1
|
Xie L, Chen J, Zheng Y, Sun Y, Zhang X, Chu L, Zhang Y. Somatic and germline aberrations in homologous recombination repair genes among Chinese high-risk breast cancer patients by multi-gene next-generation sequencing. Clin Transl Oncol 2025; 27:660-670. [PMID: 39043977 DOI: 10.1007/s12094-024-03599-x] [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: 05/12/2024] [Accepted: 07/01/2024] [Indexed: 07/25/2024]
Abstract
INTRODUCTION Recently, genes involved in homologous recombination repair (HRR) pathway have been extensively studied. However, the landscapes of HRR gene mutations remain poorly defined in Chinese high-risk breast cancer (BC) patients. Our study aims to identify the status of germline and somatic HRR gene mutations and their association with clinicopathological features in these patients. MATERIALS AND METHODS A total of 100 high-risk BC patients from our institution who underwent paired peripheral blood germline and BC tissues somatic 26 genes next-generation sequencing (NGS) from January 2018 to July 2023 were enrolled for retrospective analysis. RESULTS Out of 100 high-risk BC patients, 55 (55%) had at least one germline or somatic mutation in HRR genes. Among them, 22% carried germline pathogenic variants (19 BRCA1/2 and 3 non-BRCA genes), 9% harbored somatic pathogenic mutations (3 BRCA1/2 and 6 non-BRCA genes). Among high-risk factors, family history and early onset BC showed a correlation with HRR gene mutations (p < 0.05). BRCA1 germline and HRR gene somatic mutations showed a correlation with TNBC, but BRCA2 germline mutations were associated with Luminal B/HER2-negative BC (p < 0.05). Patients with HRR gene somatic pathogenic variant more likely had a lympho-vascular invasion and distant metastasis (p < 0.05). CONCLUSION The prevalence of HRR gene germline and somatic mutations were higher in Chinese BC patients with high risk factors. We strongly recommend that these high-risk BC patients receive comprehensive gene mutation testing, especially HRR genes, which are not only related to genetic consultation for BC patients and provide a theoretical basis for necessary prevention and individualized treatment.
Collapse
Affiliation(s)
- Ling Xie
- Department of Pathology, Jiangsu Province Hospital of Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China
| | - Jie Chen
- Department of Pathology, Jiangsu Province Hospital of Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China
| | - YanYing Zheng
- Department of Pathology, Jiangsu Province Hospital of Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China
| | - Yi Sun
- Department of Pathology, Jiangsu Province Hospital of Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China
| | - Xiang Zhang
- Department of Pathology, Jiangsu Province Hospital of Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China
| | - LeLe Chu
- Department of Pathology, Jiangsu Province Hospital of Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China
| | - YiFen Zhang
- Department of Pathology, Jiangsu Province Hospital of Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China.
| |
Collapse
|
2
|
Chapagain U, Huecker JB, Sun L. Morphologic Correlations With Homologous Recombination Deficiency in High-grade Serous Carcinomas. Int J Gynecol Pathol 2025:00004347-990000000-00217. [PMID: 39868725 DOI: 10.1097/pgp.0000000000001090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
High-grade serous carcinomas (HGSCs) with homologous recombination deficiency (HRD) respond favorably to platinum therapy and poly ADP ribose polymerase (PARP) inhibitors. Mutations in BRCA1 and BRCA2 commonly cause HRD and have been associated with Solid, pseudoEndometrioid, and Transitional-like (SET-like) histology. Mutations in other homologous recombination repair (HRR) genes as well as epigenetic changes can also result in HRD; however, morphologic correlates have not been well-explored in these cases. We hypothesized that HGSCs with HRD, regardless of the etiology, are associated with specific morphologic features. Forty-three cases of HGSC with genomic profiling, which included HRR gene mutation analysis and HRD score, were evaluated. The morphologic patterns, degree of nuclear atypia, necrosis, mitotic index, and tumor-infiltrating lymphocytes (TILs) were determined. The results showed that HRD-high status was significantly associated with the presence of BRCA1/2 mutation, SET-like morphology, geographic necrosis, and severe nuclear atypia. Additional HRR pathway genes with oncogenic mutations identified included ATM, BRIP1, BLM, FANCC, CDK12, CHEK2, RAD51C, and RAD51D. Almost one-third of HRD-high tumors did not have mutations in any HRR pathway genes identified. In conclusion, HGSC with HRD, regardless of BRCA1/2-status, was associated with SET-like morphology and more severe nuclear atypia. Identifying and reporting these patterns of tumor morphology can prompt genomic profiling with prognostic, therapeutic, and genetic counseling implications.
Collapse
Affiliation(s)
- Udita Chapagain
- Department of Pathology and Immunology, Washington University
| | - Julia B Huecker
- Center for Biostatistics and Data Science, Washington University, St. Louis, Missouri
| | - Lulu Sun
- Department of Pathology and Immunology, Washington University
| |
Collapse
|
3
|
Kechin A, Koryukov M, Mikheeva R, Filipenko M. Homologous recombination deficiency (HRD) diagnostics: underlying mechanisms and new perspectives. Cancer Metastasis Rev 2024; 44:19. [PMID: 39724448 DOI: 10.1007/s10555-024-10238-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Accepted: 12/20/2024] [Indexed: 12/28/2024]
Abstract
Homologous recombination deficiency (HRD) is considered a universal and effective sign of a tumor's sensitivity to poly(ADP-ribose) polymerase (PARP) inhibitors. HRD diagnostics have undergone several stages of transformations: from detection of point mutations in HR-related genes and large regions with loss of heterozygosity detected using single-nucleotide polymorphism arrays to whole-genome signatures of single-nucleotide variants, large genomic rearrangements (LGRs), and copy number alterations. All these methods have their own advantages and limitations. HRD tests, based on signatures of LGRs and copy number alterations, show in hindsight that some progenitor cells have possessed HRD status but not the current state of the genome. The aim of this review was to compare different methods of HRD detection and mechanisms of formation of HRD-specific LGRs. In the last several years, new data appeared implying a crucial role of proteins BRCA1 and BRCA2 in the resolution of stalled replication forks that may be associated with at least some of LGRs observed in HRD-positive tumors. Reviewing current knowledge on these mechanisms, distributions of different LGR types, and limitations of sequencing technologies and algorithms of data analysis, we offer some new perspectives on HRD diagnostics. We hope that this review will help to accelerate the development of new diagnostic approaches in this important field of molecular oncology.
Collapse
Affiliation(s)
- Andrey Kechin
- Institute of Chemical Biology and Fundamental Medicine, Novosibirsk, 630090, Russia.
- Novosibirsk State University, Novosibirsk, 630090, Russia.
| | - Maksim Koryukov
- Institute of Chemical Biology and Fundamental Medicine, Novosibirsk, 630090, Russia
- Novosibirsk State University, Novosibirsk, 630090, Russia
| | - Regina Mikheeva
- Institute of Chemical Biology and Fundamental Medicine, Novosibirsk, 630090, Russia
- Novosibirsk State University, Novosibirsk, 630090, Russia
| | - Maksim Filipenko
- Institute of Chemical Biology and Fundamental Medicine, Novosibirsk, 630090, Russia
| |
Collapse
|
4
|
Liu Z, Han L, Ji X, Wang X, Jian J, Zhai Y, Xu Y, Wang F, Wang X, Ning F. Multi-omics analysis and experiments uncover the function of cancer stemness in ovarian cancer and establish a machine learning-based model for predicting immunotherapy responses. Front Immunol 2024; 15:1486652. [PMID: 39726597 PMCID: PMC11670203 DOI: 10.3389/fimmu.2024.1486652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Accepted: 11/25/2024] [Indexed: 12/28/2024] Open
Abstract
Background The heterogeneity of cancer makes it challenging to predict its response to immunotherapy, highlighting the need to find reliable biomarkers for assessment. The sophisticated role of cancer stemness in mediating resistance to immune checkpoint inhibitors (ICIs) is still inadequately comprehended. Methods Genome-scale CRISPR screening of RNA sequencing data from Project Achilles was utilized to pinpoint crucial genes unique to Ovarian Cancer (OV). Thirteen publicly accessible OV transcriptomic datasets, seven pan-cancer ICI transcriptomic cohorts, and one single-cell RNA dataset from melanoma patients treated with PD-1 were utilized to scale a novel cancer stemness index (CSI). An OV single-cell RNA dataset was amassed and scrutinized to uncover the role of Small Nuclear Ribonucleoprotein Polypeptide E (SNRPE) in the tumor microenvironment (TME). Vitro experiments were performed to validate the function of SNRPE in promoting proliferation and migration of ovarian cancer. Results Through the analysis of extensive datasets on ovarian cancer, a specific gene set that impacts the stemness characteristics of tumors has been identified and we unveiled a negative correlation between cancer stemness, and benefits of ICI treatment in single cell ICI cohorts. This identified gene set underpinned the development of the CSI, a groundbreaking tool leveraging advanced machine learning to predict prognosis and immunotherapy responses in ovarian cancer patients. The accuracy of the CSI was further confirmed by applying PD1/PD-L1 ICI transcriptomic cohorts, with a mean AUC exceeding 0.8 for predicting tumor progression and immunotherapy benefits. Remarkably, when compared to existing immunotherapy and prognosis markers, CSI exhibited superior predictive capabilities across various datasets. Interestingly, our research unveiled that the amplification of SNRPE contribute to remodeling the TME and promoting the evasion of malignant cells from immune system recognition and SNRPE can server as a novel biomarker for predicting immunotherapy response. Conclusions A strong relationship between cancer stemness and the response to immunotherapy has been identified in our study. This finding provides valuable insights for devising efficient strategies to address immune evasion by targeting the regulation of genes associated with cellular stemness.
Collapse
Affiliation(s)
- Zhibing Liu
- Department of Oncology, Binzhou Medical University Hospital, Binzhou, Shandong, China
- Department of Oncology, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Lei Han
- Department of Reproductive Medicine, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Xiaoyu Ji
- Department of Oncology, Huashan Hospital Fudan University, Shanghai, China
| | - Xiaole Wang
- Department of Oncology, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Jinbo Jian
- Department of Oncology, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Yujie Zhai
- Department of Oncology, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Yingjiang Xu
- Department of Interventional Vascular Surgery, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Feng Wang
- Department of Oncology, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Xiuwen Wang
- Department of Oncology, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Fangling Ning
- Department of Oncology, Binzhou Medical University Hospital, Binzhou, Shandong, China
| |
Collapse
|
5
|
Azzollini J, Capone I, Duca M, Vingiani A, Piccolo A, Agnelli L, Tamborini E, Perrone F, Peissel B, Lorenzini D, Damian S, Vernieri C, Bianchi GV, Mantiero M, Ducceschi M, Polignano M, Niger M, Nichetti F, Proto C, Brambilla M, Colombo E, Stellato M, Conca E, Busico A, Manoukian S. Is tumour sequencing effective for the identification of germline BRCA1/2 pathogenic variant carriers? TUMORI JOURNAL 2024:3008916241280127. [PMID: 39311051 DOI: 10.1177/03008916241280127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
INTRODUCTION Tumour BRCA1/2 sequencing has progressively increased along with the expanding indications for poly(ADP-ribose) polymerase inhibitors. In our study, we investigated the feasibility and outcomes of a workflow for the identification of germline carriers based on tumour sequencing results. METHODS Between April 2020 and December 2022, BRCA1/2 tumour testing results from 2020 patients were reviewed. Analysed tumours included: 323 ovarian, 104 breast, 314 pancreas-biliary, 87 prostate, 374 gastrointestinal, 309 lung, and 509 less common histologies. Testing was performed through small (only BRCA1/2, 16%) or comprehensive (>50 genes) next-generation sequencing panels (84%). Patients with pathogenic/likely pathogenic variants were referred for genetic counselling and germline testing. RESULTS Tumour BRCA1/2 pathogenic variants were identified in 145 patients (7%). The pathogenic variant frequency ranged between 23% (75/323 ovarian) and 3.5% (11/314 pancreas-biliary). The highest frequency was observed in high-grade ovarian carcinomas (27%, 64/235). By 30 June 2023, 79 out of 145 patients (54%) underwent subsequent genetic counselling and germline testing. In these patients, mostly affected with ovarian carcinoma (67%, 53/79), 48 were confirmed germline pathogenic variants (61%). CONCLUSIONS In our tumour-to-germline testing approach, we observed the BRCA1/2 pathogenic variant frequency reported in other large unselected ovarian cancer cohorts, thus confirming its effectiveness in identifying putative germline carriers irrespective of eligibility for germline testing. As the range of tumours subjected to genetic testing broadens, this approach is expected to also be effective in other tumour settings for enhancing the identification of carriers, reducing the burden on genetic services, and avoiding unnecessary concerns related to germline testing.
Collapse
Affiliation(s)
- Jacopo Azzollini
- Unit of Medical Genetics, Department of Medical Oncology and Hematology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy
| | - Iolanda Capone
- Department of Diagnostic Innovation, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy
| | - Matteo Duca
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy
| | - Andrea Vingiani
- Department of Diagnostic Innovation, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milano, Italy
| | - Alberta Piccolo
- Department of Diagnostic Innovation, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy
| | - Luca Agnelli
- Department of Diagnostic Innovation, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy
| | - Elena Tamborini
- Department of Diagnostic Innovation, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy
| | - Federica Perrone
- Department of Diagnostic Innovation, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy
| | - Bernard Peissel
- Unit of Medical Genetics, Department of Medical Oncology and Hematology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy
| | - Daniele Lorenzini
- Department of Diagnostic Innovation, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milano, Italy
| | - Silvia Damian
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy
| | - Claudio Vernieri
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy
- IFOM ETS, The AIRC Institute of Molecular Oncology, Milano, Italy
| | - Giulia Valeria Bianchi
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy
| | - Mara Mantiero
- Department of Gynecologic Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy
| | - Monika Ducceschi
- Department of Gynecologic Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy
| | - Maggie Polignano
- Department of Gynecologic Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy
| | - Monica Niger
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy
| | - Federico Nichetti
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy
- Computational Oncology Group, Molecular Precision Oncology Program, National Center for Tumor Diseases and German Cancer Research Center, Heidelberg, Germany
| | - Claudia Proto
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy
| | - Marta Brambilla
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy
| | - Elena Colombo
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy
| | - Marco Stellato
- Genitourinary Medical Oncology, Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
- Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Roma, Italy
| | - Elena Conca
- Department of Diagnostic Innovation, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy
| | - Adele Busico
- Department of Diagnostic Innovation, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy
| | - Siranoush Manoukian
- Unit of Medical Genetics, Department of Medical Oncology and Hematology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy
| |
Collapse
|
6
|
Hao Y, Ji H, Gao L, Qu Z, Zhao Y, Chen J, Wang X, Ma X, Zhang G, Zhang T. Self-assembled carrier-free formulations based on medicinal and food active ingredients. Biomater Sci 2024; 12:6253-6273. [PMID: 39523875 DOI: 10.1039/d4bm00893f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
The popularity of medicinal plants, which have a unique system and are mostly used in compound form for the prevention and treatment of a wide range of diseases, is growing worldwide. In recent years, with advances in chemical separation and structural analysis techniques, many of the major bioactive molecules of medicinal plants have been identified. However, the active ingredients in medicinal plants often possess chemical characteristics, including poor water solubility, stability and bioavailability, which limit their therapeutic applications. To address this problem, self-assembly of small molecules from medicinal food sources provides a new strategy. Driven by various types of acting forces, medicinal small molecules with modifiable groups, multiple sites of action, hydrophobic side chains, and rigid backbones with self-assembly properties are able to form various supramolecular network hydrogels, nanoparticles, micelles, and other self-assemblies. This review first summarizes the forms of self-assemblies such as supramolecular network hydrogels, nanoparticles, and micelles at the level of the action site, and discusses the recent studies on the active ingredients in medicinal plants that can be used for self-assembly, in addition to summarizing the advantages of self-assemblies for a variety of disease applications, including wound healing, antitumor, anticancer, and diabetes mellitus. Finally, the problems of self-assemblers and the possible directions for future development are presented. We firmly believe that self-assemblers have the potential to develop effective compounds from drug-food homologous plants, providing valuable information for drug research and new strategies and perspectives for the modernization of Chinese medicine.
Collapse
Affiliation(s)
- Yuan Hao
- School of Chemistry and Chemical Engineering, North University of China, Taiyuan 030051, Shanxi, China.
| | - Haixia Ji
- School of Chemistry and Chemical Engineering, North University of China, Taiyuan 030051, Shanxi, China.
| | - Li Gao
- School of Chemistry and Chemical Engineering, North University of China, Taiyuan 030051, Shanxi, China.
| | - Zhican Qu
- Shanxi Nanolattix Health Technology Co., Ltd, Taiyuan 030051, Shanxi, China
| | - Yinghu Zhao
- School of Environment and Safety Engineering, North University of China, Taiyuan 030051, Shanxi, China
| | - Jiahui Chen
- School of Chemistry and Chemical Engineering, North University of China, Taiyuan 030051, Shanxi, China.
| | - Xintao Wang
- School of Chemistry and Chemical Engineering, North University of China, Taiyuan 030051, Shanxi, China.
| | - Xiaokai Ma
- School of Chemistry and Chemical Engineering, North University of China, Taiyuan 030051, Shanxi, China.
| | - Guangyu Zhang
- School of Chemistry and Chemical Engineering, North University of China, Taiyuan 030051, Shanxi, China.
| | - Taotao Zhang
- School of Chemistry and Chemical Engineering, North University of China, Taiyuan 030051, Shanxi, China.
| |
Collapse
|
7
|
Han W, Rexiati N, Wang Y, Liu T, Luo Y, Yang Z. Cases of consecutive ductal adenocarcinoma of the prostate carrying HRR mutation: case series and literature review. Oxf Med Case Reports 2024; 2024:omae124. [PMID: 39464223 PMCID: PMC11512693 DOI: 10.1093/omcr/omae124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 08/02/2024] [Accepted: 08/15/2024] [Indexed: 10/29/2024] Open
Affiliation(s)
- Weizhe Han
- Department of Urology, Zhongnan Hospital of Wuhan University, 169 Donghu Road, Wuhan 430071, China
| | - Nihati Rexiati
- Department of Urology, Zhongnan Hospital of Wuhan University, 169 Donghu Road, Wuhan 430071, China
| | - Yongzhi Wang
- Department of Urology, Zhongnan Hospital of Wuhan University, 169 Donghu Road, Wuhan 430071, China
| | - Tao Liu
- Department of Urology, Zhongnan Hospital of Wuhan University, 169 Donghu Road, Wuhan 430071, China
| | - Yongwen Luo
- Department of Urology, Zhongnan Hospital of Wuhan University, 169 Donghu Road, Wuhan 430071, China
| | - Zhonghua Yang
- Department of Urology, Zhongnan Hospital of Wuhan University, 169 Donghu Road, Wuhan 430071, China
| |
Collapse
|
8
|
Mavroeidi IA, Burghofer J, Kalbourtzis S, Taghizadeh H, Webersinke G, Piringer G, Kasper S, Schreil G, Liffers ST, Reichinger A, Kirchweger P, Heibl S, Hamacher R, Schmitt CA, Schuler M, Prager GW, Kersting D, Treckmann J, Schildhaus HU, Rumpold H, Siveke JT, Doleschal B. Understanding homologous recombination repair deficiency in biliary tract cancers: clinical implications and correlation with platinum sensitivity. ESMO Open 2024; 9:103630. [PMID: 39018588 PMCID: PMC11305189 DOI: 10.1016/j.esmoop.2024.103630] [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: 04/01/2024] [Revised: 05/28/2024] [Accepted: 05/29/2024] [Indexed: 07/19/2024] Open
Abstract
BACKGROUND Biliary tract cancers (BTCs) exhibit high mortality rates and significant heterogeneity in both clinical and molecular characteristics. This study aims to molecularly characterize a cohort of patients with BTC, with a specific focus on genomic alterations within homologous recombination repair (HRR) genes in a real-world setting. PATIENTS AND METHODS We carried out a retrospective analysis on 256 patients with BTC treated at five Austrian centers and one German comprehensive cancer center between 2016 and 2023 utilizing comprehensive genomic profiling platforms to assess HRR status and its correlation with clinical outcomes after platinum-based chemotherapy. RESULTS A total of 67 patients (27.5%) exhibited HRR gene mutations (HRRm), with the most common pathogenic alterations in BAP1 (9%), ARID1A (7.8%), and ATM (6.1%). Time to failure of the first-line strategy (TFS) between patients with HRRm and non-HRRm treated with platinum agents was 7.9 and 6.7 months, respectively [hazard ratio (HR) 0.89; P = 0.49]. The overall survival (OS) estimates at 6, 18, and 24 months were 82%, 45%, and 39% in the HRRm group (median 16.01 months) and 81%, 42%, and 22% in the HRR group (median 15.68 months), respectively (Fleming-Harrington test P = 0.0004; log-rank P = 0.022). Significance did not persist in the multivariate analysis (HR 0.72; 95% confidence interval 0.489-1.059; P = 0.095). An interaction between HRRm status and molecular-informed therapeutic strategies in later lines was noted. In the second-line treatment, OS following an irinotecan-based regimen was comparable to re-exposure to platinum-based agents (12.36 versus 10.13 months; HR 0.92; P = 0.85). No better outcome was noted for patients with HRRm versus patients with non-HRRm with second-line platinum agents (HR 1.45; P = 0.35). CONCLUSIONS Patients with HRRm with BTC showed a potential advantage in OS following platinum-based first-line chemotherapy, presumably attributed to enhanced opportunities for targetable coalterations. Further investigation is needed to outline HRR within the scope of BTCs and detail a clinically meaningful sensitivity to platinum agents or targeted approaches with poly (ADP-ribose) polymerase (PARP) inhibitors.
Collapse
Affiliation(s)
- I-A Mavroeidi
- Department of Medical Oncology, West German Cancer Center, University Hospital Essen, Essen; Division of Solid Tumor Translational Oncology, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Partner Site University Hospital Essen, Essen; Bridge Institute of Experimental Tumor Therapy, West German Cancer Center, University Hospital Essen, Essen, Germany
| | - J Burghofer
- Laboratory for Molecular Genetic Diagnostics, Ordensklinikum Linz, Linz, Austria
| | - S Kalbourtzis
- Institute of Pathology, West German Cancer Center, University Hospital Essen, Essen, Germany
| | - H Taghizadeh
- Department of Internal Medicine, Universitätsklinikum St. Pölten, St. Pölten
| | - G Webersinke
- Laboratory for Molecular Genetic Diagnostics, Ordensklinikum Linz, Linz, Austria
| | - G Piringer
- Department of Oncology and Hematology, Kepler University Hospital, Linz; Medical Faculty, Johannes Kepler University Linz, Linz
| | - S Kasper
- Department of Medical Oncology, West German Cancer Center, University Hospital Essen, Essen; Division of Solid Tumor Translational Oncology, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Partner Site University Hospital Essen, Essen
| | - G Schreil
- Department of Internal Medicine, State Hospital Pyhrn Eisenwurzen, Steyr
| | - S T Liffers
- Division of Solid Tumor Translational Oncology, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Partner Site University Hospital Essen, Essen; Bridge Institute of Experimental Tumor Therapy, West German Cancer Center, University Hospital Essen, Essen, Germany
| | - A Reichinger
- Department of Internal Medicine I for Hematology with Stem Cell Transplantation, Hemostaseology, and Medical Oncology, Ordensklinikum Linz, Linz
| | - P Kirchweger
- Medical Faculty, Johannes Kepler University Linz, Linz; Department of General and Visceral Surgery, Ordensklinikum Linz, Linz; Gastrointestinal Cancer Center, Ordensklinikum Linz, Linz
| | - S Heibl
- Department of Internal Medicine IV, Hospital Wels-Grieskirchen, Wels, Austria
| | - R Hamacher
- Department of Medical Oncology, West German Cancer Center, University Hospital Essen, Essen; Division of Solid Tumor Translational Oncology, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Partner Site University Hospital Essen, Essen
| | - C A Schmitt
- Department of Oncology and Hematology, Kepler University Hospital, Linz; Medical Faculty, Johannes Kepler University Linz, Linz
| | - M Schuler
- Department of Medical Oncology, West German Cancer Center, University Hospital Essen, Essen; Division of Solid Tumor Translational Oncology, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Partner Site University Hospital Essen, Essen; National Center for Tumor Diseases (NCT) West, Campus Essen, Essen, Germany
| | - G W Prager
- Department of Medicine I, Division of Oncology, Medical University Vienna, Vienna, Austria
| | - D Kersting
- Division of Solid Tumor Translational Oncology, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Partner Site University Hospital Essen, Essen; Department of Nuclear Medicine, University Hospital Essen, Essen
| | - J Treckmann
- Division of Solid Tumor Translational Oncology, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Partner Site University Hospital Essen, Essen; Department of General, Visceral and Transplantation Surgery, University Hospital Essen, Essen
| | - H-U Schildhaus
- Institute of Pathology, West German Cancer Center, University Hospital Essen, Essen, Germany; Discovery Life Sciences Biomarker GmbH und Pathologie Nordhessen, Kassel, Germany
| | - H Rumpold
- Medical Faculty, Johannes Kepler University Linz, Linz; Gastrointestinal Cancer Center, Ordensklinikum Linz, Linz
| | - J T Siveke
- Department of Medical Oncology, West German Cancer Center, University Hospital Essen, Essen; Division of Solid Tumor Translational Oncology, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Partner Site University Hospital Essen, Essen; Bridge Institute of Experimental Tumor Therapy, West German Cancer Center, University Hospital Essen, Essen, Germany.
| | - B Doleschal
- Medical Faculty, Johannes Kepler University Linz, Linz; Department of Internal Medicine I for Hematology with Stem Cell Transplantation, Hemostaseology, and Medical Oncology, Ordensklinikum Linz, Linz.
| |
Collapse
|
9
|
Piombino C, Pipitone S, Tonni E, Mastrodomenico L, Oltrecolli M, Tchawa C, Matranga R, Roccabruna S, D’Agostino E, Pirola M, Bacchelli F, Baldessari C, Baschieri MC, Dominici M, Sabbatini R, Vitale MG. Homologous Recombination Repair Deficiency in Metastatic Prostate Cancer: New Therapeutic Opportunities. Int J Mol Sci 2024; 25:4624. [PMID: 38731844 PMCID: PMC11083429 DOI: 10.3390/ijms25094624] [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: 03/25/2024] [Revised: 04/14/2024] [Accepted: 04/22/2024] [Indexed: 05/13/2024] Open
Abstract
More than 20% of metastatic prostate cancer carries genomic defects involving DNA damage repair pathways, mainly in homologous recombination repair-related genes. The recent approval of olaparib has paved the way to precision medicine for the treatment of metastatic prostate cancer with PARP inhibitors in this subset of patients, especially in the case of BRCA1 or BRCA2 pathogenic/likely pathogenic variants. In face of this new therapeutic opportunity, many issues remain unsolved. This narrative review aims to describe the relationship between homologous recombination repair deficiency and prostate cancer, the techniques used to determine homologous recombination repair status in prostate cancer, the crosstalk between homologous recombination repair and the androgen receptor pathway, the current evidence on PARP inhibitors activity in metastatic prostate cancer also in homologous recombination repair-proficient tumors, as well as emerging mechanisms of resistance to PARP inhibitors. The possibility of combination therapies including a PARP inhibitor is an attractive option, and more robust data are awaited from ongoing phase II and phase III trials outlined in this manuscript.
Collapse
Affiliation(s)
- Claudia Piombino
- Division of Oncology, Department of Oncology and Hematology, University Hospital of Modena, 41124 Modena, Italy; (C.P.); (S.P.); (E.T.); (L.M.); (M.O.); (C.T.); (R.M.); (S.R.); (E.D.); (M.P.); (C.B.); (M.D.); (R.S.)
| | - Stefania Pipitone
- Division of Oncology, Department of Oncology and Hematology, University Hospital of Modena, 41124 Modena, Italy; (C.P.); (S.P.); (E.T.); (L.M.); (M.O.); (C.T.); (R.M.); (S.R.); (E.D.); (M.P.); (C.B.); (M.D.); (R.S.)
| | - Elena Tonni
- Division of Oncology, Department of Oncology and Hematology, University Hospital of Modena, 41124 Modena, Italy; (C.P.); (S.P.); (E.T.); (L.M.); (M.O.); (C.T.); (R.M.); (S.R.); (E.D.); (M.P.); (C.B.); (M.D.); (R.S.)
| | - Luciana Mastrodomenico
- Division of Oncology, Department of Oncology and Hematology, University Hospital of Modena, 41124 Modena, Italy; (C.P.); (S.P.); (E.T.); (L.M.); (M.O.); (C.T.); (R.M.); (S.R.); (E.D.); (M.P.); (C.B.); (M.D.); (R.S.)
| | - Marco Oltrecolli
- Division of Oncology, Department of Oncology and Hematology, University Hospital of Modena, 41124 Modena, Italy; (C.P.); (S.P.); (E.T.); (L.M.); (M.O.); (C.T.); (R.M.); (S.R.); (E.D.); (M.P.); (C.B.); (M.D.); (R.S.)
| | - Cyrielle Tchawa
- Division of Oncology, Department of Oncology and Hematology, University Hospital of Modena, 41124 Modena, Italy; (C.P.); (S.P.); (E.T.); (L.M.); (M.O.); (C.T.); (R.M.); (S.R.); (E.D.); (M.P.); (C.B.); (M.D.); (R.S.)
| | - Rossana Matranga
- Division of Oncology, Department of Oncology and Hematology, University Hospital of Modena, 41124 Modena, Italy; (C.P.); (S.P.); (E.T.); (L.M.); (M.O.); (C.T.); (R.M.); (S.R.); (E.D.); (M.P.); (C.B.); (M.D.); (R.S.)
| | - Sara Roccabruna
- Division of Oncology, Department of Oncology and Hematology, University Hospital of Modena, 41124 Modena, Italy; (C.P.); (S.P.); (E.T.); (L.M.); (M.O.); (C.T.); (R.M.); (S.R.); (E.D.); (M.P.); (C.B.); (M.D.); (R.S.)
| | - Elisa D’Agostino
- Division of Oncology, Department of Oncology and Hematology, University Hospital of Modena, 41124 Modena, Italy; (C.P.); (S.P.); (E.T.); (L.M.); (M.O.); (C.T.); (R.M.); (S.R.); (E.D.); (M.P.); (C.B.); (M.D.); (R.S.)
| | - Marta Pirola
- Division of Oncology, Department of Oncology and Hematology, University Hospital of Modena, 41124 Modena, Italy; (C.P.); (S.P.); (E.T.); (L.M.); (M.O.); (C.T.); (R.M.); (S.R.); (E.D.); (M.P.); (C.B.); (M.D.); (R.S.)
| | - Francesca Bacchelli
- Clinical Trials Office, Division of Oncology, Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, 41124 Modena, Italy;
| | - Cinzia Baldessari
- Division of Oncology, Department of Oncology and Hematology, University Hospital of Modena, 41124 Modena, Italy; (C.P.); (S.P.); (E.T.); (L.M.); (M.O.); (C.T.); (R.M.); (S.R.); (E.D.); (M.P.); (C.B.); (M.D.); (R.S.)
| | - Maria Cristina Baschieri
- Laboratory of Cellular Therapy, Division of Oncology, Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, 41124 Modena, Italy;
| | - Massimo Dominici
- Division of Oncology, Department of Oncology and Hematology, University Hospital of Modena, 41124 Modena, Italy; (C.P.); (S.P.); (E.T.); (L.M.); (M.O.); (C.T.); (R.M.); (S.R.); (E.D.); (M.P.); (C.B.); (M.D.); (R.S.)
- Laboratory of Cellular Therapy, Division of Oncology, Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, 41124 Modena, Italy;
| | - Roberto Sabbatini
- Division of Oncology, Department of Oncology and Hematology, University Hospital of Modena, 41124 Modena, Italy; (C.P.); (S.P.); (E.T.); (L.M.); (M.O.); (C.T.); (R.M.); (S.R.); (E.D.); (M.P.); (C.B.); (M.D.); (R.S.)
| | - Maria Giuseppa Vitale
- Division of Oncology, Department of Oncology and Hematology, University Hospital of Modena, 41124 Modena, Italy; (C.P.); (S.P.); (E.T.); (L.M.); (M.O.); (C.T.); (R.M.); (S.R.); (E.D.); (M.P.); (C.B.); (M.D.); (R.S.)
| |
Collapse
|
10
|
Kong D, Wu Y, Liu Q, Huang C, Wang T, Huang Z, Gao Y, Li Y, Guo H. Functional analysis and validation of oncodrive gene AP3S1 in ovarian cancer through filtering of mutation data from whole-exome sequencing. Eur J Med Res 2024; 29:231. [PMID: 38609993 PMCID: PMC11015698 DOI: 10.1186/s40001-024-01814-7] [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: 01/19/2024] [Accepted: 03/26/2024] [Indexed: 04/14/2024] Open
Abstract
BACKGROUND High-grade serous ovarian carcinoma (HGSOC) is the most aggressive and prevalent subtype of ovarian cancer and accounts for a significant portion of ovarian cancer-related deaths worldwide. Despite advancements in cancer treatment, the overall survival rate for HGSOC patients remains low, thus highlighting the urgent need for a deeper understanding of the molecular mechanisms driving tumorigenesis and for identifying potential therapeutic targets. Whole-exome sequencing (WES) has emerged as a powerful tool for identifying somatic mutations and alterations across the entire exome, thus providing valuable insights into the genetic drivers and molecular pathways underlying cancer development and progression. METHODS Via the analysis of whole-exome sequencing results of tumor samples from 90 ovarian cancer patients, we compared the mutational landscape of ovarian cancer patients with that of TCGA patients to identify similarities and differences. The sequencing data were subjected to bioinformatics analysis to explore tumor driver genes and their functional roles. Furthermore, we conducted basic medical experiments to validate the results obtained from the bioinformatics analysis. RESULTS Whole-exome sequencing revealed the mutational profile of HGSOC, including BRCA1, BRCA2 and TP53 mutations. AP3S1 emerged as the most weighted tumor driver gene. Further analysis of AP3S1 mutations and expression demonstrated their associations with patient survival and the tumor immune response. AP3S1 knockdown experiments in ovarian cancer cells demonstrated its regulatory role in tumor cell migration and invasion through the TGF-β/SMAD pathway. CONCLUSION This comprehensive analysis of somatic mutations in HGSOC provides insight into potential therapeutic targets and molecular pathways for targeted interventions. AP3S1 was identified as being a key player in tumor immunity and prognosis, thus providing new perspectives for personalized treatment strategies. The findings of this study contribute to the understanding of HGSOC pathogenesis and provide a foundation for improved outcomes in patients with this aggressive disease.
Collapse
Affiliation(s)
- Deshui Kong
- Department of Obstetrics and Gynecology, Peking University Third Hospital, No.49 Huayuanbei Rd., Haidian District, Beijing, 100191, People's Republic of China
- National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital), Beijing, China
| | - Yu Wu
- Department of Obstetrics and Gynecology, Peking University Third Hospital, No.49 Huayuanbei Rd., Haidian District, Beijing, 100191, People's Republic of China
- National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital), Beijing, China
| | - Qiyu Liu
- Department of Obstetrics and Gynecology, Peking University Third Hospital, No.49 Huayuanbei Rd., Haidian District, Beijing, 100191, People's Republic of China
- National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital), Beijing, China
| | - Cuiyu Huang
- Department of Obstetrics and Gynecology, Peking University Third Hospital, No.49 Huayuanbei Rd., Haidian District, Beijing, 100191, People's Republic of China
- National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital), Beijing, China
| | - Tongxia Wang
- Department of Obstetrics and Gynecology, Peking University Third Hospital, No.49 Huayuanbei Rd., Haidian District, Beijing, 100191, People's Republic of China
- National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital), Beijing, China
| | - Zongyao Huang
- Department of Obstetrics and Gynecology, Peking University Third Hospital, No.49 Huayuanbei Rd., Haidian District, Beijing, 100191, People's Republic of China
- National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital), Beijing, China
| | - Yan Gao
- Department of Obstetrics and Gynecology, Peking University Third Hospital, No.49 Huayuanbei Rd., Haidian District, Beijing, 100191, People's Republic of China
- National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital), Beijing, China
| | - Yuan Li
- Department of Obstetrics and Gynecology, Peking University Third Hospital, No.49 Huayuanbei Rd., Haidian District, Beijing, 100191, People's Republic of China.
- National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital), Beijing, China.
| | - Hongyan Guo
- Department of Obstetrics and Gynecology, Peking University Third Hospital, No.49 Huayuanbei Rd., Haidian District, Beijing, 100191, People's Republic of China.
- National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital), Beijing, China.
| |
Collapse
|
11
|
Edsjö A, Gisselsson D, Staaf J, Holmquist L, Fioretos T, Cavelier L, Rosenquist R. Current and emerging sequencing-based tools for precision cancer medicine. Mol Aspects Med 2024; 96:101250. [PMID: 38330674 DOI: 10.1016/j.mam.2024.101250] [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: 11/14/2023] [Accepted: 01/22/2024] [Indexed: 02/10/2024]
Abstract
Current precision cancer medicine is dependent on the analyses of a plethora of clinically relevant genomic aberrations. During the last decade, next-generation sequencing (NGS) has gradually replaced most other methods for precision cancer diagnostics, spanning from targeted tumor-informed assays and gene panel sequencing to global whole-genome and whole-transcriptome sequencing analyses. The shift has been impelled by a clinical need to assess an increasing number of genomic alterations with diagnostic, prognostic and predictive impact, including more complex biomarkers (e.g. microsatellite instability, MSI, and homologous recombination deficiency, HRD), driven by the parallel development of novel targeted therapies and enabled by the rapid reduction in sequencing costs. This review focuses on these sequencing-based methods, puts their emergence in a historic perspective, highlights their clinical utility in diagnostics and decision-making in pediatric and adult cancer, as well as raises challenges for their clinical implementation. Finally, the importance of applying sensitive tools for longitudinal monitoring of treatment response and detection of measurable residual disease, as well as future avenues in the rapidly evolving field of sequencing-based methods are discussed.
Collapse
Affiliation(s)
- Anders Edsjö
- Department of Clinical Genetics, Pathology and Molecular Diagnostics, Office for Medical Services, Region Skåne, Lund, Sweden; Division of Pathology, Department of Clinical Sciences, Lund University, Lund, Sweden.
| | - David Gisselsson
- Department of Clinical Genetics, Pathology and Molecular Diagnostics, Office for Medical Services, Region Skåne, Lund, Sweden; Division of Clinical Genetics, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Johan Staaf
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, Medicon Village, Lund, Sweden
| | - Louise Holmquist
- Department of Clinical Genetics, Pathology and Molecular Diagnostics, Office for Medical Services, Region Skåne, Lund, Sweden
| | - Thoas Fioretos
- Department of Clinical Genetics, Pathology and Molecular Diagnostics, Office for Medical Services, Region Skåne, Lund, Sweden; Division of Clinical Genetics, Department of Laboratory Medicine, Lund University, Lund, Sweden; Clinical Genomics Lund, Science for Life Laboratory, Lund University, Lund, Sweden
| | - Lucia Cavelier
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden; Clinical Genetics and Genomics, Karolinska University Hospital, Stockholm, Sweden
| | - Richard Rosenquist
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden; Clinical Genetics and Genomics, Karolinska University Hospital, Stockholm, Sweden; Genomic Medicine Center Karolinska, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
12
|
Daly GR, AlRawashdeh MM, McGrath J, Dowling GP, Cox L, Naidoo S, Vareslija D, Hill ADK, Young L. PARP Inhibitors in Breast Cancer: a Short Communication. Curr Oncol Rep 2024; 26:103-113. [PMID: 38236558 PMCID: PMC10891270 DOI: 10.1007/s11912-023-01488-0] [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] [Accepted: 12/11/2023] [Indexed: 01/19/2024]
Abstract
PURPOSE OF REVIEW In the last decade, poly (ADP-ribose) polymerase (PARP) inhibitors have been approved in the treatment of several cancers, such as breast and ovarian cancer. This article aims to discuss the current uses, limitations, and future directions for PARP inhibitors (PARPis) in the treatment of breast cancer. RECENT FINDINGS Following the results of the OlympiAD and EMBRACA trials, PARPis were approved in HER2-negative breast cancer with a germline BRCA mutation. We reviewed this class of drugs' mechanism of action, efficacy, and limitations, as well as further studies that discussed resistance, impaired homologous recombination repair (HRR), and the combination of PARPis with other drugs. Improving understanding of HRR, increasing the ability to target resistance, and combining PARPis with other novel agents are continuing to increase the clinical utility of PARPis.
Collapse
Affiliation(s)
- Gordon R Daly
- The Department of Surgery, Royal College of Surgeons in Ireland, Dublin, Ireland.
- The Department of Surgery, Beaumont Hospital, Dublin, Ireland.
| | - Maen Monketh AlRawashdeh
- The Department of Surgery, Royal College of Surgeons in Ireland, Dublin, Ireland
- The Department of Surgery, Beaumont Hospital, Dublin, Ireland
| | - Jason McGrath
- The Department of Surgery, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Gavin P Dowling
- The Department of Surgery, Royal College of Surgeons in Ireland, Dublin, Ireland
- The Department of Surgery, Beaumont Hospital, Dublin, Ireland
| | - Luke Cox
- The Department of Surgery, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Sindhuja Naidoo
- The Department of Surgery, Royal College of Surgeons in Ireland, Dublin, Ireland
- The Department of Surgery, Beaumont Hospital, Dublin, Ireland
| | - Damir Vareslija
- The Department of Surgery, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Arnold D K Hill
- The Department of Surgery, Royal College of Surgeons in Ireland, Dublin, Ireland
- The Department of Surgery, Beaumont Hospital, Dublin, Ireland
| | - Leonie Young
- The Department of Surgery, Royal College of Surgeons in Ireland, Dublin, Ireland
| |
Collapse
|
13
|
Mahlow J, Barry M, Albertson DJ, Jo YJ, Balatico M, Seasor T, Gebrael G, Kumar SA, Sayegh N, Tripathi N, Agarwal N, Swami U, Sirohi D. Histologic patterns in prostatic adenocarcinoma are not predictive of mutations in the homologous recombination repair pathway. Hum Pathol 2024; 144:28-33. [PMID: 38278448 DOI: 10.1016/j.humpath.2024.01.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Revised: 01/17/2024] [Accepted: 01/22/2024] [Indexed: 01/28/2024]
Abstract
Somatic or germline homologous recombination repair (HRR) pathway gene mutations are commonly detected in prostate cancer, especially in advanced disease, and are associated with response to poly (ADP-ribose) polymerase (PARP) inhibitors. In this study, we evaluated whether histological patterns are predictive of HRR pathway gene mutations. The study population comprised 130 patients with advanced prostate carcinoma who underwent comprehensive genomic profiling (CGP) of tumor tissue at a CLIA-certified laboratory. HRR genes in the study included BRCA1, BRCA2, ATM, BARD1, BRIP, CHEK2, MRE11A, NBN, PALB2, RAD51C, RAD51D, EMSY, ATR, CHEK1, and FAM175A. Overall, 38 patients had mutations in BRCA1/2, 36 in other HRR genes, and 56 were negative for HRR mutations. All cases were re-reviewed and quantified by two genitourinary pathologists blinded to mutational status for the following histological patterns of prostate carcinoma: cribriform, ductal, intraductal carcinoma (IDC), small cell carcinoma, signet ring-like pattern, and lobular carcinoma-like pattern. Discordances were resolved by consensus review. Histologic patterns were analyzed for any correlation with mutations in HRR pathway genes (grouped as BRCA1/2 mutated or non-BRCA1/2 mutated) compared to tumors without mutations in HRR genes by Chi-square testing. Patterns with >20 % and >30 % of tumor volume were additionally evaluated for correlation with mutational status. We found no significant association between HRR pathway mutations and cribriform pattern, IDC, ductal carcinoma, small cell carcinoma, signet ring-like pattern, or lobular carcinoma-like patterns. Tumors with >20 % or >30 % histologic patterns by volume also demonstrated no significant association with mutational status. This study suggests that histopathologic examination alone is insufficient to distinguish prostate cancer with germline or somatic mutations in HRR pathway genes, highlighting the continuing importance of ancillary molecular diagnostics in guiding therapy selection for prostate cancer patients who may benefit from PARP inhibitors.
Collapse
Affiliation(s)
- Jon Mahlow
- Department of Pathology, University of Utah and ARUP Laboratories, Salt Lake City, UT, USA
| | - Marc Barry
- Department of Pathology, University of Utah and ARUP Laboratories, Salt Lake City, UT, USA
| | - Daniel J Albertson
- Department of Pathology, University of Utah and ARUP Laboratories, Salt Lake City, UT, USA
| | - Yeon Jung Jo
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Michael Balatico
- Department of Pathology, University of Utah and ARUP Laboratories, Salt Lake City, UT, USA
| | - Tori Seasor
- Department of Pathology, University of Utah and ARUP Laboratories, Salt Lake City, UT, USA
| | - Georges Gebrael
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Shruti A Kumar
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Nicolas Sayegh
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Nishita Tripathi
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Neeraj Agarwal
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Umang Swami
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA.
| | - Deepika Sirohi
- Department of Pathology, University of Utah and ARUP Laboratories, Salt Lake City, UT, USA.
| |
Collapse
|
14
|
Leman R, Muller E, Legros A, Goardon N, Chentli I, Atkinson A, Tranchant A, Castera L, Krieger S, Ricou A, Boulouard F, Joly F, Boucly R, Dumont A, Basset N, Coulet F, Chevalier LM, Rouleau E, Leitner K, González-Martin A, Gargiulo P, Lück HJ, Genestie C, Ray-Coquard I, Pujade-Lauraine E, Vaur D. Validation of the Clinical Use of GIScar, an Academic-developed Genomic Instability Score Predicting Sensitivity to Maintenance Olaparib for Ovarian Cancer. Clin Cancer Res 2023; 29:4419-4429. [PMID: 37756555 PMCID: PMC10618649 DOI: 10.1158/1078-0432.ccr-23-0898] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 06/12/2023] [Accepted: 08/15/2023] [Indexed: 09/29/2023]
Abstract
PURPOSE The optimal application of maintenance PARP inhibitor therapy for ovarian cancer requires accessible, robust, and rapid testing of homologous recombination deficiency (HRD). However, in many countries, access to HRD testing is problematic and the failure rate is high. We developed an academic HRD test to support treatment decision-making. EXPERIMENTAL DESIGN Genomic Instability Scar (GIScar) was developed through targeted sequencing of a 127-gene panel to determine HRD status. GIScar was trained from a noninterventional study with 250 prospectively collected ovarian tumor samples. GIScar was validated on 469 DNA tumor samples from the PAOLA-1 trial evaluating maintenance olaparib for newly diagnosed ovarian cancer, and its predictive value was compared with Myriad Genetics MyChoice (MGMC). RESULTS GIScar showed significant correlation with MGMC HRD classification (kappa statistics: 0.780). From PAOLA-1 samples, more HRD-positive tumors were identified by GIScar (258) than MGMC (242), with a lower proportion of inconclusive results (1% vs. 9%, respectively). The HRs for progression-free survival (PFS) with olaparib versus placebo were 0.45 [95% confidence interval (CI), 0.33-0.62] in GIScar-identified HRD-positive BRCA-mutated tumors, 0.50 (95% CI, 0.31-0.80) in HRD-positive BRCA-wild-type tumors, and 1.02 (95% CI, 0.74-1.40) in HRD-negative tumors. Tumors identified as HRD positive by GIScar but HRD negative by MGMC had better PFS with olaparib (HR, 0.23; 95% CI, 0.07-0.72). CONCLUSIONS GIScar is a valuable diagnostic tool, reliably detecting HRD and predicting sensitivity to olaparib for ovarian cancer. GIScar showed high analytic concordance with MGMC test and fewer inconclusive results. GIScar is easily implemented into diagnostic laboratories with a rapid turnaround.
Collapse
Affiliation(s)
- Raphaël Leman
- Laboratoire de Biologie et de Génétique du Cancer, Centre François Baclesse, Caen, France
- Inserm U1245, Cancer Brain and Genome, Normandie Université, UNICAEN, FHU G4 Génomique, Rouen, France
| | - Etienne Muller
- Laboratoire de Biologie et de Génétique du Cancer, Centre François Baclesse, Caen, France
- Inserm U1245, Cancer Brain and Genome, Normandie Université, UNICAEN, FHU G4 Génomique, Rouen, France
| | - Angelina Legros
- Laboratoire de Biologie et de Génétique du Cancer, Centre François Baclesse, Caen, France
| | - Nicolas Goardon
- Laboratoire de Biologie et de Génétique du Cancer, Centre François Baclesse, Caen, France
- Inserm U1245, Cancer Brain and Genome, Normandie Université, UNICAEN, FHU G4 Génomique, Rouen, France
| | - Imène Chentli
- Laboratoire de Biologie et de Génétique du Cancer, Centre François Baclesse, Caen, France
| | - Alexandre Atkinson
- Laboratoire de Biologie et de Génétique du Cancer, Centre François Baclesse, Caen, France
- Inserm U1245, Cancer Brain and Genome, Normandie Université, UNICAEN, FHU G4 Génomique, Rouen, France
| | - Aurore Tranchant
- Laboratoire de Biologie et de Génétique du Cancer, Centre François Baclesse, Caen, France
| | - Laurent Castera
- Laboratoire de Biologie et de Génétique du Cancer, Centre François Baclesse, Caen, France
- Inserm U1245, Cancer Brain and Genome, Normandie Université, UNICAEN, FHU G4 Génomique, Rouen, France
| | - Sophie Krieger
- Laboratoire de Biologie et de Génétique du Cancer, Centre François Baclesse, Caen, France
- Inserm U1245, Cancer Brain and Genome, Normandie Université, UNICAEN, FHU G4 Génomique, Rouen, France
| | - Agathe Ricou
- Laboratoire de Biologie et de Génétique du Cancer, Centre François Baclesse, Caen, France
- Inserm U1245, Cancer Brain and Genome, Normandie Université, UNICAEN, FHU G4 Génomique, Rouen, France
| | - Flavie Boulouard
- Laboratoire de Biologie et de Génétique du Cancer, Centre François Baclesse, Caen, France
- Inserm U1245, Cancer Brain and Genome, Normandie Université, UNICAEN, FHU G4 Génomique, Rouen, France
| | - Florence Joly
- Clinical Research, Centre François Baclesse, Caen, France
| | - Romain Boucly
- Unité d'Oncologie Moléculaire Humaine, Centre Oscar Lambret, Lille, France
| | - Aurélie Dumont
- Unité d'Oncologie Moléculaire Humaine, Centre Oscar Lambret, Lille, France
| | - Noémie Basset
- Département de Génétique Médicale, UF d'Onco-Angiogénétique et Génomique des Tumeurs Solides, Hôpital Pitié Salpêtrière APHP, Paris, France
- Sorbonne Université, Paris, France
| | - Florence Coulet
- Département de Génétique Médicale, UF d'Onco-Angiogénétique et Génomique des Tumeurs Solides, Hôpital Pitié Salpêtrière APHP, Paris, France
- Sorbonne Université, Paris, France
| | - Louise-Marie Chevalier
- Unité de Génomique Fonctionnelle, Institut de Cancérologie de l'Ouest, Angers, France
- Université Angers, Nantes Université, Inserm, CNRS, CRCI2NA, SFR ICAT, Angers, France
| | - Etienne Rouleau
- Service de Génétique des Tumeurs, Gustave Roussy, Villejuif, France
| | - Katharina Leitner
- Department of Obstetrics and Gynecology, Medical University Innsbruck, Innsbruck, Austria
- AGO Austria, Vienna, Austria
| | - Antonio González-Martin
- Department of Medical Oncology and Program in Solid Tumors-Cima, Cancer Center Clinica Universidad de Navarra, Madrid, Spain
- GEICO, Cádiz, Spain
| | - Piera Gargiulo
- Clinical Trials Unit, Istituto Nazionale Tumori IRCCS Fondazione G. Pascale, Naples, Italy
- MITO, Italy
| | - Hans-Joachim Lück
- Gynäkologisch-Onkologische Praxis Hannover, Hannover, Germany
- AGO, Wiesbaden, Germany
| | | | - Isabelle Ray-Coquard
- Association de Recherche Cancers Gynécologiques (ARCAGY), Paris, France
- Groupe d'Investigateurs Nationaux pour l'Etude des Cancers Ovariens et du sein (GINECO), France
| | - Eric Pujade-Lauraine
- Groupe d'Investigateurs Nationaux pour l'Etude des Cancers Ovariens et du sein (GINECO), France
| | - Dominique Vaur
- Laboratoire de Biologie et de Génétique du Cancer, Centre François Baclesse, Caen, France
- Inserm U1245, Cancer Brain and Genome, Normandie Université, UNICAEN, FHU G4 Génomique, Rouen, France
| |
Collapse
|
15
|
Minaguchi T, Shikama A, Akiyama A, Satoh T. Molecular biomarkers for facilitating genome‑directed precision medicine in gynecological cancer (Review). Oncol Lett 2023; 26:426. [PMID: 37664647 PMCID: PMC10472042 DOI: 10.3892/ol.2023.14012] [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: 05/18/2023] [Accepted: 07/17/2023] [Indexed: 09/05/2023] Open
Abstract
Prominent recent advancements in cancer treatment include the development and clinical application of next-generation sequencing (NGS) technologies, alongside a diverse array of novel molecular targeting therapeutics. NGS has enabled the high-speed and low-cost sequencing of whole genomes in individual patients, which has opened the era of genome-based precision medicine. The development of numerous molecular targeting agents, including anti-VEGF antibodies, poly (ADP-ribose) polymerase inhibitors and immune checkpoint inhibitors, have all improved the efficacy of systemic cancer therapy. Accumulating bench and translational research evidence has led to identification of various cancer-related biomarker profiles. In particular, companion diagnostics have been developed for some of these biomarkers, which can be clinically applied and are now widely used for guiding cancer therapies. Selecting biomarkers accurately will improve therapeutic efficacy, avoid overtreatment, enable earlier diagnosis and reduce the cost of preventing and treating gynecological cancer. Therefore, biomarkers are fast becoming indispensable tools in the practice of genome-directed precision medicine. In the present review, the current evidence of cancer-related biomarkers in the field of gynecological oncology, their molecular interpretations and future perspectives are outlined. The aim of the present review is to provide potentially useful information for the formulation of clinical trials.
Collapse
Affiliation(s)
- Takeo Minaguchi
- Department of Obstetrics and Gynecology, Institute of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Ayumi Shikama
- Department of Obstetrics and Gynecology, Institute of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Azusa Akiyama
- Department of Obstetrics and Gynecology, Institute of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Toyomi Satoh
- Department of Obstetrics and Gynecology, Institute of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| |
Collapse
|
16
|
Giannini A, Di Dio C, Di Donato V, D'oria O, Salerno MG, Capalbo G, Cuccu I, Perniola G, Muzii L, Bogani G. PARP Inhibitors in Newly Diagnosed and Recurrent Ovarian Cancer. Am J Clin Oncol 2023; 46:414-419. [PMID: 37314974 DOI: 10.1097/coc.0000000000001024] [Citation(s) in RCA: 51] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Ovarian cancer is the most lethal gynecologic malignancy, characterized by a high death-to-incidence ratio. Platinum-based chemotherapy is the mainstay of treatment for newly diagnosed and platinum-sensitive recurrent ovarian cancer. Poly (ADP-ribose) polymerase inhibitors (PARP inhibitors) have been incorporated into the treatment strategy for ovarian cancer. PARP inhibitors showed particular benefit for patients harboring defects in DNA repair pathways. Accumulating evidence showed that PARP inhibitors provide a benefit in newly diagnosed advanced ovarian cancer, even in the absence of BRCA mutation, as reported in the PRIMA, PRIME, and ATHENA-mono trials. Interestingly, the PAOLA-1 study provides another important finding, supporting the adoption of olaparib plus bevacizumab in patients with homologous recombination deficiency. Although those results are exciting, several patients develop resistance to PARP inhibitors. Hence, new combinations are under investigation to identify new treatment strategies to overcome this resistance. Currently, researchers are focused on the possibility to adopt PARP inhibitors even in the setting of platinum-resistant disease. The present critical review aims to report the current landscape and further perspective for strengthening PARP inhibitors' effectiveness in newly diagnosed and recurrent ovarian cancer.
Collapse
Affiliation(s)
- Andrea Giannini
- Department of Gynecological, Obstetrical, and Urological Sciences, "Sapienza" University of Rome, Rome, Italy
| | - Camilla Di Dio
- Department of Gynecological, Obstetrical, and Urological Sciences, "Sapienza" University of Rome, Rome, Italy
| | - Violante Di Donato
- Department of Gynecological, Obstetrical, and Urological Sciences, "Sapienza" University of Rome, Rome, Italy
| | - Ottavia D'oria
- Department of Woman's and Child's Health, Obstetrics and Gynecological Unit, San Camillo-Forlanini Hospital, Rome, Italy
| | - Maria Giovanna Salerno
- Department of Woman's and Child's Health, Obstetrics and Gynecological Unit, San Camillo-Forlanini Hospital, Rome, Italy
| | - Giuseppe Capalbo
- Department of Gynecological, Obstetrical, and Urological Sciences, "Sapienza" University of Rome, Rome, Italy
| | - Ilaria Cuccu
- Department of Gynecological, Obstetrical, and Urological Sciences, "Sapienza" University of Rome, Rome, Italy
| | - Giorgia Perniola
- Department of Gynecological, Obstetrical, and Urological Sciences, "Sapienza" University of Rome, Rome, Italy
| | - Ludovico Muzii
- Department of Gynecological, Obstetrical, and Urological Sciences, "Sapienza" University of Rome, Rome, Italy
| | - Giorgio Bogani
- Department of Gynecological, Obstetrical, and Urological Sciences, "Sapienza" University of Rome, Rome, Italy
| |
Collapse
|
17
|
Castellano G, Corti C, Boldrini L, Gervaso L, Criscitiello C, Curigliano G. Risk of thromboembolic events in patients with metastatic solid tumors treated with PARP inhibitors: A systematic review and meta-analysis of phase 3 randomized controlled trials. Cancer Treat Rev 2023; 119:102601. [PMID: 37473517 DOI: 10.1016/j.ctrv.2023.102601] [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: 05/29/2023] [Accepted: 07/15/2023] [Indexed: 07/22/2023]
Abstract
BACKGROUND AND SCOPE Poly(ADP-ribose) polymerase inhibitors (PARPi) have revolutionized cancer treatment in recent years. These drugs present a favorable safety profile, even though the potential risk of thromboembolic events (TEs) during their use has not been addressed yet. In addition, PARPi have been involved in an active scientific debate regarding non-oncologic indications, particularly during the Coronavirus Disease 2019 pandemic, including potential anti-thromboembolic effect. METHODS To clarify whether patients treated with PARPi for metastatic solid tumors are either at increased or decreased risk of TEs, we conducted a systematic review of the literature and meta-analysis, including all phase 3 randomized controlled trials (RCTs) which investigated PARPi in this setting. Search was conducted through Medline, EMBASE, Pubmed, SCOPUS and Google Scholar in February 2023, including the proceedings of the principal oncology meetings of the last 10 years, with no time restriction. For each included study, frequencies of TEs in experimental and control arm were collected. RESULTS Our search identified 2,369 reports, of which 20 were lastly selected. A total of 4,946 patients were included, across 12 different RCTs. The meta-analysis did not demonstrate either an increased or a reduced risk in TEs in patients treated with PARPi for metastatic disease (OR 1.50, range: 1.00-2.24; 95% CI; P = 0.050), with low heterogeneity and low publication bias. CONCLUSION Although our research did not confirm either increased or decreased risk of TEs for PARPi use, no safety alerts emerged. Thromboembolic risk assessment models should always be integrated in daily clinical routine, to identify high-risk patients.
Collapse
Affiliation(s)
- Grazia Castellano
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan, Italy; Department of Oncology and Hemato-Oncology (DIPO), University of Milan, Milan, Italy
| | - Chiara Corti
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan, Italy; Department of Oncology and Hemato-Oncology (DIPO), University of Milan, Milan, Italy.
| | - Laura Boldrini
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan, Italy; Department of Oncology and Hemato-Oncology (DIPO), University of Milan, Milan, Italy
| | - Lorenzo Gervaso
- Division of Gastrointestinal and Neuroendocrine Tumors, European Institute of Oncology, IRCCS, Milan, Italy; Molecular Medicine Program, University of Pavia, Pavia, Italy
| | - Carmen Criscitiello
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan, Italy; Department of Oncology and Hemato-Oncology (DIPO), University of Milan, Milan, Italy
| | - Giuseppe Curigliano
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan, Italy; Department of Oncology and Hemato-Oncology (DIPO), University of Milan, Milan, Italy
| |
Collapse
|
18
|
Jank P, Leichsenring J, Kolb S, Hoffmann I, Bischoff P, Kunze CA, Dragomir MP, Gleitsmann M, Jesinghaus M, Schmitt WD, Kulbe H, Sers C, Stenzinger A, Sehouli J, Braicu IE, Westhoff C, Horst D, Denkert C, Gröschel S, Taube ET. High EVI1 and PARP1 expression as favourable prognostic markers in high-grade serous ovarian carcinoma. J Ovarian Res 2023; 16:150. [PMID: 37525239 PMCID: PMC10388497 DOI: 10.1186/s13048-023-01239-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Accepted: 07/16/2023] [Indexed: 08/02/2023] Open
Abstract
BACKGROUND Mechanisms of development and progression of high-grade serous ovarian cancer (HGSOC) are poorly understood. EVI1 and PARP1, part of TGF-ß pathway, are upregulated in cancers with DNA repair deficiencies with DNA repair deficiencies and may influce disease progression and survival. Therefore we questioned the prognostic significance of protein expression of EVI1 alone and in combination with PARP1 and analyzed them in a cohort of patients with HGSOC. METHODS For 562 HGSOC patients, we evaluated EVI1 and PARP1 expression by immunohistochemical staining on tissue microarrays with QuPath digital semi-automatic positive cell detection. RESULTS High EVI1 expressing (> 30% positive tumor cells) HGSOC were associated with improved progression-free survival (PFS) (HR = 0.66, 95% CI: 0.504-0.852, p = 0.002) and overall survival (OS) (HR = 0.45, 95% CI: 0.352-0.563, p < 0.001), including multivariate analysis. Most interestingly, mutual high expression of both proteins identifies a group with particularly good prognosis. Our findings were proven technically and clinically using bioinformatical data sets for single-cell sequencing, copy number variation and gene as well as protein expression. CONCLUSIONS EVI1 and PARP1 are robust prognostic biomarkers for favorable prognosis in HGSOC and imply further research with respect to their reciprocity.
Collapse
Affiliation(s)
- Paul Jank
- Institute of Pathology, Philipps-University Marburg, University Hospital Marburg (UKGM), Marburg, Germany
| | - Jonas Leichsenring
- Institute of Pathology, Zytologie Und Molekulare Diagnostik, REGIOMED, Klinikum Coburg, Coburg, Germany
| | - Svenja Kolb
- Department of Gynecology, Vivantes Netzwerk Für Gesundheit GmbH Berlin, Vivantes Hospital Neukölln, Rudower Straße 48, 12351, Berlin, Germany
| | - Inga Hoffmann
- Institute of Pathology, Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt Universität Zu Berlin, CCM, Charitéplatz 1, 10117, Berlin, Germany
| | - Philip Bischoff
- Institute of Pathology, Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt Universität Zu Berlin, CCM, Charitéplatz 1, 10117, Berlin, Germany
| | - Catarina Alisa Kunze
- Institute of Pathology, Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt Universität Zu Berlin, CCM, Charitéplatz 1, 10117, Berlin, Germany
| | - Mihnea P Dragomir
- Institute of Pathology, Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt Universität Zu Berlin, CCM, Charitéplatz 1, 10117, Berlin, Germany
| | - Moritz Gleitsmann
- Institute of Pathology, Philipps-University Marburg, University Hospital Marburg (UKGM), Marburg, Germany
| | - Moritz Jesinghaus
- Institute of Pathology, Philipps-University Marburg, University Hospital Marburg (UKGM), Marburg, Germany
| | - Wolfgang D Schmitt
- Institute of Pathology, Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt Universität Zu Berlin, CCM, Charitéplatz 1, 10117, Berlin, Germany
| | - Hagen Kulbe
- Tumorbank Ovarian Cancer Network, Charité, Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität Zu Berlin, and Berlin Institute of Health, 10117, Berlin, Germany
- Department of Gynecology, European Competence Center for Ovarian Cancer, Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität Zu Berlin, and Berlin Institute of Health, 10117, Berlin, Germany
| | - Christine Sers
- Institute of Pathology, Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt Universität Zu Berlin, CCM, Charitéplatz 1, 10117, Berlin, Germany
| | | | - Jalid Sehouli
- Tumorbank Ovarian Cancer Network, Charité, Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität Zu Berlin, and Berlin Institute of Health, 10117, Berlin, Germany
- Department of Gynecology, European Competence Center for Ovarian Cancer, Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität Zu Berlin, and Berlin Institute of Health, 10117, Berlin, Germany
| | - Ioana Elena Braicu
- Tumorbank Ovarian Cancer Network, Charité, Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität Zu Berlin, and Berlin Institute of Health, 10117, Berlin, Germany
- Department of Gynecology, European Competence Center for Ovarian Cancer, Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität Zu Berlin, and Berlin Institute of Health, 10117, Berlin, Germany
| | - Christina Westhoff
- Institute of Pathology, Philipps-University Marburg, University Hospital Marburg (UKGM), Marburg, Germany
| | - David Horst
- Institute of Pathology, Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt Universität Zu Berlin, CCM, Charitéplatz 1, 10117, Berlin, Germany
| | - Carsten Denkert
- Institute of Pathology, Philipps-University Marburg, University Hospital Marburg (UKGM), Marburg, Germany
| | | | - Eliane T Taube
- Institute of Pathology, Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt Universität Zu Berlin, CCM, Charitéplatz 1, 10117, Berlin, Germany.
| |
Collapse
|
19
|
Lukashchuk N, Barnicle A, Adelman CA, Armenia J, Kang J, Barrett JC, Harrington EA. Impact of DNA damage repair alterations on prostate cancer progression and metastasis. Front Oncol 2023; 13:1162644. [PMID: 37434977 PMCID: PMC10331135 DOI: 10.3389/fonc.2023.1162644] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 06/01/2023] [Indexed: 07/13/2023] Open
Abstract
Prostate cancer is among the most common diseases worldwide. Despite recent progress with treatments, patients with advanced prostate cancer have poor outcomes and there is a high unmet need in this population. Understanding molecular determinants underlying prostate cancer and the aggressive phenotype of disease can help with design of better clinical trials and improve treatments for these patients. One of the pathways often altered in advanced prostate cancer is DNA damage response (DDR), including alterations in BRCA1/2 and other homologous recombination repair (HRR) genes. Alterations in the DDR pathway are particularly prevalent in metastatic prostate cancer. In this review, we summarise the prevalence of DDR alterations in primary and advanced prostate cancer and discuss the impact of alterations in the DDR pathway on aggressive disease phenotype, prognosis and the association of germline pathogenic alterations in DDR genes with risk of developing prostate cancer.
Collapse
Affiliation(s)
- Natalia Lukashchuk
- Translational Medicine, Oncology Research and Development (R&D), AstraZeneca, Cambridge, United Kingdom
| | - Alan Barnicle
- Translational Medicine, Oncology Research and Development (R&D), AstraZeneca, Cambridge, United Kingdom
| | - Carrie A. Adelman
- Translational Medicine, Oncology Research and Development (R&D), AstraZeneca, Cambridge, United Kingdom
| | - Joshua Armenia
- Oncology Data Science, Oncology Research and Development (R&D), AstraZeneca, Cambridge, United Kingdom
| | - Jinyu Kang
- Global Medicines Development, Oncology Research and Development (R&D), AstraZeneca, Gaithersburg, MD, United States
| | - J. Carl Barrett
- Translational Medicine, Oncology Research and Development (R&D), AstraZeneca, Waltham, MA, United States
| | - Elizabeth A. Harrington
- Translational Medicine, Oncology Research and Development (R&D), AstraZeneca, Cambridge, United Kingdom
| |
Collapse
|
20
|
Florian K, Benet-Pagès A, Berner D, Teubert A, Eck S, Arnold N, Bauer P, Begemann M, Sturm M, Kleinle S, B. Haack T, Eggermann T. Quality assurance within the context of genome diagnostics (a german perspective). MED GENET-BERLIN 2023; 35:91-104. [PMID: 38840862 PMCID: PMC10842579 DOI: 10.1515/medgen-2023-2028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/07/2024]
Abstract
The rapid and dynamic implementation of Next-Generation Sequencing (NGS)-based assays has revolutionized genetic testing, and in the near future, nearly all molecular alterations of the human genome will be diagnosable via massive parallel sequencing. While this progress will further corroborate the central role of human genetics in the multidisciplinary management of patients with genetic disorders, it must be accompanied by quality assurance measures in order to allow the safe and optimal use of knowledge ascertained from genome diagnostics. To achieve this, several valuable tools and guidelines have been developed to support the quality of genome diagnostics. In this paper, authors with experience in diverse aspects of genomic analysis summarize the current status of quality assurance in genome diagnostics, with the aim of facilitating further standardization and quality improvement in one of the core competencies of the field.
Collapse
Affiliation(s)
- Kraft Florian
- Medizinische Fakultät der RWTH AachenInstitut für Humangenetik und GenommedizinAachenDeutschland
| | - Anna Benet-Pagès
- Institut für NeurogenomikHelmholtz Zentrum MünchenNeuherbergDeutschland
| | | | | | | | - Norbert Arnold
- Universitätsklinikum Schleswig-HolsteinZentrum für familiären Brust- und Eierstockkrebs; Klinik für Gynäkologie und GeburtshilfeKielDeutschland
| | | | - Matthias Begemann
- Medizinische Fakultät der RWTH AachenInstitut für Humangenetik und GenommedizinAachenDeutschland
| | - Marc Sturm
- Universität TübingenInstitut für Medizinische Genetik und Angewandte GenomikTübingenDeutschland
| | | | - Tobias B. Haack
- Universität TübingenInstitut für Medizinische Genetik und Angewandte GenomikTübingenDeutschland
| | - Thomas Eggermann
- Medizinische Fakultät der RWTH AachenInstitut für Humangenetik und GenommedizinPauwelsstr. 3052074AachenDeutschland
| |
Collapse
|
21
|
Rădoi VE, Țurcan M, Maioru OV, Dan A, Bohîlțea LC, Dumitrescu EA, Gheorghe AS, Stănculeanu DL, Thodi G, Loukas YL, Săbău ID. Homologous Recombination Deficiency Score Determined by Genomic Instability in a Romanian Cohort. Diagnostics (Basel) 2023; 13:1896. [PMID: 37296748 PMCID: PMC10252278 DOI: 10.3390/diagnostics13111896] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 05/02/2023] [Accepted: 05/19/2023] [Indexed: 06/12/2023] Open
Abstract
The Homologous Recombination Deficiency (HRD) Score, determined by evaluating genomic instability through the assessment of loss of heterozygosity (LOH), telomeric allelic imbalance (TAI), and large-scale state transitions (LST), serves as a crucial biomarker for identifying patients who might benefit from targeted therapies, such as PARP inhibitors (PARPi). This study aimed to investigate the efficacy of HRD testing in high-grade serous ovarian carcinoma, tubal, and peritoneal cancer patients who are negative for somatic BRCA1 and BRCA2 mutations and to evaluate the impact of HRD status on Bevacizumab and PARPi therapy response. A cohort of 100 Romanian female patients, aged 42-77, was initially selected. Among them, 30 patients had unsuitable samples for HRD testing due to insufficient tumor content or DNA integrity. Using the OncoScan C.N.V. platform, HRD testing was successfully performed on the remaining 70 patients, with 20 testing negative and 50 testing positive for HRD. Among the HRD-positive patients, 35 were eligible for and benefited from PARPi maintenance therapy, resulting in a median progression-free survival (PFS) increase from 4 months to 8.2 months. Our findings support the importance of HRD testing in ovarian cancer patients, demonstrating the potential therapeutic advantage of PARPi therapy in HRD-positive patients without somatic BRCA1/2 mutations.
Collapse
Affiliation(s)
- Viorica-Elena Rădoi
- Department of Medical Genetics, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (V.-E.R.); (O.V.M.); (A.D.); (L.C.B.); (I.-D.S.)
- “Alessandrescu-Rusescu” National Institute for Maternal and Child Health, 20382 Bucharest, Romania
- Independent Researcher, 010987 Bucharest, Romania
- Sanador, 011026 Bucharest, Romania
| | - Mihaela Țurcan
- Department of Medical Genetics, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (V.-E.R.); (O.V.M.); (A.D.); (L.C.B.); (I.-D.S.)
- Independent Researcher, 010987 Bucharest, Romania
| | - Ovidiu Virgil Maioru
- Department of Medical Genetics, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (V.-E.R.); (O.V.M.); (A.D.); (L.C.B.); (I.-D.S.)
| | - Andra Dan
- Department of Medical Genetics, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (V.-E.R.); (O.V.M.); (A.D.); (L.C.B.); (I.-D.S.)
| | - Laurentiu Camil Bohîlțea
- Department of Medical Genetics, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (V.-E.R.); (O.V.M.); (A.D.); (L.C.B.); (I.-D.S.)
- “Alessandrescu-Rusescu” National Institute for Maternal and Child Health, 20382 Bucharest, Romania
| | - Elena Adriana Dumitrescu
- Department of Oncology, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (E.A.D.); (D.L.S.)
| | - Adelina Silvana Gheorghe
- Department of Oncology, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (E.A.D.); (D.L.S.)
- Department of Medical Oncology I, Institute of Oncology “Prof. Dr. Al. Trestioreanu” Bucharest, 022328 Bucharest, Romania
| | - Dana Lucia Stănculeanu
- Department of Oncology, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (E.A.D.); (D.L.S.)
- Department of Medical Oncology I, Institute of Oncology “Prof. Dr. Al. Trestioreanu” Bucharest, 022328 Bucharest, Romania
| | - Georgia Thodi
- Neoscreen Diagnostic Laboratory, Voreiou Ipeirou, 15235 Athens, Greece;
| | - Yannis L. Loukas
- School of Pharmacy, University of Athens, Panepistimiolopis, 15771 Zografou, Greece;
| | - Ileana-Delia Săbău
- Department of Medical Genetics, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (V.-E.R.); (O.V.M.); (A.D.); (L.C.B.); (I.-D.S.)
- Independent Researcher, 010987 Bucharest, Romania
| |
Collapse
|
22
|
Azzollini J, Agnelli L, Conca E, Torelli T, Busico A, Capone I, Angelini M, Tamborini E, Perrone F, Vingiani A, Lorenzini D, Peissel B, Pruneri G, Manoukian S. Prevalence of BRCA homopolymeric indels in an ION Torrent-based tumour-to-germline testing workflow in high-grade ovarian carcinoma. Sci Rep 2023; 13:7781. [PMID: 37179432 PMCID: PMC10182972 DOI: 10.1038/s41598-023-33857-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Accepted: 04/20/2023] [Indexed: 05/15/2023] Open
Abstract
Tumour DNA sequencing is essential for precision medicine since it guides therapeutic decisions but also fosters the identification of patients who may benefit from germline testing. Notwithstanding, the tumour-to-germline testing workflow presents a few caveats. The low sensitivity for indels at loci with sequences of identical bases (homopolymers) of ion semiconductor-based sequencing techniques represents a well-known limitation, but the prevalence of indels overlooked by these techniques in high-risk populations has not been investigated. In our study, we addressed this issue at the homopolymeric regions of BRCA1/2 in a retrospectively selected cohort of 157 patients affected with high-grade ovarian cancer and negative at tumour testing by ION Torrent sequencing. Variant allele frequency (VAF) of indels at each of the 29 investigated homopolymers was systematically revised with the IGV software. Thresholds to discriminate putative germline variants were defined by scaling the VAF to a normal distribution and calculating the outliers that exceeded the mean + 3 median-adjusted deviations of a control population. Sanger sequencing of the outliers confirmed the occurrence of only one of the five putative indels in both tumour and blood from a patient with a family history of breast cancer. Our results indicated that the prevalence of homopolymeric indels overlooked by ion semiconductor techniques is seemingly low. A careful evaluation of clinical and family history data would further help minimise this technique-bound limitation, highlighting cases in which a deeper look at these regions would be recommended.
Collapse
Affiliation(s)
- Jacopo Azzollini
- Unit of Medical Genetics, Department of Medical Oncology and Hematology, Fondazione IRCCS Istituto Nazionale dei Tumori, via Venezian 1, 20133, Milan, Italy
| | - Luca Agnelli
- Department of Advanced Diagnostics, Fondazione IRCCS Istituto Nazionale dei Tumori, via Venezian 1, 20133, Milan, Italy
- Medical Oncology 1 Department, Fondazione IRCCS Istituto Nazionale dei Tumori, via Venezian 1, 20133, Milan, Italy
| | - Elena Conca
- Department of Advanced Diagnostics, Fondazione IRCCS Istituto Nazionale dei Tumori, via Venezian 1, 20133, Milan, Italy
| | - Tommaso Torelli
- Department of Advanced Diagnostics, Fondazione IRCCS Istituto Nazionale dei Tumori, via Venezian 1, 20133, Milan, Italy
- Medical Oncology 1 Department, Fondazione IRCCS Istituto Nazionale dei Tumori, via Venezian 1, 20133, Milan, Italy
| | - Adele Busico
- Department of Advanced Diagnostics, Fondazione IRCCS Istituto Nazionale dei Tumori, via Venezian 1, 20133, Milan, Italy
| | - Iolanda Capone
- Department of Advanced Diagnostics, Fondazione IRCCS Istituto Nazionale dei Tumori, via Venezian 1, 20133, Milan, Italy
| | - Marta Angelini
- Department of Advanced Diagnostics, Fondazione IRCCS Istituto Nazionale dei Tumori, via Venezian 1, 20133, Milan, Italy
| | - Elena Tamborini
- Department of Advanced Diagnostics, Fondazione IRCCS Istituto Nazionale dei Tumori, via Venezian 1, 20133, Milan, Italy
| | - Federica Perrone
- Department of Advanced Diagnostics, Fondazione IRCCS Istituto Nazionale dei Tumori, via Venezian 1, 20133, Milan, Italy
| | - Andrea Vingiani
- Department of Advanced Diagnostics, Fondazione IRCCS Istituto Nazionale dei Tumori, via Venezian 1, 20133, Milan, Italy
- Oncology and Hemato-Oncology Department, University of Milan, Milan, Italy
| | - Daniele Lorenzini
- Department of Advanced Diagnostics, Fondazione IRCCS Istituto Nazionale dei Tumori, via Venezian 1, 20133, Milan, Italy
| | - Bernard Peissel
- Unit of Medical Genetics, Department of Medical Oncology and Hematology, Fondazione IRCCS Istituto Nazionale dei Tumori, via Venezian 1, 20133, Milan, Italy
| | - Giancarlo Pruneri
- Department of Advanced Diagnostics, Fondazione IRCCS Istituto Nazionale dei Tumori, via Venezian 1, 20133, Milan, Italy
- Oncology and Hemato-Oncology Department, University of Milan, Milan, Italy
| | - Siranoush Manoukian
- Unit of Medical Genetics, Department of Medical Oncology and Hematology, Fondazione IRCCS Istituto Nazionale dei Tumori, via Venezian 1, 20133, Milan, Italy.
| |
Collapse
|