1
|
Sears K, Hickey C, Vincent R, Stocks-Candelaria J, Tate J, Bumgardner C, Zhang S, Miller JB. Establishing a Variant Allele Frequency Cutoff for Manual Curation of Medical Exome Sequencing Data. J Mol Diagn 2024:S1525-1578(24)00250-2. [PMID: 39427756 DOI: 10.1016/j.jmoldx.2024.09.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 09/25/2024] [Accepted: 09/30/2024] [Indexed: 10/22/2024] Open
Abstract
Medical exome sequencing pipelines consist of various preprocessing steps to prioritize credible causal variants before a pathologist or variant curation scientist manually interprets potential findings that are then reported to patients. The variant allele frequency (VAF), reported as the fraction of sequencing reads supporting a variant call, can be used to screen for technical artifacts, yet a specific filtering threshold has yet to be established. A total of 13,122 manually curated variants, sequenced from 289 patients using the Agilent SureSelect Focused Exome enrichment kit at the University of Kentucky Clinical Genomics laboratory from October 2019 to May 2023, were evaluated. Totals of 278 single-nucleotide polymorphisms (SNPs) and 3340 SNPs as technical artifacts are clinically reported. All reported variants had a VAF between 0.33 and 0.63, and 82% (2725/3340) of sequencing artifacts had a VAF of <0.33. It is proposed that removing SNPs in which the VAF is less than approximately 0.30 reduces manual curation time by approximately 20% while capturing all medically relevant variants in medical exome sequencing data sets.
Collapse
Affiliation(s)
- Kate Sears
- Department of Pathology and Laboratory Medicine, University of Kentucky, Lexington, Kentucky
| | - Caylin Hickey
- Department of Pathology and Laboratory Medicine, University of Kentucky, Lexington, Kentucky
| | - Ryan Vincent
- Department of Pathology and Laboratory Medicine, University of Kentucky, Lexington, Kentucky
| | | | - Jason Tate
- Department of Pathology and Laboratory Medicine, University of Kentucky, Lexington, Kentucky
| | - Cody Bumgardner
- Department of Pathology and Laboratory Medicine, University of Kentucky, Lexington, Kentucky; Division of Biomedical Informatics, Department of Internal Medicine, University of Kentucky, Lexington, Kentucky
| | - Shulin Zhang
- Department of Pathology and Laboratory Medicine, University of Kentucky, Lexington, Kentucky
| | - Justin B Miller
- Department of Pathology and Laboratory Medicine, University of Kentucky, Lexington, Kentucky; Division of Biomedical Informatics, Department of Internal Medicine, University of Kentucky, Lexington, Kentucky; Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky; Department of Microbiology, Immunology, and Molecular Genetics, University of Kentucky, Lexington, Kentucky.
| |
Collapse
|
2
|
Veney DJ, Wei LY, Toland AE, Presley CJ, Hampel HL, Padamsee TJ, Lee CN, Irvin WJ, Bishop MJ, Kim JJ, Hovick SR, Senter LA, Stover DG. A video intervention to improve patient understanding of tumor genomic testing in patients with cancer. Cancer Med 2024; 13:e70095. [PMID: 39258462 PMCID: PMC11387988 DOI: 10.1002/cam4.70095] [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: 02/02/2024] [Revised: 06/14/2024] [Accepted: 07/29/2024] [Indexed: 09/12/2024] Open
Abstract
INTRODUCTION Tumor genomic testing (TGT) is standard-of-care for most patients with advanced/metastatic cancer. Despite established guidelines, patient education prior to TGT is frequently omitted. The purpose of this study was to evaluate the impact of a concise 4 min video for patient education prior to TGT. METHODS Based on a quality improvement cycle, an animated video was created to be applicable to any cancer type, incorporating culturally diverse images, available in English and Spanish. Patients undergoing standard-of-care TGT were enrolled at a tertiary academic institution and completed survey instruments prior to video viewing (T1) and immediately post-viewing (T2). Instruments included: (1) 10-question objective genomic knowledge; (2) 10-question video message-specific knowledge; (3) 11-question Trust in Provider; (4) attitudes regarding TGT. RESULTS A total of 150 participants were enrolled. For the primary objective, there was a significant increase in video message-specific knowledge (median 10 point increase; p < 0.0001) with no significant change in genomic knowledge/understanding (p = 0.89) or trust in physician/provider (p = 0.59). Results for five questions significantly improved, including the likelihood of TGT impact on treatment decision, incidental germline findings, and cost of testing. Improvement in video message-specific knowledge was consistent across demographic groups, including age, income, and education. CONCLUSIONS A concise, 3-4 min, broadly applicable video incorporating culturally diverse images administered prior to TGT significantly improved video message-specific knowledge across all demographic groups. This resource is publicly available at http://www.tumor-testing.com, with a goal to efficiently educate and empower patients regarding TGT while addressing guidelines within the flow of clinical practice.
Collapse
Affiliation(s)
- Deloris J. Veney
- Division of Medical OncologyOhio State University Comprehensive Cancer CenterColumbusOhioUSA
| | - Lai Y. Wei
- Department of Biomedical InformaticsOhio State UniversityColumbusOhioUSA
| | - Amanda E. Toland
- Division of Human Genetics, Department of Internal MedicineThe Ohio State University Comprehensive Cancer CenterColumbusOhioUSA
- Department of Cancer Biology and GeneticsThe Ohio State UniversityColumbusOhioUSA
| | - Carolyn J. Presley
- Division of Medical OncologyOhio State University Comprehensive Cancer CenterColumbusOhioUSA
| | - Heather L. Hampel
- Division of Clinical Cancer Genomics, Department of Medical Oncology and Therapeutics ResearchCity of Hope National Medical CenterDuarteCaliforniaUSA
| | - Tasleem J. Padamsee
- Division of Health Services Management and PolicyCollege of Public Health, The Ohio State UniversityColumbusOhioUSA
| | - Clara N. Lee
- Division of Health Services Management and PolicyCollege of Public Health, The Ohio State UniversityColumbusOhioUSA
- Present address:
Division of Plastic and Reconstructive SurgeryUniversity of North CarolinaChapel HillNorth CarolinaUSA
| | | | | | - James J. Kim
- Bon Secours‐Mercy Health St. ElizabethYoungstownOhioUSA
| | | | - Leigha A. Senter
- Division of Human Genetics, Department of Internal MedicineThe Ohio State University Comprehensive Cancer CenterColumbusOhioUSA
| | - Daniel G. Stover
- Division of Medical OncologyOhio State University Comprehensive Cancer CenterColumbusOhioUSA
- Department of Biomedical InformaticsOhio State UniversityColumbusOhioUSA
- Pelotonia Institute for Immuno‐Oncology, Ohio State University Comprehensive CancerColumbusOhioUSA
| |
Collapse
|
3
|
Sandoval RL, Bottosso M, Tianyu L, Polidorio N, Bychkovsky BL, Verret B, Gennari A, Cahill S, Achatz MI, Caron O, Imbert-Bouteille M, Noguès C, Mawell KN, Fortuno C, Spurdle AB, Tayob N, Andre F, Garber JE. TP53-associated early breast cancer: new observations from a large cohort. J Natl Cancer Inst 2024; 116:1246-1254. [PMID: 38569880 PMCID: PMC11308175 DOI: 10.1093/jnci/djae074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 03/19/2024] [Accepted: 03/21/2024] [Indexed: 04/05/2024] Open
Abstract
BACKGROUND A recent large, well-annotated international cohort of patients with Li-Fraumeni syndrome and early-stage breast cancer was examined for shared features. METHODS This multicenter cohort study included women with a germline TP53 pathogenic or likely pathogenic variant and nonmetastatic breast cancer diagnosed between 2002 and 2022. Clinical and genetic data were obtained from institutional registries and clinical charts. Descriptive statistics were used to summarize proportions, and differences were assessed using χ2 or Wilcoxon rank sum tests. Metachronous contralateral breast cancer risk, radiation-induced sarcoma risk, and recurrence-free survival were analyzed using the Kaplan-Meier methodology. RESULTS Among 227 women who met study criteria, the median age of first breast cancer diagnosis was 37 years (range = 21-71), 11.9% presented with bilateral synchronous breast cancer, and 18.1% had ductal carcinoma in situ only. In total, 166 (73.1%) patients underwent mastectomies, including 67 bilateral mastectomies as first breast cancer surgery. Among those patients with retained breast tissue, the contralateral breast cancer rate was 25.3% at 5 years. Among 186 invasive tumors, 72.1% were stages I to II, 48.9% were node negative, and the most common subtypes were hormone receptor-positive/HER2-negative (40.9%) and hormone receptor positive/HER2 positive (34.4%). At a median follow-up of 69.9 months (interquartile range = 32.6-125.9), invasive hormone receptor-positive/HER2-negative disease had the highest recurrence risk among the subtypes (5-year recurrence-free survival = 61.1%, P = .001). Among those who received radiation therapy (n = 79), the 5-year radiation-induced sarcoma rate was 4.8%. CONCLUSION We observed high rates of ductal carcinoma in situ, hormone receptor-positive, and HER2-positive breast cancers, with a worse outcome in the hormone receptor-positive/HER2-negative luminal tumors, despite appropriate treatment. Confirmation of these findings in further studies could have implications for breast cancer care in those with Li-Fraumeni syndrome.
Collapse
Affiliation(s)
- Renata L Sandoval
- Medical Oncology Center, Hospital Sírio-Libanês, Brasília, DF, Brazil
- Division of Cancer Genetics and Prevention, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Michele Bottosso
- Department of Surgery, Oncology and Gastroenterology, University of Padua, Padua, Italy
- Medical Oncology Department, Gustave Roussy Cancer Campus, INERM U981, Université Paris Saclay, France
| | - Li Tianyu
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Natalia Polidorio
- Breast Surgery Department, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Brittany L Bychkovsky
- Division of Cancer Genetics and Prevention, Dana-Farber Cancer Institute, Boston, MA, USA
- Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, MA
- Harvard Medical School, Boston, MA, USA
| | - Benjamin Verret
- Medical Oncology Department, Gustave Roussy Cancer Campus, INERM U981, Université Paris Saclay, France
| | - Alessandra Gennari
- Department of Translational Medicine, University of Piemonte Orientale, Novara, Italy
| | - Sophie Cahill
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA, USA
| | | | - Olivier Caron
- Medical Oncology Department, Institut Gustave Roussy, Villejuif, France
| | | | - Catherine Noguès
- Cancer Risk Management Department, Clinical Oncogenetics, Institut Paoli-Calmettes, Marseille, France
- Aix Marseille Université, INSERM, IRD, SESSTIM, Marseille, France
| | - Kara N Mawell
- Department of Medicine, Abramson Cancer Center, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Cristina Fortuno
- Population Health Program, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Amanda B Spurdle
- Population Health Program, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Nabihah Tayob
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Fabrice Andre
- Medical Oncology Department, Gustave Roussy Cancer Campus, INERM U981, Université Paris Saclay, France
| | - Judy E Garber
- Division of Cancer Genetics and Prevention, Dana-Farber Cancer Institute, Boston, MA, USA
- Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, MA
- Harvard Medical School, Boston, MA, USA
| |
Collapse
|
4
|
Summey RM, Gornstein E, Decker B, Dougherty KC, Rader JS, Hopp E. Landscape of potential germline pathogenic variants in select cancer susceptibility genes in patients with adult-type ovarian granulosa cell tumors. Cancer Med 2024; 13:e7340. [PMID: 38898688 PMCID: PMC11187164 DOI: 10.1002/cam4.7340] [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: 10/22/2023] [Revised: 05/14/2024] [Accepted: 05/18/2024] [Indexed: 06/21/2024] Open
Abstract
OBJECTIVE The objective of this study was to assess the frequency of potential germline pathogenic variants that may contribute to risk of development of adult granulosa cell tumors (AGCT) given the paucity of germline testing guidelines for these patients. METHODS This was a retrospective cross-sectional study analyzing comprehensive genomic profiling (CGP) results of AGCT with the FOXL2 p.C134W mutation submitted to Foundation Medicine between 2012 and 2022. Cases with a potential germline pathogenic variant were identified by filtering single nucleotide variants and short indels by variant allele frequency (VAF) and presence in ClinVar for select cancer susceptibility genes. Odds ratios for AGCT risk were calculated compared to a healthy population. RESULTS Prior to analysis, 595 patients were screened and 516 with a somatic FOXL2 p.C134W mutation were included. Potential germline pathogenic variants in a DNA repair-related gene (ATM, BRCA1, BRCA2, CHEK2, PALB2, PMS2, RAD51C, or RAD51D) were found in 6.6% of FOXL2-mutated AGCT. Potential germline pathogenic CHEK2 variants were found in 3.5% (18/516) of AGCT patients, a rate that was 2.8-fold higher than Genome Aggregation Database non-cancer subjects (95% CI 1.8-4.6, p < 0.001). The founder variants p.I157T (38.9%, 7/18) and p.T367fs*15 (c.1100delC; 27.8%, 5/18) were most commonly observed. CHEK2 VAF indicated frequent loss of the wildtype copy of the gene. CONCLUSIONS These results support ongoing utilization of genomic tumor profiling and confirmatory germline testing for potential germline pathogenic variants. Further prospective investigation into the biology of germline variants in this population is warranted.
Collapse
Affiliation(s)
- Rebekah M. Summey
- Division of Gynecologic Oncology, Department of Obstetrics and GynecologyMedical College of WisconsinMilwaukeeWisconsinUSA
| | | | | | | | - Janet S. Rader
- Division of Gynecologic Oncology, Department of Obstetrics and GynecologyMedical College of WisconsinMilwaukeeWisconsinUSA
| | - Elizabeth Hopp
- Division of Gynecologic Oncology, Department of Obstetrics and GynecologyMedical College of WisconsinMilwaukeeWisconsinUSA
| |
Collapse
|
5
|
Abbass MA. Colorectal Cancer Genetics: An Evolutionary Tale of Us. Clin Colon Rectal Surg 2024; 37:125-126. [PMID: 38606048 PMCID: PMC11006443 DOI: 10.1055/s-0043-1770379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/13/2024]
Affiliation(s)
- Mohammad Ali Abbass
- Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| |
Collapse
|
6
|
Fujiwara Y, Kato S, Kurzrock R. Evolution of Precision Oncology, Personalized Medicine, and Molecular Tumor Boards. Surg Oncol Clin N Am 2024; 33:197-216. [PMID: 38401905 PMCID: PMC10894322 DOI: 10.1016/j.soc.2023.12.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/26/2024]
Abstract
With multiple molecular targeted therapies available for patients with cancer that correspond to a specific genetic alteration, the selection of the best treatment is essential to ensure therapeutic efficacy. Molecular tumor boards (MTBs) play a key role in this process to deliver personalized medicine to patients with cancer in a multidisciplinary manner. Historically, personalized medicine has been offered to patients with advanced cancer, but the incorporation of molecular targeted therapies and immunotherapy into the perioperative setting requires clinicians to understand the role of the MTB. Evidence is accumulating to support feasibility and survival benefit in patients treated with matched therapy.
Collapse
Affiliation(s)
- Yu Fujiwara
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY 14263, USA.
| | - Shumei Kato
- Center for Personalized Cancer Therapy, University of California San Diego Moores Cancer Center, 3855 Health Sciences Drive, La Jolla, CA 92093, USA; Division of Hematology and Oncology, Department of Medicine, University of California San Diego Moores Cancer Center, La Jolla, CA, USA
| | - Razelle Kurzrock
- Genomic Sciences and Precision Medicine Center, Medical College of Wisconsin, Froedtert and Medical College of Wisconsin Cancer Center and Linda T. and John A. Mellowes Center for Genomic Sciences and Precision Medicine, 9200 West Wisconsin Avenue, Milwaukee, WI 53226, USA; WIN Consortium, Paris, France; University of Nebraska, Lincoln, NE, USA
| |
Collapse
|
7
|
Greene BL, Stasi SM, Ting MA, Waligorski N, Cole BL, Lockwood CM, Paulson VA, Buchan JG, Lee A, Ojemann JG, Ellenbogen RG, Stevens J, Leary SES. Looking beyond year 1 in the molecular era of pediatric brain tumor diagnosis: confirmatory testing of germline variants found on tumor sequencing. Front Oncol 2024; 14:1338022. [PMID: 38511139 PMCID: PMC10952109 DOI: 10.3389/fonc.2024.1338022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 02/20/2024] [Indexed: 03/22/2024] Open
Abstract
Purpose Somatic molecular profiling of pediatric brain tumors aids with the diagnosis and treatment of patients with a variety of high- and low-grade central nervous system neoplasms. Here, we report follow-up targeted germline evaluation for patients with possible germline variants following tumor only testing in the initial year in which somatic molecular testing was implemented at a single institution. Patients and Methods Somatic testing was completed for all tumors of the central nervous system (CNS) undergoing diagnostic workup at Seattle Children's Hospital during the study period of November 2015 to November 2016. Sequencing was performed in a College of American Pathologists-accredited, Clinical Laboratory Improvements Amendments-certified laboratory using UW-OncoPlex™ assay (version 5), a DNA-based targeted next generation sequencing panel validated to detect genetic alterations in 262 cancer-related genes. We tracked subsequent clinical evaluation and testing on a subgroup of this cohort found to have potential germline variants of interest. Results Molecular sequencing of 88 patients' tumors identified 31 patients with variants that warranted consideration of germline testing. To date, 19 (61%) patients have been tested. Testing confirmed germline variants for ten patients (31% of those identified for testing), one with two germline variants (NF1 and mosaic TP53). Eight (26%) patients died before germline testing was sent. One patient (13%) has not yet had testing. Conclusion Clinically validated molecular profiling of pediatric brain tumors identifies patients who warrant further germline evaluation. Despite this, only a subset of these patients underwent the indicated confirmatory sequencing. Further work is needed to identify barriers and facilitators to this testing, including the role of genetic counseling and consideration of upfront paired somatic-germline testing.
Collapse
Affiliation(s)
- Brittany L. Greene
- Ben Towne Center for Childhood Cancer Research, Seattle Children’s Research Institute, Seattle, WA, United States
- Department of Pediatrics, University of Washington, Seattle, WA, United States
- Cancer and Blood Disorders Center, Seattle Children’s Hospital, Seattle, WA, United States
| | - Shannon M. Stasi
- Department of Laboratories, Seattle Children’s Hospital, Seattle, WA, United States
| | - Michelle A. Ting
- Ben Towne Center for Childhood Cancer Research, Seattle Children’s Research Institute, Seattle, WA, United States
- Department of Pediatrics, University of Washington, Seattle, WA, United States
- Cancer and Blood Disorders Center, Seattle Children’s Hospital, Seattle, WA, United States
| | - Natalie Waligorski
- Cancer and Blood Disorders Center, Seattle Children’s Hospital, Seattle, WA, United States
| | - Bonnie L. Cole
- Department of Laboratories, Seattle Children’s Hospital, Seattle, WA, United States
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, United States
| | - Christina M. Lockwood
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, United States
| | - Vera A. Paulson
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, United States
| | - Jillian G. Buchan
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, United States
| | - Amy Lee
- Department of Pediatric Neurosurgery, Seattle Children’s Hospital, Seattle, WA, United States
- Department of Neurological Surgery, University of Washington, Seattle, WA, United States
| | - Jeffrey G. Ojemann
- Department of Pediatric Neurosurgery, Seattle Children’s Hospital, Seattle, WA, United States
- Department of Neurological Surgery, University of Washington, Seattle, WA, United States
| | - Richard G. Ellenbogen
- Department of Pediatric Neurosurgery, Seattle Children’s Hospital, Seattle, WA, United States
- Department of Neurological Surgery, University of Washington, Seattle, WA, United States
| | - Jeffrey Stevens
- Ben Towne Center for Childhood Cancer Research, Seattle Children’s Research Institute, Seattle, WA, United States
- Cancer and Blood Disorders Center, Seattle Children’s Hospital, Seattle, WA, United States
| | - Sarah E. S. Leary
- Ben Towne Center for Childhood Cancer Research, Seattle Children’s Research Institute, Seattle, WA, United States
- Department of Pediatrics, University of Washington, Seattle, WA, United States
- Cancer and Blood Disorders Center, Seattle Children’s Hospital, Seattle, WA, United States
| |
Collapse
|
8
|
Vandekerkhove G, Giri VN, Halabi S, McNair C, Hamade K, Bitting RL, Wyatt AW. Toward Informed Selection and Interpretation of Clinical Genomic Tests in Prostate Cancer. JCO Precis Oncol 2024; 8:e2300654. [PMID: 38547422 PMCID: PMC10994438 DOI: 10.1200/po.23.00654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 12/15/2023] [Accepted: 02/07/2024] [Indexed: 04/02/2024] Open
Abstract
Clinical genomic testing of patient germline, tumor tissue, or plasma cell-free DNA can enable a personalized approach to cancer management and treatment. In prostate cancer (PCa), broad genotyping tests are now widely used to identify germline and/or somatic alterations in BRCA2 and other DNA damage repair genes. Alterations in these genes can confer cancer sensitivity to poly (ADP-ribose) polymerase inhibitors, are linked with poor prognosis, and can have potential hereditary cancer implications for family members. However, there is huge variability in genomic tests and reporting standards, meaning that for successful implementation of testing in clinical practice, end users must carefully select the most appropriate test for a given patient and critically interpret the results. In this white paper, we outline key pre- and post-test considerations for choosing a genomic test and evaluating reported variants, specifically for patients with advanced PCa. Test choice must be based on clinical context and disease state, availability and suitability of tumor tissue, and the genes and regions that are covered by the test. We describe strategies to recognize false positives or negatives in test results, including frameworks to assess low tumor fraction, subclonal alterations, clonal hematopoiesis, and pathogenic versus nonpathogenic variants. We assume that improved understanding among health care professionals and researchers of the nuances associated with genomic testing will ultimately lead to optimal patient care and clinical decision making.
Collapse
Affiliation(s)
- Gillian Vandekerkhove
- Vancouver Prostate Centre, University of British Columbia, Vancouver, BC, Canada
- Department of Medical Oncology, BC Cancer, Vancouver, BC, Canada
| | - Veda N. Giri
- Yale School of Medicine and Yale Cancer Center, New Haven, CT
| | | | | | | | | | - Alexander W. Wyatt
- Vancouver Prostate Centre, University of British Columbia, Vancouver, BC, Canada
- Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, BC, Canada
| |
Collapse
|
9
|
Lin I, Wei A, Gebo TA, Boutros PC, Flanagan M, Kucine N, Cunniff C, Arboleda VA, Chang VY. Increased Frequency of Clonal Hematopoiesis of Indeterminate Potential in Bloom Syndrome Probands and Carriers. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.02.02.24302163. [PMID: 38370823 PMCID: PMC10871368 DOI: 10.1101/2024.02.02.24302163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/20/2024]
Abstract
Background Bloom Syndrome (BSyn) is an autosomal recessive disorder caused by biallelic germline variants in BLM, which functions to maintain genomic stability. BSyn patients have poor growth, immune defects, insulin resistance, and a significantly increased risk of malignancies, most commonly hematologic. The malignancy risk in carriers of pathogenic variants in BLM (BLM variant carriers) remains understudied. Clonal hematopoiesis of indeterminate potential (CHIP) is defined by presence of somatic mutations in leukemia-related genes in blood of individuals without leukemia and is associated with increased risk of leukemia. We hypothesize that somatic mutations driving clonal expansion may be an underlying mechanism leading to increased cancer risk in BSyn patients and BLM variant carriers. Methods To determine whether de novo or somatic variation is increased in BSyn patients or carriers, we performed and analyzed exome sequencing on BSyn and control trios. Results We discovered that both BSyn patients and carriers had increased numbers of low-frequency, putative somatic variants in CHIP genes compared to controls. Furthermore, BLM variant carriers had increased numbers of somatic variants in DNA methylation genes compared to controls. There was no statistical difference in the numbers of de novo variants in BSyn probands compared to control probands. Conclusion Our findings of increased CHIP in BSyn probands and carriers suggest that one or two germline pathogenic variants in BLM could be sufficient to increase the risk of clonal hematopoiesis. These findings warrant further studies in larger cohorts to determine the significance of CHIP as a potential biomarker of aging, cancer, cardiovascular disease, morbidity and mortality.
Collapse
Affiliation(s)
- Isabella Lin
- Department of Human Genetics, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA
- Department of Computational Medicine, David Geffen School of Medicine, UCLA
| | - Angela Wei
- Department of Human Genetics, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA
- Department of Computational Medicine, David Geffen School of Medicine, UCLA
- Interdepartmental BioInformatics Program, UCLA
| | - Tsumugi A Gebo
- Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, CA
- Institute for Precision Health, University of California Los Angeles, Los Angeles, CA
| | - P C Boutros
- Department of Human Genetics, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA
- Interdepartmental BioInformatics Program, UCLA
- Department of Urology, University of California Los Angeles, Los Angeles, CA
- Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, CA
- Institute for Precision Health, University of California Los Angeles, Los Angeles, CA
- Molecular Biology Institute, University of California, Los Angeles, CA
| | - Maeve Flanagan
- Department of Pediatrics, Weill Cornell Medicine, New York, NY
| | - Nicole Kucine
- Department of Pediatrics, Weill Cornell Medicine, New York, NY
| | - C Cunniff
- Department of Pediatrics, Weill Cornell Medicine, New York, NY
| | - V A Arboleda
- Department of Human Genetics, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA
- Department of Computational Medicine, David Geffen School of Medicine, UCLA
- Interdepartmental BioInformatics Program, UCLA
- Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, CA
- Institute for Precision Health, University of California Los Angeles, Los Angeles, CA
- Molecular Biology Institute, University of California, Los Angeles, CA
| | - V Y Chang
- Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, CA
- Institute for Precision Health, University of California Los Angeles, Los Angeles, CA
- Division of Pediatric Hematology/Oncology, UCLA, Los Angeles, CA
- Children's Discovery and Innovation Institute, UCLA, Los Angeles, CA
| |
Collapse
|
10
|
Wagner JN, Al-Bazaz M, Forstreuter A, Hammada MI, Hille J, Papingi D, Bokemeyer C, Fiedler W. Case Report of a DDX41 Germline Mutation in a Family with Multiple Relatives Suffering from Leukemia. Biomedicines 2023; 12:64. [PMID: 38255170 PMCID: PMC10813731 DOI: 10.3390/biomedicines12010064] [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: 12/07/2023] [Revised: 12/19/2023] [Accepted: 12/22/2023] [Indexed: 01/24/2024] Open
Abstract
INTRODUCTION Previously, it was assumed that genetic influence played a minor role in acute myeloid leukemia (AML). Increasing evidence of germline mutations has emerged, such as DDX41 germline mutation associated with familial AML. CASE PRESENTATION A 64-year-old male patient presented with reduced exercise tolerance and shortness of breath. Following confirmation of AML diagnosis, the patient was enrolled into the AMLSG-30-18 study with a requirement for allogenic stem cell transplantation. The sister was initially selected as a fully HLA-matched donor. However, the family history showed risks for familial AML. Due to the striking family history, further diagnostic steps were initiated to detect a germline mutation. METHODS Using NGS in the patients' bone marrow AML sample, a DDX41 mutation with a VAF of 49% was detected, raising the possibility of a germline mutation. DNA from cheek swabs and eyebrows were tested for the presence of the DDX41 mutation in all siblings. RESULTS DDX41 germline mutation was detected in 5 out of 6 siblings. The sister was excluded as a related donor and the search for an unrelated donor was initiated. CONCLUSION Obtaining family history of cancer patients plays a crucial role in oncology. If a germline mutation is suspected, further family work-up should be initiated.
Collapse
Affiliation(s)
- Jan Nicolai Wagner
- Department of Oncology, Hematology and Bone Marrow Transplantation with Division of Pneumology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (M.A.-B.); (A.F.); (M.I.H.); (J.H.); (C.B.)
| | - Maximilian Al-Bazaz
- Department of Oncology, Hematology and Bone Marrow Transplantation with Division of Pneumology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (M.A.-B.); (A.F.); (M.I.H.); (J.H.); (C.B.)
| | - Anika Forstreuter
- Department of Oncology, Hematology and Bone Marrow Transplantation with Division of Pneumology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (M.A.-B.); (A.F.); (M.I.H.); (J.H.); (C.B.)
| | - Mohammad Ibrahim Hammada
- Department of Oncology, Hematology and Bone Marrow Transplantation with Division of Pneumology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (M.A.-B.); (A.F.); (M.I.H.); (J.H.); (C.B.)
| | - Jurek Hille
- Department of Oncology, Hematology and Bone Marrow Transplantation with Division of Pneumology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (M.A.-B.); (A.F.); (M.I.H.); (J.H.); (C.B.)
| | - Dzhoy Papingi
- Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany;
| | - Carsten Bokemeyer
- Department of Oncology, Hematology and Bone Marrow Transplantation with Division of Pneumology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (M.A.-B.); (A.F.); (M.I.H.); (J.H.); (C.B.)
| | - Walter Fiedler
- Department of Oncology, Hematology and Bone Marrow Transplantation with Division of Pneumology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (M.A.-B.); (A.F.); (M.I.H.); (J.H.); (C.B.)
| |
Collapse
|
11
|
Veney D, Wei L, Toland AE, Presley CJ, Hampel H, Padamsee TJ, Lee CN, Irvin WJ, Bishop M, Kim J, Hovick SR, Senter L, Stover DG. A Video Intervention to Improve Patient Understanding of Tumor Genomic Testing in Patients with Cancer. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.12.05.23299443. [PMID: 38106014 PMCID: PMC10723483 DOI: 10.1101/2023.12.05.23299443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2023]
Abstract
Background Tumor genomic testing (TGT) has become standard-of-care for most patients with advanced/metastatic cancer. Despite established guidelines, patient education prior to TGT is variable or frequently omitted. The purpose of this study was to evaluate the impact of a concise (3-4 minute) video for patient education prior to TGT. Methods Based on a quality improvement cycle, an animated video was created to be applicable to any cancer type, incorporating culturally diverse images, available in English and Spanish. Patients undergoing standard-of care TGT were enrolled at a tertiary academic institution and completed validated survey instruments immediately prior to video viewing (T1) and immediately post-viewing (T2). Instruments included: 1) 10-question objective genomic knowledge/understanding; 2) 10-question video message-specific knowledge/recall; 3) 11-question Trust in Physician/Provider; 4) attitudes regarding TGT. The primary objective was change in outcomes from before to after the video was assessed with Wilcoxon signed rank test. Results From April 2022 to May 2023, a total of 150 participants were enrolled (MBC n=53, LC n=38, OC n=59). For the primary endpoint, there was a significant increase in video message-specific knowledge (median 10 point increase; p<0.0001) with no significant change in genomic knowledge/understanding (p=0.89) or Trust in Physician/Provider (p=0.59). Results for five questions significantly improved, including the likelihood of TGT impact on treatment decision, incidental germline findings, and cost of testing. Improvement in video message-specific knowledge was consistent across demographic groups, including age, income, and education. Individuals with less educational attainment had had greater improvement from before to after video viewing. Conclusions A concise, 3-4 minute, broadly applicable video incorporating culturally diverse images administered prior to TGT significantly improved video message-specific knowledge across all demographic groups. This resource is publicly available at http://www.tumor-testing.com, with a goal to efficiently educate and empower patients regarding TGT while addressing guidelines within the flow of clinical practice. Clinical Trial Registration ClinicalTrials.gov NCT05215769.
Collapse
Affiliation(s)
- Deloris Veney
- Division of Medical Oncology, Ohio State University Comprehensive Cancer Center, Columbus, OH, 43210 USA
| | - Lai Wei
- Department of Biomedical Informatics, Ohio State University, Columbus, OH, 43210 USA
| | - Amanda E. Toland
- Division of Human Genetics, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, OH, 43210 USA
- Department of Cancer Biology and Genetics, The Ohio State University, Columbus, OH, 43210 USA
| | - Carolyn J. Presley
- Division of Medical Oncology, Ohio State University Comprehensive Cancer Center, Columbus, OH, 43210 USA
| | - Heather Hampel
- Division of Clinical Cancer Genomics, Department of Medical Oncology and Therapeutics Research, City of Hope National Medical Center, Duarte, CA 91010
| | - Tasleem J. Padamsee
- Division of Health Services Management and Policy, College of Public Health, The Ohio State University, Columbus, OH, 43210, USA
| | - Clara N. Lee
- Division of Health Services Management and Policy, College of Public Health, The Ohio State University, Columbus, OH, 43210, USA
| | - William J. Irvin
- Bon Secours Cancer Institute at St. Francis, Richmond, Virginia, USA
| | | | - James Kim
- Bon Secours-Mercy Health St. Elizabeth, Youngstown, Ohio, USA
| | - Shelly R. Hovick
- School of Communication, Ohio State University, Columbus, OH, 43210 USA
| | - Leigha Senter
- Division of Human Genetics, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, OH, 43210 USA
| | - Daniel G. Stover
- Division of Medical Oncology, Ohio State University Comprehensive Cancer Center, Columbus, OH, 43210 USA
- Department of Biomedical Informatics, Ohio State University, Columbus, OH, 43210 USA
- Pelotonia Institute for Immuno-Oncology, Ohio State University Comprehensive Cancer, Columbus, OH, 43210 USA
| |
Collapse
|
12
|
Satake T, Kondo S, Tanabe N, Mizuno T, Katsuya Y, Sato J, Koyama T, Yoshida T, Hirata M, Yamamoto N. Pathogenic Germline Variants in BRCA1/2 and p53 Identified by Real-world Comprehensive Cancer Genome Profiling Tests in Asian Patients. CANCER RESEARCH COMMUNICATIONS 2023; 3:2302-2311. [PMID: 37916805 PMCID: PMC10644847 DOI: 10.1158/2767-9764.crc-23-0018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 07/05/2023] [Accepted: 10/26/2023] [Indexed: 11/03/2023]
Abstract
Cancer genome profiling (CGP) occasionally identifies pathogenic germline variants (PGV) in cancer susceptibility genes (CSG) as secondary findings. Here, we analyzed the prevalence and clinical characteristics of PGVs based on nationwide real-world data from CGP tests in Japan. We analyzed the genomic information and clinical characteristics of 23,928 patients with solid cancers who underwent either tumor-only (n = 20,189) or paired tumor-normal (n = 3,739) sequencing CGP tests between June 2019 and December 2021 using the comprehensive national database. We assigned clinical significance for all variants and highlighted the prevalence and characteristics of PGVs. Our primary analysis of the tumor-normal sequencing cohort revealed that 152 patients (4.1%) harbored PGVs in 15 CSGs. Among 783 germline variants, 113 were annotated as PGVs, 70 as benign variants, and 600 as variants of uncertain significance. The number of PGVs identified was highest in BRCA1/2, with 56, followed by TP53, with 18. PGVs were the most prevalent in ovarian and peritoneal cancers, including among cancer types common in Asia. In the tumor-only sequencing cohort, of the 5,184 pathogenic somatic variants across 26 CSGs, 784 (15.1%) were extracted according to the European Society for Medical Oncology recommendations for germline-focused tumor analysis. The prevalence of PGVs was similar to that previously reported in Europe and the United States. This is the largest analysis based on real-world tumor-normal sequencing tests in Asia. The more widespread use of the tumor-normal sequencing CGP test could be reasonable for evaluating PGVs. SIGNIFICANCE We analyzed real-world data from over 23,000 patients in Japan, revealing 4.1% harbored PGVs, particularly in BRCA1/2 and TP53, in CSGs. It highlights the prevalence of PGVs in Asian populations and supports the broader adoption of tumor-normal sequencing CGP tests for PGV evaluation.
Collapse
Affiliation(s)
- Tomoyuki Satake
- Department of Experimental Therapeutics, National Cancer Center Hospital, Tokyo, Japan
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Shunsuke Kondo
- Department of Experimental Therapeutics, National Cancer Center Hospital, Tokyo, Japan
- Outpatient Treatment Center, National Cancer Center Hospital, Tokyo, Japan
| | - Noriko Tanabe
- Genetic Medicine and Services, National Cancer Center Hospital, Tokyo, Japan
| | - Takaaki Mizuno
- Department of Experimental Therapeutics, National Cancer Center Hospital, Tokyo, Japan
| | - Yuki Katsuya
- Department of Experimental Therapeutics, National Cancer Center Hospital, Tokyo, Japan
| | - Jun Sato
- Department of Experimental Therapeutics, National Cancer Center Hospital, Tokyo, Japan
| | - Takafumi Koyama
- Department of Experimental Therapeutics, National Cancer Center Hospital, Tokyo, Japan
| | - Tatsuya Yoshida
- Department of Experimental Therapeutics, National Cancer Center Hospital, Tokyo, Japan
| | - Makoto Hirata
- Genetic Medicine and Services, National Cancer Center Hospital, Tokyo, Japan
| | - Noboru Yamamoto
- Department of Experimental Therapeutics, National Cancer Center Hospital, Tokyo, Japan
| |
Collapse
|
13
|
Fasola G, Barducci MC, Tozzi VD, Cavanna L, Cinieri S, Perrone F, Pinto C, Russo A, Sapino A, Grossi F, Aprile G. Implementation of Precision Oncology in the National Healthcare System: A Statement Proposal Endorsed by Italian Scientific Societies. JCO Precis Oncol 2023; 7:e2300166. [PMID: 37944071 PMCID: PMC10645411 DOI: 10.1200/po.23.00166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 06/15/2023] [Accepted: 09/08/2023] [Indexed: 11/12/2023] Open
Abstract
PURPOSE Precision oncology (PO) promises positive results for patients. To date, in Italy, the effort to implement PO has been made autonomously by regional health institutions in a top-down fashion. This approach is not very efficient and jeopardizes patients' equal access to PO. Similar outcomes have been recorded in other Western countries. We tested a method of collaboration among professionals, scientific societies, and government institutions to facilitate the delivery of PO innovation to patients' bedsides. METHOD We designed an organizational research project on the basis of a bottom-up approach. We started by observing PO-related activities in five health care authorities (HCAs) in one Italian region. We then compared the issues that emerged with those of three additional HCAs in other Italian regions. Using the results of the initial observation and adopting validated multiple-step consensus methods, we finally derived 14 statements that were approved by the four main scientific societies of oncology and pathology at the national level. RESULTS The 14 statements addressed the main issues linked to the implementation of PO in clinical practice. The strong professional consensus advocated for prompt adoption within the national healthcare system. CONCLUSIONS The consensus on the statements that were obtained shows the importance of a synergistic effort among professionals, scientific societies, and health care institutions in defining homogeneous solutions for innovation implementation within the health care system.
Collapse
Affiliation(s)
- Gianpiero Fasola
- Santa Maria della Misericordia University Hospital, ASUFC, Udine, Italy
| | - Maria C. Barducci
- Santa Maria della Misericordia University Hospital, ASUFC, Udine, Italy
| | - Valeria D. Tozzi
- Centre for Research on Health and Social Care Management (CERGAS), SDA Bocconi School of Management, Milan, Italy
| | - Luigi Cavanna
- Piacenza General Hospital, AUSL Piacenza, Piacenza, Italy
- Collegio Italiano dei Primari Oncologi Medici Ospedalieri (CIPOMO), Genova, Italy
| | - Saverio Cinieri
- Complex Medical Oncology Unit, ASL Brindisi Senatore Antonio Perrino Hospital, Brindisi, Italy
- Associazione Italiana Oncologia Medica (AIOM), Milano, Italy
| | - Francesco Perrone
- Associazione Italiana Oncologia Medica (AIOM), Milano, Italy
- Clinical Trials Unit, Istituto Nazionale Tumori-IRCCS, Fondazione G. Pascale, Napoli, Italy
| | - Carmine Pinto
- Medical Oncology, Comprehensive Cancer Centre, AUSL-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Antonio Russo
- Section of Medical Oncology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, Palermo, Italy
- Collegio degli Oncologi Medici Universitari (COMU), University of Palermo, Palermo, Italy
| | - Anna Sapino
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy
- Società Italiana di Anatomia Patologica e Citologia Diagnostica (SIAPeC), Milano, Italy
| | | | - Giuseppe Aprile
- Department of Oncology, San Bortolo General Hospital, Vicenza, Italy
| |
Collapse
|
14
|
Tung N, Dougherty KC, Gatof ES, DeLeonardis K, Hogan L, Tukachinsky H, Gornstein E, Oxnard GR, McGregor K, Keller RB. Potential pathogenic germline variant reporting from tumor comprehensive genomic profiling complements classic approaches to germline testing. NPJ Precis Oncol 2023; 7:76. [PMID: 37568048 PMCID: PMC10421918 DOI: 10.1038/s41698-023-00429-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 07/31/2023] [Indexed: 08/13/2023] Open
Abstract
Existing guidance regarding clinically informed germline testing for patients with cancer is effective for evaluation of classic hereditary cancer syndromes and established gene/cancer type associations. However, current screening methods may miss patients with rare, reduced penetrance, or otherwise occult hereditary risk. Secondary finding of suspected germline variants that may confer inherited cancer risk via tumor comprehensive genomic profiling (CGP) has the potential to help address these limitations. However, reporting practices for secondary finding of germline variants are inconsistent, necessitating solutions for transparent and coherent communication of these potentially important findings. A workflow for improved confidence detection and clear reporting of potential pathogenic germline variants (PPGV) in select cancer susceptibility genes (CSG) was applied to a research dataset from real-world clinical tumor CGP of > 125,000 patients with advanced cancer. The presence and patterns of PPGVs identified across tumor types was assessed with a focus on scenarios in which traditional clinical germline evaluation may have been insufficient to capture genetic risk. PPGVs were identified in 9.7% of tumor CGP cases using tissue- and liquid-based assays across a broad range of cancer types, including in a number of "off-tumor" contexts. Overall, PPGVs were identified in a similar proportion of cancers with National Comprehensive Cancer Network (NCCN) recommendations for germline testing regardless of family history (11%) as in all other cancer types (9%). These findings suggest that tumor CGP can serve as a tool that is complementary to traditional germline genetic evaluation in helping to ascertain inherited susceptibility in patients with advanced cancer.
Collapse
Affiliation(s)
- Nadine Tung
- Department of Medical Oncology, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | | | - Emily Stern Gatof
- Department of Medical Oncology, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Kim DeLeonardis
- Department of Medical Oncology, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Lauren Hogan
- Department of Medical Oncology, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | | | | | | | | | | |
Collapse
|
15
|
O'Brien TD, Potter AB, Driscoll CC, Goh G, Letaw JH, McCabe S, Thanner J, Kulkarni A, Wong R, Medica S, Week T, Buitrago J, Larson A, Camacho KJ, Brown K, Crist R, Conrad C, Evans-Dutson S, Lutz R, Mitchell A, Anur P, Serrato V, Shafer A, Marriott LK, Hamman KJ, Mulford A, Wiszniewski W, Sampson JE, Adey A, O'Roak BJ, Harrington CA, Shannon J, Spellman PT, Richards CS. Population screening shows risk of inherited cancer and familial hypercholesterolemia in Oregon. Am J Hum Genet 2023; 110:1249-1265. [PMID: 37506692 PMCID: PMC10432140 DOI: 10.1016/j.ajhg.2023.06.014] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Revised: 06/28/2023] [Accepted: 06/29/2023] [Indexed: 07/30/2023] Open
Abstract
The Healthy Oregon Project (HOP) is a statewide effort that aims to build a large research repository and influence the health of Oregonians through providing no-cost genetic screening to participants for a next-generation sequencing 32-gene panel comprising genes related to inherited cancers and familial hypercholesterolemia. This type of unbiased population screening can detect at-risk individuals who may otherwise be missed by conventional medical approaches. However, challenges exist for this type of high-throughput testing in an academic setting, including developing a low-cost high-efficiency test and scaling up the clinical laboratory for processing large numbers of samples. Modifications to our academic clinical laboratory including efficient test design, robotics, and a streamlined analysis approach increased our ability to test more than 1,000 samples per month for HOP using only one dedicated HOP laboratory technologist. Additionally, enrollment using a HIPAA-compliant smartphone app and sample collection using mouthwash increased efficiency and reduced cost. Here, we present our experience three years into HOP and discuss the lessons learned, including our successes, challenges, opportunities, and future directions, as well as the genetic screening results for the first 13,670 participants tested. Overall, we have identified 730 pathogenic/likely pathogenic variants in 710 participants in 24 of the 32 genes on the panel. The carrier rate for pathogenic/likely pathogenic variants in the inherited cancer genes on the panel for an unselected population was 5.0% and for familial hypercholesterolemia was 0.3%. Our laboratory experience described here may provide a useful model for population screening projects in other states.
Collapse
Affiliation(s)
- Timothy D O'Brien
- Knight Diagnostic Laboratories, Oregon Health & Science University, Portland, OR 97239, USA
| | - Amiee B Potter
- Knight Diagnostic Laboratories, Oregon Health & Science University, Portland, OR 97239, USA
| | - Catherine C Driscoll
- Knight Diagnostic Laboratories, Oregon Health & Science University, Portland, OR 97239, USA; Cancer Early Detection Advanced Research Center, Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97201, USA
| | - Gregory Goh
- Knight Diagnostic Laboratories, Oregon Health & Science University, Portland, OR 97239, USA; Cancer Early Detection Advanced Research Center, Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97201, USA
| | - John H Letaw
- Knight Diagnostic Laboratories, Oregon Health & Science University, Portland, OR 97239, USA
| | - Sarah McCabe
- Knight Diagnostic Laboratories, Oregon Health & Science University, Portland, OR 97239, USA
| | - Jane Thanner
- Information Technology Group, Oregon Health & Science University, Portland, OR 97201, USA
| | - Arpita Kulkarni
- Knight Diagnostic Laboratories, Oregon Health & Science University, Portland, OR 97239, USA
| | - Rossana Wong
- Knight Diagnostic Laboratories, Oregon Health & Science University, Portland, OR 97239, USA
| | - Samuel Medica
- Integrated Genomics Laboratory, Oregon Health & Science University, Portland, OR 97239, USA
| | - Tiana Week
- Integrated Genomics Laboratory, Oregon Health & Science University, Portland, OR 97239, USA
| | - Jacob Buitrago
- Integrated Genomics Laboratory, Oregon Health & Science University, Portland, OR 97239, USA
| | - Aaron Larson
- Integrated Genomics Laboratory, Oregon Health & Science University, Portland, OR 97239, USA
| | - Katie Johnson Camacho
- Cancer Early Detection Advanced Research Center, Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97201, USA
| | - Kim Brown
- Knight Cancer Institute, Community Outreach and Engagement, Oregon Health & Science University, Portland, OR 97201, USA
| | - Rachel Crist
- Knight Cancer Institute, Community Outreach and Engagement, Oregon Health & Science University, Portland, OR 97201, USA
| | - Casey Conrad
- Cancer Early Detection Advanced Research Center, Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97201, USA
| | - Sara Evans-Dutson
- Cancer Early Detection Advanced Research Center, Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97201, USA
| | - Ryan Lutz
- Knight Cancer Institute, Community Outreach and Engagement, Oregon Health & Science University, Portland, OR 97201, USA
| | - Asia Mitchell
- Cancer Early Detection Advanced Research Center, Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97201, USA
| | - Pavana Anur
- Cancer Early Detection Advanced Research Center, Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97201, USA
| | - Vanessa Serrato
- Knight Cancer Institute, Community Outreach and Engagement, Oregon Health & Science University, Portland, OR 97201, USA
| | - Autumn Shafer
- University of Oregon, School of Journalism and Communication, Portland, OR 97209, USA
| | | | - K J Hamman
- Department of Molecular & Medical Genetics, Oregon Health & Science University, Portland, OR 97239, USA
| | - Amelia Mulford
- Department of Molecular & Medical Genetics, Oregon Health & Science University, Portland, OR 97239, USA
| | - Wojciech Wiszniewski
- Department of Molecular & Medical Genetics, Oregon Health & Science University, Portland, OR 97239, USA
| | - Jone E Sampson
- Department of Molecular & Medical Genetics, Oregon Health & Science University, Portland, OR 97239, USA
| | - Andrew Adey
- Cancer Early Detection Advanced Research Center, Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97201, USA; Department of Molecular & Medical Genetics, Oregon Health & Science University, Portland, OR 97239, USA
| | - Brian J O'Roak
- Department of Molecular & Medical Genetics, Oregon Health & Science University, Portland, OR 97239, USA
| | - Christina A Harrington
- Integrated Genomics Laboratory, Oregon Health & Science University, Portland, OR 97239, USA; Department of Molecular & Medical Genetics, Oregon Health & Science University, Portland, OR 97239, USA
| | - Jackilen Shannon
- Knight Cancer Institute, Community Outreach and Engagement, Oregon Health & Science University, Portland, OR 97201, USA; Division of Oncological Sciences, Oregon Health & Science University, Portland, OR 97239, USA
| | - Paul T Spellman
- Cancer Early Detection Advanced Research Center, Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97201, USA; Department of Molecular & Medical Genetics, Oregon Health & Science University, Portland, OR 97239, USA
| | - C Sue Richards
- Knight Diagnostic Laboratories, Oregon Health & Science University, Portland, OR 97239, USA; Department of Molecular & Medical Genetics, Oregon Health & Science University, Portland, OR 97239, USA.
| |
Collapse
|
16
|
Mitchell OD, Gilliam K, del Gaudio D, McNeely KE, Smith S, Acevedo M, Gaduraju M, Hodge R, Ramsland ASS, Segal J, Das S, Hathaway F, Bryan DS, Tawde S, Galasinski S, Wang P, Tjota MY, Husain AN, Armato SG, Donington J, Ferguson MK, Turaga K, Churpek JE, Kindler HL, Drazer MW. Germline Variants Incidentally Detected via Tumor-Only Genomic Profiling of Patients With Mesothelioma. JAMA Netw Open 2023; 6:e2327351. [PMID: 37556141 PMCID: PMC10413174 DOI: 10.1001/jamanetworkopen.2023.27351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 06/08/2023] [Indexed: 08/10/2023] Open
Abstract
IMPORTANCE Patients with mesothelioma often have next-generation sequencing (NGS) of their tumor performed; tumor-only NGS may incidentally identify germline pathogenic or likely pathogenic (P/LP) variants despite not being designed for this purpose. It is unknown how frequently patients with mesothelioma have germline P/LP variants incidentally detected via tumor-only NGS. OBJECTIVE To determine the prevalence of incidental germline P/LP variants detected via tumor-only NGS of mesothelioma. DESIGN, SETTING, AND PARTICIPANTS A series of 161 unrelated patients with mesothelioma from a high-volume mesothelioma program had tumor-only and germline NGS performed during April 2016 to October 2021. Follow-up ranged from 18 months to 7 years. Tumor and germline assays were compared to determine which P/LP variants identified via tumor-only NGS were of germline origin. Data were analyzed from January to March 2023. MAIN OUTCOMES AND MEASURES The proportion of patients with mesothelioma who had P/LP germline variants incidentally detected via tumor-only NGS. RESULTS Of 161 patients with mesothelioma, 105 were male (65%), the mean (SD) age was 64.7 (11.2) years, and 156 patients (97%) self-identified as non-Hispanic White. Most (126 patients [78%]) had at least 1 potentially incidental P/LP germline variant. The positive predictive value of a potentially incidental germline P/LP variant on tumor-only NGS was 20%. Overall, 26 patients (16%) carried a P/LP germline variant. Germline P/LP variants were identified in ATM, ATR, BAP1, CHEK2, DDX41, FANCM, HAX1, MRE11A, MSH6, MUTYH, NF1, SAMD9L, and TMEM127. CONCLUSIONS AND RELEVANCE In this case series of 161 patients with mesothelioma, 16% had confirmed germline P/LP variants. Given the implications of a hereditary cancer syndrome diagnosis for preventive care and familial counseling, clinical approaches for addressing incidental P/LP germline variants in tumor-only NGS are needed. Tumor-only sequencing should not replace dedicated germline testing. Universal germline testing is likely needed for patients with mesothelioma.
Collapse
Affiliation(s)
- Owen D. Mitchell
- Section of Hematology/Oncology, Department of Medicine, The University of Chicago, Illinois
| | - Katie Gilliam
- Section of Hematology/Oncology, Department of Medicine, The University of Chicago, Illinois
| | | | - Kelsey E. McNeely
- Section of Hematology/Oncology, Department of Medicine, The University of Chicago, Illinois
| | - Shaili Smith
- Section of Hematology/Oncology, Department of Medicine, The University of Chicago, Illinois
| | - Maria Acevedo
- Section of Hematology/Oncology, Department of Medicine, The University of Chicago, Illinois
| | - Meghana Gaduraju
- Section of Hematology/Oncology, Department of Medicine, The University of Chicago, Illinois
| | - Rachel Hodge
- Section of Hematology/Oncology, Department of Medicine, The University of Chicago, Illinois
| | | | - Jeremy Segal
- Department of Pathology, The University of Chicago, Illinois
| | - Soma Das
- Department of Human Genetics, The University of Chicago, Illinois
| | - Feighanne Hathaway
- Section of Hematology/Oncology, Department of Medicine, The University of Chicago, Illinois
| | | | - Sanjukta Tawde
- Department of Human Genetics, The University of Chicago, Illinois
| | | | - Peng Wang
- Department of Pathology, The University of Chicago, Illinois
| | | | - Aliya N. Husain
- Department of Pathology, The University of Chicago, Illinois
| | | | | | | | - Kiran Turaga
- Department of Surgery, The University of Chicago, Illinois
| | - Jane E. Churpek
- Division of Hematology, Medical Oncology, and Palliative Care, Department of Medicine, University of Wisconsin, Madison
| | - Hedy L. Kindler
- Section of Hematology/Oncology, Department of Medicine, The University of Chicago, Illinois
| | - Michael W. Drazer
- Section of Hematology/Oncology, Department of Medicine, The University of Chicago, Illinois
| |
Collapse
|
17
|
Myint KZY, Shimabuku M, Horio R, Kaneda M, Shimizu Y, Taguchi J. Identification of circulating tumour DNA (ctDNA) from the liquid biopsy results: Findings from an observational cohort study. Cancer Treat Res Commun 2023; 35:100701. [PMID: 37094468 DOI: 10.1016/j.ctarc.2023.100701] [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: 06/22/2022] [Revised: 03/02/2023] [Accepted: 03/24/2023] [Indexed: 04/26/2023]
Abstract
Liquid biopsies can be a rapid, cost-effective and non-invasive alternative to tumour biopsies for detecting genetic mutations in somatic tumours. Genetic profiling of liquid biopsies can also be used to identify novel antigens for targeted therapy, provide updated information on disease prognosis and evaluate treatment efficacy. In this study, we aimed to examine mutations that could be identified in liquid biopsy and their distribution in a small study cohort. We studied the genomic profiles of 99 blood samples from 85 patients with 21 different types of cancer using two commercially available liquid biopsy tests. The mean circulating free DNA (cfDNA) concentration was 162.7 ± 352.3 nanograms per 20 millilitres. Amongst cfDNA, the circulating tumour DNA (ctDNA) percentage ranged from 0.006% to 90.6%. With the exception of samples with gene amplification and high microsatellite instability, the number of mutations in each sample varied from zero to 21, with an average of 5.6 mutations in each patient. Amongst these mutations, nonsynonymous mutations were the most frequently observed type of mutation (90% of the sample, with an average frequency of 3.6 mutations per patient). Mutations were observed in 76 different genes. TP53 mutations constituted more than 16% of the detectable mutations, especially in non-small cell lung cancer. All the tumour types, except the ovary, kidney and apocrine gland tumours, harboured at least one type of TP53 mutation. KRAS (mainly in pancreatic cancer) and PIK3CA (mostly in breast cancer) mutations, were responsible for an additional 10% of the mutations in the studied samples. The tumour mutations were specific to each patient, as approximately 94.7% of the mutations were so unique that there was almost no duplication amongst the patients. These findings indicate that liquid biopsy can detect specific molecular changes of tumour, which is useful for precision oncology and personalized cancer treatment.
Collapse
Affiliation(s)
- Khin Zay Yar Myint
- Centre for Advanced Medical Science and Technology, Tokyo Midtown Medical Center, Japan
| | - Masamori Shimabuku
- Centre for Advanced Medical Science and Technology, Tokyo Midtown Medical Center, Japan
| | - Ruriko Horio
- Centre for Advanced Medical Science and Technology, Tokyo Midtown Medical Center, Japan
| | - Munehisa Kaneda
- Centre for Advanced Medical Science and Technology, Tokyo Midtown Medical Center, Japan
| | - Yoko Shimizu
- Centre for Advanced Medical Science and Technology, Tokyo Midtown Medical Center, Japan
| | - Junichi Taguchi
- Centre for Advanced Medical Science and Technology, Tokyo Midtown Medical Center, Japan.
| |
Collapse
|
18
|
Karpel H, Manderski E, Pothuri B. Frequency of actionable germline pathogenic variants identified through tumor next-generation sequencing in a gynecologic cancer cohort. Int J Gynecol Cancer 2023:ijgc-2022-004142. [PMID: 36889816 DOI: 10.1136/ijgc-2022-004142] [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: 03/10/2023] Open
Abstract
BACKGROUND Tumor next-generation sequencing can identify potential germline pathogenic variants associated with cancer susceptibility. OBJECTIVE To describe the frequency of tumor sequencing results that met European Society of Medical Oncology (ESMO) recommendations for further germline genetic testing, and the frequency of germline variants among a cohort with gynecologic cancer. METHODS Patients with gynecologic cancer who underwent tumor sequencing between September 2019 and February 2022 in a large healthcare system in New York City were retrospectively identified. Eligible patients with suspected germline pathogenic variants on tumor sequencing were identified based on ESMO guidelines. Logistic regression was used to explore variables associated with referral and completion of germline testing. RESULTS Of 358 patients with gynecologic cancers who underwent tumor sequencing, 81 (22.6%) had ≥1 suspected germline variant according to ESMO guidelines. Of the 81 patients with qualifying tumor sequencing results, 56 (69.1%) received germline testing: 41/46 (89.1%) eligible patients with ovarian cancer and 15/33 (45.5%) with endometrial cancer. In the endometrial cancer cohort, 11/33 (33.3%) eligible patients were not referred for germline testing and the majority of these patients had tumor variants in genes commonly known to cause hereditary cancer. Of the 56 patients who underwent germline testing, 40 (71.4%) had pathogenic germline variants. In multivariable analysis, race/ethnicity other than non-Hispanic white was associated with lower odds of germline testing referral and completion (OR=0.1, 95% CI 0.01 to 0.5 and OR=0.2, 95% CI 0.04 to 0.6, respectively). CONCLUSION Given the high rate of pathogenic germline variant detection and the importance of identifying such variants for both patients and their family, it is imperative that eligible patients undergo germline testing. Additional education for providers on multidisciplinary guidelines and development of clinical pathways to ensure germline testing of suspected pathogenic variants identified on tumor sequencing is warranted, especially in light of the racial/ethnic inequity observed.
Collapse
Affiliation(s)
- Hannah Karpel
- New York University Grossman School of Medicine, New York, New York, USA
| | - Elizabeth Manderski
- Perlmutter Cancer Center, New York University Langone Health, New York, New York, USA
| | - Bhavana Pothuri
- Department of Obstetrics and Gynecology, Perlmutter Cancer Center, New York University Langone Health, New York, New York, USA
| |
Collapse
|
19
|
Kuzbari Z, Bandlamudi C, Loveday C, Garrett A, Mehine M, George A, Hanson H, Snape K, Kulkarni A, Allen S, Jezdic S, Ferrandino R, Westphalen CB, Castro E, Rodon J, Mateo J, Burghel GJ, Berger MF, Mandelker D, Turnbull C. Germline-focused analysis of tumour-detected variants in 49,264 cancer patients: ESMO Precision Medicine Working Group recommendations. Ann Oncol 2023; 34:215-227. [PMID: 36529447 DOI: 10.1016/j.annonc.2022.12.003] [Citation(s) in RCA: 38] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 12/01/2022] [Accepted: 12/11/2022] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND The European Society for Medical Oncology Precision Medicine Working Group (ESMO PMWG) was reconvened to update its 2018/19 recommendations on follow-up of putative germline variants detected on tumour-only sequencing, which were based on an analysis of 17 152 cancers. METHODS We analysed an expanded dataset including 49 264 paired tumour-normal samples. We applied filters to tumour-detected variants based on variant allele frequency, predicted pathogenicity and population variant frequency. For 58 cancer-susceptibility genes, we then examined the proportion of filtered tumour-detected variants of true germline origin [germline conversion rate (GCR)]. We conducted subanalyses based on the age of cancer diagnosis, specific tumour types and 'on-tumour' status (established tumour-gene association). RESULTS Analysis of 45 472 nonhypermutated solid malignancy tumour samples yielded 21 351 filtered tumour-detected variants of which 3515 were of true germline origin. 3.1% of true germline pathogenic variants were absent from the filtered tumour-detected variants. For genes such as BRCA1, BRCA2 and PALB2, the GCR in filtered tumour-detected variants was >80%; conversely for TP53, APC and STK11 this GCR was <2%. CONCLUSION Strategic germline-focused analysis can prioritise a subset of tumour-detected variants for which germline follow-up will produce the highest yield of most actionable true germline variants. We present updated recommendations around germline follow-up of tumour-only sequencing including (i) revision to 5% for the minimum per-gene GCR, (ii) inclusion of actionable intermediate penetrance genes ATM and CHEK2, (iii) definition of a set of seven 'most actionable' cancer-susceptibility genes (BRCA1, BRCA2, PALB2, MLH1, MSH2, MSH6 and RET) in which germline follow-up is recommended regardless of tumour type.
Collapse
Affiliation(s)
- Z Kuzbari
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London, UK
| | - C Bandlamudi
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, USA
| | - C Loveday
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London, UK. https://twitter.com/LovedayChey
| | - A Garrett
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London, UK. https://twitter.com/DrAliceGarrett
| | - M Mehine
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, USA
| | - A George
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London, UK; The Royal Marsden NHS Foundation Trust, London, UK
| | - H Hanson
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London, UK; South West Thames Regional Genetics Service, St George's University Hospitals NHS Foundation Trust, London, UK
| | - K Snape
- South West Thames Regional Genetics Service, St George's University Hospitals NHS Foundation Trust, London, UK. https://twitter.com/genetikos
| | - A Kulkarni
- South East Thames Regional Genetics Service, Guy's and St Thomas' NHS Foundation Trust, London, UK. https://twitter.com/Anju__Kulkarni
| | - S Allen
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London, UK
| | - S Jezdic
- Scientific and Medical Division, European Society for Medical Oncology, Lugano, Switzerland
| | - R Ferrandino
- Scientific and Medical Division, European Society for Medical Oncology, Lugano, Switzerland
| | - C B Westphalen
- Department of Medicine III and Comprehensive Cancer Center (CCC Munich LMU) University Hospital, LMU Munich, Munich, Germany
| | - E Castro
- Genitourinary Cancers Translational Research Group, Institute of Biomedical Research in Málaga (IBIMA), Málaga, Spain. https://twitter.com/Ecastromarcos
| | - J Rodon
- Investigational Cancer Therapeutics, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - J Mateo
- Vall d'Hebron Institute of Oncology (VHIO), Barcelona; Vall d'Hebron University Hospital, Barcelona, Spain
| | - G J Burghel
- North West Genomic Laboratory Hub, Manchester University NHS Foundation Trust, Manchester, UK. https://twitter.com/BurghelG
| | - M F Berger
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, USA
| | - D Mandelker
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, USA
| | - C Turnbull
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London, UK; The Royal Marsden NHS Foundation Trust, London, UK.
| |
Collapse
|
20
|
Li MM, Cottrell CE, Pullambhatla M, Roy S, Temple-Smolkin RL, Turner SA, Wang K, Zhou Y, Vnencak-Jones CL. Assessments of Somatic Variant Classification Using the Association for Molecular Pathology/American Society of Clinical Oncology/College of American Pathologists Guidelines: A Report from the Association for Molecular Pathology. J Mol Diagn 2023; 25:69-86. [PMID: 36503149 DOI: 10.1016/j.jmoldx.2022.11.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 11/09/2022] [Accepted: 11/18/2022] [Indexed: 12/13/2022] Open
Abstract
To assess the clinical implementation of the 2017 Standards and Guidelines for the Interpretation and Reporting of Sequence Variants in Cancer: A Joint Consensus Recommendation of the Association for Molecular Pathology, American Society of Clinical Oncology, and College of American Pathologists, identify content that may result in classification inconsistencies, and evaluate implementation barriers, an Association for Molecular Pathology Working Group conducted variant interpretation challenges and a guideline implementation survey. A total of 134 participants participated in the variant interpretation challenges, consisting of 11 variants in four cancer cases. Results demonstrate 86% (range, 54% to 94%) of the respondents correctly classified clinically significant variants, variants of uncertain significance, and benign/likely benign variants; however, only 59% (range, 39% to 84%) of responses agreed with the working group's consensus intended responses regarding both tiers and categories of clinical significance. In the implementation survey, 71% (157/220) of respondents have implemented the 2017 guidelines for variant classification and reporting either with or without modifications. Collectively, this study demonstrates that, although they may not yet be optimized, the 2017 guideline recommendations are being adopted for standardized somatic variant classification. The working group identified significant areas for future guideline improvement, including the need for a more granular and comprehensive classification system and education resources to meet the growing needs of both laboratory professionals and medical oncologists.
Collapse
Affiliation(s)
- Marilyn M Li
- The Variant Interpretation Testing Across Laboratories (VITAL) Somatic Working Group of the Clinical Practice Committee, Association for Molecular Pathology, Rockville, Maryland; Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania.
| | - Catherine E Cottrell
- The Variant Interpretation Testing Across Laboratories (VITAL) Somatic Working Group of the Clinical Practice Committee, Association for Molecular Pathology, Rockville, Maryland; Steve and Cindy Rasmussen Institute for Genomic Medicine, Nationwide Children's Hospital, Columbus, Ohio; Department of Pathology, The Ohio State University College of Medicine, Columbus, Ohio
| | | | - Somak Roy
- The Variant Interpretation Testing Across Laboratories (VITAL) Somatic Working Group of the Clinical Practice Committee, Association for Molecular Pathology, Rockville, Maryland; Department of Pathology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | | | - Scott A Turner
- The Variant Interpretation Testing Across Laboratories (VITAL) Somatic Working Group of the Clinical Practice Committee, Association for Molecular Pathology, Rockville, Maryland; Department of Pathology, Virginia Commonwealth University, Richmond, Virginia
| | - Kai Wang
- The Variant Interpretation Testing Across Laboratories (VITAL) Somatic Working Group of the Clinical Practice Committee, Association for Molecular Pathology, Rockville, Maryland; Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania; Raymond G. Perelman Center for Cellular and Molecular Therapeutics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Yunyun Zhou
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Cindy L Vnencak-Jones
- The Variant Interpretation Testing Across Laboratories (VITAL) Somatic Working Group of the Clinical Practice Committee, Association for Molecular Pathology, Rockville, Maryland; Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee
| |
Collapse
|
21
|
Truong H, Breen K, Nandakumar S, Sjoberg DD, Kemel Y, Mehta N, Lenis AT, Reisz PA, Carruthers J, Benfante N, Joseph V, Khurram A, Gopalan A, Fine SW, Reuter VE, Vickers AJ, Birsoy O, Liu Y, Walsh M, Latham A, Mandelker D, Stadler ZK, Pietzak E, Ehdaie B, Touijer KA, Laudone VP, Slovin SF, Autio KA, Danila DC, Rathkopf DE, Eastham JA, Chen Y, Morris MJ, Offit K, Solit DB, Scher HI, Abida W, Robson ME, Carlo MI. Gene-based Confirmatory Germline Testing Following Tumor-only Sequencing of Prostate Cancer. Eur Urol 2023; 83:29-38. [PMID: 36115772 PMCID: PMC10208030 DOI: 10.1016/j.eururo.2022.08.028] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 08/17/2022] [Accepted: 08/24/2022] [Indexed: 12/14/2022]
Abstract
BACKGROUND Tumor-only genomic profiling is an important tool in therapeutic management of men with prostate cancer. Since clinically actionable germline variants may be reflected in tumor profiling, it is critical to identify which variants have a higher risk of being germline in origin to better counsel patients and prioritize genetic testing. OBJECTIVE To determine when variants found on tumor-only sequencing of prostate cancers should prompt confirmatory germline testing. DESIGN, SETTING, AND PARTICIPANTS Men with prostate cancer who underwent both tumor and germline sequencing at Memorial Sloan Kettering Cancer Center from January 1, 2015 to January 31, 2020 were evaluated. OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS Tumor and germline profiles were analyzed for pathogenic and likely pathogenic ("pathogenic") variants in 60 moderate- or high-penetrance genes associated with cancer predisposition. The germline probability (germline/germline + somatic) of a variant was calculated for each gene. Clinical and pathologic factors were analyzed as potential modifiers of germline probability. RESULTS AND LIMITATIONS Of the 1883 patients identified, 1084 (58%) had a somatic or germline pathogenic variant in one of 60 cancer susceptibility genes, and of them, 240 (22%) had at least one germline variant. Overall, the most frequent variants were in TP53, PTEN, APC, BRCA2, RB1, ATM, and CHEK2. Variants in TP53, PTEN, or RB1 were identified in 746 (40%) patients and were exclusively somatic. Variants with the highest germline probabilities were in PALB2 (69%), MITF (62%), HOXB13 (60%), CHEK2 (55%), BRCA1 (55%), and BRCA2 (47%), and the overall germline probability of a variant in any DNA damage repair gene was 40%. Limitations were that most of the men included in the cohort had metastatic disease, and different thresholds for pathogenicity exist for somatic and germline variants. CONCLUSIONS Of patients with pathogenic variants found on prostate tumor sequencing, 22% had clinically actionable germline variants, for which the germline probabilities varied widely by gene. Our results provide an evidenced-based clinical framework to prioritize referral to genetic counseling following tumor-only sequencing. PATIENT SUMMARY Patients with advanced prostate cancer are recommended to have germline genetic testing. Genetic sequencing of a patient's prostate tumor may also identify certain gene variants that are inherited. We found that patients who had variants in certain genes, such as ones that function in DNA damage repair, identified in their prostate tumor sequencing, had a high risk for having an inherited cancer syndrome.
Collapse
Affiliation(s)
- Hong Truong
- Department of Surgery, Urology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Kelsey Breen
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Subhiksha Nandakumar
- Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Daniel D Sjoberg
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Yelena Kemel
- Niehaus Center for Inherited Cancer Genomics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Nikita Mehta
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Andrew T Lenis
- Department of Surgery, Urology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Peter A Reisz
- Department of Surgery, Urology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jessica Carruthers
- Department of Surgery, Urology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Nicole Benfante
- Department of Surgery, Urology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Vijai Joseph
- Niehaus Center for Inherited Cancer Genomics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Aliya Khurram
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Anuradha Gopalan
- Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Samson W Fine
- Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Victor E Reuter
- Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Andrew J Vickers
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ozge Birsoy
- Niehaus Center for Inherited Cancer Genomics, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ying Liu
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Michael Walsh
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Niehaus Center for Inherited Cancer Genomics, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Alicia Latham
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Diana Mandelker
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Zsofia K Stadler
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Niehaus Center for Inherited Cancer Genomics, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Medicine, Gastrointestinal Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Eugene Pietzak
- Department of Surgery, Urology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Behfar Ehdaie
- Department of Surgery, Urology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Karim A Touijer
- Department of Surgery, Urology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Vincent P Laudone
- Department of Surgery, Urology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Susan F Slovin
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Karen A Autio
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Daniel C Danila
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Dana E Rathkopf
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - James A Eastham
- Department of Surgery, Urology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Yu Chen
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Michael J Morris
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Kenneth Offit
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Niehaus Center for Inherited Cancer Genomics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - David B Solit
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Howard I Scher
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Wassim Abida
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Mark E Robson
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Niehaus Center for Inherited Cancer Genomics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Maria I Carlo
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Niehaus Center for Inherited Cancer Genomics, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
22
|
Senter L, Veney D, Surplus T, Haynam M, Adams EJ, Hampel H, Toland AE, Presley CJ, Padamsee TJ, Lee CN, Hovick SR, Stover DG. Patient Understanding of Tumor Genomic Testing: A Quality Improvement Effort. JCO Oncol Pract 2023; 19:e8-e14. [PMID: 36130146 PMCID: PMC10166357 DOI: 10.1200/op.22.00316] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 06/29/2022] [Accepted: 08/16/2022] [Indexed: 01/14/2023] Open
Abstract
PURPOSE Tumor genomic testing (TGT) has become increasingly adopted as part of standard cancer care for many cancers. Despite national guidelines around patient education before TGT, available evidence suggests that most patients' understanding of genomics remains limited, particularly lower-income and minority patients, and most patients are not informed regarding potential incidental germline findings. METHODS To investigate and address limitations in patient understanding of TGT results, a Plan-Do-Study-Act (PDSA) approach is being used to assess needs, identify opportunities for improvement, and implement approaches to optimize patient education. We reviewed published guidelines related to pre-TGT provider-patient education and to identify key points (Plan). A provider quality improvement survey was completed (Do), which highlighted inconsistency in pre-TGT discussion practice across providers and minimal discussion with patients regarding the possibility of incidental germline findings. RESULTS Patient focus groups and interviews (N = 12 patients) were completed with coding of each transcript (Study), which revealed themes including trouble differentiating TGT from other forms of testing, yet understanding that results could tailor therapy. The integration of data across this initial PDSA cycle identified consistent themes and opportunities, which were incorporated into a patient-directed, concise animated video for pre-TGT education (Act), which will form the foundation of a subsequent PDSA cycle. The video addresses how TGT may/may not inform treatment, the process for TGT using existing tissue or liquid biopsy, insurance coverage, and the potential need for germline genetics follow-up because of incidental findings. CONCLUSION This PDSA cycle reveals key gaps and opportunities for improvement in patient education before TGT.
Collapse
Affiliation(s)
- Leigha Senter
- Division of Human Genetics, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, OH
| | - Deloris Veney
- Division of Medical Oncology, Ohio State University Comprehensive Cancer Center, Columbus, OH
| | - Taylor Surplus
- School of Communication, Ohio State University, Columbus, OH
| | - Marcy Haynam
- Division of Medical Oncology, Ohio State University Comprehensive Cancer Center, Columbus, OH
| | - Elizabeth J. Adams
- Division of Medical Oncology, Ohio State University Comprehensive Cancer Center, Columbus, OH
| | - Heather Hampel
- Division of Human Genetics, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, OH
- Division of Clinical Cancer Genomics, Department of Medical Oncology and Therapeutics Research, City of Hope National Medical Center, Duarte, CA
| | - Amanda E. Toland
- Division of Human Genetics, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, OH
- Department of Cancer Biology and Genetics, The Ohio State University, Columbus, OH
| | - Carolyn J. Presley
- Division of Medical Oncology, Ohio State University Comprehensive Cancer Center, Columbus, OH
| | - Tasleem J. Padamsee
- Division of Health Services Management and Policy, College of Public Health, The Ohio State University, Columbus, OH
| | - Clara N. Lee
- Recruitment, Intervention and Survey Shared Resource, Ohio State University Comprehensive Cancer Center, Columbus, OH
| | | | - Daniel G. Stover
- Division of Medical Oncology, Ohio State University Comprehensive Cancer Center, Columbus, OH
- Department of Biomedical Informatics, Ohio State University, Columbus, OH
- Pelotonia Institute for Immuno-Oncology, Ohio State University Comprehensive Cancer, Columbus, OH
| |
Collapse
|
23
|
Bell KA, Kim R, Aronson M, Gillies B, Ali Awan A, Chun K, Hart J, Healey R, Kim L, Klaric G, Panabaker K, Sabatini PJB, Sadikovic B, Selvarajah S, Smith AC, Stockley TL, Vaags AK, Eisen A, Pollett A, Feilotter H. Development of a comprehensive approach to adult hereditary cancer testing in Ontario. J Med Genet 2022:jmg-2022-108945. [PMID: 36564171 DOI: 10.1136/jmg-2022-108945] [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: 09/16/2022] [Accepted: 12/10/2022] [Indexed: 12/25/2022]
Abstract
BACKGROUND Genetic testing for hereditary cancer susceptibility has advanced over time due to the discovery of new risk genes, improved technology and decreased cost. In the province of Ontario, testing eligibility criteria were initially developed to include hereditary breast, ovarian and colorectal cancer syndromes. The rapid evolution of genetic technologies has facilitated the ability to interrogate a large number of genes concurrently. This, coupled with new knowledge about risk genes, necessitated a coordinated approach to expanding the scope of genes and indications tested and synchronisation of access and test utilisation across the province as required in a publicly funded universal healthcare system. METHODS Ontario Health-Cancer Care Ontario convened expert working groups to develop a standardised and comprehensive cancer gene list for adults and accompanying hereditary cancer testing (HCT) criteria using an evidence-based framework and broad laboratory and clinical genetics engagement. RESULTS A standardised 76-cancer-gene panel, organised into 13 larger disease site panels and 25 single/small gene panels, was developed and endorsed by the working groups. Provincial genetic testing eligibility criteria were updated to align with the new panels and to guide clinical decision-making. In the first year following the implementation of these changes, 10 564 HCT panels were performed with an overall mutation detection rate of 12.2%. CONCLUSION Using an evidence framework and broad clinical engagement to develop and endorse an updated guidance document, cancer genetic testing for adults in Ontario is now standardised and coordinated across the province.
Collapse
Affiliation(s)
| | - Raymond Kim
- Division of Medical Oncology and Hematology, University Health Network, Toronto, Ontario, Canada.,Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Melyssa Aronson
- Zane Cohen Centre for Digestive Diseases, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Brittany Gillies
- Familial Cancer Clinic, Princess Margaret Hospital Cancer Centre, Toronto, Ontario, Canada
| | - Arif Ali Awan
- Division of Medical Oncology, Ottawa Hospital Cancer Centre, Ottawa, Ontario, Canada
| | - Kathy Chun
- The Hospital for Sick Children, Toronto, Ontario, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Jennifer Hart
- Pathology and Laboratory Medicine Program, Ontario Health, Toronto, Ontario, Canada
| | - Rachel Healey
- Pathology and Laboratory Medicine Program, Ontario Health, Toronto, Ontario, Canada
| | - Linda Kim
- Department of Laboratory Medicine and Genetics, Credit Valley Hospital Site, Mississauga, Ontario, Canada
| | - Goran Klaric
- Pathology and Laboratory Medicine Program, Ontario Health, Toronto, Ontario, Canada
| | - Karen Panabaker
- Medical Genetics Program of Southwestern Ontario, London Health Sciences Centre, London, Ontario, Canada
| | | | - Bekim Sadikovic
- Department of Pathology and Laboratory Medicine, Western University, London, Ontario, Canada.,Verspeeten Clinical Genome Centre, Western University, London, Ontario, Canada
| | - Shamini Selvarajah
- Department of Clinical Laboratory Genetics, University Health Network, Toronto, Ontario, Canada
| | | | - Tracy L Stockley
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada.,Department of Clinical Laboratory Genetics, Laboratory Medicine Program, University Health Network, Toronto, Ontario, Canada
| | - Andrea K Vaags
- Laboratory Medicine and Genetics, Trillium Health Partners, Mississauga, Ontario, Canada
| | - Andrea Eisen
- Odette Cancer Centre, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
| | - Aaron Pollett
- Pathology and Laboratory Medicine Program, Ontario Health, Toronto, Ontario, Canada.,Division of Diagnostic Medical Genetics, Sinai Health System, Toronto, Ontario, Canada
| | - Harriet Feilotter
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, Ontario, Canada
| |
Collapse
|
24
|
Lee SH, Vadlamudi C, Zhao Q, Yilmaz O, Cerda S. An institutional experience with DICER1 mutated thyroid nodules-evaluating the cytomorphology and molecular phenotype. J Am Soc Cytopathol 2022; 11:335-344. [PMID: 35934646 DOI: 10.1016/j.jasc.2022.07.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 06/27/2022] [Accepted: 07/06/2022] [Indexed: 06/15/2023]
Abstract
INTRODUCTION DICER1 mutated thyroid nodules are commonly seen in pediatric populations often, as part of DICER1 syndrome. We seek to evaluate DICER1 mutated thyroid nodules in adult populations to assess whether there exists distinctive clinical, cytologic, histologic, and molecular characteristics that underline our institutional cohort. MATERIALS AND METHODS Retrospective analysis was performed on all fine-needle aspiration (FNA) specimens with a corresponding ThyroSeq panel, to select a cohort of cases with DICER1 mutations. Clinical, radiologic, and cytology materials were reviewed, and histology was reviewed for corresponding resection cases were available. ThyroSeq panel was further scrutinized for additional molecular alterations and variant allele frequency. RESULTS DICER1 mutated thyroid nodules (n = 8), more commonly occurred in younger adults (P = 0.01) with larger (P = 0.01) nodules and only in female patients in our cohort. FNA commonly demonstrates cellular specimens with banal cytomorphologic cues including regular nuclei, inconspicuous nucleoli, smooth nuclear membranes, and abundant colloid. On retrospective review by 2 cytopathologists, the lesions were frequently diagnosed as Bethesda II (5 of 8) by both reviewers. Histology, when available, showed that all nodules were categorized as follicular adenomas (5 of 5), often demonstrating macrofollicles with papillary excrescences demonstrating bland nuclei (4 of 5). DICER1 mutational profile revealed a variant allele frequency of >40% in 25% of cases (2 of 8) and >30% in an additional 4 cases, highlighting a possible germline association. CONCLUSIONS DICER1 mutated nodules may be under-reported due to banal cytomorphologic features and may be associated with an underlying germline alteration.
Collapse
Affiliation(s)
- Soo Hyun Lee
- Department of Pathology, Boston University Medical Center, Boston, Massachusetts
| | - Charitha Vadlamudi
- Department of Pathology, Boston University Medical Center, Boston, Massachusetts
| | - Qing Zhao
- Department of Pathology, Boston University Medical Center, Boston, Massachusetts
| | - Osman Yilmaz
- Department of Pathology, Boston University Medical Center, Boston, Massachusetts.
| | - Sandra Cerda
- Department of Pathology, Boston University Medical Center, Boston, Massachusetts.
| |
Collapse
|
25
|
Somatic tumor mutations in moderate risk cancer genes: Targets for germline confirmatory testing. Cancer Genet 2022; 268-269:22-27. [PMID: 36116289 DOI: 10.1016/j.cancergen.2022.09.001] [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: 05/31/2022] [Revised: 08/11/2022] [Accepted: 09/01/2022] [Indexed: 01/25/2023]
Abstract
BACKGROUND Recent changes in oncology practice guidelines indicate that mutations in cancer susceptibility genes identified on tumor genomic profiling (TGP) should prompt confirmatory germline testing. Our study aimed to determine the proportion of patients with TGP-identified mutations in moderate risk breast and ovarian cancer genes who previously would not have been considered for germline testing. METHODS From January 2013 to September 2020, 7468 adult Stanford Health Care patients underwent TGP on solid tumor samples and 166 had TGP-identified mutations in moderate risk breast and ovarian cancer susceptibility genes (ATM, BRIP1, CHEK2, PALB2, RAD51C and RAD51D). Retrospective chart reviews were performed on 160 patients. Cases were analyzed to determine eligibility for germline testing using established NCCN criteria, and somatic and germline results were compared where both were available. RESULTS Nearly half (45.3% [73/160]) of patients would not have been eligible for germline testing if not for a TGP-identified mutation in a moderate risk breast or ovarian cancer gene. Of the 64 cases that underwent germline testing, about half (51.5% [33/64]) had results that confirmed germline origin of the TGP finding. High rates of germline confirmation were found in PALB2 (100% [5/5]), ATM (40% [14/35]), CHEK2 (61.5% [8/13]), and BRIP1 (57.1% [4/7]). CONCLUSION Our study shows that the presence of TGP-identified mutations in moderate risk breast and ovarian cancer genes increases eligibility for germline testing beyond those that would be eligible based largely on personal and family history criteria alone. Additionally, results of germline testing in these newly eligible cases supports that this expanded eligibility captures individuals with hereditary cancer syndromes that would not have otherwise been identified.
Collapse
|
26
|
Harriman JW, Espinel WF, Vagher J, Gammon A. BRCA1/2 Variants Identified Through Tumor Genomic Profiling: Assessing Genetic Counseling Outcomes. JCO Precis Oncol 2022; 6:e2100375. [DOI: 10.1200/po.21.00375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
PURPOSE As tumor genomic profiling (TGP) is increasingly used to help guide cancer treatment, BRCA variants, which may or may not be reflective of the germline genome, are being identified. As TGP use increases, it is becoming an important tool for referral to genetic counseling and identifying patients with hereditary cancer syndromes such as hereditary breast and ovarian cancer. This study explores genetic counseling referral patterns and germline implications of patients found to have pathogenic BRCA1/2 variants identified through TGP. METHODS Participants include patients at Huntsman Cancer Institute undergoing TGP through a single commercial laboratory between March 2014 and July 2018. A retrospective chart review was conducted for 62 patients found to have pathogenic variants (PVs) in BRCA1/2 on TGP. Data on genetic counseling referrals and uptake, germline test results, family history, and patient demographics were collected. RESULTS In the study time frame, 1,899 patients underwent TGP. Testing identified 67 PVs in BRCA1 (23 variants) or BRCA2 (44 variants) in 62 patients. Thirty-five patients first received a referral to a genetic counselor following TGP with 33 patients completing genetic counseling. Of the 30 patients who pursued germline genetic testing following TGP, 11 were discovered to have a previously unknown germline BRCA PV. Nine of these patients were the first in their family diagnosed with hereditary breast and ovarian cancer. CONCLUSION This study represents one institution's experience with genetic counseling referrals, uptake, and germline results following TGP. For some patients, TGP will be the first indicator of an underlying hereditary condition. Identifying patients with PVs (which may be germline) through TGP is an important new genetic counseling referral tool that can have important implications for the patient and their family.
Collapse
Affiliation(s)
| | - Whitney F. Espinel
- University of Utah, Salt Lake City, UT
- Huntsman Cancer Institute, Salt Lake City, UT
| | - Jennie Vagher
- University of Utah, Salt Lake City, UT
- Huntsman Cancer Institute, Salt Lake City, UT
| | - Amanda Gammon
- University of Utah, Salt Lake City, UT
- Huntsman Cancer Institute, Salt Lake City, UT
| |
Collapse
|
27
|
Pauley K, Koptiuch C, Greenberg S, Kohlmann W, Jeter J, Colonna S, Werner T, Kinsey C, Gilcrease G, Weis J, Whisenant J, Florou V, Garrido-Laguna I. Discrepancies between tumor genomic profiling and germline genetic testing. ESMO Open 2022; 7:100526. [PMID: 35780590 PMCID: PMC9511791 DOI: 10.1016/j.esmoop.2022.100526] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 06/06/2022] [Accepted: 06/09/2022] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND Tumor genomic profiling (TGP) often incidentally identifies germline pathogenic variants (PVs) associated with cancer predisposition syndromes. Methods used by somatic testing laboratories, including germline analysis, differ from designated germline laboratories that have optimized the identification of germline PVs. This study evaluated discrepancies between somatic and germline testing results, and their impact on patients. PATIENTS AND METHODS Chart reviews were carried out at a single institution for patients who had both somatic and designated germline genetic testing. Cases with discrepant results in which germline PVs were not detected by the somatic laboratory or in which variant classification differed are summarized. RESULTS TGP was carried out on 2811 cancer patients, 600 of whom also underwent designated germline genetic testing. Germline PVs were identified for 109 individuals. Discrepancies between germline genetic testing and tumor profiling reports were identified in 20 cases, including 14 PVs identified by designated germline genetic testing laboratories that were not reported by somatic testing laboratories and six variants with discrepant classifications between the designated germline and somatic testing laboratories. Three PVs identified by designated germline laboratories are targets for poly adenosine diphosphate-ribose polymerase (PARP) inhibitors and resulted in different treatment options. Of the PVs identified by designated germline laboratories, 60% (n = 12) were in genes with established associations to the patients' cancer, and 40% of the PVs were incidental. The majority (90%) of all discrepant findings, both contributory and incidental, changed management recommendations for these patients, highlighting the importance of comprehensive germline assessment. CONCLUSIONS Methods used by somatic laboratories, regardless of the inclusion of germline analysis, differ from those of designated germline laboratories for identifying germline PVs. Unrecognized germline PVs may harm patients by missing hereditary syndromes and targeted therapy opportunities (e.g. anti-programmed cell death protein 1 immunotherapy, PARP inhibitors). Clinicians should refer patients who meet the criteria for genetic evaluation regardless of somatic testing outcomes.
Collapse
Affiliation(s)
- K Pauley
- Family Cancer Assessment Clinic, Huntsman Cancer Institute, Salt Lake City, USA.
| | - C Koptiuch
- Family Cancer Assessment Clinic, Huntsman Cancer Institute, Salt Lake City, USA
| | - S Greenberg
- Family Cancer Assessment Clinic, Huntsman Cancer Institute, Salt Lake City, USA
| | - W Kohlmann
- Family Cancer Assessment Clinic, Huntsman Cancer Institute, Salt Lake City, USA
| | - J Jeter
- Department of Internal Medicine, Huntsman Cancer Institute, Salt Lake City, USA
| | - S Colonna
- Department of Internal Medicine, Huntsman Cancer Institute, Salt Lake City, USA
| | - T Werner
- Department of Internal Medicine, Huntsman Cancer Institute, Salt Lake City, USA
| | - C Kinsey
- Department of Internal Medicine, Huntsman Cancer Institute, Salt Lake City, USA
| | - G Gilcrease
- Department of Internal Medicine, Huntsman Cancer Institute, Salt Lake City, USA
| | - J Weis
- Department of Internal Medicine, Huntsman Cancer Institute, Salt Lake City, USA
| | - J Whisenant
- Department of Medical Oncology and Hematology, Utah Cancer Specialists, Salt Lake City, USA
| | - V Florou
- Department of Internal Medicine, Huntsman Cancer Institute, Salt Lake City, USA
| | - I Garrido-Laguna
- Department of Internal Medicine, Huntsman Cancer Institute, Salt Lake City, USA
| |
Collapse
|
28
|
Szpechcinski A, Szolkowska M, Winiarski S, Lechowicz U, Wisniewski P, Knetki-Wroblewska M. Targeted Next-Generation Sequencing of Thymic Epithelial Tumours Revealed Pathogenic Variants in KIT, ERBB2, KRAS, and TP53 in 30% of Thymic Carcinomas. Cancers (Basel) 2022; 14:3388. [PMID: 35884448 PMCID: PMC9324890 DOI: 10.3390/cancers14143388] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 07/05/2022] [Accepted: 07/11/2022] [Indexed: 01/27/2023] Open
Abstract
A better understanding of the molecular pathogenesis of thymic epithelial tumours (TETs) could revolutionise their treatment. We evaluated thymomas and thymic carcinomas by next-generation sequencing (NGS) of somatic or germline single nucleotide variants (SNVs) in genes commonly mutated in solid tumours. In total, 19 thymomas and 34 thymic carcinomas were analysed for nonsynonymous SNVs in 15 genes by targeted NGS (reference genome: hg19/GRCh37). Ten SNVs in TP53 (G154V, R158P, L194H, R267fs, R273C, R306 *, Q317 *), ERBB2 (V773M), KIT (L576P), and KRAS (Q61L) considered somatic and pathogenic/likely pathogenic were detected in 10 of 34 (29.4%) thymic carcinomas. No somatic SNVs confirmed as pathogenic/likely pathogenic were found in thymomas. Rare SNVs of uncertain or unknown functional and clinical significance, to our knowledge not reported previously in TETs, were found in ERBB2 (S703R), KIT (I690V), and FOXL2 (P157S) in 3 of 19 (16%) thymomas. The most frequent germline SNVs were TP53 P72R (94% TETs), ERBB2 I655V (40% TETs), and KIT M541L (9% TETs). No significant difference in median disease-free survival (DFS) was found between thymic carcinoma patients with and without pathogenic SNVs (p = 0.190); however, a trend toward a longer DFS was observed in the latter (16.0 vs. 30.0 months, respectively). In summary, NGS analysis of TETs revealed several SNVs in genes related to the p53, AKT, MAPK, and K-Ras signalling pathways. Thymic carcinomas showed greater genetic dysregulation than thymomas. The germline and rare SNVs of uncertain clinical significance reported in this study add to the number of known genetic alterations in TETs, thus extending our molecular understanding of these neoplasms. Druggable KIT alterations in thymic carcinomas have potential as therapeutic targets.
Collapse
Affiliation(s)
- Adam Szpechcinski
- Department of Genetics and Clinical Immunology, The Institute of Tuberculosis and Lung Diseases, 01-138 Warsaw, Poland;
| | - Malgorzata Szolkowska
- Department of Pathology, The Institute of Tuberculosis and Lung Diseases, 01-138 Warsaw, Poland
| | - Sebastian Winiarski
- Clinics of Thoracic Surgery, The Institute of Tuberculosis and Lung Diseases, 01-138 Warsaw, Poland;
| | - Urszula Lechowicz
- Department of Genetics and Clinical Immunology, The Institute of Tuberculosis and Lung Diseases, 01-138 Warsaw, Poland;
| | - Piotr Wisniewski
- Department of Pathology and Laboratory Medicine, The Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland;
| | - Magdalena Knetki-Wroblewska
- Department of Lung Cancer and Chest Tumours, The Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland;
| |
Collapse
|
29
|
Ahmed M, Mäkinen VP, Mulugeta A, Shin J, Boyle T, Hyppönen E, Lee SH. Considering hormone-sensitive cancers as a single disease in the UK biobank reveals shared aetiology. Commun Biol 2022; 5:614. [PMID: 35729236 PMCID: PMC9213416 DOI: 10.1038/s42003-022-03554-y] [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: 10/03/2021] [Accepted: 06/02/2022] [Indexed: 11/09/2022] Open
Abstract
Hormone-related cancers, including cancers of the breast, prostate, ovaries, uterine, and thyroid, globally contribute to the majority of cancer incidence. We hypothesize that hormone-sensitive cancers share common genetic risk factors that have rarely been investigated by previous genomic studies of site-specific cancers. Here, we show that considering hormone-sensitive cancers as a single disease in the UK Biobank reveals shared genetic aetiology. We observe that a significant proportion of variance in disease liability is explained by the genome-wide single nucleotide polymorphisms (SNPs), i.e., SNP-based heritability on the liability scale is estimated as 10.06% (SE 0.70%). Moreover, we find 55 genome-wide significant SNPs for the disease, using a genome-wide association study. Pair-wise analysis also estimates positive genetic correlations between some pairs of hormone-sensitive cancers although they are not statistically significant. Our finding suggests that heritable genetic factors may be a key driver in the mechanism of carcinogenesis shared by hormone-sensitive cancers.
Collapse
Affiliation(s)
- Muktar Ahmed
- Australian Centre for Precision Health, University of South Australia, Adelaide, SA, Australia. .,Department of Epidemiology, Faculty of Public Health, Jimma University Institute of Health, Jimma, Ethiopia. .,UniSA Clinical and Health Sciences, University of South Australia, Adelaide, SA, Australia. .,South Australian Health and Medical Research Institute, Adelaide, SA, Australia.
| | - Ville-Petteri Mäkinen
- Australian Centre for Precision Health, University of South Australia, Adelaide, SA, Australia.,Computational Systems Biology Program, Precision Medicine Theme, South Australian Health and Medical Research Institute, Adelaide, SA, Australia
| | - Anwar Mulugeta
- Australian Centre for Precision Health, University of South Australia, Adelaide, SA, Australia.,UniSA Clinical and Health Sciences, University of South Australia, Adelaide, SA, Australia.,South Australian Health and Medical Research Institute, Adelaide, SA, Australia
| | - Jisu Shin
- Australian Centre for Precision Health, University of South Australia, Adelaide, SA, Australia.,UniSA Allied Health & Human Performance, University of South Australia, Adelaide, SA, Australia
| | - Terry Boyle
- Australian Centre for Precision Health, University of South Australia, Adelaide, SA, Australia.,South Australian Health and Medical Research Institute, Adelaide, SA, Australia.,UniSA Allied Health & Human Performance, University of South Australia, Adelaide, SA, Australia
| | - Elina Hyppönen
- Australian Centre for Precision Health, University of South Australia, Adelaide, SA, Australia.,UniSA Clinical and Health Sciences, University of South Australia, Adelaide, SA, Australia.,South Australian Health and Medical Research Institute, Adelaide, SA, Australia
| | - Sang Hong Lee
- Australian Centre for Precision Health, University of South Australia, Adelaide, SA, Australia. .,South Australian Health and Medical Research Institute, Adelaide, SA, Australia. .,UniSA Allied Health & Human Performance, University of South Australia, Adelaide, SA, Australia.
| |
Collapse
|
30
|
Scott A, Mohan A, Austin S, Amini E, Raupp S, Pannecouk B, Kelley MJ, Narla G, Ramnath N. Integrating Medical Genetics Into Precision Oncology Practice in the Veterans Health Administration: The Time Is Now. JCO Oncol Pract 2022; 18:e966-e973. [PMID: 35258993 PMCID: PMC9191304 DOI: 10.1200/op.21.00693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 12/12/2021] [Accepted: 02/08/2022] [Indexed: 11/20/2022] Open
Abstract
PURPOSE Increased access and utilization of tumor profiling of cancers in our veteran population uncovered a modest number of potentially pathogenic germline variants (PPGVs) that require genetics referral for follow-up evaluation and germline sequencing. Challenges identified specific to the veteran population include paucity of genetics providers, either at a veteran's VA facility or nearby non-VA facilities. We sought to investigate the number of veterans who would benefit from having such resources at both local and national levels. METHODS Annotated clinical reports of mutations identified by tumor-only profiling and medical records of veterans with solid tumors at the Veterans Administration Ann Arbor Healthcare System (VA AAHS) between 2015 and 2020 were reviewed. PPGVs were identified according to society recommendations (such as ESMO and American Board of Medical Genetics and Genomics), expert review, and/or previously published criteria. After the analysis of our local VA population, these same criteria were then applied to veterans in the National Precision Oncology Program (NPOP). RESULTS Two hundred eight veterans underwent tumor profiling at the VA AAHS over the defined time period. This included 20 different primary tumor sites with over half (n = 130) being advanced cancer at diagnosis. Of these, 18 veterans (8.5%) had mutations suggestive of a PPGV. Applying these criteria to the larger NPOP database (n = 20,014), a similar percentage (6%) of PPGVs were identified. CONCLUSION These results indicate a PPGV frequency (6%-9% of veterans) consistent with the prevalence of inherited cancer predisposition syndromes in the general population, underscoring the need for medical genetics as part of standard oncologic care for veterans. We explore current and future care delivery models to optimize incorporation of medical genetics and genetic counseling to best serve veterans needing such services.
Collapse
Affiliation(s)
- Anthony Scott
- Division of Genetic Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, MI
- Division of Oncology, Veterans Affairs Ann Arbor Healthcare System, Ann Arbor, MI
| | - Arathi Mohan
- Division of Oncology, Veterans Affairs Ann Arbor Healthcare System, Ann Arbor, MI
- Division of Hematology and Oncology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI
| | - Sarah Austin
- Division of Genetic Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, MI
- Division of Oncology, Veterans Affairs Ann Arbor Healthcare System, Ann Arbor, MI
| | - Erika Amini
- Division of Genetic Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, MI
| | - Shelby Raupp
- Division of Oncology, Veterans Affairs Ann Arbor Healthcare System, Ann Arbor, MI
| | - Brittany Pannecouk
- Division of Oncology, Veterans Affairs Ann Arbor Healthcare System, Ann Arbor, MI
| | - Michael J. Kelley
- Division of Hematology Oncology, Department of Medicine, Duke University, VA Medical Center in Durham, Durham, NC
| | - Goutham Narla
- Division of Genetic Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, MI
- Division of Oncology, Veterans Affairs Ann Arbor Healthcare System, Ann Arbor, MI
| | - Nithya Ramnath
- Division of Oncology, Veterans Affairs Ann Arbor Healthcare System, Ann Arbor, MI
- Division of Hematology and Oncology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI
| |
Collapse
|
31
|
Bychkovsky BL, Li T, Sotelo J, Tayob N, Mercado J, Gomy I, Chittenden A, Kane S, Stokes S, Hughes ME, Kim JS, Umeton R, Awad MM, Konstantinopoulos PA, Yurgelun MB, Wolpin BM, Taplin ME, Newmark RE, Johnson BE, Lindeman NI, MacConaill LE, Garber JE, Lin NU. Identification and Management of Pathogenic Variants in BRCA1, BRCA2, and PALB2 in a Tumor-Only Genomic Testing Program. Clin Cancer Res 2022; 28:2349-2360. [PMID: 35363308 PMCID: PMC9167798 DOI: 10.1158/1078-0432.ccr-21-2861] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 12/20/2021] [Accepted: 03/29/2022] [Indexed: 11/16/2022]
Abstract
PURPOSE Tumor-only genomic testing can uncover somatic and germline pathogenic variants [pathogenic/likely pathogenic (P/LP)] in cancer predisposition genes. We describe the prevalence of P/LPs in BRCA1/2 and PALB2 (B1B2P2) across malignancies and the frequency of clinical germline testing (CGT) in patients with P/LPs in B1B2P2 identified on tumor-only testing. EXPERIMENTAL DESIGN Among 7,575 patients with cancer tested between 2016 and 2018 with the OncoPanel tumor-only sequencing assay, we characterized P/LP frequencies by tumor type, receipt of CGT prior to or within 12 months after OncoPanel, and factors associated with CGT. RESULTS 272 (3.6%) patients had OncoPanel-detected P/LPs in B1B2P2: 37.5% of P/LPs were in BRCA-related cancers; the remainder were in non-BRCA tumors. P/LPs were detected in ≥5% of breast, pancreatic, prostate, ovarian, nonmelanoma skin, endometrial, small cell lung, and colorectal cancers. 37.9% of patients with P/LPs received CGT prior to OncoPanel; an additional 10.7% underwent CGT within 12 months of OncoPanel. Among 132 with CGT, 88.6% had ≥1 clinical factor for CGT compared with 47.1% who did not undergo CGT. Patients with BRCA tumors were more likely to have CGT compared with those without (81.4% vs. 29.0%, P < 0.0001). Among patients with CGT, 70.5% (93/132) of P/LPs were germline. CONCLUSIONS Tumor-only genomic testing identified P/LPs in B1B2P2 in 3.6% of patients. 52.9% of patients with tumor-detected P/LPs and without CGT did not meet personal or family history criteria for CGT. In addition, some patients with tumor-detected P/LPs were not referred for CGT, especially those with non-BRCA tumors. Given implications for treatment selection and familial cancer risk, processes to reliably trigger CGT from tumor-genomic findings are needed.
Collapse
Affiliation(s)
- Brittany L. Bychkovsky
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
- Division for Cancer Genetics and Prevention, Dana-Farber Cancer Institute, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - Tianyu Li
- Department of Data Science, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Jilliane Sotelo
- Division for Cancer Genetics and Prevention, Dana-Farber Cancer Institute, Boston, Massachusetts
- Thermo Fisher Scientific, Waltham, Massachusetts
| | - Nabihah Tayob
- Harvard Medical School, Boston, Massachusetts
- Department of Data Science, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Joanna Mercado
- Division for Cancer Genetics and Prevention, Dana-Farber Cancer Institute, Boston, Massachusetts
- Genome Medical, South San Francisco, California
| | - Israel Gomy
- Division for Cancer Genetics and Prevention, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Anu Chittenden
- Division for Cancer Genetics and Prevention, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Sarah Kane
- Division for Cancer Genetics and Prevention, Dana-Farber Cancer Institute, Boston, Massachusetts
- Memorial Sloan Kettering Cancer Center, Clinical Genetics Service, New York, New York
| | - Samantha Stokes
- Division for Cancer Genetics and Prevention, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Melissa E. Hughes
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Ji Seok Kim
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
- Division for Cancer Genetics and Prevention, Dana-Farber Cancer Institute, Boston, Massachusetts
- Dewpoint Therapeutics, Boston, Massachusetts
| | - Renato Umeton
- Department of Data Science, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Mark M. Awad
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - Panagiotis A. Konstantinopoulos
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - Matthew B. Yurgelun
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
- Division for Cancer Genetics and Prevention, Dana-Farber Cancer Institute, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - Brian M. Wolpin
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - Mary-Ellen Taplin
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - Randall E. Newmark
- Massachusetts General Hospital Research Institute, Partners HealthCare, Boston, Massachusetts
| | - Bruce E. Johnson
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
- Center for Cancer Genomics, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Neal I. Lindeman
- Harvard Medical School, Boston, Massachusetts
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts
| | - Laura E. MacConaill
- Harvard Medical School, Boston, Massachusetts
- Center for Cancer Genomics, Dana-Farber Cancer Institute, Boston, Massachusetts
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts
| | - Judy E. Garber
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
- Division for Cancer Genetics and Prevention, Dana-Farber Cancer Institute, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - Nancy U. Lin
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
32
|
Montgomery B, Wang S, Rettig M, Lee B, Bates J, Pritchard C. Ordering and Interpreting Precision Oncology Studies for Adults With Advanced Solid Tumors: A Primer. Fed Pract 2022; 39:S16-S24. [PMID: 35929009 PMCID: PMC9346578 DOI: 10.12788/fp.0270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
BACKGROUND The promise of precision oncology can only be realized when genetic alterations are identified that can be leveraged to improve response and minimize toxicity. Identifying those alterations requires the knowledge to order the right test and to interpret the results correctly. This primer is designed to help clinicians order the appropriate testing for patients with specific malignancies and to give them an informed approach to interpretation. OBSERVATIONS Germline DNA is usually acquired from peripheral blood, buccal swab, or saliva collection in patients with a metastatic malignancy and can provide treatment options otherwise not available. However, germline testing does not indicate alterations that arise solely in tumor tissue. Somatic testing may be performed on primary tumor, metastatic biopsy, or circulating tumor DNA when the alteration is present at the time that the tumor developed and expected to be carried through the evolution of the tumor. CONCLUSIONS The rapid growth in technology and ability to enhance understanding of relevant tumor biology continues to improve the therapeutic landscape for individuals dealing with malignancy as does our ability to find targetable genetic alterations with the potential for meaningful clinical benefit.
Collapse
Affiliation(s)
- Bruce Montgomery
- Veterans Affairs Puget Sound Health Care System, Seattle, Washington
- University of Washington, Seattle
| | - Sunny Wang
- San Francisco Veterans Affairs Health Care System, California
- University of California San Francisco
| | - Matthew Rettig
- Greater Los Angeles Health Care System, California
- University of California, Los Angeles
| | - Benson Lee
- Greater Los Angeles Health Care System, California
| | - Jill Bates
- Durham Veterans Affairs Medical Center, North Carolina
- University of North Carolina, Chapel Hill
| | | |
Collapse
|
33
|
Higashigawa S, Matsubayashi H, Kiyozumi Y, Kado N, Nishimura S, Oishi T, Sugino T, Fushiki K, Shirasu H, Yasui H, Mamesaya N, Fukuzaki N, Kunitomo K, Horiuchi Y, Kenmotsu H, Serizawa M. Present status of germline findings in precision medicine for Japanese cancer patients: issues in the current system. Jpn J Clin Oncol 2022; 52:599-608. [PMID: 35411369 DOI: 10.1093/jjco/hyac046] [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: 09/14/2021] [Revised: 12/11/2021] [Accepted: 03/07/2022] [Indexed: 11/14/2022] Open
Abstract
OBJECTIVE Since 2019, precision cancer medicine has been covered by national insurance in Japan; however, to date, germline findings have not been fully reported. The aim of this study was to evaluate the current status and raise a problem of germline finding analysis and disclosure in Japanese precision cancer medicine. METHODS Germline findings of 52 genes were examined in 296 cases with advanced cancer by a case series study. RESULTS Six (2.0%) cases were examined by the Oncoguide™ NCC Oncopanel with germline testing, but no germline findings were reported. The remaining 290 (98.0%) cases were analyzed by FoundationOne® CDx (tumor-only testing), which recognized 404 pathogenic variants; those of BRCA1/2 were recognized in 16 (5.5%) tumors. Our institutional algorithm suggested 39 candidate germline findings in 34 cases, while the public algorithm listed at least 91 candidate germline findings. Four germline findings had been previously identified (BRCA1: 3 and ATM: 1). Nine of 30 cases with candidate germline findings excluding these known germline findings refused or deferred germline testing. Only 4 of 16 cases that received counseling underwent germline testing, and those 4 revealed 3 germline findings (BRCA2, CDK4 and RAD51C); in total, 8 (2.7%) germline findings were revealed. Reasons for refusing genetic counseling and/or germline testing included extra hospital visits, added expense for germline testing due to limited national insurance coverage, poor patient physical condition and no known family members associated with the possible germline finding. CONCLUSIONS In current Japanese precision cancer medicine, only a small fraction of the patients undergoes germline testing and demonstrated germline finding. The current results suggested a need for earlier indications for precision cancer medicine, broader insurance coverage and more efficient germline finding prediction algorithms, to increase the number of germline testings and to improve the following managements.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | - Yasue Horiuchi
- Division of Genetic Medicine Promotion.,Department of Psychiatry and Behavioral Sciences, Tokyo Metropolitan Institute of Medical Science, Setagaya-ku, Tokyo, Japan
| | | | - Masakuni Serizawa
- Clinical Research Center, Shizuoka Cancer Center, Nagaizumi-cho, Sunto-gun, Shizuoka, Japan
| |
Collapse
|
34
|
|
35
|
Bellcross CA. Hereditary Breast and Ovarian Cancer. Obstet Gynecol Clin North Am 2022; 49:117-147. [DOI: 10.1016/j.ogc.2021.11.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
36
|
Sánchez-Lorenzo L, Salas-Benito D, Villamayor J, Patiño-García A, González-Martín A. The BRCA Gene in Epithelial Ovarian Cancer. Cancers (Basel) 2022; 14:1235. [PMID: 35267543 PMCID: PMC8909050 DOI: 10.3390/cancers14051235] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Revised: 02/20/2022] [Accepted: 02/21/2022] [Indexed: 01/27/2023] Open
Abstract
Epithelial ovarian cancer (EOC) is still the most lethal gynecological cancer. Germline alterations in breast cancer 1 (gBRCA1) and breast cancer 2 (gBRCA2) genes have been identified in up to 18% of women diagnosed with EOC, and somatic mutations are found in an additional 7%. Testing of BRCA at the primary diagnosis of patients with EOC is recommended due to the implications in the genomic counseling of the patients and their families, as well as for the therapeutic implications. Indeed, the introduction of poly-(ADP ribose) polymerase inhibitors (PARPis) has changed the natural history of patients harboring a mutation in BRCA, and has resulted in a new era in the treatment of patients with ovarian cancer harboring a BRCA mutation.
Collapse
Affiliation(s)
- Luisa Sánchez-Lorenzo
- Department of Medical Oncology, Clínica Universidad de Navarra, 28027 Madrid, Spain; (L.S.-L.); (D.S.-B.); (J.V.); (A.P.-G.)
| | - Diego Salas-Benito
- Department of Medical Oncology, Clínica Universidad de Navarra, 28027 Madrid, Spain; (L.S.-L.); (D.S.-B.); (J.V.); (A.P.-G.)
| | - Julia Villamayor
- Department of Medical Oncology, Clínica Universidad de Navarra, 28027 Madrid, Spain; (L.S.-L.); (D.S.-B.); (J.V.); (A.P.-G.)
| | - Ana Patiño-García
- Department of Medical Oncology, Clínica Universidad de Navarra, 28027 Madrid, Spain; (L.S.-L.); (D.S.-B.); (J.V.); (A.P.-G.)
- Solid Tumor Program (CIMA), Clínica Universidad de Navarra, 31008 Pamplona, Spain
| | - Antonio González-Martín
- Department of Medical Oncology, Clínica Universidad de Navarra, 28027 Madrid, Spain; (L.S.-L.); (D.S.-B.); (J.V.); (A.P.-G.)
- Solid Tumor Program (CIMA), Clínica Universidad de Navarra, 31008 Pamplona, Spain
| |
Collapse
|
37
|
Hoadley A, Bass SB, Chertock Y, Brajuha J, D’Avanzo P, Kelly PJ, Hall MJ. The Role of Medical Mistrust in Concerns about Tumor Genomic Profiling among Black and African American Cancer Patients. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:ijerph19052598. [PMID: 35270290 PMCID: PMC8909390 DOI: 10.3390/ijerph19052598] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Revised: 02/18/2022] [Accepted: 02/22/2022] [Indexed: 12/13/2022]
Abstract
Tumor genomic profiling (TGP) is used in oncology practice to optimize cancer treatment and improve survival rates. However, TGP is underutilized among Black and African American (AA) patients, creating potential disparities in cancer treatment outcomes. Cost, accuracy, and privacy are barriers to genetic testing, but medical mistrust (MM) may also influence how Black and AA cancer patients perceive TGP. From December 2019 to February 2020, 112 Black and AA adults from two outpatient oncology sites in Philadelphia, PA without a known history of having TGP testing conducted completed a cross-sectional survey. Items queried included sociodemographic characteristics, clinical factors, patient-oncologist relationship quality, medical mistrust, and concerns about TGP. A k-means cluster analysis revealed two distinct psychographic clusters: high (MM-H) versus low (MM-L) medical mistrust. Clusters were not associated with any sociodemographic or clinical factors, except for age (MM-H patients older than MM-L patients, p = 0.006). Eleven TGP concerns were assessed; MM-H patients expressed greater concerns than MM-L patients, including distrust of the government, insurance carriers, and pharmaceutical companies. TGP concerns varied significantly based on level of medical mistrust, irrespective of sociodemographic characteristics. Targeted communications addressing TGP concerns may mitigate disparities in TGP uptake among those with medical mistrust.
Collapse
Affiliation(s)
- Ariel Hoadley
- Risk Communication Laboratory, Department of Social and Behavioral Sciences, College of Public Health, Temple University, 1301 Cecil B Moore Ave, Rm 947, Philadelphia, PA 19122, USA; (S.B.B.); (J.B.); (P.D.); (P.J.K.)
- Correspondence:
| | - Sarah Bauerle Bass
- Risk Communication Laboratory, Department of Social and Behavioral Sciences, College of Public Health, Temple University, 1301 Cecil B Moore Ave, Rm 947, Philadelphia, PA 19122, USA; (S.B.B.); (J.B.); (P.D.); (P.J.K.)
| | - Yana Chertock
- Fox Chase Cancer Center, Cancer Prevention and Control Program, Department of Clinical Genetics, 333 Cottman Avenue, Philadelphia, PA 19111, USA; (Y.C.); (M.J.H.)
| | - Jesse Brajuha
- Risk Communication Laboratory, Department of Social and Behavioral Sciences, College of Public Health, Temple University, 1301 Cecil B Moore Ave, Rm 947, Philadelphia, PA 19122, USA; (S.B.B.); (J.B.); (P.D.); (P.J.K.)
| | - Paul D’Avanzo
- Risk Communication Laboratory, Department of Social and Behavioral Sciences, College of Public Health, Temple University, 1301 Cecil B Moore Ave, Rm 947, Philadelphia, PA 19122, USA; (S.B.B.); (J.B.); (P.D.); (P.J.K.)
| | - Patrick J. Kelly
- Risk Communication Laboratory, Department of Social and Behavioral Sciences, College of Public Health, Temple University, 1301 Cecil B Moore Ave, Rm 947, Philadelphia, PA 19122, USA; (S.B.B.); (J.B.); (P.D.); (P.J.K.)
| | - Michael J. Hall
- Fox Chase Cancer Center, Cancer Prevention and Control Program, Department of Clinical Genetics, 333 Cottman Avenue, Philadelphia, PA 19111, USA; (Y.C.); (M.J.H.)
| |
Collapse
|
38
|
Rahman B, McEwen A, Phillips JL, Tucker K, Goldstein D, Jacobs C. Genetic and genomic learning needs of oncologists and oncology nurses in the era of precision medicine: a scoping review. Per Med 2022; 19:139-153. [PMID: 35060769 DOI: 10.2217/pme-2021-0096] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Genetic and genomic data are increasingly guiding clinical care for cancer patients. To meet the growing demand for precision medicine, patient-facing oncology staff will be a part of leading the provision of genomic testing. A scoping review was undertaken to identify the range of genetic and genomic learning needs of oncologists and oncology nurses. Learning needs were reported relating to interpretation of genomic data, clinical decision-making, patient communication and counseling, and fundamentals of genetics and genomics. There was a lack of empirical research specific to oncology nurses and their learning needs in tumor sequencing. Our findings suggest that oncologists and oncology nurses need tailored support, education and training to improve their confidence and skills in adopting genomic testing into clinical practice.
Collapse
Affiliation(s)
- Belinda Rahman
- Graduate School of Health, University of Technology Sydney, NSW, Australia
| | - Alison McEwen
- Graduate School of Health, University of Technology Sydney, NSW, Australia
| | - Jane L Phillips
- School of Nursing, Faculty of Health, Queensland University of Technology, QLD, Australia.,IMPACCT, University of Technology Sydney, NSW, Australia
| | - Katherine Tucker
- Hereditary Cancer Centre, Prince of Wales Hospital, NSW, Australia.,Prince of Wales Clinical School, University of New South Wales, NSW, Australia
| | - David Goldstein
- Prince of Wales Clinical School, University of New South Wales, NSW, Australia
| | - Chris Jacobs
- Graduate School of Health, University of Technology Sydney, NSW, Australia
| |
Collapse
|
39
|
Kordes M, Tamborero D, Lagerstedt-Robinson K, Yachnin J, Rosenquist R, Löhr JM, Gustafsson Liljefors M. Discordant Reporting of a Previously Undescribed Pathogenic Germline BRCA2 Variant in Blood and Tumor Tissue in a Patient With Pancreatic Adenocarcinoma. JCO Precis Oncol 2021; 5:974-980. [PMID: 34994625 DOI: 10.1200/po.21.00024] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Affiliation(s)
- Maximilian Kordes
- Department of Upper Abdominal Diseases, Karolinska University Hospital, Stockholm, Sweden.,Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | - David Tamborero
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden.,Science for Life Laboratory, Karolinska Institutet, Stockholm, Sweden
| | - Kristina Lagerstedt-Robinson
- Department of Clinical Genetics, Karolinska University Hospital, Stockholm, Sweden.,Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Jeffrey Yachnin
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden.,Phase-I Unit, Center for Clinical Cancer Studies, Karolinska University Hospital, Stockholm, Sweden
| | - Richard Rosenquist
- Department of Clinical Genetics, Karolinska University Hospital, Stockholm, Sweden.,Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - J-Matthias Löhr
- Department of Upper Abdominal Diseases, Karolinska University Hospital, Stockholm, Sweden.,Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | - Maria Gustafsson Liljefors
- Department of Upper Abdominal Diseases, Karolinska University Hospital, Stockholm, Sweden.,Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
40
|
Sandoval RL, Masotti C, de Macedo MP, Ribeiro MFSA, Leite ACR, Meireles SI, Bovolin RM, Santini FC, Munhoz RR, Jardim DLF, Katz A, Camargo AA, Fernandes GDS, Achatz MI. Identification of the TP53 p.R337H Variant in Tumor Genomic Profiling Should Prompt Consideration of Germline Testing for Li-Fraumeni Syndrome. JCO Glob Oncol 2021; 7:1141-1150. [PMID: 34270331 PMCID: PMC8457781 DOI: 10.1200/go.21.00097] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Revised: 04/19/2021] [Accepted: 05/25/2021] [Indexed: 01/01/2023] Open
Abstract
PURPOSE Li-Fraumeni syndrome (LFS) is rare in the worldwide population, but it is highly prevalent in the Brazilian population because of a founder mutation, TP53 p.R337H, accounting for 0.3% of south and southeastern population. Clinical criteria for LFS may not identify all individuals at risk of carrying the Brazilian founder mutation because of its lower penetrance and variable expressivity. This variant is rarely described in databases of somatic mutations. Somatic findings in tumor molecular profiling may give insight to identify individuals who might be carriers of LFS and allow the adoption of risk reduction strategies for cancer. MATERIALS AND METHODS We determined the frequency of the TP53 p.R337H variant in tumor genomic profiling from 755 consecutive Brazilian patients with pan-cancer. This is a retrospective cohort from January 2013 to March 2020 at a tertiary care center in Brazil. RESULTS The TP53 p.R337H variant was found in 2% (15 of 755) of the samples. The mutation allele frequency ranged from 30% to 91.7%. A total of seven patients were referred for genetic counseling and germline testing after tumor genomic profiling results were disclosed. All the patients who proceeded with germline testing (6 of 6) confirmed the diagnosis of LFS. Family history was available in 12 cases. Nine patients (9 of 12) did not meet LFS clinical criteria. CONCLUSION The identification of the TP53 p.R337H variant in tumor genomic profiling should be a predictive finding of LFS in the Brazilian population and should prompt testing for germline status confirmation.
Collapse
Affiliation(s)
| | - Cibele Masotti
- Department of Molecular Oncology, Hospital Sírio-Libanês, São Paulo, Brazil
| | | | | | | | | | | | | | | | | | - Artur Katz
- Department of Oncology, Hospital Sírio-Libanês, São Paulo, Brazil
| | | | | | | |
Collapse
|
41
|
Lau TTY, May CM, Sefid Dashti ZJ, Swanson L, Starks ER, Parker JDK, Moore RA, Tucker T, Bosdet I, Young SS, Santos JL, Compton K, Heidary N, Hoang L, Schrader KA, Sun S, Kwon JS, Tinker AV, Karsan A. Use of Treatment-Focused Tumor Sequencing to Screen for Germline Cancer Predisposition. J Mol Diagn 2021; 23:1145-1158. [PMID: 34197922 DOI: 10.1016/j.jmoldx.2021.06.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 05/10/2021] [Accepted: 06/07/2021] [Indexed: 10/21/2022] Open
Abstract
Next-generation sequencing assays are capable of identifying cancer patients eligible for targeted therapies and can also detect germline variants associated with increased cancer susceptibility. However, these capabilities have yet to be routinely harmonized in a single assay because of challenges with accurately identifying germline variants from tumor-only data. We have developed the Oncology and Hereditary Cancer Program targeted capture panel, which uses tumor tissue to simultaneously screen for both clinically actionable solid tumor variants and germline variants across 45 genes. Validation using 14 tumor specimens, composed of patient samples and cell lines analyzed in triplicate, demonstrated high coverage with sensitive and specific identification of single-nucleotide variants and small insertions and deletions. Average coverage across all targets remained >2000× in 198 additional patient tumor samples. Analysis of 55 formalin-fixed, paraffin-embedded tumor samples for the detection of known germline variants within a subset of cancer-predisposition genes, including one multiexon deletion, yielded a 100% detection rate, demonstrating that germline variants can be reliably detected in tumor samples using a single panel. Combining targetable somatic and actionable germline variants into a single tumor tissue assay represents a streamlined approach that can inform treatment for patients with advanced cancers as well as identify those with potential germline variants who are eligible for confirmatory testing, but would not otherwise have been identified.
Collapse
Affiliation(s)
- Tammy T Y Lau
- Canada's Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, British Columbia
| | - Christina M May
- Canada's Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, British Columbia
| | - Zahra J Sefid Dashti
- Canada's Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, British Columbia
| | - Lucas Swanson
- Canada's Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, British Columbia
| | - Elizabeth R Starks
- Canada's Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, British Columbia
| | - Jeremy D K Parker
- Canada's Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, British Columbia
| | - Richard A Moore
- Canada's Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, British Columbia
| | - Tracy Tucker
- Cancer Genetics and Genomics Laboratory, BC Cancer, Vancouver, British Columbia; Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Ian Bosdet
- Cancer Genetics and Genomics Laboratory, BC Cancer, Vancouver, British Columbia; Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Sean S Young
- Cancer Genetics and Genomics Laboratory, BC Cancer, Vancouver, British Columbia; Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Jennifer L Santos
- Hereditary Cancer Program, BC Cancer, Vancouver, British Columbia; Department of Gynecologic Oncology, BC Cancer, Vancouver, British Columbia
| | - Katie Compton
- Hereditary Cancer Program, BC Cancer, Vancouver, British Columbia
| | - Nili Heidary
- Hereditary Cancer Program, BC Cancer, Vancouver, British Columbia
| | - Lien Hoang
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | | | - Sophie Sun
- Hereditary Cancer Program, BC Cancer, Vancouver, British Columbia
| | - Janice S Kwon
- Departments of Obstetrics and Gynecology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Anna V Tinker
- Department of Medical Oncology, BC Cancer, Vancouver, British Columbia
| | - Aly Karsan
- Canada's Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, British Columbia; Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada.
| |
Collapse
|
42
|
Jacobs C, Rahman B. One size does not fit all: The case for targeted education in genetics and genomics for cancer nurses. Eur J Cancer Care (Engl) 2021; 30:e13480. [PMID: 34131987 DOI: 10.1111/ecc.13480] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Accepted: 05/30/2021] [Indexed: 01/16/2023]
Affiliation(s)
- Chris Jacobs
- Graduate School of Health, University of Technology Sydney, Sydney, NSW, Australia
| | - Belinda Rahman
- Graduate School of Health, University of Technology Sydney, Sydney, NSW, Australia
| |
Collapse
|
43
|
Takvorian SU, Balogh E, Nass S, Valentin VL, Hoffman-Hogg L, Oyer RA, Carlson RW, Meropol NJ, Sheldon LK, Shulman LN. Developing and Sustaining an Effective and Resilient Oncology Careforce: Opportunities for Action. J Natl Cancer Inst 2021; 112:663-670. [PMID: 31868912 DOI: 10.1093/jnci/djz239] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 12/04/2019] [Accepted: 12/13/2019] [Indexed: 12/12/2022] Open
Abstract
Advances in cancer care have led to improved survival, which, coupled with demographic trends, have contributed to rapid growth in the number of patients needing cancer care services. However, with increasing caseload, care complexity, and administrative burden, the current workforce is ill equipped to meet these burgeoning new demands. These trends have contributed to clinician burnout, compounding a widening workforce shortage. Moreover, family caregivers, who have unique knowledge of patient preferences, symptoms, and goals of care, are infrequently appreciated and supported as integral members of the oncology "careforce." A crisis is looming, which will hinder access to timely, high-quality cancer care if left unchecked. Stemming from the proceedings of a 2019 workshop convened by the National Cancer Policy Forum of the National Academies of Sciences, Engineering, and Medicine, this commentary characterizes the factors contributing to an increasingly strained oncology careforce and presents multilevel strategies to improve its efficiency, effectiveness, and resilience. Together, these will enable today's oncology careforce to provide high-quality care to more patients while improving the patient, caregiver, and clinician experience.
Collapse
Affiliation(s)
- Samuel U Takvorian
- Division of Hematology & Oncology, Perelman School of Medicine, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Erin Balogh
- National Cancer Policy Forum, National Academies of Sciences, Engineering, and Medicine, Washington DC, USA
| | - Sharyl Nass
- National Cancer Policy Forum and Board on Health Care Services, National Academies of Sciences, Engineering, and Medicine, Washington DC, USA
| | - Virginia L Valentin
- Division of Physician Assistant Studies, Department of Family and Preventive Medicine, University of Utah, Salt Lake City, UT, USA
| | - Lori Hoffman-Hogg
- Office of Nursing Services, Department of Veterans Affairs, Prevention Policy National Center for Health Promotion and Disease Prevention, Veterans Health Administration, Washington DC, USA
| | - Randall A Oyer
- Lancaster General Health, Penn Medicine, Lancaster, PA, USA
| | - Robert W Carlson
- National Comprehensive Cancer Network, Plymouth Meeting, PA, USA
| | | | | | - Lawrence N Shulman
- Division of Hematology & Oncology, Perelman School of Medicine, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
44
|
Stachowiak S, Jacquart A, Zimmermann MT, George B, Dong H, Geurts JL. Germline evaluation of patients undergoing tumor genomic profiling: An academic cancer center's experience with implementing a germline review protocol. J Genet Couns 2021; 30:900-910. [PMID: 33754402 DOI: 10.1002/jgc4.1392] [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: 08/19/2020] [Revised: 12/16/2020] [Accepted: 01/14/2021] [Indexed: 11/07/2022]
Abstract
Tumor genomic profiling (TGP) has the potential to identify germline variants in addition to its primary use of informing cancer treatment based on genetic alterations within the tumor. However, there are no formal consensus guidelines to identify patients who would be eligible for genetic counseling (GC) and germline testing (GT) testing in patients undergoing TGP. The purpose of this study is to describe an institutionally developed Germline Review Protocol (GRP) to evaluate adult cancer patient cases already undergoing TGP to determine GC referral eligibility. We report on our retrospective experience implementing this protocol into practice wherein 172 patients out of 638 patients reviewed (27%) were recommended for a GC referral over a 17-month time period. Of those 172 patients recommended for a GC referral, only 34 patients (20%) completed GC and GT. Among patients who received GT, 15 (44%) were positive for at least one pathogenic or likely pathogenic (P/LP) variant, seven patients (21%) were negative and 12 patients (35%) had at least 1 variant of uncertain significance (VUS). The primary reason GC and GT was not completed was because the patient moved to hospice care or was deceased. This is one of the first studies outlining the process and results of a formalized institutional protocol to facilitate patient referrals for GC and GT based on TGP results.
Collapse
Affiliation(s)
- Samantha Stachowiak
- Department of Obstetrics & Gynecology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Amanda Jacquart
- Department of Obstetrics & Gynecology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Michael T Zimmermann
- Bioinformatics Research and Development Laboratory, Genomic Sciences and Precision Medicine Center, Medical College of Wisconsin, Milwaukee, WI, USA.,Clinical and Translational Sciences Institute, Medical College of Wisconsin, Milwaukee, WI, USA.,Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Ben George
- Division of Hematology and Oncology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Huaying Dong
- Institute for Health & Equity, Division of Biostatistics, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Jennifer L Geurts
- Institute for Health and Equity, Medical College of Wisconsin, Milwaukee, WI, USA
| |
Collapse
|
45
|
Lee H, Kim EN, Lee JY, Kim JH, Oh JH, Kim WK, Cho EJ, Lim J, Chun SM, Sung CO. Characterization of early postzygotic somatic mutations through multi-organ analysis. J Hum Genet 2021; 66:777-784. [PMID: 33611337 DOI: 10.1038/s10038-021-00908-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 01/14/2021] [Accepted: 01/29/2021] [Indexed: 11/09/2022]
Abstract
Mosaicisms caused by postzygotic mutational events are of increasing interest because of their potential association with various human diseases. Postzygotic somatic mutations have not been well characterized however in terms of their developmental lineage in humans. We conducted whole-genome sequencing (WGS) and targeted deep sequencing in 15 organs across three developmental lineages from a single male fetus with polycystic kidney disease (PKD) of 21 weeks gestational age. This fetus had no detectable neurological abnormalities at autopsy but germline mutations in the PKHD1 gene were identified that may have been associated with the PKD. Eight early embryonic mosaic variants with no alteration of protein function were detected. These variants were thought to have occurred at the two or four cell stages after fertilization with a mutational pattern involving frequent C>T and T>C transitions. In our current analyses, no tendency toward organ-specific mutation occurrences was found as the eight variants were detected in all 15 organs. However different allele fractions of these variants were found in different organs, suggesting a tissue-specific asymmetric growth of cells that reflected the developmental germ layer of each organ. This indicated that somatic mutation occurrences, even in early embryogenesis, can affect specific organ development or disease. Our current analyses demonstrate that multi-organ analysis is helpful for understanding genomic mosaicism. Our results also provide insights into the biological role of mosaicism in embryonic development and disease.
Collapse
Affiliation(s)
- Hyeonjin Lee
- Department of Medical Science, Asan Medical Institute of Convergence Science and Technology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.,Center for Cancer Genome Discovery, Asan Institute for Life Science, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Eun Na Kim
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Ji-Young Lee
- Department of Medical Science, Asan Medical Institute of Convergence Science and Technology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.,Center for Cancer Genome Discovery, Asan Institute for Life Science, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Ji Hun Kim
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Ji-Hye Oh
- Department of Medical Science, Asan Medical Institute of Convergence Science and Technology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.,Center for Cancer Genome Discovery, Asan Institute for Life Science, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Won-Kyung Kim
- Department of Medical Science, Asan Medical Institute of Convergence Science and Technology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.,Center for Cancer Genome Discovery, Asan Institute for Life Science, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Eun Jeong Cho
- Department of Medical Science, Asan Medical Institute of Convergence Science and Technology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.,Center for Cancer Genome Discovery, Asan Institute for Life Science, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Jinyeong Lim
- Center for Cancer Genome Discovery, Asan Institute for Life Science, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.,Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Sung-Min Chun
- Center for Cancer Genome Discovery, Asan Institute for Life Science, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea. .,Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.
| | - Chang Ohk Sung
- Department of Medical Science, Asan Medical Institute of Convergence Science and Technology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea. .,Center for Cancer Genome Discovery, Asan Institute for Life Science, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea. .,Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
46
|
Setia N, Wang CX, Lager A, Maron S, Shroff S, Arndt N, Peterson B, Kupfer SS, Ma C, Misdraji J, Catenacci D, Hart J. Morphologic and molecular analysis of early-onset gastric cancer. Cancer 2021; 127:103-114. [PMID: 33048355 DOI: 10.1002/cncr.33213] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 07/21/2020] [Accepted: 07/28/2020] [Indexed: 01/19/2023]
Abstract
BACKGROUND Evidence suggests that early-onset gastric cancers are distinct from traditional gastric cancers; however, detailed genomic and morphologic characterization of these cancers has not been performed. METHODS Genomic analysis was performed for 81 patients with gastric cancer who were 50 years old or younger; pathology slides were available for 53 of these patients, and they were re-reviewed to perform a morphologic-molecular correlation analysis. The results were compared with corresponding cBioPortal data and The Cancer Genome Atlas (TCGA) analysis, which represent traditional gastric cancers. The TP53 molecular signature was established to determine the pattern of somatic mutational damage. Variants of potential germline origin were also identified from next-generation sequencing data. RESULTS A higher rate of CDH1 mutations (22.2% of early-onset gastric cancers vs 11.4% of traditional gastric cancers; P = .0042) but a similar rate of TP53 mutations (63% of early-onset gastric cancers vs 56.6% of traditional gastric cancers; P = .2674) were seen in early-onset cancers in comparison with traditional gastric cancers. The diffuse/mixed types correlated with the TCGA genomically stable type, and the remaining Lauren types correlated with the TCGA chromosomal instability type. Diffuse and indeterminate histologic types (overall survival, 26.25 months for the intestinal type, 20.5 months for the mixed type, 12.62 months for the diffuse type, and 9 months for the indeterminate type; P = .027) and the presence of a CDH1 gene mutation (overall survival, 9 months for mutant CDH1 and 22 months for wild-type CDH1; P = .013) significantly correlated with worse survival. The TP53 gene frequently showed transition mutations (65.5%) involving the CpG sites (49%). Variants of potential germline origin were seen in high-penetrance genes (CDH1 and APC) and moderate-penetrance genes (ATM, NBN, and MUTYH) in 9.9% of cancers. CONCLUSIONS Early-onset gastric cancer has distinct genomic alterations, such as CDH1 mutations, but shares with traditional gastric cancers a high frequency of TP53 mutations and the TP53 mutagenic signature. Diffuse and indeterminate histologic types and the presence of a CDH1 mutation are associated with worse overall survival. Endogenous factors leading to cytosine deamination and potential germline alterations in moderate-penetrance cancer susceptibility genes may be implicated in the pathogenesis of these cancers.
Collapse
Affiliation(s)
- Namrata Setia
- Department of Pathology, University of Chicago, Chicago, Illinois
| | - Cindy X Wang
- Department of Pathology, University of Chicago, Chicago, Illinois
| | - Angela Lager
- Section of Hematology/Oncology, Department of Medicine, University of Chicago, Chicago, Illinois
| | - Steve Maron
- Section of Medical Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Stuti Shroff
- Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts
| | - Nicole Arndt
- Section of Hematology/Oncology, Department of Medicine, University of Chicago, Chicago, Illinois
| | - Bryan Peterson
- Section of Hematology/Oncology, Department of Medicine, University of Chicago, Chicago, Illinois
| | - Sonia S Kupfer
- Department of Gastroenterology, University of Chicago, Chicago, Illinois
| | - Changqing Ma
- Department of Pathology, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Joseph Misdraji
- Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts
| | - Daniel Catenacci
- Section of Hematology/Oncology, Department of Medicine, University of Chicago, Chicago, Illinois
| | - John Hart
- Department of Pathology, University of Chicago, Chicago, Illinois
| |
Collapse
|
47
|
Li H, Sisoudiya SD, Martin-Giacalone BA, Khayat MM, Dugan-Perez S, Marquez-Do DA, Scheurer ME, Muzny D, Boerwinkle E, Gibbs RA, Chi YY, Barkauskas DA, Lo T, Hall D, Stewart DR, Schiffman JD, Skapek SX, Hawkins DS, Plon SE, Sabo A, Lupo PJ. Germline Cancer Predisposition Variants in Pediatric Rhabdomyosarcoma: A Report From the Children's Oncology Group. J Natl Cancer Inst 2020; 113:875-883. [PMID: 33372952 DOI: 10.1093/jnci/djaa204] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 10/15/2020] [Accepted: 12/09/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Several cancer-susceptibility syndromes are reported to underlie pediatric rhabdomyosarcoma (RMS); however, to our knowledge there have been no systematic efforts to characterize the heterogeneous genetic etiologies of this often-fatal malignancy. METHODS We performed exome-sequencing on germline DNA from 615 patients with newly diagnosed RMS consented through the Children's Oncology Group. We compared the prevalence of cancer predisposition variants in 63 autosomal-dominant cancer predisposition genes in these patients with population controls (n = 9963). All statistical tests were 2-sided. RESULTS We identified germline cancer predisposition variants in 45 RMS patients (7.3%; all FOXO1 fusion negative) across 15 autosomal dominant genes, which was statistically significantly enriched compared with controls (1.4%, P = 1.3 × 10-22). Specifically, 73.3% of the predisposition variants were found in predisposition syndrome genes previously associated with pediatric RMS risk, such as Li-Fraumeni syndrome (TP53) and neurofibromatosis type I (NF1). Notably, 5 patients had well-described oncogenic missense variants in HRAS (p.G12V and p.G12S) associated with Costello syndrome. Also, genetic etiology differed with histology, as germline variants were more frequent in embryonal vs alveolar RMS patients (10.0% vs 3.0%, P = .02). Although patients with a cancer predisposition variant tended to be younger at diagnosis (P = 9.9 × 10-4), 40.0% of germline variants were identified in those older than 3 years of age, which is in contrast to current genetic testing recommendations based on early age at diagnosis. CONCLUSIONS These findings demonstrate that genetic risk of RMS results from germline predisposition variants associated with a wide spectrum of cancer susceptibility syndromes. Germline genetic testing for children with RMS should be informed by RMS subtypes and not be limited to only young patients.
Collapse
Affiliation(s)
- He Li
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, USA
| | - Saumya D Sisoudiya
- Department of Pediatrics, Section of Hematology-Oncology, Baylor College of Medicine, Houston, TX, USA.,Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA.,Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Bailey A Martin-Giacalone
- Department of Pediatrics, Section of Hematology-Oncology, Baylor College of Medicine, Houston, TX, USA
| | - Michael M Khayat
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, USA.,Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Shannon Dugan-Perez
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, USA
| | - Deborah A Marquez-Do
- Department of Pediatrics, Section of Hematology-Oncology, Baylor College of Medicine, Houston, TX, USA
| | - Michael E Scheurer
- Department of Pediatrics, Section of Hematology-Oncology, Baylor College of Medicine, Houston, TX, USA.,Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Donna Muzny
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, USA
| | - Eric Boerwinkle
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, USA.,School of Public Health, the University of Texas Health Science Center, Houston, TX, USA
| | - Richard A Gibbs
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, USA.,Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Yueh-Yun Chi
- Children's Hospital Los Angeles, University of Southern California, Los Angeles, CA, USA
| | - Donald A Barkauskas
- QuadW Childhood Sarcoma Biostatistics and Annotation Office at the Children's Oncology Group, Monrovia, CA, USA.,Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Tammy Lo
- QuadW Childhood Sarcoma Biostatistics and Annotation Office at the Children's Oncology Group, Monrovia, CA, USA
| | - David Hall
- QuadW Childhood Sarcoma Biostatistics and Annotation Office at the Children's Oncology Group, Monrovia, CA, USA
| | - Douglas R Stewart
- Clinical Genetics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA
| | - Joshua D Schiffman
- Departments of Pediatrics and Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Stephen X Skapek
- Department of Pediatrics, the University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Douglas S Hawkins
- Division of Hematology-Oncology, Seattle Children's Hospital, University of Washington, Seattle, WA, USA
| | - Sharon E Plon
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, USA.,Department of Pediatrics, Section of Hematology-Oncology, Baylor College of Medicine, Houston, TX, USA.,Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Aniko Sabo
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, USA
| | - Philip J Lupo
- Department of Pediatrics, Section of Hematology-Oncology, Baylor College of Medicine, Houston, TX, USA.,Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
48
|
Mascarenhas E, Gelatti AC, Araújo LH, Baldotto C, Mathias C, Zukin M, Werutsky G, Pacheco P, Gomes R, de Castro G, Cordeiro de Lima VC. Comprehensive genomic profiling of Brazilian non-small cell lung cancer patients (GBOT 0118/LACOG0418). Thorac Cancer 2020; 12:580-587. [PMID: 33314759 PMCID: PMC7919136 DOI: 10.1111/1759-7714.13777] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 11/24/2020] [Accepted: 11/25/2020] [Indexed: 01/18/2023] Open
Abstract
Background The aim of this study was to carry out a descriptive analysis of the somatic genetic profile and co‐occurring mutations of non‐small cell lung cancer (NSCLC) samples from patients tested with comprehensive genomic profiling (CGP). Methods This was a retrospective cross‐sectional study of patients diagnosed with NSCLC from 2013 to 2018 in Brazil and whose samples were submitted to CGP (FoundationOne or FoundationACT) using either tumor or circulating tumor DNA (ctDNA) from plasma. Results We recovered 513 CGP results from patients, 457 (89.1%) of which were from tumors and 56 (10.9%) from plasma. The median age of patients was 64 years old, of which 51.6% were males. TP53 mutations were identified in 53.6% of tumor samples, KRAS mutations in 24.2%, EGFR activating mutations were detected in 22.5%, STK11 mutations in 11.6%, PIK3CA mutations in 8.8%, ALK rearrangements in 5.4%, BRAF mutations in 5.2%, and ERBB2 alterations in 4.9%. The most commonly comutated gene was TP53. TP53 p.R337H was observed in 4.3% of samples and was associated with somatic mutations in EGFR and ERBB2 (P < 0.00001). Tumor mutational burden (TMB) analysis was available for 80.5% of samples tested, and 5.5% of samples had high TMB (≥ 20 mutations/Mb). In conclusion, this retrospective analysis of genomic data from NSCLC patients obtained by CGP showed that common abnormalities such as EGFR mutations and ALK rearrangements had similar frequency to those previously described by other groups using others strategies. Additionally, our data confirm an association between TP53 p.R337H, supposedly germline in nature, and somatic mutations in genes of the HER family. Key points Significant findings of the study This is the first report of the prevalence of driver mutations in Brazilian NSCLC patients using comprehensive genomic profiling (CGP). The frequency of the most common driver mutations in this population was similar to that previously described in Brazil.
What this study adds TP53 was the most commonly comutated gene across samples. TP53 p.R337H was associated with somatic mutations in EGFR and ERBB2. Most samples had low TMB; only 5.5% of samples had high TMB.
Collapse
Affiliation(s)
- Eldsamira Mascarenhas
- Grupo Brasileiro de Oncologia Torácica, Porto Alegre, Brazil.,Oncologia D'or, Salvador, Brazil
| | - Ana Caroline Gelatti
- Grupo Brasileiro de Oncologia Torácica, Porto Alegre, Brazil.,Latin American Cooperative Oncology Group, Porto Alegre, Brazil.,Grupo Oncoclínicas, Porto Alegre, Brazil
| | - Luiz Henrique Araújo
- Grupo Brasileiro de Oncologia Torácica, Porto Alegre, Brazil.,Instituto COI de Educação e Pesquisa, Rio De Janeiro, Brazil.,Instituto Nacional do Câncer, Rio De Janeiro, Brazil
| | - Clarissa Baldotto
- Grupo Brasileiro de Oncologia Torácica, Porto Alegre, Brazil.,Oncologia D'or, Rio De Janeiro, Brazil
| | - Clarissa Mathias
- Grupo Brasileiro de Oncologia Torácica, Porto Alegre, Brazil.,NOB/Oncoclínicas, Salvador, Brazil
| | - Mauro Zukin
- Grupo Brasileiro de Oncologia Torácica, Porto Alegre, Brazil.,Oncologia D'or, Rio De Janeiro, Brazil
| | | | | | - Rafaela Gomes
- Latin American Cooperative Oncology Group, Porto Alegre, Brazil
| | - Gilberto de Castro
- Grupo Brasileiro de Oncologia Torácica, Porto Alegre, Brazil.,Instituto do Câncer do Estado de São Paulo, São Paulo, Brazil
| | | |
Collapse
|
49
|
Knapp E. "Somatic" Tumor Genomic Profiling and Potential Germline Implications: Ethical Considerations for Children with Cancer. THE JOURNAL OF LAW, MEDICINE & ETHICS : A JOURNAL OF THE AMERICAN SOCIETY OF LAW, MEDICINE & ETHICS 2020; 48:778-783. [PMID: 33404327 DOI: 10.1177/1073110520979389] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Affiliation(s)
- Esther Knapp
- Esther Knapp, M.D., M.B.E., is with the Division of Pediatric Hematology/Oncology, Department of Pediatrics at University of Louisville/Norton Children's Hospital
| |
Collapse
|
50
|
Mahon SM. Tumor Genomic Testing: Identifying Characteristics Associated With Germline Risk for Developing Malignancy. Clin J Oncol Nurs 2020; 24:623-626. [PMID: 33216058 DOI: 10.1188/20.cjon.623-626] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Tumor genomic testing is used primarily to facilitate the selection of the best possible treatment for a malignancy based on the genomic characteristics of the tumor. Germline genomic testing has implications for care and recommendations for cancer prevention and early detection for the patient and their family. Careful review of specific components of tumor genomic testing reports and of the family history of malignancy can help ensure that families with potential germline risk are identified and referred for genetic counseling and genetic testing.
Collapse
|