1
|
Kuang J, Li J, Zhou S, Li Y, Lin J, Huang W, Yuan X. Genomic and micro-environmental insights into drug resistance in colorectal cancer liver metastases. Discov Oncol 2025; 16:241. [PMID: 40009285 DOI: 10.1007/s12672-025-01976-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Accepted: 02/11/2025] [Indexed: 02/27/2025] Open
Abstract
BACKGROUND Colorectal cancer (CRC) is known for its high heterogeneity, with liver metastases significantly impairing survival outcomes. Understanding the tumor microenvironment (TME) and genomic alterations in metastatic sites is crucial for developing personalized therapies that overcome drug resistance and improve prognosis. METHODS We profiled 54 CRC liver metastases, comparing them with 198 other metastatic lesions and normal liver tissues. By analyzing immune cell infiltration, stromal interactions, and key genomic alterations, we constructed an 11-gene prognostic model to predict survival and immunotherapy outcomes. RESULTS CRC liver metastases with high-risk profiles demonstrated enriched follicular helper T cells, activated dendritic cells, and M2 macrophages in the TME. Frequent mutations in APC, TP53, KRAS, and PIK3CA were identified, alongside altered EGFR signaling. The 11-gene model effectively stratified patients by prognosis and predicted immunotherapy responses, emphasizing the therapeutic potential of targeting resistance mechanisms. CONCLUSIONS This study reveals how genomic and TME-driven factors contribute to drug resistance in CRC liver metastases. Integrating these insights with clinical data could advance precision therapies, addressing the evolving challenge of tumor drug resistance in CRC.
Collapse
Affiliation(s)
- Junjie Kuang
- The Second Department of Oncology, Cancer Center, Huizhou First Hospital, Huizhou, Guangdong, China
| | - Jun Li
- The Second Department of Oncology, Cancer Center, Huizhou First Hospital, Huizhou, Guangdong, China
| | - Siwei Zhou
- The Second Department of Oncology, Cancer Center, Huizhou First Hospital, Huizhou, Guangdong, China
| | - Yi Li
- The Second Department of Oncology, Cancer Center, Huizhou First Hospital, Huizhou, Guangdong, China
| | - Jinbo Lin
- The Second Department of Oncology, Cancer Center, Huizhou First Hospital, Huizhou, Guangdong, China
| | - Weizhen Huang
- The Second Department of Oncology, Cancer Center, Huizhou First Hospital, Huizhou, Guangdong, China.
| | - Xia Yuan
- The Second Department of Oncology, Cancer Center, Huizhou First Hospital, Huizhou, Guangdong, China.
| |
Collapse
|
2
|
Hussain A, Szamreta E, Ning N, Kaminski A, Shah R, Aggarwal J, Adeboyeje G. Potential barriers to implementation of next-generation sequencing in cancer management: a U.S. Physician-based survey. Future Oncol 2025; 21:231-239. [PMID: 39606845 PMCID: PMC11792822 DOI: 10.1080/14796694.2024.2430725] [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: 01/03/2024] [Accepted: 11/12/2024] [Indexed: 11/29/2024] Open
Abstract
BACKGROUND The purpose of this study was to identify barriers to physicians' NGS use and preferred strategies to alleviate these barriers. RESEARCH DESIGN AND METHODS A cross-sectional online survey link was sent to a sample of US oncologists/hematologists, surgeons, and pathologists identified through a panel. The survey collected data, from October-December 2020, on barriers to NGS testing and potential strategies. RESULTS Two hundred physicians participated (mean age: 46.2 years; 65% male; 80% White, mean years in clinical practice: 13.7). Despite the use of NGS testing by all physicians, 99.5% reported concerns/barriers. Reimbursement challenges were the most cited reason (87.5%), followed by lack of knowledge of NGS testing methodologies (81.0%), and lack of clinical utility evidence (80.0%). The most common reimbursement challenge was prior authorizations for NGS testing (72.0%), followed by knowledge of new fee codes for reimbursement or corresponding therapy (68.0%), and paperwork/administrative duties (67.5%). Surgeons were more likely to encounter challenges in using NGS testing than other physicians. CONCLUSIONS The results highlight the barriers reported by oncologists/hematologists, pathologists, and surgeons, which may impact the evolving role of NGS in the context of the overall management of cancer patients.
Collapse
Affiliation(s)
- Arif Hussain
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | | | - Ning Ning
- Evidence and Access, Open Health, Newton, MA, USA
| | | | - Ruchit Shah
- Evidence and Access, Open Health, Bethesda, MD, USA
| | | | | |
Collapse
|
3
|
Cuello M, García-Rivello H, Huamán-Garaicoa F, Irigoyen-Piñeiros P, Lara-Torres CO, Rizzo MM, Ticona-Castro M, Trejo R, Zoroquiain P. Detection of NTRK gene fusions in solid tumors: recommendations from a Latin American group of oncologists and pathologists. Future Oncol 2023; 19:2669-2682. [PMID: 38088163 DOI: 10.2217/fon-2023-0552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2023] Open
Abstract
NTRK gene fusions have been detected in more than 25 types of tumors and their prevalence is approximately 0.3% in solid tumors. This low prevalence makes identifying patients who could benefit from TRK inhibitors a considerable challenge. Furthermore, while numerous papers on the evaluation of NTRK fusion genes are available, not all countries have guidelines that are suitable for their setting, as is the case with Latin America. Therefore, a group of oncologists and pathologists from several countries in Latin America (Argentina, Chile, Ecuador, Mexico, Peru and Uruguay) met to discuss and reach consensus on how to identify patients with NTRK gene fusions in solid tumors. To do so, they developed a practical algorithm, considering their specific situation and limitations.
Collapse
Affiliation(s)
- Mauricio Cuello
- Academic Unit of Oncology, Hospital de Clínicas Dr. Manuel Quintela, Montevideo, Uruguay
| | - Hernán García-Rivello
- Departmento of Clinical Pathology, Instituto de Medicina Traslacional e Ingeniería Biomédica (IMTIB), Hospital Italiano, Buenos Aires, Argentina
| | - Fuad Huamán-Garaicoa
- Instituto de Salud Integral (ISAIN), Universidad Católica, Santiago de Guayaquil (Ecuador), Department of Pathology, Sociedad de Lucha Contra el Cáncer del Ecuador (SOLCA), Guayaquil, Ecuador
| | | | - César O Lara-Torres
- Laboratory of Molecular Pathology, Instituto Nacional de Cancerología, Ciudad de México, México
| | - Manglio M Rizzo
- Cancer Immunobiology Laboratory, Instituto de Investigaciones en Medicina Traslacional (IIMT), Facultad de Ciencias Biomédicas, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Austral, Derqui-Pilar, Argentina
- Department of Medical Oncology, Hospital Universitario Austral, Derqui-Pilar, Argentina
| | - Miguel Ticona-Castro
- Service of Medical Oncology, Hospital Nacional Edgardo Rebagliati Martins, EsSalud - Jesús María, Lima (Perú), Clínica Montefiori, La Molina, Lima, Perú
| | - Rogelio Trejo
- Department of Medical Oncology, Centro Médico Nacional Siglo XXI, Ciudad de México, México
| | - Pablo Zoroquiain
- Pathology Department, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
4
|
Alsolme E, Alqahtani S, Fageeh M, Barakeh D, Sharma NK, Mangul S, Robinson HA, Fathaddin A, Hauser CAE, Abedalthagafi M. The Genomic Landscape of Colorectal Cancer in the Saudi Arabian Population Using a Comprehensive Genomic Panel. Diagnostics (Basel) 2023; 13:2993. [PMID: 37761360 PMCID: PMC10527739 DOI: 10.3390/diagnostics13182993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Revised: 08/20/2023] [Accepted: 08/28/2023] [Indexed: 09/29/2023] Open
Abstract
PURPOSE Next-generation sequencing (NGS) technology detects specific mutations that can provide treatment opportunities for colorectal cancer (CRC) patients. PATIENTS AND METHODS We analyzed the mutation frequencies of common actionable genes and their association with clinicopathological characteristics and oncologic outcomes using targeted NGS in 107 Saudi Arabian patients without a family history of CRC. RESULTS Approximately 98% of patients had genetic alterations. Frequent mutations were observed in BRCA2 (79%), CHEK1 (78%), ATM (76%), PMS2 (76%), ATR (74%), and MYCL (73%). The APC gene was not included in the panel. Statistical analysis using the Cox proportional hazards model revealed an unusual positive association between poorly differentiated tumors and survival rates (p = 0.025). Although no significant univariate associations between specific mutations or overall mutation rate and overall survival were found, our preliminary analysis of the molecular markers for CRC in a predominantly Arab population can provide insights into the molecular pathways that play a significant role in the underlying disease progression. CONCLUSIONS These results may help optimize personalized therapy when drugs specific to a patient's mutation profile have already been developed.
Collapse
Affiliation(s)
- Ebtehal Alsolme
- Genomic Research Department, King Fahad Medical City, Riyadh 12231, Saudi Arabia; (E.A.); (D.B.)
| | - Saleh Alqahtani
- Royal Clinic and Hepatology Department, King Faisal Specialist Hospital and Research Center, Riyadh 11564, Saudi Arabia;
| | - Musa Fageeh
- Pathology Department, King Saud Medical City, Riyadh 12746, Saudi Arabia;
| | - Duna Barakeh
- Genomic Research Department, King Fahad Medical City, Riyadh 12231, Saudi Arabia; (E.A.); (D.B.)
| | - Nitesh K. Sharma
- The Titus Family Department of Clinical Pharmacy, School of Pharmacy, University of Southern California, Los Angeles, CA 90007, USA; (N.K.S.); (S.M.)
| | - Serghei Mangul
- The Titus Family Department of Clinical Pharmacy, School of Pharmacy, University of Southern California, Los Angeles, CA 90007, USA; (N.K.S.); (S.M.)
| | | | - Amany Fathaddin
- Department of Pathology, Collage of Medicine, King Saud University, Riyadh 11362, Saudi Arabia;
| | - Charlotte A. E. Hauser
- Laboratory for Nanomedicine, Biological & Environmental Science & Engineering (BESE) Division, King Abdullah University of Science and Technology (KAUST), Thuwal 23955, Saudi Arabia;
- Computational Bioscience Research Center (CBRC), King Abdullah University of Science and Technology (KAUST), Thuwal 23955, Saudi Arabia
| | - Malak Abedalthagafi
- Department of Pathology and Laboratory Medicine, Emory School of Medicine, Atlanta, GA 30307, USA
| |
Collapse
|
5
|
Pu T, Peddle A, Zhu J, Tejpar S, Verbandt S. Neoantigen identification: Technological advances and challenges. Methods Cell Biol 2023; 183:265-302. [PMID: 38548414 DOI: 10.1016/bs.mcb.2023.06.005] [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] [Indexed: 04/02/2024]
Abstract
Neoantigens have emerged as promising targets for cutting-edge immunotherapies, such as cancer vaccines and adoptive cell therapy. These neoantigens are unique to tumors and arise exclusively from somatic mutations or non-genomic aberrations in tumor proteins. They encompass a wide range of alterations, including genomic mutations, post-transcriptomic variants, and viral oncoproteins. With the advancements in technology, the identification of immunogenic neoantigens has seen rapid progress, raising new opportunities for enhancing their clinical significance. Prediction of neoantigens necessitates the acquisition of high-quality samples and sequencing data, followed by mutation calling. Subsequently, the pipeline involves integrating various tools that can predict the expression, processing, binding, and recognition potential of neoantigens. However, the continuous improvement of computational tools is constrained by the availability of datasets which contain validated immunogenic neoantigens. This review article aims to provide a comprehensive summary of the current knowledge as well as limitations in neoantigen prediction and validation. Additionally, it delves into the origin and biological role of neoantigens, offering a deeper understanding of their significance in the field of cancer immunotherapy. This article thus seeks to contribute to the ongoing efforts to harness neoantigens as powerful weapons in the fight against cancer.
Collapse
Affiliation(s)
- Ting Pu
- Digestive Oncology Unit, KULeuven, Leuven, Belgium
| | | | - Jingjing Zhu
- de Duve Institute, Université catholique de Louvain, Brussels, Belgium
| | | | | |
Collapse
|
6
|
DiBiase JF, Scharnetzki E, Edelman E, Lucas FL, Helbig P, Rueter J, Han PK, Ziller E, Jacobs EA, Anderson EC. Urban-Rural and Socioeconomic Differences in Patient Knowledge and Perceptions of Genomic Tumor Testing. JCO Precis Oncol 2023; 7:e2200631. [PMID: 36893376 PMCID: PMC10309515 DOI: 10.1200/po.22.00631] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 01/17/2023] [Indexed: 03/11/2023] Open
Abstract
PURPOSE Social determinants of health, such as rurality, income, and education, may widen health disparities by driving variation in patients' knowledge and perceptions of medical interventions. This effect may be greatest for medical technologies that are hard to understand and less accessible. This study explored whether knowledge and perceptions (expectations and attitudes) of patients with cancer toward large-panel genomic tumor testing (GTT), an emerging cancer technology, vary by patient rurality independent of other socioeconomic characteristics (education and income). METHODS Patients with cancer enrolled in a large precision oncology initiative completed surveys measuring rurality, sociodemographic characteristics, and knowledge and perceptions of GTT. We used multivariable linear models to examine differences in GTT knowledge, expectations, and attitudes by patient rurality, education, and income level. Models controlled for age, sex and clinical cancer stage and type. RESULTS Rural patients had significantly lower knowledge of GTT than urban patients using bivariate models (P = .025). However, this association disappeared when adjusting for education and income level: patients with lower educational attainment and lower income had lower knowledge and higher expectations (P ≤ .002), whereas patients with higher income had more positive attitudes (P = .005). Urban patients had higher expectations of GTT compared with patients living in large rural areas (P = .011). Rurality was not associated with attitudes. CONCLUSION Patients' education and income level are associated with knowledge, expectations, and attitudes toward GTT, whereas rurality is associated with patient expectations. These findings suggest that efforts to promote adoption of GTT should focus on improving knowledge and awareness among individuals with low education and income. These differences may lead to downstream disparities in GTT utilization, which should be explored in future research.
Collapse
Affiliation(s)
- Jessica F. DiBiase
- Center for Interdisciplinary Population and Health Research, MaineHealth Institute for Research, Portland, ME
| | - Elizabeth Scharnetzki
- Center for Interdisciplinary Population and Health Research, MaineHealth Institute for Research, Portland, ME
| | | | - F. Lee Lucas
- Center for Interdisciplinary Population and Health Research, MaineHealth Institute for Research, Portland, ME
| | | | | | - Paul K.J. Han
- Center for Interdisciplinary Population and Health Research, MaineHealth Institute for Research, Portland, ME
- Tufts University School of Medicine, Boston, MA
- Division of Cancer Control and Population Sciences, National Cancer Institute, Bethesda, MD
| | - Erika Ziller
- University of Southern Maine, Muskie School of Public Service, Portland, ME
| | - Elizabeth A. Jacobs
- Center for Interdisciplinary Population and Health Research, MaineHealth Institute for Research, Portland, ME
| | - Eric C. Anderson
- Center for Interdisciplinary Population and Health Research, MaineHealth Institute for Research, Portland, ME
- Tufts University School of Medicine, Boston, MA
| |
Collapse
|
7
|
Malapelle U, Pepe F, Pisapia P, Sgariglia R, Nacchio M, Barberis M, Bilh M, Bubendorf L, Büttner R, Cabibi D, Castiglia M, De Andrea CE, de Biase D, Dumur CI, Fontanini G, Freire J, Gristina V, Hofman P, Ilie M, Lozano MD, Merkelbach-Bruse S, Pappesch R, Pelusi N, Roma G, Russo A, Savic S, Siemanowski J, Tallini G, Tischler V, Vander Borght S, Weynand B, Xu T, Troncone G. TargetPlex FFPE-Direct DNA Library Preparation Kit for SiRe NGS panel: an international performance evaluation study. J Clin Pathol 2022; 75:416-421. [PMID: 33766954 DOI: 10.1136/jclinpath-2021-207450] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 03/01/2021] [Accepted: 03/01/2021] [Indexed: 02/05/2023]
Abstract
AIM Next generation sequencing (NGS) represents a key diagnostic tool to identify clinically relevant gene alterations for treatment-decision making in cancer care. However, the complex manual workflow required for NGS has limited its implementation in routine clinical practice. In this worldwide study, we validated the clinical performance of the TargetPlex FFPE-Direct DNA Library Preparation Kit for NGS analysis. Impressively, this new assay obviates the need for separate, labour intensive and time-consuming pre-analytical steps of DNA extraction, purification and isolation from formalin-fixed paraffin embedded (FFPE) specimens in the NGS workflow. METHODS The TargetPlex FFPE-Direct DNA Library Preparation Kit, which enables NGS analysis directly from FFPE, was specifically developed for this study by TargetPlex Genomics Pleasanton, California. Eleven institutions agreed to take part in the study coordinated by the Molecular Cytopathology Meeting Group (University of Naples Federico II, Naples, Italy). All participating institutions received a specific Library Preparation Kit to test eight FFPE samples previously assessed with standard protocols. The analytical parameters and mutations detected in each sample were then compared with those previously obtained with standard protocols. RESULTS Overall, 92.8% of the samples were successfully analysed with the TargetPlex FFPE-Direct DNA Library Preparation Kit on Thermo Fisher Scientific and Illumina platforms. Altogether, in comparison with the standard workflow, the TargetPlex FFPE-Direct DNA Library Preparation Kit was able to detect 90.5% of the variants. CONCLUSION The TargetPlex FFPE-Direct DNA Library Preparation Kit combined with the SiRe panel constitutes a convenient, practical and robust cost-saving solution for FFPE NGS analysis in routine practice.
Collapse
Affiliation(s)
| | - Francesco Pepe
- Public Health, University of Naples Federico II, Naples, Italy
| | | | | | | | - Massimo Barberis
- Clinic Unit of Histopathology and Molecular Diagnostics, Istituto Europeo di Oncologia, Milano, Italy
| | - Michel Bilh
- Department of Pathology, University Hospital Basel, Basel, Switzerland
| | - Lukas Bubendorf
- Department of Pathology, University Hospital Basel, Basel, Switzerland
| | - Reinhard Büttner
- Department of Pathology, University of Cologne, Cologne, Germany
| | - Daniela Cabibi
- Health Promotion Sciences, Maternal and Infantile Care, Internal Medicine and Medical Specialties, University of Palermo, Palermo, Italy
| | - Marta Castiglia
- Surgical, Oncological and Oral Sciences, University of Palermo, Palermo, Italy
| | | | - Dario de Biase
- Medicine (DIMES)a Hospital, Anatomic Pathology Unit, University of Bologna, Bologna, Italy
| | - Catherine I Dumur
- Molecular Diagnostic Department, Aurora Diagnostics, Jacksonville, Florida, USA
| | - Gabriella Fontanini
- Surgical, Medical, Molecular Pathology and Critical Area, University of Pisa, Pisa, Italy
| | - Javier Freire
- Pathology, Hospital Universitario Marques de Valdecilla, Santander, Spain
| | - Valerio Gristina
- Surgical, Oncological and Oral Sciences, University of Palermo, Palermo, Italy
| | | | - Marius Ilie
- Laboratory of Clinical and Experimental Pathology, Université Côte d'Azur, Nice, France
| | - Maria Dolores Lozano
- Pathology, Universidad de Navarra-Clínica Universidad de Navarra, Pamplona, Spain
| | | | - Roberto Pappesch
- Department of Pathology, University of Cologne, Cologne, Germany
| | | | - Gianluca Roma
- R&D Department, TargetPlex Genomics, Belmont, California, USA
| | - Antonio Russo
- Surgical, Oncological and Oral Sciences, University of Palermo, Palermo, Italy
| | - Spasenija Savic
- Department of Pathology, University Hospital Basel, Basel, Switzerland
| | | | | | | | - Sara Vander Borght
- Department of Pathology, Katholieke Universiteit Leuven UZ Leuven, Leuven, Belgium
| | - Birgit Weynand
- Department of Pathology, Katholieke Universiteit Leuven UZ Leuven, Leuven, Belgium
| | - Tom Xu
- R&D Department, SenseCare Medicals, Inc, Pleasanton, California, USA
| | | |
Collapse
|
8
|
Dell'Olio F. Multiplexed Liquid Biopsy and Tumor Imaging Using Surface-Enhanced Raman Scattering. BIOSENSORS 2021; 11:449. [PMID: 34821665 PMCID: PMC8615571 DOI: 10.3390/bios11110449] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 11/09/2021] [Accepted: 11/10/2021] [Indexed: 06/13/2023]
Abstract
The recent improvements in diagnosis enabled by advances in liquid biopsy and oncological imaging significantly better cancer care. Both these complementary approaches, which are used for early tumor detection, characterization, and monitoring, can benefit from applying techniques based on surface-enhanced Raman scattering (SERS). With a detection sensitivity at the single-molecule level, SERS spectroscopy is widely used in cell and molecular biology, and its capability for the in vitro detection of several types of cancer biomarkers is well established. In the last few years, several intriguing SERS applications have emerged, including in vivo imaging for tumor targeting and the monitoring of drug release. In this paper, selected recent developments and trends in SERS applications in the field of liquid biopsy and tumor imaging are critically reviewed, with a special emphasis on results that demonstrate the clinical utility of SERS.
Collapse
Affiliation(s)
- Francesco Dell'Olio
- Department of Electrical and Information Engineering, Polytechnic University of Bari, 70125 Bari, Italy
| |
Collapse
|
9
|
von Itzstein MS, Smith ML, Railey E, White CB, Dieterich JS, Garrett-Mayer L, Bruinooge SS, Freedman AN, De Moor J, Gray SW, Park JY, Yan J, Hoang AQ, Zhu H, Gerber DE. Accessing Targeted Therapies: A Potential Roadblock to Implementing Precision Oncology? JCO Oncol Pract 2021; 17:e999-e1011. [PMID: 33970688 PMCID: PMC8462665 DOI: 10.1200/op.20.00927] [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: 10/20/2020] [Revised: 02/01/2021] [Accepted: 04/14/2021] [Indexed: 11/20/2022] Open
Abstract
PURPOSE Advances in genomic techniques have led to increased use of next-generation sequencing (NGS). We evaluated the extent to which these tests guide treatment decisions. METHODS We developed and distributed a survey assessing NGS use and outcomes to a survey pool of ASCO members. Comparisons between groups were performed with Wilcoxon two-sample, chi-square, and Fisher's exact tests. RESULTS Among 178 respondents, 62% were male, 54% White, and 67% affiliated with academic centers. More than half (56%) indicated that NGS provided actionable information to a moderate or great extent. Use was highest (median ≥ 70% of cases) for lung and gastric cancer, and lowest (median < 25% of cases) in head and neck and genitourinary cancers. Approximately one third of respondents reported that, despite identification of an actionable molecular variant, patients were sometimes or often unable to access the relevant US Food and Drug Administration-approved therapy. When NGS did not provide actionable results, individuals reporting great or moderate guidance overall from NGS in treatment recommendations were more likely to request the compassionate use of an unapproved drug (P < .001), enroll on a clinical trial (P < .01), or treat off-label with a drug approved for another indication (P = .02). CONCLUSION When NGS identifies an actionable result, a substantial proportion of clinicians reported encountering challenges obtaining approved therapies on the basis of these results. Perceived overall impact of NGS appears associated with clinical behavior unrelated to actionable NGS test results, including pursuing off-label or compassionate use of unapproved therapies or referring to a clinical trial.
Collapse
Affiliation(s)
- Mitchell S. von Itzstein
- Division on Hematology and Oncology, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX
- Harold C. Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, Dallas, TX
| | | | | | - Carol B. White
- Research Advocacy Network, Plano, TX
- CBWhite, Evanston, IL
| | | | | | | | | | | | - Stacy W. Gray
- Department of Population Sciences and Medical Oncology, City of Hope, Duarte, CA
| | - Jason Y. Park
- Department of Pathology, UT Southwestern Medical Center, Dallas, TX
| | - Jingsheng Yan
- Department of Population and Data Sciences, UT Southwestern Medical Center, Dallas, TX
| | - Anh Quynh Hoang
- Department of Population and Data Sciences, UT Southwestern Medical Center, Dallas, TX
| | - Hong Zhu
- Department of Population and Data Sciences, UT Southwestern Medical Center, Dallas, TX
| | - David E. Gerber
- Division on Hematology and Oncology, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX
- Harold C. Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, Dallas, TX
- Research Advocacy Network, Plano, TX
- Department of Population and Data Sciences, UT Southwestern Medical Center, Dallas, TX
| |
Collapse
|
10
|
Rodler S, Jung A, Greif PA, Rühlmann K, Apfelbeck M, Tamalunas A, Kretschmer A, Schulz GB, Szabados B, Stief C, Heinemann V, Westphalen CB, Casuscelli J. Routine application of next-generation sequencing testing in uro-oncology-Are we ready for the next step of personalised medicine? Eur J Cancer 2021; 146:1-10. [PMID: 33535139 DOI: 10.1016/j.ejca.2020.12.024] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 12/18/2020] [Indexed: 12/24/2022]
Abstract
AIM OF THE STUDY Next-generation sequencing (NGS) might represent a valuable diagnostic tool to identify somatic alterations and enable personalised medicine in uro-oncology. We aim to determine feasibility and impact of routine NGS in clinical practice. METHODS Tumours from patients with genitourinary cancers were subjected to NGS. Results were discussed in a dedicated molecular tumour board. Statistical analyses included chi-square test and Mann-Whitney U test. RESULTS Between 2017 and 2020, 65 patients with advanced genitourinary cancers were consecutively enrolled. Number of tests increased (28 tests in 2020) and diagnostic turnaround time for generating output decreased (17.5 days [range 13-35]). Median patient's age was 62 years (range 33-84), and most NGS assays were performed upon start of systemic treatment (range 0-6 of treatment lines). 62/66 sequenced samples generated a report. Fifty samples (80.6%) showed at least one molecular alteration. Most prevalent alterations were TP53 (32.3%), PIK3CA (14.5%) and TMPRSS2-ERG (9.7%). Sequencing revealed potentially druggable targets in 29 samples (46.8%). Based on NGS results, six patients underwent therapy change, whereas for three patients, coverage of recommended off-label therapy was denied by health insurances. CONCLUSIONS NGS is increasingly feasible in clinical routine for patients with genitourinary cancers. Number of performed analyses is constantly growing, and turnaround time to therapy recommendation is decreasing. While the majority of tumours harbour clinically relevant mutations, alterations related to urologic cancers are underrepresented, thus treatment changes occurred only in a minority of patients. Further, access to target agents remains a considerable obstacle in the consequent implementation of precision uro-oncology.
Collapse
Affiliation(s)
- Severin Rodler
- Department of Urology, University Hospital, LMU Munich, Munich, Germany
| | - Andreas Jung
- Pathologisches Institut, University Hospital, LMU Munich, Munich, Germany; German Cancer Consortium, DKTK, Heidelberg, Munich, Germany; Comprehensive Cancer Center, Munich, Germany
| | - Philipp A Greif
- Department of Medicine III, University Hospital, LMU Munich, Munich, Germany
| | | | - Maria Apfelbeck
- Department of Urology, University Hospital, LMU Munich, Munich, Germany
| | | | | | - Gerald B Schulz
- Department of Urology, University Hospital, LMU Munich, Munich, Germany
| | | | - Christian Stief
- Department of Urology, University Hospital, LMU Munich, Munich, Germany
| | - Volker Heinemann
- German Cancer Consortium, DKTK, Heidelberg, Munich, Germany; Department of Medicine III, University Hospital, LMU Munich, Munich, Germany; Comprehensive Cancer Center, Munich, Germany
| | - Christoph B Westphalen
- Department of Medicine III, University Hospital, LMU Munich, Munich, Germany; Comprehensive Cancer Center, Munich, Germany
| | - Jozefina Casuscelli
- Department of Urology, University Hospital, LMU Munich, Munich, Germany; Comprehensive Cancer Center, Munich, Germany.
| |
Collapse
|
11
|
Yoon AJ, Santella RM, Wang S, Kutler DI, Carvajal RD, Philipone E, Wang T, Peters SM, Stewart CR, Momen-Heravi F, Troob S, Levin M, AkhavanAghdam Z, Shackelford AJ, Canterbury CR, Shimonosono M, Hernandez BY, McDowell BD, Nakagawa H. MicroRNA-Based Cancer Mortality Risk Scoring System and hTERT Expression in Early-Stage Oral Squamous Cell Carcinoma. JOURNAL OF ONCOLOGY 2021; 2021:8292453. [PMID: 33510789 PMCID: PMC7822680 DOI: 10.1155/2021/8292453] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 12/01/2020] [Accepted: 12/14/2020] [Indexed: 11/17/2022]
Abstract
We have previously constructed a novel microRNA (miRNA)-based prognostic model and cancer-specific mortality risk score formula to predict survival outcome in oral squamous cell carcinoma (OSCC) patients who are already categorized into "early-stage" by the TNM staging system. A total of 836 early-stage OSCC patients were assigned the mortality risk scores. We evaluated the efficacy of various treatment regimens in terms of survival benefit compared to surgery only in patients stratified into high (risk score ≥0) versus low (risk score <0) mortality risk categories. For the high-risk group, surgery with neck dissection significantly improved the 5-year survival to 75% from 46% with surgery only (p < 0.001); a Cox proportional hazard model on time-to-death demonstrated a hazard ratio of 0.37 for surgery with neck dissection (95% CI: 0.2-0.6; p=0.0005). For the low-risk group, surgery only was the treatment of choice associated with 5-year survival benefit. Regardless of treatment selected, those with risk score ≥2 may benefit from additional therapy to prevent cancer relapse. We also identified hTERT (human telomerase reverse transcriptase) as a gene target common to the prognostic miRNAs. There was 22-fold increase in the hTERT expression level in patients with risk score ≥2 compared to healthy controls (p < 0.0005). Overexpression of hTERT was also observed in the patient-derived OSCC organoid compared to that of normal organoid. The DNA cancer vaccine that targets hTERT-expressing cells currently undergoing rigorous clinical evaluation for other tumors can be repurposed to prevent cancer recurrence in these high-risk early-stage oral cancer patients.
Collapse
Affiliation(s)
- Angela J. Yoon
- Columbia University Irving Medical Center, New York, NY, USA
| | | | - Shuang Wang
- Columbia University Mailman School of Public Health, New York, NY, USA
| | | | | | | | - Tian Wang
- Columbia University Mailman School of Public Health, New York, NY, USA
| | - Scott M. Peters
- Columbia University Irving Medical Center, New York, NY, USA
| | | | | | - Scott Troob
- Columbia University Irving Medical Center, New York, NY, USA
| | | | | | | | | | | | - Brenda Y. Hernandez
- Hawaii Tumor Registry, University of Hawaii Cancer Center, Honolulu, HI, USA
| | | | | |
Collapse
|
12
|
Fenton AT, Anderson EC, Scharnetzki E, Reed K, Edelman E, Antov A, Rueter J, Han PKJ. Differences in cancer patients' and clinicians' preferences for disclosure of uncertain genomic tumor testing results. PATIENT EDUCATION AND COUNSELING 2021; 104:3-11. [PMID: 32690398 DOI: 10.1016/j.pec.2020.07.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 06/02/2020] [Accepted: 07/11/2020] [Indexed: 06/11/2023]
Abstract
OBJECTIVE To compare clinicians' and patients' preferences for disclosure of genomic tumor testing (GTT) results; to determine the sensitivity of these disclosure preferences to uncertainty about the actionability of results; and to explore factors associated with disclosure preferences. METHODS Community-based oncology clinicians (n = 94) and patients (n = 1121) were surveyed about their preferences for disclosing GTT results with varying levels of uncertainty (Tiers 1, 2, 3). Descriptive and multivariable regression analyses were used to compare clinicians' and patients' disclosure preferences and their sensitivity to uncertainty, and to explore associations between disclosure preferences and sociodemographic, clinical, and psychological factors. RESULTS Relatively more patients than clinicians preferred disclosure, and their preferences were less sensitive to the uncertainty of GTT results. For patients and clinicians, lower uncertainty sensitivity was associated with positive GTT attitudes; for patients it was also associated with greater uncertainty tolerance and knowledge of uncertainty in GTT. CONCLUSION Relatively more cancer patients than clinicians prefer disclosure of GTT results, and their preferences are less sensitive to result uncertainty. Uncertainty sensitivity in disclosure preferences is associated with GTT-related attitudes and uncertainty tolerance. PRACTICE IMPLICATIONS Differences in cancer patients' and clinicians' preferences for disclosure of uncertain GTT results warrant greater attention in cancer care.
Collapse
Affiliation(s)
- Anny T Fenton
- Center for Outcomes, Research and Evaluation, Maine Medical Center Research Institute, Portland, ME, USA
| | - Eric C Anderson
- Center for Outcomes, Research and Evaluation, Maine Medical Center Research Institute, Portland, ME, USA
| | - Elizabeth Scharnetzki
- Center for Outcomes, Research and Evaluation, Maine Medical Center Research Institute, Portland, ME, USA
| | - Kate Reed
- The Jackson Laboratory, Bar Harbor, ME, USA
| | | | | | | | - Paul K J Han
- Center for Outcomes, Research and Evaluation, Maine Medical Center Research Institute, Portland, ME, USA.
| |
Collapse
|
13
|
Liptay M, Barbosa JS, Rottenberg S. Replication Fork Remodeling and Therapy Escape in DNA Damage Response-Deficient Cancers. Front Oncol 2020; 10:670. [PMID: 32432041 PMCID: PMC7214843 DOI: 10.3389/fonc.2020.00670] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Accepted: 04/09/2020] [Indexed: 12/27/2022] Open
Abstract
Most cancers have lost a critical DNA damage response (DDR) pathway during tumor evolution. These alterations provide a useful explanation for the initial sensitivity of tumors to DNA-targeting chemotherapy. A striking example is dysfunctional homology-directed repair (HDR), e.g., due to inactivating mutations in BRCA1 and BRCA2 genes. Extensive efforts are being made to develop novel targeted therapies exploiting such an HDR defect. Inhibitors of poly(ADP-ribose) polymerase (PARP) are an instructive example of this approach. Despite the success of PARP inhibitors, the presence of primary or acquired therapy resistance remains a major challenge in clinical oncology. To move the field of precision medicine forward, we need to understand the precise mechanisms causing therapy resistance. Using preclinical models, various mechanisms underlying chemotherapy resistance have been identified. Restoration of HDR seems to be a prevalent mechanism but this does not explain resistance in all cases. Interestingly, some factors involved in DNA damage response (DDR) have independent functions in replication fork (RF) biology and their loss causes RF instability and therapy sensitivity. However, in BRCA-deficient tumors, loss of these factors leads to restored stability of RFs and acquired drug resistance. In this review we discuss the recent advances in the field of RF biology and its potential implications for chemotherapy response in DDR-defective cancers. Additionally, we review the role of DNA damage tolerance (DDT) pathways in maintenance of genome integrity and their alterations in cancer. Furthermore, we refer to novel tools that, combined with a better understanding of drug resistance mechanisms, may constitute a great advance in personalized diagnosis and therapeutic strategies for patients with HDR-deficient tumors.
Collapse
Affiliation(s)
- Martin Liptay
- Institute of Animal Pathology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Joana S. Barbosa
- Institute of Animal Pathology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Sven Rottenberg
- Institute of Animal Pathology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
- Bern Center for Precision Medicine, University of Bern, Bern, Switzerland
| |
Collapse
|
14
|
Detection of Circulating Tumor DNA with a Single-Molecule Sequencing Analysis Validated for Targeted and Immunotherapy Selection. Mol Diagn Ther 2020; 23:521-535. [PMID: 31209714 PMCID: PMC6675782 DOI: 10.1007/s40291-019-00406-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
INTRODUCTION Comprehensive genetic cancer profiling using circulating tumor DNA has enabled the detection of National Comprehensive Cancer Network (NCCN) guideline-recommended somatic alterations from a single, non-invasive blood draw. However, reliably detecting somatic variants at low variant allele fractions (VAFs) remains a challenge for next-generation sequencing (NGS)-based tests. We have developed the single-molecule sequencing (SMSEQ) platform to address these challenges. METHODS The OncoLBx assay utilizes the SMSEQ platform to optimize cell-free DNA extraction and library preparation with variant type-specific calling algorithms to improve sensitivity and specificity. OncoLBx is a pan-cancer panel for solid tumors targeting 75 genes and five microsatellite sites analyzing five classes of NCCN-recommended somatic variants: single-nucleotide variants (SNVs), insertions and deletions (indels), copy number variants (CNVs), fusions and microsatellite instability (MSI). Circulating DNA was extracted from plasma, followed by library preparation using SMSEQ. Analytical validation was performed according to recently published American College of Medical Genetics and Genomics (ACMG)/Association for Molecular Pathology (AMP) guidelines and established the limit of detection (LOD), sensitivity, specificity, accuracy and reproducibility using 126 gold-standard reference samples, healthy donor samples verified by whole-exome sequencing by an external College of American Pathologists (CAP) reference lab and cell lines with known variants. Results were analyzed using a locus-specific modeling algorithm. RESULTS We have demonstrated that OncoLBx detects VAFs of ≥ 0.1% for SNVs and indels, ≥ 0.5% for fusions, ≥ 4.5 copies for CNVs and ≥ 2% for MSI, with all variant types having specificity ≥ 99.999%. Diagnostic performance of paired samples displays 80% sensitivity and > 99.999% clinical specificity. Clinical utility and performance were assessed in 416 solid tumor samples. Variants were detected in 79% of samples, for which 87.34% of positive samples had available targeted therapy.
Collapse
|
15
|
Validating a targeted next-generation sequencing assay and profiling somatic variants in Chinese non-small cell lung cancer patients. Sci Rep 2020; 10:2070. [PMID: 32034196 PMCID: PMC7005734 DOI: 10.1038/s41598-020-58819-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Accepted: 11/29/2019] [Indexed: 02/05/2023] Open
Abstract
Non-small cell lung cancer (NSCLC) is featured with complex genomic alterations. Molecular profiling of large cohort of NSCLC patients is thus a prerequisite for precision medicine. We first validated the detection performance of a next-generation sequencing (NGS) cancer hotspot panel, OncoAim, on formalin-fixed paraffin-embedded (FFPE) samples. We then utilized OncoAim to delineate the genomic aberrations in Chinese NSCLC patients. Overall detection performance was powerful for mutations with allele frequency (MAF) ≥ 5% at >500 × coverage depth, with >99% sensitivity, high specificity (positive predictive value > 99%), 94% accuracy and 96% repeatability. Profiling 422 NSCLC FFPE samples revealed that patient characteristics, including gender, age, lymphatic spread, histologic grade and histologic subtype were significantly associated with the mutation incidence of EGFR and TP53. Moreover, RTK signaling pathway activation was enriched in adenocarcinoma, while PI(3)K pathway activation, oxidative stress pathway activation, and TP53 pathway inhibition were more prevalent in squamous cell carcinoma. Additionally, novel co-existence (e.g., variants in BRAF and PTEN) and mutual-exclusiveness (e.g., alterations in EGFR and NFE2L2) were found. Finally, we revealed distinct mutation spectrum in TP53, as well as a previously undervalued PTEN aberration. Our findings could aid in improving diagnosis, prognosis and personalized therapeutic decisions of Chinese NSCLC patients.
Collapse
|
16
|
Novo M, Castellino A, Nicolosi M, Santambrogio E, Vassallo F, Chiappella A, Vitolo U. High-grade B-cell lymphoma: how to diagnose and treat. Expert Rev Hematol 2019; 12:497-506. [DOI: 10.1080/17474086.2019.1624157] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Affiliation(s)
- Mattia Novo
- Hematology, Città della Salute e della Scienza Hospital and University, Torino, Italy
| | - Alessia Castellino
- Hematology, Città della Salute e della Scienza Hospital and University, Torino, Italy
| | - Maura Nicolosi
- Hematology, Città della Salute e della Scienza Hospital and University, Torino, Italy
| | - Elisa Santambrogio
- Hematology, Città della Salute e della Scienza Hospital and University, Torino, Italy
| | - Francesco Vassallo
- Hematology, Città della Salute e della Scienza Hospital and University, Torino, Italy
| | - Annalisa Chiappella
- Hematology, Città della Salute e della Scienza Hospital and University, Torino, Italy
| | - Umberto Vitolo
- Hematology, Città della Salute e della Scienza Hospital and University, Torino, Italy
| |
Collapse
|
17
|
Mallampati S, Duose DY, Harmon MA, Mehrotra M, Kanagal-Shamanna R, Zalles S, Wistuba II, Sun X, Luthra R. Rational "Error Elimination" Approach to Evaluating Molecular Barcoded Next-Generation Sequencing Data Identifies Low-Frequency Mutations in Hematologic Malignancies. J Mol Diagn 2019; 21:471-482. [PMID: 30794984 PMCID: PMC6521894 DOI: 10.1016/j.jmoldx.2019.01.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Revised: 10/31/2018] [Accepted: 01/18/2019] [Indexed: 12/18/2022] Open
Abstract
The emergence of highly sensitive molecular diagnostic approaches, such as droplet digital PCR, has allowed the accurate identification of low-frequency variant alleles in clinical specimens; however, the multiplex capabilities of droplet digital PCR for variant detection are inadequate. The incorporation of molecular barcodes or unique IDs into next-generation sequencing libraries through PCR has enabled the detection of low-frequency variant alleles across multiple genomic regions. However, rational library preparation and sequencing data analytic strategies that integrate molecular barcodes have rarely been applied to clinical settings. In this study, we evaluated the parameters that are crucial in the use of molecular barcodes in next-generation sequencing for genotyping clinical specimens from patients with hematologic malignancies. The uniform incorporation of molecular barcodes into DNA templates through PCR was found to be crucial, and the extent of uniformity was governed by multiple interdependent variables. An error elimination strategy was developed for removing sequencing background errors by using molecular barcode sequence information as an alternative to the conventional error correction approach. This approach was successfully used to identify mutations with frequencies as low as 0.15%, and the clonal heterogeneity of hematologic malignancies was revealed. These findings have implications for elucidating heterogeneity and temporal and spatial clonal evolution, evaluating response to therapy, and monitoring relapse in patients with hematologic malignancies.
Collapse
Affiliation(s)
- Saradhi Mallampati
- Department of Laboratory Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas; Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Dzifa Y Duose
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | | | - Meenakshi Mehrotra
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Rashmi Kanagal-Shamanna
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Stephanie Zalles
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Ignacio I Wistuba
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Xiaoping Sun
- Department of Laboratory Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Rajyalakshmi Luthra
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas; Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| |
Collapse
|
18
|
Ileana Dumbrava E, Brusco L, Daniels MS, Wathoo C, Shaw KR, Lu KH, Zheng X, Strong LC, Litton J, Arun BK, Eterovic AK, Routbort MJ, Patel KP, Qi Y, Piha-Paul SA, Subbiah V, Hong DS, Rodon J, Kopetz S, Mendelsohn J, Mills GB, Chen K, Meric-Bernstam F. Expanded analysis of secondary germline findings from matched tumor/normal sequencing identifies additional clinically significant mutations. JCO Precis Oncol 2019; 3:PO.18.00143. [PMID: 31517177 PMCID: PMC6741435 DOI: 10.1200/po.18.00143] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/17/2018] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Next-generation sequencing (NGS) for tumor molecular profiling can reveal secondary germline pathogenic and likely pathogenic variants (LPV/PV). The American College of Medical Genetics (ACMG) recommends return of secondary results for a subset of 59 genes, but other genes with evidence of clinical utility are emerging. We previously reported that 4.3% of patients who underwent NGS of a targeted panel of 201 genes had LPV/PV based on the ACMG list. Here we report the frequency of additional germline cancer-related gene variants and discuss their clinical utility. PATIENTS AND METHODS Matched tumor and germline DNA NGS of a targeted panel of 201 genes was performed in a research laboratory on samples from 1000 patients with advanced or metastatic solid tumors enrolled in a molecular testing protocol (NCT01772771). The frequency of germline LPV/PV in 54 cancer-related genes, beyond the genes in ACMG list, were analyzed. RESULTS Among 1000 patients who underwent tumor/normal DNA sequencing, 46 (4.6%) were found to have a germline LPV/PV in the following genes: AR-(5), ATM-(4), BAP1-(1), CDH1-(1), CDKN2A-(1), CHEK1-(2), CHEK2-(10), EGFR-(1), ERCC3-(4), ERCC5-(1), HNF1B-(1), HRAS-(1), MITF-(4), MLL3-(1), NF1-(3), PKHD1-(4), PTCH1-(1), and SMARCA4-(1). Thus, a total 8.7% of patients had an LPV/PV with 2 patients having 2 concomitant germline LPV/PV. Five mutations in high-penetrance hereditary cancer predisposition genes were selected to be returned to patients or their representatives: BAP1, CDH1, CDKN2A, EGFR, and SMARCA4. CONCLUSIONS Broader genomic testing is likely to identify additional secondary pathogenic germline alterations, some with potential clinical utility for return to patients and their relatives. The recommended genes for which germline results should be returned are continually changing, warranting continued study.
Collapse
Affiliation(s)
| | - Lauren Brusco
- The University of Texas MD Anderson Cancer Center, Houston, TX
| | | | - Chetna Wathoo
- The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Kenna R. Shaw
- The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Karen H. Lu
- The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Xiaofeng Zheng
- The University of Texas MD Anderson Cancer Center, Houston, TX
| | | | - Jennifer Litton
- The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Banu K. Arun
- The University of Texas MD Anderson Cancer Center, Houston, TX
| | | | | | - Keyur P. Patel
- The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Yuan Qi
- The University of Texas MD Anderson Cancer Center, Houston, TX
| | | | - Vivek Subbiah
- The University of Texas MD Anderson Cancer Center, Houston, TX
| | - David S. Hong
- The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Jordi Rodon
- The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Scott Kopetz
- The University of Texas MD Anderson Cancer Center, Houston, TX
| | - John Mendelsohn
- The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Gordon B. Mills
- The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Ken Chen
- The University of Texas MD Anderson Cancer Center, Houston, TX
| | | |
Collapse
|
19
|
Yip S, Christofides A, Banerji S, Downes MR, Izevbaye I, Lo B, MacMillan A, McCuaig J, Stockley T, Yousef GM, Spatz A. A Canadian guideline on the use of next-generation sequencing in oncology. Curr Oncol 2019; 26:e241-e254. [PMID: 31043833 PMCID: PMC6476432 DOI: 10.3747/co.26.4731] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Rapid advancements in next-generation sequencing (ngs) technology have created an unprecedented opportunity to decipher the molecular profile of tumours to more effectively prevent, diagnose, and treat cancer. Oncologists now have the option to order molecular tests that can guide treatment decisions. However, to date, most oncologists have received limited training in genomics, and they are now faced with the challenge of understanding how such tests and their interpretation align with patient management. Guidance on how to effectively use ngs technology is therefore needed to aid oncologists in applying the results of genomic tests. The Canadian guideline presented here describes best practices and unmet needs related to ngs-based testing for somatic variants in oncology, including clinical application, assay and sample selection, bioinformatics and interpretation of reports performed by laboratories, patient communication, and clinical trials.
Collapse
Affiliation(s)
- S Yip
- Cancer Genetics and Genomics Lab, BC Cancer, Vancouver, BC
| | | | - S Banerji
- Department of Medical Oncology, CancerCare Manitoba, Winnipeg, MB
| | - M R Downes
- Anatomic Pathology, Sunnybrook Health Sciences Centre, Toronto, ON
| | - I Izevbaye
- Division of Molecular Pathology, Laboratory Medicine and Pathology, University of Alberta Hospital, Edmonton, AB
| | - B Lo
- Molecular Diagnostics, The Ottawa Hospital, Ottawa, ON
| | - A MacMillan
- Provincial Medical Genetics Program, St. John's, NL
| | - J McCuaig
- Princess Margaret Cancer Centre, Toronto, ON
| | - T Stockley
- Department of Laboratory Medicine and Pathobiology, University of Toronto and University Health Network, Toronto, ON
| | - G M Yousef
- Department of Pediatric Laboratory Medicine, The Hospital for Sick Children, Toronto, ON
| | - A Spatz
- Departments of Pathology and Oncology, McGill University, McGill University Health Centre and Lady Davis Institute, Montreal, QC
| |
Collapse
|
20
|
Phan N, Hong JJ, Tofig B, Mapua M, Elashoff D, Moatamed NA, Huang J, Memarzadeh S, Damoiseaux R, Soragni A. A simple high-throughput approach identifies actionable drug sensitivities in patient-derived tumor organoids. Commun Biol 2019; 2:78. [PMID: 30820473 PMCID: PMC6389967 DOI: 10.1038/s42003-019-0305-x] [Citation(s) in RCA: 178] [Impact Index Per Article: 29.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Accepted: 01/15/2019] [Indexed: 12/25/2022] Open
Abstract
Tumor organoids maintain cell-cell interactions, heterogeneity, microenvironment, and drug response of the sample they originate from. Thus, there is increasing interest in developing tumor organoid models for drug development and personalized medicine applications. Although organoids are in principle amenable to high-throughput screenings, progress has been hampered by technical constraints and extensive manipulations required by current methods. Here we introduce a miniaturized method that uses a simplified geometry by seeding cells around the rim of the wells (mini-rings). This allows high-throughput screenings in a format compatible with automation as shown using four patient-derived tumor organoids established from two ovarian and one peritoneal high-grade serous carcinomas and one carcinosarcoma of the ovary. Using our automated screening platform, we identified personalized responses by measuring viability, number, and size of organoids after exposure to 240 kinase inhibitors. Results are available within a week from surgery, a timeline compatible with therapeutic decision-making.
Collapse
Affiliation(s)
- Nhan Phan
- Division of Hematology-Oncology, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA
- Laboratory of Stem Cell Research and Application, University of Science, Vietnam National University, HCM City, Vietnam
| | - Jenny J Hong
- Division of Hematology-Oncology, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA
| | - Bobby Tofig
- Molecular Screening Shared Resource, California NanoSystems Institute, University of California, Los Angeles, CA, 90095, USA
| | - Matthew Mapua
- Division of Hematology-Oncology, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA
| | - David Elashoff
- Department of Biostatistics, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA
| | - Neda A Moatamed
- Department of Pathology, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA
| | - Jin Huang
- Department of Obstetrics and Gynecology, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA
| | - Sanaz Memarzadeh
- Department of Obstetrics and Gynecology, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, CA, 90095, USA
- The VA Greater Los Angeles Health Care System, Los Angeles, CA, 90073, USA
- Department of Biological Chemistry, University of California, Los Angeles, CA, 90095, USA
- Molecular Biology Institute, University of California, Los Angeles, CA, 90095, USA
| | - Robert Damoiseaux
- Molecular Screening Shared Resource, California NanoSystems Institute, University of California, Los Angeles, CA, 90095, USA
- Department of Molecular and Medicinal Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA
| | - Alice Soragni
- Division of Hematology-Oncology, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA.
- Molecular Biology Institute, University of California, Los Angeles, CA, 90095, USA.
| |
Collapse
|
21
|
Zhang X, Liang Z, Wang S, Lu S, Song Y, Cheng Y, Ying J, Liu W, Hou Y, Li Y, Liu Y, Hou J, Liu X, Shao J, Tai Y, Wang Z, Fu L, Li H, Zhou X, Bai H, Wang M, Lu Y, Yang J, Zhong W, Zhou Q, Yang X, Wang J, Huang C, Liu X, Zhou X, Zhang S, Tian H, Chen Y, Ren R, Liao N, Wu C, Zhu Z, Pan H, Gu Y, Wang L, Liu Y, Zhang S, Liu T, Chen G, Shao Z, Xu B, Zhang Q, Xu R, Shen L, Wu Y, Tumor Biomarker Committee OBOCSOCO(CSCO. Application of next-generation sequencing technology to precision medicine in cancer: joint consensus of the Tumor Biomarker Committee of the Chinese Society of Clinical Oncology. Cancer Biol Med 2019; 16:189-204. [PMID: 31119060 PMCID: PMC6528448 DOI: 10.20892/j.issn.2095-3941.2018.0142] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Accepted: 12/20/2018] [Indexed: 02/05/2023] Open
Abstract
Next-generation sequencing (NGS) technology is capable of sequencing millions or billions of DNA molecules simultaneously. Therefore, it represents a promising tool for the analysis of molecular targets for the initial diagnosis of disease, monitoring of disease progression, and identifying the mechanism of drug resistance. On behalf of the Tumor Biomarker Committee of the Chinese Society of Clinical Oncology (CSCO) and the China Actionable Genome Consortium (CAGC), the present expert group hereby proposes advisory guidelines on clinical applications of NGS technology for the analysis of cancer driver genes for precision cancer therapy. This group comprises an assembly of laboratory cancer geneticists, clinical oncologists, bioinformaticians, pathologists, and other professionals. After multiple rounds of discussions and revisions, the expert group has reached a preliminary consensus on the need of NGS in clinical diagnosis, its regulation, and compliance standards in clinical sample collection. Moreover, it has prepared NGS criteria, the sequencing standard operation procedure (SOP), data analysis, report, and NGS platform certification and validation.
Collapse
Affiliation(s)
- Xuchao Zhang
- Guangdong Lung Cancer Institute, Medical Research Center, Cancer Center of Guangdong Provincial People's Hospital and Guangdong Academy of Medical Sciences, Guangzhou 510080, China
- Affiliated Guangdong Provincial People's Hospital, South China University of Technology, Guangzhou 510630, China
| | - Zhiyong Liang
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing 100006, China
| | - Shengyue Wang
- National Research Center for Translational Medicine, Shanghai, RuiJin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200240, China
| | - Shun Lu
- Lung Tumor Clinical Medical Center, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Yong Song
- Division of Respiratory Medicine, Jinling Hospital, Nanjing University School of Medicine, Nanjing 210029, China
| | - Ying Cheng
- Department of Oncology, Jilin Cancer Hospital, Changchun 132002, China
| | - Jianming Ying
- Department of Pathology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100006, China
| | - Weiping Liu
- Department of Pathology, West China Hospital, Sichuan University, Chengdu 610065, China
| | - Yingyong Hou
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai 200433, China
| | - Yangqiu Li
- Department of Hematology, First Affiliated Hospital, Institute of Hematology, School of Medicine, Jinan University, Guangzhou 519000, China
| | - Yi Liu
- Laboratory of Oncology, Affiliated Hospital of the Academy of Military Medical Sciences, Beijing 100071, China
| | - Jun Hou
- Department of Oncology, First Clinical College of South China University of Technology/Guangdong Lung Cancer Institute, Guangzhou 510060, China
| | - Xiufeng Liu
- People's Liberation Army Cancer Center of Bayi Hospital Affiliated to Nanjing University of Chinese Medicine, Nanjing 210046, China
| | - Jianyong Shao
- Department of Pathology, Sun Yat-sen University Cancer Center, Guangzhou 519000, China
| | - Yanhong Tai
- Department of Pathology, Affiliated Hospital of the Academy of Military Medical Sciences, Beijing 100071, China
| | - Zheng Wang
- Department of Pathology, Beijing Hospital, Beijing 100071, China
| | - Li Fu
- Department of Breast Cancer Pathology and Research Laboratory of Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin’s Clinical Research Center for Cancer, Tianjin 300060, China
| | - Hui Li
- Department of Oncology, Jilin Cancer Hospital, Changchun 132002, China
| | - Xiaojun Zhou
- Department of Pathology, Jinling Hospital Nanjing University School of Medicine, Nanjing 210029, China
| | - Hua Bai
- State Key Laboratory of Molecular Oncology, Department of Medical Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100006, China
| | - Mengzhao Wang
- Department of Respiratory Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing 100006, China
| | - You Lu
- Department of Oncology, West China Hospital, Sichuan University, Chengdu 610065, China
| | - Jinji Yang
- Guangdong Lung Cancer Institute, Guangdong Provincical Prople's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| | - Wenzhao Zhong
- Guangdong Lung Cancer Institute, Guangdong Provincical Prople's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| | - Qing Zhou
- Guangdong Lung Cancer Institute, Guangdong Provincical Prople's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| | - Xuening Yang
- Guangdong Lung Cancer Institute, Guangdong Provincical Prople's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| | - Jie Wang
- Department of Medical Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100006, China
| | - Cheng Huang
- Department of Thoracic Oncology, Fujian Cancer Hospital, Fujian Medical University Cancer Hospital, Fuzhou 350001, China
| | - Xiaoqing Liu
- Department of Oncology, Affiliated Hospital of the Academy of Military Medical Sciences, Beijing 100071, China
| | - Xiaoyan Zhou
- Department of Pathology, Shanghai Cancer Center, Fudan University, Shanghai 200433, China
| | - Shirong Zhang
- Center for Translational Medicine, Hangzhou First People's Hospital, Nanjing Medical University, Nanjing 210029, China
| | - Hongxia Tian
- Guangdong Lung Cancer Institute, Medical Research Center, Cancer Center of Guangdong Provincial People's Hospital and Guangdong Academy of Medical Sciences, Guangzhou 510080, China
- Affiliated Guangdong Provincial People's Hospital, South China University of Technology, Guangzhou 510630, China
| | - Yu Chen
- Guangdong Lung Cancer Institute, Medical Research Center, Cancer Center of Guangdong Provincial People's Hospital and Guangdong Academy of Medical Sciences, Guangzhou 510080, China
- Affiliated Guangdong Provincial People's Hospital, South China University of Technology, Guangzhou 510630, China
| | - Ruibao Ren
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, RuiJin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200240, China
| | - Ning Liao
- Department of Breast Cancer, Cancer Center, Guangdong Provincial People's Hospital, Guangzhou 510080, China
| | - Chunyan Wu
- Department of Pathology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200240, China
| | - Zhongzheng Zhu
- Department of Oncology, No. 113 Hospital of People's Liberation Army, Ningbo 315040, China
| | - Hongming Pan
- Department of Medical Oncology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou 310020, China
| | - Yanhong Gu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing 210029, China
| | - Liwei Wang
- Department of Oncology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200240, China
| | - Yunpeng Liu
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang 110016, China
| | - Suzhan Zhang
- Department of Oncology, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310020, China
| | - Tianshu Liu
- Department of Oncology, Zhongshan Hospital, Fudan University, Shanghai 200433, China
| | - Gong Chen
- Department of Colorectal, Sun Yat-sen University Cancer Center, Guangzhou 519000, China
| | - Zhimin Shao
- Department of Breast Surgery, Shanghai Cancer Center, Fudan University, Shanghai 200433, China
| | - Binghe Xu
- Department of Medical Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100006, China
| | - Qingyuan Zhang
- Department of Internal Medicine, The Third Affiliated Hospital of Harbin Medical University, Harbin 150030, China
| | - Ruihua Xu
- Department of Medical Oncology, Sun Yat-Sen University Cancer Center, Guangzhou 519000, China
| | - Lin Shen
- Department of Gastrointestinal Oncology, Peking University Cancer Hospital and Institute, Beijing 100142, China
| | - Yilong Wu
- Guangdong Lung Cancer Institute, Medical Research Center, Cancer Center of Guangdong Provincial People's Hospital and Guangdong Academy of Medical Sciences, Guangzhou 510080, China
- Affiliated Guangdong Provincial People's Hospital, South China University of Technology, Guangzhou 510630, China
| | | |
Collapse
|
22
|
Chen X, Chang CW, Spoerke JM, Yoh KE, Kapoor V, Baudo C, Aimi J, Yu M, Liang-Chu MMY, Suttmann R, Huw LY, Gendreau S, Cummings C, Lackner MR. Low-pass Whole-genome Sequencing of Circulating Cell-free DNA Demonstrates Dynamic Changes in Genomic Copy Number in a Squamous Lung Cancer Clinical Cohort. Clin Cancer Res 2019; 25:2254-2263. [PMID: 30617129 DOI: 10.1158/1078-0432.ccr-18-1593] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Revised: 10/01/2018] [Accepted: 01/03/2019] [Indexed: 11/16/2022]
Abstract
PURPOSE We developed a method to monitor copy number variations (CNV) in plasma cell-free DNA (cfDNA) from patients with metastatic squamous non-small cell lung cancer (NSCLC). We aimed to explore the association between tumor-derived cfDNA and clinical outcomes, and sought CNVs that may suggest potential resistance mechanisms. EXPERIMENTAL DESIGN Sensitivity and specificity of low-pass whole-genome sequencing (LP-WGS) were first determined using cell line DNA and cfDNA. LP-WGS was performed on baseline and longitudinal cfDNA of 152 patients with squamous NSCLC treated with chemotherapy, or in combination with pictilisib, a pan-PI3K inhibitor. cfDNA tumor fraction and detected CNVs were analyzed in association with clinical outcomes. RESULTS LP-WGS successfully detected CNVs in cfDNA with tumor fraction ≥10%, which represented approximately 30% of the first-line NSCLC patients in this study. The most frequent CNVs were gains in chromosome 3q, which harbors the PIK3CA and SOX2 oncogenes. The CNV landscape in cfDNA with a high tumor fraction generally matched that of corresponding tumor tissue. Tumor fraction in cfDNA was dynamic during treatment, and increases in tumor fraction and corresponding CNVs could be detected before radiographic progression in 7 of 12 patients. Recurrent CNVs, such as MYC amplification, were enriched in cfDNA from posttreatment samples compared with the baseline, suggesting a potential resistance mechanism to pictilisib. CONCLUSIONS LP-WGS offers an unbiased and high-throughput way to investigate CNVs and tumor fraction in cfDNA of patients with cancer. It may also be valuable for monitoring treatment response, detecting disease progression early, and identifying emergent clones associated with therapeutic resistance.
Collapse
Affiliation(s)
- Xiaoji Chen
- Department of Oncology Biomarker Development, Genentech, South San Francisco, California.
| | - Ching-Wei Chang
- Department of Biostatistics, Genentech, South San Francisco, California
| | - Jill M Spoerke
- Department of Oncology Biomarker Development, Genentech, South San Francisco, California
| | - Kathryn E Yoh
- Department of Oncology Biomarker Development, Genentech, South San Francisco, California
| | - Vidushi Kapoor
- Department of Oncology Biomarker Development, Genentech, South San Francisco, California
| | - Charles Baudo
- Department of Oncology Biomarker Development, Genentech, South San Francisco, California
| | - Junko Aimi
- Department of Oncology Biomarker Development, Genentech, South San Francisco, California
| | - Mamie Yu
- Department of Discovery Oncology, Genentech, South San Francisco, California
| | - May M Y Liang-Chu
- Department of Discovery Oncology, Genentech, South San Francisco, California
| | - Rebecca Suttmann
- Department of Oncology Biomarker Development, Genentech, South San Francisco, California
| | - Ling-Yuh Huw
- Department of Oncology Biomarker Development, Genentech, South San Francisco, California
| | - Steven Gendreau
- Department of Oncology Biomarker Development, Genentech, South San Francisco, California
| | - Craig Cummings
- Department of Oncology Biomarker Development, Genentech, South San Francisco, California
| | - Mark R Lackner
- Department of Oncology Biomarker Development, Genentech, South San Francisco, California.
| |
Collapse
|
23
|
Barbour JA, Wong JWH. Dysregulation of Cis-Regulatory Elements in Cancer. Clin Epigenetics 2019. [DOI: 10.1007/978-981-13-8958-0_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022] Open
|
24
|
Roberts JS, Gornick MC, Le LQ, Bartnik NJ, Zikmund-Fisher BJ, Chinnaiyan AM. Next-generation sequencing in precision oncology: Patient understanding and expectations. Cancer Med 2019; 8:227-237. [PMID: 30600607 PMCID: PMC6346219 DOI: 10.1002/cam4.1947] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 12/04/2018] [Accepted: 12/05/2018] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Implementation of precision oncology interventions poses several challenges to informed consent and patient education. This study assessed cancer patients' understanding, expectations, and outcomes regarding participation in research examining the impact of matched tumor and germline sequencing on their clinical care. METHODS A total of 297 patients (mean age: 59 years; 50% female; 96% white) with refractory, metastatic cancer were surveyed, including 217 who completed surveys both before and after undergoing integrated whole exome and transcriptome sequencing as part of a larger clinical research study. RESULTS At baseline, the vast majority of patients expected to receive several potential direct benefits from study participation, including written reports of sequencing findings (88%), greater understanding of the causes of their cancer (74%), and participation in clinical trials for which sequencing results would make them eligible (84%). In most cases, these benefits were not realized by study completion. Despite explanations from study personnel to the contrary, most participants (67%-76%) presumed that incidental germline sequencing findings relevant to noncancerous health conditions (eg, diabetes) would automatically be disclosed to them. Patients reported low levels of concern about study risks at baseline and low levels of regret about study participation at follow-up. CONCLUSIONS Findings suggest that cancer patients participating in precision oncology intervention research have largely unfulfilled expectations of direct benefits related to their study participation. Increased focus on patient education to supplement the informed consent process may help manage patients' expectations regarding the extent and likelihood of benefits received as a result of undergoing genomic sequencing.
Collapse
Affiliation(s)
- J. Scott Roberts
- Department of Health Behavior and Health EducationUniversity of Michigan School of Public HealthAnn ArborMichigan
- Center for Bioethics & Social Sciences in MedicineUniversity of Michigan Medical SchoolAnn ArborMichigan
- Rogel Cancer CenterUniversity of Michigan Medical SchoolAnn ArborMichigan
| | - Michele C. Gornick
- Center for Bioethics & Social Sciences in MedicineUniversity of Michigan Medical SchoolAnn ArborMichigan
- Rogel Cancer CenterUniversity of Michigan Medical SchoolAnn ArborMichigan
- Department of Internal MedicineUniversity of Michigan Medical SchoolAnn ArborMichigan
| | - Lan Q. Le
- Department of Health Behavior and Health EducationUniversity of Michigan School of Public HealthAnn ArborMichigan
| | - Natalie J. Bartnik
- Department of Health Behavior and Health EducationUniversity of Michigan School of Public HealthAnn ArborMichigan
| | - Brian J. Zikmund-Fisher
- Department of Health Behavior and Health EducationUniversity of Michigan School of Public HealthAnn ArborMichigan
- Center for Bioethics & Social Sciences in MedicineUniversity of Michigan Medical SchoolAnn ArborMichigan
- Rogel Cancer CenterUniversity of Michigan Medical SchoolAnn ArborMichigan
- Department of Internal MedicineUniversity of Michigan Medical SchoolAnn ArborMichigan
| | - Arul M. Chinnaiyan
- Rogel Cancer CenterUniversity of Michigan Medical SchoolAnn ArborMichigan
- Department of PathologyUniversity of Michigan Medical SchoolAnn ArborMichigan
- Michigan Center for Translational PathologyUniversity of Michigan Medical SchoolAnn ArborMichigan
- Department of UrologyUniversity of Michigan Medical SchoolAnn ArborMichigan
- Howard Hughes Medical InstituteUniversity of Michigan Medical SchoolAnn ArborMichigan
| |
Collapse
|
25
|
Abstract
INTRODUCTION Cancer is often diagnosed at late stages when the chance of cure is relatively low and although research initiatives in oncology discover many potential cancer biomarkers, few transition to clinical applications. This review addresses the current landscape of cancer biomarker discovery and translation with a focus on proteomics and beyond. Areas covered: The review examines proteomic and genomic techniques for cancer biomarker detection and outlines advantages and challenges of integrating multiple omics approaches to achieve optimal sensitivity and address tumor heterogeneity. This discussion is based on a systematic literature review and direct participation in translational studies. Expert commentary: Identifying aggressive cancers early on requires improved sensitivity and implementation of biomarkers representative of tumor heterogeneity. During the last decade of genomic and proteomic research, significant advancements have been made in next generation sequencing and mass spectrometry techniques. This in turn has led to a dramatic increase in identification of potential genomic and proteomic cancer biomarkers. However, limited successes have been shown with translation of these discoveries into clinical practice. We believe that the integration of these omics approaches is the most promising molecular tool for comprehensive cancer evaluation, early detection and transition to Precision Medicine in oncology.
Collapse
Affiliation(s)
- Ventzislava A Hristova
- a Department of Pathology , Johns Hopkins University School of Medicine , Baltimore , MD , USA
| | - Daniel W Chan
- a Department of Pathology , Johns Hopkins University School of Medicine , Baltimore , MD , USA
| |
Collapse
|
26
|
Zhang Z, Chen YC, Urs S, Chen L, Simeone DM, Yoon E. Scalable Multiplexed Drug-Combination Screening Platforms Using 3D Microtumor Model for Precision Medicine. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2018; 14:e1703617. [PMID: 30239130 DOI: 10.1002/smll.201703617] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Revised: 04/12/2018] [Indexed: 05/15/2023]
Abstract
Cancer heterogeneity is a notorious hallmark of this disease, and it is desirable to tailor effective treatments for each individual patient. Drug combinations have been widely accepted in cancer treatment for better therapeutic efficacy as compared to a single compound. However, experimental complexity and cost grow exponentially with more target compounds under investigation. The primary challenge remains to efficiently perform a large-scale drug combination screening using a small number of patient primary samples for testing. Here, a scalable, easy-to-use, high-throughput drug combination screening scheme is reported, which has the potential of screening all possible pairwise drug combinations for arbitrary number of drugs with multiple logarithmic mixing ratios. A "Christmas tree mixer" structure is introduced to generate a logarithmic concentration mixing ratio between drug pairs, providing a large drug concentration range for screening. A three-layer structure design and special inlets arrangement facilitate simple drug loading process. As a proof of concept, an 8-drug combination chip is implemented, which is capable of screening 172 different treatment conditions over 1032 3D cancer spheroids on a single chip. Using both cancer cell lines and patient-derived cancer cells, effective drug combination screening is demonstrated for precision medicine.
Collapse
Affiliation(s)
- Zhixiong Zhang
- Department of Electrical Engineering and Computer Science, University of Michigan, 1301 Beal Avenue, Ann Arbor, MI, 48109-2122, USA
| | - Yu-Chih Chen
- Department of Electrical Engineering and Computer Science, University of Michigan, 1301 Beal Avenue, Ann Arbor, MI, 48109-2122, USA
- University of Michigan Comprehensive Cancer Center, 1500 E. Medical Center Drive, Ann Arbor, MI, 48109, USA
| | - Sumithra Urs
- University of Michigan Health System, Ann Arbor, MI, 48109, USA
| | - Lili Chen
- Department of Electrical Engineering and Computer Science, University of Michigan, 1301 Beal Avenue, Ann Arbor, MI, 48109-2122, USA
| | - Diane M Simeone
- University of Michigan Health System, Ann Arbor, MI, 48109, USA
| | - Euisik Yoon
- Department of Electrical Engineering and Computer Science, University of Michigan, 1301 Beal Avenue, Ann Arbor, MI, 48109-2122, USA
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| |
Collapse
|
27
|
Lee RC, Thapa B, John T. LACES and bootstraps: the hunt for prognostic and predictive markers for adjuvant therapy in NSCLC. Transl Lung Cancer Res 2018; 7:S239-S242. [PMID: 30393612 PMCID: PMC6193923 DOI: 10.21037/tlcr.2018.09.01] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Accepted: 09/03/2018] [Indexed: 12/29/2022]
Affiliation(s)
- Rachael Chang Lee
- Olivia Newton-John Cancer Centre, Austin Health, Melbourne, Australia
| | - Bibhusal Thapa
- Olivia Newton-John Cancer Centre, Austin Health, Melbourne, Australia
| | - Thomas John
- Olivia Newton-John Cancer Centre, Austin Health, Melbourne, Australia
| |
Collapse
|
28
|
Genetic alterations crossing the borders of distinct hematopoetic lineages and solid tumors: Diagnostic challenges in the era of high-throughput sequencing in hemato-oncology. Crit Rev Oncol Hematol 2018; 126:64-79. [DOI: 10.1016/j.critrevonc.2018.03.020] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Revised: 03/03/2018] [Accepted: 03/25/2018] [Indexed: 02/07/2023] Open
|
29
|
Doostparast Torshizi A, Wang K. Next-generation sequencing in drug development: target identification and genetically stratified clinical trials. Drug Discov Today 2018; 23:1776-1783. [PMID: 29758342 DOI: 10.1016/j.drudis.2018.05.015] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2018] [Revised: 04/09/2018] [Accepted: 05/09/2018] [Indexed: 12/13/2022]
Abstract
Next-generation sequencing (NGS) enabled high-throughput analysis of genotype-phenotype relationships on human populations, ushering in a new era of genetics-informed drug development. The year 2017 was remarkable, with the first FDA-approved gene therapy for cancer (Kymriah™) and for inherited diseases (LUXTURNA™), the first multiplex NGS panel for companion diagnostics (MSK-IMPACT™) and the first drug targeting a genetic signature rather than a disease (Keytruda®). We envision that population-scale NGS with paired electronic health records (EHRs) will become a routine measure in the drug development process for the identification of novel drug targets, and that genetically stratified clinical trials could be widely adopted to improve power in precision-medicine-guided drug development.
Collapse
Affiliation(s)
- Abolfazl Doostparast Torshizi
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Kai Wang
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Department of Biomedical and Health Informatics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
30
|
Ohgami RS, Rosenwald A, Bagg A. Next-Generation Sequencing for Lymphomas: Perfecting a Pipeline for Personalized Pathobiologic and Prognostic Predictions. J Mol Diagn 2018; 20:163-165. [PMID: 29355824 DOI: 10.1016/j.jmoldx.2018.01.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Revised: 01/01/2018] [Accepted: 01/04/2018] [Indexed: 12/28/2022] Open
Abstract
This commentary highlights the article by Hung et al that details the design and implementation of a 32-gene next-generation sequencing panel for lymphomas and compares hybrid-capture with amplicon-based next-generation sequencing approaches.
Collapse
Affiliation(s)
- Robert S Ohgami
- Department of Pathology, Stanford University, Stanford, California.
| | | | - Adam Bagg
- Department of Pathology, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
31
|
Added Value of Whole-Exome and Transcriptome Sequencing for Clinical Molecular Screenings of Advanced Cancer Patients With Solid Tumors. Cancer J 2018; 24:153-162. [DOI: 10.1097/ppo.0000000000000322] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
32
|
Wadapurkar RM, Vyas R. Computational analysis of next generation sequencing data and its applications in clinical oncology. INFORMATICS IN MEDICINE UNLOCKED 2018. [DOI: 10.1016/j.imu.2018.05.003] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
|
33
|
Evolving Significance and Future Relevance of Anti-Angiogenic Activity of mTOR Inhibitors in Cancer Therapy. Cancers (Basel) 2017; 9:cancers9110152. [PMID: 29104248 PMCID: PMC5704170 DOI: 10.3390/cancers9110152] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Revised: 10/23/2017] [Accepted: 10/27/2017] [Indexed: 12/12/2022] Open
Abstract
mTOR inhibitors have demonstrated remarkable anti-tumor activity in experimental models, mainly by reducing cancer cell growth and tumor angiogenesis. Their use in cancer patients as monotherapy has, however, generated only limited benefits, increasing median overall survival by only a few months. Likewise, in other targeted therapies, cancer cells develop resistance mechanisms to overcome mTOR inhibition. Hence, novel therapeutic strategies have to be designed to increase the efficacy of mTOR inhibitors in cancer. In this review, we discuss the present and future relevance of mTOR inhibitors in cancer therapy by focusing on their effects on tumor angiogenesis.
Collapse
|
34
|
Scheerens H, Malong A, Bassett K, Boyd Z, Gupta V, Harris J, Mesick C, Simnett S, Stevens H, Gilbert H, Risser P, Kalamegham R, Jordan J, Engel J, Chen S, Essioux L, Williams JA. Current Status of Companion and Complementary Diagnostics: Strategic Considerations for Development and Launch. Clin Transl Sci 2017; 10:84-92. [PMID: 28121072 PMCID: PMC5355969 DOI: 10.1111/cts.12455] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Accepted: 01/18/2017] [Indexed: 02/06/2023] Open
Abstract
US Food and Drug Administration (FDA)‐approved diagnostic assays play an increasingly common role in managing patients to prolong lifespan while also enhancing quality of life. Diagnostic assays can be essential for the safe and effective use of therapeutics (companion diagnostic), or may inform on improving the benefit/risk ratio without restricting drug access (complementary diagnostic). This tutorial reviews strategic considerations for drug and assay development resulting in FDA‐approved companion or complementary diagnostic status.
Collapse
Affiliation(s)
- H Scheerens
- Genentech Inc., San Francisco, California, USA
| | - A Malong
- Genentech Inc., San Francisco, California, USA
| | - K Bassett
- Genentech Inc., San Francisco, California, USA
| | - Z Boyd
- Genentech Inc., San Francisco, California, USA
| | - V Gupta
- Genentech Inc., San Francisco, California, USA
| | - J Harris
- Genentech Inc., San Francisco, California, USA
| | - C Mesick
- Genentech Inc., San Francisco, California, USA
| | - S Simnett
- Genentech Inc., San Francisco, California, USA
| | | | - H Gilbert
- Genentech Inc., San Francisco, California, USA
| | - P Risser
- Genentech Inc., San Francisco, California, USA
| | | | | | - J Engel
- Roche AG, Basel, Switzerland
| | - S Chen
- Roche AG, Basel, Switzerland
| | - L Essioux
- Pharmaceutical Sciences, Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche, Ltd, Basel, Switzerland
| | | |
Collapse
|