151
|
Monsalves E, Juraschka K, Tateno T, Agnihotri S, Asa SL, Ezzat S, Zadeh G. The PI3K/AKT/mTOR pathway in the pathophysiology and treatment of pituitary adenomas. Endocr Relat Cancer 2014; 21:R331-44. [PMID: 25052915 DOI: 10.1530/erc-14-0188] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Pituitary adenomas are common intracranial neoplasms. Patients with these tumors exhibit a wide range of clinically challenging problems, stemming either from results of sellar mass effect in pituitary macroadenoma or the diverse effects of aberrant hormone production by adenoma cells. While some patients are cured/controlled by surgical resection and/or medical therapy, a proportion of patients exhibit tumors that are refractory to current modalities. New therapeutic approaches are needed for these patients. Activation of the AKT/phophotidylinositide-3-kinase pathway, including mTOR activation, is common in human neoplasia, and a number of therapeutic approaches are being employed to neutralize activation of this pathway in human cancer. This review examines the role of this pathway in pituitary tumors with respect to tumor biology and its potential role as a therapeutic target.
Collapse
Affiliation(s)
- Eric Monsalves
- Institute of Medical ScienceDepartment of Medical BiophysicsUniversity of Toronto, Toronto, Ontario, CanadaDivision of NeurosurgeryToronto Western Hospital, 399 Bathurst Street, 4W-439, Toronto, Ontario, Canada M5T 2S8Ontario Cancer InstitutePrincess Margaret Hospital, Toronto, Ontario, CanadaEndocrine Oncology Site GroupPrincess Margaret Hospital, Toronto, Ontario, CanadaDepartment of Laboratory Medicine and PathobiologyUniversity of Toronto, Toronto, Ontario, CanadaInstitute of Medical ScienceDepartment of Medical BiophysicsUniversity of Toronto, Toronto, Ontario, CanadaDivision of NeurosurgeryToronto Western Hospital, 399 Bathurst Street, 4W-439, Toronto, Ontario, Canada M5T 2S8Ontario Cancer InstitutePrincess Margaret Hospital, Toronto, Ontario, CanadaEndocrine Oncology Site GroupPrincess Margaret Hospital, Toronto, Ontario, CanadaDepartment of Laboratory Medicine and PathobiologyUniversity of Toronto, Toronto, Ontario, Canada
| | - Kyle Juraschka
- Institute of Medical ScienceDepartment of Medical BiophysicsUniversity of Toronto, Toronto, Ontario, CanadaDivision of NeurosurgeryToronto Western Hospital, 399 Bathurst Street, 4W-439, Toronto, Ontario, Canada M5T 2S8Ontario Cancer InstitutePrincess Margaret Hospital, Toronto, Ontario, CanadaEndocrine Oncology Site GroupPrincess Margaret Hospital, Toronto, Ontario, CanadaDepartment of Laboratory Medicine and PathobiologyUniversity of Toronto, Toronto, Ontario, Canada
| | - Toru Tateno
- Institute of Medical ScienceDepartment of Medical BiophysicsUniversity of Toronto, Toronto, Ontario, CanadaDivision of NeurosurgeryToronto Western Hospital, 399 Bathurst Street, 4W-439, Toronto, Ontario, Canada M5T 2S8Ontario Cancer InstitutePrincess Margaret Hospital, Toronto, Ontario, CanadaEndocrine Oncology Site GroupPrincess Margaret Hospital, Toronto, Ontario, CanadaDepartment of Laboratory Medicine and PathobiologyUniversity of Toronto, Toronto, Ontario, Canada
| | - Sameer Agnihotri
- Institute of Medical ScienceDepartment of Medical BiophysicsUniversity of Toronto, Toronto, Ontario, CanadaDivision of NeurosurgeryToronto Western Hospital, 399 Bathurst Street, 4W-439, Toronto, Ontario, Canada M5T 2S8Ontario Cancer InstitutePrincess Margaret Hospital, Toronto, Ontario, CanadaEndocrine Oncology Site GroupPrincess Margaret Hospital, Toronto, Ontario, CanadaDepartment of Laboratory Medicine and PathobiologyUniversity of Toronto, Toronto, Ontario, Canada
| | - Sylvia L Asa
- Institute of Medical ScienceDepartment of Medical BiophysicsUniversity of Toronto, Toronto, Ontario, CanadaDivision of NeurosurgeryToronto Western Hospital, 399 Bathurst Street, 4W-439, Toronto, Ontario, Canada M5T 2S8Ontario Cancer InstitutePrincess Margaret Hospital, Toronto, Ontario, CanadaEndocrine Oncology Site GroupPrincess Margaret Hospital, Toronto, Ontario, CanadaDepartment of Laboratory Medicine and PathobiologyUniversity of Toronto, Toronto, Ontario, Canada
| | - Shereen Ezzat
- Institute of Medical ScienceDepartment of Medical BiophysicsUniversity of Toronto, Toronto, Ontario, CanadaDivision of NeurosurgeryToronto Western Hospital, 399 Bathurst Street, 4W-439, Toronto, Ontario, Canada M5T 2S8Ontario Cancer InstitutePrincess Margaret Hospital, Toronto, Ontario, CanadaEndocrine Oncology Site GroupPrincess Margaret Hospital, Toronto, Ontario, CanadaDepartment of Laboratory Medicine and PathobiologyUniversity of Toronto, Toronto, Ontario, CanadaInstitute of Medical ScienceDepartment of Medical BiophysicsUniversity of Toronto, Toronto, Ontario, CanadaDivision of NeurosurgeryToronto Western Hospital, 399 Bathurst Street, 4W-439, Toronto, Ontario, Canada M5T 2S8Ontario Cancer InstitutePrincess Margaret Hospital, Toronto, Ontario, CanadaEndocrine Oncology Site GroupPrincess Margaret Hospital, Toronto, Ontario, CanadaDepartment of Laboratory Medicine and PathobiologyUniversity of Toronto, Toronto, Ontario, CanadaInstitute of Medical ScienceDepartment of Medical BiophysicsUniversity of Toronto, Toronto, Ontario, CanadaDivision of NeurosurgeryToronto Western Hospital, 399 Bathurst Street, 4W-439, Toronto, Ontario, Canada M5T 2S8Ontario Cancer InstitutePrincess Margaret Hospital, Toronto, Ontario, CanadaEndocrine Oncology Site GroupPrincess Margaret Hospital, Toronto, Ontario, CanadaDepartment of Laboratory Medicine and PathobiologyUniversity of Toronto, Toronto, Ontario, Canada
| | - Gelareh Zadeh
- Institute of Medical ScienceDepartment of Medical BiophysicsUniversity of Toronto, Toronto, Ontario, CanadaDivision of NeurosurgeryToronto Western Hospital, 399 Bathurst Street, 4W-439, Toronto, Ontario, Canada M5T 2S8Ontario Cancer InstitutePrincess Margaret Hospital, Toronto, Ontario, CanadaEndocrine Oncology Site GroupPrincess Margaret Hospital, Toronto, Ontario, CanadaDepartment of Laboratory Medicine and PathobiologyUniversity of Toronto, Toronto, Ontario, CanadaInstitute of Medical ScienceDepartment of Medical BiophysicsUniversity of Toronto, Toronto, Ontario, CanadaDivision of NeurosurgeryToronto Western Hospital, 399 Bathurst Street, 4W-439, Toronto, Ontario, Canada M5T 2S8Ontario Cancer InstitutePrincess Margaret Hospital, Toronto, Ontario, CanadaEndocrine Oncology Site GroupPrincess Margaret Hospital, Toronto, Ontario, CanadaDepartment of Laboratory Medicine and PathobiologyUniversity of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
152
|
Jiang N, Liu N, Yang F, Zhou Q, Cui R, Jiang W, He Q, Li W, Guo Y, Zeng J, Yun J, Chen X, Zhou B, Sun Y, Wang H, Chen ZG, Ma J. Hotspot mutations in common oncogenes are infrequent in nasopharyngeal carcinoma. Oncol Rep 2014; 32:1661-9. [PMID: 25109408 DOI: 10.3892/or.2014.3376] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2014] [Accepted: 07/22/2014] [Indexed: 11/05/2022] Open
Abstract
Oncogene mutations contribute to carcinogenesis and can provide potential therapeutic targets for clinical anticancer management. However, oncogene mutation patterns in nasopharyngeal carcinoma (NPC) have yet to be fully elucidated. To gain insight into mutation patterns in NPC, a high-throughput OncoCarta panel assay was used to determine 238 hotspot mutations across 19 common oncogenes in 8 NPC cell lines and 160 NPC patient samples from southern China. Statistical analyses were further conducted to identify associations between oncogene mutations and selected clinicopathological characteristics. In total, we identified 24 mutations across 11 oncogenes in 17 (10.6%) NPC patients. Four patients exhibited mutations in at least one oncogene. We also identified a PIK3CA H1047R mutant in 7 NPC cell lines. In addition, oncogene mutations showed no correlation with either risk habits (smoking and drinking) or other clinical characteristics except for TNM stage. KIT mutations were associated with poorer overall and relapse-free survival. Furthermore, KIT mutations together with age and N stage were independent prognostic factors in NPC. Taken together, the present study is the first report on mutations in multiple oncogenes in NPC. We found that hotspot oncogene mutations are infrequent in NPC patients from southern China. The lack of hotspot mutations requires a comprehensive characterization of gene mutations in NPC for developing new therapeutic targets in the future.
Collapse
Affiliation(s)
- Ning Jiang
- State Key Laboratory of Oncology in Southern China, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, P.R. China
| | - Na Liu
- State Key Laboratory of Oncology in Southern China, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, P.R. China
| | - Fan Yang
- Shenzhen Institute of Liver Diseases, The Third People's Hospital, Shenzhen, Guangdong 518112, P.R. China
| | - Qiming Zhou
- State Key Laboratory of Oncology in Southern China, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, P.R. China
| | - Ruixue Cui
- State Key Laboratory of Oncology in Southern China, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, P.R. China
| | - Wei Jiang
- State Key Laboratory of Oncology in Southern China, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, P.R. China
| | - Qingmei He
- State Key Laboratory of Oncology in Southern China, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, P.R. China
| | - Wenfei Li
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, P.R. China
| | - Ying Guo
- National Clinical Study Center for Anticancer Drugs, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, P.R. China
| | - Jing Zeng
- Department of Pathology, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, P.R. China
| | - Jingping Yun
- Department of Pathology, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, P.R. China
| | - Xinchun Chen
- Shenzhen Institute of Liver Diseases, The Third People's Hospital, Shenzhen, Guangdong 518112, P.R. China
| | - Boping Zhou
- Shenzhen Institute of Liver Diseases, The Third People's Hospital, Shenzhen, Guangdong 518112, P.R. China
| | - Ying Sun
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, P.R. China
| | - Huiyun Wang
- State Key Laboratory of Oncology in Southern China, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, P.R. China
| | - Zhuo G Chen
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Jun Ma
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, P.R. China
| |
Collapse
|
153
|
Zhou W, An G, Wei P, Chen W. Significance of p85 expression as a prognostic factor for patients with breast cancer. Oncol Lett 2014; 8:1657-1661. [PMID: 25202386 PMCID: PMC4156169 DOI: 10.3892/ol.2014.2359] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2014] [Accepted: 06/19/2014] [Indexed: 01/16/2023] Open
Abstract
p85, the regulatory subunit of phosphatidylinositol 3-kinase (PI3K), functions in the pathogenesis and progression of human breast cancers. Previous studies have observed that p85 isoforms may correlate with cancer cell proliferation. In the present study, immunohistochemical staining of p85 was performed in 126 primary breast cancers. The association between the expression levels of p85 with clinicopathological variables, subtypes and prognosis was studied. The breast cancer specimens were divided into three subgroups according to the expression levels of p85 protein. High p85 protein expression was significantly correlated with tumor grade, vascular invasion and recurrence and/or metastasis (P<0.05). Increased p85 protein expression was associated with the human epidermal growth factor receptor 2-positive and triple-negative breast cancers (P=0.008). Patients with higher p85 protein expression levels showed shorter disease-free survival and overall survival times as compared with those with lower expression levels of p85 (P<0.001). Cox proportional-hazards analysis showed that p85 protein expression was not an independent prognostic factor. Further large-scale studies are required to evaluate the significance of p85 protein expression as a prognostic marker for breast cancer.
Collapse
Affiliation(s)
- Weiwei Zhou
- Department of Oncology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, P.R. China
| | - Guangyu An
- Department of Oncology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, P.R. China
| | - Ping Wei
- Department of Pathology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, P.R. China
| | - Wenming Chen
- Department of Hematology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, P.R. China
| |
Collapse
|
154
|
DHA2, a synthesized derivative of bisbibenzyl, exerts antitumor activity against ovarian cancer through inhibition of XIAP and Akt/mTOR pathway. Food Chem Toxicol 2014; 69:163-74. [DOI: 10.1016/j.fct.2014.04.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2013] [Revised: 04/02/2014] [Accepted: 04/05/2014] [Indexed: 11/24/2022]
|
155
|
Hou W, Liu J, Chen P, Wang H, Ye BC, Qiang F. Mutation analysis of key genes in RAS/RAF and PI3K/PTEN pathways in Chinese patients with hepatocellular carcinoma. Oncol Lett 2014; 8:1249-1254. [PMID: 25120700 PMCID: PMC4114707 DOI: 10.3892/ol.2014.2253] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2013] [Accepted: 04/04/2014] [Indexed: 01/05/2023] Open
Abstract
The RAS/RAF and PI3K/PTEN signaling pathways play central roles in hepatocarcinogenesis. KRAS, NRAS, HRAS, BRAF, PIK3CA, PIK3R1 and PTEN are key cancer-related genes in the RAS/RAF and PI3K/PTEN signaling pathways. Genetic alterations in these genes often lead to the dysregulation of the two cascades. Little is known regarding the frequency of hotspot mutations in these critical components among Chinese patients with hepatocellular carcinoma (HCC). In the current study, 57 somatic hotspot mutations in 36 HCCs samples collected from Chinese patients using direct DNA sequencing method were examined. Two cases of KRAS somatic mutations (KRAS codon 61; Gln to His) were identified among 36 HCCs (5.6%). However, no mutations were found in the NRAS, HRAS, BRAF, PIK3CA, PIK3R1 and PTEN genes. These findings indicated that point mutations in the KRAS gene, but not mutations in NRAS, HRAS, BRAF, PIK3CA, PIK3R1 and PTEN genes, at a somatic level contribute to the abnormal activation of the RAS/RAF and PI3K/PTEN pathways in HCC.
Collapse
Affiliation(s)
- Wenmin Hou
- Laboratory of Biosystems and Microanalysis, State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, P.R. China ; Key Laboratory of Food Safety Research, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, P.R. China
| | - Jibin Liu
- Key Laboratory of Food Safety Research, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, P.R. China ; Tumor Institute, Nantong Tumor Hospital, Nantong, Jiangsu 226000, P.R. China
| | - Peizhan Chen
- Key Laboratory of Food Safety Research, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, P.R. China
| | - Hui Wang
- Key Laboratory of Food Safety Research, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, P.R. China
| | - Bang-Ce Ye
- Laboratory of Biosystems and Microanalysis, State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, P.R. China
| | - Fulin Qiang
- Tumor Institute, Nantong Tumor Hospital, Nantong, Jiangsu 226000, P.R. China
| |
Collapse
|
156
|
Khan KH, Yap TA, Yan L, Cunningham D. Targeting the PI3K-AKT-mTOR signaling network in cancer. CHINESE JOURNAL OF CANCER 2014; 32:253-65. [PMID: 23642907 PMCID: PMC3845556 DOI: 10.5732/cjc.013.10057] [Citation(s) in RCA: 154] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
The phosphoinositide 3-kinase-AKT-mammalian target of rapamycin (PI3K-AKT-mTOR) pathway is a frequently hyperactivated pathway in cancer and is important for tumor cell growth and survival. The development of targeted therapies against mTOR, a vital substrate along this pathway, led to the approval of allosteric inhibitors, including everolimus and temsirolimus, for the treatment of breast, renal, and pancreatic cancers. However, the suboptimal duration of response in unselected patients remains an unresolved issue. Numerous novel therapies against critical nodes of this pathway are therefore being actively investigated in the clinic in multiple tumour types. In this review, we focus on the progress of these agents in clinical development along with their biological rationale, the need of predictive biomarkers and various combination strategies, which will be useful in counteracting the mechanisms of resistance to this class of drugs.
Collapse
Affiliation(s)
- Khurum H Khan
- The Royal Marsden NHS Foundation Trust, Sutton, Surrey, UK
| | | | | | | |
Collapse
|
157
|
Beltran PJ, Calzone FJ, Mitchell P, Chung YA, Cajulis E, Moody G, Belmontes B, Li CM, Vonderfecht S, Velculescu VE, Yang G, Qi J, Slamon DJ, Konecny GE. Ganitumab (AMG 479) inhibits IGF-II-dependent ovarian cancer growth and potentiates platinum-based chemotherapy. Clin Cancer Res 2014; 20:2947-58. [PMID: 24727326 DOI: 10.1158/1078-0432.ccr-13-3448] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
PURPOSE Insulin-like growth factor 1 receptor (IGF-IR) has been implicated in the pathogenesis of ovarian cancer. Ganitumab is an investigational, fully human monoclonal antibody against IGF-IR. Here, we explore the therapeutic potential of ganitumab for the treatment of ovarian cancer. EXPERIMENTAL DESIGN The effects of ganitumab were tested in vitro against a panel of 23 established ovarian cancer cell lines. The ability of ganitumab to inhibit IGF-I-, IGF-II-, and insulin-mediated signaling was examined in vitro and in tumor xenografts using ovarian cancer models displaying IGF-IR/PI3K/AKT pathway activation by two distinct mechanisms, PTEN loss and IGF-II overexpression. Drug interactions between ganitumab and cisplatin, carboplatin, or paclitaxel were studied in vitro and in vivo. RESULTS In vitro, growth inhibition varied significantly among individual ovarian cancer cell lines. IGF-II mRNA and phospho-IGF-IR protein expression were quantitatively correlated with response to ganitumab, and PTEN mutations conferred resistance to ganitumab. Ganitumab potently inhibited baseline and IGF-I-, IGF-II-, and insulin-induced IGF-IR and IGF-IR/insulin hybrid receptor signaling in vitro and in vivo. Synergistic and additive drug interactions were seen for ganitumab and carboplatin or paclitaxel in vitro. Furthermore, ganitumab significantly increased the efficacy of cisplatin in ovarian cancer xenograft models in vivo. CONCLUSIONS These observations provide a biologic rationale to test ganitumab as a single agent or in combination with carboplatin/cisplatin and paclitaxel in patients with ovarian cancer. Moreover, assessment of tumor expression of IGF-II, phospho-IGF-IR, or PTEN status may help select patients with ovarian cancer who are most likely to benefit from ganitumab. Clin Cancer Res; 20(11); 2947-58. ©2014 AACR.
Collapse
Affiliation(s)
- Pedro J Beltran
- Authors' Affiliations: Oncology Research Therapeutic Area, Genomics Analysis Unit, Department of Pathology, Amgen Inc., Thousand Oaks; Division of Hematology-Oncology, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California; and The Ludwig Center and the Howard Hughes Medical Institute, Kimmel Cancer Center, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Frank J Calzone
- Authors' Affiliations: Oncology Research Therapeutic Area, Genomics Analysis Unit, Department of Pathology, Amgen Inc., Thousand Oaks; Division of Hematology-Oncology, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California; and The Ludwig Center and the Howard Hughes Medical Institute, Kimmel Cancer Center, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Petia Mitchell
- Authors' Affiliations: Oncology Research Therapeutic Area, Genomics Analysis Unit, Department of Pathology, Amgen Inc., Thousand Oaks; Division of Hematology-Oncology, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California; and The Ludwig Center and the Howard Hughes Medical Institute, Kimmel Cancer Center, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Young-Ah Chung
- Authors' Affiliations: Oncology Research Therapeutic Area, Genomics Analysis Unit, Department of Pathology, Amgen Inc., Thousand Oaks; Division of Hematology-Oncology, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California; and The Ludwig Center and the Howard Hughes Medical Institute, Kimmel Cancer Center, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Elaina Cajulis
- Authors' Affiliations: Oncology Research Therapeutic Area, Genomics Analysis Unit, Department of Pathology, Amgen Inc., Thousand Oaks; Division of Hematology-Oncology, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California; and The Ludwig Center and the Howard Hughes Medical Institute, Kimmel Cancer Center, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Gordon Moody
- Authors' Affiliations: Oncology Research Therapeutic Area, Genomics Analysis Unit, Department of Pathology, Amgen Inc., Thousand Oaks; Division of Hematology-Oncology, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California; and The Ludwig Center and the Howard Hughes Medical Institute, Kimmel Cancer Center, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Brian Belmontes
- Authors' Affiliations: Oncology Research Therapeutic Area, Genomics Analysis Unit, Department of Pathology, Amgen Inc., Thousand Oaks; Division of Hematology-Oncology, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California; and The Ludwig Center and the Howard Hughes Medical Institute, Kimmel Cancer Center, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Chi-Ming Li
- Authors' Affiliations: Oncology Research Therapeutic Area, Genomics Analysis Unit, Department of Pathology, Amgen Inc., Thousand Oaks; Division of Hematology-Oncology, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California; and The Ludwig Center and the Howard Hughes Medical Institute, Kimmel Cancer Center, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Steven Vonderfecht
- Authors' Affiliations: Oncology Research Therapeutic Area, Genomics Analysis Unit, Department of Pathology, Amgen Inc., Thousand Oaks; Division of Hematology-Oncology, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California; and The Ludwig Center and the Howard Hughes Medical Institute, Kimmel Cancer Center, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Victor E Velculescu
- Authors' Affiliations: Oncology Research Therapeutic Area, Genomics Analysis Unit, Department of Pathology, Amgen Inc., Thousand Oaks; Division of Hematology-Oncology, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California; and The Ludwig Center and the Howard Hughes Medical Institute, Kimmel Cancer Center, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Guorong Yang
- Authors' Affiliations: Oncology Research Therapeutic Area, Genomics Analysis Unit, Department of Pathology, Amgen Inc., Thousand Oaks; Division of Hematology-Oncology, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California; and The Ludwig Center and the Howard Hughes Medical Institute, Kimmel Cancer Center, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Jingwei Qi
- Authors' Affiliations: Oncology Research Therapeutic Area, Genomics Analysis Unit, Department of Pathology, Amgen Inc., Thousand Oaks; Division of Hematology-Oncology, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California; and The Ludwig Center and the Howard Hughes Medical Institute, Kimmel Cancer Center, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Dennis J Slamon
- Authors' Affiliations: Oncology Research Therapeutic Area, Genomics Analysis Unit, Department of Pathology, Amgen Inc., Thousand Oaks; Division of Hematology-Oncology, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California; and The Ludwig Center and the Howard Hughes Medical Institute, Kimmel Cancer Center, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Gottfried E Konecny
- Authors' Affiliations: Oncology Research Therapeutic Area, Genomics Analysis Unit, Department of Pathology, Amgen Inc., Thousand Oaks; Division of Hematology-Oncology, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California; and The Ludwig Center and the Howard Hughes Medical Institute, Kimmel Cancer Center, Johns Hopkins Medical Institutions, Baltimore, Maryland
| |
Collapse
|
158
|
Kala S, Mak ASC, Liu X, Posocco P, Pricl S, Peng L, Wong AST. Combination of Dendrimer-Nanovector-Mediated Small Interfering RNA Delivery to Target Akt with the Clinical Anticancer Drug Paclitaxel for Effective and Potent Anticancer Activity in Treating Ovarian Cancer. J Med Chem 2014; 57:2634-42. [DOI: 10.1021/jm401907z] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Shashwati Kala
- School
of Biological Sciences, University of Hong Kong, Pokfulam Road, Pokfulam, Hong Kong
| | - Abby Sin Chi Mak
- School
of Biological Sciences, University of Hong Kong, Pokfulam Road, Pokfulam, Hong Kong
| | - Xiaoxuan Liu
- Aix-Marseille University, CNRS, Centre Interdisciplinaire
de Nanoscience de Marseille, CINaM UMR 7325, Marseille Cedex 09, France
| | - Paola Posocco
- Molecular
Simulation Engineering Laboratory (MOSE), Department of Engineering
and Architecture (DEA), University of Trieste, Via Valerio 10, 34127 Trieste, Italy
- Research
Unit MOSE-DEA, National Interuniversity Consortium for Material Science
and Technology (INSTM), University of Trieste, Via Valerio 6, 34127 Trieste, Italy
| | - Sabrina Pricl
- Molecular
Simulation Engineering Laboratory (MOSE), Department of Engineering
and Architecture (DEA), University of Trieste, Via Valerio 10, 34127 Trieste, Italy
- Research
Unit MOSE-DEA, National Interuniversity Consortium for Material Science
and Technology (INSTM), University of Trieste, Via Valerio 6, 34127 Trieste, Italy
| | - Ling Peng
- Aix-Marseille University, CNRS, Centre Interdisciplinaire
de Nanoscience de Marseille, CINaM UMR 7325, Marseille Cedex 09, France
| | - Alice Sze Tsai Wong
- School
of Biological Sciences, University of Hong Kong, Pokfulam Road, Pokfulam, Hong Kong
| |
Collapse
|
159
|
Chen SF, Cao Y, Han S, Chen JZ. Insight into the structural mechanism for PKBα allosteric inhibition by molecular dynamics simulations and free energy calculations. J Mol Graph Model 2014; 48:36-46. [DOI: 10.1016/j.jmgm.2013.12.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2013] [Revised: 12/02/2013] [Accepted: 12/02/2013] [Indexed: 01/17/2023]
|
160
|
Abstract
OBJECTIVES Mucinous cystic neoplasms (MCNs) are rare, potentially curable, mucin-producing neoplasms of the pancreas. We have previously reported PIK3CA (phosphoinositide-3-kinase catalytic subunit, p110α) mutations in intraductal papillary mucinous neoplasms, another mucin-producing neoplasm of the pancreas. In this study, we analyzed the presence of PIK3CA and AKT1/PKB (V-akt murine thymoma viral oncogene homolog 1) hot-spot mutations in MCN specimens. METHODS Using the genomic DNA sequencing of tumor tissues isolated by laser capture microdissection, we evaluated 15 well-characterized MCNs for the E542K, E545K (exon 9), and H1047R (exon 20) hot-spot mutations in the PIK3CA gene and the E17K mutation in the AKT1 gene. RESULTS A hot-spot mutation (E545K) of the PIK3CA gene was detected in 1 of the 15 MCNs and further confirmed by a mutant-enriched method. Interestingly, this mutation was found to be present only in the high-grade but not in low-grade dysplastic epithelium obtained from this neoplasm and coexisted with a KRAS mutation. No mutations were identified in the AKT1 gene. CONCLUSIONS Our data, when combined with previous reports on intraductal papillary mucinous neoplasms, indicate that oncogenic activation of the PI3K pathway involving PIK3CA gene mutations can contribute to the progression of mucin-producing neoplasms but not pancreatic intraepithelial neoplasia. PIK3CA status could be useful for understanding their progression to malignancy.
Collapse
|
161
|
PIK3CA mutations frequently coexist with EGFR/KRAS mutations in non-small cell lung cancer and suggest poor prognosis in EGFR/KRAS wildtype subgroup. PLoS One 2014; 9:e88291. [PMID: 24533074 PMCID: PMC3922761 DOI: 10.1371/journal.pone.0088291] [Citation(s) in RCA: 120] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2013] [Accepted: 01/06/2014] [Indexed: 01/15/2023] Open
Abstract
PURPOSE PIK3CA gene encoding a catalytic subunit of the phosphatidylinositol-3-kinase (PI3K) is mutated and/or amplified in various neoplasia, including lung cancer. Here we investigated PIK3CA gene alterations, the expression of core components of PI3K pathway, and evaluated their clinical importance in non-small cell lung cancer (NSCLC). MATERIALS AND METHODS Oncogenic mutations/rearrangements in PIK3CA, EGFR, KRAS, HER2, BRAF, AKT1 and ALK genes were detected in tumors from 1117 patients with NSCLC. PIK3CA gene copy number was examined by fluorescent in situ hybridization and the expression of PI3K p110 subunit alpha (PI3K p110α), p-Akt, mTOR, PTEN was determined by immunohistochemistry in PIK3CA mutant cases and 108 patients without PIK3CA mutation. RESULTS PIK3CA mutation was found in 3.9% of squamous cell carcinoma and 2.7% of adenocarcinoma. Among 34 PIK3CA mutant cases, 17 tumors harbored concurrent EGFR mutations and 4 had KRAS mutations. PIK3CA mutation was significantly associated with high expression of PI3K p110α (p<0.0001), p-Akt (p = 0.024) and mTOR (p = 0.001), but not correlated with PIK3CA amplification (p = 0.463). Patients with single PIK3CA mutation had shorter overall survival than those with PIK3CA-EGFR/KRAS co-mutation or wildtype PIK3CA (p = 0.004). A significantly worse survival was also found in patients with PIK3CA mutations than those without PIK3CA mutations in the EGFR/KRAS wildtype subgroup (p = 0.043). CONCLUSIONS PIK3CA mutations frequently coexist with EGFR/KRAS mutations. The poor prognosis of patients with single PIK3CA mutation in NSCLC and the prognostic value of PIK3CA mutation in EGFR/KRAS wildtype subgroup suggest the distinct mutation status of PIK3CA gene should be determined for individual therapeutic strategies in NSCLC.
Collapse
|
162
|
Jeong JY, Kim KS, Moon JS, Song JA, Choi SH, Kim KI, Kim TH, An HJ. Targeted inhibition of phosphatidyl inositol-3-kinase p110β, but not p110α, enhances apoptosis and sensitivity to paclitaxel in chemoresistant ovarian cancers. Apoptosis 2014; 18:509-20. [PMID: 23371322 PMCID: PMC3604599 DOI: 10.1007/s10495-013-0807-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The phosphatidylinositol 3-kinase (PI3K) pathway is one of the critical signaling cascades playing important roles in the chemoresistance of human cancer cells, including ovarian cancer. In this study, we investigated the potential of targeting the PI3K p110β-isoform as a novel approach to overcome the chemoresistance in ovarian cancer. The effects on apoptosis, cell viability, proliferation and migration in chemoresistant ovarian cancer cell were determined following targeted p110β inhibition by small interfering RNA (siRNA). Seven paclitaxel (PTX)-resistant sublines (SKpacs and A2780pac) were produced from SKOV3 and A2780 ovarian cancer cell lines. We, first, evaluated the expression of PI3K p110 isoforms in chemosensitive and chemoresistant ovarian cancer cell lines and patient specimens, and found that p110β-isoform was significantly overexpressed both in a panel of ovarian cancer samples, and in PTX-resistant sublines compared with their parent cell lines. RNA interference-mediated p110β silencing augmented PTX-mediated apoptosis (31.15 ± 13.88 %) and reduced cell viability (67 %) in PTX-resistant cells, whereas targeting p110α did not show a significant change in cell viability and apoptosis. In addition, p110β silencing impaired cell proliferation (60 %) in PTX-resistant SKpac cells. We also found the combined treatment group with p110β siRNA and PTX showed a significant inhibition of tumor growth of SKpac cells compared to the PTX-only treated group in a xenograft nude mouse model. Thus, the siRNA-mediated silencing of PI3K p110β resensitizes PTX-resistant ovarian cancer cells, and may be a useful therapeutic strategy for PTX-resistant ovarian cancers.
Collapse
Affiliation(s)
- Ju-yeon Jeong
- Department of Pathology, College of Medicine, CHA University, 351 Yatap-dong, Gyeonggi-Do, Seongnam Si Bundang-gu 463-712, Republic of Korea
| | | | | | | | | | | | | | | |
Collapse
|
163
|
Kosmidou V, Oikonomou E, Vlassi M, Avlonitis S, Katseli A, Tsipras I, Mourtzoukou D, Kontogeorgos G, Zografos G, Pintzas A. Tumor Heterogeneity Revealed byKRAS,BRAF, andPIK3CAPyrosequencing:KRASandPIK3CAIntratumor Mutation Profile Differences and Their Therapeutic Implications. Hum Mutat 2014; 35:329-40. [PMID: 24352906 DOI: 10.1002/humu.22496] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2013] [Accepted: 12/05/2013] [Indexed: 01/03/2023]
Affiliation(s)
- Vivian Kosmidou
- Laboratory of Signal Mediated Gene Expression; Institute of Biology, Medicinal Chemistry and Biotechnology; National Hellenic Research Foundation; Athens 11635 Greece
| | - Eftychia Oikonomou
- Laboratory of Signal Mediated Gene Expression; Institute of Biology, Medicinal Chemistry and Biotechnology; National Hellenic Research Foundation; Athens 11635 Greece
| | - Margarita Vlassi
- Laboratory of Signal Mediated Gene Expression; Institute of Biology, Medicinal Chemistry and Biotechnology; National Hellenic Research Foundation; Athens 11635 Greece
| | - Spyros Avlonitis
- 3rd Department of Surgery; General Hospital of Athens G. Gennimatas; Athens 15669 Greece
| | - Anastasia Katseli
- 3rd Department of Surgery; General Hospital of Athens G. Gennimatas; Athens 15669 Greece
| | - Iraklis Tsipras
- 3rd Department of Surgery; General Hospital of Athens G. Gennimatas; Athens 15669 Greece
| | - Despina Mourtzoukou
- Department of Pathology; General Hospital of Athens G. Gennimatas; Athens 15669 Greece
| | - Georgios Kontogeorgos
- Department of Pathology; General Hospital of Athens G. Gennimatas; Athens 15669 Greece
| | - Georgios Zografos
- 3rd Department of Surgery; General Hospital of Athens G. Gennimatas; Athens 15669 Greece
| | - Alexander Pintzas
- Laboratory of Signal Mediated Gene Expression; Institute of Biology, Medicinal Chemistry and Biotechnology; National Hellenic Research Foundation; Athens 11635 Greece
| |
Collapse
|
164
|
Zhang XY, Birtwistle MR, Gallo JM. A General Network Pharmacodynamic Model-Based Design Pipeline for Customized Cancer Therapy Applied to the VEGFR Pathway. CPT-PHARMACOMETRICS & SYSTEMS PHARMACOLOGY 2014; 3:e92. [PMID: 24429593 PMCID: PMC3910016 DOI: 10.1038/psp.2013.65] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/19/2013] [Accepted: 10/21/2013] [Indexed: 01/02/2023]
Abstract
A unified approach to optimize multidrug chemotherapy using a pharmacokinetic (PK)/enhanced pharmacodynamic model was developed using the vascular endothelial growth factor receptor (VEGFR) signaling system. The base VEGFR network model, characterized by ligand–receptor interactions, enzyme recruitment (Grb2-Sos, phospholipase C γ (PLCγ), and phosphoinositide-3 kinase (PI3K)), and downstream mitogen-activated protein kinase and Akt cascade activation, was linked to a sunitinib (VEGFR inhibitor) PK model and underwent Sobol sensitivity analysis that revealed potential sunitinib-enhancing mechanisms. Drugs targeting these mechanisms (a VEGF inhibitor, a PI3K inhibitor, a PLCγ inhibitor, and a mitogen-activated protein kinase inhibitor) and sunitinib were input to optimization-based control analyses to design multidrug regimens that maintained 80% pERK and pAkt inhibition for 28 days while minimizing drug dose. The resultant combination regimens contained both continuous and discontinuous schedules, mostly at low doses, and were altered by oncogenic mutations. This pipeline of computational analyses demonstrates how model-based methods can capture the complexities of drug action, tailor cancer chemotherapy, and empower personalized medicine.
Collapse
Affiliation(s)
- X-Y Zhang
- Department of Pharmacology and Systems Therapeutics, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - M R Birtwistle
- Department of Pharmacology and Systems Therapeutics, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - J M Gallo
- Department of Pharmacology and Systems Therapeutics, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| |
Collapse
|
165
|
Oncogenic PIK3CA mutation and dysregulation in human salivary duct carcinoma. BIOMED RESEARCH INTERNATIONAL 2014; 2014:810487. [PMID: 24511546 PMCID: PMC3910486 DOI: 10.1155/2014/810487] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/29/2013] [Revised: 12/12/2013] [Accepted: 12/21/2013] [Indexed: 12/29/2022]
Abstract
Salivary duct carcinoma (SDC) is an aggressive malignant tumor with a high mortality, which resembles high-grade breast ductal carcinoma in morphology. The parotid gland is the most common location. Its molecular genetic characteristics remain largely unknown. We have previously reported high incidence of PIK3CA somatic mutations in head and neck squamous cell carcinoma, particularly in pharyngeal cancers. Here we examined the PIK3CA gene expression status and hotspot mutations in six cases of SDC by immunohistochemistry and genomic DNA sequencing. Immunohistochemistry showed that PIK3CA expression was elevated in all six patients with SDC. By DNA sequencing, two hotspot mutations of the PIK3CA gene, E545K (exon 9) and H1047R (exon 20), were identified in two of the six cases. Our results support that oncogenic PIK3CA is upregulated and frequently mutated in human SDC, adding evidence that PIK3CA oncogenic pathway is critical in the tumorigenesis of SDC, and may be a plausible drug target for this rare disease.
Collapse
|
166
|
Averett C, Arora S, Zubair H, Singh S, Bhardwaj A, Singh AP. Molecular Targets of Honokiol: A Promising Phytochemical for Effective Cancer Management. Enzymes 2014; 36:175-93. [PMID: 27102704 DOI: 10.1016/b978-0-12-802215-3.00009-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Honokiol is a bioactive, biphenolic phytochemical, present in the aerial parts of plants classified under the genus Magnolia. It has been an important constituent of Asian traditional medicine and is used against many ailments. Honokiol possesses potent antioxidative, anti-inflammatory, antiangiogenic, and anticancer activities by targeting a variety of signaling molecules. Consequently, there has been immense interest in exploring its utility as a novel chemopreventive and therapeutic agent against several malignancies. In this chapter, we review the structure-function relationship of honokiol and its derivative compounds, the impact of honokiol on various phenotypes associated with cancer progression and metastasis, and its prominent molecular targets and pharmacokinetics. Clearly, the available data generate significant interest in this novel phytochemical and emerging information continues to provide strong support for its potential applicability in cancer management.
Collapse
Affiliation(s)
- Courey Averett
- Department of Oncologic Sciences, Mitchell Cancer Institute, University of South Alabama, Mobile, Alabama, USA
| | - Sumit Arora
- Department of Oncologic Sciences, Mitchell Cancer Institute, University of South Alabama, Mobile, Alabama, USA
| | - Haseeb Zubair
- Department of Oncologic Sciences, Mitchell Cancer Institute, University of South Alabama, Mobile, Alabama, USA
| | - Seema Singh
- Department of Oncologic Sciences, Mitchell Cancer Institute, University of South Alabama, Mobile, Alabama, USA
| | - Arun Bhardwaj
- Department of Oncologic Sciences, Mitchell Cancer Institute, University of South Alabama, Mobile, Alabama, USA
| | - Ajay P Singh
- Department of Oncologic Sciences, Mitchell Cancer Institute, University of South Alabama, Mobile, Alabama, USA; Department of Biochemistry and Molecular Biology, College of Medicine, University of South Alabama, Mobile, Alabama, USA.
| |
Collapse
|
167
|
Acquired PIK3CA amplification causes resistance to selective phosphoinositide 3-kinase inhibitors in breast cancer. Oncogenesis 2013; 2:e83. [PMID: 24366379 PMCID: PMC3940863 DOI: 10.1038/oncsis.2013.46] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2013] [Revised: 09/21/2013] [Accepted: 10/29/2013] [Indexed: 12/11/2022] Open
Abstract
Agents targeting the PI3K/mTOR signaling axis have shown promise in early-phase clinical trials and are currently being studied in later stages of clinical development in multiple indications. Experience with other targeted agents suggests that clinical responses may be short-lived because of acquired resistance to therapy. Here, we report preclinical modeling of acquired resistance in a HER2-positive, PIK3CA mutant breast cancer cell line, KPL-4. We identified a heretofore-unreported mechanism of resistance, specifically high-level amplification of the mutant allele of PIK3CA, which resulted in a marked upregulation of PI3K signaling, enabling resistant cells to regain proliferative capacity at clinically relevant concentrations of the PI3K inhibitor, GDC-0941. We show that knockdown of the amplified PIK3CA mutant allele in these cells by small interfering RNA restored pathway signaling and sensitivity to PI3K inhibition at levels comparable to parental cells. These novel preclinical findings suggest that, in addition to assessment of other previously reported mechanisms of resistance, evaluation of PI3K copy number variation should be integrated into the exploratory analysis of biopsies obtained at disease progression.
Collapse
|
168
|
Molecular targets for epithelial ovarian cancer. Mol Oncol 2013. [DOI: 10.1017/cbo9781139046947.055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
|
169
|
PI3K. Mol Oncol 2013. [DOI: 10.1017/cbo9781139046947.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
|
170
|
Kim MS, Kim JE, Lim DY, Huang Z, Chen H, Langfald A, Lubet RA, Grubbs CJ, Dong Z, Bode AM. Naproxen induces cell-cycle arrest and apoptosis in human urinary bladder cancer cell lines and chemically induced cancers by targeting PI3K. Cancer Prev Res (Phila) 2013; 7:236-45. [PMID: 24327721 DOI: 10.1158/1940-6207.capr-13-0288] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Naproxen [(S)-6-methoxy-α-methyl-2-naphthaleneacetic acid] is a potent nonsteroidal anti-inflammatory drug that inhibits both COX-1 and COX-2 and is widely used as an over-the-counter medication. Naproxen exhibits analgesic, antipyretic, and anti-inflammatory activities. Naproxen, as well as other nonsteroidal anti-inflammatory drug, has been reported to be effective in the prevention of urinary bladder cancer in rodents. However, potential targets other than the COX isozymes have not been reported. We examined potential additional targets in urinary bladder cancer cells and in rat bladder cancers. Computer kinase profiling results suggested that phosphoinositide 3-kinase (PI3K) is a potential target for naproxen. In vitro kinase assay data revealed that naproxen interacts with PI3K and inhibits its kinase activity. Pull-down binding assay data confirmed that PI3K directly binds with naproxen in vitro and ex vivo. Western blot data showed that naproxen decreased phosphorylation of Akt, and subsequently decreased Akt signaling in UM-UC-5 and UM-UC-14 urinary bladder cancer cells. Furthermore, naproxen suppressed anchorage-independent cell growth and decreased cell viability by targeting PI3K in both cell lines. Naproxen caused an accumulation of cells at the G1 phase mediated through cyclin-dependent kinase 4, cyclin D1, and p21. Moreover, naproxen induced significant apoptosis, accompanied with increased levels of cleaved caspase-3, caspase-7, and PARP in both cell types. Naproxen-induced cell death was mainly because of apoptosis in which a prominent downregulation of Bcl-2 and upregulation of Bax were involved. Naproxen also caused apoptosis and inhibited Akt phosphorylation in rat urinary bladder cancers induced by N-butyl-N-(4-hydroxybutyl)-nitrosamine.
Collapse
Affiliation(s)
- Mi-Sung Kim
- The Hormel Institute University of Minnesota, 801 16th Avenue NE, Austin, MN 55912.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
171
|
Romano G. The role of the dysfunctional akt-related pathway in cancer: establishment and maintenance of a malignant cell phenotype, resistance to therapy, and future strategies for drug development. SCIENTIFICA 2013; 2013:317186. [PMID: 24381788 PMCID: PMC3870877 DOI: 10.1155/2013/317186] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/21/2013] [Accepted: 11/14/2013] [Indexed: 06/01/2023]
Abstract
Akt serine/threonine kinases, or PKB, are key players in the regulation of a wide variety of cellular activities, such as growth, proliferation, protection from apoptotic injuries, control of DNA damage responses and genome stability, metabolism, migration, and angiogenesis. The Akt-related pathway responds to the stimulation mediated by growth factors, cytokines, hormones, and several nutrients. Akt is present in three isoforms: Akt1, Akt2, and Akt3, which may be alternatively named PKB α , PKB β , and PKB γ , respectively. The Akt isoforms are encoded on three diverse chromosomes and their biological functions are predominantly distinct. Deregulations in the Akt-related pathway were observed in many human maladies, including cancer, cardiopathies, neurological diseases, and type-2 diabetes. This review discusses the significance of the abnormal activities of the Akt axis in promoting and sustaining malignancies, along with the development of tumor cell populations that exhibit enhanced resistance to chemo- and/or radiotherapy. This occurrence may be responsible for the relapse of the disease, which is unfortunately very often related to fatal consequences in patients.
Collapse
Affiliation(s)
- Gaetano Romano
- Department of Biology, College of Science and Technology, Temple University, Bio Life Science Building, Suite 456, 1900 N. 12th Street, Philadelphia, PA 19122, USA
| |
Collapse
|
172
|
Mierke CT. The role of focal adhesion kinase in the regulation of cellular mechanical properties. Phys Biol 2013; 10:065005. [PMID: 24304934 DOI: 10.1088/1478-3975/10/6/065005] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
The regulation of mechanical properties is necessary for cell invasion into connective tissue or intra- and extravasation through the endothelium of blood or lymph vessels. Cell invasion is important for the regulation of many healthy processes such as immune response reactions and wound healing. In addition, cell invasion plays a role in disease-related processes such as tumor metastasis and autoimmune responses. Until now the role of focal adhesion kinase (FAK) in regulating mechanical properties of cells and its impact on cell invasion efficiency is still not well known. Thus, this review focuses on mechanical properties regulated by FAK in comparison to the mechano-regulating protein vinculin. Moreover, it points out the connection between cancer cell invasion and metastasis and FAK by showing that FAK regulates cellular mechanical properties required for cellular motility. Furthermore, it sheds light on the indirect interaction of FAK with vinculin by binding to paxillin, which then impairs the binding of paxillin to vinculin. In addition, this review emphasizes whether FAK fulfills regulatory functions similar to vinculin. In particular, it discusses the differences and the similarities between FAK and vinculin in regulating the biomechanical properties of cells. Finally, this paper highlights that both focal adhesion proteins, vinculin and FAK, synergize their functions to regulate the mechanical properties of cells such as stiffness and contractile forces. Subsequently, these mechanical properties determine cellular invasiveness into tissues and provide a source sink for future drug developments to inhibit excessive cell invasion and hence, metastases formation.
Collapse
Affiliation(s)
- Claudia Tanja Mierke
- Faculty of Physics and Earth Science, Institute of Experimental Physics I, Biological Physics Division, University of Leipzig, Linnéstr. 5, D-04103 Leipzig, Germany
| |
Collapse
|
173
|
Jiang G, Huang Z, Zhang S, Wang L. PIK3CAGene Mutations and Amplifications in Chinese Patients With Ovarian Clear Cell Carcinoma. Cancer Invest 2013; 31:639-44. [DOI: 10.3109/07357907.2013.853075] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Gaofeng Jiang
- 1School of Public Health, Medical College, Wuhan University of Science and Technology, Wuhan, Hubei, China
| | | | | | | |
Collapse
|
174
|
Lin L, Gaut D, Hu K, Yan H, Yin D, Koeffler HP. Dual targeting of glioblastoma multiforme with a proteasome inhibitor (Velcade) and a phosphatidylinositol 3-kinase inhibitor (ZSTK474). Int J Oncol 2013; 44:557-62. [PMID: 24297065 PMCID: PMC3898871 DOI: 10.3892/ijo.2013.2205] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2013] [Accepted: 10/16/2013] [Indexed: 02/02/2023] Open
Abstract
Proteasome inhibitors have been proven to be effective anticancer compounds in many tumor models, including glioblastoma multiforme (GBM). In this study, we found that the proteasome inhibitor Velcade (PS-341/bortezomib) caused GBM cell death while simultaneously activating the PI3K/Akt pathway. Therefore, we sought to investigate if the PI3K inhibitor ZSTK474 would enhance the effectiveness of Velcade in anticancer therapy. Two GBM cell lines were used to detect the effects of Velcade and ZSTK474 alone or in combination in vitro. The combination of Velcade and ZSTK474 synergistically inhibited the proliferation of GBM cell lines. Cell apoptosis was increased when exposed to Velcade and ZSTK474 in combination as shown by Annexin V analysis. Treatment with both drugs led to downregulation of the p-Akt, p-4EBP1 and p-mTOR proteins as determined by western blot analysis. The anticancer ability of Velcade for glioblastoma multiforme was, therefore, enhanced by combination with the PI3K pathway inhibitor ZSTK474 in glioblastoma multiforme.
Collapse
Affiliation(s)
- Lehang Lin
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, P.R. China
| | - Daria Gaut
- Division of Hematology/Oncology, Cedars-Sinai Medical Center, UCLA School of Medicine, Los Angeles, CA 90048, USA
| | - Kaishun Hu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, P.R. China
| | - Haiyan Yan
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, P.R. China
| | - Dong Yin
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, P.R. China
| | - H Phillip Koeffler
- Division of Hematology/Oncology, Cedars-Sinai Medical Center, UCLA School of Medicine, Los Angeles, CA 90048, USA
| |
Collapse
|
175
|
Villaruz LC, Burns TF, Ramfidis VS, Socinski MA. Personalizing therapy in advanced non-small cell lung cancer. Semin Respir Crit Care Med 2013; 34:822-36. [PMID: 24258572 DOI: 10.1055/s-0033-1358552] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The recognition that non-small cell lung cancer (NSCLC) is not a single disease entity, but rather a collection of distinct molecularly driven neoplasms, has permanently shifted the therapeutic landscape of NSCLC to a personalized approach. This personalization of NSCLC therapy is typified by the dramatic response rates seen in EGFR mutant NSCLC when treated with targeted tyrosine kinase inhibitor therapy and in ALK translocation-driven NSCLC when treated with ALK inhibitors. Targeted therapeutic approaches in NSCLC necessitate consideration of more invasive biopsy techniques aimed at providing sufficient tissue for both histological determination and molecular profiling in all patients with stage IV disease both at the time of diagnosis and at the time of disease progression. Comprehensive genotyping efforts have identified oncogenic drivers in 62% lung adenocarcinomas and an increasing proportion of squamous cell carcinomas of the lung. The identification of these oncogenic drivers and the triage of patients to clinical trials evaluating novel targeted therapeutic approaches will increasingly mold a landscape of personalized lung cancer therapy where each genotype has an associated targeted therapy. This review outlines the state of personalized lung cancer therapy as it pertains to individual NSCLC genotypes.
Collapse
Affiliation(s)
- Liza C Villaruz
- Lung and Thoracic Malignancies Program, University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania
| | | | | | | |
Collapse
|
176
|
Garcia-Carracedo D, Turk AT, Fine SA, Akhavan N, Tweel BC, Parsons R, Chabot JA, Allendorf JD, Genkinger JM, Remotti HE, Su GH. Loss of PTEN expression is associated with poor prognosis in patients with intraductal papillary mucinous neoplasms of the pancreas. Clin Cancer Res 2013; 19:6830-41. [PMID: 24132918 DOI: 10.1158/1078-0432.ccr-13-0624] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
PURPOSE Previously, we reported PIK3CA gene mutations in high-grade intraductal papillary mucinous neoplasms (IPMN). However, the contribution of phosphatidylinositol-3 kinase pathway (PI3K) dysregulation to pancreatic carcinogenesis is not fully understood and its prognostic value unknown. We investigated the dysregulation of the PI3K signaling pathway in IPMN and its clinical implication. EXPERIMENTAL DESIGN Thirty-six IPMN specimens were examined by novel mutant-enriched sequencing methods for hot-spot mutations in the PIK3CA and AKT1 genes. PIK3CA and AKT1 gene amplifications and loss of heterozygosity at the PTEN locus were also evaluated. In addition, the expression levels of PDPK1/PDK1, PTEN, and Ki67 were analyzed by immunohistochemistry. RESULTS Three cases carrying the E17K mutation in the AKT1 gene and one case harboring the H1047R mutation in the PIK3CA gene were detected among the 36 cases. PDK1 was significantly overexpressed in the high-grade IPMN versus low-grade IPMN (P = 0.034) and in pancreatic and intestinal-type of IPMN versus gastric-type of IPMN (P = 0.020). Loss of PTEN expression was strongly associated with presence of invasive carcinoma and poor survival in these IPMN patients (P = 0.014). CONCLUSION This is the first report of AKT1 mutations in IPMN. Our data indicate that oncogenic activation of the PI3K pathway can contribute to the progression of IPMN, in particular loss of PTEN expression. This finding suggests the potential employment of PI3K pathway-targeted therapies for IPMN patients. The incorporation of PTEN expression status in making surgical decisions may also benefit IPMN patients and should warrant further investigation.
Collapse
Affiliation(s)
- Dario Garcia-Carracedo
- Authors' Affiliations: Herbert Irving Comprehensive Cancer Center; Departments of Pathology, Surgery, and Otolaryngology/Head and Neck Surgery; Institute for Cancer Genetics, Columbia University Medical Center; and The Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, New York
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
177
|
Li Q, Yang J, Yu Q, Wu H, Liu B, Xiong H, Hu G, Zhao J, Yuan X, Liao Z. Associations between single-nucleotide polymorphisms in the PI3K-PTEN-AKT-mTOR pathway and increased risk of brain metastasis in patients with non-small cell lung cancer. Clin Cancer Res 2013; 19:6252-60. [PMID: 24077347 DOI: 10.1158/1078-0432.ccr-13-1093] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Non-small cell lung cancer (NSCLC) metastasizes fairly often to the brain, but identifying which patients will develop brain metastases is problematic. The phosphoinositide 3-kinase (PI3K)-AKT-mTOR signaling pathway is important in the control of cell growth, tumorigenesis, and cell invasion. We hypothesized that genotype variants in this pathway could predict brain metastasis in patients with NSCLC. METHODS We genotyped 16 single-nucleotide polymorphisms (SNP) in five core genes (PIK3CA, PTEN, AKT1, AKT2, and FRAP1) by using DNA from blood samples of 317 patients with NSCLC, and evaluated potential associations with the subsequent development of brain metastasis, the cumulative incidence of which was estimated with Kaplan-Meier analysis. Multivariate Cox regression analysis was used to analyze correlations between genotype variants and the occurrence of brain metastasis. RESULTS In analysis of individual SNPs, the GT/GG genotype of AKT1: rs2498804, CT/TT genotype of AKT1: rs2494732, and AG/AA genotype of PIK3CA: rs2699887 were associated with higher risk of brain metastasis at 24-month follow-up [respective HRs, 1.860, 95% confidence interval (CI) 1.199-2.885, P = 0.006; HR 1.902, 95% CI 1.259-2.875, P = 0.002; and HR 1.933, 95% CI 1.168-3.200, P = 0.010]. We further found that these SNPs had a cumulative effect on brain metastasis risk, with that risk being highest for patients carrying both of these unfavorable genotypes (P = 0.003). CONCLUSIONS Confirmation of our findings, the first to indicate that genetic variations in PI3K-AKT-mTOR can predict brain metastasis, in prospective studies would facilitate stratification of patients for brain metastasis prevention trials.
Collapse
Affiliation(s)
- Qianxia Li
- Authors' Affiliations: Department of Oncology, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, Hubei Province, China; and Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | | | | | | | | | | | | | | | | | | |
Collapse
|
178
|
Yip WK, Choo CW, Leong VCS, Leong PP, Jabar MF, Seow HF. Molecular alterations of Ras-Raf-mitogen-activated protein kinase and phosphatidylinositol 3-kinase-Akt signaling pathways in colorectal cancers from a tertiary hospital at Kuala Lumpur, Malaysia. APMIS 2013; 121:954-66. [DOI: 10.1111/apm.12152] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2013] [Accepted: 06/24/2013] [Indexed: 12/31/2022]
Affiliation(s)
- Wai Kien Yip
- Department of Pathology; Faculty of Medicine and Health Sciences; Universiti Putra Malaysia
| | - Chee Wei Choo
- Department of Pathology; Faculty of Medicine and Health Sciences; Universiti Putra Malaysia
| | | | - Pooi Pooi Leong
- Department of Pathology; Faculty of Medicine and Health Sciences; Universiti Putra Malaysia
| | - Mohd Faisal Jabar
- Department of Surgery; Faculty of Medicine and Health Sciences; Universiti Putra Malaysia
| | - Heng Fong Seow
- Department of Pathology; Faculty of Medicine and Health Sciences; Universiti Putra Malaysia
- Institute of Bioscience; Universiti Putra Malaysia; Serdang Selangor Malaysia
| |
Collapse
|
179
|
Iacovides DC, Johnson AB, Wang N, Boddapati S, Korkola J, Gray JW. Identification and quantification of AKT isoforms and phosphoforms in breast cancer using a novel nanofluidic immunoassay. Mol Cell Proteomics 2013; 12:3210-20. [PMID: 23929892 DOI: 10.1074/mcp.m112.023119] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Breast cancer subtype-specific molecular variations can dramatically affect patient responses to existing therapies. It is thought that differentially phosphorylated protein isoforms might be a useful prognostic biomarker of drug response in the clinic. However, the accurate detection and quantitative analysis of cancer-related protein isoforms and phospho-isoforms in tumors are limited by current technologies. Using a novel, fully automated nanocapillary electrophoresis immunoassay (NanoPro(TM) 1000) designed to separate protein molecules based on their isoelectric point, we developed a reliable and highly sensitive assay for the detection and quantitation of AKT isoforms and phosphoforms in breast cancer. This assay enabled the measurement of activated AKT1/2/3 in breast cancer cells using protein produced from as few as 56 cells. Importantly, we were able to assign an identity for the phosphorylated S473 phosphoform of AKT1, the major form of activated AKT involved in multiple cancers, including breast, and a current focus in clinical trials for targeted intervention. The ability of our AKT assay to detect and measure AKT phosphorylation from very low amounts of total protein will allow the accurate evaluation of patient response to drugs targeting activated PI3K-AKT using scarce clinical specimens. Moreover, the capacity of this assay to detect and measure all three AKT isoforms using one single pan-specific antibody enables the study of the multiple and variable roles that these isoforms play in AKT tumorigenesis.
Collapse
Affiliation(s)
- Demetris C Iacovides
- Life Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, California 94720
| | | | | | | | | | | |
Collapse
|
180
|
Abstract
This article presents an overview of the PI3K/Akt/mTOR signaling pathway. As a central regulator of cell growth, protein translation, survival, and metabolism, activation of this signaling pathway contributes to the pathogenesis of many tumor types. Biochemical and genetic aberrations of this pathway observed in various cancer types are explored. Last, pathway inhibitors both in development and already approved by the Food and Drug Administration are discussed.
Collapse
|
181
|
Badinloo M, Esmaeili-Mahani S. Phosphatidylinositol 3-kinases inhibitor LY294002 potentiates the cytotoxic effects of doxorubicin, vincristine, and etoposide in a panel of cancer cell lines. Fundam Clin Pharmacol 2013; 28:414-22. [PMID: 23837575 DOI: 10.1111/fcp.12043] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2013] [Revised: 06/10/2013] [Accepted: 06/12/2013] [Indexed: 11/29/2022]
Abstract
Many novel therapeutic approaches to overcome chemoresistance have involved targeting specific signaling pathways such as the phosphatidylinositol 3-kinase (PI3K) pathway. PI3K is a known stress response pathway which is involved in the regulation of cell survival, apoptosis, and growth. Inhibition of this pathway may possibly restore or augment the effectiveness of chemotherapy. Using three human malignant cell lines, we examined the effects of LY294002 (PI3K inhibitor) on chemotherapeutic agent-induced apoptosis and cytotoxicity. An antimicrotubule agent vincristine, a topoisomerase II inhibitor etoposide, and a DNA cross-linking agent doxorubicin were used accompanied with LY294002. Cell viability was determined by MTT assay, and the induction of apoptosis was assessed by immunoblotting of caspase-3. Blocking the PI3K/Akt cascade with a PI3K inhibitor LY294002 (10 μM) increased the cytotoxic effect of vincristine and doxorubicin on SK-OV-3 cell line. Furthermore, LY294002 showed a greater promoting effect in etoposide- and doxorubicin-induced cytotoxicity on MDA-MB-468 and A549 cells. The quantity of cleaved caspase-3 in cancer cells that had combination therapy was increased compared with that in the cells treated with each drug alone. We suggest that inhibitors of the PI3K/Akt pathway in combination with chemotherapeutic agents may induce cell death effectively and be a potent modality to treat various types of cancer. The effectiveness of such combination therapy is depending to the used cell line and class of anticancer drug.
Collapse
Affiliation(s)
- Marzieh Badinloo
- Physiology Research Center, Kerman University of Medical Sciences, Jahad Blvd, 7619813159, Kerman, Iran
| | | |
Collapse
|
182
|
The Synthetic Compound Norcantharidin Induced Apoptosis in Mantle Cell Lymphoma In Vivo and In Vitro through the PI3K-Akt-NF- κ B Signaling Pathway. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2013; 2013:461487. [PMID: 23935664 PMCID: PMC3722980 DOI: 10.1155/2013/461487] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/24/2013] [Revised: 05/30/2013] [Accepted: 06/10/2013] [Indexed: 02/07/2023]
Abstract
This study aimed to elucidate the antitumor activity of norcantharidin (NCTD) against human mantle cell lymphoma (MCL). Cell proliferation and apoptosis were examined by MTS and flow cytometry. Caspase-3, -8, and -9 activities were detected with a colorimetric caspase protease assay. Apoptotic proteins—including PARP, cyclin D1, Bcl-2 family proteins, XIAP, and cIAP I—were studied by western blot. The phosphoinositide 3 kinase (PI3K) inhibitor LY294002 was used to investigate the involvement of the PI3K/Akt signaling pathway. In vivo studies were performed using Z138 cell xenografts in nude mice. NCTD inhibited proliferation and induced apoptosis of Z138 and Mino cells, both in vitro and in vivo. PI3Kp110α and p-Akt expressions were downregulated by NCTD treatment. NCTD downregulated NF-κB activity by preventing NF-κB phosphorylation and nuclear translocation. This effect was correlated with the suppression of NF-κB-regulated gene products, such as cyclin D1, BAX, survivin, Bcl-2, XIAP, and cIAP. This phenomenon was blocked by the PI3K inhibitor LY294002. Our results demonstrated that NCTD can induce growth arrest and apoptosis in MCL cells and that the mechanism may involve the PI3K/Akt/NF-κB signaling pathway. NCTD may have therapeutic and/or adjuvant therapeutic applications in the treatment of MCL.
Collapse
|
183
|
Irusta G, Maidana CP, Abramovich D, De Zúñiga I, Parborell F, Tesone M. Effects of an Inhibitor of the Gamma-Secretase Complex on Proliferation and Apoptotic Parameters in a FOXL2-Mutated Granulosa Tumor Cell Line (KGN)1. Biol Reprod 2013; 89:9. [DOI: 10.1095/biolreprod.113.108100] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
|
184
|
Stegh AH. Toward personalized cancer nanomedicine - past, present, and future. Integr Biol (Camb) 2013; 5:48-65. [PMID: 22858688 DOI: 10.1039/c2ib20104f] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Tumors are composed of highly proliferate, migratory, invasive, and therapy-evading cells. These characteristics are conferred by an enormously complex landscape of genomic, (epi-)genetic, and proteomic aberrations. Recent efforts to comprehensively catalogue these reversible and irreversible modifications have began to identify molecular mechanisms that contribute to cancer pathophysiology, serve as novel therapeutic targets, and may constitute biomarkers for early diagnosis and prediction of therapy responses. With constantly evolving technologies that will ultimately enable a complete survey of cancer genomes, the challenges for discovery cancer science and drug development are daunting. Bioinformatic and functional studies must differentiate cancer-driving and -contributing mutations from mere bystanders or 'noise', and have to delineate their molecular mechanisms of action as a function of collaborating oncogenic and tumor suppressive signatures. In addition, the translation of these genomic discoveries into meaningful clinical endpoints requires the development of co-extinction strategies to therapeutically target multiple cancer genes, to robustly deliver therapeutics to tumor sites, and to enable widespread dissemination of therapies within tumor tissue. In this perspective, I will describe the most current paradigms to study and validate cancer gene function. I will highlight advances in the area of nanotechnology, in particular, the development of RNA interference (RNAi)-based platforms to more effectively deliver therapeutic agents to tumor sites, and to modulate critical cancer genes that are difficult to target using conventional small-molecule- or antibody-based approaches. I will conclude with an outlook on the deluge of challenges that genomic and bioengineering sciences must overcome to make the long-awaited era of personalized nano-medicine a clinical reality for cancer patients.
Collapse
Affiliation(s)
- Alexander H Stegh
- Ken and Ruth Davee Department of Neurology, The Brain Tumor Institute, The Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL 60611, USA.
| |
Collapse
|
185
|
Antal CE, Newton AC. Spatiotemporal dynamics of phosphorylation in lipid second messenger signaling. Mol Cell Proteomics 2013; 12:3498-508. [PMID: 23788531 DOI: 10.1074/mcp.r113.029819] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
The plasma membrane serves as a dynamic interface that relays information received at the cell surface into the cell. Lipid second messengers coordinate signaling on this platform by recruiting and activating kinases and phosphatases. Specifically, diacylglycerol and phosphatidylinositol 3,4,5-trisphosphate activate protein kinase C and Akt, respectively, which then phosphorylate target proteins to transduce downstream signaling. This review addresses how the spatiotemporal dynamics of protein kinase C and Akt signaling can be monitored using genetically encoded reporters and provides information on how the coordination of signaling at protein scaffolds or membrane microdomains affords fidelity and specificity in phosphorylation events.
Collapse
Affiliation(s)
- Corina E Antal
- Department of Pharmacology, University of California at San Diego, La Jolla, California 92093-0721
| | | |
Collapse
|
186
|
Network nonlinearities in drug treatment. Interdiscip Sci 2013; 5:85-94. [PMID: 23740389 DOI: 10.1007/s12539-013-0165-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2012] [Revised: 12/25/2012] [Accepted: 03/22/2013] [Indexed: 10/26/2022]
Abstract
Despite major achievements in the understanding of human disease, there is a general perception that the drug development industry has failed to meet the expectations that recent advances in biotechnology should drive. One of the potential sources of failure of many next generation drugs is that their targets are embedded in highly nonlinear signaling pathways and gene networks with multiple negative and positive feedback loops of regulation. There is increasing evidence that this complex network shapes the response to external perturbations in the form of drug treatment, originating bistability, hypersensitivity, robustness, complex dose-response curves or schedule dependent activity. This review focuses on the effect of nonlinearities on signaling and gene networks involved in human disease, using tools from Nonlinear Dynamics to discuss the implications and to overcome the effects of the nonlinearities on regulatory networks.
Collapse
|
187
|
Furet P, Guagnano V, Fairhurst RA, Imbach-Weese P, Bruce I, Knapp M, Fritsch C, Blasco F, Blanz J, Aichholz R, Hamon J, Fabbro D, Caravatti G. Discovery of NVP-BYL719 a potent and selective phosphatidylinositol-3 kinase alpha inhibitor selected for clinical evaluation. Bioorg Med Chem Lett 2013; 23:3741-8. [PMID: 23726034 DOI: 10.1016/j.bmcl.2013.05.007] [Citation(s) in RCA: 333] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2013] [Revised: 04/30/2013] [Accepted: 05/04/2013] [Indexed: 12/26/2022]
Abstract
Phosphatidylinositol-3-kinase α (PI3Kα) is a therapeutic target of high interest in anticancer drug research. On the basis of a binding model rationalizing the high selectivity and potency of a particular series of 2-aminothiazole compounds in inhibiting PI3Kα, a medicinal chemistry program has led to the discovery of the clinical candidate NVP-BYL719.
Collapse
Affiliation(s)
- Pascal Furet
- Novartis Institutes for BioMedical Research, WKL-136.4.12, CH-4002 Basel, Switzerland
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
188
|
Talekar M, Ganta S, Amiji M, Jamieson S, Kendall J, Denny WA, Garg S. Development of PIK-75 nanosuspension formulation with enhanced delivery efficiency and cytotoxicity for targeted anti-cancer therapy. Int J Pharm 2013; 450:278-89. [PMID: 23632263 DOI: 10.1016/j.ijpharm.2013.04.057] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2013] [Revised: 04/15/2013] [Accepted: 04/16/2013] [Indexed: 02/07/2023]
Abstract
PIK-75 is a phosphatidylinositol 3-kinase (PI3K) inhibitor that shows selectivity toward p110-α over the other PI3K class Ia isoforms p110-β and p110-δ, but it lacks solubility, stability and other kinase selectivity. The purpose of this study was to develop folate-targeted PIK-75 nanosuspension for tumor targeted delivery and to improve therapeutic efficacy in human ovarian cancer model. High pressure homogenization was used to prepare the non-targeted and targeted PIK-75 nanosuspensions which were characterized for size, zeta potential, entrapment efficiency, morphology, saturation solubility and dissolution velocity. In vitro analysis of drug uptake, cell viability and cell survival was conducted in SKOV-3 cells. Drug pharmacokinetics and pAkt expression were determined in SKOV-3 tumor bearing mice. PIK-75 nanosuspensions showed an improvement in dissolution velocity and an 11-fold increase in saturation solubility over pre-milled PIK-75. In vitro studies in SKOV-3 cells indicated a 2-fold improvement in drug uptake and 0.4-fold decrease in IC50 value of PIK-75 following treatment with targeted nanosuspension compared to non-targeted nanosuspension. The improvement in cytotoxicity was attributed to an increase in caspase 3/7 and hROS activity. In vivo studies indicated a 5-10-fold increased PIK-75 accumulation in the tumor with both the nanosuspension formulations compared to PIK-75 suspension. The targeted nanosuspension showed an enhanced downregulation of pAkt compared to non-targeted formulation system. These results illustrate the opportunity to formulate PIK-75 as a targeted nanosuspension to enhance uptake and cytotoxicity of the drug in tumor.
Collapse
Affiliation(s)
- Meghna Talekar
- School of Pharmacy, Faculty of Medical and Health Sciences, The University of Auckland, Private Bag 92019, Auckland, New Zealand
| | | | | | | | | | | | | |
Collapse
|
189
|
Zhang Y, Duc ACE, Rao S, Sun XL, Bilbee AN, Rhodes M, Li Q, Kappes DJ, Rhodes J, Wiest DL. Control of hematopoietic stem cell emergence by antagonistic functions of ribosomal protein paralogs. Dev Cell 2013; 24:411-25. [PMID: 23449473 DOI: 10.1016/j.devcel.2013.01.018] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2011] [Revised: 11/15/2012] [Accepted: 01/20/2013] [Indexed: 01/14/2023]
Abstract
It remains controversial whether the highly homologous ribosomal protein (RP) paralogs found in lower eukaryotes have distinct functions and this has not been explored in vertebrates. Here we demonstrate that despite ubiquitous expression, the RP paralogs, Rpl22 and Rpl22-like1 (Rpl22l1) play essential, distinct, and antagonistic roles in hematopoietic development. Knockdown of Rpl22 in zebrafish embryos selectively blocks the development of T lineage progenitors after they have seeded the thymus. In contrast, knockdown of the Rpl22 paralog, Rpl22l1, impairs the emergence of hematopoietic stem cells (HSC) in the aorta-gonad-mesonephros by abrogating Smad1 expression and the consequent induction of essential transcriptional regulator, Runx1. Indeed, despite the ability of both paralogs to bind smad1 RNA, Rpl22 and Rpl22l1 have opposing effects on Smad1 expression. Accordingly, circumstances that tip the balance of these paralogs in favor of Rpl22 (e.g., Rpl22l1 knockdown or Rpl22 overexpression) result in repression of Smad1 and blockade of HSC emergence.
Collapse
Affiliation(s)
- Yong Zhang
- Blood Cell Development and Cancer Keystone, Immune Cell Development and Host Defense Program, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
190
|
Genetic deregulation of the PIK3CA oncogene in oral cancer. Cancer Lett 2013; 338:193-203. [PMID: 23597702 DOI: 10.1016/j.canlet.2013.04.005] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2012] [Revised: 03/11/2013] [Accepted: 04/09/2013] [Indexed: 01/05/2023]
Abstract
The phosphatidylinositol 3-kinase (PI3K)/Akt signaling pathway is one of the most commonly deregulated pathways in human cancers. PI3K comprises a catalytic (p110α) and regulatory subunit (p85), and p110α is encoded by the PIK3CA gene. Here, we summarize the known genetic alterations, including amplifications and mutations, of the PIK3CA oncogene in oral cancer. We discuss in detail PIK3CA mutations and their mutual exclusivity with pathway genes in addition to the incidence of PIK3CA mutations in relation to ethnicity. We describe the constitutive activation of PI3K signaling, oncogenicity, and the genetic deregulation of the PIK3CA gene and its association with oral cancer disease stage. We emphasize the importance of therapeutically targeting the genetically deregulated PIK3CA oncogene and its signaling. We also discuss the implications of targeting Akt and/or mTOR, which are the downstream effectors of PI3K that may possibly pave the way for molecular therapeutic targets for PIK3CA-driven oral carcinogenesis. Furthermore, this critical review provides a complete picture of the PIK3CA oncogene and its deregulation in oral cancer, which may facilitate early diagnosis and improve prognosis through personalized molecular targeted therapy in oral cancer.
Collapse
|
191
|
Lin J, Sampath D, Nannini MA, Lee BB, Degtyarev M, Oeh J, Savage H, Guan Z, Hong R, Kassees R, Lee LB, Risom T, Gross S, Liederer BM, Koeppen H, Skelton NJ, Wallin JJ, Belvin M, Punnoose E, Friedman LS, Lin K. Targeting activated Akt with GDC-0068, a novel selective Akt inhibitor that is efficacious in multiple tumor models. Clin Cancer Res 2013; 19:1760-72. [PMID: 23287563 DOI: 10.1158/1078-0432.ccr-12-3072] [Citation(s) in RCA: 232] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE We describe the preclinical pharmacology and antitumor activity of GDC-0068, a novel highly selective ATP-competitive pan-Akt inhibitor currently in clinical trials for the treatment of human cancers. EXPERIMENTAL DESIGN The effect of GDC-0068 on Akt signaling was characterized using specific biomarkers of the Akt pathway, and response to GDC-0068 was evaluated in human cancer cell lines and xenograft models with various genetic backgrounds, either as a single agent or in combination with chemotherapeutic agents. RESULTS GDC-0068 blocked Akt signaling both in cultured human cancer cell lines and in tumor xenograft models as evidenced by dose-dependent decrease in phosphorylation of downstream targets. Inhibition of Akt activity by GDC-0068 resulted in blockade of cell-cycle progression and reduced viability of cancer cell lines. Markers of Akt activation, including high-basal phospho-Akt levels, PTEN loss, and PIK3CA kinase domain mutations, correlate with sensitivity to GDC-0068. Isogenic PTEN knockout also sensitized MCF10A cells to GDC-0068. In multiple tumor xenograft models, oral administration of GDC-0068 resulted in antitumor activity ranging from tumor growth delay to regression. Consistent with the role of Akt in a survival pathway, GDC-0068 also enhanced antitumor activity of classic chemotherapeutic agents. CONCLUSIONS GDC-0068 is a highly selective, orally bioavailable Akt kinase inhibitor that shows pharmacodynamic inhibition of Akt signaling and robust antitumor activity in human cancer cells in vitro and in vivo. Our preclinical data provide a strong mechanistic rationale to evaluate GDC-0068 in cancers with activated Akt signaling.
Collapse
Affiliation(s)
- Jie Lin
- Genentech, South San Francisco, CA 94080, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
192
|
Clauditz TS, Gontarewicz A, Bokemeyer C, Sauter G, Knecht R, Münscher A, Wilczak W. Abundant expression of mTOR kinase in salivary gland tumors - potentials as therapy target? J Oral Pathol Med 2013; 42:769-73. [PMID: 23521157 DOI: 10.1111/jop.12064] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/28/2013] [Indexed: 12/22/2022]
Abstract
BACKGROUND Salivary gland tumors constitute 3-6% of all head and neck neoplasms in adults. Because of limited advances made in the treatment of metastatic disease, the more important is the role of new therapeutic strategies, including molecular therapy. The mammalian target of rapamycin (mTOR) has recently been established as a therapeutic target for several drugs. MATERIAL Evaluation of phospho-mTOR as a possible therapy target by patients with salivary gland tumors. Immunohistochemical semi-quantitative analyses of the expression of phospho-mTOR(Ser2448) were processed on a tissue microarray containing samples from more than 900 patients. For statistical analysis, contingency table and chi-squared test (likelihood) were used. RESULTS We observed at least weak phospho-mTOR expression in 25.6-41.2% of all 4 histological adenoma and in 36.8-61.6% of all 11 histological carcinoma subtypes analyzed. No association was seen between phospho-mTOR expression and tumor grade in mucoepidermoid carcinomas. CONCLUSION In conjunction with literature data providing the evidence for a functional role of mTOR in salivary gland tumors, we conclude that treatment with mTOR-antagonists might potentially also be efficient in wide variety of salivary gland carcinomas.
Collapse
|
193
|
De Marco C, Rinaldo N, Bruni P, Malzoni C, Zullo F, Fabiani F, Losito S, Scrima M, Marino FZ, Franco R, Quintiero A, Agosti V, Viglietto G. Multiple genetic alterations within the PI3K pathway are responsible for AKT activation in patients with ovarian carcinoma. PLoS One 2013; 8:e55362. [PMID: 23408974 PMCID: PMC3567053 DOI: 10.1371/journal.pone.0055362] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2012] [Accepted: 12/21/2012] [Indexed: 11/19/2022] Open
Abstract
The phosphatidylinositol 3-kinase (PI3K)/AKT pathway is activated in multiple cancers including ovarian carcinoma (OC). However, the relative contribution of the single components within the PI3K pathway to AKT activation in OC is still unclear. We examined 98 tumor samples from Italian OC patients for alterations in the members of the PI3K pathway. We report that AKT is significantly hyperactive in OC compared to normal tissue (n = 93; p<0.0001) and that AKT activation is preferentially observed in the elderly (>58 years old; n = 93; p<0.05). The most frequent alteration is the overexpression of the p110α catalytic subunit of PI3K (63/93, ∼68%); less frequent alterations comprise the loss of PTEN (24/89, 27%) and the overexpression of AKT1 (18/96, 19%) or AKT2 (11/88,12.5%). Mutations in the PIK3CA or KRAS genes were detected at lower frequency (12% and 10%, respectively) whereas mutations in AKT1 or AKT2 genes were absent. Although many tumors presented a single lesion (28/93, of which 23 overexpressed PIK3CA, 1 overexpressed AKT and 4 had lost PTEN), many OC (35/93) presented multiple alterations within the PI3K pathway. Apparently, aberrant PI3K signalling was mediated by activation of the canonical downstream AKT-dependent mTOR/S6K1/4EBP1 pathway and by regulation of expression of oncogenic transcription factors that include HMGA1, JUN-B, FOS and MYC but not by AKT-independent activation of SGK3. FISH analysis indicated that gene amplification of PIK3CA, AKT1 and AKT2 (but not of PI3KR1) and the loss of PTEN are common and may account for changes in the expression of the corresponding proteins. In conclusion, our results indicate that p110α overexpression represents the most frequent alteration within the PI3K/AKT pathway in OC. However, p110α overexpression may not be sufficient to activate AKT signalling and drive ovarian tumorigenesis since many tumors overexpressing PI3K presented at least one additional alteration.
Collapse
Affiliation(s)
- Carmela De Marco
- Department of Experimental and Clinical Medicine, University Magna Graecia, Catanzaro, Italy
- Biogem Scarl, Institute for Genetic Research “Gaetano Salvatore”, Ariano Irpino, Avellino, Italy
| | - Nicola Rinaldo
- Biogem Scarl, Institute for Genetic Research “Gaetano Salvatore”, Ariano Irpino, Avellino, Italy
| | - Paola Bruni
- Casa di Cura “Malzoni-Villa dei Platani”, Avellino, Italy
| | | | - Fulvio Zullo
- Department of Experimental and Clinical Medicine, University Magna Graecia, Catanzaro, Italy
| | - Fernanda Fabiani
- Department of Experimental and Clinical Medicine, University Magna Graecia, Catanzaro, Italy
| | - Simona Losito
- Fondazione “G Pascale”, National Cancer Institute, Naples, Italy
| | - Marianna Scrima
- Department of Experimental and Clinical Medicine, University Magna Graecia, Catanzaro, Italy
- Biogem Scarl, Institute for Genetic Research “Gaetano Salvatore”, Ariano Irpino, Avellino, Italy
| | | | - Renato Franco
- Fondazione “G Pascale”, National Cancer Institute, Naples, Italy
| | - Alfina Quintiero
- Department of Experimental and Clinical Medicine, University Magna Graecia, Catanzaro, Italy
| | - Valter Agosti
- Department of Experimental and Clinical Medicine, University Magna Graecia, Catanzaro, Italy
| | - Giuseppe Viglietto
- Department of Experimental and Clinical Medicine, University Magna Graecia, Catanzaro, Italy
- Biogem Scarl, Institute for Genetic Research “Gaetano Salvatore”, Ariano Irpino, Avellino, Italy
- * E-mail:
| |
Collapse
|
194
|
Wik E, Ræder MB, Krakstad C, Trovik J, Birkeland E, Hoivik EA, Mjos S, Werner HMJ, Mannelqvist M, Stefansson IM, Oyan AM, Kalland KH, Akslen LA, Salvesen HB. Lack of estrogen receptor-α is associated with epithelial-mesenchymal transition and PI3K alterations in endometrial carcinoma. Clin Cancer Res 2013; 19:1094-105. [PMID: 23319822 DOI: 10.1158/1078-0432.ccr-12-3039] [Citation(s) in RCA: 112] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE We hypothesized that estrogen receptor-α (ER-α) status in endometrial carcinomas, associated with poor prognosis, is reflected in transcriptional signatures suggesting targets for new therapy. EXPERIMENTAL DESIGN Endometrial carcinoma samples in a primary investigation cohort (n = 76) and three independent validation cohorts (n = 155/286/111) were analyzed through integrated molecular profiling. Biomarkers were assessed by immunohistochemistry (IHC), DNA oligonucleotide microarray, quantitative PCR (qPCR), single-nucleotide polymorphism (SNP) array, and Sanger sequencing in the cohorts, annotated for comprehensive histopathologic and clinical data, including follow-up. RESULTS ER-α immunohistochemical staining was strongly associated with mRNA expression of the receptor gene (ESR1) and patient survival (both P < 0.001). ER-α negativity associated with activation of genes involved in Wnt-, Sonic Hedgehog-, and TGF-β signaling in the investigation cohort, indicating epithelial-mesenchymal transition (EMT). The association between low ER-α and EMT was validated in three independent datasets. Furthermore, phosphoinositide 3-kinase (PI3K) and mTOR inhibitors were among the top-ranked drug signatures negatively correlated with the ER-α-negative tumors. Low ER-α was significantly associated with PIK3CA amplifications but not mutations. Also, low ER-α was correlated to high expression of Stathmin, a marker associated with PTEN loss, and a high PI3K activation signature. CONCLUSION Lack of ER-α in endometrial cancer is associated with EMT and reduced survival. We present a rationale for investigating ER-α's potential to predict response to PI3K/mTOR inhibitors in clinical trials and also suggest EMT inhibitors to ER-α-negative endometrial carcinomas.
Collapse
Affiliation(s)
- Elisabeth Wik
- Department of Obstetrics and Gynecology, Haukeland University Hospital, Bergen, Norway.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
195
|
Cheung M, Testa JR. Diverse mechanisms of AKT pathway activation in human malignancy. Curr Cancer Drug Targets 2013; 13:234-44. [PMID: 23297823 DOI: 10.2174/1568009611313030002] [Citation(s) in RCA: 142] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2010] [Revised: 07/20/2012] [Accepted: 01/08/2013] [Indexed: 01/12/2023]
Abstract
AKT/PKB (Protein Kinase B) are central proteins mediating signals from receptor tyrosine kinases and phosphatidylinositol 3-kinase. AKT kinases are involved in a number of important cellular processes including cell proliferation and survival, cell size in response to nutrient availability, tumor invasion/metastasis, and angiogenesis.Various components of the AKT signaling pathway are encoded by tumor suppressor genes and oncogenes whose loss or activation, respectively, plays an important role in tumorigenesis. The growing body of evidence connecting deregulated AKT signaling with sporadic human cancers and inherited cancer predisposition syndromes is discussed. We also highlight new findings regarding the involvement of activating mutations of AKT1, AKT2, and AKT3 in somatic overgrowth disorders: Proteus syndrome, hypoglycemia with hypertrophy, and hemimegalencephaly, respectively. In addition, we review recent literature documenting the various ways the AKT signaling pathway is activated in human cancers and consequences for molecularly targeted therapies.
Collapse
Affiliation(s)
- Mitchell Cheung
- Cancer Biology Program, Fox Chase Cancer Center, 333 Cottman Avenue, Philadelphia, PA 19111-2497, USA
| | | |
Collapse
|
196
|
King SM, Quartuccio SM, Vanderhyden BC, Burdette JE. Early transformative changes in normal ovarian surface epithelium induced by oxidative stress require Akt upregulation, DNA damage and epithelial-stromal interaction. Carcinogenesis 2013; 34:1125-33. [PMID: 23299406 DOI: 10.1093/carcin/bgt003] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Ovarian cancer is the deadliest gynecological malignancy due to detection of cancer at a late stage when the disease has metastasized. One likely progenitor cell type of ovarian cancer is the ovarian surface epithelium (OSE), which proliferates rapidly in the presence of inflammatory cytokines and oxidative stress following ovulation. To determine whether oxidative stress induces DNA damage leading to spontaneous transformative changes in normal OSE, an immortalized mouse OSE cell line (MOSE cells) or normal mouse ovarian organoids were treated with hydrogen peroxide (H2O2) and loss of contact inhibition was assessed by soft agar assay. In response to H2O2, OSE cells grown in 3D exhibited growth in soft agar but MOSE cells grown on 2D plastic did not, indicating a critical role for epithelial-stromal interactions in neoplastic initiation. Loss of contact inhibition in response to H2O2 correlated with an increase in proliferation, DNA damage and upregulation of the oncogene Akt1. Use of a reactive oxygen species scavenger or Akt inhibitor blocked H2O2-induced proliferation and growth in soft agar. Although parental MOSE cells did not undergo transformation by H2O2, MOSE cells stably overexpressing constitutively active myristoylated Akt or knockdown of phosphatase and tensin homolog (PTEN) exhibited loss of contact inhibition and increased proliferation. This study indicates that normal OSE undergo transformative changes induced by oxidative stress and that this process requires Akt upregulation and activation. A 3D model that retains tissue architecture is critical for studying this process and may lead to development of new intervention strategies directed at early stages of ovarian cancer.
Collapse
Affiliation(s)
- Shelby M King
- Department of Medicinal Chemistry and Pharmacognosy, University of Illinois at Chicago, Chicago, IL 60607, USA
| | | | | | | |
Collapse
|
197
|
Abstract
Alterations in PI3K signaling are common in gynecologic malignancies. Alterations detected vary with gynecologic cancer type, histologic subtypes within these, and clinical phenotypes. The distinction into type I and type II endometrial and ovarian carcinomas is reflected in distribution of changes detected in several of the PI3K members. PIK3CA mutations and amplifications are common in endometrial, ovarian, and cervical cancers. PTEN mutations and deletions are frequent in endometrial cancers. Several immunohistochemical studies of protein expression have explored these and other potential surrogate markers for PI3K pathway activation. Biomarkers to measure level of PI3K activity in clinical samples are not established. Whether amplifications, mutations, and deletions of the PI3K pathway members, and in particular change in their expression levels, result in clinically relevant pathway activation needs to be further explored. Also, to what extent these alterations drive the tumor behavior and are critical targets for therapeutics to improve patient survival needs to be further tested to establish predictive biomarkers for response to PI3K inhibition.
Collapse
Affiliation(s)
- Helga B Salvesen
- From the Department of Gynecology and Obstetrics, Haukeland University Hospital, Bergen, Norway; Institute of Clinical Sciences, University of Bergen, Bergen, Norway
| | | | | |
Collapse
|
198
|
Chen X, Ishwaran H. Pathway hunting by random survival forests. Bioinformatics 2013; 29:99-105. [PMID: 23129299 PMCID: PMC3530909 DOI: 10.1093/bioinformatics/bts643] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2012] [Revised: 07/18/2012] [Accepted: 10/17/2012] [Indexed: 01/22/2023] Open
Abstract
MOTIVATION Pathway or gene set analysis has been widely applied to genomic data. Many current pathway testing methods use univariate test statistics calculated from individual genomic markers, which ignores the correlations and interactions between candidate markers. Random forests-based pathway analysis is a promising approach for incorporating complex correlation and interaction patterns, but one limitation of previous approaches is that pathways have been considered separately, thus pathway cross-talk information was not considered. RESULTS In this article, we develop a new pathway hunting algorithm for survival outcomes using random survival forests, which prioritize important pathways by accounting for gene correlation and genomic interactions. We show that the proposed method performs favourably compared with five popular pathway testing methods using both synthetic and real data. We find that the proposed methodology provides an efficient and powerful pathway modelling framework for high-dimensional genomic data. AVAILABILITY The R code for the analysis used in this article is available upon request.
Collapse
Affiliation(s)
- Xi Chen
- Department of Biostatistics, Vanderbilt University School of Medicine, Nashville, TN 37232, USA.
| | | |
Collapse
|
199
|
Jia L, Zhang S, Ye Y, Li X, Mercado-Uribe I, Bast RC, Liu J. Paclitaxel inhibits ovarian tumor growth by inducing epithelial cancer cells to benign fibroblast-like cells. Cancer Lett 2012; 326:176-82. [PMID: 22902993 PMCID: PMC3495569 DOI: 10.1016/j.canlet.2012.08.004] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2012] [Revised: 08/03/2012] [Accepted: 08/07/2012] [Indexed: 11/25/2022]
Abstract
Paclitaxel is commonly used to treat multiple human malignancies, but its mechanism of action is still poorly defined. Human ovarian cancer SKOV3 cells (parental SKOV3) were treated with paclitaxel (1μM) for 2days, and the morphologic changes in the cells were monitored for more than 4months. Parental SKOV3 underwent a markedly morphologic transition from the epithelial to fibroblast-like phenotype following treatment with paclitaxel; the resulting cells were designated as SKOV3-P. The SKOV3-P cells' proliferative ability was assessed via a 3-[4,5-dimethylthiazol-2-yl]-2,5 diphenyl tetrazolium bromide (MTT) assay. The molecular characteristics of these cells were assessed via immunocytochemical staining and Western blot analysis. Their invasiveness and tumor formation ability was evaluated via wound-scratch and colony formation assays. The tumorigenicity of SKOV3-P cells was assessed in vivo after subcutaneous injection of tumor cells between injections of parental and paclitaxel-treated cells in nude mice. SKOV3-P cells have decreased the proliferation and invasion ability, decreased colony-forming ability when cultured in Matrigel and lost their tumor formation as compared with parental SKOV3 cells when injected in nude mice. SKOV3-P cells have decreased expression of E-cadherin, cytokeratin, Snail, PI3K, and P-Akt-Ser473, and increased expression of fibronectin, vimentin, Slug, P27, and PTEN. These results demonstrated that paclitaxel can inhibit tumor growth by inducing ovarian cancer epithelial cells toward a benign fibroblast-like phenotype through dysregulation of previously known pathways involved in the regulation of epithelial to mesenchymal transition (EMT), which may represent a novel mechanism for paclitaxel-induced tumor suppression.
Collapse
Affiliation(s)
- Lizhou Jia
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Obstetrics and Gynecology, The Affiliated People’s Hospital of Inner Mongolia Medical College, Inner Mongolia Autonomous Region, China
| | - Shiwu Zhang
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Yanfen Ye
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Cancer Research Institute, South Medical University, Guangzhou, Guangdong, China
| | - Xin Li
- Cancer Research Institute, South Medical University, Guangzhou, Guangdong, China
| | - Imelda Mercado-Uribe
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Robert C. Bast
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jinsong Liu
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
200
|
Kodigepalli KM, Dutta PS, Bauckman KA, Nanjundan M. SnoN/SkiL expression is modulated via arsenic trioxide-induced activation of the PI3K/AKT pathway in ovarian cancer cells. FEBS Lett 2012. [PMID: 23178716 DOI: 10.1016/j.febslet.2012.11.003] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
SnoN/SkiL (TGFβ regulator) is dysregulated in ovarian cancer, a disease associated with acquired drug-resistance. Arsenic trioxide (As₂O₃, used in treating APL) induces SnoN to oppose the apoptotic response in ovarian cancer cells. We now report that As₂O₃ increases phosphorylation of EGFR/p66ShcA and EGFR degradation. As₂O₃ activates Src(Y416) whose activity (inhibited by PP2) modulates EGFR activation, its interaction with Shc/Grb2, and p-AKT. Inhibition of PI3K reduces SnoN and cell survival. Although EGFR or MAPK1 siRNA did not alter SnoN expression, As₂O₃-induced cleaved PARP was reduced together with increased XIAP. Collectively, As₂O₃ mediates an initial rise in pY-Src(416) to regulate the PI3K/AKT pathway which increases SnoN and cell survival; these early events may counter the cell death response associated with increased pY-EGFR/MAPK activation.
Collapse
Affiliation(s)
- Karthik M Kodigepalli
- University of South Florida, Department of Cell Biology, Microbiology, and Molecular Biology, 4202 East Fowler Avenue, ISA2015, Tampa, FL, United States
| | | | | | | |
Collapse
|