Network


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

Hotspot


Dive into the research topics where Tanaya Neff is active.

Publication


Featured researches published by Tanaya Neff.


The Journal of Molecular Diagnostics | 2013

Combining Highly Multiplexed PCR with Semiconductor-Based Sequencing for Rapid Cancer Genotyping

Carol Beadling; Tanaya Neff; Michael C. Heinrich; Katherine Rhodes; Michael Thornton; John H. Leamon; Mark Andersen; Christopher L. Corless

There is growing demand for routine identification of actionable mutations in clinical cancer specimens. Genotyping platforms must provide rapid turnaround times and work effectively with limited amounts of formalin-fixed, paraffin-embedded (FFPE) tissue specimens that often yield poor quality DNA. We describe semiconductor-based sequencing of DNA from FFPE specimens using a single-tube, multiplexed panel of 190 amplicons targeting 46 cancer genes. With just 10 ng of input DNA, average read depths of 2000× can be obtained in 48 hours, with >95% of the reads on target. A validation set of 45 FFPE tumor specimens containing 53 point mutations previously identified with a mass spectrometry-based genotyping platform, along with 19 indels ranging from 4 to 63 bp, was used to evaluate assay performance. With a mutant allele ratio cutoff of 8%, we were able to achieve 100% sensitivity (95% CI = 97.3% to 100.0%) and 95.1% specificity (95% CI = 91.8% to 98.0%) of point mutation detection. All indels were visible by manual inspection of aligned reads; 6/9 indels ≤12 bp long were detected by the variant caller software either exactly or as mismatched nucleotides within the indel region. The rapid turnaround time and low input DNA requirements make the multiplex PCR and semiconductor-based sequencing approach a viable option for mutation detection in a clinical laboratory.


The Journal of Molecular Diagnostics | 2011

Multiplex Mutation Screening by Mass Spectrometry: Evaluation of 820 Cases from a Personalized Cancer Medicine Registry

Carol Beadling; Michael C. Heinrich; Andrea Warrick; Erin M. Forbes; Dylan Nelson; Emily Justusson; Judith Levine; Tanaya Neff; Janice Patterson; Ajia Presnell; Arin McKinley; Laura J. Winter; Christie Dewey; Amy Harlow; Oscar Barney; Brian J. Druker; Kathryn G. Schuff; Christopher L. Corless

There is an immediate and critical need for a rapid, broad-based genotyping method that can evaluate multiple mutations simultaneously in clinical cancer specimens and identify patients most likely to benefit from targeted agents now in use or in late-stage clinical development. We have implemented a prospective genotyping approach to characterize the frequency and spectrum of mutations amenable to drug targeting present in urothelial, colorectal, endometrioid, and thyroid carcinomas and in melanoma. Cancer patients were enrolled in a Personalized Cancer Medicine Registry that houses both clinical information and genotyping data, and mutation screening was performed using a multiplexed assay panel with mass spectrometry-based analysis to detect 390 mutations across 30 cancer genes. Formalin fixed, paraffin-embedded specimens were evaluated from 820 Registry patients. The genes most frequently mutated across multiple cancer types were BRAF, PIK3CA, KRAS, and NRAS. Less common mutations were also observed in AKT1, CTNNB1, FGFR2, FGFR3, GNAQ, HRAS, and MAP2K1. Notably, 48 of 77 PIK3CA-mutant cases (62%) harbored at least one additional mutation in another gene, most often KRAS. Among melanomas, only 54 of 73 BRAF mutations (74%) were the V600E substitution. These findings demonstrate the diversity and complexity of mutations in druggable targets among the different cancer types and underscore the need for a broad-spectrum, prospective genotyping approach to personalized cancer medicine.


The Journal of Molecular Diagnostics | 2015

Assessing copy number alterations in targeted, amplicon-based next-generation sequencing data.

Catherine S. Grasso; Timothy M Butler; Katherine Rhodes; Michael J. Quist; Tanaya Neff; Stephen R. Moore; Scott A. Tomlins; Erica Reinig; Carol Beadling; Mark Andersen; Christopher L. Corless

Changes in gene copy number are important in the setting of precision medicine. Recent studies have established that copy number alterations (CNAs) can be detected in sequencing libraries prepared by hybridization-capture, but there has been comparatively little attention given to CNA assessment in amplicon-based libraries prepared by PCR. In this study, we developed an algorithm for detecting CNAs in amplicon-based sequencing data. CNAs determined from the algorithm mirrored those from a hybridization-capture library. In addition, analysis of 14 pairs of matched normal and breast carcinoma tissues revealed that sequence data pooled from normal samples could be substituted for a matched normal tissue without affecting the detection of clinically relevant CNAs (>|2| copies). Comparison of CNAs identified by array comparative genomic hybridization and amplicon-based libraries across 10 breast carcinoma samples showed an excellent correlation. The CNA algorithm also compared favorably with fluorescence in situ hybridization, with agreement in 33 of 38 assessments across four different genes. Factors that influenced the detection of CNAs included the number of amplicons per gene, the average read depth, and, most important, the proportion of tumor within the sample. Our results show that CNAs can be identified in amplicon-based targeted sequencing data, and that their detection can be optimized by ensuring adequate tumor content and read coverage.


Leukemia | 2012

Newly described activating JAK3 mutations in T-cell acute lymphoblastic leukemia

T Bains; Michael C. Heinrich; Marc Loriaux; Carol Beadling; Dylan Nelson; Andrea Warrick; Tanaya Neff; Jeffrey W. Tyner; Jennifer Dunlap; Christopher L. Corless; Guang Fan

T-cell acute lymphoblastic leukemia (T-ALL) is a genetically heterogeneous disease that accounts for 10–15% of adult and 25% of childhood ALL cases. Despite advances in therapy of T-ALL, relapsed disease remains a leading cause of death. The genetic and biological determinants of treatment failure and clinical outcomes remain incompletely understood.


Modern Pathology | 2012

Phosphatidylinositol-3-kinase pathway mutations are common in breast columnar cell lesions.

Megan L. Troxell; Alayne L Brunner; Tanaya Neff; Andrea Warrick; Carol Beadling; Kelli Montgomery; Shirley Zhu; Christopher L. Corless; Robert B. West

The phosphatidylinositol-3-kinase pathway is one of the most commonly mutated pathways in invasive breast carcinoma, with PIK3CA mutations in ∼25% of invasive carcinomas, and AKT1 mutations in up to 5%. Ductal carcinoma in situ, and benign papillomas harbor similar mutations. However, activating point mutations in breast columnar cell lesions have been infrequently studied. Twenty-three breast resection specimens containing columnar cell lesions were identified; 14 with associated invasive carcinoma or carcinoma in situ. DNA extracts were prepared from formalin-fixed paraffin-embedded tissue and screened for a panel of point mutations (321 mutations in 30 genes) using a multiplex PCR panel with mass-spectroscopy readout. PIK3CA mutations were identified in 13/24 columnar cell lesions (54%) and 3/8 invasive carcinomas (37%). The mutation status of columnar cell lesions and associated carcinoma was frequently discordant. Of the 14 cases, only 5 demonstrated the same genotype in matched samples of columnar cell lesions and carcinoma (4 wild type, 1 PIK3CA H1047R). Interestingly, five patients had mutations in columnar cell lesions with wild-type carcinoma; two patients had different point mutations in columnar cell lesions and carcinoma. Only three cases had wild-type columnar cell lesion and mutated carcinoma. The 50% PIK3CA mutation prevalence in columnar cell lesions is greater than reported in most studies of invasive breast cancer. Further, columnar cell lesions and carcinoma were frequently discordant for PIK3CA/AKT1 mutation status. These findings raise interesting questions about the role of PIK3CA/AKT pathway in breast carcinogenesis, and the biologic/precursor potential of columnar cell lesions.


Human Pathology | 2012

Mucinous breast carcinomas lack PIK3CA and AKT1 mutations

Elizabeth Kehr; Julie M. Jorns; Daphne Ang; Andrea Warrick; Tanaya Neff; Michelle Degnin; Rebecca Lewis; Carol Beadling; Christopher L. Corless; Megan L. Troxell

Activating point mutations in the phosphatidylinositol-3-kinase catalytic subunit (PIK3CA) are among the most common molecular defects in invasive breast cancer. Point mutations in the downstream kinase AKT1 are seen in a minority of carcinomas. These mutations are found preferentially in estrogen receptor-positive and Her2-positive breast carcinomas; however, special morphologic types of breast cancer have not been well studied. Twenty-nine cases of pure invasive mucinous carcinoma and 9 cases of ductal carcinoma with mucinous differentiation were screened for a panel of point mutations (>321 mutations in 30 genes) using a multiplex polymerase chain reaction panel with mass spectroscopy readout. In addition, associated ductal carcinoma in situ, hyperplasia, or columnar cell lesions were separately tested where available (25 lesions). In 3 invasive cases and 15 ductal carcinoma in situ/proliferative lesions, PIK3CA hotspot mutations were, instead, tested by direct sequencing. No point mutations were identified in invasive mucinous breast carcinoma. This contrasts with the 35% frequency of PIK3CA mutations in a comparative group of invasive ductal carcinomas of no special type. Interestingly, PIK3CA hotspot point mutations were identified in associated ductal carcinoma in situ (3/14) and hyperplasia (atypical ductal hyperplasia [2/3], usual ductal hyperplasia [2/3], columnar cell change [1/5]), suggesting that PIK3CA mutations may play a role in breast epithelial proliferation. This series represents the largest study, to date, of PIK3CA genotyping in mucinous carcinoma and supports the unique pathogenetics of invasive mucinous breast carcinoma.


Applied Immunohistochemistry & Molecular Morphology | 2015

Novel Mutations in Neuroendocrine Carcinoma of the Breast: Possible Therapeutic Targets

Daphne Ang; Morgan Ballard; Carol Beadling; Andrea Warrick; Amy Schilling; Rebecca O'Gara; Marina Pukay; Tanaya Neff; Robert B. West; Christopher L. Corless; Megan L. Troxell

Primary neuroendocrine carcinoma of the breast is a rare variant, accounting for only 2% to 5% of diagnosed breast cancers, and may have relatively aggressive behavior. Mutational profiling of invasive ductal breast cancers has yielded potential targets for directed cancer therapy, yet most studies have not included neuroendocrine carcinomas. In a tissue microarray screen, we found a 2.4% prevalence (9/372) of neuroendocrine breast carcinoma, including several with lobular morphology. We then screened primary or metastatic neuroendocrine breast carcinomas (excluding papillary and mucinous) for mutations in common cancer genes using polymerase chain reaction-mass spectroscopy (643 hotspot mutations across 53 genes), or semiconductor-based next-generation sequencing analysis (37 genes). Mutations were identified in 5 of 15 tumors, including 3 with PIK3CA exon 9 E542K mutations, 2 of which also harbored point mutations in FGFR family members (FGFR1 P126S, FGFR4 V550M). Single mutations were found in each of KDR (A1065T) and HRAS (G12A). PIK3CA mutations are common in other types of breast carcinoma. However, FGFR and RAS family mutations are exceedingly rare in the breast cancer literature. Likewise, activating mutations in the receptor tyrosine kinase KDR (VEGFR2) have been reported in angiosarcomas and non–small cell lung cancers; the KDR A1065T mutation is reported to be sensitive to VEGFR kinase inhibitors, and fibroblast growth factor receptor inhibitors are in trials. Our findings demonstrate the utility of broad-based genotyping in the study of rare tumors such as neuroendocrine breast cancer.


Human Pathology | 2012

Multiplex high-throughput gene mutation analysis in acute myeloid leukemia

Jennifer Dunlap; Carol Beadling; Andrea Warrick; Tanaya Neff; William H. Fleming; Marc Loriaux; Michael C. Heinrich; Tibor Kovacsovics; Katalin Kelemen; Nicky Leeborg; Ken Gatter; Rita M. Braziel; Richard D. Press; Christopher L. Corless; Guang Fan

Classification of acute myeloid leukemia increasingly depends on genetic analysis. However, the number of known mutations in acute myeloid leukemia is expanding rapidly. Therefore, we tested a high-throughput screening method for acute myeloid leukemia mutation analysis using a multiplex mass spectrometry-based approach. To our knowledge, this is the first reported application of this approach to genotype leukemias in a clinical setting. One hundred seven acute myeloid leukemia cases were screened for mutations using a panel that covers 344 point mutations across 31 genes known to be associated with leukemia. The analysis was performed by multiplex polymerase chain reaction for mutations in genes of interest followed by primer extension reactions. Products were analyzed on a Sequenom MassARRAY system (San Diego, CA). The multiplex panel yielded mutations in 58% of acute myeloid leukemia cases with normal cytogenetics and 21% of cases with abnormal cytogenetics. Cytogenetics and routine polymerase chain reaction-based screening of NPM1, CEBPA, FLT3-ITD, and KIT was also performed on a subset of cases. When combined with the results of these standard polymerase chain reaction-based tests, the mutation frequency reached 78% in cases with normal cytogenetics. Of these, 42% harbored multiple mutations primarily involving NPM1 with NRAS, KRAS, CEBPA, PTPN11, IDH1, or FLT3. In contrast, cases with abnormal cytogenetics rarely harbored more than 1 mutation (1.5%), suggesting different underlying biology. This study demonstrates the feasibility and utility of broad-based mutation profiling of acute myeloid leukemia in a clinical setting. This approach will be helpful in defining prognostic subgroups of acute myeloid leukemia and contribute to the selection of patients for enrollment into trials with novel inhibitors.


Clinical Cancer Research | 2014

Genetic Profiling to Determine Risk of Relapse-Free Survival in High-Risk Localized Prostate Cancer

Christine M. Barnett; Michael C. Heinrich; Jeong Lim; Dylan Nelson; Carol Beadling; Andrea Warrick; Tanaya Neff; Celestia S. Higano; Mark Garzotto; David Z. Qian; Christopher L. Corless; George Thomas; Tomasz M. Beer

Purpose: The characterization of actionable mutations in human tumors is a prerequisite for the development of individualized, targeted therapy. We examined the prevalence of potentially therapeutically actionable mutations in patients with high-risk clinically localized prostate cancer. Experimental Design: Forty-eight samples of formalin-fixed paraffin-embedded prostatectomy tissue from a neoadjuvant chemotherapy trial were analyzed. DNA extracted from microdissected tumor was analyzed for 643 common solid tumor mutations in 53 genes using mass spectroscopy–based sequencing. In addition, PTEN loss and erythroblast transformation-specific–related gene (ERC) translocations were examined using immunohistochemistry (IHC) in associated tissue microarrays. Association with relapse during 5 years of follow-up was examined in exploratory analyses of the potential clinical relevance of the genetic alterations. Results: Of the 40 tumors evaluable for mutations, 10% had point mutations in potentially actionable cancer genes. Of the 47 tumors evaluable for IHC, 36% had PTEN loss and 40% had ERG rearrangement. Individual mutations were not frequent enough to determine associations with relapse. Using Kaplan–Meier analysis with a log-rank test, the 16 patients who had PTEN loss had a significantly shorter median relapse-free survival, 19 versus 106 months (P = 0.01). Conclusions: This study confirms that point mutations in the most common cancer regulatory genes in prostate cancer are rare. However, the PIK3CA/AKT pathway was mutated in 10% of our samples. Although point mutations alone did not have a statistically significant association with relapse, PTEN loss was associated with an increased relapse in high-risk prostate cancer treated with chemotherapy followed by surgery. Clin Cancer Res; 20(5); 1306–12. ©2013 AACR.


Journal of Clinical Oncology | 2013

Genomic analysis of prostate cancer.

Christine M. Barnett; Michael C. Heinrich; Dylan Nelson; Jeong Youn Lim; Carol Beadling; Andrea Warrick; Tanaya Neff; George Thomas; Mark Garzotto; Celestia S. Higano; Tomasz M. Beer; David Z. Qian

80 Background: The characterization of actionable mutations is a prerequisite for the development of individualized, targeted therapy. We sought to characterize the prevalence of potentially therapeutically actionable mutations in patients with high risk prostate cancer. METHODS 48 samples of formalin fixed paraffin embedded prostatectomy tissue from a neoadjuvant chemotherapy trial (Garzotto M, Cancer 2010 Apr 1;116(7):1699-708) were analyzed. DNA was extracted from microdissected tumor. Tumor DNA was analyzed for 643 common solid tumor point mutations in 53 genes using mass spectroscopy based sequencing (Sequenom MassArray). Low level mutations were validated using ion chip pH based sequencing (Ion Torrent). In addition, PTEN loss and ERG translocations were examined using immunohistochemistry in associated tissue microarrays. Correlation with relapse during 5 years of follow-up was examined in exploratory analyses of the potential clinical relevance of the genetic alterations examined. RESULTS Of the 40 tumors evaluable for mutations, 10% had point mutations in potentially actionable cancer genes. Of the 45 tumors evaluable for IHC, 36% had PTEN loss (6% with a heterogenous pattern) and 40% had ERG rearrangement (2% with heterogeneous pattern). Individual mutations were not frequent enough to determine associations with progression. Using Kaplan-Meier analysis with a log-rank test, the 16 patients who had PTEN loss had a significantly shorter median progression free survival, 19 vs. 106 months (p = .01). CONCLUSIONS This study confirms that point mutations in the most common cancer regulatory genes in prostate cancer are rare. However, the PIK3CA/AKT pathway was mutated in 10% of our samples. While point mutations alone did not have a statistically significant effect on relapse, PTEN loss was associated with an increased relapse in high risk prostate cancer treated with chemotherapy followed by surgery. It is yet unclear if the 16 weeks of chemotherapy played a role in selecting for tumors with these biologic features, and this will therefore be the subject of future investigations. [Table: see text].

Collaboration


Dive into the Tanaya Neff's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Celestia S. Higano

Fred Hutchinson Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge