Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46
Filter
1.
Sci Rep ; 9(1): 14020, 2019 Sep 30.
Article in English | MEDLINE | ID: mdl-31570734

ABSTRACT

Among high-grade brain tumors, glioblastoma is particularly difficult to treat, in part due to its highly infiltrative nature which contributes to the malignant phenotype and high mortality in patients. In order to better understand the signaling pathways underlying glioblastoma invasion, we performed the first large-scale CRISPR-Cas9 loss of function screen specifically designed to identify genes that facilitate cell invasion. We tested 4,574 genes predicted to be involved in trafficking and motility. Using a transwell invasion assay, we discovered 33 genes essential for invasion. Of the 11 genes we selected for secondary testing using a wound healing assay, 6 demonstrated a significant decrease in migration. The strongest regulator of invasion was mitogen-activated protein kinase 4 (MAP4K4). Targeting of MAP4K4 with single guide RNAs or a MAP4K4 inhibitor reduced migration and invasion in vitro. This effect was consistent across three additional patient derived glioblastoma cell lines. Analysis of epithelial-mesenchymal transition markers in U138 cells with lack or inhibition of MAP4K4 demonstrated protein expression consistent with a non-invasive state. Importantly, MAP4K4 inhibition limited migration in a subset of human glioma organotypic slice cultures. Our results identify MAP4K4 as a novel potential therapeutic target to limit glioblastoma invasion.


Subject(s)
Brain Neoplasms/pathology , CRISPR-Associated Protein 9 , CRISPR-Cas Systems , Glioblastoma/pathology , Intracellular Signaling Peptides and Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Brain Neoplasms/genetics , Glioblastoma/genetics , Humans , Neoplasm Invasiveness/genetics
2.
Sci Rep ; 8(1): 18002, 2018 12 20.
Article in English | MEDLINE | ID: mdl-30573757

ABSTRACT

Intratumoral genetic heterogeneity is a widely accepted characteristic of human cancer, including the most common primary malignant brain tumor, glioblastoma. However, the variability in biological behaviors amongst cells within individual tumors is not well described. Invasion into unaffected brain parenchyma is one such behavior, and a leading mechanism of tumor recurrence unaddressed by the current therapeutic armamentarium. Further, providing insight into variability of tumor cell migration within individual tumors may inform discovery of novel anti-invasive therapeutics. In this study, ex vivo organotypic slice cultures from EGFR-wild type and EGFR-amplified patient tumors were treated with the EGFR inhibitor gefitinib to evaluate potential sub-population restricted intratumoral drug-specific responses. High-resolution time-lapse microscopy and quantitative path tracking demonstrated migration of individual cells are punctuated by intermittent bursts of movement. Elevation of population aggregate mean speeds were driven by subpopulations of cells exhibiting frequent high-amplitude bursts, enriched within EGFR-amplified tumors. Treatment with gefitinib specifically targeted high-burst cell subpopulations only in EGFR-amplified tumors, decreasing bursting frequency and amplitude. We provide evidence of intratumoral subpopulations of cells with enhanced migratory behavior in human glioblastoma, selectively targeted via EGFR inhibition. These data justify use of direct human tumor slice cultures to investigate patient-specific therapies designed to limit tumor invasion.


Subject(s)
Brain Neoplasms/pathology , Glioblastoma/pathology , Cell Movement/drug effects , Cell Proliferation/drug effects , Drug Screening Assays, Antitumor , Gefitinib/pharmacology , Humans , Neoplasm Invasiveness , Primary Cell Culture , Tissue Culture Techniques , Tumor Cells, Cultured
3.
J Vis Exp ; (125)2017 07 20.
Article in English | MEDLINE | ID: mdl-28784966

ABSTRACT

Glioblastoma (GBM) continues to carry an extremely poor clinical prognosis despite surgical, chemotherapeutic, and radiation therapy. Progressive tumor invasion into surrounding brain parenchyma represents an enduring therapeutic challenge. To develop anti-migration therapies for GBM, model systems that provide a physiologically relevant background for controlled experimentation are essential. Here, we present a protocol for generating slice cultures from human GBM tissue obtained during surgical resection. These cultures allow for ex vivo experimentation without passaging through animal xenografts or single cell cultures. Further, we describe the use of time-lapse laser scanning confocal microscopy in conjunction with cell tracking to quantitatively study the migratory behavior of tumor cells and associated response to therapeutics. Slices are reproducibly generated within 90 min of surgical tissue acquisition. Retrovirally-mediated fluorescent cell labeling, confocal imaging, and tumor cell migration analyses are subsequently completed within two weeks of culture. We have successfully used these slice cultures to uncover genetic factors associated with increased migratory behavior in human GBM. Further, we have validated the model's ability to detect patient-specific variation in response to anti-migration therapies. Moving forward, human GBM slice cultures are an attractive platform for rapid ex vivo assessment of tumor sensitivity to therapeutic agents, in order to advance personalized neuro-oncologic therapy.


Subject(s)
Antineoplastic Agents/pharmacology , Brain Neoplasms/drug therapy , Cell Culture Techniques/methods , Cell Movement/drug effects , Glioblastoma/drug therapy , Models, Biological , Antineoplastic Agents/therapeutic use , Brain Neoplasms/physiopathology , Female , Glioblastoma/physiopathology , Humans , Male , Microscopy, Confocal/methods
4.
Neurosurgery ; 80(4): 590-601, 2017 04 01.
Article in English | MEDLINE | ID: mdl-27509070

ABSTRACT

BACKGROUND: Extent of resection (EOR) correlates with glioblastoma outcomes. Resectability and EOR depend on anatomical, clinical, and surgeon factors. Resectability likely influences outcome in and of itself, but an accurate measurement of resectability remains elusive. An understanding of resectability and the factors that influence it may provide a means to control a confounder in clinical trials and provide reference for decision making. OBJECTIVE: To provide proof of concept of the use of the collective wisdom of experienced brain tumor surgeons in assessing glioblastoma resectability. METHODS: We surveyed 13 academic tumor neurosurgeons nationwide to assess the resectability of newly diagnosed glioblastoma. Participants reviewed 20 cases, including digital imaging and communications in medicine-formatted pre- and postoperative magnetic resonance images and clinical vignettes. The selected cases involved a variety of anatomical locations and a range of EOR. Participants were asked about surgical goal, eg, gross total resection, subtotal resection (STR), or biopsy, and rationale for their decision. We calculated a "resectability index" for each lesion by pooling responses from all 13 surgeons. RESULTS: Neurosurgeons' individual surgical goals varied significantly ( P = .015), but the resectability index calculated from the surgeons' pooled responses was strongly correlated with the percentage of contrast-enhancing residual tumor ( R = 0.817, P < .001). The collective STR goal predicted intraoperative decision of intentional STR documented on operative notes ( P < .01) and nonresectable residual ( P < .01), but not resectable residual. CONCLUSION: In this pilot study, we demonstrate the feasibility of measuring the resectability of glioblastoma through crowdsourcing. This tool could be used to quantify resectability, a potential confounder in neuro-oncology clinical trials.


Subject(s)
Brain Neoplasms/surgery , Glioblastoma/surgery , Neoplasm, Residual/surgery , Adolescent , Adult , Aged , Brain Neoplasms/diagnostic imaging , Brain Neoplasms/pathology , Female , Glioblastoma/diagnostic imaging , Glioblastoma/pathology , Humans , Magnetic Resonance Imaging , Male , Middle Aged , Neoplasm, Residual/diagnostic imaging , Neoplasm, Residual/pathology , Neurosurgical Procedures/methods , Pilot Projects
5.
Anal Chem ; 88(5): 2770-6, 2016 Mar 01.
Article in English | MEDLINE | ID: mdl-26833093

ABSTRACT

Cell migration through three-dimensional (3D) tissue spaces is integral to many biological and pathological processes, including metastasis. Circulating tumor cells (CTCs) are phenotypically heterogeneous, and in vitro analysis of their extravasation behavior is often impeded by the inability to establish complex tissue-like extracellular matrix (ECM) environments and chemotactic gradients within microfluidic devices. We have developed a novel microfluidic strategy to manipulate surface properties of enclosed microchannels and create 3D ECM structures for real-time observation of individual migrating cells. The wettability of selective interconnected channels is controlled by a plasma pulse, enabling the incorporation of ECM exclusively within the transmigration regions. We applied this approach to collectively analyze CTC-endothelial adhesion, trans-endothelial migration, and subsequent motility of breast cancer cells (MDA-MB-231) through a 3D ECM under artificial gradients of SDF-1α. We observed migration velocities ranging from 5.12 to 12.8 µm/h, which closely correspond to single-cell migration in collagen blocks, but are significantly faster than the migration of cell aggregates. The compartmentalized microchannels separated by the porous ECM makes this in vitro assay versatile and suitable for a variety of applications such as inflammation studies, drug screening, and coculture interactions.


Subject(s)
Cell Movement , Extracellular Matrix/metabolism , Cell Line, Tumor , Human Umbilical Vein Endothelial Cells , Humans , Hydrogels , Microfluidics/instrumentation , Surface Properties
6.
J Neurooncol ; 123(3): 405-12, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26123363

ABSTRACT

Cell-mediated suppression of anti-tumor immunity is multifactorial in patients with cancer, and recent studies have focused on several distinct cellular agents that are associated with this phenomenon. This review will focus on the potential role of regulatory T cells (Tregs) and microglia in the suppression of cellular immunity observed in patients with glioblastoma. We discuss the ontogeny, basic biology, evidence for activity, and potential clinical options for targeting Tregs and microglia as part of immunotherapy in affected patients.


Subject(s)
Brain Neoplasms , Glioblastoma , Immunosuppression Therapy , Microglia/physiology , T-Lymphocytes, Regulatory/physiology , Brain Neoplasms/immunology , Brain Neoplasms/pathology , Brain Neoplasms/therapy , Glioblastoma/immunology , Glioblastoma/pathology , Glioblastoma/therapy , Humans
7.
J Cereb Blood Flow Metab ; 35(10): 1657-63, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25966956

ABSTRACT

Transient suppression of peripheral immunity is a major source of complication for patients suffering from ischemic stroke. The release of Arginase I (ArgI) from activated neutrophils has recently been associated with T-cell dysfunction in a number of pathologies. However, this pathway has not been previously explored in ischemic stroke. Using the murine model of transient middle cerebral artery occlusion, we explored effects of stroke on peripheral T-cell function and evaluated the role of neutrophils and ArgI. Stimulation of splenic T cells from post-stroke animals with anti-CD3/CD28 resulted in decreased proliferation and interferon-γ production when compared with sham-surgery controls. Flow cytometric analysis of intrasplenic leukocytes exposed the presence of a transient population of activated neutrophils that correlated quantitatively with elevated ArgI levels in culture media. In vitro activation of purified resting neutrophils from unmanipulated controls confirmed the capacity for murine neutrophils to release ArgI from preformed granules. We observed decreased expression of the L-arg-sensitive CD3ζ on T cells, consistent with decreased functional activity. Critically, L-arg supplementation restored the functional response of post-stroke T cells to mitogenic stimulation. Together, these data outline a novel mechanism of reversible, neutrophil-mediated peripheral immunosuppression related to ArgI release following ischemic stroke.


Subject(s)
Arginase/metabolism , Immune Tolerance , Neutrophil Activation , Neutrophils/enzymology , Stroke/enzymology , Animals , Arginase/blood , Arginine/pharmacology , Infarction, Middle Cerebral Artery/parasitology , Male , Mice , Mice, Inbred C57BL , Spleen/cytology , Spleen/pathology , Stroke/pathology , T-Lymphocytes
8.
J Neurooncol ; 122(1): 135-43, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25524817

ABSTRACT

Bevacizumab blocks the effects of VEGF and may allow for more aggressive radiotherapy schedules. We evaluated the efficacy and toxicity of hypofractionated intensity-modulated radiation therapy with concurrent and adjuvant temozolomide and bevacizumab in patients with newly diagnosed glioblastoma. Patients with newly diagnosed glioblastoma were treated with hypofractionated intensity modulated radiation therapy to the surgical cavity and residual tumor with a 1 cm margin (PTV1) to 60 Gy and to the T2 abnormality with a 1 cm margin (PTV2) to 30 Gy in 10 daily fractions over 2 weeks. Concurrent temozolomide (75 mg/m(2) daily) and bevacizumab (10 mg/kg) was administered followed by adjuvant temozolomide (200 mg/m(2)) on a standard 5/28 day cycle and bevacizumab (10 mg/kg) every 2 weeks for 6 months. Thirty newly diagnosed patients were treated on study. Median PTV1 volume was 131.1 cm(3) and the median PTV2 volume was 342.6 cm(3). Six-month progression-free survival (PFS) was 90 %, with median follow-up of 15.9 months. The median PFS was 14.3 months, with a median overall survival (OS) of 16.3 months. Grade 4 hematologic toxicity included neutropenia (10 %) and thrombocytopenia (17 %). Grades 3/4 non-hematologic toxicity included fatigue (13 %), wound dehiscence (7 %) and stroke, pulmonary embolism and nausea each in 1 patient. Presumed radiation necrosis with clinical decline was seen in 50 % of patients, two with autopsy documentation. The study was closed early to accrual due to this finding. This study demonstrated 90 % 6-month PFS and OS comparable to historic data in patients receiving standard treatment. Bevacizumab did not prevent radiation necrosis associated with this hypofractionated radiation regimen and large PTV volumes may have contributed to high rates of presumed radiation necrosis.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Brain Neoplasms/therapy , Chemoradiotherapy , Dose Fractionation, Radiation , Glioblastoma/therapy , Radiotherapy, Intensity-Modulated/methods , Adult , Aged , Bevacizumab/administration & dosage , Brain Neoplasms/diagnosis , Brain Neoplasms/mortality , Chemotherapy, Adjuvant , Dacarbazine/administration & dosage , Dacarbazine/analogs & derivatives , Female , Follow-Up Studies , Glioblastoma/diagnosis , Glioblastoma/mortality , Humans , Male , Middle Aged , Neoplasm Staging , Prognosis , Prospective Studies , Survival Rate , Temozolomide
9.
J Neurooncol ; 119(3): 577-81, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25074368

ABSTRACT

Over the 30 years since the formation of the AANS/CNS Section on Tumors, the breadth and scope of our activities have continued to expand. An initial focus on education and collaboration between those members of the neurosurgical community dedicated to the care of patients with neurological tumors has broadened to include development of international relationships, participation in clinical trials and efforts to define standards of care and quality metrics. As we navigate the rapidly changing environment of health care, the Section on Tumors will occupy a central role in promoting advocacy, establishing collaboration, providing education, and supporting research for the community of neurosurgeons dedicated to the care of patients with neurological tumors. This article will provide an update and status report on the development of the Section on Tumors, followed by a brief discussion of the challenges and opportunities emerging at the onset of our fourth decade of service.


Subject(s)
Neoplasms/therapy , Neurosurgery , Societies, Medical , Biomedical Research/economics , Biomedical Research/education , Biomedical Research/methods , Humans , Neurosurgery/economics , Neurosurgery/education , Patient Advocacy , Societies, Medical/economics , United States
10.
Am J Otolaryngol ; 35(3): 424-6, 2014.
Article in English | MEDLINE | ID: mdl-24499924

ABSTRACT

Optimal vascular control during neurosurgical resection of large sub-frontal meningioma is hindered by limited early access to the ethmoidal arteries. Pre-operative ligation of the ethmoidal arteries 1) induces tumor necrosis simplifying resection and 2) minimizes blood loss and operative time. Early arterial ligation is an advantage of endoscopic approaches to transnasal resection of anterior skull base meningiomas that is not appreciated in open approaches with larger meningioma. Here we present a case of a colossal meningioma where minimally invasive pre-operative ligation of ethmoidal arteries prior to a traditional open surgical approach allowed for improved vascular control and decreased surgical time.


Subject(s)
Meningeal Neoplasms/surgery , Meningioma/surgery , Preoperative Care , Arteries/surgery , Female , Humans , Ligation , Middle Aged , Nose/blood supply
11.
J Immunol ; 191(9): 4880-8, 2013 Nov 01.
Article in English | MEDLINE | ID: mdl-24078694

ABSTRACT

Despite increasing evidence that antitumor immune control exists in the pediatric brain, these findings have yet to be exploited successfully in the clinic. A barrier to development of immunotherapeutic strategies in pediatric brain tumors is that the immunophenotype of these tumors' microenvironment has not been defined. To address this, the current study used multicolor FACS of disaggregated tumor to systematically characterize the frequency and phenotype of infiltrating immune cells in the most common pediatric brain tumor types. The initial study cohort consisted of 7 pilocytic astrocytoma (PA), 19 ependymoma (EPN), 5 glioblastoma (GBM), 6 medulloblastoma (MED), and 5 nontumor brain (NT) control samples obtained from epilepsy surgery. Immune cell types analyzed included both myeloid and T cell lineages and respective markers of activated or suppressed functional phenotypes. Immune parameters that distinguished each of the tumor types were identified. PA and EPN demonstrated significantly higher infiltrating myeloid and lymphoid cells compared with GBM, MED, or NT. Additionally, PA and EPN conveyed a comparatively activated/classically activated myeloid cell-skewed functional phenotype denoted in particular by HLA-DR and CD64 expression. In contrast, GBM and MED contained progressively fewer infiltrating leukocytes and more muted functional phenotypes similar to that of NT. These findings were recapitulated using whole tumor expression of corresponding immune marker genes in a large gene expression microarray cohort of pediatric brain tumors. The results of this cross-tumor comparative analysis demonstrate that different pediatric brain tumor types exhibit distinct immunophenotypes, implying that specific immunotherapeutic approaches may be most effective for each tumor type.


Subject(s)
Brain Neoplasms/classification , Brain Neoplasms/immunology , Immunophenotyping , Myeloid Cells/immunology , T-Lymphocytes/immunology , Adolescent , Astrocytoma/immunology , Brain/immunology , Brain Neoplasms/genetics , Child , Cohort Studies , Ependymoma/immunology , Epilepsy/immunology , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Glioblastoma/immunology , HLA-DR Antigens/metabolism , Humans , Medulloblastoma/immunology , Receptors, IgG/metabolism , Tumor Microenvironment
12.
J Neurooncol ; 114(1): 111-6, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23729276

ABSTRACT

To report health-related quality of life (HRQOL) in glioblastoma (GBM) patients treated on a phase II trial of hypofractionated intensity-modulated radiotherapy (hypo-IMRT) with temozolomide (TMZ). GBM patients received postoperative hypo-IMRT to 60 Gy in 10 fractions with TMZ. HRQOL was assessed using the EORTC quality of life questionnaire core-30 and the EORTC brain cancer module, performed at baseline, RT completion, 1 mo post-RT, and every 3 mos thereafter. Changes from baseline were calculated for each specific HRQOL scale. A ≥ 10 point change in any HRQOL scale from the mean baseline score was significant. 24 patients were treated. Compliance with HRQOL assessments at baseline, RT completion, and 1, 3, 6, 9, and 12 mos post-RT was 100, 96, 92, 79, 70, 68 and 53 %, respectively. Up to 12 mos post-RT, no significant changes were seen in global health status, physical functioning, role functioning, emotional functioning, fatigue, nausea, vision, headache or seizure. Significant improvement was seen in insomnia, future uncertainty, motor dysfunction and drowsiness. Significant worsening was observed in cognitive functioning, social functioning, appetite loss and communication deficit. 60 Gy hypo-IMRT in 6-Gy fractions with TMZ does not appear to negatively impact overall HRQOL.


Subject(s)
Antineoplastic Agents, Alkylating/therapeutic use , Brain Neoplasms/therapy , Dacarbazine/analogs & derivatives , Glioblastoma/therapy , Quality of Life , Radiotherapy, Intensity-Modulated , Adult , Aged , Brain Neoplasms/psychology , Cognition/drug effects , Cognition/physiology , Cognition/radiation effects , Combined Modality Therapy , Dacarbazine/therapeutic use , Female , Glioblastoma/psychology , Humans , Karnofsky Performance Status , Male , Middle Aged , Neuropsychological Tests , Prospective Studies , Temozolomide
13.
Neuro Oncol ; 15(8): 1048-57, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23749785

ABSTRACT

BACKGROUND: Tissue invasion is a hallmark of most human cancers and remains a major source of treatment failure in patients with glioblastoma (GBM). Although EGFR amplification has been previously associated with more invasive tumor behavior, existing experimental models have not supported quantitative evaluation of interpatient differences in tumor cell migration or testing of patient-specific responses to therapies targeting invasion. To explore these questions, we optimized an ex vivo organotypic slice culture system allowing for labeling and tracking of tumor cells in human GBM slice cultures. METHODS: With use of time-lapse confocal microscopy of retrovirally labeled tumor cells in slices, baseline differences in migration speed and efficiency were determined and correlated with EGFR amplification in a cohort of patients with GBM. Slices were treated with gefitinib to evaluate anti-invasive effects associated with targeting EGFR. RESULTS: Migration analysis identified significant patient-to-patient variation at baseline. EGFR amplification was correlated with increased migration speed and efficiency compared with nonamplified tumors. Critically, gefitinib resulted in a selective and significant reduction of tumor cell migration in EGFR-amplified tumors. CONCLUSIONS: These data provide the first identification of patient-to-patient variation in tumor cell migration in living human tumor tissue. We found that EGFR-amplified GBM are inherently more efficient in their migration and can be effectively targeted by gefitinib treatment. These data suggest that stratified clinical trails are needed to evaluate gefitinib as an anti-invasive adjuvant for patients with EGFR-amplified GBM. In addition, these results provide proof of principle that primary slice cultures may be useful for patient-specific screening of agents designed to inhibit tumor invasion.


Subject(s)
Cell Movement/drug effects , ErbB Receptors/genetics , Gene Amplification , Glioblastoma/drug therapy , Protein Kinase Inhibitors/pharmacology , Quinazolines/pharmacology , Cell Proliferation/drug effects , Cohort Studies , ErbB Receptors/antagonists & inhibitors , Gefitinib , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , In Situ Hybridization, Fluorescence , Organ Culture Techniques , Tumor Cells, Cultured
14.
J Craniofac Surg ; 24(3): 953-6, 2013 May.
Article in English | MEDLINE | ID: mdl-23714919

ABSTRACT

Pneumocephalus is an exceedingly rare complication associated with neurological deficit in cases of frontoethmoid osteoma. The overarching management strategy for affected patients remains undefined. We describe the case of a 61-year-old female patient presenting with frontoethmoid osteoma manifesting as profound intraparenchymal pneumocephalus and associated neurological deficit, treated through a minimally invasive combined surgical strategy involving image-guided burr hole decompression of the pneumocephalus followed by transnasal endoscopic removal of the tumor. Using this approach, the patient rapidly recovered full neurologic function. We review the existing literature and, given the likely intraparenchymal location of pneumocephalus associated with these lesions with the potential of rapid clinical deterioration, recommend aggressive surgical management. Although these lesions can be removed from a purely endoscopic approach, we recommend burr-hole decompression of the pneumocephalus as an adjunct to ensure prompt resolution of the neurologic symptoms.


Subject(s)
Bone Neoplasms/surgery , Minimally Invasive Surgical Procedures/methods , Osteoma/surgery , Paranasal Sinus Neoplasms/surgery , Pneumocephalus/etiology , Bone Neoplasms/complications , Ethmoid Sinus/surgery , Female , Frontal Sinus/surgery , Humans , Middle Aged , Osteoma/complications , Paranasal Sinus Neoplasms/complications , Tomography, X-Ray Computed , Treatment Outcome
15.
Immunol Res ; 55(1-3): 22-33, 2013 Mar.
Article in English | MEDLINE | ID: mdl-22941561

ABSTRACT

There will be over half a million cancer-related deaths in the United States in 2012, with lung cancer being the leader followed by prostate in men and breast in women. There is estimated to be more than one and a half million new cases of cancer in 2012, making the development of effective therapies a high priority. As tumor immunologists, we are interested in the development of immunotherapies because the immune response offers exquisite specificity and the potential to target tumor cells without harming normal cells. In this review, we highlight the current advances in the field of immunotherapy and the current work being completed by laboratories at University of Colorado School of Medicine in multiple malignancies, including breast cancer, lung cancer, melanoma, thyroid cancer, and glioblastoma. This work focuses on augmenting the anti-tumor response of CD8 T cells in the blood, lymph nodes, and tumors of patients, determining biomarkers for patients who are more likely to respond to immunotherapy, and identifying additional anti-tumor and immunosuppressive cells that influence the overall response to tumors. These collaborative efforts will identify mechanisms to improve immune function, which may elucidate therapeutic targets for clinical trials to improve patient health and survival.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Neoplasms/immunology , Colorado , Humans , Immunotherapy , Neoplasms/therapy , Universities
16.
Adv Otorhinolaryngol ; 74: 138-47, 2013.
Article in English | MEDLINE | ID: mdl-23257560

ABSTRACT

Postoperative care following skull base surgery and reconstruction is critical for achieving optimal outcomes and patient satisfaction. The choice of reconstructive technique and grafting materials may significantly influence the postoperative planning and monitoring for each patient. The overarching goals of postoperative management include avoidance and early recognition of complications, facilitation of wound healing at the surgical site(s), and quick recovery for the patient. Planning for postoperative care is initiated in the preoperative and intraoperative settings. Throughout the reconstruction, the surgical team must actively and meticulously evaluate the potential effects of each maneuver in regard to the subsequent clinical course. During the immediate postoperative period, it is helpful to have clinical protocols in place that address potential surgical complications, allow for early detection of neurological or hormonal dysfunction, and maximize patient safety and comfort. Longer-term postoperative follow-up continues until symptoms resolve and the endoscopic examination is normal. During this time period, well-defined goals and issues of concern should be clearly provided to the patient. In addition, disease-dependent schedules of surveillance are critical for patients with potentially recurrent pathology. This chapter will address decision-making for such topics during postoperative care following skull base reconstruction.


Subject(s)
Endoscopy/methods , Plastic Surgery Procedures/methods , Postoperative Care , Skull Base/surgery , Surgical Flaps , Humans , Wound Healing
17.
Brain Pathol ; 22(6): 861-4, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23050872

ABSTRACT

We report a case of balloon cell melanoma metastatic to the cerebellum; the clear cell morphology prompted initial differential diagnostic considerations of metastatic renal cell carcinoma and hemangioblastoma in this site. To our knowledge this is only the second case of metastatic balloon cell melanoma to the CNS.


Subject(s)
Brain Neoplasms/secondary , Melanoma/secondary , Skin Neoplasms/pathology , Aged , Humans , Immunohistochemistry , Magnetic Resonance Imaging , Male
18.
Neurosurg Clin N Am ; 23(3): 407-16, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22748653

ABSTRACT

High-grade gliomas are rapidly progressing and generally fatal neoplasms of the brain. Chemotherapy has continued to provide only limited benefit for patients harboring these tumors. The recurrence of common mutations, combined with the similarities of many of the acquired capabilities and characteristics of solid tumors, suggest many common therapeutic targets. During the past few decades, an increased understanding of many of the cellular regulatory mechanisms associated with carcinogenesis has provided an opportunity for the development of pathway-specific small molecule targeted inhibitors (SMIs). This article reviews the use of SMIs in the treatment of high-grade glioma.


Subject(s)
Antineoplastic Agents/therapeutic use , Brain Neoplasms/drug therapy , ErbB Receptors/antagonists & inhibitors , Glioblastoma/drug therapy , Receptors, Platelet-Derived Growth Factor/antagonists & inhibitors , Receptors, Vascular Endothelial Growth Factor/antagonists & inhibitors , Clinical Trials as Topic , Humans , Phosphoinositide-3 Kinase Inhibitors , Signal Transduction , ras Proteins/antagonists & inhibitors
19.
Neurosurg Clin N Am ; 23(3): 429-37, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22748655

ABSTRACT

High-grade glioma continues to impart poor prognosis in spite of maximal treatment. Attempted gross total surgical resection followed by concurrent temozolomide and radiation therapy has become standard of care for glioblastoma. Ongoing clinical efforts have been directed at the further development of radiosensitizing agents that exploit tumor biology to maximize effects of concurrently administered radiation. The current article outlines the scientific rationale for the use of radiosensitizing agents and preliminary results from clinical trials using a variety of these approaches.


Subject(s)
Antineoplastic Agents, Alkylating/therapeutic use , Brain Neoplasms/therapy , Dacarbazine/analogs & derivatives , Glioblastoma/therapy , Radiation-Sensitizing Agents/therapeutic use , Angiogenesis Inhibitors/therapeutic use , Chemotherapy, Adjuvant , Combined Modality Therapy , Dacarbazine/therapeutic use , Humans , Radiotherapy/methods , Temozolomide
20.
J Immunol ; 189(4): 1920-7, 2012 Aug 15.
Article in English | MEDLINE | ID: mdl-22802421

ABSTRACT

Survival in the majority of high-grade astrocytoma (HGA) patients is very poor, with only a rare population of long-term survivors. A better understanding of the biological factors associated with long-term survival in HGA would aid development of more effective therapy and survival prediction. Factors associated with long-term survival have not been extensively studied using unbiased genome-wide expression analyses. In the current study, gene expression microarray profiles of HGA from long-term survivors were interrogated for discovery of survival-associated biological factors. Ontology analyses revealed that increased expression of immune function-related genes was the predominant biological factor that positively correlated with longer survival. A notable T cell signature was present within this prognostic immune gene set. Using immune cell-specific gene classifiers, both T cell-associated and myeloid linage-associated genes were shown to be enriched in HGA from long-term versus short-term survivors. Association of immune function and cell-specific genes with survival was confirmed independently in a larger publicly available glioblastoma gene expression microarray data set. Histology was used to validate the results of microarray analyses in a larger cohort of long-term survivors of HGA. Multivariate analyses demonstrated that increased immune cell infiltration was a significant independent variable contributing to longer survival, as was Karnofsky/Lansky performance score. These data provide evidence of a prognostic anti-tumor adaptive immune response and rationale for future development of immunotherapy in HGA.


Subject(s)
Astrocytoma/genetics , Astrocytoma/immunology , Brain Neoplasms/genetics , Brain Neoplasms/immunology , Survivors , Astrocytoma/mortality , Brain Neoplasms/mortality , Gene Expression Profiling , Humans , Immunohistochemistry , Kaplan-Meier Estimate , Karnofsky Performance Status , Lymphocytes, Tumor-Infiltrating/immunology , Oligonucleotide Array Sequence Analysis , Prognosis , Proportional Hazards Models
SELECTION OF CITATIONS
SEARCH DETAIL
...