1
|
Van Alsten SC, Love MI, Calhoun BC, Butler EN, Perou CM, Hoadley KA, Troester MA. Genomic Analysis Reveals Racial and Age-Related Differences in the Somatic Landscape of Breast Cancer and the Association with Socioeconomic Factors. Cancer Res 2025; 85:1327-1340. [PMID: 39879109 PMCID: PMC12034101 DOI: 10.1158/0008-5472.can-24-1788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 08/27/2024] [Accepted: 01/16/2025] [Indexed: 01/31/2025]
Abstract
Cancer genomics consortia have identified somatic drivers of breast cancer subtypes. However, these studies have predominantly included older, non-Black women, and the related socioeconomic status (SES) data are limited. Increased representation and depth of social data are crucial for understanding how health inequity is intertwined with somatic landscapes. Here, we conducted targeted sequencing on primary tumors from the Carolina Breast Cancer Study (N = 357; 52% Black; 47% <50) and compared the results with The Cancer Genome Atlas (N = 948; 18% Black; 27% <50). Race (Black vs. non-Black), age, and SES were evaluated in association with mutations, copy number alterations, and aneuploidy using generalized linear models. Pathway dysfunction was also assessed by aggregating mutation and copy number alterations. Adjusting for age, Black participants (N = 350) were significantly more likely to have TP53 and FAT1 mutations and less likely to have PIK3CA, CDH1, DDR2, and GATA3 mutations than non-Black participants. Younger participants had more GATA3 alterations and fewer KMT2C, PTEN, MAP3K1, and CDH1 alterations. Black participants had significant enrichment for MYC (8q) and PIK3CA (3q26) amplifications and higher total aneuploidy, but age was not associated with copy number variation. SES was associated with different patterns of alteration in Black versus non-Black women. Overall, Black participants showed modest differences in TP53, PIK3CA, and other alterations that further varied by SES. Race is a social construct, and varying distributions of etiologic factors across social strata may predispose Black, young, and low SES women to cancer subtypes characterized by these alterations. Significance: The collection and analysis of DNA sequencing with comprehensive socioeconomic factor associations in a large Black breast cancer patient cohort could help uncover mechanisms by which social conditions contribute to tumor biology.
Collapse
Affiliation(s)
- Sarah C. Van Alsten
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
- Department of Epidemiology, Gillings School of Public Health, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Michael I. Love
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Benjamin C. Calhoun
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
- Department of Pathology and Laboratory Medicine, School of Medicine, University of North Carolina, Chapel Hill, North Carolina
| | - Eboneé N. Butler
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
- Department of Epidemiology, Gillings School of Public Health, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Charles M. Perou
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Katherine A. Hoadley
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Melissa A. Troester
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
- Department of Epidemiology, Gillings School of Public Health, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
- Department of Pathology and Laboratory Medicine, School of Medicine, University of North Carolina, Chapel Hill, North Carolina
| |
Collapse
|
2
|
Bae-Jump VL, Sill MW, Gehrig PA, Merker JD, Corcoran DL, Pfefferle AD, Hayward MC, Walker JL, Hagemann AR, Waggoner SE, O'Cearbhaill RE, McDonald ME, Edelson MI, DiSilvestro PA, McNally AL, Fleury A, Littell RD, Ueland FR, Lankes HA, Aghajanian C. A randomized phase II/III study of paclitaxel/carboplatin/metformin versus paclitaxel/carboplatin/placebo as initial therapy for measurable stage III or IVA, stage IVB, or recurrent endometrial cancer: An NRG oncology/GOG study. Gynecol Oncol 2025; 195:66-74. [PMID: 40056832 DOI: 10.1016/j.ygyno.2025.03.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 02/28/2025] [Accepted: 03/03/2025] [Indexed: 03/10/2025]
Abstract
INTRODUCTION We evaluated the efficacy of the addition of the anti-diabetic drug metformin to standard-of-care paclitaxel and carboplatin (PC) in patients with advanced and recurrent endometrial cancer (EC). METHODS In this phase II/III trial, EC patients with chemotherapy-naïve stage III/IVA (with measurable disease) and stage IVB or recurrent (with or without measurable disease) disease were randomly assigned to PC/metformin (850 mg BID) versus PC/placebo. Metformin or placebo was continued as maintenance therapy after completion of PC until disease progression. The primary endpoint of phase II was progression-free survival (PFS). The primary endpoint of phase III was overall survival (OS). Secondary endpoints were objective response, duration of response, and toxicity. RESULTS From 3/17/2014 to 12/22/2017, 448 patients were randomized to phase II/III studies, and the data were frozen for interim analysis. The phase II study deemed metformin worthy of further investigation in the phase III study. The interim phase III analysis stopped accrual for futility on 2/1/2018. The addition of metformin to PC had a slightly higher hazard of death compared to the PC regimen (HR = 1.088; 90% CI 0.803 to 1.475), which was sufficient to close the study early. The PFS had (HR = 0.814; 90% CI 0.635 to 1.043). At a median follow-up of 10 months and 121 deaths, median OS was not determined and 28 months, on PC/placebo and PC/metformin, respectively. CONCLUSION The hazard ratios for PFS and OS endpoints was not sufficiently decreased with the addition of metformin to PC to justify continuing the trial.
Collapse
Affiliation(s)
- Victoria L Bae-Jump
- Division of Gynecologic Oncology, University of North Carolina, Chapel Hill, NC, United States of America; Lineberger Comprehensive Cancer Center, UNC Department of Genetics, University of North Carolina, Chapel Hill, NC, United States of America.
| | - Michael W Sill
- NRG Statistics and Data Management Center, Roswell Park Cancer Institute, Buffalo, NY, United States of America.
| | - Paola A Gehrig
- Division of Gynecologic Oncology, University of Virginia, Charlottesville, VA, United States of America.
| | - Jason D Merker
- Lineberger Comprehensive Cancer Center, UNC Department of Genetics, University of North Carolina, Chapel Hill, NC, United States of America.
| | - David L Corcoran
- Lineberger Comprehensive Cancer Center, UNC Department of Genetics, University of North Carolina, Chapel Hill, NC, United States of America.
| | - Adam D Pfefferle
- Lineberger Comprehensive Cancer Center, UNC Department of Genetics, University of North Carolina, Chapel Hill, NC, United States of America.
| | - Michele C Hayward
- Lineberger Comprehensive Cancer Center, UNC Department of Genetics, University of North Carolina, Chapel Hill, NC, United States of America.
| | - Joan L Walker
- Division of Gynecologic Oncology, University of Oklahoma, Oklahoma, OK, United States of America.
| | - Andrea R Hagemann
- Division of Gynecologic Oncology, Washington University School of Medicine, St. Louis, MO, United States of America.
| | - Steven E Waggoner
- Section of Gynecologic Oncology, Cleveland Clinic, Cleveland, OH, United States of America.
| | | | - Megan E McDonald
- Division of Gynecologic Oncology, University of Iowa Hospitals and Clinics, Iowa City, IA, United States of America.
| | - Mitchell I Edelson
- Hanjani Institute for Gynecologic Oncology, Jefferson Abington Hospital, Asplundh Cancer Center of Sidney Kimmel Cancer Center, Jefferson Health, Willow Grove, PA, United States of America.
| | - Paul A DiSilvestro
- Division of Gynecologic Oncology, Women & Infants Hospital, Brown University, United States of America.
| | - Amy L McNally
- Division of Gynecologic Oncology, Minnesota Oncology, Woodbury, MN, United States of America.
| | - Aimee Fleury
- Women's Cancer Center of Nevada, Las Vegas, NV, United States of America.
| | - Ramey D Littell
- Kaiser Permanente Northern California, San Francisco, CA, United States of America.
| | - Frederick R Ueland
- University of Kentucky, Obstetrics & Gynecology, Lexington, KY, United States of America.
| | - Heather A Lankes
- NRG Oncology, Operations Center-Philadelphia East, Philadelphia, PA, United States of America; Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, The Ohio State University Wexner Medical Center, Columbus, OH, United States of America.
| | - Carol Aghajanian
- Memorial Sloan Kettering Cancer Center, New York, NY, United States of America.
| |
Collapse
|
3
|
Gormley M, Adhikari A, Dudding T, Pring M, Hurley K, Macfarlane GJ, Lagiou P, Lagiou A, Polesel J, Agudo A, Alemany L, Ahrens W, Healy CM, Conway DI, Canova C, Holcatova I, Richiardi L, Znaor A, Olshan AF, Hung RJ, Liu G, Bratman S, Zhao X, Holt J, Cortez R, Gaborieau V, McKay JD, Waterboer T, Brennan P, Hayes N, Diergaarde B, Virani S. VOYAGER: an international consortium investigating the role of human papilloma virus and genetics in oral and oropharyngeal cancer risk and survival. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.02.17.25322399. [PMID: 40034767 PMCID: PMC11875266 DOI: 10.1101/2025.02.17.25322399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Head and neck cancer (HNC) is the sixth most common cancer globally. Incidence and survival rates vary significantly across geographic regions and tumor subsites. This is partly due to differences in risk factor exposure, which includes tobacco smoking, alcohol consumption and human papillomavirus (HPV) infection, alongside detection and treatment strategies. The VOYAGER (human papillomaVirus, Oral and oropharYngeal cAncer GEnomic Research) consortium is a collaboration between five large North American and European studies which generated data on 10,530 participants (7,233 cases and 3,297 controls). The primary goal of the collaboration was to improve understing of the role of HPV and genetic factors in oral cavity and oropharyngeal cancer risk and outcome. Demographic and clinical data collected by the five studies were harmonized, and HPV status was determined for the majority of cases. In addition, 999 tumors were sequenced to define somatic mutations. These activities generated a comprehensive biomedical resource that can be utilized to answer critical outsting research questions to help improve HNC prevention, early detection, treatment, and surveillance.
Collapse
Affiliation(s)
- M Gormley
- MRC Integrative Epidemiology Unit, Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
- Bristol Dental School, University of Bristol, Bristol, UK
- University Hospitals Bristol NHS Foundation Trust Bristol Dental Hospital, Bristol, UK
| | - A Adhikari
- University Hospitals Bristol NHS Foundation Trust Bristol Dental Hospital, Bristol, UK
| | - T Dudding
- MRC Integrative Epidemiology Unit, Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
- Bristol Dental School, University of Bristol, Bristol, UK
| | - M Pring
- Bristol Dental School, University of Bristol, Bristol, UK
- University Hospitals Bristol NHS Foundation Trust Bristol Dental Hospital, Bristol, UK
| | - K Hurley
- University Hospitals Bristol NHS Foundation Trust Bristol Dental Hospital, Bristol, UK
| | - G J Macfarlane
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, UK
| | - P Lagiou
- School of Medicine, National and Kapodistrian University of Athens, Greece
| | - A Lagiou
- School of Public Health, University of West Attica, Greece
| | - J Polesel
- Unit of Cancer Epidemiology, Centro di Riferimento Oncologico di Aviano (CRO) National Cancer Institute, IRCCS, Italy
| | - A Agudo
- Nutrition and Cancer Unit, Cancer Epidemiology Research Program, Catalan Institute of Oncology/IDIBELL, Barcelona, Spain
| | - L Alemany
- Infections and Cancer Unit, Cancer Epidemiology Research Program, Catalan Institute of Oncology/IDIBELL, Barcelona, Spain
- Centro de Investigación Biomédica en Red: Epidemiología y Salud Pública (CIBERESP CB06/02/0073), Madrid, Spain
| | - W Ahrens
- Epidemiological Methods and Etiological Research, Leibniz Institute for Prevention Research and Epidemiology - BIPS, Germany
| | - C M Healy
- School of Dental Science, Dublin Dental University Hospital, Trinity College Dublin, Irel
| | - D I Conway
- School of Medicine, Dentistry, and Nursing, University of Glasgow, UK
| | - C Canova
- Department of Cardiac, Thoracic and Vascular Sciences University of Padova, Italy
| | - I Holcatova
- Institute of Hygiene and Epidemiology, Charles University Prague, Czech Republic
| | - L Richiardi
- Reference Centre for Epidemiology and Cancer Prevention in Piemonte, Italy
| | - A Znaor
- Cancer Surveillance, International Agency for Research on Cancer, France
| | - A F Olshan
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina, US
| | - R J Hung
- Prosserman Centre for Population Health Research, Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, Canada
- Dalla Lana School of Public Health, University of Toronto, Toronto, Canada
| | - G Liu
- Dalla Lana School of Public Health, University of Toronto, Toronto, Canada
- Computational Biology and Medicine Program, Princess Margaret Cancer Centre, Toronto Canada
| | - S Bratman
- Department of Radiation Oncology, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, Canada
| | - X Zhao
- Department of Medicine, University of Tennessee, USA
| | - J Holt
- Department of Medicine, University of Tennessee, USA
| | - R Cortez
- Genomic Epidemiology Group, World Health Organization, International Agency for Research on Cancer, Lyon, France
| | - V Gaborieau
- Genomic Epidemiology Group, World Health Organization, International Agency for Research on Cancer, Lyon, France
| | - J D McKay
- Genomic Epidemiology Group, World Health Organization, International Agency for Research on Cancer, Lyon, France
| | - T Waterboer
- Infections and Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - P Brennan
- Genomic Epidemiology Group, World Health Organization, International Agency for Research on Cancer, Lyon, France
| | - N Hayes
- Department of Medicine, University of Tennessee, USA
| | - B Diergaarde
- Department of Human Genetics, School of Public Health, University of Pittsburgh, and UPMC Hillman Cancer Center, Pittsburgh, US
| | - S Virani
- Genomic Epidemiology Group, World Health Organization, International Agency for Research on Cancer, Lyon, France
| |
Collapse
|
4
|
Gillis N, Etheridge AS, Patil SA, Hayes DN, Hayward MC, Auman JT, Parker JS, Innocenti F. Sequencing of genes of drug response in tumor DNA and implications for precision medicine in cancer patients. THE PHARMACOGENOMICS JOURNAL 2023:10.1038/s41397-023-00299-7. [PMID: 36709390 DOI: 10.1038/s41397-023-00299-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 01/17/2023] [Accepted: 01/19/2023] [Indexed: 01/30/2023]
Abstract
Tumor DNA sequencing is becoming standard-of-care for patient treatment decisions. We evaluated genotype concordance between tumor DNA and genomic DNA from blood and catalogued functional effects of somatic mutations in 21 drug response genes in 752 solid tumor patients. Using a threshold of 10% difference between tumor and blood DNA variant allele fraction (VAF), concordance for heterogenous genotype calls was 78% and increased to 97.5% using a 30% VAF threshold. Somatic mutations were observed in all 21 drug response genes, and 44% of patients had at least one somatic mutation in these genes. In tumor DNA, eight patients had a frameshift mutation in CYP2C8, which metabolizes taxanes. Overall, somatic copy number losses were more frequent than gains, including for CYP2C19 and CYP2D6 which had the most frequent copy number losses. However, copy number gains in TPMT were more than four times as common as losses. Seven % of patients had copy number gains in ABCB1, a multidrug resistance transporter of anti-cancer agents. These results demonstrate tumor-only DNA sequencing might not be reliable to call germline genotypes of drug response variants.
Collapse
Affiliation(s)
- Nancy Gillis
- Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, USA.,Department of Cancer Epidemiology, Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Amy S Etheridge
- Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, USA.
| | - Sushant A Patil
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - D Neil Hayes
- Department of Medicine, Hematology/Oncology, University of North Carolina, Chapel Hill, NC, USA.,Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA.,Center for Cancer Research, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Michele C Hayward
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
| | - J Todd Auman
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - Joel S Parker
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA.,Department of Genetics, University of North Carolina, Chapel Hill, NC, USA
| | - Federico Innocenti
- Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, USA.,Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
| |
Collapse
|
5
|
Joo MS, Pyo KH, Chung JM, Cho BC. Artificial intelligence-based non-small cell lung cancer transcriptome RNA-sequence analysis technology selection guide. Front Bioeng Biotechnol 2023; 11:1081950. [PMID: 36873350 PMCID: PMC9975749 DOI: 10.3389/fbioe.2023.1081950] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 01/24/2023] [Indexed: 02/17/2023] Open
Abstract
The incidence and mortality rates of lung cancer are high worldwide, where non-small cell lung cancer (NSCLC) accounts for more than 85% of lung cancer cases. Recent non-small cell lung cancer research has been focused on analyzing patient prognosis after surgery and identifying mechanisms in connection with clinical cohort and ribonucleic acid (RNA) sequencing data, including single-cell ribonucleic acid (scRNA) sequencing data. This paper investigates statistical techniques and artificial intelligence (AI) based non-small cell lung cancer transcriptome data analysis methods divided into target and analysis technology groups. The methodologies of transcriptome data were schematically categorized so researchers can easily match analysis methods according to their goals. The most widely known and frequently utilized transcriptome analysis goal is to find essential biomarkers and classify carcinomas and cluster NSCLC subtypes. Transcriptome analysis methods are divided into three major categories: Statistical analysis, machine learning, and deep learning. Specific models and ensemble techniques typically used in NSCLC analysis are summarized in this paper, with the intent to lay a foundation for advanced research by converging and linking the various analysis methods available.
Collapse
Affiliation(s)
- Min Soo Joo
- School of Electrical and Electronic Engineering, College of Engineering, Yonsei University, Seoul, Republic of Korea
| | - Kyoung-Ho Pyo
- Department of Oncology, Severance Hospital, College of Medicine, Yonsei University, Seoul, Republic of Korea.,Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Republic of Korea.,Yonsei New Il Han Institute for Integrative Lung Cancer Research, Yonsei University College of Medicine, Seoul, Republic of Korea.,Division of Medical Oncology, Department of Internal Medicine and Yonsei Cancer Center, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jong-Moon Chung
- School of Electrical and Electronic Engineering, College of Engineering, Yonsei University, Seoul, Republic of Korea.,Department of Emergency Medicine, College of Medicine, Yonsei University, Seoul, Republic of Korea
| | - Byoung Chul Cho
- Division of Medical Oncology, Department of Internal Medicine and Yonsei Cancer Center, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
6
|
Zhao J, Wu Y, Chen MJ, Xu Y, Zhong W, Wang MZ. Characterization of driver mutations in Chinese non-small cell lung cancer patients using a novel targeted sequencing panel. J Thorac Dis 2022; 14:4669-4684. [PMID: 36647494 PMCID: PMC9840037 DOI: 10.21037/jtd-22-909] [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: 07/01/2022] [Accepted: 11/04/2022] [Indexed: 12/05/2022]
Abstract
Background The identification of driver mutations has greatly promoted the precise diagnosis and treatment of non-small cell lung cancer (NSCLC), but there is lack of targeted sequencing panels specifically designed and applied to Chinese NSCLC patients. This study aimed to design and validate of a novel sequencing panel for comprehensive characterization of driver mutations in Chinese NSCLC patients, facilitating further exploration of downstream pathway alterations and therapeutic utility. Methods A novel target sequencing panel including 21 driver genes was designed and examined in a cohort of 260 Chinese NSCLC patients who underwent surgery in Peking Union Medical College Hospital (PUMCH). Genetic alterations were identified and further analyzed for driver mutations, downstream pathways and therapeutic utilities. Results The most frequently identified driver mutations in PUMCH NSCLC cohort were on genes TP53 (28%), EGFR (27%) and PIK3CA (19%) for lung adenocarcinoma (LUAD), and TP53 (41%), PIK3CA (14%) and CDKN2A (13%) for lung squamous cell carcinoma (LUSC), respectively. Downstream pathway analysis revealed common pathways like G1_AND_S1_PHASES pathway were shared not only between LUAD and LUSC patients, but also among three different NSCLC cohorts, while other pathways were subtype-specific, like the unique enrichment of SHC1_EVENT_IN_EGFR_SIGNALING pathway in LUAD patients, and P38_ALPHA_BETA_DOWNSTREAM pathway in LUSC patients, respectively. About 60% of both LUAD and LUSC patients harbored driver mutations as sensitive biomarkers for different targeted therapies, covering not only frequent mutations like EGFR L858R mutation, but also rare mutations like BRAF D594N mutation. Conclusions Our study provides a novel target sequencing panel suitable for Chinese NSCLC patients, which can effectively identify driver mutations, analyze downstream pathway alterations and predict therapeutic utility. Overall it is promising to further optimize and apply this panel in clinic with convenience and effectiveness.
Collapse
Affiliation(s)
- Jing Zhao
- Department of Respiratory and Critical Care Medicine, Peking Union Medical College Hospital, Beijing, China
| | - Yang Wu
- School of Medicine, Tsinghua University, Beijing, China
| | - Min-Jiang Chen
- Department of Respiratory and Critical Care Medicine, Peking Union Medical College Hospital, Beijing, China
| | - Yan Xu
- Department of Respiratory and Critical Care Medicine, Peking Union Medical College Hospital, Beijing, China
| | - Wei Zhong
- Department of Respiratory and Critical Care Medicine, Peking Union Medical College Hospital, Beijing, China
| | - Meng-Zhao Wang
- Department of Respiratory and Critical Care Medicine, Peking Union Medical College Hospital, Beijing, China
| |
Collapse
|
7
|
Little P, Jo H, Hoyle A, Mazul A, Zhao X, Salazar AH, Farquhar D, Sheth S, Masood M, Hayward MC, Parker JS, Hoadley KA, Zevallos J, Hayes DN. UNMASC: tumor-only variant calling with unmatched normal controls. NAR Cancer 2021; 3:zcab040. [PMID: 34632388 PMCID: PMC8494212 DOI: 10.1093/narcan/zcab040] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 09/07/2021] [Accepted: 10/04/2021] [Indexed: 12/11/2022] Open
Abstract
Despite years of progress, mutation detection in cancer samples continues to require significant manual review as a final step. Expert review is particularly challenging in cases where tumors are sequenced without matched normal control DNA. Attempts have been made to call somatic point mutations without a matched normal sample by removing well-known germline variants, utilizing unmatched normal controls, and constructing decision rules to classify sequencing errors and private germline variants. With budgetary constraints related to computational and sequencing costs, finding the appropriate number of controls is a crucial step to identifying somatic variants. Our approach utilizes public databases for canonical somatic variants as well as germline variants and leverages information gathered about nearby positions in the normal controls. Drawing from our cohort of targeted capture panel sequencing of tumor and normal samples with varying tumortypes and demographics, these served as a benchmark for our tumor-only variant calling pipeline to observe the relationship between our ability to correctly classify variants against a number of unmatched normals. With our benchmarked samples, approximately ten normal controls were needed to maintain 94% sensitivity, 99% specificity and 76% positive predictive value, far outperforming comparable methods. Our approach, called UNMASC, also serves as a supplement to traditional tumor with matched normal variant calling workflows and can potentially extend to other concerns arising from analyzing next generation sequencing data.
Collapse
Affiliation(s)
- Paul Little
- Public Health Sciences, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave N, Seattle, WA 98109, USA
| | - Heejoon Jo
- Center for Cancer Research, University of Tennessee Health Science Center, 19 South Manassas, Memphis, TN 38163, USA
| | - Alan Hoyle
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, 101 Manning Drive Chapel Hill, NC 27514, USA
| | - Angela Mazul
- Otolaryngology Head and Neck Surgery, Washington University School of Medicine, 660 South Euclid Avenue, Campus Box 8115, St. Louis, MO 63110, USA
| | - Xiaobei Zhao
- Center for Cancer Research, University of Tennessee Health Science Center, 19 South Manassas, Memphis, TN 38163, USA
| | - Ashley H Salazar
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, 101 Manning Drive Chapel Hill, NC 27514, USA
| | - Douglas Farquhar
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, 101 Manning Drive Chapel Hill, NC 27514, USA
| | - Siddharth Sheth
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, 101 Manning Drive Chapel Hill, NC 27514, USA
| | - Maheer Masood
- Otolaryngology, University of Kansas Medical Center, 3901 Rainbow Blvd, Kansas City, KS 66160, USA
| | - Michele C Hayward
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, 101 Manning Drive Chapel Hill, NC 27514, USA
| | - Joel S Parker
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, 101 Manning Drive Chapel Hill, NC 27514, USA
| | - Katherine A Hoadley
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, 101 Manning Drive Chapel Hill, NC 27514, USA
| | - Jose Zevallos
- Otolaryngology Head and Neck Surgery, Washington University School of Medicine, 660 South Euclid Avenue, Campus Box 8115, St. Louis, MO 63110, USA
| | - D Neil Hayes
- Center for Cancer Research, University of Tennessee Health Science Center, 19 South Manassas, Memphis, TN 38163, USA
| |
Collapse
|
8
|
Pruneri G, De Braud F, Sapino A, Aglietta M, Vecchione A, Giusti R, Marchiò C, Scarpino S, Baggi A, Bonetti G, Franzini JM, Volpe M, Jommi C. Next-Generation Sequencing in Clinical Practice: Is It a Cost-Saving Alternative to a Single-Gene Testing Approach? PHARMACOECONOMICS - OPEN 2021; 5:285-298. [PMID: 33660227 PMCID: PMC8160052 DOI: 10.1007/s41669-020-00249-0] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 12/12/2020] [Indexed: 05/25/2023]
Abstract
OBJECTIVES This study aimed to compare the costs of a next-generation sequencing-based (NGS-based) panel testing strategy to those of a single-gene testing-based (SGT-based) strategy, considering different scenarios of clinical practice evolution. METHODS Three Italian hospitals were analysed, and four different testing pathways (paths 1, 2, 3, and 4) were identified: two for advanced non-small-cell lung cancer (aNSCLC) patients and two for unresectable metastatic colon-rectal cancer (mCRC) patients. For each path, we explored four scenarios considering the current clinical practice and its expected evolution. The 16 testing cases (4 scenarios × 4 paths) were then compared in terms of differential costs between the NGS-based and SGT-based approaches considering personnel, consumables, equipment, and overhead costs. Break-even and sensitivity analyses were performed. Data gathering, aimed at identifying the hospital setup, was performed through a semi-structured questionnaire administered to the professionals involved in testing activities. RESULTS The NGS-based strategy was found to be a cost-saving alternative to the SGT-based strategy in 15 of the 16 testing cases. The break-even threshold, the minimum number of patients required to make the NGS-based approach less costly than the SGT-based approach, varied across the testing cases depending on molecular alterations tested, techniques adopted, and specific costs. The analysis found the NGS-based approach to be less costly than the SGT-based approach in nine of the 16 testing cases at any volume of tests performed; in six cases, the NGS-based approach was found to be less costly above a threshold (and in one case, it was found to be always more expensive). Savings obtained using an NGS-based approach ranged from €30 to €1249 per patient; in the unique testing case where NGS was more costly, the additional cost per patient was €25. CONCLUSIONS An NGS-based approach may be less costly than an SGT-based approach; also, generated savings increase with the number of patients and different molecular alterations tested.
Collapse
Affiliation(s)
- Giancarlo Pruneri
- Department of the Pathology and Laboratory Medicine, Fondazione IRCCS-Istituto Nazionale dei Tumori, Milan, Italy
- School of Medicine, University of Milan, Milan, Italy
| | - Filippo De Braud
- Department of Oncology, Fondazione IRCCS-Istituto Nazionale dei Tumori, Milan, Italy
- School of Medicine, University of Milan, Milan, Italy
| | - Anna Sapino
- Pathology Unit, Candiolo Cancer Institute-FPO-IRCCS-Candiolo, Turin, Italy
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Massimo Aglietta
- Medical Oncology, Candiolo Cancer Institute-FPO-IRCCS-Candiolo, Turin, Italy
- Department of Oncology, University of Torino, Turin, Italy
| | - Andrea Vecchione
- Pathology Unit, Department of Clinical and Molecular Medicine, St. Andrea University Hospital, University of Rome La Sapienza, Rome, Italy
| | - Raffaele Giusti
- Medical Oncology Unit, St. Andrea University Hospital, Rome, Italy
| | - Caterina Marchiò
- Pathology Unit, Candiolo Cancer Institute-FPO-IRCCS-Candiolo, Turin, Italy
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Stefania Scarpino
- Pathology Unit, Department of Clinical and Molecular Medicine, St. Andrea University Hospital, University of Rome La Sapienza, Rome, Italy
| | - Anna Baggi
- Life Sciences Division, Business Integration Partners, Milan, Italy
| | - Giuseppe Bonetti
- Life Sciences Division, Business Integration Partners, Milan, Italy
| | | | - Marco Volpe
- Life Sciences Division, Business Integration Partners, Milan, Italy
| | - Claudio Jommi
- Cergas, Centre for Research on Health and Social Care Management, SDA Bocconi School of Management, Bocconi University, Milan, Italy.
| |
Collapse
|
9
|
Sullivan SA, Hawkins G, Zhao X, Jo H, Hayes N, Deng X, Bandyopadhyay D, Bae-Jump VL, Rossi EC. Genomic profiling of endometrial cancer and relationship with volume of endometrial cancer disease spread. Gynecol Oncol Rep 2021; 36:100720. [PMID: 33732849 PMCID: PMC7940789 DOI: 10.1016/j.gore.2021.100720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 01/26/2021] [Accepted: 01/31/2021] [Indexed: 11/18/2022] Open
Abstract
OBJECTIVES Lymph node (LN) metastasis and genomic profiles are important prognostic factors in endometrial cancer (EMCA). However, the prognostic significance of low volume metastasis found in sentinel lymph nodes (SLN) is unknown. We sought to determine if genomic mutations were associated with metastatic volume. METHODS Surgically staged women with EC who were enrolled in both a SLN clinical trial and tumor sequencing protocol were eligible. Relevant targets were enriched by a custom designed Agilent SureSelect hybrid capture enrichment library using standard protocols. Three specific gene mutations were evaluated, TP53, PTEN and PIK3CA in the primary tumor of patients with LN negative, LN positive and ITC disease. RESULTS 42 patients were eligible; of these, 7 (16.7%) had ITC only and 7 (16.7%) had micrometastatic or macrometastatic (LN positive) disease. No differences were seen in TP53, PIK3CA or PTEN between groups. All ITC patients with TP53 mutations were of non-endometrioid histology (2/7). Deeper myometrial invasion and lymph vascular space invasion were more likely to occur in the LN positive group (p < 0.01 for both). No patients with ITC had a recurrence in a median 67.7 months of follow-up since surgery. CONCLUSIONS This pilot investigation did not identify differences between frequency of PIK3CA, PTEN or TP53 mutations in tumors and volume of LN metastasis. Low number of ITC limited the ability to detect genomic differences, however mutations appeared to align with expected histology. More work is needed to define the relationship between genomic mutations, histology, ITC, and prognosis.
Collapse
Affiliation(s)
- Stephanie A. Sullivan
- University of North Carolina at Chapel Hill, United States
- Department of Obstetrics and Gynecology, United States
- Division of Gynecologic Oncology, United States
| | - Gabriel Hawkins
- University of North Carolina at Chapel Hill, United States
- Department of Obstetrics and Gynecology, United States
- Division of Gynecologic Oncology, United States
| | - Xiobai Zhao
- University of North Carolina at Chapel Hill, Lineberger Comprehensive Cancer Center, United States
| | - Heejoon Jo
- University of North Carolina at Chapel Hill, Lineberger Comprehensive Cancer Center, United States
| | - Neil Hayes
- University of North Carolina at Chapel Hill, Lineberger Comprehensive Cancer Center, United States
| | - Xiaoyan Deng
- Virginia Commonwealth University, Massey Cancer Center, United States
| | | | - Victoria L. Bae-Jump
- University of North Carolina at Chapel Hill, United States
- Department of Obstetrics and Gynecology, United States
- Division of Gynecologic Oncology, United States
- University of North Carolina at Chapel Hill, Lineberger Comprehensive Cancer Center, United States
| | - Emma C. Rossi
- University of North Carolina at Chapel Hill, United States
- Department of Obstetrics and Gynecology, United States
- Division of Gynecologic Oncology, United States
- University of North Carolina at Chapel Hill, Lineberger Comprehensive Cancer Center, United States
| |
Collapse
|
10
|
Beaty BT, Moon DH, Shen CJ, Amdur RJ, Weiss J, Grilley-Olson J, Patel S, Zanation A, Hackman TG, Thorp B, Blumberg JM, Patel SN, Weissler MC, Yarbrough WG, Sheets NC, Parker JS, Neil Hayes D, Weck KE, Ramkissoon LA, Mendenhall WM, Dagan R, Tan X, Gupta GP, Chera BS. PIK3CA Mutation in HPV-Associated OPSCC Patients Receiving Deintensified Chemoradiation. J Natl Cancer Inst 2021; 112:855-858. [PMID: 31747025 DOI: 10.1093/jnci/djz224] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2019] [Revised: 08/14/2019] [Accepted: 11/19/2019] [Indexed: 12/13/2022] Open
Abstract
PIK3CA is the most frequently mutated gene in human papillomavirus (HPV)-associated oropharyngeal squamous cell carcinoma (OPSCC). Prognostic implications of such mutations remain unknown. We sought to elucidate the clinical significance of PIK3CA mutations in HPV-associated OPSCC patients treated with definitive chemoradiation (CRT). Seventy-seven patients with HPV-associated OPSCC were enrolled on two phase II clinical trials of deintensified CRT (60 Gy intensity-modulated radiotherapy with concurrent weekly cisplatin). Targeted next-generation sequencing was performed. Of the 77 patients, nine had disease recurrence (two regional, four distant, three regional and distant). Thirty-four patients had mutation(s) identified; 16 had PIK3CA mutations. Patients with wild-type-PIK3CA had statistically significantly higher 3-year disease-free survival than PIK3CA-mutant patients (93.4%, 95% confidence interval [CI] = 85.0% to 99.9% vs 68.8%, 95% CI = 26.7% to 89.8%; P = .004). On multivariate analysis, PIK3CA mutation was the only variable statistically significantly associated with disease recurrence (hazard ratio = 5.71, 95% CI = 1.53 to 21.3; P = .01). PIK3CA mutation is associated with worse disease-free survival in a prospective cohort of newly diagnosed HPV-associated OPSCC patients treated with deintensified CRT.
Collapse
Affiliation(s)
- Brian T Beaty
- Department of Radiation Oncology, University of North Carolina Hospitals, Chapel Hill, NC
| | - Dominic H Moon
- Department of Radiation Oncology, University of North Carolina Hospitals, Chapel Hill, NC.,University of North Carolina, Chapel Hill, NC.,Department of Medical Oncology, University of Tennessee, Memphis, TN
| | - Colette J Shen
- Department of Radiation Oncology, University of North Carolina Hospitals, Chapel Hill, NC
| | - Robert J Amdur
- Department of Radiation Oncology, University of Florida College of Medicine, Gainesville, FL
| | | | | | | | | | | | - Brian Thorp
- Department of Otolaryngology/Head and Neck Surgery
| | | | | | | | | | - Nathan C Sheets
- Department of Radiation Oncology, University of North Carolina Hospitals, Chapel Hill, NC
| | | | | | | | | | - William M Mendenhall
- Department of Radiation Oncology, University of Florida College of Medicine, Gainesville, FL
| | - Roi Dagan
- University of Florida Health Proton Therapy Institute, Jacksonville, FL
| | | | - Gaorav P Gupta
- Department of Radiation Oncology, University of North Carolina Hospitals, Chapel Hill, NC
| | - Bhishamjit S Chera
- Department of Radiation Oncology, University of North Carolina Hospitals, Chapel Hill, NC
| |
Collapse
|
11
|
Trembath DG, Davis ES, Rao S, Bradler E, Saada AF, Midkiff BR, Snavely AC, Ewend MG, Collichio FA, Lee CB, Karachaliou GS, Ayvali F, Ollila DW, Krauze MT, Kirkwood JM, Vincent BG, Nikolaishvilli-Feinberg N, Moschos SJ. Brain Tumor Microenvironment and Angiogenesis in Melanoma Brain Metastases. Front Oncol 2021; 10:604213. [PMID: 33552976 PMCID: PMC7860978 DOI: 10.3389/fonc.2020.604213] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 11/17/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND High tumor-infiltrating lymphocytes (TILs) and hemorrhage are important prognostic factors in patients who have undergone craniotomy for melanoma brain metastases (MBM) before 2011 at the University of Pittsburgh Medical Center (UPMC). We have investigated the prognostic or predictive role of these histopathologic factors in a more contemporary craniotomy cohort from the University of North Carolina at Chapel Hill (UNC-CH). We have also sought to understand better how various immune cell subsets, angiogenic factors, and blood vessels may be associated with clinical and radiographic features in MBM. METHODS Brain tumors from the UPMC and UNC-CH patient cohorts were (re)analyzed by standard histopathology, tumor tissue imaging, and gene expression profiling. Variables were associated with overall survival (OS) and radiographic features. RESULTS The patient subgroup with high TILs in craniotomy specimens and subsequent treatment with immune checkpoint inhibitors (ICIs, n=7) trended to have longer OS compared to the subgroup with high TILs and no treatment with ICIs (n=11, p=0.059). Bleeding was significantly associated with tumor volume before craniotomy, high melanoma-specific expression of basic fibroblast growth factor (bFGF), and high density of CD31+αSMA- blood vessels. Brain tumors with high versus low peritumoral edema before craniotomy had low (17%) versus high (41%) incidence of brisk TILs. Melanoma-specific expression of the vascular endothelial growth factor (VEGF) was comparable to VEGF expression by TILs and was not associated with any particular prognostic, radiographic, or histopathologic features. A gene signature associated with gamma delta (gd) T cells was significantly higher in intracranial than same-patient extracranial metastases and primary melanoma. However, gdT cell density in MBM was not prognostic. CONCLUSIONS ICIs may provide greater clinical benefit in patients with brisk TILs in MBM. Intratumoral hemorrhage in brain metastases, a significant clinical problem, is not merely associated with tumor volume but also with underlying biology. bFGF may be an essential pathway to target. VEGF, a factor principally associated with peritumoral edema, is not only produced by melanoma cells but also by TILs. Therefore, suppressing low-grade peritumoral edema using corticosteroids may harm TIL function in 41% of cases. Ongoing clinical trials targeting VEGF in MBM may predict a lack of unfavorable impacts on TIL density and/or intratumoral hemorrhage.
Collapse
Affiliation(s)
- Dimitri G. Trembath
- Departments of Pathology and Laboratory Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Eric S. Davis
- Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Shanti Rao
- University of North Carolina School of Medicine, Chapel Hill, NC, United States
| | - Evan Bradler
- University of North Carolina School of Medicine, Chapel Hill, NC, United States
| | - Angelica F. Saada
- State University of New York Downstate Medical Center College of Medicine, Brooklyn, NY, United States
| | - Bentley R. Midkiff
- Translational Pathology Laboratory, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Anna C. Snavely
- Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Matthew G. Ewend
- Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Department of Neurosurgery, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Frances A. Collichio
- Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Department of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Carrie B. Lee
- Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Department of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Georgia-Sofia Karachaliou
- Department of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Fatih Ayvali
- Department of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - David W. Ollila
- Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Department of Surgery, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Michal T. Krauze
- Melanoma and Skin Cancer Program, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
| | - John M. Kirkwood
- Melanoma and Skin Cancer Program, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
| | - Benjamin G. Vincent
- Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Department of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Nana Nikolaishvilli-Feinberg
- Translational Pathology Laboratory, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Stergios J. Moschos
- Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Department of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| |
Collapse
|
12
|
Edris Sharif Rahmani, Azarpara H, Abazari MF, Mohajeri MR, Nasimi M, Ghorbani R, Azizpour A, Rahimi H. Novel Mutation C.7348C>T in NF1 Gene Identified by Whole-Exome Sequencing in Patient with Overlapping Clinical Symptoms of Neurofibromatosis Type 1 and Bannayan–Riley–Ruvalcaba Syndrome. CYTOL GENET+ 2020. [DOI: 10.3103/s0095452720040106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
13
|
Patel NM, Jo H, Eberhard DA, Yin X, Hayward MC, Stein MK, Hayes DN, Grilley-Olson JE. Improved Tumor Purity Metrics in Next-generation Sequencing for Clinical Practice: The Integrated Interpretation of Neoplastic Cellularity and Sequencing Results (IINCaSe) Approach. Appl Immunohistochem Mol Morphol 2020; 27:764-772. [PMID: 30102605 PMCID: PMC6887630 DOI: 10.1097/pai.0000000000000684] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Accepted: 05/08/2018] [Indexed: 12/18/2022]
Abstract
Neoplastic cellularity contributes to the analytic sensitivity of most present technologies for mutation detection, such that they underperform when stroma and inflammatory cells dilute a cancer specimen's variant fraction. Thus, tumor purity assessment by light microscopy is used to determine sample adequacy before sequencing and to interpret the significance of negative results and mutant allele fraction afterwards. However, pathologist estimates of tumor purity are imprecise and have limited reproducibility. With the advent of massively parallel sequencing, large amounts of molecular data can be analyzed by computational purity algorithms. We retrospectively compared tumor purity of 3 computational algorithms with neoplastic cellularity using hematoxylin and eosin light microscopy to determine which was best for clinical evaluation of molecular profiling. Data were analyzed from 881 cancer patients from a clinical trial cohort, LCCC1108 (UNCseq), whose tumors had targeted massively parallel sequencing. Concordance among algorithms was poor, and the specimens analyzed had high rates of algorithm failure partially due to variable tumor purity. Computational tumor purity estimates did not add value beyond the pathologist's estimate of neoplastic cellularity microscopy. To improve present methods, we propose a semiquantitative, clinically applicable strategy based on mutant allele fraction and copy number changes present within a given specimen, which when combined with the morphologic tumor purity estimate, guide the interpretation of next-generation sequencing results in cancer patients.
Collapse
Affiliation(s)
- Nirali M. Patel
- Department of Pathology and Laboratory Medicine
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC
| | - Heejoon Jo
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC
| | - David A. Eberhard
- Department of Pathology and Laboratory Medicine
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC
| | - Xiaoying Yin
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC
| | - Michele C. Hayward
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC
| | - Matthew K. Stein
- Department of Internal Medicine, Division of Hematology and Oncology
- West Cancer Center, University of Tennessee Health Science Center, Memphis, TN
| | - David Neil Hayes
- Department of Internal Medicine, Division of Hematology and Oncology
- West Cancer Center, University of Tennessee Health Science Center, Memphis, TN
| | - Juneko E. Grilley-Olson
- Department of Internal Medicine, Division of Hematology and Oncology, University of North Carolina School of Medicine
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC
| |
Collapse
|
14
|
Leslie PL, Chao YL, Tsai YH, Ghosh SK, Porrello A, Van Swearingen AED, Harrison EB, Cooley BC, Parker JS, Carey LA, Pecot CV. Histone deacetylase 11 inhibition promotes breast cancer metastasis from lymph nodes. Nat Commun 2019; 10:4192. [PMID: 31519896 PMCID: PMC6744422 DOI: 10.1038/s41467-019-12222-5] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Accepted: 08/27/2019] [Indexed: 01/28/2023] Open
Abstract
Lymph node (LN) metastases correspond with a worse prognosis in nearly all cancers, yet the occurrence of cancer spreading from LNs remains controversial. Additionally, the mechanisms explaining how cancers survive and exit LNs are largely unknown. Here, we show that breast cancer patients frequently have LN metastases that closely resemble distant metastases. In addition, using a microsurgical model, we show how LN metastasis development and dissemination is regulated by the expression of a chromatin modifier, histone deacetylase 11 (HDAC11). Genetic and pharmacologic blockade of HDAC11 decreases LN tumor growth, yet substantially increases migration and distant metastasis formation. Collectively, we reveal a mechanism explaining how HDAC11 plasticity promotes breast cancer growth as well as dissemination from LNs and suggest caution with the use of HDAC inhibitors. The prognosis of cancer patients with lymph node (LN) metastasis is worse than those without. Here, the authors report that while histone deacetylase 11 (HDAC11) inhibition suppresses tumor growth within the LN, it also promotes cancer cell migration out of the LN to form distant metastasis, and therefore suggest caution with HDAC inhibitors.
Collapse
Affiliation(s)
- Patrick L Leslie
- UNC Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Yvonne L Chao
- UNC Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Yi-Hsuan Tsai
- UNC Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Subrata K Ghosh
- UNC Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Alessandro Porrello
- UNC Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Amanda E D Van Swearingen
- UNC Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Emily B Harrison
- UNC Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Brian C Cooley
- UNC Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.,Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Joel S Parker
- UNC Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.,Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Lisa A Carey
- UNC Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.,Division of Hematology and Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Chad V Pecot
- UNC Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA. .,Division of Hematology and Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
| |
Collapse
|
15
|
Chera BS, Kumar S, Beaty BT, Marron D, Jefferys S, Green R, Goldman EC, Amdur R, Sheets N, Dagan R, Hayes DN, Weiss J, Grilley-Olson JE, Zanation A, Hackman T, Blumberg JM, Patel S, Weissler M, Tan XM, Parker JS, Mendenhall W, Gupta GP. Rapid Clearance Profile of Plasma Circulating Tumor HPV Type 16 DNA during Chemoradiotherapy Correlates with Disease Control in HPV-Associated Oropharyngeal Cancer. Clin Cancer Res 2019; 25:4682-4690. [PMID: 31088830 DOI: 10.1158/1078-0432.ccr-19-0211] [Citation(s) in RCA: 233] [Impact Index Per Article: 38.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Revised: 03/22/2019] [Accepted: 05/08/2019] [Indexed: 12/17/2022]
Abstract
PURPOSE To identify a profile of circulating tumor human papilloma virus (HPV) DNA (ctHPVDNA) clearance kinetics that is associated with disease control after chemoradiotherapy (CRT) for HPV-associated oropharyngeal squamous cell carcinoma (OPSCC). EXPERIMENTAL DESIGN A multi-institutional prospective biomarker trial was conducted in 103 patients with (i) p16-positive OPSCC, (ii) M0 disease, and (iii) receipt of definitive CRT. Blood specimens were collected at baseline, weekly during CRT, and at follow-up visits. Optimized multianalyte digital PCR assays were used to quantify ctHPVDNA (types 16/18/31/33/35) in plasma. A control cohort of 55 healthy volunteers and 60 patients with non-HPV-associated malignancy was also analyzed. RESULTS Baseline plasma ctHPVDNA had high specificity (97%) and high sensitivity (89%) for detecting newly diagnosed HPV-associated OPSCC. Pretreatment ctHPV16DNA copy number correlated with disease burden, tumor HPV copy number, and HPV integration status. We define a ctHPV16DNA favorable clearance profile as having high baseline copy number (>200 copies/mL) and >95% clearance of ctHPV16DNA by day 28 of CRT. Nineteen of 67 evaluable patients had a ctHPV16DNA favorable clearance profile, and none had persistent or recurrent regional disease after CRT. In contrast, patients with adverse clinical risk factors (T4 or >10 pack years) and an unfavorable ctHPV16DNA clearance profile had a 35% actuarial rate of persistent or recurrent regional disease after CRT (P = 0.0049). CONCLUSIONS A rapid clearance profile of ctHPVDNA may predict likelihood of disease control in patients with HPV-associated OPSCC patients treated with definitive CRT and may be useful in selecting patients for deintensified therapy.
Collapse
Affiliation(s)
- Bhishamjit S Chera
- Department of Radiation Oncology, University of North Carolina School of Medicine, Chapel Hill, North Carolina. .,Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina
| | - Sunil Kumar
- Department of Radiation Oncology, University of North Carolina School of Medicine, Chapel Hill, North Carolina.,Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina
| | - Brian T Beaty
- Department of Radiation Oncology, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| | - David Marron
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina.,Lineberger Bioinformatics Core, University of North Carolina Hospitals, Chapel Hill, North Carolina
| | - Stuart Jefferys
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina.,Lineberger Bioinformatics Core, University of North Carolina Hospitals, Chapel Hill, North Carolina
| | - Rebecca Green
- Department of Radiation Oncology, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| | - Emily C Goldman
- Department of Radiation Oncology, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| | - Robert Amdur
- Department of Radiation Oncology, University of Florida Hospitals, Gainesville, Florida
| | - Nathan Sheets
- Department of Radiation Oncology, UNC Rex Hospitals, Raleigh, North Carolina
| | - Roi Dagan
- University of Florida Health Proton Therapy Institute, Jacksonville, Florida
| | - D Neil Hayes
- West Cancer Center, University of Tennessee, Memphis, Tennessee
| | - Jared Weiss
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina.,Division of Hematology Oncology, Department of Medicine, University of North Carolina, School of Medicine, Chapel Hill, North Carolina
| | - Juneko E Grilley-Olson
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina.,Division of Hematology Oncology, Department of Medicine, University of North Carolina, School of Medicine, Chapel Hill, North Carolina
| | - Adam Zanation
- Department of Otolaryngology/Head and Neck Surgery, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| | - Trevor Hackman
- Department of Otolaryngology/Head and Neck Surgery, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| | - Jeffrey M Blumberg
- Department of Otolaryngology/Head and Neck Surgery, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| | - Samip Patel
- Department of Otolaryngology/Head and Neck Surgery, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| | - Mark Weissler
- Department of Otolaryngology/Head and Neck Surgery, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| | - Xianming M Tan
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina.,Department of Biostatistics, University of North Carolina, Chapel Hill, North Carolina
| | - Joel S Parker
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina.,Lineberger Bioinformatics Core, University of North Carolina Hospitals, Chapel Hill, North Carolina.,Department of Genetics, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| | - William Mendenhall
- Department of Radiation Oncology, University of Florida Hospitals, Gainesville, Florida
| | - Gaorav P Gupta
- Department of Radiation Oncology, University of North Carolina School of Medicine, Chapel Hill, North Carolina. .,Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina
| |
Collapse
|
16
|
Misra P, Singh S. Role of cytokines in combinatorial immunotherapeutics of non-small cell lung cancer through systems perspective. Cancer Med 2019; 8:1976-1995. [PMID: 30997737 PMCID: PMC6536974 DOI: 10.1002/cam4.2112] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Revised: 02/22/2019] [Accepted: 03/07/2019] [Indexed: 12/21/2022] Open
Abstract
Lung cancer is the leading cause of deaths related to cancer and accounts for more than a million deaths per year. Various new strategies have been developed and adapted for treatment; still the survival for 5 years is just 16% in patients with non‐small cell lung cancer (NSCLC). Most of these strategies to combat NSCLC whether it is a drug molecule or immunotherapy/vaccine candidate require a big cost and time. Integration of computational modeling with systems biology has opened new avenues for understanding complex cancer biology. Resolving the complex interactions of various pathways and their crosstalk leading to oncogenic changes could identify new therapeutic targets with lesser cost and time. Herein, this review provides an overview of various aspects of NSCLC along with available strategies for its cure concluding with our insight into how systems approach could serve as a therapeutic intervention dissecting the immunologic parameters and cross talk between various pathways involved.
Collapse
Affiliation(s)
- Pragya Misra
- National Centre for Cell ScienceSP Pune University CampusPuneIndia
| | - Shailza Singh
- National Centre for Cell ScienceSP Pune University CampusPuneIndia
| |
Collapse
|
17
|
Lu J, Han B. Liquid Biopsy Promotes Non-Small Cell Lung Cancer Precision Therapy. Technol Cancer Res Treat 2019; 17:1533033818801809. [PMID: 30244652 PMCID: PMC6153525 DOI: 10.1177/1533033818801809] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The range of potential applications of liquid biopsies for non-small cell lung cancer management is expanded by the use of circulating tumor deoxyribonucleic acid and circulating tumor cells. Principal studies have demonstrated the predictive accuracy of droplet digital polymerase chain reaction detection, next-generation sequencing, and circulating tumor cells detection in patients with non-small cell lung cancer. The translational potential of these liquid biopsy technologies promotes the improvement of sensitivity and specificity in genomic and molecular methods. Here, we highlight the realities and challenges associated with the use of liquid biopsies for the detection of non-small cell lung cancer in patients. However, liquid biopsy technologies including circulating tumor cells detection, droplet digital polymerase chain reaction detection, and next-generation sequencing detection for precision therapy in non-small cell lung cancer will show substantive clinical applications in the future.
Collapse
Affiliation(s)
- Jun Lu
- 1 Department of Pulmonary Medicine, Shanghai Chest Hospital, Shanghai Jiaotong University, Shanghai, China
| | - Baohui Han
- 1 Department of Pulmonary Medicine, Shanghai Chest Hospital, Shanghai Jiaotong University, Shanghai, China
| |
Collapse
|
18
|
Zhao X, Little P, Hoyle AP, Pegna GJ, Hayward MC, Ivanova A, Parker JS, Marron DL, Soloway MG, Jo H, Salazar AH, Papakonstantinou MP, Bouchard DM, Jefferys SR, Hoadley KA, Ollila DW, Frank JS, Thomas NE, Googe PB, Ezzell AJ, Collichio FA, Lee CB, Earp HS, Sharpless NE, Hugo W, Wilmott JS, Quek C, Waddell N, Johansson PA, Thompson JF, Hayward NK, Mann GJ, Lo RS, Johnson DB, Scolyer RA, Hayes DN, Moschos SJ. The Prognostic Significance of Low-Frequency Somatic Mutations in Metastatic Cutaneous Melanoma. Front Oncol 2019; 8:584. [PMID: 30662871 PMCID: PMC6329304 DOI: 10.3389/fonc.2018.00584] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2018] [Accepted: 11/19/2018] [Indexed: 12/12/2022] Open
Abstract
Background: Little is known about the prognostic significance of somatically mutated genes in metastatic melanoma (MM). We have employed a combined clinical and bioinformatics approach on tumor samples from cutaneous melanoma (SKCM) as part of The Cancer Genome Atlas project (TCGA) to identify mutated genes with potential clinical relevance. Methods: After limiting our DNA sequencing analysis to MM samples (n = 356) and to the CANCER CENSUS gene list, we filtered out mutations with low functional significance (snpEFF). We performed Cox analysis on 53 genes that were mutated in ≥3% of samples, and had ≥50% difference in incidence of mutations in deceased subjects versus alive subjects. Results: Four genes were potentially prognostic [RAC1, FGFR1, CARD11, CIITA; false discovery rate (FDR) < 0.2]. We identified 18 additional genes (e.g., SPEN, PDGFRB, GNAS, MAP2K1, EGFR, TSC2) that were less likely to have prognostic value (FDR < 0.4). Most somatic mutations in these 22 genes were infrequent (< 10%), associated with high somatic mutation burden, and were evenly distributed across all exons, except for RAC1 and MAP2K1. Mutations in only 9 of these 22 genes were also identified by RNA sequencing in >75% of the samples that exhibited corresponding DNA mutations. The low frequency, UV signature type and RNA expression of the 22 genes in MM samples were confirmed in a separate multi-institution validation cohort (n = 413). An underpowered analysis within a subset of this validation cohort with available patient follow-up (n = 224) showed that somatic mutations in SPEN and RAC1 reached borderline prognostic significance [log-rank favorable (p = 0.09) and adverse (p = 0.07), respectively]. Somatic mutations in SPEN, and to a lesser extent RAC1, were not associated with definite gene copy number or RNA expression alterations. High (>2+) nuclear plus cytoplasmic expression intensity for SPEN was associated with longer melanoma-specific overall survival (OS) compared to lower (≤ 2+) nuclear intensity (p = 0.048). We conclude that expressed somatic mutations in infrequently mutated genes beyond the well-characterized ones (e.g., BRAF, RAS, CDKN2A, PTEN, TP53), such as RAC1 and SPEN, may have prognostic significance in MM.
Collapse
Affiliation(s)
- Xiaobei Zhao
- Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Paul Little
- Department of Biostatistics, Gillings School of Public Health, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Alan P. Hoyle
- Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Guillaume J. Pegna
- Division of Hematology/Oncology, Department of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Michele C. Hayward
- Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Anastasia Ivanova
- Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Department of Biostatistics, Gillings School of Public Health, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Joel S. Parker
- Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - David L. Marron
- Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Matthew G. Soloway
- Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Heejoon Jo
- Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Ashley H. Salazar
- Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Michael P. Papakonstantinou
- Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Deeanna M. Bouchard
- Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Stuart R. Jefferys
- Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Katherine A. Hoadley
- Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - David W. Ollila
- Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Division of Surgical Oncology, Department of Surgery, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Melanoma Program, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Jill S. Frank
- Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Melanoma Program, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Nancy E. Thomas
- Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Melanoma Program, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Department of Dermatology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Paul B. Googe
- Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Melanoma Program, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Department of Dermatology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Ashley J. Ezzell
- Department of Cell Biology & Physiology, Histology Research Core Facility, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Frances A. Collichio
- Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Division of Hematology/Oncology, Department of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Melanoma Program, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Carrie B. Lee
- Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Division of Hematology/Oncology, Department of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Melanoma Program, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - H. Shelton Earp
- Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Norman E. Sharpless
- Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Willy Hugo
- Division of Dermatology, Department of Medicine, Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA, United States
| | - James S. Wilmott
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW, Australia
| | - Camelia Quek
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW, Australia
| | - Nicola Waddell
- Queensland Institute of Medical Research-QIMR Berghofer Medical Research Institute, Herston, QLD, Australia
| | - Peter A. Johansson
- Queensland Institute of Medical Research-QIMR Berghofer Medical Research Institute, Herston, QLD, Australia
| | - John F. Thompson
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW, Australia
| | - Nicholas K. Hayward
- Queensland Institute of Medical Research-QIMR Berghofer Medical Research Institute, Herston, QLD, Australia
| | - Graham J. Mann
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW, Australia
| | - Roger S. Lo
- Division of Dermatology, Department of Medicine, Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA, United States
| | - Douglas B. Johnson
- Department of Medicine, Vanderbilt-Ingram Cancer Center, Nashville, TN, United States
| | - Richard A. Scolyer
- Queensland Institute of Medical Research-QIMR Berghofer Medical Research Institute, Herston, QLD, Australia
| | - D. Neil Hayes
- Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Division of Hematology/Oncology, Department of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Stergios J. Moschos
- Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Division of Hematology/Oncology, Department of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Melanoma Program, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| |
Collapse
|
19
|
Rose TL, Chism DD, Alva AS, Deal AM, Maygarden SJ, Whang YE, Kardos J, Drier A, Basch E, Godley PA, Dunn MW, Kim WY, Milowsky MI. Phase II trial of palbociclib in patients with metastatic urothelial cancer after failure of first-line chemotherapy. Br J Cancer 2018; 119:801-807. [PMID: 30293995 PMCID: PMC6189143 DOI: 10.1038/s41416-018-0229-0] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Revised: 07/09/2018] [Accepted: 07/19/2018] [Indexed: 12/31/2022] Open
Abstract
Background The majority of urothelial cancers (UC) harbor alterations in retinoblastoma (Rb) pathway genes that can lead to loss of Rb tumour suppressor function. Palbociclib is an oral, selective inhibitor of CDK 4/6 that restores Rb function and promotes cell cycle arrest. Methods In this phase II trial, patients with metastatic platinum-refractory UC molecularly selected for p16 loss and intact Rb by tumour immunohistochemistry received palbociclib 125 mg p.o. daily for 21 days of a 28-day cycle. Primary endpoint was progression-free survival at 4 months (PFS4) using a Simon’s two-stage design. Next-generation sequencing including Rb pathway alterations was conducted. Results Twelve patients were enrolled and two patients (17%) achieved PFS4 with insufficient activity to advance to stage 2. No responses were seen. Median PFS was 1.9 months (95% CI 1.8–3.7 months) and median overall survival was 6.3 months (95% CI 2.2–12.6 months). Fifty-eight percent of patients had grade ≥3 hematologic toxicity. There were no CDKN2A alterations found and no correlation of Rb pathway alterations with clinical outcome. Conclusions Palbociclib did not demonstrate meaningful activity in selected patients with platinum-refractory metastatic UC. Further development of palbociclib should only be considered with improved integral biomarker selection or in rational combination with other therapies.
Collapse
Affiliation(s)
- Tracy L Rose
- Division of Hematology/Oncology, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - David D Chism
- Division of Hematology and Oncology, Department of Medicine, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Ajjai S Alva
- Division of Hematology/Oncology, University of Michigan, Ann Arbor, MI, USA
| | - Allison M Deal
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Susan J Maygarden
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Young E Whang
- Division of Hematology/Oncology, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Jordan Kardos
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Anthony Drier
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Ethan Basch
- Division of Hematology/Oncology, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Paul A Godley
- Division of Hematology/Oncology, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Mary W Dunn
- Division of Hematology/Oncology, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - William Y Kim
- Division of Hematology/Oncology, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Matthew I Milowsky
- Division of Hematology/Oncology, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| |
Collapse
|
20
|
Zhang J, Wu T, Simon J, Takada M, Saito R, Fan C, Liu XD, Jonasch E, Xie L, Chen X, Yao X, Teh BT, Tan P, Zheng X, Li M, Lawrence C, Fan J, Geng J, Liu X, Hu L, Wang J, Liao C, Hong K, Zurlo G, Parker JS, Auman JT, Perou CM, Rathmell WK, Kim WY, Kirschner MW, Kaelin WG, Baldwin AS, Zhang Q. VHL substrate transcription factor ZHX2 as an oncogenic driver in clear cell renal cell carcinoma. Science 2018; 361:290-295. [PMID: 30026228 DOI: 10.1126/science.aap8411] [Citation(s) in RCA: 139] [Impact Index Per Article: 19.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Accepted: 06/06/2018] [Indexed: 12/11/2022]
Abstract
Inactivation of the von Hippel-Lindau (VHL) E3 ubiquitin ligase protein is a hallmark of clear cell renal cell carcinoma (ccRCC). Identifying how pathways affected by VHL loss contribute to ccRCC remains challenging. We used a genome-wide in vitro expression strategy to identify proteins that bind VHL when hydroxylated. Zinc fingers and homeoboxes 2 (ZHX2) was found as a VHL target, and its hydroxylation allowed VHL to regulate its protein stability. Tumor cells from ccRCC patients with VHL loss-of-function mutations usually had increased abundance and nuclear localization of ZHX2. Functionally, depletion of ZHX2 inhibited VHL-deficient ccRCC cell growth in vitro and in vivo. Mechanistically, integrated chromatin immunoprecipitation sequencing and microarray analysis showed that ZHX2 promoted nuclear factor κB activation. These studies reveal ZHX2 as a potential therapeutic target for ccRCC.
Collapse
Affiliation(s)
- Jing Zhang
- Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA.,Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Tao Wu
- Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Jeremy Simon
- Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA.,Department of Genetics, Neuroscience Center, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Mamoru Takada
- Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - Ryoichi Saito
- Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - Cheng Fan
- Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - Xian-De Liu
- Departments of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Eric Jonasch
- Departments of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Ling Xie
- Department of Biochemistry and Biophysics, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Xian Chen
- Department of Biochemistry and Biophysics, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Xiaosai Yao
- Cancer Therapeutics and Stratified Oncology, Genome Institute of Singapore, Singapore 138672, Singapore.,Institute of Molecular and Cell Biology, Singapore 138673, Singapore
| | - Bin Tean Teh
- Institute of Molecular and Cell Biology, Singapore 138673, Singapore.,Laboratory of Cancer Epigenome, Department of Medical Sciences, National Cancer Centre, Singapore 169610, Singapore.,Cancer and Stem Cell Biology Program, Duke-NUS Medical School, Singapore 169857, Singapore
| | - Patrick Tan
- Cancer Therapeutics and Stratified Oncology, Genome Institute of Singapore, Singapore 138672, Singapore.,Cancer and Stem Cell Biology Program, Duke-NUS Medical School, Singapore 169857, Singapore
| | - Xingnan Zheng
- Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - Mingjie Li
- Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - Cortney Lawrence
- Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - Jie Fan
- Department of Pathology, Huashan Hospital, Fudan University, Shanghai, China
| | - Jiang Geng
- Department of Urology, Shanghai Tenth People's Hospital, Tongji University, Shanghai, China
| | - Xijuan Liu
- Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - Lianxin Hu
- Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - Jun Wang
- Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - Chengheng Liao
- Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - Kai Hong
- Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - Giada Zurlo
- Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - Joel S Parker
- Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - J Todd Auman
- Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - Charles M Perou
- Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - W Kimryn Rathmell
- Division of Hematology and Oncology, Department of Medicine, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - William Y Kim
- Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - Marc W Kirschner
- Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA
| | - William G Kaelin
- Howard Hughes Medical Institute, Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA
| | - Albert S Baldwin
- Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - Qing Zhang
- Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA. .,Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA.,Department of Pharmacology, University of North Carolina, Chapel Hill, NC 27599, USA
| |
Collapse
|
21
|
LCCC 1025: a phase II study of everolimus, trastuzumab, and vinorelbine to treat progressive HER2-positive breast cancer brain metastases. Breast Cancer Res Treat 2018; 171:637-648. [PMID: 29938395 DOI: 10.1007/s10549-018-4852-5] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Accepted: 06/07/2018] [Indexed: 01/09/2023]
Abstract
PURPOSE HER2 + breast cancer (BC) is an aggressive subtype with high rates of brain metastases (BCBM). Two-thirds of HER2 + BCBM demonstrate activation of the PI3K/mTOR pathway driving resistance to anti-HER2 therapy. This phase II study evaluated everolimus (E), a brain-permeable mTOR inhibitor, trastuzumab (T), and vinorelbine (V) in patients with HER2 + BCBM. PATIENTS AND METHODS Eligible patients had progressive HER2 + BCBM. The primary endpoint was intracranial response rate (RR); secondary objectives were CNS clinical benefit rate (CBR), extracranial RR, time to progression (TTP), overall survival (OS), and targeted sequencing of tumors from enrolled patients. A two-stage design distinguished intracranial RR of 5% versus 20%. RESULTS 32 patients were evaluable for toxicity, 26 for efficacy. Intracranial RR was 4% (1 PR). CNS CBR at 6 mos was 27%; at 3 mos 65%. Median intracranial TTP was 3.9 mos (95% CI 2.2-5). OS was 12.2 mos (95% CI 0.6-20.2). Grade 3-4 toxicities included neutropenia (41%), anemia (16%), and stomatitis (16%). Mutations in TP53 and PIK3CA were common in BCBM. Mutations in the PI3K/mTOR pathway were not associated with response. ERBB2 amplification was higher in BCBM compared to primary BC; ERBB2 amplification in the primary BC trended toward worse OS. CONCLUSION While intracranial RR to ETV was low in HER2 + BCBM patients, one-third achieved CNS CBR; TTP/OS was similar to historical control. No new toxicity signals were observed. Further analysis of the genomic underpinnings of BCBM to identify tractable prognostic and/or predictive biomarkers is warranted. CLINICAL TRIAL (NCT01305941).
Collapse
|
22
|
Coombs CC, Gillis NK, Tan X, Berg JS, Ball M, Balasis ME, Montgomery ND, Bolton KL, Parker JS, Mesa TE, Yoder SJ, Hayward MC, Patel NM, Richards KL, Walko CM, Knepper TC, Soper JT, Weiss J, Grilley-Olson JE, Kim WY, Earp HS, Levine RL, Papaemmanuil E, Zehir A, Hayes DN, Padron E. Identification of Clonal Hematopoiesis Mutations in Solid Tumor Patients Undergoing Unpaired Next-Generation Sequencing Assays. Clin Cancer Res 2018; 24:5918-5924. [PMID: 29866652 DOI: 10.1158/1078-0432.ccr-18-1201] [Citation(s) in RCA: 86] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Revised: 05/16/2018] [Accepted: 06/01/2018] [Indexed: 02/03/2023]
Abstract
PURPOSE In this era of precision-based medicine, for optimal patient care, results reported from commercial next-generation sequencing (NGS) assays should adequately reflect the burden of somatic mutations in the tumor being sequenced. Here, we sought to determine the prevalence of clonal hematopoiesis leading to possible misattribution of tumor mutation calls on unpaired Foundation Medicine NGS assays. EXPERIMENTAL DESIGN This was a retrospective cohort study of individuals undergoing NGS of solid tumors from two large cancer centers. We identified and quantified mutations in genes known to be frequently altered in clonal hematopoiesis (DNMT3A, TET2, ASXL1, TP53, ATM, CHEK2, SF3B1, CBL, JAK2) that were returned to physicians on clinical Foundation Medicine reports. For a subset of patients, we explored the frequency of true clonal hematopoiesis by comparing mutations on Foundation Medicine reports with matched blood sequencing. RESULTS Mutations in genes that are frequently altered in clonal hematopoiesis were identified in 65% (1,139/1,757) of patients undergoing NGS. When excluding TP53, which is often mutated in solid tumors, these events were still seen in 35% (619/1,757) of patients. Utilizing paired blood specimens, we were able to confirm that 8% (18/226) of mutations reported in these genes were true clonal hematopoiesis events. The majority of DNMT3A mutations (64%, 7/11) and minority of TP53 mutations (4%, 2/50) were clonal hematopoiesis. CONCLUSIONS Clonal hematopoiesis mutations are commonly reported on unpaired NGS testing. It is important to recognize clonal hematopoiesis as a possible cause of misattribution of mutation origin when applying NGS findings to a patient's care.See related commentary by Pollyea, p. 5790.
Collapse
Affiliation(s)
- Catherine C Coombs
- Department of Medicine, Division of Hematology and Oncology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina. .,Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Nancy K Gillis
- Department of Cancer Epidemiology, Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Xianming Tan
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Jonathan S Berg
- Department of Medicine, Division of Hematology and Oncology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina.,Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina.,Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Markus Ball
- Department of Malignant Hematology, Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Maria E Balasis
- Department of Malignant Hematology, Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Nathan D Montgomery
- Department of Pathology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Kelly L Bolton
- Memorial Sloan Kettering Cancer Center, New York, New York
| | - Joel S Parker
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina.,Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Tania E Mesa
- Molecular Genomics Core, Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Sean J Yoder
- Molecular Genomics Core, Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Michele C Hayward
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Nirali M Patel
- Department of Pathology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina.,Q Solutions - EA Genomics, Morrisville, North Carolina
| | - Kristy L Richards
- Department of Medicine, Division of Hematology and Oncology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina.,Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina.,College of Veterinary Medicine, Cornell University, Ithaca, New York
| | - Christine M Walko
- DeBartolo Family Personalized Medicine Institute, Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Todd C Knepper
- DeBartolo Family Personalized Medicine Institute, Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - John T Soper
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Jared Weiss
- Department of Medicine, Division of Hematology and Oncology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina.,Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Juneko E Grilley-Olson
- Department of Medicine, Division of Hematology and Oncology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina.,Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - William Y Kim
- Department of Medicine, Division of Hematology and Oncology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina.,Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - H Shelton Earp
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Ross L Levine
- Memorial Sloan Kettering Cancer Center, New York, New York
| | | | - Ahmet Zehir
- Memorial Sloan Kettering Cancer Center, New York, New York
| | - D Neil Hayes
- Department of Medicine, Division of Hematology and Oncology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina.,Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina.,Division of Medical Oncology at the University of Tennessee Health Science Center (UTHSC), Memphis, Tennessee
| | - Eric Padron
- Department of Malignant Hematology, Moffitt Cancer Center and Research Institute, Tampa, Florida
| |
Collapse
|
23
|
Siegel MB, He X, Hoadley KA, Hoyle A, Pearce JB, Garrett AL, Kumar S, Moylan VJ, Brady CM, Van Swearingen AE, Marron D, Gupta GP, Thorne LB, Kieran N, Livasy C, Mardis ER, Parker JS, Chen M, Anders CK, Carey LA, Perou CM. Integrated RNA and DNA sequencing reveals early drivers of metastatic breast cancer. J Clin Invest 2018; 128:1371-1383. [PMID: 29480819 PMCID: PMC5873890 DOI: 10.1172/jci96153] [Citation(s) in RCA: 131] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Accepted: 12/21/2017] [Indexed: 12/22/2022] Open
Abstract
Breast cancer metastasis remains a clinical challenge, even within a single patient across multiple sites of the disease. Genome-wide comparisons of both the DNA and gene expression of primary tumors and metastases in multiple patients could help elucidate the underlying mechanisms that cause breast cancer metastasis. To address this issue, we performed DNA exome and RNA sequencing of matched primary tumors and multiple metastases from 16 patients, totaling 83 distinct specimens. We identified tumor-specific drivers by integrating known protein-protein network information with RNA expression and somatic DNA alterations and found that genetic drivers were predominantly established in the primary tumor and maintained through metastatic spreading. In addition, our analyses revealed that most genetic drivers were DNA copy number changes, the TP53 mutation was a recurrent founding mutation regardless of subtype, and that multiclonal seeding of metastases was frequent and occurred in multiple subtypes. Genetic drivers unique to metastasis were identified as somatic mutations in the estrogen and androgen receptor genes. These results highlight the complexity of metastatic spreading, be it monoclonal or multiclonal, and suggest that most metastatic drivers are established in the primary tumor, despite the substantial heterogeneity seen in the metastases.
Collapse
Affiliation(s)
- Marni B. Siegel
- Department of Genetics
- Lineberger Comprehensive Cancer Center
| | | | | | | | - Julia B. Pearce
- Division of Hematology-Oncology, Department of Medicine, School
of Medicine
| | - Amy L. Garrett
- Division of Hematology-Oncology, Department of Medicine, School
of Medicine
| | | | | | | | | | | | - Gaorav P. Gupta
- Lineberger Comprehensive Cancer Center
- Department of Radiation Oncology, School of Medicine, University
of North Carolina (UNC) at Chapel Hill, Chapel Hill, North Carolina, USA
| | | | - Niamh Kieran
- Division of Hematology-Oncology, Department of Medicine, School
of Medicine
| | - Chad Livasy
- Department of Pathology and Laboratory Medicine, and
- Department of Pathology, Levine Cancer Institute, Carolinas
Medical Center, Carolinas HealthCare System, Charlotte, North Carolina, USA
| | - Elaine R. Mardis
- The Research Institute at Nationwide Children’s Hospital, The
Ohio State University College of Medicine, Columbus, Ohio, USA
| | - Joel S. Parker
- Department of Genetics
- Lineberger Comprehensive Cancer Center
| | - Mengjie Chen
- Department of Biostatistics, UNC at Chapel Hill, Chapel Hill,
North Carolina, USA
| | - Carey K. Anders
- Lineberger Comprehensive Cancer Center
- Division of Hematology-Oncology, Department of Medicine, School
of Medicine
| | - Lisa A. Carey
- Lineberger Comprehensive Cancer Center
- Division of Hematology-Oncology, Department of Medicine, School
of Medicine
| | - Charles M. Perou
- Department of Genetics
- Lineberger Comprehensive Cancer Center
- Department of Pathology and Laboratory Medicine, and
| |
Collapse
|
24
|
Bennett CW, Berchem G, Kim YJ, El-Khoury V. Cell-free DNA and next-generation sequencing in the service of personalized medicine for lung cancer. Oncotarget 2018; 7:71013-71035. [PMID: 27589834 PMCID: PMC5342606 DOI: 10.18632/oncotarget.11717] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Accepted: 08/11/2016] [Indexed: 12/13/2022] Open
Abstract
Personalized medicine has emerged as the future of cancer care to ensure that patients receive individualized treatment specific to their needs. In order to provide such care, molecular techniques that enable oncologists to diagnose, treat, and monitor tumors are necessary. In the field of lung cancer, cell free DNA (cfDNA) shows great potential as a less invasive liquid biopsy technique, and next-generation sequencing (NGS) is a promising tool for analysis of tumor mutations. In this review, we outline the evolution of cfDNA and NGS and discuss the progress of using them in a clinical setting for patients with lung cancer. We also present an analysis of the role of cfDNA as a liquid biopsy technique and NGS as an analytical tool in studying EGFR and MET, two frequently mutated genes in lung cancer. Ultimately, we hope that using cfDNA and NGS for cancer diagnosis and treatment will become standard for patients with lung cancer and across the field of oncology.
Collapse
Affiliation(s)
- Catherine W Bennett
- Department of Oncology, Luxembourg Institute of Health, L-1526 Luxembourg, Luxembourg
| | - Guy Berchem
- Department of Oncology, Luxembourg Institute of Health, L-1526 Luxembourg, Luxembourg.,Centre Hospitalier de Luxembourg, L-1210 Luxembourg, Luxembourg
| | - Yeoun Jin Kim
- Department of Oncology, Luxembourg Institute of Health, L-1526 Luxembourg, Luxembourg
| | - Victoria El-Khoury
- Department of Oncology, Luxembourg Institute of Health, L-1526 Luxembourg, Luxembourg
| |
Collapse
|
25
|
Kotelnikova EA, Pyatnitskiy M, Paleeva A, Kremenetskaya O, Vinogradov D. Practical aspects of NGS-based pathways analysis for personalized cancer science and medicine. Oncotarget 2018; 7:52493-52516. [PMID: 27191992 PMCID: PMC5239569 DOI: 10.18632/oncotarget.9370] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Accepted: 04/18/2016] [Indexed: 12/17/2022] Open
Abstract
Nowadays, the personalized approach to health care and cancer care in particular is becoming more and more popular and is taking an important place in the translational medicine paradigm. In some cases, detection of the patient-specific individual mutations that point to a targeted therapy has already become a routine practice for clinical oncologists. Wider panels of genetic markers are also on the market which cover a greater number of possible oncogenes including those with lower reliability of resulting medical conclusions. In light of the large availability of high-throughput technologies, it is very tempting to use complete patient-specific New Generation Sequencing (NGS) or other "omics" data for cancer treatment guidance. However, there are still no gold standard methods and protocols to evaluate them. Here we will discuss the clinical utility of each of the data types and describe a systems biology approach adapted for single patient measurements. We will try to summarize the current state of the field focusing on the clinically relevant case-studies and practical aspects of data processing.
Collapse
Affiliation(s)
- Ekaterina A Kotelnikova
- Personal Biomedicine, Moscow, Russia.,A. A. Kharkevich Institute for Information Transmission Problems, Russian Academy of Sciences, Moscow, Russia.,Institute Biomedical Research August Pi Sunyer (IDIBAPS), Hospital Clinic of Barcelona, Barcelona, Spain
| | - Mikhail Pyatnitskiy
- Personal Biomedicine, Moscow, Russia.,Orekhovich Institute of Biomedical Chemistry, Moscow, Russia.,Pirogov Russian National Research Medical University, Moscow, Russia
| | | | - Olga Kremenetskaya
- Personal Biomedicine, Moscow, Russia.,Center for Theoretical Problems of Physicochemical Pharmacology, Russian Academy of Sciences, Moscow, Russia
| | - Dmitriy Vinogradov
- Personal Biomedicine, Moscow, Russia.,A. A. Kharkevich Institute for Information Transmission Problems, Russian Academy of Sciences, Moscow, Russia.,Lomonosov Moscow State University, Moscow, Russia
| |
Collapse
|
26
|
Patel NM, Michelini VV, Snell JM, Balu S, Hoyle AP, Parker JS, Hayward MC, Eberhard DA, Salazar AH, McNeillie P, Xu J, Huettner CS, Koyama T, Utro F, Rhrissorrakrai K, Norel R, Bilal E, Royyuru A, Parida L, Earp HS, Grilley-Olson JE, Hayes DN, Harvey SJ, Sharpless NE, Kim WY. Enhancing Next-Generation Sequencing-Guided Cancer Care Through Cognitive Computing. Oncologist 2017; 23:179-185. [PMID: 29158372 PMCID: PMC5813753 DOI: 10.1634/theoncologist.2017-0170] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Accepted: 10/06/2017] [Indexed: 11/20/2022] Open
Abstract
Next‐generation sequencing (NGS) has emerged as an affordable and reproducible means to query tumors for somatic genetic anomalies. To help interpret somatic NGS data, many institutions have created a molecular tumor board to analyze the results of NGS and make recommendations. This article evaluates the utility of cognitive computing systems to analyze data for clinical decision‐making. Background. Using next‐generation sequencing (NGS) to guide cancer therapy has created challenges in analyzing and reporting large volumes of genomic data to patients and caregivers. Specifically, providing current, accurate information on newly approved therapies and open clinical trials requires considerable manual curation performed mainly by human “molecular tumor boards” (MTBs). The purpose of this study was to determine the utility of cognitive computing as performed by Watson for Genomics (WfG) compared with a human MTB. Materials and Methods. One thousand eighteen patient cases that previously underwent targeted exon sequencing at the University of North Carolina (UNC) and subsequent analysis by the UNCseq informatics pipeline and the UNC MTB between November 7, 2011, and May 12, 2015, were analyzed with WfG, a cognitive computing technology for genomic analysis. Results. Using a WfG‐curated actionable gene list, we identified additional genomic events of potential significance (not discovered by traditional MTB curation) in 323 (32%) patients. The majority of these additional genomic events were considered actionable based upon their ability to qualify patients for biomarker‐selected clinical trials. Indeed, the opening of a relevant clinical trial within 1 month prior to WfG analysis provided the rationale for identification of a new actionable event in nearly a quarter of the 323 patients. This automated analysis took <3 minutes per case. Conclusion. These results demonstrate that the interpretation and actionability of somatic NGS results are evolving too rapidly to rely solely on human curation. Molecular tumor boards empowered by cognitive computing could potentially improve patient care by providing a rapid, comprehensive approach for data analysis and consideration of up‐to‐date availability of clinical trials. Implications for Practice. The results of this study demonstrate that the interpretation and actionability of somatic next‐generation sequencing results are evolving too rapidly to rely solely on human curation. Molecular tumor boards empowered by cognitive computing can significantly improve patient care by providing a fast, cost‐effective, and comprehensive approach for data analysis in the delivery of precision medicine. Patients and physicians who are considering enrollment in clinical trials may benefit from the support of such tools applied to genomic data.
Collapse
Affiliation(s)
- Nirali M Patel
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | | | - Jeff M Snell
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Saianand Balu
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Alan P Hoyle
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Joel S Parker
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Michele C Hayward
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - David A Eberhard
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Ashley H Salazar
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | | | - Jia Xu
- IBM Watson Health, Cambridge, Massachusetts, USA
| | | | | | | | | | | | - Erhan Bilal
- IBM Research, Yorktown Heights, New York, USA
| | | | | | - H Shelton Earp
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Juneko E Grilley-Olson
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - D Neil Hayes
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | | | - Norman E Sharpless
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - William Y Kim
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Department of Urology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| |
Collapse
|
27
|
Calling Chromosome Alterations, DNA Methylation Statuses, and Mutations in Tumors by Simple Targeted Next-Generation Sequencing. J Mol Diagn 2017; 19:776-787. [DOI: 10.1016/j.jmoldx.2017.06.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Accepted: 06/01/2017] [Indexed: 12/16/2022] Open
|
28
|
SLC39A4 expression is associated with enhanced cell migration, cisplatin resistance, and poor survival in non-small cell lung cancer. Sci Rep 2017; 7:7211. [PMID: 28775359 PMCID: PMC5543149 DOI: 10.1038/s41598-017-07830-4] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Accepted: 07/04/2017] [Indexed: 12/13/2022] Open
Abstract
The zinc transporter SLC39A4 influences epithelial cell morphology and migration in various cancers; however, its role in regulating cell invasion and chemotherapeutic resistance in human lung cancer is not yet clear. Here, integrated analysis of gene expression in non-small cell lung cancer revealed that SLC39A4 expression is significantly correlated with increased tumour size and regional lymph node spread, as well as shorter overall survival (OS) and disease-free survival (DFS). SLC39A4 silencing by lentivirus-mediated shRNA blocked human lung cancer cell epithelial-mesenchymal transition and metastasis in vitro and in vivo, respectively. Moreover, SLC39A4 knockdown enhanced cancer cell sensitivity to cisplatin-induced death by inhibiting stemness in lung cancer cells. Collectively, these data suggest that SLC39A4 may be a novel therapeutic target and predictive marker of tumour metastasis in non-small cell lung cancer.
Collapse
|
29
|
Kardos J, Wobker SE, Woods ME, Nielsen ME, Smith AB, Wallen EM, Pruthi RS, Hayward MC, McGinty KA, Grilley-Olson JE, Patel NM, Weck KE, Black P, Parker JS, Milowsky MI, Hayes DN, Kim WY. Comprehensive Molecular Characterization of Urachal Adenocarcinoma Reveals Commonalities With Colorectal Cancer, Including a Hypermutable Phenotype. JCO Precis Oncol 2017; 1:1700027. [PMID: 32913973 DOI: 10.1200/po.17.00027] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Purpose Urachal adenocarcinoma is a rare type of primary bladder adenocarcinoma that comprises less than 1% of all bladder cancers. The low incidence of urachal adenocarcinomas does not allow for an evidence-based approach to therapy. Transcriptome profiling of urachal adenocarcinomas has not been previously reported. We hypothesized that an in-depth molecular understanding of urachal adenocarcinoma would uncover rational therapeutic strategies. Patients and Methods We performed targeted exon sequencing and global transcriptome profiling of 12 urachal tumors to generate a comprehensive molecular portrait of urachal adenocarcinoma. A single patient with an MSH6 mutation was treated with the anti-programmed death-ligand 1 antibody, atezolizumab. Results Urachal adenocarcinoma closely resembles colorectal cancer at the level of RNA expression, which extends previous observations that urachal tumors harbor genomic alterations that are found in colorectal adenocarcinoma. A subset of tumors was found to have alterations in genes that are associated with microsatellite instability (MSH2 and MSH6) and hypermutation (POLE). A patient with an MSH6 mutation was treated with immune checkpoint blockade, which resulted in stable disease. Conclusion Because clinical trials are next to impossible for patients with rare tumors, precision oncology may be an important adjunct for treatment decisions. Our findings demonstrate that urachal adenocarcinomas molecularly resemble colorectal adenocarcinomas at the level of RNA expression, are the first report, to our knowledge, of MSH2 and MSH6 mutations in this disease, and support the consideration of immune checkpoint blockade as a rational therapeutic treatment of this exceedingly rare tumor.
Collapse
Affiliation(s)
- Jordan Kardos
- , , , , , , , , , , , , , , , and , University of North Carolina at Chapel Hill, Chapel Hill, NC; and , University of British Columbia, Vancouver, British Columbia, Canada
| | - Sara E Wobker
- , , , , , , , , , , , , , , , and , University of North Carolina at Chapel Hill, Chapel Hill, NC; and , University of British Columbia, Vancouver, British Columbia, Canada
| | - Michael E Woods
- , , , , , , , , , , , , , , , and , University of North Carolina at Chapel Hill, Chapel Hill, NC; and , University of British Columbia, Vancouver, British Columbia, Canada
| | - Matthew E Nielsen
- , , , , , , , , , , , , , , , and , University of North Carolina at Chapel Hill, Chapel Hill, NC; and , University of British Columbia, Vancouver, British Columbia, Canada
| | - Angela B Smith
- , , , , , , , , , , , , , , , and , University of North Carolina at Chapel Hill, Chapel Hill, NC; and , University of British Columbia, Vancouver, British Columbia, Canada
| | - Eric M Wallen
- , , , , , , , , , , , , , , , and , University of North Carolina at Chapel Hill, Chapel Hill, NC; and , University of British Columbia, Vancouver, British Columbia, Canada
| | - Raj S Pruthi
- , , , , , , , , , , , , , , , and , University of North Carolina at Chapel Hill, Chapel Hill, NC; and , University of British Columbia, Vancouver, British Columbia, Canada
| | - Michele C Hayward
- , , , , , , , , , , , , , , , and , University of North Carolina at Chapel Hill, Chapel Hill, NC; and , University of British Columbia, Vancouver, British Columbia, Canada
| | - Katrina A McGinty
- , , , , , , , , , , , , , , , and , University of North Carolina at Chapel Hill, Chapel Hill, NC; and , University of British Columbia, Vancouver, British Columbia, Canada
| | - Juneko E Grilley-Olson
- , , , , , , , , , , , , , , , and , University of North Carolina at Chapel Hill, Chapel Hill, NC; and , University of British Columbia, Vancouver, British Columbia, Canada
| | - Nirali M Patel
- , , , , , , , , , , , , , , , and , University of North Carolina at Chapel Hill, Chapel Hill, NC; and , University of British Columbia, Vancouver, British Columbia, Canada
| | - Karen E Weck
- , , , , , , , , , , , , , , , and , University of North Carolina at Chapel Hill, Chapel Hill, NC; and , University of British Columbia, Vancouver, British Columbia, Canada
| | - Peter Black
- , , , , , , , , , , , , , , , and , University of North Carolina at Chapel Hill, Chapel Hill, NC; and , University of British Columbia, Vancouver, British Columbia, Canada
| | - Joel S Parker
- , , , , , , , , , , , , , , , and , University of North Carolina at Chapel Hill, Chapel Hill, NC; and , University of British Columbia, Vancouver, British Columbia, Canada
| | - Matthew I Milowsky
- , , , , , , , , , , , , , , , and , University of North Carolina at Chapel Hill, Chapel Hill, NC; and , University of British Columbia, Vancouver, British Columbia, Canada
| | - D Neil Hayes
- , , , , , , , , , , , , , , , and , University of North Carolina at Chapel Hill, Chapel Hill, NC; and , University of British Columbia, Vancouver, British Columbia, Canada
| | - William Y Kim
- , , , , , , , , , , , , , , , and , University of North Carolina at Chapel Hill, Chapel Hill, NC; and , University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
30
|
Sharma MR, Auman JT, Patel NM, Grilley-Olson JE, Zhao X, Moschos SJ, Parker JS, Yin X, Hayward MC, Polite BN, Marangon E, Posocco B, Toffoli G, Hayes DN, Innocenti F. Exceptional Chemotherapy Response in Metastatic Colorectal Cancer Associated With Hyper-Indel-Hypermutated Cancer Genome and Comutation of POLD1 and MLH1. JCO Precis Oncol 2017; 2017:PO.16.00015. [PMID: 30009279 PMCID: PMC6042871 DOI: 10.1200/po.16.00015] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
PURPOSE A73-year-old woman with metastatic colon cancer experienced a complete response to chemotherapy with dose-intensified irinotecan that has been durable for 5 years. We sequenced her tumor and germ line DNA and looked for similar patterns in publicly available genomic data from patients with colorectal cancer. PATIENTS AND METHODS Tumor DNA was obtained from a biopsy before therapy, and germ line DNA was obtained from blood. Tumor and germline DNA were sequenced using a commercial panel with approximately 250 genes. Whole-genome amplification and exome sequencing were performed for POLE and POLD1. A POLD1 mutation was confirmed by Sanger sequencing. The somatic mutation and clinical annotation data files from the colon (n = 461) and rectal (n = 171) adenocarcinoma data sets were downloaded from The Cancer Genome Atlas data portal and analyzed for patterns of mutations and clinical outcomes in patients with POLE- and/or POLD1-mutated tumors. RESULTS The pattern of alterations included APC biallelic inactivation and microsatellite instability high (MSI-H) phenotype, with somatic inactivation of MLH1 and hypermutation (estimated mutation rate > 200 per megabase). The extremely high mutation rate led us to investigate additional mechanisms for hypermutation, including loss of function of POLE. POLE was unaltered, but a related gene not typically associated with somatic mutation in colon cancer, POLD1, had a somatic mutation c.2171G>A[p.Gly724Glu]. Additionally, we noted that the high mutation rate was largely composed of dinucleotide deletions. A similar pattern of hypermutation (dinucleotide deletions, POLD1 mutations, MSI-H) was found in tumors from The Cancer Genome Atlas. CONCLUSION POLD1 mutation with associated MSI-H and hyper-indel-hypermutated cancer genome characterizes a previously unrecognized variant of colon cancer that was found in this patient with an exceptional response to chemotherapy.
Collapse
Affiliation(s)
- Manish R. Sharma
- Manish R. Sharma and Blase N. Polite, University of Chicago Medicine, Chicago, IL; James T. Auman, Nirali M. Patel, Juneko E. Grilley-Olson, Xiaobei Zhao, Stergios J. Moschos, Joel S. Parker, Xiaoying Yin, Michele C. Hayward, D. Neil Hayes, and Federico Innocenti, University of North Carolina at Chapel Hill, Chapel Hill, NC; and Elena Marangon, Bianca Posocco, and Guiseppe Toffoli, Centro di Riferimento Oncologico, Aviano, Italy
| | - James T. Auman
- Manish R. Sharma and Blase N. Polite, University of Chicago Medicine, Chicago, IL; James T. Auman, Nirali M. Patel, Juneko E. Grilley-Olson, Xiaobei Zhao, Stergios J. Moschos, Joel S. Parker, Xiaoying Yin, Michele C. Hayward, D. Neil Hayes, and Federico Innocenti, University of North Carolina at Chapel Hill, Chapel Hill, NC; and Elena Marangon, Bianca Posocco, and Guiseppe Toffoli, Centro di Riferimento Oncologico, Aviano, Italy
| | - Nirali M. Patel
- Manish R. Sharma and Blase N. Polite, University of Chicago Medicine, Chicago, IL; James T. Auman, Nirali M. Patel, Juneko E. Grilley-Olson, Xiaobei Zhao, Stergios J. Moschos, Joel S. Parker, Xiaoying Yin, Michele C. Hayward, D. Neil Hayes, and Federico Innocenti, University of North Carolina at Chapel Hill, Chapel Hill, NC; and Elena Marangon, Bianca Posocco, and Guiseppe Toffoli, Centro di Riferimento Oncologico, Aviano, Italy
| | - Juneko E. Grilley-Olson
- Manish R. Sharma and Blase N. Polite, University of Chicago Medicine, Chicago, IL; James T. Auman, Nirali M. Patel, Juneko E. Grilley-Olson, Xiaobei Zhao, Stergios J. Moschos, Joel S. Parker, Xiaoying Yin, Michele C. Hayward, D. Neil Hayes, and Federico Innocenti, University of North Carolina at Chapel Hill, Chapel Hill, NC; and Elena Marangon, Bianca Posocco, and Guiseppe Toffoli, Centro di Riferimento Oncologico, Aviano, Italy
| | - Xiaobei Zhao
- Manish R. Sharma and Blase N. Polite, University of Chicago Medicine, Chicago, IL; James T. Auman, Nirali M. Patel, Juneko E. Grilley-Olson, Xiaobei Zhao, Stergios J. Moschos, Joel S. Parker, Xiaoying Yin, Michele C. Hayward, D. Neil Hayes, and Federico Innocenti, University of North Carolina at Chapel Hill, Chapel Hill, NC; and Elena Marangon, Bianca Posocco, and Guiseppe Toffoli, Centro di Riferimento Oncologico, Aviano, Italy
| | - Stergios J. Moschos
- Manish R. Sharma and Blase N. Polite, University of Chicago Medicine, Chicago, IL; James T. Auman, Nirali M. Patel, Juneko E. Grilley-Olson, Xiaobei Zhao, Stergios J. Moschos, Joel S. Parker, Xiaoying Yin, Michele C. Hayward, D. Neil Hayes, and Federico Innocenti, University of North Carolina at Chapel Hill, Chapel Hill, NC; and Elena Marangon, Bianca Posocco, and Guiseppe Toffoli, Centro di Riferimento Oncologico, Aviano, Italy
| | - Joel S. Parker
- Manish R. Sharma and Blase N. Polite, University of Chicago Medicine, Chicago, IL; James T. Auman, Nirali M. Patel, Juneko E. Grilley-Olson, Xiaobei Zhao, Stergios J. Moschos, Joel S. Parker, Xiaoying Yin, Michele C. Hayward, D. Neil Hayes, and Federico Innocenti, University of North Carolina at Chapel Hill, Chapel Hill, NC; and Elena Marangon, Bianca Posocco, and Guiseppe Toffoli, Centro di Riferimento Oncologico, Aviano, Italy
| | - Xiaoying Yin
- Manish R. Sharma and Blase N. Polite, University of Chicago Medicine, Chicago, IL; James T. Auman, Nirali M. Patel, Juneko E. Grilley-Olson, Xiaobei Zhao, Stergios J. Moschos, Joel S. Parker, Xiaoying Yin, Michele C. Hayward, D. Neil Hayes, and Federico Innocenti, University of North Carolina at Chapel Hill, Chapel Hill, NC; and Elena Marangon, Bianca Posocco, and Guiseppe Toffoli, Centro di Riferimento Oncologico, Aviano, Italy
| | - Michele C. Hayward
- Manish R. Sharma and Blase N. Polite, University of Chicago Medicine, Chicago, IL; James T. Auman, Nirali M. Patel, Juneko E. Grilley-Olson, Xiaobei Zhao, Stergios J. Moschos, Joel S. Parker, Xiaoying Yin, Michele C. Hayward, D. Neil Hayes, and Federico Innocenti, University of North Carolina at Chapel Hill, Chapel Hill, NC; and Elena Marangon, Bianca Posocco, and Guiseppe Toffoli, Centro di Riferimento Oncologico, Aviano, Italy
| | - Blase N. Polite
- Manish R. Sharma and Blase N. Polite, University of Chicago Medicine, Chicago, IL; James T. Auman, Nirali M. Patel, Juneko E. Grilley-Olson, Xiaobei Zhao, Stergios J. Moschos, Joel S. Parker, Xiaoying Yin, Michele C. Hayward, D. Neil Hayes, and Federico Innocenti, University of North Carolina at Chapel Hill, Chapel Hill, NC; and Elena Marangon, Bianca Posocco, and Guiseppe Toffoli, Centro di Riferimento Oncologico, Aviano, Italy
| | - Elena Marangon
- Manish R. Sharma and Blase N. Polite, University of Chicago Medicine, Chicago, IL; James T. Auman, Nirali M. Patel, Juneko E. Grilley-Olson, Xiaobei Zhao, Stergios J. Moschos, Joel S. Parker, Xiaoying Yin, Michele C. Hayward, D. Neil Hayes, and Federico Innocenti, University of North Carolina at Chapel Hill, Chapel Hill, NC; and Elena Marangon, Bianca Posocco, and Guiseppe Toffoli, Centro di Riferimento Oncologico, Aviano, Italy
| | - Bianca Posocco
- Manish R. Sharma and Blase N. Polite, University of Chicago Medicine, Chicago, IL; James T. Auman, Nirali M. Patel, Juneko E. Grilley-Olson, Xiaobei Zhao, Stergios J. Moschos, Joel S. Parker, Xiaoying Yin, Michele C. Hayward, D. Neil Hayes, and Federico Innocenti, University of North Carolina at Chapel Hill, Chapel Hill, NC; and Elena Marangon, Bianca Posocco, and Guiseppe Toffoli, Centro di Riferimento Oncologico, Aviano, Italy
| | - Giuseppe Toffoli
- Manish R. Sharma and Blase N. Polite, University of Chicago Medicine, Chicago, IL; James T. Auman, Nirali M. Patel, Juneko E. Grilley-Olson, Xiaobei Zhao, Stergios J. Moschos, Joel S. Parker, Xiaoying Yin, Michele C. Hayward, D. Neil Hayes, and Federico Innocenti, University of North Carolina at Chapel Hill, Chapel Hill, NC; and Elena Marangon, Bianca Posocco, and Guiseppe Toffoli, Centro di Riferimento Oncologico, Aviano, Italy
| | - D. Neil Hayes
- Manish R. Sharma and Blase N. Polite, University of Chicago Medicine, Chicago, IL; James T. Auman, Nirali M. Patel, Juneko E. Grilley-Olson, Xiaobei Zhao, Stergios J. Moschos, Joel S. Parker, Xiaoying Yin, Michele C. Hayward, D. Neil Hayes, and Federico Innocenti, University of North Carolina at Chapel Hill, Chapel Hill, NC; and Elena Marangon, Bianca Posocco, and Guiseppe Toffoli, Centro di Riferimento Oncologico, Aviano, Italy
| | - Federico Innocenti
- Manish R. Sharma and Blase N. Polite, University of Chicago Medicine, Chicago, IL; James T. Auman, Nirali M. Patel, Juneko E. Grilley-Olson, Xiaobei Zhao, Stergios J. Moschos, Joel S. Parker, Xiaoying Yin, Michele C. Hayward, D. Neil Hayes, and Federico Innocenti, University of North Carolina at Chapel Hill, Chapel Hill, NC; and Elena Marangon, Bianca Posocco, and Guiseppe Toffoli, Centro di Riferimento Oncologico, Aviano, Italy
| |
Collapse
|
31
|
Silva GO, Siegel MB, Mose LE, Parker JS, Sun W, Perou CM, Chen M. SynthEx: a synthetic-normal-based DNA sequencing tool for copy number alteration detection and tumor heterogeneity profiling. Genome Biol 2017; 18:66. [PMID: 28390427 PMCID: PMC5385048 DOI: 10.1186/s13059-017-1193-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Accepted: 03/16/2017] [Indexed: 01/22/2023] Open
Abstract
Changes in the quantity of genetic material, known as somatic copy number alterations (CNAs), can drive tumorigenesis. Many methods exist for assessing CNAs using microarrays, but considerable technical issues limit current CNA calling based upon DNA sequencing. We present SynthEx, a novel tool for detecting CNAs from whole exome and genome sequencing. SynthEx utilizes a “synthetic-normal” strategy to overcome technical and financial issues. In terms of accuracy and precision, SynthEx is highly comparable to array-based methods and outperforms sequencing-based CNA detection tools. SynthEx robustly identifies CNAs using sequencing data without the additional costs associated with matched normal specimens.
Collapse
Affiliation(s)
- Grace O Silva
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.,Curriculum in Bioinformatics and Computational Biology, University of North Carolina, Chapel Hill, NC, 27599, USA.,Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Marni B Siegel
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.,Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Lisle E Mose
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Joel S Parker
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.,Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Wei Sun
- Public Health Division, Fred Hutchison Cancer Research Center, Seattle, WA, 98109, USA
| | - Charles M Perou
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.,Curriculum in Bioinformatics and Computational Biology, University of North Carolina, Chapel Hill, NC, 27599, USA.,Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Mengjie Chen
- Section of Genetic Medicine, Department of Medicine, The University of Chicago, 900 East 57th Street, KCBD 3220A, Chicago, IL, 60637, USA.
| |
Collapse
|
32
|
EGFR Testing in Advanced Non–Small-Cell Lung Cancer, A Mini-Review. Clin Lung Cancer 2016; 17:483-492. [DOI: 10.1016/j.cllc.2016.05.016] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Revised: 05/27/2016] [Accepted: 05/31/2016] [Indexed: 11/20/2022]
|
33
|
van Amerongen RA, Retèl VP, Coupé VM, Nederlof PM, Vogel MJ, van Harten WH. Next-generation sequencing in NSCLC and melanoma patients: a cost and budget impact analysis. Ecancermedicalscience 2016; 10:684. [PMID: 27899957 PMCID: PMC5102690 DOI: 10.3332/ecancer.2016.684] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Indexed: 02/06/2023] Open
Abstract
Next-generation sequencing (NGS) has reached the molecular diagnostic laboratories. Although the NGS technology aims to improve the effectiveness of therapies by selecting the most promising therapy, concerns are that NGS testing is expensive and that the ‘benefits’ are not yet in relation to these costs. In this study, we give an estimation of the costs and an institutional and national budget impact of various types of NGS tests in non-small-cell lung cancer (NSCLC) and melanoma patients within The Netherlands. First, an activity-based costing (ABC) analysis has been conducted on the costs of two examples of NGS panels (small- and medium-targeted gene panel (TGP)) based on data of The Netherlands Cancer Institute (NKI). Second, we performed a budget impact analysis (BIA) to estimate the current (2015) and future (2020) budget impact of NGS on molecular diagnostics for NSCLC and melanoma patients in The Netherlands. Literature, expert opinions, and a data set of patients within the NKI (n = 172) have been included in the BIA. Based on our analysis, we expect that the NGS test cost concerns will be limited. In the current situation, NGS can indeed result in higher diagnostic test costs, which is mainly related to required additional tests besides the small TGP. However, in the future, we expect that the use of whole-genome sequencing (WGS) will increase, for which it is expected that additional tests can be (partly) avoided. Although the current clinical benefits are expected to be limited, the research potentials of NGS are already an important advantage.
Collapse
Affiliation(s)
- Rosa A van Amerongen
- Department of Epidemiology and Biostatistics, VU University Medical Center, PO Box 7057, 1007 MB Amsterdam, The Netherlands
| | - Valesca P Retèl
- Department of Psychosocial Research and Epidemiology, The Netherlands Cancer Institute, PO Box 90203, 1006 BE Amsterdam, The Netherlands; School of Governance and Management, University of Twente, MB-HTSR, PO Box 217, 7500 AE Enschede, The Netherlands
| | - Veerle Mh Coupé
- Department of Epidemiology and Biostatistics, VU University Medical Center, PO Box 7057, 1007 MB Amsterdam, The Netherlands
| | - Petra M Nederlof
- Department of Molecular Diagnostics, Pathology, The Netherlands Cancer Institute, PO Box 90203, 1006 BE Amsterdam, The Netherlands
| | - Maartje J Vogel
- Department of Molecular Diagnostics, Pathology, The Netherlands Cancer Institute, PO Box 90203, 1006 BE Amsterdam, The Netherlands
| | - Wim H van Harten
- Department of Psychosocial Research and Epidemiology, The Netherlands Cancer Institute, PO Box 90203, 1006 BE Amsterdam, The Netherlands; School of Governance and Management, University of Twente, MB-HTSR, PO Box 217, 7500 AE Enschede, The Netherlands
| |
Collapse
|
34
|
Facchinetti F, Tiseo M, Di Maio M, Graziano P, Bria E, Rossi G, Novello S. Tackling ALK in non-small cell lung cancer: the role of novel inhibitors. Transl Lung Cancer Res 2016; 5:301-21. [PMID: 27413712 DOI: 10.21037/tlcr.2016.06.10] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Crizotinib is an oral inhibitor of anaplastic lymphoma kinase (ALK) with remarkable clinical activity in patients suffering from ALK-rearranged non-small cell lung cancer (NSCLC), accounting to its superiority compared to chemotherapy. Unfortunately, virtually all ALK-rearranged tumors acquire resistance to crizotinib, frequently within one year since the treatment initiation. To date, therapeutic strategies to overcome crizotinib resistance have focused on the use of more potent and structurally different compounds. Second-generation ALK inhibitors such as ceritinib (LDK378), alectinib (CH5424802/RO5424802) and brigatinib (AP26113) have shown relevant clinical activity, consequently fostering their rapid clinical development and their approval by health agencies. The third-generation inhibitor lorlatinib (PF-06463922), selectively active against ALK and ROS1, harbors impressive biological potency; its efficacy in reversing resistance to crizotinib and to other ALK inhibitors is being proven by early clinical trials. The NTRK1-3 and ROS1 inhibitor entrectinib (RXDX-101) has been reported to act against NSCLC harboring ALK fusion proteins too. Despite the quick development of these novel agents, several issues remain to be discussed in the treatment of patients suffering from ALK-rearranged NSCLC. This position paper will discuss the development, the current evidence and approvals, as long as the future perspectives of new ALK inhibitors beyond crizotinib. Clinical behaviors of ALK-rearranged NSCLC vary significantly among patients and differential molecular events responsible of crizotinib resistance account for the most important quote of this heterogeneity. The precious availability of a wide range of active anti-ALK compounds should be approached in a critical and careful perspective, in order to develop treatment strategies tailored on the disease evolution of every single patient.
Collapse
Affiliation(s)
- Francesco Facchinetti
- 1 INSERM U981, Gustave Roussy Cancer Campus, Université Paris-Sud, Villejuif, France ; 2 Medical Oncology Unit, University Hospital of Parma, Parma, Italy ; 3 Department of Oncology, AOU San Luigi (Orbassano), University of Turin, Italy ; 4 Medical Oncology, AO Ordine Mauriziano, Turin, Italy ; 5 Division of Pathology, IRCCS "Casa Sollievo della Sofferenza", San Giovanni Rotondo, Foggia, Italy ; 6 Medical Oncology, Azienda Ospedaliera Universitaria Integrata, University of Verona, Verona, Italy ; 7 Operative Unit of Pathology, Azienda USL Valle d'Aosta, Aosta, Italy
| | - Marcello Tiseo
- 1 INSERM U981, Gustave Roussy Cancer Campus, Université Paris-Sud, Villejuif, France ; 2 Medical Oncology Unit, University Hospital of Parma, Parma, Italy ; 3 Department of Oncology, AOU San Luigi (Orbassano), University of Turin, Italy ; 4 Medical Oncology, AO Ordine Mauriziano, Turin, Italy ; 5 Division of Pathology, IRCCS "Casa Sollievo della Sofferenza", San Giovanni Rotondo, Foggia, Italy ; 6 Medical Oncology, Azienda Ospedaliera Universitaria Integrata, University of Verona, Verona, Italy ; 7 Operative Unit of Pathology, Azienda USL Valle d'Aosta, Aosta, Italy
| | - Massimo Di Maio
- 1 INSERM U981, Gustave Roussy Cancer Campus, Université Paris-Sud, Villejuif, France ; 2 Medical Oncology Unit, University Hospital of Parma, Parma, Italy ; 3 Department of Oncology, AOU San Luigi (Orbassano), University of Turin, Italy ; 4 Medical Oncology, AO Ordine Mauriziano, Turin, Italy ; 5 Division of Pathology, IRCCS "Casa Sollievo della Sofferenza", San Giovanni Rotondo, Foggia, Italy ; 6 Medical Oncology, Azienda Ospedaliera Universitaria Integrata, University of Verona, Verona, Italy ; 7 Operative Unit of Pathology, Azienda USL Valle d'Aosta, Aosta, Italy
| | - Paolo Graziano
- 1 INSERM U981, Gustave Roussy Cancer Campus, Université Paris-Sud, Villejuif, France ; 2 Medical Oncology Unit, University Hospital of Parma, Parma, Italy ; 3 Department of Oncology, AOU San Luigi (Orbassano), University of Turin, Italy ; 4 Medical Oncology, AO Ordine Mauriziano, Turin, Italy ; 5 Division of Pathology, IRCCS "Casa Sollievo della Sofferenza", San Giovanni Rotondo, Foggia, Italy ; 6 Medical Oncology, Azienda Ospedaliera Universitaria Integrata, University of Verona, Verona, Italy ; 7 Operative Unit of Pathology, Azienda USL Valle d'Aosta, Aosta, Italy
| | - Emilio Bria
- 1 INSERM U981, Gustave Roussy Cancer Campus, Université Paris-Sud, Villejuif, France ; 2 Medical Oncology Unit, University Hospital of Parma, Parma, Italy ; 3 Department of Oncology, AOU San Luigi (Orbassano), University of Turin, Italy ; 4 Medical Oncology, AO Ordine Mauriziano, Turin, Italy ; 5 Division of Pathology, IRCCS "Casa Sollievo della Sofferenza", San Giovanni Rotondo, Foggia, Italy ; 6 Medical Oncology, Azienda Ospedaliera Universitaria Integrata, University of Verona, Verona, Italy ; 7 Operative Unit of Pathology, Azienda USL Valle d'Aosta, Aosta, Italy
| | - Giulio Rossi
- 1 INSERM U981, Gustave Roussy Cancer Campus, Université Paris-Sud, Villejuif, France ; 2 Medical Oncology Unit, University Hospital of Parma, Parma, Italy ; 3 Department of Oncology, AOU San Luigi (Orbassano), University of Turin, Italy ; 4 Medical Oncology, AO Ordine Mauriziano, Turin, Italy ; 5 Division of Pathology, IRCCS "Casa Sollievo della Sofferenza", San Giovanni Rotondo, Foggia, Italy ; 6 Medical Oncology, Azienda Ospedaliera Universitaria Integrata, University of Verona, Verona, Italy ; 7 Operative Unit of Pathology, Azienda USL Valle d'Aosta, Aosta, Italy
| | - Silvia Novello
- 1 INSERM U981, Gustave Roussy Cancer Campus, Université Paris-Sud, Villejuif, France ; 2 Medical Oncology Unit, University Hospital of Parma, Parma, Italy ; 3 Department of Oncology, AOU San Luigi (Orbassano), University of Turin, Italy ; 4 Medical Oncology, AO Ordine Mauriziano, Turin, Italy ; 5 Division of Pathology, IRCCS "Casa Sollievo della Sofferenza", San Giovanni Rotondo, Foggia, Italy ; 6 Medical Oncology, Azienda Ospedaliera Universitaria Integrata, University of Verona, Verona, Italy ; 7 Operative Unit of Pathology, Azienda USL Valle d'Aosta, Aosta, Italy
| |
Collapse
|
35
|
Seifert BA, O'Daniel JM, Amin K, Marchuk DS, Patel NM, Parker JS, Hoyle AP, Mose LE, Marron A, Hayward MC, Bizon C, Wilhelmsen KC, Evans JP, Earp HS, Sharpless NE, Hayes DN, Berg JS. Germline Analysis from Tumor-Germline Sequencing Dyads to Identify Clinically Actionable Secondary Findings. Clin Cancer Res 2016; 22:4087-4094. [PMID: 27083775 DOI: 10.1158/1078-0432.ccr-16-0015] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Accepted: 03/27/2016] [Indexed: 01/03/2023]
Abstract
PURPOSE To evaluate germline variants in hereditary cancer susceptibility genes among unselected cancer patients undergoing tumor-germline sequencing. EXPERIMENTAL DESIGN Germline sequence data from 439 individuals undergoing tumor-germline dyad sequencing through the LCCC1108/UNCseq™ (NCT01457196) study were analyzed for genetic variants in 36 hereditary cancer susceptibility genes. These variants were analyzed as an exploratory research study to determine whether pathogenic variants exist within the germline of patients undergoing tumor-germline sequencing. Patients were unselected with respect to indicators of hereditary cancer predisposition. RESULTS Variants indicative of hereditary cancer predisposition were identified in 19 (4.3%) patients. For about half (10/19), these findings represent new diagnostic information with potentially important implications for the patient and their family. The others were previously identified through clinical genetic evaluation secondary to suspicion of a hereditary cancer predisposition. Genes with pathogenic variants included ATM, BRCA1, BRCA2, CDKN2A, and CHEK2 In contrast, a substantial proportion of patients (178, 40.5%) had Variants of Uncertain Significance (VUS), 24 of which had VUS in genes pertinent to the presenting cancer. Another 143 had VUS in other hereditary cancer genes, and 11 had VUS in both pertinent and nonpertinent genes. CONCLUSIONS Germline analysis in tumor-germline sequencing dyads will occasionally reveal significant germline findings that were clinically occult, which could be beneficial for patients and their families. However, given the low yield for unexpected germline variation and the large proportion of patients with VUS results, analysis and return of germline results should adhere to guidelines for secondary findings rather than diagnostic hereditary cancer testing. Clin Cancer Res; 22(16); 4087-94. ©2016 AACRSee related commentary by Mandelker, p. 3987.
Collapse
Affiliation(s)
- Bryce A Seifert
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, U.S.A.,Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, U.S.A
| | - Julianne M O'Daniel
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, U.S.A.,Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, U.S.A
| | - Krunal Amin
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, U.S.A
| | - Daniel S Marchuk
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, U.S.A
| | - Nirali M Patel
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, U.S.A.,Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, U.S.A
| | - Joel S Parker
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, U.S.A.,Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, U.S.A
| | - Alan P Hoyle
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, U.S.A
| | - Lisle E Mose
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, U.S.A
| | - Andrew Marron
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, U.S.A
| | - Michele C Hayward
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, U.S.A
| | - Christopher Bizon
- Renaissance Computing Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, U.S.A
| | - Kirk C Wilhelmsen
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, U.S.A.,Renaissance Computing Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, U.S.A
| | - James P Evans
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, U.S.A.,Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, U.S.A
| | - H Shelton Earp
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, U.S.A
| | - Norman E Sharpless
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, U.S.A.,Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, U.S.A
| | - D Neil Hayes
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, U.S.A
| | - Jonathan S Berg
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, U.S.A.,Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, U.S.A
| |
Collapse
|
36
|
Shen W, Szankasi P, Sederberg M, Schumacher J, Frizzell KA, Gee EP, Patel JL, South ST, Xu X, Kelley TW. Concurrent detection of targeted copy number variants and mutations using a myeloid malignancy next generation sequencing panel allows comprehensive genetic analysis using a single testing strategy. Br J Haematol 2016; 173:49-58. [PMID: 26728869 DOI: 10.1111/bjh.13921] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Accepted: 11/19/2015] [Indexed: 02/06/2023]
Abstract
Currently, comprehensive genetic testing of myeloid malignancies requires multiple testing strategies with high costs. Somatic mutations can be detected by next generation sequencing (NGS) but copy number variants (CNVs) require cytogenetic methods including karyotyping, fluorescence in situ hybidization and microarray. Here, we evaluated a new method for CNV detection using read depth data derived from a targeted NGS mutation panel. In a cohort of 270 samples, we detected pathogenic mutations in 208 samples and targeted CNVs in 68 cases. The most frequent CNVs were 7q deletion including LUC7L2 and EZH2, TP53 deletion, ETV6 deletion, gain of RAD21 on 8q, and 5q deletion, including NSD1 and NPM1. We were also able to detect exon-level duplications, including so-called KMT2A (MLL) partial tandem duplication, in 9 cases. In the 63 cases that were negative for mutations, targeted CNVs were observed in 4 cases. Targeted CNV detection by NGS had very high concordance with single nucleotide polymorphism microarray, the current gold standard. We found that ETV6 deletion was strongly associated with TP53 alterations and 7q deletion was associated with mutations in TP53, KRAS and IDH1. This proof-of-concept study demonstrates the feasibility of using the same NGS data to simultaneously detect both somatic mutations and targeted CNVs.
Collapse
Affiliation(s)
- Wei Shen
- ARUP Laboratories, Salt Lake City, UT, USA
| | | | | | | | | | | | - Jay L Patel
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Sarah T South
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Xinjie Xu
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Todd W Kelley
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT, USA
| |
Collapse
|