62 results on '"Sylwia A. Stopka"'
Search Results
2. A non-dividing cell population with high pyruvate dehydrogenase kinase activity regulates metabolic heterogeneity and tumorigenesis in the intestine
- Author
-
Carlos Sebastian, Christina Ferrer, Maria Serra, Jee-Eun Choi, Nadia Ducano, Alessia Mira, Manasvi S. Shah, Sylwia A. Stopka, Andrew J. Perciaccante, Claudio Isella, Daniel Moya-Rull, Marianela Vara-Messler, Silvia Giordano, Elena Maldi, Niyati Desai, Diane E. Capen, Enzo Medico, Murat Cetinbas, Ruslan I. Sadreyev, Dennis Brown, Miguel N. Rivera, Anna Sapino, David T. Breault, Nathalie Y. R. Agar, and Raul Mostoslavsky
- Subjects
Science - Abstract
Metabolic reprogramming upon SIRT6 loss induces tumour formation in the intestine but the mechanism is unclear. Here, the authors show that loss of SIRT6 leads to the expansion of epithelial cells with high pyruvate dehydrogenase kinase activity resulting in enhanced stem cell activity and tumour-initiating potential
- Published
- 2022
- Full Text
- View/download PDF
3. Interim clinical trial analysis of intraoperative mass spectrometry for breast cancer surgery
- Author
-
Sankha S. Basu, Sylwia A. Stopka, Walid M. Abdelmoula, Elizabeth C. Randall, Begoña Gimenez-Cassina Lopez, Michael S. Regan, David Calligaris, Fake F. Lu, Isaiah Norton, Melissa A. Mallory, Sandro Santagata, Deborah A. Dillon, Mehra Golshan, and Nathalie Y. R. Agar
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Optimal resection of breast tumors requires removing cancer with a rim of normal tissue while preserving uninvolved regions of the breast. Surgical and pathological techniques that permit rapid molecular characterization of tissue could facilitate such resections. Mass spectrometry (MS) is increasingly used in the research setting to detect and classify tumors and has the potential to detect cancer at surgical margins. Here, we describe the ex vivo intraoperative clinical application of MS using a liquid micro-junction surface sample probe (LMJ-SSP) to assess breast cancer margins. In a midpoint analysis of a registered clinical trial, surgical specimens from 21 women with treatment naïve invasive breast cancer were prospectively collected and analyzed at the time of surgery with subsequent histopathological determination. Normal and tumor breast specimens from the lumpectomy resected by the surgeon were smeared onto glass slides for rapid analysis. Lipidomic profiles were acquired from these specimens using LMJ-SSP MS in negative ionization mode within the operating suite and post-surgery analysis of the data revealed five candidate ions separating tumor from healthy tissue in this limited dataset. More data is required before considering the ions as candidate markers. Here, we present an application of ambient MS within the operating room to analyze breast cancer tissue and surgical margins. Lessons learned from these initial promising studies are being used to further evaluate the five candidate biomarkers and to further refine and optimize intraoperative MS as a tool for surgical guidance in breast cancer.
- Published
- 2021
- Full Text
- View/download PDF
4. Multiplatform Metabolomics Studies of Human Cancers With NMR and Mass Spectrometry Imaging
- Author
-
Anya B. Zhong, Isabella H. Muti, Stephen J. Eyles, Richard W. Vachet, Kristen N. Sikora, Cedric E. Bobst, David Calligaris, Sylwia A. Stopka, Jeffery N. Agar, Chin-Lee Wu, Mari A. Mino-Kenudson, Nathalie Y. R. Agar, David C. Christiani, Igor A. Kaltashov, and Leo L. Cheng
- Subjects
metabolomics ,imaging ,mass spectrometry ,nuclear magnetic resonance spectroscopy ,lung cancer ,prostate cancer ,Biology (General) ,QH301-705.5 - Abstract
The status of metabolomics as a scientific branch has evolved from proof-of-concept to applications in science, particularly in medical research. To comprehensively evaluate disease metabolomics, multiplatform approaches of NMR combining with mass spectrometry (MS) have been investigated and reported. This mixed-methods approach allows for the exploitation of each individual technique’s unique advantages to maximize results. In this article, we present our findings from combined NMR and MS imaging (MSI) analysis of human lung and prostate cancers. We further provide critical discussions of the current status of NMR and MS combined human prostate and lung cancer metabolomics studies to emphasize the enhanced metabolomics ability of the multiplatform approach.
- Published
- 2022
- Full Text
- View/download PDF
5. Neuropeptide Localization in Lymnaea stagnalis: From the Central Nervous System to Subcellular Compartments
- Author
-
Ellen A. Wood, Sylwia A. Stopka, Linwen Zhang, Sara Mattson, Gabor Maasz, Zsolt Pirger, and Akos Vertes
- Subjects
Lymnaea stagnalis ,mass spectrometry ,neuropeptide ,central nervous system ,single neuron analysis ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 - Abstract
Due to the relatively small number of neurons (few tens of thousands), the well-established multipurpose model organism Lymnaea stagnalis, great pond snail, has been extensively used to study the functioning of the nervous system. Unlike the more complex brains of higher organisms, L. stagnalis has a relatively simple central nervous system (CNS) with well-defined circuits (e.g., feeding, locomotion, learning, and memory) and identified individual neurons (e.g., cerebral giant cell, CGC), which generate behavioral patterns. Accumulating information from electrophysiological experiments maps the network of neuronal connections and the neuronal circuits responsible for basic life functions. Chemical signaling between synaptic-coupled neurons is underpinned by neurotransmitters and neuropeptides. This review looks at the rapidly expanding contributions of mass spectrometry (MS) to neuropeptide discovery and identification at different granularity of CNS organization. Abundances and distributions of neuropeptides in the whole CNS, eleven interconnected ganglia, neuronal clusters, single neurons, and subcellular compartments are captured by MS imaging and single cell analysis techniques. Combining neuropeptide expression and electrophysiological data, and aided by genomic and transcriptomic information, the molecular basis of CNS-controlled biological functions is increasingly revealed.
- Published
- 2021
- Full Text
- View/download PDF
6. Optical Microscopy-Guided Laser Ablation Electrospray Ionization Ion Mobility Mass Spectrometry: Ambient Single Cell Metabolomics with Increased Confidence in Molecular Identification
- Author
-
Michael J. Taylor, Sara Mattson, Andrey Liyu, Sylwia A. Stopka, Yehia M. Ibrahim, Akos Vertes, and Christopher R. Anderton
- Subjects
mass spectrometry ,collisional cross section ,drift tube ion mobility separation ,laser ablation electrospray ionization ,in situ metabolomics ,ambient analysis ,Microbiology ,QR1-502 - Abstract
Single cell analysis is a field of increasing interest as new tools are continually being developed to understand intercellular differences within large cell populations. Laser-ablation electrospray ionization mass spectrometry (LAESI-MS) is an emerging technique for single cell metabolomics. Over the years, it has been validated that this ionization technique is advantageous for probing the molecular content of individual cells in situ. Here, we report the integration of a microscope into the optical train of the LAESI source to allow for visually informed ambient in situ single cell analysis. Additionally, we have coupled this ‘LAESI microscope’ to a drift-tube ion mobility mass spectrometer to enable separation of isobaric species and allow for the determination of ion collision cross sections in conjunction with accurate mass measurements. This combined information helps provide higher confidence for structural assignment of molecules ablated from single cells. Here, we show that this system enables the analysis of the metabolite content of Allium cepa epidermal cells with high confidence structural identification together with their spatial locations within a tissue.
- Published
- 2021
- Full Text
- View/download PDF
7. Metabolic Noise and Distinct Subpopulations Observed by Single Cell LAESI Mass Spectrometry of Plant Cells in situ
- Author
-
Sylwia A. Stopka, Rikkita Khattar, Beverly J. Agtuca, Christopher R. Anderton, Ljiljana Paša-Tolić, Gary Stacey, and Akos Vertes
- Subjects
single cell analysis ,in situ ,plant cells ,heterogeneity ,metabolic noise ,mass spectrometry ,Plant culture ,SB1-1110 - Abstract
Phenotypic variations and stochastic expression of transcripts, proteins, and metabolites in biological tissues lead to cellular heterogeneity. As a result, distinct cellular subpopulations emerge. They are characterized by different metabolite expression levels and by associated metabolic noise distributions. To capture these biological variations unperturbed, highly sensitive in situ analytical techniques are needed that can sample tissue embedded single cells with minimum sample preparation. Optical fiber-based laser ablation electrospray ionization mass spectrometry (f-LAESI-MS) is a promising tool for metabolic profiling of single cells under ambient conditions. Integration of this MS-based platform with fluorescence and brightfield microscopy provides the ability to target single cells of specific type and allows for the selection of rare cells, e.g., excretory idioblasts. Analysis of individual Egeria densa leaf blade cells (n = 103) by f-LAESI-MS revealed significant differences between the prespecified subpopulations of epidermal cells (n = 97) and excretory idioblasts (n = 6) that otherwise would have been masked by the population average. Primary metabolites, e.g., malate, aspartate, and ascorbate, as well as several glucosides were detected in higher abundance in the epidermal cells. The idioblasts contained lipids, e.g., PG(16:0/18:2), and triterpene saponins, e.g., medicoside I and azukisaponin I, and their isomers. Metabolic noise for the epidermal cells were compared to results for soybean (Glycine max) root nodule cells (n = 60) infected by rhizobia (Bradyrhizobium japonicum). Whereas some primary metabolites showed lower noise in the latter, both cell types exhibited higher noise for secondary metabolites. Post hoc grouping of epidermal and root nodule cells, based on the abundance distributions for certain metabolites (e.g., malate), enabled the discovery of cellular subpopulations characterized by different mean abundance values, and the magnitudes of the corresponding metabolic noise. Comparison of prespecified populations from epidermal cells of the closely related E. densa (n = 20) and Elodea canadensis (n = 20) revealed significant differences, e.g., higher sugar content in the former and higher levels of ascorbate in the latter, and the presence of species-specific metabolites. These results demonstrate that the f-LAESI-MS single cell analysis platform has the potential to explore cellular heterogeneity and metabolic noise for hundreds of tissue-embedded cells.
- Published
- 2018
- Full Text
- View/download PDF
8. WSD-0922, a novel brain-penetrant inhibitor of EGFR, promotes survival in glioblastoma mouse models
- Author
-
Jason E Conage-Pough, Sylwia A Stopka, Ju-Hee Oh, Ann C Mladek, Danielle M Burgenske, Michael S Regan, Gerard Baquer, Paul A Decker, Brett L Carlson, Katrina K Bakken, Jinqiang Zhang, Lily Liu, Claire Sun, Zhihua Mu, Wei Zhong, Nhan L Tran, William F Elmquist, Nathalie Y R Agar, Jann N Sarkaria, and Forest M White
- Subjects
Oncology ,Surgery ,Neurology (clinical) - Abstract
Background Although the epidermal growth factor receptor (EGFR) is a frequent oncogenic driver in glioblastoma (GBM), efforts to therapeutically target this protein have been largely unsuccessful. The present pre-clinical study evaluated the novel EGFR inhibitor WSD-0922. Methods We employed flank and orthotopic patient derived xenograft (PDX) models to characterize WSD-0922 and compare its efficacy to erlotinib, a potent EGFR inhibitor that failed to provide benefit for GBM patients. We performed long-term survival studies and collected short-term tumor, plasma, and whole-brain samples from mice treated with each drug. We utilized mass spectrometry to measure drug concentrations and spatial distribution and to assess the impact of each drug on receptor activity and cellular signaling networks. Results WSD-0922 inhibited EGFR signaling as effectively as erlotinib in in vitro and in vivo models. While WSD-0922 was more CNS penetrant than erlotinib in terms of total concentration, comparable concentrations of both drugs were measured at the tumor site in orthotopic models, and the concentration of free WSD-0922 in the brain was significantly less than the concentration of free erlotinib. WSD-0922 treatment provided a clear survival advantage compared to erlotinib in the GBM39 model, with marked suppression of tumor growth and most mice surviving until the end of the study. WSD-0922 treatment preferentially inhibited phosphorylation of several proteins, including those associated with EGFR inhibitor resistance and cell metabolism. Conclusions WSD-0922 is a highly potent inhibitor of EGFR in GBM, and warrants further evaluation in clinical studies.
- Published
- 2023
9. Predicting Prostate Cancer Directly from Tissue Images using Deep Learning on Mass Spectrometry Imaging and Whole Slide Imaging Data
- Author
-
Md Inzamam Ul Haque, Debangshu Mukherjee, Sylwia A Stopka, Nathalie YR Agar, Jacob Hinkle, and Olga S Ovchinnikova
- Subjects
Instrumentation - Published
- 2022
10. Interim clinical trial analysis of intraoperative mass spectrometry for breast cancer surgery
- Author
-
Mehra Golshan, Elizabeth C. Randall, Melissa Anne Mallory, David Calligaris, Sylwia A. Stopka, Sankha S. Basu, Michael S. Regan, Isaiah Norton, Deborah A. Dillon, Walid M. Abdelmoula, Begoña Gimenez-Cassina Lopez, Sandro Santagata, Fa-Ke F. Lu, and Nathalie Y. R. Agar
- Subjects
medicine.medical_specialty ,business.industry ,medicine.medical_treatment ,Lumpectomy ,Normal tissue ,Cancer ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,medicine.disease ,Surgery ,Resection ,Therapy naive ,Clinical trial ,Breast cancer ,Oncology ,medicine ,Pharmacology (medical) ,Radiology, Nuclear Medicine and imaging ,business ,Research setting ,RC254-282 - Abstract
Optimal resection of breast tumors requires removing cancer with a rim of normal tissue while preserving uninvolved regions of the breast. Surgical and pathological techniques that permit rapid molecular characterization of tissue could facilitate such resections. Mass spectrometry (MS) is increasingly used in the research setting to detect and classify tumors and has the potential to detect cancer at surgical margins. Here, we describe the ex vivo intraoperative clinical application of MS using a liquid micro-junction surface sample probe (LMJ-SSP) to assess breast cancer margins. In a midpoint analysis of a registered clinical trial, surgical specimens from 21 women with treatment naïve invasive breast cancer were prospectively collected and analyzed at the time of surgery with subsequent histopathological determination. Normal and tumor breast specimens from the lumpectomy resected by the surgeon were smeared onto glass slides for rapid analysis. Lipidomic profiles were acquired from these specimens using LMJ-SSP MS in negative ionization mode within the operating suite and post-surgery analysis of the data revealed five candidate ions separating tumor from healthy tissue in this limited dataset. More data is required before considering the ions as candidate markers. Here, we present an application of ambient MS within the operating room to analyze breast cancer tissue and surgical margins. Lessons learned from these initial promising studies are being used to further evaluate the five candidate biomarkers and to further refine and optimize intraoperative MS as a tool for surgical guidance in breast cancer.
- Published
- 2021
11. Oncometabolite d-2HG alters T cell metabolism to impair CD8
- Author
-
Giulia Notarangelo, Jessica B. Spinelli, Elizabeth M. Perez, Gregory J. Baker, Kiran Kurmi, Ilaria Elia, Sylwia A. Stopka, Gerard Baquer, Jia-Ren Lin, Alexandra J. Golby, Shakchhi Joshi, Heide F. Baron, Jefte M. Drijvers, Peter Georgiev, Alison E. Ringel, Elma Zaganjor, Samuel K. McBrayer, Peter K. Sorger, Arlene H. Sharpe, Kai W. Wucherpfennig, Sandro Santagata, Nathalie Y. R. Agar, Mario L. Suvà, and Marcia C. Haigis
- Subjects
IDH ,Carcinogenesis ,T-Lymphocytes ,CD8-Positive T-Lymphocytes ,PHENOTYPE ,Article ,Glutarates ,Interferon-gamma ,Mice ,DEHYDROGENASE ,MITOCHONDRIA ,Neoplasms ,Animals ,Humans ,Science & Technology ,IFN-GAMMA ,Multidisciplinary ,2-HYDROXYGLUTARATE ,L-Lactate Dehydrogenase ,MUTATIONS ,MAGNETIC-RESONANCE-SPECTROSCOPY ,Isocitrate Dehydrogenase ,Multidisciplinary Sciences ,ALPHA ,DIFFERENTIATION ,Gain of Function Mutation ,Mutation ,Science & Technology - Other Topics - Abstract
Gain-of-function mutations in isocitrate dehydrogenase (IDH) in human cancers result in the production of d -2-hydroxyglutarate ( d -2HG), an oncometabolite that promotes tumorigenesis through epigenetic alterations. The cancer cell–intrinsic effects of d -2HG are well understood, but its tumor cell–nonautonomous roles remain poorly explored. We compared the oncometabolite d -2HG with its enantiomer, l -2HG, and found that tumor-derived d -2HG was taken up by CD8 + T cells and altered their metabolism and antitumor functions in an acute and reversible fashion. We identified the glycolytic enzyme lactate dehydrogenase (LDH) as a molecular target of d -2HG. d -2HG and inhibition of LDH drive a metabolic program and immune CD8 + T cell signature marked by decreased cytotoxicity and impaired interferon-γ signaling that was recapitulated in clinical samples from human patients with IDH1 mutant gliomas.
- Published
- 2022
12. Convection enhanced delivery of EGFR targeting antibody-drug conjugates Serclutamab talirine and Depatux-M in glioblastoma patient-derived xenografts
- Author
-
Kendra A Porath, Michael S Regan, Jessica I Griffith, Sonia Jain, Sylwia A Stopka, Danielle M Burgenske, Katrina K Bakken, Brett L Carlson, Paul A Decker, Rachael A Vaubel, Sonja Dragojevic, Ann C Mladek, Margaret A Connors, Zeng Hu, Lihong He, Gaspar J Kitange, Shiv K Gupta, Thomas M Feldsien, Didier R Lefebvre, Nathalie Y R Agar, Jeanette E Eckel-Passow, Edward B Reilly, William F Elmquist, and Jann N Sarkaria
- Subjects
General Medicine - Abstract
Background EGFR targeting antibody-drug conjugates (ADCs) are highly effective against EGFR-amplified tumors, but poor distribution across the blood–brain barrier (BBB) limits their efficacy in glioblastoma (GBM) when administered systemically. We studied whether convection-enhanced delivery (CED) can be used to safely infuse ADCs into orthotopic patient-derived xenograft (PDX) models of EGFRvIII mutant GBM. Methods The efficacy of the EGFR-targeted ADCs depatuxizumab mafodotin (Depatux-M) and Serclutamab talirine (Ser-T) was evaluated in vitro and in vivo. CED was performed in nontumor and tumor-bearing mice. Immunostaining was used to evaluate ADC distribution, pharmacodynamic effects, and normal cell toxicity. Results Dose-finding studies in orthotopic GBM6 identified single infusion of 2 μg Ser-T and 60 μg Depatux-M as safe and effective associated with extended survival prolongation (>300 days and 95 days, respectively). However, with serial infusions every 21 days, four Ser-T doses controlled tumor growth but was associated with lethal toxicity approximately 7 days after the final infusion. Limiting dosing to two infusions in GBM108 provided profound median survival extension of over 200 days. In contrast, four Depatux-M CED doses were well tolerated and significantly extended survival in both GBM6 (158 days) and GBM108 (310 days). In a toxicity analysis, Ser-T resulted in a profound loss in NeuN+ cells and markedly elevated GFAP staining, while Depatux-M was associated only with modest elevation in GFAP staining. Conclusion CED of Depatux-M is well tolerated and results in extended survival in orthotopic GBM PDXs. In contrast, CED of Ser-T was associated with a much narrower therapeutic window.
- Published
- 2022
13. High-Throughput Analysis of Tissue-Embedded Single Cells by Mass Spectrometry with Bimodal Imaging and Object Recognition
- Author
-
Nathalie Y. R. Agar, Ellen A. Wood, Sylwia A. Stopka, Rikkita Khattar, Gary Stacey, Beverly J. Agtuca, Walid M. Abdelmoula, and Akos Vertes
- Subjects
Diagnostic Imaging ,chemistry.chemical_classification ,Spectrometry, Mass, Electrospray Ionization ,Chemistry ,Biomolecule ,Metabolite ,Laser ablation electrospray ionization ,Mass spectrometry ,Mass spectrometry imaging ,Analytical Chemistry ,chemistry.chemical_compound ,Microscopy ,Image Processing, Computer-Assisted ,Humans ,Laser Therapy ,Biological system ,Throughput (business) ,Image resolution - Abstract
In biological tissues, cell-to-cell variations stem from the stochastic and modulated expression of genes and the varying abundances of corresponding proteins. These variations are then propagated to downstream metabolite products and result in cellular heterogeneity. Mass spectrometry imaging (MSI) is a promising tool to simultaneously provide spatial distributions for hundreds of biomolecules without the need for labels or stains. Technological advances in MSI instrumentation for the direct analysis of tissue-embedded single cells are dominated by improvements in sensitivity, sample pretreatment, and increased spatial resolution but are limited by low throughput. Herein, we introduce a bimodal microscopy imaging system combined with fiber-based laser ablation electrospray ionization (f-LAESI) MSI with improved throughput ambient analysis of tissue-embedded single cells (n > 1000) to provide insight into cellular heterogeneity. Based on automated image analysis, accurate single-cell sampling is achieved by f-LAESI leading to the discovery of cellular phenotypes characterized by differing metabolite levels.
- Published
- 2021
14. Chronic convection-enhanced delivery of topotecan for patients with recurrent glioblastoma: a first-in-patient, single-centre, single-arm, phase 1b trial
- Author
-
Eleonora F Spinazzi, Michael G Argenziano, Pavan S Upadhyayula, Matei A Banu, Justin A Neira, Dominique M O Higgins, Peter B Wu, Brianna Pereira, Aayushi Mahajan, Nelson Humala, Osama Al-Dalahmah, Wenting Zhao, Akshay V Save, Brian J A Gill, Deborah M Boyett, Tamara Marie, Julia L Furnari, Tejaswi D Sudhakar, Sylwia A Stopka, Michael S Regan, Vanessa Catania, Laura Good, Stergios Zacharoulis, Meenu Behl, Petros Petridis, Sachin Jambawalikar, Akiva Mintz, Angela Lignelli, Nathalie Y R Agar, Peter A Sims, Mary R Welch, Andrew B Lassman, Fabio M Iwamoto, Randy S D’Amico, Jack Grinband, Peter Canoll, and Jeffrey N Bruce
- Subjects
Oncology ,Humans ,Glioma ,Neoplasm Recurrence, Local ,Topotecan ,Glioblastoma ,Convection ,Article - Abstract
Topotecan is cytotoxic to glioma cells but is clinically ineffective because of drug delivery limitations. Systemic delivery is limited by toxicity and insufficient brain penetrance, and, to date, convection-enhanced delivery (CED) has been restricted to a single treatment of restricted duration. To address this problem, we engineered a subcutaneously implanted catheter-pump system capable of repeated, chronic (prolonged, pulsatile) CED of topotecan into the brain and tested its safety and biological effects in patients with recurrent glioblastoma.We did a single-centre, open-label, single-arm, phase 1b clinical trial at Columbia University Irving Medical Center (New York, NY, USA). Eligible patients were at least 18 years of age with solitary, histologically confirmed recurrent glioblastoma showing radiographic progression after surgery, radiotherapy, and chemotherapy, and a Karnofsky Performance Status of at least 70. Five patients had catheters stereotactically implanted into the glioma-infiltrated peritumoural brain and connected to subcutaneously implanted pumps that infused 146 μM topotecan 200 μL/h for 48 h, followed by a 5-7-day washout period before the next infusion, with four total infusions. After the fourth infusion, the pump was removed and the tumour was resected. The primary endpoint of the study was safety of the treatment regimen as defined by presence of serious adverse events. Analyses were done in all treated patients. The trial is closed, and is registered with ClinicalTrials.gov, NCT03154996.Between Jan 22, 2018, and July 8, 2019, chronic CED of topotecan was successfully completed safely in all five patients, and was well tolerated without substantial complications. The only grade 3 adverse event related to treatment was intraoperative supplemental motor area syndrome (one [20%] of five patients in the treatment group), and there were no grade 4 adverse events. Other serious adverse events were related to surgical resection and not the study treatment. Median follow-up was 12 months (IQR 10-17) from pump explant. Post-treatment tissue analysis showed that topotecan significantly reduced proliferating tumour cells in all five patients.In this small patient cohort, we showed that chronic CED of topotecan is a potentially safe and active therapy for recurrent glioblastoma. Our analysis provided a unique tissue-based assessment of treatment response without the need for large patient numbers. This novel delivery of topotecan overcomes limitations in delivery and treatment response assessment for patients with glioblastoma and could be applicable for other anti-glioma drugs or other CNS diseases. Further studies are warranted to determine the effect of this drug delivery approach on clinical outcomes.US National Institutes of Health, The William Rhodes and Louise Tilzer Rhodes Center for Glioblastoma, the Michael Weiner Glioblastoma Research Into Treatment Fund, the Gary and Yael Fegel Foundation, and The Khatib Foundation.
- Published
- 2022
15. β-Cyclodextrin-poly (β-Amino Ester) Nanoparticles Are a Generalizable Strategy for High Loading and Sustained Release of HDAC Inhibitors
- Author
-
Michael S. Regan, Brandon E Knight, Colton W Broughton, Nathalie Y. R. Agar, Cassandra Baker, Martha J Fowler, Lindsey Sablatura, Rachael W. Sirianni, Sylwia A. Stopka, Sauradip Chaudhuri, and Sarah E. Stabenfeldt
- Subjects
Male ,Materials science ,Polyesters ,Antineoplastic Agents ,02 engineering and technology ,Hydrophobic effect ,Mice ,03 medical and health sciences ,chemistry.chemical_compound ,Drug Delivery Systems ,0302 clinical medicine ,Panobinostat ,convection enhanced delivery ,medicine ,Animals ,General Materials Science ,Amines ,chemistry.chemical_classification ,Cyclodextrin ,Brain Neoplasms ,nanoparticle ,HDACi ,beta-Cyclodextrins ,Nile red ,021001 nanoscience & nanotechnology ,nanomedicine ,Xenograft Model Antitumor Assays ,Histone Deacetylase Inhibitors ,Mice, Inbred C57BL ,chemistry ,Delayed-Action Preparations ,Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization ,030220 oncology & carcinogenesis ,drug delivery ,Drug delivery ,Biophysics ,Nanoparticles ,Nanomedicine ,Nanocarriers ,0210 nano-technology ,Hydrophobic and Hydrophilic Interactions ,Camptothecin ,Research Article ,medicine.drug - Abstract
Therapeutic development of histone deacetylase inhibitors (HDACi) has been hampered by a number of barriers to drug delivery, including poor solubility and inadequate tissue penetration. Nanoparticle encapsulation could be one approach to improve the delivery of HDACi to target tissues; however, effective and generalizable loading of HDACi within nanoparticle systems remains a long-term challenge. We hypothesized that the common terminally ionizable moiety on many HDACi molecules could be capitalized upon for loading in polymeric nanoparticles. Here, we describe the simple, efficient formulation of a novel library of β-cyclodextrin-poly (β-amino ester) networks (CDN) to achieve this goal. We observed that network architecture was a critical determinant of CDN encapsulation of candidate molecules, with a more hydrophobic core enabling effective self-assembly and a PEGylated surface enabling high loading (up to ∼30% w/w), effective self-assembly of the nanoparticle, and slow release of drug into aqueous media (up to 24 days) for the model HDACi panobinostat. We next constructed a library of CDNs to encapsulate various small, hydrophobic, terminally ionizable molecules (panobinostat, quisinostat, dacinostat, givinostat, bortezomib, camptothecin, nile red, and cytarabine), which yielded important insights into the structural requirements for effective drug loading and CDN self-assembly. Optimized CDN nanoparticles were taken up by GL261 cells in culture and a released panobinostat was confirmed to be bioactive. Panobinostat-loaded CDNs were next administered by convection-enhanced delivery (CED) to mice bearing intracranial GL261 tumors. These studies confirm that CDN encapsulation enables a higher deliverable dose of drug to effectively slow tumor growth. Matrix-assisted laser desorption/ionization (MALDI) analysis on tissue sections confirms higher exposure of tumor to drug, which likely accounts for the therapeutic effects. Taken in sum, these studies present a novel nanocarrier platform for encapsulation of HDACi via both ionic and hydrophobic interactions, which is an important step toward better treatment of disease via HDACi therapy.
- Published
- 2021
16. Deep Learning on Multimodal Chemical and Whole Slide Imaging Data for Predicting Prostate Cancer Directly from Tissue Images
- Author
-
Md Inzamam Ul Haque, Debangshu Mukherjee, Sylwia A. Stopka, Nathalie Y. R. Agar, Jacob Hinkle, and Olga S. Ovchinnikova
- Subjects
Structural Biology ,Spectroscopy - Abstract
Prostate cancer is one of the most common cancers globally and is the second most common cancer in the male population in the US. Here we develop a study based on correlating the H&E-stained biopsy data with MALDI mass-spectrometric imaging of the corresponding tissue to determine the cancerous regions and their unique chemical signatures, and variation of the predicted regions with original pathological annotations. We spatially register features obtained through deep learning from high-resolution optical micrographs of whole slide H&E stained data with MSI data to correlate the chemical signature with the tissue anatomy of the data, and then use the learned correlation to predict prostate cancer from observed H&E images using trained co-registered MSI data. We found that this system is more robust than predicting from a single imaging modality and can predict cancerous regions with ∼80% accuracy. Two chemical biomarkers were also found to be predicting the ground truth cancerous regions. This will improve on generating patient treatment trajectories by more accurately predicting prostate cancer directly from H&E-stained biopsy images.
- Published
- 2022
17. Overcoming differential tumor penetration of BRAF inhibitors using computationally guided combination therapy
- Author
-
Thomas S. C. Ng, Huiyu Hu, Stefan Kronister, Chanseo Lee, Ran Li, Luca Gerosa, Sylwia A. Stopka, Danielle M. Burgenske, Ishaan Khurana, Michael S. Regan, Sreeram Vallabhaneni, Niharika Putta, Ella Scott, Dylan Matvey, Anita Giobbie-Hurder, Rainer H. Kohler, Jann N. Sarkaria, Sareh Parangi, Peter K. Sorger, Nathalie Y. R. Agar, Heather A. Jacene, Ryan J. Sullivan, Elizabeth Buchbinder, Hannes Mikula, Ralph Weissleder, and Miles A. Miller
- Subjects
Multidisciplinary - Abstract
BRAF-targeted kinase inhibitors (KIs) are used to treat malignancies including BRAF-mutant non–small cell lung cancer, colorectal cancer, anaplastic thyroid cancer, and, most prominently, melanoma. However, KI selection criteria in patients remain unclear, as are pharmacokinetic/pharmacodynamic (PK/PD) mechanisms that may limit context-dependent efficacy and differentiate related drugs. To address this issue, we imaged mouse models of BRAF-mutant cancers, fluorescent KI tracers, and unlabeled drug to calibrate in silico spatial PK/PD models. Results indicated that drug lipophilicity, plasma clearance, faster target dissociation, and, in particular, high albumin binding could limit dabrafenib action in visceral metastases compared to other KIs. This correlated with retrospective clinical observations. Computational modeling identified a timed strategy for combining dabrafenib and encorafenib to better sustain BRAF inhibition, which showed enhanced efficacy in mice. This study thus offers principles of spatial drug action that may help guide drug development, KI selection, and combination.
- Published
- 2022
18. Metabolomic profiling of wild‐type and mutant soybean root nodules using laser‐ablation electrospray ionization mass spectrometry reveals altered metabolism
- Author
-
Minviluz G. Stacey, David W. Koppenaal, Sterling Evans, Ljiljana Paša-Tolić, Beverly J. Agtuca, Gary Stacey, Dong Xu, Yang Liu, Laith Z. Samarah, Christopher R. Anderton, Akos Vertes, and Sylwia A. Stopka
- Subjects
0106 biological sciences ,0301 basic medicine ,Spectrometry, Mass, Electrospray Ionization ,Root nodule ,Nitrogen ,Mutant ,Plant Science ,Biology ,01 natural sciences ,03 medical and health sciences ,chemistry.chemical_compound ,Symbiosis ,Nitrogen Fixation ,Genetics ,Plant defense against herbivory ,Metabolomics ,Bradyrhizobium ,Jasmonic acid ,Wild type ,food and beverages ,Cell Biology ,biology.organism_classification ,Carbon ,030104 developmental biology ,Biochemistry ,chemistry ,Mutation ,Nitrogen fixation ,Soybeans ,Root Nodules, Plant ,010606 plant biology & botany ,Bradyrhizobium japonicum - Abstract
The establishment of the nitrogen-fixing symbiosis between soybean and Bradyrhizobium japonicum is a complex process. To document the changes in plant metabolism as a result of symbiosis, we utilized laser ablation electrospray ionization-mass spectrometry (LAESI-MS) for in situ metabolic profiling of wild-type nodules, nodules infected with a B. japonicum nifH mutant unable to fix nitrogen, nodules doubly infected by both strains, and nodules formed on plants mutated in the stearoyl-acyl carrier protein desaturase (sacpd-c) gene, which were previously shown to have an altered nodule ultrastructure. The results showed that the relative abundance of fatty acids, purines, and lipids was significantly changed in response to the symbiosis. The nifH mutant nodules had elevated levels of jasmonic acid, correlating with signs of nitrogen deprivation. Nodules resulting from the mixed inoculant displayed similar, overlapping metabolic distributions within the sectors of effective (fix+ ) and ineffective (nifH mutant, fix- ) endosymbionts. These data are inconsistent with the notion that plant sanctioning is cell autonomous. Nodules lacking sacpd-c displayed an elevation of soyasaponins and organic acids in the central necrotic regions. The present study demonstrates the utility of LAESI-MS for high-throughput screening of plant phenotypes. Overall, nodules disrupted in the symbiosis were elevated in metabolites related to plant defense.
- Published
- 2020
19. Single-Cell Metabolic Profiling: Metabolite Formulas from Isotopic Fine Structures in Heterogeneous Plant Cell Populations
- Author
-
Dušan Veličković, Akos Vertes, Rikkita Khattar, Paola Parlanti, Beverly J. Agtuca, Christine A. Brantner, Christopher R. Anderton, Tina H Tran, Sylwia A. Stopka, Gary Stacey, Nikola Tolić, Ljiljana Paša-Tolić, Laith Z. Samarah, and Jared B. Shaw
- Subjects
Spectrometry, Mass, Electrospray Ionization ,education.field_of_study ,Electrospray ionization ,Laser ablation electrospray ionization ,Metabolite ,010401 analytical chemistry ,Population ,Oxygen Isotopes ,010402 general chemistry ,Mass spectrometry ,01 natural sciences ,Fourier transform ion cyclotron resonance ,0104 chemical sciences ,Analytical Chemistry ,chemistry.chemical_compound ,Metabolomics ,Single-cell analysis ,chemistry ,Biophysics ,Potassium Isotopes ,Bradyrhizobium ,Soybeans ,Single-Cell Analysis ,education - Abstract
Characterization of the metabolic heterogeneity in cell populations requires the analysis of single cells. Most current methods in single-cell analysis rely on cell manipulation, potentially altering the abundance of metabolites in individual cells. A small sample volume and the chemical diversity of metabolites are additional challenges in single-cell metabolomics. Here, we describe the combination of fiber-based laser ablation electrospray ionization (f-LAESI) with 21 T Fourier transform ion cyclotron resonance mass spectrometry (21TFTICR-MS) for in situ single-cell metabolic profiling in plant tissue. Single plant cells infected by bacteria were selected and sampled directly from the tissue without cell manipulation through mid-infrared ablation with a fine optical fiber tip for ionization by f-LAESI. Ultrahigh performance 21T-FTICR-MS enabled the simultaneous capture of isotopic fine structures (IFSs) for 47 known and 11 unknown compounds, thus elucidating their elemental compositions from single cells and providing information on metabolic heterogeneity in the cell population.
- Published
- 2020
20. In-Situ Metabolomic Analysis of Setaria viridis Roots Colonized by Beneficial Endophytic Bacteria
- Author
-
Akos Vertes, Dong Xu, Gary Stacey, Sterling Evans, Ljiljana Paša-Tolić, Yang Liu, Beverly J. Agtuca, Christopher R. Anderton, Thalita Regina Tuleski, Sylwia A. Stopka, Rose A. Monteiro, Fernanda Plucani do Amaral, and David W. Koppenaal
- Subjects
Rhizosphere ,biology ,Physiology ,Setaria viridis ,General Medicine ,Herbaspirillum seropedicae ,biology.organism_classification ,chemistry.chemical_compound ,chemistry ,Symbiosis ,Botany ,Nitrogen fixation ,Plant defense against herbivory ,Zeatin ,Agronomy and Crop Science ,Bacteria - Abstract
Over the past decades, crop yields have risen in parallel with increasing use of fossil fuel–derived nitrogen (N) fertilizers but with concomitant negative impacts on climate and water resources. There is a need for more sustainable agricultural practices, and biological nitrogen fixation (BNF) could be part of the solution. A variety of nitrogen-fixing, epiphytic, and endophytic plant growth–promoting bacteria (PGPB) are known to stimulate plant growth. However, compared with the rhizobium-legume symbiosis, little mechanistic information is available as to how PGPB affect plant metabolism. Therefore, we investigated the metabolic changes in roots of the model grass species Setaria viridis upon endophytic colonization by Herbaspirillum seropedicae SmR1 (fix+) or a fix− mutant strain (SmR54) compared with uninoculated roots. Endophytic colonization of the root is highly localized and, hence, analysis of whole-root segments dilutes the metabolic signature of those few cells impacted by the bacteria. Therefore, we utilized in-situ laser ablation electrospray ionization mass spectrometry to sample only those root segments at or adjacent to the sites of bacterial colonization. Metabolites involved in purine, zeatin, and riboflavin pathways were significantly more abundant in inoculated plants, while metabolites indicative of nitrogen, starch, and sucrose metabolism were reduced in roots inoculated with the fix− strain or uninoculated, presumably due to N limitation. Interestingly, compounds, involved in indole-alkaloid biosynthesis were more abundant in the roots colonized by the fix− strain, perhaps reflecting a plant defense response.
- Published
- 2020
21. massNet: integrated processing and classification of spatially resolved mass spectrometry data using deep learning for rapid tumor delineation
- Author
-
Walid M Abdelmoula, Sylwia A Stopka, Elizabeth C Randall, Michael Regan, Jeffrey N Agar, Jann N Sarkaria, William M Wells, Tina Kapur, and Nathalie Y R Agar
- Subjects
Statistics and Probability ,Biochemistry ,Original Papers ,Mass Spectrometry ,Computer Science Applications ,Machine Learning ,Computational Mathematics ,Mice ,Deep Learning ,Computational Theory and Mathematics ,Neoplasms ,Animals ,Metabolomics ,Molecular Biology - Abstract
Motivation Mass spectrometry imaging (MSI) provides rich biochemical information in a label-free manner and therefore holds promise to substantially impact current practice in disease diagnosis. However, the complex nature of MSI data poses computational challenges in its analysis. The complexity of the data arises from its large size, high-dimensionality and spectral nonlinearity. Preprocessing, including peak picking, has been used to reduce raw data complexity; however, peak picking is sensitive to parameter selection that, perhaps prematurely, shapes the downstream analysis for tissue classification and ensuing biological interpretation. Results We propose a deep learning model, massNet, that provides the desired qualities of scalability, nonlinearity and speed in MSI data analysis. This deep learning model was used, without prior preprocessing and peak picking, to classify MSI data from a mouse brain harboring a patient-derived tumor. The massNet architecture established automatically learning of predictive features, and automated methods were incorporated to identify peaks with potential for tumor delineation. The model’s performance was assessed using cross-validation, and the results demonstrate higher accuracy and a substantial gain in speed compared to the established classical machine learning method, support vector machine. Availability and implementation https://github.com/wabdelmoula/massNet. The data underlying this article are available in the NIH Common Fund’s National Metabolomics Data Repository (NMDR) Metabolomics Workbench under project id (PR001292) with http://dx.doi.org/10.21228/M8Q70T. Supplementary information Supplementary data are available at Bioinformatics online.
- Published
- 2022
22. A Druggable Addiction to de novo Pyrimidine Biosynthesis in Diffuse Midline Glioma
- Author
-
Sharmistha Pal, Jakub P. Kaplan, Huy Nguyen, Sylwia A. Stopka, Michael S. Regan, Quang-De Nguyen, Kristen L. Jones, Lisa A. Moreau, Andrew Perciaccante, Bradley Hunsel, Kevin X. Liu, Jingyu Peng, Mariella G. Filbin, Nathalie Y.R. Agar, Dipanjan Chowdhury, and Daphne Haas-Kogan
- Subjects
History ,Polymers and Plastics ,Business and International Management ,Industrial and Manufacturing Engineering - Published
- 2022
23. PARP-inhibition reprograms macrophages toward an anti-tumor phenotype
- Author
-
Lin Wang, Dan Wang, Olmo Sonzogni, Shizhong Ke, Qi Wang, Abhishek Thavamani, Felipe Batalini, Sylwia A. Stopka, Michael S. Regan, Steven Vandal, Shengya Tian, Jocelin Pinto, Andrew M. Cyr, Vanessa C. Bret-Mounet, Gerard Baquer, Hans P. Eikesdal, Min Yuan, John M. Asara, Yujing J. Heng, Peter Bai, Nathalie Y.R. Agar, and Gerburg M. Wulf
- Subjects
Adenosine Diphosphate ,Niacinamide ,Phenotype ,Cell Line, Tumor ,Macrophages ,Phthalazines ,CD47 Antigen ,Poly(ADP-ribose) Polymerase Inhibitors ,NAD ,Reactive Oxygen Species ,General Biochemistry, Genetics and Molecular Biology - Abstract
Poly(ADP)ribosylation inhibitors (PARPis) are toxic to cancer cells with homologous recombination (HR) deficiency but not to HR-proficient cells in the tumor microenvironment (TME), including tumor-associated macrophages (TAMs). As TAMs can promote or inhibit tumor growth, we set out to examine the effects of PARP inhibition on TAMs in BRCA1-related breast cancer (BC). The PARPi olaparib causes reprogramming of TAMs toward higher cytotoxicity and phagocytosis. A PARPi-related surge in NAD+ increases glycolysis, blunts oxidative phosphorylation, and induces reverse mitochondrial electron transport (RET) with an increase in reactive oxygen species (ROS) and transcriptional reprogramming. This reprogramming occurs in the absence or presence of PARP1 or PARP2 and is partially recapitulated by addition of NAD derivative methyl-nicotinamide (MNA). In vivo and ex vivo, the effect of olaparib on TAMs contributes to the anti-tumor efficacy of the PARPi. In vivo blockade of the “don’t-eat-me signal” with CD47 antibodies in combination with olaparib improves outcomes in a BRCA1-related BC model. publishedVersion
- Published
- 2022
24. Characterization of metabolic compartmentalization in the liver using spatially resolved metabolomics
- Author
-
Jiska van der Reest, Sylwia A. Stopka, Walid M. Abdelmoula, Daniela F. Ruiz, Shakchhi Joshi, Alison E. Ringel, Marcia C. Haigis, and Nathalie Y. R. Agar
- Abstract
Cells adapt their metabolism to physiological stimuli, and metabolic heterogeneity exists between cell types, within tissues, and subcellular compartments. The liver plays an essential role in maintaining whole-body metabolic homeostasis and is structurally defined by metabolic zones. These zones are well-understood on the transcriptomic level, but have not been comprehensively characterized on the metabolomic level. Mass spectrometry imaging (MSI) can be used to map hundreds of metabolites directly from a tissue section, offering an important advance to investigate metabolic heterogeneity in tissues compared to extraction-based metabolomics methods that analyze tissue metabolite profiles in bulk. We established a workflow for the preparation of tissue specimens for matrix-assisted laser desorption/ionization (MALDI) MSI and achieved broad coverage of central carbon, nucleotide, and lipid metabolism pathways. We used this approach to visualize the effect of nutrient stress and excess on liver metabolism. Our data revealed a highly organized metabolic compartmentalization in livers, which becomes disrupted under nutrient stress conditions. Fasting caused changes in glucose metabolism and increased the levels of fatty acids in the circulation. In contrast, a prolonged high-fat diet (HFD) caused lipid accumulation within liver tissues with clear zonal patterns. Fatty livers had higher levels of purine and pentose phosphate related metabolites, which generates reducing equivalents to counteract oxidative stress. This MALDI MSI approach allowed the visualization of liver metabolic compartmentalization at high resolution and can be applied more broadly to yield new insights into metabolic heterogeneityin vivo.
- Published
- 2021
25. Spatially resolved characterization of tissue metabolic compartments in fasted and high-fat diet livers
- Author
-
Sylwia A. Stopka, Jiska van der Reest, Walid M. Abdelmoula, Daniela F. Ruiz, Shakchhi Joshi, Alison E. Ringel, Marcia C. Haigis, and Nathalie Y. R. Agar
- Subjects
Multidisciplinary ,Liver ,Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization ,Metabolomics ,Fasting ,Diet, High-Fat - Abstract
Cells adapt their metabolism to physiological stimuli, and metabolic heterogeneity exists between cell types, within tissues, and subcellular compartments. The liver plays an essential role in maintaining whole-body metabolic homeostasis and is structurally defined by metabolic zones. These zones are well-understood on the transcriptomic level, but have not been comprehensively characterized on the metabolomic level. Mass spectrometry imaging (MSI) can be used to map hundreds of metabolites directly from a tissue section, offering an important advance to investigate metabolic heterogeneity in tissues compared to extraction-based metabolomics methods that analyze tissue metabolite profiles in bulk. We established a workflow for the preparation of tissue specimens for matrix-assisted laser desorption/ionization (MALDI) MSI that can be implemented to achieve broad coverage of central carbon, nucleotide, and lipid metabolism pathways. Herein, we used this approach to visualize the effect of nutrient stress and excess on liver metabolism. Our data revealed a highly organized metabolic tissue compartmentalization in livers, which becomes disrupted under high fat diet. Fasting caused changes in the abundance of several metabolites, including increased levels of fatty acids and TCA intermediates while fatty livers had higher levels of purine and pentose phosphate-related metabolites, which generate reducing equivalents to counteract oxidative stress. This spatially conserved approach allowed the visualization of liver metabolic compartmentalization at 30 μm pixel resolution and can be applied more broadly to yield new insights into metabolic heterogeneity in vivo.
- Published
- 2021
26. Treatment of Recurrent Glioblastoma by Chronic Convection-Enhanced Delivery of Topotecan
- Author
-
Eleonora F. Spinazzi, Michael G. Argenziano, Pavan S. Upadhyayula, Matei A. Banu, Justin A. Neira, Dominique M.O. Higgins, Peter B. Wu, Brianna Pereira, Aayushi Mahajan, Nelson Humala, Osama Al-Dalahmah, Wenting Zhao, Akshay V. Save, Deborah M. Boyett, Tamara Marie, Julia L Furnari, Tejaswi D. Sudhakar, Sylwia A. Stopka, Michael S. Regan, Vanessa Catania, Laura Good, Meenu Behl, Sachin Jambawalikar, Akiva Mintz, Angela Lignelli, Nathalie Y.R. Agar, Peter A. Sims, Mary Welch, Andrew Lassman, Fabio Iwamoto, Randy S. D’Amico, Jack Grinband, Peter Canoll, and Jeffrey N. Bruce
- Abstract
Glioblastoma, the most common primary brain malignancy, is invariably fatal. Systemic chemotherapy is ineffective mostly because of drug delivery limitations. To overcome this, we devised an internalized pump-catheter system for direct chronic convection-enhanced delivery (CED) into peritumoral brain tissue. Topotecan (TPT) by chronic CED in 5 patients with refractory glioblastoma selectively eliminated tumor cells without toxicity to normal brain. Large, stable drug distribution volumes were non-invasively monitored with MRI of co-infused gadolinium. Analysis of multiple radiographically localized biopsies taken before and after treatment showed a decreased proliferative tumor signature resulting in a shift to a slow-cycling mesenchymal/astrocytic-like population. Tumor microenvironment analysis showed an inflammatory response and preservation of neurons. This novel drug delivery strategy and innovative clinical trial paradigm overcomes current limitations in delivery and treatment response assessment as shown here for glioblastoma and is potentially applicable for other anti-glioma agents as well as other CNS diseases.
- Published
- 2021
27. De Novo Pyrimidine Synthesis is a Targetable Vulnerability in IDH Mutant Glioma
- Author
-
Januka Khanal, Daniel P. Cahill, Joseph Buehler, Diana D. Shi, Yu-Fen Lin, Misty S. Martin-Sandoval, Michael M Levitt, Keith L. Ligon, Harley I. Kornblum, Adam C. Wang, Sungwoo Lee, Dennis M. Bonal, John M. Asara, Mark A. Lehrman, Isaac S. Harris, Andreas Janzer, Cylaina E. Bird, Michael Jeffers, Ralph J. DeBerardinis, William G. Kaelin, Tammie Dang, Nathalie Y. R. Agar, Sylwia A. Stopka, Lauren C. Gattie, Stefan Gradl, Lauren G. Zacharias, Michael S. Regan, Quang-Dé Nguyen, Tak W. Mak, Sabina Signoretti, Kalil G. Abdullah, Peter Ly, Andreas Sutter, Timothy E. Richardson, Samuel K. McBrayer, Thomas P. Mathews, Milan R. Savani, Jennifer E. Endress, Min Xu, Wenhua Gao, Bofu Huang, Rebecca B. Jennings, and Ryan E. Looper
- Subjects
Purine ,IDH1 ,Mutant ,Synthetic lethality ,Biology ,medicine.disease ,chemistry.chemical_compound ,Isocitrate dehydrogenase ,chemistry ,Glioma ,Pyrimidine metabolism ,Cancer research ,medicine ,Dihydroorotate dehydrogenase - Abstract
SUMMARYMutations affecting isocitrate dehydrogenase (IDH) enzymes are prevalent in glioma, leukemia, and other cancers. Although mutant IDH inhibitors are effective against leukemia, they appear less active in aggressive glioma, underscoring the need for alternative treatment strategies. Through a chemical synthetic lethality screen, we discovered that IDH1 mutant glioma cells are hypersensitive to drugs targeting enzymes in the de novo pyrimidine nucleotide synthesis pathway, including dihydroorotate dehydrogenase (DHODH). We developed a genetically engineered mouse model of mutant IDH1-driven astrocytoma and used it and multiple patient-derived models to show that the brain-penetrant DHODH inhibitor BAY 2402234 displays monotherapy efficacy against IDH mutant gliomas. Mechanistically, this vulnerability selectively applies to de novo pyrimidine, but not purine, synthesis because glioma cells engage disparate programs to produce these nucleotide species and because IDH oncogenes increase DNA damage upon nucleotide pool imbalance. Our work outlines a tumor-selective, biomarker-guided therapeutic strategy that is poised for clinical translation.
- Published
- 2021
28. A druggable addiction to de novo pyrimidine biosynthesis in diffuse midline glioma
- Author
-
Sharmistha Pal, Jakub P. Kaplan, Huy Nguyen, Sylwia A. Stopka, Milan R. Savani, Michael S. Regan, Quang-De Nguyen, Kristen L. Jones, Lisa A. Moreau, Jingyu Peng, Marina G. Dipiazza, Andrew J. Perciaccante, Xiaoting Zhu, Bradley R. Hunsel, Kevin X. Liu, Rachid Drissi, Mariella G. Filbin, Samuel K. McBrayer, Nathalie Y.R. Agar, Dipanjan Chowdhury, and Daphne Haas-Kogan
- Subjects
De novo synthesis ,chemistry.chemical_compound ,In vivo ,DNA damage ,Chemistry ,Apoptosis ,Glioma ,Pyrimidine metabolism ,medicine ,Cancer research ,medicine.disease ,Pediatric cancer ,Uridine - Abstract
SUMMARYDiffuse midline glioma (DMG) is a uniformly fatal pediatric cancer driven by oncohistones that do not readily lend themselves to drug development. To identify druggable targets for DMG, we conducted a genome-wide CRISPR screen that reveals a DMG selective dependency on the de novo pathway for pyrimidine biosynthesis. This metabolic vulnerability reflects an elevated rate of uridine/uracil degradation that depletes DMG cells of substrates for the alternate salvage pathway for pyrimidine biosynthesis. A clinical stage inhibitor of DHODH (rate limiting enzyme in the de novo pathway) diminishes UMP pools, generates DNA damage, and induces apoptosis through suppression of replication forks--an “on target” effect, as shown by uridine rescue. MALDI mass spectroscopy imaging demonstrates that this DHODH inhibitor (BAY2402234) accumulates in brain at therapeutically relevant concentrations, suppresses de novo pyrimidine biosynthesis in vivo, and prolongs survival of mice bearing intracranial DMG xenografts, highlighting BAY2402234 as a promising therapy against DMGs.
- Published
- 2021
29. A non-dividing cell population with high pyruvate dehydrogenase kinase activity regulates metabolic heterogeneity and tumorigenesis in the intestine
- Author
-
Carlos Sebastian, Christina Ferrer, Maria Serra, Jee-Eun Choi, Nadia Ducano, Alessia Mira, Manasvi S. Shah, Sylwia A. Stopka, Andrew J. Perciaccante, Claudio Isella, Daniel Moya-Rull, Marianela Vara-Messler, Silvia Giordano, Elena Maldi, Niyati Desai, Diane E. Capen, Enzo Medico, Murat Cetinbas, Ruslan I. Sadreyev, Dennis Brown, Miguel N. Rivera, Anna Sapino, David T. Breault, Nathalie Y. R. Agar, and Raul Mostoslavsky
- Subjects
Animals ,Cell Transformation, Neoplastic ,Glycolysis ,Intestines ,Mice ,Pyruvate Dehydrogenase Acetyl-Transferring Kinase ,Neoplasms ,Sirtuins ,Neoplastic ,Multidisciplinary ,General Physics and Astronomy ,General Chemistry ,Cell Transformation ,General Biochemistry, Genetics and Molecular Biology - Abstract
Although reprogramming of cellular metabolism is a hallmark of cancer, little is known about how metabolic reprogramming contributes to early stages of transformation. Here, we show that the histone deacetylase SIRT6 regulates tumor initiation during intestinal cancer by controlling glucose metabolism. Loss of SIRT6 results in an increase in the number of intestinal stem cells (ISCs), which translates into enhanced tumor initiating potential in APCmin mice. By tracking down the connection between glucose metabolism and tumor initiation, we find a metabolic compartmentalization within the intestinal epithelium and adenomas, where a rare population of cells exhibit features of Warburg-like metabolism characterized by high pyruvate dehydrogenase kinase (PDK) activity. Our results show that these cells are quiescent cells expressing +4 ISCs and enteroendocrine markers. Active glycolysis in these cells suppresses ROS accumulation and enhances their stem cell and tumorigenic potential. Our studies reveal that aerobic glycolysis represents a heterogeneous feature of cancer, and indicate that this metabolic adaptation can occur in non-dividing cells, suggesting a role for the Warburg effect beyond biomass production in tumors.
- Published
- 2021
30. Multiplatform Metabolomics Studies of Human Cancers With NMR and Mass Spectrometry Imaging
- Author
-
Anya B. Zhong, Isabella H. Muti, Stephen J. Eyles, Richard W. Vachet, Kristen N. Sikora, Cedric E. Bobst, David Calligaris, Sylwia A. Stopka, Jeffery N. Agar, Chin-Lee Wu, Mari A. Mino-Kenudson, Nathalie Y. R. Agar, David C. Christiani, Igor A. Kaltashov, and Leo L. Cheng
- Subjects
Biochemistry, Genetics and Molecular Biology (miscellaneous) ,Molecular Biology ,Biochemistry - Abstract
The status of metabolomics as a scientific branch has evolved from proof-of-concept to applications in science, particularly in medical research. To comprehensively evaluate disease metabolomics, multiplatform approaches of NMR combining with mass spectrometry (MS) have been investigated and reported. This mixed-methods approach allows for the exploitation of each individual technique’s unique advantages to maximize results. In this article, we present our findings from combined NMR and MS imaging (MSI) analysis of human lung and prostate cancers. We further provide critical discussions of the current status of NMR and MS combined human prostate and lung cancer metabolomics studies to emphasize the enhanced metabolomics ability of the multiplatform approach.
- Published
- 2021
31. TMET-07. MOLECULAR MECHANISM OF EXQUISITE SENSITIVITY OF DIFFUSE MIDLINE GLIOMA TO DE NOVO PYRIMIDINE BIOSYNTHESIS
- Author
-
Sharmistha Pal, Milan Savani, Jakub Kaplan, Sylwia Sylwia A Stopka, Huy Nguyen, Michael Regan, Nathalie Agar, Samuel McBrayer, and Daphne Haas-Kogan
- Subjects
Cancer Research ,Oncology ,Neurology (clinical) - Abstract
Diffuse midline glioma (DMG) is a lethal pediatric brain cancer in dire need for therapeutic breakthroughs. To identify intrinsic addictions and therapeutic targets for DMG, we conducted a genome-wide k¬¬¬¬nockout CRISPR screen that identified de novo pyrimidine biosynthesis pathway as a targetable dependency. Since pyrimidine nucleotides can be synthesized by salvage and de novo pathways, identification of the latter as a dependency indicated a prominent role of de novo pathway in establishing the pyrimidine nucleotide pool in DMG. We investigated the molecular mechanism underlying this dependency using metabolic tracing of 15N-glutamine and genetic approaches. We report that DMG cells derive the majority of UMP (70%) through de novo synthesis, along with significantly elevated flux through the pyrimidine degradation pathway. High flux of 15N- labeled UMP through pyrimidine degradation in DMGs suggests that substrates (uridine and uracil) required for pyrimidine nucleotide salvage are limited, thus enhancing the dependency on de novo biosynthesis. To further confirm the causal role of pyrimidine degradation in driving de novo pathway dependency in DMG, we knocked down DPYD, the enzyme catalyzing the first committed step in pyrimidine degradation, using inducible shRNAs. Knockdown of DPYD diminished sensitivity of DMGs to de novo pathway inhibition as it rescued UMP pools and resultant DNA damage. Conversely, overexpression of DPYD in adult glioblastomas enhanced their sensitivity to de novo pyrimidine synthesis inhibition which was accompanied by greater depletion of UMP and induction of DNA damage. Consistent with this mechanism, we observed downregulation of DPYD in DMGs that acquired resistance to antagonists of de novo pyrimidine synthesis. Taken together, we have uncovered DPYD as a predictive biomarker of sensitivity to de novo pyrimidine synthesis inhibitors.
- Published
- 2022
32. De novo pyrimidine synthesis is a targetable vulnerability in IDH mutant glioma
- Author
-
Diana D. Shi, Milan R. Savani, Michael M. Levitt, Adam C. Wang, Jennifer E. Endress, Cylaina E. Bird, Joseph Buehler, Sylwia A. Stopka, Michael S. Regan, Yu-Fen Lin, Vinesh T. Puliyappadamba, Wenhua Gao, Januka Khanal, Laura Evans, Joyce H. Lee, Lei Guo, Yi Xiao, Min Xu, Bofu Huang, Rebecca B. Jennings, Dennis M. Bonal, Misty S. Martin-Sandoval, Tammie Dang, Lauren C. Gattie, Amy B. Cameron, Sungwoo Lee, John M. Asara, Harley I. Kornblum, Tak W. Mak, Ryan E. Looper, Quang-De Nguyen, Sabina Signoretti, Stefan Gradl, Andreas Sutter, Michael Jeffers, Andreas Janzer, Mark A. Lehrman, Lauren G. Zacharias, Thomas P. Mathews, Julie-Aurore Losman, Timothy E. Richardson, Daniel P. Cahill, Ralph J. DeBerardinis, Keith L. Ligon, Lin Xu, Peter Ly, Nathalie Y.R. Agar, Kalil G. Abdullah, Isaac S. Harris, William G. Kaelin, and Samuel K. McBrayer
- Subjects
Mice ,Cancer Research ,Leukemia ,Pyrimidines ,Oncology ,Brain Neoplasms ,Mutation ,Animals ,Glioma ,Enzyme Inhibitors ,Triazoles ,Salicylanilides ,Isocitrate Dehydrogenase - Abstract
Mutations affecting isocitrate dehydrogenase (IDH) enzymes are prevalent in glioma, leukemia, and other cancers. Although mutant IDH inhibitors are effective against leukemia, they seem to be less active in aggressive glioma, underscoring the need for alternative treatment strategies. Through a chemical synthetic lethality screen, we discovered that IDH1-mutant glioma cells are hypersensitive to drugs targeting enzymes in the de novo pyrimidine nucleotide synthesis pathway, including dihydroorotate dehydrogenase (DHODH). We developed a genetically engineered mouse model of mutant IDH1-driven astrocytoma and used it and multiple patient-derived models to show that the brain-penetrant DHODH inhibitor BAY 2402234 displays monotherapy efficacy against IDH-mutant gliomas. Mechanistically, this reflects an obligate dependence of glioma cells on the de novo pyrimidine synthesis pathway and mutant IDH's ability to sensitize to DNA damage upon nucleotide pool imbalance. Our work outlines a tumor-selective, biomarker-guided therapeutic strategy that is poised for clinical translation.
- Published
- 2022
33. Multimodal platform for assessing drug distribution and response in clinical trials
- Author
-
Giorgio Gaglia, Ziming Du, Louis B. Nabors, Yang Dai, Ishwar N. Kohale, Ilya Korsunsky, Brian M. Alexander, Jann N. Sarkaria, Eudocia Q. Lee, Patrick Y. Wen, Michael S. Regan, Sankha S. Basu, Soumya Raychaudhuri, Elizabeth C. Randall, Keith L. Ligon, Bianca-Maria Marin, Forest M. White, Ann C. Mladek, Sandro Santagata, Nathalie Y. R. Agar, Stuart A. Grossman, Danielle M. Burgenske, Jeffrey G. Supko, Jeffrey N. Agar, Amanda R Clark, Sylwia A. Stopka, Walid M. Abdelmoula, and Begoña Gimenez-Cassina Lopez
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,business.industry ,Clinical study design ,medicine.medical_treatment ,Phosphoproteomics ,Phases of clinical research ,Targeted therapy ,Clinical trial ,Efficacy ,Drug development ,Pharmaceutical Preparations ,Molecular Response ,Internal medicine ,Basic and Translational Investigations ,medicine ,Humans ,Neurology (clinical) ,business ,Glioblastoma - Abstract
Background Response to targeted therapy varies between patients for largely unknown reasons. Here, we developed and applied an integrative platform using mass spectrometry imaging (MSI), phosphoproteomics, and multiplexed tissue imaging for mapping drug distribution, target engagement, and adaptive response to gain insights into heterogeneous response to therapy. Methods Patient-derived xenograft (PDX) lines of glioblastoma were treated with adavosertib, a Wee1 inhibitor, and tissue drug distribution was measured with MALDI-MSI. Phosphoproteomics was measured in the same tumors to identify biomarkers of drug target engagement and cellular adaptive response. Multiplexed tissue imaging was performed on sister sections to evaluate spatial co-localization of drug and cellular response. The integrated platform was then applied on clinical specimens from glioblastoma patients enrolled in the phase 1 clinical trial. Results PDX tumors exposed to different doses of adavosertib revealed intra- and inter-tumoral heterogeneity of drug distribution and integration of the heterogeneous drug distribution with phosphoproteomics and multiplexed tissue imaging revealed new markers of molecular response to adavosertib. Analysis of paired clinical specimens from patients enrolled in the phase 1 clinical trial informed the translational potential of the identified biomarkers in studying patient’s response to adavosertib. Conclusions The multimodal platform identified a signature of drug efficacy and patient-specific adaptive responses applicable to preclinical and clinical drug development. The information generated by the approach may inform mechanisms of success and failure in future early phase clinical trials, providing information for optimizing clinical trial design and guiding future application into clinical practice.
- Published
- 2021
34. Single cell spatial analysis reveals the topology of immunomodulatory purinergic signaling in glioblastoma
- Author
-
Shannon Coy, Shu Wang, Sylwia A. Stopka, Jia-Ren Lin, Clarence Yapp, Cecily C. Ritch, Lisa Salhi, Gregory J. Baker, Rumana Rashid, Gerard Baquer, Michael Regan, Prasidda Khadka, Kristina A. Cole, Jaeho Hwang, Patrick Y. Wen, Pratiti Bandopadhayay, Mariarita Santi, Thomas De Raedt, Keith L. Ligon, Nathalie Y. R. Agar, Peter K. Sorger, Mehdi Touat, and Sandro Santagata
- Subjects
Spatial Analysis ,Multidisciplinary ,Adenosine ,Tumor Microenvironment ,General Physics and Astronomy ,Humans ,General Chemistry ,Glioma ,Single-Cell Analysis ,Child ,Glioblastoma ,5'-Nucleotidase ,General Biochemistry, Genetics and Molecular Biology - Abstract
How the glioma immune microenvironment fosters tumorigenesis remains incompletely defined. Here, we use single-cell RNA-sequencing and multiplexed tissue-imaging to characterize the composition, spatial organization, and clinical significance of extracellular purinergic signaling in glioma. We show that microglia are the predominant source of CD39, while tumor cells principally express CD73. In glioblastoma, CD73 is associated with EGFR amplification, astrocyte-like differentiation, and increased adenosine, and is linked to hypoxia. Glioblastomas enriched for CD73 exhibit inflammatory microenvironments, suggesting that purinergic signaling regulates immune adaptation. Spatially-resolved single-cell analyses demonstrate a strong spatial correlation between tumor-CD73 and microglial-CD39, with proximity associated with poor outcomes. Similar spatial organization is present in pediatric high-grade gliomas including H3K27M-mutant diffuse midline glioma. These data reveal that purinergic signaling in gliomas is shaped by genotype, lineage, and functional state, and that core enzymes expressed by tumor and myeloid cells are organized to promote adenosine-rich microenvironments potentially amenable to therapeutic targeting.
- Published
- 2021
35. Abstract 2322: Multiplatform metabolomics studies of human cancers with NMR and mass spectrometry imaging
- Author
-
Leo L. Cheng, Anya B. Zhong, Isabella H. Muti, Stephen J. Eyles, Richard W. Vachet, Sylwia A. Stopka, Kristen N. Sikora, Cedric E. Bobst, Jeffrey N. Agar, Mari A. Mino-Kenudson, Chin-Lee Wu, David C. Christiani, Igor A. Kaltashov, and Nathalie Y. Agar
- Subjects
Cancer Research ,Oncology - Abstract
Unfortunately, at present, there is no single technique that possesses all the characteristics needed to be considered an ideal global metabolite profiling tool. Thus, the use of multiple analytical platforms, such as combining the strengths of Mass spectrometry (MS) and nuclear magnetic resonance spectroscopy (NMR), for metabolic profiling can maximize coverage and generate more global metabolomic profiles. In this study, we demonstrate the utilities of the combined NMR and MSI multiplatform in our metabolomics results on human prostate and lung cancers. Statistical data on the natural history of prostate cancer (PCa) show that >70% of patients diagnosed by PSA screening will likely experience indolent disease with little impact on well-being. For about 17% of newly PSA-diagnosed patients, however, aggressive PCa proliferation ensues, truncating life expectancy. At present, no reliable clinical test can differentiate between these two groups. Using HRMAS 1HNMR followed by quantitative histology, we showed statistically significant correlations between concentrations of Spm and the amount of histologically-benign epithelial (Hb Epi) prostatic cells and glands in human cancerous prostates. However, as above discussed that using HRMAS NMR alone we cannot prove that Spm was indeed generated or resided in the Hb Epi cells. Nevertheless, using MALDI MSI, we were able to locate Spm (m/z: 203.223 ± 0.001Da) onto Hb Epi, where spermine on the PCa lesions appeared below detection limits. From these maps, for the first time, we could visualize and confirm the differential localizations of Spm in prostates. This proof of Sym relationship to prostate pathologies and its proposed PCa inhibitory effects may support further studies that are critical in differentiating aggressive from indolent PCa for disease evaluations and patient personalized treatment strategies. To search for such screening metabolomics biomarkers in lung cancer, we used HRMAS NMR to analyze 93 pairs of human LuCa tissue and serum samples, and 29 healthy human sera. A number of potential metabolite candidates capable to differentiate LuCa characteristics were identified, including glutamate, lipids, alanine, glycerylphosphorylcholine, glutamine, phosphorylcholine, etc. This list can be further expanded by analyzing metabolite composition in the serum of cancer patients and control healthy subjects using LC-MS, which offers a dramatic increase in sensitivity compared to HRMAS NMR and, therefore, is better suited for the biomarker discovery. In addition to acquiring high-resolution mass data, the high data acquisition rate allows the fragment ion mass spectra (MS/MS) to be generated for the most abundant ionic species in each chromatographic peak. This feature allows specific classes of tumor-attenuated metabolites to be identified based on the presence of unique structurally diagnostic fragment ions in MS/MS spectra. Citation Format: Leo L. Cheng, Anya B. Zhong, Isabella H. Muti, Stephen J. Eyles, Richard W. Vachet, Sylwia A. Stopka, Kristen N. Sikora, Cedric E. Bobst, Jeffrey N. Agar, Mari A. Mino-Kenudson, Chin-Lee Wu, David C. Christiani, Igor A. Kaltashov, Nathalie Y. Agar. Multiplatform metabolomics studies of human cancers with NMR and mass spectrometry imaging [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 2322.
- Published
- 2022
36. massNet: integrated processing and classification of spatially resolved mass spectrometry data using deep learning for rapid tumor delineation
- Author
-
Elizabeth C. Randall, Jeffrey N. Agar, Tina Kapur, Sylwia A. Stopka, Jann N. Sarkaria, William M. Wells, Nathalie Y. R. Agar, Walid M. Abdelmoula, and Michael S. Regan
- Subjects
Support vector machine ,Text mining ,business.industry ,Computer science ,Deep learning ,Scalability ,Code (cryptography) ,Preprocessor ,Pattern recognition ,Artificial intelligence ,Raw data ,business ,Mass spectrometry imaging - Abstract
MotivationMass spectrometry imaging (MSI) provides rich biochemical information in a label-free manner and therefore holds promise to substantially impact current practice in disease diagnosis. However, the complex nature of MSI data poses computational challenges in its analysis. The complexity of the data arises from its large size, high dimensionality, and spectral non-linearity. Preprocessing, including peak picking, has been used to reduce raw data complexity, however peak picking is sensitive to parameter selection that, perhaps prematurely, shapes the downstream analysis for tissue classification and ensuing biological interpretation.ResultsWe propose a deep learning model, massNet, that provides the desired qualities of scalability, non-linearity, and speed in MSI data analysis. This deep learning model was used, without prior preprocessing and peak picking, to classify MSI data from a mouse brain harboring a patient-derived tumor. The massNet architecture established automatically learning of predictive features, and automated methods were incorporated to identify peaks with potential for tumor delineation. The model’s performance was assessed using cross-validation, and the results demonstrate higher accuracy and a 174-fold gain in speed compared to the established classical machine learning method, support vector machine.Availability and ImplementationThe code is publicly available on GitHub.
- Published
- 2021
37. Neuropeptide Localization in
- Author
-
Ellen A, Wood, Sylwia A, Stopka, Linwen, Zhang, Sara, Mattson, Gabor, Maasz, Zsolt, Pirger, and Akos, Vertes
- Subjects
Lymnaea stagnalis ,Review ,central nervous system ,neuropeptide ,single neuron analysis ,Neuroscience ,mass spectrometry - Abstract
Due to the relatively small number of neurons (few tens of thousands), the well-established multipurpose model organism Lymnaea stagnalis, great pond snail, has been extensively used to study the functioning of the nervous system. Unlike the more complex brains of higher organisms, L. stagnalis has a relatively simple central nervous system (CNS) with well-defined circuits (e.g., feeding, locomotion, learning, and memory) and identified individual neurons (e.g., cerebral giant cell, CGC), which generate behavioral patterns. Accumulating information from electrophysiological experiments maps the network of neuronal connections and the neuronal circuits responsible for basic life functions. Chemical signaling between synaptic-coupled neurons is underpinned by neurotransmitters and neuropeptides. This review looks at the rapidly expanding contributions of mass spectrometry (MS) to neuropeptide discovery and identification at different granularity of CNS organization. Abundances and distributions of neuropeptides in the whole CNS, eleven interconnected ganglia, neuronal clusters, single neurons, and subcellular compartments are captured by MS imaging and single cell analysis techniques. Combining neuropeptide expression and electrophysiological data, and aided by genomic and transcriptomic information, the molecular basis of CNS-controlled biological functions is increasingly revealed.
- Published
- 2021
38. Heterogeneous delivery across the blood-brain barrier limits the efficacy of an EGFR-targeting antibody drug conjugate in glioblastoma
- Author
-
Michael S. Regan, Jeanette E. Eckel-Passow, Jennifer R. Cochran, Sonia Jain, Gaspar J. Kitange, Katrina K. Bakken, William F. Elmquist, Paul A. Decker, Minjee Kim, Caterina Giannini, Ian F. Parney, Rachael A. Vaubel, Ann C. Mladek, Brett L. Carlson, Terence C. Burns, Bianca-Maria Marin, Caitlyn L. Miller, Madison H McMinn, Sylwia A. Stopka, Jann N. Sarkaria, Nathalie Y. R. Agar, Danielle M. Burgenske, Forest M. White, Kendra A Porath, Jason E Conage-Pough, Juhee Oh, Surabhi Talele, Shulan Tian, Shiv K. Gupta, Lihong He, Marin B.-M., Porath K.A., Jain S., Kim M., Conage-Pough J.E., Oh J.-H., Miller C.L., Talele S., Kitange G.J., Tian S., Burgenske D.M., Mladek A.C., Gupta S.K., Decker P.A., McMinn M.H., Stopka S.A., Regan M.S., He L., Carlson B.L., Bakken K., Burns T.C., Parney I.F., Giannini C., Agar N.Y.R., Eckel-Passow J.E., Cochran J.R., Elmquist W.F., Vaubel R.A., White F.M., and Sarkaria J.N.
- Subjects
0301 basic medicine ,Drug ,Cancer Research ,Antibody-drug conjugate ,Immunoconjugates ,media_common.quotation_subject ,EGFR ,Brain tumor ,Blood–brain barrier ,Antibodies, Monoclonal, Humanized ,Depatux-M ,Depatuxizumab mafodotin ,Brain Neoplasm ,03 medical and health sciences ,Drug Delivery Systems ,0302 clinical medicine ,Antibody drug conjugate ,Cell Line, Tumor ,medicine ,Humans ,Distribution (pharmacology) ,Epidermal growth factor receptor ,ErbB Receptor ,media_common ,biology ,Brain Neoplasms ,business.industry ,medicine.disease ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,Immunoconjugate ,Pharmaceutical Preparations ,Blood-Brain Barrier ,030220 oncology & carcinogenesis ,Basic and Translational Investigations ,Drug delivery ,Cancer research ,biology.protein ,Neurology (clinical) ,business ,Glioblastoma ,Human - Abstract
Background Antibody drug conjugates (ADCs) targeting the epidermal growth factor receptor (EGFR), such as depatuxizumab mafodotin (Depatux-M), is a promising therapeutic strategy for glioblastoma (GBM) but recent clinical trials did not demonstrate a survival benefit. Understanding the mechanisms of failure for this promising strategy is critically important. Methods PDX models were employed to study efficacy of systemic vs intracranial delivery of Depatux-M. Immunofluorescence and MALDI-MSI were performed to detect drug levels in the brain. EGFR levels and compensatory pathways were studied using quantitative flow cytometry, Western blots, RNAseq, FISH, and phosphoproteomics. Results Systemic delivery of Depatux-M was highly effective in nine of 10 EGFR-amplified heterotopic PDXs with survival extending beyond one year in eight PDXs. Acquired resistance in two PDXs (GBM12 and GBM46) was driven by suppression of EGFR expression or emergence of a novel short-variant of EGFR lacking the epitope for the Depatux-M antibody. In contrast to the profound benefit observed in heterotopic tumors, only two of seven intrinsically sensitive PDXs were responsive to Depatux-M as intracranial tumors. Poor efficacy in orthotopic PDXs was associated with limited and heterogeneous distribution of Depatux-M into tumor tissues, and artificial disruption of the BBB or bypass of the BBB by direct intracranial injection of Depatux-M into orthotopic tumors markedly enhanced the efficacy of drug treatment. Conclusions Despite profound intrinsic sensitivity to Depatux-M, limited drug delivery into brain tumor may have been a key contributor to lack of efficacy in recently failed clinical trials.
- Published
- 2021
39. Abstract 1816: Phenogenomic characterization of immunomodulatory purinergic signaling in glioblastoma
- Author
-
Jaeho Hwang, Philipp Euskirchen, Sandro Santagata, Nathalie Y. R. Agar, Sylwia A. Stopka, Shannon Coy, Jia-Ren Lin, Mehdi Touat, Prasidda Khadka, Peter K. Sorger, Shu Wang, Keith L. Ligon, Rumana Rashid, Patrick Y. Wen, and Pratiti Bandopadhayay
- Subjects
Cancer Research ,Oncology ,Chemistry ,Cancer research ,medicine ,Purinergic signalling ,medicine.disease ,Glioblastoma - Abstract
INTRODUCTION: Extracellular purinergic signaling plays critical roles in the regulation of tumor growth and anti-tumor immunity via autocrine/paracrine binding of metabolites to receptors on neoplastic and non-neoplastic populations. Extracellular purine concentrations are principally mediated by the ectonucleotidase enzymes CD39 and CD73, which catabolize ATP to adenosine. Within the tumor microenvironment, neoplastic, immune, and stromal cells expressing these enzymes may co-localize to generate an immunosuppressive adenosine-rich niche. However, the cellular composition, spatial architecture and phenotypic properties of these tumor ecosystems and their relationship to tumor genotype have been poorly characterized. METHODS: We quantified CD73 expression by immunohistochemistry (IHC) in a cohort of CNS tumors [meningiomas(N=222), gliomas(N=244), ependymomas(N=44), medulloblastomas(N=24), craniopharyngiomas(N=38)]. We used publicly-available single-cell RNA-seq data and 36 marker multiplexed tissue imaging (t-CyCIF) of 139 clinically and genomically annotated glioblastomas to characterize CD39 and CD73 expressing populations, define immune architecture and tumor cell states at single cell resolution, evaluate spatial correlations, and identify markers of clinical outcome. Mass spectrometry imaging (MALDI-MSI) was employed to generate spatially-resolved quantification of purine metabolite levels in glioblastoma resections (N=9). RESULTS: IHC revealed strong CD73 expression in meningiomas and gliomas. Tumor CD73 expression was associated with poor progression-free-survival in IDH-wildtype glioblastoma (p=0.04). scRNA-seq in glioblastoma revealed that CD73 is predominantly expressed by tumor cell populations, while CD39 is predominantly expressed by monocytic (macrophage, microglial) populations. t-CyCIF showed enrichment of EGFR, Ki-67, and TP53 expression in CD73-high tumor cells at a single cell level independent of genotype, as well as significant spatial correlation between CD73 expression in tumor cells and CD39 expression in macrophages. MALDI-MSI showed significantly greater adenosine concentrations in glioblastomas with high CD73 expression. CD73 expression significantly correlated with EGFR amplification or C-terminal deletion (EGFRvIII or variants), type-II interferon signaling, and PD-L1 expression in glioblastoma. CONCLUSIONS: Phenogenomic analysis of purinergic signaling in glioblastoma revealed correlations between CD73 expression and genotype, adenosine concentration, and clinical outcome. Spatial analysis revealed interaction between macrophages CD39 expression and tumor cell CD73 expression, suggesting that these populations may interact to suppress anti-tumor immunity. Anti-CD73 therapy may provide therapeutic benefits in glioblastoma by blunting immunosuppressive and oncogenic adenosine signaling. Citation Format: Shannon Coy, Jia-Ren Lin, Shu Wang, Sylwia Stopka, Rumana Rashid, Jaeho Hwang, Prasidda Khadka, Philipp Euskirchen, Pratiti Bandopadhayay, Patrick Y. Wen, Peter K. Sorger, Nathalie Agar, Keith L. Ligon, Mehdi Touat, Sandro Santagata. Phenogenomic characterization of immunomodulatory purinergic signaling in glioblastoma [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2021; 2021 Apr 10-15 and May 17-21. Philadelphia (PA): AACR; Cancer Res 2021;81(13_Suppl):Abstract nr 1816.
- Published
- 2021
40. HGG-38. DE NOVO PYRIMIDINE SYNTHESIS INHIBITION INDUCES REPLICATION CATASTROPHE MEDIATED CELL DEATH IN DIFFUSE MIDLINE GLIOMA
- Author
-
Jakub P Kaplan, Sabine Mueller, Dipanjan Chowdhury, Daphne A. Haas-Kogan, Sylwia A. Stopka, Sharmistha Pal, Charles D. Stiles, Tabitha Cooney, Michael S. Regan, Nathalie Y. R. Agar, and Benjamin H. Kann
- Subjects
Cancer Research ,Programmed cell death ,Biology ,medicine.disease ,Oncology ,Glioma ,Pyrimidine Synthesis Inhibition ,Replication (statistics) ,medicine ,Cancer research ,AcademicSubjects/MED00300 ,AcademicSubjects/MED00310 ,Neurology (clinical) ,High Grade Gliomas - Abstract
Diffuse midline gliomas (DMG) are aggressive and lethal pediatric brain tumors that cannot be cured by conventional therapeutic modalities. Using a genome wide CRISPR screen we identified the de novo pyrimidine biosynthesis pathway as a metaboilic vulnerability in DMGs. BAY2402234 is a small molecule inhibitor of DHODH -a rate liminting enzyme in the de novo pyrimidine biosynthesis pathway. BAY2402234 induces cell death in DMG cells at low nanomolar concentrations while sparing adult glioblastoma cells and normal astrocytes. Further investigations revealed drammatic reduction in cellular UMP pools, the precursor for all pyrimidine nucleotides, after DHODH inhibition, specifically in DMG cells. Cytotoxicity of DHODH inhibition in DMG cells is rescued by exogenous uridine, supporting UMP depletion as the mechanism underlying DMG cell death and also showing that cell death is an “on target” response to BAY2402234. Cell death induced by BAY2402234 is a consequence of replication fork stalling as evident by accumulation of chromatin-bound RPA foci and g-H2AX. Stalled replication forks eventually collapse, resulting in replication catastrophy and apoptosis. Cytotoxic effects of DHODH inhibition are further exacerbated by inhibition of the intra-S checkpoint protein, ATR. Combined treatment of DMG cells with DHODH and ATR inhibitors resulted in enhanced accumulation of chromatin-bound RPA, g-H2AX, replication fork collapse and apoptosis. Importantly, in vivo studies verify that both BAY2402234 (DHODHi), and BAY1895344 (ATRi), cross the blood-brain barrier, accumulate in the brain at therapeutically relevant concentrations, and induce DNA damage in intracranial DMG xenografts in mice. Taken together, our studies have identified DHODH inhibition as a DMG-specific vulnerability resulting in cell death; the mechanism of DHODHi-induced cell death led us to identify combined inhibition of DHODH and ATR as a synergistic therapy against DMG tumors.
- Published
- 2021
41. EPCT-09. CNS LEVELS OF PANOBINOSTAT IN A NON-HUMAN PRIMATE MODEL: COMPARISON OF BLOOD AND CEREBROSPINAL FLUID PHARMACOKINETIC METHODS AND MALDI MSI
- Author
-
Josh Kramer, Cynthia McCully, Matthew Breed, W. Douglas Figg, Sylwia A. Stopka, Sara Zimmerman, Katherine E. Warren, Cody J. Peer, Thet Aye, Rafael Cruz Garcia, Nathalie Y. R. Agar, and Michael S. Regan
- Subjects
Cancer Research ,Microdialysis ,Pathology ,medicine.medical_specialty ,Non human primate ,business.industry ,Blood–brain barrier ,Maldi msi ,Translational/Early Phase Clinical Trials ,chemistry.chemical_compound ,medicine.anatomical_structure ,Cerebrospinal fluid ,Oncology ,chemistry ,Pharmacokinetics ,Panobinostat ,medicine ,AcademicSubjects/MED00300 ,Choroid plexus ,AcademicSubjects/MED00310 ,Neurology (clinical) ,business - Abstract
Adequate exposure (effective concentration over time) of a therapeutic agent at its site of action is essential for antitumor efficacy. Given constraints of repeat tissue sampling, non-human primate models predictive of pharmacokinetics in pediatric patients have been utilized to assess central nervous system (CNS) exposure. Assessment of cerebrospinal fluid (CSF) drug levels have been used to extrapolate CNS penetration but the relationship of CSF drug levels with tissue distribution is unclear. Utilizing microdialysis, we previously demonstrated geographic variability of drug permeability across the blood:brain barrier (BBB), but this technique is complex and has a high standard deviation. We, therefore, explored a novel technique, matrix-assisted laser desorption/ionization mass spectrometry imaging (MALDI MSI), to compare plasma, CSF, and tissue drug levels in a terminal non-human primate model. Panobinostat, an HDAC inhibitor in clinical trials for DIPG/DMG, was selected for study as it has previously demonstrated poor CNS tissue penetration but suggested modest clinical activity. Methods Panobinostat (p.o., dose 1.6 mg/kg) was administered to non-tumor bearing primates (n=2). One hour following administration (Tmax), blood and CSF were collected, the animal euthanized, brain and spinal cord extracted, and immediately frozen at -80. Panobinostat distribution was mapped on ex vivo sagittal tissue sections using MALDI MSI. To provide specificity and degree of permeability, anatomical structures were segmented for analysis to determine drug concentrations. Blood, CSF and tissue levels of panobinostat were measured via LC-MS/MS. Results Segmentation analysis revealed quantifiable panobinostat, particularly in the lateral ventricles and choroid plexus, and also in the subventricular zone and brainstem, although the overall panobinostat concentration was below the limit of quantitation in these areas. Conclusions Although not reflected in CSF PK, panobinostat is widely distributed in brain tissue. MALDI MSI allows regional assessment of panobinostat penetration and complements CSF pharmacokinetics.
- Published
- 2021
42. TAMI-45. PHENOGENOMIC CHARACTERIZATION OF IMMUNOMODULATORY PURINERGIC SIGNALING IN GLIOBLASTOMA
- Author
-
Sylwia A. Stopka, Mehdi Touat, Shannon Coy, Pratiti Bandopadhayay, Jia-Ren Lin, Peter K. Sorger, Jaeho Hwang, Prasidda Khadka, Rumana Rashid, Patrick Y. Wen, Philipp Euskirchen, Keith L. Ligon, Sandro Santagata, and Nathalie Y. R. Agar
- Subjects
Cancer Research ,Oncology ,medicine ,Cancer research ,Tumor Microenvironment/Angiogenesis/Metabolism/Invasion ,Neurology (clinical) ,Purinergic signalling ,Biology ,medicine.disease ,neoplasms ,Glioblastoma - Abstract
INTRODUCTION Purinergic signaling plays critical roles in the regulation of tumor growth and anti-tumor immunity via autocrine/paracrine binding of metabolites to receptors on neoplastic and non-neoplastic populations. Extracellular purine concentrations are mediated by the ectonucleotidase enzymes CD39 and CD73, which catabolize ATP to adenosine. Within tumors such as glioblastoma, neoplastic, immune, and stromal cells expressing these enzymes may co-localize to generate immunosuppressive adenosine-rich environments. However, the composition, architecture, and phenotypic properties of these tumor ecosystems and their relationship to tumor genotype are poorly characterized. METHODS We quantified CD73 expression by immunohistochemistry in a cohort of CNS tumors [meningiomas(n=222), gliomas(n=244), ependymomas(n=44), medulloblastomas(n=24), and craniopharyngiomas(n=38)]. We used publicly-available single-cell RNA-seq data and 36-marker multiplexed tissue imaging (t-CyCIF) of 139 clinically and genomically annotated glioblastoma resections to characterize CD39 and CD73-expressing populations, define the immune architecture and tumor cell-states at single cell resolution, and identify markers of clinical outcome. We used mass spectrometry imaging (MALDI-MSI) to generate spatially-resolved quantification of purine metabolite levels in glioblastoma resections (n=10). RESULTS CD73 exhibited strong expression in a subset of gliomas and meningiomas but was typically not expressed in ependymomas or medulloblastomas. CD73 expression correlated with poor progression-free-survival in IDH-wildtype glioblastoma (p=0.04). scRNA-seq and t-CyCIF in glioblastoma showed CD73 expression in tumor cells, and CD39 expression in macrophages and endothelial cells. MALDI-MSI showed significantly greater adenosine concentrations (3.5-fold;p=0.04) in glioblastomas with high CD73 expression. scRNA-seq showed direct correlations between stem-like mRNA expression, proliferation, and CD73 expression in DIPG. CD73 expression significantly correlated with EGFR amplification, interferon signaling, and PD-L1 expression in glioblastoma. CONCLUSIONS Phenogenomic analysis of purinergic immunomodulatory signaling revealed significant interplay between CD73 activity and genotype, adenosine concentration, differentiation-state, clinical outcome, and possible interaction between CD39-positive macrophages and CD73-positive neoplastic cells. Anti-CD73 therapy may provide therapeutic benefits in glioblastoma by blunting immunosuppressive and oncogenic adenosine signaling.
- Published
- 2020
43. Metabolomic Profiling of Adherent Mammalian Cells In Situ by LAESI-MS with Ion Mobility Separation
- Author
-
Sylwia A, Stopka and Akos, Vertes
- Subjects
Spectrometry, Mass, Electrospray Ionization ,Ion Mobility Spectrometry ,Metabolome ,Animals ,Humans ,Metabolomics ,Cells, Cultured - Abstract
Ambient ionization-based mass spectrometry (MS) methods coupled with ion mobility separation (IMS) have emerged as promising approaches for high-throughput in situ analysis for biomedical to environmental applications. These methods are capable of direct profiling and molecular imaging of metabolites, lipids, peptides, and xenobiotics from biological tissues with minimal sample preparation. Furthermore, employing IMS within the workflow improves the molecular coverage, resolves isobaric species, and improves biomolecule identifications through accurate collision cross section measurements. Laser ablation electrospray ionization (LAESI)-MS coupled with IMS has been successful in profiling and molecular imaging of small biomolecules directly from biological tissues and single cells. Herein, we describe a protocol for the direct analysis of adherent mammalian cells with limited perturbations by LAESI-IMS-MS. A benefit of IMS is that within the same LAESI acquisition, the spectral features related to the ESI background, washing buffer, and cell signal can be extracted and isolated separately.
- Published
- 2019
44. Metabolomic Profiling of Adherent Mammalian Cells In Situ by LAESI-MS with Ion Mobility Separation
- Author
-
Sylwia A. Stopka and Akos Vertes
- Subjects
chemistry.chemical_classification ,In situ ,0303 health sciences ,Chromatography ,Chemistry ,Biomolecule ,Laser ablation electrospray ionization ,010401 analytical chemistry ,Mass spectrometry ,01 natural sciences ,0104 chemical sciences ,Ion ,03 medical and health sciences ,Sample preparation ,Molecular imaging ,030304 developmental biology ,Ambient ionization - Abstract
Ambient ionization-based mass spectrometry (MS) methods coupled with ion mobility separation (IMS) have emerged as promising approaches for high-throughput in situ analysis for biomedical to environmental applications. These methods are capable of direct profiling and molecular imaging of metabolites, lipids, peptides, and xenobiotics from biological tissues with minimal sample preparation. Furthermore, employing IMS within the workflow improves the molecular coverage, resolves isobaric species, and improves biomolecule identifications through accurate collision cross section measurements. Laser ablation electrospray ionization (LAESI)-MS coupled with IMS has been successful in profiling and molecular imaging of small biomolecules directly from biological tissues and single cells. Herein, we describe a protocol for the direct analysis of adherent mammalian cells with limited perturbations by LAESI-IMS-MS. A benefit of IMS is that within the same LAESI acquisition, the spectral features related to the ESI background, washing buffer, and cell signal can be extracted and isolated separately.
- Published
- 2019
45. Toward Single Cell Molecular Imaging by Matrix-Free Nanophotonic Laser Desorption Ionization Mass Spectrometry
- Author
-
Sylwia A, Stopka and Akos, Vertes
- Subjects
Silicon ,Tissue Array Analysis ,Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization ,Optical Imaging ,Cell Culture Techniques ,Humans ,Hep G2 Cells ,Single-Cell Analysis ,Molecular Imaging ,Nanostructures - Abstract
In recent years, innovations in mass spectrometry imaging (MSI) have enabled simultaneous detection and mapping of biomolecules and xenobiotics directly from biological tissues and single cells. Matrix-assisted laser desorption ionization (MALDI) has been the most widely embraced MSI technique. However, this technique can exhibit ion suppression effects hindering metabolite coverage and possesses a narrow dynamic range. Nanophotonic platforms, e.g., silicon nanopost array (NAPA) structures, can be used as an alternative for matrix-free imaging of biological tissues. Here, we present a protocol for MSI of large and small adherent cell clusters by laser desorption ionization from NAPA with minimal sample preparation.
- Published
- 2019
46. Toward Single Cell Molecular Imaging by Matrix-Free Nanophotonic Laser Desorption Ionization Mass Spectrometry
- Author
-
Sylwia A. Stopka and Akos Vertes
- Subjects
chemistry.chemical_classification ,Materials science ,Silicon ,Biomolecule ,chemistry.chemical_element ,Nanotechnology ,Ion suppression in liquid chromatography–mass spectrometry ,02 engineering and technology ,010402 general chemistry ,021001 nanoscience & nanotechnology ,Laser ,01 natural sciences ,Mass spectrometry imaging ,0104 chemical sciences ,law.invention ,Matrix (chemical analysis) ,chemistry ,law ,Sample preparation ,Molecular imaging ,0210 nano-technology - Abstract
In recent years, innovations in mass spectrometry imaging (MSI) have enabled simultaneous detection and mapping of biomolecules and xenobiotics directly from biological tissues and single cells. Matrix-assisted laser desorption ionization (MALDI) has been the most widely embraced MSI technique. However, this technique can exhibit ion suppression effects hindering metabolite coverage and possesses a narrow dynamic range. Nanophotonic platforms, e.g., silicon nanopost array (NAPA) structures, can be used as an alternative for matrix-free imaging of biological tissues. Here, we present a protocol for MSI of large and small adherent cell clusters by laser desorption ionization from NAPA with minimal sample preparation.
- Published
- 2019
47. In-Situ Metabolomic Analysis of
- Author
-
Beverly J, Agtuca, Sylwia A, Stopka, Thalita R, Tuleski, Fernanda P, do Amaral, Sterling, Evans, Yang, Liu, Dong, Xu, Rose Adele, Monteiro, David W, Koppenaal, Ljiljana, Paša-Tolić, Christopher R, Anderton, Akos, Vertes, and Gary, Stacey
- Subjects
Herbaspirillum ,Nitrogen Fixation ,Host-Pathogen Interactions ,Setaria Plant ,Metabolome ,Symbiosis ,Plant Roots - Abstract
Over the past decades, crop yields have risen in parallel with increasing use of fossil fuel-derived nitrogen (N) fertilizers but with concomitant negative impacts on climate and water resources. There is a need for more sustainable agricultural practices, and biological nitrogen fixation (BNF) could be part of the solution. A variety of nitrogen-fixing, epiphytic, and endophytic plant growth-promoting bacteria (PGPB) are known to stimulate plant growth. However, compared with the rhizobium-legume symbiosis, little mechanistic information is available as to how PGPB affect plant metabolism. Therefore, we investigated the metabolic changes in roots of the model grass species
- Published
- 2019
48. Optical Microscopy-Guided Laser Ablation Electrospray Ionization Ion Mobility Mass Spectrometry: Ambient Single Cell Metabolomics with Increased Confidence in Molecular Identification
- Author
-
Sylwia A. Stopka, Sara Mattson, Yehia M. Ibrahim, Andrey V. Liyu, Michael J. Taylor, Christopher R. Anderton, and Akos Vertes
- Subjects
0301 basic medicine ,Microscope ,Materials science ,Ion-mobility spectrometry ,Endocrinology, Diabetes and Metabolism ,Electrospray ionization ,Laser ablation electrospray ionization ,drift tube ion mobility separation ,lcsh:QR1-502 ,Analytical chemistry ,laser ablation electrospray ionization ,collisional cross section ,Mass spectrometry ,mass spectrometry ,in situ metabolomics ,ambient analysis ,01 natural sciences ,Biochemistry ,Article ,lcsh:Microbiology ,law.invention ,03 medical and health sciences ,Metabolomics ,Single-cell analysis ,law ,Ionization ,Molecular Biology ,010401 analytical chemistry ,0104 chemical sciences ,030104 developmental biology - Abstract
Single cell analysis is a field of increasing interest as new tools are continually being developed to understand intercellular differences within large cell populations. Laser-ablation electrospray ionization mass spectrometry (LAESI-MS) is an emerging technique for single cell metabolomics. Over the years, it has been validated that this ionization technique is advantageous for probing the molecular content of individual cells in situ. Here, we report the integration of a microscope into the optical train of the LAESI source to allow for visually informed ambient in situ single cell analysis. Additionally, we have coupled this ‘LAESI microscope’ to a drift-tube ion mobility mass spectrometer to enable separation of isobaric species and allow for the determination of ion collision cross sections in conjunction with accurate mass measurements. This combined information helps provide higher confidence for structural assignment of molecules ablated from single cells. Here, we show that this system enables the analysis of the metabolite content of Allium cepa epidermal cells with high confidence structural identification together with their spatial locations within a tissue.
- Published
- 2021
49. DDRE-32. THERAPEUTIC TARGETING OF A NOVEL METABOLIC ADDICTION IN DIFFUSE MIDLINE GLIOMA
- Author
-
Jakub P Kaplan, Samuel K. McBrayer, Nathalie Y. R. Agar, Dipanjan Chowdhury, Tabitha Cooney, Sabine Mueller, Benjamin H. Kann, Sharmistha Pal, Bradley R Hunsel, William G. Kaelin, Sylwia A. Stopka, Charles D. Stiles, Daphne A. Haas-Kogan, and Michael S. Regan
- Subjects
Emotional vulnerability ,business.industry ,Addiction ,media_common.quotation_subject ,Childhood cancer ,Cancer ,Metabolic Drug Targets, Resistance ,medicine.disease ,Therapeutic targeting ,Blood–brain barrier ,Supplement Abstracts ,medicine.anatomical_structure ,Glioma ,medicine ,Cancer research ,AcademicSubjects/MED00300 ,AcademicSubjects/MED00310 ,Pyrimidine Nucleotides ,business ,media_common - Abstract
Diffuse midline glioma (DMG) is a uniformly fatal pediatric cancer that is in need of urgent “outside the box” therapeutic approaches. Recent studies show that tumor cells adapt to stresses created by oncogenic mutations and these oncogene-induced adaptations create vulnerabilities that can be exploited to therapeutic ends. To uncover these oncogene-induced vulnerabilities in DMGs we conducted a genome-wide CRIPSR knockout screen in three DMG lines. The top common DMG dependency pathway that we discovered is de novo pyrimidine biosynthesis. Under normal conditions pyrimidine nucleotide needs are met through the salvage pathway. However, in DMG tumorigenesis, pyrimidine nucleotide synthesis is rewired such that the cells become dependent on the de novo biosynthesis pathway. De novo pyrimidine synthesis is catalyzed by CAD, DHODH and UMPS; all three genes are identified as dependencies in our screen and have been validated using shRNA mediated gene knockdown. Interestingly, DMG cells did not exhibit a dependency on the de novo purine biosynthesis pathway. Using a small molecule inhibitor of DHODH, BAY2402234 [currently studied in phase I trial for myeloid malignancies (NCT03404726)], we have demonstrated and validated, (i) efficacy and specificity of de novo pyrimidine synthesis inhibition in vitro in DMG cells; (ii) de novo pyrimidine addiction is not attributable to cell proliferation; (iii) DHODH inhibition induces apoptosis by hindering replication and inciting DNA damage; (iv) DHODH and ATR inhibition act synergistically to induce DMG cell death; and (v) critical in vivo efficacy. The in vivo experiment documents that BAY2402234 crosses the blood-brain barrier, is present in the brain at therapeutically relevant concentrations, suppresses de novo pyrimidine biosynthesis in intracranial DMG tumors in mice, and prolongs survival of orthotopic DMG tumor bearing mice. Taken together, our studies have identified a novel metabolic vulnerability that can be translated for the treatment of DMG patients.
- Published
- 2021
50. Molecular Imaging of Biological Samples on Nanophotonic Laser Desorption Ionization Platforms
- Author
-
Sylwia A. Stopka, Andrew R. Korte, Nicholas J. Morris, Akos Vertes, Charles Rong, Javad Nazarian, Trust T. Razunguzwa, and Sridevi Yadavilli
- Subjects
MALDI imaging ,Matrix-assisted laser desorption electrospray ionization ,Analytical chemistry ,010402 general chemistry ,01 natural sciences ,Catalysis ,Mass spectrometry imaging ,law.invention ,Matrix (chemical analysis) ,Mice ,law ,Ionization ,Animals ,chemistry.chemical_classification ,Photons ,Chemistry ,Biomolecule ,Lasers ,010401 analytical chemistry ,General Chemistry ,General Medicine ,Laser ,Molecular Imaging ,0104 chemical sciences ,Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization ,Microscopy, Electron, Scanning ,Molecular imaging - Abstract
Mass spectrometry imaging (MSI) is a comprehensive tool for the analysis of a wide range of biomolecules. The mainstream method for molecular MSI is matrix-assisted laser desorption ionization, however, the presence of a matrix results in spectral interferences and the suppression of some analyte ions. Herein we demonstrate a new matrix-free MSI technique using nanophotonic ionization based on laser desorption ionization (LDI) from a highly uniform silicon nanopost array (NAPA). In mouse brain and kidney tissue sections, the distributions of over 80 putatively annotated molecular species are determined with 40 μm spatial resolution. Furthermore, NAPA-LDI-MS is used to selectively analyze metabolites and lipids from sparsely distributed algal cells and the lamellipodia of human hepatocytes. Our results open the door for matrix-free MSI of tissue sections and small cell populations by nanophotonic ionization.
- Published
- 2016
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.