Network


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

Hotspot


Dive into the research topics where Fariba Navid is active.

Publication


Featured researches published by Fariba Navid.


Cell Reports | 2014

Recurrent somatic structural variations contribute to tumorigenesis in pediatric osteosarcoma.

Xiang Chen; Armita Bahrami; Alberto S. Pappo; John Easton; James Dalton; Erin Hedlund; David W. Ellison; Sheila A. Shurtleff; Gang Wu; Lei Wei; Matthew Parker; Michael Rusch; Panduka Nagahawatte; Jianrong Wu; Shenghua Mao; Kristy Boggs; Heather L. Mulder; Donald Yergeau; Charles Lu; Li Ding; Michael Edmonson; Chunxu Qu; Jianmin Wang; Yongjin Li; Fariba Navid; Najat C. Daw; Elaine R. Mardis; Richard K. Wilson; James R. Downing; Jinghui Zhang

Pediatric osteosarcoma is characterized by multiple somatic chromosomal lesions, including structural variations (SVs) and copy number alterations (CNAs). To define the landscape of somatic mutations in pediatric osteosarcoma, we performed whole-genome sequencing of DNA from 20 osteosarcoma tumor samples and matched normal tissue in a discovery cohort, as well as 14 samples in a validation cohort. Single-nucleotide variations (SNVs) exhibited a pattern of localized hypermutation called kataegis in 50% of the tumors. We identified p53 pathway lesions in all tumors in the discovery cohort, nine of which were translocations in the first intron of the TP53 gene. Beyond TP53, the RB1, ATRX, and DLG2 genes showed recurrent somatic alterations in 29%-53% of the tumors. These data highlight the power of whole-genome sequencing for identifying recurrent somatic alterations in cancer genomes that may be missed using other methods.


Nature Medicine | 2002

Synergism between INK4a / ARF inactivation and aberrant HGF/SF signaling in rhabdomyosarcomagenesis

Richard R. Sharp; Juan A. Recio; Chamelli Jhappan; Toshiyuki Otsuka; Shiquan Liu; Yanlin Yu; Wenjing Liu; Miriam R. Anver; Fariba Navid; Lee J. Helman; Ronald A. DePinho; Glenn Merlino

Rhabdomyosarcoma (RMS) is the most common soft-tissue sarcoma in children, yet molecular events associated with the genesis and progression of this potentially fatal disease are largely unknown. For the molecules and pathways that have been implicated, genetic validation has been impeded by lack of a mouse model of RMS. Here we show that simultaneous loss of Ink4a/Arf function and disruption of c-Met signaling in Ink4a/Arf−/− mice transgenic for hepatocyte growth factor/scatter factor (HGF/SF) induces RMS with extremely high penetrance and short latency. In cultured myoblasts, c-Met activation and Ink4a/Arf loss suppress myogenesis in an additive fashion. Our data indicate that human c-MET and INK4a/ARF, situated at the nexus of pathways regulating myogenic growth and differentiation, represent critical targets in RMS pathogenesis. The marked synergism in mice between aberrant c-Met signaling and Ink4a/Arf inactivation, lesions individually implicated in human RMS, suggests a therapeutic combination to combat this devastating childhood cancer.


Cancer | 2007

Analysis of prognostic factors in ewing sarcoma family of tumors: review of St. Jude Children's Research Hospital studies.

Carlos Rodriguez-Galindo; Tiebin Liu; Matthew J. Krasin; Jianrong Wu; Catherine A. Billups; Najat C. Daw; Sheri L. Spunt; Bhaskar N. Rao; Victor M. Santana; Fariba Navid

Advances in systemic and local therapies have improved outcomes for patients with the Ewing sarcoma family of tumors (ESFT). As new treatments are developed, a critical review of data from past treatment eras is needed to identify clinically relevant risk groups.


Cancer | 2008

Combination of gemcitabine and docetaxel in the treatment of children and young adults with refractory bone sarcoma

Fariba Navid; Jennifer Willert; M. Beth McCarville; Wayne L. Furman; Amy Watkins; William Roberts; Najat C. Daw

The combination of gemcitabine and docetaxel has demonstrated promise in sarcomas diagnosed in adults. In the current study, the toxicity and efficacy of this combination were evaluated in pediatric sarcomas.


Current Cancer Drug Targets | 2010

Anti-GD2 Antibody Therapy for GD2-expressing Tumors

Fariba Navid; Victor M. Santana; Raymond C. Barfield

In the development of novel immune therapies for high-risk cancers, one goal is to find tumor targets that are not widely shared by normal cells. One such target is the surface disialoganglioside GD2. This antigen is expressed on the surface of a variety of tumors for which no curative therapies exist for patients with advanced disease. In childhood, the most common GD2-expressing tumor is neuroblastoma. GD2 is also expressed on several other high-risk tumors, including those of neuroectodermal or epithelial origin, virtually all melanomas, and approximately 50% of tumor samples from osteosarcoma and soft-tissue sarcomas. Because of the tumor-selective expression of this molecule, it is an attractive target for tumor-specific therapies such as antibody therapy. Over the last 2 decades, several anti-GD2 antibodies have been developed. To reduce both the toxicity of the antibody and the development of human anti-mouse antibodies (HAMA), research efforts have primarily focused on exploring anti-GD2 antibodies that have progressively more human elements while at the same time reducing the mouse components. This review will examine antibodies currently undergoing clinical testing as well as the most recent advances to improve antibody therapy for patients with GD2-expressing tumors.


Molecular Cancer Therapeutics | 2006

Pharmacologic inhibition of cyclin-dependent kinase 4/6 activity arrests proliferation in myoblasts and rhabdomyosarcoma-derived cells

Raya Saab; Jennifer L. Bills; Alexander P. Miceli; Colleen Anderson; Joseph D. Khoury; David W. Fry; Fariba Navid; Peter J. Houghton; Stephen X. Skapek

Myoblast cell cycle exit and differentiation are mediated in part by down-regulation of cyclin D1 and associated cyclin-dependent kinase (Cdk) activity. Because rhabdomyosarcoma may represent a malignant tumor composed of myoblast-like cells failing to exit the cell cycle and differentiate, we considered whether excess Cdk activity might contribute to this biology. Cyclin D–dependent Cdk4 and Cdk6 were expressed in most of a panel of six human rhabdomyosarcoma-derived cell lines. Cdk4 was expressed in 73% of alveolar and embryonal rhabdomyosarcoma tumors evaluated using a human tissue microarray. When challenged to differentiate by mitogen deprivation in vitro, mouse C2C12 myoblasts arrested in G1 phase of the cell cycle, whereas four in the panel of rhabdomyosarcoma cell lines failed to do so. C2C12 myoblasts maintained in mitogen-rich media and exposed to a Cdk4/Cdk6 inhibitor PD 0332991 accumulated in G1 cell cycle phase. Similar treatment of rhabdomyosarcoma cell lines caused G1 arrest and prevented cell accumulation in vitro, and it delayed growth of rhabdomyosarcoma xenografts in vivo. Consistent with a role for Cdk4/Cdk6 activity as a regulator of myogenic differentiation, we observed that PD 0332991 exposure promoted morphologic changes and enhanced the expression of muscle-specific proteins in cultured myoblasts and in the Rh30 cell line. Our findings support the concept that pharmacologic inhibition of Cdk4/Cdk6 may represent a useful therapeutic strategy to control cell proliferation and possibly promote myogenic differentiation in rhabdomyosarcoma. [Mol Cancer Ther 2006;5(5):1299–308]


Journal of Clinical Oncology | 2014

Phase I Trial of a Novel Anti-GD2 Monoclonal Antibody, Hu14.18K322A, Designed to Decrease Toxicity in Children With Refractory or Recurrent Neuroblastoma

Fariba Navid; Paul M. Sondel; Raymond C. Barfield; Barry L. Shulkin; Robert A. Kaufman; Jim A. Allay; Jacek Gan; Paul R. Hutson; Songwon Seo; KyungMann Kim; Jacob L. Goldberg; Jacquelyn A. Hank; Catherine A. Billups; Jianrong Wu; Wayne L. Furman; Lisa M. McGregor; Mario Otto; Stephen D. Gillies; Rupert Handgretinger; Victor M. Santana

PURPOSE The addition of immunotherapy, including a combination of anti-GD2 monoclonal antibody (mAb), ch14.18, and cytokines, improves outcome for patients with high-risk neuroblastoma. However, this therapy is limited by ch14.18-related toxicities that may be partially mediated by complement activation. We report the results of a phase I trial to determine the maximum-tolerated dose (MTD), safety profile, and pharmacokinetics of hu14.18K322A, a humanized anti-GD2 mAb with a single point mutation (K322A) that reduces complement-dependent lysis. PATIENTS AND METHODS Eligible patients with refractory or recurrent neuroblastoma received escalating doses of hu14.18K322A ranging from 2 to 70 mg/m(2) per day for 4 consecutive days every 28 days (one course). RESULTS Thirty-eight patients (23 males; median age, 7.2 years) received a median of two courses (range, one to 15). Dose-limiting grade 3 or 4 toxicities occurred in four of 36 evaluable patients and were characterized by cough, asthenia, sensory neuropathy, anorexia, serum sickness, and hypertensive encephalopathy. The most common non-dose-limiting grade 3 or 4 toxicities during course one were pain (68%) and fever (21%). Six of 31 patients evaluable for response by iodine-123 metaiodobenzylguanidine score had objective responses (four complete responses; two partial responses). The first-course pharmacokinetics of hu14.18K322A were best described by a two-compartment linear model. Median hu14.18K322A α (initial phase) and β (terminal phase) half-lives were 1.74 and 21.1 days, respectively. CONCLUSION The MTD, and recommended phase II dose, of hu14.18K322A is 60 mg/m(2) per day for 4 days. Adverse effects, predominately pain, were manageable and improved with subsequent courses.


Journal of Investigative Dermatology | 2015

The Genomic Landscape of Childhood and Adolescent Melanoma

Charles Lu; Jinghui Zhang; Panduka Nagahawatte; John Easton; Seungjae Lee; Zhifa Liu; Li Ding; Matthew A. Wyczalkowski; Marcus B. Valentine; Fariba Navid; Heather L. Mulder; Ruth G. Tatevossian; James Dalton; James Davenport; Zhirong Yin; Michael Edmonson; Michael Rusch; Gang Wu; Yongjin Li; Matthew Parker; Erin Hedlund; Sheila A. Shurtleff; Susana C. Raimondi; Vadodaria Bhavin; Yergeau Donald; Elaine R. Mardis; Richard Wilson; William E. Evans; David W. Ellison; Stanley Pounds

Despite remarkable advances in the genomic characterization of adult melanoma, the molecular pathogenesis of pediatric melanoma remains largely unknown. We analyzed 15 conventional melanomas (CMs), 3 melanomas arising in congenital nevi (CNMs), and 5 spitzoid melanomas (SMs), using various platforms, including whole genome or exome sequencing, the molecular inversion probe assay, and/or targeted sequencing. CMs demonstrated a high burden of somatic single-nucleotide variations (SNVs), with each case containing a TERT promoter (TERT-p) mutation, 13/15 containing an activating BRAF V600 mutation, and >80% of the identified SNVs consistent with UV damage. In contrast, the three CNMs contained an activating NRAS Q61 mutation and no TERT-p mutations. SMs were characterized by chromosomal rearrangements resulting in activated kinase signaling in 40%, and an absence of TERT-p mutations, except for the one SM that succumbed to hematogenous metastasis. We conclude that pediatric CM has a very similar UV-induced mutational spectrum to that found in the adult counterpart, emphasizing the need to promote sun protection practices in early life and to improve access to therapeutic agents being explored in adults in young patients. In contrast, the pathogenesis of CNM appears to be distinct. TERT-p mutations may identify the rare subset of spitzoid melanocytic lesions prone to disseminate.


Pediatric Blood & Cancer | 2007

Desmoplastic small round cell tumor in childhood: the St. Jude Children's Research Hospital experience.

Raya Saab; Joseph D. Khoury; Matthew J. Krasin; Andrew M. Davidoff; Fariba Navid

Desmoplastic small round cell tumor (DSRCT) is a rare, primarily intra‐abdominal tumor that has a poor outcome with current therapies.


Pediatric Hematology and Oncology | 2009

SPERM CRYOPRESERVATION PRACTICES AMONG ADOLESCENT CANCER PATIENTS AT RISK FOR INFERTILITY

James L. Klosky; Mary E. Randolph; Fariba Navid; Heather L. Gamble; Sheri L. Spunt; Monika L. Metzger; Najat C. Daw; E. Brannon Morris; Melissa M. Hudson

To assess sperm cryopreservation among males newly diagnosed with cancer aged 13 years and older, attending oncologists assigned infertility risk (yes/no) to patients and reported whether their patients engaged in sperm cryopreservation. Only 28.1% of informed at-risk patients banked sperm. Utilization of sperm banking was significantly associated with a diagnosis of central nervous system (CNS) malignancy or non-CNS solid tumor diagnosis, higher socioeconomic status, and not being a member of an Evangelical religious group. These results suggest that sperm banking is underutilized among adolescent males newly diagnosed with cancer, and that strategies to increase the engagement in this fertility preservation method are needed.

Collaboration


Dive into the Fariba Navid's collaboration.

Top Co-Authors

Avatar

Andrew M. Davidoff

St. Jude Children's Research Hospital

View shared research outputs
Top Co-Authors

Avatar

Jianrong Wu

St. Jude Children's Research Hospital

View shared research outputs
Top Co-Authors

Avatar

Matthew J. Krasin

St. Jude Children's Research Hospital

View shared research outputs
Top Co-Authors

Avatar

Victor M. Santana

University of Tennessee Health Science Center

View shared research outputs
Top Co-Authors

Avatar

Najat C. Daw

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Bhaskar N. Rao

St. Jude Children's Research Hospital

View shared research outputs
Top Co-Authors

Avatar

Catherine A. Billups

St. Jude Children's Research Hospital

View shared research outputs
Top Co-Authors

Avatar

Wayne L. Furman

St. Jude Children's Research Hospital

View shared research outputs
Top Co-Authors

Avatar

Barry L. Shulkin

St. Jude Children's Research Hospital

View shared research outputs
Researchain Logo
Decentralizing Knowledge