Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
Cell Metab ; 36(1): 62-77.e8, 2024 01 02.
Article in English | MEDLINE | ID: mdl-38134929

ABSTRACT

Glioblastoma (GBM) is a malignancy dominated by the infiltration of tumor-associated myeloid cells (TAMCs). Examination of TAMC metabolic phenotypes in mouse models and patients with GBM identified the de novo creatine metabolic pathway as a hallmark of TAMCs. Multi-omics analyses revealed that TAMCs surround the hypoxic peri-necrotic regions of GBM and express the creatine metabolic enzyme glycine amidinotransferase (GATM). Conversely, GBM cells located within these same regions are uniquely specific in expressing the creatine transporter (SLC6A8). We hypothesized that TAMCs provide creatine to tumors, promoting GBM progression. Isotopic tracing demonstrated that TAMC-secreted creatine is taken up by tumor cells. Creatine supplementation protected tumors from hypoxia-induced stress, which was abrogated with genetic ablation or pharmacologic inhibition of SLC6A8. Lastly, inhibition of creatine transport using the clinically relevant compound, RGX-202-01, blunted tumor growth and enhanced radiation therapy in vivo. This work highlights that myeloid-to-tumor transfer of creatine promotes tumor growth in the hypoxic niche.


Subject(s)
Glioblastoma , Mice , Animals , Humans , Glioblastoma/metabolism , Creatine , Hypoxia/metabolism , Myeloid Cells/metabolism , Myeloid Progenitor Cells , Cell Line, Tumor
2.
Clin Cancer Res ; 29(23): 4973-4989, 2023 12 01.
Article in English | MEDLINE | ID: mdl-37725593

ABSTRACT

PURPOSE: Glioblastoma (GBM) is the most common aggressive primary malignant brain tumor in adults with a median age of onset of 68 to 70 years old. Although advanced age is often associated with poorer GBM patient survival, the predominant source(s) of maladaptive aging effects remains to be established. Here, we studied intratumoral and extratumoral relationships between adult patients with GBM and mice with brain tumors across the lifespan. EXPERIMENTAL DESIGN: Electronic health records at Northwestern Medicine and the NCI SEER databases were evaluated for GBM patient age and overall survival. The commercial Tempus and Caris databases, as well as The Cancer Genome Atlas were profiled for gene expression, DNA methylation, and mutational changes with varying GBM patient age. In addition, gene expression analysis was performed on the extratumoral brain of younger and older adult mice with or without a brain tumor. The survival of young and old wild-type or transgenic (INK-ATTAC) mice with a brain tumor was evaluated after treatment with or without senolytics and/or immunotherapy. RESULTS: Human patients with GBM ≥65 years of age had a significantly decreased survival compared with their younger counterparts. While the intra-GBM molecular profiles were similar between younger and older patients with GBM, non-tumor brain tissue had a significantly different gene expression profile between young and old mice with a brain tumor and the eradication of senescent cells improved immunotherapy-dependent survival of old but not young mice. CONCLUSIONS: This work suggests a potential benefit for combining senolytics with immunotherapy in older patients with GBM.


Subject(s)
Brain Neoplasms , Glioblastoma , Humans , Animals , Mice , Aged , Glioblastoma/drug therapy , Glioblastoma/genetics , Glioblastoma/metabolism , Senotherapeutics , Brain Neoplasms/drug therapy , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Mutation , DNA Methylation
3.
Nat Commun ; 14(1): 1610, 2023 03 23.
Article in English | MEDLINE | ID: mdl-36959214

ABSTRACT

As a key component of the standard of care for glioblastoma, radiotherapy induces several immune resistance mechanisms, such as upregulation of CD47 and PD-L1. Here, leveraging these radiotherapy-elicited processes, we generate a bridging-lipid nanoparticle (B-LNP) that engages tumor-associated myeloid cells (TAMCs) to glioblastoma cells via anti-CD47/PD-L1 dual ligation. We show that the engager B-LNPs block CD47 and PD-L1 and promote TAMC phagocytic activity. To enhance subsequent T cell recruitment and antitumor responses after tumor engulfment, the B-LNP was encapsulated with diABZI, a non-nucleotidyl agonist for stimulator of interferon genes. In vivo treatment with diABZI-loaded B-LNPs induced a transcriptomic and metabolic switch in TAMCs, turning these immunosuppressive cells into antitumor effectors, which induced T cell infiltration and activation in brain tumors. In preclinical murine models, B-LNP/diABZI administration synergized with radiotherapy to promote brain tumor regression and induce immunological memory against glioma. In summary, our study describes a nanotechnology-based approach that hijacks irradiation-triggered immune checkpoint molecules to boost potent and long-lasting antitumor immunity against glioblastoma.


Subject(s)
Brain Neoplasms , Glioblastoma , Nanoparticles , Animals , Humans , Mice , B7-H1 Antigen/metabolism , Brain Neoplasms/drug therapy , Brain Neoplasms/radiotherapy , CD47 Antigen/metabolism , Glioblastoma/drug therapy , Glioblastoma/radiotherapy , Interferons
4.
Nat Commun ; 14(1): 1566, 2023 03 22.
Article in English | MEDLINE | ID: mdl-36949040

ABSTRACT

Whereas the contribution of tumor microenvironment to the profound immune suppression of glioblastoma (GBM) is clear, tumor-cell intrinsic mechanisms that regulate resistance to CD8 T cell mediated killing are less understood. Kinases are potentially druggable targets that drive tumor progression and might influence immune response. Here, we perform an in vivo CRISPR screen to identify glioma intrinsic kinases that contribute to evasion of tumor cells from CD8 T cell recognition. The screen reveals checkpoint kinase 2 (Chek2) to be the most important kinase contributing to escape from CD8 T-cell recognition. Genetic depletion or pharmacological inhibition of Chek2 with blood-brain-barrier permeable drugs that are currently being evaluated in clinical trials, in combination with PD-1 or PD-L1 blockade, lead to survival benefit in multiple preclinical glioma models. Mechanistically, loss of Chek2 enhances antigen presentation, STING pathway activation and PD-L1 expression in mouse gliomas. Analysis of human GBMs demonstrates that Chek2 expression is inversely associated with antigen presentation and T-cell activation. Collectively, these results support Chek2 as a promising target for enhancement of response to immune checkpoint blockade therapy in GBM.


Subject(s)
Glioblastoma , Glioma , Humans , Animals , Mice , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , B7-H1 Antigen , Checkpoint Kinase 1 , Glioma/drug therapy , Glioma/genetics , Glioma/pathology , Glioblastoma/drug therapy , Glioblastoma/genetics , CD8-Positive T-Lymphocytes , Immunity , Tumor Microenvironment
5.
Res Sq ; 2023 Jan 09.
Article in English | MEDLINE | ID: mdl-36711497

ABSTRACT

Immunotherapy has revolutionized cancer treatment but has yet to be translated into brain tumors. Studies in other solid tumors suggest a central role of B-cell immunity in driving immune-checkpoint-blockade efficacy. Using single-cell and single-nuclei transcriptomics of human glioblastoma and melanoma brain metastasis, we found that tumor-associated B-cells have high expression of checkpoint molecules, known to block B-cell-receptor downstream effector function such as plasmablast differentiation and antigen-presentation. We also identified TGFß-1/TGFß receptor-2 interaction as a crucial modulator of B-cell suppression. Treatment of glioblastoma patients with pembrolizumab induced expression of B-cell checkpoint molecules and TGFß-receptor-2. Abrogation of TGFß using different conditional knockouts expanded germinal-center-like intratumoral B-cells, enhancing immune-checkpoint-blockade efficacy. Finally, blocking αVß8 integrin (which controls the release of active TGFß) and PD-1 significantly increased B-cell-dependent animal survival and immunological memory. Our study highlights the importance of intratumoral B-cell immunity and a remodeled approach to boost the effects of immunotherapy against brain tumors.

6.
Front Immunol ; 14: 1295218, 2023.
Article in English | MEDLINE | ID: mdl-38268923

ABSTRACT

Understanding the spatial relationship and functional interaction of immune cells in glioblastoma (GBM) is critical for developing new therapeutics that overcome the highly immunosuppressive tumor microenvironment. Our study showed that B and T cells form clusters within the GBM microenvironment within a 15-µm radius, suggesting that B and T cells could form immune synapses within the GBM. However, GBM-infiltrating B cells suppress the activation of CD8+ T cells. To overcome this immunosuppression, we leveraged B-cell functions by activating them with CD40 agonism, IFNγ, and BAFF to generate a potent antigen-presenting B cells named BVax. BVax had improved antigen cross-presentation potential compared to naïve B cells and were primed to use the IL15-IL15Ra mechanism to enhance T cell activation. Compared to naïve B cells, BVax could improve CD8 T cell activation and proliferation. Compared to dendritic cells (DCs), which are the current gold standard professional antigen-presenting cell, BVax promoted highly proliferative T cells in-vitro that had a stem-like memory T cell phenotype characterized by CD62L+CD44- expression, high TCF-1 expression, and low PD-1 and granzyme B expression. Adoptive transfer of BVax-activated CD8+ T cells into tumor-bearing brains led to T cell reactivation with higher TCF-1 expression and elevated granzyme B production compared to DC-activated CD8+ T cells. Adoptive transfer of BVax into an irradiated immunocompetent tumor-bearing host promoted more CD8+ T cell proliferation than adoptive transfer of DCs. Moreover, highly proliferative CD8+ T cells in the BVax group had less PD-1 expression than those highly proliferative CD8+ T cells in the DC group. The findings of this study suggest that BVax and DC could generate distinctive CD8+ T cells, which potentially serve multiple purposes in cellular vaccine development.


Subject(s)
Glioblastoma , Humans , Granzymes , CD8-Positive T-Lymphocytes , Programmed Cell Death 1 Receptor , Antigen-Presenting Cells , Cell Proliferation , Tumor Microenvironment
7.
Clin Cancer Res ; 28(14): 3156-3169, 2022 07 15.
Article in English | MEDLINE | ID: mdl-35552677

ABSTRACT

PURPOSE: Paclitaxel (PTX) is one of the most potent and commonly used chemotherapies for breast and pancreatic cancer. Several ongoing clinical trials are investigating means of enhancing delivery of PTX across the blood-brain barrier for glioblastomas. Despite the widespread use of PTX for breast cancer, and the initiative to repurpose this drug for gliomas, there are no predictive biomarkers to inform which patients will likely benefit from this therapy. EXPERIMENTAL DESIGN: To identify predictive biomarkers for susceptibility to PTX, we performed a genome-wide CRISPR knockout (KO) screen using human glioma cells. The genes whose KO was most enriched in the CRISPR screen underwent further selection based on their correlation with survival in the breast cancer patient cohorts treated with PTX and not in patients treated with other chemotherapies, a finding that was validated on a second independent patient cohort using progression-free survival. RESULTS: Combination of CRISPR screen results with outcomes from patients with taxane-treated breast cancer led to the discovery of endoplasmic reticulum (ER) protein SSR3 as a putative predictive biomarker for PTX. SSR3 protein levels showed positive correlation with susceptibility to PTX in breast cancer cells, glioma cells, and in multiple intracranial glioma xenografts models. KO of SSR3 turned the cells resistant to PTX while its overexpression sensitized the cells to PTX. Mechanistically, SSR3 confers susceptibility to PTX through regulation of phosphorylation of ER stress sensor IRE1α. CONCLUSIONS: Our hypothesis generating study showed SSR3 as a putative biomarker for susceptibility to PTX, warranting its prospective clinical validation.


Subject(s)
Antineoplastic Agents, Phytogenic , Biomarkers, Pharmacological , Brain Neoplasms , Breast Neoplasms , Calcium-Binding Proteins , Drug Resistance, Neoplasm , Glioblastoma , Membrane Glycoproteins , Paclitaxel , Receptors, Cytoplasmic and Nuclear , Receptors, Peptide , Animals , Antineoplastic Agents, Phytogenic/therapeutic use , Brain Neoplasms/drug therapy , Brain Neoplasms/genetics , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Calcium-Binding Proteins/genetics , Cell Line, Tumor , Drug Resistance, Neoplasm/genetics , Endoribonucleases/metabolism , Female , Glioblastoma/drug therapy , Glioblastoma/genetics , Humans , Membrane Glycoproteins/genetics , Mice , Paclitaxel/therapeutic use , Prospective Studies , Protein Serine-Threonine Kinases/metabolism , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Peptide/genetics , Xenograft Model Antitumor Assays
8.
Proc Natl Acad Sci U S A ; 119(1)2022 01 04.
Article in English | MEDLINE | ID: mdl-34969858

ABSTRACT

Brain metastases are a leading cause of death in patients with breast cancer. The lack of clinical trials and the presence of the blood-brain barrier limit therapeutic options. Furthermore, overexpression of the human epidermal growth factor receptor 2 (HER2) increases the incidence of breast cancer brain metastases (BCBM). HER2-targeting agents, such as the monoclonal antibodies trastuzumab and pertuzumab, improved outcomes in patients with breast cancer and extracranial metastases. However, continued BCBM progression in breast cancer patients highlighted the need for novel and effective targeted therapies against intracranial metastases. In this study, we engineered the highly migratory and brain tumor tropic human neural stem cells (NSCs) LM008 to continuously secrete high amounts of functional, stable, full-length antibodies against HER2 (anti-HER2Ab) without compromising the stemness of LM008 cells. The secreted anti-HER2Ab impaired tumor cell proliferation in vitro in HER2+ BCBM cells by inhibiting the PI3K-Akt signaling pathway and resulted in a significant benefit when injected in intracranial xenograft models. In addition, dual HER2 blockade using anti-HER2Ab LM008 NSCs and the tyrosine kinase inhibitor tucatinib significantly improved the survival of mice in a clinically relevant model of multiple HER2+ BCBM. These findings provide compelling evidence for the use of HER2Ab-secreting LM008 NSCs in combination with tucatinib as a promising therapeutic regimen for patients with HER2+ BCBM.


Subject(s)
Antineoplastic Agents, Immunological/metabolism , Brain Neoplasms , Neoplasms, Experimental , Neural Stem Cells , Oxazoles/pharmacology , Pyridines/pharmacology , Quinazolines/pharmacology , Receptor, ErbB-2 , Animals , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Brain Neoplasms/therapy , Cell Line, Tumor , Humans , Mice , Mice, Nude , Neoplasm Metastasis , Neoplasms, Experimental/genetics , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Neoplasms, Experimental/therapy , Neural Stem Cells/metabolism , Neural Stem Cells/pathology , Neural Stem Cells/transplantation , Receptor, ErbB-2/antagonists & inhibitors , Receptor, ErbB-2/metabolism , Xenograft Model Antitumor Assays
9.
Cells ; 10(9)2021 08 31.
Article in English | MEDLINE | ID: mdl-34571905

ABSTRACT

GBM is the most common primary brain tumor in adults, and the aggressive nature of this tumor contributes to its extremely poor prognosis. Over the years, the heterogeneous and adaptive nature of GBM has been highlighted as a major contributor to the poor efficacy of many treatments including various immunotherapies. The major challenge lies in understanding and manipulating the complex interplay among the different components within the tumor microenvironment (TME). This interplay varies not only by the type of cells interacting but also by their spatial distribution with the TME. This review highlights the various immune and non-immune components of the tumor microenvironment and their consequences f the efficacy of immunotherapies. Understanding the independent and interdependent aspects of the various sub-populations encapsulated by the immune and non-immune components will allow for more targeted therapies. Meanwhile, understanding how the TME creates and responds to different environmental pressures such as hypoxia may allow for other multimodal approaches in the treatment of GBM. Ultimately, a better understanding of the GBM TME will aid in the development and advancement of more effective treatments and in improving patient outcomes.


Subject(s)
Brain Neoplasms/immunology , Brain Neoplasms/pathology , Glioblastoma/immunology , Glioblastoma/pathology , Tumor Microenvironment/immunology , Animals , Humans
10.
Cancers (Basel) ; 13(18)2021 Sep 10.
Article in English | MEDLINE | ID: mdl-34572775

ABSTRACT

Glioblastoma (GBM) is a lethal primary brain tumor. Despite extensive effort in basic, translational, and clinical research, the treatment outcomes for patients with GBM are virtually unchanged over the past 15 years. GBM is one of the most immunologically "cold" tumors, in which cytotoxic T-cell infiltration is minimal, and myeloid infiltration predominates. This is due to the profound immunosuppressive nature of GBM, a tumor microenvironment that is metabolically challenging for immune cells, and the low mutational burden of GBMs. Together, these GBM characteristics contribute to the poor results obtained from immunotherapy. However, as indicated by an ongoing and expanding number of clinical trials, and despite the mostly disappointing results to date, immunotherapy remains a conceptually attractive approach for treating GBM. Checkpoint inhibitors, various vaccination strategies, and CAR T-cell therapy serve as some of the most investigated immunotherapeutic strategies. This review article aims to provide a general overview of the current state of glioblastoma immunotherapy. Information was compiled through a literature search conducted on PubMed and clinical trials between 1961 to 2021.

11.
Front Immunol ; 12: 637146, 2021.
Article in English | MEDLINE | ID: mdl-34025646

ABSTRACT

Glioblastoma (GBM) remains an aggressive brain tumor with a high rate of mortality. Immune checkpoint (IC) molecules are expressed on tumor infiltrating lymphocytes (TILs) and promote T cell exhaustion upon binding to IC ligands expressed by the tumor cells. Interfering with IC pathways with immunotherapy has promoted reactivation of anti-tumor immunity and led to success in several malignancies. However, IC inhibitors have achieved limited success in GBM patients, suggesting that other checkpoint molecules may be involved with suppressing TIL responses. Numerous IC pathways have been described, with current testing of inhibitors underway in multiple clinical trials. Identification of the most promising checkpoint pathways may be useful to guide the future trials for GBM. Here, we analyzed the The Cancer Genome Atlas (TCGA) transcriptomic database and identified PD1 and TIGIT as top putative targets for GBM immunotherapy. Additionally, dual blockade of PD1 and TIGIT improved survival and augmented CD8+ TIL accumulation and functions in a murine GBM model compared with either single agent alone. Furthermore, we demonstrated that this combination immunotherapy affected granulocytic/polymorphonuclear (PMN) myeloid derived suppressor cells (MDSCs) but not monocytic (Mo) MDSCs in in our murine gliomas. Importantly, we showed that suppressive myeloid cells express PD1, PD-L1, and TIGIT-ligands in human GBM tissue, and demonstrated that antigen specific T cell proliferation that is inhibited by immunosuppressive myeloid cells can be restored by TIGIT/PD1 blockade. Our data provide new insights into mechanisms of GBM αPD1/αTIGIT immunotherapy.


Subject(s)
Brain Neoplasms/immunology , Glioblastoma/immunology , Immune Checkpoint Inhibitors/pharmacology , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Receptors, Immunologic/antagonists & inhibitors , Animals , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/metabolism , CD8-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Cell Proliferation/drug effects , Humans , Immune Checkpoint Proteins/metabolism , Immunotherapy/methods , Lymphocytes, Tumor-Infiltrating/metabolism , Mice , Mice, Inbred C57BL , Myeloid-Derived Suppressor Cells/drug effects , Programmed Cell Death 1 Receptor/metabolism , Receptors, Immunologic/metabolism , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology
12.
Sci Adv ; 7(8)2021 02.
Article in English | MEDLINE | ID: mdl-33597238

ABSTRACT

Glioblastoma is characterized by the robust infiltration of immunosuppressive tumor-associated myeloid cells (TAMCs). It is not fully understood how TAMCs survive in the acidic tumor microenvironment to cause immunosuppression in glioblastoma. Metabolic and RNA-seq analysis of TAMCs revealed that the arginine-ornithine-polyamine axis is up-regulated in glioblastoma TAMCs but not in tumor-infiltrating CD8+ T cells. Active de novo synthesis of highly basic polyamines within TAMCs efficiently buffered low intracellular pH to support the survival of these immunosuppressive cells in the harsh acidic environment of solid tumors. Administration of difluoromethylornithine (DFMO), a clinically approved inhibitor of polyamine generation, enhanced animal survival in immunocompetent mice by causing a tumor-specific reduction of polyamines and decreased intracellular pH in TAMCs. DFMO combination with immunotherapy or radiotherapy further enhanced animal survival. These findings indicate that polyamines are used by glioblastoma TAMCs to maintain normal intracellular pH and cell survival and thus promote immunosuppression during tumor evolution.


Subject(s)
Glioblastoma , Animals , CD8-Positive T-Lymphocytes/metabolism , Cell Survival , Eflornithine/metabolism , Eflornithine/pharmacology , Glioblastoma/metabolism , Hydrogen-Ion Concentration , Immunosuppression Therapy , Mice , Myeloid Cells/metabolism , Polyamines/metabolism , Tumor Microenvironment
13.
J Neurotrauma ; 38(13): 1821-1826, 2021 Jun 01.
Article in English | MEDLINE | ID: mdl-33238820

ABSTRACT

Penetrating brain injury (PBI) is the most devastating type of traumatic brain injury. Development of coagulopathy in the acute setting of PBI, though common, remains of unclear significance as does its reversal. The aim of this study is to investigate the relationship between coagulopathy and clinical presentation, radiographical features, and outcome in civilian patients with PBI. Eighty-nine adult patients with PBI at a Level I trauma center in Chicago, Illinois who survived acute resuscitation and with available coagulation profile were analyzed. Coagulopathy was defined as international normalized ratio [INR] >1.3, platelet count <100,000 /µL, or partial thromboplastin time >37 sec. Median age (interquartile range; IQR) of our cohort was 27 (21-35) years, and 74 (83%) were male. The intent was assault in 74 cases (83%). The mechanism of PBI was gunshot wound in all patients. Forty patients (45%) were coagulopathic at presentation. In a multiple regression model, coagulopathy was associated with lower Glasgow Coma Scale (GCS)-Motor score (odds ratio [OR], 0.67; confidence interval [CI], 0.48-0.94; p = 0.02) and transfusion of blood products (OR, 3.91; CI, 1.2-12.5; p = 0.02). Effacement of basal cisterns was the only significant radiographical features associated with coagulopathy (OR, 3.34; CI, 1.08-10.37; p = 0.04). Mortality was found to be significantly more common in coagulopathic patients (73% vs. 25%; p < 0.001). However, in our limited sample, reversal of coagulopathy at 24 h was not associated with a statistically significant improvement in outcome. The triad of coagulopathy, low post-resuscitation GCS, and radiographical effacement of basal cisterns identify a particularly ominous phenotype of PBI. The role, and potential reversal of, coagulopathy in this group warrants further investigation.


Subject(s)
Blood Coagulation Disorders/diagnostic imaging , Brain Injuries, Traumatic/diagnostic imaging , Head Injuries, Penetrating/diagnostic imaging , Injury Severity Score , Adult , Biomarkers/blood , Blood Coagulation Disorders/blood , Blood Coagulation Disorders/epidemiology , Brain Injuries, Traumatic/blood , Brain Injuries, Traumatic/epidemiology , Cohort Studies , Female , Head Injuries, Penetrating/blood , Head Injuries, Penetrating/epidemiology , Humans , Male , Prospective Studies , Retrospective Studies , Young Adult
14.
Nat Genet ; 52(4): 371-377, 2020 04.
Article in English | MEDLINE | ID: mdl-32203465

ABSTRACT

Brain metastases from lung adenocarcinoma (BM-LUAD) frequently cause patient mortality. To identify genomic alterations that promote brain metastases, we performed whole-exome sequencing of 73 BM-LUAD cases. Using case-control analyses, we discovered candidate drivers of brain metastasis by identifying genes with more frequent copy-number aberrations in BM-LUAD compared to 503 primary LUADs. We identified three regions with significantly higher amplification frequencies in BM-LUAD, including MYC (12 versus 6%), YAP1 (7 versus 0.8%) and MMP13 (10 versus 0.6%), and significantly more frequent deletions in CDKN2A/B (27 versus 13%). We confirmed that the amplification frequencies of MYC, YAP1 and MMP13 were elevated in an independent cohort of 105 patients with BM-LUAD. Functional assessment in patient-derived xenograft mouse models validated the notion that MYC, YAP1 or MMP13 overexpression increased the incidence of brain metastasis. These results demonstrate that somatic alterations contribute to brain metastases and that genomic sequencing of a sufficient number of metastatic tumors can reveal previously unknown metastatic drivers.


Subject(s)
Adenocarcinoma of Lung/genetics , Brain Neoplasms/genetics , Lung Neoplasms/genetics , Neoplasm Metastasis/genetics , Adenocarcinoma of Lung/pathology , Animals , Brain Neoplasms/pathology , Case-Control Studies , Cell Line , DNA Copy Number Variations/genetics , Female , Genes, myc/genetics , Genomics/methods , HEK293 Cells , Humans , Lung Neoplasms/pathology , Male , Matrix Metalloproteinase 13/genetics , Mice , Mice, Nude , Mutation/genetics , Neoplasm Metastasis/pathology , Transcription Factors/genetics , Exome Sequencing
15.
J Neurol Surg B Skull Base ; 80(6): 562-567, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31750041

ABSTRACT

Objective Posterior fossa meningiomas are surgically challenging tumors that are associated with high morbidity and mortality. We sought to investigate the anatomical distribution of clinically actionable mutations in posterior fossa meningioma to facilitate identifying patients amenable for systemic targeted therapy trials. Methods Targeted sequencing of clinically targetable AKT1 , SMO , and PIK3CA mutations was performed in 61 posterior fossa meningioma using Illumina NextSeq 500 to a target depth of >500 × . Samples were further interrogated for 53 cancer-relevant RNA fusions by the Archer FusionPlex panel to detect gene rearrangements. Results AKT 1 ( E17K ) mutations were detected in five cases (8.2%), four in the foramen magnum and one in the cerebellopontine angle. In contrast, none of the posterior fossa tumors harbored an SMO ( L412F ) or a PIK3CA ( E545K ) mutation. Notably, the majority of foramen magnum meningiomas (4/7, 57%) harbored an AKT1 mutation. In addition, common clinically targetable gene fusions were not detected in any of the cases. Conclusion A large subset of foramen magnum meningiomas harbor AKT1 E17K mutations and are therefore potentially amenable to targeted medical therapy. Genotyping of foramen magnum meningiomas may enable more therapeutic alternatives and guide their treatment decision process.

16.
JCI Insight ; 2(2): e88815, 2017 01 26.
Article in English | MEDLINE | ID: mdl-28138554

ABSTRACT

Clinical trials revealed limited response duration of glioblastomas to VEGF-neutralizing antibody bevacizumab. Thriving in the devascularized microenvironment occurring after antiangiogenic therapy requires tumor cell adaptation to decreased glucose, with 50% less glucose identified in bevacizumab-treated xenografts. Compared with bevacizumab-responsive xenograft cells, resistant cells exhibited increased glucose uptake, glycolysis, 13C NMR pyruvate to lactate conversion, and survival in low glucose. Glucose transporter 3 (GLUT3) was upregulated in bevacizumab-resistant versus sensitive xenografts and patient specimens in a HIF-1α-dependent manner. Resistant versus sensitive cell mitochondria in oxidative phosphorylation-selective conditions produced less ATP. Despite unchanged mitochondrial numbers, normoxic resistant cells had lower mitochondrial membrane potential than sensitive cells, confirming poorer mitochondrial health, but avoided the mitochondrial dysfunction of hypoxic sensitive cells. Thin-layer chromatography revealed increased triglycerides in bevacizumab-resistant versus sensitive xenografts, a change driven by mitochondrial stress. A glycogen synthase kinase-3ß inhibitor suppressing GLUT3 transcription caused greater cell death in bevacizumab-resistant than -responsive cells. Overexpressing GLUT3 in tumor cells recapitulated bevacizumab-resistant cell features: survival and proliferation in low glucose, increased glycolysis, impaired oxidative phosphorylation, and rapid in vivo proliferation only slowed by bevacizumab to that of untreated bevacizumab-responsive tumors. Targeting GLUT3 or the increased glycolysis reliance in resistant tumors could unlock the potential of antiangiogenic treatments.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Bevacizumab/therapeutic use , Drug Resistance, Neoplasm/genetics , Glioblastoma/drug therapy , Glucose Transporter Type 3/genetics , Glycolysis , Angiogenesis Inhibitors/pharmacology , Animals , Bevacizumab/pharmacology , Cell Line, Tumor , Cell Survival , Drug Resistance, Neoplasm/drug effects , Glioblastoma/blood supply , Glioblastoma/genetics , Glioblastoma/metabolism , Glucose/metabolism , Glucose Transporter Type 3/drug effects , Humans , Magnetic Resonance Spectroscopy , Mice , Mice, Nude , Neoplasm Transplantation , Oxidative Phosphorylation , Pyruvic Acid/metabolism , Up-Regulation
17.
Neurosurgery ; 80(3): 421-430, 2017 03 01.
Article in English | MEDLINE | ID: mdl-27218235

ABSTRACT

BACKGROUND: Glioblastoma patients can develop hydrocephalus, either obstructive, typically at diagnosis as a result of mass effect, or communicating, usually later in the disease. OBJECTIVE: To characterize the indications and efficacy of ventriculoperitoneal (VP) shunting for patients with glioblastoma-associated hydrocephalus. METHODS: Retrospective review was conducted of 841 glioblastoma patients diagnosed from 2004 to 2014, 64 (8%) of whom underwent VP shunting for symptomatic hydrocephalus, to analyze symptoms and outcomes after shunting. Overall survival and postshunt survival were analyzed with Kaplan-Meier methods, with predictors evaluated by use of Cox proportional hazards. RESULTS: Of the 64 patients who underwent shunting, 42 (66%) had communicating hydrocephalus (CH) and 22 (34%) had obstructive hydrocephalus (OH). CH patients underwent more preshunt craniotomies than those with noncommunicating hydrocephalus, with a mean of 2.3 and 0.7 surgeries, respectively ( P < .001). Ventricular entry during craniotomy occurred in 52% of CH patients vs 59% of those with OH ( P = .8). After shunting, 61% of all patients achieved symptomatic improvement, which was not associated with hydrocephalus variant ( P > .99). Hydrocephalus symptom improvement rates were as follows: headache, 77%; lethargy, 61%; and altered cognition or memory, 54%. Symptomatic improvement was more likely in patients who were younger at shunt placement (hazard ratio, 0.96; P = .045). Symptomatic improvement, shorter time between glioblastoma diagnosis and shunt placement, and CH rather than OH led to improved postshunt survival (hazard ratio = 0.24-0.99; P = .01-.04). CONCLUSION: VP shunting improves symptoms in most glioblastoma patients with suspected CH or OH, specifically younger patients. Symptomatic improvement, shorter duration between glioblastoma diagnosis and shunt placement, and CH rather than OH improve postshunt survival.


Subject(s)
Brain Neoplasms/surgery , Glioblastoma/surgery , Hydrocephalus/surgery , Ventriculoperitoneal Shunt , Adolescent , Adult , Aged , Aged, 80 and over , Brain Neoplasms/complications , Child , Female , Glioblastoma/complications , Humans , Hydrocephalus/etiology , Male , Middle Aged , Retrospective Studies , Risk Factors , Treatment Outcome , Young Adult
18.
Discov Med ; 22(122): 237-250, 2016 11.
Article in English | MEDLINE | ID: mdl-28009966

ABSTRACT

Metastases to the brain are a common complication of various cancers and are associated with poor prognosis. Management of these patients requires a multidisciplinary approach including whole-brain radiation therapy, stereotactic radiosurgery, surgical resection, chemotherapy, and supportive treatment. Because of recent technological advancements in genomics, our understanding of the genetics of brain metastases is rapidly advancing. This has led to the discovery of many potential genetic therapeutic targets in metastatic brain lesions. One of the limitations to systemic therapies is their limited ability to cross the blood brain barrier, necessitating the urgency to develop clinical trials to evaluate efficacy. A description of these genetic mutations, targeted therapies, and associated clinical trials in brain metastases from lung cancers, breast cancers, and melanoma is outlined in this review.


Subject(s)
Brain Neoplasms/genetics , Brain Neoplasms/radiotherapy , Brain Neoplasms/secondary , Brain Neoplasms/surgery , Animals , Breast Neoplasms/complications , Humans , Lung Neoplasms/complications , Melanoma/complications , Skin Neoplasms/complications
20.
J Stroke Cerebrovasc Dis ; 23(5): 1024-9, 2014.
Article in English | MEDLINE | ID: mdl-24103666

ABSTRACT

BACKGROUND: Stroke can result in varying degrees of respiratory failure. Some patients require tracheostomy in order to facilitate weaning from mechanical ventilation, long-term airway protection, or a combination of the two. Little is known about the rate and predictors of this outcome in patients with severe stroke. We aim to determine the rate of tracheostomy after severe ischemic stroke. METHODS: Using the Nationwide Inpatient Sample database from 2007 to 2009, patients hospitalized with ischemic stroke were identified based on validated International Classification of Diseases, 9th revision, Clinical Modification codes. Next, patients with stroke were stratified based on whether they were treated with or without decompressive craniectomy, and the rate of tracheostomy for each group was determined. A logistic regression analysis was used to identify predictors of tracheostomy after decompressive craniectomy. Survey weights were used to obtain nationally representative estimates. RESULTS: In 1,550,000 patients discharged with ischemic stroke nationwide, the rate of tracheostomy was 1.3% (95% confidence interval [CI], 1.2-1.4%), with a 1.3% (95% CI, 1.1-1.4%) rate in patients without decompressive craniectomy and a 33% (95% CI, 26-39%) rate in the surgical treatment group. Logistic regression analysis identified pneumonia as being significantly associated with tracheostomy after decompressive craniectomy (odds ratio, 3.95; 95% CI, 1.95-6.91). CONCLUSIONS: Tracheostomy is common after decompressive craniectomy and is strongly associated with the development of pneumonia. Given its impact on patient function and potentially modifiable associated factors, tracheostomy may warrant further study as an important patient-centered outcome among patients with stroke.


Subject(s)
Brain Ischemia/surgery , Decompressive Craniectomy , Respiratory Insufficiency/therapy , Stroke/surgery , Tracheostomy/trends , Brain Ischemia/diagnosis , Brain Ischemia/epidemiology , Chi-Square Distribution , Female , Humans , Logistic Models , Male , Middle Aged , Odds Ratio , Pneumonia/epidemiology , Respiratory Insufficiency/diagnosis , Respiratory Insufficiency/epidemiology , Retrospective Studies , Risk Factors , Severity of Illness Index , Stroke/diagnosis , Stroke/epidemiology , Tracheostomy/adverse effects , Treatment Outcome , United States/epidemiology
SELECTION OF CITATIONS
SEARCH DETAIL
...