Network


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

Hotspot


Dive into the research topics where Sarah Scollon is active.

Publication


Featured researches published by Sarah Scollon.


JAMA Oncology | 2016

Diagnostic Yield of Clinical Tumor and Germline Whole-Exome Sequencing for Children With Solid Tumors.

D. Williams Parsons; Angshumoy Roy; Yaping Yang; Tao Wang; Sarah Scollon; Katie Bergstrom; Robin A. Kerstein; Stephanie Gutierrez; Andrea K. Petersen; Abhishek Bavle; Frank Y. Lin; Dolores Lopez-Terrada; Federico A. Monzon; M. John Hicks; Karen W. Eldin; Norma M. Quintanilla; Adekunle M. Adesina; Carrie A. Mohila; William E. Whitehead; Andrew Jea; Sanjeev A. Vasudevan; Jed G. Nuchtern; Uma Ramamurthy; Amy L. McGuire; Susan G. Hilsenbeck; Jeffrey G. Reid; Donna M. Muzny; David A. Wheeler; Stacey L. Berg; Murali Chintagumpala

Importance Whole-exome sequencing (WES) has the potential to reveal tumor and germline mutations of clinical relevance, but the diagnostic yield for pediatric patients with solid tumors is unknown. Objective To characterize the diagnostic yield of combined tumor and germline WES for children with solid tumors. Design Unselected children with newly diagnosed and previously untreated central nervous system (CNS) and non-CNS solid tumors were prospectively enrolled in the BASIC3 study at a large academic childrens hospital during a 23-month period from August 2012 through June 2014. Blood and tumor samples underwent WES in a certified clinical laboratory with genetic results categorized on the basis of perceived clinical relevance and entered in the electronic health record. Main Outcomes and Measures Clinical categorization of somatic mutations; frequencies of deleterious germline mutations related to patient phenotype and incidental medically-actionable mutations. Results Of the first 150 participants (80 boys and 70 girls, mean age, 7.4 years), tumor samples adequate for WES were available from 121 patients (81%). Somatic mutations of established clinical utility (category I) were reported in 4 (3%) of 121 patients, with mutations of potential utility (category II) detected in an additional 29 (24%) of 121 patients. CTNNB1 was the gene most frequently mutated, with recurrent mutations in KIT, TSC2, and MAPK pathway genes (BRAF, KRAS, and NRAS) also identified. Mutations in consensus cancer genes (category III) were found in an additional 24 (20%) of 121 tumors. Fewer than half of somatic mutations identified were in genes known to be recurrently mutated in the tumor type tested. Diagnostic germline findings related to patient phenotype were discovered in 15 (10%) of 150 cases: 13 pathogenic or likely pathogenic dominant mutations in adult and pediatric cancer susceptibility genes (including 2 each in TP53, VHL, and BRCA1), 1 recessive liver disorder with hepatocellular carcinoma (TJP2), and 1 renal diagnosis (CLCN5). Incidental findings were reported in 8 (5%) of 150 patients. Most patients harbored germline uncertain variants in cancer genes (98%), pharmacogenetic variants (89%), and recessive carrier mutations (85%). Conclusions and Relevance Tumor and germline WES revealed mutations in a broad spectrum of genes previously implicated in both adult and pediatric cancers. Combined reporting of tumor and germline WES identified diagnostic and/or potentially actionable findings in nearly 40% of newly diagnosed pediatric patients with solid tumors.


Genome Medicine | 2014

Obtaining informed consent for clinical tumor and germline exome sequencing of newly diagnosed childhood cancer patients

Sarah Scollon; Katie Bergstrom; Robin A. Kerstein; Tao Wang; Susan G. Hilsenbeck; Uma Ramamurthy; Richard A. Gibbs; Christine M. Eng; Murali Chintagumpala; Stacey L. Berg; Laurence B. McCullough; Amy L. McGuire; Sharon E. Plon; D. Williams Parsons

BackgroundEffectively educating families about the risks and benefits of genomic tests such as whole exome sequencing (WES) offers numerous challenges, including the complexity of test results and potential loss of privacy. Research on best practices for obtaining informed consent (IC) in a variety of clinical settings is needed. The BASIC3 study of clinical tumor and germline WES in an ethnically diverse cohort of newly diagnosed pediatric cancer patients offers the opportunity to study the IC process in the setting of critical illness. We report on our experience for the first 100 families enrolled, including study participation rates, reasons for declining enrollment, assessment of clinical and demographic factors that might impact study enrollment, and preferences of parents for participation in optional genomics study procedures.MethodsA specifically trained IC team offered study enrollment to parents of eligible children for procedures including clinical tumor and germline WES with results deposited in the medical record and disclosure of both diagnostic and incidental results to the family. Optional study procedures were also offered, such as receiving recessive carrier status and deposition of data into research databases. Stated reasons for declining participation were recorded. Clinical and demographic data were collected and comparisons made between enrolled and non-enrolled patients.ResultsOver 15 months, 100 of 121 (83%) eligible families elected to enroll in the study. No significant differences in enrollment were detected based on factors such as race, ethnicity, use of Spanish interpreters and Spanish consent forms, and tumor features (central nervous system versus non-central nervous system, availability of tumor for WES). The most common reason provided for declining enrollment (10% of families) was being overwhelmed by the new cancer diagnosis. Risks specific to clinical genomics, such as privacy concerns, were less commonly reported (5.5%). More than 85% of parents consented to each of the optional study procedures.ConclusionsAn IC process was developed that utilizes a specialized IC team, active communication with the oncology team, and an emphasis on scheduling flexibility. Most parents were willing to participate in a clinical germline and tumor WES study as well as optional procedures such as genomic data sharing independent of race, ethnicity or language spoken.


Cancer | 2015

Parent decision-making around the genetic testing of children for germline TP53 mutations

Melissa A. Alderfer; Kristin Zelley; Robert B. Lindell; Ana Novokmet; Phuong L. Mai; Judy Garber; Deepika Nathan; Sarah Scollon; Nicolette M. Chun; Andrea Farkas Patenaude; James M. Ford; Sharon E. Plon; Joshua D. Schiffman; Lisa Diller; Sharon A. Savage; David Malkin; Carol A. Ford; Kim E. Nichols

Li‐Fraumeni syndrome is a rare genetic cancer predisposition syndrome caused by germline TP53 mutations. Up to 20% of mutation carriers develop cancer during childhood. The benefits of TP53 mutation testing of children are a matter of debate and knowledge of parent decision‐making around such testing is limited. The current study examined how parents make decisions regarding TP53 testing for their children.


Personalized Medicine | 2015

Illustrative case studies in the return of exome and genome sequencing results

Laura M. Amendola; Denise Lautenbach; Sarah Scollon; Barbara A. Bernhardt; Sawona Biswas; Kelly East; Jessica Everett; Marian J. Gilmore; Patricia Himes; Victoria M. Raymond; Julia Wynn; Ragan Hart; Gail P. Jarvik

Whole genome and exome sequencing tests are increasingly being ordered in clinical practice, creating a need for research exploring the return of results from these tests. A goal of the Clinical Sequencing and Exploratory Research (CSER) consortium is to gain experience with this process to develop best practice recommendations for offering exome and genome testing and returning results. Genetic counselors in the CSER consortium have an integral role in the return of results from these genomic sequencing tests and have gained valuable insight. We present seven emerging themes related to return of exome and genome sequencing results accompanied by case descriptions illustrating important lessons learned, counseling challenges specific to these tests and considerations for future research and practice.


Clinical Cancer Research | 2017

Retinoblastoma and Neuroblastoma Predisposition and Surveillance

Junne Kamihara; Franck Bourdeaut; William D. Foulkes; Jan J. Molenaar; Yael P. Mosse; Akira Nakagawara; Andreu Parareda; Sarah Scollon; Kami Wolfe Schneider; Alison H. Skalet; Lisa J. States; Michael F. Walsh; Lisa Diller; Garrett M. Brodeur

Retinoblastoma (RB) is the most common intraocular malignancy in childhood. Approximately 40% of retinoblastomas are hereditary and due to germline mutations in the RB1 gene. Children with hereditary RB are also at risk for developing a midline intracranial tumor, most commonly pineoblastoma. We recommend intensive ocular screening for patients with germline RB1 mutations for retinoblastoma as well as neuroimaging for pineoblastoma surveillance. There is an approximately 20% risk of developing second primary cancers among individuals with hereditary RB, higher among those who received radiotherapy for their primary RB tumors. However, there is not yet a clear consensus on what, if any, screening protocol would be most appropriate and effective. Neuroblastoma (NB), an embryonal tumor of the sympathetic nervous system, accounts for 15% of pediatric cancer deaths. Prior studies suggest that about 2% of patients with NB have an underlying genetic predisposition that may have contributed to the development of NB. Germline mutations in ALK and PHOX2B account for most familial NB cases. However, other cancer predisposition syndromes, such as Li–Fraumeni syndrome, RASopathies, and others, may be associated with an increased risk for NB. No established protocols for NB surveillance currently exist. Here, we describe consensus recommendations on hereditary RB and NB from the AACR Childhood Cancer Predisposition Workshop. Clin Cancer Res; 23(13); e98–e106. ©2017 AACR. See all articles in the online-only CCR Pediatric Oncology Series.


Journal of Genetic Counseling | 2016

“Not Tied Up Neatly with a Bow”: Professionals’ Challenging Cases in Informed Consent for Genomic Sequencing

Ashley N. Tomlinson; Debra Skinner; Denise L. Perry; Sarah Scollon; Myra I. Roche; Barbara A. Bernhardt

As the use of genomic technology has expanded in research and clinical settings, issues surrounding informed consent for genome and exome sequencing have surfaced. Despite the importance of informed consent, little is known about the specific challenges that professionals encounter when consenting patients or research participants for genomic sequencing. We interviewed 29 genetic counselors and research coordinators with considerable experience obtaining informed consent for genomic sequencing to understand their experiences and perspectives. As part of this interview, 24 interviewees discussed an informed consent case they found particularly memorable or challenging. We analyzed these case examples to determine the primary issue or challenge represented by each case. Challenges fell into two domains: participant understanding, and facilitating decisions about testing or research participation. Challenges related to participant understanding included varying levels of general and genomic literacy, difficulty managing participant expectations, and contextual factors that impeded participant understanding. Challenges related to facilitating decision-making included complicated family dynamics such as disagreement or coercion, situations in which it was unclear whether sequencing research would be a good use of participant time or resources, and situations in which the professional experienced disagreement or discomfort with participant decisions. The issues highlighted in these case examples are instructive in preparing genetics professionals to obtain informed consent for genomic sequencing.


Clinical Cancer Research | 2017

Recommendations for surveillance for children with leukemia-predisposing conditions

Christopher C. Porter; Todd E. Druley; Ayelet Erez; Roland P. Kuiper; Kenan Onel; Joshua D. Schiffman; Kami Wolfe Schneider; Sarah Scollon; Hamish S. Scott; Louise C. Strong; Michael F. Walsh; Kim E. Nichols

Leukemia, the most common childhood cancer, has long been recognized to occasionally run in families. The first clues about the genetic mechanisms underlying familial leukemia emerged in 1990 when Li-Fraumeni syndrome was linked to TP53 mutations. Since this discovery, many other genes associated with hereditary predisposition to leukemia have been identified. Although several of these disorders also predispose individuals to solid tumors, certain conditions exist in which individuals are specifically at increased risk to develop myelodysplastic syndrome (MDS) and/or acute leukemia. The increasing identification of affected individuals and families has raised questions around the efficacy, timing, and optimal methods of surveillance. As part of the AACR Childhood Cancer Predisposition Workshop, an expert panel met to review the spectrum of leukemia-predisposing conditions, with the aim to develop consensus recommendations for surveillance for pediatric patients. The panel recognized that for several conditions, routine monitoring with complete blood counts and bone marrow evaluations is essential to identify disease evolution and enable early intervention with allogeneic hematopoietic stem cell transplantation. However, for others, less intensive surveillance may be considered. Because few reports describing the efficacy of surveillance exist, the recommendations derived by this panel are based on opinion, and local experience and will need to be revised over time. The development of registries and clinical trials is urgently needed to enhance understanding of the natural history of the leukemia-predisposing conditions, such that these surveillance recommendations can be optimized to further enhance long-term outcomes. Clin Cancer Res; 23(11); e14–e22. ©2017 AACR. See all articles in the online-only CCR Pediatric Oncology Series.


Clinical Cancer Research | 2017

Genetic Counselor Recommendations for Cancer Predisposition Evaluation and Surveillance in the Pediatric Oncology Patient

Harriet Druker; Kristin Zelley; Rose B. McGee; Sarah Scollon; Wendy Kohlmann; Katherine A. Schneider; Kami Wolfe Schneider

As the understanding of the genetic etiology of childhood cancers increases, the need for the involvement of experts familiar with the provision of genetic counseling for this population is paramount. In October 2016, the American Association for Cancer Research organized the AACR Childhood Cancer Predisposition Workshop in which international experts in pediatric cancer predisposition met to establish surveillance guidelines for children with cancer predisposition. Identifying for whom, when, why, and how these cancer predisposition surveillance guidelines should be implemented is essential. Genetic counselors invited to this workshop provide a genetic counseling framework for oncology professionals in this article. Points of entry and recommendations regarding the provision and timing of the initial and subsequent genetic counseling sessions are addressed. The genetic counseling and testing processes are reviewed, and the psychologic impact related to surveillance is explored. Pediatric cancer genetics will continue to grow and evolve as a field, and genetic counseling services will be vital to ensure appropriate identification and management of at-risk children moving forward. Clin Cancer Res; 23(13); e91–e97. ©2017 AACR. See all articles in the online-only CCR Pediatric Oncology Series.


Clinical Cancer Research | 2017

Cancer screening recommendations and clinical management of inherited gastrointestinal cancer syndromes in childhood

Maria Isabel Achatz; Christopher C. Porter; Laurence Brugières; Harriet Druker; Thierry Frebourg; William D. Foulkes; Christian P. Kratz; Roland P. Kuiper; Jordan R. Hansford; Hector Salvador Hernandez; Katherine L. Nathanson; Wendy Kohlmann; Leslie Doros; Kenan Onel; Kami Wolfe Schneider; Sarah Scollon; Uri Tabori; Gail E. Tomlinson; D. Gareth Evans; Sharon E. Plon

Hereditary gastrointestinal cancer predisposition syndromes have been well characterized, but management strategies and surveillance remain a major challenge, especially in childhood. In October 2016, the American Association for Cancer Research organized the AACR Childhood Cancer Predisposition Workshop in which international experts in care of children with a hereditary risk of cancer met to define surveillance strategies and management of children with cancer predisposition syndromes. In this article, we review the current literature in polyposis syndromes that can be diagnosed in childhood and may be associated with an increased incidence of gastrointestinal neoplasms and other cancer types. These disorders include adenomatous polyposis syndromes (APC and MUTYH), juvenile polyposis coli (BMPR1A and SMAD4), Peutz–Jeghers Syndrome (STK11/LKB1), and PTEN hamartoma tumor syndrome (PHTS; PTEN), which can present with a more limited juvenile polyposis phenotype. Herein, the panel of experts provides recommendations for clinical diagnosis, approach to genetic testing, and focus on cancer surveillance recommendations when appropriate during the pediatric period. We also review current controversies on genetic evaluation of patients with hepatoblastoma and indications for surveillance for this tumor. Childhood cancer risks and surveillance associated with disorders involving the mismatch repair genes, including Lynch syndrome and constitutional mismatch repair deficiency (CMMRD), are discussed elsewhere in this series. Clin Cancer Res; 23(13); e107–e14. ©2017 AACR. See all articles in the online-only CCR Pediatric Oncology Series.


Journal of Genetic Counseling | 2017

A Comprehensive Review of Pediatric Tumors and Associated Cancer Predisposition Syndromes

Sarah Scollon; Amanda Knoth Anglin; Martha Thomas; Joyce Turner; Kami Wolfe Schneider

An understanding of the role of inherited cancer predisposition syndromes in pediatric tumor diagnoses continues to develop as more information is learned through the application of genomic technology. Identifying patients and their relatives at an increased risk for developing cancer is an important step in the care of this patient population. The purpose of this review is to highlight various tumor types that arise in the pediatric population and the cancer predisposition syndromes associated with those tumors. The review serves as a guide for recognizing genes and conditions to consider when a pediatric cancer referral presents to the genetics clinic.

Collaboration


Dive into the Sarah Scollon's collaboration.

Top Co-Authors

Avatar

Sharon E. Plon

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Katie Bergstrom

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Stacey L. Berg

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Amy L. McGuire

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Christine M. Eng

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Robin A. Kerstein

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Tao Wang

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Angshumoy Roy

Baylor College of Medicine

View shared research outputs
Researchain Logo
Decentralizing Knowledge