51
|
Kumar S, Singh R, Malik S, Manne U, Mishra M. Prostate cancer health disparities: An immuno-biological perspective. Cancer Lett 2018; 414:153-165. [PMID: 29154974 PMCID: PMC5743619 DOI: 10.1016/j.canlet.2017.11.011] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Revised: 11/09/2017] [Accepted: 11/11/2017] [Indexed: 02/07/2023]
Abstract
Prostate cancer (PCa) is the most commonly diagnosed malignancy in males, and, in the United States, is the second leading cause of cancer-related death for men older than 40 years. There is a higher incidence of PCa for African Americans (AAs) than for European-Americans (EAs). Investigations related to the incidence of PCa-related health disparities for AAs suggest that there are differences in the genetic makeup of these populations. Other differences are environmentally induced (e.g., diet and lifestyle), and the exposures are different. Men who immigrate from Eastern to Western countries have a higher risk of PCa than men in their native countries. However, the number of immigrants developing PCa is still lower than that of men in Western countries, suggesting that genetic factors are involved in the development of PCa. Altered genetic polymorphisms are associated with PCa progression. Androgens and the androgen receptor (AR) are involved in the development and progression of PCa. For populations with diverse racial/ethnic backgrounds, differences in lifestyle, diet, and biology, including genetic mutations/polymorphisms and levels of androgens and AR, are risk factors for PCa. Here, we provide an immuno-biological perspective on PCa in relation to racial/ethnic disparities and identify factors associated with the disproportionate incidence of PCa and its clinical outcomes.
Collapse
Affiliation(s)
- Sanjay Kumar
- Cancer Biology Research and Training Program, Department of Biological Sciences, Alabama State University, Montgomery, AL 36104, USA
| | - Rajesh Singh
- Department of Microbiology, Biochemistry, and Immunology, Morehouse School of Medicine, Atlanta, GA 30310, USA
| | - Shalie Malik
- Cancer Biology Research and Training Program, Department of Biological Sciences, Alabama State University, Montgomery, AL 36104, USA; Department of Zoology, University of Lucknow, Lucknow 226007, India
| | - Upender Manne
- Department of Pathology, Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Manoj Mishra
- Cancer Biology Research and Training Program, Department of Biological Sciences, Alabama State University, Montgomery, AL 36104, USA.
| |
Collapse
|
52
|
Schiff D, Jaeckle KA, Anderson SK, Galanis E, Giannini C, Buckner JC, Stella P, Flynn PJ, Erickson BJ, Schwerkoske JF, Kaluza V, Twohy E, Dancey J, Wright J, Sarkaria JN. Phase 1/2 trial of temsirolimus and sorafenib in the treatment of patients with recurrent glioblastoma: North Central Cancer Treatment Group Study/Alliance N0572. Cancer 2018; 124:1455-1463. [PMID: 29313954 DOI: 10.1002/cncr.31219] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Revised: 10/17/2017] [Accepted: 11/10/2017] [Indexed: 11/08/2022]
Abstract
BACKGROUND Mitogen-activated protein kinase (MAPK) activation and mammalian target of rapamycin (mTOR)-dependent signaling are hallmarks of glioblastoma. In the current study, the authors conducted a phase 1/2 study of sorafenib (an inhibitor of Raf kinase and vascular endothelial growth factor receptor 2 [VEGFR-2]) and the mTOR inhibitor temsirolimus in patients with recurrent glioblastoma. METHODS Patients with recurrent glioblastoma who developed disease progression after surgery or radiotherapy plus temozolomide and with ≤2 prior chemotherapy regimens were eligible. The phase 1 endpoint was the maximum tolerated dose (MTD), using a cohorts-of-3 design. The 2-stage phase 2 study included separate arms for VEGF inhibitor (VEGFi)-naive patients and patients who progressed after prior VEGFi. RESULTS The MTD was sorafenib at a dose of 200 mg twice daily and temsirolimus at a dose of 20 mg weekly. In the first 41 evaluable patients who were treated at the phase 2 dose, there were 7 who were free of disease progression at 6 months (progression-free survival at 6 months [PFS6]) in the VEGFi-naive group (17.1%); this finding met the prestudy threshold of success. In the prior VEGFi group, only 4 of the first 41 evaluable patients treated at the phase 2 dose achieved PFS6 (9.8%), and this did not meet the prestudy threshold for success. The median PFS for the 2 groups was 2.6 months and 1.9 months, respectively. The median overall survival for the 2 groups was 6.3 months and 3.9 months, respectively. At least 1 adverse event of grade ≥3 was observed in 75.5% of the VEGFi-naive patients and in 73.9% of the prior VEGFi patients. CONCLUSIONS The limited activity of sorafenib and temsirolimus at the dose and schedule used in the current study was observed with considerable toxicity of grade ≥3. Significant dose reductions that were required in this treatment combination compared with tolerated single-agent doses may have contributed to the lack of efficacy. Cancer 2018;124:1455-63. © 2018 American Cancer Society.
Collapse
Affiliation(s)
- David Schiff
- Division of Neuro-Oncology, Neuro-Oncology Center, University of Virginia Health System, Charlottesville, Virginia
| | | | - S Keith Anderson
- Alliance Statistics and Data Center, Mayo Clinic, Rochester, Minnesota
| | - Evanthia Galanis
- Department of Medical Oncology, Mayo Clinic, Rochester, Minnesota
| | - Caterina Giannini
- Alliance Statistics and Data Center, Mayo Clinic, Rochester, Minnesota
| | - Jan C Buckner
- Department of Medical Oncology, Mayo Clinic, Rochester, Minnesota
| | - Phillip Stella
- Medical Oncology Program, St. Joseph Mercy Health System, Ann Arbor, Michigan
| | - Patrick J Flynn
- Metro Minnesota Community Clinical Oncology Program, St. Louis Park, Minnesota
| | | | | | - Vesna Kaluza
- Medical Oncology Program, St. Joseph Mercy Health System, Ann Arbor, Michigan
| | - Erin Twohy
- Alliance Statistics and Data Center, Mayo Clinic, Rochester, Minnesota
| | - Janet Dancey
- Cancer Therapy Evaluation Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Rockville, Maryland
| | - John Wright
- Cancer Therapy Evaluation Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Rockville, Maryland
| | - Jann N Sarkaria
- Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
53
|
Ho CM, Lee FK, Huang SH, Cheng WF. Everolimus following 5-aza-2-deoxycytidine is a promising therapy in paclitaxel-resistant clear cell carcinoma of the ovary. Am J Cancer Res 2018; 8:56-69. [PMID: 29416920 PMCID: PMC5794721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Accepted: 11/29/2017] [Indexed: 06/08/2023] Open
Abstract
Our previous study showed that 5-aza-2-deoxycytidine (5-aza-dC) could inhibit tumor growth by enhancing the susceptibility of ovarian clear cell carcinoma (OCCC) to paclitaxel through decreasing AKT/mTOR expressions. The objective of the study is to evaluate the antitumor efficacy of everolimus (RAD001) and 5-aza-2-deoxycytidine (5-aza-dC) by targeting AKT/mTOR and EZH2 in OCCC. Paclitaxel-sensitive and resistant OCCC cell lines were established. In vitro proliferative and apoptotic assays and flow cytometry were performed. The expressions of EZH2, PIK3IP1, phospho-AKT, phospho-mTOR and phospho-Rictor in the OCCC cell lines were evaluated by Western blotting. In vivo animal experiments with RAD001 and 5-aza-dC were performed. RAD001 alone showed significant in vitro antitumor activity and inhibited in vivo tumor growth in paclitaxel-sensitive and resistant OCCC cells. In addition, 5-aza-dC enhanced the antitumor effects when combined with paclitaxel or RAD001 in both paclitaxel-sensitive and resistant tumors. Activation of phospho-AKT ser473 and PIK3IP1 was observed in RAD001-treated paclitaxel-sensitive and resistant OCCC cells. In contrast, inhibition of phospho-AKT ser473 and EZH2 was observed with RAD001 following 5-aza-dC treatment of paclitaxel-sensitive and resistant OCCC cells. Furthermore, RAD001 following 5-aza-dC enhanced apoptosis of paclitaxel-sensitive and resistant OCCC cells. RAD001 following 5-aza-dC may be a promising treatment strategy for the treatment of both chemo-sensitive and resistant OCCC. Further clinical studies are warranted.
Collapse
Affiliation(s)
- Chih-Ming Ho
- Gynecologic Cancer Center, Department of Obstetrics and GynecologyTaipei, Taiwan
- School of Medicine, Fu Jen Catholic UniversityHsinchuang, New Taipei, Taiwan
- School of Medicine, Taipei Medical UniversityTaipei, Taiwan
| | - Fa-Kung Lee
- Gynecologic Cancer Center, Department of Obstetrics and GynecologyTaipei, Taiwan
- School of Medicine, Fu Jen Catholic UniversityHsinchuang, New Taipei, Taiwan
| | - Shih-Hung Huang
- Department of Pathology, Cathay General HospitalTaipei, Taiwan
| | - Wen-Fang Cheng
- Department of Obstetrics and Gynecology, College of Medicine, National Taiwan UniversityTaipei, Taiwan
- Graduate Institute of Oncology, College of Medicine, National Taiwan UniversityTaipei, Taiwan
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan UniversityTaipei, Taiwan
| |
Collapse
|
54
|
Prognostic and predictive role of the PI3K-AKT-mTOR pathway in neuroendocrine neoplasms. Clin Transl Oncol 2017; 20:561-569. [PMID: 29124519 DOI: 10.1007/s12094-017-1758-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2017] [Accepted: 09/30/2017] [Indexed: 12/20/2022]
Abstract
Neuroendocrine neoplasms (NENs) are considered a heterogeneous and rare entity. Its natural history is influenced by multiple clinicopathological characteristics, which guide the management of these patients. The development of molecular biology reveals that the PI3K-AKT-mTOR pathway plays a relevant role in tumorigenesis and progression of NENs. Mammalian target of rapamycin (mTOR) inhibitors, targeted agents that block this pathway, has improved outcomes in neuroendocrine tumors (NETs). Different therapeutic approaches, such as somatostatin analogs, chemotherapy, peptide receptor radionuclide therapy, and targeted agents, have shown benefits in the treatment of NETs. However, there are not any established prognostic or predictive biomarkers to select the best therapy option to individualize treatment. Although a relation between alterations in the PI3K-AKT-mTOR pathway and clinical outcomes has not been found, these anomalies are considered attractive biomarkers. Additional molecular analysis should be integrated in future clinical trials' design to identify potential predictive or prognostic biomarkers.
Collapse
|
55
|
Song W, Filonov GS, Kim H, Hirsch M, Li X, Moon JD, Jaffrey SR. Imaging RNA polymerase III transcription using a photostable RNA-fluorophore complex. Nat Chem Biol 2017; 13:1187-1194. [PMID: 28945233 PMCID: PMC5679246 DOI: 10.1038/nchembio.2477] [Citation(s) in RCA: 188] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2016] [Accepted: 08/09/2017] [Indexed: 11/09/2022]
Abstract
Quantitative measurement of transcription rates in live cells is important for revealing mechanisms of transcriptional regulation. This is particularly challenging when measuring the activity of RNA polymerase III (Pol III), which transcribes growth-promoting small RNAs. To address this issue, we developed Corn, a genetically encoded fluorescent RNA reporter suitable for quantifying RNA transcription in cells. Corn binds and induces fluorescence of 3,5-difluoro-4-hydroxybenzylidene-imidazolinone-2-oxime, which resembles the fluorophore found in red fluorescent protein (RFP). Notably, Corn shows high photostability, enabling quantitative fluorescence imaging of mTOR-dependent Pol III transcription. We found that, unlike actinomycin D, mTOR inhibitors resulted in heterogeneous transcription suppression in individual cells. Quantitative imaging of Corn-tagged Pol III transcript levels revealed distinct Pol III transcription 'trajectories' elicited by mTOR inhibition. Together, these studies provide an approach for quantitative measurement of Pol III transcription by direct imaging of Pol III transcripts containing a photostable RNA-fluorophore complex.
Collapse
Affiliation(s)
- Wenjiao Song
- Department of Pharmacology, Weill Medical College, Cornell University, New York, NY 10065, USA
| | - Grigory S. Filonov
- Department of Pharmacology, Weill Medical College, Cornell University, New York, NY 10065, USA
| | - Hyaeyeong Kim
- Department of Pharmacology, Weill Medical College, Cornell University, New York, NY 10065, USA
| | - Markus Hirsch
- Department of Pharmacology, Weill Medical College, Cornell University, New York, NY 10065, USA
| | - Xing Li
- Department of Pharmacology, Weill Medical College, Cornell University, New York, NY 10065, USA
| | - Jared D. Moon
- Department of Pharmacology, Weill Medical College, Cornell University, New York, NY 10065, USA
| | - Samie R. Jaffrey
- Department of Pharmacology, Weill Medical College, Cornell University, New York, NY 10065, USA
| |
Collapse
|
56
|
Evolving Significance and Future Relevance of Anti-Angiogenic Activity of mTOR Inhibitors in Cancer Therapy. Cancers (Basel) 2017; 9:cancers9110152. [PMID: 29104248 PMCID: PMC5704170 DOI: 10.3390/cancers9110152] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Revised: 10/23/2017] [Accepted: 10/27/2017] [Indexed: 12/12/2022] Open
Abstract
mTOR inhibitors have demonstrated remarkable anti-tumor activity in experimental models, mainly by reducing cancer cell growth and tumor angiogenesis. Their use in cancer patients as monotherapy has, however, generated only limited benefits, increasing median overall survival by only a few months. Likewise, in other targeted therapies, cancer cells develop resistance mechanisms to overcome mTOR inhibition. Hence, novel therapeutic strategies have to be designed to increase the efficacy of mTOR inhibitors in cancer. In this review, we discuss the present and future relevance of mTOR inhibitors in cancer therapy by focusing on their effects on tumor angiogenesis.
Collapse
|
57
|
Abstract
Background: The prognosis for most patients with primary brain tumors remains poor. Recent advances in molecular and cell biology have led to a greater understanding of molecular alterations in brain tumors. These advances are being translated into new therapies that will hopefully improve the prognosis for patients with brain tumors. Methods: We reviewed the literature on small molecule targeted agents and monoclonal antibodies used in brain tumor research and brain tumor clinical trials for the past 20 years. Results: Brain tumors commonly express molecular abnormalities. These alterations can lead to the activation of cell pathways involved in cell proliferation. This knowledge has led to interest in novel anti-brain-tumor therapies targeting key components of these pathways. Many drugs and monoclonal antibodies have been developed that modulate these pathways and are in various stages of testing. Conclusions: The use of targeted therapies against brain tumors promises to improve the prognosis for patients with brain tumors. However, as the molecular pathogenesis of brain tumors has not been linked to a single genetic defect or target, molecular agents may need to be used in combinations or in tandem with cytotoxic agents. Further study of these agents in well-designed cooperative clinical trials is needed.
Collapse
Affiliation(s)
- Nicholas Butowski
- Department of Neurological Surgery, University of California, San Francisco, CA 94143-0350, USA
| | | |
Collapse
|
58
|
Gross ME, Dorff TB, Quinn DI, Diaz PM, Castellanos OO, Agus DB. Safety and Efficacy of Docetaxel, Bevacizumab, and Everolimus for Castration-resistant Prostate Cancer (CRPC). Clin Genitourin Cancer 2017; 16:S1558-7673(17)30200-8. [PMID: 28826933 PMCID: PMC7444943 DOI: 10.1016/j.clgc.2017.07.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Revised: 06/16/2017] [Accepted: 07/09/2017] [Indexed: 11/17/2022]
Abstract
BACKGROUND Previous data suggests that co-targeting mammalian target of rapamycin and angiogenic pathways may potentiate effects of cytotoxic chemotherapy. We studied combining mammalian target of rapamycin and vascular endothelial growth factor inhibition with docetaxel in castrate-resistant prostate cancer (CRPC). METHODS Eligible patients had progressive, metastatic, chemotherapy-naive CRPC. Docetaxel and bevacizumab were given intravenously day 1 with everolimus orally daily on a 21-day cycle across 3 dose levels (75:15:2.5, 75:15:5, and 65:15:5; docetaxel mg/m2, mg/kg bevacizumab, and mg everolimus, respectively). Maintenance therapy with bevacizumab/everolimus without docetaxel was allowed after ≥ 6 cycles. RESULTS Forty-three subjects were treated across all dose levels. Maximal tolerated doses for the combined therapies observed in the phase 1B portion of the trial were: docetaxel 75 mg/m2, bevacizumab 15 mg/kg, and everolimus 2.5 mg. Maximal prostate-specific antigen decline ≥ 30% and ≥ 50% was achieved in 33 (79%) and 31 (74%) of patients, respectively. Best response by modified Response Evaluation Criteria In Solid Tumors criteria in 25 subjects with measurable disease at baseline included complete or partial response in 20 (80%) patients. The median progression-free and overall survival were 8.9 months (95% confidence interval, 7.4-10.6 months) and 21.9 months (95% confidence interval, 18.4-30.3 months), respectively. Hematologic toxicities were the most common treatment-related grade ≥ 3 adverse events including: febrile neutropenia (12; 28%), lymphopenia (12; 28%), leukocytes (10; 23%), neutrophils (9; 21%), and hemoglobin (2; 5%). Nonhematologic grade ≥ 3 adverse events included: hypertension (8; 19%), fatigue (3; 7%), pneumonia (3; 7%), and mucositis (4; 5%). There was 1 treatment-related death owing to neutropenic fever and pneumonia in a patient treated at dose level 3 despite dose modifications and prophylactic growth factor support. CONCLUSIONS Docetaxel, bevacizumab, and everolimus can be safely administered in CRPC and demonstrate a significant level of anticancer activity, meeting the predetermined response criteria. However, any potential benefit of combined therapy must be balanced against increased risk for toxicities. Our results do not support the hypothesis that this combination of agents improves upon the results obtained with docetaxel monotherapy in an unselected population of chemotherapy-naive patients with CRPC.
Collapse
Affiliation(s)
- Mitchell E Gross
- Lawrence J. Ellison Institute for Transformative Medicine, University of Southern California, Los Angeles, CA; Department of Medicine/Oncology, Keck School of Medicine, University of Southern California, Los Angeles, CA.
| | - Tanya B Dorff
- Department of Medicine/Oncology, Keck School of Medicine, University of Southern California, Los Angeles, CA; USC Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA
| | - David I Quinn
- Department of Medicine/Oncology, Keck School of Medicine, University of Southern California, Los Angeles, CA; USC Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA
| | - Patricia M Diaz
- Department of Medicine/Oncology, Keck School of Medicine, University of Southern California, Los Angeles, CA
| | - Olga O Castellanos
- Department of Medicine/Oncology, Keck School of Medicine, University of Southern California, Los Angeles, CA
| | - David B Agus
- Lawrence J. Ellison Institute for Transformative Medicine, University of Southern California, Los Angeles, CA; Department of Medicine/Oncology, Keck School of Medicine, University of Southern California, Los Angeles, CA
| |
Collapse
|
59
|
Abstract
Post translational modifications (PTMs) are involved in variety of cellular activities and phosphorylation is one of the most extensively studied PTM, which regulates a number of cellular functions like cell growth, differentiation, apoptosis and cell signaling in healthy condition. However, alterations in phosphorylation pathways result in serious outcomes in the form of diseases, especially cancer. Many signalling pathways including Tyrosine kinase, MAP kinase, Cadherin-catenin complex, Cyclin-dependent kinase etc. are major players of the cell cycle and deregulation in their phosphorylation-dephosphorylation cascade has been shown to be manifested in the form of various types of cancers. Tyrosine kinase family encompasses the greatest number of oncoproteins. MAPK cascade has an importance role in cancer growth and progression. Bcl-2 family proteins serve either proapoptotic or antiapoptotic function. Cadherin-catenin complex regulates cell adhesion properties and cyclins are the key regulators of cell cycle. Altered phosphorylations in any of the above pathways are strongly associated with cancer, at the same time they serve as the potential tergets for drug development against cancer. Drugs targeting tyrosine kinase are potent anticancer drugs. Inhibitors of MEK, PI3K and ERK signalling pathways are undergoing clinical trials. Thus, drugs targeting phosphorylation pathways represent a promising area for cancer therapy.
Collapse
Affiliation(s)
- Vishakha Singh
- Department of Pharmacology and Toxicology, Ranchi Veterinary College, BAU, Kanke, Ranchi, Jharkhand, 834006, India
| | - Mahendra Ram
- Department of Pharmacology and Toxicology, Ranchi Veterinary College, BAU, Kanke, Ranchi, Jharkhand, 834006, India.
| | - Rajesh Kumar
- Department of Livestock Products Technology, Ranchi Veterinary College, BAU, Kanke, Ranchi, Jharkhand, 834006, India
| | - Raju Prasad
- Department of Pharmacology and Toxicology, Ranchi Veterinary College, BAU, Kanke, Ranchi, Jharkhand, 834006, India
| | - Birendra Kumar Roy
- Department of Pharmacology and Toxicology, Ranchi Veterinary College, BAU, Kanke, Ranchi, Jharkhand, 834006, India
| | - Kaushal Kumar Singh
- Department of Pathology, Ranchi Veterinary College, BAU, Kanke, Ranchi, Jharkhand, 834006, India
| |
Collapse
|
60
|
Ngeow J, Sesock K, Eng C. Clinical Implications for Germline PTEN Spectrum Disorders. Endocrinol Metab Clin North Am 2017; 46:503-517. [PMID: 28476234 DOI: 10.1016/j.ecl.2017.01.013] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Patients with PTEN hamartoma tumor syndrome (PHTS) may present to a variety of different subspecialties with benign and malignant clinical features. They have increased lifetime risks of breast, endometrial, thyroid, renal, and colon cancers, as well as neurodevelopmental disorders such as autism spectrum disorder. Patients and affected family members can be offered gene-directed surveillance and management. Patients who are unaffected can be spared unnecessary investigations. With longitudinal follow-up, we are likely to identify other non-cancer manifestations associated with PHTS such as metabolic, immunologic, and neurologic features.
Collapse
Affiliation(s)
- Joanne Ngeow
- Cancer Genetics Service, Division of Medical Oncology, National Cancer Centre, 11 Hospital Drive, Singapore 169610, Singapore; Genomic Medicine Institute, Cleveland Clinic, 9500 Euclid Avenue, NE-50, Cleveland, OH 44195, USA
| | - Kaitlin Sesock
- Genomic Medicine Institute, Cleveland Clinic, 9500 Euclid Avenue, NE-50, Cleveland, OH 44195, USA; Lerner Research Institute, Cleveland Clinic, 9500 Euclid Avenue, NE-50, Cleveland, OH 44195, USA; Taussig Cancer Institute, Cleveland Clinic, 9500 Euclid Avenue, NE-50, Cleveland, OH 44195, USA
| | - Charis Eng
- Genomic Medicine Institute, Cleveland Clinic, 9500 Euclid Avenue, NE-50, Cleveland, OH 44195, USA; Lerner Research Institute, Cleveland Clinic, 9500 Euclid Avenue, NE-50, Cleveland, OH 44195, USA; Taussig Cancer Institute, Cleveland Clinic, 9500 Euclid Avenue, NE-50, Cleveland, OH 44195, USA; Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, 10900 Euclid Avenue, Cleveland, OH 44106, USA; Germline High Risk Focus Group, CASE Comprehensive Cancer Center, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, USA.
| |
Collapse
|
61
|
Shen JP, Zhao D, Sasik R, Luebeck J, Birmingham A, Bojorquez-Gomez A, Licon K, Klepper K, Pekin D, Beckett A, Sanchez K, Thomas A, Kuo CC, Du D, Roguev A, Lewis NE, Chang AN, Kreisberg JF, Krogan N, Qi L, Ideker T, Mali P. Combinatorial CRISPR-Cas9 screens for de novo mapping of genetic interactions. Nat Methods 2017; 14:573-576. [PMID: 28319113 PMCID: PMC5449203 DOI: 10.1038/nmeth.4225] [Citation(s) in RCA: 255] [Impact Index Per Article: 31.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Accepted: 02/17/2017] [Indexed: 12/13/2022]
Abstract
We developed a systematic approach to map human genetic networks by combinatorial CRISPR-Cas9 perturbations coupled to robust analysis of growth kinetics. We targeted all pairs of 73 cancer genes with dual guide RNAs in three cell lines, comprising 141,912 tests of interaction. Numerous therapeutically relevant interactions were identified, and these patterns replicated with combinatorial drugs at 75% precision. From these results, we anticipate that cellular context will be critical to synthetic-lethal therapies.
Collapse
Affiliation(s)
- John Paul Shen
- Department of Medicine, Division of Genetics, University of California, San Diego; La Jolla, CA, 92093, USA
- Moores UCSD Cancer Center; La Jolla, CA, 92093, USA
- The Cancer Cell Map Initiative (CCMI), University of California, San Diego; La Jolla, CA, 92093, USA
| | - Dongxin Zhao
- The Cancer Cell Map Initiative (CCMI), University of California, San Diego; La Jolla, CA, 92093, USA
- Department of Bioengineering, University of California, San Diego; La Jolla, CA, 92093, USA
| | - Roman Sasik
- Center for Computational Biology & Bioinformatics, University of California, San Diego; La Jolla, CA, 92093, USA
| | - Jens Luebeck
- Bioinformatics & Systems Biology Program, University of California, San Diego; La Jolla, CA, 92093, USA
| | - Amanda Birmingham
- Center for Computational Biology & Bioinformatics, University of California, San Diego; La Jolla, CA, 92093, USA
| | - Ana Bojorquez-Gomez
- Department of Medicine, Division of Genetics, University of California, San Diego; La Jolla, CA, 92093, USA
| | - Katherine Licon
- Department of Medicine, Division of Genetics, University of California, San Diego; La Jolla, CA, 92093, USA
| | - Kristin Klepper
- Department of Medicine, Division of Genetics, University of California, San Diego; La Jolla, CA, 92093, USA
| | - Daniel Pekin
- Department of Medicine, Division of Genetics, University of California, San Diego; La Jolla, CA, 92093, USA
| | - Alex Beckett
- Department of Medicine, Division of Genetics, University of California, San Diego; La Jolla, CA, 92093, USA
| | - Kyle Sanchez
- Department of Medicine, Division of Genetics, University of California, San Diego; La Jolla, CA, 92093, USA
| | - Alex Thomas
- Bioinformatics & Systems Biology Program, University of California, San Diego; La Jolla, CA, 92093, USA
- Novo Nordisk Center for Biosustainability at the University of California, San Diego; La Jolla, CA 92093, USA
| | - Chih-Chung Kuo
- Department of Bioengineering, University of California, San Diego; La Jolla, CA, 92093, USA
- Novo Nordisk Center for Biosustainability at the University of California, San Diego; La Jolla, CA 92093, USA
| | - Dan Du
- The Cancer Cell Map Initiative (CCMI), University of California, San Diego; La Jolla, CA, 92093, USA
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, Department of Cell and Tissue Biology, University of California, San Francisco, CA 94143, USA
| | - Assen Roguev
- The Cancer Cell Map Initiative (CCMI), University of California, San Diego; La Jolla, CA, 92093, USA
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco CA 94143, USA University of California, San Francisco
| | - Nathan E. Lewis
- Department of Pediatrics, University of California, San Diego; La Jolla, CA, 92093, USA
- Novo Nordisk Center for Biosustainability at the University of California, San Diego; La Jolla, CA 92093, USA
| | - Aaron N. Chang
- Center for Computational Biology & Bioinformatics, University of California, San Diego; La Jolla, CA, 92093, USA
| | - Jason F. Kreisberg
- Department of Medicine, Division of Genetics, University of California, San Diego; La Jolla, CA, 92093, USA
- The Cancer Cell Map Initiative (CCMI), University of California, San Diego; La Jolla, CA, 92093, USA
| | - Nevan Krogan
- The Cancer Cell Map Initiative (CCMI), University of California, San Diego; La Jolla, CA, 92093, USA
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco CA 94143, USA University of California, San Francisco
| | - Lei Qi
- The Cancer Cell Map Initiative (CCMI), University of California, San Diego; La Jolla, CA, 92093, USA
- Department of Bioengineering, Stanford University, Stanford, CA, 94305, USA
| | - Trey Ideker
- Department of Medicine, Division of Genetics, University of California, San Diego; La Jolla, CA, 92093, USA
- Moores UCSD Cancer Center; La Jolla, CA, 92093, USA
- The Cancer Cell Map Initiative (CCMI), University of California, San Diego; La Jolla, CA, 92093, USA
- Center for Computational Biology & Bioinformatics, University of California, San Diego; La Jolla, CA, 92093, USA
| | - Prashant Mali
- Moores UCSD Cancer Center; La Jolla, CA, 92093, USA
- The Cancer Cell Map Initiative (CCMI), University of California, San Diego; La Jolla, CA, 92093, USA
- Department of Bioengineering, University of California, San Diego; La Jolla, CA, 92093, USA
| |
Collapse
|
62
|
Liu Q, Luo Q, Halim A, Song G. Targeting lipid metabolism of cancer cells: A promising therapeutic strategy for cancer. Cancer Lett 2017; 401:39-45. [PMID: 28527945 DOI: 10.1016/j.canlet.2017.05.002] [Citation(s) in RCA: 259] [Impact Index Per Article: 32.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2017] [Revised: 05/03/2017] [Accepted: 05/11/2017] [Indexed: 02/07/2023]
Abstract
One of the most important metabolic hallmarks of cancer cells is deregulation of lipid metabolism. In addition, enhancing de novo fatty acid (FA) synthesis, increasing lipid uptake and lipolysis have also been considered as means of FA acquisition in cancer cells. FAs are involved in various aspects of tumourigenesis and tumour progression. Therefore, targeting lipid metabolism is a promising therapeutic strategy for human cancer. Recent studies have shown that reprogramming lipid metabolism plays important roles in providing energy, macromolecules for membrane synthesis, and lipid signals during cancer progression. Moreover, accumulation of lipid droplets in cancer cells acts as a pivotal adaptive response to harmful conditions. Here, we provide a brief review of the crucial roles of FA metabolism in cancer development, and place emphasis on FA origin, utilization and storage in cancer cells. Understanding the regulation of lipid metabolism in cancer cells has important implications for exploring a new therapeutic strategy for management and treatment of cancer.
Collapse
Affiliation(s)
- Qiuping Liu
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400030, China
| | - Qing Luo
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400030, China
| | - Alexander Halim
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400030, China
| | - Guanbin Song
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400030, China.
| |
Collapse
|
63
|
Dou X, Wang Z, Wang L, Lu W, Ma Y, Xu S. [Analysis of Efficacy of Surgical Treatment for IIIa Small Cell Lung Cancer]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2017; 20:88-92. [PMID: 28228219 PMCID: PMC5972969 DOI: 10.3779/j.issn.1009-3419.2017.02.03] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
BACKGROUND Small cell lung cancer (SCLC) accounts for nearly 15% of all cases of cancer. As a type of highly invasive tumors, SCLC has high degree of malignancy, early and extensive metastasis, and is sensitive to chemotherapy and radiotherapy. The early treatment response rate of SCLC is high but it can also relapse rapidly without any treatment. Its median survival time is merely four to six months. Although many studies on SCLC have been conducted in recent years, its clinical treatment strategies have remained unchanged. The treatment of SCLC is still confined to chemotherapy regimens of etoposide plus cisplatin (EP) and other classic treatments because the surgical treatment of SCLC, particularly for IIIa treatment, has yet to reach a consensus. This study investigated the prognostic factors and clinical therapy effect in the comprehensive treatment of IIIa SCLC after surgical treatment. METHODS This study was conducted through the retrospective analysis of the clinical data of 78 patients with SCLC who underwent surgical treatment in Beijing Chest Hospital affiliated to Capital Medical University between January 1995 and December 1995. Through follow-up, we performed statistical analysis of each patient's gender, age, tumor size, lymph node metastasis, tumor-node-metastasis (TNM) staging, surgical methods, and adjuvant radiation and chemotherapy. RESULTS The median survival in this clinical trial team was 13.93 months. Among the participants, 47 patients accepted neoadjuvant chemotherapy and their median survival were 14.25 months. By contrast, 31 patients accepted postoperative adjuvant chemotherapy and their median survival were 13.83 months. No statistical difference was observed between the two groups. Moreover, 28 patients were of single Lymph node metastasis and their median survival was 17.1 months. By contrast, 50 patients were of multiple lymph node metastasis and their median survival was 11.9 months. Significant statistical difference was observed between the two groups (P<0.01). CONCLUSIONS In performing further evaluation of the status and value of surgical treatment in the comprehensive treatment of SCLC, several patients benefitted from IIIa SCLC surgery with comprehensive treatment.
Collapse
Affiliation(s)
- Xuejun Dou
- Department of Thoracic Surgery, Aerospace Central Hospital, Beijing 100049, China
| | - Zhiyuan Wang
- Department of Thoracic Surgery, Aerospace Central Hospital, Beijing 100049, China
| | - Liang Wang
- Department of Thoracic Surgery, Aerospace Central Hospital, Beijing 100049, China
| | - Weiqiang Lu
- Department of Thoracic Surgery, Aerospace Central Hospital, Beijing 100049, China
| | - Yunlei Ma
- Department of Thoracic Surgery, Aerospace Central Hospital, Beijing 100049, China
| | - Shaofa Xu
- Department of Thoracic Surgery, Beijing Chest Hospital, Beijing 101149, China
| |
Collapse
|
64
|
Mathur D, Stratikopoulos E, Ozturk S, Steinbach N, Pegno S, Schoenfeld S, Yong R, Murty VV, Asara JM, Cantley LC, Parsons R. PTEN Regulates Glutamine Flux to Pyrimidine Synthesis and Sensitivity to Dihydroorotate Dehydrogenase Inhibition. Cancer Discov 2017; 7:380-390. [PMID: 28255082 DOI: 10.1158/2159-8290.cd-16-0612] [Citation(s) in RCA: 101] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Revised: 12/02/2016] [Accepted: 01/17/2017] [Indexed: 01/22/2023]
Abstract
Metabolic changes induced by oncogenic drivers of cancer contribute to tumor growth and are attractive targets for cancer treatment. Here, we found that increased growth of PTEN-mutant cells was dependent on glutamine flux through the de novo pyrimidine synthesis pathway, which created sensitivity to the inhibition of dihydroorotate dehydrogenase, a rate-limiting enzyme for pyrimidine ring synthesis. S-phase PTEN-mutant cells showed increased numbers of replication forks, and inhibitors of dihydroorotate dehydrogenase led to chromosome breaks and cell death due to inadequate ATR activation and DNA damage at replication forks. Our findings indicate that enhanced glutamine flux generates vulnerability to dihydroorotate dehydrogenase inhibition, which then causes synthetic lethality in PTEN-deficient cells due to inherent defects in ATR activation. Inhibition of dihydroorotate dehydrogenase could thus be a promising therapy for patients with PTEN-mutant cancers.Significance: We have found a prospective targeted therapy for PTEN-deficient tumors, with efficacy in vitro and in vivo in tumors derived from different tissues. This is based upon the changes in glutamine metabolism, DNA replication, and DNA damage response which are consequences of inactivation of PTENCancer Discov; 7(4); 380-90. ©2017 AACR.See related article by Brown et al., p. 391This article is highlighted in the In This Issue feature, p. 339.
Collapse
Affiliation(s)
- Deepti Mathur
- Department of Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York.,Department of Integrated Cellular and Molecular Biology, Columbia University, New York, New York
| | - Elias Stratikopoulos
- Department of Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Sait Ozturk
- Department of Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Nicole Steinbach
- Department of Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York.,Department of Integrated Cellular and Molecular Biology, Columbia University, New York, New York
| | - Sarah Pegno
- Department of Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Sarah Schoenfeld
- Department of Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Raymund Yong
- Department of Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York.,Department of Neurosurgery, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Vundavalli V Murty
- Department of Pathology and Cell Biology and Institute for Cancer Genetics, Columbia University, New York, New York
| | - John M Asara
- Division of Signal Transduction, Beth Israel Deaconess Medical Center and Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Lewis C Cantley
- Meyer Cancer Center, Weill Cornell Medical College, New York, New York
| | - Ramon Parsons
- Department of Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York.
| |
Collapse
|
65
|
Lu Z, Wang J, Zheng Y, Yang S, Liu M, Chen X, Wang C, Hou G. Wild-type phosphatase and tensin homolog deleted on chromosome 10 improved the sensitivity of cells to rapamycin through regulating phosphorylation of Akt in esophageal squamous cell carcinoma. Dis Esophagus 2017; 30:1-8. [PMID: 26725440 DOI: 10.1111/dote.12448] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Esophageal squamous cell carcinoma (ESCC) is one of the most frequently diagnosed cancers in China, but the etiology and mode of carcinogenesis of this disease remain poorly understood. Phosphatase and tensin homolog deleted on chromosome 10 (PTEN), as a negative regulator of Akt/mTOR pathway, frequently mutates or is inactive in many cancers. Although mTOR has been thought a promising cancer therapeutic target, the sensitivity of tumor cells to rapamycin was still to be revaluated. In this study, we measured the effects of rapamycin on cell proliferation and phosphorylation of Akt in ESCC cells with varying degrees of differentiation. And then, the relationship between PTEN status and the sensitivity of cells to rapamycin was investigated in EC9706 cells with or without wild-type PTEN in vitro and in vivo. The results demonstrated ESCC cells with poor differentiation were insensitive to rapamycin of high concentration and rapamycin obviously promoted the phosphorylation of Akt in these cells, but it had no obvious effects on p-Akt in cells with well differentiation. Also, we showed that wild-type PTEN improved the sensitivity of poor differentiation cells to rapamycin through inhibiting phosphorylation of Akt in vitro and in vivo. This study explored the possible molecular mechanism of some ESCC cells insensitive to rapamycin and provided a measure for treating ESCC patients with PTEN inactivation using mTOR inhibitors.
Collapse
Affiliation(s)
- Z Lu
- Department of Clinical Pharmacy, School of Pharmaceutical Sciences, Zhengzhou University, China
| | - J Wang
- Department of Clinical Pharmacy, School of Pharmaceutical Sciences, Zhengzhou University, China
| | - Y Zheng
- Department of Clinical Pharmacy, School of Pharmaceutical Sciences, Zhengzhou University, China
| | - S Yang
- Department of Clinical Pharmacy, School of Pharmaceutical Sciences, Zhengzhou University, China
| | - M Liu
- Oncology Department, People's Hospital of Henan Province, China
| | - X Chen
- Department of Clinical Pharmacy, School of Pharmaceutical Sciences, Zhengzhou University, China
| | - C Wang
- Department of Clinical Pharmacy, School of Pharmaceutical Sciences, Zhengzhou University, China.,New Drug Research and Development Centre of Zhengzhou University, Zhengzhou, China
| | - G Hou
- Department of Clinical Pharmacy, School of Pharmaceutical Sciences, Zhengzhou University, China.,New Drug Research and Development Centre of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
66
|
Miyata H, Ashizawa T, Iizuka A, Kondou R, Nonomura C, Sugino T, Urakami K, Asai A, Hayashi N, Mitsuya K, Nakasu Y, Yamaguchi K, Akiyama Y. Combination of a STAT3 Inhibitor and an mTOR Inhibitor Against a Temozolomide-resistant Glioblastoma Cell Line. Cancer Genomics Proteomics 2017; 14:83-91. [PMID: 28031240 DOI: 10.21873/cgp.20021] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Revised: 12/09/2016] [Accepted: 12/15/2016] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Temozolomide-resistant (TMZ-R) glioblastoma is very difficult to treat, and a novel approach to overcome resistance is needed. MATERIALS AND METHODS The efficacy of a combination treatment of STAT3 inhibitor, STX-0119, with rapamycin was investigated against our established TMZ-resistant U87 cell line. RESULTS The growth-inhibitory effect of the combination treatment was significant against the TMZ-R U87 cell line (IC50: 78 μM for STX-0119, 30.5 μM for rapamycin and 11.3 μM for combination of the two). Western blotting analysis demonstrated that the inhibitory effect of STX-0119 on S6 and 4E-BP1 activation through regulation of YKL-40 expression occurred in addition to the inhibitory effect of rapamycin against the mTOR pathway. CONCLUSION These results suggest that the STAT3 pathway is associated with the mTOR downstream pathway mediated by YKL-40 protein, and the combination therapy of the STAT3 inhibitor and rapamycin could be worth developing as a novel therapeutic approach against TMZ-resistant relapsed gliomas.
Collapse
Affiliation(s)
- Haruo Miyata
- Immunotherapy Division, Shizuoka Cancer Center Research Institute, Shizuoka Cancer Center Hospital, Shizuoka, Japan
| | - Tadashi Ashizawa
- Immunotherapy Division, Shizuoka Cancer Center Research Institute, Shizuoka Cancer Center Hospital, Shizuoka, Japan
| | - Akira Iizuka
- Immunotherapy Division, Shizuoka Cancer Center Research Institute, Shizuoka Cancer Center Hospital, Shizuoka, Japan
| | - Ryota Kondou
- Immunotherapy Division, Shizuoka Cancer Center Research Institute, Shizuoka Cancer Center Hospital, Shizuoka, Japan
| | - Chizu Nonomura
- Immunotherapy Division, Shizuoka Cancer Center Research Institute, Shizuoka Cancer Center Hospital, Shizuoka, Japan
| | - Takashi Sugino
- Division of Pathology, Shizuoka Cancer Center Hospital, Shizuoka, Japan
| | - Kenichi Urakami
- Cancer Diagnostics Division, Shizuoka Cancer Center Research Institute, Shizuoka Cancer Center Hospital, Shizuoka, Japan
| | - Akira Asai
- Graduate School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan
| | - Nakamasa Hayashi
- Division of Neurosurgery, Shizuoka Cancer Center Hospital, Shizuoka, Japan
| | - Koichi Mitsuya
- Division of Neurosurgery, Shizuoka Cancer Center Hospital, Shizuoka, Japan
| | - Yoko Nakasu
- Division of Neurosurgery, Shizuoka Cancer Center Hospital, Shizuoka, Japan
| | - Ken Yamaguchi
- Office of the President, Shizuoka Cancer Center Hospital, Shizuoka, Japan
| | - Yasuto Akiyama
- Immunotherapy Division, Shizuoka Cancer Center Research Institute, Shizuoka Cancer Center Hospital, Shizuoka, Japan .,Division of Neurosurgery, Shizuoka Cancer Center Hospital, Shizuoka, Japan
| |
Collapse
|
67
|
Chamcheu JC, Adhami VM, Esnault S, Sechi M, Siddiqui IA, Satyshur KA, Syed DN, Dodwad SJM, Chaves-Rodriquez MI, Longley BJ, Wood GS, Mukhtar H. Dual Inhibition of PI3K/Akt and mTOR by the Dietary Antioxidant, Delphinidin, Ameliorates Psoriatic Features In Vitro and in an Imiquimod-Induced Psoriasis-Like Disease in Mice. Antioxid Redox Signal 2017; 26:49-69. [PMID: 27393705 PMCID: PMC5206770 DOI: 10.1089/ars.2016.6769] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
AIM The treatment of psoriasis remains elusive, underscoring the need for identifying novel disease targets and mechanism-based therapeutic approaches. We recently reported that the PI3K/Akt/mTOR pathway that is frequently deregulated in many malignancies is also clinically relevant for psoriasis. We also provided rationale for developing delphinidin (Del), a dietary antioxidant for the management of psoriasis. This study utilized high-throughput biophysical and biochemical approaches and in vitro and in vivo models to identify molecular targets regulated by Del in psoriasis. RESULTS A kinome-level screen and Kds analyses against a panel of 102 human kinase targets showed that Del binds to three lipid (PIK3CG, PIK3C2B, and PIK3CA) and six serine/threonine (PIM1, PIM3, mTOR, S6K1, PLK2, and AURKB) kinases, five of which belong to the PI3K/Akt/mTOR pathway. Surface plasmon resonance and in silico molecular modeling corroborated Del's direct interactions with three PI3Ks (α/c2β/γ), mTOR, and p70S6K. Del treatment of interleukin-22 or TPA-stimulated normal human epidermal keratinocytes (NHEKs) significantly inhibited proliferation, activation of PI3K/Akt/mTOR components, and secretion of proinflammatory cytokines and chemokines. To establish the in vivo relevance of these findings, an imiquimod (IMQ)-induced Balb/c mouse psoriasis-like skin model was employed. Topical treatment of Del significantly decreased (i) hyperproliferation and epidermal thickness, (ii) skin infiltration by immune cells, (iii) psoriasis-related cytokines/chemokines, (iv) PI3K/Akt/mTOR pathway activation, and (v) increased differentiation when compared with controls. Innovation and Conclusion: Our observation that Del inhibits key kinases involved in psoriasis pathogenesis and alleviates IMQ-induced murine psoriasis-like disease suggests a novel PI3K/AKT/mTOR pathway modulator that could be developed to treat psoriasis. Antioxid. Redox Signal. 26, 49-69.
Collapse
Affiliation(s)
- Jean Christopher Chamcheu
- 1 Department of Dermatology, School of Medicine and Public Health, University of Wisconsin , Madison, Wisconsin
| | - Vaqar M Adhami
- 1 Department of Dermatology, School of Medicine and Public Health, University of Wisconsin , Madison, Wisconsin
| | - Stephane Esnault
- 2 Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, School of Medicine and Public Health, University of Wisconsin , Madison, Wisconsin
| | - Mario Sechi
- 3 Department of Chemistry and Pharmacy, University of Sassari , Sassari, Italy
| | - Imtiaz A Siddiqui
- 1 Department of Dermatology, School of Medicine and Public Health, University of Wisconsin , Madison, Wisconsin
| | - Kenneth A Satyshur
- 4 Small Molecule Screening Facility, Carbone Cancer Center, School of Medicine and Public Health, University of Wisconsin , Madison, Wisconsin.,5 Middleton VA Medical Center , Madison, Wisconsin
| | - Deeba N Syed
- 1 Department of Dermatology, School of Medicine and Public Health, University of Wisconsin , Madison, Wisconsin
| | - Shah-Jahan M Dodwad
- 1 Department of Dermatology, School of Medicine and Public Health, University of Wisconsin , Madison, Wisconsin
| | - Maria-Ines Chaves-Rodriquez
- 1 Department of Dermatology, School of Medicine and Public Health, University of Wisconsin , Madison, Wisconsin.,6 Centro de Investigación en Biotecnología Instituto Tecnológico de Costa Rica , Cartago, Republica de Costa Rica
| | - B Jack Longley
- 1 Department of Dermatology, School of Medicine and Public Health, University of Wisconsin , Madison, Wisconsin
| | - Gary S Wood
- 1 Department of Dermatology, School of Medicine and Public Health, University of Wisconsin , Madison, Wisconsin
| | - Hasan Mukhtar
- 1 Department of Dermatology, School of Medicine and Public Health, University of Wisconsin , Madison, Wisconsin
| |
Collapse
|
68
|
Feng Y, Wu L. mTOR up-regulation of PFKFB3 is essential for acute myeloid leukemia cell survival. Biochem Biophys Res Commun 2017; 483:897-903. [PMID: 28082200 DOI: 10.1016/j.bbrc.2017.01.031] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2017] [Accepted: 01/08/2017] [Indexed: 11/25/2022]
Abstract
Although mTOR (mammalian target of rapamycin) activation is frequently observed in acute myeloid leukemia (AML) patients, the precise function and the downstream targets of mTOR are poorly understood. Here we revealed that PFKFB3, but not PFKFB1, PFKFB2 nor PFKFB4 was a novel downstream substrate of mTOR signaling pathway as PFKFB3 level was augmented after knocking down TSC2 in THP1 and OCI-AML3 cells. Importantly, PFKFB3 silencing suppressed glycolysis and cell proliferation of TSC2 silencing OCI-AML3 cells and activated apoptosis pathway. These results suggested that mTOR up-regulation of PFKFB3 was essential for AML cells survival. Mechanistically, Rapamycin treatment or Raptor knockdown reduced the expression of PFKFB3 in TSC2 knockdown cells, while Rictor silencing did not have such effect. Furthermore, we also revealed that mTORC1 up-regulated PFKFB3 was dependent on hypoxia-inducible factor 1α (HIF1α), a positive regulator of glycolysis. Moreover, PFKFB3 inhibitor PFK15 and rapamycin synergistically blunted the AML cell proliferation. Taken together, PFKFB3 was a promising drug target in AML patients harboring mTOR hyper-activation.
Collapse
Affiliation(s)
- Yonghuai Feng
- Department of Hematology, Peking University People's Hospital, Beijing, China; Institute of Hematology, Peking University, Beijing, China; Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China; Collaborative Innovation Center of Hematology, Peking University, Beijing, China.
| | - Liusong Wu
- The Second Department of Pediatrics, Affiliated Hospital of Zunyi Medical College, Zunyi, Guizhou, China
| |
Collapse
|
69
|
Rahman MA, Bishayee K, Sadra A, Huh SO. Oxyresveratrol activates parallel apoptotic and autophagic cell death pathways in neuroblastoma cells. Biochim Biophys Acta Gen Subj 2016; 1861:23-36. [PMID: 27815218 DOI: 10.1016/j.bbagen.2016.10.025] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Revised: 09/05/2016] [Accepted: 10/29/2016] [Indexed: 01/31/2023]
Abstract
BACKGROUND Drug resistance from apoptosis is a challenging issue with different cancer types, and there is an interest in identifying other means of inducing cytotoxicity. Here, treatment of neuroblastoma cells with oxyresveratrol (OXYRES), a natural antioxidant, led to dose-dependent cell death and increased autophagic flux along with activation of caspase-dependent apoptosis. METHODS For cell viability, we performed the CCK-8 assay. Protein expression changes were with Western blot and immunocytochemistry. Silencing of proteins was with siRNA. The readouts for cell cycle, mitochondria membrane potential, caspase-3, autophagy and apoptosis were performed with flow cytometry. RESULTS Phosphorylation of p38 MAPK increased with OXYRES treatment and inhibition of p38 reduced autophagy and cell death from OXYRES. In contrast, PI3K/AKT/mTOR signaling decreased in the target cells with OXYRES and inhibition of PI3K or mTOR enhanced OXYRES-mediated cytotoxicity with increased levels of autophagy. Modulation of either of the apoptosis and autophagy flux pathways affected the extent of cell death by OXYRES, but did not affect the indicators of these pathways with respect to each other. Both pathways were independent of ROS generation or p53 activation. CONCLUSION OXYRES led to cell death from autophagy, which was independent of apoptosis induction. The OXYRES effects were due to changes in the activity levels of p38 MAPK and PI3K/AKT/mTOR. GENERAL SIGNIFICANCE With two independent and parallel pathways for cytotoxicity induction in target cells, this study puts forward a potential utility for OXYRES or the pathways it represents as novel means of inducing cell death in neuroblastoma cells.
Collapse
Affiliation(s)
- Md Ataur Rahman
- Department of Pharmacology, College of Medicine, Institute of Natural Medicine, Hallym University, South Korea
| | - Kausik Bishayee
- Department of Pharmacology, College of Medicine, Institute of Natural Medicine, Hallym University, South Korea
| | - Ali Sadra
- Department of Pharmacology, College of Medicine, Institute of Natural Medicine, Hallym University, South Korea
| | - Sung-Oh Huh
- Department of Pharmacology, College of Medicine, Institute of Natural Medicine, Hallym University, South Korea.
| |
Collapse
|
70
|
Ong PS, Wang LZ, Dai X, Tseng SH, Loo SJ, Sethi G. Judicious Toggling of mTOR Activity to Combat Insulin Resistance and Cancer: Current Evidence and Perspectives. Front Pharmacol 2016; 7:395. [PMID: 27826244 PMCID: PMC5079084 DOI: 10.3389/fphar.2016.00395] [Citation(s) in RCA: 137] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Accepted: 10/07/2016] [Indexed: 12/16/2022] Open
Abstract
The mechanistic target of rapamycin (mTOR), via its two distinct multiprotein complexes, mTORC1, and mTORC2, plays a central role in the regulation of cellular growth, metabolism, and migration. A dysregulation of the mTOR pathway has in turn been implicated in several pathological conditions including insulin resistance and cancer. Overactivation of mTORC1 and disruption of mTORC2 function have been reported to induce insulin resistance. On the other hand, aberrant mTORC1 and mTORC2 signaling via either genetic alterations or increased expression of proteins regulating mTOR and its downstream targets have contributed to cancer development. These underlined the attractiveness of mTOR as a therapeutic target to overcome both insulin resistance and cancer. This review summarizes the evidence supporting the notion of intermittent, low dose rapamycin for treating insulin resistance. It further highlights recent data on the continuous use of high dose rapamycin analogs and related second generation mTOR inhibitors for cancer eradication, for overcoming chemoresistance and for tumor stem cell suppression. Within these contexts, the potential challenges associated with the use of mTOR inhibitors are also discussed.
Collapse
Affiliation(s)
- Pei Shi Ong
- Department of Pharmacy, Faculty of Science, National University of Singapore Singapore, Singapore
| | - Louis Z Wang
- Department of Pharmacy, Faculty of Science, National University of SingaporeSingapore, Singapore; Department of Pharmacology, Yong Loo Lin School of Medicine, National University of SingaporeSingapore, Singapore
| | - Xiaoyun Dai
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore Singapore, Singapore
| | - Sheng Hsuan Tseng
- Department of Pharmacy, Faculty of Science, National University of Singapore Singapore, Singapore
| | - Shang Jun Loo
- Department of Pharmacy, Faculty of Science, National University of Singapore Singapore, Singapore
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore Singapore, Singapore
| |
Collapse
|
71
|
Nagel R, Semenova EA, Berns A. Drugging the addict: non-oncogene addiction as a target for cancer therapy. EMBO Rep 2016; 17:1516-1531. [PMID: 27702988 PMCID: PMC5090709 DOI: 10.15252/embr.201643030] [Citation(s) in RCA: 108] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2016] [Accepted: 08/24/2016] [Indexed: 12/13/2022] Open
Abstract
Historically, cancers have been treated with chemotherapeutics aimed to have profound effects on tumor cells with only limited effects on normal tissue. This approach was followed by the development of small‐molecule inhibitors that can target oncogenic pathways critical for the survival of tumor cells. The clinical targeting of these so‐called oncogene addictions, however, is in many instances hampered by the outgrowth of resistant clones. More recently, the proper functioning of non‐mutated genes has been shown to enhance the survival of many cancers, a phenomenon called non‐oncogene addiction. In the current review, we will focus on the distinct non‐oncogenic addictions found in cancer cells, including synthetic lethal interactions, the underlying stress phenotypes, and arising therapeutic opportunities.
Collapse
Affiliation(s)
- Remco Nagel
- Division of Molecular Genetics, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Ekaterina A Semenova
- Division of Molecular Genetics, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Anton Berns
- Division of Molecular Genetics, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| |
Collapse
|
72
|
Kunati SR, Xu Y. Determination of MLN0128, an investigational antineoplastic agent, in human plasma by LC-MS/MS. Biomed Chromatogr 2016; 31. [PMID: 27554984 DOI: 10.1002/bmc.3818] [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: 12/24/2015] [Revised: 08/07/2016] [Accepted: 08/20/2016] [Indexed: 11/06/2022]
Abstract
MLN0128, an mTOR kinase inhibitor, is currently undergoing clinical investigation for treatment of a variety of cancers. To support this work, an LC-MS/MS method has been developed for the determination of MLN0128 in human plasma. A structural analog STK040263 was used as the internal standard. Both MLN0128 and the IS were first extracted from plasma using methyl tert-butyl ether; then separated on a Waters XTerra® MS C18 column using a mobile phase consisting of methanol-acetonitrile-10.0 mm ammonium formate (34:6:60, v/v/v) at a flow rate of 0.300 mL min-1 . Quantitation of MLN0128 was done by positive electrospray ionization tandem mass spectrometry in multiple-reaction-monitoring mode. This method has a total run time of <4 min with the retention times of 1.95 and 2.94 min for the IS and MLN0128, respectively. The method has been validated per the US Food and Drug Administration guidance for bioanalytical method validation. It has a calibration range of 0.100-50.0 ng mL-1 in human plasma with a correlation coefficient > 0.999. The overall assay accuracy and precision were ≤ ± 4 and ≤8%, respectively. The IS normalized recovery of MLN0128 was 98-100%. The stability studies showed that MLN0128 was stable under all tested conditions. The method developed may be useful for clinical studies of MLN0128.
Collapse
Affiliation(s)
- Sandeep R Kunati
- Department of Chemistry, Cleveland State University, Cleveland, OH, USA
| | - Yan Xu
- Department of Chemistry, Cleveland State University, Cleveland, OH, USA.,Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH, USA
| |
Collapse
|
73
|
Asimomytis A, Karanikou M, Rodolakis A, Vaiopoulou A, Tsetsa P, Creatsas G, Stefos T, Antsaklis A, Patsouris E, Rassidakis GZ. mTOR downstream effectors, 4EBP1 and eIF4E, are overexpressed and associated with HPV status in precancerous lesions and carcinomas of the uterine cervix. Oncol Lett 2016; 12:3234-3240. [PMID: 27899988 PMCID: PMC5103924 DOI: 10.3892/ol.2016.5056] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2015] [Accepted: 06/07/2016] [Indexed: 11/17/2022] Open
Abstract
The present study aims to investigate the expression levels of two critical mammalian target of rapamycin (mTOR) downstream effectors, 4E binding protein 1 (4EBP1) and eukaryotic initiation factor 4E (eIF4E) proteins, in precancerous squamous intraepithelial lesions and cancer of the uterine cervix, and their association with human papilloma virus (HPV) infection status. Uterine cervical biopsies from 73 patients were obtained, including 40 fresh-frozen samples and 42 archival formalin-fixed, paraffin-embedded tissue specimens. Whole protein extracts were analyzed for the expression of 4EBP1 and eIF4E proteins using western blotting. In addition, distribution of 4EBP1 and eIF4E protein expression and 4EBP1 phosphorylation (P-4EBP1) were analyzed by immunohistochemistry in archival tissues and correlated with the degree of dysplasia. The presence of high-risk HPV (HR-HPV) types was assessed by polymerase chain reaction. Using western blot analysis, high expression levels of 4EBP1 and eIF4E were observed in all uterine cervical carcinomas, which significantly correlated with the degree of dysplasia. By immunohistochemistry, overexpression of 4EBP1 and eIF4E was detected in 20 of 21 (95%) and 17 of 21 (81%) samples, respectively, in patients with high-grade dysplasia and carcinomas, compared with 1 of 20 (5%) and 2 of 20 (10%) samples, respectively, in patients with low-grade lesions or normal histology. All 4EBP1-positive cases tested were also positive for P-4EBP1. Furthermore, overexpression of 4EBP1 and eIF4E significantly correlated with the presence of HR-HPV oncogenic types. The present study demonstrated that critical effectors of mTOR signaling, which control protein synthesis initiation, are overexpressed in cervical high-grade dysplasia and cancer, and their levels correlate with oncogenic HPV types. These findings may provide novel targets for investigational therapeutic approaches in patients with cancer of the uterine cervix.
Collapse
Affiliation(s)
- Aristidis Asimomytis
- First Department of Pathology, National and Kapodistrian University of Athens, School of Medicine, GR-11527 Athens, Greece
| | - Maria Karanikou
- First Department of Pathology, National and Kapodistrian University of Athens, School of Medicine, GR-11527 Athens, Greece
| | - Alexander Rodolakis
- First Department of Obstetrics and Gynecology, National and Kapodistrian University of Athens, Alexandra Maternity Hospital, GR-11528 Athens, Greece
| | - Anna Vaiopoulou
- First Department of Pathology, National and Kapodistrian University of Athens, School of Medicine, GR-11527 Athens, Greece
| | - Paraskevi Tsetsa
- First Department of Obstetrics and Gynecology, National and Kapodistrian University of Athens, Alexandra Maternity Hospital, GR-11528 Athens, Greece
| | - George Creatsas
- Second Department of Obstetrics and Gynecology, National and Kapodistrian University of Athens, Aretaieion Hospital, GR-11528 Athens, Greece
| | - Theodoros Stefos
- Department of Obstetrics and Gynecology, University of Ioannina, School of Medicine, GR-45110 Ioannina, Greece
| | - Aristidis Antsaklis
- First Department of Obstetrics and Gynecology, National and Kapodistrian University of Athens, Alexandra Maternity Hospital, GR-11528 Athens, Greece
| | - Efstratios Patsouris
- First Department of Pathology, National and Kapodistrian University of Athens, School of Medicine, GR-11527 Athens, Greece
| | - George Z Rassidakis
- First Department of Pathology, National and Kapodistrian University of Athens, School of Medicine, GR-11527 Athens, Greece; Department of Pathology and Cytology, Karolinska University Hospital and Karolinska Institute, Solna 17176, Sweden
| |
Collapse
|
74
|
Wang B, Li H, Yao Q, Zhang Y, Zhu X, Xia T, Wang J, Li G, Li X, Ni S. Local in vitro delivery of rapamycin from electrospun PEO/PDLLA nanofibers for glioblastoma treatment. Biomed Pharmacother 2016; 83:1345-1352. [PMID: 27580454 DOI: 10.1016/j.biopha.2016.08.033] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Revised: 08/09/2016] [Accepted: 08/11/2016] [Indexed: 01/28/2023] Open
Abstract
Rapamycin, a mammalian target of rapamycin inhibitor and anti-proliferative agent, is used to treat glioma and other malignancies, but its effectiveness is limited by the fact that it cannot be delivered in a targeted manner to the site of the tumor. To address this issue, we fabricated a mesh via electrospinning using two biodegradable materials, poly(lactic acid) (PLA) and polyethylene oxide (PEO) as a carrier for rapamycin delivery to the tumor. Nanofiber diameter decreased with increasing PLA concentration in the mixed solution. Scanning electron microscopy analysis revealed the smooth and uniform surface morphology of hybrid fibers. Fourier transform infrared spectroscopy analysis demonstrated that rapamycin was encapsulated in the polymer solution; encapsulation efficiency was high and stable over the range of drug concentrations from 0.5-2wt%. A correlation was observed between sustained release of the drug in vitro and cytotoxicity in cultured glioma cells. These results indicate that the PEO/poly(d,l-lactic acid) nanofiber mesh can be used as a targeted delivery system for rapamycin that can limit side effects and prevent locoregional recurrence following surgical resection of glioma.
Collapse
Affiliation(s)
- Benlin Wang
- Department of Neurosurgery, Qilu Hospital, Shandong University, Jinan 250012, China
| | - Haoyuan Li
- Department of Neurosurgery, Qilu Hospital, Shandong University, Jinan 250012, China
| | - Qingyu Yao
- Department of Neurosurgery, Qilu Hospital, Shandong University, Jinan 250012, China
| | - Yulin Zhang
- Department of Neurosurgery, Qilu Hospital, Shandong University, Jinan 250012, China
| | - Xiaodong Zhu
- Affiliated Hospital of Jining Medical University, Jining 272000, China
| | - Tongliang Xia
- Department of Neurosurgery, Qilu Hospital, Shandong University, Jinan 250012, China
| | - Jian Wang
- Brain Science Research Institute, Shandong University, Jinan 250012, China; Department of Biomedicine, University of Bergen, Jonas Lies Vei 91, 5009 Bergen, Norway
| | - Gang Li
- Department of Neurosurgery, Qilu Hospital, Shandong University, Jinan 250012, China; Brain Science Research Institute, Shandong University, Jinan 250012, China
| | - Xingang Li
- Department of Neurosurgery, Qilu Hospital, Shandong University, Jinan 250012, China; Brain Science Research Institute, Shandong University, Jinan 250012, China
| | - Shilei Ni
- Department of Neurosurgery, Qilu Hospital, Shandong University, Jinan 250012, China.
| |
Collapse
|
75
|
Mohan N, Shen Y, Dokmanovic M, Endo Y, Hirsch DS, Wu WJ. VPS34 regulates TSC1/TSC2 heterodimer to mediate RheB and mTORC1/S6K1 activation and cellular transformation. Oncotarget 2016; 7:52239-52254. [PMID: 27409169 PMCID: PMC5239548 DOI: 10.18632/oncotarget.10469] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Accepted: 06/07/2016] [Indexed: 12/12/2022] Open
Abstract
VPS34 is reported to activate S6K1 and is implicated in regulating cell growth, the mechanisms of which remain elusive. Here, we describe novel mechanisms by which VPS34 upregulates mTOR/S6K1 activity via downregulating TSC2 protein and activating RheB activity. Specifically, upregulation of VPS34 lipid kinase increases local production of ptdins(3)p in the plasma membrane, which recruits PIKFYVE, a FYVE domain containing protein, to ptdins(3)p enriched regions of the plasma membrane, where VPS34 forms a protein complex with PIKFYVE and TSC1. This in turn disengages TSC2 from the TSC1/TSC2 heterodimer, leading to TSC2 ubiquitination and degradation. Downregulation of TSC2 promotes the activation of RheB and mTOR/S6K1. When VPS34 lipid kinase activity is increased by introduction of an H868R mutation, ptdins(3)p production at the plasma membrane is dramatically increased, which recruits more PIKFYVE and TSC1 molecules to the plasma membrane. This results in the enhanced TSC2 ubiquitination and degradation, and subsequent activation of RheB and mTORC1/S6K1, leading to oncogenic transformation. The role played by VPS34 in regulating mTOR/S6K1 activity and cellular transformation is underscored by the fact that the VPS34 kinase dead mutant blocks VPS34-induced recruitment of PIKFYVE and TSC1 to the plasma membrane. This study provides mechanistic insight into the cellular function of VPS34 in regulating oncogenic transformation and important indications for identifying VPS34 specific mutations in human cancers.
Collapse
Affiliation(s)
- Nishant Mohan
- Division of Biotechnology Review and Research I, Office of Biotechnology Products, Office of Pharmaceutical Quality, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, 20993, Maryland, USA
| | - Yi Shen
- Division of Biotechnology Review and Research I, Office of Biotechnology Products, Office of Pharmaceutical Quality, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, 20993, Maryland, USA
| | - Milos Dokmanovic
- Division of Biotechnology Review and Research I, Office of Biotechnology Products, Office of Pharmaceutical Quality, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, 20993, Maryland, USA
| | - Yukinori Endo
- Division of Biotechnology Review and Research I, Office of Biotechnology Products, Office of Pharmaceutical Quality, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, 20993, Maryland, USA
| | - Dianne S. Hirsch
- Division of Biotechnology Review and Research I, Office of Biotechnology Products, Office of Pharmaceutical Quality, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, 20993, Maryland, USA
| | - Wen Jin Wu
- Division of Biotechnology Review and Research I, Office of Biotechnology Products, Office of Pharmaceutical Quality, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, 20993, Maryland, USA
| |
Collapse
|
76
|
Imrali A, Mao X, Yeste-Velasco M, Shamash J, Lu Y. Rapamycin inhibits prostate cancer cell growth through cyclin D1 and enhances the cytotoxic efficacy of cisplatin. Am J Cancer Res 2016; 6:1772-1784. [PMID: 27648364 PMCID: PMC5004078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Accepted: 02/24/2016] [Indexed: 06/06/2023] Open
Abstract
Prostate cancer is the most common malignancy in Western men and hormone refractory cancer (HRPC) kills most of the patients. Chemo-resistance is a major obstacle for the treatment of prostate cancer. Platinum-complexes have been used to treat a number of malignancies including prostate cancer. However, it has limited effect to prostate cancer and with significant toxicity at higher doses. In recent years, increasing numbers of new agents targeting cancer specific pathways have become available and with low toxic side-effects. Rapamycin (Sirolimus) is an mTORC1 inhibitor, which inhibits the PI3K/Akt/mTOR signaling pathway, which is commonly altered in prostate cancer. We determined the expression of cyclin D1 and phosphorylated-mTOR proteins in association with the response to rapamycin in two androgen sensitive (22RV1 and LNCaP) and two androgen independent (DU145 and PC3) prostate cancer cell lines and found that the base-line and changes of cyclin D1 level, but not the expression level of p-mTOR, correlated with rapamycin sensitivity. We evaluated the cell killing effect of combined rapamycin and cisplatin treatment and showed that the combination had a more than additive effect in both androgen dependent and independent prostate cancer cells, which may be partially explained by the reduction of cyclin D1 expression by rapamycin. We also evaluated a range of combined treatment schedules, simultaneously or sequentially and found that continuous rapamycin treatment after a short cisplatin exposure was effective. The clinical application of these findings for prostate cancer treatment should be further investigated.
Collapse
Affiliation(s)
- Ahmet Imrali
- Centre for Molecular Oncology, Barts Cancer Institute, Queen Mary University of LondonLondon, UK
| | - Xueying Mao
- Centre for Molecular Oncology, Barts Cancer Institute, Queen Mary University of LondonLondon, UK
| | - Marc Yeste-Velasco
- Centre for Molecular Oncology, Barts Cancer Institute, Queen Mary University of LondonLondon, UK
| | - Jonathan Shamash
- Center for Experimental Cancer Medicine, Barts Cancer Institute, Queen Mary University of LondonLondon, UK
| | - Yongjie Lu
- Centre for Molecular Oncology, Barts Cancer Institute, Queen Mary University of LondonLondon, UK
| |
Collapse
|
77
|
Bhattacharya K, Maiti S, Mandal C. PTEN negatively regulates mTORC2 formation and signaling in grade IV glioma via Rictor hyperphosphorylation at Thr1135 and direct the mode of action of an mTORC1/2 inhibitor. Oncogenesis 2016; 5:e227. [PMID: 27239959 PMCID: PMC4945751 DOI: 10.1038/oncsis.2016.34] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Revised: 03/10/2016] [Accepted: 03/14/2016] [Indexed: 02/07/2023] Open
Abstract
To investigate the role of PTEN (phosphatase and tensin homolog) in mammalian target of rapamycin complex 2 (mTORC2) signaling in glioblastoma multiforme (GBM), we found higher activation of mTORC2 in PTEN(mu) cells, as evidenced by enhanced phosphorylation of mTOR (Ser2481), AKT (Ser473) and glycogen synthase kinase 3 beta (GSK3β) (Ser9) as compared with PTEN(wt) cells. In addition, PTEN(wt) cells upon PTEN depletion showed mTORC2 activation. The reduced mTORC2 signaling in PTEN(wt) cells was related to higher Rictor phosphorylation at Thr1135 residue. Phosphorylation of Rictor at Thr1135 inhibited its association with mTORC and thus there was a reduction in mTORC2 complex formation. In addition, PTEN(wt) cells expressing mutated Rictor in which Thr1135 was substituted with alanine, showed enhanced mTORC2 formation and signaling. This enhanced mTORC2 signaling promoted inactivation of GSK3β. Thus, we established the reciprocal activation of mTORC2 and GSK3β in GBM. To the best of our knowledge, this is the first report describing role of PTEN in mTORC2 formation by promoting Rictor phosphorylation (Thr1135) in GBM. Furthermore, the drug sensitivity of mTORC2 was evaluated. A newly identified carbazole alkaloid, mahanine, showed cytotoxicity in both PTEN(mu) and PTEN(wt) cells. It inhibited both mTORC1/2 and AKT completely in PTEN(mu) cells, whereas it inhibited only mTORC1 in PTEN(wt) cells. Cytotoxity and AKT-inhibitory activity of the mTORC1/2 inhibitor was increased either by depleting PTEN or in combination with phosphatidylinositol 3 kinase inhibitors in PTEN(wt) cells. In contrast, depletion of Rictor decreased the cytotoxicity of the mTORC1/2 inhibitor in PTEN(mu) cells. Thus, PTEN has an important role in mTORC2 formation and also influences the effectiveness of an mTORC1/2 inhibitor in GBM.
Collapse
Affiliation(s)
- K Bhattacharya
- Cancer Biology and Inflammatory Disorder Division, Council of Scientific and Industrial Research (CSIR)-Indian Institute of Chemical Biology, Kolkata, India
| | - S Maiti
- Cancer Biology and Inflammatory Disorder Division, Council of Scientific and Industrial Research (CSIR)-Indian Institute of Chemical Biology, Kolkata, India
| | - C Mandal
- Cancer Biology and Inflammatory Disorder Division, Council of Scientific and Industrial Research (CSIR)-Indian Institute of Chemical Biology, Kolkata, India
| |
Collapse
|
78
|
Prediction of response to everolimus in neuroendocrine tumors: evaluation of clinical, biological and histological factors. Invest New Drugs 2016; 34:654-62. [PMID: 27230034 DOI: 10.1007/s10637-016-0363-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Accepted: 05/18/2016] [Indexed: 12/19/2022]
Abstract
Objectives Several targeted therapies are available for metastatic neuroendocrine tumours (NETs) but no predictive factor of response to these treatments has been identified yet. Our aim was to identify and evaluate clinical, biological, histological and functional markers of response to everolimus. Methods We retrospectively reviewed 53 patients with NETs treated with everolimus (68 % in clinical trials). Clinical, biological and histological data were analyzed. The functional marker p-p70S6K, a main effector of the mTOR pathway, was studied by immunohistochemistry in 43 cases. Prognostic factors of progression-free survival (PFS) were studied by Kaplan Meier analysis. Results All patients had metastatic and progressive disease before everolimus treatment. Objective response was 9 % and median PFS was 8.1 (4.7-11.5) months. Hypercholesterolemia (HR = 0.13, p < 0.0001) was associated with longer PFS, whereas presence of bone metastases (HR = 3.1, p < 0.001) and overexpression of p-p70S6K by tumor cells (HR = 2.5, p = 0.01) were associated with shorter PFS under everolimus at multivariate analysis. Conclusion Clinical markers are not useful to predict response to everolimus. However, occurrence of hypercholesterolemia under treatment may be an early marker of response. Prospective studies are required to confirm these results and to assess whether p-p70S6K immunostaining is a prognostic or predictive marker of no-response to everolimus.
Collapse
|
79
|
Wick W, Gorlia T, Bady P, Platten M, van den Bent MJ, Taphoorn MJ, Steuve J, Brandes AA, Hamou MF, Wick A, Kosch M, Weller M, Stupp R, Roth P, Golfinopoulos V, Frenel JS, Campone M, Ricard D, Marosi C, Villa S, Weyerbrock A, Hopkins K, Homicsko K, Lhermitte B, Pesce G, Hegi ME. Phase II Study of Radiotherapy and Temsirolimus versus Radiochemotherapy with Temozolomide in Patients with Newly Diagnosed Glioblastoma without MGMT Promoter Hypermethylation (EORTC 26082). Clin Cancer Res 2016; 22:4797-4806. [DOI: 10.1158/1078-0432.ccr-15-3153] [Citation(s) in RCA: 112] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2015] [Accepted: 04/03/2016] [Indexed: 11/16/2022]
|
80
|
Ortolani S, Ciccarese C, Cingarlini S, Tortora G, Massari F. Suppression of mTOR pathway in solid tumors: lessons learned from clinical experience in renal cell carcinoma and neuroendocrine tumors and new perspectives. Future Oncol 2016; 11:1809-28. [PMID: 26075448 DOI: 10.2217/fon.15.81] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
The PI3K-AKT-mTOR pathway plays role in the regulation of many cellular processes. Hyperactivation of mTOR signaling has been implicated in human carcinogenesis, representing an attractive target for cancer therapy. Among other cancer subtypes, renal cell carcinoma (RCC) and neuroendocrine tumors are relevant settings in which the deregulation of mTOR pathway is of crucial importance. Different mTOR-inhibitory agents have been developed in recent years. Temsirolimus is approved for advanced RCC; everolimus is registered for the treatment of advanced RCC, pancreatic neuroendocrine tumors and postmenopausal, hormone receptor-positive/HER2-negative, advanced breast cancer. This review is focused on the description of the clinical experience with mTOR-inhibitor agents for the treatment of advanced RCC and neuroendocrine tumors, followed by an excursus on the landscape of the ongoing research in this field.
Collapse
Affiliation(s)
- Silvia Ortolani
- Medical Oncology, Azienda Ospedaliera Universitaria Integrata, University of Verona, Piazzale LA Scuro 10, 37124 Verona, Italy
| | - Chiara Ciccarese
- Medical Oncology, Azienda Ospedaliera Universitaria Integrata, University of Verona, Piazzale LA Scuro 10, 37124 Verona, Italy
| | - Sara Cingarlini
- Medical Oncology, Azienda Ospedaliera Universitaria Integrata, University of Verona, Piazzale LA Scuro 10, 37124 Verona, Italy
| | - Giampaolo Tortora
- Medical Oncology, Azienda Ospedaliera Universitaria Integrata, University of Verona, Piazzale LA Scuro 10, 37124 Verona, Italy
| | - Francesco Massari
- Medical Oncology, Azienda Ospedaliera Universitaria Integrata, University of Verona, Piazzale LA Scuro 10, 37124 Verona, Italy
| |
Collapse
|
81
|
Zatelli MC, Fanciulli G, Malandrino P, Ramundo V, Faggiano A, Colao A. Predictive factors of response to mTOR inhibitors in neuroendocrine tumours. Endocr Relat Cancer 2016; 23:R173-83. [PMID: 26666705 DOI: 10.1530/erc-15-0413] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Accepted: 12/14/2015] [Indexed: 12/13/2022]
Abstract
Medical treatment of neuroendocrine tumours (NETs) has drawn a lot of attention due to the recent demonstration of efficacy of several drugs on progression-free survival, including somatostatin analogs, small tyrosine kinase inhibitors and mTOR inhibitors (or rapalogs). The latter are approved as therapeutic agents in advanced pancreatic NETs and have been demonstrated to be effective in different types of NETs, with variable efficacy due to the development of resistance to treatment. Early detection of patients that may benefit from rapalogs treatment is of paramount importance in order to select the better treatment and avoid ineffective and expensive treatments. Predictive markers for therapeutic response are under intensive investigation, aiming at a tailored patient management and more appropriate resource utilization. This review summarizes the available data on the tissue, circulating and imaging markers that are potentially predictive of rapalog efficacy in NETs.
Collapse
Affiliation(s)
- Maria Chiara Zatelli
- Section of Endocrinology and Internal MedicineDepartment of Medical Sciences, University of Ferrara, Via Aldo Moro 8, 44124 Cona - Ferrara, ItalyNeuroendocrine Tumours UnitDepartment of Clinical and Experimental Medicine, University of Sassari - AOU Sassari, Sassari, ItalyEndocrinology UnitGaribaldi Nesima Medical Center, Catania, ItalyDepartment of Clinical Medicine and Surgery"Federico II" University of Naples, Naples, ItalyThyroid and Parathyroid Surgery UnitIstituto Nazionale per lo studio e la cura dei tumori "Fondazione G. Pascale" - IRCCS, Naples, Italy
| | - Giuseppe Fanciulli
- Section of Endocrinology and Internal MedicineDepartment of Medical Sciences, University of Ferrara, Via Aldo Moro 8, 44124 Cona - Ferrara, ItalyNeuroendocrine Tumours UnitDepartment of Clinical and Experimental Medicine, University of Sassari - AOU Sassari, Sassari, ItalyEndocrinology UnitGaribaldi Nesima Medical Center, Catania, ItalyDepartment of Clinical Medicine and Surgery"Federico II" University of Naples, Naples, ItalyThyroid and Parathyroid Surgery UnitIstituto Nazionale per lo studio e la cura dei tumori "Fondazione G. Pascale" - IRCCS, Naples, Italy
| | - Pasqualino Malandrino
- Section of Endocrinology and Internal MedicineDepartment of Medical Sciences, University of Ferrara, Via Aldo Moro 8, 44124 Cona - Ferrara, ItalyNeuroendocrine Tumours UnitDepartment of Clinical and Experimental Medicine, University of Sassari - AOU Sassari, Sassari, ItalyEndocrinology UnitGaribaldi Nesima Medical Center, Catania, ItalyDepartment of Clinical Medicine and Surgery"Federico II" University of Naples, Naples, ItalyThyroid and Parathyroid Surgery UnitIstituto Nazionale per lo studio e la cura dei tumori "Fondazione G. Pascale" - IRCCS, Naples, Italy
| | - Valeria Ramundo
- Section of Endocrinology and Internal MedicineDepartment of Medical Sciences, University of Ferrara, Via Aldo Moro 8, 44124 Cona - Ferrara, ItalyNeuroendocrine Tumours UnitDepartment of Clinical and Experimental Medicine, University of Sassari - AOU Sassari, Sassari, ItalyEndocrinology UnitGaribaldi Nesima Medical Center, Catania, ItalyDepartment of Clinical Medicine and Surgery"Federico II" University of Naples, Naples, ItalyThyroid and Parathyroid Surgery UnitIstituto Nazionale per lo studio e la cura dei tumori "Fondazione G. Pascale" - IRCCS, Naples, Italy
| | - Antongiulio Faggiano
- Section of Endocrinology and Internal MedicineDepartment of Medical Sciences, University of Ferrara, Via Aldo Moro 8, 44124 Cona - Ferrara, ItalyNeuroendocrine Tumours UnitDepartment of Clinical and Experimental Medicine, University of Sassari - AOU Sassari, Sassari, ItalyEndocrinology UnitGaribaldi Nesima Medical Center, Catania, ItalyDepartment of Clinical Medicine and Surgery"Federico II" University of Naples, Naples, ItalyThyroid and Parathyroid Surgery UnitIstituto Nazionale per lo studio e la cura dei tumori "Fondazione G. Pascale" - IRCCS, Naples, Italy
| | - Annamaria Colao
- Section of Endocrinology and Internal MedicineDepartment of Medical Sciences, University of Ferrara, Via Aldo Moro 8, 44124 Cona - Ferrara, ItalyNeuroendocrine Tumours UnitDepartment of Clinical and Experimental Medicine, University of Sassari - AOU Sassari, Sassari, ItalyEndocrinology UnitGaribaldi Nesima Medical Center, Catania, ItalyDepartment of Clinical Medicine and Surgery"Federico II" University of Naples, Naples, ItalyThyroid and Parathyroid Surgery UnitIstituto Nazionale per lo studio e la cura dei tumori "Fondazione G. Pascale" - IRCCS, Naples, Italy
| | | |
Collapse
|
82
|
Mukhopadhyay S, Frias MA, Chatterjee A, Yellen P, Foster DA. The Enigma of Rapamycin Dosage. Mol Cancer Ther 2016; 15:347-53. [PMID: 26916116 DOI: 10.1158/1535-7163.mct-15-0720] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Accepted: 11/03/2015] [Indexed: 12/20/2022]
Abstract
The mTOR pathway is a critical regulator of cell growth, proliferation, metabolism, and survival. Dysregulation of mTOR signaling has been observed in most cancers and, thus, the mTOR pathway has been extensively studied for therapeutic intervention. Rapamycin is a natural product that inhibits mTOR with high specificity. However, its efficacy varies by dose in several contexts. First, different doses of rapamycin are needed to suppress mTOR in different cell lines; second, different doses of rapamycin are needed to suppress the phosphorylation of different mTOR substrates; and third, there is a differential sensitivity of the two mTOR complexes mTORC1 and mTORC2 to rapamycin. Intriguingly, the enigmatic properties of rapamycin dosage can be explained in large part by the competition between rapamycin and phosphatidic acid (PA) for mTOR. Rapamycin and PA have opposite effects on mTOR whereby rapamycin destabilizes and PA stabilizes both mTOR complexes. In this review, we discuss the properties of rapamycin dosage in the context of anticancer therapeutics.
Collapse
Affiliation(s)
- Suman Mukhopadhyay
- Department of Biological Sciences, Hunter College of the City University of New York, New York, New York
| | - Maria A Frias
- Department of Biological Sciences, Hunter College of the City University of New York, New York, New York
| | - Amrita Chatterjee
- Department of Biological Sciences, Hunter College of the City University of New York, New York, New York
| | - Paige Yellen
- Molecular Pharmacology & Chemistry Program, Memorial Sloan-Kettering Cancer Center, New York, New York
| | - David A Foster
- Department of Biological Sciences, Hunter College of the City University of New York, New York, New York. Department of Pharmacology, Weill-Cornell Medical College, New York, New York.
| |
Collapse
|
83
|
Seront E, Rottey S, Filleul B, Glorieux P, Goeminne JC, Verschaeve V, Vandenbulcke JM, Sautois B, Boegner P, Gillain A, van Maanen A, Machiels JP. Phase II study of dual phosphoinositol-3-kinase (PI3K) and mammalian target of rapamycin (mTOR) inhibitor BEZ235 in patients with locally advanced or metastatic transitional cell carcinoma. BJU Int 2016; 118:408-15. [PMID: 26779597 DOI: 10.1111/bju.13415] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
OBJECTIVE To assess, in a multicentre phase II trial, the safety and efficacy of BEZ235, an oral pan-class I phosphoinositol-3-kinase (PI3K) and mammalian target of rapamycin (mTOR) complex1/2 inhibitor, in locally advanced or metastatic transitional cell carcinoma (TCC) after failure of platinum-based therapy. PATIENTS AND METHODS Patients with locally advanced or metastatic TCC progressing after platinum therapy were prospectively stratified by PI3K/Akt/mTOR pathway alterations, defined as PTEN loss and PIK3CA mutation. All patients received BEZ235 until progressive disease or unacceptable toxicity. The primary endpoint was the progression-free survival (PFS) rate at 16 weeks. This study was, however, closed prematurely because BEZ235 was withdrawn from further development. RESULTS A total of 20 patients (18 without and two with PI3K/Akt/mTOR alterations) were enrolled and received BEZ235. One partial response (5%) and two cases of stable disease (10%) were observed, all in patients without PI3K/mTOR pathway alterations. The PFS rate at 8 and 16 weeks was 15 and 10%, respectively; the median (range) PFS was 62 (38-588) days (95% confidence interval [CI] 53-110); and the median (range) overall survival was 127 (41-734) days (95% CI 58-309). Among the 90% of patients who experienced drug-related adverse events of any grade, 50% experienced grade 3-4 adverse events, including stomatitis (15%), fatigue (5%), nausea (5%), diarrhoea (5%), renal failure (5%), cutaneous rash (5%), hepatotoxicity (5%) and hypertension (5%). CONCLUSION BEZ235 showed modest clinical activity and an unfavourable toxicity profile in patients with advanced and pretreated TCC; however, a minority of patients experienced a clinical benefit, suggesting that a complete blockade of the PI3K/mTOR axis could improve outcome in some specific patients. Furthermore, this study showed that molecular stratification of patients for personalized medicine before treatment is feasible.
Collapse
Affiliation(s)
- Emmanuel Seront
- Department of Medical Oncology, Institut Roi Albert II, Cliniques Universitaires Saint-Luc, Université catholique de Louvain, Brussels, Belgium.,Department of Medical Oncology, Centre Hospitalier de Jolimont, Haine Saint-Paul, Belgium
| | - Sylvie Rottey
- Department of Medical Oncology, Ghent University Hospital and Heymans Institute of Pharmacology, Ghent University, Ghent, Belgium
| | - Bertrand Filleul
- Department of Medical Oncology, Centre Hospitalier de Jolimont, Haine Saint-Paul, Belgium
| | - Philippe Glorieux
- Department of Medical Oncology, Cliniques du Sud-Luxembourg-Vivalia, Arlon, Belgium
| | - Jean-Charles Goeminne
- Department of Medical Oncology, Centre de Maternité Sainte Elisabeth, Namur, Belgium
| | - Vincent Verschaeve
- Department of Medical Oncology, Grand Hopital de Charleroi, Charleroi, Belgium
| | | | - Brieuc Sautois
- Department of Medical Oncology, Centre Hospitalier Universitaire Sart-Tilman, Liège, Belgium
| | - Petra Boegner
- Department of Medical Oncology, Centre Hospitalier Epicura, Baudour, Belgium
| | - Aline Gillain
- Department of Medical Oncology, Institut Roi Albert II, Cliniques Universitaires Saint-Luc, Université catholique de Louvain, Brussels, Belgium
| | - Aline van Maanen
- Statistical Support Unit, Institut Roi Albert II, Cliniques Universitaires Saint-Luc, Université catholique de Louvain, Brussels, Belgium
| | - Jean-Pascal Machiels
- Department of Medical Oncology, Institut Roi Albert II, Cliniques Universitaires Saint-Luc, Université catholique de Louvain, Brussels, Belgium
| |
Collapse
|
84
|
Abstract
The translation initiation factor eIF4E mediates a rate-limiting process that drives selective translation of many oncongenic proteins such as cyclin D1, survivin and VEGF, thereby contributing to tumour growth, metastasis and therapy resistance. As an essential regulatory hub in cancer signalling network, many oncogenic signalling pathways appear to converge on eIF4E. Therefore, targeting eIF4E-mediated cap-dependent translation is considered a promising anticancer strategy. This paper reviews the strategies that can be used to target eIF4E, highlighting agents that target eIF4E activity at each distinct level.
Collapse
|
85
|
Rubinfeld H, Cohen O, Kammer A, Yang G, Cohen ZR, Hadani M, Shimon I. Combination of mTOR Inhibitors Augments Potency while Activating PI3K Signaling in Pituitary Tumors. Neuroendocrinology 2016; 103:592-604. [PMID: 26562326 DOI: 10.1159/000442205] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Accepted: 11/05/2015] [Indexed: 11/19/2022]
Abstract
BACKGROUND Despite the success in treating some cancers, the efficacy of the mTOR inhibitors rapalogs as anti-cancer therapeutics has been limited. AIMS We undertook to examine the effects of Torin1, a second-generation selective ATP-competitive mTOR inhibitor, in non-functioning pituitary tumor cells. During characterization of the molecular mechanisms that mediate Torin1 actions, there seemed to be a rationale for combining it with rapalogs. METHODS Proliferation assays, flow cytometry and Western blotting were applied to assess the effects of Torin1, RAD001 and their combination on an MtT/E pituitary cell line and human-derived non-functioning pituitary tumor cells. RESULTS Combined long treatments of Torin1 and RAD001 induced a pronounced reduction in cell growth and viability of both MtT/E pituitary cells and human-derived non-functioning pituitary tumor cells, superior to each drug alone. This was remarkable in the 10 nM combination and was reflected in a triggered decrease of cyclin D3 and p21/CIP expression. Interestingly, Akt-Thr308 and SIN1-Thr86 phosphorylations were robustly elevated in the combined treatment, accompanied by a reduction in PTEN expression. Phosphorylation of p70S6K was abolished in all individual and combined treatments. Akt-Ser473 phosphorylation, induced by RAD001, was reduced by the combined treatment to the same extent as when treated by Torin1 alone. CONCLUSIONS Our results suggest that the differential signaling mechanisms induced by these compounds eventually converge to lead to an efficient blockade of the PI3K/Akt/mTOR pathway in pituitary tumor cells and may facilitate a reduction in treatment dosage.
Collapse
|
86
|
Potential Therapeutic Targets in Uterine Sarcomas. Sarcoma 2015; 2015:243298. [PMID: 26576131 PMCID: PMC4632006 DOI: 10.1155/2015/243298] [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: 08/14/2015] [Accepted: 09/30/2015] [Indexed: 12/30/2022] Open
Abstract
Uterine sarcomas are rare tumors accounting for 3,4% of all uterine cancers. Even after radical hysterectomy, most patients relapse or present with distant metastases. The very limited clinical benefit of adjuvant cytotoxic treatments is reflected by high mortality rates, emphasizing the need for new treatment strategies. This review summarizes rising potential targets in four distinct subtypes of uterine sarcomas: leiomyosarcoma, low-grade and high-grade endometrial stromal sarcoma, and undifferentiated uterine sarcoma. Based on clinical reports, promising approaches for uterine leiomyosarcoma patients include inhibition of VEGF and mTOR signaling, preferably in combination with other targeted or cytotoxic compounds. Currently, the only targeted therapy approved in leiomyosarcoma patients is pazopanib, a multitargeted inhibitor blocking VEGFR, PDGFR, FGFR, and c-KIT. Additionally, preclinical evidence suggests effect of the inhibition of histone deacetylases, tyrosine kinase receptors, and the mitotic checkpoint protein aurora kinase A. In low-grade endometrial stromal sarcomas, antihormonal therapies including aromatase inhibitors and progestins have proven activity. Other potential targets are PDGFR, VEGFR, and histone deacetylases. In high-grade ESS that carry the YWHAE/FAM22A/B fusion gene, the generated 14-3-3 oncoprotein is a putative target, next to c-KIT and the Wnt pathway. The observation of heterogeneity within uterine sarcoma subtypes warrants a personalized treatment approach.
Collapse
|
87
|
Vaishampayan U, Shevrin D, Stein M, Heilbrun L, Land S, Stark K, Li J, Dickow B, Heath E, Smith D, Fontana J. Phase II Trial of Carboplatin, Everolimus, and Prednisone in Metastatic Castration-resistant Prostate Cancer Pretreated With Docetaxel Chemotherapy: A Prostate Cancer Clinical Trial Consortium Study. Urology 2015; 86:1206-11. [PMID: 26375845 DOI: 10.1016/j.urology.2015.08.008] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Revised: 08/13/2015] [Accepted: 08/13/2015] [Indexed: 11/19/2022]
Abstract
OBJECTIVE To conduct a phase II trial of the combination of carboplatin, prednisone, and everolimus in metastatic castrate-resistant prostate cancer (mCRPC) as mTOR inhibition can overcome resistance to chemotherapy in prostate cancer. METHODS Patients with progressive mCRPC pretreated with docetaxel-based regimen were eligible. Performance status of 0-1 and adequate bone marrow, renal, and liver function were required. Primary end point was time to progression. Treatment consisted of carboplatin (starting dose equal to area under the curve (AUC of 5) intravenously every 21 days along with oral everolimus 5 mg once daily and prednisone 5 mg twice daily. RESULTS Twenty-six patients were enrolled with median age of 69 years with 8 patients of African American origin. Grade 3 or 4 thrombocytopenia or neutropenia in 4 of 6 initial patients required dose adjustment of carboplatin to AUC of 4 for subsequent patients. There were no pharmacokinetic interactions between carboplatin and everolimus. The median time to progression was 2.5 months (90% confidence interval [CI], 1.8-4.3 months), and median overall survival was 12.5 months (90% CI, 7.7-18.7 months). Of 10 patients, 8 that demonstrated positive nuclear phosphorylated AKT (pAKT) staining on immunohistochemistry progressed within 9 weeks, whereas 2 patients with negative staining continued without progression for prolonged durations of 30 and 48 weeks. TSC1 gene mutations did not correlate with clinical outcome. CONCLUSION The addition of the mTOR inhibitor everolimus to carboplatin demonstrated minimal clinical efficacy in metastatic prostate cancer. pAKT testing warrants further evaluation as a predictive marker of response to everolimus therapy.
Collapse
Affiliation(s)
- Ulka Vaishampayan
- Department of Oncology, Department of Medicine, Barbara Ann Karmanos Cancer Institute, Wayne State University, Detroit, MI.
| | - Daniel Shevrin
- Department of Oncology, Northshore University Health System, Evanston, IL
| | - Mark Stein
- Department of Oncology, Cancer Institute of New Jersey, New Brunswick, NJ
| | - Lance Heilbrun
- Department of Oncology, Biostatistics Core, Barbara Ann Karmanos Cancer Institute, Detroit, MI
| | - Susan Land
- Department of Oncology, John D. Dingell Veterans Medical Center, Detroit, MI
| | - Karri Stark
- Department of Oncology, Department of Medicine, Barbara Ann Karmanos Cancer Institute, Wayne State University, Detroit, MI
| | - Jing Li
- Department of Oncology, Department of Medicine, Barbara Ann Karmanos Cancer Institute, Wayne State University, Detroit, MI
| | - Brenda Dickow
- Department of Oncology, Department of Medicine, Barbara Ann Karmanos Cancer Institute, Wayne State University, Detroit, MI
| | - Elisabeth Heath
- Department of Oncology, Department of Medicine, Barbara Ann Karmanos Cancer Institute, Wayne State University, Detroit, MI
| | - Daryn Smith
- Department of Oncology, Biostatistics Core, Barbara Ann Karmanos Cancer Institute, Detroit, MI
| | - Joseph Fontana
- Department of Oncology, John D. Dingell Veterans Medical Center, Detroit, MI
| |
Collapse
|
88
|
Abstract
mTOR (mechanistic target of rapamycin) functions as the central regulator for cell proliferation, growth and survival. Up-regulation of proteins regulating mTOR, as well as its downstream targets, has been reported in various cancers. This has promoted the development of anti-cancer therapies targeting mTOR, namely fungal macrolide rapamycin, a naturally occurring mTOR inhibitor, and its analogues (rapalogues). One such rapalogue, everolimus, has been approved in the clinical treatment of renal and breast cancers. Although results have demonstrated that these mTOR inhibitors are effective in attenuating cell growth of cancer cells under in vitro and in vivo conditions, subsequent sporadic response to rapalogues therapy in clinical trials has promoted researchers to look further into the complex understanding of the dynamics of mTOR regulation in the tumour environment. Limitations of these rapalogues include the sensitivity of tumour subsets to mTOR inhibition. Additionally, it is well known that rapamycin and its rapalogues mediate their effects by inhibiting mTORC (mTOR complex) 1, with limited or no effect on mTORC2 activity. The present review summarizes the pre-clinical, clinical and recent discoveries, with emphasis on the cellular and molecular effects of everolimus in cancer therapy.
Collapse
|
89
|
Yamazaki K, Eng C, Kuznetsov SA, Reinisch J, Yamashita DD, Walker J, Cheung C, Robey PG, Yen SLK. Missense mutation in the PTEN promoter of a patient with hemifacial hyperplasia. BONEKEY REPORTS 2015; 4:654. [PMID: 26229595 DOI: 10.1038/bonekey.2015.21] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/13/2014] [Accepted: 12/18/2014] [Indexed: 12/28/2022]
Abstract
The cellular mechanisms involved in the asymmetric facial overgrowth syndrome, hemifacial hyperplasia (HFH), are not well understood. This study was conducted to compare primary cell cultures from hyperplastic and normal HFH bone for cellular and molecular differences. Primary cultures developed from biopsies of a patient with isolated HFH showed a twofold difference in cell size and cell number between hyperplastic and normal bone. Microarray data suggested a 40% suppression of PTEN (phosphatase-tensin homolog) transcripts. Sequencing of the PTEN gene and promoter identified novel C/G missense mutation (position -1053) in the regulatory region of the PTEN promoter. Western blots of downstream pathway components showed an increase in PKBa/Akt1 phosphorylation and TOR (target of rapamcyin) signal. Sirolimus, an inhibitor of TOR, when added to overgrowth cells reversed the cell size, cell number and total protein differences between hyperplastic and normal cells. In cases of facial overgrowth, which involve PTEN/Akt/TOR dysregulation, sirolimus could be used for limiting cell overgrowth.
Collapse
Affiliation(s)
- Kiyomi Yamazaki
- Childrens Hospital Los Angeles , Los Angeles, CA, USA ; Center for Craniofacial Molecular Biology, University of Southern California , Los Angeles, CA, USA
| | - Charis Eng
- Clinical Cancer Genetics Program, Human Cancer Genetics Program, Comprehensive Cancer Center, Department of Internal Medicine, The Ohio State University , Columbus, OH, USA
| | - Sergei A Kuznetsov
- Craniofacial and Skeletal Diseases Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health , Bethesda, MD , USA
| | - John Reinisch
- Childrens Hospital Los Angeles , Los Angeles, CA, USA
| | | | - John Walker
- Childrens Hospital Los Angeles , Los Angeles, CA, USA
| | - Craig Cheung
- Center for Craniofacial Molecular Biology, University of Southern California , Los Angeles, CA, USA
| | - Pamela G Robey
- Craniofacial and Skeletal Diseases Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health , Bethesda, MD , USA
| | - Stephen L-K Yen
- Childrens Hospital Los Angeles , Los Angeles, CA, USA ; Center for Craniofacial Molecular Biology, University of Southern California , Los Angeles, CA, USA ; Craniofacial and Skeletal Diseases Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health , Bethesda, MD , USA
| |
Collapse
|
90
|
Karthik GM, Ma R, Lövrot J, Kis LL, Lindh C, Blomquist L, Fredriksson I, Bergh J, Hartman J. mTOR inhibitors counteract tamoxifen-induced activation of breast cancer stem cells. Cancer Lett 2015. [PMID: 26208432 DOI: 10.1016/j.canlet.2015.07.017] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Breast cancer cells with stem cell characteristics (CSC) are a distinct cell population with phenotypic similarities to mammary stem cells. CSCs are important drivers of tumorigenesis and the metastatic process. Tamoxifen is the most widely used hormonal therapy for estrogen receptor (ER) positive cancers. In our study, tamoxifen was effective in reducing proliferation of ER + adherent cancer cells, but not their CSC population. We isolated, expanded and incubated CSC from seven breast cancers with or without tamoxifen. By genome-wide transcriptional analysis we identified tamoxifen-induced transcriptional pathways associated with ribosomal biogenesis and mRNA translation, both regulated by the mTOR-pathway. We observed induction of the key mTOR downstream targets S6K1, S6RP and 4E-BP1 in-patient derived CSCs by tamoxifen on protein level. Using the mTOR inhibitors rapamycin, everolimus and PF-04691502 (a dual PI3K/mTOR inhibitor) and in combination with tamoxifen, significant reduction in mammosphere formation was observed. Hence, we suggest that the CSC population play a significant role during endocrine resistance through activity of the mTOR pathway. In addition, tamoxifen further stimulates the mTOR-pathway but can be antagonized using mTOR-inhibitors.
Collapse
Affiliation(s)
| | - Ran Ma
- Department of Oncology and Pathology, Karolinska Institutet, Stockholm, Sweden
| | - John Lövrot
- Department of Oncology and Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Lorand Levente Kis
- Department of Oncology and Pathology, Karolinska Institutet, Stockholm, Sweden; Department of Clinical Pathology/Cytology, Karolinska University Hospital, 17177 Stockholm, Sweden
| | - Claes Lindh
- Department of Oncology and Pathology, Karolinska Institutet, Stockholm, Sweden; Department of Clinical Pathology/Cytology, Karolinska University Hospital, 17177 Stockholm, Sweden
| | | | - Irma Fredriksson
- Department of Breast and Endocrine Surgery, Karolinska Institutet, Stockholm, Sweden; Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Jonas Bergh
- Department of Oncology and Pathology, Karolinska Institutet, Stockholm, Sweden; Radiumhemmet - Karolinska Oncology, Karolinska University Hospital, 17176 Stockholm, Sweden
| | - Johan Hartman
- Department of Oncology and Pathology, Karolinska Institutet, Stockholm, Sweden; Department of Clinical Pathology/Cytology, Karolinska University Hospital, 17177 Stockholm, Sweden.
| |
Collapse
|
91
|
Gupta S, Argilés G, Munster PN, Hollebecque A, Dajani O, Cheng JD, Wang R, Swift A, Tosolini A, Piha-Paul SA. A Phase I Trial of Combined Ridaforolimus and MK-2206 in Patients with Advanced Malignancies. Clin Cancer Res 2015; 21:5235-44. [PMID: 26187616 DOI: 10.1158/1078-0432.ccr-15-0180] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2015] [Accepted: 06/22/2015] [Indexed: 11/16/2022]
Abstract
PURPOSE The PI3K/Akt/mTOR signaling pathway is aberrantly activated in many cancers. Combining ridaforolimus, an mTOR inhibitor, with MK-2206, an Akt inhibitor, may more completely block the PI3K pathway and inhibit tumor growth. EXPERIMENTAL DESIGN This phase I study assessed dose-limiting toxicities (DLT) and maximum tolerated dose (MTD) for the combination of oral ridaforolimus plus oral MK-2206 in patients with advanced solid tumors. Efficacy was evaluated in patients with biomarker-identified estrogen receptor-positive breast cancer (low RAS gene signature and high Ki67 index) or castration-resistant prostate cancer (PTEN deficiency) with PI3K pathway addiction. RESULTS Thirty-five patients were enrolled: 11 patients in part A (three breast cancer) and 24 biomarker-eligible patients in part B (16 breast cancer, eight prostate cancer). One patient with breast cancer from part A was also found to be biomarker-eligible when tested after she had clinical response. The MTD was 10 mg/d ridaforolimus 5 d/wk + 90 mg/wk MK-2206; 1 of 17 patients experienced DLT (grade 3 rash) at this dose. The most common adverse events at MTD were rash (44.4%), stomatitis (38.9%), diarrhea (27.8%), and decreased appetite (27.8%). By investigator assessment, 2 of 16 (12.5%) evaluable patients with breast cancer had partial response; by central assessment, 2 of 14 (14.3%) evaluable patients had complete response. Two patients had durable stable disease (SD) for 416 and 285 days, respectively. No patients with prostate cancer responded; one patient had SD for ≥ 6 months. CONCLUSIONS Combination ridaforolimus and MK-2206 showed promising activity and good tolerability in heavily pretreated patients with hormone-positive and -negative breast cancer exhibiting PI3K pathway dependence.
Collapse
Affiliation(s)
- Shilpa Gupta
- H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida.
| | - Guillem Argilés
- Vall d'Hebron University Hospital and Vall d'Hebron Institute of Oncology, Barcelona, Spain
| | - Pamela N Munster
- UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, California
| | | | | | - Jonathan D Cheng
- Merck & Co., Inc., Kenilworth, New Jersey and North Wales, Pennsylvania
| | - Ruixue Wang
- Merck & Co., Inc., Kenilworth, New Jersey and North Wales, Pennsylvania
| | - Ann Swift
- Merck & Co., Inc., Kenilworth, New Jersey and North Wales, Pennsylvania
| | | | | |
Collapse
|
92
|
Rathkopf DE, Larson SM, Anand A, Morris MJ, Slovin SF, Shaffer DR, Heller G, Carver B, Rosen N, Scher HI. Everolimus combined with gefitinib in patients with metastatic castration-resistant prostate cancer: Phase 1/2 results and signaling pathway implications. Cancer 2015; 121:3853-61. [PMID: 26178426 DOI: 10.1002/cncr.29578] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Revised: 06/08/2015] [Accepted: 06/09/2015] [Indexed: 12/20/2022]
Abstract
BACKGROUND The effects of mammalian target of rapamycin (mTOR) inhibition are limited by feedback reactivation of receptor tyrosine kinase signaling in phosphatase and tensin homolog-null tumors. Thus, this study tested the combination of mTOR inhibition (everolimus) and epidermal growth factor receptor inhibition (gefitinib) in castration-resistant prostate cancer (CRPC). METHODS In phase 1, 12 patients (10 with CRPC and 2 with glioblastoma) received daily gefitinib (250 mg) with weekly everolimus (30, 50, or 70 mg). In phase 2, 27 CRPC patients received gefitinib with everolimus (70 mg). RESULTS Phase 1 revealed no pharmacokinetic interactions and no dose-limiting toxicities. In phase 2, 18 of 27 patients (67%) discontinued treatment before the 12-week evaluation because of progression as evidenced by prostate-specific antigen (PSA) levels (n = 6) or imaging (n = 5) or because of a grade 2 or higher toxicity (n = 7). Thirteen of the 37 CRPC patients (35%) exhibited a rapidly rising PSA level after they had begun treatment, and this declined upon discontinuation. Fluorodeoxyglucose positron emission tomography 24 to 72 hours after the initiation of treatment showed a decrease in the standardized uptake value consistent with mTOR inhibition in 27 of the 33 evaluable patients (82%); there was a corresponding rise in PSA in 20 of these 27 patients (74%). CONCLUSIONS The combination of gefitinib and everolimus did not result in significant antitumor activity. The induction of PSA in tumors treated with mTOR inhibitors was consistent with preclinical data showing that phosphoinositide 3-kinase (PI3K) pathway signaling feedback inhibits the androgen receptor (AR). This clinical evidence of relief of feedback inhibition promoting enhanced AR activity supports future studies combining PI3K pathway inhibitors and second-generation AR inhibitors in CRPC.
Collapse
Affiliation(s)
- Dana E Rathkopf
- Genitourinary Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York.,Weill Cornell Medical College, New York, New York
| | - Steven M Larson
- Weill Cornell Medical College, New York, New York.,Division of Nuclear Medicine, Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Aseem Anand
- Genitourinary Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York.,Division of Urological Cancers, Lund University, Lund, Sweden
| | - Michael J Morris
- Genitourinary Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York.,Weill Cornell Medical College, New York, New York
| | - Susan F Slovin
- Genitourinary Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York.,Weill Cornell Medical College, New York, New York
| | | | - Glenn Heller
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Brett Carver
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York.,Urology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Neal Rosen
- Program in Molecular Pharmacology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Howard I Scher
- Genitourinary Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York.,Weill Cornell Medical College, New York, New York
| |
Collapse
|
93
|
Abstract
New incidence of prostate cancer is a major public health issue in the Western world, and has been rising in other areas of the globe in recent years. In an effort to understanding the molecular pathogenesis of this disease, numerous cell models have been developed, arising mostly from patient biopsies. The introduction of the genetically engineered mouse in biomedical research has allowed the development of murine models that allow for the investigation of tumorigenic and metastatic processes. Current challenges to the field include lack of an animal model that faithfully recapitulates bone metastasis of prostate cancer.
Collapse
Affiliation(s)
- David Cunningham
- Department of Structural & Cellular Biology, Tulane University Health Sciences Center, New Orleans, LA, USA
| | - Zongbing You
- Department of Structural & Cellular Biology, Tulane University Health Sciences Center, New Orleans, LA, USA
- Department of Orthopaedic Surgery, Tulane University Health Sciences Center, New Orleans, LA, USA
- Tulane Cancer Center and Louisiana Cancer Research Consortium, Tulane University Health Sciences Center, New Orleans, LA, USA
- Tulane Center for Stem Cell Research and Regenerative Medicine, Tulane University Health Sciences Center, New Orleans, LA, USA
- Tulane Center for Aging, Tulane University Health Sciences Center, New Orleans, LA, USA
| |
Collapse
|
94
|
Sun JD, Ahluwalia D, Liu Q, Li W, Wang Y, Meng F, Bhupathi D, Matteucci MD, Hart CP. Combination treatment with hypoxia-activated prodrug evofosfamide (TH-302) and mTOR inhibitors results in enhanced antitumor efficacy in preclinical renal cell carcinoma models. Am J Cancer Res 2015; 5:2139-2155. [PMID: 26328245 PMCID: PMC4548326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2015] [Accepted: 06/11/2015] [Indexed: 06/04/2023] Open
Abstract
Tumors often consist of hypoxic regions which are resistant to chemo- and radiotherapy. Evofosfamide (also known as TH-302), a 2-nitroimidazole triggered hypoxia-activated prodrug, preferentially releases the DNA cross-linker bromo-isophosphoramide mustard in hypoxic cells. The intracellular kinase mTOR plays a key role in multiple pathways which are important in cancer progression. Here we investigated the enhanced efficacy profile and possible mechanisms of evofosfamide in combination with mTOR inhibitor (mTORi) everolimus or temsirolimus in renal cell carcinoma (RCC) xenograft models. The antitumor activities of the mTORi everolimus or temsirolimus alone, evofosfamide alone, or the combination were investigated in the 786-O and Caki-1 RCC cells in vitro and in vivo xenograft models. Two schedules were tested in which evofosfamide was started on the same day as the mTORi or 1 week after. Combination mechanisms were investigated by measuring a panel of pharmacodynamic biomarkers by immunohistochemistry. Antitumor efficacy in both RCC xenograft models was enhanced by the combination of evofosfamide and mTORi. Evofosfamide reduced the increased hypoxia induced by mTORi. Combination treatment induced increased DNA damage, decreased cell proliferation, and decreased survivin. Addition of mTORi did not change evofosfamide-mediated cytotoxicity in 786-O or Caki-1 cells in vitro which might suggest cell non-autonomous effects, specifically increased tumor hypoxia, are important for the in vivo combination activity. Taken together, evofosfamide potentiates the antitumor efficacy of mTOR inhibitors and inhibits the increased tumor hypoxia caused by mTOR inhibition. These studies provide a translational rationale for combining evofosfamide with mTOR inhibitors in clinical studies.
Collapse
Affiliation(s)
- Jessica D Sun
- Threshold Pharmaceuticals 170 Harbor Way, Suite 300, South San Francisco, CA 94080, USA
| | - Dharmendra Ahluwalia
- Threshold Pharmaceuticals 170 Harbor Way, Suite 300, South San Francisco, CA 94080, USA
| | - Qian Liu
- Threshold Pharmaceuticals 170 Harbor Way, Suite 300, South San Francisco, CA 94080, USA
| | - Wenwu Li
- Threshold Pharmaceuticals 170 Harbor Way, Suite 300, South San Francisco, CA 94080, USA
| | - Yan Wang
- Threshold Pharmaceuticals 170 Harbor Way, Suite 300, South San Francisco, CA 94080, USA
| | - Fanying Meng
- Threshold Pharmaceuticals 170 Harbor Way, Suite 300, South San Francisco, CA 94080, USA
| | - Deepthi Bhupathi
- Threshold Pharmaceuticals 170 Harbor Way, Suite 300, South San Francisco, CA 94080, USA
| | - Mark D Matteucci
- Threshold Pharmaceuticals 170 Harbor Way, Suite 300, South San Francisco, CA 94080, USA
| | - Charles P Hart
- Threshold Pharmaceuticals 170 Harbor Way, Suite 300, South San Francisco, CA 94080, USA
| |
Collapse
|
95
|
Bellmunt J, Werner L, Leow JJ, Mullane SA, Fay AP, Riester M, Van Hummelen P, Taplin ME, Choueiri TK, Van Allen E, Rosenberg J. Somatic Copy Number Abnormalities and Mutations in PI3K/AKT/mTOR Pathway Have Prognostic Significance for Overall Survival in Platinum Treated Locally Advanced or Metastatic Urothelial Tumors. PLoS One 2015; 10:e0124711. [PMID: 26039708 PMCID: PMC4454515 DOI: 10.1371/journal.pone.0124711] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2014] [Accepted: 03/05/2015] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND An integrative analysis was conducted to identify genomic alterations at a pathway level that could predict overall survival (OS) in patients with advanced urothelial carcinoma (UC) treated with platinum-based chemotherapy. PATIENTS AND METHODS DNA and RNA were extracted from 103 formalin-fixed paraffin embedded (FFPE) invasive high-grade UC samples and were screened for mutations, copy number variation (CNV) and gene expression analysis. Clinical data were available from 85 cases. Mutations were analyzed by mass-spectrometry based on genotyping platform (Oncomap 3) and genomic imbalances were detected by comparative genomic hybridization (CGH) analysis. Regions with threshold of log2 ratio ≥0.4, or ≤0.6 were defined as either having copy number gain or loss and significantly recurrent CNV across the set of samples were determined using a GISTIC analysis. Expression analysis on selected relevant UC genes was conducted using Nanostring. To define the co-occurrence pattern of mutations and CNV, we grouped genomic events into 5 core signal transduction pathways: 1) TP53 pathway, 2) RTK/RAS/RAF pathway, 3) PI3K/AKT/mTOR pathway, 4) WNT/CTNNB1, 5) RB1 pathway. Cox regression was used to assess pathways abnormalities with survival outcomes. RESULTS 35 samples (41%) harbored mutations on at least one gene: TP53 (16%), PIK3CA (9%), FGFR3 (2%), HRAS/KRAS (5%), and CTNNB1 (1%). 66% of patients had some sort of CNV. PIK3CA/AKT/mTOR pathway alteration (mutations+CNV) had the greatest impact on OS (p=0.055). At a gene level, overexpression of CTNNB1 (p=0.0008) and PIK3CA (p=0.02) were associated with shorter OS. Mutational status on PIK3CA was not associated with survival. Among other individually found genomic alterations, TP53 mutations (p=0.07), mTOR gain (p=0.07) and PTEN overexpression (p=0.08) have a marginally significant negative impact on OS. CONCLUSIONS Our study suggests that targeted therapies focusing on the PIK3CA/AKT/mTOR pathway genomic alterations can generate the greatest impact in the overall patient population of high-grade advanced UC.
Collapse
Affiliation(s)
- Joaquim Bellmunt
- Lank Center for Genitourinary Oncology, Dana-Farber Cancer Institute/Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States of America
| | - Lillian Werner
- Biostatistics and Computational Biology, Harvard Medical School, Dana-Farber Cancer Institute, Boston, MA, United States of America
| | - Jeffrey J. Leow
- Lank Center for Genitourinary Oncology, Dana-Farber Cancer Institute/Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States of America
| | - Stephanie A. Mullane
- Lank Center for Genitourinary Oncology, Dana-Farber Cancer Institute/Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States of America
| | - André P. Fay
- Lank Center for Genitourinary Oncology, Dana-Farber Cancer Institute/Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States of America
| | - Markus Riester
- Biostatistics and Computational Biology, Harvard Medical School, Dana-Farber Cancer Institute, Boston, MA, United States of America
| | - Paul Van Hummelen
- Center for Cancer Genome Discovery, Dana-Farber Cancer Institute, Boston, MA, United States of America
| | - Mary-Ellen Taplin
- Lank Center for Genitourinary Oncology, Dana-Farber Cancer Institute/Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States of America
| | - Toni K. Choueiri
- Lank Center for Genitourinary Oncology, Dana-Farber Cancer Institute/Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States of America
| | - Eliezer Van Allen
- Lank Center for Genitourinary Oncology, Dana-Farber Cancer Institute/Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States of America
| | - Jonathan Rosenberg
- Memorial Sloan Kettering Cancer Center, New York, NY, United States of America
| |
Collapse
|
96
|
Mohan S, Vander Broek R, Shah S, Eytan DF, Pierce ML, Carlson SG, Coupar JF, Zhang J, Cheng H, Chen Z, Van Waes C. MEK Inhibitor PD-0325901 Overcomes Resistance to PI3K/mTOR Inhibitor PF-5212384 and Potentiates Antitumor Effects in Human Head and Neck Squamous Cell Carcinoma. Clin Cancer Res 2015; 21:3946-56. [PMID: 25977343 DOI: 10.1158/1078-0432.ccr-14-3377] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Accepted: 05/04/2015] [Indexed: 01/10/2023]
Abstract
PURPOSE Head and neck squamous cell carcinomas exhibit variable sensitivity to inhibitors of the PI3K/mTOR pathway, an important target of genomic alterations in this cancer type. The mitogen-activated protein kinase kinase (MEK)/ERK/activator protein 1 (AP-1) and nuclear factor-κB (NF-κB) pathways are also frequently co-activated, but their roles in resistance mechanisms to PI3K/mTOR inhibitors and as therapeutic targets in head and neck squamous cell carcinoma (HNSCC) are not well defined. EXPERIMENTAL DESIGN We determined the IC50s of dual PI3K/mTOR inhibitor PF-05212384 (PF-384) by XTT assays in 14 HNSCC lines with PI3K/Akt/mTOR cascade alterations. In two resistant models, we further characterized the molecular, cellular, and in vivo attributes and effects of combining PF-384 with MEK inhibitor PD-0325901 (PD-901). RESULTS PF-384 IC50s varied between 0.75 and 133 nmol/L in 14 HNSCC lines with overexpression or mutations of PIK3CA, and sensitivity correlated with increased phospho-AKT(T308/S473). In resistant UMSCC-1 and -46 models, PF-384 increased G0-/G1-phase accumulation but weakly induced sub-G0 cell death. PF-384 inhibited direct targets of PI3K/mTOR, but incompletely attenuated co-activated ERK and UMSCC-1 xenograft growth in vivo. PD-901 strongly inhibited MEK/ERK targets, and the combination of PF-384 and PD-901 inhibited downstream NF-κB and AP-1 transactivation, and IL8 and VEGF production in vitro. PD-901 potently inhibited tumor growth alone and with PF384, enhanced antiproliferative, apoptotic, and anti-angiogenesis activity in vivo. CONCLUSIONS PI3K/mTOR inhibitor PF-384 exhibits variable activity in a panel of HNSCC cell lines with differing PIK3CA expression and mutation status. MEK inhibitor PD-901 overcomes resistance and enhances antitumor effects observed with PF-384 in vivo.
Collapse
Affiliation(s)
- Suresh Mohan
- Tumor Biology Section, Head and Neck Surgery Branch, NIDCD/NIH, Bethesda, Maryland. NIH Medical Research Scholars Program, Bethesda, Maryland
| | - Robert Vander Broek
- Tumor Biology Section, Head and Neck Surgery Branch, NIDCD/NIH, Bethesda, Maryland. NIH Medical Research Scholars Program, Bethesda, Maryland
| | - Sujay Shah
- Tumor Biology Section, Head and Neck Surgery Branch, NIDCD/NIH, Bethesda, Maryland
| | - Danielle F Eytan
- Tumor Biology Section, Head and Neck Surgery Branch, NIDCD/NIH, Bethesda, Maryland. NIH Medical Research Scholars Program, Bethesda, Maryland
| | - Matthew L Pierce
- Tumor Biology Section, Head and Neck Surgery Branch, NIDCD/NIH, Bethesda, Maryland. Department of Otolaryngology-Head and Neck Surgery, Georgetown University Hospital, Washington, District of Columbia
| | - Sophie G Carlson
- Tumor Biology Section, Head and Neck Surgery Branch, NIDCD/NIH, Bethesda, Maryland
| | - Jamie F Coupar
- Tumor Biology Section, Head and Neck Surgery Branch, NIDCD/NIH, Bethesda, Maryland
| | - Jialing Zhang
- Tumor Biology Section, Head and Neck Surgery Branch, NIDCD/NIH, Bethesda, Maryland
| | - Hui Cheng
- Tumor Biology Section, Head and Neck Surgery Branch, NIDCD/NIH, Bethesda, Maryland
| | - Zhong Chen
- Tumor Biology Section, Head and Neck Surgery Branch, NIDCD/NIH, Bethesda, Maryland.
| | - Carter Van Waes
- Tumor Biology Section, Head and Neck Surgery Branch, NIDCD/NIH, Bethesda, Maryland.
| |
Collapse
|
97
|
Fukuyama M, Kontani K, Katada T, Rougvie AE. The C. elegans Hypodermis Couples Progenitor Cell Quiescence to the Dietary State. Curr Biol 2015; 25:1241-8. [PMID: 25891400 DOI: 10.1016/j.cub.2015.03.016] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2014] [Revised: 01/12/2015] [Accepted: 03/12/2015] [Indexed: 12/11/2022]
Abstract
The nutritional status of an organism can greatly impact the function and behavior of stem and progenitor cells [1]. However, the regulatory circuits that inform these cells about the dietary environment remain to be elucidated. Newly hatched C. elegans larvae (L1s) halt development in "L1 arrest" or "L1 diapause" until ample food is encountered and triggers stem and progenitor cells to exit from quiescence [2]. The insulin/insulin-like growth factor signaling (IIS) pathway plays a key role in this reactivation [3, 4], but its site(s) of action have not been elucidated nor have the nutrient molecule(s) that stimulate the pathway been identified. By tissue-specifically modulating the activity of its components, we demonstrate that the IIS pathway acts in the hypodermis to regulate nutrition-responsive reactivation of neural and mesodermal progenitor cells. We identify ethanol, a likely component of the natural Caenorhabditis habitat, and amino acids as nutrients that synergistically reactivate somatic progenitor cells and upregulate expression of insulin-like genes in starved L1 larvae. The hypodermis likely senses the availability of amino acids because forced activation of the amino-acid-responsive Rag-TORC1 (target of rapamycin complex 1) pathway in this tissue can also release somatic progenitor cell quiescence in the presence of ethanol. Finally, there appears to be crosstalk between the IIS and Rag-TORC1 pathways because constitutive activation of the IIS pathway requires Rag to promote reactivation. This work demonstrates that ethanol and amino acids act as dietary cues via the IIS and Rag-TORC1 pathways in the hypodermis to coordinately control progenitor cell behavior.
Collapse
Affiliation(s)
- Masamitsu Fukuyama
- Department of Genetics, Cell Biology and Development, University of Minnesota, 321 Church Street SE, Minneapolis, MN 55455, USA; Laboratory of Physiological Chemistry, Graduate School of Pharmaceutical Sciences, University of Tokyo, 7-3-1 Hongo, Bunkyo, Tokyo 113-0033, Japan.
| | - Kenji Kontani
- Laboratory of Physiological Chemistry, Graduate School of Pharmaceutical Sciences, University of Tokyo, 7-3-1 Hongo, Bunkyo, Tokyo 113-0033, Japan
| | - Toshiaki Katada
- Laboratory of Physiological Chemistry, Graduate School of Pharmaceutical Sciences, University of Tokyo, 7-3-1 Hongo, Bunkyo, Tokyo 113-0033, Japan
| | - Ann E Rougvie
- Department of Genetics, Cell Biology and Development, University of Minnesota, 321 Church Street SE, Minneapolis, MN 55455, USA.
| |
Collapse
|
98
|
Li N, Sui J, Liu H, Zhong M, Zhang M, Wang Y, Hao F. Expression of phosphorylated Akt/mTOR and clinical significance in human ameloblastoma. Int J Clin Exp Med 2015; 8:5236-5244. [PMID: 26131097 PMCID: PMC4483961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2015] [Accepted: 03/30/2015] [Indexed: 06/04/2023]
Abstract
This study aimed to evaluate the expression of AKT and phosphorylated AKT (p-Akt) in human ameloblastoma (AB). Immunohistochemistry showed human AB was positive for Akt and Akt expression was mainly found in the cytoplasm of epithelial cells. The Akt expression in AB was significantly higher than that in normal oral mucosa (NOM), but still lower than that in oral squamous cell carcinoma (OSCC). NOM was negative for p-Akt, but AB was positive for p-Akt. In some AB tissues, p-Akt expression was found in both cytoplasm and nucleus. Akt expression in AB was significantly different from that in NOM and OSCC. The p-Akt in AB was markedly higher than that in NOM, but lower than that in OSCC. mTOR expressed in cytoplasm in AB, but not in NOM. P-mTOR expressed on cell membrane in NOM, while in cytoplasm and nucleus in Ab. Results of western blot assay showed that Akt expression was found in all the AB tissues, and increased in tissues with malignant transformation. In addition, the p-Akt expression also markedly increased in AB, but was still lower than that in OSCC tissues. Compared to NOM, mTOR and p-mTOR expression significantly increased in AB. BandScan 5.0 software was used to detect the optical density of protein bands. Results showed p-Akt, mTOR and p-mTOR expression in AB was markedly different from that in control group.
Collapse
Affiliation(s)
- Ning Li
- Department of Stomatology, Stomatological Hospital of China Medical UniversityShenyang, China
- Department of Stomatology, 202# Hospital of PLAShenyang, China
| | - Jianfu Sui
- Department of Stomatology, 202# Hospital of PLAShenyang, China
| | - Hao Liu
- Department of Stomatology, Stomatological Hospital of China Medical UniversityShenyang, China
| | - Ming Zhong
- Department of Stomatology, Stomatological Hospital of China Medical UniversityShenyang, China
| | - Min Zhang
- Department of Stomatology, Stomatological Hospital of China Medical UniversityShenyang, China
| | - Yan Wang
- Department of Stomatology, Stomatological Hospital of China Medical UniversityShenyang, China
| | - Fengyu Hao
- Department of Stomatology, Stomatological Hospital of China Medical UniversityShenyang, China
| |
Collapse
|
99
|
Lee M, Wiedemann T, Gross C, Leinhäuser I, Roncaroli F, Braren R, Pellegata NS. Targeting PI3K/mTOR Signaling Displays Potent Antitumor Efficacy against Nonfunctioning Pituitary Adenomas. Clin Cancer Res 2015; 21:3204-15. [DOI: 10.1158/1078-0432.ccr-15-0288] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2015] [Accepted: 03/27/2015] [Indexed: 11/16/2022]
|
100
|
Schreiber KH, Ortiz D, Academia EC, Anies AC, Liao C, Kennedy BK. Rapamycin-mediated mTORC2 inhibition is determined by the relative expression of FK506-binding proteins. Aging Cell 2015; 14:265-73. [PMID: 25652038 PMCID: PMC4364838 DOI: 10.1111/acel.12313] [Citation(s) in RCA: 130] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/12/2014] [Indexed: 01/09/2023] Open
Abstract
The mechanism by which the drug rapamycin inhibits the mechanistic target of rapamycin (mTOR) is of intense interest because of its likely relevance in cancer biology, aging, and other age-related diseases. While rapamycin acutely and directly inhibits mTORC1, only chronic administration of rapamycin can inhibit mTORC2 in some, but not all, cell lines or tissues. The mechanism leading to cell specificity of mTORC2 inhibition by rapamycin is not understood and is especially important because many of the negative metabolic side effects of rapamycin, reported in mouse studies and human clinical trials, have been attributed recently to mTORC2 inhibition. Here, we identify the expression level of different FK506-binding proteins (FKBPs), primarily FKBP12 and FKBP51, as the key determinants for rapamycin-mediated inhibition of mTORC2. In support, enforced reduction of FKBP12 completely converts a cell line that is sensitive to mTORC2 inhibition to an insensitive cell line, and increased expression can enhance mTORC2 inhibition. Further reduction of FKBP12 in cell lines with already low FKBP12 levels completely blocks mTORC1 inhibition by rapamycin, indicating that relative FKBP12 levels are critical for both mTORC1 and mTORC2 inhibition, but at different levels. In contrast, reduction of FKBP51 renders cells more sensitive to mTORC2 inhibition. Our findings reveal that the expression of FKBP12 and FKBP51 is the rate limiting factor that determines the responsiveness of a cell line or tissue to rapamycin. These findings have implications for treating specific diseases, including neurodegeneration and cancer, as well as targeting aging in general.
Collapse
Affiliation(s)
| | - Denise Ortiz
- The Buck Institute for Research on Aging 8001 Redwood Blvd. Novato CA 94945 USA
| | | | - Arieanna C. Anies
- The Buck Institute for Research on Aging 8001 Redwood Blvd. Novato CA 94945 USA
| | - Chen‐Yu Liao
- The Buck Institute for Research on Aging 8001 Redwood Blvd. Novato CA 94945 USA
| | - Brian K. Kennedy
- The Buck Institute for Research on Aging 8001 Redwood Blvd. Novato CA 94945 USA
| |
Collapse
|