Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Hugo F. Fernandez is active.

Publication


Featured researches published by Hugo F. Fernandez.


Cancer Cell | 2010

Leukemic IDH1 and IDH2 Mutations Result in a Hypermethylation Phenotype, Disrupt TET2 Function, and Impair Hematopoietic Differentiation

Maria E. Figueroa; Omar Abdel-Wahab; Chao Lu; Patrick S. Ward; Jay Patel; Alan Shih; Yushan Li; Neha Bhagwat; Aparna Vasanthakumar; Hugo F. Fernandez; Martin S. Tallman; Zhuoxin Sun; Kristy L. Wolniak; Justine K. Peeters; Wei Liu; Sung E. Choe; Valeria Fantin; Elisabeth Paietta; Bob Löwenberg; Jonathan D. Licht; Lucy A. Godley; Ruud Delwel; Peter J. M. Valk; Craig B. Thompson; Ross L. Levine; Ari Melnick

Cancer-associated IDH mutations are characterized by neomorphic enzyme activity and resultant 2-hydroxyglutarate (2HG) production. Mutational and epigenetic profiling of a large acute myeloid leukemia (AML) patient cohort revealed that IDH1/2-mutant AMLs display global DNA hypermethylation and a specific hypermethylation signature. Furthermore, expression of 2HG-producing IDH alleles in cells induced global DNA hypermethylation. In the AML cohort, IDH1/2 mutations were mutually exclusive with mutations in the α-ketoglutarate-dependent enzyme TET2, and TET2 loss-of-function mutations were associated with similar epigenetic defects as IDH1/2 mutants. Consistent with these genetic and epigenetic data, expression of IDH mutants impaired TET2 catalytic function in cells. Finally, either expression of mutant IDH1/2 or Tet2 depletion impaired hematopoietic differentiation and increased stem/progenitor cell marker expression, suggesting a shared proleukemogenic effect.


The New England Journal of Medicine | 2012

Prognostic relevance of integrated genetic profiling in acute myeloid leukemia

Jay Patel; Mithat Gonen; Maria E. Figueroa; Hugo F. Fernandez; Zhuoxin Sun; Janis Racevskis; Pieter Van Vlierberghe; Igor Dolgalev; Sabrena Thomas; Olga Aminova; Kety Huberman; Janice Cheng; Agnes Viale; Nicholas D. Socci; Adriana Heguy; Athena M. Cherry; Gail H. Vance; Rodney R. Higgins; Rhett P. Ketterling; Robert E. Gallagher; Mark R. Litzow; Marcel R.M. van den Brink; Hillard M. Lazarus; Jacob M. Rowe; Selina M. Luger; Adolfo A. Ferrando; Elisabeth Paietta; Martin S. Tallman; Ari Melnick; Omar Abdel-Wahab

BACKGROUND Acute myeloid leukemia (AML) is a heterogeneous disease with respect to presentation and clinical outcome. The prognostic value of recently identified somatic mutations has not been systematically evaluated in a phase 3 trial of treatment for AML. METHODS We performed a mutational analysis of 18 genes in 398 patients younger than 60 years of age who had AML and who were randomly assigned to receive induction therapy with high-dose or standard-dose daunorubicin. We validated our prognostic findings in an independent set of 104 patients. RESULTS We identified at least one somatic alteration in 97.3% of the patients. We found that internal tandem duplication in FLT3 (FLT3-ITD), partial tandem duplication in MLL (MLL-PTD), and mutations in ASXL1 and PHF6 were associated with reduced overall survival (P=0.001 for FLT3-ITD, P=0.009 for MLL-PTD, P=0.05 for ASXL1, and P=0.006 for PHF6); CEBPA and IDH2 mutations were associated with improved overall survival (P=0.05 for CEBPA and P=0.01 for IDH2). The favorable effect of NPM1 mutations was restricted to patients with co-occurring NPM1 and IDH1 or IDH2 mutations. We identified genetic predictors of outcome that improved risk stratification among patients with AML, independently of age, white-cell count, induction dose, and post-remission therapy, and validated the significance of these predictors in an independent cohort. High-dose daunorubicin, as compared with standard-dose daunorubicin, improved the rate of survival among patients with DNMT3A or NPM1 mutations or MLL translocations (P=0.001) but not among patients with wild-type DNMT3A, NPM1, and MLL (P=0.67). CONCLUSIONS We found that DNMT3A and NPM1 mutations and MLL translocations predicted an improved outcome with high-dose induction chemotherapy in patients with AML. These findings suggest that mutational profiling could potentially be used for risk stratification and to inform prognostic and therapeutic decisions regarding patients with AML. (Funded by the National Cancer Institute and others.).


The New England Journal of Medicine | 2009

Anthracycline dose intensification in acute myeloid leukemia

Hugo F. Fernandez; Zhuoxin Sun; Xiaopan Yao; Mark R. Litzow; Selina M. Luger; Elisabeth Paietta; Janis Racevskis; Gordon W. Dewald; Rhett P. Ketterling; John M. Bennett; Jacob M. Rowe; Hillard M. Lazarus; Martin S. Tallman

BACKGROUND In young adults with acute myeloid leukemia (AML), intensification of the anthracycline dose during induction therapy has improved the rate of complete remission but not of overall survival. We evaluated the use of cytarabine plus either standard-dose or high-dose daunorubicin as induction therapy, followed by intensive consolidation therapy, in inducing complete remission to improve overall survival. METHODS In this phase 3 randomized trial, we assigned 657 patients between the ages of 17 and 60 years who had untreated AML to receive three once-daily doses of daunorubicin at either the standard dose (45 mg per square meter of body-surface area) or a high dose (90 mg per square meter), combined with seven daily doses of cytarabine (100 mg per square meter) by continuous intravenous infusion. Patients who had a complete remission were offered either allogeneic hematopoietic stem-cell transplantation or high-dose cytarabine, with or without a single dose of the monoclonal antibody gemtuzumab ozogamicin, followed by autologous stem-cell transplantation. The primary end point was overall survival. RESULTS In the intention-to-treat analysis, high-dose daunorubicin, as compared with a standard dose of the drug, resulted in a higher rate of complete remission (70.6% vs. 57.3%, P<0.001) and improved overall survival (median, 23.7 vs. 15.7 months; P=0.003). The rates of serious adverse events were similar in the two groups. Median follow-up was 25.2 months. CONCLUSIONS In young adults with AML, intensifying induction therapy with a high daily dose of daunorubicin improved the rate of complete remission and the duration of overall survival, as compared with the standard dose. (ClinicalTrials.gov number, NCT00049517.)


Blood | 2009

MDS and secondary AML display unique patterns and abundance of aberrant DNA methylation

Maria E. Figueroa; Lucy Skrabanek; Yushan Li; Anchalee Jiemjit; Tamer E. Fandy; Elisabeth Paietta; Hugo F. Fernandez; Martin S. Tallman; John M. Greally; Hetty E. Carraway; Jonathan D. Licht; Steven D. Gore; Ari Melnick

Increasing evidence shows aberrant hypermethylation of genes occurring in and potentially contributing to pathogenesis of myeloid malignancies. Several of these diseases, such as myelodysplastic syndromes (MDSs), are responsive to DNA methyltransferase inhibitors. To determine the extent of promoter hypermethylation in such tumors, we compared the distribution of DNA methylation of 14 000 promoters in MDS and secondary acute myeloid leukemia (AML) patients enrolled in a phase 1 trial of 5-azacytidine and the histone deacetylase inhibitor entinostat against de novo AML patients and normal CD34(+) bone marrow cells. The MDS and secondary AML patients displayed more extensive aberrant DNA methylation involving thousands of genes than did the normal CD34(+) bone marrow cells or de novo AML blasts. Aberrant methylation in MDS and secondary AML tended to affect particular chromosomal regions, occurred more frequently in Alu-poor genes, and included prominent involvement of genes involved in the WNT and MAPK signaling pathways. DNA methylation was also measured at days 15 and 29 after the first treatment cycle. DNA methylation was reversed at day 15 in a uniform manner throughout the genome, and this effect persisted through day 29, even without continuous administration of the study drugs. This trial was registered at www.clinicaltrials.gov as J0443.


PLOS Genetics | 2012

Base-pair resolution DNA methylation sequencing reveals profoundly divergent epigenetic landscapes in acute myeloid leukemia

Altuna Akalin; Francine E. Garrett-Bakelman; Matthias Kormaksson; Jennifer Busuttil; Lu Zhang; Irina Khrebtukova; Thomas A. Milne; Yongsheng Huang; Debabrata Biswas; Jay L. Hess; C. David Allis; Robert G. Roeder; Bob Löwenberg; Ruud Delwel; Hugo F. Fernandez; Elisabeth Paietta; Martin S. Tallman; Gary P. Schroth; Christopher E. Mason; Ari Melnick; Maria E. Figueroa

We have developed an enhanced form of reduced representation bisulfite sequencing with extended genomic coverage, which resulted in greater capture of DNA methylation information of regions lying outside of traditional CpG islands. Applying this method to primary human bone marrow specimens from patients with Acute Myelogeneous Leukemia (AML), we demonstrated that genetically distinct AML subtypes display diametrically opposed DNA methylation patterns. As compared to normal controls, we observed widespread hypermethylation in IDH mutant AMLs, preferentially targeting promoter regions and CpG islands neighboring the transcription start sites of genes. In contrast, AMLs harboring translocations affecting the MLL gene displayed extensive loss of methylation of an almost mutually exclusive set of CpGs, which instead affected introns and distal intergenic CpG islands and shores. When analyzed in conjunction with gene expression profiles, it became apparent that these specific patterns of DNA methylation result in differing roles in gene expression regulation. However, despite this subtype-specific DNA methylation patterning, a much smaller set of CpG sites are consistently affected in both AML subtypes. Most CpG sites in this common core of aberrantly methylated CpGs were hypermethylated in both AML subtypes. Therefore, aberrant DNA methylation patterns in AML do not occur in a stereotypical manner but rather are highly specific and associated with specific driving genetic lesions.


Transplantation | 1997

High-dose donor bone marrow infusions to enhance allograft survival: The effect of timing

Camillo Ricordi; T. Karatzas; Jose Nery; M. Webb; Gennaro Selvaggi; Luis A. Fernandez; Farrukh A. Khan; Phillip Ruiz; Eugene R. Schiff; L. Olson; Hugo F. Fernandez; Judy A. Bean; Violet Esquenazi; Joshua Miller; Andreas G. Tzakis

BACKGROUND The development of strategies to enhance survival of transplanted organs and to potentially lower or even discontinue immunosuppressive therapy would represent a significant advance in posttransplant patient care. The aim of this clinical trial was to determine the effect of timing and dose of peripheral donor bone marrow cell (DBMC) infusion on graft and patient survival after liver transplantation. METHODS DBMC, obtained from vertebral bodies, were administered in 101 recipients of liver allografts (OLTX). There were 107 patients for whom DBMC could not be obtained; they received OLTX alone (controls). A total of 5 x 10(8)/kg DBMC were infused at day 0 (group 1; n=9); at days 0 and 11 (group 2; n=26); or at days 5 and 11 (group 3; n=26). In group 4 (n=40), patients received up to five infusions of 2 x 10(8)/kg DBMC at days 5, 14, 21, 28, and 90 after OLTX. RESULTS When the results from patients receiving two or more DBMC infusions (groups 2, 3, and 4) are considered, both patient and graft survival were significantly improved compared with the control group (P=0.02 and P=0.01, respectively). In groups 3 and 4, 88.5% and 95% of patients were alive with mean follow-up of 536 and 265 days, respectively, compared with 77.6% of patients in the control group (average follow-up of 452 days) (P=0.02). Graft survival was also significantly improved in groups 3 (88.5%) and 4 (92.5%), compared with the controls (72%) (P=0.007). CONCLUSIONS The results suggest that dose and timing of DBMC infusions may be important variables affecting allograft survival. A randomized prospective trial is now in progress to compare group 3 DBMC infusion protocol with controls receiving OLTX alone.


Bone Marrow Transplantation | 2010

5-Azacitidine for myelodysplasia before allogeneic hematopoietic cell transplantation

Teresa Field; Janelle Perkins; Y Huang; Mohamed A. Kharfan-Dabaja; Melissa Alsina; Ernesto Ayala; Hugo F. Fernandez; William Janssen; Jeffrey E. Lancet; Lia Perez; Daniel M. Sullivan; Alan F. List; Claudio Anasetti

Relapse remains a leading cause for treatment failure after hematopoietic cell transplantation (HCT) in patients with intermediate- or high-risk myelodysplastic syndrome (MDS). To discern the impact of 5-azacitine treatment pretransplant on the risk for relapse after HCT, we analyzed the post transplant outcomes of all 54 consecutive patients with MDS or chronic myelomonocytic leukemia who received HCT from HLA-compatible donors according to pretransplant 5-azacitidine exposure. Thirty patients received a median of four (1–7) cycles of 5-azacitidine, and 24 patients did not receive 5-azacitidine before HCT. The 1-year estimates of overall survival, relapse-free survival and cumulative incidence of relapse were 47, 41 and 20%, for 5-azacitidine patients and 60, 51 and 32%, respectively, for non-5-azacytidine patients. These observations suggest that outcomes are similar in both groups with a trend toward decreased early relapse in patients receiving 5-azacitidine. 5-Azacitidine may be of value in stabilizing the disease, thereby allowing time for patients to reach transplant and does not appear to affect transplant outcomes.


Blood | 2013

Serum 2-hydroxyglutarate levels predict isocitrate dehydrogenase mutations and clinical outcome in acute myeloid leukemia

Courtney D. DiNardo; Kathleen J. Propert; Alison W. Loren; Elisabeth Paietta; Zhuoxin Sun; Ross L. Levine; Kimberly Straley; Katharine E. Yen; Jay Patel; Samuel V. Agresta; Omar Abdel-Wahab; Alexander E. Perl; Mark R. Litzow; Jacob M. Rowe; Hillard M. Lazarus; Hugo F. Fernandez; David J. Margolis; Martin S. Tallman; Selina M. Luger; Martin Carroll

Cancer-associated isocitrate dehydrogenase (IDH) mutations produce the metabolite 2-hydroxyglutarate (2HG), but the clinical utility of 2HG has not been established. We studied whether 2HG measurements in acute myeloid leukemia (AML) patients correlate with IDH mutations, and whether diagnostic or remission 2HG measurements predict survival. Sera from 223 de novo AML patients were analyzed for 2HG concentration by reverse-phase liquid chromatography-mass spectrometry. Pretreatment 2HG levels ranged from 10 to 30 000 ng/mL and were elevated in IDH-mutants (median, 3004 ng/mL), compared to wild-type IDH (median, 61 ng/mL) (P < .0005). 2HG levels did not differ among IDH1 or IDH2 allelic variants. In receiver operating characteristic analysis, a discriminatory level of 700 ng/mL optimally segregated patients with and without IDH mutations, and on subsequent mutational analysis of the 13 IDH wild-type samples with 2HG levels >700 ng/mL, 9 were identified to have IDH mutations. IDH-mutant patients with 2HG levels >200 at complete remission had shorter overall survival compared to 2HG ≤200 ng/mL (hazard ratio, 3.9; P = .02). We establish a firm association between IDH mutations and serum 2HG concentration in AML, and confirm that serum oncometabolite measurements provide useful diagnostic and prognostic information that can improve patient selection for IDH-targeted therapies.


Leukemia | 2011

PHF6 mutations in adult acute myeloid leukemia

P Van Vlierberghe; Jawaharlal M. Patel; Omar Abdel-Wahab; Camille Lobry; C Hedvat; Milagros Balbín; Concha Nicolas; A R Payer; Hugo F. Fernandez; Martin S. Tallman; Elisabeth Paietta; Ari Melnick; Peter Vandenberghe; Franki Speleman; Iannis Aifantis; Jan Cools; Ross L. Levine; Adolfo A. Ferrando

Loss of function mutations and deletions encompassing the plant homeodomain finger 6 (PHF6) gene are present in about 20% of T-cell acute lymphoblastic leukemias (ALLs). Here, we report the identification of recurrent mutations in PHF6 in 10/353 adult acute myeloid leukemias (AMLs). Genetic lesions in PHF6 found in AMLs are frameshift and nonsense mutations distributed through the gene or point mutations involving the second plant homeodomain (PHD)-like domain of the protein. As in the case of T-ALL, where PHF6 alterations are found almost exclusively in males, mutations in PHF6 were seven times more prevalent in males than in females with AML. Overall, these results identify PHF6 as a tumor suppressor gene mutated in AML and extend the role of this X-linked tumor suppressor gene in the pathogenesis of hematologic tumors.


Blood | 2012

Overexpression of IL-1 receptor accessory protein in stem and progenitor cells and outcome correlation in AML and MDS

Laura Barreyro; Britta Will; Boris Bartholdy; Li Zhou; Tihomira I. Todorova; Robert F. Stanley; Susana Ben-Neriah; Cristina Montagna; Samir Parekh; Andrea Pellagatti; Jacqueline Boultwood; Elisabeth Paietta; Rhett P. Ketterling; Larry D. Cripe; Hugo F. Fernandez; Peter L. Greenberg; Martin S. Tallman; Christian Steidl; Constantine S. Mitsiades; Amit Verma; Ulrich Steidl

Cellular and interpatient heterogeneity and the involvement of different stem and progenitor compartments in leukemogenesis are challenges for the identification of common pathways contributing to the initiation and maintenance of acute myeloid leukemia (AML). Here we used a strategy of parallel transcriptional analysis of phenotypic long-term hematopoietic stem cells (HSCs), short-term HSCs, and granulocyte-monocyte progenitors from individuals with high-risk (-7/7q-) AML and compared them with the corresponding cell populations from healthy controls. This analysis revealed dysregulated expression of 11 genes, including IL-1 receptor accessory protein (IL1RAP), in all leukemic stem and progenitor cell compartments. IL1RAP protein was found to be overexpressed on the surface of HSCs of AML patients, and marked cells with the -7/7q- anomaly. IL1RAP was also overexpressed on HSCs of patients with normal karyotype AML and high-risk myelodysplastic syndrome, suggesting a pervasive role in different disease subtypes. High IL1RAP expression was independently associated with poor overall survival in 3 independent cohorts of AML patients (P = 2.2 × 10(-7)). Knockdown of IL1RAP decreased clonogenicity and increased cell death of AML cells. Our study identified genes dysregulated in stem and progenitor cells in -7/7q- AML, and suggests that IL1RAP may be a promising therapeutic and prognostic target in AML and high-risk myelodysplastic syndrome.

Collaboration


Dive into the Hugo F. Fernandez's collaboration.

Top Co-Authors

Avatar

Claudio Anasetti

University of South Florida

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Teresa Field

University of South Florida

View shared research outputs
Top Co-Authors

Avatar

Lia Perez

University of South Florida

View shared research outputs
Top Co-Authors

Avatar

Ernesto Ayala

University of South Florida

View shared research outputs
Top Co-Authors

Avatar

Janelle Perkins

University of South Florida

View shared research outputs
Top Co-Authors

Avatar

Joseph Pidala

University of South Florida

View shared research outputs
Top Co-Authors

Avatar

Melissa Alsina

University of South Florida

View shared research outputs
Top Co-Authors

Avatar

Taiga Nishihori

University of South Florida

View shared research outputs
Top Co-Authors

Avatar

Jongphil Kim

University of South Florida

View shared research outputs
Researchain Logo
Decentralizing Knowledge