Network


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

Hotspot


Dive into the research topics where Duncan H. Mak is active.

Publication


Featured researches published by Duncan H. Mak.


Journal of Clinical Oncology | 2009

Phase I/II Trial of AEG35156 X-Linked Inhibitor of Apoptosis Protein Antisense Oligonucleotide Combined With Idarubicin and Cytarabine in Patients With Relapsed or Primary Refractory Acute Myeloid Leukemia

Aaron D. Schimmer; Elihu H. Estey; Gautam Borthakur; Bing Z. Carter; Gary J. Schiller; Martin S. Tallman; Jessica K. Altman; Judith E. Karp; Jeannine Kassis; David W. Hedley; Joseph Brandwein; Wei Xu; Duncan H. Mak; Eric C. LaCasse; Christine Jacob; Stephen J. Morris; Jacques Jolivet; Michael Andreeff

PURPOSE X-linked inhibitor of apoptosis protein (XIAP) is an inhibitor of caspases 3 and 9 which are overexpressed in acute myeloid leukemia (AML) and may contribute to chemoresistance. We report on a phase I/II trial of the XIAP antisense oligonucleotide AEG35156 in combination with reinduction chemotherapy. PATIENTS AND METHODS Twenty-four patients with rapidly relapsed or refractory AML were treated with escalating doses of AEG35156 (12 to 250 mg/m(2)) as an intravenous solution over 2 hours and 32 patients were treated with the highest planned dose of 350 mg/m(2) in combination with idarubicin and high-dose cytarabine reinduction chemotherapy. Correlative studies were conducted to determine the effects of AEG35156 on levels of XIAP mRNA. RESULTS Knockdown of XIAP mRNA during treatment increased with the dose of the antisense. All patients who received 350 mg/m(2) of AEG35156 had higher than 30% target knockdown with a median maximal knockdown of 90% (range, 48% to 100%). The overall response rate was higher among the patients receiving the highest dose of AEG35156. In this group, 15 (47%) of 32 patients achieved complete response (CR)/CR with incomplete platelet count recovery (CRp) compared with only one (4%) of 24 receiving 12 to 250 mg/m(2) AEG35156. Among the patients receiving 350 mg/m(2) of AEG35156 in combination with chemotherapy, 10 (91%) of 11 who were refractory to a single induction chemotherapy regimen achieved CR/CRp after reinduction with AEG35156 and chemotherapy. AEG35156 was well tolerated save for two cases of peripheral neuropathy in patients receiving multiple doses of AEG35156. CONCLUSION At the highest dose tested, AEG35156 knocks down its target and appears very effective when combined with chemotherapy in patients with AML refractory to a single induction regimen.


Blood | 2008

Triptolide sensitizes AML cells to TRAIL-induced apoptosis via decrease of XIAP and p53-mediated increase of DR5

Bing Z. Carter; Duncan H. Mak; Wendy D. Schober; Martin Dietrich; Clemencia Pinilla; Lyubomir T. Vassilev; John C. Reed; Michael Andreeff

Acute myeloid leukemia (AML) cells are relatively resistant to tumor necrosis factor alpha-related apoptosis-inducing ligand (TRAIL). We previously reported that triptolide, a potent anticancer agent from a Chinese herb, decreases XIAP in leukemic cells. We evaluated the combination of triptolide and TRAIL and found synergistic promotion of apoptosis in AML cells. XIAP-overexpressing U937 cells (U937XIAP) were more resistant to TRAIL than U937neo cells, and inhibition of XIAP with the small-molecule inhibitor 1396-11 enhanced TRAIL-induced apoptosis, implying XIAP as a resistance factor in AML. Furthermore, triptolide increased DR5 levels in OCI-AML3, while the DR5 increase was blunted in p53-knockdown OCI-AML3 and p53-mutated U937 cells, confirming a role for p53 in the regulation of DR5. In support of this finding, disruption of MDM2-p53 binding with subsequent increase in p53 levels by nutlin3a increased DR5 levels and sensitized OCI-AML3 cells to TRAIL. The combination of 1396-11 plus nutlin3a plus TRAIL was more effective than either the 1396-11 and TRAIL or nutlin3a and TRAIL combinations in OCI-AML3 cells, further supporting the role of triptolide as a sensitizer to TRAIL-induced apoptosis in part by independent modulation of XIAP expression and p53 signaling. Thus, the combination of triptolide and TRAIL may provide a novel strategy for treating AML by overcoming critical mechanisms of apoptosis resistance.


Leukemia | 2012

MEK Inhibition Enhances ABT-737-Induced Leukemia Cell Apoptosis via Prevention of ERK activated MCL-1 induction and Modulation of MCL-1/BIM Complex

Marina Konopleva; Michele Milella; Peter P. Ruvolo; J C Watts; Maria Rosaria Ricciardi; Borys Korchin; Teresa McQueen; William G. Bornmann; Twee Tsao; P Bergamo; Duncan H. Mak; Weina Chen; James A. McCubrey; Agostino Tafuri; Michael Andreeff

Recently, strategies for acute myeloid leukemia (AML) therapy have been developed that target anti-apoptotic BCL2 family members using BH3-mimetic drugs such as ABT-737. Though effective against BCL2 and BCL-XL, ABT-737 poorly inhibits MCL-1. Here we report that, unexpectedly, ABT-737 induces activation of the extracellular receptor activated kinase and induction of MCL-1 in AML cells. MEK inhibitors such as PD0325901 and CI-1040 have been used successfully to suppress MCL-1. We report that PD0325901 blocked ABT-737-induced MCL-1 expression, and when combined with ABT-737 resulted in potent synergistic killing of AML-derived cell lines, primary AML blast and CD34+38-123+ progenitor/stem cells. Finally, we tested the combination of ABT-737 and CI-1040 in a murine xenograft model using MOLM-13 human leukemia cells.Whereas control mice and CI-1040-treated mice exhibited progressive leukemia growth, ABT-737, and to a significantly greater extent, ABT-737+CI-1040 exerted major anti-leukemia activity. Collectively, results demonstrated unexpected anti-apoptotic interaction between the BCL2 family-targeted BH3-mimetic ABT-737 and mitogen-activated protein kinase signaling in AML cells: the BH3 mimetic is not only restrained in its activity by MCL-1, but also induces its expression. However, concomitant inhibition by BH3 mimetics and MEK inhibitors could abrogate this effect and may be developed into a novel and effective therapeutic strategy for patients with AML.


Blood | 2012

Survivin is highly expressed in CD34 +38 - leukemic stem/progenitor cells and predicts poor clinical outcomes in AML

Bing Z Carter; Yihua Qiu; Xuelin Huang; Lixia Diao; Nianxiang Zhang; Kevin R. Coombes; Duncan H. Mak; Marina Konopleva; Jorge E Cortes; Hagop M Kantarjian; Gordon B. Mills; Michael Andreeff; Steven M. Kornblau

Survivin, a member of the inhibitors of apoptosis protein family, plays important roles in cell proliferation and survival and is highly expressed in various malignancies, including leukemias. To better understand its role in acute myeloid leukemia (AML), we profiled survivin expression in samples obtained from 511 newly diagnosed AML patients and in CD34(+)38(-) AML stem/progenitor cells using a validated reverse-phase protein array; we correlated its levels with clinical outcomes and with levels of other proteins in the same sample set. We found that survivin levels were higher in bone marrow than in paired peripheral blood leukemic cells (n = 140, P = .0001) and that higher survivin levels significantly predicted shorter overall (P = .016) and event-free (P = .023) survival in multivariate Cox model analysis. Importantly, survivin levels were significantly higher in CD34(+)38(-) AML stem/progenitor cells than in bulk blasts and total CD34(+) AML cells (P < .05). Survivin expression correlated with the expressions of multiple proteins involved with cell proliferation and survival. Particularly, its expression strongly correlated with HIF1α in the stem/progenitor cell compartment. These results suggest that survivin is a prognostic biomarker in AML and that survivin, which is overexpressed in AML stem/progenitor cells, remains a potentially important target for leukemia therapy.


Blood | 2010

Simultaneous activation of p53 and inhibition of XIAP enhance the activation of apoptosis signaling pathways in AML.

Bing Z. Carter; Duncan H. Mak; Wendy D. Schober; Erich Koller; Clemencia Pinilla; Lyubomir T. Vassilev; John C. Reed; Michael Andreeff

Activation of p53 by murine double minute (MDM2) antagonist nutlin-3a or inhibition of X-linked inhibitor of apoptosis (XIAP) induces apoptosis in acute myeloid leukemia (AML) cells. We demonstrate that concomitant inhibition of MDM2 by nutlin-3a and of XIAP by small molecule antagonists synergistically induced apoptosis in p53 wild-type OCI-AML3 and Molm13 cells. Knockdown of p53 by shRNA blunted the synergy, and down-regulation of XIAP by antisense oligonucleotide (ASO) enhanced nutlin-3a-induced apoptosis, suggesting that the synergy was mediated by p53 activation and XIAP inhibition. This is supported by data showing that inhibition of both MDM2 and XIAP by their respective ASOs induced significantly more cell death than either ASO alone. Importantly, p53 activation and XIAP inhibition enhanced apoptosis in blasts from patients with primary AML, even when the cells were protected by stromal cells. Mechanistic studies demonstrated that XIAP inhibition potentiates p53-induced apoptosis by decreasing p53-induced p21 and that p53 activation enhances XIAP inhibition-induced cell death by promoting mitochondrial release of second mitochondria-derived activator of caspases (SMAC) and by inducing the expression of caspase-6. Because both XIAP and p53 are presently being targeted in ongoing clinical trials in leukemia, the combination strategy holds promise for expedited translation into the clinic.


Cytometry Part A | 2015

Single-cell mass cytometry reveals intracellular survival/proliferative signaling in FLT3-ITD-mutated AML stem/progenitor cells.

Lina Han; Peng Qiu; Zhihong Zeng; Jeffrey L. Jorgensen; Duncan H. Mak; Jared K. Burks; Wendy D. Schober; Teresa McQueen; Jorge Cortes; Scott D. Tanner; Gail J. Roboz; Hagop M. Kantarjian; Steven M. Kornblau; Monica L. Guzman; Michael Andreeff; Marina Konopleva

Understanding the unique phenotypes and complex signaling pathways of leukemia stem cells (LSCs) will provide insights and druggable targets that can be used to eradicate acute myeloid leukemia (AML). Current work on AML LSCs is limited by the number of parameters that conventional flow cytometry (FCM) can analyze because of cell autofluorescence and fluorescent dye spectral overlap. Single‐cell mass cytometry (CyTOF) substitutes rare earth elements for fluorophores to label antibodies, which allows measurements of up to 120 parameters in single cells without correction for spectral overlap. The aim of this study was the evaluation of intracellular signaling in antigen‐defined stem/progenitor cell subsets in primary AML. CyTOF and conventional FCM yielded comparable results on LSC phenotypes defined by CD45, CD34, CD38, CD123, and CD99. Intracellular phosphoprotein responses to ex vivo cell signaling inhibitors and cytokine stimulation were assessed in myeloid leukemia cell lines and one primary AML sample. CyTOF and conventional FCM results were confirmed by western blotting. In the primary AML sample, we investigated the cell responses to ex vivo stimulation with stem cell factor and BEZ235‐induced inhibition of PI3K and identified activation patterns in multiple PI3K downstream signaling pathways including p‐4EBP1, p‐AKT, and p‐S6, particularly in CD34+ subsets. We evaluated multiple signaling pathways in antigen‐defined subpopulations in primary AML cells with FLT3‐ITD mutations. The data demonstrated the heterogeneity of cell phenotype distribution and distinct patterns of signaling activation across AML samples and between AML and normal samples. The mTOR targets p‐4EBP1 and p‐S6 were exclusively found in FLT3‐ITD stem/progenitor cells, but not in their normal counterparts, suggesting both as novel targets in FLT3 mutated AML. Our data suggest that CyTOF can identify functional signaling pathways in antigen‐defined subpopulations in primary AML, which may provide a rationale for designing therapeutics targeting LSC‐enriched cell populations.


Journal of the National Cancer Institute | 2014

Synergistic Targeting of AML Stem/Progenitor Cells With IAP Antagonist Birinapant and Demethylating Agents

Bing Z. Carter; Po Yee Mak; Duncan H. Mak; Yuexi Shi; Yihua Qiu; James M Bogenberger; Hong Mu; Raoul Tibes; Hui Yao; Kevin R. Coombes; Rodrigo Jacamo; Teresa McQueen; Steven M. Kornblau; Michael Andreeff

BACKGROUND Acute myeloid leukemia (AML) therapy has limited long-term efficacy because patients frequently develop disease relapse because of the inability of standard chemotherapeutic agents to target AML stem/progenitor cells. Here, we identify deregulated apoptotic components in AML stem/progenitor cells and investigate the individual and combinatorial effects of the novel inhibitor of apoptosis (IAP) protein antagonist and second mitochondrial-derived activator of caspases (SMAC) mimetic birinapant and demethylating epigenetic modulators. METHODS Protein expression was measured by reversed-phase protein array in AML patient (n = 511) and normal (n = 21) samples and by western blot in drug-treated cells. The antileukemic activity of birinapant and demethylating agents was assessed in vitro and in an in vivo AML mouse xenograft model (n = 10 mice per group). All statistical tests were two-sided. RESULTS Compared with bulk AML cells, CD34(+)38(-) AML stem/progenitors expressed increased cIAP1 and caspase-8 levels and decreased SMAC levels (one-way analysis of variance followed by Tukeys multiple comparison test, P < .001). Birinapant induced death receptor-/caspase-8-mediated apoptosis in AML cells, including in AML stem/progenitor cells, but not in normal CD34(+) cells. Demethylating agents modulated extrinsic apoptosis pathway components and, when combined with birinapant, were highly synergistic in vitro (combination index < 1), and also more effective in vivo (P < .001, by Student t test, for the median survival of birinapant plus 5-azacytadine vs birinapant alone or vs controls). CONCLUSIONS cIAP1, SMAC, and caspase-8 appear to play a role in AML stem cell survival, and synergistic targeting of these cells with birinapant and demethylating agents shows potential utility in leukemia therapy.


Cell Cycle | 2006

Regulation and targeting of Eg5, a mitotic motor protein in blast crisis CML: Overcoming imatinib resistance

Bing Z. Carter; Duncan H. Mak; Yuexi Shi; Wendy D. Schober; Rui Yu Wang; Marina Konopleva; Erich Koller; Nicholas M. Dean; Michael Andreeff

Patients with blast crisis (BC) CML frequently become resistant to Imatinib, a Bcr-Abltyrosine kinase-targeting agent. Eg5, a microtubule-associated motor protein has beendescribed to be highly expressed in BC CML by microarray analysis (Nowicki et al,Oncogene 22:3952-3963, 2003). We investigated the regulation of Eg5 by Bcr-Abltyrosine kinase and its potential as a therapeutic target in BC CML. Eg5 was highlyexpressed in all Philadelphia chromosome positive (Ph+) cell lines and BC CML patientsamples. Inhibition of Bcr-Abl by Imatinib downregulated Eg5 expression in ImatinibsensitiveKBM5 and HL-60p185 cells, but not in Imatinib-resistant KBM5-STI571,harboring a T315I mutation, and Bcr-Abl-negative HL-60 cells. Blocking Eg5 expressionwith antisense oligonucleotide (Eg5-ASO) or inhibiting its activity with the smallmoleculeEg5 inhibitor, S-trityl-L-cysteine induced G2/M cell cycle block and subsequentcell death in both Imatinib-sensitive and -resistant cells. Further, Eg5-ASO treatment ofSCID mice harboring KBM5 cell xenografts significantly prolonged the median survivalof the animals (p=0.03). Our findings suggest that Eg5 is downstream of and regulated byBcr-Abl tyrosine kinase in Philadelphia chromosome positive cells. Inhibition of Eg5expression or its activity blocks cell cycle progression and induces cell death independentof the cellular response to Imatinib. Therefore, Eg5 could be a potential therapeutic targetfor the treatment of BC CML, in particular Imatinib-resistant BC CML.


Science Translational Medicine | 2016

Combined targeting of BCL-2 and BCR-ABL tyrosine kinase eradicates chronic myeloid leukemia stem cells.

Bing Z. Carter; Po Yee Mak; Hong Mu; Hongsheng Zhou; Duncan H. Mak; Wendy D. Schober; Joel D. Leverson; Bin Zhang; Ravi Bhatia; Xuelin Huang; Jorge Cortes; Hagop M. Kantarjian; Marina Konopleva; Michael Andreeff

Inhibition of BCL-2, MCL-1, and BCL-XL eradicates CML stem cells in a mouse model and primary blast crisis samples. Stemming the regrowth of leukemia Chronic myeloid leukemia is usually held up as the poster child for targeted therapy, given the success of imatinib and related compounds such as nilotinib in treating the disease. Unfortunately, even these drugs are not perfect because they do not eliminate cancer stem cells. As a result, patients have to remain on treatment indefinitely or else face a high likelihood of relapse. Carter et al. have discovered that the antiapoptotic protein BCL-2 plays a key role in the survival of chronic myeloid leukemia stem cells and that combined treatment with a BCL-2 inhibitor and nilotinib can successfully eradicate both the active tumor cells and the stem cells, suggesting the potential for curative treatment. BCR-ABL tyrosine kinase inhibitors (TKIs) are effective against chronic myeloid leukemia (CML), but they rarely eliminate CML stem cells. Disease relapse is common upon therapy cessation, even in patients with complete molecular responses. Furthermore, once CML progresses to blast crisis (BC), treatment outcomes are dismal. We hypothesized that concomitant targeting of BCL-2 and BCR-ABL tyrosine kinase could overcome these limitations. We demonstrate increased BCL-2 expression at the protein level in bone marrow cells, particularly in Lin−Sca-1+cKit+ cells of inducible CML in mice, as determined by CyTOF mass cytometry. Further, selective inhibition of BCL-2, aided by TKI-mediated MCL-1 and BCL-XL inhibition, markedly decreased leukemic Lin−Sca-1+cKit+ cell numbers and long-term stem cell frequency and prolonged survival in a murine CML model. Additionally, this combination effectively eradicated CD34+CD38−, CD34+CD38+, and quiescent stem/progenitor CD34+ cells from BC CML patient samples. Our results suggest that BCL-2 is a key survival factor for CML stem/progenitor cells and that combined inhibition of BCL-2 and BCR-ABL tyrosine kinase has the potential to significantly improve depth of response and cure rates of chronic-phase and BC CML.


Leukemia | 2009

Inhibition of KSP by ARRY-520 induces cell cycle block and cell death via the mitochondrial pathway in AML cells.

Bing Z. Carter; Duncan H. Mak; Richard Woessner; Stefan Gross; Wendy D. Schober; Zeev Estrov; Hagop M. Kantarjian; Michael Andreeff

Kinesin spindle protein (KSP), a microtubule-associated motor protein essential for cell cycle progression, is overexpressed in many cancers and is a potential anti-tumor target. We found that inhibition of KSP by a selective inhibitor, ARRY-520, blocked cell cycle progression, leading to apoptosis in acute myeloid leukemia cell lines that express high levels of KSP. Knockdown of p53, overexpression of XIAP and mutation in caspase-8 did not significantly affect sensitivity to ARRY-520, suggesting that the response is independent of p53, XIAP and the extrinsic apoptotic pathway. Although ARRY-520 induced mitotic arrest in both HL-60 and Bcl-2-overexpressing HL-60Bcl-2 cells, cell death was blunted in HL-60Bcl-2 cells, suggesting that the apoptotic program is executed through the mitochondrial pathway. Accordingly, inhibition of Bcl-2 by ABT-737 was synergistic with ARRY-520 in HL-60Bcl-2 cells. Furthermore, ARRY-520 increased Bim protein levels prior to caspase activation in HL-60 cells. ARRY-520 significantly inhibited tumor growth of xenografts in SCID mice and inhibited AML blast but not normal colony formation, supporting a critical role for KSP in proliferation of leukemic progenitor cells. These results demonstrate that ARRY-520 potently induces cell cycle block and subsequent death in leukemic cells via the mitochondrial pathway and has the potential to eradicate AML progenitor cells.

Collaboration


Dive into the Duncan H. Mak's collaboration.

Top Co-Authors

Avatar

Michael Andreeff

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Marina Konopleva

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Wendy D. Schober

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Po Yee Mak

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Hagop M. Kantarjian

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Jorge Cortes

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Steven M. Kornblau

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Teresa McQueen

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Vivian Ruvolo

University of Texas MD Anderson Cancer Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge