Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
J Clin Oncol ; 41(26): 4236-4246, 2023 09 10.
Article in English | MEDLINE | ID: mdl-37379495

ABSTRACT

PURPOSE: Gilteritinib is a type 1 FLT3 inhibitor active as monotherapy for relapsed or refractory FLT3-mutated AML. We investigated the safety, tolerability, and efficacy of gilteritinib incorporated into intensive induction and consolidation chemotherapy, and as maintenance therapy for adult patients with newly diagnosed, non-favorable-risk AML. METHODS: In this phase IB study (2215-CL-0103; ClinicalTrials.gov identifier: NCT02236013), 103 participants were screened and 80 were allocated to treatment. The study was divided into four parts: dose escalation, dose expansion, investigation of alternate anthracycline and gilteritinib schedule, and continuous gilteritinib during consolidation. RESULTS: After dose escalation, 120 mg gilteritinib once daily was chosen for further study. There were 58 participants evaluable for response at this dose, 36 of whom harbored FLT3 mutations. For participants with FLT3-mutated AML, the composite complete response (CRc) rate was 89% (83% were conventional complete responses), all achieved after a single induction cycle. The median overall survival time was 46.1 months. Gilteritinib was well-tolerated in this context although the median time to count recovery during induction was approximately 40 days. Longer time-to-count recovery was associated with higher trough levels of gilteritinib, which, in turn, were associated with azole use. The recommended regimen is gilteritinib at a dose of 120 mg once daily from days 4 to 17 or 8 to 21 of a 7 + 3 induction with either idarubicin or daunorubicin and from day 1 continuously with high-dose cytarabine consolidation. Maintenance therapy with gilteritinib was well-tolerated. CONCLUSION: These results demonstrated the safety and tolerability of gilteritinib incorporated into an induction and consolidation chemotherapy regimen, and as single-agent maintenance therapy for patients with newly diagnosed FLT3-mutant AML. The data herein provide an important framework for the design of randomized trials comparing gilteritinib with other FLT3 inhibitors.


Subject(s)
Consolidation Chemotherapy , Leukemia, Myeloid, Acute , Adult , Humans , Neoplasm Recurrence, Local/drug therapy , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Idarubicin , Protein Kinase Inhibitors/therapeutic use , fms-Like Tyrosine Kinase 3/genetics , Mutation
2.
Blood ; 140(17): 1845-1857, 2022 10 27.
Article in English | MEDLINE | ID: mdl-35917453

ABSTRACT

Treatment results for patients with newly diagnosed FMS-like tyrosine kinase 3 (FLT3)-mutated (FLT3mut+) acute myeloid leukemia (AML) ineligible for intensive chemotherapy are disappointing. This multicenter, open-label, phase 3 trial randomized (2:1) untreated adults with FLT3mut+ AML ineligible for intensive induction chemotherapy to receive gilteritinib (120 mg/d orally) and azacitidine (GIL + AZA) or azacitidine (AZA) alone. The primary end point was overall survival (OS). At the interim analysis (August 26, 2020), a total of 123 patients were randomized to treatment (GIL + AZA, n = 74; AZA, n = 49). Subsequent AML therapy, including FLT3 inhibitors, was received by 20.3% (GIL + AZA) and 44.9% (AZA) of patients. Median OS was 9.82 (GIL + AZA) and 8.87 (AZA) months (hazard ratio, 0.916; 95% CI, 0.529-1.585; P = .753). The study was closed based on the protocol-specified boundary for futility. Median event-free survival was 0.03 month in both arms. Event-free survival defined by using composite complete remission (CRc) was 4.53 months for GIL + AZA and 0.03 month for AZA (hazard ratio, 0.686; 95% CI, 0.433-1.087; P = .156). CRc rates were 58.1% (GIL + AZA) and 26.5% (AZA) (difference, 31.4%; 95% CI, 13.1-49.7; P < .001). Adverse event (AE) rates were similar for GIL + AZA (100%) and AZA (95.7%); grade ≥3 AEs were 95.9% and 89.4%, respectively. Common AEs with GIL + AZA included pyrexia (47.9%) and diarrhea (38.4%). Gilteritinib steady-state trough concentrations did not differ between GIL + AZA and gilteritinib. GIL + AZA resulted in significantly higher CRc rates, although similar OS compared with AZA. Results support the safety/tolerability and clinical activity of upfront therapy with GIL + AZA in older/unfit patients with FLT3mut+ AML. This trial was registered at www.clinicaltrials.gov as #NCT02752035.


Subject(s)
Azacitidine , Leukemia, Myeloid, Acute , Adult , Humans , Aged , Azacitidine/adverse effects , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/diagnosis , Pyrazines/adverse effects
3.
Blood Adv ; 6(7): 2144-2155, 2022 04 12.
Article in English | MEDLINE | ID: mdl-35130342

ABSTRACT

The phase 3 Study of ASP2215 Versus Salvage Chemotherapy in Patients With Relapsed or Refractory Acute Myeloid Leukemia (AML) With FMS-like Tyrosine Kinase (FLT3) Mutation (ADMIRAL) trial demonstrated the superiority of the FLT3 inhibitor, gilteritinib, to salvage chemotherapy (SC) in patients with FLT3-mutated relapsed or refractory (R/R) AML. Baseline comutations, FLT3-internal tandem duplication (ITD) allelic ratio and length, and treatment-emergent mutations were analyzed in patients in the ADMIRAL trial. Baseline comutations were grouped according to gene subgroups (DNA methylation/hydroxymethylation, transcription, chromatin-spliceosome, receptor tyrosine kinase-Ras signaling, TP53-aneuploidy, NPM1, DNMT3A, DNMT3A/NPM1, WT-1, and IDH1/IDH2). Across all but 1 gene subgroup (TP53-aneuploidy), higher pretransplant response rates and a trend toward longer overall survival were observed with gilteritinib vs SC. Patients with DNMT3A/NPM1 comutations who received gilteritinib had the most favorable outcomes of any molecular subgroup analyzed. Survival outcomes with gilteritinib were not adversely affected by FLT3-ITD allelic ratio, FLT3-ITD length, or multiple FLT3-ITD mutations. Among patients who relapsed on gilteritinib, Ras/mitogen-activated protein kinase (MAPK) pathway and FLT3 F691L gene mutations were the most common mutational events associated with treatment resistance. However, the occurrence of Ras/MAPK pathway gene mutations at baseline did not preclude a clinical benefit from gilteritinib. Acquisition of multiple Ras/MAPK pathway gene mutations at relapse suggests a high level of pathway reactivation is needed to overcome the gilteritinib treatment effect. These findings provide insight into the R/R AML molecular profile and the impact of FLT3 inhibitors on mutational evolution associated with treatment resistance and benefit of gilteritinib across a wide spectrum of molecular and genetic subgroups in FLT3-mutated R/R AML. This trial was registered at www.clinicaltrials.gov as #NCT02421939.


Subject(s)
Leukemia, Myeloid, Acute , Pyrazines , Aniline Compounds/therapeutic use , Humans , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Protein Kinase Inhibitors/therapeutic use , Pyrazines/therapeutic use , fms-Like Tyrosine Kinase 3/genetics , fms-Like Tyrosine Kinase 3/therapeutic use
4.
Cancer Med ; 10(3): 797-805, 2021 02.
Article in English | MEDLINE | ID: mdl-33340276

ABSTRACT

The FLT3 inhibitor gilteritinib has clinical activity in patients with FLT3-mutated (FLT3mut+ ) relapsed/refractory (R/R) acute myeloid leukemia (AML). The impact of FLT3 mutation clearance and the achievement of composite complete remission (CRc) and complete remission/complete remission with partial hematologic recovery (CR/CRh) on overall survival (OS) in patients with FLT3mut+ R/R AML treated with single-agent gilteritinib in a phase 1/2 trial were evaluated. Using next-generation sequencing, a FLT3-ITD variant allele frequency of ≤10-4 was used to define FLT3-ITD clearance in patients with no morphologic leukemia (ie, CRc). A total of 108 patients with FLT3-ITD-positive (FLT3-ITD+) R/R AML were analyzed; 95 of these patients had received ≥80-mg/day gilteritinib. Ten of the 95 patients had FLT3-ITD clearance; eight of these 10 patients achieved CRc and were considered negative for measurable residual disease. There was a trend toward longer OS in patients who attained CRc with FLT3-ITD clearance (131.4 weeks) versus those who achieved CRc and did not have FLT3-ITD clearance (n = 41; 43.3 weeks; HR = 0.416; p = 0.066). Among patients treated with ≥80-mg/day gilteritinib who achieved CR/CRh (n = 24), seven had FLT3-ITD clearance. Among patients who received 120-mg/day gilteritinib, those who achieved CR/CRh had a longer median OS (70.6 weeks) and higher 52-week survival probability (66.7%) than patients who did not achieve CR/CRh (n = 71; median OS, 41.7 weeks; 52-week survival probability, 20.2%). Overall, these data suggest that gilteritinib can induce deep molecular responses in patients with FLT3-ITD+ R/R AML, and in the setting of CRc or CR/CRh, these responses may be associated with prolonged survival.


Subject(s)
Aniline Compounds/therapeutic use , Drug Resistance, Neoplasm/genetics , Leukemia, Myeloid, Acute/mortality , Mutation , Pyrazines/therapeutic use , fms-Like Tyrosine Kinase 3/genetics , Adult , Aged , Aged, 80 and over , Female , Follow-Up Studies , Humans , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Male , Middle Aged , Prognosis , Protein Kinase Inhibitors/therapeutic use , Survival Rate , Young Adult
5.
Blood Adv ; 4(3): 514-524, 2020 02 11.
Article in English | MEDLINE | ID: mdl-32040554

ABSTRACT

Gilteritinib is the first FMS-like tyrosine kinase 3 (FLT3) tyrosine kinase inhibitor (TKI) approved as monotherapy in acute myeloid leukemia with FLT3 internal tandem duplication and D835/I836 tyrosine kinase domain (TKD) mutations. Sequencing studies in patients have uncovered less common, noncanonical (NC) mutations in FLT3 and have implicated secondary TKD mutations in FLT3 TKI resistance. We report that gilteritinib is active against FLT3 NC and TKI resistance-causing mutations in vitro. A mutagenesis screen identified FLT3 F691L, Y693C/N, and G697S as mutations that confer moderate resistance to gilteritinib in vitro. Analysis of patients treated with gilteritinib revealed that 2/9 patients with preexisting NC FLT3 mutations responded and that secondary TKD mutations are acquired in a minority (5/31) of patients treated with gilteritinib. Four of 5 patients developed F691L mutations (all treated at <200 mg). These studies suggest that gilteritinib has broad activity against FLT3 mutations and limited vulnerability to resistance-causing FLT3 TKD mutations, particularly when used at higher doses.


Subject(s)
Protein Kinase Inhibitors , fms-Like Tyrosine Kinase 3 , Aniline Compounds , Humans , Mutation , Protein Kinase Inhibitors/pharmacology , Pyrazines , fms-Like Tyrosine Kinase 3/genetics
6.
Biochim Biophys Acta Mol Basis Dis ; 1863(5): 1115-1131, 2017 05.
Article in English | MEDLINE | ID: mdl-27639834

ABSTRACT

Obesity and its possible association with diseases including diabetes and cardiovascular diseases have been studied for decades for its impact on healthcare. Recent studies clearly indicate the need for developing accurate and reproducible methodologies for assessing body fat content and distribution. Body fat distribution plays a significant role in developing an insight in the underlying mechanisms in which adipose tissue is linked with various diseases. Among imaging technologies including computerized axial tomography (CAT or CT), magnetic resonance imaging (MRI), and magnetic resonance spectroscopy (MRS), MRI and MRS seem to be the best emerging techniques and together are being considered as the gold standard for body fat content and distribution. This paper reviews studies up to the present time involving different methodologies of these two emerging technologies and presents the basic concepts of MRI and MRS with required novel image analysis techniques in accurate, quantitative, and direct assessment of body fat content and distribution. This article is part of a Special Issue entitled: Oxidative Stress and Mitochondrial Quality in Diabetes/Obesity and Critical Illness Spectrum of Diseases - edited by P. Hemachandra Reddy.


Subject(s)
Adiposity , Magnetic Resonance Imaging , Obesity/diagnostic imaging , Obesity/metabolism , Tomography, X-Ray Computed , Humans
7.
Cereb Cortex ; 20(12): 2852-62, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20237243

ABSTRACT

Application of resting state functional connectivity magnetic resonance imaging (fcMRI) to the study of prematurely born infants enables assessment of the earliest forms of cerebral connectivity and characterization of its early development in the human brain. We obtained 90 longitudinal fcMRI data sets from a cohort of preterm infants aged from 26 weeks postmenstrual age (PMA) through term equivalent age at PMA-specific time points. Utilizing seed-based correlation analysis, we identified resting state networks involving varied cortical regions, the thalamus, and cerebellum. Identified networks demonstrated a regionally variable age-specific pattern of development, with more mature forms consisting of localized interhemispheric connections between homotopic counterparts. Anatomical distance was found to play a critical role in the rate of connection development. Prominent differences were noted between networks identified in term control versus premature infants at term equivalent, including in the thalamocortical connections critical for neurodevelopment. Putative precursors of the default mode network were detected in term control infants but were not identified in preterm infants, including those at term equivalent. Identified patterns of network maturation reflect the intricate relationship of structural and functional processes present throughout this important developmental period and are consistent with prior investigations of neurodevelopment in this population.


Subject(s)
Brain/growth & development , Infant, Newborn/growth & development , Nerve Net/growth & development , Neurogenesis , Female , Humans , Image Interpretation, Computer-Assisted , Magnetic Resonance Imaging , Male , Pregnancy , Premature Birth
8.
Proc Natl Acad Sci U S A ; 104(25): 10607-12, 2007 Jun 19.
Article in English | MEDLINE | ID: mdl-17556544

ABSTRACT

The human EGF receptor (HER) 2 receptor tyrosine kinase is a survival factor for human cardiomyocytes, and its inhibition may explain the increased incidence of cardiomyopathy associated with the anti-HER2 monoclonal antibody trastuzumab (Genentech, South San Francisco, CA), particularly in patients with prior exposure to cardiotoxic chemotherapies e.g., anthracyclines. Here, we show that GW2974 (HER2/EGF receptor tyrosine kinase inhibitor), but not trastuzumab, activates AMP-activated protein kinase (AMPK), initiating a metabolic stress response in human cardiomyocytes that protects against TNFalpha-induced cell death. GW2974 stimulates calcium dependent fatty acid oxidation in vitro and in the myocardium of GW2974-treated rodents. Calcium chelation or siRNA-targeted AMPK knockdown blocks GW2974 induced fatty acid oxidation. In addition, inhibition of AMPK by a specific inhibitor resulted in increased killing of cardiomyocytes. Elucidating the effects of HER2-targeted therapies on AMPK may predict for risk of cardiomyopathy and provide a novel HER2-targeted strategy designed to protect myocardium from the pro-apoptotic effects of pro-inflammatory cytokines released in response to cardiac injury by chemotherapy or acute ischemia.


Subject(s)
ErbB Receptors/antagonists & inhibitors , Multienzyme Complexes/metabolism , Myocytes, Cardiac/metabolism , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/antagonists & inhibitors , Receptor, ErbB-2/metabolism , AMP-Activated Protein Kinases , Adenosine Triphosphate/analysis , Adenosine Triphosphate/biosynthesis , Animals , Apoptosis/drug effects , Calcium/metabolism , Cells, Cultured , Enzyme Activation/drug effects , Fatty Acids/metabolism , Female , Gene Expression , Humans , Models, Biological , Myocardium/metabolism , Oxidation-Reduction , Phosphorylation/drug effects , Quinazolines/pharmacology , Rats , Rats, Sprague-Dawley
9.
Cancer Res ; 66(19): 9356-61, 2006 Oct 01.
Article in English | MEDLINE | ID: mdl-17018587

ABSTRACT

Inactivation of p53 function, which frequently occurs in tumors, can significantly modulate tumor cell sensitivity to radiation and chemotherapeutic drugs. However, in addition to acting on malignant cells, anticancer agents act on the cells of tumor stroma, causing activation of a p53 response. The effect of this response on treatment outcome has been the subject of the present study. Tumors with p53-deficient stroma were generated using mouse tumorigenic packaging cells that produce a p53 inhibitory retrovirus, encoding a dominant-negative p53 mutant. Tumors maintaining wild-type p53 in their stroma were formed by cells of similar origin but deficient in retroviral production due to the deletion of the packaging signal in the retroviral vector. Comparison of these tumor models, differing only in p53 status of their stromas, showed that tumors with p53-deficient stroma were significantly more sensitive to experimental chemotherapy and radiotherapy. A similar effect was achieved when anticancer treatment was combined with pharmacologic suppression of p53 by the cyclic form of pifithrin alpha, a small-molecule inhibitor of p53. Potentiation of the anticancer effect of chemotherapy and radiotherapy by p53 suppression in the tumor stroma is likely to be due to the increased sensitivity of p53-deficient endothelium to genotoxic stress as shown both in cell culture and in experimental tumors. Thus, reversible pharmacologic suppression of p53 may be a viable approach to improving anticancer treatment via an enhanced antiangiogenic effect of chemotherapy and radiotherapy.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Antineoplastic Agents, Alkylating/therapeutic use , Benzothiazoles/therapeutic use , Carcinoma, Lewis Lung/therapy , Cyclophosphamide/therapeutic use , Drug Resistance, Neoplasm/drug effects , Imidazoles/therapeutic use , Radiation-Sensitizing Agents/therapeutic use , Thiazoles/therapeutic use , Toluene/analogs & derivatives , Tumor Suppressor Protein p53/antagonists & inhibitors , Angiogenesis Inhibitors/pharmacology , Animals , Antineoplastic Agents, Alkylating/pharmacology , Benzothiazoles/pharmacology , Benzothiazoles/toxicity , Carcinoma, Lewis Lung/drug therapy , Carcinoma, Lewis Lung/radiotherapy , Cell Line, Transformed , Cyclophosphamide/pharmacology , Drug Screening Assays, Antitumor , Endothelial Cells/radiation effects , Fibroblasts , Imidazoles/pharmacology , Imidazoles/toxicity , Mice , Mice, Inbred C57BL , NIH 3T3 Cells , Protein Conformation , Radiation-Sensitizing Agents/pharmacology , Radiation-Sensitizing Agents/toxicity , Rats , Stromal Cells/drug effects , Stromal Cells/radiation effects , Thiazoles/pharmacology , Thiazoles/toxicity , Toluene/pharmacology , Toluene/therapeutic use , Toluene/toxicity , Transduction, Genetic , Tumor Suppressor Protein p53/chemistry , Tumor Suppressor Protein p53/genetics
10.
Proc Natl Acad Sci U S A ; 102(48): 17448-53, 2005 Nov 29.
Article in English | MEDLINE | ID: mdl-16287968

ABSTRACT

Renal cell carcinomas (RCC) commonly retain wild-type but functionally inactive p53, which is repressed by an unknown dominant mechanism. To help reveal this mechanism, we screened a diverse chemical library for small molecules capable of restoring p53-dependent transactivation in RCC cells carrying a p53-responsive reporter. Among the compounds isolated were derivatives of 9-aminoacridine (9AA), including the antimalaria drug quinacrine, which strongly induced p53 function in RCC and other types of cancer cells. Induction of p53 by these compounds does not involve genotoxic stress and is mediated by suppression of NF-kappaB activity. In contrast to agents that target IkappaB kinase 2, 9AA and quinacrine can effectively suppress both basal and inducible activities of NF-kappaB, representing inhibitors of a previously undescribed type that convert NF-kappaB from a transactivator into a transrepressor, leading to accumulation of inactive nuclear complexes with unphosphorylated Ser-536 in the p65/RelA subunit. p53 function in RCC can be restored by ectopic expression of a superrepressor of IkappaB as effectively as by 9AA-derived compounds. These findings suggest that the complete or partial repression of p53 observed in many tumors can be the result of constitutive activation of NF-kappaB. The results demonstrate, in principle, the possibility to kill cancer cells selectively through simultaneous inhibition of NF-kappaB and activation of p53 by a single small molecule and suggest anticancer applications for the well known antimalaria drug quinacrine.


Subject(s)
Aminacrine/pharmacology , Carcinoma, Renal Cell/metabolism , Gene Expression Regulation, Neoplastic/genetics , NF-kappa B/antagonists & inhibitors , Quinacrine/pharmacology , Tumor Suppressor Protein p53/metabolism , Cell Line, Tumor , Colorimetry , Humans , Structure-Activity Relationship , beta-Galactosidase/metabolism
11.
J Virol ; 79(17): 11105-14, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16103161

ABSTRACT

p53 has been well characterized as a tumor suppressor gene, but its role in antiviral defense remains unclear. A recent report has demonstrated that p53 can be induced by interferons and is activated after vesicular stomatitis virus (VSV) infection. We observed that different nononcogenic viruses, including encephalomyocarditis virus (EMCV) and human parainfluenza virus type 3 (HPIV3), induced down-regulation of p53 in infected cells. Double-stranded RNA (dsRNA) and a mutant vaccinia virus lacking the dsRNA binding protein E3L can also induce this effect, indicating that dsRNA formed during viral infection is likely the trigger for down-regulation of p53. The mechanism of down-regulation of p53 by dsRNA relies on translation inhibition mediated by the PKR and RNase L pathways. In the absence of p53, the replication of both EMCV and HPIV3 was retarded, whereas, conversely, VSV replication was enhanced. Cell cycle analysis indicated that wild-type (WT) but not p53 knockout (KO) fibroblasts undergo an early-G(1) arrest following dsRNA treatment. Moreover, in WT cells the onset of dsRNA-induced apoptosis begins after p53 levels are down-regulated, whereas p53 KO cells, which lack the early-G(1) arrest, rapidly undergo apoptosis. Hence, our data suggest that the down-regulation of p53 facilitates apoptosis, thereby limiting viral replication.


Subject(s)
Encephalomyocarditis virus/physiology , Parainfluenza Virus 3, Human/physiology , Tumor Suppressor Protein p53/metabolism , Animals , Cells, Cultured , Down-Regulation , Endoribonucleases/metabolism , Vesicular stomatitis Indiana virus/physiology , Virus Replication , eIF-2 Kinase/metabolism
12.
Cancer Res ; 64(6): 1951-8, 2004 Mar 15.
Article in English | MEDLINE | ID: mdl-15026329

ABSTRACT

Renal cell carcinoma (RCC) rarely acquires mutations in p53 tumor suppressor gene, suggesting that p53 signaling in this tumor type might be repressed by some other mechanism. In fact, all four RCC-derived cell lines we tested maintained wild-type p53 but were not capable of transactivating p53-responsive reporters and endogenous p53-responsive genes. p53 protein in RCC showed normal response to genotoxic stress, including accumulation, nuclear translocation, and activation of specific DNA binding. Functional and expression analysis of Mdm2, MdmX, and Arf showed lack of involvement of these p53 regulators in the observed defect of p53 function in RCC. However, activation of p53-mediated transactivation could be achieved by extremely high levels of p53 attained by lentivirus vector-driven transduction, suggesting the involvement of a dominant inhibitor in repression of p53-dependent transactivation in RCC. Consistently, p53 inactivation prevailed in the hybrids of RCC cells with the cells possessing fully functional p53. Remarkably, cells of normal kidney epithelium also caused partial p53 repression in cell fusion experiments, suggesting that RCC-specific p53 repression may be based on an unknown dominant mechanism also acting in normal kidney tissue.


Subject(s)
Carcinoma, Renal Cell/metabolism , Genes, Dominant , Kidney Neoplasms/metabolism , Nuclear Proteins , Signal Transduction , Tumor Suppressor Protein p53/metabolism , Carcinoma, Renal Cell/genetics , Cell Fusion , Cell Nucleus/metabolism , DNA Damage , Electrophoretic Mobility Shift Assay , Gene Expression Regulation, Neoplastic , Humans , Hybrid Cells , Kidney/metabolism , Kidney Neoplasms/genetics , Lentivirus/genetics , Luciferases , Promoter Regions, Genetic , Protein Transport , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-mdm2 , Retroviridae/genetics , Transcriptional Activation , Tumor Suppressor Protein p14ARF/metabolism , Tumor Suppressor Protein p53/deficiency , Tumor Suppressor Protein p53/genetics
13.
Diabetes ; 53(3): 821-9, 2004 Mar.
Article in English | MEDLINE | ID: mdl-14988269

ABSTRACT

The aim of the Finland-United States Investigation of NIDDM Genetics (FUSION) study is to identify genes that predispose to type 2 diabetes or are responsible for variability in diabetes-related traits via a positional cloning and positional candidate gene approach. In a previously published genome-wide scan of 478 Finnish affected sibling pair (ASP) families (FUSION 1), the strongest linkage results were on chromosomes 20 and 11. We now report a second genome-wide scan using an independent set of 242 Finnish ASP families (FUSION 2), a detailed analysis of the combined set of 737 FUSION 1 + 2 families (495 updated FUSION 1 families), and fine mapping of the regions of chromosomes 11 and 20. The strongest FUSION 2 linkage results were on chromosomes 6 (maximum logarithm of odds score [MLS] = 2.30 at 95 cM) and 14 (MLS = 1.80 at 57 cM). For the combined FUSION 1 + 2 families, three results were particularly notable: chromosome 11 (MLS = 2.98 at 82 cM), chromosome 14 (MLS = 2.74 at 58 cM), and chromosome 6 (MLS = 2.66 at 96 cM). We obtained smaller FUSION 1 + 2 MLSs on chromosomes X (MLS = 1.27 at 152 cM) and 20p (MLS = 1.21 at 20 cM). Among the 10 regions that showed nominally significant evidence for linkage in FUSION 1, four (on chromosomes 6, 11, 14, and X) also showed evidence for linkage in FUSION 2 and stronger evidence for linkage in the combined FUSION 1 + 2 sample.


Subject(s)
Chromosomes, Human, Pair 11/genetics , Chromosomes, Human, Pair 14/genetics , Chromosomes, Human, Pair 6/genetics , Diabetes Mellitus, Type 2/genetics , Genetic Predisposition to Disease/genetics , Age of Onset , Aged , Base Sequence , Body Constitution , DNA Primers , Family , Female , Finland , Genetic Markers , Genome, Human , Humans , Male , Middle Aged , Siblings
SELECTION OF CITATIONS
SEARCH DETAIL
...