Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
1.
Front Mol Biosci ; 9: 785232, 2022.
Article in English | MEDLINE | ID: mdl-35463966

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.

2.
NPJ Breast Cancer ; 7(1): 116, 2021 Sep 09.
Article in English | MEDLINE | ID: mdl-34504095

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.

3.
Nat Commun ; 9(1): 4904, 2018 11 21.
Article in English | MEDLINE | ID: mdl-30464169

ABSTRACT

Therapeutic options for the treatment of glioblastoma remain inadequate despite concerted research efforts in drug development. Therapeutic failure can result from poor permeability of the blood-brain barrier, heterogeneous drug distribution, and development of resistance. Elucidation of relationships among such parameters could enable the development of predictive models of drug response in patients and inform drug development. Complementary analyses were applied to a glioblastoma patient-derived xenograft model in order to quantitatively map distribution and resulting cellular response to the EGFR inhibitor erlotinib. Mass spectrometry images of erlotinib were registered to histology and magnetic resonance images in order to correlate drug distribution with tumor characteristics. Phosphoproteomics and immunohistochemistry were used to assess protein signaling in response to drug, and integrated with transcriptional response using mRNA sequencing. This comprehensive dataset provides simultaneous insight into pharmacokinetics and pharmacodynamics and indicates that erlotinib delivery to intracranial tumors is insufficient to inhibit EGFR tyrosine kinase signaling.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Erlotinib Hydrochloride/pharmacokinetics , Glioblastoma/drug therapy , Animals , Antineoplastic Agents/administration & dosage , ErbB Receptors/antagonists & inhibitors , Erlotinib Hydrochloride/administration & dosage , Female , Magnetic Resonance Imaging , Mice, Nude , Neoplasm Transplantation , Protein-Tyrosine Kinases/metabolism , Sequence Analysis, RNA , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
4.
J Neurooncol ; 140(2): 269-279, 2018 Nov.
Article in English | MEDLINE | ID: mdl-30128689

ABSTRACT

PURPOSE: Medulloblastoma, the most common primary pediatric malignant brain tumor, originates in the posterior fossa of the brain. Pineoblastoma, which originates within the pineal gland, is a rarer malignancy that also presents in the pediatric population. Medulloblastoma and pineoblastoma exhibit overlapping clinical features and have similar histopathological characteristics. Histopathological similarities confound rapid diagnoses of these two tumor types. We have conducted a pilot feasibility study analyzing the molecular profile of archived frozen human tumor specimens using mass spectrometry imaging (MSI) to identify potential biomarkers capable of classifying and distinguishing between medulloblastoma and pineoblastoma. METHODS: We performed matrix-assisted laser desorption ionization Fourier transform ion cyclotron resonance mass spectrometry imaging on eight medulloblastoma biopsy specimens and three pineoblastoma biopsy specimens. Multivariate statistical analyses were performed on the MSI dataset to generate classifiers that distinguish the two tumor types. Lastly, the molecules that were discriminative of tumor type were queried against the Lipid Maps database and identified. RESULTS: In this pilot study we show that medulloblastoma and pineoblastoma can be discriminated using molecular profiles determined by MSI. The highest-ranking discriminating classifiers of medulloblastoma and pineoblastoma were glycerophosphoglycerols and sphingolipids, respectively. CONCLUSION: We demonstrate proof-of-concept that medulloblastoma and pineoblastoma can be rapidly distinguished by using MSI lipid profiles. We identified biomarker candidates capable of distinguishing these two histopathologically similar tumor types. This work expands the current molecular knowledge of medulloblastoma and pineoblastoma by characterizing their lipidomic profiles, which may be useful for developing novel diagnostic, prognostic and therapeutic strategies.


Subject(s)
Brain Neoplasms/metabolism , Medulloblastoma/metabolism , Pinealoma/metabolism , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Brain Neoplasms/diagnostic imaging , Brain Neoplasms/pathology , Cerebellum/diagnostic imaging , Cerebellum/metabolism , Child , Diagnosis, Differential , Humans , Medulloblastoma/diagnostic imaging , Medulloblastoma/pathology , Pilot Projects , Pineal Gland/diagnostic imaging , Pineal Gland/metabolism , Pineal Gland/pathology , Pinealoma/diagnostic imaging , Pinealoma/pathology , Proof of Concept Study
5.
Mol Cancer Ther ; 17(9): 1893-1901, 2018 09.
Article in English | MEDLINE | ID: mdl-29970480

ABSTRACT

Controversy exists surrounding whether heterogeneous disruption of the blood-brain barrier (BBB), as seen in glioblastoma (GBM), leads to adequate drug delivery sufficient for efficacy in GBM. This question is especially important when using potent, targeted agents that have a poor penetration across an intact BBB. Efficacy of the murine double minute-2 (MDM2) inhibitor SAR405838 was tested in patient-derived xenograft (PDX) models of GBM. In vitro efficacy of SAR405838 was evaluated in PDX models with varying MDM2 expression and those with high (GBM108) and low (GBM102) expression were evaluated for flank and orthotopic efficacy. BBB permeability, evaluated using TexasRed-3 kDa dextran, was significantly increased in GBM108 through VEGFA overexpression. Drug delivery, MRI, and orthotopic survival were compared between BBB-intact (GBM108-vector) and BBB-disrupted (GBM108-VEGFA) models. MDM2-amplified PDX lines with high MDM2 expression were sensitive to SAR405838 in comparison with MDM2 control lines in both in vitro and heterotopic models. In contrast with profound efficacy observed in flank xenografts, SAR405838 was ineffective in orthotopic tumors. Although both GBM108-vector and GBM108-VEGFA readily imaged on MRI following gadolinium contrast administration, GBM108-VEGFA tumors had a significantly enhanced drug and gadolinium accumulation, as determined by MALDI-MSI. Enhanced drug delivery in GBM108-VEGFA translated into a marked improvement in orthotopic efficacy. This study clearly shows that limited drug distribution across a partially intact BBB may limit the efficacy of targeted agents in GBM. Brain penetration of targeted agents is a critical consideration in any precision medicine strategy for GBM. Mol Cancer Ther; 17(9); 1893-901. ©2018 AACR.


Subject(s)
Blood-Brain Barrier/drug effects , Brain Neoplasms/drug therapy , Glioblastoma/drug therapy , Indoles/pharmacology , Proto-Oncogene Proteins c-mdm2/antagonists & inhibitors , Spiro Compounds/pharmacology , Xenograft Model Antitumor Assays , Animals , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Line, Tumor , Female , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , Indoles/pharmacokinetics , Male , Mice , Proto-Oncogene Proteins c-mdm2/metabolism , Spiro Compounds/pharmacokinetics , Survival Analysis , Treatment Outcome , Tumor Suppressor Protein p53/metabolism
6.
Pain Pract ; 18(7): 889-894, 2018 09.
Article in English | MEDLINE | ID: mdl-29480977

ABSTRACT

Opioids are often used for analgesia via continuous intrathecal delivery by implantable devices. A higher concentration and daily dose of opioid have been postulated as risk factors for intrathecal granuloma formation. We present a 42-year-old female patient with chronic abdominal pain from refractory pancreatitis, with an intrathecal drug delivery device implanted 21 years prior, delivering continuous intrathecal morphine. After many years without concerning physical signs or complaints, with gradual increases in daily morphine dose, she presented with rapidly progressive neurologic deficits, including lower extremity, bladder, and bowel symptoms. These symptoms were determined to be secondary to mass effect and local inflammation related to an intrathecal catheter tip granuloma, detected on magnetic resonance imaging of the spine. The mass was urgently resected. On histopathologic examination, this granuloma was found to be unique, in that in addition to the expected inflammatory components, it appeared to contain precipitated nonpolarizable crystals. These were identified as precipitated morphine using liquid extraction surface analysis-tandem mass spectrometry (LESA-MS/MS) and matrix-assisted laser desorption ionization-Fourier transform ion cyclotron resonance-mass spectrometry imaging (MALDI-FTICR-MSI). In addition to the unique finding of precipitated morphine crystals, the long-term follow-up of both morphine concentration and daily dose increases provides insight into the formation of intrathecal granulomas.


Subject(s)
Analgesics, Opioid/adverse effects , Granuloma/chemically induced , Morphine/adverse effects , Spinal Cord Diseases/chemically induced , Adult , Analgesics, Opioid/administration & dosage , Female , Granuloma/diagnosis , Humans , Infusion Pumps, Implantable , Infusions, Spinal/adverse effects , Longitudinal Studies , Morphine/administration & dosage , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Spinal Cord Diseases/diagnosis , Tandem Mass Spectrometry
7.
Cancer Cell ; 32(4): 411-426.e11, 2017 10 09.
Article in English | MEDLINE | ID: mdl-28966034

ABSTRACT

Glioblastoma (GBM) is a devastating malignancy with few therapeutic options. We identify PRMT5 in an in vivo GBM shRNA screen and show that PRMT5 knockdown or inhibition potently suppresses in vivo GBM tumors, including patient-derived xenografts. Pathway analysis implicates splicing in cellular PRMT5 dependency, and we identify a biomarker that predicts sensitivity to PRMT5 inhibition. We find that PRMT5 deficiency primarily disrupts the removal of detained introns (DIs). This impaired DI splicing affects proliferation genes, whose downregulation coincides with cell cycle defects, senescence and/or apoptosis. We further show that DI programs are evolutionarily conserved and operate during neurogenesis, suggesting that they represent a physiological regulatory mechanism. Collectively, these findings reveal a PRMT5-regulated DI-splicing program as an exploitable cancer vulnerability.


Subject(s)
Brain Neoplasms/pathology , Glioma/pathology , Introns , Protein-Arginine N-Methyltransferases/physiology , Animals , Cell Cycle/drug effects , Cell Differentiation , Cell Line, Tumor , Cell Proliferation/drug effects , Glioma/drug therapy , Glioma/genetics , High-Throughput Screening Assays , Humans , Isoquinolines/pharmacology , Mice , Protein-Arginine N-Methyltransferases/antagonists & inhibitors , Pyrimidines/pharmacology , RNA Splicing
8.
Nat Commun ; 8: 15623, 2017 06 06.
Article in English | MEDLINE | ID: mdl-28585535

ABSTRACT

Culture-based blood-brain barrier (BBB) models are crucial tools to enable rapid screening of brain-penetrating drugs. However, reproducibility of in vitro barrier properties and permeability remain as major challenges. Here, we report that self-assembling multicellular BBB spheroids display reproducible BBB features and functions. The spheroid core is comprised mainly of astrocytes, while brain endothelial cells and pericytes encase the surface, acting as a barrier that regulates transport of molecules. The spheroid surface exhibits high expression of tight junction proteins, VEGF-dependent permeability, efflux pump activity and receptor-mediated transcytosis of angiopep-2. In contrast, the transwell co-culture system displays comparatively low levels of BBB regulatory proteins, and is unable to discriminate between the transport of angiopep-2 and a control peptide. Finally, we have utilized the BBB spheroids to screen and identify BBB-penetrant cell-penetrating peptides (CPPs). This robust in vitro BBB model could serve as a valuable next-generation platform for expediting the development of CNS therapeutics.


Subject(s)
Biological Transport/physiology , Blood-Brain Barrier/metabolism , Spheroids, Cellular/metabolism , Tight Junctions/metabolism , Astrocytes/metabolism , Cells, Cultured , Claudin-5/metabolism , Coculture Techniques , Endothelial Cells/metabolism , Humans , Models, Biological , Peptides/metabolism , Pericytes/metabolism , Permeability , Zonula Occludens-1 Protein/metabolism
9.
Neuro Oncol ; 19(6): 774-785, 2017 06 01.
Article in English | MEDLINE | ID: mdl-28082416

ABSTRACT

Background: Activating mutations or structural rearrangements in BRAF are identified in roughly 75% of all pediatric low-grade astrocytomas (PLGAs). However, first-generation RAF inhibitors approved for adult melanoma have poor blood-brain penetrance and are only effective on tumors that express the canonical BRAFV600E oncoprotein, which functions as a monomer. These drugs (type I antagonists that target the "DFG-in" conformation of the kinase) fail to block signaling via KIAA1549:BRAF, a truncation/fusion BRAF oncoprotein which functions as a dimer and is found in the most common form of PLGA. Methods: A panel of small molecule RAF inhibitors (including type II inhibitors, targeting the "DFG-out" conformation of the kinase) was screened for drugs showing efficacy on murine models of PLGA and on authentic human PLGA cells expressing KIAA1549:BRAF. Results: We identify a type II RAF inhibitor that serves as an equipotent antagonist of BRAFV600E, KIAA1549:BRAF, and other noncanonical BRAF oncoproteins that function as dimers. This drug (MLN2480, also known as TAK-580) has good brain penetrance and is active on authentic human PLGA cells in brain organotypic cultures. Conclusion: MLN2480 may be an effective therapeutic for BRAF mutant pediatric astrocytomas.


Subject(s)
Astrocytoma/drug therapy , Brain Neoplasms/drug therapy , Heterocyclic Compounds, 3-Ring/pharmacology , Oncogene Proteins, Fusion/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Protein Multimerization/drug effects , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , raf Kinases/antagonists & inhibitors , Animals , Astrocytoma/metabolism , Astrocytoma/pathology , Blood-Brain Barrier/drug effects , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Child , Heterocyclic Compounds, 3-Ring/chemistry , High-Throughput Screening Assays , Humans , Male , Mice , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/metabolism , Protein Kinase Inhibitors/chemistry , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins B-raf/metabolism , Tumor Cells, Cultured , Xenograft Model Antitumor Assays , raf Kinases/genetics , raf Kinases/metabolism
10.
Oncotarget ; 8(14): 22370-22384, 2017 Apr 04.
Article in English | MEDLINE | ID: mdl-26517684

ABSTRACT

Transcription factors (TFs) are a major class of protein signaling molecules that play key cellular roles in cancers such as the highly lethal brain cancer-glioblastoma (GBM). However, the development of specific TF inhibitors has proved difficult owing to expansive protein-protein interfaces and the absence of hydrophobic pockets. We uniquely defined the dimerization surface as an expansive parental pharmacophore comprised of several regional daughter pharmacophores. We targeted the OLIG2 TF which is essential for GBM survival and growth, we hypothesized that small molecules able to fit each subpharmacophore would inhibit OLIG2 activation. The most active compound was OLIG2 selective, it entered the brain, and it exhibited potent anti-GBM activity in cell-based assays and in pre-clinical mouse orthotopic models. These data suggest that (1) our multiple pharmacophore approach warrants further investigation, and (2) our most potent compounds merit detailed pharmacodynamic, biophysical, and mechanistic characterization for potential preclinical development as GBM therapeutics.


Subject(s)
Antineoplastic Agents/therapeutic use , Basic Helix-Loop-Helix Transcription Factors/antagonists & inhibitors , Brain Neoplasms/drug therapy , Drug Design , Glioblastoma/drug therapy , Guanidines/therapeutic use , Molecular Targeted Therapy , Nerve Tissue Proteins/antagonists & inhibitors , Animals , Antineoplastic Agents/pharmacology , Basic Helix-Loop-Helix Transcription Factors/chemistry , Cell Growth Processes , Cell Survival/genetics , Computer Simulation , Humans , Mice , Mice, Nude , Molecular Structure , Nerve Tissue Proteins/chemistry , Oligodendrocyte Transcription Factor 2 , Protein Binding , Protein Conformation , Small Molecule Libraries , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
11.
J Biomed Nanotechnol ; 12(6): 1297-302, 2016 Jun.
Article in English | MEDLINE | ID: mdl-27319222

ABSTRACT

Medulloblastoma is the most common childhood malignant brain tumor. The most lethal medulloblastoma subtype exhibits a high expression of the GABAA receptor α5 subunit gene and MYC amplification. New benzodiazepines have been synthesized to function as α5-GABAA receptor ligands. To compare their efficacy with that of standard-of-care treatments, we have employed a newly developed microscale implantable device that allows for high-throughput localized intratumor drug delivery and efficacy testing. Microdoses of each drug were delivered into small distinct regions of tumors, as confirmed by tissue mass spectrometry, and the local drug effect was determined by immunohistochemistry. We have identified a benzodiazepine derivative, KRM-II-08, as a new potent inhibitor in several α5-GABAA receptor expressing tumor models. This is the first instance of in vivo testing of several benzodiazepine derivatives and standard chemotherapeutic drugs within the same tumor. Obtaining high-throughput drug efficacy data within a native tumor microenvironment as detailed herein, prior to pharmacological optimization for bioavailability or safety and without systemic exposure or toxicity, may allow for rapid prioritization of drug candidates for further pharmacological optimization.


Subject(s)
Antineoplastic Agents/administration & dosage , Cerebellar Neoplasms/drug therapy , Drug Delivery Systems , Medulloblastoma/drug therapy , Animals , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/therapeutic use , Benzodiazepines/administration & dosage , Benzodiazepines/pharmacokinetics , Benzodiazepines/therapeutic use , Cell Line, Tumor , Drug Delivery Systems/instrumentation , Drug Delivery Systems/methods , Female , Humans , Mice , Mice, Nude , Molecular Imaging , Prostheses and Implants , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
12.
Cancer Res ; 76(12): 3451-62, 2016 06 15.
Article in English | MEDLINE | ID: mdl-27197198

ABSTRACT

The goal of brain tumor surgery is to maximize tumor removal without injuring critical brain structures. Achieving this goal is challenging as it can be difficult to distinguish tumor from nontumor tissue. While standard histopathology provides information that could assist tumor delineation, it cannot be performed iteratively during surgery as freezing, sectioning, and staining of the tissue require too much time. Stimulated Raman scattering (SRS) microscopy is a powerful label-free chemical imaging technology that enables rapid mapping of lipids and proteins within a fresh specimen. This information can be rendered into pathology-like images. Although this approach has been used to assess the density of glioma cells in murine orthotopic xenografts models and human brain tumors, tissue heterogeneity in clinical brain tumors has not yet been fully evaluated with SRS imaging. Here we profile 41 specimens resected from 12 patients with a range of brain tumors. By evaluating large-scale stimulated Raman imaging data and correlating this data with current clinical gold standard of histopathology for 4,422 fields of view, we capture many essential diagnostic hallmarks for glioma classification. Notably, in fresh tumor samples, we observe additional features, not seen by conventional methods, including extensive lipid droplets within glioma cells, collagen deposition in gliosarcoma, and irregularity and disruption of myelinated fibers in areas infiltrated by oligodendroglioma cells. The data are freely available in a public resource to foster diagnostic training and to permit additional interrogation. Our work establishes the methodology and provides a significant collection of reference images for label-free neurosurgical pathology. Cancer Res; 76(12); 3451-62. ©2016 AACR.


Subject(s)
Brain Neoplasms/surgery , Glioma/surgery , Spectrum Analysis, Raman/methods , Brain Neoplasms/diagnostic imaging , Brain Neoplasms/pathology , Collagen/analysis , Glioma/diagnostic imaging , Glioma/pathology , Humans
13.
Mol Cancer Ther ; 14(12): 2735-43, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26438157

ABSTRACT

PARP inhibition can enhance the efficacy of temozolomide and prolong survival in orthotopic glioblastoma (GBM) xenografts. The aim of this study was to evaluate the combination of the PARP inhibitor rucaparib with temozolomide and to correlate pharmacokinetic and pharmacodynamic studies with efficacy in patient-derived GBM xenograft models. The combination of rucaparib with temozolomide was highly effective in vitro in short-term explant cultures derived from GBM12, and, similarly, the combination of rucaparib and temozolomide (dosed for 5 days every 28 days for 3 cycles) significantly prolonged the time to tumor regrowth by 40% in heterotopic xenografts. In contrast, the addition of rucaparib had no impact on the efficacy of temozolomide in GBM12 or GBM39 orthotopic models. Using Madin-Darby canine kidney (MDCK) II cells stably expressing murine BCRP1 or human MDR1, cell accumulation studies demonstrated that rucaparib is transported by both transporters. Consistent with the influence of these efflux pumps on central nervous system drug distribution, Mdr1a/b(-/-)Bcrp1(-/-) knockout mice had a significantly higher brain to plasma ratio for rucaparib (1.61 ± 0.25) than wild-type mice (0.11 ± 0.08). A pharmacokinetic and pharmacodynamic evaluation after a single dose confirmed limited accumulation of rucaparib in the brain is associated with substantial residual PARP enzymatic activity. Similarly, matrix-assisted laser desorption/ionization mass spectrometric imaging demonstrated significantly enhanced accumulation of drug in flank tumor compared with normal brain or orthotopic tumors. Collectively, these results suggest that limited drug delivery into brain tumors may significantly limit the efficacy of rucaparib combined with temozolomide in GBM.


Subject(s)
Central Nervous System/drug effects , Glioblastoma/drug therapy , Indoles/administration & dosage , Poly(ADP-ribose) Polymerase Inhibitors/administration & dosage , ATP Binding Cassette Transporter, Subfamily B/genetics , ATP Binding Cassette Transporter, Subfamily G, Member 2 , ATP-Binding Cassette Transporters/genetics , Animals , Cell Line, Tumor , Dacarbazine/administration & dosage , Dacarbazine/analogs & derivatives , Dacarbazine/pharmacokinetics , Dogs , Glioblastoma/genetics , Glioblastoma/pathology , Humans , Indoles/pharmacokinetics , Madin Darby Canine Kidney Cells , Mice , Mice, Knockout , Poly(ADP-ribose) Polymerase Inhibitors/pharmacokinetics , Temozolomide , Xenograft Model Antitumor Assays
14.
Proc Natl Acad Sci U S A ; 112(32): 9978-83, 2015 Aug 11.
Article in English | MEDLINE | ID: mdl-26216958

ABSTRACT

We present a proof of concept study designed to support the clinical development of mass spectrometry imaging (MSI) for the detection of pituitary tumors during surgery. We analyzed by matrix-assisted laser desorption/ionization (MALDI) MSI six nonpathological (NP) human pituitary glands and 45 hormone secreting and nonsecreting (NS) human pituitary adenomas. We show that the distribution of pituitary hormones such as prolactin (PRL), growth hormone (GH), adrenocorticotropic hormone (ACTH), and thyroid stimulating hormone (TSH) in both normal and tumor tissues can be assessed by using this approach. The presence of most of the pituitary hormones was confirmed by using MS/MS and pseudo-MS/MS methods, and subtyping of pituitary adenomas was performed by using principal component analysis (PCA) and support vector machine (SVM). Our proof of concept study demonstrates that MALDI MSI could be used to directly detect excessive hormonal production from functional pituitary adenomas and generally classify pituitary adenomas by using statistical and machine learning analyses. The tissue characterization can be completed in fewer than 30 min and could therefore be applied for the near-real-time detection and delineation of pituitary tumors for intraoperative surgical decision-making.


Subject(s)
Computer Systems , Imaging, Three-Dimensional , Pituitary Neoplasms/diagnosis , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods , Humans , Neoplasm Proteins/metabolism , Pituitary Gland/pathology , Pituitary Neoplasms/pathology , Principal Component Analysis , Reproducibility of Results
15.
Anal Bioanal Chem ; 407(20): 5989-98, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26084546

ABSTRACT

Described here are the results from the profiling of the proteins arginine vasopressin (AVP) and adrenocorticotropic hormone (ACTH) from normal human pituitary gland and pituitary adenoma tissue sections, using a fully automated droplet-based liquid-microjunction surface-sampling-HPLC-ESI-MS-MS system for spatially resolved sampling, HPLC separation, and mass spectrometric detection. Excellent correlation was found between the protein distribution data obtained with this method and data obtained with matrix-assisted laser desorption/ionization (MALDI) chemical imaging analyses of serial sections of the same tissue. The protein distributions correlated with the visible anatomic pattern of the pituitary gland. AVP was most abundant in the posterior pituitary gland region (neurohypophysis), and ATCH was dominant in the anterior pituitary gland region (adenohypophysis). The relative amounts of AVP and ACTH sampled from a series of ACTH-secreting and non-secreting pituitary adenomas correlated with histopathological evaluation. ACTH was readily detected at significantly higher levels in regions of ACTH-secreting adenomas and in normal anterior adenohypophysis compared with non-secreting adenoma and neurohypophysis. AVP was mostly detected in normal neurohypophysis, as expected. This work reveals that a fully automated droplet-based liquid-microjunction surface-sampling system coupled to HPLC-ESI-MS-MS can be readily used for spatially resolved sampling, separation, detection, and semi-quantitation of physiologically-relevant peptide and protein hormones, including AVP and ACTH, directly from human tissue. In addition, the relative simplicity, rapidity, and specificity of this method support the potential of this basic technology, with further advancement, for assisting surgical decision-making. Graphical Abstract Mass spectrometry based profiling of hormones in human pituitary gland and tumor thin tissue sections.


Subject(s)
Adenoma/pathology , Adrenocorticotropic Hormone/analysis , Arginine Vasopressin/analysis , Pituitary Gland/chemistry , Pituitary Gland/pathology , Pituitary Neoplasms/pathology , Adenoma/chemistry , Amino Acid Sequence , Chromatography, High Pressure Liquid/instrumentation , Equipment Design , Female , Humans , Microinjections/instrumentation , Middle Aged , Molecular Sequence Data , Pituitary Neoplasms/chemistry , Specimen Handling/instrumentation , Spectrometry, Mass, Electrospray Ionization/instrumentation , Tandem Mass Spectrometry/instrumentation
16.
Int J Mass Spectrom ; 377: 690-698, 2015 Feb 01.
Article in English | MEDLINE | ID: mdl-25844057

ABSTRACT

Meningiomas are the most frequent intracranial tumors. The majority is benign slow-growing tumors but they can be difficult to treat depending on their location and size. While meningiomas are well delineated on magnetic resonance imaging by their uptake of contrast, surgical limitations still present themselves from not knowing the extent of invasion of the dura matter by meningioma cells. The development of tools to characterize tumor tissue in real or near real time could prevent recurrence after tumor resection by allowing for more precise surgery, i.e. removal of tumor with preservation of healthy tissue. The development of ambient ionization mass spectrometry for molecular characterization of tissue and its implementation in the surgical decision-making workflow carry the potential to fulfill this need. Here, we present the characterization of meningioma and dura mater by desorption electrospray ionization mass spectrometry to validate the technique for the molecular assessment of surgical margins and diagnosis of meningioma from surgical tissue in real-time. Nine stereotactically resected surgical samples and three autopsy samples were analyzed by standard histopathology and mass spectrometry imaging. All samples indicated a strong correlation between results from both techniques. We then highlight the value of desorption electrospray ionization mass spectrometry for the molecular subtyping/subgrouping of meningiomas from a series of forty genetically characterized specimens. The minimal sample preparation required for desorption electrospray ionization mass spectrometry offers a distinct advantage for applications relying on real-time information such as surgical decision-making. The technology here was tested to distinguish meningioma from dura mater as an approach to precisely define surgical margins. In addition we classify meningiomas into fibroblastic and meningothelial subtypes and more notably recognize meningiomas with NF2 genetic aberrations.

17.
Clin Cancer Res ; 21(8): 1916-24, 2015 Apr 15.
Article in English | MEDLINE | ID: mdl-25609063

ABSTRACT

PURPOSE: Wee1 regulates key DNA damage checkpoints, and in this study, the efficacy of the Wee1 inhibitor MK-1775 was evaluated in glioblastoma multiforme (GBM) xenograft models alone and in combination with radiation and/or temozolomide. EXPERIMENTAL DESIGN: In vitro MK-1775 efficacy alone and in combination with temozolomide, and the impact on DNA damage, was analyzed by Western blotting and γH2AX foci formation. In vivo efficacy was evaluated in orthotopic and heterotopic xenografts. Drug distribution was assessed by conventional mass spectrometry (MS) and matrix-assisted laser desorption/ionization (MALDI)-MS imaging. RESULTS: GBM22 (IC50 = 68 nmol/L) was significantly more sensitive to MK-1775 compared with five other GBM xenograft lines, including GBM6 (IC50 >300 nmol/L), and this was associated with a significant difference in pan-nuclear γH2AX staining between treated GBM22 (81% cells positive) and GBM6 (20% cells positive) cells. However, there was no sensitizing effect of MK-1775 when combined with temozolomide in vitro. In an orthotopic GBM22 model, MK-1775 was ineffective when combined with temozolomide, whereas in a flank model of GBM22, MK-1775 exhibited both single-agent and combinatorial activity with temozolomide. Consistent with limited drug delivery into orthotopic tumors, the normal brain to whole blood ratio following a single MK-1775 dose was 5%, and MALDI-MS imaging demonstrated heterogeneous and markedly lower MK-1775 distribution in orthotopic as compared with heterotopic GBM22 tumors. CONCLUSIONS: Limited distribution to brain tumors may limit the efficacy of MK-1775 in GBM.


Subject(s)
Blood-Brain Barrier/metabolism , Dacarbazine/analogs & derivatives , Glioblastoma/metabolism , Glioblastoma/pathology , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Animals , Cell Cycle Proteins/antagonists & inhibitors , DNA Damage/drug effects , Dacarbazine/pharmacokinetics , Dacarbazine/pharmacology , Disease Models, Animal , Glioblastoma/drug therapy , Glioblastoma/mortality , Humans , Mice , Nuclear Proteins/antagonists & inhibitors , Protein-Tyrosine Kinases/antagonists & inhibitors , Pyrazoles/pharmacokinetics , Pyrimidines/pharmacokinetics , Pyrimidinones , Temozolomide , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
18.
Proc Natl Acad Sci U S A ; 111(42): 15184-9, 2014 Oct 21.
Article in English | MEDLINE | ID: mdl-25246570

ABSTRACT

Distinguishing tumor from normal glandular breast tissue is an important step in breast-conserving surgery. Because this distinction can be challenging in the operative setting, up to 40% of patients require an additional operation when traditional approaches are used. Here, we present a proof-of-concept study to determine the feasibility of using desorption electrospray ionization mass spectrometry imaging (DESI-MSI) for identifying and differentiating tumor from normal breast tissue. We show that tumor margins can be identified using the spatial distributions and varying intensities of different lipids. Several fatty acids, including oleic acid, were more abundant in the cancerous tissue than in normal tissues. The cancer margins delineated by the molecular images from DESI-MSI were consistent with those margins obtained from histological staining. Our findings prove the feasibility of classifying cancerous and normal breast tissues using ambient ionization MSI. The results suggest that an MS-based method could be developed for the rapid intraoperative detection of residual cancer tissue during breast-conserving surgery.


Subject(s)
Breast Neoplasms, Male/pathology , Breast Neoplasms/pathology , Neoplasms/pathology , Spectrometry, Mass, Electrospray Ionization , Adult , Aged , Biomarkers, Tumor , Breast Neoplasms/metabolism , Breast Neoplasms, Male/metabolism , Fatty Acids/chemistry , Female , Humans , Lipids/chemistry , Male , Mastectomy , Middle Aged , Neoplasms/metabolism , Oleic Acid/chemistry , Recurrence
19.
Proc Natl Acad Sci U S A ; 111(30): 11121-6, 2014 Jul 29.
Article in English | MEDLINE | ID: mdl-24982150

ABSTRACT

For many intraoperative decisions surgeons depend on frozen section pathology, a technique developed over 150 y ago. Technical innovations that permit rapid molecular characterization of tissue samples at the time of surgery are needed. Here, using desorption electrospray ionization (DESI) MS, we rapidly detect the tumor metabolite 2-hydroxyglutarate (2-HG) from tissue sections of surgically resected gliomas, under ambient conditions and without complex or time-consuming preparation. With DESI MS, we identify isocitrate dehydrogenase 1-mutant tumors with both high sensitivity and specificity within minutes, immediately providing critical diagnostic, prognostic, and predictive information. Imaging tissue sections with DESI MS shows that the 2-HG signal overlaps with areas of tumor and that 2-HG levels correlate with tumor content, thereby indicating tumor margins. Mapping the 2-HG signal onto 3D MRI reconstructions of tumors allows the integration of molecular and radiologic information for enhanced clinical decision making. We also validate the methodology and its deployment in the operating room: We have installed a mass spectrometer in our Advanced Multimodality Image Guided Operating (AMIGO) suite and demonstrate the molecular analysis of surgical tissue during brain surgery. This work indicates that metabolite-imaging MS could transform many aspects of surgical care.


Subject(s)
Brain Neoplasms , Glioma , Glutarates/metabolism , Intraoperative Care/methods , Magnetic Resonance Imaging , Mass Spectrometry/methods , Brain Neoplasms/diagnostic imaging , Brain Neoplasms/metabolism , Brain Neoplasms/therapy , Female , Glioma/diagnostic imaging , Glioma/metabolism , Glioma/surgery , Humans , Male , Mass Spectrometry/instrumentation , Radiography
20.
Cell Metab ; 20(2): 280-94, 2014 Aug 05.
Article in English | MEDLINE | ID: mdl-25017943

ABSTRACT

The molecular mechanisms responsible for the failure of antiangiogenic therapies and how tumors adapt to these therapies are unclear. Here, we applied transcriptomic, proteomic, and metabolomic approaches to preclinical models and provide evidence for tumor adaptation to vascular endothelial growth factor blockade through a metabolic shift toward carbohydrate and lipid metabolism in tumors. During sunitinib or sorafenib treatment, tumor growth was inhibited and tumors were hypoxic and glycolytic. In sharp contrast, treatment withdrawal led to tumor regrowth, angiogenesis restoration, moderate lactate production, and enhanced lipid synthesis. This metabolic shift was associated with a drastic increase in metastatic dissemination. Interestingly, pharmacological lipogenesis inhibition with orlistat or fatty acid synthase downregulation with shRNA inhibited tumor regrowth and metastases after sunitinib treatment withdrawal. Our data shed light on metabolic alterations that result in cancer adaptation to antiangiogenic treatments and identify key molecules involved in lipid metabolism as putative therapeutic targets.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Lipids/biosynthesis , Neoplasms/drug therapy , Animals , Cell Line, Tumor , Disease Progression , Fatty Acid Synthases/antagonists & inhibitors , Fatty Acid Synthases/genetics , Fatty Acid Synthases/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Indoles/therapeutic use , Metabolomics , Mice , Mice, Inbred C57BL , Mice, Knockout , Neoplasm Metastasis , Neoplasms/metabolism , Neoplasms/pathology , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/metabolism , Niacinamide/analogs & derivatives , Niacinamide/therapeutic use , Phenylurea Compounds/therapeutic use , Proteomics , Pyrroles/therapeutic use , RNA Interference , Sorafenib , Sunitinib , Transplantation, Heterologous
SELECTION OF CITATIONS
SEARCH DETAIL