Network


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

Hotspot


Dive into the research topics where Sara M. Federico is active.

Publication


Featured researches published by Sara M. Federico.


JAMA | 2012

Association of Age at Diagnosis and Genetic Mutations in Patients With Neuroblastoma

Nai-Kong V. Cheung; Jinghui Zhang; Charles Lu; Matthew Parker; Armita Bahrami; Satish K. Tickoo; Adriana Heguy; Alberto S. Pappo; Sara M. Federico; James T. Dalton; Irene Y. Cheung; Li Ding; Robert S. Fulton; Jianmin Wang; Xiang Chen; Jared Becksfort; Jianrong Wu; Catherine A. Billups; David W. Ellison; Elaine R. Mardis; Richard Wilson; James R. Downing; Michael A. Dyer

CONTEXT Neuroblastoma is diagnosed over a wide age range from birth through young adulthood, and older age at diagnosis is associated with a decline in survivability. OBJECTIVE To identify genetic mutations that are associated with age at diagnosis in patients with metastatic neuroblastoma. DESIGN, SETTING, AND PATIENTS Whole genome sequencing was performed on DNA from diagnostic tumors and their matched germlines from 40 patients with metastatic neuroblastoma obtained between 1987 and 2009. Age groups at diagnosis included infants (0-<18 months), children (18 months-<12 years), and adolescents and young adults (≥12 years). To confirm the findings from this discovery cohort, validation testing using tumors from an additional 64 patients obtained between 1985 and 2009 also was performed. Formalin-fixed, paraffin-embedded tumor tissue was used for immunohistochemistry and fluorescence in situ hybridization. Telomere lengths were analyzed using whole genome sequencing data, quantitative polymerase chain reaction, and fluorescent in situ hybridization. MAIN OUTCOME MEASURE Somatic recurrent mutations in tumors from patients with neuroblastoma correlated with the age at diagnosis and telomere length. RESULTS In the discovery cohort (n = 40), mutations in the ATRX gene were identified in 100% (95% CI, 50%-100%) of tumors from patients in the adolescent and young adult group (5 of 5), in 17% (95% CI, 7%-36%) of tumors from children (5 of 29), and 0% (95% CI, 0%-40%) of tumors from infants (0 of 6). In the validation cohort (n = 64), mutations in the ATRX gene were identified in 33% (95% CI, 17%-54%) of tumors from patients in the adolescent and young adult group (9 of 27), in 16% (95% CI, 6%-35%) of tumors from children (4 of 25), and in 0% (95% CI, 0%-24%) of tumors from infants (0 of 12). In both cohorts (N = 104), mutations in the ATRX gene were identified in 44% (95% CI, 28%-62%) of tumors from patients in the adolescent and young adult group (14 of 32), in 17% (95% CI, 9%-29%) of tumors from children (9 of 54), and in 0% (95% CI, 0%-17%) of tumors from infants (0 of 18). ATRX mutations were associated with an absence of the ATRX protein in the nucleus and with long telomeres. CONCLUSION ATRX mutations were associated with age at diagnosis in children and young adults with stage 4 neuroblastoma. TRIAL REGISTRATION clinicaltrials.gov Identifier: NCT00588068.


Cancer Cell | 2013

Targeting Oxidative Stress in Embryonal Rhabdomyosarcoma

Xiang Chen; Elizabeth Stewart; Anang A. Shelat; Chunxu Qu; Armita Bahrami; Mark E. Hatley; Gang Wu; Cori Bradley; Justina McEvoy; Alberto S. Pappo; Sheri L. Spunt; Marcus B. Valentine; Virginia Valentine; Fred Krafcik; Walter H. Lang; Monika Wierdl; Lyudmila Tsurkan; Viktor Tolleman; Sara M. Federico; Chris Morton; Charles Lu; Li Ding; John Easton; Michael Rusch; Panduka Nagahawatte; Jianmin Wang; Matthew Parker; Lei Wei; Erin Hedlund; David Finkelstein

Rhabdomyosarcoma is a soft-tissue sarcoma with molecular and cellular features of developing skeletal muscle. Rhabdomyosarcoma has two major histologic subtypes, embryonal and alveolar, each with distinct clinical, molecular, and genetic features. Genomic analysis shows that embryonal tumors have more structural and copy number variations than alveolar tumors. Mutations in the RAS/NF1 pathway are significantly associated with intermediate- and high-risk embryonal rhabdomyosarcomas (ERMS). In contrast, alveolar rhabdomyosarcomas (ARMS) have fewer genetic lesions overall and no known recurrently mutated cancer consensus genes. To identify therapeutics for ERMS, we developed and characterized orthotopic xenografts of tumors that were sequenced in our study. High-throughput screening of primary cultures derived from those xenografts identified oxidative stress as a pathway of therapeutic relevance for ERMS.


Cancer Research | 2011

Targeting the p53 Pathway in Retinoblastoma with Subconjunctival Nutlin-3a

Rachel Brennan; Sara M. Federico; Cori Bradley; Jiakun Zhang; Jacqueline Flores-Otero; Matthew W. Wilson; Clinton F. Stewart; Fangyi Zhu; Kip Guy; Michael A. Dyer

Retinoblastoma is a rare childhood cancer of the retina that begins in utero and is diagnosed in the first years of life. The goals of retinoblastoma treatment are ocular salvage, vision preservation, and reduction of short- and long-term side effects without risking mortality because of tumor dissemination. To identify better chemotherapeutic combinations for the treatment of retinoblastoma, several groups have developed genetic mouse models and orthotopic xenograft models of human retinoblastoma for preclinical testing. Previous studies have implicated the MDMX protein in the suppression of the p53 pathway in retinoblastoma and shown that the MDM2/MDMX antagonist, Nutlin-3a, can efficiently induce p53-mediated cell death in retinoblastoma cell lines. However, Nutlin-3a cannot be administered systemically to treat retinoblastoma, because it has poor penetration across the blood-ocular barrier. Therefore, we developed an ocular formulation of Nutlin-3a, Nutlin-3a(OC), and tested the pharmacokinetics and efficacy of this new formulation in genetic and human retinoblastoma orthotopic xenograft models of retinoblastoma. Here, we show that Nutlin-3a(OC) specifically and efficiently targets the p53 pathway and that the combination of Nutlin-3a(OC) with systemic topotecan is a significantly better treatment for retinoblastoma than currently used chemotherapy in human orthotopic xenografts. Our studies provide a new standardized approach to evaluate and prioritize novel agents for incorporation into future clinical trials for retinoblastoma.


PLOS ONE | 2011

Preclinical models for neuroblastoma: establishing a baseline for treatment.

Tal Teitz; Jennifer Stanke; Sara M. Federico; Cori Bradley; Rachel Brennan; Jiakun Zhang; Melissa Johnson; Jan Sedlacik; Madoka Inoue; Ziwei M. Zhang; Sharon Frase; Jerold E. Rehg; Claudia M. Hillenbrand; David Finkelstein; Christopher Calabrese; Michael A. Dyer; Jill M. Lahti

Background Preclinical models of pediatric cancers are essential for testing new chemotherapeutic combinations for clinical trials. The most widely used genetic model for preclinical testing of neuroblastoma is the TH-MYCN mouse. This neuroblastoma-prone mouse recapitulates many of the features of human neuroblastoma. Limitations of this model include the low frequency of bone marrow metastasis, the lack of information on whether the gene expression patterns in this system parallels human neuroblastomas, the relatively slow rate of tumor formation and variability in tumor penetrance on different genetic backgrounds. As an alternative, preclinical studies are frequently performed using human cell lines xenografted into immunocompromised mice, either as flank implant or orthtotopically. Drawbacks of this system include the use of cell lines that have been in culture for years, the inappropriate microenvironment of the flank or difficult, time consuming surgery for orthotopic transplants and the absence of an intact immune system. Principal Findings Here we characterize and optimize both systems to increase their utility for preclinical studies. We show that TH-MYCN mice develop tumors in the paraspinal ganglia, but not in the adrenal, with cellular and gene expression patterns similar to human NB. In addition, we present a new ultrasound guided, minimally invasive orthotopic xenograft method. This injection technique is rapid, provides accurate targeting of the injected cells and leads to efficient engraftment. We also demonstrate that tumors can be detected, monitored and quantified prior to visualization using ultrasound, MRI and bioluminescence. Finally we develop and test a “standard of care” chemotherapy regimen. This protocol, which is based on current treatments for neuroblastoma, provides a baseline for comparison of new therapeutic agents. Significance The studies suggest that use of both the TH-NMYC model of neuroblastoma and the orthotopic xenograft model provide the optimal combination for testing new chemotherapies for this devastating childhood cancer.


Pediatric Blood & Cancer | 2013

Comparison of PET-CT and Conventional Imaging in Staging Pediatric Rhabdomyosarcoma

Sara M. Federico; Sheri L. Spunt; Matthew J. Krasin; Catherine A. Billup; Jianrong Wu; Barry L. Shulkin; Gerald Mandell; M. Beth McCarville

Over the past decade, PET–CT has been used to assess rhabdomyosarcoma (RMS) in children. However, the role of PET–CT in staging RMS is unknown.


Cancer | 2011

Subconjunctival carboplatin and systemic topotecan treatment in preclinical models of retinoblastoma

Katie Nemeth; Sara M. Federico; Angel M. Carcaboso; Ying Shen; Paula Schaiquevich; Jiakun Zhang; Merrill J. Egorin; Clinton F. Stewart; Michael A. Dyer

The authors demonstrated previously that the combination of topotecan (TPT) and carboplatin (CBP) was more effective than current chemotherapeutic combinations used to treat retinoblastoma in an orthotopic xenograft model. However, systemic coadministration of these agents is not ideal, because both agents cause dose‐limiting myelosuppression in children.


Nature | 2017

Orthotopic patient-derived xenografts of paediatric solid tumours

Elizabeth Stewart; Sara M. Federico; Xiang Chen; Anang A. Shelat; Cori Bradley; Brittney Gordon; Åsa Karlström; Nathaniel R. Twarog; Michael R. Clay; Armita Bahrami; Burgess B. Freeman; Beisi Xu; Xin Zhou; Jianrong Wu; Victoria Honnell; Monica Ocarz; Kaley Blankenship; Jason Dapper; Elaine R. Mardis; Richard Wilson; James R. Downing; Jinghui Zhang; John Easton; Alberto S. Pappo; Michael A. Dyer

Paediatric solid tumours arise from endodermal, ectodermal, or mesodermal lineages. Although the overall survival of children with solid tumours is 75%, that of children with recurrent disease is below 30%. To capture the complexity and diversity of paediatric solid tumours and establish new models of recurrent disease, here we develop a protocol to produce orthotopic patient-derived xenografts at diagnosis, recurrence, and autopsy. Tumour specimens were received from 168 patients, and 67 orthotopic patient-derived xenografts were established for 12 types of cancer. The origins of the patient-derived xenograft tumours were reflected in their gene-expression profiles and epigenomes. Genomic profiling of the tumours, including detailed clonal analysis, was performed to determine whether the clonal population in the xenograft recapitulated the patient’s tumour. We identified several drug vulnerabilities and showed that the combination of a WEE1 inhibitor (AZD1775), irinotecan, and vincristine can lead to complete response in multiple rhabdomyosarcoma orthotopic patient-derived xenografts tumours in vivo.


Developmental Biology | 2016

The Childhood Solid Tumor Network: A new resource for the developmental biology and oncology research communities

Elizabeth Stewart; Sara M. Federico; Åsa Karlström; Anang A. Shelat; András Sablauer; Alberto S. Pappo; Michael A. Dyer

Significant advances have been made over the past 25 years in our understanding of the most common adult solid tumors such as breast, colon, lung and prostate cancer. Much less is known about childhood solid tumors because they are rare and because they originate in developing organs during fetal development, childhood and adolescence. It can be very difficult to study the cellular origins of pediatric solid tumors in developing organs characterized by rapid proliferative expansion, growth factor signaling, developmental angiogenesis, programmed cell death, tissue reorganization and cell migration. Not only has the etiology of pediatric cancer remained elusive because of their developmental origins, but it also makes it more difficult to treat. Molecular targeted therapeutics that alter developmental pathway signaling may have devastating effects on normal organ development. Therefore, basic research focused on the mechanisms of development provides an essential foundation for pediatric solid tumor translational research. In this article, we describe new resources available for the developmental biology and oncology research communities. In a companion paper, we present the detailed characterization of an orthotopic xenograft of a pediatric solid tumor derived from sympathoadrenal lineage during development.


Journal of Pediatric Hematology Oncology | 2015

Subsequent Malignant Neoplasms in Pediatric Patients Initially Diagnosed with Neuroblastoma

Sara M. Federico; Heather B. Allewelt; Sheri L. Spunt; Melissa M. Hudson; Jianrong Wu; Catherine A. Billups; Jesse J. Jenkins; Victor M. Santana; Wayne L. Furman; Lisa M. McGregor

Background: Most prior studies evaluating subsequent malignant neoplasms (SMNs) in patients with neuroblastoma are restricted to long-term survivors and/or their treatment exposures. This study investigates SMNs in patients diagnosed with neuroblastoma at our institution. Methods: Records of 646 patients treated for neuroblastoma at St Jude Children’s Research Hospital between 1961 and 2005 were reviewed. Data from patients with SMNs were analyzed and the 20- and 30-year cumulative incidence of SMNs and standardized incidence ratio were calculated. Results: Twenty-one patients had a SMN. The 20- and 30-year cumulative incidences of a SMN were 2.6%±0.7% and 4.6%±1.1%, respectively. The standardized incidence ratio was 8.3 (95% confidence interval, 5.0-13.0). Five patients developed a SMN within 5 years from diagnosis. The median latency for the development of acute myeloid leukemia/myelodysplastic syndrome (n=4), sarcomas (n=7), and carcinomas (n=5) were 3.6, 9, and 24.2 years, respectively. Nine patients died from their SMN, including all with acute myeloid leukemia/myelodysplastic syndrome. Conclusions: Patients with neuroblastoma have an increased risk of secondary neoplasia. Modification of risk-adapted therapies will likely alter the affected patient population and the incidence of SMNs. Future studies are necessary to link SMNs to treatment exposures and to evaluate the risk of SMNs beyond 30 years from diagnosis.


Developmental Biology | 2015

Development and characterization of a human orthotopic neuroblastoma xenograft

Elizabeth Stewart; Anang A. Shelat; Cori Bradley; Xiang Chen; Sara M. Federico; Suresh Thiagarajan; Abbas Shirinifard; Armita Bahrami; Alberto S. Pappo; Chunxu Qu; David Finkelstein; András Sablauer; Michael A. Dyer

Neuroblastoma is a pediatric cancer of the developing sympathoadrenal lineage. The tumors are known to develop from the adrenal gland or paraspinal ganglia and have molecular and cellular features of sympathetic neurons such as dense core vesicles and catecholamine production. Here we present the detailed molecular, cellular, genetic and epigenetic characterization of an orthotopic xenograft derived from a high-risk stage 4 neuroblastoma patient. Overall, the xenografted tumor retained the high risk features of the primary tumor and showed aggressive growth and metastasis in the mouse. Also, the genome was preserved with no additional copy number variations, structural variations or aneuploidy. There were 13 missense mutations identified in the xenograft that were not present in the patient’s primary tumor and there were no new nonsense mutations. None of the missense mutations acquired in the xenograft were in known cancer genes. We also demonstrate the feasibility of using the orthotopic neuroblastoma xenograft to test standard of care chemotherapy and molecular targeted therapeutics. Finally, we optimized a new approach to produce primary cultures of the neuroblastoma xenografts for high-throughput drug screening which can be used to test new combinations of therapeutic agents for neuroblastoma.

Collaboration


Dive into the Sara M. Federico's collaboration.

Top Co-Authors

Avatar

Alberto S. Pappo

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

Michael A. Dyer

St. Jude Children's Research Hospital

View shared research outputs
Top Co-Authors

Avatar

Anang A. Shelat

St. Jude Children's Research Hospital

View shared research outputs
Top Co-Authors

Avatar

Elizabeth Stewart

St. Jude Children's Research Hospital

View shared research outputs
Top Co-Authors

Avatar

Rachel Brennan

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

Wayne L. Furman

St. Jude Children's Research Hospital

View shared research outputs
Top Co-Authors

Avatar

Armita Bahrami

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