1
|
Schilling L, Kaden J, Bán I, Berger-Höger B. Development of a generic decision guide for patients in oncology: a qualitative interview study. BMC Med Inform Decis Mak 2025; 25:125. [PMID: 40065302 PMCID: PMC11895154 DOI: 10.1186/s12911-025-02960-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Accepted: 03/03/2025] [Indexed: 03/14/2025] Open
Abstract
BACKGROUND Many patients with cancer want to be involved in healthcare decisions. For adequate participation, awareness of one's own desires and preferences and sufficient knowledge about medical measures are indispensable. In order to support patient participation, a decision guide for patients with cancer was developed as part of a larger project called TARGET, which specifically aims to improve the care of patients with rare cancer. METHODS The development of the decision guide took place from 08.2022 to 03.2023. The decision guide is a single component of a complex intervention that aims to facilitate decision support in cancer care for patients. For the development, existing development and evaluation studies of Question Prompt Lists (QPLs) were identified through systematic literature searches in the MEDLINE via PubMed, PsycInfo, and CINAHL databases. The decision guide was pre-tested for feasibility, usability, completeness and acceptance with the target groups through guided individual interviews. Sociodemographic data were collected anonymously. An expert review was conducted. The verbatim transcribed interviews were analysed using content analysis according to Kuckartz with MAXQDA. The guide has been iteratively optimized based on the results. RESULTS A generic decision guide for patients with cancer for diagnostic or treatment decisions was developed in both PDF web-based formats, based on the Ottawa Personal Decision Guide. It was supplemented with decision-related questions from QPLs for patients with cancer. The pre-test comprised seven expert reviews of (psych)oncologists and experts in evidence-based health information and ten interviews with cancer patients (n = 7), family relatives (n = 2), and one caregiver. The results were coded into nine main categories. The results indicated a good feasibility, usability and acceptability of the guide. The tool was perceived as comprehensive and appropriate. Individual elements were identified as modifiable for better comprehensibility. The target audience appreciated the decision guide as a good support option. CONCLUSION The decision guide is potentially a useful support option for patients with cancer facing medical decisions in their further course of treatment. In the TARGET project, it will be made available to patients and can be supplemented with decision coaching. Further steps for implementation into healthcare structures are necessary. CLINICAL TRIAL NUMBER Not applicable.
Collapse
Affiliation(s)
- Lia Schilling
- Institute for Public Health and Nursing Research, University of Bremen, Bremen, Germany.
| | - Jana Kaden
- Institute for Public Health and Nursing Research, University of Bremen, Bremen, Germany
| | - Isabel Bán
- Institute for Public Health and Nursing Research, University of Bremen, Bremen, Germany
| | - Birte Berger-Höger
- Institute for Public Health and Nursing Research, University of Bremen, Bremen, Germany
| |
Collapse
|
2
|
Alexandrova A, Kontareva E, Pustovalova M, Leonov S, Merkher Y. Navigating the Collective: Nanoparticle-Assisted Identification of Leader Cancer Cells During Migration. Life (Basel) 2025; 15:127. [PMID: 39860067 PMCID: PMC11766853 DOI: 10.3390/life15010127] [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: 11/27/2024] [Revised: 01/11/2025] [Accepted: 01/15/2025] [Indexed: 01/27/2025] Open
Abstract
Cancer-related deaths primarily occur due to metastasis, a process involving the migration and invasion of cancer cells. In most solid tumors, metastasis occurs through collective cell migration (CCM), guided by "cellular leaders". These leader cells generate forces through actomyosin-mediated protrusion and contractility. The cytoskeletal mechanisms employed by metastatic cells during the migration process closely resemble the use of the actin cytoskeleton in endocytosis. In our previous work, we revealed that tumor cells exhibiting high metastatic potential (MP) are more adept at encapsulating 100-200 nm nanoparticles than those with lower MP. The objective of this study was to investigate whether nanoparticle encapsulation could effectively differentiate leader tumor cells during their CCM. To achieve our objectives, we employed a two-dimensional CCM model grounded in the wound-healing ("scratch") assay, utilizing two breast cancer cell lines, MCF7 and MDA-MB-231, which display low and high migratory potential, respectively. We conducted calibration experiments to identify the "optimal time" at which cells exhibit peak speed during wound closure. Furthermore, we carried out experiments to assess nanoparticle uptake, calculating the colocalization coefficient, and employed phalloidin staining to analyze the anisotropy and orientation of actin filaments. The highest activity for low-MP cells was achieved at 2.6 h during the calibration experiments, whereas high-MP cells were maximally active at 3.9 h, resulting in 8% and 11% reductions in wound area, respectively. We observed a significant difference in encapsulation efficiency between leader and peripheral cells for both high-MP (p < 0.013) and low-MP (p < 0.02) cells. Moreover, leader cells demonstrated a considerably higher anisotropy coefficient (p < 0.029), indicating a more organized, directional structure of actin filaments compared to peripheral cells. Thus, nanoparticle encapsulation offers a groundbreaking approach to identifying the most aggressive and invasive leader cells during the CCM process in breast cancer. Detecting these cells is crucial for developing targeted therapies that can effectively curb metastasis and improve patient outcomes.
Collapse
Affiliation(s)
- Anastasia Alexandrova
- The Laboratory of Personalized Chemo-Radiation Therapy, Institute of Future Biophysics, Moscow 141700, Russia; (A.A.); (S.L.)
| | - Elizaveta Kontareva
- The Laboratory of Personalized Chemo-Radiation Therapy, Institute of Future Biophysics, Moscow 141700, Russia; (A.A.); (S.L.)
| | - Margarita Pustovalova
- The Laboratory of Personalized Chemo-Radiation Therapy, Institute of Future Biophysics, Moscow 141700, Russia; (A.A.); (S.L.)
| | - Sergey Leonov
- The Laboratory of Personalized Chemo-Radiation Therapy, Institute of Future Biophysics, Moscow 141700, Russia; (A.A.); (S.L.)
- Institute of Cell Biophysics of Russian Academy of Sciences, Pushchino 142290, Russia
| | - Yulia Merkher
- The Laboratory of Personalized Chemo-Radiation Therapy, Institute of Future Biophysics, Moscow 141700, Russia; (A.A.); (S.L.)
- Faculty of Biomedical Engineering, Technion—Israel Institute of Technology, Haifa 3200003, Israel
| |
Collapse
|
3
|
Silva PJ, Rahimzadeh V, Powell R, Husain J, Grossman S, Hansen A, Hinkel J, Rosengarten R, Ory MG, Ramos KS. Health equity innovation in precision medicine: data stewardship and agency to expand representation in clinicogenomics. Health Res Policy Syst 2024; 22:170. [PMID: 39695714 DOI: 10.1186/s12961-024-01258-9] [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: 06/13/2024] [Accepted: 11/22/2024] [Indexed: 12/20/2024] Open
Abstract
Most forms of clinical research examine a very minute cross section of the patient journey. Much of the knowledge and evidence base driving current genomic medicine practice entails blind spots arising from underrepresentation and lack of research participation in clinicogenomic databases. The flaws are perpetuated in AI models and clinical practice guidelines that reflect the lack of diversity in data being used. Participation in clinical research and biobanks is impeded in many populations due to a variety of factors that include knowledge, trust, healthcare access, administrative barriers, and technology gaps. A recent symposium brought industry, clinical, and research participants in clinicogenomics to discuss practical challenges and potential for new data sharing models that are patient centric and federated in nature and can address health disparities that might be perpetuated by lack of diversity in clinicogenomic research, biobanks, and datasets. Clinical data governance was recognized as a multiagent problem, and governance practices need to be more patient centric to address most barriers. Digital tools that preserve privacy, document provenance, and enable the management of data as intellectual property have great promise. Policy updates realigning and rationalizing clinical data governance practices are warranted.
Collapse
Affiliation(s)
- Patrick J Silva
- Texas A&M Health, School of Medicine, Health Professions Education Building 8447 Riverside Pkwy, Bryan, TX, 77807, United States of America.
- Texas A&M Institute for Bioscience and Technology, 2121 W. Holcombe Blvd, Houston, TX, 77030, United States of America.
| | - Vasiliki Rahimzadeh
- Baylor College of Medicine, 1 Baylor Plz, Houston, TX, 77030, United States of America
| | - Reid Powell
- Texas A&M Health, School of Medicine, Health Professions Education Building 8447 Riverside Pkwy, Bryan, TX, 77807, United States of America
- Texas A&M Institute for Bioscience and Technology, 2121 W. Holcombe Blvd, Houston, TX, 77030, United States of America
| | - Junaid Husain
- Greater Houston Healthconnect, 1200 Binz St Suite 1495, Houston, TX, 77004, United States of America
| | - Scott Grossman
- Merck and Co., 126 East Lincoln Avenue, Rahway, NJ, 07065, United States of America
| | - Adam Hansen
- Geneial, Houston, TX, United States of America
| | - Jennifer Hinkel
- The Data Economics Company, Los Angeles, CA, 90064, United States of America
| | - Rafael Rosengarten
- Genialis, 2726 Bissonnet St Suite 240-374, Houston, TX, 77005, United States of America
- Alliance for Artificial Intelligence in Healthcare, 1340 Smith Ave #400, Baltimore, MD, 21209, United States of America
| | - Marcia G Ory
- Department of Environmental and Occupational Health, Center for Population Health and Aging, Texas A&M University School of Public Health, 212 Adriance Lab Rd, College Station, TX, 77843, United States of America
| | - Kenneth S Ramos
- Texas A&M Health, School of Medicine, Health Professions Education Building 8447 Riverside Pkwy, Bryan, TX, 77807, United States of America.
- Texas A&M Institute for Bioscience and Technology, 2121 W. Holcombe Blvd, Houston, TX, 77030, United States of America.
- Texas A&M System, 301 Tarrow St, College Station, TX, 77840, United States of America.
| |
Collapse
|
4
|
Venetis K, Pescia C, Cursano G, Frascarelli C, Mane E, De Camilli E, Munzone E, Dellapasqua S, Criscitiello C, Curigliano G, Guerini Rocco E, Fusco N. The Evolving Role of Genomic Testing in Early Breast Cancer: Implications for Diagnosis, Prognosis, and Therapy. Int J Mol Sci 2024; 25:5717. [PMID: 38891906 PMCID: PMC11172282 DOI: 10.3390/ijms25115717] [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: 04/15/2024] [Revised: 05/13/2024] [Accepted: 05/17/2024] [Indexed: 06/21/2024] Open
Abstract
Multigene prognostic genomic assays have become indispensable in managing early breast cancer (EBC), offering crucial information for risk stratification and guiding adjuvant treatment strategies in conjunction with traditional clinicopathological parameters. The American Society of Clinical Oncology (ASCO) guidelines endorse these assays, though some clinical contexts still lack definitive recommendations. The dynamic landscape of EBC management demands further refinement and optimization of genomic assays to streamline their incorporation into clinical practice. The breast cancer community is poised at the brink of transformative advances in enhancing the clinical utility of genomic assays, aiming to significantly improve the precision and effectiveness of both diagnosis and treatment for women with EBC. This article methodically examines the testing methodologies, clinical validity and utility, costs, diagnostic frameworks, and methodologies of the established genomic tests, including the Oncotype Dx Breast Recurrence Score®, MammaPrint, Prosigna®, EndoPredict®, and Breast Cancer Index (BCI). Among these tests, Prosigna and EndoPredict® have at present been validated only on a prognostic level, while Oncotype Dx, MammaPrint, and BCI hold both a prognostic and predictive role. Oncologists and pathologists engaged in the management of EBC will find in this review a thorough comparison of available genomic assays, as well as strategies to optimize the utilization of the information derived from them.
Collapse
Affiliation(s)
- Konstantinos Venetis
- Division of Pathology, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy; (K.V.); (C.P.); (G.C.); (C.F.); (E.M.); (E.D.C.); (E.G.R.)
| | - Carlo Pescia
- Division of Pathology, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy; (K.V.); (C.P.); (G.C.); (C.F.); (E.M.); (E.D.C.); (E.G.R.)
- School of Pathology, University of Milan, 20122 Milan, Italy
| | - Giulia Cursano
- Division of Pathology, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy; (K.V.); (C.P.); (G.C.); (C.F.); (E.M.); (E.D.C.); (E.G.R.)
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy; (C.C.); (G.C.)
| | - Chiara Frascarelli
- Division of Pathology, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy; (K.V.); (C.P.); (G.C.); (C.F.); (E.M.); (E.D.C.); (E.G.R.)
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy; (C.C.); (G.C.)
| | - Eltjona Mane
- Division of Pathology, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy; (K.V.); (C.P.); (G.C.); (C.F.); (E.M.); (E.D.C.); (E.G.R.)
| | - Elisa De Camilli
- Division of Pathology, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy; (K.V.); (C.P.); (G.C.); (C.F.); (E.M.); (E.D.C.); (E.G.R.)
| | - Elisabetta Munzone
- Division of Medical Senology, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy; (E.M.); (S.D.)
| | - Silvia Dellapasqua
- Division of Medical Senology, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy; (E.M.); (S.D.)
| | - Carmen Criscitiello
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy; (C.C.); (G.C.)
- Division of New Drugs and Early Drug Development for Innovative Therapies, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy
| | - Giuseppe Curigliano
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy; (C.C.); (G.C.)
- Division of New Drugs and Early Drug Development for Innovative Therapies, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy
| | - Elena Guerini Rocco
- Division of Pathology, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy; (K.V.); (C.P.); (G.C.); (C.F.); (E.M.); (E.D.C.); (E.G.R.)
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy; (C.C.); (G.C.)
| | - Nicola Fusco
- Division of Pathology, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy; (K.V.); (C.P.); (G.C.); (C.F.); (E.M.); (E.D.C.); (E.G.R.)
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy; (C.C.); (G.C.)
| |
Collapse
|
5
|
Hong Z, Xiong J, Yang H, Mo YK. Lightweight Low-Rank Adaptation Vision Transformer Framework for Cervical Cancer Detection and Cervix Type Classification. Bioengineering (Basel) 2024; 11:468. [PMID: 38790335 PMCID: PMC11118906 DOI: 10.3390/bioengineering11050468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2024] [Revised: 05/01/2024] [Accepted: 05/02/2024] [Indexed: 05/26/2024] Open
Abstract
Cervical cancer is a major health concern worldwide, highlighting the urgent need for better early detection methods to improve outcomes for patients. In this study, we present a novel digital pathology classification approach that combines Low-Rank Adaptation (LoRA) with the Vision Transformer (ViT) model. This method is aimed at making cervix type classification more efficient through a deep learning classifier that does not require as much data. The key innovation is the use of LoRA, which allows for the effective training of the model with smaller datasets, making the most of the ability of ViT to represent visual information. This approach performs better than traditional Convolutional Neural Network (CNN) models, including Residual Networks (ResNets), especially when it comes to performance and the ability to generalize in situations where data are limited. Through thorough experiments and analysis on various dataset sizes, we found that our more streamlined classifier is highly accurate in spotting various cervical anomalies across several cases. This work advances the development of sophisticated computer-aided diagnostic systems, facilitating more rapid and accurate detection of cervical cancer, thereby significantly enhancing patient care outcomes.
Collapse
Affiliation(s)
- Zhenchen Hong
- Department of Physics and Astronomy, University of California, Riverside, CA 92521, USA
| | - Jingwei Xiong
- Graduate Group in Biostatistics, University of California, Davis, CA 95616, USA
| | - Han Yang
- Department of Chemistry, Columbia University, New York, NY 10027, USA;
| | - Yu K. Mo
- Department of Computer Science, Indiana University, Bloomington, IN 47405, USA;
- Department of Biology, Indiana University, Bloomington, IN 47405, USA
| |
Collapse
|
6
|
Aisner DL, Gocke CD, Jones D, Limson M, Morrissette J, Segal JP. The Genomics Organization for Academic Laboratories (GOAL): A vision for a genomics future for academic pathology. Acad Pathol 2023; 10:100090. [PMID: 37583476 PMCID: PMC10424130 DOI: 10.1016/j.acpath.2023.100090] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 05/26/2023] [Accepted: 06/18/2023] [Indexed: 08/17/2023] Open
Abstract
Innovative and self-sustaining clinical genomics laboratories specializing in cutting-edge oncology testing are critical to the success of academic pathology departments and resident and fellow education in molecular pathology. However, the pressures and challenges facing these laboratories are numerous, including the complexities of validating comprehensive cancer next-generation sequencing (NGS) panels, competition from commercial laboratories, and the reimbursement and regulatory hurdles inherent in high-complexity testing. Cross-institutional collaborations, including shared assay content and interpretative frameworks, are a valuable element to academic laboratory success. To address these and other needs, the Genomics Organization for Academic Laboratories (GOAL) was conceived in 2018, incorporated in 2020 and has grown to include 29 participating institutions in 2022. Here, we describe the mission of GOAL, its structure, and the outcomes and projects undertaken in its first years.
Collapse
Affiliation(s)
- Dara L. Aisner
- Department of Pathology, University of Colorado, Denver, CO, USA
| | | | - Daniel Jones
- Department of Pathology, The Ohio State University, Columbus, OH, USA
| | - Melvin Limson
- Genomics Organization for Academic Laboratories and the Association of Pathology Chairs, Wilmington, DE, USA
| | - Jennifer Morrissette
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jeremy P. Segal
- Department of Pathology, University of Chicago, Chicago, IL, USA
| |
Collapse
|
7
|
Maloberti T, De Leo A, Coluccelli S, Sanza V, Gruppioni E, Altimari A, Zagnoni S, Giunchi F, Vasuri F, Fiorentino M, Mollica V, Ferrari S, Miccoli S, Visani M, Turchetti D, Massari F, Tallini G, de Biase D. Multi-Gene Next-Generation Sequencing Panel for Analysis of BRCA1/ BRCA2 and Homologous Recombination Repair Genes Alterations Metastatic Castration-Resistant Prostate Cancer. Int J Mol Sci 2023; 24:8940. [PMID: 37240284 PMCID: PMC10219522 DOI: 10.3390/ijms24108940] [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] [Received: 04/15/2023] [Revised: 05/11/2023] [Accepted: 05/16/2023] [Indexed: 05/28/2023] Open
Abstract
Despite significant therapeutic advances, metastatic CRPC (mCRPC) remains a lethal disease. Mutations in homologous recombination repair (HRR) genes are frequent in mCRPC, and tumors harboring these mutations are known to be sensitive to PARP inhibitors. The aim of this study was to verify the technical effectiveness of this panel in the analysis of mCRPC, the frequency and type of mutations in the BRCA1/BRCA2 genes, as well as in the homologous recombination repair (HRR) genes. A total of 50 mCRPC cases were analyzed using a multi-gene next-generation sequencing panel evaluating a total of 1360 amplicons in 24 HRR genes. Of the 50 cases, 23 specimens (46.0%) had an mCRPC harboring a pathogenic variant or a variant of uncertain significance (VUS), whereas in 27 mCRPCs (54.0%), no mutations were detected (wild-type tumors). BRCA2 was the most commonly mutated gene (14.0% of samples), followed by ATM (12.0%), and BRCA1 (6.0%). In conclusion, we have set up an NGS multi-gene panel that is capable of analyzing BRCA1/BRCA2 and HRR alterations in mCRPC. Moreover, our clinical algorithm is currently being used in clinical practice for the management of patients with mCRPC.
Collapse
Affiliation(s)
- Thais Maloberti
- Solid Tumor Molecular Pathology Laboratory, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (T.M.); (A.D.L.); (V.S.); (E.G.); (A.A.); (S.Z.); (G.T.)
| | - Antonio De Leo
- Solid Tumor Molecular Pathology Laboratory, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (T.M.); (A.D.L.); (V.S.); (E.G.); (A.A.); (S.Z.); (G.T.)
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40138 Bologna, Italy; (M.F.); (D.T.)
| | - Sara Coluccelli
- Solid Tumor Molecular Pathology Laboratory, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (T.M.); (A.D.L.); (V.S.); (E.G.); (A.A.); (S.Z.); (G.T.)
| | - Viviana Sanza
- Solid Tumor Molecular Pathology Laboratory, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (T.M.); (A.D.L.); (V.S.); (E.G.); (A.A.); (S.Z.); (G.T.)
| | - Elisa Gruppioni
- Solid Tumor Molecular Pathology Laboratory, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (T.M.); (A.D.L.); (V.S.); (E.G.); (A.A.); (S.Z.); (G.T.)
| | - Annalisa Altimari
- Solid Tumor Molecular Pathology Laboratory, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (T.M.); (A.D.L.); (V.S.); (E.G.); (A.A.); (S.Z.); (G.T.)
| | - Stefano Zagnoni
- Solid Tumor Molecular Pathology Laboratory, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (T.M.); (A.D.L.); (V.S.); (E.G.); (A.A.); (S.Z.); (G.T.)
| | - Francesca Giunchi
- Pathology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (F.G.); (F.V.)
| | - Francesco Vasuri
- Pathology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (F.G.); (F.V.)
| | - Michelangelo Fiorentino
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40138 Bologna, Italy; (M.F.); (D.T.)
- Pathology Unit, Maggiore Hospital, AUSL Bologna, 40133 Bologna, Italy
| | - Veronica Mollica
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy;
| | - Simona Ferrari
- Unit of Medical Genetics, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (S.F.); (S.M.)
| | - Sara Miccoli
- Unit of Medical Genetics, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (S.F.); (S.M.)
| | - Michela Visani
- Department of Pharmacy and Biotechnology, University of Bologna, 40126 Bologna, Italy;
| | - Daniela Turchetti
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40138 Bologna, Italy; (M.F.); (D.T.)
- Unit of Medical Genetics, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (S.F.); (S.M.)
| | - Francesco Massari
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40138 Bologna, Italy; (M.F.); (D.T.)
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy;
| | - Giovanni Tallini
- Solid Tumor Molecular Pathology Laboratory, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (T.M.); (A.D.L.); (V.S.); (E.G.); (A.A.); (S.Z.); (G.T.)
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40138 Bologna, Italy; (M.F.); (D.T.)
| | - Dario de Biase
- Solid Tumor Molecular Pathology Laboratory, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (T.M.); (A.D.L.); (V.S.); (E.G.); (A.A.); (S.Z.); (G.T.)
- Department of Pharmacy and Biotechnology, University of Bologna, 40126 Bologna, Italy;
| |
Collapse
|
8
|
Hollon T, Jiang C, Chowdury A, Nasir-Moin M, Kondepudi A, Aabedi A, Adapa A, Al-Holou W, Heth J, Sagher O, Lowenstein P, Castro M, Wadiura LI, Widhalm G, Movahed-Ezazi M, Neuschmelting V, Reinecke D, von Spreckelsen N, Berger M, Hervey-Jumper S, Golfinos J, Camelo-Piragua S, Freudiger C, Lee H, Orringer D. Artificial-intelligence-based molecular classification of diffuse gliomas using rapid, label-free optical imaging. Nat Med 2023; 29:828-832. [PMID: 36959422 PMCID: PMC10445531 DOI: 10.1038/s41591-023-02252-4] [Citation(s) in RCA: 58] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 02/08/2023] [Indexed: 03/25/2023]
Abstract
Molecular classification has transformed the management of brain tumors by enabling more accurate prognostication and personalized treatment. However, timely molecular diagnostic testing for patients with brain tumors is limited, complicating surgical and adjuvant treatment and obstructing clinical trial enrollment. In this study, we developed DeepGlioma, a rapid (<90 seconds), artificial-intelligence-based diagnostic screening system to streamline the molecular diagnosis of diffuse gliomas. DeepGlioma is trained using a multimodal dataset that includes stimulated Raman histology (SRH); a rapid, label-free, non-consumptive, optical imaging method; and large-scale, public genomic data. In a prospective, multicenter, international testing cohort of patients with diffuse glioma (n = 153) who underwent real-time SRH imaging, we demonstrate that DeepGlioma can predict the molecular alterations used by the World Health Organization to define the adult-type diffuse glioma taxonomy (IDH mutation, 1p19q co-deletion and ATRX mutation), achieving a mean molecular classification accuracy of 93.3 ± 1.6%. Our results represent how artificial intelligence and optical histology can be used to provide a rapid and scalable adjunct to wet lab methods for the molecular screening of patients with diffuse glioma.
Collapse
Affiliation(s)
- Todd Hollon
- Machine Learning in Neurosurgery Laboratory, Department of Neurosurgery, University of Michigan, 1500 E. Medical Center Dr., Ann Arbor, 48105, MI, USA
| | - Cheng Jiang
- Machine Learning in Neurosurgery Laboratory, Department of Neurosurgery, University of Michigan, 1500 E. Medical Center Dr., Ann Arbor, 48105, MI, USA
| | - Asadur Chowdury
- Machine Learning in Neurosurgery Laboratory, Department of Neurosurgery, University of Michigan, 1500 E. Medical Center Dr., Ann Arbor, 48105, MI, USA
| | - Mustafa Nasir-Moin
- Department of Neurosurgery, New York University, Street, City, 10587, State, Country
| | - Akhil Kondepudi
- Machine Learning in Neurosurgery Laboratory, Department of Neurosurgery, University of Michigan, 1500 E. Medical Center Dr., Ann Arbor, 48105, MI, USA
| | - Alexander Aabedi
- Machine Learning in Neurosurgery Laboratory, Department of Neurosurgery, University of Michigan, 1500 E. Medical Center Dr., Ann Arbor, 48105, MI, USA
| | - Arjun Adapa
- Machine Learning in Neurosurgery Laboratory, Department of Neurosurgery, University of Michigan, 1500 E. Medical Center Dr., Ann Arbor, 48105, MI, USA
| | - Wajd Al-Holou
- Machine Learning in Neurosurgery Laboratory, Department of Neurosurgery, University of Michigan, 1500 E. Medical Center Dr., Ann Arbor, 48105, MI, USA
| | - Jason Heth
- Machine Learning in Neurosurgery Laboratory, Department of Neurosurgery, University of Michigan, 1500 E. Medical Center Dr., Ann Arbor, 48105, MI, USA
| | - Oren Sagher
- Machine Learning in Neurosurgery Laboratory, Department of Neurosurgery, University of Michigan, 1500 E. Medical Center Dr., Ann Arbor, 48105, MI, USA
| | - Pedro Lowenstein
- Machine Learning in Neurosurgery Laboratory, Department of Neurosurgery, University of Michigan, 1500 E. Medical Center Dr., Ann Arbor, 48105, MI, USA
| | - Maria Castro
- Machine Learning in Neurosurgery Laboratory, Department of Neurosurgery, University of Michigan, 1500 E. Medical Center Dr., Ann Arbor, 48105, MI, USA
| | - Lisa Irina Wadiura
- Machine Learning in Neurosurgery Laboratory, Department of Neurosurgery, University of Michigan, 1500 E. Medical Center Dr., Ann Arbor, 48105, MI, USA
| | - Georg Widhalm
- Machine Learning in Neurosurgery Laboratory, Department of Neurosurgery, University of Michigan, 1500 E. Medical Center Dr., Ann Arbor, 48105, MI, USA
| | - Misha Movahed-Ezazi
- Machine Learning in Neurosurgery Laboratory, Department of Neurosurgery, University of Michigan, 1500 E. Medical Center Dr., Ann Arbor, 48105, MI, USA
| | - Volker Neuschmelting
- Machine Learning in Neurosurgery Laboratory, Department of Neurosurgery, University of Michigan, 1500 E. Medical Center Dr., Ann Arbor, 48105, MI, USA
| | - David Reinecke
- Machine Learning in Neurosurgery Laboratory, Department of Neurosurgery, University of Michigan, 1500 E. Medical Center Dr., Ann Arbor, 48105, MI, USA
| | - Niklas von Spreckelsen
- Machine Learning in Neurosurgery Laboratory, Department of Neurosurgery, University of Michigan, 1500 E. Medical Center Dr., Ann Arbor, 48105, MI, USA
| | - Mitchell Berger
- Machine Learning in Neurosurgery Laboratory, Department of Neurosurgery, University of Michigan, 1500 E. Medical Center Dr., Ann Arbor, 48105, MI, USA
| | - Shawn Hervey-Jumper
- Machine Learning in Neurosurgery Laboratory, Department of Neurosurgery, University of Michigan, 1500 E. Medical Center Dr., Ann Arbor, 48105, MI, USA
| | - John Golfinos
- Machine Learning in Neurosurgery Laboratory, Department of Neurosurgery, University of Michigan, 1500 E. Medical Center Dr., Ann Arbor, 48105, MI, USA
| | - Sandra Camelo-Piragua
- Machine Learning in Neurosurgery Laboratory, Department of Neurosurgery, University of Michigan, 1500 E. Medical Center Dr., Ann Arbor, 48105, MI, USA
| | - Christian Freudiger
- Machine Learning in Neurosurgery Laboratory, Department of Neurosurgery, University of Michigan, 1500 E. Medical Center Dr., Ann Arbor, 48105, MI, USA
| | - Honglak Lee
- Machine Learning in Neurosurgery Laboratory, Department of Neurosurgery, University of Michigan, 1500 E. Medical Center Dr., Ann Arbor, 48105, MI, USA
| | - Daniel Orringer
- Department of Neurosurgery, New York University, Street, City, 10587, State, Country
| |
Collapse
|
9
|
Heuvelings DJI, Wintjens AGWE, Luyten J, Wilmink GEWA, Moonen L, Speel EJM, de Hingh IHJT, Bouvy ND, Peeters A. DNA and RNA Alterations Associated with Colorectal Peritoneal Metastases: A Systematic Review. Cancers (Basel) 2023; 15:cancers15020549. [PMID: 36672497 PMCID: PMC9856984 DOI: 10.3390/cancers15020549] [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: 12/12/2022] [Revised: 01/04/2023] [Accepted: 01/08/2023] [Indexed: 01/18/2023] Open
Abstract
BACKGROUND As colorectal cancer (CRC) patients with peritoneal metastases (PM) have a poor prognosis, new treatment options are currently being investigated for CRC patients. Specific biomarkers in the primary tumor could serve as a prediction tool to estimate the risk of distant metastatic spread. This would help identify patients eligible for early treatment. AIM To give an overview of previously studied DNA and RNA alterations in the primary tumor correlated to colorectal PM and investigate which gene mutations should be further studied. METHODS A systematic review of all published studies reporting genomic analyses on the primary tissue of CRC tumors in relation to PM was undertaken according to PRISMA guidelines. RESULTS Overall, 32 studies with 18,906 patients were included. BRAF mutations were analyzed in 17 articles, of which 10 found a significant association with PM. For all other reported genes, no association with PM was found. Two analyses with broader cancer panels did not reveal any new biomarkers. CONCLUSION An association of specific biomarkers in the primary tumors of CRC patients with metastatic spread into peritoneum could not be proven. The role of BRAF mutations should be further investigated. In addition, studies searching for potential novel biomarkers are still required.
Collapse
Affiliation(s)
- Danique J. I. Heuvelings
- NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, 6229 ER Maastricht, The Netherlands
- Correspondence:
| | - Anne G. W. E. Wintjens
- NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Julien Luyten
- Department of General Surgery, Maastricht University Medical Center (MUMC+), 6202 AZ Maastricht, The Netherlands
| | - Guus E. W. A. Wilmink
- Department of General Surgery, Maastricht University Medical Center (MUMC+), 6202 AZ Maastricht, The Netherlands
- Faculty of Science and Engineering, Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Laura Moonen
- Department of Pathology, Maastricht University Medical Center (MUMC+), 6202 AZ Maastricht, The Netherlands
- GROW–School for Oncology and Reproduction, Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Ernst-Jan M. Speel
- Department of Pathology, Maastricht University Medical Center (MUMC+), 6202 AZ Maastricht, The Netherlands
- GROW–School for Oncology and Reproduction, Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Ignace H. J. T. de Hingh
- GROW–School for Oncology and Reproduction, Maastricht University, 6229 ER Maastricht, The Netherlands
- Department of General Surgery, Catharina Ziekenhuis, 5623 EJ Eindhoven, The Netherlands
| | - Nicole D. Bouvy
- Department of General Surgery, Maastricht University Medical Center (MUMC+), 6202 AZ Maastricht, The Netherlands
- GROW–School for Oncology and Reproduction, Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Andrea Peeters
- Department of Clinical Epidemiology and Medical Technology Assessment, Maastricht University Medical Centre (MUMC+), 6202 AZ Maastricht, The Netherlands
| |
Collapse
|
10
|
Tarawneh TS, Rodepeter FR, Teply-Szymanski J, Ross P, Koch V, Thölken C, Schäfer JA, Gremke N, Mack HID, Gold J, Riera-Knorrenschild J, Wilhelm C, Rinke A, Middeke M, Klemmer A, Romey M, Hattesohl A, Jesinghaus M, Görg C, Figiel J, Chung HR, Wündisch T, Neubauer A, Denkert C, Mack EKM. Combined Focused Next-Generation Sequencing Assays to Guide Precision Oncology in Solid Tumors: A Retrospective Analysis from an Institutional Molecular Tumor Board. Cancers (Basel) 2022; 14:4430. [PMID: 36139590 PMCID: PMC9496918 DOI: 10.3390/cancers14184430] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 09/06/2022] [Accepted: 09/08/2022] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND Increasing knowledge of cancer biology and an expanding spectrum of molecularly targeted therapies provide the basis for precision oncology. Despite extensive gene diagnostics, previous reports indicate that less than 10% of patients benefit from this concept. METHODS We retrospectively analyzed all patients referred to our center's Molecular Tumor Board (MTB) from 2018 to 2021. Molecular testing by next-generation sequencing (NGS) included a 67-gene panel for the detection of short-sequence variants and copy-number alterations, a 53- or 137-gene fusion panel and an ultra-low-coverage whole-genome sequencing for the detection of additional copy-number alterations outside the panel's target regions. Immunohistochemistry for microsatellite instability and PD-L1 expression complemented NGS. RESULTS A total of 109 patients were referred to the MTB. In all, 78 patients received therapeutic proposals (70 based on NGS) and 33 were treated accordingly. Evaluable patients treated with MTB-recommended therapy (n = 30) had significantly longer progression-free survival than patients treated with other therapies (n = 17) (4.3 vs. 1.9 months, p = 0.0094). Seven patients treated with off-label regimens experienced major clinical benefits. CONCLUSION The combined focused sequencing assays detected targetable alterations in the majority of patients. Patient benefits appeared to lie in the same range as with large-scale sequencing approaches.
Collapse
Affiliation(s)
- Thomas S. Tarawneh
- Department of Hematology, Oncology and Immunology, Philipps-University Marburg, Baldingerstraße, 35043 Marburg, Germany
| | - Fiona R. Rodepeter
- Institute of Pathology, Philipps-University Marburg, Baldingerstraße, 35043 Marburg, Germany
| | - Julia Teply-Szymanski
- Institute of Pathology, Philipps-University Marburg, Baldingerstraße, 35043 Marburg, Germany
| | - Petra Ross
- Department of Hematology, Oncology and Immunology, Philipps-University Marburg, Baldingerstraße, 35043 Marburg, Germany
| | - Vera Koch
- Department of Hematology, Oncology and Immunology, Philipps-University Marburg, Baldingerstraße, 35043 Marburg, Germany
- Institute of Medical Bioinformatics and Biostatistics, Philipps-University Marburg, Hans-Meerwein-Straße 6, 35032 Marburg, Germany
| | - Clemens Thölken
- Institute of Medical Bioinformatics and Biostatistics, Philipps-University Marburg, Hans-Meerwein-Straße 6, 35032 Marburg, Germany
| | - Jonas A. Schäfer
- Department of Hematology, Oncology and Immunology, Philipps-University Marburg, Baldingerstraße, 35043 Marburg, Germany
| | - Niklas Gremke
- Department of Gynecology, Gynecologic Endocrinology and Oncology, Philipps-University Marburg, Baldingerstraße, 35043 Marburg, Germany
| | - Hildegard I. D. Mack
- Department of Hematology, Oncology and Immunology, Philipps-University Marburg, Baldingerstraße, 35043 Marburg, Germany
| | - Judith Gold
- Department of Hematology, Oncology and Immunology, Philipps-University Marburg, Baldingerstraße, 35043 Marburg, Germany
| | - Jorge Riera-Knorrenschild
- Department of Hematology, Oncology and Immunology, Philipps-University Marburg, Baldingerstraße, 35043 Marburg, Germany
| | - Christian Wilhelm
- Department of Hematology, Oncology and Immunology, Philipps-University Marburg, Baldingerstraße, 35043 Marburg, Germany
| | - Anja Rinke
- Department of Gastroenterology and Endocrinology, Philipps-University Marburg, Baldingerstraße, 35043 Marburg, Germany
| | - Martin Middeke
- Comprehensive Cancer Center Marburg, Philipps-University Marburg, Baldingerstraße, 35043 Marburg, Germany
| | - Andreas Klemmer
- Department of Pulmonary and Critical Care Medicine, Philipps-University Marburg, Baldingerstraße, 35043 Marburg, Germany
| | - Marcel Romey
- Institute of Pathology, Philipps-University Marburg, Baldingerstraße, 35043 Marburg, Germany
| | - Akira Hattesohl
- Institute of Pathology, Philipps-University Marburg, Baldingerstraße, 35043 Marburg, Germany
| | - Moritz Jesinghaus
- Institute of Pathology, Philipps-University Marburg, Baldingerstraße, 35043 Marburg, Germany
| | - Christian Görg
- Department of Hematology, Oncology and Immunology, Philipps-University Marburg, Baldingerstraße, 35043 Marburg, Germany
- Department of Gastroenterology and Endocrinology, Philipps-University Marburg, Baldingerstraße, 35043 Marburg, Germany
| | - Jens Figiel
- Department of Diagnostic and Interventional Radiology, Philipps-University Marburg, Baldingerstraße, 35043 Marburg, Germany
| | - Ho-Ryun Chung
- Institute of Medical Bioinformatics and Biostatistics, Philipps-University Marburg, Hans-Meerwein-Straße 6, 35032 Marburg, Germany
| | - Thomas Wündisch
- Comprehensive Cancer Center Marburg, Philipps-University Marburg, Baldingerstraße, 35043 Marburg, Germany
| | - Andreas Neubauer
- Department of Hematology, Oncology and Immunology, Philipps-University Marburg, Baldingerstraße, 35043 Marburg, Germany
| | - Carsten Denkert
- Institute of Pathology, Philipps-University Marburg, Baldingerstraße, 35043 Marburg, Germany
| | - Elisabeth K. M. Mack
- Department of Hematology, Oncology and Immunology, Philipps-University Marburg, Baldingerstraße, 35043 Marburg, Germany
| |
Collapse
|
11
|
Hamdan A, Ewing A. Unravelling the tumour genome: The evolutionary and clinical impacts of structural variants in tumourigenesis. J Pathol 2022; 257:479-493. [PMID: 35355264 PMCID: PMC9321913 DOI: 10.1002/path.5901] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 03/16/2022] [Accepted: 03/28/2022] [Indexed: 11/15/2022]
Abstract
Structural variants (SVs) represent a major source of aberration in tumour genomes. Given the diversity in the size and type of SVs present in tumours, the accurate detection and interpretation of SVs in tumours is challenging. New classes of complex structural events in tumours are discovered frequently, and the definitions of the genomic consequences of complex events are constantly being refined. Detailed analyses of short-read whole-genome sequencing (WGS) data from large tumour cohorts facilitate the interrogation of SVs at orders of magnitude greater scale and depth. However, the inherent technical limitations of short-read WGS prevent us from accurately detecting and investigating the impact of all the SVs present in tumours. The expanded use of long-read WGS will be critical for improving the accuracy of SV detection, and in fully resolving complex SV events, both of which are crucial for determining the impact of SVs on tumour progression and clinical outcome. Despite the present limitations, we demonstrate that SVs play an important role in tumourigenesis. In particular, SVs contribute significantly to late-stage tumour development and to intratumoural heterogeneity. The evolutionary trajectories of SVs represent a window into the clonal dynamics in tumours, a comprehensive understanding of which will be vital for influencing patient outcomes in the future. Recent findings have highlighted many clinical applications of SVs in cancer, from early detection to biomarkers for treatment response and prognosis. As the methods to detect and interpret SVs improve, elucidating the full breadth of the complex SV landscape and determining how these events modulate tumour evolution will improve our understanding of cancer biology and our ability to capitalise on the utility of SVs in the clinical management of cancer patients. © 2022 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Alhafidz Hamdan
- MRC Human Genetics Unit, Institute of Genetics and CancerUniversity of EdinburghEdinburghUK
- Cancer Research UK Edinburgh Centre, Institute of Genetics and CancerUniversity of EdinburghEdinburghUK
| | - Ailith Ewing
- MRC Human Genetics Unit, Institute of Genetics and CancerUniversity of EdinburghEdinburghUK
- Cancer Research UK Edinburgh Centre, Institute of Genetics and CancerUniversity of EdinburghEdinburghUK
| |
Collapse
|
12
|
The History and Future of Basic and Translational Cell-Free DNA Research at a Glance. Diagnostics (Basel) 2022; 12:diagnostics12051192. [PMID: 35626347 PMCID: PMC9139999 DOI: 10.3390/diagnostics12051192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 04/29/2022] [Accepted: 05/03/2022] [Indexed: 11/30/2022] Open
Abstract
We discuss the early history of the structure of DNA and its involvement in gene structure as well as its mobility in and between cells and between tissues in the form of circulating cell-free DNA (cfDNA). This is followed by a view of the present status of the studies on cfDNA and clinical applications of circulating cell-free tumor DNA (ctDNA). The future developments and roles of ctDNA are also considered.
Collapse
|
13
|
The Clinical Utility and Impact of Next Generation Sequencing in Gynecologic Cancers. Cancers (Basel) 2022; 14:cancers14051352. [PMID: 35267660 PMCID: PMC8909263 DOI: 10.3390/cancers14051352] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 03/04/2022] [Accepted: 03/04/2022] [Indexed: 11/30/2022] Open
Abstract
Simple Summary Cancer cells harbor many genetic abnormalities, but the key oncogenic pathways that lead to clinically evident cancer require driver mutations termed actionable mutations. These actionable mutations can be detected using genomic profiling or next-generation sequencing tests. This discovery has led to a tremendous change in treatment regimens from standard chemotherapy to targeted therapy where drugs are specifically targeted against these actionable mutations. Due to the cost-effectiveness and various testing platforms, utilization of these tests by oncologists has increased enormously, but the impact of targeted therapy based on these test results is still understudied. We aimed to identify the clinical utility rate of the tests and analyze the survival benefit for those receiving targeted therapy based on the test results of gynecologic cancer patients. Our findings showed high clinical utility of the tests used by gynecologic oncologists along with a significant survival benefit. Abstract Next generation sequencing (NGS) has facilitated the identification of molecularly targeted therapies. However, clinical utility is an emerging challenge. Our objective was to identify the clinical utility of NGS testing in gynecologic cancers. A retrospective review of clinico-pathologic data was performed on 299 gynecological cancers where NGS testing had been performed to identify (1) recognition of actionable targets for therapy, (2) whether the therapy changed based on the findings, and (3) the impact on survival. High grade serous carcinoma was the most common tumor (52.5%). The number of genetic alterations ranged from 0 to 25 with a mean of 2.8/case. The most altered genes were TP53, PIK3CA, BRCA1 and BRCA2. Among 299 patients, 100 had actionable alterations (79 received a targeted treatment (Group1), 29 did not receive treatment (Group 2), and there were no actionable alterations in 199 (Group3). The death rate in groups 1, 2 and 3 was 54.4%, 42.8% and 50.2%, with an average survival of 18.6, 6.6 and 10.8 months, respectively (p = 0.002). In summary, NGS testing for gynecologic cancers detected 33.4% of actionable alterations with a high clinical action rate. Along with the high clinical utility of NGS, testing also seemed to improve survival for patients who received targeted treatment.
Collapse
|
14
|
Wu L, Dias A, Diéguez L. Surface enhanced Raman spectroscopy for tumor nucleic acid: Towards cancer diagnosis and precision medicine. Biosens Bioelectron 2022; 204:114075. [DOI: 10.1016/j.bios.2022.114075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 01/13/2022] [Accepted: 02/02/2022] [Indexed: 11/25/2022]
|
15
|
Patel SB, Bookstein R, Farahani N, Chevarie-Davis M, Pao A, Aguiluz A, Riley C, Hodge JC, Alkan S, Liu Z, Deng N, Lopategui JR. Recommendations for Specimen and Therapy Selection in Colorectal Cancer. Oncol Ther 2021; 9:451-469. [PMID: 33895946 PMCID: PMC8593092 DOI: 10.1007/s40487-021-00151-7] [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] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 03/31/2021] [Indexed: 11/21/2022] Open
Abstract
INTRODUCTION Next-generation sequencing has emerged as a clinical tool for the identification of actionable mutations to triage advanced colorectal cancer patients for targeted therapies. The literature is conflicted as to whether primaries or their metastases should be selected for sequencing. Some authors suggest that either site may be sequenced, whereas others recommend sequencing the primary, the metastasis, or even both tumors. Here, we address this issue head on with a meta-analysis and provide for the first time a set of sensible recommendations to make this determination. METHODS From our own series, we include 43 tumors from 13 patients including 14 primaries, 10 regional lymph node metastases, 17 distant metastases, and two anastomotic recurrences sequenced using the 50 gene Ion AmpliSeq cancer NGS panel v2. RESULTS Based on our new cohort and a meta-analysis, we found that ~ 77% of patient-matched primary-metastatic pairs have identical alterations in these 50 cancer-associated genes. CONCLUSIONS Low tumor cellularity, tumor heterogeneity, clonal evolution, treatment status, sample quality, and/or size of the sequencing panel accounted for a proportion of the differential detection of mutations at primary and metastatic sites. The therapeutic implications of the most frequently discordant alterations (TP53, APC, PIK3CA, and SMAD4) are discussed. Our meta-analysis indicates that a subset of patients who fail initial therapy may benefit from sequencing of additional sites to identify new actionable genomic abnormalities not present in the initial analysis. Evidence-based recommendations are proposed.
Collapse
Affiliation(s)
- Snehal B Patel
- Cedars-Sinai Medical Center, Division of Molecular Pathology and Cytogenetics, Department of Pathology and Laboratory Medicine, 8700 Beverly Blvd., SSB #362, Los Angeles, CA, 90048, USA
- HeloGenika LLC, Dexter, MI, 48130, USA
| | - Robert Bookstein
- Cedars-Sinai Medical Center, Division of Molecular Pathology and Cytogenetics, Department of Pathology and Laboratory Medicine, 8700 Beverly Blvd., SSB #362, Los Angeles, CA, 90048, USA
| | - Navid Farahani
- Cedars-Sinai Medical Center, Division of Molecular Pathology and Cytogenetics, Department of Pathology and Laboratory Medicine, 8700 Beverly Blvd., SSB #362, Los Angeles, CA, 90048, USA
| | - Myriam Chevarie-Davis
- Cedars-Sinai Medical Center, Division of Molecular Pathology and Cytogenetics, Department of Pathology and Laboratory Medicine, 8700 Beverly Blvd., SSB #362, Los Angeles, CA, 90048, USA
| | - Andy Pao
- Cedars-Sinai Medical Center, Division of Molecular Pathology and Cytogenetics, Department of Pathology and Laboratory Medicine, 8700 Beverly Blvd., SSB #362, Los Angeles, CA, 90048, USA
| | - Angela Aguiluz
- Cedars-Sinai Medical Center, Division of Molecular Pathology and Cytogenetics, Department of Pathology and Laboratory Medicine, 8700 Beverly Blvd., SSB #362, Los Angeles, CA, 90048, USA
| | - Christian Riley
- Cedars-Sinai Medical Center, Division of Molecular Pathology and Cytogenetics, Department of Pathology and Laboratory Medicine, 8700 Beverly Blvd., SSB #362, Los Angeles, CA, 90048, USA
| | - Jennelle C Hodge
- Cedars-Sinai Medical Center, Division of Molecular Pathology and Cytogenetics, Department of Pathology and Laboratory Medicine, 8700 Beverly Blvd., SSB #362, Los Angeles, CA, 90048, USA
| | - Serhan Alkan
- Cedars-Sinai Medical Center, Division of Molecular Pathology and Cytogenetics, Department of Pathology and Laboratory Medicine, 8700 Beverly Blvd., SSB #362, Los Angeles, CA, 90048, USA
| | - Zhenqui Liu
- Cedars-Sinai Medical Center, Biostatistics and Bioinformatics Research Center, Samuel Oschin Comprehensive Cancer Institute, Los Angeles, CA, USA
| | - Nan Deng
- Cedars-Sinai Medical Center, Biostatistics and Bioinformatics Research Center, Samuel Oschin Comprehensive Cancer Institute, Los Angeles, CA, USA
| | - Jean R Lopategui
- Cedars-Sinai Medical Center, Division of Molecular Pathology and Cytogenetics, Department of Pathology and Laboratory Medicine, 8700 Beverly Blvd., SSB #362, Los Angeles, CA, 90048, USA.
| |
Collapse
|
16
|
Weymann D, Laskin J, Jones SJM, Roscoe R, Lim HJ, Renouf DJ, Schrader KA, Sun S, Yip S, Marra MA, Regier DA. Early-stage economic analysis of research-based comprehensive genomic sequencing for advanced cancer care. J Community Genet 2021; 13:523-538. [PMID: 34843087 PMCID: PMC8628132 DOI: 10.1007/s12687-021-00557-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Accepted: 10/06/2021] [Indexed: 01/23/2023] Open
Abstract
Genomic research is driving discovery for future population benefit. Limited evidence exists on immediate patient and health system impacts of research participation. This study uses real-world data and quasi-experimental matching to examine early-stage cost and health impacts of research-based genomic sequencing. British Columbia’s Personalized OncoGenomics (POG) single-arm program applies whole genome and transcriptome analysis (WGTA) to characterize genomic landscapes in advanced cancers. Our cohort includes POG patients enrolled between 2014 and 2015 and 1:1 genetic algorithm–matched usual care controls. We undertake a cost consequence analysis and estimate 1-year effects of WGTA on patient management, patient survival, and health system costs reported in 2015 Canadian dollars. WGTA costs are imputed and forecast using system of equations modeling. We use Kaplan-Meier survival analysis to explore survival differences and inverse probability of censoring weighted linear regression to estimate mean 1-year survival times and costs. Non-parametric bootstrapping simulates sampling distributions and enables scenario analysis, revealing drivers of incremental costs, survival, and net monetary benefit for assumed willingness to pay thresholds. We identified 230 POG patients and 230 matched controls for cohort inclusion. The mean period cost of research-funded WGTA was $26,211 (SD: $14,191). Sequencing costs declined rapidly, with WGTA forecasts hitting $13,741 in 2021. The incremental healthcare system effect (non-research expenditures) was $5203 (95% CI: 75, 10,424) compared to usual care. No overall survival differences were observed, but outcome heterogeneity was present. POG patients receiving WGTA-informed treatment experienced incremental survival gains of 2.49 months (95% CI: 1.32, 3.64). Future cost consequences became favorable as WGTA cost drivers declined and WGTA-informed treatment rates improved to 60%. Our study demonstrates the ability of real-world data to support evaluations of only-in-research health technologies. We identify situations where precision oncology research initiatives may produce survival benefit at a cost that is within healthcare systems’ willingness to pay. This economic evidence informs the early-stage healthcare impacts of precision oncology research.
Collapse
Affiliation(s)
- Deirdre Weymann
- Canadian Centre for Applied Research in Cancer Control, Cancer Control Research, BC Cancer Research Centre, 675 West 10th Avenue, Vancouver, BC, V5Z 1L3, Canada
| | - Janessa Laskin
- Division of Medical Oncology, BC Cancer, Vancouver, Canada
- Department of Medicine, Faculty of Medicine, University of British Columbia, Vancouver, Canada
| | - Steven J M Jones
- Canada's Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, Canada
- Department of Medical Genetics, Faculty of Medicine, University of British Columbia, Vancouver, Canada
- Department of Molecular Biology & Biochemistry, Simon Fraser University, Burnaby, Canada
| | - Robyn Roscoe
- Canada's Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, Canada
| | - Howard J Lim
- Division of Medical Oncology, BC Cancer, Vancouver, Canada
- Department of Medicine, Faculty of Medicine, University of British Columbia, Vancouver, Canada
| | - Daniel J Renouf
- Division of Medical Oncology, BC Cancer, Vancouver, Canada
- Department of Medicine, Faculty of Medicine, University of British Columbia, Vancouver, Canada
| | - Kasmintan A Schrader
- Department of Medical Genetics, Faculty of Medicine, University of British Columbia, Vancouver, Canada
- Department of Molecular Oncology, BC Cancer, Vancouver, Canada
| | - Sophie Sun
- Division of Medical Oncology, BC Cancer, Vancouver, Canada
- Department of Medicine, Faculty of Medicine, University of British Columbia, Vancouver, Canada
| | - Stephen Yip
- Department of Pathology & Laboratory Medicine, Faculty of Medicine, University of British Columbia, Vancouver, Canada
- Department of Pathology, BC Cancer, Vancouver, Canada
| | - Marco A Marra
- Canada's Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, Canada
- Department of Medical Genetics, Faculty of Medicine, University of British Columbia, Vancouver, Canada
| | - Dean A Regier
- Canadian Centre for Applied Research in Cancer Control, Cancer Control Research, BC Cancer Research Centre, 675 West 10th Avenue, Vancouver, BC, V5Z 1L3, Canada.
- School of Population and Public Health, University of British Columbia, Vancouver, Canada.
| |
Collapse
|
17
|
Jalloul N, Gomy I, Stokes S, Gusev A, Johnson BE, Lindeman NI, Macconaill L, Ganesan S, Garber JE, Khiabanian H. Germline Testing Data Validate Inferences of Mutational Status for Variants Detected From Tumor-Only Sequencing. JCO Precis Oncol 2021; 5:PO.21.00279. [PMID: 34820595 PMCID: PMC8608266 DOI: 10.1200/po.21.00279] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 09/20/2021] [Accepted: 10/12/2021] [Indexed: 02/06/2023] Open
Abstract
Pathogenic germline variants (PGVs) in cancer susceptibility genes are usually identified through germline testing of DNA from blood or saliva: their detection can affect treatment options and potential risk-reduction strategies for patient relatives. PGV can also be identified in tumor sequencing assays, which, when performed without patient-matched normal specimens, render determination of variants' germline or somatic origin critical. METHODS Tumor-only sequencing data from 1,608 patients were retrospectively analyzed to infer germline versus somatic status of variants using an information-theoretic, gene-independent approach. Loss of heterozygosity was also determined. Predicted mutational models were compared with clinical germline testing results. Statistical measures were computed to evaluate performance. RESULTS Tumor-only sequencing detected 3,988 variants across 70 cancer susceptibility genes for which germline testing data were available. We imputed germline versus somatic status for > 75% of all detected variants, with a sensitivity of 65%, specificity of 88%, and overall accuracy of 86% for pathogenic variants. False omission rate was 3%, signifying minimal error in misclassifying true PGV. A higher portion of PGV in known hereditary tumor suppressors were found to be retained with loss of heterozygosity in the tumor specimens (72%) compared with variants of uncertain significance (58%). CONCLUSION Analyzing tumor-only data in the context of specimens' tumor cell content allows precise, systematic exclusion of somatic variants and suggests a balance between type 1 and 2 errors for identification of patients with candidate PGV for standard germline testing. Although technical or systematic errors in measuring variant allele frequency could result in incorrect inference, misestimation of specimen purity could result in inferring somatic variants as germline in somatically mutated tumor suppressor genes. A user-friendly bioinformatics application facilitates objective analysis of tumor-only data in clinical settings.
Collapse
Affiliation(s)
- Nahed Jalloul
- Rutgers Cancer Institute of New Jersey, Rutgers University, New Brunswick, NJ
| | - Israel Gomy
- Division of Cancer Genetics and Prevention, Dana-Farber Cancer Institute, Boston, MA
| | - Samantha Stokes
- Division of Cancer Genetics and Prevention, Dana-Farber Cancer Institute, Boston, MA
| | - Alexander Gusev
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Broad Institute of MIT and Harvard, Cambridge, MA
| | - Bruce E. Johnson
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Center for Cancer Genome Discovery, Dana-Farber Cancer Institute, Boston, MA
| | - Neal I. Lindeman
- Department of Pathology, Brigham and Women's Hospital, Boston, MA
| | - Laura Macconaill
- Department of Pathology, Brigham and Women's Hospital, Boston, MA
| | - Shridar Ganesan
- Rutgers Cancer Institute of New Jersey, Rutgers University, New Brunswick, NJ
- Department of Medicine, Rutgers Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ
| | - Judy E. Garber
- Division of Cancer Genetics and Prevention, Dana-Farber Cancer Institute, Boston, MA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - Hossein Khiabanian
- Rutgers Cancer Institute of New Jersey, Rutgers University, New Brunswick, NJ
- Department of Pathology and Laboratory Medicine, Rutgers Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ
| |
Collapse
|
18
|
Huang S, Pang L, Wei C. Identification of a Four-Gene Signature With Prognostic Significance in Endometrial Cancer Using Weighted-Gene Correlation Network Analysis. Front Genet 2021; 12:678780. [PMID: 34616422 PMCID: PMC8488359 DOI: 10.3389/fgene.2021.678780] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Accepted: 08/30/2021] [Indexed: 01/01/2023] Open
Abstract
Endometrial hyperplasia (EH) is a precursor for endometrial cancer (EC). However, biomarkers for the progression from EH to EC and standard prognostic biomarkers for EC have not been identified. In this study, we aimed to identify key genes with prognostic significance for the progression from EH to EC. Weighted-gene correlation network analysis (WGCNA) was used to identify hub genes utilizing microarray data (GSE106191) downloaded from the Gene Expression Omnibus database. Differentially expressed genes (DEGs) were identified from the Uterine Corpus Endometrial Carcinoma (UCEC) dataset of The Cancer Genome Atlas database. The Limma-Voom R package was applied to detect differentially expressed genes (DEGs; mRNAs) between cancer and normal samples. Genes with |log2 (fold change [FC])| > 1.0 and p < 0.05 were considered as DEGs. Univariate and multivariate Cox regression and survival analyses were performed to identify potential prognostic genes using hub genes overlapping in the two datasets. All analyses were conducted using R Bioconductor and related packages. Through WGCNA and overlapping genes in hub modules with DEGs in the UCEC dataset, we identified 42 hub genes. The results of the univariate and multivariate Cox regression analyses revealed that four hub genes, BUB1B, NDC80, TPX2, and TTK, were independently associated with the prognosis of EC (Hazard ratio [95% confidence interval]: 0.591 [0.382–0.912], p = 0.017; 0.605 [0.371–0.986], p = 0.044; 1.678 [1.132–2.488], p = 0.01; 2.428 [1.372–4.29], p = 0.02, respectively). A nomogram was established with a risk score calculated using the four genes’ coefficients in the multivariate analysis, and tumor grade and stage had a favorable predictive value for the prognosis of EC. The survival analysis showed that the high-risk group had an unfavorable prognosis compared with the low-risk group (p < 0.0001). The receiver operating characteristic curves also indicated that the risk model had a potential predictive value of prognosis with area under the curve 0.807 at 2 years, 0.783 at 3 years, and 0.786 at 5 years. We established a four-gene signature with prognostic significance in EC using WGCNA and established a nomogram to predict the prognosis of EC.
Collapse
Affiliation(s)
- Shijin Huang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Lihong Pang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Changqiang Wei
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| |
Collapse
|
19
|
Scott RJ, Mehta A, Macedo GS, Borisov PS, Kanesvaran R, El Metnawy W. Genetic testing for homologous recombination repair (HRR) in metastatic castration-resistant prostate cancer (mCRPC): challenges and solutions. Oncotarget 2021; 12:1600-1614. [PMID: 34381565 PMCID: PMC8351605 DOI: 10.18632/oncotarget.28015] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Accepted: 06/14/2021] [Indexed: 12/16/2022] Open
Abstract
Patients with metastatic castration-resistant prostate cancer (mCRPC) have an average survival of only 13 months. Identification of novel predictive and actionable biomarkers in the homologous recombination repair (HRR) pathway in up to a quarter of patients with mCRPC has led to the approval of targeted therapies like poly-ADP ribose polymerase inhibitors (PARPi), with the potential to improve survival outcomes. The approval of PARPi has led to guideline bodies such as the National Comprehensive Cancer Network (NCCN) to actively recommend germline and or somatic HRR gene panel testing to identify patients who will benefit from PARPi. However, there are several challenges as genetic testing is still at an early stage especially in low- and middle-income countries, with cost and availability being major impediments. In addition, there are issues such as choice of optimal tissue for genetic testing, archival, storage, retrieval of tissue blocks, interpretation and classification of variants in the HRR pathway, and the need for pretest and post-test genetic counseling. This review provides insights into the HRR gene mutations prevalent in mCRPC and the challenges for a more widespread gene testing to identify actionable germline pathogenic variants and somatic mutations in the HRR pathway, and proposes a clinical algorithm to enhance the efficiency of the gene testing process.
Collapse
Affiliation(s)
- Rodney J. Scott
- Laureate Professor, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Anurag Mehta
- Director, Department of Laboratory & Transfusion Services and Director Research, Rajiv Gandhi Cancer Institute, Delhi, India
| | - Gabriel S. Macedo
- Programa de Medicina Personalizada – Coordenador, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Pavel S. Borisov
- Oncologist Urologist, FSBI “N.N. Petrov NMRC of Oncology” of the Ministry Healthcare of the Russian Federation, St Petersburg, Russia
| | - Ravindran Kanesvaran
- Deputy Head and Senior Consultant, Division of Medical Oncology, National Cancer Centre Singapore, Singapore
| | - Wafaa El Metnawy
- Professor of Molecular Pathology, Oncology Center School of Medicine, Cairo University, Giza, Egypt
| |
Collapse
|
20
|
Abstract
Technological innovation and rapid reduction in sequencing costs have enabled the genomic profiling of hundreds of cancer-associated genes as a component of routine cancer care. Tumour genomic profiling can refine cancer subtype classification, identify which patients are most likely to benefit from systemic therapies and screen for germline variants that influence heritable cancer risk. Here, we discuss ongoing efforts to enhance the clinical utility of tumour genomic profiling by integrating tumour and germline analyses, characterizing allelic context and identifying mutational signatures that influence therapy response. We also discuss the potential clinical utility of more comprehensive whole-genome and whole-transcriptome sequencing and ultra-sensitive cell-free DNA profiling platforms, which allow for minimally invasive, serial analyses of tumour-derived DNA in blood.
Collapse
Affiliation(s)
- Debyani Chakravarty
- Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA.,Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - David B Solit
- Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA. .,Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
21
|
Li Z, Fang S, Zhang R, Yu L, Zhang J, Bu D, Sun L, Zhao Y, Li J. VarBen. J Mol Diagn 2021; 23:285-299. [DOI: 10.1016/j.jmoldx.2020.11.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 10/06/2020] [Accepted: 11/17/2020] [Indexed: 02/08/2023] Open
|
22
|
Hijikata Y, Yokoyama K, Yokoyama N, Matsubara Y, Shimizu E, Nakashima M, Yamagishi M, Ota Y, Lim LA, Yamaguchi R, Ito M, Tanaka Y, Denda T, Tani K, Yotsuyanagi H, Imoto S, Miyano S, Uchimaru K, Tojo A. Successful Clinical Sequencing by Molecular Tumor Board in an Elderly Patient With Refractory Sézary Syndrome. JCO Precis Oncol 2020; 4:534-560. [DOI: 10.1200/po.19.00254] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Affiliation(s)
- Yasuki Hijikata
- Department of General Medicine, IMSUT Hospital, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Kazuaki Yokoyama
- Department of Hematology/Oncology, IMSUT Hospital, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Nozomi Yokoyama
- Department of Applied Genomics, Research Hospital, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Yasuo Matsubara
- Department of General Medicine, IMSUT Hospital, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Eigo Shimizu
- Laboratory of DNA Information Analysis, Human Genome Center, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Makoto Nakashima
- Laboratory of Tumor Cell Biology, Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Tokyo, Japan
| | - Makoto Yamagishi
- Laboratory of Tumor Cell Biology, Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Tokyo, Japan
| | - Yasunori Ota
- Department of Diagnostic Pathology, IMSUT Hospital, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Lay Ahyoung Lim
- Department of General Medicine, IMSUT Hospital, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Rui Yamaguchi
- Laboratory of DNA Information Analysis, Human Genome Center, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Mika Ito
- Division of Molecular Therapy, Advanced Clinical Research Center, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Yukihisa Tanaka
- Department of Diagnostic Pathology, IMSUT Hospital, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Tamami Denda
- Department of Diagnostic Pathology, IMSUT Hospital, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Kenzaburo Tani
- Department of General Medicine, IMSUT Hospital, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Hiroshi Yotsuyanagi
- Department of General Medicine, IMSUT Hospital, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Seiya Imoto
- Division of Health Medical Data Science, Health Intelligence Center, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Satoru Miyano
- Laboratory of DNA Information Analysis, Human Genome Center, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Kaoru Uchimaru
- Laboratory of Tumor Cell Biology, Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Tokyo, Japan
| | - Arinobu Tojo
- Department of Hematology/Oncology, IMSUT Hospital, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- Division of Health Medical Data Science, Health Intelligence Center, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
23
|
Donoghue MTA, Schram AM, Hyman DM, Taylor BS. Discovery through clinical sequencing in oncology. ACTA ACUST UNITED AC 2020; 1:774-783. [PMID: 35122052 PMCID: PMC8985175 DOI: 10.1038/s43018-020-0100-0] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Accepted: 07/15/2020] [Indexed: 12/11/2022]
Abstract
The molecular characterization of tumors now informs clinical cancer care for many patients. This advent of molecular oncology is driven by the expanding number of therapeutic biomarkers that can predict sensitivity to both approved and investigational agents. Beyond its role in driving clinical trial enrollments and guiding therapy in individual patients, large-scale clinical genomics in oncology also represents a rapidly expanding research resource for translational scientific discovery. Here, we review the progress, opportunities, and challenges of scientific and translational discovery from prospective clinical genomic screening programs now routinely conducted in cancer patients.
Collapse
|
24
|
Vashistha V, Poonnen PJ, Snowdon JL, Skinner HG, McCaffrey V, Spector NL, Hintze B, Duffy JE, Weeraratne D, Jackson GP, Kelley MJ, Patel VL. Medical oncologists' perspectives of the Veterans Affairs National Precision Oncology Program. PLoS One 2020; 15:e0235861. [PMID: 32706774 PMCID: PMC7380614 DOI: 10.1371/journal.pone.0235861] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 06/24/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND To support the rising need for testing and to standardize tumor DNA sequencing practices within the U.S. Department of Veterans Affairs (VA)'s Veterans Health Administration (VHA), the National Precision Oncology Program (NPOP) was launched in 2016. We sought to assess oncologists' practices, concerns, and perceptions regarding Next-Generation Sequencing (NGS) and the NPOP. MATERIALS AND METHODS Using a purposive total sampling approach, oncologists who had previously ordered NGS for at least one tumor sample through the NPOP were invited to participate in semi-structured interviews. Questions assessed the following: expectations for the NPOP, procedural requirements, applicability of testing results, and the summative utility of the NPOP. Interviews were assessed using an open coding approach. Thematic analysis was conducted to evaluate the completed codebook. Themes were defined deductively by reviewing the direct responses to interview questions as well as inductively by identifying emerging patterns of data. RESULTS Of the 105 medical oncologists who were invited to participate, 20 (19%) were interviewed from 19 different VA medical centers in 14 states. Five recurrent themes were observed: (1) Educational Efforts Regarding Tumor DNA Sequencing Should be Undertaken, (2) Pathology Departments Share a Critical Role in Facilitating Test Completion, (3) Tumor DNA Sequencing via NGS Serves as the Most Comprehensive Testing Modality within Precision Oncology, (4) The Availability of the NPOP Has Expanded Options for Select Patients, and (5) The Completion of Tumor DNA Sequencing through the NPOP Could Help Improve Research Efforts within VHA Oncology Practices. CONCLUSION Medical oncologists believe that the availability of tumor DNA sequencing through the NPOP could potentially lead to an improvement in outcomes for veterans with metastatic solid tumors. Efforts should be directed toward improving oncologists' understanding of sequencing, strengthening collaborative relationships between oncologists and pathologists, and assessing the role of comprehensive NGS panels within the battery of precision tests.
Collapse
Affiliation(s)
- Vishal Vashistha
- Department of Veterans Affairs, National Precision Oncology Program, Durham, NC, United States of America
- Duke Cancer Institute, Durham, NC, United states of America
- Department of Hematology and Oncology, Durham Veterans Affairs Medical Center, Durham, NC, United States of America
| | - Pradeep J. Poonnen
- Department of Veterans Affairs, National Precision Oncology Program, Durham, NC, United States of America
- Duke Cancer Institute, Durham, NC, United states of America
- Department of Hematology and Oncology, Durham Veterans Affairs Medical Center, Durham, NC, United States of America
| | | | - Halcyon G. Skinner
- College of Health, Lehigh University, Bethlehem, PA, United States of America
| | | | - Neil L. Spector
- Department of Veterans Affairs, National Precision Oncology Program, Durham, NC, United States of America
- Duke Cancer Institute, Durham, NC, United states of America
- Department of Hematology and Oncology, Durham Veterans Affairs Medical Center, Durham, NC, United States of America
| | - Bradley Hintze
- Department of Veterans Affairs, National Precision Oncology Program, Durham, NC, United States of America
| | - Jill E. Duffy
- Department of Veterans Affairs, National Precision Oncology Program, Durham, NC, United States of America
| | | | - Gretchen P. Jackson
- Watson Health, IBM, Cambridge, MA, United States of America
- Section of Surgical Sciences, Vanderbilt University Medical Center, Nashville, TN, United States of America
| | - Michael J. Kelley
- Department of Veterans Affairs, National Precision Oncology Program, Durham, NC, United States of America
- Duke Cancer Institute, Durham, NC, United states of America
- Department of Hematology and Oncology, Durham Veterans Affairs Medical Center, Durham, NC, United States of America
| | - Vimla L. Patel
- Center for Cognitive Sciences in Medicine and Public Health, The New York Academy of Medicine, New York City, NY, United States of America
| |
Collapse
|
25
|
Haga SB, Orlando LA. The enduring importance of family health history in the era of genomic medicine and risk assessment. Per Med 2020; 17:229-239. [PMID: 32320338 DOI: 10.2217/pme-2019-0091] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Improving disease risk prediction and tailoring preventive interventions to patient risk factors is one of the primary goals of precision medicine. Family health history is the traditional approach to quickly gather genetic and environmental data relevant to the patient. While the utility of family health history is well-documented, its utilization is variable, in part due to lack of patient and provider knowledge and incomplete or inaccurate data. With the advances and reduced costs of sequencing technologies, comprehensive sequencing tests can be performed as a risk assessment tool. We provide an overview of each of these risk assessment approaches, the benefits and limitations and implementation challenges.
Collapse
Affiliation(s)
- Susanne B Haga
- Center for Applied Genomics & Precision Medicine, Duke University School of Medicine, 101 Science Drive, Box 3382, Durham, NC 27708, USA
| | - Lori A Orlando
- Center for Applied Genomics & Precision Medicine, Duke University School of Medicine, 101 Science Drive, Box 3382, Durham, NC 27708, USA
| |
Collapse
|
26
|
Loh JW, Guccione C, Di Clemente F, Riedlinger G, Ganesan S, Khiabanian H. All-FIT: allele-frequency-based imputation of tumor purity from high-depth sequencing data. Bioinformatics 2020; 36:2173-2180. [PMID: 31750888 PMCID: PMC7141867 DOI: 10.1093/bioinformatics/btz865] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Revised: 11/13/2019] [Accepted: 11/19/2019] [Indexed: 01/14/2023] Open
Abstract
SUMMARY Clinical sequencing aims to identify somatic mutations in cancer cells for accurate diagnosis and treatment. However, most widely used clinical assays lack patient-matched control DNA and additional analysis is needed to distinguish somatic and unfiltered germline variants. Such computational analyses require accurate assessment of tumor cell content in individual specimens. Histological estimates often do not corroborate with results from computational methods that are primarily designed for normal-tumor matched data and can be confounded by genomic heterogeneity and presence of sub-clonal mutations. Allele-frequency-based imputation of tumor (All-FIT) is an iterative weighted least square method to estimate specimen tumor purity based on the allele frequencies of variants detected in high-depth, targeted, clinical sequencing data. Using simulated and clinical data, we demonstrate All-FIT's accuracy and improved performance against leading computational approaches, highlighting the importance of interpreting purity estimates based on expected biology of tumors. AVAILABILITY AND IMPLEMENTATION Freely available at http://software.khiabanian-lab.org. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Jui Wan Loh
- Center for Systems and Computational Biology, Rutgers University, New Brunswick, NJ, USA
- Rutgers Cancer Institute of New Jersey, Rutgers University, New Brunswick, NJ, USA
- Graduate Program in Microbiology and Molecular Genetics, Rutgers University, Piscataway, NJ, USA
| | - Caitlin Guccione
- Center for Systems and Computational Biology, Rutgers University, New Brunswick, NJ, USA
| | - Frances Di Clemente
- Rutgers Cancer Institute of New Jersey, Rutgers University, New Brunswick, NJ, USA
| | - Gregory Riedlinger
- Center for Systems and Computational Biology, Rutgers University, New Brunswick, NJ, USA
- Rutgers Cancer Institute of New Jersey, Rutgers University, New Brunswick, NJ, USA
- Department of Pathology and Laboratory Medicine, Rutgers University, New Brunswick, NJ, USA
| | - Shridar Ganesan
- Center for Systems and Computational Biology, Rutgers University, New Brunswick, NJ, USA
- Rutgers Cancer Institute of New Jersey, Rutgers University, New Brunswick, NJ, USA
- Department of Medicine, Rutgers Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ, USA
| | - Hossein Khiabanian
- Center for Systems and Computational Biology, Rutgers University, New Brunswick, NJ, USA
- Rutgers Cancer Institute of New Jersey, Rutgers University, New Brunswick, NJ, USA
- Department of Pathology and Laboratory Medicine, Rutgers University, New Brunswick, NJ, USA
| |
Collapse
|
27
|
Conway JR, Warner JL, Rubinstein WS, Miller RS. Next-Generation Sequencing and the Clinical Oncology Workflow: Data Challenges, Proposed Solutions, and a Call to Action. JCO Precis Oncol 2019; 3:PO.19.00232. [PMID: 32923847 PMCID: PMC7446333 DOI: 10.1200/po.19.00232] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/30/2019] [Indexed: 02/04/2023] Open
Abstract
PURPOSE Next-generation sequencing (NGS) of tumor and germline DNA is foundational for precision oncology, with rapidly expanding diagnostic, prognostic, and therapeutic implications. Although few question the importance of NGS in modern oncology care, the process of gathering primary molecular data, integrating it into electronic health records, and optimally using it as part of a clinical workflow remains far from seamless. Numerous challenges persist around data standards and interoperability, and clinicians frequently face difficulties in managing the growing amount of genomic knowledge required to care for patients and keep up to date. METHODS This review provides a descriptive analysis of genomic data workflows for NGS data in clinical oncology and issues that arise from the inconsistent use of standards for sharing data across systems. Potential solutions are described. RESULTS NGS technology, especially for somatic genomics, is well established and widely used in routine patient care, quality measurement, and research. Available genomic knowledge bases play an evolving role in patient management but lack harmonization with one another. Questions about their provenance and timeliness of updating remain. Potentially useful standards for sharing genomic data, such as HL7 FHIR and mCODE, remain primarily in the research and/or development stage. Nonetheless, their impact will likely be seen as uptake increases across care settings and laboratories. The specific use case of ASCO CancerLinQ, as a clinicogenomic database, is discussed. CONCLUSION Because the electronic health records of today seem ill suited for managing genomic data, other solutions are required, including universal data standards and applications that use application programming interfaces, along with a commitment on the part of sequencing laboratories to consistently provide structured genomic data for clinical use.
Collapse
Affiliation(s)
- Jake R. Conway
- Harvard Medical School, Boston, MA
- Dana-Farber Cancer Institute, Boston, MA
| | | | | | | |
Collapse
|
28
|
Manolio TA, Rowley R, Williams MS, Roden D, Ginsburg GS, Bult C, Chisholm RL, Deverka PA, McLeod HL, Mensah GA, Relling MV, Rodriguez LL, Tamburro C, Green ED. Opportunities, resources, and techniques for implementing genomics in clinical care. Lancet 2019; 394:511-520. [PMID: 31395439 PMCID: PMC6699751 DOI: 10.1016/s0140-6736(19)31140-7] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 04/09/2019] [Accepted: 05/03/2019] [Indexed: 12/19/2022]
Abstract
Advances in technologies for assessing genomic variation and an increasing understanding of the effects of genomic variants on health and disease are driving the transition of genomics from the research laboratory into clinical care. Genomic medicine, or the use of an individual's genomic information as part of their clinical care, is increasingly gaining acceptance in routine practice, including in assessing disease risk in individuals and their families, diagnosing rare and undiagnosed diseases, and improving drug safety and efficacy. We describe the major types and measurement tools of genomic variation that are currently of clinical importance, review approaches to interpreting genomic sequence variants, identify publicly available tools and resources for genomic test interpretation, and discuss several key barriers in using genomic information in routine clinical practice.
Collapse
Affiliation(s)
- Teri A Manolio
- National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA.
| | - Robb Rowley
- National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | | | - Dan Roden
- Department of Medicine, Department of Pharmacology, and Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Geoffrey S Ginsburg
- Duke Center for Applied Genomic and Precision Medicine, Duke University, Durham, NC, USA
| | - Carol Bult
- The Jackson Laboratory for Mammalian Genetics, Bar Harbor, ME, USA
| | - Rex L Chisholm
- Center for Genetic Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | | | - Howard L McLeod
- DeBartolo Family Personalized Medicine Institute, Moffitt Cancer Center, Tampa, FL, USA
| | - George A Mensah
- Center for Translation Research and Implementation Science, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Mary V Relling
- Pharmaceutical Sciences Department, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Laura Lyman Rodriguez
- National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Cecelia Tamburro
- National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Eric D Green
- National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
29
|
The Development of a Personalised Training Framework: Implementation of Emerging Technologies for Performance. J Funct Morphol Kinesiol 2019; 4:jfmk4020025. [PMID: 33467340 PMCID: PMC7739422 DOI: 10.3390/jfmk4020025] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Revised: 05/13/2019] [Accepted: 05/15/2019] [Indexed: 02/06/2023] Open
Abstract
Over the last decade, there has been considerable interest in the individualisation of athlete training, including the use of genetic information, alongside more advanced data capture and analysis techniques. Here, we explore the evidence for, and practical use of, a number of these emerging technologies, including the measurement and quantification of epigenetic changes, microbiome analysis and the use of cell-free DNA, along with data mining and machine learning. In doing so, we develop a theoretical model for the use of these technologies in an elite sport setting, allowing the coach to better answer six key questions: (1) To what training will my athlete best respond? (2) How well is my athlete adapting to training? (3) When should I change the training stimulus (i.e., has the athlete reached their adaptive ceiling for this training modality)? (4) How long will it take for a certain adaptation to occur? (5) How well is my athlete tolerating the current training load? (6) What load can my athlete handle today? Special consideration is given to whether such an individualised training framework will outperform current methods as well as the challenges in implementing this approach.
Collapse
|
30
|
Reed EK, Steinmark L, Seibert DC, Edelman E. Somatic Testing: Implications for Targeted Treatment. Semin Oncol Nurs 2019; 35:22-33. [PMID: 30660356 DOI: 10.1016/j.soncn.2018.12.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
OBJECTIVE To provide an overview of key considerations for somatic testing for the purpose of targeting cancer treatment. DATA SOURCES Literature; research reports. CONCLUSION Genomic testing of cancer cells to identify variants that drive the carcinogenic process is becoming common in clinical settings. Providers and patients need to weigh the potential benefits of testing with technologic and logistic issues. IMPLICATIONS FOR NURSING PRACTICE Testing is available for thousands of genomic variants to identify one or more to guide targeted treatment. Oncology nurses need to understand the benefits and limitations of participating in patient-centered implementation of this testing.
Collapse
|
31
|
Berland L, Heeke S, Humbert O, Macocco A, Long-Mira E, Lassalle S, Lespinet-Fabre V, Lalvée S, Bordone O, Cohen C, Leroy S, Hofman V, Hofman P, Ilié M. Current views on tumor mutational burden in patients with non-small cell lung cancer treated by immune checkpoint inhibitors. J Thorac Dis 2019; 11:S71-S80. [PMID: 30775030 DOI: 10.21037/jtd.2018.11.102] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
In the last few years, the treatment of patients with non-small cell lung cancer (NSCLC) has impressively benefitted from immunotherapy, in particular from the inhibition of immune checkpoints such as programmed cell death-1 (PD-1) and programmed cell death ligand-1 (PD-L1). However, despite the significant survival benefit for some patients with advanced NSCLC, the objective response rates (ORRs) remain relatively low no more than 20-30% with a large proportion of patients demonstrating primary resistance. Although the selection of NSCLC patients for the first-line treatment is currently guided by the expression of PD-L1 in tumor cells as detected by immunohistochemistry, this is not the case for the second-line setting. Moreover, the sensitivity and specificity of PD-L1 expression is modest which has prompted the search for additional predictive biomarkers. In this context, the assessment of the tumor mutational burden (TMB), defined as the total number of nonsynonymous mutations in the coding regions of genes, has recently emerged as an additional powerful biomarker to select patients for immunotherapy. The purpose of our review is to highlight the recent advances as well as the challenges and perspectives in the field of TMB and immunotherapy for patients with NSCLC.
Collapse
Affiliation(s)
- Léa Berland
- Laboratory of Clinical and Experimental Pathology, Pasteur Hospital, University Hospital Federation OncoAge, Antoine Lacassagne Comprehensive Cancer Center, Université Côte d'Azur, Nice, France.,UFR Médicine, Antoine Lacassagne Comprehensive Cancer Center, Université Côte d'Azur, Nice, France
| | - Simon Heeke
- Institute of Research on Cancer and Ageing of Nice (IRCAN), University Hospital Federation OncoAge, CNRS, INSERM, Antoine Lacassagne Comprehensive Cancer Center, Université Côte d'Azur, Nice, France
| | - Olivier Humbert
- Department of Nuclear Medicine, Antoine Lacassagne Comprehensive Cancer Center, Université Côte d'Azur, Nice, France
| | - Adam Macocco
- Laboratory of Clinical and Experimental Pathology, Pasteur Hospital, University Hospital Federation OncoAge, Antoine Lacassagne Comprehensive Cancer Center, Université Côte d'Azur, Nice, France.,UFR Médicine, Antoine Lacassagne Comprehensive Cancer Center, Université Côte d'Azur, Nice, France
| | - Elodie Long-Mira
- Laboratory of Clinical and Experimental Pathology, Pasteur Hospital, University Hospital Federation OncoAge, Antoine Lacassagne Comprehensive Cancer Center, Université Côte d'Azur, Nice, France.,UFR Médicine, Antoine Lacassagne Comprehensive Cancer Center, Université Côte d'Azur, Nice, France
| | - Sandra Lassalle
- Laboratory of Clinical and Experimental Pathology, Pasteur Hospital, University Hospital Federation OncoAge, Antoine Lacassagne Comprehensive Cancer Center, Université Côte d'Azur, Nice, France.,UFR Médicine, Antoine Lacassagne Comprehensive Cancer Center, Université Côte d'Azur, Nice, France
| | - Virginie Lespinet-Fabre
- Laboratory of Clinical and Experimental Pathology, Pasteur Hospital, University Hospital Federation OncoAge, Antoine Lacassagne Comprehensive Cancer Center, Université Côte d'Azur, Nice, France
| | - Salomé Lalvée
- Laboratory of Clinical and Experimental Pathology, Pasteur Hospital, University Hospital Federation OncoAge, Antoine Lacassagne Comprehensive Cancer Center, Université Côte d'Azur, Nice, France
| | - Olivier Bordone
- Laboratory of Clinical and Experimental Pathology, Pasteur Hospital, University Hospital Federation OncoAge, Antoine Lacassagne Comprehensive Cancer Center, Université Côte d'Azur, Nice, France
| | - Charlotte Cohen
- Department of Thoracic Surgery, Pasteur Hospital, University Hospital Federation OncoAge, Université Côte d'Azur, Nice, France
| | - Sylvie Leroy
- Department of Pulmonary Medicine and Oncology, Pasteur Hospital, University Hospital Federation OncoAge, Université Côte d'Azur, Nice, France
| | - Véronique Hofman
- Laboratory of Clinical and Experimental Pathology, Pasteur Hospital, University Hospital Federation OncoAge, Antoine Lacassagne Comprehensive Cancer Center, Université Côte d'Azur, Nice, France.,UFR Médicine, Antoine Lacassagne Comprehensive Cancer Center, Université Côte d'Azur, Nice, France.,Department of Thoracic Surgery, Pasteur Hospital, University Hospital Federation OncoAge, Université Côte d'Azur, Nice, France
| | - Paul Hofman
- Laboratory of Clinical and Experimental Pathology, Pasteur Hospital, University Hospital Federation OncoAge, Antoine Lacassagne Comprehensive Cancer Center, Université Côte d'Azur, Nice, France.,UFR Médicine, Antoine Lacassagne Comprehensive Cancer Center, Université Côte d'Azur, Nice, France.,Hospital-Related Biobank (BB-0033-00025), Pasteur Hospital, University Hospital Federation OncoAge, Université Côte d'Azur, Nice, France
| | - Marius Ilié
- Laboratory of Clinical and Experimental Pathology, Pasteur Hospital, University Hospital Federation OncoAge, Antoine Lacassagne Comprehensive Cancer Center, Université Côte d'Azur, Nice, France.,UFR Médicine, Antoine Lacassagne Comprehensive Cancer Center, Université Côte d'Azur, Nice, France.,Hospital-Related Biobank (BB-0033-00025), Pasteur Hospital, University Hospital Federation OncoAge, Université Côte d'Azur, Nice, France
| |
Collapse
|
32
|
Abstract
Large-scale tumor profiling studies have generated massive amounts of data that have been instrumental for the detection of recurrent driver mutations in many tumor types. These driver mutations as well as the concurrent passenger mutations are now being used for a more accurate diagnosis of the tumor and prognosis for the patient. Moreover, therapeutic inhibitors toward specific mutations are already on the market and many clinical trials are ongoing to approve novel therapeutic drugs. The broad-range identification of these somatic mutations is key to this tailored personalized medicine approach, which preferentially has to be performed by a multigene multihotspot method such as massive parallel sequencing, also called next generation sequencing (NGS). The implementation of NGS in molecular diagnostics of tumor profiling however, requires a firm validation to minimize the occurrence of false positives and false negatives, thereby yielding highly accurate and robust clinical data.Here, we describe the different performance characteristics as well as quality metrics that should be analyzed for the robust diagnostic validation of tumor profiling in order to meet the requirements of international standards specific for medical laboratories, such as the ISO15189:2012 standard. These metrics include assays that assess the precision, limit of detection, accuracy, sensitivity, specificity, and robustness of the entire workflow from DNA enrichment up to the final report.
Collapse
Affiliation(s)
- Guy Froyen
- Laboratory for Molecular Diagnostics, Department of Clinical Biology, Jessa Hospital, Hasselt, Belgium.
| | - Brigitte Maes
- Laboratory for Molecular Diagnostics, Department of Clinical Biology, Jessa Hospital, Hasselt, Belgium
| |
Collapse
|
33
|
Narrandes S, Xu W. Gene Expression Detection Assay for Cancer Clinical Use. J Cancer 2018; 9:2249-2265. [PMID: 30026820 PMCID: PMC6036716 DOI: 10.7150/jca.24744] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Accepted: 05/15/2018] [Indexed: 12/23/2022] Open
Abstract
Cancer is a genetic disease where genetic variations cause abnormally functioning genes that appear to alter expression. Proteins, the final products of gene expression, determine the phenotypes and biological processes. Therefore, detecting gene expression levels can be used for cancer diagnosis, prognosis, and treatment prediction in a clinical setting. In this review, we investigated six gene expression assay systems (qRT-PCR, DNA microarray, nCounter, RNA-Seq, FISH, and tissue microarray) that are currently being used in clinical cancer studies. Some of these methods are also commonly used in a modified way; for example, detection of DNA content or protein expression. Herein, we discuss their principles, sample preparation, design, quantification and sensitivity, data analysis, time for sample preparation and processing, and cost. We also compared these methods according to their sample selection, particularly for the feasibility of using formalin-fixed paraffin-embedded (FFPE) samples, which are routinely archived for clinical cancer studies. We intend to provide a guideline for choosing an assay method with respect to its oncological applications in a clinical setting.
Collapse
Affiliation(s)
- Shavira Narrandes
- Departments of Biochemistry and Medical Genetics, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Canada.,Research Institute of Oncology and Hematology, CancerCare Manitoba, Winnipeg, Canada
| | - Wayne Xu
- Departments of Biochemistry and Medical Genetics, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Canada.,Research Institute of Oncology and Hematology, CancerCare Manitoba, Winnipeg, Canada.,College of Pharmacy, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Canada
| |
Collapse
|
34
|
Stewart CM, Kothari PD, Mouliere F, Mair R, Somnay S, Benayed R, Zehir A, Weigelt B, Dawson SJ, Arcila ME, Berger MF, Tsui DW. The value of cell-free DNA for molecular pathology. J Pathol 2018; 244:616-627. [PMID: 29380875 DOI: 10.1002/path.5048] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Revised: 01/23/2018] [Accepted: 01/25/2018] [Indexed: 02/06/2023]
Abstract
Over the past decade, advances in molecular biology and genomics techniques have revolutionized the diagnosis and treatment of cancer. The technological advances in tissue profiling have also been applied to the study of cell-free nucleic acids, an area of increasing interest for molecular pathology. Cell-free nucleic acids are released from tumour cells into the surrounding body fluids and can be assayed non-invasively. The repertoire of genomic alterations in circulating tumour DNA (ctDNA) is reflective of both primary tumours and distant metastatic sites, and ctDNA can be sampled multiple times, thereby overcoming the limitations of the analysis of single biopsies. Furthermore, ctDNA can be sampled regularly to monitor response to treatment, to define the evolution of the tumour genome, and to assess the acquisition of resistance and minimal residual disease. Recently, clinical ctDNA assays have been approved for guidance of therapy, which is an exciting first step in translating cell-free nucleic acid research tests into clinical use for oncology. In this review, we discuss the advantages of cell-free nucleic acids as analytes in different body fluids, including blood plasma, urine, and cerebrospinal fluid, and their clinical applications in solid tumours and haematological malignancies. We will also discuss practical considerations for clinical deployment, such as preanalytical factors and regulatory requirements. Copyright © 2018 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Caitlin M Stewart
- Marie-José and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA.,Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Prachi D Kothari
- Marie-José and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA.,Department of Pediatric Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Florent Mouliere
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK.,Cancer Research UK Major Centre - Cambridge, Cancer Research UK Cambridge Institute, Cambridge, UK
| | - Richard Mair
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK.,Cancer Research UK Major Centre - Cambridge, Cancer Research UK Cambridge Institute, Cambridge, UK.,Division of Neurosurgery, Department of Clinical Neurosciences, University of Cambridge, UK
| | - Saira Somnay
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ryma Benayed
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ahmet Zehir
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Britta Weigelt
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Sarah-Jane Dawson
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Victoria, Australia.,Centre for Cancer Research, University of Melbourne, Victoria, Australia
| | - Maria E Arcila
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Michael F Berger
- Marie-José and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA.,Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Dana Wy Tsui
- Marie-José and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA.,Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| |
Collapse
|
35
|
Mehrotra M, Duose DY, Singh RR, Barkoh BA, Manekia J, Harmon MA, Patel KP, Routbort MJ, Medeiros LJ, Wistuba II, Luthra R. Versatile ion S5XL sequencer for targeted next generation sequencing of solid tumors in a clinical laboratory. PLoS One 2017; 12:e0181968. [PMID: 28767674 PMCID: PMC5540534 DOI: 10.1371/journal.pone.0181968] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Accepted: 07/10/2017] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Next generation sequencing based tumor tissue genotyping involves complex workflow and a relatively longer turnaround time. Semiconductor based next generation platforms varied from low throughput Ion PGM to high throughput Ion Proton and Ion S5XL sequencer. In this study, we compared Ion PGM and Ion Proton, with a new Ion S5XL NGS system for workflow scalability, analytical sensitivity and specificity, turnaround time and sequencing performance in a clinical laboratory. METHODS Eighteen solid tumor samples positive for various mutations as detected previously by Ion PGM and Ion Proton were selected for study. Libraries were prepared using DNA (range10-40ng) from micro-dissected formalin-fixed, paraffin-embedded (FFPE) specimens using the Ion Ampliseq Library Kit 2.0 for comprehensive cancer (CCP), oncomine comprehensive cancer (OCP) and cancer hotspot panel v2 (CHPv2) panel as per manufacturer's instructions. The CHPv2 were sequenced using Ion PGM whereas CCP and OCP were sequenced using Ion Proton respectively. All the three libraries were further sequenced individually (S540) or multiplexed (S530) using Ion S5XL. For S5XL, Ion chef was used to automate template preparation, enrichment of ion spheres and chip loading. Data analysis was performed using Torrent Suite 4.6 software on board S5XL and Ion Reporter. A limit of detection and reproducibility studies was performed using serially diluted DLD1 cell line. RESULTS A total of 241 variant calls (235 single nucleotide variants and 6 indels) expected in the studied cohort were successfully detected by S5XL with 100% and 97% concordance with Ion PGM and Proton, respectively. Sequencing run time was reduced from 4.5 to 2.5 hours with output range of 3-5 GB (S530) and 8-9.3Gb (S540). Data analysis time for the Ion S5XL is faster 1 h (S520), 2.5 h (S530) and 5 h (S540) chip, respectively as compared to the Ion PGM (3.5-5 h) and Ion Proton (8h). A limit detection of 5% allelic frequency was established along with high inter-run reproducibility. CONCLUSION Ion S5XL system simplified workflow in a clinical laboratory, was feasible for running smaller and larger panels on the same instrument, had a shorter turnaround time, and showed good concordance for variant calls with similar sensitivity and reproducibility as the Ion PGM and Proton.
Collapse
Affiliation(s)
- Meenakshi Mehrotra
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States of America.,Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States of America
| | - Dzifa Yawa Duose
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States of America
| | - Rajesh R Singh
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States of America
| | - Bedia A Barkoh
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States of America
| | - Jawad Manekia
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States of America
| | - Michael A Harmon
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States of America
| | - Keyur P Patel
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States of America
| | - Mark J Routbort
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States of America
| | - L Jeffrey Medeiros
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States of America
| | - Ignacio I Wistuba
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States of America
| | - Rajyalakshmi Luthra
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States of America.,Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States of America
| |
Collapse
|
36
|
Tack V, Dufraing K, Deans ZC, van Krieken HJ, Dequeker EMC. The ins and outs of molecular pathology reporting. Virchows Arch 2017; 471:199-207. [PMID: 28343306 DOI: 10.1007/s00428-017-2108-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Revised: 03/07/2017] [Accepted: 03/12/2017] [Indexed: 01/15/2023]
Abstract
The raid evolution in molecular pathology resulting in an increasing complexity requires careful reporting. The need for standardisation is clearer than ever. While synoptic reporting was first used for reporting hereditary genetic diseases, it is becoming more frequent in pathology, especially molecular pathology reports too. The narrative approach is no longer feasible with the growing amount of essential data present on the report, although narrative components are still necessary for interpretation in molecular pathology. On the way towards standardisation of reports, guidelines can be a helpful tool. There are several guidelines that focus on reporting in the field of hereditary diseases, but it is not always feasible to extrapolate these to the reporting of somatic variants in molecular pathology. The rise of multi-gene testing causes challenges for the laboratories. In order to provide a continuous optimisation of the laboratory testing process, including reporting, external quality assessment is essential and has already proven to improve the quality of reports. In general, a clear and concise report for molecular pathology can be created by including elements deemed important by different guidelines, adapting the report to the process flows of the laboratory and integrating the report with the laboratory information management system and the patient record.
Collapse
Affiliation(s)
- Véronique Tack
- Biomedical Quality Assurance Research Unit, Department of Public Health and Primary Care, KU Leuven, Kapucijnenvoer 35 Blok D, 3000, Leuven, Belgium
| | - Kelly Dufraing
- Biomedical Quality Assurance Research Unit, Department of Public Health and Primary Care, KU Leuven, Kapucijnenvoer 35 Blok D, 3000, Leuven, Belgium
| | - Zandra C Deans
- Department of Laboratory Medicine, UK NEQAS for Molecular Genetics, UK NEQAS Edinburgh, The Royal Infirmary of Edinburgh, Edinburgh, UK
| | - Han J van Krieken
- Department of Pathology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Elisabeth M C Dequeker
- Biomedical Quality Assurance Research Unit, Department of Public Health and Primary Care, KU Leuven, Kapucijnenvoer 35 Blok D, 3000, Leuven, Belgium.
| |
Collapse
|
37
|
Kamps R, Brandão RD, Bosch BJVD, Paulussen ADC, Xanthoulea S, Blok MJ, Romano A. Next-Generation Sequencing in Oncology: Genetic Diagnosis, Risk Prediction and Cancer Classification. Int J Mol Sci 2017; 18:ijms18020308. [PMID: 28146134 PMCID: PMC5343844 DOI: 10.3390/ijms18020308] [Citation(s) in RCA: 315] [Impact Index Per Article: 39.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Accepted: 01/19/2017] [Indexed: 12/17/2022] Open
Abstract
Next-generation sequencing (NGS) technology has expanded in the last decades with significant improvements in the reliability, sequencing chemistry, pipeline analyses, data interpretation and costs. Such advances make the use of NGS feasible in clinical practice today. This review describes the recent technological developments in NGS applied to the field of oncology. A number of clinical applications are reviewed, i.e., mutation detection in inherited cancer syndromes based on DNA-sequencing, detection of spliceogenic variants based on RNA-sequencing, DNA-sequencing to identify risk modifiers and application for pre-implantation genetic diagnosis, cancer somatic mutation analysis, pharmacogenetics and liquid biopsy. Conclusive remarks, clinical limitations, implications and ethical considerations that relate to the different applications are provided.
Collapse
Affiliation(s)
- Rick Kamps
- Department of Clinical Genetics: GROW-School for Oncology and Developmental Biology, Maastricht University Medical Centre, 6229HX Maastricht, The Netherlands.
| | - Rita D Brandão
- Department of Clinical Genetics: GROW-School for Oncology and Developmental Biology, Maastricht University Medical Centre, 6229HX Maastricht, The Netherlands.
| | - Bianca J van den Bosch
- Department of Clinical Genetics: GROW-School for Oncology and Developmental Biology, Maastricht University Medical Centre, 6229HX Maastricht, The Netherlands.
| | - Aimee D C Paulussen
- Department of Clinical Genetics: GROW-School for Oncology and Developmental Biology, Maastricht University Medical Centre, 6229HX Maastricht, The Netherlands.
| | - Sofia Xanthoulea
- Department of Gynaecology and Obstetrics: GROW-School for Oncology and Developmental Biology, Maastricht University Medical Centre, 6229HX Maastricht, The Netherlands.
| | - Marinus J Blok
- Department of Clinical Genetics: GROW-School for Oncology and Developmental Biology, Maastricht University Medical Centre, 6229HX Maastricht, The Netherlands.
| | - Andrea Romano
- Department of Gynaecology and Obstetrics: GROW-School for Oncology and Developmental Biology, Maastricht University Medical Centre, 6229HX Maastricht, The Netherlands.
| |
Collapse
|
38
|
Applications of molecular testing in surgical pathology of the head and neck. Mod Pathol 2017; 30:S104-S111. [PMID: 28060367 DOI: 10.1038/modpathol.2016.192] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Revised: 10/12/2016] [Accepted: 10/12/2016] [Indexed: 12/15/2022]
Abstract
Molecular testing in routine surgical pathology is becoming an important component of the workup of many different types of tumors. In fact, in some organ systems, guidelines now suggest that the standard of care is to obtain specific molecular panels for tumor classification and/or therapeutic planning. In the head and neck, clinically applicable molecular tests are not as abundant as in other organ systems. Most current head and neck biomarkers are utilized for diagnosis rather than as companion diagnostic tests to predict therapeutic response. As the number of potential molecular biomarker assays increases and cost pressures escalate, the pathologist must be able to navigate the molecular testing pathways. This review explores scenarios in which molecular testing might be beneficial and cost-effective in head and neck pathology.
Collapse
|
39
|
Cancer Precision Medicine: From Cancer Screening to Drug Selection and Personalized Immunotherapy. Trends Pharmacol Sci 2016; 38:15-24. [PMID: 27842888 DOI: 10.1016/j.tips.2016.10.013] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Revised: 10/20/2016] [Accepted: 10/24/2016] [Indexed: 12/22/2022]
Abstract
With the accelerating progress in basic and clinical cancer research, a huge body of new discoveries and powerful technologies has allowed us to implement a 'Cancer Precision Medicine (CPM)' system for cancer patients. The CPM system covers a wide range of cancer management including cancer screening, monitoring of relapse/recurrence, selection/prediction of effective drugs/treatments, and personalized immunotherapy. In this system individual cancer patients expect to receive personalized care: an appropriate dose of the right drug at the right time. We here aim to summarize and discuss a possible workflow for precision medicine for cancer patients by reviewing recent booming technologies and treatments that have been used or will potentially be used in the CPM system.
Collapse
|
40
|
Sohal DP, Shrotriya S, Abazeed M, Cruise M, Khorana A. Molecular characteristics of biliary tract cancer. Crit Rev Oncol Hematol 2016; 107:111-118. [DOI: 10.1016/j.critrevonc.2016.08.013] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Revised: 08/09/2016] [Accepted: 08/31/2016] [Indexed: 12/30/2022] Open
|
41
|
Shukla S, Evans JR, Malik R, Feng FY, Dhanasekaran SM, Cao X, Chen G, Beer DG, Jiang H, Chinnaiyan AM. Development of a RNA-Seq Based Prognostic Signature in Lung Adenocarcinoma. J Natl Cancer Inst 2016; 109:2905970. [PMID: 27707839 DOI: 10.1093/jnci/djw200] [Citation(s) in RCA: 138] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Accepted: 08/02/2016] [Indexed: 01/08/2023] Open
Abstract
Background Precision therapy for lung cancer will require comprehensive genomic testing to identify actionable targets as well as ascertain disease prognosis. RNA-seq is a robust platform that meets these requirements, but microarray-derived prognostic signatures are not optimal for RNA-seq data. Thus, we undertook the first prognostic analysis of lung adenocarcinoma RNA-seq data and generated a prognostic signature. Methods Lung adenocarcinoma RNA-seq and clinical data from The Cancer Genome Atlas (TCGA) were divided chronologically into training (n = 255) and validation (n = 157) cohorts. In the training cohort, prognostic association was assessed by univariate Cox analysis. A prognostic signature was built with stepwise multivariable Cox analysis. Outcomes by risk group, stage, and mutation status were analyzed with Kaplan-Meier and multivariable Cox analyses. All the statistical tests were two-sided. Results In the training cohort, 96 genes had prognostic association with P values of less than or equal to 1.00x10-4, including five long noncoding RNAs (lncRNAs). Stepwise regression generated a four-gene signature, including one lncRNA. Signature high-risk cases had worse overall survival (OS) in the TCGA validation cohort (hazard ratio [HR] = 3.07, 95% confidence interval [CI] = 2.00 to 14.62) and a University of Michigan institutional cohort (n = 67; HR = 2.05, 95% CI = 1.18 to 4.55), and worse metastasis-free survival in the TCGA validation cohort (HR = 3.05, 95% CI = 2.31 to 13.37). The four-gene prognostic signature also statistically significantly stratified overall survival in important clinical subsets, including stage I (HR = 2.78, 95% CI = 1.91 to 11.13), EGFR wild-type (HR = 3.01, 95% CI = 1.73 to 14.98), and EGFR mutant (HR = 8.99, 95% CI = 62.23 to 141.44). The four-gene prognostic signature also stood out on top when compared with other prognostic signatures. Conclusions Here, we present the first RNA-seq prognostic signature for lung adenocarcinoma that can provide a powerful prognostic tool for precision oncology as part of an integrated RNA-seq clinical sequencing program.
Collapse
Affiliation(s)
- Sudhanshu Shukla
- Department of Pathology, University of Michigan, Ann Arbor, MI.,Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI
| | - Joseph R Evans
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI
| | - Rohit Malik
- Department of Pathology, University of Michigan, Ann Arbor, MI.,Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI
| | - Felix Y Feng
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI.,Department of Radiation Oncology, University of Michigan, Ann Arbor, MI.,Comprehensive Cancer Center, University of Michigan, Ann Arbor, MI
| | - Saravana M Dhanasekaran
- Department of Pathology, University of Michigan, Ann Arbor, MI.,Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI
| | - Xuhong Cao
- Department of Pathology, University of Michigan, Ann Arbor, MI.,Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI
| | - Guoan Chen
- Department of Surgery, Section of Thoracic Surgery, University of Michigan, Ann Arbor, MI
| | - David G Beer
- Department of Surgery, Section of Thoracic Surgery, University of Michigan, Ann Arbor, MI
| | - Hui Jiang
- Department of Biostatistics, University of Michigan, Ann Arbor, MI
| | - Arul M Chinnaiyan
- Department of Pathology, University of Michigan, Ann Arbor, MI.,Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI.,Department of Biostatistics, University of Michigan, Ann Arbor, MI.,Howard Hughes Medical Institute, University of Michigan, Ann Arbor, MI
| |
Collapse
|
42
|
Paolillo C, Londin E, Fortina P. Next generation sequencing in cancer: opportunities and challenges for precision cancer medicine. Scand J Clin Lab Invest Suppl 2016; 245:S84-91. [PMID: 27542004 DOI: 10.1080/00365513.2016.1210331] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Over the past decade, testing the genes of patients and their specific cancer types has become standardized practice in medical oncology since somatic mutations, changes in gene expression and epigenetic modifications are all hallmarks of cancer. However, while cancer genetic assessment has been limited to single biomarkers to guide the use of therapies, improvements in nucleic acid sequencing technologies and implementation of different genome analysis tools have enabled clinicians to detect these genomic alterations and identify functional and disease-associated genomic variants. Next-generation sequencing (NGS) technologies have provided clues about therapeutic targets and genomic markers for novel clinical applications when standard therapy has failed. While Sanger sequencing, an accurate and sensitive approach, allows for the identification of potential novel variants, it is however limited by the single amplicon being interrogated. Similarly, quantitative and qualitative profiling of gene expression changes also represents a challenge for the cancer field. Both RT-PCR and microarrays are efficient approaches, but are limited to the genes present on the array or being assayed. This leaves vast swaths of the transcriptome, including non-coding RNAs and other features, unexplored. With the advent of the ability to collect and analyze genomic sequence data in a timely fashion and at an ever-decreasing cost, many of these limitations have been overcome and are being incorporated into cancer research and diagnostics giving patients and clinicians new hope for targeted and personalized treatment. Below we highlight the various applications of next-generation sequencing in precision cancer medicine.
Collapse
Affiliation(s)
- Carmela Paolillo
- a Department of Cancer Biology , Sidney Kimmel Medical College , Philadelphia , PA , USA
| | - Eric Londin
- b Computational Medicine Center , Thomas Jefferson University , Philadelphia , PA , USA
| | - Paolo Fortina
- a Department of Cancer Biology , Sidney Kimmel Medical College , Philadelphia , PA , USA
| |
Collapse
|
43
|
Owosho AA, Xu B, Kadempour A, Yom SK, Randazzo J, Ghossein RA, Huryn JM, Estilo CL. Metastatic solid tumors to the jaw and oral soft tissue: A retrospective clinical analysis of 44 patients from a single institution. J Craniomaxillofac Surg 2016; 44:1047-53. [PMID: 27270028 DOI: 10.1016/j.jcms.2016.05.013] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Revised: 04/11/2016] [Accepted: 05/09/2016] [Indexed: 02/06/2023] Open
Abstract
PURPOSE Metastatic solid tumors to the oral cavity are rare, frequently indicative of an end-stage disease process, and associated with poor survival rates. We performed a 20-year retrospective clinical analysis of our institution's cases of solid metastases to the oral cavity, and investigated these patients' clinical outcomes. MATERIAL AND METHODS A retrospective study of patients with metastatic solid tumors to the oral cavity over a 20-year period (October 1996 to September 2015) was conducted at Memorial Sloan Kettering Cancer Center. Patients were selected if they had a histopathologically confirmed diagnosis. Demographic, pathologic, and clinical information were reviewed to identify patient outcomes. RESULTS A total of 44 patients with metastatic non-melanocytic non-hematopoietic tumor to the oral cavity were identified: 24 males and 20 females (39 adults and 5 children) with a mean age of 54.3 years. In all, 24 cases involved the jaw and 20 cases involved the oral soft tissue. Eight patients (18.2%) had oral cavity metastases as the first indication of an occult malignancy. In adult patients, the common primary sites were the lungs (n = 9, 20%), kidney (n = 7, 16%), breast (n = 5, 11%), and colon (n = 4, 9%); and in pediatric patients the adrenal gland (3/5) was the most common site. Of the adult patients, 33 (84.6%) died of disease. From the time of metastasis diagnosis, patients with jaw metastases had a median and mean survival of 12 months and 27.7 months, respectively. In comparison, patients with oral soft tissue metastases had a median survival time of 5 months, and mean of 8 months. One pediatric patient (20%) died of disease 8 months after metastasis diagnosis. CONCLUSION Metastatic solid tumors to the oral cavity can be the first sign of a malignancy. Pediatric patients with oral cavity metastases have a better prognosis compared to adult patients. In this series, adults with oral soft tissue involvement had shorter survival times compared to patients with jaw involvement.
Collapse
Affiliation(s)
- Adepitan A Owosho
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, USA
| | - Bin Xu
- Head and Neck/Endocrine Pathology, Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, USA
| | - Arvin Kadempour
- Dental Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, USA
| | - SaeHee K Yom
- Dental Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, USA
| | - Joseph Randazzo
- Dental Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, USA
| | - Ronald A Ghossein
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, USA
| | - Joseph M Huryn
- Dental Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, USA
| | - Cherry L Estilo
- Dental Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, USA.
| |
Collapse
|
44
|
Luchi N, Capretti P, Pazzagli M, Pinzani P. Powerful qPCR assays for the early detection of latent invaders: interdisciplinary approaches in clinical cancer research and plant pathology. Appl Microbiol Biotechnol 2016; 100:5189-204. [PMID: 27112348 DOI: 10.1007/s00253-016-7541-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Revised: 04/07/2016] [Accepted: 04/10/2016] [Indexed: 12/29/2022]
Abstract
Latent invaders represent the first step of disease before symptoms occur in the host. Based on recent findings, tumors are considered to be ecosystems in which cancer cells act as invasive species that interact with the native host cell species. Analogously, in plants latent fungal pathogens coevolve within symptomless host tissues. For these reasons, similar detection approaches can be used for an early diagnosis of the invasion process in both plants and humans to prevent or reduce the spread of the disease. Molecular tools based on the evaluation of nucleic acids have been developed for the specific, rapid, and early detection of human diseases. During the last decades, these techniques to assess and quantify the proliferation of latent invaders in host cells have been transferred from the medical field to different areas of scientific research, such as plant pathology. An improvement in molecular biology protocols (especially referring to qPCR assays) specifically designed and optimized for detection in host plants is therefore advisable. This work is a cross-disciplinary review discussing the use of a methodological approach that is employed within both medical and plant sciences. It provides an overview of the principal qPCR tools for the detection of latent invaders, focusing on comparisons between clinical cancer research and plant pathology, and recent advances in the early detection of latent invaders to improve prevention and control strategies.
Collapse
Affiliation(s)
- Nicola Luchi
- National Research Council (IPSP-CNR), Institute for Sustainable Plant Protection, Via Madonna del Piano 10, 50019, Sesto Fiorentino Firenze, Italy
| | - Paolo Capretti
- National Research Council (IPSP-CNR), Institute for Sustainable Plant Protection, Via Madonna del Piano 10, 50019, Sesto Fiorentino Firenze, Italy
- Department of Agri-Food Productions and Environmental Sciences (DiSPAA), University of Florence, Piazzale delle Cascine 28, Florence, Italy
| | - Mario Pazzagli
- Department of Clinical, Experimental and Biomedical Sciences, University of Florence, Viale Pieraccini, 6, 50139, Firenze, Italy
| | - Pamela Pinzani
- Department of Clinical, Experimental and Biomedical Sciences, University of Florence, Viale Pieraccini, 6, 50139, Firenze, Italy.
| |
Collapse
|
45
|
Froyen G, Broekmans A, Hillen F, Pat K, Achten R, Mebis J, Rummens JL, Willemse J, Maes B. Validation and Application of a Custom-Designed Targeted Next-Generation Sequencing Panel for the Diagnostic Mutational Profiling of Solid Tumors. PLoS One 2016; 11:e0154038. [PMID: 27101000 PMCID: PMC4839685 DOI: 10.1371/journal.pone.0154038] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Accepted: 04/07/2016] [Indexed: 01/15/2023] Open
Abstract
The inevitable switch from standard molecular methods to next-generation sequencing for the molecular profiling of tumors is challenging for most diagnostic laboratories. However, fixed validation criteria for diagnostic accreditation are not in place because of the great variability in methods and aims. Here, we describe the validation of a custom panel of hotspots in 24 genes for the detection of somatic mutations in non-small cell lung carcinoma, colorectal carcinoma and malignant melanoma starting from FFPE sections, using 14, 36 and 5 cases, respectively. The targeted hotspots were selected for their present or future clinical relevance in solid tumor types. The target regions were enriched with the TruSeq approach starting from limited amounts of DNA. Cost effective sequencing of 12 pooled libraries was done using a micro flow cell on the MiSeq and subsequent data analysis with MiSeqReporter and VariantStudio. The entire workflow was diagnostically validated showing a robust performance with maximal sensitivity and specificity using as thresholds a variant allele frequency >5% and a minimal amplicon coverage of 300. We implemented this method through the analysis of 150 routine diagnostic samples and identified clinically relevant mutations in 16 genes including KRAS (32%), TP53 (32%), BRAF (12%), APC (11%), EGFR (8%) and NRAS (5%). Importantly, the highest success rate was obtained when using also the low quality DNA samples. In conclusion, we provide a workflow for the validation of targeted NGS by a custom-designed pan-solid tumor panel in a molecular diagnostic lab and demonstrate its robustness in a clinical setting.
Collapse
Affiliation(s)
- Guy Froyen
- Department of Clinical Biology, Jessa Hospital, Hasselt, Belgium
- * E-mail:
| | - An Broekmans
- Department of Clinical Biology, Jessa Hospital, Hasselt, Belgium
| | - Femke Hillen
- Department of Clinical Biology, Jessa Hospital, Hasselt, Belgium
| | - Karin Pat
- Department of Pneumology, Jessa Hospital, Hasselt, Belgium
| | - Ruth Achten
- Department of Pathology, Jessa Hospital, Hasselt, Belgium
| | - Jeroen Mebis
- Department of Medical Oncology, Jessa Hospital, Hasselt, Belgium
| | - Jean-Luc Rummens
- Department of Clinical Biology, Jessa Hospital, Hasselt, Belgium
| | - Johan Willemse
- Department of Clinical Biology, Jessa Hospital, Hasselt, Belgium
- Department of Clinical Biology, AZ Turnhout, Turnhout, Belgium
| | - Brigitte Maes
- Department of Clinical Biology, Jessa Hospital, Hasselt, Belgium
| |
Collapse
|
46
|
Katrib A, Hsu W, Bui A, Xing Y. "RADIOTRANSCRIPTOMICS": A synergy of imaging and transcriptomics in clinical assessment. QUANTITATIVE BIOLOGY 2016; 4:1-12. [PMID: 28529815 DOI: 10.1007/s40484-016-0061-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Recent advances in quantitative imaging and "omics" technology have generated a wealth of mineable biological "big data". With the push towards a P4 "predictive, preventive, personalized, and participatory" approach to medicine, researchers began integrating complementary tools to further tune existing diagnostic and therapeutic models. The field of radiogenomics has long pioneered such multidisciplinary investigations in neuroscience and oncology, correlating genotypic and phenotypic signatures to study structural and functional changes in relation to altered molecular behavior. Given the innate dynamic nature of complex disorders and the role of environmental and epigenetic factors in pathogenesis, the transcriptome can further elucidate serial modifications undetected at the genome level. We therefore propose "radiotranscriptomics" as a new member of the P4 medicine initiative, combining transcriptome information, including gene expression and isoform variation, and quantitative image annotations.
Collapse
Affiliation(s)
- Amal Katrib
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - William Hsu
- Department of Radiological Sciences, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Alex Bui
- Department of Radiological Sciences, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Yi Xing
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA
| |
Collapse
|
47
|
Zhu SK. Role of precision medicine in pancreatic cancer. Shijie Huaren Xiaohua Zazhi 2016; 24:4752. [DOI: 10.11569/wcjd.v24.i36.4752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
|
48
|
Lopez J, Harris S, Roda D, Yap TA. Precision Medicine for Molecularly Targeted Agents and Immunotherapies in Early-Phase Clinical Trials. TRANSLATIONAL ONCOGENOMICS 2015; 7:1-11. [PMID: 26609214 PMCID: PMC4648610 DOI: 10.4137/tog.s30533] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/02/2015] [Revised: 10/06/2015] [Accepted: 10/09/2015] [Indexed: 12/12/2022]
Abstract
Precision medicine in oncology promises the matching of genomic, molecular, and clinical data with underlying mechanisms of a range of novel anticancer therapeutics to develop more rational and effective antitumor strategies in a timely manner. However, despite the remarkable progress made in the understanding of novel drivers of different oncogenic processes, success rates for the approval of oncology drugs remain low with substantial fiscal consequences. In this article, we focus on how recent rapid innovations in technology have brought greater clarity to the biological and clinical complexities of different cancers and advanced the development of molecularly targeted agents and immunotherapies in clinical trials. We discuss the key challenges of identifying and validating predictive biomarkers of response and resistance using both tumor and surrogate tissues, as well as the hurdles associated with intratumor heterogeneity. Finally, we outline evolving strategies employed in early-phase trial designs that incorporate omics-based technologies.
Collapse
Affiliation(s)
- Juanita Lopez
- Royal Marsden NHS Foundation Trust, The Institute of Cancer Research, London, UK
| | - Sam Harris
- Royal Marsden NHS Foundation Trust, The Institute of Cancer Research, London, UK
| | - Desam Roda
- Royal Marsden NHS Foundation Trust, The Institute of Cancer Research, London, UK
| | - Timothy A Yap
- Royal Marsden NHS Foundation Trust, The Institute of Cancer Research, London, UK
| |
Collapse
|
49
|
Neuzillet C, Tijeras-Raballand A, Bourget P, Cros J, Couvelard A, Sauvanet A, Vullierme MP, Tournigand C, Hammel P. State of the art and future directions of pancreatic ductal adenocarcinoma therapy. Pharmacol Ther 2015; 155:80-104. [PMID: 26299994 DOI: 10.1016/j.pharmthera.2015.08.006] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Accepted: 08/17/2015] [Indexed: 12/12/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is expected to become the second cause of cancer-related death in 2030. PDAC is the poorest prognostic tumor of the digestive tract, with 80% of patients having advanced disease at diagnosis and 5-year survival rate not exceeding 7%. Until 2010, gemcitabine was the only validated therapy for advanced PDAC with a modest improvement in median overall survival as compared to best supportive care (5-6 vs 3 months). Multiple phase II-III studies have used various combinations of gemcitabine with other cytotoxics or targeted agents, most in vain, in attempt to improve this outcome. Over the past few years, the landscape of PDAC management has undergone major and rapid changes with the approval of the FOLFIRINOX and gemcitabine plus nab-paclitaxel regimens in patients with metastatic disease. These two active combination chemotherapy options yield an improved median overall survival (11.1 vs 8.5 months, respectively) thus making longer survival a reasonably achievable goal. This breakthrough raises some new clinical questions about the management of PDAC. Moreover, better knowledge of the environmental and genetic events that underpin multistep carcinogenesis and of the microenvironment surrounding cancer cells in PDAC has open new perspectives and therapeutic opportunities. In this new dynamic context of deep transformation in basic research and clinical management aspects of the disease, we gathered updated preclinical and clinical data in a multifaceted review encompassing the lessons learned from the past, the yet unanswered questions, and the most promising research priorities to be addressed for the next 5 years.
Collapse
Affiliation(s)
- Cindy Neuzillet
- INSERM UMR1149, Bichat-Beaujon University Hospital (AP-HP - PRES Paris 7 Diderot), 46 rue Henri Huchard, 75018 Paris, and 100 boulevard du Général Leclerc, 92110 Clichy, France; Department of Digestive Oncology, Beaujon University Hospital (AP-HP - PRES Paris 7 Diderot), 100 boulevard du Général Leclerc, 92110 Clichy, France; Department of Medical Oncology, Henri Mondor University Hospital, 51 avenue du Maréchal de Lattre de Tassigny, 94010 Créteil, France.
| | - Annemilaï Tijeras-Raballand
- Department of Translational Research, AAREC Filia Research, 1 place Paul Verlaine, 92100 Boulogne-Billancourt, France
| | - Philippe Bourget
- Department of Clinical Pharmacy, Necker-Enfants Malades University Hospital, 149 Rue de Sèvres, 75015 Paris, France
| | - Jérôme Cros
- INSERM UMR1149, Bichat-Beaujon University Hospital (AP-HP - PRES Paris 7 Diderot), 46 rue Henri Huchard, 75018 Paris, and 100 boulevard du Général Leclerc, 92110 Clichy, France; Department of Pathology, Bichat-Beaujon University Hospital (AP-HP - PRES Paris 7 Diderot), 46 rue Henri Huchard, 75018 Paris, and 100 boulevard du Général Leclerc, 92110 Clichy, France
| | - Anne Couvelard
- INSERM UMR1149, Bichat-Beaujon University Hospital (AP-HP - PRES Paris 7 Diderot), 46 rue Henri Huchard, 75018 Paris, and 100 boulevard du Général Leclerc, 92110 Clichy, France; Department of Pathology, Bichat-Beaujon University Hospital (AP-HP - PRES Paris 7 Diderot), 46 rue Henri Huchard, 75018 Paris, and 100 boulevard du Général Leclerc, 92110 Clichy, France
| | - Alain Sauvanet
- Department of Biliary and Pancreatic Surgery, Beaujon University Hospital (AP-HP - PRES Paris 7 Diderot), 100 boulevard du Général Leclerc, 92110 Clichy, France
| | - Marie-Pierre Vullierme
- Department of Radiology, Beaujon University Hospital (AP-HP - PRES Paris 7 Diderot), 100 boulevard du Général Leclerc, 92110 Clichy, France
| | - Christophe Tournigand
- Department of Medical Oncology, Henri Mondor University Hospital, 51 avenue du Maréchal de Lattre de Tassigny, 94010 Créteil, France
| | - Pascal Hammel
- INSERM UMR1149, Bichat-Beaujon University Hospital (AP-HP - PRES Paris 7 Diderot), 46 rue Henri Huchard, 75018 Paris, and 100 boulevard du Général Leclerc, 92110 Clichy, France; Department of Digestive Oncology, Beaujon University Hospital (AP-HP - PRES Paris 7 Diderot), 100 boulevard du Général Leclerc, 92110 Clichy, France
| |
Collapse
|