1
|
Winkler R, Piskor EM, Kosan C. Lessons from Using Genetically Engineered Mouse Models of MYC-Induced Lymphoma. Cells 2022; 12:37. [PMID: 36611833 PMCID: PMC9818924 DOI: 10.3390/cells12010037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 12/06/2022] [Accepted: 12/15/2022] [Indexed: 12/25/2022] Open
Abstract
Oncogenic overexpression of MYC leads to the fatal deregulation of signaling pathways, cellular metabolism, and cell growth. MYC rearrangements are found frequently among non-Hodgkin B-cell lymphomas enforcing MYC overexpression. Genetically engineered mouse models (GEMMs) were developed to understand MYC-induced B-cell lymphomagenesis. Here, we highlight the advantages of using Eµ-Myc transgenic mice. We thoroughly compiled the available literature to discuss common challenges when using such mouse models. Furthermore, we give an overview of pathways affected by MYC based on knowledge gained from the use of GEMMs. We identified top regulators of MYC-induced lymphomagenesis, including some candidates that are not pharmacologically targeted yet.
Collapse
Affiliation(s)
| | | | - Christian Kosan
- Department of Biochemistry, Center for Molecular Biomedicine (CMB), Friedrich Schiller University Jena, 07745 Jena, Germany
| |
Collapse
|
2
|
Harrington CT, Sotillo E, Dang CV, Thomas-Tikhonenko A. Tilting MYC toward cancer cell death. Trends Cancer 2021; 7:982-994. [PMID: 34481764 PMCID: PMC8541926 DOI: 10.1016/j.trecan.2021.08.002] [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: 04/17/2021] [Revised: 07/30/2021] [Accepted: 08/02/2021] [Indexed: 12/12/2022]
Abstract
MYC oncoprotein promotes cell proliferation and serves as the key driver in many human cancers; therefore, considerable effort has been expended to develop reliable pharmacological methods to suppress its expression or function. Despite impressive progress, MYC-targeting drugs have not reached the clinic. Recent advances suggest that within a limited expression range unique to each tumor, MYC oncoprotein can have a paradoxical, proapoptotic function. Here we introduce a counterintuitive idea that modestly and transiently elevating MYC levels could aid chemotherapy-induced apoptosis and thus benefit the patients as much, if not more than MYC inhibition.
Collapse
Affiliation(s)
- Colleen T Harrington
- Division of Cancer Pathobiology, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Cell and Molecular Biology Graduate Group, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Elena Sotillo
- Division of Cancer Pathobiology, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Chi V Dang
- Cell and Molecular Biology Graduate Group, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA; Molecular and Cellular Oncogenesis Program, The Wistar Institute, Philadelphia, PA 19104, USA; Ludwig Institute for Cancer Research, New York, NY 10017, USA
| | - Andrei Thomas-Tikhonenko
- Division of Cancer Pathobiology, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Cell and Molecular Biology Graduate Group, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
3
|
Szydłowski M, Garbicz F, Jabłońska E, Górniak P, Komar D, Pyrzyńska B, Bojarczuk K, Prochorec-Sobieszek M, Szumera-Ciećkiewicz A, Rymkiewicz G, Cybulska M, Statkiewicz M, Gajewska M, Mikula M, Gołas A, Domagała J, Winiarska M, Graczyk-Jarzynka A, Białopiotrowicz E, Polak A, Barankiewicz J, Puła B, Pawlak M, Nowis D, Golab J, Tomirotti AM, Brzózka K, Pacheco-Blanco M, Kupcova K, Green MR, Havranek O, Chapuy B, Juszczyński P. Inhibition of PIM Kinases in DLBCL Targets MYC Transcriptional Program and Augments the Efficacy of Anti-CD20 Antibodies. Cancer Res 2021; 81:6029-6043. [PMID: 34625423 DOI: 10.1158/0008-5472.can-21-1023] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 08/18/2021] [Accepted: 10/07/2021] [Indexed: 11/16/2022]
Abstract
The family of PIM serine/threonine kinases includes three highly conserved oncogenes, PIM1, PIM2, and PIM3, which regulate multiple pro-survival pathways and cooperate with other oncogenes such as MYC. Recent genomic CRISPR-Cas9 screens further highlighted oncogenic functions of PIMs in diffuse large B cell lymphoma (DLBCL) cells, justifying development of small molecule PIM inhibitors and therapeutic targeting of PIM kinases in lymphomas. However, detailed consequences of PIM inhibition in DLBCL remain undefined. Using chemical and genetic PIM blockade, we comprehensively characterized PIM kinase-associated pro-survival functions in DLBCL and the mechanisms of PIM inhibition-induced toxicity. Treatment of DLBCL cells with SEL24/MEN1703, a pan PIM inhibitor in clinical development, decreased BAD phosphorylation and cap-dependent protein translation, reduced MCL1 expression, and induced apoptosis. PIM kinases were tightly coexpressed with MYC in diagnostic DLBCL biopsies, and PIM inhibition in cell lines and patient-derived primary lymphoma cells decreased MYC levels as well as expression of multiple MYC-dependent genes, including PLK1. Chemical and genetic PIM inhibition upregulated surface CD20 levels in a MYC-dependent fashion. Consistently, MEN1703 and other clinically available pan-PIM inhibitors synergized with the anti-CD20 monoclonal antibody rituximab in vitro, increasing complement-dependent cytotoxicity and antibody-mediated phagocytosis. Combined treatment with PIM inhibitor and rituximab suppressed tumor growth in lymphoma xenografts more efficiently than either drug alone. Taken together, these results show that targeting PIM in DLBCL exhibits pleiotropic effects that combine direct cytotoxicity with potentiated susceptibility to anti-CD20 antibodies, justifying further clinical development of such combinatorial strategies.
Collapse
Affiliation(s)
- Maciej Szydłowski
- Dept. of Experimental Hematology, Institute of Hematology and Transfusion Medicine
| | - Filip Garbicz
- Dept. of Experimental Hematology, Institute of Hematology and Transfusion Medicine
| | - Ewa Jabłońska
- Department of Diagnostic Hematology, Institute of Hematology and Transfusion Medicine
| | - Patryk Górniak
- Dept. of Experimental Hematology, Institute of Hematology and Transfusion Medicine
| | - Dorota Komar
- Dept. of Experimental Hematology, Institute of Hematology and Transfusion Medicine
| | | | - Kamil Bojarczuk
- Department of Hematology and Medical Oncology, University Medical Center - Georg-August-Universität Göttingen
| | | | - Anna Szumera-Ciećkiewicz
- Department of Pathology and Laboratory Diagnostics, IMaria Sklodowska-Curie National Research Institute of Oncology
| | - Grzegorz Rymkiewicz
- Dept. of Pathology and Laboratory Diagnostics, National Research Institute of Oncology
| | | | | | - Marta Gajewska
- Dept. of Genetics, National Research Institute of Oncology
| | - Michal Mikula
- Maria Sklodowska-Curie Memorial Cancer Center and Institute of Oncology
| | | | | | | | | | | | - Anna Polak
- Department of Diagnostic Hematology, Institute of Hematology and Transfusion Medicine
| | | | - Bartosz Puła
- Dept. of Hematology, Institute of Hematology and Transfusion Medicine
| | - Michał Pawlak
- Dept. of Experimental Hematology, Institute of Hematology and Transfusion Medicine
| | - Dominika Nowis
- Laboratory of Experimental Medicine, Medical University of Warsaw
| | - Jakub Golab
- Department of Immunology, Medical University of Warsaw
| | | | | | | | | | - Michael R Green
- Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center
| | | | - Bjoern Chapuy
- Department of Hematology and Medical Oncology, Universitätsmedizin Göttingen
| | | |
Collapse
|
4
|
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: 2.0] [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
|
5
|
Ross J, Miron CE, Plescia J, Laplante P, McBride K, Moitessier N, Möröy T. Targeting MYC: From understanding its biology to drug discovery. Eur J Med Chem 2020; 213:113137. [PMID: 33460833 DOI: 10.1016/j.ejmech.2020.113137] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 12/22/2020] [Accepted: 12/23/2020] [Indexed: 01/06/2023]
Abstract
The MYC oncogene is considered to be a high priority target for clinical intervention in cancer patients due to its aberrant activation in more than 50% of human cancers. Direct small molecule inhibition of MYC has traditionally been hampered by its intrinsically disordered nature and lack of both binding site and enzymatic activity. In recent years, however, a number of strategies for indirectly targeting MYC have emerged, guided by the advent of protein structural information and the growing set of computational tools that can be used to accelerate the hit to lead process in medicinal chemistry. In this review, we provide an overview of small molecules developed for clinical applications of these strategies, which include stabilization of the MYC guanine quadruplex, inhibition of BET factor BRD4, and disruption of the MYC:MAX heterodimer. The recent identification of novel targets for indirect MYC inhibition at the protein level is also discussed.
Collapse
Affiliation(s)
- Julie Ross
- Institut de recherches cliniques de Montréal (IRCM), 110 Pine Ave W., Montréal, Québec, H2W 1R7, Canada
| | - Caitlin E Miron
- Department of Chemistry, McGill University, 801 Sherbrooke St. W., Montréal, Québec, H3A 0B8, Canada
| | - Jessica Plescia
- Department of Chemistry, McGill University, 801 Sherbrooke St. W., Montréal, Québec, H3A 0B8, Canada
| | - Patricia Laplante
- AmorChem II Ventures Inc., 4 Westmount Sq. Bureau 160, Westmount, Québec, H3Z 2S6, Canada
| | - Kevin McBride
- AmorChem II Ventures Inc., 4 Westmount Sq. Bureau 160, Westmount, Québec, H3Z 2S6, Canada
| | - Nicolas Moitessier
- Department of Chemistry, McGill University, 801 Sherbrooke St. W., Montréal, Québec, H3A 0B8, Canada.
| | - Tarik Möröy
- Institut de recherches cliniques de Montréal (IRCM), 110 Pine Ave W., Montréal, Québec, H2W 1R7, Canada; Département de microbiologie, infectiologie et immunologie, Université de Montréal, 2900, boul. Édouard-Montpetit, Montréal, Québec, H3T 1J4, Canada; Division of Experimental Medicine, McGill University, 801 Sherbrooke St. W., Montréal, Québec, H3A 0B8, Canada.
| |
Collapse
|
6
|
Farria AT, Plummer JB, Salinger AP, Shen J, Lin K, Lu Y, McBride KM, Koutelou E, Dent SYR. Transcriptional Activation of MYC-Induced Genes by GCN5 Promotes B-cell Lymphomagenesis. Cancer Res 2020; 80:5543-5553. [PMID: 33168647 DOI: 10.1158/0008-5472.can-20-2379] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 09/29/2020] [Accepted: 10/28/2020] [Indexed: 12/19/2022]
Abstract
Overexpression of the MYC oncoprotein is an initiating step in the formation of several cancers. MYC frequently recruits chromatin-modifying complexes to DNA to amplify the expression of cancer-promoting genes, including those regulating cell cycle, proliferation, and metabolism, yet the roles of specific modifiers in different cancer types are not well defined. Here, we show that GCN5 is an essential coactivator of cell-cycle gene expression driven by MYC overexpression and that deletion of Gcn5 delays or abrogates tumorigenesis in the Eμ-Myc mouse model of B-cell lymphoma. Our results demonstrate that Gcn5 loss impacts both expression and downstream functions of Myc. SIGNIFICANCE: Our results provide important proof of principle for Gcn5 functions in formation and progression of Myc-driven cancers, suggesting that GCN5 may be a viable target for development of new cancer therapies.
Collapse
Affiliation(s)
- Aimee T Farria
- Department of Epigenetics and Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Smithville, Texas.,The Center for Cancer Epigenetics, The University of Texas MD Anderson Cancer Center, Smithville, Texas.,Program in Genetics and Epigenetics, The University of Texas MD Anderson Cancer Center UT Health Graduate School of Biomedical Sciences, Houston, Texas
| | - Joshua B Plummer
- Department of Epigenetics and Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Smithville, Texas
| | - Andrew P Salinger
- Department of Epigenetics and Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Smithville, Texas
| | - Jianjun Shen
- Department of Epigenetics and Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Smithville, Texas.,Program in Genetics and Epigenetics, The University of Texas MD Anderson Cancer Center UT Health Graduate School of Biomedical Sciences, Houston, Texas
| | - Kevin Lin
- Department of Epigenetics and Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Smithville, Texas
| | - Yue Lu
- Department of Epigenetics and Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Smithville, Texas
| | - Kevin M McBride
- Department of Epigenetics and Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Smithville, Texas.,Program in Genetics and Epigenetics, The University of Texas MD Anderson Cancer Center UT Health Graduate School of Biomedical Sciences, Houston, Texas
| | - Evangelia Koutelou
- Department of Epigenetics and Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Smithville, Texas.,The Center for Cancer Epigenetics, The University of Texas MD Anderson Cancer Center, Smithville, Texas
| | - Sharon Y R Dent
- Department of Epigenetics and Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Smithville, Texas. .,The Center for Cancer Epigenetics, The University of Texas MD Anderson Cancer Center, Smithville, Texas.,Program in Genetics and Epigenetics, The University of Texas MD Anderson Cancer Center UT Health Graduate School of Biomedical Sciences, Houston, Texas
| |
Collapse
|
7
|
Swaminathan S, Hansen AS, Heftdal LD, Dhanasekaran R, Deutzmann A, Fernandez WDM, Liefwalker DF, Horton C, Mosley A, Liebersbach M, Maecker HT, Felsher DW. MYC functions as a switch for natural killer cell-mediated immune surveillance of lymphoid malignancies. Nat Commun 2020; 11:2860. [PMID: 32503978 PMCID: PMC7275060 DOI: 10.1038/s41467-020-16447-7] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Accepted: 05/01/2020] [Indexed: 12/12/2022] Open
Abstract
The MYC oncogene drives T- and B- lymphoid malignancies, including Burkitt's lymphoma (BL) and Acute Lymphoblastic Leukemia (ALL). Here, we demonstrate a systemic reduction in natural killer (NK) cell numbers in SRα-tTA/Tet-O-MYCON mice bearing MYC-driven T-lymphomas. Residual mNK cells in spleens of MYCON T-lymphoma-bearing mice exhibit perturbations in the terminal NK effector differentiation pathway. Lymphoma-intrinsic MYC arrests NK maturation by transcriptionally repressing STAT1/2 and secretion of Type I Interferons (IFNs). Treating T-lymphoma-bearing mice with Type I IFN improves survival by rescuing NK cell maturation. Adoptive transfer of mature NK cells is sufficient to delay both T-lymphoma growth and recurrence post MYC inactivation. In MYC-driven BL patients, low expression of both STAT1 and STAT2 correlates significantly with the absence of activated NK cells and predicts unfavorable clinical outcomes. Our studies thus provide a rationale for developing NK cell-based therapies to effectively treat MYC-driven lymphomas in the future.
Collapse
MESH Headings
- Adoptive Transfer
- Animals
- Burkitt Lymphoma/immunology
- Burkitt Lymphoma/mortality
- Cell Line, Tumor/transplantation
- Disease Models, Animal
- Gene Expression Regulation, Neoplastic/immunology
- Humans
- Immunologic Surveillance/genetics
- Interferon Type I/pharmacology
- Interferon Type I/therapeutic use
- Killer Cells, Natural/drug effects
- Killer Cells, Natural/immunology
- Killer Cells, Natural/transplantation
- Lymphoma, T-Cell/drug therapy
- Lymphoma, T-Cell/genetics
- Lymphoma, T-Cell/immunology
- Lymphoma, T-Cell/pathology
- Male
- Mice
- Primary Cell Culture
- Proto-Oncogene Proteins c-myc/genetics
- Proto-Oncogene Proteins c-myc/metabolism
- STAT1 Transcription Factor/metabolism
- STAT2 Transcription Factor/metabolism
- Signal Transduction/drug effects
- Signal Transduction/genetics
- Signal Transduction/immunology
Collapse
Affiliation(s)
- Srividya Swaminathan
- Division of Oncology, Departments of Medicine and Pathology, Stanford University, Stanford, CA, USA
- Department of Systems Biology, Beckman Research Institute of City of Hope, Duarte, CA, USA
| | - Aida S Hansen
- Division of Oncology, Departments of Medicine and Pathology, Stanford University, Stanford, CA, USA
| | - Line D Heftdal
- Division of Oncology, Departments of Medicine and Pathology, Stanford University, Stanford, CA, USA
| | - Renumathy Dhanasekaran
- Division of Oncology, Departments of Medicine and Pathology, Stanford University, Stanford, CA, USA
- Division of Gastroenterology and Hepatology, Stanford University, Stanford, CA, USA
| | - Anja Deutzmann
- Division of Oncology, Departments of Medicine and Pathology, Stanford University, Stanford, CA, USA
| | - Wadie D M Fernandez
- Division of Oncology, Departments of Medicine and Pathology, Stanford University, Stanford, CA, USA
| | - Daniel F Liefwalker
- Division of Oncology, Departments of Medicine and Pathology, Stanford University, Stanford, CA, USA
| | - Crista Horton
- Division of Oncology, Departments of Medicine and Pathology, Stanford University, Stanford, CA, USA
| | - Adriane Mosley
- Division of Oncology, Departments of Medicine and Pathology, Stanford University, Stanford, CA, USA
| | - Mariola Liebersbach
- Division of Oncology, Departments of Medicine and Pathology, Stanford University, Stanford, CA, USA
| | - Holden T Maecker
- The Human Immune Monitoring Center (HIMC), Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA, USA
| | - Dean W Felsher
- Division of Oncology, Departments of Medicine and Pathology, Stanford University, Stanford, CA, USA.
| |
Collapse
|
8
|
Oon ML, Hoppe MM, Fan S, Phyu T, Phuong HM, Tan SY, Hue SSS, Wang S, Poon LM, Chan HLE, Lee J, Chee YL, Chng WJ, de Mel S, Liu X, Jeyasekharan AD, Ng SB. The contribution of MYC and PLK1 expression to proliferative capacity in diffuse large B-cell lymphoma. Leuk Lymphoma 2019; 60:3214-3224. [PMID: 31259656 DOI: 10.1080/10428194.2019.1633629] [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: 02/16/2019] [Revised: 05/24/2019] [Accepted: 06/11/2019] [Indexed: 10/26/2022]
Abstract
Polo-like kinase-1 (PLK1) regulates the MYC-dependent kinome in aggressive B-cell lymphoma. However, the role of PLK1 and MYC toward proliferation in diffuse large B-cell lymphoma (DLBCL) is unknown. We use multiplexed fluorescent immunohistochemistry (fIHC) to evaluate the co-localization of MYC, PLK1 and Ki67 to study their association with proliferation in DLBCL. The majority (98%, 95% CI 95-100%) of MYC/PLK1-double positive tumor cells expressed Ki67, underscoring the key role of the MYC/PLK1 circuit in proliferation. However, only 38% (95% CI 23-40%) and 51% (95% CI 46-51%) of Ki67-positive cells expressed MYC and PLK1, respectively. Notably, 40% (95% CI 26-43%) of Ki67-positive cells are MYC- and PLK-negative. A stronger correlation exists between PLK1 and Ki67 expression (R = 0.74, p < .001) than with MYC and Ki67 expression (R = 0.52, p < .001). Overall, the results indicate that PLK1 has a higher association than MYC in DLBCL proliferation and there are mechanisms besides MYC and PLK1 influencing DLBCL proliferation.
Collapse
Affiliation(s)
- Ming Liang Oon
- Department of Pathology, National University Hospital, National University Health System, Singapore, Singapore
| | - Michal M Hoppe
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Shuangyi Fan
- Department of Pathology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - The Phyu
- Department of Pathology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Hoang M Phuong
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Soo-Yong Tan
- Department of Pathology, National University Hospital, National University Health System, Singapore, Singapore
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
- Department of Pathology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- A*STAR, Advanced Molecular Pathology Laboratory, Institute of Molecular and Cell Biology, Singapore, Singapore
| | - Susan Swee-Shan Hue
- Department of Pathology, National University Hospital, National University Health System, Singapore, Singapore
- A*STAR, Advanced Molecular Pathology Laboratory, Institute of Molecular and Cell Biology, Singapore, Singapore
| | - Shi Wang
- Department of Pathology, National University Hospital, National University Health System, Singapore, Singapore
| | - Li M Poon
- Department of Hematology-Oncology, National University Cancer Institute Singapore, National University Hospital, National University Health System, Singapore, Singapore
| | - Hian L E Chan
- Department of Hematology-Oncology, National University Cancer Institute Singapore, National University Hospital, National University Health System, Singapore, Singapore
| | - Joanne Lee
- Department of Hematology-Oncology, National University Cancer Institute Singapore, National University Hospital, National University Health System, Singapore, Singapore
| | - Yen L Chee
- Department of Hematology-Oncology, National University Cancer Institute Singapore, National University Hospital, National University Health System, Singapore, Singapore
| | - Wee-Joo Chng
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
- Department of Hematology-Oncology, National University Cancer Institute Singapore, National University Hospital, National University Health System, Singapore, Singapore
| | - Sanjay de Mel
- Department of Hematology-Oncology, National University Cancer Institute Singapore, National University Hospital, National University Health System, Singapore, Singapore
| | - Xin Liu
- Department of Hematology-Oncology, National University Cancer Institute Singapore, National University Hospital, National University Health System, Singapore, Singapore
| | - Anand D Jeyasekharan
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
- Department of Hematology-Oncology, National University Cancer Institute Singapore, National University Hospital, National University Health System, Singapore, Singapore
| | - Siok-Bian Ng
- Department of Pathology, National University Hospital, National University Health System, Singapore, Singapore
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
- Department of Pathology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| |
Collapse
|
9
|
Ren Y, Bi C, Zhao X, Lwin T, Wang C, Yuan J, Silva AS, Shah BD, Fang B, Li T, Koomen JM, Jiang H, Chavez JC, Pham LV, Sudalagunta PR, Wan L, Wang X, Dalton WS, Moscinski LC, Shain KH, Vose J, Cleveland JL, Sotomayor EM, Fu K, Tao J. PLK1 stabilizes a MYC-dependent kinase network in aggressive B cell lymphomas. J Clin Invest 2018; 128:5517-5530. [PMID: 30260324 PMCID: PMC6264635 DOI: 10.1172/jci122533] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Accepted: 09/20/2018] [Indexed: 12/13/2022] Open
Abstract
Concordant activation of MYC and BCL-2 oncoproteins in double-hit lymphoma (DHL) results in aggressive disease that is refractory to treatment. By integrating activity-based proteomic profiling and drug screens, polo-like kinase-1 (PLK1) was identified as an essential regulator of the MYC-dependent kinome in DHL. Notably, PLK1 was expressed at high levels in DHL, correlated with MYC expression, and connoted poor outcome. Further, PLK1 signaling augmented MYC protein stability, and in turn, MYC directly induced PLK1 transcription, establishing a feed-forward MYC-PLK1 circuit in DHL. Finally, inhibition of PLK1 triggered degradation of MYC and of the antiapoptotic protein MCL-1, and PLK1 inhibitors showed synergy with BCL-2 antagonists in blocking DHL cell growth, survival, and tumorigenicity, supporting clinical targeting of PLK1 in DHL.
Collapse
Affiliation(s)
- Yuan Ren
- Department of Laboratory Medicine and Hematopathology, Moffitt Cancer Center & Research Institute, Tampa, Florida, USA
| | - Chengfeng Bi
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Xiaohong Zhao
- Department of Laboratory Medicine and Hematopathology, Moffitt Cancer Center & Research Institute, Tampa, Florida, USA
| | - Tint Lwin
- Department of Laboratory Medicine and Hematopathology, Moffitt Cancer Center & Research Institute, Tampa, Florida, USA
| | - Cheng Wang
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Ji Yuan
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | | | | | - Bin Fang
- Proteomics Core Facility, Moffitt Cancer Center & Research Institute, Tampa, Florida, USA
| | - Tao Li
- Department of Laboratory Medicine and Hematopathology, Moffitt Cancer Center & Research Institute, Tampa, Florida, USA
| | - John M. Koomen
- Proteomics Core Facility, Moffitt Cancer Center & Research Institute, Tampa, Florida, USA
| | - Huijuan Jiang
- Department of Laboratory Medicine and Hematopathology, Moffitt Cancer Center & Research Institute, Tampa, Florida, USA
- Tianjin Medical School, Tianjin, China
| | | | - Lan V. Pham
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | | | - Lixin Wan
- Department of Molecular Oncology and
| | - Xuefeng Wang
- Department of Biostatics and Bioinformatics, Moffitt Cancer Center & Research Institute, Tampa, Florida, USA
| | | | - Lynn C. Moscinski
- Department of Laboratory Medicine and Hematopathology, Moffitt Cancer Center & Research Institute, Tampa, Florida, USA
| | | | - Julie Vose
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - John L. Cleveland
- Department of Tumor Biology, Moffitt Cancer Center & Research Institute, Tampa, Florida, USA
| | - Eduardo M. Sotomayor
- Department of Hematology & Oncology, George Washington University, Washington, DC, USA
| | - Kai Fu
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Jianguo Tao
- Department of Laboratory Medicine and Hematopathology, Moffitt Cancer Center & Research Institute, Tampa, Florida, USA
- Department of Malignant Hematology, and
| |
Collapse
|
10
|
Knauf JA, Luckett KA, Chen KY, Voza F, Socci ND, Ghossein R, Fagin JA. Hgf/Met activation mediates resistance to BRAF inhibition in murine anaplastic thyroid cancers. J Clin Invest 2018; 128:4086-4097. [PMID: 29990309 DOI: 10.1172/jci120966] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Accepted: 07/03/2018] [Indexed: 02/01/2023] Open
Abstract
Anaplastic thyroid carcinomas (ATCs) have a high prevalence of BRAF and TP53 mutations. A trial of vemurafenib in nonmelanoma BRAFV600E-mutant cancers showed significant, although short-lived, responses in ATCs, indicating that these virulent tumors remain addicted to BRAF despite their high mutation burden. To explore the mechanisms mediating acquired resistance to BRAF blockade, we generated mice with thyroid-specific deletion of p53 and dox-dependent expression of BRAFV600E, 50% of which developed ATCs after dox treatment. Upon dox withdrawal there was complete regression in all mice, although recurrences were later detected in 85% of animals. The relapsed tumors had elevated MAPK transcriptional output, and retained responses to the MEK/RAF inhibitor CH5126766 in vivo and in vitro. Whole-exome sequencing identified recurrent focal amplifications of chromosome 6, with a minimal region of overlap that included Met. Met-amplified recurrences overexpressed the receptor as well as its ligand Hgf. Growth, signaling, and viability of Met-amplified tumor cells were suppressed in vitro and in vivo by the Met kinase inhibitors PF-04217903 and crizotinib, whereas primary ATCs and Met-diploid relapses were resistant. Hence, recurrences are the rule after BRAF suppression in murine ATCs, most commonly due to activation of HGF/MET signaling, which generates exquisite dependency to MET kinase inhibitors.
Collapse
Affiliation(s)
- Jeffrey A Knauf
- Human Oncology and Pathogenesis Program.,Department of Medicine
| | | | | | | | | | - Ronald Ghossein
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - James A Fagin
- Human Oncology and Pathogenesis Program.,Department of Medicine.,Department of Medicine, Weill-Cornell Medical College, New York, New York, USA
| |
Collapse
|
11
|
D'Andrea A, Gritti I, Nicoli P, Giorgio M, Doni M, Conti A, Bianchi V, Casoli L, Sabò A, Mironov A, Beznoussenko GV, Amati B. The mitochondrial translation machinery as a therapeutic target in Myc-driven lymphomas. Oncotarget 2018; 7:72415-72430. [PMID: 27635472 PMCID: PMC5341918 DOI: 10.18632/oncotarget.11719] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Accepted: 08/25/2016] [Indexed: 12/12/2022] Open
Abstract
The oncogenic transcription factor Myc is required for the progression and maintenance of diverse tumors. This has led to the concept that Myc itself, Myc-activated gene products, or associated biological processes might constitute prime targets for cancer therapy. Here, we present an in vivo reverse-genetic screen targeting a set of 241 Myc-activated mRNAs in mouse B-cell lymphomas, unraveling a critical role for the mitochondrial ribosomal protein (MRP) Ptcd3 in tumor maintenance. Other MRP-coding genes were also up regulated in Myc-induced lymphoma, pointing to a coordinate activation of the mitochondrial translation machinery. Inhibition of mitochondrial translation with the antibiotic Tigecycline was synthetic-lethal with Myc activation, impaired respiratory activity and tumor cell survival in vitro, and significantly extended lifespan in lymphoma-bearing mice. We have thus identified a novel Myc-induced metabolic dependency that can be targeted by common antibiotics, opening new therapeutic perspectives in Myc-overexpressing tumors.
Collapse
Affiliation(s)
- Aleco D'Andrea
- Department of Experimental Oncology, European Institute of Oncology, Milan, Italy
| | - Ilaria Gritti
- Department of Experimental Oncology, European Institute of Oncology, Milan, Italy.,Present address: IRCCS San Raffaele, Functional Genomics of Cancer Unit, Division of Experimental Oncology, Milan, Italy
| | - Paola Nicoli
- Department of Experimental Oncology, European Institute of Oncology, Milan, Italy
| | - Marco Giorgio
- Department of Experimental Oncology, European Institute of Oncology, Milan, Italy
| | - Mirko Doni
- Department of Experimental Oncology, European Institute of Oncology, Milan, Italy
| | - Annalisa Conti
- Center for Genomic Science of IIT@SEMM, Fondazione Istituto Italiano di Tecnologia, Milan, Italy
| | - Valerio Bianchi
- Center for Genomic Science of IIT@SEMM, Fondazione Istituto Italiano di Tecnologia, Milan, Italy.,Present address: Hubrecht Institute-KNAW & University Medical Center Utrecht, Uppsalalaan, Utrecht, The Netherlands
| | - Lucia Casoli
- Center for Genomic Science of IIT@SEMM, Fondazione Istituto Italiano di Tecnologia, Milan, Italy
| | - Arianna Sabò
- Center for Genomic Science of IIT@SEMM, Fondazione Istituto Italiano di Tecnologia, Milan, Italy
| | - Alexandre Mironov
- The Institute of Molecular Oncology of the Italian Foundation for Cancer Research, Milan, Italy
| | - Galina V Beznoussenko
- The Institute of Molecular Oncology of the Italian Foundation for Cancer Research, Milan, Italy
| | - Bruno Amati
- Department of Experimental Oncology, European Institute of Oncology, Milan, Italy.,Center for Genomic Science of IIT@SEMM, Fondazione Istituto Italiano di Tecnologia, Milan, Italy
| |
Collapse
|
12
|
Li Y, Deutzmann A, Choi PS, Fan AC, Felsher DW. BIM mediates oncogene inactivation-induced apoptosis in multiple transgenic mouse models of acute lymphoblastic leukemia. Oncotarget 2017; 7:26926-34. [PMID: 27095570 PMCID: PMC5053622 DOI: 10.18632/oncotarget.8731] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Accepted: 04/08/2016] [Indexed: 02/07/2023] Open
Abstract
Oncogene inactivation in both clinical targeted therapies and conditional transgenic mouse cancer models can induce significant tumor regression associated with the robust induction of apoptosis. Here we report that in MYC-, RAS-, and BCR-ABL-induced acute lymphoblastic leukemia (ALL), apoptosis upon oncogene inactivation is mediated by the same pro-apoptotic protein, BIM. The induction of BIMin the MYC- and RAS-driven leukemia is mediated by the downregulation of miR-17-92. Overexpression of miR-17-92 blocked the induction of apoptosis upon oncogene inactivation in the MYC and RAS-driven but not in the BCR-ABL-driven ALL leukemia. Hence, our results provide novel insight into the mechanism of apoptosis upon oncogene inactivation and suggest that induction of BIM-mediated apoptosis may be an important therapeutic approach for ALL.
Collapse
Affiliation(s)
- Yulin Li
- Division of Oncology, Department of Medicine and Pathology, Stanford University, Stanford, CA, United States of America
| | - Anja Deutzmann
- Division of Oncology, Department of Medicine and Pathology, Stanford University, Stanford, CA, United States of America
| | - Peter S Choi
- Division of Oncology, Department of Medicine and Pathology, Stanford University, Stanford, CA, United States of America
| | - Alice C Fan
- Division of Oncology, Department of Medicine and Pathology, Stanford University, Stanford, CA, United States of America
| | - Dean W Felsher
- Division of Oncology, Department of Medicine and Pathology, Stanford University, Stanford, CA, United States of America
| |
Collapse
|
13
|
DNA-binding of the Tet-transactivator curtails antigen-induced lymphocyte activation in mice. Nat Commun 2017; 8:1028. [PMID: 29044097 PMCID: PMC5647323 DOI: 10.1038/s41467-017-01022-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Accepted: 08/14/2017] [Indexed: 12/31/2022] Open
Abstract
The Tet-On/Off system for conditional transgene expression constitutes state-of-the-art technology to study gene function by facilitating inducible expression in a timed and reversible manner. Several studies documented the suitability and versatility of this system to trace lymphocyte fate and to conditionally express oncogenes or silence tumour suppressor genes in vivo. Here, we show that expression of the tetracycline/doxycycline-controlled Tet-transactivator, while tolerated well during development and in immunologically unchallenged animals, impairs the expansion of antigen-stimulated T and B cells and thereby curtails adaptive immune responses in vivo. Transactivator-mediated cytotoxicity depends on DNA binding, but can be overcome by BCL2 overexpression, suggesting that apoptosis induction upon lymphocyte activation limits cellular and humoral immune responses. Our findings suggest a possible system-intrinsic biological bias of the Tet-On/Off system in vivo that will favour the outgrowth of apoptosis resistant clones, thus possibly confounding data published using such systems. Tet-transactivators are used for direct regulation of gene expression, RNA interference and for CRISPR/Cas9-based systems. Here the authors show that DNA-bound Tet-transactivators can induce cell death in antigen-activated lymphocytes in vivo, putting into question the use of, and in vivo data generated with, these molecular tools.
Collapse
|
14
|
p19ARF is a critical mediator of both cellular senescence and an innate immune response associated with MYC inactivation in mouse model of acute leukemia. Oncotarget 2016; 6:3563-77. [PMID: 25784651 PMCID: PMC4414137 DOI: 10.18632/oncotarget.2969] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2014] [Accepted: 12/17/2014] [Indexed: 11/30/2022] Open
Abstract
MYC-induced T-ALL exhibit oncogene addiction. Addiction to MYC is a consequence of both cell-autonomous mechanisms, such as proliferative arrest, cellular senescence, and apoptosis, as well as non-cell autonomous mechanisms, such as shutdown of angiogenesis, and recruitment of immune effectors. Here, we show, using transgenic mouse models of MYC-induced T-ALL, that the loss of either p19ARF or p53 abrogates the ability of MYC inactivation to induce sustained tumor regression. Loss of p53 or p19ARF, influenced the ability of MYC inactivation to elicit the shutdown of angiogenesis; however the loss of p19ARF, but not p53, impeded cellular senescence, as measured by SA-beta-galactosidase staining, increased expression of p16INK4A, and specific histone modifications. Moreover, comparative gene expression analysis suggested that a multitude of genes involved in the innate immune response were expressed in p19ARF wild-type, but not null, tumors upon MYC inactivation. Indeed, the loss of p19ARF, but not p53, impeded the in situ recruitment of macrophages to the tumor microenvironment. Finally, p19ARF null-associated gene signature prognosticated relapse-free survival in human patients with ALL. Therefore, p19ARF appears to be important to regulating cellular senescence and innate immune response that may contribute to the therapeutic response of ALL.
Collapse
|
15
|
Schrecengost RS, Keller SN, Schiewer MJ, Knudsen KE, Smith CD. Downregulation of Critical Oncogenes by the Selective SK2 Inhibitor ABC294640 Hinders Prostate Cancer Progression. Mol Cancer Res 2015; 13:1591-601. [PMID: 26271487 DOI: 10.1158/1541-7786.mcr-14-0626] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2014] [Accepted: 07/30/2015] [Indexed: 12/12/2022]
Abstract
UNLABELLED The bioactive sphingolipid sphingosine-1-phosphate (S1P) drives several hallmark processes of cancer, making the enzymes that synthesize S1P, that is, sphingosine kinase 1 and 2 (SK1 and SK2), important molecular targets for cancer drug development. ABC294640 is a first-in-class SK2 small-molecule inhibitor that effectively inhibits cancer cell growth in vitro and in vivo. Given that AR and Myc are two of the most widely implicated oncogenes in prostate cancer, and that sphingolipids affect signaling by both proteins, the therapeutic potential for using ABC294640 in the treatment of prostate cancer was evaluated. This study demonstrates that ABC294640 abrogates signaling pathways requisite for prostate cancer growth and proliferation. Key findings validate that ABC294640 treatment of early-stage and advanced prostate cancer models downregulate Myc and AR expression and activity. This corresponds with significant inhibition of growth, proliferation, and cell-cycle progression. Finally, oral administration of ABC294640 was found to dramatically impede xenograft tumor growth. Together, these pre-clinical findings support the hypotheses that SK2 activity is required for prostate cancer function and that ABC294640 represents a new pharmacological agent for treatment of early stage and aggressive prostate cancer. IMPLICATIONS Sphingosine kinase inhibition disrupts multiple oncogenic signaling pathways that are deregulated in prostate cancer.
Collapse
Affiliation(s)
| | - Staci N Keller
- Apogee Biotechnology Corporation, Hummelstown, Pennsylvania
| | - Matthew J Schiewer
- Department of Cancer Biology, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Karen E Knudsen
- Department of Cancer Biology, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania. Department of Urology, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania. Department of Radiation Oncology, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Charles D Smith
- Apogee Biotechnology Corporation, Hummelstown, Pennsylvania.
| |
Collapse
|
16
|
Methods to study primary tumor cells and residual tumor cells in mouse models of oncogene dependence. Methods Mol Biol 2015. [PMID: 25636480 DOI: 10.1007/978-1-4939-2297-0_19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
Abstract
The studies of oncogene dependence are aimed to understand an unfortunate and puzzling aspect of targeted anticancer treatments-their progression to drug resistance. Drug resistance develops from a pool of cells that survive the original treatment, called minimal residual disease. Mouse models based on tetracycline-dependent expression of transgenic oncogenes are used to imitate targeted oncogene blockade and to reproduce minimal residual disease in humans. Here we describe a novel method for generating oncogene-dependent mammary tumors using somatic transfer of transactivator-containing retroviruses into transgenic mice with tetracycline-dependent oncogenes and a method for measuring continuous mitotic activity in epithelial cells in real time.
Collapse
|
17
|
Zhang J, Lou X, Zellmer L, Liu S, Xu N, Liao DJ. Just like the rest of evolution in Mother Nature, the evolution of cancers may be driven by natural selection, and not by haphazard mutations. Oncoscience 2014; 1:580-90. [PMID: 25594068 PMCID: PMC4278337 DOI: 10.18632/oncoscience.83] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2014] [Accepted: 09/17/2014] [Indexed: 12/24/2022] Open
Abstract
Sporadic carcinogenesis starts from immortalization of a differentiated somatic cell or an organ-specific stem cell. The immortalized cell incepts a new or quasinew organism that lives like a parasite in the patient and usually proceeds to progressive simplification, constantly engendering intermediate organisms that are simpler than normal cells. Like organismal evolution in Mother Nature, this cellular simplification is a process of Darwinian selection of those mutations with growth- or survival-advantages, from numerous ones that occur randomly and stochastically. Therefore, functional gain of growth- or survival-sustaining oncogenes and functional loss of differentiation-sustaining tumor suppressor genes, which are hallmarks of cancer cells and contribute to phenotypes of greater malignancy, are not drivers of carcinogenesis but are results from natural selection of advantageous mutations. Besides this mutation-load dependent survival mechanism that is evolutionarily low and of an asexual nature, cancer cells may also use cell fusion for survival, which is an evolutionarily-higher mechanism and is of a sexual nature. Assigning oncogenes or tumor suppressor genes or their mutants as drivers to induce cancer in animals may somewhat coerce them to create man-made oncogenic pathways that may not really be a course of sporadic cancer formations in the human.
Collapse
Affiliation(s)
- Ju Zhang
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, P.R. China
| | - Xiaomin Lou
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, P.R. China
| | - Lucas Zellmer
- Hormel Institute, University of Minnesota, Austin, MN 55912, USA
| | - Siqi Liu
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, P.R. China
| | - Ningzhi Xu
- Laboratory of Cell and Molecular Biology, Cancer Institute, Chinese Academy of Medical Science, Beijing 100021, P.R. China
| | - D Joshua Liao
- Hormel Institute, University of Minnesota, Austin, MN 55912, USA
| |
Collapse
|
18
|
Sewastianik T, Prochorec-Sobieszek M, Chapuy B, Juszczyński P. MYC deregulation in lymphoid tumors: molecular mechanisms, clinical consequences and therapeutic implications. Biochim Biophys Acta Rev Cancer 2014; 1846:457-67. [PMID: 25199984 DOI: 10.1016/j.bbcan.2014.08.006] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2014] [Revised: 08/28/2014] [Accepted: 08/28/2014] [Indexed: 12/24/2022]
Abstract
MYC is one of the most frequently deregulated oncogenes in human malignancies. It encodes a leucine zipper transcription factor that modulates a broad spectrum of cellular genes responsible for enhancing cell proliferation, cellular metabolism, growth, angiogenesis, metastasis, genomic instability, stem cell self-renewal and reduced differentiation. MYC functions predominantly as an amplifier of expression of already active genes, potentiating the pre-existing transcriptional program, although it can also repress certain transcriptional targets. In mouse models, MYC induces lymphomas, but requires cooperation with other lesions, including inactivation of the p53 pathway, structural alterations of BCL2 family members, or increased PI3K activity. In human B-cell tumors, MYC rearrangements involving the 8q24 region and immunoglobulin heavy or light genes are a hallmark of Burkitt lymphoma (BL), but can also occur in other lymphoid malignancies, that include diffuse large B-cell lymphoma (DLBCL), B-cell lymphoma, unclassifiable, with features intermediate between DLBCL and Burkitt lymphoma (BCLU), plasma cell myeloma (PCM), mantle cell lymphoma (MCL) and plasmablastic lymphoma. For non-BL lymphoid malignancies, MYC fusions represent secondary genetic events and exist in the context of complex karyotypes. Regardless of the mechanism deregulating MYC, lymphomas over-expressing MYC are addicted to this oncogene, highlighting the potential clinical utility of MYC targeting strategies. Several promising approaches for pharmaceutical intervention have been suggested which are now in preclinical or clinical development. Herein, we therefore review the molecular pathogenetic mechanisms associated with MYC deregulation in human B-cell lymphomas and their implications for therapies targeting MYC.
Collapse
Affiliation(s)
- Tomasz Sewastianik
- Institute of Hematology and Transfusion Medicine, Department of Diagnostic Hematology, Indiry Gandhi Str. 14, 02-776 Warsaw, Poland
| | - Monika Prochorec-Sobieszek
- Institute of Hematology and Transfusion Medicine, Department of Diagnostic Hematology, Indiry Gandhi Str. 14, 02-776 Warsaw, Poland
| | - Bjoern Chapuy
- Dana-Farber Cancer Institute, Harvard Medical School, Department of Medical Oncology, 450 Brookline Avenue, Boston, MA 02215, USA
| | - Przemysław Juszczyński
- Institute of Hematology and Transfusion Medicine, Department of Diagnostic Hematology, Indiry Gandhi Str. 14, 02-776 Warsaw, Poland.
| |
Collapse
|
19
|
Li Y, Choi PS, Casey SC, Dill DL, Felsher DW. MYC through miR-17-92 suppresses specific target genes to maintain survival, autonomous proliferation, and a neoplastic state. Cancer Cell 2014; 26:262-72. [PMID: 25117713 PMCID: PMC4191901 DOI: 10.1016/j.ccr.2014.06.014] [Citation(s) in RCA: 144] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2013] [Revised: 03/23/2014] [Accepted: 06/17/2014] [Indexed: 01/04/2023]
Abstract
The MYC oncogene regulates gene expression through multiple mechanisms, and its overexpression culminates in tumorigenesis. MYC inactivation reverses turmorigenesis through the loss of distinguishing features of cancer, including autonomous proliferation and survival. Here we report that MYC via miR-17-92 maintains a neoplastic state through the suppression of chromatin regulatory genes Sin3b, Hbp1, Suv420h1, and Btg1, as well as the apoptosis regulator Bim. The enforced expression of miR-17-92 prevents MYC suppression from inducing proliferative arrest, senescence, and apoptosis and abrogates sustained tumor regression. Knockdown of the five miR-17-92 target genes blocks senescence and apoptosis while it modestly delays proliferative arrest, thus partially recapitulating miR-17-92 function. We conclude that MYC, via miR-17-92, maintains a neoplastic state by suppressing specific target genes.
Collapse
Affiliation(s)
- Yulin Li
- Division of Oncology, Department of Medicine and Pathology, Stanford University, Stanford, CA 94305, USA
| | - Peter S Choi
- Division of Oncology, Department of Medicine and Pathology, Stanford University, Stanford, CA 94305, USA
| | - Stephanie C Casey
- Division of Oncology, Department of Medicine and Pathology, Stanford University, Stanford, CA 94305, USA
| | - David L Dill
- Department of Computer Science, Stanford University, Stanford, CA 94305, USA
| | - Dean W Felsher
- Division of Oncology, Department of Medicine and Pathology, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
20
|
Addiction to multiple oncogenes can be exploited to prevent the emergence of therapeutic resistance. Proc Natl Acad Sci U S A 2014; 111:E3316-24. [PMID: 25071175 DOI: 10.1073/pnas.1406123111] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Many cancers exhibit sensitivity to the inhibition of a single genetic lesion, a property that has been successfully exploited with oncogene-targeted therapeutics. However, inhibition of single oncogenes often fails to result in sustained tumor regression due to the emergence of therapy-resistant cells. Here, we report that MYC-driven lymphomas frequently acquire activating mutations in β-catenin, including a previously unreported mutation in a splice acceptor site. Tumors with these genetic lesions are highly dependent on β-catenin for their survival and the suppression of β-catenin resulted in marked apoptosis causally related to a decrease in Bcl-xL expression. Using a novel inducible inhibitor of β-catenin, we illustrate that, although MYC withdrawal or β-catenin inhibition alone results in initial tumor regression, most tumors ultimately recurred, mimicking the clinical response to single-agent targeted therapy. Importantly, the simultaneous combined inhibition of both MYC and β-catenin promoted more rapid tumor regression and successfully prevented tumor recurrence. Hence, we demonstrated that MYC-induced tumors are addicted to mutant β-catenin, and the combined inactivation of MYC and β-catenin induces sustained tumor regression. Our results provide a proof of principle that targeting multiple oncogene addicted pathways can prevent therapeutic resistance.
Collapse
|
21
|
Abstract
The MYC proto-oncogene is an essential regulator of many normal biological programmes. MYC, when activated as an oncogene, has been implicated in the pathogenesis of most types of human cancers. MYC overexpression in normal cells is restrained from causing cancer through multiple genetically and epigenetically controlled checkpoint mechanisms, including proliferative arrest, apoptosis and cellular senescence. When pathologically activated in the correct epigenetic and genetic contexts, MYC bypasses these mechanisms and drives many of the 'hallmark' features of cancer, including uncontrolled tumour growth associated with DNA replication and transcription, cellular proliferation and growth, protein synthesis and altered cellular metabolism. MYC also dictates tumour cell fate by enforcing self-renewal and by abrogating cellular senescence and differentiation programmes. Moreover, MYC influences the tumour microenvironment, including activating angiogenesis and suppressing the host immune response. Provocatively, brief or even partial suppression of MYC back to its physiological levels of activation can lead to the restoration of intrinsic checkpoint mechanisms, resulting in acute and sustained tumour regression associated with tumour cells undergoing proliferative arrest, differentiation, senescence and apoptosis, as well as remodelling of the tumour microenvironment, recruitment of an immune response and shutdown of angiogenesis. Hence, tumours appear to be addicted to the MYC oncogene because of both tumour cell intrinsic and host-dependent mechanisms. MYC is important for the regulation of both the initiation and maintenance of tumorigenesis.
Collapse
Affiliation(s)
- Y Li
- Division of Oncology, Departments of Medicine and Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | | | | |
Collapse
|
22
|
Gabay M, Li Y, Felsher DW. MYC activation is a hallmark of cancer initiation and maintenance. Cold Spring Harb Perspect Med 2014; 4:4/6/a014241. [PMID: 24890832 DOI: 10.1101/cshperspect.a014241] [Citation(s) in RCA: 581] [Impact Index Per Article: 58.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The MYC proto-oncogene has been implicated in the pathogenesis of most types of human tumors. MYC activation alone in many normal cells is restrained from causing tumorigenesis through multiple genetic and epigenetically controlled checkpoint mechanisms, including proliferative arrest, apoptosis, and cellular senescence. When pathologically activated in a permissive epigenetic and/or genetic context, MYC bypasses these mechanisms, enforcing many of the "hallmark" features of cancer, including relentless tumor growth associated with DNA replication and transcription, cellular proliferation and growth, protein synthesis, and altered cellular metabolism. MYC mandates tumor cell fate, by inducing stemness and blocking cellular senescence and differentiation. Additionally, MYC orchestrates changes in the tumor microenvironment, including the activation of angiogenesis and suppression of the host immune response. Provocatively, brief or even partial suppression of MYC back to its physiological levels of activation can result in the restoration of intrinsic checkpoint mechanisms, resulting in acute and sustained tumor regression, associated with tumor cells undergoing proliferative arrest, differentiation, senescence, and apoptosis, as well as remodeling of the tumor microenvironment, recruitment of an immune response, and shutdown of angiogenesis. Hence, tumors appear to be "addicted" to MYC because of both tumor cell-intrinsic, cell-autonomous and host-dependent, immune cell-dependent mechanisms. Both the trajectory and persistence of many human cancers require sustained MYC activation. Multiscale mathematical modeling may be useful to predict when tumors will be addicted to MYC. MYC is a hallmark molecular feature of both the initiation and maintenance of tumorigenesis.
Collapse
Affiliation(s)
- Meital Gabay
- Division of Oncology, Departments of Medicine and Pathology, Stanford University School of Medicine, Stanford, California 94305
| | - Yulin Li
- Division of Oncology, Departments of Medicine and Pathology, Stanford University School of Medicine, Stanford, California 94305
| | - Dean W Felsher
- Division of Oncology, Departments of Medicine and Pathology, Stanford University School of Medicine, Stanford, California 94305
| |
Collapse
|
23
|
Hales EC, Taub JW, Matherly LH. New insights into Notch1 regulation of the PI3K–AKT–mTOR1 signaling axis: Targeted therapy of γ-secretase inhibitor resistant T-cell acute lymphoblastic leukemia. Cell Signal 2014; 26:149-61. [DOI: 10.1016/j.cellsig.2013.09.021] [Citation(s) in RCA: 130] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2013] [Accepted: 09/30/2013] [Indexed: 02/01/2023]
|
24
|
Nwabugwu CI, Rakhra K, Felsher DW, Paik DS. A tumor-immune mathematical model of CD4+ T helper cell dependent tumor regression by oncogene inactivation. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2013; 2013:4529-32. [PMID: 24110741 DOI: 10.1109/embc.2013.6610554] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Understanding the complex dynamics between the tumor cells and the host immune system will be key to improved therapeutic strategies against cancer. We propose an ODE-based mathematical model of both the tumor and immune system and how they respond to inactivation of the driving oncogene. Our model supports experimental results showing that cellular senescence of tumor cells is dependent on CD4+ T helper cells, leading to relapse of tumors in immunocompromised hosts.
Collapse
|
25
|
Fan AC, O'Rourke JJ, Praharaj DR, Felsher DW. Real-time nanoscale proteomic analysis of the novel multi-kinase pathway inhibitor rigosertib to measure the response to treatment of cancer. Expert Opin Investig Drugs 2013; 22:1495-509. [PMID: 23937225 DOI: 10.1517/13543784.2013.829453] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION Rigosertib (ON01910.Na), is a targeted therapeutic that inhibits multiple kinases, including PI3K and PIk-1. Rigosertib has been found to induce the proliferative arrest and apoptosis of myeloblasts but not of other normal hematopoietic cells. Rigosertib has significant clinical activity as a therapy for patients with high-risk myelodysplastic syndrome who are otherwise refractory to DNA methyltransferase inhibitors. Moreover, rigosertib has potential clinical activity in a multitude of solid tumors. AREAS COVERED The objective of this review is to evaluate the mechanism of activity, efficacy and dosing of rigosertib. Furthermore, the challenge in the clinical development of rigosertib, to identify the specific patients that are most likely to benefit from this therapeutic agent, is discussed. A PubMed search was performed using the following key words: rigosertib and ON01910.Na. EXPERT OPINION We describe the application of a novel nanoscale proteomic assay, the nanoimmunoassay, a tractable approach for measuring the activity and predicting the efficacy of rigosertib, in real-time, using limited human clinical specimens. Our strategy suggests a possible paradigm where proteomic analysis during the pre-clinical and clinical development of a therapy can be used to uncover biomarkers for the analysis and prediction of efficacy in human patients.
Collapse
Affiliation(s)
- Alice C Fan
- Stanford University School of Medicine, Division of Oncology, Departments of Medicine and Pathology , Stanford, CA , USA
| | | | | | | |
Collapse
|
26
|
In vivo imaging-based mathematical modeling techniques that enhance the understanding of oncogene addiction in relation to tumor growth. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2013; 2013:802512. [PMID: 23573174 PMCID: PMC3616361 DOI: 10.1155/2013/802512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/21/2012] [Accepted: 02/15/2013] [Indexed: 11/18/2022]
Abstract
The dependence on the overexpression of a single oncogene constitutes an exploitable weakness for molecular targeted therapy. These drugs can produce dramatic tumor regression by targeting the driving oncogene, but relapse often follows. Understanding the complex interactions of the tumor's multifaceted response to oncogene inactivation is key to tumor regression. It has become clear that a collection of cellular responses lead to regression and that immune-mediated steps are vital to preventing relapse. Our integrative mathematical model includes a variety of cellular response mechanisms of tumors to oncogene inactivation. It allows for correct predictions of the time course of events following oncogene inactivation and their impact on tumor burden. A number of aspects of our mathematical model have proven to be necessary for recapitulating our experimental results. These include a number of heterogeneous tumor cell states since cells following different cellular programs have vastly different fates. Stochastic transitions between these states are necessary to capture the effect of escape from oncogene addiction (i.e., resistance). Finally, delay differential equations were used to accurately model the tumor growth kinetics that we have observed. We use this to model oncogene addiction in MYC-induced lymphoma, osteosarcoma, and hepatocellular carcinoma.
Collapse
|
27
|
Noncanonical roles of the immune system in eliciting oncogene addiction. Curr Opin Immunol 2013; 25:246-58. [PMID: 23571026 DOI: 10.1016/j.coi.2013.02.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2012] [Revised: 02/07/2013] [Accepted: 02/08/2013] [Indexed: 02/08/2023]
Abstract
Cancer is highly complex. The magnitude of this complexity makes it highly surprising that even the brief suppression of an oncogene can sometimes result in rapid and sustained tumor regression, illustrating that cancers can be 'oncogene addicted' [1-10]. The essential implication is that oncogenes may not only fuel the initiation of tumorigenesis, but in some cases must be excessively activated to maintain a neoplastic state [11]. Oncogene suppression acutely restores normal physiological programs that effectively overrides secondary genetic events and a cancer collapses [12,13]. Oncogene addiction is the description of the dramatic and sustained regression of some cancers upon the specific inactivation of a single oncogene [1-13,14(••),15,16(••)], that can occur through tumor intrinsic [1,2,4,12], but also host immune mechanisms [17-23]. Notably, oncogene inactivation elicits a host immune response that involves specific immune effectors and cytokines that facilitate a remodeling of the tumor microenvironment including the shut down of angiogenesis and the induction of cellular senescence of tumor cells [16(••)]. Hence, immune effectors are not only critically involved in tumor prevention, initiation [17-19], and progression [20], but also appear to be essential to tumor regression upon oncogene inactivation [21,22(••),23(••)]. Understanding how the inactivation of an oncogene elicits a systemic signal in the host that prompts a deconstruction of a tumor could have important implications. The combination of oncogene-targeted therapy together with immunomodulatory therapy may be ideal for the development of both robust tumor intrinsic and immunological responses, effectively leading to sustained tumor regression.
Collapse
|
28
|
Bellovin DI, Das B, Felsher DW. Tumor dormancy, oncogene addiction, cellular senescence, and self-renewal programs. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2013; 734:91-107. [PMID: 23143977 DOI: 10.1007/978-1-4614-1445-2_6] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Cancers are frequently addicted to initiating oncogenes that elicit aberrant cellular proliferation, self-renewal, and apoptosis. Restoration of oncogenes to normal physiologic regulation can elicit dramatic reversal of the neoplastic phenotype, including reduced proliferation and increased apoptosis of tumor cells (Science 297(5578):63-64, 2002). In some cases, oncogene inactivation is associated with compete elimination of a tumor. However, in other cases, oncogene inactivation induces a conversion of tumor cells to a dormant state that is associated with cellular differentiation and/or loss of the ability to self-replicate. Importantly, this dormant state is reversible, with tumor cells regaining the ability to self-renew upon oncogene reactivation. Thus, understanding the mechanism of oncogene inactivation-induced dormancy may be crucial for predicting therapeutic outcome of targeted therapy. One important mechanistic insight into tumor dormancy is that oncogene addiction might involve regulation of a decision between self-renewal and cellular senescence. Recent evidence suggests that this decision is regulated by multiple mechanisms that include tumor cell-intrinsic, cell-autonomous mechanisms and host-dependent, tumor cell-non-autonomous programs (Mol Cell 4(2):199-207, 1999; Science 297(5578):102-104, 2002; Nature 431(7012):1112-1117, 2004; Proc Natl Acad Sci U S A 104(32):13028-13033, 2007). In particular, the tumor microenvironment, which is known to be critical during tumor initiation (Cancer Cell 7(5):411-423, 2005; J Clin Invest 121(6):2436-2446, 2011), prevention (Nature 410(6832):1107-1111, 2001), and progression (Cytokine Growth Factor Rev 21(1):3-10, 2010), also appears to dictate when oncogene inactivation elicits the permanent loss of self-renewal through induction of cellular senescence (Nat Rev Clin Oncol 8(3):151-160, 2011; Science 313(5795):1960-1964, 2006; N Engl J Med 351(21):2159-21569, 2004). Thus, oncogene addiction may be best modeled as a consequence of the interplay amongst cell-autonomous and host-dependent programs that define when a therapy will result in tumor dormancy.
Collapse
Affiliation(s)
- David I Bellovin
- Department of Medicine and Pathology, Stanford University School of Medicine, Stanford, CA 94305-5151, USA
| | | | | |
Collapse
|
29
|
|
30
|
Furth PA. Cancer prevention as biomodulation: targeting the initiating stimulus and secondary adaptations. Ann N Y Acad Sci 2013; 1271:1-9. [PMID: 23050958 PMCID: PMC3471382 DOI: 10.1111/j.1749-6632.2012.06736.x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
In a medical sense, biomodulation could be considered a biochemical or cellular response to a disease or therapeutic stimulus. In cancer pathophysiology, the initial oncogenic stimulus leads to cellular and biochemical changes that allow cells, tissue, and organism to accommodate and accept the oncogenic insult. In epithelial cell cancer development, the process of carcinogenesis is frequently characterized by sequential cellular and biochemical adaptations as cells transition through hyperplasia, dysplasia, atypical dysplasia, carcinoma in situ, and invasive cancer. In some cases, the adaptations may persist after the initial oncogenic stimulus is gone in a type of “hit-and-run” oncogenesis. These pathophysiological changes may interfere with cancer prevention therapies targeted solely to the initial oncogenic insult, perhaps contributing to resistance development. Characterization of these accommodating adaptations could provide insight for the development of cancer preventive regimens that might more effectively biomodulate preneoplastic cells toward a more normal state.
Collapse
Affiliation(s)
- Priscilla A Furth
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, USA.
| |
Collapse
|
31
|
Johnson NA, Slack GW, Savage KJ, Connors JM, Ben-Neriah S, Rogic S, Scott DW, Tan KL, Steidl C, Sehn LH, Chan WC, Iqbal J, Meyer PN, Lenz G, Wright G, Rimsza LM, Valentino C, Brunhoeber P, Grogan TM, Braziel RM, Cook JR, Tubbs RR, Weisenburger DD, Campo E, Rosenwald A, Ott G, Delabie J, Holcroft C, Jaffe ES, Staudt LM, Gascoyne RD. Concurrent expression of MYC and BCL2 in diffuse large B-cell lymphoma treated with rituximab plus cyclophosphamide, doxorubicin, vincristine, and prednisone. J Clin Oncol 2012; 30:3452-9. [PMID: 22851565 DOI: 10.1200/jco.2011.41.0985] [Citation(s) in RCA: 702] [Impact Index Per Article: 58.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
PURPOSE Diffuse large B-cell lymphoma (DLBCL) is curable in 60% of patients treated with rituximab plus cyclophosphamide, doxorubicin, vincristine, and prednisone (R-CHOP). MYC translocations, with or without BCL2 translocations, have been associated with inferior survival in DLBCL. We investigated whether expression of MYC protein, with or without BCL2 protein expression, could risk-stratify patients at diagnosis. PATIENTS AND METHODS We determined the correlation between presence of MYC and BCL2 proteins by immunohistochemistry (IHC) with survival in two independent cohorts of patients with DLBCL treated with R-CHOP. We further determined if MYC protein expression correlated with high MYC mRNA and/or presence of MYC translocation. RESULTS In the training cohort (n = 167), MYC and BCL2 proteins were detected in 29% and 44% of patients, respectively. Concurrent expression (MYC positive/BCL2 positive) was present in 21% of patients. MYC protein correlated with presence of high MYC mRNA and MYC translocation (both P < .001), but the latter was less frequent (both 11%). MYC protein expression was only associated with inferior overall and progression-free survival when BCL2 protein was coexpressed (P < .001). Importantly, the poor prognostic effect of MYC positive/BCL2 positive was validated in an independent cohort of 140 patients with DLBCL and remained significant (P < .05) after adjusting for presence of high-risk features in a multivariable model that included elevated international prognostic index score, activated B-cell molecular subtype, and presence of concurrent MYC and BCL2 translocations. CONCLUSION Assessment of MYC and BCL2 expression by IHC represents a robust, rapid, and inexpensive approach to risk-stratify patients with DLBCL at diagnosis.
Collapse
|
32
|
Abstract
National Cancer Institute has announced 24 provocative questions on cancer. Here I try to answer some of them by linking the dots of existing knowledge.
Collapse
Affiliation(s)
- Mikhail V Blagosklonny
- Department of Cell Stress Biology, Roswell Park Cancer Institute, BLSC, L3-312, Elm and Carlton Streets, Buffalo, NY 14263, USA.
| |
Collapse
|