Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 86
Filter
1.
Cell ; 157(3): 580-94, 2014 Apr 24.
Article in English | MEDLINE | ID: mdl-24726434

ABSTRACT

Developmental fate decisions are dictated by master transcription factors (TFs) that interact with cis-regulatory elements to direct transcriptional programs. Certain malignant tumors may also depend on cellular hierarchies reminiscent of normal development but superimposed on underlying genetic aberrations. In glioblastoma (GBM), a subset of stem-like tumor-propagating cells (TPCs) appears to drive tumor progression and underlie therapeutic resistance yet remain poorly understood. Here, we identify a core set of neurodevelopmental TFs (POU3F2, SOX2, SALL2, and OLIG2) essential for GBM propagation. These TFs coordinately bind and activate TPC-specific regulatory elements and are sufficient to fully reprogram differentiated GBM cells to "induced" TPCs, recapitulating the epigenetic landscape and phenotype of native TPCs. We reconstruct a network model that highlights critical interactions and identifies candidate therapeutic targets for eliminating TPCs. Our study establishes the epigenetic basis of a developmental hierarchy in GBM, provides detailed insight into underlying gene regulatory programs, and suggests attendant therapeutic strategies. PAPERCLIP:


Subject(s)
Brain Neoplasms/genetics , Brain Neoplasms/pathology , Glioblastoma/genetics , Glioblastoma/pathology , Neoplastic Stem Cells/pathology , Basic Helix-Loop-Helix Transcription Factors/metabolism , Brain Neoplasms/metabolism , Cell Differentiation , Cell Line, Tumor , Cells, Cultured , Co-Repressor Proteins/metabolism , Epigenesis, Genetic , Gene Expression Regulation, Neoplastic , Glioblastoma/metabolism , Humans , Neoplastic Stem Cells/metabolism , Nerve Tissue Proteins/metabolism , Oligodendrocyte Transcription Factor 2 , Regulatory Elements, Transcriptional , Transcription Factors/metabolism
2.
Proc Natl Acad Sci U S A ; 115(36): E8388-E8394, 2018 09 04.
Article in English | MEDLINE | ID: mdl-30082399

ABSTRACT

Aggressive neurosurgical resection to achieve sustained local control is essential for prolonging survival in patients with lower-grade glioma. However, progression in many of these patients is characterized by local regrowth. Most lower-grade gliomas harbor isocitrate dehydrogenase 1 (IDH1) or IDH2 mutations, which sensitize to metabolism-altering agents. To improve local control of IDH mutant gliomas while avoiding systemic toxicity associated with metabolic therapies, we developed a precision intraoperative treatment that couples a rapid multiplexed genotyping tool with a sustained release microparticle (MP) drug delivery system containing an IDH-directed nicotinamide phosphoribosyltransferase (NAMPT) inhibitor (GMX-1778). We validated our genetic diagnostic tool on clinically annotated tumor specimens. GMX-1778 MPs showed mutant IDH genotype-specific toxicity in vitro and in vivo, inducing regression of orthotopic IDH mutant glioma murine models. Our strategy enables immediate intraoperative genotyping and local application of a genotype-specific treatment in surgical scenarios where local tumor control is paramount and systemic toxicity is therapeutically limiting.


Subject(s)
Brain Neoplasms , Cyanides/pharmacology , Genotype , Glioma , Guanidines/pharmacology , Isocitrate Dehydrogenase/genetics , Molecular Targeted Therapy/methods , Mutation , Neoplasm Proteins/genetics , Animals , Brain Neoplasms/drug therapy , Brain Neoplasms/enzymology , Brain Neoplasms/genetics , Drug Delivery Systems/methods , Female , Glioma/drug therapy , Glioma/enzymology , Glioma/genetics , Humans , Male , Mice , Mice, SCID , Xenograft Model Antitumor Assays
3.
Skeletal Radiol ; 50(4): 723-730, 2021 Apr.
Article in English | MEDLINE | ID: mdl-32968823

ABSTRACT

OBJECTIVE: To implement an automated quality assurance tool to prospectively track discrepancies in musculoskeletal (MSK) exams submitted for second-opinion radiology interpretation at a tertiary center. METHODS: From 2013 to 2020, a standardized template was included in re-interpretation MSK reports, and a concordance assessment compared with primary interpretation was assigned. Analysis of standardized template implementation and discordance rates was performed. Of the re-interpretations that demonstrated likely clinically relevant discordance, a sample was randomly selected and the EMR was reviewed to evaluate the impact on patient care and change in medical management. RESULTS: A total of 1052 re-interpretations were identified using the standardized template. Services with higher requests for second-opinion interpretation were oncology (n = 351, 33%) and orthopedic surgery (n = 255, 24%). Overall utilization rate of the template was 65% with marked decreased during the last year (22% rate). In comparison to the primary report, there was a 30% discordance rate (n = 309) with 18% (n = 184) classified as likely clinically relevant. From the subset of discrepancies that could be clinically relevant, there was a change in management in 63% of the cases (19/30) with the re-interpretation ultimately proving correct in 80% of the cases (24/30). CONCLUSION: Implementation of a quality assurance tool embedded in the radiology workflow of second-opinion interpretations can facilitate the analysis of patient care impact; however, stricter implementation is necessary. Oncologic studies were the most common indication for re-interpretations. Although the primary and second interpretations in the majority of cases were in agreement, subspecialty MSK radiology interpretation was shown to be more accurate than primary interpretations and impacted clinical management in cases of discrepancy.


Subject(s)
Magnetic Resonance Imaging , Tomography, X-Ray Computed , Humans , Observer Variation , Referral and Consultation , Retrospective Studies
4.
J Neurooncol ; 145(1): 97-105, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31456142

ABSTRACT

BACKGROUND: H3 K27M-mutant diffuse midline glioma is a fatal malignancy with no proven medical therapies. The entity predominantly occurs in children and young adults. ONC201 is a small molecule selective antagonist of dopamine receptor D2/3 (DRD2/3) with an exceptional safety profile. Following up on a durable response in the first H3 K27M-mutant diffuse midline glioma patient who received ONC201 (NCT02525692), an expanded access program was initiated. METHODS: Patients with H3 K27M-mutant gliomas who received at least prior radiation were eligible. Patients with leptomeningeal spread were excluded. All patients received open-label ONC201 orally once every week. Safety, radiographic assessments, and overall survival were regularly assessed at least every 8Ā weeks by investigators. As of August 2018, a total of 18 patients with H3 K27M-mutant diffuse midline glioma or DIPG were enrolled to single patient expanded access ONC201 protocols. Among the 18 patients: seven adult (> 20Ā years old) and seven pediatric (< 20Ā years old) patients initiated ONC201 with recurrent disease and four pediatric patients initiated ONC201 following radiation, but prior to disease recurrence. FINDINGS: Among the 14 patients with recurrent disease prior to initiation of ONC201, median progression-free survival is 14Ā weeks and median overall survival is 17Ā weeks. Three adults among the 14 recurrent patients remain on treatment progression-free with a median follow up of 49.6 (range 41-76.1) weeks. Among the 4 pediatric patients who initiated adjuvant ONC201 following radiation, two DIPG patients remain progression-free for at least 53 and 81Ā weeks. Radiographic regressions, including a complete response, were reported by investigators in a subset of patients with thalamic and pontine gliomas, along with improvements in disease-associated neurological symptoms. INTERPRETATION: The clinical outcomes and radiographic responses in these patients provide the preliminary, and initial clinical proof-of-concept for targeting H3 K27M-mutant diffuse midline glioma with ONC201, regardless of age or location, providing rationale for robust clinical testing of the agent.


Subject(s)
Antineoplastic Agents/therapeutic use , Brain Neoplasms/drug therapy , Glioma/drug therapy , Heterocyclic Compounds, 4 or More Rings/therapeutic use , Histones/genetics , Mutation , Receptors, Dopamine D2/chemistry , Adolescent , Adult , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Child , Child, Preschool , Female , Follow-Up Studies , Glioma/genetics , Glioma/pathology , Humans , Imidazoles , Male , Prognosis , Pyridines , Pyrimidines , Survival Rate , Young Adult
5.
Neuroradiology ; 61(2): 225-227, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30565056

ABSTRACT

The T2-FLAIR mismatch sign, in which a low-grade glioma is hyperintense on T2-weighted MR and centrally hypointense on T2-weighted FLAIR MR, has been reported as 100% specific for IDH-mutant astrocytomas in several series. We report several cases of "false positive" T2-FLAIR mismatch sign occurring outside the context of IDH-mutant astrocytomas, predominantly in children or young adults with pediatric-type gliomas. These results suggest caution in the interpretation of the T2-FLAIR mismatch sign in the pediatric glioma population.


Subject(s)
Brain Neoplasms/diagnostic imaging , Glioma/diagnostic imaging , Magnetic Resonance Imaging/methods , Adolescent , Adult , Brain Neoplasms/pathology , Child , False Positive Reactions , Glioma/pathology , Humans , Infant , Male , Retrospective Studies
6.
Neuroradiology ; 61(9): 1023-1031, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31134296

ABSTRACT

PURPOSE: There is variability in survival within IDH mutant gliomas determined by chromosomal events. Copy number variation (CNV) abundance associated with survival in low-grade and IDH mutant astrocytoma has been reported. Our purpose was to correlate the extent of genome-wide CNV abundance in IDH mutant astrocytomas with MRI features. METHODS: Presurgical MRI and CNV plots derived from Illumina 850k EPIC DNA methylation arrays of 18 cases of WHO grade II-IV IDH mutant astrocytomas were reviewed. IDH mutant astrocytomas were divided into CNV stable group (CNV-S) with ≤ 3 chromosomal gains or losses and lack of focal gene amplifications and CNV unstable group (CNV-U) with > 3 large chromosomal gains/losses and/or focal amplifications. The associations between MR features, relative cerebral blood volume (rCBV), CNV abundance, and time to progression were assessed. Tumor rCBV estimates were obtained using DSC T2* perfusion analysis. RESULTS: There were nine (50%) CNV-S and nine (50%) CNV-U IDH mutant astrocytomas. CNV-U tumors showed larger mean tumor size (P = 0.004) and maximum diameter on FLAIR (P = 0.004) and also demonstrated significantly higher median rCBV than CNV-S tumors (2.62 vs 0.78, P = 0.019). CNV-U tumors tended to have shorter time to progression although without statistical significance (P = 0.393). CONCLUSIONS: Larger size/diameter and higher rCBVs were seen associated CNV-U astrocytomas, suggesting a correlation of aggressive imaging phenotype with unstable and aggressive genotype in IDH mutant astrocytomas.


Subject(s)
Astrocytoma/diagnostic imaging , Astrocytoma/genetics , Brain Neoplasms/diagnostic imaging , Brain Neoplasms/genetics , DNA Copy Number Variations/genetics , Isocitrate Dehydrogenase/genetics , Adult , Aged , Aged, 80 and over , Astrocytoma/mortality , Brain Neoplasms/mortality , Female , Humans , Magnetic Resonance Imaging , Male , Middle Aged , Phenotype , Retrospective Studies , Young Adult
7.
Int J Cancer ; 141(11): 2348-2358, 2017 12 01.
Article in English | MEDLINE | ID: mdl-28801914

ABSTRACT

Despite the current standard of multimodal management, glioblastoma (GBM) inevitably recurs and effective therapy is not available for recurrent disease. A subset of tumor cells with stem-like properties, termed GBM stem-like cells (GSCs), are considered to play a role in tumor relapse. Although oncolytic herpes simplex virus (oHSV) is a promising therapeutic for GBM, its efficacy against recurrent GBM is incompletely characterized. Transforming growth factor beta (TGF-Ɵ) plays vital roles in maintaining GSC stemness and GBM pathogenesis. We hypothesized that oHSV and TGF-Ɵ inhibitors would synergistically exert antitumor effects for recurrent GBM. Here we established a panel of patient-derived recurrent tumor models from GBMs that relapsed after postsurgical radiation and chemotherapy, based on GSC-enriched tumor sphere cultures. These GSCs are resistant to the standard-of-care temozolomide but susceptible to oHSVs G47Δ and MG18L. Inhibition of TGF-Ɵ receptor kinase with selective targeted small molecules reduced clonogenic sphere formation in all tested recurrent GSCs. The combination of oHSV and TGF-ƟR inhibitor was synergistic in killing recurrent GSCs through, in part, an inhibitor-induced JNK-MAPK blockade and increase in oHSV replication. In vivo, systemic treatment with TGF-ƟR inhibitor greatly enhanced the antitumor effects of single intratumoral oHSV injections, resulting in cures in 60% of mice bearing orthotopic recurrent GBM. These results reveal a novel synergistic interaction of oHSV therapy and TGF-Ɵ signaling blockade, and warrant further investigations aimed at clinical translation of this combination strategy for GBM patients.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Brain Neoplasms/pathology , Glioblastoma/pathology , Oncolytic Virotherapy/methods , Transforming Growth Factor beta/antagonists & inhibitors , Animals , Blotting, Western , Humans , Immunohistochemistry , Mice , Mice, SCID , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/pathology , Real-Time Polymerase Chain Reaction , Signal Transduction/drug effects , Simplexvirus , Xenograft Model Antitumor Assays
8.
J Neurooncol ; 131(3): 603-610, 2017 02.
Article in English | MEDLINE | ID: mdl-27853960

ABSTRACT

Targeting tumor angiogenesis is a potential therapeutic strategy for glioblastoma because of its high vascularization. Tivozanib is an oral pan-VEGF receptor tyrosine kinase inhibitor that hits a central pathway in glioblastoma angiogenesis. We conducted a phase II study to test the effectiveness of tivozanib in patients with recurrent glioblastoma. Ten adult patients were enrolled and treated with tivozanib 1.5Ā mg daily, 3 weeks on/1 week off in 28-day cycles. Brain MRI and blood biomarkers of angiogenesis were performed at baseline, within 24-72Ā h of treatment initiation, and monthly thereafter. Dynamic contrast enhanced MRI, dynamic susceptibility contrast MRI, and vessel architecture imaging were used to assess vascular effects. Resting state MRI was used to assess brain connectivity. Best RANO criteria responses were: 1 complete response, 1 partial response, 4 stable diseases, and 4 progressive disease (PD). Two patients were taken off study for toxicity and 8 patients were taken off study for PD. Median progression-free survival was 2.3 months and median overall survival was 8.1 months. Baseline abnormal tumor vascular permeability, blood flow, tissue oxygenation and plasma sVEGFR2 significantly decreased and plasma PlGF and VEGF increased after treatment, suggesting an anti-angiogenic effect of tivozanib. However, there were no clear structural changes in vasculature as vessel caliber and enhancing tumor volume did not significantly change. Despite functional changes in tumor vasculature, tivozanib had limited anti-tumor activity, highlighting the limitations of anti-VEGF monotherapy. Future studies in glioblastoma should leverage the anti-vascular activity of agents targeting VEGF to enhance the activity of other therapies.


Subject(s)
Antineoplastic Agents/therapeutic use , Brain Neoplasms/drug therapy , Glioblastoma/drug therapy , Neoplasm Recurrence, Local/drug therapy , Phenylurea Compounds/therapeutic use , Quinolines/therapeutic use , Receptors, Vascular Endothelial Growth Factor/antagonists & inhibitors , Administration, Oral , Aged , Biomarkers, Tumor/blood , Brain Neoplasms/blood , Brain Neoplasms/diagnostic imaging , Brain Neoplasms/pathology , Female , Glioblastoma/blood , Glioblastoma/diagnostic imaging , Glioblastoma/pathology , Humans , Male , Middle Aged , Neoplasm Recurrence, Local/diagnostic imaging , Neoplasm Recurrence, Local/pathology , Neovascularization, Pathologic/drug therapy , Receptors, Vascular Endothelial Growth Factor/blood , Survival Analysis , Treatment Outcome
9.
Oncologist ; 21(2): 214-9, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26834160

ABSTRACT

BACKGROUND: Recurrent mutations in the isocitrate dehydrogenase 1 (IDH1) and IDH2 genes, which are frequent in gliomas, result in marked accumulation of the metabolic by-product 2-hydroxyglutarate (2-HG) within tumors. In other malignancies, such as acute myeloid leukemia, presence of IDH mutation is associated with elevated 2-HG levels in serum or urine compartments. Circulating 2-HG in patients with glial malignancies has not been thoroughly investigated. METHODS: In this study, we analyzed 2-HG levels in the serum and urine of a large set of patients with IDH-mutant and IDH-wild-type glioma, and the cerebrospinal fluid (CSF) from a subset of this cohort. RESULTS: We found that 2-HG was elevated in the urine of patients with IDH-mutant versus IDH-wild-type glioma, although no significant differences in 2-HG levels were observed in the serum or the small set of CSF samples obtained. Among patients with IDH-mutant glioma, 2-HG levels did not differ based on the histopathologic grade, genetic subtype (TP53 mutant or 1p/19q codeleted), presence of a canonical (IDH1 R132H) or noncanonical (any other IDH variant) mutation, or treatment type. CONCLUSION: Our finding suggests that urinary 2-HG is increased among patients with IDH-mutant gliomas, and may represent a future surrogate, noninvasive biomarker to aid in diagnosis, prognosis, and management. IMPLICATIONS FOR PRACTICE: Patients with glioma who harbor mutations in isocitrate dehydrogenase genes showed selective elevation of the oncometabolite 2-hydroxyglutarate in the urine. Similar elevations were not identified in the serum or cerebrospinal fluid. 2-Hydroxyglutarate may serve as a useful, noninvasive biomarker to stratify patients newly diagnosed with glioma with regard to prognosis and management.


Subject(s)
Biomarkers, Tumor/urine , Glioma/urine , Glutarates/urine , Isocitrate Dehydrogenase/genetics , Adult , Aged , Female , Genotype , Glioma/blood , Glioma/genetics , Glioma/pathology , Glutarates/blood , Humans , Male , Middle Aged , Mutation
10.
J Comput Assist Tomogr ; 40(2): 238-42, 2016.
Article in English | MEDLINE | ID: mdl-26571058

ABSTRACT

PURPOSE: The aim of this study was to determine an association between fall-related hip and/or pelvic fractures and gluteus medius and minimus atrophy. METHODS: Retrospective review of 64 patients with fall-related hip/pelvic fractures and 96 age- and sex-stratified controls was performed. Gluteus medius, gluteus minimus, tensor fascia lata, and iliopsoas atrophy was scored using a standard scale. Statistical analysis was performed. RESULTS: There is a significant difference (P < 0.0001) in gluteus medius and minimus atrophy in the fracture versus control groups. Presence of gluteus atrophy was predictive of fall-related fracture (odds ratio, 2.15; 95% confidence interval, 1.08-4.31). There is no significant difference in tensor fascia lata (P = 0.47) or iliopsoas (P = 0.15) atrophy between the 2 groups. Gluteus atrophy increased with age (r = 0.41, P < 0.0001). In unilateral fractures, there is a significant difference (P = 0.0002) in ipsilateral versus contralateral gluteus medius atrophy. CONCLUSIONS: Gluteus medius and minimus muscle atrophy is greater in fall-related hip/pelvic fractures, which may predispose the elderly to falls.


Subject(s)
Accidental Falls/statistics & numerical data , Hip Fractures/diagnostic imaging , Muscle, Skeletal/diagnostic imaging , Muscular Atrophy/diagnostic imaging , Tomography, X-Ray Computed , Aged , Aged, 80 and over , Aging , Female , Hip/diagnostic imaging , Hip Fractures/complications , Humans , Male , Middle Aged , Muscular Atrophy/complications , Pelvis/injuries , Retrospective Studies
11.
Proc Natl Acad Sci U S A ; 110(47): 19059-64, 2013 Nov 19.
Article in English | MEDLINE | ID: mdl-24190997

ABSTRACT

Antiangiogenic therapy has shown clear activity and improved survival benefit for certain tumor types. However, an incomplete understanding of the mechanisms of action of antiangiogenic agents has hindered optimization and broader application of this new therapeutic modality. In particular, the impact of antiangiogenic therapy on tumor blood flow and oxygenation status (i.e., the role of vessel pruning versus normalization) remains controversial. This controversy has become critical as multiple phase III trials of anti-VEGF agents combined with cytotoxics failed to show overall survival benefit in newly diagnosed glioblastoma (nGBM) patients and several other cancers. Here, we shed light on mechanisms of nGBM response to cediranib, a pan-VEGF receptor tyrosine kinase inhibitor, using MRI techniques and blood biomarkers in prospective phase II clinical trials of cediranib with chemoradiation vs. chemoradiation alone in nGBM patients. We demonstrate that improved perfusion occurs only in a subset of patients in cediranib-containing regimens, and is associated with improved overall survival in these nGBM patients. Moreover, an increase in perfusion is associated with improved tumor oxygenation status as well as with pharmacodynamic biomarkers, such as changes in plasma placenta growth factor and sVEGFR2. Finally, treatment resistance was associated with elevated plasma IL-8 and sVEGFR1 posttherapy. In conclusion, tumor perfusion changes after antiangiogenic therapy may distinguish responders vs. nonresponders early in the course of this expensive and potentially toxic form of therapy, and these results may provide new insight into the selection of glioblastoma patients most likely to benefit from anti-VEGF treatments.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Brain Neoplasms/drug therapy , Brain Neoplasms/metabolism , Glioblastoma/drug therapy , Glioblastoma/metabolism , Oxygen/metabolism , Protein Kinase Inhibitors/pharmacology , Biomarkers, Tumor/blood , Brain Neoplasms/pathology , Brain Neoplasms/radiotherapy , DNA Modification Methylases/metabolism , DNA Repair Enzymes/metabolism , Dacarbazine/analogs & derivatives , Enzyme-Linked Immunosorbent Assay , Glioblastoma/pathology , Glioblastoma/radiotherapy , Humans , Magnetic Resonance Imaging , Polymerase Chain Reaction , Prospective Studies , Quinazolines , Receptor Protein-Tyrosine Kinases/metabolism , Statistics, Nonparametric , Temozolomide , Treatment Outcome , Tumor Suppressor Proteins/metabolism
12.
Cancer ; 121(2): 226-33, 2015 Jan 15.
Article in English | MEDLINE | ID: mdl-25204639

ABSTRACT

BACKGROUND: High-dose thiotepa, busulfan, and cyclophosphamide (TBC) with autologous stem cell transplantation (ASCT) has been used in patients with central nervous system (CNS) involvement by non-Hodgkin lymphoma (NHL). Despite limited penetration into the CNS, rituximab is active in primary CNS NHL. Therefore, high-dose rituximab was combined with TBC for ASCT in patients with CNS NHL. METHODS: A single-arm phase 2 trial using high-dose rituximab with cytarabine for stem cell mobilization followed by high-dose rituximab combined with thiotepa, busulfan, and cyclophosphamide (R-TBC) for ASCT was conducted. Doses of rituximab at 1000 mg/m(2) were given on days 1 and 8 of mobilization and on days -9 and -2 of TBC. The primary endpoint was efficacy. RESULTS: Thirty patients were enrolled. Eighteen patients had primary CNS NHL (12 with complete remission (CR)/first partial remission (PR1) and 6 with CR/PR2), and 12 patients had secondary CNS lymphoma (5 with CR/PR1 and 7 with CR/PR2 or beyond). All patients were in partial or complete remission. Twenty-nine patients proceeded to R-TBC ASCT. Two patients developed significant neurotoxicity. The 100-day nonrelapse mortality rate was 0%, and 1 patient died because of nonrelapse causes 5 months after ASCT. For all patients, at a median follow-up of 24 months (range, 12-40 months), the estimated 2-year progression-free survival rate was 81% (95% confidence interval, 59%-92%), and the 2-year overall survival rate was 93% (95% confidence interval, 76%-98%). There were no relapses or deaths among the 18 patients with primary CNS lymphoma. CONCLUSIONS: For patients with CNS involvement by B-cell NHL and especially for patients with primary CNS NHL, R-TBC ASCT shows encouraging activity and merits further study.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Central Nervous System Neoplasms/therapy , Hematopoietic Stem Cell Transplantation/methods , Induction Chemotherapy/methods , Lymphoma, Non-Hodgkin/therapy , Transplantation Conditioning/methods , Adult , Aged , Antibodies, Monoclonal, Murine-Derived/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Busulfan/administration & dosage , Central Nervous System Neoplasms/drug therapy , Central Nervous System Neoplasms/surgery , Cyclophosphamide/administration & dosage , Cytarabine/administration & dosage , Disease-Free Survival , Drug Administration Schedule , Female , Humans , Infusions, Intravenous , Lymphoma, Non-Hodgkin/drug therapy , Lymphoma, Non-Hodgkin/surgery , Male , Massachusetts , Middle Aged , Rituximab , Thiotepa/administration & dosage , Transplantation, Autologous
13.
J Neurooncol ; 125(3): 609-30, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26530264

ABSTRACT

TARGET POPULATION: These recommendations apply to adult patients with recurrent low-grade glioma (LGG) with initial pathologic diagnosis of a WHO grade II infiltrative glioma (oligodendroglioma, astrocytoma, or oligo-astrocytoma). QUESTION: Do pathologic and molecular characteristics predict outcome/malignant transformation at recurrence? RECOMMENDATIONS: IDH STATUS AND RECURRENCE: (Level III) IDH mutation status should be determined as LGGs with IDH mutations have a shortened time to recurrence. It is unclear whether knowledge of IDH mutation status provides benefit in predicting time to progression or overall survival. TP53 STATUS AND RECURRENCE: (Level III) TP53 mutations occur early in LGG pathogenesis, remain stable, and are not recommended as a marker of predisposition to malignant transformation at recurrence or other measures of prognosis. MGMT STATUS AND RECURRENCE: (Level III) Assessment of MGMT status is recommended as an adjunct to assessing prognosis as LGGs with MGMT promoter methylation are associated with shorter PFS (in the absence of TMZ) and longer post-recurrence survival (in the presence of TMZ), ultimately producing similar overall survival to LGGs without MGMT methylation. The available retrospective reports are conflicting and comparisons between reports are limited CDK2NA STATUS AND RECURRENCE: (Level III) Assessment of CDK2NA status is recommended when possible as the loss of expression of the CDK2NA via either methylation or loss of chromosome 9p is associated with malignant progression of LGGs. PROLIFERATIVE INDEX AND RECURRENCE: (Level III) It is recommended that proliferative indices (MIB-1 or BUdR) be measured in LGGs as higher proliferation indices are associated with increased likelihood of recurrence and shorter progression free and overall survival. 1P/19Q STATUS AND RECURRENCE: There is insufficient evidence to make any recommendations. QUESTION: What role does chemotherapy have in LGG recurrence? RECOMMENDATIONS: TEMOZOLOMIDE AND RECURRENCE: (Level III) Temozolomide is recommended in the therapy of recurrent LGG as it may improve clinical symptoms. Oligodendrogliomas and tumors with 1p/19q co-deletion may derive the most benefit. PCV AND RECURRENCE: (Level III) PCV is recommended in the therapy of LGG at recurrence as it may improve clinical symptoms with the strongest evidence being for oligodendrogliomas. CARBOPLATIN AND RECURRENCE : (Level III) Carboplatin is not recommended as there is no significant benefit from carboplatin as single agent therapy for recurrent LGGs. OTHER TREATMENTS (NITROSUREAS, HYDROXYUREA/IMANITIB, IRINOTECAN, PACLITAXEL) AND RECURRENCE: There is insufficient evidence to make any recommendations. It is recommended that individuals with recurrent LGGs be enrolled in a properly designed clinical trial to assess these chemotherapeutic agents. QUESTION: What role does radiation have in LGG recurrence? RECOMMENDATIONS: RADIATION AT RECURRENCE WITH NO PREVIOUS IRRADIATION: (Level III) Radiation is recommended at recurrence if there was no previous radiation treatment. RE-IRRADIATION AT RECURRENCE: (Level III) It is recommended that re-irradiation be considered in the setting of LGG recurrence as it may provide benefit in disease control. SURGERY AT RECURRENCE: There is insufficient evidence to make any specific recommendations. It is recommended that individuals with recurrent LGGs be enrolled in a properly designed clinical trial to assess the role of surgery at recurrence.


Subject(s)
Brain Neoplasms , Glioma , Neoplasm Recurrence, Local , Humans , Brain Neoplasms/pathology , Brain Neoplasms/therapy , Evidence-Based Medicine , Glioma/pathology , Glioma/therapy , Neoplasm Grading , Neoplasm Recurrence, Local/therapy
14.
J Neurooncol ; 121(3): 627-34, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25503302

ABSTRACT

Targeting specific molecular alterations in glioblastoma (GBM) might more effectively kill tumor cells and increase survival. Vandetanib inhibits epidermal growth factor receptor and vascular endothelial growth factor receptor 2. Sirolimus inhibits mammalian target of rapamycin (mTOR), a member the phosphoinositide 3-Kinase signaling pathway. We sought to determine the maximum tolerated dose (MTD) and dose-limiting toxicity (DLT) of vandetanib combined with sirolimus. Twenty-two patients (14 men; 8 women) with recurrent GBM enrolled. Median age and KPS were 52.5 years and 90 %, respectively. Patients were naive to anti-VEGF and anti-EGF therapy and mTOR inhibitors, and not on CYP3A4-inducing drugs. Vandetanib and sirolimus were orally administered on a continuous daily dosing schedule in escalating dose cohorts. Ten patients enrolled in the dose escalation phase. Twelve more enrolled at the MTD to explore progression-free survival at 6 months (PFS6) in a single arm, single stage phase II-type design. In total, 19 patients received at least one dose at the MTD, and 15 completed at least 1 cycle at MTD. MTD was 200 mg vandetanib plus 2 mg sirolimus. The DLT was elevated AST/SGOT. The most common toxicities were lymphopenia, fatigue, rash, and hypophosphatemia. For 19 patients who received at least one dose at the MTD, including seven from the phase I group, two had a partial response [10.5 %; 95 % CI (1, 33 %)] and PFS6 was 15.8 % [95 % CI (3.9, 34.9 %)]. Vandetanib and sirolimus can be safely co-administered on a continuous, daily dosing schedule.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Brain Neoplasms/drug therapy , Glioblastoma/drug therapy , Neoplasm Recurrence, Local/drug therapy , Adult , Aged , Cohort Studies , Disease-Free Survival , Female , Humans , Male , Maximum Tolerated Dose , Middle Aged , Piperidines/administration & dosage , Piperidines/adverse effects , Quinazolines/administration & dosage , Quinazolines/adverse effects , Sirolimus/administration & dosage , Sirolimus/adverse effects
15.
J Neurooncol ; 121(3): 557-63, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25411098

ABSTRACT

Tumor cell infiltration is a major mechanism of treatment escape in glioblastoma. Src is an intracellular tyrosine kinase that mediates tumor cell motility and invasiveness. We evaluated the efficacy and safety of bosutinib, a tyrosine kinase inhibitor that potently inhibits Src and Abl, in patients with recurrent glioblastoma. In this two-arm study, patients with histologically confirmed recurrent glioblastoma and ≤2 relapses, not previously treated with anti-vascular endothelial growth factor (VEGF) therapy, were administered oral bosutinib 400 mg daily. Arm A planned for 6 patients who were candidates for surgical resection to be given bosutinib for 7-9 days prior to resection. Arm B was a two-stage design phase 2 trial targeting 30 patients. The primary endpoint was progression-free survival at 6 months (PFS6) in Arm B. After 9 patients enrolled onto stage 1 of Arm B, 9 (100 %) patients progressed within 6 months. Therefore, the study met the pre-specified criteria for early closure and both Arms were closed. In Arm B, Median PFS was 7.71 weeks and median OS was 50 weeks. Best objective response was stable disease in one patient (11.1 %). Seven patients (77.8 %) had treatment-related AEs of any grade and 2 (22.2 %) were grade ≥3. Arm A was closed after 2 patients enrolled. Src activation was evident in all archival tumor samples. Bosutinib monotherapy does not appear to be effective in recurrent glioblastoma. However, Src remains a potential target based on its upregulation in tumor samples and role in glioma invasion.


Subject(s)
Aniline Compounds/therapeutic use , Antineoplastic Agents/therapeutic use , Brain Neoplasms/drug therapy , Glioblastoma/drug therapy , Neoplasm Recurrence, Local/drug therapy , Nitriles/therapeutic use , Quinolines/therapeutic use , src-Family Kinases/antagonists & inhibitors , Adult , Brain Neoplasms/mortality , Disease-Free Survival , Female , Glioblastoma/mortality , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Proportional Hazards Models , src-Family Kinases/metabolism
16.
J Neurooncol ; 125(1): 123-31, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26285766

ABSTRACT

Iniparib is a prodrug that converts to highly reactive cytotoxic metabolites intracellularly with activity in preclinical glioma models. We investigated the maximum tolerated dose (MTD) of iniparib with monthly (m) and continuous (c) temozolomide (TMZ) dosing schedules in patients with malignant gliomas (MG). Adults with newly diagnosed MG who had successfully completed ≥80% of radiation (RT) and TMZ without toxicity received mTMZ dosing (150-200 mg/m(2) days 1-5/28 days) or cTMZ dosing (75 mg/m(2)/days Ɨ 6 weeks) in conjunction with iniparib (i.v. 2 days/week) in the adjuvant setting. Iniparib was dose escalated using a modified continual reassessment method (mCRM). 43 patients (32 male; 34 GBM, 8 AA, 1 gliosarcoma; median age 54 years; median KPS 90) were enrolled across 4 dose levels. In the mTMZ group, 2/4 patients had dose limiting toxicities (DLT) at 19 mg/kg/week (rash/hypersensitivity). At 17.2 mg/kg/week, 1/9 patients had a DLT (grade 3 fatigue). Additional grade 3 toxicities were neutropenia, lymphopenia, and nausea. In the cTMZ group, one DLT (thromboembolic event) occurred at 10.2 mg/kg/week. Dose escalation stopped at 16 mg/kg/week based on mCRM. The mean maximum plasma concentration of iniparib increased with dose. Concentration of the two major circulating metabolites, 4-iodo-3-aminobenzamide and 4-iodo-3-aminobenzoic acid, was ≤5% of the corresponding iniparib concentration. Iniparib is well tolerated, at doses higher than previously investigated, in combination with TMZ after completion of RT + TMZ in patients with MG. Recommended phase 2 dosing of iniparib based on mCRM is 17.2 mg/kg/week with mTMZ and 16 mg/kg/week with cTMZ. An efficacy study of TMZ/RT + iniparib followed by TMZ + iniparib in newly diagnosed GBM using these doses has completed enrollment. Survival assessment is ongoing.


Subject(s)
Antineoplastic Agents, Alkylating/administration & dosage , Benzamides/therapeutic use , Brain Neoplasms/drug therapy , Dacarbazine/analogs & derivatives , Glioma/drug therapy , Poly(ADP-ribose) Polymerase Inhibitors/therapeutic use , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols , Brain/drug effects , Brain/pathology , Dacarbazine/administration & dosage , Dose-Response Relationship, Drug , Drug Administration Schedule , Female , Humans , Magnetic Resonance Imaging , Male , Maximum Tolerated Dose , Middle Aged , Temozolomide , Treatment Outcome
17.
Skeletal Radiol ; 44(12): 1727-33, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26260535

ABSTRACT

PURPOSE: To evaluate gluteus medius and minimus tendon pathology and muscle atrophy in older individuals using MRI. METHODS: A retrospective MRI study of 185 individuals was performed. The inclusion criterion was age ≥50. Exclusion criteria were hip surgery, fracture, infection, tumor, or inadequate image quality. Greater trochanteric bursitis was graded none, mild, moderate, or severe. Gluteus medius, gluteus minimus, and iliopsoas tendinopathy was graded normal, tendinosis, low-grade partial tear, high-grade partial tear, or full thickness tear. Gluteus medius, gluteus minimus, tensor fascia lata, and iliopsoas muscle atrophy was scored using a standard scale. Insertion site of tendinopathy and location of muscle atrophy were assessed. Descriptive and statistical analysis was performed. RESULTS: There was increasing greater trochanteric bursitis and gluteus medius and minimus tendinopathy and atrophy with advancing age with moderate to strong positive associations (p < 0.0001) for age and tendinopathy, age and atrophy, bursitis and tendinopathy, and tendinopathy and atrophy for the gluteus medius and minimus. There is a weak positive association (p < 0.0001) for age and tensor fascia lata atrophy, and no statistically significant association between age and tendinopathy or between age and atrophy for the iliopsoas. Fisher's exact tests were statistically significant (p < 0.0001) for insertion site of tendon pathology and location of muscle atrophy for the gluteus medius. CONCLUSIONS: Gluteus medius and minimus tendon pathology and muscle atrophy increase with advancing age with progression of tendinosis to low-grade tendon tears to high-grade tendon tears. There is an associated progression in atrophy of these muscles, which may be important in fall-related hip fractures.


Subject(s)
Aging/pathology , Magnetic Resonance Imaging/methods , Muscular Atrophy/epidemiology , Muscular Atrophy/pathology , Tendinopathy/epidemiology , Tendinopathy/pathology , Age Distribution , Aged , Aged, 80 and over , Buttocks/pathology , Female , Humans , Magnetic Resonance Imaging/statistics & numerical data , Male , Middle Aged , Pennsylvania/epidemiology , Prevalence , Reproducibility of Results , Risk Factors , Sensitivity and Specificity
19.
Clin Cancer Res ; 30(4): 680-686, 2024 02 16.
Article in English | MEDLINE | ID: mdl-38048045

ABSTRACT

PURPOSE: There are no effective medical therapies for patients with meningioma who progress beyond surgical and radiotherapeutic interventions. Somatostatin receptor type 2 (SSTR2) represents a promising treatment target in meningiomas. In this multicenter, single-arm phase II clinical study (NCT03971461), the SSTR2-targeting radiopharmaceutical 177Lu-DOTATATE is evaluated for its feasibility, safety, and therapeutic efficacy in these patients. PATIENTS AND METHODS: Adult patients with progressive intracranial meningiomas received 177Lu-DOTATATE at a dose of 7.4 GBq (200 mCi) every eight weeks for four cycles. 68Ga-DOTATATE PET-MRI was performed before and six months after the start of the treatment. The primary endpoint was progression-free survival (PFS) at 6 months (PFS-6). Secondary endpoints were safety and tolerability, overall survival (OS) at 12 months (OS-12), median PFS, and median OS. RESULTS: Fourteen patients (female = 11, male = 3) with progressive meningiomas (WHO 1 = 3, 2 = 10, 3 = 1) were enrolled. Median age was 63.1 (range 49.7-78) years. All patients previously underwent tumor resection and at least one course of radiation. Treatment with 177Lu-DOTATATE was well tolerated. Seven patients (50%) achieved PFS-6. Best radiographic response by modified Macdonald criteria was stable disease (SD) in all seven patients. A >25% reduction in 68Ga-DOTATATE uptake (PET) was observed in five meningiomas and two patients. In one lesion, this corresponded to >50% reduction in bidirectional tumor measurements (MRI). CONCLUSIONS: Treatment with 177Lu-DOTATATE was well tolerated. The predefined PFS-6 threshold was met in this interim analysis, thereby allowing this multicenter clinical trial to continue enrollment. 68Ga-DOTATATE PET may be a useful imaging biomarker to assess therapeutic outcome in patients with meningioma.


Subject(s)
Meningeal Neoplasms , Meningioma , Neuroendocrine Tumors , Octreotide/analogs & derivatives , Organometallic Compounds , Receptors, Somatostatin , Adult , Humans , Male , Female , Middle Aged , Aged , Meningioma/diagnostic imaging , Meningioma/radiotherapy , Meningioma/drug therapy , Radiopharmaceuticals , Organometallic Compounds/adverse effects , Positron-Emission Tomography/methods , Meningeal Neoplasms/diagnostic imaging , Meningeal Neoplasms/radiotherapy , Meningeal Neoplasms/drug therapy , Biomarkers , Neuroendocrine Tumors/pathology , Positron Emission Tomography Computed Tomography
20.
Nat Commun ; 15(1): 2742, 2024 Mar 28.
Article in English | MEDLINE | ID: mdl-38548752

ABSTRACT

The epidermal growth factor receptor, EGFR, is frequently activated in lung cancer and glioblastoma by genomic alterations including missense mutations. The different mutation spectra in these diseases are reflected in divergent responses to EGFR inhibition: significant patient benefit in lung cancer, but limited in glioblastoma. Here, we report a comprehensive mutational analysis of EGFR function. We perform saturation mutagenesis of EGFR and assess function of ~22,500 variants in a human EGFR-dependent lung cancer cell line. This approach reveals enrichment of erlotinib-insensitive variants of known and unknown significance in the dimerization, transmembrane, and kinase domains. Multiple EGFR extracellular domain variants, not associated with approved targeted therapies, are sensitive to afatinib and dacomitinib in vitro. Two glioblastoma patients with somatic EGFR G598V dimerization domain mutations show responses to dacomitinib treatment followed by within-pathway resistance mutation in one case. In summary, this comprehensive screen expands the landscape of functional EGFR variants and suggests broader clinical investigation of EGFR inhibition for cancers harboring extracellular domain mutations.


Subject(s)
Glioblastoma , Lung Neoplasms , Humans , Glioblastoma/drug therapy , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , ErbB Receptors/metabolism , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Mutation
SELECTION OF CITATIONS
SEARCH DETAIL