Network


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

Hotspot


Dive into the research topics where Kati Maharry is active.

Publication


Featured researches published by Kati Maharry.


Journal of Clinical Oncology | 2008

Prognostic Significance of, and Gene and MicroRNA Expression Signatures Associated With, CEBPA Mutations in Cytogenetically Normal Acute Myeloid Leukemia With High-Risk Molecular Features: A Cancer and Leukemia Group B Study

Guido Marcucci; Kati Maharry; Michael D. Radmacher; Krzysztof Mrózek; Tamara Vukosavljevic; Peter Paschka; Susan P. Whitman; Christian Langer; Claudia D. Baldus; Chang Gong Liu; Amy S. Ruppert; Bayard L. Powell; Andrew J. Carroll; Michael A. Caligiuri; Jonathan E. Kolitz; Richard A. Larson; Clara D. Bloomfield

PURPOSE To evaluate the prognostic significance of CEBPA mutations in the context of established molecular markers in cytogenetically normal (CN) acute myeloid leukemia (AML) and gain biologic insights into leukemogenesis of the CN-AML molecular high-risk subset (FLT3 internal tandem duplication [ITD] positive and/or NPM1 wild type) that has a significantly higher incidence of CEBPA mutations than the molecular low-risk subset (FLT3-ITD negative and NPM1 mutated). PATIENTS AND METHODS One hundred seventy-five adults age less than 60 years with untreated primary CN-AML were screened before treatment for CEBPA, FLT3, MLL, WT1, and NPM1 mutations and BAALC and ERG expression levels. Gene and microRNA (miRNA) expression profiles were obtained for the CN-AML molecular high-risk patients. RESULTS CEBPA mutations predicted better event-free (P = .007), disease-free (P = .014), and overall survival (P < .001) independently of other molecular and clinical prognosticators. Among patients with CEBPA mutations, 91% were in the CN-AML molecular high-risk group. Within this group, CEBPA mutations predicted better event-free (P < .001), disease-free (P = .004), and overall survival (P = .009) independently of other molecular and clinical characteristics and were associated with unique gene and miRNA expression profiles. The major features of these profiles were upregulation of genes (eg, GATA1, ZFPM1, EPOR, and GFI1B) and miRNAs (ie, the miR-181 family) involved in erythroid differentiation and downregulation of homeobox genes. CONCLUSION Pretreatment testing for CEBPA mutations identifies CN-AML patients with different outcomes, particularly in the molecular high-risk group, thus improving molecular risk-based classification of this large cytogenetic subset of AML. The gene and miRNA expression profiling provided insights into leukemogenesis of the CN-AML molecular high-risk group, indicating that CEBPA mutations are associated with partial erythroid differentiation.


Blood | 2008

An 86-probe-set gene-expression signature predicts survival in cytogenetically normal acute myeloid leukemia.

Klaus H. Metzeler; Manuela Hummel; Clara D. Bloomfield; Karsten Spiekermann; Jan Braess; M. C. Sauerland; Achim Heinecke; Michael D. Radmacher; Guido Marcucci; Susan P. Whitman; Kati Maharry; Peter Paschka; Richard A. Larson; Wolfgang E. Berdel; Thomas Büchner; Bernhard Wörmann; Ulrich Mansmann; Wolfgang Hiddemann; Stefan K. Bohlander; Christian Buske

Patients with cytogenetically normal acute myeloid leukemia (CN-AML) show heterogeneous treatment outcomes. We used gene-expression profiling to develop a gene signature that predicts overall survival (OS) in CN-AML. Based on data from 163 patients treated in the German AMLCG 1999 trial and analyzed on oligonucleotide microarrays, we used supervised principal component analysis to identify 86 probe sets (representing 66 different genes), which correlated with OS, and defined a prognostic score based on this signature. When applied to an independent cohort of 79 CN-AML patients, this continuous score remained a significant predictor for OS (hazard ratio [HR], 1.85; P = .002), event-free survival (HR = 1.73; P = .001), and relapse-free survival (HR = 1.76; P = .025). It kept its prognostic value in multivariate analyses adjusting for age, FLT3 ITD, and NPM1 status. In a validation cohort of 64 CN-AML patients treated on CALGB study 9621, the score also predicted OS (HR = 4.11; P < .001), event-free survival (HR = 2.90; P < .001), and relapse-free survival (HR = 3.14, P < .001) and retained its significance in a multivariate model for OS. In summary, we present a novel gene-expression signature that offers additional prognostic information for patients with CN-AML.


Blood | 2011

ASXL1 mutations identify a high-risk subgroup of older patients with primary cytogenetically normal AML within the ELN Favorable genetic category

Klaus H. Metzeler; Heiko Becker; Kati Maharry; Michael D. Radmacher; Jessica Kohlschmidt; Krzysztof Mrózek; Deedra Nicolet; Susan P. Whitman; Yue-Zhong Wu; Sebastian Schwind; Bayard L. Powell; Thomas H. Carter; Meir Wetzler; Joseph O. Moore; Jonathan E. Kolitz; Maria R. Baer; Andrew J. Carroll; Richard A. Larson; Michael A. Caligiuri; Guido Marcucci; Clara D. Bloomfield

The associations of mutations in the enhancer of trithorax and polycomb family gene ASXL1 with pretreatment patient characteristics, outcomes, and gene-/microRNA-expression profiles in primary cytogenetically normal acute myeloid leukemia (CN-AML) are unknown. We analyzed 423 adult patients for ASXL1 mutations, other prognostic gene mutations, and gene-/microRNA-expression profiles. ASXL1 mutations were 5 times more common in older (≥ 60 years) patients (16.2%) than those younger than 60 years (3.2%; P < .001). Among older patients, ASXL1 mutations associated with wild-type NPM1 (P < .001), absence of FLT3-internal tandem duplications (P = .002), mutated CEBPA (P = .01), and with inferior complete remission (CR) rate (P = .04), disease-free survival (DFS; P = .03), overall survival (OS; P = .006), and event-free survival (EFS; P = .002). Within the European LeukemiaNet (ELN) genetic categories of older CN-AML, ASXL1 mutations associated with inferior CR rate (P = .02), OS (P < .001), and EFS (P < .001) among ELN Favorable, but not among ELN Intermediate-I patients. Multivariable analyses confirmed associations of ASXL1 mutations with unfavorable CR rate (P = .03), DFS (P < .001), OS (P < .001), and EFS (P < .001) among ELN Favorable patients. We identified an ASXL1 mutation-associated gene-expression signature, but no microRNA-expression signature. This first study of ASXL1 mutations in primary CN-AML demonstrates that ASXL1-mutated older patients, particularly within the ELN Favorable group, have unfavorable outcomes and may be candidates for experimental treatment approaches.


Cancer Cell | 2010

Sp1/NFκB/HDAC/miR-29b Regulatory Network in KIT-Driven Myeloid Leukemia

Shujun Liu; Lai-Chu Wu; Jiuxia Pang; Ramasamy Santhanam; Sebastian Schwind; Yue Zhong Wu; Christopher Hickey; Jianhua Yu; Heiko Becker; Kati Maharry; Michael D. Radmacher; Chenglong Li; Susan P. Whitman; Anjali Mishra; Nicole Stauffer; Anna M. Eiring; Roger Briesewitz; Robert A. Baiocchi; Kenneth K. Chan; Peter Paschka; Michael A. Caligiuri; John C. Byrd; Carlo M. Croce; Clara D. Bloomfield; Danilo Perrotti; Ramiro Garzon; Guido Marcucci

The biologic and clinical significance of KIT overexpression that associates with KIT gain-of-function mutations occurring in subsets of acute myeloid leukemia (AML) (i.e., core binding factor AML) is unknown. Here, we show that KIT mutations lead to MYC-dependent miR-29b repression and increased levels of the miR-29b target Sp1 in KIT-driven leukemia. Sp1 enhances its own expression by participating in a NFkappaB/HDAC complex that further represses miR-29b transcription. Upregulated Sp1 then binds NFkappaB and transactivates KIT. Therefore, activated KIT ultimately induces its own transcription. Our results provide evidence that the mechanisms of Sp1/NFkappaB/HDAC/miR-29b-dependent KIT overexpression contribute to leukemia growth and can be successfully targeted by pharmacological disruption of the Sp1/NFkappaB/HDAC complex or synthetic miR-29b treatment in KIT-driven AML.


Blood | 2012

Up-regulation of a HOXA-PBX3 homeobox-gene signature following down-regulation of miR-181 is associated with adverse prognosis in patients with cytogenetically abnormal AML

Zejuan Li; Hao Huang; Yuanyuan Li; Xi Jiang; Ping Chen; Stephen Arnovitz; Michael D. Radmacher; Kati Maharry; Abdel G. Elkahloun; Xinan Yang; Chunjiang He; Miao He; Zhiyu Zhang; Konstanze Döhner; Mary Beth Neilly; Colles Price; Yves A. Lussier; Yanming Zhang; Richard A. Larson; Michelle M. Le Beau; Michael A. Caligiuri; Lars Bullinger; Ruud Delwel; Bob Löwenberg; Paul Liu; Guido Marcucci; Clara D. Bloomfield; Janet D. Rowley; Jianjun Chen

Increased expression levels of miR-181 family members have been shown to be associated with favorable outcome in patients with cytogenetically normal acute myeloid leukemia. Here we show that increased expression of miR-181a and miR-181b is also significantly (P < .05; Cox regression) associated with favorable overall survival in cytogenetically abnormal AML (CA-AML) patients. We further show that up-regulation of a gene signature composed of 4 potential miR-181 targets (including HOXA7, HOXA9, HOXA11, and PBX3), associated with down-regulation of miR-181 family members, is an independent predictor of adverse overall survival on multivariable testing in analysis of 183 CA-AML patients. The independent prognostic impact of this 4-homeobox-gene signature was confirmed in a validation set of 271 CA-AML patients. Furthermore, our in vitro and in vivo studies indicated that ectopic expression of miR-181b significantly promoted apoptosis and inhibited viability/proliferation of leukemic cells and delayed leukemogenesis; such effects could be reversed by forced expression of PBX3. Thus, the up-regulation of the 4 homeobox genes resulting from the down-regulation of miR-181 family members probably contribute to the poor prognosis of patients with nonfavorable CA-AML. Restoring expression of miR-181b and/or targeting the HOXA/PBX3 pathways may provide new strategies to improve survival substantially.


Blood | 2010

P-glycoprotein inhibition using valspodar (PSC-833) does not improve outcomes for patients younger than age 60 years with newly diagnosed acute myeloid leukemia: Cancer and Leukemia Group B study 19808

Jonathan E. Kolitz; Stephen L. George; Guido Marcucci; Ravi Vij; Bayard L. Powell; Steven L. Allen; Daniel J. DeAngelo; Thomas C. Shea; Wendy Stock; Maria R. Baer; Vera Hars; Kati Maharry; Eva Hoke; James W. Vardiman; Clara D. Bloomfield; Richard A. Larson

Cancer and Leukemia Group B 19808 (CALGB 19808) is the only randomized trial of a second-generation P-glycoprotein (Pgp) modulator in untreated patients with acute myeloid leukemia (AML) younger than age 60 years. We randomly assigned 302 patients to receive induction chemotherapy regimens consisting of cytosine arabinoside (Ara-C; A), daunorubicin (D), and etoposide (E), without (ADE) or with (ADEP) PSC-833 (P). The incidence of complete remission was 75% with both regimens. Reversible grade 3 and 4 liver and mucosal toxicities were significantly more common with ADEP. Therapy-related mortality was 7% and did not differ by induction arm. Excess cardiotoxicity was not seen with high doses of D in ADE. The median disease-free survival was 1.34 years in the ADE arm and 1.09 years in the ADEP arm (P = .74, log-rank test); the median overall survival was 1.86 years in the ADE arm and 1.69 years in the ADEP arm (P = .82). There was no evidence of a treatment difference within any identifiable patient subgroup. Inhibition of Pgp-mediated drug efflux by PSC-833 did not improve clinical outcomes in younger patients with untreated AML. This trial was registered at www.clinicaltrials.gov as #NCT00006363.


Journal of Clinical Oncology | 2014

Epigenetics Meets Genetics in Acute Myeloid Leukemia: Clinical Impact of a Novel Seven-Gene Score

Guido Marcucci; Pearlly S. Yan; Kati Maharry; David Frankhouser; Deedra Nicolet; Klaus H. Metzeler; Jessica Kohlschmidt; Krzysztof Mrózek; Yue Zhong Wu; Donna Bucci; John Curfman; Susan P. Whitman; Ann-Kathrin Eisfeld; Jason H. Mendler; Sebastian Schwind; Heiko Becker; Constance Bär; Andrew J. Carroll; Maria R. Baer; Meir Wetzler; Thomas H. Carter; Bayard L. Powell; Jonathan E. Kolitz; John C. Byrd; Christoph Plass; Ramiro Garzon; Michael A. Caligiuri; Richard Stone; Stefano Volinia; Ralf Bundschuh

PURPOSE Molecular risk stratification of acute myeloid leukemia (AML) is largely based on genetic markers. However, epigenetic changes, including DNA methylation, deregulate gene expression and may also have prognostic impact. We evaluated the clinical relevance of integrating DNA methylation and genetic information in AML. METHODS Next-generation sequencing analysis of methylated DNA identified differentially methylated regions (DMRs) associated with prognostic mutations in older (≥ 60 years) cytogenetically normal (CN) patients with AML (n = 134). Genes with promoter DMRs and expression levels significantly associated with outcome were used to compute a prognostic gene expression weighted summary score that was tested and validated in four independent patient sets (n = 355). RESULTS In the training set, we identified seven genes (CD34, RHOC, SCRN1, F2RL1, FAM92A1, MIR155HG, and VWA8) with promoter DMRs and expression associated with overall survival (OS; P ≤ .001). Each gene had high DMR methylation and lower expression, which were associated with better outcome. A weighted summary expression score of the seven gene expression levels was computed. A low score was associated with a higher complete remission (CR) rate and longer disease-free survival and OS (P < .001 for all end points). This was validated in multivariable models and in two younger (< 60 years) and two older independent sets of patients with CN-AML. Considering the seven genes individually, the fewer the genes with high expression, the better the outcome. Younger and older patients with no genes or one gene with high expression had the best outcomes (CR rate, 94% and 87%, respectively; 3-year OS, 80% and 42%, respectively). CONCLUSION A seven-gene score encompassing epigenetic and genetic prognostic information identifies novel AML subsets that are meaningful for treatment guidance.


Journal of the National Cancer Institute | 2014

Pluripotent Stem Cell miRNAs and Metastasis in Invasive Breast Cancer

Stefano Volinia; Gerard J. Nuovo; Alessandra Drusco; Stefan Costinean; Ramzey Abujarour; Caroline Desponts; Michela Garofalo; Raffaele Baffa; Rami Aeqilan; Kati Maharry; Maria Elena Sana Ramiro Garzon; Gianpiero Di Leva; Pierluigi Gasparini; Paola Dama; Jlenia Marchesini; Marco Galasso; Marco Manfrini; Carlotta Zerbinati; Fabio Corrà; Timothy Wise; Sylwia E. Wojcik; Maurizio Previati; Flavia Pichiorri; Nicola Zanesi; Hansjuerg Alder; Jeff Palatini; Kay Huebner; Charles L. Shapiro; Massimo Negrini; Andrea Vecchione

BACKGROUND The purpose of this study is to determine whether microRNA for pluripotent stem cells are also expressed in breast cancer and are associated with metastasis and outcome. METHODS We studied global microRNA profiles during differentiation of human embryonic stem cells (n =26) and in breast cancer patients (n = 33) and human cell lines (n = 35). Using in situ hybridization, we then investigated MIR302 expression in 318 untreated breast cancer patients (test cohort, n = 22 and validation cohort, n = 296). In parallel, using next-generation sequencing data from breast cancer patients (n = 684), we assessed microRNA association with stem cell markers. All statistical tests were two-sided. RESULTS In healthy tissues, the MIR302 (high)/MIR203 (low) asymmetry was exclusive for pluripotent stem cells. MIR302 was expressed in a small population of cancer cells within invasive ductal carcinoma, but not in normal breast (P < .001). Furthermore, MIR302 was expressed in the tumor cells together with stem cell markers, such as CD44 and BMI1. Conversely, MIR203 expression in 684 breast tumors negatively correlated with CD44 (Spearman correlation, Rho = -0.08, P = .04) and BMI1 (Rho = -0.11, P = .004), but positively correlated with differentiation marker CD24 (Rho = 0.15, P < .001). Primary tumors with lymph node metastasis had cancer cells showing scattered expression of MIR302 and widespread repression of MIR203. Finally, overall survival was statistically significantly shorter in patients with MIR302-positive cancer cells (P = .03). CONCLUSIONS In healthy tissues the MIR302(high)/MIR203(low) asymmetry was characteristic of embryonic and induced pluripotency. In invasive ductal carcinoma, the MIR302/MIR203 asymmetry was associated with stem cell markers, metastasis, and shorter survival.


Blood | 2012

miR-3151 interplays with its host gene BAALC and independently affects outcome of patients with cytogenetically normal acute myeloid leukemia

Ann-Kathrin Eisfeld; Guido Marcucci; Kati Maharry; Sebastian Schwind; Michael D. Radmacher; Deedra Nicolet; Heiko Becker; Krzysztof Mrózek; Susan P. Whitman; Klaus H. Metzeler; Jason H. Mendler; Yue-Zhong Wu; Sandya Liyanarachchi; Ravi Patel; Maria R. Baer; Bayard L. Powell; Thomas H. Carter; Joseph O. Moore; Jonathan E. Kolitz; Meir Wetzler; Michael A. Caligiuri; Richard A. Larson; Stephan M. Tanner; Albert de la Chapelle; Clara D. Bloomfield

High BAALC expression levels are associated with poor outcome in cytogenetically normal acute myeloid leukemia (CN-AML) patients. Recently, miR-3151 was discovered in intron 1 of BAALC. To evaluate the prognostic significance of miR-3151 expression levels and to gain insight into the biologic and prognostic interplay between miR-3151 and its host, miR-3151 and BAALC expression were measured in pretreatment blood of 179 CN-AML patients. Gene-expression profiling and miRNA-expression profiling were performed using microarrays. High miR-3151 expression was associated with shorter disease-free and overall survival, whereas high BAALC expression predicted failure of complete remission and shorter overall survival. Patients exhibiting high expression of both miR-3151 and BAALC had worse outcome than patients expressing low levels of either gene or both genes. In gene-expression profiling, high miR-3151 expressers showed down-regulation of genes involved in transcriptional regulation, posttranslational modification, and cancer pathways. Two genes, FBXL20 and USP40, were validated as direct miR-3151 targets. The results of the present study show that high expression of miR-3151 is an independent prognosticator for poor outcome in CN-AML and affects different outcome end points than its host gene, BAALC. The combination of both markers identified a patient subset with the poorest outcome. This interplay between an intronic miR and its host may have important biologic implications.


Blood | 2010

A variant allele of Growth Factor Independence 1 (GFI1) is associated with acute myeloid leukemia.

Cyrus Khandanpour; Christian Thiede; Peter J. M. Valk; Ehssan Sharif-Askari; Holger Nückel; Dietmar R. Lohmann; Bernhard Horsthemke; Winfried Siffert; Andreas Neubauer; Karl-Heinz Grzeschik; Clara D. Bloomfield; Guido Marcucci; Kati Maharry; Marilyn L. Slovak; Bert A. van der Reijden; Joop H. Jansen; Hans K. Schackert; Khashayar Afshar; Susanne Schnittger; Justine K. Peeters; Frank Kroschinsky; Gerhard Ehninger; Bob Löwenberg; Ulrich Dührsen; Tarik Möröy

The GFI1 gene encodes a transcriptional repressor, which regulates myeloid differentiation. In the mouse, Gfi1 deficiency causes neutropenia and an accumulation of granulomonocytic precursor cells that is reminiscent of a myelodysplastic syndrome. We report here that a variant allele of GFI1 (GFI1(36N)) is associated with acute myeloid leukemia (AML) in white subjects with an odds ratio of 1.6 (P < 8 x 10(-5)). The GFI1(36N) variant occurred in 1806 AML patients with an allele frequency of 0.055 compared with 0.035 in 1691 healthy control patients in 2 independent cohorts. We observed that both GFI1 variants maintain the same activity as transcriptional repressors but differ in their regulation by the AML1/ETO (RUNX1/RUNX1T1) fusion protein produced in AML patients with a t(8;21) translocation. AML1/ETO interacts and colocalizes with the more common GFI1(36S) form in the nucleus and inhibits its repressor activity. However, the variant GFI1(36N) protein has a different subnuclear localization than GFI1(36S). As a consequence, AML1/ETO does not colocalize with GFI1(36N) and is unable to inhibit its repressor activity. We conclude that both variants of GFI1 differ in their ability to be regulated by interacting proteins and that the GFI1(36N) variant form exhibits distinct biochemical features that may confer a predisposition to AML.

Collaboration


Dive into the Kati Maharry's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Bayard L. Powell

Wake Forest Baptist Medical Center

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge