Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 82
Filter
1.
Ann Allergy Asthma Immunol ; 132(5): 585-591, 2024 May.
Article in English | MEDLINE | ID: mdl-38110056

ABSTRACT

Medical evaluation for military applicants is an intricate process that requires an understanding of the terminology, standards, and guidelines. Allergy providers are often called to provide medical evaluations for patients who desire to join the military services. Without understanding the complexities and nuances of military medical evaluations, a provider may delay or not be able to assist their patient in obtaining the desired goal of joining the services. This article reviews the terminology of military medical evaluations and the guidelines and processes for these evaluations. We also focus our discussion on common allergic conditions that may be disqualifying for service and provide expert opinions of the subtleties of these conditions to provide the allergist with a practical approach to medical evaluations. Finally, we provide a list of resources that are accessible to any provider engaged in military medical evaluations for accessions.


Subject(s)
Hypersensitivity , Military Personnel , Humans , Hypersensitivity/diagnosis , Practice Guidelines as Topic
2.
Nat Commun ; 14(1): 5051, 2023 08 19.
Article in English | MEDLINE | ID: mdl-37598220

ABSTRACT

Histone deacetylases are important epigenetic regulators that have been reported to play essential roles in cancer stem cell functions and are promising therapeutic targets in many cancers including glioblastoma. However, the functionally relevant roles of specific histone deacetylases, in the maintenance of key self-renewal and growth characteristics of brain tumour stem cell (BTSC) sub-populations of glioblastoma, remain to be fully resolved. Here, using pharmacological inhibition and genetic loss and gain of function approaches, we identify HDAC2 as the most relevant histone deacetylase for re-organization of chromatin accessibility resulting in maintenance of BTSC growth and self-renewal properties. Furthermore, its specific interaction with the transforming growth factor-ß pathway related proteins, SMAD3 and SKI, is crucial for the maintenance of tumorigenic potential in BTSCs in vitro and in orthotopic xenograft models. Inhibition of HDAC2 activity and disruption of the coordinated mechanisms regulated by the HDAC2-SMAD3-SKI axis are thus promising therapeutic approaches for targeting BTSCs.


Subject(s)
Brain Stem Neoplasms , Glioblastoma , Humans , Glioblastoma/genetics , Brain , Histone Deacetylases/genetics , Neoplastic Stem Cells , Epigenesis, Genetic , Smad3 Protein/genetics , Histone Deacetylase 2/genetics
3.
Cell Chem Biol ; 30(7): 780-794.e8, 2023 07 20.
Article in English | MEDLINE | ID: mdl-37379846

ABSTRACT

Overlapping principles of embryonic and tumor biology have been described, with recent multi-omics campaigns uncovering shared molecular profiles between human pluripotent stem cells (hPSCs) and adult tumors. Here, using a chemical genomic approach, we provide biological evidence that early germ layer fate decisions of hPSCs reveal targets of human cancers. Single-cell deconstruction of hPSCs-defined subsets that share transcriptional patterns with transformed adult tissues. Chemical screening using a unique germ layer specification assay for hPSCs identified drugs that enriched for compounds that selectively suppressed the growth of patient-derived tumors corresponding exclusively to their germ layer origin. Transcriptional response of hPSCs to germ layer inducing drugs could be used to identify targets capable of regulating hPSC specification as well as inhibiting adult tumors. Our study demonstrates properties of adult tumors converge with hPSCs drug induced differentiation in a germ layer specific manner, thereby expanding our understanding of cancer stemness and pluripotency.


Subject(s)
Neoplasms , Pluripotent Stem Cells , Humans , Cell Differentiation/physiology , Neoplasms/drug therapy , Neoplasms/genetics , Genomics
4.
Ann Allergy Asthma Immunol ; 130(1): 40-45, 2023 01.
Article in English | MEDLINE | ID: mdl-35973653

ABSTRACT

BACKGROUND: Sesame allergy has been characterized in the Middle East for some time. However, it has become more widely recognized as foods containing sesame and sesame seeds have become increasingly available in Australia, Europe, and North America. With the passage of the Food Allergy Safety, Treatment, Education, and Research Act in 2021, the United States will join other countries in identifying sesame as a major food allergen and require sesame to be labeled as a food allergen beginning in 2023. OBJECTIVE: To review the literature related to sesame allergy as an increasingly recognized food allergen around the world. DATA SOURCES: English-language articles retrieved by PubMed searches with relevance to sesame allergy. STUDY SELECTIONS: Articles were included using the search terms "sesame allergy" and "sesame seed allergy." RESULTS: A total of 69 relevant articles regarding sesame allergy, relating to its prevalence, clinical presentation, natural history, allergenic epitopes, diagnosis, and treatment, were selected. CONCLUSION: In recent decades, considerable gains have been made in determining prevalence and natural history of sesame allergy. With increased recognition and prevalence come the need for reliable methods of identification of sesame allergy and approaches for management.


Subject(s)
Food Hypersensitivity , Sesamum , Humans , Allergens , Sesamum/adverse effects , Food Hypersensitivity/diagnosis , Food Hypersensitivity/epidemiology , Food Hypersensitivity/therapy , Europe , North America/epidemiology
5.
Mil Med ; 188(1-2): e421-e425, 2023 01 04.
Article in English | MEDLINE | ID: mdl-33748854

ABSTRACT

We present the case of a patient who was unable to tolerate rapid drug desensitization protocol to receive a continuous penicillin (PCN) G infusion for the treatment of neurosyphilis. A 38-year-old male with past medical history for human immunodeficiency virus, migraines, PCN allergy, doxycycline allergy, shellfish allergy, and untreated latent syphilis presented to the emergency room for a posterior migraine with associated nausea, vomiting, photophobia, right-sided paresthesias, and "shaky" vision. He was diagnosed with neurosyphilis and underwent rapid drug desensitization with the goal to receive a continuous infusion of PCN G. The patient's hospital course was complicated by intermittent drug reactions consisting of tachycardia, rash, and dyspnea, followed by periods of being able to tolerate the infusion. After being able to tolerate the recommended dose of PCN infusion, the patient was discharged home to complete the course. However, he returned almost immediately after a recurrence of symptoms at home requiring the use of intramuscular epinephrine. Ultimately, the patient was transitioned to ceftriaxone and completed the infusion course as an inpatient because of continued intermittent recurrence of drug reaction symptoms.


Subject(s)
Drug Hypersensitivity , Drug-Related Side Effects and Adverse Reactions , Hypersensitivity , Neurosyphilis , Male , Humans , Adult , Drug Hypersensitivity/complications , Drug Hypersensitivity/diagnosis , Penicillins/therapeutic use , Neurosyphilis/complications , Neurosyphilis/diagnosis , Neurosyphilis/drug therapy , Ceftriaxone/therapeutic use , Drug-Related Side Effects and Adverse Reactions/complications , Hypersensitivity/complications
6.
Cureus ; 13(10): e18969, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34812330

ABSTRACT

INTRODUCTION:  The COVID-19 pandemic taught many lessons regarding vaccine manufacturing, transportation, and delivery. Throughout the vaccination campaign, different vaccination sites reported that six or seven doses of the Pfizer/BioNTech COVID-19 vaccine could be reliably withdrawn from each multidose vial. This discrepancy was hypothesized to be caused by the differences in needle and syringe dead space volumes with differing supplies utilized at each site, but independent data regarding these volumes are lacking; therefore, we sought to objectively evaluate the volume of dead space for supplies commonly used for immunizations. MATERIALS AND METHODS: Available needles and syringes of different brands and specifications that could be used for administering vaccines were gathered. Each needle and/or syringe was weighed with a scientific, calibrated scale upon removal from the manufacturer's packaging and then filled with distilled water to simulate standard vaccine preparation. The water was then expelled, simulating vaccination, and then reweighed on the same scientific scale. The difference between the two weights was secondary to the water remaining within the needle or syringe after the simulated use, which was equivalent to the dead space volume. RESULTS: Manufacturer design, gauge, needle length, and syringe volume separately correlate with volume differences. Intuitively, the needles and syringes with the most dead space were consistently the 1.5-inch needles and the larger volume syringes, regardless of the manufacturer. CONCLUSION: The withdrawal of individual vaccine doses from a multidose vial inevitably results in vaccine volume loss due to the dead space of needles and syringes. As such, reliably obtaining seven doses of the Pfizer/BioNTech COVID-19 vaccine is improbable without utilizing specialized low dead space supplies.

7.
Fed Pract ; 38(1): 44-48, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33574649

ABSTRACT

This case study suggests that omalizumab may help prevent anaphylaxis and reduce disease burden associated with systemic mastocytosis, but further studies and formal clinical trials are needed to confirm these findings.

8.
Nat Commun ; 12(1): 979, 2021 02 12.
Article in English | MEDLINE | ID: mdl-33579912

ABSTRACT

Glioblastoma (GBM) is a deadly cancer in which cancer stem cells (CSCs) sustain tumor growth and contribute to therapeutic resistance. Protein arginine methyltransferase 5 (PRMT5) has recently emerged as a promising target in GBM. Using two orthogonal-acting inhibitors of PRMT5 (GSK591 or LLY-283), we show that pharmacological inhibition of PRMT5 suppresses the growth of a cohort of 46 patient-derived GBM stem cell cultures, with the proneural subtype showing greater sensitivity. We show that PRMT5 inhibition causes widespread disruption of splicing across the transcriptome, particularly affecting cell cycle gene products. We identify a GBM splicing signature that correlates with the degree of response to PRMT5 inhibition. Importantly, we demonstrate that LLY-283 is brain-penetrant and significantly prolongs the survival of mice with orthotopic patient-derived xenografts. Collectively, our findings provide a rationale for the clinical development of brain penetrant PRMT5 inhibitors as treatment for GBM.


Subject(s)
Antineoplastic Agents/pharmacology , Brain Neoplasms/metabolism , Glioblastoma/metabolism , Protein-Arginine N-Methyltransferases/metabolism , Animals , Apoptosis , Brain Neoplasms/drug therapy , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Cell Cycle , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Discovery , Epigenomics , Female , Gene Expression Regulation, Neoplastic , Glioblastoma/drug therapy , Glioblastoma/genetics , Glioblastoma/pathology , Humans , Mice , Neoplastic Stem Cells/metabolism , Protein-Arginine N-Methyltransferases/drug effects , Protein-Arginine N-Methyltransferases/genetics , RNA Splicing , Xenograft Model Antitumor Assays
9.
Elife ; 102021 01 11.
Article in English | MEDLINE | ID: mdl-33427645

ABSTRACT

Chromatin accessibility discriminates stem from mature cell populations, enabling the identification of primitive stem-like cells in primary tumors, such as glioblastoma (GBM) where self-renewing cells driving cancer progression and recurrence are prime targets for therapeutic intervention. We show, using single-cell chromatin accessibility, that primary human GBMs harbor a heterogeneous self-renewing population whose diversity is captured in patient-derived glioblastoma stem cells (GSCs). In-depth characterization of chromatin accessibility in GSCs identifies three GSC states: Reactive, Constructive, and Invasive, each governed by uniquely essential transcription factors and present within GBMs in varying proportions. Orthotopic xenografts reveal that GSC states associate with survival, and identify an invasive GSC signature predictive of low patient survival, in line with the higher invasive properties of Invasive state GSCs compared to Reactive and Constructive GSCs as shown by in vitro and in vivo assays. Our chromatin-driven characterization of GSC states improves prognostic precision and identifies dependencies to guide combination therapies.


Subject(s)
Cell Self Renewal , Chromatin/metabolism , Glioblastoma/secondary , Neoplastic Stem Cells/physiology , Cell Line, Tumor , Female , Humans , Male , Single-Cell Analysis
10.
Nat Cancer ; 2(2): 157-173, 2021 02.
Article in English | MEDLINE | ID: mdl-35122077

ABSTRACT

Glioblastomas harbor diverse cell populations, including rare glioblastoma stem cells (GSCs) that drive tumorigenesis. To characterize functional diversity within this population, we performed single-cell RNA sequencing on >69,000 GSCs cultured from the tumors of 26 patients. We observed a high degree of inter- and intra-GSC transcriptional heterogeneity that could not be fully explained by DNA somatic alterations. Instead, we found that GSCs mapped along a transcriptional gradient spanning two cellular states reminiscent of normal neural development and inflammatory wound response. Genome-wide CRISPR-Cas9 dropout screens independently recapitulated this observation, with each state characterized by unique essential genes. Further single-cell RNA sequencing of >56,000 malignant cells from primary tumors found that the majority organize along an orthogonal astrocyte maturation gradient yet retain expression of founder GSC transcriptional programs. We propose that glioblastomas grow out of a fundamental GSC-based neural wound response transcriptional program, which is a promising target for new therapy development.


Subject(s)
Brain Neoplasms , Glioblastoma , Brain Neoplasms/genetics , Carcinogenesis/genetics , Glioblastoma/genetics , Humans , Neoplastic Stem Cells/metabolism
11.
Cancer Res ; 80(24): 5478-5490, 2020 12 15.
Article in English | MEDLINE | ID: mdl-33106333

ABSTRACT

Cancer cells can metabolize glutamine to replenish TCA cycle intermediates, leading to a dependence on glutaminolysis for cell survival. However, a mechanistic understanding of the role that glutamine metabolism has on the survival of glioblastoma (GBM) brain tumor stem cells (BTSC) has not yet been elucidated. Here, we report that across a panel of 19 GBM BTSC lines, inhibition of glutaminase (GLS) showed a variable response from complete blockade of cell growth to absolute resistance. Surprisingly, BTSC sensitivity to GLS inhibition was a result of reduced intracellular glutamate triggering the amino acid deprivation response (AADR) and not due to the contribution of glutaminolysis to the TCA cycle. Moreover, BTSC sensitivity to GLS inhibition negatively correlated with expression of the astrocytic glutamate transporters EAAT1 and EAAT2. Blocking glutamate transport in BTSCs with high EAAT1/EAAT2 expression rendered cells susceptible to GLS inhibition, triggering the AADR and limiting cell growth. These findings uncover a unique metabolic vulnerability in BTSCs and support the therapeutic targeting of upstream activators and downstream effectors of the AADR pathway in GBM. Moreover, they demonstrate that gene expression patterns reflecting the cellular hierarchy of the tissue of origin can alter the metabolic requirements of the cancer stem cell population. SIGNIFICANCE: Glioblastoma brain tumor stem cells with low astrocytic glutamate transporter expression are dependent on GLS to maintain intracellular glutamate to prevent the amino acid deprivation response and cell death.


Subject(s)
Amino Acids/metabolism , Brain Neoplasms/metabolism , Glioblastoma/metabolism , Glutaminase/metabolism , Neoplastic Stem Cells/metabolism , Signal Transduction/drug effects , Astrocytes/metabolism , Benzeneacetamides/pharmacology , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Citric Acid Cycle/drug effects , Excitatory Amino Acid Transporter 1/metabolism , Excitatory Amino Acid Transporter 2/metabolism , Glioblastoma/pathology , Glutamic Acid/metabolism , Glutaminase/antagonists & inhibitors , Humans , Thiadiazoles/pharmacology
12.
Neuro Oncol ; 22(8): 1150-1161, 2020 08 17.
Article in English | MEDLINE | ID: mdl-32296841

ABSTRACT

BACKGROUND: Imagining ways to prevent or treat glioblastoma (GBM) has been hindered by a lack of understanding of its pathogenesis. Although overexpression of platelet derived growth factor with two A-chains (PDGF-AA) may be an early event, critical details of the core biology of GBM are lacking. For example, existing PDGF-driven models replicate its microscopic appearance, but not its genomic architecture. Here we report a model that overcomes this barrier to authenticity. METHODS: Using a method developed to establish neural stem cell cultures, we investigated the effects of PDGF-AA on subventricular zone (SVZ) cells, one of the putative cells of origin of GBM. We microdissected SVZ tissue from p53-null and wild-type adult mice, cultured cells in media supplemented with PDGF-AA, and assessed cell viability, proliferation, genome stability, and tumorigenicity. RESULTS: Counterintuitive to its canonical role as a growth factor, we observed abrupt and massive cell death in PDGF-AA: wild-type cells did not survive, whereas a small fraction of null cells evaded apoptosis. Surviving null cells displayed attenuated proliferation accompanied by whole chromosome gains and losses. After approximately 100 days in PDGF-AA, cells suddenly proliferated rapidly, acquired growth factor independence, and became tumorigenic in immune-competent mice. Transformed cells had an oligodendrocyte precursor-like lineage marker profile, were resistant to platelet derived growth factor receptor alpha inhibition, and harbored highly abnormal karyotypes similar to human GBM. CONCLUSION: This model associates genome instability in neural progenitor cells with chronic exposure to PDGF-AA and is the first to approximate the genomic landscape of human GBM and the first in which the earliest phases of the disease can be studied directly.


Subject(s)
Brain Neoplasms , Glioblastoma , Neural Stem Cells , Platelet-Derived Growth Factor , Tumor Suppressor Protein p53 , Animals , Brain Neoplasms/chemically induced , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cells, Cultured , Glioblastoma/chemically induced , Glioblastoma/genetics , Glioblastoma/metabolism , Glioblastoma/pathology , Lateral Ventricles/drug effects , Lateral Ventricles/metabolism , Lateral Ventricles/pathology , Mice , Mice, Inbred C57BL , Neural Stem Cells/drug effects , Neural Stem Cells/pathology , Platelet-Derived Growth Factor/pharmacology , Tumor Suppressor Protein p53/deficiency , Tumor Suppressor Protein p53/metabolism
13.
Neurooncol Adv ; 2(1): vdaa020, 2020.
Article in English | MEDLINE | ID: mdl-32226941

ABSTRACT

BACKGROUND: The EGFR pathway is frequently mutated in glioblastoma (GBM). However, to date, EGFR therapies have not demonstrated efficacy in clinical trials. Poor brain penetration of conventional inhibitors, lack of patient stratification for EGFR status, and mechanisms of resistance are likely responsible for the failure of EGFR-targeted therapy. We aimed to address these elements in a large panel of molecularly diverse patient-derived GBM brain tumor stem cells (BTSCs). METHODS: In vitro growth inhibition and on-target efficacy of afatinib, pacritinib, or a combination were assessed by cell viability, neurosphere formation, cytotoxicity, limiting dilution assays, and western blotting. In vivo efficacy was assessed with mass spectrometry, immunohistochemistry, magnetic resonance imaging, and intracranial xenograft models. RESULTS: We show that afatinib and pacritinib decreased BTSC growth and sphere-forming capacity in vitro. Combinations of the 2 drugs were synergistic and abrogated the activation of STAT3 signaling observed upon EGFR inhibition in vitro and in vivo. We further demonstrate that the brain-penetrant EGFR inhibitor, afatinib, improved survival in EGFRvIII mt orthotopic xenograft models. However, upregulation of the oncogenic STAT3 signaling pathway was observed following afatinib treatment. Combined inhibition with 2 clinically relevant drugs, afatinib and pacritinib, synergistically decreased BTSC viability and abrogated this compensatory mechanism of resistance to EGFR inhibition. A significant decrease in tumor burden in vivo was observed with the combinatorial treatment. CONCLUSIONS: These data demonstrate that brain-penetrant combinatorial therapies targeting the EGFR and STAT3 signaling pathways hold therapeutic promise for GBM.

14.
Mil Med ; 185(5-6): e912-e913, 2020 06 08.
Article in English | MEDLINE | ID: mdl-31819972

ABSTRACT

Omalizumab has been safely used to treat asthma and urticaria. We report a case of serum sickness-like reaction in a patient treated with omalizumab for chronic idiopathic urticaria/angioedema. An adult female experienced episodic urticaria/angioedema without repeatable trigger, ultimately receiving diagnosis of chronic idiopathic urticaria/angioedema. After initial treatment, attempts with escalating cetirizine and montelukast doses were unsuccessful due to sedation; she began treatment with subcutaneous omalizumab 150 mg monthly. Urticaria frequency partially improved after two injections; therefore, the dose was increased to 300 mg after four treatments. Several days after first 300 mg dose, she reported abdominal cramping, fatigue, fever, lymphadenopathy, and arthralgia. Aside from mild thrombocytosis, inflammatory markers were unremarkable, as were evaluations for infection, autoimmunity, and malignancy. Omalizumab was held with eventual improvement in symptoms, which did not return after discontinuation. Omalizumab is a helpful medication in treating atopic conditions, with at least theoretical risk of immune complex formation and tissue deposition causing serum sickness-like reaction. Although early publications showed very low adverse event rates, there have now been reports of serum sickness-like reactions in children and adults treated for asthma and urticaria. Determining true incidence is difficult, given rarity and non-specific nature. Previous reports described symptoms with initiation of medication, reproducible after reintroduction. While it remains to be determined which factors increase risk for serum sickness-like reaction to omalizumab, our report of an urticaria patient who exhibited symptoms with increasing dose contributes insight into the discussion regarding this adverse effect of an otherwise well-tolerated and important medication.


Subject(s)
Omalizumab/adverse effects , Serum Sickness , Adult , Anti-Allergic Agents/adverse effects , Chronic Disease , Female , Humans , Serum Sickness/chemically induced , Serum Sickness/drug therapy , Urticaria/chemically induced , Urticaria/drug therapy
15.
Invest New Drugs ; 38(4): 1137-1144, 2020 08.
Article in English | MEDLINE | ID: mdl-31707687

ABSTRACT

The PI3K/AKT/mTOR pathway activation plays a central role in glioblastoma multiforme (GBM) development and progression, and in resistance to anti-cancer therapies. Inhibition of the PI3K pathway has been shown to sensitize cultured glioma cells and tumor xenografts to the effects of temozolomide (TMZ) and radiation. Vistusertib is an oral inhibitor of mTORC1/2 complexes. The primary objective of this Canadian Cancer Trials Group phase I study was to determine the recommended phase II dose (RP2D) of vistusertib in patients with GBM receiving TMZ at first progression following primary treatment. Vistusertib was administered at a starting dose of 100 mg bid 2 days on/5 days off weekly with TMZ 150 mg/m2 daily for 5 days/28-days cycle. Dose escalation was according to a 3 + 3 design. Secondary objectives included assessment of vistusertib safety and toxicity profile, and preliminary efficacy. 15 patients were enrolled in the study (median age 66 (range 51-77), females 8). Vistusertib 125 mg BID in combination with TMZ 150 mg/m2 daily for 5 days was well tolerated. Vistusertib treatment-related adverse events were generally grade 1-2, with the most frequently reported being fatigue, gastrointestinal symptoms, and rash. Of 13 response evaluable patients, 1 patient (8%) had a partial response ongoing at 7.6 months of follow-up, and 5 patients had stable disease (38%) as best response (median duration 9.6 months, range 3.7-not yet reached). Six-month progression-free survival (PFS) rate was 26.6%. Combination of vistusertib with TMZ in GBM patients at first recurrence demonstrated a favorable safety profile at the tested dose levels.


Subject(s)
Antineoplastic Agents/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Benzamides/administration & dosage , Brain Neoplasms/drug therapy , Glioblastoma/drug therapy , Mechanistic Target of Rapamycin Complex 1/antagonists & inhibitors , Mechanistic Target of Rapamycin Complex 2/antagonists & inhibitors , Morpholines/administration & dosage , Pyrimidines/administration & dosage , Temozolomide/administration & dosage , Aged , Antineoplastic Agents/adverse effects , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Benzamides/adverse effects , Female , Humans , Male , Maximum Tolerated Dose , Middle Aged , Morpholines/adverse effects , Pyrimidines/adverse effects , Temozolomide/adverse effects
16.
Cancers (Basel) ; 11(11)2019 Oct 24.
Article in English | MEDLINE | ID: mdl-31652994

ABSTRACT

In glioblastoma (GBM), brain tumor stem cells (BTSCs) encompass heterogenous populations of multipotent, self-renewing, and tumorigenic cells, which have been proposed to be at the root of therapeutic resistance and recurrence. While the functional significance of BTSC heterogeneity remains to be fully determined, we previously distinguished relatively quiescent stem-like precursor state from the more aggressive progenitor-like precursor state. In the present study, we hypothesized that progenitor-like BTSCs arise from stem-like precursors through a mesenchymal transition and drive post-treatment recurrence. We first demonstrate that progenitor-like BTSCs display a more mesenchymal transcriptomic profile. Moreover, we show that both mesenchymal GBMs and progenitor-like BTSCs are characterized by over-activated STAT3/EMT pathways and that SLUG is the primary epithelial to mesenchymal transition (EMT) transcription factor directly regulated by STAT3 in BTSCs. SLUG overexpression in BTSCs enhances invasiveness, promotes inflammation, and shortens survival. Importantly, SLUG overexpression in a quiescent stem-like BTSC line enhances tumorigenesis. Finally, we report that recurrence is associated with SLUG-induced transcriptional changes in both BTSCs and GBM patient samples. Collectively, our findings show that a STAT3-driven precursor state transition, mediated by SLUG, may prime BTSCs to initiate more aggressive mesenchymal recurrence. Targeting the STAT3/SLUG pathway may maintain BTSCs in a quiescent stem-like precursor state, delaying recurrence and improving survival in GBM.

17.
Proc Natl Acad Sci U S A ; 116(38): 19098-19108, 2019 09 17.
Article in English | MEDLINE | ID: mdl-31471491

ABSTRACT

Glioblastoma multiforme (GBM) is the most deadly brain tumor, and currently lacks effective treatment options. Brain tumor-initiating cells (BTICs) and orthotopic xenografts are widely used in investigating GBM biology and new therapies for this aggressive disease. However, the genomic characteristics and molecular resemblance of these models to GBM tumors remain undetermined. We used massively parallel sequencing technology to decode the genomes and transcriptomes of BTICs and xenografts and their matched tumors in order to delineate the potential impacts of the distinct growth environments. Using data generated from whole-genome sequencing of 201 samples and RNA sequencing of 118 samples, we show that BTICs and xenografts resemble their parental tumor at the genomic level but differ at the mRNA expression and epigenomic levels, likely due to the different growth environment for each sample type. These findings suggest that a comprehensive genomic understanding of in vitro and in vivo GBM model systems is crucial for interpreting data from drug screens, and can help control for biases introduced by cell-culture conditions and the microenvironment in mouse models. We also found that lack of MGMT expression in pretreated GBM is linked to hypermutation, which in turn contributes to increased genomic heterogeneity and requires new strategies for GBM treatment.


Subject(s)
Biomarkers, Tumor/genetics , Brain Neoplasms/pathology , Gene Expression Regulation, Neoplastic , Genomics/methods , Glioblastoma/pathology , Neoplastic Stem Cells/pathology , Tumor Microenvironment/genetics , Adult , Aged , Aged, 80 and over , Animals , Apoptosis , Brain Neoplasms/genetics , Case-Control Studies , Cell Proliferation , DNA Methylation , Drug Resistance, Neoplasm , Female , Gene Expression Profiling , Glioblastoma/genetics , Humans , Male , Mice , Mice, SCID , Middle Aged , Neoplastic Stem Cells/metabolism , Transcriptome , Tumor Cells, Cultured , Whole Genome Sequencing , Xenograft Model Antitumor Assays
18.
Cell Rep ; 27(3): 971-986.e9, 2019 04 16.
Article in English | MEDLINE | ID: mdl-30995489

ABSTRACT

Glioblastoma therapies have remained elusive due to limitations in understanding mechanisms of growth and survival of the tumorigenic population. Using CRISPR-Cas9 approaches in patient-derived GBM stem cells (GSCs) to interrogate function of the coding genome, we identify actionable pathways responsible for growth, which reveal the gene-essential circuitry of GBM stemness and proliferation. In particular, we characterize members of the SOX transcription factor family, SOCS3, USP8, and DOT1L, and protein ufmylation as important for GSC growth. Additionally, we reveal mechanisms of temozolomide resistance that could lead to combination strategies. By reaching beyond static genome analysis of bulk tumors, with a genome-wide functional approach, we reveal genetic dependencies within a broad range of biological processes to provide increased understanding of GBM growth and treatment resistance.


Subject(s)
Brain Neoplasms/pathology , CRISPR-Cas Systems/genetics , Gene Editing/methods , Glioblastoma/pathology , Neoplastic Stem Cells/metabolism , Temozolomide/pharmacology , Animals , Brain Neoplasms/drug therapy , Brain Neoplasms/mortality , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Resistance, Neoplasm/genetics , Endopeptidases/genetics , Endopeptidases/metabolism , Endosomal Sorting Complexes Required for Transport/genetics , Endosomal Sorting Complexes Required for Transport/metabolism , Female , Gene Expression Regulation, Neoplastic/drug effects , Gene Library , Glioblastoma/drug therapy , Glioblastoma/mortality , Histone Methyltransferases/metabolism , Humans , Mice , Mice, SCID , Neoplastic Stem Cells/drug effects , Suppressor of Cytokine Signaling 3 Protein/genetics , Suppressor of Cytokine Signaling 3 Protein/metabolism , Survival Analysis , Temozolomide/therapeutic use , Ubiquitin Thiolesterase/genetics , Ubiquitin Thiolesterase/metabolism
19.
J Med Chem ; 62(5): 2651-2665, 2019 03 14.
Article in English | MEDLINE | ID: mdl-30776234

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is an aggressive, incurable cancer with a 20% 1 year survival rate. While standard-of-care therapy can prolong life in a small fraction of cases, PDAC is inherently resistant to current treatments, and novel therapies are urgently required. Histone deacetylase (HDAC) inhibitors are effective in killing pancreatic cancer cells in in vitro PDAC studies, and although there are a few clinical studies investigating combination therapy including HDAC inhibitors, no HDAC drug or combination therapy with an HDAC drug has been approved for the treatment of PDAC. We developed an inhibitor of HDACs, AES-135, that exhibits nanomolar inhibitory activity against HDAC3, HDAC6, and HDAC11 in biochemical assays. In a three-dimensional coculture model, AES-135 kills low-passage patient-derived tumor spheroids selectively over surrounding cancer-associated fibroblasts and has excellent pharmacokinetic properties in vivo. In an orthotopic murine model of pancreatic cancer, AES-135 prolongs survival significantly, therefore representing a candidate for further preclinical testing.


Subject(s)
Benzamides/pharmacology , Histone Deacetylase Inhibitors/pharmacology , Hydrocarbons, Fluorinated/pharmacology , Hydroxamic Acids/chemistry , Pancreatic Neoplasms/drug therapy , Sulfonamides/pharmacology , Animals , Apoptosis/drug effects , Benzamides/chemistry , Benzamides/pharmacokinetics , Benzamides/therapeutic use , Cell Line, Tumor , Cell Proliferation/drug effects , Coculture Techniques , Disease Models, Animal , Histone Deacetylase Inhibitors/chemistry , Histone Deacetylase Inhibitors/pharmacokinetics , Histone Deacetylase Inhibitors/therapeutic use , Humans , Hydrocarbons, Fluorinated/chemistry , Hydrocarbons, Fluorinated/pharmacokinetics , Hydrocarbons, Fluorinated/therapeutic use , Mice , Pancreatic Neoplasms/pathology , Sulfonamides/chemistry , Sulfonamides/pharmacokinetics , Sulfonamides/therapeutic use
20.
Neuron ; 101(3): 370-374, 2019 02 06.
Article in English | MEDLINE | ID: mdl-30731060

ABSTRACT

Building on Canada's strong traditions in neuroscience and ethics, neuroethics provides a backbone for the evolving Canadian Brain Research Strategy (CBRS) that, from the outset, incorporates ethically responsible discoveries in brain science into clinical, societal, educational, and commercial innovation.


Subject(s)
Neurology/ethics , Neurosciences/ethics , Psychiatry/ethics , Canada , Humans , Neurology/standards , Neurosciences/standards , Practice Guidelines as Topic , Psychiatry/standards
SELECTION OF CITATIONS
SEARCH DETAIL
...