Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
1.
Nucleic Acids Res ; 51(3): 1173-1188, 2023 02 22.
Article in English | MEDLINE | ID: mdl-36715327

ABSTRACT

The DNA mismatch repair protein MutSα recognizes wrongly incorporated DNA bases and initiates their correction during DNA replication. Dysfunctions in mismatch repair lead to a predisposition to cancer. Here, we study the homozygous mutation V63E in MSH2 that was found in the germline of a patient with suspected constitutional mismatch repair deficiency syndrome who developed colorectal cancer before the age of 30. Characterization of the mutant in mouse models, as well as slippage and repair assays, shows a mildly pathogenic phenotype. Using cryogenic electron microscopy and surface plasmon resonance, we explored the mechanistic effect of this mutation on MutSα function. We discovered that V63E disrupts a previously unappreciated interface between the mismatch binding domains (MBDs) of MSH2 and MSH6 and leads to reduced DNA binding. Our research identifies this interface as a 'safety lock' that ensures high-affinity DNA binding to increase replication fidelity. Our mechanistic model explains the hypomorphic phenotype of the V63E patient mutation and other variants in the MBD interface.


Subject(s)
DNA Mismatch Repair , DNA Repair , MutS Homolog 2 Protein , Animals , Mice , DNA/chemistry , Mutation , MutS Homolog 2 Protein/metabolism
2.
Hum Mutat ; 43(9): 1249-1258, 2022 09.
Article in English | MEDLINE | ID: mdl-35451539

ABSTRACT

The large majority of germline alterations identified in the DNA mismatch repair (MMR) gene PMS2, a low-penetrance gene for the cancer predisposition Lynch syndrome, represent variants of uncertain significance (VUS). The inability to classify most VUS interferes with personalized healthcare. The complete in vitro MMR activity (CIMRA) assay, that only requires sequence information on the VUS, provides a functional analysis-based quantitative tool to improve the classification of VUS in MMR proteins. To derive a formula that translates CIMRA assay results into the odds of pathogenicity (OddsPath) for VUS in PMS2 we used a set of clinically classified PMS2 variants supplemented by inactivating variants that were generated by an in cellulo genetic screen, as proxies for cancer-predisposing variants. Validation of this OddsPath revealed high predictive values for benign and predisposing PMS2 VUS. We conclude that the OddsPath provides an integral metric that, following the other, higher penetrance, MMR proteins MSH2, MSH6 and MLH1 can be incorporated as strong evidence type into the upcoming criteria for MMR gene VUS classification of the American College of Medical Genetics and Genomics and the Association for Molecular Pathology (ACMG/AMP).


Subject(s)
Colorectal Neoplasms, Hereditary Nonpolyposis , Mismatch Repair Endonuclease PMS2 , Colorectal Neoplasms, Hereditary Nonpolyposis/genetics , Colorectal Neoplasms, Hereditary Nonpolyposis/pathology , DNA Mismatch Repair/genetics , DNA-Binding Proteins/genetics , Genetic Testing/methods , Humans , Mismatch Repair Endonuclease PMS2/genetics , MutL Protein Homolog 1/genetics , MutS Homolog 2 Protein/genetics
3.
CRISPR J ; 2(6): 406-416, 2019 12.
Article in English | MEDLINE | ID: mdl-31742432

ABSTRACT

CRISPR-Cas9 has quickly become the method of choice for genome editing, with multiple publications describing technical advances and novel applications. It has been widely adopted as a tool for basic research and has significant translational and clinical potential. However, its usage has outpaced the establishment of essential and rigorous controls for unwanted off-target effects, manifested as small mutations, large deletions of target loci, or large-scale chromosomal rearrangements. A common application of CRISPR-Cas9 is as a tool for creating isogenic cell-line models to study the effects of precise mutations, or variants, on disease traits. Here, we describe the effect of standard CRISPR-Cas9 mutagenesis protocols on well characterized cancer cell lines. We demonstrate that commonly used methods for detecting correctly mutated clones fail to uncover large-scale rearrangements. We show that simple cytogenetic methods can be used to identify clones carrying chromosomal abnormalities and large mutations at target loci. These methods are quick and cost-efficient, and we suggest that such controls should be performed prior to publication of studies based on novel CRISPR-Cas9 mutated cancer cell lines.


Subject(s)
Chromosomal Instability/genetics , Cytogenetic Analysis/methods , Gene Editing/methods , CRISPR-Cas Systems/genetics , Cell Line, Tumor , Chromosomal Instability/physiology , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , Gene Rearrangement/genetics , Humans , Mutagenesis/genetics , Mutation , Neoplasms/genetics , RNA, Guide, Kinetoplastida/genetics , Sequence Deletion/genetics
4.
J Pathol ; 245(3): 283-296, 2018 07.
Article in English | MEDLINE | ID: mdl-29604063

ABSTRACT

Genomic instability, which is a hallmark of cancer, is generally thought to occur in the middle to late stages of tumourigenesis, following the acquisition of permissive molecular aberrations such as TP53 mutation or whole genome doubling. Tumours with somatic POLE exonuclease domain mutations are notable for their extreme genomic instability (their mutation burden is among the highest in human cancer), distinct mutational signature, lymphocytic infiltrate, and excellent prognosis. To what extent these characteristics are determined by the timing of POLE mutations in oncogenesis is unknown. Here, we have shown that pathogenic POLE mutations are detectable in non-malignant precursors of endometrial and colorectal cancer. Using genome and exome sequencing, we found that multiple driver mutations in POLE-mutant cancers show the characteristic POLE mutational signature, including those in genes conventionally regarded as initiators of tumourigenesis. In POLE-mutant cancers, the proportion of monoclonal predicted neoantigens was similar to that in other cancers, but the absolute number was much greater. We also found that the prominent CD8+ T-cell infiltrate present in POLE-mutant cancers was evident in their precursor lesions. Collectively, these data indicate that somatic POLE mutations are early, quite possibly initiating, events in the endometrial and colorectal cancers in which they occur. The resulting early onset of genomic instability may account for the striking immune response and excellent prognosis of these tumours, as well as their early presentation. © 2018 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.


Subject(s)
Adenocarcinoma/genetics , Antigens, Neoplasm/genetics , Colorectal Neoplasms/genetics , DNA Polymerase II/genetics , Endometrial Neoplasms/genetics , Mutation , Poly-ADP-Ribose Binding Proteins/genetics , Adenocarcinoma/enzymology , Adenocarcinoma/immunology , Adenocarcinoma/pathology , Antigens, Neoplasm/immunology , Antigens, Neoplasm/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/immunology , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Colorectal Neoplasms/enzymology , Colorectal Neoplasms/immunology , Colorectal Neoplasms/pathology , DNA Polymerase II/metabolism , Databases, Genetic , Endometrial Neoplasms/enzymology , Endometrial Neoplasms/immunology , Endometrial Neoplasms/pathology , Female , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic , Genetic Predisposition to Disease , Genomic Instability , Humans , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Male , Middle Aged , Neoplasm Grading , Neoplasm Staging , Phenotype , Poly-ADP-Ribose Binding Proteins/metabolism , Tumor Microenvironment , Whole Genome Sequencing
5.
Clin Cancer Res ; 24(13): 3197-3203, 2018 07 01.
Article in English | MEDLINE | ID: mdl-29559562

ABSTRACT

Purpose: Pathogenic POLE proofreading domain mutations are found in many malignancies where they are associated with ultramutation and favorable prognosis. The extent to which this prognosis depends on their sensitivity to adjuvant treatment is unknown, as is the optimal therapy for advanced-staged or recurrent POLE-mutant cancers.Experimental Design: We examined the recurrence-free survival of women with POLE-mutant and POLE-wild-type endometrial cancers (EC) in the observation arm of the randomized PORTEC-1 endometrial cancer trial (N = 245 patients with stage I endometrial cancer for analysis). Sensitivity to radiotherapy and selected chemotherapeutics was compared between Pole-mutant mouse-derived embryonic stem (mES) cells, generated using CRISPR-Cas9 (Pole mutations D275A/E275A, and cancer-associated P286R, S297F, V411L) and isogenic wild-type cell lines.Results: In the observation arm of the PORTEC-1 trial (N = 245), women with POLE-mutant endometrial cancers (N = 16) had an improved recurrence-free survival (10-year recurrence-free survival 100% vs. 80.1% for POLE-wild-type; HR, 0.143; 95% confidence interval, 0.001-0.996; P = 0.049). Pole mutations did not increase sensitivity to radiotherapy nor to chemotherapeutics in mES cells. In contrast, Pole-mutant cells displayed significantly increased sensitivity to cytarabine and fludarabine (IC50Pole P286R-mutant vs. wild-type: 0.05 vs. 0.17 µmol/L for cytarabine, 4.62 vs. 11.1 µmol/L for fludarabine; P < 0.001 for both comparisons).Conclusions: The favorable prognosis of POLE-mutant cancers cannot be explained by increased sensitivity to currently used adjuvant treatments. These results support studies exploring minimization of adjuvant therapy for early-stage POLE-mutant cancers, including endometrial and colorectal cancers. Conversely, POLE mutations result in hypersensitivity to nucleoside analogues, suggesting the use of these compounds as a potentially effective targeted treatment for advanced-stage POLE-mutant cancers. Clin Cancer Res; 24(13); 3197-203. ©2018 AACR.


Subject(s)
Biomarkers, Tumor , DNA Polymerase II/genetics , Drug Resistance, Neoplasm/genetics , Mutation , Neoplasms/genetics , Poly-ADP-Ribose Binding Proteins/genetics , Protein Interaction Domains and Motifs/genetics , Radiation Tolerance/genetics , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Chemotherapy, Adjuvant , DNA Polymerase II/chemistry , Female , Humans , Male , Neoplasm Staging , Neoplasms/mortality , Neoplasms/pathology , Neoplasms/therapy , Poly-ADP-Ribose Binding Proteins/chemistry , Prognosis , Radiotherapy, Adjuvant , Treatment Outcome
6.
Nat Commun ; 7: 11111, 2016 Apr 05.
Article in English | MEDLINE | ID: mdl-27045317

ABSTRACT

How chemotherapy affects carcinoma genomes is largely unknown. Here we report whole-exome and deep sequencing of 30 paired oesophageal adenocarcinomas sampled before and after neo-adjuvant chemotherapy. Most, but not all, good responders pass through genetic bottlenecks, a feature associated with higher mutation burden pre-treatment. Some poor responders pass through bottlenecks, but re-grow by the time of surgical resection, suggesting a missed therapeutic opportunity. Cancers often show major changes in driver mutation presence or frequency after treatment, owing to outgrowth persistence or loss of sub-clones, copy number changes, polyclonality and/or spatial genetic heterogeneity. Post-therapy mutation spectrum shifts are also common, particularly C>A and TT>CT changes in good responders or bottleneckers. Post-treatment samples may also acquire mutations in known cancer driver genes (for example, SF3B1, TAF1 and CCND2) that are absent from the paired pre-treatment sample. Neo-adjuvant chemotherapy can rapidly and profoundly affect the oesophageal adenocarcinoma genome. Monitoring molecular changes during treatment may be clinically useful.


Subject(s)
Adenocarcinoma/genetics , Antineoplastic Agents/therapeutic use , Clonal Evolution/drug effects , DNA, Neoplasm/genetics , Esophageal Neoplasms/genetics , Neoadjuvant Therapy , Neoplasm Proteins/genetics , Neoplasm Recurrence, Local/genetics , Adenocarcinoma/drug therapy , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Adult , Aged , Cyclin D2/genetics , Cyclin D2/metabolism , DNA Copy Number Variations/drug effects , DNA, Neoplasm/metabolism , Esophageal Neoplasms/drug therapy , Esophageal Neoplasms/metabolism , Esophageal Neoplasms/pathology , Exome , Female , Gene Expression Regulation, Neoplastic , Genetic Heterogeneity , Histone Acetyltransferases/genetics , Histone Acetyltransferases/metabolism , Humans , Male , Middle Aged , Mutation/drug effects , Neoplasm Proteins/metabolism , Neoplasm Recurrence, Local/drug therapy , Neoplasm Recurrence, Local/metabolism , Neoplasm Recurrence, Local/pathology , Phosphoproteins/genetics , Phosphoproteins/metabolism , RNA Splicing Factors , Ribonucleoprotein, U2 Small Nuclear/genetics , Ribonucleoprotein, U2 Small Nuclear/metabolism , Sequence Analysis, DNA , TATA-Binding Protein Associated Factors/genetics , TATA-Binding Protein Associated Factors/metabolism , Transcription Factor TFIID/genetics , Transcription Factor TFIID/metabolism
7.
Nat Rev Cancer ; 16(2): 71-81, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26822575

ABSTRACT

Although it has long been recognized that the exonucleolytic proofreading activity intrinsic to the replicative DNA polymerases Pol δ and Pol ε is essential for faithful replication of DNA, evidence that defective DNA polymerase proofreading contributes to human malignancy has been limited. However, recent studies have shown that germline mutations in the proofreading domains of Pol δ and Pol ε predispose to cancer, and that somatic Pol ε proofreading domain mutations occur in multiple sporadic tumours, where they underlie a phenotype of 'ultramutation' and favourable prognosis. In this Review, we summarize the current understanding of the mechanisms and consequences of polymerase proofreading domain mutations in human malignancies, and highlight the potential utility of these variants as novel cancer biomarkers and therapeutic targets.


Subject(s)
DNA-Directed DNA Polymerase/metabolism , Mutation , Neoplasms/genetics , DNA Polymerase II/genetics , DNA Polymerase II/metabolism , DNA Polymerase III/genetics , DNA Polymerase III/metabolism , DNA Replication , DNA-Directed DNA Polymerase/genetics , Endometrial Neoplasms/genetics , Female , Genetic Predisposition to Disease , Humans , Neoplasms/immunology , Neoplasms/pathology , Poly-ADP-Ribose Binding Proteins , Prognosis , Saccharomyces cerevisiae/genetics
8.
Lancet Gastroenterol Hepatol ; 1(3): 207-216, 2016 11.
Article in English | MEDLINE | ID: mdl-28404093

ABSTRACT

BACKGROUND: Precision cancer medicine depends on defining distinct tumour subgroups using biomarkers that may occur at very modest frequencies. One such subgroup comprises patients with exceptionally mutated (ultramutated) cancers caused by mutations that impair DNA polymerase epsilon (POLE) proofreading. METHODS: We examined the association of POLE proofreading domain mutation with clinicopathological variables and immune response in colorectal cancers from clinical trials (VICTOR, QUASAR2, and PETACC-3) and colorectal cancer cohorts (Leiden University Medical Centre 1 and 2, Oslo 1 and 2, Bern, AMC-AJCC-II, and Epicolon-1). We subsequently investigated its association with prognosis in stage II/III colorectal cancer by Cox regression of pooled individual patient data from more than 4500 cases from these studies. FINDINGS: Pathogenic somatic POLE mutations were detected in 66 (1·0%) of 6517 colorectal cancers, and were mutually exclusive with mismatch repair deficiency (MMR-D) in the 6277 cases for whom both markers were determined (none of 66 vs 833 [13·4%] of 6211; p<0·0001). Compared with cases with wild-type POLE, cases with POLE mutations were younger at diagnosis (median 54·5 years vs 67·2 years; p<0·0001), were more frequently male (50 [75·8%] of 66 vs 3577 [55·5%] of 6445; p=0·0010), more frequently had right-sided tumour location (44 [68·8%] of 64 vs 2463 [39·8%] of 6193; p<0·0001), and were diagnosed at an earlier disease stage (p=0·006, χ2 test for trend). Compared with mismatch repair proficient (MMR-P) POLE wild-type tumours, POLE-mutant colorectal cancers displayed increased CD8+ lymphocyte infiltration and expression of cytotoxic T-cell markers and effector cytokines, similar in extent to that observed in immunogenic MMR-D cancers. Both POLE mutation and MMR-D were associated with significantly reduced risk of recurrence compared with MMR-P colorectal cancers in multivariable analysis (HR 0·34 [95% CI 0·11-0·76]; p=0·0060 and 0·72 [0·60-0·87]; p=0·00035), although the difference between the groups was not significant. INTERPRETATION: POLE proofreading domain mutations identify a subset of immunogenic colorectal cancers with excellent prognosis. This association underscores the importance of rare biomarkers in precision cancer medicine, but also raises important questions about how to identify and implement them in practice. FUNDING: Cancer Research UK, Academy of Medical Sciences, Health Foundation, EU, ERC, NIHR, Wellcome Trust, Dutch Cancer Society, Dutch Digestive Foundation.


Subject(s)
Biomarkers, Tumor/genetics , Colorectal Neoplasms/genetics , DNA Polymerase II/genetics , Poly-ADP-Ribose Binding Proteins/genetics , Adult , Aged , Aged, 80 and over , Colorectal Neoplasms/diagnosis , Colorectal Neoplasms/immunology , Colorectal Neoplasms/mortality , Female , Humans , Male , Middle Aged , Mutation , Prognosis , Retrospective Studies , Survival Analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...