Network


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

Hotspot


Dive into the research topics where Guido Marcucci is active.

Publication


Featured researches published by Guido Marcucci.


Leukemia | 2001

Quantification of CBFβ/MYH11 fusion transcript by Real Time RT-PCR in patients with INV(16) acute myeloid leukemia

Guido Marcucci; Michael A. Caligiuri; Hartmut Döhner; Kj Archer; Richard F. Schlenk; K Dohner; Ea Maghraby; Clara D. Bloomfield

Amplification of the CBFβ/MYH11 fusion transcript by a qualitative reverse transcription-polymerase chain reaction (RT-PCR) has been used to detect minimal residual disease (MRD) and assess the risk for disease relapse in inv(16)(p13q22) acute myeloid leukemia (AML). This strategy has, however, produced conflicting results and because of an uncertain predictive value, its use in the clinical setting cannot be recommended. The objective of the current study was to evaluate if quantification by Real Time RT-PCR could be useful to determine levels of CBFβ/MYH11 fusion transcripts predictive of clinical outcome in inv(16)(p13q22) AML at diagnosis or during remission. Bone marrow (BM) samples from 16 patients with inv(16) AML enrolled on a German multicenter trial (AML HD93) were analyzed for levels of CBFβ/MYH11 fusion transcripts by Real Time RT-PCR at diagnosis (nu2009=u200914), during remission (nu2009=u200910) and at relapse (nu2009=u20096). The CBFβ/MYH11 transcript copy number in each sample was normalized to copies of an internal control housekeeping transcript (ie 18S). The copy number measured at diagnosis or relapse were 3 to 4 log higher that those measured during remission, following completion of induction treatment. A high CBFβ/MYH11transcript copy number at diagnosis had a significant correlation with a high percentage of BM blasts (Spearmans coefficientu2009=u2009−0.66; Pu2009=u20090.03), and a borderline correlation with a short complete remission (CR) duration (Spearmans coefficientu2009=u2009−0.51; Pu2009=u20090.07). No difference in levels of CBFβ/MYH11 fusion transcripts measured during intensification therapy was found between patients destined to relapse and those who continued in CCR (Pu2009=u20090.75). Following completion of the entire chemotherapy program, patients that during CR showed a CBFβ/MYH11 fusion transcript copy number >10 had a significantly shorter CR duration (Pu2009=u20090.002) and higher risk for disease relapse (Pu2009=u20090.05) than patients with a CBFβ/MYH11fusion transcript copy number <10. The results of the current study, therefore, suggest that it is possible to determine in remission samples a threshold of CBFβ/MYH11 transcript copy number above which relapse occurs and below which continuous CR is likely.


Blood | 2013

Lenalidomide-mediated enhanced translation of C/EBPα-p30 protein up-regulates expression of the antileukemic microRNA-181a in acute myeloid leukemia

Christopher Hickey; Sebastian Schwind; Hanna S. Radomska; Adrienne M. Dorrance; Ramasamy Santhanam; Anjali Mishra; Yue-Zhong Wu; Houda Alachkar; K. Maharry; Nicolet D; Krzysztof Mrózek; Alison Walker; Anna M. Eiring; Susan P. Whitman; Heiko Becker; Danilo Perrotti; Lai-Chu Wu; Xiaobin Zhao; Todd A. Fehniger; Ravi Vij; John C. Byrd; William Blum; Ly James Lee; Michael A. Caligiuri; Clara D. Bloomfield; Ramiro Garzon; Guido Marcucci

Recently, we showed that increased miR-181a expression was associated with improved outcomes in cytogenetically normal acute myeloid leukemia (CN-AML). Interestingly, miR-181a expression was increased in CN-AML patients harboring CEBPA mutations, which are usually biallelic and associate with better prognosis. CEBPA encodes the C/EBPα transcription factor. We demonstrate here that the presence of N-terminal CEBPA mutations and miR-181a expression are linked. Indeed, the truncated C/EBPα-p30 isoform, which is produced from the N-terminal mutant CEBPA gene or from the differential translation of wild-type CEBPA mRNA and is commonly believed to have no transactivation activity, binds to the miR-181a-1 promoter and up-regulates the microRNA expression. Furthermore, we show that lenalidomide, a drug approved for myelodysplastic syndromes and multiple myeloma, enhances translation of the C/EBPα-p30 isoform, resulting in higher miR-181a levels. In xenograft mouse models, ectopic miR-181a expression inhibits tumor growth. Similarly, lenalidomide exhibits antitumorigenic activity paralleled by increased miR-181a expression. This regulatory pathway may explain an increased sensitivity to apoptosis-inducing chemotherapy in subsets of AML patients. Altogether, our data provide a potential explanation for the improved clinical outcomes observed in CEBPA-mutated CN-AML patients, and suggest that lenalidomide treatment enhancing the C/EBPα-p30 protein levels and in turn miR-181a may sensitize AML blasts to chemotherapy.


Cell | 2018

R-2HG Exhibits Anti-tumor Activity by Targeting FTO/m6A/MYC/CEBPA Signaling

Rui Su; Lei Dong; Chenying Li; Sigrid Nachtergaele; Mark Wunderlich; Ying Qing; Xiaolan Deng; Yungui Wang; Xiaocheng Weng; Chao Hu; Mengxia Yu; Jennifer R. Skibbe; Qing Dai; Dongling Zou; Tong Wu; Kangkang Yu; Hengyou Weng; Huilin Huang; Kyle Ferchen; Xi Qin; Bin Zhang; Jun Qi; Atsuo T. Sasaki; David R. Plas; James E. Bradner; Minjie Wei; Guido Marcucci; Xi Jiang; James C. Mulloy; Jie Jin

R-2-hydroxyglutarate (R-2HG), produced at high levels by mutant isocitrate dehydrogenase 1/2 (IDH1/2) enzymes, was reported as an oncometabolite. We show here that R-2HG also exerts a broad anti-leukemic activity inxa0vitro and inxa0vivo by inhibiting leukemia cell proliferation/viability and by promoting cell-cycle arrest and apoptosis. Mechanistically, R-2HG inhibits fat mass and obesity-associated protein (FTO) activity, thereby increasing global N6-methyladenosine (m6A) RNA modification in R-2HG-sensitive leukemia cells, which in turn decreases the stability of MYC/CEBPA transcripts, leading to the suppression of relevant pathways. Ectopically expressed mutant IDH1 and S-2HG recapitulate the effects of R-2HG. High levels of FTO sensitize leukemic cells to R-2HG, whereas hyperactivation of MYC signaling confers resistance that can be reversed by the inhibition of MYC signaling. R-2HG also displays anti-tumor activity in glioma. Collectively, while R-2HG accumulated in IDH1/2 mutant cancers contributes to cancer initiation, our work demonstrates anti-tumor effectsxa0of 2HG in inhibiting proliferation/survival of FTO-high cancer cells via targeting FTO/m6A/MYC/CEBPA signaling.


Leukemia | 2016

The dual epigenetic role of PRMT5 in acute myeloid leukemia: gene activation and repression via histone arginine methylation.

Somayeh S. Tarighat; Ramasamy Santhanam; David Frankhouser; Radomska Hs; H Lai; Mirela Anghelina; Hengbin Wang; Xiaomeng Huang; Lapo Alinari; Alison Walker; Michael A. Caligiuri; Carlo M. Croce; Linsen Li; Ramiro Garzon; Chenglong Li; Robert A. Baiocchi; Guido Marcucci

Changes in the enzymatic activity of protein arginine methyltransferase (PRMT) 5 have been associated with cancer; however, the protein’s role in acute myeloid leukemia (AML) has not been fully evaluated. Here, we show that increased PRMT5 activity enhanced AML growth in vitro and in vivo while PRMT5 downregulation reduced it. In AML cells, PRMT5 interacted with Sp1 in a transcription repressor complex and silenced miR-29b preferentially via dimethylation of histone 4 arginine residue H4R3. As Sp1 is also a bona fide target of miR-29b, the miR silencing resulted in increased Sp1. This event in turn led to transcription activation of FLT3, a gene that encodes a receptor tyrosine kinase. Inhibition of PRMT5 via sh/siRNA or a first-in-class small-molecule inhibitor (HLCL-61) resulted in significantly increased expression of miR-29b and consequent suppression of Sp1 and FLT3 in AML cells. As a result, significant antileukemic activity was achieved. Collectively, our data support a novel leukemogenic mechanism in AML where PRMT5 mediates both silencing and transcription of genes that participate in a ‘yin-yang’ functional network supporting leukemia growth. As FLT3 is often mutated in AML and pharmacologic inhibition of PRMT5 appears feasible, the PRMT5–miR-29b–FLT3 network should be further explored as a novel therapeutic target for AML.


Journal of The National Comprehensive Cancer Network | 2017

Acute myeloid leukemia, version 3.2017: Clinical practice guidelines in oncology

Margaret R. O'Donnell; Martin S. Tallman; Camille N. Abboud; Jessica K. Altman; Frederick R. Appelbaum; Daniel A. Arber; Vijaya Raj Bhatt; Dale Bixby; William Blum; Steven Coutre; Marcos de Lima; Amir T. Fathi; Melanie Fiorella; James M. Foran; Steven D. Gore; Aric C. Hall; Patricia Kropf; Jeffrey E. Lancet; Lori J. Maness; Guido Marcucci; Mike G. Martin; Joseph O. Moore; Rebecca L. Olin; Deniz Peker; Daniel A. Pollyea; Keith W. Pratz; Farhad Ravandi; Paul J. Shami; Richard Stone; Stephen A. Strickland

Acute myeloid leukemia (AML) is the most common form of acute leukemia among adults and accounts for the largest number of annual deaths due to leukemias in the United States. This portion of the NCCN Guidelines for AML focuses on management and provides recommendations on the workup, diagnostic evaluation, and treatment options for younger (age <60 years) and older (age ≥60 years) adult patients.


Nature Genetics | 2016

Deregulation of DUX4 and ERG in acute lymphoblastic leukemia

Jinghui Zhang; Kelly McCastlain; Hiroki Yoshihara; Beisi Xu; Yunchao Chang; Michelle L. Churchman; Gang Wu; Yongjin Li; Lei Wei; Ilaria Iacobucci; Yu Liu; Chunxu Qu; Ji Wen; Michael Edmonson; Debbie Payne-Turner; Kerstin B Kaufmann; Shin-ichiro Takayanagi; Erno Wienholds; Esmé Waanders; Panagiotis Ntziachristos; Sofia Bakogianni; Jingjing Wang; Iannis Aifantis; Kathryn G. Roberts; Jing Ma; Guangchun Song; John Easton; Heather L. Mulder; Xiang Chen; Scott Newman

Chromosomal rearrangements deregulating hematopoietic transcription factors are common in acute lymphoblastic leukemia (ALL). Here we show that deregulation of the homeobox transcription factor gene DUX4 and the ETS transcription factor gene ERG is a hallmark of a subtype of B-progenitor ALL that comprises up to 7% of B-ALL. DUX4 rearrangement and overexpression was present in all cases and was accompanied by transcriptional deregulation of ERG, expression of a novel ERG isoform, ERGalt, and frequent ERG deletion. ERGalt uses a non-canonical first exon whose transcription was initiated by DUX4 binding. ERGalt retains the DNA-binding and transactivation domains of ERG, but it inhibits wild-type ERG transcriptional activity and is transforming. These results illustrate a unique paradigm of transcription factor deregulation in leukemia in which DUX4 deregulation results in loss of function of ERG, either by deletion or induced expression of an isoform that is a dominant-negative inhibitor of wild-type ERG function.


Leukemia | 2015

Pharmacological targeting of miR-155 via the NEDD8-activating enzyme inhibitor MLN4924 (Pevonedistat) in FLT3-ITD acute myeloid leukemia

Jihane Khalife; Radomska Hs; Ramasamy Santhanam; Xiaomeng Huang; Paolo Neviani; Jennifer N. Saultz; Hongyan Wang; Yue-Zhong Wu; Houda Alachkar; Mirela Anghelina; Adrienne M. Dorrance; John Curfman; Clara D. Bloomfield; Bruno C. Medeiros; Danilo Perrotti; Ly James Lee; Robert J. Lee; Michael A. Caligiuri; Flavia Pichiorri; Carlo M. Croce; Ramiro Garzon; Guzman Ml; Jason H. Mendler; Guido Marcucci

High levels of microRNA-155 (miR-155) are associated with poor outcome in acute myeloid leukemia (AML). In AML, miR-155 is regulated by NF-κB, the activity of which is, in part, controlled by the NEDD8-dependent ubiquitin ligases. We demonstrate that MLN4924, an inhibitor of NEDD8-activating enzyme presently being evaluated in clinical trials, decreases binding of NF-κB to the miR-155 promoter and downregulates miR-155 in AML cells. This results in the upregulation of the miR-155 targets SHIP1, an inhibitor of the PI3K/Akt pathway, and PU.1, a transcription factor important for myeloid differentiation, leading to monocytic differentiation and apoptosis. Consistent with these results, overexpression of miR-155 diminishes MLN4924-induced antileukemic effects. In vivo, MLN4924 reduces miR-155 expression and prolongs the survival of mice engrafted with leukemic cells. Our study demonstrates the potential of miR-155 as a novel therapeutic target in AML via pharmacologic interference with NF-κB-dependent regulatory mechanisms. We show the targeting of this oncogenic microRNA with MLN4924, a compound presently being evaluated in clinical trials in AML. As high miR-155 levels have been consistently associated with aggressive clinical phenotypes, our work opens new avenues for microRNA-targeting therapeutic approaches to leukemia and cancer patients.


Leukemia | 2015

Targeting Leukemia Stem Cells in vivo with AntagomiR-126 Nanoparticles in Acute Myeloid Leukemia

Adrienne M. Dorrance; Paolo Neviani; Gregory Ferenchak; Xiaomeng Huang; Deedra Nicolet; K. Maharry; Hatice Gulcin Ozer; P Hoellarbauer; Jihane Khalife; E B Hill; M Yadav; Brad Bolon; Robert J. Lee; Ly James Lee; Carlo M. Croce; Ramiro Garzon; Michael A. Caligiuri; Clara D. Bloomfield; Guido Marcucci

Current treatments for acute myeloid leukemia (AML) are designed to target rapidly dividing blast populations with limited success in eradicating the functionally distinct leukemia stem cell (LSC) population, which is postulated to be responsible for disease resistance and relapse. We have previously reported high miR-126 expression levels to be associated with a LSC-gene expression profile. Therefore, we hypothesized that miR-126 contributes to ‘stemness’ and is a viable target for eliminating the LSC in AML. Here we first validate the clinical relevance of miR-126 expression in AML by showing that higher expression of this microRNA (miR) is associated with worse outcome in a large cohort of older (⩾60 years) cytogenetically normal AML patients treated with conventional chemotherapy. We then show that miR-126 overexpression characterizes AML LSC-enriched cell subpopulations and contributes to LSC long-term maintenance and self-renewal. Finally, we demonstrate the feasibility of therapeutic targeting of miR-126 in LSCs with novel targeting nanoparticles containing antagomiR-126 resulting in in vivo reduction of LSCs likely by depletion of the quiescent cell subpopulation. Our findings suggest that by targeting a single miR, that is, miR-126, it is possible to interfere with LSC activity, thereby opening potentially novel therapeutic approaches to treat AML patients.


Leukemia | 2013

A stem cell-like gene expression signature associates with inferior outcomes and a distinct microRNA expression profile in adults with primary cytogenetically normal acute myeloid leukemia

Klaus H. Metzeler; K. Maharry; Jessica Kohlschmidt; Stefano Volinia; Krzysztof Mrózek; Heiko Becker; Deedra Nicolet; Susan P. Whitman; Jason H. Mendler; Sebastian Schwind; Ann-Kathrin Eisfeld; Yue-Zhong Wu; Bayard L. Powell; Thomas H. Carter; Meir Wetzler; Jonathan E. Kolitz; Maria R. Baer; Andrew J. Carroll; Richard Stone; Michael A. Caligiuri; Guido Marcucci; Clara D. Bloomfield

Acute myeloid leukemia (AML) is hypothesized to be sustained by self-renewing leukemia stem cells (LSCs). Recently, gene expression signatures (GES) from functionally defined AML LSC populations were reported, and expression of a ‘core enriched’ (CE) GES, representing 44 genes activated in LCSs, conferred shorter survival in cytogenetically normal (CN) AML. The prognostic impact of the CE GES in the context of other molecular markers, including gene mutations and microRNA (miR) expression alterations, is unknown and its clinical utility is unclear. We studied associations of the CE GES with known molecular prognosticators, miR expression profiles, and outcomes in 364 well-characterized CN-AML patients. A high CE score (CEhigh) associated with FLT3-internal tandem duplication, WT1 and RUNX1 mutations, wild-type CEBPA and TET2, and high ERG, BAALC and miR-155 expression. CEhigh patients had a lower complete remission (CR) rate (P=0.003) and shorter disease-free (DFS, P<0.001) and overall survival (OS, P<0.001) than CElow patients. These associations persisted in multivariable analyses adjusting for other prognosticators (CR, P=0.02; DFS, P<0.001; and OS, P<0.001). CEhigh status was accompanied by a characteristic miR expression signature. Fifteen miRs were upregulated in both younger and older CEhigh patients, including miRs relevant for stem cell function. Our results support the clinical relevance of LSCs and improve risk stratification in AML.


Cell Stem Cell | 2015

HDAC8 Inhibition Specifically Targets Inv(16) Acute Myeloid Leukemic Stem Cells by Restoring p53 Acetylation.

Jing Qi; Sandeep Singh; Wei Kai Hua; Qi Cai; Shi Wei Chao; Ling Li; Hongjun Liu; Yinwei Ho; Tinisha McDonald; Allen Lin; Guido Marcucci; Ravi Bhatia; Wei Jan Huang; Chung I. Chang; Ya Huei Kuo

Acute myeloid leukemia (AML) is driven and sustained by leukemia stem cells (LSCs) with unlimited self-renewal capacity and resistance to chemotherapy. Mutation in the TP53 tumor suppressor is relatively rare in de novo AML; however, p53 can be regulated through post-translational mechanisms. Here, we show that p53 activity is inhibited in inv(16)(+) AML LSCs via interactions with the CBFβ-SMMHC (CM) fusion protein and histone deacetylase 8 (HDAC8). HDAC8 aberrantly deacetylates p53 and promotes LSC transformation and maintenance. HDAC8 deficiency or inhibition using HDAC8-selective inhibitors (HDAC8i) effectively restores p53 acetylation and activity. Importantly, HDAC8 inhibition induces apoptosis in inv(16)(+) AML CD34(+) cells, while sparing the normal hematopoietic stem cells. Furthermore, in vivo HDAC8i administration profoundly diminishes AML propagation and abrogates leukemia-initiating capacity of both murine and patient-derived LSCs. This study elucidates an HDAC8-mediated p53-inactivating mechanism promoting LSC activity and highlights HDAC8 inhibition as a promising approach to selectively target inv(16)(+) LSCs.

Collaboration


Dive into the Guido Marcucci's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Stephen J. Forman

City of Hope National Medical Center

View shared research outputs
Top Co-Authors

Avatar

Vinod Pullarkat

City of Hope National Medical Center

View shared research outputs
Top Co-Authors

Avatar

Anthony S. Stein

City of Hope National Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ibrahim Aldoss

City of Hope National Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Joycelynne Palmer

City of Hope National Medical Center

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge