Network


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

Hotspot


Dive into the research topics where Adam C. Resnick is active.

Publication


Featured researches published by Adam C. Resnick.


Science | 2011

The genetic landscape of the childhood cancer medulloblastoma

D. Williams Parsons; Meng Li; Xiaosong Zhang; Siân Jones; Rebecca J. Leary; Jimmy Lin; Simina M. Boca; Hannah Carter; Josue Samayoa; Chetan Bettegowda; Gary L. Gallia; George I. Jallo; Zev A. Binder; Yuri Nikolsky; James Hartigan; Doug Smith; Daniela S. Gerhard; Daniel W. Fults; Scott R. VandenBerg; Mitchel S. Berger; Suely Kazue Nagahashi Marie; Sueli Mieko Oba Shinjo; Carlos Clara; Peter C. Phillips; Jane E. Minturn; Jaclyn A. Biegel; Alexander R. Judkins; Adam C. Resnick; Phillip B. Storm; Tom Curran

Genomic analysis of a childhood cancer reveals markedly fewer mutations than what is typically seen in adult cancers. Medulloblastoma (MB) is the most common malignant brain tumor of children. To identify the genetic alterations in this tumor type, we searched for copy number alterations using high-density microarrays and sequenced all known protein-coding genes and microRNA genes using Sanger sequencing in a set of 22 MBs. We found that, on average, each tumor had 11 gene alterations, fewer by a factor of 5 to 10 than in the adult solid tumors that have been sequenced to date. In addition to alterations in the Hedgehog and Wnt pathways, our analysis led to the discovery of genes not previously known to be altered in MBs. Most notably, inactivating mutations of the histone-lysine N-methyltransferase genes MLL2 or MLL3 were identified in 16% of MB patients. These results demonstrate key differences between the genetic landscapes of adult and childhood cancers, highlight dysregulation of developmental pathways as an important mechanism underlying MBs, and identify a role for a specific type of histone methylation in human tumorigenesis.


Neuro-oncology | 2010

Activating mutations in BRAF characterize a spectrum of pediatric low-grade gliomas

Mariarita Santi; Marcia S. Brose; Changqing Ma; Adam C. Resnick; Angela J. Sievert; Phillip B. Storm; Jaclyn A. Biegel

In the present study, DNA from 27 grade I and grade II pediatric gliomas, including ganglioglioma, desmoplastic infantile ganglioglioma, dysembryoplastic neuroepithelial tumor, and pleomorphic xanthoastrocytoma was analyzed using the Illumina 610K Beadchip SNP-based oligonucleotide array. Several consistent abnormalities, including gain of chromosome 7 and loss of 9p21 were observed. Based on our previous studies, in which we demonstrated BRAF mutations in 3 gangliogliomas, 31 tumors were screened for activating mutations in exons 11 and 15 of the BRAF oncogene or a KIAA1549-BRAF fusion product. There were no cases with a KIAA1549-BRAF fusion. A BRAF V600E mutation was detected in 14 of 31 tumors, which was not correlated with any consistent pattern of aberrations detected by the SNP array analysis. Tumors were also screened for mutations in codon 132 in exon 4 of IDH1, exons 2 and 3 of KRAS, and exons 2-9 of TP53. No mutations in KRAS or TP53 were identified in any of the samples, and there was only 1 IDH1 R132H mutation detected among the sample set. BRAF mutations constitute a major genetic alteration in this histologic group of pediatric brain tumors and may serve as a molecular target for biologically based inhibitors.


Brain Pathology | 2009

Duplication of 7q34 in Pediatric Low-Grade Astrocytomas Detected by High-Density Single-Nucleotide Polymorphism-Based Genotype Arrays Results in a Novel BRAF Fusion Gene

Angela J. Sievert; Eric M. Jackson; Xiaowu Gai; Hakon Hakonarson; Alexander R. Judkins; Adam C. Resnick; Leslie N. Sutton; Phillip B. Storm; Tamim H. Shaikh; Jaclyn A. Biegel

In the present study, DNA from 28 pediatric low‐grade astrocytomas was analyzed using Illumina HumanHap550K single‐nucleotide polymorphism oligonucleotide arrays. A novel duplication in chromosome band 7q34 was identified in 17 of 22 juvenile pilocytic astrocytomas and three of six fibrillary astrocytomas. The 7q34 duplication spans 2.6 Mb of genomic sequence and contains approximately 20 genes, including two candidate tumor genes, HIPK2 and BRAF. There were no abnormalities in HIPK2, and analysis of two mutation hot‐spots in BRAF revealed a V600E mutation in only one tumor without the duplication. Fluorescence in situ hybridization confirmed the 7q34 copy number change and was suggestive of a tandem duplication. Reverse transcription polymerase chain reaction‐based sequencing revealed a fusion product between KIAA1549 and BRAF. The predicted fusion product includes the BRAF kinase domain and lacks the auto‐inhibitory N‐terminus. Western blot analysis revealed phosphorylated mitogen‐activated protein kinase (MAPK) protein in tumors with the duplication, consistent with BRAF‐induced activation of the pathway. Further studies are required to determine the role of this fusion gene in downstream MAPK signaling and its role in development of pediatric low‐grade astrocytomas.


Nature Genetics | 2014

Exome sequencing identifies BRAF mutations in papillary craniopharyngiomas

Priscilla K. Brastianos; Amaro Taylor-Weiner; Peter Manley; Robert T. Jones; Dora Dias-Santagata; Aaron R. Thorner; Michael S. Lawrence; Fausto J. Rodriguez; Lindsay A. Bernardo; Laura Schubert; Ashwini Sunkavalli; Nick Shillingford; Monica L. Calicchio; Hart G.W. Lidov; Hala Taha; Maria Martinez-Lage; Mariarita Santi; Phillip B. Storm; John Y. K. Lee; James N. Palmer; Nithin D. Adappa; R. Michael Scott; Ian F. Dunn; Edward R. Laws; Chip Stewart; Keith L. Ligon; Mai P. Hoang; Paul Van Hummelen; William C. Hahn; David N. Louis

Craniopharyngiomas are epithelial tumors that typically arise in the suprasellar region of the brain. Patients experience substantial clinical sequelae from both extension of the tumors and therapeutic interventions that damage the optic chiasm, the pituitary stalk and the hypothalamic area. Using whole-exome sequencing, we identified mutations in CTNNB1 (β-catenin) in nearly all adamantinomatous craniopharyngiomas examined (11/12, 92%) and recurrent mutations in BRAF (resulting in p.Val600Glu) in all papillary craniopharyngiomas (3/3, 100%). Targeted genotyping revealed BRAF p.Val600Glu in 95% of papillary craniopharyngiomas (36 of 39 tumors) and mutation of CTNNB1 in 96% of adamantinomatous craniopharyngiomas (51 of 53 tumors). The CTNNB1 and BRAF mutations were clonal in each tumor subtype, and we detected no other recurrent mutations or genomic aberrations in either subtype. Adamantinomatous and papillary craniopharyngiomas harbor mutations that are mutually exclusive and clonal. These findings have important implications for the diagnosis and treatment of these neoplasms.


Proceedings of the National Academy of Sciences of the United States of America | 2007

Protein pyrophosphorylation by inositol pyrophosphates is a posttranslational event

Rashna Bhandari; Adolfo Saiardi; Yousef Ahmadibeni; Adele M. Snowman; Adam C. Resnick; Troels Z. Kristiansen; Henrik Molina; Akhilesh Pandey; J. Kent Werner; Krishna R. Juluri; Yong Xu; Glenn D. Prestwich; Keykavous Parang; Solomon H. Snyder

In a previous study, we showed that the inositol pyrophosphate diphosphoinositol pentakisphosphate (IP7) physiologically phosphorylates mammalian and yeast proteins. We now report that this phosphate transfer reflects pyrophosphorylation. Thus, proteins must be prephosphorylated by ATP to prime them for IP7 phosphorylation. IP7 phosphorylates synthetic phosphopeptides but not if their phosphates have been masked by methylation or pyrophosphorylation. Moreover, IP7 phosphorylated peptides are more acid-labile and more resistant to phosphatases than ATP phosphorylated peptides, indicating a different type of phosphate bond. Pyrophosphorylation may represent a novel mode of signaling to proteins.


Proceedings of the National Academy of Sciences of the United States of America | 2013

Paradoxical activation and RAF inhibitor resistance of BRAF protein kinase fusions characterizing pediatric astrocytomas

Angela J. Sievert; Shih-Shan Lang; Katie Boucher; Peter J. Madsen; Erin Slaunwhite; Namrata Choudhari; Meghan Kellet; Phillip B. Storm; Adam C. Resnick

Astrocytomas are the most common type of brain tumors in children. Activated BRAF protein kinase mutations are characteristic of pediatric astrocytomas with KIAA1549-BRAF fusion genes typifying low-grade astrocytomas and V600EBRAF alterations characterizing distinct or higher-grade tumors. Recently, BRAF-targeted therapies, such as vemurafenib, have shown great promise in treating V600E-dependent melanomas. Like V600EBRAF, BRAF fusion kinases activate MAPK signaling and are sufficient for malignant transformation; however, here we characterized the distinct mechanisms of action of KIAA1549-BRAF and its differential responsiveness to PLX4720, a first-generation BRAF inhibitor and research analog of vemurafenib. We found that in cells expressing KIAA1549-BRAF, the fusion kinase functions as a homodimer that is resistant to PLX4720 and accordingly is associated with CRAF-independent paradoxical activation of MAPK signaling. Mutagenesis studies demonstrated that KIAA1549-BRAF fusion-mediated signaling is diminished with disruption of the BRAF kinase dimer interface. In addition, the KIAA1549-BRAF fusion displays increased binding affinity to kinase suppressor of RAS (KSR), an RAF relative recently demonstrated to facilitate MEK phosphorylation by BRAF. Despite its resistance to PLX4720, the KIAA1549-BRAF fusion is responsive to a second-generation selective BRAF inhibitor that, unlike vemurafenib, does not induce activation of wild-type BRAF. Our data support the development of targeted treatment paradigms for BRAF-altered pediatric astrocytomas and also demonstrate that therapies must be tailored to the specific mutational context and distinct mechanisms of action of the mutant kinase.


Cell Metabolism | 2011

Amino Acid Signaling to mTOR Mediated by Inositol Polyphosphate Multikinase

Seyun Kim; Sangwon F. Kim; David Maag; Micah Maxwell; Adam C. Resnick; Krishna R. Juluri; Anutosh Chakraborty; Michael A. Koldobskiy; Seung Hun Cha; Roxanne K. Barrow; Adele M. Snowman; Solomon H. Snyder

mTOR complex 1 (mTORC1; mammalian target of rapamycin [mTOR] in complex with raptor) is a key regulator of protein synthesis and cell growth in response to nutrient amino acids. Here we report that inositol polyphosphate multikinase (IPMK), which possesses both inositol phosphate kinase and lipid kinase activities, regulates amino acid signaling to mTORC1. This regulation is independent of IPMKs catalytic function, instead reflecting its binding with mTOR and raptor, which maintains the mTOR-raptor association. Thus, IPMK appears to be a physiologic mTOR cofactor, serving as a determinant of mTORC1 stability and amino acid-induced mTOR signaling. Substances that block IPMK-mTORC1 binding may afford therapeutic benefit in nutrient amino acid-regulated conditions such as obesity and diabetes.


Proceedings of the National Academy of Sciences of the United States of America | 2001

FKBP12, the 12-kDa FK506-binding protein, is a physiologic regulator of the cell cycle

Bahman Aghdasi; Keqiang Ye; Adam C. Resnick; Alex Y. Huang; Hyo Chol Ha; Xin Guo; Ted M. Dawson; Valina L. Dawson; Solomon H. Snyder

FKBP12, the 12-kDa FK506-binding protein, is a ubiquitous abundant protein that acts as a receptor for the immunosuppressant drug FK506, binds tightly to intracellular calcium release channels and to the transforming growth factor β (TGF-β) type I receptor. We now demonstrate that cells from FKBP12-deficient (FKBP12−/−) mice manifest cell cycle arrest in G1 phase and that these cells can be rescued by FKBP12 transfection. This arrest is mediated by marked augmentation of p21(WAF1/CIP1) levels, which cannot be further augmented by TGF-β1. The p21 up-regulation and cell cycle arrest derive from the overactivity of TGF-β receptor signaling, which is normally inhibited by FKBP12. Cell cycle arrest is prevented by transfection with a dominant-negative TGF-β receptor construct. TGF-β receptor signaling to gene expression can be mediated by SMAD, p38, and ERK/MAP kinase (extracellular signal-regulated kinase/mitogen-activated protein kinase) pathways. SMAD signaling is down-regulated in FKBP12−/− cells. Inhibition of ERK/MAP kinase fails to affect p21 up-regulation. By contrast, activated phosphorylated p38 is markedly augmented in FKBP12−/− cells and the p21 up-regulation is prevented by an inhibitor of p38. Thus, FKBP12 is a physiologic regulator of cell cycle acting by normally down-regulating TGF-β receptor signaling.


Proceedings of the National Academy of Sciences of the United States of America | 2008

Gene deletion of inositol hexakisphosphate kinase 1 reveals inositol pyrophosphate regulation of insulin secretion, growth, and spermiogenesis

Rashna Bhandari; Krishna R. Juluri; Adam C. Resnick; Solomon H. Snyder

Inositol pyrophosphates, also designated inositol diphosphates, possess high-energy β-phosphates that can pyrophosphorylate proteins and regulate various cellular processes. They are formed by a family of inositol hexakisphosphate kinases (IP6Ks). We have created mice with a targeted deletion of IP6K1 in which production of inositol pyrophosphates is markedly diminished. Defects in the mutants indicate important roles for IP6K1 and inositol pyrophosphates in several physiological functions. Male mutant mice are sterile with defects in spermiogenesis. Mutant mice are smaller than wild-type despite normal food intake. The mutants display markedly lower circulating insulin.


Neuro-oncology | 2014

Phase II study of sorafenib in children with recurrent or progressive low-grade astrocytomas

Matthias A. Karajannis; Geneviève Legault; Michael J. Fisher; Sarah Milla; Kenneth J. Cohen; Jeffrey H. Wisoff; D. Harter; Judith D. Goldberg; Tsivia Hochman; Amanda Merkelson; Michael C. Bloom; Angela J. Sievert; Adam C. Resnick; Girish Dhall; David T. W. Jones; Andrey Korshunov; Stefan M. Pfister; Charles G. Eberhart; David Zagzag; Jeffrey C. Allen

BACKGROUND Activation of the RAS-RAF-MEK-ERK signaling pathway is thought to be the key driver of pediatric low-grade astrocytoma (PLGA) growth. Sorafenib is a multikinase inhibitor targeting BRAF, VEGFR, PDGFR, and c-kit. This multicenter phase II study was conducted to determine the response rate to sorafenib in patients with recurrent or progressive PLGA. METHODS Key eligibility criteria included age ≥ 2 years, progressive PLGA evaluable on MRI, and at least one prior chemotherapy treatment. Sorafenib was administered twice daily at 200 mg/m(2)/dose (maximum of 400 mg/dose) in continuous 28-day cycles. MRI, including 3-dimensional volumetric tumor analysis, was performed every 12 weeks. BRAF molecular testing was performed on tumor tissue when available. RESULTS Eleven patients, including 3 with neurofibromatosis type 1 (NF1), were evaluable for response; 5 tested positive for BRAF duplication. Nine patients (82%) came off trial due to radiological tumor progression after 2 or 3 cycles, including 3 patients with confirmed BRAF duplication. Median time to progression was 2.8 months (95% CI, 2.1-31.0 months). Enrollment was terminated early due to this rapid and unexpectedly high progression rate. Tumor tissue obtained from 4 patients after termination of the study showed viable pilocytic or pilomyxoid astrocytoma. CONCLUSIONS Sorafenib produced unexpected and unprecedented acceleration of tumor growth in children with PLGA, irrespective of NF1 or tumor BRAF status. In vitro studies with sorafenib indicate that this effect is likely related to paradoxical ERK activation. Close monitoring for early tumor progression should be included in trials of novel agents that modulate signal transduction.

Collaboration


Dive into the Adam C. Resnick's collaboration.

Top Co-Authors

Avatar

Angela J. Waanders

Children's Hospital of Philadelphia

View shared research outputs
Top Co-Authors

Avatar

Phillip B. Storm

Children's Hospital of Philadelphia

View shared research outputs
Top Co-Authors

Avatar

Sabine Mueller

University of California

View shared research outputs
Top Co-Authors

Avatar

Payal Jain

Children's Hospital of Philadelphia

View shared research outputs
Top Co-Authors

Avatar

Javad Nazarian

Children's National Medical Center

View shared research outputs
Top Co-Authors

Avatar

Pichai Raman

Children's Hospital of Philadelphia

View shared research outputs
Top Co-Authors

Avatar

Angela J. Sievert

Children's Hospital of Philadelphia

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Namrata Choudhari

Children's Hospital of Philadelphia

View shared research outputs
Top Co-Authors

Avatar

Yuankun Zhu

Children's Hospital of Philadelphia

View shared research outputs
Researchain Logo
Decentralizing Knowledge