30 results on '"Sunada Khadka"'
Search Results
2. Impaired anaplerosis is a major contributor to glycolysis inhibitor toxicity in glioma
- Author
-
Sunada Khadka, Kenisha Arthur, Yasaman Barekatain, Eliot Behr, Mykia Washington, Jeffrey Ackroyd, Kaitlyn Crowley, Pornpa Suriyamongkol, Yu-Hsi Lin, Cong-Dat Pham, Rafal Zielinski, Marissa Trujillo, James Galligan, Dimitra K. Georgiou, John Asara, and Florian Muller
- Subjects
Cancer metabolism ,Anaplerosis ,Collateral lethality ,Glycolysis ,Glutaminolysis ,Enolase inhibitor ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Background Reprogramming of metabolic pathways is crucial to satisfy the bioenergetic and biosynthetic demands and maintain the redox status of rapidly proliferating cancer cells. In tumors, the tricarboxylic acid (TCA) cycle generates biosynthetic intermediates and must be replenished (anaplerosis), mainly from pyruvate and glutamine. We recently described a novel enolase inhibitor, HEX, and its pro-drug POMHEX. Since glycolysis inhibition would deprive the cell of a key source of pyruvate, we hypothesized that enolase inhibitors might inhibit anaplerosis and synergize with other inhibitors of anaplerosis, such as the glutaminase inhibitor, CB-839. Methods We analyzed polar metabolites in sensitive (ENO1-deleted) and resistant (ENO1-WT) glioma cells treated with enolase and glutaminase inhibitors. We investigated whether sensitivity to enolase inhibitors could be attenuated by exogenous anaplerotic metabolites. We also determined the synergy between enolase inhibitors and the glutaminase inhibitor CB-839 in glioma cells in vitro and in vivo in both intracranial and subcutaneous tumor models. Results Metabolomic profiling of ENO1-deleted glioma cells treated with the enolase inhibitor revealed a profound decrease in the TCA cycle metabolites with the toxicity reversible upon exogenous supplementation of supraphysiological levels of anaplerotic substrates, including pyruvate. ENO1-deleted cells also exhibited selective sensitivity to the glutaminase inhibitor CB-839, in a manner rescuable by supplementation of anaplerotic substrates or plasma-like media PlasmaxTM. In vitro, the interaction of these two drugs yielded a strong synergistic interaction but the antineoplastic effects of CB-839 as a single agent in ENO1-deleted xenograft tumors in vivo were modest in both intracranial orthotopic tumors, where the limited efficacy could be attributed to the blood-brain barrier (BBB), and subcutaneous xenografts, where BBB penetration is not an issue. This contrasts with the enolase inhibitor HEX, which, despite its negative charge, achieved antineoplastic effects in both intracranial and subcutaneous tumors. Conclusion Together, these data suggest that at least for ENO1-deleted gliomas, tumors in vivo—unlike cells in culture—show limited dependence on glutaminolysis and instead primarily depend on glycolysis for anaplerosis. Our findings reinforce the previously reported metabolic idiosyncrasies of in vitro culture and suggest that cell culture media nutrient composition more faithful to the in vivo environment will more accurately predict in vivo efficacy of metabolism targeting drugs.
- Published
- 2021
- Full Text
- View/download PDF
3. Generation and Validation of an Anti-Human PANK3 Mouse Monoclonal Antibody
- Author
-
Sunada Khadka, Long Vien, Paul Leonard, Laura Bover, and Florian Muller
- Subjects
pantothenate kinases ,PANK3 ,monoclonal antibody ,CoA ,Microbiology ,QR1-502 - Abstract
Coenzyme A (CoA) is an essential co-factor at the intersection of diverse metabolic pathways. Cellular CoA biosynthesis is regulated at the first committed step—phosphorylation of pantothenic acid—catalyzed by pantothenate kinases (PANK1,2,3 in humans, PANK3 being the most highly expressed). Despite the critical importance of CoA in metabolism, the differential roles of PANK isoforms remain poorly understood. Our investigations of PANK proteins as potential precision oncology collateral lethality targets (PANK1 is co-deleted as part of the PTEN locus in some highly aggressive cancers) were severely hindered by a dearth of commercial antibodies that can reliably detect endogenous PANK3 protein. While we successfully validated commercial antibodies for PANK1 and PANK2 using CRISPR knockout cell lines, we found no commercial antibody that could detect endogenous PANK3. We therefore set out to generate a mouse monoclonal antibody against human PANK3 protein. We demonstrate that a clone (Clone MDA-299-62A) can reliably detect endogenous PANK3 protein in cancer cell lines, with band-specificity confirmed by CRISPR PANK3 knockout and knockdown cell lines. Sub-cellular fractionation shows that PANK3 is overwhelmingly cytosolic and expressed broadly across cancer cell lines. PANK3 monoclonal antibody MDA-299-62A should prove a valuable tool for researchers investigating this understudied family of metabolic enzymes in health and disease.
- Published
- 2022
- Full Text
- View/download PDF
4. Prodrugs of a 1-Hydroxy-2-oxopiperidin-3-yl Phosphonate Enolase Inhibitor for the Treatment of ENO1-Deleted Cancers
- Author
-
Victoria C. Yan, Cong-Dat Pham, Elliot S. Ballato, Kristine L. Yang, Kenisha Arthur, Sunada Khadka, Yasaman Barekatain, Prakriti Shrestha, Theresa Tran, Anton H. Poral, Mykia Washington, Sudhir Raghavan, Barbara Czako, Federica Pisaneschi, Yu-Hsi Lin, Nikunj Satani, Naima Hammoudi, Jeffrey J. Ackroyd, Dimitra K. Georgiou, Steven W. Millward, and Florian L. Muller
- Subjects
Drug Discovery ,Molecular Medicine - Published
- 2022
5. Quantification of Phosphonate Drugs by 1H–31P HSQC Shows That Rats Are Better Models of Primate Drug Exposure than Mice
- Author
-
Yasaman Barekatain, Sunada Khadka, Kristen Harris, Jorge Delacerda, Victoria C. Yan, Ko-Chien Chen, Cong-Dat Pham, Md. Nasir Uddin, Rony Avritcher, Eugene J. Eisenberg, Raghu Kalluri, Steven W. Millward, and Florian L. Muller
- Subjects
Analytical Chemistry - Abstract
The phosphonate group is a key pharmacophore in many anti-viral, anti-microbial, and anti-neoplastic drugs. Due to its high polarity and short retention time, detecting and quantifying such phosphonate-containing drugs with LC/MS-based methods is challenging and requires derivatization with hazardous reagents. Given the emerging importance of phosphonate-containing drugs, developing a practical, accessible, and safe method for their quantitation in pharmacokinetics (PK) studies is desirable. NMR-based methods are often employed in drug discovery but are seldom used for compound quantitation in PK studies. Here, we show that proton-phosphorous (1H-31P) heteronuclear single quantum correlation (HSQC) NMR allows for quantitation of the phosphonate-containing enolase inhibitor HEX in plasma and tissue at micromolar concentrations. Although mice were shown to rapidly clear HEX from circulation (over 95% in 1H-31P HSQC method to quantify phosphonate-containing drugs in complex biological samples and illustrates an important limitation of mice as preclinical model species for phosphonate-containing drugs.
- Published
- 2022
6. Anaplerotic nutrient stress drives synergy of angiogenesis inhibitors with therapeutics targeting tumor metabolism
- Author
-
Sunada Khadka, Yu-Hsi Lin, Jeffrey Ackroyd, Yi-An Chen, Yanghui Sheng, Wubin Qian, Sheng Guo, Yining Chen, Eliot Behr, Yasaman Barekatain, Md. Nasir Uddin, Kenisha Arthur, Victoria Yan, Wen-Hao Hsu, Edward Chang, Anton Poral, Theresa Tran, Surendra Chaurasia, Dimitra K. Georgiou, John M. Asara, Floris P. Barthel, Steve W. Millward, Ronald A. DePinho, and Florian L. Muller
- Abstract
Tumor angiogenesis is a cancer hallmark, and its therapeutic inhibition has provided meaningful, albeit limited, clinical benefit. While anti-angiogenesis inhibitors deprive the tumor of oxygen and essential nutrients, cancer cells activate metabolic adaptations to diminish therapeutic response. Despite these adaptations, angiogenesis inhibition incurs extensive metabolic stress, prompting us to consider such metabolic stress as aninduced vulnerabilityto therapies targeting cancer metabolism. Metabolomic profiling of angiogenesis-inhibited intracranial xenografts showed universal decrease in tricarboxylic acid cycle intermediates, corroborating a state of anaplerotic nutrient deficit or stress. Accordingly, we show strong synergy between angiogenesis inhibitors (Avastin, Tivozanib) and inhibitors of glycolysis or oxidative phosphorylation through exacerbation of anaplerotic nutrient stress in intracranial orthotopic xenografted gliomas. Our findings were recapitulated in GBM xenografts that do not have genetically predisposed metabolic vulnerabilities at baseline. Thus, our findings cement the central importance of the tricarboxylic acid cycle as the nexus of metabolic vulnerabilities and suggest clinical path hypothesis combining angiogenesis inhibitors with pharmacological cancer interventions targeting tumor metabolism for GBM tumors.
- Published
- 2023
7. Supplementary Table S5 from Targeting YAP-Dependent MDSC Infiltration Impairs Tumor Progression
- Author
-
Ronald A. DePinho, Y. Alan Wang, Lynda Chin, Mark J. McArthur, Christopher J. Logothetis, Patricia Troncoso, Qing Chang, Liren Li, Yanxia Shi, Zhihu Ding, Xiaolu Pan, Wantong Yao, Eun-Jung Jin, Baoli Hu, Pingping Hou, Sunada Khadka, Xiaoying Shang, Di Zhao, Tim Heffernan, Trang N. Tieu, Vandhana Ramamoorthy, Zhenglin Guo, Neelay Bhaskar Patel, Chang-Jiun Wu, Avnish Kapoor, Elsa M. Li-Ning-Tapia, Jianhua Zhang, Sujun Hua, Ramakrishna Konaparthi, Kun Zhao, Zhuangna Fang, Shan Jiang, Chia Chin Wu, Pingna Deng, Prasenjit Dey, Xin Lu, and Guocan Wang
- Abstract
Overlapped genes between Genes upregulated in Ptenpc-/-Smad4pc-/- tumors as compared to Ptenpc-/- tumors ({greater than or equal to}2 fold) and genes upregulated in GFP+ tumors cells from Ptenpc-/-Smad4pc-/- mice as compared to Tomato+ cells ({greater than or equal to}4 fold).
- Published
- 2023
8. Supplementary Methods, Figure Legends, Figures S1 - S7 from Targeting YAP-Dependent MDSC Infiltration Impairs Tumor Progression
- Author
-
Ronald A. DePinho, Y. Alan Wang, Lynda Chin, Mark J. McArthur, Christopher J. Logothetis, Patricia Troncoso, Qing Chang, Liren Li, Yanxia Shi, Zhihu Ding, Xiaolu Pan, Wantong Yao, Eun-Jung Jin, Baoli Hu, Pingping Hou, Sunada Khadka, Xiaoying Shang, Di Zhao, Tim Heffernan, Trang N. Tieu, Vandhana Ramamoorthy, Zhenglin Guo, Neelay Bhaskar Patel, Chang-Jiun Wu, Avnish Kapoor, Elsa M. Li-Ning-Tapia, Jianhua Zhang, Sujun Hua, Ramakrishna Konaparthi, Kun Zhao, Zhuangna Fang, Shan Jiang, Chia Chin Wu, Pingna Deng, Prasenjit Dey, Xin Lu, and Guocan Wang
- Abstract
Supplementary Figure S1. CyTOF analysis of biological samples from Ptenpc-/-Smad4pc-/- mice (Related to Figure 2). Supplementary Figure S2. Strategy used for MDSCs Isolation (Related to Figure 3). Supplementary Figure S3. Treatment scheme for Gr-1 antibody, peptibody, and Cxcr2 inhibitor SB225002. Supplementary Figure S4. IHC staining of Ki67, CD45, Vimentin, Smooth muscle actin (SMA) and Trichrome staining of mouse prostate tissues treated with IgG control or Gr1 antibody. Supplementary Figure S5. The top 10 differentially expressed genes in Ptenpc-/-Smad4pc-/- tumors as compared to Ptenpc-/- tumors, identified by microarray analysis (n=5). Figure S6. Top 10 activated oncogenic signatures identified by GSEA analysis in Ptenpc-/- Smad4pc-/- tumors as compared to Ptenpc-/- tumors (n=5). Figure S7. Clustering of primary prostate tumors from Wallace et al into MDSC-high and MDSC-low subtypes.
- Published
- 2023
9. Genetic depletion of de novo coenzyme A biosynthesis exacerbates puromycin toxicity
- Author
-
Sunada Khadka, Adam Chatoff, Nathaniel W. Snyder, Ronald DePinho, and Florian Muller
- Abstract
Puromycin is an amino nucleoside that inhibits protein synthesis by interrupting elongation of nascent peptide chains. It is a commonly used selection antibiotic in molecular biology research via engineered expression of a puromycin resistance transgene. The enzyme puromycin acetyl transferase (pac) or PuroR inactivates puromycin by N-acetylating its reactive amino group. Puromycin acetylation by pac requires the central metabolite and acetyl group donor acetyl-CoA as a substrate. We found that puromycin treatment exacerbates sensitivity of cancer cells to knockdown of pantothenate kinases, the proteins that catalyze the rate-limiting step of de novo coenzyme A production in cells. Mechanistically, we found that ablation of PANKs together with puromycin depletes acetyl-CoA levels, in a manner modulated by the dose of puromycin. Our findings provide a note of caution and context in the use of puromycin for metabolism research in that interference with the major acyl donor used for inactivating biotransformation may exacerbate toxicity under selection. Broadly, our findings also invite studies to explore how targeting CoA and acetyl-CoA synthesis may be exploited to enhance cytotoxic effects of cancer drugs that undergo acetylation.
- Published
- 2022
10. Homozygous MTAP deletion in primary human glioblastoma is not associated with elevation of methylthioadenosine
- Author
-
Kenisha Arthur, John M. Asara, Anton H. Poral, Nikunj Satani, Yu Hsi Lin, Raghu Kalluri, Florian L. Muller, Sunada Khadka, Dimitra K. Georgiou, Theresa Tran, Jeffrey J. Ackroyd, Jason T. Huse, Victoria C. Yan, Yasaman Barekatain, Eliot Itzkow Behr, Lin Wang, Ana C. deCarvalho, Ko Chien Chen, Elliot S. Ballato, John de Groot, and Roel G.W. Verhaak
- Subjects
0301 basic medicine ,Protein-Arginine N-Methyltransferases ,Science ,General Physics and Astronomy ,Purine nucleoside phosphorylase ,Antineoplastic Agents ,Biology ,Article ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,0302 clinical medicine ,Stroma ,In vivo ,Cell Line, Tumor ,medicine ,Frozen Sections ,Humans ,Metabolomics ,Molecular Targeted Therapy ,Precision Medicine ,Sequence Deletion ,Thionucleosides ,Multidisciplinary ,Deoxyadenosines ,Brain Neoplasms ,Protein arginine methyltransferase 5 ,Homozygote ,Brain ,Cancer ,Methionine Adenosyltransferase ,General Chemistry ,medicine.disease ,Xenograft Model Antitumor Assays ,Cancer metabolism ,In vitro ,CNS cancer ,030104 developmental biology ,Purine-Nucleoside Phosphorylase ,Cell culture ,Culture Media, Conditioned ,030220 oncology & carcinogenesis ,Cancer cell ,Cancer research ,Female ,Glioblastoma - Abstract
Homozygous deletion of methylthioadenosine phosphorylase (MTAP) in cancers such as glioblastoma represents a potentially targetable vulnerability. Homozygous MTAP-deleted cell lines in culture show elevation of MTAP’s substrate metabolite, methylthioadenosine (MTA). High levels of MTA inhibit protein arginine methyltransferase 5 (PRMT5), which sensitizes MTAP-deleted cells to PRMT5 and methionine adenosyltransferase 2A (MAT2A) inhibition. While this concept has been extensively corroborated in vitro, the clinical relevance relies on exhibiting significant MTA accumulation in human glioblastoma. In this work, using comprehensive metabolomic profiling, we show that MTA secreted by MTAP-deleted cells in vitro results in high levels of extracellular MTA. We further demonstrate that homozygous MTAP-deleted primary glioblastoma tumors do not significantly accumulate MTA in vivo due to metabolism of MTA by MTAP-expressing stroma. These findings highlight metabolic discrepancies between in vitro models and primary human tumors that must be considered when developing strategies for precision therapies targeting glioblastoma with homozygous MTAP deletion., The metabolite methylthioadenosine (MTA) inhibits PRMT5. Therefore, MTA accumulation due to MTA phosphorylase (MTAP) deletion has been proposed as a vulnerability for PRMT5-targeted therapy in cancer. Here, the authors show that MTA does not accumulate in MTAP-deficient cancer cells but is secreted and metabolized by MTAP-intact cells in the tumour microenvironment.
- Published
- 2021
11. Abstract 3097: Prodrugs of a 1-hydroxy-2-oxopiperidin-3-yl phosphonate enolase inhibitor for the treatment of ENO1-deleted cancers
- Author
-
Victoria C. Yan, Cong-Dat Pham, Elliot S. Ballato, Kristine L. Yang, Sunada Khadka, Yasaman Barektain, Prakriti Shrestha, Theresa Tran, Anton H. Poral, Mykia Washington, Sudhir Raghavan, Barbara Czako, Federica Pisaneschi, Yu-Hsi Lin, Nikunj Satani, Naima Hammoudi, Jeffrey J. Ackroyd, Dimitra K. Georgiou, Steven W. Millward, and Florian L. Muller
- Subjects
Cancer Research ,Oncology - Abstract
Cancers harboring homozygous deletion of the glycolytic enzyme enolase 1 (ENO1) are selectively vulnerable to inhibition of the paralogous isoform, enolase 2 (ENO2). We previously identified and characterized a competitive, small molecule phosphonate inhibitor of ENO2, 1-hydroxy-2-oxopiperidin-3-yl phosphonate (HEX), and its lipophilic bis-ester prodrug (POMHEX) in an ENO1-deleted intracranial orthotopic xenograft model of glioblastoma. Treatment with either HEX (150 mg/kg IV/IP) or POMHEX (20 mg/kg IV/IP) yielded tumor regression even after drug discontinuation. However, due to the poor pharmacokinetics of esterase-labile POMHEX, we synthesized a library of novel phosphonate prodrugs with distinct mechanisms of bioactivation and assessed their potency in D423 (ENO1-/-) cells. By conducting a prodrug structure activity relationship (SAR) study, we found that phosphonoamidate esters were efficiently bioactivated in ENO1-deleted glioma cells, while canonical McGuigan (ProTide) prodrugs were not. Other strategies, including salicylic alcohol (cycloSal) and lipid prodrugs of HEX, exhibited low micromolar IC50 values in ENO1-deleted glioma cells and improved stability in human serum over POMHEX. En route, we developed a novel class of aliphatic amine/ester prodrugs that can be broadly applied to efficiently deliver phosph(on)ate pharmacophores in cells. The activity of select prodrugs was also probed using the NCI-60 cell line screen, supporting its use to examine the relationship between prodrugs and cell line-dependent bioactivation. In sum, we have developed a novel class of phosph(on)ate prodrugs that is efficiently bioactivated in cells in vitro. Our prodrug SAR study disputes the common notion that ProTides are universally advantageous promoieties on phosph(on)ate pharmacophores and we provide mechanistic rationale for this observation with HEX. Finally, we show that the cycloSal prodrug yields efficient intracellular delivery of HEX in vitro, with a mechanism of bioactivation consistent with the GBM microenvironment, making this promoiety promising for further evaluation in vivo. Citation Format: Victoria C. Yan, Cong-Dat Pham, Elliot S. Ballato, Kristine L. Yang, Sunada Khadka, Yasaman Barektain, Prakriti Shrestha, Theresa Tran, Anton H. Poral, Mykia Washington, Sudhir Raghavan, Barbara Czako, Federica Pisaneschi, Yu-Hsi Lin, Nikunj Satani, Naima Hammoudi, Jeffrey J. Ackroyd, Dimitra K. Georgiou, Steven W. Millward, Florian L. Muller. Prodrugs of a 1-hydroxy-2-oxopiperidin-3-yl phosphonate enolase inhibitor for the treatment of ENO1-deleted cancers [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2023; Part 1 (Regular and Invited Abstracts); 2023 Apr 14-19; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2023;83(7_Suppl):Abstract nr 3097.
- Published
- 2023
12. An enolase inhibitor for the targeted treatment of ENO1-deleted cancers
- Author
-
Xiaobo Wang, Jeffrey J. Ackroyd, Yongying Jiang, Florian L. Muller, Yuting Sun, Federica Pisaneschi, Theresa Tran, Nikunj Satani, Cong-Dat Pham, Waldemar Priebe, Barbara Czako, Qi Wu, Paul G. Leonard, Ronald A. DePinho, Joseph R. Marszalek, John M. Asara, Pijus K. Mandal, Yasaman Barekatain, Susana Castro Pando, William G. Bornmann, Rafal Zielinski, Naima Hammoudi, Sunada Khadka, David Maxwell, Kenisha Arthur, Yu Hsi Lin, Quanyu Xu, Dimitra K. Georgiou, Victoria C. Yan, Zhijun Kang, and Zhenghong Peng
- Subjects
Male ,Endocrinology, Diabetes and Metabolism ,Enolase ,Antineoplastic Agents ,Mice, SCID ,Article ,Mice ,Structure-Activity Relationship ,Glycolysis Inhibition ,In vivo ,Cell Line, Tumor ,Neoplasms ,Physiology (medical) ,Glioma ,Biomarkers, Tumor ,Internal Medicine ,medicine ,Animals ,Humans ,Glycolysis ,Enzyme Inhibitors ,Precision Medicine ,Sequence Deletion ,chemistry.chemical_classification ,business.industry ,Tumor Suppressor Proteins ,Cancer ,Cell Biology ,medicine.disease ,Xenograft Model Antitumor Assays ,DNA-Binding Proteins ,Macaca fascicularis ,Enzyme ,chemistry ,Cell culture ,Phosphopyruvate Hydratase ,Cancer research ,Female ,business - Abstract
Inhibiting glycolysis remains an aspirational approach for the treatment of cancer. We have previously identified a subset of cancers harbouring homozygous deletion of the glycolytic enzyme enolase (ENO1) that have exceptional sensitivity to inhibition of its redundant paralogue, ENO2, through a therapeutic strategy known as collateral lethality. Here, we show that a small-molecule enolase inhibitor, POMHEX, can selectively kill ENO1-deleted glioma cells at low-nanomolar concentrations and eradicate intracranial orthotopic ENO1-deleted tumours in mice at doses well-tolerated in non-human primates. Our data provide an in vivo proof of principle of the power of collateral lethality in precision oncology and demonstrate the utility of POMHEX for glycolysis inhibition with potential use across a range of therapeutic settings.
- Published
- 2020
13. Impaired anaplerosis is a major contributor to glycolysis inhibitor toxicity in glioma
- Author
-
Marissa N. Trujillo, Kenisha Arthur, John M. Asara, Cong-Dat Pham, Eliot Itzkow Behr, Sunada Khadka, Yu-Hsi Lin, Mykia Washington, Kaitlyn Crowley, Rafal Zielinski, Pornpa Suriyamongkol, Jeffrey J. Ackroyd, Yasaman Barekatain, Florian L. Muller, James J. Galligan, and Dimitra K. Georgiou
- Subjects
Glutaminolysis ,Glutaminase ,Chemistry ,Research ,Enolase ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Collateral lethality ,Cancer metabolism ,POMHEX ,Citric acid cycle ,Psychiatry and Mental health ,Glycolysis Inhibition ,In vivo ,Cancer cell ,Cancer research ,Glycolysis ,CB-839 ,Enolase inhibitor ,RC254-282 ,Anaplerosis - Abstract
Background Reprogramming of metabolic pathways is crucial to satisfy the bioenergetic and biosynthetic demands and maintain the redox status of rapidly proliferating cancer cells. In tumors, the tricarboxylic acid (TCA) cycle generates biosynthetic intermediates and must be replenished (anaplerosis), mainly from pyruvate and glutamine. We recently described a novel enolase inhibitor, HEX, and its pro-drug POMHEX. Since glycolysis inhibition would deprive the cell of a key source of pyruvate, we hypothesized that enolase inhibitors might inhibit anaplerosis and synergize with other inhibitors of anaplerosis, such as the glutaminase inhibitor, CB-839. Methods We analyzed polar metabolites in sensitive (ENO1-deleted) and resistant (ENO1-WT) glioma cells treated with enolase and glutaminase inhibitors. We investigated whether sensitivity to enolase inhibitors could be attenuated by exogenous anaplerotic metabolites. We also determined the synergy between enolase inhibitors and the glutaminase inhibitor CB-839 in glioma cells in vitro and in vivo in both intracranial and subcutaneous tumor models. Results Metabolomic profiling of ENO1-deleted glioma cells treated with the enolase inhibitor revealed a profound decrease in the TCA cycle metabolites with the toxicity reversible upon exogenous supplementation of supraphysiological levels of anaplerotic substrates, including pyruvate. ENO1-deleted cells also exhibited selective sensitivity to the glutaminase inhibitor CB-839, in a manner rescuable by supplementation of anaplerotic substrates or plasma-like media PlasmaxTM. In vitro, the interaction of these two drugs yielded a strong synergistic interaction but the antineoplastic effects of CB-839 as a single agent in ENO1-deleted xenograft tumors in vivo were modest in both intracranial orthotopic tumors, where the limited efficacy could be attributed to the blood-brain barrier (BBB), and subcutaneous xenografts, where BBB penetration is not an issue. This contrasts with the enolase inhibitor HEX, which, despite its negative charge, achieved antineoplastic effects in both intracranial and subcutaneous tumors. Conclusion Together, these data suggest that at least for ENO1-deleted gliomas, tumors in vivo—unlike cells in culture—show limited dependence on glutaminolysis and instead primarily depend on glycolysis for anaplerosis. Our findings reinforce the previously reported metabolic idiosyncrasies of in vitro culture and suggest that cell culture media nutrient composition more faithful to the in vivo environment will more accurately predict in vivo efficacy of metabolism targeting drugs.
- Published
- 2021
14. Abstract 3208: Angiogenesis inhibitors strongly synergize with therapeutics targeting tumor metabolism
- Author
-
Sunada Khadka, Yu-Hsi Lin, Jeffery Ackroyd, Kenisha Arthur, Yasaman Barekatain, Anton Poral, Theresa Tran, Eliot Behr, Yining Chen, Dimitra Georgiou, Ronald Depinho, and Florian Muller
- Subjects
Cancer Research ,Oncology - Abstract
Angiogenesis inhibition has become a mainstay of oncology despite having fallen short of its early promise. As originally envisioned, angiogenesis inhibition would cut off the blood supply, deprive tumor cells of key nutrients, leading to their demise. In practice, while there is evidence that tumors under angiogenesis treatment do in fact exhibit some degree of metabolic stress, this is stress is not sufficient to induce significant cancer cell death. We posit that the full potential of angiogenesis inhibition can be realized by the combination of angiogenesis inhibition with emerging tumor metabolism targeting therapies. Because tumors under angiogenesis inhibition are already in a state of nutrient stress, the effects of metabolically targeted therapies such as amino acid depletion (e.g. asparginase, methionine restriction), inhibitors of stress adaption (AMPK and GCN2 inhibitors) or energy metabolism (e.g. IACS-010759, Metformin, POMHEX) stand to dramatically increase in potency whilst remaining selective for (angiogenic) tumor versus (non-angiogenic) normal tissue. Here, we provide proof-of-principal for this thesis. First, we performed metabolomic profiling of angiogenesis-inhibited tumors, which corroborates a state of nutrient stress in angiogenesis-inhibited tumors. Second, we demonstrate dramatic anti-neoplastic synergy (effectively curing of xenografted tumor-bearing mice, irrespective of initial tumor size), without enhanced adverse toxicities, between the anti-VEGFA antibody Avastin and enolase inhibitor HEX. The same results were recapitulated with the VEGFR inhibitor Tivozanib and HEX and the enolase inhibitor could be substituted with the Oxphos inhibitor IACS-010759, with similar effects. The synergy was observed in a broad range of tumor types, even those without clear genetic susceptibilities. Together, these results suggest that angiogenesis inhibitors synergize broadly with cancer therapies targeting metabolism, allowing the realization of the full potential of these previously disappointing drugs. Our results warrant systematic combination clinical trials between angiogenesis inhibitors and established, as well as emerging anti-metabolic cancer therapies. Citation Format: Sunada Khadka, Yu-Hsi Lin, Jeffery Ackroyd, Kenisha Arthur, Yasaman Barekatain, Anton Poral, Theresa Tran, Eliot Behr, Yining Chen, Dimitra Georgiou, Ronald Depinho, Florian Muller. Angiogenesis inhibitors strongly synergize with therapeutics targeting tumor metabolism [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2022; 2022 Apr 8-13. Philadelphia (PA): AACR; Cancer Res 2022;82(12_Suppl):Abstract nr 3208.
- Published
- 2022
15. Abstract 2395: Homozygous MTAP deletion in primary human glioblastoma is not associated with elevation of methylthioadenosine
- Author
-
Yasaman Barekatain, Jeffrey Ackroyd, Victoria Yan, Sunada Khadka, Ko-Chien Chen, Raghu Kalluri, John de Groot, Jason Huse, and Florian muller
- Subjects
Cancer Research ,Oncology - Abstract
Homozygous deletion of the CDK2NA locus frequently results in the co-deletion of methylthioadenosine phosphorylase (MTAP) in many fatal cancers such as glioblastoma multiform (GBM). In cell culture, cell lines with MTAP-deletions show elevations of its substrate metabolite, methylthioadenosine (MTA). High levels of MTA inhibit PRMT5, which sensitizes MTAP-deleted cell lines to PRMT5 and MAT2A inhibition. While extensively corroborated in vitro, the clinical efficacy of these strategies ultimately relies on equally significant accumulations of MTA in human tumors. In this work, using comprehensive metabolomic profiling, we show that MTA is primarily secreted, resulting in exceedingly high levels of extracellular MTA in vitro. We further show that primary human glioblastoma tumors minimally accumulate MTA in vivo, which is likely explained by the metabolism of MTA by MTAP-competent stromal cells. Together, these findings highlight the metabolic discrepancies between in vitro models and primary human tumors and should thus be carefully considered in the development of the precision therapies targeting MTAPhomozygous deleted GBM. Citation Format: Yasaman Barekatain, Jeffrey Ackroyd, Victoria Yan, Sunada Khadka, Ko-Chien Chen, Raghu Kalluri, John de Groot, Jason Huse, Florian muller. Homozygous MTAP deletion in primary human glioblastoma is not associated with elevation of methylthioadenosine [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2022; 2022 Apr 8-13. Philadelphia (PA): AACR; Cancer Res 2022;82(12_Suppl):Abstract nr 2395.
- Published
- 2022
16. Pharmacokinetics of Orally Administered GS-441524 in Dogs
- Author
-
Victoria C. Yan, Cong-Dat Pham, Matthew J. Yan, Alexander J. Yan, Sunada Khadka, Kenisha Arthur, Jeffrey J. Ackroyd, Dimitra K. Georgiou, Laura E. Roon, Lane R. Bushman, Peter L. Anderson, Chun Li, and Florian L. Muller
- Subjects
Coronavirus disease 2019 (COVID-19) ,Pharmacokinetics ,business.industry ,Pharmacodynamics ,Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) ,Medicine ,Clinical efficacy ,Pharmacology ,business ,Beagle ,Nucleoside ,Article ,Bioavailability - Abstract
Despite being FDA-approved for COVID-19, the clinical efficacy of remdesivir (Veklury®) remains contentious. We previously pointed out pharmacokinetic, pharmacodynamic and toxicology reasons for why its parent nucleoside GS-441524, is better suited for COVID-19 treatment. Here, we assess the oral bioavailability of GS-441524 in beagle dogs and show that plasma concentrations ∼24-fold higher than the EC50 against SARS-CoV-2 are easily and safely sustained. These data support translation of GS-441524 as an oral agent for COVID-19.
- Published
- 2021
17. Impaired Anaplerosis Is a Major Contributor to Glycolysis Inhibitor Toxicity in Glioma
- Author
-
Sunada Khadka, Kenisha Arthur, Mykia Washington, Yasaman Barekatain, Jeff Ackroyd, Eliot Behr, Pornpa Suriyamongkol, Yu-Hsi Lin, Kaitlyn Crowley, Cong-Dat Pham, Dimitra K. Georgiou, John Asara, and Florian Muller
- Abstract
Reprogramming of metabolic pathways is crucial to satisfy the bioenergetic and biosynthetic demands and maintain the redox status of rapidly proliferating cancer cells. In tumors, the tricarboxylic acid (TCA) cycle generates biosynthetic intermediates by oxidation of anaplerotic substrates, such as glucose-derived pyruvate and glutamine20 derived glutamate. We have previously documented that a subset of tumors with 1p36 homozygous deletion exhibit co-deletion of ENO1, in turn becoming extremely dependent on its redundant isoform ENO2 and sensitive to an overall enzymatic deficiency of enolase. Metabolomic profiling of ENO1-deleted glioma cells treated with an enolase inhibitor revealed a profound decrease in TCA cycle metabolites, which correlated with cell-line specific sensitivity to enolase inhibition, highlighting the importance of glycolysis derived pyruvate for anaplerosis. Correspondingly, the toxicity of the enolase inhibitor was significantly attenuated by exogenous supplementation of supraphysiological levels of anaplerotic substrates including pyruvate. These findings led us to hypothesize that cancer cells with ENO1 homozygous deletions treated with an enolase inhibitor might show exceptional sensitivity to inhibition of glutaminolysis because of reduced anaplerotic flow from glycolysis. We found that ENO1-deleted cells indeed exhibited selective sensitivity to the glutaminase inhibitor CB-839, and this sensitivity was also attenuated by exogenous supplementation of anaplerotic substrates including pyruvate. Despite these promising in vitro results, the antineoplastic effects of CB-839 as a single agent in ENO1-deleted xenograft tumors in vivo were modest in both intracranial orthotopic tumors, where the limited efficacy could be attributed to the blood brain barrier (BBB), and subcutaneous xenografts, where BBB penetration is not an issue. This contrasts with the enolase inhibitor HEX, which, despite its negative charge, achieved antineoplastic effects in both intracranial and subcutaneous tumors. Together, these data suggest that at least for 1p36-deleted gliomas, tumors in vivo—unlike cells in culture—show limited dependence on glutaminolysis and instead primarily depend on glycolysis for anaplerosis. Our findings reinforce the previously reported metabolic idiosyncrasies of the in vitro and in vivo environments as the potential reasons for the differential efficacy of metabolism targeted therapies in in vitro and in vivo systems.
- Published
- 2020
18. TAMI-62. ANGIOGENESIS INHIBITORS STRONGLY SYNERGIZE WITH THERAPEUTICS TARGETING TUMOR METABOLISM
- Author
-
Yasaman Barekatain, John M. Asara, Florian L. Muller, John de Groot, Theresa Tran, Sunada Khadka, Kenisha Arthur, Dimitra K. Georgiou, Jeffrey J. Ackroyd, Yu-Hsi Lin, Jason T. Huse, and Anton H. Poral
- Subjects
Cancer Research ,Oncology ,Angiogenesis ,business.industry ,Cancer research ,Medicine ,Tumor Microenvironment/Angiogenesis/Metabolism/Invasion ,Neurology (clinical) ,Metabolism ,business - Abstract
Angiogenesis inhibition has become a mainstay of oncology despite having fallen short of its early promise. As originally envisioned, angiogenesis inhibition would cut off the blood supply, deprive tumor cells of key nutrients, leading to their demise. In practice, while there is evidence that tumors under angiogenesis treatment do in fact exhibit some degree of metabolic stress, this is stress is not sufficient to induce significant cancer cell death. We posit that the full potential of angiogenesis inhibition can be realized by the combination of angiogenesis inhibition with emerging tumor metabolism targeting therapies. Because tumors under angiogenesis inhibition are already in a state of nutrient stress, the effects of metabolically targeted therapies such as amino acid depletion (e.g. asparginase, methionine restriction), inhibitors of stress adaption (AMPK and GCN2 inhibitors) or energy metabolism (e.g. IACS-010759, Metformin, POMHEX) stand to dramatically increase in potency whilst remaining selective for (angiogenic) tumor versus (non-angiogenic) normal tissue. Here, we provide proof-of-principal for this thesis. First, we performed metabolomic profiling of angiogenesis-inhibited tumors, which corroborates as state of nutrient stress in angiogenesis-inhibited tumors. Second, we demonstrate dramatic anti-neoplastic synergy (effectively curing of xenografted tumor-bearing mice, irrespective of initial tumor size), without enhanced adverse toxicities, between the OxPhos inhibitor IACS-010759 and the angiogenesis tyrosine kinase inhibitor, Tivozanib. The same results were recapitulated with the anti-VEGFA antibody, Avastin, and the OxPhos inhibitor could be substituted with the Enolase inhibitor HEX, with similar effects. The synergy was observed in a broad range of tumor types, even those without clear genetic susceptibilities. Together, these results suggest that Angiogenesis inhibitors synergize broadly with cancer therapies targeting metabolism, allowing the realization of the full potential of these previously disappointing drugs. Our results warrant systematic combination clinical trials between angiogenesis inhibitors and established, as well as emerging anti-metabolic cancer therapies.
- Published
- 2020
19. Bioreducible Phosphonoamidate Pro-drug Inhibitor of Enolase: Proof of Concept Study
- Author
-
Theresa Tran, Cong-Dat Pham, Anton H. Poral, Florian L. Muller, Elliot S. Ballato, Mykia Washington, Kenisha Arthur, Dimitra K. Georgiou, Prakriti Shrestha, Victoria C. Yan, Sunada Khadka, Kristine L. Yang, and Matthew J. Yan
- Subjects
Tumor hypoxia ,010405 organic chemistry ,Chemistry ,medicine.medical_treatment ,Organic Chemistry ,Enolase ,Context (language use) ,Hypoxia (medical) ,Prodrug ,01 natural sciences ,Biochemistry ,In vitro ,0104 chemical sciences ,Targeted therapy ,010404 medicinal & biomolecular chemistry ,Glycolysis Inhibition ,Drug Discovery ,medicine ,Cancer research ,medicine.symptom - Abstract
[Image: see text] Glycolysis inhibition remains aspirational in cancer therapy. We recently described a promising phosphonate inhibitor of enolase for cancers harboring homozygous deletions of ENO1. Here, we describe the application of a nitroheterocycle phosphonoamidate pro-drug pair to capitalize on tumor hypoxia. This bioreducible prodrug exhibits greater-than 2-fold potency under hypoxic conditions compared to normoxia and exhibits robust stability in biological fluids. Our work provides strong in vitro proof-of-concept for using bioreduction as a pro-drug delivery strategy in the context of enolase inhibition.
- Published
- 2020
20. Robust detection of oncometabolic aberrations by
- Author
-
Yasaman, Barekatain, Victoria C, Yan, Kenisha, Arthur, Jeffrey J, Ackroyd, Sunada, Khadka, John, De Groot, Jason T, Huse, and Florian L, Muller
- Subjects
Carbon Isotopes ,Mice, Inbred BALB C ,Magnetic Resonance Spectroscopy ,Phosphogluconate Dehydrogenase ,Diagnostic markers ,Gluconates ,Sensitivity and Specificity ,Xenograft Model Antitumor Assays ,Cancer metabolism ,Isocitrate Dehydrogenase ,Article ,Glutarates ,Cell Line, Tumor ,Mutation ,Biomarkers, Tumor ,Cancer genomics ,Animals ,Humans ,Metabolomics ,Female ,Glioblastoma - Abstract
Magnetic resonance (MR) spectroscopy has potential to non-invasively detect metabolites of diagnostic significance for precision oncology. Yet, many metabolites have similar chemical shifts, yielding highly convoluted 1H spectra of intact biological material and limiting diagnostic utility. Here, we show that hydrogen–carbon heteronuclear single quantum correlation (1H–13C HSQC) offers dramatic improvements in sensitivity compared to one-dimensional (1D) 13C NMR and significant signal deconvolution compared to 1D 1H spectra in intact biological settings. Using a standard NMR spectroscope with a cryoprobe but without specialized signal enhancing features such as magic angle spinning, metabolite extractions or 13C-isotopic enrichment, we obtain well-resolved 2D 1H–13C HSQC spectra in live cancer cells, in ex vivo freshly dissected xenografted tumors and resected primary tumors. This method can identify tumors with specific oncometabolite alterations such as IDH mutations by 2-hydroxyglutarate and PGD-deleted tumors by gluconate. Results suggest potential of 1H–13C HSQC as a non-invasive diagnostic in precision oncology., Barekatain et al. demonstrate that hydrogen–carbon heteronuclear single quantum correlation (HSQC) spectra, obtained using a standard NMR spectroscope, can detect tumours with specific oncometabolite alterations including IDH1 mutant glioblastoma, suggesting the feasibility of this method as a diagnostic tool.
- Published
- 2020
21. Robust detection of oncometabolic aberrations by 1H-13C heteronuclear single quantum correlation in live cells and intact tumors ex-vivo
- Author
-
Victoria C. Yan, Sunada Khadka, Yasaman Barekatain, John de Groot, Florian L. Muller, Kenisha Arthur, Jeffrey J. Ackroyd, and Jason T. Huse
- Subjects
0303 health sciences ,IDH1 ,Chemistry ,Metabolite ,Carbon-13 NMR ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Nuclear magnetic resonance ,Isocitrate dehydrogenase ,030220 oncology & carcinogenesis ,Cancer cell ,Magic angle spinning ,Heteronuclear single quantum coherence spectroscopy ,Ex vivo ,030304 developmental biology - Abstract
Extensive efforts have been made to use non-invasive 1H magnetic resonance (MR) spectroscopy to quantify metabolites that are diagnostic of specific disease states. Within the realm of precision oncology, these efforts have largely centered on quantifying 2-hydroxyglutarate (2-HG) in tumors harboring isocitrate dehydrogenase 1 (IDH1) mutations. As many metabolites have similar chemical shifts, the resulting 1H spectra of intact biological material are highly convoluted, limiting the application of 1H MR to high abundance metabolites. Hydrogen-Carbon Heteronuclear single quantum correlation 1H-13C HSQC is routinely employed in organic synthesis to resolve complex spectra but has received limited attention for biological studies. Here, we show that 1H-13C HSQC offers a dramatic improvement in sensitivity compared to one-dimensional (1D) 13C NMR and dramatic signal deconvolution compared to 1D 1H spectra in an intact biological setting. Using a standard NMR spectroscope without specialized signal enhancements features such as magic angle spinning, metabolite extractions or 13C-isotopic enrichment, we obtain well-resolved 2D 1H-13C HSQC spectra in live cancer cells, in ex-vivo freshly dissected xenografted tumors and resected primary tumors. We demonstrate that this method can readily identify tumors with specific genetic-driven oncometabolite alterations such as IDH mutations with elevation of 2-HG as well as PGD-homozygously deleted tumors with elevation of gluconate. These data support the potential of 1H-13C HSQC as a non-invasive diagnostic tool for metabolic precision oncology.
- Published
- 2019
- Full Text
- View/download PDF
22. OMRT-7. Angiogenesis inhibitors strongly synergize with therapeutics targeting tumor metabolism
- Author
-
Sunada Khadka
- Subjects
Bevacizumab ,Tivozanib ,Angiogenesis ,medicine.drug_class ,business.industry ,AMPK ,Cancer ,medicine.disease ,Tyrosine-kinase inhibitor ,Metformin ,Supplement Abstracts ,Final Category: Omics of Response to Therapy ,Cancer cell ,medicine ,Cancer research ,AcademicSubjects/MED00300 ,AcademicSubjects/MED00310 ,business ,medicine.drug - Abstract
Angiogenesis inhibition has become a mainstay of oncology despite having fallen short of its early promise. As originally envisioned, angiogenesis inhibition would cut off the blood supply, deprive tumor cells of key nutrients, leading to their death. In practice, while there is evidence that tumors under angiogenesis treatment do in fact exhibit some degree of metabolic stress, this is stress is not sufficient to induce significant cancer cell death. We posit that the full potential of angiogenesis inhibition can be realized by the combination of angiogenesis inhibition with emerging tumor metabolism targeting therapies. Because tumors under angiogenesis inhibition are already in a state of nutrient stress, the effects of metabolically targeted therapies such as amino acid depletion (e.g. asparginase, methionine restriction), inhibitors of stress adaption (AMPK and GCN2 inhibitors) or energy metabolism (e.g. IACS-010759, Metformin, POMHEX) stand to dramatically increase in potency whilst remaining selective for (angiogenic) tumor versus (non-angiogenic) normal tissue. Here, we provide proof-of-principal for this thesis. First, we performed metabolomic profiling of angiogenesis-inhibited tumors, which corroborates a state of nutrient stress in angiogenesis-inhibited tumors. Second, we demonstrate dramatic anti-neoplastic synergy (effectively curing of xenografted tumor-bearing mice, irrespective of initial tumor size), without enhanced adverse toxicities, between the OxPhos inhibitor IACS-010759 and the angiogenesis tyrosine kinase inhibitor, Tivozanib. The same results were recapitulated with the anti-VEGFA antibody, Avastin, and the OxPhos inhibitor could be substituted with the Enolase inhibitor HEX, with similar effects. The synergy was observed in a broad range of tumor types, even those without clear genetic susceptibilities. Together, these results suggest that angiogenesis inhibitors synergize broadly with cancer therapies targeting metabolism, allowing the realization of the full potential of these previously disappointing drugs. Our results warrant systematic combination clinical trials between angiogenesis inhibitors and established, as well as emerging anti-metabolic cancer therapies.
- Published
- 2021
23. Author Correction: An enolase inhibitor for the targeted treatment of ENO1-deleted cancers
- Author
-
Nikunj Satani, Rafal Zielinski, Kenisha Arthur, Ronald A. DePinho, Zhenghong Peng, Pijus K. Mandal, Quanyu Xu, Yasaman Barekatain, David Maxwell, Florian L. Muller, Yuting Sun, Qi Wu, Dimitra K. Georgiou, Yongying Jiang, Theresa Tran, Victoria C. Yan, Yu Hsi Lin, William G. Bornmann, Paul G. Leonard, John M. Asara, Zhijun Kang, Cong-Dat Pham, Barbara Czako, Joseph R. Marszalek, Jeffrey J. Ackroyd, Sunada Khadka, Susana Castro Pando, Waldemar Priebe, Xiaobo Wang, Naima Hammoudi, and Federica Pisaneschi
- Subjects
business.industry ,Physiology (medical) ,Endocrinology, Diabetes and Metabolism ,Published Erratum ,Enolase ,Internal Medicine ,Cancer research ,MEDLINE ,Medicine ,Cell Biology ,business - Published
- 2020
24. Targeting YAP-Dependent MDSC Infiltration Impairs Tumor Progression
- Author
-
Lynda Chin, Sujun Hua, Y. Alan Wang, Jianhua Zhang, Kun Zhao, Timothy P. Heffernan, Elsa M. Li-Ning-Tapia, Xiaolu Pan, Trang N. Tieu, Eun Jung Jin, Zhihu Ding, Vandhana Ramamoorthy, Prasenjit Dey, Ronald A. DePinho, Wantong Yao, Patricia Troncoso, Sunada Khadka, Pingping Hou, Shan Jiang, Zhuangna Fang, Avnish Kapoor, Ramakrishna Konaparthi, Qing Chang, Yanxia Shi, Christopher J. Logothetis, Xiaoying Shang, Chia Chin Wu, Neelay Bhaskar Patel, Guocan Wang, Xin Lu, Pingna Deng, Mark J. McArthur, Chang-Jiun Wu, Baoli Hu, Zhenglin Guo, Di Zhao, and Liren Li
- Subjects
Male ,0301 basic medicine ,Chemokine CXCL5 ,Stromal cell ,Protein Serine-Threonine Kinases ,Article ,Receptors, Interleukin-8B ,Mice ,03 medical and health sciences ,Prostate cancer ,Cell Line, Tumor ,Animals ,Humans ,Medicine ,PTEN ,Hippo Signaling Pathway ,Myeloid Cells ,CXC chemokine receptors ,Adaptor Proteins, Signal Transducing ,Smad4 Protein ,YAP1 ,biology ,business.industry ,PTEN Phosphohydrolase ,Prostatic Neoplasms ,YAP-Signaling Proteins ,Phosphoproteins ,medicine.disease ,030104 developmental biology ,Oncology ,Tumor progression ,CXCL5 ,Immunology ,Cancer cell ,Disease Progression ,Cancer research ,biology.protein ,business ,Signal Transduction ,Transcription Factors - Abstract
The signaling mechanisms between prostate cancer cells and infiltrating immune cells may illuminate novel therapeutic approaches. Here, utilizing a prostate adenocarcinoma model driven by loss of Pten and Smad4, we identify polymorphonuclear myeloid-derived suppressor cells (MDSC) as the major infiltrating immune cell type, and depletion of MDSCs blocks progression. Employing a novel dual reporter prostate cancer model, epithelial and stromal transcriptomic profiling identified CXCL5 as a cancer-secreted chemokine to attract CXCR2-expressing MDSCs, and, correspondingly, pharmacologic inhibition of CXCR2 impeded tumor progression. Integrated analyses identified hyperactivated Hippo–YAP signaling in driving CXCL5 upregulation in cancer cells through the YAP–TEAD complex and promoting MDSC recruitment. Clinicopathologic studies reveal upregulation and activation of YAP1 in a subset of human prostate tumors, and the YAP1 signature is enriched in primary prostate tumor samples with stronger expression of MDSC-relevant genes. Together, YAP-driven MDSC recruitment via heterotypic CXCL5–CXCR2 signaling reveals an effective therapeutic strategy for advanced prostate cancer. Significance: We demonstrate a critical role of MDSCs in prostate tumor progression and discover a cancer cell nonautonomous function of the Hippo–YAP pathway in regulation of CXCL5, a ligand for CXCR2-expressing MDSCs. Pharmacologic elimination of MDSCs or blocking the heterotypic CXCL5–CXCR2 signaling circuit elicits robust antitumor responses and prolongs survival. Cancer Discov; 6(1); 80–95. ©2015 AACR. This article is highlighted in the In This Issue feature, p. 1
- Published
- 2016
25. Eradication of ENO1-deleted Glioblastoma through Collateral Lethality
- Author
-
Barbara Czako, Theresa Tran, Florian L. Muller, Paul G. Leonard, William G. Bornmann, Marszalek, Nikunj Satani, Yu-Hsi Lin, Dimitra K. Georgiou, Federica Pisaneschi, Rafal Zielinski, Yongying Jiang, Victoria C. Yan, Quanyun Xu, David Maxwell, Pijus K. Mandal, Ronald A. DePinho, Waldemar Priebe, Jeffrey J. Ackroyd, Naima Hammoudi, Zhijun Kang, John M. Asara, Sunada Khadka, Qi Wu, Susana Castro Pando, Xiaoping Wang, Yijun Sun, and Zhenghong Peng
- Subjects
Hemolytic anemia ,0303 health sciences ,Chemistry ,Enolase ,Cell ,Cancer ,medicine.disease ,Pivaloyloxymethyl ,3. Good health ,03 medical and health sciences ,0302 clinical medicine ,medicine.anatomical_structure ,In vivo ,030220 oncology & carcinogenesis ,Glioma ,Cancer research ,medicine ,Glycolysis ,030304 developmental biology - Abstract
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.
- Published
- 2018
26. An in vivo screen identifies PYGO2 as a driver for metastatic prostate cancer
- Author
-
Chang-Jiun Wu, Yong Zang, Xiaolu Pan, Y. Alan Wang, Yanting Luo, Xin Lu, Shan Jiang, Eun-Jung Jin, Samirkumar B. Amin, Xiaoying Shang, Nora M. Navone, Xuemin Lu, William R. Morgenlander, Patricia Troncoso, Pingna Deng, Rumi Lee, Qing Chang, Ronald A. DePinho, Di Zhao, Jacqueline Weinrich, Sunada Khadka, and Shan Feng
- Subjects
0301 basic medicine ,Male ,Transcriptional Activation ,Cancer Research ,Carcinogenesis ,Mice, Nude ,Biology ,medicine.disease_cause ,Article ,Metastasis ,03 medical and health sciences ,Prostate cancer ,Mice ,Cell Line, Tumor ,medicine ,Biomarkers, Tumor ,Animals ,Humans ,Wnt Signaling Pathway ,Oncogene ,Wnt signaling pathway ,Intracellular Signaling Peptides and Proteins ,Cancer ,Prostatic Neoplasms ,Oncogenes ,medicine.disease ,Primary tumor ,Up-Regulation ,Gene Expression Regulation, Neoplastic ,030104 developmental biology ,HEK293 Cells ,Oncology ,Tumor progression ,PC-3 Cells ,Cancer research ,Disease Progression ,Lymph Nodes ,Neoplasm Grading - Abstract
Advanced prostate cancer displays conspicuous chromosomal instability and rampant copy number aberrations, yet the identity of functional drivers resident in many amplicons remain elusive. Here, we implemented a functional genomics approach to identify new oncogenes involved in prostate cancer progression. Through integrated analyses of focal amplicons in large prostate cancer genomic and transcriptomic datasets as well as genes upregulated in metastasis, 276 putative oncogenes were enlisted into an in vivo gain-of-function tumorigenesis screen. Among the top positive hits, we conducted an in-depth functional analysis on Pygopus family PHD finger 2 (PYGO2), located in the amplicon at 1q21.3. PYGO2 overexpression enhances primary tumor growth and local invasion to draining lymph nodes. Conversely, PYGO2 depletion inhibits prostate cancer cell invasion in vitro and progression of primary tumor and metastasis in vivo. In clinical samples, PYGO2 upregulation associated with higher Gleason score and metastasis to lymph nodes and bone. Silencing PYGO2 expression in patient-derived xenograft models impairs tumor progression. Finally, PYGO2 is necessary to enhance the transcriptional activation in response to ligand-induced Wnt/β-catenin signaling. Together, our results indicate that PYGO2 functions as a driver oncogene in the 1q21.3 amplicon and may serve as a potential prognostic biomarker and therapeutic target for metastatic prostate cancer. Significance: Amplification/overexpression of PYGO2 may serve as a biomarker for prostate cancer progression and metastasis. Cancer Res; 78(14); 3823–33. ©2018 AACR.
- Published
- 2018
27. Abstract 984: Synthesis of mixed, hypoxia-activated phosphoramidate esters for the inhibition of Enolase in ENO1-deleted glioblastoma
- Author
-
Kenisha Arthur, Florian L. Muller, Elliot S. Ballato, Pakriti Shrestha, Jeffrey J. Ackroyd, Sunada Khadka, Kristine L. Yang, Dimitra K. Georgiou, and Victoria C. Yan
- Subjects
Cancer Research ,Oncology ,Chemistry ,Enolase ,medicine ,Cancer research ,Phosphoramidate ,Hypoxia (medical) ,medicine.symptom ,medicine.disease ,Glioblastoma - Abstract
Background: Precision oncology is currently restricted to activated oncogenes as drug targets, with tumor suppressor deletions remaining largely in-actionable. We developed a novel method that targets tumor suppressor deletions by exploiting vulnerabilities generated by the co-deletion of neighboring metabolic housekeeping genes with critical but normally redundant functions. Proof-of-principle studies were conducted for the glycolytic enzyme Enolase. Cancers harboring the homozygous deletion of 1p36-localized ENO1 remain metabolically active and viable through redundant action of its paralogue, ENO2. Inhibition of ENO2 in cancer cells harboring ENO1-homozygous deletion discriminately kills ENO1-deleted cells. In accordance, our synthetic efforts have focused on phosphonate chemistry to generate clinically-useful substrate mimics of 2-phosphoglycerate, the natural substrate for Enolase. We have generated a phosphonohydroxamate ENO2-inhibitor termed HEX and a pro-drug of which, POMHEX, kills ENO1-homozygously deleted glioma cells in culture with strong specificity. However, the pharmacokinetics of POMHEX are poor, due to the presence of carboxyesterase in extracellular fluids, including plasma. Results: We have synthesized mechanistically different pro-drugs of HEX, utilizing nitro-aromatics as protecting groups. Nitroaromatic groups are bioactivated by nitroreductases intracellularly, in a manner that is inversely related to oxygen concentration. As such, removal of nitroaromatic pro-drug moieties and release of active drug is favored under low oxygen (hypoxic) conditions. We synthesized a series of nitroaromatic pro-drugs of HEX (the structures will be disclosed at AACR presentation, pending patent filings) and demonstrate that 1) these exhibit selective toxicity to ENO1-deleted glioma cells in culture and 2) this killing is dramatically enhanced by lowering oxygen concentrations as compared to POMHEX. We further find that redox potential of the nitroaromatic moiety correlates with the degree of hypoxia activation with the lower redox potential being associated with great oxygen dependence. Importantly, unlike POMHEX, nitroaromatic HEX pro-drugs are stable in human plasma ex-vivo. We have thus successfully synthesized a pro-drug inhibitor of Enolase that is hypoxia-activated. Conclusion and future directions: We are gearing up synthesis for pre-clinical evaluation of nitroaromatic-HEX in intracranial tumor xenograft models. While the primary purpose of Nitroaromatic HEX is for the precision oncology treatment of ENO1-homozygously deleted tumors, it has not escaped our notice that such drugs have wider applicability within the context of targeting treatment-resistant tumors cells hiding in hypoxic niches. Indeed, we have observed that virtually all glioma cell lines can be killed by nitroaromatic HEX under hypoxia. Note: This abstract was not presented at the meeting. Citation Format: Florian Muller, Victoria C. Yan, Elliot S. Ballato, Kristine L. Yang, Dimitra K. Georgiou, Kenisha Arthur, Pakriti Shrestha, Sunada Khadka, Jeffrey Ackroyd. Synthesis of mixed, hypoxia-activated phosphoramidate esters for the inhibition of Enolase in ENO1-deleted glioblastoma [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2019; 2019 Mar 29-Apr 3; Atlanta, GA. Philadelphia (PA): AACR; Cancer Res 2019;79(13 Suppl):Abstract nr 984.
- Published
- 2019
28. Bioreducible Phosphonoamidate Pro-drug Inhibitor of Enolase: Proof of Concept Study.
- Author
-
Yan, Victoria C., Yang, Kristine L., Ballato, Elliot S., Sunada Khadka, Prakriti Shrestha, Kenisha Arthur, Georgiou, Dimitra K., Mykia Washington, Tran, Theresa, Poral, Anton H., Cong-Dat Pham, Yan, Matthew J., and Muller, Florian L.
- Published
- 2020
- Full Text
- View/download PDF
29. Abstract 2831: Collateral lethality: A new target for personalized oncology
- Author
-
Marina Protopopova, Yu-Hsi Lin, Barbara Czako, Alan Y. Wang, Nikunj Satani, Maria Emilia Di Francesco, Naima Hammoudi, Florian L. Muller, Sunada Khadka, Yuting Sun, Ronald A. DePinho, Dimitra K. Georgiou, Jeffrey J. Ackroyd, and Joe Marszalek
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Collateral ,business.industry ,Internal medicine ,Personalized oncology ,medicine ,Lethality ,business - Abstract
Large-scale genomic profiling efforts, such as The Cancer Genome Atlas (TCGA) have painted an unprecedentedly detailed picture of the genetic alterations that underlie carcinogenesis, yet the key challenge remains as to how to turn this information into actionable therapies. Genomic deletions are a frequent event in diverse cancers, which inactivate a limited number of tumor suppressor genes (“driver”-events) but frequently include many chromosomal neighbors as “passengers”, some of which play critical but redundant roles in normal cellular housekeeping. The overall hypothesis we propose to test is that collateral deletions of such “passenger” genes can be utilized as novel targets of synthetic lethality, an idea which we term “collateral lethality." The large number of passenger deleted genes, playing diverse functions in cell homeostasis, offers a rich repertoire of pharmacologically targetable vulnerabilities presenting novel opportunities for the development of personalized anti-neoplastic therapies. We have provided in vitro proof-of-principal of a collaterally deleted glycolytic gene Enolase 1 (ENO1) at the 1p36 tumor suppressor locus in glioblastoma (GBM), that leads to dramatic sensitization to inhibition of the redundant paralogue, ENO2. The next step is to take this concept to the clinic. Our overall goal is to generate a clinical candidate Enolase inhibitor for tumors with ENO1-deletions, like GBM for which there is no other treatment option and the therapeutic benefit could be quite substantial. Citation Format: Yu-Hsi Lin, Naima Hammoudi, Nikunj Satani, Jeffrey Ackroyd, Sunada Khadka, Dimitra Georgiou, Joe Marszalek, Yuting Sun, Marina Protopopova, Maria E. Di Francesco, Barbara Czako, Alan Y. Wang, Ronald A. DePinho, Florian L. Muller. Collateral lethality: A new target for personalized oncology [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2018; 2018 Apr 14-18; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2018;78(13 Suppl):Abstract nr 2831.
- Published
- 2018
30. Targeting YAP-dependent MDSC infiltration impairs tumor progression
- Author
-
Xin Lu, Y. Alan Wang, Lynda Chin, Christopher J. Logothetis, Elsa M. Li Ning Tapia, Zhuangna Fang, Pingping Hou, Baoli Hu, Trang N. Tieu, Shan Jiang, Kun Zhao, Xiaoying Shang, Guocan Wang, Chia Chin Wu, Ramakrishna Konaparthi, Sujun Hua, Sunada Khadka, Timothy P. Heffernan, Vandhana Ramamoorthy, Ronald A. DePinho, Avnish Kapoor, Pingna Deng, Mark J. McArthur, Zhenglin Guo, Di Zhao, Neelay Bhaskar Patel, Qing Chang, Patricia Troncoso, Prasenjit Dey, and Jianhua Zhang
- Subjects
Pharmacology ,Cancer Research ,business.industry ,Immunology ,medicine.disease ,Bioinformatics ,law.invention ,Heterogeneous population ,Prostate cancer ,Oncology ,law ,Tumor progression ,Poster Presentation ,Myeloid cells ,medicine ,Cancer research ,Molecular Medicine ,Immunology and Allergy ,Suppressor ,business ,Solid tumor ,Infiltration (medical) - Abstract
Meeting abstracts Myeloid-derived suppressor cells (MDSCs) represent a phenotypically heterogeneous population of immature myeloid cells that play a tumor-promoting role by maintaining a state of immunological anergy and tolerance. Similar to other solid tumor types, Prostate cancer (PCa) is
- Published
- 2015
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.