1
|
Shen J, He Y, Li S, Chen H. Crosstalk of methylation and tamoxifen in breast cancer (Review). Mol Med Rep 2024; 30:180. [PMID: 39129315 PMCID: PMC11338244 DOI: 10.3892/mmr.2024.13304] [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: 06/02/2024] [Accepted: 07/23/2024] [Indexed: 08/13/2024] Open
Abstract
Tamoxifen is a widely used anti‑estrogen drug in the endocrine therapy of breast cancer (BC). It blocks estrogen signaling by competitively binding to estrogen receptor α (ERα), thereby inhibiting the growth of BC cells. However, with the long‑term application of tamoxifen, a subset of patients with BC have shown resistance to tamoxifen, which leads to low overall survival and progression‑free survival. The molecular mechanism of resistance is mainly due to downregulation of ERα expression and abnormal activation of the PI3K/AKT/mTOR signaling pathway. Moreover, the downregulation of targeted gene expression mediated by DNA methylation is an important regulatory mode to control protein expression. In the present review, methylation and tamoxifen are briefly introduced, followed by a focus on the effect of methylation on tamoxifen resistance and sensitivity. Finally, the clinical application of methylation for tamoxifen is described, including its use as a prognostic indicator. Finally, it is hypothesized that when methylation is used in combination with tamoxifen, it could recover the resistance of tamoxifen.
Collapse
Affiliation(s)
- Jin Shen
- Department of Rehabilitation, The Affiliated Zhuzhou Hospital of Xiangya Medical College, Central South University, Zhuzhou, Hunan 412000, P.R. China
| | - Yan He
- Department of Neurology, The Affiliated Zhuzhou Hospital of Xiangya Medical College, Central South University, Zhuzhou, Hunan 412000, P.R. China
| | - Shengpeng Li
- Department of Rehabilitation, The Affiliated Zhuzhou Hospital of Xiangya Medical College, Central South University, Zhuzhou, Hunan 412000, P.R. China
| | - Huimin Chen
- Department of Rehabilitation, The Affiliated Zhuzhou Hospital of Xiangya Medical College, Central South University, Zhuzhou, Hunan 412000, P.R. China
| |
Collapse
|
2
|
Yu S, Wu R, Si Y, Fan Z, Wang Y, Yao C, Sun R, Xue Y, Chen Y, Wang Z, Dong S, Wang N, Ling X, Liang Z, Bi C, Yang Y, Dong W, Sun H. Alternative splicing of ALDOA confers tamoxifen resistance in breast cancer. Oncogene 2024; 43:2901-2913. [PMID: 39164523 DOI: 10.1038/s41388-024-03134-w] [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: 02/08/2024] [Revised: 08/09/2024] [Accepted: 08/12/2024] [Indexed: 08/22/2024]
Abstract
The cancer-associated alternative splicing (AS) events generate cancer-related transcripts which are involved in uncontrolled cell proliferation and drug resistance. However, the key AS variants implicated in tamoxifen (TAM) resistance in breast cancer remain elusive. In the current study, we investigated the landscape of AS events in nine pairs of primary and relapse breast tumors from patients receiving TAM-based therapy. We unrevealed a notable association between the inclusion of exon 7.2 in the 5'untranslated region (5'UTR) of ALDOA mRNA and TAM resistance. Mechanistically, the inclusion of ALDOA exon 7.2 enhances the translation efficiency of the transcript, resulting in increased ALDOA protein expression, mTOR pathway activity, and the promotion of TAM resistance in breast cancer cells. Moreover, the inclusion of exon 7.2 in ALDOA mRNA is mediated by MSI1 via direct interaction. In addition, elevated inclusion of ALDOA exon 7.2 or expression of MSI1 is associated with an unfavorable prognosis in patients undergoing endocrine therapy. Notably, treatment with Aldometanib, an ALDOA inhibitor, effectively restrains the growth of TAM-resistant breast cancer cells in vitro and in vivo. The present study unveils the pivotal role of an AS event in ALDOA, under the regulation of MSI1, in driving TAM resistance in breast cancer. Therefore, this study provides a promising therapeutic avenue targeting ALDOA to combat TAM resistance.
Collapse
Affiliation(s)
- Shiyi Yu
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China
- Jiangsu Key Laboratory of Experimental & Translational Non-Coding RNA Research, Yangzhou University, Yangzhou, China
| | - Rui Wu
- School of Life Science, Liaoning Normal University, Dalian, China
| | - Yue Si
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China
- Jiangsu Key Laboratory of Experimental & Translational Non-Coding RNA Research, Yangzhou University, Yangzhou, China
| | - Zhehao Fan
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China
- Jiangsu Key Laboratory of Experimental & Translational Non-Coding RNA Research, Yangzhou University, Yangzhou, China
| | - Ying Wang
- Department of Thyroid and Breast Surgery, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China
| | - Chang Yao
- The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Rongmao Sun
- Department of Thyroid and Breast Surgery, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China
| | - Yaji Xue
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China
- Jiangsu Key Laboratory of Experimental & Translational Non-Coding RNA Research, Yangzhou University, Yangzhou, China
| | - Yongli Chen
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China
- Jiangsu Key Laboratory of Experimental & Translational Non-Coding RNA Research, Yangzhou University, Yangzhou, China
| | - Zheng Wang
- Department of Pathology, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China
| | - Shuangshuang Dong
- Department of Pathology, Northern Jiangsu People's Hospital Affiliated to Yangzhou University/Clinical Medical College, Yangzhou University, Yangzhou, China
| | - Ning Wang
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China
- Jiangsu Key Laboratory of Experimental & Translational Non-Coding RNA Research, Yangzhou University, Yangzhou, China
| | - Xinyue Ling
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China
- Jiangsu Key Laboratory of Experimental & Translational Non-Coding RNA Research, Yangzhou University, Yangzhou, China
| | - Zhengyan Liang
- School of Basic Medical Science, Guangdong Medical University, Dongguan, China
| | - Caili Bi
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China
- Jiangsu Key Laboratory of Experimental & Translational Non-Coding RNA Research, Yangzhou University, Yangzhou, China
| | - Yi Yang
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China.
- Jiangsu Key Laboratory of Experimental & Translational Non-Coding RNA Research, Yangzhou University, Yangzhou, China.
| | - Weibing Dong
- School of Life Science, Liaoning Normal University, Dalian, China.
| | - Haibo Sun
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China.
- Jiangsu Key Laboratory of Experimental & Translational Non-Coding RNA Research, Yangzhou University, Yangzhou, China.
- Haian Hospital of Traditional Chinese Medicine, Haian, China.
| |
Collapse
|
3
|
Walker NM, Ibuki Y, McLinden AP, Misumi K, Mitchell DC, Kleer GG, Lock AM, Vittal R, Sonenberg N, Garner AL, Lama VN. MNK-driven eIF4E phosphorylation regulates the fibrogenic transformation of mesenchymal cells and chronic lung allograft dysfunction. J Clin Invest 2024; 134:e168393. [PMID: 39145446 PMCID: PMC11324311 DOI: 10.1172/jci168393] [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: 01/09/2023] [Accepted: 06/25/2024] [Indexed: 08/16/2024] Open
Abstract
Tissue fibrosis remains unamenable to meaningful therapeutic interventions and is the primary cause of chronic graft failure after organ transplantation. Eukaryotic translation initiation factor (eIF4E), a key translational regulator, serves as convergent target of multiple upstream profibrotic signaling pathways that contribute to mesenchymal cell (MC) activation. Here, we investigate the role of MAP kinase-interacting serine/threonine kinase-induced (MNK-induced) direct phosphorylation of eIF4E at serine 209 (Ser209) in maintaining fibrotic transformation of MCs and determine the contribution of the MNK/eIF4E pathway to the pathogenesis of chronic lung allograft dysfunction (CLAD). MCs from patients with CLAD demonstrated constitutively higher eIF4E phosphorylation at Ser209, and eIF4E phospho-Ser209 was found to be critical in regulating key fibrogenic protein autotaxin, leading to sustained β-catenin activation and profibrotic functions of CLAD MCs. MNK1 signaling was upregulated in CLAD MCs, and genetic or pharmacologic targeting of MNK1 activity inhibited eIF4E phospho-Ser209 and profibrotic functions of CLAD MCs in vitro. Treatment with an MNK1/2 inhibitor (eFT-508) abrogated allograft fibrosis in an orthotopic murine lung-transplant model. Together these studies identify what we believe is a previously unrecognized MNK/eIF4E/ATX/β-catenin signaling pathway of fibrotic transformation of MCs and present the first evidence, to our knowledge, for the utility of MNK inhibitors in fibrosis.
Collapse
Affiliation(s)
- Natalie M. Walker
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Yuta Ibuki
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - A. Patrick McLinden
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Keizo Misumi
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Dylan C. Mitchell
- Department of Medicinal Chemistry, University of Michigan, Ann Arbor, Michigan, USA
| | - Gabriel G. Kleer
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Alison M. Lock
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Ragini Vittal
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Nahum Sonenberg
- Department of Biochemistry and McGill Cancer Center, McGill University, Montreal, Quebec, Canada
| | - Amanda L. Garner
- Department of Medicinal Chemistry, University of Michigan, Ann Arbor, Michigan, USA
| | - Vibha N. Lama
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| |
Collapse
|
4
|
Fard SS, Holz MK. Regulation of mRNA translation by estrogen receptor in breast cancer. Steroids 2023; 200:109316. [PMID: 37806603 PMCID: PMC10841406 DOI: 10.1016/j.steroids.2023.109316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 09/15/2023] [Accepted: 09/28/2023] [Indexed: 10/10/2023]
Abstract
Breast cancer is one of the leading causes of cancer-related fatalities and the most often diagnosed malignancy in women globally. Dysregulation of sex hormone signaling pathways mediated by the estrogen receptor (ER) in breast cancer is well characterized. Although ER is known to promote cell growth and survival by altering gene transcription, recent research suggests that its effects in cancers are also mediated through dysregulation of protein synthesis. This implies that ER can coordinately affect gene expression through both translational and transcriptional pathways, leading to the development of malignancy. In this review, we will cover the current understanding of how the ER controls mRNA translation in breast cancer and discuss any potential clinical implications of this phenomenon.
Collapse
Affiliation(s)
- Shahrzad S Fard
- Department of Cell Biology and Anatomy, Graduate School of Biomedical Sciences, New York Medical College, Valhalla, NY, USA
| | - Marina K Holz
- Department of Cell Biology and Anatomy, Graduate School of Biomedical Sciences, New York Medical College, Valhalla, NY, USA; Department of Biochemistry and Molecular Biology, Graduate School of Biomedical Sciences, New York Medical College, Valhalla, NY, USA.
| |
Collapse
|
5
|
Meng D, Zhao X, Yang YC, Navickas A, Helland C, Goodarzi H, Singh M, Bandyopadhyay S. A bi-steric mTORC1-selective inhibitor overcomes drug resistance in breast cancer. Oncogene 2023:10.1038/s41388-023-02737-z. [PMID: 37264081 DOI: 10.1038/s41388-023-02737-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 04/30/2023] [Accepted: 05/18/2023] [Indexed: 06/03/2023]
Abstract
Activation of the PI3K-mTOR pathway is central to breast cancer pathogenesis including resistance to many targeted therapies. The mTOR kinase forms two distinct complexes, mTORC1 and mTORC2, and understanding which is required for the survival of malignant cells has been limited by tools to selectively and completely impair either subcomplex. To address this, we used RMC-6272, a bi-steric molecule with a rapamycin-like moiety linked to an mTOR active-site inhibitor that displays >25-fold selectivity for mTORC1 over mTORC2 substrates. Complete suppression of mTORC1 by RMC-6272 causes apoptosis in ER+/HER2- breast cancer cell lines, particularly in those that harbor mutations in PIK3CA or PTEN, due to inhibition of the rapamycin resistant, mTORC1 substrate 4EBP1 and reduction of the pro-survival protein MCL1. RMC-6272 reduced translation of ribosomal mRNAs, MYC target genes, and components of the CDK4/6 pathway, suggesting enhanced impairment of oncogenic pathways compared to the partial mTORC1 inhibitor everolimus. RMC-6272 maintained efficacy in hormone therapy-resistant acquired cell lines and patient-derived xenografts (PDX), showed increased efficacy in CDK4/6 inhibitor treated acquired resistant cell lines versus their parental counterparts, and was efficacious in a PDX from a patient experiencing resistance to CDK4/6 inhibition. Bi-steric mTORC1-selective inhibition may be effective in overcoming multiple forms of therapy-resistance in ER+ breast cancers.
Collapse
Affiliation(s)
- Delong Meng
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, CA, USA
| | - Xin Zhao
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, CA, USA
| | - Yu Chi Yang
- Department of Biology, Revolution Medicines Inc., Redwood City, CA, USA
| | - Albertas Navickas
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA, 94158, USA
- Department of Urology, University of California, San Francisco, San Francisco, CA, USA
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA
- Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, CA, USA
- Institut Curie, PSL Research University, CNRS UMR3348, INSERM U1278, Orsay, France
| | - Ciara Helland
- Department of Biology, Revolution Medicines Inc., Redwood City, CA, USA
| | - Hani Goodarzi
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA, 94158, USA
- Department of Urology, University of California, San Francisco, San Francisco, CA, USA
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA
- Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, CA, USA
| | - Mallika Singh
- Department of Biology, Revolution Medicines Inc., Redwood City, CA, USA
| | - Sourav Bandyopadhyay
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, CA, USA.
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA.
- Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
6
|
Hany D, Vafeiadou V, Picard D. CRISPR-Cas9 screen reveals a role of purine synthesis for estrogen receptor α activity and tamoxifen resistance of breast cancer cells. SCIENCE ADVANCES 2023; 9:eadd3685. [PMID: 37172090 PMCID: PMC10181187 DOI: 10.1126/sciadv.add3685] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/14/2023]
Abstract
In breast cancer, resistance to endocrine therapies that target estrogen receptor α (ERα), such as tamoxifen and fulvestrant, remains a major clinical problem. Whether and how ERα+ breast cancers switch from being estrogen-dependent to estrogen-independent remains unclear. With a genome-wide CRISPR-Cas9 knockout screen, we identified previously unknown biomarkers and potential therapeutic targets of endocrine resistance. We demonstrate that high levels of PAICS, an enzyme involved in the de novo biosynthesis of purines, can shift the balance of ERα activity to be more estrogen-independent and tamoxifen-resistant. We find that this may be due to elevated activities of cAMP-activated protein kinase A and mTOR, kinases known to phosphorylate ERα specifically and to stimulate its activity. Genetic or pharmacological targeting of PAICS sensitizes tamoxifen-resistant cells to tamoxifen. Addition of purines renders them more resistant. On the basis of these findings, we propose the combined targeting of PAICS and ERα as a new, effective, and potentially safe therapeutic regimen.
Collapse
Affiliation(s)
- Dina Hany
- Département de Biologie Moléculaire et Cellulaire, Université de Genève, Sciences III, Quai Ernest-Ansermet 30, CH - 1211 Genève 4, Switzerland
- On leave from: Department of Pharmacology and Therapeutics Faculty of Pharmacy, Pharos University in Alexandria, Alexandria 21311, Egypt
| | - Vasiliki Vafeiadou
- Département de Biologie Moléculaire et Cellulaire, Université de Genève, Sciences III, Quai Ernest-Ansermet 30, CH - 1211 Genève 4, Switzerland
| | - Didier Picard
- Département de Biologie Moléculaire et Cellulaire, Université de Genève, Sciences III, Quai Ernest-Ansermet 30, CH - 1211 Genève 4, Switzerland
| |
Collapse
|
7
|
Si W, Kan C, Zhang L, Li F. Role of RUNX2 in breast cancer development and drug resistance (Review). Oncol Lett 2023; 25:176. [PMID: 37033103 PMCID: PMC10079821 DOI: 10.3892/ol.2023.13762] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Accepted: 03/02/2023] [Indexed: 03/17/2023] Open
Abstract
Breast cancer is the most common malignancy and ranks second among the causes of tumor-associated death in females. The recurrence and drug resistance of breast cancer are intractable due to the presence of breast cancer stem cells (BCSCs), which are adequate to initiate tumor formation and refractory to conventional remedies. Runt-related transcription factor 2 (RUNX2), a pivotal transcription factor in mammary gland and bone development, has also been related to metastatic cancer and BCSCs. State-of-the-art research has indicated the retention of RUNX2 expression in a more invasive subtype of breast cancer, and in particular, triple-negative breast cancer development and drug resistance are associated with estrogen receptor signaling pathways. The present review mainly focused on the latest updates on RUNX2 in BCSCs and their roles in breast cancer progression and drug resistance, providing insight that may aid the development of RUNX2-based diagnostics and treatments for breast cancer in clinical practice.
Collapse
Affiliation(s)
- Wentao Si
- Department of Pathophysiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Chen Kan
- Department of Pathophysiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Leisheng Zhang
- Key Laboratory of Molecular Diagnostics and Precision Medicine for Surgical Oncology in Gansu Province and NHC Key Laboratory of Diagnosis and Therapy of Gastrointestinal Tumor, Gansu Provincial Hospital, Lanzhou, Gansu 730000, P.R. China
- Key Laboratory of Radiation Technology and Biophysics, Hefei Institute of Physical Science, Chinese Academy of Sciences, Hefei, Anhui 230031, P.R. China
| | - Feifei Li
- Department of Pathophysiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| |
Collapse
|
8
|
Prabhu SA, Moussa O, Gonçalves C, LaPierre JH, Chou H, Huang F, Richard VR, Ferruzo PYM, Guettler EM, Soria-Bretones I, Kirby L, Gagnon N, Su J, Silvester J, Krisna SS, Rose AAN, Sheppard KE, Cescon DW, Mallette FA, Zahedi RP, Borchers CH, Del Rincon SV, Miller WH. Inhibition of the MNK1/2-eIF4E Axis Augments Palbociclib-Mediated Antitumor Activity in Melanoma and Breast Cancer. Mol Cancer Ther 2023; 22:192-204. [PMID: 36722142 DOI: 10.1158/1535-7163.mct-22-0092] [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: 02/08/2022] [Revised: 10/12/2022] [Accepted: 12/08/2022] [Indexed: 02/02/2023]
Abstract
Aberrant cell-cycle progression is characteristic of melanoma, and CDK4/6 inhibitors, such as palbociclib, are currently being tested for efficacy in this disease. Despite the promising nature of CDK4/6 inhibitors, their use as single agents in melanoma has shown limited clinical benefit. Herein, we discovered that treatment of tumor cells with palbociclib induces the phosphorylation of the mRNA translation initiation factor eIF4E. When phosphorylated, eIF4E specifically engenders the translation of mRNAs that code for proteins involved in cell survival. We hypothesized that cancer cells treated with palbociclib use upregulated phosphorylated eIF4E (phospho-eIF4E) to escape the antitumor benefits of this drug. Indeed, we found that pharmacologic or genetic disruption of MNK1/2 activity, the only known kinases for eIF4E, enhanced the ability of palbociclib to decrease clonogenic outgrowth. Moreover, a quantitative proteomics analysis of melanoma cells treated with combined MNK1/2 and CDK4/6 inhibitors showed downregulation of proteins with critical roles in cell-cycle progression and mitosis, including AURKB, TPX2, and survivin. We also observed that palbociclib-resistant breast cancer cells have higher basal levels of phospho-eIF4E, and that treatment with MNK1/2 inhibitors sensitized these palbociclib-resistant cells to CDK4/6 inhibition. In vivo we demonstrate that the combination of MNK1/2 and CDK4/6 inhibition significantly increases the overall survival of mice compared with either monotherapy. Overall, our data support MNK1/2 inhibitors as promising drugs to potentiate the antineoplastic effects of palbociclib and overcome therapy-resistant disease.
Collapse
Affiliation(s)
- Sathyen A Prabhu
- Lady Davis Institute, Jewish General Hospital, Montréal, Québec, Canada
- Division of Experimental Medicine, McGill University, Montréal, Québec, Canada
| | - Omar Moussa
- Lady Davis Institute, Jewish General Hospital, Montréal, Québec, Canada
- Division of Experimental Medicine, McGill University, Montréal, Québec, Canada
| | | | - Judith H LaPierre
- Lady Davis Institute, Jewish General Hospital, Montréal, Québec, Canada
- Division of Experimental Medicine, McGill University, Montréal, Québec, Canada
| | - Hsiang Chou
- Lady Davis Institute, Jewish General Hospital, Montréal, Québec, Canada
| | - Fan Huang
- Lady Davis Institute, Jewish General Hospital, Montréal, Québec, Canada
- Division of Experimental Medicine, McGill University, Montréal, Québec, Canada
| | - Vincent R Richard
- Segal Cancer Proteomics Centre, Lady Davis Institute, Jewish General Hospital, McGill University, Montréal, Québec, Canada
| | - Pault Y M Ferruzo
- Centre de recherche de l'Hôpital Maisonneuve-Rosemont, Montréal, Québec, Canada
- Department of Biochemistry and Molecular Medicine, Université de Montréal, Montréal, Québec, Canada
| | | | - Isabel Soria-Bretones
- Lady Davis Institute, Jewish General Hospital, Montréal, Québec, Canada
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Laura Kirby
- Research Division, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Natascha Gagnon
- Lady Davis Institute, Jewish General Hospital, Montréal, Québec, Canada
| | - Jie Su
- Lady Davis Institute, Jewish General Hospital, Montréal, Québec, Canada
| | - Jennifer Silvester
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | | | - April A N Rose
- Lady Davis Institute, Jewish General Hospital, Montréal, Québec, Canada
- Division of Experimental Medicine, McGill University, Montréal, Québec, Canada
- Gerald Bronfman Department of Oncology, McGill University, Montréal, Québec, Canada
| | - Karen E Sheppard
- Research Division, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Department of Biochemistry and Molecular Biology, University of Melbourne, Parkville, Victoria, Australia
| | - David W Cescon
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
- University of Toronto, Toronto, Ontario, Canada
| | - Frédérick A Mallette
- Centre de recherche de l'Hôpital Maisonneuve-Rosemont, Montréal, Québec, Canada
- Department of Biochemistry and Molecular Medicine, Université de Montréal, Montréal, Québec, Canada
- Department of Medicine, Université de Montréal, Montréal, Québec, Canada
| | - Rene P Zahedi
- Segal Cancer Proteomics Centre, Lady Davis Institute, Jewish General Hospital, McGill University, Montréal, Québec, Canada
- McGill Centre for Translational Research in Cancer, McGill University, Montréal, Québec, Canada
| | - Christoph H Borchers
- Segal Cancer Proteomics Centre, Lady Davis Institute, Jewish General Hospital, McGill University, Montréal, Québec, Canada
- Gerald Bronfman Department of Oncology, McGill University, Montréal, Québec, Canada
- McGill Centre for Translational Research in Cancer, McGill University, Montréal, Québec, Canada
| | - Sonia V Del Rincon
- Lady Davis Institute, Jewish General Hospital, Montréal, Québec, Canada
- Division of Experimental Medicine, McGill University, Montréal, Québec, Canada
- McGill Centre for Translational Research in Cancer, McGill University, Montréal, Québec, Canada
| | - Wilson H Miller
- Lady Davis Institute, Jewish General Hospital, Montréal, Québec, Canada
- Division of Experimental Medicine, McGill University, Montréal, Québec, Canada
- McGill Centre for Translational Research in Cancer, McGill University, Montréal, Québec, Canada
| |
Collapse
|
9
|
Malash I, Mansour O, Gaafar R, Shaarawy S, Abdellateif MS, Ahmed OS, Zekri ARN, Bahnassy A. Her2/EGFR-PDGFR pathway aberrations associated with tamoxifen response in metastatic breast cancer patients. J Egypt Natl Canc Inst 2022; 34:31. [DOI: 10.1186/s43046-022-00132-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 06/14/2022] [Indexed: 12/24/2022] Open
Abstract
Abstract
Background
Metastatic breast cancer (MBC) is a major health problem worldwide. Some patients improve on tamoxifen and others do not respond to treatment. Therefore, the aim of the current study is to assess genetic aberrations in the Her2/EGFR-PDGFR pathway associated with tamoxifen response in MBC patients.
Methods
This is a retrospective cohort study, including 157 hormone receptors positive, locally recurrent inoperable and/or MBC patients on tamoxifen treatment. Patients were categorized into 78 (49.7%) tamoxifen responders and 79 (50.3%) tamoxifen non-responder patients. Genetic aberrations of 84 genes involved in the Her2/EGFR-PDGFR pathway were assessed in the tumor tissue samples obtained from the patients using SA-Bioscience assay. The identified panel was correlated to patients’ response to treatment, to detect the differentially expressed genes in tamoxifen responders and non-responders.
Results
One hundred twenty-three (78.3%) patients were estrogen receptor (ER) and progesterone receptor (PR) positive, 108 (68.8%) were ER only positive, and 78 (49.7%) were PR only positive. There were 56 genes overexpressed in the refractory group compared to responders. However, only five out of these 56 genes, Janus kinase 1 (JAK1), collagen type I alpha 1 (COL1A1), GRB2-associated binding protein 1 (GAB1), fibronectin-1 (FN1), and MAP kinase-interacting serine/threonine-protein kinase (MKNK1), showed statistical significance between the two groups. Patients with bone metastasis showed a better response to treatment compared to those with metastatic deposits in other sites such as visceral metastasis (P < 0.005).
Conclusions
Genetic profiling using simple quantitative real-time polymerase chain reaction (qRT-PCR) protocols could be used to assess response to tamoxifen treatment in MBC patients. According to our data, a five-gene panel in the EGFR pathway (JAK1, COL1A1, GAB1, FN1 and MKNK1) could be used to categorize MBC patients into groups according to treatment response.
Collapse
|
10
|
Rubio A, Garland GD, Sfakianos A, Harvey RF, Willis AE. Aberrant protein synthesis and cancer development: The role of canonical eukaryotic initiation, elongation and termination factors in tumorigenesis. Semin Cancer Biol 2022; 86:151-165. [PMID: 35487398 DOI: 10.1016/j.semcancer.2022.04.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 04/11/2022] [Accepted: 04/20/2022] [Indexed: 01/27/2023]
Abstract
In tumourigenesis, oncogenes or dysregulated tumour suppressor genes alter the canonical translation machinery leading to a reprogramming of the translatome that, in turn, promotes the translation of selected mRNAs encoding proteins involved in proliferation and metastasis. It is therefore unsurprising that abnormal expression levels and activities of eukaryotic initiation factors (eIFs), elongation factors (eEFs) or termination factors (eRFs) are associated with poor outcome for patients with a wide range of cancers. In this review we discuss how RNA binding proteins (RBPs) within the canonical translation factor machinery are dysregulated in cancers and how targeting such proteins is leading to new therapeutic avenues.
Collapse
Affiliation(s)
- Angela Rubio
- MRC Toxicology Unit, University of Cambridge, Gleeson Building, Tennis Court Rd, Cambridge CB2 1QR, UK
| | - Gavin D Garland
- MRC Toxicology Unit, University of Cambridge, Gleeson Building, Tennis Court Rd, Cambridge CB2 1QR, UK
| | - Aristeidis Sfakianos
- MRC Toxicology Unit, University of Cambridge, Gleeson Building, Tennis Court Rd, Cambridge CB2 1QR, UK
| | - Robert F Harvey
- MRC Toxicology Unit, University of Cambridge, Gleeson Building, Tennis Court Rd, Cambridge CB2 1QR, UK
| | - Anne E Willis
- MRC Toxicology Unit, University of Cambridge, Gleeson Building, Tennis Court Rd, Cambridge CB2 1QR, UK.
| |
Collapse
|
11
|
Park HJ, Gregory MA, Zaberezhnyy V, Goodspeed A, Jordan CT, Kieft JS, DeGregori J. Therapeutic resistance in acute myeloid leukemia cells is mediated by a novel ATM/mTOR pathway regulating oxidative phosphorylation. eLife 2022; 11:e79940. [PMID: 36259537 PMCID: PMC9645811 DOI: 10.7554/elife.79940] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Accepted: 10/17/2022] [Indexed: 11/13/2022] Open
Abstract
While leukemic cells are susceptible to various therapeutic insults, residence in the bone marrow microenvironment typically confers protection from a wide range of drugs. Thus, understanding the unique molecular changes elicited by the marrow is of critical importance toward improving therapeutic outcomes. In this study, we demonstrate that aberrant activation of oxidative phosphorylation serves to induce therapeutic resistance in FLT3 mutant human AML cells challenged with FLT3 inhibitor drugs. Importantly, our findings show that AML cells are protected from apoptosis following FLT3 inhibition due to marrow-mediated activation of ATM, which in turn upregulates oxidative phosphorylation via mTOR signaling. mTOR is required for the bone marrow stroma-dependent maintenance of protein translation, with selective polysome enrichment of oxidative phosphorylation transcripts, despite FLT3 inhibition. To investigate the therapeutic significance of this finding, we tested the mTOR inhibitor everolimus in combination with the FLT3 inhibitor quizartinib in primary human AML xenograft models. While marrow resident AML cells were highly resistant to quizartinib alone, the addition of everolimus induced profound reduction in tumor burden and prevented relapse. Taken together, these data provide a novel mechanistic understanding of marrow-based therapeutic resistance and a promising strategy for improved treatment of FLT3 mutant AML patients.
Collapse
Affiliation(s)
- Hae J Park
- Department of Biochemistry and Molecular Genetics, School of Medicine, University of Colorado Anschutz Medical CampusAuroraUnited States
- Medical Scientist Training Program, University of Colorado Anschutz Medical CampusAuroraUnited States
| | - Mark A Gregory
- Department of Biochemistry and Molecular Genetics, School of Medicine, University of Colorado Anschutz Medical CampusAuroraUnited States
| | - Vadym Zaberezhnyy
- Department of Biochemistry and Molecular Genetics, School of Medicine, University of Colorado Anschutz Medical CampusAuroraUnited States
| | - Andrew Goodspeed
- Department of Pharmacology, University of Colorado Anschutz Medical CampusAuroraUnited States
- University of Colorado Comprehensive Cancer Center, University of Colorado Anschutz Medical CampusAuroraUnited States
| | - Craig T Jordan
- Department of Medicine, Section of Hematology, University of Colorado Anschutz Medical CampusAuroraUnited States
| | - Jeffrey S Kieft
- Department of Biochemistry and Molecular Genetics, School of Medicine, University of Colorado Anschutz Medical CampusAuroraUnited States
| | - James DeGregori
- Department of Biochemistry and Molecular Genetics, School of Medicine, University of Colorado Anschutz Medical CampusAuroraUnited States
- University of Colorado Comprehensive Cancer Center, University of Colorado Anschutz Medical CampusAuroraUnited States
- Department of Medicine, Section of Hematology, University of Colorado Anschutz Medical CampusAuroraUnited States
| |
Collapse
|
12
|
Abstract
Signaling via extracellular regulated kinase 1/2 (ERK1/2) and p90 ribosomal S6 kinase (RSK), a downstream effector, mediates numerous processes. For example, ERK1/2-RSK signaling is essential for estrogen homeostasis in the mammary gland and uterus to maintain physiological responsiveness. This review will focus on the coordination of ERK1/2-RSK2 and estrogen signaling through estrogen receptor alpha (ERα). The interrelationship and the feedback mechanisms between these pathways occurs at the level of transcription, translation, and posttranslational modification. Identifying how ERK1/2-RSK2 and estrogen signaling cooperate in homeostasis and disease may lead to novel therapeutic approaches in estrogen-dependent disorders.
Collapse
Affiliation(s)
- Deborah A Lannigan
- Correspondence: Deborah A. Lannigan, PhD, Department of Pathology, Microbiology & Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA.
| |
Collapse
|
13
|
Muñoz-Ayala A, Chimal-Vega B, García-González V. Translation initiation and its relationship with metabolic mechanisms in cancer development, progression and chemoresistance. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2022; 132:111-141. [PMID: 36088073 DOI: 10.1016/bs.apcsb.2022.05.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Pathways that regulate protein homeostasis (proteostasis) in cells range from mRNA processing to protein degradation; perturbations in regulatory mechanisms of these pathways can lead to oncogenic cellular processes. Protein synthesis modulation failures are common phenomena in cancer cells, wherein specific conditions that promote the translation of protein factors promoting carcinogenesis are present. These specific conditions may be favored by metabolic lipid alterations like those found in metabolic syndrome and obesity. Protein translation modifications have been described in obesity, favoring the translation of protein targets that benefit lipid accumulation; a determining factor is the activity of the cap-binding eukaryotic translation initiation factor 4E (eIF4E), a crosstalk in protein translation and lipogenesis. Besides, alterations of protein translation initiation steps are critical participants for the development of both pathogenic conditions, cancer, and obesity. This chapter is focused on the regulation of recognition and processing of carcinogenic-mRNA and the connections among lipid metabolism and cell signaling pathways that promote oncogenesis, tumoral microenvironment generation and potentially the development of chemoresistance. We performed an in-depth analysis of events, such as those occurring in obesity and dyslipidemias, that may influence protein translation, driving the recognition of certain mRNAs and favoring cancer development and chemoresistance.
Collapse
Affiliation(s)
- Andrea Muñoz-Ayala
- Departamento de Bioquímica, Facultad de Medicina Mexicali, Universidad Autónoma de Baja California, Mexicali, México; Laboratorio Multidisciplinario de Estudios Metabólicos y Cáncer, Universidad Autónoma de Baja California, Mexicali, México
| | - Brenda Chimal-Vega
- Departamento de Bioquímica, Facultad de Medicina Mexicali, Universidad Autónoma de Baja California, Mexicali, México; Laboratorio Multidisciplinario de Estudios Metabólicos y Cáncer, Universidad Autónoma de Baja California, Mexicali, México
| | - Victor García-González
- Departamento de Bioquímica, Facultad de Medicina Mexicali, Universidad Autónoma de Baja California, Mexicali, México; Laboratorio Multidisciplinario de Estudios Metabólicos y Cáncer, Universidad Autónoma de Baja California, Mexicali, México.
| |
Collapse
|
14
|
Design, Synthesis and Evaluation of Novel Phorbazole C Derivatives as MNK Inhibitors through Virtual High-Throughput Screening. Mar Drugs 2022; 20:md20070429. [PMID: 35877722 PMCID: PMC9319845 DOI: 10.3390/md20070429] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 06/27/2022] [Accepted: 06/27/2022] [Indexed: 12/04/2022] Open
Abstract
MNKs (mitogen-activated protein kinase-interacting protein kinases) phosphorylate eIF4E at Ser209 to control the translation of certain mRNAs and regulate the process of cell proliferation, cell migration and invasion, etc. Development of MNK inhibitors would be an effective treatment for related diseases. We used the MarineChem3D database to identify hit compounds targeting the protein MNK1 and MNK2 through high-throughput screening. Compounds from the phorbazole family showed good interactions with MNK1, and phorbazole C was selected as our hit compound. By analyzing the binding mode, we designed and synthesized 29 derivatives and evaluated their activity against MNKs, of which, six compounds showed good inhibition to MNKs. We also confirmed three interactions between this kind of compound and MNK1, which are vital for the activity. In conclusion, we report series of novel MNK inhibitors inspired from marine natural products and their relative structure–activity relationship. This will provide important information for further developing MNK inhibitors based on this kind of structure.
Collapse
|
15
|
Patel PS, Pérez-Baos S, Walters B, Orlen M, Volkova A, Ruggles K, Park CY, Schneider RJ. Translational regulation of TFH cell differentiation and autoimmune pathogenesis. SCIENCE ADVANCES 2022; 8:eabo1782. [PMID: 35749506 PMCID: PMC9232117 DOI: 10.1126/sciadv.abo1782] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Accepted: 05/09/2022] [Indexed: 06/15/2023]
Abstract
Little is known regarding T cell translational regulation. We demonstrate that T follicular helper (TFH) cells use a previously unknown mechanism of selective messenger RNA (mRNA) translation for their differentiation, role in B cell maturation, and in autoimmune pathogenesis. We show that TFH cells have much higher levels of translation factor eIF4E than non-TFH CD4+ T cells, which is essential for translation of TFH cell fate-specification mRNAs. Genome-wide translation studies indicate that modest down-regulation of eIF4E activity by a small-molecule inhibitor or short hairpin RN impairs TFH cell development and function. In mice, down-regulation of eIF4E activity specifically reduces TFH cells among T helper subtypes, germinal centers, B cell recruitment, and antibody production. In experimental autoimmune encephalomyelitis, eIF4E activity down-regulation blocks TFH cell participation in disease pathogenesis while promoting rapid remission and spinal cord remyelination. TFH cell development and its role in autoimmune pathogenesis involve selective mRNA translation that is highly druggable.
Collapse
Affiliation(s)
- Preeyam S. Patel
- Department of Microbiology, New York University School of Medicine, New York, NY 10016, USA
| | - Sandra Pérez-Baos
- Department of Microbiology, New York University School of Medicine, New York, NY 10016, USA
| | - Beth Walters
- Department of Microbiology, New York University School of Medicine, New York, NY 10016, USA
| | - Margo Orlen
- Department of Microbiology, New York University School of Medicine, New York, NY 10016, USA
| | - Angelina Volkova
- Department of Medicine, New York University School of Medicine, New York, NY 10016, USA
| | - Kelly Ruggles
- Department of Medicine, New York University School of Medicine, New York, NY 10016, USA
| | - Christopher Y. Park
- Department of Pathology, New York University School of Medicine, New York, NY 10016, USA
| | - Robert J. Schneider
- Department of Microbiology, New York University School of Medicine, New York, NY 10016, USA
| |
Collapse
|
16
|
Bou-Petit E, Hümmer S, Alarcon H, Slobodnyuk K, Cano-Galietero M, Fuentes P, Guijarro PJ, Muñoz MJ, Suarez-Cabrera L, Santamaria A, Estrada-Tejedor R, Borrell JI, Ramón y Cajal S. Overcoming Paradoxical Kinase Priming by a Novel MNK1 Inhibitor. J Med Chem 2022; 65:6070-6087. [PMID: 35417652 PMCID: PMC9059116 DOI: 10.1021/acs.jmedchem.1c01941] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Indexed: 12/23/2022]
Abstract
Targeting the kinases MNK1 and MNK2 has emerged as a valuable strategy in oncology. However, most of the advanced inhibitors are acting in an adenosine triphosphate (ATP)-competitive mode, precluding the evaluation of different binding modes in preclinical settings. Using rational design, we identified and validated the 4,6-diaryl-pyrazolo[3,4-b]pyridin-3-amine scaffold as the core for MNK inhibitors. Signaling pathway analysis confirmed a direct effect of the hit compound EB1 on MNKs, and in line with the reported function of these kinases, EB1 only affects the growth of tumor but not normal cells. Molecular modeling revealed the binding of EB1 to the inactive conformation of MNK1 and the interaction with the specific DFD motif. This novel mode of action appears to be superior to the ATP-competitive inhibitors, which render the protein in a pseudo-active state. Overcoming this paradoxical activation of MNKs by EB1 represents therefore a promising starting point for the development of a novel generation of MNK inhibitors.
Collapse
Affiliation(s)
- Elisabeth Bou-Petit
- Grup
de Química Farmacèutica, IQS School of Engineering, Universitat Ramon Llull, Via Augusta, 390, 08017 Barcelona, Spain
| | - Stefan Hümmer
- Translational
Molecular Pathology, Vall d’Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona, Psg. Vall d’Hebron 119-129, 08035 Barcelona, Spain
- Spanish
Biomedical Research Network Centre in Oncology (CIBERONC), 28029 Madrid, Spain
| | - Helena Alarcon
- Grup
de Química Farmacèutica, IQS School of Engineering, Universitat Ramon Llull, Via Augusta, 390, 08017 Barcelona, Spain
| | - Konstantin Slobodnyuk
- Translational
Molecular Pathology, Vall d’Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona, Psg. Vall d’Hebron 119-129, 08035 Barcelona, Spain
- Spanish
Biomedical Research Network Centre in Oncology (CIBERONC), 28029 Madrid, Spain
| | - Marta Cano-Galietero
- Translational
Molecular Pathology, Vall d’Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona, Psg. Vall d’Hebron 119-129, 08035 Barcelona, Spain
- Spanish
Biomedical Research Network Centre in Oncology (CIBERONC), 28029 Madrid, Spain
| | - Pedro Fuentes
- Translational
Molecular Pathology, Vall d’Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona, Psg. Vall d’Hebron 119-129, 08035 Barcelona, Spain
- Spanish
Biomedical Research Network Centre in Oncology (CIBERONC), 28029 Madrid, Spain
| | - Pedro J. Guijarro
- Translational
Molecular Pathology, Vall d’Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona, Psg. Vall d’Hebron 119-129, 08035 Barcelona, Spain
| | - María José Muñoz
- Translational
Molecular Pathology, Vall d’Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona, Psg. Vall d’Hebron 119-129, 08035 Barcelona, Spain
- Spanish
Biomedical Research Network Centre in Oncology (CIBERONC), 28029 Madrid, Spain
| | - Leticia Suarez-Cabrera
- Cell
Cycle and Cancer Laboratory, Biomedical Research Group in Urology,
Vall d’Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona, Psg. Vall d’Hebron 119-129, 08035 Barcelona, Spain
| | - Anna Santamaria
- Cell
Cycle and Cancer Laboratory, Biomedical Research Group in Urology,
Vall d’Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona, Psg. Vall d’Hebron 119-129, 08035 Barcelona, Spain
| | - Roger Estrada-Tejedor
- Grup
de Química Farmacèutica, IQS School of Engineering, Universitat Ramon Llull, Via Augusta, 390, 08017 Barcelona, Spain
| | - José I. Borrell
- Grup
de Química Farmacèutica, IQS School of Engineering, Universitat Ramon Llull, Via Augusta, 390, 08017 Barcelona, Spain
| | - Santiago Ramón y Cajal
- Translational
Molecular Pathology, Vall d’Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona, Psg. Vall d’Hebron 119-129, 08035 Barcelona, Spain
- Spanish
Biomedical Research Network Centre in Oncology (CIBERONC), 28029 Madrid, Spain
| |
Collapse
|
17
|
Tsoi H, You CP, Leung MH, Man EPS, Khoo US. Targeting Ribosome Biogenesis to Combat Tamoxifen Resistance in ER+ve Breast Cancer. Cancers (Basel) 2022; 14:1251. [PMID: 35267559 PMCID: PMC8909264 DOI: 10.3390/cancers14051251] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 02/24/2022] [Accepted: 02/27/2022] [Indexed: 02/07/2023] Open
Abstract
Breast cancer is a heterogeneous disease. Around 70% of breast cancers are estrogen receptor-positive (ER+ve), with tamoxifen being most commonly used as an adjuvant treatment to prevent recurrence and metastasis. However, half of the patients will eventually develop tamoxifen resistance. The overexpression of c-MYC can drive the development of ER+ve breast cancer and confer tamoxifen resistance through multiple pathways. One key mechanism is to enhance ribosome biogenesis, synthesising mature ribosomes. The over-production of ribosomes sustains the demand for proteins necessary to maintain a high cell proliferation rate and combat apoptosis induced by therapeutic agents. c-MYC overexpression can induce the expression of eIF4E that favours the translation of structured mRNA to produce oncogenic factors that promote cell proliferation and confer tamoxifen resistance. Either non-phosphorylated or phosphorylated eIF4E can mediate such an effect. Since ribosomes play an essential role in c-MYC-mediated cancer development, suppressing ribosome biogenesis may help reduce aggressiveness and reverse tamoxifen resistance in breast cancer. CX-5461, CX-3543 and haemanthamine have been shown to repress ribosome biogenesis. Using these chemicals might help reverse tamoxifen resistance in ER+ve breast cancer, provided that c-MYC-mediated ribosome biogenesis is the crucial factor for tamoxifen resistance. To employ these ribosome biogenesis inhibitors to combat tamoxifen resistance in the future, identification of predictive markers will be necessary.
Collapse
Affiliation(s)
| | | | | | | | - Ui-Soon Khoo
- Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China; (H.T.); (C.-P.Y.); (M.-H.L.); (E.P.S.M.)
| |
Collapse
|
18
|
Xu Y, Huangyang P, Wang Y, Xue L, Devericks E, Nguyen HG, Yu X, Oses-Prieto JA, Burlingame AL, Miglani S, Goodarzi H, Ruggero D. ERα is an RNA-binding protein sustaining tumor cell survival and drug resistance. Cell 2021; 184:5215-5229.e17. [PMID: 34559986 PMCID: PMC8547373 DOI: 10.1016/j.cell.2021.08.036] [Citation(s) in RCA: 86] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Revised: 06/28/2021] [Accepted: 08/29/2021] [Indexed: 12/15/2022]
Abstract
Estrogen receptor α (ERα) is a hormone receptor and key driver for over 70% of breast cancers that has been studied for decades as a transcription factor. Unexpectedly, we discover that ERα is a potent non-canonical RNA-binding protein. We show that ERα RNA binding function is uncoupled from its activity to bind DNA and critical for breast cancer progression. Employing genome-wide cross-linking immunoprecipitation (CLIP) sequencing and a functional CRISPRi screen, we find that ERα-associated mRNAs sustain cancer cell fitness and elicit cellular responses to stress. Mechanistically, ERα controls different steps of RNA metabolism. In particular, we demonstrate that ERα RNA binding mediates alternative splicing of XBP1 and translation of the eIF4G2 and MCL1 mRNAs, which facilitates survival upon stress conditions and sustains tamoxifen resistance of cancer cells. ERα is therefore a multifaceted RNA-binding protein, and this activity transforms our knowledge of post-transcriptional regulation underlying cancer development and drug response.
Collapse
Affiliation(s)
- Yichen Xu
- Department of Urology, University of California, San Francisco, San Francisco, CA 94158, USA; Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Peiwei Huangyang
- Department of Urology, University of California, San Francisco, San Francisco, CA 94158, USA; Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Ying Wang
- Department of Urology, University of California, San Francisco, San Francisco, CA 94158, USA; Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Lingru Xue
- Department of Urology, University of California, San Francisco, San Francisco, CA 94158, USA; Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Emily Devericks
- Department of Urology, University of California, San Francisco, San Francisco, CA 94158, USA; Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Hao G Nguyen
- Department of Urology, University of California, San Francisco, San Francisco, CA 94158, USA; Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Xiuyan Yu
- Department of Breast Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310009, China
| | - Juan A Oses-Prieto
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Alma L Burlingame
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Sohit Miglani
- Department of Urology, University of California, San Francisco, San Francisco, CA 94158, USA; Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94158, USA; Department of Biochemistry & Biophysics, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Hani Goodarzi
- Department of Urology, University of California, San Francisco, San Francisco, CA 94158, USA; Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94158, USA; Department of Biochemistry & Biophysics, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Davide Ruggero
- Department of Urology, University of California, San Francisco, San Francisco, CA 94158, USA; Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA 94158, USA; Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94158, USA.
| |
Collapse
|
19
|
Fabbri L, Chakraborty A, Robert C, Vagner S. The plasticity of mRNA translation during cancer progression and therapy resistance. Nat Rev Cancer 2021; 21:558-577. [PMID: 34341537 DOI: 10.1038/s41568-021-00380-y] [Citation(s) in RCA: 100] [Impact Index Per Article: 33.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/14/2021] [Indexed: 02/07/2023]
Abstract
Translational control of mRNAs during gene expression allows cells to promptly and dynamically adapt to a variety of stimuli, including in neoplasia in response to aberrant oncogenic signalling (for example, PI3K-AKT-mTOR, RAS-MAPK and MYC) and microenvironmental stress such as low oxygen and nutrient supply. Such translational rewiring allows rapid, specific changes in the cell proteome that shape specific cancer phenotypes to promote cancer onset, progression and resistance to anticancer therapies. In this Review, we illustrate the plasticity of mRNA translation. We first highlight the diverse mechanisms by which it is regulated, including by translation factors (for example, eukaryotic initiation factor 4F (eIF4F) and eIF2), RNA-binding proteins, tRNAs and ribosomal RNAs that are modulated in response to aberrant intracellular pathways or microenvironmental stress. We then describe how translational control can influence tumour behaviour by impacting on the phenotypic plasticity of cancer cells as well as on components of the tumour microenvironment. Finally, we highlight the role of mRNA translation in the cellular response to anticancer therapies and its promise as a key therapeutic target.
Collapse
Affiliation(s)
- Lucilla Fabbri
- Institut Curie, PSL Research University, CNRS UMR3348, INSERM U1278, Orsay, France
- Université Paris Sud, Université Paris-Saclay, CNRS UMR3348, INSERM U1278, Orsay, France
- Equipe Labellisée Ligue Nationale Contre le Cancer, Orsay, France
| | - Alina Chakraborty
- Institut Curie, PSL Research University, CNRS UMR3348, INSERM U1278, Orsay, France
- Université Paris Sud, Université Paris-Saclay, CNRS UMR3348, INSERM U1278, Orsay, France
- Equipe Labellisée Ligue Nationale Contre le Cancer, Orsay, France
| | - Caroline Robert
- INSERM U981, Gustave Roussy Cancer Campus, Villejuif, France
- Université Paris-Sud, Université Paris-Saclay, Kremlin-Bicêtre, France
- Dermato-Oncology, Gustave Roussy Cancer Campus, Villejuif, France
| | - Stéphan Vagner
- Institut Curie, PSL Research University, CNRS UMR3348, INSERM U1278, Orsay, France.
- Université Paris Sud, Université Paris-Saclay, CNRS UMR3348, INSERM U1278, Orsay, France.
- Equipe Labellisée Ligue Nationale Contre le Cancer, Orsay, France.
- Dermato-Oncology, Gustave Roussy Cancer Campus, Villejuif, France.
| |
Collapse
|
20
|
Gandhi N, Oturkar CC, Das GM. Estrogen Receptor-Alpha and p53 Status as Regulators of AMPK and mTOR in Luminal Breast Cancer. Cancers (Basel) 2021; 13:cancers13143612. [PMID: 34298826 PMCID: PMC8306694 DOI: 10.3390/cancers13143612] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 07/10/2021] [Accepted: 07/14/2021] [Indexed: 12/12/2022] Open
Abstract
Luminal breast cancer (LBC) driven by dysregulated estrogen receptor-alpha (ERα) signaling accounts for 70% of the breast cancer cases diagnosed. Although endocrine therapy (ET) is effective against LBC, about one-third of these patients fail to respond to therapy owing to acquired or inherent resistance mechanisms. Aberrant signaling via ERα, oncogenes, growth factor receptors, and mutations in tumor suppressors such as p53 impinge on downstream regulators such as AMPK and mTOR. While both AMPK and mTOR have been reported to play important roles in determining sensitivity of LBC to ET, how the ERα-p53 crosstalk impinges on regulation of AMPK and mTOR, thereby influencing therapeutic efficacy remains unknown. Here, we have addressed this important issue using isogenic breast cancer cell lines, siRNA-mediated RNA knockdown, and different modes of drug treatments. Interaction of p53 with ERα and AMPK was determined by in situ proximity ligation assay (PLA), and endogenous gene transcripts were analyzed by quantitative real-time polymerase chain reaction (qRT-PCR). Further, the effect of concurrent and sequential administration of Fulvestrant-Everolimus combination on colony formation was determined. The studies showed that in cells expressing wild type p53, as well as in cells devoid of p53, ERα represses AMPK, whereas in cells harboring mutant p53, repression of AMPK is sustained even in the absence of ERα. AMPK is a major negative regulator of mTOR, and to our knowledge, this is the first study on the contribution of AMPK-dependent regulation of mTOR by ERα. Furthermore, the studies revealed that independent of the p53 mutation status, combination of Fulvestrant and Everolimus may be a viable first line therapeutic strategy for potentially delaying resistance of ERα+/HER2- LBC to ET.
Collapse
|
21
|
Xie J, Kusnadi EP, Furic L, Selth LA. Regulation of mRNA Translation by Hormone Receptors in Breast and Prostate Cancer. Cancers (Basel) 2021; 13:3254. [PMID: 34209750 PMCID: PMC8268847 DOI: 10.3390/cancers13133254] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 06/23/2021] [Accepted: 06/24/2021] [Indexed: 12/12/2022] Open
Abstract
Breast and prostate cancer are the second and third leading causes of death amongst all cancer types, respectively. Pathogenesis of these malignancies is characterised by dysregulation of sex hormone signalling pathways, mediated by the estrogen receptor-α (ER) in breast cancer and androgen receptor (AR) in prostate cancer. ER and AR are transcription factors whose aberrant function drives oncogenic transcriptional programs to promote cancer growth and progression. While ER/AR are known to stimulate cell growth and survival by modulating gene transcription, emerging findings indicate that their effects in neoplasia are also mediated by dysregulation of protein synthesis (i.e., mRNA translation). This suggests that ER/AR can coordinately perturb both transcriptional and translational programs, resulting in the establishment of proteomes that promote malignancy. In this review, we will discuss relatively understudied aspects of ER and AR activity in regulating protein synthesis as well as the potential of targeting mRNA translation in breast and prostate cancer.
Collapse
Affiliation(s)
- Jianling Xie
- Flinders Health and Medical Research Institute, Flinders University, Bedford Park, SA 5042, Australia
| | - Eric P Kusnadi
- Translational Prostate Cancer Research, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia
- Cancer Program, Biomedicine Discovery Institute and Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC 3800, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Luc Furic
- Translational Prostate Cancer Research, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia
- Cancer Program, Biomedicine Discovery Institute and Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC 3800, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Luke A Selth
- Flinders Health and Medical Research Institute, Flinders University, Bedford Park, SA 5042, Australia
- Freemasons Centre for Male Health and Wellbeing, Flinders University, Bedford Park, SA 5042, Australia
- Adelaide Medical School, University of Adelaide, Adelaide, SA 5005, Australia
| |
Collapse
|
22
|
Inhibitory effects of Tomivosertib in acute myeloid leukemia. Oncotarget 2021; 12:955-966. [PMID: 34012509 PMCID: PMC8121614 DOI: 10.18632/oncotarget.27952] [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: 02/23/2021] [Accepted: 04/19/2021] [Indexed: 12/26/2022] Open
Abstract
The MAPK-interacting kinases 1 and 2 (MNK1/2) have generated increasing interest as therapeutic targets for acute myeloid leukemia (AML). We evaluated the therapeutic potential of the highly-selective MNK1/2 inhibitor Tomivosertib on AML cells. Tomivosertib was highly effective at blocking eIF4E phosphorylation on serine 209 in AML cells. Such inhibitory effects correlated with dose-dependent suppression of cellular viability and leukemic progenitor colony formation. Moreover, combination of Tomivosertib and Venetoclax resulted in synergistic anti-leukemic responses in AML cell lines. Mass spectrometry studies identified novel putative MNK1/2 interactors, while in parallel studies we demonstrated that MNK2 - RAPTOR - mTOR complexes are not disrupted by Tomivosertib. Overall, these findings demonstrate that Tomivosertib exhibits potent anti-leukemic properties on AML cells and support the development of clinical translational efforts involving the use of this drug, alone or in combination with other therapies for the treatment of AML.
Collapse
|
23
|
Knight JRP, Alexandrou C, Skalka GL, Vlahov N, Pennel K, Officer L, Teodosio A, Kanellos G, Gay DM, May-Wilson S, Smith EM, Najumudeen AK, Gilroy K, Ridgway RA, Flanagan DJ, Smith RCL, McDonald L, MacKay C, Cheasty A, McArthur K, Stanway E, Leach JD, Jackstadt R, Waldron JA, Campbell AD, Vlachogiannis G, Valeri N, Haigis KM, Sonenberg N, Proud CG, Jones NP, Swarbrick ME, McKinnon HJ, Faller WJ, Le Quesne J, Edwards J, Willis AE, Bushell M, Sansom OJ. MNK Inhibition Sensitizes KRAS-Mutant Colorectal Cancer to mTORC1 Inhibition by Reducing eIF4E Phosphorylation and c-MYC Expression. Cancer Discov 2021; 11:1228-1247. [PMID: 33328217 PMCID: PMC7611341 DOI: 10.1158/2159-8290.cd-20-0652] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 10/21/2020] [Accepted: 12/11/2020] [Indexed: 12/16/2022]
Abstract
KRAS-mutant colorectal cancers are resistant to therapeutics, presenting a significant problem for ∼40% of cases. Rapalogs, which inhibit mTORC1 and thus protein synthesis, are significantly less potent in KRAS-mutant colorectal cancer. Using Kras-mutant mouse models and mouse- and patient-derived organoids, we demonstrate that KRAS with G12D mutation fundamentally rewires translation to increase both bulk and mRNA-specific translation initiation. This occurs via the MNK/eIF4E pathway culminating in sustained expression of c-MYC. By genetic and small-molecule targeting of this pathway, we acutely sensitize KRASG12D models to rapamycin via suppression of c-MYC. We show that 45% of colorectal cancers have high signaling through mTORC1 and the MNKs, with this signature correlating with a 3.5-year shorter cancer-specific survival in a subset of patients. This work provides a c-MYC-dependent cotargeting strategy with remarkable potency in multiple Kras-mutant mouse models and metastatic human organoids and identifies a patient population that may benefit from its clinical application. SIGNIFICANCE: KRAS mutation and elevated c-MYC are widespread in many tumors but remain predominantly untargetable. We find that mutant KRAS modulates translation, culminating in increased expression of c-MYC. We describe an effective strategy targeting mTORC1 and MNK in KRAS-mutant mouse and human models, pathways that are also commonly co-upregulated in colorectal cancer.This article is highlighted in the In This Issue feature, p. 995.
Collapse
Affiliation(s)
| | | | - George L Skalka
- CRUK Beatson Institute, Glasgow, United Kingdom
- MRC Toxicology Unit, University of Cambridge, Cambridge, United Kingdom
| | | | - Kathryn Pennel
- Institute of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Leah Officer
- MRC Toxicology Unit, University of Cambridge, Cambridge, United Kingdom
| | - Ana Teodosio
- MRC Toxicology Unit, University of Cambridge, Cambridge, United Kingdom
| | | | - David M Gay
- CRUK Beatson Institute, Glasgow, United Kingdom
- Institute of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom
| | | | | | | | | | | | | | - Rachael C L Smith
- CRUK Beatson Institute, Glasgow, United Kingdom
- Institute of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Laura McDonald
- Drug Discovery Unit, CRUK Beatson Institute, Glasgow, United Kingdom
| | - Craig MacKay
- Drug Discovery Unit, CRUK Beatson Institute, Glasgow, United Kingdom
| | - Anne Cheasty
- CRUK Therapeutic Discovery Laboratories, Jonas Webb Building, Babraham Research Campus, Cambridge, United Kingdom
| | - Kerri McArthur
- CRUK Therapeutic Discovery Laboratories, Jonas Webb Building, Babraham Research Campus, Cambridge, United Kingdom
| | - Emma Stanway
- CRUK Therapeutic Discovery Laboratories, Jonas Webb Building, Babraham Research Campus, Cambridge, United Kingdom
| | - Joshua D Leach
- CRUK Beatson Institute, Glasgow, United Kingdom
- Institute of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom
| | | | | | | | - Georgios Vlachogiannis
- Division of Molecular Pathology, The Institute of Cancer Research, London, United Kingdom
| | - Nicola Valeri
- Division of Molecular Pathology, The Institute of Cancer Research, London, United Kingdom
- Department of Medicine, The Royal Marsden NHS Foundation Trust, London, United Kingdom
| | - Kevin M Haigis
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
- Department of Cell Biology, Harvard Medical School, Boston, Massachusetts
| | - Nahum Sonenberg
- Department of Biochemistry and Goodman Cancer Research Center, McGill University, Montreal, Quebec, Canada
| | - Christopher G Proud
- Lifelong Health, South Australian Health and Medical Research Institute, North Terrace, Adelaide, South Australia, Australia
- Department of Biological Sciences, University of Adelaide, Adelaide, South Australia, Australia
| | - Neil P Jones
- CRUK Therapeutic Discovery Laboratories, Jonas Webb Building, Babraham Research Campus, Cambridge, United Kingdom
| | - Martin E Swarbrick
- CRUK Therapeutic Discovery Laboratories, Jonas Webb Building, Babraham Research Campus, Cambridge, United Kingdom
| | | | | | - John Le Quesne
- MRC Toxicology Unit, University of Cambridge, Cambridge, United Kingdom
- Institute of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom
- Leicester Cancer Research Centre, University of Leicester, Leicester, United Kingdom
- Glenfield Hospital, Leicester University Hospitals NHS Trust, Leicester, United Kingdom
| | - Joanne Edwards
- Institute of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Anne E Willis
- MRC Toxicology Unit, University of Cambridge, Cambridge, United Kingdom
| | - Martin Bushell
- CRUK Beatson Institute, Glasgow, United Kingdom
- Institute of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Owen J Sansom
- CRUK Beatson Institute, Glasgow, United Kingdom.
- Institute of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom
| |
Collapse
|
24
|
Gruslova A, McClellan B, Balinda HU, Viswanadhapalli S, Alers V, Sareddy GR, Huang T, Garcia M, deGraffenried L, Vadlamudi RK, Brenner AJ. FASN inhibition as a potential treatment for endocrine-resistant breast cancer. Breast Cancer Res Treat 2021; 187:375-386. [PMID: 33893909 DOI: 10.1007/s10549-021-06231-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Accepted: 04/14/2021] [Indexed: 01/18/2023]
Abstract
PURPOSE The majority of breast cancers are estrogen receptor (ERα) positive making endocrine therapy a mainstay for these patients. Unfortunately, resistance to endocrine therapy is a common occurrence. Fatty acid synthase (FASN) is a key enzyme in lipid biosynthesis and its expression is commensurate with tumor grade and resistance to numerous therapies. METHODS The effect of the FASN inhibitor TVB-3166 on ERα expression and cell growth was characterized in tamoxifen-resistant cell lines, xenografts, and patient explants. Subcellular localization of ERα was assessed using subcellular fractionations. Palmitoylation and ubiquitination of ERα were assessed by immunoprecipitation. ERα and p-eIF2α protein levels were analyzed by Western blotting after treatment with TVB-3166 with or without the addition of palmitate or BAPTA. RESULTS TVB-3166 treatment leads to a marked inhibition of proliferation in tamoxifen-resistant cells compared to the parental cells. Additionally, TVB-3166 significantly inhibited tamoxifen-resistant breast tumor growth in mice and decreased proliferation of primary tumor explants compared to untreated controls. FASN inhibition significantly reduced ERα levels most prominently in endocrine-resistant cells and altered its subcellular localization. Furthermore, we showed that the reduction of ERα expression upon TVB-3166 treatment is mediated through the induction of endoplasmic reticulum stress. CONCLUSION Our preclinical data provide evidence that FASN inhibition by TVB-3166 presents a promising therapeutic strategy for the treatment of endocrine-resistant breast cancer. Further clinical development of FASN inhibitors for endocrine-resistant breast cancer should be considered.
Collapse
Affiliation(s)
| | | | | | | | - Victoria Alers
- UT Health San Antonio MD Anderson Cancer Center, San Antonio, TX, USA
| | - Gangadhara R Sareddy
- Department of Obstetrics and Gynecology, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Tim Huang
- UT Health San Antonio MD Anderson Cancer Center, San Antonio, TX, USA
| | - Michael Garcia
- UT Health San Antonio MD Anderson Cancer Center, San Antonio, TX, USA
| | | | - Ratna K Vadlamudi
- Department of Obstetrics and Gynecology, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Andrew J Brenner
- UT Health San Antonio MD Anderson Cancer Center, San Antonio, TX, USA.
- South Texas Research Facility, University of Texas Health San Antonio, STRF 2.208.58403 Floyd Curl Dr, San Antonio, TX, 78229, USA.
| |
Collapse
|
25
|
Jin X, Yu R, Wang X, Proud CG, Jiang T. Progress in developing MNK inhibitors. Eur J Med Chem 2021; 219:113420. [PMID: 33892273 DOI: 10.1016/j.ejmech.2021.113420] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Revised: 03/19/2021] [Accepted: 03/22/2021] [Indexed: 12/19/2022]
Abstract
The MNKs (mitogen-activated protein kinase-interacting protein kinases) phosphorylate eIF4E (eukaryotic initiation factor 4 E) at serine 209; eIF4E plays an important role in the translation of cytoplasmic mRNAs, all of which possess a 5' 'cap' structure to which eIF4E binds. Elevated levels of eIF4E, p-eIF4E and/or the MNK protein kinases have been found in many types of cancer, including solid tumors and leukemia. MNKs also play a role in metabolic disease. Regulation of the activities of MNKs (MNK1 and MNK2), control the phosphorylation of eIF4E, which in turn has a close relationship with the processes of tumor development, cell migration and invasion, and energy metabolism. MNK knock-out mice display no adverse effects on normal cells or phenotypes suggesting that MNK may be a potentially safe targets for the treatment of various cancers. Several MNK inhibitors or 'degraders' have been identified. Initially, some of the inhibitors were developed from natural products or based on other protein kinase inhibitors which inhibit multiple kinases. Subsequently, more potent and selective inhibitors for MNK1/2 have been designed and synthesized. Currently, three inhibitors (BAY1143269, eFT508 and ETC-206) are in various stages of clinical trials for the treatment of solid cancers or leukemia, either alone or combined with inhibitors of other protein kinase. In this review, we summarize the diverse MNK inhibitors that have been reported in patents and other literature, including those with activities in vitro and/or in vivo.
Collapse
Affiliation(s)
- Xin Jin
- School of Medicine and Pharmacy, Ocean University of China and Laboratory for Marine Drugs and Bioproducts, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China; College of Marine Life Sciences, Ocean University of China, Qingdao, China
| | - Rilei Yu
- School of Medicine and Pharmacy, Ocean University of China and Laboratory for Marine Drugs and Bioproducts, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| | - Xuemin Wang
- Lifelong Health, South Australian Health & Medical Research Institute, North Terrace, Adelaide, SA5000, Australia; School of Biomedical Sciences, University of Adelaide, Adelaide, SA, 5005, Australia
| | - Christopher G Proud
- Lifelong Health, South Australian Health & Medical Research Institute, North Terrace, Adelaide, SA5000, Australia; School of Biomedical Sciences, University of Adelaide, Adelaide, SA, 5005, Australia
| | - Tao Jiang
- School of Medicine and Pharmacy, Ocean University of China and Laboratory for Marine Drugs and Bioproducts, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China.
| |
Collapse
|
26
|
Xu Y, Liao S, Wang L, Wang Y, Wei W, Su K, Tu Y, Zhu S. Galeterone sensitizes breast cancer to chemotherapy via targeting MNK/eIF4E and β-catenin. Cancer Chemother Pharmacol 2020; 87:85-93. [PMID: 33159561 DOI: 10.1007/s00280-020-04195-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Accepted: 10/24/2020] [Indexed: 02/06/2023]
Abstract
Aberrant activation of eIF4E signalling pathway is common in breast cancer and holds potential therapeutic options. In our work, galeterone as a chemical compound under clinical trials for the treatment of prostate cancer, was identified to be effective in targeting breast cancer cells via suppressing MNK-eIF4E and β-catenin. In despite of varying IC50, galeterone at nanomolar concentrations significantly decreased viability, proliferation and migration of a panel of breast cancer cell lines regardless of clinical subtypes and genetic mutations, and to a higher extent than in normal breast cells. Galeterone significantly enhanced the effects of chemotherapeutic drugs in reducing proliferation and viability but not migration. The in vivo efficacy of galeterone as single drug alone and its ability in augmenting chemotherapy's efficacy were also shown in breast cancer xenograft mouse model. Mechanism analysis demonstrated that galeterone decreased MNK1/2 level and phosphorylation of eIF4E. In addition, galeterone decreased β-catenin level via promoting GSK-3β-mediated β-catenin degradation, and furthermore that Akt but not CK1 was involved in β-catenin degradation by galeterone. Rescue studies demonstrated that both MNK/eIF4E and β-catenin were responsible for anti-breast cancer activity of galeterone. Our study provides pre-clinical evidence to initialize clinical trials for breast cancer using galeterone in combination with chemotherapy.
Collapse
Affiliation(s)
- Yulin Xu
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Shichong Liao
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Lijun Wang
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Yuan Wang
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Wen Wei
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Ke Su
- Department of Nephrology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Yi Tu
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China.
| | - Shan Zhu
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China.
| |
Collapse
|
27
|
Yang X, Zhong W, Cao R. Phosphorylation of the mRNA cap-binding protein eIF4E and cancer. Cell Signal 2020; 73:109689. [PMID: 32535199 PMCID: PMC8049097 DOI: 10.1016/j.cellsig.2020.109689] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 05/21/2020] [Accepted: 06/02/2020] [Indexed: 12/22/2022]
Abstract
Dysregulated protein synthesis is frequently involved in oncogenesis and cancer progression. Translation initiation is thought to be the rate-limiting step in protein synthesis, and the mRNA 5' cap-binding protein eukaryotic translation initiation factor 4E (eIF4E) is a pivotal factor that initiates translation. The activities of eIF4E are regulated at multiple levels, one of which is through its phosphorylation at Serine 209 by the mitogen-activated protein kinase-interacting kinases (MNKs, including MNK1 and MNK2). Benefiting from novel mouse genetic tools and pharmacological MNK inhibitors, our understanding of a role for eIF4E phosphorylation in tumor biology and cancer therapy has greatly evolved in recent years. Importantly, recent studies have found that the level of eIF4E phosphorylation is frequently upregulated in a wide variety of human cancer types, and phosphorylation of eIF4E drives a number of important processes in cancer biology, including cell transformation, proliferation, apoptosis, metastasis and angiogenesis. The MNK-eIF4E axis is being assessed as a therapeutic target either alone or in combination with other therapies in different cancer models. As novel MNK inhibitors are being developed, experimental studies bring new hope to cure human cancers that are not responsive to traditional therapies. Herein we review recent progress on our understanding of a mechanistic role for phosphorylation of eIF4E in cancer biology and therapy.
Collapse
Affiliation(s)
- Xiaotong Yang
- School of Medicine, Tsinghua University, Beijing 100084, China; National Engineering Research Center for the Emergency Drug, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China; Department of Biomedical Sciences, University of Minnesota Medical School, Duluth, MN 55812, USA
| | - Wu Zhong
- National Engineering Research Center for the Emergency Drug, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China.
| | - Ruifeng Cao
- Department of Biomedical Sciences, University of Minnesota Medical School, Duluth, MN 55812, USA; Department of Neuroscience, University of Minnesota Medical School, Minneapolis, MN 55455, USA.
| |
Collapse
|
28
|
Prabhu SA, Moussa O, Miller WH, del Rincón SV. The MNK1/2-eIF4E Axis as a Potential Therapeutic Target in Melanoma. Int J Mol Sci 2020; 21:E4055. [PMID: 32517051 PMCID: PMC7312468 DOI: 10.3390/ijms21114055] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 05/28/2020] [Accepted: 05/29/2020] [Indexed: 12/12/2022] Open
Abstract
: Melanoma is a type of skin cancer that originates in the pigment-producing cells of the body known as melanocytes. Most genetic aberrations in melanoma result in hyperactivation of the mitogen activated protein kinase (MAPK) and phosphoinositide 3-kinase (PI3K) pathways. We and others have shown that a specific protein synthesis pathway known as the MNK1/2-eIF4E axis is often dysregulated in cancer. The MNK1/2-eIF4E axis is a point of convergence for these signaling pathways that are commonly constitutively activated in melanoma. In this review we consider the functional implications of aberrant mRNA translation in melanoma and other malignancies. Moreover, we discuss the consequences of inhibiting the MNK1/2-eIF4E axis on the tumor and tumor-associated cells, and we provide important avenues for the utilization of this treatment modality in combination with other targeted and immune-based therapies. The past decade has seen the increased development of selective inhibitors to block the action of the MNK1/2-eIF4E pathway, which are predicted to be an effective therapy regardless of the melanoma subtype (e.g., cutaneous, acral, and mucosal).
Collapse
Affiliation(s)
- Sathyen A. Prabhu
- Division of Experimental Medicine, McGill University, 1001 Decarie Boulevard, Montreal, QC H4A 3J1, Canada; (S.A.P.); (O.M.); (W.H.M.J.)
- Lady Davis Institute, Jewish General Hospital, McGill University, 3755 Côte Ste-Catherine Road, Montreal, QC H3T 1E2, Canada
| | - Omar Moussa
- Division of Experimental Medicine, McGill University, 1001 Decarie Boulevard, Montreal, QC H4A 3J1, Canada; (S.A.P.); (O.M.); (W.H.M.J.)
- Lady Davis Institute, Jewish General Hospital, McGill University, 3755 Côte Ste-Catherine Road, Montreal, QC H3T 1E2, Canada
| | - Wilson H. Miller
- Division of Experimental Medicine, McGill University, 1001 Decarie Boulevard, Montreal, QC H4A 3J1, Canada; (S.A.P.); (O.M.); (W.H.M.J.)
- Lady Davis Institute, Jewish General Hospital, McGill University, 3755 Côte Ste-Catherine Road, Montreal, QC H3T 1E2, Canada
- Department of Oncology, McGill University, 845 Sherbrooke St W, Montreal, QC H3A 0G4, Canada
- McGill Centre for Translational Research in Cancer (MCTRC), McGill University, 3755 Côte Ste-Catherine Road, Montreal, QC H3T 1E2, Canada
- Rossy Cancer Network, McGill University, 1980 Sherbrooke Ouest, #1101, Montreal, QC H3H 1E8, Canada
| | - Sonia V. del Rincón
- Division of Experimental Medicine, McGill University, 1001 Decarie Boulevard, Montreal, QC H4A 3J1, Canada; (S.A.P.); (O.M.); (W.H.M.J.)
- Lady Davis Institute, Jewish General Hospital, McGill University, 3755 Côte Ste-Catherine Road, Montreal, QC H3T 1E2, Canada
- Department of Oncology, McGill University, 845 Sherbrooke St W, Montreal, QC H3A 0G4, Canada
- McGill Centre for Translational Research in Cancer (MCTRC), McGill University, 3755 Côte Ste-Catherine Road, Montreal, QC H3T 1E2, Canada
| |
Collapse
|
29
|
Phosphorylation independent eIF4E translational reprogramming of selective mRNAs determines tamoxifen resistance in breast cancer. Oncogene 2020; 39:3206-3217. [PMID: 32066877 PMCID: PMC7142019 DOI: 10.1038/s41388-020-1210-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Revised: 01/28/2020] [Accepted: 02/05/2020] [Indexed: 11/12/2022]
Abstract
Eukaryotic translation initiation factor 4E (eIF4E) selectively promotes translation of mRNAs with atypically long and structured 5′-UTRs and has been implicated in drug resistance. Through genome-wide transcriptome and translatome analysis we revealed eIF4E overexpression could promote cellular activities mediated by ERα and FOXM1 signalling pathways. Whilst eIF4E overexpression could enhance the translation of both ERα and FOXM1, it also led to enhanced transcription of FOXM1. Polysome fractionation experiments confirmed eIF4E could modulate the translation of ERα and FOXM1 mRNA. The enhancement of FOXM1 transcription was contingent upon the presence of ERα, and it was the high levels of FOXM1 that conferred Tamoxifen resistance. Furthermore, tamoxifen resistance was conferred by phosphorylation independent eIF4E overexpression. Immunohistochemistry on 134 estrogen receptor (ER+) primary breast cancer samples confirmed that high eIF4E expression was significantly associated with increased ERα and FOXM1, and significantly associated with tamoxifen resistance. Our study uncovers a novel mechanism whereby phosphorylation independent eIF4E translational reprogramming in governing the protein synthesis of ERα and FOXM1 contributes to anti-estrogen insensitivity in ER+ breast cancer. In eIF4E overexpressing breast cancer, the increased ERα protein expression in turn enhances FOXM1 transcription, which together with its increased translation regulated by eIF4E, contributes to tamoxifen resistance. Coupled with eIF4E translational regulation, our study highlights an important mechanism conferring tamoxifen resistance via both ERα dependent and independent pathways.
Collapse
|
30
|
Lorent J, Kusnadi EP, van Hoef V, Rebello RJ, Leibovitch M, Ristau J, Chen S, Lawrence MG, Szkop KJ, Samreen B, Balanathan P, Rapino F, Close P, Bukczynska P, Scharmann K, Takizawa I, Risbridger GP, Selth LA, Leidel SA, Lin Q, Topisirovic I, Larsson O, Furic L. Translational offsetting as a mode of estrogen receptor α-dependent regulation of gene expression. EMBO J 2019; 38:e101323. [PMID: 31556460 PMCID: PMC6885737 DOI: 10.15252/embj.2018101323] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Revised: 08/28/2019] [Accepted: 08/30/2019] [Indexed: 12/25/2022] Open
Abstract
Estrogen receptor alpha (ERα) activity is associated with increased cancer cell proliferation. Studies aiming to understand the impact of ERα on cancer-associated phenotypes have largely been limited to its transcriptional activity. Herein, we demonstrate that ERα coordinates its transcriptional output with selective modulation of mRNA translation. Importantly, translational perturbations caused by depletion of ERα largely manifest as "translational offsetting" of the transcriptome, whereby amounts of translated mRNAs and corresponding protein levels are maintained constant despite changes in mRNA abundance. Transcripts whose levels, but not polysome association, are reduced following ERα depletion lack features which limit translation efficiency including structured 5'UTRs and miRNA target sites. In contrast, mRNAs induced upon ERα depletion whose polysome association remains unaltered are enriched in codons requiring U34-modified tRNAs for efficient decoding. Consistently, ERα regulates levels of U34-modifying enzymes and thereby controls levels of U34-modified tRNAs. These findings unravel a hitherto unprecedented mechanism of ERα-dependent orchestration of transcriptional and translational programs that may be a pervasive mechanism of proteome maintenance in hormone-dependent cancers.
Collapse
Affiliation(s)
- Julie Lorent
- Science for Life LaboratoryDepartment of Oncology‐PathologyKarolinska InstitutetSolnaSweden
| | - Eric P Kusnadi
- Prostate Cancer Translational Research LaboratoryPeter MacCallum Cancer CentreMelbourneVic.Australia
- Cancer ProgramBiomedicine Discovery Institute and Department of Anatomy and Developmental BiologyMonash UniversityClaytonVic.Australia
- Sir Peter MacCallum Department of OncologyUniversity of MelbourneParkvilleVic.Australia
| | - Vincent van Hoef
- Science for Life LaboratoryDepartment of Oncology‐PathologyKarolinska InstitutetSolnaSweden
| | - Richard J Rebello
- Prostate Cancer Translational Research LaboratoryPeter MacCallum Cancer CentreMelbourneVic.Australia
- Cancer ProgramBiomedicine Discovery Institute and Department of Anatomy and Developmental BiologyMonash UniversityClaytonVic.Australia
| | - Matthew Leibovitch
- Gerald Bronfman Department of Oncology and Departments of Biochemistry and Experimental MedicineLady Davis InstituteMcGill UniversityMontrealQCCanada
| | - Johannes Ristau
- Science for Life LaboratoryDepartment of Oncology‐PathologyKarolinska InstitutetSolnaSweden
| | - Shan Chen
- Science for Life LaboratoryDepartment of Oncology‐PathologyKarolinska InstitutetSolnaSweden
| | - Mitchell G Lawrence
- Prostate Cancer Translational Research LaboratoryPeter MacCallum Cancer CentreMelbourneVic.Australia
- Cancer ProgramBiomedicine Discovery Institute and Department of Anatomy and Developmental BiologyMonash UniversityClaytonVic.Australia
- Sir Peter MacCallum Department of OncologyUniversity of MelbourneParkvilleVic.Australia
| | - Krzysztof J Szkop
- Science for Life LaboratoryDepartment of Oncology‐PathologyKarolinska InstitutetSolnaSweden
| | - Baila Samreen
- Science for Life LaboratoryDepartment of Oncology‐PathologyKarolinska InstitutetSolnaSweden
| | - Preetika Balanathan
- Cancer ProgramBiomedicine Discovery Institute and Department of Anatomy and Developmental BiologyMonash UniversityClaytonVic.Australia
| | - Francesca Rapino
- Laboratory of Cancer SignalingGIGA‐InstituteUniversity of LiègeLiègeBelgium
| | - Pierre Close
- Laboratory of Cancer SignalingGIGA‐InstituteUniversity of LiègeLiègeBelgium
| | - Patricia Bukczynska
- Prostate Cancer Translational Research LaboratoryPeter MacCallum Cancer CentreMelbourneVic.Australia
| | - Karin Scharmann
- Max Planck Institute for Molecular BiomedicineMünsterGermany
- Cells‐in‐Motion Cluster of ExcellenceUniversity of MünsterMünsterGermany
| | - Itsuhiro Takizawa
- Cancer ProgramBiomedicine Discovery Institute and Department of Anatomy and Developmental BiologyMonash UniversityClaytonVic.Australia
| | - Gail P Risbridger
- Prostate Cancer Translational Research LaboratoryPeter MacCallum Cancer CentreMelbourneVic.Australia
- Cancer ProgramBiomedicine Discovery Institute and Department of Anatomy and Developmental BiologyMonash UniversityClaytonVic.Australia
- Sir Peter MacCallum Department of OncologyUniversity of MelbourneParkvilleVic.Australia
| | - Luke A Selth
- Dame Roma Mitchell Cancer Research Laboratories and Freemasons Foundation Centre for Men's HealthAdelaide Medical SchoolFaculty of Health and Medical SciencesThe University of AdelaideAdelaideSAAustralia
| | - Sebastian A Leidel
- Max Planck Institute for Molecular BiomedicineMünsterGermany
- Cells‐in‐Motion Cluster of ExcellenceUniversity of MünsterMünsterGermany
- Department of Chemistry and BiochemistryUniversity of BernBernSwitzerland
| | - Qishan Lin
- RNA Epitranscriptomics & Proteomics ResourceDepartment of ChemistryUniversity at AlbanyAlbanyNYUSA
| | - Ivan Topisirovic
- Gerald Bronfman Department of Oncology and Departments of Biochemistry and Experimental MedicineLady Davis InstituteMcGill UniversityMontrealQCCanada
| | - Ola Larsson
- Science for Life LaboratoryDepartment of Oncology‐PathologyKarolinska InstitutetSolnaSweden
| | - Luc Furic
- Prostate Cancer Translational Research LaboratoryPeter MacCallum Cancer CentreMelbourneVic.Australia
- Cancer ProgramBiomedicine Discovery Institute and Department of Anatomy and Developmental BiologyMonash UniversityClaytonVic.Australia
- Sir Peter MacCallum Department of OncologyUniversity of MelbourneParkvilleVic.Australia
| |
Collapse
|
31
|
Liu X, Miao W, Huang M, Li L, Dai X, Wang Y. Elevated Hexokinase II Expression Confers Acquired Resistance to 4-Hydroxytamoxifen in Breast Cancer Cells. Mol Cell Proteomics 2019; 18:2273-2284. [PMID: 31519767 PMCID: PMC6823848 DOI: 10.1074/mcp.ra119.001576] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 08/26/2019] [Indexed: 12/11/2022] Open
Abstract
Tamoxifen has been clinically used in treating estrogen receptor (ER)-positive breast cancer for over 30 years. The most challenging aspect associated with tamoxifen therapy is the development of resistance in initially responsive breast tumors. We applied a parallel-reaction monitoring (PRM)-based quantitative proteomic method to examine the differential expression of kinase proteins in MCF-7 and the isogenic tamoxifen-resistant (TamR) cells. We were able to quantify the relative protein expression levels of 315 kinases, among which hexokinase 2 (HK2) and mTOR were up-regulated in TamR MCF-7 cells. We also observed that the TamR MCF-7 cells exhibited elevated rate of glycolysis than the parental MCF-7 cells. In addition, we found that phosphorylation of S6K - a target of mTOR - was much lower in TamR MCF-7 cells, and this phosphorylation level could be restored upon genetic depletion or pharmacological inhibition of HK2. Reciprocally, the level of S6K phosphorylation was diminished upon overexpression of HK2 in MCF-7 cells. Moreover, we observed that HK2 interacts with mTOR, and this interaction inhibits mTOR activity. Lower mTOR activity led to augmented autophagy, which conferred resistance of MCF-7 cells toward tamoxifen. Together, our study demonstrates that elevated expression of HK2 promotes autophagy through inhibiting the mTOR-S6K signaling pathway and results in resistance of MCF-7 breast cancer cells toward tamoxifen; thus, our results uncovered, for the first time, HK2 as a potential therapeutic target for overcoming tamoxifen resistance.
Collapse
Affiliation(s)
- Xiaochuan Liu
- Department of Chemistry, University of California, Riverside, Riverside, CA 92521
| | - Weili Miao
- Department of Chemistry, University of California, Riverside, Riverside, CA 92521
| | - Ming Huang
- Environmental Toxicology Graduate Program, University of California, Riverside, Riverside, CA 92521
| | - Lin Li
- Department of Chemistry, University of California, Riverside, Riverside, CA 92521
| | - Xiaoxia Dai
- Department of Chemistry, University of California, Riverside, Riverside, CA 92521
| | - Yinsheng Wang
- Department of Chemistry, University of California, Riverside, Riverside, CA 92521; Environmental Toxicology Graduate Program, University of California, Riverside, Riverside, CA 92521.
| |
Collapse
|
32
|
Robichaud N, Sonenberg N, Ruggero D, Schneider RJ. Translational Control in Cancer. Cold Spring Harb Perspect Biol 2019; 11:cshperspect.a032896. [PMID: 29959193 DOI: 10.1101/cshperspect.a032896] [Citation(s) in RCA: 170] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The translation of messenger RNAs (mRNAs) into proteins is a key event in the regulation of gene expression. This is especially true in the cancer setting, as many oncogenes and transforming events are regulated at this level. Cancer-promoting factors that are translationally regulated include cyclins, antiapoptotic factors, proangiogenic factors, regulators of cell metabolism, prometastatic factors, immune modulators, and proteins involved in DNA repair. This review discusses the diverse means by which cancer cells deregulate and reprogram translation, and the resulting oncogenic impacts, providing insights into the complexity of translational control in cancer and its targeting for cancer therapy.
Collapse
Affiliation(s)
- Nathaniel Robichaud
- Goodman Cancer Research Centre and Department of Biochemistry, McGill University, Montreal, Quebec H3A 1A3, Canada
| | - Nahum Sonenberg
- Goodman Cancer Research Centre and Department of Biochemistry, McGill University, Montreal, Quebec H3A 1A3, Canada
| | - Davide Ruggero
- Helen Diller Family Comprehensive Cancer Center, and Departments of Urology and of Cellular and Molecular Pharmacology, University of California, San Francisco, California 94158
| | - Robert J Schneider
- NYU School of Medicine, Alexandria Center for Life Science, New York, New York 10016
| |
Collapse
|
33
|
Xie J, Merrett JE, Jensen KB, Proud CG. The MAP kinase-interacting kinases (MNKs) as targets in oncology. Expert Opin Ther Targets 2019; 23:187-199. [DOI: 10.1080/14728222.2019.1571043] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Jianling Xie
- Nutrition & Metabolism, South Australian Health & Medical Research Institute, Adelaide, Australia
| | - James E. Merrett
- Nutrition & Metabolism, South Australian Health & Medical Research Institute, Adelaide, Australia
| | - Kirk B. Jensen
- Nutrition & Metabolism, South Australian Health & Medical Research Institute, Adelaide, Australia
- School of Biological Sciences, University of Adelaide, Adelaide, Australia
| | - Christopher G. Proud
- Nutrition & Metabolism, South Australian Health & Medical Research Institute, Adelaide, Australia
- School of Biological Sciences, University of Adelaide, Adelaide, Australia
| |
Collapse
|
34
|
Liu J, Li J, Li P, Jiang Y, Chen H, Wang R, Cao F, Liu P. DLG5 suppresses breast cancer stem cell-like characteristics to restore tamoxifen sensitivity by inhibiting TAZ expression. J Cell Mol Med 2018; 23:512-521. [PMID: 30450766 PMCID: PMC6307757 DOI: 10.1111/jcmm.13954] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Revised: 07/17/2018] [Accepted: 09/12/2018] [Indexed: 01/02/2023] Open
Abstract
Tamoxifen (TAM) is a primary drug for treatment of estrogen receptor positive breast cancer. However, TAM resistance remains a serious threat to breast cancer patients and may be attributed to increased stemness of breast cancer. Here, we show that discs large homolog 5 (DLG5) expression is down‐regulated in TAM‐resistant breast cancer and cells. DLG5 silencing decreased the sensitivity to TAM and increased the frequency and stemness of CD44+/CD24− breast cancer stem cells (BCSCs) and TAZ, a transducer of the Hippo pathway, expression in MCF7 cells while DLG5 overexpression had opposite effects. TAZ silencing restored the sensitivity to TAM and reduced the frequency and stemness in TAM‐resistant breast cancer cells. Taken together, our data indicate that down‐regulated DLG5 expression increases the stemness of breast cancer cells by enhancing TAZ expression, contributing to TAM resistance in breast cancer.
Collapse
Affiliation(s)
- Jie Liu
- Center for Translational Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China.,Key Laboratory for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Juan Li
- Center for Translational Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China.,Key Laboratory for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Pingping Li
- Center for Translational Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China.,Key Laboratory for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Yina Jiang
- Department of Pathology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - He Chen
- Center for Translational Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China.,Key Laboratory for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Ruiqi Wang
- Center for Translational Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China.,Key Laboratory for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Fang Cao
- Center for Translational Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China.,Key Laboratory for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Peijun Liu
- Center for Translational Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China.,Key Laboratory for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| |
Collapse
|
35
|
Pham TND, Kumar K, DeCant BT, Shang M, Munshi SZ, Matsangou M, Ebine K, Munshi HG. Induction of MNK Kinase-dependent eIF4E Phosphorylation by Inhibitors Targeting BET Proteins Limits Efficacy of BET Inhibitors. Mol Cancer Ther 2018; 18:235-244. [PMID: 30446586 DOI: 10.1158/1535-7163.mct-18-0768] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Revised: 10/10/2018] [Accepted: 11/13/2018] [Indexed: 12/13/2022]
Abstract
BET inhibitors (BETi), which target transcription of key oncogenic genes, are currently being evaluated in early-phase clinical trials. However, because BETis show limited single-agent activity, there is increasing interest in identifying signaling pathways to enhance the efficacy of BETis. Here, we demonstrate increased MNK kinase-dependent eIF4E phosphorylation following treatment with BETis, indicating activation of a prosurvival feedback mechanism in response to BETis. BET PROTACs, which promote degradation of BET proteins, also induced eIF4E phosphorylation in cancer cells. Mechanistically, we show that the effect of BETis on MNK-eIF4E phosphorylation was mediated by p38 MAPKs. We also show that BETis suppressed RacGAP1 to induce Rac signaling-mediated eIF4E phosphorylation. Significantly, MNK inhibitors and MNK1/2 knockdown enhanced the efficacy of BETis in suppressing proliferation of cancer cells in vitro and in a syngeneic mouse model. Together, these results demonstrate a novel prosurvival feedback signaling induced by BETis, providing a mechanistic rationale for combination therapy with BET and MNK inhibitors for synergistic inhibition of cancer cells.
Collapse
Affiliation(s)
- Thao N D Pham
- Feinberg School of Medicine, Northwestern University, Chicago, Illinois.
| | - Krishan Kumar
- Feinberg School of Medicine, Northwestern University, Chicago, Illinois.,The Robert H. Lurie Comprehensive Cancer Center, Chicago, Illinois
| | - Brian T DeCant
- Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Meng Shang
- Feinberg School of Medicine, Northwestern University, Chicago, Illinois.,Jesse Brown VA Medical Center, Chicago, Illinois
| | - Samad Z Munshi
- Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Maria Matsangou
- Feinberg School of Medicine, Northwestern University, Chicago, Illinois.,The Robert H. Lurie Comprehensive Cancer Center, Chicago, Illinois
| | - Kazumi Ebine
- Feinberg School of Medicine, Northwestern University, Chicago, Illinois.,Jesse Brown VA Medical Center, Chicago, Illinois
| | - Hidayatullah G Munshi
- Feinberg School of Medicine, Northwestern University, Chicago, Illinois. .,The Robert H. Lurie Comprehensive Cancer Center, Chicago, Illinois.,Jesse Brown VA Medical Center, Chicago, Illinois
| |
Collapse
|
36
|
Ernlund AW, Schneider RJ, Ruggles KV. RIVET: comprehensive graphic user interface for analysis and exploration of genome-wide translatomics data. BMC Genomics 2018; 19:809. [PMID: 30409155 PMCID: PMC6225633 DOI: 10.1186/s12864-018-5166-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Accepted: 10/15/2018] [Indexed: 11/29/2022] Open
Abstract
Background Translatomics data, particularly genome-wide ribosome profiling and polysome profiling, provide multiple levels of gene regulatory information that can be used to assess general transcription and translation, as well translational efficiency. The increasing popularity of these techniques has resulted in multiple algorithms to detect translational regulation, typically distributed in the form of command line tools that require a basic level of programming ability. Additionally, due to the static nature of current software, dynamic transcriptional and translational comparative analysis cannot be adequately achieved. In order to streamline hypothesis generation, investigators must have the ability to manipulate and interact with their data in real-time. Results To address the lack of integration in current software, we introduce RIVET, Ribosomal Investigation and Visualization to Evaluate Translation, an R shiny based graphical user interface for translatomics data exploration and differential analysis. RIVET can analyze either microarray or RNA sequencing data from polysome profiling and ribosome profiling experiments. RIVET provides multiple choices for statistical analysis as well as integration of transcription, translation, and translational efficiency data analytics and the ability to visualize all results dynamically. Conclusions RIVET is a user-friendly tool designed for bench scientists with little to no programming background. RIVET facilitates the data analysis of translatomics data allowing for dynamic generation of results based on user-defined inputs and publication ready visualization. We expect RIVET will allow for scientists to efficiently make more comprehensive data observations that will lead to more robust hypothesis regarding translational regulation. Electronic supplementary material The online version of this article (10.1186/s12864-018-5166-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Amanda W Ernlund
- Department of Microbiology, New York University School of Medicine, New York, 10016, USA
| | - Robert J Schneider
- Department of Microbiology, New York University School of Medicine, New York, 10016, USA.
| | - Kelly V Ruggles
- Department of Medicine, Department of Translational Medicine, New York University School of Medicine, New York, 10016, USA.
| |
Collapse
|
37
|
Zakaria C, Sean P, Hoang HD, Leroux LP, Watson M, Workenhe ST, Hearnden J, Pearl D, Truong VT, Robichaud N, Yanagiya A, Tahmasebi S, Jafarnejad SM, Jia JJ, Pelin A, Diallo JS, Le Boeuf F, Bell JC, Mossman KL, Graber TE, Jaramillo M, Sonenberg N, Alain T. Active-site mTOR inhibitors augment HSV1-dICP0 infection in cancer cells via dysregulated eIF4E/4E-BP axis. PLoS Pathog 2018; 14:e1007264. [PMID: 30138450 PMCID: PMC6124814 DOI: 10.1371/journal.ppat.1007264] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Revised: 09/05/2018] [Accepted: 08/07/2018] [Indexed: 12/21/2022] Open
Abstract
Herpes Simplex Virus 1 (HSV1) is amongst the most clinically advanced oncolytic virus platforms. However, efficient and sustained viral replication within tumours is limiting. Rapamycin can stimulate HSV1 replication in cancer cells, but active-site dual mTORC1 and mTORC2 (mammalian target of rapamycin complex 1 and 2) inhibitors (asTORi) were shown to suppress the virus in normal cells. Surprisingly, using the infected cell protein 0 (ICP0)-deleted HSV1 (HSV1-dICP0), we found that asTORi markedly augment infection in cancer cells and a mouse mammary cancer xenograft. Mechanistically, asTORi repressed mRNA translation in normal cells, resulting in defective antiviral response but also inhibition of HSV1-dICP0 replication. asTORi also reduced antiviral response in cancer cells, however in contrast to normal cells, transformed cells and cells transduced to elevate the expression of eukaryotic initiation factor 4E (eIF4E) or to silence the repressors eIF4E binding proteins (4E-BPs), selectively maintained HSV1-dICP0 protein synthesis during asTORi treatment, ultimately supporting increased viral replication. Our data show that altered eIF4E/4E-BPs expression can act to promote HSV1-dICP0 infection under prolonged mTOR inhibition. Thus, pharmacoviral combination of asTORi and HSV1 can target cancer cells displaying dysregulated eIF4E/4E-BPs axis. Dysregulated mRNA translation occurs frequently in tumours due to elevated eIF4E expression or a hyperactive mTOR complex 1 (mTORC1) signaling pathway that results in the inactivation of the eIF4E binding proteins (4E-BPs). Targeting the mTORC1/4E-BPs/eIF4E axis is a promising strategy in cancer therapies and for preventing resistance to treatment. Enhanced mTORC1 activity also drives innate immune responses by modulating protein expression of antiviral genes. It was previously shown that the mTORC1 inhibitor rapamycin limits antiviral responses and promotes replication of oncolytic viruses within tumour tissues. Active-site dual mTORC1 and mTORC2 inhibitors (asTORi) have been developed for superior mTOR inhibition and anti-cancer potency but have not been studied in the context of oncolytic viral infection. We show here that prolonged treatment with asTORi strongly augments infection of HSV1-dICP0 in cancer cells, but not in normal cells, an effect modulated via eIF4E/4E-BP expression. Thus, cancer cells with dysregulated translation could be amenable to the pharmacoviral combination of HSV1 and asTORi treatment.
Collapse
Affiliation(s)
- Chadi Zakaria
- Goodman Cancer Centre, Department of Biochemistry, McGill University, Montreal, Canada
| | - Polen Sean
- Goodman Cancer Centre, Department of Biochemistry, McGill University, Montreal, Canada
| | - Huy-Dung Hoang
- Children's Hospital of Eastern Ontario Research Institute, Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Ontario, Canada
| | | | - Margaret Watson
- Children's Hospital of Eastern Ontario Research Institute, Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Ontario, Canada
| | - Samuel Tekeste Workenhe
- Department of Pathology and Molecular Medicine, MG DeGroote Institute for Infectious Disease, McMaster University, Hamilton, Ontario, Canada
| | - Jaclyn Hearnden
- Goodman Cancer Centre, Department of Biochemistry, McGill University, Montreal, Canada
| | - Dana Pearl
- Goodman Cancer Centre, Department of Biochemistry, McGill University, Montreal, Canada
| | - Vinh Tai Truong
- Goodman Cancer Centre, Department of Biochemistry, McGill University, Montreal, Canada
| | - Nathaniel Robichaud
- Goodman Cancer Centre, Department of Biochemistry, McGill University, Montreal, Canada
| | - Akiko Yanagiya
- Goodman Cancer Centre, Department of Biochemistry, McGill University, Montreal, Canada
| | - Soroush Tahmasebi
- Goodman Cancer Centre, Department of Biochemistry, McGill University, Montreal, Canada
| | | | - Jian-Jun Jia
- Children's Hospital of Eastern Ontario Research Institute, Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Ontario, Canada
| | - Adrian Pelin
- Center for Innovative Cancer Research, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Jean-Simon Diallo
- Center for Innovative Cancer Research, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Fabrice Le Boeuf
- Center for Innovative Cancer Research, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - John Cameron Bell
- Center for Innovative Cancer Research, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Karen Louise Mossman
- Department of Pathology and Molecular Medicine, MG DeGroote Institute for Infectious Disease, McMaster University, Hamilton, Ontario, Canada
| | - Tyson Ernst Graber
- Children's Hospital of Eastern Ontario Research Institute, Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Ontario, Canada
| | - Maritza Jaramillo
- INRS Institut Armand-Frappier Research Centre, Laval, Quebec, Canada
| | - Nahum Sonenberg
- Goodman Cancer Centre, Department of Biochemistry, McGill University, Montreal, Canada
- * E-mail: (NS); (TA)
| | - Tommy Alain
- Children's Hospital of Eastern Ontario Research Institute, Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Ontario, Canada
- * E-mail: (NS); (TA)
| |
Collapse
|
38
|
de la Parra C, Ernlund A, Alard A, Ruggles K, Ueberheide B, Schneider RJ. A widespread alternate form of cap-dependent mRNA translation initiation. Nat Commun 2018; 9:3068. [PMID: 30076308 PMCID: PMC6076257 DOI: 10.1038/s41467-018-05539-0] [Citation(s) in RCA: 109] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Accepted: 07/06/2018] [Indexed: 11/30/2022] Open
Abstract
Translation initiation of most mammalian mRNAs is mediated by a 5′ cap structure that binds eukaryotic initiation factor 4E (eIF4E). However, inactivation of eIF4E does not impair translation of many capped mRNAs, suggesting an unknown alternate mechanism may exist for cap-dependent but eIF4E-independent translation. We show that DAP5, an eIF4GI homolog that lacks eIF4E binding, utilizes eIF3d to facilitate cap-dependent translation of approximately 20% of mRNAs. Genome-wide transcriptomic and translatomic analyses indicate that DAP5 is required for translation of many transcription factors and receptor capped mRNAs and their mRNA targets involved in cell survival, motility, DNA repair and translation initiation, among other mRNAs. Mass spectrometry and crosslinking studies demonstrate that eIF3d is a direct binding partner of DAP5. In vitro translation and ribosome complex studies demonstrate that DAP5 and eIF3d are both essential for eIF4E-independent capped-mRNA translation. These studies disclose a widespread and previously unknown mechanism for cap-dependent mRNA translation by DAP5-eIF3d complexes. Binding of eIF4E to the 5′ cap of mRNAs is a key early step in canonical translation initiation, but the requirement for eIF4E is not universal. Here the authors show that the eIF4G homolog DAP5 interacts with eIF3 to promote cap-dependent translation of a significant number of mRNA in an eIF4E-independent manner.
Collapse
Affiliation(s)
- Columba de la Parra
- Department of Microbiology, NYU School of Medicine, New York, NY, 10016, USA.,Perlmutter Cancer Center, NYU School of Medicine, New York, NY, 10016, USA
| | - Amanda Ernlund
- Department of Microbiology, NYU School of Medicine, New York, NY, 10016, USA
| | - Amandine Alard
- Department of Microbiology, NYU School of Medicine, New York, NY, 10016, USA
| | - Kelly Ruggles
- Department of Medicine, NYU School of Medicine, New York, NY, 10016, USA
| | - Beatrix Ueberheide
- Department of Biochemistry and Molecular Pharmacology, NYU School of Medicine, New York, NY, 10016, USA
| | - Robert J Schneider
- Department of Microbiology, NYU School of Medicine, New York, NY, 10016, USA. .,Perlmutter Cancer Center, NYU School of Medicine, New York, NY, 10016, USA.
| |
Collapse
|
39
|
Peters C, Paget M, Tshilenge KT, Saha D, Antoszczyk S, Baars A, Frost T, Martuza RL, Wakimoto H, Rabkin SD. Restriction of Replication of Oncolytic Herpes Simplex Virus with a Deletion of γ34.5 in Glioblastoma Stem-Like Cells. J Virol 2018; 92:e00246-18. [PMID: 29793956 PMCID: PMC6052301 DOI: 10.1128/jvi.00246-18] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Accepted: 05/15/2018] [Indexed: 12/12/2022] Open
Abstract
Oncolytic viruses, including herpes simplex viruses (HSVs), are a new class of cancer therapeutic engineered to infect and kill cancer cells while sparing normal tissue. To ensure that oncolytic HSV (oHSV) is safe in the brain, all oHSVs in clinical trial for glioma lack the γ34.5 genes responsible for neurovirulence. However, loss of γ34.5 attenuates growth in cancer cells. Glioblastoma (GBM) is a lethal brain tumor that is heterogeneous and contains a subpopulation of cancer stem cells, termed GBM stem-like cells (GSCs), that likely promote tumor progression and recurrence. GSCs and matched serum-cultured GBM cells (ScGCs), representative of bulk or differentiated tumor cells, were isolated from the same patient tumor specimens. ScGCs are permissive to replication and cell killing by oHSV with deletion of the γ34.5 genes (γ34.5- oHSV), while patient-matched GSCs were not, implying an underlying biological difference between stem and bulk cancer cells. GSCs specifically restrict the synthesis of HSV-1 true late (TL) proteins, without affecting viral DNA replication or transcription of TL genes. A global shutoff of cellular protein synthesis also occurs late after γ34.5- oHSV infection of GSCs but does not affect the synthesis of early and leaky late viral proteins. Levels of phosphorylated eIF2α and eIF4E do not correlate with cell permissivity. Expression of Us11 in GSCs rescues replication of γ34.5- oHSV. The difference in degrees of permissivity between GSCs and ScGCs to γ34.5- oHSV illustrates a selective translational regulatory pathway in GSCs that may be operative in other stem-like cells and has implications for creating oHSVs.IMPORTANCE Herpes simplex virus (HSV) can be genetically engineered to endow cancer-selective replication and oncolytic activity. γ34.5, a key neurovirulence gene, has been deleted in all oncolytic HSVs in clinical trial for glioma. Glioblastoma stem-like cells (GSCs) are a subpopulation of tumor cells thought to drive tumor heterogeneity and therapeutic resistance. GSCs are nonpermissive for γ34.5- HSV, while non-stem-like cancer cells from the same patient tumors are permissive. GSCs restrict true late protein synthesis, despite normal viral DNA replication and transcription of all kinetic classes. This is specific for true late translation as early and leaky late transcripts are translated late in infection, notwithstanding shutoff of cellular protein synthesis. Expression of Us11 in GSCs rescues the replication of γ34.5- HSV. We have identified a cell type-specific innate response to HSV-1 that limits oncolytic activity in glioblastoma.
Collapse
Affiliation(s)
- Cole Peters
- Brain Tumor Research Center, Department of Neurosurgery, Massachusetts General Hospital, Boston, Massachusetts, USA
- Program in Virology, Harvard Medical School, Boston, Massachusetts, USA
| | - Max Paget
- Brain Tumor Research Center, Department of Neurosurgery, Massachusetts General Hospital, Boston, Massachusetts, USA
- Program in Virology, Harvard Medical School, Boston, Massachusetts, USA
| | - Kizito-Tshitoko Tshilenge
- Brain Tumor Research Center, Department of Neurosurgery, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Dipongkor Saha
- Brain Tumor Research Center, Department of Neurosurgery, Massachusetts General Hospital, Boston, Massachusetts, USA
- Department of Neurosurgery, Harvard Medical School, Boston, Massachusetts, USA
| | - Slawomir Antoszczyk
- Brain Tumor Research Center, Department of Neurosurgery, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Anouk Baars
- Brain Tumor Research Center, Department of Neurosurgery, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Thomas Frost
- Brain Tumor Research Center, Department of Neurosurgery, Massachusetts General Hospital, Boston, Massachusetts, USA
- Program in Virology, Harvard Medical School, Boston, Massachusetts, USA
| | - Robert L Martuza
- Brain Tumor Research Center, Department of Neurosurgery, Massachusetts General Hospital, Boston, Massachusetts, USA
- Department of Neurosurgery, Harvard Medical School, Boston, Massachusetts, USA
| | - Hiroaki Wakimoto
- Brain Tumor Research Center, Department of Neurosurgery, Massachusetts General Hospital, Boston, Massachusetts, USA
- Department of Neurosurgery, Harvard Medical School, Boston, Massachusetts, USA
| | - Samuel D Rabkin
- Brain Tumor Research Center, Department of Neurosurgery, Massachusetts General Hospital, Boston, Massachusetts, USA
- Program in Virology, Harvard Medical School, Boston, Massachusetts, USA
- Department of Neurosurgery, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|