Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 24
Filtrer
1.
Int J Oral Maxillofac Surg ; 51(6): 705-712, 2022 Jun.
Article de Anglais | MEDLINE | ID: mdl-34686398

RÉSUMÉ

This comprehensive literature review represents a summary of all cases of clear cell carcinoma (CCC) of the salivary glands that are documented in the literature. PubMed was used to collect available reports of CCC; 97 reports detailing 254 cases, published between 1983 and 2020, were retrieved. Clinically the tumor manifests most commonly as a painless mass or swelling on the palate, and the duration of symptoms prior to seeking care ranges from 1 week to 6 years. Metastasis, both local and distant, was reported in 13.9% of 202 total cases. Local tumor recurrence was present in 18.8% of the cases. By histopathology, CCC shows a mixture of growth patterns including solid (25.1%), nested (78.6%), sheet-like (23.5%), cords (46.1%), and trabeculae (42.4%). Immunohistochemical studies are positive for one or more cytokeratins (99.1%), PAS (95.1%), EMA (77.8%), and p63 (96.3%), but negative for S-100 (96.3%), PASD (91.1%), SMA (91.0%), and calponin (95.1%). Molecular features were reported in 115 cases; 96.0% were positive for an EWSR1 rearrangement by EWSR1 break apart FISH testing and 14.8% were positive for the rearrangement EWSR1-ATF1 tested by qPCR or targeted RNA sequencing. Clinical patterns and genetic studies imply that this tumor is the extraosseous counterpart of clear cell odontogenic carcinoma, an intraosseous odontogenic tumor of the jaws.


Sujet(s)
Adénocarcinome à cellules claires , Tumeurs des glandes salivaires , Adénocarcinome à cellules claires/génétique , Adénocarcinome à cellules claires/anatomopathologie , Humains , Hybridation fluorescente in situ , Récidive tumorale locale , Tumeurs des glandes salivaires/génétique , Tumeurs des glandes salivaires/anatomopathologie , Glandes salivaires
2.
Ann Oncol ; 30(9): 1417-1427, 2019 09 01.
Article de Anglais | MEDLINE | ID: mdl-31268127

RÉSUMÉ

BACKGROUND: NTRK1, NTRK2 and NTRK3 fusions are present in a plethora of malignancies across different histologies. These fusions represent the most frequent mechanism of oncogenic activation of these receptor tyrosine kinases, and biomarkers for the use of TRK small molecule inhibitors. Given the varying frequency of NTRK1/2/3 fusions, crucial to the administration of NTRK inhibitors is the development of optimal approaches for the detection of human cancers harbouring activating NTRK1/2/3 fusion genes. MATERIALS AND METHODS: Experts from several Institutions were recruited by the European Society for Medical Oncology (ESMO) Translational Research and Precision Medicine Working Group (TR and PM WG) to review the available methods for the detection of NTRK gene fusions, their potential applications, and strategies for the implementation of a rational approach for the detection of NTRK1/2/3 fusion genes in human malignancies. A consensus on the most reasonable strategy to adopt when screening for NTRK fusions in oncologic patients was sought, and further reviewed and approved by the ESMO TR and PM WG and the ESMO leadership. RESULTS: The main techniques employed for NTRK fusion gene detection include immunohistochemistry, fluorescence in situ hybridization (FISH), RT-PCR, and both RNA-based and DNA-based next generation sequencing (NGS). Each technique has advantages and limitations, and the choice of assays for screening and final diagnosis should also take into account the resources and clinical context. CONCLUSION: In tumours where NTRK fusions are highly recurrent, FISH, RT-PCR or RNA-based sequencing panels can be used as confirmatory techniques, whereas in the scenario of testing an unselected population where NTRK1/2/3 fusions are uncommon, either front-line sequencing (preferentially RNA-sequencing) or screening by immunohistochemistry followed by sequencing of positive cases should be pursued.


Sujet(s)
Glycoprotéines membranaires/isolement et purification , Tumeurs/diagnostic , Protéines de fusion oncogènes/isolement et purification , Récepteur trkA/isolement et purification , Récepteur trkB/isolement et purification , Récepteur trkC/isolement et purification , Marqueurs biologiques tumoraux/génétique , Marqueurs biologiques tumoraux/isolement et purification , Séquençage nucléotidique à haut débit , Humains , Immunohistochimie/normes , Hybridation fluorescente in situ/normes , Oncologie médicale/normes , Glycoprotéines membranaires/génétique , Tumeurs/traitement médicamenteux , Tumeurs/génétique , Protéines de fusion oncogènes/génétique , Médecine de précision/normes , Inhibiteurs de protéines kinases/usage thérapeutique , Récepteur trkA/génétique , Récepteur trkB/génétique , Récepteur trkC/génétique , /normes
3.
Int J Oral Maxillofac Surg ; 48(11): 1405-1410, 2019 Nov.
Article de Anglais | MEDLINE | ID: mdl-31227275

RÉSUMÉ

The purpose of this study was to summarize the currently published cases of clear cell odontogenic carcinoma (CCOC). The PubMed and Springer databases were used to collect available reports, searching for 'clear cell odontogenic carcinoma', 'CCOC', or 'clear cell ameloblastoma'. The search resulted in 75 reports detailing 107 cases between 1985 and 2018. Clinically the tumor manifests as a swelling in the posterior mandible (n=46), anterior mandible (n=33), and maxilla (n=28). Radiological analysis of 85 cases typically showed a poorly defined expansive radiolucency (n=83). Of the 70 patients with symptoms reported, 44 specified a swelling, 11 tooth mobility, seven gingival/periodontal issues, five numbness, and three decreased jaw opening. One patient presented with a neck mass. The duration of symptoms prior to seeking care was specified for 52 patients: 2 months to 1 year for 34 patients, 1-2 years for seven, 2-4 years for two, 4-7 years for six, and 7-12 years for three. The incidence of recurrence appeared to be 38 of the 88 cases where recurrence was reported. CCOC can be distinguished from other oral cancers by its distinctive histology and immunohistochemical characteristics and less aggressive behavior. Currently, treatment should be early and aggressive resection with clear surgical margins and long-term follow-up. The overall goal is to collect a cohort of patients.


Sujet(s)
Adénocarcinome à cellules claires , Tumeurs de la mâchoire , Tumeurs de la mandibule , Tumeurs odontogènes , Humains , Récidive tumorale locale
4.
Ann Oncol ; 30(7): 1121-1126, 2019 07 01.
Article de Anglais | MEDLINE | ID: mdl-30980071

RÉSUMÉ

BACKGROUND: In the ongoing phase I PROFILE 1001 study, crizotinib showed antitumor activity in patients with ROS1-rearranged advanced non-small-cell lung cancer (NSCLC). Here, we present updated antitumor activity, overall survival (OS) and safety data (additional 46.2 months follow-up) for patients with ROS1-rearranged advanced NSCLC from PROFILE 1001. PATIENTS AND METHODS: ROS1 status was determined by FISH or reverse transcriptase-polymerase chain reaction. All patients received crizotinib at a starting dose of 250 mg twice daily. RESULTS: Fifty-three patients received crizotinib, with a median duration of treatment of 22.4 months. At data cut-off, treatment was ongoing in 12 patients (23%). The objective response rate (ORR) was 72% [95% confidence interval (CI), 58% to 83%], including six confirmed complete responses and 32 confirmed partial responses; 10 patients had stable disease. Responses were durable (median duration of response 24.7 months; 95% CI, 15.2-45.3). ORRs were consistent across different patient subgroups. Median progression-free survival was 19.3 months (95% CI, 15.2-39.1). A total of 26 deaths (49%) occurred (median follow-up period of 62.6 months), and of the remaining 27 patients (51%), 14 (26%) were in follow-up at data cut-off. Median OS was 51.4 months (95% CI, 29.3 to not reached) and survival probabilities at 12, 24, 36, and 48 months were 79%, 67%, 53%, and 51%, respectively. No correlation was observed between OS and specific ROS1 fusion partner. Treatment-related adverse events (TRAEs) were mainly grade 1 or 2, per CTCAE v3.0. There were no grade ≥4 TRAEs and no TRAEs associated with permanent discontinuation. No new safety signals were reported with long-term crizotinib treatment. CONCLUSIONS: These findings serve as a new benchmark for OS in ROS1-rearranged advanced NSCLC, and continue to show the clinically meaningful benefit and safety of crizotinib in this molecular subgroup. TRIAL REGISTRATION NUMBER: ClinicalTrials.gov identifier NCT00585195.


Sujet(s)
Carcinome pulmonaire non à petites cellules/traitement médicamenteux , Crizotinib/usage thérapeutique , Réarrangement des gènes , Tumeurs du poumon/traitement médicamenteux , Protein-tyrosine kinases/génétique , Protéines proto-oncogènes/génétique , Adulte , Sujet âgé , Sujet âgé de 80 ans ou plus , Antinéoplasiques/usage thérapeutique , Carcinome pulmonaire non à petites cellules/génétique , Carcinome pulmonaire non à petites cellules/anatomopathologie , Études de cohortes , Femelle , Études de suivi , Humains , Tumeurs du poumon/génétique , Tumeurs du poumon/anatomopathologie , Mâle , Adulte d'âge moyen , Pronostic , Études rétrospectives , Taux de survie
5.
Ann Oncol ; 29(9): 1964-1971, 2018 09 01.
Article de Anglais | MEDLINE | ID: mdl-30010763

RÉSUMÉ

Background: In clinical trials of patients with anaplastic lymphoma kinase (ALK)-positive non-small-cell lung cancer (NSCLC) treated with crizotinib, evaluation of the relationship between the percentage of ALK-positive cells by fluorescence in situ hybridization (FISH)-particularly near the cut-off defining positive status-and clinical outcomes have been limited by small sample sizes. Patients and methods: Data were pooled from three large prospective trials (one single-arm and two randomized versus chemotherapy) of crizotinib in patients with ALK-positive NSCLC determined by Vysis ALK Break Apart FISH using a cut-off of ≥15% ALK-positive cells. Logistic regression and proportional hazards regression analyses were used to explore the association of percent ALK-positive cells with objective response and progression-free survival (PFS), respectively. Results: Of 11 081 screened patients, 1958 (18%) were ALK positive, 7512 (68%) were ALK negative, and 1540 (14%) were uninformative. Median percentage of ALK-positive cells was 58% in ALK-positive patients and 2% in ALK-negative patients. Of ALK-positive patients, 5% had 15%-19% ALK-positive cells; of ALK-negative patients, 2% had 10%-14% ALK-positive cells. Objective response rate for ALK-positive, crizotinib-treated patients with ≥20% ALK-positive cells was 56% (n = 700/1246), 55% (n = 725/1312) for those with ≥15% ALK-positive cells, and 38% for those with 15%-19% ALK-positive cells (n = 25/66). As a continuous variable, higher percentages of ALK-positive cells were estimated to be associated with larger differences in objective response and PFS between crizotinib and chemotherapy; however, tests for interaction between treatment and percentage of ALK-positive cells were not significant (objective response, P = 0.054; PFS, P = 0.17). Conclusions: Patients with ALK-positive NSCLC benefit from treatment with crizotinib across the full range of percentage of ALK-positive cells, supporting the clinical utility of the 15% cut-off. The small number of patients with scores near the cut-off warrant additional study given the potential for misclassification of ALK status due to technical or biologic reasons.


Sujet(s)
Kinase du lymphome anaplasique/analyse , Carcinome pulmonaire non à petites cellules/traitement médicamenteux , Crizotinib/usage thérapeutique , Tumeurs du poumon/traitement médicamenteux , Inhibiteurs de protéines kinases/usage thérapeutique , Adulte , Sujet âgé , Sujet âgé de 80 ans ou plus , Kinase du lymphome anaplasique/métabolisme , Carcinome pulmonaire non à petites cellules/mortalité , Carcinome pulmonaire non à petites cellules/anatomopathologie , Essais cliniques de phase II comme sujet , Essais cliniques de phase III comme sujet , Crizotinib/pharmacologie , Résistance aux médicaments antinéoplasiques , Femelle , Humains , Hybridation fluorescente in situ , Poumon/anatomopathologie , Tumeurs du poumon/mortalité , Tumeurs du poumon/anatomopathologie , Mâle , Adulte d'âge moyen , Survie sans progression , Études prospectives , Inhibiteurs de protéines kinases/pharmacologie , Essais contrôlés randomisés comme sujet , Jeune adulte
6.
Ann Oncol ; 29(3): 640-645, 2018 03 01.
Article de Anglais | MEDLINE | ID: mdl-29236940

RÉSUMÉ

Background: While deregulation of the cyclin D1-CDK4/6-retinoblastoma pathway is common in hormone receptor positive (HR+) breast cancer, Rb is usually intact in HR+ breast cancer, and targeted CDK 4/6 inhibitors that act upstream of Rb, are routinely being utilized in clinical practice. However, factors that can lead to clinical resistance to CDK 4/6 inhibitors are not known. Patients and methods: We identified patients who had pre- and post-genotyping in tissue and peripheral blood samples after receiving CDK 4/6 inhibitors. Genotyping was carried out in tumor tissue or blood collected before start of CDK 4/6 inhibitor and after disease progression on CDK 4/6 inhibitor, covering more than 90% of the coding region in RB1. Results: We identified detectable acquired RB1 mutations in circulating tumor DNA (ctDNA) after exposure to CDK4/6 inhibitor (palbociclib, palbociclib, ribociclib) for 5, 8, and 13 months, respectively, in three patients. The RB1 mutations included substitution in donor splicing site of exon 8 of the RB1 gene in patient #1; substitution in donor splicing site of exon 22 of RB1 gene, exon 19 deletion, exon 3 insertion in patient #2; and RB1 exon 16 H483Y mutation in patient #3. None of these RB1 mutations were present in the pre-CDK 4/6 specimen highlighting these molecular alterations, which lead to functional loss of Rb1, likely emerged under selective pressure from the CDK4/6 inhibitor potentially confering therapeutic resistance. Conclusion: This is the first clinical report to describe the emergence of somatic RB1 mutations after exposure to palbociclib or ribociclib, in patients with metastatic breast cancer. Further research is needed to validate these findings, identify how these mutations temporally emerge under selective pressure of CDK 4/6 inhibitor, and develop rational therapeutic strategies.


Sujet(s)
Antinéoplasiques/usage thérapeutique , Tumeurs du sein/traitement médicamenteux , Résistance aux médicaments antinéoplasiques/génétique , Protéines de liaison à la protéine du rétinoblastome/effets des médicaments et des substances chimiques , Protéines de liaison à la protéine du rétinoblastome/génétique , Ubiquitin-protein ligases/effets des médicaments et des substances chimiques , Ubiquitin-protein ligases/génétique , Sujet âgé , Aminopyridines/usage thérapeutique , Femelle , Génotype , Humains , Adulte d'âge moyen , Mutation/effets des médicaments et des substances chimiques , Pipérazines/usage thérapeutique , Inhibiteurs de protéines kinases/usage thérapeutique , Purines/usage thérapeutique , Pyridines/usage thérapeutique
7.
J Clin Endocrinol Metab ; 99(11): E2457-62, 2014 Nov.
Article de Anglais | MEDLINE | ID: mdl-25148236

RÉSUMÉ

BACKGROUND: The diagnosis of the follicular variant of papillary thyroid carcinoma (FVPTC) is increasingly common. Recent studies have suggested that FVPTC is heterogeneous and comprises multiple tumor types with distinct biological behaviors and underlying genetics. OBJECTIVES: The purpose of this work was to identify the prevalence of mutations and gene fusions in known oncogenes in a panel representative of the common spectrum of FVPTC diagnosed at an academic medical center and correlate the clinical and pathological features obtained at the initial diagnosis with the tumor genotype. MATERIALS AND METHODS: We performed SNaPshot genotyping on a panel of 129 FVPTCs of ≥1 cm for 90 point mutations or small deletions in known oncogenes and tumor suppressors and identified gene fusions using an anchored multiplex PCR assay targeting a panel of rearranged oncogenes. RESULTS: We identified a mutation or gene fusion in 70% (89 of 127) of cases. Mutations targeting the RAS family of oncogenes were the most frequently observed class of alterations, present in 36% (46 of 127) of cases, followed by BRAF mutation, present in 30% (38 of 127). We also detected oncogenic rearrangements not previously associated with FVPTC, including TFG-ALK and CREB3L2-PPARγ. BRAF mutation was significantly associated with unencapsulated tumor status. CONCLUSIONS: These data support the hypothesis that FVPTC is composed of distinct biological entities, with one class being identified by BRAF mutation and support the use of clinical genotyping assays that detect a diverse array of rearrangements involving ALK and PPARγ. Additional studies are necessary to identify genetic drivers in the 30% of FVPTCs with no known oncogenic alteration and to better predict behavior in tumors with known genotypes.


Sujet(s)
Carcinome papillaire folliculaire/génétique , Mutation , Fusion oncogène , Tumeurs de la thyroïde/génétique , Adulte , Sujet âgé , Carcinome papillaire folliculaire/anatomopathologie , Analyse de mutations d'ADN , Femelle , Humains , Mâle , Adulte d'âge moyen , Tumeurs de la thyroïde/anatomopathologie
8.
Br J Cancer ; 111(3): 430-6, 2014 Jul 29.
Article de Anglais | MEDLINE | ID: mdl-24960403

RÉSUMÉ

BACKGROUND: Current data suggest that platinum-based combination therapy is the standard first-line treatment for biliary tract cancer. EGFR inhibition has proven beneficial across a number of gastrointestinal malignancies; and has shown specific advantages among KRAS wild-type genetic subtypes of colon cancer. We report the combination of panitumumab with gemcitabine (GEM) and oxaliplatin (OX) as first-line therapy for KRAS wild-type biliary tract cancer. METHODS: Patients with histologically confirmed, previously untreated, unresectable or metastatic KRAS wild-type biliary tract or gallbladder adenocarcinoma with ECOG performance status 0-2 were treated with panitumumab 6 mg kg(-1), GEM 1000 mg m(-2) (10 mg m(-2) min(-1)) and OX 85 mg m(-2) on days 1 and 15 of each 28-day cycle. The primary objective was to determine the objective response rate by RECIST criteria v.1.1. Secondary objectives were to evaluate toxicity, progression-free survival (PFS), and overall survival. RESULTS: Thirty-one patients received at least one cycle of treatment across three institutions, 28 had measurable disease. Response rate was 45% and disease control rate was 90%. Median PFS was 10.6 months (95% CI 5-24 months) and median overall survival 20.3 months (95% CI 9-25 months). The most common grade 3/4 adverse events were anaemia 26%, leukopenia 23%, fatigue 23%, neuropathy 16% and rash 10%. CONCLUSIONS: The combination of gemcitabine, oxaliplatin and panitumumab in KRAS wild type metastatic biliary tract cancer showed encouraging efficacy, additional efforts of genetic stratification and targeted therapy is warranted in biliary tract cancer.


Sujet(s)
Adénocarcinome/traitement médicamenteux , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Tumeurs des voies biliaires/traitement médicamenteux , Tumeurs de la vésicule biliaire/traitement médicamenteux , Adénocarcinome/mortalité , Adénocarcinome/secondaire , Adulte , Sujet âgé , Anticorps monoclonaux/administration et posologie , Tumeurs des voies biliaires/mortalité , Tumeurs des voies biliaires/anatomopathologie , Désoxycytidine/administration et posologie , Désoxycytidine/analogues et dérivés , Survie sans rechute , Femelle , Tumeurs de la vésicule biliaire/mortalité , Tumeurs de la vésicule biliaire/anatomopathologie , Humains , Estimation de Kaplan-Meier , Métastase lymphatique , Mâle , Adulte d'âge moyen , Composés organiques du platine/administration et posologie , Oxaliplatine , Panitumumab , Protéines proto-oncogènes/génétique , Protéines proto-oncogènes p21(ras) , Résultat thérapeutique , Protéines G ras/génétique ,
9.
Ann Oncol ; 25(3): 552-563, 2014 Mar.
Article de Anglais | MEDLINE | ID: mdl-24265351

RÉSUMÉ

The fibroblast growth factor receptor (FGFR) cascade plays crucial roles in tumor cell proliferation, angiogenesis, migration and survival. Accumulating evidence suggests that in some tumor types, FGFRs are bona fide oncogenes to which cancer cells are addicted. Because FGFR inhibition can reduce proliferation and induce cell death in a variety of in vitro and in vivo tumor models harboring FGFR aberrations, a growing number of research groups have selected FGFRs as targets for anticancer drug development. Multikinase FGFR/vascular endothelial growth factor receptor (VEGFR) inhibitors have shown promising activity in breast cancer patients with FGFR1 and/or FGF3 amplification. Early clinical trials with selective FGFR inhibitors, which may overcome the toxicity constraints raised by multitarget kinase inhibition, are recruiting patients with known FGFR(1-4) status based on genomic screens. Preliminary signs of antitumor activity have been demonstrated in some tumor types, including squamous cell lung carcinomas. Rational combination of targeted therapies is expected to further increase the efficacy of selective FGFR inhibitors. Herein, we discuss unsolved questions in the clinical development of these agents and suggest guidelines for management of hyperphosphatemia, a class-specific mechanism-based toxicity. In addition, we propose standardized definitions for FGFR1 and FGFR2 gene amplification based on in situ hybridization methods. Extended access to next-generation sequencing platforms will facilitate the identification of diseases in which somatic FGFR(1-4) mutations, amplifications and fusions are potentially driving cancer cell viability, further strengthening the role of FGFR signaling in cancer biology and providing more possibilities for the therapeutic application of FGFR inhibitors.


Sujet(s)
Thérapie moléculaire ciblée , Tumeurs/traitement médicamenteux , Récepteur FGFR1/antagonistes et inhibiteurs , Récepteur FGFR1/génétique , Anticorps monoclonaux/usage thérapeutique , Facteur de croissance fibroblastique de type 3/génétique , Amplification de gène , Humains , Hyperphosphatémie/thérapie , Récepteur FGFR2/antagonistes et inhibiteurs , Récepteur FGFR2/génétique , Récepteur de type 3 des facteurs de croissance fibroblastique/antagonistes et inhibiteurs , Récepteur de type 3 des facteurs de croissance fibroblastique/génétique , Récepteur FGFR4/antagonistes et inhibiteurs , Récepteur FGFR4/génétique , Récepteurs aux facteurs de croissance endothéliale vasculaire/antagonistes et inhibiteurs
10.
Ann Oncol ; 22(12): 2616-2624, 2011 Dec.
Article de Anglais | MEDLINE | ID: mdl-22071650

RÉSUMÉ

BACKGROUND: Personalizing non-small-cell lung cancer (NSCLC) therapy toward oncogene addicted pathway inhibition is effective. Hence, the ability to determine a more comprehensive genotype for each case is becoming essential to optimal cancer care. METHODS: We developed a multiplexed PCR-based assay (SNaPshot) to simultaneously identify >50 mutations in several key NSCLC genes. SNaPshot and FISH for ALK translocations were integrated into routine practice as Clinical Laboratory Improvement Amendments-certified tests. Here, we present analyses of the first 589 patients referred for genotyping. RESULTS: Pathologic prescreening identified 552 (95%) tumors with sufficient tissue for SNaPshot; 51% had ≥1 mutation identified, most commonly in KRAS (24%), EGFR (13%), PIK3CA (4%) and translocations involving ALK (5%). Unanticipated mutations were observed at lower frequencies in IDH and ß-catenin. We observed several associations between genotypes and clinical characteristics, including increased PIK3CA mutations in squamous cell cancers. Genotyping distinguished multiple primary cancers from metastatic disease and steered 78 (22%) of the 353 patients with advanced disease toward a genotype-directed targeted therapy. CONCLUSIONS: Broad genotyping can be efficiently incorporated into an NSCLC clinic and has great utility in influencing treatment decisions and directing patients toward relevant clinical trials. As more targeted therapies are developed, such multiplexed molecular testing will become a standard part of practice.


Sujet(s)
Carcinome pulmonaire non à petites cellules/génétique , Génotype , Tumeurs du poumon/génétique , Réaction de polymérisation en chaine multiplex , Adulte , Sujet âgé , Sujet âgé de 80 ans ou plus , Marqueurs biologiques tumoraux/génétique , Carcinome pulmonaire non à petites cellules/traitement médicamenteux , Carcinome pulmonaire non à petites cellules/mortalité , Essais cliniques comme sujet , Tests diagnostiques courants , Femelle , Humains , Estimation de Kaplan-Meier , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/mortalité , Mâle , Adulte d'âge moyen , Techniques de diagnostic moléculaire , Thérapie moléculaire ciblée , Mutation , Jeune adulte
11.
Am J Transplant ; 11(7): 1464-77, 2011 Jul.
Article de Anglais | MEDLINE | ID: mdl-21668634

RÉSUMÉ

An idiopathic capillary leak syndrome ('engraftment syndrome') often occurs in recipients of hematopoietic cells, manifested clinically by transient azotemia and sometimes fever and fluid retention. Here, we report the renal pathology in 10 recipients of combined bone marrow and kidney allografts. Nine developed graft dysfunction on day 10-16 and renal biopsies showed marked acute tubular injury, with interstitial edema, hemorrhage and capillary congestion, with little or no interstitial infiltrate (≤10%) and marked glomerular and peritubular capillary (PTC) endothelial injury and loss by electron microscopy. Two had transient arterial endothelial inflammation; and 2 had C4d deposition. The cells in capillaries were primarily CD68(+) MPO(+) mononuclear cells and CD3(+) CD8(+) T cells, the latter with a high proliferative index (Ki67(+) ). B cells (CD20(+) ) and CD4(+) T cells were not detectable, and NK cells were rare. XY FISH showed that CD45(+) cells in PTCs were of recipient origin. Optimal treatment remains to be defined; two recovered without additional therapy, six were treated with anti-rejection regimens. Except for one patient, who later developed thrombotic microangiopathy and one with acute humoral rejection, all fully recovered within 2-4 weeks. Graft endothelium is the primary target of this process, attributable to as yet obscure mechanisms, arising during leukocyte recovery.


Sujet(s)
Atteinte rénale aigüe/étiologie , Transplantation de moelle osseuse/effets indésirables , Syndrome de fuite capillaire/étiologie , Transplantation rénale/effets indésirables , Atteinte rénale aigüe/anatomopathologie , Moelle osseuse/anatomopathologie , Transplantation de moelle osseuse/anatomopathologie , Syndrome de fuite capillaire/anatomopathologie , Créatinine/sang , Femelle , Rejet du greffon/anatomopathologie , Humains , Immunohistochimie , Hybridation fluorescente in situ , Transplantation rénale/anatomopathologie , Numération des leucocytes , Mâle
13.
Neurosci Lett ; 430(3): 241-5, 2008 Jan 17.
Article de Anglais | MEDLINE | ID: mdl-18055113

RÉSUMÉ

Familial amyotrophic lateral sclerosis (ALS) accounts for 10% of all ALS. Approximately 20% of cases are due to mutations in the Cu/Zn superoxide dismutase gene (SOD1). In North America, SOD1(A4V) is the most common SOD1 mutation. Carriers of the SOD1(A4V) mutation share a common phenotype with rapid disease progression and death on average occurring at 1.4 years (versus 3-5 years with other dominant SOD1 mutations). Previous studies of SOD1(A4V) carriers identified a common haplotype around the SOD1 locus, suggesting a common founder for most SOD1(A4V) patients. In the current study we sequenced the entire common haplotypic region around SOD1 to test the hypothesis that polymorphisms in either previously undescribed coding regions or non-coding regions around SOD1 are responsible for the more aggressive phenotype in SOD1(A4V)-mediated ALS. We narrowed the conserved region around the SOD1 gene in SOD1(A4V) ALS to 2.8Kb and identified five novel SNPs therein. None of these variants was specifically found in all SOD1(A4V) patients. It therefore appears likely that the aggressive nature of the SOD1(A4V) mutation is not a result of a modifying factor within the region around the SOD1 gene. Founder analysis estimates that the A4V mutation occurred 540 generations (approximately 12,000 years) ago (95% CI 480-700). The conserved minimal haplotype is statistically more similar to Asian than European population DNA sets, suggesting that the A4V mutation arose in native Asian-Americans who reached the Americas through the Bering Strait.


Sujet(s)
Sclérose latérale amyotrophique/génétique , Effet fondateur , Liaison génétique/génétique , Prédisposition génétique à une maladie/génétique , Mutation/génétique , Superoxide dismutase/génétique , Sclérose latérale amyotrophique/ethnologie , Asie/ethnologie , /génétique , Asiatiques/génétique , Analyse de mutations d'ADN , Femelle , Dépistage génétique , Génotype , Géographie , Haplotypes/génétique , Humains , Mâle , Adulte d'âge moyen , Phénotype , Polymorphisme génétique/génétique , Superoxide dismutase-1 , Taux de survie/tendances
14.
J Virol ; 74(21): 9836-44, 2000 Nov.
Article de Anglais | MEDLINE | ID: mdl-11024110

RÉSUMÉ

The multifunctional simian and human immunodeficiency virus (SIV and HIV) Nef proteins are important for virulence. We studied the importance of selected Nef functions using an SIV Nef with mutations in two regions that are required for CD4 downregulation. This Nef mutant is defective for downregulating CD4 and, in addition, for enhancing SIV infectivity and induction of SIV replication from infected quiescent peripheral blood mononuclear cells, but not for other known functions, including downregulation of class I major histocompatibility complex (MHC) cell surface expression. Replication of SIV containing this Nef variant in rhesus monkeys was attenuated early during infection. Subsequent increases in viral load coincided with selection of reversions and second-site compensatory changes in Nef. Our results indicate that the surfaces of Nef that mediate CD4 downregulation and the enhancement of virion infectivity are critical for SIV replication in vivo. Furthermore, these findings indicate that class I MHC downregulation by Nef is not sufficient for SIV virulence early in infection.


Sujet(s)
Antigènes CD4/métabolisme , Régulation négative , Produits du gène nef/métabolisme , Virus de l'immunodéficience simienne/pathogénicité , Virion/pathogénicité , Substitution d'acide aminé , Animaux , Produits du gène nef/composition chimique , Produits du gène nef/génétique , Macaca mulatta , Mutation , Analyse de séquence d'ADN , Syndrome d'immunodéficience acquise du singe/physiopathologie , Syndrome d'immunodéficience acquise du singe/virologie , Virus de l'immunodéficience simienne/physiologie , Charge virale , Virulence , Réplication virale
15.
J Virol ; 74(12): 5691-701, 2000 Jun.
Article de Anglais | MEDLINE | ID: mdl-10823877

RÉSUMÉ

Simian immunodeficiency virus (SIV) and human immunodeficiency virus type 1 (HIV-1) Nef proteins are related regulatory proteins that share several functions, including the ability to downregulate class I major histocompatibility complex (MHC) and CD4 expression on the cell surface and to alter T-cell-receptor-initiated signal transduction in T cells. We compared the mechanisms used by SIV mac239 Nef and HIV-1 Nef to downregulate class I MHC and found that the ability of SIV Nef to downregulate class I MHC requires a unique C-terminal region of the SIV mac239 Nef molecule which is not found in HIV-1 Nef. Interestingly, mutation of the PxxP motif in SIV Nef, unlike in HIV-1 Nef, does not affect class I MHC downregulation. We also found that downregulation of class I MHC by SIV Nef requires a conserved tyrosine in the cytoplasmic domain of the class I MHC heavy chain and involves accelerated endocytosis of class I complexes, as previously found with HIV-1 Nef. Thus, while SIV and HIV-1 Nef proteins use a similar mechanism to downregulate class I MHC expression, they have evolved different surfaces for molecular interactions with cell factors that regulate class I MHC traffic. Mutations in the C-terminal domain of SIV mac239 Nef selectively disrupt class I MHC downregulation, having no detectable effect on other functions of Nef, such as the downregulation of CD4 and CD3 surface expression, the stimulation of SIV virion infectivity, and the induction of SIV replication from T cells infected in the absence of stimulation. The resulting mutants will be useful reagents for studying the importance of class I MHC downregulation for SIV replication and AIDS pathogenesis in infected rhesus macaques.


Sujet(s)
Produits du gène nef/composition chimique , Produits du gène nef/métabolisme , VIH-1 (Virus de l'Immunodéficience Humaine de type 1) , Antigènes d'histocompatibilité de classe I/métabolisme , Virus de l'immunodéficience simienne , Complexe protéique adaptateur, sous-unités alpha , Protéines adaptatrices du transport vésiculaire , Motifs d'acides aminés , Séquence d'acides aminés , Animaux , Sites de fixation , Antigènes CD3/métabolisme , Antigènes CD4/métabolisme , Cellules cultivées , Régulation négative , Endocytose , Produits du gène nef/génétique , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/génétique , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/physiologie , Antigènes d'histocompatibilité de classe I/composition chimique , Antigènes d'histocompatibilité de classe I/génétique , Humains , Macaca mulatta , Protéines membranaires/métabolisme , Données de séquences moléculaires , Mutation/génétique , Liaison aux protéines , Virus de l'immunodéficience simienne/génétique , Virus de l'immunodéficience simienne/physiologie , Lymphocytes T/métabolisme , Lymphocytes T/virologie , Tyrosine/génétique , Tyrosine/métabolisme , Réplication virale , Produits du gène nef du virus de l'immunodéficience humaine
16.
J Virol ; 74(9): 4155-64, 2000 May.
Article de Anglais | MEDLINE | ID: mdl-10756028

RÉSUMÉ

SIVmac Nef contains two N-terminal tyrosines that were proposed to be part of an SH2-ligand domain and/or a tyrosine-based endocytosis signal and a putative SH3-ligand domain (P(104)xxP(107)). In the present study, we investigated the effects of combined mutations in these tyrosine and proline residues on simian immunodeficiency virus (SIV) Nef interactions with the cellular signal transduction and endocytic machinery. We found that mutation of Y(28)F, Y(39)F, P(104)A, and P(107)A (FFAA-Nef) had little effect on Nef functions such as the association with the cellular tyrosine kinase Src, downregulation of cell surface expression of CD4 and class I major histocompatibility complex, and enhancement of virion infectivity. However, mutations in the PxxP sequence reduced the ability of Nef to stimulate viral replication in primary lymphocytes. Three macaques infected with the SIVmac239 FFAA-Nef variant showed high viral loads during the acute phase of infection. Reversions in the mutated prolines were observed between 12 and 20 weeks postinfection. Importantly, reversion of A(107)-->P, which restored the ability of Nef to coprecipitate a 62-kDa phosphoprotein in in vitro kinase assays, did not precede the development of a high viral load. The Y(28)/Y(39)-->F(28)/F(39) substitutions did not revert. In conclusion, mutations in both the tyrosine residues and the putative SH3 ligand domain apparently do not disrupt major aspects of SIV Nef function in vivo.


Sujet(s)
Produits du gène nef/physiologie , Virus de l'immunodéficience simienne/physiologie , Tyrosine/physiologie , Réplication virale , Domaine d'homologie SRC , Animaux , Sites de fixation , Antigènes CD4/immunologie , Cellules COS , Lignée de cellules transformées , Régulation négative , Produits du gène nef/génétique , Produits du gène nef/immunologie , Antigènes d'histocompatibilité de classe I/immunologie , Humains , Macaca mulatta , Mutagenèse , Phosphorylation , Protein-Serine-Threonine Kinases/métabolisme , Virus de l'immunodéficience simienne/génétique , Tyrosine/métabolisme , Charge virale , p21-Activated Kinases
17.
J Infect Dis ; 181(1): 132-40, 2000 Jan.
Article de Anglais | MEDLINE | ID: mdl-10608759

RÉSUMÉ

A 36-bp deletion close to the 5' end of NEF that impaired Nef function was found in a long-term nonprogressor with human immunodeficiency virus type 1 (HIV-1) infection. Forms containing an adjacent duplication of 33 bp were also frequently observed. The duplication showed no homology to the deleted region but restored the overall length of the first variable loop of Nef. NEF alleles carrying the duplication were active in class I major histocompatibility complex (MHC-I) down-modulation and enhancement of virus infectivity. However, they showed little activity in CD4 down-regulation and were unable to stimulate viral replication in human peripheral blood mononuclear cells. Our study indicates that the enhancement of virion infectivity and the stimulation of HIV-1 replication in lymphocytes are distinct functions of Nef. Our findings also illustrate the capacity for repair of attenuating deletions in HIV-1 infection and suggest that a selective pressure for Nef-mediated MHC-I down-modulation and/or enhancement of virion infectivity exists.


Sujet(s)
Réparation de l'ADN , Gènes nef , Infections à VIH/génétique , Survivants à long terme d'une infection à VIH , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/génétique , Adulte , Allèles , Séquence d'acides aminés , Études cas-témoins , Duplication de gène , Produits du gène nef/génétique , Infections à VIH/épidémiologie , Antigènes d'histocompatibilité de classe I/biosynthèse , Homosexualité , Humains , Londres/épidémiologie , Mâle , Données de séquences moléculaires , Similitude de séquences d'acides aminés , Produits du gène nef du virus de l'immunodéficience humaine
18.
EMBO J ; 18(10): 2722-33, 1999 May 17.
Article de Anglais | MEDLINE | ID: mdl-10329619

RÉSUMÉ

The simian immunodeficiency virus (SIV) and human immunodeficiency virus type 1 (HIV-1) Nef proteins induce the endocytosis of CD4 and class I MHC molecules. Here we show that SIV Nef interacts with the AP-2 adaptor complex via two elements located in the N-terminal region of the Nef molecule, but only the N-distal element is required to induce CD4 endocytosis. This N-distal AP-2 targeting element contains no canonical endocytic signals and probably contacts the AP-2 complex via a novel interaction surface. The data support a model where SIV Nef induces CD4 endocytosis by promoting the normal interactions between the di-leucine sorting signal in the CD4 cytoplasmic domain and AP-2, but does not substitute for the CD4-AP-2 adaptor interaction. Neither element is important for the induction of class I MHC endocytosis, thus indicating that different mechanisms underlie the induction of class I MHC and CD4 endocytosis by Nef. In contrast to SIV Nef, HIV-1 Nef interacts with AP-2 via a surface containing a di-leucine endocytosis signal in the C-terminal disordered loop of Nef. The fact that genetic selection maintains similar molecular interactions via different surfaces in SIV and HIV-1 Nef proteins indicates that these interactions have critical roles for the viral life cycle in vivo.


Sujet(s)
Produits du gène nef/métabolisme , Protéines de tissu nerveux/métabolisme , Phosphoprotéines/métabolisme , Virus de l'immunodéficience simienne/métabolisme , Protéines adaptatrices du transport vésiculaire , Séquence d'acides aminés , Séquence nucléotidique , Antigènes CD4/métabolisme , Clathrine/métabolisme , Régulation négative , Endocytose , Fibroblastes , Produits du gène nef/composition chimique , Produits du gène nef/génétique , Protéines à fluorescence verte , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/métabolisme , Antigènes d'histocompatibilité de classe I/immunologie , Antigènes d'histocompatibilité de classe I/métabolisme , Humains , Cellules Jurkat , Protéines luminescentes , Données de séquences moléculaires , Mutation , Liaison aux protéines , Protéines de fusion recombinantes/génétique , Tyrosine/génétique , Produits du gène nef du virus de l'immunodéficience humaine
19.
Virology ; 257(1): 138-55, 1999 Apr 25.
Article de Anglais | MEDLINE | ID: mdl-10208928

RÉSUMÉ

Variants of the pathogenic SIVmac239 clone with changes in Nef were analyzed to assess the functional relevance of two highly conserved regions in Nef in vitro and in vivo. Changes in a region with an acidic charge (Aci-Nef), or a potential protein kinase C phosphorylation site (PKC-Nef), impaired the ability of Nef to down-regulate CD4 and MHC class I surface expression and to alter CD3-initiated signal transduction in Jurkat T cells. The Aci-Nef, but not the PKC-Nef, associated with the previously described p65 phosphoprotein. SIV containing Aci-Nef, but not SIV containing PKC-Nef, showed reduced infectivity and replication in cell culture systems. One of two rhesus macaques infected with the PKC-Nef mutant virus showed rapid reversion and progressed to disease. In the second animal no reversions and nonprogressive infection was observed. In one of two macaques infected with the Aci-Nef variant, the mutations were stable during the first 40 weeks after infection. Thereafter, variants evolved in which up to six of the eight mutated positions in Nef were reverted and functional activity in vitro was partially restored. These changes occurred concomitantly with increasing viral load and disease progression. The second animal infected with the Aci-Nef variant showed no reversions and remained asymptomatic. Our study suggests that the acidic region and conserved PKC phosphorylation site in Nef are important for SIV replication in rhesus macaques and for several in vitro Nef functions. An almost wild-type activity in in vitro infectivity and replication assays seems insufficient to confer a full nef-positive phenotype in vivo.


Sujet(s)
Produits du gène nef/métabolisme , Protéine kinase C/métabolisme , Virus de l'immunodéficience simienne/physiologie , Réplication virale , Séquence d'acides aminés , Animaux , Antigènes CD4/biosynthèse , Cellules COS , Produits du gène nef/génétique , VIH-1 (Virus de l'Immunodéficience Humaine de type 1) , Antigènes d'histocompatibilité de classe I/biosynthèse , Concentration en ions d'hydrogène , Macaca mulatta , Données de séquences moléculaires , Mutagenèse dirigée , Phosphorylation , Virus de l'immunodéficience simienne/génétique , Propriétés de surface , Produits du gène nef du virus de l'immunodéficience humaine
20.
J Virol ; 73(4): 2790-7, 1999 Apr.
Article de Anglais | MEDLINE | ID: mdl-10074126

RÉSUMÉ

The simian immunodeficiency virus macC8 (SIVmacC8) variant has been used in a European Community Concerted Action project to study the efficacy and safety of live attenuated SIV vaccines in a large number of macaques. The attenuating deletion in the SIVmacC8 nef-long terminal repeat region encompasses only 12 bp and is "repaired" in a subset of infected animals. It is unknown whether C8-Nef retains some activity. Since it seems important to use only well-characterized deletion mutants in live attenuated vaccine studies, we analyzed the relevance of the deletion, and the duplications and point mutations selected in infected macaques for Nef function in vitro. The deletion, affecting amino acids 143 to 146 (DMYL), resulted in a dramatic decrease in Nef stability and function. The initial 12-bp duplication resulted in efficient Nef expression and an intermediate phenotype in infectivity assays, but it did not significantly restore the ability of Nef to stimulate viral replication and to downmodulate CD4 and class I major histocompatibility complex cell surface expression. The additional substitutions however, which subsequently evolved in vivo, gradually restored these Nef functions. It was noteworthy that coinfection experiments in the T-lymphoid 221 cell line revealed that even SIVmac nef variants carrying the original 12-bp deletion readily outgrew an otherwise isogenic virus containing a 182-bp deletion in the nef gene. Thus, although C8-Nef is unstable and severely impaired in in vitro assays, it maintains some residual activity to stimulate viral replication.


Sujet(s)
Régulation de l'expression des gènes viraux , Gènes nef , Syndrome d'immunodéficience acquise du singe/virologie , Virus de l'immunodéficience simienne/génétique , Allèles , Séquence d'acides aminés , Animaux , Macaca , Données de séquences moléculaires , Alignement de séquences , Délétion de séquence
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE