51
|
Maurer LM, Daley JD, Mukherjee E, Venier RE, Julian CM, Bailey NG, Jacobs MF, Kumar-Sinha C, Raphael H, Periyapatna N, Weiss K, Janeway KA, Mody R, Lucas PC, McAllister-Lucas LM, Bailey KM. BRCA1-associated RING domain-1 (BARD1) loss and GBP1 expression enhance sensitivity to DNA damage in Ewing sarcoma. CANCER RESEARCH COMMUNICATIONS 2022; 2:220-232. [PMID: 36187937 PMCID: PMC9524505 DOI: 10.1158/2767-9764.crc-21-0047] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Ewing sarcoma is a fusion oncoprotein-driven primary bone tumor. A subset of patients (~10%) with Ewing sarcoma are known to harbor germline variants in a growing number of genes involved in DNA damage repair. We recently reported our discovery of a germline mutation in the DNA damage repair protein BARD1 (BRCA1-associated RING domain-1) in a patient with Ewing sarcoma. BARD1 is recruited to the site of DNA double stranded breaks via the poly(ADP-ribose) polymerase (PARP) protein and plays a critical role in DNA damage response pathways including homologous recombination. We thus questioned the impact of BARD1 loss on Ewing cell sensitivity to DNA damage and the Ewing sarcoma transcriptome. We demonstrate that PSaRC318 cells, a novel patient-derived cell line harboring a pathogenic BARD1 variant, are sensitive to PARP inhibition and by testing the effect of BARD1 depletion in additional Ewing sarcoma cell lines, we confirm that BARD1 loss enhances cell sensitivity to PARP inhibition plus radiation. Additionally, RNA-seq analysis revealed that loss of BARD1 results in the upregulation of GBP1 (guanylate-binding protein 1), a protein whose expression is associated with variable response to therapy depending on the adult carcinoma subtype examined. Here, we demonstrate that GBP1 contributes to the enhanced sensitivity of BARD1 deficient Ewing cells to DNA damage. Together, our findings demonstrate the impact of loss-of function mutations in DNA damage repair genes, such as BARD1, on Ewing sarcoma treatment response.
Collapse
Affiliation(s)
- Lisa M Maurer
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Jessica D Daley
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Elina Mukherjee
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Rosemarie E Venier
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA.,Department of Human Genetics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA
| | - Claire M Julian
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Nathanael G Bailey
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Michelle F Jacobs
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI
| | | | - Haley Raphael
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Nivitha Periyapatna
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Kurt Weiss
- Department of Orthopedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Katherine A Janeway
- Pediatric Oncology, Dana-Farber / Boston Children's Cancer and Blood Disorders Center, Boston, MA
| | - Rajen Mody
- Department of Pediatrics, University of Michigan Medical School, Ann Arbor, MI
| | - Peter C Lucas
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | | | - Kelly M Bailey
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA
| |
Collapse
|
52
|
Mas-Ponte D, McCullough M, Supek F. Spectrum of DNA mismatch repair failures viewed through the lens of cancer genomics and implications for therapy. Clin Sci (Lond) 2022; 136:383-404. [PMID: 35274136 PMCID: PMC8919091 DOI: 10.1042/cs20210682] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 02/02/2022] [Accepted: 02/28/2022] [Indexed: 12/15/2022]
Abstract
Genome sequencing can be used to detect DNA repair failures in tumors and learn about underlying mechanisms. Here, we synthesize findings from genomic studies that examined deficiencies of the DNA mismatch repair (MMR) pathway. The impairment of MMR results in genome-wide hypermutation and in the 'microsatellite instability' (MSI) phenotype-occurrence of indel mutations at short tandem repeat (microsatellite) loci. The MSI status of tumors was traditionally assessed by molecular testing of a selected set of MS loci or by measuring MMR protein expression levels. Today, genomic data can provide a more complete picture of the consequences on genomic instability. Multiple computational studies examined somatic mutation distributions that result from failed DNA repair pathways in tumors. These include analyzing the commonly studied trinucleotide mutational spectra of single-nucleotide variants (SNVs), as well as of other features such as indels, structural variants, mutation clusters and regional mutation rate redistribution. The identified mutation patterns can be used to rigorously measure prevalence of MMR failures across cancer types, and potentially to subcategorize the MMR deficiencies. Diverse data sources, genomic and pre-genomic, from human and from experimental models, suggest there are different ways in which MMR can fail, and/or that the cell-type or genetic background may result in different types of MMR mutational patterns. The spectrum of MMR failures may direct cancer evolution, generating particular sets of driver mutations. Moreover, MMR affects outcomes of therapy by DNA damaging drugs, antimetabolites, nonsense-mediated mRNA decay (NMD) inhibitors, and immunotherapy by promoting either resistance or sensitivity, depending on the type of therapy.
Collapse
Affiliation(s)
- David Mas-Ponte
- Genome Data Science, Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute for Science and Technology, Baldiri Reixac 10, Barcelona 08028, Spain
| | - Marcel McCullough
- Genome Data Science, Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute for Science and Technology, Baldiri Reixac 10, Barcelona 08028, Spain
| | - Fran Supek
- Genome Data Science, Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute for Science and Technology, Baldiri Reixac 10, Barcelona 08028, Spain
- Catalan Institution for Research and Advanced Studies (ICREA), Pg Lluís Companys, 23, Barcelona 08010, Spain
| |
Collapse
|
53
|
Komura D, Kawabe A, Fukuta K, Sano K, Umezaki T, Koda H, Suzuki R, Tominaga K, Ochi M, Konishi H, Masakado F, Saito N, Sato Y, Onoyama T, Nishida S, Furuya G, Katoh H, Yamashita H, Kakimi K, Seto Y, Ushiku T, Fukayama M, Ishikawa S. Universal encoding of pan-cancer histology by deep texture representations. Cell Rep 2022; 38:110424. [PMID: 35235802 DOI: 10.1016/j.celrep.2022.110424] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 07/11/2021] [Accepted: 02/01/2022] [Indexed: 02/07/2023] Open
Abstract
Cancer histological images contain rich biological and clinical information, but quantitative representation can be problematic and has prevented the direct comparison and accumulation of large-scale datasets. Here, we show successful universal encoding of cancer histology by deep texture representations (DTRs) produced by a bilinear convolutional neural network. DTR-based, unsupervised histological profiling, which captures the morphological diversity, is applied to cancer biopsies and reveals relationships between histologic characteristics and the response to immune checkpoint inhibitors (ICIs). Content-based image retrieval based on DTRs enables the quick retrieval of histologically similar images using The Cancer Genome Atlas (TCGA) dataset. Furthermore, via comprehensive comparisons with driver and clinically actionable gene mutations, we successfully predict 309 combinations of genomic features and cancer types from hematoxylin-and-eosin-stained images. With its mounting capabilities on accessible devices, such as smartphones, universal encoding for cancer histology has a strong impact on global equalization for cancer diagnosis and therapies.
Collapse
Affiliation(s)
- Daisuke Komura
- Department of Preventive Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 1130033, Japan
| | - Akihiro Kawabe
- Department of Preventive Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 1130033, Japan; Department of Pathology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 1130033, Japan
| | - Keisuke Fukuta
- Department of Genomic Pathology, Medical Research Institute, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 1138510, Japan
| | - Kyohei Sano
- Department of Preventive Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 1130033, Japan
| | - Toshikazu Umezaki
- Department of Preventive Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 1130033, Japan
| | - Hirotomo Koda
- Department of Preventive Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 1130033, Japan; Department of Pathology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 1130033, Japan
| | - Ryohei Suzuki
- Department of Preventive Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 1130033, Japan
| | - Ken Tominaga
- Department of Genomic Pathology, Medical Research Institute, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 1138510, Japan
| | - Mieko Ochi
- Department of Preventive Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 1130033, Japan
| | - Hiroki Konishi
- Department of Preventive Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 1130033, Japan
| | - Fumiya Masakado
- Department of Preventive Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 1130033, Japan
| | - Noriyuki Saito
- Department of Gastrointestinal Surgery, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 1130033, Japan
| | - Yasuyoshi Sato
- Department of Gastrointestinal Surgery, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 1130033, Japan
| | - Takumi Onoyama
- Department of Preventive Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 1130033, Japan; Division of Gastroenterology and Nephrology, Department of Multidisciplinary Internal Medicine, School of Medicine, Faculty of Medicine, Tottori University, 36-1 Nishicho, Yonago, Tottori 683-8504, Japan
| | - Shu Nishida
- Department of Preventive Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 1130033, Japan; Department of Pathology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 1130033, Japan
| | - Genta Furuya
- Department of Preventive Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 1130033, Japan; Department of Pathology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 1130033, Japan
| | - Hiroto Katoh
- Department of Preventive Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 1130033, Japan
| | - Hiroharu Yamashita
- Division of Digestive Surgery, Department of Surgery, Nihon University School of Medicine, 1-6 Kanda-Surugadai, Chiyoda-ku Tokyo 101-8309, Japan
| | - Kazuhiro Kakimi
- Department of Immunotherapeutics, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo 1130033, Japan
| | - Yasuyuki Seto
- Department of Gastrointestinal Surgery, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 1130033, Japan
| | - Tetsuo Ushiku
- Department of Pathology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 1130033, Japan
| | - Masashi Fukayama
- Department of Pathology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 1130033, Japan
| | - Shumpei Ishikawa
- Department of Preventive Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 1130033, Japan.
| |
Collapse
|
54
|
Toward More Comprehensive Homologous Recombination Deficiency Assays in Ovarian Cancer, Part 1: Technical Considerations. Cancers (Basel) 2022; 14:cancers14051132. [PMID: 35267439 PMCID: PMC8909526 DOI: 10.3390/cancers14051132] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 02/19/2022] [Accepted: 02/22/2022] [Indexed: 02/05/2023] Open
Abstract
Simple Summary High-grade serous ovarian cancer (HGSOC) is the most frequent and lethal form of ovarian cancer and is associated with homologous recombination deficiency (HRD) in 50% of cases. This specific alteration is associated with sensitivity to PARP inhibitors (PARPis). Despite vast prognostic improvements due to PARPis, current molecular assays assessing HRD status suffer from several limitations, and there is an urgent need for a more accurate evaluation. In these companion reviews (Part 1: Technical considerations; Part 2: Medical perspectives), we develop an integrative review to provide physicians and researchers involved in HGSOC management with a holistic perspective, from translational research to clinical applications. Abstract High-grade serous ovarian cancer (HGSOC), the most frequent and lethal form of ovarian cancer, exhibits homologous recombination deficiency (HRD) in 50% of cases. In addition to mutations in BRCA1 and BRCA2, which are the best known thus far, defects can also be caused by diverse alterations to homologous recombination-related genes or epigenetic patterns. HRD leads to genomic instability (genomic scars) and is associated with PARP inhibitor (PARPi) sensitivity. HRD is currently assessed through BRCA1/2 analysis, which produces a genomic instability score (GIS). However, despite substantial clinical achievements, FDA-approved companion diagnostics (CDx) based on GISs have important limitations. Indeed, despite the use of GIS in clinical practice, the relevance of such assays remains controversial. Although international guidelines include companion diagnostics as part of HGSOC frontline management, they also underscore the need for more powerful and alternative approaches for assessing patient eligibility to PARP inhibitors. In these companion reviews, we review and present evidence to date regarding HRD definitions, achievements and limitations in HGSOC. Part 1 is dedicated to technical considerations and proposed perspectives that could lead to a more comprehensive and dynamic assessment of HR, while Part 2 provides a more integrated approach for clinicians.
Collapse
|
55
|
Quesada S, Fabbro M, Solassol J. Toward More Comprehensive Homologous Recombination Deficiency Assays in Ovarian Cancer Part 2: Medical Perspectives. Cancers (Basel) 2022; 14:cancers14041098. [PMID: 35205846 PMCID: PMC8870335 DOI: 10.3390/cancers14041098] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 02/19/2022] [Accepted: 02/20/2022] [Indexed: 02/07/2023] Open
Abstract
Simple Summary High-grade serous ovarian cancer (HGSOC—the most frequent and aggressive form of ovarian cancer) represents an important challenge for clinicians. Half of HGSOC cases exhibit homologous recombination deficiency (HRD), mainly through alterations in BRCA1 and BRCA2. This leads to sensitivity to PARP inhibitors, a novel class of breakthrough molecules that improved HGSOC prognoses. To date, three companion diagnostic assays have received FDA approval for the evaluation of HRD status, but their use remains controversial. In this companion review (Part 1: Technical considerations; Part 2: Medical perspectives), we develop an integrative perspective, from translational research to clinical application, that could help physicians and researchers manage HGSOC. Abstract High-grade serous ovarian cancer (HGSOC) is the most frequent and aggressive form of ovarian cancer, representing an important challenge for clinicians. Half of HGSOC cases have homologous recombination deficiency (HRD), which has specific causes (mainly alterations in BRCA1/2, but also other alterations encompassed by the BRCAness concept) and consequences, both at molecular (e.g., genomic instability) and clinical (e.g., sensitivity to PARP inhibitor) levels. Based on its prevalence and clinical impact, HRD status merits investigation. To date, three PARP inhibitors have received FDA/EMA approval. For some approvals, the presence of specific molecular alterations is required. Three companion diagnostic (CDx) assays based on distinct technical and medical considerations have received FDA approval to date. However, their use remains controversial due to their technical and medical limitations. In this companion and integrated review, we take a “bench-to-bedside” perspective on HRD definition and evaluation in the context of HGSOC. Part 1 of the review adopts a molecular perspective regarding technical considerations and the development of CDx. Part 2 focuses on the clinical impact of HRD evaluation, primarily through currently validated CDx and prescription of PARP inhibitors, outlining achievements, limitations and medical perspectives.
Collapse
Affiliation(s)
- Stanislas Quesada
- Medical Oncology Department, Institut Régional du Cancer de Montpellier (ICM), 34298 Montpellier, France;
- Faculty of Medicine, University of Montpellier, 34090 Montpellier, France;
- Correspondence:
| | - Michel Fabbro
- Medical Oncology Department, Institut Régional du Cancer de Montpellier (ICM), 34298 Montpellier, France;
- Montpellier Research Cancer Institute (IRCM), Institut National de la Santé et de la Recherche Médicale (INSERM) U1194, University of Montpellier, 34298 Montpellier, France
| | - Jérôme Solassol
- Faculty of Medicine, University of Montpellier, 34090 Montpellier, France;
- Montpellier Research Cancer Institute (IRCM), Institut National de la Santé et de la Recherche Médicale (INSERM) U1194, University of Montpellier, 34298 Montpellier, France
- Department of Pathology and Onco-Biology, Centre Hospitalier Universitaire (CHU) Montpellier, 34295 Montpellier, France
| |
Collapse
|
56
|
Ramsden DA, Carvajal-Garcia J, Gupta GP. Mechanism, cellular functions and cancer roles of polymerase-theta-mediated DNA end joining. Nat Rev Mol Cell Biol 2022; 23:125-140. [PMID: 34522048 DOI: 10.1038/s41580-021-00405-2] [Citation(s) in RCA: 124] [Impact Index Per Article: 41.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/21/2021] [Indexed: 02/08/2023]
Abstract
Cellular pathways that repair chromosomal double-strand breaks (DSBs) have pivotal roles in cell growth, development and cancer. These DSB repair pathways have been the target of intensive investigation, but one pathway - alternative end joining (a-EJ) - has long resisted elucidation. In this Review, we highlight recent progress in our understanding of a-EJ, especially the assignment of DNA polymerase theta (Polθ) as the predominant mediator of a-EJ in most eukaryotes, and discuss a potential molecular mechanism by which Polθ-mediated end joining (TMEJ) occurs. We address possible cellular functions of TMEJ in resolving DSBs that are refractory to repair by non-homologous end joining (NHEJ), DSBs generated following replication fork collapse and DSBs present owing to stalling of repair by homologous recombination. We also discuss how these context-dependent cellular roles explain how TMEJ can both protect against and cause genome instability, and the emerging potential of Polθ as a therapeutic target in cancer.
Collapse
Affiliation(s)
- Dale A Ramsden
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
- Curriculum in Genetics and Molecular Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| | - Juan Carvajal-Garcia
- Curriculum in Genetics and Molecular Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Gaorav P Gupta
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
- Curriculum in Genetics and Molecular Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
- Department of Radiation Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| |
Collapse
|
57
|
Chapman BV, Liu D, Shen Y, Olamigoke OO, Lakomy DS, Barrera AMG, Stecklein SR, Sawakuchi GO, Bright SJ, Bedrosian I, Litton JK, Smith BD, Woodward WA, Perkins GH, Hoffman KE, Stauder MC, Strom EA, Arun BK, Shaitelman SF. Outcomes After Breast Radiation Therapy in a Diverse Patient Cohort With a Germline BRCA1/2 Mutation. Int J Radiat Oncol Biol Phys 2022; 112:426-436. [PMID: 34610390 PMCID: PMC9330175 DOI: 10.1016/j.ijrobp.2021.09.033] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 09/20/2021] [Accepted: 09/25/2021] [Indexed: 02/03/2023]
Abstract
PURPOSE BRCA1/2 pathogenic variant (PV) mutations confer radiation sensitivity preclinically, but there are limited data regarding breast cancer outcomes after radiation therapy (RT) among patients with documented BRCA1/2 PV mutations versus no PV mutations. METHODS AND MATERIALS This retrospective cohort study included women with clinical stage I-III breast cancer who received definitive surgery and RT and underwent BRCA1/2 genetic evaluation at the The University of Texas MD Anderson Cancer Center. Rates of locoregional recurrence (LRR), disease-specific death (DSD), toxicities, and second cancers were compared by BRCA1/2 PV status. RESULTS Of the 2213 women who underwent BRCA1/2 testing, 63% self-reported their race as White, 13.6% as Black/African American, 17.6% as Hispanic, and 5.8% as Asian/American Indian/Alaska Native; 124 had BRCA1 and 100 had BRCA2 mutations; and 1394 (63%) received regional nodal RT. The median follow-up time for all patients was 7.4 years (95% confidence interval [CI], 7.1-7.7 years). No differences were found between the groups with and without BRCA1/2 PV mutations in 10-year cumulative incidences of LRR (with mutations: 11.6% [95% CI, 7.0%-17.6%]; without mutations: 6.6% [95% CI, 5.3%-8.0%]; P = .466) and DSD (with mutations: 12.3% [95% CI, 8.0%-17.7%]; without mutations: 13.8% [95% CI, 12.0%-15.8%]; P = .716). On multivariable analysis, BRCA1/2 status was not associated with LRR or DSD, but Black/African American patients (P = .036) and Asians/American Indians/Alaska Native patients (P = .002) were at higher risk of LRR compared with White patients, and Black/African American patients were at higher risk of DSD versus White patients (P = .004). No in-field, nonbreast second cancers were observed in the BRCA1/2 PV group. Rates of acute and late grade ≥3 radiation-related toxicity in the BCRA1/2 PV group were 5.4% (n = 12) and 0.4% (n = 1), respectively. CONCLUSIONS Oncologic outcomes in a diverse cohort of patients with breast cancer who had a germline BRCA1/2 PV mutation and were treated with RT were similar to those of patients with no mutation, supporting the use of RT according to standard indications in patients with a germline BRCA1/2 PV mutation.
Collapse
Affiliation(s)
- Bhavana V. Chapman
- Department of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Diane Liu
- Department of Biostatistics, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Yu Shen
- Department of Biostatistics, University of Texas MD Anderson Cancer Center, Houston, Texas
| | | | - David S. Lakomy
- Department of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Angelica M. Gutierrez Barrera
- Department of Breast Medical Oncology and Clinical Cancer Genetics, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Shane R. Stecklein
- Department of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Gabriel O. Sawakuchi
- Department of Radiation Physics, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Scott J. Bright
- Department of Radiation Physics, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Isabelle Bedrosian
- Department of Breast Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jennifer K. Litton
- Department of Breast Medical Oncology and Clinical Cancer Genetics, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Benjamin D. Smith
- Department of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Wendy A. Woodward
- Department of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - George H. Perkins
- Department of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Karen E. Hoffman
- Department of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Michael C. Stauder
- Department of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Eric A. Strom
- Department of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Banu K. Arun
- Department of Breast Medical Oncology and Clinical Cancer Genetics, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Simona F. Shaitelman
- Department of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
58
|
El Jabbour T, Misyura M, Cowzer D, Zimmermann M, Rimkunas V, Marra A, Derakhshan F, Selenica P, Parilla M, Setton JS, Ceyhan-Birsoy O, Kemel Y, Catchings A, Ranganathan M, Ku GY, Janjigian YY, Zinda M, Koehler M, Stadler Z, Shia J, Reis-Filho JS, Mandelker D. ATM Germline Mutated Gastroesophageal Junction Adenocarcinomas: Clinical Descriptors, Molecular Characteristics and Potential Therapeutic Implications. J Natl Cancer Inst 2022; 114:761-770. [PMID: 35078243 PMCID: PMC9086803 DOI: 10.1093/jnci/djac024] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 11/22/2021] [Accepted: 01/19/2022] [Indexed: 11/14/2022] Open
Abstract
Background Gastroesophageal junction (GEJ) adenocarcinoma is a rare cancer associated with poor prognosis. The genetic factors conferring predisposition to GEJ adenocarcinoma have yet to be identified. Methods We analyzed germline testing results from 23 381 cancer patients undergoing tumor-normal sequencing, of which 312 individuals had GEJ adenocarcinoma. Genomic profiles and clinico-pathologic features were analyzed for the GEJ adenocarcinomas. Silencing of ATM and ATR was performed using validated short-interfering RNA species in GEJ, esophageal, and gastric adenocarcinoma cell lines. All statistical tests were 2-sided. Results Pathogenic or likely pathogenic ATM variants were identified in 18 of 312 patients (5.8%), and bi-allelic inactivation of ATM through loss of heterozygosity of the wild-type allele was detected in all (16 of 16) samples with sufficient tumor content. Germline ATM-mutated GEJ adenocarcinomas largely lacked somatic mutations in TP53, were more likely to harbor MDM2 amplification, and harbored statistically significantly fewer somatic single nucleotide variants (2.0 mutations/Mb vs 7.9 mutations/Mb; P < .001). A statistically significantly higher proportion of germline ATM-mutated than ATM–wild-type GEJ adenocarcinoma patients underwent a curative resection (10 [100%] vs 92 [86.8%], P = .04; Fisher’s exact test.), A synthetic lethal interaction between short-interfering RNA silencing of ATM and ATR was observed in the models analyzed. Conclusions Our results indicate that germline pathogenic variants in ATM drive oncogenesis in GEJ adenocarcinoma and might result in a distinct clinical phenotype. Given the high prevalence of germline ATM-mutated GEJ adenocarcinomas, genetic testing for individuals with GEJ adenocarcinomas may be considered to better inform prognostication, treatment decisions, and future cancer risk.
Collapse
Affiliation(s)
- Tony El Jabbour
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Maksym Misyura
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Darren Cowzer
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | | | - Antonio Marra
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Fatemeh Derakhshan
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Pier Selenica
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Megan Parilla
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jeremy S Setton
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Ozge Ceyhan-Birsoy
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Yelena Kemel
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Amanda Catchings
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Megha Ranganathan
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Geoffrey Y Ku
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Yelena Y Janjigian
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | | | - Zsofia Stadler
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jinru Shia
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jorge S Reis-Filho
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Diana Mandelker
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| |
Collapse
|
59
|
Abstract
Triple-negative breast cancer (TNBC) encompasses a heterogeneous group of fundamentally different diseases with different histologic, genomic, and immunologic profiles, which are aggregated under this term because of their lack of estrogen receptor, progesterone receptor, and human epidermal growth factor receptor 2 expression. Massively parallel sequencing and other omics technologies have demonstrated the level of heterogeneity in TNBCs and shed light into the pathogenesis of this therapeutically challenging entity in breast cancer. In this review, we discuss the histologic and molecular classifications of TNBC, the genomic alterations these different tumor types harbor, and the potential impact of these alterations on the pathogenesis of these tumors. We also explore the role of the tumor microenvironment in the biology of TNBCs and its potential impact on therapeutic response. Dissecting the biology and understanding the therapeutic dependencies of each TNBC subtype will be essential to delivering on the promise of precision medicine for patients with triple-negative disease.
Collapse
Affiliation(s)
- Fatemeh Derakhshan
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY 10021, USA;
| | - Jorge S Reis-Filho
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY 10021, USA;
| |
Collapse
|
60
|
Value of the loss of heterozygosity to BRCA1 variant classification. NPJ Breast Cancer 2022; 8:9. [PMID: 35039532 PMCID: PMC8764043 DOI: 10.1038/s41523-021-00361-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 09/21/2021] [Indexed: 11/25/2022] Open
Abstract
At least 10% of the BRCA1/2 tests identify variants of uncertain significance (VUS) while the distinction between pathogenic variants (PV) and benign variants (BV) remains particularly challenging. As a typical tumor suppressor gene, the inactivation of the second wild-type (WT) BRCA1 allele is expected to trigger cancer initiation. Loss of heterozygosity (LOH) of the WT allele is the most frequent mechanism for the BRCA1 biallelic inactivation. To evaluate if LOH can be an effective predictor of BRCA1 variant pathogenicity, we carried out LOH analysis on DNA extracted from 90 breast and seven ovary tumors diagnosed in 27 benign and 55 pathogenic variant carriers. Further analyses were conducted in tumors with PVs yet without loss of the WT allele: BRCA1 promoter hypermethylation, next-generation sequencing (NGS) of BRCA1/2, and BRCAness score. Ninety-seven tumor samples were analyzed from 26 different BRCA1 variants. A relatively stable pattern of LOH (65.4%) of WT allele for PV tumors was observed, while the allelic balance (63%) or loss of variant allele (15%) was generally seen for carriers of BV. LOH data is a useful complementary argument for BRCA1 variant classification.
Collapse
|
61
|
Sculco M, La Vecchia M, Aspesi A, Pinton G, Clavenna MG, Casalone E, Allione A, Grosso F, Libener R, Muzio A, Rena O, Baietto G, Parini S, Boldorini R, Giachino D, Papotti M, Scagliotti GV, Migliore E, Mirabelli D, Moro L, Magnani C, Ferrante D, Matullo G, Dianzani I. Malignant pleural mesothelioma: Germline variants in DNA repair genes may steer tailored treatment. Eur J Cancer 2022; 163:44-54. [PMID: 35032816 DOI: 10.1016/j.ejca.2021.12.023] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 11/25/2021] [Accepted: 12/19/2021] [Indexed: 02/02/2023]
Abstract
INTRODUCTION Malignant pleural mesothelioma (MPM) is a tumour associated with asbestos exposure. Approximately, 10% of patients with MPM carry a germline pathogenic variant (PV), mostly in DNA repair genes, suggesting the occurrence of inherited predispositions. AIM This article aimed to 1) search for new predisposing genes and assess the prevalence of PVs in DNA repair genes, by next-generation sequencing (NGS) analysis of germline DNA from 113 unselected patients with MPM and 2) evaluate whether these patients could be sensitive to tailored treatments. METHODS NGS was performed using a custom panel of 107 cancer-predisposing genes. To investigate the response to selected drugs in conditions of DNA repair insufficiency, we created a three-dimensional-MPM cell model that had a defect in ataxia telangiectasia mutated (ATM), the master regulator of DNA repair. RESULTS We identified PVs in approximately 7% of patients with MPM (8/113) and a new PV in BAP1 in a further patient with familial MPM. Most of these PVs were in genes involved or supposedly involved in DNA repair (BRCA1, BRIP1, CHEK2, SLX4, FLCN and BAP1). In vitro studies showed apoptosis induction in ATM-silenced/inhibited MPM spheroids treated with an enhancer of zeste homologue 2 inhibitor (tazemetostat). CONCLUSIONS Overall these data suggest that patients with MPM and DNA repair insufficiency may benefit from this treatment, which induces synthetic lethality.
Collapse
Affiliation(s)
- Marika Sculco
- Department of Health Sciences, Università del Piemonte Orientale, Novara, Italy
| | - Marta La Vecchia
- Department of Health Sciences, Università del Piemonte Orientale, Novara, Italy
| | - Anna Aspesi
- Department of Health Sciences, Università del Piemonte Orientale, Novara, Italy
| | - Giulia Pinton
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale, Novara, Italy
| | - Michela G Clavenna
- Department of Health Sciences, Università del Piemonte Orientale, Novara, Italy
| | | | | | - Federica Grosso
- Mesothelioma Unit, AO SS. Antonio e Biagio e Cesare Arrigo, Alessandria, Italy
| | - Roberta Libener
- Department of Integrated Activities Research and Innovation, AO SS. Antonio e Biagio e Cesare Arrigo, Alessandria, Italy
| | - Alberto Muzio
- Division of Medical Oncology, Ospedale Santo Spirito, Casale Monferrato (Alessandria), Italy
| | - Ottavio Rena
- Thoracic Surgery Unit, AOU Maggiore della Carità, Novara, Italy
| | - Guido Baietto
- Thoracic Surgery Unit, AOU Maggiore della Carità, Novara, Italy
| | - Sara Parini
- Thoracic Surgery Unit, AOU Maggiore della Carità, Novara, Italy
| | - Renzo Boldorini
- Department of Health Sciences, Section of Pathological Anatomy, Università del Piemonte Orientale, Novara, Italy
| | - Daniela Giachino
- Medical Genetics Unit, Department of Clinical and Biological Sciences, Università di Torino, AOU S. Luigi Gonzaga, Orbassano, (Torino), Italy
| | | | - Giorgio V Scagliotti
- Department of Oncology, Università di Torino, AOU S. Luigi Gonzaga, Orbassano, Torino, Italy
| | - Enrica Migliore
- Unit of Cancer Epidemiology, CPO-Piemonte and Università di Torino, Italy
| | - Dario Mirabelli
- Unit of Cancer Epidemiology, CPO-Piemonte and Università di Torino, Italy; Interdepartmental Center for Studies on Asbestos and Other Toxic Particulates "G. Scansetti", Università di Torino, Italy
| | - Laura Moro
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale, Novara, Italy
| | - Corrado Magnani
- Department of Translational Medicine, Unit of Medical Statistics, Università del Piemonte Orientale and Cancer Epidemiology, CPO Piemonte, Novara, Italy
| | - Daniela Ferrante
- Department of Translational Medicine, Unit of Medical Statistics, Università del Piemonte Orientale and Cancer Epidemiology, CPO Piemonte, Novara, Italy
| | - Giuseppe Matullo
- Department of Medical Sciences, Università di Torino, Italy; Interdepartmental Center for Studies on Asbestos and Other Toxic Particulates "G. Scansetti", Università di Torino, Italy; Medical Genetics Unit, AOU Città Della Salute e della Scienza di Torino, Italy.
| | - Irma Dianzani
- Department of Health Sciences, Università del Piemonte Orientale, Novara, Italy; Interdepartmental Center for Studies on Asbestos and Other Toxic Particulates "G. Scansetti", Università di Torino, Italy.
| |
Collapse
|
62
|
De Pauw A. Les nouvelles évolutions de l’oncogénétique en 2022. PSYCHO-ONCOLOGIE 2022. [DOI: 10.3166/pson-2021-0171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
L’oncogénétique est une spécialité de la génétique médicale en perpétuelle évolution. Mais de grands bouleversements ont eu lieu ces dernières années d’une part avec l’arrivée des PARPi dans le traitement du cancer. Ces thérapies ciblées peuvent être proposées à des patients porteurs d’altérations dans des gènes de prédisposition au cancer. La nécessité d’identifier ces patients, pour les informer des enjeux d’un test constitutionnel, a provoqué une réorganisation des consultations et des laboratoires avec la mise en place de nouveaux circuits, urgents, de consultation et d’analyse. D’autre part, la possibilité de séquençage du génome tumoral va conduire à identifier des patients porteurs de variants pathogènes pouvant être en lien avec la maladie ou non, potentiellement d’origine constitutionnelle. Dans les deux cas, une information prétest des patients reste indispensable pour préparer ceux qui seraient concernés, avec leurs familles, par une prédisposition génétique au cancer.
Collapse
|
63
|
Chen D, Gervai JZ, Póti Á, Németh E, Szeltner Z, Szikriszt B, Gyüre Z, Zámborszky J, Ceccon M, d'Adda di Fagagna F, Szallasi Z, Richardson AL, Szüts D. BRCA1 deficiency specific base substitution mutagenesis is dependent on translesion synthesis and regulated by 53BP1. Nat Commun 2022; 13:226. [PMID: 35017534 PMCID: PMC8752635 DOI: 10.1038/s41467-021-27872-7] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 12/15/2021] [Indexed: 12/25/2022] Open
Abstract
Defects in BRCA1, BRCA2 and other genes of the homology-dependent DNA repair (HR) pathway cause an elevated rate of mutagenesis, eliciting specific mutation patterns including COSMIC signature SBS3. Using genome sequencing of knock-out cell lines we show that Y family translesion synthesis (TLS) polymerases contribute to the spontaneous generation of base substitution and short insertion/deletion mutations in BRCA1 deficient cells, and that TLS on DNA adducts is increased in BRCA1 and BRCA2 mutants. The inactivation of 53BP1 in BRCA1 mutant cells markedly reduces TLS-specific mutagenesis, and rescues the deficiency of template switch-mediated gene conversions in the immunoglobulin V locus of BRCA1 mutant chicken DT40 cells. 53BP1 also promotes TLS in human cellular extracts in vitro. Our results show that HR deficiency-specific mutagenesis is largely caused by TLS, and suggest a function for 53BP1 in regulating the choice between TLS and error-free template switching in replicative DNA damage bypass.
Collapse
Affiliation(s)
- Dan Chen
- Institute of Enzymology, Research Centre for Natural Sciences, Budapest, H-1117, Hungary
| | - Judit Z Gervai
- Institute of Enzymology, Research Centre for Natural Sciences, Budapest, H-1117, Hungary
| | - Ádám Póti
- Institute of Enzymology, Research Centre for Natural Sciences, Budapest, H-1117, Hungary
| | - Eszter Németh
- Institute of Enzymology, Research Centre for Natural Sciences, Budapest, H-1117, Hungary
| | - Zoltán Szeltner
- Institute of Enzymology, Research Centre for Natural Sciences, Budapest, H-1117, Hungary
| | - Bernadett Szikriszt
- Institute of Enzymology, Research Centre for Natural Sciences, Budapest, H-1117, Hungary
| | - Zsolt Gyüre
- Institute of Enzymology, Research Centre for Natural Sciences, Budapest, H-1117, Hungary
- Doctoral School of Molecular Medicine, Semmelweis University, Budapest, H-1085, Hungary
| | - Judit Zámborszky
- Institute of Enzymology, Research Centre for Natural Sciences, Budapest, H-1117, Hungary
| | - Marta Ceccon
- IFOM Foundation-FIRC Institute of Molecular Oncology Foundation, Via Adamello 16, 20139, Milan, Italy
| | - Fabrizio d'Adda di Fagagna
- IFOM Foundation-FIRC Institute of Molecular Oncology Foundation, Via Adamello 16, 20139, Milan, Italy
- Istituto di Genetica Molecolare, Consiglio Nazionale delle Ricerche (IGM-CNR), Via Abbiategrasso 207, 27100, Pavia, Italy
| | - Zoltan Szallasi
- Computational Health Informatics Program (CHIP), Boston Children's Hospital and Harvard Medical School, Boston, MA, 02215, USA
- Danish Cancer Society Research Center, Copenhagen, 2100, Denmark
- SE-NAP, Brain Metastasis Research Group, 2nd Department of Pathology, Semmelweis University, Budapest, H-1092, Hungary
| | | | - Dávid Szüts
- Institute of Enzymology, Research Centre for Natural Sciences, Budapest, H-1117, Hungary.
| |
Collapse
|
64
|
Bonazzi VF, Kondrashova O, Smith D, Nones K, Sengal AT, Ju R, Packer LM, Koufariotis LT, Kazakoff SH, Davidson AL, Ramarao-Milne P, Lakis V, Newell F, Rogers R, Davies C, Nicklin J, Garrett A, Chetty N, Perrin L, Pearson JV, Patch AM, Waddell N, Pollock PM. Patient-derived xenograft models capture genomic heterogeneity in endometrial cancer. Genome Med 2022; 14:3. [PMID: 35012638 PMCID: PMC8751371 DOI: 10.1186/s13073-021-00990-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 10/13/2021] [Indexed: 12/12/2022] Open
Abstract
Background Endometrial cancer (EC) is a major gynecological cancer with increasing incidence. It comprises four molecular subtypes with differing etiology, prognoses, and responses to chemotherapy. In the future, clinical trials testing new single agents or combination therapies will be targeted to the molecular subtype most likely to respond. As pre-clinical models that faithfully represent the molecular subtypes of EC are urgently needed, we sought to develop and characterize a panel of novel EC patient-derived xenograft (PDX) models. Methods Here, we report whole exome or whole genome sequencing of 11 PDX models and their matched primary tumor. Analysis of multiple PDX lineages and passages was performed to study tumor heterogeneity across lineages and/or passages. Based on recent reports of frequent defects in the homologous recombination (HR) pathway in EC, we assessed mutational signatures and HR deficiency scores and correlated these with in vivo responses to the PARP inhibitor (PARPi) talazoparib in six PDXs representing the copy number high/p53-mutant and mismatch-repair deficient molecular subtypes of EC. Results PDX models were successfully generated from grade 2/3 tumors, including three uterine carcinosarcomas. The models showed similar histomorphology to the primary tumors and represented all four molecular subtypes of EC, including five mismatch-repair deficient models. The different PDX lineages showed a wide range of inter-tumor and intra-tumor heterogeneity. However, for most PDX models, one arm recapitulated the molecular landscape of the primary tumor without major genomic drift. An in vivo response to talazoparib was detected in four copy number high models. Two models (carcinosarcomas) showed a response consistent with stable disease and two models (one copy number high serous EC and another carcinosarcoma) showed significant tumor growth inhibition, albeit one consistent with progressive disease; however, all lacked the HR deficiency genomic signature. Conclusions EC PDX models represent the four molecular subtypes of disease and can capture intra-tumor heterogeneity of the original primary tumor. PDXs of the copy number high molecular subtype showed sensitivity to PARPi; however, deeper and more durable responses will likely require combination of PARPi with other agents. Supplementary Information The online version contains supplementary material available at 10.1186/s13073-021-00990-z.
Collapse
Affiliation(s)
- Vanessa F Bonazzi
- School of Biomedical Sciences, Queensland University of Technology located at the Translational Research Institute, Brisbane, QLD, Australia.,The University of Queensland Diamantina Institute, The University of Queensland, Woolloongabba, QLD, Australia
| | - Olga Kondrashova
- Department of Genetics and Computational Biology, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Deborah Smith
- Mater Health Services, South Brisbane, QLD, Australia.,Mater Pathology, Mater Research, Brisbane, QLD, Australia.,The University of Queensland, Brisbane, QLD, Australia
| | - Katia Nones
- Department of Genetics and Computational Biology, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Asmerom T Sengal
- School of Biomedical Sciences, Queensland University of Technology located at the Translational Research Institute, Brisbane, QLD, Australia
| | - Robert Ju
- School of Biomedical Sciences, Queensland University of Technology located at the Translational Research Institute, Brisbane, QLD, Australia
| | - Leisl M Packer
- School of Biomedical Sciences, Queensland University of Technology located at the Translational Research Institute, Brisbane, QLD, Australia
| | - Lambros T Koufariotis
- Department of Genetics and Computational Biology, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Stephen H Kazakoff
- Department of Genetics and Computational Biology, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Aimee L Davidson
- Department of Genetics and Computational Biology, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia.,The University of Queensland, Brisbane, QLD, Australia
| | - Priya Ramarao-Milne
- Department of Genetics and Computational Biology, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia.,The University of Queensland, Brisbane, QLD, Australia
| | - Vanessa Lakis
- Department of Genetics and Computational Biology, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Felicity Newell
- Department of Genetics and Computational Biology, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Rebecca Rogers
- Mater Pathology, Mater Research, Brisbane, QLD, Australia
| | - Claire Davies
- Mater Pathology, Mater Research, Brisbane, QLD, Australia
| | - James Nicklin
- The Wesley Hospital, Auchenflower, QLD, Australia.,Icon Cancer Centre Wesley, Auchenflower, QLD, Australia
| | - Andrea Garrett
- The Wesley Hospital, Auchenflower, QLD, Australia.,Icon Cancer Centre Wesley, Auchenflower, QLD, Australia
| | - Naven Chetty
- Mater Health Services, South Brisbane, QLD, Australia.,Mater Pathology, Mater Research, Brisbane, QLD, Australia
| | - Lewis Perrin
- Mater Health Services, South Brisbane, QLD, Australia.,Mater Pathology, Mater Research, Brisbane, QLD, Australia
| | - John V Pearson
- Department of Genetics and Computational Biology, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Ann-Marie Patch
- Department of Genetics and Computational Biology, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia.,The University of Queensland, Brisbane, QLD, Australia
| | - Nicola Waddell
- Department of Genetics and Computational Biology, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia.,The University of Queensland, Brisbane, QLD, Australia
| | - Pamela M Pollock
- School of Biomedical Sciences, Queensland University of Technology located at the Translational Research Institute, Brisbane, QLD, Australia.
| |
Collapse
|
65
|
Arai H, Elliott A, Millstein J, Xiu J, Ou FS, Innocenti F, Wang J, Battaglin F, Jayachandran P, Kawanishi N, Soni S, Zhang W, Sohal D, Goldberg RM, Hall MJ, Scott AJ, Khushman M, Hwang JJ, Lou E, Weinberg BA, Lockhart AC, Shields AF, Abraham JP, Magee D, Stafford P, Zhang J, Venook AP, Korn WM, Lenz HJ. Molecular characteristics and clinical outcomes of patients with Neurofibromin 1-altered metastatic colorectal cancer. Oncogene 2022; 41:260-267. [PMID: 34728807 PMCID: PMC8738154 DOI: 10.1038/s41388-021-02074-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 10/01/2021] [Accepted: 10/08/2021] [Indexed: 12/14/2022]
Abstract
Loss-of-function alterations of Neurofibromin 1 (NF1) activate RAS, a driver of colorectal cancer. However, the clinical implications of NF1 alterations are largely unknown. We performed a comprehensive molecular profiling of NF1-mutant colorectal cancer using data from 8150 patients included in a dataset of commercial CLIA-certified laboratory (Caris Life Sciences). In addition, NF1 expression levels were tested for associations with clinical outcomes using data from 431 patients in the CALGB/SWOG 80405 trial. In the Caris dataset, 2.2% of patients had pathogenic or presumed pathogenic NF1 mutations. NF1-mutant tumors more frequently harbored PIK3CA (25.0% vs. 16.7%) and PTEN mutations (24.0% vs. 4.2%) than wild type tumors. Gene set enrichment analysis revealed that MAPK and PI3K pathway signatures were enriched in NF1-mutant tumors. In the CALGB/SWOG 80405 cohort, low NF1 expression was associated with poor prognosis, and high NF1 expression was associated with better efficacy of cetuximab than bevacizumab. Together, we revealed concurrent genetic alterations in the PI3K pathways in NF1-mutant tumors, suggesting the need to simultaneously block MAPK and PI3K pathways in treatment. The potential of NF1 alteration as a novel biomarker for targeted therapy was highlighted, warranting further investigations in clinical settings.
Collapse
Affiliation(s)
- Hiroyuki Arai
- Division of Medical Oncology, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, USA
| | - Andrew Elliott
- Clinical & Translational Research, Medical Affairs, Caris Life Sciences, Phoenix, AZ, USA
| | - Joshua Millstein
- Department of Preventive Medicine, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, USA
| | - Joanne Xiu
- Clinical & Translational Research, Medical Affairs, Caris Life Sciences, Phoenix, AZ, USA
| | - Fang-Shu Ou
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA
| | - Federico Innocenti
- UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Jingyuan Wang
- Division of Medical Oncology, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, USA
| | - Francesca Battaglin
- Division of Medical Oncology, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, USA
| | - Priya Jayachandran
- Division of Medical Oncology, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, USA
| | - Natsuko Kawanishi
- Division of Medical Oncology, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, USA
| | - Shivani Soni
- Division of Medical Oncology, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, USA
| | - Wu Zhang
- Division of Medical Oncology, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, USA
| | - Davendra Sohal
- Division of Hematology/Oncology, University of Cincinnati, Cincinnati, OH, USA
| | | | - Michael J Hall
- Department of Clinical Genetics, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Aaron J Scott
- Division of Hematology and Oncology, Department of Medicine, Banner-University of Arizona, Tucson, AZ, USA
| | - Mohd Khushman
- Medical Oncology, Mitchell Cancer Institute, The University of South Alabama, Mobile, AL, USA
| | - Jimmy J Hwang
- Department of Solid Tumor Oncology, GI Medical Oncology, Levine Cancer Institute, Charlotte, NC, USA
| | - Emil Lou
- Division of Hematology, Oncology and Transplantation, University of Minnesota, Minneapolis, MN, USA
| | - Benjamin A Weinberg
- Ruesch Center for the Cure of Gastrointestinal Cancers, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, USA
| | - Albert Craig Lockhart
- Department of Medicine, Division of Oncology, University of Miami, Miller School of Medicine, Miami, FL, USA
| | - Anthony Frank Shields
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, USA
| | - Jim P Abraham
- Clinical & Translational Research, Medical Affairs, Caris Life Sciences, Phoenix, AZ, USA
| | - Daniel Magee
- Clinical & Translational Research, Medical Affairs, Caris Life Sciences, Phoenix, AZ, USA
| | - Phillip Stafford
- Clinical & Translational Research, Medical Affairs, Caris Life Sciences, Phoenix, AZ, USA
| | - Jian Zhang
- Clinical & Translational Research, Medical Affairs, Caris Life Sciences, Phoenix, AZ, USA
| | - Alan P Venook
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA
| | - W Michael Korn
- Clinical & Translational Research, Medical Affairs, Caris Life Sciences, Phoenix, AZ, USA
| | - Heinz-Josef Lenz
- Division of Medical Oncology, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, USA.
| |
Collapse
|
66
|
Kang HG, Hwangbo H, Kim MJ, Kim S, Lee EJ, Park MJ, Kim JW, Kim BG, Cho EH, Chang S, Lee JY, Choi JK. Aberrant Transcript Usage Is Associated with Homologous Recombination Deficiency and Predicts Therapeutic Response. Cancer Res 2022; 82:142-154. [PMID: 34711610 PMCID: PMC9397646 DOI: 10.1158/0008-5472.can-21-2023] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 09/15/2021] [Accepted: 10/25/2021] [Indexed: 01/07/2023]
Abstract
BRCA1/2 mutations account for only a small fraction of homologous recombination (HR) deficiency (HRD) cases. Recently developed genomic HRD (gHRD) tests suffer confounding factors that cause low precision in predicting samples that will respond to PARP inhibitors and DNA damaging agents. Here we present molecular and clinical evidence of transcriptional HRD (tHRD) that is based on aberrant transcript usage (aTU) of minor isoforms. Specifically, increased TU of nonfunctional isoforms of DNA repair genes was prevalent in breast and ovarian cancer with gHRD. Functional assays validated the association of aTU with impaired HR activity. Machine learning-based tHRD detection by the transcript usage (TU) pattern of key genes was superior to directly screening for gHRD or BRCA1/2 mutations in accurately predicting responses of cell lines and patients with cancer to PARP inhibitors and genotoxic drugs. This approach demonstrated the capability of tHRD status to reflect functional HR status, including in a cohort of olaparib-treated ovarian cancer with acquired platinum resistance. Diagnostic tests based on tHRD are expected to broaden the clinical utility of PARP inhibitors. SIGNIFICANCE: A novel but widespread transcriptional mechanism by which homologous recombination deficiency arises independently of BRCA1/2 mutations can be utilized as a companion diagnostic for PARP inhibitors.
Collapse
Affiliation(s)
- Hyeon Gu Kang
- Department of Bio and Brain Engineering, KAIST, Daejeon, Republic of Korea
| | - Haeun Hwangbo
- Department of Bio and Brain Engineering, KAIST, Daejeon, Republic of Korea
| | - Myung Ji Kim
- Department of Biomedical Engineering, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Sinae Kim
- Department of Biomedical Engineering, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Eun Ji Lee
- Department of Biomedical Engineering, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Min Ji Park
- Department of Biomedical Engineering, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Jae-Weon Kim
- Department of Obstetrics and Gynecology, Seoul National University Hospital, Seoul, Republic of Korea
| | - Byoung-Gie Kim
- Department of Obstetrics and Gynecology, Gynecologic Cancer Center, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Eun-Hae Cho
- Genome Research Center, GC Genome, GC Labs, Yongin-si, Gyeonggi-do, Republic of Korea.,Corresponding Authors: Eun-Hae Cho, GC Genome, GC Labs, 107 Ihyeonro, 30beon-gil, Giheung-gu, Yongin-si, Gyeonggi-do 16924, Republic of Korea. Phone: 82-31-260-9216; E-mail: ; Suhwan Chang, Department of Biomedical Engineering, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Pungnap 2(i)-dong, Songpa-gu, Seoul, Republic of Korea. Phone: 82-2-3010-2095; E-mail: ; Jung-Yun Lee, Department of Obstetrics and Gynecology, Yonsei University College of Medicine, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea. Phone: 82-2-2228-2237; E-mail: ; and Jung Kyoon Choi, YBS Building (E16-1), KAIST, 291 Daehak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea. Phone: 82-42-350-4327; E-mail:
| | - Suhwan Chang
- Department of Biomedical Engineering, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.,Corresponding Authors: Eun-Hae Cho, GC Genome, GC Labs, 107 Ihyeonro, 30beon-gil, Giheung-gu, Yongin-si, Gyeonggi-do 16924, Republic of Korea. Phone: 82-31-260-9216; E-mail: ; Suhwan Chang, Department of Biomedical Engineering, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Pungnap 2(i)-dong, Songpa-gu, Seoul, Republic of Korea. Phone: 82-2-3010-2095; E-mail: ; Jung-Yun Lee, Department of Obstetrics and Gynecology, Yonsei University College of Medicine, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea. Phone: 82-2-2228-2237; E-mail: ; and Jung Kyoon Choi, YBS Building (E16-1), KAIST, 291 Daehak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea. Phone: 82-42-350-4327; E-mail:
| | - Jung-Yun Lee
- Department of Obstetrics and Gynecology, Yonsei University College of Medicine, Seoul, Republic of Korea.,Corresponding Authors: Eun-Hae Cho, GC Genome, GC Labs, 107 Ihyeonro, 30beon-gil, Giheung-gu, Yongin-si, Gyeonggi-do 16924, Republic of Korea. Phone: 82-31-260-9216; E-mail: ; Suhwan Chang, Department of Biomedical Engineering, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Pungnap 2(i)-dong, Songpa-gu, Seoul, Republic of Korea. Phone: 82-2-3010-2095; E-mail: ; Jung-Yun Lee, Department of Obstetrics and Gynecology, Yonsei University College of Medicine, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea. Phone: 82-2-2228-2237; E-mail: ; and Jung Kyoon Choi, YBS Building (E16-1), KAIST, 291 Daehak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea. Phone: 82-42-350-4327; E-mail:
| | - Jung Kyoon Choi
- Department of Bio and Brain Engineering, KAIST, Daejeon, Republic of Korea.,PentaMedix Co., Ltd., Seongnam-si, Gyeonggi-do, Republic of Korea.,Corresponding Authors: Eun-Hae Cho, GC Genome, GC Labs, 107 Ihyeonro, 30beon-gil, Giheung-gu, Yongin-si, Gyeonggi-do 16924, Republic of Korea. Phone: 82-31-260-9216; E-mail: ; Suhwan Chang, Department of Biomedical Engineering, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Pungnap 2(i)-dong, Songpa-gu, Seoul, Republic of Korea. Phone: 82-2-3010-2095; E-mail: ; Jung-Yun Lee, Department of Obstetrics and Gynecology, Yonsei University College of Medicine, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea. Phone: 82-2-2228-2237; E-mail: ; and Jung Kyoon Choi, YBS Building (E16-1), KAIST, 291 Daehak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea. Phone: 82-42-350-4327; E-mail:
| |
Collapse
|
67
|
Pellegrino B, Tommasi C, Solinas C, Campanini N, Silini EM, Musolino A. The future potential of genome-wide mutational profiles in HRD detection in breast cancer. Expert Rev Mol Diagn 2021; 22:1-3. [PMID: 34913790 DOI: 10.1080/14737159.2022.2015328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Affiliation(s)
- Benedetta Pellegrino
- Department of Medicine and Surgery, University of Parma, Parma, Italy.,Medical Oncology and Breast Unit, University Hospital of Parma, Parma, Italy
| | - Chiara Tommasi
- Department of Medicine and Surgery, University of Parma, Parma, Italy.,Medical Oncology and Breast Unit, University Hospital of Parma, Parma, Italy
| | - Cinzia Solinas
- Department of Medical Oncology, A. Segni Hospital, Ozieri, Italy
| | | | | | - Antonino Musolino
- Department of Medicine and Surgery, University of Parma, Parma, Italy.,Medical Oncology and Breast Unit, University Hospital of Parma, Parma, Italy
| |
Collapse
|
68
|
van Wijk LM, Nilas AB, Vrieling H, Vreeswijk MPG. RAD51 as a functional biomarker for homologous recombination deficiency in cancer: a promising addition to the HRD toolbox? Expert Rev Mol Diagn 2021; 22:185-199. [PMID: 34913794 DOI: 10.1080/14737159.2022.2020102] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
INTRODUCTION Carcinomas with defects in the homologous recombination (HR) pathway are sensitive to PARP inhibitors (PARPi). A robust method to identify HR-deficient (HRD) carcinomas is therefore of utmost clinical importance. Currently available DNA-based HRD tests either scan HR-related genes such as BRCA1 and BRCA2 for the presence of pathogenic variants or identify HRD-related genomic scars or mutational signatures by using whole-exome or whole-genome sequencing data. As an alternative to DNA-based tests, functional HRD tests have been developed that assess the actual ability of tumors to accumulate RAD51 protein at DNA double strand breaks as a proxy for HR proficiency. AREAS COVERED This review presents an overview of currently available HRD tests and discuss the pros and cons of the different methodologies including their sensitivity for the identification of HRD tumors, their concordance with other HRD tests, and their capacity to predict therapy response. EXPERT OPINION With the increasing use of PARP inhibitors in the treatment of several cancers there is an urgent need to implement HRD testing in routine clinical practice. To this end, calibration of HRD thresholds and clinical validation of both DNA-based and RAD51-based HRD tests should have top-priority in the coming years.
Collapse
Affiliation(s)
- Lise M van Wijk
- Department of Human Genetics, Leiden University Medical Center, 2300 RC Leiden, the Netherlands
| | - Andreea B Nilas
- Department of Human Genetics, Leiden University Medical Center, 2300 RC Leiden, the Netherlands
| | - Harry Vrieling
- Department of Human Genetics, Leiden University Medical Center, 2300 RC Leiden, the Netherlands
| | - Maaike P G Vreeswijk
- Department of Human Genetics, Leiden University Medical Center, 2300 RC Leiden, the Netherlands
| |
Collapse
|
69
|
Zhang M, Ma SC, Tan JL, Wang J, Bai X, Dong ZY, Zhang QX. Inferring Homologous Recombination Deficiency of Ovarian Cancer From the Landscape of Copy Number Variation at Subchromosomal and Genetic Resolutions. Front Oncol 2021; 11:772604. [PMID: 34976815 PMCID: PMC8716765 DOI: 10.3389/fonc.2021.772604] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 11/19/2021] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Homologous recombination deficiency (HRD) is characterized by overall genomic instability and has emerged as an indispensable therapeutic target across various tumor types, particularly in ovarian cancer (OV). Unfortunately, current detection assays are far from perfect for identifying every HRD patient. The purpose of this study was to infer HRD from the landscape of copy number variation (CNV). METHODS Genome-wide CNV landscape was measured in OV patients from the Australian Ovarian Cancer Study (AOCS) clinical cohort and >10,000 patients across 33 tumor types from The Cancer Genome Atlas (TCGA). HRD-predictive CNVs at subchromosomal resolution were identified through exploratory analysis depicting the CNV landscape of HRD versus non-HRD OV patients and independently validated using TCGA and AOCS cohorts. Gene-level CNVs were further analyzed to explore their potential predictive significance for HRD across tumor types at genetic resolution. RESULTS At subchromosomal resolution, 8q24.2 amplification and 5q13.2 deletion were predominantly witnessed in HRD patients (both p < 0.0001), whereas 19q12 amplification occurred mainly in non-HRD patients (p < 0.0001), compared with their corresponding counterparts within TCGA-OV. The predictive significance of 8q24.2 amplification (p < 0.0001), 5q13.2 deletion (p = 0.0056), and 19q12 amplification (p = 0.0034) was externally validated within AOCS. Remarkably, pan-cancer analysis confirmed a cross-tumor predictive role of 8q24.2 amplification for HRD (p < 0.0001). Further analysis of CNV in 8q24.2 at genetic resolution revealed that amplifications of the oncogenes, MYC (p = 0.0001) and NDRG1 (p = 0.0004), located on this fragment were also associated with HRD in a pan-cancer manner. CONCLUSIONS The CNV landscape serves as a generalized predictor of HRD in cancer patients not limited to OV. The detection of CNV at subchromosomal or genetic resolution could aid in the personalized treatment of HRD patients.
Collapse
Affiliation(s)
- Meng Zhang
- Department of Obstetrics and Gynecology, Reproductive Medicine Centre, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Si-Cong Ma
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Information Management and Big Data Center, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jia-Le Tan
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jian Wang
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xue Bai
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zhong-Yi Dong
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Qing-Xue Zhang
- Department of Obstetrics and Gynecology, Reproductive Medicine Centre, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| |
Collapse
|
70
|
Lin H, Xie Y, Kong Y, Yang L, Li M. Identification of molecular subtypes and prognostic signature for hepatocellular carcinoma based on genes associated with homologous recombination deficiency. Sci Rep 2021; 11:24022. [PMID: 34912005 PMCID: PMC8674316 DOI: 10.1038/s41598-021-03432-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Accepted: 12/03/2021] [Indexed: 01/15/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is a rapidly developing digestive tract carcinoma. The prognosis of patients and side effects caused by clinical treatment should be better improved. Nonnegative matrix factorization (NMF) clustering was performed using 109 homologous recombination deficiency (HRD)-related of HCC genes from The Cancer Genome Atlas (TCGA) database. Limma was applied to analyze subtype differences. Immune scores and clinical characteristics of different subtypes were compared. An HRD signature were built with least absolute shrinkage operator (LASSO) and multivariate Cox analysis. Performance of the signature system was then assessed by Kaplan–Meier curves and receiver operating characteristic (ROC) curves. We identified two molecular subtypes (C1 and C2), with C2 showing a significantly better prognosis than C1. C1 contained 3623 differentially expressed genes. A 4-gene prognostic signature for HCC was established, and showed a high predicting accuracy in validation sets, entire TCGA data set, HCCDB18 and GSE14520 queues. Moreover, the risk score was validated as an independent prognostic marker for HCC. Our research identified two molecular subtypes of HCC, and proposed a novel scoring system for evaluating the prognosis of HCC in clinical practice.
Collapse
Affiliation(s)
- Hongsheng Lin
- Guangxi University of Chinese Medicine, Nanning, 530200, China.,Department of Laboratory, The First Affiliated Hospital of Guangxi University of Chinese Medicine, Nanning, 530023, China.,Guangxi Medical University, Nanning, 530021, China.,Department of Microbiology, School of Basic Medical Sciences, Guangxi Medical University, Nanning, 530021, China
| | - Yangyi Xie
- Guangxi University of Chinese Medicine, Nanning, 530200, China.,The First Clinical Faculty of Guangxi University of Chinese Medicine, Nanning, 530200, China
| | - Yinzhi Kong
- Guangxi University of Chinese Medicine, Nanning, 530200, China.,The First Clinical Faculty of Guangxi University of Chinese Medicine, Nanning, 530200, China
| | - Li Yang
- Guangxi University of Chinese Medicine, Nanning, 530200, China.,Department of Laboratory, The First Affiliated Hospital of Guangxi University of Chinese Medicine, Nanning, 530023, China
| | - Mingfen Li
- Guangxi University of Chinese Medicine, Nanning, 530200, China. .,Department of Laboratory, The First Affiliated Hospital of Guangxi University of Chinese Medicine, Nanning, 530023, China.
| |
Collapse
|
71
|
De Sarkar N, Dasgupta S, Chatterjee P, Coleman I, Ha G, Ang LS, Kohlbrenner EA, Frank SB, Nunez TA, Salipante SJ, Corey E, Morrissey C, Van Allen E, Schweizer MT, Haffner MC, Patel R, Hanratty B, Lucas JM, Dumpit RF, Pritchard CC, Montgomery RB, Nelson PS. Genomic attributes of homology-directed DNA repair deficiency in metastatic prostate cancer. JCI Insight 2021; 6:152789. [PMID: 34877933 PMCID: PMC8675196 DOI: 10.1172/jci.insight.152789] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 10/20/2021] [Indexed: 01/08/2023] Open
Abstract
Cancers with homology-directed DNA repair (HRR) deficiency exhibit high response rates to poly(ADP-ribose) polymerase inhibitors (PARPi) and platinum chemotherapy. Though mutations disrupting BRCA1 and BRCA2 associate with HRR deficiency (HRRd), patterns of genomic aberrations and mutation signatures may be more sensitive and specific indicators of compromised repair. Here, we evaluated whole-exome sequences from 418 metastatic prostate cancers (mPCs) and determined that one-fifth exhibited genomic characteristics of HRRd that included Catalogue Of Somatic Mutations In Cancer mutation signature 3. Notably, a substantial fraction of tumors with genomic features of HRRd lacked biallelic loss of a core HRR-associated gene, such as BRCA2. In this subset, HRRd associated with loss of chromodomain helicase DNA binding protein 1 but not with mutations in serine-protein kinase ATM, cyclin dependent kinase 12, or checkpoint kinase 2. HRRd genomic status was strongly correlated with responses to PARPi and platinum chemotherapy, a finding that supports evaluating biomarkers reflecting functional HRRd for treatment allocation.
Collapse
Affiliation(s)
| | | | | | | | - Gavin Ha
- Divisions of Human Biology.,Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Lisa S Ang
- Divisions of Human Biology.,Clinical Research
| | | | | | | | | | - Eva Corey
- Department of Urology, University of Washington, Seattle, Washington, USA
| | - Colm Morrissey
- Department of Urology, University of Washington, Seattle, Washington, USA
| | | | - Michael T Schweizer
- Division of Oncology, Department of Medicine, University of Washington, Seattle, Washington, USA
| | | | | | | | | | | | | | - Robert B Montgomery
- Division of Oncology, Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Peter S Nelson
- Divisions of Human Biology.,Clinical Research.,Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA.,Department of Laboratory Medicine and Pathology and.,Department of Urology, University of Washington, Seattle, Washington, USA.,Division of Oncology, Department of Medicine, University of Washington, Seattle, Washington, USA
| |
Collapse
|
72
|
Yordanova M, Hubert A, Hassan S. Expanding the Use of PARP Inhibitors as Monotherapy and in Combination in Triple-Negative Breast Cancer. Pharmaceuticals (Basel) 2021; 14:1270. [PMID: 34959671 PMCID: PMC8709256 DOI: 10.3390/ph14121270] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 11/27/2021] [Accepted: 11/29/2021] [Indexed: 12/31/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is the most aggressive subtype of breast cancer, and is known to be associated with a poor prognosis and limited therapeutic options. Poly (ADP-ribose) polymerase inhibitors (PARPi) are targeted therapeutics that have demonstrated efficacy as monotherapy in metastatic BRCA-mutant (BRCAMUT) TNBC patients. Improved efficacy of PARPi has been demonstrated in BRCAMUT breast cancer patients who have either received fewer lines of chemotherapy or in chemotherapy-naïve patients in the metastatic, adjuvant, and neoadjuvant settings. Moreover, recent trials in smaller cohorts have identified anti-tumor activity of PARPi in TNBC patients, regardless of BRCA-mutation status. While there have been concerns regarding the efficacy and toxicity of the use of PARPi in combination with chemotherapy, these challenges can be mitigated with careful attention to PARPi dosing strategies. To better identify a patient subpopulation that will best respond to PARPi, several genomic biomarkers of homologous recombination deficiency have been tested. However, gene expression signatures associated with PARPi response can integrate different pathways in addition to homologous recombination deficiency and can be implemented in the clinic more readily. Taken together, PARPi have great potential for use in TNBC patients beyond BRCAMUT status, both as a single-agent and in combination.
Collapse
Affiliation(s)
- Mariya Yordanova
- Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC J1K 2R1, Canada;
| | - Audrey Hubert
- Faculty of Medicine, Université de Montréal, Montréal, QC H3C 3T5, Canada;
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), l’Institut de Cancer de Montreal, Montreal, QC H2X 0A9, Canada
| | - Saima Hassan
- Faculty of Medicine, Université de Montréal, Montréal, QC H3C 3T5, Canada;
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), l’Institut de Cancer de Montreal, Montreal, QC H2X 0A9, Canada
- Division of Surgical Oncology, Department of Surgery, Centre Hospitalier de l’Université de Montréal (CHUM), Montreal, QC H2X 0C1, Canada
| |
Collapse
|
73
|
Urbina-Jara LK, Martinez-Ledesma E, Rojas-Martinez A, Rodriguez-Recio FR, Ortiz-Lopez R. DNA Repair Genes as Drug Candidates for Early Breast Cancer Onset in Latin America: A Systematic Review. Int J Mol Sci 2021; 22:13030. [PMID: 34884835 PMCID: PMC8657579 DOI: 10.3390/ijms222313030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 11/21/2021] [Accepted: 11/23/2021] [Indexed: 12/24/2022] Open
Abstract
The prevalence of breast cancer in young women (YWBC) has increased alarmingly. Significant efforts are being made to elucidate the biological mechanisms concerning the development, prognosis, and pathological response in early-onset breast cancer (BC) patients. Dysfunctional DNA repair proteins are implied in BC predisposition, progression, and therapy response, underscoring the need for further analyses on DNA repair genes. Public databases of large patient datasets such as METABRIC, TCGA, COSMIC, and cancer cell lines allow the identification of variants in DNA repair genes and possible precision drug candidates. This study aimed at identifying variants and drug candidates that may benefit Latin American (LA) YWBC. We analyzed pathogenic variants in 90 genes involved in DNA repair in public BC datasets from METABRIC, TCGA, COSMIC, CCLE, and COSMIC Cell Lines Project. Results showed that reported DNA repair germline variants in the LA dataset are underrepresented in large databases, in contrast to other populations. Additionally, only six gene repair variants in women under 50 years old from the study population were reported in BC cell lines. Therefore, there is a need for new approaches to study DNA repair variants reported in young women from LA.
Collapse
Affiliation(s)
| | | | | | | | - Rocio Ortiz-Lopez
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Monterrey 64710, Mexico; (L.K.U.-J.); (E.M.-L.); (A.R.-M.); (F.R.R.-R.)
| |
Collapse
|
74
|
Rucaparib in patients presenting a metastatic breast cancer with homologous recombination deficiency, without germline BRCA1/2 mutation. Eur J Cancer 2021; 159:283-295. [PMID: 34837859 DOI: 10.1016/j.ejca.2021.09.028] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 09/21/2021] [Accepted: 09/22/2021] [Indexed: 12/28/2022]
Abstract
BACKGROUND Breast cancer may present genomic alterations leading to homologous recombination deficiency (HRD). PARP inhibitors have proven their efficacy in patients with HER2-negative (HER2-) metastatic breast cancer (mBC) harbouring germline (g) BRCA1/2 mutations in 3 phases III trials. The single-arm phase II RUBY trial included 42 patients, 40 of whom received at least one dose of rucaparib. RUBY study assessed the efficacy of rucaparib in HER2-mBC with either high genomic loss of heterozygosity (LOH) score or non-germline BRCA1/2 mutation. PATIENTS AND METHODS The primary objective was the clinical benefit rate (CBR), and the study was powered to see 20% CBR using a 2-stage Simon design. RESULTS The primary-end point was not reached with a CBR of 13.5%. Two LOH-high patients, without somatic BRCA1/2 mutation, presented a complete and durable response (12 and 28.5 months). Whole-genome analysis was performed on 24 samples, including 5 patients who presented a clinical benefit from rucaparib. HRDetect tended to be associated with response to rucaparib, without reaching statistical significance (median HRDetect responders versus non-responders: 0.465 versus 0.040; p = 0.2135). Finally, 220 of 711 patients with mBC screened for LOH upstream from RUBY presented a high LOH score associated with a higher likelihood of death (hazard ratio = 1.39; 95% CI: 1.11-1.75; p = 0.005). CONCLUSION Our data suggest that a small subset of patients with high LOH scores without germline BRCA1/2 mutation could derive benefit from PARP inhibitors. However, the RUBY study underlines the need to develop additional biomarkers to identify selectively potential responders.
Collapse
|
75
|
Li J, M. Saville K, Ibrahim M, Zeng X, McClellan S, Angajala A, Beiser A, Andrews JF, Sun M, Koczor CA, Clark J, Hayat F, Makarov MV, Wilk A, Yates NA, Migaud ME, Sobol RW. NAD + bioavailability mediates PARG inhibition-induced replication arrest, intra S-phase checkpoint and apoptosis in glioma stem cells. NAR Cancer 2021; 3:zcab044. [PMID: 34806016 PMCID: PMC8600031 DOI: 10.1093/narcan/zcab044] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 10/20/2021] [Accepted: 11/10/2021] [Indexed: 01/31/2023] Open
Abstract
Elevated expression of the DNA damage response proteins PARP1 and poly(ADP-ribose) glycohydrolase (PARG) in glioma stem cells (GSCs) suggests that glioma may be a unique target for PARG inhibitors (PARGi). While PARGi-induced cell death is achieved when combined with ionizing radiation, as a single agent PARG inhibitors appear to be mostly cytostatic. Supplementation with the NAD+ precursor dihydronicotinamide riboside (NRH) rapidly increased NAD+ levels in GSCs and glioma cells, inducing PARP1 activation and mild suppression of replication fork progression. Administration of NRH+PARGi triggers hyperaccumulation of poly(ADP-ribose) (PAR), intra S-phase arrest and apoptosis in GSCs but minimal PAR induction or cytotoxicity in normal astrocytes. PAR accumulation is regulated by select PARP1- and PAR-interacting proteins. The involvement of XRCC1 highlights the base excision repair pathway in responding to replication stress while enhanced interaction of PARP1 with PCNA, RPA and ORC2 upon PAR accumulation implicates replication associated PARP1 activation and assembly with pre-replication complex proteins upon initiation of replication arrest, the intra S-phase checkpoint and the onset of apoptosis.
Collapse
Affiliation(s)
- Jianfeng Li
- Mitchell Cancer Institute, University of South Alabama, Mobile, AL 36604, USA,Department of Pharmacology, College of Medicine, University of South Alabama, Mobile, AL 36604, USA
| | - Kate M. Saville
- Mitchell Cancer Institute, University of South Alabama, Mobile, AL 36604, USA,Department of Pharmacology, College of Medicine, University of South Alabama, Mobile, AL 36604, USA
| | - Md Ibrahim
- Mitchell Cancer Institute, University of South Alabama, Mobile, AL 36604, USA,Department of Pharmacology, College of Medicine, University of South Alabama, Mobile, AL 36604, USA
| | - Xuemei Zeng
- Biomedical Mass Spectrometry Center, University of Pittsburgh Schools of the Health Sciences, Pittsburgh, PA 15213, USA
| | - Steve McClellan
- Mitchell Cancer Institute, University of South Alabama, Mobile, AL 36604, USA
| | - Anusha Angajala
- Mitchell Cancer Institute, University of South Alabama, Mobile, AL 36604, USA,Department of Pharmacology, College of Medicine, University of South Alabama, Mobile, AL 36604, USA
| | - Alison Beiser
- Mitchell Cancer Institute, University of South Alabama, Mobile, AL 36604, USA,Department of Pharmacology, College of Medicine, University of South Alabama, Mobile, AL 36604, USA
| | - Joel F Andrews
- Mitchell Cancer Institute, University of South Alabama, Mobile, AL 36604, USA
| | - Mai Sun
- Biomedical Mass Spectrometry Center, University of Pittsburgh Schools of the Health Sciences, Pittsburgh, PA 15213, USA
| | - Christopher A Koczor
- Mitchell Cancer Institute, University of South Alabama, Mobile, AL 36604, USA,Department of Pharmacology, College of Medicine, University of South Alabama, Mobile, AL 36604, USA
| | - Jennifer Clark
- Mitchell Cancer Institute, University of South Alabama, Mobile, AL 36604, USA,Department of Pharmacology, College of Medicine, University of South Alabama, Mobile, AL 36604, USA
| | - Faisal Hayat
- Mitchell Cancer Institute, University of South Alabama, Mobile, AL 36604, USA,Department of Pharmacology, College of Medicine, University of South Alabama, Mobile, AL 36604, USA
| | - Mikhail V Makarov
- Mitchell Cancer Institute, University of South Alabama, Mobile, AL 36604, USA,Department of Pharmacology, College of Medicine, University of South Alabama, Mobile, AL 36604, USA
| | - Anna Wilk
- Mitchell Cancer Institute, University of South Alabama, Mobile, AL 36604, USA,Department of Pharmacology, College of Medicine, University of South Alabama, Mobile, AL 36604, USA
| | - Nathan A Yates
- Biomedical Mass Spectrometry Center, University of Pittsburgh Schools of the Health Sciences, Pittsburgh, PA 15213, USA,Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Marie E Migaud
- Mitchell Cancer Institute, University of South Alabama, Mobile, AL 36604, USA,Department of Pharmacology, College of Medicine, University of South Alabama, Mobile, AL 36604, USA
| | - Robert W Sobol
- To whom correspondence should be addressed. Tel: +1 251 445 9846;
| |
Collapse
|
76
|
van der Wijngaart H, Hoes LR, van Berge Henegouwen JM, van der Velden DL, Zeverijn LJ, Roepman P, van Werkhoven E, de Leng WWJ, Jansen AML, Mehra N, Robbrecht DGJ, Labots M, de Groot DJA, Hoeben A, Hamberg P, Gelderblom H, Voest EE, Verheul HMW. Patients with Biallelic BRCA1/2 Inactivation Respond to Olaparib Treatment Across Histologic Tumor Types. Clin Cancer Res 2021; 27:6106-6114. [PMID: 34475104 DOI: 10.1158/1078-0432.ccr-21-1104] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 05/20/2021] [Accepted: 08/31/2021] [Indexed: 11/16/2022]
Abstract
PURPOSE To assess the efficacy of olaparib, a PARP inhibitor (PARPi) in patients with tumors with BRCA1/2 mutations, regardless of histologic tumor type. PATIENTS AND METHODS Patients with treatment-refractory BRCA1/2-mutated cancer were included for treatment with off-label olaparib 300 mg twice daily until disease progression or unacceptable toxicity. In Drug Rediscovery Protocol (DRUP), patients with treatment-refractory solid malignancies receive off-label drugs based on tumor molecular profiles while whole-genome sequencing (WGS) is performed on baseline tumor biopsies. The primary endpoint was clinical benefit (CB; defined as objective response or stable disease ≥ 16 weeks according to RECIST 1.1). Per protocol patients were enrolled using a Simon-like two-stage model. RESULTS Twenty-four evaluable patients with nine different tumor types harboring BRCA1/2 mutations were included, 58% had CB from treatment with olaparib. CB was observed in patients with complete loss of function (LoF) of BRCA1/2, while 73% of patients with biallelic BRCA LoF had CB. In 17 patients with and seven without current labeled indication, 10 and four patients had CB, respectively. Treatment resistance in four patients with biallelic loss might be explained by an additional oncogenic driver which was discovered by WGS, including Wnt pathway activation, FGFR amplification, and CDKN2A loss, in three tumor types. CONCLUSIONS These data indicate that using PARPis is a promising treatment strategy for patients with non-BRCA-associated histologies harboring biallelic BRCA LoF. WGS allows to accurately detect complete LoF of BRCA and homologous repair deficiency (HRD) signature as well as oncogenic drivers that may contribute to resistance, using a single assay.
Collapse
Affiliation(s)
- Hanneke van der Wijngaart
- Department of Medical Oncology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands.,Oncode Institute, Utrecht, the Netherlands
| | - Louisa R Hoes
- Oncode Institute, Utrecht, the Netherlands.,Department of Molecular Oncology & Immunology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - J Maxime van Berge Henegouwen
- Oncode Institute, Utrecht, the Netherlands.,Department of Medical Oncology, Leiden University Medical Center, Rapenburg, Leiden, the Netherlands
| | - Daphne L van der Velden
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Center, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Laurien J Zeverijn
- Oncode Institute, Utrecht, the Netherlands.,Department of Molecular Oncology & Immunology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Paul Roepman
- Hartwig Medical Foundation, Amsterdam, the Netherlands
| | - Erik van Werkhoven
- Department of Biometrics, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Wendy W J de Leng
- Department of Pathology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Anne M L Jansen
- Department of Pathology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Niven Mehra
- Department of Medical Oncology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Debbie G J Robbrecht
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - Mariette Labots
- Department of Medical Oncology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Derk Jan A de Groot
- Department of Medical Oncology, University Medical Center Groningen, Groningen, the Netherlands
| | - Ann Hoeben
- Department of Medical Oncology, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Paul Hamberg
- Department of Internal Medicine, Franciscus Gasthuis and Vlietland, Rotterdam, the Netherlands
| | - Hans Gelderblom
- Department of Medical Oncology, Leiden University Medical Center, Rapenburg, Leiden, the Netherlands
| | - Emile E Voest
- Oncode Institute, Utrecht, the Netherlands.,Department of Molecular Oncology & Immunology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Henk M W Verheul
- Department of Medical Oncology, Radboud University Medical Center, Nijmegen, the Netherlands.
| |
Collapse
|
77
|
Yoshioka KI, Kusumoto-Matsuo R, Matsuno Y, Ishiai M. Genomic Instability and Cancer Risk Associated with Erroneous DNA Repair. Int J Mol Sci 2021; 22:12254. [PMID: 34830134 PMCID: PMC8625880 DOI: 10.3390/ijms222212254] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 11/11/2021] [Accepted: 11/11/2021] [Indexed: 12/23/2022] Open
Abstract
Many cancers develop as a consequence of genomic instability, which induces genomic rearrangements and nucleotide mutations. Failure to correct DNA damage in DNA repair defective cells, such as in BRCA1 and BRCA2 mutated backgrounds, is directly associated with increased cancer risk. Genomic rearrangement is generally a consequence of erroneous repair of DNA double-strand breaks (DSBs), though paradoxically, many cancers develop in the absence of DNA repair defects. DNA repair systems are essential for cell survival, and in cancers deficient in one repair pathway, other pathways can become upregulated. In this review, we examine the current literature on genomic alterations in cancer cells and the association between these alterations and DNA repair pathway inactivation and upregulation.
Collapse
Affiliation(s)
- Ken-ichi Yoshioka
- Laboratory of Genome Stability Maintenance, National Cancer Center Research Institute, Tsukiji, Chuo-ku, Tokyo 104-0045, Japan; (R.K.-M.); (Y.M.)
| | - Rika Kusumoto-Matsuo
- Laboratory of Genome Stability Maintenance, National Cancer Center Research Institute, Tsukiji, Chuo-ku, Tokyo 104-0045, Japan; (R.K.-M.); (Y.M.)
| | - Yusuke Matsuno
- Laboratory of Genome Stability Maintenance, National Cancer Center Research Institute, Tsukiji, Chuo-ku, Tokyo 104-0045, Japan; (R.K.-M.); (Y.M.)
- Department of Applied Chemistry, Faculty of Science, Tokyo University of Science, Kagurazaka, Shinjuku-ku, Tokyo 162-8601, Japan
| | - Masamichi Ishiai
- Central Radioisotope Division, National Cancer Center Research Institute, Tsukiji, Chuo-ku, Tokyo 104-0045, Japan;
| |
Collapse
|
78
|
Endo S, Yoshino Y, Shirota M, Watanabe G, Chiba N. BRCA1/ATF1-Mediated Transactivation is Involved in Resistance to PARP Inhibitors and Cisplatin. CANCER RESEARCH COMMUNICATIONS 2021; 1:90-105. [PMID: 36860287 PMCID: PMC9973406 DOI: 10.1158/2767-9764.crc-21-0064] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 10/17/2021] [Accepted: 10/28/2021] [Indexed: 11/16/2022]
Abstract
Homologous recombination (HR)-deficient cells are sensitive to PARP inhibitors through a synthetic lethal effect. We previously developed an HR activity assay named Assay of Site-Specific HR Activity (ASHRA). Here, we evaluated the HR activity of 30 missense variants of BRCA1 by ASHRA and found that several BRCA1 variants showed intermediate HR activity, which was not clearly discerned by our previous analyses using a conventional method. HR activity measured by ASHRA was significantly correlated with sensitivity to olaparib. However, cells expressing the severely HR-deficient BRCA1-C61G variant were resistant to olaparib, and resistance was dependent on high expression of activating transcription factor 1 (ATF1), which binds to BRCA1 and activates the transcription of target genes to regulate cell proliferation. The BRCA1-C61G variant bound to ATF1 and stimulated ATF1-mediated transactivation similar to wild-type BRCA1. High expression of ATF1 conferred resistance to olaparib and cisplatin activating BRCA1/ATF1-mediated transcription without affecting HR activity in BRCA2-knockdown or RAD51-knockdown cells, but not in BRCA1-knockdown cells. These results suggest that ASHRA is a useful method to evaluate HR activity in cells and to predict the sensitivity to PARP inhibitors. The expression level of ATF1 might be an important biomarker of the effect of PARP inhibitors and platinum agents on HR-deficient tumors with the BRCA1-C61G variant or alteration of non-BRCA1 HR factors such as BRCA2 and RAD51. Significance ASHRA could evaluate HR activity in cells and predict the sensitivity to PARP inhibitors. High expression level of ATF1 may predict the resistance of BRCAness tumors with alterations of non-BRCA1 HR factors to PARP inhibitors and platinum agents.
Collapse
Affiliation(s)
- Shino Endo
- Department of Cancer Biology, Institute of Aging, Development, and Cancer, Tohoku University, Sendai, Japan
- Department of Cancer Biology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yuki Yoshino
- Department of Cancer Biology, Institute of Aging, Development, and Cancer, Tohoku University, Sendai, Japan
- Department of Cancer Biology, Tohoku University Graduate School of Medicine, Sendai, Japan
- Laboratory of Cancer Biology, Graduate School of Life Sciences, Tohoku University, Sendai, Japan
| | - Matsuyuki Shirota
- Division of Interdisciplinary Medical Science, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Gou Watanabe
- Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | - Natsuko Chiba
- Department of Cancer Biology, Institute of Aging, Development, and Cancer, Tohoku University, Sendai, Japan
- Department of Cancer Biology, Tohoku University Graduate School of Medicine, Sendai, Japan
- Laboratory of Cancer Biology, Graduate School of Life Sciences, Tohoku University, Sendai, Japan
| |
Collapse
|
79
|
Tian Y, Wei Y, Liu H, Shang H, Xu Y, Wu T, Liu W, Huang A, Dang Q, Sun Y. Significance of CD8 + T cell infiltration-related biomarkers and the corresponding prediction model for the prognosis of kidney renal clear cell carcinoma. Aging (Albany NY) 2021; 13:22912-22933. [PMID: 34606472 PMCID: PMC8544304 DOI: 10.18632/aging.203584] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 08/17/2021] [Indexed: 01/09/2023]
Abstract
Cytotoxic T cells expressing cell surface CD8 played a key role in anti-cancer immunotherapy, including kidney renal clear cell carcinoma (KIRC). Here we set out to comprehensively analyze and evaluate the significance of CD8+ T cell-related markers for patients with KIRC. We checked immune cell response in KIRC and identified cell type-specific markers and related pathways in the tumor-infiltrating CD8+ T (TIL-CD8T) cells. We used these markers to explore their prognostic signatures in TIL-CD8+ T by evaluating their prognostic efficacy and group differences at various levels. Through pan-cancer analysis, 12 of 63 up-regulated and 162 of 396 down-regulated genes in CD8+ T cells were found to be significantly correlated with the survival prognosis. Based on our highly integrated multi-platform analyses across multiple datasets, we constructed a 6-gene risk scoring model specific to TIL-CD8T. In this model, high TIL-CD8 sig score was corresponding to a higher incidence frequency of copy number variation and drug sensitivity to sorafenib. Moreover, the prognosis of patients with the same or similar immune checkpoint gene levels could be distinguished from each other by TIL-CD8 sig score.
Collapse
Affiliation(s)
- Yuan Tian
- Somatic Radiotherapy Department, Shandong Second Provincial General Hospital, Shandong Provincial ENT Hospital, Jinan 250023, Shandong, P.R. China
| | - Yumei Wei
- Head and Neck Radiotherapy Department, Shandong Provincial ENT Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250023, Shandong, P.R. China
| | - Hongmei Liu
- Radiotherapy Oncology Department, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Shandong Lung Cancer Institute, Jinan 250014, Shandong, P.R. China
| | - Heli Shang
- Radiotherapy Oncology Department, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Shandong Lung Cancer Institute, Jinan 250014, Shandong, P.R. China
| | - Yuedong Xu
- Endocrinology Department, Shandong Provincial Qianfoshan Hospital, The First Hospital Affiliated with Shandong First Medical University, Jinan 250014, Shandong, P.R. China
| | - Tong Wu
- Somatic Radiotherapy Department, Shandong Second Provincial General Hospital, Shandong Provincial ENT Hospital, Jinan 250023, Shandong, P.R. China
| | - Wei Liu
- Somatic Radiotherapy Department, Shandong Second Provincial General Hospital, Shandong Provincial ENT Hospital, Jinan 250023, Shandong, P.R. China
| | - Alan Huang
- Department of Oncology, Jinan Central Hospital, The First Hospital Affiliated with Shandong First Medical University, Jinan 250013, Shandong, P.R. China
| | - Qi Dang
- Department of Radiotherapy Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250012, Shandong, P.R. China
| | - Yuping Sun
- Phase I Clinical Trial Center, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250012, Shandong, P.R. China
| |
Collapse
|
80
|
Predicted Immunogenicity of CDK12 Biallelic Loss-of-Function Tumors Varies across Cancer Types. J Mol Diagn 2021; 23:1761-1773. [PMID: 34562615 DOI: 10.1016/j.jmoldx.2021.08.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 08/21/2021] [Accepted: 08/26/2021] [Indexed: 01/01/2023] Open
Abstract
CDK12 biallelic inactivation is associated with a distinct genomic signature of focal tandem duplications (FTDs). Gene fusions resulting from FTDs increase neoantigen load, raising interest in CDK12 as a biomarker of response to immune checkpoint inhibitors. Despite evidence of FTDs in multiple CDK12-altered cancer types, notably prostate and ovarian, report of fusion-associated neoantigen load is limited to prostate cancer. Molecular profiles were retrospectively reviewed for CDK12-biallelic (CDK12-biLOF) and -monoallelic loss-of-function (CDK12-monoLOF) in a primary cohort of >9000 tumors, representing 39 cancer types, and immune epitopes were predicted from fusions detected by whole transcriptome sequencing. CDK12-biLOF was identified for 0.3% tumors overall, most frequently in prostate cancer (4.7%). CDK12-biLOF tumors had higher mean fusion rates and fusion-associated neoantigen load than CDK12-monoLOF and CDK12-WT tumors (P < 0.01). However, concurrent mismatch repair deficiency/microsatellite instability with CDK12-biLOF associated with low fusion rates. Among CDK12-biLOF tumors, fusion-associated neoantigen load was highest in prostate and ovarian cancers, which correlated with distinct immune profiles. In a validation cohort, CDK12-biLOF tumors (0.4%) exhibited high mean fusion rates, particularly for prostate and ovarian tumors. Low fusion rates in other CDK12-biLOF tumor types warrant further investigation and highlight the value of quantitative biomarkers. Fusion rate and fusion-associated neoantigen load are linked to CDK12-biLOF in select cancers and may help to identify responders of immune checkpoint inhibitor therapy.
Collapse
|
81
|
Abécassis J, Reyal F, Vert JP. CloneSig can jointly infer intra-tumor heterogeneity and mutational signature activity in bulk tumor sequencing data. Nat Commun 2021; 12:5352. [PMID: 34504064 PMCID: PMC8429716 DOI: 10.1038/s41467-021-24992-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Accepted: 07/12/2021] [Indexed: 02/07/2023] Open
Abstract
Systematic DNA sequencing of cancer samples has highlighted the importance of two aspects of cancer genomics: intra-tumor heterogeneity (ITH) and mutational processes. These two aspects may not always be independent, as different mutational processes could be involved in different stages or regions of the tumor, but existing computational approaches to study them largely ignore this potential dependency. Here, we present CloneSig, a computational method to jointly infer ITH and mutational processes in a tumor from bulk-sequencing data. Extensive simulations show that CloneSig outperforms current methods for ITH inference and detection of mutational processes when the distribution of mutational signatures changes between clones. Applied to a large cohort of 8,951 tumors with whole-exome sequencing data from The Cancer Genome Atlas, and on a pan-cancer dataset of 2,632 whole-genome sequencing tumor samples from the Pan-Cancer Analysis of Whole Genomes initiative, CloneSig obtains results overall coherent with previous studies.
Collapse
Affiliation(s)
- Judith Abécassis
- Institut Curie, PSL Research University, Translational Research Department, INSERM, U932 Immunity and Cancer, Residual Tumor & Response to Treatment Laboratory (RT2Lab), Paris, France
- MINES ParisTech, PSL University, CBIO - Centre for Computational Biology, Paris, France
- Institut Curie, PSL Research University, Paris, France
| | - Fabien Reyal
- Institut Curie, PSL Research University, Translational Research Department, INSERM, U932 Immunity and Cancer, Residual Tumor & Response to Treatment Laboratory (RT2Lab), Paris, France
- Department of Surgery, Institut Curie, Paris, France
| | - Jean-Philippe Vert
- MINES ParisTech, PSL University, CBIO - Centre for Computational Biology, Paris, France.
- Google Research, Brain team, Paris, France.
| |
Collapse
|
82
|
Wu CH, Hsieh CS, Chang YC, Huang CC, Yeh HT, Hou MF, Chung YC, Tu SH, Chang KJ, Chattopadhyay A, Lai LC, Lu TP, Li YH, Tsai MH, Chuang EY. Differential whole-genome doubling and homologous recombination deficiencies across breast cancer subtypes from the Taiwanese population. Commun Biol 2021; 4:1052. [PMID: 34504292 PMCID: PMC8429690 DOI: 10.1038/s42003-021-02597-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Accepted: 08/05/2021] [Indexed: 12/11/2022] Open
Abstract
Whole-genome doubling (WGD) is an early macro-evolutionary event in tumorigenesis, involving the doubling of an entire chromosome complement. However, its impact on breast cancer subtypes remains unclear. Here, we performed a comprehensive and quantitative analysis of WGD and its influence on breast cancer subtypes in patients from Taiwan and consequently highlight the genomic association between WGD and homologous recombination deficiency (HRD). A higher manifestation of WGD was reported in triple-negative breast cancer, conferring high chromosomal instability (CIN), while HER2 + tumors exhibited early WGD events, with widely varied CIN levels, compared to luminal-type tumors. An association of higher activity of de novo indel signature 2 with WGD and HRD in Taiwanese breast cancer patients was reported. A control test between WGD and pseudo non-WGD samples was further employed to support this finding. The study provides a better comprehension of tumorigenesis in breast cancer subtypes, thus assisting in personalized treatment. Wu, Hsieh et al. analyze Taiwanese breast cancer patient samples using whole-exome sequencing to examine the heterogeneity and homogeneity in the timing and dependencies of somatic aberrations across disease subtypes. The authors focus on somatic alterations and related features that correlate with whole genome doubling, including homologous recombination deficiencies.
Collapse
Affiliation(s)
- Chia-Hsin Wu
- Graduate Institute of Biomedical Electronics and Bioinformatics, National Taiwan University, Taipei, Taiwan
| | - Chia-Shan Hsieh
- Genome and Systems Biology Degree Program, National Taiwan University, Taipei, Taiwan
| | | | - Chi-Cheng Huang
- Comprehensive Breast Health Center, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Hsien-Tang Yeh
- Department of Surgery, Lotung Poh-Ai Hospital, Yilan County, Taiwan
| | - Ming-Feng Hou
- Division of Breast Surgery, Department of Surgery, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yuan-Chiang Chung
- Department of Breast Surgery, Dajia Branch, Kuang Tien General Hospital, Taichung, Taiwan
| | - Shih-Hsin Tu
- Department of Surgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - King-Jen Chang
- Department of Surgery, National Taiwan University Hospital, Taipei, Taiwan
| | - Amrita Chattopadhyay
- Bioinformatics and Biostatistics Core, Centers of Genomic and Precision Medicine, National Taiwan University, Taipei, Taiwan
| | - Liang-Chuan Lai
- Graduate Institute of Physiology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Tzu-Pin Lu
- Department of Public Health, National Taiwan University, Taipei, Taiwan
| | - Yung-Hua Li
- Graduate Institute of Biomedical Electronics and Bioinformatics, National Taiwan University, Taipei, Taiwan
| | - Mong-Hsun Tsai
- Institute of Biotechnology, National Taiwan University, Taipei, Taiwan.
| | - Eric Y Chuang
- Graduate Institute of Biomedical Electronics and Bioinformatics, National Taiwan University, Taipei, Taiwan. .,Bioinformatics and Biostatistics Core, Centers of Genomic and Precision Medicine, National Taiwan University, Taipei, Taiwan. .,Department of Electrical Engineering, National Taiwan University, Taipei, Taiwan. .,Master Program for Biomedical Engineering, China Medical University, Taichung, Taiwan.
| |
Collapse
|
83
|
Mahdi H, Hafez N, Doroshow D, Sohal D, Keedy V, Do KT, LoRusso P, Jürgensmeier J, Avedissian M, Sklar J, Glover C, Felicetti B, Dean E, Mortimer P, Shapiro GI, Eder JP. Ceralasertib-Mediated ATR Inhibition Combined With Olaparib in Advanced Cancers Harboring DNA Damage Response and Repair Alterations (Olaparib Combinations). JCO Precis Oncol 2021; 5:PO.20.00439. [PMID: 34527850 PMCID: PMC8437220 DOI: 10.1200/po.20.00439] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 05/14/2021] [Accepted: 07/27/2021] [Indexed: 01/09/2023] Open
Abstract
Poly (ADP-ribose) polymerase (PARP) inhibitors have emerged as promising therapy in cancers with homologous recombination repair deficiency. However, efficacy is limited by both intrinsic and acquired resistance. The Olaparib Combinations basket trial explored olaparib alone and in combination with other homologous recombination-directed targeted therapies. Here, we report the results of the arm in which olaparib was combined with the orally bioavailable ataxia telangiectasia and RAD3-related inhibitor ceralasertib in patients with relapsed or refractory cancers harboring DNA damage response and repair alterations, including patients with BRCA-mutated PARP inhibitor-resistant high-grade serous ovarian cancer (HGSOC). PATIENTS AND METHODS Germline and somatic mutations had to be deleterious by COSMIC or ClinVar for eligibility. Olaparib was administered at 300 mg twice daily and ceralasertib at 160 mg daily on days 1-7 in 28-day cycles until progression or unacceptable toxicities. Primary end points were confirmed complete response (CR) or partial response (PR) rates and clinical benefit rate (CBR; CR + PR + stable disease [SD] at 16 weeks). RESULTS Twenty-five patients were enrolled, with median four prior therapies. Five patients required dose reductions for myelosuppression. Overall response rate was 8.3% and CBR was 62.5% among the entire cohort. Two of five patients with tumor harboring ATM mutation achieved CR or SD ongoing at 24+ months, respectively (CBR 40%). Of seven patients with PARP inhibitor-resistant HGSOC, one achieved PR (-90%) and five had SD ranging 16-72 weeks (CBR 86%). CONCLUSION Olaparib with ceralasertib demonstrated preliminary activity in ATM-mutated tumors and in PARP inhibitor-resistant BRCA1/2-mutated HGSOC. These data warrant additional studies to further confirm activity in these settings.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Colin Glover
- Oncology Early Clinical Projects, Oncology R&D, AstraZeneca, Cambridge, United Kingdom
| | - Brunella Felicetti
- Oncology Early Clinical Projects, Oncology R&D, AstraZeneca, Cambridge, United Kingdom
| | - Emma Dean
- Oncology Early Clinical Projects, Oncology R&D, AstraZeneca, Cambridge, United Kingdom
| | - Peter Mortimer
- Oncology Early Clinical Projects, Oncology R&D, AstraZeneca, Cambridge, United Kingdom
| | | | | |
Collapse
|
84
|
van de Haar J, Hoes LR, Roepman P, Lolkema MP, Verheul HMW, Gelderblom H, de Langen AJ, Smit EF, Cuppen E, Wessels LFA, Voest EE. Limited evolution of the actionable metastatic cancer genome under therapeutic pressure. Nat Med 2021; 27:1553-1563. [PMID: 34373653 DOI: 10.1038/s41591-021-01448-w] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Accepted: 06/23/2021] [Indexed: 11/08/2022]
Abstract
Genomic profiling is critical for the identification of treatment options for patients with metastatic cancer, but it remains unclear how frequently this procedure should be repeated during the course of the disease. To address this, we analyzed whole-genome sequencing (WGS) data of 250 biopsy pairs, longitudinally collected over the treatment course of 231 adult patients with a representative variety of metastatic solid malignancies. Within the biopsy interval (median, 6.4 months), patients received one or multiple lines of (mostly) standard-of-care (SOC) treatments, with all major treatment modalities being broadly represented. SOC biomarkers and biomarkers for clinical trial enrollment could be identified in 23% and 72% of biopsies, respectively. For SOC genomic biomarkers, we observed full concordance between the first and the second biopsy in 99% of pairs. Of the 219 biomarkers for clinical trial enrollment that were identified in the first biopsies, we recovered 94% in the follow-up biopsies. Furthermore, a second WGS analysis did not identify additional biomarkers for clinical trial enrollment in 91% of patients. More-frequent genomic evolution was observed when considering specific genes targeted by small-molecule inhibitors or hormonal therapies (21% and 22% of cases, respectively). Together, our data demonstrate that there is limited evolution of the actionable genome of treated metastases. A single WGS analysis of a metastatic biopsy is generally sufficient to identify SOC genomic biomarkers and to identify investigational treatment opportunities.
Collapse
Affiliation(s)
- Joris van de Haar
- Division of Molecular Oncology & Immunology, Netherlands Cancer Institute, Amsterdam, the Netherlands
- Division of Molecular Carcinogenesis, The Netherlands Cancer Institute, Amsterdam, the Netherlands
- Oncode Institute, Amsterdam, the Netherlands
| | - Louisa R Hoes
- Division of Molecular Oncology & Immunology, Netherlands Cancer Institute, Amsterdam, the Netherlands
- Oncode Institute, Amsterdam, the Netherlands
| | - Paul Roepman
- Hartwig Medical Foundation, Amsterdam, the Netherlands
| | - Martijn P Lolkema
- Department of Medical Oncology, Erasmus Medical Center Cancer Institute, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Henk M W Verheul
- Department of Medical Oncology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Hans Gelderblom
- Department of Medical Oncology, Leiden University Medical Center, Leiden, the Netherlands
| | - Adrianus J de Langen
- Department of Thoracic Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Egbert F Smit
- Department of Thoracic Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Edwin Cuppen
- Oncode Institute, Amsterdam, the Netherlands
- Hartwig Medical Foundation, Amsterdam, the Netherlands
- Center for Molecular Medicine, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Lodewyk F A Wessels
- Division of Molecular Carcinogenesis, The Netherlands Cancer Institute, Amsterdam, the Netherlands
- Oncode Institute, Amsterdam, the Netherlands
- Faculty of EEMCS, Delft University of Technology, Delft, the Netherlands
| | - Emile E Voest
- Division of Molecular Oncology & Immunology, Netherlands Cancer Institute, Amsterdam, the Netherlands.
- Oncode Institute, Amsterdam, the Netherlands.
- Center for Personalized Cancer Treatment, Rotterdam, the Netherlands.
| |
Collapse
|
85
|
Moretto R, Elliott A, Zhang J, Arai H, Germani MM, Conca V, Xiu J, Stafford P, Oberley M, Abraham J, Spetzler D, Rossini D, Antoniotti C, Marshall J, Shields A, Lopes G, Lonardi S, Pietrantonio F, Tomasello G, Passardi A, Tamburini E, Santini D, Aprile G, Masi G, Falcone A, Lenz HJ, Korn M, Cremolini C. Homologous Recombination Deficiency Alterations in Colorectal Cancer: Clinical, Molecular, and Prognostic Implications. J Natl Cancer Inst 2021; 114:271-279. [PMID: 34469533 DOI: 10.1093/jnci/djab169] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 06/10/2021] [Accepted: 07/27/2021] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Tumors with homologous recombination (HR) deficiency (HRD) show high sensitivity to platinum salts and PARP-inhibitors in several malignancies. In colorectal cancer (CRC), the role of HRD alterations is mostly unknown. METHODS Next generation sequencing, whole transcriptome sequencing and whole exome sequencing were conducted using CRC samples submitted to a commercial Clinical Laboratory Improvement Amendments (CLIA) certified laboratory. Tumors with pathogenic/presumed pathogenic mutations in 33 genes involved in the HR pathway were considered HRD, the others HR proficient (HRP). Furthermore, tumor samples from patients enrolled in the phase III TRIBE2 study comparing upfront FOLFOXIRI/bevacizumab versus FOLFOX/bevacizumab were analyzed with next generation sequencing. The analyses were separately conducted in microsatellite stable/proficient mismatch repair (MSS/pMMR) and microsatellite instable-high/deficient mismatch repair (MSI-H/dMMR) groups. All statistical tests were 2-sided. RESULTS Of 9321 CRC tumors, 1270 (13.6%) and 8051 (86.4%) were HRD and HRP, respectively. HRD tumors were more frequent among MSI-H/dMMR than MSS/pMMR tumors (73.4% vs 9.5%, p and q < 0.001). In MSS/pMMR group, HRD tumors were more frequently tumor mutational burden high (8.1% vs 2.2% P and q < 0.001) and PD-L1 positive (5.0% vs 2.4%, P and q = 0.001), enriched in all immune cell and fibroblast populations, and genomic loss of heterozygosity-high (16.2% vs 9.5%, P = .03). In the TRIBE2 study, patients with MSS/pMMR and HRD tumors (10.7%) showed longer overall survival compared to MSS/pMMR and HRP ones (40.2 vs 23.8 months; hazard ratio = 0.66; 95% confidence interval = 0.45-0.98, P = .04). Consistent results were reported in the multivariable model (hazard ratio = 0.67; 95% confidence ratio = 0.45-1.02, P = .07). No interaction effect was evident between HR groups and treatment arm. CONCLUSIONS HRD tumors are a distinctive subgroup of MSS/pMMR CRCs with specific molecular and prognostic characteristics. The potential efficacy of agents targeting the HR system and immune check-point inhibitors in this subgroup is worth of clinical investigation.
Collapse
Affiliation(s)
- Roberto Moretto
- Unit of Medical Oncology 2, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy
| | - Andrew Elliott
- Clinical & Translational Research, Medical Affairs, Caris Life Sciences, Phoenix, AZ, USA
| | - Jian Zhang
- Clinical & Translational Research, Medical Affairs, Caris Life Sciences, Phoenix, AZ, USA
| | - Hiroyuki Arai
- Division of Medical Oncology, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, USA
| | - Marco Maria Germani
- Unit of Medical Oncology 2, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy.,Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Veronica Conca
- Unit of Medical Oncology 2, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy.,Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Joanne Xiu
- Clinical & Translational Research, Medical Affairs, Caris Life Sciences, Phoenix, AZ, USA
| | - Phillip Stafford
- Clinical & Translational Research, Medical Affairs, Caris Life Sciences, Phoenix, AZ, USA
| | - Matthew Oberley
- Clinical & Translational Research, Medical Affairs, Caris Life Sciences, Phoenix, AZ, USA
| | - Jim Abraham
- Clinical & Translational Research, Medical Affairs, Caris Life Sciences, Phoenix, AZ, USA
| | - David Spetzler
- Clinical & Translational Research, Medical Affairs, Caris Life Sciences, Phoenix, AZ, USA
| | - Daniele Rossini
- Unit of Medical Oncology 2, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy.,Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Carlotta Antoniotti
- Unit of Medical Oncology 2, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy.,Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - John Marshall
- Division of Hematology/Oncology, Ruesch Center for The Cure of Gastrointestinal Cancers, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, D.C, ., USA
| | - Anthony Shields
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, USA
| | - Gilberto Lopes
- Division of Medical Oncology, Department of Medicine, Miller School of Medicine, University of Miami, Sylvester Comprehensive Cancer Center, Miami, FL, USA
| | - Sara Lonardi
- Early Phase Clinical Trial Unit, Department of Oncology, Veneto Institute of Oncology IOV-IRCCS, Padua, Italy.,Medical Oncology Unit 1, Department of Oncology, Veneto Institute of Oncology IOV-IRCCS, Padua, Italy
| | - Filippo Pietrantonio
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy.,Oncology and Hemato-oncology Department, University of Milan, Milan, Italy
| | - Gianluca Tomasello
- Oncology Unit, Oncology Department, ASST of Cremona, Cremona, Italy.,UOC Medical Oncology, IRCCS Foundation Ca' Granda Maggiore Hospital Policlinic, Milan, Italy
| | - Alessandro Passardi
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Emiliano Tamburini
- Department of Oncology and Palliative Care, Cardinale G Panico, Tricase City Hospital, Tricase, Italy
| | - Daniele Santini
- Department of Medical Oncology, University Campus Biomedico, Rome, Italy
| | - Giuseppe Aprile
- Department of Oncology, San Bortolo General Hospital, Vicenza, Italy
| | - Gianluca Masi
- Unit of Medical Oncology 2, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy.,Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Alfredo Falcone
- Unit of Medical Oncology 2, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy.,Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Heinz-Josef Lenz
- Division of Medical Oncology, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, USA
| | - Michael Korn
- Clinical & Translational Research, Medical Affairs, Caris Life Sciences, Phoenix, AZ, USA
| | - Chiara Cremolini
- Unit of Medical Oncology 2, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy.,Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| |
Collapse
|
86
|
Matis TS, Zayed N, Labraki B, de Ladurantaye M, Matis TA, Camacho Valenzuela J, Hamel N, Atayan A, Rivera B, Tabach Y, Tonin PN, Orthwein A, Mes-Masson AM, El Haffaf Z, Foulkes WD, Polak P. Current gene panels account for nearly all homologous recombination repair-associated multiple-case breast cancer families. NPJ Breast Cancer 2021; 7:109. [PMID: 34433815 PMCID: PMC8387362 DOI: 10.1038/s41523-021-00315-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Accepted: 07/26/2021] [Indexed: 12/11/2022] Open
Abstract
It was hypothesized that variants in underexplored homologous recombination repair (HR) genes could explain unsolved multiple-case breast cancer (BC) families. We investigated HR deficiency (HRD)-associated mutational signatures and second hits in tumor DNA from familial BC cases. No candidates genes were associated with HRD in 38 probands previously tested negative with gene panels. We conclude it is unlikely that unknown HRD-associated genes explain a large fraction of unsolved familial BC.
Collapse
Affiliation(s)
- Thibaut S Matis
- Department of Human Genetics, McGill University, Montreal, QC, Canada
- Cancer Research Program, Centre for Translational Biology, The Research Institute of the McGill University Health Centre, Montreal, QC, Canada
- Cancer Genetics Unit, Institut Bergonié, Bordeaux, France
| | - Nadia Zayed
- Department of Human Genetics, McGill University, Montreal, QC, Canada
- Cancer Research Program, Centre for Translational Biology, The Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | | | - Manon de Ladurantaye
- Centre de recherche du Centre hospitalier de l'Université de Montréal and Institut du cancer de Montréal, Montreal, Quebec, Canada
| | | | - José Camacho Valenzuela
- Department of Human Genetics, McGill University, Montreal, QC, Canada
- Cancer Research Program, Centre for Translational Biology, The Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Nancy Hamel
- Cancer Research Program, Centre for Translational Biology, The Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Adrienne Atayan
- Department of Human Genetics, McGill University, Montreal, QC, Canada
| | - Barbara Rivera
- Gerald Bronfman Department of Oncology, McGill University, Montreal, QC, Canada
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, QC, Canada
- IDIBELL Bellvitge Biomedical Research Institute, Barcelona, Spain
| | - Yuval Tabach
- Department of Developmental Biology and Cancer Research, Institute for Medical Research-Israel-Canada, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Patricia N Tonin
- Department of Human Genetics, McGill University, Montreal, QC, Canada
- Cancer Research Program, Centre for Translational Biology, The Research Institute of the McGill University Health Centre, Montreal, QC, Canada
- Department of Medicine, McGill University, Montréal, QC, Canada
| | - Alexandre Orthwein
- Gerald Bronfman Department of Oncology, McGill University, Montreal, QC, Canada
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, QC, Canada
| | - Anne-Marie Mes-Masson
- Centre de recherche du Centre hospitalier de l'Université de Montréal and Institut du cancer de Montréal, Montreal, Quebec, Canada
- Department of Medicine, Université de Montréal, Montreal, QC, Canada
| | - Zaki El Haffaf
- Divison de Médecine Génétique, CRCHUM, Montreal, QC, Canada
| | - William D Foulkes
- Department of Human Genetics, McGill University, Montreal, QC, Canada.
- Cancer Research Program, Centre for Translational Biology, The Research Institute of the McGill University Health Centre, Montreal, QC, Canada.
- Gerald Bronfman Department of Oncology, McGill University, Montreal, QC, Canada.
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, QC, Canada.
| | - Paz Polak
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
87
|
Bakare OO, Gokul A, Wu R, Niekerk LA, Klein A, Keyster M. Biomedical Relevance of Novel Anticancer Peptides in the Sensitive Treatment of Cancer. Biomolecules 2021; 11:1120. [PMID: 34439786 PMCID: PMC8394746 DOI: 10.3390/biom11081120] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2021] [Revised: 07/21/2021] [Accepted: 07/24/2021] [Indexed: 12/14/2022] Open
Abstract
The global increase in cancer mortality and economic losses necessitates the cautious quest for therapeutic agents with compensatory advantages over conventional therapies. Anticancer peptides (ACPs) are a subset of host defense peptides, also known as antimicrobial peptides, which have emerged as therapeutic and diagnostic candidates due to several compensatory advantages over the non-specificity of the current treatment regimens. This review aimed to highlight the ravaging incidence of cancer, the use of ACPs in cancer treatment with their mechanisms, ACP discovery and delivery methods, and the limitations for their use. This would create awareness for identifying more ACPs with better specificity, accuracy and sensitivity towards the disease. It would also promote their efficacious utilization in biotechnology, medical sciences and molecular biology to ease the severity of the disease and enable the patients living with these conditions to develop an accommodating lifestyle.
Collapse
Affiliation(s)
- Olalekan Olanrewaju Bakare
- Environmental Biotechnology Laboratory, Department of Biotechnology, University of the Western Cape, Bellville 7535, South Africa; (R.W.); (L.-A.N.)
| | - Arun Gokul
- Department of Plant Sciences, Qwaqwa Campus, University of the Free State, Phuthaditjhaba 9866, South Africa;
| | - Ruomou Wu
- Environmental Biotechnology Laboratory, Department of Biotechnology, University of the Western Cape, Bellville 7535, South Africa; (R.W.); (L.-A.N.)
| | - Lee-Ann Niekerk
- Environmental Biotechnology Laboratory, Department of Biotechnology, University of the Western Cape, Bellville 7535, South Africa; (R.W.); (L.-A.N.)
| | - Ashwil Klein
- Plant Omics Laboratory, Department of Biotechnology, University of the Western Cape, Bellville 7535, South Africa;
| | - Marshall Keyster
- Environmental Biotechnology Laboratory, Department of Biotechnology, University of the Western Cape, Bellville 7535, South Africa; (R.W.); (L.-A.N.)
| |
Collapse
|
88
|
Oshi M, Kawaguchi T, Yan L, Peng X, Qi Q, Tian W, Schulze A, McDonald KA, Narayanan S, Young J, Liu S, Morris LGT, Chan TA, Kalinski P, Matsuyama R, Otsuji E, Endo I, Takabe K. Immune cytolytic activity is associated with reduced intra-tumoral genetic heterogeneity and with better clinical outcomes in triple negative breast cancer. Am J Cancer Res 2021; 11:3628-3644. [PMID: 34354864 PMCID: PMC8332854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Accepted: 04/14/2021] [Indexed: 06/13/2023] Open
Abstract
Evaluation of the functional aspects if the tumor immune microenvironment (TIME), such as the recently introduced cytolytic activity score (CYT) index have been under the spotlight in cancer research; however, clinical relevance of immune cell killing activity in breast cancer has never been analyzed in large patient cohorts. We hypothesized that CYT reflects the immune activity of TIME and can predict patient survival. A total of 7533 breast cancer patients were analyzed as both discovery and validation cohorts. We found that high CYT was associated with advanced histological grade and triple-negative breast cancer (TNBC). High CYT in tumors was significantly associated with better survival in TNBC, but unexpectedly, not in other breast cancer subtypes. High CYT TNBC included both favorable immune-related, as well as unfavorable (suppressive) inflammation-related gene sets, and characterized by high infiltration with T cells and B cells. High CYT TNBC was associated with high homologous recombination deficiency and low somatic copy number alteration score and less mutant allele tumor heterogeneity, but not with tumor mutation burden (TMB). Although CYT was not associated with pathological complete response after neoadjuvant chemotherapy, it was significantly associated with high expression of multiple immune checkpoint molecules. In conclusion, CYT of TNBC is associated with enhanced anti-cancer immunity, less intra-tumoral heterogeneity, and with better survival.
Collapse
Affiliation(s)
- Masanori Oshi
- Department of Surgical Oncology, Roswell Park Cancer InstituteBuffalo, NY 14263, USA
- Department of Gastroenterological Surgery, Yokohama City University Graduate School of MedicineYokohama, Kanagawa, 236-0004, Japan
| | - Tsutomu Kawaguchi
- Department of Surgical Oncology, Roswell Park Cancer InstituteBuffalo, NY 14263, USA
- Department of Surgery, Kyoto Prefectural University of MedicineKyoto, Japan
| | - Li Yan
- Department of Biostatistics & Bioinformatics, Roswell Park Cancer InstituteBuffalo, NY 14263, USA
| | - Xuan Peng
- Department of Biostatistics & Bioinformatics, Roswell Park Cancer InstituteBuffalo, NY 14263, USA
| | - Qianya Qi
- Department of Biostatistics & Bioinformatics, Roswell Park Cancer InstituteBuffalo, NY 14263, USA
| | - Wanqing Tian
- Department of Biostatistics & Bioinformatics, Roswell Park Cancer InstituteBuffalo, NY 14263, USA
| | - Amy Schulze
- Department of Surgical Oncology, Roswell Park Cancer InstituteBuffalo, NY 14263, USA
| | - Kerry-Ann McDonald
- Department of Surgical Oncology, Roswell Park Cancer InstituteBuffalo, NY 14263, USA
| | - Sumana Narayanan
- Department of Surgical Oncology, Roswell Park Cancer InstituteBuffalo, NY 14263, USA
| | - Jessica Young
- Department of Surgical Oncology, Roswell Park Cancer InstituteBuffalo, NY 14263, USA
- Department of Surgery, University at Buffalo, The State University of New York Jacobs School of Medicine and Biomedical SciencesBuffalo, NY 14263, USA
| | - Song Liu
- Department of Biostatistics & Bioinformatics, Roswell Park Cancer InstituteBuffalo, NY 14263, USA
| | - Luc GT Morris
- Immunogenomics and Precision Oncology Platform, Memorial Sloan Kettering Cancer CenterNew York, NY 10065, USA
- Department of Surgery, Memorial Sloan Kettering Cancer CenterNew York, NY 10065, USA
| | - Timothy A Chan
- Immunogenomics and Precision Oncology Platform, Memorial Sloan Kettering Cancer CenterNew York, NY 10065, USA
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer CenterNew York, NY 10065, USA
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer CenterNew York, NY 10065, USA
- Center for Immunotherapy and Precision Immuno-Oncology, Cleveland ClinicCleveland, OH USA
- Lerner Research Institute and Taussig Cancer Center, Cleveland ClinicCleveland, OH, USA
| | - Pawel Kalinski
- Department of Medicine and Center for Immunotherapy, Roswell Park Cancer InstituteBuffalo, NY, USA
| | - Ryusei Matsuyama
- Department of Gastroenterological Surgery, Yokohama City University Graduate School of MedicineYokohama, Kanagawa, 236-0004, Japan
| | - Eigo Otsuji
- Department of Surgery, Kyoto Prefectural University of MedicineKyoto, Japan
| | - Itaru Endo
- Department of Gastroenterological Surgery, Yokohama City University Graduate School of MedicineYokohama, Kanagawa, 236-0004, Japan
| | - Kazuaki Takabe
- Department of Surgical Oncology, Roswell Park Cancer InstituteBuffalo, NY 14263, USA
- Department of Gastroenterological Surgery, Yokohama City University Graduate School of MedicineYokohama, Kanagawa, 236-0004, Japan
- Department of Surgery, University at Buffalo, The State University of New York Jacobs School of Medicine and Biomedical SciencesBuffalo, NY 14263, USA
- Department of Surgery, Niigata University Graduate School of Medical and Dental SciencesNiigata, Japan
- Department of Breast Surgery and Oncology, Tokyo Medical UniversityTokyo, Japan
- Department of Breast Surgery, Fukushima Medical UniversityFukushima, Japan
| |
Collapse
|
89
|
Setton J, Zinda M, Riaz N, Durocher D, Zimmermann M, Koehler M, Reis-Filho JS, Powell SN. Synthetic Lethality in Cancer Therapeutics: The Next Generation. Cancer Discov 2021; 11:1626-1635. [PMID: 33795234 PMCID: PMC8295179 DOI: 10.1158/2159-8290.cd-20-1503] [Citation(s) in RCA: 112] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 01/01/2021] [Accepted: 02/23/2021] [Indexed: 12/25/2022]
Abstract
Synthetic lethality (SL) provides a conceptual framework for tackling targets that are not classically "druggable," including loss-of-function mutations in tumor suppressor genes required for carcinogenesis. Recent technological advances have led to an inflection point in our understanding of genetic interaction networks and ability to identify a wide array of novel SL drug targets. Here, we review concepts and lessons emerging from first-generation trials aimed at testing SL drugs, discuss how the nature of the targeted lesion can influence therapeutic outcomes, and highlight the need to develop clinical biomarkers distinct from those based on the paradigms developed to target activated oncogenes. SIGNIFICANCE: SL offers an approach for the targeting of loss of function of tumor suppressor and DNA repair genes, as well as of amplification and/or overexpression of genes that cannot be targeted directly. A next generation of tumor-specific alterations targetable through SL has emerged from high-throughput CRISPR technology, heralding not only new opportunities for drug development, but also important challenges in the development of optimal predictive biomarkers.
Collapse
Affiliation(s)
- Jeremy Setton
- Memorial Sloan Kettering Cancer Center, New York, New York
| | | | - Nadeem Riaz
- Memorial Sloan Kettering Cancer Center, New York, New York
| | - Daniel Durocher
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | | | | | | | - Simon N Powell
- Memorial Sloan Kettering Cancer Center, New York, New York.
| |
Collapse
|
90
|
Zhou Q, Howard ME, Tu X, Zhu Q, Denbeigh JM, Remmes NB, Herman MG, Beltran CJ, Yuan J, Greipp PT, Boughey JC, Wang L, Johnson N, Goetz MP, Sarkaria JN, Lou Z, Mutter RW. Inhibition of ATM Induces Hypersensitivity to Proton Irradiation by Upregulating Toxic End Joining. Cancer Res 2021; 81:3333-3346. [PMID: 33597272 PMCID: PMC8260463 DOI: 10.1158/0008-5472.can-20-2960] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 12/30/2020] [Accepted: 02/11/2021] [Indexed: 12/15/2022]
Abstract
Proton Bragg peak irradiation has a higher ionizing density than conventional photon irradiation or the entrance of the proton beam profile. Whether targeting the DNA damage response (DDR) could enhance vulnerability to the distinct pattern of damage induced by proton Bragg peak irradiation is currently unknown. Here, we performed genetic or pharmacologic manipulation of key DDR elements and evaluated DNA damage signaling, DNA repair, and tumor control in cell lines and xenografts treated with the same physical dose across a radiotherapy linear energy transfer spectrum. Radiotherapy consisted of 6 MV photons and the entrance beam or Bragg peak of a 76.8 MeV spot scanning proton beam. More complex DNA double-strand breaks (DSB) induced by Bragg peak proton irradiation preferentially underwent resection and engaged homologous recombination (HR) machinery. Unexpectedly, the ataxia-telangiectasia mutated (ATM) inhibitor, AZD0156, but not an inhibitor of ATM and Rad3-related, rendered cells hypersensitive to more densely ionizing proton Bragg peak irradiation. ATM inhibition blocked resection and shunted more DSBs to processing by toxic ligation through nonhomologous end-joining, whereas loss of DNA ligation via XRCC4 or Lig4 knockdown rescued resection and abolished the enhanced Bragg peak cell killing. Proton Bragg peak monotherapy selectively sensitized cell lines and tumor xenografts with inherent HR defects, and the repair defect induced by ATM inhibitor coadministration showed enhanced efficacy in HR-proficient models. In summary, inherent defects in HR or administration of an ATM inhibitor in HR-proficient tumors selectively enhances the relative biological effectiveness of proton Bragg peak irradiation. SIGNIFICANCE: Coadministration of an ATM inhibitor rewires DNA repair machinery to render cancer cells uniquely hypersensitive to DNA damage induced by the proton Bragg peak, which is characterized by higher density ionization.See related commentary by Nickoloff, p. 3156.
Collapse
Affiliation(s)
- Qin Zhou
- Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota
| | | | - Xinyi Tu
- Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota
| | - Qian Zhu
- Department of Oncology, Mayo Clinic, Rochester, Minnesota
| | - Janet M Denbeigh
- Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota
| | | | - Michael G Herman
- Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota
| | - Chris J Beltran
- Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota
| | - Jian Yuan
- Department of Oncology, Mayo Clinic, Rochester, Minnesota
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota
| | - Patricia T Greipp
- Division of Laboratory Genetics and Genomics, Mayo Clinic, Rochester, Minnesota
| | - Judy C Boughey
- Department of Surgery, Mayo Clinic, Rochester, Minnesota
| | - Liewei Wang
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota
| | - Neil Johnson
- Molecular Therapeutics Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - Matthew P Goetz
- Division of Medical Oncology, Mayo Clinic, Rochester, Minnesota
| | - Jann N Sarkaria
- Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota
| | - Zhenkun Lou
- Department of Oncology, Mayo Clinic, Rochester, Minnesota.
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota
| | - Robert W Mutter
- Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota.
| |
Collapse
|
91
|
Gussow AB, Koonin EV, Auslander N. Identification of combinations of somatic mutations that predict cancer survival and immunotherapy benefit. NAR Cancer 2021; 3:zcab017. [PMID: 34027407 PMCID: PMC8127965 DOI: 10.1093/narcan/zcab017] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 04/18/2021] [Accepted: 04/28/2021] [Indexed: 11/14/2022] Open
Abstract
Cancer evolves through the accumulation of somatic mutations over time. Although several methods have been developed to characterize mutational processes in cancers, these have not been specifically designed to identify mutational patterns that predict patient prognosis. Here we present CLICnet, a method that utilizes mutational data to cluster patients by survival rate. CLICnet employs Restricted Boltzmann Machines, a type of generative neural network, which allows for the capture of complex mutational patterns associated with patient survival in different cancer types. For some cancer types, clustering produced by CLICnet also predicts benefit from anti-PD1 immune checkpoint blockade therapy, whereas for other cancer types, the mutational processes associated with survival are different from those associated with the improved anti-PD1 survival benefit. Thus, CLICnet has the ability to systematically identify and catalogue combinations of mutations that predict cancer survival, unveiling intricate associations between mutations, survival, and immunotherapy benefit.
Collapse
Affiliation(s)
- Ayal B Gussow
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, MD 20894, USA
| | - Eugene V Koonin
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, MD 20894, USA
| | - Noam Auslander
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, MD 20894, USA
| |
Collapse
|
92
|
Arai H, Elliott A, Xiu J, Wang J, Battaglin F, Kawanishi N, Soni S, Zhang W, Millstein J, Sohal D, Goldberg RM, Hall MJ, Scott AJ, Khushman M, Hwang JJ, Lou E, Weinberg BA, Marshall JL, Lockhart AC, Stafford P, Zhang J, Moretto R, Cremolini C, Korn WM, Lenz HJ. The Landscape of Alterations in DNA Damage Response Pathways in Colorectal Cancer. Clin Cancer Res 2021; 27:3234-3242. [PMID: 33766816 PMCID: PMC12047448 DOI: 10.1158/1078-0432.ccr-20-3635] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 01/13/2021] [Accepted: 03/22/2021] [Indexed: 11/16/2022]
Abstract
PURPOSE Defective DNA damage response (DDR) is a hallmark of cancer leading to genomic instability and is associated with chemosensitivity. Although the mismatch repair system has been extensively studied, the clinical implications of other mechanisms associated with DDR alterations in patients with colorectal cancer remain unclear. This study aimed to understand DDR pathways alterations and their association with common clinical features in patients with colorectal cancer. EXPERIMENTAL DESIGN Next-generation sequencing and whole-transcriptome sequencing were conducted using formalin-fixed paraffin-embedded samples submitted to a commercial Clinical Laboratory Improvement Amendments-certified laboratory. Samples with pathogenic or presumed pathogenic mutations in 29 specific DDR-related genes were considered as DDR-mutant (DDR-MT) and the remaining samples as DDR-wild type (DDR-WT). RESULTS Of 9,321 patients with colorectal cancer, 1,290 (13.8%) were DDR-MT. The frequency of DDR-MT was significantly higher in microsatellite instability-high (MSI-H) cases than in microsatellite stable cases (76.4% vs. 9.5%). The DDR-MT genotype was higher in the right-sided, RAS-wild, BRAF-mutant, and CMS1 subgroups. However, these associations were primarily confounded by the distribution of MSI status. Compared with the DDR-WT tumors, the DDR-MT tumors had a higher mutational burden and gene expression levels in the immune-related pathway, which were independent of MSI status. CONCLUSIONS We characterized a distinct subgroup of patients with colorectal cancer with tumors harboring mutations in the DDR-related genes. These patients more commonly had MSI-H tumors and exhibited an activated immune signature regardless of their tumor's MSI status. These findings warrant further investigations to develop personalized treatment strategies in this significant subgroup of patients with colorectal cancer.
Collapse
Affiliation(s)
- Hiroyuki Arai
- Division of Medical Oncology, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, California
| | - Andrew Elliott
- Clinical and Translational Research, Medical Affairs, Caris Life Sciences, Phoenix, Arizona
| | - Joanne Xiu
- Clinical and Translational Research, Medical Affairs, Caris Life Sciences, Phoenix, Arizona
| | - Jingyuan Wang
- Division of Medical Oncology, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, California
| | - Francesca Battaglin
- Division of Medical Oncology, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, California
| | - Natsuko Kawanishi
- Division of Medical Oncology, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, California
| | - Shivani Soni
- Division of Medical Oncology, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, California
| | - Wu Zhang
- Division of Medical Oncology, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, California
| | - Joshua Millstein
- Department of Preventive Medicine, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, California
| | - Davendra Sohal
- Division of Hematology/Oncology, University of Cincinnati, Cincinnati, Ohio
| | | | - Michael J Hall
- Department of Clinical Genetics, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - Aaron J Scott
- Department of Medicine, University of Arizona Cancer Center, Tucson, Arizona
| | - Moh'd Khushman
- Medical Oncology, Mitchell Cancer Institute, The University of South Alabama, Mobile, Alabama
| | - Jimmy J Hwang
- Department of Solid Tumor Oncology, GI Medical Oncology, Levine Cancer Institute, Charlotte, North Carolina
| | - Emil Lou
- Division of Hematology, Oncology and Transplantation, University of Minnesota, Minneapolis, Minnesota
| | - Benjamin A Weinberg
- Ruesch Center for the Cure of Gastrointestinal Cancers, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC
| | - John L Marshall
- Ruesch Center for the Cure of Gastrointestinal Cancers, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC
| | - Albert C Lockhart
- Department of Medicine, Division of Oncology, University of Miami, Miller School of Medicine, Miami, Florida
| | - Phillip Stafford
- Department of Bioinformatics, Caris Life Sciences, Phoenix, Arizona
| | - Jian Zhang
- Department of Bioinformatics, Caris Life Sciences, Phoenix, Arizona
| | - Roberto Moretto
- Unit of Medical Oncology 2, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy
| | - Chiara Cremolini
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | | | - Heinz-Josef Lenz
- Division of Medical Oncology, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, California.
| |
Collapse
|
93
|
Bååth M, Jönsson JM, Westbom Fremer S, Martín de la Fuente L, Tran L, Malander S, Kannisto P, Måsbäck A, Honeth G, Hedenfalk I. MET Expression and Cancer Stem Cell Networks Impact Outcome in High-Grade Serous Ovarian Cancer. Genes (Basel) 2021; 12:742. [PMID: 34069138 PMCID: PMC8155853 DOI: 10.3390/genes12050742] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 05/06/2021] [Accepted: 05/12/2021] [Indexed: 01/08/2023] Open
Abstract
Overexpression of the receptor tyrosine kinase MET has been linked to poor survival in several cancer types, and MET has been suggested to interact with stem cell networks. In vitro studies have further suggested a possible benefit of a combined treatment using PARP and MET inhibitors. We used a tissue microarray (TMA) with 130 samples of advanced-stage high-grade serous fallopian tube/ovarian cancer (HGSC) to investigate the prognostic value of MET protein expression alone and in combination with the stem cell factor SOX2. The possible synergistic effects of a PARP and MET inhibitor treatment were evaluated in two cell lines with BRCA1 or BRCA2 deficiency and in their BRCA1/2-proficient counterparts. Patients with tumors positive for MET had worse overall survival (log-rank test, p = 0.015) compared to patients with MET-negative tumors. The prognostic role of MET was even more prominent in the subgroup of patients with SOX2-negative tumors (p = 0.0081). No synergistic effects of the combined treatment with PARP and MET inhibitors were found in the cell lines examined. We conclude that MET expression could be used as a marker for OS in HGSC and that stemness should be taken into consideration when evaluating the mechanisms of this effect.
Collapse
Affiliation(s)
- Maria Bååth
- Division of Oncology, Department of Clinical Sciences Lund, Lund University and Skåne University Hospital, 223 81 Lund, Sweden; (M.B.); (J.-M.J.); (S.W.F.); (L.M.d.l.F.); (L.T.); (S.M.); (G.H.)
| | - Jenny-Maria Jönsson
- Division of Oncology, Department of Clinical Sciences Lund, Lund University and Skåne University Hospital, 223 81 Lund, Sweden; (M.B.); (J.-M.J.); (S.W.F.); (L.M.d.l.F.); (L.T.); (S.M.); (G.H.)
| | - Sofia Westbom Fremer
- Division of Oncology, Department of Clinical Sciences Lund, Lund University and Skåne University Hospital, 223 81 Lund, Sweden; (M.B.); (J.-M.J.); (S.W.F.); (L.M.d.l.F.); (L.T.); (S.M.); (G.H.)
- Department of Surgical Pathology, Division of Laboratory Medicine, Skåne University Hospital, 222 42 Lund, Sweden;
| | - Laura Martín de la Fuente
- Division of Oncology, Department of Clinical Sciences Lund, Lund University and Skåne University Hospital, 223 81 Lund, Sweden; (M.B.); (J.-M.J.); (S.W.F.); (L.M.d.l.F.); (L.T.); (S.M.); (G.H.)
| | - Lena Tran
- Division of Oncology, Department of Clinical Sciences Lund, Lund University and Skåne University Hospital, 223 81 Lund, Sweden; (M.B.); (J.-M.J.); (S.W.F.); (L.M.d.l.F.); (L.T.); (S.M.); (G.H.)
| | - Susanne Malander
- Division of Oncology, Department of Clinical Sciences Lund, Lund University and Skåne University Hospital, 223 81 Lund, Sweden; (M.B.); (J.-M.J.); (S.W.F.); (L.M.d.l.F.); (L.T.); (S.M.); (G.H.)
| | - Päivi Kannisto
- Division of Obstetrics and Gynaecology, Department of Clinical Sciences Lund, Lund University and Skåne University Hospital, 222 42 Lund, Sweden;
| | - Anna Måsbäck
- Department of Surgical Pathology, Division of Laboratory Medicine, Skåne University Hospital, 222 42 Lund, Sweden;
| | - Gabriella Honeth
- Division of Oncology, Department of Clinical Sciences Lund, Lund University and Skåne University Hospital, 223 81 Lund, Sweden; (M.B.); (J.-M.J.); (S.W.F.); (L.M.d.l.F.); (L.T.); (S.M.); (G.H.)
| | - Ingrid Hedenfalk
- Division of Oncology, Department of Clinical Sciences Lund, Lund University and Skåne University Hospital, 223 81 Lund, Sweden; (M.B.); (J.-M.J.); (S.W.F.); (L.M.d.l.F.); (L.T.); (S.M.); (G.H.)
| |
Collapse
|
94
|
Golan T, O'Kane GM, Denroche RE, Raitses-Gurevich M, Grant RC, Holter S, Wang Y, Zhang A, Jang GH, Stossel C, Atias D, Halperin S, Berger R, Glick Y, Park JP, Cuggia A, Williamson L, Wong HL, Schaeffer DF, Renouf DJ, Borgida A, Dodd A, Wilson JM, Fischer SE, Notta F, Knox JJ, Zogopoulos G, Gallinger S. Genomic Features and Classification of Homologous Recombination Deficient Pancreatic Ductal Adenocarcinoma. Gastroenterology 2021; 160:2119-2132.e9. [PMID: 33524400 DOI: 10.1053/j.gastro.2021.01.220] [Citation(s) in RCA: 96] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 12/29/2020] [Accepted: 01/22/2021] [Indexed: 12/20/2022]
Abstract
BACKGROUND AND AIMS Homologous recombination deficiency (HRD) in pancreatic ductal adenocarcinoma (PDAC), remains poorly defined beyond germline (g) alterations in BRCA1, BRCA2, and PALB2. METHODS We interrogated whole genome sequencing (WGS) data on 391 patients, including 49 carriers of pathogenic variants (PVs) in gBRCA and PALB2. HRD classifiers were applied to the dataset and included (1) the genomic instability score (GIS) used by Myriad's MyChoice HRD assay; (2) substitution base signature 3 (SBS3); (3) HRDetect; and (4) structural variant (SV) burden. Clinical outcomes and responses to chemotherapy were correlated with HRD status. RESULTS Biallelic tumor inactivation of gBRCA or PALB2 was evident in 43 of 49 germline carriers identifying HRD-PDAC. HRDetect (score ≥0.7) predicted gBRCA1/PALB2 deficiency with highest sensitivity (98%) and specificity (100%). HRD genomic tumor classifiers suggested that 7% to 10% of PDACs that do not harbor gBRCA/PALB2 have features of HRD. Of the somatic HRDetecthi cases, 69% were attributed to alterations in BRCA1/2, PALB2, RAD51C/D, and XRCC2, and a tandem duplicator phenotype. TP53 loss was more common in BRCA1- compared with BRCA2-associated HRD-PDAC. HRD status was not prognostic in resected PDAC; however in advanced disease the GIS (P = .02), SBS3 (P = .03), and HRDetect score (P = .005) were predictive of platinum response and superior survival. PVs in gATM (n = 6) or gCHEK2 (n = 2) did not result in HRD-PDAC by any of the classifiers. In 4 patients, BRCA2 reversion mutations associated with platinum resistance. CONCLUSIONS Germline and parallel somatic profiling of PDAC outperforms germline testing alone in identifying HRD-PDAC. An additional 7% to 10% of patients without gBRCA/PALB2 mutations may benefit from DNA damage response agents.
Collapse
Affiliation(s)
- Talia Golan
- Pancreatic Cancer Translational Research Laboratory, Oncology Institute, Sheba Medical Center, Tel Hashomer, Israel; Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.
| | - Grainne M O'Kane
- PanCuRx Translational Research Initiative, Ontario Institute for Cancer Research, Toronto, Ontario, Canada; Wallace McCain Centre for Pancreatic Cancer, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Robert E Denroche
- PanCuRx Translational Research Initiative, Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | - Maria Raitses-Gurevich
- Pancreatic Cancer Translational Research Laboratory, Oncology Institute, Sheba Medical Center, Tel Hashomer, Israel
| | - Robert C Grant
- PanCuRx Translational Research Initiative, Ontario Institute for Cancer Research, Toronto, Ontario, Canada; Wallace McCain Centre for Pancreatic Cancer, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Spring Holter
- Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Yifan Wang
- The Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada; The Goodman Cancer Research Centre of McGill University, Montreal, Quebec, Canada
| | - Amy Zhang
- PanCuRx Translational Research Initiative, Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | - Gun Ho Jang
- PanCuRx Translational Research Initiative, Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | - Chani Stossel
- Pancreatic Cancer Translational Research Laboratory, Oncology Institute, Sheba Medical Center, Tel Hashomer, Israel; Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Dikla Atias
- Pancreatic Cancer Translational Research Laboratory, Oncology Institute, Sheba Medical Center, Tel Hashomer, Israel; Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Sharon Halperin
- Pancreatic Cancer Translational Research Laboratory, Oncology Institute, Sheba Medical Center, Tel Hashomer, Israel
| | - Raanan Berger
- Pancreatic Cancer Translational Research Laboratory, Oncology Institute, Sheba Medical Center, Tel Hashomer, Israel; Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Yulia Glick
- Pancreatic Cancer Translational Research Laboratory, Oncology Institute, Sheba Medical Center, Tel Hashomer, Israel; Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - J Patrick Park
- The Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada; The Goodman Cancer Research Centre of McGill University, Montreal, Quebec, Canada
| | - Adeline Cuggia
- The Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada; The Goodman Cancer Research Centre of McGill University, Montreal, Quebec, Canada
| | - Laura Williamson
- Canada's Michael Smith Genome Sciences Centre at BC Cancer, Vancouver, British Columbia, Canada
| | - Hui-Li Wong
- BC Cancer, Vancouver Centre, Pancreas Centre BC, Canada
| | | | | | - Ayelet Borgida
- Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Anna Dodd
- Wallace McCain Centre for Pancreatic Cancer, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Julie M Wilson
- PanCuRx Translational Research Initiative, Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | - Sandra E Fischer
- Department of Laboratory Medicine and Pathobiology, University of Toronto, University Health Network, Toronto, Ontario, Canada
| | - Faiyaz Notta
- PanCuRx Translational Research Initiative, Ontario Institute for Cancer Research, Toronto, Ontario, Canada; Division of Research, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada; Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Jennifer J Knox
- PanCuRx Translational Research Initiative, Ontario Institute for Cancer Research, Toronto, Ontario, Canada; Wallace McCain Centre for Pancreatic Cancer, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - George Zogopoulos
- The Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada; The Goodman Cancer Research Centre of McGill University, Montreal, Quebec, Canada
| | - Steven Gallinger
- PanCuRx Translational Research Initiative, Ontario Institute for Cancer Research, Toronto, Ontario, Canada; Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada; Department of Surgery, University of Toronto, Toronto, Ontario, Canada; Hepatobiliary/Pancreatic Surgical Oncology Program, University Health Network, Toronto, Ontario, Canada.
| |
Collapse
|
95
|
Meier B, Volkova NV, Hong Y, Bertolini S, González-Huici V, Petrova T, Boulton S, Campbell PJ, Gerstung M, Gartner A. Protection of the C. elegans germ cell genome depends on diverse DNA repair pathways during normal proliferation. PLoS One 2021; 16:e0250291. [PMID: 33905417 PMCID: PMC8078821 DOI: 10.1371/journal.pone.0250291] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Accepted: 04/01/2021] [Indexed: 12/13/2022] Open
Abstract
Maintaining genome integrity is particularly important in germ cells to ensure faithful transmission of genetic information across generations. Here we systematically describe germ cell mutagenesis in wild-type and 61 DNA repair mutants cultivated over multiple generations. ~44% of the DNA repair mutants analysed showed a >2-fold increased mutagenesis with a broad spectrum of mutational outcomes. Nucleotide excision repair deficiency led to higher base substitution rates, whereas polh-1(Polη) and rev-3(Polζ) translesion synthesis polymerase mutants resulted in 50-400 bp deletions. Signatures associated with defective homologous recombination fall into two classes: 1) brc-1/BRCA1 and rad-51/RAD51 paralog mutants showed increased mutations across all mutation classes, 2) mus-81/MUS81 and slx-1/SLX1 nuclease, and him-6/BLM, helq-1/HELQ or rtel-1/RTEL1 helicase mutants primarily accumulated structural variants. Repetitive and G-quadruplex sequence-containing loci were more frequently mutated in specific DNA repair backgrounds. Tandem duplications embedded in inverted repeats were observed in helq-1 helicase mutants, and a unique pattern of 'translocations' involving homeologous sequences occurred in rip-1 recombination mutants. atm-1/ATM checkpoint mutants harboured structural variants specifically enriched in subtelomeric regions. Interestingly, locally clustered mutagenesis was only observed for combined brc-1 and cep-1/p53 deficiency. Our study provides a global view of how different DNA repair pathways contribute to prevent germ cell mutagenesis.
Collapse
Affiliation(s)
- Bettina Meier
- Centre for Gene Regulation and Expression, University of Dundee, Dundee, Scotland
| | - Nadezda V. Volkova
- European Molecular Biology Laboratory, European Bioinformatics Institute, Hinxton, United Kingdom
| | - Ye Hong
- Centre for Gene Regulation and Expression, University of Dundee, Dundee, Scotland
| | - Simone Bertolini
- Centre for Gene Regulation and Expression, University of Dundee, Dundee, Scotland
| | | | - Tsvetana Petrova
- Centre for Gene Regulation and Expression, University of Dundee, Dundee, Scotland
| | | | - Peter J. Campbell
- Cancer, Ageing and Somatic Mutation Program, Wellcome Sanger Institute, Hinxton, United Kingdom
- Department of Haematology, University of Cambridge, Cambridge, United Kingdom
- Department of Haematology, Addenbrooke’s Hospital, Cambridge, United Kingdom
| | - Moritz Gerstung
- European Molecular Biology Laboratory, European Bioinformatics Institute, Hinxton, United Kingdom
- European Molecular Biology Laboratory, Genome Biology Unit, Heidelberg, Germany
| | - Anton Gartner
- Centre for Gene Regulation and Expression, University of Dundee, Dundee, Scotland
- Department of Biological Sciences, School of Life Sciences, Ulsan National Institute of Science and Technology, Ulsan, Republic of Korea
- Center for Genomic Integrity, Institute for Basic Science, Ulsan, Republic of Korea
| |
Collapse
|
96
|
Mechanisms of High-Grade Serous Carcinogenesis in the Fallopian Tube and Ovary: Current Hypotheses, Etiologic Factors, and Molecular Alterations. Int J Mol Sci 2021; 22:ijms22094409. [PMID: 33922503 PMCID: PMC8122889 DOI: 10.3390/ijms22094409] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 04/19/2021] [Accepted: 04/20/2021] [Indexed: 12/15/2022] Open
Abstract
Ovarian high-grade serous carcinomas (HGSCs) are a heterogeneous group of diseases. They include fallopian-tube-epithelium (FTE)-derived and ovarian-surface-epithelium (OSE)-derived tumors. The risk/protective factors suggest that the etiology of HGSCs is multifactorial. Inflammation caused by ovulation and retrograde bleeding may play a major role. HGSCs are among the most genetically altered cancers, and TP53 mutations are ubiquitous. Key driving events other than TP53 mutations include homologous recombination (HR) deficiency, such as BRCA 1/2 dysfunction, and activation of the CCNE1 pathway. HR deficiency and the CCNE1 amplification appear to be mutually exclusive. Intratumor heterogeneity resulting from genomic instability can be observed at the early stage of tumorigenesis. In this review, I discuss current carcinogenic hypotheses, sites of origin, etiologic factors, and molecular alterations of HGSCs.
Collapse
|
97
|
Huo Y, Selenica P, Mahdi AH, Pareja F, Kyker-Snowman K, Chen Y, Kumar R, Da Cruz Paula A, Basili T, Brown DN, Pei X, Riaz N, Tan Y, Huang YX, Li T, Barnard NJ, Reis-Filho JS, Weigelt B, Xia B. Genetic interactions among Brca1, Brca2, Palb2, and Trp53 in mammary tumor development. NPJ Breast Cancer 2021; 7:45. [PMID: 33893322 PMCID: PMC8065161 DOI: 10.1038/s41523-021-00253-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Accepted: 03/24/2021] [Indexed: 02/08/2023] Open
Abstract
Inherited mutations in BRCA1, BRCA2, and PALB2 cause a high risk of breast cancer. Here, we conducted parallel conditional knockout (CKO) of Brca1, Palb2, and Brca2, individually and in combination, along with one copy of Trp53, in the mammary gland of nulliparous female mice. We observed a functional equivalence of the three genes in their basic tumor-suppressive activity, a linear epistasis of Palb2 and Brca2, but complementary roles of Brca1 and Palb2 in mammary tumor suppression, as combined ablation of either Palb2 or Brca2 with Brca1 led to delayed tumor formation. Whole-exome sequencing (WES) revealed both similarities and differences between Brca1 and Palb2 or Brca2 null tumors. Analyses of mouse mammary glands and cultured human cells showed that combined loss of BRCA1 and PALB2 led to high levels of reactive oxygen species (ROS) and increased apoptosis, implicating oxidative stress in the delayed tumor development in Brca1;Palb2 double CKO mice. The functional complementarity between BRCA1 and PALB2/BRCA2 and the role of ROS in tumorigenesis require further investigation.
Collapse
Affiliation(s)
- Yanying Huo
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, USA
- Department of Radiation Oncology, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Pier Selenica
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Amar H Mahdi
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, USA
- Department of Radiation Oncology, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
- Department of Physiology, College of Medicine, Al-Mustansiriyah University, Baghdad, Iraq
| | - Fresia Pareja
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Kelly Kyker-Snowman
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, USA
- Department of Radiation Oncology, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Ying Chen
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, USA
| | - Rahul Kumar
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Centre for Brain Research, Indian Institute of Science (IISc), Bangalore, India
| | - Arnaud Da Cruz Paula
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Thais Basili
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - David N Brown
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Xin Pei
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Nadeem Riaz
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Yongmei Tan
- Stomatological Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yu-Xiu Huang
- The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Tao Li
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, USA
- Department of Radiation Oncology, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Nicola J Barnard
- Department of Pathology and Laboratory Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Jorge S Reis-Filho
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Britta Weigelt
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| | - Bing Xia
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, USA.
- Department of Radiation Oncology, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA.
- Department of Pathology and Laboratory Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA.
| |
Collapse
|
98
|
Evolutionary Trajectories and Genomic Divergence in Localized Breast Cancers after Ipsilateral Breast Tumor Recurrence. Cancers (Basel) 2021; 13:cancers13081821. [PMID: 33920370 PMCID: PMC8069481 DOI: 10.3390/cancers13081821] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 04/07/2021] [Accepted: 04/08/2021] [Indexed: 12/30/2022] Open
Abstract
Simple Summary Ipsilateral breast tumor relapse (IBTR) occurs in 5–10% of localized breast cancers (BCs) within 10 years of incidence, despite proper treatment of the primary lesion. However, the clinical consequences of evolutionary trajectories of BC cells and their impact on IBTR remain poorly understood. Here, we conducted a longitudinal genomic analysis of 10 matched localized BC patients with IBTR. Overall, we identified the differences in homologous recombination deficiency, chromosomal instability, and somatic mutation drivers between primary and relapsed lesions. Our analyses highlighted three clonal architectures that shape by distinct mutagenic processes and subclonal diversification during relapse progression. Finally, this study provided a framework, which integrated actionable biomarkers with clonal architectures, towards improvement of future treatment decisions. Abstract The evolutionary trajectories that drive clinical and therapeutic consequences in localized breast cancers (BCs) with ipsilateral breast tumor relapse (IBTR) remain largely unknown. Analyses of longitudinal paired whole-exome sequencing data from 10 localized BC patients with IBTR reveal that, compared to primary breast tumors, homologous recombination (HR) deficiency, inactivation of the HR pathway, chromosomal instability, and somatic driver mutations are more frequent. Furthermore, three major models of evolution in IBTR are summarized, through which relative contributions of mutational signatures shift, and the subclonal diversity expansions are shown. Optimal treatment regimens are suggested by the clinically relevant molecular features, such as HR deficiency (20%) or specific alterations (30%) with sensitivity to available FDA-approved drugs. Finally, a rationale for the development of the therapeutic management framework is provided. This study sheds light on the complicated evolution patterns in IBTR and has significant clinical implications for future improvement of treatment decisions.
Collapse
|
99
|
Moukarzel LA, Ferrando L, Da Cruz Paula A, Brown DN, Geyer FC, Pareja F, Piscuoglio S, Papanastasiou AD, Fusco N, Marchiò C, Abu‐Rustum NR, Murali R, Brogi E, Wen HY, Norton L, Soslow RA, Vincent‐Salomon A, Reis‐Filho JS, Weigelt B. The genetic landscape of metaplastic breast cancers and uterine carcinosarcomas. Mol Oncol 2021; 15:1024-1039. [PMID: 33021035 PMCID: PMC8024717 DOI: 10.1002/1878-0261.12813] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Revised: 09/04/2020] [Accepted: 09/30/2020] [Indexed: 12/13/2022] Open
Abstract
Metaplastic breast carcinoma (MBC) and uterine carcinosarcoma (UCS) are rare aggressive cancers, characterized by an admixture of adenocarcinoma and areas displaying mesenchymal/sarcomatoid differentiation. We sought to define whether MBCs and UCSs harbor similar patterns of genetic alterations, and whether the different histologic components of MBCs and UCSs are clonally related. Whole-exome sequencing (WES) data from MBCs (n = 35) and UCSs (n = 57, The Cancer Genome Atlas) were reanalyzed to define somatic genetic alterations, altered signaling pathways, mutational signatures, and genomic features of homologous recombination DNA repair deficiency (HRD). In addition, the carcinomatous and sarcomatous components of an additional cohort of MBCs (n = 11) and UCSs (n = 6) were microdissected separately and subjected to WES, and their clonal relatedness was assessed. MBCs and UCSs harbored recurrent genetic alterations affecting TP53, PIK3CA, and PTEN, similar patterns of gene copy number alterations, and an enrichment in alterations affecting the epithelial-to-mesenchymal transition (EMT)-related Wnt and Notch signaling pathways. Differences were observed, however, including a significantly higher prevalence of FAT3 and FAT1 somatic mutations in MBCs compared to UCSs, and conversely, UCSs significantly more frequently harbored somatic mutations affecting FBXW7 and PPP2R1A as well as HER2 amplification than MBCs. Genomic features of HRD and biallelic alterations affecting bona fide HRD-related genes were found to be more prevalent in MBCs than in UCSs. The distinct histologic components of MBCs and UCSs were clonally related in all cases, with the sarcoma component likely stemming from a minor subclone of the carcinoma component in the samples with interpretable chronology of clonal evolution. Despite the similar histologic features and pathways affected by genetic alterations, UCSs differ from MBCs on the basis of FBXW7 and PPP2R1A mutations, HER2 amplification, and lack of HRD, supporting the notion that these entities are more than mere phenocopies of the same tumor type in different anatomical sites.
Collapse
Affiliation(s)
- Lea A. Moukarzel
- Department of SurgeryMemorial Sloan Kettering Cancer CenterNew YorkNYUSA
| | - Lorenzo Ferrando
- Department of PathologyMemorial Sloan Kettering Cancer CenterNew YorkNYUSA
- Department of Internal MedicineUniversity of GenoaItaly
| | | | - David N. Brown
- Department of PathologyMemorial Sloan Kettering Cancer CenterNew YorkNYUSA
| | - Felipe C. Geyer
- Department of PathologyMemorial Sloan Kettering Cancer CenterNew YorkNYUSA
| | - Fresia Pareja
- Department of PathologyMemorial Sloan Kettering Cancer CenterNew YorkNYUSA
| | - Salvatore Piscuoglio
- Department of PathologyMemorial Sloan Kettering Cancer CenterNew YorkNYUSA
- Visceral Surgery Research Laboratory, ClarunisDepartment of BiomedicineUniversity of BaselSwitzerland
| | - Anastasios D. Papanastasiou
- Department of PathologyMemorial Sloan Kettering Cancer CenterNew YorkNYUSA
- Department of Biomedical SciencesUniversity of West AtticaAthensGreece
| | - Nicola Fusco
- Department of PathologyMemorial Sloan Kettering Cancer CenterNew YorkNYUSA
- Division of PathologyFondazione IRCCS Ca' Grande – Ospedale Maggiore PoliclinicoMilanItaly
| | - Caterina Marchiò
- Department of PathologyMemorial Sloan Kettering Cancer CenterNew YorkNYUSA
- Department of Medical SciencesUniversity of TurinItaly
| | | | - Rajmohan Murali
- Department of PathologyMemorial Sloan Kettering Cancer CenterNew YorkNYUSA
| | - Edi Brogi
- Department of PathologyMemorial Sloan Kettering Cancer CenterNew YorkNYUSA
| | - Hannah Y. Wen
- Department of PathologyMemorial Sloan Kettering Cancer CenterNew YorkNYUSA
| | - Larry Norton
- Department of MedicineMemorial Sloan Kettering Cancer CenterNew YorkNYUSA
| | - Robert A. Soslow
- Department of PathologyMemorial Sloan Kettering Cancer CenterNew YorkNYUSA
| | | | | | - Britta Weigelt
- Department of PathologyMemorial Sloan Kettering Cancer CenterNew YorkNYUSA
| |
Collapse
|
100
|
Dror CM, Wyatt AW, Chi KN. Olaparib for the treatment of metastatic prostate cancer. Future Oncol 2021; 17:2413-2429. [PMID: 33769071 DOI: 10.2217/fon-2020-1245] [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: 11/21/2022] Open
Abstract
Recent innovations in the treatment of metastatic prostate cancer have improved patient outcomes. Nonetheless, this disease remains fatal and additional treatment approaches are needed. Greater understanding of the molecular landscape of metastatic prostate cancer has revealed recurrent alterations in key pathways amenable to therapeutic targeting. One such pathway is DNA repair, particularly alterations in genes directly or indirectly associated with homologous recombination repair found in up to one-quarter of patients with metastatic castrate-resistant prostate cancer (mCRPC). Olaparib, an inhibitor of poly-ADP-ribose polymerase, has recently gained approval for the treatment of mCRPC harboring alterations in homologous recombination repair genes. This review will provide a summary of evidence regarding PARP inhibition in the treatment of mCRPC, with a specific focus on olaparib.
Collapse
Affiliation(s)
| | - Alexander W Wyatt
- Department of Urologic Sciences, Vancouver Prostate Centre, University of British Columbia, Vancouver, BC, V5Z 4S6, Canada.,Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, BC, V5Z 4S6, Canada
| | - Kim N Chi
- BC Cancer, Vancouver, Vancouver, BC, V5Z 4S6, Canada.,Department of Urologic Sciences, Vancouver Prostate Centre, University of British Columbia, Vancouver, BC, V5Z 4S6, Canada
| |
Collapse
|