Network


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

Hotspot


Dive into the research topics where Faye R. Harris is active.

Publication


Featured researches published by Faye R. Harris.


Journal of Clinical Oncology | 2014

Identification of Independent Primary Tumors and Intrapulmonary Metastases Using DNA Rearrangements in Non–Small-Cell Lung Cancer

Stephen J. Murphy; Marie Christine Aubry; Faye R. Harris; Geoffrey C. Halling; Sarah H. Johnson; Simone Terra; Travis M. Drucker; Michael K. Asiedu; Benjamin R. Kipp; Eunhee S. Yi; Tobias Peikert; Ping Yang; George Vasmatzis; Dennis A. Wigle

PURPOSE Distinguishing independent primary tumors from intrapulmonary metastases in non-small-cell carcinoma remains a clinical dilemma with significant clinical implications. Using next-generation DNA sequencing, we developed a chromosomal rearrangement-based approach to differentiate multiple primary tumors from metastasis. METHODS Tumor specimens from patients with known independent primary tumors and metastatic lesions were used for lineage test development, which was then applied to multifocal tumors. Laser capture microdissection was performed separately for each tumor. Genomic DNA was isolated using direct in situ whole-genome amplification methodology, and next-generation sequencing was performed using an Illumina mate-pair library protocol. Sequence reads were mapped to the human genome, and primers spanning the fusion junctions were used for validation polymerase chain reaction. RESULTS A total of 41 tumor samples were sequenced (33 adenocarcinomas [ADs] and eight squamous cell carcinomas [SQCCs]), with a range of three to 276 breakpoints per tumor identified. Lung tumors predicted to be independent primary tumors based on different histologic subtype did not share any genomic rearrangements. In patients with lung primary tumors and paired distant metastases, shared rearrangements were identified in all tumor pairs, emphasizing the patient specificity of identified breakpoints. Multifocal AD and SQCC samples were reviewed independently by two pulmonary pathologists. Concordance between histology and genomic data occurred in the majority of samples. Discrepant tumor samples were resolved by genome sequencing. CONCLUSION A diagnostic lineage test based on genomic rearrangements from mate-pair sequencing demonstrates promise for distinguishing independent primary from metastatic disease in lung cancer.


PLOS Genetics | 2015

Suppression of Somatic Expansion Delays the Onset of Pathophysiology in a Mouse Model of Huntington's Disease.

Helen Budworth; Faye R. Harris; Paul T. Williams; Do Yup Lee; Amy Holt; Jens Pahnke; Bartosz Szczesny; Karina Acevedo-Torres; Sylvette Ayala-Peña; Cynthia T. McMurray

Huntington’s Disease (HD) is caused by inheritance of a single disease-length allele harboring an expanded CAG repeat, which continues to expand in somatic tissues with age. The inherited disease allele expresses a toxic protein, and whether further somatic expansion adds to toxicity is unknown. We have created an HD mouse model that resolves the effects of the inherited and somatic expansions. We show here that suppressing somatic expansion substantially delays the onset of disease in littermates that inherit the same disease-length allele. Furthermore, a pharmacological inhibitor, XJB-5-131, inhibits the lengthening of the repeat tracks, and correlates with rescue of motor decline in these animals. The results provide evidence that pharmacological approaches to offset disease progression are possible.


Modern Pathology | 2016

Integrated analysis of the genomic instability of PTEN in clinically insignificant and significant prostate cancer.

Stephen J. Murphy; R.J. Karnes; Farhad Kosari; B Edgardo R Parilla Castellar; Benjamin R. Kipp; Sarah H. Johnson; Simone Terra; Faye R. Harris; Geoffrey C. Halling; Janet L. Schaefer Klein; Aqsa Nasir; Eric J. Bergstrahl; Laureano J. Rangel; William R. Sukov; George Vasmatzis; John C. Cheville

Patients with clinically insignificant prostate cancer remain a major over-treated population. PTEN loss is one of the most recurrent alterations in prostate cancer associated with an aggressive phenotype, however, the occurrence of PTEN loss in insignificant prostate cancer has not been reported and its role in the separation of insignificant from significant prostate cancer is unclear. An integrated analysis of PTEN loss was, therefore, performed for structural variations, point mutations and protein expression in clinically insignificant (48 cases) and significant (76 cases) prostate cancers treated by radical prostatectomy. Whole-genome mate pair sequencing was performed on tumor cells isolated by laser capture microdissection to characterize PTEN structural alterations. Fluorescence in situ hybridization probes were constructed from the sequencing data to detect the spectrum of these PTEN alterations. PTEN loss by mate pair sequencing and fluorescence in situ hybridization occurred in 2% of insignificant, 13% of large volume Gleason score 6, and 46% of Gleason score 7 and higher cancers. In Gleason score 7 cancers with PTEN loss, PTEN alterations were detected in both Gleason pattern 3 and 4 in 57% of cases by mate pair sequencing, 75% by in situ hybridization and 86% by immunohistochemistry. PTEN loss by sequencing was strongly associated with TMPRSS2-ERG fusion, biochemical recurrence, PTEN loss by in situ hybridization and protein loss by immunohistochemistry. The complex nature of PTEN rearrangements was unveiled by sequencing, detailing the heterogeneous events leading to homozygous loss of PTEN. PTEN point mutation was present in 5% of clinically significant tumors and not in insignificant cancer or high-grade prostatic intraepithelial neoplasia. PTEN loss is infrequent in clinically insignificant prostate cancer, and is associated with higher grade tumors. Detection of PTEN loss in Gleason score 6 cancer in a needle biopsy specimen indicates a higher likelihood of clinically significant prostate cancer.


Scientific Reports | 2016

Quantification of Somatic Chromosomal Rearrangements in Circulating Cell-Free DNA from Ovarian Cancers.

Faye R. Harris; Irina V. Kovtun; James B. Smadbeck; Francesco Multinu; Aminah Jatoi; Farhad Kosari; Kimberly R. Kalli; Stephen J. Murphy; Geoffrey C. Halling; Sarah H. Johnson; Minetta C. Liu; Andrea Mariani; George Vasmatzis

Recently, the use of a liquid biopsy has shown promise in monitoring tumor burden. While point mutations have been extensively studied, chromosomal rearrangements have demonstrated greater tumor specificity. Such rearrangements can be identified in the tumor and subsequently detected in the plasma of patients using quantitative PCR (qPCR). In this study we used a whole-genome mate-pair protocol to characterize a landscape of genomic rearrangements in the primary tumors of ten ovarian cancer patients. Individualized tumor-specific primer panels of aberrant chromosomal junctions were identified for each case and detected by qPCR within the cell-free DNA. Selected chromosomal junctions were detected in pre-surgically drawn blood in eight of the ten patients. Of these eight, three demonstrated the continued presence of circulating tumor DNA (ctDNA) post-surgery, consistent with their documented presence of disease, and in five ctDNA was undetectable in the post-surgical blood collection, consistent with their lack of detectable disease. The ctDNA fraction was calculated using a novel algorithm designed for the unique challenges of quantifying ctDNA using qPCR to allow observations of real-time tumor dynamics. In summary, a panel of individualized junctions derived from tumor DNA could be an effective way to monitor cancer patients for relapse and therapeutic efficacy using cfDNA.


Cancer Research | 2016

Integrated Genomic Analysis of Pancreatic Ductal Adenocarcinomas Reveals Genomic Rearrangement Events as Significant Drivers of Disease

Stephen J. Murphy; Steven N. Hart; Geoffrey C. Halling; Sarah H. Johnson; James B. Smadbeck; Travis Drucker; Joema Felipe Lima; Fariborz Rakhshan Rohakhtar; Faye R. Harris; Farhad Kosari; Subbaya Subramanian; Gloria M. Petersen; Timothy D. Wiltshire; Benjamin R. Kipp; Mark J. Truty; Robert R. McWilliams; Fergus J. Couch; George Vasmatzis

Many somatic mutations have been detected in pancreatic ductal adenocarcinoma (PDAC), leading to the identification of some key drivers of disease progression, but the involvement of large genomic rearrangements has often been overlooked. In this study, we performed mate pair sequencing (MPseq) on genomic DNA from 24 PDAC tumors, including 15 laser-captured microdissected PDAC and 9 patient-derived xenografts, to identify genome-wide rearrangements. Large genomic rearrangements with intragenic breakpoints altering key regulatory genes involved in PDAC progression were detected in all tumors. SMAD4, ZNF521, and FHIT were among the most frequently hit genes. Conversely, commonly reported genes with copy number gains, including MYC and GATA6, were frequently observed in the absence of direct intragenic breakpoints, suggesting a requirement for sustaining oncogenic function during PDAC progression. Integration of data from MPseq, exome sequencing, and transcriptome analysis of primary PDAC cases identified limited overlap in genes affected by both rearrangements and point mutations. However, significant overlap was observed in major PDAC-associated signaling pathways, with all PDAC exhibiting reduced SMAD4 expression, reduced SMAD-dependent TGFβ signaling, and increased WNT and Hedgehog signaling. The frequent loss of SMAD4 and FHIT due to genomic rearrangements strongly implicates these genes as key drivers of PDAC, thus highlighting the strengths of an integrated genomic and transcriptomic approach for identifying mechanisms underlying disease initiation and progression.


Cancer Research | 2014

Genomic Rearrangements Define Lineage Relationships between Adjacent Lepidic and Invasive Components in Lung Adenocarcinoma

Stephen J. Murphy; Dennis A. Wigle; Joema Felipe Lima; Faye R. Harris; Sarah H. Johnson; Geoffrey C. Halling; Michael K. Asiedu; Charlie T. Seto; Simone Terra; Farhad Kosari; Tobias Peikert; Ping Yang; Marie Christine Aubry; George Vasmatzis

The development of adenocarcinoma of the lung is believed to proceed from in situ disease (adenocarcinoma in situ, AIS) to minimally invasive disease with prominent lepidic growth (minimally invasive adenocarcinoma, MIA), then to fully invasive adenocarcinoma (AD), but direct evidence for this model has been lacking. Because some lung adenocarcinomas show prominent lepidic growth (AD-L), we designed a study to address the lineage relationship between the lepidic (noninvasive) component (L) and the adjacent nonlepidic growth component representing invasive disease within individual tumors. Lineage relationships were evaluated by next-generation DNA sequencing to define large genomic rearrangements in microdissected tissue specimens collected by laser capture. We found a strong lineage relationship between the majority of adjacent lepidic and invasive components, supporting a putative AIS-AD transition. Notably, many rearrangements were detected in the less aggressive lepidic component, although the invasive component exhibited an overall higher rate of genomic rearrangement. Furthermore, a significant number of genomic rearrangements were present in histologically normal lung adjacent to tumor, but not in host germline DNA, suggesting field defects restricted to zonal regions near a tumor. Our results offer a perspective on the genetic pathogenesis underlying adenocarcinoma development and its clinical management.


Modern Pathology | 2015

Chromosomal rearrangements and copy number abnormalities of TP63 correlate with p63 protein expression in lung adenocarcinoma.

Marie Christine Aubry; Anja C. Roden; Stephen J. Murphy; George Vasmatzis; Sarah H. Johnson; Faye R. Harris; Geoffrey C. Halling; Ryan A. Knudson; Rhett P. Ketterling; Andrew L. Feldman

The TP63 gene encodes a member of the p53 family of transcription factors. Although TP53 is a well-known tumor suppressor gene, the role of p63 in tumorigenesis is controversial. Our group recently identified novel chromosomal rearrangements involving TP63 in approximately 6% of peripheral T-cell lymphomas, which correlated with a p63+/p40− immunohistochemical profile. As a subset of lung adenocarcinomas are p63+/p40−, we undertook the current study to examine the presence of TP63 rearrangements and correlate with p63/p40 expression. Next-generation sequencing was used to identify genomic rearrangements of TP63 in 37 adenocarcinomas. Confirmatory fluorescence in-situ hybridization (FISH) using a break-apart probe to the TP63 gene region and immunohistochemistry for p63 and p40 were performed on adenocarcinomas with TP63 rearrangements identified by mate-pair sequencing. Immunohistochemistry for p63 and p40 was performed on 45 additional adenocarcinomas, and FISH was performed on all adenocarcinomas with p63 positivity. TP63 rearrangement was identified in two adenocarcinoma specimens from a single patient. The rearrangement resulted in a complex rearrangement of 3q that fused B3GALNT1 at the 3′ intron to TP63. FISH confirmed the rearrangement in both tumors. Immunohistochemistry staining for p63 was diffuse (>80% cells+) and p40 was negative. Of the 44 additional adenocarcinomas, 13 (30%) showed p63 expression; p40 was negative in all cases. No case showed rearrangement of TP63 by a break-apart FISH. However, extra copies of the intact TP63 locus were seen in the p63-positive areas of all 12 cases, with copy numbers ranging from three to seven. We have identified a novel chromosomal rearrangement involving TP63 in a p63+/p40− lung adenocarcinoma. Break-apart FISH testing can be used to diagnose this finding. Immunohistochemistry for p63 was not specific for this rearrangement, as nearly 33% of adenocarcinomas expressed p63. Additional copies of the intact TP63 locus were also a common finding and correlated with immunohistochemistry positivity for p63.


Clinical Cancer Research | 2016

Abstract 13: Individualized approach for ovarian cancer: Identification of potential therapeutic targets based on genomic analyses, testing efficiency of treatments, and monitoring

Faye R. Harris; Geoffrey C. Halling; Marc A. Becker; Paul Haluska; George Vasmatzis; Irina V. Kovtun

Ovarian cancer (OCa) is fifth leading cause of death among women. Most of OCa patients diagnosed with high stage invasive disease. Although, majority experience remission after surgical debulking and adjuvant chemotherapy, about 80% of patients relapse and/or develop chemoresistant disease. More efficient diagnostics and therapies are needed to improve outcome in ovarian cancer. We used an integrated approach to characterize genomic rearrangements in serous OCa inordet to identify potentially targetable alterations and test treatments with corresponding drugs using matching mouse avatar models. Mate-pair next generation sequencing protocol and BIMA2 algorithm were used to identify chromosomal rearrangements, including gene fusions and copy number changes, in primary tumors. Further analysis was then performed to determine alterations that may also be therapeutically targeted. Total of 25 primary tumors were analyzed, out of which four were selected for genomically guided treatments in corresponding mouse avatars. The tumors propagated in the selected mouse lines were also sequenced and showed identical to original patient tumor landscape of rearrangements. In addition, the selected targetable alterations were validated in both patient and avatar tumor DNA. For several of the analyzed cases pre-surgical and postsurgical blood was available for detection of selected alterations in cell free circulating tumor DNA. The analysis revealed presence of selected rearrangements in pre-surgical blood of all tested cases and in a number of those that showed presence of the disease after surgery. In summary, we showed that the whole genome sequencing of DNA rearrangements in conjunction with analyses of therapeutic targets and treatment testing in mouse avatars, developed using primary tumors, is a promising approach to aid treatment decisions for individual ovarian cancer patients. Note: This abstract was not presented at the conference. Citation Format: Faye R. Harris, Geoffrey C. Halling, Marc A. Becker, Paul Haluska, Jr., George Vasmatzis, Irina V. Kovtun. Individualized approach for ovarian cancer: Identification of potential therapeutic targets based on genomic analyses, testing efficiency of treatments, and monitoring. [abstract]. In: Proceedings of the AACR Precision Medicine Series: Integrating Clinical Genomics and Cancer Therapy; Jun 13-16, 2015; Salt Lake City, UT. Philadelphia (PA): AACR; Clin Cancer Res 2016;22(1_Suppl):Abstract nr 13.


Journal of Thoracic Oncology | 2018

Neoantigenic potential of complex chromosomal rearrangements in mesothelioma

Aaron S. Mansfield; Tobias Peikert; James B. Smadbeck; Julia B. Udell; Enrique Garcia-Rivera; Laura R. Elsbernd; Courtney L. Erskine; Virginia Van Keulen; Farhad Kosari; Stephen J. Murphy; Hongzheng Ren; Vishnu V. Serla; Janet L. Schaefer Klein; Giannoula Karagouga; Faye R. Harris; Carlos Sosa; Sarah H. Johnson; Wendy K. Nevala; Svetomir N. Markovic; Aaron O. Bungum; Eric S. Edell; Haidong Dong; John C. Cheville; Marie Christine Aubry; Jin Jen; George Vasmatzis

Introduction: Malignant pleural mesothelioma is a disease primarily associated with exposure to the carcinogen asbestos. Whereas other carcinogen‐related tumors are associated with a high tumor mutation burden, mesothelioma is not. We sought to resolve this discrepancy. Methods: We used mate‐pair (n = 22), RNA (n = 28), and T cell receptor sequencing along with in silico predictions and immunologic assays to understand how structural variants of chromosomes affect the transcriptome. Results: We observed that inter‐ or intrachromosomal rearrangements were present in every specimen and were frequently in a pattern of chromoanagenesis such as chromoplexy or chromothripsis. Transcription of rearrangement‐related junctions was predicted to result in many potential neoantigens, some of which were proven to bind patient‐specific major histocompatibility complex molecules and to expand intratumoral T cell clones. T cells responsive to these predicted neoantigens were also present in a patients circulating T cell repertoire. Analysis of genomic array data from the mesothelioma cohort in The Cancer Genome Atlas suggested that multiple chromothriptic‐like events negatively impact survival. Conclusions: Our findings represent the discovery of potential neoantigen expression driven by structural chromosomal rearrangements. These results may have implications for the development of novel immunotherapeutic strategies and the selection of patients to receive immunotherapies.


Cancer Research | 2018

Abstract 5726: Rearrangement-related peptides with neoantigenic potential in malignant pleural mesothelioma

Aaron S. Mansfield; Tobias Peikert; James B. Smadbeck; Julia B. Udell; Farhad Kosari; Stephen J. Murphy; Hongzheng Ren; Vishnu V. Serla; Janet L. Schaefer Klein; Giannoula Karagouga; Faye R. Harris; Carlos Sosa; Sarah H. Johnson; Wendy K. Nevala; Svetomir N. Markovic; Aaron O. Bungum; Eric S. Edell; Haidong Dong; John C. Cheville; Marie Christine Aubry; Jin Jen; George Vasmatzis

Malignant pleural mesothelioma is a disease primarily associated with exposure to the carcinogen asbestos. Consistent with this carcinogenic exposure, cytogenetic analyses have identified multiple recurrent structural chromosomal abnormalities in this malignancy, but more recent high-throughput sequencing evaluations of point mutations suggest that there is a low mutational burden in mesothelioma. Since tumor mutational burden has been correlated with responses to treatment with immune checkpoint inhibitors such as nivolumab, it was not consistent that patients with mesothelioma and low mutation burdens would have similar response rates in clinical trials with immune checkpoint inhibitors as patients with non-small cell lung cancer which is associated with a high mutation burden. In order to reconcile these differences, and given the potential for an improved understanding of the molecular pathogenesis of mesothelioma to improve therapeutic options, we used mate-pair sequencing (MPseq) and RNA sequencing (RNAseq) to understand how structural variants affect the transcriptome. MPseq differs from standard next generation sequencing approaches by tiling the whole genome with larger fragments (2-5kb) to reliably detect structural variants such as insertions, deletions and rearrangements. Amongst 22 mesothelioma specimens there were 1535 chromosomal rearrangements (median 41, range 3-298 per specimen), that resulted in junctions or novel fusions of non-coding DNA or genes. Six-hundred thirty-seven of these rearrangements (median 22, range 5-103 range per specimen) resulted in novel fusions of genes. Many of these inter- or intra-chromosomal rearrangements were consistent with a pattern of chromoanagesis such as chromoplexy or chromothripsis. Chromosomal rearrangements detected by MPseq were used to guide analysis of RNAseq data and revealed that these chromosomal junctions resulted in the expression of 179 novel amino acid sequences (median 5, 0-51 range per specimen). To determine whether transcription of chromosomal rearrangement-related junctions have neoantigenic potential, we used in silico tools to determine whether any of the expressed junctions contained peptides that could be presented by patient-specific HLA molecules. The top candidate rearrangement-related peptides with neoantigenic potential bound patient-specific HLA molecules nearly as well or as well as a positive control in competitive binding assays. Our findings represent the discovery of potential neoantigen expression driven by structural chromosomal rearrangements. These results may have implications for the development of novel therapeutic strategies, the selection of patients to receive immunotherapy, and blood-based treatment monitoring strategies. Citation Format: Aaron S. Mansfield, Tobias Peikert, James B. Smadbeck, Julia B. Udell, Farhad Kosari, Stephen J. Murphy, Hongzheng Ren, Vishnu V. Serla, Janet L. Schaefer Klein, Giannoula Karagouga, Faye R. Harris, Carlos Sosa, Sarah H. Johnson, Wendy Nevala, Svetomir N. Markovic, Aaron O. Bungum, Eric S. Edell, Haidong Dong, John C. Cheville, Marie Christine Aubry, Jin Jen, George Vasmatzis. Rearrangement-related peptides with neoantigenic potential in malignant pleural mesothelioma [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2018; 2018 Apr 14-18; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2018;78(13 Suppl):Abstract nr 5726.

Collaboration


Dive into the Faye R. Harris's collaboration.

Researchain Logo
Decentralizing Knowledge