Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
Add more filters











Publication year range
1.
Ann Oncol ; 35(4): 364-380, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38244928

ABSTRACT

BACKGROUND: Resistance to therapies that target homologous recombination deficiency (HRD) in breast cancer limits their overall effectiveness. Multiple, preclinically validated, mechanisms of resistance have been proposed, but their existence and relative frequency in clinical disease are unclear, as is how to target resistance. PATIENTS AND METHODS: Longitudinal mutation and methylation profiling of circulating tumour (ct)DNA was carried out in 47 patients with metastatic BRCA1-, BRCA2- or PALB2-mutant breast cancer treated with HRD-targeted therapy who developed progressive disease-18 patients had primary resistance and 29 exhibited response followed by resistance. ctDNA isolated at multiple time points in the patient treatment course (before, on-treatment and at progression) was sequenced using a novel >750-gene intron/exon targeted sequencing panel. Where available, matched tumour biopsies were whole exome and RNA sequenced and also used to assess nuclear RAD51. RESULTS: BRCA1/2 reversion mutations were present in 60% of patients and were the most prevalent form of resistance. In 10 cases, reversions were detected in ctDNA before clinical progression. Two new reversion-based mechanisms were identified: (i) intragenic BRCA1/2 deletions with intronic breakpoints; and (ii) intragenic BRCA1/2 secondary mutations that formed novel splice acceptor sites, the latter being confirmed by in vitro minigene reporter assays. When seen before commencing subsequent treatment, reversions were associated with significantly shorter time to progression. Tumours with reversions retained HRD mutational signatures but had functional homologous recombination based on RAD51 status. Although less frequent than reversions, nonreversion mechanisms [loss-of-function (LoF) mutations in TP53BP1, RIF1 or PAXIP1] were evident in patients with acquired resistance and occasionally coexisted with reversions, challenging the notion that singular resistance mechanisms emerge in each patient. CONCLUSIONS: These observations map the prevalence of candidate drivers of resistance across time in a clinical setting, information with implications for clinical management and trial design in HRD breast cancers.


Subject(s)
Antineoplastic Agents , Breast Neoplasms , Female , Humans , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , BRCA1 Protein/genetics , BRCA2 Protein/genetics , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Homologous Recombination , Mutation , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors/therapeutic use , Tumor Suppressor p53-Binding Protein 1
3.
Ann Oncol ; 33(8): 769-785, 2022 08.
Article in English | MEDLINE | ID: mdl-35605746

ABSTRACT

BACKGROUND: Invasive lobular breast cancer (ILC) is the second most common type of breast cancer after invasive breast cancer of no special type (NST), representing up to 15% of all breast cancers. DESIGN: Latest data on ILC are presented, focusing on diagnosis, molecular make-up according to the European Society for Medical Oncology Scale for Clinical Actionability of molecular Targets (ESCAT) guidelines, treatment in the early and metastatic setting and ILC-focused clinical trials. RESULTS: At the imaging level, magnetic resonance imaging-based and novel positron emission tomography/computed tomography-based techniques can overcome the limitations of currently used imaging techniques for diagnosing ILC. At the pathology level, E-cadherin immunohistochemistry could help improving inter-pathologist agreement. The majority of patients with ILC do not seem to benefit as much from (neo-)adjuvant chemotherapy as patients with NST, although chemotherapy might be required in a subset of high-risk patients. No differences in treatment efficacy are seen for anti-human epidermal growth factor receptor 2 (HER2) therapies in the adjuvant setting and cyclin-dependent kinases 4 and 6 inhibitors in the metastatic setting. The clinical utility of the commercially available prognostic gene expression-based tests is unclear for patients with ILC. Several ESCAT alterations differ in frequency between ILC and NST. Germline BRCA1 and PALB2 alterations are less frequent in patients with ILC, while germline CDH1 (gene coding for E-cadherin) alterations are more frequent in patients with ILC. Somatic HER2 mutations are more frequent in ILC, especially in metastases (15% ILC versus 5% NST). A high tumour mutational burden, relevant for immune checkpoint inhibition, is more frequent in ILC metastases (16%) than in NST metastases (5%). Tumours with somatic inactivating CDH1 mutations may be vulnerable for treatment with ROS1 inhibitors, a concept currently investigated in early and metastatic ILC. CONCLUSION: ILC is a unique malignancy based on its pathological and biological features leading to differences in diagnosis as well as in treatment response, resistance and targets as compared to NST.


Subject(s)
Breast Neoplasms , Carcinoma, Ductal, Breast , Carcinoma, Lobular , Breast Neoplasms/diagnosis , Breast Neoplasms/genetics , Breast Neoplasms/therapy , Cadherins/therapeutic use , Carcinoma, Ductal, Breast/genetics , Carcinoma, Lobular/diagnosis , Carcinoma, Lobular/genetics , Carcinoma, Lobular/therapy , Female , Humans , Prognosis , Proto-Oncogene Proteins
4.
Ann Oncol ; 31(12): 1606-1622, 2020 12.
Article in English | MEDLINE | ID: mdl-33004253

ABSTRACT

BACKGROUND: Homologous recombination repair deficiency (HRD) is a frequent feature of high-grade serous ovarian, fallopian tube and peritoneal carcinoma (HGSC) and is associated with sensitivity to PARP inhibitor (PARPi) therapy. HRD testing provides an opportunity to optimise PARPi use in HGSC but methodologies are diverse and clinical application remains controversial. MATERIALS AND METHODS: To define best practice for HRD testing in HGSC the ESMO Translational Research and Precision Medicine Working Group launched a collaborative project that incorporated a systematic review approach. The main aims were to (i) define the term 'HRD test'; (ii) provide an overview of the biological rationale and the level of evidence supporting currently available HRD tests; (iii) provide recommendations on the clinical utility of HRD tests in clinical management of HGSC. RESULTS: A broad range of repair genes, genomic scars, mutational signatures and functional assays are associated with a history of HRD. Currently, the clinical validity of HRD tests in ovarian cancer is best assessed, not in terms of biological HRD status per se, but in terms of PARPi benefit. Clinical trials evidence supports the use of BRCA mutation testing and two commercially available assays that also incorporate genomic instability for identifying subgroups of HGSCs that derive different magnitudes of benefit from PARPi therapy, albeit with some variation by clinical scenario. These tests can be used to inform treatment selection and scheduling but their use is limited by a failure to consistently identify a subgroup of patients who derive no benefit from PARPis in most studies. Existing tests lack negative predictive value and inadequately address the complex and dynamic nature of the HRD phenotype. CONCLUSIONS: Currently available HRD tests are useful for predicting likely magnitude of benefit from PARPis but better biomarkers are urgently needed to better identify current homologous recombination proficiency status and stratify HGSC management.


Subject(s)
Ovarian Neoplasms , Poly(ADP-ribose) Polymerase Inhibitors , Biomarkers , Carcinoma, Ovarian Epithelial , Female , Homologous Recombination , Humans , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/genetics , Poly(ADP-ribose) Polymerase Inhibitors/therapeutic use
5.
Oncogene ; 39(24): 4780, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32427987

ABSTRACT

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

7.
Ann Oncol ; 30(9): 1437-1447, 2019 09 01.
Article in English | MEDLINE | ID: mdl-31218365

ABSTRACT

Genomic instability is a hallmark of cancer, and often is the result of altered DNA repair capacities in tumour cells. DNA damage repair defects are common in different cancer types; these alterations can also induce tumour-specific vulnerabilities that can be exploited therapeutically. In 2009, a first-in-man clinical trial of the poly(ADP-ribose) polymerase (PARP) inhibitor olaparib clinically validated the synthetic lethal interaction between inhibition of PARP1, a key sensor of DNA damage, and BRCA1/BRCA2 deficiency. In this review, we summarize a decade of PARP inhibitor clinical development, a work that has resulted in the registration of several PARP inhibitors in breast (olaparib and talazoparib) and ovarian cancer (olaparib, niraparib and rucaparib, either alone or following platinum chemotherapy as maintenance therapy). Over the past 10 years, our knowledge on the mechanism of action of PARP inhibitor as well as how tumours become resistant has been extended, and we summarise this work here. We also discuss opportunities for expanding the precision medicine approach with PARP inhibitors, identifying a wider population who could benefit from this drug class. This includes developing and validating better predictive biomarkers for patient stratification, mainly based on homologous recombination defects beyond BRCA1/BRCA2 mutations, identifying DNA repair deficient tumours in other cancer types such as prostate or pancreatic cancer, or by designing combination therapies with PARP inhibitors.


Subject(s)
Breast Neoplasms/drug therapy , Genomic Instability/drug effects , Ovarian Neoplasms/drug therapy , Poly(ADP-ribose) Polymerase Inhibitors/therapeutic use , BRCA1 Protein/genetics , BRCA2 Protein/genetics , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Female , Humans , Indazoles/therapeutic use , Indoles/therapeutic use , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Phthalazines/therapeutic use , Piperazines/therapeutic use , Piperidines/therapeutic use , Poly (ADP-Ribose) Polymerase-1/genetics
8.
Oncogene ; 33(8): 966-76, 2014 Feb 20.
Article in English | MEDLINE | ID: mdl-23474757

ABSTRACT

The gene encoding the receptor tyrosine kinase ERBB2, also known as HER2, is amplified and/or overexpressed in up to 15% of breast cancers. These tumours are characterised by an aggressive phenotype and poor clinical outcome. Although therapies targeted at ERBB2 have proven effective, many patients fail to respond to treatment or become resistant and the reasons for this are still largely unknown. Using a high-throughput functional screen we assessed whether genes found to be recurrently amplified and overexpressed in ERBB2+ve breast cancers mediate resistance to the ERBB2-targeted agent lapatinib. Lapatinib-resistant ERBB2-amplified breast cancer cell lines were screened, in the presence or absence of lapatinib, with an RNA interference library targeting 369 genes recurrently amplified and overexpressed in both ERBB2-amplified breast cancer tumours and cell lines. Small interfering RNAs targeting a number of genes caused sensitivity to lapatinib in this context. The mechanisms of resistance conferred by the identified genes were further investigated and in the case of NIBP (TRAPPC9), lapatinib resistance was found to be mediated through NF-κB signalling. Our results indicate that specific amplified and/ or overexpressed genes found in ERBB2-amplified breast cancer may mediate response to ERBB2-targeting agents.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm/genetics , Genes, erbB-2 , Quinazolines/pharmacology , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Female , High-Throughput Screening Assays , Humans , Lapatinib , RNA Interference , RNA, Small Interfering/genetics
9.
Oncogene ; 33(5): 619-31, 2014 Jan 30.
Article in English | MEDLINE | ID: mdl-23334330

ABSTRACT

Overexpression of the receptor tyrosine kinase ERBB2 (also known as HER2) occurs in around 15% of breast cancers and is driven by amplification of the ERBB2 gene. ERBB2 amplification is a marker of poor prognosis, and although anti-ERBB2-targeted therapies have shown significant clinical benefit, de novo and acquired resistance remains an important problem. Genomic profiling has demonstrated that ERBB2+ve breast cancers are distinguished from ER+ve and 'triple-negative' breast cancers by harbouring not only the ERBB2 amplification on 17q12, but also a number of co-amplified genes on 17q12 and amplification events on other chromosomes. Some of these genes may have important roles in influencing clinical outcome, and could represent genetic dependencies in ERBB2+ve cancers and therefore potential therapeutic targets. Here, we describe an integrated genomic, gene expression and functional analysis to determine whether the genes present within amplicons are critical for the survival of ERBB2+ve breast tumour cells. We show that only a fraction of the ERBB2-amplified breast tumour lines are truly addicted to the ERBB2 oncogene at the mRNA level and display a heterogeneous set of additional genetic dependencies. These include an addiction to the transcription factor gene TFAP2C when it is amplified and overexpressed, suggesting that TFAP2C represents a genetic dependency in some ERBB2+ve breast cancer cells.


Subject(s)
Breast Neoplasms/genetics , Gene Amplification/genetics , Receptor, ErbB-2/genetics , Receptor, ErbB-2/metabolism , Transcription Factor AP-2/metabolism , Breast Neoplasms/metabolism , Cell Line, Tumor , Female , Gene Expression Profiling , Humans , MCF-7 Cells , RNA Interference , RNA, Small Interfering , Receptor, ErbB-2/biosynthesis , Transcription Factor AP-2/biosynthesis
10.
Oncogene ; 32(47): 5377-87, 2013 Nov 21.
Article in English | MEDLINE | ID: mdl-23934192

ABSTRACT

Excision repair cross-complementation group 1 (ERCC1) is a DNA repair enzyme that is frequently defective in non-small cell lung cancer (NSCLC). Although low ERCC1 expression correlates with platinum sensitivity, the clinical effectiveness of platinum therapy is limited, highlighting the need for alternative treatment strategies. To discover new mechanism-based therapeutic strategies for ERCC1-defective tumours, we performed high-throughput drug screens in an isogenic NSCLC model of ERCC1 deficiency and dissected the mechanism underlying ERCC1-selective effects by studying molecular biomarkers of tumour cell response. The high-throughput screens identified multiple clinical poly (ADP-ribose) polymerase 1 and 2 (PARP1/2) inhibitors, such as olaparib (AZD-2281), niraparib (MK-4827) and BMN 673, as being selective for ERCC1 deficiency. We observed that ERCC1-deficient cells displayed a significant delay in double-strand break repair associated with a profound and prolonged G2/M arrest following PARP1/2 inhibitor treatment. Importantly, we found that ERCC1 isoform 202, which has recently been shown to mediate platinum sensitivity, also modulated PARP1/2 sensitivity. A PARP1/2 inhibitor-synthetic lethal siRNA screen revealed that ERCC1 deficiency was epistatic with homologous recombination deficiency. However, ERCC1-deficient cells did not display a defect in RAD51 foci formation, suggesting that ERCC1 might be required to process PARP1/2 inhibitor-induced DNA lesions before DNA strand invasion. PARP1 silencing restored PARP1/2 inhibitor resistance in ERCC1-deficient cells but had no effect in ERCC1-proficient cells, supporting the hypothesis that PARP1 might be required for the ERCC1 selectivity of PARP1/2 inhibitors. This study suggests that PARP1/2 inhibitors as a monotherapy could represent a novel therapeutic strategy for NSCLC patients with ERCC1-deficient tumours.


Subject(s)
Antineoplastic Agents/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , DNA-Binding Proteins/genetics , Endonucleases/genetics , Enzyme Inhibitors/therapeutic use , Lung Neoplasms/drug therapy , Phthalazines/therapeutic use , Poly(ADP-ribose) Polymerase Inhibitors , Antineoplastic Agents/pharmacology , Cell Line , Cell Line, Tumor , DNA Breaks, Double-Stranded/drug effects , DNA Repair , Enzyme Inhibitors/pharmacology , G2 Phase Cell Cycle Checkpoints/drug effects , High-Throughput Screening Assays , Humans , Indazoles/pharmacology , Indazoles/therapeutic use , Phthalazines/pharmacology , Piperazines/pharmacology , Piperazines/therapeutic use , Piperidines/pharmacology , Piperidines/therapeutic use , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerases , Protein Isoforms , RNA Interference , RNA, Small Interfering , Rad51 Recombinase/metabolism
12.
Br J Cancer ; 108(4): 983-92, 2013 Mar 05.
Article in English | MEDLINE | ID: mdl-23361057

ABSTRACT

BACKGROUND: DNA mismatch repair deficiency is present in a significant proportion of a number of solid tumours and is associated with distinct clinical behaviour. METHODS: To identify the therapeutic agents that might show selectivity for mismatch repair-deficient tumour cells, we screened a pair of isogenic MLH1-deficient and MLH1-proficient tumour cell lines with a library of clinically used drugs. To test the generality of hits in the screen, selective agents were retested in cells deficient in the MSH2 mismatch repair gene. RESULTS: We identified cytarabine and other related cytosine-based nucleoside analogues as being selectively toxic to MLH1 and MSH2-deficient tumour cells. The selective cytotoxicity we observed was likely caused by increased levels of cellular oxidative stress, as it could be abrogated by antioxidants. CONCLUSION: We propose that cytarabine-based chemotherapy regimens may represent a tumour-selective treatment strategy for mismatch repair-deficient cancers.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Antimetabolites, Antineoplastic/therapeutic use , Cytarabine/pharmacology , DNA Mismatch Repair/drug effects , DNA Repair-Deficiency Disorders/drug therapy , MutS Homolog 2 Protein/genetics , Neoplasms/genetics , Nuclear Proteins/genetics , Oxidative Stress/drug effects , Cell Line, Tumor , Humans , MutL Protein Homolog 1 , Nucleosides , Oxidative Stress/genetics
13.
Oncogene ; 30(41): 4243-60, 2011 Oct 13.
Article in English | MEDLINE | ID: mdl-21552290

ABSTRACT

Individuals with germline mutations in the tumour-suppressor gene CYLD are at high risk of developing disfiguring cutaneous appendageal tumours, the defining tumour being the highly organised cylindroma. Here, we analysed CYLD mutant tumour genomes by array comparative genomic hybridisation and gene expression microarray analysis. CYLD mutant tumours were characterised by an absence of copy-number aberrations apart from LOH chromosome 16q, the genomic location of the CYLD gene. Gene expression profiling of CYLD mutant tumours showed dysregulated tropomyosin kinase (TRK) signalling, with overexpression of TRKB and TRKC in tumours when compared with perilesional skin. Immunohistochemical analysis of a tumour microarray showed strong membranous TRKB and TRKC staining in cylindromas, as well as elevated levels of ERK phosphorylation and BCL2 expression. Membranous TRKC overexpression was also observed in 70% of sporadic BCCs. RNA interference-mediated silencing of TRKB and TRKC, as well as treatment with the small-molecule TRK inhibitor lestaurtinib, reduced colony formation and proliferation in 3D primary cell cultures established from CYLD mutant tumours. These results suggest that TRK inhibition could be used as a strategy to treat tumours with loss of functional CYLD.


Subject(s)
Neoplasms/genetics , Protein Kinases/genetics , Signal Transduction/genetics , Tumor Suppressor Proteins/genetics , Adenoma, Sweat Gland/genetics , Adenoma, Sweat Gland/metabolism , Adenoma, Sweat Gland/pathology , Carbazoles/pharmacology , Carcinoma, Adenoid Cystic/genetics , Carcinoma, Adenoid Cystic/metabolism , Carcinoma, Adenoid Cystic/pathology , Cluster Analysis , Comparative Genomic Hybridization , Deubiquitinating Enzyme CYLD , Furans , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Immunoblotting , Immunohistochemistry , Mutation , Neoplasms/metabolism , Neoplasms/pathology , Neoplasms, Basal Cell/genetics , Neoplasms, Basal Cell/metabolism , Neoplasms, Basal Cell/pathology , Oligonucleotide Array Sequence Analysis , Primary Cell Culture , Protein Kinases/metabolism , RNA Interference , Receptor, trkB/antagonists & inhibitors , Receptor, trkB/genetics , Receptor, trkB/metabolism , Receptor, trkC/antagonists & inhibitors , Receptor, trkC/genetics , Receptor, trkC/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sweat Gland Neoplasms/genetics , Sweat Gland Neoplasms/metabolism , Sweat Gland Neoplasms/pathology , Tissue Array Analysis , Tumor Cells, Cultured , Tumor Suppressor Proteins/metabolism
14.
Br J Cancer ; 103(8): 1192-200, 2010 Oct 12.
Article in English | MEDLINE | ID: mdl-20877358

ABSTRACT

BACKGROUND: The BRCA2 gene is responsible for a high number of hereditary breast and ovarian cancers, and studies of the BRCA2 biological functions are limited by the lack of models that resemble the patient's tumour features. The aim of this study was to establish and characterise a new human breast carcinoma xenograft obtained from a woman carrying a germline BRCA2 mutation. METHODS: A transplantable xenograft was obtained by grafting a breast cancer sample into nude mice. The biological and genetic profiles of the xenograft were compared with that of the patient's tumour using histology, immunohistochemistry (IHC), BRCA2 sequencing, comparative genomic hybridisation (CGH), and qRT-PCR. Tumour response to standard chemotherapies was evaluated. RESULTS: Histological profile identified the tumour as a basal-like triple-negative breast cancer. Targeted BRCA2 DNA sequencing of the xenograft showed the presence of the mutation previously identified in the carrier. Comparative genomic hybridisation array profiles of the primary tumour and the xenograft revealed a high number of similar genetic alterations. The therapeutic assessment of the xenograft showed sensitivity to anthracyclin-based chemotherapy and resistance to docetaxel. The xenograft was also highly sensitive to radiotherapy and cisplatin-based treatments. CONCLUSIONS: This study describes a new human breast cancer xenograft obtained from a BRCA2-mutated patient. This xenograft provides a new model for the pre-clinical drug development and for the exploration of the drug response biological basis.


Subject(s)
Breast Neoplasms/pathology , Carcinoma, Ductal, Breast/pathology , Genes, BRCA2 , Germ-Line Mutation , Adult , Animals , Anthracyclines/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Carcinoma, Ductal, Breast/drug therapy , Carcinoma, Ductal, Breast/genetics , Cell Culture Techniques , Cell Line, Tumor , Comparative Genomic Hybridization , DNA Mutational Analysis , Female , Germ-Line Mutation/physiology , Heterozygote , Humans , Mice , Mice, Nude , Neoplasm Transplantation , Transplantation, Heterologous , Xenograft Model Antitumor Assays/methods
15.
Oncogene ; 28(11): 1465-70, 2009 Mar 19.
Article in English | MEDLINE | ID: mdl-19182824

ABSTRACT

The BRCA1 and BRCA2 proteins are involved in the maintenance of genome stability and germ-line loss-of-function mutations in either BRCA1 or BRCA2 strongly predispose carriers to cancers of the breast and other organs. It has been demonstrated previously that inhibiting elements of the cellular DNA maintenance pathways represents a novel therapeutic approach to treating tumors in these individuals. Here, we show that inhibition of the telomere-associated protein, Tankyrase 1, is also selectively lethal with BRCA deficiency. We also demonstrate that the selectivity caused by inhibition of Tankyrase 1 is associated with an exacerbation of the centrosome amplification phenotype associated with BRCA deficiency. We propose that inhibition of Tankyrase 1 could be therapeutically exploited in BRCA-associated cancers.


Subject(s)
Genes, BRCA1/physiology , Genes, BRCA2/physiology , Neoplasms/therapy , Tankyrases/antagonists & inhibitors , Centrosome/physiology , Gene Amplification , Gene Silencing , HCT116 Cells , Humans , Neoplasms/enzymology , Neoplasms/genetics , Tankyrases/physiology
16.
J Clin Pathol ; 62(3): 195-200, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19126568

ABSTRACT

Genetic screens, where the effects of modifying gene function on cell behaviour are assessed in a systematic fashion, have for some time provided useful information to those interested in disease pathogenesis and treatment. Genetic screens exploiting the phenomenon of RNA interference (RNAi) are now becoming commonplace. This article explains the different RNAi screen formats and describes some of the applications of RNAi screening that may be pertinent to the research pathologist.


Subject(s)
Genetic Testing/methods , RNA Interference , Drug Resistance, Neoplasm/genetics , Gene Library , Humans , Myelodysplastic Syndromes/genetics , RNA, Small Interfering/genetics , Virus Diseases/genetics
17.
J Pathol ; 216(4): 399-407, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18810758

ABSTRACT

Expression profiling studies have suggested that HER2-amplified breast cancers constitute a heterogeneous group that may be subdivided according to their ER status: HER2-amplified ER-positive breast carcinomas that fall into the luminal B cluster; and HER2-amplified ER-negative cancers which form a distinct molecular subgroup, known as the erbB2 or HER2 subgroup. ER-negative breast cancer differs significantly from ER-positive disease in the pattern, type, and complexity of genetic aberrations. Here we have compared the genomic profiles of ER-positive and ER-negative HER2-amplified cancers using tiling path microarray-based comparative genomic hybridization (aCGH). Validation of the differentially amplified regions was performed in an independent series of 70 HER2-amplified breast cancers. Although HER2-amplified cancers had remarkably complex patterns of molecular genetic aberrations, ER-positive and ER-negative HER2-amplified breast carcinomas shared most molecular genetic features as defined by aCGH. Genome-wide Fisher's exact test analysis revealed that less than 1.5% of the genome was significantly differentially gained or lost in ER-positive versus ER-negative HER2-amplified cancers. However, two regions of amplification were significantly associated with ER-positive carcinomas, one of which mapped to 17q21.2 and encompassed GJC1, IGFBP4, TNS4, and TOP2A. Chromogenic in situ hybridization analysis of an independent validation series confirmed the association between ER status and TOP2A amplification. In conclusion, although hormone receptor status does not determine the overall genetic profile of HER2-amplified breast cancers, specific genetic aberrations may be characteristic of subgroups of HER2 breast cancers.


Subject(s)
Breast Neoplasms/genetics , Gene Expression Profiling , Genes, erbB-2 , Oligonucleotide Array Sequence Analysis , Receptors, Estrogen/genetics , Antigens, Neoplasm/genetics , Breast Neoplasms/pathology , DNA Topoisomerases, Type II/genetics , DNA-Binding Proteins/genetics , Female , Gene Amplification , Humans , In Situ Hybridization/methods , Poly-ADP-Ribose Binding Proteins
18.
Oncogene ; 27(8): 1036-44, 2008 Feb 14.
Article in English | MEDLINE | ID: mdl-17700519

ABSTRACT

The PPM1D gene is aberrantly amplified in a range of common cancers and encodes a protein phosphatase that is a potential therapeutic target. However, the issue of whether inhibition of PPM1D in human tumour cells that overexpress this protein compromises their viability has not yet been fully addressed. We show here, using an RNA interference (RNAi) approach, that inhibition of PPM1D can indeed reduce the viability of human tumour cells and that this effect is selective; tumour cell lines that overexpress PPM1D are sensitive to PPM1D inhibition whereas cell lines with normal levels are not. Loss of viability associated with PPM1D RNAi in human tumour cells occurs via the activation of the kinase P38. To identify chemical inhibitors of PPM1D, a high-throughput screening of a library of small molecules was performed. This strategy successfully identified a compound that selectively reduces viability of human tumour cell lines that overexpress PPM1D. As expected of a specific inhibitor, the toxicity to PPM1D overexpressing cell lines after inhibitor treatment is P38 dependent. These results further validate PPM1D as a therapeutic target and identify a proof-of-principle small molecule inhibitor.


Subject(s)
Breast Neoplasms/enzymology , Breast Neoplasms/pathology , Cyclopentanes/pharmacology , Growth Inhibitors/pharmacology , Phosphoprotein Phosphatases/antagonists & inhibitors , Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Cell Line, Tumor , Drug Resistance, Neoplasm/drug effects , Enzyme Inhibitors/pharmacology , HeLa Cells , Humans , Phosphoprotein Phosphatases/biosynthesis , Phosphoprotein Phosphatases/genetics , Protein Phosphatase 2C
19.
Oncogene ; 26(54): 7601-6, 2007 Nov 29.
Article in English | MEDLINE | ID: mdl-17563742

ABSTRACT

The DSS1 protein interacts with the breast cancer susceptibility protein BRCA2 that plays an integral role in the repair of DNA double-strand breaks (DSBs). DSS1 has also been shown to interact with components of the 26S proteasome in Saccharomyces cerevisiae and in human tumour cells. This raises the possibility of functional interplay between the DNA repair machinery and the proteasome. We show here that human DSS1 interacts with the RPN3 and RPN7 proteasome subunits and define regions of DSS1 important for the interactions with RPN3, RPN7 and BRCA2. We also show that BRCA2 interacts with RPN3 and RPN7 and that the BRCA2/RPN7 interaction is independent of DSS1. Finally, and most significantly, we demonstrate that the proteolytic activity of the proteasome is a determinant of the choice of DSB repair pathway; inhibition of proteasome proteolytic activity results in an increase in the utilization of potentially mutagenic single-strand annealing at the expense of a reduction in the level of error-free gene conversion. This confirms a functional link between DSB repair and proteasomal activity.


Subject(s)
DNA Damage , DNA Repair , Proteasome Endopeptidase Complex/genetics , Proteasome Endopeptidase Complex/metabolism , Apoptosis Regulatory Proteins , BRCA2 Protein/genetics , Breast Neoplasms/genetics , Exoribonucleases , Female , Genetic Predisposition to Disease , Hexosyltransferases , Humans , Kinetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics
20.
Article in English | MEDLINE | ID: mdl-16869747

ABSTRACT

Individuals harboring germ-line mutations in the BRCA1 or BRCA2 genes are at highly elevated risk of a variety of cancers. Ten years of research has revealed roles for BRCA1 and BRCA2 in a wide variety of cellular processes. However, it seems likely that the function of these proteins in DNA repair is critically important in maintaining genome stability. Despite this increasing knowledge of the defects present in BRCA-deficient cells, BRCA mutation carriers developing cancer are still treated similarly to sporadic cases. Here we describe our efforts, based on understanding the DNA repair defects in BRCAdeficient cells, to define the optimal existing treatment for cancers arising in BRCA mutation carriers and, additionally, the development of novel therapeutic approaches. Finally, we discuss how therapies developed to treat BRCA mutant tumors might be applied to some sporadic cancers sharing similar specific defects in DNA repair.


Subject(s)
DNA Repair/genetics , Genes, BRCA1 , Genes, BRCA2 , Germ-Line Mutation , Neoplasms/genetics , Neoplasms/therapy , Animals , Collagen Type XI/antagonists & inhibitors , DNA Damage , Female , Heterozygote , Humans , Mice , Models, Genetic , Recombination, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL