Network


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

Hotspot


Dive into the research topics where Barbara Czako is active.

Publication


Featured researches published by Barbara Czako.


Nature Chemical Biology | 2016

SF2312 is a natural phosphonate inhibitor of enolase

Paul G. Leonard; Nikunj Satani; David Maxwell; Yu Hsi Lin; Naima Hammoudi; Zhenghong Peng; Federica Pisaneschi; Todd M. Link; Gilbert R. Lee; Duoli Sun; Basvoju A. Bhanu Prasad; Maria Emilia Di Francesco; Barbara Czako; John M. Asara; Y. Alan Wang; William G. Bornmann; Ronald A. DePinho; Florian Muller

Despite being critical for energy generation in most forms of life, few if any microbial antibiotics specifically inhibit glycolysis. To develop a specific inhibitor of the glycolytic enzyme Enolase 2 for the treatment of cancers with deletion of Enolase 1, we modeled the synthetic tool compound inhibitor, Phosphonoacetohydroxamate (PhAH) into the active site of human ENO2. A ring-stabilized analogue of PhAH, with the hydroxamic nitrogen linked to the alpha-carbon by an ethylene bridge, was predicted to increase binding affinity by stabilizing the inhibitor in a bound conformation. Unexpectedly, a structure based search revealed that our hypothesized back-bone-stabilized PhAH bears strong similarity to SF2312, a phosphonate antibiotic of unknown mode of action produced by the actinomycete Micromonospora, which is active under anaerobic conditions. Here, we present multiple lines of evidence, including a novel X-ray structure, that SF2312 is a highly potent, low nM inhibitor of Enolase.


Nature Medicine | 2018

An inhibitor of oxidative phosphorylation exploits cancer vulnerability

Jennifer R. Molina; Yuting Sun; Marina Protopopova; Sonal Gera; Madhavi Bandi; Christopher A. Bristow; Timothy McAfoos; Pietro Morlacchi; Jeffrey Ackroyd; Ahmed Noor A. Agip; Gheath Alatrash; John M. Asara; Jennifer Bardenhagen; Caroline Carrillo; Christopher Carroll; Edward F. Chang; Stefan O. Ciurea; Jason B. Cross; Barbara Czako; Angela K. Deem; Naval Daver; John F. de Groot; Jian Wen Dong; Ningping Feng; Guang Gao; Mary Geck Do; Jennifer Greer; Virginia Giuliani; Jing Han; Lina Han

Metabolic reprograming is an emerging hallmark of tumor biology and an actively pursued opportunity in discovery of oncology drugs. Extensive efforts have focused on therapeutic targeting of glycolysis, whereas drugging mitochondrial oxidative phosphorylation (OXPHOS) has remained largely unexplored, partly owing to an incomplete understanding of tumor contexts in which OXPHOS is essential. Here, we report the discovery of IACS-010759, a clinical-grade small-molecule inhibitor of complex I of the mitochondrial electron transport chain. Treatment with IACS-010759 robustly inhibited proliferation and induced apoptosis in models of brain cancer and acute myeloid leukemia (AML) reliant on OXPHOS, likely owing to a combination of energy depletion and reduced aspartate production that leads to impaired nucleotide biosynthesis. In models of brain cancer and AML, tumor growth was potently inhibited in vivo following IACS-010759 treatment at well-tolerated doses. IACS-010759 is currently being evaluated in phase 1 clinical trials in relapsed/refractory AML and solid tumors.A new inhibitor targeting the mitochondrial complex I shows antitumor activity in preclinical models of acute myeloid leukemia and glioblastoma relying on oxidative phosphorylation.


bioRxiv | 2018

Eradication of ENO1-deleted Glioblastoma through Collateral Lethality

Yu-Hsi Lin; Nikunj Satani; Naima Hammoudi; Jeffrey J Ackroyd; Sunada Khadka; Victoria Yan; Dimitra K. Georgiou; Yuting Sun; Rafal Zielinski; Theresa Tran; Susana Castro Pando; Xiaobo Wang; David Maxwell; Zhenghong Peng; Federica Pisaneschi; Pijus K. Mandal; Paul G. Leonard; Quanyu Xu; Qi Wu; Yongying Jiang; Barbara Czako; Zhijun Kang; John M. Asara; Waldemar Priebe; William G. Bornmann; Joseph R. Marszalek; Ronald A. DePinho; Florian Muller

Inhibiting glycolysis remains an aspirational approach for the treatment of cancer. We recently demonstrated that SF2312, a natural product phosphonate antibiotic, is a potent inhibitor of the glycolytic enzyme Enolase with potential utility for the collateral lethality-based treatment of Enolase-deficient glioblastoma (GBM). However, phosphonates are anionic at physiological pH, limiting cell and tissue permeability. Here, we show that addition of pivaloyloxymethyl (POM) groups to SF2312 (POMSF) dramatically increases potency, leading to inhibition of glycolysis and killing of ENO1-deleted glioma cells in the low nM range. But the utility of POMSF in vivo is dose-limited by severe hemolytic anemia. A derivative, POMHEX, shows equipotency to POMSF without inducing hemolytic anemia. POMHEX can eradicate intracranial orthotopic ENO1-deleted tumors, despite sub-optimal pharmacokinetic properties. Taken together, our data provide in vivo proof-of-principal for collateral lethality in precision oncology and showcase POMHEX as a useful molecule for the study of glycolysis in cancer metabolism.


Molecular Cancer Therapeutics | 2017

Abstract A39: Pomhex, a cell-permeable high potency enolase inhibitor with utility for collateral lethality treatment of cancer

Yu-Hsi Lin; Nikunj Satani; Naima Hammoudi; Federica Pisaneschi; Paul G. Leonard; David Maxwell; Zhenghong Peng; Todd M. Link; Lee Iv R. Gilbert; Ananth Bosajou; Duoli Sun; Joe Marszalek; Yuting Sun; John S. McMurray; Pijus K. Mandal; Maria Emilia Di Francesco; Barbara Czako; Alan Wang; William G. Bornmann; Ronald A. DePinho; Florian Muller

Glycolysis inhibition is an active area of investigation for the treatment of cancer. However, few compounds have progressed beyond the cell culture stage. We have recently demonstrated that genomic passenger deletion of the glycolytic enzyme Enolase 1 (ENO1) leaves gliomas harboring such deletions solely reliant on ENO2, rendering them exquisitely sensitive to enolase inhibitors Collateral Lethality. However, the tool compound that we employed for these in vitro studies, Phosphonoacetohydroxamate (PhAH), has very poor pharmacological properties and was ineffective in vivo. We recently reported that a structural analogue of PhAH, the natural phosphonate antibiotic SF2312, is a high potency inhibitor of Enolase. While more potent than PhAH, SF2312 remains poorly cell permeable. Here, we generated a Pivaloyloxymethyl (POM) ester pro-drug derivative of SF2312, termed POMSF, which increased the potency in cell based systems by ~50-fold. POMSF is selectively active against ENO1-deleted glioma cells in culture at ~19 nM, versus μM for SF2312. However, POMSF displayed poor aqueous stability. A derivative of POMSF, termed POMHEX, showed greater stability and its active form, HEX, showed 4-fold preference for ENO1 over ENO2. Labeled 13C-glucose tracing shows that POMHEX inhibits glycolysis at the Enolase step in all cell lines tested, but with ~100-fold greater potency in ENO1-deleted lines. POMHEX selectively killed ENO1-deleted glioma cells with an IC50 Citation Format: Yu-Hsi Lin, Nikunj Satani, Naima Hammoudi, Federica Pisaneschi, Paul Leonard, David Maxwell, Zhenghong Peng, Todd Link, Lee IV R. Gilbert, Ananth Bosajou, Duoli Sun, Joe Marszalek, Yuting Sun, John S. McMurray, Pijus K. Mandal, Maria E. Di Francesco, Barbara Czako, Alan Wang, William Bornmann, Ronald A. DePinho, Florian Muller. Pomhex, a cell-permeable high potency enolase inhibitor with utility for collateral lethality treatment of cancer [abstract]. In: Proceedings of the AACR Precision Medicine Series: Opportunities and Challenges of Exploiting Synthetic Lethality in Cancer; Jan 4-7, 2017; San Diego, CA. Philadelphia (PA): AACR; Mol Cancer Ther 2017;16(10 Suppl):Abstract nr A39.


Cancer Research | 2016

Abstract 3837: Passenger deletion of ENO1 as a collateral lethality target in cancer

Yu-Hsi Lin; Nikunj Satani; Naima Hammoudi; Joe Marszalek; Yuting Sun; Marina Protopopova; Maria Emilia Di Francesco; Barbara Czako; Alan Wang; Ronald A. DePinho; Florian Muller

Large scale genomic characterization efforts such as TCGA have painted an unprecedentedly detailed picture of the genetic alterations that underlie tumorigenesis. Yet, the majority of genetic alterations are passenger rather than driver events and are considered unactionable. We have previously proposed that passenger or collateral deletions could serve as pharmacologically targetable vulnerabilities Collateral Lethality, in case passenger genes are homozygously deleted and member of a paralogous gene family carrying out an essential housekeeping function. We have presented proof-of-principal, whereby passenger deletion of the glycolytic gene Enolase 1 (ENO1) as part of the 1p36-tumor suppressor locus, renders glioma cells harboring such deletions highly sensitive to ablation of its redundant paralogue, ENO2. While our original analysis identified ENO1-homozygous deletions in Glioblastoma (GBM), recent bioinformatics analyzes, backed by immunohistochemistry, show that ENO1-homozygous deletions also occur in Hepatocellular Carcinoma and Cholangiocarcinoma. In GBM, multisector analysis of primary and recurrent tumors, indicate that ENO1 deletion is an early event which is homogenously distributed through the primary tumor and persist during recurrence. To pharmacologically exploit ENO1-deletion, we have pursued two approaches. First, we have synthesized cell-permeable prodrug derivatives of the natural Enolase inhibitor SF2312. The lead compound, POMHEX, shows potent killing of ENO1-deleted glioma cells in the low nM range while ENO1-restored isogenic or normal cells can tolerate μM doses. POMHEX has a short half-life yet can eradicate intracranial xenografted ENO1-deleted tumors, provided extensive breakdown of the blood-brain barrier. Our second approach to targeting ENO1-deletion consisted of chemical biology screening of drug libraries for the ability to kill ENO1-deleted but not isogenic rescued cells. We find that ENO1-deleted cells show a dramatic sensitization to inhibitors of the mitochondrial electron transport chain. These include tool compounds such as rotenone as well as compounds not previously associated with mitochondria, such as Mubritinib and an experimental anti-neoplastic agent previously described as a HIF1-inhibitor, now known to inhibit mitochondrial Complex I. The latter agent shows potent activity against ENO1-deleted intracranial xenografts. The likely cause for this sensitivity is the inability of ENO1-deleted cells to compensatory upregulate glycolysis in response mitochondrial inhibition, the typical response of ENO1-intact glioma cells and normal cells. Together, these data indicate that passenger deletion of ENO1 is an encouraging drug-target and provide support for collateral lethality as a viable therapeutic strategy, which, given the large number of passenger deletions in the cancer genome, may be broadly applicable. Citation Format: Yu-Hsi Lin, Nikunj Satani, Naima Hammoudi, Joe Marszalek, Yuting Sun, Marina Protopopova, Maria E. Di Francesco, Barbara Czako, Alan Wang, Ronald A. DePinho, Florian L. Muller. Passenger deletion of ENO1 as a collateral lethality target in cancer. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 3837.


Molecular Cancer Therapeutics | 2015

Abstract C183: Pomhex: a cell-permeable high potency Enolase inhibitor with in vivo anti-neoplastic activity

Yu-Hsi Lin; Joe Marszalek; Yuting Sun; Naima Hammoudi; Paul G. Leonard; David Maxwell; Nikunj Satani; Peng Zhang; Todd M. Link; Gilbert R. Lee; Maria Emilia Di Francesco; Barbara Czako; Alan Y. Want; Ronald A. DePinho; Florian Muller

Glycolysis inhibition is an active area of investigation in cancer. However, few compounds have progressed beyond the cell culture stage. We have recently demonstrated that genomic passenger deletion of the glycolytic enzyme Enolase 1 (ENO1) leaves gliomas harboring such deletions with less than 10% of normal enzymatic activity, rendering them exquisitely sensitive to enolase inhibitors. However, the tool compound that we employed for these in vitro studies, Phosphonoacetohydroxamate (PhAH), has very poor pharmacological properties and was ineffective in vivo. We performed a SAR studies to increase inhibitor specificity towards ENO2 as well as pro-druging to increase cell permeability. The lead compound generated by these efforts, termed POMHEX, is selectively active against ENO1-deleted glioma cells in culture at ∼35nM (versus μM for PhAH). Using an orthotopic intracranial xenografted model where tumor growth and response to therapy are monitored by MRI, we show that POMHEX is capable of eradicating intracranial ENO1-deleted tumors, with mice remaining recurrence-free even after treatment discontinuation. Taken together, these results reinforce that glycolysis is a viable target and provide in vivo proof-of-principal for the concept of using passenger deletions as targetable vulnerabilities in cancer therapy. Citation Format: Yu-Hsi Lin, Joe Marszalek, Yuting Sun, Naima Hammoudi, Paul Leonard, David Maxwell, Nikunj Satani, Peng Zhang, Todd Link, Gilbert Lee, Maria E. Di Francesco, Barbara Czako, Alan Y. Want, Ronald A. DePinho, Florian L. Muller. Pomhex: a cell-permeable high potency Enolase inhibitor with in vivo anti-neoplastic activity. [abstract]. In: Proceedings of the AACR-NCI-EORTC International Conference: Molecular Targets and Cancer Therapeutics; 2015 Nov 5-9; Boston, MA. Philadelphia (PA): AACR; Mol Cancer Ther 2015;14(12 Suppl 2):Abstract nr C183.


Cancer Research | 2014

Abstract 949: Identification of OXPHOS inhibitors which selectively kill tumors with specific metabolic vulnerabilities

Joseph R. Marszalek; Madhavi Bandi; Jennifer Bardenhagen; Christopher A. Bristow; Christopher Carroll; Edward F. Chang; Ninping Feng; Barbara Czako; Mary Geck Do; Jennifer Greer; Ryan Johnson; Marina Konopleva; Zhijun Kang; Gang Liu; Timothy Lofton; Timothy McAfoos; Marina Protopopova; Alessia Petrocchi; Florian Muller; Jay Theroff; Yuanqing Wu; Lynda Chin; Giulio Draetta; Philip Jones; Carlo Toniatti; Emilia Di Francesco

Inhibition of mitochondria complex I in tumors that are metabolically dependent on oxidative phosphorylation (OXPHOS) for their survival offers unique synthetic lethal opportunities. Examples of dependent contexts are AML and DLBCL, where OXPHOS is highly active and subpopulations of glioblastoma and neuroblastoma that possess genetic alterations which make them glycolysis deficient. In addition, several lines of evidence indicate that after treatment with chemo or targeted therapy, residual tumor cells become reliant on OXPHOS for their continued survival. In each of these cellular states, excessive dependence on OXPHOS renders tumor cells vulnerable to therapeutic targeting strategies that exploit this addiction. We have generated a series of novel, highly potent complex I inhibitors, which in vitro inhibit complex I with IC 50 values 50 values between 1-10 nM. Lead compounds specifically induce apoptosis with EC 50 values between 1-10 nM in OXPHOS dependent cancer models such as AML and DLBCL cell lines and in glycolysis deficient cancer cell lines. Of note, apoptosis is induced in primary AML cells but not in normal patient-derived CD34+ cells. These compounds are orally bioavailable with excellent pharmacokinetics properties in preclinical species making them appropriate tools for proof-of-concept studies in vivo. In agreement with data in cell culture, we have shown that daily oral treatment with as low as 5 -10 mg/kg of our OXPHOS inhibitors is well tolerated and induce strong regression of NB-1 (glycolysis-deficient cells) subcutaneous and intracranial xenografts. We have also demonstrated that sustained pharmacological inhibition of OXPHOS induce regression of DLBCL subcutaneous models and dramatically increase mice survival in an OCI-AML3 orthotopic xenograft model. In addition to synthetic lethality in monotherapy, we are exploring whether OXPHOS inhibition can overcome resistance to radiotherapy, chemotherapy and specific targeted therapies. Taken together, these data strongly support the notion that inhibiting OXPHOS in hypersensitive populations could be a novel, innovative therapeutic approach and justifies evaluation of OXPHOS inhibitors in a clinical setting. Citation Format: Joseph R. Marszalek, Madhavi Bandi, Jennifer Bardenhagen, Christopher Bristow, Christopher Carroll, Edward Chang, Ninping Feng, Barbara Czako, Jason Gay, Mary Geck Do, Jennifer Greer, Ryan M. Johnson, Marina Konopleva, Zhijun Kang, Gang Liu, Timothy Lofton, Timothy McAfoos, Marina Protopopova, Alessia Petrocchi, Florian Muller, Jay Theroff, Yuanqing Wu, Lynda Chin, Giulio Draetta, Philip Jones, Carlo Toniatti, Emilia Di Francesco. Identification of OXPHOS inhibitors which selectively kill tumors with specific metabolic vulnerabilities. [abstract]. In: Proceedings of the 105th Annual Meeting of the American Association for Cancer Research; 2014 Apr 5-9; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2014;74(19 Suppl):Abstract nr 949. doi:10.1158/1538-7445.AM2014-949


Archive | 2014

Heterocyclic modulators of hif activity for treatment of disease

Philip Jones; Maria Emilia Difrancesco; Alessia Petrocchi; Christopher Carroll; Joe Marszalek; Barbara Czako; Ryan Johnson; Jay Theroff


Archive | 2016

Gls1 inhibitors for treating disease

Maria Emilia Di Francesco; Philip Jones; Timothy Heffernan; Matthew M. Hamilton; Zhijun Kang; Michael J. Soth; Jason P. Burke; Kang Le; Christopher Lawrence Carroll; Wylie S. Palmer; Richard Lewis; Timothy McAfoos; Barbara Czako; Gang Liu; Jay Theroff; Zachary Herrera; Anne Yau


Archive | 2018

ENOLASE INHIBITORS AND METHODS OF TREATMENT THEREWITH

Florian Muller; David Maxwell; William G. Bornmann; Yu-hsi Lin; Basvoju A. Bhanu Prasad; Zhenghong Peng; Duoli Sun; Nikunj Satani; Emilia M. Di Francesco; Ronald A. DePinho; Barbara Czako; Federica Pisaneschi

Collaboration


Dive into the Barbara Czako's collaboration.

Top Co-Authors

Avatar

Florian Muller

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Nikunj Satani

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Ronald A. DePinho

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Yuting Sun

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Naima Hammoudi

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

David Maxwell

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Maria Emilia Di Francesco

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Yu-Hsi Lin

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Christopher Carroll

University of Texas MD Anderson Cancer Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge