Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where John Glod is active.

Publication


Featured researches published by John Glod.


The New England Journal of Medicine | 2016

Activity of Selumetinib in Neurofibromatosis Type 1–Related Plexiform Neurofibromas

Eva Dombi; Andrea Baldwin; Leigh Marcus; Michael J. Fisher; Brian Weiss; AeRang Kim; Patricia Whitcomb; Staci Martin; Lindsey Aschbacher-Smith; Tilat A. Rizvi; Jianqiang Wu; Rachel Ershler; Pamela L. Wolters; Janet Therrien; John Glod; Jean B. Belasco; Elizabeth K. Schorry; Alessandra Brofferio; Amy J. Starosta; Andrea Gillespie; Austin L. Doyle; Nancy Ratner; Brigitte C. Widemann

BACKGROUNDnEffective medical therapies are lacking for the treatment of neurofibromatosis type 1-related plexiform neurofibromas, which are characterized by elevated RAS-mitogen-activated protein kinase (MAPK) signaling.nnnMETHODSnWe conducted a phase 1 trial of selumetinib (AZD6244 or ARRY-142886), an oral selective inhibitor of MAPK kinase (MEK) 1 and 2, in children who had neurofibromatosis type 1 and inoperable plexiform neurofibromas to determine the maximum tolerated dose and to evaluate plasma pharmacokinetics. Selumetinib was administered twice daily at a dose of 20 to 30 mg per square meter of body-surface area on a continuous dosing schedule (in 28-day cycles). We also tested selumetinib using a mouse model of neurofibromatosis type 1-related neurofibroma. Response to treatment (i.e., an increase or decrease from baseline in the volume of plexiform neurofibromas) was monitored by using volumetric magnetic resonance imaging analysis to measure the change in size of the plexiform neurofibroma.nnnRESULTSnA total of 24 children (median age, 10.9 years; range, 3.0 to 18.5) with a median tumor volume of 1205 ml (range, 29 to 8744) received selumetinib. Patients were able to receive selumetinib on a long-term basis; the median number of cycles was 30 (range, 6 to 56). The maximum tolerated dose was 25 mg per square meter (approximately 60% of the recommended adult dose). The most common toxic effects associated with selumetinib included acneiform rash, gastrointestinal effects, and asymptomatic creatine kinase elevation. The results of pharmacokinetic evaluations of selumetinib among the children in this trial were similar to those published for adults. Treatment with selumetinib resulted in confirmed partial responses (tumor volume decreases from baseline of ≥20%) in 17 of the 24 children (71%) and decreases from baseline in neurofibroma volume in 12 of 18 mice (67%). Disease progression (tumor volume increase from baseline of ≥20%) has not been observed to date. Anecdotal evidence of decreases in tumor-related pain, disfigurement, and functional impairment was observed.nnnCONCLUSIONSnOur early-phase data suggested that children with neurofibromatosis type 1 and inoperable plexiform neurofibromas benefited from long-term dose-adjusted treatment with selumetinib without having excess toxic effects. (Funded by the National Institutes of Health and others; ClinicalTrials.gov number, NCT01362803 .).


Journal of Pediatric Hematology Oncology | 2016

Pediatric brain tumors: Current knowledge and therapeutic opportunities

John Glod; Gilbert J. Rahme; Harpreet Kaur; Eric H. Raabe; Eugene I. Hwang; Mark A. Israel

Great progress has been made in many areas of pediatric oncology. However, tumors of the central nervous system (CNS) remain a significant challenge. A recent explosion of data has led to an opportunity to understand better the molecular basis of these diseases and is already providing a foundation for the pursuit of rationally chosen therapeutics targeting relevant molecular pathways. The molecular biology of pediatric brain tumors is shifting from a singular focus on basic scientific discovery to a platform upon which insights are being translated into therapies.


Cancer Discovery | 2018

Antitumor Activity Associated with Prolonged Persistence of Adoptively Transferred NY-ESO-1c259T Cells in Synovial Sarcoma

Sandra P. D'Angelo; Luca Melchiori; Melinda S. Merchant; Donna B Bernstein; John Glod; Rosandra N. Kaplan; Stephan A. Grupp; William D. Tap; Karen Chagin; Gwendolyn K. Binder; Samik Basu; Daniel E. Lowther; Ruoxi Wang; Natalie Bath; Alex Tipping; Gareth Betts; Jean-Marc Navenot; Hua Zhang; Daniel K. Wells; Erin Van Winkle; Gabor Kari; Trupti Trivedi; Tom Holdich; Lini Pandite; Rafael G. Amado; Crystal L. Mackall

We evaluated the safety and activity of autologous T cells expressing NY-ESO-1c259, an affinity-enhanced T-cell receptor (TCR) recognizing an HLA-A2-restricted NY-ESO-1/LAGE1a-derived peptide, in patients with metastatic synovial sarcoma (NY-ESO-1c259T cells). Confirmed antitumor responses occurred in 50% of patients (6/12) and were characterized by tumor shrinkage over several months. Circulating NY-ESO-1c259T cells were present postinfusion in all patients and persisted for at least 6 months in all responders. Most of the infused NY-ESO-1c259T cells exhibited an effector memory phenotype following ex vivo expansion, but the persisting pools comprised largely central memory and stem-cell memory subsets, which remained polyfunctional and showed no evidence of T-cell exhaustion despite persistent tumor burdens. Next-generation sequencing of endogenous TCRs in CD8+ NY-ESO-1c259T cells revealed clonal diversity without contraction over time. These data suggest that regenerative pools of NY-ESO-1c259T cells produced a continuing supply of effector cells to mediate sustained, clinically meaningful antitumor effects.Significance: Metastatic synovial sarcoma is incurable with standard therapy. We employed engineered T cells targeting NY-ESO-1, and the data suggest that robust, self-regenerating pools of CD8+ NY-ESO-1c259T cells produce a continuing supply of effector cells over several months that mediate clinically meaningful antitumor effects despite prolonged exposure to antigen. Cancer Discov; 8(8); 944-57. ©2018 AACR.See related commentary by Keung and Tawbi, p. 914This article is highlighted in the In This Issue feature, p. 899.


Journal of Digital Imaging | 2017

ENABLE (Exportable Notation and Bookmark List Engine): an Interface to Manage Tumor Measurement Data from PACS to Cancer Databases

Nikhil Goyal; Andrea B. Apolo; Eliana D. Berman; Mohammad Hadi Bagheri; Jason E. Levine; John Glod; Rosandra N. Kaplan; Laura B. Machado; Les R. Folio

Oncologists evaluate therapeutic response in cancer trials based on tumor quantification following selected “target” lesions over time. At our cancer center, a majority of oncologists use Response Evaluation Criteria in Solid Tumors (RECIST) v1.1 quantifying tumor progression based on lesion measurements on imaging. Currently, our oncologists handwrite tumor measurements, followed by multiple manual data transfers; however, our Picture Archiving Communication System (PACS) (Carestream Health, Rochester, NY) has the ability to export tumor measurements, making it possible to manage tumor metadata digitally. We developed an interface, “Exportable Notation and Bookmark List Engine” (ENABLE), which produces prepopulated RECIST v1.1 worksheets and compiles cohort data and data models from PACS measurement data, thus eliminating handwriting and manual data transcription. We compared RECIST v1.1 data from eight patients (16 computed tomography exams) enrolled in an IRB-approved therapeutic trial with ENABLE outputs: 10 data fields with a total of 194 data points. All data in ENABLE’s output matched with the existing data. Seven staff were taught how to use the interface with a 5-min explanatory instructional video. All were able to use ENABLE successfully without additional guidance. We additionally assessed 42 metastatic genitourinary cancer patients with available RECIST data within PACS to produce a best response waterfall plot. ENABLE manages tumor measurements and associated metadata exported from PACS, producing forms and data models compatible with cancer databases, obviating handwriting and the manual re-entry of data. Automation should reduce transcription errors and improve efficiency and the auditing process.


Cancer Chemotherapy and Pharmacology | 2017

A phase I/II trial and pharmacokinetic study of mithramycin in children and adults with refractory Ewing sarcoma and EWS–FLI1 fusion transcript

Patrick J. Grohar; John Glod; Cody J. Peer; Tristan M. Sissung; Fernanda I. Arnaldez; Lauren Long; William D. Figg; Patricia Whitcomb; Lee J. Helman; Brigitte C. Widemann

PurposeIn a preclinical drug screen, mithramycin was identified as a potent inhibitor of the Ewing sarcoma EWS–FLI1 transcription factor. We conducted a phase I/II trial to determine the dose-limiting toxicities (DLT), maximum tolerated dose (MTD), and pharmacokinetics (PK) of mithramycin in children with refractory solid tumors, and the activity in children and adults with refractory Ewing sarcoma.Patients and methodsMithramycin was administered intravenously over 6xa0h once daily for 7xa0days for 28xa0day cycles. Adult patients (phase II) initially received mithramycin at the previously determined recommended dose of 25xa0µg/kg/dose. The planned starting dose for children (phase I) was 17.5xa0µg/kg/dose. Plasma samples were obtained for mithramycin PK analysis.ResultsThe first two adult patients experienced reversible grade 4 alanine aminotransferase (ALT)/aspartate aminotransferase (AST) elevation exceeding the MTD. Subsequent adult patients received mithramycin at 17.5xa0µg/kg/dose, and children at 13xa0µg/kg/dose with dexamethasone pretreatment. None of the four subsequent adult and two pediatric patients experienced cycle 1 DLT. No clinical responses were observed. The average maximal mithramycin plasma concentration in four patients was 17.8xa0±xa04.6xa0ng/mL. This is substantially below the sustained mithramycin concentrations ≥50xa0nmol/L required to suppress EWS–FLI1 transcriptional activity in preclinical studies. Due to inability to safely achieve the desired mithramycin exposure, the trial was closed to enrollment.ConclusionsHepatotoxicity precluded the administration of a mithramycin at a dose required to inhibit EWS–FLI1. Evaluation of mithramycin in patients selected for decreased susceptibility to elevated transaminases may allow for improved drug exposure.


Clinical Cancer Research | 2017

Outcomes of Children and Adolescents with Advanced Hereditary Medullary Thyroid Carcinoma Treated with Vandetanib

Ira Lignugaris Kraft; Srivandana Akshintala; Yuelin J. Zhu; Haiyan Lei; Claudia Derse-Anthony; Eva Dombi; Seth M. Steinberg; Maya Lodish; Steven G. Waguespack; Oxana Borisovna Kapustina; Elizabeth Fox; Frank M. Balis; Maria J. Merino; Paul S. Meltzer; John Glod; Jack F. Shern; Brigitte C. Widemann

Purpose: Vandetanib is well-tolerated in patients with advanced medullary thyroid carcinoma (MTC). Long-term outcomes and mechanisms of MTC progression have not been reported previously. Experimental Design: We monitored toxicities and disease status in patients taking vandetanib for hereditary, advanced MTC. Tumor samples were analyzed for molecular mechanisms of disease progression. Results: Seventeen patients [8 male, age 13 (9–17)* years] enrolled; 16 had a RET p.Met918Thr germline mutation. The duration of vandetanib therapy was 6.1 (0.1–9.7+)* years with treatment ongoing in 9 patients. Best response was partial response in 10, stable disease in 6, and progressive disease in one patient. Duration of response was 7.4 (0.6–8.7+)* and 4.9 (0.6–7.8+)* years in patients with PR and SD, respectively. Six patients died 2.0 (0.4–5.7)* years after progression. Median progression-free survival (PFS) was 6.7 years [95% confidence interval (CI): 2.3 years–undefined] and 5-year overall survival (OS) was 88.2% (95% CI: 60.6%–96.9%). Of 16 patients with a RET p.Met918Thr mutation, progression-free survival was 6.7 years (95% CI: 3.1–undefined) and 5-year overall survival was 93.8% (95% CI: 63.2%–99.1%). No patients terminated treatment because of toxicity. DNA sequencing of tissue samples (n = 11) identified an increase in copy number alterations across the genome as a potential mechanism of drug resistance [*median (range)]. Conclusions: This study demonstrates that vandetanib is safe and results in sustained responses in children and adolescents with hereditary MTC. Our preliminary molecular data suggest that an increase in copy number abnormalities may be associated with tumor progression in hereditary MTC patients treated with vandetanib. Clin Cancer Res; 24(4); 753–65. ©2017 AACR.


The Journal of Pediatrics | 2018

Multiple Endocrine Neoplasia Type 2B Presents Early in Childhood but Often Is Undiagnosed for Years

Angeliki Makri; Srivandana Akshintala; Claudia Derse-Anthony; Brigitte C. Widemann; Constantine A. Stratakis; John Glod; Maya Lodish

&NA; We describe the presenting symptoms and signs of multiple endocrine neoplasia type 2B in a cohort of children. Improved awareness of the early nonendocrine signs of multiple endocrine neoplasia type 2B could lead to earlier diagnosis before the development of medullary thyroid cancer and possibly its metastasis.


The Journal of Clinical Endocrinology and Metabolism | 2018

Pheochromocytoma in children and adolescents with multiple endocrine neoplasia type 2B

Angeliki Makri; Srivandana Akshintala; Claudia Derse-Anthony; Jaydira Del Rivero; Brigitte C. Widemann; Constantine A. Stratakis; John Glod; Maya Lodish

ContextnMultiple endocrine neoplasia type 2B (MEN2B) is characterized by early-onset medullary thyroid cancer in virtually all cases and a 50% lifetime risk of pheochromocytoma (PHEO) development. The literature on PHEO in patients with MEN2B is limited with most data being reported from adult studies that primarily address MEN2A.nnnObjectivenThe aim of the current study is to describe PHEO development in a cohort of pediatric patients with MEN2B.nnnDesignnRetrospective chart review of patients with MEN2B evaluated at the National Institutes of Health in the period between July 2007 and February 2018.nnnResultsnA total of 38 patients were identified (21 males and 17 females). Mean age at MEN2B diagnosis was 10.6 ± 3.9 years. Eight patients (21%) developed PHEO in the course of follow-up to date, all of whom were sporadic cases with the classic M918T RET mutation. PHEO was diagnosed based on biochemical and/or imaging screening studies in five patients, whereas three patients presented with symptoms of excess catecholamines. PHEO was diagnosed at a mean age 15.2 ± 4.6 (range, 10 to 25) years and 4.0 ± 3.3 years after MEN2B diagnosis. Only one patient was diagnosed with PHEO as the initial manifestation of MEN2B after she presented with hypertension and secondary amenorrhea.nnnConclusionnUndiagnosed PHEO can be associated with substantial morbidity. Current American Thyroid Association guidelines recommend PHEO screening starting at age 11 for the high-/highest risk group. The youngest patient diagnosed with PHEO in our cohort was an asymptomatic 10-year-old, suggesting that PHEO development may begin before the screening-recommended age of 11, though remains clinically undetectable and thus the current screening guidelines seem appropriate.


Clinical Endocrinology | 2018

Pregnancy on vandetanib in metastatic medullary thyroid carcinoma associated with multiple endocrine neoplasia type 2B

Nicholas Thomas; John Glod; Claudia Derse-Anthony; Emma L. Baple; Nigel Osborne; Rachel Sturley; Bijay Vaidya; Kate Newbold; Antonia Brooke

Vandetanib is a tyrosine kinase inhibitor (TKI) used in the treatment of medullary thyroid carcinoma occurring in >95% of patients with multiple endocrine neoplasia type 2b (MEN 2b). Pregnancy in women with MEN 2b on vandetanib is previously unreported and has multiple potential implications for both the mother and developing fetus [1]. We describe the case of a 22 year old woman with a background of MEN 2b who was first diagnosed aged 6 presenting with marfanoid habitus, and oral mucosal neuromas. This article is protected by copyright. All rights reserved.


Journal of Clinical Oncology | 2017

Open label, non-randomized, multi-cohort pilot study of genetically engineered NY-ESO-1 specific NY-ESO-1c259t in HLA-A2+ patients with synovial sarcoma (NCT01343043).

Crystal L. Mackall; William D. Tap; John Glod; Mihaela Druta; Warren Chow; Dejka M. Araujo; Stephan A. Grupp; Brian A. Van Tine; Karen Chagin; Erin Van Winkle; Gabor Kari; Trupti Trivedi; Elliot Norry; Tom Holdich; Arundathy N. Bartlett-Pandite; Rafael G. Amado; Sandra P. D'Angelo

Collaboration


Dive into the John Glod's collaboration.

Top Co-Authors

Avatar

Brigitte C. Widemann

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Eva Dombi

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Seth M. Steinberg

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Srivandana Akshintala

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Andrea Baldwin

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Joanne Derdak

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Pamela L. Wolters

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Patricia Whitcomb

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Paul S. Meltzer

National Institutes of Health

View shared research outputs
Researchain Logo
Decentralizing Knowledge