Try a new search

Format these results:

Searched for:

person:nicolt01

in-biosketch:true

Total Results:

73


Population Pharmacokinetics of Vemurafenib in Children With Recurrent/Refractory BRAF Gene V600E-Mutant Astrocytomas

Wang, Hechuan; Long-Boyle, Janel; Winger, Beth Apsel; Nicolaides, Theodore; Mueller, Sabine; Prados, Michael; Ivaturi, Vijay
Vemurafenib (Zelboraf) is an orally available BRAFV600E inhibitor approved for the treatment of unresectable or metastatic BRAFV600E -mutant melanoma. The primary objective of this work was to characterize the pharmacokinetics (PK) of vemurafenib in pediatric patients with recurrent/refractory astrocytomas harboring the BRAFV600E mutation. The study was also designed to evaluate the feasibility of replacing whole vemurafenib tablets with crushed tablets in young children unable to swallow tablets. Twenty-five pediatric patients (median age, 8.8 years; range, 3.3-19.2) with recurrent/refractory BRAFV600E -mutant astrocytomas received whole (n = 19) or crushed (n = 6) vemurafenib tablets twice daily. Plasma samples were collected on days 1, 15, and 22 in cycle 1 of vemurafenib treatment. Descriptive PK analyses demonstrated significant variability (approximately 6-fold) in drug exposure. A 1-compartment model with first-order absorption and elimination was developed by adjusting the vemurafenib PK model previously validated in adults with mutant BRAFV600E melanoma. After inclusion of allometric scaling on total body weight, the model adequately described the PK of vemurafenib in children between a wide age range of 3 to 19 years old. In the crushed-tablet cohort, relative bioavailability was approximately 96% (95% confidence interval, 49%-142%) compared to that seen in pediatric patients receiving whole tablets based on the preliminary comparison analysis results. Moderate intrapatient variability (48%) of vemurafenib clearance was observed. There was significant correlation (R2 = 0.83) between area under the plasma concentration-time curve and trough concentration at steady state. These results will help increase the number of pediatric patients for whom vemurafenib is accessible and facilitate improved dosing in pediatric patients with recurrent/refractory BRAFV600E astrocytomas.
PMID: 32476174
ISSN: 1552-4604
CID: 4480872

Exploring DNA Methylation for Prognosis and Analyzing the Tumor Microenvironment in Pleomorphic Xanthoastrocytoma

Tang, Karen; Kurland, David; Vasudevaraja, Varshini; Serrano, Jonathan; Delorenzo, Michael; Radmanesh, Alireza; Thomas, Cheddhi; Spino, Marissa; Gardner, Sharon; Allen, Jeffrey C; Nicolaides, Theodore; Osorio, Diana S; Finlay, Jonathan L; Boué, Daniel R; Snuderl, Matija
Pleomorphic xanthoastrocytoma (PXA) is a rare type of brain tumor that affects children and young adults. Molecular prognostic markers of PXAs remain poorly established. Similar to gangliogliomas, PXAs show prominent immune cell infiltrate, but its composition also remains unknown. In this study, we correlated DNA methylation and BRAF status with clinical outcome and explored the tumor microenvironment. We performed DNA methylation in 21 tumor samples from 18 subjects with a histological diagnosis of PXA. MethylCIBERSORT was used to deconvolute the PXA microenvironment by analyzing the associated immune cell-types. Median age at diagnosis was 16 years (range 7-32). At median follow-up of 30 months, 3-year and 5-year overall survival was 73% and 71%, respectively. Overall survival ranged from 1 to 139 months. Eleven out of 18 subjects (61%) showed disease progression. Progression-free survival ranged from 1 to 89 months. Trisomy 7 and CDKN2A/B (p16) homozygous deletion did not show any association with overall survival (p = 0.67 and p = 0.74, respectively). Decreased overall survival was observed for subjects with tumors lacking the BRAF V600E mutation (p = 0.02). PXAs had significantly increased CD8 T-cell epigenetic signatures compared with previously profiled gangliogliomas (p = 0.0019). The characterization of immune cell-types in PXAs may have implications for future development of immunotherapy.
PMID: 32594172
ISSN: 1554-6578
CID: 4503772

Phase I study of vemurafenib in children with recurrent or progressive BRAFV600E mutant brain tumors: Pacific Pediatric Neuro-Oncology Consortium study (PNOC-002)

Nicolaides, Theodore; Nazemi, Kellie J; Crawford, John; Kilburn, Lindsay; Minturn, Jane; Gajjar, Amar; Gauvain, Karen; Leary, Sarah; Dhall, Girish; Aboian, Mariam; Robinson, Giles; Long-Boyle, Janel; Wang, Hechuan; Molinaro, Annette M; Mueller, Sabine; Prados, Michael
Background: BRAFV600E mutation is present in a subset of pediatric brain tumors. Vemurafenib is an oral, selective ATP-competitive inhibitor of BRAFV600E kinase. The goal of this multi-center study conducted through the Pacific Pediatric Neuro-Oncology Consortium (PNOC) was to determine the recommended phase 2 dose (RP2D) and dose limiting toxicities (DLTs) in children < 18 years with recurrent or progressive BRAFV600E mutant brain tumors. Results: Nineteen eligible patients were enrolled. Eleven patients had received three or more prior therapies. Data reported are from the start of treatment for the first patient (April 30 2014) through August 31 2019. The RP2D was defined as 550 mg/m2 twice daily after DLT criteria adjustment for rash. Related grade ≥ 3 adverse events included secondary keratoacanthoma (n = 1); rash (n =16); and fever (n = 5). Subjects received a median of 23 cycles (range 3-63). Four patients remain on treatment. Centrally reviewed best radiographic responses included 1 complete response, 5 partial responses, and 13 stable disease. The steady-state area under the curve (AUC0-∞median) was 604 mg*h/L (range 329-1052). Methods: Vemurafenib was given starting at 550 mg/m2, twice daily which corresponds to the adult RP2D. Adverse events were graded using the NIH Common Terminology Criteria for Adverse Events (CTCAE) version 4.0. Central imaging review was performed. Pharmacokinetic sampling was performed. Conclusions: Vemurafenib has promising anti-tumor activity in recurrent BRAF V600E-positive brain tumors with manageable toxicity. A phase 2 study is ongoing (NCT01748149).
PMCID:7260122
PMID: 32523649
ISSN: 1949-2553
CID: 4478482

DECONVOLUTING MECHANISMS OF RESISTANCE TO BRAF INHIBITORS IN BRAF V600E HUMAN GLIOMA [Meeting Abstract]

Schreck, Karisa; Morin, Andrew; Zhao, Guisheng; Sanford, Bridget; Green, Adam; Jones, Kenneth; Banerjee, Anurhada; Nazemi, Kellie; Samuel, David; Kilburn, Lindsay; Solomon, David; Pratilas, Christine; Nicolaides, Theodore; Levy, Jean Mulcahy
ISI:000590061300259
ISSN: 1522-8517
CID: 4688142

POTENT, SELECTIVE, AND BRAIN PENETRANT INHIBITORS OF EXTRACELLULAR DOMAIN EGFR ONCOGENIC MUTANTS EXPRESSED IN GBM DEMONSTRATE EFFICACY IN AN INTRACRANIAL PATIENT DERIVED XENOGRAFT MODEL [Meeting Abstract]

O\Connor, Matthew; Lucas, Matt; Romashko, Darlene; Rasmussen, Sara; Fiorenza, Raffaele; Lin, Tai-An; Waters, Nigel; Trainor, George; Flohr, Alexander; Ottaviani, Giorgio; Roberts, Christopher; Buck, Elizabeth; Nicolaides, Theodore; Raleigh, David; Ozawa, Tomoko; Arista, Luca
ISI:000590061300414
ISSN: 1522-8517
CID: 4688172

AN UNUSUAL PRESENTATION OF A PEDIATRIC MIDLINE H3K27M-MUTANT TUMOR WITH DISSEMINATED CRANIOSPINAL LEPTOMENINGEAL DISEASE [Meeting Abstract]

Navarro, Ralph; Golub, Danielle; Hill, Travis; McQuinn, Michelle; Kim, Nora; Tang, Karen; Livingston, Stephanie; Cooper, Benjamin; Gardner, Sharon; Nicolaides, Theodore; William, Christopher; Zagzag, David; Hidalgo, E. Teresa
ISI:000590061300725
ISSN: 1522-8517
CID: 4688202

THE IMMUNE MICROENVIRONMENT IN LOWER GRADE GLIOMAS [Meeting Abstract]

Kumar, Anupam; Chen, Katharine; Petritsch, Claudia; Nicolaides, Theodore; Santi-Vicini, Mariarita; Perry, Arie; Bollen, Andrew; Solomon, David; Molinaro, Annette M.; Phillips, Joanna
ISI:000590061300897
ISSN: 1522-8517
CID: 4688242

Survival Benefit for Individuals With Constitutional Mismatch Repair Deficiency Syndrome Who Undergo a Surveillance Protocol: A Report From the International Replication Repair Deficiency Consortium [Meeting Abstract]

Ercan, A.; Durno, C.; Bianchi, V.; Edwards, M.; Aronson, M.; Villani, A.; Bouffet, E.; Al-Battashi, A.; Alharbi, M.; Basel, D.; Bedgood, R.; Bendel, A.; Blumenthal, D.; Bornhorst, M.; Bronsema, A.; Cairney, E.; Caroll, S.; Chamdin, A.; Chiaravalli, S.; Constantini, S.; Das, A.; Dvir, R.; Farah, R.; Foulkes, W.; Frenkel, Z.; Gardner, S.; Ghalibafian, M.; Gilpin, C.; Goudie, C.; Hamid, S. Ahmer; Hampel, H.; Hansford, J.; Harlos, C.; Hijiya, N.; Saunders, H.; Kamihara, J.; Knipstein, J.; Koschmann, C.; Larouche, V.; Lassaletta, A.; Lindhorst, S.; Ling, S.; Link, M.; DeMola, R. Loret; Luiten, R.; Lurye, M.; Maciaszek, J.; Issai, V. M.; Maher, O.; Massimino, M.; Mushtaq, N.; Newmark, M.; Nicholas, G.; Nichols, K.; Nicolaides, T.; Opocher, E.; Osborn, M.; Oshrine, B.; Pearlman, R.; Pettee, D.; Rapp, J.; Rashid, M.; Reddy, A.; Reichman, L.; Remke, M.; Robbins, G.; Sabel, M.; Samuel, D.; Scheers, I.; Sen, S.; Stearns, D.; Sumerauer, D.; Swallow, C.; Taylor, L.; Toledano, H.; Tomboc, P.; Van Damme, A.; Winer, I.; Yalon, M.; Yen, L. Y.; Zapotocky, M.; Zelcer, S.; Ziegler, D.; Zimmermann, S.; Azad, V. Fallah; Roy, S.; Tabori, U.
ISI:000581769200033
ISSN: 1545-5009
CID: 4696292

Oncogenic mutations at the dimer interface of EGFR lead to formation of covalent homo-dimers and allosteric activation of the kinase domain: A mechanism which alters the selectivity profile of oncogenic EGFR [Meeting Abstract]

Buck, E; O'Connor, M; Flohr, A; Iacone, R; Nicolaides, T; Zhang, J; Mayweg, A V; Epstein, D M
Background: Mutation of either the intracellular catalytic domain or the extracellular domain of the receptor for epidermal growth factor (EGFR) drives oncogenicity. Extracellular domain EGFR mutations are highly expressed in patients with glioblastoma. Despite clinical success with targeting EGFR catalytic site mutants, no drugs have proven effective in glioblastoma patients expressing extracellular EGFR mutations.
Method(s): Herein, we define the molecular mechanism for oncogenic activation of families of extracellular EGFR mutations and reveal how this mechanism renders current generation small molecule ATP-site inhibitors ineffective. We define these types of lesions as allosteric oncogenic mutations.
Result(s): We demonstrate that a group of the most commonly expressed extracellular domain EGFR mutants expressed in glioblastomas is activated by disulfide-bond mediated covalent homodimerization, collectively referred to as locked dimerization (LoDi- EGFR oncogenes). Strikingly, current generation small molecules binding to the active kinase conformation potently inhibit catalytic site mutants, but induce covalent dimerization and activate LoDi-EGFR receptors, manifesting in paradoxical acceleration of proliferation. Herein we describe the discovery of novel ATP competitive small molecules that potently (<50nM) inhibit the family of LoDi-EGFR oncogenes expressed in GBM, and importantly, achieve selectivity versus WT-EGFR of greater than 10-fold.
Conclusion(s): These data demonstrate how the locked-dimer mechanism of EGFR oncogenesis has profound impact on the activity of small molecules acting at the distal catalytic site, providing further evidence for "inside-out" allosteric signaling in EGFR. This provides a mechanistic understanding for the failure of current generation EGFR inhibitors to effectively treat LoDi-EGFR mutants in GBM and sets guidelines for the discovery and development of novel selective LoDi-EGFR inhibitors
EMBASE:630606777
ISSN: 1569-8041
CID: 4286082

Mechanisms of resistance to EGFR inhibition reveal metabolic vulnerabilities in human GBM

McKinney, Andrew; Lindberg, Olle R; Engler, Jane R; Chen, Katharine Y; Kumar, Anupam; Gong, Henry; Lu, Kan V; Simonds, Erin F; Cloughesy, Timothy F; Liau, Linda M; Prados, Michael; Bollen, Andrew W; Berger, Mitchel S; Shieh, Joseph T C; James, C David; Nicolaides, Theodore P; Yong, William H; Lai, Albert; Hegi, Monika E; Weiss, William A; Phillips, Joanna J
Amplification of the epidermal growth factor receptor gene (EGFR) represents one of the most commonly observed genetic lesions in glioblastoma (GBM), however, therapies targeting this signaling pathway have failed clinically. Here, using human tumors, primary patient-derived xenografts (PDXs), and a murine model for GBM, we demonstrate that EGFR inhibition leads to increased invasion of tumor cells. Further, EGFR inhibitor-treated GBM demonstrate altered oxidative stress, with increased lipid peroxidation, and generation of toxic lipid peroxidation products. A tumor cell subpopulation with elevated aldehyde dehydrogenase (ALDH) levels were determined to comprise a significant proportion of the invasive cells observed in EGFR inhibitor-treated GBM. Our analysis of ALDH1A1 protein in newly diagnosed GBM revealed detectable ALDH1A1 expression in 69% (35/51) of the cases, but in relatively low percentages of tumor cells. Analysis of paired human GBM before and after EGFR inhibitor therapy showed an increase in ALDH1A1 expression in EGFR-amplified tumors (p<0.05, n=13 tumor pairs), and in murine GBM ALDH1A1-high clones were more resistant to EGFR inhibition than ALDH1A1-low clones. Our data identify ALDH levels as a biomarker of GBM cells with high invasive potential, altered oxidative stress, and resistance to EGFR inhibition, and reveal a therapeutic target whose inhibition should limit GBM invasion.
PMID: 31270152
ISSN: 1538-8514
CID: 3968222