Network


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

Hotspot


Dive into the research topics where Yu-Hsi Lin is active.

Publication


Featured researches published by Yu-Hsi Lin.


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.


Nature Medicine | 2018

Author Correction: Mutations in the SWI/SNF complex induce a targetable dependence on oxidative phosphorylation in lung cancer

Yonathan Lissanu Deribe; Yuting Sun; Christopher Terranova; Fatima Khan; Juan Martinez-Ledesma; Guang Gao; Robert A. Mullinax; Tin Khor; Ningping Feng; Yu-Hsi Lin; Chia-Chin Wu; Claudia Reyes; Qian Peng; Frederick Robinson; Akira Inoue; Veena Kochat; Chang-Gong Liu; John M. Asara; Cesar A. Moran; Florian Muller; Jing Wang; Bingliang Fang; Vali Papadimitrakopoulou; Ignacio I. Wistuba; Kunal Rai; Joseph R. Marszalek; P. Andrew Futreal

In the version of this article originally published, information regarding several funding sources was omitted from the Acknowledgements section. The following sentences should have been included: “This work was supported by the generous philanthropic contributions to The University of Texas MD Anderson Lung Cancer Moon Shots Program, the UT Lung SPORE 5 P50 CA07090, and the MD Anderson Cancer Center Support Grant P30CA01667. V.P is supported by R01CA155196-01A1 from the National Cancer Institute.” Also, reference 18 was incorrect. The original reference was: Kim, E. S. et al. The BATTLE trial: personalizing therapy for lung cancer. Cancer Discov. 1, 44–53 (2011). It should have been: Papadimitrakopoulou, V. et al. The BATTLE-2 study: a biomarker-integrated targeted therapy study in previously treated patients with advanced non–small-cell lung cancer. J Clin. Oncol. 34, 3638–3647 (2016). The errors have been corrected in the HTML and PDF versions of this article.


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 4242: Rhodamine esters as fluorescent tumor painting agents for glioblastoma

Yu-Hsi Lin; Steven W. Millward; Seth Gammon; Nikunj Satani; Naima Hammoudi; Joshua P. Gray; Lindsay E. Kelderhouse; Argentina Ornelas; Haley Schroeder; Florian Muller

Proceedings: AACR 107th Annual Meeting 2016; April 16-20, 2016; New Orleans, LA GBM remains one of the most difficult cancers to treat. Despite surgery, radiation and chemotherapy, tumors invariably recur. There is a strong correlation between extend of resection and subsequent time to recurrence and ultimate patient survival. A key challenge for the neurosurgeon is to minimize removal normal brain tissue whilst being as aggressive as possible with regards of removal of tumor tissue. Defining tumor boundary during surgery remains challenging. Fluorescent dyes such as 5- Aminolevulinic acid, have been studied as aids for tumor delineation but have failed to gain widespread acceptance in clinical practice. It has long been known that a substantial number of cancers, including Glioblastoma, exhibit a hyperpolarization of the plasma and mitochondrial membrane potential. This hyperpolarization is manifest by increased retention of lipophilic cationic dyes (Nernstian probes), such as the SPECT-CT probes 99Tc-Sestamibi and 99Tc-Tetrofosmin in both glioma cells in culture and primary tumors. We synthesized a series of ester derivatives of Rhodamine B with high fluorescence quantum yield in the red spectrum and a very favorable biological safety profile. Utilizing a series of intracranial xenografted glioblastoma pre-clinical mouse models, we show that a single I.V. injection of 1 mg/kg fluoroethylrhodamine ester (RhoFe) results in dramatic selective fluorescence in tumor but not surrounding normal brain tissue. This was observed even in tumors with minimal breakdown of the blood brain barrier, as determined by T1-contrast enhancing agents. RhoFe-tumor-selective fluorescence can remain visible up to 72 hours after injection. Similar results were obtained with Tetramethylrhodamine ester (TMRE) and rhodamine 123 (Rho123), but unlike RhoFe, TMRE proved highly toxic, while Rho123 fluorescence was both weaker and in the green spectrum, accompanied by higher endogenous auto-fluorescence. While well tolerated in vivo, RhoFe shows strong photoxicity in cell culture, suggesting potential as a photodynamic therapy agent. Taken together, these data suggest that RhoFe may be a promising tumor painting agent, with potential utility in fluorescence guided surgery as well as photodynamic therapy. Citation Format: Yu-Hsi Lin, Steven Millward, Seth Gammon, Nikunj Satani, Naima Hammoudi, Joshua Philip Gray, Lindsay E. Kelderhouse, Argentina Ornelas, Haley Schroeder, Florian L. Muller. Rhodamine esters as fluorescent tumor painting agents for glioblastoma. [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 4242.


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.


Neuro-oncology | 2016

TMIC-14. TUMOR EVOLUTION OF GLIOMA INTRINSIC GENE EXPRESSION SUBTYPE ASSOCIATES WITH IMMUNOLOGICAL CHANGES IN THE MICROENVIRONMENT

Qianghu Wang; Xin Hu; Florian Muller; Hoon Kim; Massimo Squatrito; Tom Mikkelsen; Lisa Scarpace; Floris P. Barthel; Yu-Hsi Lin; Nikunj Satani; Emmanuel Martinez-Ledesma; Edward F. Chang; Adriana Olar; Baoli Hu; Ana C. deCarvalho; Eskil Eskilsson; Siyuan Zheng; Amy B. Heimberger; Erik P. Sulman; Do-Hyun Nam; Roel G.W. Verhaak


Cancer Research | 2018

Abstract 2831: Collateral lethality: A new target for personalized oncology

Yu-Hsi Lin; Naima Hammoudi; Nikunj Satani; Jeffrey J. Ackroyd; Sunada Khadka; Dimitra K. Georgiou; Joe Marszalek; Yuting Sun; Marina Protopopova; Maria Emilia Di Francesco; Barbara Czako; Alan Y. Wang; Ronald A. DePinho; Florian Muller


Neuro-oncology | 2017

TMIC-22. DECIPHERING GLIOMA INTRINSIC TRANSCRIPTIONAL SUBTYPES IDENTIFIES TUMOR EVOLUTION ASSOCIATES WITH CHANGES IN IMMUNE-MICROENVIRONMENT

Qianghu Wang; Baoli Hu; Xin Hu; Massimo Squatrito; Lisa Scarpace; Ana C. deCarvalho; Sali Lyu; Pengping Li; Yan Li; Floris P. Barthel; Hee Jin Cho; Yu-Hsi Lin; Nikuj Satani; Emmanuel Martinez-Ledesma; Siyuan Zheng; Edward F. Chang; Adriana Olar; Zhengdao Lan; Gaetano Finocchiaro; Joanna J. Phillips; Mitchel S. Berger; Konrad Gabrusiewicz; Guocan Wang; Eskil Eskilsson; Jian Hu; Tom Mikkelsen; Ronald A. DePinho; Florian Muller; Amy B. Heimberger; Erik P. Sulman


European Journal of Cancer | 2016

POMHEX, a cell-permeable enolase inhibitor for collateral lethality targeting of ENO1-deleted glioblastoma

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

Collaboration


Dive into the Yu-Hsi Lin'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

Naima Hammoudi

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

Barbara Czako

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

Joe Marszalek

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

Paul G. Leonard

University of Texas MD Anderson Cancer Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge