function of natural killer (NK) cells. Mechanistically, HDAC3 mediated ATF3 deacetylation, enhancing its transcriptional inhibitory activity. Targeting HDAC3 enhanced CXCL12 secretion through an ATF3-dependent pathway to stimulate NK-cell recruitment and activation. Finally, HDAC3 suppression improved the response of PTCL to conventional chemotherapy. Collectively, this study provides insights into the mechanism by which HDAC3 regulates ATF3 activity and CXCL12 secretion, leading to immune infiltration and lymphoma suppression. Combining HDAC3 inhibitors with chemotherapy may be a promising strategy for treating PTCL. Significance: Targeting HDAC3 suppresses progression of T-cell lymphoma by activating ATF3 to induce secretion of CXCL12 and promote infiltration of NK cells, providing an immunostimulatory approach for treating T-cell lymphoma patients.
Collapse
MESH Headings
- Animals
- Histone Deacetylase Inhibitors/pharmacology
- Mice
- Histone Deacetylases/metabolism
- Histone Deacetylases/genetics
- Lymphoma, T-Cell, Peripheral/pathology
- Lymphoma, T-Cell, Peripheral/immunology
- Lymphoma, T-Cell, Peripheral/metabolism
- Lymphoma, T-Cell, Peripheral/drug therapy
- Lymphoma, T-Cell, Peripheral/genetics
- Humans
- Chemokine CXCL12/metabolism
- Killer Cells, Natural/immunology
- Killer Cells, Natural/metabolism
- Activating Transcription Factor 3/metabolism
- Activating Transcription Factor 3/genetics
- Cell Line, Tumor
- Female
- Male
- Mice, Inbred C57BL
- Prognosis
Collapse
Affiliation(s)
- Jiayan Zhu
- Shanghai Institute of Hematology, Blood and Marrow Transplantation Center, Collaborative Innovation Center of Hematology, Department of Hematology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, NHC Key Laboratory of Biotechnology of Antibiotics, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Department of Hematology, Shanghai Sixth People's Hospital, Shanghai Jiaotong University, Shanghai, China
| | - Feng Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, NHC Key Laboratory of Biotechnology of Antibiotics, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Lining Wang
- Shanghai Institute of Hematology, Blood and Marrow Transplantation Center, Collaborative Innovation Center of Hematology, Department of Hematology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bo Dai
- Department of Hematology, Huashan Hospital, Fudan University, Shanghai, China
| | - Guilin Xu
- Shanghai Institute of Hematology, Blood and Marrow Transplantation Center, Collaborative Innovation Center of Hematology, Department of Hematology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Luyao Zhao
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, NHC Key Laboratory of Biotechnology of Antibiotics, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Huimin Jiang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, NHC Key Laboratory of Biotechnology of Antibiotics, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Wenhui Gao
- Shanghai Institute of Hematology, Blood and Marrow Transplantation Center, Collaborative Innovation Center of Hematology, Department of Hematology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tingting Zhang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, NHC Key Laboratory of Biotechnology of Antibiotics, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Chenxi Zhao
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, NHC Key Laboratory of Biotechnology of Antibiotics, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yun-Xuan Li
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, NHC Key Laboratory of Biotechnology of Antibiotics, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jiong Hu
- Shanghai Institute of Hematology, Blood and Marrow Transplantation Center, Collaborative Innovation Center of Hematology, Department of Hematology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ke Li
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, NHC Key Laboratory of Biotechnology of Antibiotics, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
4
|
Ghasemi N, Azizi H. Exploring Myc puzzle: Insights into cancer, stem cell biology, and PPI networks. Gene 2024; 916:148447. [PMID: 38583818 DOI: 10.1016/j.gene.2024.148447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 03/13/2024] [Accepted: 04/04/2024] [Indexed: 04/09/2024]
Abstract
"The grand orchestrator," "Universal Amplifier," "double-edged sword," and "Undruggable" are just some of the Myc oncogene so-called names. It has been around 40 years since the discovery of the Myc, and it remains in the mainstream of cancer treatment drugs. Myc is part of basic helix-loop-helix leucine zipper (bHLH-LZ) superfamily proteins, and its dysregulation can be seen in many malignant human tumors. It dysregulates critical pathways in cells that are connected to each other, such as proliferation, growth, cell cycle, and cell adhesion, impacts miRNAs action, intercellular metabolism, DNA replication, differentiation, microenvironment regulation, angiogenesis, and metastasis. Myc, surprisingly, is used in stem cell research too. Its family includes three members, MYC, MYCN, and MYCL, and each dysfunction was observed in different cancer types. This review aims to introduce Myc and its function in the body. Besides, Myc deregulatory mechanisms in cancer cells, their intricate aspects will be discussed. We will look at promising drugs and Myc-based therapies. Finally, Myc and its role in stemness, Myc pathways based on PPI network analysis, and future insights will be explained.
Collapse
Affiliation(s)
- Nima Ghasemi
- Faculty of Biotechnology, Amol University of Special Modern Technologies, Amol, Iran
| | - Hossein Azizi
- Faculty of Biotechnology, Amol University of Special Modern Technologies, Amol, Iran.
| |
Collapse
|
5
|
Pesini C, Artal L, Paúl Bernal J, Sánchez Martinez D, Pardo J, Ramírez-Labrada A. In-depth analysis of the interplay between oncogenic mutations and NK cell-mediated cancer surveillance in solid tumors. Oncoimmunology 2024; 13:2379062. [PMID: 39036370 PMCID: PMC11259085 DOI: 10.1080/2162402x.2024.2379062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 07/08/2024] [Indexed: 07/23/2024] Open
Abstract
Natural killer (NK) cells play a crucial role in antitumoral and antiviral responses. Yet, cancer cells can alter themselves or the microenvironment through the secretion of cytokines or other factors, hindering NK cell activation and promoting a less cytotoxic phenotype. These resistance mechanisms, often referred to as the "hallmarks of cancer" are significantly influenced by the activation of oncogenes, impacting most, if not all, of the described hallmarks. Along with oncogenes, other types of genes, the tumor suppressor genes are frequently mutated or modified during cancer. Traditionally, these genes have been associated with uncontrollable tumor growth and apoptosis resistance. Recent evidence suggests oncogenic mutations extend beyond modulating cell death/proliferation programs, influencing cancer immunosurveillance. While T cells have been more studied, the results obtained highlight NK cells as emerging key protagonists for enhancing tumor cell elimination by modulating oncogenic activity. A few recent studies highlight the crucial role of oncogenic mutations in NK cell-mediated cancer recognition, impacting angiogenesis, stress ligands, and signaling balance within the tumor microenvironment. This review will critically examine recent discoveries correlating oncogenic mutations to NK cell-mediated cancer immunosurveillance, a relatively underexplored area, particularly in the era dominated by immune checkpoint inhibitors and CAR-T cells. Building on these insights, we will explore opportunities to improve NK cell-based immunotherapies, which are increasingly recognized as promising alternatives for treating low-antigenic tumors, offering significant advantages in terms of safety and manufacturing suitability.
Collapse
Affiliation(s)
- Cecilia Pesini
- Aragón Health Research Institute (IIS Aragón), Biomedical Research Centre of Aragón (CIBA), Zaragoza, Spain
- Center for Biomedical Research in the Network of Infectious Diseases (CIBERINFEC), Carlos III Health Institute, Zaragoza, Spain
- Department of Microbiology, Radiology, Pediatry and Public Health, University of Zaragoza, Zaragoza, Spain
| | - Laura Artal
- Aragón Health Research Institute (IIS Aragón), Biomedical Research Centre of Aragón (CIBA), Zaragoza, Spain
- Institute of Carbochemistry (ICB-CSIC), Zaragoza, Spain
| | - Jorge Paúl Bernal
- Aragón Health Research Institute (IIS Aragón), Biomedical Research Centre of Aragón (CIBA), Zaragoza, Spain
| | - Diego Sánchez Martinez
- Aragón Health Research Institute (IIS Aragón), Biomedical Research Centre of Aragón (CIBA), Zaragoza, Spain
- Aragón I + D Foundation (ARAID), Government of Aragon, Zaragoza, Spain
| | - Julián Pardo
- Aragón Health Research Institute (IIS Aragón), Biomedical Research Centre of Aragón (CIBA), Zaragoza, Spain
- Center for Biomedical Research in the Network of Infectious Diseases (CIBERINFEC), Carlos III Health Institute, Zaragoza, Spain
- Department of Microbiology, Radiology, Pediatry and Public Health, University of Zaragoza, Zaragoza, Spain
| | - Ariel Ramírez-Labrada
- Aragón Health Research Institute (IIS Aragón), Biomedical Research Centre of Aragón (CIBA), Zaragoza, Spain
- Center for Biomedical Research in the Network of Infectious Diseases (CIBERINFEC), Carlos III Health Institute, Zaragoza, Spain
| |
Collapse
|
6
|
Miranda A, Pattnaik S, Hamilton PT, Fuss MA, Kalaria S, Laumont CM, Smazynski J, Mesa M, Banville A, Jiang X, Jenkins R, Cañadas I, Nelson BH. N-MYC impairs innate immune signaling in high-grade serous ovarian carcinoma. SCIENCE ADVANCES 2024; 10:eadj5428. [PMID: 38748789 PMCID: PMC11095474 DOI: 10.1126/sciadv.adj5428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 04/15/2024] [Indexed: 05/19/2024]
Abstract
High-grade serous ovarian cancer (HGSC) is a challenging disease, especially for patients with immunologically "cold" tumors devoid of tumor-infiltrating lymphocytes (TILs). We found that HGSC exhibits among the highest levels of MYCN expression and transcriptional signature across human cancers, which is strongly linked to diminished features of antitumor immunity. N-MYC repressed basal and induced IFN type I signaling in HGSC cell lines, leading to decreased chemokine expression and T cell chemoattraction. N-MYC inhibited the induction of IFN type I by suppressing tumor cell-intrinsic STING signaling via reduced STING oligomerization, and by blunting RIG-I-like receptor signaling through inhibition of MAVS aggregation and localization in the mitochondria. Single-cell RNA sequencing of human clinical HGSC samples revealed a strong negative association between cancer cell-intrinsic MYCN transcriptional program and type I IFN signaling. Thus, N-MYC inhibits tumor cell-intrinsic innate immune signaling in HGSC, making it a compelling target for immunotherapy of cold tumors.
Collapse
Affiliation(s)
- Alex Miranda
- Deeley Research Centre, BC Cancer, Victoria, BC V8R 6V5, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Swetansu Pattnaik
- The Kinghorn Cancer Centre and Cancer Division, Garvan Institute of Medical Research, 370 Victoria St, Darlinghurst, NSW, Australia
| | - Phineas T. Hamilton
- Deeley Research Centre, BC Cancer, Victoria, BC V8R 6V5, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | | | - Shreena Kalaria
- Deeley Research Centre, BC Cancer, Victoria, BC V8R 6V5, Canada
| | - Céline M. Laumont
- Deeley Research Centre, BC Cancer, Victoria, BC V8R 6V5, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | | | - Monica Mesa
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, BC V8P 3E6, Canada
| | - Allyson Banville
- Deeley Research Centre, BC Cancer, Victoria, BC V8R 6V5, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Xinpei Jiang
- Nuclear Dynamics and Cancer Program, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Russell Jenkins
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Israel Cañadas
- Nuclear Dynamics and Cancer Program, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Brad H. Nelson
- Deeley Research Centre, BC Cancer, Victoria, BC V8R 6V5, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, BC V8P 3E6, Canada
| |
Collapse
|
7
|
Chan KI, Zhang S, Li G, Xu Y, Cui L, Wang Y, Su H, Tan W, Zhong Z. MYC Oncogene: A Druggable Target for Treating Cancers with Natural Products. Aging Dis 2024; 15:640-697. [PMID: 37450923 PMCID: PMC10917530 DOI: 10.14336/ad.2023.0520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 05/20/2023] [Indexed: 07/18/2023] Open
Abstract
Various diseases, including cancers, age-associated disorders, and acute liver failure, have been linked to the oncogene, MYC. Animal testing and clinical trials have shown that sustained tumor volume reduction can be achieved when MYC is inactivated, and different combinations of therapeutic agents including MYC inhibitors are currently being developed. In this review, we first provide a summary of the multiple biological functions of the MYC oncoprotein in cancer treatment, highlighting that the equilibrium points of the MYC/MAX, MIZ1/MYC/MAX, and MAD (MNT)/MAX complexes have further potential in cancer treatment that could be used to restrain MYC oncogene expression and its functions in tumorigenesis. We also discuss the multifunctional capacity of MYC in various cellular cancer processes, including its influences on immune response, metabolism, cell cycle, apoptosis, autophagy, pyroptosis, metastasis, angiogenesis, multidrug resistance, and intestinal flora. Moreover, we summarize the MYC therapy patent landscape and emphasize the potential of MYC as a druggable target, using herbal medicine modulators. Finally, we describe pending challenges and future perspectives in biomedical research, involving the development of therapeutic approaches to modulate MYC or its targeted genes. Patients with cancers driven by MYC signaling may benefit from therapies targeting these pathways, which could delay cancerous growth and recover antitumor immune responses.
Collapse
Affiliation(s)
- Ka Iong Chan
- Macao Centre for Research and Development in Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR 999078, China
| | - Siyuan Zhang
- Macao Centre for Research and Development in Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR 999078, China
| | - Guodong Li
- Macao Centre for Research and Development in Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR 999078, China
| | - Yida Xu
- Macao Centre for Research and Development in Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR 999078, China
| | - Liao Cui
- Guangdong Provincial Key Laboratory of Research and Development of Natural Drugs, School of Pharmacy, Guangdong Medical University, Zhanjiang 524000, China
| | - Yitao Wang
- Macao Centre for Research and Development in Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR 999078, China
| | - Huanxing Su
- Macao Centre for Research and Development in Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR 999078, China
| | - Wen Tan
- School of Pharmacy, Lanzhou University, Lanzhou 730000, China
| | - Zhangfeng Zhong
- Macao Centre for Research and Development in Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR 999078, China
| |
Collapse
|
8
|
de Jonge AV, Duetz C, Bruins WSC, Korst CLBM, Rentenaar R, Cosovic M, Eken M, Twickler I, Nijland M, van der Poel MWM, de Heer K, Klerk CPW, Strobbe L, Oosterveld M, Boersma R, Koene HR, Roemer MGM, van Werkhoven E, Chamuleau MED, Mutis T. Distinct peripheral T-cell and NK-cell profiles in HGBL-MYC/BCL2 vs patients with DLBCL NOS. Blood Adv 2024; 8:1094-1104. [PMID: 38191686 PMCID: PMC10907399 DOI: 10.1182/bloodadvances.2023011687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 11/27/2023] [Accepted: 12/22/2023] [Indexed: 01/10/2024] Open
Abstract
ABSTRACT Patients with high-grade B-cell lymphoma with MYC and BCL2 rearrangements (HGBL-MYC/BCL2) respond poorly to immunochemotherapy compared with patients with diffuse large B-cell lymphoma not otherwise specified (DLBCL NOS) without a MYC rearrangement. This suggests a negative impact of lymphoma-intrinsic MYC on the immune system. To investigate this, we compared circulating T cells and natural killer (NK) cells of patients with HGBL-MYC/BCL2 (n = 66), patients with DLBCL NOS (n = 53), and age-matched healthy donors (HDs; n = 16) by flow cytometry and performed proliferation, cytokine production, and cytotoxicity assays. Compared with HDs, both lymphoma subtypes displayed similar frequencies of CD8+ T cells but decreased CD4+ T cells. Regulatory T-cell (Treg) frequencies were reduced only in patients with DLBCL NOS. Activated (HLA-DR+/CD38+) T cells, PD-1+CD4+ T cells, and PD-1+Tregs were increased in both lymphoma subtypes, but PD-1+CD8+ T cells were increased only in HGBL-MYC/BCL2. Patients with DLBCL NOS, but not patients with HGBL-MYC/BCL2, exhibited higher frequencies of senescent T cells than HDs. Functional assays showed no overt differences between both lymphoma groups and HDs. Deeper analyses revealed that PD-1+ T cells of patients with HGBL-MYC/BCL2 were exhausted with impaired cytokine production and degranulation. Patients with DLBCL NOS, but not patients with HGBL-MYC/BCL2, exhibited higher frequencies of NK cells expressing inhibiting receptor NKG2A. Both lymphoma subtypes exhibited lower TIM-3+- and DNAM-1+-expressing NK cells. Although NK cells of patients with HGBL-MYC/BCL2 showed less degranulation, they were not defective in cytotoxicity. In conclusion, our results demonstrate an increased exhaustion in circulating T cells of patients with HGBL-MYC/BCL2. Nonetheless, the overall intact peripheral T-cell and NK-cell functions in these patients emphasize the importance of investigating potential immune evasion in the microenvironment of MYC-rearranged lymphomas.
Collapse
Affiliation(s)
- A. Vera de Jonge
- Department of Hematology, Amsterdam UMC Location Vrije Universiteit, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
| | - Carolien Duetz
- Department of Hematology, Amsterdam UMC Location Vrije Universiteit, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
| | - Wassilis S. C. Bruins
- Department of Hematology, Amsterdam UMC Location Vrije Universiteit, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
| | - Charlotte L. B. M. Korst
- Department of Hematology, Amsterdam UMC Location Vrije Universiteit, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
| | - Rosa Rentenaar
- Department of Hematology, Amsterdam UMC Location Vrije Universiteit, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
| | - Meliha Cosovic
- Department of Hematology, Amsterdam UMC Location Vrije Universiteit, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
| | - Merve Eken
- Department of Hematology, Amsterdam UMC Location Vrije Universiteit, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
| | - Inoka Twickler
- Department of Hematology, Amsterdam UMC Location Vrije Universiteit, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
| | - Marcel Nijland
- Department of Hematology, University Medical Center Groningen, Groningen, The Netherlands
| | - Marjolein W. M. van der Poel
- Division of Hematology, Department of Internal Medicine, GROW School for Oncology and Developmental Biology, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Koen de Heer
- Department of Internal Medicine, Flevoziekenhuis, Almere, The Netherlands
| | - Clara P. W. Klerk
- Department of Internal Medicine, Dijklanderziekenhuis, Hoorn, The Netherlands
| | - Leonie Strobbe
- Department of Internal Medicine, Gelreziekenhuizen, Zutphen, The Netherlands
| | | | - Rinske Boersma
- Department of Internal Medicine, Amphia Ziekenhuis, Breda, The Netherlands
| | - Harry R. Koene
- Department of Hematology, St Antonius Ziekenhuis, Nieuwegein, The Netherlands
| | - Margaretha G. M. Roemer
- Department of Hematology, Amsterdam UMC Location Vrije Universiteit, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
| | - Erik van Werkhoven
- HOVON Foundation, Rotterdam, The Netherlands
- Department of Hematology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Martine E. D. Chamuleau
- Department of Hematology, Amsterdam UMC Location Vrije Universiteit, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
| | - Tuna Mutis
- Department of Hematology, Amsterdam UMC Location Vrije Universiteit, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
| |
Collapse
|
9
|
Xu Y, Li X, Cheng F, Zhao B, Fang M, Li Z, Meng S. Heat shock protein gp96 drives natural killer cell maturation and anti-tumor immunity by counteracting Trim28 to stabilize Eomes. Nat Commun 2024; 15:1106. [PMID: 38321029 PMCID: PMC10847424 DOI: 10.1038/s41467-024-45426-5] [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: 11/28/2022] [Accepted: 01/23/2024] [Indexed: 02/08/2024] Open
Abstract
The maturation process of natural killer (NK) cells, which is regulated by multiple transcription factors, determines their functionality, but few checkpoints specifically targeting this process have been thoroughly studied. Here we show that NK-specific deficiency of glucose-regulated protein 94 (gp96) leads to decreased maturation of NK cells in mice. These gp96-deficient NK cells exhibit undermined activation, cytotoxicity and IFN-γ production upon stimulation, as well as weakened responses to IL-15 for NK cell maturation, in vitro. In vivo, NK-specific gp96-deficient mice show increased tumor growth. Mechanistically, we identify Eomes as the downstream transcription factor, with gp96 binding to Trim28 to prevent Trim28-mediated ubiquitination and degradation of Eomes. Our study thus suggests the gp96-Trim28-Eomes axis to be an important regulator for NK cell maturation and cancer surveillance in mice.
Collapse
Affiliation(s)
- Yuxiu Xu
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS), Beijing, China
| | - Xin Li
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS), Beijing, China.
| | - Fang Cheng
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS), Beijing, China
- University of Chinese Academy of Sciences, Beijing, P.R. China
| | - Bao Zhao
- Department of Otolaryngology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, 233004, China
| | - Min Fang
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS), Beijing, China
| | - Zihai Li
- The Pelotonia Institute for Immuno-Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - Songdong Meng
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS), Beijing, China.
- University of Chinese Academy of Sciences, Beijing, P.R. China.
| |
Collapse
|
10
|
Deutzmann A, Sullivan DK, Dhanasekaran R, Li W, Chen X, Tong L, Mahauad-Fernandez WD, Bell J, Mosley A, Koehler AN, Li Y, Felsher DW. Nuclear to cytoplasmic transport is a druggable dependency in MYC-driven hepatocellular carcinoma. Nat Commun 2024; 15:963. [PMID: 38302473 PMCID: PMC10834515 DOI: 10.1038/s41467-024-45128-y] [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: 08/20/2019] [Accepted: 01/12/2024] [Indexed: 02/03/2024] Open
Abstract
The MYC oncogene is often dysregulated in human cancer, including hepatocellular carcinoma (HCC). MYC is considered undruggable to date. Here, we comprehensively identify genes essential for survival of MYChigh but not MYClow cells by a CRISPR/Cas9 genome-wide screen in a MYC-conditional HCC model. Our screen uncovers novel MYC synthetic lethal (MYC-SL) interactions and identifies most MYC-SL genes described previously. In particular, the screen reveals nucleocytoplasmic transport to be a MYC-SL interaction. We show that the majority of MYC-SL nucleocytoplasmic transport genes are upregulated in MYChigh murine HCC and are associated with poor survival in HCC patients. Inhibiting Exportin-1 (XPO1) in vivo induces marked tumor regression in an autochthonous MYC-transgenic HCC model and inhibits tumor growth in HCC patient-derived xenografts. XPO1 expression is associated with poor prognosis only in HCC patients with high MYC activity. We infer that MYC may generally regulate and require altered expression of nucleocytoplasmic transport genes for tumorigenesis.
Collapse
Affiliation(s)
- Anja Deutzmann
- Division of Oncology, Department of Medicine, Stanford University, Stanford, CA, 94305, USA
| | - Delaney K Sullivan
- Division of Oncology, Department of Medicine, Stanford University, Stanford, CA, 94305, USA
| | - Renumathy Dhanasekaran
- Division of Oncology, Department of Medicine, Stanford University, Stanford, CA, 94305, USA
- Division of Gastroenterology, Department of Medicine, Stanford University, Stanford, CA, 94305, USA
| | - Wei Li
- Center for Genetic Medicine Research, Children's National Hospital, Washington, DC, 20012, USA
- Department of Genomics and Precision Medicine, George Washington University, Washington, DC, 20012, USA
| | - Xinyu Chen
- Division of Oncology, Department of Medicine, Stanford University, Stanford, CA, 94305, USA
| | - Ling Tong
- Division of Oncology, Department of Medicine, Stanford University, Stanford, CA, 94305, USA
| | | | - John Bell
- Stanford Genome Technology Center, Stanford University, Stanford, CA, 94305, USA
| | - Adriane Mosley
- Division of Oncology, Department of Medicine, Stanford University, Stanford, CA, 94305, USA
| | - Angela N Koehler
- Koch Institute for Integrative Cancer Research at MIT, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Center for Precision Cancer Medicine, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
| | - Yulin Li
- Division of Oncology, Department of Medicine, Stanford University, Stanford, CA, 94305, USA.
- Institute for Academic Medicine, Houston Methodist and Weill Cornell Medical College, Houston, TX, 77030, USA.
| | - Dean W Felsher
- Division of Oncology, Department of Medicine, Stanford University, Stanford, CA, 94305, USA.
- Department of Pathology, Stanford University, Stanford, CA, 94305, USA.
- Stanford Cancer Institute, Stanford University, Stanford, CA, 94305, USA.
| |
Collapse
|
11
|
Hasan A, Khan NA, Uddin S, Khan AQ, Steinhoff M. Deregulated transcription factors in the emerging cancer hallmarks. Semin Cancer Biol 2024; 98:31-50. [PMID: 38123029 DOI: 10.1016/j.semcancer.2023.12.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 11/25/2023] [Accepted: 12/14/2023] [Indexed: 12/23/2023]
Abstract
Cancer progression is a multifaceted process that entails several stages and demands the persistent expression or activation of transcription factors (TFs) to facilitate growth and survival. TFs are a cluster of proteins with DNA-binding domains that attach to promoter or enhancer DNA strands to start the transcription of genes by collaborating with RNA polymerase and other supporting proteins. They are generally acknowledged as the major regulatory molecules that coordinate biological homeostasis and the appropriate functioning of cellular components, subsequently contributing to human physiology. TFs proteins are crucial for controlling transcription during the embryonic stage and development, and the stability of different cell types depends on how they function in different cell types. The development and progression of cancer cells and tumors might be triggered by any anomaly in transcription factor function. It has long been acknowledged that cancer development is accompanied by the dysregulated activity of TF alterations which might result in faulty gene expression. Recent studies have suggested that dysregulated transcription factors play a major role in developing various human malignancies by altering and rewiring metabolic processes, modifying the immune response, and triggering oncogenic signaling cascades. This review emphasizes the interplay between TFs involved in metabolic and epigenetic reprogramming, evading immune attacks, cellular senescence, and the maintenance of cancer stemness in cancerous cells. The insights presented herein will facilitate the development of innovative therapeutic modalities to tackle the dysregulated transcription factors underlying cancer.
Collapse
Affiliation(s)
- Adria Hasan
- Molecular Cell Biology Laboratory, Integral Information and Research Centre-4 (IIRC-4), Integral University, Lucknow 226026, India; Department of Bioengineering, Faculty of Engineering, Integral University, Lucknow 226026, India
| | - Naushad Ahmad Khan
- Department of Surgery, Trauma and Vascular Surgery Clinical Research, Hamad General Hospital, Doha 3050, Qatar
| | - Shahab Uddin
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha 3050, Qatar; Department of Biosciences, Integral University, Lucknow 226026, India; Animal Research Center, Qatar University, Doha, Qatar; Dermatology Institute, Academic Health System, Hamad Medical Corporation, Doha 3050, Qatar
| | - Abdul Q Khan
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha 3050, Qatar.
| | - Martin Steinhoff
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha 3050, Qatar; Animal Research Center, Qatar University, Doha, Qatar; Department of Dermatology and Venereology, Rumailah Hospital, Hamad Medical Corporation, Doha 3050, Qatar; Department of Medicine, Weill Cornell Medicine Qatar, Qatar Foundation-Education City, Doha 24144, Qatar; Department of Medicine, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA; College of Medicine, Qatar University, Doha 2713, Qatar
| |
Collapse
|
12
|
Bhin J, Yemelyanenko J, Chao X, Klarenbeek S, Opdam M, Malka Y, Hoekman L, Kruger D, Bleijerveld O, Brambillasca CS, Sprengers J, Siteur B, Annunziato S, van Haren MJ, Martin NI, van de Ven M, Peters D, Agami R, Linn SC, Boven E, Altelaar M, Jonkers J, Zingg D, Wessels LF. MYC is a clinically significant driver of mTOR inhibitor resistance in breast cancer. J Exp Med 2023; 220:e20211743. [PMID: 37642941 PMCID: PMC10465700 DOI: 10.1084/jem.20211743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 05/18/2023] [Accepted: 07/24/2023] [Indexed: 08/31/2023] Open
Abstract
Targeting the PI3K-AKT-mTOR pathway is a promising therapeutic strategy for breast cancer treatment. However, low response rates and development of resistance to PI3K-AKT-mTOR inhibitors remain major clinical challenges. Here, we show that MYC activation drives resistance to mTOR inhibitors (mTORi) in breast cancer. Multiomic profiling of mouse invasive lobular carcinoma (ILC) tumors revealed recurrent Myc amplifications in tumors that acquired resistance to the mTORi AZD8055. MYC activation was associated with biological processes linked to mTORi response and counteracted mTORi-induced translation inhibition by promoting translation of ribosomal proteins. In vitro and in vivo induction of MYC conferred mTORi resistance in mouse and human breast cancer models. Conversely, AZD8055-resistant ILC cells depended on MYC, as demonstrated by the synergistic effects of mTORi and MYCi combination treatment. Notably, MYC status was significantly associated with poor response to everolimus therapy in metastatic breast cancer patients. Thus, MYC is a clinically relevant driver of mTORi resistance that may stratify breast cancer patients for mTOR-targeted therapies.
Collapse
Affiliation(s)
- Jinhyuk Bhin
- Division of Molecular Carcinogenesis, Netherlands Cancer Institute, Amsterdam, Netherlands
- Division of Molecular Pathology, Netherlands Cancer Institute, Amsterdam, Netherlands
- Oncode Institute, Utrecht, Netherlands
- Department of Biomedical System Informatics, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Julia Yemelyanenko
- Division of Molecular Pathology, Netherlands Cancer Institute, Amsterdam, Netherlands
- Oncode Institute, Utrecht, Netherlands
| | - Xue Chao
- Division of Molecular Pathology, Netherlands Cancer Institute, Amsterdam, Netherlands
- Oncode Institute, Utrecht, Netherlands
| | - Sjoerd Klarenbeek
- Experimental Animal Pathology, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Mark Opdam
- Division of Molecular Pathology, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Yuval Malka
- Oncode Institute, Utrecht, Netherlands
- Division of Oncogenomics, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Liesbeth Hoekman
- Proteomics Facility, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Dinja Kruger
- Division of Molecular Pathology, Netherlands Cancer Institute, Amsterdam, Netherlands
- Department of Medical Oncology, Amsterdam University Medical Center, Vrije Universiteit Amsterdam/Cancer Center Amsterdam, Amsterdam, Netherlands
| | - Onno Bleijerveld
- Proteomics Facility, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Chiara S. Brambillasca
- Division of Molecular Pathology, Netherlands Cancer Institute, Amsterdam, Netherlands
- Oncode Institute, Utrecht, Netherlands
| | - Justin Sprengers
- Mouse Clinic for Cancer and Aging, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Bjørn Siteur
- Mouse Clinic for Cancer and Aging, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Stefano Annunziato
- Division of Molecular Pathology, Netherlands Cancer Institute, Amsterdam, Netherlands
- Oncode Institute, Utrecht, Netherlands
| | - Matthijs J. van Haren
- Biological Chemistry Group, Institute of Biology Leiden, Leiden University, Leiden, Netherlands
| | - Nathaniel I. Martin
- Biological Chemistry Group, Institute of Biology Leiden, Leiden University, Leiden, Netherlands
| | - Marieke van de Ven
- Mouse Clinic for Cancer and Aging, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Dennis Peters
- Core Facility Molecular Pathology and Biobanking, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Reuven Agami
- Oncode Institute, Utrecht, Netherlands
- Division of Oncogenomics, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Sabine C. Linn
- Division of Molecular Pathology, Netherlands Cancer Institute, Amsterdam, Netherlands
- Department of Pathology, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Epie Boven
- Department of Medical Oncology, Amsterdam University Medical Center, Vrije Universiteit Amsterdam/Cancer Center Amsterdam, Amsterdam, Netherlands
| | - Maarten Altelaar
- Proteomics Facility, Netherlands Cancer Institute, Amsterdam, Netherlands
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands
- Netherlands Proteomics Centre, Utrecht, Netherlands
| | - Jos Jonkers
- Division of Molecular Pathology, Netherlands Cancer Institute, Amsterdam, Netherlands
- Oncode Institute, Utrecht, Netherlands
| | - Daniel Zingg
- Division of Molecular Pathology, Netherlands Cancer Institute, Amsterdam, Netherlands
- Oncode Institute, Utrecht, Netherlands
| | - Lodewyk F.A. Wessels
- Division of Molecular Carcinogenesis, Netherlands Cancer Institute, Amsterdam, Netherlands
- Oncode Institute, Utrecht, Netherlands
| |
Collapse
|
13
|
Brown‐Suedel AN, Bouchier‐Hayes L. Got PIDD1? Natural killer cells clear polyploid cells to ensure a balanced genome. EMBO J 2023; 42:e115307. [PMID: 37691515 PMCID: PMC10577535 DOI: 10.15252/embj.2023115307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Accepted: 09/01/2023] [Indexed: 09/12/2023] Open
Abstract
Removal of polyploid cells is essential to preventing cancer and restricting tumor growth. A new study published in The EMBO Journal shows assembly of the NEMO-PIDDosome on extra centrioles. Activation of this protein complex leads to NF-κB activation that, in turn, induces NK cell-mediated cell clearance.
Collapse
Affiliation(s)
| | - Lisa Bouchier‐Hayes
- Department of Pediatrics, Division of Hematology‐OncologyBaylor College of MedicineHoustonTXUSA
| |
Collapse
|
14
|
Wang H, Lu J, Stevens T, Roberts A, Mandel J, Avula R, Ma B, Wu Y, Wang J, Land CV, Finkel T, Vockley JE, Airik M, Airik R, Muzumdar R, Gong Z, Torbenson MS, Prochownik EV. Premature aging and reduced cancer incidence associated with near-complete body-wide Myc inactivation. Cell Rep 2023; 42:112830. [PMID: 37481724 PMCID: PMC10591215 DOI: 10.1016/j.celrep.2023.112830] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 05/18/2023] [Accepted: 07/03/2023] [Indexed: 07/25/2023] Open
Abstract
MYC proto-oncogene dysregulation alters metabolism, translation, and other functions in ways that support tumor induction and maintenance. Although Myc+/- mice are healthier and longer-lived than control mice, the long-term ramifications of more complete Myc loss remain unknown. We now describe the chronic consequences of body-wide Myc inactivation initiated postnatally. "MycKO" mice acquire numerous features of premature aging, including altered body composition and habitus, metabolic dysfunction, hepatic steatosis, and dysregulation of gene sets involved in functions that normally deteriorate with aging. Yet, MycKO mice have extended lifespans that correlate with a 3- to 4-fold lower lifetime cancer incidence. Aging tissues from normal mice and humans also downregulate Myc and gradually alter many of the same Myc target gene sets seen in MycKO mice. Normal aging and its associated cancer predisposition are thus highly linked via Myc.
Collapse
Affiliation(s)
- Huabo Wang
- Division of Hematology/Oncology, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA 15224, USA
| | - Jie Lu
- Division of Hematology/Oncology, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA 15224, USA
| | - Taylor Stevens
- Division of Hematology/Oncology, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA 15224, USA
| | - Alexander Roberts
- Division of Hematology/Oncology, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA 15224, USA
| | - Jordan Mandel
- Division of Hematology/Oncology, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA 15224, USA
| | - Raghunandan Avula
- Division of Hematology/Oncology, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA 15224, USA; The University of Pittsburgh School of Medicine, Pittsburgh, PA 15224, USA
| | - Bingwei Ma
- Tongji University School of Medicine, Shanghai, China
| | - Yijen Wu
- Department of Developmental Biology, The University of Pittsburgh, Pittsburgh, PA, USA
| | - Jinglin Wang
- Division of Hematology/Oncology, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA 15224, USA; Central South University, Xiangya School of Medicine, Changsha, Hunan 410013, P.R. China
| | - Clinton Van't Land
- Division of Medical Genetics, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA 15224, USA
| | - Toren Finkel
- Division of Cardiology, The Department of Internal Medicine and the UPMC Aging Institute, Pittsburgh, PA 15224, USA
| | - Jerry E Vockley
- Division of Medical Genetics, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA 15224, USA
| | - Merlin Airik
- Division of Nephrology, Children's Hospital of Pittsburgh, Pittsburgh, PA 15224, USA
| | - Rannar Airik
- Division of Nephrology, Children's Hospital of Pittsburgh, Pittsburgh, PA 15224, USA
| | - Radhika Muzumdar
- Division of Endocrinology, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA 15224, USA
| | - Zhenwei Gong
- Division of Endocrinology, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA 15224, USA
| | - Michel S Torbenson
- Division of Laboratory Medicine and Pathology, The Mayo Clinic, Rochester, MN 55905, USA
| | - Edward V Prochownik
- Division of Hematology/Oncology, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA 15224, USA; Department of Microbiology and Molecular Genetics, UPMC, Pittsburgh, PA 15261, USA; Hillman Cancer Center of UPMC, Pittsburgh, PA 15232, USA; Pittsburgh Liver Research Center, UPMC, Pittsburgh, PA 15261, USA.
| |
Collapse
|
15
|
Khameneh HJ, Fonta N, Zenobi A, Niogret C, Ventura P, Guerra C, Kwee I, Rinaldi A, Pecoraro M, Geiger R, Cavalli A, Bertoni F, Vivier E, Trumpp A, Guarda G. Myc controls NK cell development, IL-15-driven expansion, and translational machinery. Life Sci Alliance 2023; 6:e202302069. [PMID: 37105715 PMCID: PMC10140547 DOI: 10.26508/lsa.202302069] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Revised: 04/19/2023] [Accepted: 04/19/2023] [Indexed: 04/29/2023] Open
Abstract
MYC is a pleiotropic transcription factor involved in cancer, cell proliferation, and metabolism. Its regulation and function in NK cells, which are innate cytotoxic lymphocytes important to control viral infections and cancer, remain poorly defined. Here, we show that mice deficient for Myc in NK cells presented a severe reduction in these lymphocytes. Myc was required for NK cell development and expansion in response to the key cytokine IL-15, which induced Myc through transcriptional and posttranslational mechanisms. Mechanistically, Myc ablation in vivo largely impacted NK cells' ribosomagenesis, reducing their translation and expansion capacities. Similar results were obtained by inhibiting MYC in human NK cells. Impairing translation by pharmacological intervention phenocopied the consequences of deleting or blocking MYC in vitro. Notably, mice lacking Myc in NK cells exhibited defective anticancer immunity, which reflected their decreased numbers of mature NK cells exerting suboptimal cytotoxic functions. These results indicate that MYC is a central node in NK cells, connecting IL-15 to translational fitness, expansion, and anticancer immunity.
Collapse
Affiliation(s)
- Hanif J Khameneh
- Università della Svizzera italiana, Faculty of Biomedical Sciences, Institute for Research in Biomedicine, Bellinzona, Switzerland
| | - Nicolas Fonta
- Università della Svizzera italiana, Faculty of Biomedical Sciences, Institute for Research in Biomedicine, Bellinzona, Switzerland
| | - Alessandro Zenobi
- Università della Svizzera italiana, Faculty of Biomedical Sciences, Institute for Research in Biomedicine, Bellinzona, Switzerland
| | - Charlène Niogret
- Department of Biochemistry, University of Lausanne, Epalinges, Switzerland
| | - Pedro Ventura
- Università della Svizzera italiana, Faculty of Biomedical Sciences, Institute for Research in Biomedicine, Bellinzona, Switzerland
| | - Concetta Guerra
- Università della Svizzera italiana, Faculty of Biomedical Sciences, Institute for Research in Biomedicine, Bellinzona, Switzerland
| | - Ivo Kwee
- BigOmics Analytics SA, Lugano, Switzerland
| | - Andrea Rinaldi
- Università della Svizzera italiana, Faculty of Biomedical Sciences, Institute of Oncology Research, Bellinzona, Switzerland
| | - Matteo Pecoraro
- Università della Svizzera italiana, Faculty of Biomedical Sciences, Institute for Research in Biomedicine, Bellinzona, Switzerland
| | - Roger Geiger
- Università della Svizzera italiana, Faculty of Biomedical Sciences, Institute for Research in Biomedicine, Bellinzona, Switzerland
- Università della Svizzera italiana, Faculty of Biomedical Sciences, Institute of Oncology Research, Bellinzona, Switzerland
| | - Andrea Cavalli
- Università della Svizzera italiana, Faculty of Biomedical Sciences, Institute for Research in Biomedicine, Bellinzona, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Francesco Bertoni
- Università della Svizzera italiana, Faculty of Biomedical Sciences, Institute of Oncology Research, Bellinzona, Switzerland
- Oncology Institute of Southern Switzerland, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
| | - Eric Vivier
- Aix-Marseille Université, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Centre d'Immunologie de Marseille-Luminy, Marseille, France
- Innate Pharma Research Laboratories, Marseille, France
- APHM, Hôpital de la Timone, Marseille-Immunopôle, Marseille, France
| | - Andreas Trumpp
- Division of Stem Cells and Cancer, DKFZ, Heidelberg, Germany
- HI-STEM: The Heidelberg Institute for Stem Cell Technology and Experimental Medicine gGmbH, Heidelberg, Germany
| | - Greta Guarda
- Università della Svizzera italiana, Faculty of Biomedical Sciences, Institute for Research in Biomedicine, Bellinzona, Switzerland
| |
Collapse
|
16
|
Merino A, Maakaron J, Bachanova V. Advances in NK cell therapy for hematologic malignancies: NK source, persistence and tumor targeting. Blood Rev 2023; 60:101073. [PMID: 36959057 PMCID: PMC10979648 DOI: 10.1016/j.blre.2023.101073] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Revised: 03/07/2023] [Accepted: 03/09/2023] [Indexed: 03/14/2023]
Abstract
Natural Killer (NK) cells yield promise in therapy of hematologic malignancies. The clinical experience with adoptively transferred allogeneic NK cells over past two decades has revealed safety and minimal risk of CRS or ICANS. Unlike T cells which have to be genetically altered to avoid graft vs host disease (GVHD), HLA mismatched NK cells can be infused without GVHD risk. This makes them ideal for the development of off-the-shelf products. In this review we focus on NK biology relevant to the cancer therapy, the trajectory of NK therapeutics for leukemia, lymphoma, and myeloma; and advantages of the NK cell platform. We will also discuss novel methods to enhance NK cell targeting, persistence, and function in the tumor microenvironment. The future of NK cell therapy depends on novel strategies to realize these qualities.
Collapse
Affiliation(s)
- Aimee Merino
- Division of Hematology, Oncology, and Transplantation, University of Minnesota, 420 Delaware St, Minneapolis, MN, United States of America
| | - Joseph Maakaron
- Division of Hematology, Oncology, and Transplantation, University of Minnesota, 420 Delaware St, Minneapolis, MN, United States of America
| | - Veronika Bachanova
- Division of Hematology, Oncology, and Transplantation, University of Minnesota, 420 Delaware St, Minneapolis, MN, United States of America.
| |
Collapse
|
17
|
Lordo MR, Stiff AR, Oakes CC, Mundy-Bosse BL. Effects of epigenetic therapy on natural killer cell function and development in hematologic malignancy. J Leukoc Biol 2023; 113:518-524. [PMID: 36860165 PMCID: PMC10443672 DOI: 10.1093/jleuko/qiad026] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 02/16/2023] [Accepted: 02/22/2023] [Indexed: 03/03/2023] Open
Abstract
Epigenetic therapy is an emerging field in the treatment of human cancer, including hematologic malignancies. This class of therapeutic agents approved by the US Food and Drug Administration for cancer treatment includes DNA hypomethylating agents, histone deacetylase inhibitors, IDH1/2 inhibitors, EZH2 inhibitors, and numerous preclinical targets/agents. Most studies measuring the biological effects of epigenetic therapy focus their attention on either their direct cytotoxic effects on malignant cells or their effects on modifying tumor cell antigen expression, exposing them to immune surveillance mechanisms. However, a growing body of evidence suggests that epigenetic therapy also has effects on the development and function of the immune system, including natural killer cells, which can alter their response to cancer cells. In this review, we summarize the body of literature studying the effects of different classes of epigenetic therapy on the development and/or function of natural killer cells.
Collapse
Affiliation(s)
- Matthew R. Lordo
- Comprehensive Cancer Center, The Ohio State University, 460 W. 10th Avenue, Columbus, OH 43210, USA
- Medical Scientist Training Program, Biomedical Sciences Graduate Program, The Ohio State University, 370 W. 9th Avenue, Columbus, OH 43210, USA
| | - Andrew R. Stiff
- Comprehensive Cancer Center, The Ohio State University, 460 W. 10th Avenue, Columbus, OH 43210, USA
- Physician Scientist Training Program, The Ohio State University, 370 W. 9th Avenue, Columbus, OH 43210, USA
- Division of Hematology, Department of Internal Medicine, The Ohio State University Wexner Medical Center, 460 W. 12th Avenue, Columbus, OH 43210, USA
| | - Christopher C. Oakes
- Comprehensive Cancer Center, The Ohio State University, 460 W. 10th Avenue, Columbus, OH 43210, USA
- Division of Hematology, Department of Internal Medicine, The Ohio State University Wexner Medical Center, 460 W. 12th Avenue, Columbus, OH 43210, USA
| | - Bethany L. Mundy-Bosse
- Comprehensive Cancer Center, The Ohio State University, 460 W. 10th Avenue, Columbus, OH 43210, USA
- Division of Hematology, Department of Internal Medicine, The Ohio State University Wexner Medical Center, 460 W. 12th Avenue, Columbus, OH 43210, USA
| |
Collapse
|
18
|
Kumar A, Taghi Khani A, Duault C, Aramburo S, Sanchez Ortiz A, Lee SJ, Chan A, McDonald T, Huang M, Lacayo NJ, Sakamoto KM, Yu J, Hurtz C, Carroll M, Tasian SK, Ghoda L, Marcucci G, Gu Z, Rosen ST, Armenian S, Izraeli S, Chen CW, Caligiuri MA, Forman SJ, Maecker HT, Swaminathan S. Intrinsic suppression of type I interferon production underlies the therapeutic efficacy of IL-15-producing natural killer cells in B-cell acute lymphoblastic leukemia. J Immunother Cancer 2023; 11:jitc-2022-006649. [PMID: 37217248 DOI: 10.1136/jitc-2022-006649] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/03/2023] [Indexed: 05/24/2023] Open
Abstract
BACKGROUND Type I interferons (IFN-Is), secreted by hematopoietic cells, drive immune surveillance of solid tumors. However, the mechanisms of suppression of IFN-I-driven immune responses in hematopoietic malignancies including B-cell acute lymphoblastic leukemia (B-ALL) are unknown. METHODS Using high-dimensional cytometry, we delineate the defects in IFN-I production and IFN-I-driven immune responses in high-grade primary human and mouse B-ALLs. We develop natural killer (NK) cells as therapies to counter the intrinsic suppression of IFN-I production in B-ALL. RESULTS We find that high expression of IFN-I signaling genes predicts favorable clinical outcome in patients with B-ALL, underscoring the importance of the IFN-I pathway in this malignancy. We show that human and mouse B-ALL microenvironments harbor an intrinsic defect in paracrine (plasmacytoid dendritic cell) and/or autocrine (B-cell) IFN-I production and IFN-I-driven immune responses. Reduced IFN-I production is sufficient for suppressing the immune system and promoting leukemia development in mice prone to MYC-driven B-ALL. Among anti-leukemia immune subsets, suppression of IFN-I production most markedly lowers the transcription of IL-15 and reduces NK-cell number and effector maturation in B-ALL microenvironments. Adoptive transfer of healthy NK cells significantly prolongs survival of overt ALL-bearing transgenic mice. Administration of IFN-Is to B-ALL-prone mice reduces leukemia progression and increases the frequencies of total NK and NK-cell effectors in circulation. Ex vivo treatment of malignant and non-malignant immune cells in primary mouse B-ALL microenvironments with IFN-Is fully restores proximal IFN-I signaling and partially restores IL-15 production. In B-ALL patients, the suppression of IL-15 is the most severe in difficult-to-treat subtypes with MYC overexpression. MYC overexpression promotes sensitivity of B-ALL to NK cell-mediated killing. To counter the suppressed IFN-I-induced IL-15 production in MYChigh human B-ALL, we CRISPRa-engineered a novel human NK-cell line that secretes IL-15. CRISPRa IL-15-secreting human NK cells kill high-grade human B-ALL in vitro and block leukemia progression in vivo more effectively than NK cells that do not produce IL-15. CONCLUSION We find that restoration of the intrinsically suppressed IFN-I production in B-ALL underlies the therapeutic efficacy of IL-15-producing NK cells and that such NK cells represent an attractive therapeutic solution for the problem of drugging MYC in high-grade B-ALL.
Collapse
Affiliation(s)
- Anil Kumar
- Department of Systems Biology, City of Hope Beckman Research Institute, Monrovia, California, USA
| | - Adeleh Taghi Khani
- Department of Systems Biology, City of Hope Beckman Research Institute, Monrovia, California, USA
| | - Caroline Duault
- The Human Immune Monitoring Center (HIMC), Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, California, USA
| | - Soraya Aramburo
- Department of Systems Biology, City of Hope Beckman Research Institute, Monrovia, California, USA
| | - Ashly Sanchez Ortiz
- Department of Systems Biology, City of Hope Beckman Research Institute, Monrovia, California, USA
| | - Sung June Lee
- Department of Systems Biology, City of Hope Beckman Research Institute, Monrovia, California, USA
| | - Anthony Chan
- Department of Systems Biology, City of Hope Beckman Research Institute, Monrovia, California, USA
| | - Tinisha McDonald
- The Hematopoietic Tissue Biorepository/Research Pathology Shared Resources, City of Hope, Duarte, California, USA
| | - Min Huang
- Department of Pediatrics, Stanford University School of Medicine, Stanford, California, USA
| | - Norman J Lacayo
- Department of Pediatrics, Stanford University School of Medicine, Stanford, California, USA
| | - Kathleen M Sakamoto
- Department of Pediatrics, Stanford University School of Medicine, Stanford, California, USA
| | - Jianhua Yu
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Duarte, California, USA
| | - Christian Hurtz
- Department of Cancer and Cellular Biology, Fels Cancer Institute for Personalized Medicine Temple University Lewis Katz School of Medicine, Philadelphia, Pennsylvania, USA
| | - Martin Carroll
- Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Sarah K Tasian
- Department of Pediatrics, Division of Oncology, The Children's Hospital, Philadelphia, Pennsylvania, USA
| | - Lucy Ghoda
- Hematological Malignancies Translational Science, City of Hope, Duarte, California, USA
| | - Guido Marcucci
- The Hematopoietic Tissue Biorepository/Research Pathology Shared Resources, City of Hope, Duarte, California, USA
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Duarte, California, USA
- Hematological Malignancies Translational Science, City of Hope, Duarte, California, USA
| | - Zhaohui Gu
- Department of Systems Biology, City of Hope Beckman Research Institute, Monrovia, California, USA
| | - Steven T Rosen
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Duarte, California, USA
| | - Saro Armenian
- Department of Pediatrics, City of Hope, Duarte, California, USA
| | - Shai Izraeli
- Department of Systems Biology, City of Hope Beckman Research Institute, Monrovia, California, USA
- Hematology-Oncology Department, Tel Aviv University, Tel Aviv, Israel
| | - Chun-Wei Chen
- Department of Systems Biology, City of Hope Beckman Research Institute, Monrovia, California, USA
| | - Michael A Caligiuri
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Duarte, California, USA
| | - Stephen J Forman
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Duarte, California, USA
| | - Holden T Maecker
- The Human Immune Monitoring Center (HIMC), Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, California, USA
| | - Srividya Swaminathan
- Department of Systems Biology, City of Hope Beckman Research Institute, Monrovia, California, USA
- Department of Pediatrics, City of Hope, Duarte, California, USA
| |
Collapse
|
19
|
Wang Y, Ju L, Wang G, Qian K, Jin W, Li M, Yu J, Shi Y, Wang Y, Zhang Y, Xiao Y, Wang X. DNA polymerase POLD1 promotes proliferation and metastasis of bladder cancer by stabilizing MYC. Nat Commun 2023; 14:2421. [PMID: 37105989 PMCID: PMC10140023 DOI: 10.1038/s41467-023-38160-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 04/17/2023] [Indexed: 04/29/2023] Open
Abstract
To date, most studies on the DNA polymerase, POLD1, have focused on the effect of POLD1 inactivation mutations in tumors. However, the implications of high POLD1 expression in tumorigenesis remains elusive. Here, we determine that POLD1 has a pro-carcinogenic role in bladder cancer (BLCA) and is associated to the malignancy and prognosis of BLCA. Our studies demonstrate that POLD1 promotes the proliferation and metastasis of BLCA via MYC. Mechanistically, POLD1 stabilizes MYC in a manner independent of its' DNA polymerase activity. Instead, POLD1 attenuates FBXW7-mediated ubiquitination degradation of MYC by directly binding to the MYC homology box 1 domain competitively with FBXW7. Moreover, we find that POLD1 forms a complex with MYC to promote the transcriptional activity of MYC. In turn, MYC increases expression of POLD1, forming a POLD1-MYC positive feedback loop to enhance the pro-carcinogenic effect of POLD1-MYC on BLCA. Overall, our study identifies POLD1 as a promotor of BCLA via a MYC driven mechanism and suggest its potential as biomarker for BLCA.
Collapse
Affiliation(s)
- Yejinpeng Wang
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Lingao Ju
- Department of Biological Repositories, Zhongnan Hospital of Wuhan University, Wuhan, China
- Human Genetic Resources Preservation Center of Hubei Province, Wuhan, China
- Wuhan Research Center for Infectious Diseases and Cancer, Chinese Academy of Medical Sciences, Wuhan, China
| | - Gang Wang
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China
- Department of Biological Repositories, Zhongnan Hospital of Wuhan University, Wuhan, China
- Human Genetic Resources Preservation Center of Hubei Province, Wuhan, China
| | - Kaiyu Qian
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China
- Department of Biological Repositories, Zhongnan Hospital of Wuhan University, Wuhan, China
- Human Genetic Resources Preservation Center of Hubei Province, Wuhan, China
| | - Wan Jin
- Euler Technology, ZGC Life Sciences Park, Beijing, China
| | - Mingxing Li
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Jingtian Yu
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yiliang Shi
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yongzhi Wang
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yi Zhang
- Euler Technology, ZGC Life Sciences Park, Beijing, China.
- Center for Quantitative Biology, School of Life Sciences, Peking University, Beijing, China.
| | - Yu Xiao
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China.
- Department of Biological Repositories, Zhongnan Hospital of Wuhan University, Wuhan, China.
- Human Genetic Resources Preservation Center of Hubei Province, Wuhan, China.
- Wuhan Research Center for Infectious Diseases and Cancer, Chinese Academy of Medical Sciences, Wuhan, China.
| | - Xinghuan Wang
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China.
- Wuhan Research Center for Infectious Diseases and Cancer, Chinese Academy of Medical Sciences, Wuhan, China.
- Medical Research Institute, Wuhan University, Wuhan, China.
- Institute of Urology, Wuhan University, Wuhan, China.
| |
Collapse
|
20
|
Qin A. An anti-cancer surveillance by the interplay between interferon-beta and retinoblastoma protein RB1. Front Oncol 2023; 13:1173467. [PMID: 37182173 PMCID: PMC10174298 DOI: 10.3389/fonc.2023.1173467] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Accepted: 04/05/2023] [Indexed: 05/16/2023] Open
Abstract
Interferon-beta (IFN-β), an extracellular cytokine that initiates signaling pathways for gene regulation, has been demonstrated to function as a tumor suppressor protein through lentiviral gene transduction. In this article, I review the relevant previous works and propose a cell cycle-based, tumor suppressor protein-mediated mechanism of anti-cancer surveillance. IFN-β induces a tumor cell cycle alteration that leads to S phase accumulation, senescence entry, and a loss of tumorigenicity in solid tumor cells. IFN-β does not show a significant cell cycle effect in their normal counterparts. Retinoblastoma protein RB1, another tumor suppressor protein, tightly controls the cell cycle and differentiation of normal cells, preventing them from being significantly impacted by the IFN-β effect. The interplay between IFN-β and RB1 acts as a mechanism of cell cycle-based, tumor suppressor protein-mediated anti-cancer surveillance that can selectively suppress solid tumor or proliferating transformed cells from the loss of control leading to cancer. This mechanism has important implications for the treatment of solid tumors.
Collapse
Affiliation(s)
- Albert Qin
- Medical Research & Clinical Operations, PharmaEssentia Corporation, Taipei, Taiwan
| |
Collapse
|
21
|
Cai Y, Chen X, Lu T, Yu Z, Hu S, Liu J, Zhou X, Wang X. Single-cell transcriptome analysis profiles the expression features of TMEM173 in BM cells of high-risk B-cell acute lymphoblastic leukemia. BMC Cancer 2023; 23:372. [PMID: 37095455 PMCID: PMC10123968 DOI: 10.1186/s12885-023-10830-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Accepted: 04/08/2023] [Indexed: 04/26/2023] Open
Abstract
BACKGROUND As an essential regulator of type I interferon (IFN) response, TMEM173 participates in immune regulation and cell death induction. In recent studies, activation of TMEM173 has been regarded as a promising strategy for cancer immunotherapy. However, transcriptomic features of TMEM173 in B-cell acute lymphoblastic leukemia (B-ALL) remain elusive. METHODS Quantitative real-time PCR (qRT-PCR) and western blotting (WB) were applied to determine the mRNA and protein levels of TMEM173 in peripheral blood mononuclear cells (PBMCs). TMEM173 mutation status was assessed by Sanger sequencing. Single-cell RNA sequencing (scRNA-seq) analysis was performed to explore the expression of TMEM173 in different types of bone marrow (BM) cells. RESULTS The mRNA and protein levels of TMEM173 were increased in PBMCs from B-ALL patients. Besides, frameshift mutation was presented in TMEM173 sequences of 2 B-ALL patients. ScRNA-seq analysis identified the specific transcriptome profiles of TMEM173 in the BM of high-risk B-ALL patients. Specifically, expression levels of TMEM173 in granulocytes, progenitor cells, mast cells, and plasmacytoid dendritic cells (pDCs) were higher than that in B cells, T cells, natural killer (NK) cells, and dendritic cells (DCs). Subset analysis further revealed that TMEM173 and pyroptosis effector gasdermin D (GSDMD) restrained in precursor-B (pre-B) cells with proliferative features, which expressed nuclear factor kappa-B (NF-κB), CD19, and Bruton's tyrosine kinase (BTK) during the progression of B-ALL. In addition, TMEM173 was associated with the functional activation of NK cells and DCs in B-ALL. CONCLUSIONS Our findings provide insights into the transcriptomic features of TMEM173 in the BM of high-risk B-ALL patients. Targeted activation of TMEM173 in specific cells might provide new therapeutic strategies for B-ALL patients.
Collapse
Affiliation(s)
- Yiqing Cai
- Department of Hematology, Shandong Provincial Hospital, Shandong University, No.324, Jingwu Road, Jinan, Shandong, 250021, China
| | - Xiaomin Chen
- Department of Hematology, Shandong Provincial Hospital, Shandong University, No.324, Jingwu Road, Jinan, Shandong, 250021, China
| | - Tiange Lu
- Department of Hematology, Shandong Provincial Hospital, Shandong University, No.324, Jingwu Road, Jinan, Shandong, 250021, China
| | - Zhuoya Yu
- Department of Hematology, Shandong Provincial Hospital, Shandong University, No.324, Jingwu Road, Jinan, Shandong, 250021, China
| | - Shunfeng Hu
- Department of Hematology, Shandong Provincial Hospital, Shandong University, No.324, Jingwu Road, Jinan, Shandong, 250021, China
| | - Jiarui Liu
- Department of Hematology, Shandong Provincial Hospital, Shandong University, No.324, Jingwu Road, Jinan, Shandong, 250021, China
| | - Xiangxiang Zhou
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No.324, Jingwu Road, Jinan, Shandong, 250021, China.
- Shandong Provincial Engineering Research Center of Lymphoma, Jinan, Shandong, 250021, China.
- Branch of National Clinical Research Center for Hematologic Diseases, Jinan, Shandong, 250021, China.
- National Clinical Research Center for Hematologic Diseases, the First Affiliated Hospital of Soochow University, Suzhou, 251006, China.
| | - Xin Wang
- Department of Hematology, Shandong Provincial Hospital, Shandong University, No.324, Jingwu Road, Jinan, Shandong, 250021, China.
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No.324, Jingwu Road, Jinan, Shandong, 250021, China.
- Shandong Provincial Engineering Research Center of Lymphoma, Jinan, Shandong, 250021, China.
- Branch of National Clinical Research Center for Hematologic Diseases, Jinan, Shandong, 250021, China.
- National Clinical Research Center for Hematologic Diseases, the First Affiliated Hospital of Soochow University, Suzhou, 251006, China.
| |
Collapse
|
22
|
Alburquerque-Bejar JJ, Navajas-Chocarro P, Saigi M, Ferrero-Andres A, Morillas JM, Vilarrubi A, Gomez A, Mate JL, Munoz-Marmol AM, Romero OA, Blecua P, Davalos V, Esteller M, Pros E, Llabata P, Torres-Diz M, Esteve-Codina A, Sanchez-Cespedes M. MYC activation impairs cell-intrinsic IFNγ signaling and confers resistance to anti-PD1/PD-L1 therapy in lung cancer. Cell Rep Med 2023; 4:101006. [PMID: 37044092 PMCID: PMC10140599 DOI: 10.1016/j.xcrm.2023.101006] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 07/29/2022] [Accepted: 03/17/2023] [Indexed: 04/14/2023]
Abstract
Elucidating the adaptive mechanisms that prevent host immune response in cancer will help predict efficacy of anti-programmed death-1 (PD1)/L1 therapies. Here, we study the cell-intrinsic response of lung cancer (LC) to interferon-γ (IFNγ), a cytokine that promotes immunoresponse and modulates programmed death-ligand 1 (PD-L1) levels. We report complete refractoriness to IFNγ in a subset of LCs as a result of JAK2 or IFNGR1 inactivation. A submaximal response affects another subset that shows constitutive low levels of IFNγ-stimulated genes (IγSGs) coupled with decreased H3K27ac (histone 3 acetylation at lysine 27) deposition and promoter hypermethylation and reduced IFN regulatory factor 1 (IRF1) recruitment to the DNA on IFNγ stimulation. Most of these are neuroendocrine small cell LCs (SCLCs) with oncogenic MYC/MYCL1/MYCN. The oncogenic activation of MYC in SCLC cells downregulates JAK2 and impairs IγSGs stimulation by IFNγ. MYC amplification tends to associate with a worse response to anti-PD1/L1 therapies. Hence alterations affecting the JAK/STAT pathway and MYC activation prevent stimulation by IFNγ and may predict anti-PD1/L1 efficacy in LC.
Collapse
Affiliation(s)
- Juan J Alburquerque-Bejar
- Cancer Genetics Group, Josep Carreras Leukaemia Research Institute (IJC), IJC Building, Germans Trias i Pujol, Ctra de Can Ruti, Camí de les Escoles s/n, 08916 Badalona, Barcelona, Spain
| | - Pablo Navajas-Chocarro
- Cancer Genetics Group, Josep Carreras Leukaemia Research Institute (IJC), IJC Building, Germans Trias i Pujol, Ctra de Can Ruti, Camí de les Escoles s/n, 08916 Badalona, Barcelona, Spain
| | - Maria Saigi
- Department of Medical Oncology, Catalan Institute of Oncology (ICO), Carretera de Canyet, s/n, 08916 Badalona, Barcelona, Spain
| | - Ana Ferrero-Andres
- Cancer Genetics Group, Josep Carreras Leukaemia Research Institute (IJC), IJC Building, Germans Trias i Pujol, Ctra de Can Ruti, Camí de les Escoles s/n, 08916 Badalona, Barcelona, Spain
| | - Juan M Morillas
- Cancer Genetics Group, Josep Carreras Leukaemia Research Institute (IJC), IJC Building, Germans Trias i Pujol, Ctra de Can Ruti, Camí de les Escoles s/n, 08916 Badalona, Barcelona, Spain
| | - Andrea Vilarrubi
- Cancer Genetics Group, Josep Carreras Leukaemia Research Institute (IJC), IJC Building, Germans Trias i Pujol, Ctra de Can Ruti, Camí de les Escoles s/n, 08916 Badalona, Barcelona, Spain
| | - Antonio Gomez
- Biosciences Department, Faculty of Sciences and Technology (FCT), University of Vic-Central University of Catalonia (UVic-UCC), Carrer de la Sagrada Familia, 7, 08500 Vic, Barcelona, Spain
| | - José L Mate
- Pathology Department, Hospital Universitari Germans Trias i Pujol, Carretera de Canyet, s/n, 08916 Badalona, Barcelona, Spain
| | - Ana M Munoz-Marmol
- Pathology Department, Hospital Universitari Germans Trias i Pujol, Carretera de Canyet, s/n, 08916 Badalona, Barcelona, Spain
| | - Octavio A Romero
- Cancer Genetics Group, Josep Carreras Leukaemia Research Institute (IJC), IJC Building, Germans Trias i Pujol, Ctra de Can Ruti, Camí de les Escoles s/n, 08916 Badalona, Barcelona, Spain
| | - Pedro Blecua
- Cancer Epigenetics Group, Josep Carreras Leukaemia Research Institute (IJC), IJC Building, Germans Trias i Pujol, Ctra de Can Ruti, Cami de les Escoles s/n, 08916 Badalona, Barcelona, Spain
| | - Veronica Davalos
- Cancer Epigenetics Group, Josep Carreras Leukaemia Research Institute (IJC), IJC Building, Germans Trias i Pujol, Ctra de Can Ruti, Cami de les Escoles s/n, 08916 Badalona, Barcelona, Spain
| | - Manel Esteller
- Cancer Epigenetics Group, Josep Carreras Leukaemia Research Institute (IJC), IJC Building, Germans Trias i Pujol, Ctra de Can Ruti, Cami de les Escoles s/n, 08916 Badalona, Barcelona, Spain; Centro de Investigacion Biomedica en Red Cancer (CIBERONC), Calle Monforte de Lemos, 3-5, Pabellon 11, Planta baja, 28029 Madrid, Spain; Institucio Catalana de Recerca i Estudis Avançats (ICREA), Passeig de Lluis Companys, 23, 08010 Barcelona, Spain; Physiological Sciences Department, School of Medicine and Health Sciences, University of Barcelona, Feixa Llarga, s/n, 08907 l'Hospitalet de Llobregat, Spain
| | - Eva Pros
- Cancer Genetics Group, Josep Carreras Leukaemia Research Institute (IJC), IJC Building, Germans Trias i Pujol, Ctra de Can Ruti, Camí de les Escoles s/n, 08916 Badalona, Barcelona, Spain
| | - Paula Llabata
- Cancer Genetics Group, Josep Carreras Leukaemia Research Institute (IJC), IJC Building, Germans Trias i Pujol, Ctra de Can Ruti, Camí de les Escoles s/n, 08916 Badalona, Barcelona, Spain
| | - Manuel Torres-Diz
- Cancer Genetics Group, Josep Carreras Leukaemia Research Institute (IJC), IJC Building, Germans Trias i Pujol, Ctra de Can Ruti, Camí de les Escoles s/n, 08916 Badalona, Barcelona, Spain
| | - Anna Esteve-Codina
- CNAG-CRG, Centre for Genomic Regulation (CRG), Institute of Science and Technology (BIST) and University Pompeu Fabra (UPF), Parc Cientific de Barcelona, Torre I Baldiri Reixac, 4, 08028 Barcelona, Spain
| | - Montse Sanchez-Cespedes
- Cancer Genetics Group, Josep Carreras Leukaemia Research Institute (IJC), IJC Building, Germans Trias i Pujol, Ctra de Can Ruti, Camí de les Escoles s/n, 08916 Badalona, Barcelona, Spain.
| |
Collapse
|
23
|
Smith BAH, Deutzmann A, Correa KM, Delaveris CS, Dhanasekaran R, Dove CG, Sullivan DK, Wisnovsky S, Stark JC, Pluvinage JV, Swaminathan S, Riley NM, Rajan A, Majeti R, Felsher DW, Bertozzi CR. MYC-driven synthesis of Siglec ligands is a glycoimmune checkpoint. Proc Natl Acad Sci U S A 2023; 120:e2215376120. [PMID: 36897988 PMCID: PMC10089186 DOI: 10.1073/pnas.2215376120] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 12/06/2022] [Indexed: 03/12/2023] Open
Abstract
The Siglecs (sialic acid-binding immunoglobulin-like lectins) are glycoimmune checkpoint receptors that suppress immune cell activation upon engagement of cognate sialoglycan ligands. The cellular drivers underlying Siglec ligand production on cancer cells are poorly understood. We find the MYC oncogene causally regulates Siglec ligand production to enable tumor immune evasion. A combination of glycomics and RNA-sequencing of mouse tumors revealed the MYC oncogene controls expression of the sialyltransferase St6galnac4 and induces a glycan known as disialyl-T. Using in vivo models and primary human leukemias, we find that disialyl-T functions as a "don't eat me" signal by engaging macrophage Siglec-E in mice or the human ortholog Siglec-7, thereby preventing cancer cell clearance. Combined high expression of MYC and ST6GALNAC4 identifies patients with high-risk cancers and reduced tumor myeloid infiltration. MYC therefore regulates glycosylation to enable tumor immune evasion. We conclude that disialyl-T is a glycoimmune checkpoint ligand. Thus, disialyl-T is a candidate for antibody-based checkpoint blockade, and the disialyl-T synthase ST6GALNAC4 is a potential enzyme target for small molecule-mediated immune therapy.
Collapse
Affiliation(s)
- Benjamin A. H. Smith
- Sarafan ChEM-H, Stanford University, Stanford, CA94305
- Department of Chemical and Systems Biology, Stanford University, Stanford, CA94305
| | - Anja Deutzmann
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, CA94305
| | | | - Corleone S. Delaveris
- Sarafan ChEM-H, Stanford University, Stanford, CA94305
- Department of Chemistry, Stanford University, Stanford, CA94305
| | - Renumathy Dhanasekaran
- Division of Gastroenterology and Hepatology, Department of Medicine, Stanford University School of Medicine, Stanford, CA94305
| | - Christopher G. Dove
- Division of Hematology, Department of Medicine, Stanford University, Stanford, CA94305
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA94305
| | - Delaney K. Sullivan
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, CA94305
| | - Simon Wisnovsky
- Faculty of Pharmaceutical Sciences, University of British Columbia, British Columbia, BC V6T 1Z3, Canada
| | - Jessica C. Stark
- Sarafan ChEM-H, Stanford University, Stanford, CA94305
- Department of Chemistry, Stanford University, Stanford, CA94305
| | - John V. Pluvinage
- Department of Neurology, University of California, San Francisco, CA94143
| | - Srividya Swaminathan
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, CA94305
- Department of Systems Biology, Beckman Research Institute of City of Hope, Monrovia, CA91016
- Department of Pediatrics, Beckman Research Institute of City of Hope, Duarte, CA91010
| | | | - Anand Rajan
- Department of Pathology, University of Iowa, Iowa City, IA52242
| | - Ravindra Majeti
- Division of Hematology, Department of Medicine, Stanford University, Stanford, CA94305
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA94305
| | - Dean W. Felsher
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, CA94305
- Department of Pathology, Stanford University School of Medicine, Stanford, CA94305
| | - Carolyn R. Bertozzi
- Sarafan ChEM-H, Stanford University, Stanford, CA94305
- Department of Chemistry, Stanford University, Stanford, CA94305
- Howard Hughes Medical Institute, Stanford University, Stanford, CA94305
| |
Collapse
|
24
|
Dhanasekaran R, Hansen AS, Park J, Lemaitre L, Lai I, Adeniji N, Kuruvilla S, Suresh A, Zhang J, Swamy V, Felsher DW. MYC Overexpression Drives Immune Evasion in Hepatocellular Carcinoma That Is Reversible through Restoration of Proinflammatory Macrophages. Cancer Res 2023; 83:626-640. [PMID: 36525476 PMCID: PMC9931653 DOI: 10.1158/0008-5472.can-22-0232] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 10/12/2022] [Accepted: 12/13/2022] [Indexed: 12/23/2022]
Abstract
Cancers evade immune surveillance, which can be reversed through immune-checkpoint therapy in a small subset of cases. Here, we report that the MYC oncogene suppresses innate immune surveillance and drives resistance to immunotherapy. In 33 different human cancers, MYC genomic amplification and overexpression increased immune-checkpoint expression, predicted nonresponsiveness to immune-checkpoint blockade, and was associated with both Th2-like immune profile and reduced CD8 T-cell infiltration. MYC transcriptionally suppressed innate immunity and MHCI-mediated antigen presentation, which in turn impeded T-cell response. Combined, but not individual, blockade of PDL1 and CTLA4 could reverse MYC-driven immune suppression by leading to the recruitment of proinflammatory antigen-presenting macrophages with increased CD40 and MHCII expression. Depletion of macrophages abrogated the antineoplastic effects of PDL1 and CTLA4 blockade in MYC-driven hepatocellular carcinoma (HCC). Hence, MYC is a predictor of immune-checkpoint responsiveness and a key driver of immune evasion through the suppression of proinflammatory macrophages. The immune evasion induced by MYC in HCC can be overcome by combined PDL1 and CTLA4 blockade. SIGNIFICANCE Macrophage-mediated immune evasion is a therapeutic vulnerability of MYC-driven cancers, which has implications for prioritizing MYC-driven hepatocellular carcinoma for combination immunotherapy.
Collapse
Affiliation(s)
- Renumathy Dhanasekaran
- Division of Gastroenterology and Hepatology, Department of Medicine. Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Aida S. Hansen
- Division of Oncology, Departments of Medicine and Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
- Department of Biomedicine, Aarhus University, Aarhus C 8000, Denmark
| | - Jangho Park
- Division of Gastroenterology and Hepatology, Department of Medicine. Stanford University School of Medicine, Stanford, CA 94305, USA
- Division of Oncology, Departments of Medicine and Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Lea Lemaitre
- Division of Gastroenterology and Hepatology, Department of Medicine. Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Ian Lai
- Division of Oncology, Departments of Medicine and Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Nia Adeniji
- Division of Gastroenterology and Hepatology, Department of Medicine. Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Sibu Kuruvilla
- Division of Oncology, Departments of Medicine and Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Akanksha Suresh
- Division of Gastroenterology and Hepatology, Department of Medicine. Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Josephine Zhang
- Division of Gastroenterology and Hepatology, Department of Medicine. Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Varsha Swamy
- Division of Oncology, Departments of Medicine and Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Dean W. Felsher
- Division of Oncology, Departments of Medicine and Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
| |
Collapse
|
25
|
Butyrate limits human natural killer cell effector function. Sci Rep 2023; 13:2715. [PMID: 36792800 PMCID: PMC9932090 DOI: 10.1038/s41598-023-29731-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 02/09/2023] [Indexed: 02/17/2023] Open
Abstract
The gut microbiota regulates chronic inflammation and has been implicated in the pathogenesis of a broad spectrum of disease including autoimmunity and cancer. Microbial short-chain fatty acids (SCFAs) e.g., butyrate have demonstrated immunomodulatory effects and are thought to be key mediators of the host-microbiome interaction. Here, we investigated the effect of butyrate on effector functions of blood derived human NK cells stimulated for 18 h with a combination of IL-12/IL-15, a potent mix of cytokines that drive NK cell activation. We show that butyrate has a strong anti-inflammatory effect on NK cells. NK cells cultured in the presence of butyrate expressed lower levels of activating receptors (TRAIL, NKp30, NKp44) and produced lower levels of cytokines (IFNγ, TNF-α, IL-22, granzyme B, granzyme A, perforin) in response to IL-12/IL-15. Butyrate restricted NK cell function by downregulation of mTORC1 activity, c-Myc mRNA expression and metabolism. Using a shotgun proteomic approach, we confirmed the effect of butyrate on NK cell cytokine signaling and metabolism and identified BRD2, MAT2A and EHD1 as downstream mediators of these effects. This insight into the immunomodulatory activity of butyrate on human NK cell function might help to develop new ways to limit NK cell function during chronic inflammation.
Collapse
|
26
|
Zhan M, Guo Y, Shen M, Shi X. Nanomaterial‐Boosted Tumor Immunotherapy Through Natural Killer Cells. ADVANCED NANOBIOMED RESEARCH 2022; 2. [DOI: 10.1002/anbr.202200096] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2025] Open
Abstract
Natural killer (NK)‐cell immunotherapy as an alternative to T‐cell immunotherapy has been widely used in clinical cell immunotherapy of various tumors. Despite the surprising findings, the widespread applications of NK cells are still limited by the insufficient expansion and short lifespan of adoptive NK cells in vivo, the poor penetration of NK cells in solid tumors, as well as the immunosuppressive tumor microenvironment that may cause the inactivation of NK cells. Fortunately, the emergence of nanomaterials provides many opportunities to address these vexing problems, thus overcoming the barriers faced by NK cells and promoting the tumor inhibitory efficacy of NK cells. Herein, the recent advances in the rational design of nanomaterials for boosting the NK cell‐based immunotherapy, mainly through enhancing NK cell engagement with tumors, boosting NK cell activation or expansion, as well as redirecting NK cells to tumor cells, are reviewed. Lastly, the design and preparation of next‐generation nanomaterials that aim to further boost the NK cell‐based immunotherapy are briefly discussed.
Collapse
Affiliation(s)
- Mengsi Zhan
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine College of Biological Science and Medical Engineering Donghua University Shanghai 201620 P.R. China
| | - Yunqi Guo
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine College of Biological Science and Medical Engineering Donghua University Shanghai 201620 P.R. China
| | - Mingwu Shen
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine College of Biological Science and Medical Engineering Donghua University Shanghai 201620 P.R. China
| | - Xiangyang Shi
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine College of Biological Science and Medical Engineering Donghua University Shanghai 201620 P.R. China
| |
Collapse
|
27
|
Sullivan DK, Deutzmann A, Yarbrough J, Krishnan MS, Gouw AM, Bellovin DI, Adam SJ, Liefwalker DF, Dhanasekaran R, Felsher DW. MYC oncogene elicits tumorigenesis associated with embryonic, ribosomal biogenesis, and tissue-lineage dedifferentiation gene expression changes. Oncogene 2022; 41:4960-4970. [PMID: 36207533 PMCID: PMC10257951 DOI: 10.1038/s41388-022-02458-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2021] [Revised: 08/23/2022] [Accepted: 08/26/2022] [Indexed: 11/09/2022]
Abstract
MYC is a transcription factor frequently overexpressed in cancer. To determine how MYC drives the neoplastic phenotype, we performed transcriptomic analysis using a panel of MYC-driven autochthonous transgenic mouse models. We found that MYC elicited gene expression changes mostly in a tissue- and lineage-specific manner across B-cell lymphoma, T-cell acute lymphoblastic lymphoma, hepatocellular carcinoma, renal cell carcinoma, and lung adenocarcinoma. However, despite these gene expression changes being mostly tissue-specific, we uncovered a convergence on a common pattern of upregulation of embryonic stem cell gene programs and downregulation of tissue-of-origin gene programs across MYC-driven cancers. These changes are representative of lineage dedifferentiation, that may be facilitated by epigenetic alterations that occur during tumorigenesis. Moreover, while several cellular processes are represented among embryonic stem cell genes, ribosome biogenesis is most specifically associated with MYC expression in human primary cancers. Altogether, MYC's capability to drive tumorigenesis in diverse tissue types appears to be related to its ability to both drive a core signature of embryonic genes that includes ribosomal biogenesis genes as well as promote tissue and lineage specific dedifferentiation.
Collapse
Affiliation(s)
- Delaney K Sullivan
- Division of Oncology, Departments of Medicine and Pathology, Stanford University School of Medicine, Stanford, CA, 94305, USA
- UCLA-Caltech Medical Scientist Training Program, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Anja Deutzmann
- Division of Oncology, Departments of Medicine and Pathology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Josiah Yarbrough
- Division of Oncology, Departments of Medicine and Pathology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Maya S Krishnan
- Division of Oncology, Departments of Medicine and Pathology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Arvin M Gouw
- Division of Oncology, Departments of Medicine and Pathology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - David I Bellovin
- Division of Oncology, Departments of Medicine and Pathology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Stacey J Adam
- Division of Oncology, Departments of Medicine and Pathology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Daniel F Liefwalker
- Department of Molecular and Medical Genetics, School of Medicine, Oregon Health and Science University, Portland, OR, 97239, USA
| | - Renumathy Dhanasekaran
- Division of Oncology, Departments of Medicine and Pathology, Stanford University School of Medicine, Stanford, CA, 94305, USA
- Division of Gastroenterology and Hepatology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Dean W Felsher
- Division of Oncology, Departments of Medicine and Pathology, Stanford University School of Medicine, Stanford, CA, 94305, USA.
| |
Collapse
|
28
|
Hao L, Chen Q, Chen X, Zhou Q. Integrated analysis of bulk and single-cell RNA-seq reveals the role of MYC signaling in lung adenocarcinoma. Front Genet 2022; 13:1021978. [PMID: 36299592 PMCID: PMC9589149 DOI: 10.3389/fgene.2022.1021978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 09/26/2022] [Indexed: 11/22/2022] Open
Abstract
MYC is one of the well-known oncogenes, and its important role in cancer still remains largely unknown. We obtained lung adenocarcinoma (LUAD) multi-omics data including genome, transcriptome, and single-cell sequencing data from multiple cohorts. We calculated the GSVA score of the MYC target v1 using the ssGSEA method, and obtained the genes highly correlated with this score by Spearman correlation analysis. Subsequent hierarchical clustering divided these genes into two gene sets highly associated with MYC signaling (S1 and S2). Unsupervised clustering based on these genes divided the LUAD samples into two distinct subgroups, namely, the MYC signaling inhibition group (C1) and activation group (C2). The MCP counter package in R was used to assess tumor immune cell infiltration abundance and ssGSEA was used to calculate gene set scores. The scRNA-seq was used to verify the association of MYC signaling to cell differentiation. We observed significant differences in prognosis, clinical characteristics, immune microenvironment, and genomic alterations between MYC signaling inhibition and MYC signaling activation groups. MYC-signaling is associated with genomic instability and can mediate the immunosuppressive microenvironment and promote cell proliferation, tumor stemness. Moreover, MYC-signaling activation is also subject to complex post-transcriptional regulation and is highly associated with cell differentiation. In conclusion, MYC signaling is closely related to the genomic instability, genetic alteration and regulation, the immune microenvironment landscape, cell differentiation, and disease survival in LUAD. The findings of this study provide a valuable reference to revealing the mechanism of cancer-promoting action of MYC in LUAD.
Collapse
Affiliation(s)
- Lu Hao
- Science and Education Department, Shenzhen Baoan Shiyan People’s Hospital, Shenzhen, China
| | - Qiuyan Chen
- Science and Education Department, Shenzhen Baoan Shiyan People’s Hospital, Shenzhen, China
| | - Xi Chen
- Central Laboratory, The People’s Hospital of Baoan Shenzhen, The Second Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Qing Zhou
- Central Laboratory, The People’s Hospital of Baoan Shenzhen, The Second Affiliated Hospital of Shenzhen University, Shenzhen, China
- *Correspondence: Qing Zhou,
| |
Collapse
|
29
|
Wang J, Liu X, Jin T, Cao Y, Tian Y, Xu F. NK cell immunometabolism as target for liver cancer therapy. Int Immunopharmacol 2022; 112:109193. [PMID: 36087507 DOI: 10.1016/j.intimp.2022.109193] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 08/04/2022] [Accepted: 08/22/2022] [Indexed: 11/29/2022]
Abstract
Natural killer (NK) cells are being used effectively as a potential candidate in tumor immunotherapy. However, the migration and transport of NK cells to solid tumors is inadequate. NK cell dysfunction, tumor invasiveness, and metastasis are associated with altered metabolism of NK cells in the liver cancer microenvironment. However, in liver cancers, metabolic impairment of NK cells is still not understood fully. Evidence from various sources has shown that the interaction of NK cell's immune checkpoints with its metabolic checkpoints is responsible for the regulation of the development and function of these cells. How immune checkpoints contribute to metabolic programming is still not fully understood, and how this can be beneficial needs a better understanding, but they are emerging to be incredibly compelling to rebuilding the function of NK cells in the tumor. It is expected to represent a potential aim that focuses on improving the efficacy of therapies based on NK cells for treating liver cancer. Here, the recent advancements made to understand the NK cell's metabolic reprogramming in liver cancer have been summarized, along with the possible interplay between the immune and the metabolic checkpoints in NK cell function. Finally, an overview of some potential metabolic-related targets that can be used for liver cancer therapy treatment has been presented.
Collapse
Affiliation(s)
- Junqi Wang
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Xiaolin Liu
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital of Jiaxing University, Jiaxing 314000, Zhejiang, China
| | - Tianqiang Jin
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Yuqing Cao
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Yu Tian
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Feng Xu
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang 110004, China.
| |
Collapse
|
30
|
Natural Killer Cell-Mediated Immunotherapy for Leukemia. Cancers (Basel) 2022; 14:cancers14030843. [PMID: 35159109 PMCID: PMC8833963 DOI: 10.3390/cancers14030843] [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: 12/30/2021] [Revised: 01/26/2022] [Accepted: 02/03/2022] [Indexed: 12/18/2022] Open
Abstract
Simple Summary Conventional therapies such as chemotherapy and radiation in leukemia increase infection susceptibility, adverse side effects and immune cell inactivation. Natural killer (NK) cells are the first line of defense against cancer and are critical in the recognition and cytolysis of rapidly dividing and abnormal cell populations. In this review, we describe NK cells and NK cell receptors, functional impairment of NK cells in leukemia, NK cell immunotherapies currently under investigation including monoclonal antibodies (mAbs), adoptive transfer, chimeric antigen receptor-NKs (CAR-NKs), bi-specific/tri-specific killer engagers (BiKEs/TriKEs) and potential targets of NK cell-mediated immunotherapy for leukemia in the future. Abstract Leukemia is a malignancy of the bone marrow and blood resulting from the abnormal differentiation of hematopoietic stem cells (HSCs). There are four main types of leukemia including acute myeloid leukemia (AML), acute lymphoblastic leukemia (ALL), chronic myeloid leukemia (CML), and chronic lymphocytic leukemia (CLL). While chemotherapy and radiation have been conventional forms of treatment for leukemia, these therapies increase infection susceptibility, adverse side effects and immune cell inactivation. Immunotherapies are becoming promising treatment options for leukemia, with natural killer (NK) cell-mediated therapy providing a specific direction of interest. The role of NK cells is critical for cancer cell elimination as these immune cells are the first line of defense against cancer proliferation and are involved in both recognition and cytolysis of rapidly dividing and abnormal cell populations. NK cells possess various activating and inhibitory receptors, which regulate NK cell function, signaling either inhibition and continued surveillance, or activation and subsequent cytotoxic activity. In this review, we describe NK cells and NK cell receptors, functional impairment of NK cells in leukemia, NK cell immunotherapies currently under investigation, including monoclonal antibodies (mAbs), adoptive transfer, chimeric antigen receptor-NKs (CAR-NKs), bi-specific/tri-specific killer engagers (BiKEs/TriKEs) and future potential targets of NK cell-based immunotherapy for leukemia.
Collapse
|
31
|
The MYC oncogene - the grand orchestrator of cancer growth and immune evasion. Nat Rev Clin Oncol 2022; 19:23-36. [PMID: 34508258 PMCID: PMC9083341 DOI: 10.1038/s41571-021-00549-2] [Citation(s) in RCA: 379] [Impact Index Per Article: 126.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/28/2021] [Indexed: 02/08/2023]
Abstract
The MYC proto-oncogenes encode a family of transcription factors that are among the most commonly activated oncoproteins in human neoplasias. Indeed, MYC aberrations or upregulation of MYC-related pathways by alternate mechanisms occur in the vast majority of cancers. MYC proteins are master regulators of cellular programmes. Thus, cancers with MYC activation elicit many of the hallmarks of cancer required for autonomous neoplastic growth. In preclinical models, MYC inactivation can result in sustained tumour regression, a phenomenon that has been attributed to oncogene addiction. Many therapeutic agents that directly target MYC are under development; however, to date, their clinical efficacy remains to be demonstrated. In the past few years, studies have demonstrated that MYC signalling can enable tumour cells to dysregulate their microenvironment and evade the host immune response. Herein, we discuss how MYC pathways not only dictate cancer cell pathophysiology but also suppress the host immune response against that cancer. We also propose that therapies targeting the MYC pathway will be key to reversing cancerous growth and restoring antitumour immune responses in patients with MYC-driven cancers.
Collapse
|
32
|
Ruan Y, Kim HN, Ogana HA, Wan Z, Hurwitz S, Nichols C, Abdel-Azim N, Coba A, Seo S, Loh YHE, Gang EJ, Abdel-Azim H, Hsieh CL, Lieber MR, Parekh C, Pal D, Bhojwani D, Durden DL, Kim YM. Preclinical Evaluation of a Novel Dual Targeting PI3Kδ/BRD4 Inhibitor, SF2535, in B-Cell Acute Lymphoblastic Leukemia. Front Oncol 2021; 11:766888. [PMID: 34926269 PMCID: PMC8671162 DOI: 10.3389/fonc.2021.766888] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 11/11/2021] [Indexed: 12/20/2022] Open
Abstract
The PI3K/Akt pathway—and in particular PI3Kδ—is known for its role in drug resistant B-cell acute lymphoblastic leukemia (B-ALL) and it is often upregulated in refractory or relapsed B-ALL. Myc proteins are transcription factors responsible for transcribing pro-proliferative genes and c-Myc is often overexpressed in cancers. The chromatin regulator BRD4 is required for expression of c-Myc in hematologic malignancies including B-ALL. Previously, combination of BRD4 and PI3K inhibition with SF2523 was shown to successfully decrease Myc expression. However, the underlying mechanism and effect of dual inhibition of PI3Kδ/BRD4 in B-ALL remains unknown. To study this, we utilized SF2535, a novel small molecule dual inhibitor which can specifically target the PI3Kδ isoform and BRD4. We treated primary B-ALL cells with various concentrations of SF2535 and studied its effect on specific pharmacological on-target mechanisms such as apoptosis, cell cycle, cell proliferation, and adhesion molecules expression usingin vitro and in vivo models. SF2535 significantly downregulates both c-Myc mRNA and protein expression through inhibition of BRD4 at the c-Myc promoter site and decreases p-AKT expression through inhibition of the PI3Kδ/AKT pathway. SF2535 induced apoptosis in B-ALL by downregulation of BCL-2 and increased cleavage of caspase-3, caspase-7, and PARP. Moreover, SF2535 induced cell cycle arrest and decreased cell counts in B-ALL. Interestingly, SF2535 decreased the mean fluorescence intensity (MFI) of integrin α4, α5, α6, and β1 while increasing MFI of CXCR4, indicating that SF2535 may work through inside-out signaling of integrins. Taken together, our data provide a rationale for the clinical evaluation of targeting PI3Kδ/BRD4 in refractory or relapsed B-ALL using SF2535.
Collapse
Affiliation(s)
- Yongsheng Ruan
- Department of Pediatrics, Division of Hematology, Oncology, Blood and Marrow Transplantation, Children’s Hospital Los Angeles, Norris Comprehensive Cancer Center, University of Southern California Keck School of Medicine, Los Angeles, CA, United States
- Department of Pediatrics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Hye Na Kim
- Department of Pediatrics, Division of Hematology, Oncology, Blood and Marrow Transplantation, Children’s Hospital Los Angeles, Norris Comprehensive Cancer Center, University of Southern California Keck School of Medicine, Los Angeles, CA, United States
| | - Heather A. Ogana
- Department of Pediatrics, Division of Hematology, Oncology, Blood and Marrow Transplantation, Children’s Hospital Los Angeles, Norris Comprehensive Cancer Center, University of Southern California Keck School of Medicine, Los Angeles, CA, United States
| | - Zesheng Wan
- Department of Pediatrics, Division of Hematology, Oncology, Blood and Marrow Transplantation, Children’s Hospital Los Angeles, Norris Comprehensive Cancer Center, University of Southern California Keck School of Medicine, Los Angeles, CA, United States
| | - Samantha Hurwitz
- Department of Pediatrics, Division of Hematology, Oncology, Blood and Marrow Transplantation, Children’s Hospital Los Angeles, Norris Comprehensive Cancer Center, University of Southern California Keck School of Medicine, Los Angeles, CA, United States
| | - Cydney Nichols
- Department of Pediatrics, Division of Hematology, Oncology, Blood and Marrow Transplantation, Children’s Hospital Los Angeles, Norris Comprehensive Cancer Center, University of Southern California Keck School of Medicine, Los Angeles, CA, United States
| | - Nour Abdel-Azim
- Department of Pediatrics, Division of Hematology, Oncology, Blood and Marrow Transplantation, Children’s Hospital Los Angeles, Norris Comprehensive Cancer Center, University of Southern California Keck School of Medicine, Los Angeles, CA, United States
| | - Ariana Coba
- Department of Pediatrics, Division of Hematology, Oncology, Blood and Marrow Transplantation, Children’s Hospital Los Angeles, Norris Comprehensive Cancer Center, University of Southern California Keck School of Medicine, Los Angeles, CA, United States
| | - Seyoung Seo
- Department of Pediatrics, Division of Hematology, Oncology, Blood and Marrow Transplantation, Children’s Hospital Los Angeles, Norris Comprehensive Cancer Center, University of Southern California Keck School of Medicine, Los Angeles, CA, United States
| | - Yong-Hwee Eddie Loh
- University of Southern California (USC) Libraries Bioinformatics Services, University of Southern California, Los Angeles, CA, United States
| | - Eun Ji Gang
- Department of Pediatrics, Division of Hematology, Oncology, Blood and Marrow Transplantation, Children’s Hospital Los Angeles, Norris Comprehensive Cancer Center, University of Southern California Keck School of Medicine, Los Angeles, CA, United States
| | - Hisham Abdel-Azim
- Department of Pediatrics, Division of Hematology, Oncology, Blood and Marrow Transplantation, Children’s Hospital Los Angeles, Norris Comprehensive Cancer Center, University of Southern California Keck School of Medicine, Los Angeles, CA, United States
| | - Chih-Lin Hsieh
- University of Southern California (USC) Department of Urology, University of Southern California (USC) Norris Comprehensive Cancer Center, Los Angeles, CA, United States
| | - Michael R. Lieber
- University of Southern California (USC) Department of Pathology, University of Southern California (USC) Norris Comprehensive Cancer Center, Los Angeles, CA, United States
| | - Chintan Parekh
- Department of Pediatrics, Division of Hematology, Oncology, Blood and Marrow Transplantation, Children’s Hospital Los Angeles, Norris Comprehensive Cancer Center, University of Southern California Keck School of Medicine, Los Angeles, CA, United States
| | - Dhananjaya Pal
- Department of Pediatrics, University of California San Diego, San Diego, CA, United States
| | - Deepa Bhojwani
- Department of Pediatrics, Division of Hematology, Oncology, Blood and Marrow Transplantation, Children’s Hospital Los Angeles, Norris Comprehensive Cancer Center, University of Southern California Keck School of Medicine, Los Angeles, CA, United States
| | - Donald L. Durden
- Department of Pediatrics, University of California San Diego, San Diego, CA, United States
- SignalRx Pharmaceuticals Inc., Omaha, NE, United States
| | - Yong-Mi Kim
- Department of Pediatrics, Division of Hematology, Oncology, Blood and Marrow Transplantation, Children’s Hospital Los Angeles, Norris Comprehensive Cancer Center, University of Southern California Keck School of Medicine, Los Angeles, CA, United States
- *Correspondence: Yong-Mi Kim,
| |
Collapse
|
33
|
Chen B, Deng Y, Wang B, Tian Z, Tong J, Yu B, Shi W, Tang J. Integrated analysis of long non-coding RNA-microRNA-mRNA competing endogenous RNAregulatory networks in thromboangiitis obliterans. Bioengineered 2021; 12:12023-12037. [PMID: 34787068 PMCID: PMC8810094 DOI: 10.1080/21655979.2021.2002497] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Thromboangiitis obliterans (TAO) is a non-atherosclerotic, segmental, chronic vascular inflammatory disease. Our aim was to explore the underlying mechanisms of long non-coding RNA (lncRNA)-related competing endogenous RNAs (ceRNAs) in TAO. Six blood samples were collected from patients with TAO and healthy individuals (three for each category). Total RNA was extracted from the blood of each participant and sequenced. Differentially expressed lncRNAs (DE-lncRNAs) and miRNAs (DE-miRNAs) were screened, and ceRNA networks associated with TAO were constructed. Thereafter, the genes in the ceRNA network were subjected to functional analyses. Finally, a ceRNA relationship (lncRNA NEAT1-hsa-miR-1-3p-mRNA GNA12) was selected for further validation. Analysis revealed that 347 DE-lncRNAs (150 downregulated and 197 upregulated) and 16 DE-miRNAs (3 downregulated and 13 upregulated) were identified in TAO. Further, TAO-associated ceRNA networks, which included 219 lncRNAs, 6 miRNAs, and 53 mRNAs, were proposed and subjected to gene annotation and pathway analysis. Additionally, NEAT1 and GNA12 levels were significantly upregulated, while miR-1-3p levels were evidently downregulated in TAO patients, as compared with those in healthy controls. Dual luciferase reporter assays showed that NEAT1, miR-1-3p, and GNA12 interacted with each other. We report potential TAO-associated ceRNA regulatory networks and suggest activation of NEAT1/miR-1-3p/GNA12 signaling as a novel mechanism for TAO progression.
Collapse
Affiliation(s)
- Bo Chen
- Department of Vascular Surgery, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China.,Shanghai Key Laboratory of Vascular Lesions Regulation and Remodeling, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
| | - Ying Deng
- Department of Vascular Surgery, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China.,Shanghai Key Laboratory of Vascular Lesions Regulation and Remodeling, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
| | - Bo Wang
- Department of Vascular Surgery, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China.,Shanghai Key Laboratory of Vascular Lesions Regulation and Remodeling, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
| | - Zhongyi Tian
- Department of Vascular Surgery, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China.,Shanghai Key Laboratory of Vascular Lesions Regulation and Remodeling, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
| | - Jindong Tong
- Department of Vascular Surgery, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China.,Shanghai Key Laboratory of Vascular Lesions Regulation and Remodeling, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
| | - Bo Yu
- Department of Vascular Surgery, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China.,Shanghai Key Laboratory of Vascular Lesions Regulation and Remodeling, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
| | - Weijun Shi
- Department of Vascular Surgery, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China.,Shanghai Key Laboratory of Vascular Lesions Regulation and Remodeling, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
| | - Jingdong Tang
- Department of Vascular Surgery, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China.,Shanghai Key Laboratory of Vascular Lesions Regulation and Remodeling, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
| |
Collapse
|
34
|
Chen M, Linstra R, van Vugt MATM. Genomic instability, inflammatory signaling and response to cancer immunotherapy. Biochim Biophys Acta Rev Cancer 2021; 1877:188661. [PMID: 34800547 DOI: 10.1016/j.bbcan.2021.188661] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 11/02/2021] [Accepted: 11/13/2021] [Indexed: 02/08/2023]
Abstract
Genomic and chromosomal instability are hallmarks of cancer and shape the genomic composition of cancer cells, thereby determining their behavior and response to treatment. Various genetic and epigenetic alterations in cancer have been linked to genomic instability, including DNA repair defects, oncogene-induced replication stress, and spindle assembly checkpoint malfunction. A consequence of genomic and chromosomal instability is the leakage of DNA from the nucleus into the cytoplasm, either directly or through the formation and subsequent rupture of micronuclei. Cytoplasmic DNA subsequently activates cytoplasmic DNA sensors, triggering downstream pathways, including a type I interferon response. This inflammatory signaling has pleiotropic effects, including enhanced anti-tumor immunity and potentially results in sensitization of cancer cells to immune checkpoint inhibitors. However, cancers frequently evolve mechanisms to avoid immune clearance, including suppression of inflammatory signaling. In this review, we summarize inflammatory signaling pathways induced by various sources of genomic instability, adaptation mechanisms that suppress inflammatory signaling, and implications for cancer immunotherapy.
Collapse
Affiliation(s)
- Mengting Chen
- Department of Medical Oncology, Cancer Research Center Groningen, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713GZ, Groningen, the Netherlands
| | - Renske Linstra
- Department of Medical Oncology, Cancer Research Center Groningen, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713GZ, Groningen, the Netherlands
| | - Marcel A T M van Vugt
- Department of Medical Oncology, Cancer Research Center Groningen, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713GZ, Groningen, the Netherlands.
| |
Collapse
|
35
|
Duault C, Kumar A, Taghi Khani A, Lee SJ, Yang L, Huang M, Hurtz C, Manning B, Ghoda L, McDonald T, Lacayo NJ, Sakamoto KM, Carroll M, Tasian SK, Marcucci G, Yu J, Caligiuri MA, Maecker HT, Swaminathan S. Activated natural killer cells predict poor clinical prognosis in high-risk B- and T-cell acute lymphoblastic leukemia. Blood 2021; 138:1465-1480. [PMID: 34077953 PMCID: PMC8532198 DOI: 10.1182/blood.2020009871] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2020] [Accepted: 05/05/2021] [Indexed: 11/21/2022] Open
Abstract
B- and T-cell acute lymphoblastic leukemia (B/T-ALL) may be refractory or recur after therapy by suppressing host anticancer immune surveillance mediated specifically by natural killer (NK) cells. We delineated the phenotypic and functional defects in NK cells from high-risk patients with B/T-ALL using mass cytometry, flow cytometry, and in silico cytometry, with the goal of further elucidating the role of NK cells in sustaining acute lymphoblastic leukemia (ALL) regression. We found that, compared with their normal counterparts, NK cells from patients with B/T-ALL are less cytotoxic but exhibit an activated signature that is characterized by high CD56, high CD69, production of activated NK cell-origin cytokines, and calcium (Ca2+) signaling. We demonstrated that defective maturation of NK cells into cytotoxic effectors prevents NK cells from ALL from lysing NK cell-sensitive targets as efficiently as do normal NK cells. Additionally, we showed that NK cells in ALL are exhausted, which is likely caused by their chronic activation. We found that increased frequencies of activated cytokine-producing NK cells are associated with increased disease severity and independently predict poor clinical outcome in patients with ALL. Our studies highlight the benefits of developing NK cell profiling as a diagnostic tool to predict clinical outcome in patients with ALL and underscore the clinical potential of allogeneic NK cell infusions to prevent ALL recurrence.
Collapse
Affiliation(s)
- Caroline Duault
- The Human Immune Monitoring Center, Institute for Immunity, Transplantation, and Infection, Stanford University School of Medicine, Stanford, CA
| | - Anil Kumar
- Department of Systems Biology, Beckman Research Institute of City of Hope, Monrovia, CA
| | - Adeleh Taghi Khani
- Department of Systems Biology, Beckman Research Institute of City of Hope, Monrovia, CA
| | - Sung June Lee
- Department of Systems Biology, Beckman Research Institute of City of Hope, Monrovia, CA
| | - Lu Yang
- Department of Systems Biology, Beckman Research Institute of City of Hope, Monrovia, CA
| | - Min Huang
- Division of Hematology/Oncology, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA
| | - Christian Hurtz
- Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Bryan Manning
- Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Lucy Ghoda
- Department of Hematological Malignancies and Translational Science, Beckman Research Institute of City of Hope, Duarte, CA
| | - Tinisha McDonald
- Department of Hematological Malignancies and Translational Science, Beckman Research Institute of City of Hope, Duarte, CA
| | - Norman J Lacayo
- Division of Hematology/Oncology, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA
| | - Kathleen M Sakamoto
- Division of Hematology/Oncology, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA
| | - Martin Carroll
- Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Sarah K Tasian
- Division of Oncology and Center for Childhood Cancer Research, Department of Pediatrics, Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA; and
| | - Guido Marcucci
- Department of Hematological Malignancies and Translational Science, Beckman Research Institute of City of Hope, Duarte, CA
| | - Jianhua Yu
- Department of Hematology and Hematopoietic Stem Cell Transplantation, Beckman Research Institute of City of Hope, Duarte, CA
| | - Michael A Caligiuri
- Department of Hematology and Hematopoietic Stem Cell Transplantation, Beckman Research Institute of City of Hope, Duarte, CA
| | - Holden T Maecker
- The Human Immune Monitoring Center, Institute for Immunity, Transplantation, and Infection, Stanford University School of Medicine, Stanford, CA
| | - Srividya Swaminathan
- Department of Systems Biology, Beckman Research Institute of City of Hope, Monrovia, CA
- Department of Hematological Malignancies and Translational Science, Beckman Research Institute of City of Hope, Duarte, CA
| |
Collapse
|
36
|
Fink A, Hung E, Singh I, Ben-Neriah Y. Immunity in acute myeloid leukemia: Where the immune response and targeted therapy meet. Eur J Immunol 2021; 52:34-43. [PMID: 34648664 DOI: 10.1002/eji.202048945] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Revised: 07/29/2021] [Accepted: 10/05/2021] [Indexed: 12/19/2022]
Abstract
Acute myeloid leukemia (AML) is a highly aggressive disease with high relapse and mortality rates. Recent years have shown a surge in novel therapeutic development for AML, both in clinical and preclinical stages. These developments include targeted therapies based on AML-specific molecular signatures as well as more general immune modulation and vaccination studies. In this review, we will explore the evolving arena of AML therapy and suggest some intriguing connections between immune system modulation and targeted therapy. Improved understanding of the immune system involvement in various stages of the disease and the crosstalk between immune effectors, targeted therapy, and AML cells can provide a better framework for designing the next generation of AML therapies.
Collapse
Affiliation(s)
- Avner Fink
- The Lautenberg Center for Immunology and Cancer Research, Institute of Medical Research Israel-Canada, Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Eric Hung
- The Lautenberg Center for Immunology and Cancer Research, Institute of Medical Research Israel-Canada, Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Indranil Singh
- The Lautenberg Center for Immunology and Cancer Research, Institute of Medical Research Israel-Canada, Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Yinon Ben-Neriah
- The Lautenberg Center for Immunology and Cancer Research, Institute of Medical Research Israel-Canada, Hebrew University-Hadassah Medical School, Jerusalem, Israel
| |
Collapse
|
37
|
Zhang P, He B, Cai Q, Tu G, Peng X, Zhao Z, Peng W, Yu F, Wang M, Tao Y, Wang X. Decreased IL-6 and NK Cells in Early-Stage Lung Adenocarcinoma Presenting as Ground-Glass Opacity. Front Oncol 2021; 11:705888. [PMID: 34568032 PMCID: PMC8457009 DOI: 10.3389/fonc.2021.705888] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Accepted: 08/13/2021] [Indexed: 01/06/2023] Open
Abstract
Background Lung ground-glass opacities (GGOs) are an early manifestation of lung adenocarcinoma. It is of great value to study the changes in the immune microenvironment of GGO to elucidate the occurrence and evolution of early lung adenocarcinoma. Although the changes of IL-6 and NK cells in lung adenocarcinoma have caught global attention, we have little appreciation for how IL-6 and NK cells in the lung GGO affect the progression of early lung adenocarcinoma. Methods We analyzed the RNA sequencing data of surgical specimens from 21 patients with GGO-featured primary lung adenocarcinoma and verified the changes in the expression of IL-6 and other important immune molecules in the TCGA and GEO databases. Next, we used flow cytometry to detect the protein expression levels of important Th1/Th2 cytokines in GGO and normal lung tissues and the changes in the composition ratio of tumor infiltrating lymphocytes (TILs). Then, we analyzed the effect of IL-6 on NK cells through organoid culture and immunofluorescence. Finally, we explored the changes of related molecules and pathway might be involved. Results IL-6 may play an important role in the tumor microenvironment of early lung adenocarcinoma. Further research confirmed that the decrease of IL-6 in GGO tissue is consistent with the changes in NK cells, and there seems to be a correlation between these two phenomena. Conclusion The IL-6 expression status and NK cell levels of early lung adenocarcinoma as GGO are significantly reduced, and the stimulation of IL-6 can up-regulate or activate NK cells in GGO, providing new insights into the diagnosis and pathogenesis of early lung cancer.
Collapse
Affiliation(s)
- Pengfei Zhang
- Department of Thoracic Surgery, Second Xiangya Hospital, Central South University, Changsha, China.,Hunan Key Laboratory of Early Diagnosis and Precision Therapy, Second Xiangya Hospital, Central South University, Changsha, China
| | - Boxue He
- Department of Thoracic Surgery, Second Xiangya Hospital, Central South University, Changsha, China.,Hunan Key Laboratory of Early Diagnosis and Precision Therapy, Second Xiangya Hospital, Central South University, Changsha, China
| | - Qidong Cai
- Department of Thoracic Surgery, Second Xiangya Hospital, Central South University, Changsha, China.,Hunan Key Laboratory of Early Diagnosis and Precision Therapy, Second Xiangya Hospital, Central South University, Changsha, China
| | - Guangxu Tu
- Department of Thoracic Surgery, Second Xiangya Hospital, Central South University, Changsha, China.,Hunan Key Laboratory of Early Diagnosis and Precision Therapy, Second Xiangya Hospital, Central South University, Changsha, China
| | - Xiong Peng
- Department of Thoracic Surgery, Second Xiangya Hospital, Central South University, Changsha, China.,Hunan Key Laboratory of Early Diagnosis and Precision Therapy, Second Xiangya Hospital, Central South University, Changsha, China
| | - Zhenyu Zhao
- Department of Thoracic Surgery, Second Xiangya Hospital, Central South University, Changsha, China.,Hunan Key Laboratory of Early Diagnosis and Precision Therapy, Second Xiangya Hospital, Central South University, Changsha, China
| | - Weilin Peng
- Department of Thoracic Surgery, Second Xiangya Hospital, Central South University, Changsha, China.,Hunan Key Laboratory of Early Diagnosis and Precision Therapy, Second Xiangya Hospital, Central South University, Changsha, China
| | - Fenglei Yu
- Department of Thoracic Surgery, Second Xiangya Hospital, Central South University, Changsha, China.,Hunan Key Laboratory of Early Diagnosis and Precision Therapy, Second Xiangya Hospital, Central South University, Changsha, China
| | - Min Wang
- Department of Laboratory Medicine, Second Xiangya Hospital, Central South University, Changsha, China
| | - Yongguang Tao
- Department of Thoracic Surgery, Second Xiangya Hospital, Central South University, Changsha, China.,Hunan Key Laboratory of Early Diagnosis and Precision Therapy, Second Xiangya Hospital, Central South University, Changsha, China.,Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Department of Pathology, Xiangya Hospital, Central South University, Changsha, China.,National Health Commission of the People's Republic of China (NHC), Key Laboratory of Carcinogenesis (Central South University), Cancer Research Institute and School of Basic Medicine, Central South University, Changsha, China
| | - Xiang Wang
- Department of Thoracic Surgery, Second Xiangya Hospital, Central South University, Changsha, China.,Hunan Key Laboratory of Early Diagnosis and Precision Therapy, Second Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
38
|
Liefwalker DF, Ryan M, Wang Z, Pathak KV, Plaisier S, Shah V, Babra B, Dewson GS, Lai IK, Mosley AR, Fueger PT, Casey SC, Jiang L, Pirrotte P, Swaminathan S, Sears RC. Metabolic convergence on lipogenesis in RAS, BCR-ABL, and MYC-driven lymphoid malignancies. Cancer Metab 2021; 9:31. [PMID: 34399819 PMCID: PMC8369789 DOI: 10.1186/s40170-021-00263-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 06/23/2021] [Indexed: 11/20/2022] Open
Abstract
BACKGROUND Metabolic reprogramming is a central feature in many cancer subtypes and a hallmark of cancer. Many therapeutic strategies attempt to exploit this feature, often having unintended side effects on normal metabolic programs and limited efficacy due to integrative nature of metabolic substrate sourcing. Although the initiating oncogenic lesion may vary, tumor cells in lymphoid malignancies often share similar environments and potentially similar metabolic profiles. We examined cells from mouse models of MYC-, RAS-, and BCR-ABL-driven lymphoid malignancies and find a convergence on de novo lipogenesis. We explore the potential role of MYC in mediating lipogenesis by 13C glucose tracing and untargeted metabolic profiling. Inhibition of lipogenesis leads to cell death both in vitro and in vivo and does not induce cell death of normal splenocytes. METHODS We analyzed RNA-seq data sets for common metabolic convergence in lymphoma and leukemia. Using in vitro cell lines derived in from conditional MYC, RAS, and BCR-ABL transgenic murine models and oncogene-driven human cell lines, we determined gene regulation, metabolic profiles, and sensitivity to inhibition of lipogenesis in lymphoid malignancies. We utilize preclinical murine models and transgenic primary model of T-ALL to determine the effect of lipogenesis blockade across BCR-ABL-, RAS-, and c-MYC-driven lymphoid malignancies. Statistical significance was calculated using unpaired t-tests and one-way ANOVA. RESULTS This study illustrates that de novo lipid biogenesis is a shared feature of several lymphoma subtypes. Using cell lines derived from conditional MYC, RAS, and BCR-ABL transgenic murine models, we demonstrate shared responses to inhibition of lipogenesis by the acetyl-coA carboxylase inhibitor 5-(tetradecloxy)-2-furic acid (TOFA), and other lipogenesis inhibitors. We performed metabolic tracing studies to confirm the influence of c-MYC and TOFA on lipogenesis. We identify specific cell death responses to TOFA in vitro and in vivo and demonstrate delayed engraftment and progression in vivo in transplanted lymphoma cell lines. We also observe delayed progression of T-ALL in a primary transgenic mouse model upon TOFA administration. In a panel of human cell lines, we demonstrate sensitivity to TOFA treatment as a metabolic liability due to the general convergence on de novo lipogenesis in lymphoid malignancies driven by MYC, RAS, or BCR-ABL. Importantly, cell death was not significantly observed in non-malignant cells in vivo. CONCLUSIONS These studies suggest that de novo lipogenesis may be a common survival strategy for many lymphoid malignancies and may be a clinically exploitable metabolic liability. TRIAL REGISTRATION This study does not include any clinical interventions on human subjects.
Collapse
Affiliation(s)
- Daniel F Liefwalker
- Department of Molecular and Medical Genetics, Oregon Health and Science University, Portland, OR, 97201, USA.
- Knight Cancer Institute, Oregon Health and Science University, Portland, OR, 97201, USA.
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA.
| | - Meital Ryan
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Zhichao Wang
- Department of Molecular & Cellular Endocrinology, Diabetes and Metabolism Research Institute, City of Hope Medical Center, Duarte, CA, 91010, USA
| | - Khyatiben V Pathak
- Collaborative Center for Translational Mass Spectrometry, Translational Genomics Research Institute, 445 N 5th St, Phoenix, AZ, 85004, USA
| | - Seema Plaisier
- Collaborative Center for Translational Mass Spectrometry, Translational Genomics Research Institute, 445 N 5th St, Phoenix, AZ, 85004, USA
| | - Vidhi Shah
- Department of Molecular and Medical Genetics, Oregon Health and Science University, Portland, OR, 97201, USA
- Brenden-Colson Center for Pancreatic Care, Oregon Health and Science University, Portland, OR, 97201, USA
| | - Bobby Babra
- Molecular & Cellular Biology, Oregon State University, Corvallis, Oregon, 97331, USA
| | - Gabrielle S Dewson
- Department of Molecular and Medical Genetics, Oregon Health and Science University, Portland, OR, 97201, USA
- Brenden-Colson Center for Pancreatic Care, Oregon Health and Science University, Portland, OR, 97201, USA
| | - Ian K Lai
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Adriane R Mosley
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Patrick T Fueger
- Department of Molecular & Cellular Endocrinology, Diabetes and Metabolism Research Institute, City of Hope Medical Center, Duarte, CA, 91010, USA
- Comprehensive Cancer Center, Beckman Research Institute, City of Hope Medical Center, Duarte, CA, 91010, USA
| | - Stephanie C Casey
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Lei Jiang
- Department of Molecular & Cellular Endocrinology, Diabetes and Metabolism Research Institute, City of Hope Medical Center, Duarte, CA, 91010, USA
- Comprehensive Cancer Center, Beckman Research Institute, City of Hope Medical Center, Duarte, CA, 91010, USA
| | - Patrick Pirrotte
- Collaborative Center for Translational Mass Spectrometry, Translational Genomics Research Institute, 445 N 5th St, Phoenix, AZ, 85004, USA
| | - Srividya Swaminathan
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA
- Department of Systems Biology, Beckman Research Institute of the City of Hope, Monrovia, CA, 91016, USA
- Department of Hematological Malignancies, Beckman Research Institute of City of Hope, Duarte, CA, 91010, USA
| | - Rosalie C Sears
- Department of Molecular and Medical Genetics, Oregon Health and Science University, Portland, OR, 97201, USA
- Knight Cancer Institute, Oregon Health and Science University, Portland, OR, 97201, USA
- Brenden-Colson Center for Pancreatic Care, Oregon Health and Science University, Portland, OR, 97201, USA
| |
Collapse
|
39
|
Krenz B, Gebhardt-Wolf A, Ade CP, Gaballa A, Roehrig F, Vendelova E, Baluapuri A, Eilers U, Gallant P, D'Artista L, Wiegering A, Gasteiger G, Rosenfeldt MT, Bauer S, Zender L, Wolf E, Eilers M. MYC- and MIZ1-Dependent Vesicular Transport of Double-Strand RNA Controls Immune Evasion in Pancreatic Ductal Adenocarcinoma. Cancer Res 2021; 81:4242-4256. [PMID: 34145038 DOI: 10.1158/0008-5472.can-21-1677] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 06/08/2021] [Accepted: 06/17/2021] [Indexed: 11/16/2022]
Abstract
Deregulated expression of the MYC oncoprotein enables tumor cells to evade immune surveillance, but the mechanisms underlying this surveillance are poorly understood. We show here that endogenous MYC protects pancreatic ductal adenocarcinoma (PDAC) driven by KRASG12D and TP53R172H from eradication by the immune system. Deletion of TANK-binding kinase 1 (TBK1) bypassed the requirement for high MYC expression. TBK1 was active due to the accumulation of double-stranded RNA (dsRNA), which was derived from inverted repetitive elements localized in introns of nuclear genes. Nuclear-derived dsRNA is packaged into extracellular vesicles and subsequently recognized by toll-like receptor 3 (TLR3) to activate TBK1 and downstream MHC class I expression in an autocrine or paracrine manner before being degraded in lysosomes. MYC suppressed loading of dsRNA onto TLR3 and its subsequent degradation via association with MIZ1. Collectively, these findings suggest that MYC and MIZ1 suppress a surveillance pathway that signals perturbances in mRNA processing to the immune system, which facilitates immune evasion in PDAC. SIGNIFICANCE: This study identifies a TBK1-dependent pathway that links dsRNA metabolism to antitumor immunity and shows that suppression of TBK1 is a critical function of MYC in pancreatic ductal adenocarcinoma.
Collapse
Affiliation(s)
- Bastian Krenz
- Department of Biochemistry and Molecular Biology, Theodor Boveri Institute, Biocenter, University of Würzburg, Würzburg, Germany
| | - Anneli Gebhardt-Wolf
- Department of Biochemistry and Molecular Biology, Theodor Boveri Institute, Biocenter, University of Würzburg, Würzburg, Germany
| | - Carsten P Ade
- Department of Biochemistry and Molecular Biology, Theodor Boveri Institute, Biocenter, University of Würzburg, Würzburg, Germany
| | - Abdallah Gaballa
- Department of Biochemistry and Molecular Biology, Theodor Boveri Institute, Biocenter, University of Würzburg, Würzburg, Germany
| | - Florian Roehrig
- Comprehensive Cancer Center Mainfranken, Core Unit Bioinformatics, Biocenter, University of Würzburg, Würzburg, Germany
| | - Emilia Vendelova
- Institute of Systems Immunology, University of Würzburg, Würzburg, Germany
| | - Apoorva Baluapuri
- Department of Biochemistry and Molecular Biology, Cancer Systems Biology Group, Biocenter, University of Würzburg, Würzburg, Germany
| | - Ursula Eilers
- Core Unit High-Content Microscopy, Biocenter, University of Würzburg, Würzburg, Germany
| | - Peter Gallant
- Department of Biochemistry and Molecular Biology, Theodor Boveri Institute, Biocenter, University of Würzburg, Würzburg, Germany
| | - Luana D'Artista
- Department of Medical Oncology and Pneumology (Internal Medicine VIII), University Hospital Tübingen, Tübingen, Germany
| | - Armin Wiegering
- Department of Biochemistry and Molecular Biology, Theodor Boveri Institute, Biocenter, University of Würzburg, Würzburg, Germany.,Department of General, Visceral, Transplant, Vascular and Pediatric Surgery, University Hospital Würzburg, Würzburg, Germany
| | - Georg Gasteiger
- Institute of Systems Immunology, University of Würzburg, Würzburg, Germany
| | | | - Stefan Bauer
- Institute for Immunology, BMFZ, Philipps-University of Marburg, Marburg, Germany
| | - Lars Zender
- Department of Medical Oncology and Pneumology (Internal Medicine VIII), University Hospital Tübingen, Tübingen, Germany
| | - Elmar Wolf
- Department of Biochemistry and Molecular Biology, Cancer Systems Biology Group, Biocenter, University of Würzburg, Würzburg, Germany
| | - Martin Eilers
- Department of Biochemistry and Molecular Biology, Theodor Boveri Institute, Biocenter, University of Würzburg, Würzburg, Germany.
| |
Collapse
|
40
|
Hori SS, Tong L, Swaminathan S, Liebersbach M, Wang J, Gambhir SS, Felsher DW. A mathematical model of tumor regression and recurrence after therapeutic oncogene inactivation. Sci Rep 2021; 11:1341. [PMID: 33446671 PMCID: PMC7809285 DOI: 10.1038/s41598-020-78947-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 11/24/2020] [Indexed: 12/19/2022] Open
Abstract
The targeted inactivation of individual oncogenes can elicit regression of cancers through a phenomenon called oncogene addiction. Oncogene addiction is mediated by cell-autonomous and immune-dependent mechanisms. Therapeutic resistance to oncogene inactivation leads to recurrence but can be counteracted by immune surveillance. Predicting the timing of resistance will provide valuable insights in developing effective cancer treatments. To provide a quantitative understanding of cancer response to oncogene inactivation, we developed a new 3-compartment mathematical model of oncogene-driven tumor growth, regression and recurrence, and validated the model using a MYC-driven transgenic mouse model of T-cell acute lymphoblastic leukemia. Our mathematical model uses imaging-based measurements of tumor burden to predict the relative number of drug-sensitive and drug-resistant cancer cells in MYC-dependent states. We show natural killer (NK) cell adoptive therapy can delay cancer recurrence by reducing the net-growth rate of drug-resistant cells. Our studies provide a novel way to evaluate combination therapy for personalized cancer treatment.
Collapse
Affiliation(s)
- Sharon S Hori
- Department of Radiology, Stanford University School of Medicine, Stanford, CA, USA.
- Molecular Imaging Program at Stanford, Stanford University School of Medicine, Stanford, CA, USA.
- Canary Center at Stanford for Cancer Early Detection, Stanford University School of Medicine, Palo Alto, CA, USA.
| | - Ling Tong
- Molecular Imaging Program at Stanford, Stanford University School of Medicine, Stanford, CA, USA
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Srividya Swaminathan
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Department of Systems Biology, Beckman Research Institute of the City of Hope, Monrovia, CA, USA
| | - Mariola Liebersbach
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Jingjing Wang
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Department of Oncology, The Second Xiangya Hospital of Central South University, Changsha, People's Republic of China
| | - Sanjiv S Gambhir
- Department of Radiology, Stanford University School of Medicine, Stanford, CA, USA
- Molecular Imaging Program at Stanford, Stanford University School of Medicine, Stanford, CA, USA
- Canary Center at Stanford for Cancer Early Detection, Stanford University School of Medicine, Palo Alto, CA, USA
- Department of Bioengineering, Stanford University School of Medicine, Stanford, CA, USA
- Department of Materials Science and Engineering, Stanford University, Stanford, CA, USA
| | - Dean W Felsher
- Molecular Imaging Program at Stanford, Stanford University School of Medicine, Stanford, CA, USA.
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA.
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
41
|
Riggan L, Shah S, O’Sullivan TE. Arrested development: suppression of NK cell function in the tumor microenvironment. Clin Transl Immunology 2021; 10:e1238. [PMID: 33456775 PMCID: PMC7797224 DOI: 10.1002/cti2.1238] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 12/17/2020] [Accepted: 12/18/2020] [Indexed: 12/11/2022] Open
Abstract
Natural killer (NK) cells are cytotoxic innate lymphocytes that protect against viral infection and tumor metastasis. Despite their inherent ability to kill a broad range of virally infected, stressed and transformed cells, low numbers of dysfunctional NK cells are often observed in many advanced solid human cancers. Here, we review the potential mechanisms that influence suboptimal mature NK cell recruitment and function in the tumor microenvironment (TME) of solid tumors. We further highlight current immunotherapy approaches aimed to circumvent NK cell dysfunction and discuss next-generation strategies to enhance adoptive NK cell therapy through targeting intrinsic and extrinsic checkpoints the regulate NK cell functionality in the TME. Understanding the mechanisms that drive NK cell dysfunction in the TME will lead to novel immunotherapeutic approaches in the fight against cancer.
Collapse
Affiliation(s)
- Luke Riggan
- Department of Microbiology, Immunology, and Molecular GeneticsDavid Geffen School of Medicine at UCLALos AngelesCAUSA
- Molecular Biology InstituteUniversity of CaliforniaLos AngelesCAUSA
| | - Siya Shah
- Department of Microbiology, Immunology, and Molecular GeneticsDavid Geffen School of Medicine at UCLALos AngelesCAUSA
| | - Timothy E O’Sullivan
- Department of Microbiology, Immunology, and Molecular GeneticsDavid Geffen School of Medicine at UCLALos AngelesCAUSA
- Molecular Biology InstituteUniversity of CaliforniaLos AngelesCAUSA
| |
Collapse
|
42
|
Venkatraman S, Meller J, Hongeng S, Tohtong R, Chutipongtanate S. Transcriptional Regulation of Cancer Immune Checkpoints: Emerging Strategies for Immunotherapy. Vaccines (Basel) 2020; 8:E735. [PMID: 33291616 PMCID: PMC7761936 DOI: 10.3390/vaccines8040735] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 12/01/2020] [Accepted: 12/02/2020] [Indexed: 12/19/2022] Open
Abstract
The study of immune evasion has gained a well-deserved eminence in cancer research by successfully developing a new class of therapeutics, immune checkpoint inhibitors, such as pembrolizumab and nivolumab, anti-PD-1 antibodies. By aiming at the immune checkpoint blockade (ICB), these new therapeutics have advanced cancer treatment with notable increases in overall survival and tumor remission. However, recent reports reveal that 40-60% of patients fail to benefit from ICB therapy due to acquired resistance or tumor relapse. This resistance may stem from increased expression of co-inhibitory immune checkpoints or alterations in the tumor microenvironment that promotes immune suppression. Because these mechanisms are poorly elucidated, the transcription factors that regulate immune checkpoints, known as "master regulators", have garnered interest. These include AP-1, IRF-1, MYC, and STAT3, which are known to regulate PD/PD-L1 and CTLA-4. Identifying these and other potential master regulators as putative therapeutic targets or biomarkers can be facilitated by mining cancer literature, public datasets, and cancer genomics resources. In this review, we describe recent advances in master regulator identification and characterization of the mechanisms underlying immune checkpoints regulation, and discuss how these master regulators of immune checkpoint molecular expression can be targeted as a form of auxiliary therapeutic strategy to complement traditional immunotherapy.
Collapse
Affiliation(s)
- Simran Venkatraman
- Graduate Program in Molecular Medicine, Faculty of Science Joint Program Faculty of Medicine Ramathibodi Hospital, Faculty of Medicine Siriraj Hospital, Faculty of Dentistry, Faculty of Tropical Medicine, Mahidol University, Bangkok 10400, Thailand;
| | - Jarek Meller
- Departments of Environmental and Public Health Sciences, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA;
- Division of Biomedical Informatics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45267, USA
| | - Suradej Hongeng
- Division of Hematology and Oncology, Department of Pediatrics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand;
| | - Rutaiwan Tohtong
- Graduate Program in Molecular Medicine, Faculty of Science Joint Program Faculty of Medicine Ramathibodi Hospital, Faculty of Medicine Siriraj Hospital, Faculty of Dentistry, Faculty of Tropical Medicine, Mahidol University, Bangkok 10400, Thailand;
- Department of Biochemistry, Faculty of Science, Mahidol University, Bangkok 10400, Thailand
| | - Somchai Chutipongtanate
- Pediatric Translational Research Unit, Department of Pediatrics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand
- Department of Clinical Epidemiology and Biostatistics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand
| |
Collapse
|
43
|
de Jonge AV, Mutis T, Roemer MGM, Scheijen B, Chamuleau MED. Impact of MYC on Anti-Tumor Immune Responses in Aggressive B Cell Non-Hodgkin Lymphomas: Consequences for Cancer Immunotherapy. Cancers (Basel) 2020; 12:cancers12103052. [PMID: 33092116 PMCID: PMC7589056 DOI: 10.3390/cancers12103052] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 10/12/2020] [Accepted: 10/16/2020] [Indexed: 02/08/2023] Open
Abstract
Simple Summary The human immune system has several mechanisms to attack and eliminate lymphomas. However, the MYC oncogene is thought to facilitate escape from this anti-tumor immune response. Since patients with MYC overexpressing lymphomas face a significant dismal prognosis after treatment with standard immunochemotherapy, understanding the role of MYC in regulating the anti-tumor immune response is highly relevant. In this review, we describe the mechanisms by which MYC attenuates the anti-tumor immune responses in B cell non-Hodgkin lymphomas. We aim to implement this knowledge in the deployment of novel immunotherapeutic approaches. Therefore, we also provide a comprehensive overview of current immunotherapeutic options and we discuss potential future treatment strategies for MYC overexpressing lymphomas. Abstract Patients with MYC overexpressing high grade B cell lymphoma (HGBL) face significant dismal prognosis after treatment with standard immunochemotherapy regimens. Recent preclinical studies indicate that MYC not only contributes to tumorigenesis by its effects on cell proliferation and differentiation, but also plays an important role in promoting escape from anti-tumor immune responses. This is of specific interest, since reversing tumor immune inhibition with immunotherapy has shown promising results in the treatment of both solid tumors and hematological malignancies. In this review, we outline the current understanding of impaired immune responses in B cell lymphoid malignancies with MYC overexpression, with a particular emphasis on diffuse large B cell lymphoma. We also discuss clinical consequences of MYC overexpression in the treatment of HGBL with novel immunotherapeutic agents and potential future treatment strategies.
Collapse
Affiliation(s)
- A. Vera de Jonge
- Department of Hematology, Amsterdam UMC, VU University Medical Center, Cancer Center Amsterdam, 1081HV Amsterdam, The Netherlands; (T.M.); (M.E.D.C.)
- Correspondence:
| | - Tuna Mutis
- Department of Hematology, Amsterdam UMC, VU University Medical Center, Cancer Center Amsterdam, 1081HV Amsterdam, The Netherlands; (T.M.); (M.E.D.C.)
| | - Margaretha G. M. Roemer
- Department of Pathology, Amsterdam UMC, VU University Medical Center, Cancer Center Amsterdam, 1081HV Amsterdam, The Netherlands;
| | - Blanca Scheijen
- Department of Pathology, Radboud UMC, Radboud Institute for Molecular Life Sciences, 6525GA Nijmegen, The Netherlands;
| | - Martine E. D. Chamuleau
- Department of Hematology, Amsterdam UMC, VU University Medical Center, Cancer Center Amsterdam, 1081HV Amsterdam, The Netherlands; (T.M.); (M.E.D.C.)
| |
Collapse
|