51
|
Lee S, Zhao L, Rojas C, Bateman NW, Yao H, Lara OD, Celestino J, Morgan MB, Nguyen TV, Conrads KA, Rangel KM, Dood RL, Hajek RA, Fawcett GL, Chu RA, Wilson K, Loffredo JL, Viollet C, Jazaeri AA, Dalgard CL, Mao X, Song X, Zhou M, Hood BL, Banskota N, Wilkerson MD, Te J, Soltis AR, Roman K, Dunn A, Cordover D, Eterovic AK, Liu J, Burks JK, Baggerly KA, Fleming ND, Lu KH, Westin SN, Coleman RL, Mills GB, Casablanca Y, Zhang J, Conrads TP, Maxwell GL, Futreal PA, Sood AK. Molecular Analysis of Clinically Defined Subsets of High-Grade Serous Ovarian Cancer. Cell Rep 2020; 31:107502. [PMID: 32294438 PMCID: PMC7234854 DOI: 10.1016/j.celrep.2020.03.066] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Revised: 01/02/2020] [Accepted: 03/19/2020] [Indexed: 12/30/2022] Open
Abstract
The diversity and heterogeneity within high-grade serous ovarian cancer (HGSC), which is the most lethal gynecologic malignancy, is not well understood. Here, we perform comprehensive multi-platform omics analyses, including integrated analysis, and immune monitoring on primary and metastatic sites from highly clinically annotated HGSC samples based on a laparoscopic triage algorithm from patients who underwent complete gross resection (R0) or received neoadjuvant chemotherapy (NACT) with excellent or poor response. We identify significant distinct molecular abnormalities and cellular changes and immune cell repertoire alterations between the groups, including a higher rate of NF1 copy number loss, and reduced chromothripsis-like patterns, higher levels of strong-binding neoantigens, and a higher number of infiltrated T cells in the R0 versus the NACT groups.
Collapse
Affiliation(s)
- Sanghoon Lee
- Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Li Zhao
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Christine Rojas
- Women's Health Integrated Research Center at Inova Health System, Women's Service Line, Inova Fairfax Medical Campus, Falls Church, VA, USA; Gynecologic Cancer Center of Excellence, Department of Obstetrics and Gynecology, Uniformed Services University and Walter Reed National Military Medical Center, Bethesda, MD, USA
| | - Nicholas W Bateman
- Women's Health Integrated Research Center at Inova Health System, Women's Service Line, Inova Fairfax Medical Campus, Falls Church, VA, USA; Gynecologic Cancer Center of Excellence, Department of Obstetrics and Gynecology, Uniformed Services University and Walter Reed National Military Medical Center, Bethesda, MD, USA; The Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
| | - Hui Yao
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Olivia D Lara
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Joseph Celestino
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Margaret B Morgan
- Institute for Personalized Cancer Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Tri V Nguyen
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Kelly A Conrads
- Women's Health Integrated Research Center at Inova Health System, Women's Service Line, Inova Fairfax Medical Campus, Falls Church, VA, USA; Gynecologic Cancer Center of Excellence, Department of Obstetrics and Gynecology, Uniformed Services University and Walter Reed National Military Medical Center, Bethesda, MD, USA; The Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
| | - Kelly M Rangel
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Robert L Dood
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Richard A Hajek
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Gloria L Fawcett
- Institute for Personalized Cancer Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Randy A Chu
- Institute for Personalized Cancer Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Katlin Wilson
- Women's Health Integrated Research Center at Inova Health System, Women's Service Line, Inova Fairfax Medical Campus, Falls Church, VA, USA; Gynecologic Cancer Center of Excellence, Department of Obstetrics and Gynecology, Uniformed Services University and Walter Reed National Military Medical Center, Bethesda, MD, USA; The Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
| | - Jeremy L Loffredo
- Women's Health Integrated Research Center at Inova Health System, Women's Service Line, Inova Fairfax Medical Campus, Falls Church, VA, USA; Gynecologic Cancer Center of Excellence, Department of Obstetrics and Gynecology, Uniformed Services University and Walter Reed National Military Medical Center, Bethesda, MD, USA; The Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
| | - Coralie Viollet
- The American Genome Center, Collaborative Health Initiative Research Program, Uniformed Services University of the Health Sciences, Bethesda, MD, USA; The Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
| | - Amir A Jazaeri
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Clifton L Dalgard
- The American Genome Center, Collaborative Health Initiative Research Program, Uniformed Services University of the Health Sciences, Bethesda, MD, USA; Department of Anatomy, Physiology & Genetics, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Xizeng Mao
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Xingzhi Song
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ming Zhou
- Women's Health Integrated Research Center at Inova Health System, Women's Service Line, Inova Fairfax Medical Campus, Falls Church, VA, USA; Gynecologic Cancer Center of Excellence, Department of Obstetrics and Gynecology, Uniformed Services University and Walter Reed National Military Medical Center, Bethesda, MD, USA
| | - Brian L Hood
- Women's Health Integrated Research Center at Inova Health System, Women's Service Line, Inova Fairfax Medical Campus, Falls Church, VA, USA; Gynecologic Cancer Center of Excellence, Department of Obstetrics and Gynecology, Uniformed Services University and Walter Reed National Military Medical Center, Bethesda, MD, USA; The Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
| | - Nirad Banskota
- Women's Health Integrated Research Center at Inova Health System, Women's Service Line, Inova Fairfax Medical Campus, Falls Church, VA, USA; Gynecologic Cancer Center of Excellence, Department of Obstetrics and Gynecology, Uniformed Services University and Walter Reed National Military Medical Center, Bethesda, MD, USA; The Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
| | - Matthew D Wilkerson
- The American Genome Center, Collaborative Health Initiative Research Program, Uniformed Services University of the Health Sciences, Bethesda, MD, USA; Department of Anatomy, Physiology & Genetics, Uniformed Services University of the Health Sciences, Bethesda, MD, USA; The Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
| | - Jerez Te
- The American Genome Center, Collaborative Health Initiative Research Program, Uniformed Services University of the Health Sciences, Bethesda, MD, USA; Department of Anatomy, Physiology & Genetics, Uniformed Services University of the Health Sciences, Bethesda, MD, USA; The Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
| | - Anthony R Soltis
- The American Genome Center, Collaborative Health Initiative Research Program, Uniformed Services University of the Health Sciences, Bethesda, MD, USA; The Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
| | | | | | | | - Agda Karina Eterovic
- Institute for Personalized Cancer Therapy, 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
| | - Jared K Burks
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Keith A Baggerly
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Nicole D Fleming
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Karen H Lu
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Shannon N Westin
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Robert L Coleman
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Gordon B Mills
- Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Yovanni Casablanca
- Gynecologic Cancer Center of Excellence, Department of Obstetrics and Gynecology, Uniformed Services University and Walter Reed National Military Medical Center, Bethesda, MD, USA
| | - Jianhua Zhang
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Thomas P Conrads
- Women's Health Integrated Research Center at Inova Health System, Women's Service Line, Inova Fairfax Medical Campus, Falls Church, VA, USA; Gynecologic Cancer Center of Excellence, Department of Obstetrics and Gynecology, Uniformed Services University and Walter Reed National Military Medical Center, Bethesda, MD, USA
| | - George L Maxwell
- Women's Health Integrated Research Center at Inova Health System, Women's Service Line, Inova Fairfax Medical Campus, Falls Church, VA, USA; Gynecologic Cancer Center of Excellence, Department of Obstetrics and Gynecology, Uniformed Services University and Walter Reed National Military Medical Center, Bethesda, MD, USA
| | - P Andrew Futreal
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Anil K Sood
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
52
|
Schallenberg S, Bork J, Essakly A, Alakus H, Buettner R, Hillmer AM, Bruns C, Schroeder W, Zander T, Loeser H, Gebauer F, Quaas A. Loss of the SWI/SNF-ATPase subunit members SMARCF1 (ARID1A), SMARCA2 (BRM), SMARCA4 (BRG1) and SMARCB1 (INI1) in oesophageal adenocarcinoma. BMC Cancer 2020; 20:12. [PMID: 31906887 PMCID: PMC6945480 DOI: 10.1186/s12885-019-6425-3] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2019] [Accepted: 12/03/2019] [Indexed: 12/15/2022] Open
Abstract
Background The SWI/SNF complex is an important chromatin remodeler, commonly dysregulated in cancer, with an estimated mutation frequency of 20%. ARID1A is the most frequently mutated subunit gene. Almost nothing is known about the other familiar members of the SWI/SNF complexes, SMARCA2 (BRM), SMARCA4 (BRG1) and SMARCB1 (INI1), in oesophageal adenocarcinoma (EAC). Methods We analysed a large cohort of 685 patients with EAC. We used four different antibodies to detect a loss-of-protein of ARID1A BRM, BRG1 and INI1 by immunohistochemistry and correlated these findings with molecular and clinical data. Results Loss of ARID1A, BRG1, BRM and INI1 was observed in 10.4, 3.4, 9.9 and 2% of EAC. We found a co-existing protein loss of ARID1A and BRM in 9.9% and of ARID1A and BRG1 in 2.2%. Patients with loss of ARID1A and TP53 wildtype EACs showed a shortened overall survival compared with AIRDA1A-positive tumours [median overall survival was 60.1 months (95%CI 1.2–139.9 months)] in patients with ARIDA-1A expression and 26.2 months (95%CI 3.7–19.1 months) in cases of ARIDA-1A loss (p = 0.044). Tumours with loss or expression of ARID1A and TP53 loss were not associated with a difference in survival. Only one tumour revealed high microsatellite instability (MSI-H) with concomitant ARID1A loss. All other ARID1A loss-EACs were microsatellite-stable (MSS). No predictive relevance was seen for SWI/SNF-complex alterations and simultaneous amplification of different genes (PIK3CA, KRAS, c-MYC, MET, GATA6, ERBB2). Conclusion Our work describes, for the first time, loss of one of the SWI/SNF ATPase subunit proteins in a large number of adenocarcinomas of the oesophagus. Several papers discuss possible therapeutic interventions for tumours showing a loss of function of the SWI/SNF complex, such as PARP inhibitors or PI3K and AKT inhibitors. Future studies will be needed to show whether SWI/SNF complex-deficient EACs may benefit from personalized therapy.
Collapse
Affiliation(s)
- Simon Schallenberg
- Institute of Pathology, University of Cologne, Kerpener Str. 62, 50937, Cologne, Germany.
| | - Julian Bork
- Institute of Pathology, University of Cologne, Kerpener Str. 62, 50937, Cologne, Germany
| | - Ahlem Essakly
- Institute of Pathology, University of Cologne, Kerpener Str. 62, 50937, Cologne, Germany
| | - Hakan Alakus
- Department of General, Visceral and Cancer Surgery, University of Cologne, Cologne, Germany
| | - Reinhard Buettner
- Institute of Pathology, University of Cologne, Kerpener Str. 62, 50937, Cologne, Germany
| | - Axel M Hillmer
- Institute of Pathology, University of Cologne, Kerpener Str. 62, 50937, Cologne, Germany
| | - Christiane Bruns
- Department of General, Visceral and Cancer Surgery, University of Cologne, Cologne, Germany
| | - Wolfgang Schroeder
- Department of General, Visceral and Cancer Surgery, University of Cologne, Cologne, Germany
| | - Thomas Zander
- Department I of Internal Medicine, Center for Integrated Oncology (CIO)University of Cologne, Cologne, Germany
| | - Heike Loeser
- Institute of Pathology, University of Cologne, Kerpener Str. 62, 50937, Cologne, Germany
| | - Florian Gebauer
- Department of General, Visceral and Cancer Surgery, University of Cologne, Cologne, Germany
| | - Alexander Quaas
- Institute of Pathology, University of Cologne, Kerpener Str. 62, 50937, Cologne, Germany
| |
Collapse
|
53
|
Cao Q, Wang C, Ding Y, Xu D, Qian S, Shen H, Qi J. ARID1A upregulation predicts better survival in patients with urothelial bladder carcinoma. J Int Med Res 2020; 48:300060519895687. [PMID: 31891283 PMCID: PMC7783253 DOI: 10.1177/0300060519895687] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Objective AT-rich interactive domain-containing protein 1A (ARID1A) is frequently
mutated or deficient in various types of tumors. However, the role of ARID1A
in bladder cancer remains unclear. We aimed to evaluate ARID1A expression
and its biological role and correlation with prognosis in patients with
urothelial bladder carcinoma (BUC). Methods ARID1A expression levels in BUC and normal tissues were assessed by
immunohistochemistry and correlated with clinicopathological characteristics
and patient outcomes. Downregulation of ARID1A was mimicked by transfection
with small interfering RNA in T24 bladder cancer cells, and the effects on
cell proliferation and migration were evaluated. Results ARID1A expression was significantly reduced in BUC tissues and was
significantly associated with T stage and AJCC stage. Upregulation of ARID1A
predicted a better prognosis in BUC patients. ARID1A expression and lymph
node status were identified as independent prognostic factors for overall
survival. Silencing of ARID1A promoted the proliferation of
BUC cells. Conclusions ARID1A may represent a novel diagnostic and prognostic biomarker in patients
with BUC.
Collapse
Affiliation(s)
- Qifeng Cao
- Department of Urology, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chen Wang
- Department of Urology, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yu Ding
- Department of Urology, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ding Xu
- Department of Urology, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Subo Qian
- Department of Urology, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Haibo Shen
- Department of Urology, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jun Qi
- Department of Urology, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
54
|
Myers AP, Konstantinopoulos PA, Barry WT, Luo W, Broaddus RR, Makker V, Drapkin R, Liu J, Doyle A, Horowitz NS, Meric-Bernstam F, Birrer M, Aghajanian C, Coleman RL, Mills GB, Cantley LC, Matulonis UA, Westin SN. Phase II, 2-stage, 2-arm, PIK3CA mutation stratified trial of MK-2206 in recurrent endometrial cancer. Int J Cancer 2019; 147:413-422. [PMID: 31714586 DOI: 10.1002/ijc.32783] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 09/27/2019] [Accepted: 10/09/2019] [Indexed: 12/26/2022]
Abstract
Endometrial cancers have high rates of phosphoinositide 3-kinase (PI3K) pathway alterations. MK-2206 is an allosteric inhibitor of AKT, an effector kinase of PI3K signals. We hypothesized patients with tumors harboring PIK3CA mutations would be more likely to benefit from MK-2206 than those without PIK3CA mutation. A Phase II study was performed in patients with recurrent endometrial cancer; all histologies except carcinosarcoma were eligible. Up to two prior chemotherapy lines were permitted, excluding prior treatment with PI3K pathway inhibitors. The first 18 patients were treated with MK-2206 200 mg weekly. Due to unacceptable toxicity, dose was reduced to 135 mg. Co-primary endpoints were objective response rate (ORR) and progression-free survival at 6 months (6moPFS). Thirty-seven patients were enrolled (one ineligible). By somatic PIK3CA mutation analysis, nine patients were mutant (MT) [one with partial response (PR)/6moPFS, two with 6moPFS]. Twenty-seven patients were wild-type (WT) (one PR and four 6moPFS). Most common toxicities were rash (44%), fatigue (41%), nausea (42%) and hyperglycemia (31%). Grade 3 and 4 toxicities occurred in 25 and 17% of patients, respectively. Exploratory analysis found serous histology had greater 6moPFS as compared to all other histologies (5/8 vs. 2/28, p = 0.003). PTEN expression was associated with median time to progression (p = 0.04). No other significant associations with PI3K pathway alterations were identified. There is limited single agent activity of MK-2206 in PIK3CA MT and PIK3CA WT endometrial cancer populations. Activity was detected in patients with serous histology and due to their poor outcomes warrants further study (NCT01307631).
Collapse
Affiliation(s)
- Andrea P Myers
- Division of Hematology/Oncology, Department of Medical Oncology, Dana Farber Cancer Institute, Boston, MA
| | | | | | - Weixiu Luo
- Division of Hematology/Oncology, Department of Medical Oncology, Dana Farber Cancer Institute, Boston, MA
| | - Russell R Broaddus
- Department of Pathology, University of Texas M. D. Anderson Cancer Center, Houston, TX
| | - Vicky Makker
- Department of Medicine, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, NY
| | - Ronny Drapkin
- Department of Obstetrics and Gynecology, University of Pennsylvania, Philadelphia, PA
| | - Joyce Liu
- Division of Hematology/Oncology, Department of Medical Oncology, Dana Farber Cancer Institute, Boston, MA
| | - Austin Doyle
- Cancer Therapy Evaluation Program, National Cancer Institute, Bethesda, MD
| | - Neil S Horowitz
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Dana Farber Cancer Institute, Boston, MA
| | - Funda Meric-Bernstam
- Department of Investigational Cancer Therapeutics, University of Texas M. D. Anderson Cancer Center, Houston, TX
| | - Michael Birrer
- Division of Medical Oncology, Massachusetts General Hospital, Boston, MA
| | - Carol Aghajanian
- Department of Medicine, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, NY
| | - Robert L Coleman
- Department of Gynecologic Oncology and Reproductive Medicine, University of Texas M. D. Anderson Cancer Center, Houston, TX
| | - Gordon B Mills
- Department of Systems Biology, University of Texas M. D. Anderson Cancer Center, Houston, TX
| | - Lewis C Cantley
- Department of Medicine, Weill Cornell Medical College, New York, NY
| | - Ursula A Matulonis
- Division of Hematology/Oncology, Department of Medical Oncology, Dana Farber Cancer Institute, Boston, MA
| | - Shannon N Westin
- Department of Gynecologic Oncology and Reproductive Medicine, University of Texas M. D. Anderson Cancer Center, Houston, TX
| |
Collapse
|
55
|
Huang M, Xiao J, Nasca PC, Liu C, Lu Y, Lawrence WR, Wang L, Chen Q, Lin S. Do multiple environmental factors impact four cancers in women in the contiguous United States? ENVIRONMENTAL RESEARCH 2019; 179:108782. [PMID: 31634768 DOI: 10.1016/j.envres.2019.108782] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 09/27/2019] [Accepted: 09/27/2019] [Indexed: 06/10/2023]
Abstract
BACKGROUND Though there has been an increasing concern about the effects that environmental exposures have on cancer, limited knowledge exists regarding multiple environmental factors on cancers in women. METHODS We performed a spatial autoregressive model to examine the association between the Environmental Quality Index (EQI) and mortalities of four cancers in women (breast, cervical, ovarian and uterine cancer) based on county-level data, and explored these associations by urbanicity. The EQI, which included five domains (air, water, land, built environment and sociodemographic domain) estimated from 2000 to 2005 data, was obtained from the United States Environmental Protection Agency. The mortality rates for 3107 counties in the US in 2014 were obtained from the Institute for Health Metrics and Evaluation. RESULTS We found that each unit increase in the overall EQI was positively associated with the increased mortality of breast, ovarian and uterine cancer (2.5%, 3.6% and 3.1% respectively), but was negatively associated with cervical cancer mortality. Among the environmental domains, the air and sociodemographic EQIs were positively associated with increased risks of breast, ovarian and uterine cancers. Additionally, built environment EQI was associated with breast and ovarian cancers; land EQI was associated with uterine and ovarian cancers. The sociodemographic EQI was negatively associated with cervical cancer mortality. Furthermore, we have developed a novel Environmental Quality Health Index (EQHI) in identifying environment-health risk of cancers in women at county level. CONCLUSIONS Our findings suggest that breast, ovarian, and uterine cancer mortalities are positively associated with multiple environmental factors, while cervical cancer mortality is mainly negatively associated with sociodemographic factors. The novel EQHI might help identify spatially-based environment-cancer risk.
Collapse
Affiliation(s)
- Miaoling Huang
- Department of Obstetrics and Gynecology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
| | - Jianpeng Xiao
- Guangdong Provincial Institute of Public Health, Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, 511430, China
| | - Philip C Nasca
- Department of Epidemiology and Biostatistics, University at Albany, State University of New York, Rensselaer, NY, USA
| | - Changhao Liu
- Department of Obstetrics and Gynecology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
| | - Yi Lu
- Department of Environmental Health Sciences, University at Albany, State University of New York, Rensselaer, NY, USA
| | - Wayne R Lawrence
- Department of Epidemiology and Biostatistics, University at Albany, State University of New York, Rensselaer, NY, USA
| | - Lijuan Wang
- Department of Obstetrics and Gynecology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
| | - Qing Chen
- Department of Obstetrics and Gynecology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China.
| | - Shao Lin
- Department of Epidemiology and Biostatistics, University at Albany, State University of New York, Rensselaer, NY, USA; Department of Environmental Health Sciences, University at Albany, State University of New York, Rensselaer, NY, USA.
| |
Collapse
|
56
|
Liu X, Ren Z, Xu Y, Sun W, Li Y, Rui X, Xie D, Meng X, Zheng Z. Establishment and characterization of novel human primary endometrial cancer cell line (ZJB-ENC1) and its genomic characteristic. J Cancer 2019; 10:6466-6474. [PMID: 31772679 PMCID: PMC6856739 DOI: 10.7150/jca.33013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Accepted: 08/26/2019] [Indexed: 12/12/2022] Open
Abstract
The establishment of human malignant tumor cell lines can provide abundant experimental materials for understanding the biological characteristics of tumors, studying the carcinogenesis, molecular genetics and the mechanism of metastasis and evolution. In this study, a novel cell line designated ZJB-ENC1 has been established from poorly differentiated endometrioid adenocarcinoma. Cytological results showed monolayer-cultured cells were polygonal in shape and a piling-up tendency without contact inhabitation. Immunohistochemistry analysis showed that the cells were negative for ER, PR, c-erbB2, E-CAD, CD117, and OCT3/4, but strongly positive for PTEN and P16. Meanwhile, the tumorigenicity of ZJB-ENC1 was confirmed by subcutaneous transplantation of the cells into a xenograft mouse model. In addition, the results of the whole exome sequencing revealed a unique genomic characteristic of ZJB-ENC1 cells, all common and novel SNPs and InDels were identified. In conclusion, this new stable cell line may promote basic and clinical research on endometrial cancer (EC).
Collapse
Affiliation(s)
- Xiaozhen Liu
- Institute of Cancer and Basic Medicine (ICBM), Chinese Academy of Sciences, Hangzhou, 310022, China.,The Experimental Center, Cancer Hospital of the University of Chinese Academy of Sciences, Hangzhou, 310022, China.,The Experimental Center, Zhejiang cancer hospital , Hangzhou, 310022, China
| | - Zhuozhuo Ren
- Medical Support Department, Zhejiang Provincial Hospital of Traditional Chinese Medicine, Hangzhou 310022, China
| | - Yu Xu
- Department of Hematology, The First Affiliated Hospital, Zhejiang University, School of Medicine, Hangzhou, 310022, China
| | - Wei Sun
- Institute of Cancer and Basic Medicine (ICBM), Chinese Academy of Sciences, Hangzhou, 310022, China.,The Experimental Center, Cancer Hospital of the University of Chinese Academy of Sciences, Hangzhou, 310022, China.,The Experimental Center, Zhejiang cancer hospital , Hangzhou, 310022, China
| | - Yongfeng Li
- Institute of Cancer and Basic Medicine (ICBM), Chinese Academy of Sciences, Hangzhou, 310022, China.,Department of Breast Surgery, Cancer Hospital of the University of Chinese Academy of Sciences, Hangzhou, 310022, China.,Department of Breast Surgery, Zhejiang cancer hospital, Hangzhou, 310022, China
| | - Xinmiao Rui
- The Second Clinical Department, Zhejiang Chinese Medical University, Hangzhou, 310022, China
| | - Dafei Xie
- General Surgery Department, Zhejiang Hospital, Hangzhou, 310022, China
| | - Xuli Meng
- Department of Breast Thyroid Surgery, Tongde Hospital of Zhejiang Province, Hangzhou, 310022, China
| | - Zhiguo Zheng
- Institute of Cancer and Basic Medicine (ICBM), Chinese Academy of Sciences, Hangzhou, 310022, China.,The Experimental Center, Cancer Hospital of the University of Chinese Academy of Sciences, Hangzhou, 310022, China.,The Experimental Center, Zhejiang cancer hospital , Hangzhou, 310022, China
| |
Collapse
|
57
|
Westin SN, Sill MW, Coleman RL, Waggoner S, Moore KN, Mathews CA, Martin LP, Modesitt SC, Lee S, Ju Z, Mills GB, Schilder RJ, Fracasso PM, Birrer MJ, Aghajanian C. Safety lead-in of the MEK inhibitor trametinib in combination with GSK2141795, an AKT inhibitor, in patients with recurrent endometrial cancer: An NRG Oncology/GOG study. Gynecol Oncol 2019; 155:420-428. [PMID: 31623857 DOI: 10.1016/j.ygyno.2019.09.024] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Revised: 09/26/2019] [Accepted: 09/27/2019] [Indexed: 11/25/2022]
Abstract
OBJECTIVE We sought to determine safety and efficacy of the AKT inhibitor, GSK2141795, combined with the MEK inhibitor, trametinib, in endometrial cancer. METHODS Patients with measurable recurrent endometrial cancer were eligible. One to two prior cytotoxic regimens were allowed; prior use of a MEK or PI3K pathway inhibitor was excluded. Initial trial design consisted of a KRAS mutation stratified randomized phase II with a safety lead-in evaluating the combination. For the safety lead in, the previously recommended phase 2 dose (RP2D; trametinib 1.5 mg, GSK2141795 50 mg) was chosen for Dose Level 1 (DL1). RESULTS Of 26 enrolled patients, 14 were treated on DL1 and 12 were treated on DL-1 (trametinib 1.5 mg, GSK2141795 25 mg). Most common histologies were endometrioid (58%) and serous (27%). Four of 25 (16%) patients were KRAS mutant. Dose limiting toxicities (DLTs) were assessed during cycle 1. DL1 had 8 DLTs (hypertension (n = 2), mucositis (2), rash (2), dehydration, stroke/acute kidney injury). DL1 was deemed non-tolerable so DL-1 was explored. DL-1 had no DLTs. Sixty-five percent of patients had ≥ grade 3 toxicity. There were no responses in DL1 (0%, 90%CI 0-15%) and 1 response in DL-1 (8.3%, 90%CI 0.4-33.9%). Proportion PFS at 6 months for DL1 is 14%, and 25% for DL-1. CONCLUSION The combination of trametinib and GSK2141795 had high levels of toxicity in endometrial cancer at the previously RP2D but was tolerable at a reduced dose. Due to insufficient preliminary efficacy at a tolerable dose, the Phase II study was not initiated.
Collapse
Affiliation(s)
- Shannon N Westin
- Department of Gynecologic Oncology, University of Texas M. D Anderson Cancer Center, USA.
| | - Michael W Sill
- NRG Oncology Statistics and Data Management Center Buffalo Office, Roswell Park Cancer Institute, USA.
| | - Robert L Coleman
- Department of Gynecologic Oncology, University of Texas M. D Anderson Cancer Center, USA.
| | - Steven Waggoner
- Department of Gynecologic Oncology, Case Western Reserve University, USA.
| | - Kathleen N Moore
- Department of Gynecologic Oncology, University of Oklahoma Health Sciences Center, Stephenson Cancer Center, USA.
| | - Cara A Mathews
- Department of Gynecologic Oncology, Women & Infants Hospital, USA.
| | - Lainie P Martin
- Department of Hematology/Oncology, Fox Chase Cancer Center, USA.
| | - Susan C Modesitt
- Director of Gynecologic Oncology Division, University of Virginia, USA.
| | - Sanghoon Lee
- Department of Medicine and the UVA Cancer Center, University of Virginia, USA.
| | - Zhenlin Ju
- Department of Bioinformatics and Computational Biology, University of Texas M. D Anderson Cancer Center, USA.
| | - Gordon B Mills
- Department of Medicine and the UVA Cancer Center, University of Virginia, USA.
| | - Russell J Schilder
- Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University Hospital, USA.
| | - Paula M Fracasso
- Department of Systems Biology, University of Texas M.D Anderson Cancer Center, USA.
| | | | - Carol Aghajanian
- Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, USA.
| |
Collapse
|
58
|
Woolston A, Khan K, Spain G, Barber LJ, Griffiths B, Gonzalez-Exposito R, Hornsteiner L, Punta M, Patil Y, Newey A, Mansukhani S, Davies MN, Furness A, Sclafani F, Peckitt C, Jiménez M, Kouvelakis K, Ranftl R, Begum R, Rana I, Thomas J, Bryant A, Quezada S, Wotherspoon A, Khan N, Fotiadis N, Marafioti T, Powles T, Lise S, Calvo F, Guettler S, von Loga K, Rao S, Watkins D, Starling N, Chau I, Sadanandam A, Cunningham D, Gerlinger M. Genomic and Transcriptomic Determinants of Therapy Resistance and Immune Landscape Evolution during Anti-EGFR Treatment in Colorectal Cancer. Cancer Cell 2019; 36:35-50.e9. [PMID: 31287991 PMCID: PMC6617392 DOI: 10.1016/j.ccell.2019.05.013] [Citation(s) in RCA: 193] [Impact Index Per Article: 32.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Revised: 04/01/2019] [Accepted: 05/23/2019] [Indexed: 01/05/2023]
Abstract
Despite biomarker stratification, the anti-EGFR antibody cetuximab is only effective against a subgroup of colorectal cancers (CRCs). This genomic and transcriptomic analysis of the cetuximab resistance landscape in 35 RAS wild-type CRCs identified associations of NF1 and non-canonical RAS/RAF aberrations with primary resistance and validated transcriptomic CRC subtypes as non-genetic predictors of benefit. Sixty-four percent of biopsies with acquired resistance harbored no genetic resistance drivers. Most of these had switched from a cetuximab-sensitive transcriptomic subtype at baseline to a fibroblast- and growth factor-rich subtype at progression. Fibroblast-supernatant conferred cetuximab resistance in vitro, confirming a major role for non-genetic resistance through stromal remodeling. Cetuximab treatment increased cytotoxic immune infiltrates and PD-L1 and LAG3 immune checkpoint expression, potentially providing opportunities to treat cetuximab-resistant CRCs with immunotherapy.
Collapse
Affiliation(s)
- Andrew Woolston
- Translational Oncogenomics Lab, The Institute of Cancer Research, 237 Fulham Road, London SW3 6JB, UK
| | - Khurum Khan
- GI Cancer Unit, The Royal Marsden Hospital, London SW3 6JJ, UK
| | - Georgia Spain
- Translational Oncogenomics Lab, The Institute of Cancer Research, 237 Fulham Road, London SW3 6JB, UK
| | - Louise J Barber
- Translational Oncogenomics Lab, The Institute of Cancer Research, 237 Fulham Road, London SW3 6JB, UK
| | - Beatrice Griffiths
- Translational Oncogenomics Lab, The Institute of Cancer Research, 237 Fulham Road, London SW3 6JB, UK
| | - Reyes Gonzalez-Exposito
- Translational Oncogenomics Lab, The Institute of Cancer Research, 237 Fulham Road, London SW3 6JB, UK
| | - Lisa Hornsteiner
- Translational Oncogenomics Lab, The Institute of Cancer Research, 237 Fulham Road, London SW3 6JB, UK
| | - Marco Punta
- Centre for Evolution and Cancer Bioinformatics Team, The Institute of Cancer Research, London SW3 6JB, UK
| | - Yatish Patil
- Centre for Evolution and Cancer Bioinformatics Team, The Institute of Cancer Research, London SW3 6JB, UK
| | - Alice Newey
- Translational Oncogenomics Lab, The Institute of Cancer Research, 237 Fulham Road, London SW3 6JB, UK
| | - Sonia Mansukhani
- Translational Oncogenomics Lab, The Institute of Cancer Research, 237 Fulham Road, London SW3 6JB, UK
| | - Matthew N Davies
- Translational Oncogenomics Lab, The Institute of Cancer Research, 237 Fulham Road, London SW3 6JB, UK
| | - Andrew Furness
- Cancer Institute, University College London, London WC1E 6AG, UK
| | | | - Clare Peckitt
- GI Cancer Unit, The Royal Marsden Hospital, London SW3 6JJ, UK
| | - Mirta Jiménez
- Translational Oncogenomics Lab, The Institute of Cancer Research, 237 Fulham Road, London SW3 6JB, UK
| | | | - Romana Ranftl
- Tumour Microenvironment Lab, The Institute of Cancer Research, London SW3 6JB, UK
| | - Ruwaida Begum
- GI Cancer Unit, The Royal Marsden Hospital, London SW3 6JJ, UK
| | - Isma Rana
- GI Cancer Unit, The Royal Marsden Hospital, London SW3 6JJ, UK
| | - Janet Thomas
- GI Cancer Unit, The Royal Marsden Hospital, London SW3 6JJ, UK
| | - Annette Bryant
- GI Cancer Unit, The Royal Marsden Hospital, London SW3 6JJ, UK
| | - Sergio Quezada
- Cancer Institute, University College London, London WC1E 6AG, UK
| | | | - Nasir Khan
- Department of Radiology, The Royal Marsden Hospital, London SW3 6JJ, UK
| | - Nikolaos Fotiadis
- Department of Radiology, The Royal Marsden Hospital, London SW3 6JJ, UK
| | - Teresa Marafioti
- Departments of Pathology and Histopathology, University College Hospital, London NW1 2PG, UK
| | - Thomas Powles
- Barts Cancer Institute, Queen Mary University, London EC1M 6BQ, UK
| | - Stefano Lise
- Centre for Evolution and Cancer Bioinformatics Team, The Institute of Cancer Research, London SW3 6JB, UK
| | - Fernando Calvo
- Tumour Microenvironment Lab, The Institute of Cancer Research, London SW3 6JB, UK
| | - Sebastian Guettler
- Division of Structural Biology, The Institute of Cancer Research, London SW3 6JB, UK
| | - Katharina von Loga
- Translational Oncogenomics Lab, The Institute of Cancer Research, 237 Fulham Road, London SW3 6JB, UK
| | - Sheela Rao
- GI Cancer Unit, The Royal Marsden Hospital, London SW3 6JJ, UK
| | - David Watkins
- GI Cancer Unit, The Royal Marsden Hospital, London SW3 6JJ, UK
| | | | - Ian Chau
- GI Cancer Unit, The Royal Marsden Hospital, London SW3 6JJ, UK
| | - Anguraj Sadanandam
- Systems and Precision Cancer Medicine Lab, The Institute of Cancer Research, London SW3 6JB, UK
| | | | - Marco Gerlinger
- Translational Oncogenomics Lab, The Institute of Cancer Research, 237 Fulham Road, London SW3 6JB, UK; GI Cancer Unit, The Royal Marsden Hospital, London SW3 6JJ, UK.
| |
Collapse
|
59
|
Miller JE, Metpally RP, Person TN, Krishnamurthy S, Dasari VR, Shivakumar M, Lavage DR, Cook AM, Carey DJ, Ritchie MD, Kim D, Gogoi R. Systematic characterization of germline variants from the DiscovEHR study endometrial carcinoma population. BMC Med Genomics 2019; 12:59. [PMID: 31053132 PMCID: PMC6499978 DOI: 10.1186/s12920-019-0504-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Accepted: 04/15/2019] [Indexed: 02/02/2023] Open
Abstract
Background Endometrial cancer (EMCA) is the fifth most common cancer among women in the world. Identification of potentially pathogenic germline variants from individuals with EMCA will help characterize genetic features that underlie the disease and potentially predispose individuals to its pathogenesis. Methods The Geisinger Health System’s (GHS) DiscovEHR cohort includes exome sequencing on over 50,000 consenting patients, 297 of whom have evidence of an EMCA diagnosis in their electronic health record. Here, rare variants were annotated as potentially pathogenic. Results Eight genes were identified as having increased burden in the EMCA cohort relative to the non-cancer control cohort. None of the eight genes had an increased burden in the other hormone related cancer cohort from GHS, suggesting they can help characterize the underlying genetic variation that gives rise to EMCA. Comparing GHS to the cancer genome atlas (TCGA) EMCA germline data illustrated 34 genes with potentially pathogenic variation and eight unique potentially pathogenic variants that were present in both studies. Thus, similar germline variation among genes can be observed in unique EMCA cohorts and could help prioritize genes to investigate for future work. Conclusion In summary, this systematic characterization of potentially pathogenic germline variants describes the genetic underpinnings of EMCA through the use of data from a single hospital system. Electronic supplementary material The online version of this article (10.1186/s12920-019-0504-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jason E Miller
- Department of Genetics, Institute for Biomedical Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Raghu P Metpally
- Biomedical & Translational Informatics Institute, Geisinger Health System, Danville, PA, 17822, USA
| | - Thomas N Person
- Biomedical & Translational Informatics Institute, Geisinger Health System, Danville, PA, 17822, USA
| | | | | | - Manu Shivakumar
- Biomedical & Translational Informatics Institute, Geisinger Health System, Danville, PA, 17822, USA
| | - Daniel R Lavage
- Biomedical & Translational Informatics Institute, Geisinger Health System, Danville, PA, 17822, USA
| | - Adam M Cook
- Weis Center for Research, Geisinger Medical Center, Danville, PA, 17822, USA
| | - David J Carey
- Weis Center for Research, Geisinger Medical Center, Danville, PA, 17822, USA
| | - Marylyn D Ritchie
- Department of Genetics, Institute for Biomedical Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Dokyoon Kim
- Biomedical & Translational Informatics Institute, Geisinger Health System, Danville, PA, 17822, USA.,Huck Institute of the Life Sciences, Pennsylvania State University, University Park, PA, 16802, USA.,Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA.,Institute for Biomedical Informatics, University of Pennsylvania, Philadelphia, USA
| | - Radhika Gogoi
- Weis Center for Research, Geisinger Medical Center, Danville, PA, 17822, USA.
| | | |
Collapse
|
60
|
Toumpeki C, Liberis A, Tsirkas I, Tsirka T, Kalagasidou S, Inagamova L, Anthoulaki X, Tsatsaris G, Kontomanolis EN. The Role of ARID1A in Endometrial Cancer and the Molecular Pathways Associated With Pathogenesis and Cancer Progression. In Vivo 2019; 33:659-667. [PMID: 31028182 PMCID: PMC6559907 DOI: 10.21873/invivo.11524] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Revised: 03/26/2019] [Accepted: 04/01/2019] [Indexed: 02/07/2023]
Abstract
AT-rich interaction domain 1A gene (ARID1A) encodes for a subunit of the switch/sucrose non-fermentable (SWI/SNF) complex, a chromatin remodeling complex, and it has been implicated in the pathogenesis of various cancer types. In this review, we discuss how ARID1A is linked to endometrial cancer and what molecular pathways are affected by mutation or inhibition of ARID1A. We also discuss the potential use of ARID1A not only as a prognostic biomarker, but also as a target for therapeutic interventions.
Collapse
Affiliation(s)
- Chrisavgi Toumpeki
- Department of Obstetrics and Gynecology, Democritus University of Thrace, Alexandroupolis, Greece
| | - Anastasios Liberis
- Second Department of Obstetrics and Gynecology, Hippokration General Hospital, Thessaloniki, Greece
| | - Ioannis Tsirkas
- Department of Obstetrics and Gynecology, Democritus University of Thrace, Alexandroupolis, Greece
| | - Theodora Tsirka
- Department of Molecular Biology and Genetics, University of Thrace, Alexandroupolis, Greece
| | - Sofia Kalagasidou
- Department of Obstetrics and Gynecology, Bodosakio General Hospital of Ptolemaida, Ptolemaida, Greece
| | - Lola Inagamova
- Department of Obstetrics and Gynecology, Democritus University of Thrace, Alexandroupolis, Greece
| | - Xanthoula Anthoulaki
- Department of Obstetrics and Gynecology, Democritus University of Thrace, Alexandroupolis, Greece
| | - Georgios Tsatsaris
- Department of Obstetrics and Gynecology, Democritus University of Thrace, Alexandroupolis, Greece
| | - Emmanuel N Kontomanolis
- Department of Obstetrics and Gynecology, Democritus University of Thrace, Alexandroupolis, Greece
| |
Collapse
|
61
|
Banerjee S, Akbani R, Baladandayuthapani V. Spectral Clustering via sparse graph structure learning with application to Proteomic Signaling Networks in Cancer. Comput Stat Data Anal 2019; 132:46-69. [PMID: 38774121 PMCID: PMC11106846 DOI: 10.1016/j.csda.2018.08.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Clustering methods for multivariate data exploiting the underlying geometry of the graphical structure between variables are presented. As opposed to standard approaches for graph clustering that assume known graph structures, the edge structure of the unknown graph is first estimated using sparse regression based approaches for sparse graph structure learning. Subsequently, graph clustering on the lower dimensional projections of the graph is performed based on Laplacian embeddings using a penalized k-means approach, motivated by Dirichlet process mixture models in Bayesian nonparametrics. In contrast to standard algorithmic approaches for known graphs, the proposed method allows estimation and inference for both graph structure learning and clustering. More importantly, the arguments for Laplacian embeddings as suitable projections for graph clustering are formalized by providing theoretical support for the consistency of the eigenspace of the estimated graph Laplacians. Fast computational algorithms are proposed to scale the method to large number of nodes. Extensive simulations are presented to compare the clustering performance with standard methods. The methods are applied to a novel pan-cancer proteomic data set, and protein networks and clusters are evaluated across multiple different cancer types.
Collapse
Affiliation(s)
- Sayantan Banerjee
- Operations Management & Quantitative Techniques Area, Indian Institute of ManagementIndore, Indore, India
| | - Rehan Akbani
- Dept of Bioinformatics & Computational Biology, UT MD Anderson Cancer Center,Houston, Texas, USA
| | | |
Collapse
|
62
|
Kopetz S, Mills Shaw KR, Lee JJ, Zhang J, Litzenburger B, Holla V, Kinyua W, Broaddus E, Daniels MS, Meric-Bernstam F, Broaddus RR. Use of a Targeted Exome Next-Generation Sequencing Panel Offers Therapeutic Opportunity and Clinical Benefit in a Subset of Patients With Advanced Cancers. JCO Precis Oncol 2019; 3:1800213. [PMID: 32914008 PMCID: PMC7446317 DOI: 10.1200/po.18.00213] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/13/2018] [Indexed: 12/12/2022] Open
Abstract
PURPOSE Smaller hotspot-based next-generation sequencing (NGS) panels have emerged to support standard of care therapy for patients with cancer. When standard treatments fail, it is unknown whether additional testing using an expanded panel of genes provides any benefit. The purpose of this study was to determine if larger sequencing panels that capture additional actionable genes, coupled with decision support, translates into treatment with matched therapy after frontline therapy has failed. PATIENTS AND METHODS A prospective protocol accrued 521 patients with a wide variety of refractory cancers. NGS testing using a 46- or 50-gene hotspot assay, then a 409-gene whole-exome assay, was sequentially performed in a Clinical Laboratory Improvement Amendments–certified clinical laboratory. A decision-support team annotated somatic alterations in clinically actionable genes for function and facilitated therapeutic matching. Survival and the impact of matched therapy use were determined by Kaplan-Meier estimate, log-rank test, and Cox proportional hazards regression. RESULTS The larger NGS panel identified at least one alteration in an actionable gene not previously identified in the smaller sequencing panel in 214 (41%) of 521 of enrolled patients. After the application of decision support, 41% of the alterations in actionable genes were considered to affect the function of the gene and were deemed actionable. Forty patients (40 of 214 [19%]) were subsequently treated with matched therapy. Treatment with matched therapy was associated with significantly improved overall survival compared with treatment with nonmatched therapy (P = .017). CONCLUSION Combining decision support with larger NGS panels that incorporate genes beyond those recommended in current treatment guidelines helped to identify patients who were eligible for matched therapy while improving overall treatment selection and survival. This survival benefit was restricted to a small subset of patients.
Collapse
Affiliation(s)
- Scott Kopetz
- University of Texas MD Anderson Cancer Center, Houston, TX
| | | | - J Jack Lee
- University of Texas MD Anderson Cancer Center, Houston, TX
| | - Jiexin Zhang
- University of Texas MD Anderson Cancer Center, Houston, TX
| | | | | | - Walter Kinyua
- University of Texas MD Anderson Cancer Center, Houston, TX
| | - Emily Broaddus
- University of Texas MD Anderson Cancer Center, Houston, TX
| | | | | | | |
Collapse
|
63
|
Liu Y, Xu S, Lian X, Su Y, Zhong Y, Lv R, Mo K, Zhu H, Xiaojiang W, Xu L, Wang S. Atypical GATA protein TRPS1 plays indispensable roles in mouse two-cell embryo. Cell Cycle 2019; 18:437-451. [PMID: 30712485 DOI: 10.1080/15384101.2019.1577650] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Zygotic genome activation (ZGA) is one of the most critical events at the beginning of mammalian preimplantation embryo development (PED). The mechanisms underlying mouse ZGA remain unclear although it has been widely studied. In the present study, we identified that tricho-rhino-phalangeal syndrome 1 (TRPS1), an atypical GATA family member, is an important factor for ZGA in mouse PED. We found that the Trps1 mRNA level peaked at the one-cell stage while TRPS1 protein did so at the two/four-cell stage. Knockdown of Trps1 by the microinjection of Trps1 siRNA reduced the developmental rate of mouse preimplantation embryos by approximately 30%, and increased the expression of ZGA marker genes MuERV-L and Zscan4d via suppressing the expression of major histone markers H3K4me3 and H3K27me3. Furthermore, Trps1 knockdown decreased the expression of Sox2 but increased Oct4 expression. We conclude that TRPS1 may be indispensable for zygotic genome activation during mouse PED.
Collapse
Affiliation(s)
- Yue Liu
- a Key Laboratory of Stem Cell Engineering and Regenerative Medicine , Fujian Province University
| | - Songhua Xu
- b Department of Human Anatomy, Histology and Embryology , Fujian Medical University , Fuzhou , P. R. China
| | - Xiuli Lian
- b Department of Human Anatomy, Histology and Embryology , Fujian Medical University , Fuzhou , P. R. China
| | - Yang Su
- b Department of Human Anatomy, Histology and Embryology , Fujian Medical University , Fuzhou , P. R. China
| | - Yuhuan Zhong
- b Department of Human Anatomy, Histology and Embryology , Fujian Medical University , Fuzhou , P. R. China
| | - Ruimin Lv
- b Department of Human Anatomy, Histology and Embryology , Fujian Medical University , Fuzhou , P. R. China
| | - Kaien Mo
- b Department of Human Anatomy, Histology and Embryology , Fujian Medical University , Fuzhou , P. R. China
| | - Huimin Zhu
- c Fujian Key Laboratory of Medical Bioinformatics, School of Basic Medical Sciences , Fujian Medical University , Fuzhou , P. R. China.,d Key Laboratory of Ministry of Education for Gastrointestinal Cancer, School of Basic Medical Sciences , Fujian Medical University , Fuzhou , P. R. China
| | - Wang Xiaojiang
- b Department of Human Anatomy, Histology and Embryology , Fujian Medical University , Fuzhou , P. R. China
| | - Lixuan Xu
- b Department of Human Anatomy, Histology and Embryology , Fujian Medical University , Fuzhou , P. R. China
| | - Shie Wang
- a Key Laboratory of Stem Cell Engineering and Regenerative Medicine , Fujian Province University.,b Department of Human Anatomy, Histology and Embryology , Fujian Medical University , Fuzhou , P. R. China
| |
Collapse
|
64
|
Liu L, Yang L, Chang H, Chen YN, Zhang F, Feng S, Peng J, Ren CC, Zhang XA. CP‑31398 attenuates endometrial cancer cell invasion, metastasis and resistance to apoptosis by downregulating MDM2 expression. Int J Oncol 2019; 54:942-954. [PMID: 30628640 PMCID: PMC6365028 DOI: 10.3892/ijo.2019.4681] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Accepted: 10/26/2018] [Indexed: 12/21/2022] Open
Abstract
Endometrial cancer (EC) is one of the most common malignancies of the female reproductive system, and metastasis is a major cause of mortality. In this study, we aimed to explore the role of CP-31398 in the migration, invasion and apoptosis of EC cells by its regulation of the expression of the murine double minute 2 (MDM2) gene. For this purpose, EC tissues and adjacent normal tissues were collected, and the positive expression rate of MDM2 in these tissues was assessed. Subsequently, the cellular 50% inhibitory concentration (IC50) of CP-31398 was measured. The EC RL95-2 and KLE cell lines had a higher MDM2 expression and were thus selected for use in subsequent experiments. The EC cells were then treated with CP-31398 (2 µg/ml), and were transfected with siRNA against MDM2 or an MDM2 overexpression plasmid in order to examine the effects of CP-31398 and MDM2 on EC cell activities. The expression of p53, p21, Bad, Bax, B-cell lymphoma-2 (Bcl-2), cytochrome c (Cyt-c), caspase-3, Cox-2, matrix metalloproteinase (MMP)-2 and MMP-9 was measured to further confirm the effects of CP-31398 on cell migration, invasion and apoptosis. Our results indicated that MDM2 was highly expressed in EC tissues. Notably, EC cell viability decreased with the increasing concentrations of CP-31398. The EC cells treated with CP-31398 or siRNA against MDM2 exhibited an increased apoptosis and a suppressed migration and invasion, corresponding to an increased expression of p53, p21, Bad, Bax, Cyt-c and caspase-3, as well as to a decreased expression of Bcl-2, Cox-2, MMP-2 and MMP-9. Moreover, treatment with CP-31398 and siRNA against MDM2 further enhanced these effects. Taken together, the findings of this study indicate that the CP-31398-mediated downregulation of MDM2 may suppress EC progression via its inhibitory role in EC cell migration, invasion and resistance to apoptosis. Therefore, treatment with CP-31398 may prove to be possible therapeutic strategy for EC.
Collapse
Affiliation(s)
- Ling Liu
- Department of Gynecologic Oncology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Li Yang
- Department of Gynecologic Oncology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Hui Chang
- Laboratory of Tumor Precision Medicine, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Yan-Nan Chen
- Department of Gynecologic Oncology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Feng Zhang
- Department of Gynecologic Oncology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Shuo Feng
- Department of Gynecologic Oncology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Juan Peng
- Department of Gynecologic Oncology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Chen-Chen Ren
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Xiao-An Zhang
- Department of Imaging, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| |
Collapse
|
65
|
Hong KH, Song S, Shin W, Kang K, Cho CS, Hong YT, Han K, Moon JH. A case of interdigitating dendritic cell sarcoma studied by whole-exome sequencing. Genes Genomics 2018; 40:1279-1285. [PMID: 30099721 DOI: 10.1007/s13258-018-0724-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Accepted: 07/24/2018] [Indexed: 12/18/2022]
Abstract
Interdigitating dendritic cell sarcoma (IDCS) is an aggressive neoplasm and is an extremely rare disease, with a challenging diagnosis. Etiology of IDCS is also unknown and most studies with only case reports. In our case, immunohistochemistry showed that the tumor cells were positive for S100, CD45, and CD68, but negative for CD1a and CD21. This study aimed to investigate the causative factors of IDCS by sequencing the protein-coding regions of IDCS. We performed whole-exome sequencing with genomic DNA from blood and sarcoma tissue of the IDCS patient using the Illumina Hiseq 2500 platform. After that, we conducted Sanger sequencing for validation of sarcoma-specific variants and gene ontology analysis using DAVID bioinformatics resources. Through comparing sequencing data of sarcoma with normal blood, we obtained 15 nonsynonymous single nucleotide polymorphisms (SNPs) as sarcoma-specific variants. Although the 15 SNPs were not validated by Sanger sequencing due to tumor heterogeneity and low sensitivity of Sanger sequencing, we examined the function of the genes in which each SNP is located. Based on previous studies and gene ontology database, we found that POLQ encoding DNA polymerase theta enzyme and FNIP1 encoding tumor suppressor folliculin-interacting protein might have contributed to the IDCS. Our study provides potential causative genetic factors of IDCS and plays a role in advancing the understanding of IDCS pathogenesis.
Collapse
Affiliation(s)
- Ki Hwan Hong
- Department of Otolaryngology-Head and Neck Surgery, Chonbuk National University Medical School, Jeonju, 54896, Republic of Korea
| | - Soyoung Song
- Department of Nanobiomedical Science & BK21 PLUS NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, Republic of Korea
| | - Wonseok Shin
- Department of Nanobiomedical Science & BK21 PLUS NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, Republic of Korea
| | - Keunsoo Kang
- Department of Microbiology, Dankook University, Cheonan, 31116, Republic of Korea
| | - Chun-Sung Cho
- Department of Neurosurgery, College of Medicine, Dankook University, Cheonan, 31116, Republic of Korea
| | - Yong Tae Hong
- Department of Otolaryngology-Head and Neck Surgery, Chonbuk National University Medical School, Jeonju, 54896, Republic of Korea
| | - Kyudong Han
- Department of Nanobiomedical Science & BK21 PLUS NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, Republic of Korea.
| | - Jeong Hwan Moon
- Department of Otorhinolaryngology-Head and Neck Surgery, College of Medicine, Dankook University, Cheonan, 31116, Republic of Korea.
| |
Collapse
|
66
|
Libera L, Craparotta I, Sahnane N, Chiaravalli AM, Mannarino L, Cerutti R, Riva C, Marchini S, Furlan D. Targeted gene sequencing of Lynch syndrome–related and sporadic endometrial carcinomas. Hum Pathol 2018; 81:235-244. [DOI: 10.1016/j.humpath.2018.06.029] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Revised: 06/05/2018] [Accepted: 06/15/2018] [Indexed: 01/21/2023]
|
67
|
Zhang P, Kang B, Xie G, Li S, Gu Y, Shen Y, Zhao X, Ma Y, Li F, Si J, Wang J, Chen J, Yang H, Xu X, Yang Y. Genomic sequencing and editing revealed the GRM8 signaling pathway as potential therapeutic targets of squamous cell lung cancer. Cancer Lett 2018; 442:53-67. [PMID: 30391781 DOI: 10.1016/j.canlet.2018.10.035] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Revised: 10/18/2018] [Accepted: 10/24/2018] [Indexed: 02/06/2023]
Abstract
The study sought to explore novel genetic aberration driving squamous cell lung carcinoma (LUSC). The whole exome (WES), whole genome (WGS) and target region (TS) sequencings and CRISPR-Cas9 genome editing techniques were integrated to explore and validate novel targeting candidates from LUSC primary tumors and corresponding patient-derived xenografts (PDXs). Seven genes (FGFR2, GRM1,PIK3CG, PIK3CA,ZFHX4, CSMD3, GRM8) with high frequencies of both single nucleotide variants (SNVs) and copy number variants (CNVs), and two genes (CLDN1 and RIT1) only with CNVs were identified by bioinformatics analysis. The functions of these candidates were validated through CRISPR-Cas9 system in primary PDX cells. Furthermore, we focused on the genetic and functional analysis of Metabotropic glutamate receptor 8 (GRM8), whose transcriptional activation was elucidated to promote the survival of LUSC tumor cell through inhibiting cAMP pathway and activating MAPK pathway. The SNV identified in GRM8, A112G, activated downstream signaling pathway and induced cell proliferation, which could be reversed by cAMP stimulator and MEK inhibitor. In conclusion, the components of GRM8 signaling pathway could serve as potential targets of squamous cell lung cancer carrying GRM8 activating variants.
Collapse
MESH Headings
- Animals
- Antineoplastic Agents/therapeutic use
- CRISPR-Associated Protein 9/genetics
- CRISPR-Associated Protein 9/metabolism
- CRISPR-Cas Systems
- Carcinoma, Squamous Cell/drug therapy
- Carcinoma, Squamous Cell/genetics
- Carcinoma, Squamous Cell/metabolism
- Carcinoma, Squamous Cell/pathology
- Cell Line, Tumor
- Cell Proliferation
- Cell Survival
- Clustered Regularly Interspaced Short Palindromic Repeats
- Cyclic AMP/metabolism
- Gene Editing/methods
- Gene Expression Regulation, Neoplastic
- Humans
- Lung Neoplasms/drug therapy
- Lung Neoplasms/genetics
- Lung Neoplasms/metabolism
- Lung Neoplasms/pathology
- Mice, Inbred NOD
- Mice, SCID
- Mitogen-Activated Protein Kinases/metabolism
- Molecular Targeted Therapy
- Mutation
- Receptors, Metabotropic Glutamate/genetics
- Receptors, Metabotropic Glutamate/metabolism
- Signal Transduction
- Transcriptional Activation
- Tumor Burden
- Tumor Cells, Cultured
- Exome Sequencing
Collapse
Affiliation(s)
- Panpan Zhang
- Department of Thoracic Surgery II, Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education/Beijing, Peking University Cancer Hospital and Institute, Beijing, China
| | - Bin Kang
- BGI-Shenzhen, Beishan Industrial Zone, Shenzhen, 518083, China; China National GeneBank, BGI-Shenzhen, Jinsha Road, Shenzhen, 518120, China; Guangdong Provincial Key Laboratory of Genome Read and Write, Jinsha Road, Shenzhen, 518120, China
| | - Guoyun Xie
- BGI-Shenzhen, Beishan Industrial Zone, Shenzhen, 518083, China; China National GeneBank, BGI-Shenzhen, Jinsha Road, Shenzhen, 518120, China
| | - Shaolei Li
- Department of Thoracic Surgery II, Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education/Beijing, Peking University Cancer Hospital and Institute, Beijing, China
| | - Ying Gu
- BGI-Shenzhen, Beishan Industrial Zone, Shenzhen, 518083, China; China National GeneBank, BGI-Shenzhen, Jinsha Road, Shenzhen, 518120, China; Guangdong Provincial Key Laboratory of Genome Read and Write, Jinsha Road, Shenzhen, 518120, China
| | - Yue Shen
- BGI-Shenzhen, Beishan Industrial Zone, Shenzhen, 518083, China; China National GeneBank, BGI-Shenzhen, Jinsha Road, Shenzhen, 518120, China; Guangdong Provincial Key Laboratory of Genome Read and Write, Jinsha Road, Shenzhen, 518120, China
| | - Xin Zhao
- BGI-Shenzhen, Beishan Industrial Zone, Shenzhen, 518083, China; China National GeneBank, BGI-Shenzhen, Jinsha Road, Shenzhen, 518120, China
| | - Yuanyuan Ma
- Department of Thoracic Surgery II, Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education/Beijing, Peking University Cancer Hospital and Institute, Beijing, China
| | - Fuqiang Li
- BGI-Shenzhen, Beishan Industrial Zone, Shenzhen, 518083, China; China National GeneBank, BGI-Shenzhen, Jinsha Road, Shenzhen, 518120, China
| | - Jiahui Si
- Department of Thoracic Surgery II, Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education/Beijing, Peking University Cancer Hospital and Institute, Beijing, China
| | - Jian Wang
- BGI-Shenzhen, Beishan Industrial Zone, Shenzhen, 518083, China; China National GeneBank, BGI-Shenzhen, Jinsha Road, Shenzhen, 518120, China
| | - Jinfeng Chen
- Department of Thoracic Surgery II, Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education/Beijing, Peking University Cancer Hospital and Institute, Beijing, China
| | - Huanming Yang
- BGI-Shenzhen, Beishan Industrial Zone, Shenzhen, 518083, China; China National GeneBank, BGI-Shenzhen, Jinsha Road, Shenzhen, 518120, China; Guangdong Provincial Key Laboratory of Genome Read and Write, Jinsha Road, Shenzhen, 518120, China
| | - Xun Xu
- BGI-Shenzhen, Beishan Industrial Zone, Shenzhen, 518083, China; China National GeneBank, BGI-Shenzhen, Jinsha Road, Shenzhen, 518120, China; Guangdong Provincial Key Laboratory of Genome Read and Write, Jinsha Road, Shenzhen, 518120, China.
| | - Yue Yang
- Department of Thoracic Surgery II, Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education/Beijing, Peking University Cancer Hospital and Institute, Beijing, China.
| |
Collapse
|
68
|
Davaadelger B, Murphy AR, Clare SE, Lee O, Khan SA, Kim JJ. Mechanism of Telapristone Acetate (CDB4124) on Progesterone Receptor Action in Breast Cancer Cells. Endocrinology 2018; 159:3581-3595. [PMID: 30203004 PMCID: PMC6157418 DOI: 10.1210/en.2018-00559] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Accepted: 08/30/2018] [Indexed: 12/12/2022]
Abstract
Progesterone is a steroid hormone that plays an important role in the breast. Progesterone exerts its action through binding to progesterone receptor (PR), a transcription factor. Deregulation of the progesterone signaling pathway is implicated in the formation, development, and progression of breast cancer. Next-generation selective progesterone receptor modulators (SPRMs) have potent antiprogestin activity and are selective for PR, reducing the off-target effects on other nuclear receptors. To date, there is limited information on how the newer generation of SPRMs, specifically telapristone acetate (TPA), affect PR function at the molecular level. In this study, T47D breast cancer cells were used to investigate the molecular mechanism by which TPA antagonizes PR action. Global profiling of the PR cistrome and interactome was done with chromatin immunoprecipitation sequencing (ChIP-seq) and rapid immunoprecipitation mass spectrometry. Validation studies were done on key genes and interactions. Our results demonstrate that treatment with the progestin (R5020) alone resulted in robust PR recruitment to the chromatin, and addition of TPA reduced PR recruitment globally. TPA significantly changed coregulator recruitment to PR compared with R5020. Upon conservative analysis, three proteins (TRPS1, LASP1, and AP1G1) were identified in the R5020+TPA-treated group. Silencing TRPS1 with small interfering RNA increased PR occupancy to the known PR regulatory regions and attenuated the inhibition of gene expression after TPA treatment. TRPS1 silencing alleviated the inhibition of proliferation by TPA. In conclusion, TPA decreases PR occupancy on chromatin and recruits coregulators such as TRPS1 to the PR complex, thereby regulating PR target gene expression and associated cellular responses.
Collapse
Affiliation(s)
- Batzaya Davaadelger
- Division of Reproductive Science in Medicine, Department of Obstetrics and Gynecology, Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Alina R Murphy
- Division of Reproductive Science in Medicine, Department of Obstetrics and Gynecology, Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Susan E Clare
- Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Oukseub Lee
- Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Seema A Khan
- Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - J Julie Kim
- Division of Reproductive Science in Medicine, Department of Obstetrics and Gynecology, Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois
- Correspondence: J. Julie Kim, PhD, 303 East Superior Street, Lurie 4-117, Chicago, Illinois 60611. E-mail:
| |
Collapse
|
69
|
|
70
|
Liu B, Hu FF, Zhang Q, Hu H, Ye Z, Tang Q, Guo AY. Genomic landscape and mutational impacts of recurrently mutated genes in cancers. Mol Genet Genomic Med 2018; 6:910-923. [PMID: 30107644 PMCID: PMC6305651 DOI: 10.1002/mgg3.458] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Revised: 05/29/2018] [Accepted: 07/13/2018] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Cancer genes tend to be highly mutated under positive selection. Better understanding the recurrently mutated genes (RMGs) in cancer is critical for explicating the mechanisms of tumorigenesis and providing vital clues for therapy. Although some studies have investigated functional impacts of RMGs in specific cancer types, a comprehensive analysis of RMGs and their mutational impacts across cancers is still needed. METHODS We obtained data from The Cancer Genome Atlas (TCGA) and calculated mutation rate of each gene in 31 cancer types. Functional analysis was performed to identify the important signaling pathways and enriched protein types of RMGs. In order to evaluate functional impacts of RMGs, differential expression, survival, and pairwise mutation patterns analyses were performed. RESULTS Totally, we identified 897 RMGs and 624 of them were specifically mutant in only a single cancer type. Functional analysis demonstrated that these RMGs were enriched in hydrolases, cytoskeletal protein, and pathways like MAPK, cell cycle, PI3K-Akt, ECM receptor interaction, and energy metabolism. The differentially expressed genes potentially affected by the same common RMG showed a relatively low overlap across different cancer types. For the 19 Mucin (MUC) family genes, nine of them were RMGs and four of them (MUC17, MUC5B, MUC4, and MUC16) were common RMGs shared in 8 to 17 cancer types. The results showed that recurrent mutations in MUC genes were significantly associated with better survival prognosis. Only a small part of RMGs was differentially expressed due to their own mutations and most of them were downregulated. In addition, pairwise mutation pattern analysis revealed the high frequency of co-occurred mutations among RMGs in STAD. CONCLUSION Through the functional analysis of RMGs, we found that six signaling pathways were disrupted in most cancer types and that energy metabolism was abnormal in tumors. The results also revealed a strong correlation between recurrently mutated genes from MUC family and human survival. In addition, gene expression and survival prognosis were associated with different mutation types of RMGs.
Collapse
Affiliation(s)
- Baolin Liu
- Department of Bioinformatics and Systems Biology, Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Fei-Fei Hu
- Department of Bioinformatics and Systems Biology, Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Qiong Zhang
- Department of Bioinformatics and Systems Biology, Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Hui Hu
- Department of Bioinformatics and Systems Biology, Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Zheng Ye
- Department of Bioinformatics and Systems Biology, Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China.,Department of Biochemistry and Molecular Biology, Tianjin Key Laboratory of Medical Epigenetics, Tianjin Medical University, Tianjin, China
| | - Qin Tang
- Department of Bioinformatics and Systems Biology, Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - An-Yuan Guo
- Department of Bioinformatics and Systems Biology, Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
71
|
Azimian-Zavareh V, Hossein G, Ebrahimi M, Dehghani-Ghobadi Z. Wnt11 alters integrin and cadherin expression by ovarian cancer spheroids and inhibits tumorigenesis and metastasis. Exp Cell Res 2018; 369:90-104. [PMID: 29753625 DOI: 10.1016/j.yexcr.2018.05.010] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Revised: 04/20/2018] [Accepted: 05/08/2018] [Indexed: 12/14/2022]
Abstract
The present study investigated the role of Wnt11 in multicellular tumor spheroid-like structures (MCTS) ovarian cancer cell proliferation, migration and invasion in vitro and in vivo tumorigenesis and metastasis in xenograft nude mice model. Moreover, samples from human serous ovarian cancer (SOC) were used to assess the association of Wnt11 with integrins and cadherins. The data showed that Wnt11 overexpressing SKOV-3 cells became more compact accompanied by increased expression of E-and N-cadherin and lower expression of EpCAM and CD44. The α5, β2, β3 and β6 integrin subunits expression levels were significantly reduced in Wnt11 overexpressing cells accompanied with significantly reduced disaggregation of Wnt11 overexpressing SKOV-3 MCTS on ECM components. Moreover, Wnt11 overexpressing SKOV-3 MCTS showed decreased migration, invasion as well as no tumor growth and metastasis in vivo. We found that Wnt11 significantly and negatively correlated with ITGB2, ITGB6, and EpCAM and positively with CDH-1 in high-grade SOC specimens. Our results suggest that Wnt11 impedes MCTS attachment to ECM components and therefore can affect ovarian cancer progression.
Collapse
Affiliation(s)
- Vajihe Azimian-Zavareh
- Department of Animal Physiology, School of Biology, College of Science, University of Tehran, Tehran, Iran
| | - Ghamartaj Hossein
- Department of Animal Physiology, School of Biology, College of Science, University of Tehran, Tehran, Iran.
| | - Marzieh Ebrahimi
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
| | - Zeinab Dehghani-Ghobadi
- Department of Animal Physiology, School of Biology, College of Science, University of Tehran, Tehran, Iran
| |
Collapse
|
72
|
Hsu AH, Lum MA, Shim KS, Frederick PJ, Morrison CD, Chen B, Lele SM, Sheinin YM, Daikoku T, Dey SK, Leone G, Black AR, Black JD. Crosstalk between PKCα and PI3K/AKT Signaling Is Tumor Suppressive in the Endometrium. Cell Rep 2018; 24:655-669. [PMID: 30021163 PMCID: PMC6118133 DOI: 10.1016/j.celrep.2018.06.067] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Revised: 04/25/2018] [Accepted: 06/13/2018] [Indexed: 11/17/2022] Open
Abstract
Protein kinase C (PKC) isozymes are commonly recognized as oncoproteins based on their activation by tumor-promoting phorbol esters. However, accumulating evidence indicates that PKCs can be inhibitory in some cancers, with recent findings propelling a shift in focus to understanding tumor suppressive functions of these enzymes. Here, we report that PKCα acts as a tumor suppressor in PI3K/AKT-driven endometrial cancer. Transcriptional suppression of PKCα is observed in human endometrial tumors in association with aggressive disease and poor prognosis. In murine models, loss of PKCα is rate limiting for endometrial tumor initiation. PKCα tumor suppression involves PP2A-family-dependent inactivation of AKT, which can occur even in the context of genetic hyperactivation of PI3K/AKT signaling by coincident mutations in PTEN, PIK3CA, and/or PIK3R1. Together, our data point to PKCα as a crucial tumor suppressor in the endometrium, with deregulation of a PKCα→PP2A/PP2A-like phosphatase signaling axis contributing to robust AKT activation and enhanced endometrial tumorigenesis.
Collapse
Affiliation(s)
- Alice H Hsu
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Michelle A Lum
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Kang-Sup Shim
- Department of Molecular Virology, Immunology, and Medical Genetics, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Peter J Frederick
- Department of Gynecologic Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Carl D Morrison
- Department of Pathology and Laboratory Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Baojiang Chen
- Department of Biostatistics, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Subodh M Lele
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Yuri M Sheinin
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Takiko Daikoku
- Division of Reproductive Sciences, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Sudhansu K Dey
- Division of Reproductive Sciences, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Gustavo Leone
- Department of Molecular Virology, Immunology, and Medical Genetics, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Adrian R Black
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Jennifer D Black
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198, USA.
| |
Collapse
|
73
|
Caumanns JJ, Wisman GBA, Berns K, van der Zee AGJ, de Jong S. ARID1A mutant ovarian clear cell carcinoma: A clear target for synthetic lethal strategies. Biochim Biophys Acta Rev Cancer 2018; 1870:176-184. [PMID: 30025943 DOI: 10.1016/j.bbcan.2018.07.005] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Revised: 07/12/2018] [Accepted: 07/13/2018] [Indexed: 12/20/2022]
Abstract
SWI/SNF chromatin remodeling complexes play an important role in the epigenetic regulation of chromatin structure and gene transcription. Mutual exclusive subunits in the SWI/SNF complex include the DNA targeting members ARID1A and ARID1B as well as the ATPases SMARCA2 and SMARCA4. SWI/SNF complexes are mutated across many cancer types. The highest mutation incidence is found in ARID1A, primarily consisting of deleterious mutations. Current advances have reported synthetic lethal interactions with the loss of ARID1A in several cancer types. In this review, we discuss targets that are only important for tumor growth in an ARID1A mutant context. We focus on synthetic lethal strategies with ARID1A loss in ovarian clear cell carcinoma, a cancer with the highest ARID1A mutation incidence (46-57%). ARID1A directed lethal strategies that can be exploited clinically include targeting of the DNA repair proteins PARP and ATR, and the epigenetic factors EZH2, HDAC2, HDAC6 and BRD2.
Collapse
Affiliation(s)
- Joseph J Caumanns
- Department of Gynecologic Oncology, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| | - G Bea A Wisman
- Department of Gynecologic Oncology, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| | - Katrien Berns
- Division of Molecular Carcinogenesis, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| | - Ate G J van der Zee
- Department of Gynecologic Oncology, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| | - Steven de Jong
- Department of Medical Oncology, Cancer Research Centre Groningen, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands.
| |
Collapse
|
74
|
Suhaimi SS, Ab Mutalib NS, Khor SS, Zain RRM, Syafruddin SE, Abu N, Mohd Dali AZH, Jamal R. Targeted Next-Generation Sequencing Identifies Actionable Targets in Estrogen Receptor Positive and Estrogen Receptor Negative Endometriod Endometrial Cancer. Front Pharmacol 2018; 9:750. [PMID: 30057548 PMCID: PMC6053487 DOI: 10.3389/fphar.2018.00750] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Accepted: 06/20/2018] [Indexed: 12/21/2022] Open
Abstract
Endometrioid endometrial cancer (EEC) is the commonest form of endometrial cancer and can be divided into estrogen receptor (ER) positive and negative subtypes. The mutational profiles of EEC have been shown to aid in tailoring treatment; however, little is known about the differences between the gene mutation profiles between these two subtypes. This study aims to investigate the gene mutation profile in ER positive and negative EEC, and to further elucidate the role of WHSC1 mutations in this cancer. EEC and normal endometrial tissues were obtained from 29 patients and subjected to next-generation sequencing (NGS) using Ion Ampliseq Comprehensive Cancer PanelTM targeting 409 cancer related. A total of 741 non-synonymous alterations were identified from 272 genes in ER positive subtype while 448 non-synonymous variants were identified from 221 genes in ER negative subtype. PTEN is the most frequently altered gene in ER positive subtype (64%, 7/11) while ARID1A is the most frequently altered gene in ER negative subtype (50%, 4/8). We also identified alterations in ERRB3 (36%, 4/11), GNAS (36%, 4/11), and WHSC1 (27%, 3/11) in the ER positive subtype. WHSC1 R1126H and L1268P were shown to significantly increase cell viability, proliferation, migration, and survival. In addition, reduction in ER expression sensitized EEC-1 cell with WHSC1 L1268P mutant to Fulvestrant treatment. We revealed the mutational spectra of ER positive and ER negative EEC that could lead to better understanding of the biological mechanisms of endometrial cancer and may ultimately result in improvement of treatment options and patient prognosis.
Collapse
Affiliation(s)
- Siti Syazani Suhaimi
- UKM Medical Molecular Biology Institute, Universiti Kebangsaan Malaysia, UKM Medical Center, Kuala Lumpur, Malaysia
| | - Nurul-Syakima Ab Mutalib
- UKM Medical Molecular Biology Institute, Universiti Kebangsaan Malaysia, UKM Medical Center, Kuala Lumpur, Malaysia
| | | | - Reena Rahayu Md Zain
- Department of Pathology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Saiful Effendi Syafruddin
- UKM Medical Molecular Biology Institute, Universiti Kebangsaan Malaysia, UKM Medical Center, Kuala Lumpur, Malaysia
| | - Nadiah Abu
- UKM Medical Molecular Biology Institute, Universiti Kebangsaan Malaysia, UKM Medical Center, Kuala Lumpur, Malaysia
| | - Ahmad Zailani Hatta Mohd Dali
- Department of Pathology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
- Department of Obstetrics and Gynaecology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Rahman Jamal
- UKM Medical Molecular Biology Institute, Universiti Kebangsaan Malaysia, UKM Medical Center, Kuala Lumpur, Malaysia
| |
Collapse
|
75
|
Nakamura K, Nakayama K, Ishikawa M, Minamoto T, Ishibashi T, Sato E, Sanuki K, Yamashita H, Ono R, Iida K, Sultana R, Hossain MM, Ishikawa N, Kyo S. Genetic analysis and phosphoinositide 3-kinase/protein kinase B signaling pathway status in ovarian endometrioid borderline tumor samples. Oncol Lett 2018; 16:189-194. [PMID: 29928400 PMCID: PMC6006484 DOI: 10.3892/ol.2018.8626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2016] [Accepted: 03/28/2017] [Indexed: 11/20/2022] Open
Abstract
Ovarian endometrioid borderline tumors (EBTs) are extremely rare, and are thought to be precursors of endometrioid carcinoma, beginning as adenofibroma or endometriosis and progressing in a slow, stepwise manner. In endometrioid carcinomas, a high frequency of activating mutations in phosphatase and tensin homolog (PTEN), β-catenin or AT-rich interaction domain 1A (ARID1A) genes, and the activation of the phosphoinositide 3-kinase (PI3K)/protein kinase B (AKT) signaling pathway have been observed. However, the frequency of these alterations in EBTs and how they contribute to tumor progression remain unclear. To the best of our knowledge, this is the first study to assess the status of the PI3K/AKT signaling pathway in EBTs, in association with PTEN and ARID1A mutations. PTEN mutations were observed in EBTs and also in the area of endometriosis without atypia. However, the PI3K/AKT signaling pathway was revealed to be activated only in EBTs. The observations of the present study suggest that the PTEN mutation represents an early event in EBT tumorigenesis, while additional genetic alterations may be necessary to activate the PI3K/AKT signaling pathway and induce the development of the invasive carcinoma.
Collapse
Affiliation(s)
- Kohei Nakamura
- Department of Obstetrics and Gynecology, Shimane University School of Medicine, Izumo, Shimane 693-8501, Japan
| | - Kentaro Nakayama
- Department of Obstetrics and Gynecology, Shimane University School of Medicine, Izumo, Shimane 693-8501, Japan
| | - Masako Ishikawa
- Department of Obstetrics and Gynecology, Shimane University School of Medicine, Izumo, Shimane 693-8501, Japan
| | - Toshiko Minamoto
- Department of Obstetrics and Gynecology, Shimane University School of Medicine, Izumo, Shimane 693-8501, Japan
| | - Tomoka Ishibashi
- Department of Obstetrics and Gynecology, Shimane University School of Medicine, Izumo, Shimane 693-8501, Japan
| | - Emi Sato
- Department of Obstetrics and Gynecology, Shimane University School of Medicine, Izumo, Shimane 693-8501, Japan
| | - Kaori Sanuki
- Department of Obstetrics and Gynecology, Shimane University School of Medicine, Izumo, Shimane 693-8501, Japan
| | - Hitomi Yamashita
- Department of Obstetrics and Gynecology, Shimane University School of Medicine, Izumo, Shimane 693-8501, Japan
| | - Ruriko Ono
- Department of Obstetrics and Gynecology, Shimane University School of Medicine, Izumo, Shimane 693-8501, Japan
| | - Kouji Iida
- Department of Obstetrics and Gynecology, Shimane University School of Medicine, Izumo, Shimane 693-8501, Japan
| | - Razia Sultana
- Department of Obstetrics and Gynecology, Shimane University School of Medicine, Izumo, Shimane 693-8501, Japan
| | - Mohammad Mahmud Hossain
- Department of Obstetrics and Gynecology, Shimane University School of Medicine, Izumo, Shimane 693-8501, Japan
| | - Noriyoshi Ishikawa
- Department of Organ Pathology, Shimane University School of Medicine, Izumo, Shimane 693-8501, Japan
| | - Satoru Kyo
- Department of Obstetrics and Gynecology, Shimane University School of Medicine, Izumo, Shimane 693-8501, Japan
| |
Collapse
|
76
|
Zou Y, Zhou JY, Guo JB, Wang LQ, Luo Y, Zhang ZY, Liu FY, Tan J, Wang F, Huang OP. The presence of KRAS, PPP2R1A and ARID1A mutations in 101 Chinese samples with ovarian endometriosis. Mutat Res 2018; 809:1-5. [DOI: 10.1016/j.mrfmmm.2018.03.001] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Revised: 02/23/2018] [Accepted: 03/05/2018] [Indexed: 04/10/2023]
|
77
|
Choi M, Kadara H, Zhang J, Parra ER, Rodriguez-Canales J, Gaffney SG, Zhao Z, Behrens C, Fujimoto J, Chow C, Kim K, Kalhor N, Moran C, Rimm D, Swisher S, Gibbons DL, Heymach J, Kaftan E, Townsend JP, Lynch TJ, Schlessinger J, Lee J, Lifton RP, Herbst RS, Wistuba II. Mutation profiles in early-stage lung squamous cell carcinoma with clinical follow-up and correlation with markers of immune function. Ann Oncol 2018; 28:83-89. [PMID: 28177435 DOI: 10.1093/annonc/mdw437] [Citation(s) in RCA: 85] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Background Lung squamous cell carcinoma (LUSC) accounts for 20–30% of non-small cell lung cancers (NSCLCs). There are limited treatment strategies for LUSC in part due to our inadequate understanding of the molecular underpinnings of the disease. We performed whole-exome sequencing (WES) and comprehensive immune profiling of a unique set of clinically annotated early-stage LUSCs to increase our understanding of the pathobiology of this malignancy. Methods Matched pairs of surgically resected stage I-III LUSCs and normal lung tissues (n = 108) were analyzed by WES. Immunohistochemistry and image analysis-based profiling of 10 immune markers were done on a subset of LUSCs (n = 91). Associations among mutations, immune markers and clinicopathological variables were statistically examined using analysis of variance and Fisher’s exact test. Cox proportional hazards regression models were used for statistical analysis of clinical outcome. Results This early-stage LUSC cohort displayed an average of 209 exonic mutations per tumor. Fourteen genes exhibited significant enrichment for somatic mutation: TP53, MLL2, PIK3CA, NFE2L2, CDH8, KEAP1, PTEN, ADCY8, PTPRT, CALCR, GRM8, FBXW7, RB1 and CDKN2A. Among mutated genes associated with poor recurrence-free survival, MLL2 mutations predicted poor prognosis in both TP53 mutant and wild-type LUSCs. We also found that in treated patients, FBXW7 and KEAP1 mutations were associated with poor response to adjuvant therapy, particularly in TP53-mutant tumors. Analysis of mutations with immune markers revealed that ADCY8 and PIK3CA mutations were associated with markedly decreased tumoral PD-L1 expression, LUSCs with PIK3CA mutations exhibited elevated CD45ro levels and CDKN2A-mutant tumors displayed an up-regulated immune response. Conclusion(s) Our findings pinpoint mutated genes that may impact clinical outcome as well as personalized strategies for targeted immunotherapies in early-stage LUSC.
Collapse
Affiliation(s)
- M Choi
- Department of Genetics, Howard Hughes Medical Institute, Maryland
| | - H Kadara
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - J Zhang
- Department of Biostatistics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston
| | - E R Parra
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - J Rodriguez-Canales
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - S G Gaffney
- Department of Ecology and Evolutionary Biology, Yale University, New Haven,Department of Yale School of Public Health
| | - Z Zhao
- Department of Ecology and Evolutionary Biology, Yale University, New Haven,Department of Yale School of Public Health
| | - C Behrens
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - J Fujimoto
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - C Chow
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - K Kim
- Department of Biomedical Research, Biomedical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea
| | - N Kalhor
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston
| | - C Moran
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston
| | - D Rimm
- Yale Comprehensive Cancer Center, Yale School of Medicine, Yale University, New Haven
| | - S Swisher
- Department of Thoracic and Cardiovascular Surgery, The University of Texas MD Anderson Cancer Center, Houston
| | - D L Gibbons
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - J Heymach
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - E Kaftan
- Yale Comprehensive Cancer Center, Yale School of Medicine, Yale University, New Haven
| | - J P Townsend
- Department of Ecology and Evolutionary Biology, Yale University, New Haven,Department of Yale School of Public Health
| | - T J Lynch
- Yale Comprehensive Cancer Center, Yale School of Medicine, Yale University, New Haven
| | - J Schlessinger
- Yale Comprehensive Cancer Center, Yale School of Medicine, Yale University, New Haven
| | - J Lee
- Department of Biostatistics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston
| | - R P Lifton
- Department of Genetics, Howard Hughes Medical Institute, Maryland,Yale Center for Genome Analysis, Yale School of Medicine, Yale University, New Haven, USA
| | - R S Herbst
- Yale Comprehensive Cancer Center, Yale School of Medicine, Yale University, New Haven
| | - I I Wistuba
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, USA
| |
Collapse
|
78
|
Ng PKS, Li J, Jeong KJ, Shao S, Chen H, Tsang YH, Sengupta S, Wang Z, Bhavana VH, Tran R, Soewito S, Minussi DC, Moreno D, Kong K, Dogruluk T, Lu H, Gao J, Tokheim C, Zhou DC, Johnson AM, Zeng J, Ip CKM, Ju Z, Wester M, Yu S, Li Y, Vellano CP, Schultz N, Karchin R, Ding L, Lu Y, Cheung LWT, Chen K, Shaw KR, Meric-Bernstam F, Scott KL, Yi S, Sahni N, Liang H, Mills GB. Systematic Functional Annotation of Somatic Mutations in Cancer. Cancer Cell 2018; 33:450-462.e10. [PMID: 29533785 PMCID: PMC5926201 DOI: 10.1016/j.ccell.2018.01.021] [Citation(s) in RCA: 210] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Revised: 12/07/2017] [Accepted: 01/30/2018] [Indexed: 12/11/2022]
Abstract
The functional impact of the vast majority of cancer somatic mutations remains unknown, representing a critical knowledge gap for implementing precision oncology. Here, we report the development of a moderate-throughput functional genomic platform consisting of efficient mutant generation, sensitive viability assays using two growth factor-dependent cell models, and functional proteomic profiling of signaling effects for select aberrations. We apply the platform to annotate >1,000 genomic aberrations, including gene amplifications, point mutations, indels, and gene fusions, potentially doubling the number of driver mutations characterized in clinically actionable genes. Further, the platform is sufficiently sensitive to identify weak drivers. Our data are accessible through a user-friendly, public data portal. Our study will facilitate biomarker discovery, prediction algorithm improvement, and drug development.
Collapse
Affiliation(s)
- Patrick Kwok-Shing Ng
- Institute for Personalized Cancer Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jun Li
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Kang Jin Jeong
- Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Shan Shao
- Institute for Personalized Cancer Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Hu Chen
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Graduate Program in Quantitative and Computational Biosciences, Baylor College of Medicine, Houston, TX 77030, USA
| | - Yiu Huen Tsang
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Sohini Sengupta
- Division of Oncology, Department of Medicine, Washington University, St. Louis, MO 63108, USA
| | - Zixing Wang
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | | | - Richard Tran
- Institute for Personalized Cancer Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Stephanie Soewito
- Institute for Personalized Cancer Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Darlan Conterno Minussi
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Daniela Moreno
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Kathleen Kong
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Turgut Dogruluk
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Hengyu Lu
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Jianjiong Gao
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Collin Tokheim
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21218, USA; Institute for Computational Medicine, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Daniel Cui Zhou
- Division of Oncology, Department of Medicine, Washington University, St. Louis, MO 63108, USA
| | - Amber M Johnson
- Institute for Personalized Cancer Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jia Zeng
- Institute for Personalized Cancer Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Carman Ka Man Ip
- Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Zhenlin Ju
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Matthew Wester
- Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Shuangxing Yu
- Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Yongsheng Li
- Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Christopher P Vellano
- Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Nikolaus Schultz
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Rachel Karchin
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21218, USA; Institute for Computational Medicine, Johns Hopkins University, Baltimore, MD 21218, USA; Department of Oncology, Johns Hopkins Medicine, Baltimore, MD 21287, USA
| | - Li Ding
- Division of Oncology, Department of Medicine, Washington University, St. Louis, MO 63108, USA; Siteman Cancer Center, Washington University, St. Louis, MO 63108, USA
| | - Yiling Lu
- Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Lydia Wai Ting Cheung
- HKU Shenzhen Institute of Research and Innovation, Shenzhen, China; School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR
| | - Ken Chen
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Kenna R Shaw
- Institute for Personalized Cancer Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Funda Meric-Bernstam
- Institute for Personalized Cancer Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Department of Breast Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Kenneth L Scott
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Song Yi
- Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| | - Nidhi Sahni
- Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Graduate Program in Quantitative and Computational Biosciences, Baylor College of Medicine, Houston, TX 77030, USA.
| | - Han Liang
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Graduate Program in Quantitative and Computational Biosciences, Baylor College of Medicine, Houston, TX 77030, USA.
| | - Gordon B Mills
- Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
79
|
Parra ER, Villalobos P, Zhang J, Behrens C, Mino B, Swisher S, Sepesi B, Weissferdt A, Kalhor N, Heymach JV, Moran C, Zhang J, Lee J, Rodriguez-Canales J, Gibbons D, Wistuba II. Immunohistochemical and Image Analysis-Based Study Shows That Several Immune Checkpoints are Co-expressed in Non-Small Cell Lung Carcinoma Tumors. J Thorac Oncol 2018. [PMID: 29526824 DOI: 10.1016/j.jtho.2018.03.002] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
INTRODUCTION The understanding of immune checkpoint molecules' co-expression in non-small cell lung carcinoma (NCLC) is important to potentially design combinatorial immunotherapy approaches. METHODS We studied 225 formalin-fixed, paraffin-embedded tumor tissues from stage I-III NCLCs - 142 adenocarcinomas (ADCs) and 83 squamous cell carcinomas (SCCs) - placed in tissue microarrays. Nine immune checkpoint markers were evaluated; four (programmed death ligand 1 [PD-L1], B7-H3, B7-H4, and indoleamine 2,3-dioxygenase 1 [IDO-1]) expressed predominantly in malignant cells (MCs) and five (inducible T cell costimulator, V-set immunoregulatory receptor, T-cell immunoglobulin mucin family member 3, lymphocyte activating 3, and OX40) expressed mostly in stromal tumor-associated inflammatory cells (TAICs). All markers were examined using a quantitative image analysis and correlated with clinicopathologic features, TAICs, and molecular characteristics. RESULTS Using above the median value as positive expression in MCs and high density of TAICs expressing those markers, we identified higher expression of immune checkpoints in SCC than ADC. Common simultaneous expression by MCs was PD-L1 + B7-H3 + IDO-1 in ADC and PD-L1 + B7-H3, or B7-H3 + B7-H4, in SCC. TAICs expressing checkpoint were significantly higher in current smokers than in never smokers. Almost all the immune checkpoint markers showed positive correlation with TAICs expressing inflammatory cell markers. KRAS-mutant ADC specimens showed higher expression of PD-L1 in MCs and of B7-H3, T-cell immunoglobulin mucin family member 3, and IDO-1 in TAICs than wild type. Kaplan-Meier survival curves showed worse prognosis in ADC patients with higher B7-H4 expression by MCs. CONCLUSIONS We found frequent immunohistochemical co-expression of immune checkpoints in surgically resected NCLC tumors and correlated with tumor histology, smoking history, tumor size, and the density of inflammatory cells and tumor mutational status.
Collapse
Affiliation(s)
- Edwin Roger Parra
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Pamela Villalobos
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jiexin Zhang
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Carmen Behrens
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Barbara Mino
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Stephen Swisher
- Department of Thoracic and Cardiovascular Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Boris Sepesi
- Department of Thoracic and Cardiovascular Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Annika Weissferdt
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Neda Kalhor
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - John Victor Heymach
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Cesar Moran
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jianjun Zhang
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jack Lee
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jaime Rodriguez-Canales
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Don Gibbons
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas; Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Ignacio I Wistuba
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas; Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| |
Collapse
|
80
|
Yang L, Yang G, Ding Y, Dai Y, Xu S, Guo Q, Xie A, Hu G. Inhibition of PI3K/AKT Signaling Pathway Radiosensitizes Pancreatic Cancer Cells with ARID1A Deficiency in Vitro. J Cancer 2018; 9:890-900. [PMID: 29581767 PMCID: PMC5868153 DOI: 10.7150/jca.21306] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2017] [Accepted: 01/29/2018] [Indexed: 01/05/2023] Open
Abstract
Pancreatic cancer is among the most aggressive human cancers, and is resistant to regular chemotherapy and radiotherapy. The AT-rich interactive domain containing protein 1A (ARID1A) gene, a crucial chromatin remodeling gene, mutates frequently in a broad spectrum of cancers, including pancreatic cancer. Recent evidence suggests that ARID1A acts as tumor suppressor and plays an important role in DNA damage repair (DDR). However, the effect of ARID1A on the radiosensitivity of pancreatic cancer remains unclear. Herein, we investigated the involvement of ARID1A depletion in the radioresistance of pancreatic cancer cells, and explored the underlying mechanisms. The results reveal that knockdown of ARID1A enhances the radioresistance of pancreatic cancer cells through suppressing apoptosis, impairing G2-M checkpoint arrest, strengthening DDR, and accompanying activation of PI3K/AKT signaling pathway. Moreover, upon inhibition of PI3K/AKT pathway by PI3K-inhibitor LY294002 or AKT-inhibitor mk2206, the radiosensitivity of ARID1A-deficient pancreatic cancer cells is improved in vitro via increased apoptosis and weakened DDR. Taken together, these data suggest that loss of ARID1A expression enhances radioresistance of pancreatic cancer through activation of PI3K/AKT pathway, which maybe a promising target for radiosensitization of ARID1A-deficient pancreatic cancer.
Collapse
Affiliation(s)
- Lin Yang
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Guanghai Yang
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Yingjun Ding
- Department of Physiology, University of Oklahoma Health Sciences Center, 975 N 10 th St, Oklahoma City, OK, US, 73104
| | - Yuhong Dai
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Sanpeng Xu
- Department of Pathology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Qiuyun Guo
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Aini Xie
- Department of Cardiovascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Guangyuan Hu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| |
Collapse
|
81
|
Karlsson T, Krakstad C, Tangen IL, Hoivik EA, Pollock PM, Salvesen HB, Lewis AE. Endometrial cancer cells exhibit high expression of p110β and its selective inhibition induces variable responses on PI3K signaling, cell survival and proliferation. Oncotarget 2018; 8:3881-3894. [PMID: 28002804 PMCID: PMC5354802 DOI: 10.18632/oncotarget.13989] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2016] [Accepted: 12/02/2016] [Indexed: 11/25/2022] Open
Abstract
PTEN loss and constitutive activation of the class I phosphoinositide 3-kinase (PI3K) pathway are key drivers of endometrial tumorigenesis. In some cancer types, PTEN-deficient tumors are reliant on class I PI3K p110β (encoded by PIK3CB) activity but little is known about this contribution in endometrial tumorigenesis. In this study, we find that p110β is overexpressed in a panel of 7 endometrial cancer cell lines compared to non-transformed cells. Furthermore, in 234 clinically annotated patient samples, PIK3CB mRNA levels increase significantly in the early phase of tumorigenesis from precursors to low grade primary malignant lesions whereas PIK3CA levels are higher in non-endometrioid compared to endometrioid primary tumors. While high levels of either PIK3CA or PIK3CB associate with poor prognosis, only elevated PIK3CB mRNA levels correlate with a high cell cycle signature score in clinical samples. In cancer cell lines, p110α inhibition reduces cell viability by inducing cell death in PIK3CA mutant cells while p110β inhibition delayed proliferation in PTEN-deficient cells, but not in WT cells. Taken together, our findings suggest that PIK3CB/p110β contributes to some of the pleiotropic functions of PI3K in endometrial cancer, particularly in the early steps by contributing to cell proliferation.
Collapse
Affiliation(s)
- Thomas Karlsson
- Department of Molecular Biology, University of Bergen, Bergen, Norway
| | - Camilla Krakstad
- Centre for Cancer Biomarkers, Department of Clinical Science, University of Bergen, Bergen, Norway.,Department of Biomedicine, University of Bergen, Bergen, Norway.,Department of Gynecology and Obstetrics, Haukeland University Hospital, Bergen, Norway
| | - Ingvild Løberg Tangen
- Centre for Cancer Biomarkers, Department of Clinical Science, University of Bergen, Bergen, Norway.,Department of Gynecology and Obstetrics, Haukeland University Hospital, Bergen, Norway
| | - Erling A Hoivik
- Centre for Cancer Biomarkers, Department of Clinical Science, University of Bergen, Bergen, Norway.,Department of Gynecology and Obstetrics, Haukeland University Hospital, Bergen, Norway
| | | | - Helga B Salvesen
- Centre for Cancer Biomarkers, Department of Clinical Science, University of Bergen, Bergen, Norway.,Department of Gynecology and Obstetrics, Haukeland University Hospital, Bergen, Norway
| | - Aurélia E Lewis
- Department of Molecular Biology, University of Bergen, Bergen, Norway
| |
Collapse
|
82
|
Aslan O, Cremona M, Morgan C, Cheung LW, Mills GB, Hennessy BT. Preclinical evaluation and reverse phase protein Array-based profiling of PI3K and MEK inhibitors in endometrial carcinoma in vitro. BMC Cancer 2018; 18:168. [PMID: 29426295 PMCID: PMC5807759 DOI: 10.1186/s12885-018-4035-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Accepted: 01/23/2018] [Indexed: 02/01/2023] Open
Abstract
BACKGROUND The phosphoinositide-3-kinase (PI3K) pathway is the most commonly activated pathway in cancers due to mutations at multiple nodes and loss of PTEN. Furthermore, in endometrial cancer (EC), PI3K and RAS/RAF/MEK/MAPK (RAS/MAPK herein) pathway mutations frequently co-exist. We examined the role of PI3K and RAS/MAPK pathway mutations in determining responsiveness to therapies targeted to these pathways in vitro in EC. METHODS 13 EC cell lines were profiled for their PI3K pathway and KRAS mutational and PTEN protein status and treated with one MEK- and two PI3K- targeted inhibitors alone and in combination. Expression and phosphorylation of 66 proteins were evaluated by Reverse-Phase-Protein-Array (RPPA) in 6 EC cell lines to identify signalling changes in these pathways in response to therapy. RESULTS PTEN protein loss and the absence of any tested pathway mutations are dominant negative predictors of sensitivity to MEK inhibition. KRAS-mutated cells were most sensitive to MEK inhibition, but significantly more resistant to PI3K inhibition than KRAS-wild-type cell lines. Combinations of PI3K and MEK inhibitors showed synergy or additivity in all but two cell lines tested. Treatment of KRAS-mutated cells with PI3K inhibitors and treatment of PTEN-low cells with a MEK inhibitor were most likely to induce activation of MEK/MAPK and AKT, respectively, likely indicative of feedback-loop regulation. CONCLUSIONS MEK inhibition may be a promising treatment modality, not just for ECs with mutated KRAS, but also for those with retained PTEN. Up-regulation of MEK/MAPK signalling by PI3K inhibition, and up-regulation of AKT activation by MEK inhibition may serve as potential biomarkers of likely responsiveness to each inhibitor.
Collapse
Affiliation(s)
- Ozlem Aslan
- Department of Medical Oncology, Royal College of Surgeons in Ireland, Beaumont Hospital, Dublin 9, Ireland
| | - Mattia Cremona
- Department of Medical Oncology, Royal College of Surgeons in Ireland, Beaumont Hospital, Dublin 9, Ireland
| | - Clare Morgan
- Department of Medical Oncology, Royal College of Surgeons in Ireland, Beaumont Hospital, Dublin 9, Ireland
| | - Lydia W. Cheung
- Department of Systems Biology, the University of Texas M.D. Anderson Cancer Center, Houston, TX 77030 USA
| | - Gordon B. Mills
- Department of Systems Biology, the University of Texas M.D. Anderson Cancer Center, Houston, TX 77030 USA
| | - Bryan T. Hennessy
- Department of Medical Oncology, Royal College of Surgeons in Ireland, Beaumont Hospital, Dublin 9, Ireland
| |
Collapse
|
83
|
Yang Y, Bao W, Sang Z, Yang Y, Lu M, Xi X. Microarray pathway analysis indicated that mitogen-activated protein kinase/extracellular signal-regulated kinase and insulin growth factor 1 signaling pathways were inhibited by small interfering RNA against AT-rich interactive domain 1A in endometrial cancer. Oncol Lett 2018; 15:1829-1838. [PMID: 29399196 DOI: 10.3892/ol.2017.7489] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2017] [Accepted: 10/20/2017] [Indexed: 01/08/2023] Open
Abstract
Mutations in the gene encoding AT-rich interactive domain 1A (ARID1A) are frequently observed in endometrial cancer (EC) but the molecular mechanisms linking the genetic changes remain to be fully understood. The present study aimed to elucidate the influence of ARID1A mutations on signaling pathways. Missense, synonymous and nonsense heterozygous ARID1A mutations in the EC HEC-1-A cell line were verified by Sanger sequencing. Mutated ARID1A small interfering RNA was transfected into HEC-1-A cells. Biochemical microarray analysis revealed 13 upregulated pathways, 17 downregulated pathways, 14 significantly affected disease states and functions, 662 upstream and 512 downstream genes in mutated ARID1A-depleted HEC-1-A cells, among which the mitogen-activated protein kinase/extracellular signal-regulated kinase and insulin-like growth factor-1 (IGF1) signaling pathways were the 2 most downregulated pathways. Furthermore, the forkhead box protein O1 pathway was upregulated, while the IGF1 receptor, insulin receptor substrate 1 and phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit b pathways were downregulated. Carcinoma tumorigenesis, tumor cell mitosis and tumor cell death were significantly upregulated disease states and functions, while cell proliferation and tumor growth were significantly downregulated. The results of the present study suggested that ARID1A may be a potential prognostic and therapeutic molecular drug target for the prevention of EC progression.
Collapse
Affiliation(s)
- Ye Yang
- Department of Obstetrics and Gynecology, Shanghai General Hospital of Nanjing Medical University, Shanghai 200080, P.R. China
| | - Wei Bao
- Department of Obstetrics and Gynecology, Shanghai General Hospital of Shanghai Jiaotong University, Shanghai 200080, P.R. China
| | - Zhengyu Sang
- Department of Obstetrics and Gynecology, Shanghai General Hospital of Shanghai Jiaotong University, Shanghai 200080, P.R. China
| | - Yongbing Yang
- Department of Obstetrics and Gynecology, Shanghai General Hospital of Shanghai Jiaotong University, Shanghai 200080, P.R. China
| | - Meng Lu
- Department of Obstetrics and Gynecology, Shanghai General Hospital of Shanghai Jiaotong University, Shanghai 200080, P.R. China
| | - Xiaowei Xi
- Department of Obstetrics and Gynecology, Shanghai General Hospital of Nanjing Medical University, Shanghai 200080, P.R. China
| |
Collapse
|
84
|
Arnaud O, Le Loarer F, Tirode F. BAFfling pathologies: Alterations of BAF complexes in cancer. Cancer Lett 2018; 419:266-279. [PMID: 29374542 DOI: 10.1016/j.canlet.2018.01.046] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Revised: 01/12/2018] [Accepted: 01/12/2018] [Indexed: 01/08/2023]
Abstract
To activate or repress specific genes, chromatin is constantly modified by chromatin-remodeling complexes. Among these complexes, the SWItch/Sucrose Non-Fermenting (SWI/SNF) complex, also referred to as BRG1-Associated Factor (BAF) complex, moves the nucleosome along chromatin using energy provided by ATP hydrolysis. In mammalian organisms, the SWI/SNF complex is composed of 10-15 subunits, depending on cell type, and a defect in one of these subunits can have dramatic consequences. In this review we will focus on the alterations identified in the SWI/SNF (BAF) complex subunits that lead to cancerous pathologies. While SMARCB1 was the first mutated subunit to be reported in a majority of malignant rhabdoid tumors, the advent of next-generation sequencing allowed the discovery of mutations in various SWI/SNF subunits within a broad spectrum of cancers. In most cases, the mutation leads to a loss of expression or to a truncated subunit unable to perform its function. Even though it is now commonly acknowledged that approximately 20% of all cancers present a mutation in a SWI/SNF subunit, some cancers are associated to a specific alteration of a SWI/SNF subunit, which acts either as tumor suppressor genes or as oncogenes, and therefore constitute diagnostic or prognostic biomarkers. Consistently, therapeutic strategies targeting SWI/SNF subunits or the genes affected downstream have been revealed to treat cancers.
Collapse
Affiliation(s)
- Ophelie Arnaud
- Univ Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Cancer Research Center of Lyon, Centre Léon Bérard, F-69008, Lyon, France
| | | | - Franck Tirode
- Univ Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Cancer Research Center of Lyon, Centre Léon Bérard, F-69008, Lyon, France; Department of Translational Research and Innovation, Centre Léon Bérard, F-69008, Lyon, France.
| |
Collapse
|
85
|
Chromatin remodeling gene AT-rich interactive domain-containing protein 1A suppresses gastric cancer cell proliferation by targeting PIK3CA and PDK1. Oncotarget 2018; 7:46127-46141. [PMID: 27323812 PMCID: PMC5216786 DOI: 10.18632/oncotarget.10060] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Accepted: 06/01/2016] [Indexed: 12/13/2022] Open
Abstract
The tumor suppressor gene AT-rich interactive domain-containing protein 1A (ARID1A) was frequently mutated in cancers. The modulation mechanism of ARID1A for PI3K/AKT signaling in gastric cancer (GC) remains elusive. Here, we found that depletion of endogenous ARID1A enhanced the in vitro proliferation, colony formation, cellular growth, nutrient uptake and in vivo xenograft tumor growth of GC cells. PI3K/AKT activation by ARID1A-silencing was profiled using a phospho-protein antibody array. The phosphorylation of PDK1, AKT, GSK3β and 70S6K, and the protein and mRNA expressions of PI3K and PDK1, were upregulated by ARID1A-silencing. Chromatin immunoprecipitation and luciferase reporter assay revealed that ARID1A-involved SWI/SNF complex inhibited PIK3CA and PDK1 transcription by direct binding to their promoters. Serial deletion mutation analyses revealed that the ARID1A central region containing the HIC1-binding domain, but not the ARID DNA-binding domain and the C-terminal domain, was essential for the inhibition of GC cell growth, PI3K/AKT pathway phosphorylation and its transcriptional modulation activity of PIK3CA and PDK1. The proliferation, cellular growth and glucose consumption of ARID1A-deficient GC cells were efficiently prohibited by allosteric inhibitors mk2206 and LY294002, which targeting AKT and PI3K, respectively. Both inhibitors also downregulated the phosphorylation of PI3K/AKT pathway in ARID1A-deficient GC cells. Such cells were sensitized to the treatment of LY294002, and AT7867, another inhibitor of AKT and p70S6K. The administration of LY294002 alone inhibited the in vivo growth of ARID1A- deficient GC cells in mouse xenograft model. Our study provides a novel insight into the modulatory function and mechanism of ARID1A in PI3K/AKT signaling in GC.
Collapse
|
86
|
Yuan F, Lu W. Prediction of potential drivers connecting different dysfunctional levels in lung adenocarcinoma via a protein-protein interaction network. Biochim Biophys Acta Mol Basis Dis 2017; 1864:2284-2293. [PMID: 29197663 DOI: 10.1016/j.bbadis.2017.11.018] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Revised: 11/13/2017] [Accepted: 11/23/2017] [Indexed: 12/14/2022]
Abstract
Lung cancer is a serious disease that threatens an affected individual's life. Its pathogenesis has not yet to be fully described, thereby impeding the development of effective treatments and preventive measures. "Cancer driver" theory considers that tumor initiation can be associated with a number of specific mutations in genes called cancer driver genes. Four omics levels, namely, (1) methylation, (2) microRNA, (3) mutation, and (4) mRNA levels, are utilized to cluster cancer driver genes. In this study, the known dysfunctional genes of these four levels were used to identify novel driver genes of lung adenocarcinoma, a subtype of lung cancer. These genes could contribute to the initiation and progression of lung adenocarcinoma in at least two levels. First, random walk with restart algorithm was performed on a protein-protein interaction (PPI) network constructed with PPI information in STRING by using known dysfunctional genes as seed nodes for each level, thereby yielding four groups of possible genes. Second, these genes were further evaluated in a test strategy to exclude false positives and select the most important ones. Finally, after conducting an intersection operation in any two groups of genes, we obtained several inferred driver genes that contributed to the initiation of lung adenocarcinoma in at least two omics levels. Several genes from these groups could be confirmed according to recently published studies. The inferred genes reported in this study were also different from those described in a previous study, suggesting that they can be used as essential supplementary data for investigations on the initiation of lung adenocarcinoma. This article is part of a Special Issue entitled: Accelerating Precision Medicine through Genetic and Genomic Big Data Analysis edited by Yudong Cai & Tao Huang.
Collapse
Affiliation(s)
- Fei Yuan
- Department of Science & Technology, Binzhou Medical University Hospital, Binzhou 256603, Shandong, China.
| | - WenCong Lu
- Department of Chemistry, Shanghai University, Shanghai 200072, China.
| |
Collapse
|
87
|
Berstein LM, Berlev IV, Baltrukova AN. Endometrial cancer evolution: new molecular-biologic types and hormonal-metabolic shifts. Future Oncol 2017; 13:2593-2605. [PMID: 29168655 DOI: 10.2217/fon-2017-0217] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The question hidden in the title of this manuscript (whether the topic develops or remains constant) is important for all areas of science. It is also a serious problem for endometrial cancer (EC) study. In recent times the incidence of EC gradually increases in parallel with obesity epidemics. The main point of this review was evaluation of changes in EC area in last few decades, which are not only seen in tumor incidence, but also in its biology, hormonal-metabolic characteristics of patients and in the ratio of risk and anti-risk factors. One can hope that data accumulated recently and summarized here under the notion of EC evolution will find its use for advancement of EC prevention and treatment.
Collapse
Affiliation(s)
- Lev M Berstein
- Laboratory of Oncoendocrinology, NN Petrov Research Institute of Oncology, St Petersburg, Russia
| | - Igor V Berlev
- Department of Gynecological Oncology, NN Petrov Research Institute of Oncology, St Petersburg, Russia.,Department of Obstetrics and Gynecology, Northwestern State Medical University named after II Mechnikov, St Petersburg, Russia
| | - Alexandra N Baltrukova
- Laboratory of Oncoendocrinology, NN Petrov Research Institute of Oncology, St Petersburg, Russia.,Department of Obstetrics and Gynecology, Northwestern State Medical University named after II Mechnikov, St Petersburg, Russia
| |
Collapse
|
88
|
Morton LM, Sampson JN, Armstrong GT, Chen TH, Hudson MM, Karlins E, Dagnall CL, Li SA, Wilson CL, Srivastava DK, Liu W, Kang G, Oeffinger KC, Henderson TO, Moskowitz CS, Gibson TM, Merino DM, Wong JR, Hammond S, Neglia JP, Turcotte LM, Miller J, Bowen L, Wheeler WA, Leisenring WM, Whitton JA, Burdette L, Chung C, Hicks BD, Jones K, Machiela MJ, Vogt A, Wang Z, Yeager M, Neale G, Lear M, Strong LC, Yasui Y, Stovall M, Weathers RE, Smith SA, Howell R, Davies SM, Radloff GA, Onel K, Berrington de González A, Inskip PD, Rajaraman P, Fraumeni JF, Bhatia S, Chanock SJ, Tucker MA, Robison LL. Genome-Wide Association Study to Identify Susceptibility Loci That Modify Radiation-Related Risk for Breast Cancer After Childhood Cancer. J Natl Cancer Inst 2017; 109:3855140. [PMID: 29059430 DOI: 10.1093/jnci/djx058] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Accepted: 03/08/2017] [Indexed: 12/28/2022] Open
Abstract
Background Childhood cancer survivors treated with chest-directed radiotherapy have substantially elevated risk for developing breast cancer. Although genetic susceptibility to breast cancer in the general population is well studied, large-scale evaluation of breast cancer susceptibility after chest-directed radiotherapy for childhood cancer is lacking. Methods We conducted a genome-wide association study of breast cancer in female survivors of childhood cancer, pooling two cohorts with detailed treatment data and systematic, long-term follow-up: the Childhood Cancer Survivor Study and St. Jude Lifetime Cohort. The study population comprised 207 survivors who developed breast cancer and 2774 who had not developed any subsequent neoplasm as of last follow-up. Genotyping and subsequent imputation yielded 16 958 466 high-quality variants for analysis. We tested associations in the overall population and in subgroups stratified by receipt of lower than 10 and 10 or higher gray breast radiation exposure. We report P values and pooled per-allele risk estimates from Cox proportional hazards regression models. All statistical tests were two-sided. Results Among survivors who received 10 or higher gray breast radiation exposure, a locus on 1q41 was associated with subsequent breast cancer risk (rs4342822, nearest gene PROX1 , risk allele frequency in control subjects [RAF controls ] = 0.46, hazard ratio = 1.92, 95% confidence interval = 1.49 to 2.44, P = 7.09 × 10 -9 ). Two rare variants also showed potentially promising associations (breast radiation ≥10 gray: rs74949440, 11q23, TAGLN , RAF controls = 0.02, P = 5.84 × 10 -8 ; <10 gray: rs17020562, 1q32.3, RPS6KC1 , RAF controls = 0.0005, P = 6.68 × 10 -8 ). Associations were restricted to these dose subgroups, with consistent findings in the two survivor cohorts. Conclusions Our study provides strong evidence that germline genetics outside high-risk syndromes could modify the effect of radiation exposure on breast cancer risk after childhood cancer.
Collapse
Affiliation(s)
- Lindsay M Morton
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, Bethesda, MD; Department of Epidemiology and Cancer Control, Division of Cancer Survivorship, Department of Oncology, Department of Biostatistics, Hartwell Center for Bioinformatics and Biotechnology, and Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN; Cancer Genomics Research Laboratory, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD; Departments of Medicine, Pediatrics, and Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY; Section of Hematology, Oncology and Stem Cell Transplantation, Department of Pediatrics, University of Chicago, Chicago, IL; Nationwide Children's Hospital and the Ohio State University School of Medicine, Columbus, OH; Department of Pediatrics, University of Minnesota, Minneapolis, MN; Information Management Services, Inc., Calverton, MD; Cancer Prevention and Clinical Statistics Programs and Cancer Prevention Program, Fred Hutchinson Cancer Research Center, Seattle, WA; Department of Genetics and Department of Radiation Physics, The University of Texas at MD Anderson Cancer Center, Houston, TX; Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH; Institute for Cancer Outcomes and Survivorship, University of Alabama at Birmingham, Birmingham, AL
| | - Joshua N Sampson
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, Bethesda, MD; Department of Epidemiology and Cancer Control, Division of Cancer Survivorship, Department of Oncology, Department of Biostatistics, Hartwell Center for Bioinformatics and Biotechnology, and Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN; Cancer Genomics Research Laboratory, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD; Departments of Medicine, Pediatrics, and Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY; Section of Hematology, Oncology and Stem Cell Transplantation, Department of Pediatrics, University of Chicago, Chicago, IL; Nationwide Children's Hospital and the Ohio State University School of Medicine, Columbus, OH; Department of Pediatrics, University of Minnesota, Minneapolis, MN; Information Management Services, Inc., Calverton, MD; Cancer Prevention and Clinical Statistics Programs and Cancer Prevention Program, Fred Hutchinson Cancer Research Center, Seattle, WA; Department of Genetics and Department of Radiation Physics, The University of Texas at MD Anderson Cancer Center, Houston, TX; Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH; Institute for Cancer Outcomes and Survivorship, University of Alabama at Birmingham, Birmingham, AL
| | - Gregory T Armstrong
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, Bethesda, MD; Department of Epidemiology and Cancer Control, Division of Cancer Survivorship, Department of Oncology, Department of Biostatistics, Hartwell Center for Bioinformatics and Biotechnology, and Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN; Cancer Genomics Research Laboratory, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD; Departments of Medicine, Pediatrics, and Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY; Section of Hematology, Oncology and Stem Cell Transplantation, Department of Pediatrics, University of Chicago, Chicago, IL; Nationwide Children's Hospital and the Ohio State University School of Medicine, Columbus, OH; Department of Pediatrics, University of Minnesota, Minneapolis, MN; Information Management Services, Inc., Calverton, MD; Cancer Prevention and Clinical Statistics Programs and Cancer Prevention Program, Fred Hutchinson Cancer Research Center, Seattle, WA; Department of Genetics and Department of Radiation Physics, The University of Texas at MD Anderson Cancer Center, Houston, TX; Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH; Institute for Cancer Outcomes and Survivorship, University of Alabama at Birmingham, Birmingham, AL
| | - Ting-Huei Chen
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, Bethesda, MD; Department of Epidemiology and Cancer Control, Division of Cancer Survivorship, Department of Oncology, Department of Biostatistics, Hartwell Center for Bioinformatics and Biotechnology, and Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN; Cancer Genomics Research Laboratory, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD; Departments of Medicine, Pediatrics, and Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY; Section of Hematology, Oncology and Stem Cell Transplantation, Department of Pediatrics, University of Chicago, Chicago, IL; Nationwide Children's Hospital and the Ohio State University School of Medicine, Columbus, OH; Department of Pediatrics, University of Minnesota, Minneapolis, MN; Information Management Services, Inc., Calverton, MD; Cancer Prevention and Clinical Statistics Programs and Cancer Prevention Program, Fred Hutchinson Cancer Research Center, Seattle, WA; Department of Genetics and Department of Radiation Physics, The University of Texas at MD Anderson Cancer Center, Houston, TX; Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH; Institute for Cancer Outcomes and Survivorship, University of Alabama at Birmingham, Birmingham, AL
| | - Melissa M Hudson
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, Bethesda, MD; Department of Epidemiology and Cancer Control, Division of Cancer Survivorship, Department of Oncology, Department of Biostatistics, Hartwell Center for Bioinformatics and Biotechnology, and Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN; Cancer Genomics Research Laboratory, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD; Departments of Medicine, Pediatrics, and Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY; Section of Hematology, Oncology and Stem Cell Transplantation, Department of Pediatrics, University of Chicago, Chicago, IL; Nationwide Children's Hospital and the Ohio State University School of Medicine, Columbus, OH; Department of Pediatrics, University of Minnesota, Minneapolis, MN; Information Management Services, Inc., Calverton, MD; Cancer Prevention and Clinical Statistics Programs and Cancer Prevention Program, Fred Hutchinson Cancer Research Center, Seattle, WA; Department of Genetics and Department of Radiation Physics, The University of Texas at MD Anderson Cancer Center, Houston, TX; Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH; Institute for Cancer Outcomes and Survivorship, University of Alabama at Birmingham, Birmingham, AL
| | - Eric Karlins
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, Bethesda, MD; Department of Epidemiology and Cancer Control, Division of Cancer Survivorship, Department of Oncology, Department of Biostatistics, Hartwell Center for Bioinformatics and Biotechnology, and Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN; Cancer Genomics Research Laboratory, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD; Departments of Medicine, Pediatrics, and Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY; Section of Hematology, Oncology and Stem Cell Transplantation, Department of Pediatrics, University of Chicago, Chicago, IL; Nationwide Children's Hospital and the Ohio State University School of Medicine, Columbus, OH; Department of Pediatrics, University of Minnesota, Minneapolis, MN; Information Management Services, Inc., Calverton, MD; Cancer Prevention and Clinical Statistics Programs and Cancer Prevention Program, Fred Hutchinson Cancer Research Center, Seattle, WA; Department of Genetics and Department of Radiation Physics, The University of Texas at MD Anderson Cancer Center, Houston, TX; Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH; Institute for Cancer Outcomes and Survivorship, University of Alabama at Birmingham, Birmingham, AL
| | - Casey L Dagnall
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, Bethesda, MD; Department of Epidemiology and Cancer Control, Division of Cancer Survivorship, Department of Oncology, Department of Biostatistics, Hartwell Center for Bioinformatics and Biotechnology, and Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN; Cancer Genomics Research Laboratory, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD; Departments of Medicine, Pediatrics, and Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY; Section of Hematology, Oncology and Stem Cell Transplantation, Department of Pediatrics, University of Chicago, Chicago, IL; Nationwide Children's Hospital and the Ohio State University School of Medicine, Columbus, OH; Department of Pediatrics, University of Minnesota, Minneapolis, MN; Information Management Services, Inc., Calverton, MD; Cancer Prevention and Clinical Statistics Programs and Cancer Prevention Program, Fred Hutchinson Cancer Research Center, Seattle, WA; Department of Genetics and Department of Radiation Physics, The University of Texas at MD Anderson Cancer Center, Houston, TX; Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH; Institute for Cancer Outcomes and Survivorship, University of Alabama at Birmingham, Birmingham, AL
| | - Shengchao Alfred Li
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, Bethesda, MD; Department of Epidemiology and Cancer Control, Division of Cancer Survivorship, Department of Oncology, Department of Biostatistics, Hartwell Center for Bioinformatics and Biotechnology, and Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN; Cancer Genomics Research Laboratory, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD; Departments of Medicine, Pediatrics, and Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY; Section of Hematology, Oncology and Stem Cell Transplantation, Department of Pediatrics, University of Chicago, Chicago, IL; Nationwide Children's Hospital and the Ohio State University School of Medicine, Columbus, OH; Department of Pediatrics, University of Minnesota, Minneapolis, MN; Information Management Services, Inc., Calverton, MD; Cancer Prevention and Clinical Statistics Programs and Cancer Prevention Program, Fred Hutchinson Cancer Research Center, Seattle, WA; Department of Genetics and Department of Radiation Physics, The University of Texas at MD Anderson Cancer Center, Houston, TX; Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH; Institute for Cancer Outcomes and Survivorship, University of Alabama at Birmingham, Birmingham, AL
| | - Carmen L Wilson
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, Bethesda, MD; Department of Epidemiology and Cancer Control, Division of Cancer Survivorship, Department of Oncology, Department of Biostatistics, Hartwell Center for Bioinformatics and Biotechnology, and Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN; Cancer Genomics Research Laboratory, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD; Departments of Medicine, Pediatrics, and Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY; Section of Hematology, Oncology and Stem Cell Transplantation, Department of Pediatrics, University of Chicago, Chicago, IL; Nationwide Children's Hospital and the Ohio State University School of Medicine, Columbus, OH; Department of Pediatrics, University of Minnesota, Minneapolis, MN; Information Management Services, Inc., Calverton, MD; Cancer Prevention and Clinical Statistics Programs and Cancer Prevention Program, Fred Hutchinson Cancer Research Center, Seattle, WA; Department of Genetics and Department of Radiation Physics, The University of Texas at MD Anderson Cancer Center, Houston, TX; Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH; Institute for Cancer Outcomes and Survivorship, University of Alabama at Birmingham, Birmingham, AL
| | - Deo Kumar Srivastava
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, Bethesda, MD; Department of Epidemiology and Cancer Control, Division of Cancer Survivorship, Department of Oncology, Department of Biostatistics, Hartwell Center for Bioinformatics and Biotechnology, and Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN; Cancer Genomics Research Laboratory, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD; Departments of Medicine, Pediatrics, and Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY; Section of Hematology, Oncology and Stem Cell Transplantation, Department of Pediatrics, University of Chicago, Chicago, IL; Nationwide Children's Hospital and the Ohio State University School of Medicine, Columbus, OH; Department of Pediatrics, University of Minnesota, Minneapolis, MN; Information Management Services, Inc., Calverton, MD; Cancer Prevention and Clinical Statistics Programs and Cancer Prevention Program, Fred Hutchinson Cancer Research Center, Seattle, WA; Department of Genetics and Department of Radiation Physics, The University of Texas at MD Anderson Cancer Center, Houston, TX; Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH; Institute for Cancer Outcomes and Survivorship, University of Alabama at Birmingham, Birmingham, AL
| | - Wei Liu
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, Bethesda, MD; Department of Epidemiology and Cancer Control, Division of Cancer Survivorship, Department of Oncology, Department of Biostatistics, Hartwell Center for Bioinformatics and Biotechnology, and Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN; Cancer Genomics Research Laboratory, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD; Departments of Medicine, Pediatrics, and Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY; Section of Hematology, Oncology and Stem Cell Transplantation, Department of Pediatrics, University of Chicago, Chicago, IL; Nationwide Children's Hospital and the Ohio State University School of Medicine, Columbus, OH; Department of Pediatrics, University of Minnesota, Minneapolis, MN; Information Management Services, Inc., Calverton, MD; Cancer Prevention and Clinical Statistics Programs and Cancer Prevention Program, Fred Hutchinson Cancer Research Center, Seattle, WA; Department of Genetics and Department of Radiation Physics, The University of Texas at MD Anderson Cancer Center, Houston, TX; Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH; Institute for Cancer Outcomes and Survivorship, University of Alabama at Birmingham, Birmingham, AL
| | - Guolian Kang
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, Bethesda, MD; Department of Epidemiology and Cancer Control, Division of Cancer Survivorship, Department of Oncology, Department of Biostatistics, Hartwell Center for Bioinformatics and Biotechnology, and Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN; Cancer Genomics Research Laboratory, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD; Departments of Medicine, Pediatrics, and Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY; Section of Hematology, Oncology and Stem Cell Transplantation, Department of Pediatrics, University of Chicago, Chicago, IL; Nationwide Children's Hospital and the Ohio State University School of Medicine, Columbus, OH; Department of Pediatrics, University of Minnesota, Minneapolis, MN; Information Management Services, Inc., Calverton, MD; Cancer Prevention and Clinical Statistics Programs and Cancer Prevention Program, Fred Hutchinson Cancer Research Center, Seattle, WA; Department of Genetics and Department of Radiation Physics, The University of Texas at MD Anderson Cancer Center, Houston, TX; Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH; Institute for Cancer Outcomes and Survivorship, University of Alabama at Birmingham, Birmingham, AL
| | - Kevin C Oeffinger
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, Bethesda, MD; Department of Epidemiology and Cancer Control, Division of Cancer Survivorship, Department of Oncology, Department of Biostatistics, Hartwell Center for Bioinformatics and Biotechnology, and Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN; Cancer Genomics Research Laboratory, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD; Departments of Medicine, Pediatrics, and Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY; Section of Hematology, Oncology and Stem Cell Transplantation, Department of Pediatrics, University of Chicago, Chicago, IL; Nationwide Children's Hospital and the Ohio State University School of Medicine, Columbus, OH; Department of Pediatrics, University of Minnesota, Minneapolis, MN; Information Management Services, Inc., Calverton, MD; Cancer Prevention and Clinical Statistics Programs and Cancer Prevention Program, Fred Hutchinson Cancer Research Center, Seattle, WA; Department of Genetics and Department of Radiation Physics, The University of Texas at MD Anderson Cancer Center, Houston, TX; Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH; Institute for Cancer Outcomes and Survivorship, University of Alabama at Birmingham, Birmingham, AL
| | - Tara O Henderson
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, Bethesda, MD; Department of Epidemiology and Cancer Control, Division of Cancer Survivorship, Department of Oncology, Department of Biostatistics, Hartwell Center for Bioinformatics and Biotechnology, and Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN; Cancer Genomics Research Laboratory, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD; Departments of Medicine, Pediatrics, and Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY; Section of Hematology, Oncology and Stem Cell Transplantation, Department of Pediatrics, University of Chicago, Chicago, IL; Nationwide Children's Hospital and the Ohio State University School of Medicine, Columbus, OH; Department of Pediatrics, University of Minnesota, Minneapolis, MN; Information Management Services, Inc., Calverton, MD; Cancer Prevention and Clinical Statistics Programs and Cancer Prevention Program, Fred Hutchinson Cancer Research Center, Seattle, WA; Department of Genetics and Department of Radiation Physics, The University of Texas at MD Anderson Cancer Center, Houston, TX; Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH; Institute for Cancer Outcomes and Survivorship, University of Alabama at Birmingham, Birmingham, AL
| | - Chaya S Moskowitz
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, Bethesda, MD; Department of Epidemiology and Cancer Control, Division of Cancer Survivorship, Department of Oncology, Department of Biostatistics, Hartwell Center for Bioinformatics and Biotechnology, and Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN; Cancer Genomics Research Laboratory, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD; Departments of Medicine, Pediatrics, and Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY; Section of Hematology, Oncology and Stem Cell Transplantation, Department of Pediatrics, University of Chicago, Chicago, IL; Nationwide Children's Hospital and the Ohio State University School of Medicine, Columbus, OH; Department of Pediatrics, University of Minnesota, Minneapolis, MN; Information Management Services, Inc., Calverton, MD; Cancer Prevention and Clinical Statistics Programs and Cancer Prevention Program, Fred Hutchinson Cancer Research Center, Seattle, WA; Department of Genetics and Department of Radiation Physics, The University of Texas at MD Anderson Cancer Center, Houston, TX; Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH; Institute for Cancer Outcomes and Survivorship, University of Alabama at Birmingham, Birmingham, AL
| | - Todd M Gibson
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, Bethesda, MD; Department of Epidemiology and Cancer Control, Division of Cancer Survivorship, Department of Oncology, Department of Biostatistics, Hartwell Center for Bioinformatics and Biotechnology, and Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN; Cancer Genomics Research Laboratory, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD; Departments of Medicine, Pediatrics, and Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY; Section of Hematology, Oncology and Stem Cell Transplantation, Department of Pediatrics, University of Chicago, Chicago, IL; Nationwide Children's Hospital and the Ohio State University School of Medicine, Columbus, OH; Department of Pediatrics, University of Minnesota, Minneapolis, MN; Information Management Services, Inc., Calverton, MD; Cancer Prevention and Clinical Statistics Programs and Cancer Prevention Program, Fred Hutchinson Cancer Research Center, Seattle, WA; Department of Genetics and Department of Radiation Physics, The University of Texas at MD Anderson Cancer Center, Houston, TX; Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH; Institute for Cancer Outcomes and Survivorship, University of Alabama at Birmingham, Birmingham, AL
| | - Diana M Merino
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, Bethesda, MD; Department of Epidemiology and Cancer Control, Division of Cancer Survivorship, Department of Oncology, Department of Biostatistics, Hartwell Center for Bioinformatics and Biotechnology, and Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN; Cancer Genomics Research Laboratory, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD; Departments of Medicine, Pediatrics, and Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY; Section of Hematology, Oncology and Stem Cell Transplantation, Department of Pediatrics, University of Chicago, Chicago, IL; Nationwide Children's Hospital and the Ohio State University School of Medicine, Columbus, OH; Department of Pediatrics, University of Minnesota, Minneapolis, MN; Information Management Services, Inc., Calverton, MD; Cancer Prevention and Clinical Statistics Programs and Cancer Prevention Program, Fred Hutchinson Cancer Research Center, Seattle, WA; Department of Genetics and Department of Radiation Physics, The University of Texas at MD Anderson Cancer Center, Houston, TX; Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH; Institute for Cancer Outcomes and Survivorship, University of Alabama at Birmingham, Birmingham, AL
| | - Jeannette R Wong
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, Bethesda, MD; Department of Epidemiology and Cancer Control, Division of Cancer Survivorship, Department of Oncology, Department of Biostatistics, Hartwell Center for Bioinformatics and Biotechnology, and Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN; Cancer Genomics Research Laboratory, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD; Departments of Medicine, Pediatrics, and Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY; Section of Hematology, Oncology and Stem Cell Transplantation, Department of Pediatrics, University of Chicago, Chicago, IL; Nationwide Children's Hospital and the Ohio State University School of Medicine, Columbus, OH; Department of Pediatrics, University of Minnesota, Minneapolis, MN; Information Management Services, Inc., Calverton, MD; Cancer Prevention and Clinical Statistics Programs and Cancer Prevention Program, Fred Hutchinson Cancer Research Center, Seattle, WA; Department of Genetics and Department of Radiation Physics, The University of Texas at MD Anderson Cancer Center, Houston, TX; Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH; Institute for Cancer Outcomes and Survivorship, University of Alabama at Birmingham, Birmingham, AL
| | - Sue Hammond
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, Bethesda, MD; Department of Epidemiology and Cancer Control, Division of Cancer Survivorship, Department of Oncology, Department of Biostatistics, Hartwell Center for Bioinformatics and Biotechnology, and Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN; Cancer Genomics Research Laboratory, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD; Departments of Medicine, Pediatrics, and Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY; Section of Hematology, Oncology and Stem Cell Transplantation, Department of Pediatrics, University of Chicago, Chicago, IL; Nationwide Children's Hospital and the Ohio State University School of Medicine, Columbus, OH; Department of Pediatrics, University of Minnesota, Minneapolis, MN; Information Management Services, Inc., Calverton, MD; Cancer Prevention and Clinical Statistics Programs and Cancer Prevention Program, Fred Hutchinson Cancer Research Center, Seattle, WA; Department of Genetics and Department of Radiation Physics, The University of Texas at MD Anderson Cancer Center, Houston, TX; Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH; Institute for Cancer Outcomes and Survivorship, University of Alabama at Birmingham, Birmingham, AL
| | - Joseph P Neglia
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, Bethesda, MD; Department of Epidemiology and Cancer Control, Division of Cancer Survivorship, Department of Oncology, Department of Biostatistics, Hartwell Center for Bioinformatics and Biotechnology, and Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN; Cancer Genomics Research Laboratory, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD; Departments of Medicine, Pediatrics, and Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY; Section of Hematology, Oncology and Stem Cell Transplantation, Department of Pediatrics, University of Chicago, Chicago, IL; Nationwide Children's Hospital and the Ohio State University School of Medicine, Columbus, OH; Department of Pediatrics, University of Minnesota, Minneapolis, MN; Information Management Services, Inc., Calverton, MD; Cancer Prevention and Clinical Statistics Programs and Cancer Prevention Program, Fred Hutchinson Cancer Research Center, Seattle, WA; Department of Genetics and Department of Radiation Physics, The University of Texas at MD Anderson Cancer Center, Houston, TX; Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH; Institute for Cancer Outcomes and Survivorship, University of Alabama at Birmingham, Birmingham, AL
| | - Lucie M Turcotte
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, Bethesda, MD; Department of Epidemiology and Cancer Control, Division of Cancer Survivorship, Department of Oncology, Department of Biostatistics, Hartwell Center for Bioinformatics and Biotechnology, and Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN; Cancer Genomics Research Laboratory, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD; Departments of Medicine, Pediatrics, and Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY; Section of Hematology, Oncology and Stem Cell Transplantation, Department of Pediatrics, University of Chicago, Chicago, IL; Nationwide Children's Hospital and the Ohio State University School of Medicine, Columbus, OH; Department of Pediatrics, University of Minnesota, Minneapolis, MN; Information Management Services, Inc., Calverton, MD; Cancer Prevention and Clinical Statistics Programs and Cancer Prevention Program, Fred Hutchinson Cancer Research Center, Seattle, WA; Department of Genetics and Department of Radiation Physics, The University of Texas at MD Anderson Cancer Center, Houston, TX; Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH; Institute for Cancer Outcomes and Survivorship, University of Alabama at Birmingham, Birmingham, AL
| | - Jeremy Miller
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, Bethesda, MD; Department of Epidemiology and Cancer Control, Division of Cancer Survivorship, Department of Oncology, Department of Biostatistics, Hartwell Center for Bioinformatics and Biotechnology, and Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN; Cancer Genomics Research Laboratory, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD; Departments of Medicine, Pediatrics, and Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY; Section of Hematology, Oncology and Stem Cell Transplantation, Department of Pediatrics, University of Chicago, Chicago, IL; Nationwide Children's Hospital and the Ohio State University School of Medicine, Columbus, OH; Department of Pediatrics, University of Minnesota, Minneapolis, MN; Information Management Services, Inc., Calverton, MD; Cancer Prevention and Clinical Statistics Programs and Cancer Prevention Program, Fred Hutchinson Cancer Research Center, Seattle, WA; Department of Genetics and Department of Radiation Physics, The University of Texas at MD Anderson Cancer Center, Houston, TX; Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH; Institute for Cancer Outcomes and Survivorship, University of Alabama at Birmingham, Birmingham, AL
| | - Laura Bowen
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, Bethesda, MD; Department of Epidemiology and Cancer Control, Division of Cancer Survivorship, Department of Oncology, Department of Biostatistics, Hartwell Center for Bioinformatics and Biotechnology, and Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN; Cancer Genomics Research Laboratory, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD; Departments of Medicine, Pediatrics, and Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY; Section of Hematology, Oncology and Stem Cell Transplantation, Department of Pediatrics, University of Chicago, Chicago, IL; Nationwide Children's Hospital and the Ohio State University School of Medicine, Columbus, OH; Department of Pediatrics, University of Minnesota, Minneapolis, MN; Information Management Services, Inc., Calverton, MD; Cancer Prevention and Clinical Statistics Programs and Cancer Prevention Program, Fred Hutchinson Cancer Research Center, Seattle, WA; Department of Genetics and Department of Radiation Physics, The University of Texas at MD Anderson Cancer Center, Houston, TX; Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH; Institute for Cancer Outcomes and Survivorship, University of Alabama at Birmingham, Birmingham, AL
| | - William A Wheeler
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, Bethesda, MD; Department of Epidemiology and Cancer Control, Division of Cancer Survivorship, Department of Oncology, Department of Biostatistics, Hartwell Center for Bioinformatics and Biotechnology, and Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN; Cancer Genomics Research Laboratory, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD; Departments of Medicine, Pediatrics, and Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY; Section of Hematology, Oncology and Stem Cell Transplantation, Department of Pediatrics, University of Chicago, Chicago, IL; Nationwide Children's Hospital and the Ohio State University School of Medicine, Columbus, OH; Department of Pediatrics, University of Minnesota, Minneapolis, MN; Information Management Services, Inc., Calverton, MD; Cancer Prevention and Clinical Statistics Programs and Cancer Prevention Program, Fred Hutchinson Cancer Research Center, Seattle, WA; Department of Genetics and Department of Radiation Physics, The University of Texas at MD Anderson Cancer Center, Houston, TX; Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH; Institute for Cancer Outcomes and Survivorship, University of Alabama at Birmingham, Birmingham, AL
| | - Wendy M Leisenring
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, Bethesda, MD; Department of Epidemiology and Cancer Control, Division of Cancer Survivorship, Department of Oncology, Department of Biostatistics, Hartwell Center for Bioinformatics and Biotechnology, and Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN; Cancer Genomics Research Laboratory, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD; Departments of Medicine, Pediatrics, and Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY; Section of Hematology, Oncology and Stem Cell Transplantation, Department of Pediatrics, University of Chicago, Chicago, IL; Nationwide Children's Hospital and the Ohio State University School of Medicine, Columbus, OH; Department of Pediatrics, University of Minnesota, Minneapolis, MN; Information Management Services, Inc., Calverton, MD; Cancer Prevention and Clinical Statistics Programs and Cancer Prevention Program, Fred Hutchinson Cancer Research Center, Seattle, WA; Department of Genetics and Department of Radiation Physics, The University of Texas at MD Anderson Cancer Center, Houston, TX; Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH; Institute for Cancer Outcomes and Survivorship, University of Alabama at Birmingham, Birmingham, AL
| | - John A Whitton
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, Bethesda, MD; Department of Epidemiology and Cancer Control, Division of Cancer Survivorship, Department of Oncology, Department of Biostatistics, Hartwell Center for Bioinformatics and Biotechnology, and Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN; Cancer Genomics Research Laboratory, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD; Departments of Medicine, Pediatrics, and Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY; Section of Hematology, Oncology and Stem Cell Transplantation, Department of Pediatrics, University of Chicago, Chicago, IL; Nationwide Children's Hospital and the Ohio State University School of Medicine, Columbus, OH; Department of Pediatrics, University of Minnesota, Minneapolis, MN; Information Management Services, Inc., Calverton, MD; Cancer Prevention and Clinical Statistics Programs and Cancer Prevention Program, Fred Hutchinson Cancer Research Center, Seattle, WA; Department of Genetics and Department of Radiation Physics, The University of Texas at MD Anderson Cancer Center, Houston, TX; Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH; Institute for Cancer Outcomes and Survivorship, University of Alabama at Birmingham, Birmingham, AL
| | - Laurie Burdette
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, Bethesda, MD; Department of Epidemiology and Cancer Control, Division of Cancer Survivorship, Department of Oncology, Department of Biostatistics, Hartwell Center for Bioinformatics and Biotechnology, and Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN; Cancer Genomics Research Laboratory, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD; Departments of Medicine, Pediatrics, and Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY; Section of Hematology, Oncology and Stem Cell Transplantation, Department of Pediatrics, University of Chicago, Chicago, IL; Nationwide Children's Hospital and the Ohio State University School of Medicine, Columbus, OH; Department of Pediatrics, University of Minnesota, Minneapolis, MN; Information Management Services, Inc., Calverton, MD; Cancer Prevention and Clinical Statistics Programs and Cancer Prevention Program, Fred Hutchinson Cancer Research Center, Seattle, WA; Department of Genetics and Department of Radiation Physics, The University of Texas at MD Anderson Cancer Center, Houston, TX; Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH; Institute for Cancer Outcomes and Survivorship, University of Alabama at Birmingham, Birmingham, AL
| | - Charles Chung
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, Bethesda, MD; Department of Epidemiology and Cancer Control, Division of Cancer Survivorship, Department of Oncology, Department of Biostatistics, Hartwell Center for Bioinformatics and Biotechnology, and Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN; Cancer Genomics Research Laboratory, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD; Departments of Medicine, Pediatrics, and Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY; Section of Hematology, Oncology and Stem Cell Transplantation, Department of Pediatrics, University of Chicago, Chicago, IL; Nationwide Children's Hospital and the Ohio State University School of Medicine, Columbus, OH; Department of Pediatrics, University of Minnesota, Minneapolis, MN; Information Management Services, Inc., Calverton, MD; Cancer Prevention and Clinical Statistics Programs and Cancer Prevention Program, Fred Hutchinson Cancer Research Center, Seattle, WA; Department of Genetics and Department of Radiation Physics, The University of Texas at MD Anderson Cancer Center, Houston, TX; Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH; Institute for Cancer Outcomes and Survivorship, University of Alabama at Birmingham, Birmingham, AL
| | - Belynda D Hicks
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, Bethesda, MD; Department of Epidemiology and Cancer Control, Division of Cancer Survivorship, Department of Oncology, Department of Biostatistics, Hartwell Center for Bioinformatics and Biotechnology, and Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN; Cancer Genomics Research Laboratory, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD; Departments of Medicine, Pediatrics, and Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY; Section of Hematology, Oncology and Stem Cell Transplantation, Department of Pediatrics, University of Chicago, Chicago, IL; Nationwide Children's Hospital and the Ohio State University School of Medicine, Columbus, OH; Department of Pediatrics, University of Minnesota, Minneapolis, MN; Information Management Services, Inc., Calverton, MD; Cancer Prevention and Clinical Statistics Programs and Cancer Prevention Program, Fred Hutchinson Cancer Research Center, Seattle, WA; Department of Genetics and Department of Radiation Physics, The University of Texas at MD Anderson Cancer Center, Houston, TX; Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH; Institute for Cancer Outcomes and Survivorship, University of Alabama at Birmingham, Birmingham, AL
| | - Kristine Jones
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, Bethesda, MD; Department of Epidemiology and Cancer Control, Division of Cancer Survivorship, Department of Oncology, Department of Biostatistics, Hartwell Center for Bioinformatics and Biotechnology, and Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN; Cancer Genomics Research Laboratory, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD; Departments of Medicine, Pediatrics, and Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY; Section of Hematology, Oncology and Stem Cell Transplantation, Department of Pediatrics, University of Chicago, Chicago, IL; Nationwide Children's Hospital and the Ohio State University School of Medicine, Columbus, OH; Department of Pediatrics, University of Minnesota, Minneapolis, MN; Information Management Services, Inc., Calverton, MD; Cancer Prevention and Clinical Statistics Programs and Cancer Prevention Program, Fred Hutchinson Cancer Research Center, Seattle, WA; Department of Genetics and Department of Radiation Physics, The University of Texas at MD Anderson Cancer Center, Houston, TX; Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH; Institute for Cancer Outcomes and Survivorship, University of Alabama at Birmingham, Birmingham, AL
| | - Mitchell J Machiela
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, Bethesda, MD; Department of Epidemiology and Cancer Control, Division of Cancer Survivorship, Department of Oncology, Department of Biostatistics, Hartwell Center for Bioinformatics and Biotechnology, and Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN; Cancer Genomics Research Laboratory, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD; Departments of Medicine, Pediatrics, and Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY; Section of Hematology, Oncology and Stem Cell Transplantation, Department of Pediatrics, University of Chicago, Chicago, IL; Nationwide Children's Hospital and the Ohio State University School of Medicine, Columbus, OH; Department of Pediatrics, University of Minnesota, Minneapolis, MN; Information Management Services, Inc., Calverton, MD; Cancer Prevention and Clinical Statistics Programs and Cancer Prevention Program, Fred Hutchinson Cancer Research Center, Seattle, WA; Department of Genetics and Department of Radiation Physics, The University of Texas at MD Anderson Cancer Center, Houston, TX; Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH; Institute for Cancer Outcomes and Survivorship, University of Alabama at Birmingham, Birmingham, AL
| | - Aurelie Vogt
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, Bethesda, MD; Department of Epidemiology and Cancer Control, Division of Cancer Survivorship, Department of Oncology, Department of Biostatistics, Hartwell Center for Bioinformatics and Biotechnology, and Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN; Cancer Genomics Research Laboratory, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD; Departments of Medicine, Pediatrics, and Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY; Section of Hematology, Oncology and Stem Cell Transplantation, Department of Pediatrics, University of Chicago, Chicago, IL; Nationwide Children's Hospital and the Ohio State University School of Medicine, Columbus, OH; Department of Pediatrics, University of Minnesota, Minneapolis, MN; Information Management Services, Inc., Calverton, MD; Cancer Prevention and Clinical Statistics Programs and Cancer Prevention Program, Fred Hutchinson Cancer Research Center, Seattle, WA; Department of Genetics and Department of Radiation Physics, The University of Texas at MD Anderson Cancer Center, Houston, TX; Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH; Institute for Cancer Outcomes and Survivorship, University of Alabama at Birmingham, Birmingham, AL
| | - Zhaoming Wang
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, Bethesda, MD; Department of Epidemiology and Cancer Control, Division of Cancer Survivorship, Department of Oncology, Department of Biostatistics, Hartwell Center for Bioinformatics and Biotechnology, and Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN; Cancer Genomics Research Laboratory, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD; Departments of Medicine, Pediatrics, and Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY; Section of Hematology, Oncology and Stem Cell Transplantation, Department of Pediatrics, University of Chicago, Chicago, IL; Nationwide Children's Hospital and the Ohio State University School of Medicine, Columbus, OH; Department of Pediatrics, University of Minnesota, Minneapolis, MN; Information Management Services, Inc., Calverton, MD; Cancer Prevention and Clinical Statistics Programs and Cancer Prevention Program, Fred Hutchinson Cancer Research Center, Seattle, WA; Department of Genetics and Department of Radiation Physics, The University of Texas at MD Anderson Cancer Center, Houston, TX; Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH; Institute for Cancer Outcomes and Survivorship, University of Alabama at Birmingham, Birmingham, AL
| | - Meredith Yeager
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, Bethesda, MD; Department of Epidemiology and Cancer Control, Division of Cancer Survivorship, Department of Oncology, Department of Biostatistics, Hartwell Center for Bioinformatics and Biotechnology, and Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN; Cancer Genomics Research Laboratory, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD; Departments of Medicine, Pediatrics, and Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY; Section of Hematology, Oncology and Stem Cell Transplantation, Department of Pediatrics, University of Chicago, Chicago, IL; Nationwide Children's Hospital and the Ohio State University School of Medicine, Columbus, OH; Department of Pediatrics, University of Minnesota, Minneapolis, MN; Information Management Services, Inc., Calverton, MD; Cancer Prevention and Clinical Statistics Programs and Cancer Prevention Program, Fred Hutchinson Cancer Research Center, Seattle, WA; Department of Genetics and Department of Radiation Physics, The University of Texas at MD Anderson Cancer Center, Houston, TX; Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH; Institute for Cancer Outcomes and Survivorship, University of Alabama at Birmingham, Birmingham, AL
| | - Geoffrey Neale
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, Bethesda, MD; Department of Epidemiology and Cancer Control, Division of Cancer Survivorship, Department of Oncology, Department of Biostatistics, Hartwell Center for Bioinformatics and Biotechnology, and Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN; Cancer Genomics Research Laboratory, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD; Departments of Medicine, Pediatrics, and Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY; Section of Hematology, Oncology and Stem Cell Transplantation, Department of Pediatrics, University of Chicago, Chicago, IL; Nationwide Children's Hospital and the Ohio State University School of Medicine, Columbus, OH; Department of Pediatrics, University of Minnesota, Minneapolis, MN; Information Management Services, Inc., Calverton, MD; Cancer Prevention and Clinical Statistics Programs and Cancer Prevention Program, Fred Hutchinson Cancer Research Center, Seattle, WA; Department of Genetics and Department of Radiation Physics, The University of Texas at MD Anderson Cancer Center, Houston, TX; Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH; Institute for Cancer Outcomes and Survivorship, University of Alabama at Birmingham, Birmingham, AL
| | - Matthew Lear
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, Bethesda, MD; Department of Epidemiology and Cancer Control, Division of Cancer Survivorship, Department of Oncology, Department of Biostatistics, Hartwell Center for Bioinformatics and Biotechnology, and Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN; Cancer Genomics Research Laboratory, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD; Departments of Medicine, Pediatrics, and Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY; Section of Hematology, Oncology and Stem Cell Transplantation, Department of Pediatrics, University of Chicago, Chicago, IL; Nationwide Children's Hospital and the Ohio State University School of Medicine, Columbus, OH; Department of Pediatrics, University of Minnesota, Minneapolis, MN; Information Management Services, Inc., Calverton, MD; Cancer Prevention and Clinical Statistics Programs and Cancer Prevention Program, Fred Hutchinson Cancer Research Center, Seattle, WA; Department of Genetics and Department of Radiation Physics, The University of Texas at MD Anderson Cancer Center, Houston, TX; Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH; Institute for Cancer Outcomes and Survivorship, University of Alabama at Birmingham, Birmingham, AL
| | - Louise C Strong
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, Bethesda, MD; Department of Epidemiology and Cancer Control, Division of Cancer Survivorship, Department of Oncology, Department of Biostatistics, Hartwell Center for Bioinformatics and Biotechnology, and Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN; Cancer Genomics Research Laboratory, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD; Departments of Medicine, Pediatrics, and Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY; Section of Hematology, Oncology and Stem Cell Transplantation, Department of Pediatrics, University of Chicago, Chicago, IL; Nationwide Children's Hospital and the Ohio State University School of Medicine, Columbus, OH; Department of Pediatrics, University of Minnesota, Minneapolis, MN; Information Management Services, Inc., Calverton, MD; Cancer Prevention and Clinical Statistics Programs and Cancer Prevention Program, Fred Hutchinson Cancer Research Center, Seattle, WA; Department of Genetics and Department of Radiation Physics, The University of Texas at MD Anderson Cancer Center, Houston, TX; Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH; Institute for Cancer Outcomes and Survivorship, University of Alabama at Birmingham, Birmingham, AL
| | - Yutaka Yasui
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, Bethesda, MD; Department of Epidemiology and Cancer Control, Division of Cancer Survivorship, Department of Oncology, Department of Biostatistics, Hartwell Center for Bioinformatics and Biotechnology, and Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN; Cancer Genomics Research Laboratory, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD; Departments of Medicine, Pediatrics, and Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY; Section of Hematology, Oncology and Stem Cell Transplantation, Department of Pediatrics, University of Chicago, Chicago, IL; Nationwide Children's Hospital and the Ohio State University School of Medicine, Columbus, OH; Department of Pediatrics, University of Minnesota, Minneapolis, MN; Information Management Services, Inc., Calverton, MD; Cancer Prevention and Clinical Statistics Programs and Cancer Prevention Program, Fred Hutchinson Cancer Research Center, Seattle, WA; Department of Genetics and Department of Radiation Physics, The University of Texas at MD Anderson Cancer Center, Houston, TX; Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH; Institute for Cancer Outcomes and Survivorship, University of Alabama at Birmingham, Birmingham, AL
| | - Marilyn Stovall
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, Bethesda, MD; Department of Epidemiology and Cancer Control, Division of Cancer Survivorship, Department of Oncology, Department of Biostatistics, Hartwell Center for Bioinformatics and Biotechnology, and Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN; Cancer Genomics Research Laboratory, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD; Departments of Medicine, Pediatrics, and Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY; Section of Hematology, Oncology and Stem Cell Transplantation, Department of Pediatrics, University of Chicago, Chicago, IL; Nationwide Children's Hospital and the Ohio State University School of Medicine, Columbus, OH; Department of Pediatrics, University of Minnesota, Minneapolis, MN; Information Management Services, Inc., Calverton, MD; Cancer Prevention and Clinical Statistics Programs and Cancer Prevention Program, Fred Hutchinson Cancer Research Center, Seattle, WA; Department of Genetics and Department of Radiation Physics, The University of Texas at MD Anderson Cancer Center, Houston, TX; Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH; Institute for Cancer Outcomes and Survivorship, University of Alabama at Birmingham, Birmingham, AL
| | - Rita E Weathers
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, Bethesda, MD; Department of Epidemiology and Cancer Control, Division of Cancer Survivorship, Department of Oncology, Department of Biostatistics, Hartwell Center for Bioinformatics and Biotechnology, and Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN; Cancer Genomics Research Laboratory, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD; Departments of Medicine, Pediatrics, and Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY; Section of Hematology, Oncology and Stem Cell Transplantation, Department of Pediatrics, University of Chicago, Chicago, IL; Nationwide Children's Hospital and the Ohio State University School of Medicine, Columbus, OH; Department of Pediatrics, University of Minnesota, Minneapolis, MN; Information Management Services, Inc., Calverton, MD; Cancer Prevention and Clinical Statistics Programs and Cancer Prevention Program, Fred Hutchinson Cancer Research Center, Seattle, WA; Department of Genetics and Department of Radiation Physics, The University of Texas at MD Anderson Cancer Center, Houston, TX; Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH; Institute for Cancer Outcomes and Survivorship, University of Alabama at Birmingham, Birmingham, AL
| | - Susan A Smith
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, Bethesda, MD; Department of Epidemiology and Cancer Control, Division of Cancer Survivorship, Department of Oncology, Department of Biostatistics, Hartwell Center for Bioinformatics and Biotechnology, and Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN; Cancer Genomics Research Laboratory, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD; Departments of Medicine, Pediatrics, and Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY; Section of Hematology, Oncology and Stem Cell Transplantation, Department of Pediatrics, University of Chicago, Chicago, IL; Nationwide Children's Hospital and the Ohio State University School of Medicine, Columbus, OH; Department of Pediatrics, University of Minnesota, Minneapolis, MN; Information Management Services, Inc., Calverton, MD; Cancer Prevention and Clinical Statistics Programs and Cancer Prevention Program, Fred Hutchinson Cancer Research Center, Seattle, WA; Department of Genetics and Department of Radiation Physics, The University of Texas at MD Anderson Cancer Center, Houston, TX; Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH; Institute for Cancer Outcomes and Survivorship, University of Alabama at Birmingham, Birmingham, AL
| | - Rebecca Howell
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, Bethesda, MD; Department of Epidemiology and Cancer Control, Division of Cancer Survivorship, Department of Oncology, Department of Biostatistics, Hartwell Center for Bioinformatics and Biotechnology, and Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN; Cancer Genomics Research Laboratory, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD; Departments of Medicine, Pediatrics, and Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY; Section of Hematology, Oncology and Stem Cell Transplantation, Department of Pediatrics, University of Chicago, Chicago, IL; Nationwide Children's Hospital and the Ohio State University School of Medicine, Columbus, OH; Department of Pediatrics, University of Minnesota, Minneapolis, MN; Information Management Services, Inc., Calverton, MD; Cancer Prevention and Clinical Statistics Programs and Cancer Prevention Program, Fred Hutchinson Cancer Research Center, Seattle, WA; Department of Genetics and Department of Radiation Physics, The University of Texas at MD Anderson Cancer Center, Houston, TX; Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH; Institute for Cancer Outcomes and Survivorship, University of Alabama at Birmingham, Birmingham, AL
| | - Stella M Davies
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, Bethesda, MD; Department of Epidemiology and Cancer Control, Division of Cancer Survivorship, Department of Oncology, Department of Biostatistics, Hartwell Center for Bioinformatics and Biotechnology, and Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN; Cancer Genomics Research Laboratory, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD; Departments of Medicine, Pediatrics, and Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY; Section of Hematology, Oncology and Stem Cell Transplantation, Department of Pediatrics, University of Chicago, Chicago, IL; Nationwide Children's Hospital and the Ohio State University School of Medicine, Columbus, OH; Department of Pediatrics, University of Minnesota, Minneapolis, MN; Information Management Services, Inc., Calverton, MD; Cancer Prevention and Clinical Statistics Programs and Cancer Prevention Program, Fred Hutchinson Cancer Research Center, Seattle, WA; Department of Genetics and Department of Radiation Physics, The University of Texas at MD Anderson Cancer Center, Houston, TX; Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH; Institute for Cancer Outcomes and Survivorship, University of Alabama at Birmingham, Birmingham, AL
| | - Gretchen A Radloff
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, Bethesda, MD; Department of Epidemiology and Cancer Control, Division of Cancer Survivorship, Department of Oncology, Department of Biostatistics, Hartwell Center for Bioinformatics and Biotechnology, and Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN; Cancer Genomics Research Laboratory, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD; Departments of Medicine, Pediatrics, and Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY; Section of Hematology, Oncology and Stem Cell Transplantation, Department of Pediatrics, University of Chicago, Chicago, IL; Nationwide Children's Hospital and the Ohio State University School of Medicine, Columbus, OH; Department of Pediatrics, University of Minnesota, Minneapolis, MN; Information Management Services, Inc., Calverton, MD; Cancer Prevention and Clinical Statistics Programs and Cancer Prevention Program, Fred Hutchinson Cancer Research Center, Seattle, WA; Department of Genetics and Department of Radiation Physics, The University of Texas at MD Anderson Cancer Center, Houston, TX; Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH; Institute for Cancer Outcomes and Survivorship, University of Alabama at Birmingham, Birmingham, AL
| | - Kenan Onel
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, Bethesda, MD; Department of Epidemiology and Cancer Control, Division of Cancer Survivorship, Department of Oncology, Department of Biostatistics, Hartwell Center for Bioinformatics and Biotechnology, and Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN; Cancer Genomics Research Laboratory, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD; Departments of Medicine, Pediatrics, and Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY; Section of Hematology, Oncology and Stem Cell Transplantation, Department of Pediatrics, University of Chicago, Chicago, IL; Nationwide Children's Hospital and the Ohio State University School of Medicine, Columbus, OH; Department of Pediatrics, University of Minnesota, Minneapolis, MN; Information Management Services, Inc., Calverton, MD; Cancer Prevention and Clinical Statistics Programs and Cancer Prevention Program, Fred Hutchinson Cancer Research Center, Seattle, WA; Department of Genetics and Department of Radiation Physics, The University of Texas at MD Anderson Cancer Center, Houston, TX; Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH; Institute for Cancer Outcomes and Survivorship, University of Alabama at Birmingham, Birmingham, AL
| | - Amy Berrington de González
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, Bethesda, MD; Department of Epidemiology and Cancer Control, Division of Cancer Survivorship, Department of Oncology, Department of Biostatistics, Hartwell Center for Bioinformatics and Biotechnology, and Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN; Cancer Genomics Research Laboratory, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD; Departments of Medicine, Pediatrics, and Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY; Section of Hematology, Oncology and Stem Cell Transplantation, Department of Pediatrics, University of Chicago, Chicago, IL; Nationwide Children's Hospital and the Ohio State University School of Medicine, Columbus, OH; Department of Pediatrics, University of Minnesota, Minneapolis, MN; Information Management Services, Inc., Calverton, MD; Cancer Prevention and Clinical Statistics Programs and Cancer Prevention Program, Fred Hutchinson Cancer Research Center, Seattle, WA; Department of Genetics and Department of Radiation Physics, The University of Texas at MD Anderson Cancer Center, Houston, TX; Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH; Institute for Cancer Outcomes and Survivorship, University of Alabama at Birmingham, Birmingham, AL
| | - Peter D Inskip
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, Bethesda, MD; Department of Epidemiology and Cancer Control, Division of Cancer Survivorship, Department of Oncology, Department of Biostatistics, Hartwell Center for Bioinformatics and Biotechnology, and Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN; Cancer Genomics Research Laboratory, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD; Departments of Medicine, Pediatrics, and Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY; Section of Hematology, Oncology and Stem Cell Transplantation, Department of Pediatrics, University of Chicago, Chicago, IL; Nationwide Children's Hospital and the Ohio State University School of Medicine, Columbus, OH; Department of Pediatrics, University of Minnesota, Minneapolis, MN; Information Management Services, Inc., Calverton, MD; Cancer Prevention and Clinical Statistics Programs and Cancer Prevention Program, Fred Hutchinson Cancer Research Center, Seattle, WA; Department of Genetics and Department of Radiation Physics, The University of Texas at MD Anderson Cancer Center, Houston, TX; Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH; Institute for Cancer Outcomes and Survivorship, University of Alabama at Birmingham, Birmingham, AL
| | - Preetha Rajaraman
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, Bethesda, MD; Department of Epidemiology and Cancer Control, Division of Cancer Survivorship, Department of Oncology, Department of Biostatistics, Hartwell Center for Bioinformatics and Biotechnology, and Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN; Cancer Genomics Research Laboratory, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD; Departments of Medicine, Pediatrics, and Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY; Section of Hematology, Oncology and Stem Cell Transplantation, Department of Pediatrics, University of Chicago, Chicago, IL; Nationwide Children's Hospital and the Ohio State University School of Medicine, Columbus, OH; Department of Pediatrics, University of Minnesota, Minneapolis, MN; Information Management Services, Inc., Calverton, MD; Cancer Prevention and Clinical Statistics Programs and Cancer Prevention Program, Fred Hutchinson Cancer Research Center, Seattle, WA; Department of Genetics and Department of Radiation Physics, The University of Texas at MD Anderson Cancer Center, Houston, TX; Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH; Institute for Cancer Outcomes and Survivorship, University of Alabama at Birmingham, Birmingham, AL
| | - Joseph F Fraumeni
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, Bethesda, MD; Department of Epidemiology and Cancer Control, Division of Cancer Survivorship, Department of Oncology, Department of Biostatistics, Hartwell Center for Bioinformatics and Biotechnology, and Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN; Cancer Genomics Research Laboratory, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD; Departments of Medicine, Pediatrics, and Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY; Section of Hematology, Oncology and Stem Cell Transplantation, Department of Pediatrics, University of Chicago, Chicago, IL; Nationwide Children's Hospital and the Ohio State University School of Medicine, Columbus, OH; Department of Pediatrics, University of Minnesota, Minneapolis, MN; Information Management Services, Inc., Calverton, MD; Cancer Prevention and Clinical Statistics Programs and Cancer Prevention Program, Fred Hutchinson Cancer Research Center, Seattle, WA; Department of Genetics and Department of Radiation Physics, The University of Texas at MD Anderson Cancer Center, Houston, TX; Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH; Institute for Cancer Outcomes and Survivorship, University of Alabama at Birmingham, Birmingham, AL
| | - Smita Bhatia
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, Bethesda, MD; Department of Epidemiology and Cancer Control, Division of Cancer Survivorship, Department of Oncology, Department of Biostatistics, Hartwell Center for Bioinformatics and Biotechnology, and Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN; Cancer Genomics Research Laboratory, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD; Departments of Medicine, Pediatrics, and Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY; Section of Hematology, Oncology and Stem Cell Transplantation, Department of Pediatrics, University of Chicago, Chicago, IL; Nationwide Children's Hospital and the Ohio State University School of Medicine, Columbus, OH; Department of Pediatrics, University of Minnesota, Minneapolis, MN; Information Management Services, Inc., Calverton, MD; Cancer Prevention and Clinical Statistics Programs and Cancer Prevention Program, Fred Hutchinson Cancer Research Center, Seattle, WA; Department of Genetics and Department of Radiation Physics, The University of Texas at MD Anderson Cancer Center, Houston, TX; Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH; Institute for Cancer Outcomes and Survivorship, University of Alabama at Birmingham, Birmingham, AL
| | - Stephen J Chanock
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, Bethesda, MD; Department of Epidemiology and Cancer Control, Division of Cancer Survivorship, Department of Oncology, Department of Biostatistics, Hartwell Center for Bioinformatics and Biotechnology, and Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN; Cancer Genomics Research Laboratory, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD; Departments of Medicine, Pediatrics, and Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY; Section of Hematology, Oncology and Stem Cell Transplantation, Department of Pediatrics, University of Chicago, Chicago, IL; Nationwide Children's Hospital and the Ohio State University School of Medicine, Columbus, OH; Department of Pediatrics, University of Minnesota, Minneapolis, MN; Information Management Services, Inc., Calverton, MD; Cancer Prevention and Clinical Statistics Programs and Cancer Prevention Program, Fred Hutchinson Cancer Research Center, Seattle, WA; Department of Genetics and Department of Radiation Physics, The University of Texas at MD Anderson Cancer Center, Houston, TX; Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH; Institute for Cancer Outcomes and Survivorship, University of Alabama at Birmingham, Birmingham, AL
| | - Margaret A Tucker
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, Bethesda, MD; Department of Epidemiology and Cancer Control, Division of Cancer Survivorship, Department of Oncology, Department of Biostatistics, Hartwell Center for Bioinformatics and Biotechnology, and Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN; Cancer Genomics Research Laboratory, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD; Departments of Medicine, Pediatrics, and Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY; Section of Hematology, Oncology and Stem Cell Transplantation, Department of Pediatrics, University of Chicago, Chicago, IL; Nationwide Children's Hospital and the Ohio State University School of Medicine, Columbus, OH; Department of Pediatrics, University of Minnesota, Minneapolis, MN; Information Management Services, Inc., Calverton, MD; Cancer Prevention and Clinical Statistics Programs and Cancer Prevention Program, Fred Hutchinson Cancer Research Center, Seattle, WA; Department of Genetics and Department of Radiation Physics, The University of Texas at MD Anderson Cancer Center, Houston, TX; Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH; Institute for Cancer Outcomes and Survivorship, University of Alabama at Birmingham, Birmingham, AL
| | - Leslie L Robison
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, Bethesda, MD; Department of Epidemiology and Cancer Control, Division of Cancer Survivorship, Department of Oncology, Department of Biostatistics, Hartwell Center for Bioinformatics and Biotechnology, and Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN; Cancer Genomics Research Laboratory, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD; Departments of Medicine, Pediatrics, and Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY; Section of Hematology, Oncology and Stem Cell Transplantation, Department of Pediatrics, University of Chicago, Chicago, IL; Nationwide Children's Hospital and the Ohio State University School of Medicine, Columbus, OH; Department of Pediatrics, University of Minnesota, Minneapolis, MN; Information Management Services, Inc., Calverton, MD; Cancer Prevention and Clinical Statistics Programs and Cancer Prevention Program, Fred Hutchinson Cancer Research Center, Seattle, WA; Department of Genetics and Department of Radiation Physics, The University of Texas at MD Anderson Cancer Center, Houston, TX; Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH; Institute for Cancer Outcomes and Survivorship, University of Alabama at Birmingham, Birmingham, AL
| |
Collapse
|
89
|
Gianazza E, Miller I, Guerrini U, Palazzolo L, Parravicini C, Eberini I. Gender proteomics II. Which proteins in sexual organs. J Proteomics 2017; 178:18-30. [PMID: 28988880 DOI: 10.1016/j.jprot.2017.10.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Revised: 09/26/2017] [Accepted: 10/04/2017] [Indexed: 02/08/2023]
Abstract
In continuity with the review dealing with differences by gender in non-sexual organs [1], this review collects data on the proteomes of the sexual organs as involved in human reproduction, under both physiological and pathological conditions. It also collects data on the tissue structures and biological fluids typical of pregnancy, such as placenta and amniotic fluid, as well as what may be tested on preimplantation embryos during medically assisted reproduction. The review includes as well mention to all fluids and secretions connected with sex organs and/or reproduction, including sperm and milk, to exemplify two distinctive items in male and female physiology. SIGNIFICANCE The causes of infertility are only incompletely understood; the same holds for the causes, and even the early markers, of the most frequent complications of pregnancy. To these established medical challenges, present day practice adds new issues connected with medically assisted reproduction. Omics approaches, including proteomics, are building the database for basic knowledge to possibly translate into clinical testing and eventually into medical routine in this critical branch of health care.
Collapse
Affiliation(s)
- Elisabetta Gianazza
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Via Balzaretti 9, I-20133 Milano, Italy.
| | - Ingrid Miller
- Institut für Medizinische Biochemie, Veterinärmedizinische Universität Wien, Veterinärplatz 1, A-1210 Wien, Austria
| | - Uliano Guerrini
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Via Balzaretti 9, I-20133 Milano, Italy
| | - Luca Palazzolo
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Via Balzaretti 9, I-20133 Milano, Italy
| | - Chiara Parravicini
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Via Balzaretti 9, I-20133 Milano, Italy
| | - Ivano Eberini
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Via Balzaretti 9, I-20133 Milano, Italy
| |
Collapse
|
90
|
Eritja N, Yeramian A, Chen BJ, Llobet-Navas D, Ortega E, Colas E, Abal M, Dolcet X, Reventos J, Matias-Guiu X. Endometrial Carcinoma: Specific Targeted Pathways. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 943:149-207. [PMID: 27910068 DOI: 10.1007/978-3-319-43139-0_6] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Endometrial cancer (EC) is the most common gynecologic malignancy in the western world with more than 280,000 cases per year worldwide. Prognosis for EC at early stages, when primary surgical resection is the most common initial treatment, is excellent. Five-year survival rate is around 70 %.Several molecular alterations have been described in the different types of EC. They occur in genes involved in important signaling pathways. In this chapter, we will review the most relevant altered pathways in EC, including PI3K/AKT/mTOR, RAS-RAF-MEK-ERK, Tyrosine kinase, WNT/β-Catenin, cell cycle, and TGF-β signaling pathways. At the end of the chapter, the most significant clinical trials will be briefly discussed.This information is important to identify specific targets for therapy.
Collapse
Affiliation(s)
- Nuria Eritja
- Department of Pathology and Molecular Genetics and Research Laboratory, Hospital Universitari Arnau de Vilanova, University of Lleida, IRBLLEIDA, Av Rovira Roure, 80, 25198, Lleida, Spain
- GEICEN Research Group, Department of Pathology and Molecular Genetics and Research Laboratory, Hospital Universitari Arnau de Vilanova, University of Lleida, IRBLLEIDA, Av Rovira Roure, 80, 25198, Lleida, Spain
| | - Andree Yeramian
- Department of Pathology and Molecular Genetics and Research Laboratory, Hospital Universitari Arnau de Vilanova, University of Lleida, IRBLLEIDA, Av Rovira Roure, 80, 25198, Lleida, Spain
- GEICEN Research Group, Department of Pathology and Molecular Genetics and Research Laboratory, Hospital Universitari Arnau de Vilanova, University of Lleida, IRBLLEIDA, Av Rovira Roure, 80, 25198, Lleida, Spain
| | - Bo-Juen Chen
- New York Genome Center, New York, NY, 10013, USA
| | - David Llobet-Navas
- Institute of Genetic Medicine, Newcastle University, Newcastle-Upon-Tyne, NE1 3BZ, UK
| | - Eugenia Ortega
- Department of Pathology and Molecular Genetics and Research Laboratory, Hospital Universitari Arnau de Vilanova, University of Lleida, IRBLLEIDA, Av Rovira Roure, 80, 25198, Lleida, Spain
| | - Eva Colas
- Department of Pathology and Molecular Genetics and Research Laboratory, Hospital Universitari Arnau de Vilanova, University of Lleida, IRBLLEIDA, Av Rovira Roure, 80, 25198, Lleida, Spain
- GEICEN Research Group, Department of Pathology and Molecular Genetics and Research Laboratory, Hospital Universitari Arnau de Vilanova, University of Lleida, IRBLLEIDA, Av Rovira Roure, 80, 25198, Lleida, Spain
- Research Unit in Biomedicine and Translational and Pediatric Oncology, Vall d'Hebron Research Institute, Barcelona, Spain
| | - Miguel Abal
- GEICEN Research Group, Department of Pathology and Molecular Genetics and Research Laboratory, Hospital Universitari Arnau de Vilanova, University of Lleida, IRBLLEIDA, Av Rovira Roure, 80, 25198, Lleida, Spain
- Translational Medical Oncology, Health Research Institute of Santiago (IDIS), Santiago de Compostela, Spain
| | - Xavier Dolcet
- Department of Pathology and Molecular Genetics and Research Laboratory, Hospital Universitari Arnau de Vilanova, University of Lleida, IRBLLEIDA, Av Rovira Roure, 80, 25198, Lleida, Spain
- GEICEN Research Group, Department of Pathology and Molecular Genetics and Research Laboratory, Hospital Universitari Arnau de Vilanova, University of Lleida, IRBLLEIDA, Av Rovira Roure, 80, 25198, Lleida, Spain
| | - Jaume Reventos
- GEICEN Research Group, Department of Pathology and Molecular Genetics and Research Laboratory, Hospital Universitari Arnau de Vilanova, University of Lleida, IRBLLEIDA, Av Rovira Roure, 80, 25198, Lleida, Spain
- Research Unit in Biomedicine and Translational and Pediatric Oncology, Vall d'Hebron Research Institute, Barcelona, Spain
| | - Xavier Matias-Guiu
- Department of Pathology and Molecular Genetics and Research Laboratory, Hospital Universitari Arnau de Vilanova, University of Lleida, IRBLLEIDA, Av Rovira Roure, 80, 25198, Lleida, Spain.
- GEICEN Research Group, Department of Pathology and Molecular Genetics and Research Laboratory, Hospital Universitari Arnau de Vilanova, University of Lleida, IRBLLEIDA, Av Rovira Roure, 80, 25198, Lleida, Spain.
| |
Collapse
|
91
|
Lee D, Yu EJ, Ham IH, Hur H, Kim YS. AKT inhibition is an effective treatment strategy in ARID1A-deficient gastric cancer cells. Onco Targets Ther 2017; 10:4153-4159. [PMID: 28860825 PMCID: PMC5574587 DOI: 10.2147/ott.s139664] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND The At-rich interactive domain 1A (ARID1A) is frequently mutated in gastric cancers (GCs) with a poor prognosis. Growing evidence indicates that loss of ARID1A expression leads to activation of the phosphatidylinositol 3-kinase (PI3K)/AKT pathway by AKT phosphorylation. We aim to investigate the different sensitivity for the AKT inhibitor in ARID1A-deficient GC cells. METHODS After transfection using siRNA or shRNA, the effect of ARID1A knockdown on the PI3K/AKT signaling pathway was evaluated by Western blot analysis. ARID1A-knockdown cells were treated with AKT inhibitor (GSK690693), 5-fluorouracil, or cisplatin, alone or in combination. Viability and apoptosis were analyzed using EZ-CYTOX cell viability assay and flow cytometry, respectively. RESULTS ARID1A depletion accelerated the phosphorylation of AKT and S6 in a dose-dependent manner and led to an increased proliferation of MKN-1, MKN-28, and KATO-III GC cells (P<0.001). ARID1A-deficient cells were more vulnerable to GSK690693 in comparison to the controls (P<0.001), even at very low doses. Flow cytometry confirmed the increased apoptosis in ARID1A-deficient cells treated with GSK690693 (0.01 μmol/L; P<0.001). In contrast to our expectations, ARID1A depletion did not cause resistance to 5-fluorouracil or cisplatin. Addition of GSK690693 to the conventional chemotherapy induced more decreased cell viability in ARID1A-knockdown cells (P<0.01). CONCLUSION Loss of ARID1A expression is a surrogate marker for the activation of the AKT signaling pathway and is also a reliable biomarker to predict the response for the AKT inhibitor. We anticipate that appropriate patient selection based on ARID1A expression in the tumor tissue will increase the drug sensitivity for the AKT inhibition and improve the clinical outcome.
Collapse
Affiliation(s)
- Dakeun Lee
- Department of Pathology, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Eun Ji Yu
- Department of Pathology, Ajou University School of Medicine, Suwon, Republic of Korea
| | - In-Hye Ham
- Department of Surgery, Ajou University School of Medicine, Suwon, Republic of Korea
- Brain Korea 21 Plus Research Center for Biomedical Sciences, Ajou University, Suwon, Republic of Korea
| | - Hoon Hur
- Department of Surgery, Ajou University School of Medicine, Suwon, Republic of Korea
- Brain Korea 21 Plus Research Center for Biomedical Sciences, Ajou University, Suwon, Republic of Korea
| | - You-Sun Kim
- Department of Biochemistry, Ajou University School of Medicine, Suwon, Republic of Korea
- Department of Biomedical Sciences, Graduate School, Ajou University, Suwon, Republic of Korea
| |
Collapse
|
92
|
Park J, Yoo HM, Jang W, Shin S, Kim M, Kim Y, Lee SW, Kim JG. Distribution of somatic mutations of cancer-related genes according to microsatellite instability status in Korean gastric cancer. Medicine (Baltimore) 2017; 96:e7224. [PMID: 28640116 PMCID: PMC5484224 DOI: 10.1097/md.0000000000007224] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Revised: 05/23/2017] [Accepted: 05/30/2017] [Indexed: 02/05/2023] Open
Abstract
In studies of the molecular basis of gastric cancer (GC), microsatellite instability (MSI) is one of the key factors. Somatic mutations found in GC are expected to contribute to MSI-high (H) tumorigenesis. We estimated somatic mutation distribution according to MSI status in 52 matched pair GC samples using the Ion Torrent Ion S5 XL with the AmpliSeq Cancer Hotspot panel.Seventy-five (9.8%) somatic variants consisting of 34 hotspot mutations and 41 other likely pathogenic variants were identified in 34 GC samples. The TP53 mutations was most common (35%, 26/75), followed by EGFR (8%, 6/75), HNF1A (8%, 6/75), PIK3CA (8%, 6/75), and ERBB2 (5%, 4/75). To determine MSI status, 52 matched pair samples were estimated using 15 MSI markers. Thirty-nine MS stable (S), 5 MSI-low (L), and 8 MSI-H were classified. GCs with MSI-H tended to have more variants significantly compared with GCs with MS stable (MSS) and MSI-L (standardized J-T statistic = 3.161 for number of variants; P = .002). The mean number of all variants and hotspot mutations per tumor samples only in GCs with MSI-H were 3.9 (range, 1-6) and 1.1 (range, 0-3), respectively. Whereas, the mean number of all variants and hotspot mutations per tumor samples only in GCs with MSS/MSI-L were 1 (0-5)/0.8 (0-1) and 0.5 (0-3)/0.8 (0-1), respectively.In conclusion, GC with MSI-H harbored more mutations in genes that act as a tumor suppressor or oncogene compared to GC with MSS/MSI-L. This finding suggests that the accumulation of MSIs contributes to the genetic diversity and complexities of GC. In addition, targeted NGS approach allows for detection of common and also rare clinically actionable mutations and profiles of comutations in multiple patients simultaneously. Because GC shows distinctive patterns related to ethnics, further studies pertaining to different racial/ethnic groups or cancer types may reinforce our investigations.
Collapse
Affiliation(s)
| | - Han Mo Yoo
- Division of Gastrointestinal Surgery, Department of Surgery
| | | | | | | | | | - Seung-Woo Lee
- Division of Gastroenterology, Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Jeong Goo Kim
- Division of Gastrointestinal Surgery, Department of Surgery
| |
Collapse
|
93
|
Sun C, Fang Y, Yin J, Chen J, Ju Z, Zhang D, Chen X, Vellano CP, Jeong KJ, Ng PKS, Eterovic AKB, Bhola NH, Lu Y, Westin SN, Grandis JR, Lin SY, Scott KL, Peng G, Brugge J, Mills GB. Rational combination therapy with PARP and MEK inhibitors capitalizes on therapeutic liabilities in RAS mutant cancers. Sci Transl Med 2017; 9:eaal5148. [PMID: 28566428 PMCID: PMC5919217 DOI: 10.1126/scitranslmed.aal5148] [Citation(s) in RCA: 174] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Accepted: 03/29/2017] [Indexed: 12/11/2022]
Abstract
Mutant RAS has remained recalcitrant to targeted therapy efforts. We demonstrate that combined treatment with poly(adenosine diphosphate-ribose) polymerase (PARP) inhibitors and mitogen-activated protein kinase (MAPK) kinase (MEK) inhibitors evokes unanticipated, synergistic cytotoxic effects in vitro and in vivo in multiple RAS mutant tumor models across tumor lineages where RAS mutations are prevalent. The effects of PARP and MEK inhibitor combinations are independent of BRCA1/2 and p53 mutation status, suggesting that the synergistic activity is likely to be generalizable. Synergistic activity of PARP and MEK inhibitor combinations in RAS mutant tumors is associated with (i) induction of BIM-mediated apoptosis, (ii) decrease in expression of components of the homologous recombination DNA repair pathway, (iii) decrease in homologous recombination DNA damage repair capacity, (iv) decrease in DNA damage checkpoint activity, (v) increase in PARP inhibitor-induced DNA damage, (vi) decrease in vascularity that could increase PARP inhibitor efficacy by inducing hypoxia, and (vii) elevated PARP1 protein, which increases trapping activity of PARP inhibitors. Mechanistically, enforced expression of FOXO3a, which is a target of the RAS/MAPK pathway, was sufficient to recapitulate the functional consequences of MEK inhibitors including synergy with PARP inhibitors. Thus, the ability of mutant RAS to suppress FOXO3a and its reversal by MEK inhibitors accounts, at least in part, for the synergy of PARP and MEK inhibitors in RAS mutant tumors. The rational combination of PARP and MEK inhibitors warrants clinical investigation in patients with RAS mutant tumors where there are few effective therapeutic options.
Collapse
Affiliation(s)
- Chaoyang Sun
- Department of Systems Biology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yong Fang
- Department of Systems Biology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jun Yin
- Department of Systems Biology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jian Chen
- Department of Systems Biology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Department of General Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang Province, Hangzhou 310000, China
| | - Zhenlin Ju
- Department of Bioinformatics and Computational Biology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Dong Zhang
- Department of Systems Biology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Xiaohua Chen
- Department of Systems Biology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Christopher P Vellano
- Department of Systems Biology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Kang Jin Jeong
- Department of Systems Biology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Patrick Kwok-Shing Ng
- Sheikh Khalifa Bin Zayed Al Nahyan Institute for Personalized Cancer Therapy, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Agda Karina B Eterovic
- Department of Systems Biology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Neil H Bhola
- Provost Office, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Yiling Lu
- Department of Systems Biology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Shannon N Westin
- Department of Gynecologic Oncology and Reproductive Medicine, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jennifer R Grandis
- Provost Office, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Shiaw-Yih Lin
- Department of Systems Biology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Kenneth L Scott
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Guang Peng
- Department of Clinical Cancer Prevention, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Joan Brugge
- Department of Cell Biology, Ludwig Center at Harvard, Harvard Medical School, Boston, MA 02115, USA
| | - Gordon B Mills
- Department of Systems Biology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
94
|
Lu H, Villafane N, Dogruluk T, Grzeskowiak CL, Kong K, Tsang YH, Zagorodna O, Pantazi A, Yang L, Neill NJ, Kim YW, Creighton CJ, Verhaak RG, Mills GB, Park PJ, Kucherlapati R, Scott KL. Engineering and Functional Characterization of Fusion Genes Identifies Novel Oncogenic Drivers of Cancer. Cancer Res 2017; 77:3502-3512. [PMID: 28512244 DOI: 10.1158/0008-5472.can-16-2745] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Revised: 03/07/2017] [Accepted: 04/27/2017] [Indexed: 01/22/2023]
Abstract
Oncogenic gene fusions drive many human cancers, but tools to more quickly unravel their functional contributions are needed. Here we describe methodology permitting fusion gene construction for functional evaluation. Using this strategy, we engineered the known fusion oncogenes, BCR-ABL1, EML4-ALK, and ETV6-NTRK3, as well as 20 previously uncharacterized fusion genes identified in The Cancer Genome Atlas datasets. In addition to confirming oncogenic activity of the known fusion oncogenes engineered by our construction strategy, we validated five novel fusion genes involving MET, NTRK2, and BRAF kinases that exhibited potent transforming activity and conferred sensitivity to FDA-approved kinase inhibitors. Our fusion construction strategy also enabled domain-function studies of BRAF fusion genes. Our results confirmed other reports that the transforming activity of BRAF fusions results from truncation-mediated loss of inhibitory domains within the N-terminus of the BRAF protein. BRAF mutations residing within this inhibitory region may provide a means for BRAF activation in cancer, therefore we leveraged the modular design of our fusion gene construction methodology to screen N-terminal domain mutations discovered in tumors that are wild-type at the BRAF mutation hotspot, V600. We identified an oncogenic mutation, F247L, whose expression robustly activated the MAPK pathway and sensitized cells to BRAF and MEK inhibitors. When applied broadly, these tools will facilitate rapid fusion gene construction for subsequent functional characterization and translation into personalized treatment strategies. Cancer Res; 77(13); 3502-12. ©2017 AACR.
Collapse
Affiliation(s)
- Hengyu Lu
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas
| | - Nicole Villafane
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas.,Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, Texas
| | - Turgut Dogruluk
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas
| | - Caitlin L Grzeskowiak
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas
| | - Kathleen Kong
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas
| | - Yiu Huen Tsang
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas
| | - Oksana Zagorodna
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas
| | - Angeliki Pantazi
- Division of Genetics, Brigham and Women's Hospital, Boston, Massachusetts
| | - Lixing Yang
- Department of Biomedical Informatics, Harvard Medical School, Boston, Massachusetts
| | - Nicholas J Neill
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas
| | - Young Won Kim
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas
| | - Chad J Creighton
- Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, Texas
| | - Roel G Verhaak
- The Jackson Laboratory, Genomic Medicine, Farmington, Connecticut
| | - Gordon B Mills
- Department of Systems Biology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Peter J Park
- Division of Genetics, Brigham and Women's Hospital, Boston, Massachusetts.,Department of Biomedical Informatics, Harvard Medical School, Boston, Massachusetts
| | - Raju Kucherlapati
- Division of Genetics, Brigham and Women's Hospital, Boston, Massachusetts.,Department of Genetics, Harvard Medical School, Boston, Massachusetts
| | - Kenneth L Scott
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas. .,Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, Texas
| |
Collapse
|
95
|
Le Gallo M, Rudd ML, Urick ME, Hansen NF, Zhang S, Lozy F, Sgroi DC, Vidal Bel A, Matias-Guiu X, Broaddus RR, Lu KH, Levine DA, Mutch DG, Goodfellow PJ, Salvesen HB, Mullikin JC, Bell DW. Somatic mutation profiles of clear cell endometrial tumors revealed by whole exome and targeted gene sequencing. Cancer 2017; 123:3261-3268. [PMID: 28485815 DOI: 10.1002/cncr.30745] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Revised: 02/08/2017] [Accepted: 02/14/2017] [Indexed: 01/10/2023]
Abstract
BACKGROUND The molecular pathogenesis of clear cell endometrial cancer (CCEC), a tumor type with a relatively unfavorable prognosis, is not well defined. We searched exome-wide for novel somatically mutated genes in CCEC and assessed the mutational spectrum of known and candidate driver genes in a large cohort of cases. METHODS We conducted whole exome sequencing of paired tumor-normal DNAs from 16 cases of CCEC (12 CCECs and the CCEC components of 4 mixed histology tumors). Twenty-two genes-of-interest were Sanger-sequenced from another 47 cases of CCEC. Microsatellite instability (MSI) and microsatellite stability (MSS) were determined by genotyping 5 mononucleotide repeats. RESULTS Two tumor exomes had relatively high mutational loads and MSI. The other 14 tumor exomes were MSS and had 236 validated nonsynonymous or splice junction somatic mutations among 222 protein-encoding genes. Among the 63 cases of CCEC in this study, we identified frequent somatic mutations in TP53 (39.7%), PIK3CA (23.8%), PIK3R1 (15.9%), ARID1A (15.9%), PPP2R1A (15.9%), SPOP (14.3%), and TAF1 (9.5%), as well as MSI (11.3%). Five of 8 mutations in TAF1, a gene with no known role in CCEC, localized to the putative histone acetyltransferase domain and included 2 recurrently mutated residues. Based on patterns of MSI and mutations in 7 genes, CCEC subsets molecularly resembled serous endometrial cancer (SEC) or endometrioid endometrial cancer (EEC). CONCLUSION Our findings demonstrate molecular similarities between CCEC and SEC and EEC and implicate TAF1 as a novel candidate CCEC driver gene. Cancer 2017;123:3261-8. © 2017 American Cancer Society.
Collapse
Affiliation(s)
- Matthieu Le Gallo
- Cancer Genetics and Comparative Genomics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland
| | - Meghan L Rudd
- Cancer Genetics and Comparative Genomics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland
| | - Mary Ellen Urick
- Cancer Genetics and Comparative Genomics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland
| | - Nancy F Hansen
- Cancer Genetics and Comparative Genomics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland
| | - Suiyuan Zhang
- Computational and Statistical Genomics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland
| | -
- National Institutes of Health Intramural Sequencing Center, National Institutes of Health, Bethesda, Maryland
| | - Fred Lozy
- Cancer Genetics and Comparative Genomics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland
| | - Dennis C Sgroi
- Molecular Pathology Unit, Massachusetts General Hospital, Charlestown, Massachusetts.,Center for Cancer Research, Massachusetts General Hospital, Charlestown, Massachusetts.,Department of Pathology, Harvard Medical School, Boston, Massachusetts
| | - August Vidal Bel
- Department of Pathology and Experimental Therapeutics, University of Barcelona, Barcelona, Spain.,Department of Pathology, Hospital Universitari de Bellvitge, Barcelona, Spain
| | - Xavier Matias-Guiu
- Department of Pathology, Hospital Universitari de Bellvitge, Barcelona, Spain.,Department of Pathology and Molecular Genetics/Oncological Pathology Group, Hospital Universitari Arnau de Vilanova, Universitat de Lleida, IRB Lleida, Lleida, Spain
| | - Russell R Broaddus
- Division of Surgery, Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Karen H Lu
- Division of Surgery, Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Douglas A Levine
- Gynecologic Oncology, Perlmutter Cancer Center, New York University Langone Medical Center, New York, New York
| | - David G Mutch
- Department of Obstetrics and Gynecology, Washington University School of Medicine, St. Louis, Missouri
| | - Paul J Goodfellow
- Department of Obstetrics and Gynecology, The Ohio State University Comprehensive Cancer Center, Columbus, Ohio
| | - Helga B Salvesen
- Department of Clinical Science, Center for Cancer Biomarkers, University of Bergen, Bergen, Norway.,Department of Gynecology and Obstetrics, Haukeland University Hospital, Bergen, Norway
| | - James C Mullikin
- Cancer Genetics and Comparative Genomics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland.,National Institutes of Health Intramural Sequencing Center, National Institutes of Health, Bethesda, Maryland
| | - Daphne W Bell
- Cancer Genetics and Comparative Genomics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
96
|
Kadara H, Choi M, Zhang J, Parra ER, Rodriguez-Canales J, Gaffney SG, Zhao Z, Behrens C, Fujimoto J, Chow C, Yoo Y, Kalhor N, Moran C, Rimm D, Swisher S, Gibbons DL, Heymach J, Kaftan E, Townsend JP, Lynch TJ, Schlessinger J, Lee J, Lifton RP, Wistuba II, Herbst RS. Whole-exome sequencing and immune profiling of early-stage lung adenocarcinoma with fully annotated clinical follow-up. Ann Oncol 2017; 28:75-82. [PMID: 27687306 DOI: 10.1093/annonc/mdw436] [Citation(s) in RCA: 141] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Background Lung adenocarcinomas (LUADs) lead to the majority of deaths attributable to lung cancer. We performed whole-exome sequencing (WES) and immune profiling analyses of a unique set of clinically annotated early-stage LUADs to better understand the pathogenesis of this disease and identify clinically relevant molecular markers. Methods We performed WES of 108 paired stage I-III LUADs and normal lung tissues using the Illumina HiSeq 2000 platform. Ten immune markers (PD-L1, PD-1, CD3, CD4, CD8, CD45ro, CD57, CD68, FOXP3 and Granzyme B) were profiled by imaging-based immunohistochemistry (IHC) in a subset of LUADs (n = 92). Associations among mutations, immune markers and clinicopathological variables were analyzed using ANOVA and Fisher's exact test. Cox proportional hazards regression models were used for multivariate analysis of clinical outcome. Results LUADs in this cohort exhibited an average of 243 coding mutations. We identified 28 genes with significant enrichment for mutation. SETD2-mutated LUADs exhibited relatively poor recurrence- free survival (RFS) and mutations in STK11 and ATM were associated with poor RFS among KRAS-mutant tumors. EGFR, KEAP1 and PIK3CA mutations were predictive of poor response to adjuvant therapy. Immune marker analysis revealed that LUADs in smokers and with relatively high mutation burdens exhibited increased levels of immune markers. Analysis of immunophenotypes revealed that LUADs with STK11 mutations exhibited relatively low levels of infiltrating CD4+/CD8+ T-cells indicative of a muted immune response. Tumoral PD-L1 was significantly elevated in TP53 mutant LUADs whereas PIK3CA mutant LUADs exhibited markedly down-regulated PD-L1 expression. LUADs with TP53 or KEAP1 mutations displayed relatively increased CD57 and Granzyme B levels indicative of augmented natural killer (NK) cell infiltration. Conclusion(s) Our study highlights molecular and immune phenotypes that warrant further analysis for their roles in clinical outcomes and personalized immune-based therapy of LUAD.
Collapse
Affiliation(s)
- H Kadara
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - M Choi
- Department of Genetics, Howard Hughes Medical Institute, Maryland, USA.,Department of Biomedical Sciences, Seoul National University, Seoul, Republic of Korea
| | - J Zhang
- Department of Biostatistics and Computational Biology, Indiana University Melvin and Bren Simon Cancer Center , Indianapolis , Indiana , USA
| | - E R Parra
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston
| | - J Rodriguez-Canales
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston
| | - S G Gaffney
- Department of Ecology and Evolutionary Biology, Yale University, New Haven
| | - Z Zhao
- Department of Ecology and Evolutionary Biology, Yale University, New Haven
| | - C Behrens
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - J Fujimoto
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston
| | - C Chow
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston
| | - Y Yoo
- Biomedical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea
| | | | | | - D Rimm
- Yale Comprehensive Cancer Center, Yale School of Medicine, Yale University, New Haven
| | - S Swisher
- Department of Thoracic and Cardiovascular Surgery, The University of Texas MD Anderson Cancer Center, Houston
| | - D L Gibbons
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - J Heymach
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - E Kaftan
- Yale Comprehensive Cancer Center, Yale School of Medicine, Yale University, New Haven
| | - J P Townsend
- Department of Ecology and Evolutionary Biology, Yale University, New Haven
| | - T J Lynch
- Yale Comprehensive Cancer Center, Yale School of Medicine, Yale University, New Haven
| | - J Schlessinger
- Yale Comprehensive Cancer Center, Yale School of Medicine, Yale University, New Haven
| | - J Lee
- Department of Biostatistics and Computational Biology, Indiana University Melvin and Bren Simon Cancer Center , Indianapolis , Indiana , USA
| | - R P Lifton
- Department of Genetics, Howard Hughes Medical Institute, Maryland, USA.,Yale Center for Genome Analysis, Yale School of Medicine, Yale University, New Haven, USA
| | - I I Wistuba
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston
| | - R S Herbst
- Yale Comprehensive Cancer Center, Yale School of Medicine, Yale University, New Haven
| |
Collapse
|
97
|
Molecular Alterations of TP53 are a Defining Feature of Ovarian High-Grade Serous Carcinoma: A Rereview of Cases Lacking TP53 Mutations in The Cancer Genome Atlas Ovarian Study. Int J Gynecol Pathol 2017; 35:48-55. [PMID: 26166714 DOI: 10.1097/pgp.0000000000000207] [Citation(s) in RCA: 121] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The Cancer Genome Atlas has reported that 96% of ovarian high-grade serous carcinomas (HGSCs) have TP53 somatic mutations suggesting that mutation of this gene is a defining feature of this neoplasm. In the current study, 5 gynecologic pathologists independently evaluated hematoxylin and eosin slides of 14 available cases from The Cancer Genome Atlas classified as HGSC that lacked a TP53 mutation. The histologic diagnoses rendered by these pathologists and the accompanying molecular genetic data are the subject of this report. Only 1 case (Case 5), which contained a homozygous deletion of TP53, had unanimous interobserver agreement for a diagnosis of pure HGSC. In 1 case (Case 3), all 5 observers (100%) rendered a diagnosis of HGSC; however, 3 observers (60%) noted that the histologic features were not classic for HGSC and suggested this case may have arisen from a low-grade serous carcinoma (arisen from an alternate pathway compared with the usual HGSC). In 2 cases (Cases 4 and 12), only 3 observers (60%) in each case, respectively, interpreted it as having a component of HGSC. In the remaining 10 (71%) of tumors (Cases 1, 2, 6-11, 13, and 14), the consensus diagnosis was not HGSC, with individual diagnoses including low-grade serous carcinoma, high-grade endometrioid carcinoma, HGSC, metastatic carcinoma, clear cell carcinoma, atypical proliferative (borderline) serous tumor, and adenocarcinoma, not otherwise specified. Therefore, 13 (93%) of the tumors (Cases 1-4 and 6-14) were either not a pure HGSC or represented a diagnosis other than HGSC, all with molecular results not characteristic of HGSC. Accordingly, our review of the TP53 wild-type HGSCs reported in The Cancer Genome Atlas suggests that 100% of de novo HGSCs contain TP53 somatic mutations or deletions, with the exception of the rare HGSCs that develop from a low-grade serous tumor precursor. We, therefore, propose that lack of molecular alterations of TP53 are essentially inconsistent with the diagnosis of ovarian HGSC and that tumors diagnosed as such should be rigorously reassessed to achieve correct classification.
Collapse
|
98
|
Chang YS, Huang HD, Yeh KT, Chang JG. Identification of novel mutations in endometrial cancer patients by whole-exome sequencing. Int J Oncol 2017; 50:1778-1784. [PMID: 28339086 DOI: 10.3892/ijo.2017.3919] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Accepted: 03/07/2017] [Indexed: 11/05/2022] Open
Abstract
The aim of the present study was to identify genomic alterations in Taiwanese endometrial cancer patients. This information is vitally important in Taiwan, where endometrial cancer is the second most common gynecological cancer. We performed whole-exome sequencing on DNA from 14 tumor tissue samples from Taiwanese endometrial cancer patients. We used the Genome Analysis Tool kit software package for data analysis, and the dbSNP, Catalogue of Somatic Mutations in Cancer (COSMIC) and The Cancer Genome Atlas (TCGA) databases for comparisons. Variants were validated via Sanger sequencing. We identified 143 non-synonymous mutations in 756 canonical cancer-related genes and 1,271 non-synonymous mutations in non-canonical cancer-related genes in 14 endometrial samples. PTEN, KRAS and PIK3R1 were the most frequently mutated canonical cancer-related genes. Our results revealed nine potential driver genes (MAPT, IL24, MCM6, TSC1, BIRC2, CIITA, DST, CASP8 and NOTCH2) and 21 potential passenger genes (ARMCX4, IGSF10, VPS13C, DCT, DNAH14, TLN1, ZNF605, ZSCAN29, MOCOS, CMYA5, PCDH17, UGT1A8, CYFIP2, MACF1, NUDT5, JAKMIP1, PCDHGB4, FAM178A, SNX6, IMP4 and PCMTD1). The detected molecular aberrations led to putative activation of the mTOR, Wnt, MAPK, VEGF and ErbB pathways, as well as aberrant DNA repair, cell cycle control and apoptosis pathways. We characterized the mutational landscape and genetic alterations in multiple cellular pathways of endometrial cancer in the Taiwanese population.
Collapse
Affiliation(s)
- Ya-Sian Chang
- Epigenome Research Center, China Medical University Hospital, Taichung 404, Taiwan, R.O.C
| | - Hsien-Da Huang
- Department of Biological Science and Technology, National Chiao Tung University, Hsinchu 300, Taiwan, R.O.C
| | - Kun-Tu Yeh
- Department of Pathology, Changhua Christian Hospital, Changhua 500, Taiwan, R.O.C
| | - Jan-Gowth Chang
- Epigenome Research Center, China Medical University Hospital, Taichung 404, Taiwan, R.O.C
| |
Collapse
|
99
|
Wang C, Gu C, Jeong KJ, Zhang D, Guo W, Lu Y, Ju Z, Panupinthu N, Yang JY, Gagea MM, Ng PKS, Zhang F, Mills GB. YAP/TAZ-Mediated Upregulation of GAB2 Leads to Increased Sensitivity to Growth Factor-Induced Activation of the PI3K Pathway. Cancer Res 2017; 77:1637-1648. [PMID: 28202507 DOI: 10.1158/0008-5472.can-15-3084] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2015] [Revised: 11/29/2016] [Accepted: 01/06/2017] [Indexed: 11/16/2022]
Abstract
The transcription regulators YAP and TAZ function as effectors of the HIPPO signaling cascade, critical for organismal development, cell growth, and cellular reprogramming, and YAP/TAZ is commonly misregulated in human cancers. The precise mechanism by which aberrant YAP/TAZ promotes tumor growth remains unclear. The HIPPO tumor suppressor pathway phosphorylates YAP and TAZ, resulting in cytosolic sequestration with subsequent degradation. Here, we report that the PI3K/AKT pathway, which is critically involved in the pathophysiology of endometrial cancer, interacts with the HIPPO pathway at multiple levels. Strikingly, coordinate knockdown of YAP and TAZ, mimicking activation of the HIPPO pathway, markedly decreased both constitutive and growth factor-induced PI3K pathway activation by decreasing levels of the GAB2 linker molecule in endometrial cancer lines. Furthermore, targeting YAP/TAZ decreased endometrial cancer tumor growth in vivo In addition, YAP and TAZ total and phosphoprotein levels correlated with clinical characteristics and outcomes in endometrial cancer. Thus, YAP and TAZ, which are inhibited by the HIPPO tumor suppressor pathway, modify PI3K/AKT pathway signaling in endometrial cancer. The cross-talk between these key pathways identifies potential new biomarkers and therapeutic targets in endometrial cancer. Cancer Res; 77(7); 1637-48. ©2017 AACR.
Collapse
Affiliation(s)
- Chao Wang
- Department of Obstetrics and Gynecology, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China. .,Department of Systems Biology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Chao Gu
- Department of Obstetrics and Gynecology, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China.,Department of Systems Biology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Kang Jin Jeong
- Department of Systems Biology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Dong Zhang
- Department of Systems Biology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Wei Guo
- Department of Systems Biology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Yiling Lu
- Department of Systems Biology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Zhenlin Ju
- Department of Bioinformatics and Computational Biology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Nattapon Panupinthu
- Department of Systems Biology, University of Texas MD Anderson Cancer Center, Houston, Texas.,Department of Physiology, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Ji Yeon Yang
- Department of Bioinformatics and Computational Biology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Mihai Mike Gagea
- Department of Veterinary Medicine & Surgery, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Patrick Kwok Shing Ng
- Department of Systems Biology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Fan Zhang
- Department of Systems Biology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Gordon B Mills
- Department of Systems Biology, University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
100
|
Kamps R, Brandão RD, Bosch BJVD, Paulussen ADC, Xanthoulea S, Blok MJ, Romano A. Next-Generation Sequencing in Oncology: Genetic Diagnosis, Risk Prediction and Cancer Classification. Int J Mol Sci 2017; 18:ijms18020308. [PMID: 28146134 PMCID: PMC5343844 DOI: 10.3390/ijms18020308] [Citation(s) in RCA: 315] [Impact Index Per Article: 39.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Accepted: 01/19/2017] [Indexed: 12/17/2022] Open
Abstract
Next-generation sequencing (NGS) technology has expanded in the last decades with significant improvements in the reliability, sequencing chemistry, pipeline analyses, data interpretation and costs. Such advances make the use of NGS feasible in clinical practice today. This review describes the recent technological developments in NGS applied to the field of oncology. A number of clinical applications are reviewed, i.e., mutation detection in inherited cancer syndromes based on DNA-sequencing, detection of spliceogenic variants based on RNA-sequencing, DNA-sequencing to identify risk modifiers and application for pre-implantation genetic diagnosis, cancer somatic mutation analysis, pharmacogenetics and liquid biopsy. Conclusive remarks, clinical limitations, implications and ethical considerations that relate to the different applications are provided.
Collapse
Affiliation(s)
- Rick Kamps
- Department of Clinical Genetics: GROW-School for Oncology and Developmental Biology, Maastricht University Medical Centre, 6229HX Maastricht, The Netherlands.
| | - Rita D Brandão
- Department of Clinical Genetics: GROW-School for Oncology and Developmental Biology, Maastricht University Medical Centre, 6229HX Maastricht, The Netherlands.
| | - Bianca J van den Bosch
- Department of Clinical Genetics: GROW-School for Oncology and Developmental Biology, Maastricht University Medical Centre, 6229HX Maastricht, The Netherlands.
| | - Aimee D C Paulussen
- Department of Clinical Genetics: GROW-School for Oncology and Developmental Biology, Maastricht University Medical Centre, 6229HX Maastricht, The Netherlands.
| | - Sofia Xanthoulea
- Department of Gynaecology and Obstetrics: GROW-School for Oncology and Developmental Biology, Maastricht University Medical Centre, 6229HX Maastricht, The Netherlands.
| | - Marinus J Blok
- Department of Clinical Genetics: GROW-School for Oncology and Developmental Biology, Maastricht University Medical Centre, 6229HX Maastricht, The Netherlands.
| | - Andrea Romano
- Department of Gynaecology and Obstetrics: GROW-School for Oncology and Developmental Biology, Maastricht University Medical Centre, 6229HX Maastricht, The Netherlands.
| |
Collapse
|