201
|
Teng YC, Neo JC, Wu JC, Chen YF, Kao CH, Tsai TF. Expression of a hepatitis B virus pre-S2 deletion mutant in the liver results in hepatomegaly and hepatocellular carcinoma in mice. J Pathol 2017; 241:463-474. [PMID: 27868197 DOI: 10.1002/path.4850] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2016] [Revised: 10/07/2016] [Accepted: 11/09/2016] [Indexed: 12/18/2022]
Abstract
Hepatocellular carcinoma (HCC) is the most common form of liver cancer and has a poor prognosis and a low survival rate; its incidence is on the rise. Hepatitis B virus (HBV) infection is one of the main causes of HCC. A high prevalence of pre-S deletions of HBV surface antigen, which encompass T-cell and/or B-cell epitopes, is found in HBV carriers; antiviral therapy and viral immune escape may cause and select for these HBV mutants. In particular, the presence of pre-S2 deletion mutants is an important risk factor associated with cirrhosis and HCC. We generated Alb-preΔS2 transgenic mice that express a naturally occurring pre-S2 mutant protein containing a 33-nucleotide deletion (preΔS2); the aim was to investigate its effect on hepatocarcinogenesis. After 30 months of follow-up, the liver pathology of the mice fell into four groups: G1, chronic inflammation solely; G2, chronic inflammation and fibrosis; G3, inflammation, fibrosis, and hepatomegaly accompanied by rectal prolapse (4-12%); and G4, hepatomegaly and spontaneous HCC (12-15%). Striking degeneration of the endoplasmic reticulum (ER) was present in the mouse livers at an early stage (4 months old). At 8 months, overt ER stress and the Atf6 pathway of the unfolded protein response (UPR) were induced; at the same time, metabolic pathways associated with mevalonate and cholesterol biogenesis, involving the peroxisomes and the ER, were disturbed. At 20 months and older, the protein kinase RNA-like endoplasmic reticulum kinase (PERK) pathway of the UPR was induced and the Hippo transducer Yap was activated. Together, these ultrastructural aberrations and metabolic disturbance all seem to contribute to the molecular pathogenesis and hepatocarcinogenesis present in the Alb-preΔS2 mice. These findings may contribute to the development of therapies for the liver disorders and HCC associated with pre-S2 deletion mutations among HBV carriers. Copyright © 2016 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Yuan-Chi Teng
- Program in Molecular Medicine, School of Life Sciences, National Yang-Ming University and Academia Sinica, Taipei, Taiwan.,Department of Life Sciences and Institute of Genome Sciences, National Yang-Ming University, Taipei, Taiwan
| | - Jenq Chyuan Neo
- Department of Life Sciences and Institute of Genome Sciences, National Yang-Ming University, Taipei, Taiwan
| | - Jaw-Ching Wu
- Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan.,Translational Research Division, Medical Research Department, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Yi-Fan Chen
- The PhD Program for Translational Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Cheng-Heng Kao
- Center of General Education, Chang Gung University, Taoyuan, Taiwan
| | - Ting-Fen Tsai
- Program in Molecular Medicine, School of Life Sciences, National Yang-Ming University and Academia Sinica, Taipei, Taiwan.,Department of Life Sciences and Institute of Genome Sciences, National Yang-Ming University, Taipei, Taiwan.,Genome Research Center, National Yang-Ming University, Taipei, Taiwan.,Aging and Health Research Center, National Yang-Ming University, Taipei, Taiwan.,Institute of Molecular and Genomic Medicine, National Health Research Institutes, Zhunan, Taiwan
| |
Collapse
|
202
|
García-Ruiz C, Ribas V, Baulies A, Fernández-Checa JC. Mitochondrial Cholesterol and the Paradox in Cell Death. Handb Exp Pharmacol 2017; 240:189-210. [PMID: 28035533 DOI: 10.1007/164_2016_110] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Mitochondria are considered cholesterol-poor organelles, and obtain their cholesterol load by the action of specialized proteins involved in its delivery from extramitochondrial sources and trafficking within mitochondrial membranes. Although mitochondrial cholesterol fulfills vital physiological functions, such as the synthesis of bile acids in the liver or the formation of steroid hormones in specialized tissues, recent evidence indicates that the accumulation of cholesterol in mitochondria may be a key event in prevalent human diseases, in particular in the development of steatohepatitis (SH) and its progression to hepatocellular carcinoma (HCC). Mitochondrial cholesterol accumulation promotes the transition from simple steatosis to SH due to the sensitization to oxidative stress and cell death. However, mitochondrial cholesterol loading in HCC determines apoptosis resistance and insensitivity to chemotherapy. These opposing functions of mitochondrial cholesterol in SH and HCC define its paradoxical role in cell death as a pro- and anti-apoptotic factor. Further understanding of this conundrum may be useful to modulate the progression from SH to HCC by targeting mitochondrial cholesterol trafficking.
Collapse
Affiliation(s)
- Carmen García-Ruiz
- Department of Cell Death and Proliferation, Instituto Investigaciones Biomedicas de Barcelona, CSIC, C/Rosello 161, 08036, Barcelona, Spain
- Liver Unit, Hospital Clinic, IDIBAPS, Barcelona, Spain
- Centro de Investigación Biomédica en Red (CIBERehd), Barcelona, Spain
- Keck School of Medicine, USC, University of Southern California Research Center for Alcohol Liver and Pancreatic Diseases and Cirrhosis, Los Angeles, CA, USA
| | - Vicente Ribas
- Department of Cell Death and Proliferation, Instituto Investigaciones Biomedicas de Barcelona, CSIC, C/Rosello 161, 08036, Barcelona, Spain
- Liver Unit, Hospital Clinic, IDIBAPS, Barcelona, Spain
- Centro de Investigación Biomédica en Red (CIBERehd), Barcelona, Spain
| | - Anna Baulies
- Department of Cell Death and Proliferation, Instituto Investigaciones Biomedicas de Barcelona, CSIC, C/Rosello 161, 08036, Barcelona, Spain
- Liver Unit, Hospital Clinic, IDIBAPS, Barcelona, Spain
- Centro de Investigación Biomédica en Red (CIBERehd), Barcelona, Spain
| | - Jose C Fernández-Checa
- Department of Cell Death and Proliferation, Instituto Investigaciones Biomedicas de Barcelona, CSIC, C/Rosello 161, 08036, Barcelona, Spain.
- Liver Unit, Hospital Clinic, IDIBAPS, Barcelona, Spain.
- Centro de Investigación Biomédica en Red (CIBERehd), Barcelona, Spain.
- Keck School of Medicine, USC, University of Southern California Research Center for Alcohol Liver and Pancreatic Diseases and Cirrhosis, Los Angeles, CA, USA.
| |
Collapse
|
203
|
Halim L, Parente-Pereira AC, Maher J. Prospects for immunotherapy of acute myeloid leukemia using γδ T cells. Immunotherapy 2017; 9:111-114. [PMID: 28128710 DOI: 10.2217/imt-2016-0139] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
MESH Headings
- Animals
- Antigens, Neoplasm/immunology
- Antigens, Viral/immunology
- Cancer Vaccines/immunology
- Cross Reactions
- Cytotoxicity, Immunologic
- Humans
- Immunotherapy, Adoptive/methods
- Immunotherapy, Adoptive/trends
- Interleukin-17/metabolism
- Leukemia, Myeloid, Acute/immunology
- Leukemia, Myeloid, Acute/therapy
- Lymphocyte Activation
- Lymphocytes, Tumor-Infiltrating/immunology
- Receptors, Antigen, T-Cell, gamma-delta/metabolism
- T-Lymphocytes/immunology
- T-Lymphocytes/transplantation
- Tumor Microenvironment
Collapse
Affiliation(s)
- Leena Halim
- CAR Mechanics Group, Division of Cancer Studies, King's College London, Guy's Hospital Campus, Great Maze Pond, London SE1 9RT, UK
| | - Ana Catarina Parente-Pereira
- CAR Mechanics Group, Division of Cancer Studies, King's College London, Guy's Hospital Campus, Great Maze Pond, London SE1 9RT, UK
| | - John Maher
- CAR Mechanics Group, Division of Cancer Studies, King's College London, Guy's Hospital Campus, Great Maze Pond, London SE1 9RT, UK
- Department of Clinical Immunology & Allergy, King's College Hospital NHS Foundation Trust, London, UK
- Department of Immunology, Eastbourne Hospital, East Sussex, UK
| |
Collapse
|
204
|
Tanaka K, Osada H, Murakami-Tonami Y, Horio Y, Hida T, Sekido Y. Statin suppresses Hippo pathway-inactivated malignant mesothelioma cells and blocks the YAP/CD44 growth stimulatory axis. Cancer Lett 2017; 385:215-224. [DOI: 10.1016/j.canlet.2016.10.020] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Revised: 09/28/2016] [Accepted: 10/10/2016] [Indexed: 12/12/2022]
|
205
|
Holic N, Frin S, Seye AK, Galy A, Fenard D. Improvement of De Novo Cholesterol Biosynthesis Efficiently Promotes the Production of Human Immunodeficiency Virus Type 1-Derived Lentiviral Vectors. Hum Gene Ther Methods 2016; 28:67-77. [PMID: 28042946 DOI: 10.1089/hgtb.2016.150] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The use of lentiviral vectors (LVs) for gene transfer in research, technological, or clinical applications requires the production of large amounts of vector. Mass production of clinical-grade LVs remains a challenge and limits certain perspectives for therapeutic use. Some improvements in LV production protocols have been possible by acting on multiple steps of the production process. The addition of animal-derived cholesterol to the culture medium of producer cells is known to increase the infectivity of LVs. To avoid the use of this animal-derived product in clinical settings, an alternative approach is to increase de novo the production of cholesterol by overexpressing a crucial cholesterogenic enzyme, namely, 3-hydroxy-3-methylglutaryl-coenzyme A reductase (HMGCR). This project evaluates the impact of such an approach on the production, infectivity, and stability of LVs. We demonstrated that the overexpression of human HMGCR isoform 1 (hHMGCR1) in LV producer cells efficiently increased de novo cholesterol biosynthesis and enhanced by 2- to 3-fold the physical and infectious titers of LVs. We also observed that LVs produced in hHMGCR1-overexpressing cells were comparable in stability to LVs produced under classical conditions and were capable of transducing human CD34+ hematopoietic stem/progenitor cells efficiently. Interestingly, we also showed that LV production in the absence of fetal calf serum (FCS) but under hHMGCR1-overexpressing conditions allowed a viral production yield comparable to that achieved under classical conditions in high FCS content, leading the way to the establishment of new LV production protocols on adherent cells without serum.
Collapse
Affiliation(s)
- Nathalie Holic
- 1 Généthon , Evry, France
- 2 INSERM , UMR_S951, Généthon, Evry, France
- 3 Université Evry Val d'Essonne , UMR_S951, Evry, France
| | - Sophie Frin
- 1 Généthon , Evry, France
- 2 INSERM , UMR_S951, Généthon, Evry, France
| | - Ababacar K Seye
- 1 Généthon , Evry, France
- 2 INSERM , UMR_S951, Généthon, Evry, France
| | - Anne Galy
- 1 Généthon , Evry, France
- 2 INSERM , UMR_S951, Généthon, Evry, France
- 3 Université Evry Val d'Essonne , UMR_S951, Evry, France
| | - David Fenard
- 1 Généthon , Evry, France
- 2 INSERM , UMR_S951, Généthon, Evry, France
- 3 Université Evry Val d'Essonne , UMR_S951, Evry, France
| |
Collapse
|
206
|
Wang T, Seah S, Loh X, Chan CW, Hartman M, Goh BC, Lee SC. Simvastatin-induced breast cancer cell death and deactivation of PI3K/Akt and MAPK/ERK signalling are reversed by metabolic products of the mevalonate pathway. Oncotarget 2016; 7:2532-44. [PMID: 26565813 PMCID: PMC4823053 DOI: 10.18632/oncotarget.6304] [Citation(s) in RCA: 95] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Accepted: 10/14/2015] [Indexed: 02/07/2023] Open
Abstract
Statins purportedly exert anti-tumoral effects on breast cancer. However, the biologic mechanisms for these actions are not fully elucidated. The aims of this study were 1) to explore the effects of simvastatin on apoptosis, proliferation as well as PI3K/Akt/mTOR and MAPK/ERK pathway in a window-of-opportunity breast cancer trial; 2) to further confirm findings from the clinical trial by functional studies; 3) to explore the regulatory role of mevalonate pathway on the anti-tumoral effects of simvastatin. In clinical samples, simvastatin led to increase in cleaved caspase-3 (p = 0.002) and decreased trend for Ki67 (p = 0.245). Simvastatin markedly suppressed PI3K/Akt/mTOR signalling by activating PTEN (p = 0.005) and by dephosphorylating Akt (p = 0.002) and S6RP (p = 0.033); it also inhibited MAPK/ERK pathway by dephosphorylating c-Raf (p = 0.018) and ERK1/2 (p = 0.002). In ER-positive (MCF-7, T47D) and ER-negative (MDA-MB-231, BT-549) breast cancer cells, simvastatin treatment consistently induced apoptosis and inhibited proliferation by deregulating caspase cascades and cell cycle proteins in a dose dependent manner. Concordantly, simvastatin strongly suppressed PI3K/Akt/mTOR pathway by enhancing PTEN expression and by further sequentially dephosphorylating downstream cascades including Akt, mTOR, p70S6K, S6RP and 4E-BP1. Furthermore, simvastatin significantly inhibited MAPK/ERK pathway by dephosphorylating sequential cascades such as c-Raf, MEK1/2 and ERK1/2. These simvastatin anti-tumoral effects were reversed by metabolic products of the mevalonate pathway, including mevalonate, farnesyl pyrophosphate and geranylgeranyl pyrophosphate. These findings shed light on the biological and potential anti-tumoral effects of simvastatin in breast cancer.
Collapse
Affiliation(s)
- Tingting Wang
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Serena Seah
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Xinyi Loh
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Ching-Wan Chan
- Department of Surgery, National University Cancer Institute, National University Health System, Singapore, Singapore
| | - Mikael Hartman
- Department of Surgery, National University Cancer Institute, National University Health System, Singapore, Singapore
| | - Boon-Cher Goh
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore.,Department of Haematology and Oncology, National University Cancer Institute, National University Health System, Singapore, Singapore
| | - Soo-Chin Lee
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore.,Department of Haematology and Oncology, National University Cancer Institute, National University Health System, Singapore, Singapore
| |
Collapse
|
207
|
Sakellakis M, Akinosoglou K, Kostaki A, Spyropoulou D, Koutras A. Statins and risk of breast cancer recurrence. BREAST CANCER-TARGETS AND THERAPY 2016; 8:199-205. [PMID: 27853392 PMCID: PMC5104306 DOI: 10.2147/bctt.s116694] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Background The primary end point of our study was to test whether the concurrent use of a statin is related to a lower risk of recurrence and increased relapse-free survival in patients with early breast cancer. Materials and methods We reviewed 610 female patients with stage I, II, or III breast cancer who had been surgically treated and who had subsequently received at least adjuvant chemotherapy in order to prevent recurrence. Results Among the 610 patients with breast cancer, 83 (13.6%) were receiving a statin on a chronic basis for other medical purposes. Overall, statin users displayed longer mean relapse-free survival (16.6 vs 10.2 years, P=0.028). After data had been adjusted for patient and disease characteristics, statin users maintained a lower risk of recurrence. This favorable outcome in statin users was particularly evident when we included only younger patients in the analysis (20 vs 10 years, P=0.006). Conclusion Statins may be linked to a favorable outcome in early breast cancer patients, especially in younger age-groups.
Collapse
Affiliation(s)
- Minas Sakellakis
- Department of Medicine, Division of Oncology, University Hospital, Patras Medical School, Patras
| | - Karolina Akinosoglou
- Department of Medicine, Division of Oncology, University Hospital, Patras Medical School, Patras
| | - Anastasia Kostaki
- Department of Statistics, Athens University of Economics and Business, Athens, Greece
| | - Despina Spyropoulou
- Department of Medicine, Division of Oncology, University Hospital, Patras Medical School, Patras
| | - Angelos Koutras
- Department of Medicine, Division of Oncology, University Hospital, Patras Medical School, Patras
| |
Collapse
|
208
|
Mullen PJ, Yu R, Longo J, Archer MC, Penn LZ. The interplay between cell signalling and the mevalonate pathway in cancer. Nat Rev Cancer 2016; 16:718-731. [PMID: 27562463 DOI: 10.1038/nrc.2016.76] [Citation(s) in RCA: 477] [Impact Index Per Article: 53.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The mevalonate (MVA) pathway is an essential metabolic pathway that uses acetyl-CoA to produce sterols and isoprenoids that are integral to tumour growth and progression. In recent years, many oncogenic signalling pathways have been shown to increase the activity and/or the expression of MVA pathway enzymes. This Review summarizes recent advances and discusses unique opportunities for immediately targeting this metabolic vulnerability in cancer with agents that have been approved for other therapeutic uses, such as the statin family of drugs, to improve outcomes for cancer patients.
Collapse
Affiliation(s)
- Peter J Mullen
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada M5G 1L7
| | - Rosemary Yu
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada M5G 1L7
- Department of Medical Biophysics, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada M5G 1L7
| | - Joseph Longo
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada M5G 1L7
- Department of Medical Biophysics, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada M5G 1L7
| | - Michael C Archer
- Department of Medical Biophysics, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada M5G 1L7
- Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada M5S 3E2
| | - Linda Z Penn
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada M5G 1L7
- Department of Medical Biophysics, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada M5G 1L7
| |
Collapse
|
209
|
Reed GA, Schiller GJ, Kambhampati S, Tallman MS, Douer D, Minden MD, Yee KW, Gupta V, Brandwein J, Jitkova Y, Gronda M, Hurren R, Shamas-Din A, Schuh AC, Schimmer AD. A Phase 1 study of intravenous infusions of tigecycline in patients with acute myeloid leukemia. Cancer Med 2016; 5:3031-3040. [PMID: 27734609 PMCID: PMC5119957 DOI: 10.1002/cam4.845] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Revised: 06/07/2016] [Accepted: 07/08/2016] [Indexed: 01/27/2023] Open
Abstract
Acute myeloid leukemia (AML) cells meet the higher energy, metabolic, and signaling demands of the cell by increasing mitochondrial biogenesis and mitochondrial protein translation. Blocking mitochondrial protein synthesis through genetic and chemical approaches kills human AML cells at all stages of development in vitro and in vivo. Tigecycline is an antimicrobial that we found inhibits mitochondrial protein synthesis in AML cells. Therefore, we conducted a phase 1 dose‐escalation study of tigecycline administered intravenously daily 5 of 7 days for 2 weeks to patients with AML. A total of 27 adult patients with relapsed and refractory AML were enrolled in this study with 42 cycles being administered over seven dose levels (50–350 mg/day). Two patients experienced DLTs related to tigecycline at the 350 mg/day level resulting in a maximal tolerated dose of tigecycline of 300 mg as a once daily infusion. Pharmacokinetic experiments showed that tigecycline had a markedly shorter half‐life in these patients than reported for noncancer patients. No significant pharmacodynamic changes or clinical responses were observed. Thus, we have defined the safety of once daily tigecycline in patients with refractory AML. Future studies should focus on schedules of the drug that permit more sustained target inhibition.
Collapse
Affiliation(s)
| | - Gary J Schiller
- David Geffen School of Medicine at UCLA, Los Angeles, California
| | | | - Martin S Tallman
- Leukemia Service, Department of Medicine, Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Dan Douer
- Leukemia Service, Department of Medicine, Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Mark D Minden
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Karen W Yee
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Vikas Gupta
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Joseph Brandwein
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada.,Department of Medicine, University of Alberta, Edmonton, Alberta, Canada
| | - Yulia Jitkova
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Marcela Gronda
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Rose Hurren
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Aisha Shamas-Din
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Andre C Schuh
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Aaron D Schimmer
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| |
Collapse
|
210
|
Statin use and breast cancer survival and risk: a systematic review and meta-analysis. Oncotarget 2016; 6:42988-3004. [PMID: 26472026 PMCID: PMC4767486 DOI: 10.18632/oncotarget.5557] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Accepted: 09/06/2015] [Indexed: 12/16/2022] Open
Abstract
The purpose of this study is to determine the associations between statin use and breast cancer survival and risk by performing a systematic review and meta-analysis. We searched PubMed, Embase and Web of Science up to August 2015 for identifying relevant prospective or case-control studies, or randomized clinical trials. Five prospective studies involving 60,911 patients reported the association between statin use and breast cancer mortality. Eleven prospective studies, 12 case-control studies and 9 randomized clinical trials involving 83,919 patients reported the association between statin use and breast cancer risk. After pooling estimates from all available studies, there was a significantly negative association between pre-diagnosis statin use and breast cancer mortality (for overall survival (OS): hazard ratio (HR) = 0.68, 95% confidence interval (CI) 0.54–0.84; for disease specific survival (DSS): HR = 0.72, 95% CI 0.53–0.99). There was also a significant inverse association between post-diagnosis statin use and breast cancer DSS (HR = 0.65, 95% CI 0.43–0.98), although the association with breast cancer OS did not reach statistical significance (HR = 0.71, 95% CI 0.48–1.07). Additionally, there was a non-linear relationship for the duration of post-diagnosis statin use with breast cancer specific mortality. On the other hand, with regards to the relationship between statin use and breast cancer risk, no significant association was detected. Our analyses suggest that although statin use may not influence breast cancer risk, the use of statin may be associated with decrease mortality of breast cancer patients. Further large-scale studies are warranted to validate our findings.
Collapse
|
211
|
Goldman AR, Bitler BG, Schug Z, Conejo-Garcia JR, Zhang R, Speicher DW. The Primary Effect on the Proteome of ARID1A-mutated Ovarian Clear Cell Carcinoma is Downregulation of the Mevalonate Pathway at the Post-transcriptional Level. Mol Cell Proteomics 2016; 15:3348-3360. [PMID: 27654507 PMCID: PMC5098034 DOI: 10.1074/mcp.m116.062539] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Indexed: 12/24/2022] Open
Abstract
Inactivating mutations in ARID1A, which encodes a subunit of the SWI/SNF chromatin-remodeling complex, are found in over half of ovarian clear cell carcinoma cases and more broadly across most types of cancers. To identify ARID1A-dependent changes in intracellular signaling pathways, we performed proteome analyses of isogenic ovarian clear cell carcinoma cell lines with or without ARID1A expression. Knockout of ARID1A in an ovarian clear cell carcinoma cell line with wild-type ARID1A, OVCA429, primarily resulted in downregulation of the mevalonate pathway, an important metabolic pathway involved in isoprenoid synthesis, cholesterol synthesis, and other downstream pathways. In a complementary experiment, expression of wild-type ARID1A in an ovarian clear cell carcinoma cell line containing mutated ARID1A, OVISE, affected the mevalonate pathway in a reciprocal manner. A striking aspect of these analyses was that, although only 5% of the detected proteome showed significant abundance changes, most proteins in the mevalonate pathway were coordinately affected by ARID1A status. There were generally corresponding changes when comparing the proteomics data to our previously published microarray data for ectopic expression of ARID1A in the OVISE cell line. However, ARID1A-dependent changes were not detected for genes within the mevalonate pathway. This discrepancy suggests that the mevalonate pathway is not regulated directly by ARID1A-mediated transcription and may be regulated post-transcriptionally. We conclude that ARID1A status indirectly influences the mevalonate pathway and probably influences other processes including glycogen metabolism and 14-3-3-mediated signaling. Further, our findings demonstrate that changes in mRNA levels are sometimes poor indicators of signaling pathways affected by gene manipulations in cancer cells.
Collapse
Affiliation(s)
- Aaron R Goldman
- From the ‡Molecular and Cellular Oncogenesis Program, The Wistar Institute, 3601 Spruce St., Philadelphia, Pennsylvania 19104
| | - Benjamin G Bitler
- §Gene Expression and Regulation Program, The Wistar Institute, 3601 Spruce St., Philadelphia, Pennsylvania 19104
| | - Zachary Schug
- From the ‡Molecular and Cellular Oncogenesis Program, The Wistar Institute, 3601 Spruce St., Philadelphia, Pennsylvania 19104
| | - Jose R Conejo-Garcia
- ¶Tumor Microenvironment and Metastasis Program, The Wistar Institute, 3601 Spruce St., Philadelphia, Pennsylvania 19104
| | - Rugang Zhang
- §Gene Expression and Regulation Program, The Wistar Institute, 3601 Spruce St., Philadelphia, Pennsylvania 19104
| | - David W Speicher
- From the ‡Molecular and Cellular Oncogenesis Program, The Wistar Institute, 3601 Spruce St., Philadelphia, Pennsylvania 19104; .,‖The Center for Systems and Computational Biology, The Wistar Institute, 3601 Spruce St., Philadelphia, Pennsylvania 19104
| |
Collapse
|
212
|
Navarro-Imaz H, Rueda Y, Fresnedo O. SND1 overexpression deregulates cholesterol homeostasis in hepatocellular carcinoma. Biochim Biophys Acta Mol Cell Biol Lipids 2016; 1861:988-996. [DOI: 10.1016/j.bbalip.2016.05.011] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2015] [Revised: 05/18/2016] [Accepted: 05/24/2016] [Indexed: 01/06/2023]
|
213
|
Kopecka J, Porto S, Lusa S, Gazzano E, Salzano G, Giordano A, Desiderio V, Ghigo D, Caraglia M, De Rosa G, Riganti C. Self-assembling nanoparticles encapsulating zoledronic acid revert multidrug resistance in cancer cells. Oncotarget 2016; 6:31461-78. [PMID: 26372812 PMCID: PMC4741618 DOI: 10.18632/oncotarget.5058] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Accepted: 08/27/2015] [Indexed: 01/23/2023] Open
Abstract
The overexpression of ATP binding cassette (ABC) transporters makes tumor cells simultaneously resistant to several cytotoxic drugs. Impairing the energy metabolism of multidrug resistant (MDR) cells is a promising chemosensitizing strategy, but many metabolic modifiers are too toxic in vivo. We previously observed that the aminobisphosphonate zoledronic acid inhibits the activity of hypoxia inducible factor-1α (HIF-1α), a master regulator of cancer cell metabolism. Free zoledronic acid, however, reaches low intratumor concentration. We synthesized nanoparticle formulations of the aminobisphosphonate that allow a higher intratumor delivery of the drug. We investigated whether they are effective metabolic modifiers and chemosensitizing agents against human MDR cancer cells in vitro and in vivo. At not toxic dosage, nanoparticles carrying zoledronic acid chemosensitized MDR cells to a broad spectrum of cytotoxic drugs, independently of the type of ABC transporters expressed. The nanoparticles inhibited the isoprenoid synthesis and the Ras/ERK1/2-driven activation of HIF-1α, decreased the transcription and activity of glycolytic enzymes, the glucose flux through the glycolysis and tricarboxylic acid cycle, the electron flux through the mitochondrial respiratory chain, the synthesis of ATP. So doing, they lowered the ATP-dependent activity of ABC transporters, increasing the chemotherapy efficacy in vitro and in vivo. These effects were more pronounced in MDR cells than in chemosensitive ones and were due to the inhibition of farnesyl pyrophosphate synthase (FPPS), as demonstrated in FPPS-silenced tumors. Our work proposes nanoparticle formulations of zoledronic acid as the first not toxic metabolic modifiers, effective against MDR tumors.
Collapse
Affiliation(s)
- Joanna Kopecka
- Department of Oncology, University of Torino, Torino, Italy
| | - Stefania Porto
- Department of Biochemistry, Biophysics and General Pathology, Second University of Naples, Naples, Italy
| | - Sara Lusa
- Department of Pharmacy, Federico II University of Naples, Naples, Italy
| | - Elena Gazzano
- Department of Oncology, University of Torino, Torino, Italy
| | - Giuseppina Salzano
- Center for Pharmaceutical Biotechnology and Nanomedicine, Northeastern University, Boston, MA, USA
| | - Antonio Giordano
- Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, College of Science and Technology, Temple University, Philadelphia, PA, USA.,Department of Medicine, Surgery and Neuroscience University of Siena, Siena, Italy
| | - Vincenzo Desiderio
- Department of Experimental Medicine, Second University of Naples, Naples, Italy
| | - Dario Ghigo
- Department of Oncology, University of Torino, Torino, Italy
| | - Michele Caraglia
- Department of Biochemistry, Biophysics and General Pathology, Second University of Naples, Naples, Italy.,Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, College of Science and Technology, Temple University, Philadelphia, PA, USA
| | - Giuseppe De Rosa
- Department of Pharmacy, Federico II University of Naples, Naples, Italy
| | - Chiara Riganti
- Department of Oncology, University of Torino, Torino, Italy
| |
Collapse
|
214
|
Genome-wide RNAi analysis reveals that simultaneous inhibition of specific mevalonate pathway genes potentiates tumor cell death. Oncotarget 2016; 6:26909-21. [PMID: 26353928 PMCID: PMC4694962 DOI: 10.18632/oncotarget.4817] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Accepted: 08/12/2015] [Indexed: 01/03/2023] Open
Abstract
The mevalonate (MVA) pathway is often dysregulated or overexpressed in many cancers suggesting tumor dependency on this classic metabolic pathway. Statins, which target the rate-limiting enzyme of this pathway, 3-hydroxy-3-methylglutaryl-CoA reductase (HMGCR), are promising agents currently being evaluated in clinical trials for anti-cancer efficacy. To uncover novel targets that potentiate statin-induced apoptosis when knocked down, we carried out a pooled genome-wide short hairpin RNA (shRNA) screen. Genes of the MVA pathway were amongst the top-scoring targets, including sterol regulatory element binding transcription factor 2 (SREBP2), 3-hydroxy-3-methylglutaryl-coenzyme A synthase 1 (HMGCS1) and geranylgeranyl diphosphate synthase 1 (GGPS1). Each gene was independently validated and shown to significantly sensitize A549 cells to statin-induced apoptosis when knocked down. SREBP2 knockdown in lung and breast cancer cells completely abrogated the fluvastatin-induced upregulation of sterol-responsive genes HMGCR and HMGCS1. Knockdown of SREBP2 alone did not affect three-dimensional growth of lung and breast cancer cells, yet in combination with fluvastatin cell growth was disrupted. Taken together, these results show that directly targeting multiple levels of the MVA pathway, including blocking the sterol-feedback loop initiated by statin treatment, is an effective and targetable anti-tumor strategy.
Collapse
|
215
|
Agabiti SS, Liang Y, Wiemer AJ. Molecular mechanisms linking geranylgeranyl diphosphate synthase to cell survival and proliferation. Mol Membr Biol 2016; 33:1-11. [PMID: 27537059 DOI: 10.1080/09687688.2016.1213432] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Geranylgeranyl diphosphate is a 20-carbon isoprenoid phospholipid whose lipid moiety can be post-translationally incorporated into proteins to promote membrane association. The process of geranylgeranylation has been implicated in anti-proliferative effects of clinical agents that inhibit enzymes of the mevalonate pathway (i.e. statins and nitrogenous bisphosphonates) as well as experimental agents that deplete geranylgeranyl diphosphate. Inhibitors of geranylgeranyl diphosphate synthase are an attractive way to block geranylgeranylation because they possess a calcium-chelating substructure to allow localization to bone and take advantage of a unique position of the enzyme within the biosynthetic pathway. Here, we describe recent advances in geranylgeranyl diphosphate synthase expression and inhibitor development with a particular focus on the molecular mechanisms that link geranylgeranyl diphosphate to cell proliferation via geranylgeranylated small GTPases.
Collapse
Affiliation(s)
- Sherry S Agabiti
- a Department of Pharmaceutical Sciences , University of Connecticut , Storrs , CT , USA
| | - Yilan Liang
- a Department of Pharmaceutical Sciences , University of Connecticut , Storrs , CT , USA
| | - Andrew J Wiemer
- a Department of Pharmaceutical Sciences , University of Connecticut , Storrs , CT , USA.,b Institute for Systems Genomics, University of Connecticut , Storrs , CT , USA
| |
Collapse
|
216
|
Mitochondria, cholesterol and cancer cell metabolism. Clin Transl Med 2016; 5:22. [PMID: 27455839 PMCID: PMC4960093 DOI: 10.1186/s40169-016-0106-5] [Citation(s) in RCA: 111] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Accepted: 06/26/2016] [Indexed: 12/15/2022] Open
Abstract
Given the role of mitochondria in oxygen consumption, metabolism and cell death regulation, alterations in mitochondrial function or dysregulation of cell death pathways contribute to the genesis and progression of cancer. Cancer cells exhibit an array of metabolic transformations induced by mutations leading to gain-of-function of oncogenes and loss-of-function of tumor suppressor genes that include increased glucose consumption, reduced mitochondrial respiration, increased reactive oxygen species generation and cell death resistance, all of which ensure cancer progression. Cholesterol metabolism is disturbed in cancer cells and supports uncontrolled cell growth. In particular, the accumulation of cholesterol in mitochondria emerges as a molecular component that orchestrates some of these metabolic alterations in cancer cells by impairing mitochondrial function. As a consequence, mitochondrial cholesterol loading in cancer cells may contribute, in part, to the Warburg effect stimulating aerobic glycolysis to meet the energetic demand of proliferating cells, while protecting cancer cells against mitochondrial apoptosis due to changes in mitochondrial membrane dynamics. Further understanding the complexity in the metabolic alterations of cancer cells, mediated largely through alterations in mitochondrial function, may pave the way to identify more efficient strategies for cancer treatment involving the use of small molecules targeting mitochondria, cholesterol homeostasis/trafficking and specific metabolic pathways.
Collapse
|
217
|
Gruenbacher G, Gander H, Rahm A, Idzko M, Nussbaumer O, Thurnher M. Ecto-ATPase CD39 Inactivates Isoprenoid-Derived Vγ9Vδ2 T Cell Phosphoantigens. Cell Rep 2016; 16:444-456. [PMID: 27346340 DOI: 10.1016/j.celrep.2016.06.009] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2015] [Revised: 04/29/2016] [Accepted: 05/26/2016] [Indexed: 01/11/2023] Open
Abstract
In humans, Vγ9Vδ2 T cells respond to self and pathogen-associated, diphosphate-containing isoprenoids, also known as phosphoantigens (pAgs). However, activation and homeostasis of Vγ9Vδ2 T cells remain incompletely understood. Here, we show that pAgs induced expression of the ecto-ATPase CD39, which, however, not only hydrolyzed ATP but also abrogated the γδ T cell receptor (TCR) agonistic activity of self and microbial pAgs (C5 to C15). Only mevalonate-derived geranylgeranyl diphosphate (GGPP, C20) resisted CD39-mediated hydrolysis and acted as a regulator of CD39 expression and activity. GGPP enhanced macrophage differentiation in response to the tissue stress cytokine interleukin-15. In addition, GGPP-imprinted macrophage-like cells displayed increased capacity to produce IL-1β as well as the chemokine CCL2 and preferentially activated CD161-expressing CD4(+) T cells in an innate-like manner. Our studies reveal a previously unrecognized immunoregulatory function of CD39 and highlight a particular role of GGPP among pAgs.
Collapse
Affiliation(s)
- Georg Gruenbacher
- Immunotherapy Unit, Department of Urology, Medical University of Innsbruck and K1 Center Oncotyrol-Center for Personalized Cancer Medicine, 6020 Innsbruck, Austria
| | - Hubert Gander
- Immunotherapy Unit, Department of Urology, Medical University of Innsbruck and K1 Center Oncotyrol-Center for Personalized Cancer Medicine, 6020 Innsbruck, Austria
| | - Andrea Rahm
- Immunotherapy Unit, Department of Urology, Medical University of Innsbruck and K1 Center Oncotyrol-Center for Personalized Cancer Medicine, 6020 Innsbruck, Austria
| | - Marco Idzko
- Department of Pulmonary Medicine, University Medical Center Freiburg, 79106 Freiburg, Germany
| | - Oliver Nussbaumer
- Peter Gorer Department of Immunobiology, King's College London, London SE1 9RT, UK
| | - Martin Thurnher
- Immunotherapy Unit, Department of Urology, Medical University of Innsbruck and K1 Center Oncotyrol-Center for Personalized Cancer Medicine, 6020 Innsbruck, Austria.
| |
Collapse
|
218
|
Sharon C, Baranwal S, Patel NJ, Rodriguez-Agudo D, Pandak WM, Majumdar APN, Krystal G, Patel BB. Inhibition of insulin-like growth factor receptor/AKT/mammalian target of rapamycin axis targets colorectal cancer stem cells by attenuating mevalonate-isoprenoid pathway in vitro and in vivo. Oncotarget 2016; 6:15332-47. [PMID: 25895029 PMCID: PMC4558155 DOI: 10.18632/oncotarget.3684] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Accepted: 03/06/2015] [Indexed: 01/06/2023] Open
Abstract
We observed a co-upregulation of the insulin-like growth factor receptor (IGF-1R)/AKT/mammalian target of rapamycin (mTOR) [InAT] axis and the mevalonate-isoprenoid biosynthesis (MIB) pathways in colorectal cancer stem cells (CSCs) in an unbiased approach. Hence, we hypothesized that the InAT axis might regulate the MIB pathway to govern colorectal CSCs growth. Stimulation (IGF-1) or inhibition (IGF-1R depletion and pharmacological inhibition of IGF-1R/mTOR) of the InAT axis produced induction or attenuation of CSC growth as well as expression of CSC markers and self-renewal factors respectively. Intriguingly, activation of the InAT axis (IGF-1) caused significant upregulation of the MIB pathway genes (both mRNA and protein); while its inhibition produced the opposite effects in colonospheres. More importantly, supplementation with dimethylallyl- and farnesyl-PP, MIB metabolites downstream of isopentenyl-diphosphate delta isomerase (IDI), but not mevalonate and isopentenyl-pp that are upstream of IDI, resulted in a near-complete reversal of the suppressive effect of the InAT axis inhibitors on CSCs growth. The latter findings suggest a specific regulation of the MIB pathway by the InAT axis distal to the target of statins that inhibit 3-hydroxy-3-methyl-glutaryl-CoA reductase (HMGCR). Effects of IGF-1R inhibition on colonic CSCs proliferation and the MIB pathway were confirmed in an ‘in vivo’ HCT-116 xenograft model. These observations establish a novel mechanistic link between the InAT axis that is commonly deregulated in colorectal cancer and the MIB pathway in regulation of colonic CSCs growth. Hence, the InAT-MIB corridor is a novel target for developing paradigm shifting optimum anti-CSCs therapies for colorectal cancer.
Collapse
Affiliation(s)
- Chetna Sharon
- Division of Hematology, Oncology and Palliative Care, Virginia Commonwealth University, Richmond, VA, USA
| | - Somesh Baranwal
- Division of Hematology, Oncology and Palliative Care, Virginia Commonwealth University, Richmond, VA, USA
| | | | - Daniel Rodriguez-Agudo
- Hunter Holmes McGuire VA Medical Center, Richmond, VA, USA.,Department of Medicine, Division of Gastroenterology, Virginia Commonwealth University, Richmond, VA, USA
| | - William M Pandak
- Hunter Holmes McGuire VA Medical Center, Richmond, VA, USA.,Department of Medicine, Division of Gastroenterology, Virginia Commonwealth University, Richmond, VA, USA
| | | | - Geoffrey Krystal
- Hunter Holmes McGuire VA Medical Center, Richmond, VA, USA.,Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, USA.,Division of Hematology, Oncology and Palliative Care, Virginia Commonwealth University, Richmond, VA, USA
| | - Bhaumik B Patel
- Hunter Holmes McGuire VA Medical Center, Richmond, VA, USA.,Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, USA.,Division of Hematology, Oncology and Palliative Care, Virginia Commonwealth University, Richmond, VA, USA
| |
Collapse
|
219
|
MicroRNA: a connecting road between apoptosis and cholesterol metabolism. Tumour Biol 2016; 37:8529-54. [PMID: 27105614 DOI: 10.1007/s13277-016-4988-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Accepted: 02/10/2016] [Indexed: 12/15/2022] Open
Abstract
Resistance to apoptosis leads to tumorigenesis and failure of anti-cancer therapy. Recent studies also highlight abrogated lipid/cholesterol metabolism as one of the root causes of cancer that can lead to metastatic transformations. Cancer cells are dependent on tremendous supply of cellular cholesterol for the formation of new membranes and continuation of cell signaling. Cholesterol homeostasis network tightly regulates this metabolic need of cancer cells on cholesterol and other lipids. Genetic landscape is also shared between apoptosis and cholesterol metabolism. MicroRNAs (miRNAs) are the new fine tuners of signaling pathways and cellular processes and are known for their ability to post-transcriptionally repress gene expression in a targeted manner. This review summarizes the current knowledge about the cross talk between apoptosis and cholesterol metabolism via miRNAs. In addition, we also emphasize herein recent therapeutic modulations of specific miRNAs and their promising potential for the treatment of deadly diseases including cancer and cholesterol related pathologies. Understanding of the impact of miRNA-based regulation of apoptosis and metabolic processes is still at its dawn and needs further research for the development of future miRNA-based therapies. As both these physiological processes affect cellular homeostasis, we believe that this comprehensive summary of miRNAs modulating both apoptosis and cholesterol metabolism will open uncharted territory for scientific exploration and will provide the foundation for discovering novel drug targets for cancer and metabolic diseases.
Collapse
|
220
|
|
221
|
Lee YM, Chang WC, Ma WL. Hypothesis: solid tumours behave as systemic metabolic dictators. J Cell Mol Med 2016; 20:1076-85. [PMID: 26843513 PMCID: PMC4882994 DOI: 10.1111/jcmm.12794] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Accepted: 12/22/2015] [Indexed: 02/06/2023] Open
Abstract
Current knowledge regarding mechanisms of carcinogenesis in human beings centres around the accumulation of genetic instability, amplified cellular signalling, disturbed cellular energy metabolism and microenvironmental regulation governed by complicated cell-cell interactions. In this article, we provide an alternative view of cancer biology. We propose that cancer behaves as a systemic dictator that interacts with tissues throughout the body to control their metabolism and eventually homeostasis. The mechanism of development of this endocrine organ-like tumour (EOLT) tissue might be the driving force for cancer progression. Here, we review the literature that led to the development of this hypothesis. The EOLT phenotype can be defined as a tumour that alters systemic homeostasis. The literature indicates that the EOLT phenotype is present throughout cancer progression. The feedback mechanism that governs the interaction between tumours and various organs is unknown. We believe that investigating the mechanism of EOLT development may advance the current knowledge of regulation within the tumour macroenvironment and consequently lead to new diagnostic methods and therapy.
Collapse
Affiliation(s)
- Yang-Ming Lee
- Sex Hormone Research Center, Graduate Institution of Clinical Medical Science, China Medical University, Taichung, Taiwan.,Department of Endocrinology and Metabolism, Changhua Christian Hospital, Changhua, Taiwan
| | - Wei-Chun Chang
- Sex Hormone Research Center, Department of Gynecology and Obstetric, China Medical University Hospital, Taichung, Taiwan
| | - Wen-Lung Ma
- Sex Hormone Research Center, Graduate Institution of Clinical Medical Science, China Medical University, Taichung, Taiwan.,Sex Hormone Research Center, Department of Gynecology and Obstetric, China Medical University Hospital, Taichung, Taiwan
| |
Collapse
|
222
|
Distinct cholesterogenic and lipidogenic gene expression patterns in ovarian cancer - a new pool of biomarkers. Genes Cancer 2016; 6:472-9. [PMID: 26807200 PMCID: PMC4701226 DOI: 10.18632/genesandcancer.87] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Cancer cells display different metabolic requirements compared to nonmalignant cells imposed by their need for rapid proliferation. Alterations in cellular metabolic pathways of lipid and cholesterol synthesis have been linked to tumorigenesis and cancer progression but have not been exploited in clinical diagnosis. Here, the expression of genes related to cholesterol/lipid metabolism was measured with semiquantitative and real-time RT-PCR in RNA isolated from normal, benign and cancer ovarian tissues. We found that both SREBF2 and its target gene DHCR7 are downregulated in ovarian cancer tissues. On the contrary, SREBF1c and its target SCD1 were upregulated. The steroidogenesis regulator PDE8B was found downregulated. Oncomine analysis supported these findings, and further revealed that in ovarian cancers, the SREBF1-regulated lipidogenic pathway is activated while the SREBF2-regulated cholesterogenic pathway is repressed based on expression profiles of HMGCR and DHCR7. In conclusion, we show that ovarian cancer cells display distinct lipidogenic and cholesterogenic gene expression profiles with potential applications in the development of new biomarkers and/or treatment of ovarian cancer. Reduced cholesterol and enhanced lipid synthesis and SCD1 expression may provide an explanation for the previously reported increased membrane fluidity of ovarian cancer cells, a finding that merits further investigation.
Collapse
|
223
|
Trojan PJJ, Bohatch-Junior MS, Otuki MF, Souza-Fonseca-Guimarães F, Svidnicki PV, Nogaroto V, Fernandes D, Krum EA, Favero GM. Pravastatin induces cell cycle arrest and decreased production of VEGF and bFGF in multiple myeloma cell line. BRAZ J BIOL 2016; 76:59-65. [PMID: 26909624 DOI: 10.1590/1519-6984.11914] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2014] [Accepted: 10/17/2014] [Indexed: 01/19/2023] Open
Abstract
Multiple myeloma (MM) is a B cell bone marrow neoplasia characterized by inflammation with an intense secretion of growth factors that promote tumor growth, cell survival, migration and invasion. The aim of this study was to evaluate the effects of pravastatin, a drug used to reduce cholesterol, in a MM cell line.Cell cycle and viability were determinate by Trypan Blue and Propidium Iodide. IL6, VEGF, bFGF and TGFβ were quantified by ELISA and qRT-PCR including here de HMG CoA reductase. It was observed reduction of cell viability, increase of cells in G0/G1 phase of the cell cycle and reducing the factors VEGF and bFGF without influence on 3-Methyl-Glutaryl Coenzyme A reductase expression.The results demonstrated that pravastatin induces cell cycle arrest in G0/G1 and decreased production of growth factors in Multiple Myeloma cell line.
Collapse
Affiliation(s)
- P J J Trojan
- Laboratório Multidisciplinar de Ciências Biológicas e da Saúde, Universidade Estadual de Ponta Grossa, Ponta Grossa, PR, Brazil
| | - M S Bohatch-Junior
- Laboratório Multidisciplinar de Ciências Biológicas e da Saúde, Universidade Estadual de Ponta Grossa, Ponta Grossa, PR, Brazil
| | - M F Otuki
- Departamento de Ciências Farmacêuticas, Universidade Estadual de Ponta Grossa, Ponta Grossa, PR, Brazil
| | - F Souza-Fonseca-Guimarães
- Unit Cytokines and Inflammation, Department Infection and Epidemiology Institut Pasteur, Paris, France
| | - P V Svidnicki
- Departamento de Biologia Molecular, Estrutural e Genética, Universidade Estadual de Ponta Grossa, Ponta Grossa, PR, Brazil
| | - V Nogaroto
- Departamento de Biologia Molecular, Estrutural e Genética, Universidade Estadual de Ponta Grossa, Ponta Grossa, PR, Brazil
| | - D Fernandes
- Departamento de Ciências Farmacêuticas, Universidade Estadual de Ponta Grossa, Ponta Grossa, PR, Brazil
| | - E A Krum
- Departamento de Ciências Farmacêuticas, Universidade Estadual de Ponta Grossa, Ponta Grossa, PR, Brazil
| | - G M Favero
- Laboratório Multidisciplinar de Ciências Biológicas e da Saúde, Universidade Estadual de Ponta Grossa, Ponta Grossa, PR, Brazil
| |
Collapse
|
224
|
Benatti P, Chiaramonte ML, Lorenzo M, Hartley JA, Hochhauser D, Gnesutta N, Mantovani R, Imbriano C, Dolfini D. NF-Y activates genes of metabolic pathways altered in cancer cells. Oncotarget 2016; 7:1633-50. [PMID: 26646448 PMCID: PMC4811486 DOI: 10.18632/oncotarget.6453] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Accepted: 11/15/2015] [Indexed: 12/21/2022] Open
Abstract
The trimeric transcription factor NF-Y binds to the CCAAT box, an element enriched in promoters of genes overexpressed in tumors. Previous studies on the NF-Y regulome identified the general term metabolism as significantly enriched. We dissect here in detail the targeting of metabolic genes by integrating analysis of NF-Y genomic binding and profilings after inactivation of NF-Y subunits in different cell types. NF-Y controls de novo biosynthetic pathways of lipids, teaming up with the master SREBPs regulators. It activates glycolytic genes, but, surprisingly, is neutral or represses mitochondrial respiratory genes. NF-Y targets the SOCG (Serine, One Carbon, Glycine) and Glutamine pathways, as well as genes involved in the biosynthesis of polyamines and purines. Specific cancer-driving nodes are generally under NF-Y control. Altogether, these data delineate a coherent strategy to promote expression of metabolic genes fuelling anaerobic energy production and other anabolic pathways commonly altered in cancer cells.
Collapse
Affiliation(s)
- Paolo Benatti
- Dipartimento di Scienze della Vita, Università di Modena e Reggio Emilia, Modena, Italy
| | | | - Mariangela Lorenzo
- Dipartimento di Bioscienze, Università degli Studi di Milano, Milano, Italy
| | - John A. Hartley
- Cancer Research UK Drug-DNA Interactions Research Group, UCL Cancer Institute, Paul O'Gorman Building, University College London, London, UK
| | - Daniel Hochhauser
- Cancer Research UK Drug-DNA Interactions Research Group, UCL Cancer Institute, Paul O'Gorman Building, University College London, London, UK
| | - Nerina Gnesutta
- Dipartimento di Bioscienze, Università degli Studi di Milano, Milano, Italy
| | - Roberto Mantovani
- Dipartimento di Bioscienze, Università degli Studi di Milano, Milano, Italy
| | - Carol Imbriano
- Dipartimento di Scienze della Vita, Università di Modena e Reggio Emilia, Modena, Italy
| | - Diletta Dolfini
- Dipartimento di Bioscienze, Università degli Studi di Milano, Milano, Italy
| |
Collapse
|
225
|
Marcuzzi A, Piscianz E, Loganes C, Vecchi Brumatti L, Knowles A, Bilel S, Tommasini A, Bortul R, Zweyer M. Innovative Target Therapies Are Able to Block the Inflammation Associated with Dysfunction of the Cholesterol Biosynthesis Pathway. Int J Mol Sci 2015; 17:47. [PMID: 26729102 PMCID: PMC4730292 DOI: 10.3390/ijms17010047] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Revised: 12/23/2015] [Accepted: 12/24/2015] [Indexed: 01/24/2023] Open
Abstract
The cholesterol pathway is an essential biochemical process aimed at the synthesis of bioactive molecules involved in multiple crucial cellular functions. The end products of this pathway are sterols, such as cholesterol, which are essential components of cell membranes, precursors of steroid hormones, bile acids and other molecules such as ubiquinone. Several diseases are caused by defects in this metabolic pathway: the most severe forms of which cause neurological involvement (psychomotor retardation and cerebellar ataxia) as a result of a variety of cellular impairments, including mitochondrial dysfunction. These pathologies are induced by convergent mechanisms in which the mitochondrial unit plays a pivotal role contributing to defective apoptosis, autophagy and mitophagy processes. Unraveling these mechanisms would contribute to the development of effective drug treatments for these disorders. In addition, the development of biochemical models could have a substantial impact on the understanding of the mechanism of action of drugs that act on this pathway in multifactor disorders. In this review we will focus in particular on inhibitors of cholesterol synthesis, mitochondria-targeted drugs and inhibitors of the inflammasome.
Collapse
Affiliation(s)
- Annalisa Marcuzzi
- Department of Medicine, Surgery and Health Sciences, University of Trieste, Piazzale Europa 1, Trieste 34128, Italy.
| | - Elisa Piscianz
- Institute for Maternal and Child Health-IRCCS "Burlo Garofolo", via dell'Istria, 65/1, Trieste 34137, Italy.
| | - Claudia Loganes
- Department of Medicine, Surgery and Health Sciences, University of Trieste, Piazzale Europa 1, Trieste 34128, Italy.
| | - Liza Vecchi Brumatti
- Institute for Maternal and Child Health-IRCCS "Burlo Garofolo", via dell'Istria, 65/1, Trieste 34137, Italy.
| | - Alessandra Knowles
- Institute for Maternal and Child Health-IRCCS "Burlo Garofolo", via dell'Istria, 65/1, Trieste 34137, Italy.
| | - Sabrine Bilel
- Cluster in Biomedicine (CBM scrl), Trieste 34128, Italy.
| | - Alberto Tommasini
- Institute for Maternal and Child Health-IRCCS "Burlo Garofolo", via dell'Istria, 65/1, Trieste 34137, Italy.
| | - Roberta Bortul
- Department of Medicine, Surgery and Health Sciences, University of Trieste, Piazzale Europa 1, Trieste 34128, Italy.
| | - Marina Zweyer
- Department of Medicine, Surgery and Health Sciences, University of Trieste, Piazzale Europa 1, Trieste 34128, Italy.
| |
Collapse
|
226
|
Gómez de Cedrón M, Ramírez de Molina A. Microtargeting cancer metabolism: opening new therapeutic windows based on lipid metabolism. J Lipid Res 2015; 57:193-206. [PMID: 26630911 DOI: 10.1194/jlr.r061812] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Indexed: 01/04/2023] Open
Abstract
Metabolic reprogramming has emerged as a hallmark of cancer. MicroRNAs are noncoding RNAs that posttranscriptionally repress the expression of target mRNAs implicated in multiple physiological processes, including apoptosis, differentiation, and cancer. MicroRNAs can affect entire biological pathways, making them good candidates for therapeutic intervention compared with classical single target approaches. Moreover, microRNAs may become more relevant in the fine-tuning adaptation to stress situations, such as oncogenic events, hypoxia, nutrient deprivation, and oxidative stress. Furthermore, artificial microRNAs can be designed to modulate the expression of multiple targets of a specific pathway. In this review, we describe the metabolic reprogramming associated to cancer, with a special interest in the altered lipid metabolism. Next, we describe specific features of microRNAs that make them relevant to target cancer cell metabolism. Finally, in an attempt to open new therapeutic windows, we emphasize two exciting scenarios for microRNA-mediated intervention that need to be further explored: 1) the cooperation between FA biosynthesis (lipogenesis) and FA oxidation as complementary partners for the survival of cancer cells; and 2) the regulation of the intracellular lipid content modulating both lipid storage into lipid droplets, and lipid mobilization through lipolysis and/or lipophagy.
Collapse
Affiliation(s)
- Marta Gómez de Cedrón
- Molecular Oncology and Nutritional Genomics of Cancer Group, IMDEA (Madrid Institute of Advanced Studies)-Food, CEI UAM + CSIC, Madrid, Spain
| | - Ana Ramírez de Molina
- Molecular Oncology and Nutritional Genomics of Cancer Group, IMDEA (Madrid Institute of Advanced Studies)-Food, CEI UAM + CSIC, Madrid, Spain
| |
Collapse
|
227
|
Influence of Serum and Hypoxia on Incorporation of [14C]-d-Glucose or [14C]-l-Glutamine into Lipids and Lactate in Murine Glioblastoma Cells. Lipids 2015; 50:1167-84. [DOI: 10.1007/s11745-015-4075-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2015] [Accepted: 08/28/2015] [Indexed: 12/29/2022]
|
228
|
Haskins JW, Zhang S, Means RE, Kelleher JK, Cline GW, Canfrán-Duque A, Suárez Y, Stern DF. Neuregulin-activated ERBB4 induces the SREBP-2 cholesterol biosynthetic pathway and increases low-density lipoprotein uptake. Sci Signal 2015; 8:ra111. [PMID: 26535009 DOI: 10.1126/scisignal.aac5124] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Cholesterol is a lipid that is critical for steroid hormone production and the integrity of cellular membranes, and, as such, it is essential for cell growth. The epidermal growth factor receptor (EGFR) family member ERBB4, which forms signaling complexes with other EGFR family members, can undergo ligand-induced proteolytic cleavage to release a soluble intracellular domain (ICD) that enters the nucleus to modify transcription. We found that ERBB4 activates sterol regulatory element binding protein-2 (SREBP-2) to enhance low-density lipoprotein (LDL) uptake and cholesterol biosynthesis. Expression of the ERBB4 ICD in mammary epithelial cells or activation of ERBB4 with the ligand neuregulin 1 (NRG1) induced the expression of SREBP target genes involved in cholesterol biosynthesis, including HMGCR and HMGCS1, and lipid uptake, LDLR, which encodes the LDL receptor. Addition of NRG1 increased the abundance of the cleaved, mature form of SREBP-2 through a pathway that was blocked by addition of inhibitors of PI3K (phosphatidylinositol 3-kinase) or dual inhibition of mammalian target of rapamycin complex 1 (mTORC1) and mTORC2, but not by inhibition of AKT or mTORC1. Pharmacological inhibition of the activity of SREBP site 1 protease or of all EGFR family members (with lapatinib), but not EGFR alone (with erlotinib), impaired NRG1-induced expression of cholesterol biosynthesis genes. Collectively, our findings indicated that activation of ERBB4 promotes SREBP-2-regulated cholesterol metabolism. The connections of EGFR and ERBB4 signaling with SREBP-2-regulated cholesterol metabolism are likely to be important in ERBB-regulated developmental processes and may contribute to metabolic remodeling in ERBB-driven cancers.
Collapse
Affiliation(s)
- Jonathan W Haskins
- Department of Pathology and Yale Cancer Center, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Shannon Zhang
- Department of Pathology and Yale Cancer Center, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Robert E Means
- Department of Pathology and Yale Cancer Center, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Joanne K Kelleher
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Gary W Cline
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Alberto Canfrán-Duque
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Yajaira Suárez
- Department of Pathology and Yale Cancer Center, Yale University School of Medicine, New Haven, CT 06510, USA. Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT 06510, USA
| | - David F Stern
- Department of Pathology and Yale Cancer Center, Yale University School of Medicine, New Haven, CT 06510, USA.
| |
Collapse
|
229
|
Predictive value of blood lipid association with response to neoadjuvant chemoradiotherapy in colorectal cancer. Tumour Biol 2015; 37:4955-61. [PMID: 26531721 DOI: 10.1007/s13277-015-4320-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Accepted: 10/26/2015] [Indexed: 12/11/2022] Open
Abstract
The aim of this research was to explore whether blood lipid parameters could predict tumor regression grading (TRG) and compare with the predictive value of carcinoembryonic antigen (CEA) in patients with locally advanced colorectal cancer (LARC) treated with neoadjuvant chemoradiotherapy (nCRT). Between June 2011 and January 2015, the records of 176 patients with primary colorectal adenocarcinoma treated with nCRT followed by radical surgery were reviewed retrospectively. Total cholesterol (TC), triglyceride (TG), low-density lipoprotein (LDL), high-density lipoprotein (HDL), and pre-CEA were measured before nCRT, and post-CEA was measured before surgery. A total of 129 (73.3 %) good responders (TRG 3-4) and 47 (26.7 %) poor responders (TRG 0-2) were assessed after the nCRT. TC, LDL, HDL, and ΔCEA were 6.56 ± 0.95, 3.08 ± 0.72, and 1.43 ± 0.25 mmol/L and -0.69 ± 8.33 μg/mL in poor responders compared with 5.15 ± 1.29, 2.39 ± 0.5, and 1.37 ± 0.32 mmol/L and 16.67 ± 30.18 μg/mL in good responders, respectively (p < 0.05). TG, pre-CEA, and post-CEA were not significantly different. Multivariate logistic regression analysis revealed TC and ΔCEA as independent factors in predicting TRG; TC showed a sensitivity of 62.79 %, a specificity of 91.49 %, a Youden index of 0.543, a cutoff value of 5.52, and an AUC of 0.800 compared with ΔCEA (sensitivity 76.74 %, specificity 65.96 %, Youden index 0.427, and AUC 0.761). TC has a better predictive value than ΔCEA and hence might serve as a predictor of TRG in LARC patients.
Collapse
|
230
|
Legut M, Cole DK, Sewell AK. The promise of γδ T cells and the γδ T cell receptor for cancer immunotherapy. Cell Mol Immunol 2015; 12:656-68. [PMID: 25864915 PMCID: PMC4716630 DOI: 10.1038/cmi.2015.28] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2015] [Accepted: 03/01/2015] [Indexed: 12/13/2022] Open
Abstract
γδ T cells form an important part of adaptive immune responses against infections and malignant transformation. The molecular targets of human γδ T cell receptors (TCRs) remain largely unknown, but recent studies have confirmed the recognition of phosphorylated prenyl metabolites, lipids in complex with CD1 molecules and markers of cellular stress. All of these molecules are upregulated on various cancer types, highlighting the potential importance of the γδ T cell compartment in cancer immunosurveillance and paving the way for the use of γδ TCRs in cancer therapy. Ligand recognition by the γδ TCR often requires accessory/co-stimulatory stress molecules on both T cells and target cells; this cellular stress context therefore provides a failsafe against harmful self-reactivity. Unlike αβ T cells, γδ T cells recognise their targets irrespective of HLA haplotype and therefore offer exciting possibilities for off-the-shelf, pan-population cancer immunotherapies. Here, we present a review of known ligands of human γδ T cells and discuss the promise of harnessing these cells for cancer treatment.
Collapse
MESH Headings
- Antigen Presentation
- Antigens, CD1/genetics
- Antigens, CD1/immunology
- Antigens, Neoplasm/genetics
- Antigens, Neoplasm/immunology
- Clinical Trials as Topic
- Gene Expression Regulation, Neoplastic/immunology
- Hemiterpenes/immunology
- Humans
- Immunotherapy/methods
- Ligands
- Models, Molecular
- Monitoring, Immunologic
- Neoplasms/genetics
- Neoplasms/immunology
- Neoplasms/pathology
- Neoplasms/therapy
- Organophosphorus Compounds/immunology
- Phosphorylation
- Protein Structure, Tertiary
- Receptors, Antigen, T-Cell, gamma-delta/genetics
- Receptors, Antigen, T-Cell, gamma-delta/immunology
- Signal Transduction
- T-Lymphocytes/immunology
- T-Lymphocytes/pathology
- T-Lymphocytes/transplantation
Collapse
Affiliation(s)
- Mateusz Legut
- Division of Infection and Immunity and Systems Immunity University Research Institute, Cardiff University School of Medicine, Cardiff, UK
| | - David K Cole
- Division of Infection and Immunity and Systems Immunity University Research Institute, Cardiff University School of Medicine, Cardiff, UK
| | - Andrew K Sewell
- Division of Infection and Immunity and Systems Immunity University Research Institute, Cardiff University School of Medicine, Cardiff, UK
| |
Collapse
|
231
|
p53 regulates the mevalonate pathway in human glioblastoma multiforme. Cell Death Dis 2015; 6:e1909. [PMID: 26469958 PMCID: PMC4632304 DOI: 10.1038/cddis.2015.279] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2015] [Revised: 08/04/2015] [Accepted: 08/09/2015] [Indexed: 11/30/2022]
Abstract
The mevalonate (MVA) pathway is an important metabolic pathway implicated in multiple aspects of tumorigenesis. In this study, we provided evidence that p53 induces the expression of a group of enzymes of the MVA pathway including 3′-hydroxy-3′-methylglutaryl-coenzyme A reductase, MVA kinase, farnesyl diphosphate synthase and farnesyl diphosphate farnesyl transferase 1, in the human glioblastoma multiforme cell line, U343 cells, and in normal human astrocytes, NHAs. Genetic and pharmacologic perturbation of p53 directly influences the expression of these genes. Furthermore, p53 is recruited to the gene promoters in designated p53-responsive elements, thereby increasing their transcription. Such effect was abolished by site-directed mutagenesis in the p53-responsive element of promoter of the genes. These findings highlight another aspect of p53 functions unrelated to tumor suppression and suggest p53 as a novel regulator of the MVA pathway providing insight into the role of this pathway in cancer progression.
Collapse
|
232
|
Lewis CA, Brault C, Peck B, Bensaad K, Griffiths B, Mitter R, Chakravarty P, East P, Dankworth B, Alibhai D, Harris AL, Schulze A. SREBP maintains lipid biosynthesis and viability of cancer cells under lipid- and oxygen-deprived conditions and defines a gene signature associated with poor survival in glioblastoma multiforme. Oncogene 2015; 34:5128-40. [PMID: 25619842 DOI: 10.1038/onc.2014.439] [Citation(s) in RCA: 183] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2014] [Revised: 11/04/2014] [Accepted: 12/05/2014] [Indexed: 12/16/2022]
Abstract
Oxygen and nutrient limitation are common features of the tumor microenvironment and are associated with cancer progression and induction of metastasis. The inefficient vascularization of tumor tissue also limits the penetration of other serum-derived factors, such as lipids and lipoproteins, which can be rate limiting for cell proliferation and survival. Here we have investigated the effect of hypoxia and serum deprivation on sterol regulatory element-binding protein (SREBP) activity and the expression of lipid metabolism genes in human glioblastoma multiforme (GBM) cancer cells. We found that SREBP transcriptional activity was induced by serum depletion both in normoxic and hypoxic cells and that activation of SREBP was required to maintain the expression of fatty acid and cholesterol metabolism genes under hypoxic conditions. Moreover, expression of stearoyl-CoA desaturase, the enzyme required for the generation of mono-unsaturated fatty acids, and fatty acid-binding protein 7, a regulator of glioma stem cell function, was strongly dependent on SREBP function. Inhibition of SREBP function blocked lipid biosynthesis in hypoxic cancer cells and impaired cell survival under hypoxia and in a three-dimensional spheroid model. Finally, gene expression analysis revealed that SREBP defines a gene signature that is associated with poor survival in glioblastoma.
Collapse
Affiliation(s)
- C A Lewis
- Gene Expression Analysis Laboratory, Cancer Research UK London Research Institute, London, UK
| | - C Brault
- Theodor-Boveri-Institute, Biocenter, Am Hubland, Würzburg, Germany
| | - B Peck
- Gene Expression Analysis Laboratory, Cancer Research UK London Research Institute, London, UK
| | - K Bensaad
- CRUK Growth Factor Group, The Weatherall Institute of Molecular Medicine, University of Oxford, Headington, Oxford, UK
| | - B Griffiths
- Gene Expression Analysis Laboratory, Cancer Research UK London Research Institute, London, UK
| | - R Mitter
- Bioinformatics and Biostatistics Service, Cancer Research UK London Research Institute, London, UK
| | - P Chakravarty
- Bioinformatics and Biostatistics Service, Cancer Research UK London Research Institute, London, UK
| | - P East
- Bioinformatics and Biostatistics Service, Cancer Research UK London Research Institute, London, UK
| | - B Dankworth
- Theodor-Boveri-Institute, Biocenter, Am Hubland, Würzburg, Germany
| | - D Alibhai
- Light Microscopy Core, Cancer Research UK London Research Institute, London, UK
| | - A L Harris
- CRUK Growth Factor Group, The Weatherall Institute of Molecular Medicine, University of Oxford, Headington, Oxford, UK
| | - A Schulze
- Gene Expression Analysis Laboratory, Cancer Research UK London Research Institute, London, UK
- Theodor-Boveri-Institute, Biocenter, Am Hubland, Würzburg, Germany
- Comprehensive Cancer Center Mainfranken Würzburg, Germany
| |
Collapse
|
233
|
Qiu Z, Yuan W, Chen T, Zhou C, Liu C, Huang Y, Han D, Huang Q. HMGCR positively regulated the growth and migration of glioblastoma cells. Gene 2015; 576:22-7. [PMID: 26432005 DOI: 10.1016/j.gene.2015.09.067] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2015] [Revised: 08/12/2015] [Accepted: 09/23/2015] [Indexed: 12/14/2022]
Abstract
The metabolic program of cancer cells is significant different from the normal cells, which makes it possible to develop novel strategies targeting cancer cells. Mevalonate pathway and its rate-limiting enzyme HMG-CoA reductase (HMGCR) have shown important roles in the progression of several cancer types. However, their roles in glioblastoma cells remain unknown. In this study, up-regulation of HMGCR in the clinical glioblastoma samples was observed. Forced expression of HMGCR promoted the growth and migration of U251 and U373 cells, while knocking down the expression of HMGCR inhibited the growth, migration and metastasis of glioblastoma cells. Molecular mechanism studies revealed that HMGCR positively regulated the expression of TAZ, an important mediator of Hippo pathway, and the downstream target gene connective tissue growth factor (CTGF), suggesting HMGCR might activate Hippo pathway in glioblastoma cells. Taken together, our study demonstrated the oncogenic roles of HMGCR in glioblastoma cells and HMGCR might be a promising therapeutic target.
Collapse
Affiliation(s)
- Zhihua Qiu
- Department of Neurosurgery, Central Hospital of Zhuzhou, Zhuzhou, Hunan Province 412000, PR China
| | - Wen Yuan
- Department of Neurosurgery, Central Hospital of Zhuzhou, Zhuzhou, Hunan Province 412000, PR China
| | - Tao Chen
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong Province 510120, PR China
| | - Chenzhi Zhou
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong Province 510120, PR China
| | - Chao Liu
- Department of Neurosurgery, Central Hospital of Zhuzhou, Zhuzhou, Hunan Province 412000, PR China
| | - Yongkai Huang
- Department of Neurosurgery, Central Hospital of Zhuzhou, Zhuzhou, Hunan Province 412000, PR China
| | - Deqing Han
- Department of Neurosurgery, Central Hospital of Zhuzhou, Zhuzhou, Hunan Province 412000, PR China.
| | - Qinghui Huang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong Province 510120, PR China; Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong Province 510120, PR China.
| |
Collapse
|
234
|
Aberrant de novo cholesterogenesis: Clinical significance and implications. Clin Chim Acta 2015; 450:356-61. [PMID: 26386164 DOI: 10.1016/j.cca.2015.09.014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2015] [Revised: 09/12/2015] [Accepted: 09/15/2015] [Indexed: 01/23/2023]
Abstract
Human cells can acquire cholesterol from the circulation but also have the ability to synthesize it via de novo cholesterogenesis (DC). Cholesterol absorption and de novo cholesterogenesis are the key processes that modulate cholesterol homeostasis in the human body. The endogenous biosynthesis of cholesterol substantially contributes to the whole-body cholesterol pool. Additionally, dysregulation of this pathway is associated with diverse medical conditions. The present review focuses on our current understanding of the cholesterogenic pathway and the various different factors regulating this pathway. It also highlights dysregulation of this pathway in various physiological and pathological conditions including cardiovascular diseases, type II diabetes, obesity and viral infections.
Collapse
|
235
|
Gabitova L, Restifo D, Gorin A, Manocha K, Handorf E, Yang DH, Cai KQ, Klein-Szanto AJ, Cunningham D, Kratz LE, Herman GE, Golemis EA, Astsaturov I. Endogenous Sterol Metabolites Regulate Growth of EGFR/KRAS-Dependent Tumors via LXR. Cell Rep 2015; 12:1927-38. [PMID: 26344763 DOI: 10.1016/j.celrep.2015.08.023] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Revised: 07/10/2015] [Accepted: 08/07/2015] [Indexed: 02/04/2023] Open
Abstract
Meiosis-activating sterols (MAS) are substrates of SC4MOL and NSDHL in the cholesterol pathway and are important for normal organismal development. Oncogenic transformation by epidermal growth factor receptor (EGFR) or RAS increases the demand for cholesterol, suggesting a possibility for metabolic interference. To test this idea in vivo, we ablated Nsdhl in adult keratinocytes expressing KRAS(G12D). Strikingly, Nsdhl inactivation antagonized the growth of skin tumors while having little effect on normal skin. Loss of Nsdhl induced the expression of ATP-binding cassette (ABC) transporters ABCA1 and ABCG1, reduced the expression of low-density lipoprotein receptor (LDLR), decreased intracellular cholesterol, and was dependent on the liver X receptor (LXR) α. Importantly, EGFR signaling opposed LXRα effects on cholesterol homeostasis, whereas an EGFR inhibitor synergized with LXRα agonists in killing cancer cells. Inhibition of SC4MOL or NSDHL, or activation of LXRα by sterol metabolites, can be an effective strategy against carcinomas with activated EGFR-KRAS signaling.
Collapse
Affiliation(s)
- Linara Gabitova
- Molecular Therapeutics Program, Fox Chase Cancer Center, Philadelphia, PA 19111, USA; Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Tatarstan 420000, Russia
| | - Diana Restifo
- Molecular Therapeutics Program, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | - Andrey Gorin
- Molecular Therapeutics Program, Fox Chase Cancer Center, Philadelphia, PA 19111, USA; Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Tatarstan 420000, Russia
| | - Kunal Manocha
- Molecular Therapeutics Program, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | - Elizabeth Handorf
- Biostatistics and Bioinformatics Facility, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | - Dong-Hua Yang
- Molecular Therapeutics Program, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | - Kathy Q Cai
- Histopathology Facility, Fox Chase Cancer Center, Philadelphia, PA 19111, USA; Cancer Biology Program, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | - Andres J Klein-Szanto
- Histopathology Facility, Fox Chase Cancer Center, Philadelphia, PA 19111, USA; Cancer Biology Program, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | - David Cunningham
- The Research Institute at Nationwide Children's Hospital and Department of Pediatrics, The Ohio State University, Columbus, OH 43205, USA
| | - Lisa E Kratz
- Kennedy Krieger Institute, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Gail E Herman
- The Research Institute at Nationwide Children's Hospital and Department of Pediatrics, The Ohio State University, Columbus, OH 43205, USA
| | - Erica A Golemis
- Molecular Therapeutics Program, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | - Igor Astsaturov
- Molecular Therapeutics Program, Fox Chase Cancer Center, Philadelphia, PA 19111, USA; Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Tatarstan 420000, Russia.
| |
Collapse
|
236
|
Hsieh AL, Walton ZE, Altman BJ, Stine ZE, Dang CV. MYC and metabolism on the path to cancer. Semin Cell Dev Biol 2015; 43:11-21. [PMID: 26277543 DOI: 10.1016/j.semcdb.2015.08.003] [Citation(s) in RCA: 261] [Impact Index Per Article: 26.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2015] [Revised: 07/17/2015] [Accepted: 08/09/2015] [Indexed: 12/13/2022]
Abstract
The MYC proto-oncogene is frequently deregulated in human cancers, activating genetic programs that orchestrate biological processes to promote growth and proliferation. Altered metabolism characterized by heightened nutrients uptake, enhanced glycolysis and glutaminolysis and elevated fatty acid and nucleotide synthesis is the hallmark of MYC-driven cancer. Recent evidence strongly suggests that Myc-dependent metabolic reprogramming is critical for tumorigenesis, which could be attenuated by targeting specific metabolic pathways using small drug-like molecules. Understanding the complexity of MYC-mediated metabolic re-wiring in cancers as well as how MYC cooperates with other metabolic drivers such as mammalian target of rapamycin (mTOR) will provide translational opportunities for cancer therapy.
Collapse
Affiliation(s)
- Annie L Hsieh
- Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Zandra E Walton
- Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Brian J Altman
- Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Zachary E Stine
- Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Chi V Dang
- Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Division of Hematology-Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
237
|
Na YR, Hong JH, Lee MY, Jung JH, Jung D, Kim YW, Son D, Choi M, Kim KP, Seok SH. Proteomic Analysis Reveals Distinct Metabolic Differences Between Granulocyte-Macrophage Colony Stimulating Factor (GM-CSF) and Macrophage Colony Stimulating Factor (M-CSF) Grown Macrophages Derived from Murine Bone Marrow Cells. Mol Cell Proteomics 2015; 14:2722-32. [PMID: 26229149 DOI: 10.1074/mcp.m115.048744] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2015] [Indexed: 12/27/2022] Open
Abstract
Macrophages are crucial in controlling infectious agents and tissue homeostasis. Macrophages require a wide range of functional capabilities in order to fulfill distinct roles in our body, one being rapid and robust immune responses. To gain insight into macrophage plasticity and the key regulatory protein networks governing their specific functions, we performed quantitative analyses of the proteome and phosphoproteome of murine primary GM-CSF and M-CSF grown bone marrow derived macrophages (GM-BMMs and M-BMMs, respectively) using the latest isobaric tag based tandem mass tag (TMT) labeling and liquid chromatography-tandem mass spectrometry (LC-MS/MS). Strikingly, metabolic processes emerged as a major difference between these macrophages. Specifically, GM-BMMs show significant enrichment of proteins involving glycolysis, the mevalonate pathway, and nitrogen compound biosynthesis. This evidence of enhanced glycolytic capability in GM-BMMs is particularly significant regarding their pro-inflammatory responses, because increased production of cytokines upon LPS stimulation in GM-BMMs depends on their acute glycolytic capacity. In contrast, M-BMMs up-regulate proteins involved in endocytosis, which correlates with a tendency toward homeostatic functions such as scavenging cellular debris. Together, our data describes a proteomic network that underlies the pro-inflammatory actions of GM-BMMs as well as the homeostatic functions of M-BMMs.
Collapse
Affiliation(s)
- Yi Rang Na
- From the ‡Department of Microbiology and Immunology, and Institute of Endemic Disease, Seoul National University College of Medicine, 103 Daehak-ro, Chongno-gu, Seoul 110-799, South Korea
| | - Ji Hye Hong
- §Department of Applied Chemistry, Kyung Hee University, 1732 Deogyeong-daero, Giheung-gu, Yongin-si, Gyeonggi-do 446-701, South Korea
| | - Min Yong Lee
- §Department of Applied Chemistry, Kyung Hee University, 1732 Deogyeong-daero, Giheung-gu, Yongin-si, Gyeonggi-do 446-701, South Korea
| | - Jae Hun Jung
- §Department of Applied Chemistry, Kyung Hee University, 1732 Deogyeong-daero, Giheung-gu, Yongin-si, Gyeonggi-do 446-701, South Korea
| | - Daun Jung
- From the ‡Department of Microbiology and Immunology, and Institute of Endemic Disease, Seoul National University College of Medicine, 103 Daehak-ro, Chongno-gu, Seoul 110-799, South Korea
| | - Young Won Kim
- From the ‡Department of Microbiology and Immunology, and Institute of Endemic Disease, Seoul National University College of Medicine, 103 Daehak-ro, Chongno-gu, Seoul 110-799, South Korea
| | - Dain Son
- From the ‡Department of Microbiology and Immunology, and Institute of Endemic Disease, Seoul National University College of Medicine, 103 Daehak-ro, Chongno-gu, Seoul 110-799, South Korea
| | - Murim Choi
- ¶Department of Biomedical Science, Seoul National University College of Medicine, 103 Daehak-ro, Chongno-gu, Seoul 110-799, South Korea
| | - Kwang Pyo Kim
- §Department of Applied Chemistry, Kyung Hee University, 1732 Deogyeong-daero, Giheung-gu, Yongin-si, Gyeonggi-do 446-701, South Korea;
| | - Seung Hyeok Seok
- From the ‡Department of Microbiology and Immunology, and Institute of Endemic Disease, Seoul National University College of Medicine, 103 Daehak-ro, Chongno-gu, Seoul 110-799, South Korea
| |
Collapse
|
238
|
Kobayashi Y, Kashima H, Wu RC, Jung JG, Kuan JC, Gu J, Xuan J, Sokoll L, Visvanathan K, Shih IM, Wang TL. Mevalonate Pathway Antagonist Suppresses Formation of Serous Tubal Intraepithelial Carcinoma and Ovarian Carcinoma in Mouse Models. Clin Cancer Res 2015; 21:4652-62. [PMID: 26109099 DOI: 10.1158/1078-0432.ccr-14-3368] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2014] [Accepted: 06/08/2015] [Indexed: 12/13/2022]
Abstract
PURPOSE Statins are among the most frequently prescribed drugs because of their efficacy and low toxicity in treating hypercholesterolemia. Recently, statins have been reported to inhibit the proliferative activity of cancer cells, especially those with TP53 mutations. Because TP53 mutations occur in almost all ovarian high-grade serous carcinoma (HGSC), we determined whether statins suppressed tumor growth in animal models of ovarian cancer. EXPERIMENTAL DESIGN Two ovarian cancer mouse models were used. The first one was a genetically engineered model, mogp-TAg, in which the promoter of oviduct glycoprotein-1 was used to drive the expression of SV40 T-antigen in gynecologic tissues. These mice spontaneously developed serous tubal intraepithelial carcinomas (STICs), which are known as ovarian cancer precursor lesions. The second model was a xenograft tumor model in which human ovarian cancer cells were inoculated into immunocompromised mice. Mice in both models were treated with lovastatin, and effects on tumor growth were monitored. The molecular mechanisms underlying the antitumor effects of lovastatin were also investigated. RESULTS Lovastatin significantly reduced the development of STICs in mogp-TAg mice and inhibited ovarian tumor growth in the mouse xenograft model. Knockdown of prenylation enzymes in the mevalonate pathway recapitulated the lovastatin-induced antiproliferative phenotype. Transcriptome analysis indicated that lovastatin affected the expression of genes associated with DNA replication, Rho/PLC signaling, glycolysis, and cholesterol biosynthesis pathways, suggesting that statins have pleiotropic effects on tumor cells. CONCLUSIONS The above results suggest that repurposing statin drugs for ovarian cancer may provide a promising strategy to prevent and manage this devastating disease.
Collapse
Affiliation(s)
- Yusuke Kobayashi
- Department of Pathology, School of Medicine, Johns Hopkins University, Baltimore, Maryland. The Sidney Kimmel Comprehensive Cancer Center, School of Medicine, Johns Hopkins University, Baltimore, Maryland. Department of Obstetrics and Gynecology, School of Medicine, Keio University, Tokyo, Japan
| | - Hiroyasu Kashima
- Department of Pathology, School of Medicine, Johns Hopkins University, Baltimore, Maryland. The Sidney Kimmel Comprehensive Cancer Center, School of Medicine, Johns Hopkins University, Baltimore, Maryland
| | - Ren-Chin Wu
- Department of Pathology, School of Medicine, Johns Hopkins University, Baltimore, Maryland. The Sidney Kimmel Comprehensive Cancer Center, School of Medicine, Johns Hopkins University, Baltimore, Maryland. Department of Pathology, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan
| | - Jin-Gyoung Jung
- Department of Pathology, School of Medicine, Johns Hopkins University, Baltimore, Maryland. The Sidney Kimmel Comprehensive Cancer Center, School of Medicine, Johns Hopkins University, Baltimore, Maryland
| | - Jen-Chun Kuan
- Department of Pathology, School of Medicine, Johns Hopkins University, Baltimore, Maryland. The Sidney Kimmel Comprehensive Cancer Center, School of Medicine, Johns Hopkins University, Baltimore, Maryland
| | - Jinghua Gu
- Department of Electrical and Computer Engineering, Virginia Polytechnic Institute and State University, Blacksburg, Virginia
| | - Jianhua Xuan
- Department of Electrical and Computer Engineering, Virginia Polytechnic Institute and State University, Blacksburg, Virginia
| | - Lori Sokoll
- Department of Pathology, School of Medicine, Johns Hopkins University, Baltimore, Maryland. The Sidney Kimmel Comprehensive Cancer Center, School of Medicine, Johns Hopkins University, Baltimore, Maryland
| | - Kala Visvanathan
- The Sidney Kimmel Comprehensive Cancer Center, School of Medicine, Johns Hopkins University, Baltimore, Maryland. Department of Epidemiology, School of Public Health, Johns Hopkins University, Baltimore, Maryland
| | - Ie-Ming Shih
- Department of Pathology, School of Medicine, Johns Hopkins University, Baltimore, Maryland. The Sidney Kimmel Comprehensive Cancer Center, School of Medicine, Johns Hopkins University, Baltimore, Maryland. Department of Gynecology/Obstetrics, School of Medicine, Johns Hopkins University, Baltimore, Maryland
| | - Tian-Li Wang
- Department of Pathology, School of Medicine, Johns Hopkins University, Baltimore, Maryland. The Sidney Kimmel Comprehensive Cancer Center, School of Medicine, Johns Hopkins University, Baltimore, Maryland. Department of Gynecology/Obstetrics, School of Medicine, Johns Hopkins University, Baltimore, Maryland.
| |
Collapse
|
239
|
Mohamed AD, Tremblay AM, Murray GI, Wackerhage H. The Hippo signal transduction pathway in soft tissue sarcomas. Biochim Biophys Acta Rev Cancer 2015; 1856:121-9. [PMID: 26050962 DOI: 10.1016/j.bbcan.2015.05.006] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Revised: 05/27/2015] [Accepted: 05/28/2015] [Indexed: 12/11/2022]
Abstract
Sarcomas are rare cancers (≈1% of all solid tumours) usually of mesenchymal origin. Here, we review evidence implicating the Hippo pathway in soft tissue sarcomas. Several transgenic mouse models of Hippo pathway members (Nf2, Mob1, LATS1 and YAP1 mutants) develop various types of sarcoma. Despite that, Hippo member genes are rarely point mutated in human sarcomas. Instead, WWTR1-CAMTA1 and YAP1-TFE3 fusion genes are found in almost all cases of epithelioid haemangioendothelioma. Also copy number gains of YAP1 and other Hippo members occur at low frequencies but the most likely cause of perturbed Hippo signalling in sarcoma is the cross-talk with commonly mutated cancer genes such as KRAS, PIK3CA, CTNNB1 or FBXW7. Current Hippo pathway-targeting drugs include compounds that target the interaction between YAP and TEAD G protein-coupled receptors (GPCR) and the mevalonate pathway (e.g. statins). Given that many Hippo pathway-modulating drugs are already used in patients, this could lead to early clinical trials testing their efficacy in different types of sarcoma.
Collapse
Affiliation(s)
- Abdalla D Mohamed
- School of Medical Sciences, University of Aberdeen, AB25 2ZD Scotland, UK
| | - Annie M Tremblay
- Stem Cell Program, Children's Hospital, Boston, MA 02115, USA; Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA
| | - Graeme I Murray
- School of Medicine and Dentistry, University of Aberdeen, AB25 2ZD Scotland, UK
| | - Henning Wackerhage
- School of Medical Sciences, University of Aberdeen, AB25 2ZD Scotland, UK.
| |
Collapse
|
240
|
Reznik E, Sander C. Extensive decoupling of metabolic genes in cancer. PLoS Comput Biol 2015; 11:e1004176. [PMID: 25961905 PMCID: PMC4427321 DOI: 10.1371/journal.pcbi.1004176] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2014] [Accepted: 02/04/2015] [Indexed: 12/21/2022] Open
Abstract
Tumorigenesis requires the re-organization of metabolism to support malignant proliferation. We examine how the altered metabolism of cancer cells is reflected in the rewiring of co-expression patterns among metabolic genes. Focusing on breast and clear-cell kidney tumors, we report the existence of key metabolic genes which act as hubs of differential co-expression, showing significantly different co-regulation patterns between normal and tumor states. We compare our findings to those from classical differential expression analysis, and counterintuitively observe that the extent of a gene's differential co-expression only weakly correlates with its differential expression, suggesting that the two measures probe different features of metabolism. Focusing on this discrepancy, we use changes in co-expression patterns to highlight the apparent loss of regulation by the transcription factor HNF4A in clear cell renal cell carcinoma, despite no differential expression of HNF4A. Finally, we aggregate the results of differential co-expression analysis into a Pan-Cancer analysis across seven distinct cancer types to identify pairs of metabolic genes which may be recurrently dysregulated. Among our results is a cluster of four genes, all components of the mitochondrial electron transport chain, which show significant loss of co-expression in tumor tissue, pointing to potential mitochondrial dysfunction in these tumor types.
Collapse
Affiliation(s)
- Ed Reznik
- Computational Biology Program, Memorial Sloan Kettering Cancer Center, New York, New York, United States of America
- * E-mail:
| | - Chris Sander
- Computational Biology Program, Memorial Sloan Kettering Cancer Center, New York, New York, United States of America
| |
Collapse
|
241
|
Abstract
Liver X receptors (LXRs) are members of the nuclear receptor superfamily of DNA-binding transcription factors and act as sensors of cholesterol homeostasis. Under normal conditions, when intracellular cholesterol concentration increases, cells synthesize oxysterols and activate the LXR transcriptional network to drive cholesterol efflux and reduce cholesterol influx and synthesis. During normal and cancer cell proliferation, there is a net uncoupling between intracellular cholesterol increase and LXR activation resulting from the reduced intracellular oxysterol concentration. This review dissects the novel mechanisms of a previously unrecognized metabolic uncoupling, supporting the activation of the LXR axis as a bona fide therapeutic approach in cancer.
Collapse
Affiliation(s)
- Fabiola Bovenga
- Clinica Medica Cesare Frugoni, Dipartimento Interdisciplinare di Medicina, University of Bari Aldo Moro, 70124 Bari, Italy; National Cancer Institute, IRCCS Istituto Oncologico Giovanni Paolo II, 70124 Bari, Italy
| | - Carlo Sabbà
- Clinica Medica Cesare Frugoni, Dipartimento Interdisciplinare di Medicina, University of Bari Aldo Moro, 70124 Bari, Italy
| | - Antonio Moschetta
- Clinica Medica Cesare Frugoni, Dipartimento Interdisciplinare di Medicina, University of Bari Aldo Moro, 70124 Bari, Italy; National Cancer Institute, IRCCS Istituto Oncologico Giovanni Paolo II, 70124 Bari, Italy.
| |
Collapse
|
242
|
Bjarnadottir O, Kimbung S, Johansson I, Veerla S, Jönsson M, Bendahl PO, Grabau D, Hedenfalk I, Borgquist S. Global Transcriptional Changes Following Statin Treatment in Breast Cancer. Clin Cancer Res 2015; 21:3402-11. [PMID: 25840970 DOI: 10.1158/1078-0432.ccr-14-1403] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2014] [Accepted: 03/29/2015] [Indexed: 11/16/2022]
Abstract
BACKGROUND Statins purportedly exert antitumoral effects, but the underlying mechanisms are currently not fully elucidated. The aim of this study was to explore potential statin-induced effects on global gene expression profiles in primary breast cancer. EXPERIMENTAL DESIGN This window-of-opportunity phase II trial enrolled 50 newly diagnosed breast cancer patients prescribed atorvastatin (80 mg/day) for 2 weeks presurgically. Pre- and posttreatment tumor samples were analyzed using Significance Analysis of Microarrays (SAM) to identify differentially expressed genes. Similarly, SAM and gene ontology analyses were applied to gene expression data derived from atorvastatin-treated breast cancer cell lines (MCF7, BT474, SKBR3, and MDAMB231) comparing treated and untreated cells. The Systematic Motif Analysis Retrieval Tool (SMART) was used to identify enriched transcription factor-binding sites. Literature Vector Analysis (LitVAn) identified gene module functionality, and pathway analysis was performed using GeneGo Pathways Software (MetaCore; https://portal.genego.com/). RESULTS Comparative analysis of gene expression profiles in paired clinical samples revealed 407 significantly differentially expressed genes (FDR = 0); 32 upregulated and 375 downregulated genes. Restricted filtration (fold change ≥1.49) resulted in 21 upregulated and 46 downregulated genes. Significantly upregulated genes included DUSP1, RHOB1, GADD45B, and RGS1. Pooled results from gene ontology, LitVAn and SMART analyses identified statin-induced effects on the apoptotic and MAPK pathways among others. Comparative analyses of gene expression profiles in breast cancer cell lines showed significant upregulation of the mevalonate and proapoptotic pathways following atorvastatin treatment. CONCLUSIONS We report potential statin-induced changes in global tumor gene expression profiles, indicating MAPK pathway inhibition and proapoptotic events.
Collapse
Affiliation(s)
- Olöf Bjarnadottir
- Division of Oncology and Pathology, Department of Clinical Sciences, Lund University, Lund, Sweden. Department of Oncology, Skåne University Hospital, Lund, Sweden
| | - Siker Kimbung
- Division of Oncology and Pathology, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Ida Johansson
- Division of Oncology and Pathology, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Srinivas Veerla
- Division of Oncology and Pathology, Department of Clinical Sciences, Lund University, Lund, Sweden. SciBlu genomics, Lund University, Sweden
| | - Mats Jönsson
- Division of Oncology and Pathology, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Pär-Ola Bendahl
- Division of Oncology and Pathology, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Dorthe Grabau
- Division of Oncology and Pathology, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Ingrid Hedenfalk
- Division of Oncology and Pathology, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Signe Borgquist
- Division of Oncology and Pathology, Department of Clinical Sciences, Lund University, Lund, Sweden. Department of Oncology, Skåne University Hospital, Lund, Sweden.
| |
Collapse
|
243
|
Abstract
Whereas resting T cells, which have low metabolic requirements, use oxidative phosphorylation (OXPHOS) to maximize their generation of ATP, activated T cells, similar to tumor cells, shift metabolic activity to aerobic glycolysis, which also fuels mevalonate metabolism. Both sterol and nonsterol derivatives of mevalonate affect T cell function. The intracellular availability of sterols, which is dynamically regulated by different classes of transcription factors, represents a metabolic checkpoint that modulates T cell responses. The electron carrier ubiquinone, which is modified with an isoprenoid membrane anchor, plays a pivotal role in OXPHOS, which supports the proliferation of T cells. Isoprenylation also mediates the plasma membrane attachment of the Ras, Rho, and Rab guanosine triphosphatases, which are involved in T cell immunological synapse formation, migration, proliferation, and cytotoxic effector responses. Finally, multiple phosphorylated mevalonate derivatives can act as danger signals for innate-like γδ T cells, thus contributing to the immune surveillance of stress, pathogens, and tumors. We highlight the importance of the mevalonate pathway in the metabolic reprogramming of effector and regulatory T cells.
Collapse
Affiliation(s)
- Martin Thurnher
- Immunotherapy Unit, Department of Urology, Medical University of Innsbruck and Oncotyrol, K1 Center for Personalized Cancer Medicine, 6020 Innsbruck, Austria.
| | - Georg Gruenbacher
- Immunotherapy Unit, Department of Urology, Medical University of Innsbruck and Oncotyrol, K1 Center for Personalized Cancer Medicine, 6020 Innsbruck, Austria
| |
Collapse
|
244
|
Squalene epoxidase (SQLE) promotes the growth and migration of the hepatocellular carcinoma cells. Tumour Biol 2015; 36:6173-9. [DOI: 10.1007/s13277-015-3301-x] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2015] [Accepted: 03/01/2015] [Indexed: 01/22/2023] Open
|
245
|
Ahern TP, Lash TL, Damkier P, Christiansen PM, Cronin-Fenton DP. Statins and breast cancer prognosis: evidence and opportunities. Lancet Oncol 2015; 15:e461-8. [PMID: 25186049 DOI: 10.1016/s1470-2045(14)70119-6] [Citation(s) in RCA: 138] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Much preclinical and epidemiological evidence supports the anticancer effects of statins. Epidemiological evidence does not suggest an association between statin use and reduced incidence of breast cancer, but does support a protective effect of statins--especially simvastatin--on breast cancer recurrence. Here, we argue that the existing evidence base is sufficient to justify a clinical trial of breast cancer adjuvant therapy with statins and we advocate for such a trial to be initiated without delay. If a protective effect of statins on breast cancer recurrence is supported by trial evidence, then the indications for a safe, well tolerated, and inexpensive treatment can be expanded to improve outcomes for breast cancer survivors. We discuss several trial design opportunities--including candidate predictive biomarkers of statin safety and efficacy--and offer solutions to the key challenges involved in the enrolment, follow-up, and analysis of such a trial.
Collapse
Affiliation(s)
- Thomas P Ahern
- Departments of Surgery and Biochemistry, University of Vermont College of Medicine, Burlington, VT, USA.
| | - Timothy L Lash
- Department of Clinical Epidemiology, Aarhus University Hospital, Aarhus, Denmark; Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Per Damkier
- Department of Clinical Biochemistry and Pharmacology, Odense University Hospital, Odense, Denmark
| | - Peer M Christiansen
- Unit of Breast and Endocrine Surgery, Aarhus University Hospital, Aarhus, Denmark; Danish Breast Cancer Cooperative Group, Copenhagen, Denmark
| | | |
Collapse
|
246
|
Greife A, Tukova J, Steinhoff C, Scott SD, Schulz WA, Hatina J. Establishment and characterization of a bladder cancer cell line with enhanced doxorubicin resistance by mevalonate pathway activation. Tumour Biol 2015; 36:3293-300. [DOI: 10.1007/s13277-014-2959-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2014] [Accepted: 12/05/2014] [Indexed: 02/06/2023] Open
|
247
|
McMahon KM, Foit L, Angeloni NL, Giles FJ, Gordon LI, Thaxton CS. Synthetic high-density lipoprotein-like nanoparticles as cancer therapy. Cancer Treat Res 2015; 166:129-50. [PMID: 25895867 PMCID: PMC4418545 DOI: 10.1007/978-3-319-16555-4_6] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/13/2023]
Abstract
High-density lipoproteins (HDL) are diverse natural nanoparticles that carry cholesterol and are best known for the role that they play in cardiovascular disease. However, due to their unique targeting capabilities, diverse molecular cargo, and natural functions beyond cholesterol transport, it is becoming increasingly appreciated that HDLs are critical to cancer development and progression. Accordingly, this chapter highlights ongoing research focused on the connections between HDL and cancer in order to design new drugs and targeted drug delivery vehicles. Research is focused on synthesizing biomimetic HDL-like nanoparticles (NP) that can be loaded with diverse therapeutic cargo (e.g., chemotherapies, nucleic acids, proteins) and specifically targeted to cancer cells. Beyond drug delivery, new data is emerging that HDL-like NPs may be therapeutically active in certain tumor types, for example, B cell lymphoma. Overall, HDL-like NPs are becoming increasingly appreciated as targeted, biocompatible, and efficient therapies for cancer, and may soon become indispensable agents in the cancer therapeutic armamentarium.
Collapse
Affiliation(s)
- Kaylin M. McMahon
- Northwestern University, Feinberg School of Medicine, Department of Urology, Tarry 16-703, 303 E. Chicago Ave. Chicago, IL 60611 United States
- Simpson Querrey Institute (SQI), 303 E. Superior St, Chicago, IL 60611 United States
| | - Linda Foit
- Northwestern University, Feinberg School of Medicine, Department of Urology, Tarry 16-703, 303 E. Chicago Ave. Chicago, IL 60611 United States
- Simpson Querrey Institute (SQI), 303 E. Superior St, Chicago, IL 60611 United States
| | - Nicholas L. Angeloni
- Northwestern University, Feinberg School of Medicine, Department of Urology, Tarry 16-703, 303 E. Chicago Ave. Chicago, IL 60611 United States
- Simpson Querrey Institute (SQI), 303 E. Superior St, Chicago, IL 60611 United States
| | - Francis J. Giles
- Northwestern Medicine Developmental Therapeutics Institute, Northwestern University, 645 N. Michigan Ave, Chicago, IL 60611, USA
| | - Leo I. Gordon
- Department of Medicine, Division of Hematology/Oncology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL 60611
| | - C. Shad Thaxton
- Northwestern University, Feinberg School of Medicine, Department of Urology, Tarry 16-703, 303 E. Chicago Ave. Chicago, IL 60611 United States
- Simpson Querrey Institute (SQI), 303 E. Superior St, Chicago, IL 60611 United States
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL 60611
- International Institute for Nanotechnology (IIN), Northwestern University, 2145 Sheridan Rd. Evanston IL. 60208, United States
- Corresponding Author:
| |
Collapse
|
248
|
Pandyra A, Penn LZ. Targeting tumor cell metabolism via the mevalonate pathway: Two hits are better than one. Mol Cell Oncol 2014; 1:e969133. [PMID: 27308369 PMCID: PMC4905210 DOI: 10.4161/23723548.2014.969133] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2014] [Revised: 08/22/2014] [Accepted: 08/22/2014] [Indexed: 11/19/2022]
Abstract
Statins are promising anticancer agents that target the mevalonate pathway. Tumor cells are sensitive to depletion of mevalonate-derived products but this activity triggers a homeostatic feedback loop that blunts statin efficacy. We showed that dipyridamole inhibits this feedback response and potentiates statin antitumor activity. This study identifies statins plus dypridamole as a preclinically effective combination of approved agents.
Collapse
Affiliation(s)
- Aleksandra Pandyra
- Princess Margaret Cancer Centre, Toronto, ON, Canada; Department of Medical Biophysics; University of Toronto ; Toronto, ON, Canada
| | - Linda Z Penn
- Princess Margaret Cancer Centre, Toronto, ON, Canada; Department of Medical Biophysics; University of Toronto ; Toronto, ON, Canada
| |
Collapse
|
249
|
Foit L, Giles FJ, Gordon LI, Thaxton CS. Synthetic high-density lipoprotein-like nanoparticles for cancer therapy. Expert Rev Anticancer Ther 2014; 15:27-34. [PMID: 25487833 DOI: 10.1586/14737140.2015.990889] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
High-density lipoproteins (HDLs) are a diverse group of natural nanoparticles that are most well known for their role in cholesterol transport. However, HDLs have diverse functions that provide significant opportunities for cancer therapy. Presented is a focused review of the ways that synthetic versions of HDL have been used as targeted therapies for cancer, and as vehicles for the delivery of diverse therapeutic cargo to cancer cells. As such, synthetic HDLs are likely to play a central role in the development of next-generation cancer therapies.
Collapse
Affiliation(s)
- Linda Foit
- Department of Urology, Feinberg School of Medicine, Northwestern University, Tarry 16-703, 303 E. Chicago Ave. Chicago, IL 60611, USA
| | | | | | | |
Collapse
|
250
|
Parente-Pereira AC, Shmeeda H, Whilding LM, Zambirinis CP, Foster J, van der Stegen SJC, Beatson R, Zabinski T, Brewig N, Sosabowski JK, Mather S, Ghaem-Maghami S, Gabizon A, Maher J. Adoptive Immunotherapy of Epithelial Ovarian Cancer with Vγ9Vδ2 T Cells, Potentiated by Liposomal Alendronic Acid. THE JOURNAL OF IMMUNOLOGY 2014; 193:5557-5566. [DOI: 10.4049/jimmunol.1402200] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
Abstract
Abstract
Adoptive immunotherapy using γδ T cells harnesses their natural role in tumor immunosurveillance. The efficacy of this approach is enhanced by aminobisphosphonates such as zoledronic acid and alendronic acid, both of which promote the accumulation of stimulatory phosphoantigens in target cells. However, the inefficient and nonselective uptake of these agents by tumor cells compromises the effective clinical exploitation of this principle. To overcome this, we have encapsulated aminobisphosphonates within liposomes. Expanded Vγ9Vδ2 T cells from patients and healthy donors displayed similar phenotype and destroyed autologous and immortalized ovarian tumor cells, following earlier pulsing with either free or liposome-encapsulated aminobisphosphonates. However, liposomal zoledronic acid proved highly toxic to SCID Beige mice. By contrast, the maximum tolerated dose of liposomal alendronic acid was 150-fold higher, rendering it much more suited to in vivo use. When injected into the peritoneal cavity, free and liposomal alendronic acid were both highly effective as sensitizing agents, enabling infused γδ T cells to promote the regression of established ovarian tumors by over one order of magnitude. Importantly however, liposomal alendronic acid proved markedly superior compared with free drug following i.v. delivery, exploiting the “enhanced permeability and retention effect” to render advanced tumors susceptible to γδ T cell–mediated shrinkage. Although folate targeting of liposomes enhanced the sensitization of folate receptor–α+ ovarian tumor cells in vitro, this did not confer further therapeutic advantage in vivo. These findings support the development of an immunotherapeutic approach for ovarian and other tumors in which adoptively infused γδ T cells are targeted using liposomal alendronic acid.
Collapse
Affiliation(s)
- Ana C. Parente-Pereira
- *King’s College London, King’s Health Partners Integrated Cancer Centre, London SE1 9RT, United Kingdom
| | - Hilary Shmeeda
- †Shaare Zedek Medical Center, Department of Oncology, Jerusalem 91031, Israel
| | - Lynsey M. Whilding
- *King’s College London, King’s Health Partners Integrated Cancer Centre, London SE1 9RT, United Kingdom
- ‡Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital, London W12 0HS, United Kingdom
| | | | - Julie Foster
- §Centre for Molecular Oncology and Imaging, John Vane Science Centre, Institute of Cancer, Barts and the London School of Medicine and Dentistry, London EC1M 6BQ, United Kingdom
| | | | - Richard Beatson
- *King’s College London, King’s Health Partners Integrated Cancer Centre, London SE1 9RT, United Kingdom
| | - Tomasz Zabinski
- *King’s College London, King’s Health Partners Integrated Cancer Centre, London SE1 9RT, United Kingdom
| | - Nancy Brewig
- ‡Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital, London W12 0HS, United Kingdom
| | - Jane K. Sosabowski
- §Centre for Molecular Oncology and Imaging, John Vane Science Centre, Institute of Cancer, Barts and the London School of Medicine and Dentistry, London EC1M 6BQ, United Kingdom
| | - Stephen Mather
- §Centre for Molecular Oncology and Imaging, John Vane Science Centre, Institute of Cancer, Barts and the London School of Medicine and Dentistry, London EC1M 6BQ, United Kingdom
| | - Sadaf Ghaem-Maghami
- ‡Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital, London W12 0HS, United Kingdom
| | - Alberto Gabizon
- †Shaare Zedek Medical Center, Department of Oncology, Jerusalem 91031, Israel
- ¶Hebrew University School of Medicine, Jerusalem 91905, Israel
| | - John Maher
- *King’s College London, King’s Health Partners Integrated Cancer Centre, London SE1 9RT, United Kingdom
- ‖Department of Immunology, Barnet Hospital, Barnet, Hertfordshire EN5 3DJ, United Kingdom; and
- #Department of Clinical Immunology and Allergy, King’s College Hospital National Health Service Foundation Trust, London SE5 9RS, United Kingdom
| |
Collapse
|