Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
Neurooncol Pract ; 7(1): 118-126, 2020 Jan.
Article in English | MEDLINE | ID: mdl-32257290

ABSTRACT

BACKGROUND: Leptomeningeal dissemination (LD) in adults is an exceedingly rare complication of low-grade neuroepithelial CNS tumors (LGNs). We aimed to determine relative incidence, clinical presentation, and predictors of outcome. METHODS: We searched the quality control database of the Section of Neuro-Oncology, Yale Cancer Center, for patients with LGN (WHO grade I/II) seen between 2002 and 2017. For cases complicated by LD, we recorded demographics, clinical signs, histopathological diagnosis, and imaging findings. A comprehensive literature review was performed. RESULTS: Eleven consecutive patients with LD were identified, representing 2.3% of individuals with LGN seen at our institution between 2002 and 2017 (n = 475). Ependymoma was the predominant histological entity. Mean time interval from diagnosis of LGN to LD was 38.6 ± 10 months. Symptoms were mostly attributed to communicating hydrocephalus. Tumor deposits of LD were either nodular or linear with variable enhancement (nonenhancing lesions in 4 of 11 patients). Localized (surgery, radiosurgery, involved-field, or craniospinal radiation therapy) or systemic treatments (chemotherapy) were provided. All patients progressed radiographically. Median overall survival after LD was 102 months. Survival was prolonged when a combination of localized and systemic therapies was administered (188.5 vs 25.5 months; P = .03). Demographics and tumor spectrum reported in the literature were similar to our cohort. CONCLUSIONS: LD is a rare complication of LGNs. A high level of suspicion is required for timely diagnosis as early symptoms are nonspecific and commonly do not occur until years after initial tumor diagnosis. Repeated aggressive treatment appears to be beneficial in improving survival.

2.
Nat Genet ; 48(10): 1253-9, 2016 10.
Article in English | MEDLINE | ID: mdl-27548314

ABSTRACT

RNA polymerase II mediates the transcription of all protein-coding genes in eukaryotic cells, a process that is fundamental to life. Genomic mutations altering this enzyme have not previously been linked to any pathology in humans, which is a testament to its indispensable role in cell biology. On the basis of a combination of next-generation genomic analyses of 775 meningiomas, we report that recurrent somatic p.Gln403Lys or p.Leu438_His439del mutations in POLR2A, which encodes the catalytic subunit of RNA polymerase II (ref. 1), hijack this essential enzyme and drive neoplasia. POLR2A mutant tumors show dysregulation of key meningeal identity genes, including WNT6 and ZIC1/ZIC4. In addition to mutations in POLR2A, NF2, SMARCB1, TRAF7, KLF4, AKT1, PIK3CA, and SMO, we also report somatic mutations in AKT3, PIK3R1, PRKAR1A, and SUFU in meningiomas. Our results identify a role for essential transcriptional machinery in driving tumorigenesis and define mutually exclusive meningioma subgroups with distinct clinical and pathological features.


Subject(s)
Meningeal Neoplasms/genetics , Meningioma/genetics , Mutation , RNA Polymerase II/genetics , Catalytic Domain/genetics , Chromosomes, Human, Pair 22 , Cohort Studies , DNA Mutational Analysis , Enhancer Elements, Genetic , Exome , Gene Expression Regulation, Neoplastic , Genotype , Humans , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/genetics , Meningeal Neoplasms/classification , Meningioma/classification , Neurofibromin 2/genetics , Tumor Necrosis Factor Receptor-Associated Peptides and Proteins/genetics
3.
ACS Nano ; 10(4): 4209-18, 2016 04 26.
Article in English | MEDLINE | ID: mdl-26967254

ABSTRACT

The blood-brain barrier (BBB) is partially disrupted in brain tumors. Despite the gaps in the BBB, there is an inadequate amount of pharmacological agents delivered into the brain. Thus, the low delivery efficiency renders many of these agents ineffective in treating brain cancer. In this report, we proposed an "autocatalytic" approach for increasing the transport of nanoparticles into the brain. In this strategy, a small number of nanoparticles enter into the brain via transcytosis or through the BBB gaps. After penetrating the BBB, the nanoparticles release BBB modulators, which enables more nanoparticles to be transported, creating a positive feedback loop for increased delivery. Specifically, we demonstrated that these autocatalytic brain tumor-targeting poly(amine-co-ester) terpolymer nanoparticles (ABTT NPs) can readily cross the BBB and preferentially accumulate in brain tumors at a concentration of 4.3- and 94.0-fold greater than that in the liver and in brain regions without tumors, respectively. We further demonstrated that ABTT NPs were capable of mediating brain cancer gene therapy and chemotherapy. Our results suggest ABTT NPs can prime the brain to increase the systemic delivery of therapeutics for treating brain malignancies.


Subject(s)
Blood-Brain Barrier/metabolism , Brain Neoplasms/diagnostic imaging , Brain Neoplasms/therapy , Nanoparticles/chemistry , Animals , Antineoplastic Agents/administration & dosage , Biological Transport , Cell Line, Tumor , Decanoic Acids/chemistry , Drug Delivery Systems , Ethanolamines/chemistry , Female , Genetic Therapy , Heterografts , Humans , Matrix Metalloproteinase 2/chemistry , Mice , Mice, Inbred C57BL , Optical Imaging , Paclitaxel/administration & dosage , Permeability , Polymers/chemistry , Purines/chemistry , Pyrazoles/chemistry , Scorpion Venoms/chemistry , Transcytosis , Tumor Microenvironment
4.
J Neurooncol ; 121(3): 441-9, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25403507

ABSTRACT

Current therapy for glioblastoma multiforme (GBM) is largely ineffective, with nearly universal tumor recurrence. The failure of current therapy is primarily due to the lack of approaches for the efficient delivery of therapeutics to diffuse tumors in the brain. In our prior study, we developed brain-penetrating nanoparticles that are capable of penetrating brain tissue and distribute over clinically relevant volumes when administered via convection-enhanced delivery (CED). We demonstrated that these particles are capable of efficient delivery of chemotherapeutics to diffuse tumors in the brain, indicating that they may serve as a groundbreaking approach for the treatment of GBM. In the original study, nanoparticles in the brain were imaged using positron emission tomography (PET). However, clinical translation of this delivery platform can be enabled by engineering a non-invasive detection modality using magnetic resonance imaging (MRI). For this purpose, we developed chemistry to incorporate superparamagnetic iron oxide (SPIO) into the brain-penetrating nanoparticles. We demonstrated that SPIO-loaded nanoparticles, which retain the same morphology as nanoparticles without SPIO, have an excellent transverse (T(2)) relaxivity. After CED, the distribution of nanoparticles in the brain (i.e., in the vicinity of injection site) can be detected using MRI and the long-lasting signal attenuation of SPIO-loaded brain-penetrating nanoparticles lasted over a one-month timecourse. Development of these nanoparticles is significant as, in future clinical applications, co-administration of SPIO-loaded nanoparticles will allow for intraoperative monitoring of particle distribution in the brain to ensure drug-loaded nanoparticles reach tumors as well as for monitoring the therapeutic benefit with time and to evaluate tumor relapse patterns.


Subject(s)
Antineoplastic Agents/administration & dosage , Drug Delivery Systems/methods , Lactic Acid/administration & dosage , Magnetic Resonance Imaging/methods , Nanoparticles/administration & dosage , Neuroimaging/methods , Polyglycolic Acid/administration & dosage , Animals , Brain Neoplasms/drug therapy , Convection , Ferric Compounds , Glioblastoma/drug therapy , Humans , Image Processing, Computer-Assisted , Nanoparticles/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer , Rats , Rats, Sprague-Dawley
5.
Proc Natl Acad Sci U S A ; 110(29): 11751-6, 2013 Jul 16.
Article in English | MEDLINE | ID: mdl-23818631

ABSTRACT

Current therapy for glioblastoma multiforme is insufficient, with nearly universal recurrence. Available drug therapies are unsuccessful because they fail to penetrate through the region of the brain containing tumor cells and they fail to kill the cells most responsible for tumor development and therapy resistance, brain cancer stem cells (BCSCs). To address these challenges, we combined two major advances in technology: (i) brain-penetrating polymeric nanoparticles that can be loaded with drugs and are optimized for intracranial convection-enhanced delivery and (ii) repurposed compounds, previously used in Food and Drug Administration-approved products, which were identified through library screening to target BCSCs. Using fluorescence imaging and positron emission tomography, we demonstrate that brain-penetrating nanoparticles can be delivered to large intracranial volumes in both rats and pigs. We identified several agents (from Food and Drug Administration-approved products) that potently inhibit proliferation and self-renewal of BCSCs. When loaded into brain-penetrating nanoparticles and administered by convection-enhanced delivery, one of these agents, dithiazanine iodide, significantly increased survival in rats bearing BCSC-derived xenografts. This unique approach to controlled delivery in the brain should have a significant impact on treatment of glioblastoma multiforme and suggests previously undescribed routes for drug and gene delivery to treat other diseases of the central nervous system.


Subject(s)
Antineoplastic Agents/therapeutic use , Blood-Brain Barrier/metabolism , Dithiazanine/therapeutic use , Drug Delivery Systems/methods , Glioblastoma/drug therapy , Nanoparticles/administration & dosage , Animals , Antineoplastic Agents/administration & dosage , Dithiazanine/administration & dosage , Fluorescence , Kaplan-Meier Estimate , Positron-Emission Tomography , Rats , Swine
6.
Science ; 339(6123): 1077-80, 2013 Mar 01.
Article in English | MEDLINE | ID: mdl-23348505

ABSTRACT

We report genomic analysis of 300 meningiomas, the most common primary brain tumors, leading to the discovery of mutations in TRAF7, a proapoptotic E3 ubiquitin ligase, in nearly one-fourth of all meningiomas. Mutations in TRAF7 commonly occurred with a recurrent mutation (K409Q) in KLF4, a transcription factor known for its role in inducing pluripotency, or with AKT1(E17K), a mutation known to activate the PI3K pathway. SMO mutations, which activate Hedgehog signaling, were identified in ~5% of non-NF2 mutant meningiomas. These non-NF2 meningiomas were clinically distinctive-nearly always benign, with chromosomal stability, and originating from the medial skull base. In contrast, meningiomas with mutant NF2 and/or chromosome 22 loss were more likely to be atypical, showing genomic instability, and localizing to the cerebral and cerebellar hemispheres. Collectively, these findings identify distinct meningioma subtypes, suggesting avenues for targeted therapeutics.


Subject(s)
Brain Neoplasms/genetics , Kruppel-Like Transcription Factors/genetics , Meningeal Neoplasms/genetics , Meningioma/genetics , Proto-Oncogene Proteins c-akt/genetics , Receptors, G-Protein-Coupled/genetics , Tumor Necrosis Factor Receptor-Associated Peptides and Proteins/genetics , Adult , Aged , Aged, 80 and over , Brain Neoplasms/classification , Brain Neoplasms/pathology , Chromosomes, Human, Pair 22/genetics , DNA Mutational Analysis , Female , Genes, Neurofibromatosis 2 , Genomic Instability , Genomics , Humans , Kruppel-Like Factor 4 , Male , Meningeal Neoplasms/classification , Meningeal Neoplasms/pathology , Meningioma/classification , Meningioma/pathology , Middle Aged , Mutation , Neoplasm Grading , Smoothened Receptor
7.
Hematol Oncol Clin North Am ; 26(4): 757-77, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22794282

ABSTRACT

In the United States, approximately 65,000 people are diagnosed with primary brain tumors each year, with an incidence of 19.3 cases per 100,000 person-years. These numbers represent a wide spectrum of disease, from benign to malignant, and prognosis varies widely based on disease. Treatment of primary brain tumors most often uses a combination of surgery and radiation. However, over the past several generations, technological advancements have significantly altered the treatment paradigm. This article reviews the current role of neurosurgery and radiation therapy in the management of primary brain tumors.


Subject(s)
Brain Neoplasms/therapy , Brain/pathology , Brain Neoplasms/radiotherapy , Brain Neoplasms/surgery , Diagnostic Imaging , Humans
8.
Hematol Oncol Clin North Am ; 26(4): 797-809, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22794284

ABSTRACT

Low-grade gliomas are uncommon tumors whose optimal management remains to be determined. Although well-designed clinical trials have been mounted to address certain aspects of postoperative radiotherapeutic management, additional studies are required to refine management based on tumor-specific and patient-specific variables. There is mounting evidence that the relative completeness of surgical resection can improve survival, and the molecular and histopathologic characterization of the glioma requires adequate samples for analysis. Current imaging and operative techniques can direct surgical resection, and the same imaging techniques can help monitor patients postoperatively and predict prognosis.


Subject(s)
Brain Neoplasms/diagnosis , Brain Neoplasms/therapy , Glioma/diagnosis , Glioma/therapy , Brain/pathology , Brain Neoplasms/etiology , Diagnostic Imaging , Glioma/etiology , Humans , Neoplasm Grading
9.
Cancer J ; 18(1): 20-5, 2012.
Article in English | MEDLINE | ID: mdl-22290253

ABSTRACT

Glioblastoma multiforme is a highly infiltrative tumor that typically has a central region of necrosis surrounded by contrast-enhancing proliferative tumor cells surrounded by diffuse isolated tumor cells that migrate into the brain. The goal of surgery is often directed toward the central necrotic region and the imaging-defined enhancing margin. To limit morbidity from removing functional brain tissue, the infiltrative tumor cells found in surrounding brain are generally not considered part of the surgical target. This is also the site where tumors recur after treatment. It is well accepted by most surgeons and neuro-oncologists that, when possible, aggressive resection of malignant gliomas is the preferred initial step in management. Although there are limited randomized prospective studies that address extent of resection and survival, the benefit of aggressive surgical resection will not be debated in this report. Tumor resection to the maximum extent that is safely possible can decrease tumor burden and thereby enhance the effects of adjuvant therapies, improve symptoms from mass effect, reduce the frequency of seizures, and provide tissue for pathological and genomic studies to better identify and test novel therapy.Surgery for glioblastoma is highly dependent on imaging. Magnetic resonance imaging can provide an anatomic definition of the lesion and functional capacity of critical cortical regions and allow for precise localization within the brain. The common use of stereotactic guidance, intraoperative imaging, functional magnetic resonance imaging, and physiologic monitoring have enhanced the surgeon's ability to achieve aggressive tumor removal while protecting the patient from neurologic impairment. This review will address the use of these techniques as an important first step in managing patients with glioblastoma.


Subject(s)
Brain Neoplasms/surgery , Glioblastoma/surgery , Aged , Aged, 80 and over , Brain Neoplasms/drug therapy , Brain Neoplasms/pathology , Brain Neoplasms/radiotherapy , Combined Modality Therapy , Glioblastoma/drug therapy , Glioblastoma/pathology , Glioblastoma/radiotherapy , Humans , Middle Aged
10.
Adv Drug Deliv Rev ; 64(7): 701-5, 2012 May 15.
Article in English | MEDLINE | ID: mdl-22210134

ABSTRACT

The central nervous system (CNS) poses a unique challenge for drug delivery. The blood-brain barrier significantly hinders the passage of systemically delivered therapeutics and the brain extracellular matrix limits the distribution and longevity of locally delivered agents. Polymeric nanoparticles represent a promising solution to these problems. Over the past 40years, substantial research efforts have demonstrated that polymeric nanoparticles can be engineered for effective systemic and local delivery of therapeutics to the CNS. Moreover, many of the polymers used in nanoparticle fabrication are both biodegradable and biocompatible, thereby increasing the clinical utility of this strategy. Here, we review the major advances in the development of polymeric nanoparticles for drug delivery to the CNS.


Subject(s)
Central Nervous System Diseases/drug therapy , Drug Delivery Systems/trends , Nanoparticles/administration & dosage , Polymers/administration & dosage , Animals , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Central Nervous System/drug effects , Central Nervous System/metabolism , Central Nervous System Diseases/metabolism , Drug Carriers/administration & dosage , Drug Carriers/chemistry , Drug Carriers/metabolism , Drug Delivery Systems/methods , Humans , Nanoparticles/chemistry , Polymers/chemistry , Polymers/metabolism
11.
Nat Mater ; 11(1): 82-90, 2011 Dec 04.
Article in English | MEDLINE | ID: mdl-22138789

ABSTRACT

Many synthetic polycationic vectors for non-viral gene delivery show high efficiency in vitro, but their usually excessive charge density makes them toxic for in vivo applications. Here we describe the synthesis of a series of high molecular weight terpolymers with low charge density, and show that they exhibit efficient gene delivery, some surpassing the efficiency of the commercial transfection reagents Polyethylenimine and Lipofectamine 2000. The terpolymers were synthesized via enzyme-catalyzed copolymerization of lactone with dialkyl diester and amino diol, and their hydrophobicity adjusted by varying the lactone content and by selecting a lactone comonomer of specific ring size. Targeted delivery of the pro-apoptotic TRAIL gene to tumour xenografts by one of the terpolymers results in significant inhibition of tumour growth, with minimal toxicity both in vitro and in vivo. Our findings suggest that the gene delivery ability of the terpolymers stems from their high molecular weight and increased hydrophobicity, which compensates for their low charge density.


Subject(s)
Gene Transfer Techniques , Polyamines/chemistry , Polymers/chemistry , Animals , Cell Line, Tumor , Female , Flow Cytometry , Genetic Therapy/methods , HEK293 Cells , Humans , In Situ Nick-End Labeling , Mice , Mice, Nude , Nanoparticles/chemistry , Nanoparticles/toxicity , Neoplasms/therapy , Polyamines/toxicity , Polymers/toxicity , Toxicity Tests
12.
Drug Deliv Transl Res ; 1(1): 34-42, 2011 Feb 01.
Article in English | MEDLINE | ID: mdl-21691426

ABSTRACT

Direct delivery of chemotherapy agents to the brain via degradable polymer delivery systems-such as Gliadel®-is a clinically proven method for treatment of glioblastoma multiforme, but there are important limitations with the current technology-including the requirement for surgery, profound local tissue toxicity, and limitations in diffusional penetration of agents-that limit its application and effectiveness. Here, we demonstrate another technique for direct, controlled delivery of chemotherapy to the brain that provides therapeutic benefit with fewer limitations. In our new approach, camptothecin (CPT)-loaded poly(lacticco-glycolic acid) (PLGA) nanoparticles are infused via convection-enhanced delivery (CED) to a stereotactically defined location in the brain, allowing simultaneous control of location, spread, and duration of drug release. To test this approach, CPT-PLGA nanoparticles (~100 nm in diameter) were synthesized with 25% drug loading. When these nanoparticles were incubated in culture with 9L gliosarcoma cells, the IC50 of CPT-PLGA nanoparticles was 0.04 µM, compared to 0.3 µM for CPT alone. CPT-PLGA nanoparticles stereotactically delivered by CED improved survival in rats with intracranial 9L tumors: the median survival for rats treated with CPT-PLGA nanoparticles (22 days) was significantly longer than unloaded nanoparticles (15 days) and free CPT infusion (17 days). CPT-PLGA nanoparticle treatment also produced significantly more long-term survivors (30% of animals were free of disease at 60 days) than any other treatment. CPT was present in tissues harvested up to 53 days post-infusion, indicating prolonged residence at the local site of administration. These are the first results to demonstrate the effectiveness of combining polymer-controlled release nanoparticles with CED in treating fatal intracranial tumors.

14.
J Neurooncol ; 95(3): 437-443, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19554263

ABSTRACT

Tumors of the optic chiasm are relatively uncommon and usually associated with phakomatoses such as neurofibromatosis. Even more rare is the presentation of a primary, non-exophytic, isolated optic chiasm germ cell tumor (GCT). These tumors have imaging characteristics nearly indistinguishable from optic chiasmatic gliomas (OCGs). Herein we describe two cases of young men who presented with similar findings of progressive, painless visual loss and hypothalamic-pituitary-adrenal axis dysfunction including diabetes insipidus. Brain imaging was non-diagnostic and suggestive of an OCG. Pathology demonstrated GCTs in each case highlighting the importance of biopsy confirmation of the diagnosis. Both patients underwent a pterional craniotomy and sub-frontal approach to the optic chiasm. The chiasm was diffusely enlarged and discolored in each case without evidence of sellar, suprasellar or perichiasmatic pathology. Pathology demonstrated a malignant mixed GCT in the first patient and a germinoma in the second. This case series highlights the importance of tissue biopsy for patients with progressive symptoms from optic chiasm tumors. Furthermore, this is the first report of a primary, non-exophytic malignant mixed GCT. As the treatment regimens differ widely between optic chiasm GCTs and chiasm gliomas, tissue diagnosis is important.


Subject(s)
Magnetic Resonance Imaging , Neoplasms, Germ Cell and Embryonal/pathology , Optic Chiasm/pathology , Optic Nerve Neoplasms/pathology , Biopsy , Child , Craniotomy , Diabetes Insipidus/pathology , Diabetes Insipidus/surgery , Diabetes Insipidus/therapy , Humans , Male , Neoplasms, Germ Cell and Embryonal/surgery , Neoplasms, Germ Cell and Embryonal/therapy , Optic Chiasm/surgery , Optic Nerve Neoplasms/surgery , Optic Nerve Neoplasms/therapy , Young Adult
15.
J Neurooncol ; 92(3): 253-9, 2009 May.
Article in English | MEDLINE | ID: mdl-19357953

ABSTRACT

Over the past two decades, the accumulated clinical and research experience has improved our understanding the biology of WHO grade II gliomas (G2G). While there have been relatively few randomized clinical trials in this population, those that exist and the experience from clinical reports have enhanced our understanding of how these tumors progressively increase in size, accumulate additional genetic mutations and ultimately transform into high-grade lesions. Our ability to reliably predict the time sequence of this transformation remains a challenge; however, recent findings have started to clarify selection criteria for adjuvant treatment. G2G remain a fatal disease for many patients. Continued investigation into the biology of these lesions will likely provide the information needed to select more appropriate therapy based on biological and genetic differences in these unique lesions. Some of this information will be derived from the study of high-grade lesions. However, experience has shown that much of the work on high-grade lesions is also applicable to low-grade lesions.


Subject(s)
Brain Neoplasms/diagnosis , Brain Neoplasms/therapy , Glioma/diagnosis , Glioma/therapy , Clinical Trials as Topic , Humans , World Health Organization
17.
J Neurooncol ; 92(2): 203-9, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19030779

ABSTRACT

Fibrous histiocytomas are rare lesions, more commonly encountered in soft tissues and bones. They are uncommon as an intracranial lesion. Although there have been several reports about malignant fibrous histiocytomas, less is known about the benign variant of these intracranial tumors as they are often misclassified as other types of tumors. We describe a child who presented with seizure and was subsequently found to have a large temporal lesion. Pathology revealed benign fibrous histiocytoma. We also review other cases reported in the literature in an effort to provide further insight into the diagnosis and management of this rare tumor.


Subject(s)
Brain Neoplasms/pathology , Brain Neoplasms/surgery , Histiocytoma, Benign Fibrous/pathology , Histiocytoma, Benign Fibrous/surgery , Brain Neoplasms/complications , Child , Female , Histiocytoma, Benign Fibrous/complications , Humans , Immunohistochemistry , Microscopy, Electron, Transmission , Neurosurgical Procedures , Seizures/etiology
18.
J Neurosci ; 28(8): 1882-93, 2008 Feb 20.
Article in English | MEDLINE | ID: mdl-18287505

ABSTRACT

Metastatic tumors and malignant gliomas make up the majority of cancers in the brain. They are invariably fatal and there is currently no cure. From in vitro comparisons of a number of viruses, we selected one that appeared the best in selectively killing glioblastoma cells. This replication-competent virus, the glioma-adapted vesicular stomatis virus strain VSVrp30a, was used for in vivo tests with the underlying view that infection of tumor cells will lead to an increase in the number of viruses subsequently released to kill additional tumor cells. Intravenous injection of VSVrp30a expressing a green fluorescent protein reporter, rapidly targeted and destroyed multiple types of human and mouse tumors implanted in the mouse brain, including glioblastoma and mammary tumors. When tumors were implanted both in the brain and peripherally, emulating systemic cancer metastasis, tumors inside and outside the brain were simultaneously infected. Intranasal inoculation, leading to olfactory nerve transport of the virus into the brain, selectively infected and killed olfactory bulb tumors. Neither control cortical wounds nor transplanted normal mouse or human cells were targeted, indicating viral tumor selectivity. Control viruses, including pseudorabies, adeno-associated, or replication-deficient VSV, did not infect the brain tumor. Confocal laser time-lapse imaging through a cranial window showed that intravenous VSV infects the tumor at multiple sites and kills migrating tumor cells. Disrupted tumor vasculature, suggested by dye leakage, may be the port of entry for intravenously delivered VSV. Quantitative PCR analysis of how VSVrp30a selectively infected tumor cells suggested multiple mechanisms, including cell surface binding and internalization.


Subject(s)
Brain Neoplasms/pathology , Brain Neoplasms/secondary , Glioma/pathology , Glioma/secondary , Vesiculovirus , Animals , Antineoplastic Agents/administration & dosage , Brain Neoplasms/drug therapy , Carcinoma/drug therapy , Carcinoma/pathology , Carcinoma/secondary , Cell Line, Tumor , Cells, Cultured , Glioma/drug therapy , Humans , Mice , Mice, Nude , Mice, SCID , Xenograft Model Antitumor Assays/methods
19.
J Clin Oncol ; 25(7): 837-44, 2007 Mar 01.
Article in English | MEDLINE | ID: mdl-17327604

ABSTRACT

PURPOSE: Glioblastoma multiforme (GBM) is a devastating brain tumor with a median survival of 6 months after recurrence. Cintredekin besudotox (CB) is a recombinant protein consisting of interleukin-13 (IL-13) and a truncated form of Pseudomonas exotoxin (PE38QQR). Convection-enhanced delivery (CED) is a locoregional-administration method leading to high-tissue concentrations with large volume of distributions. We assessed the use of intracerebral CED to deliver CB in patients with recurrent malignant glioma (MG). PATIENTS AND METHODS: Three phase I clinical studies evaluated intracerebral CED of CB along with tumor resection. The main objectives were to assess the tolerability of various concentrations and infusion durations; tissue distribution; and methods for optimizing delivery. All patients underwent tumor resection followed by a single intraparenchymal infusion (in addition to the intraparenchymal one following resection), with a portion of patients who had a preresection intratumoral infusion. RESULTS: A total of 51 patients with MG were treated including 46 patients with GBM. The maximum tolerated intraparenchymal concentration was 0.5 microg/mL and tumor necrosis was observed at this concentration. Infusion durations of up to 6 days were well tolerated. Postoperative catheter placement appears to be important for optimal drug distribution. CB- and procedure-related adverse events were primarily limited to the CNS. Overall median survival for GBM patients is 42.7 weeks and 55.6 weeks for patients with optimally positioned catheters with patient follow-up extending beyond 5 years. CONCLUSION: CB appears to have a favorable risk-benefit profile. CED is a complex delivery method requiring catheter placement via a second procedure to achieve accurate catheter positioning, better drug distribution, and better outcome.


Subject(s)
Drug Delivery Systems/methods , Exotoxins/administration & dosage , Glioma/drug therapy , Immunotoxins/administration & dosage , Interleukin-13/administration & dosage , Supratentorial Neoplasms/drug therapy , Adult , Aged , Catheterization , Convection , Exotoxins/adverse effects , Exotoxins/pharmacokinetics , Female , Glioma/mortality , Humans , Interleukin-13/adverse effects , Interleukin-13/pharmacokinetics , Male , Middle Aged , Neoplasm Recurrence, Local , Recombinant Fusion Proteins , Supratentorial Neoplasms/mortality , Tissue Distribution
20.
J Neurooncol ; 82(2): 221-5, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17029014

ABSTRACT

OBJECTIVE: A subset of patients with malignant glioma comes to medical attention before their masses show rim enhancement and central necrosis. Tumors in those cases are frequently located in eloquent areas of the brain. Tissue diagnosis is limited to stereotactic biopsy providing limited material for accurate grading. We conducted this study to determine whether imaging characteristics of early stages of malignant gliomas could aid in timely definitive diagnosis. METHODS: We retrospectively analyzed patients with newly diagnosed malignant glioma seen at the Yale Brain Tumor Center between 2002 and 2005. Patients with typical radiographic presentation were excluded. RESULTS: Of 89 patients, eight meeting the inclusion criteria were identified. In five patients, patchy or small nodular enhancing lesions without central necrosis were present within the tumor mass. Diffusion-weighted imaging (DWI) showed areas of increased signal intensity in all cases. Apparent diffusion coefficient maps (ADC) revealed low-signal intensity in corresponding areas. At the time of imaging, biopsy was performed in seven patients but diagnosis of malignant glioma could only be established prior to further tumor growth in four cases. CONCLUSIONS: The diagnosis in the early stages of malignant glioma can be challenging in a subset of cases. Information obtained through DWI should be incorporated in the clinical decision-making process. Mass lesions displaying decreased water diffusion indicating high cellularity, are suggestive of a high-grade glioma. Biopsies are recommended. However, even when biopsies are inconclusive, a strong suspicion of malignant glioma should be considered.


Subject(s)
Brain Neoplasms/diagnosis , Diffusion Magnetic Resonance Imaging , Glioma/diagnosis , Aged , Early Diagnosis , Female , Humans , Male , Middle Aged
SELECTION OF CITATIONS
SEARCH DETAIL
...