Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 195
Filter
Add more filters

Publication year range
1.
Mol Cell ; 82(16): 2939-2951.e5, 2022 08 18.
Article in English | MEDLINE | ID: mdl-35793673

ABSTRACT

PARP1 rapidly detects DNA strand break damage and allosterically signals break detection to the PARP1 catalytic domain to activate poly(ADP-ribose) production from NAD+. PARP1 activation is characterized by dynamic changes in the structure of a regulatory helical domain (HD); yet, there are limited insights into the specific contributions that the HD makes to PARP1 allostery. Here, we have determined crystal structures of PARP1 in isolated active states that display specific HD conformations. These captured snapshots and biochemical analysis illustrate HD contributions to PARP1 multi-domain and high-affinity interaction with DNA damage, provide novel insights into the mechanics of PARP1 allostery, and indicate how HD active conformations correspond to alterations in the catalytic region that reveal the active site to NAD+. Our work deepens the understanding of PARP1 catalytic activation, the dynamics of the binding site of PARP inhibitor compounds, and the mechanisms regulating PARP1 retention on DNA damage.


Subject(s)
DNA Damage , NAD , Catalytic Domain , DNA Repair , NAD/metabolism , Poly (ADP-Ribose) Polymerase-1/metabolism , Poly Adenosine Diphosphate Ribose , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology
2.
Mol Cell ; 81(4): 767-783.e11, 2021 02 18.
Article in English | MEDLINE | ID: mdl-33333017

ABSTRACT

Chromatin is a barrier to efficient DNA repair, as it hinders access and processing of certain DNA lesions. ALC1/CHD1L is a nucleosome-remodeling enzyme that responds to DNA damage, but its precise function in DNA repair remains unknown. Here we report that loss of ALC1 confers sensitivity to PARP inhibitors, methyl-methanesulfonate, and uracil misincorporation, which reflects the need to remodel nucleosomes following base excision by DNA glycosylases but prior to handover to APEX1. Using CRISPR screens, we establish that ALC1 loss is synthetic lethal with homologous recombination deficiency (HRD), which we attribute to chromosome instability caused by unrepaired DNA gaps at replication forks. In the absence of ALC1 or APEX1, incomplete processing of BER intermediates results in post-replicative DNA gaps and a critical dependence on HR for repair. Hence, targeting ALC1 alone or as a PARP inhibitor sensitizer could be employed to augment existing therapeutic strategies for HRD cancers.


Subject(s)
Chromatin Assembly and Disassembly , DNA Helicases/metabolism , DNA-Binding Proteins/metabolism , Neoplasm Proteins/metabolism , Neoplasms, Experimental/metabolism , Nucleosomes/metabolism , Poly(ADP-ribose) Polymerases/metabolism , Animals , DNA Helicases/genetics , DNA Replication/drug effects , DNA-(Apurinic or Apyrimidinic Site) Lyase/genetics , DNA-(Apurinic or Apyrimidinic Site) Lyase/metabolism , DNA-Binding Proteins/genetics , Homologous Recombination/drug effects , Mice , Mice, Knockout , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Neoplasms, Experimental/genetics , Nucleosomes/genetics , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Poly(ADP-ribose) Polymerases/genetics
3.
Cell ; 145(1): 30-8, 2011 Apr 01.
Article in English | MEDLINE | ID: mdl-21458666

ABSTRACT

As cancer cell genomes are unveiled at a breathtaking pace, the genetic principles at play in cancer are emerging in all their complexity, prompting the assessment of classical genetic interaction models. Here, we discuss the implications of these findings for cancer progression and heterogeneity and for the development of new therapeutic approaches.


Subject(s)
Epistasis, Genetic , Neoplasms/genetics , Animals , Disease Progression , Humans , Mutation , Neoplasms/physiopathology
4.
Nature ; 585(7825): 447-452, 2020 09.
Article in English | MEDLINE | ID: mdl-32908313

ABSTRACT

Genomic instability is a hallmark of cancer, and has a central role in the initiation and development of breast cancer1,2. The success of poly-ADP ribose polymerase inhibitors in the treatment of breast cancers that are deficient in homologous recombination exemplifies the utility of synthetically lethal genetic interactions in the treatment of breast cancers that are driven by genomic instability3. Given that defects in homologous recombination are present in only a subset of breast cancers, there is a need to identify additional driver mechanisms for genomic instability and targeted strategies to exploit these defects in the treatment of cancer. Here we show that centrosome depletion induces synthetic lethality in cancer cells that contain the 17q23 amplicon, a recurrent copy number aberration that defines about 9% of all primary breast cancer tumours and is associated with high levels of genomic instability4-6. Specifically, inhibition of polo-like kinase 4 (PLK4) using small molecules leads to centrosome depletion, which triggers mitotic catastrophe in cells that exhibit amplicon-directed overexpression of TRIM37. To explain this effect, we identify TRIM37 as a negative regulator of centrosomal pericentriolar material. In 17q23-amplified cells that lack centrosomes, increased levels of TRIM37 block the formation of foci that comprise pericentriolar material-these foci are structures with a microtubule-nucleating capacity that are required for successful cell division in the absence of centrosomes. Finally, we find that the overexpression of TRIM37 causes genomic instability by delaying centrosome maturation and separation at mitotic entry, and thereby increases the frequency of mitotic errors. Collectively, these findings highlight TRIM37-dependent genomic instability as a putative driver event in 17q23-amplified breast cancer and provide a rationale for the use of centrosome-targeting therapeutic agents in treating these cancers.


Subject(s)
Breast Neoplasms/genetics , Centrosome/metabolism , Centrosome/pathology , Chromosomes, Human, Pair 17/genetics , Tripartite Motif Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Antineoplastic Agents/pharmacology , Breast Neoplasms/pathology , Cell Line, Tumor , Centrosome/drug effects , Female , G2 Phase , Genomic Instability , Humans , Mitosis/genetics , Protein Serine-Threonine Kinases/metabolism , Tripartite Motif Proteins/genetics , Ubiquitin-Protein Ligases/genetics
5.
Br J Cancer ; 130(11): 1828-1840, 2024 May.
Article in English | MEDLINE | ID: mdl-38600325

ABSTRACT

BACKGROUND: Invasive Lobular Carcinoma (ILC) is a morphologically distinct breast cancer subtype that represents up to 15% of all breast cancers. Compared to Invasive Breast Carcinoma of No Special Type (IBC-NST), ILCs exhibit poorer long-term outcome and a unique pattern of metastasis. Despite these differences, the systematic discovery of robust prognostic biomarkers and therapeutically actionable molecular pathways in ILC remains limited. METHODS: Pathway-centric multivariable models using statistical machine learning were developed and tested in seven retrospective clinico-genomic cohorts (n = 996). Further external validation was performed using a new RNA-Seq clinical cohort of aggressive ILCs (n = 48). RESULTS AND CONCLUSIONS: mRNA dysregulation scores of 25 pathways were strongly prognostic in ILC (FDR-adjusted P < 0.05). Of these, three pathways including Cell-cell communication, Innate immune system and Smooth muscle contraction were also independent predictors of chemotherapy response. To aggregate these findings, a multivariable machine learning predictor called PSILC was developed and successfully validated for predicting overall and metastasis-free survival in ILC. Integration of PSILC with CRISPR-Cas9 screening data from breast cancer cell lines revealed 16 candidate therapeutic targets that were synthetic lethal with high-risk ILCs. This study provides interpretable prognostic and predictive biomarkers of ILC which could serve as the starting points for targeted drug discovery for this disease.


Subject(s)
Breast Neoplasms , Carcinoma, Lobular , Humans , Female , Breast Neoplasms/genetics , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Carcinoma, Lobular/drug therapy , Carcinoma, Lobular/genetics , Carcinoma, Lobular/pathology , Carcinoma, Lobular/metabolism , Prognosis , Retrospective Studies , Biomarkers, Tumor/genetics , Machine Learning , Middle Aged , Gene Expression Regulation, Neoplastic , Neoplasm Invasiveness
6.
Nature ; 560(7716): 117-121, 2018 08.
Article in English | MEDLINE | ID: mdl-30022168

ABSTRACT

53BP1 is a chromatin-binding protein that regulates the repair of DNA double-strand breaks by suppressing the nucleolytic resection of DNA termini1,2. This function of 53BP1 requires interactions with PTIP3 and RIF14-9, the latter of which recruits REV7 (also known as MAD2L2) to break sites10,11. How 53BP1-pathway proteins shield DNA ends is currently unknown, but there are two models that provide the best potential explanation of their action. In one model the 53BP1 complex strengthens the nucleosomal barrier to end-resection nucleases12,13, and in the other 53BP1 recruits effector proteins with end-protection activity. Here we identify a 53BP1 effector complex, shieldin, that includes C20orf196 (also known as SHLD1), FAM35A (SHLD2), CTC-534A2.2 (SHLD3) and REV7. Shieldin localizes to double-strand-break sites in a 53BP1- and RIF1-dependent manner, and its SHLD2 subunit binds to single-stranded DNA via OB-fold domains that are analogous to those of RPA1 and POT1. Loss of shieldin impairs non-homologous end-joining, leads to defective immunoglobulin class switching and causes hyper-resection. Mutations in genes that encode shieldin subunits also cause resistance to poly(ADP-ribose) polymerase inhibition in BRCA1-deficient cells and tumours, owing to restoration of homologous recombination. Finally, we show that binding of single-stranded DNA by SHLD2 is critical for shieldin function, consistent with a model in which shieldin protects DNA ends to mediate 53BP1-dependent DNA repair.


Subject(s)
DNA Repair , Multiprotein Complexes/metabolism , Tumor Suppressor p53-Binding Protein 1/metabolism , Animals , CRISPR-Cas Systems , Cell Line , DNA Breaks, Double-Stranded , DNA, Single-Stranded/genetics , Female , Genes, BRCA1 , Humans , Immunoglobulin Class Switching/genetics , Mice , Models, Biological , Multiprotein Complexes/chemistry , Multiprotein Complexes/deficiency , Multiprotein Complexes/genetics , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Telomere-Binding Proteins/metabolism , Tumor Suppressor Protein p53/deficiency
7.
PLoS Genet ; 17(2): e1009354, 2021 02.
Article in English | MEDLINE | ID: mdl-33591981

ABSTRACT

The RB1 tumor suppressor is recurrently mutated in a variety of cancers including retinoblastomas, small cell lung cancers, triple-negative breast cancers, prostate cancers, and osteosarcomas. Finding new synthetic lethal (SL) interactions with RB1 could lead to new approaches to treating cancers with inactivated RB1. We identified 95 SL partners of RB1 based on a Drosophila screen for genetic modifiers of the eye phenotype caused by defects in the RB1 ortholog, Rbf1. We validated 38 mammalian orthologs of Rbf1 modifiers as RB1 SL partners in human cancer cell lines with defective RB1 alleles. We further show that for many of the RB1 SL genes validated in human cancer cell lines, low activity of the SL gene in human tumors, when concurrent with low levels of RB1 was associated with improved patient survival. We investigated higher order combinatorial gene interactions by creating a novel Drosophila cancer model with co-occurring Rbf1, Pten and Ras mutations, and found that targeting RB1 SL genes in this background suppressed the dramatic tumor growth and rescued fly survival whilst having minimal effects on wild-type cells. Finally, we found that drugs targeting the identified RB1 interacting genes/pathways, such as UNC3230, PYR-41, TAK-243, isoginkgetin, madrasin, and celastrol also elicit SL in human cancer cell lines. In summary, we identified several high confidence, evolutionarily conserved, novel targets for RB1-deficient cells that may be further adapted for the treatment of human cancer.


Subject(s)
Neoplasms/genetics , Phosphotransferases (Alcohol Group Acceptor)/genetics , RNA Splicing , Retinoblastoma Protein/genetics , Signal Transduction , Ubiquitin/metabolism , Animals , Animals, Genetically Modified , Cell Line, Tumor , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Eye Abnormalities/genetics , Eye Abnormalities/metabolism , Humans , Neoplasms/metabolism , Neoplasms/pathology , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Phosphotransferases (Alcohol Group Acceptor)/metabolism , RNA Interference , Retinoblastoma Protein/deficiency , Retinoblastoma Protein/metabolism , Species Specificity , Survival Analysis , Synthetic Lethal Mutations/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , ras Proteins/genetics , ras Proteins/metabolism
8.
Cancer Treat Res ; 186: 13-23, 2023.
Article in English | MEDLINE | ID: mdl-37978128

ABSTRACT

PARP inhibitors now have proven utility in the treatment of homologous recombination (HR) defective cancers. These drugs, and the synthetic lethality effect they exploit, have not only taught us how to approach the treatment of HR defective cancers but have also illuminated how resistance to a synthetic lethal approach can occur, how cancer-associated synthetic lethal effects are perhaps more complex than we imagine, how the better use of biomarkers could improve the success of treatment and even how drug resistance might be targeted. Here, we discuss some of the lessons learnt from the study of PARP inhibitor synthetic lethality and how these lessons might have wider application. Specifically, we discuss the concept of synthetic lethal penetrance, phenocopy effects in cancer such as BRCAness, synthetic lethal resistance, the polygenic and complex nature of synthetic lethal interactions, how evolutionary double binds could be exploited in treatment as well as future horizons for the field.


Subject(s)
Antineoplastic Agents , Neoplasms , Humans , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors/therapeutic use , Synthetic Lethal Mutations , Neoplasms/drug therapy , Antineoplastic Agents/therapeutic use
9.
Semin Cancer Biol ; 61: 121-131, 2020 04.
Article in English | MEDLINE | ID: mdl-31698086

ABSTRACT

Ovarian clear cell carcinomas (OCCC) are rare aggressive, chemo-resistant tumours comprising approximately 13% of all epithelial ovarian cancers, which have distinct clinical and molecular features, when compared to other gynaecological malignancies. At present, there are no specific licensed targeted therapies for OCCC, although a number of candidate targets have been identified. This review focuses on recent knowledge underpinning our understanding of the pathogenesis of OCCC including direct and synthetic-lethal therapeutic strategies in particular focussing on ARID1A deficiency. We also discuss current targeted clinical trials and immunotherapeutic approaches.


Subject(s)
Adenocarcinoma, Clear Cell/etiology , Carcinoma, Ovarian Epithelial/etiology , Genomics , Translational Research, Biomedical , Adenocarcinoma, Clear Cell/diagnosis , Adenocarcinoma, Clear Cell/metabolism , Adenocarcinoma, Clear Cell/therapy , Biomarkers , Carcinoma, Ovarian Epithelial/diagnosis , Carcinoma, Ovarian Epithelial/metabolism , Carcinoma, Ovarian Epithelial/therapy , DNA Copy Number Variations , DNA-Binding Proteins/genetics , Disease Management , Epigenesis, Genetic , Female , Genetic Association Studies , Genetic Predisposition to Disease , Genomics/methods , Humans , Mutation , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Signal Transduction , Transcription Factors/genetics
10.
Int J Gynecol Cancer ; 31(11): 1471-1475, 2021 11.
Article in English | MEDLINE | ID: mdl-34518240

ABSTRACT

BACKGROUND: ARID1A (AT-rich interactive domain containing protein 1A) loss-of-function mutations have been reported in gynecological cancers, including rarer subtypes such as clear cell carcinoma. Preclinical studies indicate that ARID1A mutant cancers display sensitivity to ATR inhibition while tumors without ARID1A mutations may be sensitive to Ataxia telangiectasia and Rad3 related (ATR) inhibitors in combination with poly-ADP ribose polymerase (PARP) inhibitors. PRIMARY OBJECTIVE: To determine whether the ATR inhibitor, ceralasertib, has clinical activity as a single agent and in combination with the PARP inhibitor, olaparib, in patients with ARID1A 'loss' and 'no loss' clear cell carcinomas and other relapsed gynecological cancers. STUDY HYPOTHESIS: ARID1A deficient clear cell carcinoma of the ovary or endometrium is sensitive to ATR inhibition, while the combination of ATR and PARP inhibition has activity in other gynecological tumors, irrespective of ARID1A status. TRIAL DESIGN: ATARI (ENGOT/GYN1/NCRI) is a multicenter, international, proof-of-concept, phase II, parallel cohort trial assessing ceralasertib activity as a single agent and in combination with olaparib in ARID1A stratified gynecological cancers. Patients with relapsed ovarian/endometrial clear cell carcinoma with ARID1A loss will receive ceralasertib monotherapy (cohort 1A). Relapsed ovarian/endometrial clear cell carcinoma patients with no ARID1A loss (cohort 2) or patients with other histological subtypes (endometrioid, carcinosarcoma, cervical) (cohort 3) will receive combination therapy (olaparib/ceralasertib). Treatment will continue until disease progression. MAJOR INCLUSION/EXCLUSION CRITERIA: Patients with histologically confirmed recurrent clear cell (ovarian, endometrial, or endometriosis related), endometrioid (ovarian, endometrial, or endometriosis related), cervical (adenocarcinomas or squamous), or carcinosarcomas (ovarian or endometrial) are eligible. Patients progressing after ≥1 prior platinum with evidence of measurable (RECIST v1.1) radiological disease progression since last systemic anticancer therapy and prior to trial entry are eligible. Previous ATR or PARP inhibitor treatment is not permissible. PRIMARY ENDPOINT: Best overall objective response rate (RECIST v1.1). SAMPLE SIZE: A minimum of 40 and a maximum of 116. ESTIMATED DATES FOR COMPLETING ACCRUAL AND PRESENTING RESULTS: Accrual is anticipated to be complete by the second quarter of 2022, with reporting of results by the fourth quarter of 2022. Overall accrual targets and reporting timelines are dependent on individual cohort progression to stage 2. TRIAL REGISTRATION NUMBER: NCT0405269.


Subject(s)
Indoles/administration & dosage , Morpholines/administration & dosage , Ovarian Neoplasms/drug therapy , Phthalazines/administration & dosage , Piperazines/administration & dosage , Poly(ADP-ribose) Polymerase Inhibitors/administration & dosage , Protein Kinase Inhibitors/administration & dosage , Pyrimidines/administration & dosage , Sulfonamides/administration & dosage , DNA-Binding Proteins , Endometrial Neoplasms , Female , Humans , Multicenter Studies as Topic , Neoplasm Recurrence, Local/drug therapy , Transcription Factors
11.
Am J Hum Genet ; 100(4): 650-658, 2017 Apr 06.
Article in English | MEDLINE | ID: mdl-28343630

ABSTRACT

Intellectual disability (ID) is a highly heterogeneous disorder involving at least 600 genes, yet a genetic diagnosis remains elusive in ∼35%-40% of individuals with moderate to severe ID. Recent meta-analyses statistically analyzing de novo mutations in >7,000 individuals with neurodevelopmental disorders highlighted mutations in PPM1D as a possible cause of ID. PPM1D is a type 2C phosphatase that functions as a negative regulator of cellular stress-response pathways by mediating a feedback loop of p38-p53 signaling, thereby contributing to growth inhibition and suppression of stress-induced apoptosis. We identified 14 individuals with mild to severe ID and/or developmental delay and de novo truncating PPM1D mutations. Additionally, deep phenotyping revealed overlapping behavioral problems (ASD, ADHD, and anxiety disorders), hypotonia, broad-based gait, facial dysmorphisms, and periods of fever and vomiting. PPM1D is expressed during fetal brain development and in the adult brain. All mutations were located in the last or penultimate exon, suggesting escape from nonsense-mediated mRNA decay. Both PPM1D expression analysis and cDNA sequencing in EBV LCLs of individuals support the presence of a stable truncated transcript, consistent with this hypothesis. Exposure of cells derived from individuals with PPM1D truncating mutations to ionizing radiation resulted in normal p53 activation, suggesting that p53 signaling is unaffected. However, a cell-growth disadvantage was observed, suggesting a possible effect on the stress-response pathway. Thus, we show that de novo truncating PPM1D mutations in the last and penultimate exons cause syndromic ID, which provides additional insight into the role of cell-cycle checkpoint genes in neurodevelopmental disorders.


Subject(s)
Exons , Intellectual Disability/genetics , Mutation , Protein Phosphatase 2C/genetics , Adolescent , Cell Cycle , Child , Child, Preschool , Humans , Intellectual Disability/pathology , Young Adult
12.
Cancer ; 125(12): 1963-1972, 2019 06 15.
Article in English | MEDLINE | ID: mdl-30835824

ABSTRACT

Substantial progress has been made in understanding ovarian cancer at the molecular and cellular level. Significant improvement in 5-year survival has been achieved through cytoreductive surgery, combination platinum-based chemotherapy, and more effective treatment of recurrent cancer, and there are now more than 280,000 ovarian cancer survivors in the United States. Despite these advances, long-term survival in late-stage disease has improved little over the last 4 decades. Poor outcomes relate, in part, to late stage at initial diagnosis, intrinsic drug resistance, and the persistence of dormant drug-resistant cancer cells after primary surgery and chemotherapy. Our ability to accelerate progress in the clinic will depend on the ability to answer several critical questions regarding this disease. To assess current answers, an American Association for Cancer Research Special Conference on "Critical Questions in Ovarian Cancer Research and Treatment" was held in Pittsburgh, Pennsylvania, on October 1-3, 2017. Although clinical, translational, and basic investigators conducted much of the discussion, advocates participated in the meeting, and many presentations were directly relevant to patient care, including treatment with poly adenosine diphosphate ribose polymerase (PARP) inhibitors, attempts to improve immunotherapy by overcoming the immune suppressive effects of the microenvironment, and a better understanding of the heterogeneity of the disease.


Subject(s)
Antineoplastic Agents/therapeutic use , Immunotherapy/methods , Ovarian Neoplasms/diagnosis , Ovarian Neoplasms/drug therapy , Patient-Centered Care , Poly(ADP-ribose) Polymerase Inhibitors/therapeutic use , Congresses as Topic , Drug Resistance, Neoplasm , Female , Humans , Societies, Scientific , Tumor Microenvironment
13.
Gut ; 67(10): 1780-1792, 2018 10.
Article in English | MEDLINE | ID: mdl-28830912

ABSTRACT

OBJECTIVE: Oesophageal cancer is the seventh most common cause of cancer-related death worldwide. Disease relapse is frequent and treatment options are limited. DESIGN: To identify new biomarker-defined therapeutic approaches for patients with oesophageal cancer, we integrated the genomic profiles of 17 oesophageal tumour-derived cell lines with drug sensitivity data from small molecule inhibitor profiling, identifying drug sensitivity effects associated with cancer driver gene alterations. We also interrogated recently described RNA interference screen data for these tumour cell lines to identify candidate genetic dependencies or vulnerabilities that could be exploited as therapeutic targets. RESULTS: By integrating the genomic features of oesophageal tumour cell lines with siRNA and drug screening data, we identified a series of candidate targets in oesophageal cancer, including a sensitivity to inhibition of the kinase BTK in MYC amplified oesophageal tumour cell lines. We found that this genetic dependency could be elicited with the clinical BTK/ERBB2 kinase inhibitor, ibrutinib. In both MYC and ERBB2 amplified tumour cells, ibrutinib downregulated ERK-mediated signal transduction, cMYC Ser-62 phosphorylation and levels of MYC protein, and elicited G1 cell cycle arrest and apoptosis, suggesting that this drug could be used to treat biomarker-selected groups of patients with oesophageal cancer. CONCLUSIONS: BTK represents a novel candidate therapeutic target in oesophageal cancer that can be targeted with ibrutinib. On the basis of this work, a proof-of-concept phase II clinical trial evaluating the efficacy of ibrutinib in patients with MYC and/or ERBB2 amplified advanced oesophageal cancer is currently underway (NCT02884453). TRIAL REGISTRATION NUMBER: NCT02884453; Pre-results.


Subject(s)
Esophageal Neoplasms , Proto-Oncogene Proteins c-myc/genetics , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Receptor, ErbB-2/genetics , Adenine/analogs & derivatives , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Drug Discovery/methods , Esophageal Neoplasms/drug therapy , Esophageal Neoplasms/genetics , Humans , Pharmacogenetics , Pharmacogenomic Testing/methods , Piperidines , RNA Interference/drug effects , Signal Transduction/genetics , Xenograft Model Antitumor Assays
14.
N Engl J Med ; 373(18): 1697-708, 2015 Oct 29.
Article in English | MEDLINE | ID: mdl-26510020

ABSTRACT

BACKGROUND: Prostate cancer is a heterogeneous disease, but current treatments are not based on molecular stratification. We hypothesized that metastatic, castration-resistant prostate cancers with DNA-repair defects would respond to poly(adenosine diphosphate [ADP]-ribose) polymerase (PARP) inhibition with olaparib. METHODS: We conducted a phase 2 trial in which patients with metastatic, castration-resistant prostate cancer were treated with olaparib tablets at a dose of 400 mg twice a day. The primary end point was the response rate, defined either as an objective response according to Response Evaluation Criteria in Solid Tumors, version 1.1, or as a reduction of at least 50% in the prostate-specific antigen level or a confirmed reduction in the circulating tumor-cell count from 5 or more cells per 7.5 ml of blood to less than 5 cells per 7.5 ml. Targeted next-generation sequencing, exome and transcriptome analysis, and digital polymerase-chain-reaction testing were performed on samples from mandated tumor biopsies. RESULTS: Overall, 50 patients were enrolled; all had received prior treatment with docetaxel, 49 (98%) had received abiraterone or enzalutamide, and 29 (58%) had received cabazitaxel. Sixteen of 49 patients who could be evaluated had a response (33%; 95% confidence interval, 20 to 48), with 12 patients receiving the study treatment for more than 6 months. Next-generation sequencing identified homozygous deletions, deleterious mutations, or both in DNA-repair genes--including BRCA1/2, ATM, Fanconi's anemia genes, and CHEK2--in 16 of 49 patients who could be evaluated (33%). Of these 16 patients, 14 (88%) had a response to olaparib, including all 7 patients with BRCA2 loss (4 with biallelic somatic loss, and 3 with germline mutations) and 4 of 5 with ATM aberrations. The specificity of the biomarker suite was 94%. Anemia (in 10 of the 50 patients [20%]) and fatigue (in 6 [12%]) were the most common grade 3 or 4 adverse events, findings that are consistent with previous studies of olaparib. CONCLUSIONS: Treatment with the PARP inhibitor olaparib in patients whose prostate cancers were no longer responding to standard treatments and who had defects in DNA-repair genes led to a high response rate. (Funded by Cancer Research UK and others; ClinicalTrials.gov number, NCT01682772; Cancer Research UK number, CRUK/11/029.).


Subject(s)
Antineoplastic Agents/therapeutic use , DNA Repair , Enzyme Inhibitors/therapeutic use , Phthalazines/therapeutic use , Piperazines/therapeutic use , Poly(ADP-ribose) Polymerase Inhibitors , Prostatic Neoplasms/drug therapy , Adult , Aged , Anemia/chemically induced , Ataxia Telangiectasia Mutated Proteins/genetics , DNA Repair/genetics , Drug Resistance, Neoplasm , Enzyme Inhibitors/adverse effects , Fatigue/chemically induced , Genes, BRCA2 , Genes, Tumor Suppressor , Humans , Male , Middle Aged , Mutation , Neoplasm Metastasis/drug therapy , Phthalazines/adverse effects , Piperazines/adverse effects , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology
15.
Nature ; 481(7381): 287-94, 2012 Jan 18.
Article in English | MEDLINE | ID: mdl-22258607

ABSTRACT

Genomic instability is one of the most pervasive characteristics of tumour cells and is probably the combined effect of DNA damage, tumour-specific DNA repair defects, and a failure to stop or stall the cell cycle before the damaged DNA is passed on to daughter cells. Although these processes drive genomic instability and ultimately the disease process, they also provide therapeutic opportunities. A better understanding of the cellular response to DNA damage will not only inform our knowledge of cancer development but also help to refine the classification as well as the treatment of the disease.


Subject(s)
DNA Damage , Neoplasms/genetics , Neoplasms/therapy , DNA Damage/drug effects , DNA Repair/drug effects , Genomic Instability/drug effects , Humans , Molecular Targeted Therapy , Neoplasms/pathology , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/pathology , Poly(ADP-ribose) Polymerase Inhibitors , Poly(ADP-ribose) Polymerases/metabolism , Tumor Microenvironment/drug effects
16.
J Biol Chem ; 291(32): 16686-98, 2016 08 05.
Article in English | MEDLINE | ID: mdl-27268052

ABSTRACT

We demonstrate the usefulness of synthetic lethal screening of a conditionally BCL6-deficient Burkitt lymphoma cell line, DG75-AB7, with a library of small molecules to determine survival pathways suppressed by BCL6 and suggest mechanism-based treatments for lymphoma. Lestaurtinib, a JAK2 inhibitor and one of the hits from the screen, repressed survival of BCL6-deficient cells in vitro and reduced growth and proliferation of xenografts in vivo BCL6 deficiency in DG75-AB7 induced JAK2 mRNA and protein expression and STAT3 phosphorylation. Surface IL10RA was elevated by BCL6 deficiency, and blockade of IL10RA repressed STAT3 phosphorylation. Therefore, we define an IL10RA/JAK2/STAT3 pathway each component of which is repressed by BCL6. We also show for the first time that JAK2 is a direct BCL6 target gene; BCL6 bound to the JAK2 promoter in vitro and was enriched by ChIP-seq. The place of JAK2 inhibitors in the treatment of diffuse large B-cell lymphoma has not been defined; we suggest that JAK2 inhibitors might be most effective in poor prognosis ABC-DLBCL, which shows higher levels of IL10RA, JAK2, and STAT3 but lower levels of BCL6 than GC-DLBCL and might be usefully combined with novel approaches such as inhibition of IL10RA.


Subject(s)
Burkitt Lymphoma/drug therapy , Carbazoles/pharmacology , Interleukin-10 Receptor alpha Subunit/metabolism , Janus Kinase 2/antagonists & inhibitors , Lymphoma, Large B-Cell, Diffuse/drug therapy , Proto-Oncogene Proteins c-bcl-6/biosynthesis , STAT3 Transcription Factor/metabolism , Animals , Burkitt Lymphoma/genetics , Burkitt Lymphoma/metabolism , Cell Line, Tumor , Furans , Humans , Interleukin-10 Receptor alpha Subunit/genetics , Janus Kinase 2/genetics , Janus Kinase 2/metabolism , Lymphoma, Large B-Cell, Diffuse/genetics , Lymphoma, Large B-Cell, Diffuse/metabolism , Mice , Mice, SCID , Proto-Oncogene Proteins c-bcl-6/genetics , STAT3 Transcription Factor/genetics , Xenograft Model Antitumor Assays
17.
Br J Cancer ; 117(1): 113-123, 2017 Jun 27.
Article in English | MEDLINE | ID: mdl-28535155

ABSTRACT

BACKGROUND: Elevated APOBEC3B expression in tumours correlates with a kataegic pattern of localised hypermutation. We assessed the cellular phenotypes associated with high-level APOBEC3B expression and the influence of p53 status on these phenotypes using an isogenic system. METHODS: We used RNA interference of p53 in cells with inducible APOBEC3B and assessed DNA damage response (DDR) biomarkers. The mutational effects of APOBEC3B were assessed using whole-genome sequencing. In vitro small-molecule inhibitor sensitivity profiling was used to identify candidate therapeutic vulnerabilities. RESULTS: Although APOBEC3B expression increased the incorporation of genomic uracil, invoked DDR biomarkers and caused cell cycle arrest, inactivation of p53 circumvented APOBEC3B-induced cell cycle arrest without reversing the increase in genomic uracil or DDR biomarkers. The continued expression of APOBEC3B in p53-defective cells not only caused a kataegic mutational signature but also caused hypersensitivity to small-molecule DDR inhibitors (ATR, CHEK1, CHEK2, PARP, WEE1 inhibitors) as well as cisplatin/ATR inhibitor and ATR/PARP inhibitor combinations. CONCLUSIONS: Although loss of p53 might allow tumour cells to tolerate elevated APOBEC3B expression, continued expression of this enzyme might impart a number of therapeutic vulnerabilities upon tumour cells.


Subject(s)
Cell Cycle Checkpoints/genetics , Cytidine Deaminase/genetics , DNA Damage/genetics , Gene Expression Regulation, Neoplastic , Minor Histocompatibility Antigens/genetics , Tumor Suppressor Protein p53/genetics , Ataxia Telangiectasia Mutated Proteins/antagonists & inhibitors , Blotting, Western , CRISPR-Cas Systems , Cell Cycle Checkpoints/drug effects , Cell Cycle Proteins/antagonists & inhibitors , Cell Line , Checkpoint Kinase 1/antagonists & inhibitors , Checkpoint Kinase 2/antagonists & inhibitors , Cisplatin/pharmacology , Cytidine Deaminase/metabolism , DNA Damage/drug effects , G2 Phase Cell Cycle Checkpoints , Gene Knockout Techniques , HEK293 Cells , Humans , Minor Histocompatibility Antigens/metabolism , Mutation , Nuclear Proteins/antagonists & inhibitors , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Protein-Tyrosine Kinases/antagonists & inhibitors , RNA Interference , Uracil/metabolism
18.
Annu Rev Med ; 66: 455-70, 2015.
Article in English | MEDLINE | ID: mdl-25341009

ABSTRACT

The genetic concept of synthetic lethality, in which the combination or synthesis of mutations in multiple genes results in cell death, provides a framework to design novel therapeutic approaches to cancer. Already there are promising indications, from clinical trials exploiting this concept by using poly(ADP-ribose) polymerase (PARP) inhibitors in patients with germline BRCA1 or BRCA2 gene mutations, that this approach could be beneficial. We discuss the biological rationale for BRCA-PARP synthetic lethality, how the synthetic lethal approach is being assessed in the clinic, and how mechanisms of resistance are starting to be dissected. Applying the synthetic lethal concept to target non-BRCA-mutant cancers also has clear potential, and we discuss how some of the principles learned in developing PARP inhibitors might also drive the development of additional genetic approaches.


Subject(s)
Breast Neoplasms/drug therapy , Enzyme Inhibitors/therapeutic use , Neoplasms, Cystic, Mucinous, and Serous/drug therapy , Ovarian Neoplasms/drug therapy , Poly(ADP-ribose) Polymerase Inhibitors , Benzamides/therapeutic use , Breast Neoplasms/genetics , Drug Resistance, Neoplasm , Female , Genes, BRCA1 , Genes, BRCA2 , Humans , Mutation , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms, Cystic, Mucinous, and Serous/genetics , Ovarian Neoplasms/genetics , Phthalazines/therapeutic use , Piperazines/therapeutic use , Poly (ADP-Ribose) Polymerase-1
19.
J Pathol ; 239(2): 197-205, 2016 06.
Article in English | MEDLINE | ID: mdl-26969893

ABSTRACT

Cutaneous cylindroma is an adnexal tumour with apocrine differentiation. A predisposition to multiple cylindromas is seen in patients with Brooke-Spiegler syndrome, who carry germline mutations in the tumour suppressor gene CYLD. Previous studies of inherited cylindromas have highlighted the frequent presence of bi-allelic truncating CYLD mutations as a recurrent driver mutation. We have previously shown that sporadic cylindromas express either MYB-NFIB fusion transcripts or show evidence of MYB activation in the absence of such fusions. Here, we investigated inherited cylindromas from several families with germline CYLD mutations for the presence of MYB activation. Strikingly, none of the inherited CYLD-defective (n = 23) tumours expressed MYB-NFIB fusion transcripts. However, MYB expression was increased in the majority of tumours (69%) and global gene expression analysis revealed that well-established MYB target genes were up-regulated in CYLD-defective tumours. Moreover, knock-down of MYB expression caused a significant reduction in cylindroma cell proliferation, suggesting that MYB is also a key player and oncogenic driver in inherited cylindromas. Taken together, our findings suggest molecular heterogeneity in the pathogenesis of sporadic and inherited cutaneous cylindromas, with convergence on MYB activation. © 2016 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.


Subject(s)
Germ-Line Mutation , Neoplastic Syndromes, Hereditary/genetics , Oncogene Proteins, Fusion/genetics , Skin Neoplasms/genetics , Tumor Suppressor Proteins/genetics , Adult , Aged , Aged, 80 and over , Cell Proliferation , Deubiquitinating Enzyme CYLD , Female , Gene Expression Profiling , Genotype , Humans , In Situ Hybridization, Fluorescence , Male , Middle Aged , Neoplastic Syndromes, Hereditary/metabolism , Neoplastic Syndromes, Hereditary/pathology , Oligonucleotide Array Sequence Analysis , Oncogene Proteins, Fusion/metabolism , Phenotype , Sequence Analysis, DNA , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Tumor Suppressor Proteins/metabolism
20.
J Pathol ; 240(3): 315-328, 2016 11.
Article in English | MEDLINE | ID: mdl-27512948

ABSTRACT

The initiation and progression of breast cancer from the transformation of the normal epithelium to ductal carcinoma in situ (DCIS) and invasive disease is a complex process involving the acquisition of genetic alterations and changes in gene expression, alongside microenvironmental and recognized histological alterations. Here, we sought to comprehensively characterise the genomic and transcriptomic features of the MCF10 isogenic model of breast cancer progression, and to functionally validate potential driver alterations in three-dimensional (3D) spheroids that may provide insights into breast cancer progression, and identify targetable alterations in conditions more similar to those encountered in vivo. We performed whole genome, exome and RNA sequencing of the MCF10 progression series to catalogue the copy number and mutational and transcriptomic landscapes associated with progression. We identified a number of predicted driver mutations (including PIK3CA and TP53) that were acquired during transformation of non-malignant MCF10A cells to their malignant counterparts that are also present in analysed primary breast cancers from The Cancer Genome Atlas (TCGA). Acquisition of genomic alterations identified MYC amplification and previously undescribed RAB3GAP1-HRAS and UBA2-PDCD2L expressed in-frame fusion genes in malignant cells. Comparison of pathway aberrations associated with progression showed that, when cells are grown as 3D spheroids, they show perturbations of cancer-relevant pathways. Functional interrogation of the dependency on predicted driver events identified alterations in HRAS, PIK3CA and TP53 that selectively decreased cell growth and were associated with progression from preinvasive to invasive disease only when cells were grown as spheroids. Our results have identified changes in the genomic repertoire in cell lines representative of the stages of breast cancer progression, and demonstrate that genetic dependencies can be uncovered when cells are grown in conditions more like those in vivo. The MCF10 progression series therefore represents a good model with which to dissect potential biomarkers and to evaluate therapeutic targets involved in the progression of breast cancer. © 2016 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.


Subject(s)
Breast Neoplasms/genetics , Carcinoma, Ductal, Breast/genetics , Carcinoma, Intraductal, Noninfiltrating/genetics , Models, Biological , Phosphatidylinositol 3-Kinases/genetics , Transcriptome , Breast Neoplasms/pathology , Carcinoma, Ductal, Breast/pathology , Carcinoma, Intraductal, Noninfiltrating/pathology , Cell Line, Tumor , Cell Transformation, Neoplastic , Class I Phosphatidylinositol 3-Kinases , DNA, Neoplasm/chemistry , DNA, Neoplasm/genetics , Disease Progression , Exome/genetics , Female , Gene Expression Regulation, Neoplastic , Genome , High-Throughput Nucleotide Sequencing , Humans , Mutation , Phosphatidylinositol 3-Kinases/metabolism , Sequence Analysis, DNA , Spheroids, Cellular , Tumor Suppressor Protein p53/genetics
SELECTION OF CITATIONS
SEARCH DETAIL