1
|
Sorscher S. A dual biomarker in non-small cell lung cancer that predicts Li Fraumeni syndrome : Lung cancer and Li Fraumeni. Fam Cancer 2024; 23:469-471. [PMID: 39235550 DOI: 10.1007/s10689-024-00418-8] [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: 05/13/2024] [Accepted: 08/22/2024] [Indexed: 09/06/2024]
Abstract
Non-small cell lung cancer is the most common cause of cancer death globally. When apparent incidental pathogenic germline variants (PGVs) are uncovered with routine next generation sequencing (NGS) of NSCLCs, germline testing (GT) is recommended to confirm that PGV. Because it is far more common that an uncovered tumor TP53 variant is related to a somatic event than an incidental PGV, however, GT for Li Fraumeni syndrome (LFS) is not recommended based solely on uncovering a NSCLC TP53 variant. Because nearly all tumor EGFR variants are also somatic in origin, GT is not recommended based solely on uncovering a tumor EGFR variant.However, there is evidence that patients with coexisting NSCLC variants in both EGFR and TP53 have significant likelihoods of having LFS. For patients with LFS, there are recommended measures for prevention and early detection of LFS-associated cancers and cascade GT of relatives for LFS. Although co-existing genetic variants in NSCLC are not currently used as a biomarker for GT to identify patients with PGVs, given the evidence reviewed here, select patients with NSCLCs that harbor this dual biomarker (i.e., co-existing TP53/EGFR variants) might reasonably be considered for GT for LFS.
Collapse
Affiliation(s)
- Steven Sorscher
- Not currently affiliated with any institution or company, Winston-Salem, NC, 27104, USA.
| |
Collapse
|
2
|
van de Haar J, Roepman P, Andre F, Balmaña J, Castro E, Chakravarty D, Curigliano G, Czarnecka AM, Dienstmann R, Horak P, Italiano A, Marchiò C, Monkhorst K, Pritchard CC, Reardon B, Russnes HEG, Sirohi B, Sosinsky A, Spanic T, Turnbull C, Van Allen E, Westphalen CB, Tamborero D, Mateo J. ESMO Recommendations on clinical reporting of genomic test results for solid cancers. Ann Oncol 2024; 35:954-967. [PMID: 39112111 DOI: 10.1016/j.annonc.2024.06.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 06/24/2024] [Accepted: 06/25/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND Genomic tumour profiling has a crucial role in the management of patients with solid cancers, as it helps selecting and prioritising therapeutic interventions based on prognostic and predictive biomarkers, as well as identifying markers of hereditary cancers. Harmonised approaches to interpret the results of genomic testing are needed to support physicians in their decision making, prevent inequalities in precision medicine and maximise patient benefit from available cancer management options. METHODS The European Society for Medical Oncology (ESMO) Translational Research and Precision Medicine Working Group assembled a group of international experts to propose recommendations for preparing clinical genomic reports for solid cancers. These recommendations aim to foster best practices in integrating genomic testing within clinical settings. After review of available evidence, several rounds of surveys and focused discussions were conducted to reach consensus on the recommendation statements. Only consensus recommendations were reported. Recommendation statements were graded in two tiers based on their clinical importance: level A (required to maintain common standards in reporting) and level B (optional but necessary to achieve ideal practice). RESULTS Genomics reports should present key information in a front page(s) followed by supplementary information in one or more appendices. Reports should be structured into sections: (i) patient and sample details; (ii) assay and data analysis characteristics; (iii) sample-specific assay performance and quality control; (iv) genomic alterations and their functional annotation; (v) clinical actionability assessment and matching to potential therapy indications; and (vi) summary of the main findings. Specific recommendations to prepare each of these sections are made. CONCLUSIONS We present a set of recommendations aimed at structuring genomics reports to enhance physician comprehension of genomic profiling results for solid cancers. Communication between ordering physicians and professionals reporting genomic data is key to minimise uncertainties and to optimise the impact of genomic tests in patient care.
Collapse
Affiliation(s)
- J van de Haar
- Department of Molecular Oncology & Immunology, Netherlands Cancer Institute, Amsterdam
| | - P Roepman
- Medical Department, Hartwig Medical Foundation, Amsterdam, The Netherlands
| | - F Andre
- INSERM U981, Gustave Roussy, Villejuif; Department of Cancer Medicine, Gustave Roussy, Villejuif; Faculty of Medicine, Université Paris-Saclay, Kremlin Bicêtre, France
| | - J Balmaña
- Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron Barcelona Hospital Campus, Barcelona
| | - E Castro
- Department of Medical Oncology, Hospital Universitario 12 de Octubre, Instituto de Investigación Sanitaria Hospital 12 de Octubre, Madrid, Spain
| | - D Chakravarty
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, USA
| | - G Curigliano
- Early Drug Development for Innovative Therapies Division, Istituto Europeo di Oncologia, IRCCS, Milan; Department of Oncology and Hemato-Oncology, University of Milano, Milan, Italy
| | - A M Czarnecka
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw; Department of Experimental Pharmacology, Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland
| | - R Dienstmann
- Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron Barcelona Hospital Campus, Barcelona; University of Vic-Central University of Catalonia, Vic, Spain; Oncoclínicas, São Paulo, Brazil
| | - P Horak
- Department of Translational Medical Oncology, National Center for Tumor Diseases (NCT) Heidelberg, Heidelberg; German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - A Italiano
- Early Phase Trials and Sarcoma Units, Institut Bergonie, Bordeaux; DITEP, Gustave Roussy, Villejuif; Faculty of Medicine, University of Bordeaux, Bordeaux, France
| | - C Marchiò
- Department of Medical Sciences, University of Turin, Turin; Division of Pathology, Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy
| | - K Monkhorst
- Department of Pathology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - C C Pritchard
- Department of Laboratory Medicine and Pathology, Brotman Baty Institute for Precision Medicine, University of Washington, Seattle
| | - B Reardon
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston; Division of Population Sciences, Dana-Farber Cancer Institute, Boston, USA
| | - H E G Russnes
- Department of Pathology, Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital, Radiumhospitalet, Oslo; Institute for Clinical Medicine, University of Oslo, Oslo, Norway
| | - B Sirohi
- Department of Medical Oncology, Vedanta Medical Research Foundation (BALCO Medical Centre), Raipur, India
| | | | - T Spanic
- Europa Donna Slovenia, Ljubljana, Slovenia
| | - C Turnbull
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London; The Royal Marsden NHS Foundation Trust, London, UK
| | - E Van Allen
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston; Division of Population Sciences, Dana-Farber Cancer Institute, Boston, USA; Harvard Medical School, Harvard University, Boston, USA
| | - C B Westphalen
- Comprehensive Cancer Center Munich & Department of Medicine III, University Hospital, LMU Munich, Munich; German Cancer Consortium (DKTK), Partner Site Munich, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - D Tamborero
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden.
| | - J Mateo
- Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron Barcelona Hospital Campus, Barcelona.
| |
Collapse
|
3
|
Meng F, Li H, Jin R, Yang A, Luo H, Li X, Wang P, Zhao Y, Chervova O, Tang K, Cheng S, Hu B, Li Y, Sheng J, Yang F, Carbone D, Chen K, Wang J. Spatial immunogenomic patterns associated with lymph node metastasis in lung adenocarcinoma. Exp Hematol Oncol 2024; 13:106. [PMID: 39468696 PMCID: PMC11514955 DOI: 10.1186/s40164-024-00574-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 10/13/2024] [Indexed: 10/30/2024] Open
Abstract
BACKGROUND Lung adenocarcinoma (LUAD) with lymph node (LN) metastasis is linked to poor prognosis, yet the underlying mechanisms remain largely undefined. This study aimed to elucidate the immunogenomic landscape associated with LN metastasis in LUAD. METHODS We employed broad-panel next-generation sequencing (NGS) on a cohort of 257 surgically treated LUAD patients to delineate the molecular landscape of primary tumors and identify actionable driver-gene alterations. Additionally, we used multiplex immunohistochemistry (mIHC) on a propensity score-matched cohort, which enabled us to profile the immune microenvironment of primary tumors in detail while preserving cellular metaclusters, interactions, and neighborhood functional units. By integrating data from NGS and mIHC, we successfully identified spatial immunogenomic patterns and developed a predictive model for LN metastasis, which was subsequently validated independently. RESULTS Our analysis revealed distinct immunogenomic alteration patterns associated with LN metastasis stages. Specifically, we observed increased mutation frequencies in genes such as PIK3CG and ATM in LN metastatic primary tumors. Moreover, LN positive primary tumors exhibited a higher presence of macrophage and regulatory T cell metaclusters, along with their enriched neighborhood units (p < 0.05), compared to LN negative tumors. Furthermore, we developed a novel predictive model for LN metastasis likelihood, designed to inform non-surgical treatment strategies, optimize personalized therapy plans, and potentially improve outcomes for patients who are ineligible for surgery. CONCLUSIONS This study offers a comprehensive analysis of the genetic and immune profiles in LUAD primary tumors with LN metastasis, identifying key immunogenomic patterns linked to metastatic progression. The predictive model derived from these insights marks a substantial advancement in personalized treatment, underscoring its potential to improve patient management.
Collapse
Affiliation(s)
- Fanjie Meng
- Department of Thoracic Surgery, Beijing Institute of Respiratory Medicine and Beijing Chao Yang Hospital, Capital Medical University, Beijing, China
| | - Hao Li
- Department of Thoracic Surgery, Institution of Thoracic Oncology, Peking University People's Hospital, No.11 Xizhimen South Street, Beijing, 100044, Xicheng District, China
| | - Ruoyi Jin
- Department of Thoracic Surgery, Institution of Thoracic Oncology, Peking University People's Hospital, No.11 Xizhimen South Street, Beijing, 100044, Xicheng District, China
- Thoracic Oncology Institute & Research Unit of Intelligence Diagnosis and Treatment in Early Non-Small Cell Lung Cancer, Peking University People's Hospital, Beijing, China
- Institute of Advanced Clinical Medicine, Peking University, Beijing, China
| | - Airong Yang
- Kanghui Biotechnology Co., Ltd, Shenyang, China
| | - Hao Luo
- Cancer Center, Daping Hospital Army Medical University, Chongqing, China
| | - Xiao Li
- Department of Thoracic Surgery, Institution of Thoracic Oncology, Peking University People's Hospital, No.11 Xizhimen South Street, Beijing, 100044, Xicheng District, China
| | - Peiyu Wang
- Department of Thoracic Surgery, Institution of Thoracic Oncology, Peking University People's Hospital, No.11 Xizhimen South Street, Beijing, 100044, Xicheng District, China
- Thoracic Oncology Institute & Research Unit of Intelligence Diagnosis and Treatment in Early Non-Small Cell Lung Cancer, Peking University People's Hospital, Beijing, China
- Institute of Advanced Clinical Medicine, Peking University, Beijing, China
| | - Yaxing Zhao
- Infinity Scope Biotechnology Co., Ltd., Hangzhou, China
| | - Olga Chervova
- University College London Cancer Institute, University College London, London, UK
| | - Kaicheng Tang
- Infinity Scope Biotechnology Co., Ltd., Hangzhou, China
| | - Sida Cheng
- Department of Thoracic Surgery, Institution of Thoracic Oncology, Peking University People's Hospital, No.11 Xizhimen South Street, Beijing, 100044, Xicheng District, China
| | - Bin Hu
- Department of Thoracic Surgery, Beijing Institute of Respiratory Medicine and Beijing Chao Yang Hospital, Capital Medical University, Beijing, China
| | - Yun Li
- Department of Thoracic Surgery, Institution of Thoracic Oncology, Peking University People's Hospital, No.11 Xizhimen South Street, Beijing, 100044, Xicheng District, China
| | - Jianpeng Sheng
- College of Artificial Intelligence, Nanjing University of Aeronautics and Astronautics, Nanjing, China
- Chinese Institutes for Medical Research, Beijing, China
| | - Fan Yang
- Department of Thoracic Surgery, Institution of Thoracic Oncology, Peking University People's Hospital, No.11 Xizhimen South Street, Beijing, 100044, Xicheng District, China
| | - David Carbone
- James Thoracic Oncology Center, Ohio State University, Columbus, USA
| | - Kezhong Chen
- Department of Thoracic Surgery, Institution of Thoracic Oncology, Peking University People's Hospital, No.11 Xizhimen South Street, Beijing, 100044, Xicheng District, China.
- Thoracic Oncology Institute & Research Unit of Intelligence Diagnosis and Treatment in Early Non-Small Cell Lung Cancer, Peking University People's Hospital, Beijing, China.
- Institute of Advanced Clinical Medicine, Peking University, Beijing, China.
| | - Jun Wang
- Department of Thoracic Surgery, Institution of Thoracic Oncology, Peking University People's Hospital, No.11 Xizhimen South Street, Beijing, 100044, Xicheng District, China.
| |
Collapse
|
4
|
van Schaik LF, Engelhardt EG, Wilthagen EA, Steeghs N, Fernández Coves A, Joore MA, van Harten WH, Retèl VP. Factors for a broad technology assessment of comprehensive genomic profiling in advanced cancer, a systematic review. Crit Rev Oncol Hematol 2024; 202:104441. [PMID: 39002790 DOI: 10.1016/j.critrevonc.2024.104441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 06/12/2024] [Accepted: 07/06/2024] [Indexed: 07/15/2024] Open
Abstract
Comprehensive Genomic Profiling (CGP) allows for the identification of many targets. Reimbursement decision-making is, however, challenging because besides the health benefits of on-label treatments and costs, other factors related to diagnostic and treatment pathways may also play a role. The aim of this study was to identify which other factors are relevant for the technology assessment of CGP and to summarize the available evidence for these factors. After a scoping search and two expert sessions, five factors were identified: feasibility, test journey, wider implications of diagnostic results, organisation of laboratories, and "scientific spillover". Subsequently, a systematic search identified 83 studies collecting mainly evidence for the factors "test journey" and "wider implications of diagnostic results". Its nature was, however, of limited value for decision-making. We recommend the use of comparative strategies, uniformity in outcome definitions, and the inclusion of a comprehensive set of factors in future evidence generation.
Collapse
Affiliation(s)
- L F van Schaik
- Division of Psychosocial Research and Epidemiology, The Netherlands Cancer Institute, P.O. Box 90103, Amsterdam 1006 BE, the Netherlands; Erasmus School of Health Policy and Management, Erasmus University Rotterdam, Rotterdam, the Netherlands.
| | - E G Engelhardt
- Division of Psychosocial Research and Epidemiology, The Netherlands Cancer Institute, P.O. Box 90103, Amsterdam 1006 BE, the Netherlands.
| | - E A Wilthagen
- Scientific Information Service, Netherlands Cancer Institute, Antoni van Leeuwenhoek, Plesmanlaan 121, Amsterdam CX 1066, the Netherlands.
| | - N Steeghs
- Department of Medical Oncology, Netherlands Cancer Institute, Plesmanlaan 121, Amsterdam CX 1066, the Netherlands.
| | - A Fernández Coves
- Department of Clinical Epidemiology and Medical Technology Assessment (KEMTA), P. Debyelaan 25, Oxford Building, P.O. Box 5800a, Maastricht, Limburg, the Netherlands; Care and Public Health Research Institute (CAPHRI), Maastricht University, Maastricht, The Netherlands.
| | - M A Joore
- Department of Clinical Epidemiology and Medical Technology Assessment (KEMTA), P. Debyelaan 25, Oxford Building, P.O. Box 5800a, Maastricht, Limburg, the Netherlands; Care and Public Health Research Institute (CAPHRI), Maastricht University, Maastricht, The Netherlands.
| | - W H van Harten
- Division of Psychosocial Research and Epidemiology, The Netherlands Cancer Institute, P.O. Box 90103, Amsterdam 1006 BE, the Netherlands; Department of Health Technology and Services Research, University of Twente, Enschede, the Netherlands.
| | - V P Retèl
- Division of Psychosocial Research and Epidemiology, The Netherlands Cancer Institute, P.O. Box 90103, Amsterdam 1006 BE, the Netherlands; Erasmus School of Health Policy and Management, Erasmus University Rotterdam, Rotterdam, the Netherlands.
| |
Collapse
|
5
|
Ibrahim MB, Adnani Y, Clément GJ, Lacroix L, Loriot Y, Besse B, Massard C, Rouleau E. Assessing actionability and incidental findings of germline variants in two precision oncology trials. Eur J Cancer 2024; 210:114256. [PMID: 39154486 DOI: 10.1016/j.ejca.2024.114256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 07/18/2024] [Accepted: 07/18/2024] [Indexed: 08/20/2024]
Abstract
INTRODUCTION High-throughput sequencing techniques have revolutionized oncology. Paired germline-tumor DNA analysis has emerged as a comprehensive strategy to uncover actionable alterations in advanced cancer patients (ACP) enrolled in precision oncology trials. However, challenges persist in variant interpretation and managing incidental germline findings. METHODS We conducted a study involving 288 ACP from MOSCATO (NCT01566019) and MATCHR (NCT02517892) trials to assess germline variants impacting cancer-related genes. Germline DNA sequencing was performed using a panel of 250 cancer-related genes, and the results were discussed during tumor molecular board sessions. RESULTS Germline pathogenic variants (PV) were classified according to the ESCAT classification. Lung cancer (36.8 %), followed by prostate (18.4 %) and breast cancer (17.7 %), comprised the most prevalent tumor types. PVs were found in 12.5 % of patients. Most PVs were classified as ESCAT X (63.9 %), highlighting limited therapeutic actionability. Notably,2 % of patients had actionable variants (ESCAT I-A/II-A). Incidental findings included 7.3 % of patients with PVs in cancer-predisposition genes, with 2.4 % having very high-risk potential, necessitating mandatory oncogenetic counseling. Nearly one in five patients (21.9 %) had at least one VUS. DISCUSSION Our study underscores the significance of germline sequencing in identifying actionable alterations and the need for improved variant interpretation as well as pretest counseling plans in precision oncology trials.
Collapse
Affiliation(s)
- Maria Baz Ibrahim
- Department of oncogenetics and biochemistry, Paul Brousse University Hospital (APHP), 12 Av. Paul Vaillant Couturier, 94800 Villejuif, France.
| | - Yahia Adnani
- Bioinformatics Core Facility, Gustave Roussy Cancer Campus, Villejuif, France
| | | | - Ludovic Lacroix
- Molecular Biopathology, Medical Biology and Pathology Department, Gustave-Roussy Cancer Campus, Villejuif, France
| | - Yohann Loriot
- Department of Medical Oncology, Gustave Roussy, 94805 Villejuif, France
| | - Benjamin Besse
- Department of Medical Oncology, Gustave Roussy, 94805 Villejuif, France
| | | | - Etienne Rouleau
- Cancer Genetics Laboratory, Medical Biology and Pathology Department, Gustave-Roussy Cancer Campus, Villejuif, France
| |
Collapse
|
6
|
Shimada S, Yamada T, Minamoto A, Matsukawa M, Yabe I, Tada H, Oda K, Ueki A, Higashigawa S, Morikawa M, Sato Y, Hirasawa A, Ogawa M, Kondo T, Yoshioka M, Kanai M, Muto M, Kosugi S. Nationwide survey of the secondary findings in cancer genomic profiling: survey including liquid biopsy. J Hum Genet 2024:10.1038/s10038-024-01294-x. [PMID: 39289525 DOI: 10.1038/s10038-024-01294-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 08/23/2024] [Accepted: 08/28/2024] [Indexed: 09/19/2024]
Abstract
We surveyed the status of the secondary finding (SF) disclosure in comprehensive genome profiling (CGP) in 2020. The situation has changed: increase in the number of hospitals that provide CGP, an update to the Comprehensive Tumor Genomic Profiling: Materials for Review of Secondary Findings (CTGPMRSF), and the addition of a liquid biopsy test, FoundationOne® Liquid CDx (F1L). Moreover, the actual situation was unclear because the 2020 survey did not include all designated and cooperative hospitals. Herein, we conducted a questionnaire survey of all designated-core, designated, and cooperative hospitals to identify the current status and challenges concerning SF in the CGP in 2022. A total of 82.1% of the hospitals responded and 77.7% of the response was from cooperative hospitals. Approximately 80% of the hospitals used CTGPMRSF. SF disclosure, confirmatory test implementation, and SF confirmation rates were 12.4%, 31.6%, and 46.6% for FoundationOne® CDx (F1CDx), respectively, and 6.8%, 31.8%, and 70.7% for F1L, respectively. The implementation rate of the confirmatory test was substantially higher in hospitals with genetic experts and in hospitals that could conduct confirmatory tests on the same day. Our survey provides insight into how SF is handled in Japan. The percentage of cases leading to confirmatory tests has gradually increased, although challenges such as insurance coverage limitations and varied understanding of SF among patients and healthcare providers persist. With the increasing use of whole-genome analysis, our findings will provide valuable insights into establishing an effective SF disclosure system.
Collapse
Affiliation(s)
- Saki Shimada
- Department of Medical Ethics and Medical Genetics, Kyoto University Graduate School of Medicine, Kyoto, Japan
- Clinical Genetics Center, Kansai Medical University Hospital, Osaka, Japan
| | - Takahiro Yamada
- Department of Medical Ethics and Medical Genetics, Kyoto University Graduate School of Medicine, Kyoto, Japan.
- Division of Clinical Genetics, Hokkaido University Hospital, Sapporo, Japan.
- Secondary Findings Working Group (SFWG), Liaison Council for Core Hospitals for Cancer Genomic Medicine, Tokyo, Japan.
| | - Akari Minamoto
- Clinical Genetics Unit, Kyoto University Hospital, Kyoto, Japan
| | - Manami Matsukawa
- Department of Medical Ethics and Medical Genetics, Kyoto University Graduate School of Medicine, Kyoto, Japan
- Secondary Findings Working Group (SFWG), Liaison Council for Core Hospitals for Cancer Genomic Medicine, Tokyo, Japan
- Department of Genetic Medicine and Services, National Cancer Center Hospital, Tokyo, Japan
| | - Ichiro Yabe
- Division of Clinical Genetics, Hokkaido University Hospital, Sapporo, Japan
- Secondary Findings Working Group (SFWG), Liaison Council for Core Hospitals for Cancer Genomic Medicine, Tokyo, Japan
| | - Hiroshi Tada
- Secondary Findings Working Group (SFWG), Liaison Council for Core Hospitals for Cancer Genomic Medicine, Tokyo, Japan
- Department of Breast and Endocrine Surgical Oncology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Katsutoshi Oda
- Secondary Findings Working Group (SFWG), Liaison Council for Core Hospitals for Cancer Genomic Medicine, Tokyo, Japan
- Division of Integrative Genomics, The University of Tokyo, Tokyo, Japan
| | - Arisa Ueki
- Secondary Findings Working Group (SFWG), Liaison Council for Core Hospitals for Cancer Genomic Medicine, Tokyo, Japan
- Center for Medical Genetics, Keio Cancer Center, Keio University School of Medicine, Tokyo, Japan
| | - Satomi Higashigawa
- Secondary Findings Working Group (SFWG), Liaison Council for Core Hospitals for Cancer Genomic Medicine, Tokyo, Japan
- Division of Clinical Genetics, Shizuoka Cancer Center, Shizuoka, Japan
| | - Maki Morikawa
- Secondary Findings Working Group (SFWG), Liaison Council for Core Hospitals for Cancer Genomic Medicine, Tokyo, Japan
- Medical Genome Center, Nagoya University Hospital, Nagoya, Japan
| | - Yuki Sato
- Secondary Findings Working Group (SFWG), Liaison Council for Core Hospitals for Cancer Genomic Medicine, Tokyo, Japan
- Department of Genetic Counseling, Osaka University Hospital, Osaka, Japan
| | - Akira Hirasawa
- Secondary Findings Working Group (SFWG), Liaison Council for Core Hospitals for Cancer Genomic Medicine, Tokyo, Japan
- Department of Clinical Genomic Medicine, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Masanobu Ogawa
- Department of Medical Ethics and Medical Genetics, Kyoto University Graduate School of Medicine, Kyoto, Japan
- Secondary Findings Working Group (SFWG), Liaison Council for Core Hospitals for Cancer Genomic Medicine, Tokyo, Japan
- Department of Clinical Genetics and Medicine, Kyushu University Hospital, Fukuoka, Japan
| | - Tomohiro Kondo
- Secondary Findings Working Group (SFWG), Liaison Council for Core Hospitals for Cancer Genomic Medicine, Tokyo, Japan
- Department of Medical Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Masahiro Yoshioka
- Secondary Findings Working Group (SFWG), Liaison Council for Core Hospitals for Cancer Genomic Medicine, Tokyo, Japan
- Department of Medical Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Masashi Kanai
- Secondary Findings Working Group (SFWG), Liaison Council for Core Hospitals for Cancer Genomic Medicine, Tokyo, Japan
- Department of Medical Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
- Cancer Center, Kansai Medical University Hospital, Osaka, Japan
| | - Manabu Muto
- Department of Medical Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Shinji Kosugi
- Department of Medical Ethics and Medical Genetics, Kyoto University Graduate School of Medicine, Kyoto, Japan
- Secondary Findings Working Group (SFWG), Liaison Council for Core Hospitals for Cancer Genomic Medicine, Tokyo, Japan
| |
Collapse
|
7
|
Lococo F, De Paolis E, Evangelista J, Dell'Amore A, Giannarelli D, Chiappetta M, Campanella A, Sassorossi C, Cancellieri A, Calabrese F, Conca A, Vita E, Minucci A, Bria E, Castello A, Urbani A, Rea F, Margaritora S, Scambia G. Comparative Analysis of Comprehensive Genomic Profile in Thymomas and Recurrent Thymomas Reveals Potentially Actionable Mutations for Target Therapies. Int J Mol Sci 2024; 25:9560. [PMID: 39273507 PMCID: PMC11394945 DOI: 10.3390/ijms25179560] [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: 07/15/2024] [Revised: 08/31/2024] [Accepted: 09/02/2024] [Indexed: 09/15/2024] Open
Abstract
Molecular profiles of thymomas and recurrent thymomas are far from being defined. Herein, we report an analysis of a comprehensive genetic profile (CGP) in a highly selected cohort of recurrent thymomas. Among a cohort of 426 thymomas, the tissue was available in 23 recurrent tumors for matching the biomolecular results obtained from primary and relapse samples. A control group composed of non-recurrent thymoma patients was selected through a propensity score match analysis. CGP was performed using the NGS Tru-SightOncology assay to evaluate TMB, MSI, and molecular alterations in 523 genes. CGP does not differ when comparing initial tumor with tumor relapse. A significantly higher frequency of cell cycle control genes alterations (100.0% vs. 57.1%, p = 0.022) is detected in patients with early recurrence (<32 months) compared to late recurrent cases. The CGPs were similar in recurrent thymomas and non-recurrent thymomas. Finally, based on NGS results, an off-label treatment or clinical trial could be potentially proposed in >50% of cases (oncogenic Tier-IIC variants). In conclusion, CGPs do not substantially differ between initial tumor vs. tumor recurrence and recurrent thymomas vs. non-recurrent thymomas. Cell cycle control gene alterations are associated with an early recurrence after thymectomy. Multiple target therapies are potentially available by performing a comprehensive CGP, suggesting that a precision medicine approach on these patients could be further explored.
Collapse
Affiliation(s)
- Filippo Lococo
- Thoracic Surgery, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
- Thoracic Surgery, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Elisa De Paolis
- Clinical Chemistry, Biochemistry and Molecular Biology Operations (UOC), Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
- Departmental Unit of Molecular and Genomic Diagnostics, Genomics Core Facility, Gemelli Science and Technology Park (G-STeP), Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Jessica Evangelista
- Thoracic Surgery, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
- Thoracic Surgery, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Andrea Dell'Amore
- Thoracic Surgery Unit, Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, 35122 Padova, Italy
| | - Diana Giannarelli
- Epidemiology and Biostatistics Facility, Gemelli Science and Technology Park (G-STeP), Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Marco Chiappetta
- Thoracic Surgery, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Annalisa Campanella
- Thoracic Surgery, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Carolina Sassorossi
- Thoracic Surgery, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | | | - Fiorella Calabrese
- Pathology Unit, Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, 35122 Padova, Italy
| | - Alessandra Conca
- Pathology Unit, Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, 35122 Padova, Italy
| | - Emanuele Vita
- UOSD Oncologia Toraco-Polmonare, Comprehensive Cancer Center, Fondazione Policlinico Universitario Ago-stino Gemelli IRCCS, 00168 Rome, Italy
| | - Angelo Minucci
- Pathology Unit, Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, 35122 Padova, Italy
| | - Emilio Bria
- UOSD Oncologia Toraco-Polmonare, Comprehensive Cancer Center, Fondazione Policlinico Universitario Ago-stino Gemelli IRCCS, 00168 Rome, Italy
- UOC Oncologia Medica, Ospedale Isola Tiberina-Gemelli Isola, 00186 Roma, Italy
| | - Angelo Castello
- Nuclear Medicine Department, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Andrea Urbani
- Clinical Chemistry, Biochemistry and Molecular Biology Operations (UOC), Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Federico Rea
- Thoracic Surgery Unit, Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, 35122 Padova, Italy
| | - Stefano Margaritora
- Thoracic Surgery, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
- Thoracic Surgery, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Giovanni Scambia
- Division of Gynecologic Oncology, Fondazione Policlinico Universitario A. Gemelli-IRCCS, 00168 Rome, Italy
| |
Collapse
|
8
|
Rives TA, Collard J, Li N, Yan D, Dietrich CS, Miller RW, Ueland FR, Pickarski J, Kolesar JM. Clinical Utility of Molecular Tumor Board Review for Identification of Possible Germline Pathogenic Variants on Tumor Next-Generation Sequencing Reports. JCO Precis Oncol 2024; 8:e2400301. [PMID: 39259913 PMCID: PMC11404756 DOI: 10.1200/po.24.00301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 07/10/2024] [Accepted: 07/31/2024] [Indexed: 09/13/2024] Open
Abstract
PURPOSE Tumor next-generation sequencing (NGS) testing identifies possible germline pathogenic variants (PGPVs), creating a dilemma for appropriate recognition, triage, and management. The objective of this study was to determine the clinical utility of an institutional molecular tumor board (MTB) in assessing tumor NGS reports for PGPVs. METHODS Our institutional MTB reviews all NGS reports to provide treatment and further testing recommendations, including genetic counseling referral and consideration of genetic testing (GC/GT). We studied the patients reviewed by the MTB who were recommended for GC/GT to determine the frequency of referral to a GC, germline test completion, rate of pathogenic germline variants (PGVs), factors related to PGVs, and germline conversion rate (GCR). RESULTS Of the 2,355 patients reviewed by the MTB during the study period, 609 (25.9%) had a recommendation for GC/GT. Of the 609 with a GC/GT recommendation, only 181 (29.7%) were referred for GC/GT by their treating physicians, and only 107 (17.6%) completed GT. Of the 107 patients completing GT, 29 (26%) had a confirmed PGV. The only factors significantly associated with PGVs were testing due to a PGPV and higher mean variant allele fraction on the tumor NGS. Only 40 patients with a GC/GT recommendation (14.3%) due to a PGPV completed GT; however, the GCR was 42.5% (n = 17/40). CONCLUSION The MTB review of PGPV is clinically valuable, identifying PGPV in 12% of patients undergoing tumor NGS and a GCR of 42.5%. Rates of GC/GT completion were relatively low due to under-referral by treating physicians. Given the high GCR, the authors encourage institutional algorithms to help increase GC/GT rates for patients found to have PGPV following tumor NGS testing.
Collapse
Affiliation(s)
- Taylor A Rives
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of Kentucky, Lexington, KY
| | - James Collard
- Markey Cancer Center, University of Kentucky, Lexington, KY
| | - Ning Li
- Department of Biostatistics, College of Public Health, University of Kentucky, Lexington, KY
| | - Donglin Yan
- Department of Biostatistics, College of Public Health, University of Kentucky, Lexington, KY
| | - Charles S Dietrich
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of Kentucky, Lexington, KY
- Markey Cancer Center, University of Kentucky, Lexington, KY
| | - Rachel W Miller
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of Kentucky, Lexington, KY
- Markey Cancer Center, University of Kentucky, Lexington, KY
| | - Frederick R Ueland
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of Kentucky, Lexington, KY
- Markey Cancer Center, University of Kentucky, Lexington, KY
| | | | - Jill M Kolesar
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of Kentucky, Lexington, KY
- Markey Cancer Center, University of Kentucky, Lexington, KY
- College of Pharmacy, University of Kentucky, Lexington, KY
| |
Collapse
|
9
|
Luo M, Wong D, Zelley K, Wu J, Schubert J, Denenberg EH, Fanning EA, Chen J, Gallo D, Golenberg N, Patel M, Conlin LK, Maxwell KN, Wertheim GB, Surrey LF, Zhong Y, Brodeur GM, MacFarland SP, Li MM. Identification of TP53 germline variants in pediatric patients undergoing tumor testing: strategy and prevalence. J Natl Cancer Inst 2024; 116:1356-1365. [PMID: 38702830 DOI: 10.1093/jnci/djae102] [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: 01/08/2024] [Revised: 04/08/2024] [Accepted: 04/30/2024] [Indexed: 05/06/2024] Open
Abstract
BACKGROUND TP53 alterations are common in certain pediatric cancers, making identification of putative germline variants through tumor genomic profiling crucial for disease management. METHODS We analyzed TP53 alterations in 3123 tumors from 2788 pediatric patients sequenced using tumor-only or tumor-normal paired panels. Germline confirmatory testing was performed when indicated. Somatic and germline variants were classified based on published guidelines. RESULTS In 248 tumors from 222 patients, 284 tier 1/2 TP53 sequence and small copy number variants were detected. Following germline classification, 86.6% of 142 unique variants were pathogenic or likely pathogenic. Confirmatory testing on 118 patients revealed germline TP53 variants in 28 of them (23 pathogenic or likely pathogenic and 5 of uncertain significance), suggesting a minimum Li-Fraumeni syndrome incidence of 0.8% (23/2788) in this cohort, 10.4% (23/222) in patients with TP53 variant-carrying tumors, and 19.5% (23/118) with available normal samples. About 25% (7/28) of patients with germline TP53 variants did not meet Li-Fraumeni syndrome diagnostic or testing criteria, while 20.9% (28/134) with confirmed or inferred somatic origins did. TP53 biallelic inactivation occurred in 75% of germline carrier tumors and was also prevalent in other groups, causing an elevated tumor-observed variant allelic fraction. Somatic evidence, however, including low variant allele fraction correctly identified only 27.8% (25/90) of patients with confirmed somatic TP53 variants. CONCLUSION The high incidence and variable phenotype of Li-Fraumeni syndrome in this cohort highlights the importance of assessing germline status of TP53 variants identified in all pediatric tumors. Without clear somatic evidence, distinguishing somatic from germline origins is challenging. Classifying germline and somatic variants should follow appropriate guidelines.
Collapse
Affiliation(s)
- Minjie Luo
- Division of Genomic Diagnostics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Derek Wong
- Division of Genomic Diagnostics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Kristin Zelley
- Division of Oncology, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Jinhua Wu
- Division of Genomic Diagnostics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Jeffery Schubert
- Division of Genomic Diagnostics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Elizabeth H Denenberg
- Division of Genomic Diagnostics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Elizabeth A Fanning
- Division of Genomic Diagnostics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Jiani Chen
- Division of Genomic Diagnostics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Daniel Gallo
- Division of Genomic Diagnostics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Netta Golenberg
- Division of Genomic Diagnostics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Maha Patel
- Division of Genomic Diagnostics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Laura K Conlin
- Division of Genomic Diagnostics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Kara N Maxwell
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, PA, USA
| | - Gerald B Wertheim
- Division of Genomic Diagnostics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Lea F Surrey
- Division of Genomic Diagnostics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Yiming Zhong
- Division of Genomic Diagnostics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Garrett M Brodeur
- Division of Oncology, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Suzanne P MacFarland
- Division of Oncology, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Marilyn M Li
- Division of Genomic Diagnostics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Division of Oncology, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
10
|
Jacobs MF, Stoffel EM. Genetic and other risk factors for pancreatic ductal adenocarcinoma (PDAC). Fam Cancer 2024; 23:221-232. [PMID: 38573398 DOI: 10.1007/s10689-024-00372-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Accepted: 03/07/2024] [Indexed: 04/05/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is often diagnosed at an advanced stage, resulting in poor prognosis and low 5-year survival rates. While early evidence suggests increased long-term survival in those with screen-detected resectable cancers, surveillance imaging is currently only recommended for individuals with a lifetime risk of PDAC ≥ 5%. Identification of risk factors for PDAC provides opportunities for early detection, risk reducing interventions, and targeted therapies, thus potentially improving patient outcomes. Here, we summarize modifiable and non-modifiable risk factors for PDAC. We review hereditary cancer syndromes associated with risk for PDAC and their implications for patients and their relatives. In addition, other biologically relevant pathways and environmental and lifestyle risk factors are discussed. Future work may focus on elucidating additional genetic, environmental, and lifestyle risk factors that may modify PDAC risk to continue to identify individuals at increased risk for PDAC who may benefit from surveillance and risk reducing interventions.
Collapse
Affiliation(s)
- Michelle F Jacobs
- Division of Genetic Medicine, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Elena M Stoffel
- Division of Gastroenterology, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan, USA.
| |
Collapse
|
11
|
Tung N, Ricker C, Messersmith H, Balmaña J, Domchek S, Stoffel EM, Almhanna K, Arun B, Chavarri-Guerra Y, Cohen SA, Cragun D, Crew KD, Hall MJ, Idos G, Lopez G, Pal T, Pirzadeh-Miller S, Pritchard C, Rana HQ, Swami U, Vidal GA. Selection of Germline Genetic Testing Panels in Patients With Cancer: ASCO Guideline. J Clin Oncol 2024; 42:2599-2615. [PMID: 38759122 DOI: 10.1200/jco.24.00662] [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: 03/29/2024] [Accepted: 04/03/2024] [Indexed: 05/19/2024] Open
Abstract
PURPOSE To guide use of multigene panels for germline genetic testing for patients with cancer. METHODS An ASCO Expert Panel convened to develop recommendations on the basis of a systematic review of guidelines, consensus statements, and studies of germline and somatic genetic testing. RESULTS Fifty-two guidelines and consensus statements met eligibility criteria for the primary search; 14 studies were identified for Clinical Question 4. RECOMMENDATIONS Patients should have a family history taken and recorded that includes details of cancers in first- and second-degree relatives and the patient's ethnicity. When more than one gene is relevant based on personal and/or family history, multigene panel testing should be offered. When considering what genes to include in the panel, the minimal panel should include the more strongly recommended genes from Table 1 and may include those less strongly recommended. A broader panel may be ordered when the potential benefits are clearly identified, and the potential harms from uncertain results should be mitigated. Patients who meet criteria for germline genetic testing should be offered germline testing regardless of results from tumor testing. Patients who would not normally be offered germline genetic testing based on personal and/or family history criteria but who have a pathogenic or likely pathogenic variant identified by tumor testing in a gene listed in Table 2 under the outlined circumstances should be offered germline testing.Additional information is available at www.asco.org/molecular-testing-and-biomarkers-guidelines.
Collapse
Affiliation(s)
- Nadine Tung
- Beth Israel Deaconess Medical Center, Sharon, MA
| | | | | | | | | | | | | | - Banu Arun
- University of Texas MD Anderson Cancer Center, Houston, TX
| | - Yanin Chavarri-Guerra
- Instituto Nacional de Ciencias Médicas y Nutrición, Salvador Zubirán, Mexico City, Mexico
| | | | | | | | | | - Gregory Idos
- City of Hope Comprehensive Cancer Center, Duarte, CA
| | - Ghecemy Lopez
- USC Norris Comprehensive Cancer Center, Los Angeles, CA
| | - Tuya Pal
- Vanderbilt-Ingram Cancer Center, Nashville, TN
| | - Sara Pirzadeh-Miller
- Simmons Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX
| | | | | | - Umang Swami
- Huntsman Cancer Institute at the University of Utah, Salt Lake City, UT
| | - Gregory A Vidal
- The West Cancer Center and Research Institute and The University of Tennessee Health Sciences Center, Germantown, TN
| |
Collapse
|
12
|
Pongcharoen S, Kaewsringam N, Somaparn P, Roytrakul S, Maneerat Y, Pintha K, Topanurak S. Immunopeptidomics in the cancer immunotherapy era. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2024; 5:801-817. [PMID: 39280250 PMCID: PMC11390293 DOI: 10.37349/etat.2024.00249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Accepted: 06/06/2024] [Indexed: 09/18/2024] Open
Abstract
Cancer is the primary cause of death worldwide, and conventional treatments are painful, complicated, and have negative effects on healthy cells. However, cancer immunotherapy has emerged as a promising alternative. Principle of cancer immunotherapy is the re-activation of T-cell to combat the tumor that presents the peptide antigen on major histocompatibility complex (MHC). Those peptide antigens are identified with the set of omics technology, proteomics, genomics, and bioinformatics, which referred to immunopeptidomics. Indeed, immunopeptidomics can identify the neoantigens that are very useful for cancer immunotherapies. This review explored the use of immunopeptidomics for various immunotherapies, i.e., peptide-based vaccines, immune checkpoint inhibitors, oncolytic viruses, and chimeric antigen receptor T-cell. We also discussed how the diversity of neoantigens allows for the discovery of novel antigenic peptides while post-translationally modified peptides diversify the overall peptides binding to MHC or so-called MHC ligandome. The development of immunopeptidomics is keeping up-to-date and very active, particularly for clinical application. Immunopeptidomics is expected to be fast, accurate and reliable for the application for cancer immunotherapies.
Collapse
Affiliation(s)
- Sutatip Pongcharoen
- Division of Immunology, Department of Medicine, Faculty of Medicine, Naresuan University, Phitsanulok 65000, Thailand
| | - Nongphanga Kaewsringam
- Department of Molecular Tropical Medicine and Genetics, Faculty of Tropical Medicine, Mahidol University, Bangkok 10400, Thailand
| | - Poorichaya Somaparn
- Center of Excellence in Systems Biology, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
| | - Sittiruk Roytrakul
- Functional Proteomics Technology Laboratory, National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Khlong Nueng, Khlong Luang 12120, Pathum Thani, Thailand
| | - Yaowapa Maneerat
- Department of Tropical Pathology, Faculty of Tropical Medicine, Mahidol University, Bangkok 10400, Thailand
| | - Komsak Pintha
- Division of Biochemistry, School of Medical Sciences, University of Phayao, Phayao 56000, Thailand
| | - Supachai Topanurak
- Department of Molecular Tropical Medicine and Genetics, Faculty of Tropical Medicine, Mahidol University, Bangkok 10400, Thailand
| |
Collapse
|
13
|
Kiyozumi Y, Matsubayashi H, Todaka A, Ashida R, Nishimura S, Kado N, Higashigawa S, Harada R, Ishihara E, Horiuchi Y, Honda G, Kenmotsu H, Serizawa M, Urakami K. Two Japanese families with familial pancreatic cancer with suspected pathogenic variants of CDKN2A: a case report. Hered Cancer Clin Pract 2024; 22:11. [PMID: 38961426 PMCID: PMC11223274 DOI: 10.1186/s13053-024-00283-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 06/24/2024] [Indexed: 07/05/2024] Open
Abstract
BACKGROUND Germline mutations in CDKN2A result in Familial Atypical Multiple Mole Melanoma Syndrome (FAMMM) (OMIM #155,601), which is associated with an increased risk of pancreatic ductal adenocarcinoma and melanoma. FAMMM has been reported globally, but it is quite rare in Japan. We report two families with familial pancreatic cancer with suspected pathogenic variants of CDKN2A that were incidentally identified through comprehensive genomic profiling. CASE PRESENTATION The first case is a 74-year-old woman with a diagnosis of pancreatic carcinoma with multiple liver metastases. She had family histories of pancreatic cancer, but no personal or family history of malignant melanoma. Whole exon sequencing detected a germline CDKN2A variant evaluated as likely pathogenic. The results were disclosed to her daughters after she died, and the same CDKN2A variant was detected in one of the daughter. The daughter was referred to a nearby hospital for her clinical management. The second case is a 65-year-old man with pancreatic ductal adenocarcinoma. He had family histories of pancreatic cancer, but no personal or family history of malignant melanoma. He underwent a comprehensive genomic profiling test using pancreatic cancer tissue, and detected a presumed germline pathogenic variant of CDKN2A. Germline testing confirmed the same CDKN2A variant. Genetic analysis of his relatives produced negative results. Other blood relatives are scheduled for genetic analysis in the future. We report two families with familial pancreatic cancer with suspected pathogenic variants of CDKN2A that were incidentally identified through comprehensive genomic profiling. CONCLUSIONS In current Japanese precision medicine, comprehensive genetic analysis can reveal rare genetic syndromes and offer us the opportunity to provide health management for patients and their relatives. However, gene-specific issues are raised in terms of the evaluation of a variant's pathogenicity and the extent of surveillance of the at-risk organs due to a lack of genetic and clinical data concerning CDKN2A variant carriers in Japan.
Collapse
Affiliation(s)
- Yoshimi Kiyozumi
- Division of Genetic Medicine Promotion, Shizuoka Cancer Center, 1007, Shimonagakubo, Nagaizumi, Suntogun, Shizuoka, 411-8777, Japan
| | - Hiroyuki Matsubayashi
- Division of Genetic Medicine Promotion, Shizuoka Cancer Center, 1007, Shimonagakubo, Nagaizumi, Suntogun, Shizuoka, 411-8777, Japan.
| | - Akiko Todaka
- Division of Gastrointestinal Oncology, Shizuoka Cancer Center, 1007, Shimonagakubo, Nagaizumi, Suntogun, Shizuoka, 411-8777, Japan
- Department of Medical Oncology and Hematology, Oita University Faculty of Medicine, 1-1, Idaigaoka, Hasama-machi, Yufu, Oita, 879-5593, Japan
| | - Ryo Ashida
- Division of Hepato-Biliary-Pancreatic Surgery, Shizuoka Cancer Center, 1007, Shimonagakubo, Nagaizumi, Suntogun, Shizuoka, 411-8777, Japan
| | - Seiichiro Nishimura
- Division of Genetic Medicine Promotion, Shizuoka Cancer Center, 1007, Shimonagakubo, Nagaizumi, Suntogun, Shizuoka, 411-8777, Japan
| | - Nobuhiro Kado
- Division of Genetic Medicine Promotion, Shizuoka Cancer Center, 1007, Shimonagakubo, Nagaizumi, Suntogun, Shizuoka, 411-8777, Japan
| | - Satomi Higashigawa
- Division of Genetic Medicine Promotion, Shizuoka Cancer Center, 1007, Shimonagakubo, Nagaizumi, Suntogun, Shizuoka, 411-8777, Japan
| | - Rina Harada
- Division of Genetic Medicine Promotion, Shizuoka Cancer Center, 1007, Shimonagakubo, Nagaizumi, Suntogun, Shizuoka, 411-8777, Japan
| | - Eiko Ishihara
- Division of Genetic Medicine Promotion, Shizuoka Cancer Center, 1007, Shimonagakubo, Nagaizumi, Suntogun, Shizuoka, 411-8777, Japan
| | - Yasue Horiuchi
- Division of Genetic Medicine Promotion, Shizuoka Cancer Center, 1007, Shimonagakubo, Nagaizumi, Suntogun, Shizuoka, 411-8777, Japan
- Research Institute, of Shizuoka Cancer Center, 1007, Shimonagakubo, Nagaizumi, Suntogun, Shizuoka, 411-8777, Japan
| | - Goichi Honda
- Division of Genetic Medicine Promotion, Shizuoka Cancer Center, 1007, Shimonagakubo, Nagaizumi, Suntogun, Shizuoka, 411-8777, Japan
- Research Institute, of Shizuoka Cancer Center, 1007, Shimonagakubo, Nagaizumi, Suntogun, Shizuoka, 411-8777, Japan
| | - Hirotsugu Kenmotsu
- Division of Genetic Medicine Promotion, Shizuoka Cancer Center, 1007, Shimonagakubo, Nagaizumi, Suntogun, Shizuoka, 411-8777, Japan
| | - Masakuni Serizawa
- Research Institute, of Shizuoka Cancer Center, 1007, Shimonagakubo, Nagaizumi, Suntogun, Shizuoka, 411-8777, Japan
| | - Kenichi Urakami
- Research Institute, of Shizuoka Cancer Center, 1007, Shimonagakubo, Nagaizumi, Suntogun, Shizuoka, 411-8777, Japan
| |
Collapse
|
14
|
Balciuniene J, Ning Y, Lazarus HM, Aikawa V, Sherpa S, Zhang Y, Morrissette JJD. Cancer cytogenetics in a genomics world: Wedding the old with the new. Blood Rev 2024; 66:101209. [PMID: 38852016 DOI: 10.1016/j.blre.2024.101209] [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: 03/15/2024] [Revised: 04/30/2024] [Accepted: 05/02/2024] [Indexed: 06/10/2024]
Abstract
Since the discovery of the Philadelphia chromosome in 1960, cytogenetic studies have been instrumental in detecting chromosomal abnormalities that can inform cancer diagnosis, treatment, and risk assessment efforts. The initial expansion of cancer cytogenetics was with fluorescence in situ hybridization (FISH) to assess submicroscopic alterations in dividing or non-dividing cells and has grown into the incorporation of chromosomal microarrays (CMA), and next generation sequencing (NGS). These molecular technologies add additional dimensions to the genomic assessment of cancers by uncovering cytogenetically invisible molecular markers. Rapid technological and bioinformatic advances in NGS are so promising that the idea of performing whole genome sequencing as part of routine patient care may soon become economically and logistically feasible. However, for now cytogenetic studies continue to play a major role in the diagnostic testing and subsequent assessments in leukemia with other genomic studies serving as complementary testing options for detection of actionable genomic abnormalities. In this review, we discuss the role of conventional cytogenetics (karyotyping, chromosome analysis) and FISH studies in hematological malignancies, highlighting the continued clinical utility of these techniques, the subtleties and complexities that are relevant to treating physicians and the unique strengths of cytogenetics that cannot yet be paralleled by the current high-throughput molecular technologies. Additionally, we describe how CMA, optical genome mapping (OGM), and NGS detect abnormalities that were beyond the capacity of cytogenetic studies and how an integrated approach (broad molecular testing) can contribute to the detection of actionable targets and variants in malignancies. Finally, we discuss advances in the field of genomic testing that are bridging the advantages of individual (single) cell based cytogenetic testing and broad genomic testing.
Collapse
Affiliation(s)
- Jorune Balciuniene
- Division of Genomic Diagnostics, Department of Pathology and Laboratory Medicine, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Yi Ning
- Department of Pathology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Hillard M Lazarus
- Department of Medicine, Case Western Reserve University, Cleveland, OH, United States of America
| | - Vania Aikawa
- Division of Precision and Computational Diagnostics, Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Sarina Sherpa
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Yanming Zhang
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Jennifer J D Morrissette
- Division of Precision and Computational Diagnostics, Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
15
|
Mosele MF, Westphalen CB, Stenzinger A, Barlesi F, Bayle A, Bièche I, Bonastre J, Castro E, Dienstmann R, Krämer A, Czarnecka AM, Meric-Bernstam F, Michiels S, Miller R, Normanno N, Reis-Filho J, Remon J, Robson M, Rouleau E, Scarpa A, Serrano C, Mateo J, André F. Recommendations for the use of next-generation sequencing (NGS) for patients with advanced cancer in 2024: a report from the ESMO Precision Medicine Working Group. Ann Oncol 2024; 35:588-606. [PMID: 38834388 DOI: 10.1016/j.annonc.2024.04.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 04/17/2024] [Accepted: 04/18/2024] [Indexed: 06/06/2024] Open
Abstract
BACKGROUND Advancements in the field of precision medicine have prompted the European Society for Medical Oncology (ESMO) Precision Medicine Working Group to update the recommendations for the use of tumour next-generation sequencing (NGS) for patients with advanced cancers in routine practice. METHODS The group discussed the clinical impact of tumour NGS in guiding treatment decision using the ESMO Scale for Clinical Actionability of molecular Targets (ESCAT) considering cost-effectiveness and accessibility. RESULTS As for 2020 recommendations, ESMO recommends running tumour NGS in advanced non-squamous non-small-cell lung cancer, prostate cancer, colorectal cancer, cholangiocarcinoma, and ovarian cancer. Moreover, it is recommended to carry out tumour NGS in clinical research centres and under specific circumstances discussed with patients. In this updated report, the consensus within the group has led to an expansion of the recommendations to encompass patients with advanced breast cancer and rare tumours such as gastrointestinal stromal tumours, sarcoma, thyroid cancer, and cancer of unknown primary. Finally, ESMO recommends carrying out tumour NGS to detect tumour-agnostic alterations in patients with metastatic cancers where access to matched therapies is available. CONCLUSION Tumour NGS is increasingly expanding its scope and application within oncology with the aim of enhancing the efficacy of precision medicine for patients with cancer.
Collapse
Affiliation(s)
- M F Mosele
- INSERM U981, Gustave Roussy, Villejuif; Department of Cancer Medicine, Gustave Roussy, Villejuif, France
| | - C B Westphalen
- Comprehensive Cancer Center Munich & Department of Medicine III, University Hospital, LMU Munich, Munich
| | - A Stenzinger
- Institute of Pathology, University Hospital Heidelberg and Center for Personalized Medicine (ZPM), Heidelberg, Germany
| | - F Barlesi
- INSERM U981, Gustave Roussy, Villejuif; Department of Cancer Medicine, Gustave Roussy, Villejuif, France; Faculty of Medicine, Université Paris-Saclay, Kremlin Bicêtre
| | - A Bayle
- Faculty of Medicine, Université Paris-Saclay, Kremlin Bicêtre; Drug Development Department (DITEP), Gustave Roussy, Villejuif; Oncostat U1018, Inserm, Université Paris-Saclay, labeled Ligue Contre le Cancer, Villejuif; Service de Biostatistique et Epidémiologie, Gustave Roussy, Villejuif
| | - I Bièche
- Department of Genetics, Institut Curie, INSERM U1016, Université Paris Cité, Paris, France
| | - J Bonastre
- Oncostat U1018, Inserm, Université Paris-Saclay, labeled Ligue Contre le Cancer, Villejuif; Service de Biostatistique et Epidémiologie, Gustave Roussy, Villejuif
| | - E Castro
- Department of Medical Oncology, Hospital Universitario 12 de Octubre, Instituto de Investigación Sanitaria Hospital 12 de Octubre, Madrid
| | - R Dienstmann
- Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron Barcelona Hospital Campus, Barcelona; University of Vic-Central University of Catalonia, Vic, Spain; Oncoclínicas, São Paulo, Brazil
| | - A Krämer
- Clinical Cooperation Unit Molecular Hematology/Oncology, German Cancer Research Center (DKFZ), Heidelberg; Department of Internal Medicine V, University of Heidelberg, Heidelberg, Germany
| | - A M Czarnecka
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw; Department of Experimental Pharmacology, Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland
| | - F Meric-Bernstam
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - S Michiels
- Oncostat U1018, Inserm, Université Paris-Saclay, labeled Ligue Contre le Cancer, Villejuif; Service de Biostatistique et Epidémiologie, Gustave Roussy, Villejuif
| | - R Miller
- Department of Medical Oncology, University College London, London; Department of Medical Oncology, St Bartholomew's Hospital, London, UK
| | - N Normanno
- Scientific Directorate, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - J Reis-Filho
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York
| | - J Remon
- Department of Cancer Medicine, Gustave Roussy, Villejuif, France
| | - M Robson
- Breast Medicine and Clinical Genetics Services, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, USA
| | - E Rouleau
- Tumor Genetics Service, Medical Biology and Pathology Department, Gustave Roussy, Villejuif, France
| | - A Scarpa
- Section of Pathology, Department of Diagnostics and Public Health, University of Verona-School of Medicine, Verona, Italy
| | - C Serrano
- Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron Barcelona Hospital Campus, Barcelona
| | - J Mateo
- Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron Barcelona Hospital Campus, Barcelona
| | - F André
- INSERM U981, Gustave Roussy, Villejuif; Department of Cancer Medicine, Gustave Roussy, Villejuif, France; Faculty of Medicine, Université Paris-Saclay, Kremlin Bicêtre.
| |
Collapse
|
16
|
West EC, Chiappetta M, Mattingly AA, Congedo MT, Evangelista J, Campanella A, Sassorossi C, Flamini S, Rossi T, Pistoni M, Abenavoli L, Margaritora S, Lococo F, Boccuto L. BRCA1-associated protein 1: Tumor predisposition syndrome and Kury-Isidor syndrome, from genotype-phenotype correlation to clinical management. Clin Genet 2024; 105:589-595. [PMID: 38506155 DOI: 10.1111/cge.14507] [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: 11/29/2023] [Revised: 01/27/2024] [Accepted: 02/07/2024] [Indexed: 03/21/2024]
Abstract
The BAP1 tumor suppressor gene encodes a deubiquitinase enzyme involved in several cellular activities, including DNA repair and apoptosis. Germline pathogenic variants in BAP1 have been associated with heritable conditions including BAP1 tumor predisposition syndrome 1 (BAP1-TPDS1) and a neurodevelopmental disorder known as Kury-Isidor syndrome (KURIS). Both these conditions are caused by monoallelic, dominant alterations of BAP1 but have never been reported in the same subject or family, suggesting a mutually exclusive genotype-phenotype correlation. This distinction is extremely important considering the early onset and aggressive nature of the types of cancer reported in individuals with TPDS1. Genetic counseling in subjects with germline BAP1 variants is fundamental to predicting the effect of the variant and the expected phenotype, assessing the potential risk of developing cancer for the tested subject and the family members who may carry the same variant and providing the multidisciplinary clinical team with the proper information to establish precise surveillance and management protocols.
Collapse
Affiliation(s)
- Elizabeth Casey West
- Healthcare Genetics and Genomics, School of Nursing, Clemson University, Clemson, South Carolina, USA
| | - Marco Chiappetta
- Thoracic Surgery, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Aubrey Anne Mattingly
- Healthcare Genetics and Genomics, School of Nursing, Clemson University, Clemson, South Carolina, USA
| | - Maria Teresa Congedo
- Thoracic Surgery, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Jessica Evangelista
- Thoracic Surgery, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Annalisa Campanella
- Thoracic Surgery, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Carolina Sassorossi
- Thoracic Surgery, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Sara Flamini
- Thoracic Surgery, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Teresa Rossi
- Laboratory of Translational Research, Azienda USL-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Mariaelena Pistoni
- Laboratory of Translational Research, Azienda USL-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Ludovico Abenavoli
- Department of Health Sciences, University "Magna Græcia", Catanzaro, Italy
| | - Stefano Margaritora
- Thoracic Surgery, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
- Catholic University of the Sacred Heart, Rome, Italy
| | - Filippo Lococo
- Thoracic Surgery, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
- Catholic University of the Sacred Heart, Rome, Italy
| | - Luigi Boccuto
- Healthcare Genetics and Genomics, School of Nursing, Clemson University, Clemson, South Carolina, USA
| |
Collapse
|
17
|
Sanabria-Salas MC, Pedroza-Duran A, Díaz-Casas SE, Nuñez Lemus M, Grillo-Ardila CF, Briceño-Morales X, García-Mora M, Ángel-Aristizábal J, Mariño Lozano IF, Suarez Rodríguez RA, Guzmán Abisaab LH. Management and Clinical Outcomes of Breast Cancer in Women Diagnosed with Hereditary Cancer Syndromes in a Clinic-Based Sample from Colombia. Cancers (Basel) 2024; 16:2020. [PMID: 38893140 PMCID: PMC11171067 DOI: 10.3390/cancers16112020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 05/08/2024] [Accepted: 05/09/2024] [Indexed: 06/21/2024] Open
Abstract
This study aimed to investigate prognosis and survival differences in 82 breast cancer patients with germline pathogenic/likely pathogenic variants (PVs) treated and followed at the Breast Unit of the Instituto Nacional de Cancerología, Colombia (INC-C) between 2018 and 2021. Median age at diagnosis was 46 years, with 62.2% presenting locally advanced tumors, 47.6% histological grade 3, and 35.4% with triple-negative breast cancer (TNBC) subtype. Most carriers, 74.4% (61/82), had PVs in known breast cancer susceptibility genes (i.e., "associated gene carriers" group, considered inherited breast cancer cases): BRCA2 (30), BRCA1 (14), BARD1 (4), RAD51D (3), TP53 (2), PALB2 (2), ATM (2), CHEK2 (1), RAD51C (1), NF1 (1), and PTEN (1). BRCA1-2 represented 53.7%, and homologous recombination DNA damage repair (HR-DDR) genes associated with breast cancer risk accounted for 15.9%. Patients with PVs in non-breast-cancer risk genes were combined in a different category (21/82; 25.6%) (i.e., "non-associated gene carriers" group, considered other breast cancer cases). Median follow-up was 38.1 months, and 24% experienced recurrence, with 90% being distant. The 5-year Disease-Free Survival (DFS) for inherited breast cancer cases was 66.5%, and for other breast cancer cases it was 88.2%. In particular, for carriers of PVs in the BRCA2 gene, it was 37.6%. The 5-year Overall Survival (OS) rates ranged from 68.8% for those with PVs in BRCA2 to 100% for those with PVs in other HR-DDR genes. Further studies are crucial for understanding tumor behavior and therapy response differences among Colombian breast cancer patients with germline PVs.
Collapse
Affiliation(s)
- María Carolina Sanabria-Salas
- Instituto Nacional de Cancerología, Calle 1 N. 9-85, Bogotá 111511, Colombia; (A.P.-D.); (S.E.D.-C.); (M.N.L.); (X.B.-M.); (M.G.-M.); (J.Á.-A.); (I.F.M.L.); (R.A.S.R.); (L.H.G.A.)
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, 610 University Avenue, Toronto, ON M5G 2C1, Canada
| | - Ana Pedroza-Duran
- Instituto Nacional de Cancerología, Calle 1 N. 9-85, Bogotá 111511, Colombia; (A.P.-D.); (S.E.D.-C.); (M.N.L.); (X.B.-M.); (M.G.-M.); (J.Á.-A.); (I.F.M.L.); (R.A.S.R.); (L.H.G.A.)
| | - Sandra E. Díaz-Casas
- Instituto Nacional de Cancerología, Calle 1 N. 9-85, Bogotá 111511, Colombia; (A.P.-D.); (S.E.D.-C.); (M.N.L.); (X.B.-M.); (M.G.-M.); (J.Á.-A.); (I.F.M.L.); (R.A.S.R.); (L.H.G.A.)
| | - Marcela Nuñez Lemus
- Instituto Nacional de Cancerología, Calle 1 N. 9-85, Bogotá 111511, Colombia; (A.P.-D.); (S.E.D.-C.); (M.N.L.); (X.B.-M.); (M.G.-M.); (J.Á.-A.); (I.F.M.L.); (R.A.S.R.); (L.H.G.A.)
| | - Carlos F. Grillo-Ardila
- Department of Obstetrics & Gynecology, School of Medicine, Universidad Nacional de Colombia, Avenida Carrera 30 N. 45-3, Bogotá 111321, Colombia;
| | - Ximena Briceño-Morales
- Instituto Nacional de Cancerología, Calle 1 N. 9-85, Bogotá 111511, Colombia; (A.P.-D.); (S.E.D.-C.); (M.N.L.); (X.B.-M.); (M.G.-M.); (J.Á.-A.); (I.F.M.L.); (R.A.S.R.); (L.H.G.A.)
| | - Mauricio García-Mora
- Instituto Nacional de Cancerología, Calle 1 N. 9-85, Bogotá 111511, Colombia; (A.P.-D.); (S.E.D.-C.); (M.N.L.); (X.B.-M.); (M.G.-M.); (J.Á.-A.); (I.F.M.L.); (R.A.S.R.); (L.H.G.A.)
| | - Javier Ángel-Aristizábal
- Instituto Nacional de Cancerología, Calle 1 N. 9-85, Bogotá 111511, Colombia; (A.P.-D.); (S.E.D.-C.); (M.N.L.); (X.B.-M.); (M.G.-M.); (J.Á.-A.); (I.F.M.L.); (R.A.S.R.); (L.H.G.A.)
| | - Iván Fernando Mariño Lozano
- Instituto Nacional de Cancerología, Calle 1 N. 9-85, Bogotá 111511, Colombia; (A.P.-D.); (S.E.D.-C.); (M.N.L.); (X.B.-M.); (M.G.-M.); (J.Á.-A.); (I.F.M.L.); (R.A.S.R.); (L.H.G.A.)
| | - Raúl Alexis Suarez Rodríguez
- Instituto Nacional de Cancerología, Calle 1 N. 9-85, Bogotá 111511, Colombia; (A.P.-D.); (S.E.D.-C.); (M.N.L.); (X.B.-M.); (M.G.-M.); (J.Á.-A.); (I.F.M.L.); (R.A.S.R.); (L.H.G.A.)
| | - Luis Hernán Guzmán Abisaab
- Instituto Nacional de Cancerología, Calle 1 N. 9-85, Bogotá 111511, Colombia; (A.P.-D.); (S.E.D.-C.); (M.N.L.); (X.B.-M.); (M.G.-M.); (J.Á.-A.); (I.F.M.L.); (R.A.S.R.); (L.H.G.A.)
| |
Collapse
|
18
|
Laguna JC, Pastor B, Nalda I, Hijazo-Pechero S, Teixido C, Potrony M, Puig-Butillé JA, Mezquita L. Incidental pathogenic germline alterations detected through liquid biopsy in patients with solid tumors: prevalence, clinical utility and implications. Br J Cancer 2024; 130:1420-1431. [PMID: 38532104 PMCID: PMC11059286 DOI: 10.1038/s41416-024-02607-9] [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: 09/15/2023] [Revised: 01/14/2024] [Accepted: 01/25/2024] [Indexed: 03/28/2024] Open
Abstract
Liquid biopsy, a minimally invasive approach for detecting tumor biomarkers in blood, has emerged as a leading-edge technique in cancer precision medicine. New evidence has shown that liquid biopsies can incidentally detect pathogenic germline variants (PGVs) associated with cancer predisposition, including in patients with a cancer for which genetic testing is not recommended. The ability to detect these incidental PGV in cancer patients through liquid biopsy raises important questions regarding the management of this information and its clinical implications. This incidental identification of PGVs raises concerns about cancer predisposition and the potential impact on patient management, not only in terms of providing access to treatment based on the tumor molecular profiling, but also the management of revealing genetic predisposition in patients and families. Understanding how to interpret this information is essential to ensure proper decision-making and to optimize cancer treatment and prevention strategies. In this review we provide a comprehensive summary of current evidence of incidental PGVs in cancer predisposition genes identified by liquid biopsy in patients with cancer. We critically review the methodological considerations of liquid biopsy as a tool for germline diagnosis, clinical utility and potential implications for cancer prevention, treatment, and research.
Collapse
Affiliation(s)
- Juan Carlos Laguna
- Medical Oncology Department, Hospital Clinic of Barcelona, Barcelona, Spain
- Laboratory of Translational Genomics and Targeted Therapies in Solid Tumors, IDIBAPS, Barcelona, Spain
| | - Belén Pastor
- Medical Oncology Department, Hospital Clinic of Barcelona, Barcelona, Spain
| | - Irene Nalda
- Medical Oncology Department, Hospital Clinic of Barcelona, Barcelona, Spain
- Laboratory of Translational Genomics and Targeted Therapies in Solid Tumors, IDIBAPS, Barcelona, Spain
| | - Sara Hijazo-Pechero
- Preclinical and Experimental Research in Thoracic Tumors (PRETT), Oncobell, Bellvitge Biomedical Research Institute (IDIBELL), l'Hospitalet de Llobregat, Barcelona, Spain
| | - Cristina Teixido
- Laboratory of Translational Genomics and Targeted Therapies in Solid Tumors, IDIBAPS, Barcelona, Spain
- Department of Medicine, University of Barcelona, Barcelona, Spain
- Department of Pathology, Hospital Clinic of Barcelona, Barcelona, Spain
| | - Miriam Potrony
- Biochemistry and Molecular Genetics Department, Hospital Clínic of Barcelona, IDIBAPS, Barcelona, Spain
- CIBER of Rare Diseases (CIBERER), Barcelona, Spain
| | - Joan Antón Puig-Butillé
- CIBER of Rare Diseases (CIBERER), Barcelona, Spain
- Molecular Biology CORE, Hospital Clínic of Barcelona, IDIBAPS, Barcelona, Spain
| | - Laura Mezquita
- Medical Oncology Department, Hospital Clinic of Barcelona, Barcelona, Spain.
- Laboratory of Translational Genomics and Targeted Therapies in Solid Tumors, IDIBAPS, Barcelona, Spain.
- Department of Medicine, University of Barcelona, Barcelona, Spain.
| |
Collapse
|
19
|
McDevitt T, Durkie M, Arnold N, Burghel GJ, Butler S, Claes KBM, Logan P, Robinson R, Sheils K, Wolstenholme N, Hanson H, Turnbull C, Hume S. EMQN best practice guidelines for genetic testing in hereditary breast and ovarian cancer. Eur J Hum Genet 2024; 32:479-488. [PMID: 38443545 PMCID: PMC11061103 DOI: 10.1038/s41431-023-01507-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 11/07/2023] [Accepted: 11/21/2023] [Indexed: 03/07/2024] Open
Abstract
Hereditary Breast and Ovarian Cancer (HBOC) is a genetic condition associated with increased risk of cancers. The past decade has brought about significant changes to hereditary breast and ovarian cancer (HBOC) diagnostic testing with new treatments, testing methods and strategies, and evolving information on genetic associations. These best practice guidelines have been produced to assist clinical laboratories in effectively addressing the complexities of HBOC testing, while taking into account advancements since the last guidelines were published in 2007. These guidelines summarise cancer risk data from recent studies for the most commonly tested high and moderate risk HBOC genes for laboratories to refer to as a guide. Furthermore, recommendations are provided for somatic and germline testing services with regards to clinical referral, laboratory analyses, variant interpretation, and reporting. The guidelines present recommendations where 'must' is assigned to advocate that the recommendation is essential; and 'should' is assigned to advocate that the recommendation is highly advised but may not be universally applicable. Recommendations are presented in the form of shaded italicised statements throughout the document, and in the form of a table in supplementary materials (Table S4). Finally, for the purposes of encouraging standardisation and aiding implementation of recommendations, example report wording covering the essential points to be included is provided for the most common HBOC referral and reporting scenarios. These guidelines are aimed primarily at genomic scientists working in diagnostic testing laboratories.
Collapse
Affiliation(s)
- Trudi McDevitt
- Department of Clinical Genetics, Children's Health Ireland at Crumlin, Dublin, Ireland.
| | - Miranda Durkie
- Sheffield Diagnostic Genetics Service, North East and Yorkshire Genomic Laboratory Hub, Sheffield Children's NHS Foundation Trust Western Bank, Sheffield, UK
| | - Norbert Arnold
- UKSH Campus Kiel, Gynecology and Obstetrics, Institut of Clinical Chemistry, Institut of Clinical Molecular Biology, Kiel, Germany
| | - George J Burghel
- Manchester University NHS Foundation Trust, North West Genomic Laboratory Hub, Manchester, UK
| | - Samantha Butler
- Central and South Genomic Laboratory Hub, West Midlands Regional Genetics Laboratory, Birmingham Women's and Children's NHS Foundation Trust, Birmingham, UK
| | | | - Peter Logan
- HSCNI / Belfast Trust Laboratories, Regional Molecular Diagnostics Service, Belfast, Northern Ireland
| | - Rachel Robinson
- Leeds Teaching Hospitals NHS Trust, Genetics Department, Leeds, UK
| | | | | | - Helen Hanson
- St George's University Hospitals NHS Foundation Trust, Clinical Genetics, London, UK
| | | | - Stacey Hume
- University of British Columbia, Pathology and Laboratory Medicine, Vancouver, British Columbia, Canada
| |
Collapse
|
20
|
Olmos D, Lorente D, Alameda D, Cattrini C, Romero-Laorden N, Lozano R, Lopez-Casas PP, Jambrina A, Capone C, Vanden Broecke AM, Trevisan M, Van Sanden S, Jürgens A, Herrera-Imbroda B, Castro E. Treatment patterns and outcomes in metastatic castration-resistant prostate cancer patients with and without somatic or germline alterations in homologous recombination repair genes. Ann Oncol 2024; 35:458-472. [PMID: 38417742 DOI: 10.1016/j.annonc.2024.01.011] [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: 10/04/2023] [Revised: 01/30/2024] [Accepted: 01/31/2024] [Indexed: 03/01/2024] Open
Abstract
BACKGROUND Although germline BRCA mutations have been associated with adverse outcomes in prostate cancer (PC), understanding of the association between somatic/germline alterations in homologous recombination repair (HRR) genes and treatment outcomes in metastatic castration-resistant PC (mCRPC) is limited. The aim of this study was to investigate the prevalence and outcomes associated with somatic/germline HRR alterations, particularly BRCA1/2, in patients initiating first-line (1L) mCRPC treatment with androgen receptor signalling inhibitors (ARSi) or taxanes. PATIENTS AND METHODS Data from 729 mCRPC patients were pooled for CAPTURE from four multicentre observational studies. Eligibility required 1L treatment with ARSi or taxanes, adequate tumour samples and biomarker panel results. Patients underwent paired normal and tumour DNA analyses by next-generation sequencing using a custom gene panel including ATM, BRCA1, BRCA2, BRIP1, CDK12, CHEK2, FANCA, HDAC2, PALB2, RAD51B and RAD54L. Patients were divided into subgroups based on somatic/germline alteration(s): with BRCA1/2 mutations (BRCA); with HRR mutations except BRCA1/2 (HRR non-BRCA); and without HRR alterations (non-HRR). Patients without BRCA1/2 mutations were classified as non-BRCA. Radiographic progression-free survival (rPFS), progression-free survival 2 (PFS2) and overall survival (OS) were assessed. RESULTS Of 729 patients, 96 (13.2%), 127 (17.4%) and 506 (69.4%) were in the BRCA, HRR non-BRCA and non-HRR subgroups, respectively. BRCA patients performed significantly worse for all outcomes than non-HRR or non-BRCA patients (P < 0.05), while PFS2 and OS were significantly shorter for BRCA than HRR non-BRCA patients (P < 0.05). HRR non-BRCA patients also had significantly worse rPFS, PFS2 and OS than non-HRR patients. Exploratory analyses suggested that for BRCA patients, there were no significant differences in outcomes associated with 1L treatment choice (ARSi or taxanes) or with the somatic/germline origin of the alterations. CONCLUSIONS Worse outcomes were observed for mCRPC patients in the BRCA subgroup compared with non-BRCA subgroups, either HRR non-BRCA or non-HRR. Despite its heterogeneity, the HRR non-BRCA subgroup presented worse outcomes than the non-HRR subgroup. Screening early for HRR mutations, especially BRCA1/2, is crucial in improving mCRPC patient prognosis.
Collapse
Affiliation(s)
- D Olmos
- Hospital Universitario 12 de Octubre, Instituto de Investigación Sanitaria Hospital 12 de Octubre, Madrid.
| | - D Lorente
- Instituto Valenciano de Oncología, Valencia; Hospital Provincial de Castellón, Castellón de la Plana
| | - D Alameda
- Instituto de Investigación Biomédica de Málaga, Málaga, Spain
| | - C Cattrini
- Maggiore della Carità University Hospital, Novara, Italy
| | - N Romero-Laorden
- Cátedra UAM-Fundación Instituto Roche de Medicina Personalizada de Precisión, Hospital Universitario de La Princesa, Madrid
| | - R Lozano
- Hospital Universitario de Salamanca, Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain
| | - P P Lopez-Casas
- Hospital Universitario 12 de Octubre, Instituto de Investigación Sanitaria Hospital 12 de Octubre, Madrid
| | - A Jambrina
- Hospital Universitario 12 de Octubre, Instituto de Investigación Sanitaria Hospital 12 de Octubre, Madrid
| | - C Capone
- Janssen Inc., Issy-les-Moulineaux, France
| | | | - M Trevisan
- Janssen Pharmaceuticals, Zug, Switzerland
| | | | | | - B Herrera-Imbroda
- Instituto de Investigación Biomédica de Málaga, Málaga, Spain; Hospital Universitario Virgen de la Victoria, Málaga, Spain
| | - E Castro
- Hospital Universitario 12 de Octubre, Instituto de Investigación Sanitaria Hospital 12 de Octubre, Madrid; Instituto de Investigación Biomédica de Málaga, Málaga, Spain.
| |
Collapse
|
21
|
Casolino R, Beer PA, Chakravarty D, Davis MB, Malapelle U, Mazzarella L, Normanno N, Pauli C, Subbiah V, Turnbull C, Westphalen CB, Biankin AV. Interpreting and integrating genomic tests results in clinical cancer care: Overview and practical guidance. CA Cancer J Clin 2024; 74:264-285. [PMID: 38174605 DOI: 10.3322/caac.21825] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 11/07/2023] [Accepted: 11/27/2023] [Indexed: 01/05/2024] Open
Abstract
The last decade has seen rapid progress in the use of genomic tests, including gene panels, whole-exome sequencing, and whole-genome sequencing, in research and clinical cancer care. These advances have created expansive opportunities to characterize the molecular attributes of cancer, revealing a subset of cancer-associated aberrations called driver mutations. The identification of these driver mutations can unearth vulnerabilities of cancer cells to targeted therapeutics, which has led to the development and approval of novel diagnostics and personalized interventions in various malignancies. The applications of this modern approach, often referred to as precision oncology or precision cancer medicine, are already becoming a staple in cancer care and will expand exponentially over the coming years. Although genomic tests can lead to better outcomes by informing cancer risk, prognosis, and therapeutic selection, they remain underutilized in routine cancer care. A contributing factor is a lack of understanding of their clinical utility and the difficulty of results interpretation by the broad oncology community. Practical guidelines on how to interpret and integrate genomic information in the clinical setting, addressed to clinicians without expertise in cancer genomics, are currently limited. Building upon the genomic foundations of cancer and the concept of precision oncology, the authors have developed practical guidance to aid the interpretation of genomic test results that help inform clinical decision making for patients with cancer. They also discuss the challenges that prevent the wider implementation of precision oncology.
Collapse
Affiliation(s)
- Raffaella Casolino
- Wolfson Wohl Cancer Research Center, School of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - Philip A Beer
- Wolfson Wohl Cancer Research Center, School of Cancer Sciences, University of Glasgow, Glasgow, UK
- Hull York Medical School, York, UK
| | | | - Melissa B Davis
- Department of Surgery, Weill Cornell Medicine, New York City, New York, USA
| | - Umberto Malapelle
- Department of Public Health, University of Naples Federico II, Naples, Italy
| | - Luca Mazzarella
- Laboratory of Translational Oncology and Division of Gastrointestinal Medical Oncology and Neuroendocrine Tumors IEO European Institute of Oncology, IRCCS, Milan, Italy
| | - Nicola Normanno
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori, IRCCS "Fondazione G. Pascale", Naples, Italy
| | - Chantal Pauli
- Department of Pathology and Molecular Pathology, University Hospital Zurich, Zurich, Switzerland
| | - Vivek Subbiah
- Sarah Cannon Research Institute, Nashville, Tennessee, USA
| | - Clare Turnbull
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London, UK
- National Cancer Registration and Analysis Service, National Health Service (NHS) England, London, UK
- Cancer Genetics Unit, The Royal Marsden NHS Foundation Trust, London, UK
| | - C Benedikt Westphalen
- Department of Medicine III, Ludwig Maximilians University (LMU) Hospital Munich, Munich, Germany
- Comprehensive Cancer Center, LMU Hospital Munich, Munich, Germany
- German Cancer Consortium, LMU Hospital Munich, Munich, Germany
| | - Andrew V Biankin
- Wolfson Wohl Cancer Research Center, School of Cancer Sciences, University of Glasgow, Glasgow, UK
- West of Scotland Pancreatic Unit, Glasgow Royal Infirmary, Glasgow, UK
- South Western Sydney Clinical School, Liverpool, New South Wales, Australia
| |
Collapse
|
22
|
Tashkandi H, Younes IE. Advances in Molecular Understanding of Polycythemia Vera, Essential Thrombocythemia, and Primary Myelofibrosis: Towards Precision Medicine. Cancers (Basel) 2024; 16:1679. [PMID: 38730632 PMCID: PMC11083661 DOI: 10.3390/cancers16091679] [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: 04/02/2024] [Revised: 04/22/2024] [Accepted: 04/23/2024] [Indexed: 05/13/2024] Open
Abstract
Myeloproliferative neoplasms (MPNs), including Polycythemia Vera (PV), Essential Thrombocythemia (ET), and Primary Myelofibrosis (PMF), are characterized by the clonal proliferation of hematopoietic stem cells leading to an overproduction of hematopoietic cells. The last two decades have seen significant advances in our understanding of the molecular pathogenesis of these diseases, with the discovery of key mutations in the JAK2, CALR, and MPL genes being pivotal. This review provides a comprehensive update on the molecular landscape of PV, ET, and PMF, highlighting the diagnostic, prognostic, and therapeutic implications of these genetic findings. We delve into the challenges of diagnosing and treating patients with prognostic mutations, clonal evolution, and the impact of emerging technologies like next-generation sequencing and single-cell genomics on the field. The future of MPN management lies in leveraging these molecular insights to develop personalized treatment strategies, aiming for precision medicine that optimizes outcomes for patients. This article synthesizes current knowledge on molecular diagnostics in MPNs, underscoring the critical role of genetic profiling in enhancing patient care and pointing towards future research directions that promise to further refine our approach to these complex disorders.
Collapse
Affiliation(s)
- Hammad Tashkandi
- Department of Pathology and Laboratory Medicine, Moffitt Cancer Center, Tampa, FL 33612, USA
| | - Ismail Elbaz Younes
- Department of Laboratory Medicine and Pathology, Division of Hematopathology, University of Minnesota, Minneapolis, MN 55455, USA;
| |
Collapse
|
23
|
Rofes P, Dueñas N, del Valle J, Navarro M, Balmaña J, Ramón y Cajal T, Tuset N, Castillo C, González S, Brunet J, Capellá G, Lázaro C, Pineda M. Tumor analysis of MMR genes in Lynch-like syndrome: Challenges associated with results interpretation. Cancer Med 2024; 13:e7041. [PMID: 38558366 PMCID: PMC10983805 DOI: 10.1002/cam4.7041] [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: 11/18/2023] [Revised: 01/24/2024] [Accepted: 02/09/2024] [Indexed: 04/04/2024] Open
Abstract
BACKGROUND Up to 70% of suspected Lynch syndrome patients harboring MMR deficient tumors lack identifiable germline pathogenic variants in MMR genes, being referred to as Lynch-like syndrome (LLS). Previous studies have reported biallelic somatic MMR inactivation in a variable range of LLS-associated tumors. Moreover, translating tumor testing results into patient management remains controversial. Our aim is to assess the challenges associated with the implementation of tumoral MMR gene testing in routine workflows. METHODS Here, we present the clinical characterization of 229 LLS patients. MMR gene testing was performed in 39 available tumors, and results were analyzed using two variant allele frequency (VAF) thresholds (≥5% and ≥10%). RESULTS AND DISCUSSION More biallelic somatic events were identified at VAF ≥ 5% than ≥10% (35.9% vs. 25.6%), although the rate of nonconcordant results regarding immunohistochemical pattern increased (30.8% vs. 20.5%). Interpretation difficulties question the current utility of the identification of MMR somatic hits in the diagnostic algorithm of suspected LS cases.
Collapse
Affiliation(s)
- Paula Rofes
- Hereditary Cancer ProgramCatalan Institute of Oncology, Molecular Mechanisms and Experimental Therapy in Oncology Program, Institut d'Investigació Biomèdica de Bellvitge – IDIBELLL'Hospitalet de LlobregatSpain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC)MadridSpain
| | - Núria Dueñas
- Hereditary Cancer ProgramCatalan Institute of Oncology, Molecular Mechanisms and Experimental Therapy in Oncology Program, Institut d'Investigació Biomèdica de Bellvitge – IDIBELLL'Hospitalet de LlobregatSpain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC)MadridSpain
| | - Jesús del Valle
- Hereditary Cancer ProgramCatalan Institute of Oncology, Molecular Mechanisms and Experimental Therapy in Oncology Program, Institut d'Investigació Biomèdica de Bellvitge – IDIBELLL'Hospitalet de LlobregatSpain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC)MadridSpain
| | - Matilde Navarro
- Hereditary Cancer ProgramCatalan Institute of Oncology, Molecular Mechanisms and Experimental Therapy in Oncology Program, Institut d'Investigació Biomèdica de Bellvitge – IDIBELLL'Hospitalet de LlobregatSpain
| | - Judith Balmaña
- Hereditary Cancer Genetics Group, Vall d'Hebron Institute of Oncology (VHIO)Vall d'Hebron HospitalBarcelonaSpain
| | | | - Noemí Tuset
- Medical Oncology DepartmentArnau de Vilanova University HospitalLleidaSpain
| | - Carmen Castillo
- Hereditary Cancer ProgramCatalan Institute of Oncology, Molecular Mechanisms and Experimental Therapy in Oncology Program, Institut d'Investigació Biomèdica de Bellvitge – IDIBELLL'Hospitalet de LlobregatSpain
| | - Sara González
- Hereditary Cancer ProgramCatalan Institute of Oncology, Molecular Mechanisms and Experimental Therapy in Oncology Program, Institut d'Investigació Biomèdica de Bellvitge – IDIBELLL'Hospitalet de LlobregatSpain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC)MadridSpain
| | - Joan Brunet
- Hereditary Cancer ProgramCatalan Institute of Oncology, Molecular Mechanisms and Experimental Therapy in Oncology Program, Institut d'Investigació Biomèdica de Bellvitge – IDIBELLL'Hospitalet de LlobregatSpain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC)MadridSpain
- Hereditary Cancer ProgramCatalan Institute of Oncology – IDIBGiGironaSpain
| | - Gabriel Capellá
- Hereditary Cancer ProgramCatalan Institute of Oncology, Molecular Mechanisms and Experimental Therapy in Oncology Program, Institut d'Investigació Biomèdica de Bellvitge – IDIBELLL'Hospitalet de LlobregatSpain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC)MadridSpain
| | - Conxi Lázaro
- Hereditary Cancer ProgramCatalan Institute of Oncology, Molecular Mechanisms and Experimental Therapy in Oncology Program, Institut d'Investigació Biomèdica de Bellvitge – IDIBELLL'Hospitalet de LlobregatSpain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC)MadridSpain
| | - Marta Pineda
- Hereditary Cancer ProgramCatalan Institute of Oncology, Molecular Mechanisms and Experimental Therapy in Oncology Program, Institut d'Investigació Biomèdica de Bellvitge – IDIBELLL'Hospitalet de LlobregatSpain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC)MadridSpain
| |
Collapse
|
24
|
Carballal S, Balaguer F, Bujanda L, Capellá G, González Santiago S, Jover R, Moreira L, Pineda M, Ruiz-Ponte C, Sánchez Heras AB, Serrano Blanch R, Soto JL, Vidal Tocino R, Cubiella J. Use of multi-gene panels in patients at high risk of hereditary digestive cancer: position statement of AEG, SEOM, AEGH and IMPaCT-GENÓMICA consortium. GASTROENTEROLOGIA Y HEPATOLOGIA 2024; 47:293-318. [PMID: 37315767 DOI: 10.1016/j.gastrohep.2023.06.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 06/04/2023] [Accepted: 06/07/2023] [Indexed: 06/16/2023]
Abstract
This position statement, sponsored by the Asociación Española de Gastroenterología, the Sociedad Española de Oncología Médica, the Asociación Española de Genética Humana and the IMPaCT-Genómica Consortium aims to establish recommendations for use of multi-gene panel testing in patients at high risk of hereditary gastrointestinal and pancreatic cancer. To rate the quality of the evidence and the levels of recommendation, we used the methodology based on the GRADE system (Grading of Recommendations Assessment, Development and Evaluation). We reached a consensus among experts using a Delphi method. The document includes recommendations on clinical scenarios where multi-gene panel testing is recommended in colorectal cancer, polyposis syndromes, gastric and pancreatic cancer, as well as the genes to be considered in each clinical scenario. Recommendations on the evaluation of mosaicisms, counseling strategies in the absence of an index subject and, finally, constitutional analysis after identification of pathogenic tumor variants are also made.
Collapse
Affiliation(s)
- Sabela Carballal
- Servicio de Gastroenterología, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Centro de Investigación Biomédica en Red en Enfermedades Hepáticas y Digestivas (CIBEREHD), Barcelona, España.
| | - Francesc Balaguer
- Servicio de Gastroenterología, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Centro de Investigación Biomédica en Red en Enfermedades Hepáticas y Digestivas (CIBEREHD), Barcelona, España
| | - Luis Bujanda
- Servicio de Aparato Digestivo, Hospital Universitario Donostia, Instituto Biodonostia. Universidad del País Vasco (UPV/EHU), CIBEREHD, San Sebastián, Guipúzcoa, España
| | - Gabriel Capellá
- Programa de Cáncer Hereditario, Instituto Catalán de Oncología, Programa ONCOBELL, IDIBELL, Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), L'Hospitalet de Llobregat, Barcelona, España
| | | | - Rodrigo Jover
- Servicio de Medicina Digestiva, Hospital General Universitario Dr. Balmis, Instituto de Investigación Sanitaria de Alicante (ISABIAL), Departamento de Medicina Clínica, Universidad Miguel Hernández, Alicante, España
| | - Leticia Moreira
- Servicio de Gastroenterología, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Centro de Investigación Biomédica en Red en Enfermedades Hepáticas y Digestivas (CIBEREHD), Barcelona, España
| | - Marta Pineda
- Programa de Cáncer Hereditario, Instituto Catalán de Oncología, Programa ONCOBELL, IDIBELL, Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), L'Hospitalet de Llobregat, Barcelona, España
| | - Clara Ruiz-Ponte
- Fundación Pública Galega de Medicina Xenómica (SERGAS), Instituto de Investigación Sanitaria de Santiago (IDIS), Grupo de Medicina Xenomica (USC), Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERer), Santiago de Compostela, La Coruña, España
| | - Ana Beatriz Sánchez Heras
- Unidad de Consejo Genético en Cáncer, Servicio de Oncología Médica, Hospital General Universitario de Elche, Fundación para el Fomento de la Investigación Sanitaria y Biomédica de la Comunitat Valenciana (FISABIO), Elche, Alicante, España
| | - Raquel Serrano Blanch
- Unidad de Consejo Genético en Cáncer, Unidad de Gestión Clínica de Oncología Médica, H.U. Reina Sofía de Córdoba. Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), CIBERONC, Universidad de Córdoba (UCO), Córdoba, España
| | - José Luis Soto
- Unidad de Genética Molecular, Hospital General Universitario de Elche, FISABIO, Elche, Alicante, España
| | - Rosario Vidal Tocino
- Servicio de Oncología Médica, Complejo Asistencial Universitario de Salamanca, Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, España
| | - Joaquín Cubiella
- Servicio de Aparato Digestivo, Hospital Universitario de Ourense, Grupo de Investigación en Oncología Digestiva-Ourense (GIODO), CIBEREHD, Ourense, España.
| |
Collapse
|
25
|
Kage H, Akiyama N, Chang H, Shinozaki‐Ushiku A, Ka M, Kawata J, Muto M, Okuma Y, Okita N, Tsuchihara K, Kikuchi J, Shirota H, Hayashi H, Kokuryo T, Yachida S, Hirasawa A, Kubo M, Kenmotsu H, Tanabe M, Ushiku T, Muto K, Seto Y, Oda K. Patient survey on cancer genomic medicine in Japan under the national health insurance system. Cancer Sci 2024; 115:954-962. [PMID: 38273803 PMCID: PMC10920978 DOI: 10.1111/cas.16065] [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: 09/06/2023] [Revised: 12/09/2023] [Accepted: 12/17/2023] [Indexed: 01/27/2024] Open
Abstract
In Japan, comprehensive genomic profiling (CGP) tests have been reimbursed under the national health care system for solid cancer patients who have finished standard treatment. More than 50,000 patients have taken the test since June 2019. We performed a nation-wide questionnaire survey between March 2021 and July 2022. Questionnaires were sent to 80 designated Cancer Genomic Medicine Hospitals. Of the 933 responses received, 370 (39.7%) were web based and 563 (60.3%) were paper based. Most patients (784, 84%) first learned about CGP tests from healthcare professionals, and 775 (83.1%) gave informed consent to their treating physician. At the time of informed consent, they were most worried about test results not leading to novel treatment (536, 57.4%). On a scale of 0-10, 702 respondents (75.2%) felt that the explanations of the test result were easy to understand (7 or higher). Ninety-one patients (9.8%) started their recommended treatment. Many patients could not receive recommended treatment because no approved drugs or clinical trials were available (102/177, 57.6%). Ninety-eight patients (10.5%) did not wish their findings to be disclosed. Overall satisfaction with the CGP test process was high, with 602 respondents (64.5%) giving a score of 7-10. The major reason for choosing 0-6 was that the CGP test result did not lead to new treatment (217/277, 78.3%). In conclusion, satisfaction with the CGP test process was high. Patients and family members need better access to information. More patients need to be treated with genomically matched therapy.
Collapse
Affiliation(s)
- Hidenori Kage
- Department of Clinical GenomicsThe University of Tokyo HospitalTokyoJapan
- Next‐Generation Precision Medicine Development Laboratory, Graduate School of MedicineThe University of TokyoTokyoJapan
| | - Nana Akiyama
- Department of Clinical GenomicsThe University of Tokyo HospitalTokyoJapan
| | - Hyangri Chang
- Department of Clinical GenomicsThe University of Tokyo HospitalTokyoJapan
| | - Aya Shinozaki‐Ushiku
- Department of Clinical GenomicsThe University of Tokyo HospitalTokyoJapan
- Division of Integrative Genomics, Graduate School of MedicineThe University of TokyoTokyoJapan
- Department of Pathology, Graduate School of MedicineThe University of TokyoTokyoJapan
| | - Mirei Ka
- Division of Integrative Genomics, Graduate School of MedicineThe University of TokyoTokyoJapan
| | - Junichi Kawata
- Department of Public PolicyThe Institute of Medical Science, The University of TokyoTokyoJapan
| | - Manabu Muto
- Department of Therapeutic OncologyKyoto University Graduate School of MedicineKyotoJapan
| | - Yusuke Okuma
- Center for Cancer Genomics and Advanced TherapeuticsNational Cancer CenterTokyoJapan
| | - Natsuko Okita
- Center for Cancer Genomics and Advanced TherapeuticsNational Cancer CenterTokyoJapan
| | - Katsuya Tsuchihara
- Department of Genetic Medicine and ServicesNational Cancer Center Hospital EastChibaJapan
| | - Junko Kikuchi
- Division of Clinical Cancer GenomicsHokkaido University HospitalSapporoJapan
| | - Hidekazu Shirota
- Department of Clinical OncologyTohoku University HospitalSendaiJapan
| | - Hideyuki Hayashi
- Genomics Unit, Keio Cancer CenterKeio University School of MedicineTokyoJapan
| | - Toshio Kokuryo
- Division of Surgical Oncology, Department of SurgeryNagoya University Graduate School of MedicineNagoyaJapan
| | - Shinichi Yachida
- Department of Cancer Genome Informatics, Graduate School of MedicineOsaka UniversityOsakaJapan
| | - Akira Hirasawa
- Department of Clinical Genomic MedicineOkayama University Graduate School of Medicine, Dentistry and Pharmaceutical SciencesOkayamaJapan
| | - Makoto Kubo
- Department of Breast Surgical OncologyKyushu University HospitalFukuokaJapan
| | | | - Masahiko Tanabe
- Department of Breast and Endocrine Surgery, Graduate School of MedicineThe University of TokyoTokyoJapan
| | - Tetsuo Ushiku
- Department of Pathology, Graduate School of MedicineThe University of TokyoTokyoJapan
| | - Kaori Muto
- Department of Public PolicyThe Institute of Medical Science, The University of TokyoTokyoJapan
| | - Yasuyuki Seto
- Department of Gastrointestinal Surgery, Graduate School of MedicineThe University of TokyoTokyoJapan
| | - Katsutoshi Oda
- Department of Clinical GenomicsThe University of Tokyo HospitalTokyoJapan
- Division of Integrative Genomics, Graduate School of MedicineThe University of TokyoTokyoJapan
| |
Collapse
|
26
|
Nowlen CJ, Daniels M, Uzunparmak B, Ileana Dumbrava EE, Yuan Y, Patel KP, Rayes N, Harkenrider J, Wathoo C, Veazie J, Luna KA, Wang W, Horombe C, Javle M, Ahnert JR, Yap TA, Arun B, Lu KH, Meric-Bernstam F. Limited Independent Follow-Up with Germline Testing of Variants Detected in BRCA1 and BRCA2 by Tumor-Only Sequencing. JOURNAL OF IMMUNOTHERAPY AND PRECISION ONCOLOGY 2024; 7:7-17. [PMID: 38327755 PMCID: PMC10846638 DOI: 10.36401/jipo-23-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 08/28/2023] [Accepted: 10/10/2023] [Indexed: 02/09/2024]
Abstract
Introduction Genomic profiling is performed in patients with advanced or metastatic cancer, in order to direct cancer treatment, often sequencing tumor-only, without a matched germline comparator. However, because many of the genes analyzed on tumor profiling overlap with those known to be associated with hereditary cancer predisposition syndromes (HCPS), tumor-only profiling can unknowingly uncover germline pathogenic (P) and likely pathogenic variants (LPV). In this study, we evaluated the number of patients with P/LPVs identified in BRCA1 and BRCA2 (BRCA1/2) via tumor-only profiling, then determined the germline testing outcomes for those patients. Methods A retrospective chart review was performed to identify patients with BRCA1/2 variants on tumor-only genomic profiling, and whether they had germline testing. Results This study found that of 2923 patients with 36 tumor types who underwent tumor-only testing, 554 had a variant in BRCA1/2 (19.0%); 119 of the 554 patients (21.5%) had a P/LP BRCA1/2 variant, representing 4.1% of the overall population who underwent genomic profiling. Seventy-three (61.3%) of 119 patients with BRCA1/2 P/LPV on tumor-only testing did not undergo germline testing, 34 (28.6%) had already had germline testing before tumor-only testing, and 12 (10.1%) underwent germline testing after tumor-only testing. Twenty-eight germline BRCA1/2 P/LPVs were detected, 24 in those who had prior germline testing, and 4 among the 12 patients who had germline testing after tumor-only testing. Conclusion Tumor-only testing is likely to identify P/LPVs in BRCA1/2. Efforts to improve follow-up germline testing is needed to improve identification of germline BRCA1/2 alterations.
Collapse
Affiliation(s)
- Carol J. Nowlen
- The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Molly Daniels
- Department of Clinical Cancer Genetics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Burak Uzunparmak
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ecaterina E. Ileana Dumbrava
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ying Yuan
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Keyur P. Patel
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Nadine Rayes
- Department of Clinical Cancer Genetics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jacqueline Harkenrider
- Department of Obstetrics, Gynecology, and Reproductive Sciences, The University of Texas Health Science Center at Houston John P. and Katherine G. McGovern Medical School, Houston, TX, USA
| | - Chetna Wathoo
- Institute for Personalized Cancer Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jennifer Veazie
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Krystle A. Luna
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Wanlin Wang
- Institute for Personalized Cancer Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Chacha Horombe
- Department of Enterprise Development & Integration, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Milind Javle
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jordi Rodon Ahnert
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Timothy A. Yap
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Banu Arun
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Karen H. Lu
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Funda Meric-Bernstam
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Institute for Personalized Cancer Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
27
|
Koster R, Schipper LJ, Giesbertz NAA, van Beek D, Mendeville M, Samsom KG, Rosenberg EH, Hogervorst FBL, Roepman P, Boelens MC, Bosch LJW, van den Berg JG, Meijer GA, Voest EE, Cuppen E, Ruijs MWG, van Wezel T, van der Kolk L, Monkhorst K. Impact of genetic counseling strategy on diagnostic yield and workload for genome-sequencing-based tumor diagnostics. Genet Med 2024; 26:101032. [PMID: 38006283 DOI: 10.1016/j.gim.2023.101032] [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: 07/17/2023] [Revised: 11/17/2023] [Accepted: 11/19/2023] [Indexed: 11/26/2023] Open
Abstract
PURPOSE Genome sequencing (GS) enables comprehensive molecular analysis of tumors and identification of hereditary cancer predisposition. According to guidelines, directly determining pathogenic germline variants (PGVs) requires pretest genetic counseling, which is cost-ineffective. Referral for genetic counseling based on tumor variants alone could miss relevant PGVs and/or result in unnecessary referrals. METHODS We validated GS for detection of germline variants and simulated 3 strategies using paired tumor-normal GS data of 937 metastatic patients. In strategy-1, genetic counseling before tumor testing allowed direct PGV analysis. In strategy-2 and -3, germline testing and referral for post-test genetic counseling is based on tumor variants using Dutch (strategy-2) or Europen Society for Medical Oncology (ESMO) Precision Medicine Working Group (strategy-3) guidelines. RESULTS In strategy-1, PGVs would be detected in 50 patients (number-needed-to counsel; NTC = 18.7). In strategy-2, 86 patients would have been referred for genetic counseling and 43 would have PGVs (NTC = 2). In strategy-3, 94 patients would have been referred for genetic counseling and 32 would have PGVs (NTC = 2.9). Hence, 43 and 62 patients, respectively, were unnecessarily referred based on a somatic variant. CONCLUSION Both post-tumor test counseling strategies (2 and 3) had significantly lower NTC, and strategy-2 had the highest PGV yield. Combining pre-tumor test mainstreaming and post-tumor test counseling may maximize the clinically relevant PGV yield and minimize unnecessary referrals.
Collapse
Affiliation(s)
- Roelof Koster
- The Netherlands Cancer Institute, Amsterdam, The Netherlands.
| | - Luuk J Schipper
- The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | | | | | | | - Kris G Samsom
- The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | | | | | - Paul Roepman
- Hartwig Medical Foundation, Amsterdam, The Netherlands
| | | | - Linda J W Bosch
- The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | | | - Gerrit A Meijer
- The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Emile E Voest
- The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Edwin Cuppen
- Hartwig Medical Foundation, Amsterdam, The Netherlands
| | | | - Tom van Wezel
- The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | | | - Kim Monkhorst
- The Netherlands Cancer Institute, Amsterdam, The Netherlands.
| |
Collapse
|
28
|
Hayashi H, Kunimasa K, Tanishima S, Nakamura K, Ishikawa M, Kato Y, Aimono E, Kawano R, Nishihara H. Germline BRCA2 variant with low variant allele frequency detected in tumor-only comprehensive genomic profiling. Cancer Sci 2024; 115:682-686. [PMID: 38086530 PMCID: PMC10859595 DOI: 10.1111/cas.16043] [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: 09/29/2023] [Revised: 11/20/2023] [Accepted: 11/28/2023] [Indexed: 02/13/2024] Open
Abstract
Germline BRCA1/2 variants in comprehensive genomic profiling (CGP) often exhibit variant allele frequency (VAF) exceeding 50%. However, when genomic loss occurs at the ipsilateral allele, including the germline variant in tumor cells, the VAF is low. This case report presents a patient with uterine sarcoma with a pathogenic BRCA2 mutation and low VAF in tumor-only CGP, which was later identified as a germline variant. When genomic alterations in BRCA1/2 are identified in tumor-only CGP, the possible germline origin of the variants should be considered, even if their VAF is very low.
Collapse
Affiliation(s)
- Hideyuki Hayashi
- Genomics Unit, Keio Cancer CenterKeio University School of MedicineTokyoJapan
| | - Kei Kunimasa
- Department of Thoracic OncologyOsaka International Cancer InstituteOsakaJapan
| | - Shigeki Tanishima
- Department of Biomedical Informatics DevelopmentMitsubishi Electric Software Co., LtdTokyoJapan
| | - Kohei Nakamura
- Genomics Unit, Keio Cancer CenterKeio University School of MedicineTokyoJapan
| | - Marin Ishikawa
- Genomics Unit, Keio Cancer CenterKeio University School of MedicineTokyoJapan
| | - Yasutaka Kato
- Genomics Unit, Keio Cancer CenterKeio University School of MedicineTokyoJapan
| | - Eriko Aimono
- Genomics Unit, Keio Cancer CenterKeio University School of MedicineTokyoJapan
| | - Ryutaro Kawano
- Genomics Unit, Keio Cancer CenterKeio University School of MedicineTokyoJapan
| | - Hiroshi Nishihara
- Genomics Unit, Keio Cancer CenterKeio University School of MedicineTokyoJapan
| |
Collapse
|
29
|
Bourlon MT, Valdez P, Castro E. Development of PARP inhibitors in advanced prostate cancer. Ther Adv Med Oncol 2024; 16:17588359231221337. [PMID: 38205078 PMCID: PMC10777773 DOI: 10.1177/17588359231221337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 11/29/2023] [Indexed: 01/12/2024] Open
Abstract
The relatively high prevalence of alterations in the homologous recombination repair (HRR) pathway described in advanced prostate cancer provides a unique opportunity to develop therapeutic strategies that take advantage of the decreased tumor ability to repair DNA damage. Poly ADP-ribose polymerase (PARP) inhibitors have been demonstrated to improve the outcomes of metastatic castration-resistant prostate cancer (mCRPC) patients with HRR defects, particularly in those with BRCA1/2 alterations. To expand the benefit of PARPi to patients without detectable HRR alterations, multiple studies are addressing potential synergies between PARP inhibition (PARPi) and androgen receptor pathway inhibitors (ARSi), radiation, radioligand therapy, chemotherapy, or immunotherapy, and these strategies are also being evaluated in the hormone-sensitive setting. In this review, we summarize the development of PARPi in prostate cancer, the potential synergies, and combinations being investigated as well as the future directions of PARPi for the management of the disease.
Collapse
Affiliation(s)
- Maria Teresa Bourlon
- Hemato-Oncology Department, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Paola Valdez
- Hemato-Oncology Department, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Elena Castro
- Department of Medical Oncology, Hospital Universitario 12 de Octubre, Av. Cordoba s/n, 28041, Madrid, Spain
| |
Collapse
|
30
|
Kleiblová P, Černá M, Zemánková P, Matějková K, Nehasil P, Hojný J, Horáčková K, Janatová M, Soukupová J, Šťastná B, Kleibl Z. Parallel DNA/RNA NGS Using an Identical Target Enrichment Panel in the Analysis of Hereditary Cancer Predisposition. Folia Biol (Praha) 2024; 70:62-73. [PMID: 38830124 DOI: 10.14712/fb2024070010062] [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: 06/05/2024]
Abstract
Germline DNA testing using the next-gene-ration sequencing (NGS) technology has become the analytical standard for the diagnostics of hereditary diseases, including cancer. Its increasing use places high demands on correct sample identification, independent confirmation of prioritized variants, and their functional and clinical interpretation. To streamline these processes, we introduced parallel DNA and RNA capture-based NGS using identical capture panel CZECANCA, which is routinely used for DNA analysis of hereditary cancer predisposition. Here, we present the analytical workflow for RNA sample processing and its analytical and diagnostic performance. Parallel DNA/RNA analysis allowed credible sample identification by calculating the kinship coefficient. The RNA capture-based approach enriched transcriptional targets for the majority of clinically relevant cancer predisposition genes to a degree that allowed analysis of the effect of identified DNA variants on mRNA processing. By comparing the panel and whole-exome RNA enrichment, we demonstrated that the tissue-specific gene expression pattern is independent of the capture panel. Moreover, technical replicates confirmed high reproducibility of the tested RNA analysis. We concluded that parallel DNA/RNA NGS using the identical gene panel is a robust and cost-effective diagnostic strategy. In our setting, it allows routine analysis of 48 DNA/RNA pairs using NextSeq 500/550 Mid Output Kit v2.5 (150 cycles) in a single run with sufficient coverage to analyse 226 cancer predisposition and candidate ge-nes. This approach can replace laborious Sanger confirmatory sequencing, increase testing turnaround, reduce analysis costs, and improve interpretation of the impact of variants by analysing their effect on mRNA processing.
Collapse
Affiliation(s)
- Petra Kleiblová
- Institute of Biology and Medical Genetics, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic.
- Institute of Medical Biochemistry and Laboratory Diagnostics, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic.
| | - Marta Černá
- Institute of Medical Biochemistry and Laboratory Diagnostics, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic
| | - Petra Zemánková
- Institute of Medical Biochemistry and Laboratory Diagnostics, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic
- Institute of Pathological Physiology, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Kateřina Matějková
- Department of Genetics and Microbiology, Faculty of Science, Charles University, Prague, Czech Republic
- Institute of Medical Biochemistry and Laboratory Diagnostics, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic
| | - Petr Nehasil
- Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic
- Institute of Medical Biochemistry and Laboratory Diagnostics, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic
- Institute of Pathological Physiology, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Jan Hojný
- Institute of Pathology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic
| | - Klára Horáčková
- Institute of Medical Biochemistry and Laboratory Diagnostics, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic
| | - Markéta Janatová
- Institute of Medical Biochemistry and Laboratory Diagnostics, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic
| | - Jana Soukupová
- Institute of Medical Biochemistry and Laboratory Diagnostics, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic
| | - Barbora Šťastná
- Department of Biochemistry, Faculty of Science, Charles University, Prague, Czech Republic
- Institute of Medical Biochemistry and Laboratory Diagnostics, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic
| | - Zdeněk Kleibl
- Institute of Medical Biochemistry and Laboratory Diagnostics, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic
| |
Collapse
|
31
|
Calabrese M, Saporita I, Turco F, Gillessen S, Castro E, Vogl UM, Di Stefano RF, Carfì FM, Poletto S, Farinea G, Tucci M, Buttigliero C. Synthetic Lethality by Co-Inhibition of Androgen Receptor and Polyadenosine Diphosphate-Ribose in Metastatic Prostate Cancer. Int J Mol Sci 2023; 25:78. [PMID: 38203248 PMCID: PMC10779404 DOI: 10.3390/ijms25010078] [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: 10/18/2023] [Revised: 12/17/2023] [Accepted: 12/18/2023] [Indexed: 01/12/2024] Open
Abstract
Androgen receptor pathway inhibitors (ARPI) and polyadenosine diphosphate-ribose inhibitors (PARPi) are part of the standard of care in patients with metastatic castration-resistant prostate cancer (mCRPC). There is biological evidence that the association of ARPI and PARPi could have a synergistic effect; therefore, several ongoing clinical trials are investigating the efficacy of this combination with preliminary results that are not perfectly concordant in identifying patients who can obtain the most benefit from this therapeutic option. The purpose of this review is to describe the PARPi mechanisms of action and to analyze the biological mechanisms behind the interplay between the androgen receptor and the PARPi system to better understand the rationale of the ARPI + PARPi combinations. Furthermore, we will summarize the preliminary results of the ongoing studies on these combinations, trying to understand in which patients to apply. Finally, we will discuss the clinical implications of this combination and its possible future perspectives.
Collapse
Affiliation(s)
- Mariangela Calabrese
- Department of Oncology, University of Turin, AOU San Luigi Gonzaga, 10043 Orbassano, Italy; (M.C.); (I.S.); (F.T.); (R.F.D.S.); (F.M.C.); (S.P.); (G.F.)
| | - Isabella Saporita
- Department of Oncology, University of Turin, AOU San Luigi Gonzaga, 10043 Orbassano, Italy; (M.C.); (I.S.); (F.T.); (R.F.D.S.); (F.M.C.); (S.P.); (G.F.)
| | - Fabio Turco
- Department of Oncology, University of Turin, AOU San Luigi Gonzaga, 10043 Orbassano, Italy; (M.C.); (I.S.); (F.T.); (R.F.D.S.); (F.M.C.); (S.P.); (G.F.)
- Ente Ospedaliero Cantonale—Istituto Oncologico della Svizzera Italiana, 6500 Bellinzona, Switzerland; (S.G.); (U.M.V.)
| | - Silke Gillessen
- Ente Ospedaliero Cantonale—Istituto Oncologico della Svizzera Italiana, 6500 Bellinzona, Switzerland; (S.G.); (U.M.V.)
- Department of Medical Oncology, Faculty of Biomedical Sciences, Università della Svizzera Italiana, 6900 Lugano, Switzerland
| | - Elena Castro
- Hospital Universitario 12 de Octubre, 28041 Madrid, Spain;
| | - Ursula Maria Vogl
- Ente Ospedaliero Cantonale—Istituto Oncologico della Svizzera Italiana, 6500 Bellinzona, Switzerland; (S.G.); (U.M.V.)
| | - Rosario Francesco Di Stefano
- Department of Oncology, University of Turin, AOU San Luigi Gonzaga, 10043 Orbassano, Italy; (M.C.); (I.S.); (F.T.); (R.F.D.S.); (F.M.C.); (S.P.); (G.F.)
| | - Federica Maria Carfì
- Department of Oncology, University of Turin, AOU San Luigi Gonzaga, 10043 Orbassano, Italy; (M.C.); (I.S.); (F.T.); (R.F.D.S.); (F.M.C.); (S.P.); (G.F.)
| | - Stefano Poletto
- Department of Oncology, University of Turin, AOU San Luigi Gonzaga, 10043 Orbassano, Italy; (M.C.); (I.S.); (F.T.); (R.F.D.S.); (F.M.C.); (S.P.); (G.F.)
| | - Giovanni Farinea
- Department of Oncology, University of Turin, AOU San Luigi Gonzaga, 10043 Orbassano, Italy; (M.C.); (I.S.); (F.T.); (R.F.D.S.); (F.M.C.); (S.P.); (G.F.)
| | - Marcello Tucci
- Department of Medical Oncology, Cardinal Massaia Hospital, 14100 Asti, Italy;
| | - Consuelo Buttigliero
- Department of Oncology, University of Turin, AOU San Luigi Gonzaga, 10043 Orbassano, Italy; (M.C.); (I.S.); (F.T.); (R.F.D.S.); (F.M.C.); (S.P.); (G.F.)
| |
Collapse
|
32
|
Mosteiro M, Azuara D, Villatoro S, Alay A, Gausachs M, Varela M, Baixeras N, Pijuan L, Ajenjo-Bauza M, Lopez-Doriga A, Teulé Á, Solanes A, Palmero R, Brenes J, Jové M, Padrones S, Moreno V, Cordero D, Matías-Guiu X, Lázaro C, Nadal E. Molecular profiling and feasibility using a comprehensive hybrid capture panel on a consecutive series of non-small-cell lung cancer patients from a single centre. ESMO Open 2023; 8:102197. [PMID: 38070435 PMCID: PMC10774954 DOI: 10.1016/j.esmoop.2023.102197] [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: 07/03/2023] [Revised: 11/04/2023] [Accepted: 11/13/2023] [Indexed: 12/31/2023] Open
Abstract
BACKGROUND Targeted next-generation sequencing (NGS) is recommended to screen actionable genomic alterations (GAs) in patients with non-small-cell lung cancer (NSCLC). We determined the feasibility to detect actionable GAs using TruSight™ Oncology 500 (TSO500) in 200 consecutive patients with NSCLC. MATERIALS AND METHODS DNA and RNA were sequenced on an Illumina® NextSeq 550 instrument and processed using the TSO500 Docker pipeline. Clinical actionability was defined within the molecular tumour board following European Society for Medical Oncology (ESMO) guidelines for oncogene-addicted NSCLC. Overall survival (OS) was estimated as per the presence of druggable GAs and treatment with targeted therapy. RESULTS Most patients were males (69.5%) and former or current smokers (86.5%). Median age was 64 years. The most common histological type and tumour stage were lung adenocarcinoma (81%) and stage IV (64%), respectively. Sequencing was feasible in most patients (93.5%) and actionable GAs were found in 26.5% of patients. A high concordance was observed between single-gene testing and TSO500 NGS panel. Patients harbouring druggable GAs and receiving targeted therapy achieved longer OS compared to patients without druggable GAs. Conversely, patients with druggable GAs not receiving targeted therapy had a trend toward shorter OS compared with driver-negative patients. CONCLUSIONS Hybrid capture sequencing using TSO500 panel is feasible to analyse clinical samples from patients with NSCLC and is an efficient tool for screening actionable GAs.
Collapse
Affiliation(s)
- M Mosteiro
- Department of Medical Oncology, Catalan Institute of Oncology (ICO), L'Hospitalet del Llobregat, Barcelona, Spain; Preclinical and Experimental Research in Thoracic Tumors (PReTT), Molecular Mechanisms and Experimental Therapy in Oncology Program (Oncobell), Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain
| | - D Azuara
- Laboratory Core Molecular Analysis (L-CAM), Hospital Universitari de Bellvitge and Catalan Institute of Oncology, L'Hospitalet del Llobregat, Barcelona, Spain; Hereditary Cancer Program, Molecular Mechanisms and Experimental Therapy in Oncology (Oncobell) Program, Catalan Institute of Oncology (ICO), Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet del Llobregat, Barcelona, Spain
| | - S Villatoro
- Laboratory Core Molecular Analysis (L-CAM), Hospital Universitari de Bellvitge and Catalan Institute of Oncology, L'Hospitalet del Llobregat, Barcelona, Spain; Department of Pathology, Hospital Universitari de Bellvitge and Catalan Institute of Oncology (ICO), L'Hospitalet del Llobregat, Barcelona, Spain
| | - A Alay
- Preclinical and Experimental Research in Thoracic Tumors (PReTT), Molecular Mechanisms and Experimental Therapy in Oncology Program (Oncobell), Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain; Unit of Bioinformatics for Precision Oncology (UBOP), Catalan Institute of Oncology (ICO), L'Hospitalet de Llobregat, Barcelona, Spain
| | - M Gausachs
- Department of Medical Oncology, Catalan Institute of Oncology (ICO), L'Hospitalet del Llobregat, Barcelona, Spain; Preclinical and Experimental Research in Thoracic Tumors (PReTT), Molecular Mechanisms and Experimental Therapy in Oncology Program (Oncobell), Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain
| | - M Varela
- Laboratory Core Molecular Analysis (L-CAM), Hospital Universitari de Bellvitge and Catalan Institute of Oncology, L'Hospitalet del Llobregat, Barcelona, Spain; Department of Pathology, Hospital Universitari de Bellvitge and Catalan Institute of Oncology (ICO), L'Hospitalet del Llobregat, Barcelona, Spain
| | - N Baixeras
- Department of Pathology, Hospital Universitari de Bellvitge and Catalan Institute of Oncology (ICO), L'Hospitalet del Llobregat, Barcelona, Spain
| | - L Pijuan
- Department of Pathology, Hospital Universitari de Bellvitge and Catalan Institute of Oncology (ICO), L'Hospitalet del Llobregat, Barcelona, Spain
| | - M Ajenjo-Bauza
- Unit of Bioinformatics for Precision Oncology (UBOP), Catalan Institute of Oncology (ICO), L'Hospitalet de Llobregat, Barcelona, Spain
| | - A Lopez-Doriga
- Preclinical and Experimental Research in Thoracic Tumors (PReTT), Molecular Mechanisms and Experimental Therapy in Oncology Program (Oncobell), Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain; Unit of Bioinformatics for Precision Oncology (UBOP), Catalan Institute of Oncology (ICO), L'Hospitalet de Llobregat, Barcelona, Spain; Centro de Investigación Biomédica en Red de Epidemiologia y Salud Pública (CIBERESP), Madrid, Spain
| | - Á Teulé
- Department of Medical Oncology, Catalan Institute of Oncology (ICO), L'Hospitalet del Llobregat, Barcelona, Spain; Hereditary Cancer Program, Molecular Mechanisms and Experimental Therapy in Oncology (Oncobell) Program, Catalan Institute of Oncology (ICO), Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet del Llobregat, Barcelona, Spain
| | - A Solanes
- Department of Medical Oncology, Catalan Institute of Oncology (ICO), L'Hospitalet del Llobregat, Barcelona, Spain; Hereditary Cancer Program, Molecular Mechanisms and Experimental Therapy in Oncology (Oncobell) Program, Catalan Institute of Oncology (ICO), Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet del Llobregat, Barcelona, Spain
| | - R Palmero
- Department of Medical Oncology, Catalan Institute of Oncology (ICO), L'Hospitalet del Llobregat, Barcelona, Spain; Preclinical and Experimental Research in Thoracic Tumors (PReTT), Molecular Mechanisms and Experimental Therapy in Oncology Program (Oncobell), Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain
| | - J Brenes
- Department of Medical Oncology, Catalan Institute of Oncology (ICO), L'Hospitalet del Llobregat, Barcelona, Spain; Preclinical and Experimental Research in Thoracic Tumors (PReTT), Molecular Mechanisms and Experimental Therapy in Oncology Program (Oncobell), Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain
| | - M Jové
- Department of Medical Oncology, Catalan Institute of Oncology (ICO), L'Hospitalet del Llobregat, Barcelona, Spain; Preclinical and Experimental Research in Thoracic Tumors (PReTT), Molecular Mechanisms and Experimental Therapy in Oncology Program (Oncobell), Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain
| | - S Padrones
- Department of Respiratory Medicine, Hospital Universitari de Bellvitge, L'Hospitalet del Llobregat, Barcelona, Spain
| | - V Moreno
- Centro de Investigación Biomédica en Red de Epidemiologia y Salud Pública (CIBERESP), Madrid, Spain; Oncology Data Analytics Program, Catalan Institute of Oncology (ICO), Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain; Department of Clinical Sciences, Faculty of Medicine, and Universitat de Barcelona Institute of Complex Systems (UBICS), University of Barcelona, Barcelona, Spain
| | - D Cordero
- Preclinical and Experimental Research in Thoracic Tumors (PReTT), Molecular Mechanisms and Experimental Therapy in Oncology Program (Oncobell), Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain; Department of Pathology, Hospital Universitari de Bellvitge and Catalan Institute of Oncology (ICO), L'Hospitalet del Llobregat, Barcelona, Spain; Unit of Bioinformatics for Precision Oncology (UBOP), Catalan Institute of Oncology (ICO), L'Hospitalet de Llobregat, Barcelona, Spain; Centro de Investigación Biomédica en Red de Epidemiologia y Salud Pública (CIBERESP), Madrid, Spain.
| | - X Matías-Guiu
- Laboratory Core Molecular Analysis (L-CAM), Hospital Universitari de Bellvitge and Catalan Institute of Oncology, L'Hospitalet del Llobregat, Barcelona, Spain; Department of Pathology, Hospital Universitari de Bellvitge and Catalan Institute of Oncology (ICO), L'Hospitalet del Llobregat, Barcelona, Spain.
| | - C Lázaro
- Laboratory Core Molecular Analysis (L-CAM), Hospital Universitari de Bellvitge and Catalan Institute of Oncology, L'Hospitalet del Llobregat, Barcelona, Spain; Hereditary Cancer Program, Molecular Mechanisms and Experimental Therapy in Oncology (Oncobell) Program, Catalan Institute of Oncology (ICO), Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet del Llobregat, Barcelona, Spain; Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain.
| | - E Nadal
- Department of Medical Oncology, Catalan Institute of Oncology (ICO), L'Hospitalet del Llobregat, Barcelona, Spain; Preclinical and Experimental Research in Thoracic Tumors (PReTT), Molecular Mechanisms and Experimental Therapy in Oncology Program (Oncobell), Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain.
| |
Collapse
|
33
|
Benusiglio PR, Dardenne A, Fallet V, Cadranel J. Emerging cancer risks in BRCA2 pathogenic germline variant carriers. Eur J Hum Genet 2023; 31:1355-1356. [PMID: 37758835 PMCID: PMC10689811 DOI: 10.1038/s41431-023-01465-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 09/13/2023] [Indexed: 09/29/2023] Open
Affiliation(s)
- Patrick R Benusiglio
- Sorbonne Université, UF d'Oncogénétique Clinique, Département de Génétique Médicale, Hôpital Pitié-Salpêtrière, AP-HP, 47-83 Boulevard de l'Hôpital, F-75013, Paris, France.
- Sorbonne Université, INSERM, Unité Mixte de Recherche Scientifique 938 et SIRIC CURAMUS, Centre de Recherche Saint-Antoine (CRSA), Equipe Instabilité des Microsatellites et Cancer, F-75012, Paris, France.
| | - Antoine Dardenne
- Sorbonne Université, Service de Chirurgie Digestive, Hôpital Saint-Antoine, APHP, F-75012, Paris, France
| | - Vincent Fallet
- Sorbonne Université, GRC-04 Theranoscan et Service de Pneumologie et Oncologie Thoracique, Hôpital Tenon, APHP, F-75020, Paris, France
| | - Jacques Cadranel
- Sorbonne Université, GRC-04 Theranoscan et Service de Pneumologie et Oncologie Thoracique, Hôpital Tenon, APHP, F-75020, Paris, France
| |
Collapse
|
34
|
Lalloo F, Kulkarni A, Chau C, Nielsen M, Sheaff M, Steele J, van Doorn R, Wadt K, Hamill M, Torr B, Tischkowitz M, Hanson H. Clinical practice guidelines for the diagnosis and surveillance of BAP1 tumour predisposition syndrome. Eur J Hum Genet 2023; 31:1261-1269. [PMID: 37607989 PMCID: PMC10620132 DOI: 10.1038/s41431-023-01448-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 06/22/2023] [Accepted: 08/01/2023] [Indexed: 08/24/2023] Open
Abstract
BRCA1-associated protein-1 (BAP1) is a recognised tumour suppressor gene. Germline BAP1 pathogenic/likely pathogenic variants are associated with predisposition to multiple tumours, including uveal melanoma, malignant pleural and peritoneal mesothelioma, renal cell carcinoma and specific non-malignant neoplasms of the skin, as part of the autosomal dominant BAP1-tumour predisposition syndrome. The overall lifetime risk for BAP1 carriers to develop at least one BAP1-associated tumour is up to 85%, although due to ascertainment bias, current estimates of risk are likely to be overestimated. As for many rare cancer predisposition syndromes, there is limited scientific evidence to support the utility of surveillance and, therefore, management recommendations for BAP1 carriers are based on expert opinion. To date, European recommendations for BAP1 carriers have not been published but are necessary due to the emerging phenotype of this recently described syndrome and increased identification of BAP1 carriers via large gene panels or tumour sequencing. To address this, the Clinical Guideline Working Group of the CanGene-CanVar project in the United Kingdom invited European collaborators to collaborate to develop guidelines to harmonize surveillance programmes within Europe. Recommendations with respect to BAP1 testing and surveillance were achieved following literature review and Delphi survey completed by a core group and an extended expert group of 34 European specialists including Geneticists, Ophthalmologists, Oncologists, Dermatologists and Pathologists. It is recognised that these largely evidence-based but pragmatic recommendations will evolve over time as further data from research collaborations informs the phenotypic spectrum and surveillance outcomes.
Collapse
Affiliation(s)
- Fiona Lalloo
- Manchester Centre for Genomic Medicine, Manchester University NHS Foundation Trust, Manchester, UK
| | - Anju Kulkarni
- Department of Clinical Genetics, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Cindy Chau
- Department of Ophthalmology, Leiden University Medical Center, Leiden, the Netherlands
| | - Maartje Nielsen
- Department of Clinical Genetics, Leiden University Medical Center, Leiden, the Netherlands
| | - Michael Sheaff
- Department of Cellular Pathology, Barts Health NHS Trust, London, UK
| | - Jeremy Steele
- Department of Oncology, Barts Health NHS Trust, London, UK
| | - Remco van Doorn
- Department of Dermatology, Leiden University Medical Center, Leiden, the Netherlands
| | - Karin Wadt
- Department of Clinical Genetics, Copenhagen University Hospital, Copenhagen, Denmark
| | - Monica Hamill
- Division of Genetics and Epidemiology, Institute of Cancer Research, Sutton, London, UK
| | - Beth Torr
- Division of Genetics and Epidemiology, Institute of Cancer Research, Sutton, London, UK
| | - Marc Tischkowitz
- Department of Medical Genetics, National Institute for Health Research Cambridge Biomedical Research Centre, University of Cambridge, Cambridge, UK
| | - Helen Hanson
- Division of Genetics and Epidemiology, Institute of Cancer Research, Sutton, London, UK.
- South West Thames Regional Genetics Service, St George's University Hospitals NHS Foundation Trust, London, UK.
| |
Collapse
|
35
|
Stout LA, Hunter C, Schroeder C, Kassem N, Schneider BP. Clinically significant germline pathogenic variants are missed by tumor genomic sequencing. NPJ Genom Med 2023; 8:30. [PMID: 37833309 PMCID: PMC10575977 DOI: 10.1038/s41525-023-00374-9] [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: 05/24/2023] [Accepted: 09/29/2023] [Indexed: 10/15/2023] Open
Abstract
A germline pathogenic variant may be present even if the results of tumor genomic sequencing do not suggest one. There are key differences in the assay design and reporting of variants between germline and somatic laboratories. When appropriate, both tests should be completed to aid in therapy decisions and determining optimal screening and risk-reduction interventions.
Collapse
Affiliation(s)
- Leigh Anne Stout
- Indiana University School of Medicine, Indianapolis, IN, USA
- Indiana University Health Precision Genomics, Indianapolis, IN, USA
| | - Cynthia Hunter
- Indiana University Health Precision Genomics, Indianapolis, IN, USA
| | | | - Nawal Kassem
- Indiana University School of Medicine, Indianapolis, IN, USA
- Indiana University Health Precision Genomics, Indianapolis, IN, USA
| | - Bryan P Schneider
- Indiana University School of Medicine, Indianapolis, IN, USA.
- Indiana University Health Precision Genomics, Indianapolis, IN, USA.
| |
Collapse
|
36
|
Farinea G, Crespi V, Listì A, Righi L, Bironzo P, Merlini A, Malapelle U, Novello S, Scagliotti GV, Passiglia F. The Role of Germline Mutations in Thoracic Malignancies: Between Myth and Reality. J Thorac Oncol 2023; 18:1146-1164. [PMID: 37331604 DOI: 10.1016/j.jtho.2023.05.028] [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: 03/15/2023] [Revised: 05/15/2023] [Accepted: 05/23/2023] [Indexed: 06/20/2023]
Abstract
Considering the established contribution of environmental factors to the development of thoracic malignancies, the inherited susceptibility of these tumors has rarely been explored. However, the recent introduction of next-generation sequencing-based tumor molecular profiling in the real-word setting enabled us to deeply characterize the genomic background of patients with lung cancer with or without smoking-related history, increasing the likelihood of detecting germline mutations with potential prevention and treatment implications. Pathogenic germline variants have been detected in 2% to 3% of patients with NSCLC undergoing next-generation sequencing analysis, whereas the proportion of germline mutations associated with the development of pleural mesothelioma widely varies across different studies, ranging between 5% and 10%. This review provides an updated summary of emerging evidence about germline mutations in thoracic malignancies, focusing on pathogenetic mechanisms, clinical features, therapeutic implications, and screening recommendations for high-risk individuals.
Collapse
Affiliation(s)
- Giovanni Farinea
- Department of Oncology, University of Turin, San Luigi Hospital, Orbassano, Turin, Italy
| | - Veronica Crespi
- Department of Oncology, University of Turin, San Luigi Hospital, Orbassano, Turin, Italy
| | - Angela Listì
- Department of Oncology, University of Turin, San Luigi Hospital, Orbassano, Turin, Italy
| | - Luisella Righi
- Department of Oncology, University of Turin, San Luigi Hospital, Orbassano, Turin, Italy
| | - Paolo Bironzo
- Department of Oncology, University of Turin, San Luigi Hospital, Orbassano, Turin, Italy
| | - Alessandra Merlini
- Department of Oncology, University of Turin, San Luigi Hospital, Orbassano, Turin, Italy
| | - Umberto Malapelle
- Department of Public Health, University Federico II of Naples, Naples, Italy
| | - Silvia Novello
- Department of Oncology, University of Turin, San Luigi Hospital, Orbassano, Turin, Italy
| | | | - Francesco Passiglia
- Department of Oncology, University of Turin, San Luigi Hospital, Orbassano, Turin, Italy
| |
Collapse
|
37
|
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
|
38
|
Speight B, Hanson H, Turnbull C, Hardy S, Drummond J, Khorashad J, Wragg C, Page P, Parkin NW, Rio-Machin A, Fitzgibbon J, Kulasekararaj AG, Hamblin A, Talley P, McVeigh TP, Snape K. Germline predisposition to haematological malignancies: Best practice consensus guidelines from the UK Cancer Genetics Group (UKCGG), CanGene-CanVar and the NHS England Haematological Oncology Working Group. Br J Haematol 2023; 201:25-34. [PMID: 36744544 DOI: 10.1111/bjh.18675] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 01/10/2023] [Accepted: 01/17/2023] [Indexed: 02/07/2023]
Abstract
The implementation of whole genome sequencing and large somatic gene panels in haematological malignancies is identifying an increasing number of individuals with either potential or confirmed germline predisposition to haematological malignancy. There are currently no national or international best practice guidelines with respect to management of carriers of such variants or of their at-risk relatives. To address this gap, the UK Cancer Genetics Group (UKCGG), CanGene-CanVar and the NHS England Haematological Oncology Working Group held a workshop over two days on 28-29th April 2022, with the aim of establishing consensus guidelines on relevant clinical and laboratory pathways. The workshop focussed on the management of disease-causing germline variation in the following genes: DDX41, CEBPA, RUNX1, ANKRD26, ETV6, GATA2. Using a pre-workshop survey followed by structured discussion and in-meeting polling, we achieved consensus for UK best practice in several areas. In particular, high consensus was achieved on issues regarding standardised reporting, variant classification, multidisciplinary team working and patient support. The best practice recommendations from this meeting may be applicable to an expanding number of other genes in this setting.
Collapse
Affiliation(s)
- Beverley Speight
- East Anglian Medical Genetics Service, Addenbrooke's Treatment Centre, Addenbrooke's Hospital, Cambridge, UK
| | - Helen Hanson
- South West Thames Regional Genetics Service, St George's University Hospitals NHS Foundation Trust, London, UK
- Institute of Cancer Research, Sutton, London, UK
| | - Clare Turnbull
- Institute of Cancer Research, Sutton, London, UK
- Cancer Genetics Unit, The Royal Marsden NHS Foundation Trust, London, UK
| | - Steven Hardy
- National Disease Registration Service, NHS Digital, London, UK
| | - James Drummond
- East Anglian Medical Genetics Service, Addenbrooke's Treatment Centre, Addenbrooke's Hospital, Cambridge, UK
| | | | - Christopher Wragg
- South West Genomics Laboratory Hub, Bristol Genetics Laboratory, North Bristol NHS Trust, Pathology Building, Southmead Hospital, Bristol, UK
| | - Paula Page
- West Midlands Regional Genetics Laboratory, Birmingham Women's and Children's NHS Foundation Trust, Birmingham, UK
| | - Nicholas W Parkin
- Molecular Pathology Laboratory, Synnovis Analytics, King's College Hospital, London, UK
| | - Ana Rio-Machin
- Centre for Genomics and Computational Biology, Barts Cancer Institute, Queen Mary University of London, London, UK
| | - Jude Fitzgibbon
- Centre for Genomics and Computational Biology, Barts Cancer Institute, Queen Mary University of London, London, UK
| | - Austin Gladston Kulasekararaj
- King's College Hospital NHS Foundation Trust, London, UK
- National Institute for Health and Care Research and Wellcome King's Research Facility, London, UK
- King's College London, London, UK
| | - Angela Hamblin
- Oxford University Hospitals NHS Foundation Trust and Central and South Genomic Laboratory Hub, Oxford, UK
| | - Polly Talley
- Genomics Unit, NHS UK and NHS Improvement, Leeds, UK
- North East and Yorkshire Genomic Laboratory Hub, Leeds, UK
| | - Terri P McVeigh
- Institute of Cancer Research, Sutton, London, UK
- Cancer Genetics Unit, The Royal Marsden NHS Foundation Trust, London, UK
| | - Katie Snape
- South West Thames Regional Genetics Service, St George's University Hospitals NHS Foundation Trust, London, UK
- King's College Hospital NHS Foundation Trust, London, UK
| | | |
Collapse
|