Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
Clin Cancer Res ; 30(16): 3378-3387, 2024 Aug 15.
Article in English | MEDLINE | ID: mdl-38860976

ABSTRACT

Replication repair deficiency (RRD) is a pan-cancer mechanism characterized by abnormalities in the DNA mismatch repair (MMR) system due to pathogenic variants in the PMS2, MSH6, MSH2, or MLH1 genes, and/or in the polymerase-proofreading genes POLE and POLD1. RRD predisposition syndromes (constitutional MMR deficiency, Lynch, and polymerase proofreading-associated polyposis) share overlapping phenotypic and biological characteristics. Moreover, cancers stemming from germline defects of one mechanism can acquire somatic defects in another, leading to complete RRD. Here we describe the recent advances in the diagnostics, surveillance, and clinical management for children with RRD syndromes. For patients with constitutional MMR deficiency, new data combining clinical insights and cancer genomics have revealed genotype-phenotype associations and helped in the development of novel functional assays, diagnostic guidelines, and surveillance recommendations. Recognition of non-gastrointestinal/genitourinary malignancies, particularly aggressive brain tumors, in select children with Lynch and polymerase proofreading-associated polyposis syndromes harboring an RRD biology have led to new management considerations. Additionally, universal hypermutation and microsatellite instability have allowed immunotherapy to be a paradigm shift in the treatment of RRD cancers independent of their germline etiology. These advances have also stimulated a need for expert recommendations about genetic counseling for these patients and their families. Future collaborative work will focus on newer technologies such as quantitative measurement of circulating tumor DNA and functional genomics to tailor surveillance and clinical care, improving immune surveillance; develop prevention strategies; and deliver these novel discoveries to resource-limited settings to maximize benefits for patients globally.


Subject(s)
DNA Repair-Deficiency Disorders , Humans , Child , DNA Repair-Deficiency Disorders/genetics , DNA Repair-Deficiency Disorders/diagnosis , Young Adult , Adolescent , DNA Mismatch Repair/genetics , DNA Replication/genetics , Genetic Predisposition to Disease , Neoplastic Syndromes, Hereditary/genetics , Neoplastic Syndromes, Hereditary/therapy , Neoplastic Syndromes, Hereditary/diagnosis , Colorectal Neoplasms, Hereditary Nonpolyposis/genetics , Colorectal Neoplasms, Hereditary Nonpolyposis/diagnosis , Colorectal Neoplasms, Hereditary Nonpolyposis/therapy , Microsatellite Instability
2.
Bull Cancer ; 109(1): 76-82, 2022 Jan.
Article in French | MEDLINE | ID: mdl-34799081

ABSTRACT

PARP inhibitors are effective in different types of tumors such as ovarian, breast, prostate and pancreatic cancer. Many studies are in progress and may lead to prescription evolution. PARP inhibitors prescription is almost reserved to patients with a constitutional BRCA mutation or a somatic BRCA alteration or a tumor with a deficiency in homologous recombination. Nowadays, the diagnosis of homologous recombination deficit, HRD, is possible with the prescription of a myChoice CDx (Myriad) test. PARP inhibitors are studied in association with chemotherapy and targeted therapies but also with radiotherapy and with immune checkpoint inhibitors. Access to PARP inhibitors is challenged with the emergence of resistance mechanism. Various trials are now studying the possibility of reversing these resistance mechanisms.


Subject(s)
DNA Repair-Deficiency Disorders/diagnosis , Drug Resistance, Neoplasm , Homologous Recombination , Poly(ADP-ribose) Polymerase Inhibitors/therapeutic use , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , DNA Damage , Drug Resistance, Neoplasm/genetics , Female , Genes, BRCA1 , Genes, BRCA2 , Humans , Indazoles/therapeutic use , Indoles/therapeutic use , Male , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/genetics , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/genetics , Phthalazines/therapeutic use , Piperazines/therapeutic use , Piperidines/therapeutic use , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/genetics , Recombinational DNA Repair
3.
J Med Genet ; 58(9): 648-652, 2021 09.
Article in English | MEDLINE | ID: mdl-32843487

ABSTRACT

Pathogenic variants in BRCA1 gene in heterozygous state are known to be associated with breast-ovarian cancer susceptibility; however, biallelic variants cause a phenotype recognised as Fanconi anaemia complementation group S. Due to its rarity, medical management and preventive screening measures are insufficiently understood. Here, we present nine individuals (one new and eight previously presented) with biallelic variants in BRCA1 gene, to delineate clinical features in comparison with other chromosome instability syndromes and understand the patients' health risk. Features seen in these 9 individuals (7 females/2 males) include prenatal and postnatal growth failure (9/9), microcephaly (9/9), hypo/hyperpigmented lesions (9/9), facial dysmorphism (9/9), mild developmental delay (8/9) and early-onset solid tumours (5/9). None presented bone marrow failure or immunodeficiency. Individuals with biallelic variants in BRCA1 also showed chromosomal instability by mitomycin and diepoxybutane test. The phenotype caused by biallelic BRCA1 variants is best framed between Fanconi anaemia and Nijmegen syndrome, yet distinct due to lack of bone marrow failure and immunodeficiency. We hypothesise that disease class should be reframed and medical management in people with biallelic variants in BRCA1 should emphasise on detection of solid tumour development and avoiding exposure to ionising radiation.


Subject(s)
BRCA1 Protein/genetics , DNA Repair-Deficiency Disorders/diagnosis , DNA Repair-Deficiency Disorders/genetics , Genetic Association Studies , Genetic Predisposition to Disease , Mutation , Phenotype , Alleles , Biomarkers , Genetic Association Studies/methods , Humans , Male , Pedigree , Symptom Assessment
4.
J Clin Res Pediatr Endocrinol ; 12(3): 315-318, 2020 09 02.
Article in English | MEDLINE | ID: mdl-31752481

ABSTRACT

Progeria syndrome is a rare disorder in childhood which causes accelerated systemic aging. Due to the accelerated aging process, disorders which normally occur only in old age will appear in these children at a much younger age. We report two children with progeria syndrome, in whom fulminant diabetes mellitus manifested at a very early age.


Subject(s)
Diabetes Mellitus/etiology , Progeria/complications , Progeria/diagnosis , Adolescent , Age Factors , Age of Onset , Child , DNA Repair-Deficiency Disorders/complications , DNA Repair-Deficiency Disorders/diagnosis , Diabetes Mellitus/diagnosis , Fatal Outcome , Female , Humans , Male
5.
Nat Rev Dis Primers ; 5(1): 64, 2019 09 19.
Article in English | MEDLINE | ID: mdl-31537806

ABSTRACT

Fanconi anaemia (FA), ataxia telangiectasia (A-T), Nijmegen breakage syndrome (NBS) and Bloom syndrome (BS) are clinically distinct, chromosome instability (or breakage) disorders. Each disorder has its own pattern of chromosomal damage, with cells from these patients being hypersensitive to particular genotoxic drugs, indicating that the underlying defect in each case is likely to be different. In addition, each syndrome shows a predisposition to cancer. Study of the molecular and genetic basis of these disorders has revealed mechanisms of recognition and repair of DNA double-strand breaks, DNA interstrand crosslinks and DNA damage during DNA replication. Specialist clinics for each disorder have provided the concentration of expertise needed to tackle their characteristic clinical problems and improve outcomes. Although some treatments of the consequences of a disorder may be possible, for example, haematopoietic stem cell transplantation in FA and NBS, future early intervention to prevent complications of disease will depend on a greater understanding of the roles of the affected DNA repair pathways in development. An important realization has been the predisposition to cancer in carriers of some of these gene mutations.


Subject(s)
DNA Repair-Deficiency Disorders/diagnosis , DNA Repair-Deficiency Disorders/genetics , Ataxia Telangiectasia/diagnosis , Ataxia Telangiectasia/genetics , Ataxia Telangiectasia/physiopathology , Bloom Syndrome/diagnosis , Bloom Syndrome/genetics , Bloom Syndrome/physiopathology , DNA Damage/genetics , DNA Repair-Deficiency Disorders/physiopathology , Fanconi Anemia/diagnosis , Fanconi Anemia/genetics , Fanconi Anemia/physiopathology , Humans , Nijmegen Breakage Syndrome/diagnosis , Nijmegen Breakage Syndrome/genetics , Nijmegen Breakage Syndrome/physiopathology
6.
Brain Dev ; 41(2): 150-157, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30301590

ABSTRACT

OBJECTIVES: Defects in DNA damage responses or repair mechanisms cause numerous rare inherited diseases, referred to as "DNA-repair defects" or "DNA damage deficiency", characterized by neurodegeneration, immunodeficiency, and/or cancer predisposition. Early accurate diagnosis is important for informing appropriate clinical management; however, diagnosis is frequently challenging and can be delayed, due to phenotypic heterogeneity. Comprehensive genomic analysis could overcome this disadvantage. The objectives of this study were to determine the prevalence of ataxia-telangiectasia (A-T) and A-T-like DNA-repair defects in Japan and to determine the utility of comprehensive genetic testing of presumptively diagnosed patients in facilitating early diagnosis. METHODS: A nationwide survey of diseases presumably caused by DNA-repair defects, including A-T, was performed. Additionally, comprehensive next-generation sequencing (NGS) analysis, targeting known disease-causing genes, was conducted. RESULTS: Sixty-three patients with A-T or other diseases with characteristics of DNA-repair defects were identified. Thirty-four patients were genetically or clinically definitively diagnosed with A-T (n = 22) or other DNA-repair defects (n = 12). Genetic analysis of 17 presumptively diagnosed patients revealed one case of ataxia with oculomotor apraxia type 1 (AOA1); one ataxia with oculomotor apraxia type 2 (AOA2); two types of autosomal dominant spinocerebellar ataxia (SCA5, SCA29); two CACNA1A-related ataxias; one microcephaly with or without chorioretinopathy, lymphedema, or mental retardation (MCLMR); and one autosomal dominant KIF1A-related disorder with intellectual deficit, cerebellar atrophy, spastic paraparesis, and optic nerve atrophy. The diagnostic yield was 58.8%. CONCLUSION: Comprehensive genetic analysis of targeted known disease-causing genes by NGS is a powerful diagnostic tool for subjects with indistinguishable neurological phenotypes resembling DNA-repair defects.


Subject(s)
Ataxia Telangiectasia/epidemiology , Ataxia Telangiectasia/genetics , DNA Repair-Deficiency Disorders/epidemiology , DNA Repair-Deficiency Disorders/genetics , Adolescent , Adult , Asian People/genetics , Ataxia Telangiectasia/diagnosis , Child , Child, Preschool , DNA Repair-Deficiency Disorders/diagnosis , Early Diagnosis , Female , Genetic Predisposition to Disease , Genetic Testing , Humans , Japan/epidemiology , Male , Middle Aged , Young Adult
7.
J Natl Cancer Inst ; 110(7): 704-713, 2018 07 01.
Article in English | MEDLINE | ID: mdl-29788099

ABSTRACT

Defective DNA repair is a common hallmark of cancer. Homologous recombination is a DNA repair pathway of clinical interest due to the sensitivity of homologous recombination-deficient cells to poly-ADP ribose polymerase (PARP) inhibitors. The measurement of homologous recombination deficiency (HRD) in cancer is therefore vital to the appropriate design of clinical trials incorporating PARP inhibitors. However, methods to identify HRD in tumors are varied and controversial. Understanding existing and new methods to measure HRD is important to their appropriate use in clinical trials and practice. The aim of this review is to summarize the biology and clinical validation of current methods to measure HRD, to aid decision-making for patient stratification and translational research in PARP inhibitor trials. We discuss the current clinical development of PARP inhibitors, along with established indicators for HRD such as germline BRCA1/2 mutation status and clinical response to platinum-based therapy. We then examine newer assays undergoing clinical validation, including 1) somatic mutations in homologous recombination genes, 2) "genomic scar" assays using array-based comparative genomic hybridization (aCGH), single nucleotide polymorphism (SNP) analysis or mutational signatures derived from next-generation sequencing, 3) transcriptional profiles of HRD, and 4) phenotypic or functional assays of protein expression and localization. We highlight the strengths and weaknesses of each of these assays, for consideration during the design of studies involving PARP inhibitors.


Subject(s)
Biomarkers, Tumor , DNA Repair-Deficiency Disorders/diagnosis , Homologous Recombination/genetics , Neoplasms/genetics , Biomarkers, Tumor/genetics , DNA Repair/genetics , DNA Repair-Deficiency Disorders/genetics , Decision Support Techniques , Genetic Testing/methods , Humans , Neoplasms/diagnosis , Neoplasms/drug therapy , Neoplasms/pathology , Patient Selection , Poly(ADP-ribose) Polymerase Inhibitors/therapeutic use , Validation Studies as Topic
8.
J Allergy Clin Immunol ; 141(1): 322-328.e10, 2018 01.
Article in English | MEDLINE | ID: mdl-28392333

ABSTRACT

BACKGROUND: Rare DNA breakage repair disorders predispose to infection and lymphoreticular malignancies. Hematopoietic cell transplantation (HCT) is curative, but coadministered chemotherapy or radiotherapy is damaging because of systemic radiosensitivity. We collected HCT outcome data for Nijmegen breakage syndrome, DNA ligase IV deficiency, Cernunnos-XRCC4-like factor (Cernunnos-XLF) deficiency, and ataxia-telangiectasia (AT). METHODS: Data from 38 centers worldwide, including indication, donor, conditioning regimen, graft-versus-host disease, and outcome, were analyzed. Conditioning was classified as myeloablative conditioning (MAC) if it contained radiotherapy or alkylators and reduced-intensity conditioning (RIC) if no alkylators and/or 150 mg/m2 fludarabine or less and 40 mg/kg cyclophosphamide or less were used. RESULTS: Fifty-five new, 14 updated, and 18 previously published patients were analyzed. Median age at HCT was 48 months (range, 1.5-552 months). Twenty-nine patients underwent transplantation for infection, 21 had malignancy, 13 had bone marrow failure, 13 received pre-emptive transplantation, 5 had multiple indications, and 6 had no information. Twenty-two received MAC, 59 received RIC, and 4 were infused; information was unavailable for 2 patients. Seventy-three of 77 patients with DNA ligase IV deficiency, Cernunnos-XLF deficiency, or Nijmegen breakage syndrome received conditioning. Survival was 53 (69%) of 77 and was worse for those receiving MAC than for those receiving RIC (P = .006). Most deaths occurred early after transplantation, suggesting poor tolerance of conditioning. Survival in patients with AT was 25%. Forty-one (49%) of 83 patients experienced acute GvHD, which was less frequent in those receiving RIC compared with those receiving MAC (26/56 [46%] vs 12/21 [57%], P = .45). Median follow-up was 35 months (range, 2-168 months). No secondary malignancies were reported during 15 years of follow-up. Growth and developmental delay remained after HCT; immune-mediated complications resolved. CONCLUSION: RIC HCT resolves DNA repair disorder-associated immunodeficiency. Long-term follow-up is required for secondary malignancy surveillance. Routine HCT for AT is not recommended.


Subject(s)
DNA Breaks, Double-Stranded , DNA Repair-Deficiency Disorders/genetics , DNA Repair-Deficiency Disorders/therapy , DNA Repair , Hematopoietic Stem Cell Transplantation , Adolescent , Alleles , Child , Child, Preschool , DNA Repair-Deficiency Disorders/diagnosis , DNA Repair-Deficiency Disorders/mortality , Female , Follow-Up Studies , Graft vs Host Disease/diagnosis , Graft vs Host Disease/etiology , Hematopoietic Stem Cell Transplantation/adverse effects , Hematopoietic Stem Cell Transplantation/methods , Humans , Infant , Kaplan-Meier Estimate , Male , Mutation , Prognosis , Treatment Outcome , Virus Diseases , Young Adult
9.
Clin Cancer Res ; 23(11): e23-e31, 2017 Jun 01.
Article in English | MEDLINE | ID: mdl-28572264

ABSTRACT

DNA repair syndromes are heterogeneous disorders caused by pathogenic variants in genes encoding proteins key in DNA replication and/or the cellular response to DNA damage. The majority of these syndromes are inherited in an autosomal-recessive manner, but autosomal-dominant and X-linked recessive disorders also exist. The clinical features of patients with DNA repair syndromes are highly varied and dependent on the underlying genetic cause. Notably, all patients have elevated risks of syndrome-associated cancers, and many of these cancers present in childhood. Although it is clear that the risk of cancer is increased, there are limited data defining the true incidence of cancer and almost no evidence-based approaches to cancer surveillance in patients with DNA repair disorders. This article is the product of the October 2016 AACR Childhood Cancer Predisposition Workshop, which brought together experts from around the world to discuss and develop cancer surveillance guidelines for children with cancer-prone disorders. Herein, we focus on the more common of the rare DNA repair disorders: ataxia telangiectasia, Bloom syndrome, Fanconi anemia, dyskeratosis congenita, Nijmegen breakage syndrome, Rothmund-Thomson syndrome, and Xeroderma pigmentosum. Dedicated syndrome registries and a combination of basic science and clinical research have led to important insights into the underlying biology of these disorders. Given the rarity of these disorders, it is recommended that centralized centers of excellence be involved directly or through consultation in caring for patients with heritable DNA repair syndromes. Clin Cancer Res; 23(11); e23-e31. ©2017 AACRSee all articles in the online-only CCR Pediatric Oncology Series.


Subject(s)
DNA Repair-Deficiency Disorders/genetics , DNA Repair/genetics , Early Detection of Cancer , Neoplasms/genetics , Ataxia Telangiectasia/diagnosis , Ataxia Telangiectasia/genetics , Bloom Syndrome/diagnosis , Bloom Syndrome/genetics , Child , DNA Repair-Deficiency Disorders/diagnosis , DNA Repair-Deficiency Disorders/pathology , Fanconi Anemia/diagnosis , Fanconi Anemia/genetics , Humans , Neoplasms/diagnosis , Neoplasms/pathology , Xeroderma Pigmentosum/diagnosis , Xeroderma Pigmentosum/genetics
10.
Clin Exp Dermatol ; 42(5): 523-526, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28543586

ABSTRACT

Phototherapy is a useful noninvasive therapy, but it can induce cutaneous malignant tumours, including squamous cell carcinoma (SCC) and basal cell carcinoma (BCC). We report on a 79-year-old man who had long-standing mycosis fungoides for 40 years, which had been treated with psoralen ultraviolet A therapy for 37 years at a dose of approximately 5000 J/cm2 . Approximately 6 years before presentation, numerous types of cutaneous malignancies, including actinic keratosis, BCC and SCC, had begun to develop all over the patient's body. We hypothesized that he was experiencing a pathogenesis similar to patients with xeroderma pigmentosum (XP), and we therefore assessed his DNA repair capacity. Based on these investigations, the patient was eventually diagnosed as non-XP, even though we detected that his DNA repair capacity was slightly lower than that of normal controls, which may have led to the skin cancers. We speculate that multiple skin malignancies can be induced by long-term phototherapy in patients with slightly impaired DNA repair capacity.


Subject(s)
DNA Repair-Deficiency Disorders/diagnosis , Mycosis Fungoides/radiotherapy , Neoplasms, Radiation-Induced , Skin Neoplasms/pathology , Ultraviolet Therapy/adverse effects , Aged , Carcinoma, Basal Cell/etiology , Carcinoma, Basal Cell/pathology , Carcinoma, Squamous Cell/etiology , DNA Repair-Deficiency Disorders/complications , Humans , Male , Melanoma/etiology , Melanoma/pathology , Skin Neoplasms/etiology , Skin Neoplasms/radiotherapy
11.
J Pathol ; 242(2): 165-177, 2017 06.
Article in English | MEDLINE | ID: mdl-28299801

ABSTRACT

Homologous recombination (HR) DNA repair-deficient (HRD) breast cancers have been shown to be sensitive to DNA repair targeted therapies. Burgeoning evidence suggests that sporadic breast cancers, lacking germline BRCA1/BRCA2 mutations, may also be HRD. We developed a functional ex vivo RAD51-based test to identify HRD primary breast cancers. An integrated approach examining methylation, gene expression, and whole-exome sequencing was employed to ascertain the aetiology of HRD. Functional HRD breast cancers displayed genomic features of lack of competent HR, including large-scale state transitions and specific mutational signatures. Somatic and/or germline genetic alterations resulting in bi-allelic loss-of-function of HR genes underpinned functional HRD in 89% of cases, and were observed in only one of the 15 HR-proficient samples tested. These findings indicate the importance of a comprehensive genetic assessment of bi-allelic alterations in the HR pathway to deliver a precision medicine-based approach to select patients for therapies targeting tumour-specific DNA repair defects. Copyright © 2017 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Subject(s)
BRCA1 Protein/genetics , BRCA2 Protein/genetics , Breast Neoplasms/genetics , DNA Repair-Deficiency Disorders/genetics , Rad51 Recombinase/genetics , Recombinational DNA Repair , Adult , Aged , Aged, 80 and over , Breast Neoplasms/diagnosis , Breast Neoplasms, Male/diagnosis , Breast Neoplasms, Male/genetics , DNA Repair-Deficiency Disorders/diagnosis , Female , Germ-Line Mutation , Homologous Recombination , Humans , Loss of Heterozygosity , Male , Middle Aged , Mutation , Young Adult
12.
Gastroenterology ; 151(5): 870-878.e3, 2016 11.
Article in English | MEDLINE | ID: mdl-27443823

ABSTRACT

BACKGROUND & AIMS: Colonoscopy provides incomplete protection from colorectal cancer (CRC), but determinants of post-colonoscopy CRC are not well understood. We compared clinical features and molecular characteristics of CRCs diagnosed at different time intervals after a previous colonoscopy. METHODS: We performed a population-based, cross-sectional study of incident CRC cases in Denmark (2007-2011), categorized as post-colonoscopy or detected during diagnostic colonoscopy (in patients with no prior colonoscopy). We compared prevalence of proximal location and DNA mismatch repair deficiency (dMMR) in CRC tumors, relative to time since previous colonoscopy, using logistic regression and cubic splines to assess temporal variation. RESULTS: Of 10,365 incident CRCs, 725 occurred after colonoscopy examinations (7.0%). These were more often located in the proximal colon (odds ratio [OR], 2.34; 95% confidence interval [CI], 1.90-2.89) and were more likely to have dMMR (OR, 1.26; 95% CI, 1.00-1.59), but were less likely to be metastatic at presentation (OR, 0.65; 95% CI, 0.48-0.89) compared with CRCs diagnosed in patients with no prior colonoscopy. The highest proportions of proximal and/or dMMR tumors were observed in CRCs diagnosed 3-6 years after colonoscopy, but these features were still more frequent among cancers diagnosed up to 10 years after colonoscopy. The relative excess of dMMR tumors was most pronounced in distal cancers. In an analysis of 85 cases detected after colonoscopy, we found BRAF mutations in 23% of tumors and that 7% of cases had features of Lynch syndrome. Colonoscopy exams were incomplete in a higher proportion of cases diagnosed within <1 year (in 38%) than in those diagnosed within 1-10 years after colonoscopy (16%). CONCLUSIONS: In a study of incident CRC cases in Denmark, we observed that tumors found in patients who have undergone colonoscopy are more often proximal and have dMMR compared to CRCs detected in patients without previous colonoscopies. The excess of right-sided tumors and the modest independent effects of dMMR reinforce the importance of proper colonoscopic examination of the proximal large bowel.


Subject(s)
Adenocarcinoma/diagnosis , Adenocarcinoma/genetics , Adenoma/diagnosis , Adenoma/genetics , Colonoscopy , Colorectal Neoplasms/diagnosis , Colorectal Neoplasms/genetics , Adenocarcinoma/epidemiology , Adenocarcinoma/pathology , Adenoma/epidemiology , Adenoma/pathology , Adult , Aged , Aged, 80 and over , Colorectal Neoplasms/epidemiology , Colorectal Neoplasms/pathology , Cross-Sectional Studies , DNA Mismatch Repair , DNA Repair-Deficiency Disorders/diagnosis , DNA Repair-Deficiency Disorders/epidemiology , Denmark/epidemiology , Female , Humans , Incidence , Logistic Models , Male , Middle Aged , Registries , Time Factors
14.
Br J Cancer ; 108(10): 2079-87, 2013 May 28.
Article in English | MEDLINE | ID: mdl-23652311

ABSTRACT

BACKGROUND: Microsatellite instability (MSI) is a molecular phenotype due to defective DNA mismatch repair (MMR) system. It is used to predict outcome of colorectal tumours and to screen tumours for Lynch syndrome (LS). A pentaplex panel composed of five mononucleotide markers has been largely recommended for determination of the MSI status. However, its sensitivity may be taken in default in occasional situations. The aim of the study was to optimise this panel for the detection of MSI. METHODS: We developed an assay allowing co-amplification of six mononucleotide repeat markers (BAT25, BAT26, BAT40, NR21, NR22, NR27) and one polymorphic dinucleotide marker (D3S1260) in a single reaction. Performances of the new panel were evaluated on a cohort of patients suspected of LS. RESULTS: We demonstrate that our assay is technically as easy to use as the pentaplex assay. The hexaplex panel shows similar performances for the identification of colorectal and non-MSH6-deficient tumours. On the other hand, the hexaplex panel has higher sensitivity for the identification of MSH6-deficient tumours (94.7% vs 84.2%) and MMR-deficient tumours other than colorectal cancer (92.9% vs 85.7%). CONCLUSION: The hexaplex panel could thus be an attractive alternative to the pentaplex panel for the identification of patients with LS.


Subject(s)
Biomarkers, Tumor , DNA Mismatch Repair/genetics , Early Detection of Cancer/methods , Microsatellite Repeats , Neoplasms/diagnosis , Adult , Biomarkers, Tumor/analysis , Biomarkers, Tumor/genetics , Case-Control Studies , DNA Repair-Deficiency Disorders/diagnosis , DNA Repair-Deficiency Disorders/genetics , Female , Fluorescence , Genes, Neoplasm , Humans , Microsatellite Instability , Microsatellite Repeats/physiology , Middle Aged , Neoplasms/genetics , Polymerase Chain Reaction/methods
15.
Handb Clin Neurol ; 113: 1637-50, 2013.
Article in English | MEDLINE | ID: mdl-23622385

ABSTRACT

Deficient repair of ubiquitous errors in the genome risks faulty transcription or replication. Its direct and indirect phenotypic consequences are rare, complex, dementing, lethal disorders of children with inadequately understood overlapping genotypes and variable severity. Mutations of CSA or CSB responsible for impaired transcription-coupled repair cause Cockayne syndrome (CS). Its characteristics are (1) profound growth deficiency affecting all tissues, including the brain, (2) premature aging marked by cachexia, vascular disease, exocrine deficiency, and osteopenia, but not cancer, and (3) a selective degenerative disorder of central and peripheral myelin and by neuronal loss in the retina and inner ear, and in the cerebellum and basal ganglia where it is associated with calcification. Xeroderma pigmentosum (XP) can arise from mutations of at least eight genes involved in global genomic repair. Severe XPA and XPC cause innumerable carcinomas and melanomas in light-exposed eyes and skin, and enhanced risk of visceral cancers. XPA and XPD and others can cause childhood XP neurological disease with widespread neuronal loss, axonal sensorimotor neuropathy, and dwarfing. Four genes, including XPD, can cause trichothiodystrophy (TTD) with sulfur-deficient, brittle, tiger-tail hair, and growth and developmental inadequacy. CSB or XPD can cause the severe congenital cerebro-oculofacioskeletal (COFS) CS-like syndrome with joint contractures, cataracts, and early death. Three XP genes can also cause XP/CS complex. Much more needs to be learned about these and other disorders of DNA repair to enable prevention and treatment.


Subject(s)
DNA Repair-Deficiency Disorders/diagnosis , DNA Repair/genetics , DNA Repair-Deficiency Disorders/genetics , Humans , Phenotype
16.
Ann Dermatol Venereol ; 139 Suppl 3: S78-82, 2012 Nov.
Article in French | MEDLINE | ID: mdl-23260522

ABSTRACT

The occurrence of abnormally pigmented skin lesions is a common phenomenon and often associated with the influence of ultraviolet radiation (UV) and other sources of DNA damage. Pigmentary lesions induced by UV radiation and other sources of DNA damage occur in healthy individuals, but human diseases with defective DNA repair represent important models which allow the investigation of possible underlying molecular mechanisms leading to hypo- and hyperpigmentations. There are several hereditary diseases which are known to go along with genetic defects of DNA repair mechanisms comprising Xeroderma pigmentosum (XP), Cockayne syndrome (CS), Trichothiodystrophy (TTD), Werner syndrome (WS), Bloom syndrome (BS), Fanconi anemia (FA) and Ataxia telangiectasia (AT). These diseases share clinical characteristics including poikilodermatic skin changes such as hypo-and hyperpigmentation. Since UV radiation is the most common source of DNA damage which can cause pigmentary lesions both in healthy individuals and in patients with genetic deficiency in DNA repair, in the present article, we focus on pigmentary lesions in patients with XP as an example of a disease associated with genetic defects in DNA repair.


Subject(s)
DNA Damage/physiology , DNA Repair-Deficiency Disorders/diagnosis , DNA Repair-Deficiency Disorders/genetics , Photosensitivity Disorders/diagnosis , Photosensitivity Disorders/genetics , Pigmentation Disorders/diagnosis , Pigmentation Disorders/genetics , Ultraviolet Rays/adverse effects , Xeroderma Pigmentosum/diagnosis , Xeroderma Pigmentosum/genetics , Adult , Carcinoma, Basal Cell/diagnosis , Carcinoma, Basal Cell/genetics , Carcinoma, Basal Cell/physiopathology , Carcinoma, Basal Cell/prevention & control , Carcinoma, Squamous Cell/diagnosis , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/physiopathology , Carcinoma, Squamous Cell/prevention & control , Child , DNA Damage/genetics , DNA Repair-Deficiency Disorders/physiopathology , DNA Repair-Deficiency Disorders/prevention & control , Facial Neoplasms/diagnosis , Facial Neoplasms/genetics , Facial Neoplasms/physiopathology , Facial Neoplasms/prevention & control , Female , Genetic Predisposition to Disease/genetics , Humans , Male , Neoplasms, Multiple Primary/diagnosis , Neoplasms, Multiple Primary/genetics , Neoplasms, Multiple Primary/physiopathology , Neoplasms, Multiple Primary/prevention & control , Neoplasms, Radiation-Induced/diagnosis , Neoplasms, Radiation-Induced/genetics , Neoplasms, Radiation-Induced/physiopathology , Neoplasms, Radiation-Induced/prevention & control , Photosensitivity Disorders/physiopathology , Photosensitivity Disorders/prevention & control , Pigmentation Disorders/physiopathology , Pigmentation Disorders/prevention & control , Skin/physiopathology , Skin/radiation effects , Sunscreening Agents/administration & dosage , Syndrome , Xeroderma Pigmentosum/physiopathology
17.
Eur J Dermatol ; 22(4): 443-55, 2012.
Article in English | MEDLINE | ID: mdl-22436139

ABSTRACT

The human genome is constantly exposed to various sources of DNA damage. Ineffective protection from this damage leads to genetic instability which can ultimately give rise to somatic disease, causing mutations. Therefore our organism commands a number of highly conserved and effective mechanisms responsible for DNA repair. If these repair mechanisms are defective due to germline mutations in relevant genes, rare diseases with DNA repair deficiencies can arise. Today, a limited number of rare hereditary diseases characterized by genetic defects of DNA repair mechanisms is known, comprising ataxia telangiectasia, Nijmegen breakage syndrome, Werner syndrome, Bloom Syndrome, Fanconi anemia, xeroderma pigmentosum, Cockayne syndrome, trichothiodystrophy. Although heterogeneous in respect to selected symptoms, these rare disorders share many clinical features such as growth retardation, neurological disorders, premature ageing, skin alterations including abnormal pigmentation, telangiectasia, xerosis cutis, pathological wound healing as well as an increased risk of developing different types of cancer. Based on the clinical similarities of symptoms as well as the predominant diagnostic technology available, many of these rare disorders were formerly classified as genodermatoses with cancer predisposition or chromosomal breakage symptoms. These pathological conditions not only severely impair patients with these rare genetic diseases but also represent symptoms affecting large parts of the general population.


Subject(s)
DNA Repair-Deficiency Disorders , DNA Repair-Deficiency Disorders/diagnosis , DNA Repair-Deficiency Disorders/genetics , Humans , Phenotype
18.
Eur J Pediatr ; 170(3): 295-308, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21337117

ABSTRACT

The syndromic primary immunodeficiencies are disorders in which not only the immune system but also other organ systems are affected. Other features most commonly involve the ectodermal, skeletal, nervous, and gastrointestinal systems. Key in identifying syndromic immunodeficiencies is the awareness that increased susceptibility to infections or immune dysregulation in a patient known to have other symptoms or special features may hint at an underlying genetic syndrome. Because the extraimmune clinical features can be highly variable, it is more difficult establishing the correct diagnosis. Nevertheless, correct diagnosis at an early age is important because of the possible treatment options. Therefore, diagnostic work-up is best performed in a center with extensive expertise in this field, having immunologists and clinical geneticists, as well as adequate support from a specialized laboratory at hand. This paper provides the general pediatrician with the main clinical features that are crucial for the recognition of these syndromes.


Subject(s)
DNA Repair-Deficiency Disorders/diagnosis , Immunologic Deficiency Syndromes/diagnosis , Chromosome Aberrations , DNA Repair-Deficiency Disorders/genetics , DNA Repair-Deficiency Disorders/immunology , Disease Susceptibility , Humans , Immunologic Deficiency Syndromes/genetics , Immunologic Deficiency Syndromes/immunology , Infections/etiology
19.
Adv Exp Med Biol ; 685: 106-10, 2010.
Article in English | MEDLINE | ID: mdl-20687499

ABSTRACT

Although the term, "trichothiodystrophy" (TTD) refers to the hair anomalies in this group of patients, this is a heterogeneous, multisystem disease in which any or every organ in the body may be affected. Neuroectodermal derived tissues are particularly likely to be involved. This term was introduced by Price et alin 1980 to designate patients with sulfur-deficient brittle hair, which they recognized as a marker for this complex disease and designated it as a "neuroectodermal symptom complex". Patients with TTD have brittle hair and nails (associated with reduced content ofcysteine-rich matrix proteins), ichthyotic skin and physical and mental growth retardation. Ichthyosis is usually apparent at birth but much less so after the first few weeks of life. Other frequently associated features include ocular cataracts, infections and maternal complications related to pregnancy. Atrophy of subcutaneous fat may also be present. TTD occurs in a pattern of inheritance consistent with an autosomal recessive condition. The disease is extremely heterogeneous in severity and extent, with some patients showing no neurological deficiency. Others show severe, multisystem disease. Many patients die at a young age, most commonly due to infectious disease. TTD is part of a more broadly defined group of diseases identified as IBIDS (ichthyosis, brittle hair, impaired intelligence, decreased fertility and short stature). Photosensitive cases are also identified as PIBIDS (photosensitivity with IBIDS). Cases without manifest ichthyosis are also identified as PBIDS. These syndromes defy rigorous definition because of clinical variation between patients. The original two cases were described by Tay in oriental siblings, whose parents were first cousins; thus the disease is also known as Tay syndrome. The hairs in patients with TTD have a distinctive, diagnostically useful appearance on polarized light microscopy consisting of alternating light and dark bands known as the "tiger tail" anomaly. Diagnosis may be confirmed by sulfur content analysis ofhair shafts, which shows decreased sulfur and cysteine content. Approximately half of patients with TTD have photosensitivity, which correlates with a nudeotide excision repair (NER) defect. These patients are designated as having trichothiodystrophy-photosensitive (TTDP). Non-photosensitivepatients are designated as having trichothiodystrophy-nonphotosensitive (TTDN). Skin cancer is very rare in sun-sensitive TTD.


Subject(s)
DNA Repair-Deficiency Disorders , Nail Diseases , Trichothiodystrophy Syndromes , Animals , DNA Repair/genetics , DNA Repair-Deficiency Disorders/classification , DNA Repair-Deficiency Disorders/diagnosis , DNA Repair-Deficiency Disorders/genetics , DNA Repair-Deficiency Disorders/metabolism , DNA Repair-Deficiency Disorders/pathology , Female , Hair/metabolism , Hair/pathology , Hair Diseases/classification , Hair Diseases/diagnosis , Hair Diseases/genetics , Hair Diseases/metabolism , Hair Diseases/pathology , Humans , Male , Nail Diseases/classification , Nail Diseases/diagnosis , Nail Diseases/genetics , Nail Diseases/metabolism , Nail Diseases/pathology , Pregnancy , Pregnancy Complications/classification , Pregnancy Complications/diagnosis , Pregnancy Complications/genetics , Pregnancy Complications/metabolism , Pregnancy Complications/pathology , Skin Neoplasms/classification , Skin Neoplasms/diagnosis , Skin Neoplasms/genetics , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Sulfur/deficiency , Sulfur/metabolism , Trichothiodystrophy Syndromes/classification , Trichothiodystrophy Syndromes/diagnosis , Trichothiodystrophy Syndromes/genetics , Trichothiodystrophy Syndromes/metabolism , Trichothiodystrophy Syndromes/pathology
20.
Expert Rev Mol Med ; 12: e9, 2010 Mar 18.
Article in English | MEDLINE | ID: mdl-20298636

ABSTRACT

DNA-repair pathways recognise and repair DNA damaged by exogenous and endogenous agents to maintain genomic integrity. Defects in these pathways lead to replication errors, loss or rearrangement of genomic material and eventually cell death or carcinogenesis. The creation of diverse lymphocyte receptors to identify potential pathogens requires breaking and randomly resorting gene segments encoding antigen receptors. Subsequent repair of the gene segments utilises ubiquitous DNA-repair proteins. Individuals with defective repair pathways are found to be immunodeficient and many are radiosensitive. The role of repair proteins in the development of adaptive immunity by VDJ recombination, antibody isotype class switching and affinity maturation by somatic hypermutation has become clearer over the past few years, partly because of identification of the genes involved in human disease. We describe the mechanisms involved in the development of adaptive immunity relating to DNA repair, and the clinical consequences and treatment of the primary immunodeficiency resulting from such defects.


Subject(s)
DNA Repair-Deficiency Disorders/complications , Immunologic Deficiency Syndromes/complications , Adaptive Immunity , Animals , DNA Damage , DNA Repair/genetics , DNA Repair-Deficiency Disorders/diagnosis , DNA Repair-Deficiency Disorders/immunology , DNA Repair-Deficiency Disorders/therapy , Humans , Immunologic Deficiency Syndromes/immunology , Immunologic Deficiency Syndromes/therapy , Polymorphism, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL