51 results on '"Mclendon, Re"'
Search Results
2. A Modified Nucleoside 6-Thio-2'-Deoxyguanosine Exhibits Antitumor Activity in Gliomas.
- Author
-
Yu S, Wei S, Savani M, Lin X, Du K, Mender I, Siteni S, Vasilopoulos T, Reitman ZJ, Ku Y, Wu D, Liu H, Tian M, Chen Y, Labrie M, Charbonneau CM, Sugarman E, Bowie M, Hariharan S, Waitkus M, Jiang W, McLendon RE, Pan E, Khasraw M, Walsh KM, Lu Y, Herlyn M, Mills G, Herbig U, Wei Z, Keir ST, Flaherty K, Liu L, Wu K, Shay JW, Abdullah K, Zhang G, and Ashley DM
- Subjects
- Animals, Cell Line, Tumor, Deoxyguanosine analogs & derivatives, Humans, Mice, Nucleosides therapeutic use, Proteomics, Thionucleosides, Xenograft Model Antitumor Assays, Brain Neoplasms drug therapy, Brain Neoplasms genetics, Brain Neoplasms pathology, Glioma drug therapy, Glioma genetics, Glioma pathology
- Abstract
Purpose: To investigate the therapeutic role of a novel telomere-directed inhibitor, 6-thio-2'-deoxyguanosine (THIO) in gliomas both in vitro and in vivo ., Experimental Design: A panel of human and mouse glioma cell lines was used to test therapeutic efficacy of THIO using cell viability assays, flow cytometric analyses, and immunofluorescence. Integrated analyses of RNA sequencing and reverse-phase protein array data revealed the potential antitumor mechanisms of THIO. Four patient-derived xenografts (PDX), two patient-derived organoids (PDO), and two xenografts of human glioma cell lines were used to further investigate the therapeutic efficacy of THIO., Results: THIO was effective in the majority of human and mouse glioma cell lines with no obvious toxicity against normal astrocytes. THIO as a monotherapy demonstrated efficacy in three glioma cell lines that had acquired resistance to temozolomide. In addition, THIO showed efficacy in four human glioma cell lines grown as neurospheres by inducing apoptotic cell death. Mechanistically, THIO induced telomeric DNA damage not only in glioma cell lines but also in PDX tumor specimens. Integrated computational analyses of transcriptomic and proteomic data indicated that THIO significantly inhibited cell invasion, stem cell, and proliferation pathways while triggering DNA damage and apoptosis. Importantly, THIO significantly decreased tumor proliferation in two PDO models and reduced the tumor size of a glioblastoma xenograft and a PDX model., Conclusions: The current study established the therapeutic role of THIO in primary and recurrent gliomas and revealed the acute induction of telomeric DNA damage as a primary antitumor mechanism of THIO in gliomas., (©2021 American Association for Cancer Research.)
- Published
- 2021
- Full Text
- View/download PDF
3. Targeting PD-L1 Initiates Effective Antitumor Immunity in a Murine Model of Cushing Disease.
- Author
-
Kemeny HR, Elsamadicy AA, Farber SH, Champion CD, Lorrey SJ, Chongsathidkiet P, Woroniecka KI, Cui X, Shen SH, Rhodin KE, Tsvankin V, Everitt J, Sanchez-Perez L, Healy P, McLendon RE, Codd PJ, Dunn IF, and Fecci PE
- Subjects
- Adenoma drug therapy, Adenoma immunology, Adenoma pathology, Adolescent, Adult, Aged, Aged, 80 and over, Animals, Cell Line, Tumor, Disease Models, Animal, Female, Humans, Male, Mice, Mice, Inbred C57BL, Middle Aged, Pituitary ACTH Hypersecretion immunology, Pituitary ACTH Hypersecretion pathology, Pituitary Neoplasms immunology, Pituitary Neoplasms pathology, Survival Rate, Young Adult, Antibodies, Monoclonal pharmacology, B7-H1 Antigen antagonists & inhibitors, B7-H1 Antigen metabolism, Immunotherapy methods, Pituitary ACTH Hypersecretion drug therapy, Pituitary Neoplasms drug therapy, T-Lymphocytes immunology
- Abstract
Purpose: Although pituitary adenoma is classified as benign, Cushing disease is associated with significant morbidity due to the numerous sequelae of elevated cortisol levels. Successful therapy for Cushing disease remains elusive due to high rates of treatment-refractory recurrence. The frequent emergence of lymphocytic hypophysitis following checkpoint blockade for other cancers, as well as the expression of PD-L1 on pituitary adenomas, suggest a role for immunotherapy., Experimental Design: This study confirms PD-L1 expression on functioning pituitary adenomas and is the first to evaluate the efficacy of checkpoint blockade (anti-PD-L1) therapy in a preclinical model of Cushing disease., Results: Herein, treatment with anti-PD-L1 was successful in reducing adrenocorticotropic hormone plasma levels, decreasing tumor growth, and increasing survival in our model. Furthermore, tumor-infiltrating T cells demonstrated a pattern of checkpoint expression similar to other checkpoint blockade-susceptible tumors., Conclusions: This suggests that immunotherapy, particularly blockade of the PD1/PD-L1 axis, may be a novel therapeutic option for refractory Cushing disease. Clinical investigation is encouraged., (©2019 American Association for Cancer Research.)
- Published
- 2020
- Full Text
- View/download PDF
4. MTAP Loss Promotes Stemness in Glioblastoma and Confers Unique Susceptibility to Purine Starvation.
- Author
-
Hansen LJ, Sun R, Yang R, Singh SX, Chen LH, Pirozzi CJ, Moure CJ, Hemphill C, Carpenter AB, Healy P, Ruger RC, Chen CJ, Greer PK, Zhao F, Spasojevic I, Grenier C, Huang Z, Murphy SK, McLendon RE, Friedman HS, Friedman AH, Herndon JE 2nd, Sampson JH, Keir ST, Bigner DD, Yan H, and He Y
- Subjects
- Animals, Apoptosis, Biomarkers, Tumor genetics, Cell Proliferation, Glioblastoma genetics, Glioblastoma metabolism, Humans, Mice, Neoplastic Stem Cells metabolism, Prognosis, Purine-Nucleoside Phosphorylase genetics, Survival Rate, Tumor Cells, Cultured, Xenograft Model Antitumor Assays, Biomarkers, Tumor metabolism, Gene Expression Regulation, Neoplastic, Glioblastoma pathology, Neoplastic Stem Cells pathology, Purine-Nucleoside Phosphorylase metabolism, Purines metabolism
- Abstract
Homozygous deletion of methylthioadenosine phosphorylase ( MTAP ) is one of the most frequent genetic alterations in glioblastoma (GBM), but its pathologic consequences remain unclear. In this study, we report that loss of MTAP results in profound epigenetic reprogramming characterized by hypomethylation of PROM1 /CD133-associated stem cell regulatory pathways. MTAP deficiency promotes glioma stem-like cell (GSC) formation with increased expression of PROM1 /CD133 and enhanced tumorigenicity of GBM cells and is associated with poor prognosis in patients with GBM. As a combined consequence of purine production deficiency in MTAP -null GBM and the critical dependence of GSCs on purines, the enriched subset of CD133
+ cells in MTAP -null GBM can be effectively depleted by inhibition of de novo purine synthesis. These findings suggest that MTAP loss promotes the pathogenesis of GBM by shaping the epigenetic landscape and stemness of GBM cells while simultaneously providing a unique opportunity for GBM therapeutics. SIGNIFICANCE: This study links the frequently mutated metabolic enzyme MTAP to dysregulated epigenetics and cancer cell stemness and establishes MTAP status as a factor for consideration in characterizing GBM and developing therapeutic strategies., (©2019 American Association for Cancer Research.)- Published
- 2019
- Full Text
- View/download PDF
5. A Rationally Designed Fully Human EGFRvIII:CD3-Targeted Bispecific Antibody Redirects Human T Cells to Treat Patient-derived Intracerebral Malignant Glioma.
- Author
-
Gedeon PC, Schaller TH, Chitneni SK, Choi BD, Kuan CT, Suryadevara CM, Snyder DJ, Schmittling RJ, Szafranski SE, Cui X, Healy PN, Herndon JE 2nd, McLendon RE, Keir ST, Archer GE, Reap EA, Sanchez-Perez L, Bigner DD, and Sampson JH
- Subjects
- Animals, Antibodies, Bispecific pharmacology, CD3 Complex immunology, Cell Line, Tumor, ErbB Receptors immunology, Female, Gene Expression Regulation, Neoplastic immunology, Glioma immunology, Glioma pathology, Humans, Lymphocyte Activation drug effects, Lymphocyte Activation immunology, Male, Mice, Recombinant Proteins genetics, Recombinant Proteins pharmacology, Single-Chain Antibodies immunology, Single-Chain Antibodies pharmacology, T-Lymphocytes drug effects, T-Lymphocytes immunology, Xenograft Model Antitumor Assays, CD3 Complex antagonists & inhibitors, ErbB Receptors antagonists & inhibitors, Glioma drug therapy, Immunotherapy
- Abstract
Purpose: Conventional therapy for malignant glioma fails to specifically target tumor cells. In contrast, substantial evidence indicates that if appropriately redirected, T cells can precisely eradicate tumors. Here we report the rational development of a fully human bispecific antibody (hEGFRvIII-CD3 bi-scFv) that redirects human T cells to lyse malignant glioma expressing a tumor-specific mutation of the EGFR (EGFRvIII). Experimental Design: We generated a panel of bispecific single-chain variable fragments and optimized design through successive rounds of screening and refinement. We tested the ability of our lead construct to redirect naïve T cells and induce target cell-specific lysis. To test for efficacy, we evaluated tumor growth and survival in xenogeneic and syngeneic models of glioma. Tumor penetrance following intravenous drug administration was assessed in highly invasive, orthotopic glioma models. Results: A highly expressed bispecific antibody with specificity to CD3 and EGFRvIII was generated (hEGFRvIII-CD3 bi-scFv). Antibody-induced T-cell activation, secretion of proinflammatory cytokines, and proliferation was robust and occurred exclusively in the presence of target antigen. hEGFRvIII-CD3 bi-scFv was potent and target-specific, mediating significant lysis of multiple malignant glioma cell lines and patient-derived malignant glioma samples that heterogeneously express EGFRvIII. In both subcutaneous and orthotopic models, well-engrafted, patient-derived malignant glioma was effectively treated despite heterogeneity of EGFRvIII expression; intravenous hEGFRvIII-CD3 bi-scFv administration caused significant regression of tumor burden ( P < 0.0001) and significantly extended survival ( P < 0.0001). Similar efficacy was obtained in highly infiltrative, syngeneic glioma models, and intravenously administered hEGFRvIII-CD3 bi-scFv localized to these orthotopic tumors. Conclusions: We have developed a clinically translatable bispecific antibody that redirects human T cells to safely and effectively treat malignant glioma. On the basis of these results, we have developed a clinical study of hEGFRvIII-CD3 bi-scFv for patients with EGFRvIII-positive malignant glioma. Clin Cancer Res; 24(15); 3611-31. ©2018 AACR ., (©2018 American Association for Cancer Research.)
- Published
- 2018
- Full Text
- View/download PDF
6. Adaptive Evolution of the GDH2 Allosteric Domain Promotes Gliomagenesis by Resolving IDH1 R132H -Induced Metabolic Liabilities.
- Author
-
Waitkus MS, Pirozzi CJ, Moure CJ, Diplas BH, Hansen LJ, Carpenter AB, Yang R, Wang Z, Ingram BO, Karoly ED, Mohney RP, Spasojevic I, McLendon RE, Friedman HS, He Y, Bigner DD, and Yan H
- Subjects
- Animals, Brain Neoplasms metabolism, Brain Neoplasms mortality, Brain Neoplasms pathology, Evolution, Molecular, Gene Expression Regulation, Neoplastic, Gene Knock-In Techniques, Glioma metabolism, Glioma mortality, Glioma pathology, Glutamate Dehydrogenase chemistry, Glutamate Dehydrogenase genetics, Humans, Isocitrate Dehydrogenase genetics, Male, Mice, Inbred NOD, Mice, Inbred Strains, Mutagenesis, Site-Directed, Prosencephalon embryology, Protein Domains, Transgenes, Brain Neoplasms genetics, Glioma genetics, Glutamate Dehydrogenase metabolism, Isocitrate Dehydrogenase metabolism
- Abstract
Hotspot mutations in the isocitrate dehydrogenase 1 ( IDH1 ) gene occur in a number of human cancers and confer a neomorphic enzyme activity that catalyzes the conversion of α-ketoglutarate (αKG) to the oncometabolite D-(2)-hydroxyglutarate (D2HG). In malignant gliomas, IDH1
R132H expression induces widespread metabolic reprogramming, possibly requiring compensatory mechanisms to sustain the normal biosynthetic requirements of actively proliferating tumor cells. We used genetically engineered mouse models of glioma and quantitative metabolomics to investigate IDH1R132H -dependent metabolic reprogramming and its potential to induce biosynthetic liabilities that can be exploited for glioma therapy. In gliomagenic neural progenitor cells, IDH1R132H expression increased the abundance of dipeptide metabolites, depleted key tricarboxylic acid cycle metabolites, and slowed progression of murine gliomas. Notably, expression of glutamate dehydrogenase GDH2, a hominoid-specific enzyme with relatively restricted expression to the brain, was critically involved in compensating for IDH1R132H -induced metabolic alterations and promoting IDH1R132H glioma growth. Indeed, we found that recently evolved amino acid substitutions in the GDH2 allosteric domain conferred its nonredundant, glioma-promoting properties in the presence of IDH1 mutation. Our results indicate that among the unique roles for GDH2 in the human forebrain is its ability to limit IDH1R132H -mediated metabolic liabilities, thus promoting glioma growth in this context. Results from this study raise the possibility that GDH2-specific inhibition may be a viable therapeutic strategy for gliomas with IDH mutations. Significance: These findings show that the homonid-specific brain enzyme GDH2 may be essential to mitigate metabolic liabilities created by IDH1 mutations in glioma, with possible implications to leverage its therapeutic management by IDH1 inhibitors. Cancer Res; 78(1); 36-50. ©2017 AACR ., (©2017 American Association for Cancer Research.)- Published
- 2018
- Full Text
- View/download PDF
7. Dendritic Cells Enhance Polyfunctionality of Adoptively Transferred T Cells That Target Cytomegalovirus in Glioblastoma.
- Author
-
Reap EA, Suryadevara CM, Batich KA, Sanchez-Perez L, Archer GE, Schmittling RJ, Norberg PK, Herndon JE 2nd, Healy P, Congdon KL, Gedeon PC, Campbell OC, Swartz AM, Riccione KA, Yi JS, Hossain-Ibrahim MK, Saraswathula A, Nair SK, Dunn-Pirio AM, Broome TM, Weinhold KJ, Desjardins A, Vlahovic G, McLendon RE, Friedman AH, Friedman HS, Bigner DD, Fecci PE, Mitchell DA, and Sampson JH
- Subjects
- Adoptive Transfer, Adult, Aged, CD8-Positive T-Lymphocytes immunology, Cytomegalovirus, Dendritic Cells metabolism, Female, Humans, Male, Middle Aged, Phosphoproteins metabolism, T-Lymphocytes transplantation, Treatment Outcome, Viral Matrix Proteins metabolism, Brain Neoplasms therapy, Dendritic Cells immunology, Glioblastoma therapy, Immunotherapy, Adoptive methods, T-Lymphocytes immunology
- Abstract
Median survival for glioblastoma (GBM) remains <15 months. Human cytomegalovirus (CMV) antigens have been identified in GBM but not normal brain, providing an unparalleled opportunity to subvert CMV antigens as tumor-specific immunotherapy targets. A recent trial in recurrent GBM patients demonstrated the potential clinical benefit of adoptive T-cell therapy (ATCT) of CMV phosphoprotein 65 (pp65)-specific T cells. However, ex vivo analyses from this study found no change in the capacity of CMV pp65-specific T cells to gain multiple effector functions or polyfunctionality, which has been associated with superior antitumor efficacy. Previous studies have shown that dendritic cells (DC) could further enhance tumor-specific CD8
+ T-cell polyfunctionality in vivo when administered as a vaccine. Therefore, we hypothesized that vaccination with CMV pp65 RNA-loaded DCs would enhance the frequency of polyfunctional CMV pp65-specific CD8+ T cells after ATCT. Here, we report prospective results of a pilot trial in which 22 patients with newly diagnosed GBM were initially enrolled, of which 17 patients were randomized to receive CMV pp65-specific T cells with CMV-DC vaccination (CMV-ATCT-DC) or saline (CMV-ATCT-saline). Patients who received CMV-ATCT-DC vaccination experienced a significant increase in the overall frequencies of IFNγ+ , TNFα+ , and CCL3+ polyfunctional, CMV-specific CD8+ T cells. These increases in polyfunctional CMV-specific CD8+ T cells correlated ( R = 0.7371, P = 0.0369) with overall survival, although we cannot conclude this was causally related. Our data implicate polyfunctional T-cell responses as a potential biomarker for effective antitumor immunotherapy and support a formal assessment of this combination approach in a larger randomized study. Significance: A randomized pilot trial in patients with GBM implicates polyfunctional T-cell responses as a biomarker for effective antitumor immunotherapy. Cancer Res; 78(1); 256-64. ©2017 AACR ., (©2017 American Association for Cancer Research.)- Published
- 2018
- Full Text
- View/download PDF
8. Cic Loss Promotes Gliomagenesis via Aberrant Neural Stem Cell Proliferation and Differentiation.
- Author
-
Yang R, Chen LH, Hansen LJ, Carpenter AB, Moure CJ, Liu H, Pirozzi CJ, Diplas BH, Waitkus MS, Greer PK, Zhu H, McLendon RE, Bigner DD, He Y, and Yan H
- Subjects
- Animals, Cell Line, Tumor, Cells, Cultured, Gene Expression Profiling methods, Humans, Mice, Inbred C57BL, Mice, Inbred NOD, Mice, Knockout, Mice, SCID, Mice, Transgenic, Oligodendroglioma pathology, Cell Differentiation genetics, Cell Proliferation genetics, Mutation, Neural Stem Cells metabolism, Oligodendroglioma genetics, Repressor Proteins genetics
- Abstract
Inactivating mutations in the transcriptional repression factor Capicua ( CIC ) occur in approximately 50% of human oligodendrogliomas, but mechanistic links to pathogenesis are unclear. To address this question, we generated Cic -deficient mice and human oligodendroglioma cell models. Genetic deficiency in mice resulted in a partially penetrant embryonic or perinatal lethal phenotype, with the production of an aberrant proliferative neural population in surviving animals. In vitro cultured neural stem cells derived from Cic conditional knockout mice bypassed an EGF requirement for proliferation and displayed a defect in their potential for oligodendrocyte differentiation. Cic is known to participate in gene suppression that can be relieved by EGFR signal, but we found that cic also activated expression of a broad range of EGFR-independent genes. In an orthotopic mouse model of glioma, we found that Cic loss potentiated the formation and reduced the latency in tumor development. Collectively, our results define an important role for Cic in regulating neural cell proliferation and lineage specification, and suggest mechanistic explanations for how CIC mutations may impact the pathogenesis and therapeutic targeting of oligodendroglioma. Cancer Res; 77(22); 6097-108. ©2017 AACR ., (©2017 American Association for Cancer Research.)
- Published
- 2017
- Full Text
- View/download PDF
9. Histone H3.3K27M Represses p16 to Accelerate Gliomagenesis in a Murine Model of DIPG.
- Author
-
Cordero FJ, Huang Z, Grenier C, He X, Hu G, McLendon RE, Murphy SK, Hashizume R, and Becher OJ
- Subjects
- Animals, Brain Stem Neoplasms genetics, Cell Line, Tumor, Cyclin-Dependent Kinase Inhibitor p16 genetics, Disease Models, Animal, Glioma genetics, Histones genetics, Mice, Mice, Inbred C57BL, Brain Stem Neoplasms metabolism, Brain Stem Neoplasms pathology, Cyclin-Dependent Kinase Inhibitor p16 metabolism, Glioma metabolism, Glioma pathology, Histones metabolism
- Abstract
Diffuse intrinsic pontine glioma (DIPG) is a highly aggressive pediatric brainstem tumor genetically distinguished from adult GBM by the high prevalence of the K27M mutation in the histone H3 variant H3.3 ( H3F3A ). This mutation reprograms the H3K27me3 epigenetic landscape of DIPG by inhibiting the H3K27-specific histone methyltransferase EZH2. This globally reduces H3K27me2/3, critical repressive marks responsible for cell fate decisions, and also causes focal gain of H3K27me3 throughout the epigenome. To date, the tumor-driving effects of H3.3K27M remain largely unknown. Here, it is demonstrated that H3.3K27M cooperates with PDGF-B in vivo, enhancing gliomagenesis and reducing survival of p53 wild-type (WT) and knockout murine models of DIPG. H3.3K27M expression drives increased proliferation of tumor-derived murine neurospheres, suggesting that cell-cycle deregulation contributes to increased malignancy in mutant tumors. RNA sequencing on tumor tissue from H3.3K27M-expressing mice indicated global upregulation of PRC2 target genes, and a subset of newly repressed genes enriched in regulators of development and cell proliferation. Strikingly, H3.3K27M induced targeted repression of the p16/ink4a ( CDKN2A ) locus, a critical regulator of the G
0 -G1 to S-phase transition. Increased levels of H3K27me3 were observed at the p16 promoter; however, pharmacologic reduction of methylation at this promoter did not rescue p16 expression. Although DNA methylation is also present at this promoter, it is not K27M dependent. Intriguingly, inhibition of DNA methylation restores p16 levels and is cytotoxic against murine tumor cells. Importantly, these data reveal that H3.3K27M-mediated p16 repression is an important mechanism underlying the proliferation of H3.3K27M tumor cells, as in vivo cdkn2a knockout eliminates the survival difference between H3.3K27M and H3.3WT tumor-bearing mice. Implications: This study shows that H3.3K27M mutation and PDGF signaling act in concert to accelerate gliomagenesis in a genetic mouse model and identifies repression of p16 tumor suppressor as a target of H3.3K27M, highlighting the G1 -S cell-cycle transition as a promising therapeutic avenue. Mol Cancer Res; 15(9); 1243-54. ©2017 AACR ., (©2017 American Association for Cancer Research.)- Published
- 2017
- Full Text
- View/download PDF
10. Mutant IDH1 Disrupts the Mouse Subventricular Zone and Alters Brain Tumor Progression.
- Author
-
Pirozzi CJ, Carpenter AB, Waitkus MS, Wang CY, Zhu H, Hansen LJ, Chen LH, Greer PK, Feng J, Wang Y, Bock CB, Fan P, Spasojevic I, McLendon RE, Bigner DD, He Y, and Yan H
- Subjects
- Animals, Brain Neoplasms genetics, Cell Differentiation, Cell Proliferation, Cells, Cultured, DNA Methylation, Gene Knock-In Techniques, Humans, Lateral Ventricles pathology, Mice, Mice, Transgenic, Neural Stem Cells cytology, Neural Stem Cells pathology, Oligodendrocyte Transcription Factor 2 genetics, Promoter Regions, Genetic, Tumor Microenvironment, Brain Neoplasms pathology, Isocitrate Dehydrogenase genetics, Lateral Ventricles cytology, Mutation
- Abstract
IDH1 mutations occur in the majority of low-grade gliomas and lead to the production of the oncometabolite, D-2-hydroxyglutarate (D-2HG). To understand the effects of tumor-associated mutant IDH1 (IDH1-R132H) on both the neural stem cell (NSC) population and brain tumorigenesis, genetically faithful cell lines and mouse model systems were generated. Here, it is reported that mouse NSCs expressing Idh1-R132H displayed reduced proliferation due to p53-mediated cell-cycle arrest as well as a decreased ability to undergo neuronal differentiation. In vivo , Idh1-R132H expression reduced proliferation of cells within the germinal zone of the subventricular zone (SVZ). The NSCs within this area were dispersed and disorganized in mutant animals, suggesting that Idh1-R132H perturbed the NSCs and the microenvironment from which gliomas arise. In addition, tumor-bearing animals expressing mutant Idh1 displayed a prolonged survival and also overexpressed Olig2, features consistent with IDH1-mutated human gliomas. These data indicate that mutant Idh1 disrupts the NSC microenvironment and the candidate cell-of-origin for glioma; thus, altering the progression of tumorigenesis. In addition, this study provides a mutant Idh1 brain tumor model that genetically recapitulates human disease, laying the foundation for future investigations on mutant IDH1 -mediated brain tumorigenesis and targeted therapy. Implications: Through the use of a conditional mutant mouse model that confers a less aggressive tumor phenotype, this study reveals that mutant Idh1 impacts the candidate cell-of-origin for gliomas. Mol Cancer Res; 15(5); 507-20. ©2017 AACR ., (©2017 American Association for Cancer Research.)
- Published
- 2017
- Full Text
- View/download PDF
11. Long-term Survival in Glioblastoma with Cytomegalovirus pp65-Targeted Vaccination.
- Author
-
Batich KA, Reap EA, Archer GE, Sanchez-Perez L, Nair SK, Schmittling RJ, Norberg P, Xie W, Herndon JE 2nd, Healy P, McLendon RE, Friedman AH, Friedman HS, Bigner D, Vlahovic G, Mitchell DA, and Sampson JH
- Subjects
- Adjuvants, Immunologic, Aged, Brain Neoplasms immunology, Brain Neoplasms mortality, Combined Modality Therapy, Dacarbazine administration & dosage, Dacarbazine analogs & derivatives, Dendritic Cells immunology, Disease-Free Survival, Female, Glioblastoma immunology, Glioblastoma mortality, Granulocyte-Macrophage Colony-Stimulating Factor, Humans, Kaplan-Meier Estimate, Male, Middle Aged, Phosphoproteins immunology, T-Lymphocytes, Regulatory immunology, Temozolomide, Viral Matrix Proteins immunology, Antineoplastic Agents therapeutic use, Brain Neoplasms therapy, Cancer Vaccines therapeutic use, Dendritic Cells transplantation, Glioblastoma therapy, Phosphoproteins therapeutic use, Viral Matrix Proteins therapeutic use
- Abstract
Purpose: Patients with glioblastoma have less than 15-month median survival despite surgical resection, high-dose radiation, and chemotherapy with temozolomide. We previously demonstrated that targeting cytomegalovirus pp65 using dendritic cells (DC) can extend survival and, in a separate study, that dose-intensified temozolomide (DI-TMZ) and adjuvant granulocyte macrophage colony-stimulating factor (GM-CSF) potentiate tumor-specific immune responses in patients with glioblastoma. Here, we evaluated pp65-specific cellular responses following DI-TMZ with pp65-DCs and determined the effects on long-term progression-free survival (PFS) and overall survival (OS). Experimental Design: Following standard-of-care, 11 patients with newly diagnosed glioblastoma received DI-TMZ (100 mg/m
2 /d × 21 days per cycle) with at least three vaccines of pp65 lysosome-associated membrane glycoprotein mRNA-pulsed DCs admixed with GM-CSF on day 23 ± 1 of each cycle. Thereafter, monthly DI-TMZ cycles and pp65-DCs were continued if patients had not progressed. Results: Following DI-TMZ cycle 1 and three doses of pp65-DCs, pp65 cellular responses significantly increased. After DI-TMZ, both the proportion and proliferation of regulatory T cells (Tregs) increased and remained elevated with serial DI-TMZ cycles. Median PFS and OS were 25.3 months [95% confidence interval (CI), 11.0-∞] and 41.1 months (95% CI, 21.6-∞), exceeding survival using recursive partitioning analysis and matched historical controls. Four patients remained progression-free at 59 to 64 months from diagnosis. No known prognostic factors [age, Karnofsky performance status (KPS), IDH-1/2 mutation, and MGMT promoter methylation] predicted more favorable outcomes for the patients in this cohort. Conclusions: Despite increased Treg proportions following DI-TMZ, patients receiving pp65-DCs showed long-term PFS and OS, confirming prior studies targeting cytomegalovirus in glioblastoma. Clin Cancer Res; 23(8); 1898-909. ©2017 AACR ., (©2017 American Association for Cancer Research.)- Published
- 2017
- Full Text
- View/download PDF
12. A Three-Dimensional Organoid Culture System Derived from Human Glioblastomas Recapitulates the Hypoxic Gradients and Cancer Stem Cell Heterogeneity of Tumors Found In Vivo.
- Author
-
Hubert CG, Rivera M, Spangler LC, Wu Q, Mack SC, Prager BC, Couce M, McLendon RE, Sloan AE, and Rich JN
- Subjects
- Animals, Heterografts, Humans, Mice, Brain Neoplasms pathology, Cell Hypoxia, Glioblastoma pathology, Neoplastic Stem Cells pathology, Organoids pathology
- Abstract
Many cancers feature cellular hierarchies that are driven by tumor-initiating cancer stem cells (CSC) and rely on complex interactions with the tumor microenvironment. Standard cell culture conditions fail to recapitulate the original tumor architecture or microenvironmental gradients and are not designed to retain the cellular heterogeneity of parental tumors. Here, we describe a three-dimensional culture system that supports the long-term growth and expansion of tumor organoids derived directly from glioblastoma specimens, including patient-derived primary cultures, xenografts, genetically engineered glioma models, or patient samples. Organoids derived from multiple regions of patient tumors retain selective tumorigenic potential. Furthermore, organoids could be established directly from brain metastases not typically amenable to in vitro culture. Once formed, tumor organoids grew for months and displayed regional heterogeneity with a rapidly dividing outer region of SOX2(+), OLIG2(+), and TLX(+) cells surrounding a hypoxic core of primarily non-stem senescent cells and diffuse, quiescent CSCs. Notably, non-stem cells within organoids were sensitive to radiotherapy, whereas adjacent CSCs were radioresistant. Orthotopic transplantation of patient-derived organoids resulted in tumors displaying histologic features, including single-cell invasiveness, that were more representative of the parental tumor compared with those formed from patient-derived sphere cultures. In conclusion, we present a new ex vivo model in which phenotypically diverse stem and non-stem glioblastoma cell populations can be simultaneously cultured to explore new facets of microenvironmental influences and CSC biology. Cancer Res; 76(8); 2465-77. ©2016 AACR., (©2016 American Association for Cancer Research.)
- Published
- 2016
- Full Text
- View/download PDF
13. Differential Immune Microenvironments and Response to Immune Checkpoint Blockade among Molecular Subtypes of Murine Medulloblastoma.
- Author
-
Pham CD, Flores C, Yang C, Pinheiro EM, Yearley JH, Sayour EJ, Pei Y, Moore C, McLendon RE, Huang J, Sampson JH, Wechsler-Reya R, and Mitchell DA
- Subjects
- Animals, Biomarkers, Disease Models, Animal, Immunophenotyping, Lymphocyte Count, Lymphocytes, Tumor-Infiltrating immunology, Lymphocytes, Tumor-Infiltrating metabolism, Lymphocytes, Tumor-Infiltrating pathology, Medulloblastoma drug therapy, Medulloblastoma genetics, Medulloblastoma mortality, Mice, Mice, Knockout, Myeloid Cells immunology, Myeloid Cells metabolism, Myeloid Cells pathology, Patched Receptors, Phenotype, Receptors, Cell Surface deficiency, Receptors, Cell Surface genetics, T-Lymphocyte Subsets immunology, T-Lymphocyte Subsets metabolism, T-Lymphocyte Subsets pathology, Tumor Microenvironment genetics, Antineoplastic Agents pharmacology, Medulloblastoma immunology, Medulloblastoma pathology, Programmed Cell Death 1 Receptor antagonists & inhibitors, Tumor Microenvironment drug effects, Tumor Microenvironment immunology
- Abstract
Purpose: Despite significant strides in the identification and characterization of potential therapeutic targets for medulloblastoma, the role of the immune system and its interplay with the tumor microenvironment within these tumors are poorly understood. To address this, we adapted two syngeneic animal models of human Sonic Hedgehog (SHH)-driven and group 3 medulloblastoma for preclinical evaluation in immunocompetent C57BL/6 mice., Experimental Design and Results: Multicolor flow cytometric analyses were used to phenotype and characterize immune infiltrating cells within established cerebellar tumors. We observed significantly higher percentages of dendritic cells, infiltrating lymphocytes, myeloid-derived suppressor cells, and tumor-associated macrophages in murine SHH model tumors compared with group 3 tumors. However, murine group 3 tumors had higher percentages of CD8(+) PD-1(+) T cells within the CD3 population. PD-1 blockade conferred superior antitumor efficacy in animals bearing intracranial group 3 tumors compared with SHH group tumors, indicating that immunologic differences within the tumor microenvironment can be leveraged as potential targets to mediate antitumor efficacy. Further analysis of anti-PD-1 monoclonal antibody localization revealed binding to PD-1(+) peripheral T cells, but not tumor infiltrating lymphocytes within the brain tumor microenvironment. Peripheral PD-1 blockade additionally resulted in a marked increase in CD3(+) T cells within the tumor microenvironment., Conclusions: This is the first immunologic characterization of preclinical models of molecular subtypes of medulloblastoma and demonstration that response to immune checkpoint blockade differs across subtype classification. Our findings also suggest that effective anti-PD-1 blockade does not require that systemically administered antibodies penetrate the brain tumor microenvironment., (©2015 American Association for Cancer Research.)
- Published
- 2016
- Full Text
- View/download PDF
14. Tumor-infiltrating lymphocytes in glioblastoma are associated with specific genomic alterations and related to transcriptional class.
- Author
-
Rutledge WC, Kong J, Gao J, Gutman DA, Cooper LA, Appin C, Park Y, Scarpace L, Mikkelsen T, Cohen ML, Aldape KD, McLendon RE, Lehman NL, Miller CR, Schniederjan MJ, Brennan CW, Saltz JH, Moreno CS, and Brat DJ
- Subjects
- Brain Neoplasms genetics, Brain Neoplasms mortality, Brain Neoplasms pathology, Gene Expression Profiling, Glioblastoma mortality, Glioblastoma pathology, Humans, Oligonucleotide Array Sequence Analysis, Prognosis, Survival Rate, Biomarkers, Tumor genetics, DNA Methylation, Gene Dosage, Glioblastoma genetics, Lymphocytes, Tumor-Infiltrating pathology, Mutation genetics
- Abstract
Purpose: Tumor-infiltrating lymphocytes (TIL) have prognostic significance in many cancers, yet their roles in glioblastoma have not been fully defined. We hypothesized that TILs in glioblastoma are associated with molecular alterations, histologies, and survival., Experimental Design: We used data from The Cancer Genome Atlas (TCGA) to investigate molecular, histologic, and clinical correlates of TILs in glioblastomas. Lymphocytes were categorized as absent, present, or abundant in histopathologic images from 171 TCGA glioblastomas. Associations were examined between lymphocytes and histologic features, mutations, copy number alterations, CpG island methylator phenotype, transcriptional class, and survival. We validated histologic findings using CD3G gene expression., Results: We found a positive correlation between TILs and glioblastomas with gemistocytes, sarcomatous cells, epithelioid cells, and giant cells. Lymphocytes were enriched in the mesenchymal transcriptional class and strongly associated with mutations in NF1 and RB1. These mutations are frequent in the mesenchymal class and characteristic of gemistocytic, sarcomatous, epithelioid, and giant cell histologies. Conversely, TILs were rare in glioblastomas with small cells and oligodendroglioma components. Lymphocytes were depleted in the classical transcriptional class and in EGF receptor (EGFR)-amplified and homozygous PTEN-deleted glioblastomas. These alterations are characteristic of glioblastomas with small cells and glioblastomas of the classical transcriptional class. No association with survival was shown., Conclusions: TILs were enriched in glioblastomas of the mesenchymal class, strongly associated with mutations in NF1 and RB1 and typical of histologies characterized by these mutations. Conversely, TILs were depleted in the classical class, EGFR-amplified, and homozygous PTEN-deleted tumors and rare in histologies characterized by these alterations., (©2013 AACR.)
- Published
- 2013
- Full Text
- View/download PDF
15. Construction of an immunotoxin, D2C7-(scdsFv)-PE38KDEL, targeting EGFRwt and EGFRvIII for brain tumor therapy.
- Author
-
Chandramohan V, Bao X, Keir ST, Pegram CN, Szafranski SE, Piao H, Wikstrand CJ, McLendon RE, Kuan CT, Pastan IH, and Bigner DD
- Subjects
- Antigens, Neoplasm immunology, Brain Neoplasms pathology, Brain Neoplasms therapy, Cell Line, Tumor, Epitopes isolation & purification, ErbB Receptors antagonists & inhibitors, ErbB Receptors genetics, Flow Cytometry, Glioblastoma pathology, Glioblastoma therapy, Humans, Immunoglobulin G genetics, Immunoglobulin G immunology, Immunotoxins genetics, Surface Plasmon Resonance, Xenograft Model Antitumor Assays, Brain Neoplasms immunology, ErbB Receptors immunology, Glioblastoma immunology, Immunotoxins administration & dosage
- Abstract
Purpose: The EGF receptor gene (EGFR) is most frequently amplified and overexpressed, along with its deletion mutant, EGFRvIII, in glioblastoma. We tested the preclinical efficacy of the recombinant immunotoxin, D2C7-(scdsFv)-PE38KDEL, which is reactive with a 55-amino acid (AA) region present in the extracellular domain of both EGFRwt (583-637 AAs) and EGFRvIII (292-346 AAs) proteins., Experimental Design: The binding affinity and specificity of D2C7-(scdsFv)-PE38KDEL for EGFRwt and EGFRvIII were measured by surface-plasmon resonance and flow cytometry. In vitro cytotoxicity of D2C7-(scdsFv)-PE38KDEL was measured by inhibition of protein synthesis in human EGFRwt-transfected NR6 (NR6W), human EGFRvIII-transfected NR6 (NR6M), EGFRwt-overexpressing A431-epidermoid-carcinoma, and glioblastoma xenograft cells (43, D08-0493MG, D2159MG, and D270MG). In vivo antitumor efficacy of D2C7-(scdsFv)-PE38KDEL was evaluated using 43, NR6M, and D270MG orthotopic tumor models., Results: The KD of D2C7-(scdsFv)-PE38KDEL for EGFRwt and EGFRvIII was 1.6×10(-9) mol/L and 1.3×10(-9) mol/L, respectively. Flow cytometry with NR6W and NR6M cells confirmed the specificity of D2C7-(scdsFv)-PE38KDEL for EGFRwt and EGFRvIII. The D2C7-(scdsFv)-PE38KDEL IC50 was 0.18 to 2.5 ng/mL on cells expressing EGFRwt (NR6W, A431, 43, and D08-0493MG). The D2C7-(scdsFv)-PE38KDEL IC50 was approximately 0.25 ng/mL on EGFRvIII-expressing cells (NR6M) and on EGFRwt- and EGFRvIII-expressing glioblastoma xenograft cells (D2159MG and D270MG). Significantly, in intracranial tumor models of 43, NR6M, and D270MG, treatment with D2C7-(scdsFv)-PE38KDEL by convection-enhanced delivery prolonged survival by 310% (P=0.006), 28% (P=0.002), and 166% (P=0.001), respectively., Conclusions: In preclinical studies, the D2C7-(scdsFv)-PE38KDEL immunotoxin exhibited significant potential for treating brain tumors expressing EGFRwt, EGFRvIII, or both., (©2013 AACR.)
- Published
- 2013
- Full Text
- View/download PDF
16. A phase I/II trial of pazopanib in combination with lapatinib in adult patients with relapsed malignant glioma.
- Author
-
Reardon DA, Groves MD, Wen PY, Nabors L, Mikkelsen T, Rosenfeld S, Raizer J, Barriuso J, McLendon RE, Suttle AB, Ma B, Curtis CM, Dar MM, and de Bono J
- Subjects
- Adult, Angiogenesis Inhibitors administration & dosage, Anticonvulsants administration & dosage, Anticonvulsants pharmacokinetics, Antineoplastic Agents pharmacokinetics, Antineoplastic Combined Chemotherapy Protocols, Brain Neoplasms pathology, Disease-Free Survival, Drug Delivery Systems, ErbB Receptors metabolism, Glioma pathology, Humans, Indazoles, Lapatinib, Neoplasm Recurrence, Local drug therapy, Neoplasm Recurrence, Local pathology, Neoplasm Staging, Pyrimidines pharmacokinetics, Quinazolines pharmacokinetics, Recurrence, Sulfonamides pharmacokinetics, Antineoplastic Agents administration & dosage, Brain Neoplasms drug therapy, Glioma drug therapy, Pyrimidines administration & dosage, Quinazolines administration & dosage, Sulfonamides administration & dosage
- Abstract
Purpose: Increased mitogenic signaling and angiogenesis, frequently facilitated by somatic activation of EGF receptor (EGFR; ErbB1) and/or loss of PTEN, and VEGF overexpression, respectively, drive malignant glioma growth. We hypothesized that patients with recurrent glioblastoma would exhibit differential antitumor benefit based on tumor PTEN/EGFRvIII status when treated with the antiangiogenic agent pazopanib and the ErbB inhibitor lapatinib., Experimental Design: A phase II study evaluated the antitumor activity of pazopanib 400 mg/d plus lapatinib 1,000 mg/d in patients with grade 4 malignant glioma and known PTEN/EGFRvIII status not receiving enzyme-inducing anticonvulsants (EIAC). The phase II study used a two-stage Green-Dahlberg design for futility. An independent, parallel phase I component determined the maximum-tolerated regimen (MTR) of pazopanib and lapatinib in patients with grade 3/4 glioma receiving EIACs., Results: The six-month progression-free survival (PFS) rates in phase II (n = 41) were 0% and 15% in the PTEN/EGFRvIII-positive and PTEN/EGFRvIII-negative cohorts, respectively, leading to early termination. Two patients (5%) had a partial response and 14 patients (34%) had stable disease lasting 8 or more weeks. In phase I (n = 34), the MTR was not reached. On the basis of pharmacokinetic and safety review, a regimen of pazopanib 600 mg plus lapatinib 1,000 mg, each twice daily, was considered safe. Concomitant EIACs reduced exposure to pazopanib and lapatinib., Conclusions: The antitumor activity of this combination at the phase II dose tested was limited. Pharmacokinetic data indicated that exposure to lapatinib was subtherapeutic in the phase II evaluation. Evaluation of intratumoral drug delivery and activity may be essential for hypothesis-testing trials with targeted agents in malignant glioma., (©2012 AACR.)
- Published
- 2013
- Full Text
- View/download PDF
17. Disruption of wild-type IDH1 suppresses D-2-hydroxyglutarate production in IDH1-mutated gliomas.
- Author
-
Jin G, Reitman ZJ, Duncan CG, Spasojevic I, Gooden DM, Rasheed BA, Yang R, Lopez GY, He Y, McLendon RE, Bigner DD, and Yan H
- Subjects
- Astrocytoma genetics, Brain Neoplasms metabolism, Cell Line, Tumor, Genotype, Glioblastoma genetics, Glioma metabolism, Humans, Isocitrate Dehydrogenase metabolism, Brain Neoplasms genetics, Glioma genetics, Glutarates metabolism, Isocitrate Dehydrogenase genetics, Mutation
- Abstract
Point mutations at Arg132 of the cytoplasmic NADP(+)-dependent isocitrate dehydrogenase 1 (IDH1) occur frequently in gliomas and result in a gain of function to produce the "oncometabolite" D-2-hydroxyglutarate (D-2HG). The mutated IDH1 allele is usually associated with a wild-type IDH1 allele (heterozygous) in cancer. Here, we identify 2 gliomas that underwent loss of the wild-type IDH1 allele but retained the mutant IDH1 allele following tumor progression from World Health Organization (WHO) grade III anaplastic astrocytomas to WHO grade IV glioblastomas. Intratumoral D-2HG was 14-fold lower in the glioblastomas lacking wild-type IDH1 than in glioblastomas with heterozygous IDH1 mutations. To characterize the contribution of wild-type IDH1 to cancer cell D-2HG production, we established an IDH1-mutated astrocytoma (IMA) cell line from a WHO grade III anaplastic astrocytoma. Disruption of the wild-type IDH1 allele in IMA cells by gene targeting resulted in an 87-fold decrease in cellular D-2HG levels, showing that both wild-type and mutant IDH1 alleles are required for D-2HG production in glioma cells. Expression of wild-type IDH1 was also critical for mutant IDH1-associated D-2HG production in the colorectal cancer cell line HCT116. These insights may aid in the development of therapeutic strategies to target IDH1-mutated cancers.
- Published
- 2013
- Full Text
- View/download PDF
18. OTX2 is critical for the maintenance and progression of Shh-independent medulloblastomas.
- Author
-
Adamson DC, Shi Q, Wortham M, Northcott PA, Di C, Duncan CG, Li J, McLendon RE, Bigner DD, Taylor MD, and Yan H
- Subjects
- Animals, Blotting, Western, Cerebellar Neoplasms metabolism, Disease Progression, Gene Dosage, Genes, myc genetics, Hedgehog Proteins genetics, Humans, In Situ Hybridization, Fluorescence, Medulloblastoma metabolism, Mice, Mice, Nude, Oligonucleotide Array Sequence Analysis, Polymerase Chain Reaction, Polymorphism, Single Nucleotide, Cerebellar Neoplasms genetics, Gene Expression Regulation, Neoplastic, Medulloblastoma genetics, Otx Transcription Factors genetics
- Abstract
OTX2 is a developmentally regulated transcription factor involved in early morphogenesis of the central nervous system. This gene is amplified and overexpressed in medulloblastoma cell lines, but the nature and extent of its genetic alterations in primary tumors have not been evaluated. Analysis of a large cohort of primary medulloblastomas revealed frequent focal copy number gain of a region minimally containing OTX2 as a single gene. OTX2 copy number gain was restricted to tumor subtypes that did not express a molecular signature of Wnt or Shh pathway activation. FISH analysis revealed copy number gain in a subset of cells within medulloblastoma samples, suggesting a late event in tumor progression. Gain of OTX2 copy number was associated with the presence of anaplastic histologic features and shorter survival in medulloblastoma patients. In support of a functional role, ectopic OTX2 expression enhanced proliferation and tumorigenicity of immortalized primary cells, whereas OTX2 knockdown in medulloblastoma cells prolonged the survival of animals bearing xenograft tumors. Mechanistic investigations revealed upregulation of MYC as a potential mechanism whereby OTX2 promotes tumor progression. Our findings define OTX2 as an important oncogenic driver in medulloblastoma.
- Published
- 2010
- Full Text
- View/download PDF
19. Glioblastoma proto-oncogene SEC61gamma is required for tumor cell survival and response to endoplasmic reticulum stress.
- Author
-
Lu Z, Zhou L, Killela P, Rasheed AB, Di C, Poe WE, McLendon RE, Bigner DD, Nicchitta C, and Yan H
- Subjects
- Brain Neoplasms metabolism, Brain Neoplasms pathology, Cell Growth Processes genetics, Endoplasmic Reticulum metabolism, Endoplasmic Reticulum pathology, ErbB Receptors biosynthesis, ErbB Receptors genetics, Gene Amplification, Genes, erbB-1, Glioblastoma metabolism, Glioblastoma pathology, HeLa Cells, Humans, Membrane Proteins biosynthesis, Proto-Oncogene Mas, RNA, Small Interfering genetics, SEC Translocation Channels, Tunicamycin pharmacology, Brain Neoplasms genetics, Endoplasmic Reticulum genetics, Glioblastoma genetics, Membrane Proteins genetics
- Abstract
Glioblastoma multiforme is the most prevalent type of adult brain tumor and one of the deadliest tumors known to mankind. The genetic understanding of glioblastoma multiforme is, however, limited, and the molecular mechanisms that facilitate glioblastoma multiforme cell survival and growth within the tumor microenvironment are largely unknown. We applied digital karyotyping and single nucleotide polymorphism arrays to screen for copy-number changes in glioblastoma multiforme samples and found that the most frequently amplified region is at chromosome 7p11.2. The high resolution of digital karyotyping and single nucleotide polymorphism arrays permits the precise delineation of amplicon boundaries and has enabled identification of the minimal region of amplification at chromosome 7p11.2, which contains two genes, EGFR and SEC61gamma. SEC61gamma encodes a subunit of a heterotrimeric protein channel located in the endoplasmic reticulum (ER). In addition to its high frequency of gene amplification in glioblastoma multiforme, SEC61gamma is also remarkably overexpressed in 77% of glioblastoma multiforme but not in lower-grade gliomas. The small interfering RNA-mediated knockdown of SEC61gamma expression in tumor cells led to growth suppression and apoptosis. Furthermore, we showed that pharmacologic ER stress agents induce SEC61gamma expression in glioblastoma multiforme cells. Together, these results indicate that aberrant expression of SEC61gamma serves significant roles in glioblastoma multiforme cell survival likely via a mechanism that is involved in the cytoprotective ER stress-adaptive response to the tumor microenvironment.
- Published
- 2009
- Full Text
- View/download PDF
20. Phase II trial of Gliadel plus O6-benzylguanine in adults with recurrent glioblastoma multiforme.
- Author
-
Quinn JA, Jiang SX, Carter J, Reardon DA, Desjardins A, Vredenburgh JJ, Rich JN, Gururangan S, Friedman AH, Bigner DD, Sampson JH, McLendon RE, Herndon JE 2nd, Threatt S, and Friedman HS
- Subjects
- Adult, Aged, Antineoplastic Combined Chemotherapy Protocols adverse effects, Brain Neoplasms metabolism, Carmustine, Decanoic Acids adverse effects, Female, Glioblastoma metabolism, Guanine administration & dosage, Guanine adverse effects, Humans, Male, Middle Aged, O(6)-Methylguanine-DNA Methyltransferase metabolism, Polyesters adverse effects, Recurrence, Survival Analysis, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Brain Neoplasms drug therapy, Decanoic Acids administration & dosage, Glioblastoma drug therapy, Guanine analogs & derivatives, Polyesters administration & dosage
- Abstract
Purpose: This phase II trial was designed to define the efficacy of Gliadel wafers in combination with an infusion of O6-benzylguanine (O6-BG) that suppresses tumor O6-alkylguanine-DNA alkyltransferase (AGT) levels in patients with recurrent glioblastoma multiforme for 5 days and to evaluate the safety of this combination therapy., Experimental Design: This was a phase II, open-label, single center trial. On gross total resection of the tumor, up to eight Gliadel wafers were implanted. Bolus infusion of O6-BG was administered at 120 mg/m2 over 1 hour on days 1, 3, and 5, along with a continuous infusion at 30 mg/m2/d. The primary end points were 6-month overall survival (OS) and safety, and the secondary end points were 1-year, 2-year, and median OS., Results: Fifty-two patients were accrued. The 6-month OS was 82% [95% confidence interval (95% CI), 72-93%]. The 1- and 2-year OS rates were 47% (95% CI, 35-63%) and 10% (95% CI, 3-32%), respectively. The median OS was 50.3 weeks (95% CI, 36.1-69.4 weeks). Treatment-related toxicity with this drug combination included grade 3 hydrocephalus (9.6%), grade 3 cerebrospinal fluid (CSF) leak (19.2%), and grade 3 CSF/brain infection (13.4%)., Conclusion: The efficacy of implanted Gliadel wafers may be improved with the addition of O6-BG. Although systemically administered O6-BG can be coadministered with Gliadel wafers safely, it may increase the risk of hydrocephalus, CSF leak, and CSF/brain infection. Future trials are required to verify that inhibition of tumor AGT levels by O6-BG results in increased efficacy of Gliadel wafers without added toxicity.
- Published
- 2009
- Full Text
- View/download PDF
21. Mismatch repair deficiency does not mediate clinical resistance to temozolomide in malignant glioma.
- Author
-
Maxwell JA, Johnson SP, McLendon RE, Lister DW, Horne KS, Rasheed A, Quinn JA, Ali-Osman F, Friedman AH, Modrich PL, Bigner DD, and Friedman HS
- Subjects
- Adult, Aged, Aged, 80 and over, Dacarbazine pharmacology, Female, Glioblastoma genetics, Humans, Male, Middle Aged, O(6)-Methylguanine-DNA Methyltransferase genetics, Temozolomide, Antineoplastic Agents, Alkylating pharmacology, Base Pair Mismatch, Brain Neoplasms drug therapy, Brain Neoplasms genetics, DNA Mismatch Repair, Dacarbazine analogs & derivatives, Drug Resistance, Neoplasm, Glioma drug therapy, Glioma genetics
- Abstract
Purpose: A major mechanism of resistance to methylating agents, including temozolomide, is the DNA repair protein O(6)-alkylguanine-DNA alkyltransferase (AGT). Preclinical data indicates that defective DNA mismatch repair (MMR) results in tolerance to temozolomide regardless of AGT activity. The purpose of this study was to determine the role of MMR deficiency in mediating resistance in samples from patients with both newly diagnosed malignant gliomas and those who have failed temozolomide therapy., Experimental Design: The roles of AGT and MMR deficiency in mediating resistance in glioblastoma multiforme were assessed by immunohistochemistry and microsatellite instability (MSI), respectively. The mutation status of the MSH6 gene, a proposed correlate of temozolomide resistance, was determined by direct sequencing and compared with data from immunofluorescent detection of MSH6 protein and reverse transcription-PCR amplification of MSH6 RNA., Results: Seventy percent of newly diagnosed and 78% of failed-therapy glioblastoma multiforme samples expressed nuclear AGT protein in > or = 20% of cells analyzed, suggesting alternate means of resistance in 20% to 30% of cases. Single loci MSI was observed in 3% of patient samples; no sample showed the presence of high MSI. MSI was not shown to correlate with MSH6 mutation or loss of MSH6 protein expression., Conclusions: Although high AGT levels may mediate resistance in a portion of these samples, MMR deficiency does not seem to be responsible for mediating temozolomide resistance in adult malignant glioma. Accordingly, the presence of a fraction of samples exhibiting both low AGT expression and MMR proficiency suggests that additional mechanisms of temozolomide resistance are operational in the clinic.
- Published
- 2008
- Full Text
- View/download PDF
22. Targeting cancer stem cells through L1CAM suppresses glioma growth.
- Author
-
Bao S, Wu Q, Li Z, Sathornsumetee S, Wang H, McLendon RE, Hjelmeland AB, and Rich JN
- Subjects
- AC133 Antigen, Animals, Antigens, CD immunology, Blotting, Western, Brain Neoplasms immunology, Brain Neoplasms metabolism, Cell Division, Cell Separation, Cell Survival, Flow Cytometry, Fluorescent Antibody Technique, Glioma immunology, Glioma metabolism, Glycoproteins immunology, Humans, Mice, Mice, Nude, Neoplastic Stem Cells immunology, Neoplastic Stem Cells metabolism, Neural Cell Adhesion Molecule L1 genetics, Peptides immunology, Polymerase Chain Reaction, Brain Neoplasms pathology, Glioma pathology, Neoplastic Stem Cells pathology, Neural Cell Adhesion Molecule L1 metabolism
- Abstract
Malignant gliomas are lethal cancers that display striking cellular heterogeneity. A highly tumorigenic glioma tumor subpopulation, termed cancer stem cells or tumor-initiating cells, promotes therapeutic resistance and tumor angiogenesis. Therefore, targeting cancer stem cells may improve patient survival. We interrogated the role of a neuronal cell adhesion molecule, L1CAM, in glioma stem cells as L1CAM regulates brain development and is expressed in gliomas. L1CAM(+) and CD133(+) cells cosegregated in gliomas, and levels of L1CAM were higher in CD133(+) glioma cells than normal neural progenitors. Targeting L1CAM using lentiviral-mediated short hairpin RNA (shRNA) interference in CD133(+) glioma cells potently disrupted neurosphere formation, induced apoptosis, and inhibited growth specifically in glioma stem cells. We identified a novel mechanism for L1CAM regulation of cell survival as L1CAM knockdown decreased expression of the basic helix-loop-helix transcription factor Olig2 and up-regulated the p21(WAF1/CIP1) tumor suppressor in CD133(+) glioma cells. To determine if targeting L1CAM was sufficient to reduce glioma stem cell tumor growth in vivo, we targeted L1CAM in glioma cells before injection into immunocompromised mice or directly in established tumors. In each glioma xenograft model, shRNA targeting of L1CAM expression in vivo suppressed tumor growth and increased the survival of tumor-bearing animals. Together, these data show that L1CAM is required for maintaining the growth and survival of CD133(+) glioma cells both in vitro and in vivo, and L1CAM may represent a cancer stem cell-specific therapeutic target for improving the treatment of malignant gliomas and other brain tumors.
- Published
- 2008
- Full Text
- View/download PDF
23. Issues of diagnostic review in brain tumor studies: from the Brain Tumor Epidemiology Consortium.
- Author
-
Davis FG, Malmer BS, Aldape K, Barnholtz-Sloan JS, Bondy ML, Brännström T, Bruner JM, Burger PC, Collins VP, Inskip PD, Kruchko C, McCarthy BJ, McLendon RE, Sadetzki S, Tihan T, Wrensch MR, and Buffler PA
- Subjects
- Chicago, Humans, Incidence, Observer Variation, Registries, Reproducibility of Results, United States epidemiology, World Health Organization, Brain Neoplasms classification, Brain Neoplasms epidemiology
- Abstract
Epidemiologists routinely conduct centralized single pathology reviews to minimize interobserver diagnostic variability, but this practice does not facilitate the combination of studies across geographic regions and institutions where diagnostic practices differ. A meeting of neuropathologists and epidemiologists focused on brain tumor classification issues in the context of protocol needs for consortial studies (http://epi.grants.cancer.gov/btec/). It resulted in recommendations relevant to brain tumors and possibly other rare disease studies. Two categories of brain tumors have enough general agreement over time, across regions, and between individual pathologists that one can consider using existing diagnostic data without further review: glioblastomas and meningiomas (as long as uniform guidelines such as those provided by the WHO are used). Prospective studies of these tumors benefit from collection of pathology reports, at a minimum recording the pathology department and classification system used in the diagnosis. Other brain tumors, such as oligodendroglioma, are less distinct and require careful histopathologic review for consistent classification across study centers. Epidemiologic study protocols must consider the study specific aims, diagnostic changes that have taken place over time, and other issues unique to the type(s) of tumor being studied. As diagnostic changes are being made rapidly, there are no readily available answers on disease classification issues. It is essential that epidemiologists and neuropathologists collaborate to develop appropriate study designs and protocols for specific hypothesis and populations.
- Published
- 2008
- Full Text
- View/download PDF
24. Targeting methylguanine-DNA methyltransferase in the treatment of neuroblastoma.
- Author
-
Wagner LM, McLendon RE, Yoon KJ, Weiss BD, Billups CA, and Danks MK
- Subjects
- Adaptor Proteins, Signal Transducing metabolism, Animals, Camptothecin pharmacology, Cell Line, Tumor, Cell Proliferation drug effects, DNA Modification Methylases analysis, DNA Modification Methylases metabolism, DNA Repair Enzymes analysis, DNA Repair Enzymes metabolism, Dacarbazine pharmacology, Enzyme Inhibitors pharmacology, Guanine analogs & derivatives, Guanine pharmacology, Humans, Irinotecan, Mice, Mice, SCID, MutL Protein Homolog 1, MutS Homolog 2 Protein metabolism, Neuroblastoma pathology, Nuclear Proteins metabolism, Temozolomide, Tumor Suppressor Proteins analysis, Tumor Suppressor Proteins metabolism, Antineoplastic Agents, Phytogenic pharmacology, Camptothecin analogs & derivatives, DNA Modification Methylases antagonists & inhibitors, DNA Repair Enzymes antagonists & inhibitors, Dacarbazine analogs & derivatives, Drug Resistance, Neoplasm drug effects, Neuroblastoma enzymology, Tumor Suppressor Proteins antagonists & inhibitors
- Abstract
Purpose: The combination of temozolomide and irinotecan has preclinical schedule-dependent synergy against neuroblastoma but is not curative for relapsed high-risk patients. We hypothesized that the DNA repair protein methylguanine-DNA methyltransferase (MGMT) is an important resistance factor, and that inactivation of MGMT would sensitize neuroblastoma cells to these agents., Experimental Design: MGMT protein expression was assessed in 74 primary neuroblastoma tumors. Growth inhibition assays were done to determine the IC(50) and the extent of synergy observed with various concentrations of temozolomide, irinotecan, and the MGMT-inactivating agent O(6)-benzylguanine, using cultured syngeneic neuroblastoma cells with either low or high levels of MGMT expression. We then assessed efficacy in a mouse xenograft model of metastatic neuroblastoma., Results: MGMT was expressed by all 74 tumors evaluated. Pretreatment of neuroblastoma cells with O(6)-benzylguanine reduced the IC(50) of temozolomide by 10-fold regardless of level of MGMT expression, and pretreatment with BG followed by temozolomide + irinotecan further reduced the IC(50) in cells with high MGMT expression another 10-fold, to well below clinically achievable concentrations. The combination index was 0.27 to 0.30 for all three drugs in both cell lines, indicating strong synergy. Survival at 100 days for mice with metastatic neuroblastoma was 56% with three-drug treatment, compared with untreated controls (0%, P < 0.001) or temozolomide + irinotecan (10%, P = 0.081)., Conclusions: MGMT is widely expressed in primary neuroblastoma tumors, and is a relevant therapeutic target. Both in vitro and in vivo studies suggest inactivation of MGMT with O(6)-benzylguanine may increase the activity of temozolomide and irinotecan against neuroblastoma.
- Published
- 2007
- Full Text
- View/download PDF
25. AAL881, a novel small molecule inhibitor of RAF and vascular endothelial growth factor receptor activities, blocks the growth of malignant glioma.
- Author
-
Sathornsumetee S, Hjelmeland AB, Keir ST, McLendon RE, Batt D, Ramsey T, Yusuff N, Rasheed BK, Kieran MW, Laforme A, Bigner DD, Friedman HS, and Rich JN
- Subjects
- Animals, Antineoplastic Agents therapeutic use, Aorta, Biopsy, Cattle, Cell Line, Tumor, Endothelium, Vascular cytology, Endothelium, Vascular drug effects, Glioma drug therapy, Humans, raf Kinases genetics, Cell Division drug effects, Glioma pathology, Isoquinolines pharmacology, Receptors, Vascular Endothelial Growth Factor antagonists & inhibitors, raf Kinases antagonists & inhibitors
- Abstract
Malignant gliomas are highly proliferative and angiogenic cancers resistant to conventional therapies. Although RAS and RAF mutations are uncommon in gliomas, RAS activity is increased in gliomas. Additionally, vascular endothelial growth factor and its cognate receptors are highly expressed in gliomas. We now report that AAL881, a novel low-molecular weight inhibitor of the kinase activities associated with B-RAF, C-RAF (RAF-1), and VEGF receptor-2 (VEGFR2), showed activity against glioma cell lines and xenografts. In culture, AAL881 inhibited the downstream effectors of RAF in a concentration-dependent manner, with inhibition of proliferation associated with a G(1) cell cycle arrest, induction of apoptosis, and decreased colony formation. AAL881 decreased the proliferation of bovine aortic endothelial cells as well as the tumor cell secretion of vascular endothelial growth factor and inhibited the invasion of glioma cells through an artificial extracellular matrix. Orally administered AAL881 was well tolerated with minimal weight loss in non-tumor-bearing mice. Established s.c. human malignant glioma xenografts grown in immunocompromised mice treated with a 10-day course of oral AAL881 exhibited growth delays relative to control tumors, frequently resulting in long-term complete regressions. AAL881 treatment extended the survival of immunocompromised mice bearing orthotopic glioma xenografts compared with placebo controls. The intraparenchymal portions of orthotopic AAL881-treated tumors underwent widespread necrosis consistent with vascular disruption compared with the subarachnoid elements. These effects are distinct from our prior experience with VEGFR2 inhibitors, suggesting that targeting RAF itself or in combination with VEGFR2 induces profound tumor responses in gliomas and may serve as a novel therapeutic approach in patients with malignant gliomas.
- Published
- 2006
- Full Text
- View/download PDF
26. Stem cell-like glioma cells promote tumor angiogenesis through vascular endothelial growth factor.
- Author
-
Bao S, Wu Q, Sathornsumetee S, Hao Y, Li Z, Hjelmeland AB, Shi Q, McLendon RE, Bigner DD, and Rich JN
- Subjects
- Animals, Biopsy, Glioblastoma blood supply, Glioblastoma pathology, Humans, Male, Mice, Mice, Nude, Middle Aged, Transplantation, Heterologous, Glioma blood supply, Glioma pathology, Neovascularization, Pathologic pathology, Stem Cells pathology, Vascular Endothelial Growth Factor A physiology
- Abstract
Malignant gliomas are highly lethal cancers dependent on angiogenesis. Critical tumor subpopulations within gliomas share characteristics with neural stem cells. We examined the potential of stem cell-like glioma cells (SCLGC) to support tumor angiogenesis. SCLGC isolated from human glioblastoma biopsy specimens and xenografts potently generated tumors when implanted into the brains of immunocompromised mice, whereas non-SCLGC tumor cells isolated from only a few tumors formed secondary tumors when xenotransplanted. Tumors derived from SCLGC were morphologically distinguishable from non-SCLGC tumor populations by widespread tumor angiogenesis, necrosis, and hemorrhage. To determine a potential molecular mechanism for SCLGC in angiogenesis, we measured the expression of a panel of angiogenic factors secreted by SCLGC. In comparison with matched non-SCLGC populations, SCLGC consistently secreted markedly elevated levels of vascular endothelial growth factor (VEGF), which were further induced by hypoxia. In an in vitro model of angiogenesis, SCLGC-conditioned medium significantly increased endothelial cell migration and tube formation compared with non-SCLGC tumor cell-conditioned medium. The proangiogenic effects of glioma SCLGC on endothelial cells were specifically abolished by the anti-VEGF neutralizing antibody bevacizumab, which is in clinical use for cancer therapy. Furthermore, bevacizumab displayed potent antiangiogenic efficacy in vivo and suppressed growth of xenografts derived from SCLGC but limited efficacy against xenografts derived from a matched non-SCLGC population. Together these data indicate that stem cell-like tumor cells can be a crucial source of key angiogenic factors in cancers and that targeting proangiogenic factors from stem cell-like tumor populations may be critical for patient therapy.
- Published
- 2006
- Full Text
- View/download PDF
27. Phase 1 trial of gefitinib plus sirolimus in adults with recurrent malignant glioma.
- Author
-
Reardon DA, Quinn JA, Vredenburgh JJ, Gururangan S, Friedman AH, Desjardins A, Sathornsumetee S, Herndon JE 2nd, Dowell JM, McLendon RE, Provenzale JM, Sampson JH, Smith RP, Swaisland AJ, Ochs JS, Lyons P, Tourt-Uhlig S, Bigner DD, Friedman HS, and Rich JN
- Subjects
- Administration, Oral, Adult, Aged, Biomarkers, Tumor analysis, Dose-Response Relationship, Drug, Drug Administration Schedule, Female, Gefitinib, Humans, Male, Middle Aged, Recurrence, Treatment Outcome, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Glioma drug therapy, Quinazolines administration & dosage, Sirolimus administration & dosage
- Abstract
Purpose: To determine the maximum tolerated dose (MTD) and dose-limiting toxicity (DLT) of gefitinib, a receptor tyrosine kinase inhibitor of the epidermal growth factor receptor, plus sirolimus, an inhibitor of the mammalian target of rapamycin, among patients with recurrent malignant glioma., Patients and Methods: Gefitinib and sirolimus were administered on a continuous daily dosing schedule at dose levels that were escalated in successive cohorts of malignant glioma patients at any recurrence who were stratified based on concurrent use of CYP3A-inducing anticonvulsants [enzyme-inducing antiepileptic drugs, (EIAED)]. Pharmacokinetic and archival tumor biomarker data were also assessed., Results: Thirty-four patients with progressive disease after prior radiation therapy and chemotherapy were enrolled, including 29 (85%) with glioblastoma multiforme and 5 (15%) with anaplastic glioma. The MTD was 500 mg of gefitinib plus 5 mg of sirolimus for patients not on EIAEDs and 1,000 mg of gefitinib plus 10 mg of sirolimus for patients on EIAEDs. DLTs included mucositis, diarrhea, rash, thrombocytopenia, and hypertriglyceridemia. Gefitinib exposure was not affected by sirolimus administration but was significantly lowered by concurrent EIAED use. Two patients (6%) achieved a partial radiographic response, and 13 patients (38%) achieved stable disease., Conclusion: We show that gefitinib plus sirolimus can be safely coadministered on a continuous, daily dosing schedule, and established the recommended dose level of these agents in combination for future phase 2 clinical trials.
- Published
- 2006
- Full Text
- View/download PDF
28. ZD6474, a novel tyrosine kinase inhibitor of vascular endothelial growth factor receptor and epidermal growth factor receptor, inhibits tumor growth of multiple nervous system tumors.
- Author
-
Rich JN, Sathornsumetee S, Keir ST, Kieran MW, Laforme A, Kaipainen A, McLendon RE, Graner MW, Rasheed BK, Wang L, Reardon DA, Ryan AJ, Wheeler C, Dimery I, Bigner DD, and Friedman HS
- Subjects
- Animals, Cell Line, Tumor, Cell Movement drug effects, Cell Proliferation drug effects, Central Nervous System Neoplasms metabolism, Central Nervous System Neoplasms pathology, Dose-Response Relationship, Drug, Ependymoma drug therapy, Ependymoma pathology, ErbB Receptors metabolism, Glioma drug therapy, Glioma pathology, Humans, Immunohistochemistry, Ki-67 Antigen analysis, Mice, Mice, Nude, Mitogen-Activated Protein Kinase 1 metabolism, Mitogen-Activated Protein Kinase 3 metabolism, Neovascularization, Pathologic metabolism, Neovascularization, Pathologic pathology, Neovascularization, Pathologic prevention & control, Phosphorylation drug effects, Receptors, Vascular Endothelial Growth Factor metabolism, Xenograft Model Antitumor Assays, Central Nervous System Neoplasms drug therapy, ErbB Receptors antagonists & inhibitors, Piperidines pharmacology, Quinazolines pharmacology, Receptors, Vascular Endothelial Growth Factor antagonists & inhibitors
- Abstract
Purpose: Primary central nervous system (CNS) tumors represent a diverse group of tumor types with heterogeneous molecular mechanisms that underlie their formation and maintenance. CNS tumors depend on angiogenesis and often display increased activity of ErbB-associated pathways. Current nonspecific therapies frequently have poor efficacy in many of these tumor types, so there is a pressing need for the development of novel targeted therapies., Experimental Design: ZD6474 is a novel, orally available low molecular weight inhibitor of the kinase activities associated with vascular endothelial growth factor receptor-2 and epidermal growth factor receptor. We hypothesized that ZD6474 may provide benefit in the treatment of several CNS tumor types., Results: In mice bearing established s.c. tumor xenografts of CNS tumors (malignant glioma and ependymoma) or rhabdomyosarcoma, a limited course of ZD6474 treatment produced significant tumor growth delays and a high rate of partial tumor regression in most models examined. Mice with i.c. malignant glioma xenografts treated with ZD6474 experienced a significant prolongation of survival. Tumors from mice treated with ZD6474 displayed a lower proliferative index and disrupted tumor vascularity. Notably, some of these models are insensitive to low molecular weight kinase inhibitors targeting only vascular endothelial growth factor receptor-2 or epidermal growth factor receptor functions, suggesting that the combined disruption of both epidermal growth factor receptor and vascular endothelial growth factor receptor-2 activities may significantly increase tumor control., Conclusions: In conclusion, ZD6474 shows significant activity against xenograft models of several primary human CNS tumor types. Consideration for clinical development in this disease setting seems warranted.
- Published
- 2005
- Full Text
- View/download PDF
29. Gene expression profiling and genetic markers in glioblastoma survival.
- Author
-
Rich JN, Hans C, Jones B, Iversen ES, McLendon RE, Rasheed BK, Dobra A, Dressman HK, Bigner DD, Nevins JR, and West M
- Subjects
- Aged, Brain Neoplasms metabolism, Cyclin-Dependent Kinase Inhibitor p16 biosynthesis, Cyclin-Dependent Kinase Inhibitor p16 genetics, Doublecortin Domain Proteins, ErbB Receptors biosynthesis, ErbB Receptors genetics, Female, Gene Expression Profiling, Genetic Markers genetics, Glioblastoma metabolism, Humans, Loss of Heterozygosity, Male, Membrane Glycoproteins biosynthesis, Membrane Glycoproteins genetics, Microtubule-Associated Proteins biosynthesis, Microtubule-Associated Proteins genetics, Middle Aged, Neuropeptides biosynthesis, Neuropeptides genetics, Osteonectin biosynthesis, Osteonectin genetics, PTEN Phosphohydrolase, Phosphoric Monoester Hydrolases biosynthesis, Phosphoric Monoester Hydrolases genetics, Reproducibility of Results, Semaphorins, Survival Rate, Tumor Suppressor Protein p53 biosynthesis, Tumor Suppressor Protein p53 genetics, Tumor Suppressor Proteins biosynthesis, Tumor Suppressor Proteins genetics, Brain Neoplasms genetics, Glioblastoma genetics
- Abstract
Despite the strikingly grave prognosis for older patients with glioblastomas, significant variability in patient outcome is experienced. To explore the potential for developing improved prognostic capabilities based on the elucidation of potential biological relationships, we did analyses of genes commonly mutated, amplified, or deleted in glioblastomas and DNA microarray gene expression data from tumors of glioblastoma patients of age >50 for whom survival is known. No prognostic significance was associated with genetic changes in epidermal growth factor receptor (amplified in 17 of 41 patients), TP53 (mutated in 11 of 41 patients), p16INK4A (deleted in 15 of 33 patients), or phosphatase and tensin homologue (mutated in 15 of 41 patients). Statistical analysis of the gene expression data in connection with survival involved exploration of regression models on small subsets of genes, based on computational search over multiple regression models with cross-validation to assess predictive validity. The analysis generated a set of regression models that, when weighted and combined according to posterior probabilities implied by the statistical analysis, identify patterns in expression of a small subset of genes that are associated with survival and have value in assessing survival risks. The dominant genes across such multiple regression models involve three key genes-SPARC (Osteonectin), Doublecortex, and Semaphorin3B-which play key roles in cellular migration processes. Additional analysis, based on statistical graphical association models constructed using similar computational analysis methods, reveals other genes which support the view that multiple mediators of tumor invasion may be important prognostic factor in glioblastomas in older patients.
- Published
- 2005
- Full Text
- View/download PDF
30. Identification of OTX2 as a medulloblastoma oncogene whose product can be targeted by all-trans retinoic acid.
- Author
-
Di C, Liao S, Adamson DC, Parrett TJ, Broderick DK, Shi Q, Lengauer C, Cummins JM, Velculescu VE, Fults DW, McLendon RE, Bigner DD, and Yan H
- Subjects
- Brain Neoplasms metabolism, Cell Growth Processes drug effects, Cell Growth Processes genetics, Cell Line, Tumor, Gene Amplification, Homeodomain Proteins antagonists & inhibitors, Homeodomain Proteins biosynthesis, Humans, Medulloblastoma metabolism, Medulloblastoma pathology, Nerve Tissue Proteins antagonists & inhibitors, Nerve Tissue Proteins biosynthesis, Oncogenes drug effects, Oncogenes genetics, Otx Transcription Factors, RNA, Small Interfering genetics, RNA, Small Interfering pharmacology, Trans-Activators antagonists & inhibitors, Trans-Activators biosynthesis, Antineoplastic Agents pharmacology, Brain Neoplasms genetics, Homeodomain Proteins genetics, Medulloblastoma drug therapy, Medulloblastoma genetics, Nerve Tissue Proteins genetics, Trans-Activators genetics, Tretinoin pharmacology
- Abstract
Through digital karyotyping of permanent medulloblastoma cell lines, we found that the homeobox gene OTX2 was amplified more than 10-fold in three cell lines. Gene expression analyses showed that OTX2 transcripts were present at high levels in 14 of 15 (93%) medulloblastomas with anaplastic histopathologic features. Knockdown of OTX2 expression by siRNAs inhibited medulloblastoma cell growth in vitro, whereas pharmacologic doses of all-trans retinoic acid repressed OTX2 expression and induced apoptosis only in medulloblastoma cell lines that expressed OTX2. These observations suggest that OTX2 is essential for the pathogenesis of anaplastic medulloblastomas and that these tumors may be amenable to therapy with all-trans-retinoic acid.
- Published
- 2005
31. Mutations of PIK3CA in anaplastic oligodendrogliomas, high-grade astrocytomas, and medulloblastomas.
- Author
-
Broderick DK, Di C, Parrett TJ, Samuels YR, Cummins JM, McLendon RE, Fults DW, Velculescu VE, Bigner DD, and Yan H
- Subjects
- Astrocytoma pathology, Brain Neoplasms pathology, Class I Phosphatidylinositol 3-Kinases, DNA, Neoplasm genetics, Humans, Medulloblastoma pathology, Oligodendroglioma pathology, Phosphoric Monoester Hydrolases genetics, Astrocytoma genetics, Brain Neoplasms genetics, Medulloblastoma genetics, Mutation, Oligodendroglioma genetics, Phosphatidylinositol 3-Kinases genetics
- Abstract
The phosphatidylinositol 3'-kinase pathway is activated in multiple advanced cancers, including glioblastomas, through inactivation of the PTEN tumor suppressor gene. Recently, mutations in PIK3CA, a member of the family of phosphatidylinositol 3'-kinase catalytic subunits, were identified in a significant fraction (25-30%) of colorectal cancers, gastric cancers, and glioblastomas and in a smaller fraction of breast and lung cancers. These mutations were found to cluster into two major "hot spots" located in the helical and catalytic domains. To determine whether PIK3CA is genetically altered in brain tumors, we performed a large-scale mutational analysis of the helical and catalytic domains. A total of 13 mutations of PIK3CA within these specific domains were identified in anaplastic oligodendrogliomas, anaplastic astrocytomas, glioblastoma multiforme, and medulloblastomas, whereas no mutations were identified in ependymomas or low-grade astrocytomas. These observations implicate PIK3CA as an oncogene in a wider spectrum of adult and pediatric brain tumors and suggest that PIK3CA may be a useful diagnostic marker or a therapeutic target in these cancers.
- Published
- 2004
- Full Text
- View/download PDF
32. Efficacy of intracerebral microinfusion of trastuzumab in an athymic rat model of intracerebral metastatic breast cancer.
- Author
-
Grossi PM, Ochiai H, Archer GE, McLendon RE, Zalutsky MR, Friedman AH, Friedman HS, Bigner DD, and Sampson JH
- Subjects
- Animals, Antibodies, Monoclonal administration & dosage, Antibodies, Monoclonal, Humanized, Antineoplastic Agents administration & dosage, Disease Models, Animal, Female, Infusions, Parenteral, Neoplasm Metastasis, Rats, Rats, Nude, Survival Analysis, Trastuzumab, Antibodies, Monoclonal therapeutic use, Antineoplastic Agents therapeutic use, Brain Neoplasms drug therapy, Brain Neoplasms secondary, Breast Neoplasms drug therapy, Breast Neoplasms pathology
- Abstract
Purpose: The monoclonal antibody (MAb) trastuzumab (Herceptin) effectively treats HER2-overexpressing extracerebral breast neoplasms. Delivery of such macromolecule therapeutic agents to intracerebral metastases, however, is limited by the tight junctions characteristic of the cerebral vasculature. Direct intracerebral microinfusion (ICM) is a technique that bypasses this blood-brain barrier and allows for a greater delivery of drugs directly into intracerebral tumors., Experimental Design: A human breast cancer cell line transfected to overexpress HER2, MCF-7/HER2-18, was transplanted into the cerebrum of athymic rats. Saline, trastuzumab, or an isotype-matched control MAb was delivered systemically or by ICM to assess toxicity and efficacy., Results: No clinical or histological toxicity related to trastuzumab was evident under any of the conditions studied. Delivery of trastuzumab (2 mg/kg) i.p. led to a median survival of 26.5 days, whereas treatment with trastuzumab (2 mg/kg) by ICM increased the median survival by 96% to 52 days, with two of nine rats surviving >120 days (P = 0.009). Treatment with an isotype-matched control MAb (16 mg/kg) resulted in a median survival of 21 days, which did not differ significantly from the survival of rats treated by ICM with saline (16 days; P = 0.42). Treatment by ICM with trastuzumab (16 mg/kg) led to a median survival of 45 days, with 2 of 10 rats surviving >120 days. These results represent 181% and 114% increases in median survival over the saline and MAb controls, respectively (P < 0.001)., Conclusion: ICM of trastuzumab is safe and superior to systemic delivery as therapy for HER2-overexpressing intracerebral neoplasms in an athymic rat model.
- Published
- 2003
33. Brain tumors in mice are susceptible to blockade of epidermal growth factor receptor (EGFR) with the oral, specific, EGFR-tyrosine kinase inhibitor ZD1839 (iressa).
- Author
-
Heimberger AB, Learn CA, Archer GE, McLendon RE, Chewning TA, Tuck FL, Pracyk JB, Friedman AH, Friedman HS, Bigner DD, and Sampson JH
- Subjects
- 3T3 Cells, Administration, Oral, Animals, Antineoplastic Agents pharmacology, Blotting, Western, DNA Mutational Analysis, Enzyme Inhibitors pharmacology, Flow Cytometry, Gefitinib, Humans, Male, Mice, Mice, Inbred BALB C, Mice, Nude, Neoplasm Transplantation, Phosphorylation, Protein-Tyrosine Kinases antagonists & inhibitors, Signal Transduction, Time Factors, Tumor Cells, Cultured, Tyrosine metabolism, Brain Neoplasms drug therapy, Brain Neoplasms metabolism, ErbB Receptors antagonists & inhibitors, Quinazolines pharmacology
- Abstract
Iressa (ZD1839) is a p.o.-active, selective, epidermal growth factor receptor tyrosine kinase inhibitor (EGFR-TKI) that blocks signal transduction pathways implicated in cancer cell proliferation, survival, and host-dependent processes promoting cancer growth. EGFR is up-regulated in primary malignant tumors of the central nervous system (CNS) and in many systemic tumors that metastasize to the CNS. The purpose of our study was to evaluate the efficacy and toxicity of p.o.-administered ZD1839 for the treatment of established intracerebral (i.c.) tumors expressing EGFR or the tumorigenic mutated variant EGFRvIII, which is constitutively phosphorylated. Oral administration of ZD1839 at 50 or 100 mg/kg/day for 3 weeks in athymic mice with established i.c. A431 human epidermoid carcinoma expressing EGFR increased median survival by 88% (P = 0.009) and 105% (P < 0.001), respectively. Additionally, there was no evidence of systemic or CNS toxicity. However, ZD1839 failed to inhibit either s.c. or i.c. in vivo tumor growth when tumorigenicity was conferred by EGFRvIII. Western blotting revealed that treatment with ZD1839 virtually ablated phosphorylation of EGFR Tyr-1173 in A431 tumors. However, treatment of NR6M tumors with ZD1839 only partially decreased phosphorylation of EGFRvIII Tyr-1173 while up-regulating overall expression, suggesting that EGFRvIII may not be susceptible to the same molecular mechanisms of tyrosine kinase inhibition as EGFR. In conclusion, ZD1839 is active in a brain tumor model expressing EGFR, but not EGFRvIII, as EGFR mutations may lead to relative therapeutic resistance. On the basis of these observations, we believe that clinical trials of ZD1839 against brain tumors expressing EGFR are warranted, but that special consideration should be given to tumors that coexpress EGFRvIII.
- Published
- 2002
34. Targeted delivery in primary and metastatic brain tumors: summary report of the seventh annual meeting of the Blood-Brain Barrier Disruption Consortium.
- Author
-
Doolittle ND, Abrey LE, Ferrari N, Hall WA, Laws ER, McLendon RE, Muldoon LL, Peereboom D, Peterson DR, Reynolds CP, Senter P, and Neuwelt EA
- Subjects
- Brain Neoplasms pathology, Brain Neoplasms secondary, Clinical Trials as Topic, Combined Modality Therapy, Drug Implants, Humans, Antineoplastic Agents administration & dosage, Blood-Brain Barrier drug effects, Brain Neoplasms drug therapy, Drug Delivery Systems
- Abstract
The November 2000 NIH report of the Brain Tumor Progress Review Group identified delivering and targeting therapeutic agents as a priority in the treatment of malignant brain tumors. For this reason, the seventh annual Blood-Brain Barrier Disruption Consortium meeting, partially funded by an NIH R13 Grant, focused on recent advances in targeted delivery to the central nervous system, clinical trials for primary and metastatic brain tumors using enhanced chemotherapy delivery, and strategies to lessen the toxicities associated with dose intensive treatments, using thiols.
- Published
- 2002
35. A genetically tractable model of human glioma formation.
- Author
-
Rich JN, Guo C, McLendon RE, Bigner DD, Wang XF, and Counter CM
- Subjects
- Animals, Antigens, Polyomavirus Transforming biosynthesis, Antigens, Polyomavirus Transforming genetics, Astrocytes metabolism, Astrocytes pathology, Astrocytes physiology, Catalytic Domain, DNA-Binding Proteins, Genes, ras, Glioma pathology, Humans, Mice, Mice, Inbred BALB C, Mice, Nude, Mice, SCID, Retroviridae genetics, Telomerase biosynthesis, Telomerase genetics, Tumor Cells, Cultured, Cell Transformation, Viral genetics, Glioma genetics, RNA
- Abstract
Gliomas remain one of the deadliest forms of cancer. Improved therapeutics will require a better understanding of the molecular nature of these tumors. We, therefore, mimicked the most common genetic changes found in grade III-IV gliomas, disruption of the p53 and RB pathways and activation of telomere maintenance and independence from growth factors, through the ectopic expression of the SV40 T/t-Ag oncogene, an oncogenic form of H-ras (H-ras(V12G)), and the human telomerase catalytic subunit hTERT in normal human astrocytes. The resulting cells displayed many of the hallmarks of grade III-IV gliomas, including greatly expanded life span and growth in soft agar and, most importantly, were tumorigenic with pathology consistent with grade III-IV neuroectodermal tumors in mice. This model system will, for the first time, allow the biological significance of selected genetic alterations to be studied in human gliomas.
- Published
- 2001
36. Temozolomide delivered by intracerebral microinfusion is safe and efficacious against malignant gliomas in rats.
- Author
-
Heimberger AB, Archer GE, McLendon RE, Hulette C, Friedman AH, Friedman HS, Bigner DD, and Sampson JH
- Subjects
- Animals, Antineoplastic Agents, Alkylating therapeutic use, Brain drug effects, Catheterization, Dacarbazine therapeutic use, Humans, Male, Neoplasm Transplantation, Rats, Rats, Nude, Temozolomide, Time Factors, Tumor Cells, Cultured, Antineoplastic Agents, Alkylating administration & dosage, Brain Neoplasms drug therapy, Catheters, Indwelling, Dacarbazine administration & dosage, Dacarbazine analogs & derivatives, Drug Delivery Systems, Glioma drug therapy
- Abstract
Intracerebral microinfusion (ICM) is an innovative technique of delivering therapeutic agents throughout large portions of the brain that circumvents the blood-brain barrier, minimizes systemic toxicity, and provides a homogeneous distribution of the infused agent. Temozolomide is a novel methylating agent with proven efficacy against malignant gliomas (MGs) after systemic administration but with dose-limiting myelotoxicity. Because MGs rarely metastasize, systemic drug delivery is unnecessary. Therefore, we evaluated the efficacy and toxicity of ICM with temozolomide in an athymic rat model of human MGs. Treatment of rats by ICM with temozolomide 3 days after intracerebral challenge with D54 human MG xenograft increased median survival by 128% compared with rats treated by ICM with saline, by 113% compared with rats treated with i.p. saline, and by 100% compared with rats treated with i.p. temozolomide (P < 0.001). Delay of treatment until 9 days after tumor challenge still resulted in a 23% increase in median survival in rats treated by ICM of temozolomide compared with rats treated with i.p. temozolomide. In addition, overall, 21.7% of rats treated by ICM with temozolomide survived for > 100 days without clinical or histological evidence of tumor. The dose of temozolomide delivered by ICM in this study was limited only by drug solubility, and no neurological or systemic toxicity could be attributed to ICM with temozolomide. Therefore, ICM of temozolomide may offer significant advantages in the treatment of MGs.
- Published
- 2000
37. A public database for gene expression in human cancers.
- Author
-
Lal A, Lash AE, Altschul SF, Velculescu V, Zhang L, McLendon RE, Marra MA, Prange C, Morin PJ, Polyak K, Papadopoulos N, Vogelstein B, Kinzler KW, Strausberg RL, and Riggins GJ
- Subjects
- Brain metabolism, Cloning, Molecular, Glioblastoma genetics, Humans, Internet, Models, Theoretical, RNA, Messenger analysis, Reverse Transcriptase Polymerase Chain Reaction, Tissue Distribution, Databases, Factual, Gene Expression, Neoplasms genetics
- Abstract
A public database, SAGEmap, was created as a component of the Cancer Genome Anatomy Project to provide a central location for depositing, retrieving, and analyzing human gene expression data. This database uses serial analysis of gene expression to quantify transcript levels in both malignant and normal human tissues. By accessing SAGEmap (http://www.ncbi.nlm.nih.gov/SAGE) the user can compare transcript populations between any of the posted libraries. As an initial demonstration of the database's utility, gene expression in human glioblastomas was compared with that of normal brain white matter. Of the 47,174 unique transcripts expressed in these two tissues, 471 (1.0%) were differentially expressed by more than 5-fold (P<0.001). Classification of these genes revealed functions consistent with the biological properties of glioblastomas, in particular: angiogenesis, transcription, and cell cycle related genes.
- Published
- 1999
38. Regional treatment of epidermal growth factor receptor vIII-expressing neoplastic meningitis with a single-chain immunotoxin, MR-1.
- Author
-
Archer GE, Sampson JH, Lorimer IA, McLendon RE, Kuan CT, Friedman AH, Friedman HS, Pastan IH, and Bigner DD
- Subjects
- Animals, Antibody Specificity, ErbB Receptors biosynthesis, ErbB Receptors genetics, Exotoxins toxicity, Female, Humans, Immunoglobulin Variable Region toxicity, Immunotoxins toxicity, Injections, Spinal, Meningeal Neoplasms metabolism, Mice, Mice, Nude, Mutation, Protein Synthesis Inhibitors pharmacology, Rats, Rats, Nude, Tumor Cells, Cultured, Pseudomonas aeruginosa Exotoxin A, ADP Ribose Transferases, Bacterial Toxins, ErbB Receptors immunology, ErbB Receptors metabolism, Exotoxins pharmacology, Immunoglobulin Variable Region pharmacology, Immunotoxins pharmacology, Meningeal Neoplasms drug therapy, Virulence Factors
- Abstract
The incidence of neoplastic meningitis is on the rise. Neoplastic meningitis can result from a direct seeding of the neuraxis by primary brain tumors or by hematogeneous spread of systemic solid tumors. A frequent genetic alteration in primary brain tumors such as gliomas is an in-frame deletion in the epidermal growth factor receptor (EGFR) gene EGFRvIII, which brings together what were normally distant polypeptide sequences in the intact receptor. A novel glycine is formed at the fusion junction, resulting in a unique and tumor-specific target. By using phage display, we have isolated a single-chain antibody specific for the EGFRvIII mutation and expressed it with a modified form of the Pseudomonas exotoxin to form the immunotoxin MR1scFvPE38KDEL (MR-1). The multiple dose toxicity and therapeutic efficacy of MR-1 immunotoxin were tested in an athymic rat model of neoplastic meningitis. The maximally tolerated doses in non-tumor-bearing rats were three doses of 3 microg each. For therapeutic studies, the target was a neoplastic meningitis induced by intrathecal inoculation of the EGFRvIII-expressing human glioma U87MG.deltaEGFR. A dose escalation study compared the survival of three equal doses of 1, 2, and 3 microg of MR-1 immunotoxin with saline or 3 microg of the control immunotoxin specific for the interleukin 2 receptor, anti-Tac. All animals treated with three doses of saline or 3 microg of anti-Tac died, with median survival of 7 and 10 days, respectively. There were 75% (six of eight) long-term survivors in the group treated with three doses of 1 microg and 57% (four of seven) long-term survivors in the groups treated with three doses of either 2 or 3 microg of MR-1 immunotoxin. None of the MR-1 immunotoxin-treated groups reached median survival by the termination of the study at 53 days. Therefore, median survival was estimated to be >53 days, resulting in an estimated increase in median survival of >657% compared with saline and 430% versus anti-Tac. Compartmental therapy with three doses of 2 microg of MR-1 immunotoxin is effective in the treatment of EGFRvIII-expressing neoplastic meningitis. This dose was found to have no clinical or histopathological effects on non-tumor-bearing animals. MR-1 immunotoxin is, therefore, considered specific and safe within its therapeutic window. Phase I clinical trials for tumors invading the intrathecal space that express the EGFRvIII target should be initiated.
- Published
- 1999
39. Treatment of neoplastic meningitis with intrathecal temozolomide.
- Author
-
Sampson JH, Archer GE, Villavicencio AT, McLendon RE, Friedman AH, Bishop WR, Bigner DD, and Friedman HS
- Subjects
- Animals, Antineoplastic Agents, Alkylating administration & dosage, Antineoplastic Agents, Alkylating pharmacokinetics, Antineoplastic Agents, Alkylating toxicity, Dacarbazine administration & dosage, Dacarbazine pharmacokinetics, Dacarbazine therapeutic use, Dacarbazine toxicity, Drug Screening Assays, Antitumor, Humans, Neoplasm Transplantation, Rats, Rats, Nude, Solubility, Subarachnoid Space, Temozolomide, Transplantation, Heterologous, Antineoplastic Agents, Alkylating therapeutic use, Dacarbazine analogs & derivatives, Meningeal Neoplasms drug therapy, Meningeal Neoplasms secondary
- Abstract
Neoplastic meningitis (NM) results from leptomeningeal dissemination of cancers arising within the central nervous system or metastasizing to the leptomeninges from systemic neoplasms. The inability to produce therapeutic drug levels intrathecally (i.t.) with systemic administration and the minimal efficacy of chemotherapeutic agents currently available for direct i.t. use limit therapy. Temozolomide [8-carbamoyl-3-methylimidazo[5,1-d]-1,2,3,5-tetrazin-4([3H])-one] is a novel methylating agent with proven activity against intraparenchymal malignant gliomas (MGs). Insolubility of the standard formulation prevents its efficacious use as an i.t. agent, however. To overcome this obstacle, we have developed a unique microcrystalline formulation of temozolomide with greatly enhanced solubility. Treatment of athymic rats bearing subarachnoid MER- human MG xenografts with four doses of i.t. microcrystalline temozolomide over a 2-week period produced a 142% increase in median survival at individual doses of 2.2 micromol (P = 0.0073) and a >367% increase in median survival at individual doses of 6.8 micromol (P = 0.0015). At the higher dose tested, three of eight rats treated developed no neurological symptoms and had no evidence of residual tumor on histological examination after treatment. Use of this microcrystalline formulation in athymic rats bearing subarachnoid MER+ human MG xenografts increased median survival >132% (P < 0.0058) at both dose levels tested. Toxicity directly attributable to the i.t. administration of microcrystalline temozolomide was exhibited in the highest dose groups only and was limited to small patchy areas of focal demyelination involving <5% of spinal cord long tracks.
- Published
- 1999
40. PTEN gene mutations are seen in high-grade but not in low-grade gliomas.
- Author
-
Rasheed BK, Stenzel TT, McLendon RE, Parsons R, Friedman AH, Friedman HS, Bigner DD, and Bigner SH
- Subjects
- Adult, Aged, Aged, 80 and over, Brain Neoplasms pathology, Child, DNA Mutational Analysis, DNA, Neoplasm genetics, Disease Progression, Female, Genes, p53, Glioma pathology, Humans, Loss of Heterozygosity, Male, Medulloblastoma genetics, Medulloblastoma pathology, Middle Aged, Polymorphism, Single-Stranded Conformational, Sequence Deletion, Brain Neoplasms genetics, Genes, Tumor Suppressor, Glioma genetics
- Abstract
The PTEN gene, located on 10q23, has recently been implicated as a candidate tumor suppressor gene in brain, breast and prostate tumors. In the present study, 123 brain tumors, including various grades and histological types of gliomas occurring in children and adults, were analyzed for PTEN mutations by SSCP assay and sequencing. Mutations in the PTEN gene were found in 13 of 42 adult glioblastomas and 3 of 13 adult anaplastic astrocytomas, whereas none of the 21 low-grade adult gliomas or the 22 childhood gliomas of all grades showed mutations. The single medulloblastoma with a mutation was a recurrent tumor that also possessed a p53 mutation. High-grade adult gliomas with PTEN mutations included cases that also contained gene amplification or p53 gene mutations, as well as cases that did not contain either of these abnormalities. There was no obvious relationship between presence of PTEN mutation and survival; however, there was a tendency for PTEN mutations to occur in older age group patients. This analysis suggest that PTEN gene mutations are restricted to high-grade adult gliomas and that this abnormality is independent of the presence or absence of gene amplification or p53 gene mutation in these tumors.
- Published
- 1997
41. Cell surface localization and density of the tumor-associated variant of the epidermal growth factor receptor, EGFRvIII.
- Author
-
Wikstrand CJ, McLendon RE, Friedman AH, and Bigner DD
- Subjects
- Animals, Antigen-Antibody Complex metabolism, Biopsy, Cell Compartmentation, Cell Membrane metabolism, Central Nervous System Neoplasms pathology, Endocytosis, Flow Cytometry, Fluorescent Antibody Technique, Indirect, Glioma pathology, Humans, Mice, Microscopy, Confocal, Transfection, Tumor Cells, Cultured, Central Nervous System Neoplasms metabolism, ErbB Receptors metabolism, Glioma metabolism
- Abstract
The potential of therapeutic targeting of tumor cell surface epidermal growth factor receptors (EGFRs) by modified ligands or specific antibodies has been limited by the normal tissue distribution of the receptor. The identification and characterization of a variant of this receptor, EGFRvIII, which is not expressed in normal tissues but has been described in gliomas, non-small cell lung carcinomas, and breast carcinomas, has provided a highly specific, internalizing target for antibody-mediated approaches. To determine the feasibility of immunotargeting EGFRvIII, we have assessed the qualitative distribution and quantitative expression at both the population and cellular levels of EGFRvIII in 21 biopsy samples of human gliomas by indirect analytical and quantitative flow cytometry and by immunohistochemical assay of frozen and formalin-fixed tissue. Consistent with previous reports, 50% of gliomas tested (1 of 2 anaplastic astrocytomas, 7 of 12 glioblastoma multiforme, and 2 of 6 oligodendrogliomas) expressed EGFRvIII, as determined by a minimum of 2 separate assays. Minimum estimates of the proportion of positive tumor cells in these populations ranged from 37-86%; in four of five cases in which quantitation of the EGFRvIII density/cell was performed, values of 2.7-6.8 x 10(5) were obtained with monoclonal antibody (mAb) L8A4 (EGFRvIII specific), levels consistent with successful in vivo immunotargeting. Confocal microscopic analysis confirmed that the subcellular localization of EGFRvIII was identical to that described for EGFR: predominant cell membrane expression, with some perinuclear distribution suggestive of localization to the Golgi region. Neither EGFR nor EGFRvIII was found within the nucleus. This study establishes for the first time that approximately 50% of human glioma biopsies contain cell populations expressing a sufficient number of membrane-expressed EGFRvIIIs to mediate specific anti-EGFRvIII mAb localization. Coupled with previous demonstrations of the rapid internalization of specific mAb-EGFRvIII complexes and the susceptibility of the targeted cells to isotope or toxin-mediated cytotoxicity, this study establishes the validity of targeting EGFRvIII for therapy of mutant receptor-positive gliomas, breast carcinomas, and non-small cell lung carcinomas.
- Published
- 1997
42. Intrathecal 131I-labeled antitenascin monoclonal antibody 81C6 treatment of patients with leptomeningeal neoplasms or primary brain tumor resection cavities with subarachnoid communication: phase I trial results.
- Author
-
Brown MT, Coleman RE, Friedman AH, Friedman HS, McLendon RE, Reiman R, Felsberg GJ, Tien RD, Bigner SH, Zalutsky MR, Zhao XG, Wikstrand CJ, Pegram CN, Herndon JE 2nd, Vick NA, Paleologos N, Fredericks RK, Schold SC Jr, and Bigner DD
- Subjects
- Adolescent, Adult, Aged, Animals, Antibodies, Monoclonal immunology, Brain Neoplasms mortality, Child, Preschool, Female, Humans, Male, Meningeal Neoplasms mortality, Mice, Middle Aged, Radiotherapy Dosage, Antibodies, Monoclonal therapeutic use, Brain Neoplasms radiotherapy, Iodine Radioisotopes therapeutic use, Meningeal Neoplasms radiotherapy, Radioimmunotherapy adverse effects
- Abstract
We aimed to determine the maximum tolerated dose (MTD) of 131I-labeled 81C6 in patients with leptomeningeal neoplasms or brain tumor resection cavities with subarachnoid communication and to identify any objective responses. 81C6 is a murine IgG monoclonal antibody that reacts with tenascin in gliomas/carcinomas but does not react with normal adult brain. 131I-labeled 81C6 delivers intrathecal (IT) radiation to these neoplasms. This study was a Phase I trial in which patients were treated with a single IT dose of 131I-labeled 81C6. Cohorts of three to six patients were treated with escalating doses of 131I (starting dose, 40 mCi; 20 mCi escalations) on 10 mg 81C6. MTD is defined as the highest dose resulting in serious toxicity in no more than two of six patients. Serious toxicity is defined as grade III/IV nonhematological toxicity or major hematological toxicity. We treated 31 patients (8 pediatric and 23 adult). Eighteen had glioblastoma multiforme. Patients were treated with 131I doses from 40 to 100 mCi. Hematological toxicity was dose limiting and correlated with the administered 131I dose. No grade III/IV nonhematological toxicities were encountered. A partial response occurred in 1 patient and disease stabilization occurred in 13 (42%) of 31 patients. Twelve patients are alive (median follow-up, > 320 days); five are progression free >409 days median posttreatment. The MTD of a single IT administration of 131I-labeled 81C6 in adults is 80 mCi 131I-labeled 81C6. The MTD in pediatric patients was not reached at 131I doses up to 40 mCi normalized for body surface area.
- Published
- 1996
43. Efficacy of compartmental administration of immunotoxin LMB-1 (B3-LysPE38) in a rat model of carcinomatous meningitis.
- Author
-
Bigner DD, Archer GE, McLendon RE, Friedman HS, Fuchs HE, Pai LH, Herndon JE 2nd, and Pastan IH
- Subjects
- Animals, Carcinoma immunology, Carcinoma pathology, Disease Models, Animal, Drug Screening Assays, Antitumor, Humans, Meningeal Neoplasms immunology, Meningeal Neoplasms pathology, Rats, Rats, Nude, Tumor Cells, Cultured, Pseudomonas aeruginosa Exotoxin A, ADP Ribose Transferases, Antibodies, Monoclonal administration & dosage, Bacterial Toxins, Carcinoma therapy, Exotoxins administration & dosage, Immunotoxins administration & dosage, Lewis Blood Group Antigens immunology, Meningeal Neoplasms therapy, Virulence Factors
- Abstract
LMB-1 (B3-LysPE38) is an immunotoxin composed of the tumor-reactive monoclonal antibody B3 and a genetically engineered form of Pseudomonas exotoxin. Monoclonal antibody B3 reacts with a carbohydrate epitope that is found on a number of solid tumors (e.g., breast, ovarian, and lung carcinomas) that frequently invade the intrathecal space, causing neoplastic meningitis. The Pseudomonas exotoxin has been engineered to remove the binding domain to eliminate nonspecific binding. A model of human neoplastic meningitis using rats bearing the human epidermoid carcinoma A431 was used for therapeutic studies of immunotoxin LMB-1. Therapy was initiated 3 days after injection of the tumor cells, which was one third of the median survival time of untreated rats. A single intrathecal injection of 40 microgram increased median survival from 9 days with saline injection to 16 days (78%, P < 0.001), and a single dose of 200 microgram increased median survival to 25 days (188%, P < 0. 001). Three doses of 40 or 200 microgram given on days 3, 6, and 8 significantly increased the median survival of 9.5 days associated with saline injection to 40.5 days (326% increase) and 33.0 days (247% increase), respectively, with two long-term survivors (191-day survival) in each treatment group. LMB-1 had no therapeutic effect on the treatment of two B3 antigen-negative neoplastic meningitis models. Treatment of the antigen-positive A431 neoplastic meningitis with B3 alone or a nonspecific monoclonal, MOPC, coupled to the engineered Pseudomonas exotoxin produced no survival effects. Nontumor-bearing athymic rats showed no toxicity with a single dose of either 40 microgram or 200 microgram, or 3 doses of 40 microgram. However, when they were given three doses of 200 microgram, these rats showed weight loss and loss of neurological function, and two of eight animals died. These studies indicate that, in the range of the most therapeutically effective dosage, the immunotoxin LMB-1 is tolerated in the intrathecal space and should be considered for human intrathecal trials.
- Published
- 1995
44. Intraarterial administration of melphalan for treatment of intracranial human glioma xenografts in athymic rats.
- Author
-
Kurpad SN, Friedman HS, Archer GE, McLendon RE, Petros WM, Fuchs HE, Guaspari A, and Bigner DD
- Subjects
- Animals, Brain drug effects, Brain Neoplasms metabolism, Brain Neoplasms mortality, Dose-Response Relationship, Drug, Drug Screening Assays, Antitumor, Glioma metabolism, Glioma mortality, Humans, Injections, Intra-Arterial, Male, Melphalan pharmacokinetics, Rats, Rats, Nude, Transplantation, Heterologous, Brain Neoplasms drug therapy, Glioma drug therapy, Melphalan administration & dosage
- Abstract
Malignant gliomas will affect 15,000-17,000 Americans each year and carry a dismal prognosis. Adjuvant chemotherapy is hampered by inadequate drug delivery, systemic toxicity, and a markedly variable biological sensitivity. Intraarterial (i.a.) therapy may enhance selectivity by improving tumor drug delivery and reducing systemic toxicity. Using melphalan given i.a., we studied the therapy of intracranial human glioma xenografts in male athymic nude rats (mean weight, 300 g) which were inoculated intracerebrally with D-54 MG and D-456 MG. On Days 6 and 7 (D-54 MG) or Days 9 and 10 (D-456 MG), rats randomized by body weight and treated with single-dose melphalan given i.a. at 0.5 or 0.75 mg produced significantly higher median survival (D-54 MG, Days 33 and 32; D-456 MG, Days 52 and 54, respectively) compared with i.a. saline (D-54 MG, Day 14, P < 0.001; D-456 MG, Day 24, P = 0.000) or melphalan given i.v. at 0.75 mg and 0.9 mg (D-54 MG only; Day 19, P < 0.001; Day 23, P < 0.001, respectively) and at 0.5 and 0.75 mg (D-456 MG only; Day 26 for both doses, P = 0.00). Although a dose-dependent increase in median survival (D-54 MG, 0.25 mg, Day 18; 0.5 mg, Day 28.5; 0.75 mg, Day 32.5) was observed with i.a. administered melphalan, no significant difference was apparent between 0.5 and 0.75 mg in either tumor model (D-54 MG, P = 0.15; D-456 MG, P = 0.37). Toxicity studies in nontumor-bearing athymic rats yielded a maximum tolerated dose of 0.8 mg for i.a. administered melphalan. This dosage was superior in spite of different xenograft permeabilities (apparent mean blood-to-tissue transport [K] values for alpha-aminoisobutyric acid, 5.8 for D-54 MG and 1.3 for D-456 MG). Pharmacokinetic experiments demonstrated a significant first pass advantage for i.a. (versus i.v.) melphalan. The short plasma half-life, marked antiglioma activity, and lack of requirement for metabolic activation indicate that i.a. melphalan holds considerable promise for human glioma therapy.
- Published
- 1995
45. Monoclonal antibodies against EGFRvIII are tumor specific and react with breast and lung carcinomas and malignant gliomas.
- Author
-
Wikstrand CJ, Hale LP, Batra SK, Hill ML, Humphrey PA, Kurpad SN, McLendon RE, Moscatello D, Pegram CN, and Reist CJ
- Subjects
- Amino Acid Sequence, Animals, Antibodies, Monoclonal isolation & purification, Antibodies, Monoclonal metabolism, Antibody Specificity, Base Sequence, Breast Neoplasms immunology, Breast Neoplasms metabolism, ErbB Receptors classification, ErbB Receptors genetics, Female, Glioma immunology, Glioma metabolism, Humans, Immunohistochemistry, Kinetics, Lung Neoplasms immunology, Lung Neoplasms metabolism, Mice, Mice, Inbred BALB C, Molecular Sequence Data, Neoplasm Transplantation, Polymerase Chain Reaction, RNA, Messenger analysis, RNA, Messenger genetics, Transcription, Genetic, Transplantation, Heterologous, Tumor Cells, Cultured, Antibodies, Monoclonal immunology, Breast Neoplasms ultrastructure, ErbB Receptors immunology, Glioma ultrastructure, Lung Neoplasms ultrastructure
- Abstract
Despite molecular biological advances in understanding human cancers, translation into therapy has been less forthcoming; targeting neoplastic cells still requires that tumor-specific markers, preferably those on the cell surface, be identified. The epidermal growth factor receptor (EGFR) exists in a deletion-mutant form, EGFRvIII, which has been identified by genetic and immunological means in a subset of gliomas and non-small cell lung carcinomas. Specific polyvalent antisera to the extracellular portion of the variant were readily induced, but immunization using a synthetic linear peptide representing the unique EGFRvIII primary sequence has been unsuccessful in mice or macaques. We report here five specific monoclonal antibodies (mAbs) developed through long-term immunization protocols using the EGFRvIII-specific synthetic peptide and the intact variant in different formats that maintained secondary and tertiary conformation. These mAbs identify the EGFRvIII on the cell surface with relatively high affinity (KA range, 0.13 to 2.5 x 10(9) M-1) by live cell Scatchard analysis. These mAbs are specific for EGFRvIII as determined by RIA, ELISA, Western blot, analytical flow cytometry, autophosphorylation, and immunohistochemistry. Isolating specific mAbs enabled us to analyze normal and neoplastic human tissue and establish that EGFRvIII is truly tumor specific for subsets of breast carcinomas and for previously reported non-small cell lung carcinomas and gliomas. Also, this receptor is not expressed by any normal human tissues thus far examined, including elements of the peripheral, central nervous, and lymphoid systems. With mAbs, we identified a higher incidence of EGFRvIII positivity in gliomas than previously described and identified an EGFRvIII-positive subset of breast tumors; also, we observed that the EGFRvIII epitope is not expressed in normal tissues, and we demonstrated the localizing and therapeutic potential of the mAbs for tumors expressing this epitope. Our observations strongly warrant development of this mAb-antigen system as therapy for breast, lung, and central nervous system tumors.
- Published
- 1995
46. Intrathecal melphalan therapy of human neoplastic meningitis in athymic nude rats.
- Author
-
Friedman HS, Archer GE, McLendon RE, Schuster JM, Colvin OM, Guaspari A, Blum R, Savina PA, Fuchs HE, and Bigner DD
- Subjects
- Animals, Brain Neoplasms mortality, Demyelinating Diseases chemically induced, Disease Models, Animal, Drug Screening Assays, Antitumor, Female, Glioma mortality, Humans, Injections, Spinal, Melphalan adverse effects, Meningitis etiology, Meningitis mortality, Rats, Rats, Nude, Rhabdomyosarcoma mortality, Subarachnoid Space, Transplantation, Heterologous, Tumor Cells, Cultured, Brain Neoplasms drug therapy, Glioma drug therapy, Melphalan administration & dosage, Meningitis drug therapy, Rhabdomyosarcoma drug therapy
- Abstract
We report the activity and toxicity of intrathecal melphalan in the treatment of human neoplastic meningitis in the subarachnoid space of athymic nude rats. Animals received injections via chronic indwelling subarachnoid catheters with 5 x 10(5) or 5 x 10(6) TE-671 human rhabdomyosarcoma cells or 5 x 10(6) D-54 MG human glioma cells and were treated with melphalan on days 8, 5, or 5, respectively. Melphalan toxicity in nontumor-bearing rats was assessed at single doses of a 2.0, 3.0, 4.0, or 5.0 mM solution, with clinical and histological evidence of neurotoxicity observed at the 4.0 and 5.0 mM levels. Multiple-dose toxicity studies using a dosing schedule of twice a week for two weeks with a 0.25, 0.5, 0.75, 1.0, 1.5, or 2 mM solution revealed dose-dependent clinical and histological evidence for toxicity at all dosages. Treatment of TE-671 with a single dose of 2.0 mM intrathecal melphalan produced an increase in median survival of 442% compared with saline controls (P < 0.003). Comparison of a single dose of 1.0 or 2.0 mM melphalan with a multiple dose regimen at 0.25 or 0.5 mM melphalan in the treatment of TE-671 revealed increases in median survival of 50% for 1.0 mM, 57% for 2.0 mM, 79% for 0.5 mM, and 111% for 0.25 mM concentrations. Comparison of a single dose of 1 mM melphalan with multiple doses of 0.25 mM melphalan in the treatment of D-54 MG revealed an increase in median survival of 475+% for each of the regimens. Intrathecal melphalan may be an important new addition in the treatment of neoplastic meningitis and is currently being evaluated clinically in a Phase 1 trial.
- Published
- 1994
47. Radioimmunotherapy of neoplastic meningitis in rats using an alpha-particle-emitting immunoconjugate.
- Author
-
Zalutsky MR, McLendon RE, Garg PK, Archer GE, Schuster JM, and Bigner DD
- Subjects
- Animals, Antibodies, Monoclonal metabolism, Astatine pharmacokinetics, Female, Humans, Meningitis etiology, Rats, Rats, Nude, Alpha Particles therapeutic use, Antibodies, Monoclonal therapeutic use, Astatine therapeutic use, Meningeal Neoplasms radiotherapy, Meningitis radiotherapy, Radioimmunotherapy methods, Rhabdomyosarcoma radiotherapy
- Abstract
Because of their short range and high linear energy transfer, alpha-particles may be particularly effective in the treatment of neoplastic meningitis. Monoclonal antibody 81C6 was labeled with alpha-particle-emitting 211At using N-succinimidyl3-[211At]astatobenzoate, and the efficacy and toxicity of this immunoconjugate were evaluated in an athymic rat model. Animals were given injections via a chronic indwelling catheter with 5 x 10(5) TE-671 human rhabdomyosarcoma cells and treated 8 days later with single intrathecal doses of either saline or 4-18 microCi of 211At-labeled specific 81C6 antibody or isotype-matched control 211At-labeled 45.6 antibody. In the first experiment, 4, 7, and 13 microCi 211At-labeled 81C6 produced statistically significant (P = 0.004-0.02) increases in median survival of 33, 29, and 51%, respectively, as compared with saline. Two of 10 animals receiving the 13-microCi dose lived for 6 months before being killed for histological analysis. In the second experiment, 12 microCi of 211At-labeled 45.6 did not increase median survival significantly relative to saline control, while 12 microCi of 211At-labeled 81C6 increased median survival by 113% (P < 0.005) and resulted in 33% apparent cures. Five of 10 animals receiving 18 microCi of 211At-labeled 81C6 survived until they were killed at 295 days. An additional study was performed in animals given intrathecal injections of 5 x 10(6) TE-671 cells and given a single dose of 18 microCi of 211At-labeled 81C6 or 211At-labeled 45.6. At this higher cell number, significantly prolonged survival was still seen for specific antibody as compared with saline (P < 0.001) and control antibody (P < 0.05). These results suggest that treatment with 211At-labeled monoclonal antibodies may be a valuable approach for neoplastic meningitis.
- Published
- 1994
48. Alterations of the TP53 gene in human gliomas.
- Author
-
Rasheed BK, McLendon RE, Herndon JE, Friedman HS, Friedman AH, Bigner DD, and Bigner SH
- Subjects
- Adolescent, Adult, Age Factors, Aged, Aged, 80 and over, Brain Neoplasms mortality, Brain Neoplasms pathology, Child, Child, Preschool, Chromosome Aberrations, Chromosomes, Human, Pair 17, Female, Gene Amplification genetics, Gene Deletion, Glioma mortality, Glioma pathology, Heterozygote, Humans, Immunohistochemistry, Infant, Male, Middle Aged, Polymerase Chain Reaction, Brain Neoplasms genetics, Genes, p53 genetics, Glioma genetics, Mutation genetics
- Abstract
Glial tumors of all grades and histological types from 72 adults and 48 children were analyzed for mutations of the TP53 gene, loss of heterozygosity (LOH) for 17p, and accumulation of TP53 protein to determine whether the incidence and type of TP53 alterations differ among tumors of different histological type and between tumors from adults and children. These tumors were also evaluated for LOH for chromosome 10 and for amplification of the epidermal growth factor receptor, C-MYC, N-MYC, GLI, platelet-derived growth factor receptor-alpha, and murine double minute 2 genes to determine the patterns of molecular alterations involved in the progression of these neoplasms. Seventeen of the 120 tumors contained mutations of the TP53 gene. One of the tumors with TP53 gene mutation was from one of the 48 patients less than 18 years of age. Twelve of the 17 tumors with mutations occurred among the 27 patients in the 18-45-year age group, while 4 tumors with mutations were among the 45 patients more than 45 years old. There was also an increased incidence of TP53 mutation in patients with anaplastic astrocytoma histology. However, no significant association between presence of TP53 mutation and patient survival was observed. These studies demonstrate that TP53 gene mutations are a common mechanism for glial cell neoplasms in the 18-45-year age group but are unrelated to progression and advanced histological grade. LOH for chromosome 10 and gene amplification, however, occurring in 82 and 40%, respectively, of glioblastoma multiforme, whether seen alone or along with TP53 gene alterations, are related to advanced histological grade of the tumor. In childhood gliomas, in contrast, TP53 gene alterations, LOH for 17p and 10q, and gene amplification are uncommon in tumors of all grades, suggesting that presently unknown mechanisms are responsible for the genesis and progression of these tumors.
- Published
- 1994
49. Intraarterial therapy of human glioma xenografts in athymic rats using 4-hydroperoxycyclophosphamide.
- Author
-
Schuster JM, Friedman HS, Archer GE, Fuchs HE, McLendon RE, Colvin OM, and Bigner DD
- Subjects
- Animals, Body Weight drug effects, Brain Neoplasms metabolism, Cyclophosphamide pharmacokinetics, Cyclophosphamide pharmacology, Cyclophosphamide toxicity, Dose-Response Relationship, Drug, Glioma metabolism, Humans, Injections, Intra-Arterial, Injections, Intravenous, Male, Mice, Mice, Nude, Rats, Rats, Nude, Transplantation, Heterologous, Brain Neoplasms drug therapy, Cyclophosphamide analogs & derivatives, Glioma drug therapy
- Abstract
The addition of chemotherapy, notably using nitrosoureas, in the treatment of patients with glioblastoma multiforme has resulted in only modest improvements in long-term patient survival over the use of surgical intervention and irradiation alone. Intraarterial (i.a.) chemotherapy offers the potential benefit of increasing tumor drug delivery because of first-pass drug uptake, while minimizing systemic drug levels and toxicity. We have now investigated the i.a. therapy of intracerebral human glioma xenografts in athymic rats with 4-hydroperoxycyclophosphamide (4-HC), a preactivated derivative of cyclophosphamide. Athymic male rats were given intracerebral injections of the human glioma line D-54 MG. On Day 5 after injection, the rats were randomized (n = 8-10) by body weight (mean weight, approximately 300 g). In one set of experiments, each group received either i.v. saline, i.a. saline, 6 mg i.a. 4-HC, 6 mg i.v. 4-HC (6 mg), or 12 mg i.v. 4-HC. Intraarterial 4-HC produced significant increases in median survival (Day 24) compared with i.a. saline controls (140% increase), equivalent doses given i.v. (71% increase), and twice the equivalent dose given i.v. (50% increase) (by Wilcoxon rank sum analysis, P < 0.05 is statistically significant). The i.a. maximum tolerated dose was subsequently determined to be approximately 12.5 mg in non-tumor-bearing rats. Further experiments demonstrated a dose-response increase in survival for i.a. dosages of 6, 9, and 12.5 mg with significant improvement when compared with saline controls and 12.5 mg i.v. Pharmacokinetic experiments also demonstrated a significant first-pass uptake advantage for i.a. (versus i.v.) administered 4-HC. The short plasma half-life and marked antiglioma activity of 4-HC, without the need for hepatic activation, suggest a therapeutic application of this drug in the i.a. treatment of brain tumors.
- Published
- 1993
50. Lactotetraose series ganglioside 3',6'-isoLD1 in tumors of central nervous and other systems in vitro and in vivo.
- Author
-
Wikstrand CJ, Longee DC, McLendon RE, Fuller GN, Friedman HS, Fredman P, Svennerholm L, and Bigner DD
- Subjects
- Animals, Antibodies, Monoclonal isolation & purification, Antibody Specificity, Carbohydrate Sequence, Chromatography, High Pressure Liquid, Female, Humans, Immunohistochemistry, Mice, Mice, Inbred BALB C, Mice, Nude, Molecular Sequence Data, Neoplasm Transplantation, Transplantation, Heterologous, Tumor Cells, Cultured, Central Nervous System Neoplasms chemistry, Gangliosides analysis, Oligosaccharides analysis
- Abstract
Two monoclonal antibodies, DMAb-21 and DMAb-22, directed against the lactotetraose series ganglioside-associated epitope IV3NeuAc,III6-NeuAcLcOse4Cer (3',6'-isoLD1), were found to define the minimum binding epitope NeuAc(or NeuGc)alpha 2-3Gal beta 1-3(NeuAc or NeuGc)alpha 2-6GlcNAc. The distribution of 3',6'-isoLD1 in cultured cell lines and derived xenografts of primary tumors of the human central nervous system and of embryonal or neuroectodermal tumor derivation was determined. Only 4 of 26 cell lines, 3 teratomas and 1 pancreatic adenocarcinoma, expressed detectable 3',6'-isoLD1 when cultured in vitro; none of 14 tested glioma lines, including 2 that expressed the monosialo-precursor IV3NeuAcLcOse4Cer in vitro, expressed detectable levels. Expression of 3',6'-isoLD1 was more frequent when neoplastic cells were grown in xenograft form in athymic mice; 4 of 10 glioma and 2 of 2 teratoma xenograft ganglioside extracts were positive for 3',6'-isoLD1. The absence of 3',6'-isoLD1 in cultured tumor cells of the central nervous system and its proportional increased presence in tumor cells of the same origin grown in vivo further supports previous studies suggesting that ganglioside expression may be modified by environmental forces. The expression of lacto series gangliosides both in vitro and in vivo by teratoma and pancreatic adenocarcinoma cells, as opposed to only in vivo expression by glioma cells, suggests that tissue-specific forces may also exist. Immunohistochemical localization of 3',6'-isoLD1 in frozen sections of primary central nervous system neoplasms including those of glial and nonglial origin was performed; 20 of 30 (67%) of glial tumors were positive. Among nonglial tumors, 21 of 34 (62%) of epithelial cancers were reactive with anti-3',6'-isoLD1 monoclonal antibodies; notably negative were carcinomas of the ovary and lung carcinomas of all subtypes. Lymphomas and infiltrative lymphocytes were uniformly negative. The restriction of 3',6'-isoLD1 expression within the human central nervous system to periods of fetal-neonatal astroglial proliferation, to intense reactive astrocytosis, and to primary neoplasms, and the production of specific monoclonal antibodies to this epitope provide a specific complex for immunolocalization and, eventually, immunotherapy.
- Published
- 1993
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.